Next Article in Journal
A Computational Framework for Prediction and Analysis of Cancer Signaling Dynamics from RNA Sequencing Data—Application to the ErbB Receptor Signaling Pathway
Next Article in Special Issue
Comprehensive Exome Analysis of Immunocompetent Metastatic Head and Neck Cancer Models Reveals Patient Relevant Landscapes
Previous Article in Journal
Blockage of Store-Operated Ca2+ Influx by Synta66 is Mediated by Direct Inhibition of the Ca2+ Selective Orai1 Pore
Previous Article in Special Issue
Role of Oral Bacteria in the Development of Oral Squamous Cell Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Regulation of NFκB Signalling by Ubiquitination: A Potential Therapeutic Target in Head and Neck Squamous Cell Carcinoma?

Tumor Biology Section, Head and Neck Surgery Branch, National Institute of Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, USA
*
Authors to whom correspondence should be addressed.
Cancers 2020, 12(10), 2877; https://doi.org/10.3390/cancers12102877
Submission received: 24 August 2020 / Revised: 22 September 2020 / Accepted: 29 September 2020 / Published: 7 October 2020
(This article belongs to the Special Issue Head and Neck Cancers)

Abstract

:

Simple Summary

Head and neck cancer is the sixth most common cancer worldwide and typically caused by smoking and alcohol consumption, or infection with Human Papillomavirus (HPV). Currently, treatment options include surgery, radiotherapy, and/or chemotherapy. However, whilst the survival rate in HPV+ cancer patients is better than those without HPV, survival rates have not improved greatly in recent years, and recurrence rates are high. Ubiquitination is a critical regulatory mechanism that can promote the activation or termination of signal cascades, such as the NFκB pathway, which plays an important role in the pathogenesis and therapeutic resistance of head and neck cancer. In this review, we discuss how NFκB signalling is regulated by ubiquitination and how the ubiquitin pathway is deregulated in head and neck cancer, highlighting how the this pathway may be targeted to inhibit NFκB signalling.

Abstract

Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide, with over 600,000 cases per year. The primary causes for HNSCC include smoking and alcohol consumption, with an increasing number of cases attributed to infection with Human Papillomavirus (HPV). The treatment options for HNSCC currently include surgery, radiotherapy, and/or platinum-based chemotherapeutics. Cetuximab (targeting EGFR) and Pembrolizumab (targeting PD-1) have been approved for advanced stage, recurrent, and/or metastatic HNSCC. Despite these advances, whilst HPV+ HNSCC has a 3-year overall survival (OS) rate of around 80%, the 3-year OS for HPV− HNSCC is still around 55%. Aberrant signal activation of transcription factor NFκB plays an important role in the pathogenesis and therapeutic resistance of HNSCC. As an important mediator of inflammatory signalling and the immune response to pathogens, the NFκB pathway is tightly regulated to prevent chronic inflammation, a key driver of tumorigenesis. Here, we discuss how NFκB signalling is regulated by the ubiquitin pathway and how this pathway is deregulated in HNSCC. Finally, we discuss the current strategies available to target the ubiquitin pathway and how this may offer a potential therapeutic benefit in HNSCC.

Graphical Abstract

1. Introduction

Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide, with over 600,000 cases and around 300,000 deaths per year [1]. Current therapies include surgery, radiotherapy, and standard chemotherapies [2]. However, targeted therapies have been approved for use in the advanced stage, recurrent, and/or metastatic HNSCC, including anti-epidermal growth factor receptor (EGFR) (cetuximab) and anti-programmed cell death protein (PD-1) therapy (pembrolizumab) [3,4,5]. Despite these advances, whilst HPV+ HNSCC has a 3-year overall survival (OS) rate of around 80%, the 3-year OS for HPV− HNSCC is still around 55% [1]. While the early stage disease has survival rates of 70–90%, the locally advanced disease can have survival rates <50% [1].
The recent increase in survival rate is largely due to an epidemiological shift in the causes of HNSCC. In low- and middle-income countries, the primary known risk factors for most HNSCC cases (>90%) remain alcohol and tobacco use [6]. However, in high-income countries, such as the USA and Western Europe, over 70% of cases involve infection with human papillomavirus (HPV) [1,7]. HPV is an important carcinogenic factor, accounting for around 5% of all cancers worldwide [8]. The most common cancer associated with HPV infection is cervical cancer, which is primarily caused by the high-risk HPV types 16 and 18 (70%) [9]; in contrast, HPV+ HNSCC is almost always due to HPV16 infection (over 90%) [1]. Interestingly, the incidence of HPV+ oropharyngeal cancers is higher in current smokers than never smokers, suggesting that smoking may also play a role in the clinical outcome of these patients [10].
The differences between these cancers manifest in several characteristics (Table 1). HPV− cancers are decreasing, occur in older adults, and localized in any region of the head and neck; in contrast, HPV+ HNSCC cases are increasing, mostly occurs in younger adults and is localized primarily in the oropharynx and the base of the tongue (BOT) [1,2]. In clinical terms, HPV− HNSCC are primarily well differentiated with a generally keratinized histology, whereas HPV+ HNSCC tumors are usually poorly differentiated or have basaloid histology [11]. HPV+ HNSCC also have a better prognosis, respond better to chemoradiation therapy, and have higher survival rates than HPV− HNSCC [1]. Thus, while most HPV+ HNSCC patients are diagnosed at a clinically advanced stage with regional lymph node metastases, after treatment, they are less likely to develop local-regional recurrence, secondary malignancies, or distant metastases. Thus, it is clear that HNSCC cases can be separated into (at least) two distinct subtypes, dependent on HPV status, that result in different clinical outcomes.
As one of the hallmarks of cancer [12], the role of inflammation in tumor initiation and progression has become an important area of investigation. Inflammatory conditions can induce signalling and genetic damage to initiate or promote tumorigenesis; furthermore, genetic and epigenetic changes, as well as the HPV oncogenes, can generate an inflammatory microenvironment that further supports tumor progression [6,13]. Many inflammatory cytokines and signalling pathways are implicated in the pathogenesis of HNSCC [14,15]; the expression of these inflammatory mediators and intrinsic resistance of tumor cells can be driven by the oncogenic insults known to contribute to the development of HNSCC [14,16,17].
An important driver and inducer of inflammatory mediators, which promotes cell survival and therapeutic resistance, is the transcription factor nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) [18]. In HNSCC, NFκB is often aberrantly activated and is a common mediator of the effects of numerous pro-inflammatory cytokines (TNFα and IL-1), as well as other signalling pathways targeted by oncogenic mutations, genetic copy number alterations (CNA), or viral oncogenes [14,19,20]. NFκB drives the expression of a repertoire of genes that promote malignant progression, including cell proliferation and survival, therapeutic resistance, and pro-inflammatory factors [21,22].
An important process in the regulation of signalling pathways is protein ubiquitination, which plays a pivotal role in the activation and termination of many signalling pathways, including the NFκB pathway [23]. Thus, defects in ubiquitination can result in aberrant NFκB activation, or the failure to terminate NFκB signalling.
In this review, we will discuss how the ubiquitin system regulates NFκB signalling and how this may contribute to the aberrant NFκB activity observed in HNSCC. Additionally, we will summarize the current methods of modulating the ubiquitin system that could potentially be used to inhibit NFκB signalling in HNSCC.

2. The NFκB Signalling Pathway

The difference in clinical response of HPV+ and HPV− HNSCC is not fully understood; however, recent developments in the genomic and transcriptomic analysis of primary HNSCC cases have provided insight into the difference in clinical response [14,17]. Subsequent studies have examined the alterations in specific pathways and how these relate to the pathogenesis of HNSCC and have highlighted a critical role for aberrant NFκB activity.
The NFκB family of transcription factors are homo- or heterodimers that regulate a large number of genes involved in inflammation, immunity, and cell survival [24]. In humans, the NFκB family has five known members: p105/p50 (NFκB1), p100/p52 (NFκB2), p65 (RELA), c-REL, and RELB [24]. Each family member contains an N-terminal Rel homology domain (RHD), regulating their dimerization, DNA binding ability, and nuclear localization. The RHD also binds to the inhibitory of κB proteins (IκBs), critical regulators of NFκB, which sequester it in the cytoplasm in unstimulated cells [24]. p50 and p52 do not contain their own transactivation domains (TAD); they are generated from the precursor proteins, p105 and p100, respectively, following phosphorylation, ubiquitination, and proteasomal degradation of a C-terminal Inhibitor-kappaB kinase (IκB)-like domain [25,26]. Homodimers of p50 or p52 cannot activate gene transcription without additional co-factors and therefore function as gene repressors [27]. p50 and p52 can also bind to a TAD-containing family member to generate a functional NFκB heterodimer. p65, c-REL and RELB contain a TAD capable of activating gene expression. The TAD of RELs contains sites for post-translational phosphorylation and/or acetylation, which enhances transactivation and duration of DNA binding [28].
The IκB family consists of several ankyrin repeat-containing proteins, including IκBα, IκBβ, IκBɛ, IκB-δ (IκBNS), IκB-ζ, and BCL-IκBα, IκBβ and IκBɛ inhibit DNA binding and enhance export and sequestration of NFκB in the cytoplasm, whereas IκB-δ, IκB-ζ, and Bcl-3 function in the nucleus as co-activators of NFκB [29]. Whereas the majority of IκBs serve as inhibitors of NFκB, the IκBζ, and Bcl-3 isoforms instead potentiate NFκB transactivation in the nucleus [30].

2.1. Canonical NFκB Signalling

The mechanisms of activation and function of the canonical (classical) and non-canonical (alternative) NFκB pathways are often distinct; however, studies have revealed that cross-activation of these pathways occurs at several levels via kinase function, processing of p100/p105 precursors, and transcription [31]. The canonical NFκB pathway plays an important role in biological processes such as innate and adaptive immunity and cell survival. A variety of stimuli activate canonical NFκB signalling, which converges upon a trimeric IκB kinase (IKK) complex, consisting of catalytic subunits IKKα and IKKβ and the regulatory subunit NFκB essential modifier (NEMO, also known as IKKγ). IKKβ and NEMO serve essential and non-redundant roles in the activation of canonical NFκB signalling, whereas IKKa regulates both canonical and non-canonical pathways [31]. IKKα and IKKβ can phosphorylate IκB proteins at two N-terminal serine residues, triggering their ubiquitination and proteasomal degradation, thus allowing NFκB to enter the nucleus and activate stimulus-specific gene programs.
Canonical NFκB pathway is exemplified by activation via proinflammatory cytokines such as Tumor Necrosis Factor α (TNFα) or Interleukin-1β (IL-1β) (Figure 1). In TNFα mediated signalling, TNFα binding to the TNF receptor (TNFR1) initiates recruitment of a complex consisting of Tumor necrosis factor receptor type 1-associated DEATH domain (TRADD), TNF-receptor-associated factor 2 (TRAF2), receptor-interacting serine/threonine-protein kinase 1 (RIP1), and cellular Inhibitor of Apoptosis 1/2 (cIAP1/2). This leads to the K63-polyubiquitination of RIP1 by the heterodimer UBC13-UBE2V1, an E2 protein complex, and cIAP1/2, which are E3 ligases [32].
The resulting poly-ubiquitin chains form a scaffold for the recruitment of TAB2 and NEMO and activation of the transforming growth factor β-activated kinase 1 (TAK1) and IKK complexes, respectively. Subsequently, TAK1 induces the phosphorylation of IKKβ, which subsequently induces IκBα phosphorylation, proteasomal degradation, and NFκB nuclear translocation and activation [33,34].
In IL-1 signalling, the ligand-bound IL-1 receptor (IL-1R) recruits the adapter protein MyD88, resulting in the recruitment of the kinases Interleukin 1 Receptor Associated Kinase (IRAK) 1 and IRAK4 [35]. IRAK1/ 4 can then dissociate from the receptor and interact with the E3 ligase TRAF6, leading to the formation of K63-polyubiquitin chains on TRAF6 and IRAK1, serving as a binding platform for the TAK1 complex via TAB2 [36]. As with TNFα signalling, TAK1 then activates the IKK complex [33].

2.2. Non-Canonical NFκB Signalling

While the canonical NFκB pathway can be activated by several inflammatory cytokines and other stimuli, the non-canonical, or alternative, NFκB pathway is activated by a different set of cytokine/receptor pairs in the TNFR superfamily (Figure 2). These typically do not contain death domains and include the BAFF receptor (BAFFR), CD40, and lymphotoxin B receptor (LTβR), activated by BAFF, CD40L, and LTβ, respectively [37]. Non-canonical NFκB signalling is driven by NFκB-inducing kinase (NIK); in unstimulated cells, NIK expression is low due to its persistent ubiquitination and proteasomal degradation via the TRAF3 E3 ligase in a complex containing TRAF2, TRAF3, and cIAP1/2 [38].
Upon ligand binding, TRAF3 is polyubiquitinated and degraded by the TRAF2/TRAF3/cIAP complex, allowing the cytoplasmic accumulation of NIK. This allows NIK to induce the phosphorylation and activation of IKKα, which in turn phosphorylates p100, promoting its partial proteasomal degradation. This liberates the p52/RELB dimer which can then translocate into the nucleus to drive gene expression [37].

2.3. Ubiquitin-Mediated Regulation of NFκB Signalling Pathway

Ubiquitination plays an essential role in the regulation of NFκB [23]. Ubiquitin is a small, highly conserved, ubiquitously expressed protein found in all eukaryotic species, from yeast to humans [39]. Ubiquitin regulates a wide range of cellular functions, including protein degradation and the modulation of signal transduction, which is achieved through the conjugation of ubiquitin to protein substrates through a stepwise, enzymatic cascade [40,41]. This process requires three enzyme classes: E1 ubiquitin-activating enzymes, E2 ubiquitin-conjugating enzymes, and E3 ubiquitin ligases (Figure 3). The human genome encodes for eight E1 proteins (only two E1 proteins are involved in ubiquitination), around 50 E2 proteins, and over 700 E3 proteins, demonstrating the complexity of the ubiquitin system [41]. In addition, ubiquitin can be removed by deubiquitinating enzymes (DUBs), of which there has been around 100 characterized in 7 distinct families [42].
Ubiquitin contains 76 amino acids, including seven lysine residues—at amino acid position 6 (K6), K11, K27, K33, K48, and K63; the C-terminus of ubiquitin can be conjugated to any of these seven lysines on another ubiquitin, forming polyubiquitin chains of different linkages [43] Additionally, ubiquitin can be directly attached to the N-terminal methionine of another ubiquitin to form linear polyubiquitin chains [43].
On a structural basis, three types of ubiquitin linkages have been identified: monoubiquitination, polyubiquitination, and branched ubiquitination [40]. Each type of ubiquitination, and upon which lysine ubiquitin conjugation occurs, results in different biological outcomes, demonstrating the complexity of the ubiquitin system. Monoubiquitination is involved in the regulation of DNA damage repair; for example, RNF2-mediated monoubiquitination of γH2AX is required for the initiation of the DNA damage response (DDR) via the recruitment of Ataxia-Telangiectasia Mutated (ATM) and Mediator of DNA damage checkpoint protein 1 (MDC1) [44]. K48-polyubiquitination is most commonly associated with marking its protein substrate for proteasomal degradation [45]; however, it also has a proteasome-independent function [46]. K63-polyubiquitination is involved in creating ligand-receptor mediated scaffolds for signalling proteins, exemplified by its role in the activation of NFκB signalling [32]. K6-polyubiquitination has mostly been associated with the response to DNA damage; however, it may also have an important role in endoplasmic reticulum (ER) homeostasis and mitophagy [47]. K27, K29, and K33-polyubiquitination are the least understood ubiquitin linkages. K27-polyubiquitination is associated with the regulation of the DDR [48], whereas K29-polyubiquitination is associated with the proteasomal function [49]. K33-polyubiquitination plays an important role in anterograde trafficking [50].
The most well studied polyubiquitin chain is K48-polyubiquitination, which promotes the proteasomal degradation of protein substrates [45]. K11-polyubiquitination can also promote proteasomal degradation [51]; in addition, K11-polyubiquitination plays an important role in NFκB signalling [52]. Finally, the M1 linear-polyubiquitination has a primary role in promoting NFκB signalling [53].
Branched ubiquitin chains are poorly understood, likely due to detection difficulties [54]. Branched K48-/K11 and K48-/K29-polyubiquitination are strong degradation signals [54]. Branched K63-/K48-polyubiquitination enhances NFκB signalling by preventing K63-polyubiquitin deubiquitination [55]; however, K63-polyubiquitination also can promote the formation of branched K63-/K48- polyubiquitination, promoting proteasomal degradation [56].
Ubiquitination is a reversible post-translational modification and can be removed by deubiquitinases (DUBs). Over 100 DUBs have currently been characterized in seven distinct families [42]: ubiquitin C-terminal hydrolases (UCHs), ubiquitin-specific proteases (USPs), ovarian tumor proteases (OTUs), the Machado-Josephin domain superfamily (MJD), the MINDY family, and the ZUFSP family members function as cysteine proteases, whereas JAB1/MPN/MOV34 metalloenzymes (JAMMs) are zinc-dependent metalloproteases [42]. These enzymes have different specificities; some cleave only one kind of ubiquitin linkage (e.g., OTULIN; M1 linkages) or multiple ubiquitin linkages (e.g., CYLD; primarily M1 and K63 linkages) [57].

2.3.1. Canonical NFκB Signalling

At the receptor level, an essential step in cytokine mediated NFκB activation is the K63-polyubiquitination of RIP1 and NEMO. Several studies have demonstrated that the E2 proteins UBC13/UEV1A are important regulators of K63-polyubiquitination; these proteins act in concert with cIAP1/2 and TRAF6 to induce K63-polyubiquitination on RIP1 and NEMO, respectively [58]. Additionally, the E3 ligase Parkin can induce RIP1 K63-polyubiquitination (Figure 2; [59]). K63-polyubiquitin chains on RIP1 serve as a binding platform for the regulatory proteins TAB1 and TAB2 through their ubiquitin binding domains (UBDs) [60].
This leads to the recruitment, autophosphorylation and activation of TAK1, and subsequent downstream activation of the IKK complex and IκBα degradation [33]. Furthermore, the K63-polyubiquitination of TAK1 by TRAF2 and TRAF6 is essential for TAK1 activation and downstream signalling [32]. Additionally, NEMO itself can bind to K63-polyubiquitination on RIP1; this brings the IKK complex into close proximity to TAK1, promoting the phosphorylation and activation of the IKK complex [61].
In IL-1β signalling, activation of IRAK1 via autophosphorylation results in the phosphorylation of the E3 ligase Pellino 3; this induces Pellino 3-dependent K63-polyubiquitination of IRAK1, recruitment of NEMO and the downstream activation of NFκB activity [62].
Linear ubiquitination also plays a key role in NFκB activation. M1 linear- polyubiquitination is mediated by the E3 ligase Linear UBiquitin chain Assembly Complex (LUBAC) in concert with the E2 protein UBCH7; LUBAC contains the subunits HOIL-1L, HOIP, and Sharpin [63]. Upon K63-polyubiquitination of RIP1 or TRAF6, LUBAC is recruited and mediates the linear ubiquitination of RIP1 and/or NEMO; these functions are essential for canonical NFκB signalling [53].
In addition to K63- and linear polyubiquitination, components of the NFκB are also extensively modified by atypical polyubiquitin chains. RIP1 is also K11-polyubiquitinated; this is mediated by UBCH5 and cIAP1 [52]. These K11-polyubiquitin chains are TNFα inducible and were bound by NEMO, suggesting they may play an important role in downstream NFκB activation [52]. Furthermore, TNFα can induce UBCH5C/cIAP1 mediated K6-polyubiquitination on NEMO, which mediates TNFα-induced NFκB activation [47]. The E3 ligase tripartite motif protein 23 (TRIM23) can induce K27-polyubiquitin chains in response to viral infection, as this promotes anti-viral NFκB activity [64].
A major step in the activation of the canonical NFκB pathway is the proteasomal degradation of its negative regulator IκBα via the addition of K48-polyubiquitin chains, which occurs after phosphorylation of IκBα at serine 32 and 36 by the IKK complex [34]. IκBα ubiquitination is mediated by a UBC4/5 family member and the E3 ligase Skp1–Cullin1–F-box protein (SCF)β-TrCP (Figure 2; [34]). The degradation of IκBα releases the p65/p50 heterodimer, unveiling their nuclear localization sequences and cleft for DNA binding. Thus, proteasomal degradation of IκBα is essential for the activation of the canonical NFκB pathway. SCFβ-TrCP also promotes the degradation of IRAK1 in an IL-1β-dependent manner; this is required for TAK1-mediated NFκB activation [36].
In addition to the activation of NFκB, ubiquitination can also inhibit NFκB signalling. cIAP1 and SIAH2 promote the K48-polyubiquitination of TRAF2, attenuating NFκB activity [65,66]. Further downstream, K48-polyubiquitination of IKKα and β results in their proteasomal degradation, inhibiting NFκB activity [67]. IKKβ K48-polyubiquitination is mediated by the KEAP1-Cul3-RBX1 E3 ligase complex [67]; however, the E3 ligase for IKKα is unclear. The E3 ligase ITCH induces K48-polyubiquitinated of TAK1 [68]. Furthermore, the E3 ligase TRAF7 induces K29-polyubiquitination of NEMO and p65, facilitating the lysosomal degradation of NEMO and p65, inhibiting NFκB activity [69]. Additionally, the NFκB protein p65 is K48-polyubiquitinated by several E3 ligases, including inhibitor of growth protein 4 (ING4) [70], suppressor of cytokine signalling 1 (SOCS1) [71], Peroxisome proliferator-activated receptor γ (PPARγ) [72], HERC (82) and PDZ- and LIM-domain containing protein 2 (PDLIM2) [73].
The removal of ubiquitin by deubiquitinases (DUBs) also plays a key role in the NFκB pathway. Well characterized DUBs involved in NFκB signalling are A20 (TNFα-Induced Protein 3, TNFAIP3, a member of the OTU family, and CYLD, a member of the USP family [74,75]. Unusually, A20 can also function as a deubiquitinase and an E3 ligase. A20 regulates NFκB signalling via several mechanisms. A20 can remove K63-polyubiquitin chains from RIP1; TNFα stimulation activates NFκB and induces A20 expression as part of a negative feedback loop [76]. IKKα mediated phosphorylation of the adapter protein TAX1BP1 leads to the formation of the A20 ubiquitin-editing complex containing TAX1BP1, ITCH, and RNFThis functions as a deubiquitinase complex to remove K63-polyubiquitin chains from RIP1 [77]. A20 also induces K48-polyubiquitination of RIP1 and UBC13, promoting their proteasomal degradation [78]. Furthermore, A20 inhibits the interaction between the UBC13 and TRAF2/6 and cIAP1/2, inhibiting K63-polyubiquitin chain formation [79]. Additionally, the binding of A20 to K63-polyubiquitin chains of RIP1 can inhibit TAK1-mediated phosphorylation of IKKβ [80]. Finally, A20 binds to linear-polyubiquitin chains, preventing the association between LUBAC and NEMO [81]. All of these functions of A20 inhibit downstream NFκB signalling.
The deubiquitinase CYLD removes K63-polyubiquitin chains from RIP1, TRAF2, TRAF6, TAK1, and NEMO, as well as removing M1-linear polyubiquitination from RIP1 [23]. As such, CYLD functions as a tumor suppressor gene in several cancers [75]. Other deubiquitinases can also modulate NFκB signalling. USP4, USP20, and USP21 can remove K63-polyubiquitin chains from RIP1 (USP4 and USP21) and TRAF6 (USP20), respectively [82,83,84]. Furthermore, USP4 can remove K63-polyubiquitin chains from TAK1, whereas USP21 can remove K63-polyubiquitin chains from TRAFConversely, the OTU family member OTUB1 can stabilize both cIAP1/2 and UBC13, resulting in enhanced NFκB signalling [85]. Additionally, USP11 and USP15 can deubiquitinate and stabilize IκBα, promoting NFκB nuclear translocation [86].
The deubiquitinases OTUD7b, USP48, and USP7 can regulate NFκB signalling at multiple levels. OTUD7b can remove K11- and K63-polyubiquitin chains from RIP1 [87]. In contrast, USP7 can remove K48-polyubiquitin chains from IKKα and p65, thereby promoting NFκB signalling [88], and USP48 stabilizes TRAF2 and p65 by the removal of K48- polyubiquitin chains [89,90].

2.3.2. Non-Canonical NFκB Signalling

In contrast to canonical NFκB signalling, the regulation of non-canonical NFκB signalling is not as well understood (Figure 3). In unstimulated cells, NIK is constantly turned-over by TRAF3, which, in combination with another E3 ligase C terminus of HSC70-Interacting Protein (CHIP), promotes the K48-polyubiquitination and proteasomal degradation of NIK [38,91]. Upon ligand activation, cIAP-mediated K48-polyubiquitination of TRAF3 promotes its proteasomal degradation; this induces NIK accumulation and the subsequent activation of IKKα [38]. Interestingly, the E3 ligase ZFP91 can stabilize and activate NIK [92]. Downstream, SCFβ-TrCP can induce K48-polyubiquitination of both p100 and p105, whereas SCFFBXW7α can induce K48-polyubiquitination of p100, targeting them for partial proteasomal degradation [29]. This liberates p52 and p50, inducing their nuclear translocation and subsequent NFκB activation.
The only deubiquitinases identified to play a role in non-canonical NFκB signalling are OTUD7b and OTUBOTUD7b and OTUB1 remove K48-polyubiquitin chains from TRAF3 and cIAP1, respectively [85,87]; therefore, OTUD7b and OTUB1 can inhibit both the canonical and non-canonical NFκB pathways.
It is thus evident that the modulation of NFκB signalling by the ubiquitin system is complex, with many E3 ligases and DUBs targeting multiple components to ensure efficient regulation. This suggests that defects in the ubiquitin system can result in chronic NFκB signalling and promote cancer progression.

3. Ubiquitin-Mediated NFκB Signalling in Head and Neck Squamous Cell Carcinoma

NFκB Signalling in HNSCC

NFκB signalling drives cell proliferation, survival, therapeutic resistance, and pro-inflammatory factors [93,94]. As mentioned previously, NFκB is often aberrantly activated in HNSCC, including oropharyngeal, laryngeal, hypopharyngeal, and tongue cancers [95,96,97,98]. Additionally, NFκB signalling plays an important role in other cancers of the head and neck distinct from HNSCC, such as salivary gland cancer and nasopharyngeal carcinoma [99,100,101]. Early studies from our lab demonstrated that key components of the canonical NFκB pathway are aberrantly activated in HNSCC and are further activated by TNFα via the IKK complex [22,102,103]. Furthermore, recent studies have demonstrated the activation of non-canonical NFκB signalling, involving NIK and IKKα [104,105,106]. Both of the NFκB pathways have also been shown to interact with other pro-oncogenic signalling pathways, including the EGFR, STAT3, and AP-1 pathways [104,107,108].
The activation of NFκB in HNSCC occurs through various mechanisms, including somatic mutations, genetic copy number alterations (CNA), or the expression of the HPV viral oncogenes [14,19,20]. Several genome wide studies have highlighted the prevalence of several alterations in the ubiquitin system implicated in the aberrant activation of NFκB signalling in HNSCC (Table 2).
In the TCGA HNSCC dataset, frequent amplification and overexpression of BIRC2/3 (on chromosome 11q22), encoding cIAP1/2, was observed [109]. These E3 ligases are the critical mediator of TNFα signalling, promoting K63-polyubiquitination of RIP1 and K48-polyubiquitination and subsequent proteasomal degradation of TRAF3 [36]. BIRC2 is more commonly amplified and overexpressed in HPV- HNSCC, while BIRC3 overexpression appears to be more common in HPV+ HNSCC [109]. Additional ubiquitin components are upregulated at the mRNA level; OTUB1 mRNA levels are high in HNSCC, without significant copy number alterations [14].
A key finding of the TCGA study was the identification of common mutations/deletions in the CYLD and TRAF3 genes in HPV+ HNSCC [17,110]. CYLD removes both K63-polyubiquitin and M1 linear-ubiquitin chains, inhibiting NFκB signalling at several different steps [111]. TRAF3 is a negative regulator of both classical and alternate NFκB [38,112]. Further studies demonstrated that these mutations in CYLD/TRAF3, or the loss of TRAF3, resulted in enhanced NFκB signalling and promoted HPV+ HNSCC [105,110]. Intriguingly, loss of CYLD/TRAF3 was observed in a subset of HPV+ HNSCC with episomal HPV, suggesting loss may enhance the establishment of persistent infection and susceptibility to transformation [110]. Furthermore, similar genomic alterations in CYLD and/or TRAF3 were observed salivary cancers and in a cohort of nasopharyngeal carcinomas negative for the Epstein Barr Virus (EBV) [101,113,114].
The mutational analysis also demonstrated that SCFFBXW7 is one of the most commonly mutated genes in HPV− HNSCC [17]; this E3 ligase promotes the degradation of the NFκB protein p100, inhibiting NFκB activity [112]. FBXW7 ‘hot-spot’ mutations (R505 > R479 > R465) occur in the substrate binding domain; these mutations inhibit the interaction between SCFFBXW7 and its substrates [115]. Therefore, these mutations may promote the stabilization of p100, potentially increasing the inactive precursor for stimuli-induced processing, thereby enhancing NFκB activity [116].
Cullin ring ligase 3 (CUL3) is an E3 ligase, and KEAP1 is a CUL3-specific adapter protein. The most well studied substrate inhibited by the CUL3-KEAP1 complex is Nuclear factor erythroid 2-related factor 2 (NRF2), a transcription factor that regulates antioxidant and stress-responsive genes [117]. Co-activated NRF2 and NF-kB may cooperate in transcriptional activation [117]. The CUL3-KEAP1 complex also targets IKKβ for proteasomal degradation, inhibiting NFκB signalling [67]. Thus, mutations, or loss, of either CUL3 or KEAP1 can result in increased inflammatory signalling through co-activation of NFκB and NRFAdditionally, in non-tumorigenic Retinal Pigment Epithelium RPE cells, CUL3 inactivation in combination with TTP53 inactivation results in an oncogenic phenotype driven by NFκB and AP-1 signalling [118]. In line with this, mutations in CUL3 significantly co-occur with mutations in TTP53 in HNSCC [119], suggesting that the CUL3-KEAP1 E3 ligase complex may function in concert with deleted, mutated, or degraded TTP53 in HNSCC to promote NFκB, AP-1, and NRF2 activation.
These differences in genomic alterations in HPV+ and HPV− HNSCC demonstrate that the activation of NFκB signalling can occur by different mechanisms. However, HPV itself can also induce NFκB activation; HPV E6 has been shown to promote NFκB activation by several mechanisms [19,20]. Interestingly, E6 mediates the proteasomal degradation CYLD in hypoxic conditions [20], an environment that occurs in the poorly vascularized center of cervical lymph nodes common in HPV+ HNSCC. Recently, it has been shown that E6 can also activate the GTPase Rac1, which regulates NFκB activity in cervical cancer cells [19]. Thus, it is clear that HPV can induce NFκB activity by a number of mechanisms that could potentially be specifically targeted for therapy for HPV+ HNSCC.

4. Targeting the Ubiquitin System to Inhibit NFκB Signalling in HNSCC

The success of kinase inhibitors in cancer treatment over the past 30 years, along with recent research into how defects in the ubiquitin system contribute to disease, has prompted the pharmaceutical industry to investigate the ubiquitin system as a potential therapeutic target [120]. So far, progress has been slow, and only a few small molecule inhibitors have been successfully developed and are clinically approved or in clinical trials. This may be due to the complex and dynamic nature of ubiquitination, which involves many protein-protein interactions that have been challenging to disrupt by small molecules with the necessary specificity to avoid toxicity.
In spite of this, recent advances in the understanding of ubiquitination biology have identified new small molecule inhibitors targeting components of the ubiquitin system that are efficacious in several diseases, including cancer [120]. Below, we will review how targeting various components of the ubiquitin system could be utilized to regulate the NFκB pathway in HNSCC.

4.1. Targeting the Proteasome

The most advanced inhibitors of the ubiquitin system are proteasome inhibitors. The inhibition of the proteasome deregulates protein turnover, resulting in the stabilization of tumor suppressors, such as TP53 [121]. Additionally, proteasome inhibition can inhibit the NFκB pathway by allowing the accumulation of phosphorylated IκBα [22]. Thus, proteasome inhibition can have multiple effects that inhibit cell proliferation and promote apoptosis.
Since the mid 1990s, proteasome inhibitors have been tested in many cancers, including multiple myeloma (MM) and various lymphomas [121]. Currently, two proteasome inhibitors are approved by the FDA, bortezomib (Velcade®, Millenium Pharmaceuticals, Cambridge, MA, USA) and carfilzomib (Kyprolis®, Amgen, Thousand Oaks, CA, USA) [122]. Bortezomib is a first-generation proteasome inhibitor, which reversibly binds to the catalytic site of the proteasome [123]. Bortezomib is currently FDA approved for the treatment of MM and mantle-cell lymphoma (MCL). Studies have shown that bortezomib’s efficacy in MM and MCL is likely due to multiple mechanisms, including the inhibition of proliferation and the induction of apoptosis [124,125]. Additionally, bortezomib inhibits NFκB signalling due to the stabilization of IκBα [126]. Unfortunately, bortezomib treatment often results in intrinsic or acquired resistance in many patients [123]. Therefore, the second-generation of proteasome inhibitors were designed to overcome this; carfilzomib is an irreversible inhibitor of the proteasome that is currently approved for the treatment of MM that is refractory to at least two previous therapeutics, including bortezomib [127].
Proteasome inhibitors have been tested as a potential therapeutic option in HNSCC. In early clinical studies, bortezomib treatment resulted in clinical tumor shrinkage or necrosis in a subset of patients [22,128]. In both mouse and human squamous cell carcinomas, bortezomib treatment was shown to inhibit NFκB signalling [22,129]. Furthermore, it inhibited canonical NFκB signalling to a greater extent than non-canonical NFκB signalling [18]. When combined with anti-EGFR antibody cetuximab and radiation, these effects were found to be negated by the inhibitory effect of bortezomib on the degradation of EGFR, which can promote resistance via activation of NFκB and MAPK-AP-1 axes [130]. As a result of early recurrences observed, this phase I clinical trial of bortezomib in combination with chemoradiotherapy and cetuximab in stage IV HNSCC was halted (Table 3). Additional pre-clinical studies demonstrated that when combined with histone deacetylase (HDAC) inhibitors or cisplatin, bortezomib enhanced the induction of apoptosis and therapeutic responses [131]. However, these combinations were relatively toxic and offered too narrow a therapeutic window in preclinical models.

4.2. Targeting E1 Activating Enzymes

Ubiquitin activating enzymes (UBEs or E1 enzymes) are the first step of the ubiquitination cascade (Figure 1). E1 enzymes catalyze the formation of a thioester bond between the C-terminal carboxyl group of ubiquitin and the cysteine residue of E1 in an ATP-dependent manner [120]. To date, eight E1 enzymes have been identified in humans that regulate the addition of ubiquitin and ubiquitin-like proteins (UBLs) to protein substrates. Only two E1 enzymes, UBA1 and UBA6, control the ubiquitination of all proteins.
PYR-41 and PYZD-4409 were the earliest identified inhibitors for UBA1; both inhibitors irreversibly inhibit UBA1 by covalent attachment to the active cysteine (Cys632) residue [134,135]. Both inhibitors have demonstrated some anti-tumor effects in vitro in leukemia cell lines and can inhibit NFκB signalling. However, the apoptosis observed is likely due to off target effects caused by the global inhibition of ubiquitination [134]. A more potent inhibitor of UBA1 called MLN7243 (also known as TAK-243) forms a covalent adduct with ubiquitin and, in contrast to PYR-41 and PYZD-4409, inhibits UBA6 as well as UBAMLN7243 has shown anti-tumor activity in vivo in immunodeficient mice subcutaneously grafted primary human AML cells [136]. However, in a phase I dose-escalation clinical trial (clinicaltrial.gov identifier: NCT02045095) in advanced solid tumors, over one third of patients had serious adverse effects, suggesting that such global inhibition of ubiquitination is toxic.
In addition to the two E1 activating enzymes involved in the ubiquitination pathway, the E1 activating enzyme UBA3 should be mentioned. UBA3, also known as the neuronal precursor cell, expressed, developmentally down-regulated 8 (NEDD8)-activating enzyme (NAE)2, is the E1 activating enzyme of the ubiquitin-like protein NEDD8 [137]. This E1 functions in concert with an additional subunit, NAE1; the heterodimer of NAE1 and 2 is normally just referred as NAE.
The addition of NEDD8 to protein substrates is termed NEDDylation. Whilst this pathway is not part of the ubiquitin system, NEDDylation is essential for the activation and function of the Cullin Ring Ligase (CRL) family of E3 ligases [138]. The cullin subunit of CRLs is NEDDylated, and CRL E3 ligase is often involved in cell proliferation. Several are deregulated in cancer, including HNSCC (17); thus, inhibition of CRL E3 ligases results in the accumulation of CRL substrates.
MLN4924 (Pevonedistat) is an NAE inhibitor that forms a covalent adduct with NEDD8 and is currently in Phase I/II trials for various types of leukemia (clinicaltrial.gov identifier: NCT03268954). Initial experiments showed that MLN4924 induced DNA damage in vitro and limited tumor growth when used to treat immunocompromised mice in vivo [139].
Several studies have shown that MLN4924 inhibits NFκB signalling; in both chronic lymphocytic leukemia (CLL) and non-Hodgkin lymphoma, MLN4924 stabilized phosphorylated IκBα, inhibiting p65 and p50 nuclear translocation, NFκB activity and inducing apoptosis [139]. In HNSCC, MLN4924 has anti-tumor effects, inducing apoptosis and sensitizing HNSCC cells to radiation and TRAIL-induced cell death [140,141,142]. However, the anti-tumor mechanism of MLN4924 in HNSCC is still unclear, and no clinical trials in HNSCC have been performed.

4.3. Targeting E2 Conjugating Enzymes

The E2 ubiquitin-conjugating enzymes interact with numerous E3 ligases to transfer ubiquitin molecules onto substrate proteins (Figure 1). Over 30 identified E2 genes in humans [143]; therefore, targeting E2 enzymes should provide more selectivity than E1 enzymes. However, only a few inhibitors targeting E2 conjugating enzymes have been discovered so far, with none currently in clinical trials.
NSC697923 is an irreversible, covalent inhibitor of the UBC13/UBE2V1 heterodimer that catalyzes the formation of K63-polyubiquitin chains [144,145]. Inhibition occurs by covalent attachment of NSC697923 to the catalytic cysteine of UBC13, preventing the conjugation of ubiquitin [145]. Interestingly, NSC697923 was originally identified in a screen for molecules inhibiting the NFκB pathway [144]. The research demonstrated that NSC697923 suppressed the proliferation of diffuse large B-cell lymphoma (DLBCL) cells in vitro, via the inhibition of the NFκB pathway [144]. Another UBC13 inhibitor, BAY117082, was first thought to inhibit the IKK complex, as it inhibited IκBα phosphorylation [146]; however, BAY117082 covalently attaches to the catalytic cysteine of UBC13 and UBCH7 [145]. This results in the inhibition of both K63-polyubiquitin and M1 linear-polyubiquitin chain formation in lymphoma cells, promoting cell death.

4.4. Targeting E3 Ligases

As the largest enzyme class involved in the ubiquitination cascade, over 700 E3 ligases are currently identified [41]. These are subdivided into at least three sub-classes, based on structural classification: the Really Interesting New Gene (RING) E3s, which function as a scaffold to bring ubiquitin-conjugated E2 enzymes in close contact with their substrates; Homologous to E6-AP Carboxyl Terminus (HECT) E3s, which catalyze the transfer of ubiquitin to their own cysteine residues before transfer to substrates, and a third subfamily, RING-Between-RING (RBR) E3s, which function as a hybrid between RING and HECT E3s [41].
As a large family of enzymes that use distinct catalytic mechanisms, the targeting of E3 ligases should offer a high degree of specificity, resulting in less toxicity and having greater therapeutic potential. Many E3 ligase inhibitors have been identified; here, we will focus on E3 ligases, which have been implicated in NFκB signalling.
As mention previously, MLN4924 can impact the function of the CRL sub-family of E3 ligases. CRLs play a critical role in NFκB activation [147]. The SCF E3 ligase complex containing CUL1 is the most well characterized to date. SCFβ-TrCP induces K48-polyubiquitination and the subsequent proteasomal degradation of IκBα, as well as the partial cleavage of the NFκB proteins p100 and p105 [34,148]. Furthermore, p100 is also a target for SCFFBXW7α [116]. The KEAP1-Cul3-RBX1 CRL E3 ligase complex, containing CUL3, induces IKKβ K48-polyubiquitination and proteasomal degradation [67]. MLN4924 inhibits the function of all cullin proteins; thus, inhibition of NAE could inhibit proliferation and induce cell death in HNSCC by inhibiting the NFκB pathway.
Of particular interest, the KEAP1, and CUL3 genes are among the most frequently mutated proteins in HNSCC [17,119]. Therefore, loss of function mutations in these three genes may contribute to the constitutive activation of NFκB in HNSCC and in other cancers [118]. This suggests that the inhibition of cullin activity via MLN4924 may function similarly to loss of function mutations observed in these genes in HNSCC patients and may have the potential to inhibit the NFκB pathway in these cancers.
The dual function of cIAP1 and 2 has made them an interesting potential therapeutic target in cancer by inhibiting NFκB and downstream inflammatory signalling and the induction of apoptosis though the extrinsic pathway [149]. Compounds targeting cIAPs, so called IAP antagonists, are mimicked after the natural IAP antagonist, Second Mitochondria-derived Activator of Caspases (SMAC)—thus, they are also called SMAC mimetics. Upon mitochondrial release, SMAC is cleaved and dimerizes. The N-terminal domain of SMAC contains a four amino acid sequence (AVPI; alanine (A), valine (V), proline (P), isoleucine (I)); this sequence binds to the BIR3 and BIR2 domain of IAPs [149]. IAP antagonists mimic this sequence and bind to IAPs. Upon binding, cIAP1/2 undergo a conformation change, autoubiquitination, and proteasomal degradation. In addition, some SMAC mimetics target XIAP, preventing it from binding to caspase 3, 7, and 9, thereby inducing caspase activation and apoptotic cell death [150]. The IAP antagonists developed thus far are monovalent or bivalent, based on how many BIR domains are bound by the IAP antagonist. The mechanism of action IAP antagonists is primarily the enhancement of TNFα-dependent apoptosis rather than inhibition of NFκB; despite the potential for canonical NFκB signalling inhibition, cIAP depletion results in stabilization of NIK, thereby activating non-canonical NFκB signalling [149]. This results in autocrine TNFα-signalling by tumor cells, as well as enhancing TNFα production in cytotoxic T lymphocytes. Altogether, increased TNFα in the microenvironment and the induction of caspases in tumor cells favors the induction of apoptotic and necroptotic cell death. IAP antagonist-induced apoptosis is also enhanced in combination with other death ligands, such as TRAIL [151].
A number of IAP antagonists are currently in clinical trials; four monovalent (Debio-1143, GDC-0917, LCL-161, and GDC-0152), two bivalent (birinapant and HGS-1029), and a synthetic based on the binding pocket rather than an AVPI peptidomimetic (ASTX-660). In HNSCC, several IAP antagonists have been shown to have activity in vitro. Birinapant and Smac-164 sensitize HNSCC cancer cells to standard chemotherapeutics, TNFα and TRAIL [152]. In addition, Debio-1143 and LCL161 sensitize HNSCC cells to radiation therapy [64,65]. Interestingly, HNSCC cell lines with amplification of the Fas-associated death domain (FADD), which mediates TNFR induced cell death, had an increased sensitivity to IAP antagonists and combination therapy with radiation, and resulted in an enhanced expression of TNFα [109]. Furthermore, IAP antagonists enhanced sensitivity to death ligands in HPV+ HNSCC that lack FADD copy gains. In some of these lines, IAP antagonists enhanced TP53 expression, which is targeted for degradation by HPV EIn these studies, ASTX660, in combination with TNFα and TRAIL, induced cell death in HNSCC cell lines and radiation-induced immunogenic death in pre-clinical models [151,153]. ASTX660 is currently in phase I trials in advanced solid tumors and lymphomas (Table 3; NCT02503423). Recently, Debio-1143 was granted breakthrough therapy designation by the Food and Drug Administration (FDA) to treat untreated, unresectable, and locally advanced HNSCC in combination with standard chemoradiation therapy. The phase II study results (Table 3; NCT02022098) presented at the ESMO Congress 2019 in Barcelona, Spain revealed a significant improvement for the primary endpoint locoregional control rate, 18 months after CRT (21% improvement vs. control arm) and a progression-free survival (PFS) benefit vs. the CRT + placebo arm after a 2-year follow-up period (HR = 0.37, p = 0.007). The drug showed a manageable safety profile, and that it did not compromise the full delivery of standard CRT. Therefore, these data suggest that IAP antagonists offer clinical benefit in HNSCC patients, particularly when the underlying genetics or HPV status of a tumor are known, making them an attractive target in personalized medicine. Currently, Debio 1143 is in phase I/II trials in combination with nivolumab, an anti-PD-1 monoclonal antibody approved for treating a number of cancers, including HNSCC (Table 3; NCT04122625).

4.5. Targeting Deubiquitinase

Like E3 ligases, several deubiquitinases are deregulated and implicated in cancer development [154]. Due to their well-defined active site, deubiquitinases are currently gaining much interest as potential drug targets. Among all known deubiquitinases, USP7 inhibitors are the most advanced due to its critical role in regulating TP53 function by deubiquitinating the TP53 negative regulator MDM2 [155]. Furthermore, USP7 directly deubiquitinates RELA/p65, promoting NFκB signalling [156]. The early USP7 inhibitors P5091 and HBX19818 were modestly efficacious, fairly selective, and induced TP53-dependent cell death in vivo [157]. Importantly, P5091 induced apoptosis in MM patient cells, including those resistant to prior treatments such as bortezomib [157]. More recently, highly potent and specific USP7 inhibitors demonstrated high efficacy in vitro and MM xenografts in mice [158]. Despite these positive studies, USP7 inhibitors may only stabilize wild-type TP53, whereas the majority of tumors harbor mutant TP53; therefore, the clinical efficacy of these inhibitors will depend on the genetic background of the tumor.
The removal of ubiquitin chains from target proteins during proteasomal degradation is performed by proteasome-associated deubiquitinases, including UCHL5 and USP14 [159]. These deubiquitinases trim polyubiquitin chains to allow substrate proteins entry into the proteasome for degradation. The inhibitor b-AP15 targets both UCHL5 and USP14 and leads to the accumulation of ubiquitinated substrates [160]. Furthermore, due to its effects on proteasomal degradation, b-AP15 can result in the inhibition of NFκB signalling [161]. b-AP15 demonstrated exhibited excellent efficacy in different in vivo solid tumor models [160]. VLX1570, a b-AP15 analogue with higher potency and higher selectivity for USP14, demonstrated anti-tumor activity, and enhanced survival in MM xenografts [162]. However, a recently reported Phase 1 study of VLX1570 in multiple myeloma patients showed that despite some preliminary anti-tumor effects, treatment resulted in severe toxicity in several patients, similar to previous observations after treatment with proteasome inhibitors [163]. USP14 has also been shown to regulate NFκB via the deubiquitination of IκBα [164]. More selective USP14 inhibitors are now available; IU1, which inhibits USP14 but not UCHL5, led to tumor regression in a TP53-deficient mouse tumor model [165].

5. Conclusions

Many of the ubiquitin activating and proteasomal degradation steps hold great potential as drug targets. However, they also suffer from issues of low specificity. In contrast, advances in computational chemistry and novel technologies, including mass spectrometry and high-throughput screening, the development and success of specific E3 ligase and deubiquitinase inhibitors with more selective anti-cancer activity may be possible. These new technologies may enable the development of innovative therapeutic approaches that target protein-protein interactions, or co-opt the ubiquitin system, as is the case with protein-targeting chimeric molecules, which bind to protein and induce their proteasomal degradation (PROTACs) [166]. It is clear that the NFκB pathway is essential for the proliferation and survival of HNSCC, in vitro and in vivo. Furthermore, targeting the ubiquitin system is an attractive therapeutic target in HNSCC due to the potential to target the NFκB pathway. The use of proteasome inhibitors has demonstrated that this strategy has clinical benefit in multiple myeloma, but toxicity, lack of specificity, and resistance mechanisms emerged as a challenge in HNSCC. Thus, further research into the role of specific enzymes of the ubiquitin system and how they regulate the NFκB pathway may offer novel therapeutic strategies in HNSCC.

Funding

This research was supported by the Intramural Research Program of the NIH, NIDCD. ELM, ZC and CVW are supported by the NIDCD intramural project Z01-DC-00016.

Acknowledgments

We thank Clint Allen, MD (Translational Tumor Immunology Program, Head and Neck Surgery Branch, NIDCD, NIH) and Vassiliki Saloura, MD, PhD (Thoracic and GI Malignancies Branch, NCI) for critical reading of this review and providing helpful suggestions.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Chow, L.Q.M. Head and neck cancer. N. Engl. J. Med. 2020, 382, 60–72. [Google Scholar] [CrossRef] [PubMed]
  2. Cramer, J.D.; Burtness, B.; Le, Q.T.; Ferris, R.L. The changing therapeutic landscape of head and neck cancer. Nat. Rev. Clin. Oncol. 2019, 16, 669–683. [Google Scholar] [CrossRef] [PubMed]
  3. Beckham, T.H.; Barney, C.; Healy, E.; Wolfe, A.R.; Branstetter, A.; Yaney, A.; Riaz, N.; McBride, S.M.; Tsai, C.J.; Kang, J.; et al. Platinum-based regimens versus cetuximab in definitive chemoradiation for human papillomavirus-unrelated head and neck cancer. Int. J. Cancer 2020, 147, 107–115. [Google Scholar] [CrossRef] [PubMed]
  4. Bauman, J.E.; Ohr, J.; Gooding, W.E.; Ferris, R.L.; Duvvuri, U.; Kim, S.; Johnson, J.T.; Soloff, A.C.; Wallweber, G.; Winslow, J.; et al. Phase I Study of Ficlatuzumab and Cetuximab in Cetuximab-Resistant, Recurrent/Metastatic Head and Neck Cancer. Cancers 2020, 12, 1537. [Google Scholar] [CrossRef]
  5. Powell, S.F.; Gold, K.A.; Gitau, M.M.; Sumey, C.J.; Lohr, M.M.; McGraw, S.C.; Nowak, R.K.; Jensen, A.W.; Blanchard, M.J.; Fischer, C.D.; et al. Safety and Efficacy of Pembrolizumab With Chemoradiotherapy in Locally Advanced Head and Neck Squamous Cell Carcinoma: A Phase IB Study. J. Clin. Oncol. 2020, 1, 190315. [Google Scholar] [CrossRef]
  6. Leemans, C.R.; Braakhuis, B.J.M.; Brakenhoff, R.H. Response to correspondence on the molecular biology of head and neck cancer. Nat. Rev. Cancer 2011, 11, 382. [Google Scholar] [CrossRef] [Green Version]
  7. Kobayashi, K.; Hisamatsu, K.; Suzui, N.; Hara, A.; Tomita, H.; Miyazaki, T. A Review of HPV-Related Head and Neck Cancer. J. Clin. Med. 2018, 7, 241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Chaturvedi, A.K.; Engels, E.A.; Pfeiffer, R.M.; Hernandez, B.Y.; Xiao, W.; Kim, E.; Jiang, B.; Goodman, M.T.; Sibug-Saber, M.; Cozen, W.; et al. Human Papillomavirus and Rising Oropharyngeal Cancer Incidence in the United States. J. Clin. Oncol. 2011, 29, 4294–4301. [Google Scholar] [CrossRef] [PubMed]
  9. Lowy, D.R.; Schiller, J.T. Reducing HPV-associated cancer globally. Cancer Prev. Res. 2012, 5, 18–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Chaturvedi, A.K.; D’Souza, G.; Gillison, M.L.; Katki, H.A. Burden of HPV-positive oropharynx cancers among ever and never smokers in the U.S. population. Oral Oncol. 2016, 60, 61–67. [Google Scholar] [CrossRef] [PubMed]
  11. Gillison, M.L.; Koch, W.M.; Capone, R.B.; Spafford, M.; Westra, W.H.; Wu, L.; Zahurak, M.L.; Daniel, R.W.; Viglione, M.; Symer, D.E.; et al. Evidence for a causal association between human papillomavirus and a subset of head and neck cancers. J. Natl. Cancer Inst. 2000, 92, 709–720. [Google Scholar] [CrossRef] [PubMed]
  12. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Walch-Rückheim, B.; Mavrova, R.; Henning, M.; Vicinus, B.; Kim, Y.J.; Bohle, R.M.; Juhasz-Böss, I.; Solomayer, E.F.; Smola, S. Stromal fibroblasts induce CCL20 through IL6/C/EBPβ to support the recruitment of Th17 cells during cervical cancer progression. Cancer Res. 2015, 15, 5248–5259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Yang, X.; Cheng, H.; Chen, J.; Wang, R.; Saleh, A.; Si, H.; Lee, S.; Guven-Maiorov, E.; Keskin, O.; Gursoy, A.; et al. Head and Neck Cancers Promote an Inflammatory Transcriptome through Coactivation of Classic and Alternative NF-κB Pathways. Cancer Immunol. Res. 2019, 7, 1760–1774. [Google Scholar] [CrossRef]
  15. Yadav, A.; Kumar, B.; Datta, J.; Teknos, T.N.; Kumar, P. IL-6 promotes head and neck tumor metastasis by inducing epithelial-mesenchymal transition via the JAK-STAT3-SNAIL signaling pathway. Mol. Cancer Res. 2011, 9, 1658–1667. [Google Scholar] [CrossRef] [Green Version]
  16. Gao, J.; Zhao, S.; Halstensen, T.S. Increased interleukin-6 expression is associated with poor prognosis and acquired cisplatin resistance in head and neck squamous cell carcinoma. Oncol. Rep. 2016, 35, 3265–3274. [Google Scholar] [CrossRef] [Green Version]
  17. The Cancer Genome Atlas Network. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nat. Cell Biol. 2015, 517, 576–582. [Google Scholar] [CrossRef] [Green Version]
  18. Xia, Y.; Shen, S.; Verma, I.M. NF- B, an Active Player in Human Cancers. Cancer Immunol. Res. 2014, 2, 823–830. [Google Scholar] [CrossRef] [Green Version]
  19. Morgan, E.L.; Macdonald, A. Autocrine STAT3 activation in HPV positive cervical cancer through a virus-driven Rac1—NFκB—IL-6 signalling axis. PLoS Pathog. 2019, 15, e1007835. [Google Scholar] [CrossRef] [Green Version]
  20. An, J.; Mo, D.; Liu, H.; Veena, M.S.; Srivatsan, E.S.; Massoumi, R.; Rettig, M.B. Inactivation of the CYLD Deubiquitinase by HPV E6 Mediates Hypoxia-Induced NF-κB Activation. Cancer Cell 2008, 14, 394–407. [Google Scholar] [CrossRef] [Green Version]
  21. Jost, P.J.; Ruland, J. Aberrant NF-κB signaling in lymphoma: Mechanisms, consequences, and therapeutic implications. Blood 2006, 109, 2700–2707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Allen, C.; Saigal, K.; Nottingham, L.; Arun, P.; Chen, Z.; Van Waes, C. Bortezomib-induced apoptosis with limited clinical response is accompanied by inhibition of canonical but not alternative nuclear factor-{kappa}B subunits in head and neck cancer. Clin. Cancer Res. 2008, 1, 4175–4185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Liu, S.; Chen, Z.J. Expanding role of ubiquitination in NF-κB signaling. Cell Res. 2010, 21, 6–21. [Google Scholar] [CrossRef] [PubMed]
  24. Karin, M. How NF-kappaB is activated: The role of the IkappaB kinase (IKK) complex. Oncogene 1999, 22, 6867–6874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Liou, H.; Nolan, G.; Ghosh, S.; Fujita, T.; Baltimore, D. The NF-kappa B p50 precursor, p105, contains an internal I kappa B-like inhibitor that preferentially inhibits p50. EMBO J. 1992, 11, 3003–3009. [Google Scholar] [CrossRef]
  26. Beg, A.A.; Ruben, S.M.; Scheinman, R.I.; Haskill, S.; Rosen, C.A.; Baldwin, A.S. I kappa B interacts with the nuclear localization sequences of the subunits of NF-kappa B: A mechanism for cytoplasmic retention. Genes Dev. 1992, 6, 1899–1913. [Google Scholar] [CrossRef] [Green Version]
  27. Tong, X.; Yin, L.; Washington, R.; Rosenberg, D.W.; Giardina, C. The p50-p50 NF-κB complex as a stimulus-specific repressor of gene activation. Mol. Cell. Biochem. 2004, 265, 171–183. [Google Scholar] [CrossRef]
  28. Chen, L.-F.; Mu, Y.; Greene, W.C. Acetylation of RelA at discrete sites regulates distinct nuclear functions of NF-kappaB. EMBO J. 2002, 21, 6539–6548. [Google Scholar] [CrossRef] [Green Version]
  29. Oeckinghaus, A.; Ghosh, S. The NF-kappaB family of transcription factors and its regulation. Cold Spring Harb Perspect Biol. 2009, 1, a000034. [Google Scholar] [CrossRef]
  30. Zhang, Q.; Didonato, J.A.; Karin, M.; McKeithan, T.W. BCL3 encodes a nuclear protein which can alter the subcellular location of NF-kappa B proteins. Mol. Cell. Biol. 1994, 14, 3915–3926. [Google Scholar] [CrossRef] [Green Version]
  31. Shih, V.F.-S.; Tsui, R.; Caldwell, A.; Hoffmann, A. A single NFκB system for both canonical and non-canonical signaling. Cell Res. 2010, 21, 86–102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Wang, G.; Gao, Y.; Li, L.; Jin, G.; Cai, Z.; Chao, J.-I.; Lin, H.-K. K63-Linked Ubiquitination in Kinase Activation and Cancer. Front. Oncol. 2012, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Wang, C.; Deng, L.; Hong, M.; Akkaraju, G.R.; Inoue, J.-I.; Chen, Z.J. TAK1 is a ubiquitin-dependent kinase of MKK and IKK. Nat. Cell Biol. 2001, 412, 346–351. [Google Scholar] [CrossRef] [PubMed]
  34. Winston, J.T.; Strack, P.; Beer-Romero, P.; Chu, C.Y.; Elledge, S.J.; Harper, J.W. The SCFbeta-TRCP-ubiquitin ligase complex associates specifically with phosphorylated destruction motifs in IkappaBalpha and beta-catenin and stimulates IkappaBalpha ubiquitination in vitro. Genes Dev. 1999, 13, 270–283. [Google Scholar] [CrossRef] [PubMed]
  35. Li, X.; Commane, M.; Jiang, Z.; Stark, G.R. IL-1-induced NFkappa B and c-Jun N-terminal kinase (JNK) activation diverge at IL-1 receptor-associated kinase (IRAK). Proc. Natl. Acad. Sci. USA 2001, 98, 4461–4465. [Google Scholar] [CrossRef] [Green Version]
  36. Cui, W.; Xiao, N.; Xiao, H.; Zhou, H.; Yu, M.; Gu, J.; Li, X. β-TrCP-Mediated IRAK1 Degradation Releases TAK1-TRAF6 from the Membrane to the Cytosol for TAK1-Dependent NF-κB Activation. Mol. Cell. Biol. 2012, 32, 3990–4000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Sun, S.-C. Non-canonical NF-κB signaling pathway. Cell Res. 2010, 21, 71–85. [Google Scholar] [CrossRef] [Green Version]
  38. Zarnegar, B.J.; Wang, Y.; Mahoney, D.J.; Dempsey, P.W.; Cheung, H.H.; He, J.; Shiba, T.; Yang, X.; Yeh, W.-C.; Mak, T.W.; et al. Noncanonical NF-κB activation requires coordinated assembly of a regulatory complex of the adaptors cIAP1, cIAP2, TRAF2 and TRAF3 and the kinase NIK. Nat. Immunol. 2008, 9, 1371–1378. [Google Scholar] [CrossRef] [Green Version]
  39. Clague, M.J.; Urbé, S. Ubiquitin: Same Molecule, Different Degradation Pathways. Cell 2010, 143, 682–685. [Google Scholar] [CrossRef] [Green Version]
  40. Komander, D.; Clague, M.J.; Urbé, S. Breaking the chains: Structure and function of the deubiquitinases. Nat. Rev. Mol. Cell Biol. 2009, 10, 550–563. [Google Scholar] [CrossRef]
  41. Buetow, L.; Huang, D.T. Structural insights into the catalysis and regulation of E3 ubiquitin ligases. Nat. Rev. Mol. Cell Biol. 2016, 17, 626–642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Walden, M.; Masandi, S.K.; Pawłowski, K.; Zeqiraj, E. Pseudo-DUBs as allosteric activators and molecular scaffolds of protein complexes. Biochem. Soc. Trans. 2018, 46, 453–466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Swatek, K.N.; Komander, D. Ubiquitin modifications. Cell Res. 2016, 26, 399–422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Pan, M.-R.; Peng, G.; Hung, W.-C.; Lin, S.-Y. Monoubiquitination of H2AX Protein Regulates DNA Damage Response Signaling. J. Biol. Chem. 2011, 286, 28599–28607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Thrower, J.S.; Hoffman, L.; Rechsteiner, M.; Pickart, C.M. Recognition of the polyubiquitin proteolytic signal. EMBO J. 2000, 19, 94–102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Mukhopadhyay, D.; Riezman, H. Proteasome-Independent Functions of Ubiquitin in Endocytosis and Signaling. Science 2007, 315, 201–205. [Google Scholar] [CrossRef] [Green Version]
  47. Michel, M.A.; Swatek, K.N.; Hospenthal, M.K.; Komander, D. Ubiquitin Linkage-Specific Affimers Reveal Insights into K6-Linked Ubiquitin Signaling. Mol. Cell 2017, 68, 233–246.e5. [Google Scholar] [CrossRef] [Green Version]
  48. Gatti, M.; Pinato, S.; Maiolica, A.; Rocchio, F.; Prato, M.G.; Aebersold, R.; Penengo, L. RNF168 Promotes Noncanonical K27 Ubiquitination to Signal DNA Damage. Cell Rep. 2015, 10, 226–238. [Google Scholar] [CrossRef] [Green Version]
  49. Kristariyanto, Y.A.; Rehman, S.A.A.; Campbell, D.G.; Morrice, N.A.; Johnson, C.; Toth, R.; Kulathu, Y. K29-selective ubiquitin binding domain reveals structural basis of specificity and heterotypic nature of k29 polyubiquitin. Mol. Cell 2015, 58, 83–94. [Google Scholar] [CrossRef] [Green Version]
  50. Yuan, W.-C.; Lee, Y.-R.; Lin, S.-Y.; Chang, L.-Y.; Tan, Y.P.; Hung, C.-C.; Kuo, J.-C.; Liu, C.-H.; Lin, M.-Y.; Xu, M.; et al. K33-Linked Polyubiquitination of Coronin 7 by Cul3-KLHL20 Ubiquitin E3 Ligase Regulates Protein Trafficking. Mol. Cell 2014, 54, 586–600. [Google Scholar] [CrossRef] [Green Version]
  51. Xu, P.; Duong, D.M.; Seyfried, N.T.; Cheng, D.; Xie, Y.; Robert, J.; Rush, J.; Hochstrasser, M.; Finley, D.; Peng, J. Quantitative Proteomics Reveals the Function of Unconventional Ubiquitin Chains in Proteasomal Degradation. Cell 2009, 137, 133–145. [Google Scholar] [CrossRef] [Green Version]
  52. Dynek, J.N.; Goncharov, T.; Dueber, E.C.; Fedorova, A.V.; Izrael-Tomasevic, A.; Phu, L.; Helgason, E.; Fairbrother, W.J.; Deshayes, K.; Kirkpatrick, D.S.; et al. c-IAP1 and UbcH5 promote K11-linked polyubiquitination of RIP1 in TNF signalling. EMBO J. 2010, 29, 4198–4209. [Google Scholar] [CrossRef]
  53. Haas, T.L.; Emmerich, C.H.; Gerlach, B.; Schmukle, A.C.; Cordier, S.M.; Rieser, E.; Feltham, R.; Vince, J.; Warnken, U.; Wenger, T.; et al. Recruitment of the Linear Ubiquitin Chain Assembly Complex Stabilizes the TNF-R1 Signaling Complex and Is Required for TNF-Mediated Gene Induction. Mol. Cell 2009, 36, 831–844. [Google Scholar] [CrossRef] [PubMed]
  54. Meyer, H.-J.; Rape, M. Enhanced Protein Degradation by Branched Ubiquitin Chains. Cell 2014, 157, 910–921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Ohtake, F.; Saeki, Y.; Ishido, S.; Kanno, J.; Tanaka, K. The K48-K63 Branched Ubiquitin Chain Regulates NF-κB Signaling. Mol. Cell 2016, 64, 251–266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Ohtake, F.; Tsuchiya, H.; Saeki, Y.; Tanaka, K. K63 ubiquitylation triggers proteasomal degradation by seeding branched ubiquitin chains. Proc. Natl. Acad. Sci. USA 2018, 115, E1401–E1408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Ritorto, M.S.; Ewan, R.; Perez-Oliva, A.B.; Knebel, A.; Buhrlage, S.J.; Wightman, M.; Kelly, S.M.; Wood, N.T.; Virdee, S.; Gray, N.S.; et al. Screening of DUB activity and specificity by MALDI-TOF mass spectrometry. Nat. Commun. 2014, 5, 4763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. O’Donnell, M.A.; Legarda-Addison, D.; Skountzos, P.; Yeh, W.-C.; Ting, A.T. Ubiquitination of RIP1 regulates an NF-kappaB-independent cell-death switch in TNF signalling. Curr. Biol. 2007, 17, 418–424. [Google Scholar] [CrossRef] [Green Version]
  59. Wang, Y.; Shan, B.; Liang, Y.; Wei, H.; Yuan, J. Parkin regulates NF-κB by mediating site-specific ubiquitination of RIPK. Cell Death Dis. 2018, 9, 732. [Google Scholar] [CrossRef] [PubMed]
  60. Kanayama, A.; Seth, R.B.; Sun, L.; Ea, C.K.; Hong, M.; Shaito, A.; Chiu, Y.H.; Deng, L.; Chen, Z.J. TAB2 and TAB3 Activate the NF-κB Pathway through Binding to Polyubiquitin Chains. Mol. Cell 2004, 15, 535–548. [Google Scholar] [CrossRef]
  61. Laplantine, E.; Fontan, E.; Chiaravalli, J.; Lopez, T.; Lakisic, G.; Veron, M.; Agou, F.; Israel, A. NEMO specifically recognizes K63-linked poly-ubiquitin chains through a new bipartite ubiquitin-binding domain. EMBO J. 2009, 28, 2885–2895. [Google Scholar] [CrossRef]
  62. Ordureau, A.; Smith, H.; Windheim, M.; Peggie, M.; Carrick, E.; Morrice, N.; Cohen, P. The IRAK-catalysed activation of the E3 ligase function of Pellino isoforms induces the Lys63-linked polyubiquitination of IRAK. Biochem. J. 2007, 409, 43–52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Spit, M.; Rieser, E.; Walczak, H. Linear ubiquitination at a glance. J. Cell Sci. 2019, 132, jcs208512. [Google Scholar] [CrossRef] [Green Version]
  64. Arimoto, K.-I.; Funami, K.; Saeki, Y.; Tanaka, K.; Okawa, K.; Takeuchi, O.; Akira, S.; Murakami, Y.; Shimotohno, K. Polyubiquitin conjugation to NEMO by triparite motif protein 23 (TRIM23) is critical in antiviral defense. Proc. Natl. Acad. Sci. USA 2010, 107, 15856–15861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Li, X.; Yang, Y.; Ashwell, J.D. TNF-RII and c-IAP1 mediate ubiquitination and degradation of TRAF. Nat. Cell Biol. 2002, 416, 345–347. [Google Scholar] [CrossRef] [Green Version]
  66. Habelhah, H.; Frew, I.J.; Laine, A.; Janes, P.W.; Relaix, F.; Sassoon, D.; Bowtell, D.D.; Ronai, Z. Stress-induced decrease in TRAF2 stability is mediated by Siah2. EMBO J. 2002, 21, 5756–5765. [Google Scholar] [CrossRef] [PubMed]
  67. Lee, D.-F.; Kuo, H.-P.; Liu, M.; Chou, C.-K.; Xia, W.; Du, Y.; Shen, J.; Chen, C.-T.; Huo, L.; Hsu, M.-C.; et al. KEAP1 E3 Ligase-Mediated Downregulation of NF-κB Signaling by Targeting IKKβ. Mol. Cell 2009, 36, 131–140. [Google Scholar] [CrossRef] [Green Version]
  68. Ahmed, N.; Zeng, M.; Sinha, I.; Polin, L.; Wei, W.-Z.; Rathinam, C.; Flavell, R.; Massoumi, R.; Venuprasad, K. The E3 ligase Itch and deubiquitinase Cyld act together to regulate Tak1 and inflammation. Nat. Immunol. 2011, 12, 1176–1183. [Google Scholar] [CrossRef] [Green Version]
  69. Zotti, T.; Uva, A.; Ferravante, A.; Vessichelli, M.; Scudiero, I.; Ceccarelli, M.; Vito, P. TRAF7 protein promotes Lys-29-linked polyubiquitination of IκB kinase (IKKγ)/NF-κB essential modulator (NEMO) and p65/RelA protein and represses NF-κB activation. J. Biol. Chem. 2011, 286, 22924–22933. [Google Scholar] [CrossRef] [Green Version]
  70. Coles, A.H.; Gannon, H.; Cerny, A.; Kurt-Jones, E.; Jones, S.N. Inhibitor of growth-4 promotes IkappaB promoter activation to suppress NF-kappaB signalling and innate immunity. Proc. Natl. Acad. Sci. USA 2010, 107, 11423–11428. [Google Scholar] [CrossRef] [Green Version]
  71. Strebovsky, J.; Walker, P.; Lang, R.; Dalpke, A.H. Suppressor of cytokine signalling 1 (SOCS1) limits NFkappaB signalling by decreasing p65 stability within the cell nucleus. FASEB J. 2011, 25, 863–874. [Google Scholar] [CrossRef] [PubMed]
  72. Hou, Y.; Moreau, F.; Chadee, K. PPARγ is an E3 ligase that induces the degradation of NFκB/p. Nat. Commun. 2012, 3, 1300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Tanaka, T.; Grusby, M.J.; Kaisho, T. PDLIM2-mediated termination of transcription factor NF-kappaB activation by intranuclear sequestration and degradation of the p65 subunit. Nat. Immunol. 2007, 8, 584–591. [Google Scholar] [CrossRef] [PubMed]
  74. Hymowitz, S.G.; Wertz, I.E. A20: From ubiquitin editing to tumour suppression. Nat. Rev. Cancer 2010, 10, 332–341. [Google Scholar] [CrossRef]
  75. Massoumi, R. CYLD: A deubiquitination enzyme with multiple roles in cancer. Futur. Oncol. 2011, 7, 285–297. [Google Scholar] [CrossRef]
  76. Werner, S.L.; Kearns, J.D.; Zadorozhnaya, V.; Lynch, C.; O’Dea, E.; Boldin, M.; Ma, A.; Baltimore, D.; Hoffmann, A. Encoding NF- B temporal control in response to TNF: Distinct roles for the negative regulators I B and A. Genes Dev. 2008, 22, 2093–2101. [Google Scholar] [CrossRef] [Green Version]
  77. Shembade, N.; Harhaj, N.S.; Liebl, D.J.; Harhaj, E.W. Essential role for TAX1BP1 in the termination of TNF-alpha-, IL-1- and LPS-mediated NF-kappaB and JNK signalling. EMBO J. 2007, 26, 3910–3922. [Google Scholar] [CrossRef] [Green Version]
  78. Bellail, A.C.; Olson, J.J.; Yang, X.; Chen, Z.J.; Hao, C. A20 ubiquitin ligase-mediated polyubiquitination of RIP1 inhibits caspase-8 cleavage and TRAIL-induced apoptosis in glioblastoma. Cancer Discov. 2012, 2, 140–155. [Google Scholar] [CrossRef] [Green Version]
  79. Shembade, N.; Ma, A.; Harhaj, E.W. Inhibition of NF-kappaB signalling by A20 through disruption of ubiquitin enzyme complexes. Science 2010, 327, 1135–1139. [Google Scholar] [CrossRef] [Green Version]
  80. Skaug, B.; Chen, J.; Du, F.; He, J.; Ma, A.; Chen, Z.J. Direct, Noncatalytic Mechanism of IKK Inhibition by A. Mol. Cell 2011, 44, 559–571. [Google Scholar] [CrossRef] [Green Version]
  81. Verhelst, K.; Carpentier, I.; Kreike, M.; Meloni, L.; Verstrepen, L.; Kensche, T.; Dikic, I.; Beyaert, R. A20 inhibits LUBAC-mediated NF-κB activation by binding linear polyubiquitin chains via its zinc finger. EMBO J. 2012, 31, 3845–3855. [Google Scholar] [CrossRef] [PubMed]
  82. Hou, X.; Wang, L.; Zhang, L.; Pan, X.; Zhao, W. Ubiquitin-specific protease 4 promotes TNF-α-induced apoptosis by deubiquitination of RIP1 in head and neck squamous cell carcinoma. FEBS Lett. 2013, 587, 311–316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Jean-Charles, P.-Y.; Zhang, L.; Wu, J.-H.; Han, S.-O.; Brian, L.; Freedman, N.J.; Shenoy, S.K. Ubiquitin-specific Protease 20 Regulates the Reciprocal Functions of β-Arrestin2 in Toll-like Receptor 4-promoted Nuclear Factor κB (NFκB) Activation*. J. Biol. Chem. 2016, 291, 7450–7464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Xu, G.; Tan, X.; Wang, H.; Sun, W.; Shi, Y.; Burlingame, S.; Gu, X.; Cao, G.; Zhang, T.; Qin, J.; et al. Ubiquitin-specific Peptidase 21 Inhibits Tumor Necrosis Factor α-induced Nuclear Factor κB Activation via Binding to and Deubiquitinating Receptor-interacting Protein. J. Biol. Chem. 2009, 285, 969–978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Goncharov, T.; Niessen, K.; De Almagro, M.C.; Izrael-Tomasevic, A.; Fedorova, A.V.; Varfolomeev, E.; Arnott, D.; Deshayes, K.; Kirkpatrick, D.S.; Vucic, D. OTUB1 modulates c-IAP1 stability to regulate signalling pathways. EMBO J. 2013, 32, 1103–1114. [Google Scholar] [CrossRef] [PubMed]
  86. Lee, E.-W.; Seong, D.; Seo, J.; Jeong, M.; Lee, H.-K.; Song, J. USP11-dependent selective cIAP2 deubiquitylation and stabilization determine sensitivity to Smac mimetics. Cell Death Differ. 2015, 22, 1463–1476. [Google Scholar] [CrossRef] [Green Version]
  87. Hu, H.; Brittain, G.C.; Chang, J.-H.; Puebla-Osorio, N.; Jin, J.; Zal, A.; Xiao, Y.; Cheng, X.; Chang, M.; Fu, Y.-X.; et al. OTUD7B controls non-canonical NF-κB activation through deubiquitination of TRAF. Nat. Cell Biol. 2013, 494, 371–374. [Google Scholar] [CrossRef] [Green Version]
  88. Colleran, A.; Collins, P.E.; O’Carroll, C.; Ahmed, A.; Mao, X.; McManus, B.; Kiely, P.A.; Burstein, E.; Carmody, R.J. Deubiquitination of NF-κB by Ubiquitin-Specific Protease-7 promotes transcription. Proc. Natl. Acad. Sci. USA 2012, 110, 618–623. [Google Scholar] [CrossRef] [Green Version]
  89. Schweitzer, K.; Naumann, M. CSN-associated USP48 confers stability to nuclear NF-κB/RelA by trimming K48-linked Ub-chains. Biochim. Biophys. Acta (BBA)-Bioenerg. 2015, 1853, 453–469. [Google Scholar] [CrossRef] [Green Version]
  90. Li, S.; Wang, D.; Zhao, J.; Weathington, N.M.; Shang, D.; Zhao, Y. The deubiquitinating enzyme USP48 stabilizes TRAF2 and reduces E-cadherin-mediated adherens junctions. FASEB J. 2017, 32, 230–242. [Google Scholar] [CrossRef] [Green Version]
  91. Jiang, B.; Shen, H.; Chen, Z.; Yin, L.; Zan, L.; Rui, L. Carboxyl Terminus of HSC70-interacting Protein (CHIP) Down-regulates NF-κB-inducing Kinase (NIK) and Suppresses NIK-induced Liver Injury*. J. Biol. Chem. 2015, 290, 11704–11714. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Jin, X.; Jin, H.R.; Jung, H.S.; Lee, S.J.; Lee, J.-H.; Lee, J.J. An atypical E3 ligase zinc finger protein 91 stabilizes and activates NF-kappaB-inducing kinase via Lys63-linked ubiquitination. J. Biol. Chem. 2010, 285, 30539–30547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Karin, M.; Cao, Y.; Greten, F.R.; Li, Z.-W. NF-kappaB in cancer: From innocent bystander to major culprit. Nat. Rev. Cancer 2002, 2, 301–310. [Google Scholar] [CrossRef] [PubMed]
  94. Karin, M. NF-kappaB as a critical link between inflammation and cancer. Cold Spring Harb. Perspect. Biol. 2009, 1, a000141. [Google Scholar] [CrossRef] [PubMed]
  95. Gaykalova, D.A.; Manola, J.B.; Ozawa, H.; Zizkova, V.; Morton, K.; Bishop, J.A.; Sharma, R.; Zhang, C.; Michailidi, C.; Considine, M.; et al. NF-κB and stat3 transcription factor signatures differentiate HPV-positive and HPV-negative head and neck squamous cell carcinoma. Int. J. Cancer 2015, 137, 1879–1889. [Google Scholar] [CrossRef] [PubMed]
  96. Giopanou, I.; Lilis, I.; Papadaki, H.; Papadas, T.; Stathopoulos, G.T. A link between RelB expression and tumor progression in laryngeal cancer. Oncotarget 2017, 8, 114019–114030. [Google Scholar] [CrossRef] [Green Version]
  97. Vageli, D.P.; Doukas, S.G.; Sasaki, C.T. Inhibition of NF-κB prevents the acidic bile-induced oncogenic mRNA phenotype, in human hypopharyngeal cells. Oncotarget 2017, 9, 5876–5891. [Google Scholar] [CrossRef] [Green Version]
  98. Gupta, S.; Kumar, P.; Kaur, H.; Sharma, N.; Gupta, S.; Saluja, D.; Bharti, A.C.; Das, B. Constitutive activation and overexpression of NF-κB/c-Rel in conjunction with p50 contribute to aggressive tongue tumorigenesis. Oncotarget 2018, 9, 33011–33029. [Google Scholar] [CrossRef] [Green Version]
  99. Sun, Z.-J.; Chen, G.; Hu, X.; Zhang, W.; Liu, Y.; Zhu, L.-X.; Zhou, Q.; Zhao, Y.-F. Activation of PI3K/Akt/IKK-alpha/NF-kappaB signalling pathway is required for the apoptosis-evasion in human salivary adenoid cystic carcinoma: Its inhibition by quercetin. Apoptosis 2010, 15, 850–863. [Google Scholar] [CrossRef]
  100. Yao, X.; Wang, Y.; Duan, Y.; Zhang, Q.; Li, P.; Jin, R.; Tao, Y.; Zhang, W.; Wang, X.; Jing, C.; et al. IGFBP2 promotes salivary adenoid cystic carcinoma metastasis by activating cademia and industry. Signal Transduct. Target. Ther. 2019, 4, 38–46. [Google Scholar]
  101. Li, Y.Y.; Chung, G.T.; Lui, V.W.; To, K.F.; Ma, B.B.; Chow, C.; John, K.; Woo, S.; Yip, K.Y.; Seo, J.; et al. Exome and genome sequencing of nasopharynx cancer identifies NF-κB pathway activating mutations. Nat. Commun. 2017, 8, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Duffey, D.C.; Crowl-Bancroft, C.V.; Chen, Z.; Ondrey, F.G.; Nejad-Sattari, M.; Dong, G.; Van Waes, C. Inhibition of transcription factor nuclear factor-κB by a mutant inhibitor-κBα attenuates resistance of human head and neck squamous cell carcinoma to TNF-α caspase-mediated cell death. Br. J. Cancer 2000, 83, 1367–1374. [Google Scholar] [CrossRef] [PubMed]
  103. Yang, X.; Lu, H.; Yan, B.; Romano, R.-A.; Bian, Y.; Friedman, J.; Duggal, P.; Allen, C.; Chuang, R.; Ehsanian, R.; et al. ΔNp63 versatilely regulates a Broad NF-κB gene program and promotes squamous epithelial proliferation, migration, and inflammation. Cancer Res. 2011, 71, 3688–3700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Nottingham, L.K.; Yan, C.H.; Yang, X.; Si, H.; Coupar, J.; Bian, Y.; Cheng, T.-F.; Allen, C.; Arun, P.; Gius, D.; et al. Aberrant IKKα and IKKβ cooperatively activate NF-κB and induce EGFR/AP1 signaling to promote survival and migration of head and neck cancer. Oncogene 2013, 33, 1135–1147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Zhang, J.; Chen, T.; Yang, X.; Cheng, H.; Späth, S.S.; Clavijo, P.E.; Chen, J.; Silvin, C.; Issaeva, N.; Su, X.; et al. Attenuated TRAF3 Fosters Activation of Alternative NF-κB and Reduced Expression of Antiviral Interferon, TP53, and RB to Promote HPV-Positive Head and Neck Cancers. Cancer Res. 2018, 78, 4613–4626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Das, R.; Coupar, J.; Clavijo, P.E.; Saleh, A.; Cheng, T.-F.; Yang, X.; Chen, J.; Van Waes, C.; Chen, Z. Lymphotoxin-β receptor-NIK signaling induces alternative RELB/NF-κB2 activation to promote metastatic gene expression and cell migration in head and neck cancer. Mol. Carcinog. 2018, 58, 411–425. [Google Scholar] [CrossRef] [PubMed]
  107. Sriuranpong, V.; Park, J.I.; Amornphimoltham, P.; Patel, V.; Nelkin, B.D.; Gutkind, J.S. Epidermal growth factor receptor-independent constitutive activation of STAT3 in head and neck squamous cell carcinoma is mediated by the autocrine/paracrine stimulation of the interleukin 6/gp130 cytokine system. Cancer Res. 2003, 63, 2948–2956. [Google Scholar]
  108. Squarize, C.H.; Castilho, R.M.; Sriuranpong, V.; Pinto, D.S.; Gutkind, J.S. Molecular cross-talk between the NFkappaB and STAT3 signalling pathways in head and neck squamous cell carcinoma. Neoplasia 2006, 8, 733–746. [Google Scholar] [CrossRef] [Green Version]
  109. Eytan, D.F.; Snow, G.E.; Carlson, S.; Derakhshan, A.; Saleh, A.M.; Schiltz, S.; Cheng, H.; Mohan, S.; Cornelius, S.; Coupar, J.F.; et al. SMAC Mimetic Birinapant plus Radiation Eradicates Human Head and Neck Cancers with Genomic Amplifications of Cell Death Genes FADD and BIRC. Cancer Res. 2016, 76, 5442–5454. [Google Scholar] [CrossRef] [Green Version]
  110. Hajek, M.; Sewell, A.; Kaech, S.; Burtness, B.; Yarbrough, W.G.; Issaeva, N. TRAF3/CYLD mutations identify a distinct subset of human papillomavirus-associated head and neck squamous cell carcinoma. Cancer 2017, 123, 1778–1790. [Google Scholar] [CrossRef] [Green Version]
  111. Hrdinka, M.; Fiil, B.K.; Zucca, M.; Leske, D.; Bagola, K.; Yabal, M.; Elliott, P.R.; Damgaard, R.B.; Komander, D.; Jost, P.J.; et al. CYLD Limits Lys63- and Met1-Linked Ubiquitin at Receptor Complexes to Regulate Innate Immune Signaling. Cell Rep. 2016, 14, 2846–2858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Zarnegar, B.; Yamazaki, S.; He, J.Q.; Cheng, G. Control of canonical NF-kappaB activation through the NIK-IKK complex pathway. Proc. Natl. Acad. Sci. USA 2008, 105, 3503–3508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Suzuki, S.; Ohmori, Y.; Sakashita, H.; Hiroi, M.; Fukuda, M. Loss of CYLD might be associated with development of salivary gland tumors. Oncol. Rep. 2008, 19, 1421–1427. [Google Scholar] [CrossRef]
  114. Rito, M.; Mitani, Y.; Bell, D.; Mariano, F.V.; Almalki, S.T.; Pytynia, K.B.; Fonseca, I.; El-Naggar, A.K. Frequent and differential mutations of the CYLD gene in basal cell salivary neoplasms: Linkage to tumor development and progression. Mod. Pathol. 2018, 31, 1064–1072. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Davis, R.J.; Welcker, M.; Clurman, B.E. Tumor suppression by the Fbw7 ubiquitin ligase: Mechanisms and opportunities. Cancer Cell 2014, 26, 455–464. [Google Scholar] [CrossRef] [Green Version]
  116. Arabi, A.; Ullah, K.; Branca, R.M.M.; Johansson, J.; Bandarra, D.; Haneklaus, M.; Fu, J.; Ariës, I.; Nilsson, P.; Boer, M.L.D.; et al. Proteomic screen reveals Fbw7 as a modulator of the NF-κB pathway. Nat. Commun. 2012, 3, 976. [Google Scholar] [CrossRef]
  117. Thu, K.L.; Pikor, L.A.; Chari, R.; Wilson, I.M.; MacAulay, C.E.; English, J.C.; Tsao, M.S.; Gazdar, A.F.; Lam, S.; Lam, W.L.; et al. Genetic disruption of KEAP1/CUL3 E3 ubiquitin ligase complex components is a key mechanism of NF-kappaB pathway activation in lung cancer. J. Thorac. Oncol. 2011, 6, 1521–1529. [Google Scholar] [CrossRef] [Green Version]
  118. Drainas, A.P.; Lambuta, R.A.; Ivanova, I.; Serçin, Ö.; Sarropoulos, I.; Smith, M.L.; Efthymiopoulos, T.; Raeder, B.; Stütz, A.M.; Waszak, S.M.; et al. Genome-wide Screens Implicate Loss of Cullin Ring Ligase 3 in Persistent Proliferation and Genome Instability in TP53-Deficient Cells. Cell Rep. 2020, 31, 107465. [Google Scholar] [CrossRef]
  119. Ge, Z.; Leighton, J.S.; Wang, T.C.; Peng, X.; Chen, Z.; Chen, H.; Sun, Y.; Yao, F.; Li, J.; Zhang, H.; et al. Integrated Genomic Analysis of the Ubiquitin Pathway across Cancer Types. Cell Rep. 2018, 23, 213–226.e3. [Google Scholar] [CrossRef] [Green Version]
  120. Huang, X.; Dixit, V.M. Drugging the undruggables: Exploring the ubiquitin system for drug development. Cell Res. 2016, 26, 484–498. [Google Scholar] [CrossRef]
  121. Hideshima, T.; Richardson, P.G.; Anderson, K.C. Mechanism of Action of Proteasome Inhibitors and Deacetylase Inhibitors and the Biological Basis of Synergy in Multiple Myeloma. Mol. Cancer Ther. 2011, 10, 2034–2042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Kisselev, A.F.; Van Der Linden, W.A.; Overkleeft, H.S. Proteasome Inhibitors: An Expanding Army Attacking a Unique Target. Chem. Biol. 2012, 19, 99–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Oerlemans, R.; Franke, N.E.; Assaraf, Y.G.; Cloos, J.; Van Zantwijk, I.; Berkers, C.R.; Scheffer, G.L.; Debipersad, K.; Vojtekova, K.; Lemos, C.; et al. Molecular basis of bortezomib resistance: Proteasome subunit β5 (PSMB5) gene mutation and overexpression of PSMB5 protein. Blood 2008, 112, 2489–2499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Robak, P.; Robak, T. Bortezomib for the Treatment of Hematologic Malignancies: 15 Years Later. Drugs R&D 2019, 19, 73–92. [Google Scholar] [CrossRef] [Green Version]
  125. Horton, T.M.; Whitlock, J.A.; Lu, X.; O’Brien, M.M.; Borowitz, M.J.; Devidas, M.; Raetz, E.A.; Brown, P.A.; Carroll, W.L.; Hunger, S.P. Bortezomib reinduction chemotherapy in high-risk ALL in first relapse: A report from the Children’s Oncology Group. Br. J. Haematol. 2019, 186, 274–285. [Google Scholar] [CrossRef]
  126. Sung, M.H.; Bagain, L.; Chen, Z.; Karpova, T.; Yang, X.; Silvin, C.; Voss, T.C.; McNally, J.G.; Van Waes, C.; Hager, G.L. Dynamic effect of bortezomib on nuclear factor-κB activity and gene expression in tumor cells. Mol. Pharmacol. 2008, 74, 1215–1222. [Google Scholar] [CrossRef] [Green Version]
  127. Siegel, D.S.; Martin, T.; Wang, M.; Vij, R.; Jakubowiak, A.J.; Lonial, S.; Trudel, S.; Kukreti, V.; Bahlis, N.; Alsina, M.; et al. A phase 2 study of single-agent carfilzomib (PX-171-003-A1) in patients with relapsed and refractory multiple myeloma. Blood 2012, 120, 2817–2825. [Google Scholar] [CrossRef] [Green Version]
  128. Chung, C.H.; Aulino, J.; Muldowney, N.J.; Hatakeyama, H.; Baumann, J.; Burkey, B.; Netterville, J.; Sinard, R.; Yarbrough, W.G.; Cmelak, A.J.; et al. Nuclear factor-kappa B pathway and response in a phase II trial of bortezomib and docetaxel in patients with recurrent and/or metastatic head and neck squamous cell carcinoma. Ann. Oncol. 2010, 21, 864–870. [Google Scholar] [CrossRef]
  129. Chen, Z.; Ricker, J.L.; Malhotra, P.S.; Nottingham, L.; Bagain, L.; Lee, T.-L.; Yeh, N.T.; Van Waes, C. Differential bortezomib sensitivity in head and neck cancer lines corresponds to proteasome, nuclear factor-kappaB and activator protein-1 related mechanisms. Mol. Cancer Ther. 2008, 7, 1949–1960. [Google Scholar] [CrossRef] [Green Version]
  130. Argiris, A.; Duffy, A.G.; Kummar, S.; Simone, N.L.; Arai, Y.; Kim, S.W.; Rudy, S.F.; Kannabiran, V.R.; Yang, X.; Jang, M.; et al. Early tumor progression associated with enhanced EGFR signalling with bortezomib, cetuximab, and radiotherapy for head and neck cancer. Clin. Cancer Res. 2011, 17, 5755–5764. [Google Scholar] [CrossRef] [Green Version]
  131. Duan, J.; Friedman, J.; Nottingham, L.; Chen, Z.; Ara, G.; Van Waes, C. Nuclear factor-kappaB p65 small interfering RNA or proteasome inhibitor bortezomib sensitizes head and neck squamous cell carcinomas to classic histone deacetylase inhibitors and novel histone deacetylase inhibitor PXD. Mol. Cancer Ther. 2007, 6, 37–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Van Waes, C.; Chang, A.A.; Lebowitz, P.F.; Druzgal, C.H.; Chen, Z.; Elsayed, Y.A.; Sunwoo, J.B.; Rudy, S.F.; Morris, J.C.; Mitchell, J.B.; et al. Inhibition of nuclear factor-κB and target genes during combined therapy with proteasome inhibitor bortezomib and reirradiation in patients with recurrent head-and-neck squamous cell carcinoma. Int. J. Radiat. Oncol. 2005, 63, 1400–1412. [Google Scholar] [CrossRef] [PubMed]
  133. Kubicek, G.J.; Axelrod, R.S.; Machtay, M.; Ahn, P.H.; Anne, P.R.; Fogh, S.E.; Cognetti, D.; Myers, T.J.; Curran, W.J.; Dicker, A.P. Phase I Trial Using the Proteasome Inhibitor Bortezomib and Concurrent Chemoradiotherapy for Head-and-Neck Malignancies. Int. J. Radiat. Oncol. 2012, 83, 1192–1197. [Google Scholar] [CrossRef] [PubMed]
  134. Xu, G.W.; Ali, M.; Wood, T.E.; Wong, D.; MacLean, N.; Wang, X.; Gronda, M.; Skrtic, M.; Li, X.; Hurren, R.; et al. The ubiquitin-activating enzyme E1 as a therapeutic target for the treatment of leukemia and multiple myeloma. Blood 2010, 115, 2251–2259. [Google Scholar] [CrossRef] [Green Version]
  135. Chen, C.; Meng, Y.; Wang, L.; Wang, H.X.; Tian, C.; Pang, G.D.; Li, H.H.; Du, J. Ubiquitin-activating enzyme E 1 inhibitor PYR 41 attenuates angiotensin II-induced activation of dendritic cells via the Iκ B a/NF-κ B and MKP 1/ERK/STAT 1 pathways. Immunology 2014, 142, 307–319. [Google Scholar] [CrossRef]
  136. Barghout, S.H.; Schimmer, A.D. The ubiquitin-activating enzyme, UBA1, as a novel therapeutic target for AML. Oncotarget 2018, 9, 34198–34199. [Google Scholar] [CrossRef] [PubMed]
  137. Soucy, T.A.; Dick, L.R.; Smith, P.G.; Milhollen, M.A.; Brownell, J.E. The NEDD8 Conjugation Pathway and Its Relevance in Cancer Biology and Therapy. Genes Cancer 2010, 1, 708–716. [Google Scholar] [CrossRef] [Green Version]
  138. Duda, D.M.; Borg, L.A.; Scott, D.C.; Hunt, H.W.; Hammel, M.; Schulman, B.A. Structural Insights into NEDD8 Activation of Cullin-RING Ligases: Conformational Control of Conjugation. Cell 2008, 134, 995–1006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Godbersen, J.C.; Humphries, L.A.; Danilova, O.V.; Kebbekus, P.E.; Brown, J.R.; Eastman, A.; Danilov, A.V. The Nedd8-activating enzyme inhibitor MLN4924 thwarts microenvironment-driven NF-κB activation and induces apoptosis in chronic lymphocytic leukemia B cells. Clin. Cancer Res. 2014, 20, 1576–1589. [Google Scholar] [CrossRef] [Green Version]
  140. Zhao, L.; Yue, P.; Lonial, S.; Khuri, F.R.; Sun, S.-Y. The NEDD8-activating enzyme inhibitor, MLN4924, cooperates with TRAIL to augment apoptosis through facilitating c-FLIP degradation in head and neck cancer cells. Mol. Cancer Ther. 2011, 10, 2415–2425. [Google Scholar] [CrossRef] [Green Version]
  141. Vanderdys, V.; Allak, A.; Guessous, F.; Benamar, M.; Read, P.W.; Jameson, M.J.; Abbas, T. The Neddylation Inhibitor Pevonedistat (MLN4924) Suppresses and Radiosensitizes Head and Neck Squamous Carcinoma Cells and Tumors. Mol. Cancer Ther. 2017, 17, 368–380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Zhang, W.; Liang, Y.; Li, L.; Wang, X.; Yan, Z.; Dong, C.; Zeng, M.S.; Zhong, Q.; Liu, X.K.; Yu, J.; et al. The Nedd8-activating enzyme inhibitor MLN 4924 (TAK-924/Pevonedistat) induces apoptosis via c-Myc-Noxa axis in head and neck squamous cell carcinoma. Cell Prolif. 2019, 52, e12536. [Google Scholar] [CrossRef] [PubMed]
  143. Stewart, M.D.; Ritterhoff, T.; Klevit, R.E.; Brzovic, P.S. E2 enzymes: More than just middle men. Cell Res. 2016, 26, 423–440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Pulvino, M.; Liang, Y.; Oleksyn, D.; DeRan, M.; Van Pelt, E.; Shapiro, J.; Sanz, I.; Chen, L.; Zhao, J. Inhibition of proliferation and survival of diffuse large B-cell lymphoma cells by a small-molecule inhibitor of the ubiquitin-conjugating enzyme Ubc13-Uev1A. Blood 2012, 120, 1668–1677. [Google Scholar] [CrossRef] [PubMed]
  145. Hodge, C.; Edwards, R.A.; Markin, C.J.; McDonald, D.; Pulvino, M.; Huen, M.S.Y.; Zhao, J.; Spyracopoulos, L.; Hendzel, M.J.; Glover, J.N.M. Covalent Inhibition of Ubc13 Affects Ubiquitin Signaling and Reveals Active Site Elements Important for Targeting. ACS Chem. Biol. 2015, 10, 1718–1728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Strickson, S.; Campbell, D.G.; Emmerich, C.H.; Knebel, A.; Plater, L.; Ritorto, M.S.; Shpiro, N.; Cohen, P. The anti-inflammatory drug BAY 11-7082 suppresses the MyD88-dependent signalling network by targeting the ubiquitin system. Biochem. J. 2013, 451, 427–437. [Google Scholar] [CrossRef] [Green Version]
  147. Fouad, S.; Wells, O.S.; Hill, M.A.; D’Angiolella, V. Cullin Ring Ubiquitin Ligases (CRLs) in Cancer: Responses to Ionizing Radiation (IR) Treatment. Front. Physiol. 2019, 10. [Google Scholar] [CrossRef]
  148. Cohen, S.; Achbert-Weiner, H.; Ciechanover, A. Dual effects of IkappaB kinase beta-mediated phosphorylation on p105 Fate: SCF(beta-TrCP)-dependent degradation and SCF(beta-TrCP)-independent processing. Mol. Cell Biol. 2004, 24, 475–486. [Google Scholar] [CrossRef] [Green Version]
  149. Varfolomeev, E.; Blankenship, J.W.; Wayson, S.M.; Fedorova, A.V.; Kayagaki, N.; Garg, P.; Zobel, K.; Dynek, J.N.; Elliott, L.O.; Wallweber, H.J.; et al. IAP Antagonists Induce Autoubiquitination of c-IAPs, NF-κB Activation, and TNFα-Dependent Apoptosis. Cell 2007, 131, 669–681. [Google Scholar] [CrossRef] [Green Version]
  150. Derakhshan, A.; Chen, Z.; Van Waes, C. Therapeutic Small Molecules Target Inhibitor of Apoptosis Proteins in Cancers with Deregulation of Extrinsic and Intrinsic Cell Death Pathways. Clin. Cancer Res. 2016, 23, 1379–1387. [Google Scholar] [CrossRef] [Green Version]
  151. Xiao, R.; An, Y.; Ye, W.; Derakhshan, A.; Cheng, H.; Yang, X.; Allen, C.; Chen, Z.; Schmitt, N.C.; Van Waes, C. Dual Antagonist of cIAP/XIAP ASTX660 Sensitizes HPV− and HPV+ Head and Neck Cancers to TNFα, TRAIL, and Radiation Therapy. Clin. Cancer Res. 2019, 25, 6463–6474. [Google Scholar] [CrossRef] [PubMed]
  152. Matzinger, O.; Viertl, D.; Tsoutsou, P.; Kadi, L.; Rigotti, S.; Zanna, C.; Wiedemann, N.; Vozenin, M.-C.; Vuagniaux, G.; Bourhis, J. The radiosensitizing activity of the SMAC-mimetic, Debio 1143, is TNFα-mediated in head and neck squamous cell carcinoma. Radiother. Oncol. 2015, 116, 495–503. [Google Scholar] [CrossRef] [PubMed]
  153. Ye, W.; Gunti, S.; Allen, C.T.; Hong, Y.; Clavijo, P.E.; Van Waes, C.; Schmitt, N.C. ASTX660, an antagonist of cIAP1/2 and XIAP, increases antigen processing machinery and can enhance radiation-induced immunogenic cell death in preclinical models of head and neck cancer. Oncoimmunology 2020, 9, 1710398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Lai, K.P.; Chen, J.; Tse, W.K.F. Role of Deubiquitinases in Human Cancers: Potential Targeted Therapy. Int. J. Mol. Sci. 2020, 21, 2548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Li, M.; Brooks, C.L.; Kon, N.; Gu, W. A dynamic role of HAUSP in the p53-Mdm2 pathway. Mol. Cell 2004, 13, 879–886. [Google Scholar] [CrossRef]
  156. Mitxitorena, I.; Somma, D.; Mitchell, J.P.; Lepistö, M.; Tyrchan, C.; Smith, E.L.; Kiely, P.A.; Walden, H.; Keeshan, K.; Carmody, R.J. The deubiquitinase USP7 uses a distinct ubiquitin-like domain to deubiquitinate NF-κB subunits. J. Biol. Chem. 2020, 295, 11754–11763. [Google Scholar] [CrossRef]
  157. Chauhan, D.; Tian, Z.; Nicholson, B.; Kumar, K.S.; Zhou, B.; Carrasco, R.; McDermott, J.L.; Leach, C.A.; Fulcinniti, M.; Kodrasov, M.P.; et al. A small molecule inhibitor of ubiquitin-specific protease-7 induces apoptosis in multiple myeloma cells and overcomes bortezomib resistance. Cancer Cell 2012, 22, 345–358. [Google Scholar] [CrossRef] [Green Version]
  158. Turnbull, A.P.; Ioannidis, S.; Krajewski, W.W.; Pinto-Fernandez, A.; Heride, C.; Martin, A.C.L.; Tonkin, L.M.; Townsend, E.C.; Buker, S.M.; Lancia, D.R.; et al. Molecular basis of USP7 inhibition by selective small-molecule inhibitors. Nature 2017, 550, 481–486. [Google Scholar] [CrossRef]
  159. Harrigan, J.A.; Jacq, X.; Martin, N.M.; Jackson, S.P. Deubiquitylating enzymes and drug discovery: Emerging opportunities. Nat. Rev. Drug Discov. 2017, 17, 57–78. [Google Scholar] [CrossRef]
  160. D’Arcy, P.; Brnjic, S.; Olofsson, M.H.; Fryknäs, M.; Lindsten, K.; De Cesare, M.; Perego, P.; Sadeghi, B.; Hassan, M.; Larsson, R.; et al. Inhibition of proteasome deubiquitinating activity as a new cancer therapy. Nat. Med. 2011, 17, 1636–1640. [Google Scholar] [CrossRef]
  161. Cai, J.; Xia, X.; Liao, Y.; Liu, N.; Guo, Z.; Chen, J.; Yang, L.; Long, H.; Yang, Q.; Zhang, X.; et al. A novel deubiquitinase inhibitor b-AP15 triggers apoptosis in both androgen receptor-dependent and -independent prostate cancers. Oncotarget 2017, 8, 63232–63246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Wang, X.; Mazurkiewicz, M.; Hillert, E.K.; Olofsson, M.H.; Pierrou, S.; Hillertz, P.; Gullbo, J.; Selvaraju, K.; Paulus, A.; Akhtar, S.; et al. The proteasome deubiquitinase inhibitor VLX1570 shows selectivity for ubiquitin-specific protease-14 and induces apoptosis of multiple myeloma cells. Sci. Rep. 2016, 6, 26979–27015. [Google Scholar] [CrossRef] [PubMed]
  163. Rowinsky, E.K.; Paner, A.; Berdeja, J.G.; Paba-Prada, C.; Venugopal, P.; Porkka, K.; Gullbo, J.; Linder, S.; Loskog, A.S.I.; Richardson, P.G.; et al. Phase 1 study of the protein deubiquitinase inhibitor VLX1570 in patients with relapsed and/or refractory multiple myeloma. Investig. New Drugs 2020, 71, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Mialki, R.K.; Zhao, J.; Wei, J.; Mallampalli, D.F.; Zhao, Y. Overexpression of USP14 Protease Reduces I-κB Protein Levels and Increases Cytokine Release in Lung Epithelial Cells. J. Biol. Chem. 2013, 288, 15437–15441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Ma, Y.-S.; Wang, X.-F.; Zhang, Y.-J.; Luo, P.; Long, H.-D.; Li, L.; Yang, H.-Q.; Xie, R.-T.; Jia, C.-Y.; Lu, G.-X.; et al. Inhibition of USP14 Deubiquitinating Activity as a Potential Therapy for Tumors with p53 Deficiency. Mol. Ther. Oncolytics 2020, 16, 147–157. [Google Scholar] [CrossRef] [Green Version]
  166. Sun, X.; Gao, H.; Yang, Y.; He, M.; Wu, Y.; Song, Y.; Tong, Y.; Rao, Y. PROTACs: Great opportunities for academia and industry. Signal Transduct. Target. Ther. 2019, 4, 1–33. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The canonical NFκB signalling pathway induced by TNF-α and IL-1β and the role of ubiquitination. In TNFα signalling, binding of TNFα to its receptor TNFR1 leads to the recruitment of TRADD, TRAF2, RIP1 and cIAPs. This leads to K63-polyubiquitination on TRAF2, cIAP1/2 and RIP1, providing a binding platform for the TAK1 complex via TABIn IL-1β signalling, binding of IL-1β to its receptor IL-1R allows binding of the adapter protein MyD88 to the receptor TIR domain, resulting in the recruitment of the kinases Interleukin 1 Receptor Associated Kinase (IRAK) 1 and IRAKIRAK1 and 4 can then dissociate from the receptor and interact with the E3 ligase TRAF6, leading to the formation of K63-polyubiquitin chains on TRAF6 and IRAK1, serving as a binding platform for the TAK1 complex via TABIrrespective of the stimuli, LUBAC binding to K63-polyubiquitin chains on cIAPs stabilize the complex and induce M1-polyubiquitin chains on RIP1 and the recruitment of IKK complex via NEMO. The IKK complex is then activated by TAK1, resulting in the phosphorylation of IκBα and its subsequent proteasomal degradation. This allows nuclear translocation of the NFκB subunits and NFκB activation. The other proteins and ubiquitin linkages depicted in the figure are discussed in the text. For clarity, E2 ligases and some E3 ligases are not shown in the figure but are discussed in the text. Dark blue, core NFκB pathway components; Light blue, adapter proteins; Purple, E3 ligases; Red, Deubiquitinases.
Figure 1. The canonical NFκB signalling pathway induced by TNF-α and IL-1β and the role of ubiquitination. In TNFα signalling, binding of TNFα to its receptor TNFR1 leads to the recruitment of TRADD, TRAF2, RIP1 and cIAPs. This leads to K63-polyubiquitination on TRAF2, cIAP1/2 and RIP1, providing a binding platform for the TAK1 complex via TABIn IL-1β signalling, binding of IL-1β to its receptor IL-1R allows binding of the adapter protein MyD88 to the receptor TIR domain, resulting in the recruitment of the kinases Interleukin 1 Receptor Associated Kinase (IRAK) 1 and IRAKIRAK1 and 4 can then dissociate from the receptor and interact with the E3 ligase TRAF6, leading to the formation of K63-polyubiquitin chains on TRAF6 and IRAK1, serving as a binding platform for the TAK1 complex via TABIrrespective of the stimuli, LUBAC binding to K63-polyubiquitin chains on cIAPs stabilize the complex and induce M1-polyubiquitin chains on RIP1 and the recruitment of IKK complex via NEMO. The IKK complex is then activated by TAK1, resulting in the phosphorylation of IκBα and its subsequent proteasomal degradation. This allows nuclear translocation of the NFκB subunits and NFκB activation. The other proteins and ubiquitin linkages depicted in the figure are discussed in the text. For clarity, E2 ligases and some E3 ligases are not shown in the figure but are discussed in the text. Dark blue, core NFκB pathway components; Light blue, adapter proteins; Purple, E3 ligases; Red, Deubiquitinases.
Cancers 12 02877 g001
Figure 2. The non-canonical NFκB signalling pathway and the role of ubiquitination. In the absence of ligand (left), the TRAF2/TRAF3/cIAP complex facilitates the proteasomal degradation of NIK. This results in the sequestration of RelB in the cytoplasm by p100, the precursor of the NFκB protein pFollowing ligand: receptor engagement, TRAF3 is recruited to the receptor and is degraded by the combined functions of TRAF2 and cIAP1/The loss of TRAF3 allows the accumulation of NIK, leading to the phosphorylation and activation of IKKα. IKKα then facilitates the partial degradation of p100, allowing the activated p52/RelA heterodimer to translocate into the nucleus. The other proteins and ubiquitin linkages depicted in the figure are discussed in the text. For simplification, E2 ligases are not shown in the figure. Dark blue, core NFκB pathway components; Purple, E3 ligases; Red, Deubiquitinases.
Figure 2. The non-canonical NFκB signalling pathway and the role of ubiquitination. In the absence of ligand (left), the TRAF2/TRAF3/cIAP complex facilitates the proteasomal degradation of NIK. This results in the sequestration of RelB in the cytoplasm by p100, the precursor of the NFκB protein pFollowing ligand: receptor engagement, TRAF3 is recruited to the receptor and is degraded by the combined functions of TRAF2 and cIAP1/The loss of TRAF3 allows the accumulation of NIK, leading to the phosphorylation and activation of IKKα. IKKα then facilitates the partial degradation of p100, allowing the activated p52/RelA heterodimer to translocate into the nucleus. The other proteins and ubiquitin linkages depicted in the figure are discussed in the text. For simplification, E2 ligases are not shown in the figure. Dark blue, core NFκB pathway components; Purple, E3 ligases; Red, Deubiquitinases.
Cancers 12 02877 g002
Figure 3. The ubiquitin enzymatic cascade and the ubiquitin-proteasome system (USP). (1) A diagram of the ubiquitin enzymatic cascade. Ubiquitin is added to protein substrates via the E1-E2-E3 enzymatic cascade; (A) E1 activating enzymes, which catalyze the addition of ubiquitin to E1 enzymes is ATP-dependent [39]; (B) E2 conjugating enzymes; (C) E3 ligases, which catalyze the addition of ubiquitin to protein substrates and (D) deubiquitinases, which catalyze the removal of ubiquitin from protein substrates. The addition of ubiquitin to protein substrates can have serval consequences, including (2) the modulation of signal transduction or protein recruitment and localization or (3) proteasomal degradation, which requires ATP. Highlighted in red are components of the ubiquitin system that have been targeted with small molecule inhibitors and are discussed in this review.
Figure 3. The ubiquitin enzymatic cascade and the ubiquitin-proteasome system (USP). (1) A diagram of the ubiquitin enzymatic cascade. Ubiquitin is added to protein substrates via the E1-E2-E3 enzymatic cascade; (A) E1 activating enzymes, which catalyze the addition of ubiquitin to E1 enzymes is ATP-dependent [39]; (B) E2 conjugating enzymes; (C) E3 ligases, which catalyze the addition of ubiquitin to protein substrates and (D) deubiquitinases, which catalyze the removal of ubiquitin from protein substrates. The addition of ubiquitin to protein substrates can have serval consequences, including (2) the modulation of signal transduction or protein recruitment and localization or (3) proteasomal degradation, which requires ATP. Highlighted in red are components of the ubiquitin system that have been targeted with small molecule inhibitors and are discussed in this review.
Cancers 12 02877 g003
Table 1. Overview of the key differences between HPV− and HPV+ HNSCC.
Table 1. Overview of the key differences between HPV− and HPV+ HNSCC.
FeaturesHPV− HNSCCHPV+ HNSCC
Clinical and epidemiological characteristics
IncidenceDecreasingIncreasing
AgeDetected mostly in older adultsDetected mostly in young adults
Tumor LocationAll parts of the head and neckOropharynx, especially the tonsil and base of the tongue
PrognosisPoorGood
Response to TherapyUnfavorable response to chemoradiationFavorable response to chemoradiation
SurvivalLow survival rateHigh survival rate
Biological and histological characteristics
TP53 StatusHighly mutatedDegraded by HPV E6
p16INK4a StatusDecreased expression, due to inactivating mutations, deletions or methylationFrequently overexpressed
Genomic StabilityGenome is highly unstableGenome is less unstable
Regional MetastasisFrequent. Less bulkyMore frequent, bulky
Distant MetastasisFrequentRare
Secondary malignancyFrequentRare
HistologyModerately or well differentiated squamous cell carcinoma; keratinizedPoorly differentiated squamous cell carcinoma; basaloid
Table 2. Overview of ubiquitin-related genes modulated in HNSCC and have a potential role in NFκB activity.
Table 2. Overview of ubiquitin-related genes modulated in HNSCC and have a potential role in NFκB activity.
GeneGenomic StatusKnown Role in NFκB in HNSCC?
BIRC2/3Amplification, mRNA upregulation
TRAF3Deletion, mutation
OTUB1mRNA upregulationX
CYLDMutation
FBXW7αMutationX
CUL3MutationX
KEAP1MutationX
Table 3. Overview of clinical trials for ubiquitin-modulating compound for HNSCC.
Table 3. Overview of clinical trials for ubiquitin-modulating compound for HNSCC.
CompoundCo-TherapyPhaseClinical Trial Number or Reference
BortezomibCetuximab and radiation +/− cisplatinINCT01445405
BortezomibReirradiationI[132]
BortezomibDocetaxelII[128]
BortezomibRadiation + cisplatinI[133]
Debio 1143Radiation + cisplatinI/IINCT02022098
Debio 1143NivolumabINCT04122625
ASTX660N/AINCT02503423
BirinapantRadiationINCT03803774

Share and Cite

MDPI and ACS Style

Morgan, E.L.; Chen, Z.; Van Waes, C. Regulation of NFκB Signalling by Ubiquitination: A Potential Therapeutic Target in Head and Neck Squamous Cell Carcinoma? Cancers 2020, 12, 2877. https://doi.org/10.3390/cancers12102877

AMA Style

Morgan EL, Chen Z, Van Waes C. Regulation of NFκB Signalling by Ubiquitination: A Potential Therapeutic Target in Head and Neck Squamous Cell Carcinoma? Cancers. 2020; 12(10):2877. https://doi.org/10.3390/cancers12102877

Chicago/Turabian Style

Morgan, Ethan L., Zhong Chen, and Carter Van Waes. 2020. "Regulation of NFκB Signalling by Ubiquitination: A Potential Therapeutic Target in Head and Neck Squamous Cell Carcinoma?" Cancers 12, no. 10: 2877. https://doi.org/10.3390/cancers12102877

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