Open access

Introductory Chapter: Protein Kinases as Promising Targets for Drug Design against Cancer

Written By

Rohit Bhatia and Rajesh K. Singh

Published: 08 December 2021

DOI: 10.5772/intechopen.100315

From the Edited Volume

Protein Kinases - Promising Targets for Anticancer Drug Research

Edited by Rajesh Kumar Singh

Chapter metrics overview

318 Chapter Downloads

View Full Metrics

1. Introduction

Cancer is one of the most dreadful and highly prevailing life-threatening ailments of the modern age. Despite a great advancement in the health sector, still it is the leading cause of mortality around the globe [1, 2]. The continuous research is in progress for several years to design therapeutic agents against cancer with greater efficacy, specificity, and least toxicity. For the past two decades, the protein kinase family has been greatly focused by the researchers for drug development against cancer. There are about 538 protein kinase enzymes that are encoded by the human genome, which function mainly by transferring a γ-phosphate group from the ATP site toward amino acid residues such as serine, threonine, or tyrosine residues [3, 4, 5]. It is evident that several members of this protein kinase family have tendencies to initiate and develop human cancers [6, 7]. The recently developed small molecules as potential kinase inhibitors in the therapy of a variety of cancers have witnessed the significance of kinases as a target against cancers. Moreover, these are in second place as a target for drugs after the G-protein-coupled receptors [8]. Protein kinases are associated with the promotion of cell proliferation, migration, and survival and, when they are dysregulated/overexpressed, lead to oncogenesis [9, 10]. During the past decades, it has been observed that human malignancies are largely associated with modulation or dysfunction of protein and lipid kinases due to the deactivation of phosphatases resulting from chromosomal abnormalities or mutations [11, 12]. It is worth notable that the anti-inflammatory kinases such as EGFR, VEGFR, BCR-ABL, ALK, KIT, HER2, and several others are involved in the development of solid cancers including chronic lymphoid leukemia, lymphoblastic leukemia, mantle cell lymphoma, myelogenous lymphoma, and several other types of cancers [13]. These kinases show a pro-tumor effect associated with loss of normal kinase functioning followed by mutations and associations with high-regulatory T cell pathogens [14]. These pathogens ultimately activate the anti-inflammatory kinases and initiate the development of solid cancers. The role of some kinases in the development of cancers has been depicted in Figure 1.

Figure 1.

Impact of protein kinases in development of cancer.

Kinase amplifications are able to play diagnostic, prognostic, therapeutic as well as biomarker roles in cancer [15]. The amplifications of EGFR have been well seen in a variety of cancers including non-small cell lung cancer, colorectal cancer, bladder cancer, pancreatic, and breast cancer, whereas ERBB2 amplifications are associated with esophageal, gastric, breast, and ovarian cancers [16, 17, 18]. Overexpression of EGFR, ERBB2, EPHA2, and AKT2 are the best examples of biomarkers for cancers [19, 20].

Advertisement

2. Progress in the development of protein kinase inhibitors against cancer

In the past two decades, there has been a remarkable progress made in the drug development process involving protein kinases as a target. The first FDA approved drug imatinib was launched in 2001 against chronic myeloid leukemia which inhibits Abelson (ABL) tyrosine kinase [21]. It was proved to be a blockbuster drug with polypharmacological effects. From 2001 to 2021, in a span of 20 years, there has been an extraordinary progress made with the discovery of more potent and specific small-molecule kinase inhibitors and about 70 new drugs have got approval in this time span [22]. These drugs have left a promising positive impact to improve the drug design strategies and therapy to treat the cancers and conditions associated with it. Table 1 comprises the details of kinase inhibitor drugs approved by the FDA from 2015 to 2021 [23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58].

S. No.DrugBrand nameYear of approvalInhibitory targetIndicationReferences
1.PalbociclibIbrance2015CDK4/6 inhibitorAdvanced metastatic breast cancer[23]
2.LenvatinibLenvima2015VEGFR1/2/3 inhibitorProgressive/differentiated thyroid cancer[24]
3.CobimetinibCotellic2015MEK inhibitorMelanoma[25]
4.OsimertinibTagrisso2015EGFR inhibitorNon-small cell lung carcinomas with specific mutations[26]
5.NecitumumabPortrazza2015EGFR antibodyAdvanced (metastatic) squamous non-small cell lung cancer[27]
6.AlectinibAlecensa2015ALK inhibitorNon-small cell lung cancer[28]
7.OlaratumabLartruvo2016PDGFRA inhibitorSoft tissue sarcoma[29]
8.RibociclibKisqali2016CDK4/6 inhibitorAdvanced breast cancer[30]
9.BrigatinibAlunbrig2017ALK and EGFR inhibitorNon-small cell lung cancer[31]
10.CopanlisibAliqopa2017PI3K inhibitorRelapsed follicular lymphoma[32]
11.AbemaciclibVerzenio2017CDK4/6 inhibitorsAdvanced metastatic breast cancer[33]
12.AcalabrutinibCalquence2017BTK inhibitorMantle cell lymphoma[34]
13.BinimetinibMektovi2018MEK inhibitorUnresectable or metastatic melanoma[35]
14.EncorafenibBraftovi2018MEK inhibitorUnresectable or metastatic melanoma[36]
15.DuvelisibCopiktra2018PI3K inhibitorRefractory chronic lymphocytic leukemia, small lymphocytic lymphoma, and follicular lymphoma[37]
16.DacomitinibVizimpro2018EGFR inhibitorMetastatic non-small cell lung cancer[38]
17.LorlatinibLorbrena2018ALK and ROS1 inhibitorMetastatic non-small cell lung cancer[39]
18.GilteritinibXospata2018AXL inhibitorRelapsed or refractory acute myeloid leukemia[40]
19.ErdafitinibBalversa2019FGFR inhibitorLocally advanced or metastatic bladder cancer[41]
20.AlpelisibPiqray2019PI3K inhibitorBreast cancer[42]
21.PexidartinibTuralio2019inhibitor of CSF1, KIT, and FLT3Symptomatic tenosynovial giant cell tumor[43]
22.EntrectinibRozlytrek2019inhibitor of ALK, ROS1, TKI, and TRKA/B/CMetastatic non-small cell lung cancer[44]
23.ZanubrutinibBrukinsa2019BTK inhibitorMantle cell lymphoma[45]
24.AvapritinibAyvakit2020PDGFRA receptor kinase inhibitorMetastatic gastrointestinal stromal tumors[46]
25.SelumetinibKoselugo2020BRAF kinase inhibitorNeurofibromatosis type I[47]
26.TucatinibTukyssa2020EBBR2 inhibitorMetastatic HER2-positive breast cancer[48]
27.PemigatinibPemazyre2020FGFR2 inhibitorAdvanced/metastatic or surgically unresectable cholangiocarcinoma[49]
28.CapmatinibTabrecta2020MET kinase inhibitorMetastatic non-small cell lung cancer[50]
29.SelpercatinibRetevmo2020RET receptor kinaseNon-small cell lung cancer, metastatic medullary thyroid cancer, or advanced or metastatic thyroid cancer[51]
30.RipretinibQinlock2020PDGFRA and KIT receptor kinase inhibitorGist[52]
31.PralsetinibGavreto2020RET receptor kinase inhibitorThyroid cancer, non-small cell lung cancer[53]
32.MargetuximabMargenza2020HER2 inhibitorHER2-positive breast cancer[54]
33.TrilaciclibCosela2021CDK4/6 inhibitorExtensive-stage small cell lung cancer[55]
34.InfigratinibTruseltiq2021FGFR2 inhibitorCholangiocarcinomas with FGFR2 fusion proteins[56]
35.TepotinibTepmetco2021Met KinaseMet mutation-positive non-small cell lung carcinoma[57]
36.TivozanibFotvida2021VEGFR2 inhibitorRenal cell carcinoma[58]

Table 1.

FDA-approved kinase inhibitors against various cancers during 2015–2021.

The modern strategies adopted for the development of selective kinase inhibitors include synthesis along with structure-based design approaches facilitated by molecular docking, crystallographic studies, and NMR spectroscopy [59]. It is surprising that alone USA has filed more than 10 thousand patent applications for kinase inhibitors since 2001. Beyond the discovery of small-molecule kinase inhibitors, kinase-targeted antibodies have also been postulated against different cancers such as cetuximab (colorectal, head, and neck cancer), trastuzumab (breast cancer) [60]. Various small-molecule kinase inhibitors have different inhibitory modes and on the basis of these modes, these inhibitors have been divided into five categories (Figure 2). Type I inhibitors contain a heterocyclic moiety in their structure to occupy purine binding pocket and serves as a template for side chains to occupy the hydrophobic region. These inhibitors are basically ATP-binding site competitors and mimic the purine ring of ATP. These bind to the active conformational side and cause alteration of structural conformation [61]. Type II inhibitors target the inactive conformation and occupy the catalytic region of the unphosphorylated inactive conformation. These kinases explore the new binding patterns in the hydrophobic pocket associated with conformational changes of phenylalanine residue of the Asp-Phe-Gly (DFG) system [62]. Type III inhibitors are regarded as allosteric inhibitors and exhibit their action via binding to the outer catalytic ATP-binding site and alter kinase activity in an allosteric way. Type IV kinase inhibitors are regarded as substrate-directed inhibitors and undergo reversible binding outside the ATP pocket. These are non-competitive inhibitors and do not compete with ATP [63]. Type V inhibitors are covalent inhibitors and bind through an irreversible covalent bond to catalytic nucleophilic cysteine active site of the enzyme.

Figure 2.

Inhibitory patterns of different kinase inhibitors.

From a clinical point of view, it has been observed that kinase target anticancer therapies have more success rate than the other cancer therapies. But it is also evident in the past few years EGFR/VEGF-targeting molecules have given unsatisfactory results [64, 65]. Instead, success stories have been seen with molecules targeting kinase B, phosphatidylinositol kinase delta and gamma, kinase I, tyrosine kinase, nerve growth receptors Wee 1-like kinases in Phase 1 clinical trials. The latest explored targets Aurora kinases have led to the development of two inhibitors palbociclib and ribociclib which have passed phase III clinical trials [66]. The modern developments on kinases are following the precision therapy that has been based upon the genomic data. The detailed genetic studies on tumors and drivers involved in the generation of tumors have resulted in tremendous advantages for patients who need effective therapy.

Advertisement

3. Investigations on kinase inhibitory potentials of natural products

The continuous research is in progress for several years to design synthetic and natural chemotherapeutic agents against cancer with selective cytotoxic efficacy and minimum toxicity [67, 68, 69, 70]. The contribution of molecules from natural sources in kinase-mediated anticancer research cannot be ignored. The kinase modulating properties of natural molecules has brought a new paradigm in the screening of kinase inhibitors. Toward this direction, small molecules like polyphenols have revealed tremendous potentials to bind with kinases like tyrosine kinase followed by alteration of phosphorylation leading to modulation of multi-signaling mechanisms. The explored natural compounds in this direction are curcumins, resveratrol, quercetin, cyrysitin, myricetin, luteolin, apigenin, anthocyanin, genistein, epigallocatechin gallate, fisetin, astaxanthin, and tetrahydrocurcumins and many more. Polyphenols such as resveratrol [71], quercetin [72], curcumin [73] and tea extracts [74] have revealed promising EGFR inhibition [75]. Curcumin and chrysin have receptor RON blocker activity in tumor cells [76, 77]. Natural products have also shown Abl, JAK-2, c-Met, c-SRC, and serine kinase inhibitory potentials [78, 79, 80]. Resveratrol also has modulatory effects on the expression of Akt in breast, uterine, skin, and prostate cancers [81, 82]. It binds to the ATP site competitively as well as reversibly. Myricetin has reported inhibition of cell proliferation by binding to Akt. Beyond these significant activities, several reports in the literature are available evidencing the inhibitory and modulatory effects of natural products on mTOR, CDK, Aurora kinases, B-raf kinases, PI3K [83, 84, 85], etc. Many natural molecules bind directly to the oncogenic kinases and alter the cell signaling involved in tumor progression by modifying the phosphorylation process. Several other classes of natural compounds are under investigation for their kinase-modulating activities.

Advertisement

4. Conclusions and future perspectives

The therapeutic implication of protein kinases against a variety of cancers is well known from past decades. Also, it is well established that deregulation, mutations, and overexpression of these kinases are important triggers for the development of cancers. Several kinase inhibitors are already reported who prevent cancer by modulating the protein kinases by following different mechanisms and several inhibitors are under investigation. Despite tremendous advancements in kinase drug development, still, a large number of kinases are unexplored. It is also worth notable that most of the available kinase inhibitors work through binding to ATP sites. A great challenge in clinical implication of kinase inhibitors is the development of drug resistance of cancer stem cells. It develops due to the loss of activity of some important kinases. Therefore, strategies to overcome this resistance are the requirement of the hour. In the therapeutics of cancer, the kinase inhibitors have been proven to be well tolerated as compared to the traditional therapies.

Advertisement

Abbreviations

ABLAbelson murine leukemia viral oncogene
AblAbelson murine leukemia
Aktprotein kinase B
ALKanaplastic lymphoma kinase
BRAFproto-oncogene
BTKBruton agammaglobulinemia tyrosine kinase
CDKcyclin-dependent kinase
c-Metc-MET proto-oncogene
c-SRCproto-oncogene tyrosine-protein kinase
CTKcytoplasmic tyrosine kinase
EGFRepidermal growth factor receptor
ERBB2V-Erb-B2 avian erythroblastic leukemia viral oncogene homolog
FGFRsfibroblast growth factor receptors
HER-2human epidermal growth factorreceptor-2
JAK2Janus kinase 2
MAPKmitogen-activated protein kinases
MEKMEK kinase gene
PDGFRsplatelet-derived growth factor receptors
PI3Kphosphatidylinositol-3-kinase
PI3KCAphosphatidylinositol-4,5-bisphosphate 3-kinase
RTKreceptor tyrosine kinase
VEGFR-2vascular endothelial growth factor receptor 2

References

  1. 1. Bhatia R, Rawal RK. Coumarin hybrids: Promising scaffolds in the treatment of breast cancer. Mini Reviews in Medicinal Chemistry. 2019;19(17):1443-1458
  2. 2. Nagai H, Kim YH. Cancer prevention from the perspective of global cancer burden patterns. Journal of Thoracic Disease. 2017;9(3):448-451
  3. 3. Bhullar KS, Lagarón NO, McGowan EM, et al. Kinase-targeted cancer therapies: Progress, challenges and future directions. Molecular Cancer. 2018;17:48
  4. 4. Berndt N, Karim RM, Schönbrunn E. Advances of small molecule targeting of kinases. Current Opinion in Chemical Biology. 2017;39:126-132
  5. 5. Cohen P, Cross D, Jänne PA. Kinase drug discovery 20 years after imatinib: Progress and future directions. Nature Reviews. Drug Discovery. 2021;20:551-569
  6. 6. Pottier C, Fresnais M, Gilon M, Jérusalem G, Longuespée R, Sounni NE. Tyrosine kinase inhibitors in cancer: Breakthrough and challenges of targeted therapy. Cancers. 2020;12(3):731
  7. 7. Yu JS, Cui W. Proliferation, survival and metabolism: The role of PI3K/AKT/mTOR signalling in pluripotency and cell fate determination. Development. 2016;143:3050-3060
  8. 8. Ardito F, Giuliani M, Perrone D, Troiano G, Lo ML. The crucial role of protein phosphorylation in cell signaling and its use as targeted therapy (review). International Journal of Molecular Medicine. 2017;40(2):271-280
  9. 9. Lemmon MA, Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell. 2010;141:1117-1134
  10. 10. 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
  11. 11. Bononi A, Agnoletto C, De Marchi E, et al. Protein kinases and phosphatases in the control of cell fate. Enzyme Research. 2011;2011:329098
  12. 12. Kannaiyan R, Mahadevan D. A comprehensive review of protein kinase inhibitors for cancer therapy. Expert Review of Anticancer Therapy. 2018;18(12):1249-1270
  13. 13. Alkharsah KR. VEGF upregulation in viral infections and its possible therapeutic implications. International Journal of Molecular Sciences. 2018;19(6):1642
  14. 14. Soroceanu L, Akhavan A, Cobbs CS. Platelet-derived growth factor-alpha receptor activation is required for human cytomegalovirus infection. Nature. 2008;455(7211):391-395
  15. 15. Nukaga S, Yasuda H, Tsuchihara K, Hamamoto J, Masuzawa K, Kawada I, et al. Amplification of egfr wild-type alleles in non-small cell lung cancer cells confers acquired resistance to mutation-selective egfr tyrosine kinase inhibitors. Cancer Research. 2017;77:2078-2089
  16. 16. Khan SA, Zeng Z, Shia J, Paty PB. Egfr gene amplification and kras mutation predict response to combination targeted therapy in metastatic colorectal cancer. Pathology Oncology Research. 2017;23:673-677
  17. 17. Zhou C, Zhu L, Ji J, Ding F, Wang C, Cai Q , et al. Egfr high expression, but not kras status, predicts sensitivity of pancreatic cancer cells to nimotuzumab treatment in vivo. Current Cancer Drug Targets. 2017;17:89-97
  18. 18. Morey AL, Brown B, Farshid G, Fox SB, Francis GD, McCue G, et al. Determining her2 (erbb2) amplification status in women with breast cancer: Final results from the australian in situ hybridisation program. Pathology. 2016;48:535-542
  19. 19. Wang YK, Yang BF, Yun T, Zhu CY, Li CY, Jiang B, et al. Methods and significance of the combined detection of her2 gene amplification and chemosensitivity in gastric cancer. Cancer Biomarkers. 2017;21:439-447
  20. 20. Xu CW, Wang WX, Wu MJ, Zhu YC, Zhuang W, Lin G, et al. Comparison of the c-met gene amplification between primary tumor and metastatic lymph nodes in non-small cell lung cancer. Thoracic Cancer. 2017;8:417-422
  21. 21. Blanke CD, Demetri GD, von Mehren M, Heinrich MC, Eisenberg B, Fletcher JA, et al. Long-term results from a randomized phase II trial of standard versus higher-dose imatinib mesylate for patients with unresectable or metastatic gastrointestinal stromal tumors expressing KIT. Journal of Clinical Oncology. 2008;26:620-625
  22. 22. Cohen P. Protein kinases—The major drug targets of the twenty-first century? Nature Reviews. Drug Discovery. 2002;1:309-315
  23. 23. Wedam S, Fashoyin-Aje L, Bloomquist E, Tang S, Sridhara R, Goldberg KB, et al. FDA approval summary: Palbociclib for male patients with metastatic breast cancer. Clinical Cancer Research. 2020;26(6):1208-1212
  24. 24. Nair A, Reece K, Donoghue MB, Yuan WV, Rodriguez L, Keegan P, et al. FDA supplemental approval summary: Lenvatinib for the treatment of unresectable hepatocellular carcinoma. The Oncologist. 2021;26(3):e484-e491
  25. 25. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-osimertinib-first-line-treatment-metastatic-nsclc-most-common-egfr-mutations [Accessed: 4 September 2021]
  26. 26. Greig SL. Osimertinib: First global approval. Drugs. 2016;76(2):263-273
  27. 27. Fala LP. Necitumumab: An IgG1 monoclonal antibody, FDA approved for advanced squamous non-small-cell lung cancer. American Health & Drug Benefits. 2016;9(Spec Feature):119-122
  28. 28. Larkins E, Blumenthal GM, Chen H, He K, Agarwal R, Gieser G, et al. FDA approval: Alectinib for the treatment of metastatic, ALK-positive non-small cell lung cancer following crizotinib. Clinical Cancer Research. 2016;22(21):5171-5176
  29. 29. Shirley M. Olaratumab: First global approval. Drugs. 2017;77(1):107-112
  30. 30. Shah A, Bloomquist E, Tang S, Fu W, Bi Y, Liu Q , et al. FDA approval: Ribociclib for the treatment of postmenopausal women with hormone receptor-positive, HER2-negative advanced or metastatic breast cancer. Clinical Cancer Research. 2018;24(13):2999-3004
  31. 31. https://www.fda.gov/drugs/resources-information-approved-drugs/brigatinib [Accessed: 4 September 2021]
  32. 32. Kumar A, Bhatia R, Chawla P, Anghore D, Saini V, Rawal RK. Copanlisib: Novel PI3K inhibitor for the treatment of lymphoma. Anti-Cancer Agents in Medicinal Chemistry. 2020;20(10):1158-1172
  33. 33. Voli L, Mamyrbékova J, Bazureau J. Abemaciclib, a recent novel FDA-Approved small molecule inhibiting cyclin-dependant kinase 4/6 for the treatment of metastatic breast cancer: A mini-review. Open Journal of Medicinal Chemistry. 2020;10:128-138
  34. 34. https://www.cancer.gov/news-events/cancer-currents-blog/2017/acalabrutinib-fda-mantle-cell-lymphoma [Accessed: 4 September 2021]
  35. 35. Shirley M. Encorafenib and binimetinib: First global approvals. Drugs. 2018;78(12):1277-1284
  36. 36. Kopetz S, Grothey A, Yaeger R, Van Cutsem E, Desai J, Yoshino T, et al. Encorafenib, binimetinib, and cetuximab in BRAF V600E-mutated colorectal cancer. The New England Journal of Medicine. 2019;381(17):1632-1643
  37. 37. Rodrigues DA, Sagrillo FS, Fraga CAM. Duvelisib: A 2018 novel FDA-approved small molecule inhibiting phosphoinositide 3-kinases. Pharmaceuticals. 2019;12(2):69
  38. 38. Shirley M. Dacomitinib: First global approval. Drugs. 2018;78(18):1947-1953
  39. 39. Solomon BJ, Besse B, Bauer TM, Felip E, Soo RA, Camidge DR, et al. Lorlatinib in patients with ALK-positive non-small-cell lung cancer: Results from a global phase 2 study. The Lancet Oncology. 2018;19(12):1654-1667
  40. 40. Perl AE, Martinelli G, Cortes JE, Neubauer A, Berman E, Paolini S, et al. Gilteritinib or chemotherapy for relapsed or refractory FLT3-mutated AML. The New England Journal of Medicine. 2019;381(18):1728-1740
  41. 41. Montazeri K, Bellmunt J. Erdafitinib for the treatment of metastatic bladder cancer. Expert Review of Clinical Pharmacology. 2020;13(1):1-6
  42. 42. Tayyar Y, Idris A, Vidimce J, Ferreira DA, McMillan NA. Alpelisib and radiotherapy treatment enhances Alisertib-mediated cervical cancer tumor killing. American Journal of Cancer Research. 2021;11(6):3240-3251
  43. 43. Benner B, Good L, Quiroga D, et al. Pexidartinib, a novel small molecule CSF-1R inhibitor in use for tenosynovial giant cell tumor: A systematic review of pre-clinical and clinical development. Drug Design, Development and Therapy. 2020;14:1693-1704
  44. 44. Sartore-Bianchi A, Pizzutilo EG, Marrapese G, Tosi F, Cerea G, Siena S. Entrectinib for the treatment of metastatic NSCLC: Safety and efficacy. Expert Review of Anticancer Therapy. 2020;20(5):333-341
  45. 45. Trotman J, Opat S, Gottlieb D, Simpson D, Marlton P, Cull G, et al. Zanubrutinib for the treatment of patients with Waldenström macroglobulinemia: 3 years of follow-up. Blood. 2020;136(18):2027-2037
  46. 46. Heinrich MC, Jones RL, von Mehren M, Schöffski P, Serrano C, Kang YK, et al. Avapritinib in advanced PDGFRA D842V-mutant gastrointestinal stromal tumour (NAVIGATOR): A multicentre, open-label, phase 1 trial. The Lancet Oncology. 2020;21:7935-7946
  47. 47. Gross AM, Wolters PL, Dombi E, Baldwin A, Whitcomb P, Fisher MJ, et al. Selumetinib in children with inoperable plexiform neurofibromas. The New England Journal of Medicine. 2020;382:1430-1442
  48. 48. Murth RK, Loi S, Okines A, Paplomata E, Hamilton E, Hurvitz SA, et al. Tucatinib, trastuzumab, and capecitabine for HER2-positive metastatic breast cancer. The New England Journal of Medicine. 2020;382:597-609
  49. 49. Abou-Alfa GK, Sahai V, Hollebecque A, Vaccaro G, Melisi D, Al-Rajabi R, et al. Pemigatinib for previously treated, locally advanced or metastatic cholangiocarcinoma: A multicentre, open-label, phase 2 study. The Lancet Oncology. 2020;21:671-684
  50. 50. Wolf J, Seto T, Han JY, Reguart N, Garon EB, Groen HJM, et al. Capmatinibin MET exon 14-mutated or MET-amplified non-small-cell lung cancer. The New England Journal of Medicine. 2020;383:944-957
  51. 51. Wirth LJ, Sherman E, Robinson B, Solomon B, Kang H, Lorch J, et al. Efficacy of selpercatinib in RET-altered thyroid cancers. The New England Journal of Medicine. 2020;383:825-835
  52. 52. Blay JY, Serrano C, Heinrich MC, Zalcberg J, Bauer S, Gelderblom H, et al. Ripretinib in patients with advanced gastrointestinal stromal tumours (INVICTUS): A double-blind, randomised, placebo controlled, phase 3 trial. The Lancet Oncology. 2020;21:923-934
  53. 53. Hu M, Subbiah V, Wirth LJ, Schuler M, Mansfield AS, Brose MS, et al. Results from the registrational phase I/II ARROW trial of pralsetinib (BLU-667) in patients (pts) with advanced RET mutation positive medullary thyroid cancer (RET + MTC). ESMO Virtual Congress. 2020;31:S1084
  54. 54. Rugo HS, Im SA, Cardoso F, Cortés J, Curigliano G, Musolino A, et al. Efficacy of margetuximab vs trastuzumab in patients with pretreated ERBB2-positive advanced breast cancer: A phase 3 randomized clinical trial. JAMA Oncology. 2021;7(4):573-584
  55. 55. Tam CS, Opat S, Simpson D, Cull G, Munoz J, Phillips TJ, et al. Zanubrutinib for the treatment of relapsed or refractory mantle cell lymphoma. Blood Advances. 2021;5(12):2577-2585
  56. 56. Huynh H, Lee LY, Goh KY, et al. Infigratinib mediates vascular normalization, impairs metastasis, and improves chemotherapy in hepatocellular carcinoma. Hepatology. 2019;69(3):943-958
  57. 57. Tanaka H, Taima K, Makiguchi T, Nakagawa J, Niioka T, Tasaka S. Activity and bioavailability of tepotinib for leptomeningeal metastasis of NSCLC with MET exon 14 skipping mutation. Cancer Communications. 2021;41(1):83-87
  58. 58. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-tivozanib-relapsed-or-refractory-advanced-renal-cell-carcinoma [Accessed: 4 September 2021]
  59. 59. Jonker DJ, O’Callaghan CJ, Karapetis CS, Zalcberg JR, Tu D, Au H-J, et al. Cetuximab for the treatment of colorectal cancer. The New England Journal of Medicine. 2007;357:2040-2048
  60. 60. Hudis CA. Trastuzumab—Mechanism of action and use in clinical practice. The New England Journal of Medicine. 2007;357:39-51
  61. 61. Dar AC, Shokat KM. The evolution of protein kinase inhibitors from antagonists to agonists of cellular signaling. Annual Review of Biochemistry. 2011;80:769-795
  62. 62. Roskoski R. Classification of small molecule protein kinase inhibitors based upon the structures of their drug-enzyme complexes. Pharmacological Research. 2016;103:26-48
  63. 63. Zuccotto F, Ardini E, Casale E, Angiolini M. Through the “gatekeeper door”: Exploiting the active kinase conformation. Journal of Medicinal Chemistry. 2009;53:2681-2694
  64. 64. Takeda M, Nakagawa K. Toxicity profile of epidermal growth factor receptor tyrosine kinase inhibitors in patients with epidermal growth factor receptor gene mutation-positive lung cancer. Molecular and Clinical Oncology. 2017;6:3-6
  65. 65. Yr L, Zhu W, Jl Z, Jq H, Zhao YZ, Zhang W, et al. The evaluation of efficacy and safety of sunitinib on EGFR-TKI pretreated advanced non-small cell lung cancer patients in China. The Clinical Respiratory Journal. 2014;8:206-212
  66. 66. Knapp S, Sundström M. Recently targeted kinases and their inhibitors—The path to clinical trials. Current Opinion in Pharmacology. 2014;17:58-63
  67. 67. Kumar S, Sharma B, Bhardwaj TR, Singh RK. Design, synthesis and studies on novel polymeric prodrugs of erlotinib for colon drug delivery. Anti-Cancer Agents in Medicinal Chemistry. 2021;21(3):383-392
  68. 68. Singh RK, Prasad DN, Bhardwaj TR. Synthesis, in vitro/in vivo evaluation and in silico physicochemical study of prodrug approach for brain targeting of alkylating agent. Medicinal Chemistry Research. 2013;22:5324-5336
  69. 69. Singh RK, Kumar S, Prasad DN, Bhardwaj TR. Therapeutic journey of nitrogen mustard as alkylating anticancer agents: Historic to future perspectives. European Journal of Medicinal Chemistry. 2018;151:401-433
  70. 70. Singh RK, Kumar S, Prasad DN, Bhardwaj TR. Reversible redox system based drug design for targeting alkylating agent across brain. Medicinal Chemistry Research. 2014;23(5):2405-2416
  71. 71. Jeong KJ, Cho KH, Panupinthu N, Kim H, Kang J, Park CG, et al. EGFR mediates LPA-induced proteolytic enzyme expression and ovarian cancer invasion: Inhibition by resveratrol. Molecular Oncology. 2013;7:121-129
  72. 72. Huang C-Y, Chan C-Y, Chou I-T, Lien C-H, Hung H-C, Lee M-F. Quercetin induces growth arrest through activation of FOXO1 transcription factor in EGFR-overexpressing oral cancer cells. The Journal of Nutritional Biochemistry. 2013;24:1596-1603
  73. 73. Sun X-D, Liu X-E, Huang D-S. Curcumin induces apoptosis of triple-negative breast cancer cells by inhibition of EGFR expression. Molecular Medicine Reports. 2012;6:1267-1270
  74. 74. Masuda M, Wakasaki T, Toh S, Shimizu M, Adachi S. Chemoprevention of head and neck cancer by green tea extract: EGCG—The role of EGFR signaling and “lipid raft”. Journal of Oncology. 2011;2011:540148
  75. 75. Nicholson R, Gee J, Harper M. EGFR and cancer prognosis. European Journal of Cancer. 2001;37:9-15
  76. 76. Sakurai R, Villarreal P, Husain S, Liu J, Sakurai T, Tou E, et al. Curcumin protects the developing lung against long-term hyperoxic injury. American Journal of Physiology—Lung Cellular and Molecular Physiology. 2013;305:L301-L311
  77. 77. Narasimhan M, Ammanamanchi S. Curcumin blocks RON tyrosine kinase-mediated invasion of breast carcinoma cells. Cancer Research. 2008;68:5185-5192
  78. 78. Greuber EK, Smith-Pearson P, Wang J, Pendergast AM. Role of ABL family kinases in cancer: From leukaemia to solid tumours. Nature Reviews. Cancer. 2013;13:559-571
  79. 79. Byun S, Lee KW, Jung SK, Lee EJ, Hwang MK, Lim SH, et al. Luteolin inhibits protein kinase Cε and c-Src activities and UVB induced skin cancer. Cancer Research. 2010;70:2415-2423
  80. 80. Wolanin K, Magalska A, Mosieniak G, Klinger R, Mckenna S, Vejda S, et al. Curcumin affects components of the chromosomal passenger complex and induces mitotic catastrophe in apoptosis-resistant Bcr-Abl expressing cells. Molecular Cancer Research. 2006;4:457-469
  81. 81. Jiang H, Shang X, Wu H, Gautam SC, Al-Holou S, Li C, et al. Resveratrol downregulates PI3K/Akt/mTOR signaling pathways in human U251 glioma cells. Journal of Experimental Therapeutics & Oncology. 2009;8:25
  82. 82. Fröjdö S, Cozzone D, Vidal H, Pirola L. Resveratrol is a class IA phosphoinositide 3-kinase inhibitor. The Biochemical Journal. 2007;406:511-518
  83. 83. Park D, Jeong H, Lee MN, Koh A, Kwon O, Yang YR, et al. Resveratrol induces autophagy by directly inhibiting mTOR through ATP competition. Scientific Reports. 2016;6:21772
  84. 84. Kumamoto T, Fujii M, Hou D-X. Akt is a direct target for myricetin to inhibit cell transformation. Molecular and Cellular Biochemistry. 2009;332:33-41
  85. 85. Weir NM, Selvendiran K, Kutala VK, Tong L, Vishwanath S, Rajaram M, et al. Curcumin induces G2/M arrest and apoptosis in cisplatin-resistant human ovarian cancer cells by modulating Akt and p38 MAPK. Cancer Biology and Therapy. 2007;6:178-184

Written By

Rohit Bhatia and Rajesh K. Singh

Published: 08 December 2021