Next Article in Journal
The Benefits and Challenges of Antibiotics–Non-Steroidal Anti-Inflammatory Drugs Non-Covalent Reaction
Next Article in Special Issue
The Mosquito Larvicidal Activity of Lignans from Branches of Cinnamomum camphora chvar. Borneol
Previous Article in Journal
Boosting Electrocatalytic Oxidation of Formic Acid on Ir(IV)-Doped PdAg Alloy Nanodendrites with Sub-5 nm Branches
Previous Article in Special Issue
Horse Chestnut Saponins–Escins, Isoescins, Transescins, and Desacylescins
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anticancer Potentials of the Lignan Magnolin: A Systematic Review

by
Md. Shimul Bhuia
1,†,
Polrat Wilairatana
2,*,†,
Raihan Chowdhury
1,
Asraful Islam Rakib
1,
Hossam Kamli
3,
Ahmad Shaikh
3,
Henrique D. M. Coutinho
4,* and
Muhammad Torequl Islam
1,*
1
Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
2
Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
3
Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia
4
Department of Biological Chemistry, Regional University of Cariri, Crato 63105-000, CE, Brazil
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2023, 28(9), 3671; https://doi.org/10.3390/molecules28093671
Submission received: 9 March 2023 / Revised: 20 April 2023 / Accepted: 21 April 2023 / Published: 23 April 2023
(This article belongs to the Special Issue Advances in Natural Products and Their Biological Activities)

Abstract

:
Magnolin is a naturally occurring, multi-bioactive lignan molecule with inherent anticancer effects. This study aims to summarize the botanical origins and anticancer properties of magnolin. For this, a recent (as of March 2023) literature review was conducted using various academic search engines, including PubMed, Springer Link, Wiley Online, Web of Science, Science Direct, and Google Scholar. All the currently available information about this phytochemical and its role in various cancer types has been gathered and investigated. Magnolin is a compound found in many different plants. It has been demonstrated to have anticancer activity in numerous experimental models by inhibiting the cell cycle (G1 and G2/M phase); inducing apoptosis; and causing antiinvasion, antimetastasis, and antiproliferative effects via the modulation of several pathways. In conclusion, magnolin showed robust anticancer activity against many cancer cell lines by altering several cancer signaling pathways in various non- and pre-clinical experimental models, making it a promising plant-derived chemotherapeutic option for further clinical research.

1. Introduction

Cancer is a broad collection of illnesses that can begin in practically any organ or tissue of the body [1]. The World Health Organization (WHO) stated that approximately 10 million cancer-related deaths occurred worldwide in 2020, accounting for nearly one-sixth of all deaths. Furthermore, around 400,000 children are diagnosed with cancer each year (https://www.who.int/health-topics/cancer, accessed on 5 March 2023). These diseases occur when atypical cells multiply uncontrollably, invade neighboring tissues beyond their usual boundaries, and metastasize to other organs [2]. The underlying anomaly that leads to the formation of cancer is the persistent and uncontrolled growth of cancerous cells [3]. Cancer cells do not behave in the same way as normal cells, and they do not respond appropriately to the signals that govern normal cell behavior. Instead, cancer cells grow and divide without regulation, invade healthy tissues and organs, and eventually metastasize to other parts of the body [4]. The process of cancer development is seen as a complex series of steps at the cellular level, which includes mutation and selection for cells with enhanced abilities for proliferation, survival, invasion, and metastasis. These steps occur progressively, with cells acquiring more and more of these traits over time [5]. The standard process of the biological system can be changed by daily exposure to dangerous toxicants, which eventually results in genetic damage [6]. The development of cancer is a multifaceted process that occurs over a series of steps, as we know, and as a result, numerous factors may influence the likelihood of cancer development. Therefore, it is overly simplistic to attribute most cancers to a single cause [7]. Despite the complexity of cancer development, several factors, such as radiation, viruses, and chemicals, have been identified to cause cancer in both humans and laboratory animals. These agents have been found to induce cancer through various mechanisms [8,9,10]. A poor diet with insufficient fruits and vegetables and lifestyle choices such as smoking and drinking alcohol can also raise the risk of developing cancer [11,12].
The treatment plan for cancer is determined based on several factors, such as the type and stage of the cancer, as well as the patient’s overall health and age. Treatment options may include surgery, radiation therapy, immunotherapy, chemotherapy, and targeted therapy. Early detection and treatment can elevate the chances of survival and reduce the risk of complications [13,14,15]. For the majority of human cancers, chemotherapy offers variable levels of effectiveness as a treatment while offering curative care for specific cancer types [16,17]. Current anticancer drugs have multiple limitations, including resistance, toxicity, limited efficacy, and cost. Cancer cells can develop resistance to anticancer drugs, which hinders their effectiveness. Additionally, many anticancer drugs cause significant side effects that impact patients’ quality of life. A lack of specificity can also cause healthy cells to be affected, leading to toxicity and side effects [18,19,20]. This is why we need to develop more safe and effective novel anticancer drugs.
Natural remedies are becoming increasingly prevalent worldwide as conventional or supplementary treatments for curable and incurable diseases and for encouraging health and disease prevention [21]. Due to their negligible side effects and good safety records, they are widely used in various diseases as effective therapeutic routes [22]. Lead compounds are abundant in plants and are used in drug discovery. Most (60–70%) of the antibacterial and anticancer medications used in clinical settings are natural compounds or their derivatives [23,24].
Naturally occurring lignans are a group of secondary metabolites that are found in a variety of sources, such as seeds, nuts, cereals, vegetables, and fruits. Lignans serve a range of purposes in plants, and their multifaceted functions are significant for various organisms, including humans [25,26,27]. They are mainly liable for the defense mechanism and are distinguished by the presence of two terminal phenyl groups in their chemical structure [28]. Phytochemicals that belong to the lignan class exhibit diverse biological features, including antioxidant, anti-inflammatory, antiestrogenic, anticarcinogenic, and antitumor activities [29,30]. Several epidemiological studies have suggested that lignans may lower the risk of cardiovascular disease. However, the impact of lignans on other chronic ailments, such as breast cancer, is still debated [31]. Additionally, the dietary intake of lignans has been linked with a diminished risk of other types of cancer, including colon cancer, gastric adenocarcinoma, and esophageal carcinoma. However, there have been very few human studies conducted on this topic [32]. The scientific community has previously mentioned the discovery and development of lignan derivatives for the creation of anticancer therapeutics. Examples of such drugs include etoposide, teniposide, and etopophos [33].
Magnolin is a lignan (C23H28O7, Figure 1), chemically known as (3S,3aR,6S,6aR)-3-(3,4-dimethoxy phenyl)-6-(3,4,5-tri methoxyphenyl)-1,3,3a,4,6,6a-hexahydrofuro[3,4-c] furan, that has been isolated from the Magnolia genus [34,35], which is a large genus of about 210 flowering species in the family Magnoliaceae [36], and has been shown to have health-enhancing properties (Figure 2) in in vivo and in vitro test systems [37,38,39,40,41,42,43,44]. Magnolin exhibits various pharmacological activities, including anti-oxidative, anti-inflammatory, and vasodilatory effects. It has also been found to have protective effects against contrast-induced nephropathy and has been shown to inhibit the migration and invasion of lung cancer cells [41,43]. Additionally, magnolin has been found to be capable of suppressing cell proliferation and transformation by suppressing the ERKs/RSK2 signaling pathways and impairing the G1/S cell-cycle transition [45]. This review focuses on magnolin’s general anticancer mechanism. For this goal, we have compiled the most recent information on this optimistic anticancer agent’s anticancer activity.

2. Results and Discussion

2.1. Database Reports

A total of 163 pieces of evidence were identified through the databases of PubMed and Science Direct, as well as through other sources such as Google Scholar and Research Gate. These databases covered 31.90, 66.10, and 2.00% of the evidence, respectively. Based on the inclusion criteria, this study includes 42 reports. Among them, 7.35% of the reports pertain to the anticancer activities of magnolin, while 18.40% of the reports pertain to its botanical sources, pharmacological activities, and pharmacokinetic features. Among the different cancers, 1.22, 1.22, 1.22, 0.6, 1.22, 1.22, and 0.6% of the cases correspond to breast cancer, lung cancer, ovarian cancer, liver cancer, prostate cancer, colon cancer, and pancreatic carcinoma, respectively. Meanwhile, 1.80% of the reports correspond to mouse models, and 7.35% correspond to in vitro experiments (investigated on different cell lines). After reviewing the titles, abstracts, and contents of the reports, 121 were deemed irrelevant to the study and were subsequently excluded. A logical flowchart for this study is exhibited in Figure 2.

2.2. Botanical Sources

Medicinal plants are considered significant resources for discovering novel drugs worldwide [46,47]. They hold cultural and economic importance to local communities and have been utilized in traditional and popular medicine as well as in the development of pharmaceuticals [48,49]. The commercial and scientific interest in medicinal plants as a source of primary materials for the herbal pharmaceutical industries is increasing rapidly due to the growing global demand [50]. The World Health Organization (WHO) reports that a significant proportion of individuals in developing countries (70–95%) rely on medicinal plants for their primary healthcare needs. However, despite their widespread use, only a small fraction (15%) of medicinal plants worldwide has been analyzed to measure their potential therapeutic benefits based on their phytochemical and phytopharmacological properties [51]. Magnolin, a biologically active compound, has been extracted from plants of the Magnolia genus [35]. Three species of magnolia, including Magnolia denudata, Magnolia sprengeri, and Magnolia biondii, have been recognized in the pharmacopoeia and are known for their therapeutic benefits [42]. The compound source is a component of the flowers, leaves, seeds, roots, and aerial portions of numerous species, according to accounts in the literature. The botanical origins of this lignan are shown in Table 1.

2.3. Cellular and Molecular Anticancer Mechanisms of Magnolin

New precision medicine treatments for cancer are developed by utilizing information obtained from changes that occur at the molecular level in cancer genes and their associated signaling pathways. These days, it is widely acknowledged that signaling pathways and molecular networks play crucial roles in carrying out and regulating vital cellular processes that promote cell survival and growth and are hence largely responsible for the development of cancer as well as its potential treatment [77]. In cancer, two pathways, namely, the PI3K/AKT/mTOR signal transduction pathway and the Ras/MEK pathway, are commonly activated or mutated [78]. These pathways are closely interlinked in transmitting signals from receptor tyrosine kinases (RTKs) to intracellular effector proteins and cell cycle regulators, thus regulating upstream cellular signals [79]. Some other important signaling pathways that develop cancer and can be the target of therapeutic development include cell cycle, Hippo signaling [80], Notch signaling [81], Myc signaling [82,83], oxidative stress response/Nrf2 [84,85], TGFb signaling [86], β-catenin/Wnt signaling [87], and p53 signaling pathways [88]. CDK4/6 inhibitors (CDK4/6i) have made significant advancements in cancer treatment among all the signaling pathways targeting CDK4/6 activation [89].
Due to the growing number of deaths caused by cancer, it is essential to develop therapeutic strategies that have fewer cytotoxic side effects and are less prone to resistance. Natural compounds have been found to possess multiple beneficial activities, such as promoting overall health and providing cancer treatment [90]. Natural products have demonstrated preferential advantages against cancer cells when compared to normal cells. Additionally, their chemical structures can serve as models for the development of innovative drugs [90,91]. Using these models, drugs can be formulated that offer comparable or superior benefits to those provided by natural products. Moreover, these drugs may have fewer side effects and lower chances of resistance compared to the original natural products [92,93].
According to various studies in the documented literature on magnolin’s anticancer action, this therapeutic substance may prevent cancer by inhibiting the cell cycle, preventing metastasis, inducing apoptosis, and suppressing cell proliferation and migration [94] (Figure 3). The effective dose and the mechanism of action may differ depending on the kind of cancer (Table 2). Furthermore, the following provides comprehensive information on magnolin’s anticancer mechanism (Figure 3).

2.4. Magnolin against Various Cancers

2.4.1. Breast Cancer

Breast cancer is the most often diagnosed cancer and a significant cause of mortality in women worldwide [104,105]. It is the second most common cause of cancer-related deaths among women. The pathogenesis of breast cancer is a complex, multi-step process with associated factors such as multiple cell types, and its prevention remains a significant challenge globally [106,107]. It has the ability to spread to other portions of the body, such as the bone, liver, lung, and brain, making it an incurable disease in most cases. However, if the disease is detected early, the prognosis and chances of survival are generally favorable [106,108]. The treatment of breast cancer involves a multidisciplinary approach that encompasses various methods such as locoregional therapy, which involves surgery and radiation therapy, as well as systemic therapy. Systemic therapy consists of several types of treatments, including hormone therapy for chemotherapy, hormone-positive disease, anti-HER2 therapy for HER2-positive disease, and more recently, immunotherapy [109].
According to recent studies, the treatment of MDA-MB-231 breast cancer cells with magnolin suppresses proliferation and invasion and promotes apoptosis. In addition, magnolin significantly downregulated the expression of CDK1, BCL2, MMP2, and MMP9 and upregulated the cleaved caspases 3 and 9 [95]; caspase-9, which plays a crucial role in regulating apoptosis, is involved in various stimuli such as oncogenes, DNA-damaging agents, cellular stress, and apoptotic processes. Moreover, many chemotherapeutic drugs utilize caspase-9 to initiate apoptosis [95,110]. Breast cancer migration can be inhibited by preventing the phosphorylation of ERK in the affected cells [111]. The compound also diminished the phosphorylation of MEK1/2 and extracellular-signal-regulated kinase (ERK)1/2. Magnolin is known to modulate the activity of caspase proteins through the MEK-ERK-BCL2 axis. The ERK protein is situated downstream of MEK and has been identified as a significant factor in influencing tumor growth in various cancer studies [112]. In a prior study, the treatment of MDA-MB-231 breast cancer cell lines with magnolin reduced the bone loss and bone-resorbing activity caused by breast cancer through suppressed mRNA expression and the secretion of MMP-9 and cathepsin K activities. Magnolin also prevented the start and development of the “vicious cycle” between cancer metastases and the bone microenvironment by preventing PTHrP synthesis in osteoclastic bone resorption breast cancer cells [96]. When used against the cancer cell line MCF-7, Vo et al. observed that magnolin showed only moderate cytotoxic effects [57].

2.4.2. Lung Cancer

GLOBOCAN, the International Agency for Research on Cancer, provided cancer statistics in 2020 worldwide, with an appraised 19.3 million new cancer cases, in which lung cancer remained the primary cancer killer [113,114]. The mechanisms and processes that underlie the development of lung cancer are intricate and not entirely comprehended [115]. The primary reason for lung cancer is tobacco smoking, and about 80% to 90% of cases are attributed to smoking cigarettes [116]. Continued exposure can cause genetic mutations, which interfere with protein synthesis and disrupt the cell cycle, ultimately promoting the development of cancer. BCL2, MYC, and p53 are the most common genetic mutations linked with small-cell lung cancer, while non-small-cell lung cancer is most commonly caused by mutations in EGFR, KRAS, and p16 [115,117,118].
The ERKs/RSK2 signaling pathway is a promising target for cancer research and therapy, as it plays a critical role in cancer cell survival, proliferation, and invasion. The inhibition of this pathway promotes cancer cell death and enhances the effectiveness of chemotherapy [119]. Additionally, targeting this pathway may have potential in cancer prevention, as it is involved in cancer initiation and progression. Overall, exploring the therapeutic potential of the ERKs/RSK2 pathway may lead to the development of effective cancer treatments and preventive measures [97,119]. Recent research has shown that magnolin can increase lung cancer cells’ chemosensitivity by preventing cell proliferation, migration, and invasion. Magnolin inhibits cell migration by inhibiting ERKs/RSK2 signaling, which NF-κB-mediated Cox-2 gene expression enhances. Moreover, findings have shown that magnolin suppressed the activity of MMP-2 and MMP-9 [97], two crucial enzymes involved in focal adhesion and cell migration [120]. Lee and colleagues showed in their previous study that magnolin prevented the phosphorylation of RSK2 and its downstream targets, including c-Jun, ATF1, and AP-1, which is a dimer composed of Jun and Fos proteins. Additionally, magnolin’s ability to block ERKs hindered EGF-induced anchorage-independent cell transformation and colony expansion of A549 human lung cancer cells and NIH3T3 cells expressing Ras (G12V) when grown in soft agar [98]. So, targeting the ERKs/RSK2 signaling pathway is an important approach for developing chemoprevention and cancer therapy.

2.4.3. Liver Cancer

Regarding cancer-related mortality among men, liver cancer is the third most common [121]. It is a malignant tumor that often develops in individuals who have a history of long-term liver disease and cirrhosis, and it is characterized by its aggressive nature [122]. In cancer, numerous signaling pathways undergo changes, and in liver cancer specifically, certain pathways are observed to be disrupted. The primary pathways that play a role in the development of hepatocellular carcinoma (HCC) are those that regulate the signaling of growth factors such as insulin-like growth factor (IGF), epidermal growth factor (EGF), platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), and hepatocyte growth factor (HGF/MET) [123]. The Ras/Raf/MEK/ERK and PI3K/AKT/mTOR cascades are the primary signaling pathways that are activated after the activation of receptor tyrosine kinases [124]. Tumor development, cell cycle progression, apoptosis, and survival depend on the PI3K/AKT/mTOR signaling system [54,125,126]. According to several studies, over 50% of hepatocellular carcinoma patients have an active phosphatidylinositol-3 kinase PI3K/AKT/mTOR pathway [127,128]. The PI3K-AKT/mTOR and ERK/MEK pathways were revealed to be the targets of magolin’s synergistic action with the BRAF inhibitor (SB590885) in halting the progression of liver cancer (Bel-7402 and SK-Hep1) cells in vitro [99], which are the best characterized and most frequently activated intracellular pathways in liver cancer [129]. Additionally, Llovet et al. (2008) stated that impeding the MEK/ERK pathway can lead to a reduction in cancer cell proliferation [130].

2.4.4. Ovarian Cancer

Most deaths from gynecologic cancers are caused by ovarian cancer, and it is one of the most prevalent female malignancies [131,132]. Age, early menarche, null parity, late menopause, environment, and family history are risk factors [133,134,135], out of which, genetic and epigenetic factors are deemed the most significant among all the factors [136], but breastfeeding and pregnancy reduce this risk [137,138]. Individuals afflicted with this deadly illness have a mere 45.6% 5-year survival rate [139]. Patients with advanced ovarian cancer have the worst prognosis regarding curative therapeutic options, with cytoreductive surgery and cisplatin-based chemotherapy as the cornerstones of care. However, many patients show intrinsic or acquired resistance to cisplatin-based treatment regimens [140,141,142]. In this situation, magnolin therapy reduced the accumulation of G1 and G2/M cell cycle phases and the activation of ERK1/2 in TOV-112D ovarian cancer cells, resulting in the suppression of the ERK1/2-MEK pathway, a vital signaling mechanism that facilitates ligand-stimulated signals by inhibiting the initiation of cell proliferation, differentiation, and cell survival [143]. Of particular note, magnolin prevented the colony formation of TOV-112D cells on soft agar. Furthermore, in xenograft tissues, magnolin increased the proteins p16Ink4a and p27Kip1, which are essential indicators of cellular senescence [45]. Like this, magnolin, which inhibits ERK1 and ERK2 with IC50 values of 16 and 68 nM, restrained ovarian cancer TOV-112D cell proliferation and colony expansion. Notably, cellular senescence was induced in TOV-112 when magnolin administration reduced the phosphorylation of ATF-1 at Ser63, a downstream target of RSK2 [100].

2.4.5. Prostate Cancer

Prostate cancer (PC) is the second most commonly diagnosed cancer and the second leading cause of cancer-associated deaths in men after lung cancer [144,145]. It is a complex and diverse type of cancer that can be categorized based on aggressiveness, grade, and age of onset. Exogenous factors such as diet, sexual behavior, physical activity, and occupation may influence the progression of the disease, but only three established risk factors are ethnicity, age, and heredity (first-line relatives or relatives with early-onset PC [146]. Magnolin has an IC50 value of 0.51 μM and is a potent antiproliferative agent against PANC-1 prostate cancer cells. Additionally, it can cause the PANC-1 cell to form suppressive colonies in a concentration-dependent manner. Furthermore, the PANC-1 cell was subjected to an inhibitory effect on migration caused by MMP3. Additionally, magnolin was substantially less hazardous to normal human cells than the growth-inhibiting positive control medication doxorubicin (6.96% and 30.48%, respectively, at 5 g/mL) [76]. In a prior study, magnolin prevented the growth and survival of the tumor in PC3 and Du145 prostate cancer cells by causing cell cycle arrest through P53/P21 activation and inducing apoptosis both in vitro and in vivo. Additionally, magnolin reduced the activation of Akt protein kinase and increased the expression of cleaved caspase3 to inhibit cell growth and promote death [43].

2.4.6. Pancreatic Carcinoma

Pancreatic cancer is a form of carcinoma that originates from the cells lining the pancreatic duct, specifically known as pancreatic ductal carcinoma [147]. It is currently the fourth leading cause of cancer-associated deaths. However, due to its aggressive nature, it is expected to become the second most prevalent cause of cancer-associated deaths by 2030 [148]. The cytotoxic properties of lignans isolated from the plant Zanthoxylum alatum were investigated in pancreatic carcinoma cell lines (MIA-PaCa) and lung carcinoma cell lines (A549) using the MTT assay. The results indicated that 4′-O-demethyl magnolin extracted from Zanthoxylum alatum exhibited remarkable cytotoxic activity against these cell lines [101].

2.4.7. Colorectal Cancer

Colorectal cancer is among the most commonly diagnosed malignancies worldwide, and it is one of the leading causes of cancer-associated death [149,150]. Several risk factors, including age, family history, diet, lifestyle, obesity, alcohol intake, and smoking, influence the occurrence of colorectal cancer. Environmental variables may also significantly impact the development of colorectal cancer [151,152,153]. The treatment of colorectal cancer is being revolutionized by cancer immunotherapy, which employs immune checkpoint inhibitors to empower the body’s immune system to more effectively detect and eradicate cancerous cells [154]. Autophagy improves the efficacy of immunotherapy by facilitating the optimal release of immunostimulatory signals. This is achieved through the delivery of antigens to immune cells, such as antigen-presenting cells and CD8+ cytotoxic T lymphocytes [155]. Magnolin inhibits cell growth and causes cell cycle arrest in HCT116 and HT29 cells in the G2/M phase [102]. Similarly, by encouraging autophagy and cell cycle arrest via the LIF/Stat3/Mcl-1 pathway, magnolin inhibits the formation of tumors in HCT116 and SW480 colorectal cancer cells in vitro and in vivo without exhibiting any observable adverse effects [103]. Recent data from various sources also indicate that controlling autophagy may offer crucial insights into halting the growth and spread of cancer and improving cancer treatments [156,157,158]. The biological efficacy of lignans, which includes magnolin derived from the aboveground portions of Artemisia gorgonum Webb, has been examined to determine their ability to destroy cells in different types of mammalian tumor cell lines and in normal murine cells (CFU-GM). The findings showed that magnolin exhibited only slight potency and selectivity against murine colon, and there was no significant evidence of its ability to destroy human tumor cells [44].

2.5. Pharmacokinetic Features

Pharmacokinetics (PKs) plays a crucial role in drug discovery by helping to improve the absorption, distribution, metabolism, and excretion (ADME) properties of potential drug candidates. The ultimate aim is to create a clinical candidate that achieves the desired efficacy and safety profile by attaining a concentration–time profile in the body that is appropriate [159,160,161]. Assessing the PKs of herbal medicinal drugs, including the extent of oral bioavailability, can help to establish a connection between pharmacological data and clinical effects. It can also facilitate the development of appropriate dosage regimens by providing valuable information for rational drug design [160,162]. Pharmacokinetics and pharmacogenetics are significant factors in drug discovery that impact the success of treatment. In the case of cancer treatment, these factors are crucial due to the elevated levels of toxicity associated with many cancer medications. Therefore, comprehending how drugs are metabolized, how they interact with their targets, and how genetic variations can impact drug response is crucial for developing effective and safe treatments for cancer [163]. Various in vivo studies revealed the PK properties necessary to understand the ADME of magnolin.
In 2010, Kim and colleagues first conducted an experiment using live rats to investigate the PKs of magnolin, which is the major active phytochemical in Magnolia fargesii. They administered the compound intravenously and orally in various doses and found that the oral bioavailability ranged from 54.3% to 76.4% for the doses they tested. They also discovered that magnolin bound strongly to plasma proteins in rats, with a consistent binding value of 71.3% to 80.5% across a concentration range of 500 to 10,000 ng/mL. The researchers found that the PKs of magnolin were not affected by the dosage, regardless of whether it was administered intravenously or orally [164]. The process of drug metabolism is a critical aspect of pharmacokinetics and plays a significant role in drug development [165]. Studies have shown that in rats, magnolin undergoes metabolism to produce 4’-O-desmethyl magnolin, 4″-O-desmethyl magnolin, and their respective glucuronides [166]. Another study reported that Aspergillus niger selectively metabolizes magnolin to produce 4’-desmethyl magnolin and 4’-desmethyl-5’-hydroxymagnolin [167]. According to Kim et al. (2011), magnolin is predicted to be metabolized into its oxidative metabolites by several CYP enzymes, including CYP2C8, CYP2C19, CYP2C9, and CYP3A4. As a result, there is potential for drug interactions when co-administered with drugs that can inhibit or diminish CYP2C enzymes [168]. In cases of elimination, magnolin is rapidly absorbed when administered orally and is almost entirely eliminated through metabolic processes [164].

3. Methodology

3.1. Literature Searching Strategy

An up-to-date (until March 2023) search was made in the various databases, such as PubMed, Springer Link, Wiley Online, Web of Science, ScienceDirect, and Google Scholar, with the common keyword “magnolin”, which was then paired with “cancer,” “tumor,” “anticancer activity,” “antiproliferation activity,” “cytotoxic activity,” “antitumor activity,” “human cancer,” “biological activities,” “pharmacological effects,” “pharmacological activities,” “biological sources,” “chemical features,” “in vivo studies”, or “in vitro studies “. No time and language restrictions were imposed in the search criteria. The studies were evaluated in-depth, with information regarding the sources, dose/concentration, test system, proposed action mechanism, and overall conclusion and recommendation.

3.2. Inclusion Criteria

  • Studies carried out in vitro, ex vivo, or in vivo with or without utilizing laboratory animals, including mice, rats, rabbits, and humans, and their derived tissues or cells.
  • Studies with anticancer activities and botanical sources of magnolin.
  • Studies with magnolin or its derivatives or preparations.
  • Magnolin or its derivatives provide joint activity with other chemical compounds.
  • Studies which do or do not imply possible mechanisms of action.

3.3. Exclusion Criteria

  • Studies demonstrated data duplication and titles and/or abstracts not meeting the inclusion criteria.
  • Magnolin in other studies not covering the current issue.
  • Papers written in languages other than English.
  • Studies without full text available.
  • Case reports, letters, editorials, and commentaries

4. Conclusions

Cancer remains one of the most fearsome diseases in the world, causing a significant number of deaths every year. Among the other strategies, chemotherapeutic approaches are considered one of the most potential and effective therapeutic modalities in cancer. Natural products and their derivatives are the potential sources of anticancer lead compounds. This review suggests that, to date, magnolin has been isolated from 23 medicinal plants under 13 genera. Magnolin between 10 to 125 μM showed anticancer effects against breast, lung, liver, ovarian, prostate, and colon cancers, while its derivative, 4′-O-demethyl magnolin, is evidently effective against lung and pancreatic cancers. Magnolin exerts anticancer effects against various cancer cell lines or systems through diverse mechanisms that include cytotoxicity, apoptotic cell death, cell cycle arrest, antimigratory, antiproliferative, and anti-invasion pathways. However, according to the database reports, more work has been done on its anticancer effects against breast and lung cancers. The cytotoxicity, apoptosis, and antiproliferative effects of magnolin were predominant among the anticancer mechanisms. More research is necessary on this hopeful anticancer lead compound.

Author Contributions

Data search and writing (equal contribution), M.S.B. and R.C.; methodology, data search, A.I.R., H.K. and A.S.; conceptualization, design, and final editing, M.T.I., P.W. and H.D.M.C. All authors have read and agreed to the published version of the manuscript.

Funding

The research did not receive any financial support from external sources.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The processed data are available from the corresponding author upon request.

Acknowledgments

The authors extend their appreciation to the Deanship of Scientific Research at King Khalid University, Abha, Saudi Arabia, for funding this work through small group Research Project under grant number RGP.1/191/43.

Conflicts of Interest

The authors state that they have no conflict of interest to disclose.

References

  1. Upadhyay, A. Cancer: An unknown territory; rethinking before going ahead. Genes Dis. 2021, 8, 655–661. [Google Scholar] [CrossRef]
  2. Jones, P.A.; Baylin, S.B. The epigenomics of cancer. Cell 2007, 128, 683–692. [Google Scholar] [CrossRef]
  3. Evan, G.I.; Vousden, K.H. Proliferation, cell cycle and apoptosis in cancer. Nature 2001, 411, 342–348. [Google Scholar] [CrossRef] [PubMed]
  4. Zink, D.; Fischer, A.H.; Nickerson, J.A. Nuclear structure in cancer cells. Nat. Rev. Cancer 2004, 4, 677–687. [Google Scholar] [CrossRef]
  5. Farber, E.; Rubin, H. Cellular adaptation in the origin and development of cancer. Cancer Res. 1991, 51, 2751–2761. [Google Scholar] [PubMed]
  6. Wu, X.; Cobbina, S.J.; Mao, G.; Xu, H.; Zhang, Z.; Yang, L. A review of toxicity and mechanisms of individual and mixtures of heavy metals in the environment. Environ. Sci. Pollut. Res. 2016, 23, 8244–8259. [Google Scholar] [CrossRef]
  7. Armstrong, B.; Doll, R. Environmental factors and cancer incidence and mortality in different countries, with special reference to dietary practices. Int. J. Cancer 1975, 15, 617–631. [Google Scholar] [CrossRef] [PubMed]
  8. Javier, R.T.; Butel, J.S. The history of tumor virology. Cancer Res. 2008, 68, 7693–7706. [Google Scholar] [CrossRef] [PubMed]
  9. Rakoff-Nahoum, S. Cancer issue: Why cancer and inflammation? Yale J. Biol. Med. 2006, 79, 123. [Google Scholar]
  10. Trichopoulos, D.; Li, F.P.; Hunter, D.J. What causes cancer? Sci. Am. 1996, 275, 80–87. [Google Scholar] [CrossRef]
  11. Ng, R.; Sutradhar, R.; Yao, Z.; Wodchis, W.P.; Rosella, L.C. Smoking, drinking, diet and physical activity—Modifiable lifestyle risk factors and their associations with age to first chronic disease. Int. J. Epidemiol. 2020, 49, 113–130. [Google Scholar] [CrossRef]
  12. Ames, B.N.; Gold, L.S.; Willett, W.C. The causes and prevention of cancer. Proc. Natl. Acad. Sci. USA 1995, 92, 5258–5265. [Google Scholar] [CrossRef] [PubMed]
  13. Zeng, Y. Advances in mechanism and treatment strategy of cancer. Cell. Mol. Biol. 2018, 64, 1–3. [Google Scholar] [CrossRef]
  14. Mansouri, V.; Beheshtizadeh, N.; Gharibshahian, M.; Sabouri, L.; Varzandeh, M.; Rezaei, N. Recent advances in regenerative medicine strategies for cancer treatment. Biomed. Pharmacother. 2021, 141, 111875. [Google Scholar] [CrossRef]
  15. Maher, E.; Coia, L.; Duncan, G.; Lawton, P. Treatment strategies in advanced and metastatic cancer: Differences in attitude between the USA, Canada and Europe. Int. J. Radiat. Oncol. Biol. Phys. 1992, 23, 239–244. [Google Scholar] [CrossRef]
  16. Furue, H. Chemotherapy cancer treatment during the past sixty years. Gan Kagaku Ryoho Cancer Chemother. 2003, 30, 1404–1411. [Google Scholar]
  17. Ruggeri, B.A.; Camp, F.; Miknyoczki, S. Animal models of disease: Pre-clinical animal models of cancer and their applications and utility in drug discovery. Biochem. Pharmacol. 2014, 87, 150–161. [Google Scholar] [CrossRef]
  18. Martinez-Outschoorn, U.E.; Peiris-Pagés, M.; Pestell, R.G.; Sotgia, F.; Lisanti, M.P. Cancer metabolism: A therapeutic perspective. Nat. Rev. Clin. Oncol. 2017, 14, 11–31. [Google Scholar] [CrossRef]
  19. Pommier, Y.; Leo, E.; Zhang, H.; Marchand, C. DNA topoisomerases and their poisoning by anticancer and antibacterial drugs. Chem. Biol. 2010, 17, 421–433. [Google Scholar] [CrossRef] [PubMed]
  20. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  21. Zhang, A.; Sun, H.; Wang, X. Recent advances in natural products from plants for treatment of liver diseases. Eur. J. Med. Chem. 2013, 63, 570–577. [Google Scholar] [CrossRef]
  22. Sasidharan, S.; Chen, Y.; Saravanan, D.; Sundram, K.; Latha, L.Y. Extraction, isolation and characterization of bioactive compounds from plants’ extracts. Afr. J. Tradit. Complement. Altern. Med. 2011, 8, 1–10. [Google Scholar] [CrossRef] [PubMed]
  23. Ebob, O.T.; Babiaka, S.B.; Ntie-Kang, F. Natural products as potential lead compounds for drug discovery against SARS-CoV-2. Nat. Prod. Bioprospecting 2021, 11, 611–628. [Google Scholar] [CrossRef] [PubMed]
  24. Sikder, L.; Khan, M.R.; Smrity, S.Z.; Islam, M.T.; Khan, S.A. Phytochemical and pharmacological investigation of the ethanol extract of Byttneria pilosa Roxb. Clin. Phytosci. 2022, 8, 1–8. [Google Scholar] [CrossRef]
  25. Teponno, R.B.; Kusari, S.; Spiteller, M. Recent advances in research on lignans and neolignans. Nat. Prod. Rep. 2016, 33, 1044–1092. [Google Scholar] [CrossRef]
  26. Ražná, K.; Nôžková, J.; Vargaová, A.; Harenčár, Ľ.; Bjelková, M. Biological functions of lignans in plants. Agriculture 2021, 67, 155–165. [Google Scholar] [CrossRef]
  27. Magoulas, G.E.; Papaioannou, D. Bioinspired syntheses of dimeric hydroxycinnamic acids (lignans) and hybrids, using phenol oxidative coupling as key reaction, and medicinal significance thereof. Molecules 2014, 19, 19769–19835. [Google Scholar] [CrossRef]
  28. Kalinová, J.P.; Marešová, I.; Tříska, J.; Vrchotová, N. Distribution of lignans in Panicum miliaceum, Fagopyrum esculentum, Fagopyrum tataricum, and Amaranthus hypochondriacus. J. Food Compos. Anal. 2022, 106, 104283. [Google Scholar] [CrossRef]
  29. Ionkova, I. Anticancer lignans-from discovery to biotechnology. Mini Rev. Med. Chem. 2011, 11, 843–856. [Google Scholar] [CrossRef]
  30. Durazzo, A.; Zaccaria, M.; Polito, A.; Maiani, G.; Carcea, M. Lignan content in cereals, buckwheat and derived foods. Foods 2013, 2, 53–63. [Google Scholar] [CrossRef]
  31. Peterson, J.; Dwyer, J.; Adlercreutz, H.; Scalbert, A.; Jacques, P.; McCullough, M.L. Dietary lignans: Physiology and potential for cardiovascular disease risk reduction. Nutr. Rev. 2010, 68, 571–603. [Google Scholar] [CrossRef]
  32. Rodríguez-García, C.; Sánchez-Quesada, C.; Toledo, E.; Delgado-Rodríguez, M.; Gaforio, J.J. Naturally lignan-rich foods: A dietary tool for health promotion? Molecules 2019, 24, 917. [Google Scholar] [CrossRef] [PubMed]
  33. Su, G.-Y.; Wang, K.-W.; Wang, X.-Y.; Wu, B. Bioactive lignans from Zanthoxylum planispinum with cytotoxic potential. Phytochem. Lett. 2015, 11, 120–126. [Google Scholar] [CrossRef]
  34. Miyazawa, M.; Kasahara, H.; Kameoka, H. Phenolic lignans from flower buds of Magnolia fargesii. Phytochemistry 1992, 31, 3666–3668. [Google Scholar] [CrossRef]
  35. Pan, J.-X.; Hensens, O.D.; Zink, D.L.; Chang, M.N.; Hwang, S.-B. Lignans with platelet activating factor antagonist activity from Magnolia biondii. Phytochemistry 1987, 26, 1377–1379. [Google Scholar] [CrossRef]
  36. Lee, Y.D. Use of Magnolia (Magnolia grandiflora) Seeds in Medicine, and Possible Mechanisms of Action. In Nuts and Seeds in Health and Disease Prevention; Elsevier: Amsterdam, The Netherlands, 2011; pp. 727–732. [Google Scholar]
  37. Kim, J.Y.; Lim, H.J.; Kim, J.S.; Lee, H.J.; Kim, H.D.; Jeon, R.; Ryu, J.-H. In vitro anti-inflammatory activity of lignans isolated from Magnolia fargesii. Bioorganic Med. Chem. Lett. 2009, 19, 937–940. [Google Scholar] [CrossRef]
  38. Ibarra-Alvarado, C.; Rojas, A.; Mendoza, S.; Bah, M.; Gutiérrez, D.; Hernández-Sandoval, L.; Martínez, M. Vasoactive and antioxidant activities of plants used in Mexican traditional medicine for the treatment of cardiovascular diseases. Pharm. Biol. 2010, 48, 732–739. [Google Scholar] [CrossRef] [PubMed]
  39. Wang, F.; Zhang, G.; Zhou, Y.; Gui, D.; Li, J.; Xing, T.; Wang, N. Magnolin protects against contrast-induced nephropathy in rats via antioxidation and antiapoptosis. Oxidative Med. Cell. Longev. 2014, 2014, 203458. [Google Scholar] [CrossRef]
  40. Uto, T.; Tung, N.H.; Ohta, T.; Shoyama, Y. (+)-Magnolin enhances melanogenesis in melanoma cells and three-dimensional human skin equivalent; involvement of PKA and p38 MAPK signaling pathways. Planta Med. 2022, 88, 1199–1208. [Google Scholar] [CrossRef]
  41. Ma, P.; Che, D.; Zhao, T.; Zhang, Y.; Li, C.; An, H.; Zhang, T.; He, H. Magnolin inhibits IgE/Ag-induced allergy in vivo and in vitro. Int. Immunopharmacol. 2019, 76, 105867. [Google Scholar] [CrossRef]
  42. Shen, Y.; Pang, E.C.; Xue, C.C.; Zhao, Z.; Lin, J.; Li, C.G. Inhibitions of mast cell-derived histamine release by different Flos Magnoliae species in rat peritoneal mast cells. Phytomedicine 2008, 15, 808–814. [Google Scholar] [CrossRef] [PubMed]
  43. Huang, Y.; Zou, X.; Zhang, X.; Wang, F.; Zhu, W.; Zhang, G.; Xiao, J.; Chen, M. Magnolin inhibits prostate cancer cell growth in vitro and in vivo. Biomed. Pharmacother. 2017, 87, 714–720. [Google Scholar] [CrossRef] [PubMed]
  44. Ortet, R.; Prado, S.; Regalado, E.L.; Valeriote, F.A.; Media, J.; Mendiola, J.; Thomas, O.P. Furfuran lignans and a flavone from Artemisia gorgonum Webb and their in vitro activity against Plasmodium falciparum. J. Ethnopharmacol. 2011, 138, 637–640. [Google Scholar] [CrossRef] [PubMed]
  45. Song, J.H.; Lee, C.J.; An, H.J.; Yoo, S.M.; Kang, H.C.; Lee, J.Y.; Kim, K.D.; Kim, D.J.; Lee, H.S.; Cho, Y.Y. Magnolin targeting of ERK1/2 inhibits cell proliferation and colony growth by induction of cellular senescence in ovarian cancer cells. Mol. Carcinog. 2019, 58, 88–101. [Google Scholar] [CrossRef] [PubMed]
  46. Chen, S.-L.; Yu, H.; Luo, H.-M.; Wu, Q.; Li, C.-F.; Steinmetz, A. Conservation and sustainable use of medicinal plants: Problems, progress, and prospects. Chin. Med. 2016, 11, 37. [Google Scholar] [CrossRef] [PubMed]
  47. Nalawade, S.M.; Sagare, A.P.; Lee, C.-Y.; Kao, C.-L.; Tsay, H.-S. Studies on tissue culture of Chinese medicinal plant resources in Taiwan and their sustainable utilization. Bot. Bull. Acad. Sin. 2003, 44, 79–98. [Google Scholar]
  48. Kaky, E.; Gilbert, F. Using species distribution models to assess the importance of Egypt’s protected areas for the conservation of medicinal plants. J. Arid Environ. 2016, 135, 140–146. [Google Scholar] [CrossRef]
  49. Jamshidi-Kia, F.; Lorigooini, Z.; Amini-Khoei, H. Medicinal plants: Past history and future perspective. J. Herbmed Pharmacol. 2017, 7, 1–7. [Google Scholar] [CrossRef]
  50. Bieski, I.G.C.; Leonti, M.; Arnason, J.T.; Ferrier, J.; Rapinski, M.; Violante, I.M.P.; Balogun, S.O.; Pereira, J.F.C.A.; Figueiredo, R.d.C.F.; Lopes, C.R.A.S. Ethnobotanical study of medicinal plants by population of valley of Juruena region, legal Amazon, Mato Grosso, Brazil. J. Ethnopharmacol. 2015, 173, 383–423. [Google Scholar] [CrossRef]
  51. De Luca, V.; Salim, V.; Atsumi, S.M.; Yu, F. Mining the biodiversity of plants: A revolution in the making. Science 2012, 336, 1658–1661. [Google Scholar] [CrossRef]
  52. Chen, C.; Huang, Y.; Chen, H.; Chen, Y.; Hsu, H. On the Ca++-Antagonistic Principles of the Flower Buds of Magnolia fargesii1. Planta Med. 1988, 54, 438–440. [Google Scholar] [CrossRef] [PubMed]
  53. Peng, C.-Y.; Liu, Y.-Q.; Deng, Y.-H.; Wang, Y.-H.; Zhou, X.-J. Lignans from the bark of Zanthoxylum simulans. J. Asian Nat. Prod. Res. 2015, 17, 232–238. [Google Scholar] [CrossRef] [PubMed]
  54. Feng, T.; Zheng, L.; Liu, F.; Xu, X.; Mao, S.; Wang, X.; Liu, J.; Lu, Y.; Zhao, W.; Yu, X. Growth factor progranulin promotes tumorigenesis of cervical cancer via PI3K/Akt/mTOR signaling pathway. Oncotarget 2016, 7, 58381. [Google Scholar] [CrossRef] [PubMed]
  55. Zhao, W.; Zhou, T.; Fan, G.; Chai, Y.; Wu, Y. Isolation and purification of lignans from Magnolia biondii Pamp by isocratic reversed-phase two-dimensional liquid chromatography following microwave-assisted extraction. J. Sep. Sci. 2007, 30, 2370–2381. [Google Scholar] [CrossRef]
  56. Ma, Y.; Han, G. Biologically active lignins from Magnolia biondii Pamp. Zhongguo Zhongyao Zazhi China J. Chin. Mater. Med. 1995, 20, 102–104. [Google Scholar]
  57. Vo, T.N.; Nguyen, P.L.; Tuong, L.T.; Pratt, L.M.; Vo, P.N.; Nguyen, K.P.P.; Nguyen, N.S. Lignans and triterpenes from the root of Pseuderanthemum carruthersii var. atropurpureum. Chem. Pharm. Bull. 2012, 60, 1125–1133. [Google Scholar] [CrossRef]
  58. Hong, P.T.L.; Kim, H.J.; Kim, W.K.; Nam, J.H. Flos magnoliae constituent fargesin has an anti-allergic effect via ORAI1 channel inhibition. Korean J. Physiol. Pharmacol. Off. J. Korean Physiol. Soc. Korean Soc. Pharmacol. 2021, 25, 251–258. [Google Scholar] [CrossRef]
  59. Arango-De la Pava, L.D.; Zamilpa, A.; Trejo-Espino, J.L.; Domínguez-Mendoza, B.E.; Jiménez-Ferrer, E.; Pérez-Martínez, L.; Trejo-Tapia, G. Synergism and subadditivity of verbascoside-lignans and-iridoids binary mixtures isolated from castilleja tenuiflora benth. on NF-κB/AP-1 inhibition activity. Molecules 2021, 26, 547. [Google Scholar] [CrossRef]
  60. Radulović, N.S.; Mladenović, M.Z.; Ðorđević, N.D. Chemotypification of Astrantia major L. (Apiaceae): Essential-Oil and Lignan Profiles of Fruits. Chem. Biodivers. 2012, 9, 1320–1337. [Google Scholar] [CrossRef]
  61. Qiao, D.; Gan, L.-S.; Mo, J.-X.; Zhou, C.-X. Chemical constituents of Acorus calamus. Zhongguo Zhong Yao Za Zhi China J. Chin. Mater. Med. 2012, 37, 3430–3433. [Google Scholar]
  62. Chávez, D.; Acevedo, L.A.; Mata, R. Tryptamine derived amides and acetogenins from the seeds of Rollinia mucosa. J. Nat. Prod. 1999, 62, 1119–1122. [Google Scholar] [CrossRef]
  63. Jo, Y.-H.; Seo, G.-U.; Yuk, H.-G.; Lee, S.-C. Antioxidant and tyrosinase inhibitory activities of methanol extracts from Magnolia denudata and Magnolia denudata var. purpurascens flowers. Food Res. Int. 2012, 47, 197–200. [Google Scholar] [CrossRef]
  64. Seo, Y. Antioxidant activity of the chemical constituents from the flower buds of Magnolia denudata. Biotechnol. Bioprocess Eng. 2010, 15, 400–406. [Google Scholar] [CrossRef]
  65. Li, J.; Tanaka, M.; Kurasawa, K.; Ikeda, T.; Nohara, T. Studies of the chemical constituents of the flower buds of Magnolia kobus and M. salicifolia. J. Nat. Med. 2007, 61, 222–223. [Google Scholar] [CrossRef]
  66. Lee, H.J.; Seo, S.M.; Lee, O.K.; Jo, H.J.; Kang, H.Y.; Choi, D.H.; Paik, K.H.; Khan, M. Lignans from the bark of Magnolia kobus. Helv. Chim. Acta 2008, 91, 2361–2366. [Google Scholar] [CrossRef]
  67. Alegrio, L.V.; Fo, R.B.; Gottlieb, O.R.; Maia, J.G.S. Lignans and neolignans from Licaria armeniaca. Phytochemistry 1981, 20, 1963–1965. [Google Scholar] [CrossRef]
  68. Barbosa-Filho, J.M.; Yoshida, M.; Gottlieb, O.R. Neolignans from the fruits of Licaria armeniaca. Phytochemistry 1986, 26, 319–321. [Google Scholar] [CrossRef]
  69. Ahmed, A.A.; Mahmoud, A.A.; Ali, E.T.; Tzakou, O.; Couladis, M.; Mabry, T.J.; Gáti, T.; Tóth, G. Two highly oxygenated eudesmanes and 10 lignans from Achillea holosericea. Phytochemistry 2002, 59, 851–856. [Google Scholar] [CrossRef]
  70. Dutra, L.M.; Costa, E.V.; de Souza Moraes, V.R.; de Lima Nogueira, P.C.; Vendramin, M.E.; Barison, A.; do Nascimento Prata, A.P. Chemical constituents from the leaves of Annona pickelii (Annonaceae). Biochem. Syst. Ecol. 2012, 41, 115–118. [Google Scholar] [CrossRef]
  71. Wu, H.-B.; Liu, T.-T.; Zhang, Z.-X.; Wang, W.-S.; Zhu, W.-W.; Li, L.-F.; Li, Y.-R.; Chen, X. Leaves of Magnolia liliflora Desr. as a high-potential by-product: Lignans composition, antioxidant, anti-inflammatory, anti-phytopathogenic fungal and phytotoxic activities. Ind. Crops Prod. 2018, 125, 416–424. [Google Scholar] [CrossRef]
  72. Trifunović, S.; Vajs, V.; Tešević, V.; Djoković, D.; Milosavljević, S. Lignans from the plant species Achillea lingulata. J. Serb. Chem. Soc. 2003, 68, 277–280. [Google Scholar] [CrossRef]
  73. Öksüz, S.; Ulubelen, A.; Tuzlaci, E. Constituents of Achillea teretifolia. Fitoterapia 1990, 61, 283. [Google Scholar]
  74. Jain, N.; Srivastava, S.; Aggarwal, K.; Ramesh, S.; Kumar, S. Essential oil composition of Zanthoxylum alatum seeds from northern India. Flavour Fragr. J. 2001, 16, 408–410. [Google Scholar] [CrossRef]
  75. Nishino, C.; Mitsui, T. Lignans from Hernandia Ovigera linn. Tetrahedron Lett. 1973, 14, 335–338. [Google Scholar] [CrossRef]
  76. Sayed, A.M.; El-Hawary, S.S.; Abdelmohsen, U.R.; Ghareeb, M.A. Antiproliferative potential of Physalis peruviana-derived magnolin against pancreatic cancer: A comprehensive in vitro and in silico study. Food Funct. 2022, 13, 11733–11743. [Google Scholar] [CrossRef]
  77. Yip, H.Y.K.; Papa, A. Signaling pathways in cancer: Therapeutic targets, combinatorial treatments, and new developments. Cells 2021, 10, 659. [Google Scholar] [CrossRef]
  78. Vogelstein, B.; Kinzler, K.W. Cancer genes and the pathways they control. Nat. Med. 2004, 10, 789–799. [Google Scholar] [CrossRef]
  79. Pons-Tostivint, E.; Thibault, B.; Guillermet-Guibert, J. Targeting PI3K signaling in combination cancer therapy. Trends Cancer 2017, 3, 454–469. [Google Scholar] [CrossRef]
  80. Pan, D. The hippo signaling pathway in development and cancer. Dev. Cell 2010, 19, 491–505. [Google Scholar] [CrossRef]
  81. Callahan, R.; Egan, S.E. Notch signaling in mammary development and oncogenesis. J. Mammary Gland. Biol. Neoplasia 2004, 9, 145–163. [Google Scholar] [CrossRef]
  82. Xu, J.; Chen, Y.; Olopade, O.I. MYC and breast cancer. Genes Cancer 2010, 1, 629–640. [Google Scholar] [CrossRef] [PubMed]
  83. Zaytseva, O.; Kim, N.-H.; Quinn, L.M. MYC in brain development and cancer. Int. J. Mol. Sci. 2020, 21, 7742. [Google Scholar] [CrossRef]
  84. Schmidlin, C.J.; Shakya, A.; Dodson, M.; Chapman, E.; Zhang, D.D. The intricacies of NRF2 regulation in cancer. Semin. Cancer Biol. 2021, 76, 110–119. [Google Scholar] [CrossRef]
  85. LeBoeuf, S.E.; Wu, W.L.; Karakousi, T.R.; Karadal, B.; Jackson, S.R.; Davidson, S.M.; Wong, K.-K.; Koralov, S.B.; Sayin, V.I.; Papagiannakopoulos, T. Activation of oxidative stress response in cancer generates a druggable dependency on exogenous non-essential amino acids. Cell Metab. 2020, 31, 339–350.e4. [Google Scholar] [CrossRef] [PubMed]
  86. Fabregat, I.; Fernando, J.; Mainez, J.; Sancho, P. TGF-beta signaling in cancer treatment. Curr. Pharm. Des. 2014, 20, 2934–2947. [Google Scholar] [CrossRef] [PubMed]
  87. Sanchez-Vega, F.; Mina, M.; Armenia, J.; Chatila, W.K.; Luna, A.; La, K.C.; Dimitriadoy, S.; Liu, D.L.; Kantheti, H.S.; Saghafinia, S. Oncogenic signaling pathways in the cancer genome atlas. Cell 2018, 173, 321–337. [Google Scholar] [CrossRef]
  88. Sui, X.; Jin, L.; Huang, X.; Geng, S.; He, C.; Hu, X. p53 signaling and autophagy in cancer: A revolutionary strategy could be developed for cancer treatment. Autophagy 2011, 7, 565–571. [Google Scholar] [CrossRef]
  89. Rader, J.; Russell, M.R.; Hart, L.S.; Nakazawa, M.S.; Belcastro, L.T.; Martinez, D.; Li, Y.; Carpenter, E.L.; Attiyeh, E.F.; Diskin, S.J. Dual CDK4/CDK6 Inhibition Induces Cell-Cycle Arrest and Senescence in NeuroblastomaCDK4/6 Inhibition in Neuroblastoma. Clin. Cancer Res. 2013, 19, 6173–6182. [Google Scholar] [CrossRef]
  90. Hashem, S.; Ali, T.A.; Akhtar, S.; Nisar, S.; Sageena, G.; Ali, S.; Al-Mannai, S.; Therachiyil, L.; Mir, R.; Elfaki, I. Targeting cancer signaling pathways by natural products: Exploring promising anti-cancer agents. Biomed. Pharmacother. 2022, 150, 113054. [Google Scholar] [CrossRef]
  91. Shah, U.; Shah, R.; Acharya, S.; Acharya, N. Novel anticancer agents from plant sources. Chin. J. Nat. Med. 2013, 11, 16–23. [Google Scholar] [CrossRef]
  92. Darnell, J.E., Jr. STATs and gene regulation. Science 1997, 277, 1630–1635. [Google Scholar] [CrossRef]
  93. Cragg, G.M.; Grothaus, P.G.; Newman, D.J. Impact of natural products on developing new anti-cancer agents. Chem. Rev. 2009, 109, 3012–3043. [Google Scholar] [CrossRef] [PubMed]
  94. Patel, D.K. Therapeutic Effectiveness of Magnolin on cancers and other Human complications. Pharmacol. Res. Mod. Chin. Med. 2022, 6, 100203. [Google Scholar] [CrossRef]
  95. Wang, J.; Zhang, S.; Huang, K.; Shi, L.; Zhang, Q. Magnolin inhibits proliferation and invasion of breast cancer MDA-MB-231 cells by targeting the ERK1/2 signaling pathway. Chem. Pharm. Bull. 2020, 68, 421–427. [Google Scholar] [CrossRef] [PubMed]
  96. Jun, A.Y.; Kim, H.-J.; Park, K.-K.; Son, K.H.; Lee, D.H.; Woo, M.-H.; Chung, W.-Y. Tetrahydrofurofuran-type lignans inhibit breast cancer-mediated bone destruction by blocking the vicious cycle between cancer cells, osteoblasts and osteoclasts. Investig. New Drugs 2014, 32, 1–13. [Google Scholar] [CrossRef]
  97. Lee, C.-J.; Lee, M.-H.; Yoo, S.-M.; Choi, K.-I.; Song, J.-H.; Jang, J.-H.; Oh, S.-R.; Ryu, H.-W.; Lee, H.-S.; Surh, Y.-J. Magnolin inhibits cell migration and invasion by targeting the ERKs/RSK2 signaling pathway. BMC Cancer 2015, 15, 576. [Google Scholar] [CrossRef]
  98. Lee, C.-J.; Lee, H.S.; Ryu, H.W.; Lee, M.-H.; Lee, J.Y.; Li, Y.; Dong, Z.; Lee, H.-K.; Oh, S.-R.; Cho, Y.-Y. Targeting of magnolin on ERKs inhibits Ras/ERKs/RSK2-signaling-mediated neoplastic cell transformation. Carcinogenesis 2014, 35, 432–441. [Google Scholar] [CrossRef]
  99. Wang, W.; Xiao, Y.; Li, S.; Zhu, X.; Meng, L.; Song, C.; Yu, C.; Jiang, N.; Liu, Y. Synergistic activity of magnolin combined with B-RAF inhibitor SB590885 in hepatocellular carcinoma cells via targeting PI3K-AKT/mTOR and ERK MAPK pathway. Am. J. Transl. Res. 2019, 11, 3816. [Google Scholar]
  100. Yoo, S.-M.; Lee, C.-J.; Kim, S.-M.; Cho, S.-Y.; Park, J.; Cho, Y.-Y. Molecular mechanisms of magnolin resistance in ovarian cancer cells. Cancer Res. 2018, 78, 4028. [Google Scholar] [CrossRef]
  101. Mukhija, M.; Dhar, K.L.; Kalia, A.N. Bioactive Lignans from Zanthoxylum alatum Roxb. stem bark with cytotoxic potential. J. Ethnopharmacol. 2014, 152, 106–112. [Google Scholar] [CrossRef]
  102. Park, J.; Lee, G.E.; An, H.J.; Lee, C.J.; Cho, E.S.; Kang, H.C.; Lee, J.Y.; Lee, H.S.; Choi, J.S.; Kim, D.J.; et al. Kaempferol sensitizes cell proliferation inhibition in oxaliplatin-resistant colon cancer cells. Arch. Pharm. Res. 2021, 44, 1091–1108. [Google Scholar] [CrossRef]
  103. Yu, H.; Yin, S.; Zhou, S.; Shao, Y.; Sun, J.; Pang, X.; Han, L.; Zhang, Y.; Gao, X.; Jin, C.; et al. Magnolin promotes autophagy and cell cycle arrest via blocking LIF/Stat3/Mcl-1 axis in human colorectal cancers. Cell Death Dis. 2018, 9, 702. [Google Scholar] [CrossRef] [PubMed]
  104. Waks, A.G.; Winer, E.P. Breast Cancer Treatment: A Review. JAMA 2019, 321, 288–300. [Google Scholar] [CrossRef]
  105. Kim, K.M.; Jung, J. Upregulation of G Protein-Coupled Estrogen Receptor by Chrysin-Nanoparticles Inhibits Tumor Proliferation and Metastasis in Triple Negative Breast Cancer Xenograft Model. Front. Endocrinol. 2020, 11, 560605. [Google Scholar] [CrossRef]
  106. Sun, Y.-S.; Zhao, Z.; Yang, Z.-N.; Xu, F.; Lu, H.-J.; Zhu, Z.-Y.; Shi, W.; Jiang, J.; Yao, P.-P.; Zhu, H.-P. Risk factors and preventions of breast cancer. Int. J. Biol. Sci. 2017, 13, 1387. [Google Scholar] [CrossRef] [PubMed]
  107. Key, T.J.; Verkasalo, P.K.; Banks, E. Epidemiology of breast cancer. Lancet Oncol. 2001, 2, 133–140. [Google Scholar] [CrossRef]
  108. DeSantis, C.E.; Fedewa, S.A.; Goding Sauer, A.; Kramer, J.L.; Smith, R.A.; Jemal, A. Breast cancer statistics, 2015: Convergence of incidence rates between black and white women. CA Cancer J. Clin. 2016, 66, 31–42. [Google Scholar] [CrossRef] [PubMed]
  109. Smolarz, B.; Nowak, A.Z.; Romanowicz, H. Breast Cancer—Epidemiology, Classification, Pathogenesis and Treatment (Review of Literature). Cancers 2022, 14, 2569. [Google Scholar] [CrossRef]
  110. Chan, Y.W.; Ma, H.T.; Wong, W.; Ho, C.C.; On, K.F.; Poon, R.Y. CDK1 inhibitors antagonize the immediate apoptosis triggered by spindle disruption but promote apoptosis following the subsequent rereplication and abnormal mitosis. Cell Cycle 2008, 7, 1449–1461. [Google Scholar] [CrossRef]
  111. Liu, X.; Zhao, W.; Wang, W.; Lin, S.; Yang, L. Puerarin suppresses LPS-induced breast cancer cell migration, invasion and adhesion by blockage NF-κB and Erk pathway. Biomed. Pharmacother. 2017, 92, 429–436. [Google Scholar] [CrossRef] [PubMed]
  112. Sun, Q.; Liang, Y.; Zhang, T.; Wang, K.; Yang, X. ER-α36 mediates estrogen-stimulated MAPK/ERK activation and regulates migration, invasion, proliferation in cervical cancer cells. Biochem. Biophys. Res. Commun. 2017, 487, 625–632. [Google Scholar] [CrossRef] [PubMed]
  113. 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]
  114. Ushijima, T.; Clark, S.J.; Tan, P. Mapping genomic and epigenomic evolution in cancer ecosystems. Science 2021, 373, 1474–1479. [Google Scholar] [CrossRef] [PubMed]
  115. Cagle, P.T.; Allen, T.C.; Olsen, R.J. Lung cancer biomarkers: Present status and future developments. Arch. Pathol. Lab. Med. 2013, 137, 1191–1198. [Google Scholar] [CrossRef] [PubMed]
  116. Minna, J.D.; Roth, J.A.; Gazdar, A.F. Focus on lung cancer. Cancer Cell 2002, 1, 49–52. [Google Scholar] [CrossRef] [PubMed]
  117. Lindeman, N.I.; Cagle, P.T.; Beasley, M.B.; Chitale, D.A.; Dacic, S.; Giaccone, G.; Jenkins, R.B.; Kwiatkowski, D.J.; Saldivar, J.-S.; Squire, J. Molecular testing guideline for selection of lung cancer patients for EGFR and ALK tyrosine kinase inhibitors: Guideline from the College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology. J. Thorac. Oncol. 2013, 8, 823–859. [Google Scholar] [PubMed]
  118. Lindeman, N.I.; Cagle, P.T.; Aisner, D.L.; Arcila, M.E.; Beasley, M.B.; Bernicker, E.H.; Colasacco, C.; Dacic, S.; Hirsch, F.R.; Kerr, K. Updated molecular testing guideline for the selection of lung cancer patients for treatment with targeted tyrosine kinase inhibitors: Guideline from the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. Arch. Pathol. Lab. Med. 2018, 142, 321–346. [Google Scholar] [PubMed]
  119. Yoo, S.-M.; Cho, S.J.; Cho, Y.-Y. Molecular targeting of ERKs/RSK2 signaling axis in cancer prevention. J. Cancer Prev. 2015, 20, 165. [Google Scholar] [CrossRef]
  120. Chen, J.-S.; Huang, X.-H.; Wang, Q.; Huang, J.-Q.; Zhang, L.-J.; Chen, X.-L.; Lei, J.; Cheng, Z.-X. Sonic hedgehog signaling pathway induces cell migration and invasion through focal adhesion kinase/AKT signaling-mediated activation of matrix metalloproteinase (MMP)-2 and MMP-9 in liver cancer. Carcinogenesis 2013, 34, 10–19. [Google Scholar] [CrossRef]
  121. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef]
  122. Liu, C.-Y.; Chen, K.-F.; Chen, P.-J. Treatment of liver cancer. Cold Spring Harb. Perspect. Med. 2015, 5, a021535. [Google Scholar] [CrossRef]
  123. Dhanasekaran, R.; Bandoh, S.; Roberts, L.R. Molecular pathogenesis of hepatocellular carcinoma and impact of therapeutic advances. F1000Research 2016, 5, 879. [Google Scholar] [CrossRef]
  124. Whittaker, S.; Marais, R.; Zhu, A. The role of signaling pathways in the development and treatment of hepatocellular carcinoma. Oncogene 2010, 29, 4989–5005. [Google Scholar] [CrossRef] [PubMed]
  125. Wu, Q.; Liu, T.-Y.; Hu, B.-C.; Li, X.; Wu, Y.-T.; Sun, X.-T.; Jiang, X.-W.; Wang, S.; Qin, X.-C.; Ding, H.-W. CK-3, a novel methsulfonyl pyridine derivative, suppresses hepatocellular carcinoma proliferation and invasion by blocking the PI3K/AKT/mTOR and MAPK/ERK pathways. Front. Oncol. 2021, 11, 717626. [Google Scholar] [CrossRef] [PubMed]
  126. Xie, G.; Wang, Z.; Chen, Y.; Zhang, S.; Feng, L.; Meng, F.; Yu, Z. Dual blocking of PI3K and mTOR signaling by NVP-BEZ235 inhibits proliferation in cervical carcinoma cells and enhances therapeutic response. Cancer Lett. 2017, 388, 12–20. [Google Scholar] [CrossRef] [PubMed]
  127. Matter, M.S.; Decaens, T.; Andersen, J.B.; Thorgeirsson, S.S. Targeting the mTOR pathway in hepatocellular carcinoma: Current state and future trends. J. Hepatol. 2014, 60, 855–865. [Google Scholar] [CrossRef] [PubMed]
  128. Kunter, I.; Erdal, E.; Nart, D.; Yilmaz, F.; Karademir, S.; Sagol, O.; Atabey, N. Active form of AKT controls cell proliferation and response to apoptosis in hepatocellular carcinoma. Oncol. Rep. 2014, 31, 573–580. [Google Scholar] [CrossRef]
  129. Diniz, P.H.; Silva, S.D.; Vidigal, P.V.; Xavier, M.A.; Lima, C.X.; Faria, L.C.; Ferrari, T.C. Expression of MAPK and PI3K/AKT/mTOR proteins according to the chronic liver disease etiology in hepatocellular carcinoma. J. Oncol. 2020, 2020, 4609360. [Google Scholar] [CrossRef]
  130. Llovet, J.M.; Ricci, S.; Mazzaferro, V.; Hilgard, P.; Gane, E.; Blanc, J.-F.; De Oliveira, A.C.; Santoro, A.; Raoul, J.-L.; Forner, A. Sorafenib in advanced hepatocellular carcinoma. N. Engl. J. Med. 2008, 359, 378–390. [Google Scholar] [CrossRef]
  131. Tringler, B.; Liu, W.; Corral, L.; Torkko, K.C.; Enomoto, T.; Davidson, S.; Lucia, M.S.; Heinz, D.E.; Papkoff, J.; Shroyer, K.R. B7-H4 overexpression in ovarian tumors. Gynecol. Oncol. 2006, 100, 44–52. [Google Scholar] [CrossRef]
  132. Gray, J.W.; Suzuki, S.; Kuo, W.-L.; Polikoff, D.; Deavers, M.; Smith-McCune, K.; Berchuck, A.; Pinkel, D.; Albertson, D.; Mills, G.B. Specific keynote: Genome copy number abnormalities in ovarian cancer. Gynecol. Oncol. 2003, 88, S16–S21. [Google Scholar] [CrossRef] [PubMed]
  133. Reid, B.M.; Permuth, J.B.; Sellers, T.A. Epidemiology of ovarian cancer: A review. Cancer Biol. Med. 2017, 14, 9–32. [Google Scholar] [CrossRef]
  134. Omeragić, F.; Ljuca, D. Rana detekcija raka jajnika u Federaciji Bosne i Hercegovine i uloga porodične medicine. Med. Glas. 2007, 4, 77–81. [Google Scholar]
  135. Trentham-Dietz, A.; Newcomb, P.A.; Nichols, H.B.; Hampton, J.M. Breast cancer risk factors and second primary malignancies among women with breast cancer. Breast Cancer Res. Treat. 2007, 105, 195–207. [Google Scholar] [CrossRef] [PubMed]
  136. Bast, R.C., Jr.; Hennessy, B.; Mills, G.B. The biology of ovarian cancer: New opportunities for translation. Nat. Rev. Cancer 2009, 9, 415–428. [Google Scholar] [CrossRef]
  137. Gwinn, M.L.; Lee, N.C.; Rhodes, P.H.; Layde, P.M.; Rubin, G.L. Pregnancy, breast feeding, and oral contraceptives and the risk of epithelial ovarian cancer. J. Clin. Epidemiol. 1990, 43, 559–568. [Google Scholar] [CrossRef] [PubMed]
  138. Gao, X.; Wang, B.; Wei, X.; Men, K.; Zheng, F.; Zhou, Y.; Zheng, Y.; Gou, M.; Huang, M.; Guo, G. Anticancer effect and mechanism of polymer micelle-encapsulated quercetin on ovarian cancer. Nanoscale 2012, 4, 7021–7030. [Google Scholar] [CrossRef]
  139. Oronsky, B.; Ray, C.M.; Spira, A.I.; Trepel, J.B.; Carter, C.A.; Cottrill, H.M. A brief review of the management of platinum-resistant–platinum-refractory ovarian cancer. Med. Oncol. 2017, 34, 1–7. [Google Scholar] [CrossRef]
  140. Youness, R.A.; Kamel, R.; Elkasabgy, N.A.; Shao, P.; Farag, M.A. Recent advances in tannic acid (gallotannin) anticancer activities and drug delivery systems for efficacy improvement; a comprehensive review. Molecules 2021, 26, 1486. [Google Scholar] [CrossRef]
  141. Pokhriyal, R.; Hariprasad, R.; Kumar, L.; Hariprasad, G. Chemotherapy Resistance in Advanced Ovarian Cancer Patients. Biomark Cancer 2019, 11, 1179299x19860815. [Google Scholar] [CrossRef]
  142. Elsayed, A.M.; Amero, P.; Salama, S.A.; Abdelaziz, A.H.; Lopez-Berestein, G.; Rodriguez-Aguayo, C. Back to the Future: Rethinking the Great Potential of lncRNA(S) for Optimizing Chemotherapeutic Response in Ovarian Cancer. Cancers 2020, 12, 2406. [Google Scholar] [CrossRef]
  143. Steinmetz, R.; Wagoner, H.A.; Zeng, P.; Hammond, J.R.; Hannon, T.S.; Meyers, J.L.; Pescovitz, O.H. Mechanisms regulating the constitutive activation of the extracellular signal-regulated kinase (ERK) signaling pathway in ovarian cancer and the effect of ribonucleic acid interference for ERK1/2 on cancer cell proliferation. Mol. Endocrinol. 2004, 18, 2570–2582. [Google Scholar] [CrossRef]
  144. Sak, K.; Lust, H.; Kase, M.; Saar, M.; Jaal, J. Suppression of taxanes cytotoxicity by citrus flavonoid hesperetin in PPC-1 human prostate cancer cells. AntiCancer Res. 2018, 38, 6209–6215. [Google Scholar] [CrossRef] [PubMed]
  145. Sathiakumar, N.; Delzell, E.; Morrisey, M.A.; Falkson, C.; Yong, M.; Chia, V.; Blackburn, J.; Arora, T.; Kilgore, M.L. Mortality following bone metastasis and skeletal-related events among men with prostate cancer: A population-based analysis of US Medicare beneficiaries, 1999–2006. Prostate Cancer Prostatic Dis. 2011, 14, 177–183. [Google Scholar] [CrossRef]
  146. Nguyen-Nielsen, M.; Borre, M. Diagnostic and therapeutic strategies for prostate cancer. Semin. Nucl. Med. 2016, 46, 484–490. [Google Scholar] [CrossRef] [PubMed]
  147. Kamisawa, T.; Wood, L.D.; Itoi, T.; Takaori, K. Pancreatic cancer. Lancet 2016, 388, 73–85. [Google Scholar] [CrossRef]
  148. Dariya, B.; Alam, A.; Nagaraju, G.P. Biology, pathophysiology, and epidemiology of pancreatic cancer. In Theranostic Approach for Pancreatic Cancer; Elsevier: Amsterdam, The Netherlands, 2019; pp. 1–50. [Google Scholar]
  149. Arnold, M.; Sierra, M.S.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global patterns and trends in colorectal cancer incidence and mortality. Gut 2017, 66, 683–691. [Google Scholar] [CrossRef]
  150. Siegel, R.L.; Miller, K.D.; Fedewa, S.A.; Ahnen, D.J.; Meester, R.G.S.; Barzi, A.; Jemal, A. Colorectal cancer statistics, 2017. CA Cancer J. Clin. 2017, 67, 177–193. [Google Scholar] [CrossRef] [PubMed]
  151. Rawla, P.; Sunkara, T.; Barsouk, A. Epidemiology of colorectal cancer: Incidence, mortality, survival, and risk factors. Prz. Gastroenterol. 2019, 14, 89–103. [Google Scholar] [CrossRef]
  152. Haggar, F.A.; Boushey, R.P. Colorectal cancer epidemiology: Incidence, mortality, survival, and risk factors. Clin. Colon Rectal Surg. 2009, 22, 191–197. [Google Scholar] [CrossRef]
  153. Shuwen, H.; Yinhang, W.; Xingming, Z.; Jing, Z.; Jinxin, L.; Wei, W.; Kefeng, D. Using whole-genome sequencing (WGS) to plot colorectal cancer-related gut microbiota in a population with varied geography. Gut Pathog. 2022, 14, 50. [Google Scholar] [CrossRef]
  154. Bousbaa, H. Novel Anticancer Strategies. Pharmaceutics 2021, 13, 275. [Google Scholar] [CrossRef]
  155. Jiang, G.M.; Tan, Y.; Wang, H.; Peng, L.; Chen, H.T.; Meng, X.J.; Li, L.L.; Liu, Y.; Li, W.F.; Shan, H. The relationship between autophagy and the immune system and its applications for tumor immunotherapy. Mol. Cancer 2019, 18, 17. [Google Scholar] [CrossRef] [PubMed]
  156. Amaravadi, R.; Kimmelman, A.C.; White, E. Recent insights into the function of autophagy in cancer. Genes Dev. 2016, 30, 1913–1930. [Google Scholar] [CrossRef]
  157. White, E.; Mehnert, J.M.; Chan, C.S. Autophagy, Metabolism, and Cancer. Clin. Cancer Res. 2015, 21, 5037–5046. [Google Scholar] [CrossRef]
  158. Li, X.; He, S.; Ma, B. Autophagy and autophagy-related proteins in cancer. Mol. Cancer 2020, 19, 12. [Google Scholar] [CrossRef] [PubMed]
  159. Reichel, A.; Lienau, P. Pharmacokinetics in drug discovery: An exposure-centred approach to optimising and predicting drug efficacy and safety. New Approaches Drug Discov. 2016, 232, 235–260. [Google Scholar]
  160. Bhuia, M.S.; Islam, T.; Rokonuzzman, M.; Shamsh Prottay, A.A.; Akter, F.; Hossain, M.I.; Chowdhury, R.; Kazi, M.A.; Khalipha, A.B.R.; Coutinho, H.D.M. Modulatory effects of phytol on the antiemetic property of domperidone, possibly through the D2 receptor interaction pathway: In vivo and in silico studies. 3 Biotech 2023, 13, 116. [Google Scholar] [CrossRef]
  161. Bhuia, M.S.; Rahaman, M.M.; Islam, T.; Bappi, M.H.; Sikder, M.I.; Hossain, K.N.; Akter, F.; Al Shamsh Prottay, A.; Rokonuzzman, M.; Gürer, E.S. Neurobiological effects of gallic acid: Current perspectives. Chin. Med. 2023, 18, 27. [Google Scholar] [CrossRef]
  162. Bhattaram, V.A.; Graefe, U.; Kohlert, C.; Veit, M.; Derendorf, H. Pharmacokinetics and bioavailability of herbal medicinal products. Phytomedicine 2002, 9, 1–33. [Google Scholar] [CrossRef]
  163. Hassan, M.; Sallam, H.; Hassan, Z. The role of pharmacokinetics and pharmacodynamics in early drug development with reference to the cyclin-dependent kinase (Cdk) inhibitor-roscovitine. Sultan Qaboos Univ. Med. J. 2011, 11, 165. [Google Scholar] [PubMed]
  164. Kim, N.J.; Song, W.Y.; Yoo, S.D.; Oh, S.-R.; Lee, H.-K.; Lee, H.S. Pharmacokinetics of magnolin in rats. Arch. Pharmacal Res. 2010, 33, 933–938. [Google Scholar] [CrossRef]
  165. Muhamad, N.; Na-Bangchang, K. Metabolite profiling in anticancer drug development: A systematic review. Drug Des. Dev. Ther. 2020, 14, 1401–1444. [Google Scholar] [CrossRef] [PubMed]
  166. Miyazawa, M.; Kasahara, H.; Kameoka, H. Biotransformation of lignans: Metabolism of (+)-Yangabin in Spodoptera litura. Nat. Prod. Lett. 1996, 8, 87–88. [Google Scholar] [CrossRef]
  167. Miyazawa, M.; Kasahara, H.; Kameoka, H. Biotransformation of lignans: A specific microbial oxidation of (+)-eudesmin and (+)-magnolin by Aspergillus niger. Phytochemistry 1993, 34, 1501–1507. [Google Scholar] [CrossRef]
  168. Kim, D.K.; Liu, K.-H.; Jeong, J.H.; Ji, H.Y.; Oh, S.-R.; Lee, H.-K.; Lee, H.S. In vitro metabolism of magnolin and characterization of cytochrome P450 enzymes responsible for its metabolism in human liver microsomes. Xenobiotica 2011, 41, 358–371. [Google Scholar] [CrossRef]
Figure 1. (a) Two-dimensional chemical structure of magnolin and (b) different pharmacological activities of magnolin based on literature.
Figure 1. (a) Two-dimensional chemical structure of magnolin and (b) different pharmacological activities of magnolin based on literature.
Molecules 28 03671 g001
Figure 2. Schematic presentation of data collection, inclusion and exclusion criteria (PM: PubMed; SD: Science Direct; MS: miscellaneous (e.g., Google Scholar and Research Gate)).
Figure 2. Schematic presentation of data collection, inclusion and exclusion criteria (PM: PubMed; SD: Science Direct; MS: miscellaneous (e.g., Google Scholar and Research Gate)).
Molecules 28 03671 g002
Figure 3. Anticancer mechanisms of magnolin.
Figure 3. Anticancer mechanisms of magnolin.
Molecules 28 03671 g003
Table 1. Biological sources of magnolin.
Table 1. Biological sources of magnolin.
Plant NamePlant PartReferences
Magnolia fargesiiFlower buds[37,52]
Zanthoxylum simulansBark[53]
M. biondiiFlower buds[54,55,56]
Pseuderanthemum carruthersiiRoot[57]
M. officinalisFlower buds[58]
M. sargentiana and M. sprengeri-[42]
Castilleja tenuifloraAerial part of roots[59]
Astrantia majorFruits[60]
Artemisia gorgonum WebbLeaves and flowers[44]
Acorus calamus-[61]
Rollinia mucosaSeeds[62]
M. denudataFlowers[63,64]
M. kobusFlower buds, bark[65,66]
M. salicifoliaFlower buds[65]
Licaria armeniacaTrunk wood, fruits[67,68]
Achillea holosericea.Aerial parts[69]
Annona pickeliiLeaves[70]
M. lilifloraLeaves[71]
A. gypsicolaAerial parts and roots[72,73]
Z. alatumSeeds[74]
Hernandia ovigeraLeaves[75]
Physalis peruvianaFruits[76]
Table 2. Pharmacological mechanisms of magnolin involved in its anticancer activities.
Table 2. Pharmacological mechanisms of magnolin involved in its anticancer activities.
Type of CancerExperimental Model/
Cell Line
Tested
Concentrations
Efficacy, IC50
(Exposure Time)
Anticancer Effects and MechanismsReference
Breast cancerMDA-MB-231 20 and 50 µM30.34 µM (24 h)↓MEK1/2, ↓ERK1/2, ↓CDK1, ↓BCL2, ↓MMP 2 and 9, ↑CASPASES3 and 9
↓proliferation, ↓invasion, ↑apoptosis
[95]
MDA-MB-23110, 20, 40, 60, 80, and 100 µM-↓mRNA expression, ↓PTHrP, ↓MMP9, ↓cathepsin K, ↑RANKL/OPG ratio
↓bone loss and bone-resorbing activity
[96]
-MCF-7 100 µg/mL-↑cytotoxicity[57]
Lung cancerJB6CL41, NCI-H1975 and A549 15, 30, and 60 µM-↓ERK1/2, ↓MMP2, ↓MMP9, ↓RSK2, ↓migration, ↓invasion[97]
NIH3T3, A549 15, 30, and 60 µM-↓ERK1, ↓ERK2, ↓RSK2, ↓ATF1, ↓AP1, ↓proliferation, ↓migration[98]
Hepatocellular carcinomaBEL-7402 and SK-HEP125, 50, 75 100, and 125 μM-↓MEK, ↓PI3K, ↓AKT,
↓proliferation
[99]
Ovarian cancerTOV-112D 15, 30, and 60 µM-↓ERK1, ↓ERK2, ↑P16Ink4a, ↑P27Kip1, ↓G1 and G2/M phase, ↓proliferation[45]
TOV-112D -16 nM 68 nM↓ERK1, ↓ERK2, ↓ATF1, ↓RSK2
↓proliferation
[100]
Prostate cancerPANC-1-0.51 µM↓MMP3, ↓proliferation, ↓migration[76]
PC3 and DU14550 and 100 µM-↓AKT, ↓BCL2, ↑BAX, ↑CASPASE3,
↑P53, ↑P21
↓G1, G2 phase, ↑apoptosis
[43]
Pancreatic carcinomaMIA-PaCa--↑cytotoxicity[101]
Colon cancerHCT116 and HT29--↓G2/M-phases, ↓proliferation[102]
Colorectal cancerHCT116 and SW48010, 20, 30, and 40 µM-↓LIF, ↓STAT3, ↓MCL1
↑ autophagy ↓cell cycle
[103]
Arrows (↑ and ↓) show an increase or decrease in the obtained variables. MEK1/2: mitogen-activated protein kinase kinase 1 and 2; ERK1/2: extracellular signal-regulated kinase 1 and 2; CDK1: cyclin-dependent kinase 1; BCL2: B-cell lymphoma 2; MMP 2 and 9: matrix metalloproteinases 2 and 9; RSK2: ribosomal S6 kinase 2; ATF1: activating transcription factor 1; AP1: activator protein 1; PI3K: phosphoinositide 3-kinase; AKT: protein kinase B; BAX: Bcl-2-associated X protein; LIF: leukemia inhibitory factor; STAT3: signal transducer and activator of transcription 3; MCL1: myeloid leukemia 1; P53: tumor protein P53; P21: tumor protein P21.
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

Bhuia, M.S.; Wilairatana, P.; Chowdhury, R.; Rakib, A.I.; Kamli, H.; Shaikh, A.; Coutinho, H.D.M.; Islam, M.T. Anticancer Potentials of the Lignan Magnolin: A Systematic Review. Molecules 2023, 28, 3671. https://doi.org/10.3390/molecules28093671

AMA Style

Bhuia MS, Wilairatana P, Chowdhury R, Rakib AI, Kamli H, Shaikh A, Coutinho HDM, Islam MT. Anticancer Potentials of the Lignan Magnolin: A Systematic Review. Molecules. 2023; 28(9):3671. https://doi.org/10.3390/molecules28093671

Chicago/Turabian Style

Bhuia, Md. Shimul, Polrat Wilairatana, Raihan Chowdhury, Asraful Islam Rakib, Hossam Kamli, Ahmad Shaikh, Henrique D. M. Coutinho, and Muhammad Torequl Islam. 2023. "Anticancer Potentials of the Lignan Magnolin: A Systematic Review" Molecules 28, no. 9: 3671. https://doi.org/10.3390/molecules28093671

Article Metrics

Back to TopTop