Next Article in Journal
Photodynamic Therapy for the Treatment of Bowen’s Disease: A Review on Efficacy, Non-Invasive Treatment Monitoring, Tolerability, and Cosmetic Outcome
Previous Article in Journal
The Role of SOX2 and SOX9 Transcription Factors in the Reactivation-Related Functional Properties of NT2/D1-Derived Astrocytes
Previous Article in Special Issue
Tephrosin Suppresses the Chemoresistance of Paclitaxel-Resistant Ovarian Cancer via Inhibition of FGFR1 Signaling Pathway
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Natural Products as Regulators against Matrix Metalloproteinases for the Treatment of Cancer

1
Department of Chemistry, Faculty of Science, Mawlana Bhashani Science and Technology University, Santosh, Tangail 1902, Bangladesh
2
Department of Chemistry Education, Kongju National University, Gongju 32588, Chungcheongnam-do, Republic of Korea
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(4), 794; https://doi.org/10.3390/biomedicines12040794
Submission received: 29 February 2024 / Revised: 21 March 2024 / Accepted: 29 March 2024 / Published: 3 April 2024
(This article belongs to the Special Issue Natural Products to Prevent Cancer Progression)

Abstract

:
Cancers are currently the major cause of mortality in the world. According to previous studies, matrix metalloproteinases (MMPs) have an impact on tumor cell proliferation, which could lead to the onset and progression of cancers. Therefore, regulating the expression and activity of MMPs, especially MMP-2 and MMP-9, could be a promising strategy to reduce the risk of cancers. Various studies have tried to investigate and understand the pathophysiology of cancers to suggest potent treatments. In this review, we summarize how natural products from marine organisms and plants, as regulators of MMP-2 and MMP-9 expression and enzymatic activity, can operate as potent anticancer agents.

1. Introduction

Cancers represent a danger to human health, and they are the second leading cause of death worldwide [1]. Currently, people living in poor countries bear the brunt of sickness; a recent survey revealed that about 10 million cancer-related deaths and 19.3 million new cases were reported globally in 2020 [1,2]. The leading cause of cancer-related deaths worldwide was lung cancer (1.79 million) followed by other cancers, including liver cancer (0.83 million), stomach cancer (0.77 million), breast cancer (0.68 million), and other cancers (Figure 1) [3].
According to research on 29 cancer diseases across 204 nations, the overall cost of cancer to the world economy between 2020 and 2050 will be USD 25.2 trillion. About half of those will be attributed to five different forms of cancer: lung, liver, breast, colon, and leukemia [4]. Some of the key participants in the growth and aggressiveness of cancer cells of glycolytic enzymes could be lactate dehydrogenase (LDH), caspases, cyclin-dependent kinases, redox–detox enzymes, matrix metalloproteinases-2/9 (MMP-2 and MMP-9), and NAD+-dependent enzymes [5,6].
When a family member has a greater incidence of a certain type of cancer, it has been feasible to determine whether the disease runs in the family. The most common hereditary cancer type is colorectal cancer, by mutation of DNA MMR genes. In addition, breast and ovarian cancer are due to mutations of the BRCA1 as well as BRCA2 genes, respectively. However, 20–30% of instances of breast cancer, which affects 10 in 100 women, are genetically predisposed to BRCA1 and BRCA2 gene mutations [7,8]. Moreover, rapid industrialization and inappropriate pesticide usage are also responsible for lung, liver, and prostate cancer [9].
Particularly, it is difficult to diagnose cancer until it reaches the metastasis stage, consequently lowering longevity [10]. To cure cancer, different types of medical technology have been used including radiation, surgery, and systemic hormonal therapy. Surgery has been performed on about 56% and 18% of those in stages I and II. The majority of people (62%) in stage III receive chemotherapy to treat cancer, though expensive. As an alternative to surgery and chemotherapy, the administration of natural compounds will be used to modify the genes of cancer cells to mitigate cancerous death [11].
Throughout malignancy, tumor cells interact with surrounding endothelial cells as well as growth regulators, cytokines, and extracellular matrix (ECM) components [12], such as gelatin, collagen, elastin, and fibronectin. The four features of cancer (angiogenesis, invasion, migration, and metastasis) are dependent on the exterior microenvironment. MMPs are crucial because they increase the interplay between cells and the extracellular matrix by breaking down several types of cell adhesion molecules [13,14]. The ECM exists in muscle niches and consists of proteins, polysaccharides, RNA, and other materials, and it is essential for maintaining homeostasis and regulating the development of skeletal muscle [15]. Recent research by Liu et al. has demonstrated that collagen I, an important component of the extracellular matrix, stimulates focal adhesion kinase activation, which in turn controls NF-κB nuclear translocation and improves myoblast migration [16]. Thus, it could be concluded that ECM plays a critical role in maintaining the physiological function of satellite cells as well as skeletal muscle development. ECM variations are primarily caused by two mechanisms: (1) chemical modification, which alters the biochemical and structural properties of the ECM, and (2) proteolytic degradation, which releases bioactive ECM fragments and ECM-bound factors and may be essential for the release of cellular restrictions such as migratory barriers [17]. The degradation of the ECM by MMPs is a major factor of change in cell shape. For instance, when epithelial cells lose their polarity and cell–cell adhesion is disrupted, it facilitates the movement and infiltration of these cancer cells [18,19]. Hence, MMPs promote tumor growth and spread through the capillary endothelium [20].
The history of the therapeutic use of natural resources dates back thousands of years when medicinal plants rich in phytochemicals and microbes were an important constituent of medicines. This knowledge is frequently passed down from one generation to the next. Generally, natural bioactive materials derived from plants, minerals, animals, and microorganisms have been utilized to treat a wide range of illnesses [21]. Natural products could inhibit MMPs by interacting with or chelating out the catalytic Zn(II) of MMPs. Preclinical studies have shown that the hydroxamate-based MMP inhibitors, including Batimastat and Marimastat, have potential to cure cancer [22,23]. However, the anticipated outcomes of studies on patients were insufficient. Later, structural investigations using X-ray crystallography and computer simulations were used to create several hydroxamate derivatives, especially helping to increase their sensitivity as MMP inhibitors to limit cancer [22]. In this review, we first briefly address MMPs and their critical roles in human health and cancer progression; then, we address natural products from marine organisms and plants that could regulate MMP-2 and MMP-9 expression levels and activities. We tried to provide general knowledge for development of new drugs with the potential to control MMPs, leading to the successful treatment of cancers.

1.1. Basic Information on MMPs

MMPs are Zn(II)-dependent endopeptidase [24]. MMPs have several domains including (i) membrane anchor domain and (ii) Zn(II)-binding domain, together forming the (iii) active site for substrate catalysis (Figure 2). MMPs are released by cells as (iv) proenzymes (propeptide), which are dormant and must undergo chemical adaptation by reactive oxygen species (ROS) to catalytically activated proteases [25]; (v) fibronectin repeats, which enable them to bind big substrates like collagen and effectively break gelatins [26]; and (vi) a hemopexin domain, which plays a role in ties to both substrates and inhibitors [27,28,29]. Furthermore, MMP-9 possesses a distinct (64 amino acid) linker sequence that joins the hemopexin domain, known as (vii) the O-glycosylated domain, to (viii) the active site. The most recent atypical MMP is MMP-23; it contains an immunoglobulin-like domain and cysteine-rich domain linked to the C-terminus and a transmembrane domain bound to the N-terminus [30]. Therefore, by trapping cis Kv1.3, MMP-23 affects cellular functions whereas in trans-Kv1.3, it blocks extracellular functions. Breaking the bonds in protein affects cytokine and chemokine signaling. However, MMP-23 activity in melanoma cancers can assist disease progression through these mechanisms [31]. The primary function of MMPs is to disintegrate ECM components. MMP-mediated breakage of ECM components plays a role in triggering the discharge of growth hormones from the ECM, propagation, and tissue reconstruction of cancer cells [32].
Among multiple MMPs, MMP-2 and MMP-9 have been reported to be involved in pathology of cancers. MMP-2 (gelatinase A) could break down type I, type II, and type III collagen and is produced by stroma cells in the majority of organs, including hematopoietic, endothelial, dendritic, fibroblast, mast, and macrophage cells. MMP-9 (gelatinase B) is prevalent in healthy tissues and released by dendritic, hematopoietic, macrophage, neutrophil, fibroblast, and lymphocyte cells [33,34].
The structures of MMP-2 and MMP-9 are similar, but slightly different at the catalytic center. For MMP-2, the catalytic center is composed of Zn(II) coordinated by three histidines (His201, His205, and His211) and glutamic acid (Glu202). The structure of the catalytic domain of human MMP-9 is composed of the Zn(II) coordinated by three histidine residues (His401, His405, and His411) and glutamic acid residue (Glu402) (Figure 3) [35].

1.2. Normal and Disease Conditions

The ECM aids in intercellular communication, signal transduction, and the regulation of biological processes including cell division and death [36]. ECM remodeling is an important and necessary process that takes place in both physiologically normal and diseased conditions. During its remodeling, cells entirely or partially destroy certain components of the ECM, such as laminin, collagen, and fibrin [37]. MMPs are among the proteases that mediate the breakdown process by inducing remodeling in both temporal and spatial dimensions [38]. Therefore, regulation of MMPs is strictly maintained to keep homeostasis. Dysregulation in any regulatory system leads to an aberrant expression of MMPs, which can exacerbate tissue degradation disorders such as fibrotic illnesses, osteoarthritis/arthritis, neurological illnesses including glaucoma, Parkinson’s disease, Alzheimer’s disease, and Japanese encephalitis, which have been linked to aberrant MMP expression [39,40]. Under normal circumstances, MMP expression is typically relatively low; nonetheless, higher MMP levels have been seen in several cancer types and are associated with increased tumor development and proliferation [41].

1.3. Growth of Cancer Cells through Blood

Cancer cell invasion, proliferation, and metastasis are caused by the degradation of ECM proteins [42]. Many ECM components, such as collagen, laminin, gelatin, fibronectin, elastin, and fibrin, are disrupted by MMPs and are linked to the integrity of the basement membrane. Because of this, MMPs play a critical role in the processes of cancer cell metastasis, which includes the growth of metastatic tumors, basal membrane degradation and subsequent invasion into blood or lymphatic vessels, activation of various growth factors required for angiogenesis, and extravasation into new tissues (Figure 4) [43]. Cancer cells rely on the overexpression of MMP-2 and MMP-9 to travel from the source cells to nearby tissues [43]. Furthermore, the degradation of basement membrane components such as collagen and laminin can cleave endothelial/epithelial cell barrier proteins [44]. When the tumor cells have propagated to a secondary organ, MMPs help to modify the tissue microenvironment to ensure the survival of metastatic cells. By liberating vascular endothelial growth factor (VEGF) from angiogenic islets, MMP-9 is known to initiate the angiogenic migration in cancer, [45] whereas MMP-2 controls the bioavailability of dissolved VEGF-A and tumor vascular patterning [46]. For instance, MMP-9 can cleave the interleukin-2 receptor-α (IL-2R-α) in order to inhibit the growth of lymphocytes called T cells [47]. Several previous studies claim that MMPs inhibit TGF-α activation, thereby decreasing T cells’ capacity to combat tumors [48,49]. It has also been observed that MMPs generate soluble TNF-α, which encourages cancer cells to undergo apoptosis [50]. Blocking MMPs seemed to be a viable strategy against cancer and metastatic growths. MMPs are important regulators for the progression of cancer, particularly metastasis. A number of MMP inhibitors have been practically investigated in the past decade for their potential for curbing metastasis and treating tumors. Marimastat was among of the first MMP inhibitors and underwent phase III clinical trials. The overall survival of patients with pancreatic cancer was found to be improved by Marimastat; however, the side effects of the treatments included musculoskeletal toxicity [51]. Particular MMP inhibitors are currently being developed to combat various cancers; though, blocking MMPs alone may not be enough to stop the progression of cancer.

1.4. Related Diseases

Some metalloproteinase-related polymorphisms and genes have an impact on the development of hypersensitivity pneumonia, an inflammatory illness brought by an overreaction of the immune system to particular organic particle inhalation [52]. Lung function decline appears to be associated with remodeling of the ECM in the airways and respiratory interstice. The variability of circulating active MMP-9 levels is examined during renal replacement therapy (RRT) for a disease that is associated with a high mortality risk and may be made worse by persistent inflammation [53].
Aggressive brain tumors have been revealed to have higher tissue levels of MMP-2 and MMP-9 [54,55,56,57]. Patients with malignant brain tumors also had a lower duration of survival when their tissues had positive levels of MMP-2. Cerebrospinal fluids of patients with brain malignancies have been found to contain both latent and active versions of MMP-2 and MMP-9 [58]. The activity of plasma MMP-2 and MMP-9 is considerably elevated in metastatic prostate cancer [59]. Moreover, upregulation of MMP-2 in tumor cells was linked to reduced disease-free survival. Metalloproteinase (MMP-2 and MMP-9) were found to be important signs of cancer return upon analysis of MMP-2 and MMP-9 levels in radical prostatectomy tissues [60,61]. MMP-9 plays an important role in the onset, progression, and metastasis of gastric [62], lung [63], colon [64], and breast cancers [64]. A high level of MMP-2 and MMP-9 has been found in several cancer types, such as those of the bladder, breast, bronchopulmonary, cervical, colon, glioma, larynx, lung, ovary, melanoma, myeloma, esophagus, pancreas, prostate, skin, and stomach [65].

2. Regulators of MMP-2 and/or MMP-9

2.1. Advantages of Applying Natural Products

Various natural products that are extracted from plants and marine species have potential of medicinal properties. They could be used to cure cancer and other illnesses for a long time because of their effectiveness against various impairments [66]. These compounds’ fabulous offshoots against cancer cells indicate that their oral administration is a tremendous discovery of science. Moreover, it has been revealed that almost 60% of natural products such as flavonoids, tannins, alkaloids, and terpenoids could act as anticarcinogenic agents [67,68]. They can control the growth, migration, and apoptosis of cancer cells by altering metabolic pathways as well as biological systems. Preparing anticarcinogenic medicine in a laboratory is expensive, tedious, and time-consuming, whereas the usage of natural products as drugs is relatively easy [69]. However, researchers take into consideration that natural products may be detrimental to patients due to their potent toxicity concerns [70,71].

2.2. Natural Products from Marine Organisms

Emerging research over the previous fifty years has demonstrated the positive effects of several natural compounds originating from marine life (Table 1) in both the avoidance and amelioration of cancer [72]. New bioactive agents have been abundantly produced by marine natural products [73,74,75]. The sea’s distinct climatic circumstances and variety of marine ecosystems are mostly unexplored sources of new compounds with noteworthy chemical novelty [76,77,78].
Lemnalol (Table 1), a compound that comes from soft corals (Lemnalia cervicornis and Lemnalia tenuis Verseveldt), exhibits anti-inflammatory properties on mast cells (MCs) and osteoclast function. An immunohistochemical analysis revealed that it reduces MC infiltration and degranulation, and this impact may be partially attributed to decreased MMP-9 activity [79]. Ageladine A is one kind of natural product derived from Agelas Nakamura (a marine organism). While the N-methylated derivatives failed to inhibit MMP-9, they suppressed both MMP-2 and MMP-9 proteolytic activity, based on in vitro research on isolated enzymes (Table 1) [80]. Many MMP inhibitors act by chelating out the Zn(II) from the catalytic domain [80].
Additionally, 11-Epi-sinularoide acetate (11-epi-SA) was isolated from a soft coral Sinularia flexibilis (Table 1). In different concentrations, it inhibited cell migration and invasion in hepatocellular carcinoma cells (HA22T cells). MMP-2 and MMP-9 activity and expression levels significantly diminished at non-toxic dosages (7.98 M), indicating that the action was highly related to the regulation of MMPs or their inhibitors [81,82]. Dihydroaustrasulfone alcohol (DA) was extracted from marine coral and exhibits antioxidant as well as anticancer properties (Table 1). Moreover, numerous dosage-dependent studies reveal that it inhibits the invasion and activity of human non-small-cell lung cancer cells (NSCLC A549 cells).
Gelatin zymography analysis also showed significantly decreased levels and actions of MMP-2 and MMP-9. These results established a connection between DA’s antimetastatic effect and the suppression of colonization-related enzymes [83,84].
Marimastat is a major MMP inhibitor, which hinders the activity of MMP-2 and MMP-9. Regardless, the t-butyl and α-hydroxyl groups, respectively, increased solubility in water, and it was demonstrated to be oral permeable [85]. The effectiveness of this medication against models of breast and lung metastases has been shown in preclinical studies [86]. Although it has several harmful side effects, such as cell death, fibrosis, bleeding, and weight loss, it showed fabulous improvement in patients having advanced gastric cancer [86,87]. In particular, Aplysina aerophoba sponges produce the brominated antibiotic aeroplysinin-1, which sets off chemical defense feedback. It could exert antitumor and antiangiogenic effects. MMP-2 levels were lower in a study [88] that used multiple human endothelial cell lines (Table 1). Fluid isolates of Aplysina aerophoba were very effective in reducing protein and mRNA expression rates linked to MMP-2 and MMP-9 in rat astrocyte conditions [89].

2.3. Natural Products from Plants

Many plants contain phenolic substances. A few of these compounds have been connected to invaluable well-being and illness impacts [90]. They showed some fabulous beneficial properties including anticancer, antiviral, and cell reinforcement, as well as energizing vital properties [90]. The sources and effects of natural products on the actions and expression of MMP-2 and MMP-9 are summarized in Table 2.
Recently, multiple studies accomplished research on the anti-inflammatory and anticancer activities of quercetin (Que), which impedes human hepatocarcinoma cell line (HCCLM3 cells) propagation and invasion. Moreover, Que reduces levels of MMP-2 and MMP-9 indicating linking antimigratory and antiinvasive actions [91]. MMP-2 and MMP-9 levels were diminished upon treatment with Que for human oral cancer cells (HSC-6 and SCC-9) [92]. In the case of a cardiopulmonary resuscitation study, rats treated with 50 mg/kg Que for five consecutive days exhibited remarkably fewer ROS, less irritation, and less MMP-2 expression [93]. Hypertension examined using the kidneys of animals who were fed Que for 21 days (10 mg/kg/day) manifested a decrease in artery ROS levels and MMP-2 actions, detected by immunofluorescence and situ zymography methods [94].
Kaempferol (Kae), a bioactive molecule, has some unique features such as cardioprotective, anticancer, antidiabetic, anti-inflammatory, antiaging, and antiallergic properties [95]. It inhibited nuclear translocation of the transcription factor AP-1 to the MMP-2 promoter, decreased MMP-2 production, and prevented migration and invasion in human tongue squamous cell carcinoma cells (SCC4 cells) [96]. Kae is a phytoestrogen slowing the development of cancer and carcinogenesis. It reduces the production of proteins linked to metastasis, such as MMP-2 and MMP-9, in the MCF-7 breast cancer cell line [97].
Naringenin (Nar), extracted from multiple fruits, has anti-inflammatory and anticancer actions. Earlier research looks at how fast Nar changed the proliferation of lung cancer cells (A549 cells). After gelatin zymography, Nar lessens MMP-2 and MMP-9 levels based on its concentration [98]. Furthermore, naringenin treatment significantly reduced the nuclear translocation of NF-κB. Numerous genes, including MMP-2 and MMP-9, are modulated by the transcription factor NF-κB to control a range of cellular functions, for instance, adhesion, inflammation, and cancer metastasis [99]. Luteoin (Lut), mostly discovered in fruits, vegetables, and herbs, has anti-inflammatory and antioxidant features. The effect of Lut on MMP-2 and MMP-9 in the development of colon cancer was induced by azoxymethane (AOM) in BALB/c mice to investigate impacts of Lut on MMP-2 and MMP-9. Initially, MMP-2 and MMP-9 expression was increased, but intraperitoneal Lut administration (15 mg/kg) reduced the level of their expression after 21 days [100]. Recent studies both in vitro and in vivo using subcutaneous injection in nude mice (A2780 cells) have shown that the activity of MMP-2 and MMP-9 declines in ovarian cancer cells [101,102].
Myricetin (Myr) is abundant in vegetables, fruits, beverages, and certain therapeutic plants. Several studies found that it has antioxidant, anti-inflammatory, anticancer, and neurological functions [103]. It fights against cancer cells through several pathways, including (a) control of MMP-2 and MMP-9 activities and (b) reduction of MMP-2 protein production in colorectal cancer cells (COLO 205). Moreover, gelatin gel zymography analysis of isolated MMP-2 [104] utilizing breast cancer cells (MDA-Mb-231) indicates a strong and direct interaction between Myr and MMP-2 and blocks breast cancer metastasis by lowering the action of MMP-2 and MMP-9 [105].
Another study looked at how Myr affected the growth and spread of radioresistant lung cancer cells (A549-IR cells) via the suppression of MMP-2 and MMP-9 expression. Experimental data have shown that Myr prevents the growth and migration of cancer in A549 cells [106].
Scutellaria baicalensis Georgi (Lamiaceae) generates a bioactive baicalein (Bai) used as traditional Chinese medicine. With its antioxidant, antiviral, anti-inflammatory, antiangiogenic, and anticancer properties, Bai has a multitude of beneficial advantages [107]. Numerous investigations reveal that Bai functions as an antitumor agent via multiple mechanisms, including the regulation of MMP-2 and MMP-9 expression [108]. Further research on the anti-proliferative properties of Bai in melanoma cell lines (A375 and SK-MEL-28) revealed a significant reduction in MMP-2 expression in Bai-treated cells [109]. Moreover, Bai research on pancreatic neuroendocrine tumor cell line BON1 revealed a correlation between decreased MMP-2 and MMP-9 levels and a reduction in tumor invasion and migration [110]. The latest findings indicate that Bai contributes to the prevention of osteosarcoma metastasis. Following treatment with Bai, the invasive capacity of human osteosarcoma cells (CRL-1427 cells) was reduced as a consequence of low production of MMP-9 and MMP-2 [111]. Genistein is a flavonoid isolated from the Leguminosae Genistin rhizome. Its effects include antitumor, antibacterial, antioxidant, hypolipidemic, and estrogen-like properties. It may show beneficial antitumor effect by inhibiting angiogenesis and inducing tumor cell programmed death. Genistein had the potential to stop the human colon cancer cell line HCT116 from propagating because it is a potent MMP-9 inhibitor that prevents the growth and spread of tumors investigated in vivo tests in mice [112]. Another naturally occurring MMP inhibitor is Silibinin isolate from milk thistle seeds, which has antioxidant and anticarcinogenic properties. It behaves as a chemopreventive agent to stop skin cancer and influences how breast cancer metastasizes. By inhibiting the MEK/ERK cascade in a dose-sensitive way, it suppresses the production of MMP-9 in mice. Silibinin also inhibits cancer cell migration, and MMP-9 expression in thyroid and breast cancer cells is reduced by protecting the ECM [113,114,115].
The compounds that appear to be active in Panax root are termed ginsenosides (GSs). They are a class of glycosylated triterpenes, which could be referred to as saponins. Some studies highlight the diverse medicinal benefits of GSs on heart disease and hormonal, immunological, and neurological functions [116,117,118]. Additionally, it has been proved that GSs suppress the proliferation of cancers via transforming MMP-2 and MMP-9 [119,120]. Blocking the expression of these enzymes could inhibit the formation of cancerous tumors. Due to their ability to reduce MMP levels, GSs have the potential to be effective cancer chemopreventive medications [119,120,121,122]. Given that MMPs have a widely recognized influence on adipogenesis, GSs could modulate MMP activity and reduce adipogenesis in 3T3-L1 adipocytes [116].
Amenoflavone is a polyphenolic constituent that was found in Selaginella tamariscina and has been demonstrated to possess a variety of therapeutic properties [123,124]. These properties include neuroprotective, antiallergic, antioxidant, and antitumor properties. Amentoflavone suppresses NF-κB activation and modifies the expression of antimetastatic proteins, following in vivo experiments conducted on B16F-10 melanoma cells [125]. Amentoflavone-induced cell cycle depletion and apoptosis, which had an impact on the respiratory system in vitro in breast cancer cells, may limit the proliferation of cells [124]. The antimetastatic actions of amentoflavone by inhibition of MMPs, especially MMP-2 and MMP-9, have been believed to be involved in tumor progression, partly because of their ability to degrade collagen type IV, one of the major components of basement membranes [126].
Sulforaphane is a naturally found in large amounts in cruciferous vegetables, such as cabbage and broccoli, and has anticancer and anti-inflammatory properties [127]. It has been reported that consuming cruciferous vegetables could prevent stomach cancer. Sulforaphane is currently observed to initiate apoptosis and block the cell cycle of human colorectal cancer [128]. In addition, sulforaphane may make hepatoma cancer cells more susceptible to TRAIL-induced apoptosis through DR5 overexpression driven by ROS [129]. Recently, fundamental processes of the influence of sulforaphane on the nicotine-mediated stimulation of MMP-9 has been reported. It suggests sulforaphane impedes the nicotine-related MMP-9 activity by blocking the ROS-mediated NF-κB and MAPK (p38 MAPK, Erk1/2)/AP-1 signaling pathways that serve to treat abdominal cancer cells in humans [130].
Caffeic acid (CA), a strong and selective MMP-9 activity and transcription inhibitor, was isolated from the plant Euonymus alatus [133]. It is a common phenolic acid found naturally in fruits, vegetables, wine, olive oil, and coffee [131]. It has antioxidant, antiallergic, and antiproliferative functions. Caffeine has been demonstrated to have antioxidant qualities in normal cells and pro-oxidant characteristics in cancer cells [132]. Ceramide-induced NF-κB-binding capability is effectively inhibited by CA [133]. According to recent studies, CA inhibits MMP-9 enzymatic activity and gene expression, both of which are critical for colon cancer invasion and metastasis [134].
Combitastatins are derived from the willow tree Combretum caffrum in South Africa. Among combitastatins, Combretastatin A-4 (CA-4) is the most popular product that can bind to tubulin MMP-2 and MMP-9 [135]. CA-4 has antimetastatic qualities and could downregulate the production of MMPs, mostly MMP-2 and MMP-9, in A549 cells [136,137,138,139,140]. Naturally occurring plant-based compounds have recently drawn more interest due to their potential role as cancer preventatives for those who have substantial cancer risk [141,142]. The main sources of pterostilbene are blueberries and several grape varieties [143]. Pterostilbene includes similar features to resveratrol, such as antiproliferative, anti-inflammatory, anticancer, and antioxidant properties [144,145,146]. A recent study showed that pterostilbene inhibited the generation of MMP-2 and decreased the capacity of the cells to migrate via stimulating Erk1/2 in A7r5 cells [147]. Decursin, a coumarin derivative present in the roots of Angelica gigas, has long been utilized for remedying anemia [148]. Cell cycle death appears when it comes into contact with breast, prostate, bladder, and colon cancer cells [149,150,151]. According to recent studies, decursin inhibits the expression of NF-κB in breast cells and macrophages hence limiting MMP-9 generation [151,152,153].
Researchers’ interest in natural products has grown significantly in recent years because of their possible chemopreventive roles in human health and illnesses. Marine- and plant-based natural products are known to have significant antioxidant properties that fight against cellular oxidative stress and ROS, lowering the risk of diseases. Additionally, these natural products could contribute to the development of treatments for many chronic ailments including infectious diseases, cancer, diabetes, cardiovascular diseases (CVDs), neurodegenerative diseases, and inflammatory disorders [154]. In this regard, we emphasized and summarized some natural products and their mechanisms of action against a range of serious human illnesses.

2.4. Mechanism of MMP Regulation by Natural Products

Over two decades, it has been demonstrated that cell inactivation could be a barrier to curing cancer [155]. It is widely known that MMPs are responsible for cell death because they induce metastasis and invasion of normal cells by degrading the ECM as well as the basement membrane [156]. The Chinese started to use the natural product Bai, which was extracted from the roots of Scutelaria baicalensis as an anticarcinogenic agent. Natural MMP regulators could deteriorate the activity and expression of MMPs in order to cure cancers after taking them in a daily diet. These active ingredients pass through the cell membrane and interact directly or indirectly with MMPs [157,158,159,160,161]. After revealing positive aspects of MMP regulation by Bai, other nations began to widely appreciate Bai as well as take it as a drug [156]. Although the mechanism of action relies on the type of natural products, the proposed suppression pathways of activity of natural products against MMPs are briefly described. The natural products enlisted in this review could regulate the expression and activity of MMP-2 and MMP-9 through (a) breaking hydrogen bonds of A-T or C-T of DNA and distortion of MMPs geometry around the Zn(II) center [162,163,164], (b) damaging the chromosomal sequence of DNA by generating ROS [165,166,167,168], (c) activating PKC-δ with the help of NADPH and ROS generation to activate p38-MARK [169,170,171], (d) inactivating the vascular endothelial growth factor (VEGF) [172], and (e) suppressing NF-κB signaling [173,174,175].

2.5. Difficulties of Natural Products Using as Medications

Natural products have been used extensively to treat cancer. Nevertheless, their poor pharmacokinetic traits, low water solubility, low biocompatibility, limited oral bioavailability, and volatility make it difficult to employ them in clinical applications on their own [176]. Furthermore, there is little yield from spontaneous isolation of natural products [177]. Additionally, using these natural products frequently results in serious adverse consequences, such as myelosuppression, dizziness, vomiting, stomatitis, exhaustion, diarrhea, gastrointestinal pain, peripheral nerve damage, and baldness [176,177]. Despite these difficulties, natural products still have potential as medications for various diseases, including cancers as summarized above.

3. Conclusions and Future Prospects

This study summarized how natural products from natural fruits and vegetables as well as marine organisms have been shown to have positive impacts on cancer by downregulating the actions and/or levels of MMP-2 and MMP-9. The therapeutic effects of numerous natural products obtained from botanical and aqu atic sources in the recovery from deadly human cancers have been studied. As MMPs are implicated in the development of such disorders, a variety of natural products have been used to control MMP generation and activity. Multiple natural products have been identified as powerful inhibitors of MMP-2 and MMP-9 expression. Chemoresistance has been commonly endured by many traditional chemotherapeutics, which is the main concern and hurdle in cancer therapy along with radiation resistance. Natural products are ideal candidates for chemosensitizing cancer cells and increasing the efficacy of existing drugs. Furthermore, additional research is required to pinpoint particular regulatory systems downregulating the activity and/or upregulating the expression levels of MMP-2 and MMP-9, governed by natural products.

Author Contributions

Ideas, M.T.I. and H.J.L.; Writing—original draft, M.T.I. and N.H.J.; Literature search, M.T.I., N.H.J. and H.J.L.; Writing—review and editing, N.H.J. and H.J.L.; Supervision, Project administration, and Funding acquisition, H.J.L.; Funding acquisition, H.J.L. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIT) (2022R1F1A1074609 to H.J.L.).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–464. [Google Scholar] [CrossRef] [PubMed]
  2. Torre, L.A.; Bray, F.; Siegel, R.L.; Ferlay, J.; Lortet-Tieulent, J.; Jemal, A. Global cancer statistics, 2012. CA Cancer J. Clin. 2015, 65, 87–108. [Google Scholar] [CrossRef] [PubMed]
  3. Sung, H.; Ferlay, J.; Siege, R.L. Global Cancer Statistics 2020 GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA. Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  4. Chen, S.; Cao, Z.; Prettner, K.; Kuhn, M.; Yang, J.; Jiao, L.; Wang, Z.; Li, W.; Geldsetzer, P.; Bärnighausen, T.; et al. Estimates and projections of the global economic cost of 29 cancers in 204 countries and territories from 2020 to 2050. JAMA Oncol. 2023, 9, 465–472. [Google Scholar] [CrossRef] [PubMed]
  5. Baig, M.H. Enzyme targeting strategies for prevention and treatment of cancer: Implications for cancer therapy. Semin. Cancer Biol. 2019, 56, 1–11. [Google Scholar] [CrossRef] [PubMed]
  6. Campagna, R.; Vignini, A. NAD+ Homeostasis and NAD+-Consuming Enzymes: Implications for Vascular Health. Antioxidants 2023, 12, 376. [Google Scholar] [CrossRef] [PubMed]
  7. Lynch, H.T.; de la Chapelle, A. Hereditary Colorectal Cancer. N. Eng. J. Med. 2003, 348, 919–932. [Google Scholar] [CrossRef] [PubMed]
  8. Ford, D.; Easton, D.F.; Peto, J. Estimates of the gene frequency of BRCA1 and its contribution to breast and ovarian cancer incidence. Am. J. Hum. Genet. 1995, 57, 1457–1462. [Google Scholar] [PubMed]
  9. Eala, M.A.B.; Robredo, J.P.G.; Dee, E.C.; Lin, V.; Lagmay, A.M.F.A. Climate crisis and cancer: Perspectives from the hardest hit. Lancet Oncol. 2022, 23, e92. [Google Scholar] [CrossRef]
  10. Kanis, M.J.; Kolev, V.; Getrajdman, J.; Zakashansky, K.; Cohen, C.; Rahaman, J. Carcinosarcoma of the ovary: A single institution experience and review of the literature. Eur. J. Gynaecol. Oncol. 2016, 37, 75–79. [Google Scholar]
  11. Miller, K.D. Cancer treatment and survivorship statistics, 2019. CA Cancer J. Clin. 2019, 69, 363–385. [Google Scholar] [CrossRef] [PubMed]
  12. Murphy, G. The ADAMs: Signalling scissors in the tumour microenvironment. Nat. Rev. Cancer 2008, 8, 932–941. [Google Scholar] [CrossRef]
  13. Coussens, L.M.; Fingleton, B.; Matrisian, L.M. Matrix metalloproteinase inhibitors and cancer: Trials and tribulations. Science 2002, 295, 2387–2392. [Google Scholar] [CrossRef] [PubMed]
  14. Egeblad, M.; Werb, Z. New functions for the matrix metalloproteinases in cancer progression. Nat. Rev. Cancer. 2002, 2, 161–174. [Google Scholar] [CrossRef]
  15. Huang, G. alpha3(V) collagen is critical for glucose homeostasis in mice due to defects in pancreatic islets and peripheral tissues. J. Clin. Investig. 2011, 121, 769–783. [Google Scholar] [CrossRef]
  16. Liu, X. Type I collagen promotes the migration and myogenic diferentiation of C2C12 myoblasts via the release of interleukin-6 mediated by FAK/NF-kappaB p65 activation. Food Funct. 2020, 11, 328–338. [Google Scholar] [CrossRef] [PubMed]
  17. Kai, F.; Drain, A.P.; Weaver, V.M. The Extracellular Matrix Modulates the Metastatic Journey. Dev. Cell. 2019, 49, 332–346. [Google Scholar] [CrossRef]
  18. Thiery, J.P. Epithelial–mesenchymal transitions in tumour progression. Nat. Rev. Cancer. 2002, 2, 442–454. [Google Scholar] [CrossRef]
  19. Yu, D. Quercetin inhibits Epithelial—Mesenchymal transition, decreases invasiveness and metastasis, and reverses IL-6 induced Epithelial—Mesenchymal transition, expression of MMP by inhibiting STAT3 signaling in pancreatic cancer cells. OncoTargets Ther. 2017, 10, 4719–4729. [Google Scholar] [CrossRef]
  20. Nagase, H.; Visse, R.; Murphy, G. Structure and function of matrix metalloproteinases and TIMPs. Cardiovasc. Res. 2006, 69, 562–573. [Google Scholar] [CrossRef]
  21. Corzo, L.; Fernández-Novoa, L.; Carrera, I.; Martínez, O.; Rodríguez, S.; Alejo, R.; Cacabelos, R. Nutrition, health, and disease: Role of selected marine and vegetal nutraceuticals. Nutrients 2020, 12, 747. [Google Scholar] [CrossRef] [PubMed]
  22. Shi, Y.; Ma, X.; Fang, G.; Tian, X.; Ge, C. Matrix metalloproteinase inhibitors (MMPIs) as attractive therapeutic targets: Recent progress and current challenges. NanoImpact 2021, 21, 100293. [Google Scholar] [CrossRef] [PubMed]
  23. Winer, A.; Adams, S.; Mignatti, P. Matrix metalloproteinase inhibitors in cancer therapy: Turning past failures into future successes. Mol. Cancer Ther. 2018, 17, 1147–1155. [Google Scholar] [CrossRef] [PubMed]
  24. Choi, H.; Kim, E.; Choi, J.Y.; Park, E.; Lee, H.J. Potent therapeutic targets for treatment of Alzheimer’s disease: Amyloid degrading enzymes. Bull. Korean Chem. Soc. 2021, 42, 1419–1429. [Google Scholar] [CrossRef]
  25. Zucker, S.; Vacirca, J. Role of matrix metalloproteinases (MMPs) in colorectal cancer. Cancer Metastasis Rev. 2004, 23, 101–117. [Google Scholar] [CrossRef] [PubMed]
  26. Boon, L.; Ugarte-Berzal, E.; Vandooren, J.; Opdenakker, G. Protease propeptide structures, mechanisms of activation, and functions. Crit. Rev. Biochem. Mol. Biol. 2020, 55, 111–165. [Google Scholar] [CrossRef] [PubMed]
  27. Steffensen, B.; Wallon, U.M.; Overall, C.M. Extracellular Matrix Binding Properties of Recombinant Fibronectin Type II-like Modules of Human 72-kDa Gelatinase/Type IV Collagenase. J. Biol. Chem. 1995, 270, 11555–11566. [Google Scholar] [CrossRef] [PubMed]
  28. Roeb, E. The Matrix Metalloproteinase 9 (MMP-9) Hemopexin Domain Is a Novel Gelatin Binding Domain and Acts as an Antagonist. J. Biol. Chem. 2002, 277, 50326–50332. [Google Scholar] [CrossRef]
  29. Goldberg, G.I.; Strongin, A.; Collier, I.E.; Genrich, L.T.; Marmer, B.L. Interaction of 92-kDa type IV collagenase with the tissue inhibitor of metalloproteinases prevents dimerization, complex formation with interstitial collagenase, and activation of the proenzyme with stromelysin. J. Biol. Chem. 1992, 267, 4583–4591. [Google Scholar] [CrossRef]
  30. Galea, C.A.; Nguyen, H.M.; George Chandy, K.; Smith, B.J.; Norton, R.S. Domain structure and function of matrix metalloprotease 23 (MMP23): Role in potassium channel trafficking. Cell. Mol. Life Sci. 2014, 71, 1191–1210. [Google Scholar] [CrossRef]
  31. Pei, D.; Kang, T.; Qi, H. Cysteine array matrix metalloproteinase (CA-MMP)/ MMP-23 is a type II transmembrane matrix metalloproteinase regulated by a single cleavage for both secretion and activation. J. Biol. Chem. 2000, 275, 33988–33997. [Google Scholar] [CrossRef] [PubMed]
  32. Alaseem, A.; Alhazzani, K.; Dondapati, P.; Alobid, S.; Bishayee, A.; Rathinavelu, A. Matrix Metalloproteinases: A challenging paradigm of cancer management. Semin. Cancer Biol. 2019, 56, 100–115. [Google Scholar] [CrossRef] [PubMed]
  33. Jeleniewicz, W. MMP-2 mRNA Expression in Ovarian Cancer Tissues Predicts Patients Response to Platinum-Taxane Chemotherapy. Anticancer. Res. 2019, 39, 1821–1827. [Google Scholar] [CrossRef]
  34. Li, H.; Qiu, Z.; Li, F.; Wang, C. The relationship between MMP-2 and MMP-9 expression levels with breast cancer incidence and prognosis. Oncol. Lett. 2017, 14, 5865–5870. [Google Scholar] [CrossRef] [PubMed]
  35. Bode, W.; Fernandez-Catalan, C.; Tschesche, H.; Grams, F.; Nagase, H.; Maskos, H.N.K. Structural properties of matrix metalloproteinases. Cell. Mol. Life Sci. 1999, 55, 639–652. [Google Scholar] [CrossRef] [PubMed]
  36. Lu, P.; Takai, K.; Weaver, V.M.; Werb, Z. Extracellular Matrix Degradation and Remodeling in Development and Disease. Cold Spring Harb. Perspect. Biol. 2011, 3, a005058. [Google Scholar] [CrossRef] [PubMed]
  37. Cabral-Pacheco, G.A. The Roles of Matrix Metalloproteinases and Their Inhibitors in Human Diseases. Int. J. Mol. Sci. 2020, 21, 9739. [Google Scholar] [CrossRef] [PubMed]
  38. Kessenbrock, K.; Plaks, V.; Werb, Z. Matrix Metalloproteinases: Regulators of the Tumor Microenvironment. Cell 2010, 141, 52–67. [Google Scholar] [CrossRef] [PubMed]
  39. Bonnans, C.; Chou, J.; Werb, Z. Remodelling the extracellular matrix in development and disease. Nat. Rev. Mol. Cell. Biol. 2014, 15, 786–801. [Google Scholar] [CrossRef]
  40. Singh, D.; Srivastava, S.K.; Chaudhuri, T.K.; Upadhyay, G. Multifaceted role of matrix metalloproteinases (MMPs). Front. Mol. Biosci. 2015, 2, 19. [Google Scholar] [CrossRef]
  41. Jiang, Y.; Goldberg, I.D.; Shi, Y.E. Complex roles of tissue inhibitors of metalloproteinases in cancer. Oncogene 2002, 21, 2245–2252. [Google Scholar] [CrossRef] [PubMed]
  42. Verma, S. Matrix metalloproteinases and gastrointestinal cancers: Impacts of dietary antioxidants. World J. Biol. Chem. 2014, 5, 355. [Google Scholar] [CrossRef] [PubMed]
  43. Deryugina, E.I.; Quigley, J.P. Matrix metalloproteinases and tumor metastasis. Cancer Metastasis Rev. 2006, 25, 9–34. [Google Scholar] [CrossRef] [PubMed]
  44. Ichikawa, Y.; Ishikawa, T.; Momiyama, N.; Kamiyama, M.; Sakurada, H.; Matsuyama, R.; Hasegawa, S.; Chishima, T.; Hamaguchi, Y.; Fujii, S.; et al. Matrilysin (MMP-7) degrades VE-cadherin and accelerates accumulation of beta-catenin in the nucleus of human umbilical vein endothelial cells. Oncol. Rep. 2006, 2, 737–744. [Google Scholar] [CrossRef]
  45. Bergers, G. Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nat. Cell Biol. 2000, 2, 737–744. [Google Scholar] [CrossRef] [PubMed]
  46. Lee, S.; Jilani, S.M.; Nikolova, G.V.; Carpizo, D.; Iruela-Arispe, M.L. Processing of VEGF-A by matrix metalloproteinases regulates bioavailability and vascular patterning in tumors. J. Cell Biol. 2005, 169, 681–691. [Google Scholar] [CrossRef] [PubMed]
  47. Hsu, S.M.; Ho, H.N.; Lien, H.C.; Huang, S.C.; Lin, R.H. A novel role of metalloproteinase in cancer-mediated immunosuppression. Cancer Res. 2001, 61, 237–242. [Google Scholar]
  48. Yu, Q.; Stamenkovic, I. Cell surface-localized matrix metalloproteinase-9 proteolytically activates TGF-β and promotes tumor invasion and angiogenesis. Genes Dev. 2000, 14, 163–176. [Google Scholar] [CrossRef]
  49. Gorelik, L.; Flavell, R.A. Immune-mediated eradication of tumors through the blockade of transforming growth factor-β signaling in T cells. Nat. Med. 2001, 7, 1118–1122. [Google Scholar] [CrossRef]
  50. Wang, X.; Lin, Y. Tumor necrosis factor and cancer, buddies or foes? Acta Pharmacol. Sin. 2008, 29, 1275–1288. [Google Scholar] [CrossRef]
  51. Bramhall, S.R.; Rosemurgy, A.; Brown, P.D.; Bowry, C.; Buckels, J.A.C. Marimastat as First-Line Therapy for Patients with Unresectable Pancreatic Cancer: A Randomized Trial. J. Clin. Oncol. 2001, 19, 3447–3455. [Google Scholar] [CrossRef] [PubMed]
  52. Santiago-Ruiz, L.; Buendía-Roldán, I.; Pérez-Rubio, G.; Ambrocio-Ortiz, E.; Mejía, M.; Montaño, M.; Falfán-Valencia, R. MMP2 Polymorphism Affects Plasma Matrix Metalloproteinase (MMP)-2 Levels, and Correlates with the Decline in Lung Function in Hypersensitivity Pneumonitis Positive to Autoantibodies Patients. Biomolecules 2009, 9, 574. [Google Scholar] [CrossRef] [PubMed]
  53. Rodríguez-Sánchez, E. Variations in Circulating Active MMP-9 Levels during Renal Replacement Therapy. Biomolecules 2020, 10, 505. [Google Scholar] [CrossRef] [PubMed]
  54. Sawaya, R.E. Expression and localization of 72 kDa type IV collagenase (MMP-2) in human malignant gliomas in vivo. Clin. Exp. Metastasis. 1996, 14, 35–42. [Google Scholar] [CrossRef] [PubMed]
  55. Jäälinojä, J.; Herva, R.; Korpela, M.; Höyhtyä, M.; Turpeenniemi-Hujanen, T. Matrix metalloproteinase 2 (MMP-2) immunoreactive protein is associated with poor grade and survival in brain neoplasms. J. Neurooncol. 2000, 46, 81–90. [Google Scholar] [CrossRef] [PubMed]
  56. Rao, J.S. Expression and localization of 92 kDa type IV collagenase/gelatinase B (MMP-9) in human gliomas. Clin. Exp. Metastasis 1996, 14, 12–18. [Google Scholar] [CrossRef] [PubMed]
  57. Choe, G.; Park, J.K.; Jouben-Steele, L. Activematrix metalloproteinase 9 expression is associated with primary glioblastoma subtype. Clin. Cancer Res. 2002, 8, 2894–2901. [Google Scholar] [PubMed]
  58. Friedberg, M.H.; Glantz, M.J.; Klempner, M.S.; Cole, B.F.; Perides, G. Specific matrix metalloproteinase profiles in the cerebrospinal fluid correlated with the presence of malignant astrocytomas, brain metastases, and carcinomatous meningitis. Cancer 1998, 82, 923–930. [Google Scholar] [CrossRef]
  59. Morgia, G. Matrix metalloproteinases as diagnostic (MMP-13) and prognostic (MMP-2, MMP-9) markers of prostate cancer. Urol. Res. 2005, 33, 44–50. [Google Scholar] [CrossRef]
  60. Miyake, H.; Muramaki, M.; Kurahashi, T.; Takenaka, A.; Fujisawa, M. Expression of potential molecular markers in prostate cancer: Correlation with clinicopathological outcomes in patients undergoing radical prostatectomy. Urol. Oncol. 2010, 28, 145–151. [Google Scholar] [CrossRef]
  61. Trudel, D.; Fradet, Y.; Meyer, F.; Harel, F.; Têtu, B. Membrane-type-1 matrix metalloproteinase, matrix metalloproteinase 2, and tissue inhibitor of matrix proteinase 2 in prostate cancer: Identification of patients with poor prognosis by immunohistochemistry. Hum. Pathol. 2008, 39, 731–739. [Google Scholar] [CrossRef] [PubMed]
  62. Liu, L.; Ye, Y.; Zhu, X. MMP-9 secreted by tumor associated macrophages promoted gastric cancer metastasis through a PI3K/AKT/Snail pathway. Biomed. Pharmacother. 2019, 117, 109096. [Google Scholar] [CrossRef] [PubMed]
  63. Zhang, H.; Zhao, B.; Zhai, Z.G.; Zheng, J.D.; Wang, Y.K.; Zhao, Y.Y. Expression and clinical significance of MMP-9 and P53 in lung cancer. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 1358–1365. [Google Scholar] [PubMed]
  64. Ruan, P.; Gu, Y.; Zhou, H. Expression and clinical significance of CD74 and MMP-9 in colon adenocarcinomas. J. BUON 2020, 25, 927–932. [Google Scholar]
  65. Ran, Z.; Hou, L.; Guo, H.; Wang, K.; Li, X. Expression of VEGF, COX-2 and MMP-9 in breast cancer and their relationship with ultrasound findings. Int. J. Clin. Exp. Pathol. 2018, 11, 4264–4269. [Google Scholar] [PubMed]
  66. Choudhari, A.S.; Mandave, P.C.; Deshpande, M.; Ranjekar, P.; Prakash, O. Phytochemicals in Cancer Treatment: From Preclinical Studies to Clinical Practice. Front. Pharmacol. 2020, 10, 1614. [Google Scholar] [CrossRef] [PubMed]
  67. Rayan, A.; Raiyn, J.; Falah, M. Nature is the best source of anticancer drugs: Indexing natural products for their anticancer bioactivity. PLoS ONE 2017, 12, e0187925. [Google Scholar] [CrossRef]
  68. Mamta, S.; Saxena, J.; Rajeev, N.; Singh, D.; Gupta, A. Phytochemistry of medicinal plants. J. Pharmacogn. Phytochem. 2013, 1, 8192. [Google Scholar]
  69. Hughes, J.; Rees, S.; Kalindjian, S.; Philpott, K. Principles of early drug discovery. Br. J. Pharmacol. 2011, 162, 1239–1249. [Google Scholar] [CrossRef]
  70. Ahuja, V.; Bokan, S.; Sharma, S. Predicting toxicities in humans by nonclinical safety testing: An update with particular reference to anticancer compounds. Drug Discov. Today 2017, 22, 127–132. [Google Scholar] [CrossRef]
  71. Tewari, D.; Rawat, P.; Singh, P.K. Adverse drug reactions of anticancer drugs derived from natural sources. Food Chem. Toxicol. 2019, 123, 522–535. [Google Scholar] [CrossRef] [PubMed]
  72. Dyshlovoy, S.A.; Honecker, F. Marine compounds and cancer: 2017 updates. Mar. Drugs 2018, 16, 41. [Google Scholar] [CrossRef] [PubMed]
  73. Mayer, A.M.; Hamann, M.T. Marine pharmacology in 2001–2002: Marine compounds with anthelmintic, antibacterial, anticoagulant, antidiabetic, antifungal, anti-inflammatory, antimalarial, antiplatelet, antiprotozoal, antituberculosis, and antiviral activities; affecting the cardiovascular, immune and nervous systems and other miscellaneous mechanisms of action. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2005, 140, 265–286. [Google Scholar] [PubMed]
  74. Mayer, A.M.; Rodríguez, A.D.; Berlinck, R.G.; Fusetani, N. Marine pharmacology in 2007–8: Marine compounds with antibacterial, anticoagulant, antifungal, anti-inflammatory, antimalarial, antiprotozoal, antituberculosis, and antiviral activities; affecting the immune and nervous system, and other miscellaneous mechanisms of action. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2011, 153, 191–222. [Google Scholar] [PubMed]
  75. Veiga-Santos, P.; Pelizzaro-Rocha, K.J.; Santos, A.O.; Ueda-Nakamura, T.; Dias Filho, B.P.; Silva, S.O.; Sudatti, D.B.; Bianco, E.M.; RPereira, C.; Nakamura, C.V. In vitro anti-trypanosomal activity of elatol isolated from red seaweed Laurencia dendroidea. Parasitology 2010, 137, 1661–1670. [Google Scholar] [CrossRef] [PubMed]
  76. Gerwick, W.H.; Moore, B.S. Lessons from the past and charting the future of marine natural products drug discovery and chemical biology. Chem. Biol. 2012, 19, 85–98. [Google Scholar] [CrossRef] [PubMed]
  77. Haefner, B. Drugs from the deep: Marine natural products as drug candidates. Drug Discov. Today 2023, 8, 536–544. [Google Scholar] [CrossRef] [PubMed]
  78. Simmons, T.L.; Andrianasolo, E.; McPhail, K.; Flatt, P.; Gerwick, W.H. Marine natural products as anticancer drugs. Mol. Cancer Ther. 2005, 4, 333–342. [Google Scholar] [CrossRef] [PubMed]
  79. Lee, H.P.; Lin, Y.Y.; Duh, C.Y.; Huang, S.Y.; Wang, H.M.; Wu, S.F.; Lin, S.C.; Jean, Y.H.; Wen, Z.H. Lemnalol attenuates mast cell activation and osteoclast activity in a gouty arthritis model. J. Pharm. Pharmacol. 2015, 67, 274–285. [Google Scholar] [CrossRef]
  80. Fujita, M.; Nakao, Y.; Matsunaga, S.; Seiki, M.; Itoh, Y.; Yamashita, J.; Van Soest, R.W.; Fusetani, N. Ageladine A: An Antiangiogenic Matrixmetalloproteinase Inhibitor from the Marine Sponge Agelas N Akamurai. J. Am. Chem. Soc. 2003, 125, 15700–15701. [Google Scholar] [CrossRef]
  81. Lin, J.J.; Su, J.H.; Tsai, C.C.; Chen, Y.J.; Liao, M.H.; Wu, Y.J. 11-epi-Sinulariolide acetate reduces cell migration and invasion of human hepatocellular carcinoma by reducing the activation of ERK1/2, p38MAPK and FAK/PI3K/AKT/mTOR signaling pathways. Mar. Drugs 2014, 12, 4783–4798. [Google Scholar] [CrossRef] [PubMed]
  82. Cheng, T.C.; Din, Z.H.; Su, J.H.; Wu, Y.J.; Liu, C.I. Sinulariolide suppresses cell migration and invasion by inhibiting matrix metalloproteinase-2/-9 and urokinase through the PI3K/AKT/mTOR signaling pathway in human bladder cancer cells. Mar. Drugs 2017, 15, 238. [Google Scholar] [CrossRef] [PubMed]
  83. Chen, S.C.; Chien, Y.C.; Pan, C.H.; Sheu, J.H.; Chen, C.Y.; Wu, C.H. Inhibitory effect of dihydroaustrasulfone alcohol on the migration of human non-small cell lung carcinoma A549 cells and the antitumor effect on a Lewis lung carcinoma-bearing tumor model in C57BL/6J mice. Mar. Drugs 2014, 12, 196–213. [Google Scholar] [CrossRef] [PubMed]
  84. Li, P.C.; Sheu, M.J.; Ma, W.F.; Pan, C.H.; Sheu, J.H.; Wu, C.H. Anti-restenotic roles of dihydroaustrasulfone alcohol involved in inhibiting PDGF-BB-stimulated proliferation and migration of vascular smooth muscle cells. Mar. Drugs 2015, 13, 3046–3060. [Google Scholar] [CrossRef] [PubMed]
  85. Whittaker, M.; Floyd, C.D.; Brown, P.; Gearing, A.J. Design and Therapeutic Application of Matrix Metalloproteinase Inhibitors. Chem. Rev. 2001, 101, 2205–2206. [Google Scholar] [CrossRef] [PubMed]
  86. Yip, D.; Ahmad, A.; Karapetis, C.S.; Hawkins, C.A.; Harper, P.G. Matrix metalloproteinase inhibitors: Applications in oncology. Investig. New Drugs 1999, 17, 387–399. [Google Scholar] [CrossRef] [PubMed]
  87. Nelson, A.R.; Fingleton, B.; Rothenberg, M.L.; Matrisian, L.M. Matrix metalloproteinases: Biologic activity and clinical implications. J. Clin. Oncol. 2000, 18, 1135–1149. [Google Scholar] [CrossRef] [PubMed]
  88. Martínez-Poveda, B.; García-Vilas, J.A.; Cardenas, C.; Melgarejo, E.; Quesada, A.R.; Medina, M.A. The brominated compound aeroplysinin-1 inhibits proliferation and the expression of key pro-inflammatory molecules in human endothelial and monocyte cells. PLoS ONE 2013, 8, e55203. [Google Scholar] [CrossRef] [PubMed]
  89. Di Bari, G.; Gentile, E.; Latronico, T.; Corriero, G.; Fasano, A.; Nonnis Marzano, C.; Liuzzi, G.M. Inhibitory effect of aqueous extracts from marine sponges on the activity and expression of gelatinases A (MMP-2) and B (MMP-9) in rat astrocyte cultures. PLoS ONE 2015, 10, e0129322. [Google Scholar] [CrossRef]
  90. Andrade, S.; Ramalho, M.J.; Loureiro, J.A.; Pereira, M.D. Natural compounds for Alzheimer’s disease therapy: A systematic review of preclinical and clinical studies. Int. J. Mol. Sci. 2019, 20, 2313. [Google Scholar] [CrossRef]
  91. Lu, J.; Wang, Z.; Li, S.; Xin, Q.; Yuan, M.; Li, H.; Song, X.; Gao, H.; Pervaiz, N.; Sun, X.; et al. Quercetin inhibits the migration and invasion of HCCLM3 cells by suppressing the expression of p-Akt1, matrix metalloproteinase (MMP) MMP-2, and MMP-9. Medical science monitor: Int. J. Clin. Exp. Med. Res. 2018, 24, 2583. [Google Scholar] [CrossRef] [PubMed]
  92. Zhao, J.; Fang, Z.; Zha, Z.; Sun, Q.; Wang, H.; Sun, M.; Qiao, B. Quercetin inhibits cell viability, migration and invasion by regulating miR-16/HOXA10 axis in oral cancer. Eur. J. Pharmacol. 2019, 847, 11–18. [Google Scholar] [CrossRef] [PubMed]
  93. Wang, D.; Lou, X.; Jiang, X.M.; Yang, C.; Liu, X.L.; Zhang, N. Quercetin protects against inflammation, MMP-2 activation and apoptosis induction in rat model of cardiopulmonary resuscitation through modulating Bmi-1 expression. Mol. Med. Rep. 2018, 18, 610–616. [Google Scholar] [CrossRef] [PubMed]
  94. Pereira, S.C.; Parente, J.M.; Belo, V.A.; Mendes, A.S.; Gonzaga, N.A.; do Vale, G.T.; Ceron, C.S.; Tanus-Santos, J.E.; Tirapelli, C.R.; Castro, M.M. Quercetin decreases the activity of matrix metalloproteinase-2 and ameliorates vascular remodeling in renovascular hypertension. Atherosclerosis 2018, 270, 146–153. [Google Scholar] [CrossRef] [PubMed]
  95. Imran, M.; Rauf, A.; Shah, Z.A.; Saeed, F.; Imran, A.; Arshad, M.U.; Ahmad, B.; Bawazeer, S.; Atif, M.; Peters, D.G.; et al. Chemo-preventive and therapeutic effect of the dietary flavonoid kaempferol: A comprehensive review. Phytother. Res. 2019, 3, 263–275. [Google Scholar] [CrossRef] [PubMed]
  96. Lin, C.W.; Chen, P.N.; Chen, M.K.; Yang, W.E.; Tang, C.H.; Yang, S.F.; Hsieh, Y.S. Kaempferol reduces matrix metalloproteinase-2 expression by down-regulating ERK1/2 and the activator protein-1 signaling pathways in oral cancer cells. PLoS ONE 2013, 8, e80883. [Google Scholar] [CrossRef] [PubMed]
  97. Lee, G.A.; Choi, K.C.; Hwang, K.A. Kaempferol, a phytoestrogen, suppressed triclosan-induced epithelial-mesenchymal transition and metastatic-related behaviors of MCF-7 breast cancer cells. Environ. Toxicol. Pharmacol. 2017, 49, 48–57. [Google Scholar] [CrossRef] [PubMed]
  98. Chang, H.L.; Chang, Y.M.; Lai, S.C.; Chen, K.M.; Wang, K.C.; Chiu, T.T.; Chang, F.H.; Hsu, L.S. Naringenin inhibits migration of lung cancer cells via the inhibition of matrix metalloproteinases-2 and-9. Exp Ther. Med. 2017, 13, 739–744. [Google Scholar] [CrossRef] [PubMed]
  99. Pandurangan, A.K.; Dharmalingam, P.; Sadagopan, S.K.; Ganapasam, S. Luteolin inhibits matrix metalloproteinase 9 and 2 in azoxymethane-induced colon carcinogenesis. Hum. Exp. Toxicol. 2014, 33, 1176–1185. [Google Scholar] [CrossRef]
  100. Luqman, S.; Pezzuto, J.M. NFκB: A promising target for natural products in cancer chemoprevention. Phytother. Res. 2010, 24, 949–963. [Google Scholar] [CrossRef]
  101. Liu, H.; Zeng, Z.; Wang, S.; Li, T.; Mastriani, E.; Li, Q.H.; Bao, H.X.; Zhou, Y.J.; Wang, X.; Liu, Y.; et al. Main components of pomegranate, ellagic acid and luteolin, inhibit metastasis of ovarian cancer by down-regulating MMP-2 and MMP-9. Cancer Biol. Ther. 2017, 18, 990–999. [Google Scholar] [CrossRef] [PubMed]
  102. Yao, X.; Jiang, W.; Yu, D.; Yan, Z. Luteolin inhibits proliferation and induces apoptosis of human melanoma cells in vivo and in vitro by suppressing MMP-2 and MMP-9 through the PI3K/AKT pathway. Food Funct. 2019, 10, 703–712. [Google Scholar] [CrossRef] [PubMed]
  103. Gupta, G.; Siddiqui, M.A.; Khan, M.M.; Ajmal, M.; Ahsan, R.; Rahaman, M.A.; Ahmad, M.A.; Arshad, M.; Khushtar, M. Current pharmacological trends on myricetin. Drug Res. 2020, 70, 448–454. [Google Scholar] [CrossRef] [PubMed]
  104. Ko, C.H.; Shen, S.C.; Lee, T.J.; Chen, Y.C. Myricetin inhibits matrix metalloproteinase 2 protein expression and enzyme activity in colorectal carcinoma cells. Mol. Cancer Ther. 2005, 4, 281–290. [Google Scholar] [CrossRef] [PubMed]
  105. Ci, Y.; Zhang, Y.; Liu, Y.; Lu, S.; Cao, J.; Li, H.; Zhang, J.; Huang, Z.; Zhu, X.; Gao, J.; et al. Myricetin suppresses breast cancer metastasis through down-regulating the activity of matrix metalloproteinase (MMP)-2/9. Phytother. Res. 2018, 32, 1373–1381. [Google Scholar] [CrossRef] [PubMed]
  106. Kang, H.R.; Moon, J.Y.; Ediriweera, M.K.; Song, Y.W.; Cho, M.; Kasiviswanathan, D.; Cho, S.K. Dietary flavonoid myricetin inhibits invasion and migration of radioresistant lung cancer cells (A549-IR) by suppressing MMP-2 and MMP-9 expressions through inhibition of the FAK-ERK signaling pathway. Food Sci. Nutr. 2020, 8, 2059–2067. [Google Scholar] [CrossRef] [PubMed]
  107. Sun, Z.Q.; Meng, F.H.; Tu, L.X.; Sun, L. Myricetin attenuates the severity of seizures and neuroapoptosis in pentylenetetrazole kindled mice by regulating the of BDNF-TrkB signaling pathway and modulating matrix metalloproteinase-9 and GABAA. Exp. Ther. Med. 2019, 17, 3083–3091. [Google Scholar] [CrossRef] [PubMed]
  108. Chandrashekar, N.; Selvamani, A.; Subramanian, R.; Pandi, A.; Thiruvengadam, D. Baicalein inhibits pulmonary carcinogenesis-associated inflammation and interferes with COX-2, MMP-2 and MMP-9 expressions in-vivo. Toxicol. Appl. Pharmacol. 2012, 261, 10–21. [Google Scholar] [CrossRef] [PubMed]
  109. Yang, X.; Zhang, C.; Li, Y.; Jiang, J. Baicalein restrains proliferation, migration, and invasion of human malignant melanoma cells by down-regulating colon cancer associated transcript-1. Braz. J. Med. Biol. 2021, 54, e8934. [Google Scholar] [CrossRef]
  110. Tian, Y.; Zhen, L.; Bai, J.A.; Mei, Y.; Li, Z.; Lin, A.; Li, X. Anticancer effects of baicalein in pancreatic neuroendocrine tumors in vitro and in vivo. Pancreas 2017, 46, 1076. [Google Scholar] [CrossRef]
  111. Lin, H.; Hao, Y.; Wan, X.; He, J.; Tong, Y. Baicalein inhibits cell development, metastasis and EMT and induces apoptosis by regulating ERK signaling pathway in osteosarcoma. J. Recept. Signal Transduct. 2020, 40, 49–57. [Google Scholar] [CrossRef]
  112. Li, W.; Saji, S.; Sato, F.; Noda, M.; Toi, M. Potential clinical applications of matrix metalloproteinase inhibitors and their future prospects. Int. J. Biol. Marker. 2013, 28, 117–130. [Google Scholar] [CrossRef] [PubMed]
  113. Kim, S.; Kim, S.H.; Hur, S.M.; Lee, S.K.; Kim, W.W.; Kim, J.S.; Kim, J.H.; Choe, J.H.; Nam, S.J.; Lee, J.E.; et al. Silibinin prevents TPA-induced MMP-9 expression by down-regulation of COX-2 in human breast cancer cells. J. Ethnopharmacol. 2009, 126, 252–257. [Google Scholar] [CrossRef]
  114. Singh, R.P.; Agarwal, R. Flavonoid antioxidant silymarin and skin cancer. Antioxid Redox Signal. 2002, 4, 655–663. [Google Scholar] [CrossRef]
  115. Nguyen, T.T.; Ding, D.; Wolter, W.R.; Pérez, R.L.; Champion, M.M.; Mahasenan, K.V.; Hesek, D.; Lee, M.; Schroeder, V.A.; Jones, J.I.; et al. Validation of matrix metalloproteinase-9 (MMP-9) as a novel target for treatment of diabetic foot ulcers in humans and discovery of a potent and selective small-molecule MMP-9 inhibitor that accelerates healing. J. Med. Chem. 2018, 61, 8825–8837. [Google Scholar] [CrossRef]
  116. Gillis, C.N. Panax ginseng pharmacology: A nitric oxide link? Biochem. Pharmacol. 1997, 54, 1–8. [Google Scholar] [CrossRef]
  117. Attele, A.S.; Wu, J.A.; Yuan, C.S. Ginseng pharmacology: Multiple constituents and multiple actions. Biochem. Pharmacol. 1999, 58, 1685–1693. [Google Scholar] [CrossRef] [PubMed]
  118. Lu, J.M.; Yao, Q.; Chen, C. Ginseng compounds: An update on their molecular mechanisms and medical applications. Curr. Vasc. Pharmacol. 2009, 7, 293–302. [Google Scholar] [CrossRef] [PubMed]
  119. Yue, P.Y.; Wong, D.Y.; Wu, P.K.; Leung, P.Y.; Mak, N.K.; Yeung, H.W.; Liu, L.; Cai, Z.; Jiang, Z.H.; Fan, T.P.D.; et al. The angiosuppressive effects of 20 (R)-ginsenoside Rg3. Biochem. Pharmacol. 2006, 72, 37–445. [Google Scholar] [CrossRef]
  120. Xu, T.M.; Cui, M.H.; Ying, X.I.N.; Gu, L.P.; Jiang, X.; Su, M.M.; Wang, D.D.; Wang, W.J. Inhibitory effect of ginsenoside Rg3 on ovarian cancer metastasis. Chin. Med. J. 2008, 121, 1394–1397. [Google Scholar] [CrossRef]
  121. Yoon, J.H.; Choi, Y.J.; Cha, S.W.; Lee, S.G. Anti-metastatic effects of ginsenoside Rd via inactivation of MAPK signaling and induction of focal adhesion formation. Phytomedicine 2012, 19, 284–292. [Google Scholar] [CrossRef]
  122. Ho, Y.L.; Li, K.C.; Chao, W.; Chang, Y.S.; Huang, G.J. Korean red ginseng suppresses metastasis of human hepatoma SK-Hep1 cells by inhibiting matrix metalloproteinase-2/-9 and urokinase plasminogen activator. Evid. Based Complement. Alternat. 2012, 2012, 1–8. [Google Scholar] [CrossRef]
  123. Pei, J.S.; Liu, C.C.; Hsu, Y.N.; Lin, L.L.; Wang, S.C.; Chung, J.G.; Bau, D.T.; Lin, S.S. Amentoflavone induces cell-cycle arrest and apoptosis in MCF-7 human breast cancer cells via mitochondria-dependent pathway. In Vivo 2012, 26, 963–970. [Google Scholar]
  124. Shin, D.H.; Bae, Y.C.; Kim-Han, J.S.; Lee, J.H.; Choi, I.Y.; Son, K.H.; Kang, S.S.; Kim, W.K.; Han, B.H. Polyphenol amentoflavone affords neuroprotection against neonatal hypoxic-ischemic brain damage via multiple mechanisms. J. Neurochem. 2006, 96, 561–572. [Google Scholar] [CrossRef]
  125. Guruvayoorappan, C.; Kuttan, G. Effect of amentoflavone on the inhibition of pulmonary metastasis induced by B16F-10 melanoma cells in C57BL/6 mice. Integr. Cancer Ther. 2007, 6, 185–197. [Google Scholar] [CrossRef] [PubMed]
  126. Guruvayoorappan, C.; Kuttan, G. Amentoflavone inhibits experimental tumor metastasis through a regulatory mechanism involving MMP-2, MMP-9, prolyl hydroxylase, lysyl oxidase, VEGF, ERK-1, ERK-2, STAT-1, NM23 and cytokines in lung tissues of C57BL/6 mice. Immunopharmacol. Immunotoxicol. 2008, 30, 711–727. [Google Scholar] [CrossRef] [PubMed]
  127. Fimognari, C.; Hrelia, P. Sulforaphane as a promising molecule for fighting cancer. Mutat Res. 2007, 635, 90–104. [Google Scholar] [CrossRef]
  128. Gamet-Payrastre, L.; Li, P.; Lumeau, S.; Cassar, G.; Dupont, M.A.; Chevolleau, S.; Gasc, N.; Tulliez, J.; Tercé, F. Sulforaphane, a naturally occurring isothiocyanate, induces cell cycle arrest and apoptosis in HT29 human colon cancer cells. Cancer Res. 2000, 60, 1426–1433. [Google Scholar] [PubMed]
  129. Kim, H. Sulforaphane Sensitizes Tumor Necrosis Factor–Related Apoptosis-Inducing Ligand (TRAIL)–Resistant Hepatoma Cells to TRAIL-Induced Apoptosis through Reactive Oxygen Species–Mediated Up-regulation of DR5. Cancer Res. 2016, 66, 1740–1750. [Google Scholar] [CrossRef]
  130. Xia, Y.; Kang, T.W.; Jung, Y.D.; Zhang, C.; Lian, S. Sulforaphane inhibits nonmuscle invasive bladder cancer cells proliferation through suppression of HIF-1α-mediated glycolysis in hypoxia. J. Agric. Food Chem. 2019, 67, 7844–7854. [Google Scholar] [CrossRef]
  131. Shahidi, F.; Naczk, M. Phenolics in Food and Nutraceuticals; CRC Press: Boca Raton, FL, USA, 2003; Chapters 4–6; pp. 83–330. [Google Scholar]
  132. Nardini, M.; Leonardi, F.; Scaccini, C.; Virgili, F. Modulation of ceramide-induced NF-κB binding activity and apoptotic response by caffeic acid in U937 cells: Comparison with other antioxidants. Free Radic. Biol. Med. 2001, 30, 722–733. [Google Scholar] [CrossRef] [PubMed]
  133. Chung, T.W.; Moon, S.K.; Chang, Y.C.; Ko, J.H.; Lee, Y.C.; Cho, G.; Kim, S.H.; Kim, J.G.; Kim, C.H. Novel and therapeutic effect of caffeic acid and caffeic acid phenyl ester on hepatocarcinoma cells: Complete regression of hepatoma growth and metastasis by dual mechanism. FASEB J. 2004, 18, 1670–1681. [Google Scholar] [CrossRef] [PubMed]
  134. Kanimozhi, G.; Prasad, N.R. Anticancer effect of caffeic acid on human cervical cancer cells. In Coffee in Health and Disease Prevention; Academic Press: Cambridge, MA, USA, 2015; pp. 655–661. [Google Scholar]
  135. Pettit, G.R.; Singh, S.B.; Hamel, E.; Lin, C.M.; Alberts, D.S.; Garcia-Kendal, D. Isolation and structure of the strong cell growth and tubulin inhibitor combretastatin A-4. Experientia 1989, 45, 209–211. [Google Scholar] [CrossRef] [PubMed]
  136. Kanthou, C.; Tozer, G.M. Tumour targeting by microtubule-depolymerising vascular disrupting agents. Expert Opin. Ther. Targets. 2007, 11, 1443–1457. [Google Scholar] [CrossRef]
  137. Tozer, G.M.; Kanthou, C.; Baguley, B.C. Disrupting tumour blood vessels. Nat. Rev. Cancer. 2005, 5, 423–435. [Google Scholar] [CrossRef] [PubMed]
  138. Kanthou, C.; Tozer, G.M. Microtubule depolymerizing vascular disrupting agents: Novel therapeutic agents for oncology and other pathologies. Int. J. Exp. Pathol. 2009, 90, 284–294. [Google Scholar] [CrossRef] [PubMed]
  139. Mahal, K.; Biersack, B.; Caysa, H.; Schobert, R.; Mueller, T. Combretastatin A-4 derived imidazoles show cytotoxic, antivascular, and antimetastatic effects based on cytoskeletal reorganisation. Investig. New Drugs 2015, 33, 541–554. [Google Scholar] [CrossRef] [PubMed]
  140. Huang, X.; Chen, Y.; Zhong, W.; Liu, Z.; Zhang, H.; Zhang, B.; Wang, H. Novel combretastatin A-4 derivative containing aminophosphonates as dual inhibitors of tubulin and matrix metalloproteinases for lung cancer treatment. Eur. J. Med. Chem. 2022, 244, 114817. [Google Scholar] [CrossRef] [PubMed]
  141. Ravindranath, M.H.; Ramasamy, V.; Moon, S.; Ruiz, C.; Muthugounder, S. Differential growth suppression of human melanoma cells by tea (Camellia sinensis) epicatechins (ECG, EGC and EGCG). Evid.-Based Complement. Altern. Med. 2009, 6, 523–530. [Google Scholar] [CrossRef]
  142. Shankar, S.; Ganapathy, S.; Hingorani, S.R.; Srivastava, R.K. EGCG inhibits growth, invasion, angiogenesis and metastasis of pancreatic cancer. Front. Biosci. 2008, 13, 440–452. [Google Scholar] [CrossRef]
  143. Rimando, A.M.; Cuendet, M.; Desmarchelier, C.; Mehta, R.G.; Pezzuto, J.M.; Duke, S.O. Cancer chemopreventive and antioxidant activities of pterostilbene, a naturally occurring analogue of resveratrol. J. Agric. Food Chem. 2002, 50, 3453–3457. [Google Scholar] [CrossRef] [PubMed]
  144. Stivala, L.A.; Savio, M.; Carafoli, F.; Perucca, P.; Bianchi, L.; Maga, G.; Forti, L.; Pagnoni, U.M.; Albini, A.; Prosperi, E.; et al. Specific structural determinants are responsible for the antioxidant activity and the cell cycle effects of resveratrol. Biol. Chem. 2001, 276, 22586–22594. [Google Scholar] [CrossRef] [PubMed]
  145. Pan, M.H.; Chang, Y.H.; Badmaev, V.; Nagabhushanam, K.; Ho, C.T. Pterostilbene induces apoptosis and cell cycle arrest in human gastric carcinoma cells. J Agric Food Chem. 2007, 55, 7777–7785. [Google Scholar] [CrossRef] [PubMed]
  146. McCormack, D.; McFadden, D. Pterostilbene and cancer: Current review. J. Surg. Res. 2012, 173, e53–e61. [Google Scholar] [CrossRef] [PubMed]
  147. Lin, H.C.; Hsieh, M.J.; Peng, C.H.; Yang, S.F.; Huang, C.N. Pterostilbene Inhibits Vascular Smooth Muscle Cells Migration and Matrix Metalloproteinase-2 through Modulation of MAPK Pathway. J. Food Sci. 2015, 80, H2331–H2335. [Google Scholar] [CrossRef] [PubMed]
  148. Kim, J.M.; Noh, E.M.; Kim, M.S.; Hwang, J.K.; Hwang, H.Y.; Ryu, D.G.; Kim, H.J.; Yu, H.N.; You, Y.O.; Kim, J.S.; et al. Decursin prevents TPA-induced invasion through suppression of PKCα/p38/NF-κB-dependent MMP-9 expression in MCF-7 human breast carcinoma cells. Int. J. Oncol. 2014, 44, 1607–1613. [Google Scholar] [CrossRef] [PubMed]
  149. Yim, D.; Singh, R.P.; Agarwal, C.; Lee, S.; Chi, H.; Agarwal, R. A novel anticancer agent, decursin, induces G1 arrest and apoptosis in human prostate carcinoma cells. Cancer Res. 2005, 65, 1035–1044. [Google Scholar] [CrossRef] [PubMed]
  150. Jiang, C.; Guo, J.; Wang, Z.; Xiao, B.; Lee, H.J.; Lee, E.O.; Kim, S.H.; Lu, J. Decursin and decursinol angelate inhibit estrogen-stimulated and estrogen-independent growth and survival of breast cancer cells. Breast Cancer Res. 2007, 9, R77. [Google Scholar] [CrossRef] [PubMed]
  151. Kim, W.J.; Lee, S.J.; Choi, Y.D.; Moon, S.K. Decursin inhibits growth of human bladder and colon cancer cells via apoptosis, G1-phase cell cycle arrest and extracellular signal-regulated kinase activation. Int. J. Mol. Med. 2010, 25, 635–641. [Google Scholar]
  152. Kim, J.H.; Jeong, J.H.; Jeon, S.T.; Kim, H.; Ock, J.; Suk, K.; Kim, S.I.; Song, K.S.; Lee, W.H. Decursin inhibits induction of inflammatory mediators by blocking nuclear factor-κB activation in macrophages. Mol. Pharmacol. 2006, 69, 1783–1790. [Google Scholar] [CrossRef]
  153. Lee, S.H.; Lee, J.H.; Kim, E.J.; Kim, W.J.; Suk, K.; Kim, J.H.; Song, G.Y.; Lee, W.H. A novel derivative of decursin, CSL-32, blocks migration and production of inflammatory mediators and modulates PI3K and NF-κB activities in HT1080 cells. Cell Biol. Int. 2012, 36, 683–688. [Google Scholar] [CrossRef] [PubMed]
  154. Rudrapal, M.; Khairnar, S.J.; Khan, J.; Dukhyil, A.B.; Ansari, M.A.; Palai, S.; Devi, R. Dietary polyphenols and their role in oxidative stress-induced human diseases: Insights into protective effects, antioxidant potentials and mechanism (s) of action. Front. Pharmacol. 2022, 13, 806470. [Google Scholar] [CrossRef] [PubMed]
  155. D’Aguanno, S.; Del Bufalo, D. Inhibition of anti-apoptotic Bcl-2 proteins in preclinical and clinical studies: Current overview in cancer. Cells 2020, 9, 1287. [Google Scholar] [CrossRef] [PubMed]
  156. Wang, L.; Ling, Y.; Chen, Y.; Li, C.L.; Feng, F.; You, Q.D.; Lu, N.; Guo, Q.L. Flavonoid baicalein suppresses adhesion, migration and invasion of MDA-MB-231 human breast cancer cells. Cancer Lett. 2010, 297, 42–48. [Google Scholar] [CrossRef] [PubMed]
  157. Maikoo, S.; Makayane, D.; Booysen, I.N.; Ngubane, P.; Khathi, A. Ruthenium compounds as potential therapeutic agents for type 2 diabetes mellitus. Eur. J. Med. Chem. 2021, 213, 113064. [Google Scholar] [CrossRef]
  158. Gong, G.; Du, W.; Xu, J.; Huang, X.; Yin, G. Regulation of heteronuclear Pt–Ru complexes on the fibril formation and cytotoxicity of human islet amyloid polypeptide. J. Inorg. Biochem. 2018, 189, 7–16. [Google Scholar] [CrossRef]
  159. Lee, T.Y.; Chei, W.S.; Ju, H.; Lee, M.S.; Lee, J.W.; Suh, J. A Co (III) complex cleaving soluble oligomers of h-IAPP in the presence of polymeric aggregates of h-IAPP. Bioorg. Med. Chem. Lett. 2012, 22, 5689–5693. [Google Scholar] [CrossRef]
  160. Gong, G.; Wang, W.; Du, W. Binuclear ruthenium complexes inhibit the fibril formation of human islet amyloid polypeptide. RSC Adv. 2017, 7, 18512–18522. [Google Scholar] [CrossRef]
  161. Yang, Y.; Wang, Q.; Luo, J.; Jiang, Y.; Zhou, R.; Tong, S.; Wang, Z.; Tong, Q. Superoxide dismutase mimic, MnTE-2-PyP enhances rectal anastomotic strength in rats after preoperative chemoradiotherapy. Oxid. Med. Cell. Longev. 2020, 2020, 3509859. [Google Scholar] [CrossRef]
  162. Pacor, S.; Zorzet, S.; Cocchietto, M.; Bacac, M.; Vadori, M.; Turrin, C.; Gava, B.; Castellarin, A.; Sava, G. Intratumoral NAMI-A treatment triggers metastasis reduction, which correlates to CD44 regulation and tumor infiltrating lymphocyte recruitment. J. Pharmacol. Exp. Ther. 2004, 310, 737–744. [Google Scholar] [CrossRef]
  163. Gu, L.; Li, X.; Ran, Q.; Kang, C.; Lee, C.; Shen, J. Antimetastatic activity of novel ruthenium (III) pyridine complexes. Cancer Med. 2016, 5, 2850–2860. [Google Scholar] [CrossRef] [PubMed]
  164. Wu, J.; Yang, T.; Wang, X.; Li, W.; Pang, M.; Sun, H.; Liang, H.; Yang, F. Development of a multi-target anticancer Sn(ii) pyridine-2-carboxaldehyde thiosemicarbazone complex. Dalton Trans. 2021, 50, 10909–10921. [Google Scholar] [CrossRef] [PubMed]
  165. Ruiz, M.C.; Perelmulter, K.; Levín, P.; Romo, A.I.; Lemus, L.; Bollati-Fogolín, M.; León, I.E.; Di Virgilio, A.L. Antiproliferative activity of two copper (II) complexes on colorectal cancer cell models: Impact on ROS production, apoptosis induction and NF-κB inhibition. Eur. J. Pharm. Sci. 2022, 169, 106092. [Google Scholar] [CrossRef]
  166. Levín, P.; Ruiz, M.C.; Romo, A.I.; Nascimento, O.R.; Di Virgilio, A.L.; Oliver, A.G.; Ayala, A.P.; Diógenes, I.C.; León, I.E.; Lemus, L. Water-mediated reduction of [Cu(dmp)2(CH3CN)]2+: Implications of the structure of a classical complex on its activity as an anticancer drug. Inorg. Chem. Front. 2021, 8, 3238–3252. [Google Scholar] [CrossRef]
  167. Balsa, L.M.; Ruiz, M.C.; de la Parra, L.S.M.; Baran, E.J.; León, I.E. Anticancer and antimetastatic activity of copper (II)-tropolone complex against human breast cancer cells, breast multicellular spheroids and mammospheres. J. Inorg. Biochem. 2020, 204, 110975. [Google Scholar] [CrossRef] [PubMed]
  168. Zec, M.; Srdic-Rajic, T.; Konic-Ristic, A.; Todorovic, T.; Andjelkovic, K.; Filipovic-Ljeskovic, I.; Radulovic, S. Anti-metastatic and anti-angiogenic properties of potential new anti-cancer drugs based on metal complexes of selenosemicarbazones. Curr. Med. Chem. Anticancer Agents 2012, 12, 1071–1080. [Google Scholar] [CrossRef] [PubMed]
  169. Lin, H.; Wang, Y.; Lai, H.; Li, X.; Chen, T. Iron (II)–Polypyridyl Complexes Inhibit the Growth of Glioblastoma Tumor and Enhance TRAIL-Induced Cell Apoptosis. Chem. Asian J. 2018, 13, 2730–2738. [Google Scholar] [CrossRef]
  170. Stefàno, E.; Muscella, A.; Benedetti, M.; De Castro, F.; Fanizzi, F.P.; Marsigliante, S. Antitumor and antimigration effects of a new Pt compound on neuroblastoma cells. Biochem. Pharmacol. 2022, 202, 115124. [Google Scholar] [CrossRef] [PubMed]
  171. Muscella, A.; Vetrugno, C.; Calabriso, N.; Cossa, L.G.; De Pascali, S.A.; Fanizzi, F.P.; Marsigliante, S. [Pt (O,O’-acac)(γ-acac)(DMS)] alters SH-SY5Y cell migration and invasion by the inhibition of Na+/H+ exchanger isoform 1 occurring through a PKC-ε/ERK/mTOR Pathway. PLoS ONE 2014, 9, e112186. [Google Scholar] [CrossRef]
  172. Elie, B.T.; Fernández-Gallardo, J.; Curado, N.; Cornejo, M.A.; Ramos, J.W.; Contel, M. Bimetallic titanocene-gold phosphane complexes inhibit invasion, metastasis, and angiogenesis-associated signaling molecules in renal cancer. Eur. J. Med. Chem. 2019, 161, 310–322. [Google Scholar] [CrossRef]
  173. Li, Z.; Wang, Q.; Li, L.; Chen, Y.; Cui, J.; Liu, M.; Zhang, N.; Liu, Z.; Han, J.; Wang, Z. Ketoprofen and loxoprofen platinum (iv) complexes displaying antimetastatic activities by inducing DNA damage, inflammation suppression, and enhanced immune response. J. Med. Chem. 2021, 64, 17920–17935. [Google Scholar] [CrossRef] [PubMed]
  174. Chen, Y.; Wang, Q.; Li, Z.; Liu, Z.; Zhao, Y.; Zhang, J.; Liu, M.; Wang, Z.; Li, D.; Han, J. Naproxen platinum (iv) hybrids inhibiting cycloxygenases and matrix metalloproteinases and causing DNA damage: Synthesis and biological evaluation as antitumor agents in vitro and in vivo. J. Chem. Soc. 2020, 49, 5192–5204. [Google Scholar] [CrossRef] [PubMed]
  175. Becceneri, A.B.; Fuzer, A.M.; Plutin, A.M.; Batista, A.A.; Lelievre, S.A.; Cominetti, M.R. Three-dimensional cell culture models for metallodrug testing: Induction of apoptosis and phenotypic reversion of breast cancer cells by the trans-[Ru(PPh3)2(N,N-dimethyl-N-thiophenylthioureato-k2O,S)(bipy)]PF6 complex. Inorg. Chem. Front. 2020, 7, 2909–2919. [Google Scholar] [CrossRef]
  176. Wen, T.; Song, L.; Hua, S. Perspectives and controversies regarding the use of natural products for the treatment of lung cancer. Cancer Med. 2021, 10, 2396–2422. [Google Scholar] [CrossRef]
  177. Jing, Q.; Hu, X.; Ma, Y.; Mu, J.; Liu, W.; Xu, F.; Li, Z.; Bai, J.; Hua, H.; Li, D. Marine-derived natural lead compound disulfide-linked dimer psammaplin A: Biological activity and structural modification. Mar. Drugs 2019, 17, 384. [Google Scholar] [CrossRef]
Figure 1. A schematic representation of the number of different cancer deaths globally.
Figure 1. A schematic representation of the number of different cancer deaths globally.
Biomedicines 12 00794 g001
Figure 2. A brief description of the structure of MMPs and arrangement of their functional domains.
Figure 2. A brief description of the structure of MMPs and arrangement of their functional domains.
Biomedicines 12 00794 g002
Figure 3. Catalytic site of MMP-2 (left) and MMP-9 (right). The N, O, C, and H atoms are presented in blue, brown, gray, and white, respectively. Zn(II) is indicated in orange circle.
Figure 3. Catalytic site of MMP-2 (left) and MMP-9 (right). The N, O, C, and H atoms are presented in blue, brown, gray, and white, respectively. Zn(II) is indicated in orange circle.
Biomedicines 12 00794 g003
Figure 4. Role of MMP-2 and MMP-9 in cancer progression.
Figure 4. Role of MMP-2 and MMP-9 in cancer progression.
Biomedicines 12 00794 g004
Table 1. Marine-derived natural product downregulators for MMP-2 and MMP-9.
Table 1. Marine-derived natural product downregulators for MMP-2 and MMP-9.
ProductNameSourcesMMP-2MMP-9ModelsRefs.
Biomedicines 12 00794 i001LemnalolLemnalia sp.UnknownSuppressionRats[79]
Biomedicines 12 00794 i002Ageladine AAgelas nakamuraiSuppressionSuppressionIn vitro[80]
Biomedicines 12 00794 i00311-Episinularoide acetateSinularia flexibilisSuppressionSuppressionHA22T cells[81,82]
Biomedicines 12 00794 i004Dihydroaustrasulfone alcoholCladiella australisSuppressionSuppressionA549 cells VSMC[83,84]
Biomedicines 12 00794 i005MarimastatAlgaeUnknownSuppressionMice[85,86,87]
Biomedicines 12 00794 i006Aeroplysinin-1Aplysina aerophobaSuppressionUnknownEndothelial cells[88,89]
Table 2. Flavonoids and other compounds from fruits and vegetables, which can modulate MMP-2 and MMP-9 activity and/or levels.
Table 2. Flavonoids and other compounds from fruits and vegetables, which can modulate MMP-2 and MMP-9 activity and/or levels.
ProductNameSourcesMMP-2MMP-9ModelsRefs.
Biomedicines 12 00794 i007QuercetinVegetables, seeds, and grainsSuppressionSuppressionHSC-6 cells
SCC-9 cells
2K1C rats
[91,92,93,94]
Biomedicines 12 00794 i008KaempferolTea, cabbage, beans, tomato, strawberries, and grapesSuppression
Suppression
Unknown SuppressionSCC-4 cells
MCF-7 cells
[95,96,97]
Biomedicines 12 00794 i009NaringeninGrapefruit, bergamot, orange, tomatoes, and cocoaSuppressionSuppressionA549 cells[98,99]
Biomedicines 12 00794 i010LuteoinSalvia, broccoli, parsley, thyme, green pepper, and artichokeSuppression
Suppression
Suppression
Suppression
Suppression
Suppression
Mice
A2780 cells,
mice
[100,101,102]
Biomedicines 12 00794 i011MyricetinTomatoes, nuts, oranges, berries, tea, and red wineSuppression
Suppression
Suppression
Suppression
Unknown
Suppression
Suppression
Suppression
COLO 205 cells
MDA-Mb-231 cells
A549 cells
[103,104,105,106]
Biomedicines 12 00794 i012BaicaleinRoot of
Scutellaria
Baicalensis
Suppression
Suppression
Suppression
Suppression
Unknown Unknown
Suppression
Suppression
SK-MEL-28
BON1 cells
CRL-1427 cells
A375 cells, mice
[107,108,109,110,111]
Biomedicines 12 00794 i013GenisteinFood,
soya products
UnknownSuppressionMice[112]
Biomedicines 12 00794 i014SilibininFruitsUnknownSuppressionMice[113,114,115]
Biomedicines 12 00794 i015GinsenosidePanax rootSuppressionSuppression3T3-L1 cells[116,117,118,119,120,121,122]
Biomedicines 12 00794 i016AmenoflavoneSelaginella tamariscinaSuppressionSuppressionBreast cancer cells
B16F-10 cells
[123,124,125,126]
Biomedicines 12 00794 i017SulforaphaneCabbage and broccoliUnknownSuppressionAbdominal cancer cells[127,128,129,130]
Biomedicines 12 00794 i018Caffeic acidFruits, vegetables, wine, olive oil, and coffeeUnknownSuppressionColon cancer cells[131,132,133,134]
Biomedicines 12 00794 i019Combretastatin A-4Willow treeSuppressionSuppressionA549 cells[135,136,137,138,139,140]
Biomedicines 12 00794 i020PterostilbeneBlueberries and grapeSuppressionUnknownA7r5 cells[141,142,143,144,145,146,147]
Biomedicines 12 00794 i021DecursinAngelica gigasUnknownSuppressionBreast cancer cells[148,149,150,151,152,153]
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

Islam, M.T.; Jang, N.H.; Lee, H.J. Natural Products as Regulators against Matrix Metalloproteinases for the Treatment of Cancer. Biomedicines 2024, 12, 794. https://doi.org/10.3390/biomedicines12040794

AMA Style

Islam MT, Jang NH, Lee HJ. Natural Products as Regulators against Matrix Metalloproteinases for the Treatment of Cancer. Biomedicines. 2024; 12(4):794. https://doi.org/10.3390/biomedicines12040794

Chicago/Turabian Style

Islam, Md. Towhedul, Nak Han Jang, and Hyuck Jin Lee. 2024. "Natural Products as Regulators against Matrix Metalloproteinases for the Treatment of Cancer" Biomedicines 12, no. 4: 794. https://doi.org/10.3390/biomedicines12040794

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