The Potential of Integrative Cancer Treatment Using Melatonin and the Challenge of Heterogeneity in Population-Based Studies: A Case Report of Colon Cancer and a Literature Review
Abstract
:1. Introduction
2. Materials and Methods
3. Results
3.1. Colonoscopy, Computed Tomography, and Histology
3.2. Systemic Chemotherapy
3.3. Clinical Manifestations in Using Melatonin
3.4. Aspirin, Food Supplements, and Nutrition
3.5. Clinical Parameters and Their Dynamics
3.5.1. Tumor-Associated Systemic Inflammation
3.5.2. Tumor Markers
3.5.3. Hemostasis Disorders
4. Discussion
4.1. Relevance of the Use of Melatonin in Integrative Cancer Treatment
4.1.1. Physiological Function of Melatonin
4.1.2. Anticancer Activity of Melatonin and Its Preliminary Clinical Studies
4.1.3. Safety and Bioavailability of Exogenous Melatonin
4.2. Integrative Approach to Cancer Treatment
4.2.1. Zinc and Selenium
4.2.2. Vitamin D3
4.2.3. Polyphenols
4.2.4. Aspirin
4.3. N-of-1 Study Design
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Bouyahya, A.; Mechchate, H.; Oumeslakht, L.; Zeouk, I.; Aboulaghras, S.; Balahbib, A.; Zengin, G.; Kamal, M.A.; Gallo, M.; Montesano, D.; et al. The Role of Epigenetic Modifications in Human Cancers and the Use of Natural Compounds as Epidrugs: Mechanistic Pathways and Pharmacodynamic Actions. Biomolecules 2022, 12, 367. [Google Scholar] [CrossRef] [PubMed]
- Kasprzak, A. The Role of Tumor Microenvironment Cells in Colorectal Cancer (CRC) Cachexia. Int. J. Mol. Sci. 2021, 22, 1565. [Google Scholar] [CrossRef] [PubMed]
- Lissoni, P.; Rovelli, F.; Vigorè, L.; Messina, G.; Lissoni, A.; Porro, G.; Di Fede, G. How to Monitor the Neuroimmune Biological Response in Patients Affected by Immune Alteration-Related Systemic Diseases. Methods Mol. Biol. 2018, 1781, 171–191. [Google Scholar] [CrossRef] [PubMed]
- Aguilar-Cazares, D.; Chavez-Dominguez, R.; Marroquin-Muciño, M.; Perez-Medina, M.; Benito-Lopez, J.J.; Camarena, A.; Rumbo-Nava, U.; Lopez-Gonzalez, J.S. The systemic-level repercussions of cancer-associated inflammation mediators produced in the tumor microenvironment. Front. Endocrinol. 2022, 13, 929572. [Google Scholar] [CrossRef] [PubMed]
- Zappavigna, S.; Cossu, A.M.; Grimaldi, A.; Bocchetti, M.; Ferraro, G.A.; Nicoletti, G.F.; Filosa, R.; Caraglia, M. Anti-Inflammatory Drugs as Anticancer Agents. Int. J. Mol. Sci. 2020, 21, 2605. [Google Scholar] [CrossRef] [PubMed]
- Admasu, F.T.; Dejenie, T.A.; Ayehu, G.W.; Zewde, E.A.; Dessie, G.; Adugna, D.G.; Enyew, E.F.; Geto, Z.; Abebe, E.C. Evaluation of thromboembolic event, basic coagulation parameters, and associated factors in patients with colorectal cancer: A multicenter study. Front. Oncol. 2023, 13, 1143122. [Google Scholar] [CrossRef] [PubMed]
- O’Brien, K.; Ried, K.; Binjemain, T.; Sali, A. Integrative Approaches to the Treatment of Cancer. Cancers 2022, 14, 5933. [Google Scholar] [CrossRef] [PubMed]
- 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, 209–249. [Google Scholar] [CrossRef] [PubMed]
- Heudobler, D.; Lüke, F.; Vogelhuber, M.; Klobuch, S.; Pukrop, T.; Herr, W.; Gerner, C.; Pantziarka, P.; Ghibelli, L.; Reichle, A. Anakoinosis: Correcting Aberrant Homeostasis of Cancer Tissue-Going Beyond Apoptosis Induction. Front. Oncol. 2019, 9, 1408. [Google Scholar] [CrossRef] [PubMed]
- Nowak-Sliwinska, P.; Scapozza, L.; Ruiz i Altaba, A. Drug repurposing in oncology: Compounds, pathways, phenotypes and computational approaches for colorectal cancer. Biochim. Biophys. Acta Rev. Cancer 2019, 1871, 434–454. [Google Scholar] [CrossRef] [PubMed]
- Lüke, F.; Harrer, D.C.; Pantziarka, P.; Pukrop, T.; Ghibelli, L.; Gerner, C.; Reichle, A.; Heudobler, D. Drug Repurposing by Tumor Tissue Editing. Front. Oncol. 2022, 12, 900985. [Google Scholar] [CrossRef] [PubMed]
- Heudobler, D.; Rechenmacher, M.; Lüke, F.; Vogelhuber, M.; Klobuch, S.; Thomas, S.; Pukrop, T.; Hackl, C.; Herr, W.; Ghibelli, L.; et al. Clinical Efficacy of a Novel Therapeutic Principle, Anakoinosis. Front. Pharmacol. 2018, 9, 1357. [Google Scholar] [CrossRef] [PubMed]
- Block, K.I.; Gyllenhaal, C.; Lowe, L.; Amedei, A.; Amin, A.R.M.R.; Amin, A.; Aquilano, K.; Arbiser, J.; Arreola, A.; Arzumanyan, A. Designing a broad-spectrum integrative approach for cancer prevention and treatment. Semin. Cancer Biol. 2015, 35, S276–S304. [Google Scholar] [CrossRef]
- Bondy, S.C.; Campbell, A. Mechanisms Underlying Tumor Suppressive Properties of Melatonin. Int. J. Mol. Sci. 2018, 19, 2205. [Google Scholar] [CrossRef] [PubMed]
- Mihanfar, A.; Yousefi, B.; Azizzadeh, B.M. Interactions of melatonin with various signaling pathways: Implications for cancer therapy. Cancer Cell Int. 2022, 22, 420. [Google Scholar] [CrossRef]
- Chok, K.C.; Ng, C.H.; Koh, R.Y.; Ng, K.Y.; Chye, S.M. The potential therapeutic actions of melatonin in colorectal cancer. Horm. Mol. Biol. Clin. Investig. 2019, 39, 20190001. [Google Scholar] [CrossRef] [PubMed]
- Gil-Martín, E.; Egea, J.; Reiter, R.J.; Romero, A. The emergence of melatonin in oncology: Focus on colorectal cancer. Med. Res. Rev. 2019, 39, 2239–2285. [Google Scholar] [CrossRef]
- Evans, S.O.; Khairuddin, P.F.; Jameson, M.B. Optimising Selenium for Modulation of Cancer Treatments. Anticancer Res. 2017, 37, 6497–6509. [Google Scholar] [CrossRef] [PubMed]
- Maalmi, H.; Walter, V.; Jansen, L.; Boakye, D.; Schöttker, B.; Hoffmeister, M.; Brenner, H. Association between Blood 25-Hydroxyvitamin D Levels and Survival in Colorectal Cancer Patients: An Updated Systematic Review and Meta-Analysis. Nutrients 2018, 10, 896. [Google Scholar] [CrossRef]
- Arora, I.; Sharma, M.; Tollefsbol, T.O. Combinatorial Epigenetics Impact of Polyphenols and Phytochemicals in Cancer Prevention and Therapy. Int. J. Mol. Sci. 2019, 20, 4567. [Google Scholar] [CrossRef] [PubMed]
- Liu, S.; Lin, Y.; Huang, S.; Xue, S.; Huang, R.; Chen, L.; Wang, C. Identifying the long-term survival beneficiary of chemotherapy for stage N1c sigmoid colon cancer. Sci. Rep. 2022, 12, 16909. [Google Scholar] [CrossRef] [PubMed]
- Lin, Q.; Zhou, H.; Shi, S.; Lin, J.; Yan, W. The Prognostic Value of Adjuvant Chemotherapy in Colon Cancer with Solitary Tumor Deposit. Front. Oncol. 2022, 12, 916091. [Google Scholar] [CrossRef] [PubMed]
- Luo, Z.; Zhu, M.G.; Zhang, Z.Q.; Ye, F.J.; Huang, W.H.; Luo, X.Z. Increased expression of Ki-67 is a poor prognostic marker for colorectal cancer patients: A meta analysis. BMC Cancer 2019, 19, 123. [Google Scholar] [CrossRef] [PubMed]
- Xiong, D.-D.; Lin, X.-G.; He, R.-Q.; Pan, D.-H.; Luo, Y.-H.; Dang, Y.-W.; Luo, D.-Z.; Chen, G.; Peng, Z.-G.; Gan, T.-Q. Ki67/MIB-1 predicts better prognoses in colorectal cancer patients received both surgery and adjuvant radio-chemotherapy: A meta-analysis of 30 studies. Int. J. Clin. Exp. Med. 2017, 10, 1788–1804. [Google Scholar]
- Fluge, Ø.; Gravdal, K.; Carlsen, E.; Vonen, B.; Kjellevold, K.; Refsum, S.; Lilleng, R.; Eide, T.J.; Halvorsen, T.B.; Tveit, K.M.; et al. Norwegian Gastrointestinal Cancer Group. Expression of EZH2 and Ki-67 in colorectal cancer and associations with treatment response and prognosis. Br. J. Cancer 2009, 101, 1282–1289. [Google Scholar] [CrossRef] [PubMed]
- Munro, A.J.; Lain, S.; Lane, D.P. P53 abnormalities and outcomes in colorectal cancer: A systematic review. Br. J. Cancer 2005, 92, 434–444. [Google Scholar] [CrossRef] [PubMed]
- Williams, D.S.; Mouradov, D.; Browne, C.; Palmieri, M.; Elliott, M.J.; Nightingale, R.; Fang, C.G.; Li, R.; Mariadason, J.M.; Faragher, I. Overexpression of TP53 protein is associated with the lack of adjuvant chemotherapy benefit in patients with stage III colorectal cancer. Mod. Pathol. 2020, 33, 483–495. [Google Scholar] [CrossRef] [PubMed]
- Oh, H.J.; Bae, J.M.; Wen, X.; Jung, S.; Kim, Y.; Kim, K.J.; Cho, N.Y.; Kim, J.H.; Han, S.W.; Kim, T.Y. p53 expression status is associated with cancer-specific survival in stage III and high-risk stage II colorectal cancer patients treated with oxaliplatin-based adjuvant chemotherapy. Br. J. Cancer 2019, 120, 797–805. [Google Scholar] [CrossRef] [PubMed]
- Zaanan, A.; Cuilliere-Dartigues, P.; Guilloux, A.; Parc, Y.; Louvet, C.; de Gramont, A.; Tiret, E.; Dumont, S.; Gayet, B.; Validire, P. Impact of p53 expression and microsatellite instability on stage III colon cancer disease-free survival in patients treated by 5-fluorouracil and leucovorin with or without oxaliplatin. Ann. Oncol. 2010, 21, 772–780. [Google Scholar] [CrossRef] [PubMed]
- Ji, R.; Huang, G.; Xu, J.; Yu, X.; Zhou, A.; Du, C. Splenomegaly during oxaliplatin-based chemotherapy: Impact on blood parameters and anti-neoplastic treatment. Transl. Cancer Res. 2022, 11, 1880–1888. [Google Scholar] [CrossRef] [PubMed]
- Selvy, M.; Pereira, B.; Kerckhove, N.; Gonneau, C.; Feydel, G.; Pétorin, C.; Vimal-Baguet, A.; Melnikov, S.; Kullab, S.; Hebbar, M.; et al. Long-Term Prevalence of Sensory Chemotherapy-Induced Peripheral Neuropathy for 5 Years after Adjuvant FOLFOX Chemotherapy to Treat Colorectal Cancer: A Multicenter Cross-Sectional Study. J. Clin. Med. 2020, 9, 2400. [Google Scholar] [CrossRef] [PubMed]
- Areti, A.; Komirishetty, P.; Akuthota, M.; Malik, R.A.; Kumar, A. Melatonin prevents mitochondrial dysfunction and promotes neuroprotection by inducing autophagy during oxaliplatin-evoked peripheral neuropathy. J. Pineal. Res. 2017, 62, 3. [Google Scholar] [CrossRef]
- Wang, Y.; Wang, P.; Zheng, X.; Du, X. Therapeutic strategies of melatonin in cancer patients: A systematic review and meta-analysis. Onco Targets Ther. 2018, 11, 7895–7908. [Google Scholar] [CrossRef]
- Nahleh, Z.; Pruemer, J.; Lafollette, J.; Sweany, S. Melatonin, a promising role in taxane-related neuropathy. Clin. Med. Insights Oncol. 2010, 4, 35–41. [Google Scholar] [CrossRef]
- Ali, M.; Aziz, T. The Combination of Zinc and Melatonin Enhanced Neuroprotection and Attenuated Neuropathy in Oxaliplatin-Induced Neurotoxicity. Drug Des. Devel. Ther. 2022, 16, 3447–3463. [Google Scholar] [CrossRef] [PubMed]
- Dehmer, S.P.; O’Keefe, L.R.; Evans, C.V.; Guirguis-Blake, J.M.; Perdue, L.A.; Maciosek, M.V. Aspirin Use to Prevent Cardiovascular Disease and Colorectal Cancer: Updated Modeling Study for the US Preventive Services Task Force. JAMA. 2022, 327, 1598–1607. [Google Scholar] [CrossRef]
- Xiao, S.; Xie, W.; Fan, Y.; Zhou, L. Timing of Aspirin Use among Patients with Colorectal Cancer in Relation to Mortality: A Systematic Review and Meta-Analysis. JNCI Cancer Spectr. 2021, 5, pkab067. [Google Scholar] [CrossRef] [PubMed]
- Radomska, D.; Czarnomysy, R.; Radomski, D.; Bielawska, A.; Bielawski, K. Selenium as a Bioactive Micronutrient in the Human Diet and Its Cancer Chemopreventive Activity. Nutrients 2021, 13, 1649. [Google Scholar] [CrossRef] [PubMed]
- Hu, W.; Zhao, C.; Hu, H.; Yin, S. Food Sources of Selenium and Its Relationship with Chronic Diseases. Nutrients 2021, 13, 1739. [Google Scholar] [CrossRef] [PubMed]
- Baker, J.R.; Umesh, S.; Jenab, M.; Schomburg, L.; Tjønneland, A.; Olsen, A.; Boutron-Ruault, M.-C.; Rothwell, J.A.; Severi, G.; Katzke, V.; et al. Prediagnostic Blood Selenium Status and Mortality among Patients with Colorectal Cancer in Western European Populations. Biomedicines 2021, 9, 1521. [Google Scholar] [CrossRef] [PubMed]
- Pozharitskaya, O.N.; Karlina, M.V.; Shikov, A.N.; Kosman, V.M.; Makarova, M.N.; Makarov, V.G. Determination and pharmacokinetic study of taxifolin in rabbit plasma by high-performance liquid chromatography. Phytomedicine 2009, 16, 244–251. [Google Scholar] [CrossRef]
- Van Blarigan, E.L.; Meyerhardt, J.A. Role of physical activity and diet after colorectal cancer diagnosis. J. Clin. Oncol. 2015, 33, 1825–1834. [Google Scholar] [CrossRef] [PubMed]
- Fadelu, T.; Zhang, S.; Niedzwiecki, D.; Ye, X.; Saltz, L.B.; Mayer, R.J.; Mowat, R.B.; Whittom, R.; Hantel, A.; Benson, A.B.; et al. Nut Consumption and Survival in Patients with Stage III Colon Cancer: Results from CALGB 89803 (Alliance). J. Clin. Oncol. 2018, 36, 1112–1120. [Google Scholar] [CrossRef] [PubMed]
- Kristo, A.S.; Klimis-Zacas, D.; Sikalidis, A.K. Protective Role of Dietary Berries in Cancer. Antioxidants 2016, 5, 37. [Google Scholar] [CrossRef] [PubMed]
- Filippini, T.; Malavolti, M.; Borrelli, F.; Izzo, A.A.; Fairweather-Tait, S.J.; Horneber, M.; Vinceti, M. Green tea (Camellia sinensis) for the prevention of cancer. Cochrane Database Syst. Rev. 2020, 3, CD005004. [Google Scholar] [CrossRef]
- Mahomoodally, M.F.; Aumeeruddy, M.Z.; Rengasamy, K.R.R.; Roshan, S.; Hammad, S.; Pandohee, J.; Hu, X.; Zengin, G. Ginger and its active compounds in cancer therapy: From folk uses to nano-therapeutic applications. Semin. Cancer Biol. 2021, 69, 140–149. [Google Scholar] [CrossRef] [PubMed]
- Walter, V.; Jansen, L.; Ulrich, A.; Roth, W.; Bläker, H.; Chang-Claude, J.; Hoffmeister, M.; Brenner, H. Alcohol consumption and survival of colorectal cancer patients: A population-based study from Germany. Am. J. Clin. Nutr. 2016, 103, 1497–1506. [Google Scholar] [CrossRef] [PubMed]
- Romagnolo, D.F.; Donovan, M.G.; Doetschman, T.C.; Selmin, O.I. n-6 Linoleic Acid Induces Epigenetics Alterations Associated with Colonic Inflammation and Cancer. Nutrients 2019, 11, 171. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, S.; Li, H.; Yu, D.; Cai, H.; Gao, J.; Gao, Y.; Luu, H.N.; Tran, H.; Xiang, Y.B.; Zheng, W. Dietary fatty acids and colorectal cancer risk in men: A report from the Shanghai Men’s Health Study and a meta-analysis. Int. J. Cancer 2021, 148, 77–89. [Google Scholar] [CrossRef] [PubMed]
- Simopoulos, A.P. The importance of the omega-6/omega-3 fatty acid ratio in cardiovascular disease and other chronic diseases. Exp. Biol. Med. 2008, 233, 674–688. [Google Scholar] [CrossRef]
- Bojková, B.; Winklewski, P.J.; Wszedybyl-Winklewska, M. Dietary Fat and Cancer-Which Is Good, Which Is Bad, and the Body of Evidence. Int. J. Mol. Sci. 2020, 21, 4114. [Google Scholar] [CrossRef] [PubMed]
- Shahjehan, F.; Merchea, A.; Cochuyt, J.J.; Li, Z.; Colibaseanu, D.T.; Kasi, P.M. Body Mass Index and Long-Term Outcomes in Patients with Colorectal Cancer. Front. Oncol. 2018, 8, 620. [Google Scholar] [CrossRef]
- Delpino, F.M.; Figueiredo, L.M. Melatonin supplementation and anthropometric indicators of obesity: A systematic review and meta-analysis. Nutrition 2021, 91–92, 111399. [Google Scholar] [CrossRef] [PubMed]
- Alam, W.; Ullah, H.; Santarcangelo, C.; Di Minno, A.; Khan, H.; Daglia, M.; Arciola, C.R. Micronutrient Food Supplements in Patients with Gastro-Intestinal and Hepatic Cancers. Int. J. Mol. Sci. 2021, 22, 8014. [Google Scholar] [CrossRef] [PubMed]
- Fasinu, P.S.; Rapp, G.K. Herbal Interaction with Chemotherapeutic Drugs-A Focus on Clinically Significant Findings. Front. Oncol. 2019, 9, 1356. [Google Scholar] [CrossRef] [PubMed]
- Duda-Chodak, A.; Tarko, T. Possible Side Effects of Polyphenols and Their Interactions with Medicines. Molecules 2023, 28, 2536. [Google Scholar] [CrossRef] [PubMed]
- Yasueda, A.; Urushima, H.; Ito, T. Efficacy and Interaction of Antioxidant Supplements as Adjuvant Therapy in Cancer Treatment: A Systematic Review. Integr. Cancer Ther. 2016, 15, 17–39. [Google Scholar] [CrossRef]
- Florido, J.; Rodriguez-Santana, C.; Martinez-Ruiz, L.; López-Rodríguez, A.; Acuña-Castroviejo, D.; Rusanova, I.; Escames, G. Understanding the Mechanism of Action of Melatonin, Which Induces ROS Production in Cancer Cells. Antioxidants 2022, 11, 1621. [Google Scholar] [CrossRef] [PubMed]
- Greten, F.R.; Grivennikov, S.I. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity 2019, 51, 27–41. [Google Scholar] [CrossRef] [PubMed]
- Rumba, R.; Cipkina, S.; Cukure, F.; Vanags, A. Systemic and local inflammation in colorectal cancer. Acta Med. Litu. 2018, 25, 185–196. [Google Scholar] [CrossRef] [PubMed]
- Misiewicz, A.; Dymicka-Piekarska, V. Fashionable, but What is Their Real Clinical Usefulness? NLR, LMR, and PLR as a Propmising Indicator in Colorectal Cancer Prognosis: A Systematic Review. J. Inflamm. Res. 2023, 16, 69–81. [Google Scholar] [CrossRef] [PubMed]
- Yamamoto, T.; Kawada, K.; Obama, K. Inflammation-Related Biomarkers for the Prediction of Prognosis in Colorectal Cancer Patients. Int. J. Mol. Sci. 2021, 22, 8002. [Google Scholar] [CrossRef] [PubMed]
- Xu, X.R.; Yousef, G.M.; Ni, H. Cancer and platelet crosstalk: Opportunities and challenges for aspirin and other antiplatelet agents. Blood 2018, 131, 1777–1789. [Google Scholar] [CrossRef] [PubMed]
- Nielsen, S.R.; Schmid, M.C. Macrophages as Key Drivers of Cancer Progression and Metastasis. Mediators Inflamm. 2017, 2017, 9624760. [Google Scholar] [CrossRef] [PubMed]
- Liang, L.; Zhu, J.; Jia, H.; Huang, L.; Li, D.; Li, Q.; Li, X. Predictive value of pretreatment lymphocyte count in stage II colorectal cancer and in high-risk patients treated with adjuvant chemotherapy. Oncotarget 2016, 7, 1014–1028. [Google Scholar] [CrossRef] [PubMed]
- Yasui, K.; Shida, D.; Nakamura, Y.; Ahiko, Y.; Tsukamoto, S.; Kanemitsu, Y. Postoperative, but not preoperative, inflammation-based prognostic markers are prognostic factors in stage III colorectal cancer patients. Br. J. Cancer 2021, 124, 933–941. [Google Scholar] [CrossRef] [PubMed]
- Herold, Z.; Herold, M.; Lohinszky, J.; Szasz, A.M.; Dank, M.; Somogyi, A. Longitudinal changes in personalized platelet count metrics are good indicators of initial 3-year outcome in colorectal cancer. World J. Clin. Cases 2022, 10, 6825–6844. [Google Scholar] [CrossRef] [PubMed]
- Fu, J.; Zhu, J.; Du, F.; Zhang, L.; Li, D.; Huang, H.; Tian, T.; Liu, Y.; Zhang, L.; Liu, Y.; et al. Prognostic Inflammatory Index Based on Preoperative Peripheral Blood for Predicting the Prognosis of Colorectal Cancer Patients. Cancers 2021, 13, 3. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Jia, H.; Yu, W.; Xu, Y.; Li, X.; Li, Q.; Cai, S. Nomograms for predicting prognostic value of inflammatory biomarkers in colorectal cancer patients after radical resection. Int. J. Cancer 2016, 139, 220–231. [Google Scholar] [CrossRef] [PubMed]
- Lissoni, P.; Messina, G.; Borsotti, G.; Alessio, T.; Stefano, F.; Simonetta, T.; Di Fede, G. Modulation of Immune and Anti-Tumor Effects of Cancer Immunotherapy with Anti-Pd-1 Monoclonal Antibodies by the Pineal Hormone Melatonin: Preliminary Clinical Results. J. Immuno. Allerg. 2020, 1, 1–6. [Google Scholar] [CrossRef]
- Lissoni, P.; Barni, S.; Tancini, G.; Ardizzoia, A.; Ricci, G.; Aldeghi, R.; Brivio, F.; Tisi, E.; Rovelli, F.; Rescaldani, R.; et al. A randomised study with subcutaneous low-dose interleukin 2 alone vs interleukin 2 plus the pineal neurohormone melatonin in advanced solid neoplasms other than renal cancer and melanoma. Br. J. Cancer 1994, 69, 196–199. [Google Scholar] [CrossRef] [PubMed]
- Noh, O.K.; Oh, S.Y.; Kim, Y.B.; Suh, K.W. Prognostic Significance of Lymphocyte Counts in Colon Cancer Patients Treated with FOLFOX Chemotherapy. World J. Surg. 2017, 41, 2898–2905. [Google Scholar] [CrossRef] [PubMed]
- Idos, G.E.; Kwok, J.; Bonthala, N.; Kysh, L.; Gruber, S.B.; Qu, C. The Prognostic Implications of Tumor Infiltrating Lymphocytes in Colorectal Cancer: A Systematic Review and Meta-Analysis. Sci. Rep. 2020, 10, 3360. [Google Scholar] [CrossRef] [PubMed]
- Wei, Y.; Zhang, X.; Wang, G.; Zhou, Y.; Luo, M.; Wang, S.; Hong, C. The impacts of pretreatment circulating eosinophils and basophils on prognosis of stage I-III colorectal cancer. Asia. Pac. J. Clin. Oncol. 2018, 14, e243–e251. [Google Scholar] [CrossRef] [PubMed]
- Gao, L.; Yuan, C.; Fu, J.; Tian, T.; Huang, H.; Zhang, L.; Li, D.; Liu, Y.; Meng, S.; Liu, Y. Prognostic scoring system based on eosinophil- and basophil-related markers for predicting the prognosis of patients with stage II and stage III colorectal cancer: A retrospective cohort study. Front. Oncol. 2023, 13, 1182944. [Google Scholar] [CrossRef] [PubMed]
- Akinbo, D.B.; Ajayi, O.I. Thrombotic Pathogenesis and Laboratory Diagnosis in Cancer Patients, An Update. Int. J. Gen. Med. 2023, 16, 259–272. [Google Scholar] [CrossRef] [PubMed]
- Lu, S.L.; Ye, Z.H.; Ling, T.; Liang, S.Y.; Li, H.; Tang, X.Z.; Xu, Y.S.; Tang, W.Z. High pretreatment plasma D-dimer predicts poor survival of colorectal cancer: Insight from a meta-analysis of observational studies. Oncotarget 2017, 8, 81186–81194. [Google Scholar] [CrossRef] [PubMed]
- Liu, C.; Ning, Y.; Chen, X.; Zhu, Q. D-Dimer level was associated with prognosis in metastatic colorectal cancer: A Chinese patients based cohort study. Medicine 2020, 99, e19243. [Google Scholar] [CrossRef] [PubMed]
- Shibutani, M.; Kashiwagi, S.; Fukuoka, T.; Iseki, Y.; Kasashima, H.; Kitayama, K.; Maeda, K. Prognostic Role of Preoperative D-dimer Levels in Patients with Stage I-III Colorectal Cancer. Cancer Diagn. Progn. 2023, 3, 38–43. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Ye, J.; Luo, Q.; Kuang, M.; Mao, M.; Dai, S.; Wang, X. Prediction of Poor Outcomes in Patients with Colorectal Cancer: Elevated Preoperative Prothrombin Time (PT) and Activated Partial Thromboplastin Time (APTT). Cancer Manag. Res. 2020, 12, 5373–5384. [Google Scholar] [CrossRef]
- Tønnesen, J.; Pallisgaard, J.; Rasmussen, P.V.; Ruwald, M.H.; Lamberts, M.; Nouhravesh, N.; Strange, J.; Gislason, G.H.; Hansen, M.L. Risk and timing of venous thromboembolism in patients with gastrointestinal cancer: A nationwide Danish cohort study. BMJ Open 2023, 13, e062768. [Google Scholar] [CrossRef] [PubMed]
- Khorana, A.A.; Kuderer, N.M.; Culakova, E.; Lyman, G.H.; Francis, C.W. Development and validation of a predictive model for chemotherapy-associated thrombosis. Blood 2008, 111, 4902–4907. [Google Scholar] [CrossRef] [PubMed]
- Peterson, E.A.; Lee, A.Y.Y. Update from the clinic: What’s new in the diagnosis of cancer-associated thrombosis? Hematology Am. Soc. Hematol. Educ. Program. 2019, 2019, 167–174. [Google Scholar] [CrossRef] [PubMed]
- Bosch, F.T.M.; Mulder, F.I.; Kamphuisen, P.W.; Middeldorp, S.; Bossuyt, P.M.; Büller, H.R.; van Es, N. Primary thromboprophylaxis in ambulatory cancer patients with a high Khorana score: A systematic review and meta-analysis. Blood Adv. 2020, 4, 5215–5225. [Google Scholar] [CrossRef] [PubMed]
- Hasan, Z.T.; Atrakji, D.M.Q.Y.M.A.A.; Mehuaiden, D.A.K. The Effect of Melatonin on Thrombosis, Sepsis and Mortality Rate in COVID-19 Patients. Int. J. Infect. Dis. 2022, 114, 79–84. [Google Scholar] [CrossRef] [PubMed]
- Shohrati, M.; Mohammadi, A.; Najafi, A.; Sharifzadeh, M.; Sharifnia, H.; Abdollahi, M.; Salesi, M.; Sahebnasagh, A.; Mojtahedzadeh, M. Evaluation of the Effects of Melatonin Supplementation on Coagulation in Patients with Haemorrhagic Stroke; A Randomized, Double-Blind, Controlled Trial. Front. Emerg. Med. 2021, 5, e32. [Google Scholar] [CrossRef]
- Pashalieva, I.; Decheva, L.; Stancheva, E.; Nyagolov, Y.; Negrev, N. Melatonin and luzindole–induced effects on integral blood coagulation parameters in rats. Comptes Rendus Acad. Bulg. Sci. 2014, 67, 1269–1274. [Google Scholar]
- Lissoni, P.; Mandala, M.; Rossini, F.; Fumagalli, L.; Barni, S. Growth Factors: Thrombopoietic Property of the Pineal Hormone Melatonin. Hematology 1999, 4, 335–343. [Google Scholar] [CrossRef] [PubMed]
- Lissoni, P.; Barni, S.; Mandalà, M.; Ardizzoia, A.; Paolorossi, F.; Vaghi, M.; Longarini, R.; Malugani, F.; Tancini, G. Decreased toxicity and increased efficacy of cancer chemotherapy using the pineal hormone melatonin in metastatic solid tumour patients with poor clinical status. Eur. J. Cancer 1999, 35, 1688–1692. [Google Scholar] [CrossRef]
- Esmaeili, A.; Nassiri Toosi, M.; Taher, M.; Bayani, J.; Namazi, S. Melatonin effect on platelet count in patients with liver disease. Gastroenterol. Hepatol. Bed Bench 2021, 14, 356–361. [Google Scholar] [PubMed]
- Girish, K.S.; Paul, M.; Thushara, R.M.; Hemshekhar, M.; Shanmuga Sundaram, M.; Rangappa, K.S.; Kemparaju, K. Melatonin elevates apoptosis in human platelets via ROS mediated mitochondrial damage. Biochem. Biophys. Res. Commun. 2013, 438, 198–204. [Google Scholar] [CrossRef] [PubMed]
- Frenck, R.W., Jr.; Mansour, A.; Nakhla, I.; Sultan, Y.; Putnam, S.; Wierzba, T.; Morsy, M.; Knirsch, C. Short-course azithromycin for the treatment of uncomplicated typhoid fever in children and adolescents. Clin. Infect. Dis. 2004, 38, 951–957. [Google Scholar] [CrossRef] [PubMed]
- Yang, C.J.; Hwang, J.J.; Hung, J.Y.; Chong, I.W.; Huang, M.S. Extreme thrombocytosis under the treatment by amoxicillin/clavulanate. Pharm. World Sci. 2006, 28, 326–328. [Google Scholar] [CrossRef] [PubMed]
- Timp, J.F.; Cannegieter, S.C.; Tichelaar, V.; Braekkan, S.K.; Rosendaal, F.R.; le Cessie, S.; Lijfering, W.M. Antibiotic use as a marker of acute infection and risk of first and recurrent venous thrombosis. Br. J. Haematol. 2017, 176, 961–970. [Google Scholar] [CrossRef] [PubMed]
- Panebianco, C.; Andriulli, A.; Pazienza, V. Pharmacomicrobiomics: Exploiting the drug-microbiota interactions in anticancer therapies. Microbiome 2018, 6, 92. [Google Scholar] [CrossRef] [PubMed]
- Mohamed, A.; Menon, H.; Chulkina, M.; Yee, N.S.; Pinchuk, I.V. Drug–Microbiota Interaction in Colon Cancer Therapy: Impact of Antibiotics. Biomedicines 2021, 9, 259. [Google Scholar] [CrossRef] [PubMed]
- LaCourse, K.D.; Zepeda-Rivera, M.; Kempchinsky, A.G.; Baryiames, A.; Minot, S.S.; Johnston, C.D.; Bullman, S. The cancer chemotherapeutic 5-fluorouracil is a potent Fusobacterium nucleatum inhibitor and its activity is modified by intratumoral microbiota. Cell Rep. 2022, 41, 111625. [Google Scholar] [CrossRef]
- Brennan, C.A.; Nakatsu, G.; Gallini Comeau, C.A.; Drew, D.A.; Glickman, J.N.; Schoen, R.E.; Chan, A.T.; Garrett, W.S. Aspirin Modulation of the Colorectal Cancer-Associated Microbe Fusobacterium nucleatum. mBio 2021, 12, e00547-21. [Google Scholar] [CrossRef] [PubMed]
- Smorodin, E.P. Prospects and Challenges of the Study of Anti-Glycan Antibodies and Microbiota for the Monitoring of Gastrointestinal Cancer. Int. J. Mol. Sci. 2021, 22, 11608. [Google Scholar] [CrossRef] [PubMed]
- Huangfu, S.C.; Zhang, W.B.; Zhang, H.R.; Li, Y.; Zhang, Y.R.; Nie, J.L.; Chu, X.; Chen, C.S.; Jiang, H.P.; Pan, J.H. Clinicopathological and prognostic significance of Fusobacterium nucleatum infection in colorectal cancer: A meta-analysis. J. Cancer 2021, 12, 1583–1591. [Google Scholar] [CrossRef]
- Liu, R.Y.; Li, L.; Zhang, Z.T.; Wu, T.; Lin, S.; Zhang, X.T. Clinical efficacy of melatonin as adjunctive therapy to non-surgical treatment of periodontitis: A systematic review and meta-analysis. Inflammopharmacology 2022, 30, 695–704. [Google Scholar] [CrossRef] [PubMed]
- Fowler, S.; Hoedt, E.C.; Talley, N.J.; Keely, S.; Burns, G.L. Circadian Rhythms and Melatonin Metabolism in Patients with Disorders of Gut-Brain Interactions. Front. Neurosci. 2022, 16, 825246. [Google Scholar] [CrossRef]
- Liu, W.; Zhou, Z.; Dong, D.; Sun, L.; Zhang, G. Sex Differences in the Association between Night Shift Work and the Risk of Cancers: A Meta-Analysis of 57 Articles. Dis. Markers 2018, 2018, 7925219. [Google Scholar] [CrossRef] [PubMed]
- Talib, W.H.; Alsayed, A.R.; Abuawad, A.; Daoud, S.; Mahmod, A.I. Melatonin in Cancer Treatment: Current Knowledge and Future Opportunities. Molecules 2021, 26, 2506. [Google Scholar] [CrossRef] [PubMed]
- Kvetnoy, I.; Ivanov, D.; Mironova, E.; Evsyukova, I.; Nasyrov, R.; Kvetnaia, T.; Polyakova, V. Melatonin as the Cornerstone of Neuroimmunoendocrinology. Int. J. Mol. Sci. 2022, 23, 1835. [Google Scholar] [CrossRef] [PubMed]
- Esteban-Zubero, E.; López-Pingarrón, L.; Alatorre-Jiménez, M.A.; Ochoa-Moneo, P.; Buisac-Ramón, C.; Rivas-Jiménez, M.; Castán-Ruiz, S.; Antoñanzas-Lombarte, Á.; Tan, D.X.; García, J.J.; et al. Melatonin’s role as a co-adjuvant treatment in colonic diseases: A review. Life Sci. 2017, 170, 72–81. [Google Scholar] [CrossRef] [PubMed]
- Hibberd, T.J.; Ramsay, S.; Spencer-Merris, P.; Dinning, P.G.; Zagorodnyuk, V.P.; Spencer, N.J. Circadian rhythms in colonic function. Front. Physiol. 2023, 14, 1239278. [Google Scholar] [CrossRef] [PubMed]
- Iesanu, M.I.; Zahiu, C.D.M.; Dogaru, I.-A.; Chitimus, D.M.; Pircalabioru, G.G.; Voiculescu, S.E.; Isac, S.; Galos, F.; Pavel, B.; O’Mahony, S.M.; et al. Melatonin–Microbiome Two-Sided Interaction in Dysbiosis-Associated Conditions. Antioxidants 2022, 11, 2244. [Google Scholar] [CrossRef] [PubMed]
- Zhang, N.; Sundquist, J.; Sundquist, K.; Ji, J. Use of Melatonin Is Associated with Lower Risk of Colorectal Cancer in Older Adults. Clin. Transl. Gastroenterol. 2021, 12, e00396. [Google Scholar] [CrossRef] [PubMed]
- Jensen, L.D.; Oliva, D.; Andersson, B.Å.; Lewin, F. A multidisciplinary perspective on the complex interactions between sleep, circadian, and metabolic disruption in cancer patients. Cancer Metastasis Rev. 2021, 40, 1055–1071. [Google Scholar] [CrossRef] [PubMed]
- Innominato, P.F.; Giacchetti, S.; Bjarnason, G.A.; Focan, C.; Garufi, C.; Coudert, B.; Iacobelli, S.; Tampellini, M.; Durando, X.; Mormont, M.C.; et al. Prediction of overall survival through circadian rest-activity monitoring during chemotherapy for metastatic colorectal cancer. Int. J. Cancer 2012, 131, 2684–2692. [Google Scholar] [CrossRef] [PubMed]
- Kos-Kudla, B.; Ostrowska, Z.; Kozlowski, A.; Marek, B.; Ciesielska-Kopacz, N.; Kudla, M.; Kajdaniuk, D.; Strzelczyk, J.; Staszewicz, P. Circadian rhythm of melatonin in patients with colorectal carcinoma. Neuro. Endocrinol. Lett. 2002, 23, 239–242. [Google Scholar] [PubMed]
- Minich, D.M.; Henning, M.; Darley, C.; Fahoum, M.; Schuler, C.B.; Frame, J. Is Melatonin the “Next Vitamin D”?: A Review of Emerging Science, Clinical Uses, Safety, and Dietary Supplements. Nutrients 2022, 14, 3934. [Google Scholar] [CrossRef] [PubMed]
- Anghel, L.; Baroiu, L.; Popazu, C.R.; Pătraș, D.; Fotea, S.; Nechifor, A.; Ciubara, A.; Nechita, L.; Mușat, C.L.; Stefanopol, I.A.; et al. Benefits and adverse events of melatonin use in the elderly (Review). Exp. Ther. Med. 2022, 23, 219. [Google Scholar] [CrossRef] [PubMed]
- Gurunathan, S.; Qasim, M.; Kang, M.H.; Kim, J.H. Role and Therapeutic Potential of Melatonin in Various Type of Cancers. Onco Targets Ther. 2021, 14, 2019–2052. [Google Scholar] [CrossRef] [PubMed]
- Xin, Z.; Jiang, S.; Jiang, P.; Yan, X.; Fan, C.; Di, S.; Wu, G.; Yang, Y.; Reiter, R.J.; Ji, G. Melatonin as a treatment for gastrointestinal cancer: A review. J. Pineal Res. 2015, 58, 375–387. [Google Scholar] [CrossRef] [PubMed]
- Zarezadeh, M.; Khorshidi, M.; Emami, M.; Janmohammadi, P.; Kord-Varkaneh, H.; Mousavi, S.M.; Mohammed, S.H.; Saedisomeolia, A.; Alizadeh, S. Melatonin supplementation and pro-inflammatory mediators: A systematic review and meta-analysis of clinical trials. Eur. J. Nutr. 2020, 59, 1803–1813. [Google Scholar] [CrossRef] [PubMed]
- Lissoni, P. Biochemotherapy with standard chemotherapies plus the pineal hormone melatonin in the treatment of advanced solid neoplasms. Pathol. Biol. 2007, 55, 201–204. [Google Scholar] [CrossRef] [PubMed]
- Lissoni, P. Is there a role for melatonin in supportive care? Support. Care Cancer 2002, 10, 110–116. [Google Scholar] [CrossRef] [PubMed]
- Lissoni, P.; Chilelli, M.; Villa, S.; Cerizza, L.; Tancini, G. Five years survival in metastatic non-small cell lung cancer patients treated with chemotherapy alone or chemotherapy and melatonin: A randomized trial. J. Pineal Res. 2003, 35, 12–15. [Google Scholar] [CrossRef]
- Cerea, G.; Vaghi, M.; Ardizzoia, A.; Villa, S.; Bucovec, R.; Mengo, S.; Gardani, G.; Tancini, G.; Lissoni, P. Biomodulation of cancer chemotherapy for metastatic colorectal cancer: A randomized study of weekly low-dose irinotecan alone versus irinotecan plus the oncostatic pineal hormone melatonin in metastatic colorectal cancer patients progressing on 5-fluorouracil-containing combinations. Anticancer Res. 2003, 23, 1951–1954. [Google Scholar] [PubMed]
- Seely, D.; Wu, P.; Fritz, H.; Kennedy, D.A.; Tsui, T.; Seely, A.J.; Mills, E. Melatonin as adjuvant cancer care with and without chemotherapy: A systematic review and meta-analysis of randomized trials. Integr. Cancer Ther. 2012, 11, 293–303. [Google Scholar] [CrossRef] [PubMed]
- Wu, H.; Liu, J.; Yin, Y.; Zhang, D.; Xia, P.; Zhu, G. Therapeutic Opportunities in Colorectal Cancer: Focus on Melatonin Antioncogenic Action. Biomed. Res. Int. 2019, 2019, 9740568. [Google Scholar] [CrossRef] [PubMed]
- Su, S.C.; Hsieh, M.J.; Yang, W.E.; Chung, W.H.; Reiter, R.J.; Yang, S.F. Cancer metastasis: Mechanisms of inhibition by melatonin. J. Pineal Res. 2017, 62, 1. [Google Scholar] [CrossRef]
- Lissoni, P.; Rovelli, F.; Porro, G.; Brivio, F.; Fumagalli, L.; Lissoni, A.; Messina, G.; Cenaj, V.; Di Fede, G. Treatment of Advanced Cancer related Lymphocytopenia: Comparison among the Effects of Subcutaneous Low-dose Interleukin-2, High-dose Pineal Hormone Melatonin and Checkpoint Inhibitors. J. Cancer Res. Oncobiol. 2018, 1, 112. [Google Scholar] [CrossRef]
- Vigoré, L.; Messina, G.; Brivio, F.; Fumagalli, L.; Rovelli, F.; Di Fede, G.; Lissoni, P. Psychoneuroendocrine modulation of regulatory T lymphocyte system: In vivo and in vitro effects of the pineal immunomodulating hormone melatonin. In Vivo 2010, 24, 787–789. [Google Scholar] [PubMed]
- Lissoni, P.; Messina, G.; Lissoni, A.; Franco, R. The psychoneuroendocrine-immunotherapy of cancer: Historical evolution and clinical results. J. Res. Med. Sci. 2017, 22, 45. [Google Scholar] [CrossRef] [PubMed]
- Bitzer-Quintero, O.K.; Ortiz, G.G.; Jaramillo-Bueno, S.; Ramos-González, E.J.; Márquez-Rosales, M.G.; Delgado-Lara, D.L.C.; Torres-Sánchez, E.D.; Tejeda-Martínez, A.R.; Ramirez-Jirano, J. Psycho-Neuro-Endocrine-Immunology: A Role for Melatonin in This New Paradigm. Molecules 2022, 27, 4888. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Li, S.; Zhou, Y.; Meng, X.; Zhang, J.J.; Xu, D.P.; Li, H.B. Melatonin for the prevention and treatment of cancer. Oncotarget 2017, 8, 39896–39921. [Google Scholar] [CrossRef] [PubMed]
- Ma, Z.; Xu, L.; Liu, D.; Zhang, X.; Di, S.; Li, W.; Zhang, J.; Reiter, R.J.; Han, J.; Li, X.; et al. Utilizing Melatonin to Alleviate Side Effects of Chemotherapy: A Potentially Good Partner for Treating Cancer with Ageing. Oxid. Med. Cell. Longev. 2020, 2020, 6841581. [Google Scholar] [CrossRef] [PubMed]
- Onseng, K.; Johns, N.P.; Khuayjarernpanishk, T.; Subongkot, S.; Priprem, A.; Hurst, C.; Johns, J. Beneficial Effects of Adjuvant Melatonin in Minimizing Oral Mucositis Complications in Head and Neck Cancer Patients Receiving Concurrent Chemoradiation. J. Altern. Complement. Med. 2017, 23, 957–963. [Google Scholar] [CrossRef] [PubMed]
- Farhood, B.; Goradel, N.H.; Mortezaee, K.; Khanlarkhani, N.; Salehi, E.; Nashtaei, M.S.; Mirtavoos-Mahyari, H.; Motevaseli, E.; Shabeeb, D.; Musa, A.E.; et al. Melatonin as an adjuvant in radiotherapy for radioprotection and radiosensitization. Clin. Transl. Oncol. 2019, 21, 268–279. [Google Scholar] [CrossRef] [PubMed]
- Nuszkiewicz, J.; Woźniak, A.; Szewczyk-Golec, K. Ionizing Radiation as a Source of Oxidative Stress—The Protective Role of Melatonin and Vitamin D. Int. J. Mol. Sci. 2020, 21, 5804. [Google Scholar] [CrossRef] [PubMed]
- Jadid, M.F.S.; Aghaei, E.; Taheri, E.; Seyyedsani, N.; Chavoshi, R.; Abbasi, S.; Khorrami, A.; Goleij, P.; Hajazimian, S.; Taefehshokr, S.; et al. Melatonin increases the anticancer potential of doxorubicin in Caco-2 colorectal cancer cells. Environ. Toxicol. 2021, 36, 1061–1069. [Google Scholar] [CrossRef]
- Lissoni, P.; Rovelli, F.; Brivio, F.; Messina, G.; Lissoni, A.; Pensato, S.; Di Fede, G. Five year-survivals with high-dose melatonin and other antitumor pineal hormones in advanced cancer patients eligible for the only palliative therapy. Res. J. Oncol. 2018, 2, 2. [Google Scholar]
- Lissoni, P.; Cazzaniga, M.; Tancini, G.; Scardino, E.; Musci, R.; Barni, S.; Maffezzini, M.; Meroni, T.; Rocco, F.; Conti, A.; et al. Reversal of clinical resistance to LHRH analogue in metastatic prostate cancer by the pineal hormone melatonin: Efficacy of LHRH analogue plus melatonin in patients progressing on LHRH analogue alone. Eur. Urol. 1997, 31, 178–181. [Google Scholar] [CrossRef] [PubMed]
- Zharinov, G.M.; Bogomolov, O.A.; Chepurnaya, I.V.; Neklasova, N.Y.; Anisimov, V.N. Melatonin increases overall survival of prostate cancer patients with poor prognosis after combined hormone radiation treatment. Oncotarget 2020, 11, 3723–3729. [Google Scholar] [CrossRef] [PubMed]
- Harpsøe, N.G.; Andersen, L.P.; Gögenur, I.; Rosenberg, J. Clinical pharmacokinetics of melatonin: A systematic review. Eur. J. Clin. Pharmacol. 2015, 71, 901–909. [Google Scholar] [CrossRef] [PubMed]
- Zetner, D.; Andersen, L.P.; Rosenberg, J. Pharmacokinetics of alternative administration routes of melatonin: A systematic review. Drug Res. 2016, 66, 169–173. [Google Scholar] [CrossRef] [PubMed]
- Hrushesky, W.J.M.; Lis, C.G.; Levin, R.D.; Grutsch, J.F.; Birdsall, T.; Wood, P.A.; Huff, D.F.Q.; Reynolds, J.L.; Pearl, D.K.; Shen, X.; et al. Daily evening melatonin prolongs survival among patients with advanced non-small-cell lung cancer. Biol. Rhythm. Res. 2022, 53, 1043–1057. [Google Scholar] [CrossRef]
- Seely, D.; Legacy, M.; Auer, R.C.; Fazekas, A.; Delic, E.; Anstee, C.; Angka, L.; Kennedy, M.A.; Tai, L.H.; Zhang, T.; et al. Adjuvant melatonin for the prevention of recurrence and mortality following lung cancer resection (AMPLCaRe): A randomized placebo controlled clinical trial. EClinicalMedicine 2021, 33, 100763. [Google Scholar] [CrossRef] [PubMed]
- Shiu, S.Y.; Law, I.C.; Lau, K.W.; Tam, P.C.; Yip, A.W.; Ng, W.T. Melatonin slowed the early biochemical progression of hormone-refractory prostate cancer in a patient whose prostate tumor tissue expressed MT1 receptor subtype. J. Pineal Res. 2003, 35, 177–182. [Google Scholar] [CrossRef] [PubMed]
- Neri, B.; de Leonardis, V.; Gemelli, M.T.; di Loro, F.; Mottola, A.; Ponchietti, R.; Raugei, A.; Cini, G. Melatonin as biological response modifier in cancer patients. Anticancer Res. 1998, 18, 1329–1332. [Google Scholar]
- Lissoni, P.; Brivio, F.; Fumagalli, L.; Messina, G.; Vigoré, L.; Parolini, D.; Colciago, M.; Rovelli, F. Neuroimmunomodulation in medical oncology: Application of psychoneuroimmunology with subcutaneous low-dose IL-2 and the pineal hormone melatonin in patients with untreatable metastatic solid tumors. Anticancer Res. 2008, 28, 1377–1381. [Google Scholar] [PubMed]
- Berk, L.; Berkey, B.; Rich, T.; Hrushesky, W.; Blask, D.; Gallagher, M.; Kudrimoti, M.; McGarry, R.C.; Suh, J.; Mehta, M. Randomized phase II trial of high-dose melatonin and radiation therapy for RPA class 2 patients with brain metastases (RTOG 0119). Int. J. Radiat. Oncol. Biol. Phys. 2007, 68, 852–857. [Google Scholar] [CrossRef] [PubMed]
- Sookprasert, A.; Johns, N.P.; Phunmanee, A.; Pongthai, P.; Cheawchanwattana, A.; Johns, J.; Konsil, J.; Plaimee, P.; Porasuphatana, S.; Jitpimolmard, S. Melatonin in patients with cancer receiving chemotherapy: A randomized, double-blind, placebo-controlled trial. Anticancer Res. 2014, 34, 7327–7337. [Google Scholar] [PubMed]
- Oh, S.N.; Myung, S.K.; Jho, H.J. Analgesic Efficacy of Melatonin: A Meta-Analysis of Randomized, Double-Blind, Placebo-Controlled Trials. J. Clin. Med. 2020, 9, 1553. [Google Scholar] [CrossRef] [PubMed]
- Sedighi Pashaki, A.; Mohammadian, K.; Afshar, S.; Gholami, M.H.; Moradi, A.; Javadinia, S.A.; Keshtpour Amlashi, Z. A Randomized, Controlled, Parallel-Group, Trial on the Effects of Melatonin on Fatigue Associated with Breast Cancer and Its Adjuvant Treatments. Integr. Cancer. Ther. 2021, 20, 1534735420988343. [Google Scholar] [CrossRef]
- Hansen, M.V.; Andersen, L.T.; Madsen, M.T.; Hageman, I.; Rasmussen, L.S.; Bokmand, S.; Rosenberg, J.; Gögenur, I. Effect of melatonin on depressive symptoms and anxiety in patients undergoing breast cancer surgery: A randomized, double-blind, placebo-controlled trial. Breast Cancer Res. Treat. 2014, 145, 683–695. [Google Scholar] [CrossRef]
- Zaki, N.F.; Sabri, Y.M.; Farouk, O.; Abdelfatah, A.; Spence, D.W.; Bahammam, A.S.; Pandi-Perumal, S. Depressive Symptoms, Sleep Profiles and Serum Melatonin Levels in a Sample of Breast Cancer Patients. Nat. Sci. Sleep 2020, 12, 135–149. [Google Scholar] [CrossRef] [PubMed]
- Innominato, P.F.; Lim, A.S.; Palesh, O.; Clemons, M.; Trudeau, M.; Eisen, A.; Wang, C.; Kiss, A.; Pritchard, K.I.; Bjarnason, G.A. The effect of melatonin on sleep and quality of life in patients with advanced breast cancer. Support. Care Cancer 2016, 24, 1097–10105. [Google Scholar] [CrossRef] [PubMed]
- Palmer, A.C.S.; Zortea, M.; Souza, A.; Santos, V.; Biazús, J.V.; Torres, I.L.S.; Fregni, F.; Caumo, W. Clinical impact of melatonin on breast cancer patients undergoing chemotherapy; effects on cognition, sleep and depressive symptoms: A randomized, double-blind, placebo-controlled trial. PLoS ONE 2020, 15, e0231379. [Google Scholar] [CrossRef] [PubMed]
- Fan, R.; Bu, X.; Yang, S.; Tan, Y.; Wang, T.; Chen, H.; Li, X. Effect of melatonin on quality of life and symptoms in patients with cancer: A systematic review and meta-analysis of randomised controlled trials. BMJ Open 2022, 12, e060912. [Google Scholar] [CrossRef] [PubMed]
- Gummadi, A.C.; Guddati, A.K. Genetic Polymorphisms in Pharmaceuticals and Chemotherapy. World J. Oncol. 2021, 12, 149–154. [Google Scholar] [CrossRef] [PubMed]
- Posadzki, P.P.; Bajpai, R.; Kyaw, B.M.; Roberts, N.J.; Brzezinski, A.; Christopoulos, G.I.; Divakar, U.; Bajpai, S.; Soljak, M.; Dunleavy, G.; et al. Melatonin and health: An umbrella review of health outcomes and biological mechanisms of action. BMC Med. 2018, 16, 18. [Google Scholar] [CrossRef] [PubMed]
- Reiter, R.J.; Rosales-Corral, S.A.; Tan, D.-X.; Acuna-Castroviejo, D.; Qin, L.; Yang, S.-F.; Xu, K. Melatonin, a Full Service Anti-Cancer Agent: Inhibition of Initiation, Progression and Metastasis. Int. J. Mol. Sci. 2017, 18, 843. [Google Scholar] [CrossRef] [PubMed]
- Cho, J.H.; Bhutani, S.; Kim, C.H.; Irwin, M.R. Anti-inflammatory effects of melatonin: A systematic review and meta-analysis of clinical trials. Brain Behav. Immun. 2021, 93, 245–253. [Google Scholar] [CrossRef] [PubMed]
- Hardeland, R. Melatonin in aging and disease -multiple consequences of reduced secretion, options and limits of treatment. Aging Dis. 2012, 3, 194–225. [Google Scholar] [PubMed]
- Griñan-Lison, C.; Blaya-Cánovas, J.L.; López-Tejada, A.; Ávalos-Moreno, M.; Navarro-Ocón, A.; Cara, F.E.; González-González, A.; Lorente, J.A.; Marchal, J.A.; Granados-Principal, S. Antioxidants for the Treatment of Breast Cancer: Are We There Yet? Antioxidants 2021, 10, 205. [Google Scholar] [CrossRef] [PubMed]
- Andersen, L.P.; Gögenur, I.; Rosenberg, J.; Reiter, R.J. The Safety of Melatonin in Humans. Clin. Drug Investig. 2016, 36, 169–175. [Google Scholar] [CrossRef] [PubMed]
- Besag, F.M.C.; Vasey, M.J.; Lao, K.S.J.; Wong, I.C.K. Adverse Events Associated with Melatonin for the Treatment of Primary or Secondary Sleep Disorders: A Systematic Review. CNS Drugs 2019, 33, 1167–1186. [Google Scholar] [CrossRef] [PubMed]
- Menczel Schrire, Z.; Phillips, C.L.; Chapman, J.L.; Duffy, S.L.; Wong, G.; D’Rozario, A.L.; Comas, M.; Raisin, I.; Saini, B.; Gordon, C.J.; et al. Safety of higher doses of melatonin in adults: A systematic review and meta-analysis. J. Pineal Res. 2022, 72, e12782. [Google Scholar] [CrossRef] [PubMed]
- Foley, H.M.; Steel, A.E. Adverse events associated with oral administration of melatonin: A critical systematic review of clinical evidence. Complement. Ther. Med. 2019, 42, 65–81. [Google Scholar] [CrossRef] [PubMed]
- Zetner, D.; Andersen, L.P.K.; Alder, R.; Jessen, M.L.; Tolstrup, A.; Rosenberg, J. Pharmacokinetics and Safety of Intravenous, Intravesical, Rectal, Transdermal, and Vaginal Melatonin in Healthy Female Volunteers: A Cross-Over Study. Pharmacology 2021, 106, 169–176. [Google Scholar] [CrossRef]
- Gooneratne, N.S.; Edwards, A.Y.; Zhou, C.; Cuellar, N.; Grandner, M.A.; Barrett, J.S. Melatonin pharmacokinetics following two different oral surge-sustained release doses in older adults. J. Pineal Res. 2012, 52, 437–445. [Google Scholar] [CrossRef] [PubMed]
- Wang, C.; Huo, X.; Tian, X.; Xu, M.; Dong, P.; Luan, Z.; Wang, X.; Zhang, B.; Zhang, B.; Huang, S.; et al. Inhibition of melatonin metabolism in humans induced by chemical components from herbs and effective prediction of this risk using a computational model. Br. J. Pharmacol. 2016, 173, 3261–3275. [Google Scholar] [CrossRef]
- Bayat Mokhtari, R.; Homayouni, T.S.; Baluch, N.; Morgatskaya, E.; Kumar, S.; Das, B.; Yeger, H. Combination therapy in combating cancer. Oncotarget 2017, 8, 38022–38043. [Google Scholar] [CrossRef] [PubMed]
- Muraro, E.; Vinante, L.; Fratta, E.; Bearz, A.; Höfler, D.; Steffan, A.; Baboci, L. Metronomic Chemotherapy: Anti-Tumor Pathways and Combination with Immune Checkpoint Inhibitors. Cancers 2023, 15, 2471. [Google Scholar] [CrossRef] [PubMed]
- Simsek, C.; Esin, E.; Yalcin, S. Metronomic Chemotherapy: A Systematic Review of the Literature and Clinical Experience. J. Oncol. 2019, 2019, 5483791. [Google Scholar] [CrossRef]
- Filippi, R.; Lombardi, P.; Depetris, I.; Fenocchio, E.; Quarà, V.; Chilà, G.; Aglietta, M.; Leone, F. Rationale for the use of metronomic chemotherapy in gastrointestinal cancer. Expert Opin. Pharmacother. 2018, 19, 1451–1463. [Google Scholar] [CrossRef] [PubMed]
- Cheng, S.-H.; Chiou, H.-Y.C.; Wang, J.-W.; Lin, M.-H. Reciprocal Regulation of Cancer-Associated Fibroblasts and Tumor Microenvironment in Gastrointestinal Cancer: Implications for Cancer Dormancy. Cancers 2023, 15, 2513. [Google Scholar] [CrossRef] [PubMed]
- Nicolas, A.; Carré, M.; Pasquier, E. Metronomics: Intrinsic Anakoinosis Modulator? Front. Pharmacol. 2018, 9, 689. [Google Scholar] [CrossRef] [PubMed]
- Sauer, S.; Reed, D.R.; Ihnat, M.; Hurst, R.E.; Warshawsky, D.; Barkan, D. Innovative Approaches in the Battle against Cancer Recurrence: Novel Strategies to Combat Dormant Disseminated Tumor Cells. Front. Oncol. 2021, 11, 659963. [Google Scholar] [CrossRef] [PubMed]
- Hardeland, R. Melatonin and the Programming of Stem Cells. Int. J. Mol. Sci. 2022, 23, 1971. [Google Scholar] [CrossRef] [PubMed]
- Samec, M.; Liskova, A.; Koklesova, L.; Zhai, K.; Varghese, E.; Samuel, S.M.; Šudomová, M.; Lucansky, V.; Kassayova, M.; Pec, M.; et al. Metabolic Anti-Cancer Effects of Melatonin: Clinically Relevant Prospects. Cancers 2021, 13, 3018. [Google Scholar] [CrossRef] [PubMed]
- Skrajnowska, D.; Bobrowska-Korczak, B. Role of Zinc in Immune System and Anti-Cancer Defense Mechanisms. Nutrients 2019, 11, 2273. [Google Scholar] [CrossRef] [PubMed]
- Gandin, V.; Khalkar, P.; Braude, J.; Fernandes, A.P. Organic selenium compounds as potential chemotherapeutic agents for improved cancer treatment. Free Radic. Biol. Med. 2018, 127, 80–97. [Google Scholar] [CrossRef] [PubMed]
- Wessels, I.; Maywald, M.; Rink, L. Zinc as a Gatekeeper of Immune Function. Nutrients 2017, 9, 1286. [Google Scholar] [CrossRef] [PubMed]
- Franklin, R.B.; Costello, L.C. The important role of the apoptotic effects of zinc in the development of cancers. J. Cell. Biochem. 2009, 106, 750–757. [Google Scholar] [CrossRef] [PubMed]
- Liu, S.; Sima, X.; Liu, X.; Chen, H. Zinc Finger Proteins: Functions and Mechanisms in Colon Cancer. Cancers 2022, 14, 5242. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Cao, D.; Huang, Y.; Chen, B.; Chen, Z.; Wang, R.; Dong, Q.; Wei, Q.; Liu, L. Zinc Intakes and Health Outcomes: An Umbrella Review. Front. Nutr. 2022, 9, 798078. [Google Scholar] [CrossRef] [PubMed]
- Hosui, A.; Kimura, E.; Abe, S.; Tanimoto, T.; Onishi, K.; Kusumoto, Y.; Sueyoshi, Y.; Matsumoto, K.; Hirao, M.; Yamada, T.; et al. Long-Term Zinc Supplementation Improves Liver Function and Decreases the Risk of Developing Hepatocellular Carcinoma. Nutrients 2018, 10, 1955. [Google Scholar] [CrossRef] [PubMed]
- Lubiński, J.; Jaworowska, E.; Derkacz, R.; Marciniak, W.; Białkowska, K.; Baszuk, P.; Scott, R.J.; Lubiński, J.A. Survival of Laryngeal Cancer Patients Depending on Zinc Serum Level and Oxidative Stress Genotypes. Biomolecules 2021, 11, 865. [Google Scholar] [CrossRef] [PubMed]
- Nakanishi, K.; Toyoshima, M.; Ichikawa, G.; Suzuki, S. Zinc deficiency is associated with gynecologic cancer recurrence. Front. Oncol. 2022, 12, 1025060. [Google Scholar] [CrossRef] [PubMed]
- Lin, Y.S.; Lin, L.C.; Lin, S.W. Effects of zinc supplementation on the survival of patients who received concomitant chemotherapy and radiotherapy for advanced nasopharyngeal carcinoma: Follow-up of a double-blind randomized study with subgroup analysis. Laryngoscope 2009, 119, 1348–1352. [Google Scholar] [CrossRef] [PubMed]
- Hoppe, C.; Kutschan, S.; Dörfler, J.; Büntzel, J.; Büntzel, J.; Huebner, J. Zinc as a complementary treatment for cancer patients: A systematic review. Clin. Exp. Med. 2021, 21, 297–313. [Google Scholar] [CrossRef] [PubMed]
- Ribeiro, S.M.; Braga, C.B.; Peria, F.M.; Domenici, F.A.; Martinez, E.Z.; Feres, O.; da Rocha, J.J.; da Cunha, S.F. Effect of Zinc Supplementation on Antioxidant Defenses and Oxidative Stress Markers in Patients Undergoing Chemotherapy for Colorectal Cancer: A Placebo-Controlled, Prospective Randomized Trial. Biol. Trace Elem. Res. 2016, 169, 8–16. [Google Scholar] [CrossRef]
- Sun, Y.; Wang, Z.; Gong, P.; Yao, W.; Ba, Q.; Wang, H. Review on the health-promoting effect of adequate selenium status. Front. Nutr. 2023, 10, 1136458. [Google Scholar] [CrossRef] [PubMed]
- Misra, S.; Boylan, M.; Selvam, A.; Spallholz, J.E.; Björnstedt, M. Redox-Active Selenium Compounds—From Toxicity and Cell Death to Cancer Treatment. Nutrients 2015, 7, 3536–3556. [Google Scholar] [CrossRef] [PubMed]
- Radomska, D.; Czarnomysy, R.; Radomski, D.; Bielawski, K. Selenium Compounds as Novel Potential Anticancer Agents. Int. J. Mol. Sci. 2021, 22, 1009. [Google Scholar] [CrossRef] [PubMed]
- Avery, J.C.; Hoffmann, P.R. Selenium, Selenoproteins, and Immunity. Nutrients 2018, 10, 1203. [Google Scholar] [CrossRef] [PubMed]
- Zakharia, Y.; Bhattacharya, A.; Rustum, Y.M. Selenium targets resistance biomarkers enhancing efficacy while reducing toxicity of anti-cancer drugs: Preclinical and clinical development. Oncotarget 2018, 9, 10765–10783. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.J.; Choi, M.C.; Park, J.M.; Chung, A.S. Antitumor Effects of Selenium. Int. J. Mol. Sci. 2021, 22, 11844. [Google Scholar] [CrossRef] [PubMed]
- Fakih, M.G.; Pendyala, L.; Brady, W.; Smith, P.F.; Ross, M.E.; Creaven, P.J.; Badmaev, V.; Prey, J.D.; Rustum, Y.M. A Phase I and pharmacokinetic study of selenomethionine in combination with a fixed dose of irinotecan in solid tumors. Cancer Chemother. Pharmacol. 2008, 62, 499–508. [Google Scholar] [CrossRef] [PubMed]
- Fakih, M.G.; Pendyala, L.; Smith, P.F.; Creaven, P.J.; Reid, M.E.; Badmaev, V.; Azrak, R.G.; Prey, J.D.; Lawrence, D.; Rustum, Y.M. A phase I and pharmacokinetic study of fixed-dose selenomethionine and irinotecan in solid tumors. Clin. Cancer Res. 2006, 12, 1237–1244. [Google Scholar] [CrossRef] [PubMed]
- Karamali, M.; Nourgostar, S.; Zamani, A.; Vahedpoor, Z.; Asemi, Z. The favourable effects of long-term selenium supplementation on regression of cervical tissues and metabolic profiles of patients with cervical intraepithelial neoplasia: A randomised, double-blind, placebo-controlled trial. Br. J. Nutr. 2015, 114, 2039–2045. [Google Scholar] [CrossRef] [PubMed]
- Brodin, O.; Eksborg, S.; Wallenberg, M.; Asker-Hagelberg, C.; Larsen, E.H.; Mohlkert, D.; Lenneby-Helleday, C.; Jacobsson, H.; Linder, S.; Misra, S.; et al. Pharmacokinetics and Toxicity of Sodium Selenite in the Treatment of Patients with Carcinoma in a Phase I Clinical Trial: The SECAR Study. Nutrients 2015, 7, 4978–4994. [Google Scholar] [CrossRef] [PubMed]
- Song, M.; Kumaran, M.N.; Gounder, M.; Gibbon, D.G.; Nieves-Neira, W.; Vaidya, A.; Hellmann, M.; Kane, M.P.; Buckley, B.; Shih, W.; et al. Phase I trial of selenium plus chemotherapy in gynecologic cancers. Gynecol. Oncol. 2018, 150, 478–486. [Google Scholar] [CrossRef] [PubMed]
- Short, S.P.; Pilat, J.M.; Williams, C.S. Roles for selenium and selenoprotein P in the development, progression, and prevention of intestinal disease. Free Radic. Biol. Med. 2018, 127, 26–35. [Google Scholar] [CrossRef] [PubMed]
- Psathakis, D.; Wedemeyer, N.; Oevermann, E.; Krug, F.; Siegers, C.P.; Bruch, H.P. Blood selenium and glutathione peroxidase status in patients with colorectal cancer. Dis. Colon Rectum. 1998, 41, 328–335. [Google Scholar] [CrossRef] [PubMed]
- Demircan, K.; Bengtsson, Y.; Sun, Q.; Brange, A.; Vallon-Christersson, J.; Rijntjes, E.; Malmberg, M.; Saal, L.H.; Rydén, L.; Borg, Å.; et al. Serum selenium, selenoprotein P and glutathione peroxidase 3 as predictors of mortality and recurrence following breast cancer diagnosis: A multicentre cohort study. Redox Biol. 2021, 47, 102145. [Google Scholar] [CrossRef] [PubMed]
- Muecke, R.; Micke, O.; Schomburg, L.; Buentzel, J.; Kisters, K.; Adamietz, I.A.; On behalf of AKTE. Selenium in Radiation Oncology—15 Years of Experiences in Germany. Nutrients 2018, 10, 483. [Google Scholar] [CrossRef] [PubMed]
- Handa, E.; Puspitasari, I.M.; Abdulah, R.; Yamazaki, C.; Kameo, S.; Nakano, T.; Koyama, H. Recent advances in clinical studies of selenium supplementation in radiotherapy. J. Trace Elem. Med. Biol. 2020, 62, 126653. [Google Scholar] [CrossRef] [PubMed]
- Knox, S.J.; Jayachandran, P.; Keeling, C.A.; Stevens, K.J.; Sandhu, N.; Stamps-DeAnda, S.L.; Savic, R.; Shura, L.; Buyyounouski, M.K.; Grimes, K. Results from a Phase 1 Study of Sodium Selenite in Combination with Palliative Radiation Therapy in Patients with Metastatic Cancer. Transl. Oncol. 2019, 12, 1525–1531. [Google Scholar] [CrossRef] [PubMed]
- Garbo, S.; Di Giacomo, S.; Łażewska, D.; Honkisz-Orzechowska, E.; Di Sotto, A.; Fioravanti, R.; Zwergel, C.; Battistelli, C. Selenium-Containing Agents Acting on Cancer-A New Hope? Pharmaceutics 2022, 15, 104. [Google Scholar] [CrossRef] [PubMed]
- Ivory, K.; Prieto, E.; Spinks, C.; Armah, C.N.; Goldson, A.J.; Dainty, J.R.; Nicoletti, C. Selenium supplementation has beneficial and detrimental effects on immunity to influenza vaccine in older adults. Clin. Nutr. 2017, 36, 407–415. [Google Scholar] [CrossRef] [PubMed]
- Kenfield, S.A.; Van Blarigan, E.L.; DuPre, N.; Stampfer, M.J.; L Giovannucci, E.; Chan, J.M. Selenium supplementation and prostate cancer mortality. J. Natl. Cancer. Inst. 2014, 107, 360. [Google Scholar] [CrossRef] [PubMed]
- Barchielli, G.; Capperucci, A.; Tanini, D. The Role of Selenium in Pathologies: An Updated Review. Antioxidants 2022, 11, 251. [Google Scholar] [CrossRef] [PubMed]
- Wu, X.; Hu, W.; Lu, L.; Zhao, Y.; Zhou, Y.; Xiao, Z.; Zhang, L.; Zhang, H.; Li, X.; Li, W.; et al. Repurposing vitamin D for treatment of human malignancies via targeting tumor microenvironment. Acta Pharm. Sin. B 2019, 9, 203–219. [Google Scholar] [CrossRef] [PubMed]
- Jeon, S.M.; Shin, E.A. Exploring vitamin D metabolism and function in cancer. Exp. Mol. Med. 2018, 50, 1–14. [Google Scholar] [CrossRef] [PubMed]
- Huang, D.; Lei, S.; Wu, Y.; Weng, M.; Zhou, Y.; Xu, J.; Xia, D.; Xu, E.; Lai, M.; Zhang, H. Additively protective effects of vitamin D and calcium against colorectal adenoma incidence, malignant transformation and progression: A systematic review and meta-analysis. Clin. Nutr. 2020, 39, 2525–2538. [Google Scholar] [CrossRef] [PubMed]
- Na, S.Y.; Kim, K.B.; Lim, Y.J.; Song, H.J. Vitamin D and Colorectal Cancer: Current Perspectives and Future Directions. J. Cancer Prev. 2022, 27, 147–156. [Google Scholar] [CrossRef]
- Yuan, C.; Sato, K.; Hollis, B.W.; Zhang, S.; Niedzwiecki, D.; Ou, F.S.; Chang, I.W.; O’Neil, B.H.; Innocenti, F.; Lenz, H.J.; et al. Plasma 25-Hydroxyvitamin D Levels and Survival in Patients with Advanced or Metastatic Colorectal Cancer: Findings from CALGB/SWOG 80405 (Alliance). Clin. Cancer Res. 2019, 25, 7497–7505. [Google Scholar] [CrossRef] [PubMed]
- Zhou, J.; Ge, X.; Fan, X.; Wang, J.; Miao, L.; Hang, D. Associations of vitamin D status with colorectal cancer risk and survival. Int. J. Cancer 2021, 149, 606–614. [Google Scholar] [CrossRef]
- Emmanouilidou, G.; Kalopitas, G.; Bakaloudi, D.R.; Karanika, E.; Theocharidou, E.; Germanidis, G.; Chourdakis, M. Vitamin D as a chemopreventive agent in colorectal neoplasms. A systematic review and meta-analysis of randomized controlled trials. Pharmacol. Ther. 2022, 237, 108252. [Google Scholar] [CrossRef] [PubMed]
- Akutsu, T.; Okada, S.; Hirooka, S.; Ikegami, M.; Ohdaira, H.; Suzuki, Y.; Urashima, M. Effect of Vitamin D on Relapse-Free Survival in a Subgroup of Patients with p53 Protein-Positive Digestive Tract Cancer: A Post Hoc Analysis of the AMATERASU Trial. Cancer Epidemiol. Biomarkers Prev. 2020, 29, 406–413. [Google Scholar] [CrossRef] [PubMed]
- Yonaga, H.; Okada, S.; Akutsu, T.; Ohdaira, H.; Suzuki, Y.; Urashima, M. Effect Modification of Vitamin D Supplementation by Histopathological Characteristics on Survival of Patients with Digestive Tract Cancer: Post Hoc Analysis of the AMATERASU Randomized Clinical Trial. Nutrients 2019, 11, 2547. [Google Scholar] [CrossRef] [PubMed]
- Barry, E.L.; Peacock, J.L.; Rees, J.R.; Bostick, R.M.; Robertson, D.J.; Bresalier, R.S.; Baron, J.A. Vitamin D Receptor Genotype, Vitamin D3 Supplementation, and Risk of Colorectal Adenomas: A Randomized Clinical Trial. JAMA Oncol. 2017, 3, 628–635. [Google Scholar] [CrossRef] [PubMed]
- Chandler, P.D.; Chen, W.Y.; Ajala, O.N.; Hazra, A.; Cook, N.; Bubes, V.; Lee, I.M.; Giovannucci, E.L.; Willett, W.; Buring, J.E.; et al. Effect of Vitamin D3 Supplements on Development of Advanced Cancer: A Secondary Analysis of the VITAL Randomized Clinical Trial. JAMA Netw. Open 2020, 3, e2025850. [Google Scholar] [CrossRef] [PubMed]
- Cháirez-Ramírez, M.H.; de la Cruz-López, K.G.; García-Carrancá, A. Polyphenols as Antitumor Agents Targeting Key Players in Cancer-Driving Signaling Pathways. Front. Pharmacol. 2021, 12, 710304. [Google Scholar] [CrossRef] [PubMed]
- Farhan, M.; Rizvi, A. Understanding the Prooxidant Action of Plant Polyphenols in the Cellular Microenvironment of Malignant Cells: Role of Copper and Therapeutic Implications. Front. Pharmacol. 2022, 13, 929853. [Google Scholar] [CrossRef] [PubMed]
- Sharma, E.; Attri, D.C.; Sati, P.; Dhyani, P.; Szopa, A.; Sharifi-Rad, J.; Hano, C.; Calina, D.; Cho, W.C. Recent updates on anticancer mechanisms of polyphenols. Front. Cell. Dev. Biol. 2022, 10, 1005910. [Google Scholar] [CrossRef] [PubMed]
- Wang, M.; Liu, X.; Chen, T.; Cheng, X.; Xiao, H.; Meng, X.; Jiang, Y. Inhibition and potential treatment of colorectal cancer by natural compounds via various signaling pathways. Front. Oncol. 2022, 12, 956793. [Google Scholar] [CrossRef] [PubMed]
- Gavrilas, L.I.; Cruceriu, D.; Mocan, A.; Loghin, F.; Miere, D.; Balacescu, O. Plant-Derived Bioactive Compounds in Colorectal Cancer: Insights from Combined Regimens with Conventional Chemotherapy to Overcome Drug-Resistance. Biomedicines 2022, 10, 1948. [Google Scholar] [CrossRef] [PubMed]
- Pochet, S.; Lechon, A.S.; Lescrainier, C.; De Vriese, C.; Mathieu, V.; Hamdani, J.; Souard, F. Herb-anticancer drug interactions in real life based on VigiBase, the WHO global database. Sci. Rep. 2022, 12, 14178. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Zhang, T.; Chen, G.Y. Flavonoids and Colorectal Cancer Prevention. Antioxidants 2018, 7, 187. [Google Scholar] [CrossRef] [PubMed]
- Shin, C.M.; Lee, D.H.; Seo, A.Y.; Lee, H.J.; Kim, S.B.; Son, W.C.; Kim, Y.K.; Lee, S.J.; Park, S.H.; Kim, N.; et al. Green tea extracts for the prevention of metachronous colorectal polyps among patients who underwent endoscopic removal of colorectal adenomas: A randomized clinical trial. Clin. Nutr. 2018, 37, 452–458. [Google Scholar] [CrossRef] [PubMed]
- Seufferlein, T.; Ettrich, T.; Menzler, S.; Messmann, H.; Kleber, G.; Zipprich, A.; Frank-Gleich, S.; Algül, H.; Metter, K.; Odemar, F.; et al. MIRACLE: Green tea extract versus placebo for the prevention of colorectal adenomas: A randomized, controlled trial. Ann. Oncol. 2019, 30, v869. [Google Scholar] [CrossRef]
- Bracci, L.; Fabbri, A.; Del Cornò, M.; Conti, L. Dietary Polyphenols: Promising Adjuvants for Colorectal Cancer Therapies. Cancers 2021, 13, 4499. [Google Scholar] [CrossRef] [PubMed]
- Cueva, C.; Silva, M.; Pinillos, I.; Bartolomé, B.; Moreno-Arribas, M.V. Interplay between Dietary Polyphenols and Oral and Gut Microbiota in the Development of Colorectal Cancer. Nutrients 2020, 12, 625. [Google Scholar] [CrossRef]
- Scholl, C.; Lepper, A.; Lehr, T.; Hanke, N.; Schneider, K.L.; Brockmöller, J.; Seufferlein, T.; Stingl, J.C. Population nutrikinetics of green tea extract. PLoS ONE 2018, 13, e0193074. [Google Scholar] [CrossRef] [PubMed]
- Almatroodi, S.A.; Almatroudi, A.; Khan, A.A.; Alhumaydhi, F.A.; Alsahli, M.A.; Rahmani, A.H. Potential Therapeutic Targets of Epigallocatechin Gallate (EGCG), the Most Abundant Catechin in Green Tea, and Its Role in the Therapy of Various Types of Cancer. Molecules 2020, 25, 3146. [Google Scholar] [CrossRef] [PubMed]
- Farhan, M. Green Tea Catechins: Nature’s Way of Preventing and Treating Cancer. Int. J. Mol. Sci. 2022, 23, 10713. [Google Scholar] [CrossRef] [PubMed]
- Oketch-Rabah, H.A.; Roe, A.L.; Rider, C.V.; Bonkovsky, H.L.; Giancaspro, G.I.; Navarro, V.; Paine, M.F.; Betz, J.M.; Marles, R.J.; Casper, S.; et al. United States Pharmacopeia (USP) comprehensive review of the hepatotoxicity of green tea extracts. Toxicol. Rep. 2020, 7, 386–402. [Google Scholar] [CrossRef] [PubMed]
- Wang, D.; Wei, Y.; Wang, T.; Wan, X.; Yang, C.S.; Reiter, R.J.; Zhang, J. Melatonin attenuates (-)-epigallocatehin-3-gallate-triggered hepatotoxicity without compromising its downregulation of hepatic gluconeogenic and lipogenic genes in mice. J. Pineal Res. 2015, 59, 497–507. [Google Scholar] [CrossRef]
- Zhang, L.; He, Y.; Wu, X.; Zhao, G.; Zhang, K.; Yang, C.S.; Reiter, R.J.; Zhang, J. Melatonin and (-)-Epigallocatechin-3-Gallate: Partners in Fighting Cancer. Cells 2019, 8, 745. [Google Scholar] [CrossRef] [PubMed]
- Sunil, C.; Xu, B. An insight into the health-promoting effects of taxifolin (dihydroquercetin). Phytochemistry 2019, 166, 112066. [Google Scholar] [CrossRef] [PubMed]
- Das, A.; Baidya, R.; Chakraborty, T.; Samanta, A.K.; Roy, S. Pharmacological basis and new insights of taxifolin: A comprehensive review. Biomed. Pharmacother. 2021, 142, 112004. [Google Scholar] [CrossRef] [PubMed]
- Wang, R.; Zhu, X.; Wang, Q.; Li, X.; Wang, E.; Zhao, Q.; Wang, Q.; Cao, H. The anti-tumor effect of taxifolin on lung cancer via suppressing stemness and epithelial-mesenchymal transition in vitro and oncogenesis in nude mice. Ann. Transl. Med. 2020, 8, 590. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Hu, L.; Zhou, T.; Gong, X.; Jiang, R.; Li, H.; Kuang, G.; Wan, J.; Li, H. Taxifolin inhibits breast cancer cells proliferation, migration and invasion by promoting mesenchymal to epithelial transition via β-catenin signaling. Life Sci. 2019, 232, 116617. [Google Scholar] [CrossRef] [PubMed]
- Xie, J.; Pang, Y.; Wu, X. Taxifolin suppresses the malignant progression of gastric cancer by regulating the AhR/CYP1A1 signaling pathway. Int. J. Mol. Med. 2021, 48, 197. [Google Scholar] [CrossRef] [PubMed]
- Razak, S.; Afsar, T.; Ullah, A.; Almajwal, A.; Alkholief, M.; Alshamsan, A.; Jahan, S. Taxifolin, a natural flavonoid interacts with cell cycle regulators causes cell cycle arrest and causes tumor regression by activating Wnt/β-catenin signaling pathway. BMC Cancer 2018, 18, 1043. [Google Scholar] [CrossRef] [PubMed]
- Inoue, T.; Fu, B.; Nishio, M.; Tanaka, M.; Kato, H.; Tanaka, M.; Itoh, M.; Yamakage, H.; Ochi, K.; Ito, A.; et al. Novel Therapeutic Potentials of Taxifolin for Obesity-Induced Hepatic Steatosis, Fibrogenesis, and Tumorigenesis. Nutrients 2023, 15, 350. [Google Scholar] [CrossRef] [PubMed]
- Alanezi, A.A.; Almuqati, A.F.; Alfwuaires, M.A.; Alasmari, F.; Namazi, N.I.; Althunibat, O.Y.; Mahmoud, A.M. Taxifolin Prevents Cisplatin Nephrotoxicity by Modulating Nrf2/HO-1 Pathway and Mitigating Oxidative Stress and Inflammation in Mice. Pharmaceuticals 2022, 15, 1310. [Google Scholar] [CrossRef]
- Unver, E.; Tosun, M.; Olmez, H.; Kuzucu, M.; Cimen, F.K.; Suleyman, Z. The Effect of Taxifolin on Cisplatin-Induced Pulmonary Damage in Rats: A Biochemical and Histopathological Evaluation. Mediat. Inflamm. 2019, 2019, 3740867. [Google Scholar] [CrossRef] [PubMed]
- Kurt, N.; Türkeri, Ö.N.; Suleyman, B.; Bakan, N. The effect of taxifolin on high-dose-cisplatin-induced oxidative liver injury in rats. Adv. Clin. Exp. Med. 2021, 30, 1025–1030. [Google Scholar] [CrossRef] [PubMed]
- Chen, H.-J.; Chung, Y.-L.; Li, C.-Y.; Chang, Y.-T.; Wang, C.C.N.; Lee, H.-Y.; Lin, H.-Y.; Hung, C.-C. Taxifolin Resensitizes Multidrug Resistance Cancer Cells via Uncompetitive Inhibition of P-Glycoprotein Function. Molecules 2018, 23, 3055. [Google Scholar] [CrossRef] [PubMed]
- Orlova, S.V.; Tatarinov, V.V.; Nikitina, E.A.; Sheremeta, A.V.; Ivlev, V.A.; Vasil’ev, V.G.; Paliy, K.V.; Goryainov, S.V. Bioavailability and Safety of Dihydroquercetin (Review). Pharm. Chem. J. 2022, 55, 1133–1137. [Google Scholar] [CrossRef] [PubMed]
- Tao, D.L.; Tassi Yunga, S.; Williams, C.D.; McCarty, O.J.T. Aspirin and antiplatelet treatments in cancer. Blood 2021, 137, 3201–3211. [Google Scholar] [CrossRef] [PubMed]
- Elwood, P.; Protty, M.; Morgan, G.; Pickering, J.; Delon, C.; Watkins, J. Aspirin and cancer: Biological mechanisms and clinical outcomes. Open Biol. 2022, 12, 220124. [Google Scholar] [CrossRef] [PubMed]
- Li, P.; Ning, Y.; Li, M.; Cai, P.; Siddiqui, A.D.; Liu, E.Y.; Hadley, M.; Wu, F.; Pan, S.; Dixon, R.A.F.; et al. Aspirin Is Associated with Reduced Rates of Venous Thromboembolism in Older Patients with Cancer. J. Cardiovasc. Pharmacol. Ther. 2020, 25, 456–465. [Google Scholar] [CrossRef] [PubMed]
- Veettil, S.K.; Lim, K.G.; Ching, S.M.; Saokaew, S.; Phisalprapa, P.; Chaiyakunapruk, N. Effects of aspirin and non-aspirin nonsteroidal anti-inflammatory drugs on the incidence of recurrent colorectal adenomas: A systematic review with meta-analysis and trial sequential analysis of randomized clinical trials. BMC Cancer 2017, 17, 763. [Google Scholar] [CrossRef] [PubMed]
- Loomans-Kropp, H.A.; Pinsky, P.; Cao, Y.; Chan, A.T.; Umar, A. Association of Aspirin Use with Mortality Risk among Older Adult Participants in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. JAMA Netw. Open 2019, 2, e1916729. [Google Scholar] [CrossRef] [PubMed]
- Elwood, P.C.; Morgan, G.; Delon, C.; Protty, M.; Galante, J.; Pickering, J.; Watkins, J.; Weightman, A.; Morris, D. Aspirin and cancer survival: A systematic review and meta-analyses of 118 observational studies of aspirin and 18 cancers. Ecancermedicalscience 2021, 15, 1258. [Google Scholar] [CrossRef] [PubMed]
- McNeil, J.J.; Gibbs, P.; Orchard, S.G.; Lockery, J.E.; Bernstein, W.B.; Cao, Y.; Ford, L.; Haydon, A.; Kirpach, B.; Macrae, F.; et al. Effect of Aspirin on Cancer Incidence and Mortality in Older Adults. J. Natl. Cancer Inst. 2021, 113, 258–265. [Google Scholar] [CrossRef] [PubMed]
- Konturek, P.C.; Konturek, S.J.; Celinski, K.; Slomka, M.; Cichoz-Lach, H.; Bielanski, W.; Reiter, R.J. Role of melatonin in mucosal gastroprotection against aspirin-induced gastric lesions in humans. J. Pineal Res. 2010, 48, 318–323. [Google Scholar] [CrossRef] [PubMed]
- Gouda, M.A.; Buschhorn, L.; Schneeweiss, A.; Wahida, A.; Subbiah, V. N-of-1 Trials in Cancer Drug Development. Cancer Discov. 2023, 13, 1301–1309. [Google Scholar] [CrossRef] [PubMed]
- Samuel, J.P.; Wootton, S.H.; Tyson, J.E. N-of-1 trials: The epitome of personalized medicine? J. Clin. Transl. Sci. 2023, 7, e161. [Google Scholar] [CrossRef] [PubMed]
- Bradbury, J.; Avila, C.; Grace, S. Practice-Based Research in Complementary Medicine: Could N-of-1 Trials Become the New Gold Standard? Healthcare 2020, 8, 15. [Google Scholar] [CrossRef]
- Schork, N.J.; Beaulieu-Jones, B.; Liang, W.S.; Smalley, S.; Goetz, L.H. Exploring human biology with N-of-1 clinical trials. Camb. Prism. Precis. Med. 2023, 1, e12. [Google Scholar] [CrossRef]
- Punja, S.; Nikles, C.J.; Senior, H.; Mitchell, G.; Schmid, C.H.; Heussler, H.; Witmans, M.; Vohra, S. Melatonin in Youth: N-of-1 trials in a stimulant-treated ADHD Population (MYNAP): Study protocol for a randomized controlled trial. Trials 2016, 17, 375. [Google Scholar] [CrossRef] [PubMed]
- Saadeh, C.; Bright, D.; Rustem, D. Precision Medicine in Oncology Pharmacy Practice. Acta Med. Acad. 2019, 48, 90–104. [Google Scholar] [CrossRef]
- Karim, S.; Benn, R.; Carlson, L.E.; Fouladbakhsh, J.; Greenlee, H.; Harris, R.; Henry, N.L.; Jolly, S.; Mayhew, S.; Spratke, L.; et al. Integrative Oncology Education: An Emerging Competency for Oncology Providers. Curr. Oncol. 2021, 28, 853–862. [Google Scholar] [CrossRef] [PubMed]
- Blackston, J.W.; Chapple, A.G.; McGree, J.M.; McDonald, S.; Nikles, J. Comparison of Aggregated N-of-1 Trials with Parallel and Crossover Randomized Controlled Trials Using Simulation Studies. Healthcare 2019, 7, 137. [Google Scholar] [CrossRef] [PubMed]
- Vanderhout, S.; Nicholls, S.; Monfaredi, Z.; Hampel, C.; Ashdown, L.; Bilodeau, M.; Rich, S.; Shea, B.; Fergusson, D. Facilitating and supporting the engagement of patients, families and caregivers in research: The “Ottawa model” for patient engagement in research. Res. Involv. Engagem. 2022, 8, 25. [Google Scholar] [CrossRef] [PubMed]
Clinical Parameters | Before Surgery | After Surgery | During and after CT | 54 Months after Diagnosis | Reference Ranges |
---|---|---|---|---|---|
Neutrophils * | 3.90 | 3.08 | 2.00–1.69–1.85 | 3.46 | 2.20–7.60 |
Lymphocytes * | 1.38 | 1.50 | 1.85–2.61–2.05 | 1.96 | 1.00–3.60 |
Monocytes * | 0.71 | 0.61 | 0.70–1.01–0.61 | 0.64 | 0.20–1.00 |
Platelets * | 164 | 145 | 75–227–96–110 | 142 | 150–400 |
NLR | 2.83 | 2.05 | 0.77–0.91–1.61–0.92 | 1.77 | <2.72 |
LMR | 1.94 | 2.46 | 2.58–3.36–2.43–4.79 | 3.06 | >2.83 |
PLR | 118.8 | 96.7 | 40.5–106.1–41.0–96.3 | 72.5 | <219.0 |
C-reactive protein, mg/L | <1.0 | 11.0 | <1.0 | 1.0 | <5.0 |
Eosinophils * | 0.05 | 0.30–0.18 | 0.22–0.34–0.20 | 0.16 | 0.10–0.40 |
RBC, number × 1012/L | 4.35 | 3.83 | 3.72–3.39–3.56 | 4.08 | 4.50–6.00 |
Hb, g/L | 136 | 116 | 113–117 | 128 | 130–180 |
CEA, μg/L | 6 | 1.4 | 1.2–1.7 | 1.70 | <3.8 ** |
CA 19-9, kU/L | 13 | 6 | 7–9 | 8 | <27 |
PSA, μg/L | 5.70 | 4.62–4.17–7.61–5.28 | 6.40 | <4.00 | |
D-dimer, mg/L | 0.61 | 1.86–1.43–0.43 | <0.50 | ||
APTT ***, s | 38.7 | 42.9 | 36.6–39.2–44.2 | 28.6–38.2 |
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. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Smorodin, E.; Chuzmarov, V.; Veidebaum, T. The Potential of Integrative Cancer Treatment Using Melatonin and the Challenge of Heterogeneity in Population-Based Studies: A Case Report of Colon Cancer and a Literature Review. Curr. Oncol. 2024, 31, 1994-2023. https://doi.org/10.3390/curroncol31040149
Smorodin E, Chuzmarov V, Veidebaum T. The Potential of Integrative Cancer Treatment Using Melatonin and the Challenge of Heterogeneity in Population-Based Studies: A Case Report of Colon Cancer and a Literature Review. Current Oncology. 2024; 31(4):1994-2023. https://doi.org/10.3390/curroncol31040149
Chicago/Turabian StyleSmorodin, Eugeniy, Valentin Chuzmarov, and Toomas Veidebaum. 2024. "The Potential of Integrative Cancer Treatment Using Melatonin and the Challenge of Heterogeneity in Population-Based Studies: A Case Report of Colon Cancer and a Literature Review" Current Oncology 31, no. 4: 1994-2023. https://doi.org/10.3390/curroncol31040149
APA StyleSmorodin, E., Chuzmarov, V., & Veidebaum, T. (2024). The Potential of Integrative Cancer Treatment Using Melatonin and the Challenge of Heterogeneity in Population-Based Studies: A Case Report of Colon Cancer and a Literature Review. Current Oncology, 31(4), 1994-2023. https://doi.org/10.3390/curroncol31040149