Next Article in Journal
Short Culture of Bovine Hepatocytes Biopsied from Dairy Cows as a Model for Toxicological Studies—CYP 1A1 Activity Response to Zearalenone Treatment
Previous Article in Journal
Therapeutic Efficiency of Proteins Secreted by Glial Progenitor Cells in a Rat Model of Traumatic Brain Injury
Previous Article in Special Issue
Cyclooxygenase Inhibition Alters Proliferative, Migratory, and Invasive Properties of Human Glioblastoma Cells In Vitro
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Cyclooxygenase and Cancer: Fundamental Molecular Investigations

Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, Via E. Orabona 4, 70125 Bari, Italy
Int. J. Mol. Sci. 2023, 24(15), 12342; https://doi.org/10.3390/ijms241512342
Submission received: 28 July 2023 / Accepted: 1 August 2023 / Published: 2 August 2023
(This article belongs to the Special Issue Cyclooxygenase and Cancer: Fundamental Molecular Investigations)
The involvement of prostaglandins in cancer was first observed in human esophageal carcinoma cells, whose invasive and metastatic potential in nude mice was found to be related to PGE2 and PGF2a production [1]. Since then, the evaluation of prostaglandins (in particular PGE2) and cyclooxygenases (in particular COX-2) in distinct neoplastic diseases, along with the investigations of their effects on tumor cell biology properties and tumor progression, have resulted in a very extensive, perhaps overwhelming, scientific literature.
The topic is still of great interest (in the PubMed database, the search “cyclooxygenase and cancer” yields more than 5000 results in the last 10 years), considering its present and potential applications in medical practice. Epidemiological and preclinical evidence indeed suggests that agents with anti-inflammatory COX-targeting activity, such as aspirin and non-steroidal anti-inflammatory drugs (NSAIDs), have the potential to prevent or delay cancer initiation and improve the therapeutic efficacy of cytotoxic agents and radiotherapy, as well as targeted agents and immune checkpoint inhibitors [2,3,4].
The Special Issue “Cyclooxygenase and Cancer: Fundamental Molecular Investigations” of the International Journal of Molecular Sciences includes five research papers, four of which concern the effects of NSAIDs on glioblastoma [5,6,7] and prostate [8] cancer cells, and one which explores a pro-proliferative COX-2 mechanism that does not involve classical prostaglandin receptor signaling [9].
Glioblastoma multiforme (GBM) is the most common and aggressive malignant glioma, accounting for about 50% of all primary malignant brain tumors in adults [10,11]. GBM has a very poor prognosis, with a median survival rate of only 14–17 months with standard treatment including maximal safe resection, adjuvant radiotherapy, and chemotherapy with temozolomide [12]. The GBM-associated increased expression of COX-2 and PGE2 has long been known [13,14], and extensive data suggest that elevated COX-2 activity in tumor cells and the glioblastoma microenvironment [15] can facilitate the acquisition of cancer hallmark capabilities [16] and tumor progression. Therefore, the COX-2-PGE2 axis may be a potential therapeutic target for which mechanistic investigations of effects on cancer cell phenotype can provide a solid rationale.
In their research paper, Ferreira et al. [5] investigated the effects of COX-1 and COX-2 inhibition upon cell proliferation, migration, and invasive properties of human GBM cells in vitro. First, the mRNA expression of PTGS1 (COX-1) and PTGS2 (COX-2) genes was evaluated in differing grades of glioma using the GlioVis software [17] for analysis of brain tumor expression in the TCGA (The Cancer Genome Atlas) and CGGA (Chinese Glioma Genome Atlas) datasets. The expression of both COX-1 and COX-2 was found to be significantly increased in grade IV GBMs in comparison with lower-grade gliomas. Having confirmed the expression of both COX isoforms in the GBM cell lines under study, the effects of non-selective (ibuprofen) or selective (SC560 for COX-1 and NS398 for COX-2) inhibitors were investigated. Overall, the results showed that COX-2 as well as COX-1 activity is important to the normal function of GBM cells in in vitro conditions, thus suggesting a coordinated pathophysiological role of both isoforms [18] in glioblastoma. In this study, the relevance of the PGE2 receptors EP2 and EP4 to the control of GBM cell proliferation and migration was also identified. The concomitant pharmacological inhibition of EP2 and EP4 caused a significant decrease in cell migration, which was not reverted by exogenous PGE2. Finally, a poorly explored area of GBM cell biology, i.e., the control of matrix metalloproteinase (MMP) expression and activity by the prostanoid pathway, was also investigated. Interestingly, MMP2 expression was significantly positively correlated with PTGS1 (COX-1) in GBM tissue. The finding that COX-1 inhibition affects MMP2 protein expression and extracellular matrix-modifying activity of GBM cells in vitro suggests a novel therapeutic target for drug development.
In addition to the current GBM therapy, novel feasible or potential targets have recently emerged [19] and are actively investigated in clinical trials [20]. As reported in the systematic review by Da Silva et al. [21], targeted therapies in GBM clinical trials can be grouped into four categories: targeting the potential for unlimited replication, growth autonomy and migration, cell cycle and apoptosis, and angiogenesis. According to a large body of research, the anticancer activity of NSAIDs depends upon their ability to interfere with tumorigenic signaling pathways [22]. In more detail, both traditional NSAIDs and COX-2 selective inhibitors can modulate many different signaling pathways, such as NF-κB, phosphodiesterases, NSAID-activated genes (NAG-1), peroxisome proliferator-activated receptors (PPAR), the Wnt pathway, cell kinetic effects, the Akt pathway, and pro-resolving mediators [23].
The increased expression of anti-apoptotic Bcl-2 family members has been described in a wide range of solid tumors and in GBM, where the levels of anti-apoptotic BCL-xL and MCL-1 are consistently increased with respect to non-malignant cells and tissues [24].
In the research paper on the effects of the non-selective COX inhibitor indomethacin on GBM cells [6], Chang C. Y. et al. extend their previous observations on indomethacin-induced glioma apoptosis involving the ceramide/protein phosphatase 2A (PP2A)/Akt axis [25]. The authors show that indomethacin can induce oxidative stress and endoplasmic reticulum (ER) stress, as well as Ask1 and p38 activation, in glioma cells. Interestingly, mechanistic investigations further indicated that the oxidative stress/ER stress/Ask1/p38 cascade is an alternative regulator of the PP2A/Akt axis, resulting in Mcl-1 and FLIP downregulation and eventually glioma apoptosis.
Even aspirin can induce tumor cell apoptosis, in most cases involving the anti-apoptotic Mcl-1 protein downregulation [26,27,28,29]. In their second paper in this Special Issue, Chang C. Y. et al. investigate the apoptotic potential of aspirin towards GBM, focusing on the molecular bases of crosstalk between anti-apoptotic and pro-apoptotic Bcl-2 family proteins and underlying apoptotic programs [7]. The authors show that the glioma cell-killing effects of aspirin involve mitochondria-mediated apoptosis accompanied by ER stress, Noxa upregulation, Mcl-1 downregulation, Bax mitochondrial distribution and oligomerization, and caspase activation. Data from genetic and pharmacological studies reveal that the axis of ER stress comprises an apoptotic cascade leading to Noxa upregulation and apoptosis. Importantly, the apoptotic programs and mediators triggered by aspirin in glioma cells were duplicated in tumor-bearing BALB/c nude mice. These findings, along with the previously reported involvement of ER stress in indomethacin-induced Mcl-1 downregulation, support ER stress as a valuable target for intervention in glioma cell apoptosis [30].
Prostate cancer (PCa) is the most common tumor in men, and it has an increasing incidence worldwide due to an aging population and increased detection [31]. Since androgens regulate prostate cancer growth, androgen deprivation therapy (ADT) is the first-line approach for advanced PCa. However, the duration of the ADT response is limited (18–24 months), and most patients progress to the more aggressive castration-resistant prostate cancer (CRPC). Multiple mechanisms for castration resistance have been proposed [32], including immune and inflammatory signaling in both cancer cells and the tumor microenvironment [33].
In their research paper [8], Benelli et al. investigated the effects of the COX-2 inhibitor celecoxib on two androgen-resistant LNCaP sublines (i.e., PDB and MDB), which recapitulate some phenotypic features of PCa evolution to CRPC [34]. PDB cells mimic the clinical condition in which cancer cells are partially exposed to androgens, whereas MDB cells mimic the clinical condition in which cancer cells survive despite a completely hormone-deprived microenvironment. Benelli et al. demonstrate that constitutive activation of ErbB family receptors controlling AKT/AR/GSK3β/P38/NF-κB and hnRNP K signaling nodes emerges in PCa cells during the progression to CRPC. Importantly, bioinformatic analyses of human prostate cancer datasets support the relevance of these pathways in PCa progression. All these molecules are simultaneously modulated by celecoxib treatment. Celecoxib reduced cell growth and induced apoptosis through AKT blockade, cleavage of poly (ADP-ribose) polymerase-1 (PARP-1), and proteasomal degradation of the anti-apoptotic protein Mcl-1. Epidermal growth factor receptor (EGFR), ErbB2, and ErbB3 degradation, and heterogeneous nuclear ribonucleoprotein K (hnRNP K) downregulation further amplified the inhibition of androgen signaling. Moreover, celecoxib reduced the invasive phenotype of CRPC cells by modulating NF-κB activity and reduced tumor growth in mouse xenografts when administered in association with the anti-EGFR receptor antibody cetuximab, thus suggesting a novel therapeutic strategy to hinder signal transduction during CRPC progression.
In the last paper of this Special Issue, Saadi et al. explore an unexpected and surprising biological property of the COX-2 protein, independent from its enzymatic activity. Based on previous data that shows that the continued exposure of COX-2 to arachidonic acid leads to the appearance of lower molecular weight COX-2 fragments [35], in their research paper [9], Saadi et al. confirm the presence of COX-2 immunoreactive fragments in a murine model of glioblastoma as well as in patient-derived colorectal cancer tissues. To provide proof of principle that COX-2 fragments can have biological effects independently from enzymatic activity, a COX-2 mutant that undergoes spontaneous cleavage was used. The K598R point mutation (i.e., arginine for lysine) at the carboxyl-terminus of COX-2 causes the occurrence of several COX-2 immunoreactive fragments in nuclear compartments and significantly enhances cell proliferation. From a mechanistic point of view, transcriptomic analyses show that K598R COX-2 significantly affects the expression of genes involved in RNA metabolism, and subsequent proteomics suggest that it is associated with proteins that regulate mRNA processing. The authors report a similar increase in proliferation by expressing just that catalytic domain of COX-2 (ΔNT-COX-2), which is completely devoid of catalytic activity in the absence of its other domains. Moreover, they show that the ΔNT-COX-2 protein also interacts in the nucleus with β-catenin, a central regulator of gene transcription. Together, these data strongly suggest that the cleavage products of COX-2 can affect cell proliferation through mechanisms that are independent of prostaglandin synthesis. Overall, the results provide a possible explanation for the poor therapeutic efficacy of NSAIDs in tumors where COX-2 expression clearly correlates with a worse prognosis. Additional studies on COX-2 fragments in human tumors, as well as on the mechanisms underlying their production and effects on tumor cell biology, will help to better understand this unexpected biological property of COX-2.

Funding

This research received no external funding.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Botha, J.H.; Robinson, K.M.; Ramchurren, N.; Reddi, K.; Norman, R.J. Human esophageal carcinoma cell lines: Prostaglandin production, biological properties, and behavior in nude mice. J. Natl. Cancer Inst. 1986, 76, 1053. [Google Scholar]
  2. Crusz, S.M.; Balkwill, F.R. Inflammation and cancer: Advances and new agents. Nat. Rev. Clin. Oncol. 2015, 12, 584. [Google Scholar] [CrossRef] [PubMed]
  3. Thiruchenthooran, V.; Sanchez-Lopez, E.; Gliszczyńska, A. Perspectives of the Application of Non-Steroidal Anti-Inflammatory Drugs in Cancer Therapy: Attempts to Overcome Their Unfavorable Side Effects. Cancers 2023, 15, 475. [Google Scholar] [CrossRef] [PubMed]
  4. Ramos-Inza, S.; Ruberte, A.C.; Sanmartin, C.; Sharma, A.K.; Plano, D. NSAIDs: Old Acquaintance in the Pipeline for Cancer Treatment and Prevention—Structural Modulation, Mechanisms of Action, and Bright Future. J. Med. Chem. 2021, 64, 16380. [Google Scholar] [CrossRef] [PubMed]
  5. Ferreira, M.T.; Miyake, J.A.; Gomes, R.N.; Feitoza, F.; Stevannato, P.B.; da Cunha, A.S.; Serachi, F.O.; Panagopoulos, A.T.; Colquhoun, A. Cyclooxygenase Inhibition Alters Proliferative, Migratory, and Invasive Properties of Human Glioblastoma Cells In Vitro. Int. J. Mol. Sci. 2021, 22, 4297. [Google Scholar] [CrossRef]
  6. Chang, C.Y.; Li, J.R.; Wu, C.C.; Wang, J.D.; Liao, S.L.; Chen, W.Y.; Wang, W.Y.; Chen, C.J. Endoplasmic Reticulum Stress Contributes to Indomethacin-Induced Glioma Apoptosis. Int. J. Mol. Sci. 2020, 21, 557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Chang, C.Y.; Pan, P.H.; Li, J.R.; Ou, Y.C.; Wang, J.D.; Liao, S.L.; Chen, W.Y.; Wang, W.Y.; Chen, C.J. Aspirin Induced Glioma Apoptosis through Noxa Upregulation. Int. J. Mol. Sci. 2020, 21, 4219. [Google Scholar] [CrossRef]
  8. Benelli, R.; Barboro, P.; Costa, D.; Astigiano, S.; Barbieri, O.; Capaia, M.; Poggi, A.; Ferrari, N. Multifocal Signal Modulation Therapy by Celecoxib: A Strategy for Managing Castration-Resistant Prostate Cancer. Int. J. Mol. Sci. 2019, 20, 6091. [Google Scholar] [CrossRef] [Green Version]
  9. Saadi, E.; Sood, R.; Dromi, I.; Srouji, R.; Hatoum, O.A.; Tal, S.; Barki-Harrington, L. Limited Proteolysis of Cyclooxygenase-2 Enhances Cell Proliferation. Int. J. Mol. Sci. 2020, 21, 3195. [Google Scholar] [CrossRef]
  10. Grochans, S.; Cybulska, A.M.; Simińska, D.; Korbecki, J.; Kojder, K.; Chlubek, D.; Baranowska-Bosiacka, I. Epidemiology of Glioblastoma Multiforme–Literature Review. Cancers 2022, 14, 2412. [Google Scholar] [CrossRef]
  11. Molinaro, A.M.; Taylor, J.W.; Wiencke, J.K.; Wrensch, M.R. Genetic and molecular epidemiology of adult diffuse glioma. Nat. Rev. Neurol. 2019, 15, 405. [Google Scholar] [CrossRef]
  12. Wen, P.Y.; Weller, M.; Lee, E.Q.; Alexander, B.M.; Barnholtz-Sloan, J.S.; Barthel, F.P.; Batchelor, T.T.; Bindra, R.S.; Chang, S.M.; Chiocca, E.A.; et al. Glioblastoma in adults: A society for neuro-oncology (SNO) and European society of neuro-oncology (EANO) consensus review on current management and future directions. Neuro Oncol. 2020, 22, 1073. [Google Scholar] [CrossRef]
  13. Temel, S.G.; Kahveci, Z. Cyclooxygenase-2 expression in astrocytes and microglia in human oligodendroglioma and astrocytoma. J. Mol. Histol. 2009, 40, 369. [Google Scholar] [CrossRef]
  14. Qiu, J.; Shi, Z.; Jiang, J. Cyclooxygenase-2 in glioblastoma multiforme. Drug Discov. Today 2017, 22, 148. [Google Scholar] [CrossRef] [Green Version]
  15. Dean, P.T.; Hooks, S.B. Pleiotropic effects of the COX-2/PGE2 axis in the glioblastoma tumor microenvironment. Front. Oncol. 2023, 12, 1116014. [Google Scholar] [CrossRef]
  16. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31. [Google Scholar] [CrossRef]
  17. Bowman, R.L.; Wang, Q.; Carro, A.; Verhaak, R.G.W.; Squatrito, M. GlioVis data portal for visualization and analysis of brain tumor expression datasets. Neuro Oncol. 2017, 19, 131. [Google Scholar] [CrossRef] [Green Version]
  18. Pannunzio, A.; Coluccia, M. Cyclooxygenase-1 (COX-1) and COX-1 Inhibitors in Cancer: A Review of Oncology and Medicinal Chemistry Literature. Pharmaceuticals 2018, 11, 101. [Google Scholar] [CrossRef] [Green Version]
  19. Yang, K.; Wu, Z.; Zhang, H.; Zhang, N.; Wu, W.; Wang, Z.; Dai, Z.; Zhang, X.; Zhang, L.; Peng, Y.; et al. Glioma Targeted Therapy: Insight into Future of Molecular Approaches. Mol. Cancer 2022, 21, 39. [Google Scholar] [CrossRef]
  20. Muzyka, L.; Goff, N.K.; Choudhary, N.; Koltz, M.T. Systematic Review of Molecular Targeted Therapies for Adult-Type Diffuse Glioma: An Analysis of Clinical and Laboratory Studies. Int. J. Mol. Sci. 2023, 24, 10456. [Google Scholar] [CrossRef]
  21. Cruz Da Silva, E.; Mercier, M.-C.; Etienne-Selloum, N.; Dontenwill, M.; Choulier, L. A Systematic Review of Glioblastoma-Targeted Therapies in Phases II, III, IV Clinical Trials. Cancers 2021, 13, 1795. [Google Scholar] [CrossRef] [PubMed]
  22. Gurpinar, E.; Grizzle, W.E.; Piazza, G.A. NSAIDs inhibit tumorigenesis, but how? Clin. Cancer Res. 2014, 20, 1104. [Google Scholar] [CrossRef] [Green Version]
  23. Kolawole, O.R.; Kashfi, K. NSAIDs and Cancer Resolution: New Paradigms beyond Cyclooxygenase. Int. J. Mol. Sci. 2022, 23, 1432. [Google Scholar] [CrossRef] [PubMed]
  24. Koessinger, A.L.; Cloix, C.; Koessinger, D.; Heiland, D.H.; Bock, F.J.; Strathdee, K.; Kinch, K.; Martínez-Escardó, L.; Paul, N.R.; Nixon, C.; et al. Increased apoptotic sensitivity of glioblastoma enables therapeutic targeting by BH3-mimetics. Cell Death Differ. 2022, 29, 2089. [Google Scholar] [CrossRef]
  25. Chang, C.Y.; Li, J.R.; Wu, C.C.; Wang, J.D.; Yang, C.P.; Chen, W.Y.; Wang, W.Y.; Chen, C.J. Indomethacin induced glioma apoptosis involving ceramide signals. Exp. Cell. Res. 2018, 365, 66. [Google Scholar] [CrossRef]
  26. Iglesias-Serret, D.; Piqué, M.; Barragán, M.; Cosialls, A.M.; Santidrián, A.F.; González-Gironès, D.M.; Coll-Mulet, L.; de Frias, M.; Pons, G.; Gil, J. Aspirin induces apoptosis in human leukemia cells independently of NF-κB and MAPKs through alteration of the Mcl-1/Noxa balance. Apoptosis 2010, 15, 219. [Google Scholar] [CrossRef] [PubMed]
  27. Li, G.; Zhang, S.; Fang, H.; Yan, B.; Zhao, Y.; Feng, L.; Ma, X.; Ye, X. Aspirin overcomes Navitoclax-resistance in hepatocellular carcinoma cells through suppression of Mcl-1. Biochem. Biophys. Res. Commun. 2013, 434, 809. [Google Scholar] [CrossRef]
  28. Park, I.S.; Jo, J.R.; Hong, H.; Nam, K.Y.; Kim, J.B.; Hwang, S.H.; Choi, M.-S.; Ryu, N.-H.; Jang, H.-J.; Lee, S.-H.; et al. Aspirin induces apoptosis in YD-8 human oral squamous carcinoma cells through activation of caspases, down-regulation of Mcl-1, and inactivation of ERK-1/2 and AKT. Toxicol. Vitr. 2010, 24, 713. [Google Scholar] [CrossRef]
  29. Yue, W.; Zheng, X.; Lin, Y.; Yang, C.S.; Xu, Q.; Carpizo, D.; Huang, H.; DiPaola, R.S.; Tan, X.L. Metformin combined with aspirin significantly inhibit pancreatic cancer cell growth in vitro and in vivo by suppressing anti-apoptotic proteins Mcl-1 and Bcl-2. Oncotarget 2015, 28, 21208. [Google Scholar] [CrossRef] [Green Version]
  30. Obacz, J.; Avril, T.; Le Reste, P.-J.; Urra, H.; Quillien, V.; Hetz, C.; Chevet, E. Endoplasmic reticulum proteostasis in glioblastoma—From molecular mechanisms to therapeutic perspectives. Sci. Signal. 2017, 10, 470. [Google Scholar] [CrossRef] [Green Version]
  31. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209. [Google Scholar] [CrossRef]
  32. Karantanos, T.; Corn, P.G.; Thompson, T.C. Prostate cancer progression after androgen deprivation therapy: Mechanisms of castrate resistance and novel therapeutic approaches. Oncogene 2013, 32, 5501. [Google Scholar] [CrossRef]
  33. Zhong, S.; Huang, C.; Chen, Z.; Chen, Z.; Luo, J.-L. Targeting Inflammatory Signaling in Prostate Cancer Castration Resistance. J. Clin. Med. 2021, 10, 5000. [Google Scholar] [CrossRef]
  34. Ferrari, N.; Granata, I.; Capaia, M.; Piccirillo, M.; Guarracino, M.R.; Venè, R.; Brizzolara, A.; Petretto, A.; Inglese, E.; Morini, M.; et al. Adaptive phenotype drives resistance to androgen deprivation therapy in prostate cancer. Cell Commun. Signal. 2017, 15, 51. [Google Scholar] [CrossRef] [Green Version]
  35. Saadi, E.; Tal, S.; Barki-Harrington, L. Substrate-inactivated cyclooxygenase-2 is disposed of by exosomes through the ER-Golgi pathway. Biochem. J. 2018, 475, 3141. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Coluccia, M. Cyclooxygenase and Cancer: Fundamental Molecular Investigations. Int. J. Mol. Sci. 2023, 24, 12342. https://doi.org/10.3390/ijms241512342

AMA Style

Coluccia M. Cyclooxygenase and Cancer: Fundamental Molecular Investigations. International Journal of Molecular Sciences. 2023; 24(15):12342. https://doi.org/10.3390/ijms241512342

Chicago/Turabian Style

Coluccia, Mauro. 2023. "Cyclooxygenase and Cancer: Fundamental Molecular Investigations" International Journal of Molecular Sciences 24, no. 15: 12342. https://doi.org/10.3390/ijms241512342

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