Open access peer-reviewed chapter

Phosphorylation of NF-κB in Cancer

Written By

Matthew Martin, Antja-Voy Hartley, Jiamin Jin, Mengyao Sun and Tao Lu

Submitted: 24 April 2018 Reviewed: 19 December 2018 Published: 24 April 2019

DOI: 10.5772/intechopen.83650

From the Edited Volume

Adenosine Triphosphate in Health and Disease

Edited by Gyula Mozsik

Chapter metrics overview

1,875 Chapter Downloads

View Full Metrics

Abstract

The proinflammatory transcription factor nuclear factor-κB (NF-κB) has emerged as a central player in inflammatory responses and tumor development since its discovery three decades ago. In general, aberrant NF-κB activity plays a critical role in tumorigenesis and acquired resistance to chemotherapy. This aberrant NF-κB activity frequently involves several post-translational modifications of NF-κB, including phosphorylation. In this chapter, we will specifically cover the phosphorylation sites reported on the p65 subunit of NF-κB and their relationship to cancer. Importantly, phosphorylation is catalyzed by different kinases using adenosine triphosphate (ATP) as the phosphorus donor. These kinases are frequently hyperactive in cancers and thus may serve as potential therapeutic targets to treat different cancers.

Keywords

  • kinase
  • NF-κB
  • phosphorylation
  • post-translational modifications

1. Introduction

1.1 Basic nuclear factor-κB (NF-κB) family and signaling pathways

So what is NF-κB? In mammals, NF-κB is a collective term for a small family of dimeric transcription factors [comprising p65 (RelA) and RelB, c-Rel, p50/p105 (NF-κB1), and p52/p100 (NF-κB2)]. All NF-κB proteins share a Rel homology domain (RHD), which is responsible for DNA binding and dimerization. Only p65, RelB, and c-Rel contain potent transactivation domains within sequences from C-terminal to the RHD. Therefore, p50 and p52 cannot act as transcriptional activators by themselves. Dimers of these two proteins have been reported to repress NF-κB-dependent transcription in vivo, most likely by competing with other transcriptionally active dimers. These proteins form homo- and heterodimers, and their activity is regulated by the canonical or alternative pathways described as following [1]. A simple diagram of canonical NF-κB signaling, which will be the focus of this chapter, is shown in Figure 1. The canonical pathway is activated by multiple stimuli, including proinflammatory cytokines (e.g., tumor necrosis factor, TNF; interleukin 1, IL-1), and the components of the bacterial wall (Lipopolysaccharide, LPS). Exterior signals lead to the phosphorylation and degradation of the inhibitory complex IκB, which is modulated by the IκB kinase (IKK), and its degradation allows for the release of the typical NF-κB heterodimer, p65/p50, to translocate into the nucleus. NF-κB binds to its cognate DNA elements and can transcriptionally activate different target genes among which 200–500 genes have been implicated in cell survival/apoptosis, cell growth, immune response, and inflammation [2].

Figure 1.

Pathway of canonical and non-canonical NF-κB signaling. Under the canonical pathway of NF-κB signaling, activation of the NF-κB is initiated by a stimulus resulting in phosphorylation and subsequent proteasomal degradation of IκBα. This allows the release of the p65/p50 heterodimer into the nucleus, where they can bind to their cognate DNA elements and promote NF-κB target gene expression [1]. On the other hand, under noncanonical activation of NF-κB, NIK (NF-κB-inducing kinase) leads to activation of IKKα in this pathway. Subsequent phosphorylation of the NF-κB1 precursor molecule, p100, triggers partial proteolysis giving rise to p52, which preferentially dimerizes with RelB. This allows translocation of p52/RelB to the nucleus where they can bind to cognate DNA elements and promote gene transcription [1]. Figure adapted and simplified from Hoesel et al. [103].

The alternative or noncanonical pathway is activated by the members of the TNF cytokine family, such as B-cell activating factor (BAFF), cluster of differentiation 40 ligand (CD40L), receptor activator of nuclear factor-κB ligand (RANKL), and lymphotoxin-β2 (LTβ2), and requires recruitment of the p52/RelB dimers to activate transcription. Firstly, activation of NIK (NF-κB-inducing kinase) leads to activation of IKKα in this pathway. This event leads to the subsequent phosphorylation of the NF-κB1 precursor molecule, p100, and triggers partial proteolysis to give rise to p52, which preferentially dimerizes with RelB [1]. The p52/RelB heterodimer then translocates to the nucleus where they can bind to cognate DNA elements and promote gene transcription (Figure 1).

1.2 Important role of NF-κB in cancer

NF-κB was first discovered by Dr. Ranjan Sen in 1986 [3]. This family of transcription factors plays important roles in the regulation of apoptosis, proliferation, inflammation, and immune response in both normal and cancer cells. Generally, in normal cells, the central transcription factor NF-κB is transiently activated in response to certain stimuli. However, cancer cells usually exhibit sustained activation of NF-κB [4, 5] which significantly contributes to their survival. Moreover, NF-κB activity plays critical roles in many of the well-known “hallmarks” of cancer, via its regulation of target genes involved in tumor cell proliferation, suppression of apoptosis, activation of angiogenesis as well as induction of the epithelial-to-mesenchymal transition (EMT) phenotype, a critical step in metastasis [6]. Constitutively active NF-κB has been found in many types of cancer. For instance, in thyroid cancer, oncogenic proteins including “rearranged during transfection” (RET), “Rat Sarcoma” (RAS), and “v-Raf murine sarcoma viral oncogene homolog B” (BRAF) were shown to induce NF-κB activation, which in turn activated proliferative and antiapoptotic signaling pathways [7]. Moreover, in renal cell carcinoma (RCC), NF-κB is constitutively activated. The phosphorylated p65, a major subunit of NF-κB, exhibited a significant increase in the RCC samples compared with corresponding normal tissues [8]. Furthermore, Nogueira et al. showed that in glioblastoma (GBM), deletion of IκB showed a phenotype similar to that of epidermal growth factor receptor (EGFR) amplification in the pathogenesis of GBM. This was also correlated with low survival rates in affected patients [2].

Importantly, our laboratory also found that in colon cancer, NF-κB can be activated by the Y-box protein 1 (YBX1), a critical event correlated with increased colon cancer cell proliferation and anchorage-independent growth [9, 10]. Additionally, in pancreatic cancer, a mutant oncogenic KrasG12D (glycine to aspartic acid) mutation induced positive feedback loops of interleukin 1α (IL-1α) and p62 expression to sustain constitutive IKKβ (inhibitor of NF-κB kinase subunit β)/NF-κB activation [11]. In breast cancer, moderately elevated NF-κB led to chronic inflammatory conditions that result in some cells escaping immune surveillance [12]. Additionally, NF-κB activation was shown to upregulate the expression of cyclin D1, cyclin-dependent kinase 2 (CDK2), and c-Myc, which drives cell cycle progression and causes uncontrolled cell proliferation [13]. Moreover, in breast cancer, NF-κB has been shown to induce and stabilize the expression of EMT markers (Snail and twist-related protein1) [14], a pivotal process in tumor metastasis. In addition to the role of NF-κB in solid tumors as described thus far, Gasparini et al. has thoroughly reviewed the important tumorgenic role of NF-κB in hematological malignancies, including acute lymphocytic lymphoma (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), chromic myeloid leukemia (CML), B lymphomas, diffuse large B-cell lymphomas (DLBCLs), Hodgkin’s lymphoma, adult T-cell lymphomas (ATLL), anaplastic large-cell lymphomas (ALCL), and multiple myeloma, which will not be further discussed in this chapter [15]. Overall, these studies highlight a prominent role of dysregulated NF-κB in multiple aspects of cancer progression in both solid tumors and hematological malignancies.

Advertisement

2. Role of phosphorylation of NF-κB in cancer

2.1 Phosphorylation of the p65 subunit of NF-ĸB and the role of its kinases in cancer

Phosphorylation is a critical modification for NF-κB activation and plays an indispensable role in the regulation of its target genes. Moreover, as mentioned above, many of these target genes contribute to the hallmarks of cancer such as cellular proliferation, antiapoptosis as well as enhanced angiogenesis via vascular endothelial growth factor expression, among others [6]. Thus, understanding how phosphorylation of NF-κB contributes to these cancer phenotypes is a critical step in effectively limiting NF-κB activity [16]. Generally speaking, phosphorylation requires phosphorus which is supplied by the donor molecule adenosine tri-phosphate (ATP). Although several members of the NF-κB family of proteins are reported to be subjected to phosphorylation, p65 stands out as the most frequently modified subunit (Table 1). Furthermore, scientists have found that p65 can be phosphorylated by a variety of different kinases, some of which are themselves frequently overactive in cancer. For instance, p65 phosphorylation at serine 536 (S536) by IKKβ has been shown to be critical for TNFα-induced transformation of mouse epidermal cells [17]. Additional studies have also reported a role for p65 S536 phosphorylation in mediating expression of matrix metalloproteinase 1 (MMP-1) in lymphomas, wherein high MMP-1 expression correlated with lymphatic invasion and lymph node metastasis [18]. Another study with an immortalized prostate cell line, PNT1a, showed a role for phosphorylated S536 of p65 in cell motility and transformation [19].

Known phosphorylation sites of p65Cancers involvedCell line discoveredReferences
S205No cancers currently knownHEK 293 cells[104]
T254Breast cancerBT20 and MCF-7 cells[105]
S529Breast cancerHeLa cells[33]
S536Bone cancerHeLa and BC-3 cells[35]
S276Head and neck cancers, breast cancerHNSCC cells[34]
S281No cancers currently knownMEF cells[106]
S311No cancers currently knownHEK 293 and MEF cells[107]
T435No cancers currently knownSiHa cells[108]
S468No cancers currently knownHeLa cells[109]
T505No cancers currently knownNARF2 and Hs68 cells[110]
S535No cancers currently knownHeLa cells[111]
S316No cancers currently knownHEK 293 cells[41]
S547No cancers currently knownHEK 293 cells[112]

Table 1.

List of known phosphorylation sites on the p65 subunit of NF-κB and their relationship to cancer.

As mentioned above, phosphorylation events on NF-κB are mediated by a variety of kinases. It is therefore unsurprising that the action of these kinases has been tightly regulated to maintain normal cellular function. However, deregulation of kinase activity can have detrimental downstream effects, which also involves the aberrant activation of NF-κB and its target genes to promote a cancer phenotype. Several kinases have been shown to have critical roles in the regulation of the p65 subunit of NF-κB. For example, glycogen synthase kinase 3 beta (GSK3β) and TRAF-associated NF-κB activator TBK1 ((TANK)-binding kinase 1) have been shown to be critical activators of NF-κB signaling [20, 21] by targeting p65 for phosphorylation on S536. This phosphorylation leads to enhanced NF-κB transactivation both in vitro and in vivo [22, 23, 24]. Another well-known kinase involved in modifying p65 is protein kinase A (PKAc). PKAc is typically activated following IκBα-degradation, leading to PKAc-mediated phosphorylation of p65 on S276 [25]. This phosphorylation event causes recruitment of histone acetyltransferases including cAMP response element-binding (CREB)-binding protein (CBP) and p300. The net effect is displacement of p50-histone deacetylase (HDAC)-1 complex from DNA, which increases p65 transactivation ability [26, 27]. Other kinases can also phosphorylate p65 at S276. These include mitogen- and stress-activated protein kinase-1 and 2 (MSK1, 2), proto-oncogene serine/threonine-protein kinase PIM-1 (PIM-1), ribosomal s6 kinase (RSK) p90, and protein kinase C α (PKCα) [28, 29, 30, 31]. Moreover, casein kinase II (CK2), which phosphorylates p65 on S529, has been implicated in breast cancer [32, 33]. Another study demonstrated a role for p65 S276 phosphorylation by protein kinase A (PKA) in promoting a malignant phenotype in head and neck squamous cell carcinoma (HNSCC). Here, the authors found that S276 phosphorylation was prevalent in the nucleus of HNSCC samples but cytoplasmic in normal mucosa. Furthermore, this TNF-α-induced nuclear p65-S276 phosphorylation was significantly inhibited by the PKA inhibitor H-89, which in turn suppressed NF-κB activity, target gene expression, cell proliferation, and induced cell death via G1/S phase arrest [34]. Other p65-targeting kinases implicated in cancer include cell division protein kinase 6 (CDK6) and PIM-1, which phosphorylate p65 at S536 and S276, respectively [29, 35]. Both PIM1 and CDK6 have been shown to be overexpressed in a variety of cancers including hematological cancers, prostate cancer, pancreatic cancer, gastric cancer, head and neck cancer, liver cancer, glioblastomas, medulloblastomas, colon cancer, and lung cancers [36, 37, 38, 39]. However, their exact roles in regulating p65 phosphorylation in these cancers are yet to be understood. In gastric cancer cells, Aurora Kinase A (AURKA) was also shown to phosphorylate S536 in in vivo and in vitro models [40] whereby overexpression of AURKA induced a significant increase in NF-κB p65 and phospho-p65 (S536) protein levels. Interestingly, protein kinase C δ (PKCδ), a member of novel PKC isoforms, has also been implicated in a number of cancers including breast, pancreatic, prostate, and melanoma tumor cells but has been shown to regulate p65 transactivation in a phosphorylation-independent manner [41]. Our laboratory has also recently reported a novel phosphorylation site on S316 of p65, a modification mediated by the kinase CKII [41]. We showed that S316 phosphorylation was necessary for NF-κB activation and target gene expression. Collectively, these examples indicate the importance and sophistication of p65 phosphorylation and their corresponding kinases in regulating NF-κB signaling in the context of cancer.

2.2 Importance and effects of phosphorylation of p65 in modulating chemoresistance

Several studies have indicated a role for NF-κB hyperactivity in the development of resistance to chemotherapeutics via downregulation of antisurvival and upregulation of prosurvival target genes and pathways [42, 43, 44]. In one study for example, gemcitabine-resistant pancreatic cancer cells were rendered sensitive to gemcitabine upon knockdown of p65 [42]. These and other accounts of NF-κB-mediated chemoresistance have been extensively reviewed by others such as Li, Sethi, and Godwin et al., which will not be further discussed in this chapter [45, 46]. However, the specific contribution of dysregulated p65 phosphorylation to chemoresistance is less well understood and requires further exploration. Nonetheless, a few reports suggest that upstream kinases involved in chemoresistance can modulate p65 phosphorylation levels in this context. For instance, siRNA-mediated depletion of IKKα in HT1080 human fibrosarcoma cells was shown to decrease phosphorylation of p65 in response to doxorubicin, thus severely impairing the ability of doxorubicin to initiate NF-κB DNA-binding activity. These findings suggest that IKKα plays a critical role in NF-κB-mediated chemoresistance in response to doxorubicin and potentially serves as a therapeutic target for improving chemotherapeutic response [47]. Other studies have shown that p65, in a hyperphosphorylated state, can be correlated with resistance to thymidylate synthases and irinotecan in stomach and colon cancers, respectively [44, 48, 49]. Doxorubicin resistance in lung cancer has also been correlated with p65 S536 phosphorylation states [47]. Additionally, multiple myelomas have exhibited increased p65 S536 phosphorylation within melphalan- or doxorubicin-resistant cells [50].

Advertisement

3. p65 modifying kinases as potential therapeutic targets

3.1 Current therapeutics used to treat cancers with constitutive NF-κB activity

The NF-κB pathway is widely considered an attractive therapeutic target in a broad range of cancers. Yet, despite the efforts to develop NF-κB inhibitors, none has been clinically approved. This is largely due to immune-related toxicities associated with global NF-κB suppression [51]. Furthermore, the high complexity of the NF-κB signaling network presents another unique challenge for developing specific NF-κB inhibitors. To further complicate matters, some standard anticancer agents can inadvertently activate the NF-κB pathway via induction of proinflammatory cytokines such as IL-1β and TNF-α and cellular stressors such as reactive oxygen species (ROS), or by activating DNA-repair mechanisms [52]. Finally, constitutive NF-κB activity can also be achieved via secreted cytokines and chemokines from inflammatory cells within the tumor microenvironment [52]. Taken together, consideration of all these factors is imperative when strategizing the development of the most effective and least toxic anticancer agents.

Currently, the use of NF-κB inhibitors has mainly been combined with other agents [47, 50]. Some of these combinatorial therapies have shown promising effectiveness and have been made it as far as the clinical trial phase. For example, combination of irinotecan with the proteasome and NF-κB inhibitor bortezomib was shown to increase sensitivity of colon cancer cells to irinotecan [53]. A separate study showed that bortezomib could sensitize non-small cell lung cancer (NSCLC) cells to sodium butyrate, which acts to inhibit histone deacetylases [54]. Moreover, several clinical trials testing the efficacy of inhibitors against IKK to target solid tumors have been undertaken. For example, perturbation of IKKβ with the inhibitory ML120B led to synergistic enhancement of vincristine cytotoxicity in lymphoma. These results implicate IKK disruption using inhibitors as a useful adjunct therapy with standard chemotherapeutics. Other attempted trials using IKK inhibitors, such as CHS-828, EB-1627, and IMD-1041, as single or combinatorial agents unfortunately produced toxicity concerns for patients [55, 56, 57].

Other examples of combinatorial therapies include the use of NF-κB inhibitors Bay11-7082 and sulfasalazine in combination with more commonly used chemotherapeutics such as 5-fluorouracil and cisplatin to synergistically reduce colon cancer cell growth [58]. Other indirect means of targeting NF-κB such as inhibition of upstream kinases have also shown promise. For instance, one study using pancreatic cancer cells showed that inhibition of GSK3β by a small molecule inhibitor reduced phosphorylation of p65 at S536 resulting in decreased NF-κB activity and cell growth [59]. Another study with the chemical compound ursolic acid showed reduced p65 phosphorylation via inhibition of IKKβ, which impaired overall cell growth in leukemia cell lines [60]. Other studies with proteasome inhibitors, including Tosyl phenylalanyl chloromethyl ketone (TPCK) and Tosyl-L-lysyl-chloromethane hydrochloride (TLCK), demonstrated that not only do these inhibitors target IKKβ, but they were also able to reduce overall phosphorylation levels of NF-κB [61, 62]. In summary, these studies suggest there may be many benefits to targeting hyperactive NF-κB signaling and, in particular, the kinases that regulate NF-κB in various cancers.

3.2 Benefits and pitfalls for targeting kinases in cancers

The development of small-molecule kinase inhibitors for the treatment of cancer has continued to be of intense interest. Notably, many inhibitors have received FDA approval with approximately another 150 are in preclinical and clinical phase trials. Despite these important advances, many factors have confounded the clinical efficacy of these kinase-targeted drugs including the challenges of tumor heterogeneity and microenvironment as well as the emergence of mutations that confer drug resistance. Another major challenge of kinase inhibition is that of the development of adverse side effects. Some classic examples of this include dermatologic complications and cardiotoxicity associated with inhibition of EGFR and vascular endothelial growth factor receptor (VEGFR), respectively. Furthermore, there is an urgent need to develop relevant models of resistance in response to kinase inhibitors in efforts to overcome this resistance via potential synergistic combinatorial therapies.

Another critical issue facing clinical trial design with kinase-targeted agents is that of determining the types of tumors that are most likely to respond to specific kinase inhibitors and thus identify the subsets of patients who will likely benefit from these treatments. To combat this issue, many studies have been dedicated to identifying certain “kinase dependencies” in cancer cells that would make them more susceptible to inhibition. These so-called dependencies are primarily based on the existence of constitutively activate kinases achieved by gene mutation, amplification, or fusion. Among the potential approaches to identifying signatures of kinase dependency are proteomic profiling, next-generation sequencing and various applications utilizing phospho-specific antibodies against numerous specific kinase substrates. Additional mechanisms of kinase dependency include impairment of the function of phosphatases, the negative regulators of phosphorylation as is the case with mutations in the phosphatase and tensin homolog (PTEN) tumor suppressor gene. The consequence of PTEN loss is signal propagation through downstream kinases such as Akt. Moreover, growing evidence from isogenic human and mouse models also suggests that this type of indirect avenue of kinase dependency may be analogous to direct, activating mutations in the kinases themselves.

Finally, there are also cases in which the beneficial effect of a kinase inhibitor is counteracted by an additional genetic lesion in a compensatory signaling pathway. Therefore, studies to identify such secondary events are urgently needed. Taken together, these evidences underscore the critical need to optimize the use of kinase inhibitors against cancers by continued detailed molecular characterization of tumor tissues. It will be critical to develop new compounds that circumvent acquired resistance to the first-generation kinase inhibitors for patients with refractory disease.

3.3 Cutting edge therapeutics for treating cancers with constitutively active kinases

Since the mid-1970s, numerous studies have highlighted a crucial role for kinases in promoting tumorigenesis and metastasis [63, 64, 65, 66, 67]. This is of no surprise, since a clear majority of protein kinases promote critical cellular functions pertinent to cancer progression, including cell proliferation, survival, and migration [68, 69, 70, 71, 72]. Hence, targeting mutated, overexpressed, or hyperactive kinases represents an important and promising clinical niche for developing drugs for cancer therapy [67, 73, 74]. Interestingly, inhibitors against kinases account for approximately 25% of all current drug discovery efforts. So far, ~37 inhibitors with activity targeted to one or multiple kinases have been approved for clinical use [67, 75]. These range from highly selective monoclonal antibodies to more broad-spectrum synthetic or natural small molecules that have achieved a significant increase in patient survival rate in cancers [67]. For example, treatment with imatinib and dasatinib, which are highly potent and selective tyrosine kinase inhibitors against BCR-ABL, produces more favorable outcomes compared to conventional cytotoxic therapy for patients with chronic myeloid leukemia (CML) [76, 77, 78]. Similarly, broad-spectrum inhibitors have also been used with great success. For example, CHIR-258, which targets multiple kinases, inhibits VEGFR, platelet-derived growth factor receptor (PDGFR), FMS-like tyrosine kinase-3 (FLT-3), mast/stem cell growth factor receptor (c-Kit), and fibroblast growth factor receptor (FGFR) in multiple myeloma patients and is most effective for killing tumors harboring the translocation t [4, 14] (p16.3;q32.3) with increased expression and activating mutations of FGFR3 [79, 80, 81].

Some other major inhibitors to enter the market include those targeting key oncogenic kinase drug targets such as ERBB2 (e.g., afatinib), HER2 (e.g., trastuzumab), and VEGFRs (e.g., sorafenib, a small molecule inhibitor used for the treatment of renal cell, liver. and thyroid cancers) [82, 83, 84, 85, 86, 87]. Another successful targeting strategy has been the use of a monoclonal antibody against VEGFR (bevacizumab). Bevacizumab is used in combination with chemotherapy to treat patients with metastatic colon cancer, and its widespread use has resulted in significant improvement in survival outcomes [88]. Other examples include inhibitors against Kit, PDGFRs, proto-oncogene tyrosine-protein kinase Src (SRC), mechanistic target of rapamycin and FK506-binding protein 12-rapamycin-associated protein 1 (mTOR), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), serine/threonine-protein kinase B (PKB or Akt)—Raf (BRAF), and epidermal growth factor receptor (EGFR) (e.g., cetuximab, panitumumab), all of which act to activate significant tumor cell signaling pathways such as NF-ĸB [67, 89]. For instance, several FDA-approved kinase inhibitors, although not perceived as direct NF-ĸB inhibitors, have been shown to suppress NF-κB signaling [89]. These include inhibitors targeting EGFR in breast and lung cancer, Akt in breast cancer, GSK-3β in pancreatic cancer, NIK in melanoma, BRAF in multiple myeloma, and IKK in brain and liver cancer [89, 90, 91]. Specifically, GSK2118436, PLX4720, sorafenib, and PLX4032 are all drugs which are currently being used to target B-RafV600E in advanced cancers with elevated NF-ĸB activity [73, 92, 93, 94]. Finally, small molecule inhibitors targeting Akt, which include perifosine, GSK690693, VQD002, and MK2206, are also being tested clinically [95, 96]. In summary, these studies highlight the importance of inhibition of distinct kinase signaling pathways as a means of minimizing cytotoxic effects on noncancerous cells, thus bestowing selective killing of tumor cells and improving patient clinical outcomes.

Advertisement

4. Conclusion and future directions

In summary, this chapter highlights a significant role of NF-ĸB phosphorylation in driving the initiation and progression of several cancers as well as chemoresistance to first-line therapies. Furthermore, we emphasized the relationship between p65 phosphorylation and the role that constitutively active kinases play in promoting the cancer phenotype, via utilization of ATP as the phosphate donor. Finally, we underlined the well-established therapeutic potential of targeting these kinases in the treatment of various cancers. Despite these encouraging data, we acknowledge that the difficulties with drug resistance and toxicity continue to present critical challenges for the use of kinase inhibitors in both clinical and experimental oncology. Furthermore, issues related to the inadequate understanding of the selectivity of the kinase inhibitors have also plagued the successful clinical utility of these inhibitors. Nevertheless, a key challenge for overcoming this enigma is to identify the most efficacious, complementary, and least toxic combinations of kinase inhibitors for targeted cancer treatment [97, 98]. This will likely lead to the development of multimodal treatment initiatives that evade the treatment-related drug resistance. Finally, it is well known that cancers have heterogeneous populations of cells, and these may differentially contribute to chemoresistance [99, 100]. To address this, many efforts are underway to eliminate cancer stem cells as main culprits of this intrinsic heterogeneity, which will undoubtedly improve our understanding of better drug design and efficiency [101, 102].

Advertisement

Acknowledgments

This publication is made possible, in part, with support from the Indiana Clinical and Translational Sciences Institute (CTSI) funded from the National Institutes of Health, National Center for Advancing Translational Sciences, Clinical and Translational Sciences Award (to TL); V foundation Kay Yow Cancer Fund (Grant 4486242 to TL); NIH-NIGMS Grant (#1R01GM120156-01A1 (to TL); and 100 VOH Grant (#2987613 to TL), as well as NIH-NCI Grant (#1R03CA223906-01).

References

  1. 1. Jost PJ, Ruland J. Aberrant NF-κB signaling in lymphoma: Mechanisms, consequences, and therapeutic implications. Blood. 2007;109(7):2700-2707
  2. 2. Nogueira L et al. The NFκB pathway: A therapeutic target in glioblastoma. Oncotarget. 2011;2(8):646
  3. 3. Sen R, Baltimore D. Multiple nuclear factors interact with the immunoglobulin enhancer sequences. Cell. 1986;46(5):705-716
  4. 4. Lu T, Stark GR. NF-κB: Regulation by methylation. Cancer Research. 2015;75(18):3692-3695
  5. 5. Vaiopoulos AG, Athanasoula KC, Papavassiliou AG. NF-κB in colorectal cancer. Journal of Molecular Medicine. 2013;91(9):1029-1037
  6. 6. Xia Y, Shen S, Verma IM. NF-κB, an active player in human cancers. Cancer Immunology Research. 2014;2(9):823-830
  7. 7. Pacifico F, Leonardi A. Role of NF-κB in thyroid cancer. Molecular and Cellular Endocrinology. 2010;321(1):29-35
  8. 8. Morais C et al. The emerging role of nuclear factor kappa B in renal cell carcinoma. The International Journal of Biochemistry & Cell Biology. 2011;43(11):1537-1549
  9. 9. Martin M et al. Novel serine 176 phosphorylation of YBX1 activates NF-κB in colon cancer. Journal of Biological Chemistry. 2017. jbc-M116
  10. 10. Prabhu L et al. Critical role of phosphorylation of serine 165 of YBX1 on the activation of NF-κB in colon cancer. Oncotarget. 2015;6(30):29396
  11. 11. Prabhu L et al. Critical role of NF-κB in pancreatic cancer. Oncotarget. 2014;5(22):10969
  12. 12. Smyth MJ, Dunn GP, Schreiber RD. Cancer immunosurveillance and immunoediting: The roles of immunity in suppressing tumor development and shaping tumor immunogenicity. Advances in Immunology. 2006;90:1-50
  13. 13. Hinz M et al. NF-κB function in growth control: Regulation of cyclin D1 expression and G0/G1-to-S-phase transition. Molecular and Cellular Biology. 1999;19(4):2690-2698
  14. 14. Wu Y et al. Stabilization of snail by NF-κB is required for inflammation-induced cell migration and invasion. Cancer Cell. 2009;15(5):416-428
  15. 15. Gasparini C et al. NF-κB pathways in hematological malignancies. Cellular and Molecular Life Sciences. 2014;71(11):2083-2102
  16. 16. Viatour P et al. Phosphorylation of NF-κB and IκB proteins: Implications in cancer and inflammation. Trends in Biochemical Sciences. 2005;30(1):43-52
  17. 17. Hu J et al. Insufficient p65 phosphorylation at S536 specifically contributes to the lack of NF-κB activation and transformation in resistant JB6 cells. Carcinogenesis. 2004;25(10):1991-2003
  18. 18. Wang Y-P et al. Astrocyte elevated gene-1 is associated with metastasis in head and neck squamous cell carcinoma through p65 phosphorylation and upregulation of MMP1. Molecular Cancer. 2013;12(1):109
  19. 19. Zhang L et al. Function of phosphorylation of NF-kB p65 ser536 in prostate cancer oncogenesis. Oncotarget. 2015;6(8):6281
  20. 20. Bonnard M et al. Deficiency of T2K leads to apoptotic liver degeneration and impaired NF-κB-dependent gene transcription. The EMBO Journal. 2000;19(18):4976-4985
  21. 21. Hoeflich KP et al. Requirement for glycogen synthase kinase-3β in cell survival and NF-κB activation. Nature. 2000;406(6791):86
  22. 22. Schwabe RF, Brenner DA. Role of glycogen synthase kinase-3 in TNF-α-induced NF-κB activation and apoptosis in hepatocytes. American Journal of Physiology-Gastrointestinal and Liver Physiology. 2002;283(1):G204-G211
  23. 23. Fujita F et al. Identification of NAP1, a regulatory subunit of IκB kinase-related kinases that potentiates NF-κB signaling. Molecular and Cellular Biology. 2003;23(21):7780-7793
  24. 24. Ly Philip TT et al. Inhibition of GSK3β-mediated BACE1 expression reduces Alzheimer-associated phenotypes. The Journal of Clinical Investigation. 2012;123(1)
  25. 25. Zhong H et al. The transcriptional activity of NF-κB is regulated by the IκB-associated PKAc subunit through a cyclic AMP–independent mechanism. Cell. 1997;89(3):413-424
  26. 26. Zhong H, Voll RE, Ghosh S. Phosphorylation of NF-κB p65 by PKA stimulates transcriptional activity by promoting a novel bivalent interaction with the coactivator CBP/p300. Molecular Cell. 1998;1(5):661-671
  27. 27. Zhong H et al. The phosphorylation status of nuclear NF-κB determines its association with CBP/p300 or HDAC-1. Molecular Cell. 2002;9(3):625-636
  28. 28. Vermeulen L et al. Transcriptional activation of the NF-κB p65 subunit by mitogen-and stress-activated protein kinase-1 (MSK1). The EMBO Journal. 2003;22(6):1313-1324
  29. 29. Nihira K et al. Pim-1 controls NF-κB signalling by stabilizing RelA/p65. Cell Death and Differentiation. 2010;17(4):689
  30. 30. Wang H et al. Proteinase-activated receptors induce interleukin-8 expression by intestinal epithelial cells through ERK/RSK90 activation and histone acetylation. The FASEB Journal. 2010;24(6):1971-1980
  31. 31. Wang Y et al. M-CSF induces monocyte survival by activating NF-κB p65 phosphorylation at Ser276 via protein kinase C. PLoS One. 2011;6(12):e28081
  32. 32. Duncan JS, Litchfield DW. Too much of a good thing: the role of protein kinase CK2 in tumorigenesis and prospects for therapeutic inhibition of CK2. Biochimica et Biophysica Acta (BBA)-Proteins and Proteomics. 2008;1784(1):33-47
  33. 33. Wang D et al. TNFalpha-induced phosphorylation of RelA/p65 on Ser529 is controlled by casein kinase II. Journal of Biological Chemistry. 2000
  34. 34. Arun P et al. Nuclear NF-κB p65 phosphorylation at serine 276 by protein kinase A contributes to the malignant phenotype of head and neck cancer. Clinical Cancer Research. 2009;15(19):5974-5984
  35. 35. Buss H et al. Cyclin-dependent kinase 6 phosphorylates NF-κB P65 at serine 536 and contributes to the regulation of inflammatory gene expression. PLoS One. 2012;7(12):e51847
  36. 36. Valdman A et al. Pim-1 expression in prostatic intraepithelial neoplasia and human prostate cancer. The Prostate. 2004;60(4):367-371
  37. 37. Lam YP, di Tomaso E, Ng H-K, Pang JCS, Roussel MF, Hjelm NM. Expression of p19 INK4d, CDK4, CDK6 in glioblastoma multiforme. British Journal of Neurosurgery. 2000;14(1):28-32
  38. 38. Mendrzyk F et al. Genomic and protein expression profiling identifies CDK6 as novel independent prognostic marker in medulloblastoma. Journal of Clinical Oncology. 2005;23(34):8853-8862
  39. 39. Igarashi K et al. Activation of cyclin D1-related kinase in human lung adenocarcinoma. British Journal of Cancer. 1999;81(4):705
  40. 40. Katsha A et al. Aurora kinase A promotes inflammation and tumorigenesis in mice and human gastric neoplasia. Gastroenterology. 2013;145(6):1312-1322
  41. 41. Wang B et al. Role of novel serine 316 phosphorylation of the p65 subunit of NF-κB in differential gene regulation. Journal of Biological Chemistry. 2015;jbc-M115
  42. 42. Pan X et al. Nuclear factor-κB p65/relA silencing induces apoptosis and increases gemcitabine effectiveness in a subset of pancreatic cancer cells. Clinical Cancer Research. 2008;14(24):8143-8151
  43. 43. Zemskova M et al. The PIM1 kinase is a critical component of a survival pathway activated by docetaxel and promotes survival of docetaxel-treated prostate cancer cells. Journal of Biological Chemistry. 2008;283(30):20635-20644
  44. 44. Uetsuka H et al. Inhibition of inducible NF-κB activity reduces chemoresistance to 5-fluorouracil in human stomach cancer cell line. Experimental Cell Research. 2003;289(1):27-35
  45. 45. Li F, Gautam S. Targeting transcription factor NF-κB to overcome chemoresistance and radioresistance in cancer therapy. Biochimica et Biophysica Acta (BBA)-Reviews on Cancer. 2010;1805(2):167-180
  46. 46. Godwin P et al. Targeting nuclear factor-kappa B to overcome resistance to chemotherapy. Frontiers in Oncology. 2013;3:120
  47. 47. Bednarski BK et al. Active roles for inhibitory κB kinases α and β in nuclear factor-κB–mediated chemoresistance to doxorubicin. Molecular Cancer Therapeutics. 2008;7(7):1827-1835
  48. 48. Guo J et al. Enhanced chemosensitivity to irinotecan by RNA interference-mediated down-regulation of the nuclear factor-κB p65 subunit. Clinical Cancer Research. 2004;10(10):3333-3341
  49. 49. Wang W, McLeod HL, Cassidy J. Disulfiram-mediated inhibition of NF-κB activity enhances cytotoxicity of 5-fluorouracil in human colorectal cancer cell lines. International Journal of Cancer. 2003;104(4):504-511
  50. 50. Baumann P et al. Alkylating agents induce activation of NFκB in multiple myeloma cells. Leukemia Research. 2008;32(7):1144-1147
  51. 51. Dai Y et al. Blockade of histone deacetylase inhibitor-induced RelA/p65 acetylation and NF-κB activation potentiates apoptosis in leukemia cells through a process mediated by oxidative damage, XIAP downregulation, and c-Jun N-terminal kinase 1 activation. Molecular and Cellular Biology. 2005;25(13):5429-5444
  52. 52. Wu Z-H et al. Molecular linkage between the kinase ATM and NF-κB signaling in response to genotoxic stimuli. Science. 2006;311(5764):1141-1146
  53. 53. Cusack JC et al. Enhanced chemosensitivity to CPT-11 with proteasome inhibitor PS-341: Implications for systemic nuclear factor-κB inhibition. Cancer Research. 2001;61(9):3535-3540
  54. 54. Denlinger CE et al. Combined proteasome and histone deacetylase inhibition in non–small cell lung cancer. The Journal of Thoracic and Cardiovascular Surgery. 2004;127(4):1078-1086
  55. 55. Ravaud A et al. Phase I study and pharmacokinetic of CHS-828, a guanidino-containing compound, administered orally as a single dose every 3 weeks in solid tumours: An ECSG/EORTC study. European Journal of Cancer. 2005;41(5):702-707
  56. 56. Huang J-J et al. Novel IKKβ inhibitors discovery based on the co-crystal structure by using binding-conformation-based and ligand-based method. European Journal of Medicinal Chemistry. 2013;63:269-278
  57. 57. Matsumoto R et al. Inhibition of IκB phosphorylation by a novel IKK inhibitor IMD-1041 attenuates myocardial dysfunction after infarction. Immunology, Endocrine & Metabolic Agents in Medicinal Chemistry. 2012;12(2):137-142
  58. 58. Levidou G et al. Expression of nuclear factor κB in human gastric carcinoma: Relationship with IκBa and prognostic significance. Virchows Archiv. 2007;450(5):519-527
  59. 59. Ougolkov AV et al. Glycogen synthase kinase-3β participates in nuclear factor κB–mediated gene transcription and cell survival in pancreatic cancer cells. Cancer Research. 2005;65(6):2076-2081
  60. 60. Shishodia S et al. Ursolic acid inhibits nuclear factor-κB activation induced by carcinogenic agents through suppression of IκBα kinase and p65 phosphorylation: Correlation with down-regulation of cyclooxygenase 2, matrix metalloproteinase 9, and cyclin D1. Cancer Research. 2003;63(15):4375-4383
  61. 61. Natarajan K et al. Caffeic acid phenethyl ester is a potent and specific inhibitor of activation of nuclear transcription factor NF-kappa B. Proceedings of the National Academy of Sciences. 1996;93(17):9090-9095
  62. 62. Finco TS, Beg AA, Baldwin AS. Inducible phosphorylation of I kappa B alpha is not sufficient for its dissociation from NF-kappa B and is inhibited by protease inhibitors. Proceedings of the National Academy of Sciences. 1994;91(25):11884-11888
  63. 63. Drake JM, Lee JK, Witte ON. Clinical targeting of mutated and wild-type protein tyrosine kinases in cancer. Molecular and Cellular Biology. 2014;MCB-01592
  64. 64. Gross S et al. Targeting cancer with kinase inhibitors. The Journal of Clinical Investigation. 2015;125(5):1780-1789
  65. 65. Paul MK, Mukhopadhyay AK. Tyrosine kinase—Role and significance in cancer. International Journal of Medical Sciences. 2004;1(2):101
  66. 66. Shchemelinin I, Sefc L, Necas E. Protein kinases, their function and implication in cancer and other diseases. Folia Biologica. 2006;52(3):81
  67. 67. Bhullar KS et al. Kinase-targeted cancer therapies: Progress, challenges and future directions. Molecular Cancer. 2018;17(1):48
  68. 68. Lemmon MA, Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell. 2010;141(7):1117-1134
  69. 69. Kaistha BP et al. Key role of dual specificity kinase TTK in proliferation and survival of pancreatic cancer cells. British Journal of Cancer. 2014;111(9):1780
  70. 70. Duncan JS et al. Regulation of cell proliferation and survival: Convergence of protein kinases and caspases. Biochimica et Biophysica Acta (BBA): Proteins and Proteomics. 2010;1804(3):505-510
  71. 71. Cain RJ, Ridley AJ. Phosphoinositide 3-kinases in cell migration. Biology of the Cell. 2009;101(1):13-29
  72. 72. Huang C, Jacobson K, Schaller MD. MAP kinases and cell migration. Journal of Cell Science. 2004;117(20):4619-4628
  73. 73. Regad T. Targeting RTK signaling pathways in cancer. Cancers. 2015;7(3):1758-1784
  74. 74. Maurer G, Tarkowski B, Baccarini M. Raf kinases in cancer—Roles and therapeutic opportunities. Oncogene. 2011;30(32):3477
  75. 75. Xu J et al. Comparison of FDA approved kinase targets to clinical trial ones: Insights from their system profiles and drug-target interaction networks. BioMed Research International. 2016
  76. 76. Steinberg M. Dasatinib: A tyrosine kinase inhibitor for the treatment of chronic myelogenous leukemia and Philadelphia chromosome—Positive acute lymphoblastic leukemia. Clinical Therapeutics. 2007;29(11):2289-2308
  77. 77. Kantarjian H et al. Nilotinib in imatinib-resistant CML and Philadelphia chromosome–positive ALL. New England Journal of Medicine. 2006;354(24):2542-2551
  78. 78. Schultz KR et al. Improved early event-free survival with imatinib in Philadelphia chromosome–positive acute lymphoblastic leukemia: A children's oncology group study. Journal of Clinical Oncology. 2009;27(31):5175
  79. 79. Chesi M et al. Frequent translocation t (4; 14)(p16. 3; q32. 3) in multiple myeloma is associated with increased expression and activating mutations of fibroblast growth factor receptor 3. Nature Genetics. 1997;16(3):260
  80. 80. De Menezes, Lopes DE, et al. CHIR-258: A potent inhibitor of FLT3 kinase in experimental tumor xenograft models of human acute myelogenous leukemia. Clinical Cancer Research. 2005;11(14):5281-5291
  81. 81. Trudel S et al. CHIR-258, a novel, multitargeted tyrosine kinase inhibitor for the potential treatment of t (4; 14) multiple myeloma. Blood. 2005;105(7):2941-2948
  82. 82. Fabian MA et al. A small molecule–kinase interaction map for clinical kinase inhibitors. Nature Biotechnology. 2005;23(3):329
  83. 83. Yang JC-H et al. Afatinib versus cisplatin-based chemotherapy for EGFR mutation-positive lung adenocarcinoma (LUX-lung 3 and LUX-lung 6): Analysis of overall survival data from two randomised, phase 3 trials. The Lancet Oncology. 2015;16(2):141-151
  84. 84. Soria J-C et al. Afatinib versus erlotinib as second-line treatment of patients with advanced squamous cell carcinoma of the lung (LUX-lung 8): An open-label randomised controlled phase 3 trial. The Lancet Oncology. 2015;16(8):897-907
  85. 85. Kato T et al. Afatinib versus cisplatin plus pemetrexed in Japanese patients with advanced non-small cell lung cancer harboring activating EGFR mutations: Subgroup analysis of LUX-lung 3. Cancer Science. 2015;106(9):1202-1211
  86. 86. Jackisch C et al. Subcutaneous versus intravenous formulation of trastuzumab for HER2-positive early breast cancer: Updated results from the phase III HannaH study. Annals of Oncology. 2014;26(2):320-325
  87. 87. Pitoia F, Jerkovich F. Selective use of sorafenib in the treatment of thyroid cancer. Drug Design, Development and Therapy. 2016;10:1119
  88. 88. Ilic I, Jankovic S, Ilic M. Bevacizumab combined with chemotherapy improves survival for patients with metastatic colorectal cancer: Evidence from meta analysis. PLoS One. 2016;11(8):e0161912
  89. 89. Chaturvedi MM et al. NF-κB addiction and its role in cancer:‘one size does not fit all’. Oncogene. 2011;30(14):1615
  90. 90. Pianetti S et al. Her-2/neu overexpression induces NF-κB via a PI3-kinase/Akt pathway involving calpain-mediated degradation of IκB-α that can be inhibited by the tumor suppressor PTEN. Oncogene. 2001;20(11):1287
  91. 91. Keats JJ et al. Promiscuous mutations activate the noncanonical NF-κB pathway in multiple myeloma. Cancer Cell. 2007;12(2):131-144
  92. 92. Lin K et al. The role of B-RAF mutations in melanoma and the induction of EMT via dysregulation of the NF-κB/snail/RKIP/PTEN circuit. Genes & Cancer. 2010;1(5):409-420
  93. 93. Li W et al. The relationship between BRAFV600E, NF-κB and TgAb expression in papillary thyroid carcinoma. Pathology-Research and Practice. 2017;213(3):183-188
  94. 94. Fuchs O. Targeting of NF-kappaB signaling pathway, other signaling pathways and epigenetics in therapy of multiple myeloma. Cardiovascular & Haematological Disorders-Drug Targets. 2013;13(1):16-34
  95. 95. Mattmann ME, Stoops SL, Lindsley CW. Inhibition of Akt with small molecules and biologics: Historical perspective and current status of the patent landscape. Expert Opinion on Therapeutic Patents. 2011;21(9):1309-1338
  96. 96. Hussain AR et al. Cross-talk between NFkB and the PI3-kinase/AKT pathway can be targeted in primary effusion lymphoma (PEL) cell lines for efficient apoptosis. PLoS One. 2012;7(6):e39945
  97. 97. Saha D et al. Combinatorial effects of VEGFR kinase inhibitor axitinib and oncolytic virotherapy in mouse and human glioblastoma stem-like cell models. Clinical Cancer Research. 2018;clincanres-1717
  98. 98. Ahronian LG, Corcoran RB. Strategies for monitoring and combating resistance to combination kinase inhibitors for cancer therapy. Genome Medicine. 2017;9(1):37
  99. 99. Asim BM et al. Intratumoral heterogeneity and chemoresistance in nonseminomatous germ cell tumor of the testis. Oncotarget. 2016;7(52):86280
  100. 100. Brooks MD, Burness ML, Wicha MS. Therapeutic implications of cellular heterogeneity and plasticity in breast cancer. Cell Stem Cell. 2015;17(3):260-271
  101. 101. Prasetyanti PR, Medema JP. Intra-tumor heterogeneity from a cancer stem cell perspective. Molecular Cancer. 2017;16(1):41
  102. 102. Dragu DL et al. Therapies targeting cancer stem cells: Current trends and future challenges. World Journal of Stem Cells. 2015;7(9):1185
  103. 103. Hoesel B, Schmid JA. The complexity of NF-κB signaling in inflammation and cancer. Molecular Cancer. 2013;12(1):86
  104. 104. Han X et al. Phosphorylation of mini-chromosome maintenance 3 (MCM3) by Chk1 negatively regulates DNA replication and checkpoint activation. Journal of Biological Chemistry. 2015:jbc-M114
  105. 105. Ryo A et al. Regulation of NF-κB signaling by Pin1-dependent prolyl isomerization and ubiquitin-mediated proteolysis of p65/RelA. Molecular Cell. 2003;12(6):1413-1426
  106. 106. Anrather J, Racchumi G, Iadecola C. Cis-acting element-specific transcriptional activity of differentially phosphorylated nuclear factor-κB. Journal of Biological Chemistry. 2005;280(1):244-252
  107. 107. Duran A, Diaz-Meco MT, Moscat J. Essential role of RelA Ser311 phosphorylation by ζPKC in NF-κB transcriptional activation. The EMBO Journal. 2003;22(15):3910-3918
  108. 108. Yeh PY et al. Suppression of MEK/ERK signaling pathway enhances cisplatin-induced NF-κB activation by protein phosphatase 4-mediated NF-κB p65 Thr dephosphorylation. Journal of Biological Chemistry. 2004;279(25):26143-26148
  109. 109. Buss H et al. Phosphorylation of serine 468 by GSK-3β negatively regulates basal p65 NF-κB activity. Journal of Biological Chemistry. 2004;279(48):49571-49574
  110. 110. Rocha S et al. Regulation of NF-κB and p53 through activation of ATR and Chk1 by the ARF tumour suppressor. The EMBO Journal. 2005;24(6):1157-1169
  111. 111. Bae JS et al. Phosphorylation of NF-κB by calmodulin-dependent kinase IV activates anti-apoptotic gene expression. Biochemical and Biophysical Research Communications. 2003;305(4):1094-1098
  112. 112. Sabatel H et al. Phosphorylation of p65 (RelA) on Ser547 by ATM represses NF-κB-dependent transcription of specific genes after genotoxic stress. PLoS One. 2012;7(6):e38246

Written By

Matthew Martin, Antja-Voy Hartley, Jiamin Jin, Mengyao Sun and Tao Lu

Submitted: 24 April 2018 Reviewed: 19 December 2018 Published: 24 April 2019