Next Article in Journal
Biomimetic Theranostic Agents with Superior NIR-II Photoacoustic and Magnetic Resonance Imaging Performance for Targeted Photothermal Therapy of Prostate Cancer
Previous Article in Journal
Polymeric Micelles Formulation of Combretastatin Derivatives with Enhanced Solubility, Cytostatic Activity and Selectivity against Cancer Cells
Previous Article in Special Issue
Potential Roles of Melatonin in Doxorubicin-Induced Cardiotoxicity: From Cellular Mechanisms to Clinical Application
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Therapeutic Potential of Melatonin Counteracting Chemotherapy-Induced Toxicity in Breast Cancer Patients: A Systematic Review

1
Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
2
Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain
3
Institute Teófilo Hernando for Drug Discovery, Department of Pharmacology, School of Medicine, Autonomous University of Madrid, 28029 Madrid, Spain
4
Faculty of Health, Camilo José Cela University of Madrid (UCJC), 28692 Madrid, Spain
5
Neuropsychopharmacology Unit, Hospital 12 de Octubre Research Institute, 28041 Madrid, Spain
6
Department of Biochemistry, Genetics and Immunology, Faculty of Biology, University of Vigo, 36310 Vigo, Spain
*
Authors to whom correspondence should be addressed.
Pharmaceutics 2023, 15(6), 1616; https://doi.org/10.3390/pharmaceutics15061616
Submission received: 23 February 2023 / Revised: 5 May 2023 / Accepted: 26 May 2023 / Published: 30 May 2023
(This article belongs to the Special Issue Recent Advances in Melatonin for Cancer Therapy)

Abstract

:
The purpose of this systematic review is to provide an overview of the existing knowledge on the therapeutic potential of melatonin to counteract the undesirable effects of chemotherapy in breast cancer patients. To this aim, we summarized and critically reviewed preclinical- and clinical-related evidence according to the PRISMA guidelines. Additionally, we developed an extrapolation of melatonin doses in animal studies to the human equivalent doses (HEDs) for randomized clinical trials (RCTs) with breast cancer patients. For the revision, 341 primary records were screened, which were reduced to 8 selected RCTs that met the inclusion criteria. We assembled the evidence drawn from these studies by analyzing the remaining gaps and treatment efficacy and suggested future translational research and clinical trials. Overall, the selected RCTs allow us to conclude that melatonin combined with standard chemotherapy lines would derive, at least, a better quality of life for breast cancer patients. Moreover, regular doses of 20 mg/day seemed to increase partial response and 1-year survival rates. Accordingly, this systematic review leads us to draw attention to the need for more RCTs to provide a comprehensive view of the promising actions of melatonin in breast cancer and, given the safety profile of this molecule, adequate translational doses should be established in further RCTs.

1. Introduction

Cancer is one of the most prevalent diseases and one of the leading causes of death worldwide [1], accounting for ~10 million deaths each year, which alarmingly continues to rapidly increase worldwide. The International Agency for Research on Cancer (IARC) has recently estimated a worsening of the global burden of the disease, from 14.1 million new cases (in 2012) to 21.7 million expected by 2030 [2]. In this same sense, it is expected that up to 40% of the adult population of men and women will be diagnosed with cancer throughout their lives, especially in developed countries. In the case of breast cancer, one of the reasons argued to explain the increasing trend in prevalence rates is industrialization, particularly artificial light at night (ALAN) [3]. Indeed, several studies have reported that prolonged and repeated exposure to ALAN [4] or sleep disturbance women with rotating night shifts [5] are key risk factors for this disease. Indeed, breast cancer is the most common cancer in women worldwide, accounting for ~25% of all cancer cases and affecting more than 2.2 million women each year [6]. Moreover, it is the second leading cause of cancer death in women (1 in 6 cancer deaths), exceeded only by lung cancer. However, despite the increase in overall cancer incidence, hopefully, the mortality rate of some tumors is being contained, which can be attributed to the immense research effort of the last decades that has allowed promising advances in immunotherapy by harnessing innate and/or adaptive immunity and precision medicine [7]. In this regard, breast cancer is a paradigmatic example of a highly prevalent tumor, whose in-deep study has achieved significant improvements in molecular pathogenesis, biomarkers to reach an early diagnosis and better outline the prognosis of patients, and therapeutic targets that improve the efficacy of clinical management. Additionally, the extension of prediagnostic screenings among the general population and/or risk cohorts has made it possible to routinely detect early tumors or even identify asymptomatic cases.
The multiplicity of treatments currently available to reduce disease severity and increase total and disease-free survival, including surgery, radiation therapy, chemotherapy, hormone therapy, targeted therapy, and immunotherapy, has been supplemented by multimodal combined therapies, which preserve therapeutic efficacy and patient life expectancy while reducing undesirable side-effects in diagnosed patients [8]. Despite this, the outcome of the patients, especially for metastatic breast cancer, is not satisfactory due to high recurrence and low survival rates [9]. Furthermore, some of the conventional treatment modalities are associated with non-specific toxicity responsible for detrimental side effects that have a significant impact on the quality of life of patients [10]. In this context, adjuvant radio/chemotherapy dosage regimens that achieve maximum drug efficacy confer a therapeutic advantage, although they may also result in off-target toxicity. Thus, thrombo and lymphocytopenia and other undesirable hematological and immunological disorders, together with fatigue, insomnia, cognitive impairment, peripheral neuropathy, depression, and psychological stress, are some of the adverse side effects of conventional chemotherapy lines in breast cancer [11]. Therefore, the development of effective practices in clinical interventions and a complementary adjunctive defense against toxicity associated with breast cancer therapy is urgent.
Increasing evidence suggests that melatonin (N-acetyl-5-methoxytryptamine) is a cell protector endowed with systemic effects against many types of cancer [12,13,14,15,16], including breast cancer [17,18,19,20]. In this regard, melatonin possesses antioxidant properties, which may help to protect cells from damage caused by free radicals and other harmful substances that can contribute to cancer development [21]. Specifically, melatonin directly scavenges free radicals, such as hydroxyl radical, peroxynitrite anion, singlet oxygen, and superoxide anion, thus protecting cells from oxidative stress [22]. Remarkably, the elimination of free radicals non-enzymatically transforms melatonin into metabolites with greater antioxidant capacity, which enabling the removal of 10 reactive species per molecule [23]. Antioxidative protection is especially relevant in the mitochondria of the extrapineal tissues, where more than 90% of body melatonin is synthesized and which, in addition to protecting organelles from reactive species, facilitates energy production [24]. In particular, melatonin activates transcription factors that control the expression of mitochondrial genes, such as complex I and complex IV, as well as influences ATP production and energy metabolism [21]. Furthermore, melatonin upregulates various antioxidant enzymes, such as superoxide dismutase, catalase, and glutathione peroxidase [22], which are responsible for neutralizing free radicals and preventing oxidative insults. Overall, the direct and indirect antioxidant activity of melatonin works together to ensure cell homeostasis and thereby promote health and well-being. Accordingly, several studies have found that individuals with higher melatonin levels show a lower risk of developing breast cancer, and melatonin supplementation may help inhibit the growth and spread of breast cancer cells [25,26]. Given the promising therapeutic potential of melatonin for breast cancer patients, there is great interest in conducting carefully designed multicenter clinical trials enrolling large prospective cohorts to fully understand its potential benefits and risks.
Further, two membrane-associated G protein-coupled melatonin receptors (named MT1 and MT2) are widely expressed in various tissues and organs, including the breast [27]. Some investigations have found that the activation of melatonin receptors in breast cancer cells may have antiproliferative and pro-apoptotic effects, leading to growth inhibition [28,29] and control of tumor angiogenesis [30]. In this respect, it has been reported that melatonin inhibits the development of breast cancer via the MT1 receptor, with an inverse correlation between the expression of the MT1 protein and the degree of malignancy of the tumors [31]. This fact reveals that MT1 may be used as a pathologic biomarker to evaluate breast carcinoma tissue [32]. On the other hand, estrogen has a significant role in the development and progression of breast cancer. Thus, estrogen/progesterone receptor-positive breast cancers include the most common types of breast cancer (75% of all cases). In this sense, melatonin is able to modulate the estrogen/ER signal transduction pathway, displaying an antiestrogenic effect and, therefore, inhibiting the growth of human breast tumors [32,33].
Melatonin has been shown to have anti-inflammatory properties by regulating several immune mechanisms that (i) enhance natural killer cell activity, (ii) inhibit the production of pro-inflammatory cytokines such as interleukin-1 beta (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α) [34,35], and (iii) protect hematopoietic progenitor cells from radiation therapy and chemotherapy [36]. Likewise, melatonin can inhibit the activation of nuclear factor-kappa B (NF-κB), which reduces the expression of pro-inflammatory genes and helps dampen the inflammatory response [37]. Consequently, the important melatonin-driven modulation of immune and inflammatory responses may be useful for controlling tumor progression and the clinical management of breast cancer patients [38]. Indeed, melatonin has aroused increasing expectations in recent years as an adjuvant in breast cancer therapy, reducing side effects and enhancing the efficacy of chemotherapy [39,40,41]. However, despite promising results from preclinical studies, the use of melatonin as an adjuvant in the treatment of breast cancer is still in the early stages of study, with some results suggesting potential benefits [42] and others not finding any effects [43]. Factors that may contribute to the variability in preliminary clinical results include differences in patient cohorts, dosing regimens, or study design. Therefore, further research to determine optimal dosing protocols and additional randomized clinical studies are necessary.

2. Materials and Methods

Systematic reviews and meta-analyses are accurate and reliable tools of evidence-based practice that are becoming the gold standard for collecting evidence related to any topic of interest and facilitating updating, the critical synthesis of information from sometimes confusing and contradictory literature, and optimizing the decision-making by professionals, avoiding biases and misunderstandings. In this regard, we performed a selection and bibliographic analysis according to the systematic review methodology of the PRISMA statement [44]. The review protocol was designed and validated by all authors. Specifically, following the PRISMA guidelines, we compiled the initial studies, assessed the quality and relationship of each one with the topic of interest, extracted their relevant data, and then carried out a critical synthesis. This method was selected to collect all the primary studies that may contain relevant RCTs regarding melatonin co-treatment in breast cancer up to 26 December 2022. We chose the PRISMA protocol to ensure the transparency and reproducibility of the criteria used for the selection and processing of the literature. Below, we describe the step-by-step PRISMA methodology.

2.1. Search Strategy

The systematic search for the primary reports was carried out in the main repositories of biomedical information, including Medline (https://pubmed.ncbi.nlm.nih.gov (accessed on 26 December 2022)), Scopus (https://www.scopus.com (accessed on 26 December 2022)), Web of Science (https://www.webofscience.com/wos/woscc/basic-search (accessed on 26 December 2022)), and the Cochrane library (https://www.cochranelibrary.com (accessed on 26 December 2022)). The identification of the relevant literature was conducted by means of key terms and Boolean operators combined in the following search algorithm: ((((breast cancer[Title/Abstract]) AND melatonin[Title/Abstract]) AND English[Language])) NOT REVIEW[Publication Type] AND (Intervention Studies OR intervention OR controlled trial OR “randomized” OR “randomized” OR “random” OR “randomly” OR “placebo” OR “assignment”).
When available, Medical Subject Heading (MESH) terms were used to reinforce the identification of all studies focused on the topic matter of the revision. Moreover, a systematic search was completed with hand searches.

2.2. Inclusion and Exclusion Criteria

Reviews were excluded and studies included were restricted to those published in the English language (26 December 2022). Of the results obtained following the aforementioned search strategy, only RCTs that co-administered melatonin in combination with at least one other chemotherapeutic agent to breast cancer patients were considered. Likewise, only studies comparing melatonin administration with a control or placebo group were included. Routes other than the oral administration (e.g., topical, systemic) of melatonin were excluded from the study.

2.3. Study Selection

Search results were exported to EndNote (https://www.myendnoteweb.com (accessed on 26 December 2022)), and duplicate publications were automatically or manually removed. Thereafter, eligible articles were selected based on their title, abstract, or full content when required. Studies included RCTs with chemotherapy-treated cohorts and control groups. Due to the low number of RCTs available in the literature, all studies dealing with combined chemotherapy and melatonin regimens were admitted. On the other hand, any disagreement regarding eligibility and/or inclusion was resolved by consensus.

2.4. Quality Assessment of Bias of the Selected Studies

To assess the methodological quality of the selected studies, we used the Cochrane collaboration tool (RoB 2) for assessing the risk of bias [45]. For each RCT, several sources to assess the risk of bias were considered. Indeed, RCTs included were assessed for the following domains of bias using the Cochrane tool RoB 2. Domain 1 refers to the randomization process; Domain 2 indicates the risk of deviations from the intended interventions; Domain 3 reflects any missing outcome data; Domain 4 is for outcome measurement; and Domain 5 refers to the selection of the reported result.
The domains for each study were graded as low risk, high risk, or of some concern risk of bias depending on whether available information was adequate, inadequate, or insufficient, respectively. The classification was developed independently by two authors (E.R. and A.R.), aside from the algorithm result of the RoB 2 toll. After grading, with the assistance of RoB 2 toll, the overall quality of each study was considered, and the final scores were discussed by all authors.
The described protocol is fully reproducible and allows for the inclusion of emerging information through newly updated or continually updated systematic reviews, which are also known as living systematic reviews [46].

3. Results

3.1. Selection and Identification of Relevant Studies

After the algorithmic and manual screenings of the aforementioned databases, 341 records were initially identified, of which 27 duplicates were removed. Of the 314 records reviewed, 198 were considered irrelevant and 33 corresponded to animal studies. In addition, the exclusion criteria applied were non-randomized controlled studies and non-chemotherapy-treated patients. Finally, only eight primary studies of breast cancer patients treated with chemotherapy and melatonin could be considered for the analysis. A detailed flowchart of the selection process is represented in Figure 1.

3.2. Characteristics of the Studies

Table 1 summarizes the main features of the included studies. We observed variations concerning doses, study design, and criteria for estimating patient outcomes in the selected RCTs. Overall, five articles included doses from 18–20 mg/day and three from 3–10 mg/day administered from 10 days to 36 months. Three studies included a placebo group, and a comparison was made with a non-melatonin-treated group in five studies, where patients were only treated with chemotherapy. It should be noted that it is highly probable that the variability of patient outcomes was also conditioned by the length of the trials.
The summary of the risk of bias (Table 2) was elaborated using the Cochrane collaboration tool RoB 2. In the case of insufficient information for domain assessment, the study was classified as “no information”. The quality assessment and risk of bias on five different quality domains reflected that none of the selected studies were classified as having a high risk of bias. As can be seen in Table 2, the overall quality of the RCTs considered ranged from low risk to some concerns. Due to the nature of the selected outcomes, no critical concerns were detected in the eight studies.

4. Comparative Doses

In the literature, several in vivo studies included melatonin as a co-treatment for breast cancer models. These preclinical investigations pointed out that co-treatment with melatonin would positively influence several clinical outcomes in breast cancer patients. Table 3 summarizes the effects of melatonin co-treatment in animal breast cancer studies. Research in this field showed the therapeutic potential of melatonin to improve the results in treated animals compared with those cancer animals not treated with indoleamine. One of the most frequent observations was the ability of melatonin to reduce tumor size [40,56,57,58,59,60,61,62,63], decrease the risk of metastasis [57,63,64,65,66] and angiogenesis [56,58,61,63,64], and enhance tumor stabilization rates [56,61,67,68,69].
To establish a correlation between melatonin doses in animal studies and RCTs, we calculated the human equivalent doses (HEDs) [70] (Table 3), which are based on the body surface area (BSA) of the animal calculated from body weight and length. The HED calculation is a critical step in ensuring that a drug dose is safe and effective for humans. It helps to avoid potential adverse effects or under-dosing that could compromise the efficacy of a drug. However, it is important to note that the HED has limitations, such as individual variability in metabolism and drug response, which must be taken into account in clinical trials. In this regard, the translation of optimal melatonin doses can be difficult to define because they vary depending on melatonin receptor desensitization, which can result from a variety of mechanisms, including receptor functionality (downregulation, internalization, conformational changes) and/or oscillations in downstream signaling pathways [71]. Additionally, the response of melatonin receptors to high doses of melatonin may depend on the receptor subtype, the treatment schedule (duration and frequency of exposure), and/or the cellular environment [72]. Therefore, although the exogenous administration of melatonin may be beneficial by potentiating antioxidant activity, it may also induce inappropriate responses, such as decreased responsiveness or even pro-oxidant effects [73]. In summary, it is important to carefully consider the dose and duration of melatonin treatment to achieve therapeutic efficacy and minimize possible spurious effects. These estimates are necessary to correctly translate active doses from animal to human studies and understand preclinical results in the context of RCTs. Table 3 also depicts some relevant findings of animal models related, to some extent, to those observed in breast cancer RCTs screened.
The observed outcomes in the selected RCTs are not as positive for melatonin co-administration as reported in preclinical studies. Although, the RCTs with 20 mg/day of melatonin showed very promising results in reducing chemotherapy toxicity, displaying higher partial responses and higher 1-year survival rates. Comparing results with preclinical data, we observed more extensive and encouraging findings as HED doses increased.
When effective animal doses were extrapolated to humans, the HED ranged from 1 to 486 mg per day. The eight clinical trials selected by the PRISMA guidelines tested doses from 3 to 20 mg of melatonin per day. This means that in most animal studies, equivalent doses calculated were significantly greater, even 24-fold greater than those administered to breast cancer patients in RCTs.

5. Discussion

Systematic methods based on standardized and explicit protocols make it possible to compile relevant research (primary papers or reports) related to a specific question (a topic of interest) and analyze the information according to objective quality criteria. The backbone of the systematic reviews is schematically reflected in the flow diagram in order to facilitate future reproductions and/or updates of the results and conclusions reached at any time. Thus, the present work is the first systematic review providing a comprehensive analysis of the evidence regarding the effectiveness of melatonin in counteracting chemotherapy-induced toxicity in breast cancer patients. The broad pleiotropy of this indoleamine gives it a presumed pivotal role in many signaling pathways impaired by malignant transformation and, due to its anti-estrogen activity, it is suggested as an ideal candidate for the prevention and/or regulation of the response to treatments. Thus, as far as breast cancer is concerned, diverse recent studies have pointed out that supplemental melatonin can synergistically potentiate drug cytotoxicity. Thus, in vivo and in vitro evidence in breast cancer cells support adjuvant melatonin-sensitized therapy-resistant, primary HER2-positive breast cancer cells for chemotherapy through increased oxidative stress in the endoplasmic reticulum [40]. Similarly, melatonin synergized the effectiveness of several chemotherapy lines by enhancing apoptosis of breast cancer cells [76,77,78,79] or inhibiting the progression of triple-negative breast cancer in a xenograft mouse model through the downregulation of two competing endogenous RNA antagonists of miR-101 [80]. Preclinical results also suggest that melatonin pretreatment prior to radiation sensitizes breast cancer cells to ionizing radiation [81], definitively emphasizing the translational importance of designing melatonin as an adjuvant in radiochemotherapy regimens [82]. Melatonin has also demonstrated proactivity in disrupting the EMT program that directs the implantation of vascular structures, and in VEGF-mediated paracrine regulation of the tumor microenvironment responsible for estrogen production and resistance to anti-angiogenic drugs [83,84,85]. In addition to breast tumor-suppressive capacity by favorably mediating therapy efficacy, supplementation with melatonin restores nocturnal endogenous levels disturbed by chemotherapy, improving sleep quality, circadian cyclicity, and the overall welfare of treated patients. It should be mentioned that in other studies, prescribed melatonin failed to affect breast cancer biomarkers in postmenopausal women with a prior history of breast cancer who had completed active cancer treatment [86], as well as ameliorate insomnia, fatigue, and other detrimental cancer-related symptomatologies [87].
In view of the functional biology of melatonin in breast cancer, the PRISMA methodology allowed the selection of RCTs that addressed this issue and evaluated the protective effects of oral melatonin in breast cancer patients treated with chemotherapy. Additionally, some promising evidence has been reported, such as the finding that topical melatonin counteracts the effects of radiotherapy in breast cancer patients [42]. To date, only eight RCTs have studied melatonin in this regard [47,48,49,50,51,52,53,54,55]. Among them, six tested melatonin at doses ranging from 10 to 20 mg/day and two at 3 mg/day. Overall, they involved 403 breast cancer patients, all of whom had different chemotherapy regimens. These studies showed that compared with placebo or non-treated patients, melatonin co-treatment reduced drug-induced toxicity, promoted higher partial responses, and increased 1-year survival rates. Correspondingly, adjuvant therapy with melatonin should be studied in this context as a way to reduce these side effects and improve the quality of life of breast cancer patients.
Additionally, we first calculated the translational doses of melatonin from the preclinical studies with animals to HED values and subsequently compared them to the doses used in RCTs to interrogate whether doses from experimental studies would be effective for human patients with breast cancer. Of note, the doses used in the reported RCTs have shown melatonin to be effective in reducing chemotherapy-induced toxicity at doses of 10 to 20 mg/day, as well as improving tumor regression rates at doses of 20 mg/day. RCTs with lower dosages of 3 mg/day revealed that melatonin improved sleep quality and fatigue. Mills et al. [88] observed that doses of melatonin ranging from 20–40 mg/day were effective in reducing the risk of cancer. However, it is important to emphasize that more research is needed to determine the optimal translational dose of melatonin in breast cancer patients. The dosages used in studies may not always be directly applicable to human patients, and individual variability in response to melatonin must also be taken into account.
Remarkably, no serious adverse effects were reported in these eight RCTs as a consequence of melatonin co-treatment. Moreover, several chemotherapy-induced toxicities, such as thrombocytopenia, myelosuppression, malaise, asthenia, stomatitis and neuropathy, leukopenia, and anemia or cardiac complications, were less common in melatonin-treated patients [47,48,49,50,53,54]. Therefore, we can conclude that melatonin does not appear to have safety concerns, but rather counteracts chemotherapy-related adverse effects. In this line, melatonin’s safety has been extensively reviewed in the literature [89,90,91].
Several limitations should be considered when reaching conclusions from the selected RCTs for this review. These limitations include the small number of RCTs performed that were available in the literature. Concerning the methodology of the RCTs, the lack of blinding the participants (medical staff and patients) increases the risk of incurring biases. However, objective assessments of patient outcomes would not likely be altered by insufficient blinding. Nonetheless, the heterogeneity of chemotherapy cycles and outcomes limited general conclusions because they made direct comparisons or meta-analyses on the efficacy of melatonin as a coadjuvant in breast cancer therapy impossible.
One of the limiting factors that prevent the direct extrapolation of the results from animal studies to humans is the fact that experimental animals (mainly rodents; mice and rats) tend to be nocturnal, whereas humans are diurnal. Intriguingly, circadian rhythm patterns can affect cancer development and progression [92]. Nocturnal animals have different metabolic rates than daytime animals, which may affect the absorption, distribution, and elimination of drugs and other active substances used in cancer treatment [93]. Furthermore, nocturnal animals exhibit different immune responses than humans and also different genetic backgrounds than humans, which may affect disease susceptibility, gene expression, drug metabolism, and other biological processes [94]. Therefore, while animal studies can provide valuable insights into cellular biochemistry and how it is affected by pharmacological and other interventions, it is important to keep in mind the limitations of extrapolating findings from research models to humans.
Melatonin is used as part of circadian-based regimens to maximize drug efficacy and tolerability (oncochronotherapies) by timing the delivery to match the body’s circadian rhythms. Thus, it has been suggested that administering melatonin at the appropriate phase of the circadian cycle may enhance its anti-tumor activity and reduce the side effects of chemotherapy and radiation therapy [95]. In this regard, while the optimal dosage of melatonin is an important consideration in cancer research, the timing of administration is also crucial to maximize its anti-cancer and other beneficial effects. The need for researchers to carefully consider the chronodependence of melatonin administration in their investigations and take into account the circadian rhythms in animal studies and clinical trials must be emphasized.
A great number of in vivo and in vitro reports on the efficacy of melatonin and its mechanisms of action against cancer models can be found in the literature [96,97], including its ability to regulate cell proliferation, apoptosis, and immune system function. Nevertheless, despite the promising actions reported in this systematic review, only a few RCT-reported studies have been conducted to date. Although the available clinical research is scarce, some RCTs have treated post-operative breast cancer patients with no additional chemotherapy and compared them with the placebo group, finding that melatonin led to better sleep quality and reduced depressive symptoms [98,99,100]. However, more clinical studies are needed to determine the optimal dose, timing, and route of administration of melatonin in cancer-treated patients.

6. Conclusions

Despite numerous animal-based studies available in the literature, only a few clinical trials confirming the benefits of melatonin co-treatment for breast cancer patients have been developed. The eight RCTs included in this review provide evidence for the safety and positive outcomes of melatonin as a therapeutic agent. However, these results are not sufficient evidence to conclude that clinical co-administration of melatonin may be effective due to the small number of trials carried out and the heterogeneity of treatment conditions and, consequently, of the outcomes reported. Furthermore, the doses to be administered in future RCTs should be reconsidered since we demonstrated that the direct extrapolation of effective doses in animal models to humans leads to doses that are likely too low to significantly influence tumor regression, metastasis, and/or angiogenesis. Therefore, more RCTs with more accurate melatonin doses are needed to confirm whether this indoleamine can generate synergies with chemotherapy that improve outcomes in breast cancer patients. In this regard, the potential benefits of melatonin co-treatment in improving the quality of life for cancer patients make it a promising avenue for future research.

Author Contributions

Conceptualization, A.R. and E.R.; formal analysis, E.R.; investigation, A.R., E.R. and E.G.-M.; resources, E.R., E.G.-M., F.L.-M. and J.E.; writing—original draft preparation, A.R. and E.R.; writing—review and editing, A.R., E.R., E.G.-M., F.L.-M. and J.E.; supervision, A.R. and E.R.; project administration, A.R.; funding acquisition, A.R. All authors have read and agreed to the published version of the manuscript.

Funding

This review was funded by UCJC (ONCOMEL 2021-23 and INFLAMAMEL 2022-07 projects).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Butterfield, L.H. Cancer vaccines. Bmj 2015, 350, h988. [Google Scholar] [CrossRef] [PubMed]
  2. Lybaert, L.; Vermaelen, K.; De Geest, B.G.; Nuhn, L. Immunoengineering through cancer vaccines—A personalized and multi-step vaccine approach towards precise cancer immunity. J. Control. Release 2018, 289, 125–145. [Google Scholar] [CrossRef]
  3. Schwimmer, H.; Metzer, A.; Pilosof, Y.; Szyf, M.; Machnes, Z.M.; Fares, F.; Harel, O.; Haim, A. Light at night and melatonin have opposite effects on breast cancer tumors in mice assessed by growth rates and global DNA methylation. Chronobiol. Int. 2014, 31, 144–150. [Google Scholar] [CrossRef]
  4. Stevens, R.G. Light-at-night, circadian disruption and breast cancer: Assessment of existing evidence. Int. J. Epidemiol. 2009, 38, 963–970. [Google Scholar] [CrossRef] [PubMed]
  5. Schernhammer, E.S.; Laden, F.; Speizer, F.E.; Willett, W.C.; Hunter, D.J.; Kawachi, I.; Colditz, G.A. Rotating night shifts and risk of breast cancer in women participating in the nurses’ health study. J. Natl. Cancer Inst. 2001, 93, 1563–1568. [Google Scholar] [CrossRef] [PubMed]
  6. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  7. Nagai, H.; Kim, Y.H. Cancer prevention from the perspective of global cancer burden patterns. J. Thorac. Dis. 2017, 9, 448–451. [Google Scholar] [CrossRef] [PubMed]
  8. Fisusi, F.A.; Akala, E.O. Drug Combinations in Breast Cancer Therapy. Pharm. Nanotechnol. 2019, 7, 3–23. [Google Scholar] [CrossRef]
  9. Arpino, G.; Milano, M.; De Placido, S. Features of aggressive breast cancer. Breast 2015, 24, 594–600. [Google Scholar] [CrossRef]
  10. Kerr, A.J.; Dodwell, D.; McGale, P.; Holt, F.; Duane, F.; Mannu, G.; Darby, S.C.; Taylor, C.W. Adjuvant and neoadjuvant breast cancer treatments: A systematic review of their effects on mortality. Cancer Treat. Rev. 2022, 105, 102375. [Google Scholar] [CrossRef]
  11. Di Nardo, P.; Lisanti, C.; Garutti, M.; Buriolla, S.; Alberti, M.; Mazzeo, R.; Puglisi, F. Chemotherapy in patients with early breast cancer: Clinical overview and management of long-term side effects. Expert. Opin. Drug. Saf. 2022, 21, 1341–1355. [Google Scholar] [CrossRef] [PubMed]
  12. Pathipaka, R.; Thyagarajan, A.; Sahu, R.P. Melatonin as a Repurposed Drug for Melanoma Treatment. Med. Sci. 2023, 11, 9. [Google Scholar] [CrossRef]
  13. Ngai, Z.N.; Chok, K.C.; Ng, K.Y.; Koh, R.Y.; Chye, S.M. Potential role of melatonin in prevention and treatment of lung cancer. Horm. Mol. Biol. Clin. Investig. 2022, 43, 485–503. [Google Scholar] [CrossRef] [PubMed]
  14. Moradian, F.; Pourhanifeh, M.H.; Mehrzadi, S.; Karimi-Behnagh, A.; Hosseinzadeh, A. Therapeutic potentials of melatonin in the treatment of lymphoma: A review of current evidence. Fundam. Clin. Pharmacol. 2022, 36, 777–789. [Google Scholar] [CrossRef] [PubMed]
  15. Shen, D.; Ju, L.; Zhou, F.; Yu, M.; Ma, H.; Zhang, Y.; Liu, T.; Xiao, Y.; Wang, X.; Qian, K. The inhibitory effect of melatonin on human prostate cancer. Cell. Commun. Signal. 2021, 19, 34. [Google Scholar] [CrossRef]
  16. Gil-Martin, E.; Egea, J.; Reiter, R.J.; Romero, A. The emergence of melatonin in oncology: Focus on colorectal cancer. Med. Res. Rev. 2019, 39, 2239–2285. [Google Scholar] [CrossRef]
  17. Kubatka, P.; Zubor, P.; Busselberg, D.; Kwon, T.K.; Adamek, M.; Petrovic, D.; Opatrilova, R.; Gazdikova, K.; Caprnda, M.; Rodrigo, L.; et al. Melatonin and breast cancer: Evidences from preclinical and human studies. Crit. Rev. Oncol. Hematol. 2018, 122, 133–143. [Google Scholar] [CrossRef]
  18. Kong, X.; Gao, R.; Wang, Z.; Wang, X.; Fang, Y.; Gao, J.; Reiter, R.J.; Wang, J. Melatonin: A Potential Therapeutic Option for Breast Cancer. Trends Endocrinol. Metab. 2020, 31, 859–871. [Google Scholar] [CrossRef]
  19. Samanta, S. Melatonin: A Potential Antineoplastic Agent in Breast Cancer. J. Environ. Pathol. Toxicol. Oncol. 2022, 41, 55–84. [Google Scholar] [CrossRef]
  20. Franco, P.I.R.; do Carmo Neto, J.R.; Milhomem, A.C.; Machado, J.R.; Miguel, M.P. Antitumor effect of melatonin on breast cancer in experimental models: A systematic review. Biochim. Biophys. Acta Rev. Cancer 2023, 1878, 188838. [Google Scholar] [CrossRef]
  21. Florido, J.; Rodriguez-Santana, C.; Martinez-Ruiz, L.; Lopez-Rodriguez, A.; Acuna-Castroviejo, D.; Rusanova, I.; Escames, G. Understanding the Mechanism of Action of Melatonin, Which Induces ROS Production in Cancer Cells. Antioxidants 2022, 11, 1621. [Google Scholar] [CrossRef]
  22. Rodriguez, C.; Mayo, J.C.; Sainz, R.M.; Antolin, I.; Herrera, F.; Martin, V.; Reiter, R.J. Regulation of antioxidant enzymes: A significant role for melatonin. J. Pineal Res. 2004, 36, 1–9. [Google Scholar] [CrossRef]
  23. Hardeland, R. Melatonin, Its Metabolites and Their Interference with Reactive Nitrogen Compounds. Molecules 2021, 26, 4105. [Google Scholar] [CrossRef] [PubMed]
  24. Suofu, Y.; Li, W.; Jean-Alphonse, F.G.; Jia, J.; Khattar, N.K.; Li, J.; Baranov, S.V.; Leronni, D.; Mihalik, A.C.; He, Y.; et al. Dual role of mitochondria in producing melatonin and driving GPCR signaling to block cytochrome c release. Proc. Natl. Acad. Sci. USA 2017, 114, E7997–E8006. [Google Scholar] [CrossRef] [PubMed]
  25. Jia, Y.; Lu, Y.; Wu, K.; Lin, Q.; Shen, W.; Zhu, M.; Huang, S.; Chen, J. Does night work increase the risk of breast cancer? A systematic review and meta-analysis of epidemiological studies. Cancer Epidemiol. 2013, 37, 197–206. [Google Scholar] [CrossRef]
  26. Schernhammer, E.S.; Hankinson, S.E. Urinary melatonin levels and postmenopausal breast cancer risk in the Nurses’ Health Study cohort. Cancer Epidemiol. Biomarkers Prev. 2009, 18, 74–79. [Google Scholar] [CrossRef]
  27. Liu, J.; Clough, S.J.; Hutchinson, A.J.; Adamah-Biassi, E.B.; Popovska-Gorevski, M.; Dubocovich, M.L. MT1 and MT2 Melatonin Receptors: A Therapeutic Perspective. Annu. Rev. Pharmacol. Toxicol. 2016, 56, 361–383. [Google Scholar] [CrossRef]
  28. Girgert, R.; Hanf, V.; Emons, G.; Gründker, C. Membrane-bound melatonin receptor MT1 down-regulates estrogen responsive genes in breast cancer cells. J. Pineal Res. 2009, 47, 23–31. [Google Scholar] [CrossRef] [PubMed]
  29. Yuan, L.; Collins, A.R.; Dai, J.; Dubocovich, M.L.; Hill, S.M. MT(1) melatonin receptor overexpression enhances the growth suppressive effect of melatonin in human breast cancer cells. Mol. Cell. Endocrinol. 2002, 192, 147–156. [Google Scholar] [CrossRef]
  30. Ma, Q.; Reiter, R.J.; Chen, Y. Role of melatonin in controlling angiogenesis under physiological and pathological conditions. Angiogenesis 2020, 23, 91–104. [Google Scholar] [CrossRef] [PubMed]
  31. Jablonska, K.; Pula, B.; Zemla, A.; Owczarek, T.; Wojnar, A.; Rys, J.; Ambicka, A.; Podhorska-Okolow, M.; Ugorski, M.; Dziegiel, P. Expression of melatonin receptor MT1 in cells of human invasive ductal breast carcinoma. J. Pineal Res. 2013, 54, 334–345. [Google Scholar] [CrossRef] [PubMed]
  32. Goyal, R.; Gupta, T.; Bal, A.; Sahni, D.; Singh, G. Role of Melatonin in Breast Carcinoma: Correlation of Expression Patterns of Melatonin-1 Receptor With Estrogen, Progesterone, and HER2 Receptors. Appl. Immunohistochem. Mol. Morphol. 2020, 28, 518–523. [Google Scholar] [CrossRef] [PubMed]
  33. Menendez-Menendez, J.; Martinez-Campa, C. Melatonin: An Anti-Tumor Agent in Hormone-Dependent Cancers. Int. J. Endocrinol. 2018, 2018, 3271948. [Google Scholar] [CrossRef]
  34. Mannino, G.; Caradonna, F.; Cruciata, I.; Lauria, A.; Perrone, A.; Gentile, C. Melatonin reduces inflammatory response in human intestinal epithelial cells stimulated by interleukin-1beta. J. Pineal Res. 2019, 67, e12598. [Google Scholar] [CrossRef]
  35. Cho, J.H.; Bhutani, S.; Kim, C.H.; Irwin, M.R. Anti-inflammatory effects of melatonin: A systematic review and meta-analysis of clinical trials. Brain Behav. Immun. 2021, 93, 245–253. [Google Scholar] [CrossRef] [PubMed]
  36. Tan, S.S.; Zhan, W.; Poon, C.J.; Han, X.; Marre, D.; Boodhun, S.; Palmer, J.A.; Mitchell, G.M.; Morrison, W.A. Melatonin promotes survival of nonvascularized fat grafts and enhances the viability and migration of human adipose-derived stem cells via down-regulation of acute inflammatory cytokines. J. Tissue Eng. Regen. Med. 2018, 12, 382–392. [Google Scholar] [CrossRef] [PubMed]
  37. Minocha, T.; Das, M.; Rai, V.; Verma, S.S.; Awasthee, N.; Gupta, S.C.; Haldar, C.; Yadav, S.K. Melatonin induces apoptosis and cell cycle arrest in cervical cancer cells via inhibition of NF-kappaB pathway. Inflammopharmacology 2022, 30, 1411–1429. [Google Scholar] [CrossRef]
  38. Moslehi, M.; Moazamiyanfar, R.; Dakkali, M.S.; Rezaei, S.; Rastegar-Pouyani, N.; Jafarzadeh, E.; Mouludi, K.; Khodamoradi, E.; Taeb, S.; Najafi, M. Modulation of the immune system by melatonin; implications for cancer therapy. Int. Immunopharmacol. 2022, 108, 108890. [Google Scholar] [CrossRef]
  39. Rohilla, S.; Singh, M.; Priya, S.; Almalki, W.H.; Haniffa, S.M.; Subramaniyan, V.; Fuloria, S.; Fuloria, N.K.; Sekar, M.; Singh, S.K.; et al. Exploring the Mechanical Perspective of a New Anti-Tumor Agent: Melatonin. J. Environ. Pathol. Toxicol. Oncol. 2023, 42, 1–16. [Google Scholar] [CrossRef] [PubMed]
  40. Sang, X.; Li, L.; Rui, C.; Liu, Y.; Liu, Z.; Tao, Z.; Cheng, H.; Liu, P. Induction of EnR stress by Melatonin enhances the cytotoxic effect of Lapatinib in HER2-positive breast cancer. Cancer Lett. 2021, 518, 82–93. [Google Scholar] [CrossRef]
  41. Estirado, S.; Fernández-Delgado, E.; Viñuelas-Zahínos, E.; Luna-Giles, F.; Rodríguez, A.B.; Pariente, J.A.; Espino, J. Pro-Apoptotic and Anti-Migration Properties of a Thiazoline-Containing Platinum(II) Complex in MDA-MB-231 Breast Cancer Cells: The Role of Melatonin as a Synergistic Agent. Antioxidants 2022, 11, 1971. [Google Scholar] [CrossRef] [PubMed]
  42. Zetner, D.; Kamby, C.; Gülen, S.; Christophersen, C.; Paulsen, C.B.; Piga, E.; Hoffmeyer, B.; Mahmood, F.; Rosenberg, J. Quality-of-life outcomes following topical melatonin application against acute radiation dermatitis in patients with early breast cancer: A double-blind, randomized, placebo-controlled trial. J. Pineal Res. 2023, 74, e12840. [Google Scholar] [CrossRef] [PubMed]
  43. Pistiolis, L.; Khaki, D.; Kovács, A.; Olofsson Bagge, R. The Effect of Melatonin Intake on Survival of Patients with Breast Cancer-A Population-Based Registry Study. Cancers 2022, 14, 5884. [Google Scholar] [CrossRef] [PubMed]
  44. Liberati, A.; Altman, D.G.; Tetzlaff, J.; Mulrow, C.; Gotzsche, P.C.; Ioannidis, J.P.; Clarke, M.; Devereaux, P.J.; Kleijnen, J.; Moher, D. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: Explanation and elaboration. PLoS Med. 2009, 6, e1000100. [Google Scholar] [CrossRef] [PubMed]
  45. Higgins, J.; Savovic, J.; Page, M.J.; Elbers, R.G.; Sterne, J.A.C. Assessing risk of bias in a randomized trial. In Cochrane Handbook for Systematic Reviews of Interventions Version 6.3; Higgins, J.P.T.T.J., Chandler, J., Cumpston, M., Li, T., Page, M.J., Welch, V.A., Eds.; John Wiley & Sons: UK, London, 2022. [Google Scholar]
  46. Elliott, J.H.; Turner, T.; Clavisi, O.; Thomas, J.; Higgins, J.P.; Mavergames, C.; Gruen, R.L. Living systematic reviews: An emerging opportunity to narrow the evidence-practice gap. PLoS Med. 2014, 11, e1001603. [Google Scholar] [CrossRef]
  47. Lissoni, P.; Ardizzoia, A.; Barni, S.; Paolorossi, F.; Tancini, G.; Meregalli, S.; Esposti, D.; Zubelewicz, B.; Braczowski, R. A randomized study of tamoxifen alone versus tamoxifen plus melatonin in estrogen receptor-negative heavily pretreated metastatic breast-cancer patients. Oncol. Rep. 1995, 2, 871–873. [Google Scholar] [CrossRef]
  48. Lissoni, P.; Tancini, G.; Barni, S.; Paolorossi, F.; Ardizzoia, A.; Conti, A.; Maestroni, G. Treatment of cancer chemotherapy-induced toxicity with the pineal hormone melatonin. Support. Care Cancer 1997, 5, 126–129. [Google Scholar] [CrossRef]
  49. Lissoni, P.; Barni, S.; Mandalà, M.; Ardizzoia, A.; Paolorossi, F.; Vaghi, M.; Longarini, R.; Malugani, F.; Tancini, G. Decreased toxicity and increased efficacy of cancer chemotherapy using the pineal hormone melatonin in metastatic solid tumour patients with poor clinical status. Eur. J. Cancer 1999, 35, 1688–1692. [Google Scholar] [CrossRef]
  50. Palmer, A.C.S.; Souza, A.; Dos Santos, V.S.; Cavalheiro, J.A.C.; Schuh, F.; Zucatto, A.E.; Biazus, J.V.; Torres, I.; Fregni, F.; Caumo, W. The Effects of Melatonin on the Descending Pain Inhibitory System and Neural Plasticity Markers in Breast Cancer Patients Receiving Chemotherapy: Randomized, Double-Blinded, Placebo-Controlled Trial. Front. Pharmacol. 2019, 10, 1382. [Google Scholar] [CrossRef]
  51. Palmer, A.C.S.; Zortea, M.; Souza, A.; Santos, V.; Biazús, J.V.; Torres, I.L.S.; Fregni, F.; Caumo, W. Clinical impact of melatonin on breast cancer patients undergoing chemotherapy; effects on cognition, sleep and depressive symptoms: A randomized, double-blind, placebo-controlled trial. PLoS ONE 2020, 15, e0231379. [Google Scholar] [CrossRef]
  52. Semiglazova, T.Y.; Osipov, M.; Krivorotko, P.; Protsenko, S.; Semiglazov, V.; Donskih, R.; Klimenko, V.; Tsirlina, E.; Berstein, L.; Anisimov, V.; et al. Neoadjuvant endocrine therapy in combination with melatonin and metformin in locally advanced breast cancer. Ann. Oncol. 2019, 30, v99–v100. [Google Scholar] [CrossRef]
  53. Semiglazova, T.Y.; Osipov, M.; Krivorotko, P.; Semiglazov, V.; Protsenko, S.; Berstein, L.; Tsirlina, E.; Klimenko, V.; Donskih, R.; Anisimov, V.; et al. Melatonin and metformin in neoadjuvant chemotherapy in locally advanced breast cancer. Ann. Oncol. 2019, 30, v100. [Google Scholar] [CrossRef]
  54. Sedighi Pashaki, A.; Mohammadian, K.; Afshar, S.; Gholami, M.H.; Moradi, A.; Javadinia, S.A.; Keshtpour Amlashi, Z. A Randomized, Controlled, Parallel-Group, Trial on the Effects of Melatonin on Fatigue Associated with Breast Cancer and Its Adjuvant Treatments. Integr. Cancer Ther. 2021, 20, 1534735420988343. [Google Scholar] [CrossRef] [PubMed]
  55. Talaee, N.; Ebrahimpour, S.; Sfandbod, M.; Majedi, H.; Kohneloo, A.J.; Gholami, K.; Jahangard-Rafsanjani, Z. Effect of Melatonin on Paclitaxel-Associated Acute and Chronic Pain: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Pharm. Sci. 2022, 28, 579–588. [Google Scholar] [CrossRef]
  56. Odeh, L.H.; Talib, W.H.; Basheti, I.A. Synergistic effect of thymoquinone and melatonin against breast cancer implanted in mice. J. Cancer Res. Ther. 2018, 14, S324–S330. [Google Scholar] [CrossRef] [PubMed]
  57. Cos, S.; Fernández, R.; Güézmes, A.; Sánchez-Barceló, E.J. Influence of melatonin on invasive and metastatic properties of MCF-7 human breast cancer cells. Cancer Res. 1998, 58, 4383–4390. [Google Scholar]
  58. Jardim-Perassi, B.V.; Arbab, A.S.; Ferreira, L.C.; Borin, T.F.; Varma, N.R.; Iskander, A.S.; Shankar, A.; Ali, M.M.; de Campos Zuccari, D.A. Effect of melatonin on tumor growth and angiogenesis in xenograft model of breast cancer. PLoS ONE 2014, 9, e85311. [Google Scholar] [CrossRef] [PubMed]
  59. Colombo, J.; Jardim-Perassi, B.V.; Ferreira, J.P.S.; Braga, C.Z.; Sonehara, N.M.; Júnior, R.P.; Moschetta, M.G.; Girol, A.P.; Zuccari, D. Melatonin Differentially Modulates NF-кB Expression in Breast and Liver Cancer Cells. Anticancer. Agents Med. Chem. 2018, 18, 1688–1694. [Google Scholar] [CrossRef]
  60. Jardim-Perassi, B.V.; Alexandre, P.A.; Sonehara, N.M.; de Paula-Junior, R.; Reis Júnior, O.; Fukumasu, H.; Chammas, R.; Coutinho, L.L.; Zuccari, D. RNA-Seq transcriptome analysis shows anti-tumor actions of melatonin in a breast cancer xenograft model. Sci. Rep. 2019, 9, 966. [Google Scholar] [CrossRef]
  61. Kubatka, P.; Bojková, B.; Kassayová, M.; Orendáš, P.; Kajo, K.; Výbohová, D.; Kružliak, P.; Adamicová, K.; Péč, M.; Stollárová, N.; et al. Combination of Pitavastatin and melatonin shows partial antineoplastic effects in a rat breast carcinoma model. Acta Histochem. 2014, 116, 1454–1461. [Google Scholar] [CrossRef]
  62. Moselhy, S.S.; Al mslmani, M.A. Chemopreventive effect of lycopene alone or with melatonin against the genesis of oxidative stress and mammary tumors induced by 7,12 dimethyl(a)benzanthracene in sprague dawely female rats. Mol. Cell. Biochem. 2008, 319, 175–180. [Google Scholar] [CrossRef] [PubMed]
  63. Talib, W.H.; Saleh, S. Propionibacterium acnes Augments Antitumor, Anti-Angiogenesis and Immunomodulatory Effects of Melatonin on Breast Cancer Implanted in Mice. PLoS ONE 2015, 10, e0124384. [Google Scholar] [CrossRef] [PubMed]
  64. Karadas, A.K.; Dilmac, S.; Aytac, G.; Tanriover, G. Melatonin decreases metastasis, primary tumor growth and angiogenesis in a mice model of breast cancer. Hum. Exp. Toxicol. 2021, 40, 1545–1557. [Google Scholar] [CrossRef] [PubMed]
  65. Oshiba, R.T.; Touson, E.; Elsherbini, Y.M.; Abdraboh, M.E. Melatonin: A regulator of the interplay between FoxO1, miR96, and miR215 signaling to diminish the growth, survival, and metastasis of murine adenocarcinoma. Biofactors 2021, 47, 740–753. [Google Scholar] [CrossRef]
  66. Borin, T.F.; Arbab, A.S.; Gelaleti, G.B.; Ferreira, L.C.; Moschetta, M.G.; Jardim-Perassi, B.V.; Iskander, A.S.; Varma, N.R.; Shankar, A.; Coimbra, V.B.; et al. Melatonin decreases breast cancer metastasis by modulating Rho-associated kinase protein-1 expression. J. Pineal Res. 2016, 60, 3–15. [Google Scholar] [CrossRef]
  67. Panchenko, A.V.; Tyndyk, M.L.; Maydin, M.A.; Baldueva, I.A.; Artemyeva, A.S.; Kruglov, S.S.; Kireeva, G.S.; Golubev, A.G.; Belyaev, A.M.; Anisimov, V.N. Melatonin Administered before or after a Cytotoxic Drug Increases Mammary Cancer Stabilization Rates in HER2/Neu Mice. Chemotherapy 2020, 65, 42–50. [Google Scholar] [CrossRef]
  68. Kassayová, M.; Bobrov, N.; Strojný, L.; Orendáš, P.; Demečková, V.; Jendželovský, R.; Kubatka, P.; Kisková, T.; Kružliak, P.; Adamkov, M.; et al. Anticancer and Immunomodulatory Effects of Lactobacillus plantarum LS/07, Inulin and Melatonin in NMU-induced Rat Model of Breast Cancer. Anticancer. Res. 2016, 36, 2719–2728. [Google Scholar]
  69. Orendáš, P.; Kubatka, P.; Bojková, B.; Kassayová, M.; Kajo, K.; Výbohová, D.; Kružliak, P.; Péč, M.; Adamkov, M.; Kapinová, A.; et al. Melatonin potentiates the anti-tumour effect of pravastatin in rat mammary gland carcinoma model. Int. J. Exp. Pathol. 2014, 95, 401–410. [Google Scholar] [CrossRef]
  70. Reagan-Shaw, S.; Nihal, M.; Ahmad, N. Dose translation from animal to human studies revisited. Faseb. J. 2008, 22, 659–661. [Google Scholar] [CrossRef]
  71. Gerdin, M.J.; Masana, M.I.; Dubocovich, M.L. Melatonin-mediated regulation of human MT(1) melatonin receptors expressed in mammalian cells. Biochem. Pharmacol. 2004, 67, 2023–2030. [Google Scholar] [CrossRef]
  72. Hardeland, R. Divergent Importance of Chronobiological Considerations in High- and Low-dose Melatonin Therapies. Diseases 2021, 9, 18. [Google Scholar] [CrossRef]
  73. Reiter, R.J.; Tan, D.X.; Galano, A. Melatonin: Exceeding expectations. Physiology 2014, 29, 325–333. [Google Scholar] [CrossRef] [PubMed]
  74. Talib, W.H. A ketogenic diet combined with melatonin overcomes cisplatin and vincristine drug resistance in breast carcinoma syngraft. Nutrition 2020, 72, 110659. [Google Scholar] [CrossRef]
  75. Anisimov, V.N.; Alimova, I.N.; Baturin, D.A.; Popovich, I.G.; Zabezhinski, M.A.; Manton, K.G.; Semenchenko, A.V.; Yashin, A.I. The effect of melatonin treatment regimen on mammary adenocarcinoma development in HER-2/neu transgenic mice. Int. J. Cancer 2003, 103, 300–305. [Google Scholar] [CrossRef] [PubMed]
  76. Alonso-González, C.; Menéndez-Menéndez, J.; González-González, A.; González, A.; Cos, S.; Martínez-Campa, C. Melatonin enhances the apoptotic effects and modulates the changes in gene expression induced by docetaxel in MCF-7 human breast cancer cells. Int. J. Oncol. 2018, 52, 560–570. [Google Scholar] [CrossRef]
  77. Woo, S.M.; Min, K.J.; Kwon, T.K. Melatonin-mediated Bim up-regulation and cyclooxygenase-2 (COX-2) down-regulation enhances tunicamycin-induced apoptosis in MDA-MB-231 cells. J. Pineal Res. 2015, 58, 310–320. [Google Scholar] [CrossRef] [PubMed]
  78. Sabzichi, M.; Samadi, N.; Mohammadian, J.; Hamishehkar, H.; Akbarzadeh, M.; Molavi, O. Sustained release of melatonin: A novel approach in elevating efficacy of tamoxifen in breast cancer treatment. Colloids Surf. B. Biointerfaces 2016, 145, 64–71. [Google Scholar] [CrossRef] [PubMed]
  79. Tran, Q.H.; Hoang, D.H.; Song, M.; Choe, W.; Kang, I.; Kim, S.S.; Ha, J. Melatonin and doxorubicin synergistically enhance apoptosis via autophagy-dependent reduction of AMPKα1 transcription in human breast cancer cells. Exp. Mol. Med. 2021, 53, 1413–1422. [Google Scholar] [CrossRef]
  80. Yang, A.; Peng, F.; Zhu, L.; Li, X.; Ou, S.; Huang, Z.; Wu, S.; Peng, C.; Liu, P.; Kong, Y. Melatonin inhibits triple-negative breast cancer progression through the Lnc049808-FUNDC1 pathway. Cell. Death Dis. 2021, 12, 712. [Google Scholar] [CrossRef]
  81. Alonso-González, C.; González, A.; Martínez-Campa, C.; Gómez-Arozamena, J.; Cos, S. Melatonin sensitizes human breast cancer cells to ionizing radiation by downregulating proteins involved in double-strand DNA break repair. J. Pineal Res. 2015, 58, 189–197. [Google Scholar] [CrossRef]
  82. Ben-David, M.A.; Elkayam, R.; Gelernter, I.; Pfeffer, R.M. Melatonin for Prevention of Breast Radiation Dermatitis: A Phase II, Prospective, Double-Blind Randomized Trial. Isr. Med. Assoc. J. 2016, 18, 188–192. [Google Scholar] [PubMed]
  83. Lacerda, J.Z.; Ferreira, L.C.; Lopes, B.C.; Aristizábal-Pachón, A.F.; Bajgelman, M.C.; Borin, T.F.; Zuccari, D. Therapeutic Potential of Melatonin in the Regulation of MiR-148a-3p and Angiogenic Factors in Breast Cancer. Microrna 2019, 8, 237–247. [Google Scholar] [CrossRef] [PubMed]
  84. Maroufi, N.F.; Amiri, M.; Dizaji, B.F.; Vahedian, V.; Akbarzadeh, M.; Roshanravan, N.; Haiaty, S.; Nouri, M.; Rashidi, M.R. Inhibitory effect of melatonin on hypoxia-induced vasculogenic mimicry via suppressing epithelial-mesenchymal transition (EMT) in breast cancer stem cells. Eur. J. Pharmacol. 2020, 881, 173282. [Google Scholar] [CrossRef] [PubMed]
  85. Alvarez-García, V.; González, A.; Alonso-González, C.; Martínez-Campa, C.; Cos, S. Regulation of vascular endothelial growth factor by melatonin in human breast cancer cells. J. Pineal Res. 2013, 54, 373–380. [Google Scholar] [CrossRef]
  86. Schernhammer, E.S.; Giobbie-Hurder, A.; Gantman, K.; Savoie, J.; Scheib, R.; Parker, L.M.; Chen, W.Y. A randomized controlled trial of oral melatonin supplementation and breast cancer biomarkers. Cancer Causes Control. 2012, 23, 609–616. [Google Scholar] [CrossRef]
  87. Lund Rasmussen, C.; Klee Olsen, M.; Thit Johnsen, A.; Petersen, M.A.; Lindholm, H.; Andersen, L.; Villadsen, B.; Groenvold, M.; Pedersen, L. Effects of melatonin on physical fatigue and other symptoms in patients with advanced cancer receiving palliative care: A double-blind placebo-controlled crossover trial. Cancer 2015, 121, 3727–3736. [Google Scholar] [CrossRef]
  88. Mills, E.; Wu, P.; Seely, D.; Guyatt, G. Melatonin in the treatment of cancer: A systematic review of randomized controlled trials and meta-analysis. J. Pineal Res. 2005, 39, 360–366. [Google Scholar] [CrossRef]
  89. Andersen, L.P.; Gogenur, I.; Rosenberg, J.; Reiter, R.J. The Safety of Melatonin in Humans. Clin. Drug. Investig. 2016, 36, 169–175. [Google Scholar] [CrossRef]
  90. Foley, H.M.; Steel, A.E. Adverse events associated with oral administration of melatonin: A critical systematic review of clinical evidence. Complement. Ther. Med. 2019, 42, 65–81. [Google Scholar] [CrossRef]
  91. Seabra, M.L.; Bignotto, M.; Pinto, L.R., Jr.; Tufik, S. Randomized, double-blind clinical trial, controlled with placebo, of the toxicology of chronic melatonin treatment. J. Pineal Res. 2000, 29, 193–200. [Google Scholar] [CrossRef]
  92. Zhou, J.; Wang, J.; Zhang, X.; Tang, Q. New Insights Into Cancer Chronotherapies. Front. Pharmacol. 2021, 12, 741295. [Google Scholar] [CrossRef] [PubMed]
  93. Hrushesky, W.J. Circadian timing of cancer chemotherapy. Science 1985, 228, 73–75. [Google Scholar] [CrossRef]
  94. Ozturk, N.; Ozturk, D.; Kavakli, I.H.; Okyar, A. Molecular Aspects of Circadian Pharmacology and Relevance for Cancer Chronotherapy. Int. J. Mol. Sci. 2017, 18, 2168. [Google Scholar] [CrossRef] [PubMed]
  95. Levi, F. Circadian chronotherapy for human cancers. Lancet Oncol. 2001, 2, 307–315. [Google Scholar] [CrossRef] [PubMed]
  96. Li, Y.; Li, S.; Zhou, Y.; Meng, X.; Zhang, J.J.; Xu, D.P.; Li, H.B. Melatonin for the prevention and treatment of cancer. Oncotarget 2017, 8, 39896–39921. [Google Scholar] [CrossRef]
  97. Talib, W.H.; Alsayed, A.R.; Abuawad, A.; Daoud, S.; Mahmod, A.I. Melatonin in Cancer Treatment: Current Knowledge and Future Opportunities. Molecules 2021, 26, 2506. [Google Scholar] [CrossRef]
  98. Hansen, M.V.; Madsen, M.T.; Andersen, L.T.; Hageman, I.; Rasmussen, L.S.; Bokmand, S.; Rosenberg, J.; Gögenur, I. Effect of Melatonin on Cognitive Function and Sleep in relation to Breast Cancer Surgery: A Randomized, Double-Blind, Placebo-Controlled Trial. Int. J. Breast Cancer 2014, 2014, 416531. [Google Scholar] [CrossRef]
  99. Madsen, M.T.; Hansen, M.V.; Andersen, L.T.; Hageman, I.; Rasmussen, L.S.; Bokmand, S.; Rosenberg, J.; Gögenur, I. Effect of melatonin on sleep in the perioperative period after breast cancer surgery: A randomized, double-blind, placebo-controlled trial. J. Clin. Sleep Med. 2016, 12, 225–233. [Google Scholar] [CrossRef]
  100. Hansen, M.V.; Andersen, L.T.; Madsen, M.T.; Hageman, I.; Rasmussen, L.S.; Bokmand, S.; Rosenberg, J.; Gögenur, I. Effect of melatonin on depressive symptoms and anxiety in patients undergoing breast cancer surgery: A randomized, double-blind, placebo-controlled trial. Breast Cancer Res. Treat. 2014, 145, 683–695. [Google Scholar] [CrossRef]
Figure 1. PRISMA flow diagram.
Figure 1. PRISMA flow diagram.
Pharmaceutics 15 01616 g001
Table 1. Characteristics of the included trials with breast cancer patients.
Table 1. Characteristics of the included trials with breast cancer patients.
StudyCountrySample SizeMedian AgeMel
mg/day
CTPeriodImproved Outcome
[47]Italy4065–6620 tamoxifen6–7 monthsCT-induced toxicity
Partial response
1-year survival rate
[48]Italy315920 mitoxantrone5–7 monthsCT-induced toxicity
[49]Italy775920 doxorubicin paclitaxel
mitoxantrone
36–12 monthsTumor regression rate
1-year survival rate
CT-induced toxicity a
[50,51]Brazil365420 CT10 daysPain perception
Cognitive function, sleep quality, and depressive symptoms.
[52]Russia54683 toremifene4 monthsSleep quality
[53]Russia53563 NACT6 monthsCT-induced fatigue and sleep quality
[54]Iran7850–46 18 CT and RT25 weeksCT-induced fatigue
[55]Iran3451–52 10 paclitaxel12 weeksNeuropathic chronic pain
CT = chemotherapy; RT = radiotherapy; NACT = anthracycline and taxan-containing neoadjuvant chemotherapy. a Results were not significant in mitoxantrone-treated patients.
Table 2. Quality assessment of randomized clinical trials according to the revised guidelines of the Cochrane collaboration tool RoB 2 for the risk of bias.
Table 2. Quality assessment of randomized clinical trials according to the revised guidelines of the Cochrane collaboration tool RoB 2 for the risk of bias.
Risk Domain/Reference[47][48][49][50][52][53][54][55]
Randomization process++++++++
Deviations from intended interventions++++!!++
Missing outcome data++++++++
Measurement of the outcome++++++++
Selection of the reported result!!!+++++
Overall!!!++!++
Green (+) = low risk; yellow (!) = some concerns.
Table 3. Therapeutic results of preclinical studies and a comparison of the active doses in animal studies and RCTs (the HED extrapolation dose for human adults).
Table 3. Therapeutic results of preclinical studies and a comparison of the active doses in animal studies and RCTs (the HED extrapolation dose for human adults).
Animal ModelRef.FindingsMelatonin Daily DoseAdministration PeriodDaily HED for a 60 kg Adult
Mouse[67]Enhances stabilization rates0.2 mg/kg3 weeks1 mg
[56]Reduces tumor size, cell growth, and angiogenesis2 mg/kg2 weeks10 mg
[74]Diminishes angiogenesis and cell survival2 mg/kg2 weeks
[75]Inhibits mammary carcinogenesis2.5–3.1 mg/kg9 months12–15 mg
[57]Reduces tumor size and formation and metastasis
Longer survival
5 mg/kg10 weeks24 mg
[65]Diminishes cell growth, survival, and metastasis20 mg/kg4 weeks97 mg
[64]Diminishes metastasis and angiogenesis30 mg/kg27 days146 mg
[58]Reduces tumor size and angiogenesis40 mg/kg3 weeks195 mg
[59]Reduces tumor size40 mg/Kg3 weeks
[60]Reduces tumor size40 mg/kg 3 weeks
[40]Reduces tumor size and inhibits cell growth50 mg/kg27 days243 mg
[66]Diminishes metastasis100 mg/kg2/5 weeks486 mg
Rat[68]Antiproliferative and immunomodulatory effects1.2 mg/kg16 weeks12 mg
[61]Decreases tumor frequency and tumor size and extends tumor latency. Antiangiogenic, bone metabolism2.1 mg/kg15 weeks20mg
[69]Decreases tumor frequency and extends latency2.4 mg/kg15 weeks23 mg
[62]Reduces tumor size2.5 mg/kg120 days24 mg
[63]Reduces tumor size
Diminishes metastasis and angiogenesis
10 mg/kg2 weeks97 mg
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ramos, E.; Egea, J.; López-Muñoz, F.; Gil-Martín, E.; Romero, A. Therapeutic Potential of Melatonin Counteracting Chemotherapy-Induced Toxicity in Breast Cancer Patients: A Systematic Review. Pharmaceutics 2023, 15, 1616. https://doi.org/10.3390/pharmaceutics15061616

AMA Style

Ramos E, Egea J, López-Muñoz F, Gil-Martín E, Romero A. Therapeutic Potential of Melatonin Counteracting Chemotherapy-Induced Toxicity in Breast Cancer Patients: A Systematic Review. Pharmaceutics. 2023; 15(6):1616. https://doi.org/10.3390/pharmaceutics15061616

Chicago/Turabian Style

Ramos, Eva, Javier Egea, Francisco López-Muñoz, Emilio Gil-Martín, and Alejandro Romero. 2023. "Therapeutic Potential of Melatonin Counteracting Chemotherapy-Induced Toxicity in Breast Cancer Patients: A Systematic Review" Pharmaceutics 15, no. 6: 1616. https://doi.org/10.3390/pharmaceutics15061616

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop