Next Article in Journal
AhR, PXR and CAR: From Xenobiotic Receptors to Metabolic Sensors
Next Article in Special Issue
Promising and Minimally Invasive Biomarkers: Targeting Melanoma
Previous Article in Journal
NK92 Expressing Anti-BCMA CAR and Secreted TRAIL for the Treatment of Multiple Myeloma: Preliminary In Vitro Assessment
Previous Article in Special Issue
PAK1 and Therapy Resistance in Melanoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

CAR NK Cell Therapy for the Treatment of Metastatic Melanoma: Potential & Prospects

Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
*
Author to whom correspondence should be addressed.
Cells 2023, 12(23), 2750; https://doi.org/10.3390/cells12232750
Submission received: 25 October 2023 / Revised: 22 November 2023 / Accepted: 26 November 2023 / Published: 30 November 2023
(This article belongs to the Special Issue Recent Advances in the Field of Metastatic Melanoma)

Abstract

:
Melanoma is among the most lethal forms of cancer, accounting for 80% of deaths despite comprising just 5% of skin cancer cases. Treatment options remain limited due to the genetic and epigenetic mechanisms associated with melanoma heterogeneity that underlie the rapid development of secondary drug resistance. For this reason, the development of novel treatments remains paramount to the improvement of patient outcomes. Although the advent of chimeric antigen receptor-expressing T (CAR-T) cell immunotherapies has led to many clinical successes for hematological malignancies, these treatments are limited in their utility by their immune-induced side effects and a high risk of systemic toxicities. CAR natural killer (CAR-NK) cell immunotherapies are a particularly promising alternative to CAR-T cell immunotherapies, as they offer a more favorable safety profile and have the capacity for fine-tuned cytotoxic activity. In this review, the discussion of the prospects and potential of CAR-NK cell immunotherapies touches upon the clinical contexts of melanoma, the immunobiology of NK cells, the immunosuppressive barriers preventing endogenous immune cells from eliminating tumors, and the structure and design of chimeric antigen receptors, then finishes with a series of proposed design innovations that could improve the efficacy CAR-NK cell immunotherapies in future studies.

1. Introduction

Melanoma, arising from the malignant transformation of melanocytes, is an aggressive and potentially lethal form of skin cancer. It is often caused by solar UV radiation, but there are myriad other environmental, immunological, and genetic risk factors that can also contribute to an individual’s predisposition towards melanoma [1,2]. For instance, heritable mutations such as in MC1R, CDK4, and CDKN2A, as well as genetic conditions like xeroderma pigmentosum (XP), have all been associated with an increased risk of melanoma development [3]. Melanoma is one of three major skin cancers, the other two being squamous cell carcinoma and basal cell carcinoma. Though melanoma accounts for only 5% of new skin cancer cases, it is ultimately responsible for approximately 80% of skin cancer deaths [4]. The incidence and mortality rates for malignant melanoma have steadily increased over the past few decades, particularly amongst older patients, whereas survival rates have remained relatively constant [4,5].
Resistance to a broad spectrum of therapies presents an ongoing challenge for the treatment of melanoma. Melanoma mutations occur at an exceptionally high rate, in turn driving the rapid development of treatment resistance [6]. Chemotherapeutics are often ineffective treatments for melanoma: standard chemotherapeutic agents such as dacarbazine elicit clinically positive responses in only 5–10% of patients [7,8]. If diagnosed early, melanoma could potentially be cured with surgery. However, roughly 30% of patients develop metastatic lesions in various organs following surgical ablation of primary tumors [9]. The propensity of melanoma for metastatic spread arises partly from the developmental origins of melanocytes, which are derived from the neural crest and consequently follow similar patterns of cellular migration to the brain and gastrointestinal tract [10]. Prognoses for patients with metastatic melanoma remain markedly poor. As the number of metastatic sites increases, the efficacy of treatments dwindles. Targeted therapies have improved patient outcomes; however, the effectiveness of these drugs is short-lived. For example, treatment with the BRAF inhibitors (BRAFi) vemurafenib and dabrafenib initially induced positive clinical responses in roughly 70% of BRAF-mutant patients [11], but the majority of responders subsequently developed BRAFi resistance in less than a year [12].
The 5-year survival rate for patients with metastatic melanoma is less than 5% [13] and highlights the limitations of current treatments. In recent years, the emergence of chimeric antigen receptor (CAR)-T cell therapies has garnered great success in the treatment of hematological cancers [14]. However, the application of CAR-T cell therapies is constrained by the associated risk of immunological systemic toxicities such as graft-versus-host disease (GvHD), immune effector cell-associated neurologic syndrome (ICANS), and cytokine release syndrome (CRS). GvHD and CRS tend to arise from immunoreactivity due to donor-host mismatching, whereas ICANS is an example of on-target, off-tumor toxicity. All of these can be fatal if left untreated [15,16,17,18,19]. More broadly, CAR-T cell therapies impose the risk of on-target, off-tumor toxicities that could cause potentially irreversible damage to healthy tissues that expressed the targeted antigen at low levels [20,21,22]. In addition, CAR-T cells are generally equipped with single-antigen specificity, so they are especially vulnerable to impaired efficacy in the event of antigen loss. Tumor heterogeneity is observed across many solid tumor types [23,24,25,26], including melanoma [27], which can further compromise the efficacy of CAR-T cell therapies if the targeted antigen is only expressed in a subset of tumor cells or becomes downregulated through acquired resistance [21,28,29].
CAR-NK cells are just beginning to gain traction as an alternative design to CAR-T cell therapies. Equipping natural killer (NK) cells with a CAR would retain the intrinsic cytotoxic capabilities of the transduced NK cell, while the phenotypic and mechanistic advantages of NK cells over T cells could allow CAR-NK cell therapies to soon become established as a viable treatment for melanoma. In contrast to T cells, adoptive cell therapies (ACTs) involving NK cells generally have a substantially lower risk of systemic toxicities. Furthermore, unlike T cells, NK cell activation does not require HLA matching for antigen recognition and its cytotoxicity mechanisms are regulated by multiple activating and inhibitory receptors [30,31,32]. This unique aspect of NK cell activity allows GvHD to be avoided in allogeneic NK cell therapies [31,33,34,35,36,37,38,39,40], which in turn enables allogeneic NK cells to be made readily available as an “off-the-shelf” treatment [41]. By extension, this implies that the acquisition of NK cells is generally much easier and more cost-efficient than T cells. Thus, CAR-NK cells have great potential as an immunotherapy treatment for melanoma.

2. Biology and Function of NK Cells in the Context of Melanoma

Accounting for 5–15% of the total cells in whole (peripheral) blood, NK cells are innate cytotoxic lymphocytes that can be found in blood, lung, liver, spleen, and bone marrow as well as in uterus, mucosal, and other tissues [42]. The two primary functions of NK cells are to maintain immune homeostasis and to eliminate stressed, infected, or potentially cancerous cells [43,44]. Generally, NK cells are characterized by the absence of CD3/TCR molecules and the presence of CD16 and CD56 (CD56+CD16+CD3-) phenotypes. Based on CD56 expression levels, NK cells can be further classified into CD56dim and CD56bright subsets [32]. CD56dim NK cells account for 85–95% of the NK cell population within peripheral blood and co-express the Fcγ receptor CD16 that enables their participation as effector cells in antibody-dependent cell cytotoxicity (ADCC) [45,46]. The potent cytotoxic activity of the mature CD56dim NK cells distinguishes them from the immature CD56bright NK cells. CD56bright NK cells account for only 5–15% of NK cells within peripheral blood, but, unlike CD56dim NK cells, CD56bright NK cells exhibit limited cytotoxic activity. Instead, CD56bright NK cells predominantly function as immunoregulators by secreting cytokines and chemokines such as IL-12, IL-15, IL-18, IFNγ, and TNF-α that then recruit other immune effectors against target cells [47,48]. Such NK cell activities are precisely regulated by signals transferred through its receptors [32,44]. Structurally, NK cell receptors are divided into the immunoglobulin-like receptor (IgSF) superfamily and the C-type lectin-like receptor (CLSF) superfamily [48]. Functionally, NK cell receptors are classified as either activating or inhibitory receptors; both functional types are included within either superfamily [49,50]. The IgSF includes natural cytotoxicity receptors (NCRs), human killer cell immunoglobulin-like receptors (KIRs), and leukocyte immunoglobulin-like receptors (LILRs) [50]. Killer cell lectin-like receptors (KLRs) are the primary constituents of the CLSF [50].

2.1. NK Cell Activating Receptors

NK cell activation signals are mediated by a diverse group of receptors. The main NK cell activating receptors (NKARs) include NCRs, NKG2D, NKG2C, DNAM-1 (CD226), CD16, and activating KIRs (aKIRs). NCRs play a large role in NK cell-mediated cytotoxicity against malignant cells [51]. The three NCRs expressed by NK cells are NKp46 (NCR1), NKp44 (NCR2), and NKp30 (NCR3) [52]. NKp46 and NKp30 constitutively express in resting-state and activated NK cells, while NKp44 expresses restrictedly in activated NK cells [53]. Ongoing challenges in experimental designs leave many NCR ligands undiscovered, but a notable exception to this trend was the discovery of the B7-H6 protein as an activating ligand for NKp30 [54]. NKG2D is a CLSF NKAR expressed by all NK cells [52] and participates extensively in activation signal transduction. NKG2D can recognize a broad range of activating ligands including MICA, MICB, and UL16-binding proteins 1–6 (ULBP1-6) [55]. In addition, NKG2D is unusual in that it can mediate signal transduction via multiple possible signaling pathways [53,55]. MICA is an NKG2D ligand that has been reported to be strongly expressed in melanoma cells [56]. CD226 is an IgSF coactivating receptor that acts synergistically with other NKARs to induce NK cell activation. CD155 and CD112 are the two known CD226 ligands. CD112 is expressed in roughly 26% of melanoma cases, whereas CD155 is expressed in most primary and metastatic melanoma cases [57]. Although surface expression of CD155 can induce NK cell activation, melanoma cells secrete a soluble form of CD155 that inhibits CD226-mediated NK cell cytotoxicity [58], which may explain the correlation between CD155 expression and negative melanoma prognostic markers such as metastatic emergence and treatment resistance [59,60]. CD16 is also a member of the IgSF and similarly functions as an NKAR. Finally, aKIRs are IgSF receptors, and depending on how many Ig-like domains they possess, they can be further classified as either KIR2DS or KIR3DS, where the “S” denotes short cytoplasmic tails. aKIR are a subtype of HLA-I dependent KIRs with shorter intracellular domains and primarily participate in NK cell activation through the conduction of activation signals [61]. Individual aKIRs may vary in the particular HLA molecule that they recognize: KIR2DS1, KIR2DS2, and KIR2DS4 recognize HLA-C, while KIR3DS1 recognizes HLA-B [62].
Although CD16 binding to its IgG ligand alone is sufficient to induce the release of lytic granules from NK cells [63,64,65], individual NKARs generally cannot induce NK cell cytotoxicity alone; rather, NK cell cytotoxicity is initiated by the collective, synergistic signaling from multiple co-activated receptors. Activation signaling is influenced by the contributions of several other structures, many of which are formed from dimerization. One such example is the CD94:NKG2C heterodimeric complex, and much like aKIRs:DAP12, its primary functions involve the transmission of activating signals [62].

2.2. NK Cell Inhibitory Receptors

The chief inhibitory NK receptors include Inhibitory KIRs (iKIRs) and Killer cell lectin-like receptor D1(KLRD1). iKIRs typically have long cytoplasmic tails and account for the majority of all KIRs. iKIRs are classified with the same conventions as aKIRs. Long cytoplasmic tails are indicated by a trailing “L” to leave finalized categories of KIR2DL and KIR3DL. Despite having a long cytoplasmic tail, KIR2DL4 is one of the exceptions to the cytoplasmic tail trend due to its dual ability to transduce activating signals as well via its arginine–tyrosine activation motif [66]. Like aKIRs, iKIRs are also MHC-I dependent and are similarly varied in their HLA molecule specificity. KIR2DL1, KIR2DL2, and KIR2DL3 recognize various HLA-B and HLA-C allotypes. KIR3DL1 recognizes Bw4 epitopes among HLA-B and some HLA-A allotypes, while KIR3DL2 recognizes certain HLA-A allotypes [52]. Prognoses for metastatic melanoma have been reported to shift according to KIR expression. Lower expression of the aKIR KIR2DS5 corresponds to faster rates of melanoma progression [67]. Likewise, higher expression of the inhibitory KIR2DL2 receptor corresponds to greater melanoma progression [67]. An earlier study [68] reported that HLA-C-bound KIR2DL4 appeared to have protective roles in melanoma, where the KIR2DL3:HLA-C complex appeared to induce NK cell activation; the alternate explanation of spurious allelic variation has yet to be ruled out [69,70]. KLRD1 (CD94) heterodimerized with NKG2A (CD94:NKG2A) functions as an inhibitory receptor that has specificity towards HLA-E. HLA-E is typically expressed at negligible levels in melanoma cells, but surface expression of HLA-E can become drastically upregulated in the presence of IFNγ, which is produced in high quantities by NK cells [52]. The presence of NK cells could therefore induce HLA-E upregulation, and in turn, enable tumor resistance towards NK cell cytotoxicity [52]. This phenomenon is further corroborated by the restoration of cytotoxic efficacy via NKG2A inhibition, although this may result in IFNγ upregulation [71,72,73]. LIRs are IgR members that participate predominantly in inhibitory signal transduction. Examples of LIRs include LIR1 (ILT2), which notably demonstrates broad specificity to HLA-A, HLA-B, and HLA-G [62]. Like NKARs, inhibitory receptors can also transduce signals synergistically with coreceptors such as TACTILE (CD96) and TIGIT, the latter of which recognizes CD112, CD113, and CD155 [74,75] (Figure 1).

2.3. NK Cell Cytotoxicity and Significance to Tumor Cell Clearance

NK cell cytotoxicity can be activated through two different mechanisms: by secreting lytic granules containing perforin and granzymes to degrade vital cell structures of the target cell, or by expressing TRAIL or FasL on their cell surface to trigger apoptotic cascades in target cells when either ligand interacts with the corresponding receptor [76]. Strikingly, NK cell cytotoxicity is neither dependent on antigen recognition nor restricted to one particular antigen [77]. Instead, NK cell activities are mediated by an intricate balance of germline-encoded activating or inhibitory receptors that can elicit NK cell responses as needed yet still ensure self-tolerance. Though the absence of self-identifying surface HLA expression is generally necessary for NK cells to be able to activate their cytotoxicity, this alone is not sufficient. Attenuated inhibitory signals must also be accompanied by activating signals from receptor recognition of specific ligands. Stimulatory ligands are only upregulated in stressed cells, so under normal circumstances, the inhibitory effects of HLA-I expression by healthy cells combined with the absence of activating ligands ensures that NK cells remain tolerant toward them [78]. Malignantly transformed cells, on the other hand, tend to downregulate the surface expression of HLA-I and upregulate stress-induced activating ligands, which initiate NK cell cytotoxicity against cancerous cells [79].
NK cells fulfill such a critical role in tumor cell clearance that the degree of NK cell infiltration in tumors can be used as a heuristic predictor of patient outcomes. Infiltrated NK cells in melanoma tumors were found to be positively correlated with patient survival rates [80,81]. The findings from a later study described a negative correlation between patient survival rates and the proportion of weakly cytotoxic CD56bright NK cells in circulation, which implicitly validated that the proportion of strongly cytotoxic CD56dim NK cells in circulation or infiltrated into tumors indeed correlated positively with patient survival [82]. The occurrence of both tumorigenesis and metastatic invasion is contingent upon tumor cell escape of NK cell-mediated destruction [32]. As such, the development of novel therapies that can disrupt immune escape mechanisms is paramount to patient survival.

3. NK Cell Immunosuppression within the Tumor Microenvironment

The tumor microenvironment (TME) is an evolving, complex, and dynamic entity that is characterized by metabolic and chemical conditions that are unfavorable for an immunological response to the growing tumor. Ongoing crosstalk between melanoma cells, immune cells, and stromal cells within the TME gives rise to a variety of functional shifts across several different cell types. Different cell types may not necessarily shift in the same direction, potentially shifting in activity to work in opposition to one another; however, phenotypic shifts tend to favor pro-tumoral states and often involve secretory activity. Several pro-tumoral cell types within the TME are capable of altering NK cell cytotoxicity, and the assortment of cell types that are present have already been summarized excellently in previous publications [83,84,85,86]. Here, we will focus on the three TME cell types that tend to exert the greatest influence over NK cell activity: regulatory T (Treg) cells, cancer-associated fibroblasts (CAFs), and tumor-associated macrophages (TAMs) (Figure 2).
The combined efforts of melanoma cells and pro-tumoral non-malignant cells give rise to four main NK cell immunosuppressive mechanisms by which tumor cells evade NK cell cytotoxicity: (1) disrupting receptor-ligand interactions responsible for NK cell activation; (2) amplifying receptor-ligand interactions that inhibit NK cell activity; (3) evading NK cell cytotoxicity with the assistance of non-malignant TME cells; and (4) escaping immune detection due to the immunosuppressive effect of inhospitable microenvironmental conditions (Figure 3).
First, melanoma cells can circumvent NK cell cytotoxicity by disrupting ligand-receptor interactions that drive NK cell activation. This may be accomplished through the melanoma-mediated downregulation of NKAR expression on NK cell surfaces, which effectively places a constraint on the strength of NK cell activation signals. For instance, melanoma cells highly express indoleamine-pyrrole 2,3-dioxygenase (IDO1) and prostaglandin E2 (PGE2), both of which have immunosuppressive activity. PGE2 can directly modulate NK cell receptor expression, whereas IDO1 catalyzes the production of L-kynurenine, which can then interact with NK cells and drive downregulation of major NKARs such as NKG2D, NKp30, and NKp44 [87,88,89,90,91,92,93]. Melanoma cells may also interfere with NK cell activation by downregulation of NKAR activating ligand expression or/and clearing away NKAR-activating ligands from melanoma cell surfaces via metalloprotease-mediated cleavage. In the presence of the BRAFi vemurafenib, BRAFV600E-mutant melanoma cells displayed strong downregulation of the NKAR ligands MICA, CD155, and B7-H6 [94], which may result in melanoma cell resistance towards NK cell cytotoxicity as a direct byproduct of acquired BRAFi resistance. The metalloproteases ADAM10, ADAM17, matrix metalloprotein (MMP)-25, and MMP-14 have been reported to be highly expressed in melanoma [95,96,97]. In addition, proteolytic shedding of the NKAR ligands MICA, ULBP2, and B7-H6 was often observed in melanoma samples [98]. Metastatic melanoma cells frequently shed NKAR-activating ligands such as the NKG2D ligands MICA and MICB [99]. The soluble ligands freed by metalloprotease-mediated cleavage can still interact with the binding sites of NKARs, thus inhibiting NKAR activation by obstructing surface-bound activating ligands from reaching their intended binding sites [100,101]. Higher concentrations of soluble ligands freed by proteolytic cleavage have been associated with advanced melanoma, which may be a factor underlying poor prognoses for individuals with late-stage or metastatic melanoma [97,102]. Inhibiting ligand cleavage sites attenuated ligand shedding and prevented metalloprotease recognition, thereby restoring NK cell cytotoxicity [97,103].
Second, melanoma cells can suppress NK cell cytotoxicity by promoting inhibitory NK cell receptor-ligand interactions. Tumor cells upregulate their surface expression of inhibitory ligands such as CD111 [104], CD112 [105,106], CD155 [106,107], CD200 [108], PD-L1 [109,110], HLA-E [111], and HLA-G [112,113]. Expression of non-classical HLA-G and HLA-E allotypes is observed across numerous cancers. HLA-E interacts with the inhibitory NK receptor complex CD94:NKG2A to suppress NK cell activity [114,115]. Likewise, non-classical HLA-G is recognized by LIR1 and similarly drives inhibitory signaling in NK cells upon binding [112]. Melanoma is also suspected to promote the upregulation of inhibitory receptors in NK cells [52]. The inhibitory receptors Tim-3 and TIGIT are expressed in both NK and T cells to regulate effector cell activity, and both were found to have immunosuppressive effects on NK cell activity [32,116]. A later study demonstrated that NK cell exhaustion was successfully reversed upon blockade of Tim-3 [117]. TIGIT recognizes CD111, CD112, CD155 (PVR), and PVRL2, where each interaction has immunosuppressive effects on NK cell activity [105,106]. TIGIT is inferred to directly suppress NK cell cytotoxicity based upon previous reports that TIGIT and CD155 have inhibitory effects on NK cell production of IFNγ [107]. Cancer cells, including melanoma cells, can also evade immune responses through exosome-mediated delivery of inhibitory receptor ligands to NK cells [118,119].
Third, non-malignant cells in the TME can assist with melanoma immune evasion. Depending on the cell type and environment, TME cells may intercept and inhibit NK cell activity directly via cell-cell interactions or by secreting immunosuppressive signaling molecules. For example, CAFs are fibroblasts within the TME that have been redirected to a pro-tumoral state resembling the phenotype of myofibroblasts [120] and secrete a variety of immunosuppressive cytokines, growth factors, and proteases that promote extracellular matrix (ECM) remodeling into a more favorable environment for tumor growth [121]. CAFs have a key role as front-line regulators of the anti-tumor activity from tumor-infiltrating immune cells [122]. Proteases secreted by CAFs further obstruct NK cell activity by participating in the proteolytic cleavage of NKAR ligands from melanoma cell surfaces [123]. Treg cells in the TME can similarly secrete immunosuppressive signaling molecules that can potentially impair NK cell cytotoxicity [124]. CD39/CD73 ectonucleotidase activity cleaves ATP to produce adenosine, which has the two-fold immunosuppressive effect of promoting CD4+ Treg cell maturation [125,126] and halting activity for most cytotoxic effector cells. For instance, adenosine directly inhibits NK cell infiltration and activation [127]. The elevated adenosine levels can also prevent macrophages from activation to target tumor cells [90]. IDO secreted by TAMs and MDSCs, as well as PGE2 produced from CAFs, promote the recruitment of Treg cells; in turn, this may further lead to intensified immunosuppression. TAMs were originally tumor-infiltrating immune cells that were driven from the phagocytic M1 phenotype to the angiogenic M2 phenotype through cell-cell interactions with melanoma (Figure 2) [90,128]. Furthermore, the acidic conditions of the TME promote the expression of the M2 phenotype [129,130]. TAM-secreted molecules may also disrupt treatment mechanisms. For example, TAMs secrete interleukin 1β (IL-1β) in response to the acidic conditions, which then intensely promote angiogenesis, tumor cell migration, tumor cell proliferation, and metastasis [129,130,131]. Analogously, CAFs secrete arginase into the TME, which appears to impair the arginine-dependent tumor cell detection mechanisms in cytotoxic T cells [132]. In cases of advanced disease progression, metastasis-associated macrophages (MAMs) may begin to express membrane-bound TGF-β that further compromises NK cell anti-tumor immunity [133].
Finally, immune evasion of NK cells may arise due to the inhospitable biochemical or environmental conditions of the TME [134]. Melanoma tumors do not use oxidative phosphorylation and are therefore heavily reliant on glycolysis for energy. Acidic byproducts from glycolysis and fermentation start to accumulate until the TME becomes distinctly acidic relative to its surroundings [135]. Moreover, uncontrolled tumor cell proliferation depletes the surrounding oxygen levels and results in hypoxic local conditions [136]. NK cell activation mechanisms, metabolism, and proliferative capacity are all dysregulated by the acidic and hypoxic conditions within the TME [82,106,108,113,114,132,137,138].
Inducing the expression of a CAR into NK cells helps redirect them towards malignant cells that express the target antigen and contributes activation signaling to the NK cell, thereby improving the chances that the NK cell can be cytotoxic against tumor cells despite the immunosuppressive effects of the TME. Both CAR-T cells and CAR-NK cells display enhanced cytotoxic activation capabilities alongside greater specificity against TAMs than their ordinary lymphocyte counterparts, and depending on the design, CAR-T and CAR-NK cells may be engineered with additional modifications that further increase their efficacy.

4. Car NK Cell Therapies to Treat Melanoma

4.1. Design & Production of CAR-NK Cells

CAR-NK cells generally express a CAR that mechanistically imitates a TCR activation signaling pathway; in most cases, CAR-NK cells are still created by transducing NK cells with the same CAR-encoding genes that would be used to create CAR-T cells. The CAR itself consists of four modular components: a single-chain variable fragment (scFv) domain, a hinge domain, a transmembrane domain, and one or multiple intracellular signaling domains. The extracellular scFv tumor antigen binding domain confers the specificity of CAR cells. This domain consists of a heavy (VH) and light (VL) variable chain fragment that is connected by a linker [139]. The location and abundance of the epitope, the order of the variable chain fragments, and the length of the linker must all be taken into consideration when designing the scFv domain [15]. The hinge domain links the scFv domain to the transmembrane domain and the hinge domain length influences the formation of immunological synapses as it facilitates access to tumor antigens. Membrane-distal tumor epitopes are more accessible with longer hinges, while membrane-proximal epitopes are better suited to shorter hinges [140]. The hinge domain of CAR moieties is often assembled using IgG-based hinges derived from IgG1, IgG2, and IgG4, often including the CH2 or CH3 IgG domains; other hinge domain constructs have made use of peptide sequences derived from CD28 or CD8α [140]. The transmembrane domain consists of a hydrophobic α-helix that anchors the scFv and hinge domains to the NK cell membrane [15,139]. CD4, CD28, and CD8α are the most commonly used transmembrane domains, but the choice of transmembrane domain may impact the degree of cell activation. The intracellular signaling domain governs the strength and nature of the activation signal. Differences in this intracellular domain distinguish between successive generations of CAR-transduced cells. First-generation CAR cells contained only the CD3ζ activation domain, while second- and third-generation CAR cells included one or two co-stimulatory molecules, respectively; of those molecules, CD28 and 4-1BB are frequently used [139,141]. Fourth-generation CAR cells are more broadly defined than the preceding generations but collectively seek to address the limitations of their predecessors.
Several fourth-generation CARs are constructed with a fragmented design. Universal CAR cells possess a fragmented and interchangeable antigen-specific extracellular domain that allows multiple cancer types or antigens to be targeted using the same combination of transmembrane and intracellular domains [15]. ON-switch CAR cells contain a fragmented intracellular domain that requires the presence of a particular small molecule for the functional CAR to be assembled; in other words, ON-switch CAR activation can be controlled through the administration of a drug [142]. Dual CARs comprise another subset of fourth-generation CARs; as the name implies, these constructs make use of two scFv domains simultaneously. AND-gated CAR cells require the expression of both target antigens on a given cell for signal propagation, which can enable non-specific tumor antigens to be targeted with better safety, provided that the other scFv domain is then targeted against a tumor-specific antigen. In contrast, OR-gated CAR cells activate when at least one of the two scFv domains has become bound to a target antigen to maximize tumor recognition rates. In both types of dual CARs, the scFv domains are able to share the same transmembrane and intracellular domains; in the case of OR-gated CARs, the dual scFv domains may simply involve the co-expression of two fully functional CAR constructs on the same cell [1]. Fourth-generation CARs also include a subset that can target the TME. In inhibitory CAR cells, the extracellular domains of inhibitory receptors are spliced together with the intracellular activating domains of CAR cells, allowing immunosuppressive signals to be redirected into activating signals [15]. Finally, T cells Redirected for Universal Cytokine Killing (TRUCK) are structurally similar to 3rd-generation CARs but carry an additional CAR-inducible transgenic “payload” that is to be delivered to the tumor site to attenuate immunosuppression by the TME [143]. Cytokine-mediated TME alteration is accompanied by a risk of systemic toxicities, so this approach should be used with caution, if at all [144].

4.2. Sources of NK Cells

NK cells suitable for CAR transduction can be obtained from several sources, but the process of isolating, purifying, and expanding primary NK cells can be difficult and inefficient. These limitations can be bypassed through NK cell lines, which are readily expanded in vitro [145]. The representative NK-92 cell line is an especially popular platform for many CAR-NK cell studies. Notably, CAR-NK92 cells were successfully used to treat renal cell carcinoma and high-risk rhabdomyosarcomas, and this may suggest that CAR-NK cells can elicit similar outcomes in additional cancer types [146,147]. One drawback of NK cell lines is their origin from malignant tissues, causing them to present an inherent risk of tumorigenicity [148]. As a precaution, NK cell lines must first be irradiated to prevent their persistence in vivo before they can be safely administered, but the shortened lifespan of irradiated cells may place a constraint on their clinical efficacy [149]. Transducing a drug-inducible suicide gene into CAR-NK cells may be an advantageous alternative to irradiation, as this would grant clinicians more agency in determining the duration of effect [150]. Umbilical cord blood is another viable source of NK cells. NK cells derived from cord blood possess an especially powerful capacity for expansion, giving them great clinical value in terms of scalability [151,152,153]. To minimize heterogeneity in the NK cell population between CD56dim and CD56bright phenotypes, cytokine co-stimulation ex vivo can induce the maturation into active CD56dim effector NK cells that have appreciable cytotoxicity and durable persistence [154]. Peripheral blood is another source of NK cells rich in mature NK cells; however, despite the strong cytolytic activity and a tendency to persist in blood for longer periods, these NK cells are usually difficult to expand in vitro [154]. Peripheral blood NK cells are also at varying stages of maturation, which can result in an unfavorably heterogeneous cell population following expansion [154]. Finally, induced pluripotent stem cells (iPSCs) are another potential source of NK cells. Extraction of iPSCs from cord or peripheral blood yields an immature population that can be transduced with a CAR construct of interest. Subsequent incubation in a cytokine cocktail containing various interleukins and growth factors can then induce their differentiation into CAR-equipped NK cells [155]. This offers the benefit of a more homogenous population of CAR-NK cells, but at the cost of reduced cytotoxicity on account of a relatively larger proportion of immature cells [154].

4.3. Pre-Clinical CAR-NK Studies with Meaningful Implications for Melanoma

Numerous pre-clinical trials have studied CAR-NK cell therapies as a potential treatment for several different solid malignancies. Most of the existing pre-clinical data pertains to glioblastoma, breast cancer, ovarian cancer, and pancreatic cancer [154], but applications of CAR-NK cell therapies have currently been extended to colorectal cancer [155], neuroblastoma [156], hepatocellular carcinoma [157], gastric cancer [158], small cell lung cancer [159], and lung adenocarcinoma [160]. Importantly, multiple pre-clinical studies assess antigen targets that could be clinically relevant to melanoma (Table 1).
Pre-melanosome protein (PMEL; PMEL17; glycoprotein 100, gp100) ordinarily plays a role in melanin synthesis and is highly specific to melanoma [181]. Because PMEL is broadly overexpressed (61–90%) in melanoma patients [175,181,182], independent of their stage of disease progression, PMEL is of especially great interest as a therapeutic target. CAR-NK92MI cells bearing TCR-like CARs were assembled by selecting the TCR-like antibody GPA7 against the PMEL/HLA-A2 complex and then fused to the intracellular domain of a CD3-ζ chain [175]. In the context of HLA-A2, the anti-PMEL CAR-NK cells were able to recognize melanoma cells and exhibited enhanced cytotoxic activity.
The disialoganglioside GD2 is overexpressed across multiple forms of cancer, such as glioblastoma [183], Ewing sarcoma [184], small cell lung cancer [185], melanoma [186], and other cancers, yet GD2 expression is generally restricted in healthy tissues [187]. Because of its strong cancer specificity, GD2 is a common target for several cancer immunotherapies [188]. GD2 is expressed by roughly 40% of melanoma tumors [182], and it follows that both CAR-T and CAR-NK cell therapies have been targeted against GD2 [172,173,189]. Mitwasi et al. generated anti-GD2 CAR-NK92 cells expressing CARs with either a conventional scFv domain or a novel IgG4-based antigen-binding domain and found that both CAR-NK92 variants demonstrated enhanced cytotoxic activity against melanoma cells both in vitro and in vivo; the IgG4-based CAR-NK92 variant exhibited a longer half-life than the conventional scFv-based variant but at the expense of reduced cytotoxic activity [172].
B7-H3 (CD276) is a transmembrane protein within the B7 glycoprotein family that is generally understood to be a negative regulator of effector cell activity [190,191]. Melanoma and several other solid tumors are known to express B7-H3, which may have an inhibitory effect on NK cell activity [192,193] and has grown to be a popular target for numerous immunotherapies, including both CAR-T cells and CAR-NK cells [161,194,195,196,197,198,199]. CAR-NK92 cells transduced with an anti-B7-H3 CAR exhibited strong cytotoxicity against melanoma cells independently of NKG2A expression or knockout, suggesting that anti-B7-H3 CAR-NK92 cells had displayed resistance towards NK inhibitory signaling mechanisms that are believed to underlie tumoral immune evasion [164]. Transforming growth factor (TGF)-β is another signaling molecule within the TME that has been associated with impaired cytotoxicity of both NK and CAR-NK cells, but this can be circumvented by co-transducing NK92-MI cells with both the anti-B7-H3 CAR and a TGF-β dominant negative receptor (DNR), thereby rescuing impaired cytotoxic activity [196].
Vascular endothelial growth factor receptor 2 (VEGFR-2) is an angiogenic receptor tyrosine kinase (RTK) that is upregulated in a subset of melanoma tumor specimens and is thought to play a role in tumor cell migration in metastasis [200]. In both T cells and YT NK cells, anti-VEGFR2 CAR transduction with an alternative design- replacing the scFv domain conventionally derived from monoclonal antibodies with an analogous fibronectin type III (Fn3) domain- successfully produced cytotoxicity against melanoma cells [201].

4.4. Potential Targets for CAR-NK Cell Therapies in Melanoma

Although melanoma has not yet been explicitly targeted in a CAR-NK clinical trial, there are a number of existing CAR-NK clinical trials that are currently recruiting or underway for various solid tumors. Several antigens from those studies may also apply to melanoma. A select few studies are discussed below, with more studies summarized in Table 2.
Mucin-1 (MUC1) is a transmembrane glycoprotein overexpressed in several solid tumors, such as breast cancer and melanoma. MUC1 could interact with ErbB2 and other RTKs involved in the PI3K/AKT signaling pathway, which in turn is associated with melanoma progression and acquired BRAF inhibitor resistance [201]. A recent study has shown that MUC1 participates in tumor cell migration as a driving force in metastasis in lung cancer, as well as melanoma [226]. A phase I clinical trial employed anti-PDL1/MUC1 dual CAR-NK cells against a range of solid tumors and found that this construct achieved a stable response in 9 out of 13 participants (~70%) [179]. By extension, this construct may be able to produce similar results if used to treat melanoma.
CLDN6 (cell adhesion protein Claudin-6), which plays a role in epithelial tight junction and cell-cell adhesion interactions, has recently been identified as a potential biomarker of melanoma and a myriad of other solid tumors [227]. A single-arm clinical trial (NCT05410717) is currently recruiting participants to investigate the therapeutic potential of anti-CLDN6 CAR-NK cells for the treatment of CLDN6-positive advanced solid tumors [166].
GD3 (ganglioside GD3) is in the same family of surface glycoproteins as GD2 and has been reported to be overexpressed in melanoma by at least 10-fold, relative to healthy melanocytes [228]. Upregulation of GD3 amplifies malignant invasion in melanoma cells by increasing adhesion signals and recruiting integrins [229,230]. Furthermore, GD3 was associated with malignant growth and invasion mediated by p130Cas and paxillin [229]. Previous pre-clinical studies have targeted GD3 as a melanoma antigen for both TCR-T and CAR-T cells [218,219], hinting that anti-GD3 CAR-NK cells could be used with similar success. Clinical implications for anti-GD3 CAR-NK cells may be particularly strong for melanoma brain metastases, which have been linked to GD3 upregulation [231], since CAR-NK cells offer an advantageous safety profile that can reduce the risk of on-target, off-tumor toxicities despite the tumor sites’ close proximity to ICANS-vulnerable brain regions.
αvβ3 (vitronectin receptor; integrin αvβ3) is expressed in melanoma, glioblastoma, pancreatic cancer, and other solid malignancies, where it plays a role in cell-cell and cell-ECM interactions [232]. Stimulation of integrin αvβ3 has led to activation of invasion by melanoma in vitro [233]. Alterations to patterns of glycosylation and sialylation of integrin αvβ3 have been reported in melanoma [234], hinting at its clinical significance as an antigen target. Anti-αvβ3 CAR-T cell therapies have been shown to effectively eliminate malignant cells in pre-clinical studies of glioblastoma, melanoma, and other solid malignancies [224,225]. The anti-αvβ3 CAR construct can be translated to CAR-NK cells, where it would be expected to confer advantages such as improved safety and cost-efficiency over CAR-T cells.
CD126 (interleukin 6 receptor; IL-6R) is predominantly expressed in B cells and epithelial cells. Increased apoptosis and decreased malignant growth have both been reported after antibody-mediated inhibition of IL-6 signaling [235], which provided the basis for a pre-clinical CAR-T cell study that targeted CD126. Mishra et al. developed anti-CD126 CAR-T cells that demonstrated broad efficacy against a panel of cancer types, including metastatic melanoma, both in vitro and in vivo [205]. In light of this success, anti-CD126 CAR-NK cells may be able to improve upon the clinical viability of this CAR-T cell construct. Other than this study, there is a scarcity of data on this antigen target, which leaves the door open to many possible routes for further research. Future CAR-NK studies may further elaborate upon the clinical value of CD126 as an antigen target.
Axl and MerTK are closely related RTKs that have both been associated with drug resistance and immunological resistance in melanoma [236,237,238,239] Antibody-mediated inhibition of Axl appeared to restore patient susceptibility to BRAF inhibitors [240]. Axl inhibition was also associated with melanoma cell motility and therefore required increased security to safeguard against the risk of an invasion [241,242]. MerTK expression is upregulated in BRAFi-resistant melanoma cells and TAMs [243,244]. Correspondingly, MerTK inhibition promoted the apoptosis of melanoma cells [245], along with suppression of tumor proliferation and growth [246].
CD271 (nerve growth factor receptor; NGFR) is upregulated in a subset of melanoma tumors and is implicated in resistance to both drug treatments and immune responses. After exposure to differentiation antigen-specific cytotoxic T cells, melanoma cells exhibited CD271 upregulation, which may predict resistance to anti-PD-1 therapies and immune cell exclusion from tumors [247]. CD271 is also associated with melanoma intra-tumoral heterogeneity [248] and metastatic migration of tumor cells [249]. Importantly, CD271 has been linked to immune escape in multiple reports. CD271 can be induced by IFNγ, which is produced in high volumes by NK cells, thus contributing to immune escape by driving the downregulation of melanoma antigens [250,251]. If future CAR-NK studies can be designed to secrete a synthetic peptide that can inhibit CD271, this may be able to restore endogenous effector cell recruitment for concerted tumor cell elimination alongside CAR-NK cells.

5. Perspectives and Future Prospects for CAR-NK Cell Therapies to Treat Melanoma

Current challenges faced by CAR-NK cell therapies include their limitation to surface antigens, neglect to structurally optimize the CAR for NK cell signal transduction, their susceptibility to immunosuppression by both the TME and malignant cells, their insufficient infiltration into tumors, and the general struggle with tumor clearance due to pro-tumoral signaling pathways that accelerate malignant growth. Future CAR-NK cell studies are likely to benefit from strengthening their cytotoxic activation as they seek out solutions to the challenges described (Figure 4).

5.1. Structural Optimization of the CAR to Enhance CAR-NK Cell Activation

Signal optimization is an important consideration for the optimization of CAR structures. Although TCR-T activation is generally more sensitive than CAR-T activation [252], this disparity does not necessarily carry over to CAR-NK and TCR-NK cells. NK cells are equipped with several pathways that can transduce activating signals, so it would be just as promising to try NK-specific signaling components. Presently, most CAR-NK cell constructs still use CARs designed to imitate TCR signaling [253] and may suffer from impaired activity as a result. CAR components usable in CAR-T cells may be incompatible with CAR-NK cell activity, and vice-versa. For instance, the effectiveness of 4-1BB as a costimulatory molecule in CAR-NK cells is contested, where conflicting findings hinted at an underlying incompatibility [254]; the unique significance of the adapter proteins DAP10 and DAP12 in NK cell signal transduction lends further credence to this idea [255]. CAR-NK cell therapies have been shown to perform best when equipped with components that have been tailored to NK cell signaling in studies [256,257,258]. A study on CAR-NK cell designs reported that CAR-NK cell cytotoxicity was highest for the CAR designed with an NKG2D transmembrane domain and substituting 2B4 and DAP10 as the chosen costimulatory molecules. All of these signaling components had been derived directly from NK-specific signaling components [259]. Likewise, a CAR-NK cell construct containing an NK-derived DNAM1 transmembrane domain outperformed an equivalent design bearing a T cell-specific CD28 transmembrane domain [157]. CAR-NK cell constructs possessing the NK-derived DAP12 signaling motif also outperformed equivalent constructs bearing the CD3ζ chain signaling motif [260,261], and this is a particularly noteworthy feat in that it established that TCR signaling mechanisms were not necessarily the best route to achieve NK cell activation.

5.2. Integration of TCR Structural Elements to Expand the Range of Targetable Antigens

Like antibodies, CAR-NK cells express a CAR that is similarly restricted to surface antigen targets. Finding ways to extend the range of potential antigen targets for CAR-NK cells has the potential to be pivotal for this prognosis. TCRs permit T cells to target intracellular antigens via MHC-I recognition [262], but despite this, engineered TCR-T cell therapies remain impractical due to their inherent vulnerability to TCR subunit mispairing between existing and transduced TCRs, which can lead to serious off-target toxicities [263,264]. NK cells do not naturally express TCRs, so the genes for a TCR can be transduced into NK cells without incurring the risks of subunit mispairing. Recently, TCR-NK cells expressing a transgenic TCR have been produced successfully [265]. A similar study reported that transgenic TCRs could be safely co-expressed alongside a CAR in CAR-T cells [211].
The TCR/CAR dual receptor layout would therefore permit CAR-NK cells to target the full breadth of melanoma antigens. However, this design can be consolidated into a mono-receptor TCR/CAR hybrid layout. The recent development of TCR-CARs involved the fusion of a TCR antigen recognition moiety to the intracellular CAR signaling moiety, successfully combining the strengths of both receptor types [266]. Future CAR-NK studies could further refine this design by incorporating NK-specific design elements in the intracellular CAR signaling domain.

5.3. Targeting Melanoma Cell Signaling Pathways

Melanoma cells frequently exhibit mutations to driver genes enriched in several protein kinase signaling pathways that result in the aberrant hyperactivation of these pathways. BRAF mutations are the most common type of mutation in melanoma, and this can affect multiple signaling pathways within the MAPK/ERK signaling network [267,268]. Protumoral signaling pathways in melanoma are generally governed by one or more RTKs that were overexpressed frequently in melanoma cells [269]. Inhibited tumor invasion, growth, and proliferation, as well as drug and immunological resistance, have all been reported following RTK inhibition [270].
Inhibition of the signaling pathways by inhibitors used in combination with CAR-NK cell targeting specific tyrosine kinase receptors could inhibit melanoma invasion and proliferation or kill the tumor cells directly. Alternatively, CAR-NK cells could be similarly designed to secrete an inducible peptide inhibitor for a myriad of melanoma cell receptors, analogous to the TRUCK concept in CAR-T cells [173,189]. For instance, the transmembrane protein LRIG1 is expressed by NK cells [271], and the soluble ectodomain released by proteolytic cleavage (sLRIG1) has been shown to efficiently impede malignant growth in models of glioma by inhibiting several RTKs, chiefly within EGFR receptor family [179,272]. Furthermore, sLRIG1-mediated EGFR inhibition was demonstrated to inhibit melanoma growth and proliferation both in vitro [273], hinting at a favorable likelihood of similar effects in vivo. sLRIG1 is already available commercially as a reagent, so it can be inferred that DNA and peptide sequences are both already known. If sLRIG1 secretion were to be coupled to antigen recognition by the scFv of a CAR-NK cell then CAR-NK cells could potentially be used as both an effector cell and as a vehicle for localized drug delivery simultaneously. Future studies may be able to rely upon methods such as phage display [274] to elicit similarly viable peptides for use as receptor inhibitors.

5.4. Targeting Cell Motility to Enhance Immune Infiltration and Impair Malignant Invasion

Chemotaxis influences the activity of both effector and target cells; the motility of melanoma cells is a major driving force in metastatic migration, while NK cell motility can heuristically measure the degree of immune cell infiltration within a tumor. It follows that NK cell cytotoxicity, and by extension their clinical efficacy, can be enhanced in the presence of chemotactic cytokines and chemokines. NK cells must be able to migrate to metastatic sites if they are to eliminate metastases effectively. Chemokine receptors on NK cells such as CXCR3 enable robust NK cell chemoattraction [275]; conversely, CXCR3-deficient NK cells failed to migrate to metastatic sites [276], suggesting that increasing CXCR3 in CAR-NK cells may promote CAR-NK cells’ tumor site infiltration and enhance the CAR-NK mediated tumor clearance. Chemotactic motility can also be impacted by the expression of autophagy genes. By blocking the autophagy gene Beclin1 (BECN1) in a model of melanoma, inhibition of CCL5 was alleviated and led to inhibited growth of melanoma cells concomitantly to increased NK cell infiltration into the tumor [277].

5.5. Targeting Non-Malignant Cell Types in the TME

The assortment of cell types within the TME do not act as isolated contributors to immune escape; rather, they form a complex cell signaling network involving many different cell-cell interactions. In lieu of targeting a novel candidate melanoma surface antigen, CAR-NK cells that are alternatively redirected against antigens expressed by non-malignant TME cell types would most likely reduce the population of cells secreting immunosuppressive signals and improve the odds of preserving NK cell cytotoxicity.
CAFs may promote drug resistance and metastatic invasion in melanoma, so their destruction would likely have great clinical value. Co-culture experiments indicate that CAFs are drivers of tumor cell invasion [278]. The relationship between CAFs and RTKs expressed by melanoma cells, which mediate signaling that leads to increased MAPK activation, has also been implicated in BRAF inhibitor resistance in BRAF-mutant patients [279]. BRAF inhibitors can reprogram CAFs to drive matrix remodeling in melanoma [280], and it follows that CAFs may influence melanoma cell responsiveness to combinatorial therapy involving both BRAF and MEK inhibitors [281]. Inhibition of CAFs by β-catechin was associated with pronounced decreases in tumoral vascularization, which hints at the importance of CAFs in metastatic development and tumor progression [282]. CAFs have also been identified as mediators of immune escape as demonstrated by the impairment of CD8+ cytotoxic T cell activity via arginase-mediated interference with tumor cell recognition [132]. CAFs have also been reported to mediate inhibitory phenotypic shifts in NK cells [283]. Fortunately, CAFs are identifiable by a number of marker genes, including FAP [284], Col11A1 [285], and Acta2 [286], thus they could be specifically targeted by CAR-NK cell therapies in the coming years.
TAMs are another significant contributor to poor patient outcomes in melanoma. Crosstalk between CAFs and TAMs has been noted to exacerbate tumorigenesis and immune escape [287], so if CAFs are not a viable target for CAR-NK cells, then it may be worthwhile to target TAMs instead. TAMs are macrophages that have been functionally shifted to the pro-inflammatory M2 phenotype by melanoma cells [90]. M2 macrophages are known to inhibit the activity of anti-tumoral M1 macrophages [288]. In addition, TAMs express the β3-adrenoreceptor and secrete adrenomedullin, which are both associated with increased vascularization and malignant invasion in melanoma [90,289]. Therefore, the elimination of TAMs would be expected to improve immunological responsiveness against tumor cells. To date, there is at least one example of a CAR-NK cell therapy directed against TAMs, in which anti-CSF1R CAR-NK92MI and CAR-T cells were developed and exhibited strong anti-TAM activity [290]. Future CAR-NK cell studies may be able to elicit comparable successes by targeting other known TAM markers such as CCL5, CD163, or AHR [291].
Finally, Treg cells are secretory T cells that produce a plethora of immunosuppressive signaling molecules that protect melanoma tumors from immune destruction [292,293,294,295]. Importantly, Treg secretion of perforin and granzyme-B are directly associated with NK cell inhibition [296]. A decrease in the Treg cell population would correspond to a lessened immunosuppressive environment for CAR-NK cells. Recently, a CAR-NK cell study targeting CD25 was published to target Treg cells that were newly reprogrammed to a pro-tumoral state [297]. Other markers such as galectin-9 [298] and PD-L1/L2 [299,300] may also be potential targets for CAR-NK cell therapies that are targeted against Treg cells.

5.6. Incoporation with Deletion of Inhibitory Receptor Using Innovated Methods

NK cell inhibitory receptors are potential immune checkpoint therapeutic targets [32]. Blocking these inhibitory receptors could enhance the capability of NK cells to killing the tumor cells [115,200]. CRISPR/Cas9 technology has enabled site-specific inhibitory receptor gene deletion. Recent study has demonstrated that combining adeno-associated viral (AAV) system with CRISPR/Cas9 technology could successfully insert the gene into NK cells with site-specifically [301]. This innovative method may be the most promising approach to develop next generation CAR-NK cells.

5.7. Clinical Consideration of Pharmaceutical Impacts on CAR-NK Cell Efficacy

As a final consideration, it is imperative that future CAR-NK cell therapies be optimized to operate in tandem with drug therapies. NK cells are directly responsible for the clinical successes of BRAF inhibitors [302] and acquired resistance to BRAF inhibitors has been associated with increased melanoma cell susceptibility to NK cell lysis [303]. If subsequent studies can clarify the potential risk of BRAF inhibitor cytotoxicity towards CAR-NK cells, the clinical versatility of cell immunotherapies may be confirmed. BRAF inhibitors may begin to be administered as an adjuvant to CAR-NK cell therapies, or perhaps CAR-NK cells may become a follow-up therapy to BRAF inhibitors by capitalizing on the phenotypic shifts associated with acquired drug resistance [304]. Future studies that explore the dynamics between NK cells and other drug therapies would shed light on the compatibility of CAR-NK cells with other adjuvant therapies. Promising clinical implications may be revealed by these studies, as well as potentially unfavorable interactions that clinicians should consider when formulating treatment plans for melanoma patients.

6. Conclusions

The rapid development of drug resistance contributes significantly to the ongoing clinical challenges of melanoma treatment. The persistently high mortality rate of melanoma patients, especially those with metastatic melanoma, is indicative of an ongoing, unmet need for a sufficiently efficacious treatment. CAR-T cell therapies have marked a significant milestone for improving patient outcomes for hematological malignancies, but the accompanying risks and limitations of CAR-T cell therapies suggest that CAR-NK cells may be a more suitable alternative immunotherapy. NK cells are mechanically distinct from T cells, which enables them to continue to target tumor cells even in cases of antigen loss, but this comes at the cost of increased sensitivity to inhibitory signals secreted within the TME. As CAR-NK cell therapies continue to grow and improve, the impact of these limitations will be lessened with time. There is a favorable degree of flexibility for the myriad of therapeutic strategies that may be useful for future studies on CAR-NK cells. Further research will provide greater insights into the clinical utility of CAR-NK cells for the treatment of melanoma.

Funding

This work was funded in part by the Intramural Research Program at the National Institutes of Health and the Staff Scientist/Staff Clinician Research Award to YY from the National Cancer Institute Center for Cancer Research.

Acknowledgments

The figures were created with BioRender 23. We apologize to all authors whose work could not be cited due to space limitations.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Conforti, C.; Zalaudek, I. Epidemiology and Risk Factors of Melanoma: A Review. Dermatol. Pract. Concept. 2021, 11, 2021161S. [Google Scholar] [CrossRef]
  2. Strashilov, S.; Yordanov, A. Aetiology and Pathogenesis of Cutaneous Melanoma: Current Concepts and Advances. Int. J. Mol. Sci. 2021, 22, 6395. [Google Scholar] [CrossRef]
  3. Sundararajan, S.; Thida, A.M.; Yadlapati, S.; Koya, S. Metastatic Melanoma; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  4. National Cancer Institute. SEER Cancer Stat Facts: Melanoma of the Skin. National Cancer Institute. In National Cancer Institute; 2022. Available online: https://seer.cancer.gov/statfacts/html/melan.html (accessed on 26 December 2022).
  5. Saginala, K.; Barsouk, A.; Aluru, J.S.; Rawla, P.; Barsouk, A. Epidemiology of Melanoma. Med. Sci. 2021, 9, 63. [Google Scholar] [CrossRef]
  6. Davis, E.J.; Johnson, D.B.; Sosman, J.A.; Chandra, S. Melanoma: What do all the mutations mean? Cancer 2018, 124, 3490–3499. [Google Scholar] [CrossRef] [PubMed]
  7. Chapman, P.B.; Hauschild, A.; Robert, C.; Haanen, J.B.; Ascierto, P.; Larkin, J.; Dummer, R.; Garbe, C.; Testori, A.; Maio, M.; et al. Improved Survival with Vemurafenib in Melanoma with BRAF V600E Mutation. N. Engl. J. Med. 2011, 364, 2507–2516. [Google Scholar] [CrossRef] [PubMed]
  8. Chapman, P.B.; Einhorn, L.H.; Meyers, M.L.; Saxman, S.; Destro, A.N.; Panageas, K.S.; Begg, C.B.; Agarwala, S.S.; Schuchter, L.M.; Ernstoff, M.S.; et al. Phase III Multicenter Randomized Trial of the Dartmouth Regimen versus Dacarbazine in Patients with Metastatic Melanoma. J. Clin. Oncol. 1999, 17, 2745–2751. [Google Scholar] [CrossRef] [PubMed]
  9. Sandru, A.; Voinea, S.; Panaitescu, E.; Blidaru, A. Survival rates of patients with metastatic malignant melanoma. J. Med. Life 2014, 7, 572–576. [Google Scholar] [PubMed]
  10. Wessely, A.; Steeb, T.; Berking, C.; Heppt, M.V. How Neural Crest Transcription Factors Contribute to Melanoma Heterogeneity, Cellular Plasticity, and Treatment Resistance. Int. J. Mol. Sci. 2021, 22, 5761. [Google Scholar] [CrossRef]
  11. Priantti, J.N.; Vilbert, M.; Madeira, T.; Moraes, F.C.A.; Hein, E.C.K.; Saeed, A.; Cavalcante, L. Efficacy and Safety of Rechallenge with BRAF/MEK Inhibitors in Advanced Melanoma Patients: A Systematic Review and Meta-Analysis. Cancers 2023, 15, 3754. [Google Scholar] [CrossRef]
  12. Davis, L.E.; Shalin, S.C.; Tackett, A.J. Current state of melanoma diagnosis and treatment. Cancer Biol. Ther. 2019, 20, 1366–1379. [Google Scholar] [CrossRef]
  13. Kalal, B.S.; Upadhya, D.; Pai, V.R. Chemotherapy resistance mechanisms in advanced skin cancer. Oncol. Rev. 2017, 11, 19–25. [Google Scholar] [CrossRef] [PubMed]
  14. June, C.H.; O’Connor, R.S.; Kawalekar, O.U.; Ghassemi, S.; Milone, M.C. CAR T cell immunotherapy for human cancer. Science 2018, 359, 1361–1365. [Google Scholar] [CrossRef] [PubMed]
  15. Pfefferle, A.; Huntington, N.D. You Have Got a Fast CAR: Chimeric Antigen Receptor NK Cells in Cancer Therapy. Cancers 2020, 12, 706. [Google Scholar] [CrossRef]
  16. Lee, D.W.; Santomasso, B.D.; Locke, F.L.; Ghobadi, A.; Turtle, C.J.; Brudno, J.N.; Maus, M.V.; Park, J.H.; Mead, E.; Pavletic, S.; et al. ASTCT Consensus Grading for Cytokine Release Syndrome and Neurologic Toxicity Associated with Immune Effector Cells. Biol. Blood Marrow Transplant. 2019, 25, 625–638. [Google Scholar] [CrossRef] [PubMed]
  17. Schuster, S.J.; Bishop, M.R.; Tam, C.S.; Waller, E.K.; Borchmann, P.; McGuirk, J.P.; Jäger, U.; Jaglowski, S.; Andreadis, C.; Westin, J.R.; et al. Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B-Cell Lymphoma. N. Engl. J. Med. 2019, 380, 45–56. [Google Scholar] [CrossRef] [PubMed]
  18. Neelapu, S.S.; Locke, F.L.; Bartlett, N.L.; Lekakis, L.J.; Miklos, D.B.; Jacobson, C.A.; Braunschweig, I.; Oluwole, O.O.; Siddiqi, T.; Lin, Y.; et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. N. Engl. J. Med. 2017, 377, 2531–2544. [Google Scholar] [CrossRef]
  19. Liu, P.; Liu, M.; Lyu, C.; Lu, W.; Cui, R.; Wang, J.; Li, Q.; Mou, N.; Deng, Q.; Yang, D. Acute Graft-Versus-Host Disease after Humanized Anti-CD19-CAR T Therapy in Relapsed B-ALL Patients after Allogeneic Hematopoietic Stem Cell Transplant. Front. Oncol. 2020, 10, 799206. [Google Scholar] [CrossRef] [PubMed]
  20. Zhang, Y.; Li, Y.; Cao, W.; Wang, F.; Xie, X.; Li, Y.; Wang, X.; Guo, R.; Jiang, Z.; Guo, R. Single-Cell Analysis of Target Antigens of CAR-T Reveals a Potential Landscape of “On-Target, Off-Tumor Toxicity”. Front. Immunol. 2021, 12, 799206. [Google Scholar] [CrossRef]
  21. Morgan, R.A.; Yang, J.C.; Kitano, M.; E Dudley, M.; Laurencot, C.M.; Rosenberg, S.A. Case Report of a Serious Adverse Event Following the Administration of T Cells Transduced with a Chimeric Antigen Receptor Recognizing ERBB2. Mol. Ther. 2010, 18, 843–851. [Google Scholar] [CrossRef]
  22. Castellarin, M.; Sands, C.; Da, T.; Scholler, J.; Graham, K.; Buza, E.; Fraietta, J.A.; Zhao, Y.; June, C.H. A rational mouse model to detect on-target, off-tumor CAR T cell toxicity. JCI Insight 2020, 5, e136012. [Google Scholar] [CrossRef]
  23. Laskowski, T.J.; Biederstädt, A.; Rezvani, K. Natural killer cells in antitumour adoptive cell immunotherapy. Nat. Rev. Cancer 2022, 22, 557–575. [Google Scholar] [CrossRef]
  24. Dagogo-Jack, I.; Shaw, A.T. Tumour heterogeneity and resistance to cancer therapies. Nat. Rev. Clin. Oncol. 2018, 15, 81–94. [Google Scholar] [CrossRef] [PubMed]
  25. Yu, T.; Gao, X.; Zheng, Z.; Zhao, X.; Zhang, S.; Li, C.; Liu, G. Intratumor Heterogeneity as a Prognostic Factor in Solid Tumors: A Systematic Review and Meta-Analysis. Front. Oncol. 2021, 11, 744064. [Google Scholar] [CrossRef] [PubMed]
  26. 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] [PubMed]
  27. Grzywa, T.M.; Paskal, W.; Włodarski, P.K. Intratumor and Intertumor Heterogeneity in Melanoma. Transl. Oncol. 2017, 10, 956–975. [Google Scholar] [CrossRef] [PubMed]
  28. O’rourke, D.M.; Nasrallah, M.P.; Desai, A.; Melenhorst, J.J.; Mansfield, K.; Morrissette, J.J.D.; Martinez-Lage, M.; Brem, S.; Maloney, E.; Shen, A.; et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci. Transl. Med. 2017, 9, eaaa0984. [Google Scholar] [CrossRef] [PubMed]
  29. Ahmed, N.; Brawley, V.S.; Hegde, M.; Robertson, C.; Ghazi, A.; Gerken, C.; Liu, E.; Dakhova, O.; Ashoori, A.; Corder, A.; et al. Human Epidermal Growth Factor Receptor 2 (HER2)—Specific Chimeric Antigen Receptor—Modified T Cells for the Immunotherapy of HER2-Positive Sarcoma. J. Clin. Oncol. 2015, 33, 1688–1696. [Google Scholar] [CrossRef]
  30. Pitcovski, J.; Shahar, E.; Aizenshtein, E.; Gorodetsky, R. Melanoma antigens and related immunological markers. Crit. Rev. Oncol. Hematol. 2017, 115, 36–49. [Google Scholar] [CrossRef]
  31. Shi, J.; Tricot, G.; Szmania, S.; Rosen, N.; Garg, T.K.; Malaviarachchi, P.A.; Moreno, A.; DuPont, B.; Hsu, K.C.; Baxter-Lowe, L.A.; et al. Infusion of haplo-identical killer immunoglobulin-like receptor ligand mismatched NK cells for relapsed myeloma in the setting of autologous stem cell transplantation. Br. J. Haematol. 2008, 143, 641–653. [Google Scholar] [CrossRef]
  32. Yu, Y. The Function of NK Cells in Tumor Metastasis and NK Cell-Based Immunotherapy. Cancers 2023, 15, 2323. [Google Scholar] [CrossRef]
  33. Arai, S.; Meagher, R.; Swearingen, M.; Myint, H.; Rich, E.; Martinson, J.; Klingemann, H. Infusion of the allogeneic cell line NK-92 in patients with advanced renal cell cancer or melanoma: A phase I trial. Cytotherapy 2008, 10, 625–632. [Google Scholar] [CrossRef] [PubMed]
  34. Li, H.K.; Hsiao, C.W.; Yang, S.H.; Yang, H.P.; Wu, T.S.; Lee, C.Y.; Lin, Y.L.; Pan, J.; Cheng, Z.F.; Lai, Y.D.; et al. A Novel off-the-Shelf Trastuzumab-Armed NK Cell Therapy (ACE1702) Using Antibody-Cell-Conjugation Technology. Cancers 2021, 13, 2724. [Google Scholar] [CrossRef] [PubMed]
  35. Williams, B.A.; Law, A.D.; Routy, B.; Denhollander, N.; Gupta, V.; Wang, X.-H.; Chaboureau, A.; Viswanathan, S.; Keating, A. A phase I trial of NK-92 cells for refractory hematological malignancies relapsing after autologous hematopoietic cell transplantation shows safety and evidence of efficacy. Oncotarget 2017, 8, 89256–89268. [Google Scholar] [CrossRef] [PubMed]
  36. Rujkijyanont, P.; Chan, W.K.; Eldridge, P.W.; Lockey, T.; Holladay, M.; Rooney, B.; Davidoff, A.M.; Leung, W.; Vong, Q. Ex Vivo Activation of CD56+ Immune Cells That Eradicate Neuroblastoma. Cancer Res. 2013, 73, 2608–2618. [Google Scholar] [CrossRef] [PubMed]
  37. Otegbeye, F.; Cooper, B.; Caimi, P.; Zamborsky, K.; Reese-Koc, J.; Hillian, A.; Hernandez-Collazo, Y.; Lee, G.; Boughan, K.; Tomlinson, B.; et al. A Phase I Study to Determine the Maximum Tolerated Dose of Ex Vivo Expanded Natural Killer Cells Derived from Unrelated, HLA-Disparate Adult Donors. Transplant. Cell Ther. 2022, 28, 250.e1–250.e8. [Google Scholar] [CrossRef] [PubMed]
  38. Mansour, A.G.; Teng, K.-Y.; Li, Z.; Zhu, Z.; Chen, H.; Tian, L.; Ali, A.; Zhang, J.; Lu, T.; Ma, S.; et al. Off-the-shelf CAR–engineered natural killer cells targeting FLT3 enhance killing of acute myeloid leukemia. Blood Adv. 2023, 7, 6225–6239. [Google Scholar] [CrossRef] [PubMed]
  39. Caruso, S.; De Angelis, B.; Del Bufalo, F.; Ciccone, R.; Donsante, S.; Volpe, G.; Manni, S.; Guercio, M.; Pezzella, M.; Iaffaldano, L.; et al. Safe and effective off-the-shelf immunotherapy based on CAR.CD123-NK cells for the treatment of acute myeloid leukaemia. J. Hematol. Oncol. 2022, 15, 163. [Google Scholar] [CrossRef]
  40. Portillo, A.L.; Hogg, R.; Poznanski, S.M.; Rojas, E.A.; Cashell, N.J.; Hammill, J.A.; Chew, M.V.; Shenouda, M.M.; Ritchie, T.M.; Cao, Q.T.; et al. Expanded human NK cells armed with CAR uncouple potent anti-tumor activity from off-tumor toxicity against solid tumors. iScience 2021, 24, 102619. [Google Scholar] [CrossRef]
  41. Ruggeri, L.; Capanni, M.; Urbani, E.; Perruccio, K.; Shlomchik, W.D.; Tosti, A.; Posati, S.; Rogaia, D.; Frassoni, F.; Aversa, F.; et al. Effectiveness of Donor Natural Killer Cell Alloreactivity in Mismatched Hematopoietic Transplants. Science 2002, 295, 2097–2100. [Google Scholar] [CrossRef]
  42. Daher, M.; Garcia, L.M.; Li, Y.; Rezvani, K. CAR-NK cells: The next wave of cellular therapy for cancer. Clin. Transl. Immunol. 2021, 10, e1274. [Google Scholar] [CrossRef]
  43. Sojka, D.K.; Plougastel-Douglas, B.; Yang, L.; A Pak-Wittel, M.; Artyomov, M.N.; Ivanova, Y.; Zhong, C.; Chase, J.M.; Rothman, P.B.; Yu, J.; et al. Tissue-resident natural killer (NK) cells are cell lineages distinct from thymic and conventional splenic NK cells. Elife 2014, 3, e01659. [Google Scholar] [CrossRef] [PubMed]
  44. Marofi, F.; Rahman, H.S.; Thangavelu, L.; Dorofeev, A.; Bayas-Morejón, F.; Shirafkan, N.; Shomali, N.; Chartrand, M.S.; Jarahian, M.; Vahedi, G.; et al. Renaissance of armored immune effector cells, CAR-NK cells, brings the higher hope for successful cancer therapy. Stem Cell Res. Ther. 2021, 12, 200. [Google Scholar] [CrossRef] [PubMed]
  45. Morice, W.G. The Immunophenotypic Attributes of NK Cells and NK-Cell Lineage Lymphoproliferative Disorders. Am. J. Clin. Pathol. 2007, 127, 881–886. [Google Scholar] [CrossRef] [PubMed]
  46. Xie, G.; Dong, H.; Liang, Y.; Ham, J.D.; Rizwan, R.; Chen, J. CAR-NK cells: A promising cellular immunotherapy for cancer. EBioMedicine 2020, 59, 102975. [Google Scholar] [CrossRef] [PubMed]
  47. Lorenzo-Herrero, S.; López-Soto, A.; Sordo-Bahamonde, C.; Gonzalez-Rodriguez, A.P.; Vitale, M.; Gonzalez, S. NK Cell-Based Immunotherapy in Cancer Metastasis. Cancers 2018, 11, 29. [Google Scholar] [CrossRef] [PubMed]
  48. Poli, A.; Michel, T.; Thérésine, M.; Andrès, E.; Hentges, F.; Zimmer, J. CD56bright natural killer (NK) cells: An important NK cell subset. Immunology 2009, 126, 458–465. [Google Scholar] [CrossRef] [PubMed]
  49. Radaev, S.; Sun, P.D. Structure and Function of Natural Killer Cell Surface Receptors. Annu. Rev. Biophys. Biomol. Struct. 2003, 32, 93–114. [Google Scholar] [CrossRef]
  50. Rangarajan, S.; Mariuzza, R.A. Natural Killer Cell Receptors. In Structural Biology in Immunology; Elsevier: Amsterdam, The Netherlands, 2018; pp. 101–125. [Google Scholar] [CrossRef]
  51. Kruse, P.H.; Matta, J.; Ugolini, S.; Vivier, E. Natural cytotoxicity receptors and their ligands. Immunol. Cell Biol. 2014, 92, 221–229. [Google Scholar] [CrossRef]
  52. Lee, H.; Da Silva, I.P.; Palendira, U.; Scolyer, R.A.; Long, G.V.; Wilmott, J.S. Targeting NK Cells to Enhance Melanoma Response to Immunotherapies. Cancers 2021, 13, 1363. [Google Scholar] [CrossRef]
  53. Paul, S.; Lal, G. The Molecular Mechanism of Natural Killer Cells Function and Its Importance in Cancer Immunotherapy. Front. Immunol. 2017, 8, 1124. [Google Scholar] [CrossRef]
  54. Brandt, C.S.; Baratin, M.; Yi, E.C.; Kennedy, J.; Gao, Z.; Fox, B.; Haldeman, B.; Ostrander, C.D.; Kaifu, T.; Chabannon, C.; et al. The B7 family member B7-H6 is a tumor cell ligand for the activating natural killer cell receptor NKp30 in humans. J. Exp. Med. 2009, 206, 1495–1503. [Google Scholar] [CrossRef]
  55. Duan, S.; Guo, W.; Xu, Z.; He, Y.; Liang, C.; Mo, Y.; Wang, Y.; Xiong, F.; Guo, C.; Li, Y.; et al. Natural killer group 2D receptor and its ligands in cancer immune escape. Mol. Cancer 2019, 18, 29. [Google Scholar] [CrossRef] [PubMed]
  56. Cluxton, C.D.; Spillane, C.; O’Toole, S.A.; Sheils, O.; Gardiner, C.M.; O’Leary, J.J. Suppression of Natural Killer cell NKG2D and CD226 anti-tumour cascades by platelet cloaked cancer cells: Implications for the metastatic cascade. PLoS ONE 2019, 14, e0211538. [Google Scholar] [CrossRef] [PubMed]
  57. Tarazona, R.; Duran, E.; Solana, R. Natural Killer Cell Recognition of Melanoma: New Clues for a More Effective Immunotherapy. Front. Immunol. 2016, 6, 649. [Google Scholar] [CrossRef]
  58. Okumura, G.; Iguchi-Manaka, A.; Murata, R.; Yamashita-Kanemaru, Y.; Shibuya, A.; Shibuya, K. Tumor-derived soluble CD155 inhibits DNAM-1–mediated antitumor activity of natural killer cells. J. Exp. Med. 2020, 217, e20191290. [Google Scholar] [CrossRef]
  59. Bevelacqua, V.; Bevelacqua, Y.; Candido, S.; Skarmoutsou, E.; Amoroso, A.; Guarneri, C.; Strazzanti, A.; Gangemi, P.; Mazzarino, M.C.; D’amico, F.; et al. Nectin like-5 overexpression correlates with the malignant phenotype in cutaneous melanoma. Oncotarget 2012, 3, 882–892. [Google Scholar] [CrossRef]
  60. Lepletier, A.; Madore, J.; O’Donnell, J.S.; Johnston, R.L.; Li, X.Y.; McDonald, E.; Ahern, E.; Kuchel, A.; Eastgate, M.; Pearson, S.A.; et al. Tumor CD155 Expression Is Associated with Resistance to Anti-PD1 Immunotherapy in Metastatic Melanoma. Clin. Cancer Res. 2020, 26, 3671–3681. [Google Scholar] [CrossRef]
  61. Ivarsson, M.A.; Michaã«Lsson, J.; Fauriat, C. Activating killer cell Ig-like receptors in health and disease. Front. Immunol. 2014, 5, 184. [Google Scholar] [CrossRef]
  62. Chen, Y.; Lu, D.; Churov, A.; Fu, R. Research Progress on NK Cell Receptors and Their Signaling Pathways. Mediat. Inflamm. 2020, 2020, 6437057. [Google Scholar] [CrossRef]
  63. Tsukerman, P.; Stern-Ginossar, N.; Yamin, R.; Ophir, Y.; Stanietsky, A.M.N.; Mandelboim, O. Expansion of CD16 positive and negative human NK cells in response to tumor stimulation. Eur. J. Immunol. 2014, 44, 1517–1525. [Google Scholar] [CrossRef]
  64. Lee, H.R.; Son, C.H.; Koh, E.K.; Bae, J.H.; Kang, C.D.; Yang, K.; Park, Y.S. Expansion of cytotoxic natural killer cells using irradiated autologous peripheral blood mononuclear cells and anti-CD16 antibody. Sci. Rep. 2017, 7, 11075. [Google Scholar] [CrossRef] [PubMed]
  65. Romee, R.; Foley, B.; Lenvik, T.; Wang, Y.; Zhang, B.; Ankarlo, D.; Luo, X.; Cooley, S.; Verneris, M.; Walcheck, B.; et al. NK cell CD16 surface expression and function is regulated by a disintegrin and metalloprotease-17 (ADAM17). Blood 2013, 121, 3599–3608. [Google Scholar] [CrossRef] [PubMed]
  66. Rajagopalan, S.; Fu, J.; Long, E.O. Cutting Edge: Induction of IFN-γ Production but Not Cytotoxicity by the Killer Cell Ig-Like Receptor KIR2DL4 (CD158d) in Resting NK Cells. J. Immunol. 2001, 167, 1877–1881. [Google Scholar] [CrossRef] [PubMed]
  67. Kandilarova, S.M.; Paschen, A.; Mihaylova, A.; Ivanova, M.; Schadendorf, D.; Naumova, E. The Influence of HLA and KIR Genes on Malignant Melanoma Development and Progression. Arch. Immunol. Ther. Exp. 2016, 64 (Suppl. S1), 73–81. [Google Scholar] [CrossRef] [PubMed]
  68. Campillo, J.A.; Legaz, I.; López-Álvarez, M.R.; Bolarín, J.M.; Heras, B.L.; Muro, M.; Minguela, A.; Moya-Quiles, M.R.; Blanco-García, R.; Martínez-Banaclocha, H.; et al. KIR gene variability in cutaneous malignant melanoma: Influence of KIR2D/HLA-C pairings on disease susceptibility and prognosis. Immunogenetics 2013, 65, 333–343. [Google Scholar] [CrossRef] [PubMed]
  69. Moradi, S.; Stankovic, S.; O’connor, G.M.; Pymm, P.; MacLachlan, B.J.; Faoro, C.; Retière, C.; Sullivan, L.C.; Saunders, P.M.; Widjaja, J.; et al. Structural plasticity of KIR2DL2 and KIR2DL3 enables altered docking geometries atop HLA-C. Nat. Commun. 2021, 12, 2173. [Google Scholar] [CrossRef] [PubMed]
  70. Frazier, W.R.; Steiner, N.; Hou, L.; Dakshanamurthy, S.; Hurley, C.K. Allelic Variation in KIR2DL3 Generates a KIR2DL2-like Receptor with Increased Binding to its HLA-C Ligand. J. Immunol. 2013, 190, 6198–6208. [Google Scholar] [CrossRef] [PubMed]
  71. Nguyen, S.; Beziat, V.; Dhedin, N.; Kuentz, M.; Vernant, J.P.; Debre, P.; Vieillard, V. HLA-E upregulation on IFN-γ-activated AML blasts impairs CD94/NKG2A-dependent NK cytolysis after haplo-mismatched hematopoietic SCT. Bone Marrow Transplant. 2009, 43, 693–699. [Google Scholar] [CrossRef]
  72. André, P.; Denis, C.; Soulas, C.; Bourbon-Caillet, C.; Lopez, J.; Arnoux, T.; Bléry, M.; Bonnafous, C.; Gauthier, L.; Morel, A.; et al. Anti-NKG2A mAb Is a Checkpoint Inhibitor that Promotes Anti-Tumor Immunity by Unleashing Both T and NK Cells. Cell 2018, 175, 1731–1743.e13. [Google Scholar] [CrossRef]
  73. Kamiya, T.; Seow, S.V.; Wong, D.; Robinson, M.; Campana, D. Blocking expression of inhibitory receptor NKG2A overcomes tumor resistance to NK cells. J. Clin. Investig. 2019, 129, 2094–2106. [Google Scholar] [CrossRef]
  74. Chauvin, J.M.; Pagliano, O.; Fourcade, J.; Sun, Z.; Wang, H.; Sander, C.; Kirkwood, J.M.; Chen, T.H.T.; Maurer, M.; Korman, A.J.; et al. TIGIT and PD-1 impair tumor antigen–specific CD8+ T cells in melanoma patients. J. Clin. Investig. 2015, 125, 2046–2058. [Google Scholar] [CrossRef] [PubMed]
  75. Inozume, T.; Yaguchi, T.; Furuta, J.; Harada, K.; Kawakami, Y.; Shimada, S. Melanoma Cells Control Antimelanoma CTL Responses via Interaction between TIGIT and CD155 in the Effector Phase. J. Investig. Dermatol. 2016, 136, 255–263. [Google Scholar] [CrossRef]
  76. Prager, I.; Watzl, C. Mechanisms of natural killer cell-mediated cellular cytotoxicity. J. Leukoc. Biol. 2019, 105, 1319–1329. [Google Scholar] [CrossRef] [PubMed]
  77. Elahi, R.; Heidary, A.H.; Hadiloo, K.; Esmaeilzadeh, A. Chimeric Antigen Receptor-Engineered Natural Killer (CAR NK) Cells in Cancer Treatment; Recent Advances and Future Prospects. Stem Cell Rev. Rep. 2021, 17, 2081–2106. [Google Scholar] [CrossRef]
  78. Chan, C.J.; Smyth, M.J.; Martinet, L. Molecular mechanisms of natural killer cell activation in response to cellular stress. Cell Death Differ. 2014, 21, 5–14. [Google Scholar] [CrossRef] [PubMed]
  79. Du, N.; Guo, F.; Wang, Y.; Cui, J. NK Cell Therapy: A Rising Star in Cancer Treatment. Cancers 2021, 13, 4129. [Google Scholar] [CrossRef]
  80. Cursons, J.; Souza-Fonseca-Guimaraes, F.; Foroutan, M.; Anderson, A.; Hollande, F.; Hediyeh-Zadeh, S.; Behren, A.; Huntington, N.D.; Davis, M.J. A Gene Signature Predicting Natural Killer Cell Infiltration and Improved Survival in Melanoma Patients. Cancer Immunol. Res. 2019, 7, 1162–1174. [Google Scholar] [CrossRef]
  81. Ali, T.H.; Pisanti, S.; Ciaglia, E.; Mortarini, R.; Anichini, A.; Garofalo, C.; Tallerico, R.; Santinami, M.; Gulletta, E.; Ietto, C.; et al. Enrichment of CD56dimKIR+CD57+ highly cytotoxic NK cells in tumour-infiltrated lymph nodes of melanoma patients. Nat. Commun. 2014, 5, 5639. [Google Scholar] [CrossRef]
  82. de Jonge, K.; Ebering, A.; Nassiri, S.; Hajjami, H.M.-E.; Ouertatani-Sakouhi, H.; Baumgaertner, P.; Speiser, D.E. Circulating CD56bright NK cells inversely correlate with survival of melanoma patients. Sci. Rep. 2019, 9, 4487. [Google Scholar] [CrossRef]
  83. Attrill, G.H.; Ferguson, P.M.; Palendira, U.; Long, G.V.; Wilmott, J.S.; Scolyer, R.A. The tumour immune landscape and its implications in cutaneous melanoma. Pigment. Cell Melanoma Res. 2021, 34, 529–549. [Google Scholar] [CrossRef]
  84. Vitale, M.; Cantoni, C.; Pietra, G.; Mingari, M.C.; Moretta, L. Effect of tumor cells and tumor microenvironment on NK-cell function. Eur. J. Immunol. 2014, 44, 1582–1592. [Google Scholar] [CrossRef] [PubMed]
  85. Falcone, I.; Conciatori, F.; Bazzichetto, C.; Ferretti, G.; Cognetti, F.; Ciuffreda, L.; Milella, M. Tumor Microenvironment: Implications in Melanoma Resistance to Targeted Therapy and Immunotherapy. Cancers 2020, 12, 2870. [Google Scholar] [CrossRef] [PubMed]
  86. Amalinei, C.; Grigoraș, A.; Lozneanu, L.; Căruntu, I.D.; Giușcă, S.E.; Balan, R.A. The Interplay between Tumour Microenvironment Components in Malignant Melanoma. Medicina 2022, 58, 365. [Google Scholar] [CrossRef] [PubMed]
  87. Kim, S.-H.; Roszik, J.; Cho, S.-N.; Ogata, D.; Milton, D.R.; Peng, W.; Menter, D.G.; Ekmekcioglu, S.; Grimm, E.A. The COX2 Effector Microsomal PGE2 Synthase 1 is a Regulator of Immunosuppression in Cutaneous Melanoma. Clin. Cancer Res. 2019, 25, 1650–1663. [Google Scholar] [CrossRef] [PubMed]
  88. Holt, D.; Ma, X.; Kundu, N.; Fulton, A. Prostaglandin E2 (PGE2) suppresses natural killer cell function primarily through the PGE2 receptor EP4. Cancer Immunol. Immunother. 2011, 60, 1577–1586. [Google Scholar] [CrossRef] [PubMed]
  89. Ferreira, M.; Krykbaeva, I.; Damsky, W.; Kluger, H.M.; Bosenberg, M. Evaluating the role of the COX2/PGE2 pathway in anti-melanoma immunity. J. Clin. Oncol. 2019, 37 (Suppl. S14), e14114. [Google Scholar] [CrossRef]
  90. 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]
  91. Hornyák, L.; Dobos, N.; Koncz, G.; Karányi, Z.; Páll, D.; Szabó, Z.; Halmos, G.; Székvölgyi, L. The Role of Indoleamine-2,3-Dioxygenase in Cancer Development, Diagnostics, and Therapy. Front. Immunol. 2018, 9, 151. [Google Scholar] [CrossRef]
  92. Zhang, J.; Han, X.; Hu, X.; Jin, F.; Gao, Z.; Yin, L.; Qin, J.; Yin, F.; Li, C.; Wang, Y. IDO1 impairs NK cell cytotoxicity by decreasing NKG2D/NKG2DLs via promoting miR-18a. Mol. Immunol. 2018, 103, 144–155. [Google Scholar] [CrossRef]
  93. Pietra, G.; Manzini, C.; Rivara, S.; Vitale, M.; Cantoni, C.; Petretto, A.; Balsamo, M.; Conte, R.; Benelli, R.; Minghelli, S.; et al. Melanoma Cells Inhibit Natural Killer Cell Function by Modulating the Expression of Activating Receptors and Cytolytic Activity. Cancer Res. 2012, 72, 1407–1415. [Google Scholar] [CrossRef]
  94. López-Cobo, S.; Pieper, N.; Campos-Silva, C.; García-Cuesta, E.M.; Reyburn, H.T.; Paschen, A.; Valés-Gómez, M. Impaired NK cell recognition of vemurafenib-treated melanoma cells is overcome by simultaneous application of histone deacetylase inhibitors. Oncoimmunology 2018, 7, e1392426. [Google Scholar] [CrossRef] [PubMed]
  95. Peruzzi, G.; Femnou, L.; Gil-Krzewska, A.; Borrego, F.; Weck, J.; Krzewski, K.; Coligan, J.E. Membrane-Type 6 Matrix Metalloproteinase Regulates the Activation-Induced Downmodulation of CD16 in Human Primary NK Cells. J. Immunol. 2013, 191, 1883–1894. [Google Scholar] [CrossRef]
  96. Liu, G.; Atteridge, C.L.; Wang, X.; Lundgren, A.D.; Wu, J.D. Cutting Edge: The Membrane Type Matrix Metalloproteinase MMP14 Mediates Constitutive Shedding of MHC Class I Chain-Related Molecule a Independent of a Disintegrin and Metalloproteinases. J. Immunol. 2010, 184, 3346–3350. [Google Scholar] [CrossRef] [PubMed]
  97. Chitadze, G.; Lettau, M.; Bhat, J.; Wesch, D.; Steinle, A.; Fürst, D.; Mytilineos, J.; Kalthoff, H.; Janssen, O.; Oberg, H.; et al. Shedding of endogenous MHC class I-related chain molecules A and B from different human tumor entities: Heterogeneous involvement of the “a disintegrin and metalloproteases” 10 and 17. Int. J. Cancer 2013, 133, 1557–1566. [Google Scholar] [CrossRef]
  98. Schlecker, E.; Fiegler, N.; Arnold, A.; Altevogt, P.; Rose-John, S.; Moldenhauer, G.; Sucker, A.; Paschen, A.; von Strandmann, E.P.; Textor, S.; et al. Metalloprotease-Mediated Tumor Cell Shedding of B7-H6, the Ligand of the Natural Killer Cell–Activating Receptor NKp30. Cancer Res. 2014, 74, 3429–3440. [Google Scholar] [CrossRef] [PubMed]
  99. Deng, W.; Gowen, B.G.; Zhang, L.; Wang, L.; Lau, S.; Iannello, A.; Xu, J.; Rovis, T.L.; Xiong, N.; Raulet, D.H. A shed NKG2D ligand that promotes natural killer cell activation and tumor rejection. Science 2015, 348, 136–139. [Google Scholar] [CrossRef]
  100. Zhang, J.; Basher, F.; Wu, J.D. NKG2D Ligands in Tumor Immunity: Two Sides of a Coin. Front. Immunol. 2015, 6, 97. [Google Scholar] [CrossRef]
  101. Ichise, H.; Tsukamoto, S.; Hirashima, T.; Konishi, Y.; Oki, C.; Tsukiji, S.; Iwano, S.; Miyawaki, A.; Sumiyama, K.; Terai, K.; et al. Functional visualization of NK cell-mediated killing of metastatic single tumor cells. Elife 2022, 11, e76269. [Google Scholar] [CrossRef]
  102. de Andrade, L.F.; Kumar, S.; Luoma, A.M.; Ito, Y.; da Silva, P.H.A.; Pan, D.; Pyrdol, J.W.; Yoon, C.H.; Wucherpfennig, K.W. Inhibition of MICA and MICB Shedding Elicits NK-Cell–Mediated Immunity against Tumors Resistant to Cytotoxic T Cells. Cancer Immunol. Res. 2020, 8, 769–780. [Google Scholar] [CrossRef]
  103. de Andrade, L.F.; Tay, R.E.; Pan, D.; Luoma, A.M.; Ito, Y.; Badrinath, S.; Tsoucas, D.; Franz, B.; May, K.F., Jr.; Harvey, C.J.; et al. Antibody-mediated inhibition of MICA and MICB shedding promotes NK cell–driven tumor immunity. Science 2018, 359, 1537–1542. [Google Scholar] [CrossRef]
  104. Wang, X.; Xing, Z.; Chen, H.; Yang, H.; Wang, Q.; Xing, T. High expression of nectin-1 indicates a poor prognosis and promotes metastasis in hepatocellular carcinoma. Front. Oncol. 2022, 12, 953529. [Google Scholar] [CrossRef] [PubMed]
  105. Deuss, F.A.; Gully, B.S.; Rossjohn, J.; Berry, R. Recognition of nectin-2 by the natural killer cell receptor T cell immunoglobulin and ITIM domain (TIGIT). J. Biol. Chem. 2017, 292, 11413–11422. [Google Scholar] [CrossRef] [PubMed]
  106. Stanietsky, N.; Simic, H.; Arapovic, J.; Toporik, A.; Levy, O.; Novik, A.; Levine, Z.; Beiman, M.; Dassa, L.; Achdout, H.; et al. The interaction of TIGIT with PVR and PVRL2 inhibits human NK cell cytotoxicity. Proc. Natl. Acad. Sci. USA 2009, 106, 17858–17863. [Google Scholar] [CrossRef] [PubMed]
  107. Li, M.; Xia, P.; Du, Y.; Liu, S.; Huang, G.; Chen, J.; Zhang, H.; Hou, N.; Cheng, X.; Zhou, L.; et al. T-cell Immunoglobulin and ITIM Domain (TIGIT) Receptor/Poliovirus Receptor (PVR) Ligand Engagement Suppresses Interferon-γ Production of Natural Killer Cells via β-Arrestin 2-mediated Negative Signaling. J. Biol. Chem. 2014, 289, 17647–17657. [Google Scholar] [CrossRef] [PubMed]
  108. Siva, A.; Xin, H.; Qin, F.; Oltean, D.; Bowdish, K.S.; Kretz-Rommel, A. Immune modulation by melanoma and ovarian tumor cells through expression of the immunosuppressive molecule CD200. Cancer Immunol. Immunother. 2008, 57, 987–996. [Google Scholar] [CrossRef] [PubMed]
  109. Saputro, R.D.; Rinonce, H.T.; Iramawasita, Y.; Ridho, M.R.; Pudjohartono, M.F.; Anwar, S.L.; Setiaji, K.; Aryandono, T. Potential prognostic value of PD-L1 and NKG2A expression in Indonesian patients with skin nodular melanoma. BMC Res. Notes 2021, 14, 206. [Google Scholar] [CrossRef] [PubMed]
  110. Lee, J.H.; Shklovskaya, E.; Lim, S.Y.; Carlino, M.S.; Menzies, A.M.; Stewart, A.; Pedersen, B.; Irvine, M.; Alavi, S.; Yang, J.Y.H.; et al. Transcriptional downregulation of MHC class I and melanoma de-differentiation in resistance to PD-1 inhibition. Nat. Commun. 2020, 11, 1897. [Google Scholar] [CrossRef]
  111. Soulas, C.; Remark, R.; Brezar, V.; Lopez, J.; Bonnet, E.; Caraguel, F.; Lalanne, A.; Hoffmann, C.; Denis, C.; Arnoux, T.; et al. Abstract 2714: Combination of monalizumab and durvalumab as a potent immunotherapy treatment for solid human cancers. Cancer Res. 2018, 78 (Suppl. S13). [Google Scholar] [CrossRef]
  112. Carosella, E.D.; Rouas-Freiss, N.; Roux, D.T.L.; Moreau, P.; LeMaoult, J. HLA-G: An Immune Checkpoint Molecule. Adv. Immunol. 2015, 127, 33–144. [Google Scholar] [CrossRef]
  113. Lin, A.; Yan, W.H. Heterogeneity of HLA-G Expression in Cancers: Facing the Challenges. Front. Immunol. 2018, 9, 2164. [Google Scholar] [CrossRef]
  114. Fisher, J.G.; Doyle, A.D.P.; Graham, L.V.; Khakoo, S.I.; Blunt, M.D. Disruption of the NKG2A:HLA-E Immune Checkpoint Axis to Enhance NK Cell Activation against Cancer. Vaccines 2022, 10, 1993. [Google Scholar] [CrossRef] [PubMed]
  115. Liu, X.; Song, J.; Zhang, H.; Liu, X.; Zuo, F.; Zhao, Y.; Zhao, Y.; Yin, X.; Guo, X.; Wu, X.; et al. Immune checkpoint HLA-E:CD94-NKG2A mediates evasion of circulating tumor cells from NK cell surveillance. Cancer Cell 2023, 41, 272–287.e9. [Google Scholar] [CrossRef] [PubMed]
  116. Fourcade, J.; Sun, Z.; Benallaoua, M.; Guillaume, P.; Luescher, I.F.; Sander, C.; Kirkwood, J.M.; Kuchroo, V.; Zarour, H.M. Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen–specific CD8+ T cell dysfunction in melanoma patients. J. Exp. Med. 2010, 207, 2175–2186. [Google Scholar] [CrossRef]
  117. da Silva, I.P.; Gallois, A.; Jimenez-Baranda, S.; Khan, S.; Anderson, A.C.; Kuchroo, V.K.; Osman, I.; Bhardwaj, N. Reversal of NK-Cell Exhaustion in Advanced Melanoma by Tim-3 Blockade. Cancer Immunol. Res. 2014, 2, 410–422. [Google Scholar] [CrossRef] [PubMed]
  118. Whiteside, T.L. Exosomes and tumor-mediated immune suppression. J. Clin. Investig. 2016, 126, 1216–1223. [Google Scholar] [CrossRef]
  119. Sharma, P.; Diergaarde, B.; Ferrone, S.; Kirkwood, J.M.; Whiteside, T.L. Melanoma cell-derived exosomes in plasma of melanoma patients suppress functions of immune effector cells. Sci. Rep. 2020, 10, 92. [Google Scholar] [CrossRef] [PubMed]
  120. 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]
  121. Mao, X.; Xu, J.; Wang, W.; Liang, C.; Hua, J.; Liu, J.; Zhang, B.; Meng, Q.; Yu, X.; Shi, S. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: New findings and future perspectives. Mol. Cancer 2021, 20, 131. [Google Scholar] [CrossRef]
  122. Harper, J.; Sainson, R.C.A. Regulation of the anti-tumour immune response by cancer-associated fibroblasts. Semin. Cancer Biol. 2014, 25, 69–77. [Google Scholar] [CrossRef]
  123. Ziani, L.; Ben Safta-Saadoun, T.; Gourbeix, J.; Cavalcanti, A.; Robert, C.; Favre, G.; Chouaib, S.; Thiery, J. Melanoma-associated fibroblasts decrease tumor cell susceptibility to NK cell-mediated killing through matrix-metalloproteinases secretion. Oncotarget 2017, 8, 19780–19794. [Google Scholar] [CrossRef]
  124. Du, Y.; Fang, Q.; Zheng, S.G. Regulatory T Cells: Concept, Classification, Phenotype, and Biological Characteristics. In T Regulatory Cells in Human Health and Diseases; Springer: Berlin/Heidelberg, Germany, 2021; pp. 1–31. [Google Scholar] [CrossRef]
  125. Allard, B.; Longhi, M.S.; Robson, S.C.; Stagg, J. The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets. Immunol. Rev. 2017, 276, 121–144. [Google Scholar] [CrossRef] [PubMed]
  126. Cekic, C.; Day, Y.-J.; Sag, D.; Linden, J. Myeloid Expression of Adenosine A2A Receptor Suppresses T and NK Cell Responses in the Solid Tumor Microenvironment. Cancer Res. 2014, 74, 7250–7259. [Google Scholar] [CrossRef] [PubMed]
  127. Wang, H.; Yang, L.; Wang, D.; Zhang, Q.; Zhang, L. Pro-tumor activities of macrophages in the progression of melanoma. Hum. Vaccin. Immunother. 2017, 13, 1556–1562. [Google Scholar] [CrossRef] [PubMed]
  128. Bohn, T.; Rapp, S.; Luther, N.; Klein, M.; Bruehl, T.J.; Kojima, N.; Lopez, P.A.; Hahlbrock, J.; Muth, S.; Endo, S.; et al. Tumor immunoevasion via acidosis-dependent induction of regulatory tumor-associated macrophages. Nat. Immunol. 2018, 19, 1319–1329. [Google Scholar] [CrossRef] [PubMed]
  129. Chanmee, T.; Ontong, P.; Konno, K.; Itano, N. Tumor-Associated Macrophages as Major Players in the Tumor Microenvironment. Cancers 2014, 6, 1670–1690. [Google Scholar] [CrossRef]
  130. Rébé, C.; Ghiringhelli, F. Interleukin-1β and Cancer. Cancers 2020, 12, 1791. [Google Scholar] [CrossRef]
  131. Smith, M.P.; Sanchez-Laorden, B.; O’Brien, K.; Brunton, H.; Ferguson, J.; Young, H.; Dhomen, N.; Flaherty, K.T.; Frederick, D.T.; Cooper, Z.A.; et al. The Immune Microenvironment Confers Resistance to MAPK Pathway Inhibitors through Macrophage-Derived TNFα. Cancer Discov. 2014, 4, 1214–1229. [Google Scholar] [CrossRef]
  132. Érsek, B.; Silló, P.; Cakir, U.; Molnár, V.; Bencsik, A.; Mayer, B.; Mezey, E.; Kárpáti, S.; Pós, Z.; Németh, K. Melanoma-associated fibroblasts impair CD8+ T cell function and modify expression of immune checkpoint regulators via increased arginase activity. Cell. Mol. Life Sci. 2021, 78, 661–673. [Google Scholar] [CrossRef]
  133. Brownlie, D.; Doughty-Shenton, D.; Soong, D.Y.; Nixon, C.; O Carragher, N.; Carlin, L.M.; Kitamura, T. Metastasis-associated macrophages constrain antitumor capability of natural killer cells in the metastatic site at least partially by membrane bound transforming growth factor β. J. Immunother. Cancer 2021, 9, e001740. [Google Scholar] [CrossRef]
  134. D’aguanno, S.; Mallone, F.; Marenco, M.; Del Bufalo, D.; Moramarco, A. Hypoxia-dependent drivers of melanoma progression. J. Exp. Clin. Cancer Res. 2021, 40, 159. [Google Scholar] [CrossRef]
  135. Böhme, I.; Bosserhoff, A.K. Acidic tumor microenvironment in human melanoma. Pigment. Cell Melanoma Res. 2016, 29, 508–523. [Google Scholar] [CrossRef] [PubMed]
  136. Muz, B.; de la Puente, P.; Azab, F.; Azab, A.K. The role of hypoxia in cancer progression, angiogenesis, metastasis, and resistance to therapy. Hypoxia 2015, 3, 83–92. [Google Scholar] [CrossRef] [PubMed]
  137. Terrén, I.; Orrantia, A.; Vitallé, J.; Zenarruzabeitia, O.; Borrego, F. NK Cell Metabolism and Tumor Microenvironment. Front. Immunol. 2019, 10, 2278. [Google Scholar] [CrossRef] [PubMed]
  138. Garcés-Lázaro, I.; Kotzur, R.; Cerwenka, A.; Mandelboim, O. NK Cells Under Hypoxia: The Two Faces of Vascularization in Tumor and Pregnancy. Front. Immunol. 2022, 13, 924775. [Google Scholar] [CrossRef]
  139. Lu, H.; Zhao, X.; Li, Z.; Hu, Y.; Wang, H. From CAR-T Cells to CAR-NK Cells: A Developing Immunotherapy Method for Hematological Malignancies. Front. Oncol. 2021, 11, 720501. [Google Scholar] [CrossRef] [PubMed]
  140. Guedan, S.; Calderon, H.; Posey, A.D., Jr.; Maus, M.V. Engineering and Design of Chimeric Antigen Receptors. Mol. Ther. Methods Clin. Dev. 2018, 12, 145–156. [Google Scholar] [CrossRef] [PubMed]
  141. Guedan, S.; Posey, A.D., Jr.; Shaw, C.; Wing, A.; Da, T.; Patel, P.R.; McGettigan, S.E.; Casado-Medrano, V.; Kawalekar, O.U.; Uribe-Herranz, M.; et al. Enhancing CAR T cell persistence through ICOS and 4-1BB costimulation. JCI Insight 2018, 3, e96976. [Google Scholar] [CrossRef] [PubMed]
  142. Wu, C.Y.; Roybal, K.T.; Puchner, E.M.; Onuffer, J.; Lim, W.A. Remote control of therapeutic T cells through a small molecule–gated chimeric receptor. Science 2015, 350, aab4077. [Google Scholar] [CrossRef]
  143. Christodoulou, I.; Ho, W.J.; Marple, A.; Ravich, J.W.; Tam, A.; Rahnama, R.; Fearnow, A.; Rietberg, C.; Yanik, S.; E Solomou, E.; et al. Engineering CAR-NK cells to secrete IL-15 sustains their anti-AML functionality but is associated with systemic toxicities. J. Immunother. Cancer 2021, 9, e003894. [Google Scholar] [CrossRef]
  144. Wrona, E.; Borowiec, M.; Potemski, P. CAR-NK Cells in the Treatment of Solid Tumors. Int. J. Mol. Sci. 2021, 22, 5899. [Google Scholar] [CrossRef]
  145. Zhang, Q.; Tian, K.; Xu, J.; Zhang, H.; Li, L.; Fu, Q.; Chai, D.; Li, H.; Zheng, J. Synergistic Effects of Cabozantinib and EGFR-Specific CAR-NK-92 Cells in Renal Cell Carcinoma. J. Immunol. Res. 2017, 2017, 6915912. [Google Scholar] [CrossRef]
  146. Gossel, L.D.H.; Heim, C.; Pfeffermann, L.-M.; Moser, L.M.; Bönig, H.B.; Klingebiel, T.E.; Bader, P.; Wels, W.S.; Merker, M.; Rettinger, E. Retargeting of NK-92 Cells against High-Risk Rhabdomyosarcomas by Means of an ERBB2 (HER2/Neu)-Specific Chimeric Antigen Receptor. Cancers 2021, 13, 1443. [Google Scholar] [CrossRef] [PubMed]
  147. Navarrete-Galvan, L.; Guglielmo, M.; Amaya, J.C.; Smith-Gagen, J.; Lombardi, V.C.; Merica, R.; Hudig, D. Optimizing NK-92 serial killers: Gamma irradiation, CD95/Fas-ligation, and NK or LAK attack limit cytotoxic efficacy. J. Transl. Med. 2022, 20, 151. [Google Scholar] [CrossRef] [PubMed]
  148. Klingemann, H.; Boissel, L.; Toneguzzo, F. Natural Killer Cells for Immunotherapy—Advantages of the NK-92 Cell Line over Blood NK Cells. Front. Immunol. 2016, 7, 91. [Google Scholar] [CrossRef]
  149. Maddineni, S.; Silberstein, J.L.; Sunwoo, J.B. Emerging NK cell therapies for cancer and the promise of next generation engineering of iPSC-derived NK cells. J. Immunother. Cancer 2022, 10, e004693. [Google Scholar] [CrossRef] [PubMed]
  150. Guercio, M.; Manni, S.; Boffa, I.; Caruso, S.; Di Cecca, S.; Sinibaldi, M.; Abbaszadeh, Z.; Camera, A.; Ciccone, R.; Polito, V.A.; et al. Inclusion of the Inducible Caspase 9 Suicide Gene in CAR Construct Increases Safety of CAR.CD19 T Cell Therapy in B-Cell Malignancies. Front. Immunol. 2021, 12, 755639. [Google Scholar] [CrossRef] [PubMed]
  151. Mehta, R.S.; Shpall, E.J.; Rezvani, K. Cord Blood as a Source of Natural Killer Cells. Front. Med. 2016, 2, 93. [Google Scholar] [CrossRef]
  152. Shaim, H.; Yvon, E. Cord blood: A promising source of allogeneic natural killer cells for immunotherapy. Cytotherapy 2015, 17, 1–2. [Google Scholar] [CrossRef]
  153. Liu, E.; Tong, Y.; Dotti, G.; Shaim, H.; Savoldo, B.; Mukherjee, M.; Orange, J.; Wan, X.; Lu, X.; Reynolds, A.; et al. Cord blood NK cells engineered to express IL-15 and a CD19-targeted CAR show long-term persistence and potent antitumor activity. Leukemia 2018, 32, 520–531. [Google Scholar] [CrossRef]
  154. Li, Y.; Yin, J.; Li, T.; Huang, S.; Yan, H.; Leavenworth, J.; Wang, X. NK cell-based cancer immunotherapy: From basic biology to clinical application. Sci. China Life Sci. 2015, 58, 1233–1245. [Google Scholar] [CrossRef]
  155. Xiao, L.; Cen, D.; Gan, H.; Sun, Y.; Huang, N.; Xiong, H.; Jin, Q.; Su, L.; Liu, X.; Wang, K.; et al. Adoptive Transfer of NKG2D CAR mRNA-Engineered Natural Killer Cells in Colorectal Cancer Patients. Mol. Ther. 2019, 27, 1114–1125. [Google Scholar] [CrossRef] [PubMed]
  156. Focaccetti, C.; Benvenuto, M.; Pighi, C.; Vitelli, A.; Napolitano, F.; Cotugno, N.; Fruci, D.; Palma, P.; Rossi, P.; Bei, R.; et al. DNAM-1-chimeric receptor-engineered NK cells, combined with Nutlin-3a, more effectively fight neuroblastoma cells in vitro: A proof-of-concept study. Front. Immunol. 2022, 13, 886319. [Google Scholar] [CrossRef] [PubMed]
  157. Huang, Y.; Zeng, J.; Liu, T.; Xu, Q.; Song, X.; Zeng, J. DNAM1 and 2B4 Costimulatory Domains Enhance the Cytotoxicity of Anti-GPC3 Chimeric Antigen Receptor-Modified Natural Killer Cells Against Hepatocellular Cancer Cells in vitro. Cancer Manag. Res. 2020, 12, 3247–3255. [Google Scholar] [CrossRef] [PubMed]
  158. Cao, B.; Liu, M.; Huang, J.; Zhou, J.; Li, J.; Lian, H.; Huang, W.; Guo, Y.; Yang, S.; Lin, L.; et al. Development of mesothelin-specific CAR NK-92 cells for the treatment of gastric cancer. Int. J. Biol. Sci. 2021, 17, 3850–3861. [Google Scholar] [CrossRef]
  159. Liu, M.; Huang, W.; Guo, Y.; Zhou, Y.; Zhi, C.; Chen, J.; Li, J.; He, J.; Lian, H.; Zhou, J.; et al. CAR NK-92 cells targeting DLL3 kill effectively small cell lung cancer cells in vitro and in vivo. J. Leukoc. Biol. 2022, 112, 901–911. [Google Scholar] [CrossRef] [PubMed]
  160. Peng, Y.; Zhang, W.; Chen, Y.; Zhang, L.; Shen, H.; Wang, Z.; Tian, S.; Yang, X.; Cui, D.; He, Y.; et al. Engineering c-Met-CAR NK-92 cells as a promising therapeutic candidate for lung adenocarcinoma. Pharmacol. Res. 2023, 188, 106656. [Google Scholar] [CrossRef]
  161. Yang, S.; Cao, B.; Zhou, G.; Zhu, L.; Wang, L.; Zhang, L.; Kwok, H.F.; Zhang, Z.; Zhao, Q. Targeting B7-H3 Immune Checkpoint with Chimeric Antigen Receptor-Engineered Natural Killer Cells Exhibits Potent Cytotoxicity against Non-Small Cell Lung Cancer. Front. Pharmacol. 2020, 11, 1089. [Google Scholar] [CrossRef]
  162. Pekar, L.; Klausz, K.; Busch, M.; Valldorf, B.; Kolmar, H.; Wesch, D.; Oberg, H.-H.; Krohn, S.; Boje, A.S.; Gehlert, C.L.; et al. Affinity Maturation of B7-H6 Translates into Enhanced NK Cell–Mediated Tumor Cell Lysis and Improved Proinflammatory Cytokine Release of Bispecific Immunoligands via NKp30 Engagement. J. Immunol. 2021, 206, 225–236. [Google Scholar] [CrossRef]
  163. Klapdor, R.; Wang, S.; Morgan, M.; Dörk, T.; Hacker, U.; Hillemanns, P.; Büning, H.; Schambach, A. Characterization of a Novel Third-Generation Anti-CD24-CAR against Ovarian Cancer. Int. J. Mol. Sci. 2019, 20, 660. [Google Scholar] [CrossRef]
  164. Grote, S.; Ureña-Bailén, G.; Chan, K.C.-H.; Baden, C.; Mezger, M.; Handgretinger, R.; Schleicher, S. In Vitro Evaluation of CD276-CAR NK-92 Functionality, Migration and Invasion Potential in the Presence of Immune Inhibitory Factors of the Tumor Microenvironment. Cells 2021, 10, 1020. [Google Scholar] [CrossRef]
  165. Klapdor, R.; Wang, S.; Morgan, M.A.; Zimmermann, K.; Hachenberg, J.; Büning, H.; Dörk, T.; Hillemanns, P.; Schambach, A. NK Cell-Mediated Eradication of Ovarian Cancer Cells with a Novel Chimeric Antigen Receptor Directed against CD44. Biomedicines 2021, 9, 1339. [Google Scholar] [CrossRef]
  166. Zhang, Z.; He, B. CLDN6-CAR-NK Cell Therapy for Advanced Solid Tumors. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT05410717 (accessed on 9 October 2023).
  167. Murakami, T.; Nakazawa, T.; Natsume, A.; Nishimura, F.; Nakamura, M.; Matsuda, R.; Omoto, K.; Tanaka, Y.; Shida, Y.; Park, Y.-S.; et al. Novel Human NK Cell Line Carrying CAR Targeting EGFRvIII Induces Antitumor Effects in Glioblastoma Cells. Anticancer Res. 2018, 38, 5049–5056. [Google Scholar] [CrossRef] [PubMed]
  168. Müller, N.; Michen, S.; Tietze, S.; Töpfer, K.; Schulte, A.; Lamszus, K.; Schmitz, M.; Schackert, G.; Pastan, I.; Temme, A. Engineering NK Cells Modified with an EGFRvIII-specific Chimeric Antigen Receptor to Overexpress CXCR4 Improves Immunotherapy of CXCL12/SDF-1α-secreting Glioblastoma. J. Immunother. 2015, 38, 197–210. [Google Scholar] [CrossRef] [PubMed]
  169. Liu, Y.; Zhou, Y.; Huang, K.; Fang, X.; Li, Y.; Wang, F.; An, L.; Chen, Q.; Zhang, Y.; Shi, A.; et al. Targeting epidermal growth factor-overexpressing triple-negative breast cancer by natural killer cells expressing a specific chimeric antigen receptor. Cell Prolif. 2020, 53, e12858. [Google Scholar] [CrossRef] [PubMed]
  170. Han, J.; Chu, J.; Chan, W.K.; Zhang, J.; Wang, Y.; Cohen, J.B.; Victor, A.; Meisen, W.H.; Kim, S.-H.; Grandi, P.; et al. CAR-Engineered NK Cells Targeting Wild-Type EGFR and EGFRvIII Enhance Killing of Glioblastoma and Patient-Derived Glioblastoma Stem Cells. Sci. Rep. 2015, 5, 11483. [Google Scholar] [CrossRef]
  171. Sahm, C.; Schönfeld, K.; Wels, W.S. Expression of IL-15 in NK cells results in rapid enrichment and selective cytotoxicity of gene-modified effectors that carry a tumor-specific antigen receptor. Cancer Immunol. Immunother. 2012, 61, 1451–1461. [Google Scholar] [CrossRef] [PubMed]
  172. Mitwasi, N.; Feldmann, A.; Arndt, C.; Koristka, S.; Berndt, N.; Jureczek, J.; Loureiro, L.R.; Bergmann, R.; Máthé, D.; Hegedüs, N.; et al. “UniCAR”-modified off-the-shelf NK-92 cells for targeting of GD2-expressing tumour cells. Sci. Rep. 2020, 10, 2141. [Google Scholar] [CrossRef] [PubMed]
  173. Rudek, L.S.; Zimmermann, K.; Galla, M.; Meyer, J.; Kuehle, J.; Stamopoulou, A.; Brand, D.; Sandalcioglu, I.E.; Neyazi, B.; Moritz, T.; et al. Generation of an NFκB-Driven Alpharetroviral “All-in-One” Vector Construct as a Potent Tool for CAR NK Cell Therapy. Front. Immunol. 2021, 12, 751138. [Google Scholar] [CrossRef]
  174. Kailayangiri, S.; Altvater, B.; Spurny, C.; Jamitzky, S.; Schelhaas, S.; Jacobs, A.H.; Wiek, C.; Roellecke, K.; Hanenberg, H.; Hartmann, W.; et al. Targeting Ewing sarcoma with activated and GD2-specific chimeric antigen receptor-engineered human NK cells induces upregulation of immune-inhibitory HLA-G. Oncoimmunology 2017, 6, e1250050. [Google Scholar] [CrossRef]
  175. Zhang, G.; Liu, R.; Zhu, X.; Wang, L.; Ma, J.; Han, H.; Wang, X.; Zhang, G.; He, W.; Wang, W.; et al. Retargeting NK-92 for anti-melanoma activity by a TCR-like single-domain antibody. Immunol. Cell Biol. 2013, 91, 615–624. [Google Scholar] [CrossRef]
  176. Zhang, C.; Burger, M.C.; Jennewein, L.; Genßler, S.; Schönfeld, K.; Zeiner, P.; Hattingen, E.; Harter, P.N.; Mittelbronn, M.; Tonn, T.; et al. ErbB2/HER2-Specific NK Cells for Targeted Therapy of Glioblastoma. J. Natl. Cancer Inst. 2016, 108, djv375. [Google Scholar] [CrossRef] [PubMed]
  177. Schönfeld, K.; Sahm, C.; Zhang, C.; Naundorf, S.; Brendel, C.; Odendahl, M.; Nowakowska, P.; Bönig, H.; Köhl, U.; Kloess, S.; et al. Selective Inhibition of Tumor Growth by Clonal NK Cells Expressing an ErbB2/HER2-Specific Chimeric Antigen Receptor. Mol. Ther. 2015, 23, 330–338. [Google Scholar] [CrossRef] [PubMed]
  178. Merker, M.; Wagner, J.; Kreyenberg, H.; Heim, C.; Moser, L.M.; Wels, W.S.; Bonig, H.; Ivics, Z.; Ullrich, E.; Klingebiel, T.; et al. ERBB2-CAR-Engineered Cytokine-Induced Killer Cells Exhibit Both CAR-Mediated and Innate Immunity Against High-Risk Rhabdomyosarcoma. Front. Immunol. 2020, 11, 581468. [Google Scholar] [CrossRef] [PubMed]
  179. Li, Q.; Wang, Y.; Lin, M.; Xia, L.; Bao, Y.; Sun, X.; Yang, L. Abstract A014: Phase I clinical trial with PD-1/MUC1 CAR-pNK92 immunotherapy. Cancer Immunol. Res. 2019, 7 (Suppl. S2), A014. [Google Scholar] [CrossRef]
  180. Kulemzin, S.V.; Gorchakov, A.A.; Chikaev, A.N.; Kuznetsova, V.V.; Volkova, O.Y.; Matvienko, D.A.; Petukhov, A.V.; Zaritskey, A.Y.; Taranin, A.V. VEGFR2-specific FnCAR effectively redirects the cytotoxic activity of T cells and YT NK cells. Oncotarget 2018, 9, 9021–9029. [Google Scholar] [CrossRef] [PubMed]
  181. Zhang, S.; Chen, K.; Liu, H.; Jing, C.; Zhang, X.; Qu, C.; Yu, S. PMEL as a Prognostic Biomarker and Negatively Associated With Immune Infiltration in Skin Cutaneous Melanoma (SKCM). J. Immunother. 2021, 44, 214–223. [Google Scholar] [CrossRef]
  182. Strobel, S.B.; Machiraju, D.; Hülsmeyer, I.; Becker, J.C.; Paschen, A.; Jäger, D.; Wels, W.S.; Bachmann, M.; Hassel, J.C. Expression of Potential Targets for Cell-Based Therapies on Melanoma Cells. Life 2021, 11, 269. [Google Scholar] [CrossRef]
  183. Golinelli, G.; Grisendi, G.; Prapa, M.; Bestagno, M.; Spano, C.; Rossignoli, F.; Bambi, F.; Sardi, I.; Cellini, M.; Horwitz, E.M.; et al. Targeting GD2-positive glioblastoma by chimeric antigen receptor empowered mesenchymal progenitors. Cancer Gene Ther. 2020, 27, 558–570. [Google Scholar] [CrossRef]
  184. Kailayangiri, S.; Altvater, B.; Meltzer, J.; Pscherer, S.; Luecke, A.; Dierkes, C.; Titze, U.; Leuchte, K.; Landmeier, S.; Hotfilder, M.; et al. The ganglioside antigen GD2 is surface-expressed in Ewing sarcoma and allows for MHC-independent immune targeting. Br. J. Cancer 2012, 106, 1123–1133. [Google Scholar] [CrossRef]
  185. Reppel, L.; Tsahouridis, O.; Akulian, J.; Davis, I.J.; Lee, H.; Fucà, G.; Weiss, J.; Dotti, G.; Pecot, C.V.; Savoldo, B. Targeting disialoganglioside GD2 with chimeric antigen receptor-redirected T cells in lung cancer. J. Immunother. Cancer 2022, 10, e003897. [Google Scholar] [CrossRef]
  186. Yu, J.; Wu, X.; Yan, J.; Yu, H.; Xu, L.; Chi, Z.; Sheng, X.; Si, L.; Cui, C.; Dai, J.; et al. Anti-GD2/4-1BB chimeric antigen receptor T cell therapy for the treatment of Chinese melanoma patients. J. Hematol. Oncol. 2018, 11, 1. [Google Scholar] [CrossRef] [PubMed]
  187. Doronin, I.I.; Vishnyakova, P.A.; Kholodenko, I.V.; Ponomarev, E.D.; Ryazantsev, D.Y.; Molotkovskaya, I.M.; Kholodenko, R.V. Ganglioside GD2 in reception and transduction of cell death signal in tumor cells. BMC Cancer 2014, 14, 295. [Google Scholar] [CrossRef] [PubMed]
  188. Suzuki, M.; Cheung, N.K.V. Disialoganglioside GD2 as a therapeutic target for human diseases. Expert Opin. Ther. Targets 2015, 19, 349–362. [Google Scholar] [CrossRef] [PubMed]
  189. Zimmermann, K.; Kuehle, J.; Dragon, A.C.; Galla, M.; Kloth, C.; Rudek, L.S.; Sandalcioglu, I.E.; Neyazi, B.; Moritz, T.; Meyer, J.; et al. Design and Characterization of an “All-in-One” Lentiviral Vector System Combining Constitutive Anti-GD2 CAR Expression and Inducible Cytokines. Cancers 2020, 12, 375. [Google Scholar] [CrossRef] [PubMed]
  190. Castellanos, J.R.; Purvis, I.J.; Labak, C.M.; Guda, M.R.; Tsung, A.J.; Velpula, K.K.; Asuthkar, S. B7-H3 role in the immune landscape of cancer. Am. J. Clin. Exp. Immunol. 2017, 6, 66–75. [Google Scholar] [PubMed]
  191. Hofmeyer, K.A.; Ray, A.; Zang, X. The contrasting role of B7-H3. Proc. Natl. Acad. Sci. USA 2008, 105, 10277–10278. [Google Scholar] [CrossRef] [PubMed]
  192. Chen, H.; Duan, X.; Deng, X.; Huang, Y.; Zhou, X.; Zhang, S.; Zhang, X.; Liu, P.; Yang, C.; Liu, G.; et al. EBV-Upregulated B7-H3 Inhibits NK cell–Mediated Antitumor Function and Contributes to Nasopharyngeal Carcinoma Progression. Cancer Immunol. Res. 2023, 11, 830–846. [Google Scholar] [CrossRef]
  193. Lee, Y.H.; Martin-Orozco, N.; Zheng, P.; Li, J.; Zhang, P.; Tan, H.; Park, H.J.; Jeong, M.; Chang, S.H.; Kim, B.S.; et al. Inhibition of the B7-H3 immune checkpoint limits tumor growth by enhancing cytotoxic lymphocyte function. Cell Res. 2017, 27, 1034–1045. [Google Scholar] [CrossRef]
  194. Zhang, Z.; Jiang, C.; Liu, Z.; Yang, M.; Tang, X.; Wang, Y.; Zheng, M.; Huang, J.; Zhong, K.; Zhao, S.; et al. B7-H3-Targeted CAR-T Cells Exhibit Potent Antitumor Effects on Hematologic and Solid Tumors. Mol. Ther. Oncolytics 2020, 17, 180–189. [Google Scholar] [CrossRef]
  195. Yamato, M.; Hasegawa, J.; Maejima, T.; Hattori, C.; Kumagai, K.; Watanabe, A.; Nishiya, Y.; Shibutani, T.; Aida, T.; Hayakawa, I.; et al. DS-7300a, a DNA Topoisomerase I Inhibitor, DXd-Based Antibody–Drug Conjugate Targeting B7-H3, Exerts Potent Antitumor Activities in Preclinical Models. Mol. Cancer Ther. 2022, 21, 635–646. [Google Scholar] [CrossRef]
  196. Chaudhry, K.; Geiger, A.; Dowlati, E.; Lang, H.; Sohai, D.K.; Hwang, E.I.; Lazarski, C.A.; Yvon, E.; Holdhoff, M.; Jones, R.; et al. Co-transducing B7H3 CAR-NK cells with the DNR preserves their cytolytic function against GBM in the presence of exogenous TGF-β. Mol. Ther. Methods Clin. Dev. 2022, 27, 415–430. [Google Scholar] [CrossRef] [PubMed]
  197. Tang, X.; Wang, Y.; Huang, J.; Zhang, Z.; Liu, F.; Xu, J.; Guo, G.; Wang, W.; Tong, A.; Zhou, L. Administration of B7-H3 targeted chimeric antigen receptor-T cells induce regression of glioblastoma. Signal Transduct. Target. Ther. 2021, 6, 125. [Google Scholar] [CrossRef] [PubMed]
  198. Majzner, R.G.; Theruvath, J.L.; Nellan, A.; Heitzeneder, S.; Cui, Y.; Mount, C.W.; Rietberg, S.P.; Linde, M.H.; Xu, P.; Rota, C.; et al. CAR T Cells Targeting B7-H3, a Pan-Cancer Antigen, Demonstrate Potent Preclinical Activity Against Pediatric Solid Tumors and Brain Tumors. Clin. Cancer Res. 2019, 25, 2560–2574. [Google Scholar] [CrossRef] [PubMed]
  199. Li, S.; Zhang, M.; Wang, M.; Wang, H.; Wu, H.; Mao, L.; Zhang, M.; Li, H.; Zheng, J.; Ma, P.; et al. B7-H3 specific CAR-T cells exhibit potent activity against prostate cancer. Cell Death Discov. 2023, 9, 147. [Google Scholar] [CrossRef] [PubMed]
  200. Molhoek, K.R.; Erdag, G.; Rasamny, J.; Murphy, C.; Deacon, D.; Patterson, J.W.; Slingluff, C.L.; Brautigan, D.L. VEGFR-2 expression in human melanoma: Revised assessment. Int. J. Cancer 2011, 129, 2807–2815. [Google Scholar] [CrossRef] [PubMed]
  201. Kufe, D.W. MUC1-C oncoprotein as a target in breast cancer: Activation of signaling pathways and therapeutic approaches. Oncogene 2013, 32, 1073–1081. [Google Scholar] [CrossRef] [PubMed]
  202. Wei, J.; Sun, H.; Zhang, A.; Wu, X.; Li, Y.; Liu, J.; Duan, Y.; Xiao, F.; Wang, H.; Lv, M.; et al. A novel AXL chimeric antigen receptor endows T cells with anti-tumor effects against triple negative breast cancers. Cell Immunol. 2018, 331, 49–58. [Google Scholar] [CrossRef]
  203. DeRenzo, C. B7-H3-Specific Chimeric Antigen Receptor Autologous T-Cell Therapy for Pediatric Patients with Solid Tumors (3CAR). ClinicalTrials.gov identifier: NCT04897321. Updated 8 August 2023. Available online: https://clinicaltrials.gov/study/NCT04897321 (accessed on 10 October 2023).
  204. Yang, M.; Tang, X.; Zhang, Z.; Gu, L.; Wei, H.; Zhao, S.; Zhong, K.; Mu, M.; Huang, C.; Jiang, C.; et al. Tandem CAR-T cells targeting CD70 and B7-H3 exhibit potent preclinical activity against multiple solid tumors. Theranostics 2020, 10, 7622–7634. [Google Scholar] [CrossRef]
  205. Mishra, A.K.; Kemler, I.; Dingli, D. Preclinical development of CD126 CAR-T cells with broad antitumor activity. Blood Cancer J. 2021, 11, 3. [Google Scholar] [CrossRef]
  206. Rataj, F.; Jacobi, S.J.; Stoiber, S.; Asang, F.; Ogonek, J.; Tokarew, N.; Cadilha, B.L.; van Puijenbroek, E.; Heise, C.; Duewell, P.; et al. High-affinity CD16-polymorphism and Fc-engineered antibodies enable activity of CD16-chimeric antigen receptor-modified T cells for cancer therapy. Br. J. Cancer 2019, 120, 79–87. [Google Scholar] [CrossRef]
  207. Lu, Y.; Huang, H.; Chen, C.; Wang, H.; Wu, H.; Liu, X.; Zhao, W.; Gao, C.; Tan, S. Developing CD70-Targeted CAR-T Cells for the Treatment of Hematological and Solid Malignancies. Blood 2022, 140 (Suppl. S1), 12695. [Google Scholar] [CrossRef]
  208. Yang, J. Administering Peripheral Blood Lymphocytes Transduced with a CD70-Binding Chimeric Antigen Receptor to People with CD70 Expressing Cancers. ClinicalTrials.gov identifier: NCT02830724. Updated 1 September 2023. Available online: https://clinicaltrials.gov/study/NCT02830724 (accessed on 10 October 2023).
  209. Shah, P.D.; Huang, A.C.; Xu, X.; Orlowski, R.; Amaravadi, R.K.; Schuchter, L.M.; Zhang, P.; Tchou, J.; Matlawski, T.; Cervini, A.; et al. Phase I Trial of Autologous RNA-electroporated cMET-directed CAR T Cells Administered Intravenously in Patients with Melanoma and Breast Carcinoma. Cancer Res. Commun. 2023, 3, 821–829. [Google Scholar] [CrossRef] [PubMed]
  210. Kang, C.H.; Kim, Y.; Lee, D.Y.; Choi, S.U.; Lee, H.K.; Park, C.H. c-Met-Specific Chimeric Antigen Receptor T Cells Demonstrate Anti-Tumor Effect in c-Met Positive Gastric Cancer. Cancers 2021, 13, 5738. [Google Scholar] [CrossRef] [PubMed]
  211. Simon, B.; Harrer, D.C.; Schuler-Thurner, B.; Schuler, G.; Uslu, U. Arming T Cells with a gp100-Specific TCR and a CSPG4-Specific CAR Using Combined DNA- and RNA-Based Receptor Transfer. Cancers 2019, 11, 696. [Google Scholar] [CrossRef] [PubMed]
  212. Krug, C.; Birkholz, K.; Paulus, A.; Schwenkert, M.; Schmidt, P.; Hoffmann, N.; Hombach, A.; Fey, G.; Abken, H.; Schuler, G.; et al. Stability and activity of MCSP-specific chimeric antigen receptors (CARs) depend on the scFv antigen-binding domain and the protein backbone. Cancer Immunol. Immunother. 2015, 64, 1623–1635. [Google Scholar] [CrossRef]
  213. Wiesinger, M.; März, J.; Kummer, M.; Schuler, G.; Dörrie, J.; Schuler-Thurner, B.; Schaft, N. Clinical-Scale Production of CAR-T Cells for the Treatment of Melanoma Patients by mRNA Transfection of a CSPG4-Specific CAR under Full GMP Compliance. Cancers 2019, 11, 1198. [Google Scholar] [CrossRef] [PubMed]
  214. Fan, J.; Yu, Y.; Yan, L.; Yuan, Y.; Sun, B.; Yang, D.; Liu, N.; Guo, J.; Zhang, J.; Zhao, X. GAS6-based CAR-T cells exhibit potent antitumor activity against pancreatic cancer. J. Hematol. Oncol. 2023, 16, 77. [Google Scholar] [CrossRef]
  215. Gargett, T.; Yu, W.; Dotti, G.; Yvon, E.S.; Christo, S.N.; Hayball, J.D.; Lewis, I.D.; Brenner, M.K.; Brown, M.P. GD2-specific CAR T Cells Undergo Potent Activation and Deletion Following Antigen Encounter but can be Protected from Activation-induced Cell Death by PD-1 Blockade. Mol. Ther. 2016, 24, 1135–1149. [Google Scholar] [CrossRef]
  216. Kaplan, R. A Phase I Trial of T Cells Expressing an Anti-GD2 Chimeric Antigen Receptor in Children and Young Adults with GD2+ Solid Tumors. ClinicalTrials.gov identifier: NCT02107963. Updated 24 August 2023. Available online: https://clinicaltrials.gov/study/NCT02107963 (accessed on 10 October 2023).
  217. Omer, B. C7R-GD2.CART Cells for Patients With Relapsed or Refractory Neuroblastoma and Other GD2 Positive Cancers (GAIL-N). ClinicalTrial.gov identifier: NCT03635632. Updated 2 August 2023. Available online: https://clinicaltrials.gov/study/NCT03635632 (accessed on 10 October 2023).
  218. Lo, A.S.Y.; Ma, Q.; Liu, D.L.; Junghans, R.P. Anti-GD3 Chimeric sFv-CD28/T-Cell Receptor ζ Designer T Cells for Treatment of Metastatic Melanoma and Other Neuroectodermal Tumors. Clin. Cancer Res. 2010, 16, 2769–2780. [Google Scholar] [CrossRef]
  219. Yun, C.O.; Nolan, K.F.; Beecham, E.J.; Reisfeld, R.A.; Junghans, P. Targeting of T Lymphocytes to Melanoma Cells through Chimeric Anti-GD3 Immunoglobulin T-Cell Receptors. Neoplasia 2000, 2, 449–459. [Google Scholar] [CrossRef]
  220. Forsberg, E.M.; Lindberg, M.F.; Jespersen, H.; Alsén, S.; Bagge, R.O.; Donia, M.; Svane, I.M.; Nilsson, O.; Ny, L.; Nilsson, L.M.; et al. HER2 CAR-T Cells Eradicate Uveal Melanoma and T-cell Therapy–Resistant Human Melanoma in IL2 Transgenic NOD/SCID IL2 Receptor Knockout Mice. Cancer Res. 2019, 79, 899–904. [Google Scholar] [CrossRef] [PubMed]
  221. Park, J.; Talukder, A.H.; Lim, S.A.; Kim, K.; Pan, K.; Melendez, B.; Bradley, S.D.; Jackson, K.R.; Khalili, J.S.; Wang, J.; et al. SLC45A2: A Melanoma Antigen with High Tumor Selectivity and Reduced Potential for Autoimmune Toxicity. Cancer Immunol. Res. 2017, 5, 618–629. [Google Scholar] [CrossRef] [PubMed]
  222. Shivde, R.; Jaishankar, D.; Thomas, A.; Le Poole, I. 549 Design and in vitro efficacy of TRP-1 CAR T cells to target melanoma. J. Investig. Dermatol. 2021, 141, S95. [Google Scholar] [CrossRef]
  223. Lanitis, E.; Kosti, P.; Ronet, C.; Cribioli, E.; Rota, G.; Spill, A.; Reichenbach, P.; Zoete, V.; Laniti, D.D.; Coukos, G.; et al. VEGFR-2 redirected CAR-T cells are functionally impaired by soluble VEGF-A competition for receptor binding. J. Immunother. Cancer 2021, 9, e002151. [Google Scholar] [CrossRef]
  224. Cobb, D.A.; de Rossi, J.; Liu, L.; An, E.; Lee, D.W. Targeting of the alphavbeta3 integrin complex by CAR-T cells leads to rapid regression of diffuse intrinsic pontine glioma and glioblastoma. J. Immunother. Cancer 2022, 10, e003816. [Google Scholar] [CrossRef] [PubMed]
  225. Wallstabe, L.; Mades, A.; Frenz, S.; Einsele, H.; Rader, C.; Hudecek, M. CAR T cells targeting αvβ3 integrin are effective against advanced cancer in preclinical models. Adv. Cell Gene Ther. 2018, 1, e11. [Google Scholar] [CrossRef] [PubMed]
  226. Wang, X.; Lan, H.; Li, J.; Su, Y.; Xu, L. Muc1 promotes migration and lung metastasis of melanoma cells. Am. J. Cancer Res. 2015, 5, 2590–2604. [Google Scholar] [PubMed]
  227. Zhang, C.; Guo, C.; Li, Y.; Liu, K.; Zhao, Q.; Ouyang, L. Identification of Claudin-6 as a Molecular Biomarker in Pan-Cancer Through Multiple Omics Integrative Analysis. Front. Cell Dev. Biol. 2021, 9, 726656. [Google Scholar] [CrossRef]
  228. Carubia, J.M.; Yu, R.K.; Macala, L.J.; Kirkwood, J.M.; Varga, J.M. Gangliosides of normal and neoplastic human melanocytes. Biochem. Biophys. Res. Commun. 1984, 120, 500–504. [Google Scholar] [CrossRef]
  229. Hamamura, K.; Furukawa, K.; Hayashi, T.; Hattori, T.; Nakano, J.; Nakashima, H.; Okuda, T.; Mizutani, H.; Hattori, H.; Ueda, M.; et al. Ganglioside GD3 promotes cell growth and invasion through p130Cas and paxillin in malignant melanoma cells. Proc. Natl. Acad. Sci. USA 2005, 102, 11041–11046. [Google Scholar] [CrossRef]
  230. Ohkawa, Y.; Miyazaki, S.; Hamamura, K.; Kambe, M.; Miyata, M.; Tajima, O.; Ohmi, Y.; Yamauchi, Y.; Furukawa, K.; Furukawa, K. Ganglioside GD3 Enhances Adhesion Signals and Augments Malignant Properties of Melanoma Cells by Recruiting Integrins to Glycolipid-enriched Microdomains. J. Biol. Chem. 2010, 285, 27213–27223. [Google Scholar] [CrossRef] [PubMed]
  231. Ramos, R.I.; Bustos, M.A.; Wu, J.; Jones, P.; Chang, S.C.; Kiyohara, E.; Tran, K.; Zhang, X.; Stern, S.L.; Izraely, S.; et al. Upregulation of cell surface GD3 ganglioside phenotype is associated with human melanoma brain metastasis. Mol. Oncol. 2020, 14, 1760–1778. [Google Scholar] [CrossRef] [PubMed]
  232. Huang, R.; Rofstad, E.K. Integrins as therapeutic targets in the organ-specific metastasis of human malignant melanoma. J. Exp. Clin. Cancer Res. 2018, 37, 92. [Google Scholar] [CrossRef] [PubMed]
  233. Seftor, R.E.; Seftor, E.A.; Gehlsen, K.R.; Stetler-Stevenson, W.G.; Brown, P.D.; Ruoslahti, E.; Hendrix, M.J. Role of the alpha v beta 3 integrin in human melanoma cell invasion. Proc. Natl. Acad. Sci. USA 1992, 89, 1557–1561. [Google Scholar] [CrossRef] [PubMed]
  234. Pocheć, E.; Bubka, M.; Rydlewska, M.; Janik, M.; Pokrywka, M.; Lityńska, A. Aberrant glycosylation of αvβ3 integrin is associated with melanoma progression. Anticancer Res. 2015, 35, 2093–2103. [Google Scholar] [PubMed]
  235. Scheller, J.; Chalaris, A.; Schmidt-Arras, D.; Rose-John, S. The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim. Biophys. Acta Mol. Cell Res. 2011, 1813, 878–888. [Google Scholar] [CrossRef] [PubMed]
  236. Zuo, Q.; Liu, J.; Huang, L.; Qin, Y.; Hawley, T.; Seo, C.; Merlino, G.; Yu, Y. AXL/AKT axis mediated-resistance to BRAF inhibitor depends on PTEN status in melanoma. Oncogene 2018, 37, 3275–3289. [Google Scholar] [CrossRef]
  237. Tworkoski, K.A.; Platt, J.T.; Bacchiocchi, A.; Bosenberg, M.; Boggon, T.J.; Stern, D.F. MERTK controls melanoma cell migration and survival and differentially regulates cell behavior relative to AXL. Pigment. Cell Melanoma Res. 2013, 26, 527–541. [Google Scholar] [CrossRef]
  238. Tizpa, E.; Young, H.J.; Bonjoc, K.-J.C.; Chang, C.-W.; Liu, Y.; Foulks, J.M.; Chaudhry, A. Role of AXL in metastatic melanoma and impact of TP-0903 as a novel therapeutic option for melanoma brain metastasis. J. Clin. Oncol. 2020, 38 (Suppl. S15), e22021. [Google Scholar] [CrossRef]
  239. Boshuizen, J.; Koopman, L.A.; Krijgsman, O.; Shahrabi, A.; van den Heuvel, E.G.; A Ligtenberg, M.A.; Vredevoogd, D.W.; Kemper, K.; Kuilman, T.; Song, J.-Y.; et al. Cooperative targeting of melanoma heterogeneity with an AXL antibody-drug conjugate and BRAF/MEK inhibitors. Nat. Med. 2018, 24, 203–212. [Google Scholar] [CrossRef]
  240. Nyakas, M.; Fleten, K.G.; Haugen, M.H.; Engedal, N.; Sveen, C.; Farstad, I.N.; Flørenes, V.A.; Prasmickaite, L.; Mælandsmo, G.M.; Seip, K. AXL inhibition improves BRAF-targeted treatment in melanoma. Sci. Rep. 2022, 12, 5076. [Google Scholar] [CrossRef]
  241. Nicolè, L.; Cappello, F.; Cappellesso, R.; Piccin, L.; Ventura, L.; Guzzardo, V.; Del Fiore, P.; Chiarion-Sileni, V.; Tos, A.P.D.; Mocellin, S.; et al. RIPK3 and AXL Expression Study in Primary Cutaneous Melanoma Unmasks AXL as Predictor of Sentinel Node Metastasis: A Pilot Study. Front. Oncol. 2021, 11, 728319. [Google Scholar] [CrossRef]
  242. Shao, H.; Teramae, D.; Wells, A. Axl contributes to efficient migration and invasion of melanoma cells. PLoS ONE 2023, 18, e0283749. [Google Scholar] [CrossRef] [PubMed]
  243. Xue, G.; Kohler, R.; Tang, F.; Hynx, D.; Wang, Y.; Orso, F.; Prêtre, V.; Ritschard, R.; Hirschmann, P.; Cron, P.; et al. mTORC1/autophagy-regulated MerTK in mutant BRAFV600 melanoma with acquired resistance to BRAF inhibition. Oncotarget 2017, 8, 69204–69218. [Google Scholar] [CrossRef] [PubMed]
  244. Rodriguez-Garcia, A.; Lynn, R.C.; Poussin, M.; Eiva, M.A.; Shaw, L.C.; O’connor, R.S.; Minutolo, N.G.; Casado-Medrano, V.; Lopez, G.; Matsuyama, T.; et al. CAR-T cell-mediated depletion of immunosuppressive tumor-associated macrophages promotes endogenous antitumor immunity and augments adoptive immunotherapy. Nat. Commun. 2021, 12, 877. [Google Scholar] [CrossRef]
  245. Schlegel, J.; Sambade, M.J.; Sather, S.; Moschos, S.J.; Tan, A.-C.; Winges, A.; DeRyckere, D.; Carson, C.C.; Trembath, D.G.; Tentler, J.J.; et al. MERTK receptor tyrosine kinase is a therapeutic target in melanoma. J. Clin. Investig. 2013, 123, 2257–2267. [Google Scholar] [CrossRef] [PubMed]
  246. Sinik, L.; Minson, K.A.; Tentler, J.J.; Carrico, J.; Bagby, S.M.; Robinson, W.A.; Kami, R.; Burstyn-Cohen, T.; Eckhardt, S.G.; Wang, X.; et al. Inhibition of MERTK Promotes Suppression of Tumor Growth in BRAF Mutant and BRAF Wild-Type Melanoma. Mol. Cancer Ther. 2019, 18, 278–288. [Google Scholar] [CrossRef] [PubMed]
  247. Boshuizen, J.; Vredevoogd, D.W.; Krijgsman, O.; Ligtenberg, M.A.; Blankenstein, S.; de Bruijn, B.; Frederick, D.T.; Kenski, J.C.N.; Parren, M.; Brüggemann, M.; et al. Reversal of pre-existing NGFR-driven tumor and immune therapy resistance. Nat. Commun. 2020, 11, 3946. [Google Scholar] [CrossRef]
  248. 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]
  249. Radke, J.; Roßner, F.; Redmer, T. CD271 determines migratory properties of melanoma cells. Sci. Rep. 2017, 7, 9834. [Google Scholar] [CrossRef]
  250. Furuta, J.; Inozume, T.; Harada, K.; Shimada, S. CD271 on Melanoma Cell Is an IFN-γ-Inducible Immunosuppressive Factor that Mediates Downregulation of Melanoma Antigens. J. Investig. Dermatol. 2014, 134, 1369–1377. [Google Scholar] [CrossRef] [PubMed]
  251. Lehmann, J.; Caduff, N.; Krzywińska, E.; Stierli, S.; Salas-Bastos, A.; Loos, B.; Levesque, M.P.; Dummer, R.; Stockmann, C.; Münz, C.; et al. Escape from NK cell tumor surveillance by NGFR-induced lipid remodeling in melanoma. Sci. Adv. 2023, 9, eadc8825. [Google Scholar] [CrossRef] [PubMed]
  252. Teppert, K.; Wang, X.; Anders, K.; Evaristo, C.; Lock, D.; Künkele, A. Joining Forces for Cancer Treatment: From “TCR versus CAR” to “TCR and CAR. ” Int. J. Mol. Sci. 2022, 23, 14563. [Google Scholar] [CrossRef] [PubMed]
  253. Khawar, M.B.; Sun, H. CAR-NK Cells: From Natural Basis to Design for Kill. Front. Immunol. 2021, 12, 707542. [Google Scholar] [CrossRef] [PubMed]
  254. Oberschmidt, O.; Kloess, S.; Koehl, U. Redirected Primary Human Chimeric Antigen Receptor Natural Killer Cells as an “Off-the-Shelf Immunotherapy” for Improvement in Cancer Treatment. Front. Immunol. 2017, 8, 654. [Google Scholar] [CrossRef] [PubMed]
  255. Ruppel, K.E.; Fricke, S.; Köhl, U.; Schmiedel, D. Taking Lessons from CAR-T Cells and Going Beyond: Tailoring Design and Signaling for CAR-NK Cells in Cancer Therapy. Front. Immunol. 2022, 13, 822298. [Google Scholar] [CrossRef] [PubMed]
  256. Zenere, G.; Olwenyi, O.A.; Byrareddy, S.N.; Braun, S.E. Optimizing intracellular signaling domains for CAR NK cells in HIV immunotherapy: A comprehensive review. Drug Discov. Today 2019, 24, 983–991. [Google Scholar] [CrossRef]
  257. Voynova, E.; Hawk, N.; Flomerfelt, F.A.; Telford, W.G.; Gress, R.E.; Kanakry, J.A.; Kovalovsky, D. Increased Activity of a NK-Specific CAR-NK Framework Targeting CD3 and CD5 for T-Cell Leukemias. Cancers 2022, 14, 524. [Google Scholar] [CrossRef]
  258. Lee, Y.E.; Ju, A.; Choi, H.W.; Kim, J.-C.; Kim, E.E.; Kim, T.S.; Kang, H.J.; Kim, S.-Y.; Jang, J.-Y.; Ku, J.-L.; et al. Rationally designed redirection of natural killer cells anchoring a cytotoxic ligand for pancreatic cancer treatment. J. Control. Release 2020, 326, 310–323. [Google Scholar] [CrossRef]
  259. Li, Y.; Hermanson, D.L.; Moriarity, B.S.; Kaufman, D.S. Human iPSC-Derived Natural Killer Cells Engineered with Chimeric Antigen Receptors Enhance Anti-Tumor Activity. Cell Stem Cell 2018, 23, 181–192.e5. [Google Scholar] [CrossRef]
  260. Blake, S.J.; Dougall, W.C.; Miles, J.J.; Teng, M.W.; Smyth, M.J. Molecular Pathways: Targeting CD96 and TIGIT for Cancer Immunotherapy. Clin. Cancer Res. 2016, 22, 5183–5188. [Google Scholar] [CrossRef] [PubMed]
  261. Jan, C.I.; Huang, S.W.; Canoll, P.; Bruce, J.N.; Lin, Y.C.; Pan, C.M.; Lu, H.M.; Chiu, S.C.; Cho, D.Y. Targeting human leukocyte antigen G with chimeric antigen receptors of natural killer cells convert immunosuppression to ablate solid tumors. J. Immunother. Cancer 2021, 9, e003050. [Google Scholar] [CrossRef] [PubMed]
  262. Xu, X.; Li, H.; Xu, C. Structural understanding of T cell receptor triggering. Cell Mol. Immunol. 2020, 17, 193–202. [Google Scholar] [CrossRef]
  263. Ferrara, J.; Reddy, P.; Paczesny, S.; Reimann, C.; Cortivo, L.D.; Hacein-Bey-Abina, S.; Fischer, A.; André-Schmutz, I.; Cavazzana-Calvo, M.; Ribas, A.; et al. Immunotherapy through T-cell receptor gene transfer induces severe graft-versus-host disease. Immunotherapy 2010, 2, 791–794. [Google Scholar] [CrossRef] [PubMed]
  264. Bijen, H.M.; van der Steen, D.M.; Hagedoorn, R.S.; Wouters, A.K.; Wooldridge, L.; Falkenburg, J.F.; Heemskerk, M.H. Preclinical Strategies to Identify Off-Target Toxicity of High-Affinity TCRs. Mol. Ther. 2018, 26, 1206–1214. [Google Scholar] [CrossRef] [PubMed]
  265. Parlar, A.; Sayitoglu, E.C.; Ozkazanc, D.; Georgoudaki, A.M.; Pamukcu, C.; Aras, M.; Josey, B.J.; Chrobok, M.; Branecki, S.; Zahedimaram, P.; et al. Engineering antigen-specific NK cell lines against the melanoma-associated antigen tyrosinase via TCR gene transfer. Eur. J. Immunol. 2019, 49, 1278–1290. [Google Scholar] [CrossRef] [PubMed]
  266. Walseng, E.; Köksal, H.; Sektioglu, I.M.; Fåne, A.; Skorstad, G.; Kvalheim, G.; Gaudernack, G.; Inderberg, E.M.; Wälchli, S. A TCR-based Chimeric Antigen Receptor. Sci. Rep. 2017, 7, 10713. [Google Scholar] [CrossRef] [PubMed]
  267. Wellbrock, C.; Arozarena, I. The Complexity of the ERK/MAP-Kinase Pathway and the Treatment of Melanoma Skin Cancer. Front. Cell Dev. Biol. 2016, 4, 33. [Google Scholar] [CrossRef]
  268. Rauch, N.; Rukhlenko, O.S.; Kolch, W.; Kholodenko, B.N. MAPK kinase signalling dynamics regulate cell fate decisions and drug resistance. Curr. Opin. Struct. Biol. 2016, 41, 151–158. [Google Scholar] [CrossRef]
  269. Kunz, M.; Vera, J. Modelling of Protein Kinase Signaling Pathways in Melanoma and Other Cancers. Cancers 2019, 11, 465. [Google Scholar] [CrossRef]
  270. Guo, W.; Wang, H.; Li, C. Signal pathways of melanoma and targeted therapy. Signal Transduct. Target. Ther. 2021, 6, 424. [Google Scholar] [CrossRef]
  271. Wang, Y.; Poulin, E.J.; Coffey, R.J. LRIG1 is a triple threat: ERBB negative regulator, intestinal stem cell marker and tumour suppressor. Br. J. Cancer 2013, 108, 1765–1770. [Google Scholar] [CrossRef]
  272. Johansson, M.; Oudin, A.; Tiemann, K.; Bernard, A.; Golebiewska, A.; Keunen, O.; Fack, F.; Stieber, D.; Wang, B.; Hedman, H.; et al. The soluble form of the tumor suppressor Lrig1 potently inhibits in vivo glioma growth irrespective of EGF receptor status. Neuro Oncol. 2013, 15, 1200–1211. [Google Scholar] [CrossRef]
  273. Billing, O.; Holmgren, Y.; Nosek, D.; Hedman, H.; Hemmingsson, O. LRIG1 is a conserved EGFR regulator involved in melanoma development, survival and treatment resistance. Oncogene 2021, 40, 3707–3718. [Google Scholar] [CrossRef]
  274. Wu, C.H.; Liu, I.J.; Lu, R.M.; Wu, H.C. Advancement and applications of peptide phage display technology in biomedical science. J. Biomed. Sci. 2016, 23, 8. [Google Scholar] [CrossRef]
  275. Garner, H.; de Visser, K.E. Immune crosstalk in cancer progression and metastatic spread: A complex conversation. Nat. Rev. Immunol. 2020, 20, 483–497. [Google Scholar] [CrossRef]
  276. Kim, J.; Kim, J.S.; Lee, H.K.; Kim, H.S.; Park, E.J.; Choi, J.E.; Choi, Y.J.; Shin, B.R.; Kim, E.Y.; Hong, J.T.; et al. CXCR3-deficient natural killer cells fail to migrate to B16F10 melanoma cells. Int. Immunopharmacol. 2018, 63, 66–73. [Google Scholar] [CrossRef] [PubMed]
  277. Mgrditchian, T.; Arakelian, T.; Paggetti, J.; Noman, M.Z.; Viry, E.; Moussay, E.; Van Moer, K.; Kreis, S.; Guerin, C.; Buart, S.; et al. Targeting autophagy inhibits melanoma growth by enhancing NK cells infiltration in a CCL5-dependent manner. Proc. Natl. Acad. Sci. USA 2017, 114, E9271–E9279. [Google Scholar] [CrossRef]
  278. Jobe, N.P.; Rösel, D.; Dvořánková, B.; Kodet, O.; Lacina, L.; Mateu, R.; Smetana, K.; Brábek, J. Simultaneous blocking of IL-6 and IL-8 is sufficient to fully inhibit CAF-induced human melanoma cell invasiveness. Histochem. Cell Biol. 2016, 146, 205–217. [Google Scholar] [CrossRef] [PubMed]
  279. Straussman, R.; Morikawa, T.; Shee, K.; Barzily-Rokni, M.; Qian, Z.R.; Du, J.; Davis, A.; Mongare, M.M.; Gould, J.; Frederick, D.T.; et al. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature 2012, 487, 500–504. [Google Scholar] [CrossRef] [PubMed]
  280. Liu, T.; Zhou, L.; Xiao, Y.; Andl, T.; Zhang, Y. BRAF Inhibitors Reprogram Cancer-Associated Fibroblasts to Drive Matrix Remodeling and Therapeutic Escape in Melanoma. Cancer Res. 2022, 82, 419–432. [Google Scholar] [CrossRef] [PubMed]
  281. Morales, D.; Vigneron, P.; Ferreira, I.; Hamitou, W.; Magnano, M.; Mahenthiran, L.; Lok, C.; Vayssade, M. Fibroblasts Influence Metastatic Melanoma Cell Sensitivity to Combined BRAF and MEK Inhibition. Cancers 2021, 13, 4761. [Google Scholar] [CrossRef]
  282. Zhou, L.; Yang, K.; Wickett, R.R.; Kadekaro, A.L.; Zhang, Y. Targeted deactivation of cancer-associated fibroblasts by β-catenin ablation suppresses melanoma growth. Tumor Biol. 2016, 37, 14235–14248. [Google Scholar] [CrossRef]
  283. Balsamo, M.; Scordamaglia, F.; Pietra, G.; Manzini, C.; Cantoni, C.; Boitano, M.; Queirolo, P.; Vermi, W.; Facchetti, F.; Moretta, A.; et al. Melanoma-associated fibroblasts modulate NK cell phenotype and antitumor cytotoxicity. Proc. Natl. Acad. Sci. USA 2009, 106, 20847–20852. [Google Scholar] [CrossRef] [PubMed]
  284. Kahounová, Z.; Kurfürstová, D.; Bouchal, J.; Kharaishvili, G.; Navrátil, J.; Remšík, J.; Šimečková, K.; Študent, V.; Kozubík, A.; Souček, K. The fibroblast surface markers FAP, anti-fibroblast, and FSP are expressed by cells of epithelial origin and may be altered during epithelial-to-mesenchymal transition. Cytom. Part A 2018, 93, 941–951. [Google Scholar] [CrossRef] [PubMed]
  285. Vázquez-Villa, F.; García-Ocaña, M.; Galván, J.A.; García-Martínez, J.; García-Pravia, C.; Menéndez-Rodríguez, P.; González-del Rey, C.; Barneo-Serra, L.; de los Toyos, J.R. COL11A1/(pro)collagen 11A1 expression is a remarkable biomarker of human invasive carcinoma-associated stromal cells and carcinoma progression. Tumor Biol. 2015, 36, 2213–2222. [Google Scholar] [CrossRef] [PubMed]
  286. Mezawa, Y.; Orimo, A. The roles of tumor- and metastasis-promoting carcinoma-associated fibroblasts in human carcinomas. Cell Tissue Res. 2016, 365, 675–689. [Google Scholar] [CrossRef]
  287. Gunaydin, G. CAFs Interacting With TAMs in Tumor Microenvironment to Enhance Tumorigenesis and Immune Evasion. Front. Oncol. 2021, 11, 668349. [Google Scholar] [CrossRef]
  288. Chen, P.; Huang, Y.; Bong, R.; Ding, Y.; Song, N.; Wang, X.; Song, X.; Luo, Y. Tumor-Associated Macrophages Promote Angiogenesis and Melanoma Growth via Adrenomedullin in a Paracrine and Autocrine Manner. Clin. Cancer Res. 2011, 17, 7230–7239. [Google Scholar] [CrossRef]
  289. Filippi, L.; Bruno, G.; Domazetovic, V.; Favre, C.; Calvani, M. Current Therapies and New Targets to Fight Melanoma: A Promising Role for the β3-Adrenoreceptor. Cancers 2020, 12, 1415. [Google Scholar] [CrossRef]
  290. Zhang, P.; Zhao, S.; Wu, C.; Li, J.; Li, Z.; Wen, C.; Hu, S.; An, G.; Meng, H.; Zhang, X.; et al. Effects of CSF1R-targeted chimeric antigen receptor-modified NK92MI & T cells on tumor-associated macrophages. Immunotherapy 2018, 10, 935–949. [Google Scholar] [CrossRef]
  291. Wang, L.; He, T.; Liu, J.; Tai, J.; Wang, B.; Chen, Z.; Quan, Z. Pan-cancer analysis reveals tumor-associated macrophage communication in the tumor microenvironment. Exp. Hematol. Oncol. 2021, 10, 31. [Google Scholar] [CrossRef] [PubMed]
  292. Dixon, M.L.; Luo, L.; Ghosh, S.; Grimes, J.M.; Leavenworth, J.D.; Leavenworth, J.W. Remodeling of the tumor microenvironment via disrupting Blimp1+ effector Treg activity augments response to anti-PD-1 blockade. Mol. Cancer 2021, 20, 150. [Google Scholar] [CrossRef] [PubMed]
  293. Salmi, S.; Lin, A.; Hirschovits-Gerz, B.; Valkonen, M.; Aaltonen, N.; Sironen, R.; Siiskonen, H.; Pasonen-Seppänen, S. The role of FoxP3+ regulatory T cells and IDO+ immune and tumor cells in malignant melanoma—An immunohistochemical study. BMC Cancer 2021, 21, 641. [Google Scholar] [CrossRef]
  294. Watson, M.J.; Vignali, P.D.A.; Mullett, S.J.; Overacre-Delgoffe, A.E.; Peralta, R.M.; Grebinoski, S.; Menk, A.V.; Rittenhouse, N.L.; DePeaux, K.; Whetstone, R.D.; et al. Metabolic support of tumour-infiltrating regulatory T cells by lactic acid. Nature 2021, 591, 645–651. [Google Scholar] [CrossRef] [PubMed]
  295. Lainé, A.; Labiad, O.; Hernandez-Vargas, H.; This, S.; Sanlaville, A.; Léon, S.; Dalle, S.; Sheppard, D.; Travis, M.A.; Paidassi, H.; et al. Regulatory T cells promote cancer immune-escape through integrin αvβ8-mediated TGF-β activation. Nat. Commun. 2021, 12, 6228. [Google Scholar] [CrossRef] [PubMed]
  296. Li, C.; Jiang, P.; Wei, S.; Xu, X.; Wang, J. Regulatory T cells in tumor microenvironment: New mechanisms, potential therapeutic strategies and future prospects. Mol. Cancer 2020, 19, 116. [Google Scholar] [CrossRef] [PubMed]
  297. Dehbashi, M.; Hojati, Z.; Motovali-Bashi, M.; Ganjalikhany, M.R.; Cho, W.C.; Shimosaka, A.; Navabi, P.; Ganjalikhani-Hakemi, M. A Novel CAR Expressing NK Cell Targeting CD25 with the Prospect of Overcoming Immune Escape Mechanism in Cancers. Front. Oncol. 2021, 11, 649710. [Google Scholar] [CrossRef]
  298. Kageshita, T.; Kashio, Y.; Yamauchi, A.; Seki, M.; Abedin, M.J.; Nishi, N.; Shoji, H.; Nakamura, T.; Ono, T.; Hirashima, M. Possible role of galectin-9 in cell aggregation and apoptosis of human melanoma cell lines and its clinical significance. Int. J. Cancer 2002, 99, 809–816. [Google Scholar] [CrossRef]
  299. Francisco, L.M.; Salinas, V.H.; Brown, K.E.; Vanguri, V.K.; Freeman, G.J.; Kuchroo, V.K.; Sharpe, A.H. PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. J. Exp. Med. 2009, 206, 3015–3029. [Google Scholar] [CrossRef]
  300. Unger, W.W.J.; Laban, S.; Kleijwegt, F.S.; van der Slik, A.R.; Roep, B.O. Induction of Treg by monocyte-derived DC modulated by vitamin D3 or dexamethasone: Differential role for PD-L1. Eur. J. Immunol. 2009, 39, 3147–3159. [Google Scholar] [CrossRef] [PubMed]
  301. Kararoudi, M.N.; Likhite, S.; Elmas, E.; Yamamoto, K.; Schwartz, M.; Sorathia, K.; Pereira, M.d.S.F.; Sezgin, Y.; Devine, R.D.; Lyberger, J.M.; et al. Optimization and validation of CAR transduction into human primary NK cells using CRISPR and AAV. Cell Rep. Methods 2022, 2, 100236. [Google Scholar] [CrossRef] [PubMed]
  302. de Andrade, L.F.; Ngiow, S.F.; Stannard, K.; Rusakiewicz, S.; Kalimutho, M.; Khanna, K.K.; Tey, S.-K.; Takeda, K.; Zitvogel, L.; Martinet, L.; et al. Natural Killer Cells Are Essential for the Ability of BRAF Inhibitors to Control BRAFV600E-Mutant Metastatic Melanoma. Cancer Res. 2014, 74, 7298–7308. [Google Scholar] [CrossRef] [PubMed]
  303. Frazao, A.; Rethacker, L.; Jeudy, G.; Colombo, M.; Pasmant, E.; Avril, M.-F.; Toubert, A.; Moins-Teisserenc, H.; Roelens, M.; Dalac, S.; et al. BRAF inhibitor resistance of melanoma cells triggers increased susceptibility to natural killer cell-mediated lysis. J. Immunother. Cancer 2020, 8, e000275. [Google Scholar] [CrossRef]
  304. Sottile, R.; Pangigadde, P.N.; Tan, T.; Anichini, A.; Sabbatino, F.; Trecroci, F.; Favoino, E.; Orgiano, L.; Roberts, J.; Ferrone, S.; et al. HLA class I downregulation is associated with enhanced NK-cell killing of melanoma cells with acquired drug resistance to BRAF inhibitors. Eur. J. Immunol. 2016, 46, 409–419. [Google Scholar] [CrossRef]
Figure 1. Natural killer cell signaling receptor repertoire. Natural killer cells are equipped with a diverse assortment of activating and inhibitory signaling receptors that mediate NK cell activity through combined signal propagation. Activating receptors, including NCRs, NKG2D, DNAM-1, CD16, and aKIRs transmit the signals to promote NK cell activation and cytotoxicity, while inhibitory receptors, including CD96, TIGIT, LIR1 and iKIRs, induce the inhibition signaling to block the NK cell activation. KIR2DL4 could introduce activating or inhibitory signaling into NK cells based on its content and ligands.
Figure 1. Natural killer cell signaling receptor repertoire. Natural killer cells are equipped with a diverse assortment of activating and inhibitory signaling receptors that mediate NK cell activity through combined signal propagation. Activating receptors, including NCRs, NKG2D, DNAM-1, CD16, and aKIRs transmit the signals to promote NK cell activation and cytotoxicity, while inhibitory receptors, including CD96, TIGIT, LIR1 and iKIRs, induce the inhibition signaling to block the NK cell activation. KIR2DL4 could introduce activating or inhibitory signaling into NK cells based on its content and ligands.
Cells 12 02750 g001
Figure 2. Functional shifts between anti-tumor immunity and immunosuppression within the tumor microenvironment. Several non-malignant immune and stromal cells undergo phenotypic alterations through direct cell-cell interactions with melanoma cells and through the influence of chemical conditions within the tumor microenvironment. M1: phagocytic macrophage phenotype. M2: pro-inflammatory macrophage phenotype. N1: cytotoxic neutrophil phenotype. N2: pro-inflammatory neutrophil phenotype. APC-DC: functional antigen-presenting dendritic cell. Tol-DC: dysfunctional tolerogenic dendritic cell. Teff: CD8+ cytotoxic T cell. Th: CD4+ pro-cytotoxic helper T cell. Treg: CD4+ immunosuppressive regulatory T cell. NK: natural killer cell. P-Fib: primary fibroblast. CAF: cancer-associated fibroblast.
Figure 2. Functional shifts between anti-tumor immunity and immunosuppression within the tumor microenvironment. Several non-malignant immune and stromal cells undergo phenotypic alterations through direct cell-cell interactions with melanoma cells and through the influence of chemical conditions within the tumor microenvironment. M1: phagocytic macrophage phenotype. M2: pro-inflammatory macrophage phenotype. N1: cytotoxic neutrophil phenotype. N2: pro-inflammatory neutrophil phenotype. APC-DC: functional antigen-presenting dendritic cell. Tol-DC: dysfunctional tolerogenic dendritic cell. Teff: CD8+ cytotoxic T cell. Th: CD4+ pro-cytotoxic helper T cell. Treg: CD4+ immunosuppressive regulatory T cell. NK: natural killer cell. P-Fib: primary fibroblast. CAF: cancer-associated fibroblast.
Cells 12 02750 g002
Figure 3. The four main mechanisms of immune escape. (A) Disruption of NK cell activation through at least three mechanisms: Tumor cells can express molecules such as IDO1 and PGE2 to mediate the inhibition of NKAR expression in NK cells. Also, tumor cells often downregulate the NKAR ligand expression on tumor cell surfaces or/and shed their NKAR ligands proteolytically. (B) Promotion of NK-inhibiting receptor-ligand interactions via upregulation of inhibitory ligand expression on melanoma cells. (C) Immune escape assisted by non-malignant constituent cells within the tumor microenvironment through secreted immunosuppressive cytokines and chemokines. (D) Immunological dysfunction resulting from hypoxic and acidic chemical conditions within the tumor microenvironment.
Figure 3. The four main mechanisms of immune escape. (A) Disruption of NK cell activation through at least three mechanisms: Tumor cells can express molecules such as IDO1 and PGE2 to mediate the inhibition of NKAR expression in NK cells. Also, tumor cells often downregulate the NKAR ligand expression on tumor cell surfaces or/and shed their NKAR ligands proteolytically. (B) Promotion of NK-inhibiting receptor-ligand interactions via upregulation of inhibitory ligand expression on melanoma cells. (C) Immune escape assisted by non-malignant constituent cells within the tumor microenvironment through secreted immunosuppressive cytokines and chemokines. (D) Immunological dysfunction resulting from hypoxic and acidic chemical conditions within the tumor microenvironment.
Cells 12 02750 g003
Figure 4. A diagram of future CAR-NK cell development for treatment of melanoma. These studies will focus strengthening CAR-NK cytotoxic activation for maximized to killing the metastatic and drug resistant melanoma, including structural optimization of CAR with NKG2D or DNAM1 transmembrane domain and 2B4 or DAP10 intracellular signaling domain, integration of TCR structural elements in CAR-NK cells, targeting receptor tyrosine kinase signaling with sLRIG1, integration of CXCR3 to enhance CAR-NK cells infiltration, targeting markers of non-malignant cells in TME, corporation of blocking NK cell inhibitory receptors, as well as optimization with drug therapies.
Figure 4. A diagram of future CAR-NK cell development for treatment of melanoma. These studies will focus strengthening CAR-NK cytotoxic activation for maximized to killing the metastatic and drug resistant melanoma, including structural optimization of CAR with NKG2D or DNAM1 transmembrane domain and 2B4 or DAP10 intracellular signaling domain, integration of TCR structural elements in CAR-NK cells, targeting receptor tyrosine kinase signaling with sLRIG1, integration of CXCR3 to enhance CAR-NK cells infiltration, targeting markers of non-malignant cells in TME, corporation of blocking NK cell inhibitory receptors, as well as optimization with drug therapies.
Cells 12 02750 g004
Table 1. Current pre-clinical and clinical CAR-NK cell studies relevant to melanoma.
Table 1. Current pre-clinical and clinical CAR-NK cell studies relevant to melanoma.
TargetStudy TypeClinical Trial StatusCancer TypeReference
5T4ClinicalActive/RecruitingSolid tumorsNCT05194709
B7-H3Preclinical-Non-small cell lung cancer[161]
B7-H6Preclinical-Solid tumors[162]
CD24Preclinical-Ovarian cancer[163]
CD276Preclinical-Solid tumors[164]
CD44Preclinical-Ovarian cancer[165]
CLDN6ClinicalActive/RecruitingSolid tumors[166]
c-MetPreclinical-Lung adenocarcinoma[160]
EGFR/EGFRvIIIPreclinical-Glioblastoma[167]
-Glioblastoma[168]
-Glioblastoma[169]
-Glioblastoma[170]
EpCAMPreclinical-Solid tumors[171]
GD2Preclinical-GD2+ solid tumors[172]
-GD2+ solid tumors[173]
-Ewing sarcoma[174]
gp100Preclinical-melanoma[175]
HER2Preclinical-Rhabdomyosarcoma[146]
-Glioblastoma[176]
-Solid tumors[177]
-Rhabdomyosarcoma[178]
MesothelinPreclinical-Gastric cancer[158]
MUC1ClinicalCompletedSolid tumors[179]
NKG2DLClinicalActive/RecruitingSolid tumorsNCT05528341
RecruitingSolid tumorsNCT03415100
ROBO1ClinicalUnknownSolid tumorsNCT03940820
VEGFR2Preclinical-Solid tumors[180]
Table 2. Potential targets for future CAR-NK cell immunotherapies for melanoma based on CAR-T cell preclinical and clinical studies.
Table 2. Potential targets for future CAR-NK cell immunotherapies for melanoma based on CAR-T cell preclinical and clinical studies.
TargetStudy TypeClinical Trial StatusCancer TypeReference
AXLPreclinical-Triple-negative breast cancers[202]
B7-H3ClinicalActive, Not RecruitingSolid tumorsNCT04483778
RecruitingSolid tumorsNCT04897321
RecruitingSolid tumorsNCT05190185
Preclinical-Solid tumors[194]
-Glioblastoma[197]
-Solid tumors[198]
-Prostate cancer[199]
-Solid tumors[203]
B7-H3/CD70Preclinical-Solid tumors[204]
CD126Preclinical-Solid tumors[205]
CD16Preclinical-Solid tumors[206]
CD70ClinicalRecruitingSolid tumorsNCT02830724
Preclinical-CD70+ cancers[207]
-CD70+ cancers[208]
c-MetClinicalCompletedMelanoma, breast carcinoma[209]
Preclinical-Gastric cancer[210]
CSPG4Preclinical-Solid tumors[211]
-Solid tumors[212]
-Melanoma[213]
GAS6Preclinical-Pancreatic cancer[214]
GD2ClinicalCompletedMelanoma[186,215]
CompletedSolid tumors[216]
CompletedNeuroblastoma[217]
RecruitingSolid tumorsNCT03635632
CompletedSolid tumorsNCT02107963
Preclinical-Lung cancer[185]
-Solid tumors[189]
-Solid tumors[215]
GD3Preclinical-Melanoma[218]
-Melanoma[219]
HER2Preclinical-HER2+ sarcomas[29]
-HER2+ melanoma subtypes[220]
IL13Ra2ClinicalRecruitingSolid tumorsNCT04119024
Multiple targets (NY-ESO-1, DR5, EGFRvIII, c-Met)ClinicalUnknownSolid tumorsNCT03638206
SLC45A2Preclinical-Melanoma[221]
TRP-1Preclinical-Melanoma[222]
VEGFR2ClinicalTerminatedSolid tumorsNCT01218867
Preclinical-Solid tumors[180]
-Solid tumors[223]
αvβ3Preclinical-Glioma and glioblastoma[224]
-Advanced cancers[225]
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

Hibler, W.; Merlino, G.; Yu, Y. CAR NK Cell Therapy for the Treatment of Metastatic Melanoma: Potential & Prospects. Cells 2023, 12, 2750. https://doi.org/10.3390/cells12232750

AMA Style

Hibler W, Merlino G, Yu Y. CAR NK Cell Therapy for the Treatment of Metastatic Melanoma: Potential & Prospects. Cells. 2023; 12(23):2750. https://doi.org/10.3390/cells12232750

Chicago/Turabian Style

Hibler, Winston, Glenn Merlino, and Yanlin Yu. 2023. "CAR NK Cell Therapy for the Treatment of Metastatic Melanoma: Potential & Prospects" Cells 12, no. 23: 2750. https://doi.org/10.3390/cells12232750

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