Topic Highlight Open Access
Copyright ©2014 Baishideng Publishing Group Inc. All rights reserved.
World J Clin Oncol. Oct 10, 2014; 5(4): 588-594
Published online Oct 10, 2014. doi: 10.5306/wjco.v5.i4.588
Apoptosis block as a barrier to effective therapy in non small cell lung cancer
Ian Paul, J Mark Jones, Department of Cardiothoracic Surgery, Royal Victoria Hospital, Belfast BT12 6BA, Northern Ireland, United Kingdom
Author contributions: Paul I conceived and constructed the manuscript; Jones JM edited and critically reviewed the manuscript.
Correspondence to: J Mark Jones, MD, FRCSEd(CTh), Department of Cardiothoracic Surgery, Royal Victoria Hospital, Grosvenor Road, Belfast BT12 6BA, Northern Ireland, United Kingdom. mark.jones@belfasttrust.hscni.net
Telephone: +44-28-90636544 Fax: +44-28-90312907
Received: January 10, 2014
Revised: February 28, 2014
Accepted: June 10, 2014
Published online: October 10, 2014

Abstract

Lung cancer, is the most common cause of cancer death in men and second only to breast cancer in women. Currently, the first line therapy of choice is platinum-based combination chemotherapy. A therapeutic plateau has been reached with the prognosis for patients with advanced non-small cell lung cancer (NSCLC) remaining poor. New biomarkers of prognosis as well as new therapies focusing on molecular targets are emerging helping to identify patients who are likely to benefit from therapy. Despite this, drug resistance remains the major cause for treatment failure. In this article we review the role of apoptosis in mediating drug resistance in NSCLC. Better understanding of this fundamental biological process may provide a rationale for overcoming the current therapeutic plateau.

Key Words: Apoptosis, Lung cancer, Adjuvant therapy, Mitochondria, BAX, BAK, BCL2

Core tip: A therapeutic plateau has been reached with the treatment of non-small cell lung cancers with platinum-based combination chemotherapy. New biomarkers of prognosis as well as new therapies focusing on molecular targets are emerging helping to identify patients who are likely to benefit from therapy. These are as yet only available to the minority of patients. Drug resistance remains the major cause for treatment failure. Apoptosis block as a mechanism for drug resistance and potential routes to overcome this will be reviewed.



INTRODUCTION

Lung cancer is the leading cause of cancer death in the United Kingdom accounting for 24% of all male related cancer deaths and 20% in females[1]. Similar trends are seen in the United States with lung cancer representing the most common cause of cancer death in men and women, recently overtaking breast cancer in the latter[2]. In the United States, more than 213000 new cases were diagnosed in 2007 with a total of 1.61 million cases worldwide in 2008[2,3]. The majority of these cancers (75%-80%) are non-small cell lung cancers (NSCLC)[4].

Surgery is the mainstay of treatment for early stage and localised disease (Stage I and II and selected IIIA). Multimodal therapy is the norm for regionally advanced disease in the form of adjuvant chemotherapy. Palliative chemotherapy forms the mainstay of treatment in patients with advanced metastatic disease[5]. Five year survival rates following lung resection for NSCLC are IA-73%, IB-54%, IIA-48%, IIB-38%, IIIA-25%[6]. The majority of patients have advanced disease at the time of diagnosis and therefore are not surgical candidates. This is illustrated by the fact that in the United Kingdom only 14% of patients diagnosed go on to have surgical resection[7]. In those patients that are surgical candidates, more than 50% will develop a recurrence. Adjuvant chemotherapy has been used with limited success to decrease the recurrence rates but this has only yielded a survival benefit of 5%-15%[8].

For patients not suitable for surgery due to advanced disease or those who have suffered recurrence following resection, chemotherapy forms the mainstay of treatment with evidence that platinum based therapies are most effective in the first line setting[9,10]. Despite this, in a recent trial of platinum combination therapies in advanced NSCLC, only 30% of patients showed objective disease response and a significant proportion suffered toxic side-effects such as neutropenia (27%), anaemia (10%), thrombocytopenia (13%), alopecia (21%) and nausea (4%). During the trial, deaths due to study drug toxicity were in the region of 1%[11]. This demonstrates the major problem of drug resistance in NSCLC to standard platinum based therapies and the associated toxicities.

MOLECULAR TARGETED THERAPY

A recent major advance in the management of NSCLC has been the identification of activating mutations in the tyrosine kinase domain of the epidermal growth factor receptor (EGFR)[12]. These mutations, found in 10% of NSCLC specimens is the United States and Europe, are associated with increased sensitivity to EGFR tyrosine kinase inhibitors erlotinib and gefitinib[13]. EGFR mutations are often accompanied by gene amplification[14]. Mutation of the EGFR can lead to signal transduction independent of ligand stimulation, relaying signals to the PI3K-AKT-mTOR pathway involved in cell survival, or to the RAS-RAF-MEK-ERK pathway involved in cell proliferation[15]. The fact that kinase inhibition leads to apoptosis in cells with mutant EGFR supports the notion that these cells are ‘‘addicted’’ to signalling via the mutant proteins[16]. This explains the dramatic response to EGFR tyrosine kinase inhibitors in EGFR mutated NSCLC with associated improvement in survival[17,18].

Molecular profiling in NSCLC has revealed a number of other mutations such as EML4-ALK translocation[19]. This is associated with extreme preclinical sensitivity to respective ALK kinase inhibition in both preclinical and clinical settings[20,21]. Additional molecular subclasses with associated somatic gene alterations have been discovered, predominantly in lung adenocarcinomas and include mutations of BRAF[22], Her2[23] and PIKC3A[24]. The continual discovery of new molecular subclasses represents significant progress in the treatment of NSCLC but they should not necessarily be viewed as a panacea as resistance to these novel targeted therapies are being reported[25]. The above molecular subclasses still only account for less than half all new cases of the disease. The majority of patients still rely on standard combination chemotherapy of platinum based doublet regimens.

In addition to molecular tumour markers, there is increasing interest in developing biomarkers associated with NSCLC. These may be predictive for response to treatment or prognostic. An area of interest in our unit relates to the link between plasma fibrinogen and NSCLC. In early stage disease amenable to surgical resection, raised fibrinogen is associated with increased risk of incomplete resection and correlates with T stage of tumour[26]. More recently, serum fibrinogen has been shown to be an independent predictor for both disease recurrence and overall survival in both resectable and advanced disease[27,28] The mechanism by which fibrinogen acts as a marker is not fully understood. Its role as an extracellular matrix, in promoting angiogenesis and metastasis has been proposed[29-32].

APOPTOSIS AND DRUG RESISTANCE

Cisplatin (cis-diamminedichloroplatinum II) is the most effective platinum based therapy in the first line setting for NSCLC[33]. It forms DNA-platinum covalent adducts resulting in inhibition of DNA replication, suppression of RNA transcription and protein translation, attempted DNA repair as well as disturbance of the cell cycle and the activation of apoptosis[34].

Apoptosis serves as a natural barrier to cancer development[35]. Normally the accumulation of genetic mutations required to drive uncontrolled cell cycle progression and tumorigenesis would result in triggering of apoptosis within a cell[36]. The ability of cancer cells to evade apoptosis is, therefore, an essential hallmark of cancer[37]. Defects in apoptosis, harboured by cancer cells, not only underpin tumorigenesis but also drug resistance[38]. Resistance of NSCLC cells to a diverse range of cytotoxic therapies suggests a defect in apoptosis signalling[39]. Further understanding of the mechanisms by which apoptosis resistance occurs and how this can be overcome will be important in order to administer a more rational approach to anticancer drug design and therapy.

APOPTOSIS

Apoptosis, a genetically encoded programme of cell death, was originally defined based on the morphological features observed as the cell died; nuclear condensation, nuclear and cellular fragmentation, membrane blebbing and phagocytosis of the dying cell in the absence of inflammation[40]. The process of apoptosis is conserved in a wide range of multicellular organisms from worms to humans and plays a key role in normal development and homeostasis. The apoptosis phenotype is produced through the activity of cysteine-aspartic proteases, a family of cysteine proteases termed caspases. A hierarchical cascade of activation occurs which results in apoptosis. Two groups of caspases have been classified; the initiator caspases and the effector caspases.

The initiator caspases, caspase-8, -9 and -10, are activated early in apoptosis signalling and have restricted cleavage targets, limited to self cleavage, the effector caspases and BID (caspase-8). In contrast, the effector caspases, caspase-3, -6 and -7, have hundreds of cleavage sites broadly distributed throughout the cell[41]. The effector caspases are held in the cytosol as inactive dimers. The activating event, catalyzed by the initiator caspases, involves conversion to catalytically active enzymes by cleavage in the linker region between the large and small active subunits. This allows intramolecular rearrangements to form an enzymatically active dimer[42]. Caspase-3 and -7 display highly similar substrate specificity and carry out redundant but essential functions in apoptotic cell death as mouse embryonic fibroblasts lacking both enzymes are resistant to intrinsic and extrinsic apoptotic stimuli[43].

Caspase-8 and -10 are involved in the death receptor signalling pathway whereas caspase-9 is involved in mitochondrial apoptosis.

It is clear caspase activation is a critical step in the execution of apoptosis and is generally a terminal event for the cell. Regulation of the process occurs through the extrinsic, death receptor apoptosis pathway and the intrinsic mitochondrial apoptosis pathway.

EXTRINSIC APOPTOSIS PATHWAY

Apoptosis occurs via the extrinsic apoptosis pathway as a result of signalling through death receptors expressed on the surface of mammalian cells. In the 1970’s it was identified that certain products of lymphocytes and macrophages caused the lysis of certain types of cells, especially tumour cells. This product was termed ‘tumour necrosis factor (TNF)[44,45]. It has been established that TNF has its effect via cell surface receptor binding and activation. At least 18 TNF family ligands and 29 receptors have been identified in humans[46]. In cancer research, interest has grown around the use of TNF-related apoptosis-inducing ligand (TRAIL) and targeting its receptors, members of the TNF superfamily, since the observation that recombinant human (rh) TRAIL induces apoptosis in various tumour cells but not in normal cells[47].

TRAIL activates apoptosis by binding to specific transmembrane receptors TRAIL-R1 (DR4) and TRAIL-R2 (DR5)[48,49]. It can also bind truncated, non-functional receptors TRAIL-R3 and TRAIL-R4 known as decoy receptors (DcR1 and DcR2)[50]. Upon binding TRAIL to the death receptors (DR4, DR5), trimerization of the receptors occurs and a complex is formed termed the death-inducing signalling complex (DISC). This is the key step for subsequent initiator caspase activation to occur.

INTRINSIC APOPTOSIS PATHWAY

The other major apoptosis pathway leading to caspase activation and cell death is the Intrinsic (mitochondrial) apoptosis pathway. As DISC formation is the key step in extrinsic (death receptor) signalling so mitochondrial outer membrane permeabilization (MOMP) is the key requirement for caspase activation and apoptosis via the mitochondrial apoptosis pathway. As the name suggests, this apoptosis pathway is engaged primarily as a result of internal cellular stresses such as DNA damage, ER stress[41].

MOMP during apoptosis is primarily controlled by the BCL2 family of proteins[51]. The pro-apoptotic members, BAX[52] and BAK[53], contain BH1-3 domains. BAX and BAK are often referred to as effector proteins as there is an absolute requirement for their activation at the outer mitochondrial membrane for MOMP to occur[54].

A diverse range of death signals caused by eg DNA damage, growth factor deprivation leads to a shift in the balance of anti- and pro-apoptotic BCL2 family members with signals produced to engage MOMP. Activation of a “multidomain” proapoptotic member, BAX or BAK, is an essential gateway to mitochondrial dysfunction required for cell death in response to these diverse stimuli[54].

Upon receipt of a death signal, BAX translocates to the mitochondrial surface where BAK already resides[55,56]. A detailed sequence of events from BAX translocation to the OMM, subsequent activation and MOMP has been described recently[57]. The requirement for activated BAX and/or BAK for MOMP to occur is clear, the mechanism by which they bring about MOMP is not. The key feature of MOMP in bringing about caspase activation and apoptosis is cytochrome C release. This is clear from evidence that cells lacking cytochrome C fail to activate capases in response to UV irradiation, serum withdrawal, or staurosporine[58]. The primary function of cytochrome C is in oxidative phosphorylation as it is a key component of the electron transport chain and it is found loosely associated with the mitochondrial inner membrane. The other important mitochondrial intermembrane space protein released during MOMP is second mitochondria-derived activator of caspase (SMAC) also termed DIABLO (direct IAP binding protein with low pI). It binds XIAP’s, antagonising their ability to inhibit caspases[59,60].

APOPTOSIS BLOCK IN CANCER

Having outlined how apoptosis proceeds, the mechanisms by which cancer cells evade this process will be explored. Block in mitochondrial apoptosis has been broadly divided into three groups[61]: (1) Class A block occurs when normal generation of proapoptotic activators by aberrant behavior is inhibited. The mechanisms by which aberrant behaviour, such as genomic instability and oncogene activation, generate death signals via BH3-only proteins is as yet poorly understood. This is an area of ongoing study; (2) Class B block occurs when there is a significant loss of the BCL2 family effectors, BAX and BAK; and (3) Class C block occurs when increased expression of an anti-apoptotic BCL2 family protein is present, thereby inhibiting or sequestering pro-apoptotic BH3-only proteins. In this scenario, the cell has generated an appropriate BH3-only death signal but it is inhibited by opposing anti-apoptotic expression. Cells in this state are referred to as “primed for death” and are “addicted” to the overexpression of the anti-apoptotic protein[62].

Apoptosis blocks such as described above may determine the sensitivity of a cancer cell to a wide range of diverse chemotherapy agents and explain frequently observed phenomenon of multidrug resistance[51]. The fact that being primed for death (class C block) is apparently more common in tumours than in normal cells may help explain why chemotherapy is often more toxic to cancers[51].

Having classified apoptosis block in tumours, a new technique for determining what type of block a given cell employs has been developed termed BH3 profiling[61,62]. The basic method involves incubation of mitochondria isolated from tumour cells with a panel of BH3 peptides. By assessing the pattern of response, the type of apoptosis block present can be identified. This may be used in the future to select drugs targeting anti-apoptotic proteins as a strategy for improving efficacy of drug treatments.

EXPRESSION OF BCL2 FAMILY MEMBERS IN NSCLC

Much focus has been placed on the role of BCL2 as a prognostic predictor in NSCLC. Many studies over the last 15-20 years have assessed its expression in many differing solid tumours. Given its function as an antiapoptotic protein, it would be predicted that overexpression would result in a more aggressive and treatment resistant phenotype. The published data is mixed with regard to its role as a prognostic marker.

A meta-analysis from 2003 compiled 28 studies from 1993 to 1999 which report the expression of BCL2 in NSCLC and the prognostic value of its expression in the primary tumour[63]. Immunohistochemistry was used to detect expression in all studies. Of the 28 studies included, 11 concluded Bcl-2 expression was a good prognostic marker, 14 concluded it was not prognostic for survival with only 3 linking Bcl-2 expression to poor prognosis. Having performed the meta-analysis, the authors concluded that Bcl-2 positive tumours had a significantly better survival than those with Bcl-2 negative tumours. Given its function in the apoptotic pathway this would appear a paradoxical conclusion. One theory suggests that loss of Bcl-2 expression correlates with tumour de-differentiation and therefore a more aggressive phenotype[64]. As discussed above, increased expression of BCL2 would likely confer a class C block in apoptosis and as such these tumours would be primed for death perhaps explaining the increased survival. This should be interpreted with caution as a result of heterogenous treatment patients in these studies received. Given the highly complex interplay between Bcl-2 and its other family members in regulating apoptosis, it is not surprising that the study of one anti-apoptotic member yielded such a result. As knowledge about the interplay between Bcl-2 family members improves, other targets or combination of targets may be more relevant to study rather than a single protein in isolation.

The crucial role the proapoptotic multidomain proteins BAX and BAK play in mitochondrial outer membrane permeabilisation warrants further study as prognostic markers. Loss of both proteins would confer a class B apoptosis block. Many studies exist reporting the status of BAX and BAK expression in NSCLC but again only report in isolation and each is considered separately as prognostic markers.

Altered BAX expression is frequently reported in NSCLC[65-68]. None of these studies conclude that altered BAX expression has significant value as a prognostic marker, although none have investigated the expression of BAK together with BAX. Fewer studies report the incidence of altered BAK expression[69,70]. These studies report the incidence of BAK loss at 42% and 59%. They also include data on BAX expression with loss reported in 34% and 47%. Neither study reports the incidence of double loss. Both conclude that no prognostic value is attributed to BAX or BAK expression in NSCLC but again each protein is analysed in isolation. Data from the International Adjuvant Lung Cancer Trial (IALT) suggests a trend toward increasing chemosensitivity with increasing BAX level[71]. The converse effect of BAX negativity was not reported.

STRATEGIES TO OVERCOME MITOCHONDRIAL APOPTOSIS BLOCK

Given the frequent loss of expression of each protein, it is likely a significant portion of patients with NSCLC will have BAX and BAK double loss and given the evidence that this results in a highly apoptosis resistant phenotype it would be important to assess the impact on double loss on both survival and response to standard chemotherapy and radiotherapy in NSCLC.

Given what is known about the mitochondrial apoptosis pathway, class B block in apoptosis is likely to prove resistant to a range of targeted therapies. BH3 mimetics would be predicted to be ineffective due to absence of effector proteins BAX and BAK. Alternative strategies will be required to treat these potentially multidrug resistant cancers. In vitro evidence exists to show that in the absence of BAX and BAK, detachment of hexokinase from mitochondrial VDAC can lead to cytochrome C release and therefore mediate cell death[72]. This can be achieved using either a competitive peptide, or by inhibiting AKT (which in turn regulates hexokinase interaction with VDAC) and prove a rational approach to treating cancers exhibiting Akt activation or an imbalance in the expression of antiapoptotic and proapoptotic members of the Bcl-2 family.

CONCLUSION

NSCLC presents a major health burden worldwide. Platinum based chemotherapy is the mainstay of treatment in the clinic today, although denovo and acquired drug resistance has resulted in a therapeutic plateau since its introduction over 30 years ago. Novel targeted therapies are beginning to emerge that induce apoptosis in certain molecular subclasses. Apoptosis resistance underpins tumorigenesis and drug resistance. Understanding how apoptosis resistance occurs in NSCLC will allow tailoring of therapy and development of novel targets to overcome this problem.

Footnotes

P- Reviewer: Hattori N, Kawai H, Vlachostergios PJ, Zhang YJ S- Editor: Wen LL L- Editor: A E- Editor: Lu YJ

References
1.   Available from: http: //info.cancerresearchuk.org/cancerstats/types/lung/mortality/?a=5441#source1].  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Ramalingam S, Belani C. Systemic chemotherapy for advanced non-small cell lung cancer: recent advances and future directions. Oncologist. 2008;13 Suppl 1:5-13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 201]  [Cited by in F6Publishing: 256]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
3.  Ferlay J, Parkin DM, Steliarova-Foucher E. Estimates of cancer incidence and mortality in Europe in 2008. Eur J Cancer. 2010;46:765-781.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Klastersky J, Awada A. Milestones in the use of chemotherapy for the management of non-small cell lung cancer (NSCLC). Crit Rev Oncol Hematol. 2012;81:49-57.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Dempke WC, Suto T, Reck M. Targeted therapies for non-small cell lung cancer. Lung Cancer. 2010;67:257-274.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 122]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
6.  Lim E, Harris G, Patel A, Adachi I, Edmonds L, Song F. Preoperative versus postoperative chemotherapy in patients with resectable non-small cell lung cancer: systematic review and indirect comparison meta-analysis of randomized trials. J Thorac Oncol. 2009;4:1380-1388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 122]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
7.   Available from: http: //www.ic.nhs.uk/webfiles/Services/NCASP/audits and reports/NHSIC_National_Lung_Cancer_Audit_2010_V1.0.pdf.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Sedrakyan A, Van Der Meulen J, O’Byrne K, Prendiville J, Hill J, Treasure T. Postoperative chemotherapy for non-small cell lung cancer: A systematic review and meta-analysis. J Thorac Cardiovasc Surg. 2004;128:414-419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 95]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
9.  Belani CP, Langer C. First-line chemotherapy for NSCLC: an overview of relevant trials. Lung Cancer. 2002;38 Suppl 4:13-19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 24]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
10.  Sörenson S, Glimelius B, Nygren P. A systematic overview of chemotherapy effects in non-small cell lung cancer. Acta Oncol. 2001;40:327-339.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 47]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
11.  Scagliotti GV, Parikh P, von Pawel J, Biesma B, Vansteenkiste J, Manegold C, Serwatowski P, Gatzemeier U, Digumarti R, Zukin M. Phase III study comparing cisplatin plus gemcitabine with cisplatin plus pemetrexed in chemotherapy-naive patients with advanced-stage non-small-cell lung cancer. J Clin Oncol. 2008;26:3543-3551.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Paez JG, Jänne PA, Lee JC, Tracy S, Greulich H, Gabriel S, Herman P, Kaye FJ, Lindeman N, Boggon TJ. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science. 2004;304:1497-1500.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7278]  [Cited by in F6Publishing: 7326]  [Article Influence: 366.3]  [Reference Citation Analysis (0)]
13.  Pao W, Girard N. New driver mutations in non-small-cell lung cancer. Lancet Oncol. 2011;12:175-180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 822]  [Cited by in F6Publishing: 861]  [Article Influence: 66.2]  [Reference Citation Analysis (0)]
14.  Ladanyi M, Pao W. Lung adenocarcinoma: guiding EGFR-targeted therapy and beyond. Mod Pathol. 2008;21 Suppl 2:S16-S22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 253]  [Cited by in F6Publishing: 278]  [Article Influence: 17.4]  [Reference Citation Analysis (0)]
15.  Pao W, Iafrate AJ, Su Z. Genetically informed lung cancer medicine. J Pathol. 2011;223:230-240.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 50]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
16.  Riely GJ, Politi KA, Miller VA, Pao W. Update on epidermal growth factor receptor mutations in non-small cell lung cancer. Clin Cancer Res. 2006;12:7232-7241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 270]  [Cited by in F6Publishing: 300]  [Article Influence: 17.6]  [Reference Citation Analysis (0)]
17.  Rosell R, Moran T, Queralt C, Porta R, Cardenal F, Camps C, Majem M, Lopez-Vivanco G, Isla D, Provencio M. Screening for epidermal growth factor receptor mutations in lung cancer. N Engl J Med. 2009;361:958-967.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1687]  [Cited by in F6Publishing: 1749]  [Article Influence: 116.6]  [Reference Citation Analysis (0)]
18.  Rosell R, Carcereny E, Gervais R, Vergnenegre A, Massuti B, Felip E, Palmero R, Garcia-Gomez R, Pallares C, Sanchez JM. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2012;13:239-246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3670]  [Cited by in F6Publishing: 4169]  [Article Influence: 347.4]  [Reference Citation Analysis (0)]
19.  Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y, Ishikawa S, Fujiwara S, Watanabe H, Kurashina K, Hatanaka H. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature. 2007;448:561-566.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3816]  [Cited by in F6Publishing: 3914]  [Article Influence: 230.2]  [Reference Citation Analysis (0)]
20.  Bang ELKY, Shaw AT, Camidge DR, Iafrate AJ, Maki RG, Solomon BJ, Ou SJ, Salgia R, Clark JW. Clinical activity of the oral ALK inhibitor PF-02341066 in ALK-positive patients with non-small cell lung cancer (NSCLC). ASCO. 2010;28:2010.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Shaw AT, Kim DW, Nakagawa K, Seto T, Crinó L, Ahn MJ, De Pas T, Besse B, Solomon BJ, Blackhall F. Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med. 2013;368:2385-2394.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2534]  [Cited by in F6Publishing: 2584]  [Article Influence: 234.9]  [Reference Citation Analysis (0)]
22.  Brose MS, Volpe P, Feldman M, Kumar M, Rishi I, Gerrero R, Einhorn E, Herlyn M, Minna J, Nicholson A. BRAF and RAS mutations in human lung cancer and melanoma. Cancer Res. 2002;62:6997-7000.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Cappuzzo F, Bemis L, Varella-Garcia M. HER2 mutation and response to trastuzumab therapy in non-small-cell lung cancer. N Engl J Med. 2006;354:2619-2621.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 176]  [Cited by in F6Publishing: 189]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
24.  Stephens P, Hunter C, Bignell G, Edkins S, Davies H, Teague J, Stevens C, O’Meara S, Smith R, Parker A. Lung cancer: intragenic ERBB2 kinase mutations in tumours. Nature. 2004;431:525-526.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 555]  [Cited by in F6Publishing: 567]  [Article Influence: 28.4]  [Reference Citation Analysis (0)]
25.  Bean J, Riely GJ, Balak M, Marks JL, Ladanyi M, Miller VA, Pao W. Acquired resistance to epidermal growth factor receptor kinase inhibitors associated with a novel T854A mutation in a patient with EGFR-mutant lung adenocarcinoma. Clin Cancer Res. 2008;14:7519-7525.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 214]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
26.  Jones JM, McGonigle NC, McAnespie M, Cran GW, Graham AN. Plasma fibrinogen and serum C-reactive protein are associated with non-small cell lung cancer. Lung Cancer. 2006;53:97-101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 78]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
27.  Zhu JF, Cai L, Zhang XW, Wen YS, Su XD, Rong TH, Zhang LJ. High plasma fibrinogen concentration and platelet count unfavorably impact survival in non-small cell lung cancer patients with brain metastases. Chin J Cancer. 2014;33:96-104.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Sheng L, Luo M, Sun X, Lin N, Mao W, Su D. Serum fibrinogen is an independent prognostic factor in operable nonsmall cell lung cancer. Int J Cancer. 2013;133:2720-2725.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Simpson-Haidaris PJ, Rybarczyk B. Tumors and fibrinogen. The role of fibrinogen as an extracellular matrix protein. Ann N Y Acad Sci. 2001;936:406-425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 173]  [Cited by in F6Publishing: 177]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
30.  Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med. 1986;315:1650-1659.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3033]  [Cited by in F6Publishing: 2976]  [Article Influence: 78.3]  [Reference Citation Analysis (0)]
31.  Biggerstaff JP, Seth NB, Meyer TV, Amirkhosravi A, Francis JL. Fibrin monomer increases platelet adherence to tumor cells in a flowing system: a possible role in metastasis? Thromb Res. 1998;92:S53-S58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 14]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
32.  Palumbo JS, Potter JM, Kaplan LS, Talmage K, Jackson DG, Degen JL. Spontaneous hematogenous and lymphatic metastasis, but not primary tumor growth or angiogenesis, is diminished in fibrinogen-deficient mice. Cancer Res. 2002;62:6966-6972.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Hotta K, Matsuo K, Ueoka H, Kiura K, Tabata M, Tanimoto M. Meta-analysis of randomized clinical trials comparing Cisplatin to Carboplatin in patients with advanced non-small-cell lung cancer. J Clin Oncol. 2004;22:3852-3859.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 288]  [Cited by in F6Publishing: 309]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
34.  Wang G, Reed E, Li QQ. Molecular basis of cellular response to cisplatin chemotherapy in non-small cell lung cancer (Review). Oncol Rep. 2004;12:955-965.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Adams JM, Cory S. The Bcl-2 apoptotic switch in cancer development and therapy. Oncogene. 2007;26:1324-1337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1458]  [Cited by in F6Publishing: 1525]  [Article Influence: 89.7]  [Reference Citation Analysis (0)]
36.  Lowe SW, Cepero E, Evan G. Intrinsic tumour suppression. Nature. 2004;432:307-315.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 936]  [Cited by in F6Publishing: 928]  [Article Influence: 46.4]  [Reference Citation Analysis (0)]
37.  Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19834]  [Cited by in F6Publishing: 18741]  [Article Influence: 780.9]  [Reference Citation Analysis (0)]
38.  Johnstone RW, Ruefli AA, Lowe SW. Apoptosis: a link between cancer genetics and chemotherapy. Cell. 2002;108:153-164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1680]  [Cited by in F6Publishing: 1660]  [Article Influence: 75.5]  [Reference Citation Analysis (0)]
39.  Joseph B, Lewensohn R, Zhivotovsky B. Role of apoptosis in the response of lung carcinomas to anti-cancer treatment. Ann N Y Acad Sci. 2000;926:204-216.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 34]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
40.  Wyllie AH, Kerr JF, Currie AR. Cell death: the significance of apoptosis. Int Rev Cytol. 1980;68:251-306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4980]  [Cited by in F6Publishing: 4820]  [Article Influence: 109.5]  [Reference Citation Analysis (0)]
41.  Tait SW, Green DR. Mitochondria and cell death: outer membrane permeabilization and beyond. Nat Rev Mol Cell Biol. 2010;11:621-632.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1714]  [Cited by in F6Publishing: 1819]  [Article Influence: 129.9]  [Reference Citation Analysis (0)]
42.  Chai J, Wu Q, Shiozaki E, Srinivasula SM, Alnemri ES, Shi Y. Crystal structure of a procaspase-7 zymogen: mechanisms of activation and substrate binding. Cell. 2001;107:399-407.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 189]  [Cited by in F6Publishing: 177]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
43.  Fuentes-Prior P, Salvesen GS. The protein structures that shape caspase activity, specificity, activation and inhibition. Biochem J. 2004;384:201-232.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 653]  [Cited by in F6Publishing: 609]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
44.  Granger GA, Shacks SJ, Williams TW, Kolb WP. Lymphocyte in vitro cytotoxicity: specific release of lymphotoxin-like materials from tuberculin-sensitive lymphoid cells. Nature. 1969;221:1155-1157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 81]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
45.  Carswell EA, Old LJ, Kassel RL, Green S, Fiore N, Williamson B. An endotoxin-induced serum factor that causes necrosis of tumors. Proc Natl Acad Sci USA. 1975;72:3666-3670.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2760]  [Cited by in F6Publishing: 2795]  [Article Influence: 57.0]  [Reference Citation Analysis (0)]
46.  Zhang G. Tumor necrosis factor family ligand-receptor binding. Curr Opin Struct Biol. 2004;14:154-160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 55]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
47.  Walczak H, Miller RE, Ariail K, Gliniak B, Griffith TS, Kubin M, Chin W, Jones J, Woodward A, Le T. Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo. Nat Med. 1999;5:157-163.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1924]  [Cited by in F6Publishing: 1897]  [Article Influence: 75.9]  [Reference Citation Analysis (0)]
48.  Pan G, O’Rourke K, Chinnaiyan AM, Gentz R, Ebner R, Ni J, Dixit VM. The receptor for the cytotoxic ligand TRAIL. Science. 1997;276:111-113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1333]  [Cited by in F6Publishing: 1332]  [Article Influence: 49.3]  [Reference Citation Analysis (0)]
49.  Sheridan JP, Marsters SA, Pitti RM, Gurney A, Skubatch M, Baldwin D, Ramakrishnan L, Gray CL, Baker K, Wood WI. Control of TRAIL-induced apoptosis by a family of signaling and decoy receptors. Science. 1997;277:818-821.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1275]  [Cited by in F6Publishing: 1278]  [Article Influence: 47.3]  [Reference Citation Analysis (0)]
50.  Stegehuis JH, de Wilt LH, de Vries EG, Groen HJ, de Jong S, Kruyt FA. TRAIL receptor targeting therapies for non-small cell lung cancer: current status and perspectives. Drug Resist Updat. 2010;13:2-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 63]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
51.  Letai AG. Diagnosing and exploiting cancer’s addiction to blocks in apoptosis. Nat Rev Cancer. 2008;8:121-132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 437]  [Cited by in F6Publishing: 418]  [Article Influence: 26.1]  [Reference Citation Analysis (0)]
52.  Oltvai ZN, Milliman CL, Korsmeyer SJ. Bcl-2 heterodimerizes in vivo with a conserved homolog, Bax, that accelerates programmed cell death. Cell. 1993;74:609-619.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4399]  [Cited by in F6Publishing: 4433]  [Article Influence: 143.0]  [Reference Citation Analysis (0)]
53.  Chittenden T, Harrington EA, O’Connor R, Flemington C, Lutz RJ, Evan GI, Guild BC. Induction of apoptosis by the Bcl-2 homologue Bak. Nature. 1995;374:733-736.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 541]  [Cited by in F6Publishing: 575]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
54.  Wei MC, Zong WX, Cheng EH, Lindsten T, Panoutsakopoulou V, Ross AJ, Roth KA, MacGregor GR, Thompson CB, Korsmeyer SJ. Proapoptotic BAX and BAK: a requisite gateway to mitochondrial dysfunction and death. Science. 2001;292:727-730.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2959]  [Cited by in F6Publishing: 2983]  [Article Influence: 129.7]  [Reference Citation Analysis (0)]
55.  Gao CF, Ren S, Zhang L, Nakajima T, Ichinose S, Hara T, Koike K, Tsuchida N. Caspase-dependent cytosolic release of cytochrome c and membrane translocation of Bax in p53-induced apoptosis. Exp Cell Res. 2001;265:145-151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 90]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
56.  De Giorgi F, Lartigue L, Bauer MK, Schubert A, Grimm S, Hanson GT, Remington SJ, Youle RJ, Ichas F. The permeability transition pore signals apoptosis by directing Bax translocation and multimerization. FASEB J. 2002;16:607-609.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Lovell JF, Billen LP, Bindner S, Shamas-Din A, Fradin C, Leber B, Andrews DW. Membrane binding by tBid initiates an ordered series of events culminating in membrane permeabilization by Bax. Cell. 2008;135:1074-1084.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 438]  [Cited by in F6Publishing: 444]  [Article Influence: 27.8]  [Reference Citation Analysis (0)]
58.  Li K, Li Y, Shelton JM, Richardson JA, Spencer E, Chen ZJ, Wang X, Williams RS. Cytochrome c deficiency causes embryonic lethality and attenuates stress-induced apoptosis. Cell. 2000;101:389-399.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 391]  [Cited by in F6Publishing: 395]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
59.  Verhagen AM, Ekert PG, Pakusch M, Silke J, Connolly LM, Reid GE, Moritz RL, Simpson RJ, Vaux DL. Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins. Cell. 2000;102:43-53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1742]  [Cited by in F6Publishing: 1652]  [Article Influence: 68.8]  [Reference Citation Analysis (0)]
60.  Du C, Fang M, Li Y, Li L, Wang X. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell. 2000;102:33-42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2467]  [Cited by in F6Publishing: 2390]  [Article Influence: 99.6]  [Reference Citation Analysis (0)]
61.  Deng J, Carlson N, Takeyama K, Dal Cin P, Shipp M, Letai A. BH3 profiling identifies three distinct classes of apoptotic blocks to predict response to ABT-737 and conventional chemotherapeutic agents. Cancer Cell. 2007;12:171-185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 402]  [Cited by in F6Publishing: 397]  [Article Influence: 23.4]  [Reference Citation Analysis (0)]
62.  Certo M, Del Gaizo Moore V, Nishino M, Wei G, Korsmeyer S, Armstrong SA, Letai A. Mitochondria primed by death signals determine cellular addiction to antiapoptotic BCL-2 family members. Cancer Cell. 2006;9:351-365.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 965]  [Cited by in F6Publishing: 958]  [Article Influence: 53.2]  [Reference Citation Analysis (0)]
63.  Martin B, Paesmans M, Berghmans T, Branle F, Ghisdal L, Mascaux C, Meert AP, Steels E, Vallot F, Verdebout JM. Role of Bcl-2 as a prognostic factor for survival in lung cancer: a systematic review of the literature with meta-analysis. Br J Cancer. 2003;89:55-64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 122]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
64.  Daniel JC, Smythe WR. The role of Bcl-2 family members in non-small cell lung cancer. Semin Thorac Cardiovasc Surg. 2004;16:19-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
65.  Apolinario RM, van der Valk P, de Jong JS, Deville W, van Ark-Otte J, Dingemans AM, van Mourik JC, Postmus PE, Pinedo HM, Giaccone G. Prognostic value of the expression of p53, bcl-2, and bax oncoproteins, and neovascularization in patients with radically resected non-small-cell lung cancer. J Clin Oncol. 1997;15:2456-2466.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Groeger AM, Esposito V, De Luca A, Cassandro R, Tonini G, Ambrogi V, Baldi F, Goldfarb R, Mineo TC, Baldi A. Prognostic value of immunohistochemical expression of p53, bax, Bcl-2 and Bcl-xL in resected non-small-cell lung cancers. Histopathology. 2004;44:54-63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 68]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
67.  Shabnam MS, Srinivasan R, Wali A, Majumdar S, Joshi K, Behera D. Expression of p53 protein and the apoptotic regulatory molecules Bcl-2, Bcl-XL, and Bax in locally advanced squamous cell carcinoma of the lung. Lung Cancer. 2004;45:181-188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 38]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
68.  Yaren A, Oztop I, Kargi A, Ulukus C, Onen A, Sanli A, Binicier O, Yilmaz U, Alakavuklar M. Bax, bcl-2 and c-kit expression in non-small-cell lung cancer and their effects on prognosis. Int J Clin Pract. 2006;60:675-682.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 28]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
69.  Berrieman HK, Smith L, O’Kane SL, Campbell A, Lind MJ, Cawkwell L. The expression of Bcl-2 family proteins differs between nonsmall cell lung carcinoma subtypes. Cancer. 2005;103:1415-1419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
70.  Borner MM, Brousset P, Pfanner-Meyer B, Bacchi M, Vonlanthen S, Hotz MA, Altermatt HJ, Schlaifer D, Reed JC, Betticher DC. Expression of apoptosis regulatory proteins of the Bcl-2 family and p53 in primary resected non-small-cell lung cancer. Br J Cancer. 1999;79:952-958.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 69]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
71.  Brambilla E, Dunant A, Filipits M, Lantuejoul S, Tarayre M, David-Boudet L, Pirker R, Soria J, Pignon J, Le Chevalier T. O-021 Prognostic and predictive role of alterations of the P53-bax-bcl2pathway of apoptosis in the IALT (International Adjuvant Lung Cancer Trial). Lung cancer (Amsterdam, Netherlands). 2005;49:S10-S11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
72.  Majewski N, Nogueira V, Bhaskar P, Coy PE, Skeen JE, Gottlob K, Chandel NS, Thompson CB, Robey RB, Hay N. Hexokinase-mitochondria interaction mediated by Akt is required to inhibit apoptosis in the presence or absence of Bax and Bak. Mol Cell. 2004;16:819-830.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 476]  [Cited by in F6Publishing: 485]  [Article Influence: 24.3]  [Reference Citation Analysis (0)]