The Multifaceted Role of Alpha-Lipoic Acid in Cancer Prevention, Occurrence, and Treatment
Abstract
:1. Introduction
2. ALA and Tumor Prevention
2.1. Role in Maintaining Redox Homeostasis in the Body
- ALA and DHLA exhibit a high redox potential (320 mV), which enables more effective maintenance of the reductive state within the body compared with that observed for the endogenous reduced/oxidized glutathione (GSH/GSSG) system (240 mV). Additionally, these molecules interfere with various signaling pathways activated by oxidative stress, preventing the excessive production of reactive oxygen species [4,23];
- ALA can directly quench various reactive species, including ROS, reactive nitrogen species, hydroxyl radicals (HO•), hypochlorous acid (HclO), and singlet oxygen (1O2); it provides indirect antioxidant protection through metal chelation (ALA primarily binds Cu2+ and Zn2+, while DHLA can bind Cu2+, Zn2+, Pb2+, Hg2+, and Fe3+) and the regeneration of certain endogenous antioxidants, such as vitamin E, vitamin C, and glutathione [4,5,23];
- Many antioxidants exhibit limited bioavailability due to their instability in the bloodstream or hydrophilicity, which restricts their passage through cell membranes. However, ALA and DHLA exhibit amphipathic properties, allowing them to be distributed in both hydrophilic environments (e.g., plasma) and lipophilic environments (e.g., cell membranes). This enables ALA to function extracellularly and to protect key intracellular molecules [4] (Figure 2).Figure 2. Sources of ROS and the antioxidative effects of ALA. Sources of ROS: The blue section on the left side of the figure outlines the extracellular and intracellular sources of ROS production. Extracellular sources of ROS include environmental pollutants, radiation, toxic stimuli, microbial infections, and the inflammation they induce. Intracellularly, both normal and abnormal cellular activities can induce ROS production, including mitochondrial oxidative phosphorylation, ER stress and the UPR, enzymatic reactions, peroxisome metabolism, and NADPH oxidase activity. Antioxidative effects of ALA: The red section on the right side of the figure summarizes the role of ALA as a multifunctional antioxidant in protecting cells from excessive ROS damage. ALA can directly quench various reactive oxygen species, including ROS, RNS, HO•, HClO, and 1O2. It also provides indirect antioxidant protection through metal chelation (e.g., Cu2+ and Fe3+) and the regeneration of certain endogenous antioxidants (e.g., vitamin E, vitamin C, and glutathione). Additionally, ALA leverages its amphiphilic properties to distribute in both hydrophilic environments (e.g., plasma) and lipophilic environments (e.g., cell membranes). Finally, ALA’s antioxidative action effectively protects mitochondria, ensuring efficient ATP production and thereby maintaining the proper function of the sodium–potassium pump [24]. (Arrows indicate promotion, lines indicate inhibition). Created with BioRender.com (accessed on 22 July 2024). RNS: reactive nitrogen species.Figure 3. Carcinogenic factors in environmental pollution and chronic poisoning and changes in redox homeostasis during increased ROS levels. (A) Carcinogenic factors in environmental pollution and chronic poisoning: Under long-term exposure and chronic intake, metalloids (e.g., As), toxic metals (e.g., Hg), synthetic toxic substances (e.g., pesticides), and organic toxins (e.g., formaldehyde) can exhibit strong carcinogenic potential. Additionally, the sources of these toxic substances may include water, food, or air, as well as factors related to occupational exposure. This is also associated with long-term groundwater and crop contamination. (B) Changes in redox homeostasis during increased ROS levels: Under normal physiological conditions, the oxidative and reductive pressures in the body should be in a dynamic balance, with oxidants and antioxidants relatively balanced. In the case of pathological ROS increase, the reductive capacity will be progressively limited by the oxidants. When the oxidative stress exceeds the body’s maximum reductive capacity, it will induce related pathological outcomes. Created with BioRender.com (accessed on 22 July 2024). As: arsenic; Hg: mercury.
2.2. Anti-Infective and Anti-Carcinogenic Effects of Chronic Inflammation
- Chronic inflammation induced by infection can activate signaling pathways such as the NF-κB, JAK/STAT, and Rb pathways, promoting the synthesis of inflammatory factors and cellular stress states, leading to somatic mutations and/or oncogene activation;
- Pathogen-secreted byproducts and/or metabolites may alter local environmental conditions (e.g., redox homeostasis, pH levels) to favor their replication, often resulting in chromosomal DNA damage in the affected tissues or organs;
- Some pathogens possess the capacity to modify the immune microenvironment, resulting in the induction of disordered immune responses. This phenomenon has been observed in the co-induction of malignant B-cell clones by malaria parasites and Epstein-Barr virus (EBV), which produce immunosuppressive cytokines that induce T-cell apoptosis and/or trigger the recruitment of myeloid-derived suppressor cells and Tregs, as observed in the cytokine storm induced by COVID-19;
- Persistent infection symptoms can maintain local tissues in a state of constant stimulation and healing after damage, which may potentially lead to genetic mutations, epigenetic changes, and protein modifications. These changes may result in the activation of oncogenes and the inhibition of anticancer processes. Furthermore, prolonged inflammatory responses may also result in immune system exhaustion, thereby facilitating the escape of tumour cells from immune surveillance [29,30,31,32].
- ALA helps maintain redox homeostasis, preventing the redox state from being skewed in favor of pathogen proliferation [36];
- ALA can regenerate depleted glutathione (GSH) during viral replication and internalization. The endogenous antioxidant glutathione is associated with leukocyte proliferation, enhanced immune system function, and effective antiviral protection. A reduction in glutathione levels can impair the Na+/H+ antiporter, resulting in a reduction in intracellular pH and creating an environment conducive to viral replication [36,37];
- ALA can increase TCA cycle activity, enhancing ATP production and providing energy for immune activities and healing. Additionally, inflammation suppression can indirectly enhance immune system function, preventing hypersensitive reactions such as cytokine storms [38].
2.3. Effects of Detoxification on Environmental Pollution and Chronic Toxicity
- ALA can scavenge the excessive ROS generated by poisoning, maintain redox balance, and reduce damage to crucial structures such as DNA and mitochondria;
- For metal toxins, ALA has chelating properties, reducing the cellular uptake of toxic metals and potentially promoting their excretion by forming complexes [58];
- ALA facilitates the regeneration of reduced glutathione through the Nrf2/ARE signalling pathway, thereby reducing the interference with normal antioxidant functions caused by toxins;
- ALA has been demonstrated to be particularly effective in the detoxification of mercury. In addition to chelating Hg2+, ALA can mitigate mercury toxicity by interacting with selenide compounds. This interaction results in the formation of stable complexes with thiol groups in selenium enzymes such as thioredoxin reductase, leading to mercury separation [59,60];
- ALA can also work synergistically with dimercaptosuccinic acid (DMSA), a known heavy metal detoxifier, to lower arsenic levels in rat models [61].
2.4. Anti-Carcinogenic Effects against Other Carcinogenic Factors
3. ALA and Its Anticancer Effects
3.1. Anticancer Cell Proliferation
3.2. Pro-Apoptotic Effects on Cancer Cells
3.3. Anti-Migration, Invasion, and EMT Effects
3.4. Unproven Hypotheses and Other Effects
4. The Adjuvant Role of ALA in Cancer Treatment
4.1. Enhancing Chemotherapy and Radiotherapy Effects
4.2. Reducing Adverse Effects after Anticancer Treatment
4.3. Auxiliary Role in Cancer Nanomedicine
- ALA is a member of the liposome family and possesses natural disulfide bonds that enable it to remain relatively stable in systemic circulation and rapidly degrade upon absorption by cancer cells, exerting its effects. Furthermore, ALA exhibits strong amphiphilicity, excellent biocompatibility, and easy chemical modification, rendering it one of the most promising candidates for anticancer drug delivery [153,154,155,156,157];
- In normal cells, ALA acts as an antioxidant by clearing ROS. However, in cancer cells, it can exert pro-oxidative effects, inducing pathways that restrict cancer progression. This indicates that nanostructures containing ALA, in conjunction with anticancer drugs, exhibit synergistic effects, increasing selectivity and providing protective and reparative functions against the side effects of several anticancer drugs that mediate ROS generation;
- ALA is a naturally occurring compound with nontoxic properties that effectively avoids the toxicity associated with drug accumulation caused by the enhanced permeability and retention (EPR) effect when it is administered systemically to treat solid tumors [158].
4.4. Other Auxiliary Effects
5. Future Prospects and Questions
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Zhao, D.; Yang, K.; Guo, H.; Zeng, J.; Wang, S.; Xu, H.; Ge, A.; Zeng, L.; Chen, S.; Ge, J. Mechanisms of ferroptosis in Alzheimer’s disease and therapeutic effects of natural plant products: A review. Biomed. Pharmacother. 2023, 164, 114312. [Google Scholar] [CrossRef] [PubMed]
- Rochette, L.; Ghibu, S. Mechanics Insights of Alpha-Lipoic Acid against Cardiovascular Diseases during COVID-19 Infection. Int. J. Mol. Sci. 2021, 22, 7979. [Google Scholar] [CrossRef] [PubMed]
- Gomes, M.B.; Negrato, C.A. Alpha-lipoic acid as a pleiotropic compound with potential therapeutic use in diabetes and other chronic diseases. Diabetol. Metab. Syndr. 2014, 6, 80. [Google Scholar] [CrossRef] [PubMed]
- Rochette, L.; Ghibu, S.; Muresan, A.; Vergely, C. Alpha-lipoic acid: Molecular mechanisms and therapeutic potential in diabetes. Can. J. Physiol. Pharmacol. 2015, 93, 1021–1027. [Google Scholar] [CrossRef] [PubMed]
- Shay, K.P.; Moreau, R.F.; Smith, E.J.; Smith, A.R.; Hagen, T.M. Alpha-lipoic acid as a dietary supplement: Molecular mechanisms and therapeutic potential. Biochim. Biophys. Acta 2009, 1790, 1149–1160. [Google Scholar] [CrossRef] [PubMed]
- Wedan, R.J.; Longenecker, J.Z.; Nowinski, S.M. Mitochondrial fatty acid synthesis is an emergent central regulator of mammalian oxidative metabolism. Cell Metab. 2024, 36, 36–47. [Google Scholar] [CrossRef]
- Sies, H. Oxidative Stress: Concept and Some Practical Aspects. Antioxidants 2020, 9, 852. [Google Scholar] [CrossRef] [PubMed]
- Biewenga, G.P.; Haenen, G.R.; Bast, A. The pharmacology of the antioxidant lipoic acid. Gen. Pharmacol. 1997, 29, 315–331. [Google Scholar] [CrossRef] [PubMed]
- Viana, M.D.M.; Lauria, P.S.S.; Lima, A.A.d.; Opretzka, L.C.F.; Marcelino, H.R.; Villarreal, C.F. Alpha-Lipoic Acid as an Antioxidant Strategy for Managing Neuropathic Pain. Antioxidants 2022, 11, 2420. [Google Scholar] [CrossRef]
- Farhat, D.; Ghayad, S.E.; Icard, P.; Le Romancer, M.; Hussein, N.; Lincet, H. Lipoic acid-induced oxidative stress abrogates IGF-1R maturation by inhibiting the CREB/furin axis in breast cancer cell lines. Oncogene 2020, 39, 3604–3610. [Google Scholar] [CrossRef]
- Darenskaya, M.; Kolesnikov, S.; Semenova, N.; Kolesnikova, L. Diabetic Nephropathy: Significance of Determining Oxidative Stress and Opportunities for Antioxidant Therapies. Int. J. Mol. Sci. 2023, 24, 12378. [Google Scholar] [CrossRef] [PubMed]
- Di Meo, S.; Reed, T.T.; Venditti, P.; Victor, V.M. Role of ROS and RNS Sources in Physiological and Pathological Conditions. Oxid. Med. Cell Longev. 2016, 2016, 1245049. [Google Scholar] [CrossRef]
- Dragomanova, S.; Miteva, S.; Nicoletti, F.; Mangano, K.; Fagone, P.; Pricoco, S.; Staykov, H.; Tancheva, L. Therapeutic Potential of Alpha-Lipoic Acid in Viral Infections, including COVID-19. Antioxidants 2021, 10, 1294. [Google Scholar] [CrossRef] [PubMed]
- Yin, H.; Xu, L.; Porter, N.A. Free radical lipid peroxidation: Mechanisms and analysis. Chem. Rev. 2011, 111, 5944–5972. [Google Scholar] [CrossRef]
- Román-Pintos, L.M.; Villegas-Rivera, G.; Rodríguez-Carrizalez, A.D.; Miranda-Díaz, A.G.; Cardona-Muñoz, E.G. Diabetic Polyneuropathy in Type 2 Diabetes Mellitus: Inflammation, Oxidative Stress, and Mitochondrial Function. J. Diabetes Res. 2016, 2016, 3425617. [Google Scholar] [CrossRef] [PubMed]
- Xiao, W.H.; Bennett, G.J. Effects of mitochondrial poisons on the neuropathic pain produced by the chemotherapeutic agents, paclitaxel and oxaliplatin. Pain 2012, 153, 704–709. [Google Scholar] [CrossRef] [PubMed]
- Green, D.R.; Galluzzi, L.; Kroemer, G. Mitochondria and the autophagy-inflammation-cell death axis in organismal aging. Science 2011, 333, 1109–1112. [Google Scholar] [CrossRef] [PubMed]
- Ludman, T.; Melemedjian, O.K. Bortezomib-induced aerobic glycolysis contributes to chemotherapy-induced painful peripheral neuropathy. Mol. Pain 2019, 15, 1744806919837429. [Google Scholar] [CrossRef]
- Bjørklund, G.; Aaseth, J.; Crisponi, G.; Rahman, M.M.; Chirumbolo, S. Insights on alpha lipoic and dihydrolipoic acids as promising scavengers of oxidative stress and possible chelators in mercury toxicology. J. Inorg. Biochem. 2019, 195, 111–119. [Google Scholar] [CrossRef]
- Checa, J.; Aran, J.M. Reactive Oxygen Species: Drivers of Physiological and Pathological Processes. J. Inflamm. Res. 2020, 13, 1057–1073. [Google Scholar] [CrossRef]
- Holley, A.K.; Bakthavatchalu, V.; Velez-Roman, J.M.; St Clair, D.K. Manganese superoxide dismutase: Guardian of the powerhouse. Int. J. Mol. Sci. 2011, 12, 7114–7162. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Huang, T.T.; Carlson, E.J.; Melov, S.; Ursell, P.C.; Olson, J.L.; Noble, L.J.; Yoshimura, M.P.; Berger, C.; Chan, P.H.; et al. Dilated cardiomyopathy and neonatal lethality in mutant mice lacking manganese superoxide dismutase. Nat. Genet. 1995, 11, 376–381. [Google Scholar] [CrossRef] [PubMed]
- Rochette, L.; Ghibu, S.; Richard, C.; Zeller, M.; Cottin, Y.; Vergely, C. Direct and indirect antioxidant properties of α-lipoic acid and therapeutic potential. Mol. Nutr. Food Res. 2013, 57, 114–125. [Google Scholar] [CrossRef] [PubMed]
- Abdal Dayem, A.; Hossain, M.K.; Lee, S.B.; Kim, K.; Saha, S.K.; Yang, G.-M.; Choi, H.Y.; Cho, S.-G. The Role of Reactive Oxygen Species (ROS) in the Biological Activities of Metallic Nanoparticles. Int. J. Mol. Sci. 2017, 18, 120. [Google Scholar] [CrossRef] [PubMed]
- Oikonomopoulou, K.; Brinc, D.; Kyriacou, K.; Diamandis, E.P. Infection and cancer: Revaluation of the hygiene hypothesis. Clin. Cancer Res. 2013, 19, 2834–2841. [Google Scholar] [CrossRef] [PubMed]
- de Martel, C.; Georges, D.; Bray, F.; Ferlay, J.; Clifford, G.M. Global burden of cancer attributable to infections in 2018: A worldwide incidence analysis. Lancet Glob. Health 2020, 8, e180–e190. [Google Scholar] [CrossRef] [PubMed]
- Boshoff, C.; Weiss, R. AIDS-related malignancies. Nat. Rev. Cancer 2002, 2, 373–382. [Google Scholar] [CrossRef] [PubMed]
- Moon, E.-J.; Jeong, C.-H.; Jeong, J.-W.; Kim, K.R.; Yu, D.-Y.; Murakami, S.; Kim, C.W.; Kim, K.-W. Hepatitis B virus X protein induces angiogenesis by stabilizing hypoxia-inducible factor-1alpha. FASEB J. 2004, 18, 382–384. [Google Scholar] [CrossRef] [PubMed]
- van Tong, H.; Brindley, P.J.; Meyer, C.G.; Velavan, T.P. Parasite Infection, Carcinogenesis and Human Malignancy. EBioMedicine 2017, 15, 12–23. [Google Scholar] [CrossRef]
- Tasaka, S.; Amaya, F.; Hashimoto, S.; Ishizaka, A. Roles of oxidants and redox signaling in the pathogenesis of acute respiratory distress syndrome. Antioxid. Redox Signal 2008, 10, 739–753. [Google Scholar] [CrossRef]
- Pagano, G.; Manfredi, C.; Pallardó, F.V.; Lyakhovich, A.; Tiano, L.; Trifuoggi, M. Potential roles of mitochondrial cofactors in the adjuvant mitigation of proinflammatory acute infections, as in the case of sepsis and COVID-19 pneumonia. Inflamm. Res. 2021, 70, 159–170. [Google Scholar] [CrossRef] [PubMed]
- Chêne, A.; Donati, D.; Guerreiro-Cacais, A.O.; Levitsky, V.; Chen, Q.; Falk, K.I.; Orem, J.; Kironde, F.; Wahlgren, M.; Bejarano, M.T. A molecular link between malaria and Epstein-Barr virus reactivation. PLoS Pathog. 2007, 3, e80. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Mu, J.; Du, J.; Feng, Y.; Xu, W.; Bai, M.; Zhang, H. Alpha-lipoic acid could attenuate the effect of chemerin-induced diabetic nephropathy progression. Iran. J. Basic. Med. Sci. 2021, 24, 1107–1116. [Google Scholar] [CrossRef] [PubMed]
- Biewenga, G.P.; Dorstijn, M.A.; Verhagen, J.V.; Haenen, G.R.; Bast, A. Reduction of lipoic acid by lipoamide dehydrogenase. Biochem. Pharmacol. 1996, 51, 233–238. [Google Scholar] [CrossRef]
- Kaur, D.; Behl, T.; Sehgal, A.; Singh, S.; Sharma, N.; Chigurupati, S.; Alhowail, A.; Abdeen, A.; Ibrahim, S.F.; Vargas-De-La-Cruz, C.; et al. Decrypting the potential role of α-lipoic acid in Alzheimer’s disease. Life Sci. 2021, 284, 119899. [Google Scholar] [CrossRef] [PubMed]
- Rudzite, V.; Berzinsh, J.; Grivane, I.; Fuchs, D.; Baier-Bitterlich, G.; Wachter, H. Serum tryptophan, kynurenine, and neopterin in patients with Guillain-Barre-syndrome (GBS) and multiple sclerosis (MS). Adv. Exp. Med. Biol. 1996, 398, 183–187. [Google Scholar] [PubMed]
- Rózsa, E.; Robotka, H.; Vécsei, L.; Toldi, J. The Janus-face kynurenic acid. J. Neural Transm. 2008, 115, 1087–1091. [Google Scholar] [CrossRef] [PubMed]
- Nowinski, S.M.; Van Vranken, J.G.; Dove, K.K.; Rutter, J. Impact of Mitochondrial Fatty Acid Synthesis on Mitochondrial Biogenesis. Curr. Biol. 2018, 28, R1212–R1219. [Google Scholar] [CrossRef]
- Cheung, E.C.; Vousden, K.H. The role of ROS in tumour development and progression. Nat. Rev. Cancer 2022, 22, 280–297. [Google Scholar] [CrossRef]
- Lood, C.; Blanco, L.P.; Purmalek, M.M.; Carmona-Rivera, C.; De Ravin, S.S.; Smith, C.K.; Malech, H.L.; Ledbetter, J.A.; Elkon, K.B.; Kaplan, M.J. Neutrophil extracellular traps enriched in oxidized mitochondrial DNA are interferogenic and contribute to lupus-like disease. Nat. Med. 2016, 22, 146–153. [Google Scholar] [CrossRef]
- Papayannopoulos, V. Neutrophil extracellular traps in immunity and disease. Nat. Rev. Immunol. 2018, 18, 134–147. [Google Scholar] [CrossRef] [PubMed]
- Park, S.; Karunakaran, U.; Jeoung, N.H.; Jeon, J.-H.; Lee, I.-K. Physiological effect and therapeutic application of alpha lipoic acid. Curr. Med. Chem. 2014, 21, 3636–3645. [Google Scholar] [CrossRef] [PubMed]
- Scaramuzza, A.; Giani, E.; Redaelli, F.; Ungheri, S.; Macedoni, M.; Giudici, V.; Bosetti, A.; Ferrari, M.; Zuccotti, G.V. Alpha-Lipoic Acid and Antioxidant Diet Help to Improve Endothelial Dysfunction in Adolescents with Type 1 Diabetes: A Pilot Trial. J. Diabetes Res. 2015, 2015, 474561. [Google Scholar] [CrossRef] [PubMed]
- Pande, M.; Flora, S.J.S. Lead induced oxidative damage and its response to combined administration of alpha-lipoic acid and succimers in rats. Toxicology 2002, 177, 187–196. [Google Scholar] [CrossRef] [PubMed]
- Patrick, L. Mercury toxicity and antioxidants: Part 1: Role of glutathione and alpha-lipoic acid in the treatment of mercury toxicity. Altern. Med. Rev. 2002, 7, 456–471. [Google Scholar]
- Shindyapina, A.V.; Komarova, T.V.; Sheshukova, E.V.; Ershova, N.M.; Tashlitsky, V.N.; Kurkin, A.V.; Yusupov, I.R.; Mkrtchyan, G.V.; Shagidulin, M.Y.; Dorokhov, Y.L. The Antioxidant Cofactor Alpha-Lipoic Acid May Control Endogenous Formaldehyde Metabolism in Mammals. Front. Neurosci. 2017, 11, 651. [Google Scholar] [CrossRef] [PubMed]
- De Gregori, I.; Fuentes, E.; Rojas, M.; Pinochet, H.; Potin-Gautier, M. Monitoring of copper, arsenic and antimony levels in agricultural soils impacted and non-impacted by mining activities, from three regions in Chile. J. Environ. Monit. 2003, 5, 287–295. [Google Scholar] [CrossRef] [PubMed]
- Román, M.D.; Niclis, C.; Aballay, L.R.; Lantieri, M.J.; Díaz, M.D.e.P.; Muñoz, S.E. Do Exposure to Arsenic, Occupation and Diet Have Synergistic Effects on Prostate Cancer Risk? Asian Pac. J. Cancer Prev. 2018, 19, 1495–1501. [Google Scholar] [PubMed]
- Pershagen, G. Lung cancer mortality among men living near an arsenic-emitting smelter. Am. J. Epidemiol. 1985, 122, 684–694. [Google Scholar] [CrossRef]
- Shi, C.; Zhou, X.; Zhang, J.; Wang, J.; Xie, H.; Wu, Z. α-Lipoic acid protects against the cytotoxicity and oxidative stress induced by cadmium in HepG2 cells through regeneration of glutathione by glutathione reductase via Nrf2/ARE signaling pathway. Environ. Toxicol. Pharmacol. 2016, 45, 274–281. [Google Scholar] [CrossRef]
- Macias-Barragan, J.; Huerta-Olvera, S.G.; Hernandez-Cañaveral, I.; Pereira-Suarez, A.L.; Montoya-Buelna, M. Cadmium and α-lipoic acid activate similar de novo synthesis and recycling pathways for glutathione balance. Environ. Toxicol. Pharmacol. 2017, 52, 38–46. [Google Scholar] [CrossRef]
- Ebert, F.; Weiss, A.; Bültemeyer, M.; Hamann, I.; Hartwig, A.; Schwerdtle, T. Arsenicals affect base excision repair by several mechanisms. Mutat. Res. 2011, 715, 32–41. [Google Scholar] [CrossRef]
- Holcomb, N.; Goswami, M.; Han, S.G.; Scott, T.; D’Orazio, J.; Orren, D.K.; Gairola, C.G.; Mellon, I. Inorganic arsenic inhibits the nucleotide excision repair pathway and reduces the expression of XPC. DNA Repair 2017, 52, 70–80. [Google Scholar] [CrossRef] [PubMed]
- Hartwig, A. Metal interaction with redox regulation: An integrating concept in metal carcinogenesis? Free Radic. Biol. Med. 2013, 55, 63–72. [Google Scholar] [CrossRef] [PubMed]
- Nollen, M.; Ebert, F.; Moser, J.; Mullenders, L.H.F.; Hartwig, A.; Schwerdtle, T. Impact of arsenic on nucleotide excision repair: XPC function, protein level, and gene expression. Mol. Nutr. Food Res. 2009, 53, 572–582. [Google Scholar] [CrossRef]
- Zhou, Q.; Xi, S. A review on arsenic carcinogenesis: Epidemiology, metabolism, genotoxicity and epigenetic changes. Regul. Toxicol. Pharmacol. 2018, 99, 78–88. [Google Scholar] [CrossRef]
- Bjørklund, G.; Aaseth, J.; Chirumbolo, S.; Urbina, M.A.; Uddin, R. Effects of arsenic toxicity beyond epigenetic modifications. Environ. Geochem. Health 2018, 40, 955–965. [Google Scholar] [CrossRef] [PubMed]
- Muhammad, M.T.; Khan, M.N. Kinetics, mechanistic and synergistic studies of Alpha lipoic acid with hydrogen peroxide. J. Saudi Chem. Soc. 2017, 21, 123–131. [Google Scholar] [CrossRef]
- Jadán-Piedra, C.; Vélez, D.; Devesa, V. In vitro evaluation of dietary compounds to reduce mercury bioavailability. Food Chem. 2018, 248, 353–359. [Google Scholar] [CrossRef]
- Carvalho, C.M.L.; Lu, J.; Zhang, X.; Arnér, E.S.J.; Holmgren, A. Effects of selenite and chelating agents on mammalian thioredoxin reductase inhibited by mercury: Implications for treatment of mercury poisoning. FASEB J. 2011, 25, 370–381. [Google Scholar] [CrossRef]
- Kokilavani, V.; Devi, M.A.; Sivarajan, K.; Panneerselvam, C. Combined efficacies of DL-alpha-lipoic acid and meso 2,3 dimercaptosuccinic acid against arsenic induced toxicity in antioxidant systems of rats. Toxicol. Lett. 2005, 160, 1–7. [Google Scholar] [CrossRef] [PubMed]
- Xu, G.; Zhao, Y.; Tao, Y.; Xiong, C.; Lv, M.; Gao, Q.; Zhang, F.; An, Z.; Wu, W. Lias overexpression alleviates pulmonary injury induced by fine particulate matter in mice. Environ. Geochem. Health 2023, 45, 6585–6603. [Google Scholar] [CrossRef] [PubMed]
- Araujo, J.A.; Nel, A.E. Particulate matter and atherosclerosis: Role of particle size, composition and oxidative stress. Part. Fibre Toxicol. 2009, 6, 24. [Google Scholar] [CrossRef] [PubMed]
- Chakraborti, D.; Rahman, M.M.; Mukherjee, A.; Alauddin, M.; Hassan, M.; Dutta, R.N.; Pati, S.; Mukherjee, S.C.; Roy, S.; Quamruzzman, Q.; et al. Groundwater arsenic contamination in Bangladesh—21 Years of research. J. Trace Elem. Med. Biol. 2015, 31, 237–248. [Google Scholar] [CrossRef] [PubMed]
- Mondal, P.; Majumder, C.B.; Mohanty, B. Laboratory based approaches for arsenic remediation from contaminated water: Recent developments. J. Hazard. Mater. 2006, 137, 464–479. [Google Scholar] [CrossRef] [PubMed]
- Aposhian, H.V.; Morgan, D.L.; Queen, H.L.S.; Maiorino, R.M.; Aposhian, M.M. Vitamin C, glutathione, or lipoic acid did not decrease brain or kidney mercury in rats exposed to mercury vapor. J. Toxicol. Clin. Toxicol. 2003, 41, 339–347. [Google Scholar] [CrossRef] [PubMed]
- Trewin, A.J.; Lundell, L.S.; Perry, B.D.; Patil, K.V.; Chibalin, A.V.; Levinger, I.; McQuade, L.R.; Stepto, N.K. Effect of N-acetylcysteine infusion on exercise-induced modulation of insulin sensitivity and signaling pathways in human skeletal muscle. Am. J. Physiol. Endocrinol. Metab. 2015, 309, E388–E397. [Google Scholar] [CrossRef] [PubMed]
- Merry, T.L.; McConell, G.K. Do reactive oxygen species regulate skeletal muscle glucose uptake during contraction? Exerc. Sport. Sci. Rev. 2012, 40, 102–105. [Google Scholar] [CrossRef]
- Mody, R.J.; Wu, Y.-M.; Lonigro, R.J.; Cao, X.; Roychowdhury, S.; Vats, P.; Frank, K.M.; Prensner, J.R.; Asangani, I.; Palanisamy, N.; et al. Integrative Clinical Sequencing in the Management of Refractory or Relapsed Cancer in Youth. JAMA 2015, 314, 913–925. [Google Scholar] [CrossRef]
- Fasipe, B.; Faria, A.; Laher, I. Potential for Novel Therapeutic Uses of Alpha Lipoic Acid. Curr. Med. Chem. 2023, 30, 3942–3954. [Google Scholar] [CrossRef]
- Farhat, D.; Lincet, H. Lipoic acid a multi-level molecular inhibitor of tumorigenesis. Biochim. Biophys. Acta Rev. Cancer 2020, 1873, 188317. [Google Scholar] [CrossRef] [PubMed]
- Jeon, M.J.; Kim, W.G.; Lim, S.; Choi, H.-J.; Sim, S.; Kim, T.Y.; Shong, Y.K.; Kim, W.B. Alpha lipoic acid inhibits proliferation and epithelial mesenchymal transition of thyroid cancer cells. Mol. Cell. Endocrinol. 2016, 419, 113–123. [Google Scholar] [CrossRef] [PubMed]
- Tripathy, J.; Tripathy, A.; Thangaraju, M.; Suar, M.; Elangovan, S. α-Lipoic acid inhibits the migration and invasion of breast cancer cells through inhibition of TGFβ signaling. Life Sci. 2018, 207, 15–22. [Google Scholar] [CrossRef]
- Kuban-Jankowska, A.; Gorska-Ponikowska, M.; Wozniak, M. Lipoic Acid Decreases the Viability of Breast Cancer Cells and Activity of PTP1B and SHP2. Anticancer. Res. 2017, 37, 2893–2898. [Google Scholar] [PubMed]
- Balavenkatraman, K.K.; Aceto, N.; Britschgi, A.; Mueller, U.; Bence, K.K.; Neel, B.G.; Bentires-Alj, M. Epithelial protein-tyrosine phosphatase 1B contributes to the induction of mammary tumors by HER2/Neu but is not essential for tumor maintenance. Mol. Cancer Res. 2011, 9, 1377–1384. [Google Scholar] [CrossRef] [PubMed]
- Farhat, D.; Léon, S.; Ghayad, S.E.; Gadot, N.; Icard, P.; Le Romancer, M.; Hussein, N.; Lincet, H. Lipoic acid decreases breast cancer cell proliferation by inhibiting IGF-1R via furin downregulation. Br. J. Cancer 2020, 122, 885–894. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.; Nie, Q.; Gao, M.; Yang, L.; Xiang, J.-W.; Xiao, Y.; Liu, F.-Y.; Gong, X.-D.; Fu, J.-L.; Wang, Y.; et al. The transcription factor CREB acts as an important regulator mediating oxidative stress-induced apoptosis by suppressing αB-crystallin expression. Aging 2020, 12, 13594–13617. [Google Scholar] [CrossRef]
- Glaviano, A.; Foo, A.S.C.; Lam, H.Y.; Yap, K.C.H.; Jacot, W.; Jones, R.H.; Eng, H.; Nair, M.G.; Makvandi, P.; Geoerger, B.; et al. PI3K/AKT/mTOR signaling transduction pathway and targeted therapies in cancer. Mol. Cancer 2023, 22, 138. [Google Scholar] [CrossRef] [PubMed]
- Yu, J.S.L.; Cui, W. Proliferation, survival and metabolism: The role of PI3K/AKT/mTOR signalling in pluripotency and cell fate determination. Development 2016, 143, 3050–3060. [Google Scholar] [CrossRef]
- Park, S.; Choi, S.K.; Choi, Y.; Moon, H.-S. AMPK/p53 Axis Is Essential for α-Lipoic Acid-Regulated Metastasis in Human and Mouse Colon Cancer Cells. J. Investig. Med. 2015, 63, 882–885. [Google Scholar] [CrossRef]
- Li, W.; Saud, S.M.; Young, M.R.; Chen, G.; Hua, B. Targeting AMPK for cancer prevention and treatment. Oncotarget 2015, 6, 7365–7378. [Google Scholar] [CrossRef] [PubMed]
- Park, K.-G.; Min, A.-K.; Koh, E.H.; Kim, H.S.; Kim, M.-O.; Park, H.-S.; Kim, Y.-D.; Yoon, T.-S.; Jang, B.K.; Hwang, J.S.; et al. Alpha-lipoic acid decreases hepatic lipogenesis through adenosine monophosphate-activated protein kinase (AMPK)-dependent and AMPK-independent pathways. Hepatology 2008, 48, 1477–1486. [Google Scholar] [CrossRef]
- Lee, W.J.; Song, K.-H.; Koh, E.H.; Won, J.C.; Kim, H.S.; Park, H.-S.; Kim, M.-S.; Kim, S.-W.; Lee, K.-U.; Park, J.-Y. α-Lipoic acid increases insulin sensitivity by activating AMPK in skeletal muscle. Biochem. Biophys. Res. Commun. 2005, 332, 885–891. [Google Scholar] [CrossRef] [PubMed]
- Yue, L.; Ren, Y.; Yue, Q.; Ding, Z.; Wang, K.; Zheng, T.; Chen, G.; Chen, X.; Li, M.; Fan, L. α-Lipoic Acid Targeting PDK1/NRF2 Axis Contributes to the Apoptosis Effect of Lung Cancer Cells. Oxid. Med. Cell Longev. 2021, 2021, 6633419. [Google Scholar] [CrossRef]
- Phiboonchaiyanan, P.P.; Chanvorachote, P. Suppression of a cancer stem-like phenotype mediated by alpha-lipoic acid in human lung cancer cells through down-regulation of β-catenin and Oct-4. Cell Oncol. 2017, 40, 497–510. [Google Scholar] [CrossRef]
- Fan, W.; Li, X. The SIRT1-c-Myc axis in regulation of stem cells. Front. Cell Dev. Biol. 2023, 11, 1236968. [Google Scholar] [CrossRef]
- Elbadawy, M.; Usui, T.; Yamawaki, H.; Sasaki, K. Emerging Roles of C-Myc in Cancer Stem Cell-Related Signaling and Resistance to Cancer Chemotherapy: A Potential Therapeutic Target Against Colorectal Cancer. Int. J. Mol. Sci. 2019, 20, 2340. [Google Scholar] [CrossRef]
- Akari, M.; Toshiyuki, M.; Atsuko, N.; Yasuko, K.; Mayuko, I.; Satoru, M. Cell Cycle Regulation via the p53, PTEN, and BRCA1 Tumor Suppressors. In New Aspects in Molecular and Cellular Mechanisms of Human Carcinogenesis; Dmitry, B., Ed.; IntechOpen: Rijeka, Croatia, 2016; Chapter 2. [Google Scholar]
- Weinberg, F.; Hamanaka, R.; Wheaton, W.W.; Weinberg, S.; Joseph, J.; Lopez, M.; Kalyanaraman, B.; Mutlu, G.M.; Budinger, G.R.S.; Chandel, N.S. Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proc. Natl. Acad. Sci. USA 2010, 107, 8788–8793. [Google Scholar] [CrossRef]
- Radisky, D.C.; Levy, D.D.; Littlepage, L.E.; Liu, H.; Nelson, C.M.; Fata, J.E.; Leake, D.; Godden, E.L.; Albertson, D.G.; Nieto, M.A.; et al. Rac1b and reactive oxygen species mediate MMP-3-induced EMT and genomic instability. Nature 2005, 436, 123–127. [Google Scholar] [CrossRef] [PubMed]
- Nieborowska-Skorska, M.; Kopinski, P.K.; Ray, R.; Hoser, G.; Ngaba, D.; Flis, S.; Cramer, K.; Reddy, M.M.; Koptyra, M.; Penserga, T.; et al. Rac2-MRC-cIII-generated ROS cause genomic instability in chronic myeloid leukemia stem cells and primitive progenitors. Blood 2012, 119, 4253–4263. [Google Scholar] [CrossRef]
- Sosa, V.; Moliné, T.; Somoza, R.; Paciucci, R.; Kondoh, H.; Lleonart, M.E. Oxidative stress and cancer: An overview. Ageing Res. Rev. 2013, 12, 376–390. [Google Scholar] [CrossRef] [PubMed]
- Park, Y.-J.; Yoo, S.-A.; Kim, W.-U. Role of endoplasmic reticulum stress in rheumatoid arthritis pathogenesis. J. Korean Med. Sci. 2014, 29, 2–11. [Google Scholar] [CrossRef] [PubMed]
- Pibiri, M.; Sulas, P.; Camboni, T.; Leoni, V.P.; Simbula, G. α-Lipoic acid induces Endoplasmic Reticulum stress-mediated apoptosis in hepatoma cells. Sci. Rep. 2020, 10, 7139. [Google Scholar] [CrossRef] [PubMed]
- Dörsam, B.; Fahrer, J. The disulfide compound α-lipoic acid and its derivatives: A novel class of anticancer agents targeting mitochondria. Cancer Lett. 2016, 371, 12–19. [Google Scholar] [CrossRef] [PubMed]
- Göder, A.; Nagel, G.; Kraus, A.; Dörsam, B.; Seiwert, N.; Kaina, B.; Fahrer, J. Lipoic acid inhibits the DNA repair protein O 6-methylguanine-DNA methyltransferase (MGMT) and triggers its depletion in colorectal cancer cells with concomitant autophagy induction. Carcinogenesis 2015, 36, 817–831. [Google Scholar] [CrossRef] [PubMed]
- Kothari, I.R.; Mazumdar, S.; Sharma, S.; Italiya, K.; Mittal, A.; Chitkara, D. Docetaxel and alpha-lipoic acid co-loaded nanoparticles for cancer therapy. Ther. Deliv. 2019, 10, 227–240. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Su, Z.; Tavana, O.; Gu, W. Understanding the complexity of p53 in a new era of tumor suppression. Cancer Cell 2024, 42, 946–967. [Google Scholar] [CrossRef] [PubMed]
- Yu, W.; Zhang, L.; Wei, Q.; Shao, A. O6-Methylguanine-DNA Methyltransferase (MGMT): Challenges and New Opportunities in Glioma Chemotherapy. Front. Oncol. 2019, 9, 1547. [Google Scholar] [CrossRef] [PubMed]
- Kaina, B.; Christmann, M.; Naumann, S.; Roos, W.P. MGMT: Key node in the battle against genotoxicity, carcinogenicity and apoptosis induced by alkylating agents. DNA Repair 2007, 6, 1079–1099. [Google Scholar] [CrossRef]
- Aoyama, S.; Okimura, Y.; Fujita, H.; Sato, E.F.; Umegaki, T.; Abe, K.; Inoue, M.; Utsumi, K.; Sasaki, J. Stimulation of membrane permeability transition by alpha-lipoic acid and its biochemical characteristics. Physiol. Chem. Phys. Med. NMR 2006, 38, 1–20. [Google Scholar]
- Isenberg, J.S.; Klaunig, J.E. Role of the mitochondrial membrane permeability transition (MPT) in rotenone-induced apoptosis in liver cells. Toxicol. Sci. 2000, 53, 340–351. [Google Scholar] [CrossRef] [PubMed]
- Frigo, E.; Tommasin, L.; Lippe, G.; Carraro, M.; Bernardi, P. The Haves and Have-Nots: The Mitochondrial Permeability Transition Pore across Species. Cells 2023, 12, 1409. [Google Scholar] [CrossRef] [PubMed]
- Bonora, M.; Pinton, P. The mitochondrial permeability transition pore and cancer: Molecular mechanisms involved in cell death. Front. Oncol. 2014, 4, 302. [Google Scholar] [CrossRef] [PubMed]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
- Fidler, I.J. The pathogenesis of cancer metastasis: The ‘seed and soil’ hypothesis revisited. Nat. Rev. Cancer 2003, 3, 453–458. [Google Scholar] [CrossRef]
- Chaffer, C.L.; Weinberg, R.A. A perspective on cancer cell metastasis. Science 2011, 331, 1559–1564. [Google Scholar] [CrossRef]
- Zachary, I. Focal adhesion kinase. Int. J. Biochem. Cell Biol. 1997, 29, 929–934. [Google Scholar] [CrossRef]
- Huang, Y.; Hong, W.; Wei, X. The molecular mechanisms and therapeutic strategies of EMT in tumor progression and metastasis. J. Hematol. Oncol. 2022, 15, 129. [Google Scholar] [CrossRef]
- Hao, Y.; Baker, D.; Ten Dijke, P. TGF-β-Mediated Epithelial-Mesenchymal Transition and Cancer Metastasis. Int. J. Mol. Sci. 2019, 20, 2767. [Google Scholar] [CrossRef]
- Baribeau, S.; Chaudhry, P.; Parent, S.; Asselin, É. Resveratrol inhibits cisplatin-induced epithelial-to-mesenchymal transition in ovarian cancer cell lines. PLoS ONE 2014, 9, e86987. [Google Scholar] [CrossRef]
- Yamasaki, M.; Iwase, M.; Kawano, K.; Sakakibara, Y.; Suiko, M.; Ikeda, M.; Nishiyama, K. α-Lipoic acid suppresses migration and invasion via downregulation of cell surface β1-integrin expression in bladder cancer cells. J. Clin. Biochem. Nutr. 2014, 54, 18–25. [Google Scholar] [CrossRef]
- Lee, H.S.; Na, M.H.; Kim, W.K. α-Lipoic acid reduces matrix metalloproteinase activity in MDA-MB-231 human breast cancer cells. Nutr. Res. 2010, 30, 403–409. [Google Scholar] [CrossRef]
- Pradella, D.; Naro, C.; Sette, C.; Ghigna, C. EMT and stemness: Flexible processes tuned by alternative splicing in development and cancer progression. Mol. Cancer 2017, 16, 8. [Google Scholar] [CrossRef]
- Zhang, Z.; Xu, Y. FZD7 accelerates hepatic metastases in pancreatic cancer by strengthening EMT and stemness associated with TGF-β/SMAD3 signaling. Mol. Med. 2022, 28, 82. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.; Weinberg, R.A. Epithelial-mesenchymal transition: At the crossroads of development and tumor metastasis. Dev. Cell 2008, 14, 818–829. [Google Scholar] [CrossRef] [PubMed]
- Thiery, J.P.; Acloque, H.; Huang, R.Y.J.; Nieto, M.A. Epithelial-mesenchymal transitions in development and disease. Cell 2009, 139, 871–890. [Google Scholar] [CrossRef]
- Kalluri, R.; Weinberg, R.A. The basics of epithelial-mesenchymal transition. J. Clin. Investig. 2009, 119, 1420–1428. [Google Scholar] [CrossRef]
- Yadav, D.; Rao, G.S.N.K.; Paliwal, D.; Singh, A.; Shadab, S. Insight into the Basic Mechanisms and Various Modulation Strategies Involved in Cancer Drug Resistance. Curr. Cancer Drug Targets 2023, 23, 778–791. [Google Scholar] [CrossRef] [PubMed]
- Vaupel, P.; Mayer, A. Hypoxia in cancer: Significance and impact on clinical outcome. Cancer Metastasis Rev. 2007, 26, 225–239. [Google Scholar] [CrossRef]
- Harris, A.L. Hypoxia—A key regulatory factor in tumour growth. Nat. Rev. Cancer 2002, 2, 38–47. [Google Scholar] [CrossRef]
- Kalyane, D.; Raval, N.; Maheshwari, R.; Tambe, V.; Kalia, K.; Tekade, R.K. Employment of enhanced permeability and retention effect (EPR): Nanoparticle-based precision tools for targeting of therapeutic and diagnostic agent in cancer. Mater. Sci. Eng. C Mater. Biol. Appl. 2019, 98, 1252–1276. [Google Scholar] [CrossRef]
- Semenza, G.L. Targeting HIF-1 for cancer therapy. Nat. Rev. Cancer 2003, 3, 721–732. [Google Scholar] [CrossRef] [PubMed]
- Keith, B.; Johnson, R.S.; Simon, M.C. HIF1α and HIF2α: Sibling rivalry in hypoxic tumour growth and progression. Nat. Rev. Cancer 2011, 12, 9–22. [Google Scholar] [CrossRef] [PubMed]
- Jain, I.H.; Zazzeron, L.; Goli, R.; Alexa, K.; Schatzman-Bone, S.; Dhillon, H.; Goldberger, O.; Peng, J.; Shalem, O.; Sanjana, N.E.; et al. Hypoxia as a therapy for mitochondrial disease. Science 2016, 352, 54–61. [Google Scholar] [CrossRef] [PubMed]
- Burr, S.P.; Costa, A.S.H.; Grice, G.L.; Timms, R.T.; Lobb, I.T.; Freisinger, P.; Dodd, R.B.; Dougan, G.; Lehner, P.J.; Frezza, C.; et al. Mitochondrial Protein Lipoylation and the 2-Oxoglutarate Dehydrogenase Complex Controls HIF1α Stability in Aerobic Conditions. Cell Metab. 2016, 24, 740–752. [Google Scholar] [CrossRef] [PubMed]
- Cowman, S.J.; Koh, M.Y. Revisiting the HIF switch in the tumor and its immune microenvironment. Trends Cancer 2022, 8, 28–42. [Google Scholar] [CrossRef] [PubMed]
- Tsvetkov, P.; Coy, S.; Petrova, B.; Dreishpoon, M.; Verma, A.; Abdusamad, M.; Rossen, J.; Joesch-Cohen, L.; Humeidi, R.; Spangler, R.D.; et al. Copper induces cell death by targeting lipoylated TCA cycle proteins. Science 2022, 375, 1254–1261. [Google Scholar] [CrossRef] [PubMed]
- Galaris, D.; Pantopoulos, K. Oxidative stress and iron homeostasis: Mechanistic and health aspects. Crit. Rev. Clin. Lab. Sci. 2008, 45, 1–23. [Google Scholar] [CrossRef] [PubMed]
- Hentze, M.W.; Muckenthaler, M.U.; Andrews, N.C. Balancing acts: Molecular control of mammalian iron metabolism. Cell 2004, 117, 285–297. [Google Scholar] [CrossRef]
- Finney, L.A.; O’Halloran, T.V. Transition metal speciation in the cell: Insights from the chemistry of metal ion receptors. Science 2003, 300, 931–936. [Google Scholar] [CrossRef]
- Hernández-Rabaza, V.; López-Pedrajas, R.; Almansa, I. Progesterone, Lipoic Acid, and Sulforaphane as Promising Antioxidants for Retinal Diseases: A Review. Antioxidants 2019, 8, 53. [Google Scholar] [CrossRef] [PubMed]
- Blyth, B.J.; Cole, A.J.; MacManus, M.P.; Martin, O.A. Radiation therapy-induced metastasis: Radiobiology and clinical implications. Clin. Exp. Metastasis 2018, 35, 223–236. [Google Scholar] [CrossRef] [PubMed]
- Andarawewa, K.L.; Erickson, A.C.; Chou, W.S.; Costes, S.V.; Gascard, P.; Mott, J.D.; Bissell, M.J.; Barcellos-Hoff, M.H. Ionizing radiation predisposes nonmalignant human mammary epithelial cells to undergo transforming growth factor beta induced epithelial to mesenchymal transition. Cancer Res. 2007, 67, 8662–8670. [Google Scholar] [CrossRef] [PubMed]
- Yadav, P.; Shankar, B.S. Radio resistance in breast cancer cells is mediated through TGF-β signalling, hybrid epithelial-mesenchymal phenotype and cancer stem cells. Biomed. Pharmacother. 2019, 111, 119–130. [Google Scholar] [CrossRef] [PubMed]
- Sari Susanna, T. TRPV Family Ion Channels in the Mammary Epithelium: Role in Normal Tissue Homeostasis and along Breast Cancer Progression. In Ion Transporters; Zuzana Sevcikova, T., Ed.; IntechOpen: Rijeka, Croatia, 2022; Chapter 4. [Google Scholar]
- Yu, Z.; Xu, C.; Song, B.; Zhang, S.; Chen, C.; Li, C.; Zhang, S. Tissue fibrosis induced by radiotherapy: Current understanding of the molecular mechanisms, diagnosis and therapeutic advances. J. Transl. Med. 2023, 21, 708. [Google Scholar] [CrossRef] [PubMed]
- Puchsaka, P.; Chaotham, C.; Chanvorachote, P. α-Lipoic acid sensitizes lung cancer cells to chemotherapeutic agents and anoikis via integrin β1/β3 downregulation. Int. J. Oncol. 2016, 49, 1445–1456. [Google Scholar] [CrossRef] [PubMed]
- Li, B.J.; Hao, X.Y.; Ren, G.H.; Gong, Y. Effect of lipoic acid combined with paclitaxel on breast cancer cells. Genet. Mol. Res. 2015, 14, 17934–17940. [Google Scholar] [CrossRef] [PubMed]
- Nur, G.; Nazıroğlu, M.; Deveci, H.A. Synergic prooxidant, apoptotic and TRPV1 channel activator effects of alpha-lipoic acid and cisplatin in MCF-7 breast cancer cells. J. Recept. Signal Transduct. Res. 2017, 37, 569–577. [Google Scholar] [CrossRef]
- Shah, A.; Hoffman, E.M.; Mauermann, M.L.; Loprinzi, C.L.; Windebank, A.J.; Klein, C.J.; Staff, N.P. Incidence and disease burden of chemotherapy-induced peripheral neuropathy in a population-based cohort. J. Neurol. Neurosurg. Psychiatry 2018, 89, 636–641. [Google Scholar] [CrossRef]
- Banach, M.; Juranek, J.K.; Zygulska, A.L. Chemotherapy-induced neuropathies-a growing problem for patients and health care providers. Brain Behav. 2017, 7, e00558. [Google Scholar] [CrossRef]
- Aromolaran, K.A.; Goldstein, P.A. Ion channels and neuronal hyperexcitability in chemotherapy-induced peripheral neuropathy; cause and effect? Mol. Pain. 2017, 13, 1744806917714693. [Google Scholar] [CrossRef] [PubMed]
- Grisold, W.; Cavaletti, G.; Windebank, A.J. Peripheral neuropathies from chemotherapeutics and targeted agents: Diagnosis, treatment, and prevention. Neuro Oncol. 2012, 14 (Suppl. S4), iv45–iv54. [Google Scholar] [CrossRef]
- Hu, S.; Huang, K.M.; Adams, E.J.; Loprinzi, C.L.; Lustberg, M.B. Recent Developments of Novel Pharmacologic Therapeutics for Prevention of Chemotherapy-Induced Peripheral Neuropathy. Clin. Cancer Res. 2019, 25, 6295–6301. [Google Scholar] [CrossRef] [PubMed]
- Argyriou, A.A.; Bruna, J.; Marmiroli, P.; Cavaletti, G. Chemotherapy-induced peripheral neurotoxicity (CIPN): An update. Crit. Rev. Oncol. Hematol. 2012, 82, 51–77. [Google Scholar] [CrossRef] [PubMed]
- Low, P.A.; Nickander, K.K.; Tritschler, H.J. The roles of oxidative stress and antioxidant treatment in experimental diabetic neuropathy. Diabetes 1997, 46 (Suppl. S2), S38–S42. [Google Scholar] [CrossRef]
- Sun, H.; Guo, X.; Wang, Z.; Wang, P.; Zhang, Z.; Dong, J.; Zhuang, R.; Zhou, Y.; Ma, G.; Cai, W. Alphalipoic Acid Prevents Oxidative Stress and Peripheral Neuropathy in Nab-Paclitaxel-Treated Rats through the Nrf2 Signalling Pathway. Oxid. Med. Cell Longev. 2019, 2019, 3142732. [Google Scholar] [CrossRef]
- Gedlicka, C.; Kornek, G.V.; Schmid, K.; Scheithauer, W. Amelioration of docetaxel/cisplatin induced polyneuropathy by alpha-lipoic acid. Ann. Oncol. 2003, 14, 339–340. [Google Scholar] [CrossRef]
- Guo, Y.; Jones, D.; Palmer, J.L.; Forman, A.; Dakhil, S.R.; Velasco, M.R.; Weiss, M.; Gilman, P.; Mills, G.M.; Noga, S.J.; et al. Oral alpha-lipoic acid to prevent chemotherapy-induced peripheral neuropathy: A randomized, double-blind, placebo-controlled trial. Support. Care Cancer 2014, 22, 1223–1231. [Google Scholar] [CrossRef]
- Salehi, B.; Berkay Yılmaz, Y.; Antika, G.; Boyunegmez Tumer, T.; Fawzi Mahomoodally, M.; Lobine, D.; Akram, M.; Riaz, M.; Capanoglu, E.; Sharopov, F.; et al. Insights on the Use of α-Lipoic Acid for Therapeutic Purposes. Biomolecules 2019, 9, 356. [Google Scholar] [CrossRef]
- Shi, J.; Kantoff, P.W.; Wooster, R.; Farokhzad, O.C. Cancer nanomedicine: Progress, challenges and opportunities. Nat. Rev. Cancer 2017, 17, 20–37. [Google Scholar] [CrossRef]
- Cho, K.; Wang, X.; Nie, S.; Chen, Z.G.; Shin, D.M. Therapeutic nanoparticles for drug delivery in cancer. Clin. Cancer Res. 2008, 14, 1310–1316. [Google Scholar] [CrossRef] [PubMed]
- Hubbell, J.A.; Chilkoti, A. Chemistry. Nanomaterials for drug delivery. Science 2012, 337, 303–305. [Google Scholar] [CrossRef] [PubMed]
- Kievit, F.M.; Zhang, M. Cancer nanotheranostics: Improving imaging and therapy by targeted delivery across biological barriers. Adv. Mater. 2011, 23, H217–H247. [Google Scholar] [CrossRef] [PubMed]
- Mehlen, P.; Puisieux, A. Metastasis: A question of life or death. Nat. Rev. Cancer 2006, 6, 449–458. [Google Scholar] [CrossRef] [PubMed]
- Riehemann, K.; Schneider, S.W.; Luger, T.A.; Godin, B.; Ferrari, M.; Fuchs, H. Nanomedicine--challenge and perspectives. Angew. Chem. Int. Ed. Engl. 2009, 48, 872–897. [Google Scholar] [CrossRef] [PubMed]
- Ling, L.; Ismail, M.; Du, Y.; Yao, C.; Li, X. Lipoic acid-derived cross-linked liposomes for reduction-responsive delivery of anticancer drug. Int. J. Pharm. 2019, 560, 246–260. [Google Scholar] [CrossRef] [PubMed]
- Fang, H.; Zhao, X.; Gu, X.; Sun, H.; Cheng, R.; Zhong, Z.; Deng, C. CD44-Targeted Multifunctional Nanomedicines Based on a Single-Component Hyaluronic Acid Conjugate with All-Natural Precursors: Construction and Treatment of Metastatic Breast Tumors in Vivo. Biomacromolecules 2020, 21, 104–113. [Google Scholar] [CrossRef] [PubMed]
- Yang, F.; Chen, Y.; Zhang, J.; Liao, C.; Zhang, S. Cross-linked (R)-(+)-lipoic acid nanoparticles with prodrug loading for synergistic cancer therapy. J. Mater. Chem. B 2021, 9, 1583–1591. [Google Scholar] [CrossRef]
- Jia, M.; Lu, R.; Liu, C.; Zhou, X.; Li, P.; Zhang, S. In Situ Implantation of Chitosan Oligosaccharide-Doped Lipoic Acid Hydrogel Breaks the “Vicious Cycle” of Inflammation and Residual Tumor Cell for Postoperative Skin Cancer Therapy. ACS Appl. Mater. Interfaces 2023, 15, 32824–32838. [Google Scholar] [CrossRef]
- Jing, P.; Luo, Y.; Chen, Y.; Tan, J.; Liao, C.; Zhang, S. Aspirin-Loaded Cross-Linked Lipoic Acid Nanodrug Prevents Postoperative Tumor Recurrence by Residual Cancer Cell Killing and Inflammatory Microenvironment Improvement. Bioconjug Chem. 2023, 34, 366–376. [Google Scholar] [CrossRef]
- Cortes, J.E.; Lin, T.L.; Asubonteng, K.; Faderl, S.; Lancet, J.E.; Prebet, T. Efficacy and safety of CPX-351 versus 7 + 3 chemotherapy by European LeukemiaNet 2017 risk subgroups in older adults with newly diagnosed, high-risk/secondary AML: Post hoc analysis of a randomized, phase 3 trial. J. Hematol. Oncol. 2022, 15, 155. [Google Scholar] [CrossRef] [PubMed]
- Cucinotto, I.; Fiorillo, L.; Gualtieri, S.; Arbitrio, M.; Ciliberto, D.; Staropoli, N.; Grimaldi, A.; Luce, A.; Tassone, P.; Caraglia, M.; et al. Nanoparticle albumin bound Paclitaxel in the treatment of human cancer: Nanodelivery reaches prime-time? J. Drug Deliv. 2013, 2013, 905091. [Google Scholar] [CrossRef] [PubMed]
- Lu, H.; Zha, S.; Zhang, W.; Wang, Q.; Jiang, D.; Xu, X.; Zheng, X.; Qiu, M.; Shan, C. A systematic review and meta-analysis of nab-paclitaxel mono-chemotherapy for metastatic breast cancer. BMC Cancer 2021, 21, 830. [Google Scholar] [CrossRef]
- Martín, M. nab-Paclitaxel dose and schedule in breast cancer. Breast Cancer Res. 2015, 17, 81. [Google Scholar] [CrossRef] [PubMed]
- Huang, Y.-P.; Jin, H.-Y.; Yu, H.-P. Inhibitory effects of alpha-lipoic acid on oxidative stress in the rostral ventrolateral medulla in rats with salt-induced hypertension. Int. J. Mol. Med. 2017, 39, 430–436. [Google Scholar] [CrossRef]
- Mason, S.A.; Trewin, A.J.; Parker, L.; Wadley, G.D. Antioxidant supplements and endurance exercise: Current evidence and mechanistic insights. Redox Biol. 2020, 35, 101471. [Google Scholar] [CrossRef] [PubMed]
- Donato, A.J.; Uberoi, A.; Bailey, D.M.; Wray, D.W.; Richardson, R.S. Exercise-induced brachial artery vasodilation: Effects of antioxidants and exercise training in elderly men. Am. J. Physiol. Heart Circ. Physiol. 2010, 298, H671–H678. [Google Scholar] [CrossRef] [PubMed]
- Rossman, M.J.; Trinity, J.D.; Garten, R.S.; Ives, S.J.; Conklin, J.D.; Barrett-O’Keefe, Z.; Witman, M.A.H.; Bledsoe, A.D.; Morgan, D.E.; Runnels, S.; et al. Oral antioxidants improve leg blood flow during exercise in patients with chronic obstructive pulmonary disease. Am. J. Physiol. Heart Circ. Physiol. 2015, 309, H977–H985. [Google Scholar] [CrossRef] [PubMed]
- Khan, H.; Singh, T.G.; Dahiya, R.S.; Abdel-Daim, M.M. α-Lipoic Acid, an Organosulfur Biomolecule a Novel Therapeutic Agent for Neurodegenerative Disorders: An Mechanistic Perspective. Neurochem. Res. 2022, 47, 1853–1864. [Google Scholar] [CrossRef]
- Soreze, Y.; Boutron, A.; Habarou, F.; Barnerias, C.; Nonnenmacher, L.; Delpech, H.; Mamoune, A.; Chrétien, D.; Hubert, L.; Bole-Feysot, C.; et al. Mutations in human lipoyltransferase gene LIPT1 cause a Leigh disease with secondary deficiency for pyruvate and alpha-ketoglutarate dehydrogenase. Orphanet J. Rare Dis. 2013, 8, 192. [Google Scholar] [CrossRef]
- Cai, Y.; He, Q.; Liu, W.; Liang, Q.; Peng, B.; Li, J.; Zhang, W.; Kang, F.; Hong, Q.; Yan, Y.; et al. Comprehensive analysis of the potential cuproptosis-related biomarker LIAS that regulates prognosis and immunotherapy of pan-cancers. Front. Oncol. 2022, 12, 952129. [Google Scholar] [CrossRef] [PubMed]
- Cai, Z.; Li, C.-F.; Han, F.; Liu, C.; Zhang, A.; Hsu, C.-C.; Peng, D.; Zhang, X.; Jin, G.; Rezaeian, A.-H.; et al. Phosphorylation of PDHA by AMPK Drives TCA Cycle to Promote Cancer Metastasis. Mol. Cell 2020, 80, 263–278. [Google Scholar] [CrossRef] [PubMed]
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
Yan, S.; Lu, J.; Chen, B.; Yuan, L.; Chen, L.; Ju, L.; Cai, W.; Wu, J. The Multifaceted Role of Alpha-Lipoic Acid in Cancer Prevention, Occurrence, and Treatment. Antioxidants 2024, 13, 897. https://doi.org/10.3390/antiox13080897
Yan S, Lu J, Chen B, Yuan L, Chen L, Ju L, Cai W, Wu J. The Multifaceted Role of Alpha-Lipoic Acid in Cancer Prevention, Occurrence, and Treatment. Antioxidants. 2024; 13(8):897. https://doi.org/10.3390/antiox13080897
Chicago/Turabian StyleYan, Shuai, Jiajie Lu, Bingqing Chen, Liuxia Yuan, Lin Chen, Linglin Ju, Weihua Cai, and Jinzhu Wu. 2024. "The Multifaceted Role of Alpha-Lipoic Acid in Cancer Prevention, Occurrence, and Treatment" Antioxidants 13, no. 8: 897. https://doi.org/10.3390/antiox13080897