Please cite this paper as:
Chakraborty, S., Sarkar, S., Chattopadhyay, A. and Bandyopadhyay, D. 2023. The dual actions of melatonin as a potential oncostatic agent and a protector against chemotherapy induced toxicity. Melatonin Research. 6, 2 (Jun. 2023), 189-214. DOI:https://doi.org/https://doi.org/10.32794/mr112500149.
Review
The dual-actions of melatonin as a potential oncostatic agent and a protector against hemotherapy-induced toxicity
Souradipta Chakraborty1, Swaimanti Sarkar1, Aindrila Chattopadhyay2, Debasish Bandyopadhyay1*
Oxidative Stress and Free Radical Biology Laboratory, Department of Physiology, University of Calcutta, 92, APC Road, Kolkata-700009, India
2Department of Physiology, Vidyasagar College,39, Sankar Ghosh Lane, Kolkata-700006, India
*Correspondence: debasish63@gmail.com, Tel: +91-9433072066
Running title: Melatonin against cancer and chemotherapy toxicity
Received: February 11, 2023; Accepted: June 12, 2023
ABSTRACT
Cancer is one of the most complicated and arduous diseases, causing immense physical and emotional tribulations in the life of patients. Carcinogens can lead to genetic mutations and cancer progression either by directly binding to DNA covalently, forming cross-links, or indirectly via the generation of oxidative stress and/or by other recondite mechanisms. Despite being the most widely used treatment, chemotherapy has several adverse consequences, including acute and/or chronic toxicities. Numerous studies have demonstrated melatonin being a potential anticancer molecule with multiple activities including prevention of the initiation, promotion, and progression of cancer. In addition to its role as a potent antioxidant, melatonin exhibits its cytostatic effects by arresting the mutated cell in the G0/G1 phase, preventing epithelial-to-mesenchymal transition and inciting the immune battle against tumours, possibly by dampening MMP activities. Melatonin inhibits the MAP-K/ERK and p38 pathways and regulates NF-ĸB-mediated inflammatory responses. Melatonin exerts its anti-angiogenic activity by curbing VEGF levels, while its anti-estrogenic activity by inhibiting the cellular uptake of linoleic acid (LA). In addition, melatonin reduces the toxicities of the chemotherapy while improving its effectiveness in cancer treatment. The purpose of this review is to assemble the knowledge available on melatonin’s oncostatic role and its protective effects against chemotherapy-induced toxicities. Further studies are needed to investigate the adjunctive role of melatonin with chemotherapy in the clinical setting and to corroborate its effectiveness in cancer cure.
Key words: Melatonin, cancer, chemotherapy, anticancer drug, oncostatic, toxicity
1. INTRODUCTION
Cancer is one of the major causes of death worldwide, and its rising trend will cause serious consequence for premature mortality in the years to come (1). In 2020, it was responsible for approximately 10 million fatalities, with an estimated 19.3 million new cases in both sexes across all cancer types. Among them around 58.3% of cancer fatalities occurred in Asia (2).
Accumulating evidence suggests various risk factors leading to cancer initiation and progression. These include smoking, alcohol abuse, cancer-inflicting viruses, diet, pollution, ionizing and non-ionizing radiation, heavy metals, medicinal drugs, and others (3-5). Besides, several acquired or inherited factors may emerge as prominent aetiologies of cancer (6, 7). Both intrinsic and extrinsic factors are capable of inducing DNA damage. Mutations in genes that have the sustained DNA damage due to impaired repair mechanisms promote cell proliferation and impair apoptosis, causing tumorigenesis (8-11). DNA damage caused by oxidative stress can specifically cause cytosolic chromatin fragments and further aggravates this damage (12). In general, genome instability is a driving force behind intratumor heterogeneity and the progression of cancer (13, 14).
Currently, the main treatments for cancers include radiation, chemotherapy, and surgery. The use of these therapies singly or in combination has limited effectiveness, therefore, identification and development of new cancer-preventive agents are required (15). Classical anticancer therapies like radiotherapy and chemotherapy impact both normal and malignant cells and elicit side effects like nausea, fatigue, vomiting, pain, etc. (16, 17). They also lead to disruption in normal cell-growth signalling and DNA damage which result in organ-specific toxicities and cell death (18, 19).
Melatonin (N-acetyl-5-methoxytryptamine), is a molecule produced principally by the pineal glands in humans and other animals in response to darkness (20). It participates in the regulation of circadian rhythms but also exerts its versatile biological activities including cell cycle regulation, maintenance of mitochondrial function, bone metabolism, reproductive modulation and endocrine homeostasis (21, 22).
Multiple studies have shown the anti-carcinogenic effect of melatonin through its receptor-dependent and independent mechanisms (23). Furthermore, melatonin has shown its potential in ameliorating the toxicity induced by chemotherapeutic drugs while increasing the efficiency of chemotherapy with reducing adverse effects. This therapeutic potential is redolent of melatonin’s possible efficiency as an adjuvant of anticancer therapies (24, 25).
This review explores the current knowledge regarding the therapeutic potential of melatonin against cancer as well as reducing the side effects of chemo-therapeutic drugs, while highlighting its potential mechanisms.
2. CARCINOGENS AND CARCINOGENESIS
In early epidemiological studies of cancer, the first recognized carcinogens were associated with certain occupations (26). Yamagiwa and Ichikawa have identified that toxic chemicals are capable of causing cancer directly (27). After being biologically activated to reactive intermediates, environmental toxins have been shown to induce mutations in key genes. These chemicals play a critical role in promoting tumorigenesis by stimulating the proliferation of cells containing oncogenic mutations. Additionally, these chemicals can provoke oxidative damage to cellular macromolecules and disrupt metabolic pathways leading to carcinogenesis (28-31). The term of carcinogens refers to substances that, irrespective of their origin, are capable of causing DNA damage and mutations via several mechanisms, therefore, resulting in cancer development (32-35).These are agents that increase the prevalence of cancer in relevant organisms compared to concurrent and/or historic controls (36). Some common carcinogenic mechanisms include oxidative stress, interactions with cellular receptors, chronic inflammation, immunosuppression, and epigenetic changes such as modifications of histones and cellular structural components (37).
The IARC (International Agency for Research on Cancer) has defined ten key attributes of human carcinogens, and as per their specifications, a carcinogen 1. Can be activated metabolically or is electrophilic; 2. Is genotoxic; 3. Alters DNA repair or disrupts genomic stability; 4. Promotes epigenetic alterations; 5. Generates oxidative stress; 6. Produce chronic inflammation; 7. Is immunosuppressive; 8. Influences receptor-mediated effects; 9. Results in immortalization; 10. Alters nutrient supply, cell proliferation, and cell death (33, 38-40).
3. MODE OF ACTION OF CARCINOGENS
Cancer pathogenesis involves a complex interaction between extrinsic and intrinsic factors. It usually manifests through an uncontrolled proliferation of genetically altered cells (41). Fieser and others have proposed that carcinogen metabolism plays a critical role in cancer development. These were later confirmed by animal studies conducted by James and Elizabeth Miller (42-46). The study of Omura and Sato revealed that the carcinogenic chemicals are metabolized principally by cytochrome p450s or cyps, which are considered phase I xenobiotic-metabolizing enzymes (47, 31). Furthermore, phase II xenobiotic-metabolizing enzymes (like glutathione-S-transferases, Uridine diphosphate-glucuronosyltransferases, etc.) have also been identified to play a crucial role in the metabolism of carcinogenic chemicals (28). Chemical carcinogens can also lead to polymorphisms in phase I and II xenobiotic-metabolizing enzymes, which have been associated with a modified risk of cancer development (29, 30, 48).
Based on their carcinogenic mechanisms, all carcinogens can be categorized as genotoxic or non-genotoxic one (29). The summary and modes of actions of common carcinogens are described in Table 1.
Table 1: Summary and modes of action of common carcinogens.
CLASSIFICATION OF CARCINOGENIC AGENTS | MODE OF ACTION | REFERENCES |
Genotoxic carcinogen
Direct-acting or activation- independent carcinogens
Indirect-actingor activation-dependent carcinogens | They interact with the genomic DNA of the target cell and damage it, resulting in mutations that are transmitted down to the daughter cells.
1. Carcinogens that are capable of producing cancer without being metabolized. 2. They generally exist as highly reactive electrophilic molecules, that directly bind to and interact with DNA and other cellular macromolecules. 3. They frequently lead to tumor formation at the location of their exposure, because of their high reactivity. 4. The Ames mutagenesis bioassay typically detects these carcinogens without additional metabolic activation. 5. Some examples of these carcinogens include Infrared and Ultraviolet radiation, alkylating agents, epoxides, sulphate, and alkyl esters, propiolactone, halo ethers (bis (chloromethyl) ether), halogen derivatives like mustard gasses, imines, nitrosamides, and nitrosoureas, etc.
1. Most DNA-reactive carcinogenic agents possess a procarcinogenic form. 2. The bioactivation of procarcinogens via P450 can lead to the formation of the carcinogen’s DNA binding and reactive form. 3. Indirect-acting carcinogens are generally unable to cause cancer at the application site, rather, they are more likely to cause cancer in the tissues that can activate metabolically the procarcinogenic form into its DNA-reactive form. 4. Aflatoxins and polycyclic aromatic hydrocarbons (PAHs) are examples of DNA-reactive indirect-acting carcinogens.
| (49)
(50-54)
(50, 55-59)
|
Non-genotoxic or non-DNA reactive carcinogens | 1. Many chemicals can cause cancer through pathways that do not entail the direct binding of chemicals or their metabolites to DNA or bring about heritable genetic changes. 2. Most of the time, non-DNA reactive carcinogens cause tumor formation in tissues that exhibit a high rate of spontaneous tumorigenesis. 3. These agents that work via non-DNA reactive mechanisms are frequently associated with the promotion of the tumor stage of the carcinogenesis process, requiring long-term exposure and relatively high levels for tumour to develop. 4. There are several mechanisms by which they exert their carcinogenic effect-like a2u-globulin binding, sustained cytotoxicity and proliferation of cells, the generation of oxidative stress, and receptor-mediated effects (cytochrome P450 induction, proliferation of peroxisomes), and altered DNA methylation. 5. Examples of non-genotoxic carcinogens include: endocrine modifiers, dichlorodiphenyltrichloroethane (DDT), carbon tetrachloride (CCl4), etc. | (50, 60-62) |
In addition to these mechanisms, some alterations in a subset of tumor suppressor genes and proto-oncogenes have been explicitly linked to cancer initiation (46). The SPI-1 (Spi-1 proto-oncogene) transcription factor contributes to cancer progression through the regulation of oncogene transcription (63). Some examples of tumor-suppressor genes are retinoblastoma 1 protein (RB1) and P53. The inactivation or absence of tumor suppressor genes due to mutations leads to cancer (64).
Tissue-specific alterations are also elicited by some hormonally active agents like steroids, peptide hormones, and biogenic amines that interact with selective receptors to exert such alterations. Trophic hormones have been shown to cause cell proliferation in the organs they target to. When hormonal regulatory mechanisms are compromised, certain hormones display a persistent hike in their circulatory levels. Such activities may contribute to tumor development. The development of ovarian neoplasms by decreasing estradiol levels and increasing luteinizing hormone levels, as well as, in rats, the initiation of thyroid tumors by phenobarbital-type P450 inducers, are two well-studied examples (46, 65, 66).
4. ANTICANCER AGENTS
Chemotherapy, which involves the use of low molecular weight drugs, is among the most effective cancer treatments available, and the search for new drugs persists along with conventional therapeutic practices (67, 68). During the 1940s, the utilization of chemotherapy was initiated with nitrogen mustards, which are incredibly potent alkylating agents, and antimetabolites. Because of the early success of these initial strategies, an enormous number of other anticancer medications have been developed (69). Currently, with the increasing prevalence of cancer, cytotoxic drug treatments are gaining popularity, and these therapeutic approaches are capable of completely curing some cancers. However, the success of cancer treatment varies greatly depending on the types of cancers and the stages of diagnosis (70, 67).
Conventional Anticancer agents are categorized based on their mode of action and include DNA-interactive agents like alkylating agents (such as cyclophosphamide), microtubular poisons (e.g., paclitaxel), antimetabolites (like 5-fluorouracil [5-FU]), topoisomerase inhibitors (such as irinotecan), hormones, molecular targeting agents, cytotoxic antibodies (e.g., bleomycin), and other biological agents (71,72).
The main objective of chemotherapy is to prevent the proliferation of tumours, thereby averting invasion and metastasis. However, chemotherapy can damage healthy cells (73). Although traditional cytotoxic antineoplastic drugs, particularly platinum-based drugs like carboplatin, oxaliplatin, and cisplatin have achieved major success over the past decades,they generally have narrow therapeutic windows. This was primarily attributed to their high toxicity, lack of tumor specificity, poor water solubility, and the possibility of inherent and acquired drug resistance. All these have severely limited their use (74-79). Other frequently clinical used anticancer drugs including paclitaxel, fluorouracil, and doxorubicin also exhibit similar limitations (76). These drugs also foster acute and long-term side effects, which may be reversible and irreversible (80). The toxic effects of some anticancer drugs on different tissues have been discussed in detail in Table 2. Such adverse toxicities not only limit the therapeutic potential of anticancer drugs but also decrease the rate of survival and quality of life of cancer patients. Thus, the demanding for new anticancer agents, which are more efficacious and less toxic was urgently required (76).
Table 2: Summary of classic anticancer drug-induced organ toxicities.
ANTICANCER DRUG-INDUCED ORGAN TOXICITY | CONSEQUENCES | REFERENCES |
Hematological toxicity | a. Chemotherapywith carboplatin, cyclophosphamide, melphalan, etc. may jeopardize activity of proliferating haematopoietic precursor cells, resulting in the depletion offormed elements, and causing life-threatening infections and hemorrhages. b. Peripheral cytopenia due to the suppression of the bone marrow is a frequently observed chemotherapy side effect that can lead to acute and chronic marrow destruction. c. Cyclophosphamide has been reported to alter hematopoietic factors, leading to myelotoxicity. | (81-84) |
Cardiotoxicity | a. Cardiotoxicity is known to be caused by anthracenediones and alkylating agents. b. ROS-mediated oxidative damage lies at the core of these drug-mediated cardiotoxicities. c. Both prostate and breast cancer drugs are included in cardiotoxicity warnings on d. Some drugs can lead to cardiotoxicity by inhibition of mitochondrial protection pathways. e. Taxanes are responsible for the prolongation of the QT interval, leading to atrial fibrillation and bradycardia. f. Damage to cardiomyocytes and necrosis caused by thromboembolism and vascular damage are reported during alkylating drug usage. | (85-89) |
Gastrointestinal toxicity | a. The side effects of GI like nausea, bloating, vomiting, constipation, ulceration, and particularly diarrhea caused by chemotherapy are major hurdles resulting in delays, reduction of doses, and cessation of treatment in many cancer patients. b. Mucositis, which involves inflammation and loss of epithelial cells in the barrier lining of the GI (gastrointestinal) tract, is one of the most severe side effects of chemotherapy. Stomatitis, diarrhea, dysphagia, oesophagitis, proctitis with pain and bleeding, and oral ulceration arise from chemotherapy. c. The most commonly used anticancer agents for colorectal cancer, such as 5-fluorouracil, cisplatin, oxaliplatin and irinotecan are linked with mucositis and gastrointestinal dysfunction. | (90-97) |
Neurotoxicity | a. The neurotoxicity caused by vinca alkaloids (vindesine, vincristine, and vinblastine) may be dosage dependent and includes peripheral, autonomic, and cranial neurotoxicity. b. Classic anticancer agents including doxorubicin, paclitaxel, cisplatin and current anticancer agents like bortezomib and trastuzumab can cause peripheral neuropathy. c. Several studies have demonstrated that chemotherapy can cause short and long-term mood changes as well as cognitive deficits, including impairment in learning, attention, and concentration, speed of information processing, short-term and working memory, and executive decisions (like decision-making, multitasking, language) d. Oxaliplatin can cause tight junction disassembling and brain dysfunction in the endothelial cell line (RBE4) of rat brains. | (98-102)
|
Nephrotoxicity | a. Another vital side effect is nephrotoxicity since many patients receiving chemotherapy present impaired renal function. b. The activation of the p53, DNAase 1, and MAP kinase pathways, as well as the increase in TNF-α are involved in cisplatin-induced nephrotoxicity. c. Cyclophosphamide causes hemorrhagic cystitis, which is closely associated with hyponatremia. d. Nephrotoxicity has also been reported with biologics like cetuximab and bevacizumab. Bevacizumab can cause proteinuria via immune complex-mediated focal glomerulonephritis, and hypomagnesemia is caused by cetuximab by inactivating the TRPM6 magnesium channel. e. Essentially, both biologics and small molecules impair renal excretion by affecting glomerular perfusion and vasculature, resulting in nephrotoxicity. f. There have been reports of renal failure, which may be to some extent attributed to rhabdomyolysis, associated with the use of a marine-derived alkylating agent, trabectedin for treating advanced sarcoma of soft tissue. | (103-105) |
Hepatotoxicity | a. The most commonly used antimetabolite, methotrexate, has been linked to hepatotoxicity through the activation of inflammatory pathways, ROS generation, and cytokines. b. Peroxidation of membrane lipids and disruption of mitochondrial energy metabolism induced by doxorubicin can also lead to liver damage. c. Fatal hepatotoxicity along with severe reactions has been caused by Imatinib. | (103, 106, 107) |
5. MELATONIN, AS AN ONCOSTATIC AGENT
In 1958, Aaron Lerner discovered and isolated melatonin from the bovine pineal gland (108). The pineal gland secretes melatonin as its primary hormone. Extrapineal sources of melatonin include the bone marrow cells, retina, skin, platelets, Harderian gland, lymphocytes, cerebellum, and particularly the digestive tract of vertebrates (109). Melatonin is produced not only in animal tissues but also in plants and their derivatives. Melatonin has been identified in wine, olive oil, tomatoes, beer, and juices. Darkness is a prerequisite for pineal melatonin synthesis and its secretion has circadian and in seasonal rhythms regulated via the light-sensitive retino-pineal pathway (110).
Melatonin exerts multiple effects, both through receptor-independent and receptor (MT1 and MT2)-dependent mechanisms (111-114). It is well known for its antioxidant and anti-inflammatory properties, and its effect on sleep-wake cycles, metabolism, and reproduction. The effects of melatonin on cancer have been investigated extensively in recent decades identifying its anti-proliferative, anti-oxidative, pro-apoptotic, cytotoxic, and differentiative activities, as well as its potential to modulate epigenetic processes in cancer cells (110).
6. MELATONIN AS AN ANTIOXIDANT
An imbalance between the production and elimination of free radicals results in oxidative stress (115, 116). In addition to DNA damage, oxidative stress causes damage to other macromolecules, which also contributes to tumor development and progression (117). Furthermore, excess free radicals can disrupt intercellular signalling linked to tumorigenesis (116). Several mechanisms are responsible for the antioxidative function of melatonin, including its direct role as a free radical scavenger, the enhancement of activity of antioxidant enzymes and their gene expression, and the suppression of pro-oxidant enzymes. To preserve mitochondrial function and chelating Fe2+/3+ to inhibit harmful Fenton reaction are also the mechanisms of melatonin to reduce ROS production (118). In addition to protecting DNA from oxidative damage, melatonin maintains genomic stability (119). It also regulates oxidative stress by impeding the NF-ĸB pathway (120). It’s noteworthy that melatonin's antioxidant activity is conditional regarding the reduction of oxidative stress. Under certain circumstances, such as in malignancies, melatonin may paradoxically play the role of a pro-oxidant, causing cell death by apoptosis (121).
7. MELATONIN IN THE INHIBITION OF TUMOR PROGRESSION
Melatonin is capable to inhibit the proliferation of neoplasms via cytostatic and cytotoxic mechanisms, making it an effective antitumor agent (122). The cytostatic action of melatonin was demonstrated in a breast cancer cells, MCF-7 cells. In these cells, melatonin restrains them in the G0/G1 phase or delays their transition to the S phase of the cell cycle (122-124). The inhibitory effect of melatonin on proliferation of ovarian cancer cells (OVCAR-429) was also observed in a dose- and time-dependent manner when the cells were incubated with melatonin (at a concentration of 400-800 µM) for 24-72 h (125).
Melatonin has been demonstrated to have anti-metastatic properties in some types of cancer, specifically by preventing EMT (epithelial-to-mesenchymal transition) (126). Melatonin significantly reduces MAP-K/ERK signalling activity, which is regulated by the human epidermal growth factor receptor 2 (HER2) known for promoting human breast cancer cell invasion and metastasis (127, 128). Melatonin’s anti-invasive effect may be attributed to its suppression of the p38 pathway and down-regulation of metalloproteinases-2 and -9 activity and expression (129).
The antitumor properties of melatonin including the potential to induce apoptosis, an effect observed only in cancer cells lead to a substantial reduction in the volume of cancer and an improvement in patient health (15, 130-132). In cultured human adenocarcinoma cells treated with melatonin, caspase 3 activity was increased, the expressions of Bax and p53 were up-regulated but Bcl-2 was down-regulated (133).
8. ANTI-ANGIOGENIC EFFECT OF MELATONIN
The process of angiogenesis involves the formation of new blood vessels and is crucial to wound healing, embryonic development, and carcinogenesis. Tumors that lack vascular support are prone to necrosis and even apoptosis (134). In both normal and malignant cells, VEGF (vascular endothelial growth factor) is essential for new blood vessel development and for promoting angiogenesis. VEGF has three key receptors: VEGF-1, 2, and 3. However, its main function is mediated via VEGF-1(135-138). Declining circulating VEGF levels in serum correlate with the strong response seen in patients with static-metastatic cancers (139).
Melatonin inhibits HIF-1α (Hypoxia-inducible factor 1-alpha) in prostate cancer cells by suppressing the translation of proteins (140). Importantly, melatonin, supresses tumor growth by inhibition angiogenesis but in normal cells it stimulates angiogenesis to promote wound healing (141).
9. EFFECT OF MELATONIN ON THE IMMUNE SYSTEM
Numerous studies have shown a strong association between the immune system and melatonin (142-145). Melatonin can regulate cytokine production and immune cell proliferation via its receptors present in immune cells (146, 147). Melatonin can restore impaired T-helper cell activity, modulate lymphocytes, induce the proliferation of T-lymphocytes, protect CD4+ T-cells against apoptosis, and prevent the apoptosis of B-cell precursors in the bone marrow (148). It triggers the generation of IL-2, IL-10, IL-6, IL-12, and IFN-γ in mononucleated cells, resulting in a TH-1 lymphocyte response (149). It stimulates the activity of natural killer cells in humans as well as increases the antitumor properties of the immune system (150). Liu et al. found that melatonin suppressed the regulatory T cells (Tregs) in mice bearing gastric cancer tumors, leading to cell death (151).
10. MELATONIN’S ANTI-ESTROGENIC PROPERTIES AND INHIBITION OF LINOLEIC ACID UPTAKE IN CANCER CELLS
Numerous investigations have revealed that estrogen is critical to the proliferation of neoplastic breast epithelium (152-154). Estrogen receptor expression increases as cancer progresses (154, 155). Studies have demonstrated that melatonin reduced the proliferation of breast cancer by inhibiting estrogen receptors (156, 157). Moreover, treatment with melatonin inhibited the proliferation of human breast cancer stem cells by down-regulating the transcription factor OCT4 and estrogen receptor-a (158).
In addition to its interactions with estrogen, melatonin decreases linoleic acid (LA) uptake, a crucial component essential for the initiation of the cellular pathways linked to hormone-dependent malignancies of the breast (159).
11. EPIGENETIC ACTIONS OF MELATONIN
The ability of melatonin to switch genes on and off can aid in controlling epigenetic regulations. For instance, melatonin can control transcription factors that are affected by inflammatory and nitro-oxidative conditions (159). Inflammation is caused by pathways governed by the family of activator protein-1 (AP-1) and NF-κB. These pathways directly lead to the activation of pro-inflammatory mediators and cytokines such as interleukins, TNF-α, cyclooxygenase-2 (COX-2), matrix metalloproteinases (MMPs), and cytokine-inducible nitric oxide synthase (iNOS). The activation of these transcription factors triggers epigenetic modification via altering the chromatin's structure, either by acetylating histones or by methylating DNA (160). According to numerous studies, melatonin selectively suppresses COX-2, iNOS and MMPs resulting in the inhibition of NF-κB (161-163). The schematic diagram (Figure 1) shows the potential role of melatonin as an oncostatic molecule.
Fig. 1. The potential roles of melatonin as an oncostatic molecule.
Carcinogens can lead to cell mutation and cancer formation and progression either directly by binding with DNA to form DNA cross-links or indirectly by the generation of free radicals and induction of the Cytochrome P450 enzyme. Tumor suppressor genes (RB1 and P53) and oncogenes (SPI-1) along with increased levels of luteinizing hormone and decreased levels of estradiol contribute to cancer development.Melatonin, in addition to its role as a potent antioxidant, exhibits cytostatic effects by arresting the mutated cell in the G0 or G1 phase or delaying its transition to the S phase. It also prevents epithelial-to-mesenchymal transition. Melatonin is also involved in the upregulation of the immune system. It significantly suppresses the Mapk/ERK and p38 pathways and regulate the NF-ĸB pathway. Melatonin exerts its anti-angiogenic properties by curbing VEGF levels. It also exhibits anti-estrogenic properties and inhibits LA uptake by the cells.
Green tranangle: melatonin, Blue box: anticancer drugs, PXR: Pregnane X Receptor, RXR: retinoid X receptor, TMEFF1: Transmembrane Protein with EGF-like and 2 Follistatin-like domains 1, FATP: Fatty acid transport proteins, ERE: estrogen response element, E2: estradiol, 13-HODE: 13-Hydroxyoctadecadienoic acid, Ras: rat sarcoma
12. THE ROLE OF MELATONIN IN THE PREVENTION OF ANTICANCER DRUG-MEDIATED TOXICITY
Melatonin’s oncostatic and immunomodulatory properties support its use in conjugation with conventional anticancer treatment (164). Melatonin and classic chemotherapeutic drugs as a chemohormonotherapeutic combination are further substantiated by its ability to lower the toxicity of anticancer drugs (165). The multifaceted role of melatonin in mitigating various anticancer drug-induced toxicity is discussed in Table 3.
Table 3: Summary of the preventive role of melatonin against toxicities of various anticancer drugs.
TOXICITY INDUCED BY ANTICANCER DRUGS | PREVENTIVE ROLE OF MELATONIN | REFERENCES
|
ANTHRACYCLIN-INDUCED TOXICITY | l Melatonin exerted cardioprotective effects against damage induced by doxorubicin in rats, by decreasing ST segments elevation, increasing the R-amplitude, reducing serum levels of cardiac injury markers, protecting the activity of antioxidant enzymes, decreasing lipid peroxidation, and modifying serum lipid profile. l Melatonin, when given with daunorubicin, decreased the proportion of apoptotic cardiomyocytes and suppressed nitrosative stress induced by epirubicin. | (166-168) |
PLATINUM DRUG-INDUCED TOXICITY | l Melatonin inhibited cell death induced by cisplatin and increased the expression of antiapoptotic Bcl-2 gene and protein while decreasing levels of phosphorylated apoptotic protein p53, Bax, and cleaved caspase 3 and also significantly reduced cisplatin-induced DNA fragmentation and renal cytotoxicity. l Melatonin protected against peripheral neuropathy induced by oxaliplatin. l Melatonin reduced the levels of mitochondrial lipid peroxidation and protein carbonyl content induced by oxaliplatin and controlled the alterations in mitochondrial enzymatic and non-enzymatic antioxidants as well as complex respiratory enzymes. l In preclinical and clinical studies on neuroinflammatory pain, melatonin exerted antiallodynic and anti-inflammatory effects. | (169-172) |
ALKYLATING AGENT-INDUCED TOXICITY | l Melatonin exerted anti-inflammatory effects mitigated toxicity induced by alkylatingagentsvia the mechanism of inhibition of iNOS and scavenging peroxynitrite. l Melatonin had powerful antigenotoxic effects and inhibited chromosomal aberrations induced by cyclophosphamide toxicity in mice, possibly by the elevated antioxidant status by melatonin. l Melatonin may also ameliorate the spermatological toxicity induced by busulfan in male patients. | (173-177) |
ANTIMETABOLITES-INDUCED TOXICITY | l Pretreatment with melatonin reduced methotrexate-induced oxidative stress, altered antioxidant enzyme activity, and increased the activity of myeloperoxidase, implying that melatonin might reduce renal damage through anti-inflammatory and antioxidant actions. l Melatonin also alleviated the cognitive defects which were the result of 5-fluorouracil-induced hippocampal neurodegeneration.
| (178-180) |
MITOTIC INHIBITORS-INDUCED TOXICITY | l Melatonin treatment during taxane chemotherapy was associated with a lower occurrence of peripheral neuropathy. l Melatonin reduced mitochondrial damage and neuropathic pain caused by paclitaxel. | (181) |
MOLECULAR TARGETED ANTIBODIES-INDUCED TOXICITY | l The humanized monoclonal antibody trastuzumab targets the extracellular domain of HER2 and is used in HER2-positive breast cancer patients as a crucial part of adjuvant treatment and metastasis therapy. Nonetheless, its serious side effects restrict its use. l The levels of serum CK-MB and oxidative stress markers were highly increased following trastuzumab treatment; melatonin administration reversed these changes, returning them close to normal levels, implying that melatonin is beneficial in reducing cardiotoxicity induced by trastuzumab. | (182-184) |
13. MELATONIN AS A POTENTIAL ADJUVANT THERAPEUTIC IN CANCER TREATMENT
Several studies have examined the effectiveness of melatonin as an anticancer treatment in combination with chemotherapy in cancers such as cervical, breast, colon, lung, hematological, hepatic, and others (185). Melatonin exerts a range of anticancer properties at various stages of tumor progression and metastasis (186). Furthermore, melatonin in conjugation with chemotherapies not only increased the efficacy of anticancer drugs (187-189) but improved the cancer-related symptoms like cachexia, delirium, and insomnia (110).
This indoleamine can also enhance the anticancer activity of doxorubicin by preventing its outflow from cancer cells mediated by P-glycoprotein (190). In the MCF-7 human breast cancer cells, doxorubicin and melatonin in combination were more effective in eliciting apoptosis, depolarization of the mitochondrial membrane, and activation of caspase 3 and caspase 9 than doxorubicin alone (191). It promotes cisplatin-induced cell cycle arrest and apoptosis in human lung adenocarcinoma cells (192). Melatonin significantly enhanced the activities of 5-fluorouracil to inhibit colon cancer cell proliferation, formation of colonies, cell migration, and invasion. Melatonin and 5-fluorouracil synergistically induced cell cycle arrest via caspase/PARP-dependent apoptosis pathway activation (193). Melatonin also enhanced paclitaxel’s antitumor effects in the endoplasmic reticulum of endometrial cancer cell line expressing MT1 receptor (25).
Melatonin at a dose of 20 mg/day orally in combination with chemotherapy increased the survival of patients by 1 year and potentiated tumor regression (194). Another study showed that combination of etoposide cisplatin and melatonin had a 6% chance of survival rate after five years, whereas no patients survived who received etoposide and cisplatin only (195). Melatonin, when combined with radiotherapy, increased the survival rate by 153.5 months as compared to 64.0 months in patients without melatonin co-treatment (196). Furthermore, it significantly reduced myelosuppression, and neuropathy, indicating a better tolerance to chemotherapy for patients treated with anticancer drugs (197).
14. EMERGING ROLE OF MELATONIN IN THE MODULATION OF TUMOR MICROENVIRONMENT
Recent work indicates that the melatonergic pathway is present in all cells of humans, primarily mitochondrial melatonergic pathway is proposed to be a significant target of tumor fluxes in the tumor microenvironment (198, 199). Notably, the melatonergic pathway does not always control the production of melatonin but, regulate the N-acetylserotonin (NAS)/melatonin ratio, including from the pineal gland at night (200). Melatonin can be ‘backward’ converted to NAS by a number of factors, including the aryl hydrocarbon receptor (AhR)-induced cytochrome P450 (CYP)1A2 and CYP1b1 (201). As tumor-induced kynurenine release activates the AhR to modulate immune responses in the tumor microenvironment, AhR activation is an important aspect of tumor microenvironment pathophysiology, including via AhR-induced NAS (202). NAS, a brain-derived neurotrophic factor (BDNF) mimic can activate the BDNF receptor, tyrosine receptor kinase B (TrkB) (203). This would indicate that an increase in NAS/melatonin ratio may be of importance to tumor survival, that is, AhR activation stimulates TrkB activity to increase the proliferation and survival of cancer stem-like cells (204).
Regulation of tumor initiation and pathophysiology by melatonin may be partly mediated via its uptake by mitochondria via the peptide transporter (PEPT)1/2 and the organic anion transporter (OAT)3 (205-207). Melatonin not only has antagonistic interactions with the AhR, but can also induce the mitochondrial melatonergic pathway in cells of the tumor microenvironment, thereby altering the intercellular ‘homeostatic’ interactions of cells in the tumor microenvironment (208). The intercellular interactions of melatonin on tumor microenvironments with relevance to both the pathoetiology and physiology of cancer is required for further investigation.
15. SUMMARY AND CONCLUSION
An array of studies has looked into the melatonin as a powerful salutary molecule with diverse biological activities including oncostatic, anti-oxidative, immunomodulatory, and anti-inflammatory properties. Results from these studies have shed light to use melatonin as a potential adjuvant of chemotherapy (209,210).
Melatonin reduces the interaction of carcinogens with DNA and decreases the oxidative stress caused by carcinogens. Melatonin also protects against damage to cellular macromolecules and other elusive mechanisms. The antitumor effects of melatonin are amplified by its activities blocking cell proliferation, enhancing tumor apoptotic pathways, strengthening the immune system, and suppressing angiogenesis. In addition, melatonin reduces toxicities of various antineoplastic drugs on normal cells, therefore, enhances the tolerance of patients to chemotherapy.
This review explores melatonin's potential as an adjuvant in combination with chemotherapy for improving survival and quality of life for cancer patients. Further studies are required to unravel the clinical outcomes of melatonin as an adjuvant with classic anticancer drugs. Furthermore, mechanisms of melatonin in mitigating the toxicities of anticancer drugs should be further explored. In conclusion, our review encourages further research and clinical trials to identify the effectiveness of melatonin in inhibiting carcinogens, checking cancer development, and ameliorating toxicities induced by anticancer drugs. Understanding the underlying molecular events will present a clear picture of the mode of action of melatonin, and its role as a potential adjuvant in cancer therapy will obtain greater recognition.
ACKNOWLEDGEMENTS
Souradipta Chakraborty is extremely grateful for the financial assistance that she has received as a Junior Research Fellow(JRF) [200510284471] under the UGC scheme, Govt. of India. Swaimanti Sarkar is receiving financial assistance as a Senior Research Fellow (SRF) [709/(CSIR-UGC NET DEC. 2018] under the Joint CSIR-UGC scheme, Govt. of India. Dr. Aindrila Chattopadhyay is supported by funds available to her from Department of Science and Technology, Govt. of West Bengal. Prof. Debasish Bandyopadhyay thankfully acknowledges the support he received from Departmental BI Grant and DST-PURSE Program awarded to the University of Calcutta.
AUTHORSHIP
The concept of the review article was developed by Dr. DB, Dr. AC, SS and SC. Moreover, SC and SS contributed in drafting the manuscript, prepared the figures, and edited it. Dr. DB and Dr. AC revised the manuscript critically and finally approved it.
CONFLICT OF INTEREST
Authors declare no conflict of interest.
REFERENCES
Bray F, Laversanne M, Weiderpass E, Soerjomataram I. (2021). The ever-increasing importance of cancer as a leading cause of premature death worldwide. Cancer 127 (16): 3029-3030. doi:10.1002/cncr.33587.
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F (2021) Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA. Cancer J. Clin.71 (3): 209–249. doi:10.3322/caac.21660.
Danaei G, Vander Hoorn S, Lopez AD, Murray CJ, Ezzati M, Comparative Risk Assessment collaborating group (Cancers) (2005) Causes of cancer in the world: comparative risk assessment of nine behavioural and environmental risk factors. Lancet 366 (9499): 1784–1793. doi:10.1016/S0140-6736(05)67725-2.
Najafi M, Cheki M, Rezapoor S, Geraily G, Motevaseli E, Carnovale C, Clementi E, Shirazi A (2018). Metformin: Prevention of genomic instability and cancer: A review. Mutat Res Genet Toxicol. Environ. Mutagen. 827: 1–8. doi:10.1016/j.mrgentox.2018.01.007.
Stewart BW, Wild CP (2014) “Cancer etiology”. World Cancer Report 2014. World Health Organization, pp. 81-176. Available online: http://www.searo.who.int/publications/bookstore/documents/9283204298/en/ [Google Scholar].
Boffetta P (2000) Molecular epidemiology. J. Intern. Med. 248 (6): 447–454. doi:10.1046/j.1365-2796.2000.00777.x.
Boffetta P, Islami F (2013) The contribution of molecular epidemiology to the identification of human carcinogens: current status and future perspectives. Ann. Oncol. 24 (4): 901–908. doi:10.1093/annonc/mds543.
Goldar S, Khaniani MS, Derakhshan S M, Baradaran B (2015) Molecular mechanisms of apoptosis and roles in cancer development and treatment. Asian Pac. J. Cancer Prev. 16 (6): 2129–2144. doi:10.7314/apjcp.2015.16.6.2129.
TorgovnickA, Schumacher B (2015) DNA repair mechanisms in cancer development and therapy. Front. Genet. 6: 157. doi:10.3389/fgene.2015.00157.
Basu A K (2018) DNA Damage, Mutagenesis and Cancer. Int. J. Mol. Sci. 19 (4): 970. doi:10.3390/ijms19040970.
Vilenchi MM, Knudson AG (2003) Endogenous DNA double-strand breaks: production, fidelity of repair, and induction of cancer. Proc. Natl Acad. Sci. USA. 100 (22): 12871–12876. doi:10.1073/pnas.2135498100.
Han X, Chen H, Gong H, Tang X, Huang N, Xu W, Tai H, Zhang G, Zhao T, Gong C, Wang S, Yang Y, Xiao H (2020) Autolysosomal degradation of cytosolic chromatin fragments antagonizes oxidative stress-induced senescence. J. Biol. Chem. 295 (14): 4451–4463. doi:10.1074/jbc.RA119.010734.
Liu X, Li, Huang Q, Zhang Z, Zhou L, Deng Y, ZhouM, Fleenor DE, Wang H, Kastan M B, Li CY (2017) Self-inflicted DNA double-strand breaks sustain tumorigenicity and stemness of cancer cells. Cell Res. 27 (6): 764–783. doi:10.1038/cr.2017.41
Raynaud F, Mina M, Tavernari D, Ciriello G (2018) Pan-cancer inference of intra-tumor heterogeneity reveals associations with different forms of genomic instability. PLoS Genet. 14 (9): e1007669. doi:10.1371/journal.pgen.1007669.
Talib WH (2018) Melatonin and Cancer Hallmarks. Molecules 23 (3): 518. doi:10.3390/molecules23030518.
Dantzer R, Meagher MW, Cleeland CS (2012) Translational approaches to treatment-induced symptoms in cancer patients. Nat. Rev. Clin. Oncol. 9 (7): 414–426. doi:10.1038/nrclinonc.2012.88.
Love RR, Leventhal H, Easterling DV, Nerenz DR (1989) Side effects and emotional distress during cancer chemotherapy. Cancer 63 (3): 604–612. doi:10.1002/1097-0142(19890201)63:3<604::aid-cncr2820630334>3.0.co;2-2.
Cleeland C, Allen JD, Roberts SA, Brell JM, Giralt SA, Khakoo AY, Kirch RA, Kwitkowski VE, Liao Z, Skillings J (2012) Reducing the toxicity of cancer therapy: recognizing needs, taking action. Nat. Rev. Clin. Oncol. 9 (8): 471–478. doi:10.1038/nrclinonc.2012.99.
Pich O, Muiños F, Lolkema MP, Steeghs N, Gonzalez-Perez A, Lopez-Bigas N (2019) The mutational footprints of cancer therapies. Na.t Genet. 51 (12): 1732–1740. doi:10.1038/s41588-019-0525-5.
Slominski AT, HardelandR, Zmijewski MA, Slominski RM, Reiter RJ, Paus R (2018) Melatonin: A Cutaneous Perspective on its Production, Metabolism, and Functions. J. Invest. Dermatol. 138 (3): 490–499. doi:10.1016/j.jid.2017.10.025.
Arendt J. (2006). Melatonin and human rhythms. Chronobiol. Int. 23 (1-2): 21–37. doi:10.1080/07420520500464361.
Reiter RJ (1991) Melatonin: the chemical expression of darkness. Mo.l Cell Endocrinol.79 (1-3): C153–C158. doi:10.1016/0303-7207(91)90087-9.
Srinivasan V, Spence DW, Pandi-Perumal SR, TrakhtI,Cardinali D P (2008) Therapeutic actions of melatonin in cancer: possible mechanisms. Integr. Cancer Ther. 7 (3): 189–203.doi:10.1177/1534735408322846.
Ma Z, Xu L, Liu D, Zhang X, Di S, Li W, Zhang J, Reiter RJ, Han J, Li X, Yan X (2020) Utilizing Melatonin to Alleviate Side Effects of Chemotherapy: A Potentially Good Partner for Treating Cancer with Ageing. Oxid Med Cell Longev. 2020: 6841581. doi:10.1155/2020/6841581.
Sanchez-Barcelo EJ, Mediavilla MD, Alonso-Gonzalez C, Reiter RJ (2012) Melatonin uses in oncology: breast cancer prevention and reduction of the side effects of chemotherapy and radiation. Expert.OpinInvestig. Drugs 21 (6): 819–831. doi:10.1517/13543784.2012.681045.
Blackadar CB (2016) Historical review of the causes of cancer. World J. Clin. Oncol. 7 (1): 54–86.doi:10.5306/wjco.v7.i1.54.
Yamagiwa K, Ichikawa K (1977) Experimental study of the pathogenesis of carcinoma. CA Cancer J. Clin. 27 (3): 174–181. doi:10.3322/canjclin.27.3.174.
Kiyohara C, Otsu A, Shirakawa T, Fukuda S, Hopkin JM (2002) Genetic polymorphisms and lung cancer susceptibility: a review. Lung Cancer 37 (3): 241–256. doi:10.1016/s0169-5002(02)00107-1.
Tsuchiya Y, Sato T, Kiyohara C, Yoshida K, Ogoshi K, Nakamura K, Yamamoto M (2002) Genetic polymorphisms of cytochrome P450 1A1 and risk of gallbladder cancer. J. Exp. Clin. Cancer Res. 21 (1): 119–124. PMID: 12071517.
Zhang YJ (2010) Interactions of chemical carcinogens and genetic variation in hepatocellular carcinoma. World J. Hepatol. 2 (3): 94–102. doi:10.4254/wjh.v2.i3.94.
Peters JM, Gonzalez FJ (2018) The Evolution of Carcinogenesis. Toxicol. Sci. 165(2): 272–276. doi:10.1093/toxsci/kfy184.
Louten J (2016) Viruses and Cancer. Essential Human Virology. Elsevier pp 155-170. doi:10.1016/B978-0-12-800947-5.00009-0.
Smith MT, Guyton KZ, Gibbons CF, Fritz JM, Portier CJ, Rusyn I, DeMarini DM, Caldwell JC, Kavlock RJ, Lambert PF, Hecht SS, Bucher JR, Stewart BW, Baan RA, Cogliano VJ, Straif K (2016) Key Characteristics of Carcinogens as a Basis for Organizing Data on Mechanisms of Carcinogenesis. Environ. Health Perspect. 124 (6): 713–721. doi:10.1289/ehp.1509912.
Chakarov S, Petkova R, RussevGC, Zhelev N (2014) DNA damage and mutation. Types of DNA damage. BioDiscovery 11: e8957. DOI: 10.7750/BioDiscovery.2014.11.1.
Barnes JL, Zubair M, John K, Poirier MC, Martin FL (2018) Carcinogens and DNA damage. Biochem. Soc. Trans 46 (5): 1213–1224. doi:10.1042/BST20180519.
Naito A (2010) Nongenotoxic carcinogenesis. Comprehensive Toxicology (3rd edition). Elsevier. pp 35-48. doi: 10.1016/B978-0-08-046884-6.01403-2.
Birkett N, Al-Zoughool M, Bird M, Baan RA, Zielinski J, Krewski D (2019) Overview of biological mechanisms of human carcinogens. J. Toxicol. Environ. Health B Crit. Rev. 22 (7-8): 288–359. doi:10.1080/10937404.2019.1643539.
Bus JS (2017) IARC use of oxidative stress as key mode of action characteristic for facilitating cancer classification: Glyphosate case example illustrating a lack of robustness in interpretative implementation. Regul. Toxicol. Pharmacol. 86: 157–166. doi:10.1016/j.yrtph.2017.03.004.
TroskoJE(2017) Reflections on the use of 10 IARC carcinogenic characteristics for an objective approach to identifying and organizing results from certain mechanistic studies. Toxicol. Res. Appl. 1:239784731771083. doi: 10.1177/2397847317710837.
Smith CJ, Perfetti TA, Hayes AW (2021) Categorizing the characteristics of human carcinogens: a need for specificity. Arch.Toxicol. 95: 2883–2889. doi:10.1007/s00204-021-03109-w.
Thomas RD (1986). Drinking Water and Health: Volume 6. Washington (DC): National Academies Press (US); 5, Mechanisms of Carcinogenesis. National Research Council (US) Safe Drinking Water Committee. Available from: https://www.ncbi.nlm.nih.gov/books/NBK219109/. DOI: 10.17226/921.
Fieser LF (1938) Carcinogenic activity, structure and chemical reactivity of polynuclear hydrocarbons. Am. J. Cancer Res. 34: 37−124. doi: 10.1158/ajc.1938.37.
Hill J (1761) Cautions against the immoderate use of snuff: founded on the known qualities of the tobacco plant; and the effects it must produce when this way taken into the body: and enforced by instances of persons who have perished miserably of diseases occasioned or rendered incurable by its use. by dr. j. hill (The 2nd). Printed for R. Baldwin in Pater-noster Row and J. Jackson in St. James's-Street. http://0find.galegroup.com.biblio.eui.eu/ecco/infomark.do?contentSet=ECCOArticles&docType=ECCOArticles&bookId=1329700300&type=getFullCitation&tabID=T001&prodId=ECCO&docLevel=TEXT_GRAPHICS&version=1.0&source=library&userGroupName=europeo.
Miller E C, Miller JA (1947) The presence and significance of bound amino azodyes in the livers of rats fed pdimethylaminoazobenzene. Cancer Res. 7: 468−480. Corpus ID: 44390356.
MILLER EC (1951) Studies on the formation of protein-bound derivatives of 3,4-benzpyrene in the epidermal fraction of mouse skin. Cancer Res. 11 (2): 100–108. PMID: 14812434.
Miller JA (1970) Carcinogenesis by chemicals: an overview--G. H. A. Clowes memorial lecture. Cancer Res. 30 (3): 559–576. PMID: 4915745.
Omura T, Sato R (1964) The carbon monoxide-binding pigment of liver microsomes. i. evidence for its hemoprotein nature. J. Biol. Chem. 239: 2370–2378. doi: 10.1016/S0021-9258(20)82244-3.
Omiecinski CJ, Vanden Heuvel JP, Perdew GH, Peters JM (2011) Xenobiotic metabolism, disposition, and regulation by receptors: from biochemical phenomenon to predictors of major toxicities. Toxicol. Sci. 120 (Suppl 1): S49–S75. doi:10.1093/toxsci/kfq338.
Klaunig JE (2020). Carcinogenesis. An introduction to interdisciplinary toxicology. Academic press. pp 97-110. doi: 10.1016/B978-0-12-813602-7.00008-9.
Klaunig JE, Kamendulis LM (2010) Carcinogenicity. Comprehensive Toxicology (Second Edition). Elsevier. pp 117-138. doi: 10.1016/B978-0-08-046884-6.00315-8.
Ravanat JL, Douki T (2016) UV and ionizing radiations induced DNA damage, differences and similarities. Radiat. Phys. Chem. 128: 92-102. doi: 10.1016/j.radphyschem.2016.07.007.
CohenSM, Arnold LL (2011) Chemical carcinogenesis. Toxicol. Sci. 120 (Suppl 1): S76–S92. doi:10.1093/toxsci/kfq365.
Hebels DG, Briedé JJ, Khampang R, Kleinjans JC, de Kok TM (2010) Radical mechanisms in nitrosamine- and nitrosamide-induced whole-genome gene expression modulations in Caco-2 cells. Toxicol. Sci. 116 (1): 194–205. doi:10.1093/toxsci/kfq121.
Kondo N, Takahashi A, Ono K, Ohnishi T (2010) DNA damage induced by alkylating agents and repair pathways. J. Nucleic Acids 2010: 543531. doi:10.4061/2010/543531.
Wohak LE, Krais AM, Kucab JE, Stertmann J, Øvrebø S, Seidel A, Phillips DH, Arlt VM (2016) Carcinogenic polycyclic aromatic hydrocarbons induce CYP1A1 in human cells via a p53-dependent mechanism. Arch.Toxicol. 90 (2): 291–304. doi:10.1007/s00204-014-1409-1.
Sridhar J, Goyal N, Liu J, Foroozesh M (2017) Review of ligand specificity factors for CYP1A subfamily enzymes from molecular modeling studies reported to-date. Molecules 22 (7): 1143. doi:10.3390/molecules22071143.
Guengerich FP (2000) Metabolism of chemical carcinogens. Carcinogenesis 21 (3): 345-351. doi:10.1093/carcin/21.3.345.
Conney AH (1982) Induction of microsomal enzymes by foreign chemicals and carcinogenesis by polycyclic aromatic hydrocarbons: G. H. A. Clowes Memorial Lecture. Cancer Res. 42 (12): 4875–4917. doi: 0008-5472/82/0042-OOOO.
Miller EC, Miller JA (1981) Searches for ultimate chemical carcinogens and their reactions with cellular macromolecules. Cancer 47 (10): 2327–2345. doi:10.1002/1097-0142(19810515)47:10<2327::aid-cncr2820471003>3.0.co;2-z.
Melnick RL, Kohn MC, Portier CJ (1996) Implications for risk assessment of suggested nongenotoxic mechanisms of chemical carcinogenesis. Environ. Health Perspect 104 (Suppl 1): 123–134. doi:10.1289/ehp.96104s1123.
Williams GM (2001) Mechanisms of chemical carcinogenesis and application to human cancer risk assessment. Toxicology 166 (1-2): 3–10. doi:10.1016/s0300-483x(01)00442-5.
Hernández LG, van Steeg H, Luijten M, van Benthem J (2009) Mechanisms of non-genotoxic carcinogens and importance of a weight of evidence approach. Mutat. Res. 682 (2-3): 94–109. doi:10.1016/j.mrrev.2009.07.002.
Tao L, Wang X, Zhou Q (2020) Long noncoding RNA SNHG16 promotes the tumorigenicity of cervical cancer cells by recruiting transcriptional factor SPI1 to upregulate PARP9. Cell Biol. Int. 44 (3): 773–784. doi:10.1002/cbin.11272.
Nenclares P, Harrington KJ (2020) The biology of cancer. Medicine 48 (2): 67-72. doi: 10.1016/j.mpmed.2019.11.001.
Capen CC, DayanAD, Green S (1995) Receptor-mediated mechanisms in carcinogenesis: an overview. Mutat. Res. 333 (1-2):215-224. doi:10.1016/0027-5107(95)00148-4.
McClain MR(1993) Mechanistic considerations for the relevance of animal data on thyroid neoplasia to human risk assessment. Mutat. Res. 333 (1-2): 131-142. doi:10.1016/0027-5107(95)00139-5.
Meegan MJ, O'Boyle NM (2019) Special Issue "Anticancer Drugs". Pharmaceuticals (Basel)12 (3): 134. doi:10.3390/ph12030134.
Sikes RA (2007) Chemistry and pharmacology of anticancer drugs. Br. J. Cancer 97 (12): 1713. doi: 10.1038/sj.bjc.6604075.
Shewach DS, Kuchta RD (2009) Introduction to cancer chemotherapeutics. Chem. Rev. 109 (7): 2859–2861. doi:10.1021/cr900208x.
Nussbaumer S, Bonnabry P, Veuthey JL, Fleury-Souverain S (2011) Analysis of anticancer drugs: a review. Talanta 85 (5): 2265–2289. doi:10.1016/j.talanta.2011.08.034.
Galluzzi L, Buqué A, Kepp O, Zitvogel L, Kroemer G. (2015) Immunological effects of conventional chemotherapy and targeted anticancer agents. Cancer cell 28 (6): 690–714. doi:10.1016/j.ccell.2015.10.012.
Thurston DE (2006) “Chemistry and Pharmacology of Anticancer Drugs”. CRC Press. Oxford. doi: 10.1201/9781420008906.
Amjad MT, Chidharla A, Kasi A (2022) Cancer Chemotherapy. In: StatPearls. Treasure Island (FL): StatPearls Publishing. PMID: 33232037.
Smith IE, Evans BD (1985) Carboplatin (JM8) as a single agent and in combination in the treatment of small cell lung cancer. Cancer Treat. Rev. 12 (Suppl A): 73–75. doi:10.1016/0305-7372(85)90021-0.
Extra JM, Espie M, Calvo F, Ferme C, Mignot L, Marty M (1990) Phase I study of oxaliplatin in patients with advanced cancer. Cancer Chemother. Pharmacol. 25 (4): 299–303. doi:10.1007/BF00684890.
Rosenberg B, VanCamp L, Trosko JE, Mansour VH (1969) Platinum compounds: a new class of potent antitumour agents. Nature 222 (5191): 385–386. doi:10.1038/222385a0.
Garbutcheon-Singh KB, Leverett P, Myers S, Aldrich-Wrigh JR (2013) Cytotoxic platinum(II) intercalators that incorporate 1R,2Rdiaminocyclopentane. Dalton Trans 42: 918-926. doi:10.1039/c2dt31323e.
Lowenthal RM, Eaton K (1996) Toxicity of chemotherapy. Hematol. Oncol. Clin. North Am. 10 (4): 967–990. doi:10.1016/s0889-8588(05)70378-6.
Redmond KM, Wilson TR, Johnston PG, Longley DB (2008) Resistance mechanisms to cancer chemotherapy. Front. Biosci. 13: 5138–5154. doi:10.2741/3070.
Remesh A (2017) Toxicities of anticancer drugs and its management. Int. J. Basic Clin. Pharmacol. 1 (1): 2-12 .doi: 10.5455/2319-2003.ijbcp000812.
George M. Brenner, Craig W. Stevens. (2010). Antineoplastic drugs, Text book of Pharmacology. Saunders Elsevier. 493-511. Paperback ISBN: 9780323391665.
Hoagland HC (1982) Hematologic complications of cancer chemotherapy. Semin. Oncol. 9 (1): 95–102. doi: 10.5555/URI:PII:0093775482900112.
Gupta S, Tannous R, Friedman M (2001) Incidence of anaemia in CHOP-treated intermediate- grade nonHodgkin’s lymphoma (IGNHL). Eur. J. Cancer. 37: S94:339.10.1016/S0959-8049(01)80831-5.
Liu M, Tan H, Zhang X, Liu Z, Cheng Y, Wang D, Wang F (2014) Hematopoietic effects and mechanisms of Fufang e׳jiao jiang on radiotherapy and chemotherapy-induced myelosuppressed mice. J. Ethnopharmacol. 152 (3): 575–584. doi:10.1016/j.jep.2014.02.012.
Cardinale D, Ciceri F, Latini R (2018) Anthracycline-induced cardiotoxicity: A multicenter randomised trial comparing two strategies for guiding prevention with enalapril: The International CardioOncology Society-one trial. Eur. J. Cancer 94: 126-137. doi:10.1016/j.ejca.2018.02.005.
Adão R, de Keulenaer G, Leite-Moreira A, &Brás-Silva (2013). Cardiotoxicity associated with cancer therapy: pathophysiology and prevention strategies. Rev. Port. Cardiol. 32 (5): 395–409. doi:10.1016/j.repc.2012.11.002.
Curigliano G, Cardinale D, Dent S, Criscitiello C, Aseyev O, Lenihan D, Cipolla CM (2016) Cardiotoxicity of anticancer treatments: Epidemiology, detection, and management. CA. Cancer J. Clin. 66 (4): 309–325. doi:10.3322/caac.21341.
Rowinsky EK, McGuire WP, Guarnieri T, Fisherman JS, Christian MC, Donehower RC (1991) Cardiac disturbances during the administration of taxol. J. Clin. Oncol. 9 (9): 1704–1712. doi:10.1200/JCO.1991.9.9.1704.
Al-Majed AA, Sayed-Ahmed MM, Al-Yahya AA, Aleisa AM, Al-Rejaie SS, Al-Shabanah OA (2006) Propionyl-L-carnitine prevents the progression of cisplatin-induced cardiomyopathy in a carnitine-depleted rat model. Pharmacol. Res. 53 (3): 278–286. doi:10.1016/j.phrs.2005.12.005.
Lee CS, Ryan, EJ, Doherty GA (2014) Gastro-intestinal toxicity of chemotherapeutics in colorectal cancer: the role of inflammation. World J. Gastroenterol. 20 (14): 3751–3761. doi:10.3748/wjg.v20.i14.3751.
Xue H, Sawyer MB, WischmeyerPE,Baracos VE (2011) Nutrition modulation of gastrointestinal toxicity related to cancer chemotherapy: from preclinical findings to clinical strategy. PEN J. Parenter Enteral. Nutr. 35 (1): 74–90. doi:10.1177/0148607110377338.
Benson AB 3rd, Ajani JA, Catalano RB, Engelking C, Kornblau SM, Martenson JA, Jr McCallum R, Mitchell E, O'Dorisio TM, Vokes EE,Wadler S (2004) Recommended guidelines for the treatment of cancer treatment-induced diarrhea. J. Clin. Oncol. 22 (14): 2918–2926. doi:10.1200/JCO.2004.04.132.
Denlinger CS, Barsevick AM (2009) The challenges of colorectal cancer survivorship. J. Natl. Compr. Canc. Netw. 7 (8): 883–894. doi:10.6004/jnccn.2009.0058.
PetersonDE, Boers-Doets CB, Bensadoun RJ, Herrstedt J, ESMO Guidelines Committee (2015) Management of oral and gastrointestinal mucosal injury: ESMO Clinical Practice Guidelines for diagnosis, treatment, and follow-up. Ann. Oncol. 26 (Suppl 5): v139–v151. doi:10.1093/annonc/mdv202.
Sonis ST, Elting LS, Keefe D, Peterson DE, Schubert M, Hauer-Jensen M, Bekele BN, Raber-Durlacher J, Donnelly JP, Rubenstein EB, Mucositis Study Section of the Multinational Association for Supportive Care in Cancer, & International Society for Oral Oncology (2004). Perspectives on cancer therapy-induced mucosal injury: pathogenesis, measurement, epidemiology, and consequences for patients. Cancer 100 (9 Suppl): 1995–2025. doi:10.1002/cncr.20162.
Duncan M, Grant G (2003) Oral and intestinal mucositis - causes and possible treatments. Aliment Pharmacol. Ther. 18 (9): 853–874. doi:10.1046/j.1365-2036.2003.01784.x.
McQuade RM, Al Thaalibi M, Nurgali K (2020). Impact of chemotherapy-induced enteric nervous system toxicity on gastrointestinal mucositis. Curr. Opin. Support Palliat. Care 14 (3): 293–300. doi:10.1097/SPC.0000000000000515.
Blijham GH (1993) Prevention and treatment of organ toxicity during high-dose chemotherapy: an overview. Anticancer Drugs 4 (5):527-533. doi:10.1097/00001813-199310000-00001.
Han Y, Smith MT (2013) Pathobiology of cancer chemotherapy-induced peripheral neuropathy (CIPN). Front. Pharmacol. 4: 156. doi:10.3389/fphar.2013.00156.
Dietrich J (2020) Neurotoxicity of cancer therapies. Continuum (Minneap Minn) 26 (6): 1646–1672. doi:10.1212/CON.0000000000000943.
Bompaire F, Durand T, Léger-Hardy I, Psimaras D, Ricard D (2017) Chemotherapy-related cognitive impairment or « chemobrain »: concept and state of art. Troubles cognitifschimio-induitsou « chemobrain»: concept et état de l’art. Geriatr Psychol.NeuropsychiatrVieil 15 (1): 89–98. doi:10.1684/pnv.2017.0659.
Branca JJV, Morucci G, Pacini A (2018) Cadmium-induced neurotoxicity: still much ado. Neural Regen. Res. 13 (11): 1879–1882. doi:10.4103/1673-5374.239434.
Basak D, Arrighi S, Darwiche Y, Deb S (2021) Comparison of anticancer drug toxicities: paradigm shift in adverse effect profile. Life (Basel) 12 (1): 48. doi:10.3390/life12010048.
Townsend DM, Deng M, Zhang L, Lapus MG, Hanigan MH (2003) Metabolism of Cisplatin to a nephrotoxin in proximal tubule cells. J. Am. Soc. Nephrol. 14 (1): 1–10. doi:10.1097/01.asn.0000042803.28024.92.
Sahni V, Choudhury D Ahmed,Z. (2009) Chemotherapy-associated renal dysfunction. Nat. Rev. Nephrol. 5 (8): 450–462. doi:10.1038/nrneph.2009.97.
Mudd TW, Guddati A. (2021) Management of hepatotoxicity of chemotherapy and targeted agents. Am. J. Cancer Res. 11 (7): 3461–3474. PMID: 34354855 PMCID: PMC8332851.
Han JM, Yee J, Cho S, Gwak HS (2020) Factors influencing imatinib-induced hepatotoxicity. Cancer Res. Treat. 52 (1): 181–188. doi:10.4143/crt.2019.131.
LERNER AB, CASE JD, TAKAHASHI Y (1960) Isolation of melatonin and 5-methoxyindole-3-acetic acid from bovine pineal glands. J. Biol. Chem. 235: 1992–1997. doi: 10.1016/s0021-9258(18)69351-2.
1Tordjman S, Chokron S, Delorme R, Charrier A, Bellissant E, Jaafari N, Fougerou C (2017) Melatonin: Pharmacology, functions and therapeutic benefits. Curr. Neuropharmacol. 15 (3): 434–443. doi:10.2174/1570159X14666161228122115.
Favero G, Moretti,E, Bonomini F, Reiter RJ, Rodella LF, Rezzani R. (2018) Promising antineoplastic actions of melatonin. Front. Pharmacol. 9: 1086. doi:10.3389/fphar.2018.01086.
Reppert SM, Godson C, Mahle CD, Weaver DR, Slaugenhaupt SA, Gusella JF (1995) Molecular characterization of a second melatonin receptor expressed in human retina and brain: the Mel1b melatonin receptor. Proc. Natl. Acad. Sci. USA. 92 (19): 8734–8738. doi:10.1073/pnas.92.19.8734.
Slominski RM, Reiter RJ, Schlabritz-Loutsevitch N, Ostrom RS, Slominski AT (2012) Melatonin membrane receptors in peripheral tissues: distribution and functions. Mol. Cell Endocrinol. 351 (2): 152–166. doi:10.1016/j.mce.2012.01.004.
Reiter RJ, Tan DX, Galano A (2014) Melatonin: exceeding expectations. Physiology (Bethesda) 29 (5): 325–333. doi:10.1152/physiol.00011.2014.
Dubocovich ML, Markowska M (2005) Functional MT1 and MT2 melatonin receptors in mammals. Endocrine 27 (2): 101–110. doi:10.1385/ENDO:27:2:101.
Reiter RJ, Mayo J, Tan, X., Sainz RM, Alatorre-Jimenez M, Qin L (2016) Melatonin as an antioxidant: under promises but over delivers. J. Pineal Res. 61 (3): 253–278. doi:10.1111/jpi.12360.
Bonmati-Carrion MA, Tomas-Loba A (2021) Melatonin and cancer: A polyhedral network where the source matters.Antioxidants (Basel). 10 (2): 210. doi:10.3390/antiox10020210.
Halladin N, Busch, SE, Jensen SE, Hansen HS, Zaremba T, Aarøe J, Rosenberg ,&Gögenur I (2014) Intracoronary and systemic melatonin to patients with acute myocardial infarction: protocol for the IMPACT trial. Dan. Med. J. 61 (2): A4773. doi: 10.1007/s00380-014-0589-1.
Karbownik M, Lewinski A, Reiter RJ (2001) Anticarcinogenic actions of melatonin which involve antioxidative processes: comparison with other antioxidants. Int. J. Biochem. Cell Biol. 33 (8): 735–753. doi:10.1016/s1357-2725(01)00059-0.
Galano A, Tan DX, Reiter RJ (2018) Melatonin: A Versatile Protector against Oxidative DNA Damage. Molecules 23 (3): 530. doi:10.3390/molecules23030530.
Guo Y, Su J., Li T, Zhang Q, Bu, Wang Q, Lai D (2017) Melatonin ameliorates restraint stress-induced oxidative stress and apoptosis in testicular cells via NF-κB/iNOS and Nrf2/ HO-1 signaling pathway. Sci. Rep. 7 (1): 9599. doi:10.1038/s41598-017-09943-2.
Zhang HM, Zhang (2014). Melatonin: a well-documented antioxidant with conditional pro-oxidant actions. J. Pineal Res. 57 (2): 131–146. doi:10.1111/jpi.12162.
Proietti , Cucina, A, Reiter RJ, Bizzarri M (2013) Molecular mechanisms of melatonin's inhibitory actions on breast cancers. Cell. Mol. Life Sci. 70 (12): 2139–2157. doi:10.1007/s00018-012-1161-8.
Cos S, Recio J Sánchez-Barceló EJ (1996). Modulation of the length of the cell cycle time of MCF-7 human breast cancer cells by melatonin. Life Sci. 58 (9): 811–816. doi:10.1016/0024-3205(95)02359-3.
Nooshinfar E, Bashash D, Safaroghli-Azar A, Bayati S, Rezaei-Tavirani M, Ghaffari S H, Akbari ME (2016) Melatonin promotes ATO-induced apoptosis in MCF-7 cells: Proposing novel therapeutic potential for breast cancer. Biomed. Pharmacother. 83: 456–465. doi:10.1016/j.biopha.2016.07.004.
Shen CJ, Chang CC, Chen YT, Lai CS, Hsu YC (2016). Melatonin suppresses the growth of ovarian cancer cell lines (OVCAR-429 and PA-1) and potentiates the effect of g1 arrest by targeting CDKs. Int. J. Mol. Sci. 17 (2): 176. doi:10.3390/ijms17020176.
Gonçalves NdoN, Colombo J, Lopes JR, Gelaleti GB, Moschetta MG, Sonehara NM, Hellmén E, ZanonCdeF, Oliani SM, Zuccari DA (2016) Effect of melatonin in epithelial mesenchymal transition markers and invasive properties of breast cancer stem cells of canine and human cell lines. PloS one 11 (3): e0150407. doi:10.1371/journal.pone.0150407.
Mao L, SummersW, Xiang S, Yuan L, Dauchy RT, Reynolds A, Wren-Dail MA, Pointer D, Frasch T, Blask DE, Hill SM (2016) Melatonin represses metastasis in her2-postive human breast cancer cells by suppressing RSK2 expression. Mol. Cancer Res. 14 (11): 1159–1169. doi:10.1158/1541-7786.MCR-16-0158.
Spigel DR, Burstein HJ (2002) HER2 overexpressing metastatic breast cancer. Curr. Treat. Options Oncol. 3 (2): 163–174. doi:10.1007/s11864-002-0062-8.
Mao L, Yuan L, Slakey LM, Jones FE, Burow ME, Hill SM (2010). Inhibition of breast cancer cell invasion by melatonin is mediated through regulation of the p38 mitogen-activated protein kinase signaling pathway. Breast Cancer Res. 12 (6): R107. doi:10.1186/bcr2794.
Seely D, Wu P, Frit H., Kennedy DA, Tsui T, Seely AJ, Mills E (2012) Melatonin as adjuvant cancer care with and without chemotherapy: a systematic review and meta-analysis of randomized trials. Integr. Cancer Ther. 11 (4): 293–303. doi:10.1177/1534735411425484.
Chovancova B, Hudecova S, Lencesova L, Babula P, Rezuchova I, Penesova A, Grman M, MoravcikR, Zeman M, Krizanova O (2017) Melatonin-induced changes in cytosolic calcium might be responsible for apoptosis induction in tumour cells. Cell Physiol. Biochem. 44 (2): 763–777. doi:10.1159/000485290.
Fulda S (2018 Therapeutic opportunities based on caspase modulation. Semin. Cell Dev. Biol. 82: 150–157. doi:10.1016/j.semcdb.2017.12.008.
Chuffa LGA, Reiter R, Lupi LA (2017) Melatonin as a promising agent to treat ovarian cancer: molecular mechanisms. Carcinogenesis 38 (10): 945–952. doi:10.1093/carcin/bgx054.
Nishida N, Yano H, Nishida T, Kamura T, Kojir M (2006). Angiogenesis in cancer. Vasc. Health Risk Manag. 2 (3): 213–219. doi:10.2147/vhrm.2006.2.3.213.
HicklinDJ, Ellis LM (2005) Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. J. Clin. Oncol. 23 (5): 1011–1027. doi:10.1200/JCO.2005.06.081.
Pradeep CR, Sunila ES, Kuttan G (2005) Expression of vascular endothelial growth factor (VEGF) and VEGF receptors in tumor angiogenesis and malignancies. Integr. Cancer Ther. 4 (4): 315–321. doi:10.1177/1534735405282557.
Shinkaruk S, Bayle M, Lain, Deleris G (2003) Vascular endothelial cell growth factor (VEGF), an emerging target for cancer chemotherapy. Curr. Med. Chem. Anticancer Agents 3 (2): 95–117. doi:10.2174/1568011033353452.
Moreira IS, Fernandes PA, Ramos MJ (2007) Vascular endothelial growth factor (VEGF) inhibition--a critical review. Anticancer Agents Med. Chem. 7 (2): 223–245.doi:10.2174/187152007780058687.
Colombo J, Maciel JM, Ferreira LC, Silva RFDA, Zuccari DA (2016) Effects of melatonin on HIF-1α and VEGF expression and on the invasive properties of hepatocarcinoma cells. Oncol. Lett. 12 (1): 231–237. doi:10.3892/ol.2016.4605.
Park JW, Hwang MS, Suh SI, Baek WK (2009) Melatonin down-regulates HIF-1 alpha expression through inhibition of protein translation in prostate cancer cells. J. Pineal Res. 46 (4): 415–421. doi:10.1111/j.1600-079X.2009.00678.x.
Soybir G, Topuzlu C, Odabaş O, Dolay K, Bili A, Kökso F (2003) The effects of melatonin on angiogenesis and wound healing. Surg. Today 33 (12): 896–901. doi:10.1007/s00595-003-2621-3.
Labrecque N, Cermakian N (2015) Circadian clocks in the immune system. J. Biol. Rhythms. 30 (4): 277–290. doi:10.1177/0748730415577723.
Ozkanlar , Kara A, Sengul E, Simsek N, Karadeniz A, Kurt N (2016) Melatonin modulates the immune system response and inflammation in diabetic rats experimentally-induced by alloxan. Horm. Metab. Res. 48 (2): 137–144. doi:10.1055/s-0035-1548937.
Vinther AG, Claesson MH (2015) The influence of melatonin on immune system and cancer. Int. J. Cancer Clin. Res. 2: 024. doi: 10.23937/2378-3419/2/4/1024.
Ren W, Liu G, Chen S, Yin J, Wang J, Tan B, Wu G, Bazer FW, PengY, Li T, Reiter RJ. Yin Y (2017) Melatonin signaling in T cells: Functions and applications. J. Pineal Res. 62 (3): 10.1111/jpi.12394. doi:10.1111/jpi.12394.
Carpentieri AR, Peralta Lopez ME, Aguilar J, Solá VM (2017) Melatonin and periodontal tissues: Molecular and clinical perspectives. Pharmacol. Res. 125 (Pt B): 224–231. doi:10.1016/j.phrs.2017.09.003.
Singh M, Jadhav HR (2014) Melatonin: functions and ligands. Drug Discov. Today 19 (9): 1410–1418. doi:10.1016/j.drudis.2014.04.014.
Carrillo-Vico A, Lardone P, Alvarez-Sánchez N, Rodríguez-Rodríguez A, Guerrero JM (2013) Melatonin: buffering the immune system. Int. J. Mol. Sci. 14 (4): 8638–8683. doi:10.3390/ijms14048638.
García-Mauriño S, Pozo D, Carrillo-Vico A, Calvo JR, Guerrero JM (1999) Melatonin activates Th1 lymphocytes by increasing IL-12 production. Life Sci. 65 (20): 2143–2150. doi:10.1016/s0024-3205(99)00479-8.
Miller SC, Pandi-Perumal SR, Esquifino AI, Cardinali DP, Maestroni GJ (2006) The role of melatonin in immuno-enhancement: potential application in cancer. Int. J. Exp. Pathol. 87 (2): 81–87. doi:10.1111/j.0959-9673.2006.00474.x.
Liu H, Xu L, We JE, Xie MR, Wang SE, Zhou RX (2011) Role of CD4+ CD25+ regulatory T cells in melatonin-mediated inhibition of murine gastric cancer cell growth in vivo and in vitro. Anat. Rec. (Hoboken) 294 (5): 781–788. doi:10.1002/ar.21361.
Bouris P, Skandalis SS, Piperigko Z, Afratis N, Karamanou K, Aletras AJ, Moustakas A, Theocharis AD, Karamanos NK (2015) Estrogen receptor alpha mediates epithelial to mesenchymal transition, expression of specific matrix effectors and functional properties of breast cancer cells. Matrix. Biol. 43: 42–60. doi:10.1016/j.matbio.2015.02.008.
Huang B, Warner M, Gustafsson JA (2015) Estrogen receptors in breast carcinogenesis and endocrine therapy. Mol. Cell Endocrinol. 418 (Pt 3): 240-244. doi:10.1016/j.mce.2014.11.015.
Santen RJ, Yue W, Wang JP (2015) Estrogen metabolites and breast cancer. Steroids 99 (Pt A): 61–66. doi:10.1016/j.steroids.2014.08.003.
Soysal SD, Kilic IB, Regenbrecht CR, Schneider S, Muenst S, Kilic N, Güth U, Dietel M, Terracciano LM,Kilic E (2015) Status of estrogen receptor 1 (ESR1) gene in mastopathy predicts subsequent development of breast cancer. Breast Cancer Res. Treat. 151 (3): 709–715. doi:10.1007/s10549-015-3427-y.
Lopes J, Arnosti D, Trosko JE, Tai MH, Zuccari D (2016) Melatonin decreases estrogen receptor binding to estrogen response elements sites on the OCT4 gene in human breast cancer stem cells. Genes Cancer 7 (5-6): 209–217. doi:10.18632/genesandcancer.107.
Sánchez-Barceló EJ, Cos S, Mediavilla D, Martínez-Campa C, González A, Alonso-González C (2005) Melatonin-estrogen interactions in breast cancer. J. Pineal Res. 38 (4): 217–222. doi:10.1111/j.1600-079X.2004.00207.x.
Hill S M, Belancio VP, Dauchy RT, Xiang S, Brimer S, Mao L, Hauch A, Lundberg PW, Summers W, Yuan L, Frasch T, Blask D (2015) Melatonin: an inhibitor of breast cancer. Endocr. Relat. Cancer 22 (3): R183–R204. doi:10.1530/ERC-15-0030.
Monayo SM, Liu X (2022) The Prospective Application of Melatonin in Treating Epigenetic Dysfunctional Diseases. Front. Pharmacol. 13: 867500. doi:10.3389/fphar.2022.867500.
Saha S, Buttari B, Panieri E, Profumo E, &Saso L (2020) An overview of Nrf2 signaling pathway and its role in inflammation. Molecules 25 (22): 5474. doi:10.3390/molecules25225474.
Yi C, Zhang Y, Yu Z, Xiao Y, Wang J, Qiu H, Yu W, Tang R, Yuan Y, Guo W, Deng W (2014) Melatonin enhances the anti-tumor effect of fisetin by inhibiting COX-2/iNOS and NF-κB/p300 signaling pathways. PloS one 9 (7): e99943. doi:10.1371/journal.pone.0099943.
Shrestha S, Zhu J, Wang Q, Du X, Liu F, Jiang J, Song J, Xing J, Sun D, Hou Q, Peng Y, Zhao J, Sun X, Song X (2017) Melatonin potentiates the antitumor effect of curcumin by inhibiting IKKβ/NF-κB/COX-2 signaling pathway. Int. J. Oncol. 51 (4): 1249–1260. doi:10.3892/ijo.2017.4097.
Qin W, Lu W, Li H, Yuan X, Li B, Zhang Q, Xiu R (2012) Melatonin inhibits IL1β-induced MMP9 expression and activity in human umbilical vein endothelial cells by suppressing NF-κB activation. J. Endocrinol. 214 (2): 145–153. doi:10.1530/JOE-12-0147.
Lissoni P, Tancini G, Barni S, Paolorossi F, Ardizzoia A, Conti A, Maestroni G (1997) Treatment of cancer chemotherapy-induced toxicity with the pineal hormone melatonin. Support Care Cancer 5 (2): 126–129. doi:10.1007/BF01262569.
Regelson W, Pierpaoli W (1987) Melatonin: a rediscovered antitumor hormone? Its relation to surface receptors; sex steroid metabolism, immunologic response, and chronobiologic factors in tumor growth and therapy. Cancer Invest. 5 (4): 379–385. doi: 10.1080/07357908709170112.
Bilginoğlu A, Aydın D, Ozsoy S, Aygün H (2014) Protective effect of melatonin on adriamycin-induced cardiotoxicity in rats. Turk Kardiyol Dern Ars. 42 (3): 265–273. doi:10.5543/tkda.2014.36089.
Dziegiel P, Surowiak P, Rabczyński J, Zabel M (2002) Effect of melatonin on cytotoxic effects of daunorubicin on myocardium and on transplantable Morris hepatoma in rats. Pol. J. Pathol. 53 (4): 201–204. PMID: 12597337.
Guven A, Yavuz O, Cam M, Ercan F, Bukan N, Comunoglu C (2007) Melatonin protects against epirubicin-induced cardiotoxicity. Acta Histochem. 109 (1): 52-60. doi:10.1016/j.acthis.2006.09.007.
Bennukul K, Numkliang S, Leardkamolkarn V (2014) Melatonin attenuates cisplatin-induced HepG2 cell death via the regulation of mTOR and ERCC1 expressions. World J. Hepatol. 6 (4): 230–242. doi:10.4254/wjh.v6.i4.230.
Areti A, Komirishetty P, Akuthota M, Malik RA, Kumar A (2017) Melatonin prevents mitochondrial dysfunction and promotes neuroprotection by inducing autophagy during oxaliplatin-evoked peripheral neuropathy. J. Pineal Res. 62 (3): 10.1111/jpi.12393. doi:10.1111/jpi.12393.
Waseem M, Tabassum H, Parvez S (2016) Neuroprotective effects of melatonin as evidenced by abrogation of oxaliplatin induced behavioral alterations, mitochondrial dysfunction and neurotoxicity in rat brain. Mitochondrion 30: 168–176. doi:10.1016/j.mito.2016.08.001.
Wang YS, Li YY, Cui W, Li LB, Zhang ZC, Tian BP, Zhang GS (2017) Melatonin Attenuates Pain Hypersensitivity and Decreases Astrocyte-Mediated Spinal Neuroinflammation in a Rat Model of Oxaliplatin-Induced Pain. Inflammation 40 (6): 2052–2061. doi:10.1007/s10753-017-0645-y.
Ucar M, Korkmaz A, Reiter RJ, et al. (2007) Melatonin alleviates lung damage induced by the chemical warfare agent nitrogen mustard. Toxicol. Lett. 173 (2): 124-131. doi:10.1016/j.toxlet.2007.07.005.
Macit E, Yaren H, Aydin I, Kunak ZI, Yaman H, Onguru O, Uysal B, Korkmaz A, Turel S, Kenar L (2013) The protective effect of melatonin and S-methylisothiourea treatments in nitrogen mustard induced lung toxicity in rats. Environ. Toxicol. Pharmacol. 36 (3): 1283–1290. doi:10.1016/j.etap.2013.10.001.
Shokrzadeh M, Naghshvar F, Ahmadi A, Chabra A, Jeivad F (2014) The potential ameliorative effects of melatonin against cyclophosphamide-induced DNA damage in murine bone marrow cells. Eur. Rev. Med. Pharmacol. Sci. 18 (5): 605–611. PMID: 24668699.
Ferreira SG, Peliciari-Garcia RA, Takahashi-Hyodo SA, Rodrigues AC, Amaral FG, Berra CM, Bordin S, Curi R, Cipolla-Neto J (2013) Effects of melatonin on DNA damage induced by cyclophosphamide in rats. Braz. J. Med. Biol. Res. 46 (3): 278–286. doi:10.1590/1414-431x20122230.
Cui Y, Ren L, Li B, Fang J, Zhai Y, He X, Du E, Miao,Y, Hua, J, PengS (2017) Melatonin relieves busulfan-induced spermatogonial stem cell apoptosis of mouse testis by inhibiting endoplasmic reticulum stress. Cell. Physiol. Biochem. 44 (6): 2407–2421. doi:10.1159/000486165.
Abraham P, Kolli VK, Rabi S (2010) Melatonin attenuates methotrexate-induced oxidative stress and renal damage in rats. Cell Biochem. Funct. 28 (5): 426–433. doi:10.1002/cbf.1676.
Oguz E, Kocarslan S, Tabur S, Sezen H, Yilmaz Z, Aksoy N (2015) Effects of lycopene alone or combined with melatonin on methotrexate-induced nephrotoxicity in rats. Asian Pac. J. Cancer Prev. 16 (14): 6061–6066. doi:10.7314/apjcp.2015.16.14.6061.
Sirichoat A, Suwannakot K, Chaisawang P, Pannangrong W, Aranarochana A, Wigmore P,Welbat JU (2020) Melatonin attenuates 5-fluorouracil-induced spatial memory and hippocampal neurogenesis impairment in adult rats. Life Sci. 248: 117468. doi:10.1016/j.lfs.2020.117468.
Galley HF, McCormick B, Wilson KL, Lowes DA, Colvin L, Torsney C (2017) Melatonin limits paclitaxel-induced mitochondrial dysfunction in vitro and protects against paclitaxel-induced neuropathic pain in the rat. J. Pineal Res. 63 (4): e12444. doi:10.1111/jpi.12444.
Gianni L, Dafni U, Gelber RD, Azambuja E, Muehlbauer S, Goldhirsch A, Untch M, Smith I, Baselga J, Jackisch C, Cameron D, Mano M, Pedrini JL, Veronesi A, Mendiola C, Pluzanska A, Semiglazov V, Vrdoljak E, Eckart MJ, Shen Z, Herceptin Adjuvant (HERA) Trial Study Team (2011). Treatment with trastuzumab for 1 year after adjuvant chemotherapy in patients with HER2-positive early breast cancer: a 4-year follow-up of a randomised controlled trial. Lancet Oncol. 12 (3): 236–244. doi:10.1016/S1470-2045(11)70033-X.
Ghani EA, Kerr I, Dada R (2014) Grade 3 trastuzumab-induced neutropenia in breast cancer patient. J. Oncol. Pharm. Pract. 20 (2): 154–157. doi:10.1177/1078155213487394.
Ozturk M, Ozler M, Kurt YG, Ozturk B, Uysal B, Ersoz N, Yasar M, Demirbas S, Kurt B, Acikel C, Oztas Y, Arpaci F, Topal T, Ozet A, Ataergin S, Kuzhan O, Oter S, Korkmaz A (2011) Efficacy of melatonin, mercaptoethylguanidine and 1400W in doxorubicin- and trastuzumab-induced cardiotoxicity. J. Pineal Res. 50 (1): 89–96. doi:10.1111/j.1600-079X.2010.00818.x.
Reiter RJ, Rosales-Corral SA, Tan DX, Acuna-Castroviejo D, Qin L, Yang S, Xu K (2017) Melatonin, a full service anti-cancer agent: inhibition of initiation, progression and metastasis. Int. J. Mol. Sci. 18 (4): 843. doi:10.3390/ijms18040843.
Li Y, Li S, Zhou Y, Meng X, Zhang JJ, Xu DP, Li HB (2017) Melatonin for the prevention and treatment of cancer. Oncotarget 8 (24): 39896–39921. doi:10.18632/oncotarget.16379.
Lee JH, Yoon YM, Han YS, Yun CW, Lee, SH (2018) Melatonin promotes apoptosis of oxaliplatin-resistant colorectal cancer cells through inhibition of cellular prion protein. Anticancer Res. 38 (4): 1993–2000. doi:10.21873/anticanres.12437.
Sakatani A, Sonohara F, Goel A (2019) Melatonin-mediated downregulation of thymidylate synthase as a novel mechanism for overcoming 5-fluorouracil associated chemoresistance in colorectal cancer cells. Carcinogenesis 40 (3): 422–431. doi:10.1093/carcin/bgy186.
Liu Z, Sang X, Wang M, Liu Y, Liu J, Wang X, Liu P, Cheng H (2021) Melatonin potentiates the cytotoxic effect of Neratinib in HER2+ breast cancer through promoting endocytosis and lysosomal degradation of HER2. Oncogene 40 (44): 6273–6283. doi:10.1038/s41388-021-02015-w.
Fic M, Gomulkiewicz A, Grzegrzolka J, Podhorska-Okolow M, Zabel M, Dziegiel P, Jablonska K (2017) The impact of melatonin on colon cancer cells' resistance to doxorubicin in an in vitro study. Int. J. Mol. Sci. 18 (7): 1396. doi:10.3390/ijms18071396.
Koşar PA, Nazıroğlu M, Övey İS, Çiğ B (2016) Synergic effects of doxorubicin and melatonin on apoptosis and mitochondrial oxidative stress in MCF-7 breast cancer cells: Involvement of TRPV1 channels. J. Membr. Biol. 249 (1-2): 129–140. doi:10.1007/s00232-015-9855-0.
Chen L, Liu L, Li Y, Gao J (2018) Melatonin increases human cervical cancer HeLa cells apoptosis induced by cisplatin via inhibition of JNK/Parkin/mitophagy axis. In Vitro Cell Dev. Biol. Anim. 54 (1): 1–10. doi:10.1007/s11626-017-0200-z.
Gao Y, Xiao X, Zhang C, Yu W, Guo W, Zhang Z, Li Z, Feng X, Hao J, Zhang K, Xiao B, Chen M, Huang W, Xiong S, Wu X, Deng W (2017) Melatonin synergizes the chemotherapeutic effect of 5-fluorouracil in colon cancer by suppressing PI3K/AKT and NF-κB/iNOS signaling pathways. J. Pineal Res. 62 (2): 10.1111/jpi.12380. doi:10.1111/jpi.12380.
Lissoni P, Barni S, Mandala M, Ardizzoia A, Paolorossi F, Vaghi M, et al. (1999) 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 35 (12): 1688–1692. doi:10.1016/s0959-8049(99)00159-8.
Lissoni P, Chilelli M, Villa S, Cerizza L, Tancini G (2003) 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. 35 (1): 12–15. doi:10.1034/j.1600-079x.2003.00032.x.
Zharinov GM, Bogomolov OA, Chepurnaya IV, Neklasova NY, Anisimov VN (2020) Melatonin increases overall survival of prostate cancer patients with poor prognosis after combined hormone radiation treatment. Oncotarget 11 (41): 3723–3729. doi:10.18632/oncotarget.27757.
Reiter RJ, Tan DX, Sainz RM, Mayo JC, Lopez-Burillo S (2002) Melatonin: reducing the toxicity and increasing the efficacy of drugs. J. Pharm.Pharmacol. 54 (10): 1299–1321.doi:10.1211/002235702760345374.
Tan DX, Manchester LC, Liu X, Rosales-Corral SA, Acuna-Castroviejo D, Reiter RJ (2013) Mitochondria and chloroplasts as the original sites of melatonin synthesis: a hypothesis related to melatonin's primary function and evolution in eukaryotes. J. Pineal Res. 54 (2): 127–138. doi:10.1111/jpi.12026.
Anderson G (2022) Tumor microenvironment and metabolism: role of the mitochondrial melatonergic pathway in determining intercellular interactions in a new dynamic homeostasis. Int. J. Mol. Sci. 24 (1): 311. doi:10.3390/ijms24010311.
Yao JK, Dougherty GG.Jr, Reddy RD, Keshavan MS, Montrose DM, Matson WR, Rozen S, Krishnan RR, McEvoy J, Kaddurah-Daouk R (2010) Altered interactions of tryptophan metabolites in first-episode neuroleptic-naive patients with schizophrenia. Mol. Psychiatry 15 (9): 938–953. doi:10.1038/mp.2009.33.
Mokkawes T, de Visser SP (2023) Melatonin activation by cytochrome P450 isozymes: How does CYP1A2 compare to CYP1A1?. Int. J. Mol. Sci. 24 (4): 3651. doi:10.3390/ijms24043651.
Liu Y, Liang X, Dong W, Fang Y, Lv J, Zhang T, Fiskesund R, Xie J, Liu J, Yin X, Jin X, Chen D, Tang K, Ma J, Zhang H, Yu J, Yan J, Liang H, Mo S, Cheng F, Huang B (2018) Tumor-repopulating cells induce PD-1 expression in CD8+ T cells by transferring kynurenine and AhR activation. Cancer Cell 33 (3): 480–494.e7. doi:10.1016/j.ccell.2018.02.005.
Jang SW, Liu X, Yepes M, Shepherd KR, Miller GW, Liu Y, Wilson WD, Xiao G, Blanchi B, Sun YE, Ye K (2010) A selective TrkB agonist with potent neurotrophic activities by 7,8-dihydroxyflavone. Proc. Natl. Acad. Sci. USA 107 (6): 2687–2692.doi:10.1073/pnas.0913572107.
Anderson G, Reiter RJ (2019) Glioblastoma: Role of mitochondria n-acetylserotonin/melatonin ratio in mediating effects of mir-451 and aryl hydrocarbon receptor and in coordinating wider biochemical changes. Int. J. Tryptophan. Res. 12: 1178646919855942. doi:10.1177/1178646919855942.
Reiter RJ, Sharma R, Ma Q, Rosales-Corral SA, Acuna-Castroviejo D,Escames G (2019) Inhibition of mitochondrial pyruvate dehydrogenase kinase: a proposed mechanism by whichmelatonin causes cancer cells to overcome aerobic glycolysis, limit tumor growth and reverseinsensitivity to chemotherapy. Melatonin Res. 2 (3): 105-119. doi:10.32794/mr11250033.
Anderson G (2019) Daytime orexin and night-time melatonin regulation of mitochondria melatoninroles in circadian oscillations systemically and centrally in breast cancer symptomatology. Melatonin Res. 2 (4): 1-8; doi: 10.32794/mr11250037.
Huo X, Wang C, Yu Z, Peng Y, Wang S, Feng S, Zhang S, Tian X, Sun C, Liu K, Deng S, Ma X (2017) Human transporters, PEPT1/2, facilitate melatonin transportation into mitochondria of cancer cells: An implication of the therapeutic potential. J. Pineal Res. 62 (4): 10.1111/jpi.12390. doi:10.1111/jpi.12390.
Anderson G (2020) Tumourmicroenvironment: roles of the aryl hydrocarbon receptor, O-glcnacylation, acetyl-coa and melatonergic pathway in regulating dynamic metabolic interactions across cell types-tumour microenvironment and metabolism. Int. J. Mol. Sci. 22 (1): 141. doi:10.3390/ijms22010141.
Das NK, Samanta S (2022) The potential anti-cancer effects of melatonin on breast cancer. Explor. Med. 3: 112–27.doi: 10.37349/emed.2022.00078.
Wang L, Wang C, Choi, WS (2022) Use of melatonin in cancer treatment: Where are we?.Int. J. Mol. Sci. 23 (7): 3779. doi:10.3390/ijms23073779.
This work is licensed under a Creative Commons Attribution 4.0 International License