Abstract
Reactive Oxygen Species Modulator 1 (ROMO1) was first discovered in 2006, and its structural characteristics were elucidated by Lee et al. in 2018. This novel protein resides in the inner mitochondrial membrane and exerts control over the production of reactive oxygen species (ROS) by modulating membrane potential and permeability. ROS, in turn, plays a multifaceted role in cancer progression: at low concentrations, it serves as a critical player in cell signaling, influencing tumor suppression and immune system maintenance; at moderate concentrations, it promotes cancer progression, while high concentrations induce apoptosis. ROMO1, as a key regulator of intracellular ROS, significantly impacts cancer cell invasion and growth. Existing literature demonstrates that overexpression of ROMO1 is strongly associated with lymph node metastasis and a dismal prognosis in cancer patients, making it a promising prognostic factor for solid malignant tumors. ROMO1 can be investigated by various methods including immunohistochemistry (IHC) which is one very suitable method in our opinion.
Introduction
Cancer continues to be a leading cause of mortality worldwide, particularly among aging populations. In 2019, there were approximately 23.6 million new cancer cases and 10.0 million cancer-related deaths globally [1]. In 2020, lung cancer ranked as the most common malignancy among men, accounting for 15.4 % of newly diagnosed cases, followed by prostate and colorectal cancers [2]. Among women in 2020, breast cancer took the lead, representing 25.8 % of all new cases, followed by colorectal, lung, and cervical cancers [2].
When it comes to both preventing and treating tumors, the immune system is crucial [3]. Therapies focused on the immune system, such as cancer vaccines, target specific antigens expressed on cancer cell surfaces while sparing healthy cells from drug-related toxicity [4].
Due to tumor diversity and the fact that cancer vaccines target only a limited number of antigens, cancer cells that do not express these antigens or a group of cancer cells with mutations that alter surface antigens can evade immune control and create new tumor populations that can resist treatment with vaccines encoding the same tumor-associated antigens [5, 6]. Therefore, the discovery of new potential biomarkers for cancer development and further treatment is of great importance.
Intratumoral heterogeneity is a hallmark of cancer progression, with malignancies becoming increasingly diverse as the disease advances [7]. Because cancer is a progressive disease, throughout its course, malignancies typically become more diverse. As a result of this heterogeneity, the majority of the tumor may contain a variety of cells with distinct molecular signatures and varying degrees of sensitivity to treatment [7]. Increasing reactive oxygen species (ROS) levels have a significant impact on heterogeneity. Under low concentrations, reactive oxygen species may function as signaling molecules that promote tumorigenesis and heterogeneity, whereas under high concentrations, these species may function as cancer modulators due to their deleterious, genotoxic, or proapoptotic effect on cancer cells [8]. This effect is due to ROS’s capacity to induce genetic and epigenetic alterations in DNA structure [8]. One way to produce ROS in the mitochondria is through the Reactive Oxygen Species Modulator 1 (ROMO1) [9].
Reactive oxygen species modulator 1 – what is it?
Reactive Oxygen Species Modulator 1 (ROMO1), identified in 2006, has been the subject of extensive research regarding its role in ROS production. This protein resides within the inner mitochondrial membrane and generates ROS via Complex III in the electron transport chain. Common ROS types produced include hydrogen peroxide, the hydroxyl radical, and the superoxide anion, generated through the partial reduction of atmospheric oxygen [3].
The structural characteristics of ROMO1 were reported by Lee et al. in 2018 [10]. It consists of two transmembrane domains (TMDs) linked by a basic loop, each containing an alpha helix. TMD2 comprises polar amino acids (K58, T59, Q62, S63, T66, and T69), while TMD1 includes a hydrophobic alpha helix. By forming homo-oligomers in the inner mitochondrial membrane, the amphipathic TMD facilitates the creation of non-selective cation channels in cells. As per Lee et al. [10], ROMO1 functions as a non-selective cation channel with properties akin to viroporins, enabling it to alter the membrane potential and permeability of mitochondrial membranes.
ROMO1 has a notable impact on cancer cell invasion and proliferation, contributing to an ongoing inflammatory response. Multiple studies have examined ROMO1’s role in cancer progression and its potential as a prognostic factor in various cancer types [9]. It has been associated with lymphatic metastasis and a less favorable prognosis [11].
ROMO 1 – methods for examination
ROMO1 can be examined using many methods, however, the most common ones include-RNA sequencing, used to measure gene expression, ELISA and Western blotting, PCR and immunohistochemistry (IHC) (Figure 1).

Methods for ROMO1 analysis.
The novel protein is a critical factor in cancer, impacting malignancy through its influence on redox balance, mitochondrial dynamics, and apoptosis resistance.
Redox Regulation: ROMO1 generates ROS, which, at high levels due to ROMO1 overactivity, can damage DNA, proteins, and lipids, promoting genetic instability and cancer progression [12]. Mitochondrial Dynamics: ROMO1’s location in mitochondria affects their function and structure [13]. Dysregulation by ROMO1 can lead to energy problems, increased ROS production, and resistance to cell death, contributing to cancer cell survival. Apoptosis Resistance: ROMO1 disrupts the balance of apoptosis-regulating factors within mitochondria, making cancer cells less responsive to apoptotic signals, thereby facilitating their survival and growth despite treatment [14].
A 2022 study by Wang et al. [3] tested the importance of ROMO1 as a biomarker for prostate cancer progression using information from the TCGA data set and various model analyses. This data comes from RNA sequencing. They concluded that ROMO1 is a significant gene strongly linked with the microenvironment of prostate cancer tumors, and the essential signaling pathways implicated were identified. In light of this, they proposed that ROMO1 may serve as a biomarker and therapeutic target for prostate cancer. Another study in 2020 examined ROMO1’s role in bladder cancer and oxidative stress using quantitative real-time PCR [15].
ROMO1 expression was significantly correlated with tumor size and histopathological grading. The expression level of the ROMO1 gene was measured in malignant and adjacent healthy tissues and found to be significantly higher in bladder cancerous tissues than in adjacent healthy tissues [15]. ELISA method and Western blotting were used to measure ROMO1 in patients with non-small cellular lung cancer (NSCLC), where the team tested the potential of ROMO1 as a diagnostic biomarker [16]. They concluded that the serum levels or ROMO1 correlate positively with ROMO1 in cancerous tissues. In addition to that, the serum ROMO1 levels of NSCLC patients were substantially higher than those of healthy individuals or those with benign lung diseases [16]. The different methods of detection of ROMO1 are shown in Table 1.
Possible methods for examining ROMO1.
Experimental method | Purpose and application | Types of cancer tested |
---|---|---|
Immunohistochemistry (IHC) | Allows identification of the protein and semiquantitative histologic evaluation of its expression. | Cervical [17], lung [18], [19], [20], [21], [22], hepatocellular [23], colorectal [11] |
Polymerase Chain Reaction (PCR) | Amplify and quantify ROMO1 gene expression at the transcription level. | Bladder [15], hepatocellular [23], gastric [24], glioblastoma [25, 26] |
Western Blotting | Detect and quantify ROMO1 protein levels in cell lysates and tissues. | Lung [16], colorectal [27] |
Enzyme-Linked Immunosorbent Assay (ELISA) | Quantify ROMO1 protein concentration in biological samples. | Lung [16], ovarian [28], endometrial [28] |
Gene silencing – siRNA, shRNA | Suppress ROMO1 gene expression to study its effects. | Colorectal [27], prostate [20], glioblastoma [25, 26] |
ROS detection assays | Measure reactive oxygen species (ROS) levels with altered ROMO1 expression. | Glioblastoma [25, 26], colorectal [27] |
Functional assays | Study ROMO1’s role in apoptosis, cell migration, proliferation, etc. | Colorectal [27], gastric [24] |
ROMO1 and immunohistochemistry
Immunohistochemistry (IHC) offers several compelling reasons and advantages for detecting ROMO1 as a potential diagnostic biomarker and therapeutic target in cancer research:
Tissue Localization: IHC allows precise localization of ROMO1 within tissue samples. This spatial information is very important for understanding ROMO1’s role in cancer development and progression. It helps us identify specific cellular and subcellular locations where ROMO1 is overexpressed or dysregulated.
Protein Expression Profiling: ROMO1 protein expression levels in tumor tissues can be evaluated by IHC. The associations between ROMO1 expression and disease severity, prognosis, and therapeutic response can be determined using this information.
Validation of Biomarker Potential: IHC is also a reliable technique for confirming ROMO1’s potential as a diagnostic biomarker. Researchers can evaluate the specificity and sensitivity of ROMO1 as a diagnostic biomarker by contrasting its expression in healthy and malignant tissues.
Patient Stratification: Patient classification based on ROMO1 expression levels can benefit from IHC. By enabling tailored treatment plans, this information can assist clinicians in identifying patients who are more likely to benefit from ROMO1-targeted medicines.
Monitoring Treatment Response: IHC can be used to monitor changes in ROMO1 expression following treatment. Decreased ROMO1 levels post-treatment may indicate therapeutic efficacy, providing a means to assess treatment response.
Clinical Translation: There are several studies that have tested the expression of ROMO1 using IHC and in all of them the results are similar-overexpression of the protein leads to poorer prognosis.
ROMO1 and colorectal cancer (CRC)
A study carried out by Kim et al. [11] provided information that ROMO1 can be used as a prognostic marker for CRC. The high ROMO1 expression in cancer tissues was substantially associated with early recurrence and poor survival in patients who underwent curative resection. ROMO1 expression was substantially associated with the survival of the entire cohort when patients with stage IV CRC were included. In addition, they discovered that ROMO1 overexpression was substantially associated with increased lymph node ratio and lymphatic invasion of primary tumors, as well as decreased survival in patients with CRC.
ROMO1 and non-small cell lung cancer (NSCLC)
Several studies have employed immunohistochemistry to investigate ROMO1 expression in lung cancer patients. A 2020 study found that ROMO1 levels increase significantly with advanced disease stages [18, 22]. Overexpression of the protein is also associated with lymph node metastasis, progression, and poor prognosis [18, 22, 29]. Another study concluded that the overexpression of ROMO1 is associated with worse survival outcomes in patients who received surgical resection, conventional fractionated radiotherapy, and chemotherapy [21]. Furthermore, ROMO1 overexpression was associated with a poor response to platinum-based chemotherapy and reduced survival in advanced NSCLC patients [19]. Kwock et al. [20] demonstrated that ROMO1 overexpression was linked with poor response to treatment and short survival in patients treated with EGFR-tyrosine kinase inhibitors. Kong et al. tested the significance of ROMO1 in NSCLC stage III and concluded that the overexpression of the marker leads to worse survival and early metastasis in local lymph nodes [30].
ROMO1 and hepatocellular carcinoma (HCC)
In a 2021 study of HCC patients, ROMO1 expression was detected using various methods, including IHC [23]. The results demonstrated that increased ROMO1 expression was associated with vascular invasion, larger tumor size, worse dedifferentiation, and poorer survival.
ROMO1 and cervical cancer (CC)
In 2022, we conducted an immunohistochemical study to assess the expression of ROMO1 at different stages of the disease in cervical cancer (CC). To our knowledge, this marks the first study to employ immunohistochemistry to investigate ROMO1’s role in CC progression. Our retrospective study encompassed 75 patients. Since there isn’t an established scoring system for ROMO1, we utilized the H-score, Allred score, and combined score to evaluate and analyze our findings. Our results indicate that ROMO1 levels are significantly higher in early-stage tumors compared to advanced-stage tumors [17]. Among the scoring systems, the H-score yielded the most significant p-value, making it the preferred method for assessing ROMO1 expression via immunohistochemistry. Furthermore, the values of the combined score for the N stage provide additional evidence that ROMO1 plays a role in cancer progression and metastasis.
We observed significantly elevated ROMO1 levels in FIGO1 stage compared to FIGO2 and FIGO3, as indicated by both scoring methods (H-score: FIGO1 vs. FIGO2, p=0.00012; FIGO1 vs. FIGO3, p=0.0008; Allred score: FIGO1 vs. FIGO2, p=0.0029; FIGO1 vs. FIGO3, p=0.012). Moreover, a statistically significant difference was noted based on the H-score between patients with and without metastatic lymph nodes (p=0.033). These findings shed light on the potential significance of ROMO1 in CC progression and its possible role as a prognostic marker.
In Table 2 we summarize the clinical significance of ROMO1 expression (Table 2).
Clinical significance of ROMO1 expression.
Cancer type | Expression | Results/prognosis |
---|---|---|
Non-small cell lung cancer [18], [19], [20], [21], [22] | High expression in the advanced stage | Lymph node metastasis; lymphatic invasion; poor prognosis; poor response to treatment; potential predictive and prognostic biomarker |
Colorectal cancer [11] | High expression in the advanced stage | High lymph node ratio; prognostic biomarker |
Hepatocellular cancer [23] | High expression | Vascular invasion; bigger tumor size; worse differentiation and worse survival; potential predictive and prognostic biomarker |
Cervical cancer [17] | High expression in the early stage | Metastatic lymph nodes |
Discussion
Gynecological cancers are a significant public health concern, and early detection through screening is crucial for effective management. Nowadays the only gynecological cancer that is screenable is carcinoma of the cervix (CC) – there is no such option for endometrial, ovarian, and vulvar cancer [31]. Even though a screening programme is available for CC, in 2020 there was a 5.7 % increase in new cases and an 8.9 % increase in morbidity compared to 2018 [32]. That is why new prognostic and predictive biomarkers are required by clinicians.
Bearing in mind that Lee et al. [10] described the structure of ROMO1 very recently – in 2018, this novel protein shows great perspective as a potential biomarker, showing different results according to the type of cancer being studied. The methods by which it can be examined are different, but we decided to use IHC for the following reasons. IHC can be highly specific, as it relies on antibodies that are designed to target a particular protein of interest. This specificity ensures that we are detecting ROMO1 and no other proteins with similar properties. IHC is also very compatible with different tissue samples. It can be used both on paraffin-embedded or frozen sections, allowing for flexibility in sample preparation. This method is suitable for use in clinical practice because it can assess protein expression in patient samples. If ROMO1 is implicated in a particular disease or condition, IHC can be employed for diagnostic or prognostic purposes. Of course, a very alluring reason to use IHC is its price-it is much cheaper than other possible methods, such as RNA sequencing for example.
Different studies [18, 22, 29, 30] using IHC show that ROMO1 expression levels are higher in advanced stages, while in CC it is exactly the opposite – its expression is higher in the early stages of the disease – it may be owing to the viral component in the carcinogenesis of CC.
ROMO1 is responsible for a rise in intracellular ROS levels. We, however, have not tested what role this biomarker plays in the tumor microenvironment. In the study carried out in 2022, the authors concluded that ROMO1 has a high degree of infiltration, a bad prognosis, and a strong connection with CD8+T cells, all of which are associated with tumor immune infiltration [3]. ROMO1-induced ROS accumulation can promote angiogenesis [14], facilitating tumor vascularization and nutrient supply. Additionally, elevated ROS levels may suppress anti-tumor immune responses, allowing cancer cells to evade immune surveillance [33]. Thus, ROMO1’s impact on the tumor microenvironment highlights its potential as a crucial mediator of cancer progression and therapeutic resistance.
Conclusions
ROMO1 regulates redox, mitochondrial dynamics, and apoptosis. Its increasing significance in cancer biology is fascinating and therapeutically significant. It affects ROS levels, mitochondrial dynamics, and the tumor microenvironment. We strongly believe that immunohistochemistry is a promising method to test for ROMO1 expression, as it is inexpensive, accessible, and straightforward to use.
The potential of ROMO1 as both a diagnostic biomarker and therapeutic target opens new avenues for personalized cancer treatment. However, challenges persist. An in-depth understanding of ROMO1’s molecular control and interactions with other cellular components is essential for targeted therapeutics. Clinical translation necessitates the study of ROMO1’s context-dependent effects across various cancer types and patient cohorts. Establishing ROMO1 as a diagnostic biomarker and therapeutic target requires robust preclinical and clinical investigations.
-
Research ethics: Not applicable.
-
Informed consent: Not applicable.
-
Author contributions: All authors have accepted responsibility for the entire content of this manuscript and approved its submission.
-
Competing interests: Authors state no conflict of interest.
-
Research funding: None declared.
-
Data availability: The authors declare that all related data are available concerning researchers by the corresponding author’s email.
References
1. Kocarnik, JM, Compton, K, Dean, FE, Fu, W, Gaw, BL, Harvey, JD, et al.. Cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life years for 29 cancer groups from 2010 to 2019: a systematic analysis for the global burden of disease study 2019. JAMA Oncol 2022;8:420–44. https://doi.org/10.1001/jamaoncol.2021.6987.Suche in Google Scholar PubMed PubMed Central
2. World Cancer Research Fund International. Worldwide cancer data [Online]. https://www.wcrf.org/cancer-trends/worldwide-cancer-data/ [Accessed 20 Jul 2023].Suche in Google Scholar
3. Wang, L, Liu, X, Liu, Z, Wang, Y, Fan, M, Yin, J, et al.. Network models of prostate cancer immune microenvironments identify ROMO1 as a heterogeneity and prognostic marker. Sci Rep 2022;12:192. https://doi.org/10.1038/s41598-021-03946-w.Suche in Google Scholar PubMed PubMed Central
4. Perez, CR, De Palma, M. Engineering dendritic cell vaccines to improve cancer immunotherapy. Nat Commun 2019;10:5408. https://doi.org/10.1038/s41467-019-13368-y.Suche in Google Scholar PubMed PubMed Central
5. McLaughlin, J, Han, G, Schalper, KA, Carvajal-Hausdorf, D, Pelekanou, V, Rehman, J, et al.. Quantitative assessment of the heterogeneity of PD-L1 expression in non-small-cell lung cancer. JAMA Oncol 2016;2:46–54. https://doi.org/10.1001/jamaoncol.2015.3638.Suche in Google Scholar PubMed PubMed Central
6. Ebert, LM, Yu, W, Gargett, T, Brown, MP. Logic-gated approaches to extend the utility of chimeric antigen receptor T-cell technology. Biochem Soc Trans 2018;46:391–401. https://doi.org/10.1042/bst20170178.Suche in Google Scholar PubMed
7. Dagogo-Jack, I, Shaw, AT. Tumour heterogeneity and resistance to cancer therapies. Nat Rev Clin Oncol 2018;15:81–94. https://doi.org/10.1038/nrclinonc.2017.166.Suche in Google Scholar PubMed
8. de Sá Junior, PL, Câmara, DAD, Porcacchia, AS, Fonseca, PMM, Jorge, SD, Araldi, RP, et al.. The roles of ROS in cancer heterogeneity and therapy. Oxid Med Cell Longev 2017;2017:2467940–12. https://doi.org/10.1155/2017/2467940.Suche in Google Scholar PubMed PubMed Central
9. National Cancer Institute. The cancer genome atlas program [Online]. https://www.cancer.gov/about-nci/organization/ccg/research/structural-genomics/tcga [Accessed on 10 Jul 2023].Suche in Google Scholar
10. Lee, GY, You, DG, Lee, HR, Hwang, SW, Lee, CJ, Yoo, YD. ROMO1 is a mitochondrial nonselective cation channel with viroporin-like characteristics. J Cell Biol 2018;217:2059–71. https://doi.org/10.1083/jcb.201709001.Suche in Google Scholar PubMed PubMed Central
11. Kim, HJ, Jo, MJ, Kim, BR, Kim, JL, Jeong, YA, Na, YJ, et al.. Reactive oxygen species modulator-1 (ROMO1) predicts unfavorable prognosis in colorectal cancer patients. PLoS One 2017;12:e0176834. https://doi.org/10.1371/journal.pone.0176834.Suche in Google Scholar PubMed PubMed Central
12. Juan, CA, Pérez de la Lastra, JM, Plou, FJ, Pérez-Lebeña, E. The chemistry of reactive oxygen species (ROS) revisited: outlining their role in biological macromolecules (DNA, lipids and proteins) and induced pathologies. Int J Mol Sci 2021;22:4642. https://doi.org/10.3390/ijms22094642.Suche in Google Scholar PubMed PubMed Central
13. Norton, M, Ng, AC, Baird, S, Dumoulin, A, Shutt, T, Mah, N, et al.. ROMO1 is an essential redox-dependent regulator of mitochondrial dynamics. Sci Signal 2014;7:ra10. https://doi.org/10.1126/scisignal.2004374.Suche in Google Scholar PubMed
14. Kennel, KB, Greten, FR. Immune cell-produced ROS and their impact on tumor growth and metastasis. Redox Biol 2021;42:101891. https://doi.org/10.1016/j.redox.2021.101891.Suche in Google Scholar PubMed PubMed Central
15. Ghasemi, H, Amini, MA, Pegah, A, Azizi, E, Tayebinia, H, Khanverdilou, S, et al.. Overexpression of reactive oxygen species modulator 1 is associated with advanced grades of bladder cancer. Mol Biol Rep 2020;47:6497–505. https://doi.org/10.1007/s11033-020-05702-1.Suche in Google Scholar PubMed PubMed Central
16. Lee, SH, Lee, JS, Lee, EJ, Min, KH, Hur, GY, Lee, SH, et al.. Serum reactive oxygen species modulator 1 (ROMO1) as a potential diagnostic biomarker for non-small cell lung cancer. Lung Cancer 2014;85:175–81. https://doi.org/10.1016/j.lungcan.2014.05.023.Suche in Google Scholar PubMed
17. Tsoneva, E, Dimitrova, PD, Metodiev, M, Shivarov, V, Vasileva-Slaveva, M, Yordanov, A. The effects of ROMO1 on cervical cancer progression. Pathol Res Pract 2023;248:154561. https://doi.org/10.1016/j.prp.2023.154561.Suche in Google Scholar PubMed
18. Kim, T, Cha, YJ, Park, JH, Kim, A, Choi, YJ, Park, HJ. Reactive oxygen species modulator 1 expression predicts lymph node metastasis and survival in early-stage non-small cell lung cancer. PLoS One 2020;15:e0239670. https://doi.org/10.1371/journal.pone.0239670.Suche in Google Scholar PubMed PubMed Central
19. Lee, SH, Choi, SI, Lee, JS, Kim, CH, Jung, WJ, Lee, EJ, et al.. Reactive oxygen species modulator 1 (ROMO1) predicts poor outcomes in advanced non-small cell lung cancer patients treated with platinum-based chemotherapy. Cancer Res Treat 2017;49:141–9. https://doi.org/10.4143/crt.2016.133.Suche in Google Scholar PubMed PubMed Central
20. Kwack, WG, Sung, JY, Lee, SH. Overexpression of reactive oxygen species modulator 1 predicts unfavorable clinical outcome in EGFR-mutant lung adenocarcinomas treated with targeted therapy. Front Oncol 2021;11:770230. https://doi.org/10.3389/fonc.2021.770230.Suche in Google Scholar PubMed PubMed Central
21. Kong, M, Sung, JY, Lee, SH. Reactive oxygen species modulator 1 is associated with poor survival in patients with non-small cell lung cancer after stereotactic fractionated radiosurgery: a retrospective pilot study. Onco Targets Ther 2020;13:8173–80. https://doi.org/10.2147/ott.s266344.Suche in Google Scholar
22. Kim, HJ, Jo, MJ, Kim, BR, Kim, JL, Jeong, YA, Na, YJ, et al.. Overexpression of ROMO1 is an unfavorable prognostic biomarker and a predictor of lymphatic metastasis in non-small cell lung cancer patients. Onco Targets Ther 2018;11:4233–46. https://doi.org/10.2147/OTT.S161587.Suche in Google Scholar PubMed PubMed Central
23. Chung, JS, Park, S, Park, SH, Park, ER, Cha, PH, Kim, BY, et al.. Overexpression of Romo1 promotes production of reactive oxygen species and invasiveness of hepatic tumor cells. Gastroenterology 2012;143:1084–94.e7. https://doi.org/10.1053/j.gastro.2012.06.038.Suche in Google Scholar PubMed
24. Amini, MA, Karimi, J, Khodadadi, I, Tavilani, H, Talebi, SS, Afshar, B. Overexpression of ROMO1 and OMA1 are potentially biomarkers and predict unfavorable prognosis in gastric cancer. J Gastrointest Cancer 2020;51:939–46. https://doi.org/10.1007/s12029-019-00330-w.Suche in Google Scholar PubMed
25. Yu, MO, Song, NH, Park, KJ, Park, DH, Kim, SH, Chae, YS, et al.. Romo1 is associated with ROS production and cellular growth in human gliomas. J Neuro Oncol 2015;121:73–81. https://doi.org/10.1007/s11060-014-1608-x.Suche in Google Scholar PubMed
26. Sun, G, Cao, Y, Qian, C, Wan, Z, Zhu, J, Guo, J, et al.. Romo1 is involved in the immune response of glioblastoma by regulating the function of macrophages. Aging (Albany NY) 2020;12:1114–27. https://doi.org/10.18632/aging.102648.Suche in Google Scholar PubMed PubMed Central
27. Jo, MJ, Kim, BG, Park, SH, Kim, HJ, Jeong, S, Kim, BR, et al.. ROMO1 inhibition induces TRAIL-mediated apoptosis in colorectal cancer. Cancers (Basel) 2020;12:2358. https://doi.org/10.3390/cancers12092358.Suche in Google Scholar PubMed PubMed Central
28. Turan, H, Vitale, SG, Kahramanoglu, I, Della Corte, L, Giampaolino, P, Azemi, A, et al.. Diagnostic and prognostic role of TFF3, ROMO-1, NF-кB and SFRP4 as biomarkers for endometrial and ovarian cancers: a prospective observational translational study. Arch Gynecol Obstet 2022;306:2105–14. https://doi.org/10.1007/s00404-022-06563-8.Suche in Google Scholar PubMed PubMed Central
29. Lee, SH, Min, JW, Lee, JS, Kim, CH, Yoo, YD, Lee, EJ, et al.. Reactive oxygen species modulator 1 (ROMO1) overexpression is an independent predictor of poor survival in NSCLC patients who undergo surgical resection. Lung Cancer 2015;87:45–52. https://doi.org/10.1016/j.lungcan.2014.11.004.Suche in Google Scholar PubMed
30. Kong, M, Sung, JY, Lee, SH. Reactive oxygen species modulator 1 as an adverse prognostic marker in stage III non-small cell lung cancer treated with radiotherapy: a retrospective pilot study. Onco Targets Ther 2019;12:8263–73. https://doi.org/10.2147/OTT.S217514.Suche in Google Scholar PubMed PubMed Central
31. Golia D’Augè, T, Giannini, A, Bogani, G, Di Dio, C, Laganà, AS, Di Donato, V, et al.. Prevention, screening, treatment and follow-up of gynecological cancers: state of art and future perspectives. Clin Exp Obstet Gynecol 2023;50:1–6. https://doi.org/10.31083/j.ceog5008160.Suche in Google Scholar
32. Arbyn, M, Weiderpass, E, Bruni, L, de Sanjosé, S, Saraiya, M, Ferlay, J, et al.. Estimates of incidence and mortality of cervical cancer in 2018: a worldwide analysis. Lancet Glob Health 2020;8:e191–203. https://doi.org/10.1016/S2214-109X(19)30482-6.Suche in Google Scholar PubMed PubMed Central
33. Liu, R, Peng, L, Zhou, L, Huang, Z, Zhou, C, Huang, C. Oxidative stress in cancer immunotherapy: molecular mechanisms and potential applications. Antioxidants (Basel) 2022;11:853. https://doi.org/10.3390/antiox11050853.Suche in Google Scholar PubMed PubMed Central
© 2023 the author(s), published by De Gruyter, Berlin/Boston
This work is licensed under the Creative Commons Attribution 4.0 International License.
Artikel in diesem Heft
- Frontmatter
- Review Articles
- Mitochondrial thermogenesis in cancer cells
- Application of indocyanine green in the management of oral cancer: a literature review
- Long non-coding RNA, FOXP4-AS1, acts as a novel biomarker of cancers
- The role of synthetic peptides derived from bovine lactoferricin against breast cancer cell lines: a mini-review
- Single cell RNA sequencing – a valuable tool for cancer immunotherapy: a mini review
- Research Articles
- Global patterns and temporal trends in ovarian cancer morbidity, mortality, and burden from 1990 to 2019
- The association between NRF2 transcriptional gene dysregulation and IDH mutation in Grade 4 astrocytoma
- More than just a KRAS inhibitor: DCAI abrogates the self-renewal of pancreatic cancer stem cells in vitro
- DUSP1 promotes pancreatic cancer cell proliferation and invasion by upregulating nephronectin expression
- IMMT promotes hepatocellular carcinoma formation via PI3K/AKT/mTOR pathway
- MiR-100-5p transfected MSCs-derived exosomes can suppress NSCLC progression via PI3K-AKT-mTOR
- Inhibitory function of CDK12i combined with WEE1i on castration-resistant prostate cancer cells in vitro and in vivo
- Prognostic potential of m7G-associated lncRNA signature in predicting bladder cancer response to immunotherapy and chemotherapy
- Case Reports
- A rare FBXO25–SEPT14 fusion in a patient with chronic myeloid leukemia treatment to tyrosine kinase inhibitors: a case report
- Stage I duodenal adenocarcinoma cured by a short treatment cycle of pembrolizumab: a case report
- Rapid Communication
- ROMO1 – a potential immunohistochemical prognostic marker for cancer development
- Article Commentary
- A commentary: Role of MTA1: a novel modulator reprogramming mitochondrial glucose metabolism
Artikel in diesem Heft
- Frontmatter
- Review Articles
- Mitochondrial thermogenesis in cancer cells
- Application of indocyanine green in the management of oral cancer: a literature review
- Long non-coding RNA, FOXP4-AS1, acts as a novel biomarker of cancers
- The role of synthetic peptides derived from bovine lactoferricin against breast cancer cell lines: a mini-review
- Single cell RNA sequencing – a valuable tool for cancer immunotherapy: a mini review
- Research Articles
- Global patterns and temporal trends in ovarian cancer morbidity, mortality, and burden from 1990 to 2019
- The association between NRF2 transcriptional gene dysregulation and IDH mutation in Grade 4 astrocytoma
- More than just a KRAS inhibitor: DCAI abrogates the self-renewal of pancreatic cancer stem cells in vitro
- DUSP1 promotes pancreatic cancer cell proliferation and invasion by upregulating nephronectin expression
- IMMT promotes hepatocellular carcinoma formation via PI3K/AKT/mTOR pathway
- MiR-100-5p transfected MSCs-derived exosomes can suppress NSCLC progression via PI3K-AKT-mTOR
- Inhibitory function of CDK12i combined with WEE1i on castration-resistant prostate cancer cells in vitro and in vivo
- Prognostic potential of m7G-associated lncRNA signature in predicting bladder cancer response to immunotherapy and chemotherapy
- Case Reports
- A rare FBXO25–SEPT14 fusion in a patient with chronic myeloid leukemia treatment to tyrosine kinase inhibitors: a case report
- Stage I duodenal adenocarcinoma cured by a short treatment cycle of pembrolizumab: a case report
- Rapid Communication
- ROMO1 – a potential immunohistochemical prognostic marker for cancer development
- Article Commentary
- A commentary: Role of MTA1: a novel modulator reprogramming mitochondrial glucose metabolism