Flyer

Archives in Cancer Research

  • ISSN: 2254-6081
  • Journal h-index: 14
  • Journal CiteScore: 3.77
  • Journal Impact Factor: 4.09
  • Average acceptance to publication time (5-7 days)
  • Average article processing time (30-45 days) Less than 5 volumes 30 days
    8 - 9 volumes 40 days
    10 and more volumes 45 days
Awards Nomination 20+ Million Readerbase
Indexed In
  • China National Knowledge Infrastructure (CNKI)
  • CiteFactor
  • OCLC- WorldCat
  • Publons
  • Geneva Foundation for Medical Education and Research
  • Euro Pub
  • Google Scholar
  • J-Gate
  • Secret Search Engine Labs
  • International Committee of Medical Journal Editors (ICMJE)
  • Zenodo
Share This Page

Review Article - (2015) Volume 3, Issue 3

Clinical Challenges to Current Molecularly Targeted Therapies in Lung Cancer

Gagan Chhabra, Ashley Eggert and Neelu Puri*
Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, Illinois, USA
Corresponding Author: Neelu Puri, Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, Illinois, USA,Tel: +18153955678, E-mail: neelupur@uic.edu
Related article at Pubmed, Scholar Google
Visit for more related articles at Archives in Cancer Research

Abstract

Lung cancer is difficult to treat with a poor prognosis and a five year survival of 15%. Current molecularly targeted therapies are initially effective in non-small cell lung cancer (NSCLC) patients; however, they are plagued with difficulties including induced resistance and small therapeutically responsive populations. This mini review describes the mechanism of resistance to several molecularly targeted therapies which are currently being used to treat NSCLC. The major targets discussed are c-Met, EGFR, HER2, ALK, VEGFR, and BRAF. The first generation tyrosine kinase inhibitors (TKIs) resulted in resistance; however, second and third generation TKIs are being developed, which are generally more efficacious and have potential to treat NSCLC patients with resistance to first generation TKIs. Combination therapies could also be effective in preventing TKI resistance in NSCLC patients.

Keywords

NSCLC, Molecularly targeted therapies, TKI, Resistance

Introduction

The focus of current lung cancer treatment has been shifted from more traditional options to newly developed molecularly targeted therapies. Many of the molecularly targeted therapies are utilized to target specific biomarkers that are commonly overexpressed and have important roles in tumorigenesis; these biomarkers contribute to cancer-related processes such as cell proliferation, survival and migration. While initially effective, many targeted therapies have been associated with increased drug resistance after their initial use. Acquired resistance to current molecularly targeted therapies in lung cancer presents a major clinical challenge. Current research focuses on identifying potential novel biomarkers and mechanisms involved in resistance to these therapies. There are several clinical challenges associated with current molecularly targeted therapies including the induction of various types of resistance mechanisms, which are not clearly defined, and the lack of effective combinatorial therapies designed to prevent and overcome the problem of drug resistance in lung cancer.

Current Therapies

Common molecularly targeted therapies target receptor tyrosine kinases (RTKs) including hepatocyte growth factor receptor (HGFR/c-Met), epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER2), anaplastic lymphoma kinase (ALK), and endothelial growth factor receptor (VEGFR), which are commonly mutated in NSCLC cases [1]. Recently, v-Raf murine sarcoma viral oncogene homolog B1 (BRAF) has also been shown as a potential target for treatment of advanced NSCLC patients having mutated BRAF. Mutations in these RTKs cause uncontrolled upregulation and amplification of various downstream signaling pathways including MAP kinase (mitogenactivated protein kinases), PI3K (phosphoinositide 3-kinase)/AKT (protein kinase B) and mTOR (mammalian target of rapamycin) pathways; these pathways are responsible for cell survival, proliferation, migration, protein synthesis, and angiogenesis of cancerous cells [2]. In order to inhibit cell growth and proliferation, many tyrosine kinase inhibitor inhibitors (TKIs) have been developed that act by binding to RTKs and inhibiting their downstream signaling cascades [1].
c-Met is a RTK for the ligand hepatocyte growth factor (HGF), which is secreted by mesenchymal cells and cancer cells [3]. There have been several monoclonal antibodies designed to target c-Met/HGF including rilotumumab (AMG 102), ficlatuzumab (AV- 299), onartuzumab (MetMAb), as well as TKIs including tivantinib (ARQ-197), cabozantinib (XL184/BMS-907351), crizotinib (PF- 2341066), and foretinib (XL880, GSK1363089) [4]. For each of these TKIs, resistance is a major concern [5] and several mechanisms for resistance have already been proposed. One study showed that MET-dependent NSCLC cells that had become resistant displayed high levels of c-Met and KRAS (kirsten rat sarcoma viral oncogene homolog) amplification, leading to downstream MAP kinase activity [6]. Another study found that inhibition of c-Met in Met-amplified NSCLC led to activation of the EGFR pathway [7]. However, in a gastric carcinoma cellline, a mutation in the c-Met activation loop has been shown to destabilize autoinhibitory conformational change, ultimately causing constitutive expression which could be a possible mechanism of c-Met TKI resistance [8].
Epidermal growth factor receptor (EGFR) is a transmembrane receptor that plays an essential role in regulating cell proliferation, survival, and growth [9]. EGFR TKIs inhibit receptor phosphorylation and downstream signaling by binding to the intracellular EGFR TK domain. The first generation of EGFR TKIs bind reversibly to the ATP binding site of the EGFR TK domain; due to high binding affinity for this domain, an inhibition of RTK activity is observed [10]. However, prolonged use of EGFR-TKIs can lead to distinct drug resistance patterns. The dominant resistance pattern is a common T790M secondary mutation. The T790M mutation induces resistance by interfering with TKIs binding to the ATP binding domain [11]. D761Y, T854A and L747S (Table 1) are additional secondary mutations that cause resistance; these arise subsequent to the EGFR TKI sensitizing L858R mutation [12]. Our recent studies indicate that the activation of alternative signaling pathways such as PI3K/mTOR and Wnt may also cause resistance to EGFR TKIs [13,14]. A second generation EGFR TKI, afatinib, which irreversibly binds to the ATP binding pocket of EGFR, was suggested to have potential to overcome TKI resistance. This inhibitor is efficacious in NSCLC patients who have T790M mutation which confers resistance to EGFR TKIs such as erlotinib [15], however it also has been shown to inhibit wild type EGFR that may result in dose limiting toxicities. AZD9291, CO-1686, and HM61713 are the third generation of TKIs that target both the sensitizing mutations and the T790M resistance mutation while sparing the wild type EGFR [16,17] and show potential to overcome resistance. HER2, another member of the EGFR family, also activates downstream signaling pathways such as RAS, PI3K, MAPK, and SRC. The HER2 TKI lapatinib and the HER2 antibody trastuzumab are originally very effective at blocking HER2 signaling, but their effectiveness decreases over time. This may be due to the T798M mutation [18]; however, the mechanism through which the T798M mutation confers resistance may be due to increased EGFR ligand production [18].
The anaplastic lymphoma kinase (ALK) is a RTK typically expressed in the central and peripheral nervous system regions [19,20]. ALK gene amplification, mutation and rearrangement are known to be associated with tumor development in lung cancer patients [21,22]; approximately 5% of NSCLC cases are diagnosed with ALK gene rearrangement [22]. Crizotinib, a small-molecule ALK TKI was the first FDA approved drug to treat patients with ALKrearranged NSCLC. However, the efficacy of crizotinib is limited to approximately one year due to the emergence of resistance patterns. Point mutations including L1196M, C1156Y, G1269A and F1174L in the kinase domain of ALK have been observed in biopsies from patients treated with crizotinib, a first generation ALK TKI, and have been found perturbing crizotinib binding to render it less effective [23-25]. Another study identified G1202R, S1206Y and 1151Tins point mutations in crizotinib treated ALK-positive NSCLC patients. Ceritinib, alectinib, and AP26113 are amongst the second generation of ALK TKIs with improved selectivity and potency compared to crizotinib. However, mutations in the ALK gene conferring resistance to alectinib (G1123S, G1202R, I1171T/ N/S, and V1180L) and ceritinib (G1202R and F1174C/V) have also been found (Table 1) [26-31].
Overexpression of vascular endothelial growth factor (VEGF), an angiogenic factor, and its receptors are related to poor prognosis in NSCLC patients [32]. Bevacizumab (a monoclonal antibody that targets VEGF) and aflibercept (a recombinant fusion protein that binds strongly to VEGF) are being explored clinically to block VEGF pathways in NSCLC patients [33,34]. Acquired resistance to anti-VEGF therapy usually occurs via several distinct mechanisms [35] including expression of additional proangiogenic pathways including platelet derived growth factor (PDGF) and fibroblast derived growth factor (FGF) [36].
BRAF (v-Raf murine sarcoma viral oncogene homolog B1) is a member of the RAF serine/threonine protein kinases family. Mutations in BRAF have been shown to be associated with tumor development in NSCLC with a frequency of 2–3%. Recently, a BRAF inhibitor dabrafenib, the first drug of its class, is shown to be effective for the treatment of advanced NSCLC patients with BRAF V600E mutation in a phase II clinical study [37]. However, one study reported acquired resistance to dabrafenib in a patient after 8 months of response. An acquired G12D mutation (Table 1) in KRAS has been suggested to be primarily responsible for acquired dabrafenib resistance in this patient [38]. Further studies are required to understand the therapeutic potential of this inhibitor.

Conclusion

Although current molecularly targeted therapies are very effective for NSCLC patients, almost all patients eventually acquire resistance to these therapies. To combat this resistance against first generation TKIs, second and third generation TKIs have been developed. These new generations of TKIs are either completing clinical trials or have been FDA approved to treat NSCLC patients. However, their therapeutic potential needs to be further validated and established. Various secondary mutations and alternative signaling pathways have been identified as distinct resistance patterns for several TKIs targeting EGFR, c-Met, and ALK. However, further studies are required to determine the specific mechanisms of acquired resistance to HER2, VEGFR and BRAF. Combinatorial strategies could be effective in overcoming TKI resistance in lung cancer patients. These strategies may require targeting both mutations involved in resistance and alternative signaling pathways.

Tables at a glance

Table icon
Table 1
 
 
7348

References

  1. Domvri K, Zarogoulidis P, Darwiche K, Browning RF, Li Q, et al. (2013) Molecular Targeted Drugs and Biomarkers in NSCLC, the Evolving Role of Individualized Therapy. J Cancer 4: 736-754.
  2. Ciuffreda L, Incani UC, Steelman LS, Abrams SL, Falcone I, et al. (2014) Signaling intermediates (MAPK and PI3K) as therapeutic targets in NSCLC. Curr Pharm Des 20: 3944-3957.
  3. Grata ML, Rocchi L, Farinatti MT, Mantovani P (1988) [Acute cytomegalovirus hepatitis in non transfused subjects]. ClinTer 127: 49-52.
  4. Sadiq AA, Salgia R (2013) MET as a possible target for non-small-cell lung cancer. J ClinOncol 31: 1089-1096.
  5. Sierra JR, Cepero V, Giordano S (2010) Molecular mechanisms of acquired resistance to tyrosine kinase targeted therapy. Mol Cancer 9: 75.
  6. Cepero V, Sierra JR, Corso S, Ghiso E, Casorzo L, et al. (2010) MET and KRAS gene amplification mediates acquired resistance to MET tyrosine kinase inhibitors. Cancer Res 70: 7580-7590.
  7. McDermott U, Pusapati RV, Christensen JG, Gray NS, Settleman J (2010) Acquired resistance of non-small cell lung cancer cells to MET kinase inhibition is mediated by a switch to epidermal growth factor receptor dependency. Cancer research 70:1625-1634.
  8. Qi J, McTigue MA, Rogers A, Lifshits E, Christensen JG, et al. (2011) Multiple mutations and bypass mechanisms can contribute to development of acquired resistance to MET inhibitors. Cancer Res 71: 1081-1091.
  9. Mendelsohn J, Baselga J (2006) Epidermal growth factor receptor targeting in cancer. SeminOncol 33: 369-385.
  10. Sattler M, Abidoye O, Salgia R (2008) EGFR-targeted therapeutics: focus on SCCHN and NSCLC. ScientificWorldJournal 8: 909-919.
  11. Gazdar AF (2009) Activating and resistance mutations of EGFR in non-small-cell lung cancer: role in clinical response to EGFR tyrosine kinase inhibitors. Oncogene 28: S24-31.
  12. Ray M, Salgia R, Vokes EE (2009) The role of EGFR inhibition in the treatment of non-small cell lung cancer. Oncologist 14: 1116-1130.
  13. Botting GM, Rastogi I, Chhabra G, Nlend M, Puri N (2015) Mechanism of Resistance and Novel Targets Mediating Resistance to EGFR and c-Met Tyrosine Kinase Inhibitors in Non-Small Cell Lung Cancer. PloS one 10: e0136155.
  14. Fong JT, Jacobs RJ, Moravec DN, Uppada SB, Botting GM, et al. (2013) Alternative signaling pathways as potential therapeutic targets for overcoming EGFR and c-Met inhibitor resistance in non-small cell lung cancer. PloS one 8: e78398.
  15. Engle JA, Kolesar JM2 (2014) Afatinib: A first-line treatment for selected patients with metastatic non-small-cell lung cancer. Am J Health Syst Pharm 71: 1933-1938.
  16. Finlay MR, Anderton M, Ashton S, Ballard P, Bethel PA, et al. (2014) Discovery of a potent and selective EGFR inhibitor (AZD9291) of both sensitizing and T790M resistance mutations that spares the wild type form of the receptor. J Med Chem 57: 8249-8267.
  17. Peters S, Zimmermann S, Adjei AA (2014) Oral epidermal growth factor receptor tyrosine kinase inhibitors for the treatment of non-small cell lung cancer: comparative pharmacokinetics and drug-drug interactions. Cancer treatment reviews 40: 917-926.
  18. Rexer BN, Ghosh R, Narasanna A, Estrada MV, Chakrabarty A, et al. (2013) Human breast cancer cells harboring a gatekeeper T798M mutation in HER2 overexpress EGFR ligands and are sensitive to dual inhibition of EGFR and HER2. Clinical cancer research : an official journal of the American Association for Cancer Research. 19: 5390-5401.
  19. Wellstein A (2012) ALK receptor activation, ligands and therapeutic targeting in glioblastoma and in other cancers. Front Oncol 2: 192.
  20. Souttou B, Carvalho NB, Raulais D, Vigny M (2001) Activation of anaplastic lymphoma kinase receptor tyrosine kinase induces neuronal differentiation through the mitogen-activated protein kinase pathway. The Journal of biological chemistry 276: 9526-9531.
  21. Shaw AT, Solomon B (2011) Targeting anaplastic lymphoma kinase in lung cancer. Clinical cancer research: an official journal of the American Association for Cancer Research 17: 2081-2086.
  22. Proietti A, Alì G, Pelliccioni S, Lupi C, Sensi E, et al. (2014) Anaplastic lymphoma kinase gene rearrangements in cytological samples of non-small cell lung cancer: comparison with histological assessment. Cancer Cytopathol 122: 445-453.
  23. Choi YL, Soda M, Yamashita Y, Ueno T, Takashima J, et al. (2010) EML4-ALK mutations in lung cancer that confer resistance to ALK inhibitors. N Engl J Med 363: 1734-1739.
  24. Katayama R, Shaw AT, Khan TM, Mino-Kenudson M, Solomon BJ, et al. (2012) Mechanisms of acquired crizotinib resistance in ALK-rearranged lung Cancers. SciTransl Med 4: 120ra17.
  25. Doebele RC, Pilling AB, Aisner DL, Kutateladze TG, Le AT, et al. (2012) Mechanisms of resistance to crizotinib in patients with ALK gene rearranged non-small cell lung cancer. Clin Cancer Res 18: 1472-1482.
  26. Toyokawa G, Inamasu E, Shimamatsu S, Yoshida T, Nosaki K, et al. (2015) Identification of a Novel ALK G1123S Mutation in a Patient with ALK-rearranged Non-small-cell Lung Cancer Exhibiting Resistance to Ceritinib. Journal of thoracic oncology: official publication of the International Association for the Study of Lung Cancer 10: e55-7.
  27. Ignatius SH, Azada M, Hsiang DJ, Herman JM, Kain TS, et al. (2014) Next-generation sequencing reveals a Novel NSCLC ALK F1174V mutation and confirms ALK G1202R mutation confers high-level resistance to alectinib (CH5424802/RO5424802) in ALK-rearranged NSCLC patients who progressed on crizotinib. Journal of thoracic oncology: official publication of the International Association for the Study of Lung Cancer 9: 549-553.
  28. Ou SH, Klempner SJ, Greenbowe JR, Azada M, Schrock AB, et al. (2014) Identification of a novel HIP1-ALK fusion variant in Non-Small-Cell Lung Cancer (NSCLC) and discovery of ALK I1171 (I1171N/S) mutations in two ALK-rearranged NSCLC patients with resistance to Alectinib. Journal of thoracic oncology: official publication of the International Association for the Study of Lung Cancer 9: 1821-1825.
  29. Toyokawa G, Hirai F, Inamasu E, Yoshida T, Nosaki K, et al. (2014) Secondary mutations at I1171 in the ALK gene confer resistance to both Crizotinib and Alectinib. Journal of thoracic oncology: official publication of the International Association for the Study of Lung Cancer 9: e86-87.
  30. Katayama R, Friboulet L, Koike S, Lockerman EL, Khan TM, et al. (2014) Two novel ALK mutations mediate acquired resistance to the next-generation ALK inhibitor alectinib. Clinical cancer research: an official journal of the American Association for Cancer Research 20: 5686-5696.
  31. Friboulet L, Li N, Katayama R, Lee CC, Gainor JF, et al. (2014) The ALK inhibitor ceritinib overcomes crizotinib resistance in non-small cell lung cancer. Cancer Discov 4: 662-673.
  32. Piperdi B, Merla A, Perez-Soler R (2014) Targeting angiogenesis in squamous non-small cell lung cancer. Drugs 74: 403-413.
  33. Di Costanzo F, Mazzoni F, MicolMela M, Antonuzzo L, Checcacci D, et al. (2008) Bevacizumab in non-small cell lung cancer. Drugs 68: 737-746.
  34. Korpanty G, Smyth E, Carney DN (2011) Update on anti-angiogenic therapy in non-small cell lung cancer: Are we making progress? Journal of thoracic disease 3:19-29.
  35. Bottsford-Miller JN, Coleman RL, Sood AK (2012) Resistance and escape from antiangiogenesis therapy: clinical implications and future strategies. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 30: 4026-4034.
  36. Lambrechts D, Lenz HJ, de Haas S, Carmeliet P, Scherer SJ (2013) Markers of response for the antiangiogenic agent bevacizumab. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 31: 1219-1230.
  37. Planchard D, Kim TM, Mazieres J, Quoix E, Riely GJ, et al. (2014) Dabrafenib in patients with braf v600e-mutant advanced non-small cell lung cancer (nsclc): a multicenter, open-label, phase ii trial (BRF113928). Ann Oncol 25.
  38. Rudin CM, Hong K, Streit M (2013) Molecular characterization of acquired resistance to the BRAF inhibitor dabrafenib in a patient with BRAF-mutant non-small-cell lung cancer. Journal of thoracic oncology: official publication of the International Association for the Study of Lung Cancer 8: e41-42