You are on page 1of 9

Biomedicine & Pharmacotherapy 141 (2021) 111873

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

The role of Hypoxia-Inducible Factor-1alpha and its signaling in melanoma


Mohammad Malekan a, b, Mohammad Ali Ebrahimzadeh c, *, Fateme Sheida d
a
Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
b
Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
c
Pharmaceutical Sciences Research Center, School of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
d
Student Research Committee, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran

A R T I C L E I N F O A B S T R A C T

Keywords: Adaptation to the loss of O2 is regulated via the activity of hypoxia-inducible factors such as Hypoxia-Inducible
Hypoxia Factor-1 (HIF-1). HIF-1 acts as a main transcriptional mediator in the tissue hypoxia response that regulates over
HIF-1α 1000 genes related to low oxygen tension. The role of HIF-1α in oncogenic processes includes angiogenesis,
Melanoma
tumor metabolism, cell proliferation, and metastasis, which has been examined in various malignancies, such as
Signaling pathways
Diagnostic/prognostic biomarker
melanoma. Melanoma is accompanied by a high death rate and a cancer type whose incidence has risen over the
Melanoma treatment last decades. The linkage between O2 loss and melanogenesis had extensively studied over decades. Recent
studies revealed that HIF-1α contributes to melanoma progression via different signaling pathways such as PI3K/
Akt/mTOR, RAS/RAF/MEK/ERK, JAK/STAT, Wnt/β-catenin, Notch, and NF-κB. Also, various microRNAs (miRs)
are known to mediate the HIF-1α role in melanoma. Therefore, HIF-1α offers a diagnostic/prognostic biomarker
and a candidate for targeted therapy in melanoma.

1. Introduction (PHD1–3), which finally leads to their recognition by the von


Hippel-Lindau (VHL) protein their subsequent degradation by the pro­
In most solid tumors, adaptation to the reduction of O2 availability is teasome. In hypoxic conditions, PHDs activity is blocked, and VHL
regulated via the activity of hypoxia-inducible factors such as Hypoxia- cannot identify HIF-1α subunits, leading to its nuclear pathological ef­
Inducible Factor-1 (HIF-1) [1]. HIF-1 is a primary transcriptional fect [6]. Therefore, HIF-1α plays a role in angiogenesis mediated by
mediator in the tissue hypoxia response and the main regulator of O2 vascular endothelial growth factor (VEGF), cellular metabolism, tumor
homeostasis that regulates over 1000 genes related to low oxygen ten­ invasion and metastasis, and cell survival [7–10].
sion [1,2]. Also, HIF-1 is a heterodimeric complex consisting of a Melanoma is caused by the malignant change melanocytes, which
constitutively expressed beta subunit (HIF-1β, also known as the aryl typically occur in the skin but may rarely occur in the eye (uveal mel­
hydrocarbon receptor nuclear translocator [ARNT]) and an anoma) and mouth (oral melanoma); the pigment-producing cells lined
oxygen-regulated alpha subunit (HIF-1α) [3]. These proteins are a in the dermal-epidermal junction and the hair follicles [11–13]. Mela­
member of the basic helix-loop-helix–Per-ARNT-Sim (bHLH–PAS) fam­ noma is accompanied by a high mortality rate and a cancer type whose
ily. bHLH and PAS are needed for heterodimer arrangement between the incidence has increased over the last decades. Only in the United States,
HIF-1β and HIF-1α subunits, and the basic downstream area affords near 4.9 million adults have received skin cancer treatment from 2007 to
particular binding to the hormone response element (HRE) DNA chain 2011 [14,15].
[4]. Two transcription stimulator domains, C-terminal (C-TAD) and N- Extensive studies were conducted on various aspects of the rela­
terminal (N-TAD), were identified. The C-TAD has been indicated to tionship between melanoma and HIF-1α [16,17]. For example, Martí­
interact with CREB-binding protein (CBP) and p300 co-activators to nez-García et al. studied clinical staging and potential associations
activate gene transcription [3]. HIF-1α also includes an between HIF-1α expressions and melanoma in the prospective multi­
oxygen-dependent degradation domain (ODDD) that mediates center cohort study; they concluded that increasing of positively labeled
oxygen-regulated stability [5]. Under normoxic conditions, critical key HIF-1α melanoma cells might have the potential of serving as a mela­
proline residues HIFa subunits are hydroxylated by prolyl hydroxylases noma tumor prognosis and phenotype biomarker [8]. Hence, the present

* Corresponding author.
E-mail addresses: zadeh20@gmail.com, MA.Ebrahimzadeh@mazum.ac.ir (M.A. Ebrahimzadeh).

https://doi.org/10.1016/j.biopha.2021.111873
Received 26 May 2021; Received in revised form 25 June 2021; Accepted 28 June 2021
Available online 2 July 2021
0753-3322/© 2021 The Authors. Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
M. Malekan et al. Biomedicine & Pharmacotherapy 141 (2021) 111873

review aimed to address whether HIF-1α can serve as a diagnostic Akt phosphorylation and activity, leading to reduced apoptosis and
/prognostic biomarker and a candidate for targeted therapy in increased proliferation signaling [27,28]. A PTEN mutation has been
melanoma. reported in about 30–50% of melanoma cell lines [29]. It seems to cause
amplification of the gene PI3K, located in chromosome 3q26, and en­
2. HIF-1α and signaling pathways codes the p110a catalytic subunit of PI3K. This gene is frequently
amplified in several human cancers [30].
HIF-1α mediates the transcription of genes involved in different
processes, including stress adaptation, metabolism, apoptosis, tumor
growth, angiogenesis, and invasion [18]. Recent studies also revealed 2.2. The RAS/RAF/MEK/ERK pathway
that HIF-1α takes part in different signaling pathways such as
PI3K/Akt/mTOR, RAS/RAF/MEK/ERK, JAK/STAT, Wnt/β‑catenin, Some studies revealed that the MAPK/ERK pathway or RAS/RAF/
Notch, and NF‑κB [19,20]. These pathways and associated complex MEK/ERK pathway could regulate HIF-1α expression regulation [31,
changes appear to affect tumor growth control, metabolism, motility, 32]. Specific growth factors activate RAS, which in turn stimulates
and cell apoptosis in melanoma [21] (Fig. 1). RAS/RAF/MEK/ERK kinase stream [33]. This signaling pathway even­
tually increases mRNA translation rate into HIF-1α protein [33,34].
Also, ERK phosphorylates the co-activator CBP/p300, so it raises
2.1. The PI3K/Akt/mTOR pathway HIF-1α/p300 complex production. Interestingly, ERK affects the regu­
lation of HIF-1α synthesis and its transcriptional activation [33]. The
HIF-1α is located downstream of the mechanistic target of rapamycin RAS/RAF/MEK/ERK signaling pathway is central to the pathogenesis of
(mTOR) complex 1, which is individually regulated by the phosphati­ melanoma. It is frequently activated by oncogenic mutations, such as
dylinositol 3-kinase/protein kinase B (PI3K/Akt) signaling pathway B-RAF, N-RAS, and mutations at upstream membrane receptors (e.g.,
[22]. Amplified signaling through PI3K/Akt, and its downstream target, KIT) [35]. Mutations in B-RAF seem to be the main common genetic
mTORC1, increases HIF-1α-dependent gene expression in several cancer modification in 50–70% of melanoma cases [36,37]. N-RAS mutation is
types, including melanoma [23,24]. the other major oncogenic cause in melanoma progression and happens
The PI3K⁄Akt pathway activation in cancer is due mainly to muta­ in about 20–25% of all cases [36,38]. Furthermore, MEK’s importance in
tions in the tumor suppressor gene PTEN [25]. The PTEN gene produces RAS/RAF/MEK/ERK pathway activation has been revealed in mela­
a phosphatase whose chief act is degeneration of PI3K products by noma. Indeed, MEK acts as a mediator of melanoma oncogenesis and
dephosphorylating phosphatidylinositol 3,4,5-trisphosphate and phos­ plays a role in developing inhibiting apoptosis and cellular trans­
phatidylinositol 3,4-bisphosphate [26]. Loss of PTEN function increases formation [39].

Fig. 1. HIF1 complex and related signaling pathways.

2
M. Malekan et al. Biomedicine & Pharmacotherapy 141 (2021) 111873

2.3. The Wnt/β-catenin pathway Ploper et al. showed that MITF stabilization made a rise in multi­
vesicular body biosynthesis, which in turn enhanced Wnt signaling,
HIF-1α has a role both downstream and upstream of the Wnt/β-cat­ resulting in a positive-feedback loop that may account for melanoma
enin signaling pathway; indeed, these have a mutual regulative function proliferative stages [45].
[40,41]. The Wnt/β-catenin pathway is associated with the PI3K/Akt
pathway; also, β-catenin is involved in activating the RAS/RAF/ME­
K/ERK pathway [42,43]. So, Wnt/β-catenin can indirectly adjust HIF-1α 2.4. The JAK/STAT pathway
by PI3K/Akt and RAS/RAF/MEK/ERK pathways as an upstream
pathway. The signal transducer and activator of transcription (STAT) family
Wu et al., like Liu et al. in their studies, expressed HIF-1α acts up­ proteins consist of seven transcription factors, and the anus kinase (JAK)
stream of the Wnt/β-catenin pathway. These results showed that the loss family has four members [46,47]. They are involved in the activation of
of O2 in the cell environment elevated the expression of HIF-1α, sub­ target genes in response to growth factors and cytokines [47].
sequently activated the Wnt/β-catenin signaling pathway, and raised Recent studies have been shown that hypoxia activates the JAK⁄
β-catenin expression [40,41]. Microphthalmia Transcription Factor STAT signaling pathway in several tumor cell lines and animal models
(MITF) is the main determinant of both melanoma progression and via HIF-1α [48,49]. This pathway has also been indicated to work with
melanocyte development; Wnt/β-catenin signaling regulates the HIF1 in the activation of VEGF and haptoglobin gene transcription
expression of MITF. So this pathway has been promoted in the patho­ during hypoxia [50]. Multiple mechanisms have been inferred to
genesis of both malignant melanomas and benign melanocytic nevi [44]. mediate STAT’s effect on cancer formation, such as STAT impact on
immune surveillance and control of apoptosis, growth factor signaling,

Fig. 2. Representative Hypoxia/HIF-1α regulatory genes and their effects on melanoma progression.

3
M. Malekan et al. Biomedicine & Pharmacotherapy 141 (2021) 111873

and angiogenesis; all are likely also to impact melanoma [51,52]. committing the glucose to the glycolytic cycle [74,75]. HIF-1α also in­
Guenterberg et al. showed that IL-29 is expressed on melanoma cell duces transcription of pyruvate dehydrogenase kinase 1 (PDK1). PDK1
lines and activation with this cytokine led to the expression of multiple blocks the transformation of pyruvate to the acetyl CoA via inhibiting
genes and an increase in apoptosis via Jak-STAT signal transduction pyruvate dehydrogenase activity [75]. Multiple studies have examined
[53]. Another study suggests an increase of JAK/STAT signaling in the metabolism of melanoma cancer cells under hypoxia conditions [76,
melanoma tumor cell lines by genetic inhibition of the negative regu­ 77].
lator PTPN2 potentiated IFN-γ response in vivo and in vitro. Hence this Scott et al. have shown that all melanoma cell lines produce more
pathway may be a target to develop immunotherapy efficacy [54]. lactate and utilize more glucose than normal melanocytes [76]. In
another study, Koch and his colleagues conducted a study that widely
3. Hypoxia/HIF-1α in melanoma progression investigated the characterization of glycolysis-related gene expression in
malignant melanoma. They observed significant up-regulation of HIF-1α
The decreased availability of oxygen, hypoxia, is a major feature of target genes GLUT1, HK2, and lactate dehydrogenase-A (LDH-A) in
solid tumors, which increases poor clinical outcomes in cancer patients melanoma cell lines under hypoxic conditions. Moreover, GLUT1
[55]. Indeed, massive tumor-cell proliferation separates cells from the expression was significantly elevated in metastatic and BRAF-V600E
vasculature, then oxygen and nutrients shall be inadequate for the mutated melanoma cell lines. GLUT1 expression in melanoma tissue
rapidly growing tumor cells [56]. Developmental responses to hypoxia samples associated significantly with HK1, LDH-A, and Mono­
are mainly regulated by HIF- 1α that is encoded by the HIF1A gene [57]. carboxylate transporter 1 (MCT1) expression, which confirmed a
Hypoxia activates HIF-1α that promotes tumor growth by regulating the glycolytic phenotype of melanoma cell lines [77].
expression of genes involved in angiogenesis, metabolism, cell prolif­
eration, metastasis, and other biological mechanisms; these processes 3.3. Cell proliferation or cell death
finally lead to melanoma progression [58,59] (Fig. 2).
Hypoxia induces a chain of effects that lead to tumor cell prolifera­
3.1. Angiogenesis tion; however, in severe conditions, hypoxia can also help cell death by
the presence in tumors of a central necrotic zone [78]. In fact, HIF-1α
Angiogenesis is the new blood vessel organization from preexisting affects cell proliferation and death by regulating the expression of
ones and is required for tumor progression [60]. Angiogenic factors such cellular myelocytomatosis (C-MYC), insulin-like growth factor 2 (IGF2),
as vascular endothelial growth factor (VEGF), Nitric oxide synthase and other regulators of the cell cycle and death pathways [59]. Highly
(NOS), and transforming growth factor (TGF) activate tumor angio­ variable levels of hypoxia can be expected to accompany the dynamics
genesis [61–63]. The major regulator of angiogenesis is VEGF and its of tumor mass spatiotemporal growth so that a multitude of tumor cell
receptors, which are mainly associated with HIF-1α protein expression responses are represented [78]. C-MYC is a human oncogene that is
[64]. HIF-1α is translocated into the nucleus under hypoxic conditions, commonly altered in many forms of cancer, including melanoma [79,
activating a wide array of downstream genes, including VEGF [65]. 80]. C-MYC overexpression in cancer cells can increase the HIF-1α
In melanoma, vascular formation is highly seen, and there are expression and that of VEGF.
extensive studies that supporting HIF-1α and angiogenesis for the tumor Moreover, It is determined that C-MYC sets HIF-1α at the post-
progression and treatment of melanoma [66–68]. Simonetti et al. con­ transcriptional level. C-MYC is also essential for regulating angiogen­
ducted a study to recognize the correlation between angiogenesis and esis and contributes to tumor proliferation [79]. Mouriaux et al. con­
hypoxia in primary oral melanoma development. They used immuno­ ducted a retrospective analysis on uveal melanoma tumors from 88
staining markers of CD34, VEGF, and HIF-1α in 16 malignant primary patients. They concluded that increased HIF-1α expression correlated
melanomas patients. It was observed that there are positive correlations with cell proliferation and vascular markers CD31 and VEGF-A expres­
between VEGF and HIF-1α expression in invasive front vessels. Finally, sion [81]. Several studies stated that the HIF-1α induced BH3-only
they concluded that patients survival is significantly associated with protein family of cell death factors via BNIP3 and BNIP3L genes in
high HIF-1α expression and angiogenic activity of endothelial VEGF hypoxia conditions [78,82,83]. Wu et al. showed that α-melanocyte
[69]. stimulating hormone (α-MSH) stimulated the HIF-1α signaling, and then
the expression of BNIP3/BNIP3L is induced, thereby promoting the
3.2. Metabolism apoptosis and autophagy in melanoma cells [84].

It has been indicated that cancer cells have different metabolism 3.4. Metastasis
compared to normal cells; perhaps the most prominent variation in
cancer cells is aerobic glycolysis or the Warburg effect [70]. In this Activation of the HIF-1α is one of the most prominent mechanisms
condition, glucose metabolism is mainly fermentative rather than res­ that increase metastasis and enhances tumor aggressiveness [85].
piratory, with raised lactate production, even when enough O2 present HIF-1α regulates the expression of genes encoding metastasis, including
to support mitochondrial function [71]. Indeed, switching to aerobic fibronectin 1 (FN1), lysyl oxidase-like 2 (LOXL2), urokinase plasmin­
glycolysis is a critical adaptive response to hypoxia. Glycolysis has low ogen activator receptor (uPAR), epithelial-mesenchymal transition
efficiency in ATP production than mitochondrial respiration; therefore, (EMT) inducers, and other components involved in the invasion
the high glycolysis in the tumor for energy production causes cancer [86–88]. Lin et al. expressed that HIF-1α-related pathways such as
cells to consume more glucose [71,72]. MAPK, NF-κB, and PI3K/AKT mediated the expression of EMT genes that
HIF-1α is the main transcription factor for the induction of many initiated the metastatic cascade of melanoma. For example, the
genes encoding glycolytic enzymes and glucose transporters, allowing HIF-1α/NF-κB axis directly regulates EMT through the gene product
hypoxic tumor cells to take up more glucose efficiently [72]. The transcription. Then, this axis indirectly acts through the transcriptional
up-regulation of glucose transporter proteins 1 and 3 (GLUT1 and up-regulation of the metastasis inducer Snail [89].
GLUT3) is a critical characteristic in cancer cells regulated by HIF-1α. In another study, Li et al. reasoned that luteolin inhibited melanoma
GLUT transporter’s main cellular role is to facilitate glucose entry into cell migration and invasion by inhibiting the HIF-1α/VEGF signaling
the cell [73]. pathway. Luteolin effectively repressed EMT by decreased N-cadherin
Moreover, HIF-1α can up-regulate hexokinase 1 and 2 levels (HK1 and vimentin expression and increased E-cadherin in mRNA and protein
and HK2) in tumor cells. HK is the early glycolytic enzyme that enables levels. Besides, they stated that luteolin induces its anti-metastatic ac­
glucose conversion to glucose-6-phosphate in cancer cells, hence tion by reducing the p-Akt, HIF-1α, VEGF-A, p- VEGFR-2, MMP-2 MMP-

4
M. Malekan et al. Biomedicine & Pharmacotherapy 141 (2021) 111873

9 proteins expression [90]. 5. HIF-1α as a diagnostic/prognostic biomarker in melanoma

4. MicroRNAs as mediators of the HIF-1α role in melanoma HIF-1α has continuously been remarked as one of the diagnostic/
prognostic biomarkers for various cancers, including colorectal cancer
Extensive studies have examined the relationship between micro­ [103], pancreatic cancer [104], glioblastoma [105], as well as mela­
RNAs (miRs) and HIF-1α in melanoma. Some miRs have inhibitory ef­ noma [106,107]. Nevertheless, there are few studies about the diag­
fects on HIF-1α expression in melanoma tumors; however, there are nostic/prognostic marker role of HIF-1α in melanoma. Fridman et al. did
miRs that increase HIF-1α expression in melanogenesis [10,91–94]. a study on morphological characteristics of B16 rats melanoma pro­
Hwang et al. investigate the expression of four miRs, including miR-210, gression with a high and low resistance to hypoxia. They expressed that
miR-218, miR-224, and miR-452, in the human melanoma cell lines. it is necessary to consider hypoxia and hypoxia-inducible factors during
They determined that decreased expression of these miRs in the TGFβ1 exploring new prognostic markers for melanoma progression in mice
pathway-expressing melanoma cells restricted the cell cycle; these re­ [108].
sults suggested that HIF-1α may regulate the biological behavior of the Another study showed that HIF-1α target genes, including GLUT1,
TGF-β1 pathway-expressing melanoma cells and the expression of some HK2, and LDH-A, are up-regulated under hypoxic conditions. On the
genes through miR-210, miR-218, miR-224, and miR-452 [95]. other hand, Cyclin D1 expression was associated with GLUT1, HK1,
Another study revealed that miR-652/HIF-1α axis promoted migra­ LDH-A in human melanoma tissue samples. They stated that Cyclin D1
tion and proliferation of MUM-2B and MEL270 uveal melanoma cells; expression level could be a prognostic marker for melanoma patients’
indeed, miR-652 promoted HIF-1α signaling via suppression of Ho­ outcomes [77]. Xia et al. indicated that miR-652/HIF-1α/HOXA9 axis
meobox A9 (HOXA9) [96]. Chen et al. showed that HIF-1α was the could be a useful biomarker for uveal melanoma patients’ prognosis;
miR-138 direct target, and miR-138 negatively regulated the HIF-1α they stated that miR-652 enhances HIF-1α signaling via inhibition of
protein expression in WM451 melanoma cells. HIF-1α also has an HOXA9. Also, HOXA9 silencing weakens the inhibitor function of
important role in glycolysis; upregulation of miR-138 notably inhibited miR-652, and then cell proliferation rate and migration potential are
the glycolysis level. All in all, they concluded miR-138 suppressed the decreased in uveal melanoma cells [96].
invasion, proliferation, and glycolysis in malignant melanoma cells by
targeting HIF‑1α [97]. Table 1 summarizes studies examining miRs and 6. Targeting HIF-1α in melanoma treatment
HIF-1α relationships in melanoma based on a systematic search in
PUBMED and SCOPUS. Extensive studies are conducted on the potential of HIF-1α and

Table 1
Summary of studies that investigated miRs associated with HIF-1α.
MicroRNA Relationship Function Target gene / Cell type (s) Ref.
with HIF-1α Signaling pathway

miR-210 Up-regulated by Decreased expression of these four miRNAs in TGFβ1 pathway- Bnip3 / TGFβ1 0380-MMU, UACC-3093, 0620- [95]
miR-218 HIF-1α expressing melanoma cells lines restricts the cell cycle, while their pathway LNA, 0708-LND, and UACC-647
miR-224 overexpression in melanoma cells enhances the expression of the
miR-452 hypoxic response gene Bnip3
miR-33a Inhibits HIF-1α Overexpression of miR-33a significantly inhibited melanoma PDK3 / miR-33a/HIF- A375, WM451 and SK-MEL-1 [93]
tumorigenesis by inhibition of HIF-1α/PDK3 1α signaling axis
miR-652 Promotes HIF-1α Overexpression of miR-652 directly inhibited HOXA9 expression and miR-652/HOXA9/ MUM-2B and MEL270 [96]
led to the activation of the HIF-1 signaling pathway HIF-1 axis
miR-18b Inhibits HIF-1α miR-18b inhibits the growth of melanoma cells by directly targeting miR-18b/HIF-1α axis B16 and A375 [94]
HIF-1α in vitro and in vivo
miR-33a-5p Inhibits HIF-1α miR-33a-5p acts as a tumor suppressor by inhibiting HIF-1α-mediated Glycolysis / miR-33a- A375, WM35, WM451, and SK- [98]
glycolysis 5p/ HIF-1α axis MEL-1
miR-33a-3p Inhibits HIF-1α miR-33a-3p enhanced sensitivity to radiotherapy in melanoma cells Glycolysis / miR-33a- WM35, WM451, and A375 [99]
via repression of the hypoxia-inducible factor 1α (HIF-1α)/lactate 3p/ HIF-1α axis
dehydrogenase A glycolysis axis. Moreover, HIF-1α, after the
identification of the miR-33a-3p promoter region, via histone
deacetylase 2 reduces the expression of miR-33a-3p, in this regard,
creates a negative feedback loop is created.
miR-33b Inhibits HIF-1α Expression of HIF-1α negatively regulated by miR-33b at the post- Glycolysis / miR- WM35, WM451 and SK-MEL-1 [92]
transcriptional level in melanoma cells. Overexpression of HIF-1α 33b/HIF-1α axis
reverse the inhibitory effect of miR-33b on the proliferation and
glycolysis in melanoma cells.
miR-138 Inhibits HIF-1α miRNA-138 directly targeted HIF1α, and miRNA-138 overexpression miR-138/ HIF-1α WM35, A375 melanoma cells and [100]
or HIF1α inhibition significantly repressed the growth and metastasis HEK293
of melanoma in vivo
miR‑138 Inhibits HIF-1α miR-138 seems to inhibit proliferation, invasion, and glycolysis in Glycolysis / miR- WM451 melanoma cells and [97]
melanoma cells, potentially via the direct inhibition of HIF-1α. 138/ HIF-1α axis normal human melanocyte cell
line
miR-155 Inhibits HIF-1α The level of HIF-1α is a direct target of miR-155, and It is indicated miR-155/HIF-1α B16-F10 melanoma cells [101]
that miR-155 deficiency promoted melanoma growth
miR-211 Inhibits HIF-1α Expression of miR-211 reduced HIF-1α protein levels and decreased PDK4 / miR-211/ A375, and WM1552C, as well as [91]
cell growth during hypoxia. HIF-1α protein loss was associated with HIF-1α axis the human epidermal melanocyte
the downregulation of miR-211 target the gene PDK4. cell line HEM-l
miR-210 Inhibits HIF-1α The highest level of miR-210 expression caused HIF-1α gene NADH/NAD+/ miR- SK-MEL-30 [10]
silencing; their results suggest the potential use of targeted HIF- 1α 210/ HIF-1α
and miR-210 respectively gene silencing in the therapy of malignant
melanoma
miR‑199a‑5p Inhibits HIF-1α Overexpression of miR‑199a‑5p repressed cell proliferation and miR‑199a‑5p/HIF-1α B16 and HME1 melanoma cell [102]
arrested the cell cycle in the G1 phase; it also significantly lines
downregulated the expression of HIF-1α in vivo.

5
M. Malekan et al. Biomedicine & Pharmacotherapy 141 (2021) 111873

related target genes in melanoma treatment. One approach is sup­ showed encouraging inhibitive effects on melanoma cell migration and
pressing tumors via miRs, that HIF-1α regulates in tumor hypoxia con­ metastasis, proliferation, glycogenesis and glycolysis, protein synthesis,
ditions [109,110]. MiR-33a [93], miR-33b [92], miR‑199a‑5p [102], and energy metabolism. This study provided a preclinical treatment
miR-138 [97,100], and miR-18b [94] are miRs investigated as a sup­ strategy related to HIF-1α and its signaling pathways [124].
pressor of melanoma as presented in Table 1.
Moreover, other approaches based on using the HIF-1α oncogenic 7. Concluding remarks and future directions
mechanisms are recommended and practiced in various studies on
melanoma tumors inhibiting and curing melanoma patients [111–113]. This review showed that HIF-1α takes part in different signaling
Park et al. studied vanillin (an organic phenolic aldehyde) as an anti- pathways such as PI3K/Akt/mTOR, RAS/RAF/MEK/ERK, JAK/STAT,
metastatic and anti-cancer intervention in malignant melanoma cells. Wnt/β‑catenin. These pathways are associated with melanoma angio­
They observed that vanillin down-regulates the expression of HIF-1α genesis, metabolism, cell proliferation, metastasis. Also, it is pointed
protein and related HIF-1α target genes, including FN1, LOXL2, and different miRNAs engage in HIF-1α-related axes. HIF-1α not only can be
uPAR, which are involved in melanoma metastasis. Indeed, the vanillin a diagnostic/prognostic biomarker for melanoma patients but also a
exerted its anti-metastatic and anti-cancer effect via the STAT3/HIF-1α novel therapeutic approach. However, a lot remains to be done to clarify
signaling pathway under hypoxic conditions [111]. Liu et al. conducted the mechanisms associated with and the implication for treatment and
a study targeting HIF-1α through transferrin receptor-mediated endo­ melanoma diagnosis.
cytosis with the transferrin–polyethylenimine–short-hairpin RNA More research into the relevance of HIF-1α induced gene products as
(Tf-PEI-shRNA) complex for malignant melanoma repressing. They also markers of prognosis should start and develop. The expansion of anti-
reported that their method to target HIF-1α holds promise as a clinical angiogenic, anti-growth, anti-glycolysis, and anti-metastasis agents
gene therapy approach for malignant melanoma [112]. Another study based on HIF-1α with potential as a melanoma therapy has led the way
indicated that HIF- 1a negatively sets TET2 gene and protein expression; in demonstrating that the tumors’ hypoxic response can be targeted.
therapeutic interventions to inhibit HIF-1α via adjuvant ascorbic acid
may promote a critical tumor’s reexpression suppressor genes in ma­ Financial disclosure
lignant melanoma cells [114].
Zheng and his colleagues showed that the knockdown of F-box only This research did not receive any specific grant from funding
protein 22 (FBXO22) repressed angiogenesis, cell migration, and inva­ agencies in the public, commercial, or not-for-profit sectors.
sion by the HIF-1α/VEGF pathway in melanoma cell lines [115]. Kluza
et al. presented a preclinical validation of the HIF-1α and pyruvate de­ CRediT authorship contribution statement
hydrogenase kinase-3 (PDK3) pathway as a new therapeutic interven­
tion in metastatic melanoma. They declared that dichloroacetate (DCA) All authors actively contributed to writing the manuscript. Also, M.
presents its antitumor effects via blockades of the HIF-1a pathway, ul­ M. helped draw figures for the project, and M. A. E. developed the
timately reduce glycolysis and promoted mitochondrial respiration via a theoretical framework and contributed the final version of the
specific reduction in the expression of PDK3 [116]. Li et al. offered manuscript.
luteolin as a possible therapeutic candidate in melanoma, which acted
through the HIF-1α/VEGF signaling pathway and EMT markers [90]. Conflict of interest statement
Licarete et al. conducted a study on B16.F10 murine melanoma cells.
They handled simvastatin cytotoxicity to target HIF‑1α for anti-cancer There is no actual or potential conflict of interest and our funding
strategies against melanoma [117]. Spinella et al. presented an endo­ source has no role in study design, in the collection, analysis and
thelin B receptor antagonist, which prevented invasive behavior of interpretation of data, in the writing of the report, and in the decision to
melanomas via HIF-1α [118]. Smolarczyk et al. introduced anti-vascular submit the paper for publication.
agents with HIF-1α inhibitors, which repress melanoma tumors [119].
Zhao et al. determined that selective inhibition of HIF-1α and glycolysis Acknowledgments
regulated by pyruvate dehydrogenase kinase 1 (PDK1) through
2-methoxyestradiol (2-MeOE2) enhances radio-sensitivity of melanoma None to declare.
cells [120].
Also, Shah et al. introduced vitamin K3 (menadione) as an E3 References
ubiquitin ligase Siah2 inhibitor leading to melanoma tumorigenesis
blockage. They declared that Siah2 ubiquitin ligase regulates hypoxia [1] D. Flügel, A. Görlach, C. Michiels, T. Kietzmann, Glycogen synthase kinase 3
phosphorylates hypoxia-inducible factor 1α and mediates its destabilization in a
response and some related pathways such as MAPK. It was showed that VHL-independent manner, Mol. Cell. Biol. 27 (9) (2007) 3253–3265, https://doi.
menadione significantly suppressed the growth of melanoma xenograft org/10.1128/mcb.00015-07.
tumors [121]. Martí-Díaz et al. used acriflavine as a potent inhibitor of [2] J. Valencak, H. Kittler, K. Schmid, M. Schreiber, M. Raderer, M. Gonzalez-
Inchaurraga, P. Birner, H. Pehamberger, Prognostic relevance of hypoxia
HIF-1α dimerization to examine its potential in melanoma treatment. inducible factor-1α expression in patients with melanoma, Clin. Exp. Dermatol.:
They found that acriflavine via inhibiting the HIF-1α/PDK1 axis and Exp. Dermatol. 34 (8) (2009) e962–e964, https://doi.org/10.1111/j.1365-
blockage of necessary adaptive mechanisms for melanoma cell survival 2230.2009.03706.x.
[3] Q. Ke, M. Costa, Hypoxia-inducible factor-1 (HIF-1), Mol. Pharmacol. 70 (5)
under metabolic stress could be a new promising targeted therapy
(2006) 1469–1480, https://doi.org/10.1124/mol.106.027029.
approach for melanoma [122]. Furthermore, Indian researchers showed [4] S. Koyasu, M. Kobayashi, Y. Goto, M. Hiraoka, H. Harada, Regulatory
that Cinnamaldehyde suppressed melanoma cell angiogenesis and mechanisms of hypoxia-inducible factor 1 activity: two decades of knowledge,
Cancer Sci. 109 (3) (2018) 560–571, https://doi.org/10.1111/cas.13483.
metastasis induced by hypoxia. Indeed, Cinnamaldehyde acts through
[5] Z.-j Liu, G.L. Semenza, H.-f Zhang, Hypoxia-inducible factor 1 and breast cancer
targeting PI3/Akt/mTOR pathway, leading to reduction of HIF-1α pro­ metastasis, J. Zhejiang Univ. -Sci. B 16 (1) (2015) 32–43, https://doi.org/
tein content [123]. 10.1631/jzus.b1400221.
A group of Malaysian researchers preclinically carried out a study to [6] F. Meléndez-Rodríguez, A.A. Urrutia, D. Lorendeau, G. Rinaldi, O. Roche,
N. Böğürcü-Seidel, M. Ortega Muelas, C. Mesa-Ciller, G. Turiel, A. Bouthelier,
determine whether Phyllanthus (P. amarus, P. niruri, P. urinaria, and P. Hernansanz-Agustín, A. Elorza, E. Escasany, Q. Li, M. Torres-Capelli, D. Tello,
P. watsonii) could be a medication for skin melanoma treatment or not. E. Fuertes, E. Fraga, A. Martínez-Ruiz, B. Pérez, J.M. Giménez-Bachs, A.S. Salinas-
Interestingly, they observed that cancer-related progressive pathways Sánchez, T. Acker, R. Sánchez Prieto, S.M. Fendt, K. De Bock, J. Aragonés, HIF1α
suppresses tumor cell proliferation through inhibition of aspartate biosynthesis,
like NFκB, MAPK/ERK, hypoxia suppressed by down-regulation of pan- Cell Rep. 26 (9) (2019) 2257–2265, https://doi.org/10.1016/j.
Ras, c-Raf, RSK, Akt, and HIF-1α. They concluded that these compounds celrep.2019.01.106.

6
M. Malekan et al. Biomedicine & Pharmacotherapy 141 (2021) 111873

[7] A. Giatromanolaki, E. Sivridis, C. Kouskoukis, K.C. Gatter, A.L. Harris, M. [30] J.Á.F. Vara, E. Casado, J. de Castro, P. Cejas, C. Belda-Iniesta, M. González-Barón,
I. Koukourakis, Hypoxia-inducible factors 1α and 2α are related to vascular PI3K/Akt signalling pathway and cancer, Cancer Treat. Rev. 30 (2) (2004)
endothelial growth factor expression and a poorer prognosis in nodular malignant 193–204, https://doi.org/10.1016/j.ctrv.2003.07.007.
melanomas of the skin, Melanoma Res. 13 (5) (2003) 493–501, https://doi.org/ [31] J. Wan, W. Wu, Hyperthermia induced HIF-1a expression of lung cancer through
10.1097/00008390-200310000-00008. AKT and ERK signaling pathways, J. Exp. Clin. Cancer Res.: CR 35 (1) (2016) 119,
[8] M.Á. Martínez-García, E. Riveiro-Falkenbach, J.L. Rodríguez-Peralto, E. Nagore, https://doi.org/10.1186/s13046-016-0399-7.
A. Martorell-Calatayud, F. Campos-Rodríguez, R. Farré, L. Hernández Blasco, [32] F. Loayza-Puch, Y. Yoshida, T. Matsuzaki, C. Takahashi, H. Kitayama, M. Noda,
J. Bañuls Roca, E. Chiner Vives, A. Sánchez-de-la-Torre, J. Abad Capa, J. Hypoxia and RAS-signaling pathways converge on, and cooperatively
M. Montserrat, I. Almendros, A. Pérez-Gil, V. Cabriada Nuño, I. Cano-Pumarega, downregulate, the RECK tumor-suppressor protein through microRNAs,
J. Corral Peñafiel, T. Diaz Cambriles, O. Mediano, J. Dalmau Arias, D. Gozal, Oncogene 29 (18) (2010) 2638–2648, https://doi.org/10.1038/onc.2010.23.
N. Spanish Sleep, A prospective multicenter cohort study of cutaneous melanoma: [33] N. Sang, D.P. Stiehl, J. Bohensky, I. Leshchinsky, V. Srinivas, J. Caro, MAPK
clinical staging and potential associations with HIF-1α and VEGF expressions, signaling up-regulates the activity of hypoxia-inducible factors by its effects on
Melanoma Res. 27 (6) (2017) 558–564, https://doi.org/10.1097/ p300, J. Biol. Chem. 278 (16) (2003) 14013–14019, https://doi.org/10.1074/jbc.
cmr.0000000000000393. m209702200.
[9] X. Jin, L. Dai, Y. Ma, J. Wang, Z. Liu, Implications of HIF-1α in the tumorigenesis [34] G.L. Semenza, Signal transduction to hypoxia-inducible factor 1, Biochem.
and progression of pancreatic cancer, Cancer Cell Int. 20 (1) (2020) 1–11, Pharmacol. 64 (5–6) (2002) 993–998, https://doi.org/10.1016/s0006-2952(02)
https://doi.org/10.1186/s12935-020-01370-0. 01168-1.
[10] I. Špaková, W.F. Graier, M. Rabajdová, K. Dubayová, V. Nagyová, M. Mareková, [35] J.A. McCubrey, L.S. Steelman, W.H. Chappell, S.L. Abrams, E.W. Wong, F. Chang,
Hypoxia factors suppression effect on the energy metabolism of a malignant B. Lehmann, D.M. Terrian, M. Milella, A. Tafuri, F. Stivala, M. Libra, J. Basecke,
melanoma cell SK-MEL-30, Eur. Rev. Med. Pharmacol. Sci. 24 (2020) 4909–4920, C. Evangelisti, A.M. Martelli, R.A. Franklin, Roles of the Raf/MEK/ERK pathway
https://doi.org/10.26355/eurrev_202005_21180. in cell growth, malignant transformation and drug resistance, Biochim. Biophys.
[11] B. Bedogni, M.B. Powell, Skin hypoxia: a promoting environmental factor in Acta (BBA)-Mol. Cell Res. 1773 (8) (2007) 1263–1284, https://doi.org/10.1016/
melanomagenesis, Cell Cycle 5 (12) (2006) 1258–1261, https://doi.org/10.4161/ j.bbamcr.2006.10.001.
cc.5.12.2810. [36] D. Pjanova, et al., The role of the meiotic component in reproduction of B-RAF-
[12] L. Dong, S. You, Q. Zhang, S. Osuka, N.S. Devi, S. Kaluz, J.H. Ferguson, H. Yang, mutated melanoma: a review and “Brainstorming” session, Melanoma (2020),
G. Chen, B. Wang, H.E. Grossniklaus, E.G. Van Meir, Arylsulfonamide 64B https://doi.org/10.5772/intechopen.93641.
inhibits hypoxia/HIF-induced expression of c-Met and CXCR4 and reduces [37] A.X. Wang, X.Y. Qi, Targeting RAS/RAF/MEK/ERK signaling in metastatic
primary tumor growth and metastasis of uveal melanoma, Clin. Cancer Res. 25 melanoma, IUBMB Life 65 (9) (2013) 748–758, https://doi.org/10.1002/
(7) (2019) 2206–2218, https://doi.org/10.1158/1078-0432.ccr-18-1368. iub.1193.
[13] M.H. Smith, I. Bhattacharyya, D.M. Cohen, N.M. Islam, S.G. Fitzpatrick, L. [38] H. Yang, B. Higgins, K. Kolinsky, K. Packman, Z. Go, R. Iyer, S. Kolis, S. Zhao,
J. Montague, D.D. Damm, C.B. Fowler, Melanoma of the oral cavity: an analysis of R. Lee, J.F. Grippo, K. Schostack, M.E. Simcox, D. Heimbrook, G. Bollag, F. Su,
46 new cases with emphasis on clinical and histopathologic characteristics, Head RG7204 (PLX4032), a selective BRAFV600E inhibitor, displays potent antitumor
Neck Pathol. 10 (3) (2016) 298–305. activity in preclinical melanoma models, Cancer Res. 70 (13) (2010) 5518–5527,
[14] C.W. Rundle, et al., Epidemiologic burden of skin cancer in the US and https://doi.org/10.1158/0008-5472.can-10-0646.
worldwide, Curr. Dermatol. Rep. (2020) 1–14, https://doi.org/10.1016/j. [39] S.A. Byron, D.C. Loch, C.L. Wellens, A. Wortmann, J. Wu, J. Wang, K. Nomoto, P.
oooo.2015.05.030. M. Pollock, Sensitivity to the MEK inhibitor E6201 in melanoma cells is
[15] C. Karimkhani, A.C. Green, T. Nijsten, M.A. Weinstock, R.P. Dellavalle, associated with mutant BRAF and wildtype PTEN status, Mol. Cancer 11 (1)
M. Naghavi, C. Fitzmaurice, The global burden of melanoma: results from the (2012) 75, https://doi.org/10.1186/1476-4598-11-75.
Global Burden of Disease Study 2015, Br. J. Dermatol. 177 (1) (2017) 134–140, [40] C. Wu, et al., Wnt/β-catenin coupled with HIF-1α/VEGF signaling pathways
https://doi.org/10.1111/bjd.15510. involved in galangin neurovascular unit protection from focal cerebral ischemia,
[16] B. Zbytek, D.L. Peacock, T.N. Seagroves, A. Slominski, Putative role of HIF Sci. Rep. 5 (1) (2015) 1–11, https://doi.org/10.1038/srep16151.
transcriptional activity in melanocytes and melanoma biology, Derm. [41] H.L. Liu, D. Liu, G.R. Ding, P.F. Liao, J.W. Zhang, Hypoxia-inducible factor-1α
-Endocrinol. 5 (2) (2013) 239–251, https://doi.org/10.4161/derm.22678. and Wnt/β-catenin signaling pathways promote the invasion of hypoxic gastric
[17] B. Xu, X. Zhang, Y. Gao, J. Song, B. Shi, Microglial Annexin A3 promoted the cancer cells, Mol. Med. Rep. 12 (3) (2015) 3365–3373, https://doi.org/10.3892/
development of melanoma via activation of hypoxia-inducible factor-1α/vascular mmr.2015.3812.
endothelial growth factor signaling pathway, J. Clin. Lab. Anal. 35 (2) (2021), [42] M. Lau, C. Klausen, P. Leung, E-cadherin inhibits tumor cell growth by
23622, https://doi.org/10.1002/jcla.23622. suppressing PI3K/Akt signaling via β-catenin-Egr1-mediated PTEN expression,
[18] D. Singh, R. Arora, P. Kaur, B. Singh, R. Mannan, S. Arora, Overexpression of Oncogene 30 (24) (2011) 2753–2766, https://doi.org/10.1038/onc.2011.6.
hypoxia-inducible factor and metabolic pathways: possible targets of cancer, Cell [43] H. Ji, J. Wang, H. Nika, D. Hawke, S. Keezer, Q. Ge, B. Fang, X. Fang, D. Fang, D.
Biosci. 7 (1) (2017) 1–9, https://doi.org/10.1186/s13578-017-0190-2. W. Litchfield, K. Aldape, Z. Lu, EGF-induced ERK activation promotes CK2-
[19] Q. Zhang, Z. Han, Y. Zhu, J. Chen, W. Li, Role of hypoxia inducible factor-1 in mediated disassociation of α-catenin from β-catenin and transactivation of
cancer stem cells, Mol. Med. Rep. 23 (1) (2020) 1, https://doi.org/10.3892/ β-catenin, Mol. Cell 36 (4) (2009) 547–559, https://doi.org/10.1016/j.
mmr.2020.11655. molcel.2009.09.034.
[20] P. Lopez-Bergami, B. Fitchman, Ze Ronai, Understanding signaling cascades in [44] O.M. Lucero, D.W. Dawson, R.T. Moon, A.J. Chien, A re-evaluation of the
melanoma, Photochem. Photobiol. 84 (2) (2008) 289–306, https://doi.org/ “oncogenic” nature of Wnt/β-catenin signaling in melanoma and other cancers,
10.1111/j.1751-1097.2007.00254.x. Curr. Oncol. Rep. 12 (5) (2010) 314–318, https://doi.org/10.1007/s11912-010-
[21] C.Z. Michaylira, H. Nakagawa, Hypoxic microenvironment as a cradle for 0114-3.
melanoma development and progression, Cancer Biol. Ther. 5 (5) (2006) [45] D. Ploper, V.F. Taelman, L. Robert, B.S. Perez, B. Titz, H.W. Chen, T.G. Graeber,
476–479, https://doi.org/10.4161/cbt.5.5.2749. E. von Euw, A. Ribas, E.M. De Robertis, MITF drives endolysosomal biogenesis
[22] M. Shimobayashi, M.N. Hall, Making new contacts: the mTOR network in and potentiates Wnt signaling in melanoma cells, Proc. Natl. Acad. Sci. 112 (5)
metabolism and signalling crosstalk, Nat. Rev. Mol. Cell Biol. 15 (3) (2014) (2015) E420–E429, https://doi.org/10.1073/pnas.1424576112.
155–162, https://doi.org/10.1038/nrm3757. [46] J.E. Darnell, STATs and gene regulation, Science 277 (5332) (1997) 1630–1635,
[23] L. Soumoy, C. Schepkens, M. Krayem, A. Najem, V. Tagliatti, G.E. Ghanem, https://doi.org/10.1126/science.277.5332.1630.
S. Saussez, J.M. Colet, F. Journe, Metabolic reprogramming in metastatic [47] B. Wingelhofer, H.A. Neubauer, P. Valent, X. Han, S.N. Constantinescu, P.
melanoma with acquired resistance to targeted therapies: integrative T. Gunning, M. Müller, R. Moriggl, Implications of STAT3 and STAT5 signaling on
metabolomic and proteomic analysis, Cancers 12 (5) (2020), 1323, https://doi. gene regulation and chromatin remodeling in hematopoietic cancer, Leukemia 32
org/10.3390/cancers12051323. (8) (2018) 1713–1726, https://doi.org/10.1038/s41375-018-0117-x.
[24] M. Chen, J. Lu, W. Wei, Y. Lv, X. Zhang, Y. Yao, L. Wang, T. Ling, X. Zou, Effects [48] M.Z. Noman, S. Buart, J. Van Pelt, C. Richon, M. Hasmim, N. Leleu, W.
of proton pump inhibitors on reversing multidrug resistance via downregulating M. Suchorska, A. Jalil, Y. Lecluse, F. El Hage, M. Giuliani, C. Pichon, B. Azzarone,
V-ATPases/PI3K/Akt/mTOR/HIF-1α signaling pathway through TSC1/2 complex N. Mazure, P. Romero, F. Mami-Chouaib, S. Chouaib, The cooperative induction
and Rheb in human gastric adenocarcinoma cells in vitro and in vivo, of hypoxia-inducible factor-1α and STAT3 during hypoxia induced an impairment
OncoTargets Ther. 11 (2018) 6705–6722, https://doi.org/10.2147/ott.s161198. of tumor susceptibility to CTL-mediated cell lysis, J. Immunol. 182 (6) (2009)
[25] L.S. Steelman, F.E. Bertrand, J.A. McCubrey, The complexity of PTEN: mutation, 3510–3521, https://doi.org/10.4049/jimmunol.0800854.
marker and potential target for therapeutic intervention, Expert Opin. Ther. [49] S.-H. Kang, M.O. Yu, K.J. Park, S.G. Chi, D.H. Park, Y.G. Chung, Activated STAT3
Targets 8 (6) (2004) 537–550, https://doi.org/10.1517/14728222.8.6.537. regulates hypoxia-induced angiogenesis and cell migration in human
[26] C.-Y. Chen, J. Chen, L. He, B.L. Stiles, PTEN: tumor suppressor and metabolic glioblastoma, Neurosurgery 67 (5) (2010) 1386–1395, https://doi.org/10.1227/
regulator, Front. Endocrinol. 9 (2018), 338, https://doi.org/10.3389/ neu.0b013e3181f1c0cd.
fendo.2018.00338. [50] M.J. Gray, J. Zhang, L.M. Ellis, G.L. Semenza, D.B. Evans, S.S. Watowich, G.
[27] J. Hojný, N. Hájková, O. Kodet, Comprehensive analysis of PTEN in primary E. Gallick, HIF-1α, STAT3, CBP/p300 and Ref-1/APE are components of a
cutaneous melanoma, Folia Biol. 66 (2020) 7–16. transcriptional complex that regulates Src dependent hypoxic expression of VEGF
[28] P. Guldberg, P. Thor Straten, A. Birck, V. Ahrenkiel, A.F. Kirkin, J. Zeuthen, in pancreatic and prostrate carcinomas, Oncogene 24 (19) (2005) 3110–3120,
Disruption of the MMAC1/PTEN gene by deletion or mutation is a frequent event https://doi.org/10.1038/sj.onc.1208513.
in malignant melanoma, Cancer Res. 57 (17) (1997) 3660–3663. [51] J. Bromberg, Stat proteins and oncogenesis, J. Clin. Investig. 109 (9) (2002)
[29] H. Tsao, X. Zhang, E. Benoit, F.G. Haluska, Identification of PTEN/MMAC1 1139–1142, https://doi.org/10.1172/jci0215617.
alterations in uncultured melanomas and melanoma cell lines, Oncogene 16 (26) [52] D.W. Kulesza, K. Ramji, M. Maleszewska, J. Mieczkowski, M. Dabrowski,
(1998) 3397–3402, https://doi.org/10.1038/sj.onc.1201881. S. Chouaib, B. Kaminska, Search for novel STAT3-dependent genes reveals

7
M. Malekan et al. Biomedicine & Pharmacotherapy 141 (2021) 111873

SERPINA3 as a new STAT3 target that regulates invasion of human melanoma [77] A. Koch, E.V. Ebert, T. Seitz, P. Dietrich, M. Berneburg, A. Bosserhoff,
cells, Lab. Investig. 99 (11) (2019) 1607–1621, https://doi.org/10.1038/s41374- C. Hellerbrand, Characterization of glycolysis-related gene expression in
019-0288-8. malignant melanoma, Pathol. Res. Pract. 216 (1) (2020), 152752, https://doi.
[53] K.D. Guenterberg, V.P. Grignol, E.T. Raig, J.M. Zimmerer, A.N. Chan, F. org/10.1016/j.prp.2019.152752.
M. Blaskovits, G.S. Young, G.J. Nuovo, B.L. Mundy, G.B. Lesinski, W.E. Carson, [78] M.C. Brahimi-Horn, J. Chiche, J. Pouysségur, Hypoxia and cancer, J. Mol. Med.
Interleukin-29 binds to melanoma cells inducing Jak-STAT signal transduction 85 (12) (2007) 1301–1307, https://doi.org/10.1007/s00109-007-0281-3.
and apoptosis, Mol. Cancer Ther. 9 (2) (2010) 510–520. [79] C. Chen, S. Cai, G. Wang, X. Cao, X. Yang, X. Luo, Y. Feng, J. Hu, c-Myc enhances
[54] N. Luo, L. Formisano, P.I. Gonzalez-Ericsson, V. Sanchez, P.T. Dean, S. colon cancer cell-mediated angiogenesis through the regulation of HIF-1α,
R. Opalenik, M.E. Sanders, R.S. Cook, C.L. Arteaga, D.B. Johnson, J.M. Balko, Biochem. Biophys. Res. Commun. 430 (2) (2013) 505–511, https://doi.org/
Melanoma response to anti-PD-L1 immunotherapy requires JAK1 signaling, but 10.1016/j.bbrc.2012.12.006.
not JAK2, Oncoimmunology 7 (6) (2018), 1438106, https://doi.org/10.1158/ [80] S. Mannava, V. Grachtchouk, L.J. Wheeler, M. Im, D. Zhuang, E.G. Slavina, C.
1535-7163.mct-09-0461. K. Mathews, D.S. Shewach, M.A. Nikiforov, Direct role of nucleotide metabolism
[55] B. Muz, P. de la Puente, F. Azab, A.K. Azab, The role of hypoxia in cancer in C-MYC-dependent proliferation of melanoma cells, Cell Cycle 7 (15) (2008)
progression, angiogenesis, metastasis, and resistance to therapy, Hypoxia 3 2392–2400, https://doi.org/10.4161/cc.6390.
(2015) 83–92, https://doi.org/10.2147/hp.s93413. [81] F. Mouriaux, F. Sanschagrin, C. Diorio, S. Landreville, F. Comoz, E. Petit,
[56] J. Schödel, S. Grampp, E.R. Maher, H. Moch, P.J. Ratcliffe, P. Russo, D.R. Mole, M. Bernaudin, A.P. Rousseau, D. Bergeron, M. Morcos, Increased HIF-1α
Hypoxia, hypoxia-inducible transcription factors, and renal cancer, Eur. Urol. 69 expression correlates with cell proliferation and vascular markers CD31 and
(4) (2016) 646–657, https://doi.org/10.1016/j.eururo.2015.08.007. VEGF-A in uveal melanoma, Investig. Ophthalmol. Vis. Sci. 55 (3) (2014)
[57] D.W. Yoon, D. So, S. Min, J. Kim, M. Lee, R. Khalmuratova, C.H. Cho, J.W. Park, 1277–1283, https://doi.org/10.1167/iovs.13-13345.
H.W. Shin, Accelerated tumor growth under intermittent hypoxia is associated [82] J. Pouysségur, F. Dayan, N.M. Mazure, Hypoxia signalling in cancer and
with hypoxia-inducible factor-1-dependent adaptive responses to hypoxia, approaches to enforce tumour regression, Nature 441 (7092) (2006) 437–443,
Oncotarget 8 (37) (2017) 61592–61603, https://doi.org/10.18632/ https://doi.org/10.1038/nature04871.
oncotarget.18644. [83] M.C. Brahimi-Horn, J. Pouysségur, The hypoxia-inducible factor and tumor
[58] L. Li, F. Ren, C. Qi, L. Xu, Y. Fang, M. Liang, J. Feng, B. Chen, W. Ning, J. Cao, progression along the angiogenic pathway, Int. Rev. Cytol. 242 (2005) 157–214,
Intermittent hypoxia promotes melanoma lung metastasis via oxidative stress and https://doi.org/10.1016/s0074-7696(04)42004-x.
inflammation responses in a mouse model of obstructive sleep apnea, Respir. Res. [84] J.-C. Wu, H.E. Tsai, G.S. Liu, C.S. Wu, M.H. Tai, Autophagic cell death
19 (1) (2018) 28, https://doi.org/10.1186/s12931-018-0727-x. participates in POMC-induced melanoma suppression, Cell Death Discov. 4 (1)
[59] E.-J. Yeo, Hypoxia and aging, Exp. Mol. Med. 51 (6) (2019) 1–15, https://doi. (2018) 1–11, https://doi.org/10.1038/s41420-018-0070-5.
org/10.1038/s12276-019-0233-3. [85] D. Liao, C. Corle, T.N. Seagroves, R.S. Johnson, Hypoxia-inducible factor-1α is a
[60] J. Fang, Q. Zhou, L.Z. Liu, C. Xia, X. Hu, X. Shi, B.H. Jiang, Apigenin inhibits key regulator of metastasis in a transgenic model of cancer initiation and
tumor angiogenesis through decreasing HIF-1α and VEGF expression, progression, Cancer Res. 67 (2) (2007) 563–572, https://doi.org/10.1158/0008-
Carcinogenesis 28 (4) (2007) 858–864, https://doi.org/10.1093/carcin/bgl205. 5472.can-06-2701.
[61] D. Shweiki, A. Itin, D. Soffer, E. Keshet, Vascular endothelial growth factor [86] X. Lu, Y. Kang, Hypoxia and hypoxia-inducible factors: master regulators of
induced by hypoxia may mediate hypoxia-initiated angiogenesis, Nature 359 metastasis, Clin. Cancer Res. 16 (24) (2010) 5928–5935, https://doi.org/
(6398) (1992) 843–845, https://doi.org/10.1038/359843a0. 10.1158/1078-0432.ccr-10-1360.
[62] C. Brinkmann, A. Metten, P. Scriba, C.V. Tagarakis, P. Wahl, J. Latsch, K. Brixius, [87] A. Laurenzana, A. Chillà, C. Luciani, S. Peppicelli, A. Biagioni, F. Bianchini,
W. Bloch, Hypoxia and hyperoxia affect serum angiogenic regulators in t2dm men E. Tenedini, E. Torre, A. Mocali, L. Calorini, F. Margheri, G. Fibbi, M. Del Rosso,
during cycling, Int. J. Sports Med. 38 (02) (2017) 92–98, https://doi.org/ uPA/uPAR system activation drives a glycolytic phenotype in melanoma cells,
10.1055/s-0042-116823. Int. J. Cancer 141 (6) (2017) 1190–1200, https://doi.org/10.1002/ijc.30817.
[63] C.M. Failla, M. Carbo, V. Morea, Positive and negative regulation of angiogenesis [88] M.-H. Yang, M.Z. Wu, S.H. Chiou, P.M. Chen, S.Y. Chang, C.J. Liu, S.C. Teng, K.
by soluble vascular endothelial growth factor receptor-1, Int. J. Mol. Sci. 19 (5) J. Wu, Direct regulation of TWIST by HIF-1α promotes metastasis, Nat. Cell Biol.
(2018) 1306, https://doi.org/10.3390/ijms19051306. 10 (3) (2008) 295–305, https://doi.org/10.1038/ncb1691.
[64] D. Zagzag, H. Zhong, J.M. Scalzitti, E. Laughner, J.W. Simons, G.L. Semenza, [89] K. Lin, S. Baritaki, L. Militello, G. Malaponte, Y. Bevelacqua, B. Bonavida, The
Expression of hypoxia-inducible factor 1α in brain tumors: association with role of B-RAF mutations in melanoma and the induction of EMT via dysregulation
angiogenesis, invasion, and progression, Cancer: Interdiscip. Int. J. Am. Cancer of the NF-κB/Snail/RKIP/PTEN circuit, Genes Cancer 1 (5) (2010) 409–420,
Soc. 88 (11) (2000) 2606–2618. https://doi.org/10.1002/1097-0142(20000601) https://doi.org/10.1177/1947601910373795.
88:11%3C2606::AID-CNCR25%3E3.0.CO;2-W. [90] C. Li, Q. Wang, S. Shen, X. Wei, G. Li, HIF-1α/VEGF signaling-mediated
[65] S.-T. Cha, P.S. Chen, G. Johansson, C.Y. Chu, M.Y. Wang, Y.M. Jeng, S.L. Yu, J. epithelial–mesenchymal transition and angiogenesis is critically involved in anti-
S. Chen, K.J. Chang, S.H. Jee, C.T. Tan, M.T. Lin, M.L. Kuo, MicroRNA-519c metastasis effect of luteolin in melanoma cells, Phytother. Res. 33 (3) (2019)
suppresses hypoxia-inducible factor-1α expression and tumor angiogenesis, 798–807, https://doi.org/10.1002/ptr.6273.
Cancer Res. 70 (7) (2010) 2675–2685, https://doi.org/10.1158/0008-5472.can- [91] J. Mazar, F. Qi, B. Lee, J. Marchica, S. Govindarajan, J. Shelley, J.L. Li, A. Ray, R.
09-2448. J. Perera, MicroRNA 211 functions as a metabolic switch in human melanoma
[66] P. Corrie, B. Basu, K. Ahmad Zaki, Targeting angiogenesis in melanoma: prospects cells, Mol. Cell. Biol. 36 (7) (2016) 1090–1108, https://doi.org/10.1128/
for the future, Ther. Adv. Med. Oncol. 2 (6) (2010) 367–380, https://doi.org/ mcb.00762-15.
10.1177/1758834010380101. [92] Y. Zhao, C. Wu, L. Li, MicroRNA‑33b inhibits cell proliferation and glycolysis by
[67] Q. Xu, G.Q. Zhao, J. Zhao, H. Lin, Y.Y. Mou, Q. Wang, W.R. Sun, Expression and targeting hypoxia‑inducible factor‑1α in malignant melanoma, Exp. Ther. Med.
significance of factors related to angiogenesis in choroidal melanoma, Int. J. 14 (2) (2017) 1299–1306, https://doi.org/10.3892/etm.2017.4702.
Ophthalmol. 4 (1) (2011) 49–54, https://doi.org/10.3980/j.issn.2222- [93] J. Zhou, D. Xu, H. Xie, J. Tang, R. Liu, J. Li, S. Wang, X. Chen, J. Su, X. Zhou,
3959.2011.01.11. K. Xia, Q. He, J. Chen, W. Xiong, P. Cao, K. Cao, miR-33a functions as a tumor
[68] D. Su, et al., Effect of Huaier on melanoma invasion, metastasis, and angiogenesis, suppressor in melanoma by targeting HIF-1α, Cancer Biol. Ther. 16 (6) (2015)
BioMed. Res. Int. 2020 (2020) 1–8, https://doi.org/10.1155/2020/8163839. 846–855, https://doi.org/10.1080/15384047.2015.1030545.
[69] O. Simonetti, G. Lucarini, C. Rubini, G. Goteri, A. Zizzi, S. Staibano, [94] Y. Chen, Z. Zhang, C. Luo, Z. Chen, J. Zhou, MicroRNA-18b inhibits the growth of
A. Campanati, G. Ganzetti, R. Di Primio, A. Offidani, Microvessel density and malignant melanoma via inhibition of HIF-1α-mediated glycolysis, Oncol. Rep. 36
VEGF, HIF-1α expression in primary oral melanoma: correlation with prognosis, (1) (2016) 471–479, https://doi.org/10.3892/or.2016.4824.
Oral Dis. 19 (6) (2013) 620–627, https://doi.org/10.1111/odi.12048. [95] H.W. Hwang, L.L. Baxter, S.K. Loftus, J.C. Cronin, N.S. Trivedi, B. Borate, W.
[70] J.-w Kim, P. Gao, C.V. Dang, Effects of hypoxia on tumor metabolism, Cancer J. Pavan, Distinct micro RNA expression signatures are associated with melanoma
Metastas. Rev. 26 (2) (2007) 291–298, https://doi.org/10.1007/s10555-007- subtypes and are regulated by HIF 1A, Pigment Cell Melanoma Res. 27 (5) (2014)
9060-4. 777–787, https://doi.org/10.1111/pcmr.12255.
[71] W. Zeng, P. Liu, W. Pan, S.R. Singh, Y. Wei, Hypoxia and hypoxia inducible [96] Z. Xia, C. Yang, X. Yang, S. Wu, Z. Feng, L. Qu, X. Chen, L. Liu, Y. Ma, miR-652
factors in tumor metabolism, Cancer Lett. 356 (2) (2015) 263–267, https://doi. promotes proliferation and migration of uveal melanoma cells by targeting
org/10.1016/j.canlet.2014.01.032. HOXA9, Med. Sci. Monit.: Int. Med. J. Exp. Clin. Res. 25 (2019) 8722–8732,
[72] R. Courtnay, D.C. Ngo, N. Malik, K. Ververis, S.M. Tortorella, T.C. Karagiannis, https://doi.org/10.12659/msm.917099.
Cancer metabolism and the Warburg effect: the role of HIF-1 and PI3K, Mol. Biol. [97] Y. Chen, K.E. Cao, S. Wang, J. Chen, B. He, G.U. He, Y. Chen, B. Peng, J. Zhou,
Rep. 42 (4) (2015) 841–851, https://doi.org/10.1007/s11033-015-3858-x. MicroRNA-138 suppresses proliferation, invasion and glycolysis in malignant
[73] K. Adekola, S.T. Rosen, M. Shanmugam, Glucose transporters in cancer melanoma cells by targeting HIF-1α, Exp. Ther. Med. 11 (6) (2016) 2513–2518,
metabolism, Curr. Opin. Oncol. 24 (6) (2012) 650–654, https://doi.org/10.1097/ https://doi.org/10.3892/etm.2016.3220.
cco.0b013e328356da72. [98] K. Cao, J. Li, J. Chen, L. Qian, A. Wang, X. Chen, W. Xiong, J. Tang, S. Tang,
[74] G.L. Semenza, HIF-1: upstream and downstream of cancer metabolism, Curr. Y. Chen, Y. Chen, Y. Cheng, J. Zhou, microRNA-33a-5p increases radiosensitivity
Opin. Genet. Dev. 20 (1) (2010) 51–56, https://doi.org/10.1016/j. by inhibiting glycolysis in melanoma, Oncotarget 8 (48) (2017) 83660–83672,
gde.2009.10.009. https://doi.org/10.18632/oncotarget.19014.
[75] M. Nagy, HIF-1 is the commander of gateways to cancer, J. Cancer Sci. Ther. 3 (2) [99] Y. Liu, D. He, M. Xiao, Y. Zhu, J. Zhou, K. Cao, Long noncoding RNA LINC00518
(2011) 35–40, https://doi.org/10.4172/1948-5956.1000054. induces radioresistance by regulating glycolysis through an miR-33a-3p/HIF-1α
[76] D.A. Scott, A.D. Richardson, F.V. Filipp, C.A. Knutzen, G.G. Chiang, Z.A. Ronai, A. negative feedback loop in melanoma, Cell Death Dis. 12 (3) (2021), 245, https://
L. Osterman, J.W. Smith, Comparative metabolic flux profiling of melanoma cell doi.org/10.1038/s41419-021-03523-z.
lines beyond the warburg effect, J. Biol. Chem. 286 (49) (2011) 42626–42634,
https://doi.org/10.1074/jbc.m111.282046.

8
M. Malekan et al. Biomedicine & Pharmacotherapy 141 (2021) 111873

[100] H. Qiu, F. Chen, M. Chen, MicroRNA-138 negatively regulates the hypoxia- B. Van Houten, S.J. Moschos, Mitochondrial respiration-an important therapeutic
inducible factor 1α to suppress melanoma growth and metastasis, Biol. Open 8 (8) target in melanoma, PLoS One 7 (8) (2012), e40690, https://doi.org/10.1371/
(2019), bio042937, https://doi.org/10.1242/bio.042937. journal.pone.0040690.
[101] J. Wang, F. Yu, X. Jia, S. Iwanowycz, Y. Wang, S. Huang, W. Ai, D. Fan, Micro [114] A.P. Fischer, S.L. Miles, Silencing HIF-1α induces TET2 expression and augments
RNA-155 deficiency enhances the recruitment and functions of myeloid-derived ascorbic acid induced 5-hydroxymethylation of DNA in human metastatic
suppressor cells in tumor microenvironment and promotes solid tumor growth, melanoma cells, Biochem. Biophys. Res. Commun. 490 (2) (2017) 176–181,
Int. J. Cancer 136 (6) (2015) E602–E613, https://doi.org/10.1002/ijc.29151. https://doi.org/10.1016/j.bbrc.2017.06.017.
[102] X. Yang, S. Lei, J. Long, X. Liu, Q. Wu, MicroRNA-199a-5p inhibits tumor [115] Y. Zheng, H. Chen, Y. Zhao, X. Zhang, J. Liu, Y. Pan, J. Bai, H. Zhang, Knockdown
proliferation in melanoma by mediating HIF-1α, Mol. Med. Rep. 13 (6) (2016) of FBXO22 inhibits melanoma cell migration, invasion and angiogenesis via the
5241–5247, https://doi.org/10.3892/mmr.2016.5202. HIF-1α/VEGF pathway, Investig. New Drugs 38 (1) (2020) 20–28, https://doi.
[103] D. Sabry, S. El-Deek, M. Maher, M. El-Baz, H.M. El-Bader, E. Amer, E.A. Hassan, org/10.1007/s10637-019-00761-z.
W. Fathy, H. El-Deek, Role of miRNA-210, miRNA-21 and miRNA-126 as [116] J. Kluza, P. Corazao-Rozas, Y. Touil, M. Jendoubi, C. Maire, P. Guerreschi,
diagnostic biomarkers in colorectal carcinoma: impact of HIF-1α-VEGF signaling A. Jonneaux, C. Ballot, S. Balayssac, S. Valable, A. Corroyer-Dulmont,
pathway, Mol. Cell. Biochem. 454 (1–2) (2019) 177–189, https://doi.org/ M. Bernaudin, M. Malet-Martino, E.M. de Lassalle, P. Maboudou, P. Formstecher,
10.1007/s11010-018-3462-1. R. Polakowska, L. Mortier, P. Marchetti, Inactivation of the HIF-1α/PDK3
[104] R. Qin, T.C. Smyrk, N.R. Reed, R.L. Schmidt, T. Schnelldorfer, S.T. Chari, G. signaling axis drives melanoma toward mitochondrial oxidative metabolism and
M. Petersen, A.H. Tang, Combining clinicopathological predictors and molecular potentiates the therapeutic activity of pro-oxidants, Cancer Res. 72 (19) (2012)
biomarkers in the oncogenic K-RAS/Ki67/HIF-1α pathway to predict survival in 5035–5047, https://doi.org/10.1158/0008-5472.can-12-0979.
resectable pancreatic cancer, Br. J. Cancer 112 (3) (2015) 514–522, https://doi. [117] E. Licarete, A. Sesarman, V.F. Rauca, L. Luput, L. Patras, M. Banciu, HIF‑1α acts as
org/10.1038/bjc.2014.659. a molecular target for simvastatin cytotoxicity in B16. F10 melanoma cells
[105] A. Lo Dico, C. Martelli, C. Diceglie, G. Lucignani, L. Ottobrini, Hypoxia-inducible cultured under chemically induced hypoxia, Oncol. Lett. 13 (5) (2017)
factor-1α activity as a switch for glioblastoma responsiveness to temozolomide, 3942–3950, https://doi.org/10.3892/ol.2017.5928.
Front. Oncol. 8 (2018) 249, https://doi.org/10.3389/fonc.2018.00249. [118] F. Spinella, L. Rosanò, V. Di Castro, S. Decandia, M.R. Nicotra, P.G. Natali,
[106] C. Potter, A. Harris, Diagnostic, prognostic and therapeutic implications of A. Bagnato, Endothelin-1 and endothelin-3 promote invasive behavior via
carbonic anhydrases in cancer, Br. J. Cancer 89 (1) (2003) 2–7, https://doi.org/ hypoxia-inducible factor-1α in human melanoma cells, Cancer Res. 67 (4) (2007)
10.1038/sj.bjc.6600936. 1725–1734, https://doi.org/10.1158/0008-5472.can-06-2606.
[107] H. Mirzaei, S. Gholamin, S. Shahidsales, A. Sahebkar, M.R. Jaafari, H.R. Mirzaei, [119] R. Smolarczyk, T. Cichoń, E. Pilny, M. Jarosz-Biej, A. Poczkaj, N. Kułach, S. Szala,
S.M. Hassanian, A. Avan, MicroRNAs as potential diagnostic and prognostic Combination of anti-vascular agent-DMXAA and HIF-1α inhibitor-digoxin inhibits
biomarkers in melanoma, Eur. J. Cancer 53 (2016) 25–32, https://doi.org/ the growth of melanoma tumors, Sci. Rep. 8 (1) (2018) 1–9, https://doi.org/
10.1016/j.ejca.2015.10.009. 10.1038/s41598-018-25688-y.
[108] I.A. Fridman, E.A. Ponomarenko, O.V. Makarova, E.A. Postovalova, N. [120] H. Zhao, H. Jiang, Z. Li, Y. Zhuang, Y. Liu, S. Zhou, Y. Xiao, C. Xie, F. Zhou,
A. Zolotova, D.N. Khochanskiy, V.A. Mkhitarov, I.S. Tsvetkov, A.M. Kosyreva, Y. Zhou, 2-Methoxyestradiol enhances radiosensitivity in radioresistant
Morphological characteristic of melanoma B16 progression in C57BL/6 mice with melanoma MDA-MB-435R cells by regulating glycolysis via HIF-1α/PDK1 axis,
high and low resistance to hypoxia, Bull. Exp. Biol. Med. 168 (3) (2020) 390–394, Int. J. Oncol. 50 (5) (2017) 1531–1540, https://doi.org/10.3892/ijo.2017.3924.
https://doi.org/10.1007/s10517-020-04716-w. [121] M. Shah, J.L. Stebbins, A. Dewing, J. Qi, M. Pellecchia, Z.A. Ronai, Inhibition of
[109] A. Thyagarajan, A. Shaban, R.P. Sahu, MicroRNA-directed cancer therapies: Siah2 ubiquitin ligase by vitamin K3 (menadione) attenuates hypoxia and MAPK
implications in melanoma intervention, J. Pharmacol. Exp. Ther. 364 (1) (2018) signaling and blocks melanoma tumorigenesis, Pigment Cell Melanoma Res. 22
1–12, https://doi.org/10.1124/jpet.117.242636. (6) (2009) 799–808, https://doi.org/10.1111/j.1755-148x.2009.00628.x.
[110] S. Sabarimurugan, M. Madurantakam Royam, A. Das, S. Das, G. K M, R. Jayaraj, [122] R. Martí-Díaz, M.F. Montenegro, J. Cabezas-Herrera, C.R. Goding, J.N. Rodríguez-
Systematic review and meta-analysis of the prognostic significance of miRNAs in López, L. Sánchez-del-Campo, Acriflavine, a potent inhibitor of HIF-1α, disturbs
melanoma patients, Mol. Diagn. Ther. 22 (6) (2018) 653–669, https://doi.org/ glucose metabolism and suppresses ATF4-protective pathways in melanoma
10.1007/s40291-018-0357-5. under non-hypoxic conditions, Cancers 13 (1) (2021), 102, https://doi.org/
[111] E.-J. Park, Y.M. Lee, T.I. Oh, B.M. Kim, B.O. Lim, J.H. Lim, Vanillin suppresses cell 10.3390/cancers13010102.
motility by inhibiting STAT3-mediated HIF-1α mRNA expression in malignant [123] K. Patra, S. Jana, A. Sarkar, D.P. Mandal, S. Bhattacharjee, The inhibition of
melanoma cells, Int. J. Mol. Sci. 18 (3) (2017), 532, https://doi.org/10.3390/ hypoxia-induced angiogenesis and metastasis by cinnamaldehyde is mediated by
ijms18030532. decreasing HIF-1α protein synthesis via PI3K/Akt pathway, Biofactors 45 (3)
[112] Y. Liu, J. Tao, Y. Li, J. Yang, Y. Yu, M. Wang, X. Xu, C. Huang, W. Huang, J. Dong, (2019) 401–415, https://doi.org/10.1002/biof.1499.
L. Li, J. Liu, G. Shen, Y. Tu, Targeting hypoxia-inducible factor-1α with [124] Y.-Q. Tang, I.B. Jaganath, R. Manikam, S.D. Sekaran, Inhibition of MAPKs, Myc/
Tf–PEI–shRNA complex via transferrin receptor–mediated endocytosis inhibits Max, NFκB, and hypoxia pathways by Phyllanthus prevents proliferation,
melanoma growth, Mol. Ther. 17 (2) (2009) 269–277, https://doi.org/10.1038/ metastasis and angiogenesis in human melanoma (MeWo) cancer cell line, Int. J.
mt.2008.266. Med. Sci. 11 (6) (2014) 564–577, https://doi.org/10.7150/ijms.7704.
[113] M. Barbi de Moura, G. Vincent, S.L. Fayewicz, N.W. Bateman, B.L. Hood, M. Sun,
J. Suhan, S. Duensing, Y. Yin, C. Sander, J.M. Kirkwood, D. Becker, T.P. Conrads,

You might also like