Next Article in Journal
The Immunopathology of Pulmonary Rejection after Murine Lung Transplantation
Previous Article in Journal
Novel Vitamin D3 Hydroxymetabolites Require Involvement of the Vitamin D Receptor or Retinoic Acid-Related Orphan Receptors for Their Antifibrogenic Activities in Human Fibroblasts
Previous Article in Special Issue
Autophagy and Apoptosis in Rabies Virus Replication
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

CD133-Dependent Activation of Phosphoinositide 3-Kinase /AKT/Mammalian Target of Rapamycin Signaling in Melanoma Progression and Drug Resistance

by
Naji Kharouf
1,2,
Thomas W. Flanagan
3,
Abdulhadi A. Alamodi
4,
Youssef Al Hmada
5,
Sofie-Yasmin Hassan
6,
Hosam Shalaby
7,
Simeon Santourlidis
8,
Sarah-Lilly Hassan
9,
Youssef Haikel
1,2,10,
Mossad Megahed
11,
Robert T. Brodell
5 and
Mohamed Hassan
1,2,12,*
1
Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France
2
Department of Operative Dentistry and Endodontics, Dental Faculty, University of Strasbourg, 67000 Strasbourg, France
3
Department of Pharmacology and Experimental Therapeutics, LSU Health Sciences Center, New Orleans, LA 70112, USA
4
College of Health Sciences, Jackson State University, Jackson, MS 39213, USA
5
Department of Pathology, University of Mississippi Medical Center, Jackson, MS 39216, USA
6
Department of Pharmacy, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Dusseldorf, Germany
7
Department of Urology, School of Medicine, Tulane University, New Orleans, LA 70112, USA
8
Epigenetics Core Laboratory, Institute of Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany
9
Department of Chemistry, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Dusseldorf, Germany
10
Pôle de Médecine et Chirurgie Bucco-Dentaire, Hôpital Civil, Hôpitaux Universitaire de Strasbourg, 67000 Strasbourg, France
11
Clinic of Dermatology, University Hospital of Aachen, 52074 Aachen, Germany
12
Research Laboratory of Surgery-Oncology, Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
*
Author to whom correspondence should be addressed.
Cells 2024, 13(3), 240; https://doi.org/10.3390/cells13030240
Submission received: 29 November 2023 / Revised: 24 January 2024 / Accepted: 25 January 2024 / Published: 26 January 2024
(This article belongs to the Special Issue PI3K/AKT/mTOR Signaling Network in Human Health and Diseases 2.0)

Abstract

:
Melanoma frequently harbors genetic alterations in key molecules leading to the aberrant activation of PI3K and its downstream pathways. Although the role of PI3K/AKT/mTOR in melanoma progression and drug resistance is well documented, targeting the PI3K/AKT/mTOR pathway showed less efficiency in clinical trials than might have been expected, since the suppression of the PI3K/mTOR signaling pathway-induced feedback loops is mostly associated with the activation of compensatory pathways such as MAPK/MEK/ERK. Consequently, the development of intrinsic and acquired resistance can occur. As a solid tumor, melanoma is notorious for its heterogeneity. This can be expressed in the form of genetically divergent subpopulations including a small fraction of cancer stem-like cells (CSCs) and non-cancer stem cells (non-CSCs) that make the most of the tumor mass. Like other CSCs, melanoma stem-like cells (MSCs) are characterized by their unique cell surface proteins/stemness markers and aberrant signaling pathways. In addition to its function as a robust marker for stemness properties, CD133 is crucial for the maintenance of stemness properties and drug resistance. Herein, the role of CD133-dependent activation of PI3K/mTOR in the regulation of melanoma progression, drug resistance, and recurrence is reviewed.
Keywords:
melanoma; CSCs; CD133; PI3K; AKT; mTOR

1. Introduction

Human malignant melanoma is a highly aggressive skin cancer characterized by its heterogeneity, propensity to metastasize to distant organs, and the potential for developing resistance to conventional and even the newest targeted therapeutics as measured by progression-free and overall survival [1,2,3]. As a heterogeneous tumor, malignant melanoma exists in the form of genetically divergent subpopulations containing melanoma initiating cells/cancer stem-like cells (CSCs) as a small fraction, and non-cancer stem cells (non-CSCs) that form most of the tumor mass [4,5]. Like other CSCs, melanoma stem-like cells (MSCs) are characterized by their unique surface proteins and aberrant signaling pathways [4,5,6], which are either in a causal or consequential relationship to melanoma progression, treatment resistance, and recurrence [5,6,7,8]. CD133 (prominin-1) is one of the most important cancer stem cells (CSCs) that is widely expressed in the CSC subpopulation derived from a large variety of human malignancies, including melanoma [5,6,8]. Beyond its role as a reliable CSC marker for the identification of CSC populations [7,8], accumulating evidence has indicated that CD133 is responsible for CSC tumorigeneses and chemoresistance [4,8].
Aberrant activation of the phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways play a significant role in melanoma development, progression, and drug resistance. Melanoma is characterized by its heterogeneity as a consequence of genetic and non-genetic alterations that are frequently associated with the activation of the PI3K and MAPK pathways [9,10,11]. Although the dual inhibition of BRAF and MEK by their specific inhibitors has demonstrated significant treatment successes in advanced melanoma in patients with the BRAF mutation [12,13], most patients showed only short response durations along with the development of both intrinsic and acquired resistance [14]. The preclinical investigation of PI3K inhibitors as a monotherapy showed no significant advances in clinical trials [15,16]. The limited therapeutic efficiency of the inhibitors of PI3K pathway seems to result from the suppression of feedback loops mediated by PI3K/mTOR leading to the activation of compensatory pathways such as MAPK/MEK/ERK [17,18].
Melanoma treatment remains a major challenge in clinical oncology. Poor prognosis, disease progression, and drug resistance in melanoma are closely associated with the activation of PI3K [19,20]. Thus, targeting the PI3K/AKT/mTOR and MAPK/MEK/ERK pathways is a promising therapeutic approach that might improve treatment outcomes.
The main function of PI3K is to phosphorylate inositol-containing lipids (PtdIns) at the D3 OH group of the inositol ring. Consequently, the generation of PtdIns (3)P in the inner leaflet of membrane bilayers facilitates the recruitment of cytosolic proteins to initiate multi-functional signaling cascades [21].
Three different classes of PI3K have been identified [22,23]. Although the three PI3K classes, namely class I, class II, and class III, are similar in structure, they differ in their function. For example, class I of PI3Ks is directly involved in signaling downstream of plasma membrane-bound receptors, whereas the two other classes are involved in the regulation of vesicular trafficking [22].
The activation of PI3Ks is regulated by receptor–ligand ligation that in turn mediates the internalization of signaling receptors from the plasma membrane into early endosomes, where the receptors can be sorted into late endosomes/lysosomes for either degradation or recycling back to the plasma membrane [24,25]. Functionally, the PI3K class I is involved in the phosphorylation of PtdIns P2 to yield PtdIns P3 that is frequently altered in cancer, thereby serving as a potentially important therapeutic target [26,27,28].
Investigators have primarily focused on the heterodimer PI3K. It is composed of a catalytic subunit (P110) and a regulatory subunit (P85). The regulatory subunits contain non-receptor tyrosine kinase Src Homology 2 (SH2) and 3 (SH3) domains [29,30]. These SH2 and SH3 domains are characterized by their ability to interact with target proteins containing tyrosine kinase-binding sites [29,30].
It is notable that the activation of PI3K in many cancer types, including melanoma, is mostly associated with tumor development, progression, and drug resistance [31,32,33,34,35]. Melanoma frequently harbors genetic alterations in the key molecules of the PI3K pathway that can drive the aberrant activation of the PI3K pathway and its downstream pathways [27,36].
The conserved serine/threonine kinase mTOR, the mammalian target of rapamycin, is a downstream effector of the PI3K/AKT pathway and forms two distinct multiprotein complexes, mTORC1 and mTORC2 [9,10]. mTORC1 is sensitive to rapamycin and activates S6K1 and 4EBP1, both of which are involved in mRNA translation [37,38]. The activation of mTOR is mediated by diverse stimuli including growth factors, nutrients, energy, and stress signals, as well as signaling pathways, such as PI3K and MAPK [37,38].
Although the role of PI3K/AKT/mTOR in melanoma progression and drug resistance is well described, the inhibitors of the PI3K/AKT/mTOR pathway have demonstrated only limited success in clinical trials, particularly when applied as a monotherapy [35,36,37]. This shortcoming of PI3K/AKT/mTOR inhibitors as therapeutic targets in melanoma treatment is attributed to feedback loops derived from the inhibition of PI3K/mTOR leading to the activation/reactivation of aberrant signaling pathways like RAS/RAF/MEK/ERK [39,40].
This review provides insight into the role of CD133 signaling in the PI3K/mTOR pathway which can lead to melanoma progression, drug resistance, and recurrence, and the impact of the CD133/PI3K/AKT/mTOR pathway as a therapeutic target for melanoma treatment.

2. Melanoma Heterogeneity and Plasticity

Melanocytes are of neuroectodermal origin. They have the potential to migrate throughout the body and ultimately are localized in the skin, uveal tract, mucosa, inner ear, and rectum [41,42]. Based on the wide distribution of melanocytes, the development of melanoma can occur even outside these more common areas.
Despite access to early diagnostics and increasingly available access to primary care, both melanoma incidence and mortality rates are increasing worldwide. Current reports reveal that around 1.7% of all newly diagnosed skin cancers are melanomas, while patients dying from melanoma account for 75% of skin cancer deaths and nearly 0.7% of all cancer mortality [41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65]. Incidence rates differ significantly across the globe with the highest rates in Australia, New Zealand, Europe, and North America; the lowest incidence rate has been noted in Africa [46,47]. While the geographical dependency of melanoma development has been widely established, ethnicity, lifestyle (UV exposure), and genetic background are the most significant factors contributing to the development of melanoma [48,49].
Tumor heterogeneity is a significant challenge and is the main cause of drug resistance which ultimately leads to therapeutic failure [50,51,52,53,54]. It affects the therapeutic targets and shapes the tumor microenvironment influencing drug resistance [50]. Tumor heterogeneity is a tumor phenomenon that occurs spatially and temporally during tumor development with the aim of continuously triggering the reprogramming of the tumor microenvironment [50].
Tumor heterogeneity is attributed to the presence of subpopulations of cells that differ in their phenotype and biological behavior [56,57]. These subpopulations can exist within a tumor in the form of intra-tumoral or inter-tumoral heterogeneity. These two types of tumoral heterogeneity can occur in the same histopathological tumor subtype within a patient (intra-tumoral heterogeneity) or in the same tumor in different patients (inter-tumoral heterogeneity) [57,58].
The development of tumor heterogeneity in various cancer types including melanoma is mediated by genomic- (intrinsic factors) and non-genomic (extrinsic factors)-dependent mechanisms. Genomic tumor heterogeneity-dependent mechanisms are derived from significant alterations in the intrinsic factors including genome, transcriptome, epigenome, and proteome [60,61,62,63,64,65]. Consequently, melanomas are among the most common heterogeneous cancers. For example, melanomas are characterized by diverse genomic alterations in the form of functional mutations to NRAS, BRAF, KIT, CDK4, and MITF, loss of CDKN2A, PTEN, ARID2, and NF expression, and epigenetic changes to PTEN, CDKN2A, RAC1, and P53 genes [16,66,67]. In addition, the development of tumor heterogeneity in melanoma is the result of differential expression in the same tumor to various functional proteins, such as stem cell markers CD20, CD133, ABCB5, CD271, JARID1B, and ALDH1 [68,69,70].
Non-genomic (extrinsic factors) tumor heterogeneity-dependent mechanisms are the consequence of tumor-associated inflammation, and crosstalk between the tumor and its microenvironment [70,71]. Non-genomic/phenotypic heterogeneity demonstrates phenotypic variability between tumor subpopulations, which share the same genome, while trait differences among these tumor subpopulations are not related to the genetic differences between the identified tumor subpopulations [71,72,73]. In contrast to intrinsic factor-dependent mechanisms, the extrinsic factor-mediated tumor heterogeneity includes pH, hypoxia, and crosstalk between tumor cells and the components of the tumor microenvironment, particularly in association with stromal cells [50,71,72].
The contribution of inflammation to the development of tumor heterogeneity has been reported in several studies [74,75]. Inflammation is caused by a variety of pathogenic and environmental factors which lead to the induction of several mechanisms in the form of oxidative stress, upregulation of hypoxia inducible factor 1-alpha, and the production of pro-inflammatory cytokines [74,75,76,77].
The mechanisms involved in the development of phenotypic and functional heterogeneity of melanoma including genomic and non-genomic-dependent mechanisms are summarized in Figure 1.
In contrast to melanoma heterogeneity, the development of plasticity in melanoma cells is attributed to significant alterations in the expression profile of stromal and immune cells, and changes in the extracellular matrix of the melanoma microenvironment [75,78]. Cancer cell plasticity describes the ability of cancer cells to shift dynamically between differentiated and undifferentiated states to promote long-term tumor cell growth and transition into CSCs.
Melanoma cells have the potential to switch their phenotype during tumor progression from a proliferative and differentiated phenotype to a more invasive and dedifferentiated phenotype [79,80]. Genetic alterations, influences from the tumor microenvironment, and epigenetic changes belong to the phenotypic plasticity and high heterogeneity that is known to be characteristic of melanoma [81]. Melanoma cells continuously undergo reversible alteration between a proliferative/differentiated and an invasive/dedifferentiated phenotype, an epithelial-to-mesenchymal transition (EMT)-like process [82,83]. Thus, the transition of melanoma cells into an invasive phenotype facilitates melanoma dissemination from a primary tumor to distant sites [84,85]. However, phenotype alteration is mediated mainly by a hypoxic tumor microenvironment and inflammatory signal-dependent mechanisms [85,86,87,88,89].
In addition to the reversible alteration between a proliferative/differentiated and an invasive/dedifferentiated phenotype [79,90,91], occurrence of melanoma cell plasticity is mediated by both cell-autonomous mechanisms and tumor microenvironment-dependent signals [90,92]. Accordingly, cancer cell/stromal cell-dependent mechanisms can impact the regulation of the phenotypic plasticity of melanoma cells [90,91,92,93,94,95]. For example, cancer-associated fibroblasts (CAFs), the major component in the tumor microenvironment, have been shown to play key roles in the regulation of melanoma cell plasticity via hepatocyte growth factor (HGF)-dependent mechanism [96,97] and insulin-like growth factor receptor signaling-dependent mechanisms [98].
Thus, understanding the biological complexity of cancer cell plasticity and its role in melanoma progression and relapse may lead to the development of new therapeutic approaches for the treatment of melanoma. Also, the functional analysis of specific cell markers and key molecules of aberrant signaling pathways is urgently needed, particularly those closely associated with the maintenance of stemness properties and the regulation of the cross talk between melanoma cells and their microenvironment. The mechanisms regulating melanoma plasticity are outlined in Figure 2.

3. Melanoma Stem Cells

The processes of melanoma development start in mature melanocytes [99,100]. Although the accumulated evidence indicates that early-stage precursors of melanocytes exist in the dermis [101,102], other reports have suggested that the earliest origins of cutaneous melanoma may have occurred in extrafollicular melanocyte stem cells [100,101,102]. This is probably because the melanoma stem cells (MSCs) are derived either from a transformed melanocyte, from a transformed melanocyte stem cell, or from a combination of both sources. Like CSCs, MSCs are characterized by their ability to self-renew and differentiate [103,104,105]. The generation of these subpopulations is mediated by genetic segregation and epigenetic alterations via the transcriptional regulation of genes associated with stemness properties and dysregulation of aberrant signaling pathways such as FOXM1 signaling [106,107,108]. Consequently, melanoma cells undergo intrinsically asymmetric cell divisions of stem cell lineage to produce two daughter cells, both significantly different in their genetic material and phenotype [109,110,111]. One of these daughter cells is characterized by its stemness properties and is referred to as CSC, whereas the other daughter cell (non-CSC) represents a larger portion of the overall tumor mass [4,5,9,110,111]. In addition to the deregulation of aberrant signaling pathways for tumor growth and survival, MSCs are notable for their expression of CD20, CD133, CD166, CD271, ABCB5, Nestin, and CD105 surface markers [4,5,8]. Thus, understanding the pathways controlling self-renewal, expansion, and differentiation of MSCs and how UV radiation alters and disrupts melanocyte lineage pathways will bring greater insight into the origin of melanomas. The development of CSCs from normal stem/progenitor cells (Figure 3A) and from cancer cells (Figure 3B) is outlined in detail in Figure 3.
Beyond their identification in various malignancies as part of the tumor mass, CSCs are characterized by their ability to confer self-renewal, differentiation, tumor initiation, metastasis, recurrence, and drug resistance [5,6,8,112]. Like CSCs of different tumor types, MSCs have been functionally characterized in vitro and in vivo [5,6,9,100,102]. In addition to their stemness properties, MSCs demonstrate the expression of stem cell markers including CD20, CD105, CD133, CD146, CD166, CD271, ABCB5, and Nestin [5]. Apart from their function as stem cell marker proteins, such as CD133, the proteins have also been discussed for their functional role in the regulation of MSCs maintenance and resistance [8].

4. CD133

The stem cell marker CD133 (prominin-1/AC133) has a molecular weight of 120 kDa and is encoded by the PROM1 gene [113,114]. It is a member of pentaspan transmembrane glycoproteins [114]. CD133 localizes specifically to cellular membranes with an extracellular N-terminal domain, 5-transmembrane domains separating two large glycosylated extracellular loops, two small intracellular loops, and an intracellular C-terminal domain [114,115]. The involvement of the CD133 protein in the maintenance of melanoma stemness properties and drug resistance is mediated through its C-terminal domain, which contains tyrosine binding sites located on tyrosine 828 (Tyr828) and tyrosine 852 (Tyr852) residues [9,116]. These two tyrosine residues are phosphorylation targets of the non-receptor tyrosine kinase (NRTK) Fyn [116]. The structure and the various domains of CD133 protein are illustrated in Figure 4.
Many studies have demonstrated that increased CD133 expression is associated with high tumorigenicity and metastatic potential for melanoma cells [117,118,119]. Also, CD133 protein has been implicated in the regulation of tumor resistance [120,121,122].
CD133-expressing CSCs have been shown to exhibit resistance to chemotherapy and radiation therapy in addition to being associated with poor prognosis in various cancers [122]. We and others have demonstrated that CD133+ cancer cells confer resistance to many chemotherapeutic agents such as caffeic acid phenethyl ester [5], Taxol [6], and fotemustine [8]. Accordingly, CD133-dependent mechanisms have been shown to be involved in the development of melanoma resistance to chemotherapy [8].
The contribution of CD133 to the regulation of CSC functions such as self-renewal, differentiation, and drug resistance are likely mediated by the NRTK, Fyn-dependent mechanism via the phosphorylation of the Tyr828 residue located on the cytoplasmic domain of CD133 [8,116]. Our laboratory has demonstrated that the phosphorylation of Tyr828 is essential for triggering the activation of PI3K and its downstream dependent signaling pathways in melanoma [8].
The PI3K/AKT pathway is one of the most important networks with the highest mutation frequency in human cancers [123,124,125]. Both PI3K/AKT/NF-κB and PI3K/AKT/mTOR are the two main mutated pathways involved in apoptosis and tumorigenesis facilitating the development of melanoma resistance to anti-cancer agents [126]. Dysregulation of major key molecules in these signaling pathways is associated with drug resistance and melanoma progression. Also, the elevated activation of PI3K pathway has been suggested as triggering melanoma progression through the activation of the PI3K/AKT/NF-κB axis [27,127,128].
In addition to the key role of CD133 in chemoresistance, we and others have demonstrated the cellular mechanisms by which the CD133 protein triggers activation of the PI3K pathway both in melanoma [8].
The PI3K/AKT/mTOR signal to downstream proteins leads to the development of tumor resistance [129,130]. Also, the PI3K/AKT/mTOR pathway has been shown to play a crucial role in a variety of biological and physiological processes including cell survival and growth, and transcription and translation, which are implicated in the development of drug resistance [18,33,131]. Abnormal activation of the PI3K/AKT/mTOR pathway in different tumor types including melanoma has been suggested as the key mechanism through which tumors evade drug toxicity [126,132,133]. Thus, CD133 mediates the activation of the PI3K/AKT pathway in melanoma [8,116]. The activation of mTORC1 through the PI3K-dependent activation of both AKT and PDK-1 has been reported in CD133 positive cells [8,116].
In addition to the frequent mutation to AKT family members, mutations to the PI3K/AKT/mTOR pathway are common in melanoma [33,34,35]. Mutation in PTEN has been shown to effectively restrain the PI3K/AKT/mTOR growth-promoting signaling cascade in primary and metastatic melanoma patients [131,132,133,134,135,136]. Also, the activation of PI3K/AKT results in the activation of mTORC1 that subsequently leads to the phosphorylation of the downstream molecules, p70S6K1, and eukaryotic initiation factor 4EBP1 to affect mRNA translation and protein synthesis [132,137]. The role of the Fyn-stimulated CD133 signal to PI3K is considered a key mechanism in the regulation of both PI3K/AKT and PI3K/NF-κB pathways. To that end, Fyn-stimulated CD133 signaling to PI3K is expected to trigger the activation of the mTOR and the associated biological consequences in melanoma. In addition, the references describing the functional role of Fyn-stimulated CD133 signal in melanoma have been mentioned (Table 1). The mechanisms are thought to be mediated by an Fyn-stimulated CD133 signal leading to the activation of PI3K and its downstream pathways, as shown in the proposed model (Figure 5).

5. Non-Receptor Tyrosine Kinases

In contrast to receptor tyrosine kinases (RTKs), non-receptor tyrosine kinases (NRTKs) do not possess either an extracellular ligand-binding domain or a transmembrane domain-spanning region [138,139,140]. Therefore, NRTKs are either localized in the cytoplasm [136], or anchored to the cell membrane through their amino terminal modification [141], or are underlying nuclear translocation [142]. NRTKs are characterized by their modular construction enzymes and individual domains that are connected by disordered regions in the form of loop connecting domains [140]. The catalytic domain of the NRTKs is of critical importance and is about 275 residues in length [143,144]. The structure of the catalytic domain is organized in two lobes. The small lobe functions as a binding site for ATP, whereas the large one functions as a binding site for the protein substrate [145]. The binding of both ATP and substrate to the corresponding domains catalyzes phosphate transfer in the cleft between the small and the large lobes [146]. In addition to their enzyme activity, NTRKs are characterized by their sequence preference around the target tyrosine [147]. The most Src-preferred sequence is Glu–Glu/Asp–Ile–Tyr–Gly/Glu–Glu–Phe, while the Abl-preferred sequence is Ile/Val–Tyr–Gly–Val–Leu/Val [148,149]. The difference in the preferred sequences around tyrosine in Src and Abl is evidence of their specific target substrates. NRTKs contain not only a tyrosine kinase domain, but also possess other domains that can mediate protein–protein, protein–lipid, and protein–DNA interactions [144,150]. Key protein–protein interaction domains in NRTKs are the Src homology 2 (SH2) and 3 (SH3) domains. SH2 is the longer domain of 100 residues and is characterized by the ability to bind, in a sequence-specific manner, phosphotyrosine (pTyr) but not unphosphorylated tyrosine (Tyr) residues [150,151,152,153]. In contrast to the SH2 domain, the SH3 domain is a small domain with 60 residues and is characterized by its ability to bind proline-containing sequences to form polyproline type II helixes [140,141,145]. While most NRTKs possess SH domains, there are also some that lack SH2 and SH3 domains [154,155]. These NRTKs possess some subfamily-specific domains that are essential for protein–protein interactions [156]. Among these subfamilies are the specific domains that target enzymes to the cytoplasmic part of the cytokine receptors [157,158], such as the JAK family, the integrin-binding domain, and the focal adhesion-binding domain (FAK family) [115,159]. The NRTK Abl also contains additional interaction domains including the F actin-binding domain and DNA-binding domain that contains a nuclear localization signal and exists both in the nucleus and cytoplasm [160,161,162]. Conversely, in addition to SH2 and SH3 domains, the NRTKs Btk/TEC subfamily possesses a pleckstrin homology (PH) domain [163,164]. PH domains are characterized by their ability to bind to phosphatidylinositol (PI) lipids [165,166].
PI lipids are key players in many trafficking and signaling pathways [167,168]. The phosphorylation of PI lipides is mediated primarily by a family of lipid kinases, the phosphatidylinositol-3-kinases (PI3Ks) [169,170,171].
PI3Ks are a family of enzymes that contain important cellular signaling regulators [18,172]. The activation of PI3K is mediated by either G-protein-coupled receptors or receptors with an intrinsic/associated protein tyrosine kinase activity in the form of an extracellular stimuli-dependent mechanism [173,174]. In addition to their activation by the direct interaction with the small GTPase Ras, the activation of PI3K by an NRTK-dependent mechanism has also been demonstrated in melanoma [175]. The structure of various NRTK families and their functional domains is illustrated in Figure 6.

6. Non-Receptor Tyrosine Kinase Fyn

Fyn is a tyrosine-specific phospho-transferase that is a member of the large Src family of NRTKs. While the formal crystal structure of the full-length Fyn protein has not been described, the mode of regulation of Fyn tyrosine kinase activity is like the Src family kinases [175,176]. Fyn is a 59 kDa protein comprised of 537 amino acids encoded by the Fyn gene that can be spliced to produce three isoforms [177]. The first identified isoform is isoform 1 (Fyn[B]); isoform 2 (Fyn [T]) is highly expressed in T-cells and differs from the isoform in the linker region between the SH2 and SH1 domains [178]. Isoform 3 has been detected in blood cells and differs from isoform 1 via the absence of sequence 233–287 [179,180]. Like members of the Src family, Fyn shares the conserved structure that consists of consecutive SH1, SH2, and SH3 domains (Figure 7). The SH1 domain is the catalytic tyrosine kinase, while the SH2 domain binds to tyrosine-phosphorylated substrates. Specifically, the SH2 domain of Fyn binds the phosphorylated tyrosine Y528 residue in its carboxyl terminal tail under basal conditions in vivo [116,181]. Repression of Fyn kinase activity is achieved via intra-molecular interactions between the SH3 domain and a polyproline type II linker helix that connects the SH2 and the SH1 domains. For the Fyn kinase, the tyrosine Y528 negative regulatory site is phosphorylated by C-terminal src Kinase (CSK), a cytoplasmic protein–tyrosine kinase [181]. FCSK homology kinase (CHK) is a second enzyme that catalyzes the phosphorylation of this inhibitory tyrosine Y528 [182]. CHK binds Src family members with a high affinity, independent of CHK catalytic activity, which may be sufficient to inhibit Src family kinase activity [183,184]. The dephosphorylation of the Y528 residue by protein tyrosine phosphatases rPTPα, SHP1/2, PTP1B, PTPε, and CD45 can release the SH2 domain and activate the enzyme [181,185,186]. In addition, the subfamily composed of Fyn, Src, and Lyn kinases contains dual acylation sites in the amino-terminal SH4 domain, which is thought to be partially responsible for lipid raft micro-domain association [181,186,187,188].
As is widely documented, the Src family kinase/focal adhesion kinase (FAK) complex is a signaling platform that is known to play a crucial role in the regulation of oncogenic growth factor receptor-dependent downstream pathways [189,190]. This observation is described more in the case of melanoma in Xiphophorus fish, in which the oncogenic EGF receptor orthologue Xiphophorus melanoma receptor kinase (Xmrk) effects the continuous activation of the Src family kinase Fyn that is strongly involved in promoting many tumorigenic events [190,191,192].
The Xiphophorus fish, which is derived from the crosses between X. maculatus (the southern platyfish) and X. hellerii (the green swordtail) species of the fish genus Xiphophorus [191,192], can spontaneously develop malignant melanoma via a proto-oncogene encoding a receptor tyrosine kinase-designated Xmrk-dependent mechanism [191]. The encoded Xmrk protein is structurally related to the human EGFR with an extracellular ligand-binding domain, a transmembrane domain, and intracellular.
The melanoma formation in Xiphoporus is initiated by overexpression of the EGFR-related receptor tyrosine kinase Xmrk. This receptor is activated in fish melanoma as well as in a melanoma-derived cell line (PSM) resulting in constitutive Xmrk-mediated mitogenic signaling. The elevated expression of Xmrk is the initial cell-type-specific event in melanoma formation in Xiphoporus [110]. Xmrk-mediated transformation potential is cell-type-specific signal transduction-dependent mechanism [193]. The NRTK Fyn has been identified as a substrate of Xmrk in Xiphoporus melanoma cells [181,194,195]. In addition, Xmrk has been reported to contain binding sites for growth factor receptor-bound protein 2 (GRB2), Src homology and collagen (Shc), Fyn, and PLCg [196]. Also, the binding of Grb2 to the activated Xmrk has been shown to trigger activation of the MAP kinase pathway [194]; meanwhile, the binding of Fyn to the Xmrk receptor is mediated through its SH2 domain [195].
FYN is highly expressed in many cancers and promotes cancer growth and metastasis through diverse biological functions such as cell growth, apoptosis, and motility migration, as well as the development of drug resistance in many tumors [12,151,177,178,179,180,181]. In addition, FYN is involved in the regulation of multiple cancer-related signaling pathways, including interactions with ERK, COX-2, STAT5, MET, and AKT [196]. FYN is therefore an attractive therapeutic target for various tumor types, and suppressing FYN can improve the prognosis and prolong the life of patients.

7. Receptor and Non-Receptor Tyrosine Kinase-Mediated Pathways to Melanoma Progression and Drug Resistance

In human melanomas, both PI3K/PTEN/AKT and RAS/RAF/MEK/ERK signaling pathways are the key routes whose constitutive activation results from genetic alteration [19,20]. Frequent mutations in RAF, RAS, and PTEN are largely associated with defects in cell death machinery, abnormal proliferation, angiogenesis, and invasion and thereby contribute in great part to melanoma progression and drug resistance [19,20,137,197].
The PI3K signaling pathway is one of the most frequently altered pathways in human cancer leading to oncogenic transformation, tumor initiation, and progression, in addition to the regulation of tumor apoptosis and autophagy [22,23,24]. Meanwhile, the PI3K and its lipid product phosphatidylinositol-3,4,5-trisphosphate (PIP3) are involved in the activation of multiple downstream signaling proteins [198]. Of note, the protein kinase AKT is the most studied downstream effector protein of PI3K [72,199,200].
The discovery of PI3K-dependent downstream effector signaling molecules such as the PDK1/mTORC2/SGK axis underscores the importance of PI3K in the regulation of a variety of cellular functions [200,201]. The PI3K/PDK1/mTOR/SGK pathway has been shown to compensate for the function of the PI3K/AKT pathway in the promotion of tumor survival, progression, migration, and drug resistance [20,82,202].
The target of rapamycin (TOR) was first identified in the budding yeast Saccharomyces cerevisiae [203]. The structurally and functionally conserved mammalian counterpart, mTOR, was identified based on the biochemical and inhibitory properties of rapamycin in mammalian cells [9,204,205,206].
mTOR includes two functionally distinct protein complexes, namely mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2) [207]. mTORC1 is composed of mTOR, raptor, mLST8, and two negative regulators, PRAS40 and DEPTOR [207,208]. Meanwhile, mTORC2 is composed of the conserved mTOR, RICTOR (mAVO3), mSin1, and mLST8 (GβL), as well as less conserved proteins such as PRR5/Proctor, PRR5L, and DEPTOR [207,209,210].
The PI3K-Akt-mammalian target of rapamycin (mTOR) pathway is intracellular and is aberrantly upregulated in different tumor types, including melanoma [124]. The PI3K/AKT and PI3K/AKT-mTOR pathways are essential in the regulation of biochemical and biological processes both in normal and cancer cells [131,208,209,210,211,212]. Dysregulation of these pathways is frequently associated with genetic/epigenetic alterations and poor treatment outcomes in a variety of human cancers including melanoma [209,210].
The activation of PI3K through an NRTK/Fyn-induced CD133 signal has been reported both in melanoma [116] and glioma cells [211]. The phosphorylation of tyrosine kinase Tyr828 and Tyr852 residues by NRTK Fyn was found to trigger downstream pathways of PI3K, including PI3K/AKT/MDM2 and PI3K/AKT/MKP-1 [8]. Activation of the PI3K/AKT/MDM2 pathway results in the destabilization of the p53 protein, while the activation of the PI3K/AKT/MKP-1 pathway results in the inhibition of mitogen-activated protein kinases (MAPKs) JNK and p38, and the activation of both PI3K/AKT/MDM2 and PI3K/AKT/MKP-1 pathways leads to the inhibition of fotemustine-induced apoptosis [8]. Although the characterization of Fyn-stimulated CD133/PI3K/mTOR is not described in human melanoma, the mechanism of Fyn-stimulated CD133 signal to the PI3/AKT axis seems to be a general mechanism in solid tumors [116,211]. However, the activation of PI3/AKT is expected to mediate the Fyn-stimulated CD133 signal to mTOR in the form of generating an Fyn/CD133/AKT/mTOR axis. Thus, targeting the Fyn/CD133/AKT/mTOR axis likely possesses a therapeutic impact for melanoma treatment. Figure 8 demonstrates a proposed model for both RTK- and NRTK-mediated signaling pathways, PI3K/AKT, PI3K/AKT/mTOR, and RAS/RAF/MEK/ERK, in different tumor types including melanoma.
The activation of mTORC1 is mediated by the inactivation of both TSC1 and TSC2 following their phosphorylation via the PI3K/AKT pathway [205]. In addition to its functional role in the regulation of cell growth, proliferation, and survival in response to sensing mitogen, energy, and nutrient signals, the mTORC1 is involved in the regulation of many functional proteins including the regulation of eukaryotic elongation factor 2 (eEF2) kinase [212,213], CLIP-170 (cytoplasmic linker protein-170) [214], ornithine decarboxylase (ODC) [215], hypoxia-inducible factor 1α (HIF-1α) [216], protein phosphatase 2A (PP2A) [217], lipin [218], PKCδ and PKCε [219], protein phosphatase 2A (PP2A) [2], p21Cip1and p27Kip1cyclin-dependent kinase inhibitors [220,221], and retinoblastoma protein (Rb) [222].
The second complex of mTOR, namely mTORC2, has been demonstrated as playing an important role in the promotion of cancer cell survival, proliferation, growth, and motility based on its ability to enhance the phosphorylation of AktSer473, the key regulator of the insulin/PI3K pathway [9,223]. To that end, the permanent activation of AKT is associated with feedback reactivation of mTORC2 [224]. Thus, targeting mTORC1 and/or mTORC2 may have a therapeutic impact on the treatment of different tumor types, including melanoma. The mechanisms of RTK-mediated signaling to the PI3K/AKT/mTOR pathway and its biological consequences in melanoma cells are outlined in Figure 9.

8. Melanoma Progression and Drug Resistance Are Attributed to Dysregulation of PI3K/AKT/mTOR Pathways

Dysregulation of the PI3K/AKT/mTOR pathways contributes to the pathogenesis of melanoma [210,225]. The initiation of primary melanoma occurs as horizontal lesions that appear in the form of a thin plaque on the epidermis [226], and therefore is known as the radial growth phase (RGP) that can proceed to form the vertical growth phase (VGP) characterized by its invasiveness and propensity to metastasize to distant organs. The transition of RGP into VGP has been reported to be mostly associated with AKT activation, since the occurrence of melanoma metastasis is frequently associated, to a large degree, with AKT/mTOR activities [227]. Thus, the enhancement of the aggressiveness and metastasis of melanoma has been suggested to occur through AKT-dependent mechanisms [228,229]. In this process, AKT acts as a molecular switch that is linked with the elevated activation of mTOR and S6K1, enhancement of angiogenesis, and the accumulation of reactive oxygen species (ROS) that can enhance further aggressiveness and metastasis in melanoma [209].
As is widely documented, the development of melanoma is attributed, in great part, to intensive skin exposure to both UVA and UVB rays that contribute to the dysregulation of the PI3K/AKT/mTOR pathway [230,231]. Thus, abnormal cellular alterations in response to genetic and non-genetic modifications of genes or proteins of cutaneous intracellular networks are expected to destroy skin tissue homeostasis, and ultimately lead to the development of different types of skin cancer [82,232].
Accumulated evidence indicates that activation of the mTOR pathway is strongly associated with the pathogenesis of melanoma [40,233]. Meanwhile, constitutive activation of mTOR has been reported as inhibiting autophagic cell death and dysregulation of the normal cell cycle [234,235]. Given that the molecular basis and targets of most melanomas have been described in detail, the development of anticancer agents with the ability to maintain skin tissue homeostasis and integrity is a promising therapeutic strategy. Overall, targeting PI3K, AKT, and mTOR proteins by a specific inhibitor may offer a therapeutic option for the treatment of different tumor types including melanoma.

9. PI3K/AKT/mTOR Pathway as a Therapeutic Target in Melanoma Treatment

Successful prevention of tumor development and progression requires the long-term administration of anticancer agents that have to be well tolerated by patients. Previous and current analyses of signaling pathways in different tumor types have revealed that the tumor driving-signaling pathways of melanoma contain key molecules that can serve as therapeutic targets for melanoma treatment [236,237,238].
In addition to limited clinical benefit as a monotherapy in melanoma treatment, treatment with PI3K-targeted inhibitors is associated with the development of intrinsic and acquired resistance [239,240]. The development of these resistance mechanisms has been shown to be the consequences of PI3K inhibitor-induced insulin release, leading to hyperphosphorylation of insulin growth factor receptor(s) (IGF1R) [241,242]. As consequence, the phosphorylated IGF1R becomes able to mediate the reactivation of the PI3K signaling axis in tumors and rescues AKT and S6 phosphorylation via insulin receptor substrate (IRS) adaptor molecule-dependent mechanisms [241,243,244]. Insulin-mediated feedback loops can evade the biological consequences of PI3Kα, mTORC1, and AKT inhibition [17,245,246].
Both PI3K/AKT/mTOR and RAF/MEK/ERK signaling cascades are derived from numerous feedback loops and are interconnected at multiple points of crosstalk. Inhibition of one of these pathways can result in the activation of the other signaling cascade [247]. Thus, dual targeting of both pathways may improve treatment efficacy, leading to better clinical outcomes. Numerous in vitro and in vivo preclinical studies have revealed that dual targeting of signaling pathways, such as the PI3K/AKT/mTOR and RAF/MEK/ERK pathways, is a clinically relevant treatment option [248]. For example, pyridinyl imidazole compounds have been reported to simultaneously target both the BRAF oncogene and mTORC1 signaling in human melanoma cells [249].
Although the immune checkpoint inhibitors have been utilized as an alternative therapeutic option for melanoma treatment [250,251,252], tumor microenvironment-induced immunosuppressive effects have been shown to impair the therapeutic efficacy of immune checkpoint inhibitors [253]. In contrast to inhibitors of the immune checkpoint, PI3K/AKT/mTOR and MAPK/MEK/ERK inhibitors have been shown to suppress tumor growth and block tumor microenvironment-mediated tumor growth and metastasis. For example, the inhibition of PI3K/AKT/mTOR was found to increase immunogenicity and thereby enhance tumor sensitivity to immunotherapy [254]. Thus, the combination of MAPK/MEK/ERK- and PI3K/AKT/mTOR-targeted therapies may overcome resistance to immunotherapy [255,256]. Combinatorial or sequential treatments are expected to benefit melanoma patients, particularly those with mutations in both PI3K and MAPK pathways [255,256].
Many of the multiple targeted inhibitors for Fyn/CD133, PI3K, AKT, or mTOR have been evaluated and tested in in vitro melanoma model and in phase I and II clinical trials in melanoma. These include the inhibition of the Fyn/Stat pathway by chalcone derivative in melanoma [257], the inhibitor of Fyn/CD133 (Saracatinib) that has been evaluated via a Phase II study in metastatic melanoma [258], the inhibitors of PI3K (BAY-80-6946) [259] and of buparlisib [260], the inhibitors of AKT including perifosine in patients with metastatic melanoma [261], and MK-2206 in BRAF wild-type melanoma [262]. Meanwhile, the dual inhibitor of PI3K/mTOR (NVP-BEZ235) has been studied in melanoma [263]. Also, the mTOR analogs including everolimus [264] and temsirolimus [265] have been tested on their inhibitory effect on mTORC1 in melanoma; conversely, mTOR kinase inhibitors were found to target both mTORC1 and mTORC2 [266,267]. In addition, the most common references describing the reliability of the Fyn/CD133/PI3K/mTOR pathway as a therapeutic target in melanoma treatment have been provided in Table 2 and the possible therapeutic targets of the Fyn/CD133/PI3K/Akt/mTOR pathway in melanoma are outlined in Figure 10.

10. Conclusions

Malignant melanoma is a deadly disease with a poor prognosis. Obtaining complete tumor remission is difficult because of the presence of a heterogeneous subpopulation of CSCs. However, the identification of CSCs in melanoma and other cancers has led to promising advances that may soon impact the management of these cancers. CSCs are also responsible for therapeutic resistance that leads to tumor relapse. Specific signaling mechanisms are required for the maintenance of CSCs in tumors that can maintain their microenvironment. CSCs are becoming priority targets for the development of novel antitumor therapy. The tumor milieu is a critical regulator of melanoma-specific CSC-driven angiogenesis and metastasis. Signaling effectors from ECM or stromal cells can act as EMT or MET inducers or may regulate dormancy at metastatic sites in CSCs. CSC-dependent melanoma progression is mediated by MAPK/ERK and PI3K/Akt/mTOR pathways. Given the unique biology of CSCs, there is great need to develop novel and promising approaches for CSC-targeted cancer therapy. Combinatorial and/or sequential inhibition of CD133 signaling to PI3K/AKT/mTOR and PI3K/RAS/RAF/MEK/ERK pathways, after first-line immunotherapy, may extend the anti-tumor response for melanoma patients. This is especially true for those harboring genetic alterations in key molecules of both the PI3K/AKT/mTOR and MAPK/MEK/ERK pathways. The development of clinically relevant pharmacological inhibitors to block the function of both PI3K/AKT/mTOR and MAPK/MEK/ERK could provide new avenues for well-designed studies that assess the tolerability and efficacy of a new therapeutic approach for the treatment of melanoma.

Author Contributions

Conceptualization, Y.A.H., R.T.B., M.H., N.K., A.A.A., T.W.F., H.S. and Y.H.; methodology, S.-Y.H., S.-L.H., S.S., M.H., A.A.A., T.W.F., H.S. and Y.H.; software, N.K., M.H., A.A.A. and T.W.F.; validation, M.H., A.A.A., Y.H., R.T.B. and S.S.; formal analysis, N.K., M.H. and T.W.F.; investigation, M.H., Y.H., A.A.A., Y.A.H., R.T.B. and S.S.; resources, Y.H., M.H., A.A.A., Y.A.H. and R.T.B.; data curation, M.H., N.K., Y.A.H., R.T.B. and S.S.; writing, M.H., T.W.F., N.K., H.S., A.A.A. and Y.H.; writing—review and editing, M.H., Y.H., A.A.A. and S.S.; visualization, M.H., S.-Y.H., A.A.A. and S.-L.H.; supervision, M.H., S.S., A.A.A. and Y.H.; project administration, M.H., S.-Y.H., S.S., H.S., A.A.A. and S.-L.H.; funding acquisition, M.H., Y.H. and M.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

Robert T. Brodell is a principal investigator for clinical trials (Novartis and Sanofi), the Corevitas psoriasis biologic registry, and owns stock in Veradermics, Inc. He has served on advisory boards for Amgen and Novan. The remaining authors have no conflicts of interest to disclose.

References

  1. Boumahdi, S.; de Sauvage, F.J. The great escape: Tumour cell plasticity in resistance to targeted therapy. Nat. Rev. Drug Discov. 2020, 19, 39–56. [Google Scholar] [CrossRef]
  2. Li, Z.; Jia, H.; Zhang, B.; Zhang, Y.; Li, H.; Song, P. The clinical features, treatment, and prognosis of primary mediastinal malignant melanoma: A case report. Medicine 2017, 96, e6436. [Google Scholar] [CrossRef]
  3. Hamid, O.; Robert, C.; Daud, A.; Hodi, F.S.; Hwu, W.J.; Kefford, R.; Wolchok, J.D.; Hersey, P.; Joseph, R.; Weber, J.S.; et al. Five-year survival outcomes for patients with advanced melanoma treated with pembrolizumab in KEYNOTE-001. Ann. Oncol. 2019, 30, 582–588. [Google Scholar] [CrossRef] [PubMed]
  4. El-Khattouti, A.; Sheehan, N.T.; Monico, J.; Drummond, H.A.; Haikel, Y.; Brodell, R.T.; Megahed, M.; Hassan, M. CD133+ melanoma subpopulation acquired resistance to caffeic acid phenethyl ester-induced apoptosis is attributed to the elevated expression of ABCB5: Significance for melanoma treatment. Cancer Lett. 2015, 357, 83–104. [Google Scholar] [CrossRef] [PubMed]
  5. El-Khattouti, A.; Selimovic, D.; Haïkel, Y.; Megahed, M.; Gomez, C.R.; Hassan, M. Identification and analysis of CD133(+) melanoma stem-like cells conferring resistance to taxol: An insight into the mechanisms of their resistance and response. Cancer Lett. 2014, 343, 123–133. [Google Scholar] [CrossRef] [PubMed]
  6. Barzegar Behrooz, A.; Syahir, A.; Ahmad, S. CD133: Beyond a cancer stem cell biomarker. J. Drug Target. 2019, 27, 257–269. [Google Scholar] [CrossRef] [PubMed]
  7. Kumar, D.; Kumar, S.; Gorain, M.; Tomar, D.; Patil, H.S.; Radharani, N.N.V.; Kumar, T.V.S.; Patil, T.V.; Thulasiram, H.V.; Kundu, G.C. Notch1-MAPK Signaling Axis Regulates CD133. J. Investig. Dermatol. 2016, 136, 2462–2474. [Google Scholar] [CrossRef]
  8. Jamal, S.M.E.; Alamodi, A.; Wahl, R.U.; Grada, Z.; Shareef, M.A.; Hassan, S.Y.; Murad, F.; Hassan, S.L.; Santourlidis, S.; Gomez, C.R.; et al. Melanoma stem cell maintenance and chemo-resistance are mediated by CD133 signal to PI3K-dependent pathways. Oncogene 2020, 39, 5468–5478. [Google Scholar] [CrossRef] [PubMed]
  9. Inoki, K.; Ouyang, H.; Li, Y.; Guan, K.L. Signaling by target of rapamycin proteins in cell growth control. Microbiol. Mol. Biol. Rev. 2005, 69, 79–100. [Google Scholar] [CrossRef] [PubMed]
  10. Zhang, S.; Cao, S.; Gong, M.; Zhang, W.; Zhu, Z.; Wu, S.; Yue, Y.; Qian, W.; Ma, Q.; Wang, S.; et al. Mechanically activated ion channel Piezo1 contributes to melanoma malignant progression through AKT/mTOR signaling. Cancer Biol. Ther. 2022, 23, 336–347. [Google Scholar] [CrossRef]
  11. Sanchez, J.N.; Wang, T.; Cohen, M.S. BRAF and MEK Inhibitors: Use and Resistance in BRAF-Mutated Cancers. Drugs 2018, 78, 549–566. [Google Scholar] [CrossRef]
  12. Adashek, J.J.; Menta, A.K.; Reddy, N.K.; Desai, A.P.; Roszik, J.; Subbiah, V. Tissue-Agnostic Activity of BRAF plus MEK Inhibitor in BRAF V600-Mutant Tumors. Mol. Cancer Ther. 2022, 21, 871–878. [Google Scholar] [CrossRef]
  13. Mak, G.; Arkenau, H.T.; Chin, M. Resistance surveillance in a BRAF mutant melanoma patient on long-term BRAF-inhibitor treatment. Melanoma Res. 2014, 24, 408–412. [Google Scholar] [CrossRef] [PubMed]
  14. Savoia, P.; Zavattaro, E.; Cremona, O. Clinical Implications of Acquired BRAF Inhibitors resistance in Melanoma. Int. J. Mol. Sci. 2020, 21, 9730. [Google Scholar] [CrossRef]
  15. Haluska, F.; Pemberton, T.; Ibrahim, N.; Kalinsky, K. The RTK/RAS/BRAF/PI3K pathways in melanoma: Biology, small molecule inhibitors, and potential applications. Semin. Oncol. 2007, 34, 546–554. [Google Scholar] [CrossRef]
  16. Khaddour, K.; Maahs, L.; Avila-Rodriguez, A.M.; Maamar, Y.; Samaan, S.; Ansstas, G. Melanoma Targeted Therapies beyond. Cancers 2021, 13, 5847. [Google Scholar] [CrossRef]
  17. Rozengurt, E.; Soares, H.P.; Sinnet-Smith, J. Suppression of feedback loops mediated by PI3K/mTOR induces multiple overactivation of compensatory pathways: An unintended consequence leading to drug resistance. Mol. Cancer Ther. 2014, 13, 2477–2488. [Google Scholar] [CrossRef]
  18. Corrales, E.; Levit-Zerdoun, E.; Metzger, P.; Mertes, R.; Lehmann, A.; Münch, J.; Lemke, S.; Kowar, S.; Boerries, M. PI3K/AKT signaling allows for MAPK/ERK pathway independency mediating dedifferentiation-driven Drug resistance in melanoma. Cell Commun. Signal. 2022, 20, 187. [Google Scholar] [CrossRef]
  19. Yajima, I.; Kumasaka, M.Y.; Thang, N.D.; Goto, Y.; Takeda, K.; Yamanoshita, O.; Iida, M.; Ohgami, N.; Tamura, H.; Kawamoto, Y.; et al. RAS/RAF/MEK/ERK and PI3K/PTEN/AKT Signaling in Malignant Melanoma Progression and Therapy. Dermatol. Res. Pract. 2012, 2012, 354191. [Google Scholar] [CrossRef] [PubMed]
  20. Ye, Y.; Jin, L.; Wilmott, J.S.; Hu, W.L.; Yosufi, B.; Thorne, R.F.; Liu, T.; Rizos, H.; Yan, X.G.; Dong, L.; et al. PI(4,5)P2 5-phosphatase A regulates PI3K/Akt signalling and has a tumour suppressive role in human melanoma. Nat. Commun. 2013, 4, 1508. [Google Scholar] [CrossRef] [PubMed]
  21. De Craene, J.O.; Bertazzi, D.L.; Bär, S.; Friant, S. Phosphoinositides, Major Actors in Membrane Trafficking and Lipid Signaling Pathways. Int. J. Mol. Sci. 2017, 18, 634. [Google Scholar] [CrossRef]
  22. Bilanges, B.; Posor, Y.; Vanhaesebroeck, B. PI3K isoforms in cell signalling and vesicle trafficking. Nat. Rev. Mol. Cell Biol. 2019, 20, 515–534. [Google Scholar] [CrossRef]
  23. Jean, S.; Kiger, A.A. Classes of phosphoinositide 3-kinases at a glance. J. Cell Sci. 2014, 127 Pt 5, 923–928. [Google Scholar] [CrossRef]
  24. Siempelkamp, B.D.; Rathinaswamy, M.K.; Jenkins, M.L.; Burke, J.E. Molecular mechanism of activation of class IA phosphoinositide 3-kinases (PI3Ks) by membrane-localized HRas. J. Biol. Chem. 2017, 292, 12256–12266. [Google Scholar] [CrossRef]
  25. Vidal, S.; Bouzaher, Y.H.; El Motiam, A.; Seoane, R.; Rivas, C. Overview of the regulation of the class IA PI3K/AKT pathway by SUMO. Semin. Cell Dev. Biol. 2022, 132, 51–61. [Google Scholar] [CrossRef]
  26. Fox, M.; Mott, H.R.; Owen, D. Class IA PI3K regulatory subunits: p110-independent roles and structures. Biochem. Soc. Trans. 2020, 48, 1397–1417. [Google Scholar] [CrossRef] [PubMed]
  27. Parkman, G.L.; Foth, M.; Kircher, D.A.; Holmen, S.L.; McMahon, M. The role of PI3’-lipid signalling in melanoma initiation, progression and maintenance. Exp. Dermatol. 2022, 31, 43–56. [Google Scholar] [CrossRef] [PubMed]
  28. Rodgers, S.J.; Ferguson, D.T.; Mitchell, C.A.; Ooms, L.M. Regulation of PI3K effector signalling in cancer by the phosphoinositide phosphatases. Biosci. Rep. 2017, 37, BSR20160432. [Google Scholar] [CrossRef] [PubMed]
  29. Kang, H.; Schneider, H.; Rudd, C.E. Phosphatidylinositol 3-kinase p85 adaptor function in T-cells. Co-stimulation and regulation of cytokine transcription independent of associated p110. J. Biol. Chem. 2002, 277, 912–921. [Google Scholar] [CrossRef]
  30. Jiang, X.; Chen, S.; Asara, J.M.; Balk, S.P. Phosphoinositide 3-kinase pathway activation in phosphate and tensin homolog (PTEN)-deficient prostate cancer cells is independent of receptor tyrosine kinases and mediated by the p110beta and p110delta catalytic subunits. J. Biol. Chem. 2010, 285, 14980–14989. [Google Scholar] [CrossRef]
  31. Sipeki, S.; Koprivanacz, K.; Takács, T.; Kurilla, A.; László, L.; Vas, V.; Buday, L. Novel Roles of SH2 and SH3 Domains in Lipid Binding. Cells 2021, 10, 1191. [Google Scholar] [CrossRef] [PubMed]
  32. Candido, S.; Salemi, R.; Piccinin, S.; Falzone, L.; Libra, M. The PIK3CA H1047R Mutation Confers Resistance to BRAF and MEK Inhibitors in A375 Melanoma Cells through the Cross-Activation of MAPK and PI3K-Akt Pathways. Pharmaceutics 2022, 14, 590. [Google Scholar] [CrossRef] [PubMed]
  33. Wu, X.; Yu, J.; Yan, J.; Dai, J.; Si, L.; Chi, Z.; Sheng, X.; Cui, C.; Ma, M.; Tang, H.; et al. PI3K/AKT/mTOR pathway inhibitors inhibit the growth of melanoma cells with mTOR H2189Y mutations in vitro. Cancer Biol. Ther. 2018, 19, 584–589. [Google Scholar] [CrossRef] [PubMed]
  34. Kong, Y.; Si, L.; Li, Y.; Wu, X.; Xu, X.; Dai, J.; Tang, H.; Ma, M.; Chi, Z.; Sheng, X.; et al. Analysis of mTOR Gene Aberrations in Melanoma Patients and Evaluation of Their Sensitivity to PI3K-AKT-mTOR Pathway Inhibitors. Clin. Cancer Res. 2016, 22, 1018–1027. [Google Scholar] [CrossRef]
  35. Sinnberg, T.; Lasithiotakis, K.; Niessner, H.; Schittek, B.; Flaherty, K.T.; Kulms, D.; Maczey, E.; Campos, M.; Gogel, J.; Garbe, C.; et al. Inhibition of PI3K-AKT-mTOR signaling sensitizes melanoma cells to cisplatin and temozolomide. J. Investig. Dermatol. 2009, 129, 1500–1515. [Google Scholar] [CrossRef]
  36. Hocker, T.L.; Singh, M.K.; Tsao, H. Melanoma genetics and therapeutic approaches in the 21st century: Moving from the benchside to the bedside. J. Investig. Dermatol. 2008, 128, 2575–2595. [Google Scholar] [CrossRef]
  37. Pópulo, H.; Lopes, J.M.; Soares, P. The mTOR signalling pathway in human cancer. Int. J. Mol. Sci. 2012, 13, 1886–1918. [Google Scholar] [CrossRef]
  38. Moschetta, M.; Reale, A.; Marasco, C.; Vacca, A.; Carratù, M.R. Therapeutic targeting of the mTOR-signalling pathway in cancer: Benefits and limitations. Br. J. Pharmacol. 2014, 171, 3801–3813. [Google Scholar] [CrossRef]
  39. Indini, A.; Fiorilla, I.; Ponzone, L.; Calautti, E.; Audrito, V. NAD/NAMPT and mTOR Pathways in Melanoma: Drivers of Drug Resistance and Prospective Therapeutic Targets. Int. J. Mol. Sci. 2022, 23, 9985. [Google Scholar] [CrossRef] [PubMed]
  40. Karbowniczek, M.; Spittle, C.S.; Morrison, T.; Wu, H.; Henske, E.P. mTOR is activated in the majority of malignant melanomas. J. Investig. Dermatol. 2008, 128, 980–987. [Google Scholar] [CrossRef] [PubMed]
  41. Song, Y.; Karakousis, G.C. Melanoma of unknown primary. J. Surg. Oncol. 2019, 119, 232–241. [Google Scholar] [CrossRef] [PubMed]
  42. Shain, A.H.; Yeh, I.; Kovalyshyn, I.; Sriharan, A.; Talevich, E.; Gagnon, A.; Dummer, R.; North, J.; Pincus, L.; Ruben, B.; et al. The Genetic Evolution of Melanoma from Precursor Lesions. N. Engl. J. Med. 2015, 373, 1926–1936. [Google Scholar] [CrossRef] [PubMed]
  43. Katzendobler, S.; Do, A.; Weller, J.; Rejeski, K.; Dorostkar, M.M.; Albert, N.L.; Forbrig, R.; Niyazi, M.; Egensperger, R.; Tonn, J.C.; et al. The value of stereotactic biopsy of primary and recurrent brain metastases in the era of precision medicine. Front. Oncol. 2022, 12, 1014711. [Google Scholar] [CrossRef] [PubMed]
  44. Ward-Peterson, M.; Acuña, J.M.; Alkhalifah, M.K.; Nasiri, A.M.; Al-Akeel, E.S.; Alkhaldi, T.M.; Dawari, S.A.; Aldaham, S.A. Association Between Race/Ethnicity and Survival of Melanoma Patients in the United States Over 3 Decades: A Secondary Analysis of SEER Data. Medicine 2016, 95, e3315. [Google Scholar] [CrossRef] [PubMed]
  45. Saginala, K.; Barsouk, A.; Aluru, J.S.; Rawla, P. Epidemiology of Melanoma. Med. Sci. 2021, 9, 63. [Google Scholar] [CrossRef] [PubMed]
  46. Haggar, F.A.; Boushey, R.P. Colorectal cancer epidemiology: Incidence, mortality, survival, and risk factors. Clin. Colon Rectal Surg. 2009, 22, 191–197. [Google Scholar] [CrossRef] [PubMed]
  47. Volkovova, K.; Bilanicova, D.; Bartonova, A.; Letašiová, S.; Dusinska, M. Associations between environmental factors and incidence of cutaneous melanoma. Review. Environ. Health 2012, 11 (Suppl. S1), S12. [Google Scholar] [CrossRef]
  48. Budden, T.; Bowden, N.A. The role of altered nucleotide excision repair and UVB-induced DNA damage in melanomagenesis. Int. J. Mol. Sci. 2013, 14, 1132–1151. [Google Scholar] [CrossRef]
  49. Kumar, R.; Deep, G.; Agarwal, R. An Overview of Ultraviolet B Radiation-Induced Skin Cancer Chemoprevention by Silibinin. Curr. Pharmacol. Rep. 2015, 1, 206–215. [Google Scholar] [CrossRef]
  50. Zhang, A.; Miao, K.; Sun, H.; Deng, C.X. Tumor heterogeneity reshapes the tumor microenvironment to influence drug resistance. Int. J. Biol. Sci. 2022, 18, 3019–3033. [Google Scholar] [CrossRef]
  51. Yang, E.; Wang, X.; Gong, Z.; Yu, M.; Wu, H.; Zhang, D. Exosome-mediated metabolic reprogramming: The emerging role in tumor microenvironment remodeling and its influence on cancer progression. Signal Transduct. Target. Ther. 2020, 5, 242. [Google Scholar] [CrossRef]
  52. McQuerry, J.A.; Chang, J.T.; Bowtell, D.D.L.; Cohen, A.; Bild, A.H. Mechanisms and clinical implications of tumor heterogeneity and convergence on recurrent phenotypes. J. Mol. Med. 2017, 95, 1167–1178. [Google Scholar] [CrossRef] [PubMed]
  53. Burrell, R.A.; McGranahan, N.; Bartek, J.; Swanton, C. The causes and consequences of genetic heterogeneity in cancer evolution. Nature 2013, 501, 338–345. [Google Scholar] [CrossRef] [PubMed]
  54. Hida, K.; Ohga, N.; Akiyama, K.; Maishi, N.; Hida, Y. Heterogeneity of tumor endothelial cells. Cancer Sci. 2013, 104, 1391–1395. [Google Scholar] [CrossRef] [PubMed]
  55. Lim, S.Y.; Menzies, A.M.; Rizos, H. Mechanisms and strategies to overcome resistance to molecularly targeted therapy for melanoma. Cancer 2017, 123, 2118–2129. [Google Scholar] [CrossRef] [PubMed]
  56. Tellez-Gabriel, M.; Ory, B.; Lamoureux, F.; Heymann, M.F.; Heymann, D. Tumour Heterogeneity: The Key Advantages of Single-Cell Analysis. Int. J. Mol. Sci. 2016, 17, 2142. [Google Scholar] [CrossRef]
  57. Sun, X.X.; Yu, Q. Intra-tumor heterogeneity of cancer cells and its implications for cancer treatment. Acta Pharmacol. Sin. 2015, 36, 1219–1227. [Google Scholar] [CrossRef]
  58. Sanli, Y.; Leake, J.; Odu, A.; Xi, Y.; Subramaniam, R.M. Tumor Heterogeneity on FDG PET/CT and Immunotherapy: An Imaging Biomarker for Predicting Treatment Response in Patients With Metastatic Melanoma. AJR Am. J. Roentgenol. 2019, 212, 1318–1326. [Google Scholar] [CrossRef]
  59. Crucitta, S.; Cucchiara, F.; Mathijssen, R.; Mateo, J.; Jager, A.; Joosse, A.; Passaro, A.; Attili, I.; Petrini, I.; van Schaik, R.; et al. Treatment-driven tumour heterogeneity and drug resistance: Lessons from solid tumours. Cancer Treat. Rev. 2022, 104, 102340. [Google Scholar] [CrossRef]
  60. Shannan, B.; Perego, M.; Somasundaram, R.; Herlyn, M. Heterogeneity in Melanoma. Cancer. Treat. Res. 2016, 167, 1–15. [Google Scholar] [CrossRef]
  61. Katenkamp, D. Cellular heterogeneity. Explanation for changing of tumor phenotype and biologic behavior in soft tissue sarcomas. Pathol. Res. Pract. 1988, 183, 698–705. [Google Scholar] [CrossRef] [PubMed]
  62. Jamal-Hanjani, M.; Quezada, S.A.; Larkin, J.; Swanton, C. Translational implications of tumor heterogeneity. Clin. Cancer Res. 2015, 21, 1258–1266. [Google Scholar] [CrossRef]
  63. Sanna, A.; Harbst, K.; Johansson, I.; Christensen, G.; Lauss, M.; Mitra, S.; Rosengren, F.; Häkkinen, J.; Vallon-Christersson, J.; Olsson, H.; et al. Tumor genetic heterogeneity analysis of chronic sun-damaged melanoma. Pigment Cell Melanoma Res. 2020, 33, 480–489. [Google Scholar] [CrossRef]
  64. Testa, U.; Castelli, G.; Pelosi, E. Melanoma: Genetic Abnormalities, Tumor Progression, Clonal Evolution and Tumor Initiating Cells. Med. Sci. 2017, 5, 28. [Google Scholar] [CrossRef]
  65. Pipek, O.; Vizkeleti, L.; Doma, V.; Alpár, D.; Bödör, C.; Kárpáti, S.; Timar, J. The Driverless Triple-Wild-Type (BRAF, RAS, KIT) Cutaneous Melanoma: Whole Genome Sequencing Discoveries. Cancers 2023, 15, 1712. [Google Scholar] [CrossRef]
  66. Guo, W.; Wang, H.; Li, C. Signal pathways of melanoma and targeted therapy. Signal Transduct. Target. Ther. 2021, 6, 424. [Google Scholar] [CrossRef] [PubMed]
  67. Palmieri, G.; Colombino, M.; Casula, M.; Manca, A.; Mandalà, M.; Cossu, A.; Italian Melanoma Intergroup for the Italian Melanoma Intergroup (IMI). Molecular Pathways in Melanomagenesis: What We Learned from Next-Generation Sequencing Approaches. Curr. Oncol. Rep. 2018, 20, 86. [Google Scholar] [CrossRef] [PubMed]
  68. Grzywa, T.M.; Paskal, W.; Włodarski, P.K. Intratumor and Intertumor Heterogeneity in Melanoma. Transl. Oncol. 2017, 10, 956–975. [Google Scholar] [CrossRef]
  69. Ng, M.F.; Simmons, J.L.; Boyle, G.M. Heterogeneity in Melanoma. Cancers 2022, 14, 3030. [Google Scholar] [CrossRef]
  70. Kumar, D.; Gorain, M.; Kundu, G.; Kundu, G.C. Therapeutic implications of cellular and molecular biology of cancer stem cells in melanoma. Mol. Cancer 2017, 16, 7. [Google Scholar] [CrossRef]
  71. Hass, R.; von der Ohe, J.; Ungefroren, H. Impact of the Tumor Microenvironment on Tumor Heterogeneity and Consequences for Cancer Cell Plasticity and Stemness. Cancers 2020, 12, 3716. [Google Scholar] [CrossRef]
  72. Manning, B.D.; Toker, A. AKT/PKB Signaling: Navigating the Network. Cell 2017, 169, 381–405. [Google Scholar] [CrossRef]
  73. Shlyakhtina, Y.; Moran, K.L.; Portal, M.M. Genetic and Non-Genetic Mechanisms Underlying Cancer Evolution. Cancers 2021, 13, 1380. [Google Scholar] [CrossRef]
  74. Hirata, E.; Sahai, E. Tumor Microenvironment and Differential Responses to Therapy. Cold Spring Harb. Perspect. Med. 2017, 7, a026781. [Google Scholar] [CrossRef]
  75. Hölzel, M.; Tüting, T. Inflammation-Induced Plasticity in Melanoma Therapy and Metastasis. Trends Immunol. 2016, 37, 364–374. [Google Scholar] [CrossRef]
  76. Shou, Y.; Yang, L.; Yang, Y.; Zhu, X.; Li, F.; Xu, J. Determination of hypoxia signature to predict prognosis and the tumor immune microenvironment in melanoma. Mol. Omics 2021, 17, 307–316. [Google Scholar] [CrossRef]
  77. Umansky, V.; Sevko, A. Overcoming immunosuppression in the melanoma microenvironment induced by chronic inflammation. Cancer Immunol. Immunother. 2012, 61, 275–282. [Google Scholar] [CrossRef]
  78. Ahmed, F.; Haass, N.K. Microenvironment-Driven Dynamic Heterogeneity and Phenotypic Plasticity as a Mechanism of Melanoma Therapy Resistance. Front. Oncol. 2018, 8, 173. [Google Scholar] [CrossRef] [PubMed]
  79. Granados, K.; Poelchen, J.; Novak, D.; Utikal, J. Cellular Reprogramming-A Model for Melanoma Cellular Plasticity. Int. J. Mol. Sci. 2020, 21, 8274. [Google Scholar] [CrossRef] [PubMed]
  80. Bettum, I.J.; Gorad, S.S.; Barkovskaya, A.; Pettersen, S.; Moestue, S.A.; Vasiliauskaite, K.; Tenstad, E.; Øyjord, T.; Risa, Ø.; Nygaard, V.; et al. Metabolic reprogramming supports the invasive phenotype in malignant melanoma. Cancer Lett. 2015, 366, 71–83. [Google Scholar] [CrossRef] [PubMed]
  81. Falletta, P.; Goding, C.R.; Vivas-García, Y. Connecting Metabolic Rewiring With Phenotype Switching in Melanoma. Front. Cell Dev. Biol. 2022, 10, 930250. [Google Scholar] [CrossRef]
  82. Karami Fath, M.; Ebrahimi, M.; Nourbakhsh, E.; Zia Hazara, A.; Mirzaei, A.; Shafieyari, S.; Salehi, A.; Hoseinzadeh, M.; Payandeh, Z.; Barati, G. PI3K/Akt/mTOR signaling pathway in cancer stem cells. Pathol. Res. Pract. 2022, 237, 154010. [Google Scholar] [CrossRef]
  83. Rambow, F.; Marine, J.C.; Goding, C.R. Melanoma plasticity and phenotypic diversity: Therapeutic barriers and opportunities. Genes Dev. 2019, 33, 1295–1318. [Google Scholar] [CrossRef]
  84. Pagliuca, C.; Di Leo, L.; De Zio, D. New Insights into the Phenotype Switching of Melanoma. Cancers 2022, 14, 6118. [Google Scholar] [CrossRef] [PubMed]
  85. Qian, J.; Olbrecht, S.; Boeckx, B.; Vos, H.; Laoui, D.; Etlioglu, E.; Wauters, E.; Pomella, V.; Verbandt, S.; Busschaert, P.; et al. A pan-cancer blueprint of the heterogeneous tumor microenvironment revealed by single-cell profiling. Cell Res. 2020, 30, 745–762. [Google Scholar] [CrossRef] [PubMed]
  86. Hossain, S.M.; Eccles, M.R. Phenotype Switching and the Melanoma Microenvironment; Impact on Immunotherapy and Drug Resistance. Int. J. Mol. Sci. 2023, 24, 1601. [Google Scholar] [CrossRef] [PubMed]
  87. Chen, Z.; Han, F.; Du, Y.; Shi, H.; Zhou, W. Hypoxic microenvironment in cancer: Molecular mechanisms and therapeutic interventions. Signal Transduct. Target. Ther. 2023, 8, 70. [Google Scholar] [CrossRef] [PubMed]
  88. Zhao, M.; Wang, S.; Zuo, A.; Zhang, J.; Wen, W.; Jiang, W.; Chen, H.; Liang, D.; Sun, J.; Wang, M. HIF-1α/JMJD1A signaling regulates inflammation and oxidative stress following hyperglycemia and hypoxia-induced vascular cell injury. Cell. Mol. Biol. Lett. 2021, 26, 40. [Google Scholar] [CrossRef] [PubMed]
  89. Yang, L.; Lin, P.C. Mechanisms that drive inflammatory tumor microenvironment, tumor heterogeneity, and metastatic progression. Semin. Cancer Biol. 2017, 47, 185–195. [Google Scholar] [CrossRef]
  90. Benboubker, V.; Boivin, F.; Dalle, S.; Caramel, J. Cancer Cell Phenotype Plasticity as a Driver of Immune Escape in Melanoma. Front. Immunol. 2022, 13, 873116. [Google Scholar] [CrossRef]
  91. Knappe, N.; Novak, D.; Weina, K.; Bernhardt, M.; Reith, M.; Larribere, L.; Hölzel, M.; Tüting, T.; Gebhardt, C.; Umansky, V.; et al. Directed Dedifferentiation Using Partial Reprogramming Induces Invasive Phenotype in Melanoma Cells. Stem Cells 2016, 34, 832–846. [Google Scholar] [CrossRef]
  92. Diazzi, S.; Tartare-Deckert, S.; Deckert, M. The mechanical phenotypic plasticity of melanoma cell: An emerging driver of therapy cross-resistance. Oncogenesis 2023, 12, 7. [Google Scholar] [CrossRef] [PubMed]
  93. Vandamme, N.; Berx, G. Melanoma cells revive an embryonic transcriptional network to dictate phenotypic heterogeneity. Front. Oncol. 2014, 4, 352. [Google Scholar] [CrossRef] [PubMed]
  94. Romano, V.; Belviso, I.; Venuta, A.; Ruocco, M.R.; Masone, S.; Aliotta, F.; Fiume, G.; Montagnani, S.; Avagliano, A.; Arcucci, A. Influence of Tumor Microenvironment and Fibroblast Population Plasticity on Melanoma Growth, Therapy Resistance and Immunoescape. Int. J. Mol. Sci. 2021, 22, 5283. [Google Scholar] [CrossRef] [PubMed]
  95. Bae, H.J.; Liu, S.; Jin, P.; Stroncek, D. Mesenchymal stromal cell plasticity and the tumor microenvironment. Emerg. Top. Life Sci. 2017, 1, 487–492. [Google Scholar] [CrossRef] [PubMed]
  96. Tsang, M.; Quesnel, K.; Vincent, K.; Hutchenreuther, J.; Postovit, L.M.; Leask, A. Insights into Fibroblast Plasticity: Cellular Communication Network 2 Is Required for Activation of Cancer-Associated Fibroblasts in a Murine Model of Melanoma. Am. J. Pathol. 2020, 190, 206–221. [Google Scholar] [CrossRef] [PubMed]
  97. Papaccio, F.; Kovacs, D.; Bellei, B.; Caputo, S.; Migliano, E.; Cota, C.; Picardo, M. Profiling Cancer-Associated Fibroblasts in Melanoma. Int. J. Mol. Sci. 2021, 22, 7255. [Google Scholar] [CrossRef]
  98. Yoshida, M.; Selvan, S.; McCue, P.A.; DeAngelis, T.; Baserga, R.; Fujii, A.; Rui, H.; Mastrangelo, M.J.; Sato, T. Expression of insulin-like growth factor-1 receptor in metastatic uveal melanoma and implications for potential autocrine and paracrine tumor cell growth. Pigment Cell Melanoma Res. 2014, 27, 297–308. [Google Scholar] [CrossRef]
  99. White, R.M.; Zon, L.I. Melanocytes in development, regeneration, and cancer. Cell Stem Cell 2008, 3, 242–252. [Google Scholar] [CrossRef]
  100. Centeno, P.P.; Pavet, V.; Marais, R. The journey from melanocytes to melanoma. Nat. Rev. Cancer 2023, 23, 372–390. [Google Scholar] [CrossRef]
  101. Cichorek, M.; Wachulska, M.; Stasiewicz, A.; Tymińska, A. Skin melanocytes: Biology and development. Postepy Dermatol. Alergol. 2013, 30, 30–41. [Google Scholar] [CrossRef]
  102. Li, L.; Fukunaga-Kalabis, M.; Herlyn, M. Isolation and cultivation of dermal stem cells that differentiate into functional epidermal melanocytes. Methods Mol. Biol. 2012, 806, 15–29. [Google Scholar] [CrossRef]
  103. Hoerter, J.D.; Bradley, P.; Casillas, A.; Chambers, D.; Weiswasser, B.; Clements, L.; Gilbert, S.; Jiao, A. Does melanoma begin in a melanocyte stem cell? J. Skin Cancer 2012, 2012, 571087. [Google Scholar] [CrossRef] [PubMed]
  104. Grichnik, J.M.; Ali, W.N.; Burch, J.A.; Byers, J.D.; Garcia, C.A.; Clark, R.E.; Shea, C.R. KIT expression reveals a population of precursor melanocytes in human skin. J. Investig. Dermatol. 1996, 106, 967–971. [Google Scholar] [CrossRef] [PubMed]
  105. Zabierowski, S.E.; Fukunaga-Kalabis, M.; Li, L.; Herlyn, M. Dermis-derived stem cells: A source of epidermal melanocytes and melanoma? Pigment Cell Melanoma Res. 2011, 24, 422–429. [Google Scholar] [CrossRef] [PubMed]
  106. Kyrgidis, A.; Tzellos, T.G.; Triaridis, S. Melanoma: Stem cells, sun exposure and hallmarks for carcinogenesis, molecular concepts and future clinical implications. J. Carcinog. 2010, 9, 3. [Google Scholar] [CrossRef]
  107. Valyi-Nagy, K.; Kormos, B.; Ali, M.; Shukla, D.; Valyi-Nagy, T. Stem cell marker CD271 is expressed by vasculogenic mimicry-forming uveal melanoma cells in three-dimensional cultures. Mol. Vis. 2012, 18, 588–592. [Google Scholar]
  108. sher, G.; Masoodi, T.; Patil, K.; Akhtar, S.; Kuttikrishnan, S.; Ahmad, A.; Uddin, S. Dysregulated FOXM1 signaling in the regulation of cancer stem cells. Semin. Cancer Biol. 2022, 86 Pt 3, 107–121. [Google Scholar] [CrossRef]
  109. Faião-Flores, F.; Smalley, K.S.M. Get with the Program! Stemness and Reprogramming in Melanoma Metastasis. J. Investig. Dermatol. 2018, 138, 10–13. [Google Scholar] [CrossRef]
  110. Pine, S.R.; Liu, W. Asymmetric cell division and template DNA co-segregation in cancer stem cells. Front. Oncol. 2014, 4, 226. [Google Scholar] [CrossRef] [PubMed]
  111. Gómez-López, S.; Lerner, R.G.; Petritsch, C. Asymmetric cell division of stem and progenitor cells during homeostasis and cancer. Cell. Mol. Life Sci. 2014, 71, 575–597. [Google Scholar] [CrossRef]
  112. Schatton, T.; Frank, M.H. Cancer stem cells and human malignant melanoma. Pigment Cell Melanoma Res. 2008, 21, 39–55. [Google Scholar] [CrossRef]
  113. Fargeas, C.A.; Florek, M.; Huttner, W.B.; Corbeil, D. Characterization of prominin-2, a new member of the prominin family of pentaspan membrane glycoproteins. J. Biol. Chem. 2003, 278, 8586–8596. [Google Scholar] [CrossRef]
  114. Corbeil, D.; Karbanová, J.; Fargeas, C.A.; Jászai, J. Prominin-1 (CD133): Molecular and Cellular Features Across Species. Adv. Exp. Med. Biol. 2013, 777, 3–24. [Google Scholar] [CrossRef] [PubMed]
  115. Liu, C.; Li, Y.; Xing, Y.; Cao, B.; Yang, F.; Yang, T.; Ai, Z.; Wei, Y.; Jiang, J. The Interaction between Cancer Stem Cell Marker CD133 and Src Protein Promotes Focal Adhesion Kinase (FAK) Phosphorylation and Cell Migration. J. Biol. Chem. 2016, 291, 15540–15550. [Google Scholar] [CrossRef] [PubMed]
  116. Wei, D.; Labbé, D.; Fontaine, N.; Lamy, S.; Beaulieu, E.; Gingras, D.; Béliveau, R. The stem cell marker CD133 (prominin-1) is phosphorylated on cytoplasmic tyrosine-828 and tyrosine-852 by Src and Fyn tyrosine kinases. Biochemistry 2009, 48, 3998–4007. [Google Scholar] [CrossRef]
  117. González-Herrero, I.; Romero-Camarero, I.; Cañueto, J.; Cardeñoso-Álvarez, E.; Fernández-López, E.; Pérez-Losada, J.; Sánchez-García, I.; Román-Curto, C. CD133+ cell content correlates with tumour growth in melanomas from skin with chronic sun-induced damage. Br. J. Dermatol. 2013, 169, 830–837. [Google Scholar] [CrossRef] [PubMed]
  118. Madjd, Z.; Erfani, E.; Gheytanchi, E.; Moradi-Lakeh, M.; Shariftabrizi, A.; Asadi-Lari, M. Expression of CD133 cancer stem cell marker in melanoma: A systematic review and meta-analysis. Int. J. Biol. Markers 2016, 31, e118–e125. [Google Scholar] [CrossRef] [PubMed]
  119. Simbulan-Rosenthal, C.M.; Gaur, A.; Zhou, H.; AbdusSamad, M.; Qin, Q.; Dougherty, R.; Aljehane, L.; Kuo, L.W.; Vakili, S.; Karna, K.; et al. CD133 Is Associated with Increased Melanoma Cell Survival after Multikinase Inhibition. J. Oncol. 2019, 2019, 6486173. [Google Scholar] [CrossRef] [PubMed]
  120. Lai, I.C.; Shih, P.H.; Yao, C.J.; Yeh, C.T.; Wang-Peng, J.; Lui, T.N.; Chuang, S.E.; Hu, T.S.; Lai, T.Y.; Lai, G.M. Elimination of cancer stem-like cells and potentiation of temozolomide sensitivity by Honokiol in glioblastoma multiforme cells. PLoS ONE 2015, 10, e0114830. [Google Scholar] [CrossRef] [PubMed]
  121. Yang, Y.P.; Chien, Y.; Chiou, G.Y.; Cherng, J.Y.; Wang, M.L.; Lo, W.L.; Chang, Y.L.; Huang, P.I.; Chen, Y.W.; Shih, Y.H.; et al. Inhibition of cancer stem cell-like properties and reduced chemoradioresistance of glioblastoma using microRNA145 with cationic polyurethane-short branch PEI. Biomaterials 2012, 33, 1462–1476. [Google Scholar] [CrossRef] [PubMed]
  122. Tseng, L.M.; Huang, P.I.; Chen, Y.R.; Chen, Y.C.; Chou, Y.C.; Chen, Y.W.; Chang, Y.L.; Hsu, H.S.; Lan, Y.T.; Chen, K.H.; et al. Targeting signal transducer and activator of transcription 3 pathway by cucurbitacin I diminishes self-renewing and radiochemoresistant abilities in thyroid cancer-derived CD133+ cells. J. Pharmacol. Exp. Ther. 2012, 341, 410–423. [Google Scholar] [CrossRef] [PubMed]
  123. Davies, M.A. The role of the PI3K-AKT pathway in melanoma. Cancer J. 2012, 18, 142–147. [Google Scholar] [CrossRef] [PubMed]
  124. Rascio, F.; Spadaccino, F.; Rocchetti, M.T.; Castellano, G.; Stallone, G.; Netti, G.S.; Ranieri, E. The Pathogenic Role of PI3K/AKT Pathway in Cancer Onset and Drug Resistance: An Updated Review. Cancers 2021, 13, 3949. [Google Scholar] [CrossRef] [PubMed]
  125. Yuan, T.L.; Cantley, L.C. PI3K pathway alterations in cancer: Variations on a theme. Oncogene 2008, 27, 5497–5510. [Google Scholar] [CrossRef]
  126. Wang, J.; Cai, S.; Xiong, Q.; Weng, D.; Wang, Q.; Ma, Z. PIK3R2 predicts poor outcomes for patients with melanoma and contributes to the malignant progression via PI3K/AKT/NF-κB axis. Clin. Transl. Oncol. 2023, 25, 1402–1412. [Google Scholar] [CrossRef] [PubMed]
  127. Ueda, Y.; Richmond, A. NF-kappaB activation in melanoma. Pigment Cell Res. 2006, 19, 112–124. [Google Scholar] [CrossRef]
  128. Amiri, K.I.; Richmond, A. Role of nuclear factor-kappa B in melanoma. Cancer Metastasis Rev. 2005, 24, 301–313. [Google Scholar] [CrossRef]
  129. Qi, X.; Chen, Y.; Liu, S.; Liu, L.; Yu, Z.; Yin, L.; Fu, L.; Deng, M.; Liang, S.; Lü, M. Sanguinarine inhibits melanoma invasion and migration by targeting the FAK/PI3K/AKT/mTOR signalling pathway. Pharm. Biol. 2023, 61, 696–709. [Google Scholar] [CrossRef]
  130. Khan, K.H.; Yap, T.A.; Yan, L.; Cunningham, D. Targeting the PI3K-AKT-mTOR signaling network in cancer. Chin. J. Cancer 2013, 32, 253–265. [Google Scholar] [CrossRef]
  131. Babchia, N.; Calipel, A.; Mouriaux, F.; Faussat, A.M.; Mascarelli, F. The PI3K/Akt and mTOR/P70S6K signaling pathways in human uveal melanoma cells: Interaction with B-Raf/ERK. Investig. Ophthalmol. Vis. Sci. 2010, 51, 421–429. [Google Scholar] [CrossRef]
  132. Gil, D.; Zarzycka, M.; Pabijan, J.; Lekka, M.; Dulińska-Litewka, J. Dual targeting of melanoma translation by MNK/eIF4E and PI3K/mTOR inhibitors. Cell. Signal. 2023, 109, 110742. [Google Scholar] [CrossRef]
  133. Caporali, S.; Alvino, E.; Lacal, P.M.; Levati, L.; Giurato, G.; Memoli, D.; Caprini, E.; Antonini Cappellini, G.C.; D’Atri, S. Targeting the PI3K/AKT/mTOR pathway overcomes the stimulating effect of dabrafenib on the invasive behavior of melanoma cells with acquired resistance to the BRAF inhibitor. Int. J. Oncol. 2016, 49, 1164–1174. [Google Scholar] [CrossRef]
  134. Madhunapantula, S.V.; Robertson, G.P. The PTEN-AKT3 signaling cascade as a therapeutic target in melanoma. Pigment Cell Melanoma Res. 2009, 22, 400–419. [Google Scholar] [CrossRef]
  135. Gong, C.; Xia, H. Resveratrol suppresses melanoma growth by promoting autophagy through inhibiting the PI3K/AKT/mTOR signaling pathway. Exp. Ther. Med. 2020, 19, 1878–1886. [Google Scholar] [CrossRef]
  136. Robertson, G.P. Functional and therapeutic significance of Akt deregulation in malignant melanoma. Cancer Metastasis Rev. 2005, 24, 273–285. [Google Scholar] [CrossRef]
  137. McCubrey, J.A.; Steelman, L.S.; Chappell, W.H.; Abrams, S.L.; Wong, E.W.; Chang, F.; Lehmann, B.; Terrian, D.M.; Milella, M.; Tafuri, A.; et al. Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance. Biochim. Biophys. Acta 2007, 1773, 1263–1284. [Google Scholar] [CrossRef]
  138. Gocek, E.; Moulas, A.N.; Studzinski, G.P. Non-receptor protein tyrosine kinases signaling pathways in normal and cancer cells. Crit. Rev. Clin. Lab. Sci. 2014, 51, 125–137. [Google Scholar] [CrossRef] [PubMed]
  139. Srivastava, A.K.; Chiasson, J.L. Comparative characterization of receptor and non-receptor associated protein tyrosine kinases. Biochim. Biophys. Acta 1989, 996, 13–18. [Google Scholar] [CrossRef] [PubMed]
  140. Siveen, K.S.; Prabhu, K.S.; Achkar, I.W.; Kuttikrishnan, S.; Shyam, S.; Khan, A.Q.; Merhi, M.; Dermime, S.; Uddin, S. Role of Non Receptor Tyrosine Kinases in Hematological Malignances and its Targeting by Natural Products. Mol. Cancer 2018, 17, 31. [Google Scholar] [CrossRef] [PubMed]
  141. Kim, J.; Lee, C.D.; Rath, A.; Davidson, A.R. Recognition of non-canonical peptides by the yeast Fus1p SH3 domain: Elucidation of a common mechanism for diverse SH3 domain specificities. J. Mol. Biol. 2008, 377, 889–901. [Google Scholar] [CrossRef] [PubMed]
  142. Ahmed, I.; Calle, Y.; Sayed, M.A.; Kamal, J.M.; Rengaswamy, P.; Manser, E.; Meiners, S.; Nur-E-Kamal, A. Cdc42-dependent nuclear translocation of non-receptor tyrosine kinase, ACK. Biochem. Biophys. Res. Commun. 2004, 314, 571–579. [Google Scholar] [CrossRef] [PubMed]
  143. Engen, J.R.; Wales, T.E.; Hochrein, J.M.; Meyn, M.A.; Banu Ozkan, S.; Bahar, I.; Smithgall, T.E. Structure and dynamic regulation of Src-family kinases. Cell. Mol. Life Sci. 2008, 65, 3058–3073. [Google Scholar] [CrossRef] [PubMed]
  144. Prieto-Echagüe, V.; Miller, W.T. Regulation of ack-family nonreceptor tyrosine kinases. J. Signal Transduct. 2011, 2011, 742372. [Google Scholar] [CrossRef] [PubMed]
  145. Tabernero, L.; Aricescu, A.R.; Jones, E.Y.; Szedlacsek, S.E. Protein tyrosine phosphatases: Structure-function relationships. FEBS J. 2008, 275, 867–882. [Google Scholar] [CrossRef] [PubMed]
  146. Du, Z.; Lovly, C.M. Mechanisms of receptor tyrosine kinase activation in cancer. Mol. Cancer 2018, 17, 58. [Google Scholar] [CrossRef] [PubMed]
  147. Marholz, L.J.; Zeringo, N.A.; Lou, H.J.; Turk, B.E.; Parker, L.L. In Silico Design and in Vitro Characterization of Universal Tyrosine Kinase Peptide Substrates. Biochemistry 2018, 57, 1847–1851. [Google Scholar] [CrossRef]
  148. Luechapanichkul, R.; Chen, X.; Taha, H.A.; Vyas, S.; Guan, X.; Freitas, M.A.; Hadad, C.M.; Pei, D. Specificity profiling of dual specificity phosphatase vaccinia VH1-related (VHR) reveals two distinct substrate binding modes. J. Biol. Chem. 2013, 288, 6498–6510. [Google Scholar] [CrossRef]
  149. Songyang, Z.; Shoelson, S.E.; Chaudhuri, M.; Gish, G.; Pawson, T.; Haser, W.G.; King, F.; Roberts, T.; Ratnofsky, S.; Lechleider, R.J. SH2 domains recognize specific phosphopeptide sequences. Cell 1993, 72, 767–778. [Google Scholar] [CrossRef]
  150. Miller, W.T. Determinants of substrate recognition in nonreceptor tyrosine kinases. Acc. Chem. Res. 2003, 36, 393–400. [Google Scholar] [CrossRef]
  151. Rickles, R.J.; Botfield, M.C.; Weng, Z.; Taylor, J.A.; Green, O.M.; Brugge, J.S.; Zoller, M.J. Identification of Src, Fyn, Lyn, PI3K and Abl SH3 domain ligands using phage display libraries. EMBO J. 1994, 13, 5598–5604. [Google Scholar] [CrossRef]
  152. Jaber Chehayeb, R.; Boggon, T.J. SH2 Domain Binding: Diverse FLVRs of Partnership. Front. Endocrinol. 2020, 11, 575220. [Google Scholar] [CrossRef] [PubMed]
  153. Wagner, M.J.; Stacey, M.M.; Liu, B.A.; Pawson, T. Molecular mechanisms of SH2- and PTB-domain-containing proteins in receptor tyrosine kinase signaling. Cold Spring Harb. Perspect. Biol. 2013, 5, a008987. [Google Scholar] [CrossRef] [PubMed]
  154. Schumacher, C.; Knudsen, B.S.; Ohuchi, T.; Di Fiore, P.P.; Glassman, R.H.; Hanafusa, H. The SH3 domain of Crk binds specifically to a conserved proline-rich motif in Eps15 and Eps15R. J. Biol. Chem. 1995, 270, 15341–15347. [Google Scholar] [CrossRef]
  155. Mehrabipour, M.; Jasemi, N.S.K.; Dvorsky, R.; Ahmadian, M.R. A Systematic Compilation of Human SH3 Domains: A Versatile Superfamily in Cellular Signaling. Cells 2023, 12, 2054. [Google Scholar] [CrossRef] [PubMed]
  156. Williams, J.C.; Wierenga, R.K.; Saraste, M. Insights into Src kinase functions: Structural comparisons. Trends Biochem. Sci. 1998, 23, 179–184. [Google Scholar] [CrossRef]
  157. Hu, X.; Li, J.; Fu, M.; Zhao, X.; Wang, W. The JAK/STAT signaling pathway: From bench to clinic. Signal Transduct. Target. Ther. 2021, 6, 402. [Google Scholar] [CrossRef]
  158. Levine, S.J. Molecular mechanisms of soluble cytokine receptor generation. J. Biol. Chem. 2008, 283, 14177–14181. [Google Scholar] [CrossRef]
  159. Sun, Y.; Chen, Y.; Zhan, L.; Zhang, L.; Hu, J.; Gao, Z. The role of non-receptor protein tyrosine kinases in the excitotoxicity induced by the overactivation of NMDA receptors. Rev. Neurosci. 2016, 27, 283–289. [Google Scholar] [CrossRef]
  160. Chen, S.; Brier, S.; Smithgall, T.E.; Engen, J.R. The Abl SH2-kinase linker naturally adopts a conformation competent for SH3 domain binding. Protein Sci. 2007, 16, 572–581. [Google Scholar] [CrossRef]
  161. Donaldson, L.W.; Gish, G.; Pawson, T.; Kay, L.E.; Forman-Kay, J.D. Structure of a regulatory complex involving the Abl SH3 domain, the Crk SH2 domain, and a Crk-derived phosphopeptide. Proc. Natl. Acad. Sci. USA 2002, 99, 14053–14058. [Google Scholar] [CrossRef] [PubMed]
  162. Owen, L.M.; Bax, N.A.; Weis, W.I.; Dunn, A.R. The C-terminal actin-binding domain of talin forms an asymmetric catch bond with F-actin. Proc. Natl. Acad. Sci. USA 2022, 119, e2109329119. [Google Scholar] [CrossRef]
  163. Murayama, K.; Kato-Murayama, M.; Mishima, C.; Akasaka, R.; Shirouzu, M.; Fukui, Y.; Yokoyama, S. Crystal structure of the Bruton’s tyrosine kinase PH domain with phosphatidylinositol. Biochem. Biophys. Res. Commun. 2008, 377, 23–28. [Google Scholar] [CrossRef]
  164. Fukuda, M.; Kojima, T.; Kabayama, H.; Mikoshiba, K. Mutation of the pleckstrin homology domain of Bruton’s tyrosine kinase in immunodeficiency impaired inositol 1,3,4,5-tetrakisphosphate binding capacity. J. Biol. Chem. 1996, 271, 30303–30306. [Google Scholar] [CrossRef] [PubMed]
  165. Lee, S.B.; Várnai, P.; Balla, A.; Jalink, K.; Rhee, S.G.; Balla, T. The pleckstrin homology domain of phosphoinositide-specific phospholipase Cdelta4 is not a critical determinant of the membrane localization of the enzyme. J. Biol. Chem. 2004, 279, 24362–24371. [Google Scholar] [CrossRef]
  166. Overduin, M.; Kervin, T.A. The phosphoinositide code is read by a plethora of protein domains. Expert Rev. Proteom. 2021, 18, 483–502. [Google Scholar] [CrossRef]
  167. Hirsch, E.; Gulluni, F.; Martini, M. Phosphoinositides in cell proliferation and metabolism. Adv. Biol. Regul. 2020, 75, 100693. [Google Scholar] [CrossRef] [PubMed]
  168. Bridges, D.; Saltiel, A.R. Phosphoinositides: Key modulators of energy metabolism. Biochim. Biophys. Acta 2015, 1851, 857–866. [Google Scholar] [CrossRef]
  169. Beziau, A.; Brand, D.; Piver, E. The Role of Phosphatidylinositol Phosphate Kinases during Viral Infection. Viruses 2020, 12, 1124. [Google Scholar] [CrossRef] [PubMed]
  170. Burke, J.E. Structural Basis for Regulation of Phosphoinositide Kinases and Their Involvement in Human Disease. Mol. Cell 2018, 71, 653–673. [Google Scholar] [CrossRef] [PubMed]
  171. Jackson, C.L.; Walch, L.; Verbavatz, J.M. Lipids and Their Trafficking: An Integral Part of Cellular Organization. Dev. Cell 2016, 39, 139–153. [Google Scholar] [CrossRef]
  172. Toker, A.; Cantley, L.C. Signalling through the lipid products of phosphoinositide-3-OH kinase. Nature 1997, 387, 673–676. [Google Scholar] [CrossRef]
  173. Liu, P.; Cheng, H.; Roberts, T.M.; Zhao, J.J. Targeting the phosphoinositide 3-kinase pathway in cancer. Nat. Rev. Drug Discov. 2009, 8, 627–644. [Google Scholar] [CrossRef]
  174. Desale, S.E.; Chidambaram, H.; Chinnathambi, S. G-protein coupled receptor, PI3K and Rho signaling pathways regulate the cascades of Tau and amyloid-β in Alzheimer’s disease. Mol. Biomed. 2021, 2, 17. [Google Scholar] [CrossRef]
  175. Taichman, R.; Merida, I.; Torigoe, T.; Gaulton, G.N.; Reed, J.C. Evidence that protein tyrosine kinase p56-Lck regulates the activity of phosphatidylinositol-3’-kinase in interleukin-2-dependent T-cells. J. Biol. Chem. 1993, 268, 20031–20036. [Google Scholar] [CrossRef]
  176. Karnitz, L.M.; Sutor, S.L.; Abraham, R.T. The Src-family kinase, Fyn, regulates the activation of phosphatidylinositol 3-kinase in an interleukin 2-responsive T cell line. J. Exp. Med. 1994, 179, 1799–1808. [Google Scholar] [CrossRef]
  177. Tang, X.; Feng, Y.; Ye, K. Src-family tyrosine kinase fyn phosphorylates phosphatidylinositol 3-kinase enhancer-activating Akt, preventing its apoptotic cleavage and promoting cell survival. Cell Death Differ. 2007, 14, 368–377. [Google Scholar] [CrossRef]
  178. Peng, S.; Fu, Y. FYN: Emerging biological roles and potential therapeutic targets in cancer. J. Transl. Med. 2023, 21, 84. [Google Scholar] [CrossRef]
  179. Matrone, C.; Petrillo, F.; Nasso, R.; Ferretti, G. Fyn Tyrosine Kinase as Harmonizing Factor in Neuronal Functions and Dysfunctions. Int. J. Mol. Sci. 2020, 21, 4444. [Google Scholar] [CrossRef] [PubMed]
  180. Goldsmith, J.F.; Hall, C.G.; Atkinson, T.P. Identification of an alternatively spliced isoform of the fyn tyrosine kinase. Biochem. Biophys. Res. Commun. 2002, 298, 501–504. [Google Scholar] [CrossRef]
  181. Vatish, M.; Yamada, E.; Pessin, J.E.; Bastie, C.C. Fyn kinase function in lipid utilization: A new upstream regulator of AMPK activity? Arch. Physiol. Biochem. 2009, 115, 191–198. [Google Scholar] [CrossRef] [PubMed]
  182. Baer, A.; Colon-Moran, W.; Xiang, J.; Stapleton, J.T.; Bhattarai, N. Src-family kinases negatively regulate NFAT signaling in resting human T cells. PLoS ONE 2017, 12, e0187123. [Google Scholar] [CrossRef]
  183. Chong, Y.P.; Mulhern, T.D.; Zhu, H.J.; Fujita, D.J.; Bjorge, J.D.; Tantiongco, J.P.; Sotirellis, N.; Lio, D.S.; Scholz, G.; Cheng, H.C. A novel non-catalytic mechanism employed by the C-terminal Src-homologous kinase to inhibit Src-family kinase activity. J. Biol. Chem. 2004, 279, 20752–20766. [Google Scholar] [CrossRef]
  184. Advani, G.; Lim, Y.C.; Catimel, B.; Lio, D.S.S.; Ng, N.L.Y.; Chüeh, A.C.; Tran, M.; Anasir, M.I.; Verkade, H.; Zhu, H.J.; et al. Csk-homologous kinase (Chk) is an efficient inhibitor of Src-family kinases but a poor catalyst of phosphorylation of their C-terminal regulatory tyrosine. Cell Commun. Signal. 2017, 15, 29. [Google Scholar] [CrossRef]
  185. den Hertog, J.; Ostman, A.; Böhmer, F.D. Protein tyrosine phosphatases: Regulatory mechanisms. FEBS J. 2008, 275, 831–847. [Google Scholar] [CrossRef]
  186. Vacaresse, N.; Møller, B.; Danielsen, E.M.; Okada, M.; Sap, J. Activation of c-Src and Fyn kinases by protein-tyrosine phosphatase RPTPalpha is substrate-specific and compatible with lipid raft localization. J. Biol. Chem. 2008, 283, 35815–35824. [Google Scholar] [CrossRef]
  187. Liang, X.; Lu, Y.; Wilkes, M.; Neubert, T.A.; Resh, M.D. The N-terminal SH4 region of the Src family kinase Fyn is modified by methylation and heterogeneous fatty acylation: Role in membrane targeting, cell adhesion, and spreading. J. Biol. Chem. 2004, 279, 8133–8139. [Google Scholar] [CrossRef]
  188. Boggon, T.J.; Eck, M.J. Structure and regulation of Src family kinases. Oncogene 2004, 23, 7918–7927. [Google Scholar] [CrossRef]
  189. Teutschbein, J.; Schartl, M.; Meierjohann, S. Interaction of Xiphophorus and murine Fyn with focal adhesion kinase. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2009, 149, 168–174. [Google Scholar] [CrossRef]
  190. Zhao, X.; Guan, J.L. Focal adhesion kinase and its signaling pathways in cell migration and angiogenesis. Adv. Drug Deliv. Rev. 2011, 63, 610–615. [Google Scholar] [CrossRef] [PubMed]
  191. Meierjohann, S.; Wende, E.; Kraiss, A.; Wellbrock, C.; Schartl, M. The oncogenic epidermal growth factor receptor variant Xiphophorus melanoma receptor kinase induces motility in melanocytes by modulation of focal adhesions. Cancer Res. 2006, 66, 3145–3152. [Google Scholar] [CrossRef]
  192. Wittbrodt, J.; Adam, D.; Malitschek, B.; Mäueler, W.; Raulf, F.; Telling, A.; Robertson, S.M.; Schartl, M. Novel putative receptor tyrosine kinase encoded by the melanoma-inducing Tu locus in Xiphophorus. Nature 1989, 341, 415–421. [Google Scholar] [CrossRef] [PubMed]
  193. Wittbrodt, J.; Lammers, R.; Malitschek, B.; Ullrich, A.; Schartl, M. The Xmrk receptor tyrosine kinase is activated in Xiphophorus malignant melanoma. EMBO J. 1992, 11, 4239–4246. [Google Scholar] [CrossRef] [PubMed]
  194. Wellbrock, C.; Schartl, M. Activation of phosphatidylinositol 3-kinase by a complex of p59fyn and the receptor tyrosine kinase Xmrk is involved in malignant transformation of pigment cells. Eur. J. Biochem. 2000, 267, 3513–3522. [Google Scholar] [CrossRef]
  195. Wellbrock, C.; Schartl, M. Multiple binding sites in the growth factor receptor Xmrk mediate binding to p59fyn, GRB2 and Shc. Eur. J. Biochem. 1999, 260, 275–283. [Google Scholar] [CrossRef] [PubMed]
  196. Alexanian, A.; Miller, B.; Chesnik, M.; Mirza, S.; Sorokin, A. Post-translational regulation of COX2 activity by FYN in prostate cancer cells. Oncotarget 2014, 5, 4232–4243. [Google Scholar] [CrossRef] [PubMed]
  197. Czarnecka, A.M.; Bartnik, E.; Fiedorowicz, M.; Rutkowski, P. Targeted Therapy in Melanoma and Mechanisms of Resistance. Int. J. Mol. Sci. 2020, 21, 4576. [Google Scholar] [CrossRef]
  198. Denley, A.; Gymnopoulos, M.; Kang, S.; Mitchell, C.; Vogt, P.K. Requirement of phosphatidylinositol(3,4,5)trisphosphate in phosphatidylinositol 3-kinase-induced oncogenic transformation. Mol. Cancer Res. 2009, 7, 1132–1138. [Google Scholar] [CrossRef]
  199. Lien, E.C.; Dibble, C.C.; Toker, A. PI3K signaling in cancer: Beyond AKT. Curr. Opin. Cell Biol. 2017, 45, 62–71. [Google Scholar] [CrossRef]
  200. Dieterle, A.M.; Böhler, P.; Keppeler, H.; Alers, S.; Berleth, N.; Drießen, S.; Hieke, N.; Pietkiewicz, S.; Löffler, A.S.; Peter, C.; et al. PDK1 controls upstream PI3K expression and PIP3 generation. Oncogene 2014, 33, 3043–3053. [Google Scholar] [CrossRef]
  201. Orlacchio, A.; Ranieri, M.; Brave, M.; Arciuch, V.A.; Forde, T.; De Martino, D.; Anderson, K.E.; Hawkins, P.; Di Cristofano, A. SGK1 Is a Critical Component of an AKT-Independent Pathway Essential for PI3K-Mediated Tumor Development and Maintenance. Cancer Res. 2017, 77, 6914–6926. [Google Scholar] [CrossRef]
  202. Liu, R.; Chen, Y.; Liu, G.; Li, C.; Song, Y.; Cao, Z.; Li, W.; Hu, J.; Lu, C.; Liu, Y. PI3K/AKT pathway as a key link modulates the multidrug resistance of cancers. Cell Death Dis. 2020, 11, 797. [Google Scholar] [CrossRef]
  203. Loewith, R.; Hall, M.N. Target of rapamycin (TOR) in nutrient signaling and growth control. Genetics 2011, 189, 1177–1201. [Google Scholar] [CrossRef]
  204. Thoreen, C.C.; Kang, S.A.; Chang, J.W.; Liu, Q.; Zhang, J.; Gao, Y.; Reichling, L.J.; Sim, T.; Sabatini, D.M.; Gray, N.S. An ATP-competitive mammalian target of rapamycin inhibitor reveals rapamycin-resistant functions of mTORC1. J. Biol. Chem. 2009, 284, 8023–8032. [Google Scholar] [CrossRef]
  205. Dibble, C.C.; Cantley, L.C. Regulation of mTORC1 by PI3K signaling. Trends Cell Biol. 2015, 25, 545–555. [Google Scholar] [CrossRef]
  206. Kim, L.C.; Cook, R.S.; Chen, J. mTORC1 and mTORC2 in cancer and the tumor microenvironment. Oncogene 2017, 36, 2191–2201. [Google Scholar] [CrossRef]
  207. Yang, H.; Rudge, D.G.; Koos, J.D.; Vaidialingam, B.; Yang, H.J.; Pavletich, N.P. mTOR kinase structure, mechanism and regulation. Nature 2013, 497, 217–223. [Google Scholar] [CrossRef]
  208. Sharma, N.; Nanta, R.; Sharma, J.; Gunewardena, S.; Singh, K.P.; Shankar, S.; Srivastava, R.K. PI3K/AKT/mTOR and sonic hedgehog pathways cooperate together to inhibit human pancreatic cancer stem cell characteristics and tumor growth. Oncotarget 2015, 6, 32039–32060. [Google Scholar] [CrossRef]
  209. Chamcheu, J.C.; Roy, T.; Uddin, M.B.; Banang-Mbeumi, S.; Chamcheu, R.N.; Walker, A.L.; Liu, Y.Y.; Huang, S. Role and Therapeutic Targeting of the PI3K/Akt/mTOR Signaling Pathway in Skin Cancer: A Review of Current Status and Future Trends on Natural and Synthetic Agents Therapy. Cells 2019, 8, 803. [Google Scholar] [CrossRef]
  210. Mafi, S.; Mansoori, B.; Taeb, S.; Sadeghi, H.; Abbasi, R.; Cho, W.C.; Rostamzadeh, D. mTOR-Mediated Regulation of Immune Responses in Cancer and Tumor Microenvironment. Front. Immunol. 2021, 12, 774103. [Google Scholar] [CrossRef] [PubMed]
  211. Wei, Y.; Jiang, Y.; Zou, F.; Liu, Y.; Wang, S.; Xu, N.; Xu, W.; Cui, C.; Xing, Y.; Cao, B.; et al. Activation of PI3K/Akt pathway by CD133-p85 interaction promotes tumorigenic capacity of glioma stem cells. Proc. Natl. Acad. Sci. USA 2013, 110, 6829–6834. [Google Scholar] [CrossRef] [PubMed]
  212. Wang, X.; Regufe da Mota, S.; Liu, R.; Moore, C.E.; Xie, J.; Lanucara, F.; Agarwala, U.; Pyr Dit Ruys, S.; Vertommen, D.; Rider, M.H.; et al. Eukaryotic elongation factor 2 kinase activity is controlled by multiple inputs from oncogenic signaling. Mol. Cell. Biol. 2014, 34, 4088–4103. [Google Scholar] [CrossRef] [PubMed]
  213. Hijazi, M.; Casado, P.; Akhtar, N.; Alvarez-Teijeiro, S.; Rajeeve, V.; Cutillas, P.R. eEF2K Activity Determines Synergy to Cotreatment of Cancer Cells With PI3K and MEK Inhibitors. Mol. Cell. Proteom. 2022, 21, 100240. [Google Scholar] [CrossRef] [PubMed]
  214. Bracho-Valdés, I.; Moreno-Alvarez, P.; Valencia-Martínez, I.; Robles-Molina, E.; Chávez-Vargas, L.; Vázquez-Prado, J. mTORC1- and mTORC2-interacting proteins keep their multifunctional partners focused. IUBMB Life 2011, 63, 896–914. [Google Scholar] [CrossRef] [PubMed]
  215. Origanti, S.; Nowotarski, S.L.; Carr, T.D.; Sass-Kuhn, S.; Xiao, L.; Wang, J.Y.; Shantz, L.M. Ornithine decarboxylase mRNA is stabilized in an mTORC1-dependent manner in Ras-transformed cells. Biochem. J. 2012, 442, 199–207. [Google Scholar] [CrossRef]
  216. Cam, H.; Easton, J.B.; High, A.; Houghton, P.J. mTORC1 signaling under hypoxic conditions is controlled by ATM-dependent phosphorylation of HIF-1α. Mol. Cell 2010, 40, 509–520. [Google Scholar] [CrossRef]
  217. Wong, P.M.; Feng, Y.; Wang, J.; Shi, R.; Jiang, X. Regulation of autophagy by coordinated action of mTORC1 and protein phosphatase 2A. Nat. Commun. 2015, 6, 8048. [Google Scholar] [CrossRef]
  218. Peterson, T.R.; Sengupta, S.S.; Harris, T.E.; Carmack, A.E.; Kang, S.A.; Balderas, E.; Guertin, D.A.; Madden, K.L.; Carpenter, A.E.; Finck, B.N.; et al. mTOR complex 1 regulates lipin 1 localization to control the SREBP pathway. Cell 2011, 146, 408–420. [Google Scholar] [CrossRef]
  219. Muscella, A.; Vetrugno, C.; Calabriso, N.; Cossa, L.G.; De Pascali, S.A.; Fanizzi, F.P.; Marsigliante, S. [Pt(O,O’-acac)(γ-acac)(DMS)] alters SH-SY5Y cell migration and invasion by the inhibition of Na+/H+ exchanger isoform 1 occurring through a PKC-ε/ERK/mTOR Pathway. PLoS ONE 2014, 9, e112186. [Google Scholar] [CrossRef] [PubMed]
  220. Huang, S.; Liu, L.N.; Hosoi, H.; Dilling, M.B.; Shikata, T.; Houghton, P.J. p53/p21(CIP1) cooperate in enforcing rapamycin-induced G(1) arrest and determine the cellular response to rapamycin. Cancer Res. 2001, 61, 3373–3381. [Google Scholar] [PubMed]
  221. Nourse, J.; Firpo, E.; Flanagan, W.M.; Coats, S.; Polyak, K.; Lee, M.H.; Massague, J.; Crabtree, G.R.; Roberts, J.M. Interleukin-2-mediated elimination of the p27Kip1 cyclin-dependent kinase inhibitor prevented by rapamycin. Nature 1994, 372, 570–573. [Google Scholar] [CrossRef] [PubMed]
  222. Zhang, J.; Xu, K.; Liu, P.; Geng, Y.; Wang, B.; Gan, W.; Guo, J.; Wu, F.; Chin, Y.R.; Berrios, C.; et al. Inhibition of Rb Phosphorylation Leads to mTORC2-Mediated Activation of Akt. Mol. Cell 2016, 62, 929–942. [Google Scholar] [CrossRef]
  223. Massi, D.; Mihic-Probst, D.; Schadendorf, D.; Dummer, R.; Mandalà, M. Dedifferentiated melanomas: Morpho-phenotypic profile, genetic reprogramming and clinical implications. Cancer Treat. Rev. 2020, 88, 102060. [Google Scholar] [CrossRef]
  224. Rodrik-Outmezguine, V.S.; Chandarlapaty, S.; Pagano, N.C.; Poulikakos, P.I.; Scaltriti, M.; Moskatel, E.; Baselga, J.; Guichard, S.; Rosen, N. mTOR kinase inhibition causes feedback-dependent biphasic regulation of AKT signaling. Cancer Discov. 2011, 1, 248–259. [Google Scholar] [CrossRef] [PubMed]
  225. Mercurio, L.; Albanesi, C.; Madonna, S. Recent Updates on the Involvement of PI3K/AKT/mTOR Molecular Cascade in the Pathogenesis of Hyperproliferative Skin Disorders. Front. Med. 2021, 8, 665647. [Google Scholar] [CrossRef]
  226. Urso, C. Are growth phases exclusive to cutaneous melanoma? J. Clin. Pathol. 2004, 57, 560. [Google Scholar] [CrossRef]
  227. Ciarletta, P.; Foret, L.; Ben Amar, M. The radial growth phase of malignant melanoma: Multi-phase modelling, numerical simulations and linear stability analysis. J. R. Soc. Interface 2011, 8, 345–368. [Google Scholar] [CrossRef]
  228. Sinha, S.; Singh, S.K.; Jangde, N.; Ray, R.; Rai, V. p32 promotes melanoma progression and metastasis by targeting EMT markers, Akt/PKB pathway, and tumor microenvironment. Cell Death Dis. 2021, 12, 1012. [Google Scholar] [CrossRef] [PubMed]
  229. Zbytek, B.; Carlson, J.A.; Granese, J.; Ross, J.; Mihm, M.C.; Slominski, A. Current concepts of metastasis in melanoma. Expert Rev. Dermatol. 2008, 3, 569–585. [Google Scholar] [CrossRef]
  230. Lo, J.A.; Fisher, D.E. The melanoma revolution: From UV carcinogenesis to a new era in therapeutics. Science 2014, 346, 945–949. [Google Scholar] [CrossRef]
  231. Sample, A.; He, Y.Y. Mechanisms and prevention of UV-induced melanoma. Photodermatol. Photoimmunol. Photomed. 2018, 34, 13–24. [Google Scholar] [CrossRef]
  232. Piérard, G.E. Cell proliferation in cutaneous malignant melanoma: Relationship with neoplastic progression. Int. Sch. Res. Not. Dermatol. 2012, 2012, 828146. [Google Scholar] [CrossRef]
  233. Leonardi, G.C.; Falzone, L.; Salemi, R.; Zanghì, A.; Spandidos, D.A.; Mccubrey, J.A.; Candido, S.; Libra, M. Cutaneous melanoma: From pathogenesis to therapy (Review). Int. J. Oncol. 2018, 52, 1071–1080. [Google Scholar] [CrossRef] [PubMed]
  234. Li, Y.C.; He, S.M.; He, Z.X.; Li, M.; Yang, Y.; Pang, J.X.; Zhang, X.; Chow, K.; Zhou, Q.; Duan, W.; et al. Plumbagin induces apoptotic and autophagic cell death through inhibition of the PI3K/Akt/mTOR pathway in human non-small cell lung cancer cells. Cancer Lett. 2014, 344, 239–259. [Google Scholar] [CrossRef]
  235. Xie, X.; White, E.P.; Mehnert, J.M. Coordinate autophagy and mTOR pathway inhibition enhances cell death in melanoma. PLoS ONE 2013, 8, e55096. [Google Scholar] [CrossRef] [PubMed]
  236. Shtivelman, E.; Davies, M.Q.; Hwu, P.; Yang, J.; Lotem, M.; Oren, M.; Flaherty, K.T.; Fisher, D.E. Pathways and therapeutic targets in melanoma. Oncotarget 2014, 5, 1701–1752. [Google Scholar] [CrossRef] [PubMed]
  237. Tentori, L.; Lacal, P.M.; Graziani, G. Challenging resistance mechanisms to therapies for metastatic melanoma. Trends Pharmacol. Sci. 2013, 34, 656–666. [Google Scholar] [CrossRef] [PubMed]
  238. Wang, B.; Zhang, W.; Zhang, G.; Kwong, L.; Lu, H.; Tan, J.; Sadek, N.; Xiao, M.; Zhang, J.; Labrie, M.; et al. Targeting mTOR signaling overcomes acquired resistance to combined BRAF and MEK inhibition in BRAF-mutant melanoma. Oncogene 2021, 40, 5590–5599. [Google Scholar] [CrossRef] [PubMed]
  239. Fenton, S.E.; Sosman, J.A.; Chandra, S. Resistance mechanisms in melanoma to immuneoncologic therapy with checkpoint inhibitors. Cancer Drug Resist. 2019, 2, 744–761. [Google Scholar] [CrossRef] [PubMed]
  240. Patel, M.; Eckburg, A.; Gantiwala, S.; Hart, Z.; Dein, J.; Lam, K.; Puri, N. Resistance to Molecularly Targeted Therapies in Melanoma. Cancers 2021, 13, 1115. [Google Scholar] [CrossRef]
  241. Kearney, A.L.; Norris, D.M.; Ghomlaghi, M.; Kin Lok Wong, M.; Humphrey, S.J.; Carroll, L.; Yang, G.; Cooke, K.C.; Yang, P.; Geddes, T.A.; et al. Akt phosphorylates insulin receptor substrate to limit PI3K-mediated PIP3 synthesis. Elife 2021, 10, e66942. [Google Scholar] [CrossRef] [PubMed]
  242. Suleymanova, N.; Crudden, C.; Worrall, C.; Dricu, A.; Girnita, A.; Girnita, L. Enhanced response of melanoma cells to MEK inhibitors following unbiased IGF-1R down-regulation. Oncotarget 2017, 8, 82256–82267. [Google Scholar] [CrossRef] [PubMed]
  243. Leroy, C.; Ramos, P.; Cornille, K.; Bonenfant, D.; Fritsch, C.; Voshol, H.; Bentires-Alj, M. Activation of IGF1R/p110β/AKT/mTOR confers resistance to α-specific PI3K inhibition. Breast Cancer Res. 2016, 18, 41. [Google Scholar] [CrossRef] [PubMed]
  244. Hemmings, B.A.; Restuccia, D.F. PI3K-PKB/Akt pathway. Cold Spring Harb. Perspect. Biol. 2012, 4, a011189. [Google Scholar] [CrossRef]
  245. Glaviano, A.; Foo, A.S.C.; Lam, H.Y.; Yap, K.C.H.; Jacot, W.; Jones, R.H.; Eng, H.; Nair, M.G.; Makvandi, P.; Geoerger, B.; et al. PI3K/AKT/mTOR signaling transduction pathway and targeted therapies in cancer. Mol. Cancer 2023, 22, 138. [Google Scholar] [CrossRef] [PubMed]
  246. Chiarini, F.; Evangelisti, C.; Lattanzi, G.; McCubrey, J.A.; Martelli, A.M. Advances in understanding the mechanisms of evasive and innate resistance to mTOR inhibition in cancer cells. Biochim. Biophys. Acta Mol. Cell Res. 2019, 1866, 1322–1337. [Google Scholar] [CrossRef]
  247. Steelman, L.S.; Chappell, W.H.; Abrams, S.L.; Kempf, R.C.; Long, J.; Laidler, P.; Mijatovic, S.; Maksimovic-Ivanic, D.; Stivala, F.; Mazzarino, M.C.; et al. Roles of the Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR pathways in controlling growth and sensitivity to therapy-implications for cancer and aging. Aging 2011, 3, 192–222. [Google Scholar] [CrossRef]
  248. Li, Q.; Li, Z.; Luo, T.; Shi, H. Targeting the PI3K/AKT/mTOR and RAF/MEK/ERK pathways for cancer therapy. Mol. Biomed. 2022, 3, 47. [Google Scholar] [CrossRef]
  249. Palušová, V.; Renzová, T.; Verlande, A.; Vaclová, T.; Medková, M.; Cetlová, L.; Sedláčková, M.; Hříbková, H.; Slaninová, I.; Krutá, M.; et al. Dual Targeting of BRAF and mTOR Signaling in Melanoma Cells with Pyridinyl Imidazole Compounds. Cancers 2020, 12, 1516. [Google Scholar] [CrossRef]
  250. Carlino, M.S.; Larkin, J.; Long, G.V. Immune checkpoint inhibitors in melanoma. Lancet 2021, 398, 1002–1014. [Google Scholar] [CrossRef]
  251. Fukuda, K.; Okamura, K.; Riding, R.L.; Fan, X.; Afshari, K.; Haddadi, N.S.; McCauley, S.M.; Guney, M.H.; Luban, J.; Funakoshi, T.; et al. AIM2 regulates anti-tumor immunity and is a viable therapeutic target for melanoma. J. Exp. Med. 2021, 218, e20200962. [Google Scholar] [CrossRef]
  252. Weber, R.; Fleming, V.; Hu, X.; Nagibin, V.; Groth, C.; Altevogt, P.; Utikal, J.; Umansky, V. Myeloid-Derived Suppressor Cells Hinder the Anti-Cancer Activity of Immune Checkpoint Inhibitors. Front. Immunol. 2018, 9, 1310. [Google Scholar] [CrossRef]
  253. Simiczyjew, A.; Dratkiewicz, E.; Mazurkiewicz, J.; Ziętek, M.; Matkowski, R.; Nowak, D. The Influence of Tumor Microenvironment on Immune Escape of Melanoma. Int. J. Mol. Sci. 2020, 21, 8359. [Google Scholar] [CrossRef] [PubMed]
  254. Deken, M.A.; Gadiot, J.; Jordanova, E.S.; Lacroix, R.; van Gool, M.; Kroon, P.; Pineda, C.; Geukes Foppen, M.H.; Scolyer, R.; Song, J.Y.; et al. Targeting the MAPK and PI3K pathways in combination with PD1 blockade in melanoma. Oncoimmunology 2016, 5, e1238557. [Google Scholar] [CrossRef] [PubMed]
  255. Rager, T.; Eckburg, A.; Patel, M.; Qiu, R.; Gantiwala, S.; Dovalovsky, K.; Fan, K.; Lam, K.; Roesler, C.; Rastogi, A.; et al. Treatment of Metastatic Melanoma with a Combination of Immunotherapies and Molecularly Targeted Therapies. Cancers 2022, 14, 3779. [Google Scholar] [CrossRef] [PubMed]
  256. Aasen, S.N.; Parajuli, H.; Hoang, T.; Feng, Z.; Stokke, K.; Wang, J.; Roy, K.; Bjerkvig, R.; Knappskog, S.; Thorsen, F. Effective Treatment of Metastatic Melanoma by Combining MAPK and PI3K Signaling Pathway Inhibitors. Int. J. Mol. Sci. 2019, 20, 4235. [Google Scholar] [CrossRef] [PubMed]
  257. Tang, L.; Long, J.; Li, K.; Zhang, X.; Chen, X.; Peng, C. A novel chalcone derivative suppresses melanoma cell growth through targeting Fyn/Stat3 pathway. Cancer Cell Int. 2020, 20, 256. [Google Scholar] [CrossRef]
  258. Gangadhar, T.C.; Clark, J.I.; Karrison, T.; Gajewski, T.F. Phase II study of the Src kinase inhibitor saracatinib 260-Gangadhar, T.C.; Clark, J.I.; Karrison, T.; Gajewski, T.F. Phase II study of the Src kinase inhibitor saracatinib (AZD0530) in metastatic melanoma. Investig. New Drugs 2013, 31, 769–773. [Google Scholar] [CrossRef]
  259. Schneider, P.; Schön, M.; Pletz, N.; Seitz, C.S.; Liu, N.; Ziegelbauer, K.; Zachmann, K.; Emmert, S.; Schön, M.P. The novel PI3 kinase inhibitor, BAY 80-6946, impairs melanoma growth in vivo and in vitro. Exp. Dermatol. 2014, 23, 579–584. [Google Scholar] [CrossRef]
  260. Amaral, T.; Niessner, H.; Sinnberg, T.; Thomas, I.; Meiwes, A.; Garbe, C.; Garzarolli, M.; Rauschenberg, R.; Eigentler, T.; Meier, F. An open-label, single-arm, phase II trial of buparlisib in patients with melanoma brain metastases not eligible for surgery or radiosurgery-the BUMPER study. Neurooncol. Adv. 2020, 2, vdaa140. [Google Scholar] [CrossRef]
  261. Tran, K.B.; Kolekar, S.; Jabed, A.; Jaynes, P.; Shih, J.H.; Wang, Q.; Flanagan, J.U.; Rewcastle, G.W.; Baguley, B.C.; Shepherd, P.R. Diverse mechanisms activate the PI 3-kinase/mTOR pathway in melanomas: Implications for the use of PI 3-kinase inhibitors to overcome resistance to inhibitors of BRAF and MEK. BMC Cancer 2021, 21, 136. [Google Scholar] [CrossRef] [PubMed]
  262. Ernst, D.S.; Eisenhauer, E.; Wainman, N.; Davis, M.; Lohmann, R.; Baetz, T.; Belanger, K.; Smylie, M. Phase II study of perifosine in previously untreated patients with metastatic melanoma. Investig. New Drugs 2005, 23, 569–575. [Google Scholar] [CrossRef] [PubMed]
  263. Rebecca, V.W.; Massaro, R.R.; Fedorenko, I.V.; Sondak, V.K.; Anderson, A.R.; Kim, E.; Amaravadi, R.K.; Maria-Engler, S.S.; Messina, J.L.; Gibney, G.T.; et al. Inhibition of autophagy enhances the effects of the AKT inhibitor MK-2206 when combined with paclitaxel and carboplatin in BRAF wild-type melanoma. Pigment Cell Melanoma Res. 2014, 27, 465–478. [Google Scholar] [CrossRef] [PubMed]
  264. Sznol, J.A.; Jilaveanu, L.B.; Kluger, H.M. Studies of NVP-BEZ235 in melanoma. Curr. Cancer Drug Targets 2013, 13, 165–174. [Google Scholar] [CrossRef] [PubMed]
  265. Bernard, S.; Poon, A.C.; Tam, P.M.; Mutsaers, A.J. Investigation of the effects of mTOR inhibitors rapamycin and everolimus in combination with carboplatin on canine malignant melanoma cells. BMC Vet. Res. 2021, 17, 382. [Google Scholar] [CrossRef]
  266. Slingluff, C.L.; Petroni, G.R.; Molhoek, K.R.; Brautigan, D.L.; Chianese-Bullock, K.A.; Shada, A.L.; Smolkin, M.E.; Olson, W.C.; Gaucher, A.; Chase, C.M.; et al. Clinical activity and safety of combination therapy with temsirolimus and bevacizumab for advanced melanoma: A phase II trial (CTEP 7190/Mel47). Clin. Cancer Res. 2013, 19, 3611–3620. [Google Scholar] [CrossRef]
  267. Gopal, Y.N.; Rizos, H.; Chen, G.; Deng, W.; Frederick, D.T.; Cooper, Z.A.; Scolyer, R.A.; Pupo, G.; Komurov, K.; Sehgal, V.; et al. Inhibition of mTORC1/2 overcomes resistance to MAPK pathway inhibitors mediated by PGC1α and oxidative phosphorylation in melanoma. Cancer Res. 2014, 74, 7037–7047. [Google Scholar] [CrossRef]
  268. Espona-Fiedler, M.; Soto-Cerrato, V.; Hosseini, A.; Lizcano, J.M.; Guallar, V.; Quesada, R.; Gao, T.; Pérez-Tomás, R. Identification of dual mTORC1 and mTORC2 inhibitors in melanoma cells: Prodigiosin vs. obatoclax. Biochem. Pharmacol. 2012, 83, 489–496. [Google Scholar] [CrossRef]
Figure 1. Overview of the genomic and non-genomic-dependent mechanisms regulating the development of melanoma heterogeneity and the biological consequences.
Figure 1. Overview of the genomic and non-genomic-dependent mechanisms regulating the development of melanoma heterogeneity and the biological consequences.
Cells 13 00240 g001
Figure 2. Cellular plasticity in melanoma. Melanoma cells can switch between a differentiated/proliferative/cancer stem cells (CSCs) and a dedifferentiated/invasive phenotype/non-stem cancer cells via mechanism-mediated. Melanoma plasticity is mediated by genetic and epigenetic alterations of melanoma cells and by tumor microenvironmental secretory products (e.g., growth factors and cytokines), activation of cancer-associated fibroblasts (CAFs), or tumor-associated macrophages (TAMs). Cancer cells can be reprogrammed towards pluripotency. Phenotype switch of melanoma increases their plasticity and is responsible for tumor growth, invasion, and drug resistance.
Figure 2. Cellular plasticity in melanoma. Melanoma cells can switch between a differentiated/proliferative/cancer stem cells (CSCs) and a dedifferentiated/invasive phenotype/non-stem cancer cells via mechanism-mediated. Melanoma plasticity is mediated by genetic and epigenetic alterations of melanoma cells and by tumor microenvironmental secretory products (e.g., growth factors and cytokines), activation of cancer-associated fibroblasts (CAFs), or tumor-associated macrophages (TAMs). Cancer cells can be reprogrammed towards pluripotency. Phenotype switch of melanoma increases their plasticity and is responsible for tumor growth, invasion, and drug resistance.
Cells 13 00240 g002
Figure 3. Model for the development of cancer stem cells (CSCs) from both tumor and normal cells. (A) Development of CSCs from cancer cells is mediated by the activation of aberrant signaling pathways via driver mutation-dependent mechanisms and transcriptional activation of self-renewal genes. Cancer cells become programmed to divide into cancer progenitor cells and CSCs. Cancer progenitor cells are genetically programmed to divide into one differentiated cell and one CSC. Once the dedifferentiation process of the differentiated cell has been completed, it can be transformed into a CSC that ultimately undergoes abnormal asymmetric cell division to produce genetically divergent subpopulations including CSCs and non-CSCs. (B) The development of CSCs from the activation of self-renewal genes normal stem/progenitor cells via multiple genetic mutations and dedifferentiation-dependent mechanisms. Like cancer cell-derived CSCs, CSCs derived from normal stem/progenitor cells undergo abnormal asymmetric cell division to produce two genetically divergent subpopulations including CSC and non-CSC.
Figure 3. Model for the development of cancer stem cells (CSCs) from both tumor and normal cells. (A) Development of CSCs from cancer cells is mediated by the activation of aberrant signaling pathways via driver mutation-dependent mechanisms and transcriptional activation of self-renewal genes. Cancer cells become programmed to divide into cancer progenitor cells and CSCs. Cancer progenitor cells are genetically programmed to divide into one differentiated cell and one CSC. Once the dedifferentiation process of the differentiated cell has been completed, it can be transformed into a CSC that ultimately undergoes abnormal asymmetric cell division to produce genetically divergent subpopulations including CSCs and non-CSCs. (B) The development of CSCs from the activation of self-renewal genes normal stem/progenitor cells via multiple genetic mutations and dedifferentiation-dependent mechanisms. Like cancer cell-derived CSCs, CSCs derived from normal stem/progenitor cells undergo abnormal asymmetric cell division to produce two genetically divergent subpopulations including CSC and non-CSC.
Cells 13 00240 g003
Figure 4. Structure of CD133 protein. CD133 is a 97 kDa transmembrane glycoprotein that contains an extracellular N-terminal domain, five transmembrane (TM) segments, two small intracellular loops (IC I and IC II), two extracellular loops (EC I and EC II), and an intracellular C-terminal domain. The two extracellular loops contain nine N-linked glycan (G) residues. The EC loop I contains 5 N-glycosylation sites, while EC II contains 4 glycosylation sites. The intracellular C-terminal domain contains two Tyrosine (Tyr828) and (Tyr852) residues.
Figure 4. Structure of CD133 protein. CD133 is a 97 kDa transmembrane glycoprotein that contains an extracellular N-terminal domain, five transmembrane (TM) segments, two small intracellular loops (IC I and IC II), two extracellular loops (EC I and EC II), and an intracellular C-terminal domain. The two extracellular loops contain nine N-linked glycan (G) residues. The EC loop I contains 5 N-glycosylation sites, while EC II contains 4 glycosylation sites. The intracellular C-terminal domain contains two Tyrosine (Tyr828) and (Tyr852) residues.
Cells 13 00240 g004
Figure 5. Function of CD133 protein. Phosphorylation of Tyr828 and Tyr852 residues of CD133 by non-receptor tyrosine kinase (NRTK) Fyn triggers the activation of PI3K- and NRAS-dependent pathways in melanoma. Upon the phosphorylation of both Tyr828 and Tyr852 residues, located on the cytoplasmic N-terminal domain of CD133, the phosphorylated Tyr828 (pTyr828) and Tyr852 (pTyr852) recruit the regulatory subunit of PI3K, p85, and the adaptor protein Grb2 to mediate the activation of PI3K/PDK-1/AKT and PI3K/AKT/MDM, and NRAS/RAFMEK pathways, respectively. The activated PI3K/PDK-1/AKT pathway induces the inhibition of p27 and the activated PI3K/AKT/MDM2 pathway induces the ubiquitination of p53. Meanwhile, the activated RAS/RAFMEK pathway induces the activation of ERK. Consequently, the activation of Fyn-stimulated signaling to downstream pathways of both PI3K and Grb2 results in melanoma growth and resistance.
Figure 5. Function of CD133 protein. Phosphorylation of Tyr828 and Tyr852 residues of CD133 by non-receptor tyrosine kinase (NRTK) Fyn triggers the activation of PI3K- and NRAS-dependent pathways in melanoma. Upon the phosphorylation of both Tyr828 and Tyr852 residues, located on the cytoplasmic N-terminal domain of CD133, the phosphorylated Tyr828 (pTyr828) and Tyr852 (pTyr852) recruit the regulatory subunit of PI3K, p85, and the adaptor protein Grb2 to mediate the activation of PI3K/PDK-1/AKT and PI3K/AKT/MDM, and NRAS/RAFMEK pathways, respectively. The activated PI3K/PDK-1/AKT pathway induces the inhibition of p27 and the activated PI3K/AKT/MDM2 pathway induces the ubiquitination of p53. Meanwhile, the activated RAS/RAFMEK pathway induces the activation of ERK. Consequently, the activation of Fyn-stimulated signaling to downstream pathways of both PI3K and Grb2 results in melanoma growth and resistance.
Cells 13 00240 g005
Figure 6. Structure of non-receptor tyrosine kinase families and their functional domains. Non-receptor tyrosine kinases (NRTKs) such as ABl, CSK, SYK, Src, TEC, FAK, FES, JAK, and ACK kinases are made up of an N-terminal region and a large C-terminal region. The N-terminus contains the common kinase domain that spans ~300 residues. The N-terminal region of NRTKs contains a number of extra SRC homology 4 (SH4), 3 (SH3), and 2 (SH2), catalytic kinase (SH1), Pleckstrin homology (PH), four-point-one, ezrin, radixin, moesin (FERM), Janus homology (JH2) domain 2, and FES/FER/Cdc-42-interacting protein homology (FCH) domains, with Btk-type zinc finger (Btk), coiled coil (CC) motives, proline-rich region (pr), Janus homology domain 2 (JH2). The C-terminal region of NRTKs contains SH3, CRIB, and Cdc42/Rac-interactive (CRIB) domains as well as DNA-binding (DNA), actin-binding (Actin), and focal adhesion targeting (FAT) domains.
Figure 6. Structure of non-receptor tyrosine kinase families and their functional domains. Non-receptor tyrosine kinases (NRTKs) such as ABl, CSK, SYK, Src, TEC, FAK, FES, JAK, and ACK kinases are made up of an N-terminal region and a large C-terminal region. The N-terminus contains the common kinase domain that spans ~300 residues. The N-terminal region of NRTKs contains a number of extra SRC homology 4 (SH4), 3 (SH3), and 2 (SH2), catalytic kinase (SH1), Pleckstrin homology (PH), four-point-one, ezrin, radixin, moesin (FERM), Janus homology (JH2) domain 2, and FES/FER/Cdc-42-interacting protein homology (FCH) domains, with Btk-type zinc finger (Btk), coiled coil (CC) motives, proline-rich region (pr), Janus homology domain 2 (JH2). The C-terminal region of NRTKs contains SH3, CRIB, and Cdc42/Rac-interactive (CRIB) domains as well as DNA-binding (DNA), actin-binding (Actin), and focal adhesion targeting (FAT) domains.
Cells 13 00240 g006
Figure 7. Fyn kinase structure and regulation. Fyn kinase consists of SH1, SH2, SH3, and SH4 domains. The SH2 domain binds the phosphorylated Tyr528 (pTyr528) in the C-terminus to keep Fyn in an inactive conformation. Tyr528 is dephosphorylated by phosphatases (PTPs) to keep the structure open allowing for phosphorylation of Tyr416 in the catalytic SH1 domain.
Figure 7. Fyn kinase structure and regulation. Fyn kinase consists of SH1, SH2, SH3, and SH4 domains. The SH2 domain binds the phosphorylated Tyr528 (pTyr528) in the C-terminus to keep Fyn in an inactive conformation. Tyr528 is dephosphorylated by phosphatases (PTPs) to keep the structure open allowing for phosphorylation of Tyr416 in the catalytic SH1 domain.
Cells 13 00240 g007
Figure 8. Proposed model for models of both non-receptor tyrosine kinase- (NRTKs) and receptor tyrosine kinase-mediated signaling pathways in tumor progression and drug resistance. (A) NRTK-mediated signaling pathways to tumor progression and drug resistance. NRTK Fyn stimulates Stat3, CD133 signaling to PI3K/AKT/mTOR, PI3K/PDK-1, PI3K/AKT/MDM2, and Grb2/Grb7/RAS/RAF/MEK/ERK pathways in melanoma by the phosphorylation of tyrosine kinase Tyr828 and Tyr852 residues located on the cytoplasmic C-terminal domain of the CD133 protein to recruit both p85 and Grb2 proteins, respectively. P85 serves to mediate Fyn-stimulated CD133 signal to PI3K to enhance the activation of several pathways including PI3K/PDK-1/AKT/mTOR/S6K and/or PI3K/AKT/mTOR/S6K; PI3K/PDK-1/AKT/4E-BP1 and/or PI3K/AKT/4E-BP1; PI3K/PDK-1/AKT/MDM2 and/or PI3K/AKT/MDM2; PI3K/PDK-1/AKT/MKP-1 and/or PI3K/1/AKT/MKP-1. Meanwhile, Grb2serves to mediate Fyn-stimulated CD133 signal to enhance the activation of NRAS/BRAF/MEK/ERK pathway. Finally, the biological consequences of Fyn-stimulated signal to CD133 in melanoma include the enhancement of protein synthesis, cell cycle progression, cell growth, survival, and drug resistance. (B) RTK-mediated signaling pathways to tumor progression and drug resistance. By binding RTKs to their corresponding receptors, tyrosine kinases (TKs) bind ligands to initiate the signaling pathway via intermediate molecule, insulin receptor substrate (IRS). Then, the activated PI3K becomes able to phosphorylate the phosphatidylinositol 4,5-bisphosphate (PIP2) to phosphatidylinositol 3,4,5-trisphosphate (PIP3), a process that is reversed by PTEN. Meanwhile, at the cell membrane, proteins with pleckstrin homology domains are then phosphorylated via PIP3 (phosphoinositide-dependent protein kinase-1 (PDK1) and AKT). Also, PDK1 can phosphorylate critical residues on AKT. The tumor suppressor complex of TSC1/TSC2 normally inhibits mTOR activation via the brain-enriched Ras homologue (Rheb). Activated AKT prevents this inhibition and leads to activation of the mTOR/Raptor complex 1 (mTORC1). This complex can be inhibited by rapamycin and its analogues. Consequently, mTORC1 leads to the activation of downstream proteins involved in the initiation of protein synthesis, leading to cell growth. Also, activation of RTKs able to initiate MAPK pathway signaling, leading to tumor progression, proliferation, and drug resistance. Activation of the MAPK pathway may also enhance PI3K signaling. MEK, MAPK/ERK kinase.
Figure 8. Proposed model for models of both non-receptor tyrosine kinase- (NRTKs) and receptor tyrosine kinase-mediated signaling pathways in tumor progression and drug resistance. (A) NRTK-mediated signaling pathways to tumor progression and drug resistance. NRTK Fyn stimulates Stat3, CD133 signaling to PI3K/AKT/mTOR, PI3K/PDK-1, PI3K/AKT/MDM2, and Grb2/Grb7/RAS/RAF/MEK/ERK pathways in melanoma by the phosphorylation of tyrosine kinase Tyr828 and Tyr852 residues located on the cytoplasmic C-terminal domain of the CD133 protein to recruit both p85 and Grb2 proteins, respectively. P85 serves to mediate Fyn-stimulated CD133 signal to PI3K to enhance the activation of several pathways including PI3K/PDK-1/AKT/mTOR/S6K and/or PI3K/AKT/mTOR/S6K; PI3K/PDK-1/AKT/4E-BP1 and/or PI3K/AKT/4E-BP1; PI3K/PDK-1/AKT/MDM2 and/or PI3K/AKT/MDM2; PI3K/PDK-1/AKT/MKP-1 and/or PI3K/1/AKT/MKP-1. Meanwhile, Grb2serves to mediate Fyn-stimulated CD133 signal to enhance the activation of NRAS/BRAF/MEK/ERK pathway. Finally, the biological consequences of Fyn-stimulated signal to CD133 in melanoma include the enhancement of protein synthesis, cell cycle progression, cell growth, survival, and drug resistance. (B) RTK-mediated signaling pathways to tumor progression and drug resistance. By binding RTKs to their corresponding receptors, tyrosine kinases (TKs) bind ligands to initiate the signaling pathway via intermediate molecule, insulin receptor substrate (IRS). Then, the activated PI3K becomes able to phosphorylate the phosphatidylinositol 4,5-bisphosphate (PIP2) to phosphatidylinositol 3,4,5-trisphosphate (PIP3), a process that is reversed by PTEN. Meanwhile, at the cell membrane, proteins with pleckstrin homology domains are then phosphorylated via PIP3 (phosphoinositide-dependent protein kinase-1 (PDK1) and AKT). Also, PDK1 can phosphorylate critical residues on AKT. The tumor suppressor complex of TSC1/TSC2 normally inhibits mTOR activation via the brain-enriched Ras homologue (Rheb). Activated AKT prevents this inhibition and leads to activation of the mTOR/Raptor complex 1 (mTORC1). This complex can be inhibited by rapamycin and its analogues. Consequently, mTORC1 leads to the activation of downstream proteins involved in the initiation of protein synthesis, leading to cell growth. Also, activation of RTKs able to initiate MAPK pathway signaling, leading to tumor progression, proliferation, and drug resistance. Activation of the MAPK pathway may also enhance PI3K signaling. MEK, MAPK/ERK kinase.
Cells 13 00240 g008
Figure 9. Downstream effectors of RTK-mediated pathways and their biological consequences. Upon receptor tyrosine kinase (RTK) stimulation by their corresponding ligands, both PI3K/Akt/mTOR and Grb2/RAS/RAF/MEK/ERK signaling pathways can be activated in melanoma. The activation of PI3K/AKT and NRAS/RAF/MEK signaling pathways by RTKs can promote several cellular functions via the activation of MDM2, NF-κB, and CREB to enhance p53 degradation as well as promote the transcription of proliferative and anti-apoptotic genes. PI3K/AKT-induced activation of mTOR phosphorylates 4E-BP1, the inhibitor of elF-4E, to allow the release of the active form of elf-4E to initiate the translation processes of angiogenic or cell cycle gene targets. PI3K/AKT can inhibit the phosphorylation of downstream effectors, namely caspase-9, GSK-3ß, p27, and p21 to induce cell cycle progression and inhibition of apoptosis.
Figure 9. Downstream effectors of RTK-mediated pathways and their biological consequences. Upon receptor tyrosine kinase (RTK) stimulation by their corresponding ligands, both PI3K/Akt/mTOR and Grb2/RAS/RAF/MEK/ERK signaling pathways can be activated in melanoma. The activation of PI3K/AKT and NRAS/RAF/MEK signaling pathways by RTKs can promote several cellular functions via the activation of MDM2, NF-κB, and CREB to enhance p53 degradation as well as promote the transcription of proliferative and anti-apoptotic genes. PI3K/AKT-induced activation of mTOR phosphorylates 4E-BP1, the inhibitor of elF-4E, to allow the release of the active form of elf-4E to initiate the translation processes of angiogenic or cell cycle gene targets. PI3K/AKT can inhibit the phosphorylation of downstream effectors, namely caspase-9, GSK-3ß, p27, and p21 to induce cell cycle progression and inhibition of apoptosis.
Cells 13 00240 g009
Figure 10. Selected agents targeting the Fyn/CD133/PI3K/Akt/mTOR pathway. Fyn, PI3KAKT, mTORC1, and mTORC1/mTORC2 inhibitors.
Figure 10. Selected agents targeting the Fyn/CD133/PI3K/Akt/mTOR pathway. Fyn, PI3KAKT, mTORC1, and mTORC1/mTORC2 inhibitors.
Cells 13 00240 g010
Table 1. Studies focused on CD133-dependent P13K/AKT pathway signaling and its link to mTOR activation in melanoma progression.
Table 1. Studies focused on CD133-dependent P13K/AKT pathway signaling and its link to mTOR activation in melanoma progression.
Description of the StudyReferences
Melanoma progression and treatment resistance are mediated by CD133 signaling to the PI3K pathwayJamal et al., 2020 [8]
Inhibition of melanoma growth in an autophagy-dependent manner through inhibition of PI3K/AKT/mTOR signalingGong et al., 2020 [135]
Inhibition of the PI3K/AKT/mTOR pathway can efficiently counteract dabrafenib-induced stimulation of the invasive capacity of melanoma cells with required resistance.Caporali et al., 2014 [133]
Table 2. Studies focused on the reliability of Fyn/CD133/PI3K/mTOR pathway as therapeutic target in melanoma treatment.
Table 2. Studies focused on the reliability of Fyn/CD133/PI3K/mTOR pathway as therapeutic target in melanoma treatment.
Description of the Study Therapeutic TargetReferences
Phase II study of the Src kinase inhibitor saracatinib (AZD0530) in metastatic melanomaNRTK, FynTang et al., 2020; Gangadhar, et al., 2013 [257,258]
Clinical reliability of PI3K pathway as therapeutic target for melanoma treatment PI3KAmaral et al., 2020; Schneider et al., 2014; Tran et al., 2021 [259,260,261]
Clinical relevance of AKT as therapeutic in melanoma treatmentAKTErnst et al., 2005; Rebecca et al., 2014 [262,263]
Target both mTORC1 and mTORC2 based on their clinical reliability in melanoma treatmentmTORC1/mTORC2Sznol et al., 2013; Bernard et al., 2021; Slingluff et al., 2013; Gopal et al., 2014; Espona-Fiedler et al., 2012 [264,265,266,267,268]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kharouf, N.; Flanagan, T.W.; Alamodi, A.A.; Al Hmada, Y.; Hassan, S.-Y.; Shalaby, H.; Santourlidis, S.; Hassan, S.-L.; Haikel, Y.; Megahed, M.; et al. CD133-Dependent Activation of Phosphoinositide 3-Kinase /AKT/Mammalian Target of Rapamycin Signaling in Melanoma Progression and Drug Resistance. Cells 2024, 13, 240. https://doi.org/10.3390/cells13030240

AMA Style

Kharouf N, Flanagan TW, Alamodi AA, Al Hmada Y, Hassan S-Y, Shalaby H, Santourlidis S, Hassan S-L, Haikel Y, Megahed M, et al. CD133-Dependent Activation of Phosphoinositide 3-Kinase /AKT/Mammalian Target of Rapamycin Signaling in Melanoma Progression and Drug Resistance. Cells. 2024; 13(3):240. https://doi.org/10.3390/cells13030240

Chicago/Turabian Style

Kharouf, Naji, Thomas W. Flanagan, Abdulhadi A. Alamodi, Youssef Al Hmada, Sofie-Yasmin Hassan, Hosam Shalaby, Simeon Santourlidis, Sarah-Lilly Hassan, Youssef Haikel, Mossad Megahed, and et al. 2024. "CD133-Dependent Activation of Phosphoinositide 3-Kinase /AKT/Mammalian Target of Rapamycin Signaling in Melanoma Progression and Drug Resistance" Cells 13, no. 3: 240. https://doi.org/10.3390/cells13030240

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop