Open access peer-reviewed chapter

The Role of Kinase Inhibitors in Cancer Therapies

Written By

Gizem Kursunluoglu, Duygu Erdogan, Elcin Cagatay, Esra Bulut Atalay, Seminay Guler, Yonca Gungor and Hulya Ayar Kayali

Reviewed: 25 June 2021 Published: 11 October 2021

DOI: 10.5772/intechopen.99070

From the Edited Volume

Protein Kinases - Promising Targets for Anticancer Drug Research

Edited by Rajesh Kumar Singh

Chapter metrics overview

450 Chapter Downloads

View Full Metrics

Abstract

Protein kinases are enzymes that transfer a phosphate group to the threonine, serine, or tyrosine residues of the target protein, regulating its activity. The activity of these enzymes are very important and strictly regulated in the cell as they promote cell proliferation, survival, and migration. In the case of any dysregulation of these enzymes, they can be associated with cancer initiation and progression. Small-molecule kinase inhibitors approved by the FDA for their improved clinical benefits are currently used in targeted therapy for the treatment of various cancers. So far, there are 62 FDA-approved therapeutic agents targeting different protein kinases, eight of which were approved in 2020. Today, kinase inhibitors are used as FDA approved cancer agents and newly developed ones are evaluated in clinical trials. Those protein kinase inhibitors can be grouped as growth factor receptor inhibitors, Ras/Raf/Mek inhibitors, phosphoinositide 3-kinase (PI3K) and cyclin dependent kinase inhibitors, other targets, and agents such as protein kinase c and 3 phosphoinositide-dependent kinase 1. In this chapter, these kinases, their pathways, and their inhibitors will be discussed in detail.

Keywords

  • cancer therapy
  • clinical use
  • inhibitor
  • protein kinase
  • drug

1. Introduction

Protein kinases (PKs) are enzymes that regulate the activity of a protein by adding phosphate group to specific amino acids using ATP as a resource, thereby resulting in a conformational change of that protein. More specifically, they add terminal γ-phosphate group to the serine, threonine or tyrosine residues of the target proteins and this process is called phosphorylation. Phosphorylation of a protein changes its activity, location or downstream function that can result in amplification of the first signal [1]. Furthermore, phosphorylation can also alter biological activities such as transcription and translation. In addition, it can have inhibitory or stimulatory effect on the target. In either case, it has a significant role in regulation of cellular activities.

PK family is one of the largest protein family comprising more than 500 different kinases in the human proteome that is encoded by 2% of the human genome [2]. PKs in regulating protein activity have a massive effect on cell signaling pathways involved in metabolism, immune responses, growth, differentiation, migration, and adaptation.

Because of the potency of PKs, changes in their expression levels or patterns can impact various pathways resulting in disease development including cancers, as well as metabolic and developmental disorders.

PKs are tightly regulated to restrain their potency and any dysregulation could result in a diseased state. Either a kinase itself or the pathway that the kinase is regulated is mutated in many cancers [3, 4, 5] and those PKs can be classified as proto-oncogenes. In fact, in 1978, the first identified proto-oncogene encodes a tyrosine kinase called c-Src [6]. Overexpression or dysregulation of oncogenic kinases results in altered signaling pathways and oncogenic transformation. In other words, any perturbation on the regulation of oncogenic kinases can result in anchorage-independent uncontrolled growth and proliferation as well as angiogenesis and metastasis. Exploring the role of kinases in cancer development and progression have highlighted the potential of kinase targeted therapies. In this chapter, we will focus on the kinases’ role in oncogenic pathways and the kinase inhibitors targeting these pathways as a therapeutic option.

Protein kinase inhibitors (PKIs) are antineoplastic substances that are used to block the constant or overactivity of dysregulated protein kinases. The first developed PKI called imatinib mesylate (Gleevec, STI571, or CP57148B) targets a fusion protein called “BCR-ABL” which is a constantly active tyrosine kinase that is observed in chronic myeloid leukemia (CML). In addition, imatinib directly inhibits ARG, KIT, and PDGFR tyrosine kinases and used to treat blood neoplasia other than CML and solid tumors that stem from activation of these tyrosine kinases. Consequently, the remarkable success of imatinib had a major impact on researchers to focus on the development of other targeted PKIs that could be potential cancer therapeutics. Following imatinib, other BCL-ABL inhibitors such as nilotinib, dasatinib, bosutinib and ponatinib were developed to overcome imatinib-resistant mutants [7].

The success of imatinib had opened the door to exploration of other oncogenic kinases in other signaling pathways. Those kinase inhibitors can be grouped as growth factor receptor inhibitors, Ras/Raf/Mek inhibitors, phosphoinositide 3-kinase (PI3K) and cyclin dependent kinase inhibitors, other targets and agents such as protein kinase c and 3 phosphoinositide-dependent kinase 1 (PDK1). This chapter will focus on the kinases’ role in oncogenic pathways and the kinase inhibitors targeting these pathways as a therapeutic option.

Advertisement

2. Growth factor receptors

Development, growth and homeostasis of multicellular organisms are controlled by growth factors. Growth factors show their activity by binding to their receptors and are required for cell–cell communications for embryonic tissue induction, cell survival, fate determination, apoptosis, cell migration and tissue specialization. These receptors transmit extracellular signals to the intracellular region in two ways: activation of intracellular transporters or direct translocation of the receptor to the nucleus [8].

The feature of cancer is its continuous growth. Growth factors include epidermal growth factor (EGF), vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), transforming growth factor (TGF), insulin-like growth factor (IGF) and platelet-derived growth factor (PDGF) and they stimulate proliferation, migration and invasion of cancer cell and stromal cell, thereby regulating tumor growth, angiogenesis and metastasis (Figure 1) [9]. Table 1 shows molecules that inhibits growth factor receptors.

Figure 1.

Kinase inhibitors and their inhibition pathways. Cetuximab binds to EGFR with a higher affinity than EGF and competitively inhibits its binding. Other drugs compete with ATP, inhibits the autophosphorylation process. 1st generation EGFR inhibitors gefitinib, erlotinib and icotinib are not effective against the receptors containing exon 19 deletion and L858R mutations. On the other hand, 3rd generation drug osimertinib is effective against EGFR containing exon 19 deletion, L858R and T790M mutations. Bevacizumab binds to VEGFR ligand VEGF, inhibiting its binding with VEGFR. Other inhibitors of VEGFR, FGFR, IGF-R1 and PDGFR target autophosphorylation mechanism, thereby prohibiting the downstream activation of other proteins.

Table 1.

Structures of growth factor receptors inhibitors.

2.1 Epidermal growth factor receptor

One of the most studied drugs in human cancer is the EGF family of receptor tyrosine kinases (RTKs), also called ErbB or HER receptors [8]. Family of EGF receptor (EGFR) comprises four receptor proteins: ErbB-1/EGFR-1 to −4 (also called HER 1–4). These proteins are expressed on cell surface and have alike structure: an intracellular domain with kinase activity, a transmembrane domain and an extracellular domain with ligand binding site [10]. The binding of EGFR ligand initiates homo- and heterodimers between receptors, activating cascades of mitogenic and anti-apoptotic signal [11].

The dimerization is required to activate the intracellular tyrosine kinase domain and C-terminal tail phosphorylation. Its autophosphorylation then promotes either directly or by adaptor proteins, the signal transduction pathways including Ras/MAPK, PI(3)kinase/Akt, PLCg1/PKC, and STAT pathways [8] in modulating differentiation, cellular proliferation, and survival [11].

EGFR has been extensively studied for cancer therapeutics due to mutations, deletions, and overexpression in tumors [11]. EGFR is generally investigated in non-small cell lung cancer (NSCLC): its amplification is ∼80% and its mutation is 20% [12, 13]. When compared to chemotherapy used in NSCLC patients, EGFR TKIs have more than twice the progression-free survival (PFS) with actionable mutations of EGFR. Moreover, they have better objective response rates (ORRs), response time, life quality, and reduction in treatment- associated toxicity [14]. First generation EGFR-tyrosine kinase inhibitors (TKIs) bind reversibly to EGFR and block ATP-TK domain binding and this blockage causes cell death by inhibiting cell proliferation [15]. Some examples of these EGFR-TKIs are gefitinib, erlotinib, and icotinib [14]. When looked at the second-generation EGFR-TKIs these irreversibly bind to EGFR. Afatinib and dacomitinib can be given examples as second generation [14]. Although dacomitinib has improved median PFS, hazard ratio, and median overall survival [16, 17] compared to first-line treatment, the usage of afatinib and dacomitinib in clinical practice may be limited due to having increased toxicities [18].

EGFR T790M mutation causes almost 50% resistance to EGFR-TKIs of first and second generations [19]. As an EGFR TKI of third-generation, osimertinib inhibits both EGFR T790M mutations and EGFR-sensitizing [20] and it is promising for central nervous system (CNS) metastatic patients which is due to EGFR mutation in NSCLC patients [20] or after treatment with EGFR-TKIs of first- or second-generation [21]. Other EGFR TKIs as a third-generation for advanced NSCLC with mutation in EGFR T790M are almonertinib [22], furmonertinib (AST2818) [23], BPI-7711 [24], lazertinib (YH25448) [25], and nazartinib (EGF816) [26], and they have exhibited acceptable safeties and promising efficacies. Almonertinib was also approved by China National Medical Products Administration (NMPA).

Monoclonal antibodies (mAbs), cetuximab and panitumumab, have been used against EGFR and are approved for metastatic colorectal cancer (CRC) treatment. Only small patient subgroups to cetuximab and panitumumab indicate clinical benefit. The best response to cetuximab and panitumumab is seen in patients with the combination of wild type KRAS, BRAF, and PIK3CA and PTEN protein express [27]. Trastuzumab is another mAb used in EGFR/HER2 pathway for the treatment of HER2-positive breast cancers [28].

2.2 Vascular endothelial growth factor receptor

Vascular endothelial growth factors (VEGF) are a family of polypeptides contain highly conserved receptor binding domain in disulfide-node [29]. There are two types of VEGF; VEGF-A and VEGF-B, which bind their receptors named RTK. VEGF members display plural interactions with RTKs are the critical factors for blood vessel formation which cause cord formation and tubulogenesis, differentiation of endothelial cells, proliferation, migration. In mammals, the vascular endothelial growth factor receptor (VEGFR), transmembrane tyrosine kinase receptors, comprises three members VEGFR-1 (Flt-1), VEGFR-2 (KDR/Flk-1) and VEGFR-3 (Flt-4), and control the generation of blood and lymphatic vessels [30].

VEGFRs have seven immunoglobulin (Ig)-like domains on the extracellular site and two split tyrosine kinase domains in the intracellular site [10]. In adults, while the vascular endothelial cells are posess largely VEGFR-1 and VEGFR-2 in their structure, the lymphatic endothelial cells have VEGFR-3 [31]. The literature shows that VEGF pathway is critical for renal cell carcinoma (RCC) initiation and progression, and VEGFRs targeted TKIs have been used for most favorable RCC treatment strategy.

In recent years, VEGFR-associated multi-targeted TKIs have been revealed as antı-tumor agents for cancer treatment [18]. VEGFR-targeted therapeutic agents have become the main element used for RCC patients treatment [31]. The FDA has approved 6 small molecules, named as sunitinib, sorafenib, axitinib, pazopanib, cabozantinib and lenvatinib, that inhibit VEGFR1/2/3 for use in RCC treatment. Additionally, for clinical trials, seven VEGFR inhibitors; vandetanib, vorolanib, anlotinib, MGCD516, regorafenib, tivozanib, and apatinib are under review [31].

Not only VEGFRs targeted and block by these drugs, also some other receptors generally overexpressed in RCC, platelet-derived growth factor (PDGF) receptors a and b (PDGFR-a/b), stem cell factor receptor (c-KIT), FMS-like tyrosine kinase-3 (FLT3), tyrosine protein kinase MET, rearranged during transfection (RET), tyrosine kinase that contains immunoglobulin-like loops and epidermal growth factor-similar domains-2 (Tie2), fibroblast growth factor receptor (FGFR), the GAS6 receptor (AXL), serine/threonine protein kinase Raf-1, colony stimulating factor1 receptor (CSF1R), and discoidin domain receptor (DDR) targeted by these small drugs. In addition, growth, proliferation and angiogenesis of tumor cells are significantly inhibited by the effects of VEGFR1/2/3 inhibitors. The humanized IgG1 monoclonal antibody approved in 2009, Bevacizumab can bind VEGF and for use in first-line therapy for RCC in combination with IFN-α [32].

Besides, in hepatocellular carcinoma (HCC), the efficiency of VEGFR-associated multi-targeted TKIs was demonstrated. Especially Sorafenib can target VEGFR, PDGFR, FGFR, and other signaling targets and prefered as first line therapy in inoperable cases [33, 34]. In phase II/III studies of Donafenib, VEGFR-associated multitargeted TKI, is shown a higher overall survival rate than sorafenib in advanced HCC cases [35]. Moreover, regorafenib [36], apatinib [37] and cabozantinib [38] are used for second-line treatment of HCC. More recently, it is shown that Pembrolizumab is anti programmed cell death protein-1 (PD-1) antibody and lenvatinib combination therapies demonstrated promising anti-tumor effect on untreated/inoperable HCC patients and this combination named as breakthrıough therapy by FDA [39].

For lung cancer therapy (on NSCLC [40] and SCLC [41]), Anlotinib showed promising results and was approved for further-line therapy by(National Medical Products Administration of China (NMPA) [18]. Furthermore, similar results identified in thyroid carcinoma and soft tissue sarcoma (STS) patients [42].

2.3 Fibroblast growth factor receptor

Fibroblast growth factor receptors (FGFRs), which are members of receptor tyrosine kinase can be thought as single pass membrane proteins due to the cellular membrane covering in a single region [43]. There are four transmembrane proteins (FGFR1–4) for the FGFR family and different isoforms of them with changed ligand specificity [10]. Upon binding of the different FGF ligands to different FGF receptors, auto-phosphorylation and kinase activation are initiated by FGFR dimerization. This binding causes cell death inhibition and uncontrolled growth, respectively due to downstream anti-apoptotic PI3K/AKT signals and mitogenic growth signals (MAPK) so the interaction FGF-FGFR plays a critical role in tumorigenesis. Moreover, the downstream of PLC/PKC pathway joins the MAPK pathway to promote cell growth [10].

The use of FGFR pathway downstream molecules as targets in anti-cancer drugs has attracted attention, and these drugs are small compounds and antibodies against cancer driver mutations in FGFRs and related signaling molecules [43]. Most FGFR-TKIs belong to multiple target TKIs [18].

FGFR2 changes are associated with the cholangiocarcinoma (CCA). Pemigatinib, a FGFR1–3 TKI, has been accepted for the treatment of locally advanced or metastatic CCA [44]. Some other FGFR-TKIs (such as futibatinib, infigratinib) also have the potential to be used for CCA based on their results [45].

In addition to CCA, FGFR2 changes also play a role in urothelial carcinoma (UC). Fusions and mutations in FGFR2/3 are seen in 20% of patients with UC [46]. Treatment with erdafitinib, a FGFR1–4 TKI, has been accepted for the adult patients with previously treated metastatic or locally advanced FGFR2/3-mutated UC. Response was more promising as second-line therapy for advanced UC compared to antibody-drug conjugates such as enfortumab vedotin or sacituzumab govitecan [47] and pembrolizumab [48]. In addition, other inhibitors of pan-FGFR are investigated and for example infigratinib (BGJ 398) is examined in the treatment of UC holding a FGFR3 mutation [47].

2.4 Transforming growth factor-β receptor

Transforming growth factor-beta (TGF-β) is a cytokine which has different functions and modulates cell growth and differentiation, extracellular matrix production, apoptosis, angiogenesis, cell motility, and cellular immune responses. Interestingly, TGF- β shows different effects on tumorigenesis. Although it acts as a tumor suppressor in the early stages, it advances tumor growth by producing a more suitable environment for tumor invasion and metastasis in later stages [49].

There are three membrane receptors for the TGF-β receptor (TGF-βR) family: TβRI, TβRII and TβRIII. Their expression in various cell types control different cellular functions by altering signals upon ligand binding of TGF-β [10]. When high binding affinity is established between activated TGF-β and TβRII signaling is started. The binding needs altering the conformation of TβRII by engagement of TβRIII [49]. Receptors transphosphorylation is triggered after binding is created between TGF-β ligand and transmembrane receptor serine/threonine kinase (type I and II) complex. After that, SMAD proteins in C-terminal serine are phosphorylated by activated receptors. SMAD complexes which are activated control target genes transcription by migrating to the nucleus [50], thereby controlling cell proliferation, migration, survival, and differentiation [10].

TGF-β can signal via intracellular Smad proteins and some Smad independent pathways involving ERK, MAP kinase, PI3K, JNK, p38, and AKT [49]. The Smad pathway is very important in the antiproliferative properties of TGF-β and modifications in the Smad system by missense mutations [51, 52]. Moreover, blocking of the phosphorylation process or Smad 2/3 complex formation has been demonstrated to be effective in tumor development [53]. The TGF-β overexpression has been determined in many tumors containing cancers of the breast, colon, liver, stomach, lung, esophagus, kidney, prostate, pancreas, brain, and malignant melanoma, as well as certain hematological malignancies [54, 55, 56, 57, 58, 59, 60, 61]. Some small-molecule TKIs of TβR II and TβR III can block the signaling pathway of TGF-β-mediated receptor.

Galunisertib (LY2157299) monohydrate has shown powerful as a TβR I inhibitor by reducing Smad2 phosphorylation in pancreatic, colorectal cancer, lung cancer [62] and ovarian cancer [63]. LY2157299 is another promising inhibitor of Smad2 phosphorylation for hepatocellular carcinoma models [64]. It moved to Phase II and was approved by the FDA for liver cancer as an orphan drug in 2013. LY2109761 for metastatic NSCLC [65], colorectal cancer [66] could also investigated as a kinase inhibitor. In addition, Ki26894 blocks Smad2 phosphorylation by binding to the TβR I-ATP domain and its activity was demonstrated in breast cancer [67] and gastric cancer [68].

2.5 Insuline-like growth factor receptor

The insulin-like growth factor receptor (IGFR) family includes two cell membrane receptors, named as IGF-IR and IGF-IIR. IGF-IR (that also forms a heterodimer with the insulin receptor [IR]) has higher affinity to insulin-like growth factor 1 (IGF-I) but IGF-II comparatively has lower affinity. Although IGF-IR, receptor tyrosine kinase, has the triggering effects on IGF-I and IGF-II and thus on cell proliferation, migration and invasion, IGF-IIR lacks kinase activity [9].

Pathways activated by the presence of IGF and progressive pathological and physiological processes take place through proper receptors, named as IGFR. The mature IGF-IR have homodimer structure, containing α2 and β2 chains bounded with disulfide bonds. The intracellular domain is autophosphorylated by the binding of ligands and the downstream processes continue with the activation of a number of proteins. In several carcinomas, proliferation, transformation and metastasis are induced by overexpression of IGFIR genes. IGF-IIR, called mannose-6 phosphate receptor, M6P, is formed as a single polypeptide chain and performs as a “scavenger receptor” that suppresses tumor growth, modulates invasiveness, and blocks angiogenesis. Mutated IGFIIR locus could be found in lung cells and early phase hepatocellular carcinoma [10].

Especially, cancer associated macrophages, tumor cells and liver cells secrete IGF. The increased risk of breast and prostate cancer is correlated with high levels of IGF in the circulating system [69, 70]. For smokers,there is a moderate correlation between IGF level and lung cancer risk [71]. IGF-I is not related to colorectal cancer but takes increased risk for colorectal cancer [72]. Multiple signaling pathways of PI3K/Akt, JAK/STAT, MAPK, Src and focal adhesion kinase (FAK) lead to the proliferation, survival, and migration of cancer cells by binding IGF to its appropriate receptor, are activated.

In many human tumors, especially mesenchymal, epithelial and hematopoietic cancers, the growth of cancer cells, metastasis and the formation of drug resistance can be associated with the activation of IGF signaling pathways [73, 74, 75].

The IGF-IR has been evaluated as a target protein in cancer treatment. There are some small-molecule inhibitors and anti-IGF-IR mAbs used in pre-clinical models and clinical trials. A dual IGF-IR/Insulin receptor inhibitor, linsitinib (OSI-906), is investigated in phase II in recurrent small cell lung cancer patients. However, clinical activity failed to show improvement in small cell lung cancer, metastatic or advanced adrenocortical carcinoma, metastatic colorectal cancer, advanced NSLC, gastrointestinal stromal tumors and metastatic prostate cancer. AXL1717 is studied in early phase with relapsed malignant astrocytomas and can show long-term stable disease and patients’ survival [9].

2.6 Platelet derived growth factor receptor

Platelets produce platelet-derived growth factor (PDGF) and are secreted from both epithelial and mesenchymal cells [76]. The PDGF family has five isoforms (PDGF AA, BB, AB, CC, and DD) that bind to two RTKs, PDGFRα and PDGFRβ. After activation of PDGFRα and β, they promote cell proliferation, migration and survival through initiating signaling pathway with the inclusion of the extracellular signal-regulated kinase 1/2 (ERK) and phosphatidylinositol 3-kinase (PI3K)/AKT [77].

Overexpressed platelet-derived growth factor receptor (PDGFR) is associated with the formation of various human tumors like glioma, neurofibroma, ovarian cancer, prostate cancer, and non-small cell lung carcinoma. In addition, PDGF accelerates angiogenesis by increasing VEGF expression and development of cancer-related fibroblasts that directly or indirectly affect tumor formation. Moreover, PDGF plays a role in gene amplification [76].

To enhance anti-angiogenesis effect and suppress tumor growth PDGFR is targeted by most VEGFR-related multiple kinase inhibitors such as sorafenib, sunitinib, regorafenib, lenvatinib, axitinib, pazopanib, anlotinib, famitinib, donafenib and cediranib. These inhibitors can be critical to treat various cancer types [18].

Advertisement

3. Ras/Raf/MEK pathway inhibitors

Ras/Raf/MEK pathway inhibitors are important as kinase inhibitors in various cancer treatment. Firstly, the Ras protein known as Ras GTP-binding protein is the member of small G protein family, which has an important role in transmitting growth factor and relay signals from activated growth factor receptors (GFR) (Figure 2).

Figure 2.

Downstream molecular pathways that are activated upon stimulation of receptor tyrosine kinases (RTKs). The main pathways include protein kinase C (PKC), Ras–Raf–Mek and PI3K pathways.

Ras proteins are regulated by a GDP-GTP cycle, different form is carried out when bound to GDP or GTP, inactive form is Ras-GDP and active form is Ras-GTP [78]. Therefore this cycle has been stimulated via receptor activation then Ras proteins bind to GTP (active form), cellular proliferation and other effects are promoted [79]. Ras protein associate with cell transformation such as cell growth, differentiation, apoptosis, cell migration, this Ras regulation is carried out by modulating some signaling molecules by translocating them to the plasma membrane for activation. Since Ras proteins play critical role as a branch point in signal transduction and orchestrate the activity of multiple signaling pathways such as Raf/MEK/ERK [78].

Numerous of the signal transduction pathways are occurred through protein kinases regulating of cellular metabolism [80]. One of the most important key protein kinases are Ras, Raf and MEK for targeting of the anticancer drug and also, Ras mutations are quite common as they are identified in about 30% of cancers [81, 82, 83]. Ras protein activations are depended on the cancer type, for instance N-Ras in lymphoid and myeloid cancers, K-Ras in colon and pancreatic cancers, H-Ras in bladder and kidney cancers [84]. One of the main reasons for this situation is thought to be the disorder in protein kinase activity. Thus, the protein kinase enzyme family is seen as one of the most important drug targets of the 21st century in many diseases, especially cancer, and there are 62 FDA-approved therapeutic agents targeting different protein kinases, eight of which were approved in 2020 [85]. Thus, today, kinase inhibitors are both used as FDA approved cancer agents and evaluated in clinical trials. Protein kinase inhibitors related to the Ras / Raf / MEK pathway, which is the subject of this part, are structurally classified as small molecules and antisense oligonucleotides and structure of Ras/Raf/MEK kinase inhibitors is shown in Table 2.

Table 2.

Structures of Ras–Raf–MEK kinase inhibitors.

3.1 Ras kinase inhibitors

Ras protein, which has the ability to induce different growth and proliferation pathways of the cell, is located in the cell membrane. And most importantly, overactivation of mutated Ras can induce to make progress in cancer. Farnesyl Transferase enzyme catalyzes the first step of the reaction in the posttranslational modification of both the normal and mutated Ras gene, thus it is easier to settle on the cell membrane. FTase, one of the tansferase enzymes, targets Ras proteins, adding a farnesyl group to it. After the essential transfer step, the activity of the Ras protein has been inhibited by Ftase [86, 87]. Therefore, FTase enzyme and the downstream pathways of the Ras protein are considered to be one of the most important groups of targets that can be medicated in cancer therapy [88]. The potential anti-cancer agent effect of Farnesyl Transferase inhibitors acting as kinase inhibitors which is affecting Ras and Ras downstream will be reviewed in this part. Various FTase inhibitors such as tipifarnib/R115777, lonafarnib/SCH66336, L-778123 and BMS-214662 have been used primarily in clinical trials and phase studies [89].

The antitumor activity of tipifarnib, a potent and selective farnesyltransferase inhibitor, also known as R115777, has been evaluated the antiproliferative effect of tipifarnib, 53 human tumor cell lines have been studied and 75% were obtained to be sensitive to R115777 [90, 91]. The strong potency effect of Tipifarnib was detected in SU86.86 human pancreatic cells, CAPAN-2 human pancreatic cells and NCI-H441 human lung cells and these cells espacially include the KRAS 12 mutation cells [90, 91]. Tipifarnib has been studied as a phase 2 study on 249 adult patients with refractory urothelial carcinoma (UC) and HRAS mutation. This study shows that tipifarnib is effective in previously treated metastatic UC patients by inhibiting the processing of newly synthesized proteins [92]. In refractory advanced colorectal cancer as a Phase III study, it had an acceptable toxicity profile and was well tolerated but did not improve overall survival according to the best supportive [93], and R115777 was not effective in metastatic colorectal cancer patients in another Phase II study [94]. Another phase two studies of R115777 emphazied that antitumor properties were not observed in metastatic c pancreatic cancer and did not improve overall survival in advanced non-small cell lung cancer [95, 96]. Farnesyl transferase inhibitor R115777, one of the protein kinase inhibitors, which are thought to be effective in the Ras pathway, has being continuing to be evaluated different combination therapies studies as well as single therapy [97].

Lonafarnib (SCH66336) as a clinical candidate FTase inhibitor, showed antitumor activity in vivo lung, colon, pancreas, bladder and prostate human tumor xenograft models [98]. Various antiproliferative effects of the SCH66336 agent were observed in eight human astrocytoma cell lines with different concentrations of IC50 values ​​(0.6 mM - 32.3 mM) [99]. Growth inhibitory effects of the drug SCH66336 were observed in human tumor xenografts with various tumor models (colon, lung, pancreatic, prostatic carcinoma and a H-ras transgenic mouse model) [100]. In the other phase 2 studies, Lonafarnib, which was used as a combined treatment with paclitaxel, had clinical benefit and low toxicity in patients with non-small cell lung carcinoma, while the targeted response of Lonafarnib was not observed in 5-fluorouracil and irinotecan-resistant metastatic colorectal cancer and gastrointestinal toxicity was observed in single therapy [97] .

L-778123, a peptidomimetic farnesyl protein transferase (FPTase) inhibitor, was administered to patients with solid malignancies developed in phase 1 studies and their toxicity levels were investigated. While unacceptable toxicity was observed at the doses of L-778123 given as 1120 mg / m2 / day, the dose of 560 mg/m2/day was well tolerated. Also, no objective tumor response was observed following drug administration in this phase 1 study [101]. When the toxicity of L-778123 and radiotherapy combination was evaluated in phase 1 studies, it was found to be at an acceptable level in pancreatic cancer patients [102]. Although there are no important studies on cancer treatment in phase 1, this drug has been stopped in its clinical development due to its severe and unexpected toxicity [103].

3.2 Raf kinase inhibitors

Receptor tyrosine kinase effector Raf derives its name from “Rapidly Accelerated Fibrosarcoma” [104]. There are 3 different isoforms of Raf family related to serine/threonine protein kinases, namely Raf-1, A-Raf and B-Raf [105]. Critical steps are needed for Raf activation to occur, these are Raf-1 phosphorylation, binding of Raf protein to Ras-GTP, oligomerization of Raf protein, interaction of Raf protein with membrane lipids, and conformational changes in Ras-induced Raf protein [106]. Phosphorylation of Raf kinases plays an important role in cell cycle regulation, proliferation and differentiation, cell survival, apoptosis, and many cellular processes [107]. Furthermore, Raf kinase isoforms become overactive in a variety of solid tumors such as renal cell carcinoma (RCC), hepatocellular carcinoma (HCC), non-small cell lung cancer (NSCLC), and papillary thyroid carcinoma. BAY 43–9006 is a potent Raf kinase inhibitor with significant activity in four different types of human tumors, including colon, pancreatic, lung and ovarian tumors. The in vivo study of BAY 43–9006 shows that after 14 days of injection, tumor growth is strongly suppressed in athymic mice modeled with human tumors [107]. It suggests that BAY 43–9006 may have clinical potential as a cancer therapeutic agent with a new mechanism of action with its antitumor activity [107]. One of the other important raf kinase inhibitors is ISIS 5132 (CGP 69846A), for which phase I and II studies have also been performed. This Raf kinase inhibitor provides inhibition of c-Raf mRNA expression and inhibits the proliferation of lung, colon, cervical, prostate and ovarian carcinoma cell lines [105]. According to the phase1 and phase 2 studies performed with ISIS 5132, no significant response occurred in small cell or non-small cell lung carcinoma, hormone-resistant prostate cancer, and Colorectal cancer [105]. There are two FDA approved molecules among Raf Kinase Inhibitors, one of which is Dabrafenib (GSK2118436), which is effective in BRAFV600E/K melanomas, BRAFV600E NSCLC, BRAFV600E anaplastic thyroid cancers, and Encorafenib (LGX818), which is suitable for combination therapy with binimetinib for BRAFV600E/K melanomas [85].

3.3 MEK kinase inhibitors

One of the most important involved in cancer biology downstream targets of Ras is mitogen activated extracellular signal regulated kinase (MEK) [108]. Various MEK inhibitors, one of the first selective inhibitors of mitogen-activated protein kinase (MAPK) pathway activation, were investigated in phase 1 and 2 studies [109]. CI-1040, the first MEK inhibitor participating in the clinical study, was tested on 66 patients, while a partial response was observed in one patient with pancreatic cancer, stable disease was observed in 19 patients with various solid tumors such as non-small cell lung, breast and colon cancer [110]. PD 0325901, a second generation MEK inhibitor, was studied in 27 patients, while a partial response was observed in two patients with melanoma, and stable disease was observed in eight patients with various solid tumors [111]. There are 4 different FDA approved small molecule protein kinase inhibitors, for which “MEK1/2” is the primary target. These FDA approved MEK protein kinase inhibitors, Binimetinib (MEK162), Cobimetinib (GDC-0973), Selumetinib (AZD6224), Trametinib (GSK1120212), are respectively effective in these diseases; Combination therapy with encorafenib for BRAFV600E/K melanomas, melanomas in combination with vemurafenib, Neurofibromatosis type I, BRAFV600E/K melanomas, BRAFV600E NSCLC [85].

Advertisement

4. Phosphoinositide 3-kinase (PI3K) pathway inhibitors

The activation of PI3K, a superfamily of lipid kinases, leads to the production of lipid seconder molecule phosphatidylinositol-3,4,5-trisphosphate (PIP3) which recruits phosphatidylinositide-dependent protein kinase (PDK1) and Akt protein kinases to the plasma membrane. Akt that activated and phosphorylated from PDK1 and mTOR, phosphorylates several target proteins either at the plasma membrane or in the cytosol and nucleus (Figure 2). Cell proliferation controlled by the PI3K pathway is partially dependent on a large kinase protein called mammalian target of rapamycin (mTOR). mTOR complex 1 plays a role in cell growth and survival by stimulating nutrient uptake and metabolism. Akt activates mTOR independently of phosphorylation at complex 1 [112].

PI3K has three different classes (I, II and III) and four different isoforms (α, β, γ, δ). While it has central physiological roles in cancer, diabetes, and aging, the isoform within each class has distinct roles.Several small molecules have been produced with good pharmacological properties that have been tested in various cancers to selectively inhibit PI3K, AKT or, mTOR [113].

Idelalisib and Copanlisip are FDA-approved drugs. Idelalisib has been selectively developed for the delta isoform of PI3K p110 inhibitor. This purine-quinazoline derivative has been used for the treatment of patients with CLL, relapsed follicular B-cell non-Hodgkin lymphoma (NHL), and relapsed small lymphocytic leukemia (SLL) [114]. The combination study with rituximab resulted in a significant increase in the response rate and overall survival on CLL patients [115]. Copanlisip is the inhibitor of the pan-class I PI3K, which shows preferential activity against p110α and p110δ as compared with p110β and p110γ. It has been used for the treatment of various subtypes of indolent and aggressive malignant lymphoma such as NHL, relapsed follicular lymphoma (FL), and CLL in phase II study. This small molecule, administered intravenously, becomes advantageous compared to other FDA-approved drugs, exhibiting a favorable toxicity profile and maintaining efficacy [116].

Rapamycin and derivatives (CCI-779 and RAD001) inhibit both mTOR and the downstream kinase target such as p70S6 kinase (p70S6K) and 4E-binding protein-1 (4E-BP1). The derivatives under clinical study have been shown to suppress the proliferation and growth of various tumor cell lines by blocking the G1-S transition in the cell cycle [117]. Furthermore, the immunosuppressant effect of rapamycin has been observed in several case studies, with transplanted patients reducing the incidence of cancer [118]. In ongoing clinical studies in prostate cancer in which the PI3K/Akt/mTOR pathway is highly expressed, it was observed that rapamycin and its derivatives were well tolerated in prostate cancer patients and decreased mTOR level [119]. In addition to the antiproliferative effect, RAD001 is also used as an immunosuppressant for the treatment of several malignancies in phase I study [120]. CCI779 that is the soluble ester of rapamycin [121] has been used for the treatment of several cancers in phase II study. This drug candidate has been used for T-cell leukemia, prostate, breast, and SCLC, as well as glioma and melanoma cell lines. CCI779 showed apoptosis induction, delaying the growth of the tumor, encouraging survival [105]. In combined treatment with apatinib, the treatment inhibits the proliferation and migration of small cell lung cancer cell lines [122].

Recently synthesized CYH33 is a novel PI3Kα inhibitor in phase 1b clinical trial for esophageal squamous cell carcinoma (ESCC) and breast cancer therapy. In vitro and in vivo studies show the anti-proliferative activity of this drug candidate to cell lines, induction of G1 phase arrest, and down-regulation of phosphorylated ERK in solid tumors [123]. The structure of all PI3K pathway inhibitors is shown in Table 3.

Table 3.

Structures of PI3K pathway inhibitors both in clinical development and FDA approved.

Advertisement

5. Cyclin-dependent kinase (CDK) inhibitors

CDKs found in a family of serine/threonine kinases are key regulators in the various phases of the cell cycle (Figure 3). CDKs ensure the continuation of the cell cycle by phosphorylating critical target proteins that are necessary to proceed to the next stage. Especially cells need to regulate the phosphorylation of kinases to sustain constant division in the presence of abnormal ploidy. Cyclin proteins, differently from CDKs, are synthesized at certain stages of the cell cycle. When the various cyclin form complexes with their target CDKs, it regulates the cell cycle transitions by enabling them to be phosphorylated and activated. For example, both cyclin D/CDK4 and cyclin D/CDK6 complexes direct phosphorylation of the Retinoblastoma gene (RB), which induces the separation of E2F to allow the transcription of the genes necessary for the proceed G1 to S phase transition. CDK inhibitors have been used in cancer therapy to interfere with the limitless replicative potential which is one of the hallmarks of cancer cells [124].

Figure 3.

Cyclin dependent kinases (CDKs) in the cell cycle and their inhibitors. CDKs are a family of serine/threonine kinases and key regulators in the cell cycle.

Palbociclib (PD 0332991) [128], Ribociclib (LEE011) [125], and Abemaciclib (LY2835219) [126] are novel dual inhibitors of both CDK4 and CKD6, that were approved by the FDA. Abemaciclib, more potent against CDK4 (IC50s of 2 nM for CDK4 and 10 nM for CDK6) [126]. These drugs have using for the treatment of postmenopausal women with HR-positive, HER2-negative advanced, or metastatic breast cancer. The action mechanism is that inhibiting the phosphorylation of Rb protein in the G1 phase results in cell cycle arrest [127]. Palbociclib has a synergistic effect in combination therapy with either letrozole or fulvestrant [128]. Abemaciclib also has a synergistic effect with gemcitabine [126].

BMS-387032 (SNS-032) is an aminothiazole, selectively designed for CDKs 2, 7, and 9 (with IC50s of 38 nM, 62 nM, and 4 nM, respectively) inhibition. Its activity was shown to inhibit both the cell cycle and the expression of anti-apoptotic proteins in various carcinoma models [129]. A phase I dose-escalation clinical trial study carried out to evaluate the safety, and clinical efficacy demonstrates limited clinical activity in heavily pretreated CLL and MM patients [130].

CYC202 (Seliciclib) shows potent inhibitors for broad CDK such as Cdk2, Cdk1, Cdk7, and Cdk9 (with IC50s of 0.1, 2.7, 0.5, and 0.8 mM, respectively), competing at their ATP binding sites [131]. This small molecule showed antitumor activity inducing apoptosis in multiple myeloma cell lines and has assayed in phase II study resulted in the dosing schedule being tolerable in nasopharyngeal carcinoma patients [132].

E7070 is a novel chloroindolyl-sulfonamide anticancer agent, which leads to induces arrest at the G1-S boundary and in company with mitigation in the expression of CDK2. E7070 has been assayed for melanoma cancer therapy in phase II and the results showed that CDK activity can be inhibited in tumor cells, but the dose and schedule applied are not suitable for single-agent chemotherapy for melanoma cancer treatment [133].

Flavopiridol has potent strong activity on several CDKs (CDK1, 2, 4, 6, and 7). In addition to controlling the cell cycle, it exhibits more than one action mechanism by showing an antiproliferative effect to leukemias and lymphoma cell lines [134, 135]. Phase I and phase II studies have been tested on various progressive tumors and it has been observed that flavopiridol has no effect on metastatic renal carcinoma [135]. The structure of all CDK kinase inhibitors is shown in Table 4.

Table 4.

Structures of CDK kinase inhibitors both in clinical development and FDA approved.

Advertisement

6. Other protein kinase inhibitors

Protein kinase C (PKC) is a family of serine/threonine kinases. They have important role in regulating a range of cellular functions including gene expression, differentiation, proliferation, cell cycle, apoptosis and cell migration. The PKC family includes 12 isoenzymes that can be divided into three groups depending on activation requirements. Conventional PKCs are calcium dependent and they require negatively charged phospholipid and diacylglycerol for activation. Novel PKCs are calcium independent, as conventional PKCs their activation requires phospholipid and diacylglycerol. Atypical PKCs are both calcium and diacylglycerol independent protein kinase group [136].

Regarding the PKC as a receptor for tumor-promoting phorbol esters, the researcher targets PKC for the potential treatment of cancer.

PKC is activated by phorbol esters and this event prevents cell death. Therefore, the inhibition of PKCα results in apoptosis. PKC activity has been reported to increase in many types of cancer, suggesting that PKC has important role in tumor formation. There are many PKC inhibitor candidates in clinical development for the treatment of cancer (Figure 2) [137].

First class of PKC inhibitors with anti-cancer activity are bryostatins. They are macrocyclic lactones extracted form marine bryozoan Bugula nerutina. Interaction of bryostatins with regulatory domain of PKC causes downregulation of the enzyme. Besides anti-proliferative, apoptotic and cytotoxic effects of Bryostatins on cancer cells, they also have immunomodulatory functions. Bryostatins provide the development of tumor-specific cytotoxic T-lymphocytes and stimulates the release of different cytokines such as TNF, IL-6. Both the immunomodulatory effect and downregulation of PKC has participated in antitumor effect of bryostatins [137, 138].

Second class of PKC inhibitors are staurosporines. Staurosporine is a microbial alkaloid derived from Streptomyces species organisms. In addition to PKC, staurosporine has shown the activity aganist different kinases including, pyruvate dehydrogenase kinase 1 (PDK1), PKA and PTK. Several staurosporines are under in clinical research such as UCN-01 (7-hydroxystaurosporine) or midostaurin (N-benzoyl staurosporine, PKC412) [138]. UCN-01 (7-hydroxy-staurosporine) has been reported to have more PKC inhibitory effects than staurosporine [105, 137]. UCN-01 causes cell cycle arrest in G1 step, thus leading the cell to death.

There is a growing interest in antisense therapy for PKC inhibitors. The structures of PKC inhibitors are shown in Table 5. ISIS 3521, an antisense therapy agent, is a phosphorothioate antisense oligodeoxynucleotide. ISIS 3521 binds to 3′ untranslated region (UTR) of human PKC-α messenger RNA (mRNA). This hybridization is then cleaved by RNase H and resulted in inhibition of PKC-α expression. Based on in vitro and in vivo studies results, ISIS 3521 may be a potential treatment agent for cancer patients [139].

Table 5.

Structures of other kinases’ inhibitors.

Besides PKC inhibitor classes for cancer therapies described above, there are other agents including safingol, quercetin, antiestrogens, and miltefosine. Safingol is a synthetic sphingoid base analogue and safingol was the first to enter clinical trials. Quercetin belongs to flavonoids and widely distributed in nature. Quercetin inhibits different classes of protein kinases including PKC, phosphatidyl inositol-3 kinase. The PKC inhibitory effect of the classical antagonism of estradiol at the estrogen receptor level has been mentioned in many studies. However, the antiestrogen tamoxifen and its analogues have also been shown to inhibit PKC at very low concentrations. Miltefosine is an alkylphosphocholine and it shows its activation by preventing phospholipid metabolism. Miltefosine’s antitumor activity is possibly associated with its abilitiy to inhibit PKC, but there has also been multiple alternative actions [137].

Although most protein kinase C inhibitors target the PKC-α, inhibitors targeting PKC-β have been investigated. PKC-β belongs to major PKC isoform classes. Hyperglycemia activated PKC-β leads to diabetic kidney diseases. LY333531 named as Ruboxistaurin selectively inhibits PKC-β. Recent studies showed that LY333531 significantly reduced PKC activity and PKC-β protein expression in the kidney [140, 141].

Advertisement

7. Conclusions

Protein kinases are potent oncogenes because of their ability to activate or inhibit other proteins. Furthermore, the ability to activate other protein kinases results in producing an exponential signal. In other words, a tiny signal can lead to a huge cellular response. This property of protein kinases makes their strict regulation crucial, and any dysregulation can lead to catastrophic outcomes. Many cancers are caused by the dysregulation of such oncogenic kinases, and inhibitors of those shown to increase overall survival in cancer patients. Although some tumors may gain resistance to some of those protein kinase inhibitors (PKIs), promising results of PKI administered cancer patients have led to developing new PKIs to treat PKI-related cancers [142].

References

  1. 1. Johnson LN and Lewis RJ, Structural basis for control by phosphorylation. Chemical Reviews, 2001.101: p. 2209–42
  2. 2. Manning G, Whyte DB, Martinez R, et al., The protein kinase complement of the human genome. Science, 2002. 298: p. 1912–34
  3. 3. Wu, W., Cho, C., Lee, C., Fan, D., Wu, K., Yu, J., and Sung, J. (2010). Dysregulation of cellular signaling in gastric cancer. Cancer Letters, 295(2), 144-153. DOI: 10.1016/j.canlet.2010.04.025
  4. 4. Ding, L., Wang, H., Lang, W., and Xiao, L. (2002). Protein Kinase C-ε Promotes Survival of Lung Cancer Cells by Suppressing Apoptosis through Dysregulation of the Mitochondrial Caspase Pathway. Journal Of Biological Chemistry, 277(38), 35305-35313. DOI: 10.1074/jbc.m201460200
  5. 5. Nadella, K., and Kirschner, L. (2005). Disruption of Protein Kinase A Regulation Causes Immortalization and Dysregulation of D-Type Cyclins. Cancer Research, 65(22), 10307-10315. DOI: 10.1158/0008-5472.can-05-3183
  6. 6. Stehelin D, Varmus HE, Bishop JM, et al., DNA related to the transforming gene(s) of avian sarcoma viruses is present in normal avian DNA. Nature, 1976.260: p. 170–3
  7. 7. Bitencourt, R., Zalcberg, I., and Louro, I. (2011). Imatinib resistance: a review of alternative inhibitors in chronic myeloid leukemia. Revista Brasileira De Hematologia E Hemoterapia, 33(6), 470-475. DOI: 10.5581/1516-8484.20110124
  8. 8. Wieduwilt MJ, Moasser M. The epidermal growth factor receptor family: biology driving targeted therapeutics. Cellular and Molecular Life Sciences. 2008. 65(10): 1566-1584. DOI: 10.1007/s00018-008-7440-8.
  9. 9. Hua H, Kong Q, Yin J, Zhang J, Jiang Y. Insulin-like growth factor receptor signaling in tumorigenesis and drug resistance: a challenge for cancer therapy. Journal of Hematology & Oncology. 2020. 13(1):64. DOI: 10.1186/s13045-020-00904-3.
  10. 10. Tiash S, Chowdhury EH. Growth factor receptors: promising drug targets in cancer. Journal of Cancer Metastasis and Treatment. 2015. 1:190-200. DOI: 10.4103/2394-4722.163151.
  11. 11. London M, Gallo E. Epidermal growth factor receptor (EGFR) involvement in epithelial-derived cancers and its current antibody-based immunotherapies. Cell biology international. 2020. 44(6): 1267-1282. DOI: 10.1002/cbin.11340.
  12. 12. Lynch T J, Bell D W, Sordella R, Gurubhagavatula S, Okimoto R A, Brannigan BW, Harris PL, Haserlat SM, Supko JG, Haluska FG, Louis DN, Christiani DC, Settleman J, Haber DA. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non–small-cell lung cancer to gefitinib. New England Journal of Medicine. 2004. 350(21):2129-2139. DOI: 10.1056/NEJMoa040938
  13. 13. Pao W, Chmielecki J. Rational, biologically based treatment of EGFR-mutant non-small-cell lung cancer. Nature Reviews Cancer. 2010. 10(11):760–74. DOI: 10.1038/nrc2947.
  14. 14. Karachaliou N, Fernandez-Bruno M, Bracht JP, Rosell RJTCR. EGFR first-and second-generation TKIs—there is still place for them in EGFR-mutant NSCLC patients. Translational Cancer Research. 2018. 8:23-47. DOI: 10.21037/tcr.2018.10.06.
  15. 15. Bartholomew C, Eastlake L, Dunn P, Yiannakis D. EGFR targeted therapy in lung cancer; an evolving story. Respiratory medicine case reports. 2017. 20: 137-40. DOI: 10.1016/j.rmcr.2017.01.016.
  16. 16. Wu YL, Cheng Y, Zhou X, Lee KH, Nakagawa K, Niho S, Tsuji F, Linke R, Rosell R, Corral J, Migliorino MR, Pluzanski A, Sbar E, Wang T, White JL, Nadanaciva S, Sandin R, Mok TS. Dacomitinib versus geftinib as frst-line treatment for patients with EGFR-mutation-positive non-small-cell lung cancer (ARCHER 1050): a randomised, open-label, phase 3 trial. Lancet Oncology. 2017. 18(11):1454–66. DOI: 10.1016/S1470-2045(17)30608-3.
  17. 17. Mok TS, Cheng Y, Zhou X, Lee KH, Nakagawa K, Niho S, Lee M, Linke R, Rosell R, Corral J, Migliorino MR, Pluzanski A, Sbar E, Wang T, White JL, Wu YL. Improvement in overall survival in a randomized study that compared dacomitinib with geftinib in patients with advanced non-small-cell lung cancer and EGFR-activating mutations. Journal of Clinical Oncology. 2018. 36(22):2244–50. DOI: 10.1200/JCO.2018.78.7994.
  18. 18. Huang L, Jiang S, Shi Y. Tyrosine kinase inhibitors for solid tumors in the past 20 years (2001–2020). Journal of Hematology & Oncology. 2020. 13(1), 1-23. DOI: 10.1186/s13045-020-00977-0.
  19. 19. Lim SM, Syn NL, Cho BC, Ross AS. Acquired resistance to EGFR targeted therapy in non-small cell lung cancer: Mechanisms and therapeutic strategies. Cancer Treatment Reviews. 2018. 65:1–10. DOI: 10.1016/j.ctrv.2018.02.006.
  20. 20. Mok TS, Wu Y-L, Ahn M-J, Garassino MC, Kim HR, Ramalingam SS, Shepherd FA, He Y, Akamatsu H, Theelen WSME, Lee CK, Sebastian M, Templeton A, Mann H, Marotti M, Ghiorghiu S,Papadimitrakopoulou VA. Osimertinib or platinum-pemetrexed in EGFR T790M—positive lung cancer. New England Journal of Medicine. 2016. 376(7):629–40. DOI: 10.1056/NEJMoa1612674.
  21. 21. Reungwetwattana T, Nakagawa K, Cho BC, Cobo M, Cho EK, Bertolini A, Bohnet S, Zhou C, Lee KH, Nogami N, Okamoto I, Leighl N, Hodge R, McKeown A, Brown AP, Rukazenkov Y, Ramalingam SS, Vansteenkiste J. CNS response to osimertinib versus standard epidermal growth factor receptor tyrosine kinase inhibitors in patients with untreated EGFR-mutated advanced non-small-cell lung cancer. Journal of Clinical Oncology. 2018. 36(33):3290–7. DOI: 10.1200/JCO.2018.78.3118.
  22. 22. Lu S, Wang Q, Zhang G, Dong X, Yang C, Song Y, Chang G, Lu Y, Pan H, Chiu CH, Wang Z, Feng J, Zhou J, Xu X, Guo R, Chen J, Yang H, Chen Y, Yu Z, Shiah HS, Wang CC, Yang N, Fang J, Wang P, Wang K, Hu Y, He J, Wang Z, Shi J, Chen S, Wu Q, Sun C, Li C, Wei H, Cheng Y, Su WC, Hsia TC, Cui J, Sun Y, Yang JCH. A multicenter, open-label, single-arm, phase II study: the third generation EGFR tyrosine kinase inhibitor almonertinib for pretreated EGFR T790M-positive locally advanced or metastatic non-small cell lung cancer (APOLLO). Cancer Research. 2020 (80) (16 Supplement) CT190; DOI: 10.1158/1538-7445.AM2020-CT190.
  23. 23. Shi Y, Zhang S, Hu X, Feng J, Ma Z, Zhou J, Yang N, Wu L, Liao W, Zhong D, Han X, Wang Z, Zhang X, Qin S, Ying K, Feng J, Fang J, Liu L, Jiang Y. Safety, clinical activity, and pharmacokinetics of alfutinib (AST2818) in patients with advanced NSCLC with EGFR T790M mutation. Journal of Thoracic Oncology. 2020. 15(6):1015–26. DOI: 10.1016/j.jtho.2020.01.010.
  24. 24. Shi Y, Fang J, Shu Y, Wang D, Yu H, Zhao Y, Zhang L, Zhu B, Li X, Chen G, Shi J, Zheng R, Huang J, Yang S, Long J, Gao W, Greco M, Hu G, Li X. OA01.08 a phase I study to evaluate safety and antitumor activity of BPI-7711 in EGFRM+/T790M+ advanced or recurrent NSCLC patients. Journal of Thoracic Oncology. 2019. 14(11):1126–7. DOI: 10.1016/j.jtho.2019.09.022.
  25. 25. Ahn M, Han J, Lee KH, Kim SW, Kim DW, Lee YG, Cho EK, Kim JH, Lee GW, Lee JS, Min YJ, Kim JS, Lee SS, Kim HR, Hong MH, Ahn JS, Sun JM, Kim HT, Lee DH, Kim S, Cho BC. Lazertinib in patients with EGFR mutationpositive advanced non-small-cell lung cancer: results from the dose escalation and dose expansion parts of a frst-in-human, open-label, multicentre, phase 1–2 study. The Lancet Oncology. 2019. 20(12):1681–90. DOI: 10.1016/S1470-2045(19)30504-2.
  26. 26. Tan DSW, Leighl NB, Riely GJ, Yang JCH, Sequist LV, Wolf J, Seto T, Felip E, Aix SP, Jonnaert M, Pan C, Tan EY, Ko J, Moody SE, Kim DW. Safety and efcacy of nazartinib (EGF816) in adults with EGFR-mutant non-small-cell lung carcinoma: a multicentre, open-label, phase 1 study. The Lancet Respiratory Medicine. 2020. 8(6):561–72. DOI: 10.1016/S2213-2600(19)30267-X.
  27. 27. Merla A, Goel S. Novel drugs targeting the epidermal growth factor receptor and its downstream pathways in the treatment of colorectal cancer: a systematic review. Chemotherapy research and practice, 2012.2012:387172. DOI: 10.1155/2012/387172.
  28. 28. Cardoso F, Senkus E, Costa A, Papadopoulos E, Aapro M, Andre F, Harbeck N, Lopez BA, Barrios CH, Bergh J, Biganzoli L, Boers-Doets CB, Cardoso MJ, Carey LA, Cortés J, Curigliano G, Diéras V, El Saghir NS, Eniu A, Fallowfield L, Francis PA, Gelmon K, Johnston SRD, Kaufman B, Koppikar S, Krop IE, Mayer M, Nakigudde G, Offersen BV, Ohno S, Pagani O, Paluch-Shimon S, Penault-Llorca F, Prat A, Rugo HS, Sledge GW, Spence D, Thomssen C, Vorobiof DA, Xu B, Norton L, Winer EP. 4th ESOESMO International Consensus Guidelines for Advanced Breast Cancer (ABC 4). Annals of Oncology. 2018. 29(8):1634–57. DOI: 10.1093/annonc/mdy192.
  29. 29. Kannaiyan R, Mahadevan D. A comprehensive review of protein kinase inhibitors for cancer therapy. Expert review of anticancer therapy. 2018. 18(12), 1249-1270. DOI: 10.1080/14737140.2018.1527688.
  30. 30. Guo S, Colbert LS, Fuller M, Zhang Y, Gonzalez-Perez RR. Vascular endothelial growth factor receptor-2 in breast cancer. Biochimica et Biophysica Acta (BBA)-Reviews on Cancer. 2010; 1806: 108-121. DOI: https://doi.org/10.1016/j.bbcan.2010.04.004
  31. 31. Li W, Feng C, Di W, Hong S, Chen H, Ejaz M, Yang Y, Xu TR. Clinical use of vascular endothelial growth factor receptor inhibitors for the treatment of renal cell carcinoma. European Journal of Medicinal Chemistry. 2020; 200:112482. DOI: https://doi.org/10.1016/j.ejmech.2020.112482
  32. 32. Escudier B, Porta C, Schmidinger M, Rioux-Leclercq N, Bex A, Khoo V, Gruenvald V, Horwich A, ESMO Guidelines Committee. Renal cell carcinoma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Annals of Oncology. 2016; 27:v58-v68. DOI: https://doi.org/10.1093/annonc/mdw328
  33. 33. Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, Cosmo de Olivera A, Santoro A, Raoul JL, Forner A, Schwartz M, Porta C, Zeuzem S, Bolondi L, Greten TF, Galle PR, Seitz JF, Borbath I, Haussinger D, Giannaris T, Shan M, Moscovici M, Voliotis D, Bruix J. Sorafenib in advanced hepatocellular carcinoma. The New England Journal of Medicine. 2008;359:378–90. DOI: https://doi.org/10.1056/nejmoa0708857
  34. 34. Cheng A, Kang Y, Chen Z, Tsao CJ, Qin S, Kim JS, Luo R, Feng J, Ye S, Yang TS, Xu J, Sun Y, Liang H, Liu J, Wang J, Tak WY, Pan H, Burock K, Zou J, Voliotis D, Guan Z. Eficacy and safety of sorafenib in patients in the Asia-Pacifc region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial. Lancet Oncology. 2009;10:25–34. DOI: https://doi.org/10.1016/s1470-2045(08)70285-7
  35. 35. Bi F, Qin S, Gu S, Bai Y, Chen Z, Wang Z, Ying J, Lu Y, Meng Z, Pan H, Yang P, Zhang H, Chen X, Xu A, Liu X, Meng Q, Wu L, Chen F. Donafenib versus sorafenib as fİrst-line therapy in advanced hepatocellular carcinoma: An open-label, randomized, multicenter phase II/III trial. Journal of Clinical Oncology. 2020;38:4506-4506. DOI: 10.1200/JCO.2020.38.15_suppl.4506
  36. 36. Bruix J, Qin S, Merle P, Granito A, Huang YH, Bodoky G, Pracht M, Yokosuka O, Rosmorduc O, Breder V, Gerolami R, Masi G, Ross PJ, Song T, Bronowicki JP, Hourmand IO, Kudo M, Cheng AL, Llovet JM, Finn RS, LeBerre MA, Baumhauer A, Meinhardt G, Han G. Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2017;389:56–66. DOI: https://doi.org/10.1016/s0140-6736(16)32453-9
  37. 37. Li Q, Qin S, Gu S, Chen X, Lin L, Wang Z, Xu A, Chen X, Zhou C, Ren Z, Yang L, Xu L, Bai Y, Chen L, Li J, Pan H, Cao B, FanG W, Yan P, Jin C. Apatinib as second-line therapy in Chinese patients with advanced hepatocellular carcinoma: a randomized, placebo-controlled, double-blind, phase III study. J Clin Oncol. 2020;38:4507-4507. DOI: 10.1200/JCO.2020.38.15_suppl.4507
  38. 38. Abou-Alfa G,K Meyer T, Cheng AL, El-Khoueiry AB, Rimassa L, Ryoo BY, Cicin I, Merle P, Chen Y, Park JW, Blanc JF, Bolondi L, Klümpen HJ, Chan SL, Zagonel V, Pressiani T, Ryu MH, Venook AP, Hessel C, Borgman-Hagey AE, Schwab G, Kelley RK. Cabozantinib in patients with advanced and progressing hepatocellular carcinoma. The New England Journal of Medicine. 2018;379:54–63. DOI: https://doi.org/10.1056/nejmoa1717002
  39. 39. Zhu AX, Finn RS, Ikeda M, Sung MW, Baron AD, Kudo M, Okusaka T, Kobayashi M, Kumada H, Kaneko S, Pracht M, Mamontov K, Meyer T, Mody K, Kubota T, Dutcus CE, Saito K, Siegel AB, Dubrovsky L, Llovet JM. A phase Ib study of lenvatinib (LEN) plus pembrolizumab (PEMBRO) in unresectable hepatocellular carcinoma (uHCC). Journal of Clinical Oncology. 2020;38: 4519-4519. DOI: 10.1200/JCO.2020.38.15_suppl.4519
  40. 40. Han B, Li K, Wang Q, Zhang L, Shi J, Wang Z, Cheng Y, He J, Shi Y, Zhao Y, Yu H, Zhao Y, Chen W, Luo Y, Wu L, Wang X, Pirker R, Nan K, Jin F, Dong J, Li B, Sun Y. Efect of anlotinib as a third-line or further treatment on overall survival of patients with advanced non-small cell lung cancer: the ALTER 0303 phase 3 randomized clinical trial. JAMA OncolOGY. 2018;4:1569–75. DOI: https://doi.org/10.1001/jamaoncol.2018.3039
  41. 41. Cheng Y, Wang Q, Li K, Shi J, Wu L, Han B, Chen G, He J, Wang J, Qin H, Li X. OA13.03 anlotinib as third-line or furtherline treatment in relapsed SCLC: a Multicentre, Randomized, Double-Blind Phase 2 Trial. Journal of Thoracic Oncology. 2018. 13(10): 351-352. DOI: 10.1016/j.jtho.2018.08.308
  42. 42. Poddubskaya E, Baranova M, Allina D, Smirnov PY, Albert EA, Kirilchev AP, Aleshin AA, Sekacheva MI. Suntsova MV. Personalized prescription of tyrosine kinase inhibitors in unresectable metastatic cholangiocarcinoma. Experimental Hematology & Oncology. 2018. 7:21. DOI: 10.1186/s40164-018-0113-x.
  43. 43. Sobhani N, Chumei F, Flores-Villanueva OP, Generali D, Li, Y. The fibroblast growth factor receptors in breast cancer: from oncogenesis to better treatments. International journal of molecular sciences. 2020. 21(6): 2011. DOI: 10.3390/ijms21062011.
  44. 44. Vogel A, Sahai V, Hollebecque A, Vaccaro G, Melisi D, Al-Rajabi R, Paulson AS, Borad MJ, Gallinson D, Murphy AG, Oh D-Y, Dotan E, Catenacci DV, Van Cutsem E, Lihou CF, Zhen H, Féliz L, Abou-Alfa K. FIGHT-202: a phase II study of pemigatinib in patients (pts) with previously treated locally advanced or metastatic cholangiocarcinoma (CCA). Annals of Oncology. 2019. 30:v876. DOI: 10.1093/annonc/mdz394.031
  45. 45. Javle M, Kelley RK, Roychowdhury S, Weiss KH, et al. AB051. P-19. A phase II study of infgratinib (BGJ398) in previously-treated advanced cholangiocarcinoma containing FGFR2 fusions. Hepatobiliary Surgery and Nutrition. 2019.8(Suppl 1):AB051. DOI: 10.21037/hbsn.2019.AB051
  46. 46. Knowles MA, Hurst CD. Molecular biology of bladder cancer: new insights into pathogenesis and clinical diversity. Nature Reviews Cancer. 2015. 15(1):25–41. DOI: 10.1038/nrc3817
  47. 47. Pal SK, Rosenberg JE, Hofman-Censits JH, Berger R, Quinn DI, et al. Efficacy of BGJ398, a Fibroblast Growth Factor Receptor 1-3 Inhibitor, in Patients with Previously Treated Advanced Urothelial Carcinoma with FGFR3 Alterations. Cancer Discovery. 2018. 8(7):812. DOI: 10.1158/2159-8290.CD-18-0229
  48. 48. Bellmunt J, de Wit R, Vaughn DJ, Fradet Y, Lee J-L, Fong L, Vogelzang NJ, Climent MA, Petrylak DP, Choueiri TK, Necchi A, Gerritsen W, Gurney H, Quinn DI, Culine S, Sternberg CN, Mai Y, Poehlein CH, Perini RF, Bajorin DF. Pembrolizumab as secondline therapy for advanced urothelial carcinoma. The New England Journal of Medicine. 2017. 376(11):1015–26. DOI: 10.1056/NEJMoa1613683
  49. 49. Haque S, Morris, JC. Transforming growth factor-β: A therapeutic target for cancer. Human vaccines & immunotherapeutics. 2017. 13(8):1741-1750. DOI: 10.1080/21645515.2017.1327107
  50. 50. Ma X, Cui Z, Du Z, Lin H. Transforming growth factor-β signaling, a potential mechanism associated with diabetes mellitus and pancreatic cancer?. Journal of cellular physiology. 2020. 235(9):5882-5892. DOI: 10.1002/jcp.29605
  51. 51. Hahn SA, Schutte M, Hoque AT, Moskaluk CA, da Costa LT, Rozenblum E, Weinstein CL, Fischer A, Yeo CJ, Hruban RH, Kern SE. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science. 1996. 271(5247):350-3. DOI: 10.1126/science.271.5247.350
  52. 52. Riggins GJ, Kinzler KW, Vogelstein B, Thiagalingam S. Frequency of Smad gene mutations in human cancers. Cancer Research. 1997. 57(13):2578-80.
  53. 53. Halder SK, Beauchamp RD, Datta PK. Smad7 induces tumorigenicity by blocking TGF-beta-induced growth inhibition and apoptosis. Experimental Cell Research. 2005. 307(1):231-46. DOI: 10.1016/j.yexcr.2005.03.009
  54. 54. Bruna A, Darken RS, Rojo F, Ocana A, Penuelas S, Arias A, Paris R, Tortosa A, Mora J, Baselga J, Seoane J. High TGFbeta-Smad activity confers poor prognosis in glioma patients and promotes cell proliferation depending on the methylation of the PDGF-B gene. Cancer Cell. 2007. 11(2):147-60. DOI: 10.1016/j.ccr.2006.11.023
  55. 55. Chod J, Zavadova E, Halaska MJ, Strnad P, Fucikova T, Rob L. Preoperative transforming growth factor-beta 1 (TGF-beta 1) plasma levels in operable breast cancer patients. European Journal of Gynaecological Oncology. 2008. 29(6):613-6. PMID: 19115689
  56. 56. Fischer JR, Darjes H, Lahm H, Schindel M, Drings P, Krammer PH. Constitutive secretion of bioactive transforming growth factor beta 1 by small cell lung cancer cell lines. European Journal of Cancer. 1994. 30A (14):2125-9. DOI: 10.1016/0959-8049(94)00364-B
  57. 57. Labidi SI, Menetrier-Caux C, Chabaud S, Chassagne C, Sebban C, Gargi T, Biron P, Blay JY, Ghesquieres H. Serum cytokines in follicular lymphoma. Correlation of TGF-beta and VEGF with survival. Annals of hematology. 2010. 89(1):25-33. DOI: 10.1007/s00277-009-0777-8
  58. 58. Langenskiold M, Holmdahl L, Falk P, Angenete E, Ivarsson ML. Increased TGF-beta 1 protein expression in patients with advanced colorectal cancer. Journal of Surgical Oncology. 2008. 97(5):409-15. DOI: 10.1002/jso.20961
  59. 59. Shariat SF, Walz J, Roehrborn CG, Zlotta AR, Perrotte P, Suardi N, Saad F, Karakiewicz PI. External validation of a biomarker-based preoperative nomogram predicts biochemical recurrence after radical prostatectomy. Journal of Clınıcal Oncology. 2008. 26 (9):1526-31. DOI: 10.1200/JCO.2007.12.4669
  60. 60. Shirai Y, Kawata S, Tamura S, Ito N, Tsushima H, Takaishi K, Kiso S, Matsuzawa Y. Plasma transforming growth factor-beta 1 in patients with hepatocellular carcinoma. Comparison with chronic liver diseases. Cancer. 1994. 73(9):2275-9. DOI: 10.1002/1097-0142(19940501)73:9<2275::AID-CNCR2820730907>3.0.CO;2-T
  61. 61. von Rahden BH, Stein HJ, Feith M, Puhringer F, Theisen J, Siewert€ JR, Sarbia M. Overexpression of TGF-beta1 in esophageal (Barrett’s) adenocarcinoma is associated with advanced stage of disease and poor prognosis. Molecular Carcinogenesis. 2006. 45(10):786-94. DOI: 10.1002/mc.20259
  62. 62. Calon A, Lonardo E, Berenguer-Llergo A., Espinet E., Hernando-Momblona X., Iglesias M, Sevillano M, Palomo-Ponce S, Tauriello DVF, Byrom D, Cortina C, Morral C, Barceló C, Tosi S, Riera A, Attolini CSO, Rossell D, Sancho E, Batlle, E. Stromal gene expression defines poor-prognosis subtypes in colorectal cancer. Nature Genetics. 2015. 47(4): 320– U362. DOI: 10.1038/ng.3225
  63. 63. Alsina-Sanchis E, Figueras A, Lahiguera A, Vidal A, Casanovas O, Graupera M, Villanueva A, Vinals, F. The TGF beta pathway stimulates ovarian cancer cell proliferation by increasing IGF1R levels. International Journal of Cancer. 2016. 139(8): 1894– 1903. DOI: 10.1002/ijc.30233
  64. 64. Serova M, Tijeras-Raballand A, Dos Santos C, Muller N, Benhadji K, Paradis V, Faivre S, Raymond E, de Gramont A. Abstract 2094: Effects of TGF-beta signaling inhibition with LY2157299 in hepatocarcinoma models and in ex vivo whole tumor tissue samples from patient specimen. Cancer Research. 2013. 73(8 Suppl):nr 2094. DOI: 10.1158/1538-7445.AM2013-2094
  65. 65. Ganapathy V, Ge R, Grazioli A, Xie W, Banach-Petrosky W, Kang Y, Lonning S, McPherson J, Yingling JM, Biswas S, Mundy GR, Reiss M. Targeting the transforming growth factor-β pathway inhibits human basal-like breast cancer metastasis. Molecular cancer. 2010. 9(1): 1-16. DOI: 10.1186/1476-4598-9-122
  66. 66. Zhang B., Halder SK, Kashikar ND, Cho Y-J., Datta A, Gorden, DL, Datta PK. Antimetastatic role of Smad4 signaling in colorectal cancer. Gastroenterology. 2010. 138(3): 969– U220. DOI: 10.1053/j.gastro.2009.11.004
  67. 67. Ehata S, Hanyu A, Fujime M, Katsuno Y, et al. Ki26894, a novel transforming growth factor-beta type I receptor kinase inhibitor, inhibits in vitro invasion and in vivo bone metastasis of a human breast cancer cell line. Cancer Science. 2007. 98(1): 127– 133. 10.1111/j.1349-7006.2006.00357.x
  68. 68. Shinto O, Yashiro M, Kawajiri H, Shimizu K, Shimizu T, Miwa A, Hirakawa, K. Inhibitory effect of a TGF beta receptor type-I inhibitor, Ki26894, on invasiveness of scirrhous gastric cancer cells. British Journal of Cancer. 2010. 102(5): 844– 851. DOI: 10.1038/sj.bjc.6605561
  69. 69. Roddam AW, Allen NE, Appleby P, Key TJ, et al. Insulin like growth factors, their binding proteins, and prostate cancer risk: analysis of individual patient data from 12 prospective studies. Ann Intern Med. 2008. 149:461–71. DOI: 10.7326/0003-4819-149-7-200810070-00006
  70. 70. Price AJ, Allen NE, Appleby PN, Crowe FL, et al. Insulin like growth factor-I concentration and risk of prostate cancer: results from the European Prospective Investigation into Cancer and Nutrition. Cancer Epidemiology, Biomarkers & Prevention. 2012. 21:1531–41. DOI: 10.1158/1055-9965.EPI-12-0481-T
  71. 71. Ho GY, Zheng SL, Cushman M, Perez-Soler R, Kim M, Xue X, Wang T, Schlecht NF, Tinker L, Rohan TE, Wassertheil-Smoller S, Wallace R, Chen C, Xu J, Yu H. Associations of insulin and IGFBP-3 with lung cancer susceptibility in current smokers. Journal of the National Cancer Institute. 2016. 108(7). DOI: 10.1093/jnci/djw012
  72. 72. Yoon YS, Keum N, Zhang X, Cho E, Giovannucci EL. Circulating levels of IGF-1, IGFBP-3, and IGF-1/IGFBP-3 molar ratio and colorectal adenomas: a meta-analysis. Cancer epidemiology. 2015. 39(6):1026-1035. DOI: 10.1016/j.canep.2015.09.004
  73. 73. Chng WJ, Gualberto A, Fonseca R. IGF-1R is overexpressed in poor-prognostic subtypes of multiple myeloma. Leukemia. 2006. 20(1):174-176. DOI: 10.1038/sj.leu.2403997
  74. 74. Sprynski AC., Hose D, Kassambara A, Vincent L, Jourdan M, Rossi JF, Goldschmidt H., Klein B. Insulin is a potent myeloma cell growth factor through insulin/IGF-1 hybrid receptor activation. Leukemia. 2010. 24(11): 1940-1950. DOI: 10.1038/leu.2010.192
  75. 75. Vishwamitra D, George SK, Shi P, Kaseb AO, Amin HM. Type I insulin-like growth factor receptor signaling in hematological malignancies. Oncotarget. 2017. 8(1): 1814. DOI: 10.18632/oncotarget.12123
  76. 76. Lin LH, Lin JS, Yang CC, Cheng HW, Chang KW, Liu CJ. Overexpression of platelet-derived growth factor and its receptor are correlated with oral tumorigenesis and poor prognosis in oral squamous cell carcinoma. International journal of molecular sciences. 2020. 21(7):2360. DOI: 10.3390/ijms21072360
  77. 77. Kim S, You D, Jeong Y, Yoon SY, Kim SA, Lee J. E. Inhibition of platelet-derived growth factor receptor synergistically increases the pharmacological effect of tamoxifen in estrogen receptor α positive breast cancer. Oncology Letters. 2006. 21(4):1-8. DOI: 10.3892/ol.2021.12555
  78. 78. Olson MF, Marais R. Ras protein signalling. Seminars in immunology. 2000;12:63-73. DOI: 10.1006/smim.2000.0208
  79. 79. Boguski MS, McCormick F. Proteins regulating Ras and its relatives. Nature. 1993;366:643-654. DOI: 10.1038/366643a0
  80. 80. Levitzki A. Protein kinase inhibitors as a therapeutic modality. Accounts of chemical research. 2003;36:462-469. DOI: 10.1021/ar0201207
  81. 81. Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949-954. DOI: 10.1038/nature00766
  82. 82. Pollock PM, Meltzer PS. Lucky draw in the gene raffle. Nature. 2002;417:906-907. DOI: 10.1038/417906a
  83. 83. Pruitt K, Der CJ. Ras and Rho regulation of the cell cycle and oncogenesis. Cancer letters. 2001;171:1-10. DOI: 10.1016/s0304-3835(01)00528-6
  84. 84. Bos JL. ras oncogenes in human cancer: a review. Cancer research. 1989;49:4682-4689.
  85. 85. Roskoski R, Jr. Properties of FDA-approved small molecule protein kinase inhibitors: A 2021 update. Pharmacological research. 2021;165:105463. DOI: 10.1016/j.phrs.2021.105463
  86. 86. Bagchi S, Rathee P, Jayaprakash V, Banerjee S. Farnesyl Transferase Inhibitors as Potential Anticancer Agents. Mini reviews in medicinal chemistry. 2018;18:1611-1623. DOI: 10.2174/1389557518666180801110342
  87. 87. Wang Q, Chen F, Liu P, Mu Y, Sun S, Yuan X, et al. Scaffold-based analysis of nonpeptide oncogenic FTase inhibitors using multiple similarity matching, binding affinity scoring and enzyme inhibition assay. Journal of molecular graphics & modelling. 2021;105:107898. DOI: 10.1016/j.jmgm.2021.107898
  88. 88. Wang J, Yao X, Huang J. New tricks for human farnesyltransferase inhibitor: cancer and beyond. MedChemComm. 2017;8:841-854. DOI: 10.1039/c7md00030h
  89. 89. Klochkov SG, Neganova ME, Yarla NS, Parvathaneni M, Sharma B, Tarasov VV, et al. Implications of farnesyltransferase and its inhibitors as a promising strategy for cancer therapy. Seminars in cancer biology. 2019;56:128-134. DOI: 10.1016/j.semcancer.2017.10.010
  90. 90. End DW, Smets G, Todd AV, Applegate TL, Fuery CJ, Angibaud P, et al. Characterization of the antitumor effects of the selective farnesyl protein transferase inhibitor R115777 in vivo and in vitro. Cancer research. 2001;61:131-137.
  91. 91. Korzeniecki C, Priefer R. Targeting KRAS mutant cancers by preventing signaling transduction in the MAPK pathway. European journal of medicinal chemistry. 2021;211:113006. DOI: 10.1016/j.ejmech.2020.113006
  92. 92. Lee HW, Sa JK, Gualberto A, Scholz C, Sung HH, Jeong BC, et al. A Phase II Trial of Tipifarnib for Patients with Previously Treated, Metastatic Urothelial Carcinoma Harboring HRAS Mutations. Clinical cancer research : an official journal of the American Association for Cancer Research. 2020;26:5113-5119. DOI: 10.1158/1078-0432.CCR-20-1246
  93. 93. Rao S, Cunningham D, de Gramont A, Scheithauer W, Smakal M, Humblet Y, et al. Phase III double-blind placebo-controlled study of farnesyl transferase inhibitor R115777 in patients with refractory advanced colorectal cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2004;22:3950-3957. DOI: 10.1200/JCO.2004.10.037
  94. 94. Whitehead RP, McCoy S, Macdonald JS, Rivkin SE, Neubauer MA, Dakhil SR, et al. Phase II trial of R115777 (NSC #70818) in patients with advanced colorectal cancer: a Southwest Oncology Group study. Investigational new drugs. 2006;24:335-341. DOI: 10.1007/s10637-005-4345-3
  95. 95. Cohen SJ, Ho L, Ranganathan S, Abbruzzese JL, Alpaugh RK, Beard M, et al. Phase II and pharmacodynamic study of the farnesyltransferase inhibitor R115777 as initial therapy in patients with metastatic pancreatic adenocarcinoma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2003;21:1301-1306. DOI: 10.1200/JCO.2003.08.040
  96. 96. Adjei AA, Mauer A, Bruzek L, Marks RS, Hillman S, Geyer S, et al. Phase II study of the farnesyl transferase inhibitor R115777 in patients with advanced non-small-cell lung cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2003;21:1760-1766. DOI: 10.1200/JCO.2003.09.075
  97. 97. Baranyi M, Buday L, Hegedus B. K-Ras prenylation as a potential anticancer target. Cancer metastasis reviews. 2020;39:1127-1141. DOI: 10.1007/s10555-020-09902-w
  98. 98. Liu M, Bryant MS, Chen J, Lee S, Yaremko B, Lipari P, et al. Antitumor activity of SCH 66336, an orally bioavailable tricyclic inhibitor of farnesyl protein transferase, in human tumor xenograft models and wap-ras transgenic mice. Cancer research. 1998;58:4947-4956.
  99. 99. Feldkamp MM, Lau N, Roncari L, Guha A. Isotype-specific Ras·GTP-Levels Predict the Efficacy of Farnesyl Transferase Inhibitors against Human Astrocytomas Regardless of <em>Ras</em> Mutational Status. Cancer research. 2001;61:4425-4431.
  100. 100. Morgillo F, Lee HY. Lonafarnib in cancer therapy. Expert opinion on investigational drugs. 2006;15:709-719. DOI: 10.1517/13543784.15.6.709
  101. 101. Britten CD, Rowinsky EK, Soignet S, Patnaik A, Yao SL, Deutsch P, et al. A phase I and pharmacological study of the farnesyl protein transferase inhibitor L-778,123 in patients with solid malignancies. Clinical cancer research : an official journal of the American Association for Cancer Research. 2001;7:3894-3903.
  102. 102. Martin NE, Brunner TB, Kiel KD, DeLaney TF, Regine WF, Mohiuddin M, et al. A phase I trial of the dual farnesyltransferase and geranylgeranyltransferase inhibitor L-778,123 and radiotherapy for locally advanced pancreatic cancer. Clinical cancer research : an official journal of the American Association for Cancer Research. 2004;10:5447-5454. DOI: 10.1158/1078-0432.CCR-04-0248
  103. 103. Caponigro F, Casale M, Bryce J. Farnesyl transferase inhibitors in clinical development. Expert opinion on investigational drugs. 2003;12:943-954. DOI: 10.1517/13543784.12.6.943
  104. 104. Leicht DT, Balan V, Kaplun A, Singh-Gupta V, Kaplun L, Dobson M, et al. Raf kinases: function, regulation and role in human cancer. Biochimica et biophysica acta. 2007;1773:1196-1212. DOI: 10.1016/j.bbamcr.2007.05.001
  105. 105. Dancey J, Sausville EA. Issues and progress with protein kinase inhibitors for cancer treatment. Nature reviews Drug discovery. 2003;2:296-313. DOI: 10.1038/nrd1066
  106. 106. Avruch J, Khokhlatchev A, Kyriakis JM, Luo Z, Tzivion G, Vavvas D, et al. Ras activation of the Raf kinase: tyrosine kinase recruitment of the MAP kinase cascade. Recent progress in hormone research. 2001;56:127-155. DOI: 10.1210/rp.56.1.127
  107. 107. Lyons JF, Wilhelm S, Hibner B, Bollag G. Discovery of a novel Raf kinase inhibitor. Endocrine-related cancer. 2001;8:219-225. DOI: 10.1677/erc.0.0080219
  108. 108. Neuzillet C, Tijeras-Raballand A, de Mestier L, Cros J, Faivre S, Raymond E. MEK in cancer and cancer therapy. Pharmacology & therapeutics. 2014;141:160-171. DOI: 10.1016/j.pharmthera.2013.10.001
  109. 109. Roberts PJ, Der CJ. Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene. 2007;26:3291-3310. DOI: 10.1038/sj.onc.1210422
  110. 110. Lorusso PM, Adjei AA, Varterasian M, Gadgeel S, Reid J, Mitchell DY, et al. Phase I and pharmacodynamic study of the oral MEK inhibitor CI-1040 in patients with advanced malignancies. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2005;23:5281-5293. DOI: 10.1200/JCO.2005.14.415
  111. 111. Wang D, Boerner SA, Winkler JD, LoRusso PM. Clinical experience of MEK inhibitors in cancer therapy. Biochimica et biophysica acta. 2007;1773:1248-1255. DOI: 10.1016/j.bbamcr.2006.11.009
  112. 112. Fruman, D. A, Rommel C. PI3K and cancer: lessons, challenges and opportunities. Nature reviews Drug discovery. 2014; 13: 140-156. DOI: 10.1038/nrd4204
  113. 113. Kannaiyan R, & Mahadevan, D. A comprehensive review of protein kinase inhibitors for cancer therapy. Expert review of anticancer therapy. 2018; 18: 1249-1270. DOI: 10.1080/14737140.2018.1527688
  114. 114. Markham, A. Idelalisib: first global approval. Drugs. 2014; 74: 1701-1707. DOI: 10.1007/s40265-014-0285-6
  115. 115. O'Brien S M, Lamanna N, Kipps T J, Flinn I, Zelenetz A D, Burger J A, Coutre S E. A phase 2 study of idelalisib plus rituximab in treatment-naive older patients with chronic lymphocytic leukemia. Blood, The Journal of the American Society of Hematology. 2015; 126: 2686-2694. DOI: 10.1182/blood-2015-03-630947
  116. 116. Dreyling M, Morschhauser F, Bouabdallah K, Bron D, Cunningham D, Assouline S E, Zinzani P L. Phase II study of copanlisib, a PI3K inhibitor, in relapsed or refractory, indolent or aggressive lymphoma. Annals of Oncology. 2017; 28: 2169-2178. DOI: 10.1093/annonc/mdx289
  117. 117. Fasolo, A., Sessa, C. Current and future directions in mammalian target of rapamycin inhibitors development. Expert opinion on investigational drugs. 2011; 20: 381-394. DOI: 10.1517/13543784.2011.541154
  118. 118. Lu, X., Paliogiannis, P., Calvisi, D. F., Chen, X. Role of the mammalian target of rapamycin pathway in liver cancer: from molecular genetics to targeted therapies. Hepatology, 2021; 73: 49-61. DOI: 10.1002/hep.31310
  119. 119. Faes, S., Demartines, N., Dormond, O. Mechanistic Target of Rapamycin Inhibitors in Renal Cell Carcinoma: Potential, Limitations, and Perspectives. Frontiers in Cell and Developmental Biology. 2021; 9, 459. DOI: 10.3389/fcell.2021.636037
  120. 120. Günther A, Baumann P, Burger R, Kellner C, Klapper W, Schmidmaier R, Gramatzki, M. Activity of everolimus (RAD001) in relapsed and/or refractory multiple myeloma: a phase I study. Haematologica. 2015; 100(4), 541. DOI: 10.3324/haematol.2014.116269
  121. 121. Dudkin L, Dilling M B, Cheshire P J, Harwood F C, Hollingshead M, Arbuck S G, Houghton, P J. Biochemical correlates of mTOR inhibition by the rapamycin ester CCI-779 and tumor growth inhibition. Clinical Cancer Research. 2001; 7: 1758-1764.
  122. 122. Liu C, Zhang H, Li Y, Zhang Z, Shi R, Xu S, Chen J. Apatinib Combined with CCI-779 Inhibits the Proliferation and Migration of Small Cell Lung Cancer NCI-H446 Cells In Vitro. Zhongguo fei ai za zhi= Chinese Journal of Lung Cancer. 2020; 23: 216-222. DOI: 10.3779/j.issn.1009-3419.2020.104.08
  123. 123. Xiang H Y, Wang X, Chen Y H, Zhang X, Tan C, Wang Y, Yang C H. Identification of methyl (5-(6-((4-(methylsulfonyl) piperazin-1-yl) methyl)-4-morpholinopyrrolo [2, 1-f][1, 2, 4] triazin-2-yl)-4-(trifluoromethyl) pyridin-2-yl) carbamate (CYH33) as an orally bioavailable, highly potent, PI3K alpha inhibitor for the treatment of advanced solid tumors. European Journal of Medicinal Chemistry. 2018; 209: 112913. DOI: 10.1016/j.ejmech.2020.112913
  124. 124. Asghar U, Witkiewicz A K, Turner N C, Knudsen E S. The history and future of targeting cyclin-dependent kinases in cancer therapy. Nature reviews Drug discovery. 2015; 14: 130-146. DOI: 10.1038/nrd4504
  125. 125. Samson, K. LEE011 CDK Inhibitor Showing Early Promise in Drug-Resistant Cancers. Oncol. Times. 2014; 36, 39–40. DOI: 10.1097/01.COT.0000444043.33304.c1
  126. 126. Gelbert L M, Cai S, Lin X, Sanchez-Martinez C, Del Prado M, Lallena M J, de Dios A. Preclinical characterization of the CDK4/6 inhibitor LY2835219: in-vivo cell cycle-dependent/independent anti-tumor activities alone/in combination with gemcitabine. Investigational new drugs. 2014; 32: 825-837. DOI: 10.1007/s10637-014-0120-7
  127. 127. Łukasik P, Baranowska-Bosiacka I, Kulczycka K, Gutowska I. Inhibitors of Cyclin-Dependent Kinases: Types and Their Mechanism of Action. International Journal of Molecular Sciences. 2021; 22: 2806. DOI: 10.3390/ijms22062806
  128. 128. Diéras V, Harbeck N, Joy A A, Gelmon K, Ettl J, Verma S, Finn R S. Palbociclib with letrozole in postmenopausal women with ER+/HER2− advanced breast cancer: hematologic safety analysis of the randomized PALOMA-2 trial. The oncologist. 2019; 24: 1514. DOI: 10.1634/theoncologist.2019-0019
  129. 129. Nuwayhid S J, Hyde J, Aleshin A, Walker D H, Arkin M R. SNS-032 is a potent and selective inhibitor of CDK2, 7 and 9 and induces cell death by inhibiting cell cycle progression and the expression of antiapoptotic proteins. 2006
  130. 130. Tong W G, Chen R, Plunkett W, Siegel D, Sinha R, Harvey R D, Wierda W G. Phase I and pharmacologic study of SNS-032, a potent and selective Cdk2, 7, and 9 inhibitor, in patients with advanced chronic lymphocytic leukemia and multiple myeloma. Journal of clinical oncology. 2010; 28: 3015. DOI: 10.1200/JCO.2009.26.1347
  131. 131. McClue S J, Blake D, Clarke R, Cowan A, Cummings L, Fischer P M, Lane D P. In vitro and in vivo antitumor properties of the cyclin dependent kinase inhibitor CYC202 (R-roscovitine). International journal of cancer. 2002; 102: 463-468. DOI: 10.1002/ijc.10738
  132. 132. Alsfouk A A, Alshibl H M, Altwaijry N A, Alsfouk B A, Al-Abdullah E S. Synthesis and biological evaluation of seliciclib derivatives as potent and selective CDK9 inhibitors for prostate cancer therapy. Monatshefte für Chemie-Chemical Monthly. 2021; 152:109-120. DOI: 10.1007/s00706-020-02727-x
  133. 133. Smyth J F, Aamdal S, Awada A, Dittrich C, Caponigro F, Schöffski P, Punt C J A. Phase II study of E7070 in patients with metastatic melanoma. Annals of oncology. 2005; 16: 158-161. DOI: 10.1093/annonc/mdi016
  134. 134. Sedlacek H, Czech J, Naik R, Kaur G, Worland P, Losiewicz M, Sausville E. Flavopiridol (L86 8275; NSC 649890), a new kinase inhibitor for tumor therapy. International journal of oncology. 1996; 9: 1143-1168. DOI: 10.3892/ijo.9.6.1143
  135. 135. Sedlacek H H. Mechanisms of action of flavopiridol. Critical reviews in oncology/hematology. 2001; 38: 139-170. DOI: 10.1016/S1040-8428(00)00124-4
  136. 136. Teicher BA. Protein Kinase C as a Therapeutic Target. Clinical cancer research. 2006;11:5336-45. DOI: 10.1158/1078-0432.ccr-06-0945
  137. 137. Swannie HC, Kaye SB. Protein Kinase C Inhibitors. Current oncology reports. 2002;4:37-46. DOI: 10.1007/s11912-002-0046-7
  138. 138. Cooke M, Magimaidas A, Casado-Medrano V, Kazanietz MG. Protein kinase C in cancer: The top Five Unanswered Questions. Molecular carcinogenesis. 2017;56:1531-1542. DOI: 10.1002/mc.22617
  139. 139. Marshall JL, Eisenberg SG, Johnson MD, Hanfelt J, Dorr FA, El-Ashry D, Oberst M, Fuxman Y, Holmlund J, Malik S. A phase II trial of ISIS 3521 in patients with metastatic colorectal cancer. Clinical colorectal cancer. 2004;4: 268-274. DOI: 10.3816/ccc.2004.n.026
  140. 140. Kunt T, Forst T, Kazda C, Harzer O, Engelbach M, Löbig M, Beyer J, Pfützner A. The beta-specific protein kinase C inhibitor ruboxistaurin (LY333531) suppresses glucose-induced adhesion of human monocytes to endothelial cells in vitro. Journal of diabetes science and technology. 2007;6: 929-935. DOI: 10.1177/193229680700100620
  141. 141. Yonggui W, Guozhong W, Xiangming Q, Hui L, Hao Q, Jijia S, Shantan L. Protein Kinase C β Inhibitor LY333531 Attenuates Intercellular Adhesion Molecule-1 and Monocyte Chemotactic Protein-1 Expression in the Kidney in Diabetic Rats. Journal of Pharmacological Sciences. 2006;4: 335-343. DOI: 10.1254/jphs.FP0050896
  142. 142. Kumari, A., Silakari, O., & Singh, R. (2018). Recent advances in colony stimulating factor-1 receptor/c-FMS as an emerging target for various therapeutic implications. Biomedicine & Pharmacotherapy, 103, 662-679. doi: 10.1016/j.biopha.2018.04.046

Written By

Gizem Kursunluoglu, Duygu Erdogan, Elcin Cagatay, Esra Bulut Atalay, Seminay Guler, Yonca Gungor and Hulya Ayar Kayali

Reviewed: 25 June 2021 Published: 11 October 2021