Next Article in Journal
TAK1 Mediates ROS Generation Triggered by the Specific Cephalosporins through Noncanonical Mechanisms
Previous Article in Journal
P27 Protects Neurons from Ischemic Damage by Suppressing Oxidative Stress and Increasing Autophagy in the Hippocampus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Prognostic Significance of COX-2 Overexpression in BRAF-Mutated Middle Eastern Papillary Thyroid Carcinoma

by
Sandeep Kumar Parvathareddy
1,†,
Abdul K. Siraj
1,†,
Padmanaban Annaiyappanaidu
1,
Saif S. Al-Sobhi
2,
Fouad Al-Dayel
3 and
Khawla S. Al-Kuraya
1,*
1
Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia
2
Department of Surgery, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia
3
Department of Pathology, King Faisal Specialist Hospital and Research Centre, P.O. Box 3354, Riyadh 11211, Saudi Arabia
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2020, 21(24), 9498; https://doi.org/10.3390/ijms21249498
Submission received: 8 November 2020 / Revised: 27 November 2020 / Accepted: 10 December 2020 / Published: 14 December 2020
(This article belongs to the Section Molecular Oncology)

Abstract

:
The cyclooxygenase-2 (COX-2)–prostaglandin E2 (PGE2) pathway has been implicated in carcinogenesis, with BRAF mutation shown to promote PGE2 synthesis. This study was conducted to evaluate COX-2 expression in a large cohort of Middle Eastern papillary thyroid carcinoma (PTC), and further evaluate the prognostic significance of COX-2 expression in strata of BRAF mutation status. BRAF mutation analysis was performed using Sanger sequencing, and COX-2 expression was evaluated immunohistochemically using tissue microarray (TMA). COX-2 overexpression, noted in 43.2% (567/1314) of cases, was significantly associated with poor prognostic markers such as extra-thyroidal extension, lymph-node metastasis, and higher tumor stage. COX-2 was also an independent predictor of poor disease-free survival (DFS). Most notably, the association of COX-2 expression with DFS differed by BRAF mutation status. COX-2 overexpression was associated with poor DFS in BRAF-mutant but not BRAF wild-type PTCs, with a multivariate-adjusted hazard ratio of 2.10 (95% CI = 1.52–2.92; p < 0.0001) for COX-2 overexpressed tumors in BRAF-mutant PTC. In conclusion, the current study shows that COX-2 plays a key role in prognosis of PTC patients, especially in BRAF-mutated tumors. Our data suggest the potential therapeutic role of COX-2 inhibition in patients with BRAF-mutated PTC.

1. Introduction

Thyroid cancer is the most common endocrine malignancy. Its incidence is steadily rising worldwide [1,2], with the most common histology being papillary thyroid carcinoma (PTC) [3]. The incidence of PTC in Saudi Arabia is high—it is the second most common cancer affecting Saudi women, after breast cancer [4]. Although PTC is an indolent and slow-growing malignancy that can be successfully treated, there are patients that progress to more aggressive disease, with recurrence and metastasis [5,6,7]. It is critical to identify this subset of patients who might benefit from more aggressive therapy. Therefore, identification of a molecular marker that can be useful for prognostication of PTC patients is critically needed.
Tumor-promoting inflammation is a promising target for cancer therapy [8] due to its role in promoting cancer progression by increasing several growth, angiogenic, and immunosuppressive factors [9]. One of the important processes causing cancer inflammation is the cyclooxygenase-2 (COX-2) pathway. COX is a group of enzymes that are required for prostaglandin E2 (PGE2) synthesis [10]. In normal physiological status, COX-2 and PGE2 are upregulated and act as pro-inflammatory factors [11]. Although COX-2 expression is usually undetectable in normal tissue, it has been observed to be overexpressed in several human cancers [12,13,14,15]. Many studies have described the mechanisms by which COX-2 can promote carcinogenesis, including inhibition of apoptosis, increase in cell proliferation, stimulation of angiogenesis, and development of a tumor-promoting inflammatory microenvironment [9,16,17,18,19,20].
COX-2 overexpression in different thyroid cancer histotypes has also been investigated in several studies [21,22,23], the majority of which have been aimed at evaluation of its prognostic relevance by correlating the expression levels with patients’ clinicopathological features. Other studies have attempted to estimate the prognostic value of COX-2 expression in thyroid cancer [15,24]. Those studies found that COX-2 overexpression in thyroid cancer is associated with aggressive clinical behavior and tumor recurrence [15,25].
Another characteristic of PTC is that a single nucleotide mutation in the BRAF gene, V600E, is detected in a majority of PTC patients [26,27], often associated with aggressive disease [7,28]. BRAFV600E mutations have been shown to induce oncogenic cellular proliferation by constitutively activating the mitogen-activated protein (MAP) kinase pathway [29]. A recent study by Kosumin et al. [30] showed the interactive role of BRAF mutation status and COX-2 overexpression in the prognostication of colorectal cancer patients, suggesting that upregulation ofMAP kinase pathway mediates overexpression of COX-2 in BRAFV600E tumor cells.
Therefore, we conducted this study to evaluate COX-2 overexpression in a large cohort (n = 1335) of Middle Eastern PTC, and to analyze its correlation with clinicopathological parameters. Then we further evaluated the prognostic significance of COX-2 expression in strata of BRAF mutation status.

2. Results

2.1. Patient Characteristics

The mean age of the study population was 40.4 years (SD = ±16.1 years), with a male-to-female ratio of 1:3. A majority of the cases were of classical variant (67.1%) and Stage I tumors (83.4%). A nearly equal proportion of tumors exhibited presence and absence of extra-thyroidal extension, as well as multifocality (Table 1). Patient characteristics stratified by BRAF mutation status is presented in Table 1.

2.2. Clinicopathological Associations of COX-2 Expression in PTC

We performed immunohistochemical analysis to look for COX-2 protein expression in our cohort of 1335 PTC cases. Immunohistochemistry data were interpretable in 1314 cases, and cytoplasmic staining was noted (Figure 1). COX-2 overexpression, noted in 43.2% (567/1314) of cases, was significantly associated with older age (p < 0.0001), extra-thyroidal extension (p < 0.0001), T4 tumors (p = 0.0094), lymph-node metastasis (p = 0.0003), and advanced stage (p < 0.0001) (Table 2). A significant association was also observed between COX-2 overexpression and poor disease-free survival (DFS; p < 0.0001) (Table 2; Figure 2A). Multivariate analysis using the Cox regression model, after adjusting for possible confounding factors of survival, showed that COX-2 overexpression was an independent predictor of poor DFS (HR = 1.57; 95% CI = 1.22–2.03; p = 0.0004) (Table 3).

2.3. COX-2 Expression and BRAF Mutation in PTC

We found a significant association between COX-2 overexpression and BRAF mutation (p = 0.0055) in our cohort. Next, we examined the prognostic association of COX-2 expression in the strata of BRAF mutation status. Using Kaplan–Meier survival analyses, we found COX-2 expression was associated with a significantly shorter DFS in BRAF-mutant PTC, but not in BRAF wild-type cases (Figure 2B,C). Multivariate analysis showed COX-2 overexpression was associated with shorter DFS in BRAF-mutant PTC cases (HR = 2.10; 95% CI = 1.52–2.92; p < 0.0001) (Table 4).

3. Discussion

We conducted this study using a large cohort of more than 1300 Middle Eastern PTC tumors, and found a frequency of COX-2 expression of 43.2%. There was a strong association of COX-2 expression with adverse markers of PTC prognosis, most notably extra-thyroidal extension, lymph-node metastasis, and higher tumor stage. A recent study by Fu et al. [15] demonstrated a similar association of COX-2 expression with aggressive clinicopathological features such as extra-thyroidal extension and multifocality in 252 PTC samples. Several previous studies have explored the usefulness of COX-2 as a biomarker of thyroid malignancies and its potential role in PTC carcinogenesis [15,31,32,33], but the prognostic role of COX-2 in PTC is still controversial [34,35]. Our current study shows that elevated COX-2 expression is an independent predictor of poor DFS in patients with PTC.
COX-2 mainly generates prostaglandin E2 (PGE2). The COX-2–PGE2 pathway is known to play an important role in tumor progression, and its oncogenic role has been shown in several tumor types [36,37,38,39]. However, recent evidence shows that PGE2 is important in modifying the tumor microenvironment to induce immune tolerance [40,41,42]. PGE2 induces alterations in cytokine balance and causes suppression of lymphocyte proliferation following mitogen stimulation and inhibition of dendritic cells [43,44,45]. Given the known function of PGE2 in modulating the tumor microenvironment to suppress an immune response [46], our data clearly show the strong relationship among cancer progression, aggressiveness, lymph-node metastasis, and COX-2 expression.
We observed a differential prognostic association of COX-2 expression according to BRAF mutation status in our study. The prognostic association of COX-2 expression was significantly pronounced in BRAF-mutated PTC compared to BRAF wild-type PTC. A similar observation has been documented recently in CRC [30]. This further supports the role of BRAF mutation in the acceleration of the production of PGE2 via COX-2 [41,47,48], and highlights how a subset of PTC might use increased COX-2 activity as a possible pathway that affects the survival of patients with the BRAF mutation.
Our study reveals the necessity for additional prognostic biomarkers for PTC, especially markers of tumor-associated inflammation, which could be used to tailor therapeutic approaches and improve patient survival. Although our findings suggest that inhibition of COX-2 in BRAF-mutated PTC might be a good therapeutic strategy, clinical trials and further clinical research is needed to explore the therapeutic advantage of adding COX-2 inhibitors to the current iodine therapy in BRAF-mutated PTC patients. The findings of our study are summarized in Figure 3.

4. Materials and Methods

4.1. Sample Selection

The study included 1335 PTC patients diagnosed between 1989 and 2018 at King Faisal Specialist Hospital and Research Center (Riyadh, Saudi Arabia) with available archival tissue samples. Clinicopathological data were collected from case records (Table 1). The hospital’s institutional review board approved the collection of archival samples. For this study, since only archived paraffin tissue blocks were used, a waiver of consent was obtained from the Research Advisory Council (RAC) under project RAC# 2110 031.

4.2. DNA Isolation

DNA was extracted from PTC formalin-fixed and paraffin-embedded (FFPE) tumor tissues using the Gentra DNA isolation kit (Gentra, Minneapolis, MN, USA) according to the manufacturer’s protocols as elaborated in previous studies [49].

4.3. Sanger Sequencing Analysis

The sequencing of entire coding and splicing regions of exon 15 in BRAF genes, and exon 2 and 3 in HRAS and NRAS genes, in the 1335 PTC samples was carried out using Sanger sequencing technology. Primer 3 online software was used to design the primers (available upon request). PCR and Sanger sequencing analysis were conducted as described previously [50]. The reference sequence was downloaded from NCBI GenBank. The sequencing results were compared with the reference sequence by Mutation Surveyor V4.04 (Soft Genetics, LLC, State College, PA, USA).

4.4. Tissue Microarray (TMA) Construction and Immunohistochemistry (IHC) Analysis

The tissue microarray (TMA) format was used for immunohistochemical analysis of the PTC samples. TMA was constructed as previously described [51]. A modified semiautomatic robotic precision instrument (Beecher Instruments, Woodland, WI, USA) was used to punch tissue cylinders with a diameter of 0.6 mm from a representative tumor area of the donor tissue block into the recipient paraffin block. Two 0.6 mm cores of PTC were arrayed from each case.
Tissue microarray slides were processed and stained manually as described previously [52], using a primary antibody against COX-2 (ab15191, 1:1000 dilution, pH 6.0, Abcam, Cambridge, UK). The Dako Envision Plus System kit was used as the secondary detection system with 3,30-diaminobenzidine as chromogen. All slides were counterstained with hematoxylin, dehydrated, cleared, and mounted. The negative controls included omission of the primary antibody. Normal tissues of different organ system were also included in the TMA to serve as controls. Only fresh-cut slides were stained simultaneously to minimize the influence of slide aging and maximize reproducibility of the experiment.
A cytoplasmic staining was observed and scored. COX-2 was scored as described previously [53] using the H score. The intensity of staining was scored from 0–3 (0: absent, 1+: weak, 2+: moderate, 3+: strong), and the proportion of tumor-cell staining for a particular intensity was recorded in 5% increments in the range of 0–100. A final H score was assigned using the following formula: H score = (1 × (% cells 1+) + 2 × (% cells 2+) + 3 × (% cells 3+)). This H score ranged from 0–300. Two scores per tumor were analyzed to minimize the number of missing/uninterpretable spots. However, the higher of the two scores was used as the final score. X-tile plots were constructed for the assessment of biomarkers and the optimization of cut-off points based on an outcome described earlier [54]. Based on X-tile plots, PTC cases were classified into two subgroups: those with an H score of 0, defined as negative expression of COX-2, and those with an H score > 0, defined as overexpression.

4.5. Statistical Analysis

The associations between clinico-pathological variables and protein expression were derived using contingency table analysis and Chi square tests. The Mantel–Cox log-rank test was used to evaluate disease-free survival. Survival curves were generated using the Kaplan–Meier method. The Cox proportional-hazards regression model was used for multivariate analysis. Two-sided tests were used for statistical analyses, with a limit of significance defined as p value < 0.05. Data analyses were performed using the JMP11.0 (SAS Institute, Inc., Cary, NC, USA) software package.

5. Conclusions

Recently, the possibility of combining different therapies, including biological therapies and kinase inhibitors, has contributed to tailoring the treatment of cancer patients and improving their survival. Our data show that COX-2 correlates with PTC disease-free survival in BRAF-mutated tumors, representing a useful prognostic marker for risk stratification of thyroid cancer patients. These findings have clinical relevance because they provide a rationale to test COX-2 inhibition as a potential treatment to prevent PTC progression and enhance the antitumor activity of other cancer therapies to treat patients with aggressive PTC and BRAF mutations.

Author Contributions

Conceptualization, K.S.A.-K.; Methodology, S.K.P. and A.K.S.; Software, S.K.P.; Validation, S.K.P. and P.A.; Formal analysis, S.K.P., A.K.S. and P.A.; Investigation, S.K.P., P.A., S.S.A.-S. and F.A.-D.; Resources, S.K.P., S.S.A.-S. and F.A.-D.; Data curation, S.K.P., A.K.S. and P.A.; Writing—Original draft preparation, K.S.A.-K.; Writing—Review and editing, S.K.P., A.K.S. and K.S.A.-K.; Visualization, A.K.S. and S.K.P.; Supervision, K.S.A.-K. and A.K.S.; Project administration, K.S.A.-K. and A.K.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors would like to thank Felisa DeVera for technical assistance.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

COX-2Cyclooxygenase-2
PGE2Prostaglandin E2
PTCPapillary thyroid carcinoma
DFSDisease-free survival
FFPEFormalin-fixed and paraffin-embedded
PCRPolymerase chain reaction
TMATissue microarray
IHCImmunohistochemistry
SDStandard deviation
HRHazard ratio
CIConfidence interval

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. La Vecchia, C.; Malvezzi, M.; Bosetti, C.; Garavello, W.; Bertuccio, P.; Levi, F.; Negri, E. Thyroid cancer mortality and incidence: A global overview. Int. J. Cancer 2015, 136, 2187–2195. [Google Scholar] [CrossRef] [PubMed]
  3. Lim, H.; Devesa, S.S.; Sosa, J.A.; Check, D.; Kitahara, C.M. Trends in thyroid cancer incidence and mortality in the United States, 1974–2013. JAMA 2017, 317, 1338–1348. [Google Scholar] [CrossRef] [PubMed]
  4. Alrawaji, A.; Alshahrani, Z.; Alzahrani, W.; Alomran, F.; Almadouj, A.; Alshehri, S.; Alzahrani, A.; Bazarbashi, S.; Alhashmi, H.; Almutlaq, H.; et al. Cancer Incidence Report Saudi Arabia 2015. In Saudi Cancer Registry; Saudi Health Council: Riyadh, Saudi Arabia, 2018. [Google Scholar]
  5. Ritter, A.; Mizrachi, A.; Bachar, G.; Vainer, I.; Shimon, I.; Hirsch, D.; Diker-Cohen, T.; Duskin-Bitan, H.; Robenshtok, E. Detecting recurrence following lobectomy for thyroid cancer: Role of thyroglobulin and thyroglobulin antibodies. J. Clin. Endocrinol. Metab. 2020, 105, dgaa152. [Google Scholar] [CrossRef] [PubMed]
  6. Tumino, D.; Frasca, F.; Newbold, K. Updates on the management of advanced, metastatic, and radioiodine refractory differentiated thyroid cancer. Front. Endocrinol. 2017, 8, 312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Xing, M.; Alzahrani, A.S.; Carson, K.A.; Shong, Y.K.; Kim, T.Y.; Viola, D.; Elisei, R.; Bendlová, B.; Yip, L.; Mian, C. Association between BRAF V600E mutation and recurrence of papillary thyroid cancer. J. Clin. Oncol. 2015, 33, 42. [Google Scholar] [CrossRef] [Green Version]
  8. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  9. Wang, D.; DuBois, R.N. Eicosanoids and cancer. Nat. Rev. Cancer 2010, 10, 181–193. [Google Scholar] [CrossRef]
  10. Funk, C.D. Prostaglandins and leukotrienes: Advances in eicosanoid biology. Science 2001, 294, 1871–1875. [Google Scholar] [CrossRef] [Green Version]
  11. Dubois, R.N.; Abramson, S.B.; Crofford, L.; Gupta, R.A.; Simon, L.S.; Van De Putte, L.B.; Lipsky, P.E. Cyclooxygenase in biology and disease. FASEB J. 1998, 12, 1063–1073. [Google Scholar] [CrossRef] [Green Version]
  12. Petkova, D.; Clelland, C.; Ronan, J.; Pang, L.; Coulson, J.; Lewis, S.; Knox, A. Overexpression of cyclooxygenase-2 in non-small cell lung cancer. Respir. Med. 2004, 98, 164–172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Agrawal, U.; Kumari, N.; Vasudeva, P.; Mohanty, N.K.; Saxena, S. Overexpression of COX2 indicates poor survival in urothelial bladder cancer. Ann. Diagn. Pathol. 2018, 34, 50–55. [Google Scholar] [CrossRef] [PubMed]
  14. Ogino, S.; Kirkner, G.J.; Nosho, K.; Irahara, N.; Kure, S.; Shima, K.; Hazra, A.; Chan, A.T.; Dehari, R.; Giovannucci, E.L. Cyclooxygenase-2 expression is an independent predictor of poor prognosis in colon cancer. Clin. Cancer Res. 2008, 14, 8221–8227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Fu, X.; Zhang, H.; Chen, Z.; Yang, Z.; Shi, D.; Liu, T.; Chen, W.; Yao, F.; Su, X.; Deng, W. TFAP2B overexpression contributes to tumor growth and progression of thyroid cancer through the COX-2 signaling pathway. Cell Death Dis. 2019, 10, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Xiao, Y.; Teng, Y.; Zhang, R.; Luo, L. Antitumor effect of the selective COX-2 inhibitor celecoxib on endometrial adenocarcinoma in vitro and in vivo. Oncol. Lett. 2012, 4, 1219–1224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Xu, L.; Stevens, J.; Hilton, M.B.; Seaman, S.; Conrads, T.P.; Veenstra, T.D.; Logsdon, D.; Morris, H.; Swing, D.A.; Patel, N.L. COX-2 inhibition potentiates antiangiogenic cancer therapy and prevents metastasis in preclinical models. Sci. Transl. Med. 2014, 6, 242ra84. [Google Scholar] [CrossRef]
  18. Xu, B.; Wang, Y.; Yang, J.; Zhang, Z.; Zhang, Y.; Du, H. Celecoxib induces apoptosis but up-regulates VEGF via endoplasmic reticulum stress in human colorectal cancer in vitro and in vivo. Cancer Chemother. Pharmacol. 2016, 77, 797–806. [Google Scholar] [CrossRef]
  19. Liu, B.; Qu, L.; Yan, S. Cyclooxygenase-2 promotes tumor growth and suppresses tumor immunity. Cancer Cell Int. 2015, 15, 106. [Google Scholar] [CrossRef] [Green Version]
  20. Greenhough, A.; Smartt, H.J.; Moore, A.E.; Roberts, H.R.; Williams, A.C.; Paraskeva, C.; Kaidi, A. The COX-2/PGE 2 pathway: Key roles in the hallmarks of cancer and adaptation to the tumour microenvironment. Carcinogenesis 2009, 30, 377–386. [Google Scholar] [CrossRef] [Green Version]
  21. Krawczyk-Rusiecka, K.; Lewiński, A. Cyclooxygenase-2 expression and its association with thyroid lesions. Arch. Med. Sci. 2010, 6, 653. [Google Scholar] [CrossRef]
  22. Krawczyk-Rusiecka, K.; Wojciechowska-Durczynska, K.; Cyniak-Magierska, A.; Zygmunt, A.; Lewinski, A. Assessment of cyclooxygenase-1 and 2 gene expression levels in chronic autoimmune thyroiditis, papillary thyroid carcinoma and nontoxic nodular goitre. Thyroid Res. 2014, 7, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Haynik, D.M.; Prayson, R.A. Immunohistochemical expression of cyclooxygenase 2 in follicular carcinomas of the thyroid. Arch. Pathol. Lab. Med. 2005, 129, 736–741. [Google Scholar] [PubMed]
  24. Perisa, M.M.; Sarcevic, B.; Troselj, K.G.; Grsic, K.; Sitic, S.; Seiwerth, S. Expression of nm23-H1 and COX-2 in thyroid papillary carcinoma and microcarcinoma. Oncol. Lett. 2017, 13, 3547–3555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Ji, B.; Liu, Y.; Zhang, P.; Wang, Y.; Wang, G. COX-2 expression and tumor angiogenesis in thyroid carcinoma patients among northeast Chinese population-result of a single-center study. Int. J. Med. Sci. 2012, 9, 237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Ge, J.; Wang, J.; Wang, H.; Jiang, X.; Liao, Q.; Gong, Q.; Mo, Y.; Li, X.; Li, G.; Xiong, W. The BRAF V600E mutation is a predictor of the effect of radioiodine therapy in papillary thyroid cancer. J. Cancer 2020, 11, 932. [Google Scholar] [CrossRef] [Green Version]
  27. Yan, C.; Huang, M.; Li, X.; Wang, T.; Ling, R. Relationship between BRAF V600E and clinical features in papillary thyroid carcinoma. Endocr. Connect. 2019, 8, 988–996. [Google Scholar] [CrossRef] [Green Version]
  28. Gan, X.; Shen, F.; Deng, X.; Feng, J.; Lu, J.; Cai, W.; Peng, L.; Zheng, W.; Wang, W.; Huang, P. Prognostic implications of the BRAF-V600E mutation in papillary thyroid carcinoma based on a new cut-off age stratification. Oncol. Lett. 2020, 19, 631–640. [Google Scholar] [CrossRef] [Green Version]
  29. Wan, P.T.; Garnett, M.J.; Roe, S.M.; Lee, S.; Niculescu-Duvaz, D.; Good, V.M.; Project, C.G.; Jones, C.M.; Marshall, C.J.; Springer, C.J. Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell 2004, 116, 855–867. [Google Scholar] [CrossRef] [Green Version]
  30. Kosumi, K.; Hamada, T.; Zhang, S.; Liu, L.; da Silva, A.; Koh, H.; Twombly, T.S.; Mima, K.; Morikawa, T.; Song, M. Prognostic association of PTGS2 (COX-2) over-expression according to BRAF mutation status in colorectal cancer: Results from two prospective cohorts and CALGB 89803 (Alliance) trial. Eur. J. Cancer 2019, 111, 82–93. [Google Scholar] [CrossRef]
  31. Meng, X.-Y.; Zhang, Q.; Li, Q.; Lin, S.; Li, J. Immunohistochemical levels of cyclo-oxygenase-2, matrix metalloproteinase-9 and vascular endothelial growth factor in papillary thyroid carcinoma and their clinicopathological correlations. J. Int. Med. Res. 2014, 42, 619–627. [Google Scholar] [CrossRef] [Green Version]
  32. Erdem, H.; Gündogdu, C.; Şipal, S. Correlation of E-cadherin, VEGF, COX-2 expression to prognostic parameters in papillary thyroid carcinoma. Exp. Mol. Pathol. 2011, 90, 312–317. [Google Scholar] [CrossRef] [PubMed]
  33. Giaginis, C.; Alexandrou, P.; Delladetsima, I.; Karavokyros, I.; Danas, E.; Giagini, A.; Patsouris, E.; Theocharis, S. Clinical significance of Hu-antigen receptor (HuR) and cyclooxygenase-2 (COX-2) expression in human malignant and benign thyroid lesions. Pathol. Oncol. Res. 2016, 22, 189–196. [Google Scholar] [CrossRef] [PubMed]
  34. Cunha, L.L.; Marcello, M.A.; Nonogaki, S.; Morari, E.C.; Soares, F.A.; Vassallo, J.; Ward, L.S. CD 8+ tumour-infiltrating lymphocytes and COX2 expression may predict relapse in differentiated thyroid cancer. Clin. Endocrinol. 2015, 83, 246–253. [Google Scholar] [CrossRef] [PubMed]
  35. Kim, H.R.; Si, Y.; Lee, Y.S.; Kim, J.S.; Jeon, H.M.; Park, W.C. The Clinical Significance of Expression of Cyclooxygenase-2 in the Prognosis of the Papillary Thyroid Cancer. Korean J. Endocr. Surg. 2007, 7, 75–79. [Google Scholar] [CrossRef]
  36. Wang, D.; Fu, L.; Sun, H.; Guo, L.; DuBois, R.N. Prostaglandin E2 promotes colorectal cancer stem cell expansion and metastasis in mice. Gastroenterology 2015, 149, 1884–1895.e4. [Google Scholar] [CrossRef] [Green Version]
  37. Liu, Y.; Sun, H.; Hu, M.; Zhang, Y.; Chen, S.; Tighe, S.; Zhu, Y. The role of cyclooxygenase-2 in colorectal carcinogenesis. Clin. Colorectal Cancer 2017, 16, 165–172. [Google Scholar] [CrossRef]
  38. Xie, C.; Xu, X.; Wang, X.; Wei, S.; Shao, L.; Chen, J.; Cai, J.; Jia, L. Cyclooxygenase-2 induces angiogenesis in pancreatic cancer mediated by prostaglandin E2. Oncol. Lett. 2018, 16, 940–948. [Google Scholar] [CrossRef] [Green Version]
  39. Hashemi Goradel, N.; Najafi, M.; Salehi, E.; Farhood, B.; Mortezaee, K. Cyclooxygenase-2 in cancer: A review. J. Cell. Physiol. 2019, 234, 5683–5699. [Google Scholar] [CrossRef]
  40. Mizuno, R.; Kawada, K.; Sakai, Y. Prostaglandin E2/EP Signaling in the Tumor Microenvironment of Colorectal Cancer. Int. J. Mol. Sci. 2019, 20, 6254. [Google Scholar] [CrossRef] [Green Version]
  41. Zelenay, S.; Van Der Veen, A.G.; Böttcher, J.P.; Snelgrove, K.J.; Rogers, N.; Acton, S.E.; Chakravarty, P.; Girotti, M.R.; Marais, R.; Quezada, S.A. Cyclooxygenase-dependent tumor growth through evasion of immunity. Cell 2015, 162, 1257–1270. [Google Scholar] [CrossRef] [Green Version]
  42. Muller, A.J.; Scherle, P.A. Targeting the mechanisms of tumoral immune tolerance with small-molecule inhibitors. Nat. Rev. Cancer 2006, 6, 613–625. [Google Scholar] [CrossRef]
  43. Zhang, Q.; Zhu, B.; Li, Y. Resolution of cancer-promoting inflammation: A new approach for anticancer therapy. Front. Immunol. 2017, 8, 71. [Google Scholar] [CrossRef] [Green Version]
  44. Göbel, C.; Breitenbuecher, F.; Kalkavan, H.; Hähnel, P.; Kasper, S.; Hoffarth, S.; Merches, K.; Schild, H.; Lang, K.; Schuler, M. Functional expression cloning identifies COX-2 as a suppressor of antigen-specific cancer immunity. Cell Death Dis. 2014, 5, e1568. [Google Scholar] [CrossRef] [Green Version]
  45. Wu, A.A.; Drake, V.; Huang, H.-S.; Chiu, S.; Zheng, L. Reprogramming the tumor microenvironment: Tumor-induced immunosuppressive factors paralyze T cells. Oncoimmunology 2015, 4, e1016700. [Google Scholar] [CrossRef] [PubMed]
  46. Liu, M.; Matsumura, N.; Mandai, M.; Li, K.; Yagi, H.; Baba, T.; Suzuki, A.; Hamanishi, J.; Fukuhara, K.; Konishi, I. Classification using hierarchical clustering of tumor-infiltrating immune cells identifies poor prognostic ovarian cancers with high levels of COX expression. Mod. Pathol. 2009, 22, 373–384. [Google Scholar] [CrossRef] [PubMed]
  47. Yu, H.; Lee, H.; Herrmann, A.; Buettner, R.; Jove, R. Revisiting STAT3 signalling in cancer: New and unexpected biological functions. Nat. Rev. Cancer 2014, 14, 736–746. [Google Scholar] [CrossRef] [PubMed]
  48. Shi, C.; Yang, Y.; Xia, Y.; Okugawa, Y.; Yang, J.; Liang, Y.; Chen, H.; Zhang, P.; Wang, F.; Han, H. Novel evidence for an oncogenic role of microRNA-21 in colitis-associated colorectal cancer. Gut 2016, 65, 1470–1481. [Google Scholar] [CrossRef]
  49. Abubaker, J.; Jehan, Z.; Bavi, P.; Sultana, M.; Al-Harbi, S.; Ibrahim, M.; Al-Nuaim, A.; Ahmed, M.; Amin, T.; Al-Fehaily, M. Clinicopathological analysis of papillary thyroid cancer with PIK3CA alterations in a Middle Eastern population. J. Clin. Endocrinol. Metab. 2008, 93, 611–618. [Google Scholar] [CrossRef] [Green Version]
  50. Bu, R.; Siraj, A.K.; Al-Obaisi, K.A.; Beg, S.; Al Hazmi, M.; Ajarim, D.; Tulbah, A.; Al-Dayel, F.; Al-Kuraya, K.S. Identification of novel BRCA founder mutations in Middle Eastern breast cancer patients using capture and Sanger sequencing analysis. Int. J. Cancer 2016, 139, 1091–1097. [Google Scholar] [CrossRef] [Green Version]
  51. Siraj, A.; Bavi, P.; Abubaker, J.; Jehan, Z.; Sultana, M.; Al-Dayel, F.; Al-Nuaim, A.; Alzahrani, A.; Ahmed, M.; Al-Sanea, O. Genome-wide expression analysis of Middle Eastern papillary thyroid cancer reveals c-MET as a novel target for cancer therapy. J. Pathol. 2007, 213, 190–199. [Google Scholar] [CrossRef]
  52. Bavi, P.; Jehan, Z.; Atizado, V.; Al-Dossari, H.; Al-Dayel, F.; Tulbah, A.; Amr, S.S.; Sheikh, S.S.; Ezzat, A.; El-Solh, H. Prevalence of fragile histidine triad expression in tumors from Saudi Arabia: A tissue microarray analysis. Cancer Epidemiol. Prev. Biomark. 2006, 15, 1708–1718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Ahmed, M.; Hussain, A.R.; Siraj, A.K.; Uddin, S.; Al-Sanea, N.; Al-Dayel, F.; Al-Assiri, M.; Beg, S.; Al-Kuraya, K.S. Co-targeting of Cyclooxygenase-2 and FoxM1 is a viable strategy in inducing anticancer effects in colorectal cancer cells. Mol. Cancer 2015, 14, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Camp, R.L.; Dolled-Filhart, M.; Rimm, D.L. X-tile: A new bio-informatics tool for biomarker assessment and outcome-based cut-point optimization. Clin. Cancer Res. 2004, 10, 7252–7259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Cyclooxygenase-2 (COX-2) immunohistochemical staining in papillary thyroid carcinoma (PTC) tissue microarray (TMA). Representative examples of tumors showing (A) high expression and (B) low expression of COX-2 (20×/0.70 objective on an Olympus BX 51 microscope (Olympus America Inc., Center Valley, PA, USA), with the inset showing a 40×/0.85 aperture magnified view of the same TMA spot).
Figure 1. Cyclooxygenase-2 (COX-2) immunohistochemical staining in papillary thyroid carcinoma (PTC) tissue microarray (TMA). Representative examples of tumors showing (A) high expression and (B) low expression of COX-2 (20×/0.70 objective on an Olympus BX 51 microscope (Olympus America Inc., Center Valley, PA, USA), with the inset showing a 40×/0.85 aperture magnified view of the same TMA spot).
Ijms 21 09498 g001
Figure 2. Survival analyses of COX-2 protein expression in the overall cohort and based on BRAF mutation status. (A) Kaplan–Meier survival plot showing statistically significant poor disease-free survival in COX-2 high-expression cases compared to COX-2 low-expression cases (p < 0.0001); (B) Kaplan–Meier survival plot showing no statistically significant difference in disease-free survival for COX-2 expression in BRAF wild-type PTC (p = 0.7675); (C) Kaplan–Meier survival plot showing statistically significant poor disease-free survival for cases with COX-2 overexpression in BRAF-mutant PTC (p < 0.0001).
Figure 2. Survival analyses of COX-2 protein expression in the overall cohort and based on BRAF mutation status. (A) Kaplan–Meier survival plot showing statistically significant poor disease-free survival in COX-2 high-expression cases compared to COX-2 low-expression cases (p < 0.0001); (B) Kaplan–Meier survival plot showing no statistically significant difference in disease-free survival for COX-2 expression in BRAF wild-type PTC (p = 0.7675); (C) Kaplan–Meier survival plot showing statistically significant poor disease-free survival for cases with COX-2 overexpression in BRAF-mutant PTC (p < 0.0001).
Ijms 21 09498 g002
Figure 3. Schematic presentation of the study findings and future directions.
Figure 3. Schematic presentation of the study findings and future directions.
Ijms 21 09498 g003
Table 1. Patient characteristics according to BRAF mutation status.
Table 1. Patient characteristics according to BRAF mutation status.
Total BRAF Wild-Type BRAF Mutant
No.%No.%No.%
Total1335 58443.775156.3
Age at surgery (years)
Mean ± SD40.4 ± 16.136.6 ± 15.543.3 ± 15.9
<55108781.450987.257877.0
≥5524818.67512.817323.0
Sex
Male33024.713823.619225.6
Female100575.344676.455974.4
Histologic subtype
Classical variant89567.131453.758177.4
Follicular variant23217.418531.7476.3
Tall cell variant1188.8213.69712.9
Other variants906.76411.0263.5
Extrathyroidal extension
Present59044.217830.541254.9
Absent74555.840669.533945.1
Multifocality
Yes67050.225944.441154.7
No66549.832555.634045.3
pT
T136928.517631.319326.4
T226620.613824.612817.5
T354942.421237.733746.0
T41108.5366.47410.1
pN
N052739.528048.024732.9
N168351.124541.943858.3
Nx1259.45910.1668.8
pM
M0120595.451893.768796.8
M1584.6356.3233.2
TNM Stage
I 108483.449987.558580.1
II14811.4518.99713.3
III211.640.7172.3
IV-A171.320.4152.1
IV-B302.3142.5162.2
NRAS mutation
Present856.48314.320.3
Absent124493.649885.774699.7
HRAS mutation
Present302.3284.220.3
Absent129897.755395.274599.7
Table 2. Association of clinicopathological characteristics with Cox-2 expression in PTC.
Table 2. Association of clinicopathological characteristics with Cox-2 expression in PTC.
Total Cox-2 PositiveCox-2 Negativep Value
No.%No.%No.%
Total1314 56743.274756.8
Age at surgery (years)
<55107281.641839.065461.0<0.0001
≥5524218.414961.69338.4
Sex
Male32024.414645.617454.40.3049
Female99475.642142.417457.6
Histologic subtype
Classical variant87966.939444.848555.20.1542
Follicular variant22917.48436.714563.3
Tall cell variant1168.85244.86455.2
Other variants906.93741.15358.9
Extrathyroidal extension
Present57944.129150.328849.7<0.0001
Absent73555.927637.645962.4
Multifocality
Yes65850.129544.836355.20.2175
No65649.927241.538458.5
pT
T136528.714038.422561.60.0094
T226520.810138.116461.9
T353842.225347.028553.0
T41068.35350.05350.0
pN
N051643.319437.632262.40.0003
N167556.732548.235051.8
pM
M0118695.450942.967757.10.3575
M1574.62849.12950.9
TNM Stage
I 106983.441538.865461.2<0.0001
II14611.48658.96041.1
III201.61785.0315.0
IV-A171.31058.8741.2
IV-B292.32172.4827.6
BRAF mutation
Present74056.334446.539653.50.0055
Absent57443.722338.935161.1
5-year disease-free survival 69.5 81.7<0.0001
Table 3. Univariate and multivariate analysis of COX-2 expression using the Cox proportional-hazards model.
Table 3. Univariate and multivariate analysis of COX-2 expression using the Cox proportional-hazards model.
Clinical ParametersUnivariateMultivariate
Hazard Ratio (95% CI)p-ValueHazard Ratio (95% CI)p-Value
Age
≥55 years (vs. <55 years)2.82 (2.21–3.57)<0.00012.04 (1.51–2.76)<0.0001
Sex
Male (vs. Female)0.59 (0.47–0.74)<0.00010.66 (0.51–0.85)0.0016
Extrathyroidal extension
Present (vs. Absent)2.89 (2.24–3.77)<0.00012.26 (1.72–2.99)<0.0001
pM
M1 (vs. M0)4.52 (3.12–6.34)<0.00012.84 (1.74–4.63)<0.0001
Stage
IV (vs. I–III)4.40 (2.84–6.52)<0.00010.83 (0.45–1.54)0.5560
COX-2 IHC
Overexpression (vs. low expression)1.66 (1.32–2.08)<0.00011.57 (1.22–2.03)0.0004
Table 4. Univariate and multivariate analysis of COX-2 expression in BRAF-mutant PTC using the Cox proportional-hazards model.
Table 4. Univariate and multivariate analysis of COX-2 expression in BRAF-mutant PTC using the Cox proportional-hazards model.
Clinical ParametersUnivariateMultivariate
Risk Ratio (95% CI)p-ValueRisk Ratio (95% CI)p-Value
Age
≥55 years (vs. <55 years)2.45 (1.84–3.25)<0.00011.75 (1.22–2.48)0.0020
Sex
Male (vs. Female)0.55 (0.42–0.74)<0.00010.52 (0.39–0.72)<0.0001
Extrathyroidal extension
Present (vs. Absent)2.77 (1.96–4.01)<0.00012.11 (1.46–3.10)<0.0001
pM
M1 (vs. M0)4.69 (2.64–7.72)<0.00014.44 (1.61–11.71)0.0036
Stage
IV (vs. I–III)3.15 (1.77–5.16)<0.00010.49 (0.17–1.25)0.1656
COX-2 IHC
Overexpression (vs. low expression)2.10 (1.59–2.79)<0.00012.10 (1.52–2.92)<0.0001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Parvathareddy, S.K.; Siraj, A.K.; Annaiyappanaidu, P.; Al-Sobhi, S.S.; Al-Dayel, F.; Al-Kuraya, K.S. Prognostic Significance of COX-2 Overexpression in BRAF-Mutated Middle Eastern Papillary Thyroid Carcinoma. Int. J. Mol. Sci. 2020, 21, 9498. https://doi.org/10.3390/ijms21249498

AMA Style

Parvathareddy SK, Siraj AK, Annaiyappanaidu P, Al-Sobhi SS, Al-Dayel F, Al-Kuraya KS. Prognostic Significance of COX-2 Overexpression in BRAF-Mutated Middle Eastern Papillary Thyroid Carcinoma. International Journal of Molecular Sciences. 2020; 21(24):9498. https://doi.org/10.3390/ijms21249498

Chicago/Turabian Style

Parvathareddy, Sandeep Kumar, Abdul K. Siraj, Padmanaban Annaiyappanaidu, Saif S. Al-Sobhi, Fouad Al-Dayel, and Khawla S. Al-Kuraya. 2020. "Prognostic Significance of COX-2 Overexpression in BRAF-Mutated Middle Eastern Papillary Thyroid Carcinoma" International Journal of Molecular Sciences 21, no. 24: 9498. https://doi.org/10.3390/ijms21249498

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop