Molecules 29 05275 v2
Molecules 29 05275 v2
1 Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, Tsarigradsko Shosse,
73, 1113 Sofia, Bulgaria; zasheva.diana@yahoo.com (D.Z.); silvina_z@abv.bg (S.Z.)
2 Agrobioinstitute, Agricultural Academy, bul. “Dragan Tsankov” 8, 1164 Sofia, Bulgaria;
mladenovpetko@yahoo.com (P.M.); d_djilianov@abi.bg (D.D.); dmoyankova@abi.bg (D.M.)
3 Institute of Plant Physiology and Genetics, Bulgarian Academy of Science, “Acad. Georgi Bonchev” Str.,
Bl. 21, 1113 Sofia, Bulgaria; zlatina.go@abv.bg (Z.G.); stamova@bio21.bas.bg (M.G.);
valentin.velinov82@gmail.com (V.V.)
4 Institute of Molecular Biology “Rumen Tzanev”, Bulgarian Academy of Sciences, “Acad. Georgi Bonchev”
Str., Bl. 21, 1113 Sofia, Bulgaria; ialexandar@yahoo.com
* Correspondence: lpsimova@yahoo.co.uk
Abstract: Breast cancer is the second leading cause of death among women, and the number of
mortal cases in diagnosed patients is constantly increasing. The search for new plant compounds
with antitumor effects is very important because of the side effects of conventional therapy and the
development of drug resistance in cancer cells. The use of plant substances in medicine has been
Citation: Zasheva, D.; Mladenov, P.; well known for centuries, but the exact mechanism of their action is far from being elucidated. The
Zapryanova, S.; Gospodinova, Z.; molecular mechanisms of cytotoxicity exerted by secondary metabolites and bioactive peptides of
Georgieva, M.; Alexandar, I.; Velinov, plant origin on breast cancer cell lines are the subject of this review.
V.; Djilianov, D.; Moyankova, D.;
Simova-Stoilova, L. Cytotoxic Effects Keywords: breast cancer; MCF7; MDA-MB231; cytotoxicity; secondary metabolites; bioactive
of Plant Secondary Metabolites and peptides
Naturally Occurring Bioactive
Peptides on Breast Cancer Model
Systems: Molecular Mechanisms.
Molecules 2024, 29, 5275. https://
1. Introduction
doi.org/10.3390/molecules29225275
The problem of effective cancer therapy is becoming increasingly important. The
Academic Editors: Agata number of patients diagnosed with a tumor and the number of mortal cases among those
Poniewierska-Baran and Maciej
patients have been expanding over the past few years. In 2018 alone, the number of dead
Tarnowski
cancer patients was about 10 million, as reported by the World Health Organization [1].
Received: 30 September 2024 Concerning breast cancer, 1.38 million women were diagnosed in 2012 [2], and by 2020
Revised: 30 October 2024 this number had increased to 2.3 million [3]. Breast cancer is the most common cancer
Accepted: 6 November 2024 among women worldwide by 2021, although about 1% of breast cancer patients are men [4].
Published: 7 November 2024 Breast cancer cases account for 12% of all cancer cases and can be found in teenage girls,
in women of reproductive age, and in menopausal women with preliminarily developed
climax changes [5].
Commonly used methods of breast cancer treatment are surgery, chemotherapy, and
Copyright: © 2024 by the authors.
radiation therapy. Surgical methods are invasive with a long recovery period and usually
Licensee MDPI, Basel, Switzerland.
affect a woman’s quality of life. The surgical method is followed by chemotherapy and
This article is an open access article
radiotherapy or a combination of them. Chemotherapeutic anticancer drugs are usually
distributed under the terms and
conditions of the Creative Commons
synthetic drugs and have cytotoxic effects. They are classified into several groups depend-
Attribution (CC BY) license (https://
ing on the mechanism of action on cells. The following groups are used in anticancer
creativecommons.org/licenses/by/ therapy: microtubule-interacting agents (vincristine, taxol) [6], drugs with a topoisomerase-
4.0/). inhibiting effect (doxorubicin, podophyllotoxin) [7], alkylating agents such as melphalan,
methotrexate, blocking cell metabolism [8], and DNA interacting agents such as doc-
etaxel [9]. The molecular mechanisms of commonly used antitumor drugs are not very
specific; they kill not only cancer cells but their targets could also be normal cells. This fact
explains many side effects of chemotherapeutics. The same is the case with radiotherapy.
These conventional therapeutic methods are associated with loss of normal blood cells, loss
of body mass, and loss of hair; some of them are associated with vomiting and dizziness [2].
Long-term use of drugs in anticancer therapy can lead to the emergence of resistance to
them in cancer cells (multidrug resistance, MDR) [10]. The MDR of cancer cells results from
molecular mechanisms related to the inability of administered drugs to penetrate into the
cells and trigger a mechanism of cancer cell elimination. These facts make drugs ineffective,
and cancer development continues progressively. The probability of the emergence of
multidrug resistance in cancer patients, together with the strong impact of the side effects
of chemotherapy drugs on the treated organism, makes the search for new anticancer
therapeutics so necessary and significant. Possible substances that could be expected to
have little or no side effects are certain substances of plant origin. Plants have been used in
medicine since ancient times. Generally, Chinese traditional medicine is based on them;
Ayurveda in India is based on therapeutic methods using plants and substances of plant
origin. Intensive studies of plant secondary metabolites and bioactive peptides are being
carried out on cancer cell lines, a model system that enables the testing of plant derivatives
on cell lines with characteristics of different types of breast cancer. The mechanisms of their
cytotoxicity and their targets can be established at the cellular and molecular levels. The
subject of this review will be studies of well-characterized substances among secondary
metabolites and polypeptides of plant origin with cytotoxic effects on breast cancer cell
lines with different characteristics and the molecular mechanisms mediating the reduction
in cancer cell number as a result of plant substances application. Breast cancer cell lines are
well-characterized and convenient model systems with the same cell signaling mechanisms
as the primary breast tumors. The possible new search for metabolites with specific targets,
depending on cell-line characteristics and cancer characteristics, will be discussed. The
discovery of plant polypeptides targeting specific proteases with a high impact on cancer
cell proliferation, invasion metastasis formation, and angiogenesis will be elucidated.
Future prospects of searching for new plant metabolites and polypeptides with breast
anticancer effects will be suggested.
2. Breast Cancer Types and Signal Pathways Related to the Breast Cancer Development
Depending on cancer origin, the types of breast cancer are ductal (85% of breast
cancer cases; lobular, originating from the lobes (9–14%), and very aggressive atypical
inflammatory forms (1–6% of the cases) [11]. The characteristics of the transformed cells
are also very important. Depending on the expression of cell receptors—estrogen receptor
(ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2),
breast cancer types are divided into four molecular subtypes: basal (triple negative, which
does not express ER, PR, and HER2 (11 % of BC cases with an aggressive phenotype and
lack of response to hormone replacement therapy), luminal A (ER+/PR+/HER2−, 60%
of BC cases), luminal B (ER+/PR+/HER2+, 15% of BC cases), and HER2 overexpressing
(ER-/PR−/HER2+, 14% of BC cases). The cellular transformation of breast cancer is
characterized by changes in signaling pathways as a result of some risk factors such as
genetic and environmental that could be predisposed to it, followed by changes in molecular
mechanisms accompanied by mutations in different groups of genes: tumor-suppressor
genes (p53, NF1, NF2), DNA repair system genes (PTEN, BRCA1 and BRCA2 genes),
oncogenes (RAS, MYC, Bcl2, RAF), and genes involved in cell growth and metabolism [12].
These mutations lead to the acquisition of uncontrollable division of cancer cells, and
invasion of the tissues where they are located and, therefore, can form blood vessels
and metastasize.
The main signaling pathways associated with breast cancer transformation and pro-
gression (Figure 1) depend on the expression of the estrogen receptor and its associated
PI3K/AKT/mTOR signaling pathway is another one with an impact on brea
development; more PI3K mutations are features of breast cancer predisposition. T
to cellular dedifferentiation of basal and luminal progenitor cells, rendering
Molecules 2024, 29, 5275 multilineage potential [50]. Akt kinase and downstream mTOR hyperactivation 3 of 23
resistance to endocrine therapy [51]. Inhibition of mTOR is a good candidate for
therapy due to the possibility of restoring the antiestrogen sensitivity of cancer
signaling—ER
The knowledge alpha
of (ERα—a membrane
the molecular receptor) andofER
mechanisms beta (ERβ—a
breast cancer nuclear recep-
transformation pr
tor) [13–16]. All of them are linked with the processes of cell development directed by
opportunity to search for new targeted therapeutic drugs that are more effective
changes in cell cycle control and proliferation, cell differentiation and cell death, and cell
invasive.
migration andThemotility
interest
[17].inThe
plant‐derived
pathway associated compounds with anticancer
with cell proliferation is mediated activity
revived in recent years and intense research on the molecular mechanisms
by ERα interaction with cyclin D. Cyclin D is an activator of cyclin-dependent kinases 4 of the
and 6, which mediate cell cycle progression from G1 to S phase
especially targeting cancer signaling pathways, is being performed. Canc[17,18]. This type of cancer
responds to hormone replacement therapy, and estrogen-dependent cell line models such
especially
as MCF7 arebreast
used incancer,
studies toare diseases
search related
for targeted to different
therapeutic blockingchanges
estrogen in the genome,
receptor α
stress,
in high and
levelsaand
change
estrogeninreceptor
immune β infunctions; the search
low levels, whose for molecules
homodimers which
are located in the mod
key elements and
cell cytoplasm, associated with breast
their dimerization cancer
is needed transformation
for activation and nuclear istranslocation.
a challenge in th
They direct the transcription of different genes in the nucleus, or they are cofactors of
related to breast cancer therapy.
various other transcription factors [18–21].
3.1. Flavonoids
Flavonoids belong to the group of substances containing polyphenols, where they
have been described as being about 4000 and they contain 15-carbon derivatives of beta-
gamma-pyrone. They are divided into different classes, such as flavanones, flavans, proan-
thocyanidins [7], coumarins, and coumarin-related compounds [53]. A comparative study
of flavonoids from different subclasses and their effects on breast cancer cell lines estab-
lished the leading role of the 2,3-double bond in the C-ring in cytotoxicity linked with
mitochondrial impairment [54]. Docking analysis of 98 flavonoids with GLUT1 trans-
porter, overexpressed in several carcinomas, has shown that the transporter is a target
for flavonoids. Screening for cytotoxicity on a carcinoma cell line has shown that eight
of them (apigenin, kaempferol, eupatilin, luteolin, hispidulin, isosinensetin, sinensetin,
and nobiletin) reduce cell viability to 50% by inhibition of GLUT1 transporter, the critical
pharmacophores of flavonoids inhibitors being 3′ hydrophobic groups and hydrogen bond
acceptors [55]. The molecular mechanism of glucose metabolism inhibition in tumor cells
by forming the flavonoid–GLUT1 transporter complex is shown in Figure 2. The group of
flavonoids like genistein, which is isolated from soya and soya products, induces cell cycle
arrest in the G2/M phase in the triple-negative MDA-MB231 cell line, and the mechanism
of the arrest is mediated by ERK1/2 kinase pathway activation and downregulation of
Cdk1, cyclin B1, and Cdc25 C [56]. Genistein, in combination with doxorubicin, exhibited a
synergistic effect on MCF-7/Adr drug-resistant cells by an increase in the intracellular accu-
mulation of doxorubicin and inhibition of HER2/neu expression [57]. It was demonstrated
that the bioflavonoid quercetin, which is characterized by low in vivo toxicity, increases the
inhibitory effect of doxorubicin in MCF-7 Adr-resistant breast cancer cells [58]. Combined
again with doxorubicin, quercetin induced rejection of 4T1 breast cancer in mice [59]. The
natural flavonoid naringenin associated with doxorubicin synergistically suppressed the
growth and migration of MCF7 cells [60].
Molecules 2024, 29, 5275 6 of 23
Molecules 2024, 29, 5275 6 of 23
Figure2.2.Molecular
Figure Molecularinteraction
interactionmechanisms
mechanisms ofof suppression
suppression of of breast
breast cancer
cancer survival
survival andand mitigation
mitigation
by natural phenylpropanoid glycosides, flavonoids, terpenoids, and coumarins. Structures of
by natural phenylpropanoid glycosides, flavonoids, terpenoids, and coumarins. Structures of com-
compounds are represented by ball and stick diagrams with their charged groups involved in
pounds are represented by ball and stick diagrams with their charged groups involved in interactions.
interactions. The structure of myconoside was downloaded from Japan Chemical Substance
The structure(Nikkaji);
Dictionary of myconoside was downloaded
the common from Japan Chemical
feature pharmacophores Substance
of flavonoids for Dictionary
interaction(Nikkaji);
with Glut 1
the common feature
represented pharmacophores
with green of flavonoids
and blue spheres for interaction
is according with Glut 1ofrepresented
to [55]; structures with
celastrol and green
conferone
and
wereblue spheres is according
downloaded to [55];Proteins
from PubChem. structuresof of celastrol and
interactions are conferone
represented were
by downloaded from
ribbons downloaded
PubChem. Proteins ofin
from PDB illustrated interactions
the contextare represented
of the processesby andribbons
pathwaysdownloaded from PDB
in breast cancer cells.illustrated
Red dashed
inlines represent
the context of the
the interactions
processes andof pathways
inhibition inof breast
metabolites
cancerwith
cells.target proteins.
Red dashed lines represent the
interactions of inhibition of metabolites with target proteins.
Coumarins
Coumarins
Among flavonoids, coumarins have a very important role in breast cancer research
Among flavonoids, coumarins have a very important role in breast cancer research
on model cell lines. Coumarin is 1,2‐benzopyrone or 2H‐1‐benzopyran‐2‐one. It can be
on model cell lines. Coumarin is 1,2-benzopyrone or 2H-1-benzopyran-2-one. It can be
found in various plant organs (roots, seeds, and leaves). Coumarins are widely available
found in various plant organs (roots, seeds, and leaves). Coumarins are widely avail-
in plants—800 coumarin derivatives are known from about 100 plant families and 600
able in plants—800 coumarin derivatives are known from about 100 plant families and
genera [53,61]. They have a polyphenolic structure and are colorless. The coumarins are
600 genera [53,61]. They have a polyphenolic structure and are colorless. The coumarins
divided
are dividedinto four
into groups—simple
four groups—simple coumarins,
coumarins, composed
composed of of
hydroxylated,
hydroxylated, alkoxilated, and
alkoxilated,
and alkylated coumarin derivatives and their glycosides; pyranocoumarins, which areare
alkylated coumarin derivatives and their glycosides; pyranocoumarins, which
structuresofofsix-furan
structures six‐furanrings
ringsand
andfused
fusedwithwiththethe benzene
benzene ring
ring furanocoumarins,
furanocoumarins, which
which
comprisefurane
comprise furanering
ringand
andcoumarin
coumarinfusionfusion [62];
[62]; and
and pyrone‐substituted
pyrone-substituted coumarins.
coumarins. This
This
group is divided into three subgroups: 4‐hydroxycoumarin, 3‐phenylcoumarin,
group is divided into three subgroups: 4-hydroxycoumarin, 3-phenylcoumarin, and 3,4- and 3,4‐
benzocoumarin. Coumarins have biological effects on various diseases,
benzocoumarin. Coumarins have biological effects on various diseases, including cancer. including cancer.
Themechanisms
The mechanisms of their
of their anticancer
anticancer effectseffects are related
are related to the induction
to the induction of through
of apoptosis apoptosis
through
the the activation
activation of a caspaseofcascade
a caspase cascade
initiated by initiated
caspase 9,by caspase
which 9, whichbyisamediated
is mediated by a
decrease in
decrease in antiapoptotic
antiapoptotic Bcl2 expression Bcl2 expression
levels [63]. Theirlevels [63]. Their
anticancer effectsanticancer
have beeneffects have on
established been
established
the MCF7 breast on cancer
the MCF7 breast
cell model line.cancer cell with
Treatment model line. Treatment
coumarin with coumarin
derivatives increased P21
derivatives increased P21 protein expression and arrested cells in the G0/G1 phase [64],
Molecules 2024, 29, 5275 7 of 23
protein expression and arrested cells in the G0/G1 phase [64], Three synthesized coumarins
derived from triphenylethylene inhibit angiogenesis of breast cancer cell lines and more
precisely, compound TCH-5c changes endothelial cell cytoskeleton organization and migra-
tion of EA.hy926 endothelial cells [64]. Cellular treatment with them affects different cell
signaling and kinase-dependent pathways that direct cancer cells to apoptosis or cell cycle
arrest [63,64]. The coumarin group targets the key cell cycle regulator cdc25 and is a good
option for targeted therapy of breast cancer [65]. Oral uptake of coumarins is effective. They
are absorbed rapidly in the gastrointestinal tract and penetrate cells by passive diffusion
through the lipid membrane. Coumarins are rapidly metabolized in the liver/excreted by
the kidney, and only 2–6% reach the system circulation [66,67]. The toxicological studies of
coumarins are very controversial. Synthetic coumarins have been shown to cause acute
chronic and cancerogenic effects in mice and rats [66]. In contrast, studies on human and
cynomolgus monkey liver fragments or hepatocytes have shown relative resistance to
coumarin toxicity [66]. The use of standard breast anticancer drugs results in a multidrug
resistance phenotype. Some of the plant coumarins suppress Pgp-mediated drug efflux in
the MCF7 cell line, and thus, they are promising for overcoming multidrug resistance [68].
The mechanism of multidrug resistance suppression of conferone is presented in Figure 2.
3.3. Polyphenols
3.3.1. Curcumin
Curcumin (diferuloylmethane) has a polyphenolic structure and is a yellow powder
derived from a plant extract of Curcuma longa. This substance is the subject of many studies
related to its anticancer effects. Its effects mediate the inhibition of the transcription factor
NFkB in the MCF7 breast cancer cell line [69] after reducing the expression of its target
gene, such as COX2 and cyclin D, leading to apoptosis [6,7]. It can affect cells by binding to
the tubulin-like taxol and thereby block mitosis [69]. It has an effect on normal cells and
is therefore used in combination with other drugs. Paclitaxel and curcumin reduced cell
viability of breast cancer cell lines MCF7 and MDA-MB231 through apoptosis, activation of
caspases 3/7 and protein expression of nuclear NfkB transcription factor [70]. Curcumin
has effects on cell culture models of various origins as an effective inhibitor of tyrosine-
regulated kinase 2, a positive regulator of the 26S proteasome, which disrupts it and leads
to impaired cell proliferation in the triple-negative breast cancer cell line MDA-MB468 [71].
The effect of curcumin on cell viability was favored by binding to jacalin molecules, which
act as a natural ligand of Thomsen–Friedenreich (TF) tumor-associated antigen in this
triple-negative cell line MDA-MB231 [72]. It blocks kinase pathways by NfKb signaling
in combination with taxol in higher doses and can bind to DNA molecules, thus blocking
cell cycle progression; besides, it can block angiogenesis, targeting the VEGF-VEGFR2
signaling pathway [69,70]. Other studies related to the combined treatment of curcumin
with standardly used drugs on breast cancer cell lines show an improvement in the an-
Molecules 2024, 29, 5275 8 of 23
3.3.2. Saponins
Indicacin is a polyphenol from the group of 3-terpenoid saponins purified from a
methanol extract of Fagonia indika. It has an effect on breast cancer cell lines of different
origins (MDA-MB-468 and MCF7) through PARP cleavage, caspase 3 activation, DNA
fragmentation, and apoptosis activation [84]. Morus alba metabolite lectin has an antiprolif-
erative effect on the MCF7 cell line [85]. Platycodin D from Placticodon grandifloras has a
cytotoxic effect, activates caspases, and induces apoptosis in the MCF7 cell line [86]. The
in vivo activity of platycodin D was established on mice with tumors induced by an injec-
tion of human metastatic breast cancer cells. Oral administration of platycodin D inhibited
cell-induced osteolysis and blocked osteoclast formation and osteoclast-mediated bone
resorption [87]. In combination with docetaxel, platycodin enhanced the antiproliferative
effect in MCF7 and MDA-MB231 cell lines [88]. The side effects of platycodin D, as of
the other saponins, are related to the induction of hemolytic activity [89], which could be
solved by chemical modifications of their structure.
3.3.3. Myconoside
The anticancer effects of the phenyl propanoid glycoside myconoside, identified in the
methanol extract fraction of the resurrection plant Haberlea rhodopensis, were established on
MCF7 and MDA-MB231 breast cancer cell lines. The cytotoxic and antiproliferative effects
of the myconoside-enriched fraction have been demonstrated, and the docking analysis
of myconoside with estrogen receptor, glucose transporter, and MYST acetyltransferase
provides a basis for the explanation of the molecular mechanisms of their anticancer
effects [90] and prospects for the future search of targeted therapy based on myconoside
treatment. The molecular mechanism of the myconoside effect on the MCF7 cell line is
presented in Figure 2. Docking analysis predicts that it blocks estrogen receptors, glucose
transporter GLUT1, and MYST acetyltransferase and, in this way, reduces cell growth and
proliferation. MDA-MB231 triple negative cell line is influenced by linking to the glucose
transporter and MYST acetyltransferase [90].
Molecules 2024, 29, 5275 9 of 23
3.4. Plant Metabolites with Anticancer Effects on Triple Negative Cell Lines
Different groups of plant metabolites have anticancer effects on triple-negative cell
lines that do not express estrogen receptors, progesterone receptors, and the epidermal
growth factor receptor (HER1). These characteristics make them unresponsive to hormone
replacement therapy and with a higher invasive and metastatic potential [91].
3.4.1. Piperine
Piperine from black pepper arrests MDA-MB231 and MDA-MB468 cells by blocking
the activation of the phosphatidyl inositol 3 kinase pathway and triggering the caspase-
dependent mitochondrial apoptosis pathway. Piperine-treated cells showed lower MMP2/9
expression and migration potential [92]. The effects of alkaloids such as piperlongumine
(Piper longum), berberine, a quaternary ammonium alkaloid extracted from Coptis chinensis,
indirubin-3-monoxime found in Indigo naturalis, are associated with the blocking of kinase
signaling pathways and reducing the migratory potential of triple-negative breast cancer
cells [93–95]. Piperine, as other described alkaloid agents in this paragraph, does not have
side effects, and the application of piperine in combination with other conventional drugs
like paclitaxel is very effective. It is absorbed by the intestinal tract and metabolized in the
liver and, kidney, and excreted in bile and urine [96].
3.4.2. Terpenoids
The terpenoid group is another plant metabolite group with established effects on
triple-negative breast cancer cell lines. Tanshinone I and Tanshinone IIA found in the Dan
Shen root of Salvia miltiorrhiza affect the MDA-MB231 cell line by reducing cell growth and
vascular endothelial growth factor (VEGF) expression, thereby reducing the proliferation
level via mTOR/p70S6K/4 E-BP1 signaling pathway [97]. Eupalinolide J (EJ), a sesquiter-
pene lactone found in Eupatorium lindleyanum, astragaloside IV, an active triterpenoid from
Radix astragali found in the roots of Astragalus membranaceus Bunge, Betulinic acid, and Dil-
lenia suffruticosa Martelli root extract KHF16 (24-acetylisodahurinol-3-O-D-xylopyranoside),
a triterpenoid found in the rhizomes of Cimicifuga foetida, the pseudopterosins, a class of
marine diterpene glycosides, extracted from the gorgonian sea whip Antillogorgia elisabethae,
have anticancer effects on triple-negative breast cancer cell lines. The effects of terpenoids
on triple-negative cell lines can be classified based on their molecular mechanisms leading
to cell death. KHF16 (24-acetylisodachurinol-3-O-D-xylopyranoside) triggers cell cycle
arrest and apoptosis in some triple-negative cell lines, promoting G2/M phase cell cycle
arrest and NF-Kb pathway-mediated necrosis [98]. The terpenoids inhibit cell proliferation
by multiple targets. The molecular mechanism of their anticancer activity is related to the
NfkB pathway. Triterpene celastrole inhibits tumor growth of mouse xenographs of the
triple-negative cell lin MDA-MB-435 e to 60% by NfKb inactivation and activates apoptotic
effects induced by tumor necrosis factor α. NfkB inactivation inhibits its DNA binding
capacity (Figure 2). The celastrole molecule suppresses NfkB, targeting its cysteine 179 [99].
The effects of terpenoids on triple-negative cell lines are limited in clinical trial translation
because of their poor absorption and low availability, which implies the need for their
structural modification in synthetic analogs. Their use in combination with standard drugs
like doxorubicin, cis platina, and paclitaxel diminishes multidrug resistance of tumor cells
and increases the effectiveness of chemotherapy [100].
Well-characterized polyphenols with their anticancer mechanisms clarified on cancer
cell lines of different origins are described in Table 1.
Molecules 2024, 29, 5275 10 of 23
Molecules 2024, 29, 5275 10 of 23
Table 1. Plant secondary metabolites with anticancer effects on breast cancer cell lines with established molecular mechanism.
TableTable
1. Plant secondary
1. Plant metabolites
secondary with anticancer
metabolites effectseffects
with anticancer on breast cancercancer
on breast cell lines
cell with
lines established molecular
with established mechanism.
molecular mechanism.
Chemical Structure (PubChem Database‐ Established Established
Chemical Structure (PubChem Database‐ Established Established
Metabolites/ Established
Anticancer Established
Molecular
Metabolites/ Chemical Structure (PubChem Database- Anticancer Molecular References
Metabolites/ https://pubchem.ncbi.nlm.nih.gov/ accessed on 20 September Anticancer Molecular
Plant Origin https://pubchem.ncbi.nlm.nih.gov/ accessed
https://pubchem.ncbi.nlm.nih.gov/ on 20on
Accessed September Activity on Breast Cancer Cell Mechanism of Anticancer References
References
Plant Origin
Plant Origin 2024) 2024) Activity
Activity onon Breast
Breast Cancer Cell
Cancer Mechanism
Mechanism of Anticancer
Anticancer
20 September
2024) Models Activity
Cell Models
Models Activity
Activity
Essential
Essentialchemical
chemical structure
structure
Essential chemical structure
Apoptosis induction,
Vinca alkaloids Group of substances used in Apoptosis induction,
Vinca alkaloids
Vinca alkaloids Group of substances used in Apoptosis binding to DNAbinding
induction, molecules
to DNA
Taxol (Paclitaxel) Groupstandard chemotherapy
of substances that have
used in standard binding to DNA molecules
Taxol (Paclitaxel)
Taxol (Paclitaxel) standard chemotherapy that have and cell
molecules proliferation
and arrest,
cell proliferation arrest, [6–8]
Derived from chemotherapy
effects onthat have effects
different breastoncancer and cell proliferation arrest, [6–8][6–8]
Derived from from
Derived effects on different breast development
cancer of multidrug
development of multidrugresistance
Catharanthus roseusroseus different breast cancer cell lines
cell lines development of multidrug
Catharanthus
Catharanthus roseus cell lines in cells
resistance in cells
resistance in cells
Molecules 2024, 29, 5275 11 of 23
Table 1. Cont.
Established Established
Molecules 2024, 29, 5275 Chemical Structure (PubChem Database- 11 of 23
Metabolites/
Molecules 2024, 29, 5275 Anticancer Molecular 11 of 23
https://pubchem.ncbi.nlm.nih.gov/ Accessed on References
Plant Origin Activity on Breast Cancer Mechanism of Anticancer
20 September 2024)
Cell Models Activity
Blocking
Blocking activation
activation of
of
Phosphatidyl
Phosphatidyl inositol 33
Blocking activation inositol
of Phosphatidyl
Piperine
Piperine
Piperine MDA‐MB‐231
MDA-MB-231 and and MDA‐MB‐468
MDA-MB-468
MDA‐MB‐231 and MDA‐MB‐468 kinase
inositol pathway
3 kinase
kinase and
pathway
pathway andand [92]
[92] [92]
from Piper Piper longum
fromlongum extract triple-negative cell lines
triple‐negative cell lines triggering the caspase-dependent
triggering the caspase‐
from Piper longum extract
extract triple‐negative cell lines triggering the caspase‐
mitochondrial apoptosis
dependent
dependent mitochondrial
mitochondrial
apoptosis
apoptosis
Blockage
Blockage of
of the
the kinase
kinase signal
signal
Piperlongumine
Piperlongumine Triple‐negative breast cancer
Triple‐negative breast cancer cellcell Blockage pathways,
pathways, signal
of the kinase
Piperlongumine [93]
from Triple-negative breast lines
cancer cell lines pathways, [93] [93]
from Piper
fromlongum
Piper longum extract
Piper longum
extract
extract lines decrease
decrease
decrease
in
in migration
migration
in migration potential
potential
potential
Apoptosis
Apoptosis activation
activation by by
inhibition
inhibition of
of NfkB
NfkB
transcription
transcription factor
factor followed
followed
by
by reduction
reduction of
of its
its target
target
Curcumin
Curcumin Effects
Effects on
on MCF‐7
MCF‐7 cell
cell line
line and
and genes
genes expression
expression
isolated
isolated from
from extract
extract of
of triple‐negative
triple‐negative cell
cell lines
lines like
like (COX2,
(COX2, cyclin
cyclin D),
D), [6,7,68–72]
[6,7,68–72]
Curcuma
Curcuma longa
longa MDA‐MB‐231, MDA‐MB‐468
MDA‐MB‐231, MDA‐MB‐468 Inhibition of tyrosine‐
Inhibition of tyrosine‐
regulated
regulated kinase
kinase 2, 2,
perturbation of
perturbation of 26S 26S
proteasome
proteasome andand cell
cell cycle
cycle
arrest
arrest
Molecules 2024, 29, 5275 12 of 23
Blockage of the kinase signal
Piperlongumine Triple‐negative breast cancer cell pathways,
Table 1. Cont.
[93]
from Piper longum extract lines decrease in migration
potential
Established Established
Chemical Structure (PubChem Database-
Metabolites/ Anticancer Molecular
https://pubchem.ncbi.nlm.nih.gov/ Accessed on References
Plant Origin Activity on Breast Cancer Mechanism of Anticancer
20 September 2024)
Cell Models Activity
Apoptosis activation by
inhibition of NfkB
transcription
Apoptosis factor
activation by followed
inhibition of
NfkBby reduction factor
transcription of its followed
target by
Curcumin Effects on MCF‐7 cell line and reduction
genes expressiongenes
of its target
Curcumin Effects on MCF-7 cell line and
expression
isolatedisolated
from extract of of
from extract triple‐negative
triple-negative celllike
cell lines lines like (COX2, cyclin D), [6,7,68–72]
[6,7,68–72]
(COX2, cyclin D),
Curcuma
Curcuma longalonga MDA-MB-231,
MDA‐MB‐231,MDA-MB-468
MDA‐MB‐468Inhibition Inhibition of tyrosine‐
of tyrosine-regulated kinase
regulated
2, perturbation kinase
of 26S 2,
proteasome and
cell cycle arrest
perturbation of 26S
Molecules 2024, 29, 5275 12 of 23
proteasome and cell cycle
arrest
Myconoside *
Myconoside * MCF‐7
MCF-7 Cytotoxic and
Cytotoxic and antiproliferative [90] [90]
from Haberlea rhodopensis
from Haberlea rhodopensis MDA‐MB‐231
MDA-MB-231 antiproliferative effectseffects
Cytotoxic
Platycodin D ** effect
MCF‐7 [86]
from Placticodon grandifloras activation of caspases and
apoptosis
Myconoside **
Myconoside MCF‐7 Cytotoxic and
and
MCF‐7 Cytotoxic [90]
from Haberlea rhodopensis
Myconoside * Table 1. Cont. MDA‐MB‐231
MCF‐7 antiproliferative
Cytotoxic effects
and [90]
from Haberlea rhodopensis MDA‐MB‐231 antiproliferative effects [90]
from Haberlea rhodopensis MDA‐MB‐231 antiproliferative effects
Established Established
Chemical Structure (PubChem Database-
Metabolites/ Anticancer Molecular
https://pubchem.ncbi.nlm.nih.gov/ Accessed on References
Plant Origin Activity on Breast Cancer Mechanism of Anticancer
20 September 2024)
Cell Models Activity
Cytotoxic
Cytotoxic
Platycodin D
Platycodin D **
** effect
Cytotoxic
Platycodin MCF‐7 effect
Cytotoxic [86]
Platycodin
from Placticodon ** D **
Placticodon grandifloras
grandifloras
D MCF‐7
MCF-7 activation of caspases and
effect
effect
activation of caspases and [86] [86]
from from Placticodon grandifloras MCF‐7 [86]
from Placticodon grandifloras activation of apoptosis
activationcaspases and apoptosis
of caspases and
apoptosis
apoptosis
Reduction of
Reduction of cell
cell growth
growth
Tanshinone Reduction of cellof
growth and VEGF
II Tanshinone
Tanshinone
Tanshinone
Tanshinone IIA IIA
I Tanshinone
IIA and VEGF
and VEGF
Reduction
expression,
expression,
cell growth
expression,
decrease in proliferation
Tanshinone
the Dan
the Danthe
Shen
Shen
I Tanshinone
Danroot
root Salvia
Shenofroot ofIIA
of Salvia Salvia MDA‐MB‐231
MDA-MB-231
MDA‐MB‐231 decrease in proliferation
and VEGF proliferation
expression, via [97] [97]
via decrease in
mTOR/p70S6K/4 via
E-BP1 signaling [97]
miltiorrhiza
Shen root of Salvia
the Danmiltiorrhiza MDA‐MB‐231 mTOR/p70S6K/4
decrease in E‐BP1
proliferation via [97]
miltiorrhiza pathway E‐BP1
mTOR/p70S6K/4
miltiorrhiza signaling pathway
signaling pathway
mTOR/p70S6K/4 E‐BP1
signaling pathway
Blockage of
of the kinase
kinase signal
signal
Berberine
Berberine Triple‐negative breast
Triple‐negative breast cancer cell Blockage
cancer cell Blockage of the
the kinase signal
Berberine
Triple-negative breast lines
cancer cellcancer
pathways,
Blockage
lines cell pathways, of
pathways, thedecrease
kinase
decrease
decrease
in
signal
in
in migration
[94,95]
[94,95]
[94,95]
from Coptis
Berberine
from Coptisfrom chinensis
Coptis chinensis
chinensis Triple‐negative breast
lines
Molecules 2024, 29, 5275 migration
pathways, potential
decrease
potentialin
potential
migration [94,95]
13 of 23
from Coptis chinensis lines
migration potential
Eupalinolide
Eupalinolide Triple‐negative breast cancer cell Triggering
Triggering
of cellofcycle
cell arrest
cycle and
Triple-negative breast cancer cell lines [98] [98]
from Eupatorium
from Eupatorium lindleyanum
lindleyanum lines apoptosis
arrest and apoptosis
Table 1. Cont.
Eupalinolide
Eupalinolide Triple‐negative
Triple‐negative breast cancer cell Triggering of
of cell
cell cycle
Establishedbreast cancer cell Triggering
Established cycle [98]
[98]
from Eupatorium
from lindleyanum Chemical Structure (PubChem Database- lines
lines arrest and
and apoptosis
arrestMolecular
apoptosis
Metabolites/ Anticancer
https://pubchem.ncbi.nlm.nih.gov/ Accessed on References
Plant Origin Activity on Breast Cancer Mechanism of Anticancer
20 September 2024)
Cell Models Activity
Cell
Cell cycle
cycle arrest
arrest inin G2/M
G2/M
phase
phase mediated
mediated by
by ERK1/2
ERK1/2
Genistein
Genistein
Genistein
Cell cycle arrest in G2/M phase
kinase
kinase
mediated bypathway
pathway activation
activation
ERK1/2 kinase pathway [56]
isolated from
from soya
isolatedisolated fromand
soya soyasoya
and soya
and soya MDA‐MB‐231
MDA‐MB‐231
MDA-MB-231 [56] [56]
and
activationdownregulation
and downregulation
and downregulation of
of of
products
products
products Cdk1, cyclin
Cdk1,
Cdk1, cyclinB1
cyclin B1and
B1 and
andCdc25
Cdc25
Cdc25C
C
C
Note: The
The chemical
Note: Note: chemical structures
structures of plant metabolites
metabolitesareare from PubChem: https://pubchem.ncbi.nlm.nih.gov/ accessed onon 2020 September 2024.
2024. ** The myconoside
The chemical structuresofofplant
plant metabolites are from
from PubChem:
PubChem: https://pubchem.ncbi.nlm.nih.gov/
https://pubchem.ncbi.nlm.nih.gov/ accessedaccessed
on 20 September September
2024. * The myconosideThe myconoside
structure from Haberlea
structure
structure from
from Haberlea
rhodopensis Haberlea
as rhodopensis
rhodopensis
determined by MS/MS as determined
determinedinby
as identification by MS/MS
MS/MS
reference identification
identification
[90]. in
in reference
** The Platycodin D structure[90].
reference [90]. **
** The Platycodin
Thefollowing
is described Platycodin D
D structure
structure
the link: is
is described
described following
following the
the link:
link:
https://www.selleckchem.com/products/platycodin-
https://www.selleckchem.com/products/platycodin‐d.html
d.html accessed on 20 September 2024. *** KHF 16 structure accessed
https://www.selleckchem.com/products/platycodin‐d.html is described
accessed on 20
20 September
onfollowing the [98].2024.
September 2024. *** KHF 16
*** KHF 16 structure
structure is
is described
described following
following thethe [98].
[98].
Molecules 2024, 29, 5275 15 of 23
4.1. Lunasin
Lunasin is a soybean-derived bioactive peptide of 5.5 kDA (43 amino acids, SKWQHQQ
DSCRKQLQGVNLTPC-EKHIMEKIQG-RGD-DDDDDDDD, four structural fragments).
It was discovered in 1987, and its antimitotic and cell death effects were established
12 years later [107]. The RGD motif (Arg-Gly-Asp) competes with integrins, suppressing
the integrin-mediated signaling pathway [108]. This motif mediates the binding of the
peptide to receptors and its internalization into tumor cells; besides, binding through
the RGD motif could directly activate caspase-3 and promote apoptosis. The C-terminal
aspartic acid residues can interact with chromatin, and the second fragment of nine amino
acids is responsible for binding to the histone core [107]. Lunasin structure presents highly
disordered and flexible features typical for intrinsically disordered proteins, which partic-
ipate in transcription and translation regulation, DNA condensation, cell cycle, mitosis,
and apoptosis [107]. Lunasin has no inhibiting effects on the normal breast cancer cell
line MCF-10A, whereas 50% inhibition of proliferation in the triple-negative MDA-MB-231
breast cancer cell line is achieved at a twice lower concentration than that of the hormone-
responsive MCF-7 cell line. In both cancer cell lines, lunasin treatment decreased aromatase
gene expression and activity, vascular endothelial growth factor (VEGF) secretion and
cell vitality, and induced cell apoptosis [107]. Estrogen receptor (ER)α gene expression
Molecules 2024, 29, 5275 16 of 23
was decreased by lunasin treatment, and ERβ gene levels were significantly increased in
MDA-MB-231 cells [109]. Transcriptomic and proteomic analysis of breast cancer cell line
MDA-MB-231 treated with synthetic lunasin and lunasin from overexpressing transgenic
maize demonstrated apoptosis activation by significant upregulation of cysteinyl aspartate
specific proteinase (CASP) 3, CASP 7, and CASP 14, almost 10-fold increase in Bax/Bcl-2 ra-
tio, and down-regulation of DNA replication genes [110,111]. In the MCF7 cell line, lunasin
up-regulated tumor suppressor phosphatase and tensin homolog deleted in chromosome
ten (PTEN) promoter activity, increased PTEN transcript and protein levels and enhanced
nuclear PTEN localization, and the induced apoptosis was p53-independent [112]. In
both MCF7 and MDA-MB-231 cell lines, 10–20 µM lunasin inhibited the expression of
matrix metalloproteinase (MMP)-2/-9, the phosphorylation of focal adhesion kinase (FAK),
Src, Akt, ERK and nucleus translocation of NF-κB, thus demonstrating the possibility of
metastasis suppression in breast cancer cells through integrin-mediated FAK/Akt/ERK
and NF-κB signaling pathways and downregulation of MMP-2/-9 [108]. The anticancer
action of lunasin is preserved by a protease inhibitor Bowman–Birk type [106].
with conventional drugs with herbs [129]. In this respect, it is important to know the
molecular mechanisms of action in the active constituents and to predict the interactions
with a chemotherapeutic, which could be of three kinds—synergism, an additive effect,
or antagonism (if two compounds bind to the same target). Knowledge of the molecular
mechanism of cytotoxic action, target sites, doses, and pharmacokinetics will be at hand to
go beyond the trial-and-error approach and predict possible interactions. In this respect,
the use of model cell lines with defined molecular characteristics close to those of in vivo
tumor cells is of enormous importance to check the effectiveness of certain combinations;
however, the experiments on cell lines cannot be extrapolated directly to cancer situation
in vivo [129].
Knowledge of the exact molecular mechanisms of the observed cytotoxic effects
permits combined treatments with substances targeting different pathways of cytotoxic
action. Despite recent progress, a largely unexplored field is the use of drugs, which target
the activated proteolytic machinery in tumor cells [113] and the combined treatment with
natural substances and/or drugs with different molecular targets, which could additionally
destabilize malignant cells and could be a way to reverse the development of multidrug
resistance [101,114].
Author Contributions: Conceptualization and writing—original draft preparation, D.Z., P.M., Z.G.
and L.S.-S.; writing—review and editing, S.Z., M.G., I.A., V.V., D.D. and D.M.; visualization, D.Z.,
P.M. and M.G. All authors have read and agreed to the published version of the manuscript.
Funding: The research was funded by NSF of Bulgaria, Grant number KΠ-06-H41/6.
Informed Consent Statement: Not applicable.
Data Availability Statement: Data sharing is not applicable.
Conflicts of Interest: The authors declare no conflicts of interest.
References
1. Zarei, O.; Benvenuti, S.; Ustun-Alkan, F.; Hamzeh-Mivehroud, M.; Dastmalchi, S. Strategies of targeting the extracellular domain
of RON tyrosine kinase receptor for cancer therapy and drug delivery. J. Cancer Res. Clin. Oncol. 2016, 142, 2429–2446. [CrossRef]
2. Plackal Adimuriyil George, B.; Abrahamse, H. A review on novel breast cancer therapies: Photodynamic therapy and plant
derived agent induced cell death mechanisms. Anti-Cancer Agents Med. Chem. (Former. Curr. Med. Chem.-Anti-Cancer Agents) 2016,
16, 793–801. [CrossRef] [PubMed]
3. Available online: https://www.iarc.who.int/news-events/current-and-future-burden-of-breast-cancer-global-statistics-for-
2020-and-2040/ (accessed on 25 February 2024).
4. Avtanski, D.; Poretsky, L. Phyto-polyphenols as potential inhibitors of breast cancer metastasis. Mol. Med. 2018, 24, 29. [CrossRef]
[PubMed]
5. Available online: https://www.breastcancer.org/facts-statistics (accessed on 25 February 2024).
6. Stierle, A.; Strobel, G.; Stierle, D. Taxol and taxane production by Taxomyces andreanae, an endophytic fungus of Pacific yew.
Science 1993, 260, 214–216. [CrossRef] [PubMed]
7. Lindholm, P. Cytotoxic Components of Plant Origin -Biological and Chemical Diversity. Ph.D. Thesis, Phaculty of Pharmacy,
Upsala University, Uppsala, Sweden, 2005.
8. Demain, A.L.; Vaishnav, P. Natural products for cancer chemotherapy. Microb. Biotechnol. 2011, 4, 687–699. [CrossRef] [PubMed]
9. Mansoori, B.; Mohammadi, A.; Davudian, S.; Shirjang, S.; Baradaran, B. The different mechanisms of cancer drug resistance: A
brief review. Adv. Pharm. Bull. 2017, 7, 339–348. [CrossRef]
10. Mazumder, K.; Biswas, B.; Raja, I.M.; Fukase, K. A review of cytotoxic plants of the Indian subcontinent and a broad-spectrum
analysis of their bioactive compounds. Molecules 2020, 25, 1904. [CrossRef]
11. Kumar, V.; Green, S.; Stack, G.; Berry, M.; Jin, J.R.; Chambon, P. Functional domains of the human estrogen receptor. Cell 1987, 51,
941–951. [CrossRef]
12. Osborne, C.K.; Schiff, R.; Fuqua, S.A.; Shou, J. Estrogen receptor: Current understanding of its activation and modulation. Clin.
Cancer Res. 2001, 7, 4338s–4342s.
13. Renoir, J.M.; Marsaud, V.; Lazennec, G. Estrogen receptor signaling as a target for novel breast cancer therapeutics. Biochem.
Pharmacol. 2013, 85, 449–465. [CrossRef]
14. Cheskis, B.J.; Greger, J.G.; Nagpal, S.; Freedman, L.P. Signaling by estrogens. J. Cell. Physiol. 2007, 213, 610–617. [CrossRef]
[PubMed]
15. Sever, R.; Brugge, J.S. Signal transduction in cancer. Cold Spring Harb. Perspect. Med. 2015, 5, a006098. [CrossRef] [PubMed]
Molecules 2024, 29, 5275 19 of 23
16. Miziak, P.; Baran, M.; Błaszczak, E.; Przybyszewska-Podstawka, A.; Kałafut, J.; Smok-Kalwat, J.; Dmoszy ´nska-Graniczka, M.;
Kiełbus, M.; Stepulak, A. Estrogen Receptor Signaling in Breast Cancer. Cancers 2023, 15, 4689. [CrossRef] [PubMed]
17. Clusan, L.; Ferrière, F.; Flouriot, G.; Pakdel, F. A Basic Review on Estrogen Receptor Signaling Pathways in Breast Cancer. Int. J.
Mol. Sci. 2023, 24, 6834. [CrossRef] [PubMed]
18. Bjornstrom, L.; Sjoberg, M. Mechanisms of estrogen receptor signaling: Convergence of genomic and nongenomic actions on
target genes. Mol. Endocrinol. 2005, 19, 833–842. [CrossRef]
19. Saha Roy, S.; Vadlamudi, R.K. Role of estrogen receptor signaling in breast cancer metastasis. Int. J. Breast Cancer 2012, 2012,
654698. [CrossRef]
20. Roskoski, R., Jr. The ErbB/HER family of protein-tyrosine kinases and cancer. Pharmacol. Res. 2014, 79, 34–74. [CrossRef]
21. Arteaga, C.L.; Engelman, J.A. ERBB receptors: From oncogene discovery to basic science to mechanism-based cancer therapeutics.
Cancer Cell 2014, 25, 282–303. [CrossRef]
22. Wee, P.; Wang, Z. Epidermal growth factor receptor cell proliferation signaling pathways. Cancers 2017, 9, 52. [CrossRef]
23. Wieduwilt, M.J.; Moasser, M.M. The epidermal growth factor receptor family: Biology driving targeted therapeutics. Cell. Mol.
Life Sci. 2008, 65, 1566–1584. [CrossRef]
24. Garrett, T.P.; McKern, N.M.; Lou, M.; Elleman, T.C.; Adams, T.E.; Lovrecz, G.O.; Kofler, M.; Jorissen, R.N.; Nice, E.C.; Burgess,
A.W.; et al. The crystal structure of a truncated ErbB2 ectodomain reveals an active conformation, poised to interact with other
ErbB receptors. Mol. Cell 2003, 11, 495–505. [CrossRef] [PubMed]
25. Graus-Porta, D.; Beerli, R.R.; Daly, J.M.; Hynes, N.E. ErbB-2, the preferred heterodimerization partner of all ErbB receptors, is a
mediator of lateral signaling. EMBO J. 1997, 16, 1647–1655. [CrossRef] [PubMed]
26. Burgess, A.W. EGFR family: Structure physiology signalling and therapeutic targets. Growth Factors 2008, 26, 263–274. [CrossRef]
27. Mayer, I.A.; Arteaga, C.L. The pi3K/AKT pathway as a target for cancer treatment. Annu. Rev. Med. 2016, 67, 11–28. [CrossRef]
28. Ono, M.; Kuwano, M. Molecular mechanisms of epidermal growth factor receptor (EGFR) activation and response to gefitinib
and other EGFR-targeting drugs. Clin. Cancer Res. 2006, 12, 7242–7251. [CrossRef]
29. Yousefnia, S.; Seyed Forootan, F.; Seyed Forootan, S.; Nasr Esfahani, M.H.; Gure, A.O.; Ghaedi, K. Mechanistic pathways of
malignancy in breast cancer stem cells. Front. Oncol. 2020, 10, 452. [CrossRef]
30. Hosseini, H.; Obradović, M.M.; Hoffmann, M.; Harper, K.L.; Sosa, M.S.; Werner-Klein, M.; Nanduri, L.K.; Werno, C.; Ehrl, C.;
Maneck, M.; et al. Early dissemination seeds metastasis in breast cancer. Nature 2016, 540, 552. [CrossRef] [PubMed]
31. Feng, Y.; Spezia, M.; Huang, S.; Yuan, C.; Zeng, Z.; Zhang, L.; Ji, X.; Liu, W.; Huang, B.; Luo, W.; et al. Breast cancer development
and progression: Risk factors, cancer stem cells, signaling pathways, genomics, and molecular pathogenesis. Genes Dis. 2018, 5,
77–106. [CrossRef]
32. Yang, K.; Wang, X.; Zhang, H.; Wang, Z.; Nan, G.; Li, Y.; Zhang, F.; Mohammed, M.K.; Haydon, R.C.; Luu, H.H.; et al. The
evolving roles of canonical WNT signaling in stem cells and tumorigenesis: Implications in targeted cancer therapies. Lab. Investig.
2016, 96, 116–136. [CrossRef]
33. Mohammed, M.K.; Shao, C.; Wang, J.; Wei, Q.; Wang, X.; Collier, Z.; Tang, S.; Liu, H.; Zhang, F.; Huang, J.; et al. Wnt/beta-catenin
signaling plays an ever-expanding role in stem cell self-renewal, tumorigenesis and cancer chemoresistance. Genes Dis. 2016, 3,
11–40. [CrossRef]
34. Clevers, H.; Nusse, R. Wnt/beta-catenin signaling and disease. Cell 2012, 149, 1192–1205. [CrossRef] [PubMed]
35. Nusse, R.; Clevers, H. Wnt/beta-Catenin signaling, disease, and emerging therapeutic modalities. Cell 2017, 169, 985–999.
[CrossRef] [PubMed]
36. Ugolini, F.; Adélaïde, J.; Charafe-Jauffret, E.; Nguyen, C.; Jacquemier, J.; Jordan, B.; Birnbaum, D.; Pébusque, M.J. Differential
expression assay of chromosome arm 8p genes identifies Frizzled-related (FRP1/FRZB) and Fibroblast Growth Factor Receptor 1
(FGFR1) as candidate breast cancer genes. Oncogene 1999, 18, 1903–1910. [CrossRef] [PubMed]
37. Veeck, J.; Geisler, C.; Noetzel, E.; Alkaya, S.; Hartmann, A.; Knüchel, R.; Dahl, E. Epigenetic inactivation of the secreted frizzled-
related protein-5 (SFRP5) gene in human breast cancer is associated with unfavorable prognosis. Carcinogenesis 2008, 29, 991–998.
[CrossRef]
38. Khramtsov, A.I.; Khramtsova, G.F.; Tretiakova, M.; Huo, D.; Olopade, O.I.; Goss, K.H. Wnt/beta-catenin pathway activation is
enriched in basal-like breast cancers and predicts poor outcome. Am. J. Pathol. 2010, 176, 2911–2920. [CrossRef]
39. Lin, S.Y.; Xia, W.; Wang, J.C.; Kwong, K.Y.; Spohn, B.; Wen, Y.; Pestell, R.G.; Hung, M.C. Beta-catenin, a novel prognosticmarker for
breast cancer: Its roles in cyclin D1 expression and cancer progression. Proc. Natl. Acad. Sci. USA 2000, 97, 4262–4266. [CrossRef]
40. Schade, B.; Lesurf, R.; Sanguin-Gendreau, V.; Bui, T.; Deblois, G.; O’Toole, S.A.; Millar, E.K.; Zardawi, S.J.; Lopez-Knowles, E.;
Sutherland, R.L.; et al. beta-Catenin signaling is a critical event in ErbB2-mediated mammary tumor progression. Cancer Res.
2013, 73, 4474–4487. [CrossRef]
41. Nurse, P.M. Nobel Lecture. Cyclin dependent kinases and cell cycle control. Biosci. Rep. 2002, 22, 487–499. [CrossRef] [PubMed]
42. Sutherland, R.L.; Musgrove, E.A. Cyclins and breast cancer. J. Mammary Gland. Biol. Neoplasia 2004, 9, 95–104. [CrossRef]
43. Radtke, F.; Raj, K. The role of Notch in tumorigenesis: Oncogene or tumour suppressor? Nat. Rev. Cancer 2003, 3, 756–767.
[CrossRef]
44. Reedijk, M.; Odorcic, S.; Chang, L.; Zhang, H.; Miller, N.; McCready, D.R.; Lockwood, G.; Egan, S.E. High-level coexpression
of JAG1 and NOTCH1 is observed in human breast cancer and is associated with poor overall survival. Cancer Res. 2005, 65,
8530–8537. [CrossRef] [PubMed]
Molecules 2024, 29, 5275 20 of 23
45. Dickson, B.C.; Mulligan, A.M.; Zhang, H.; Lockwood, G.; O’Malley, F.P.; Egan, S.E.; Reedijk, M. High-level JAG1mRNA and
protein predict poor outcome in breast cancer. Mod. Pathol. 2007, 20, 685–693. [CrossRef] [PubMed]
46. García-Zaragoza, E.; Pérez-Tavarez, R.; Ballester, A.; Lafarga, V.; Jiménez-Reinoso, A.; Ramírez, Á.; Murillas, R.; Gallego, M.I.
Intraepithelial paracrine Hedgehog signaling induces the expansion of ciliated cells that express diverse progenitor cell markers
in the basal epithelium of the mouse mammary gland. Dev. Biol. 2012, 372, 28–44. [CrossRef] [PubMed]
47. Fiaschi, M.; Rozell, B.; Bergstrom, A.; Toftgard, R.; Kleman, M.I. Targeted expression of GLI1 in the mammary gland disrupts
pregnancy-induced maturation and causes lactation failure. J. Biol. Chem. 2007, 282, 36090–36101. [CrossRef] [PubMed]
48. Miah, S.; Martin, A.; Lukong, K.E. Constitutive activation of breasttumor kinase accelerates cell migration and tumor growth
in vivo. Oncogenesis 2012, 1, e11. [CrossRef]
49. Koren, S.; Reavie, L.; Couto, J.P.; De Silva, D.; Stadler, M.B.; Roloff, T.; Britschgi, A.; Eichlisberger, T.; Kohler, H.; Aina, O.; et al.
PIK3CA(H1047R) induces multipotency and multi-lineage mammary tumours. Nature 2015, 525, 114–118. [CrossRef]
50. Kim, E.K.; Kim, H.A.; Koh, J.S.; Kim, M.S.; Kim, K.I.; Lee, J.I.; Moon, N.M.; Ko, E.; Noh, W.C. Phosphorylated S6K1 is a possible
marker for endocrine therapy resistance in hormone receptor-positive breast cancer. Breast Cancer Res. Treat. 2011, 126, 93–99.
[CrossRef] [PubMed]
51. Yang, Y.; Leonard, M.; Zhang, Y.; Zhao, D.; Mahmoud, C.; Khan, S.; Wang, J.; Lower, E.E.; Zhang, X. HER2-Driven breast
tumorigenesis relies upon interactions of the estrogen receptor with coactivator MED1. Cancer Res. 2018, 78, 422–435. [CrossRef]
52. Shrihastini, V.; Muthuramalingam, P.; Adarshan, S.; Sujitha, M.; Chen, J.T.; Shin, H.; Ramesh, M. Plant derived bioactive
compounds, their anti-cancer effects and in silico approaches as an alternative target treatment strategy for breast cancer: An
updated overview. Cancers 2021, 13, 6222. [CrossRef]
53. Seca, A.M.; Pinto, D.C. Plant secondary metabolites as anticancer agents: Successes in clinical trials and therapeutic application.
Int. J. Mol. Sci. 2018, 19, 263. [CrossRef]
54. Yadegarynia, S.; Pham, A.; Ng, A.; Nguyen, D.; Lialiutska, T.; Bortolazzo, A.; Sivryuk, V.; Bremer, M.; White, J.B. Profiling
flavonoid cytotoxicity in human breast cancer cell lines: Determination of structure-function relationships. Nat. Prod. Commun.
2012, 7(10), 1295–1304. [CrossRef] [PubMed]
55. Sun, Y.; Duan, X.; Wang, F.; Tan, H.; Hu, J.; Bai, W.; Wang, X.; Wang, B.; Hu, J. Inhibitory Effects of Flavonoids on Glucose
Transporter 1 (GLUT1): From Library Screening to Biological Evaluation to Structure-Activity Relationship. Toxicology 2023, 488,
153475. [CrossRef] [PubMed]
56. Li, Z.; Li, J.; Mo, B.; Hu, C.; Liu, H.; Qi, H.; Wang, X.; Xu, J. Genistein induces G2/M cell cycle arrest via stable activation of
ERK1/2 pathway in MDA-MB-231 breast cancer cells. Cell Biol. Toxicol. 2008, 24, 401–409. [CrossRef]
57. Xue, J.P.; Wang, G.; Zhao, Z.B.; Wang, Q.; Shi, Y. Synergistic cytotoxic effect of genistein and doxorubicin on drug-resistant human
breast cancer MCF-7/Adr cells. Oncol. Rep. 2014, 32, 1647–1653. [CrossRef] [PubMed]
58. Scambia, G.; Ranelletti, F.O.; Panici, P.B.; De Vincenzo, R.; Bonanno, G.; Ferrandina, G.; Piantelli, M.; Bussa, S.; Rumi, C.;
Cianfriglia, M.; et al. Quercetin potentiates the effect of adriamycin in a multidrug resistant MCF-7 human breast-cancer cell line:
P-glycoprotein as a possible target. Cancer Chemother. Pharmacol. 1994, 34, 459–464. [CrossRef]
59. Du, G.; Lin, H.; Yang, Y.; Zhang, S.; Wu, X.; Wang, M.; Ji, L.; Lu, L.; Yu, L.; Han, G. Dietary quercetin combining intratumoral
doxorubicin injection synergistically induces rejection of established breast cancer in mice. Int. Immunopharmacol. 2010, 10,
819–826. [CrossRef]
60. Effat, H.; Abosharaf, H.A.; Radwan, A.M. Combined effects of naringin and doxorubicin on the JAK/STAT signaling pathway
reduce the development and spread of breast cancer cells. Sci. Rep. 2024, 14, 2824. [CrossRef]
61. Küpeli Akkol, E.; Genç, Y.; Karpuz, B.; Sobarzo-Sánchez, E.; Capasso, R. Coumarins and coumarin-related compounds in
pharmacotherapy of cancer. Cancers 2020, 12, 1959. [CrossRef]
62. Lake, B. Synthesis & pharmacological investigation of 4-hydroxy coumarin derivatives & shown as anti-coagulant. Food Chem.
Tox. 1999, 3, 412–423.
63. Lacy, A.; O’Kennedy, R. Studies on coumarins and coumarin-related compounds to determine their therapeutic role in the
treatment of cancer. Curr. Pharm. Des. 2004, 10, 3797–3811. [CrossRef]
64. Cui, N.; Lin, D.D.; Shen, Y.; Shi, J.G.; Wang, B.; Zhao, M.Z.; Zheng, L.; Chen, H.; Shi, J.H. Triphenylethylene-coumarin hybrid
TCH-5c suppresses tumorigenic progression in breast cancer mainly through the inhibition of angiogenesis. Anti-Cancer Agents
Med. Chem. 2019, 19, 1253–1261. [CrossRef] [PubMed]
65. Valente, S.; Bana, E.; Viry, E.; Bagrel, D.; Kirsch, G. Synthesis and biological evaluation of novel coumarin-based inhibitors of
Cdc25 phosphatases. Bioorg. Med. Chem. Lett. 2010, 20, 5827–5830. [CrossRef] [PubMed]
66. Lake, B. Coumarin Metabolism, Toxicity and Carcinogenicity: Relevance for Human Risk Assessment. Food Chem. Tox. 1999, 37,
423–453. [CrossRef]
67. Bogan, D.; Deasy, B.; O’Kennedy, R.; Smyth, M. Determination of Free and Total 7-hydroxycoumarin in Urine and Serum by
Capillary Electrophoresis. J. Chromatogr. B 1995, 663, 371–378. [CrossRef]
68. Singh, A.; Singh, K.; Kaur, K.; Singh, A.; Sharma, A.; Kaur, K.; Kaur, J.; Kaur, G.; Kaur, U.; Kaur, H.; et al. Coumarin as an Elite
Scaffold in Anti-Breast Cancer Drug Development: Design Strategies, Mechanistic Insights, and Structure–Activity Relationships.
Biomedicines 2024, 12, 1192. [CrossRef]
69. Liu, J.L.; Pan, Y.Y.; Chen, O.; Luan, Y.; Xue, X.; Zhao, J.J.; Liu, L.; Jia, H.Y. Curcumin inhibits MCF-7 cells by modulating the NF-κB
signaling pathway. Oncol. Lett. 2017, 14, 5581–5584. [CrossRef]
Molecules 2024, 29, 5275 21 of 23
70. Cianfruglia, L.; Minnelli, C.; Laudadio, E.; Scirè, A.; Armeni, T. Side effects of curcumin: Epigenetic and antiproliferative
implications for normal dermal fibroblast and breast cancer cells. Antioxidants 2019, 8, 382. [CrossRef]
71. Calaf, G.M.; Ponce-Cusi, R.; Carrión, F. Curcumin and paclitaxel induce cell death in breast cancer cell lines. Oncol. Rep. 2018, 40,
2381–2388. [CrossRef] [PubMed]
72. Banerjee, S.; Ji, C.; Mayfield, J.E.; Goel, A.; Xiao, J.; Dixon, J.E.; Guo, X. Ancient drug curcumin impedes 26S proteasome activity
by direct inhibition of dual-specificity tyrosine-regulated kinase 2. Proc. Natl. Acad. Sci. USA 2018, 115, 8155–8160. [CrossRef]
73. Petrova, L.; Gergov, N.; Stoup, M.; Zapryanova, S.; Van Damme, E.J.; Lebègue, N.; Liberelle, M.; Zasheva, D.; Bogoeva, V.
Jacalin-Curcumin Complex Sensitizes the Breast Cancer MDA-MB-231 Cell Line. Int. J. Mol. Sci. 2023, 24, 17399. [CrossRef]
74. Wen, C.; Fu, L.; Huang, J.; Dai, Y.; Wang, B.; Xu, G.; Wu, L.; Zhou, H. Curcumin Reverses Doxorubicin Resistance via Inhibition
the Efflux Function of ABCB4 in Doxorubicin-resistant Breast Cancer Cells. Mol. Med. Rep. 2019, 19, 5162–5168. [CrossRef]
[PubMed]
75. Chen, W.-C.; Lai, Y.-A.; Lin, Y.-C.; Ma, J.-W.; Huang, L.-F.; Yang, N.-S.; Ho, C.-T.; Kuo, S.-C.; Way, T.-D. Curcumin Suppresses
Doxorubicin-Induced Epithelial–Mesenchymal Transition via the Inhibition of TGF-β and PI3K/AKT Signaling Pathways in
Triple-Negative Breast Cancer Cells. J. Agric. Food Chem. 2013, 61, 11817–11824. [CrossRef]
76. Ke, C.S.; Liu, H.S.; Yen, C.H.; Huang, G.C.; Cheng, H.C.; Huang, C.Y.; Su, C.L. Curcumin-induced aurora-a suppression not only
causes mitotic defect and cell cycle arrest but also alters chemosensitivity to anticancer drugs. J. Nutr. Biochem. 2014, 25, 526–539.
[CrossRef]
77. Aggarwal, B.B.; Shishodia, S.; Takada, Y.; Banerjee, S.; Newman, R.A.; Bueso-Ramos, C.E.; Price, J.E. Curcumin suppresses the
paclitaxel-induced nuclear factor-kappab pathway in breast cancer cells and inhibits lung metastasis of human breast cancer in
nude mice. Clin. Cancer Res. 2005, 11, 7490–7498. [CrossRef]
78. Mayo, B.; Penroz, S.; Torres, K.; Simón, L. Curcumin Administration Routes in Breast Cancer Treatment. Int. J. Mol. Sci. 2024, 25,
11492. [CrossRef]
79. Bayet-Robert, M.; Kwiatowski, F.; Leheurteur, M.; Gachon, F.; Planchat, E.; Abrial, C.; Mouret-Reynier, M.-A.; Durando, X.;
Barthomeuf, C.; Chollet, P. Phase I Dose Escalation Trial of Docetaxel plus Curcumin in Patients with Advanced and Metastatic
Breast Cancer. Cancer Biol. Ther. 2010, 9, 8–14. [CrossRef] [PubMed]
80. Ryan, J.L.; Heckler, C.E.; Ling, M.; Katz, A.; Williams, J.P.; Pentland, A.P.; Morrow, G.R. Curcumin for Radiation Dermatitis: A
Randomized, Double-Blind, Placebo-Controlled Clinical Trial of Thirty Breast Cancer Patients. Radiat. Res. 2013, 180, 34–43.
[CrossRef]
81. Lao, C.D.; Ruffin, M.T.; Normolle, D.; Heath, D.D.; Murray, S.I.; Bailey, J.M.; Boggs, M.E.; Crowell, J.; Rock, C.L.; Brenner, D.E.
Dose escalation of a curcuminoid formulation. BMC Complement. Altern. Med. 2006, 6, 10. [CrossRef]
82. Hewlings, S.J.; Kalman, D.S. Curcumin: A Review of Its’ Effects on Human Health. Foods 2017, 6, 92. [CrossRef]
83. Luca, S.V.; Macovei, I.; Bujor, A.; Miron, A.; Skalicka-Woźniak, K.; Aprotosoaie, A.C.; Trifan, A. Bioactivity of dietary polyphenols:
The role of metabolites. Crit. Rev. Food Sci. Nutr. 2019, 60, 626–659. [CrossRef]
84. Waheed, A.; Barker, J.; Barton, S.J.; Owen, C.P.; Ahmed, S.; Carew, M.A. A novel steroidal saponin glycoside from Fagonia indica
induces cell-selective apoptosis or necrosis in cancer cells. Eur. J. Pharm. Sci. 2012, 47, 464–473. [CrossRef] [PubMed]
85. Deepa, M.; Priya, S. Purification and characterization of a novel anti-proliferative lectin from Morus alba L. leaves. Protein Pept.
Lett. 2012, 19, 839–845. [CrossRef] [PubMed]
86. Yu, J.S.; Kim, A.K. Platycodin D induces apoptosis in MCF-7 human breast cancer cells. J. Med. Food 2010, 13, 298–305. [CrossRef]
87. Lee, S.K.; Park, K.K.; Kim, H.J.; Kim, K.R.; Kang, E.J.; Kim, Y.L.; Yoon, H.; Kim, Y.S.; Chung, W.Y. Platycodin D Blocks Breast
Cancer-Induced Bone Destruction by Inhibiting Osteoclastogenesis and the Growth of Breast Cancer Cells. Cell Physiol. Biochem.
2015, 36, 1809–1820. [CrossRef]
88. Tang, Z.H.; Li, T.; Gao, H.W.; Sun, W.; Chen, X.P.; Wang, Y.T.; Lu, J.J. Platycodin D from Platycodonis Radix enhances the
anti-proliferative effects of doxorubicin on breast cancer MCF-7 and MDA-MB-231 cells. Chin. Med. 2014, 9, 16. [CrossRef]
[PubMed]
89. Sarikahya, N.B.; Nalbantsoy, A.; Top, H.; Gokturk, R.S.; Sumbul, H.; Kirmizigul, S. Immunomodulatory, hemolytic and cytotoxic
activity potentials of triterpenoid saponins from eight Cephalaria species. Phytomedicine 2018, 38, 135–144. [CrossRef] [PubMed]
90. Zasheva, D.; Mladenov, P.; Rusanov, K.; Simova, S.; Zapryanova, S.; Simova-Stoilova, L.; Moyankova, D.; Djilianov, D. Fractions
of methanol extracts from the Resurrection plant Haberlea rhodopensis have anti-breast cancer effects in model cell systems.
Separations 2023, 10, 388. [CrossRef]
91. Thilagavathi, R.; Priyankha, S.; Kannan, M.; Prakash, M.; Selvam, C. Compounds from diverse natural origin against triple-
negative breast cancer: A comprehensive review. Chem. Biol. Drug Des. 2023, 101, 218–243. [CrossRef]
92. Greenshields, A.L.; Doucette, C.D.; Sutton, K.M.; Madera, L.; Annan, H.; Yaffe, P.B.; Knickle, A.F.; Dong, Z.; Hoskin, D.W. Piperine
inhibits the growth and motility of triple-negative breast cancer cells. Cancer Lett. 2015, 357, 129–140. [CrossRef]
93. Delaney, L.M.; Farias, N.; Ghassemi Rad, J.; Fernando, W.; Annan, H.; Hoskin, D.W. The natural alkaloid Piperlongumine inhibits
metastatic activity and epithelial-to-Mesenchymal transition of triple-negative mammary carcinoma cells. Nutr. Cancer 2020, 73,
2397–2410. [CrossRef]
94. Zhai, H.Y.; Zhao, C.; Zhang, N.; Jin, M.N.; Tang, S.A.; Qin, N.; Kong, D.X.; Duan, H.Q. Alkaloids from Pachysandra terminalis
inhibit breast cancer invasion and have potential for development as antimetastasis therapeutic agents. J. Nat. Prod. 2012, 75,
1305–1311. [CrossRef] [PubMed]
Molecules 2024, 29, 5275 22 of 23
95. Ali Abdalla, Y.O.; Subramaniam, B.; Nyamathulla, S.; Shamsuddin, N.; Arshad, N.M.; Mun, K.S.; Awang, K.; Nagoor, N.H.
Natural products for cancer therapy: A review of their mechanism of actions and toxicity in the past decade. J. Trop. Med. 2022,
2022, 5794350. [CrossRef] [PubMed]
96. Quijia, C.R.; Chorilli, M. Piperine for treating breast cancer: A review of molecular mechanisms, combination with anticancer
drugs, and nanosystems. Phytother. Res. 2022, 36, 147–163. [CrossRef] [PubMed]
97. Li, G.; Shan, C.; Liu, L.; Zhou, T.; Zhou, J.; Hu, X.; Chen, Y.; Cui, H.; Gao, N. Tanshinone IIA inhibits HIF-1α and VEGF expression
in breast cancer cells via mTOR/p70S6K/RPS6/4E-BP1 signaling pathway. PLoS ONE 2015, 10, e0117440. [CrossRef]
98. Kong, Y.; Chen, C. KHF16, a new cycloartane triterpenoid isolated from Cimicifuga foetida, suppresses triple-negative breast
cancer by inhibiting the NF-κB signaling pathway. Cancer Res. 2015, 75 (Suppl. S15), 3798. [CrossRef]
99. Yang, H.; Ping Dou, Q. Targeting apoptosis pathway with natural terpenoids: Implications for treatment of breast and prostate
cancer. Curr. Drug Targets 2010, 11, 733–744. [CrossRef]
100. Kamran, S.; Sinniah, A.; Abdulghani, M.A.; Alshawsh, M.A. Therapeutic Potential of Certain Terpenoids as Anticancer Agents: A
Scoping Review. Cancers 2022, 14, 1100. [CrossRef]
101. Huang, Y.; Peng, H.; Zeng, A.; Song, L. The role of peptides in reversing chemoresistance of breast cancer: Current facts and
future prospects. Front. Pharmacol. 2023, 14, 1188477. [CrossRef]
102. Ghaly, G.; Tallima, H.; Dabbish, E.; Badr ElDin, N.; Abd El-Rahman, M.K.; Ibrahim, M.A.A.; Shoeib, T. Anti-Cancer Peptides:
Status and Future Prospects. Molecules 2023, 28, 1148. [CrossRef]
103. Fang, X.Y.; Chen, W.; Fan, J.T.; Song, R.; Wang, L.; Gu, Y.H.; Zeng, G.Z.; Shen, Y.; Wu, X.F.; Tan, N.H.; et al. Plant cyclopeptide
RA-V kills human breast cancer cells by inducing mitochondria-mediated apoptosis through blocking PDK1–AKT interaction.
Toxicol. Appl. Pharmacol. 2013, 267, 95–103. [CrossRef]
104. Chiangjong, W.; Chutipongtanate, S.; Hongeng, S. Anticancer peptide: Physicochemical property, functional aspect and trend in
clinical application. Int. J. Oncol. 2020, 57, 678–696. [CrossRef] [PubMed]
105. Li, Q.Z.; Zhou, Z.R.; Hu, C.Y.; Li, X.B.; Chang, Y.Z.; Liu, Y.; Wang, Y.L.; Zhou, X.W. Recent advances of bioactive pro-
teins/polypeptides in the treatment of breast cancer. Food Sci. Biotechnol. 2023, 32, 265–282. [CrossRef] [PubMed]
106. Hsieh, C.C.; Hernández-Ledesma, B.; Jeong, H.J.; Park, J.H.; de Lumen, B.O. Complementary roles in cancer prevention: Protease
inhibitor makes the cancer preventive peptide lunasin bioavailable. PLoS ONE 2010, 5, e8890. [CrossRef] [PubMed]
107. Kaufman-Szymczyk, A.; Kaczmarek, W.; Fabianowska-Majewska, K.; Lubecka-Gajewska, K. Lunasin and Its Epigenetic Impact in
Cancer Chemoprevention. Int. J. Mol. Sci. 2023, 24, 9187. [CrossRef]
108. Jiang, Q.; Pan, Y.; Cheng, Y.; Li, H.; Liu, D.; Li, H. Lunasin suppresses the migration and invasion of breast cancer cells by
inhibiting matrix metalloproteinase-2/-9 via the FAK/Akt/ERK and NF-κB signaling pathways. Oncol. Rep. 2016, 36, 253–262.
[CrossRef]
109. Hsieh, C.C.; Wu, C.H.; Peng, S.H.; Chang, C.H. Seed-derived peptide lunasin suppressed breast cancer cell growth by regulating
inflammatory mediators, aromatase, and estrogen receptors. Food Nutr. Res. 2023, 67, 8991. [CrossRef]
110. Hao, Y.; Guo, H.; Hong, Y.; Fan, X.; Su, Y.; Yang, X.; Ren, G. Lunasin peptide promotes lysosome-mitochondrial mediated
apoptosis and mitotic termination in MDA-MB-231 cells. Food Sci. Hum. Wellness 2022, 11, 1598–1606. [CrossRef]
111. Hao, Y.; Wu, B.; Li, M.; Yuan, M.; Qiao, L.; Zhao, J.; Zheng, X.; Li, X.; Wang, Y.; Wang, Y.; et al. Functional exploration of lunasin
peptide in transgenic maize (Zea mays L.) and its role in controlling mitophagy in MDA-MB-231 cells. Food Biosci. 2024, 58, 103726.
[CrossRef]
112. Pabona, J.M.P.; Dave, B.; Su, Y.; Montales, M.T.E.; De Lumen, B.O.; De Mejia, E.G.; Rahal, O.M.; Simmen, R.C. The soybean
peptide lunasin promotes apoptosis of mammary epithelial cells via induction of tumor suppressor PTEN: Similarities and
distinct actions from soy isoflavone genistein. Genes Nutr. 2013, 8, 79–90. [CrossRef]
113. Song, R.; Qiao, W.; He, J.; Huang, J.; Luo, Y.; Yang, T. Proteases and their modulators in cancer therapy: Challenges and
opportunities. J. Med. Chem. 2021, 64, 2851–2877. [CrossRef]
114. Hoelzen, L.; Mitschke, J.; Schoenichen, C.; Hess, M.E.; Ehrenfeld, S.; Boerries, M.; Miething, C.; Brummer, T.; Reinheckel, T. RNA
Interference screens discover proteases as synthetic lethal partners of PI3K inhibition in breast cancer cells. Theranostics 2022, 12,
4348. [CrossRef] [PubMed]
115. Hashemi, M.; Paskeh, M.D.A.; Orouei, S.; Abbasi, P.; Khorrami, R.; Dehghanpour, A.; Esmaeili, N.; Ghahremanzade, A.; Zandieh,
M.A.; Peymani, M.; et al. Towards dual function of autophagy in breast cancer: A potent regulator of tumor progression and
therapy response. Biomed. Pharmacother. 2023, 161, 114546. [CrossRef]
116. Cid-Gallegos, M.S.; Corzo-Ríos, L.J.; Jiménez-Martínez, C.; Sánchez-Chino, X.M. Protease inhibitors from plants as therapeutic
agents—A review. Plant Foods Hum. Nutr. 2022, 77, 20–29. [CrossRef] [PubMed]
117. Gitlin-Domagalska, A.; Maciejewska, A.; D˛ebowski, D. Bowman-Birk inhibitors: Insights into family of multifunctional proteins
and peptides with potential therapeutical applications. Pharmaceuticals 2020, 13, 421. [CrossRef]
118. Moy, L.Y.; Billings, P.C. A proteolytic activity in a human breast cancer cell line which is inhibited by the anticarcinogenic
Bowman-Birk protease inhibitor. Cancer Lett. 1994, 85, 205–210. [CrossRef]
119. Chen, Y.W.; Huang, S.C.; Lin-Shiau, S.Y.; Lin, J.K. Bowman–Birk inhibitor abates proteasome function and suppresses the
proliferation of MCF7 breast cancer cells through accumulation of MAP kinase phosphatase-1. Carcinogenesis 2005, 26, 1296–1306.
[CrossRef] [PubMed]
Molecules 2024, 29, 5275 23 of 23
120. Joanitti, G.A.; Azevedo, R.B.; Freitas, S.M. Apoptosis and lysosome membrane permeabilization induction on breast cancer cells
by an anticarcinogenic Bowman–Birk protease inhibitor from Vigna unguiculata seeds. Cancer Lett. 2010, 293, 73–81. [CrossRef]
121. Mehdad, A.; Brumana, G.; Souza, A.A.; Barbosa JA, R.G.; Ventura, M.M.; De Freitas, S.M. A Bowman–Birk inhibitor induces
apoptosis in human breast adenocarcinoma through mitochondrial impairment and oxidative damage following proteasome 20S
inhibition. Cell Death Discov. 2016, 2, 15067. [CrossRef]
122. Kyani, S.; Akrami, H.; Mostafaei, A.; Akbari, S.; Salehi, Z. Inhibitory effect of Bowman–Birk protease inhibitor on autophagy in
MDAMB231 breast cancer cell line. J. Cancer Res. Ther. 2021, 17, 504–509. [CrossRef]
123. Zhang, L.; Wan, X.S.; Donahue, J.J.; Ware, J.H.; Kennedy, A.R. Effects of the Bowman-Birk Inhibitor on Clonogenic Survival and
Cisplatin- or Radiation-Induced Cytotoxicity in Human Breast, Cervical, and Head and Neck Cancer Cells. Nutr. Cancer 1999, 33,
165–173. [CrossRef]
124. Lanza, A.; Tava, A.; Catalano, M.; Ragona, L.; Singuaroli, I.; della Cuna FS, R.; della Cuna, G.R. Effects of the Medicago scutellata
trypsin inhibitor (MsTI) on cisplatin-induced cytotoxicity in human breast and cervical cancer cells. Anticancer Res. 2004, 24,
227–234. [PubMed]
125. Gueven, N.; Dittmann, K.; Mayer, C.; Rodemann, H.P. The radioprotective potential of the Bowman–Birk protease inhibitor is
independent of its secondary structure. Cancer Lett. 1998, 125, 77–82. [CrossRef] [PubMed]
126. Dittmann, K.; Löffler, H.; Bamberg, M.; Rodemann, H.P. Bowman–Birk proteinase inhibitor (BBI) modulates radiosensitivity and
radiation induced differentiation of human fibroblasts in culture. Radiother. Oncol. 1995, 34, 137–143. [CrossRef] [PubMed]
127. Lozon, L.; Ramadan, W.S.; Kawaf, R.R.; Al-Shihabi, A.M.; El-Awady, R. Decoding cell death signalling: Impact on the response of
breast cancer cells to approved therapies. Life Sci. 2024, 342, 122525. [CrossRef] [PubMed]
128. Naeem, M.; Iqbal, M.O.; Khan, H.; Ahmed, M.M.; Farooq, M.; Aadil, M.M.; Jamaludin, M.I.; Hazafa, A.; Tsai, W.-C. A Review of
Twenty Years of Research on the Regulation of Signaling Pathways by Natural Products in Breast Cancer. Molecules 2022, 27, 3412.
[CrossRef]
129. HemaIswarya, S.; Doble, M. Potential synergism of natural products in the treatment of cancer. Phytother. Res. An. Int. J. Devoted
Pharmacol. Toxicol. Eval. Nat. Prod. Deriv. 2006, 20, 239–249. [CrossRef]
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.