ArticlePDF AvailableLiterature Review

Targeting Notch signaling pathways with natural bioactive compounds: a promising approach against cancer

Frontiers
Frontiers in Pharmacology
Authors:

Abstract and Figures

Notch signaling pathway is activated abnormally in solid and hematological tumors, which perform essential functions in cell differentiation, survival, proliferation, and angiogenesis. The activation of Notch signaling and communication among Notch and other oncogenic pathways heighten malignancy aggressiveness. Thus, targeting Notch signaling offers opportunities for improved survival and reduced disease incidence. Already, most attention has been given to its role in the cancer cells. Recent research shows that natural bioactive compounds can change signaling molecules that are linked to or interact with the Notch pathways. This suggests that there may be a link between Notch activation and the growth of tumors. Here, we sum up the natural bioactive compounds that possess inhibitory effects on human cancers by impeding the Notch pathway and preventing Notch crosstalk with other oncogenic pathways, which provoke further study of these natural products to derive rational therapeutic regimens for the treatment of cancer and develop novel anticancer drugs. This review revealed Notch as a highly challenging but promising target in oncology.
Notch receptors are initially produced as a solitary polypeptide in signal-receiving cells. These receptors are subsequently divided by Furin-like convertase(s) in the trans-Golgi network (S1) and combine to create a heterodimer. During trafficking, this heterodimeric receptor is conveyed to the cellular membrane. In the meantime, Notch ligands in sender cells can attach to Notch receptors in receiver cells. The contact between the receptor and ligand triggers a second cleavage (S2) in the extracellular domain, which is facilitated by the ADAM (A disintegrin and metalloprotease). The Notch extracellular domain (NECD) has a role in the binding of the ligand. Subsequently, a third cleavage (S3) takes place within the transmembrane domain, facilitated by the gamma-secretase function of the presenilin, Nicastrin, Anterior pharynx-defective 1 (APH-1), and Presenilin enhancer 2 (PEN-2) multi-protein complex. Lastly, the intracellular domain of Notch (NICD) is liberated and migrates to the nucleus, where it interacts with the transcription factor CSL (CBF1, Suppressor of Hairless, Lag-1). This connection results in the stimulation of transcription by blocking co-repressors and simultaneously attracting co-activators like mastermind, so facilitating the transcription of Notch target genes. Note: Mastermind-like (MAML); Histone acetyl transferases (HATs); Ski-interacting protein (SKIP); MYC proto-oncogene (Myc); Cold-inducible RNA-binding protein (CIR); Histone deacetylases (HDAC); Nuclear receptor corepressor 2 (SMRT); C-terminal binding protein (CtBP).
… 
This content is subject to copyright.
Targeting Notch signaling
pathways with natural bioactive
compounds: a promising
approach against cancer
Jia Yang
1
,
2
,
3
, Qihui Sun
1
,
4
, Xiaoyun Liu
5
, Yong Yang
5
,
Rong Rong
1
,
2
*, Peiyu Yan
2
* and Ying Xie
3
*
1
College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China,
2
State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and
Technology, Taipa, Macao SAR, China,
3
State Key Laboratory of Traditional Chinese Medicine Syndrome,
The Second Afliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong,
China,
4
School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China,
5
Key
Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University
of Traditional Chinese Medicine, Jinan, Shandong, China
Notch signaling pathway is activated abnormally in solid and hematological
tumors, which perform essential functions in cell differentiation, survival,
proliferation, and angiogenesis. The activation of Notch signaling and
communication among Notch and other oncogenic pathways heighten
malignancy aggressiveness. Thus, targeting Notch signaling offers
opportunities for improved survival and reduced disease incidence. Already,
most attention has been given to its role in the cancer cells. Recent research
shows that natural bioactive compounds can change signaling molecules that are
linked to or interact with the Notch pathways. This suggests that there may be a
link between Notch activation and the growth of tumors. Here, we sum up the
natural bioactive compounds that possess inhibitory effects on human cancers by
impeding the Notch pathway and preventing Notch crosstalk with other
oncogenic pathways, which provoke further study of these natural products
to derive rational therapeutic regimens for the treatment of cancer and develop
novel anticancer drugs. This review revealed Notch as a highly challenging but
promising target in oncology.
KEYWORDS
notch signaling, crosstalk, oncogenic pathways, phytochemicals, cancer
OPEN ACCESS
EDITED BY
Antonella DAnneo,
University of Palermo, Italy
REVIEWED BY
Magesh Muthu,
Wayne State University, United States
Armel Hervé Nwabo Kamdje,
University of Garoua, Cameroon
Maria Pia Felli,
Sapienza University of Rome, Italy
*CORRESPONDENCE
Ying Xie,
leoxieying16@outlook.com
Peiyu Yan,
pyyan@must.edu.mo
Rong Rong,
rosierong@163.com
RECEIVED 05 April 2024
ACCEPTED 27 June 2024
PUBLISHED 18 July 2024
CITATION
Yang J, Sun Q, Liu X, Yang Y, Rong R, Yan P and
Xie Y (2024), Targeting Notch signaling
pathways with natural bioactive compounds: a
promising approach against cancer.
Front. Pharmacol. 15:1412669.
doi: 10.3389/fphar.2024.1412669
COPYRIGHT
© 2024 Yang, Sun, Liu, Yang, Rong, Yan and Xie.
This is an open-access article distributed under
the terms of the Creative Commons Attribution
License (CC BY). The use, distribution or
reproduction in other forums is permitted,
provided the original author(s) and the
copyright owner(s) are credited and that the
original publication in this journal is cited, in
accordance with accepted academic practice.
No use, distribution or reproduction is
permitted which does not comply with these
terms.
Abbreviations: Notch1/2/3/4, Notch receptor 1/2/3/4; DLL1/3/4, Delta-like protein 1/3/4; ADAM, A
disintegrin and metalloprotease; NECD, extracellular domain; NICD, intracellular domain of Notch; CSL,
CBF1, Suppressor of Hairless, Lag-1; EGCG, Epigallocatechin-3-gallate; GSIs, gamma-secretase
inhibitors; RBPJ, recombination signal-binding protein of immunoglobulin kappa J region; PTCH,
patched receptor; SMO, smoothened; SUFU, suppressor of fused; COS2, costal 2; PKA, protein
kinase A; SLIMB, supernumerary limbs; Ci/Gli, Cubitus interruptus/zinc nger protein-Glioma-
associated oncogene homolog1; ErbB2, HER2/Neu; PDGF, platelet-derived growth factor; EGFR,
epidermal growth factor receptor; TGF-β, transforming growth factor type beta; PTEN, phosphatase
and tensin homolog; PP2A, protein phosphatase 2; TCF/LEF, T-cell factor/lymphoid enhancer factor;
PI3K, Phosphatidylinositol 3-Kinase; AKT, protein kinase B; ERK, extracellular signal-regulated kinase;
mTOR, mammalian target of rapamycin.
Frontiers in Pharmacology frontiersin.org01
TYPE Review
PUBLISHED 18 July 2024
DOI 10.3389/fphar.2024.1412669
GRAPHICAL ABSTRACT
1 Introduction
Cancer is a highly destructive illness that poses a signicant threat to
human wellbeing (Bray et al., 2021). Treatment choices include
radiotherapy, chemotherapy, surgery, immunotherapy, and targeted
therapy (Ullah et al., 2023). However, there has been limited
improvement in the poor prognosis, adverse reactions, drug resistance,
and high recurrence rates. Therefore, discovering new therapeutic drugs
or targets for cancer is required. Notch, an ideal molecular target, is
predominantly expressed in cancerous cells (Espinoza and Miele, 2013).
Notch signaling is a highly conserved pathway that is present in
various types of human malignancies, such as breast, colorectal,
lung, pancreatic, prostate tumors, and glioblastoma (Majumder
et al., 2021). It plays a crucial role in various cellular processes,
including cell proliferation, differentiation, and the maintenance of
cancer stem cells (Zhou et al., 2022).
Clinical research demonstrates an adverse correlation between the
level of Notch expression and patient survival in different kinds of
cancer, indicating that increased Notch activation is associated with
strengthened cancer aggressiveness (Takebe et al., 2014). Thus,
modulating the expression levels of the Notch pathway is a potential
therapeutic approach to treating cancer. Several inhibitors targeting the
Notch pathway have been developed, but their safety and efcacy are
still being evaluated and are not in clinical use (Takebe et al., 2014).
Searching for natural inhibitors of Notch with low toxicity and high
safety,aswellaselucidatingtheir mechanism, could to some extent
address the gap of inhibitors that have not yet been utilized in clinical
practice. On the other hand, recent work indicates that oncogenic
pathways affected by natural bioactive compounds may be linked to or
interact with the Notch pathways, suggesting a potential connection
between Notch signaling and tumor progression.
In this review, we summarize the role of bioactive natural
products in regulating the Notch signaling pathway, particularly
focusing on its activation and interaction with other oncogenic
signaling pathways in various types of cancer. The aim of this study
is to investigate the potential therapeutic applications of natural
products targeting Notch in cancer treatment.
2 Notch signaling pathway
The Notch signaling pathway includes two primary categories:
the canonical pathway and the non-canonical pathway. The
canonical Notch signaling pathway plays a crucial role in
determining cell fate, including cellular communication,
differentiation of embryos and tissues, gene expression, as well as
the development of both benign and malignant diseases (Yamamoto
et al., 2014).
The architecture of the pathway comprises Notch receptors
(Notch 14), Notch receptor ligands, namely, Delta-like (DLL1,
-3, and -4) or Serrate-like (commonly known as Jagged 1 and Jagged
2), transcription elements, co-activators, co-repressors, and
Frontiers in Pharmacology frontiersin.org02
Yang et al. 10.3389/fphar.2024.1412669
downstream effectors (Zhou et al., 2022). The activation of the
Notch signaling pathway occurs when the Notch ligand combines
with the Notch receptor in signal-receiving cells. The Notch
receptors undergo three cleavages before translocating into the
nucleus in order to regulate the transcription of target genes
(Majumder et al., 2021). It is noteworthy that a diverse range of
cellular activities can be triggered by identical signaling pathways in
distinct circumstances. Considering this, cancer progression
activities initiated by Notch and its cross talks are dependent on
the context (Guo et al., 2011). The Notch signaling pathway is
summarized as depicted in Figure 1.
3 Phytochemicals treated cancers via
modulating Notch pathway
Various studies have shown that natural bioactive compounds
offer substantial protection against cancer by inuencing Notch
signaling pathways. This includes suppressing Notch pathway
activation, inhibiting gamma-secretase expression, Notch
transcription complex, and Notch downstream target genes, as
well as preventing communication between Notch and other
cancer-causing pathways.
3.1 Phytochemicals suppressed the Notch
signaling pathways activation
Notch is initiated by the binding of the extracellular domain of
the Notch receptors (Notch1-4) to its ligands (DLL 1, -3, -4, and
Jagged1, -2), which are typically present on the surface of
neighboring signal-sending cells (Reichrath and Reichrath, 2020).
Activation of the Notch signaling pathway controls cell fate and
proliferation in both non-cancerous and cancerous diseases (Zhou
et al., 2022). The following topics are addressed in our discussion on
the regulation of Notch activation using natural products: 1) the
FIGURE 1
Notch receptors are initially produced as a solitary polypeptide in signal-receiving cells. These receptors are subsequently divided by Furin-like
convertase(s) in the trans-Golgi network (S1) and combine to create a heterodimer. During trafcking, this heterodimeric receptor is conveyed to the
cellular membrane. In the meantime, Notch ligands in sender cells can attach to Notch receptors in receiver cells. The contact between the receptor and
ligand triggers a second cleavage (S2) in the extracellular domain, which is facilitated by the ADAM (A disintegrin and metalloprotease). The Notch
extracellular domain (NECD) has a role in the binding of the ligand. Subsequently, a third cleavage (S3) takes place within the transmembrane domain,
facilitated by the gamma-secretase function of the presenilin, Nicastrin, Anterior pharynx-defective 1 (APH-1), and Presenilin enhancer 2 (PEN-2) multi-
protein complex. Lastly, the intracellular domain of Notch (NICD) is liberated and migrates to the nucleus, where it interacts with the transcription factor
CSL (CBF1, Suppressor of Hairless, Lag-1). This connection results in the stimulation of transcription by blocking co-repressors and simultaneously
attracting co-activators like mastermind, so facilitating the transcription of Notch target genes. Note: Mastermind-like (MAML); Histone acetyl
transferases (HATs); Ski-interacting protein (SKIP); MYC proto-oncogene (Myc); Cold-inducible RNA-binding protein (CIR); Histone deacetylases (HDAC);
Nuclear receptor corepressor 2 (SMRT); C-terminal binding protein (CtBP).
Frontiers in Pharmacology frontiersin.org03
Yang et al. 10.3389/fphar.2024.1412669
FIGURE 2
Phytochemicals suppress the activation of the Notch signaling pathway.
TABLE 1 Phytochemicals on Notch downstream target genes in different types of cancer.
Phytochemical Cancer type Hes-1 Hes-5 Hey-1 Hey-2 References
Curcumin Colorectal Carcinoma Chen et al. (2017a)
Pancreatic Cancer Wang et al. (2006a)
Melanoma Cancer Tang and Cao (2022)
Cholangiocarcinoma Koprowski et al. (2015)
Esophageal Cancer Subramaniam et al. (2012)
Honokiol Colon Cancer Ponnurangam et al. (2012)
Curcumin Colon Cancer Bounaama et al. (2012)
EGCG T-ALL Wang et al. (2020)
Tongue Cancer Wei et al. (2022)
3,6-Dihydroxyavone Breast Cancer Chen et al. (2016a)
Resveratrol Osteosarcoma ↓↓↓ Liu et al. (2016)
Triptolide Triple-Negative Breast Cancer ↓↓Ramamoorthy et al. (2021)
Silybin Hepatocellular Carcinoma Zhang et al. (2013)
Withaferin A Breast Cancer ↓↓ Lee et al. (2012)
Carvacrol Prostate Cancer Khan et al. (2019)
Retinoic Acid Glioblastoma Ying et al. (2011)
Oleandrin T-ALL ↓↓ Arai et al. (2018a)
Cowanin T-ALL ↓↓ Arai et al. (2018b)
showed decreased expression.
Frontiers in Pharmacology frontiersin.org04
Yang et al. 10.3389/fphar.2024.1412669
modulation of Notch ligands and receptors expression, and 2) the
interference with ligand-receptor binding.
Epigallocatechin-3-gallate (EGCG), the main polyphenol in
green tea, decreased the expression of Notch receptors including
Notch1 (Hossain et al., 2012;Lee et al., 2013;Toden et al., 2016;
Wang et al., 2020;Ashry et al., 2022) and Notch2 (Jin et al., 2013)in
several cancer models. Notch was activated in the tongue cancer
mice and EGCG signicantly blocked the activation by decreasing
the expression of Notch1, which induced cell apoptosis and
inhibited the proliferation (Wei et al., 2022). In addition, EGCG
also promotes suppression of Notch1 and cleaved Notch1 in 5-
Fluorouracil -resistant cell lines (Toden et al., 2016), suggesting that
EGCG may overcome the 5-Fluorouracil resistance by modulating
the activation of Notch. However, no studies have indicated that
EGCG could regulate Notch ligands.
Resveratrol, isolated from grapes, peanuts, and blueberries,
possesses antitumor and antioxidant properties. It has the
potential to inhibit cell cycle and growth (Pinchot et al., 2011;Yu
X. M. et al., 2013;Zhang et al., 2014), stimulate apoptosis and re-
differentiation (Yu XM. et al., 2013), prevent tumor recurrence
(Giordano et al., 2023), and impede autophagy (Giordano et al.,
2021) in cancer through downregulation of Notch receptors and
ligands, including Notch1, Notch2, Notch4, DLL1, DLL4, and
Jagged1. These studies indicated that resveratrol could hinder the
activation of the Notch pathway through repression of Notch
receptors and ligands in several tumors. However, Notch
signaling might not play a universally critical role in human
medulloblastoma cells given its limited relevance to resveratrol-
induced differentiation and apoptosis (Lacerda-Abreu et al., 2021).
The combined use of a Notch inhibitor and resveratrol
demonstrated greater effectiveness in blocking autophagy,
suggesting its potential as a therapeutic intervention for cancer
(Giordano et al., 2021).
Curcumin, also termed diferuloylmethane, a bright yellow
bioactive compound derived from the plants of the Curcuma
longa species. Curcumin decreased the levels of Notch1 (Liu
et al., 2014;Chen G. P. et al., 2017;Gupta et al., 2017;Siddappa
et al., 2017;Li et al., 2018;Zhdanovskaya et al., 2022), Notch3
(Zhdanovskaya et al., 2022) and Jagged1 (Kiesel and Stan, 2022),
leading to cell death mediated by DNA damage and hindering
cellular self-renewal, apoptosis (Howells et al., 2011;Liu et al.,
2014), motility, migration, and invasion. Importantly, curcumin
could upregulate the levels of miR222-3p (Tang and Cao, 2022)
and miR-34a (Toden et al., 2018) while downregulating miR-27a
(Han et al., 2020), paralogous proteins, and gamma-secretase
protein complex (Subramaniam et al., 2012), ultimately leading
FIGURE 3
An illustrative depiction of the phytochemicals could inuence the interaction between the Notch and Wnt/Hedgehog signaling pathways. In the
gures provided, the term promotesignies that the chemical(s) could enhance the expression or activity of the specic protein. Conversely, the term
inhibitsuggests that the compound(s) can decrease the expression of the protein or hinder its activity.
Frontiers in Pharmacology frontiersin.org05
Yang et al. 10.3389/fphar.2024.1412669
to the inactivation of Notch1. Curcumin treatment suppressed
invasion in human osteosarcoma U2OS cells, and overexpressing
Notch1 reversed this effect (Sha et al., 2016), indicating that the
antitumor effect of curcumin is mediated through Notch1.
However, curcumin had no impact on the expression of
Notch1, but it did hinder the DNA-binding capacity of
cleaved Notch1 in human prostate DU145 cells (Yang
et al., 2017).
Silybin, derived from milk thistle seed, is mainly used in
chronic liver disease and is well known for its anti-
inammatory, hepatoprotective, antiviral, neuroprotective, and
cardioprotective. Results regarding the efcacy of silybin as a
Notch modulator are clear. Notably, the antitumor activity of
silybin was weakened by recombinant human Jagged1,
suggesting that Jagged1 may be utilizedasananti-tumortarget
(Zhang et al., 2013;Kim et al., 2014). However, the synergistic
effect of Notch1 siRNA in vitro or DAPT (C
23
H
26
F
2
N
2
O
4
), a well-
known Notch inhibitor) in vivo, augmented the anticancer potency
of silybin, suggesting that silybin may improve the efcacy of
Notch inhibitors.
Activation of Notch signaling has been implicated in
pathogenesis of various hematologic tumors including
leukemias (Hubmann et al., 2020;Zhang et al., 2022;Skelin
et al., 2024) and lymphomas (Alderuccio and Lossos, 2022).
For example, in lymphopoiesis, the Notch1 activation may be
involved in the correct differentiation of T and B cells (Okatani
et al., 2024). In chronic lymphocytic leukaemia, Notch1 may
represent a potential druggable target (Pozzo et al., 2022),
suggesting that phytochemicals, such as EGCG and curcumin,
may be benecial in the treatment of chronic lymphocytic
leukemia (CLL). Suppressing the activity of Notch1 or
Notch2 resulted in an enhanced response of CLL cells to
udarabine (Del Papa et al., 2019) or venetoclax (Fiorcari
et al., 2022), respectively, indicating that natural compounds
with the ability to decrease Notch activation may be able to
overcome resistance in CLL. It is crucial to highlight that in T cell
acute lymphoblastic leukemia (T-ALL), the Notch1 activating
mutations typically occurs laterinasequenceofgenomic
damages, suggesting that Notch1-based therapies may impede
the efcacy targeted Notch1 therapy (De Bie et al., 2018).
Other agents could also inhibit Notch pathway activation by
controlling the expression of both Notch receptor and ligand, such
as emodin (Deng et al., 2015), carvacrol (Khan et al., 2019),
withaferin A (Lee et al., 2012) and quercetin (Okuhashi et al.,
2011;Peiffer et al., 2020). In summary, several natural bioactive
compounds suppress Notch activation to downregulate cancer
aggressiveness as shown in Figure 2.
3.2 Phytochemicals modulate the Notch
signaling pathway
Phytochemicals may play an anti-tumor role by modulating
other components of the Notch pathway, such as the gamma
secretase complex, the Notch transcription complex and Notch
downstream target genes.
FIGURE 4
An illustrative depiction of the phytochemicals could inuence the interaction between the Notch and EGFR/PDGF/TGF-β/VEGF
signaling pathways.
Frontiers in Pharmacology frontiersin.org06
Yang et al. 10.3389/fphar.2024.1412669
3.2.1 Gamma-secretase inhibitors
The gamma-secretase complex, which consists of presenilin,
Nicastrin, APH-1, and PEN-2 proteins, plays a crucial role in
producing the NICD. Inhibition of secretase activity has shown
signicant antitumor efcacy in cancers, including lung cancer,
colorectal cancer, melanoma, and ovarian cancer (Takebe et al.,
2014). Several gamma-secretase inhibitors (GSIs) have advanced
from preclinical testing to early clinical stages and observed
antitumor activity (Mccaw et al., 2021). Unfortunately, clinical
trials had to be stopped due to serious adverse events, such as
gastrointestinal toxicities, skin disorders, and diarrhea, making the
use of inhibitors less attractive for Notch blockade to arrival
antitumor effects. Therefore, it is necessary to explore new
strategies via inhibiting gamma-secretases.
Cucurbitacin B and I, decrease the expression of Notch receptors
and ligands, repressing the activity of gamma-secretases,
suppressing NICD production, binding to Notch1, subsequently
repressing downstream genes of Notch, inhibiting tumor growth in
colon cancer (Dandawate et al., 2020;Mccaw et al., 2021). Celastrol
and triptolide both reduced the expression of Notch1 and its
downstream target proteins, hence regulating the renewal of stem
cells in triple negative breast cancer (Ramamoorthy et al., 2021).
Additionally, quercetin also decreases the expression of all ve
proteins (presenilin1, presenilin2, Nicastrin, APH1, PEN2) of the
gamma-secretase complex in colorectal cancer (Li et al., 2020).
Combining quercetin with a gamma-secretase inhibitor
(Okuhashi et al., 2011) or ionizing radiation (Liao et al., 2023)
could enhance the overall anti-tumor efcacy, implying that
quercetin may be used to reduce the toxicity of GSIs.
3.2.2 Notch transcription complex inhibitors
The interaction between NICD and CSL plays a crucial role in
determining the activation or deactivation of downstream genes in
the nucleus within the Notch signaling pathway. Phytochemicals
inhibit the binding of CSL and NICD, as well as the expression of co-
activators, while boosting the expression of co-repressors, resulting
in antitumor effects. For example, silybin suppresses the Notch
signaling system by activating the apoptotic pathway, leading to the
inhibition of NICD activity in human cancer cells. Silybin reduces
the expression of intracellular domain of Notch1 (N1ICD) and RBPJ
(recombination signal-binding protein of immunoglobulin kappa J
region) activity in hepatocellular carcinoma with a CSL-dependent
manner (Zhang et al., 2013), suggesting that blocking CSL and
N1ICD interactions is a good idea in tumor therapy. Resveratrol
FIGURE 5
An illustrative depiction of the phytochemicals could inuence the interaction between the Notch and PI3K/AKT/mTOR signaling pathways.
Frontiers in Pharmacology frontiersin.org07
Yang et al. 10.3389/fphar.2024.1412669
TABLE 2 Phytochemicals on Notch and other pathways crosstalk.
Phytochemical Cancer type Cross talk Reference
EGCG Neuroblastoma Notch, AKT, VEGF Hossain et al. (2012)
Liver cancer Notch, Hedgehog, NF-κBAshry et al. (2022)
Resveratrol Cervical cancer Notch, STAT3, Wnt Zhang et al. (2014)
Ovarian cancer Notch, STAT3, Wnt Zhong et al. (2015)
T-ALL Notch, PI3K/AKT Cecchinato et al. (2007)
Ovarian cancer Notch, PTEN, AKT Kim et al. (2019)
Emodin Glioma stem cells Notch, Wnt, STAT3 Kim et al. (2015)
Glioma stem cells EGFR, Notch, Wnt, STAT3 Kim et al. (2018)
Quercetin Breast cancer Notch, PI3K/AKT Cao et al. (2018)
Breast cancer Notch, Death-associated factor 6 Takebe et al. (2014)
Histiocytic lymphoma Notch, AKT, mTOR, VEGF Chen et al. (2016b)
Curcumin Oral cancer Notch, NF-κBLiao et al. (2011)
Lung cancer Notch, HIF1α, VEGF, NF-κBLi et al. (2018)
Pancreatic cancer Notch, Hippo Zhou et al. (2016)
Triptolide Brain cancer Notch, PI3K/AKT Zhang et al. (2018)
Triptonide Gastric cancer Notch, NF-κBXiang et al. (2020)
Trichostatin A Gastric cancer Notch, NF-κBYao et al. (2012)
Genistein Breast cancer Notch, Death-associated factor 6 Peiffer et al. (2020)
Pancreatic cancer Notch, NF-κBWang et al. (2006c)
Colon cancer Notch, NF-κB, Ecadherin Zhou et al. (2017)
Pancreatic cancer Notch, NF-κBWang et al. (2006b)
Prostate cancer Notch, AKT, foxm1 Wang et al. (2011a)
Breast cancer Notch, NF-κBPan et al. (2012)
Pancreatic cancer Notch, EMT Wang et al. (2009)
Pancreatic cancer Notch, Cancer Stem Cells Wang et al. (2011b)
Silybin Breast cancer Notch, AKT, ERK Kim et al. (2014)
Berberine Gastric cancer Notch, MAPK, NF-κBWang et al. (2021)
Withaferin A T-ALL Notch, eIF2A Sanchez-Martin et al. (2017)
Colon cancer Notch, JNK Koduru et al. (2010)
Colon cancer Notch, AKT, Bcl-2 Koduru et al. (2009)
Ovarian cancer Notch, AKT, Bcl-2 Zhang et al. (2012)
Physciosporin Colorectal cancer Notch, Hedgehog Yang et al. (2019)
(-)-Gossypol Glioma stem cells Notch, Hedgehog Linder et al. (2019)
Cordycepin Breast cancer Notch, Hedgehog Liu et al. (2020)
Esc-3 Ovarian cancer Notch, Wnt Fu et al. (2017)
8,12-Dimethoxysanguinarine Breast cancer Notch, NF-κB, PI3K/AKT, Wnt Yang et al. (2023)
Okadaic Acid Breast cancer Notch, PI3K/AKT, NF-κBLi et al. (2016)
Adriamycin Or Paclitaxel Breast cancer Notch, VEGF Zhang et al. (2016)
(Continued on following page)
Frontiers in Pharmacology frontiersin.org08
Yang et al. 10.3389/fphar.2024.1412669
alters the DNA methylation patterns in human breast cancer cells
and inhibits the cancer-causing Notch signaling by controlling the
co-activator (MAML2) transcriptional activity through epigenetic
processes (Lubecka et al., 2016). Similarly, resveratrol also inhibits
the co-activator protein ASCL1 in carcinoid tumors (Pinchot et al.,
2011). Resveratrol could enhance the recruitment of the co-
repressor (HDAC1) in human glioma cells (Yang et al., 2016).
Therefore, it is crucial to limit the activation of co-activators or
promote the activation of co-repressors in tumor cells to avoid the
development of cancer.
3.2.3 Notch downstream target genes inhibitors
The ultimate stage in Notch signaling is the transcription of
downstream target genes. The Hes and Hey protein families are the
most extensively studied targets of Notch (Guo et al., 2011).
Hes1 has been demonstrated to promote the activation of the
Notch signaling pathway, thereby promoting the proliferation
and inhibiting the apoptosis. Natural products, such as oleandrin
and cowanin, have been shown to depress the expression of Hes1/5,
thus controlling the progression of T-ALL (Arai et al., 2018a;Arai
et al., 2018b). Several phytochemicals have shown potential in
treating cancer by inuencing the activity of specic genes
downstream of the Notch signaling pathway, as indicated in Table 1.
3.3 Crosstalk between Notch and other
oncogenic pathways
In human cancers, Notch signaling plays multiple roles in
different tissues, acting as both a tumor suppressor and a tumor
promoter. Activation of the Notch signaling pathway upregulates
multiple factors, which subsequently transmit bidirectional signals
among cancerous, stromal, and endothelial cells (Rizzo et al., 2008;
Yamamoto et al., 2014). Thus, it is expected that Notch signaling
intersects with multiple oncogenic signaling pathways, including
Wnt and Hedgehog signaling, growth factors like epidermal growth
factor receptor (EGFR), transforming growth factor type beta (TGF-
β), and vascular endothelial growth factor (VEGF) oncogenic
kinases, and transcription factors like Nuclear factor kappa-B
(NF-κB), PI3K (phosphatidylinositol 3-kinase)/AKT (protein
kinase B)/mTOR (mammalian target of rapamycin) (Guo et al.,
2011). The crosstalk between Notch and other oncogenic signaling
pathways is crucial for various cellular processes such as cell
proliferation, migration, invasion, metastasis, angiogenesis, and
the self-renewal of cancer stem cells, suggesting that Notch
signaling could be a promising approach for cancer therapy.
Here, we have attempted to summarize how natural products
regulate the complexity of cellular responses due to the crosstalk
between signaling pathways.
3.3.1 Interacting with developmental
signaling pathways
During embryogenesis, the Notch pathway interacts with other
developmental pathways, including Wnt and Hedgehog signaling
pathways, which operate in coordination across various types of
cancer (Guo et al., 2011). Interaction among Notch, Hedgehog, and
Wnt signaling pathways involved in regulating self-renewal,
proliferation, and differentiation, ensuring correct organogenesis
(Cerdan and Bhatia, 2010). Therefore, comprehensive knowledge of
the interactive functions of these pathways in cancer may provide
novel options for cancer treatment as shown in Figure 3.
The Wnt signaling pathway is a major developmental pathway
that determines cell proliferation, differentiation, tissue
homeostasis, and epithelial-mesenchymal interactions during
embryogenesis (Arai et al., 2018b;Manoranjan et al., 2020). The
pathway comprises two major categories: the canonical Wnt
pathway, which involves key elements such as Wnt, β-catenin,
and TCF/LEF (T-cell factor/lymphoid enhancer factor)
transcription factors, and the non-canonical Wnt-calcium
pathway, which regulates intracellular calcium levels and
cytoskeleton of cells (Vlashi et al., 2023). Therefore, the
suppression of Wnt activity has received extensive attention in
the study of cancer cells, presenting an opportunity for cancer
treatment (Olson et al., 2006;Seifert and Mlodzik, 2007).
A recent study revealed the expression of Notch ligands
inhibited the transformation of human mammary epithelial cells
induced by Wnt1, suggesting that the involvement of Notch-Wnt
communication in breast tumorigenesis (Ayyanan et al., 2006). ESC-
3, a novel cytotoxic compound, induces apoptosis via inhibiting both
Notch and Wnt/β-catenin pathways, resulting in a reduction in
tumor growth in an ovarian cancer xenograft model (Fu et al., 2017).
Emodin also hinders the Wnt pathway by reducing the level of active
β-catenin in human glioma stem cells (Kim et al., 2018).
β-catenin has been shown to activate Notch signaling by
upregulating the expression of the Notch ligand Jagged1 (Rodilla
et al., 2009;Kode et al., 2014). The efcacy of Bruceine D, which is
derived from the Chinese herb Brucea javanica (L.) Merr, to inhibit
Jagged1 was found to be synergistic following β-catenin knockdown
and reversed following overexpression (Cheng et al., 2017).
However, the levels of β-catenin remained unchanged in
Jagged1 knockdown cells (Cheng et al., 2017). It is reasonable to
assume that the tumorigenesis mediated by β-catenin could be
blocked by Notch inhibitors, such as resveratrol, 8,12-
dimethoxysanguinarine, and emodin. Conversely, the activated
Wnt signal boosts the accumulation of β-catenin in the nucleus.
Subsequently, specic target genes undergo transcriptional
activation, leading to the development of cancer (MacDonald
et al., 2009). Resveratrol inhibited the proliferation of ovarian
cancer cells by reducing the expression of β-catenin and Hes1,
TABLE 2 (Continued) Phytochemicals on Notch and other pathways crosstalk.
Phytochemical Cancer type Cross talk Reference
Nimbolide Oral cancer Notch, VEGF, MMP Kowshik et al. (2017)
Artemisinin Breast cancer Notch, VEGF, HIF1αDong et al. (2020)
Z-Ajoene Glioblastoma multiforme Notch, AKT, TGFβJung et al. (2014)
Frontiers in Pharmacology frontiersin.org09
Yang et al. 10.3389/fphar.2024.1412669
suggesting the simultaneous suppression of the biological functions
of Notch and Wnt signaling (Zhong et al., 2015). In summary, the
regulation of β-catenin is vital in the crosstalk between Wnt and
Notch signaling.
The Hedgehog is a developmental signaling pathway that plays
an essential role in embryogenesis, tissue polarity, tissue
regeneration, and carcinogenesis (Lum and Beachy, 2004;Li
et al., 2012). The Hedgehog pathway consists of several key
components, namely, Hedgehog, Patched receptor (PTCH),
Smoothened (SMO), cytosolic intermediates like Suppressor of
Fused (SUFU) and Costal 2 (COS2), as well as inhibitors such as
Protein Kinase A (PKA) and Supernumerary Limbs (SLIMB).
Additionally, the pathway involves the transcription factors
Cubitus interruptus/Zinc Finger protein -Glioma-associated
oncogene homolog1 (Ci/Gli) (Cortes et al., 2019). Hedgehog has
three ligands, Sonic hedgehog, which is critical for neuronal
development, Indian hedgehog, which is critical for skeletal
development, and Desert hedgehog, which is critical for gonadal
development (Lee et al., 2016).
According to the literature, Hedgehog is controlled by the Notch
signaling pathway, mainly through the downstream effector of
Notch that controls the transport of Hedgehog component and
Gli levels (Xia et al., 2022). Consequently, Notch target genes play a
central role in controlling Gli gene transcription (Jacobs and Huang,
2019). Physciosporin, derived from P. granulata, decreased the
transcriptional activity of the Gli, Hes1 and CSL (Yang et al.,
2019). Additionally, it markedly reduced the formation of
spheroids in human CSC221 cells that overexpressed Gli1/2 or
Delta EN1 (a membrane-bound and S2-cleaved type of human
Notch1). Nevertheless, it did not decrease the formation of
spheroids in cells that overexpressed both Gli1/2 and Delta EN1,
proposing that physciosporin can downregulate the cancer stemness
of human colon cells by controlling the Sonic Hedgehog and Notch
signaling pathways (Yang et al., 2019). Additionally, NUMB
endocytic adaptor protein (NUMB) negatively regulates Notch
signaling by binding directly to NICD, preventing NICD from
initiating gene transcription (Flores et al., 2014). NUMB is also a
suppressor of Hedgehog signaling and specically targets Gli1 for
ubiquitination through the action of Itch (Di Marcotullio et al.,
2006). Quercetin decreases tumor growth by inhibiting the
activation of NUMB and Hedgehog signaling pathway (Salama
et al., 2019), suggesting that NUMB has potential as a biomarker
for cancer.
Additionally, modulation of the Notch pathway by the
Hedgehog signaling pathway, mainly through inhibition of
Hedgehog downstream effectors, such as Gli proteins and Hes1
(Wall et al., 2009;Dave et al., 2011). Hedgehog signaling may induce
Hes1 expression in Hep2 cells (human), however, this effect can be
reversed by EGCG, which acts as an inhibitor for NICD (Ashry et al.,
2022). Therefore, the Hedgehog signaling pathway may have an
impact on the activity of NICD, leading to the promotion of
Hes1 production. Cordycepin, also known as 3-deoxyadenosine,
encompasses anti-inammatory, antioxidant, and anti-cancer
activities (Tuli et al., 2014). Cordycepin suppressed the
transcriptional activity of Gli, which in turn reduced the
expression of Notch1, Notch3, Jagged1 and Hes1 in human
triple-negative breast cancer cells (MDA-MB-231 cell line) (Liu
et al., 2020). Notably, knocking out of Gli obstructed cordycepin-
induced inuences on the apoptotic, EMT, and Notch pathways,
demonstrating that the regulation of Notch by cordycepin is
dependent on Gli in breast cancer (Liu et al., 2020). Evidence
indicates that the Notch ligand, Jagged 2 is induced by Hedgehog
signaling during carcinogenesis (Katoh, 2007). For example,
cyclopamine inhibits the expression of Notch1, Notch2, Notch3,
Jagged2, and DLL1 correlated with the downregulation of the sonic
hedgehog odontogenic keratocytes (Ren et al., 2012). In summary,
the regulation of processing and nuclear translocating of Gli or
NUMB is vital in the crosstalk between Hedgehog and
Notch signaling.
3.3.2 Interacting with growth factors
Numerous growth factors exert a variety of actions in cancers.
For example, the oncogene gene mutations, amplication or
overexpression of HER2/Neu (ErbB2), platelet-derived growth
factor (PDGF), EGFR and TGF-β(Wang et al., 2010). Growth
factors pathways are associated with the progression of cancers,
including proliferation, invasion, and tumor growth. Not
surprisingly, the interaction between growth factors and the
Notch signaling pathway is frequently observed in several
cancers. A diagram illustrating the interplay among different
genes of Notch, EGFR, PDGF, TGF-β, and VEGF is presented
in Figure 4.
The role of the Notch-EGFR crosstalk has been discovered in
various types of cancer, including breast (Baker et al., 2014), lung
(Konishi et al., 2007), brain (Purow et al., 2008) cancer. Recently,
researchers have continued to explain crosstalk between Notch and
EGFR to dissect the mechanisms for a better understanding of how
cancer cells apply the Notch pathway to counteract the inhibitory
effects of EGFR targeting. Notch signaling could regulate EGFR
activity via regulating the nuclear translocation of the
Notch1 intracellular domain. For example, dioscin markedly
enhanced the expression of Notch1 and Jagged1, and enhanced
the activity of gamma-secretase, leading to EGFR, VEGF, and
Notch-dependent target genes (Xu et al., 2018). Conversely, the
decrease in activity of Notch3 greatly inhibited the growth and
stimulated the programmed cell death of the human ErbB2-negative
tumor cell lines (Yamaguchi et al., 2008). On the other hand, EGFR
signaling may also regulate the Notch pathway. Emodin inhibited
the growth of human glioma stem cells through the induction of
proteasomal degradation of EGFR/EGFRvIII, which subsequently
hindered the activation of stemness signaling pathways, specically
the Notch pathway (Kim et al., 2018).
PDGF, which is produced in carcinomas, primarily affects the
non-epithelial tumor stroma, and stimulates angiogenesis
(Papadopoulos and Lennartsson, 2018;Li et al., 2022). The
available literature indicates that the PDGF receptor is a novel
Notch target gene (Wu et al., 2021;Vimalraj, 2022). PDGF-D is
a vital factor in the aggressiveness of breast tumors, which is causally
associated with the activation of Notch1 (Ahmad et al., 2011).
Furthermore, the downregulation of PDGF-D results in the
inactivation of the Notch1/Twist1 axis, potentially reversing
epithelial-mesenchymal transition and inhibiting the progression
of colorectal cancer (Chen J. H. et al., 2017). Unfortunately, there are
currently no documented ndings on the identication of natural
compounds that modulate the communication between PDGF-D
and Notch signaling in cancer. Nevertheless, studies have
Frontiers in Pharmacology frontiersin.org10
Yang et al. 10.3389/fphar.2024.1412669
demonstrated that lycopene, a naturally occurring carotenoid found
in tomatoes, can effectively suppress the growth of certain cancer
cells (Wu et al., 2007). Additionally, it has been observed that
lycopene directly binds to PDGF-BB, indicating that its ability to
inhibit PDGF may play a role in its anti-tumor properties (Wu
et al., 2007).
TGF-β, a multifunctional cytokine, plays a key role in the cancer
pathogenesis involved in cell growth, differentiation, adhesion, and
apoptosis (Gu and Feng, 2018). The diverse roles of TGF-βin cancer
are inuenced by its interactions with several signaling pathways,
such as Hedgehog, Wnt, PI3K/AKT, Notch, and RAS-ERK
(extracellular signal-regulated kinase). Heyl, a downstream
effector of the Notch signaling pathway, inhibits the action of
TGF-βby interacting with activated Smads (Han et al., 2014),
providing novel strategies and perspectives for treating breast
cancer. Moreover, Z-ajoene, which is derived from garlic, has
demonstrated a variety of biological activities, such as anti-
proliferative, antioxidant, and antitumor (Naznin et al., 2010;
Jung et al., 2014). Z-ajoene reduced levels of Notch target genes,
and TGF-βis the key mediator of the Z-ajoene effect on glioblastoma
multiforme cancer stem cells (Jung et al., 2014). Furthermore, the
TGF-βpathway is critical in maintaining stem cell properties in
cancer cells (Gurska et al., 2019).
VEGF, a signicant angiogenic factor in both normal and
abnormal blood vessel formation, is frequently overexpressed in
human tumors, leading to the malignancy progression across
several tumors and low survival rates of patients (Yang and
Cao, 2022;Ghalehbandi et al., 2023). Both the VEGF and
Notch signaling pathways have been indicated to be key
regulators in developmental and pathological angiogenesis,
including in cancers (Li and Harris, 2009;Blanco and Gerhardt,
2013). VEGF modulates the expression of Notch signaling
components, specically by upregulating the expression of
DLL4 and Notch receptors, which then promotes the activation
of Notch signaling (Patel et al., 2005;Hainaud et al., 2006;Ridgway
et al., 2006). The combination of survivin shRNA and EGCG
markedly decreased angiogenic (VEGF and b-FGF) factors and
then inhibited Notch1 expression in neuroblastoma, reasonably
inferring that blocking VEGF may enhance the sensitivity of
tumors to anti-Notch therapy (Hossain et al., 2012). Nimbolide,
obtained from neem leaves and owers, inhibited the activity of
MMP and blocked Notch and VEGF signaling pathways by
targeting miR-21 (Kowshik et al., 2017). Curcumin also could
decrease the tumor weight and size via downregulating the
expression of Notch, HIF-1, VEGF, and NF-κB(Li et al., 2018).
Although VEGF induces Notch signaling, expression of VEGF
ligands and receptors appear to be also regulated by Notch signaling.
For example, quercetin decreased Notch1 expression and then
suppressed the expression of angiogenesis-associated proteins
hypoxia-inducible factor alpha (HIF1α) and VEGF in histiocytic
lymphoma (Chen X. et al., 2016). In breast cancer, artemisinin, an
anti-malarial active compound derived from the sweet wormwood
plant, downregulates the expression of Notch1, DLL4, and Jagged1,
reducing VEGF and HIF-1αlevels (Dong et al., 2020). Providing a
feedback mechanism, another study has found that the inhibition of
Notch signaling or the suppression of Notch4/DLL3 leads to a
decrease in endothelial markers and the function of tumor-
derived endothelial cells following treatment with adriamycin or
paclitaxel via VEGF receptor (Zhang et al., 2016). These data provide
new prospects for the antiangiogenic therapy of human cancer.
3.3.3 Interacting with oncogenic kinases and
transcription factors
The NF-κB pathway comprises two major categories: the
canonical IKK pathway which is dependent on IκB proteins
through an individual kinase signalosome multiprotein complex,
while the non-canonical pathway is triggered by ligands like CD40L
and lymphotoxin (Dolcet et al., 2005). NF-κB activation is evident in
several cancer types and is also associated with the initiation of
tumor angiogenesis (Sarkar and Li, 2008;Prasad et al., 2010). Many
publications have described how natural products can regulate the
NF-κB pathway through Notch, and vice versa, by various context-
dependent mechanisms (Rajasinghe and Gupta, 2017;Ferrandino
et al., 2018;Hossain et al., 2018;Grazioli et al., 2020). Firstly,
evidence shows that Notch serves as a crucial regulator of NF-κB
at an upstream level and regulates NF-κB pathway members (Mishra
et al., 2021). RBPJ functions as a potent transcriptional inhibitor of
p100/p52, but its inhibitory effects can be reversed by activated
Notch1, implying that p100/p52 is regulated by Notch signaling and
can be considered a target gene of Notch (Oswald et al., 1998).
Triptonide, a natural small molecule extracted from Tripterygium
wilfordii Hook F,efciently inhibits tumor growth and metastasis
via decreasing the levels of Notch1 downstream proteins RBPJ, IKK
alpha, IKK beta, reducing p52 phosphorylation (Xiang et al., 2020).
Notch1 co-localizes with IKK at NF-κB-responsive promoter sites,
enhancing IκB kinase activity and thereby maintaining NF-κB
activity (Song et al., 2008). And inactivation of NF-κB DNA-
binding activity by genistein was partly impeded by the
overexpression of Notch1 in cDNA-transfected BxPC-3 cells
(Wang et al., 2006b). Genistein also inhibits tumor growth in
pancreatic (Wang et al., 2006b), colon (Zhou et al., 2017), and
triple-negative breast cancers (Pan et al., 2012) by inhibiting NF-κB
activity via the Notch1 pathway. Curcumin downregulates
Notch1 expression, blocks Notch1 activation, and inactivates NF-
κB DNA-binding activity (Gupta et al., 2017).
Secondly, NF-κB subunits regulate the transcription of Notch
family members. This nding is supported by Wang et al. (2001)
which revealed that the N-terminal region of Notch1 NICD
specically associated with the p50 subunit and prevented it
from binding to DNA in NTera-2 cells. Lastly, not all cancer
cells exhibit a close association between the activation of the NF-
κB pathway and the Notch pathway. For example, trichostatin A,
derived from streptomyces, caused gastric cancer cells growth
arrest and apoptosis by controlling NF-κBandp21
WAF1/CIP1
,
without the involvement of the Notch pathway (Yao et al.,
2012). Another is berberine, which inhibits the Notch, MAPK,
and NF-κB signaling pathways by regulating circRNA (Wang
et al., 2021). Sentrin-specic protease-2 (SENP2) could act as a
tumor suppressor in CLL cells via suppressing the Notch and NF-
κBsignalingpathways(Chen et al., 2019). Nevertheless, there is
currently no natural products that regulates the activity of
SENP2 in the therapy of hematologic tumors. Betanidin,
obtained from red beets, could directly bind to SENP2 and
may be used as a treatment for CLL cells (Taghvaei et al.,
2022). These results suggest novel understandings of the
crosstalk between Notch and NF-κB.
Frontiers in Pharmacology frontiersin.org11
Yang et al. 10.3389/fphar.2024.1412669
Thirdly, in cancer biology, as mentioned above, the Notch and
NF-κB pathways promote cancer progression by regulating each
others activities. The Notch-NF-κB network are typically involved
in regulation of inammatory disease (Liubomirski and Ben-Baruch,
2020). Nevertheless, the growth of tumor and its reaction to
treatment are controlled by inammation, which can either
stimulate or inhibit the advancement of tumors, potentially
leading to contrasting impacts on the effectiveness of therapy
(Zhao et al., 2021). Thus, an optimal approach is evidently
necessary to address the interaction between the Notch and NF-
κB pathways to devise innovative treatments for the treatment of
inammatory illnesses. Utilizing small molecule inhibitors that
specically target NF-κB, which is the downstream of Notch, and
not necessarily the Notch blockers, may serve as an effective
therapeutic method to disturb the interplay and reduce
inammation. Many of the transformational events in cancers are
the consequence of amplied signaling within the PI3K/AKT
pathway (Hynes and Stoelzle, 2009;Dillon and Muller, 2010).
The PI3K/AKT/mTOR pathway is crucial in regulating multiple
cellular activities, such as growth, proliferation, metabolism,
motility, migration, invasion, angiogenesis, survival, and
autophagy (McAuliffe et al., 2010). And mTOR is a crucial
protein kinase that frequently acts as a downstream effector of
the PI3K/AKT signaling pathway in various cancer cell types
(Carino et al., 2008). mTOR can also phosphorylate AKT
(Saxton and Sabatini, 2017). The PI3K/AKT pathway plays a
crucial role in promoting EMT during the development of cancer
(Sabbah et al., 2008). Crosstalk between PI3K/AKT and Notch
pathways has been described in prostate cancer (Wang et al.,
2011a), T-ALL (Cecchinato et al., 2007), colon cancer (Koduru
et al., 2009), brain cancer (Xiang et al., 2020) as well as breast cancer
(Cao et al., 2018). Kim et al. (2014) reported that overexpression of
Notch1 reversed the suppression of ERK and AKT phosphorylation
caused by silybin. Additional reports also demonstrated that inhibit
Notch1 activation by Withaferin A, which resulted in the
downregulation of pAKT and Bcl-2 expression (Koduru et al.,
2009). Kim et al. (2019) found that induces cell death through
ROS-dependent downregulation of Notch1, which negatively
controlled the expression of PTEN (Phosphatase and tensin
homolog) and AKT signaling. Curcumin, an inhibitor of Notch,
increases PTEN expression and decreases AKT phosphorylation in
chronic myelogenous leukemia (Chen et al., 2015). Notch inhibits
the dephosphorylation of PI3K/AKT by blocking the activation of
PP2A (Protein phosphatase 2) and PTEN, leading to the promotion
of cancers malignant progression (Li et al., 2016). The
downregulation of Notch1 activity by okadaic acid, a PP2A
inhibitor, decreases low cell invasion in breast cancer via
inactivating the AKT, mTOR, and NF-κB signaling pathways (Li
et al., 2016). Moreover, through the downregulation of Notch1/
PTEN/AKT signaling, resveratrol causes the demise of ovarian
cancer cells (Kim et al., 2019).
AKT serves as a regulator of Notch signaling. On the one hand,
Notch1 downregulation is linked to AKT in the induction of cell
growth inhibition and death by genistein in prostate cancer (Wang
et al., 2011a). On the other hand, resveratrol suppresses Notch
signaling in T-ALL cells, resulting in a reduction in AKT activity and
modulating the operation of interacting signaling systems
(Cecchinato et al., 2007). Therefore, strategies aimed at
modulation of Notch-PI3K/AKT/mTOR signaling have the
potential to enable therapeutic intervention. A schematic diagram
depicting the interactions among the Notch, PI3K/AKT/mTOR, and
NF-κB as shown in Figure 5.
3.3.4 Others
In addition, many connections between the Notch pathway and
other signaling pathways such as DXX6, ERK, and Hippo signaling
may also contribute to the complexity of tumor angiogenesis and the
challenge of Notch as a promising target.
The regulation of Notch-related pathways is also irregular with
bioactive natural compounds. Regarding breast cancer, the
repression of Notch4 protein and its downstream targets by
quercetin appears to be independent of death-associated factor 6,
whereas genistein is known to repress Notch signaling solely
through the presence of death-associated factor 6 (Peiffer et al.,
2020). Hippo signaling compounds could regulate Notch pathways.
In pancreatic cancer, curcumin reduces the expression of Yes-
associated protein and transcriptional coactivator with PDZ-
binding motif, two paralogous proteins belonging to the Hippo
signaling pathway, which results in the subsequent suppression of
Notch1 expression (Zhou et al., 2016). Kim et al. (2014) reported
that overexpression of Notch1 prevented silybin-induced inhibition
of ERK and AKT phosphorylation. We summarized the cross talk
between Notch and other signaling pathways in different cancers
with phytochemicals in Table 2.
4 Conclusion and perspective
Recently, there has been an increasing attraction in developing
clinically potent antagonists of the Notch signaling pathway.
Antitumor activity has been observed from gamma-secretase
inhibitors and monoclonal antibodies administered as single
agents in early clinical trials (Takebe et al., 2014). However,
achieving Notch-directed therapeutics remains an unattained
goal. Currently, no compounds, including chemicals and
phytochemicals, that interfere with Notch signaling have been
approved for using in cancer patients. Additionally, it is
imperative to develop competent tools and precise research
techniques to understand the heterogeneity and complexity of the
Notch signaling pathway in cancer.
The research summarized above has shown that Notch
activation and interactions with other oncogenic pathways
strongly promote the malignant phenotype, in vitro and in vivo.
To some extent, natural bioactive compounds can inhibit Notch
activation and cross talk with other oncogenic pathways without any
signicant side effects. Further research is needed to elucidate the
signicance and mechanisms of Notch pathway activation in cancer,
and to determine whether Notch-based therapies in combination
with chemotherapy or other biologically targeted drugs could
enhance clinical anti-tumor efcacy. And whether there is a need
to discover more sophisticated and potent Notch inhibitors.
Targeting Notch is critical because the pathway communicates
extensively with other signaling pathways, such as Wnt,
Hedgehog signaling, growth factors such as EGFR, TGF-βand
VEGF, oncogenic kinases, and transcription factors such as NF-
κB, PI3K/AKT/mTOR. Therefore, the multi-target properties of
Frontiers in Pharmacology frontiersin.org12
Yang et al. 10.3389/fphar.2024.1412669
natural products and drugs designed to inhibit Notch oncogenic
signaling crosstalk may have potential for therapeutic intervention.
In addition, an important future direction for Notch targeting is
to ascertain the functions of the Notch pathway in various cancer
cells and to develop a biomarker for cancer and stromal sensitivity.
Author contributions
JY: Conceptualization, Writingoriginal draft, Writingreview
and editing. QS: Writingreview and editing. XL: Writingreview
and editing. YY: Methodology, Writingreview and editing. RR:
Funding acquisition, Investigation, Writingreview and editing. PY:
Writingreview and editing. YX: Conceptualization, Supervision,
Writingreview and editing.
Funding
The authors declare that nancial support was received for the
research, authorship, and/or publication of this article. This work
was supported by National Natural Science Foundation of China
(No. 82274397); Natural Science Foundation of Shandong Province
(No. ZR2021LZY012); Opening project of shanghai key laboratory
of compound Chinese medicines (No. 21DZ2270500).
Acknowledgments
Figures in this review were created by BioRender.
Conict of interest
The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could be
construed as a potential conict of interest.
Publishers note
All claims expressed in this article are solely those of the authors
and do not necessarily represent those of their afliated organizations,
or those of the publisher, the editors and the reviewers. Any product
that may be evaluated in this article, or claim that may be made by its
manufacturer, is not guaranteed or endorsed by the publisher.
References
Ahmad, A., Wang, Z. W., Kong, D. J., Ali, R., Ali, S., Banerjee, S., et al. (2011). Platelet-
derived growth factor-D contributes to aggressiveness of breast cancer cells by up-
regulating Notch and NF-κB signaling pathways. Breast Cancer Res. Treat. 126 (1),
1525. doi:10.1007/s10549-010-0883-2
Alderuccio, J. P., and Lossos, I. S. (2022). NOTCH signaling in the pathogenesis of
splenic marginal zone lymphoma-opportunities for therapy. Leukemia Lymphoma 63
(2), 279290. doi:10.1080/10428194.2021.1984452
Arai, M. A., Akamine, R., Hayashi, N., Koyano, T., Kowithayakorn, T., and Ishibashi,
M. (2018a). The notch inhibitors isolated from Nerium indicum.J. Nat. Prod. 81 (5),
12351240. doi:10.1021/acs.jnatprod.7b01031
Arai, M. A., Akamine, R., Tsuchiya, A., Yoneyama, T., Koyano, T., Kowithayakorn, T.,
et al. (2018b). The Notch inhibitor cowanin accelerates nicastrin degradation. Sci. Rep.
8, 5376. doi:10.1038/s41598-018-23698-4
Ashry, R., Elhussiny, M., Abdellatif, H., Elkashty, O., Abdel-Ghaffar, H. A., Gaballa, E.
T., et al. (2022). Genetic interpretation of the impacts of Honokiol and EGCG on
apoptotic and self-renewal pat sways in HEp-2 human laryngeal CD44
high
cancer stem
cells, Nutrition and Cancer-an. Int. J. 74 (6), 21522173. doi:10.1080/01635581.2021.
1981404
Ayyanan, A., Civenni, G., Ciarloni, L., Morel, C., Mueller, N., Lefort, K., et al. (2006).
Increased Wnt signaling triggers oncogenic conversion of human breast epithelial cells
by a Notch-dependent mechanism. Proc. Natl. Acad. Sci. U. S. A. 103 (10), 37993804.
doi:10.1073/pnas.0600065103
Baker, A. T., Zlobin, A., and Osipo, C. (2014). Notch- EGFR/HER2 bidirectional
crosstalk in breast cancer. Front. Oncol. 4, 360. doi:10.3389/fonc.2014.00360
Blanco, R., and Gerhardt, H. (2013). VEGF and Notch in tip and stalk cell selection.
Cold Spring Harb. Perspect. Med. 3 (1), a006569. doi:10.1101/cshperspect.a006569
Bounaama, A., Djerdjouri, B., Laroche-Clary, A., Le Morvan, V., and Robert, J.
(2012). Short curcumin treatment modulates oxidative stress, arginase activity,
aberrant crypt foci, and TGF-β1 and HES-1 transcripts in 1,2-dimethylhydrazine-
colon carcinogenesis in mice. Toxicology 302 (2-3), 308317. doi:10.1016/j.tox.2012.
08.014
Bray, F., Laversanne, M., Weiderpass, E., and Soerjomataram, I. (2021). The ever-
increasing importance of cancer as a leading cause of premature death worldwide.
Cancer 127 (16), 30293030. doi:10.1002/cncr.33587
Cao, L. B., Yang, Y. X., Ye, Z. Y., Lin, B. H., Zeng, J. C., Li, C. H., et al. (2018).
Quercetin-3-methyl ether suppresses human breast cancer stem cell formation by
inhibiting the Notch1 and PI3K/Akt signaling pathways. Int. J. Mol. Med. 42 (3),
16251636. doi:10.3892/ijmm.2018.3741
Carino, C., Olawaiye, A. B., Cherls, S., Serikawa, T., Lynch, M. P., Rueda, B. R., et al.
(2008). Leptin regulation of proangiogenic molecules in benign and cancerous
endometrial cells. Int. J. Cancer 123 (12), 27822790. doi:10.1002/ijc.23887
Cecchinato, V., Chiaramonte, R., Nizzardo, M., Cristofaro, B., Basile, A., Sherbet, G.
V., et al. (2007). Resveratrol-induced apoptosis in human T-cell acute lymphoblastic
leukaemia MOLT-4 cells. Biochem. Pharmacol. 74 (11), 15681574. doi:10.1016/j.bcp.
2007.08.001
Cerdan, C., and Bhatia, M. (2010). Novel roles for Notch, Wnt and Hedgehog in
hematopoesis derived from human pluripotent stem cells. Int. J. Dev. Biol. 54 (6-7),
955963. doi:10.1387/ijdb.103067cc
Chen, G. P., Zhang, Y., Xu, Z. Y., Yu, J. F., and Wei, X. (2017a). Curcumin combined
with cis-platinum promote the apoptosis of human colorectal cancer HT29 cells and
mechanism. Int. J. Clin. Exp. Pathology 10 (12), 1149611505.
Chen, J. H., Yuan, W. Z., Wu, L., Tang, Q., Xia, Q. H., Ji, J. T., et al. (2017b ). PDGF-D
promotes cell growth, aggressiveness, angiogenesis and EMT transformation of
colorectal cancer by activation of Notch1/Twist1 pathway. Oncotarget 8 (6),
99619973. doi:10.18632/oncotarget.14283
Chen, J. L., Chang, H., Peng, X. L., Gu, Y. Y., Yi, L., Zhang, Q. Y., et al. (2016a). 3,6-
dihydroxyavone suppresses the epithelial-mesenchymal transition in breast cancer
cells by inhibiting the Notch signaling pathway. Sci. Rep. 6, 28858. doi:10.1038/
srep28858
Chen, J. Z., Xu, T. F., and Chen, C. (2015). The critical roles of miR-21 in anti-cancer
effects of curcumin. Ann. Transl. Med. 3 (21), 330. doi:10.3978/j.issn.2305-5839.2015.
09.20
Chen, X., Dong, X. S., Gao, H. Y., Jiang, Y. F., Jin, Y. L., Chang, Y. Y., et al. (2016b).
Suppression of HSP27 increases the anti-tumor effects of quercetin in human leukemia
U937 cells. Mol. Med. Rep. 13 (1), 689696. doi:10.3892/mmr.2015.4600
Chen, X. L., Wang, S. F., Liang, X. T., Liang, H. X., Wang, T. T., Wu, S. Q., et al. (2019).
SENP2 exerts an anti-tumor effect on chronic lymphocytic leukemia cells through the
inhibition of the Notch and NF-B signaling pathways. Int. J. Oncol. 54 (2), 455466.
doi:10.3892/ijo.2018.4635
Cheng, Z. Y., Yuan, X., Qu, Y., Li, X., Wu, G. Z., Li, C. W., et al. (2017). Bruceine D
inhibits hepatocellular carcinoma growth by targeting β-catenin/jagged1 pathways.
Cancer Lett. 403, 195205. doi:10.1016/j.canlet.2017.06.014
Cortes, J. E., Gutzmer, R., Kieran, M. W., and Solomon, J. A. (2019). Hedgehog
signaling inhibitors in solid and hematological cancers. Cancer Treat. Rev. 76, 4150.
doi:10.1016/j.ctrv.2019.04.005
Dandawate, P., Subramaniam, D., Panovich, P., Standing, D., Krishnamachary, B.,
Kaushik, G., et al. (2020). Cucurbitacin B and I inhibits colon cancer growth by targeting
the Notch signaling pathway. Sci. Rep. 10 (1), 1290. doi:10.1038/s41598-020-57940-9
Dave, R. K., Ellis, T., Toumpas, M. C., Robson, J. P., Julian, E., Adolphe, C., et al.
(2011). Sonic Hedgehog and Notch signaling can cooperate to regulate neurogenic
divisions of neocortical progenitors. Plos One 6 (2), e14680. doi:10.1371/journal.pone.
0014680
Frontiers in Pharmacology frontiersin.org13
Yang et al. 10.3389/fphar.2024.1412669
De Bie, J., Demeyer, S., Alberti-Servera, L., Geerdens, E., Segers, H., Broux, M., et al.
(2018). Single-cell sequencing reveals the origin and the order of mutation acquisition in
T-cell acute lymphoblastic leukemia. Leukemia 32 (6), 13581369. doi:10.1038/s41375-
018-0127-8
Del Papa, B., Baldoni, S., Dorillo, E., De Falco, F., Rompietti, C., Cecchini, D., et al.
(2019). Decreased NOTCH1 activation correlates with response to ibrutinib in chronic
lymphocytic leukemia. Clin. Cancer Res. 25 (24), 75407553. doi:10.1158/1078-0432.
CCR-19-1009
Deng, G., Ju, X., Meng, Q., Yu, Z. J., and Ma, L. B. (2015). Emodin inhibits the
proliferation of PC3 prostate cancer cells in vitro via the Notch signaling pathway. Mol.
Med. Rep. 12 (3), 44274433. doi:10.3892/mmr.2015.3923
Dillon, R. L., and Muller, W. J. (2010). Distinct biological roles for the AKT family in
mammary tumor progression. Cancer Res. 70 (11), 42604264. doi:10.1158/0008-5472.
CAN-10-0266
Di Marcotullio, L., Ferretti, E., Greco, A., De Smaele, E., Po, A., Sico, M. A., et al.
(2006). Numb is a suppressor of Hedgehog signalling and targets Gli1 for Itch-
dependent ubiquitination. Nat. Cell Biol. 8 (12), 14151423. doi:10.1038/ncb1510
Dolcet, X., Llobet, D., Pallares, J., and Matias-Guiu, X. (2005). NF-kB in development
and progression of human cancer. Virchows Arch. 446 (5), 475482. doi:10.1007/
s00428-005-1264-9
Dong, J., Chen, Y. L., Yang, W. H., Zhang, X., and Li, L. (2020). Antitumor and anti-
angiogenic effects of artemisinin on breast tumor xenografts in nude mice. Res.
Veterinary Sci. 129, 6669. doi:10.1016/j.rvsc.2020.01.005
Espinoza, I., and Miele, L. (2013). Notch inhibitors for cancer treatment. Pharmacol.
Ther. 139 (2), 95110. doi:10.1016/j.pharmthera.2013.02.003
Ferrandino, F., Grazioli, P., Bellavia, D., Campese, A. F., Screpanti, I., and Felli, M. P.
(2018). Notch and NF-κB: coach and players of regulatory T-cell response in cancer.
Front. Immunol. 9, 2165. doi:10.3389/mmu.2018.02165
Fiorcari, S., Maffei, R., Atene, C. G., Mesini, N., Maccaferri, M., Leonardi, G., et al.
(2022). Notch2 increases the resistance to venetoclax-induced apoptosis in chronic
lymphocytic leukemia B cells by inducing mcl-1. Front. Oncol. 11, 777587. doi:10.3389/
fonc.2021.777587
Flores, A. N., McDermott, N., Meunier, A., and Marignol, L. (2014). NUMB
inhibition of NOTCH signalling as a therapeutic target in prostate cancer. Nat. Rev.
Urol. 11 (9), 499507. doi:10.1038/nrurol.2014.195
Fu, Q. R., Song, W., Deng, Y. T., Li, H. L., Mao, X. M., Lin, C. L., et al. (2017). ESC-3
induces apoptosis of human ovarian carcinomas through Wnt/β-catenin and Notch
signaling in vitro and in vivo.Int. J. Oncol. 50 (1), 241251. doi:10.3892/ijo.2016.3773
Ghalehbandi, S., Yuzugulen, J., Pranjol, M. Z. I., and Pourgholami, M. H. (2023). The
role of VEGF in cancer-induced angiogenesis and research progress of drugs targeting
VEGF. Eur. J. Pharmacol. 949, 175586. doi:10.1016/j.ejphar.2023.175586
Giordano, F., DAmico, M., Montalto, F. I., Malivindi, R., Chimento, A., Conforti, F.
L., et al. (2023). Cdk4 regulates glioblastoma cell invasion and stemness and is target of a
Notch inhibitor plus resveratrol combined treatment. Int. J. Mol. Sci. 24 (12), 10094.
doi:10.3390/ijms241210094
Giordano, F., Montalto, F. I., Panno, M. L., Andò, S., and De Amicis, F. (2021). A
Notch inhibitor plus Resveratrol induced blockade of autophagy drives glioblastoma cell
death by promoting a switch to apoptosis. Am. J. Cancer Res. 11 (12), 59335950.
Grazioli, P., Orlando, A., Giordano, N., Noce, C., Peruzzi, G., Scafetta, G., et al. (2020).
NF-κB1 regulates immune environment and outcome of Notch-dependent T-Cell acute
lymphoblastic leukemia. Front. Immunol. 11, 541. doi:10.3389/mmu.2020.00541
Gu, S. C., and Feng, X. H. (2018). TGF-βsignaling in cancer. Acta Biochimica
Biophysica Sinica 50 (10), 941949. doi:10.1093/abbs/gmy092
Guo, S. C., Liu, M. L., and Gonzalez-Perez, R. R. (2011). Role of Notch and its
oncogenic signaling crosstalk in breast cancer. Biochimica Biophysica Acta-Reviews
Cancer 1815 (2), 197213. doi:10.1016/j.bbcan.2010.12.002
Gupta,A.P.,Khan,S.,Manzoor,M.M.,Yadav,A.K.,Sharma,G.,Anand,R.,
et al. (2017). Anticancer curcumin: natural analogues and structure-activity
relationship. Stud. Nat. Prod. Chem. 54, 355401. doi:10.1016/b978-0-444-
63929-5.00010-3
Gurska, L. M., Ames, K., and Gritsman, K. (2019). Signaling pathways in leukemic
stem cells. Adv. Exp. Med. Biol. 1143, 139. doi:10.1007/978-981-13-7342-8_1
Hainaud, P., Contrerès, J. O., Villemain, A., Liu, L. X., Plouët, J., Tobelem, G., et al.
(2006). The role of the vascular endothelial growth factor-Delta-like 4 ligand/Notch4-
ephrin B2 cascade in tumor vessel remodeling and endothelial cell functions. Cancer
Res. 66 (17), 85018510. doi:10.1158/0008-5472.CAN-05-4226
Han,L.F.,Diehl,A.,Nguyen,N.K.,Korangath,P.,Teo,W.W.,Cho,S.W.,etal.
(2014). The Notch pathway inhibits TGFβsignaling in breast cancer through
HEYL-mediated crosstalk. Cancer Res. 74 (22), 65096518. doi:10.1158/0008-
5472.CAN-14-0816
Han, Z. F., Zhang, J. Z., Zhang, K., and Zhao, Y. H. (2020). Curcumin inhibits cell
viability, migration, and invasion of thymic carcinoma cells via downregulation of
microRNA-27a. Phytotherapy Res. 34 (7), 16291637. doi:10.1002/ptr.6629
Hossain, F., Sorrentino, C., Ucar, D. A., Peng, Y., Matossian, M., Wyczechowska, D.,
et al. (2018). Notch signaling regulates mitochondrial metabolism and NF-κB activity in
triple-negative breast cancer cells via IKKα-dependent non-canonical pathways. Front.
Oncol. 8, 575. doi:10.3389/fonc.2018.00575
Hossain, M. M., Banik, N. L., and Ray, S. K. (2012). Survivin knockdown increased
anti-cancer effects of (-)-epigallocatechin-3-gallate in human malignant neuroblastoma
SK-N-BE2 and SH-SY5Y cells. Exp. Cell Res. 318 (13), 15971610. doi:10.1016/j.yexcr.
2012.03.033
Howells, L. M., Sale, S., Sriramareddy, S. N., Irving, G. R. B., Jones, D. J. L., Ottley, C. J.,
et al. (2011). Curcumin ameliorates oxaliplatin-induced chemoresistance in
HCT116 colorectal cancer cells in vitro and in vivo.Int. J. Cancer 129 (2), 476486.
doi:10.1002/ijc.25670
Hubmann, R., Schnabl, S., Araghi, M., Schmidl, C., Rendeiro, A. F., Hilgarth, M., et al.
(2020). Targeting nuclear NOTCH2 by gliotoxin recovers a tumor-suppressor
NOTCH3 activity in CLL. Cells 9 (6), 1484. doi:10.3390/cells9061484
Hynes, N. E., and Stoelzle, T. (2009). Key signalling nodes in mammary gland
development and cancer: myc. Breast Cancer Res. 11 (5), 210. doi:10.1186/
bcr2406
Jacobs, C. T., and Huang, P. (2019). Notch signalling maintains Hedgehog
responsiveness via a Gli-dependent mechanism during spinal cord patterning in
zebrash. Elife 8, e49252. doi:10.7554/eLife.49252
Jin, H. Y., Gong, W., Zhang, C. X., and Wang, S. M. (2013). Epigallocatechin gallate
inhibits the proliferation of colorectal cancer cells by regulating Notch signaling.
Oncotargets Ther. 6, 145153. doi:10.2147/OTT.S40914
Jung, Y., Park, H., Zhao, H. Y., Jeon, R., Ryu, J. H., and Kim, W. Y. (2014). Systemic
approaches identify a garlic-derived chemical, Z-ajoene, as a glioblastoma multiforme
cancer stem cell-specic targeting agent. Mol. Cells 37 (7), 547553. doi:10.14348/
molcells.2014.0158
Katoh, M. (2007). Networking of WNT, FGF, Notch, BMP, and Hedgehog signaling
pathways during carcinogenesis. Stem Cell Rev. 3(1),3038. doi:10.1007/s12015-007-
0006-6
Khan, F., Singh, V. K., Saeed, M., Kausar, M. A., and Ansari, I. A. (2019). Carvacrol
induced program cell death and cell cycle arrest in androgen-independent human
prostate cancer cells via inhibition of Notch signaling. Anti-Cancer Agents Med. Chem.
19 (13), 15881608. doi:10.2174/1871520619666190731152942
Kiesel, V. A., and Stan, S. D. (2022). Modulation of Notch signaling pathway by
bioactive dietary agents. Int. J. Mol. Sci. 23 (7), 3532. doi:10.3390/ijms23073532
Kim,J.,Lee,J.S.,Jung,J.,Lim,I.,Lee,J.Y.,andPark,M.J.(2015).
Emodin suppresses maintenance of stemness by augmenting proteosomal
degradation of epidermal growth factor receptor/epidermal growth factor
receptor variant iii in glioma stem cells. Stem Cells Dev. 24 (3), 284295.
doi:10.1089/scd.2014.0210
Kim, S. Y., Kim, J. Y., Shin, W. S., Lee, S. J., Chi, S. G., Lee, J. Y., et al. (2018).
Saccharina japonica Extract suppresses stemness of glioma stem cells by degrading
epidermal growth factor receptor/epidermal growth factor receptor variant iii. J. Med.
Food 21 (5), 496505. doi:10.1089/jmf.2017.3992
Kim, T. H., Park, J. H., and Woo, J. S. (2019). Resveratrol induces cell death through
ROS-dependent downregulation of Notch1/PTEN/Akt signaling in ovarian cancer cells.
Mol. Med. Rep. 19 (4), 33533360. doi:10.3892/mmr.2019.9962
Kim, T. H., Woo, J. S., Kim, Y. K., and Kim, K. H. (2014). Silibinin induces cell death
through reactive oxygen species-dependent downregulation of Notch-1/ERK/Akt
signaling in human breast cancer cells. J. Pharmacol. Exp. Ther. 349 (2), 268278.
doi:10.1124/jpet.113.207563
Kode, A., Manavalan, J. S., Mosialou, I., Bhagat, G., Rathinam, C. V., Luo, N., et al.
(2014). Leukaemogenesis induced by an activating β-catenin mutation in osteoblasts.
Nature 506 (7487), 240244. doi:10.1038/nature12883
Koduru, S., Kumar, R., Srinivasan, S., and Damodaran, C. (2009). 1078 Inactivation of
Notch signaling by Withaferin-A in human colon cancer. EJC Suppl. 7 (2), 108. doi:10.
1016/s1359-6349(09)70371-4
Koduru, S., Kumar, R., Srinivasan, S., Evers, M. B., and Damodaran, C. (2010). Notch-
1 inhibition by Withaferin-A: a therapeutic target against colon carcinogenesis. Mol.
Cancer Ther. 9 (1), 202210. doi:10.1158/1535-7163.MCT-09-0771
Konishi, J., Kawaguchi, K. S., Vo, H., Haruki, N., Gonzalez, A., Carbone, D. P., et al.
(2007). Gamma-secretase inhibitor prevents Notch3 activation and reduces
proliferation in human lung cancers. Cancer Res. 67 (17), 80518057. doi:10.1158/
0008-5472.CAN-07-1022
Koprowski, S., Sokolowski, K., Kunnimalaiyaan, S., Gamblin, T. C., and
Kunnimalaiyaan, M. (2015). Curcumin-mediated regulation of Notch1/hairy and
enhancer of split-1/survivin: molecular targeting in cholangiocarcinoma. J. Surg. Res.
198 (2), 434440. doi:10.1016/j.jss.2015.03.029
Kowshik, J., Mishra, R., Sophia, J., Rautray, S., Anbarasu, K., Reddy, G. D., et al.
(2017). Nimbolide upregulates RECK by targeting miR-21 and HIF-1αin cell lines and
in a hamster oral carcinogenesis model. Sci. Rep. 7, 2045. doi:10.1038/s41598-017-
01960-5
Lacerda-Abreu, M. A., Russo-Abrahao, T., and Meyer-Fernandes, J. R. (2021).
Resveratrol is an inhibitor of sodium-dependent inorganic phosphate transport in
triple-negative MDA-MB-231 breast cancer cells. Cell Biol. Int. 45 (8), 17681775.
doi:10.1002/cbin.11616
Frontiers in Pharmacology frontiersin.org14
Yang et al. 10.3389/fphar.2024.1412669
Lee, J., Sehrawat, A., and Singh, S. V. (2012). Withaferin A causes activation of
Notch2 and Notch4 in human breast cancer cells. Breast Cancer Res. Treat. 136 (1),
4556. doi:10.1007/s10549-012-2239-6
Lee, R. T. H., Zhao, Z. H., and Ingham, P. W. (2016). Hedgehog signalling.
Development 143 (3), 367372. doi:10.1242/dev.120154
Lee, S. H., Nam, H. J., Kang, H. J., Kwon, H. W., and Lim, Y. C. (2013).
Epigallocatechin-3-gallate attenuates head and neck cancer stem cell traits through
suppression of Notch pathway. Eur. J. Cancer 49 (15), 32103218. doi:10.1016/j.ejca.
2013.06.025
Li, J. L., and Harris, A. L. (2009). Crosstalk of VEGF and Notch pathways in tumour
angiogenesis: therapeutic implications. Front. Bioscience-Landmark 14, 30943110.
doi:10.2741/3438
Li, L., Zhang, J., Xiong, N. Y., Li, S., Chen, Y., Yang, H., et al. (2016). Notch-1 signaling
activates NF-κB in human breast carcinoma MDA-MB-231 cells via PP2A-dependent
AKT pathway. Med. Oncol. 33 (4), 33. doi:10.1007/s12032-016-0747-7
Li, S. Y. S., Johnson, R., Smyth, L. C. D., and Dragunow, M. (2022). Platelet-derived
growth factor signalling in neurovascular function and disease. Int. J. Biochem. Cell Biol.
145, 106187. doi:10.1016/j.biocel.2022.106187
Li, X. J., Ma, S. J., Yang, P. Y., Sun, B. X., Zhang, Y., Sun, Y. H., et al. (2018). Anticancer
effects of curcumin on nude mice bearing lung cancer A549 cell subsets SP and NSP
cells. Oncol. Lett. 16 (5), 67566762. doi:10.3892/ol.2018.9488
Li, Y., Wang, Z. C., Jin, J. J., Zhu, S. X., He, G. Q., Li, S. H., et al. (2020). Quercetin
pretreatment enhances the radiosensitivity of colon cancer cells by targeting Notch-1
pathway. Biochem. Biophysical Res. Commun. 523 (4), 947953. doi:10.1016/j.bbrc.
2020.01.048
Li, Y. W., Maitah, M. Y., Ahmad, A., Kong, D., Bao, B., and Sarkar, F. H. (2012).
Targeting the Hedgehog signaling pathway for cancer therapy. Expert Opin. Ther.
Targets 16 (1), 4966. doi:10.1517/14728222.2011.617367
Liao,S.K.,Xia,J.,Chen,Z.W.,Zhang,S.H.,Ahmad,A.,Miele,L.,etal.(2011).Inhibitory
effect of curcumin on oral carcinoma CAL-27 cells via suppression of Notch-1 and NF-κB
signaling pathways. J. Cell. Biochem. 112 (4), 10551065. doi:10.1002/jcb.23019
Liao, W. H., Zhang, L. L., Chen, X., Xiang, J. Y., Zheng, Q., Chen, N. Z., et al. (2023).
Targeting cancer stem cells and signaling pathways through phytochemicals: a
promising approach against colorectal cancer. Phytomedicine 108, 20. doi:10.1016/j.
phymed.2022.154524
Linder, B., Wehle, A., Hehlgans, S., Bonn, F., Dikic, I., Rödel, F., et al. (2019). Arsenic
Trioxide and (-)-Gossypol synergistically target glioma stem-like cells via inhibition of
Hedgehog and Notch signaling. Cancers 11 (3), 350. doi:10.3390/cancers11030350
Liu, C. Y., Qi, M., Li, L., Yuan, Y., Wu, X. P., and Fu, J. S. (2020). Natural cordycepin
induces apoptosis and suppresses metastasis in breast cancer cells by inhibiting the
Hedgehog pathway. Food & Funct. 11 (3), 21072116. doi:10.1039/c9fo02879j
Liu, X., Zhang, Q., Li, Y., Zhao, D. X., and Gu, R. (2016). Dual mechanism of action of
resveratrol in Notch signaling pathway activation in osteosarcoma. Trop. J. Pharm. Res.
15 (1), 101106. doi:10.4314/tjpr.v15i1.14
Liu, Z. C., Yang, Z. X., Zhou, J. S., Zhang, H. T., Huang, Q. K., Dang, L. L., et al. (2014).
Curcumin regulates hepatoma cell proliferation and apoptosis through the Notch
signaling pathway. Int. J. Clin. Exp. Med. 7 (3), 714718.
Liubomirski, Y., and Ben-Baruch, A. (2020). Notch-inammation networks in
regulation of breast cancer progression. Cells 9 (7), 1576. doi:10.3390/cells9071576
Lubecka, K., Kurzava, L., Flower, K., Buvala, H., Zhang, H., Teegarden, D., et al.
(2016). Stilbenoids remodel the DNA methylation patterns in breast cancer cells and
inhibit oncogenic Notch signaling through epigenetic regulation of
MAML2 transcriptional activity. Carcinogenesis 37 (7), 656668. doi:10.1093/carcin/
bgw048
Lum, L., and Beachy, P. A. (2004). The Hedgehog response network: sensors, switches,
and routers. Science 304 (5678), 17551759. doi:10.1126/science.1098020
MacDonald, B. T., Tamai, K., and He, X. (2009). Wnt/beta-catenin signaling:
components, mechanisms, and diseases. Dev. Cell 17 (1), 926. doi:10.1016/j.devcel.
2009.06.016
Majumder, S., Crabtree, J. S., Golde, T. E., Minter, L. M., Osborne, B. A., and Miele, L.
(2021). Targeting Notch in oncology: the path forward. Nat. Rev. Drug Discov. 20 (2),
125144. doi:10.1038/s41573-020-00091-3
Manoranjan, B., Adile, A. A., Venugopal, C., and Singh, S. K. (2020). WNT: an
unexpected tumor suppressor in medulloblastoma. Oncology 7 (6), 1834903. doi:10.
1080/23723556.2020.1834903
McAuliffe, P. F., Meric-Bernstam, F., Mills, G. B., and Gonzalez-Angulo, A. M. (2010).
Deciphering the role of PI3K/Akt/mTOR pathway in breast cancer biology and
pathogenesis. Clin. Breast Cancer 10, S59S65. doi:10.3816/CBC.2010.s.013
Mccaw, T. R., Inga, E., Chen, H., JaskulaSztul, R., Dudeja, V., Bibb, J. A., et al. (2021).
Gamma secretase inhibitors in cancer: a current perspective on clinical performance.
Oncologist 26 (4), e608e621. doi:10.1002/onco.13627
Mishra, A. K., Sharma, V., Mutsuddi, M., and Mukherjee, A. (2021). Signaling cross-
talk during development: context-specic networking of Notch, NF-kB and JNK
signaling pathways in Drosophila.Cell. Signal. 82, 109937. doi:10.1016/j.cellsig.2021.
109937
Naznin, M. T., Maeda, T., and Morita, N. (2010). Antioxidant functions of E- and
Z-Ajoene derived from Japanese Garlic. Int. J. Food Prop. 13 (4), 821829. doi:10.1080/
10942910902895218
Okatani, T., Nishimura, M. F., Egusa, Y., Yoshida, S., Nishimura, Y., Nishikori, A.,
et al. (2024). Analysis of Notch1 protein expression in methotrexate-associated
lymphoproliferative disorders. J. Clin. Exp. Hematop. 64 (1), 19. doi:10.3960/jslrt.
23038
Okuhashi, Y., Itoh, M., Nara, N., and Tohda, S. (2011). Effects of combination of
Notch inhibitor plus Hedgehog inhibitor or Wnt inhibitor on growth of leukemia cells.
Anticancer Res. 31 (3), 893896.
Olson, L. E., Tollkuhn, J., Scafoglio, C., Krones, A., Zhang, J., Ohgi, K. A., et al. (2006).
Homeodomain-mediated beta-catenin-dependent switching events dictate cell-lineage
determination. Cell 125 (3), 593605. doi:10.1016/j.cell.2006.02.046
Oswald, F., Liptay, S., Adler, G., and Schmid, R. M. (1998). NF-kappaB2 is a putative
target gene of activated Notch-1 via RBP-Jkappa. Mol. Cell. Biol. 18 (4), 20772088.
doi:10.1128/mcb.18.4.2077
Pan, H., Zhou, W. B., He, W., Liu, X. A., Ding, Q., Ling, L. J., et al. (2012). Genistein
inhibits MDA-MB-231 triple-negative breast cancer cell growth by inhibiting NF-κB
activity via the Notch-1 pathway. Int. J. Mol. Med. 30 (2), 337343. doi:10.3892/ijmm.
2012.990
Papadopoulos, N., and Lennartsson, J. (2018). The PDGF/PDGFR pathway as a drug
target. Mol. Aspects Med. 62, 7588. doi:10.1016/j.mam.2017.11.007
Patel, N. S., Li, J. L., Generali, D., Poulsom, R., Cranston, D. W., and Harris, A. L.
(2005). Up-regulation of delta-like 4 ligand in human tumor vasculature and the role of
basal expression in endothelial cell function. Cancer Res. 65 (19), 86908697. doi:10.
1158/0008-5472.CAN-05-1208
Peiffer, D. S., Ma, E., Wyatt, D., Albain, K. S., and Osipo, C. (2020). DAXX-inducing
phytoestrogens inhibit ER+ tumor initiating cells and delay tumor development.
Npj Breast Cancer 6 (1), 37. doi:10.1038/s41523-020-00178-5
Pinchot, S. N., Jaskula-Sztul, R., Ning, L., Peters, N. R., Cook, M. R., Kunnimalaiyaan,
M., et al. (2011). Identication and validation of Notch pathway activating compounds
through a novel high-throughput screening method. Cancer 117 (7), 13861398. doi:10.
1002/cncr.25652
Ponnurangam, S., Mammen, J. M. V., Ramalingam, S., He, Z. Y., Zhang, Y. C., Umar,
S., et al. (2012). Honokiol in combination with radiation targets notch signaling to
inhibit colon cancer stem cells. Mol. Cancer Ther. 11 (4), 963972. doi:10.1158/1535-
7163.MCT-11-0999
Pozzo,F.,Bittolo,T.,Tissino,E.,Zucchetto, A., Bomben, R., Polcik, L., et al. (2022).
Multiple mechanisms of NOTCH1 activation in chronic lymphocytic leukemia:
NOTCH1 mutations and beyond. Cancers 14 (12), 2997. doi:10.3390/
cancers14122997
Prasad, S., Ravindran, J., and Aggarwal, B. B. (2010). NF-kappaB and cancer: how
intimate is this relationship. Mol. Cell. Biochem. 336 (1-2), 2537. doi:10.1007/s11010-
009-0267-2
Purow, B. W., Sundaresan, T. K., Burdick, M. J., Kefas, B. A., Comeau, L. D.,
Hawkinson, M. P., et al. (2008). Notch-1 regulates transcription of the epidermal
growth factor receptor through p53. Carcinogenesis 29 (5), 918925. doi:10.1093/carcin/
bgn079
Rajasinghe,L.D.,andGupta,S.V.(2017).Tocotrienol-richmixtureinhibitscell
proliferation and induces apoptosis via down-regulation of the Notch-1/NF-
κB pathways in NSCLC cells. Nutr. Diet. Suppl. 9, 103114. doi:10.2147/
nds.s129891
Ramamoorthy, P., Dandawate, P., Jensen, R. A., and Anant, S. (2021). Celas trol
and Triptolide suppress stemness in triple negative breast cancer: notch as a
therapeutic target for stem cells. Biomedicines 9 (5), 482. doi:10.3390/
biomedicines9050482
Reichrath, J., and Reichrath, S. (2020). Notch pathway and inherited diseases:
challenge and promise. Adv. Exp. Med. Biol. 1218, 159187. doi:10.1007/978-3-030-
34436-8_9
Ren, C. C., Amm, H. M., DeVilliers, P., Wu, Y. X., Deatherage, J. R., Liu, Z. Y., et al.
(2012). Targeting the Sonic Hedgehog pathway in keratocystic odontogenic tumor.
J. Biol. Chem. 287 (32), 2711727125. doi:10.1074/jbc.M112.367680
Ridgway, J., Zhang, G., Wu, Y., Stawicki, S., Liang, W. C., Chanthery, Y., et al. (2006).
Inhibition of Dll4 signalling inhibits tumour growth by deregulating angiogenesis.
Nature 444 (7122), 10831087. doi:10.1038/nature05313
Rizzo, P., Osipo, C., Foreman, K., Golde, T., Osborne, B., and Miele, L. (2008).
Rational targeting of Notch signaling in cancer. Oncogene 27 (38), 51245131. doi:10.
1038/onc.2008.226
Rodilla, V., Villanueva, A., Obrador-Hevia, A., Robert-Moreno, A., Fernández-
Majada, V., Grilli, A., et al. (2009). Jagged1 is the pathological link between Wnt
and Notch pathways in colorectal cancer. Proc. Natl. Acad. Sci. U. S. A. 106 (15),
63156320. doi:10.1073/pnas.0813221106
Sabbah, M., Emami, S., Redeuilh, G., Julien, S., Prévost, G., Zimber, A., et al. (2008).
Molecular signature and therapeutic perspective of the epithelial-to-mesenchymal
transitions in epithelial cancers. Drug Resist. Updat. 11 (4-5), 123151. doi:10.1016/
j.drup.2008.07.001
Frontiers in Pharmacology frontiersin.org15
Yang et al. 10.3389/fphar.2024.1412669
Salama, Y. A., El-karef, A., El Gayyar, A. M., and Abdel-Rahman, N. (2019). Beyond
its antioxidant properties: quercetin targets multiple signalling pathways in
hepatocellular carcinoma in rats. Life Sci. 236, 116933. doi:10.1016/j.lfs.2019.116933
Sanchez-Martin, M., Ambesi-Impiombato, A., Qin, Y., Herranz, D., Bansal, M.,
Girardi, T., et al. (2017). Synergistic antileukemic therapies in Notch1-induced T-ALL.
Proc. Natl. Acad. Sci. U. S. A. 114 (8), 20062011. doi:10.1073/pnas.1611831114
Sarkar, F. H., and Li, Y. W. (2008). NF-kappaB: a potential target for cancer
chemoprevention and therapy. Front. Bioscience-Landmark 13, 29502959. doi:10.
2741/2900
Saxton, R. A., and Sabatini, D. M. (2017). mTOR signaling in growth, Metabolism,
and Disease. Cell 168 (6), 960976. doi:10.1016/j.cell.2017.02.004
Seifert, J. R. K., and Mlodzik, M. (2007). Frizzled/PCP signalling: a conserved
mechanism regulating cell polarity and directed motility. Nat. Rev. Genet. 8 (2),
126138. doi:10.1038/nrg2042
Sha, J., Li, J., Wang, W., Pan, L., Cheng, J., Li, L., et al. (2016). Curcumin induces G0/
G1 arrest and apoptosis in hormone independent prostate cancer DU-145 cells by down
regulating Notch signaling. Biomed. Pharmacother. 84, 177184. doi:10.1016/j.biopha.
2016.09.037
Siddappa, G., Kulsum, S., Ravindra, D. R., Kumar, V. V., Raju, N., Ragha van, N., et al.
(2017). Curcumin and metformin-mediated chemoprevention of oral cancer is
associated with inhibition of cancer stem cells. Mol. Carcinog. 56 (11), 24462460.
doi:10.1002/mc.22692
Skelin, J., Matulic, M., Milkovic, L., Heckel, D., Skoko, J., Skreb, K. A., et al. (2024).
Analysis of primary chronic lymphocytic leukemia cellssignaling pathways.
Biomedicines 12 (3), 524. doi:10.3390/biomedicines12030524
Song, L. L., Peng, Y., Yun, J., Rizzo, P., Chaturvedi, V., Weijzen, S., et al. (2008).
Notch-1 associates with IKKalpha and regulates IKK activity in cervical cancer cells.
Oncogene 27 (44), 58335844. doi:10.1038/onc.2008.190
Subramaniam, D., Ponnurangam, S., Ramamoorthy, P., Standing, D., Battafarano, R.
J., Anant, S., et al. (2012). Curcumin induces cell death in esophageal cancer cells
through modulating Notch signaling. Plos One 7 (2), e30590. doi:10.1371/journal.pone.
0030590
Taghvaei, S., Sabouni, F., and Minuchehr, Z. (2022). Identication of natural products
as SENP2 inhibitors for targeted therapy in heart failure. Front. Pharmacol. 13, 817990.
doi:10.3389/fphar.2022.817990
Takebe, N., Nguyen, D., and Yang, S. X. (2014). Targeting Notch signaling pathway in
cancer: clinical development advances and challenges. Pharmacol. Ther. 141 (2),
140149. doi:10.1016/j.pharmthera.2013.09.005
Tang, Y. Q., and Cao, Y. M. (2022). Curcumin inhibits the growth and metastasis of
melanoma via miR-222-3p/SOX10/Notch Axis. Dis. Markers 2022, 3129781. doi:10.
1155/2022/3129781
Toden, S., Ravindranathan, P., Gu, J. H., Cardenas, J., Yuchang, M., and Goel, A.
(2018). Oligomeric proanthocyanidins (OPCs) target cancer stem-like cells and
suppress tumor organoid formation in colorectal cancer. Sci. Rep. 8, 3335. doi:10.
1038/s41598-018-21478-8
Toden, S., Tran, H. M., Tovar-Camargo, O. A., Okugawa, Y., and Goel, A. (2016).
Epigallocatechin-3-gallate targets cancer stem-like cells and enhances 5-uorouracil
chemosensitivity in colorectal cancer. Oncotarget 7 (13), 1615816171. doi:10.18632/
oncotarget.7567
Tuli, H. S., Sandhu, S. S., and Sharma, A. K. (2014). Pharmacological and therapeutic
potential of Cordyceps with special reference to Cordycepin. 3 Biotech. 4 (1), 112.
doi:10.1007/s13205-013-0121-9
Ullah, A., Ullah, N., Nawaz, T., and Aziz, T. (2023). Molecular mechanisms of
sanguinarine in cancer prevention and treatment. Anti-Cancer Agents Med. Chem. 23
(7), 765778. doi:10.2174/1871520622666220831124321
Vimalraj,S.(2022).AconcisereviewofVEGF,PDGF,FGF,Notch,angiopoietin,
and HGF signalling in tumor angiogenesis with a focus on alternative approaches and
future directions. Int. J. Biol. Macromol. 221, 14281438. doi:10.1016/j.ijbiomac.
2022.09.129
Vlashi, R., Zhang, X. E., Wu, M. R., and Chen, G. Q. (2023). Wnt signaling: essential
roles in osteoblast differentiation, bone metabolism and therapeutic implications for
bone and skeletal disorders. Genes & Dis. 10 (4), 12911317. doi:10.1016/j.gendis.2022.
07.011
Wall, D. S., Mears, A. J., McNeill, B., Mazerolle, C., Thurig, S., Wang, Y. P., et al.
(2009). Progenitor cell proliferation in the retina is dependent on Notch-independent
Sonic Hedgehog/Hes1 activity. J. Cell Biol. 184 (1), 101112. doi:10.1083/jcb.200805155
Wang, J. H., Shelly, L., Miele, L., Boykins, R., Norcross, M. A., and Guan, E. (2001).
Human Notch-1 inhibits NF-κB activity in the nucleus through a direct interaction
involving a novel domain. J. Immunol. 167 (1), 289295. doi:10.4049/jimmunol.167.
1.289
Wang, M., Sun, L. T., Wang, L., and Sun, Y. N. (2021). Effects of berberine on circular
rna expression proles in human gastric cancer cells. Evidence-Based Complementary
Altern. Med. 2021, 6688629. doi:10.1155/2021/6688629
Wang, S. E., Yu, Y., Criswell, T. L., DeBusk, L. M., Lin, P. C., Zent, R., et al. (2010).
Oncogenic mutations regulate tumor microenvironment through induction of growth
factors and angiogenic mediators. Oncogene 29 (23), 33353348. doi:10.1038/onc.
2010.112
Wang, Y. N., Wang, J., Yang, H. N., Zhang, B. L., Zhang, P., Sun, P. Y., et al. (2020).
The oxidation of (-)-epigallocatechin-3-gallate inhibits T-cell acute lymphoblastic
leukemia cell line HPB-ALL via the regulation of Notch1 expression. Rsc Adv. 10
(3), 16791684. doi:10.1039/c9ra08459b
Wang, Z., Zhang, Y., Banerjee, S., Li, Y., and Sarkar, F. H. (2006c). Inhibition of
nuclear factor κB activity by genistein is mediated via Notch-1 signaling pathway in
pancreatic cancer cells. Int. J. Cancer 118 (8), 19301936. doi:10.1002/ijc.21589
Wang, Z. W., Ahmad, A., Li, Y. W., Azmi, A. S., Miele, L., and Sarkar, F. H. (2011b).
Targeting Notch to eradicate pancreatic cancer stem cells for cancer therapy. Anticancer
Res. 31 (4), 11051113.
Wang, Z. W., Li, Y. W., Ahmad, A., Banerjee, S., Azmi, A. S., Kong, D. J., et al. (2011a).
Down-regulation of Notch-1 is associated with Akt and FoxM1 in inducing cell growth
inhibition and apoptosis in prostate cancer cells. J. Cell. Biochem. 112 (1), 7888. doi:10.
1002/jcb.22770
Wang, Z. W., Li, Y. W., Kong, D., Banerjee, S., Ahmad, A., Azmi, A. S., et al. (2009).
Acquisition of epithelial-mesenchymal transition phenotype of gemcitabine-resistant
pancreatic cancer cells is linked with activation of the Notch signaling pathway. Cancer
Res. 69 (6), 24002407. doi:10.1158/0008-5472.CAN-08-4312
Wang, Z. W., Zhang, Y. X., Banerjee, S., Li, Y. W., and Sarkar, F. H. (2006a). Notch-1
down-regulation by curcumin is associated with the inhibition of cell growth and the
induction of apoptosis in pancreatic cancer cells. Cancer 106 (11), 25032513. doi:10.
1002/cncr.21904
Wang, Z. W., Zhang, Y. X., Li, Y. W., Banerjee, S., Liao, J., and Sarkar, F. H. (2006b).
Down-regulation of Notch-1 contributes to cell growth inhibition and apoptosis in
pancreatic cancer cells. Mol. Cancer Ther. 5 (3), 483493. doi:10.1158/1535-7163.MCT-
05-0299
Wei, H., Ge, Q., Zhang, L. Y., Xie, J., Gan, R. H., Lu, Y. G., et al. (2022). EGCG inhibits
growth of tumoral lesions on lip and tongue of K-Ras transgenic mice through the
Notch pathway. J. Nutr. Biochem. 99, 108843. doi:10.1016/j.jnutbio.2021.108843
Wu, D., Wang, S. L., Oliveira, D. V., Del Gaudio, F., Vanlandewijck, M., Lebouvier, T.,
et al. (2021). The infantile myobromatosis Notch3 L1519P mutation leads to
hyperactivated ligand-independent Notch signaling and increased PDGFRB
expression. Dis. Models Mech. 14 (2), dmm046300. doi:10.1242/dmm.046300
Wu, W. B., Chiang, H. S., Fang, J. Y., and Hung, C. F. (2007). Inhibitory effect of
lycopene on PDGF-BB-induced signalling and migration in human dermal broblasts: a
possible target for cancer. Biochem. Soc. Trans. 35 (Pt 5), 13771378. doi:10.1042/
BST0351377
Xia, R., Xu, M., Yang, J., and Ma, X. (2022). The role of Hedgehog and Notch signaling
pathway in cancer. Mol. Biomed. 3 (1), 44. doi:10.1186/s43556-022-00099-8
Xiang, S. F., Zhao, Z., Zhang, T., Zhang, B., Meng, M., Cao, Z. F., et al. (2020).
Triptonide effectively suppresses gastric tumor growth and metastasis through
inhibition of the oncogenic Notch1 and NF-κB signaling pathways. Toxicol. Appl.
Pharmacol. 388, 114870. doi:10.1016/j.taap.2019.114870
Xu, L. N., Gu, L. N., Tao, X. F., Xu, Y. W., Qi, Y., Yin, L. H., et al. (2018). Effect of
dioscin on promoting liver regeneration via activating Notch1/Jagged1 signal pathway.
Phytomedicine 38, 107117. doi:10.1016/j.phymed.2017.11.006
Yamaguchi, N., Oyama, T., Ito, E., Satoh, H., Azuma, S., Hayashi, M., et al. (2008).
Notch3 signaling pathway plays crucial roles in the proliferation of ErbB2-negative
human breast cancer cells. Cancer Res. 68 (6), 18811888. doi:10.1158/0008-5472.CAN-
07-1597
Yamamoto, S., Schulze, K. L., and Bellen, H. J. (2014). Introduction to notch signaling.
Methods Mol. Biol. 1187, 114. doi:10.1007/978-1-4939-1139-4_1
Yang, J. Z., Wang, C. L., Zhang, Z. J., Chen, X. J., Jia, Y. S., Wang, B., et al. (2017).
Curcumin inhibits the survival and metastasis of prostate cancer cells via the Notch-1
signaling pathway. Apmis 125 (2), 134140. doi:10.1111/apm.12650
Yang, P., Zhou, X., and Xie, Y. (2023). Cytotoxic Effects of the benzophenanthridine
alkaloids isolated from Eomecon chionantha on MCF-7 cells and its potential
mechanism. Chem. Biodivers. 20 (1), e202200871. doi:10.1002/cbdv.202200871
Yang, Y., Nguyen, T. T., Pereira, I., Hur, J. S., and Kim, H. (2019). Lichen secondary
metabolite physciosporin decreases the stemness potential of colorectal cancer cells.
Biomolecules 9 (12), 797. doi:10.3390/biom9120797
Yang, Y. L., and Cao, Y. H. (2022). The impact of VEGF on cancer metastasis and
systemic disease. Seminars Cancer Biol. 86, 251261. doi:10.1016/j.semcancer.2022.
03.011
Yang, Y. T., Cui, J. J., Xue, F., Overstreet, A. M., Zhan, Y. P., Shan, D. P., et al. (2016).
Resveratrol represses pokemon expression in human glioma cells. Mol. Neurobiol. 53
(2), 12661278. doi:10.1007/s12035-014-9081-2
Yao, J., Qian, C. J., Ye, B., Zhang, X., and Liang, Y. (2012). ERK inhibition enhances
TSA-induced gastric cancer cell apoptosis via NF-κB-dependent and Notch-
independent mechanism. Life Sci. 91 (5-6), 186193. doi:10.1016/j.lfs.2012.06.034
Ying, M., Wang, S., Sang, Y., Sun, P., Lal, B., Goodwin, C. R., et al. (2011). Regulation
of glioblastoma stem cells by retinoic acid: role for Notch pathway inhibition. Oncogene
30 (31), 34543467. doi:10.1038/onc.2011.58
Frontiers in Pharmacology frontiersin.org16
Yang et al. 10.3389/fphar.2024.1412669
Yu, X. M., Jaskula-Sztul, R., Ahmed, K., Harrison, A. D., Kunnimalaiyaan, M., and
Chen, H. (2013a). Resveratrol induces differentiation markers expression in anaplastic
thyroid carcinoma via activation of Notch1 signaling and suppresses cell growth. Mol.
Cancer Ther. 12 (7), 12761287. doi:10.1158/1535-7163.MCT-12-0841
Yu, X. M., Jaskula-Sztul, R., Ahmed, K., Harrison, A. D., Kunnimalaiyaan, M., and
Chen, H. (2013b). Resveratrol induces differentiation markers expression in anaplastic
thyroid carcinoma via activation of Notch1 signaling and suppresses cell growth. Mol.
Cancer Ther. 12 (7), 12761287. doi:10.1158/1535-7163.MCT-12-0841
Zhang, H. F., Li, H., Liu, Z. G., Ge, A., Guo, E. Y., Liu, S. X., et al. (2018). Triptolide
inhibits the proliferation and migration of medulloblastoma Daoy cells by upregulation
of microRNA-138. J. Cell. Biochem. 119 (12), 98669877. doi:10.1002/jcb.27307
Zhang, P., He, D. X., Chen, Z., Pan, Q. X., Du, F. F., Zang, X., et al. (2016).
Chemotherapy enhances tumor vascularization via Notch signaling-mediated
formation of tumor-derived endothelium in breast cancer. Biochem. Pharmacol. 118,
1830. doi:10.1016/j.bcp.2016.08.008
Zhang, P., Li, H., Yang, B., Yang, F., Zhang, L. L., Kong, Q. Y., et al. (2014). Biological
signicance and therapeutic implication of resveratrol-inhibited Wnt, Notch and
STAT3 signaling in cervical cancer cells. Genes Cancer 5 (5-6), 154164. doi:10.
18632/genesandcancer.15
Zhang, Q. K., Zhu, Z. S., Guan, J. Q., Hu, Y. Y., Zhou, W. J., Ye, W. C., et al. (2022).
Hes1 controls proliferation and apoptosis in chronic lymphoblastic leukemia cells by
modulating PTEN expression. Mol. Biotechnol. 64 (12), 14191430. doi:10.1007/
s12033-022-00476-2
Zhang, S., Yang, Y., Liang, Z. X., Duan, W. X., Yang, J., Yan, J. J., et al. (2013). Silybin-
mediated inhibition of Notch signaling exerts antitumor activity in human hepatocellular
carcinoma cells. Plos One 8 (12), e83699. doi:10.1371/journal.pone.0083699
Zhang, X., Samadi, A. K., Roby, K. F., Timmermann, B., and Cohen, M. S. (2012).
Inhibition of cell growth and induction of apoptosis in ovarian carcinoma cell lines
CaOV3 and SKOV3 by natural withanolide Withaferin A. Gynecol. Oncol. 124 (3),
606612. doi:10.1016/j.ygyno.2011.11.044
Zhao, H., Wu, L., Yan, G., Chen, Y., Zhou, M., Wu, Y., et al. (2021). Inammation and
tumor progression: signaling pathways and targeted intervention. Signal Transduct.
Target Ther. 6 (1), 263. doi:10.1038/s41392-021-00658-5
Zhdanovskaya, N., Lazzari, S., Caprioglio, D., Firrincieli, M., Maioli, C., Pace, E., et al.
(2022). Identication of a novel curcumin derivative inuencing Notch pathway and
DNA damage as a potential therapeutic agent in T-ALL. Cancers 14 (23), 5772. doi:10.
3390/cancers14235772
Zhong,L.X.,Li,H.,Wu,M.L.,Liu,X.Y.,Zhong,M.J.,Chen,X.Y.,etal.
(2015). Inhibition of STAT3 signaling as critical molecular event in resveratrol-
suppressed ovarian cancer cells. J. Ovarian Res. 8, 25. doi:10.1186/s13048-015-
0152-4
Zhou, B. H., Lin, W. L., Long, Y. L., Yang, Y. K., Zhang, H., Wu, K. M., et al. (2022).
Notch signaling pathway: architecture, disease, and therapeutics. Signal Transduct.
Target. Ther. 7 (1), 95. doi:10.1038/s41392-022-00934-y
Zhou, P. P., Wang, C. L., Hu, Z. B., Chen, W. R., Qi, W. T., and Li, A. K. (2017).
Genistein induces apoptosis of colon cancer cells by reversal of epithelial-to-
mesenchymal via a Notch1/NF-κB/slug/E-cadherin pathway. Bmc Cancer 17, 813.
doi:10.1186/s12885-017-3829-9
Zhou, X. X., Su, J. N., Feng, S. Y., Wang, L. X., Yin, X. Y., Yan, J. Z., et al. (2016).
Antitumor activity of curcumin is involved in down-regulation of YAP/TAZ expression
in pancreatic cancer cells. Oncotarget 7 (48), 7907679088. doi:10.18632/
oncotarget.12596
Frontiers in Pharmacology frontiersin.org17
Yang et al. 10.3389/fphar.2024.1412669
... The Notch signaling pathways are divided into canonical and non-canonical pathways. The canonical pathway plays a significant role in cell fate determination and intercellular communication, regulating embryonic development, tissue differentiation, and gene regulation, as well as contributing to both benign and malignant diseases [34]. The Notch signaling pathway involves multiple steps for the maturation and activation of Notch proteins [35] (Figure 2). ...
Article
Full-text available
Notch signaling is an evolutionarily conserved, multifunctional pathway involved in cell fate determination and immune modulation and contributes to the pathogenesis of autoinflammatory diseases. Emerging evidence reveals a bidirectional interaction between Notch and the gut microbiota (GM), whereby GM composition is capable of modulating Notch signaling through the binding of microbial elements to Notch receptors, leading to immune modulation. Furthermore, Notch regulates the GM by promoting SCFA-producing bacteria while suppressing proinflammatory strains. Beneficial microbes, such as Lactobacillus and Akkermansia muciniphila, modulate Notch and reduce proinflammatory cytokine production (such as IL-6 and TNF-α). The interaction between GM and Notch can either amplify or attenuate inflammatory pathways in inflammatory bowel diseases (IBDs), Behçet’s disease, and PAPA syndrome. Together, these findings provide novel therapeutic perspectives for autoinflammatory diseases by targeting the GM via probiotics or inhibiting Notch signaling. This review focuses on Notch–GM crosstalk and how GM-based and/or Notch-targeted approaches may modulate immune responses and promote better clinical outcomes.
... Genistein suppresses the expression of critical molecules like cyclin B1, Bcl-2, and Bcl-XL, which are essential for cancer cell proliferation and resistance to apoptosis. Cyclin B1 regulates cell cycle progression, and its reduction halts cell division, while Bcl-2 and Bcl-XL are anti-apoptotic proteins whose downregulation shifts the balance toward apoptosis, eliminating TNBC cells (Yang et al. 2024). ...
Article
Full-text available
Breast cancer remains one of the leading causes of cancer-related deaths among women worldwide. Genistein (Gen), a phytoestrogen soy isoflavone, has emerged as a promising agent in the prevention and treatment of breast cancer due to its ability to function as a natural selective estrogen receptor modulator (SERM). This review explores the multifaceted mechanisms through which Gen and its derivatives exert their anticancer effects, including modulation of the PI3K/Akt signaling pathway, regulation of apoptosis, inhibition of angiogenesis, and impacts on DNA methylation and enzyme functions. We discuss the dual roles of Gen in both enhancing and inhibiting estrogen receptor (ER)-dependent pathways., highlighting its complex interactions with ERα and ERβ. Furthermore, the review examines the synergistic effect of combining Gen with conventional chemotherapeutic agents such as doxorubicin, cisplatin, and selenium, as well as other natural compounds like lycopene. Clinical studies suggest that while isoflavones may not significantly influence breast cancer progression in general, the high consumption of soy isoflavones is associated with reduced recurrence rates in breast cancer survivors. Importantly, Gen’s ability to modulate key signaling pathways and enhance the efficacy of existing treatments improves its potential as a valuable adjunct in breast cancer therapy. In conclusion, Gen and its derivatives offer a novel and promising approach for treatment of breast cancer. Continued research into their mechanisms of action and clinical applications will be essential in optimizing their therapeutic potential and translating these findings into effective clinical interventions.
Article
Full-text available
Chronic lymphocytic leukemia (CLL) is a lymphoproliferative disorder characterized by a specific expansion of mature B-cell clones. We hypothesized that the disease has a heterogeneous clinical outcome that depends on the genes and signaling pathways active in the malignant clone of the individual patient. It was found that several signaling pathways are active in CLL, namely, NOTCH1, the Ikaros family genes, BCL2, and NF-κB, all of which contribute to cell survival and the proliferation of the leukemic clone. Therefore, we analyzed primary CLL cells for the gene and protein expression of NOTCH1, DELTEX1, HES1, and AIOLOS in both peripheral blood lymphocytes (PBLs) and the bone marrow (BM) of patients, as well as the expression of BCL2 and miRNAs to see if they correlate with any of these genes. BCL2 and AIOLOS were highly expressed in all CLL samples as previously described, but we show here for the first time that AIOLOS expression was higher in the PBLs than in the BM. On the other hand, NOTCH1 activation was higher in the BM. In addition, miR-15a, miR-181, and miR-146 were decreased and miR-155 had increased expression in most samples. The activation of the NOTCH pathway in vitro increases the susceptibility of primary CLL cells to apoptosis despite high BCL2 expression.
Article
Full-text available
Methotrexate (MTX)-associated lymphoproliferative disorder (MTX-LPD) is a lymphoproliferative disorder in patients treated with MTX. The mechanism of pathogenesis is still elusive, but it is thought to be a complex interplay of factors, such as underlying autoimmune disease activity, MTX use, Epstein-Barr virus infection, and aging. The NOTCH genes encode receptors for a signaling pathway that regulates various fundamental cellular processes, such as proliferation and differentiation during embryonic development. Mutations of NOTCH1 have been reported in B-cell tumors, including chronic lymphocytic leukemia/lymphoma, mantle cell lymphoma, and diffuse large B-cell lymphoma (DLBCL). Recently, it has also been reported that NOTCH1 mutations are found in post-transplant lymphoproliferative disorders, and in CD20-positive cells in angioimmunoblastic T-cell lymphoma, which might be associated with lymphomagenesis in immunodeficiency. In this study, to investigate the association of NOTCH1 in the pathogenesis of MTX-LPD, we evaluated protein expression of Notch1 in nuclei immunohistochemically in MTX-LPD cases [histologically DLBCL-type (n = 24) and classical Hodgkin lymphoma (CHL)-type (n = 24)] and de novo lymphoma cases [DLBCL (n = 19) and CHL (n = 15)]. The results showed that among MTX-LPD cases, the expression of Notch1 protein was significantly higher in the DLBCL type than in the CHL type (P < 0.001). In addition, among DLBCL morphology cases, expression of Notch1 tended to be higher in MTX-LPD than in the de novo group; however this difference was not significant (P = 0.0605). The results showed that NOTCH1 may be involved in the proliferation and tumorigenesis of B cells under the use of MTX. Further research, including genetic studies, is necessary.
Article
Full-text available
Glioblastoma multiforme (GBM) is one of the most aggressive types of cancer characterized by poor patient outcomes. To date, it is believed that the major cause of its recurrence and chemoresistance is represented by the enrichment of GBM stem cells (GSCs) sustained by the abnormal activation of a number of signaling pathways. In this study, we found that in GBM cells, treatment with low toxicity doses of the γ-secretase inhibitor RO4929097 (GSI), blocking the Notch pathway activity, in combination with resveratrol (RSV) was able to reverse the basal mesenchymal phenotype to an epithelial-like phenotype, affecting invasion and stemness interplay. The mechanism was dependent on cyclin D1 and cyclin-dependent kinase (CDK4), leading to a reduction of paxillin (Pxn) phosphorylation. Consequently, we discovered the reduced interaction of Pxn with vinculin (Vcl), which, during cell migration, transmits the intracellular forces to the extracellular matrix. The exogenous expression of a constitutively active Cdk4 mutant prevented the RSV + GSI inhibitory effects in GBM cell motility/invasion and augmented the expression of stemness-specific markers, as well as the neurosphere sizes/forming abilities in untreated cells. In conclusion, we propose that Cdk4 is an important regulator of GBM stem-like phenotypes and invasive capacity, highlighting how the combined treatment of Notch inhibitors and RSV could be prospectively implemented in the novel therapeutic strategies to target Cdk4 for these aggressive brain tumors.
Article
Full-text available
Notch and Hedgehog signaling are involved in cancer biology and pathology, including the maintenance of tumor cell proliferation, cancer stem-like cells, and the tumor microenvironment. Given the complexity of Notch signaling in tumors, its role as both a tumor promoter and suppressor, and the crosstalk between pathways, the goal of developing clinically safe, effective, tumor-specific Notch-targeted drugs has remained intractable. Drugs developed against the Hedgehog signaling pathway have affirmed definitive therapeutic effects in basal cell carcinoma; however, in some contexts, the challenges of tumor resistance and recurrence leap to the forefront. The efficacy is very limited for other tumor types. In recent years, we have witnessed an exponential increase in the investigation and recognition of the critical roles of the Notch and Hedgehog signaling pathways in cancers, and the crosstalk between these pathways has vast space and value to explore. A series of clinical trials targeting signaling have been launched continually. In this review, we introduce current advances in the understanding of Notch and Hedgehog signaling and the crosstalk between pathways in specific tumor cell populations and microenvironments. Moreover, we also discuss the potential of targeting Notch and Hedgehog for cancer therapy, intending to promote the leap from bench to bedside.
Article
Full-text available
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy considered curable by modern clinical management. Nevertheless, the prognosis for T-ALL high-risk cases or patients with relapsed and refractory disease is still dismal. Therefore, there is a keen interest in developing more efficient and less toxic therapeutic approaches. T-ALL pathogenesis is associated with Notch signaling alterations, making this pathway a highly promising target in the fight against T-ALL. Here, by exploring the anti-leukemic capacity of the natural polyphenol curcumin and its derivatives, we found that curcumin exposure impacts T-ALL cell line viability and decreases Notch signaling in a dose- and time-dependent fashion. However, our findings indicated that curcumin-mediated cell outcomes did not depend exclusively on Notch signaling inhibition, but might be mainly related to compound-induced DNA-damage-associated cell death. Furthermore, we identified a novel curcumin-based compound named CD2066, endowed with potentiated anti-proliferative activity in T-ALL compared to the parent molecule curcumin. At nanomolar concentrations, CD2066 antagonized Notch signaling, favored DNA damage, and acted synergistically with the CDK1 inhibitor Ro3306 in T-ALL cells, thus representing a promising novel candidate for developing therapeutic agents against Notch-dependent T-ALL.
Article
Full-text available
Historically, natural plant-derived drugs received a great impact of consideration in the treatment of several human-associated disorders. Cancer is a devastating disease and the second most cause of mortality. Sanguinarine (SANG), a naturally isolated plant alkaloidal agent, possesses chemo-preventive effects. Several studies have revealed that SANG impedes tumor metastasis and development by disrupting a wide range of cell signaling pathways and its molecular targets such as BCL-2, MAPKs, Akt, NF-κB, ROS, and microRNAs (miRNAs). Although its low chemical stability and poor oral bioavailability continue to be a key issue in its use as a medicinal molecule. A novel method (e.g., liposomes, nanoparticles, and micelles) and alternative analogs provide an exciting approach to alleviate these problems and broaden its pharmacokinetic profile. Cancer-specific miRNAs expression is synchronized by SANG, which has also been uncertain. In this critical study, we review the utilization of SANG mimics and nano-technologies to improve its support in cancer. We focus on recently disclosed studies on SANG anti-cancer properties.
Article
Angiogenesis is a double-edged sword; it is a mechanism that defines the boundary between health and disease. In spite of its central role in physiological homeostasis, it provides the oxygen and nutrition needed by tumor cells to proceed from dormancy if pro-angiogenic factors tip the balance in favor of tumor angiogenesis. Among pro-angiogenic factors, vascular endothelial growth factor (VEGF) is a prominent target in therapeutic methods due to its strategic involvement in the formation of anomalous tumor vasculature. In addition, VEGF exhibits immune-regulatory properties which suppress immune cell antitumor activity. VEGF signaling through its receptors is an integral part of tumoral angiogenic approaches. A wide variety of medicines have been designed to target the ligands and receptors of this pro-angiogenic superfamily. Herein, we summarize the direct and indirect molecular mechanisms of VEGF to demonstrate its versatile role in the context of cancer angiogenesis and current transformative VEGF-targeted strategies interfering with tumor growth.
Article
Seven benzophenanthridine alkaloids (1‐7) were obtained from the 75% EtOH extract of Eomecon chionantha, and exhibited moderate biological activity against MCF‐7 cells. 8,12‐dimethoxysanguinarine (1, DSG) strongly decreased the cell viability of MCF‐7 cell lines with an IC50 value of 7.12 μΜ. Based on RNA‐sequencing measure and KEGG pathway enrichment analysis results, the significant differentially expressed genes (DEGs) were associated with in Pathways in Cancer and PI3K‐AKT signaling pathways in DSG treated group. The potential molecular regulatory mechanisms underlying the effect of DSG induces necroptosis in MCF‐7 cells via molecular docking, TEM analysis, and ROS measurement. Besides, DEGs of bone metastasis‐related genes such as PI3K, IGF1R, Notch, and Wnt mRNA were significantly downregulated in the DSG‐treated group on MCF‐7 cells. DSG might be selected as a bone metastasis proteins inhibitor of IL‐1β, IL‐6, IκBα, IGF1R, Notch, NF‐κB, PTHrp, PI3K, PKB/AKT, PTEN, TNF‐α, and Wnt via molecular docking. DSG suppressed bone metastasis by regulating the expression levels of IL‐1β, IL‐6, PTH, CROSS, TP1NP, and OSTEOC on MCF‐7 cells using ELISA measurement. Thus, our findings reveal that DSG could be a leadcompound for suppressing tumor cells to bone metastasis in breast cancer cells.
Article
Background Cancer stem cells (CSCs) are strongly associated with high tumourigenicity, chemotherapy or radiotherapy resistance, and metastasis and recurrence, particularly in colorectal cancer (CRC). Therefore, targeting CSCs may be a promising approach. Recently, discovery and research on phytochemicals that effectively target colorectal CSCs have been gaining popularity because of their broad safety profile and multi-target and multi-pathway modes of action. Purpose This review aimed to elucidate and summarise the effects and mechanisms of phytochemicals with potential anti-CSC agents that could contribute to the better management of CRC. Methods We reviewed PubMed, EMBASE, Web of Science, Ovid, ScienceDirect and China National Knowledge Infrastructure databases from the original publication date to March 2022 to review the mechanisms by which phytochemicals inhibit CRC progression by targeting CSCs and their key signalling pathways. Phytochemicals were classified and summarised based on the mechanisms of action. Results We observed that phytochemicals could affect the biological properties of colorectal CSCs. Phytochemicals significantly inhibit self-renewal, migration, invasion, colony formation, and chemoresistance and induce apoptosis and differentiation of CSCs by regulating the Wnt/β-catenin pathway (e.g., diallyl trisulfide and genistein), the phosphatidylinositol-3-kinase/protein kinase B/mammalian target of rapamycin pathway (e.g., caffeic acid and piperlongumine), the neurogenic locus notch homolog protein pathway (e.g., honokiol, quercetin, and α-mangostin), the Janus kinase-signal transducer and activator of transcription pathway (e.g., curcumin, morin, and ursolic acid), and other key signalling pathways. It is worth noting that several phytochemicals, such as resveratrol, silibinin, evodiamine, and thymoquinone, highlight multi-target and multi-pathway effects in restraining the malignant biological behaviour of CSCs. Conclusions This review demonstrates the potential of targeted therapies for colorectal CSCs using phytochemicals. Phytochemicals could serve as novel therapeutic agents for CRC and aid in drug development.
Article
Angiogenesis forms new vessels from existing ones. Abnormal angiogenesis, which is what gives tumor microenvironments their distinctive features, is characterised by convoluted, permeable blood vessels with a variety of shapes and high perfusion efficiency. Tumor angiogenesis controls cancer growth by allowing invasion and metastasis and is highly controlled by signalling networks. Therapeutic techniques targeting VEGF, PDGF, FGF Notch, Angiopoietin, and HGF signalling restrict the tumor's vascular supply. Numerous pathways regulate angiogenesis, and when one of those processes is blocked, the other pathways may step in to help. VEGF signalling inhibition alone has limits as an antiangiogenic therapy, and additional angiogenic pathways such as FGF, PDGF, Notch, angiopoietin, and HGF are important. For the treatment of advanced solid tumors, there are also new, emerging medicines that target multiple angiogenic pathways. Recent therapies block numerous signalling channels concurrently. This study focuses on ‘alternative’ methods to standard antiangiogenic medicines, such as cyclooxygenase-2 blocking, oligonucleotide binding complementary sites to noncoding RNAs to regulate mRNA target, matrix metalloproteinase inhibition and CRISPR/Cas9 based gene edition and dissecting alternative angiogenesis mechanism in tumor microenvironment.