Skip to main content
Genes & Cancer logoLink to Genes & Cancer
editorial
. 2010 Dec;1(12):1200–1210. doi: 10.1177/1947601911407324

The Genomics of Lung Adenocarcinoma

Opportunities for Targeted Therapies

Heidi Greulich 1,2,
Editors: Irwin H Gelman, Marius Sudol
PMCID: PMC3092285  PMID: 21779443

Abstract

Standard cytotoxic chemotherapy is effective for some cancers, but for many others, available treatments offer only a limited survival benefit. Lung adenocarcinoma is one such cancer, responsible for approximately half of lung cancer deaths each year. Development of targeted therapies is thought to hold the most promise for successfully treating this disease, but a targeted approach is dependent on understanding the genomic state of the tumor cells. Exon-directed sequencing of large numbers of lung adenocarcinoma tumor samples has provided an initial low-resolution image of the somatic mutation profile of these tumors. Such cancer sequencing studies have confirmed the high frequency of TP53 and KRAS mutations in lung adenocarcinoma, have found inactivating mutations in known tumor suppressor genes not previously associated with lung adenocarcinoma, and have identified oncogenic mutations of EGFR upon which the first targeted therapy for lung adenocarcinoma patients was based. Additional candidate oncogenes await functional validation. It is anticipated that upcoming whole-exome and whole-genome lung adenocarcinoma sequencing experiments will reveal a more detailed landscape of somatic mutations that can be exploited for therapeutic purposes.

Keywords: lung adenocarcinoma, EGFR, cancer sequencing, targeted therapy

Introduction

Lung cancer is the leading cause of cancer death in the United States and worldwide, accounting for over 150,000 deaths annually in the United States alone.1 The overall 5-year survival rate for lung cancer is only 16%, largely driven by the high frequency of late diagnosis, resulting in nonresectable tumors.1 Lung cancer can be histologically subclassified into 4 major categories: lung adenocarcinoma, squamous cell carcinoma, and large cell carcinoma, comprising non–small cell lung cancer (NSCLC), and small cell carcinoma of the lung.2 Lung adenocarcinoma, an epithelial cancer of glandular origin, is the most prevalent of these lung cancer diagnoses, including in never-smokers.3

The abysmal survival rate for lung adenocarcinoma reflects the inadequacy of traditional cytotoxic chemotherapy for this disease; therapies targeted to tumor cell vulnerabilities instead hold the most promise for the future. Somatic mutations that activate oncogenes frequently result in tumor cell dependency on the altered oncogene products,4,5 a property exploited by the prototypical targeted therapy, imatinib mesylate. Imatinib mesylate inhibits the Bcr-Abl fusion protein, resulting from a recurrent translocation in chronic myelogenous leukemia.6 Imatinib additionally inhibits activated forms of the related tyrosine kinases KIT and PDGFRA and has been successfully used in gastrointestinal stromal tumors harboring mutations in these genes.7 The identification of recurring oncogenic lesions in lung adenocarcinoma upon which the tumor cell depends for survival may therefore lead to novel lung cancer therapies.

A large-scale exon-directed sequencing experiment, the Tumor Sequencing Project (TSP), was undertaken in order to begin to address the question of recurring somatic mutations in lung adenocarcinoma. In this experiment, all coding exons of 623 cancer-related genes were sequenced in 188 tumor/normal DNA pairs, resulting in the identification of 1,013 nonsynonymous somatic mutations.8 Statistical analysis indicated that 26 genes were mutated at a rate significantly higher than the background mutation rate, indicative of positive selection (Fig. 1). These 26 significantly mutated genes included several well-characterized oncogenes and tumor suppressor genes already known to be involved in lung cancer, KRAS, TP53, STK11, EGFR, and CDKN2A. In addition, a number of significantly mutated genes not previously reported in lung adenocarcinoma were identified, including known tumor suppressor genes and several tyrosine kinase genes that represent candidate oncogenes pending functional validation.

Figure 1.

Figure 1.

Significantly mutated genes from the lung adenocarcinoma Tumor Sequencing Project. Adapted from Ding et al.8

Here, I describe the state of knowledge of the genomics of lung adenocarcinoma as advanced by the TSP experiment with special attention to therapeutic implications. The upcoming wave of whole-exome and whole-genome lung adenocarcinoma sequencing results, facilitated by next-generation sequencing technologies, will likely revolutionize our understanding of the genomics of this disease once more.

Mutually Exclusive Oncogenic Alterations

Somatic alterations of 5 lung adenocarcinoma oncogenes, KRAS, EGFR, ALK, ERBB2, and BRAF, are interestingly mutually exclusive and are represented in over 50% of lung adenocarcinomas.9,10 In fact, patients with mutations in these 5 genes may account for up to 90% of Asian never-smokers with the disease.11 The ability to therapeutically inhibit the functions of these 5 altered genes would therefore represent significant progress in the battle against lung cancer.

KRAS

Mutations in KRAS, the most frequently mutated oncogene in lung adenocarcinoma described to date, have been known for some time.12,13 KRAS encodes a low molecular weight GTPase that signals through RAF and ERK when GTP bound.14,15 Similar to KRAS mutations found in other tumor types, mutations that replace Gly 12 with any one of several other amino acids are especially common, with substitutions at Gly 13 and Gln 61 also observed, at a combined frequency of 32%.8 These mutations are activating and oncogenic, causing a reduction in GTPase activity and an increase in GTP-bound protein, resulting in increased mitogenic signaling through RAF.12,14,15

Despite the high frequency of KRAS mutations in lung adenocarcinoma and other cancers, it has proven difficult to exploit mutant KRAS as a therapeutic target. Early efforts were aimed at blocking C-terminal farnesylation, a posttranslational modification required for protein activity.16 Phase III clinical trials of farnesyl transferase inhibitors in solid tumors did not show any statistically significant overall survival benefit, possibly because of the alternate KRAS prenylation activity of geranylgeranyl transferase I, resulting in continued membrane association in the presence of farnesyl transferase inhibitors.16,17

Inhibition of downstream signaling proteins RAF and MEK might also be expected to inhibit growth of tumors cells harboring KRAS mutations, but this approach has been largely unsuccessful as well. Although a combination of PI3K and MEK inhibition can reverse lung adenocarcinomas in transgenic mice driven by KRAS G12D,18 phase II trials of MEK inhibitors as single agents in unselected NSCLC patients have shown a lack of efficacy thus far.19-21 Treatment with sorafenib, a small molecule inhibitor of BRAF and CRAF and several other kinases, resulted in stable disease for 59% of unselected NSCLC patients in a phase II trial, but no responses were observed.22 Moreover, preclinical studies demonstrated that treatment of KRAS mutant cells with a specific BRAF inhibitor paradoxically activated the RAF-MEK-ERK pathway in a CRAF-dependent manner, indicating that BRAF inhibitors are not suitable for use in tumor cells harboring KRAS mutations.23-25

One current area of active research in targeting lung adenocarcinoma cells harboring KRAS mutations involves a synthetic lethal approach,26 whereby inhibition of a second protein causes cell death only in KRAS mutant cells. Interestingly, several RNA-interference synthetic lethal screens have recently been completed in KRAS mutant and wild-type cell lines, identifying the kinases STK33, TBK1, and PLK1 as possible synthetic lethal therapeutic targets.27-29 Additional experiments in tumor cell lines dependent on mutant KRAS for survival or mouse models of lung cancer driven by mutant KRAS pinpointed inhibition or knockdown of NFκB, CDK4, SYK, integrin β6, and RON as synthetic lethal with KRAS mutation.30-32 Whether any of these synthetic lethal interactions translate to a lung cancer therapy remains to be determined.

EGFR

Recurring mutations of the epidermal growth factor receptor (EGFR) tyrosine kinase were first reported in lung adenocarcinoma in 2004 in about 10% of Western patients and over 40% of East Asian patients,33-35 although the biology of this ethnic disparity remains unclear. Mutations were initially identified in 3 kinase domain exons, encoding G719S or G719C in exon 18, small in-frame deletions in exon 19, and L858R or L861Q in exon 21. The observed mutations were determined to be constitutively activating and oncogenic36 and importantly correlated with patient response to gefitinib and erlotinib, small molecule inhibitors of EGFR.33-35 By contrast, oncogenic small in-frame insertions of exon 20 were subsequently discovered in lung adenocarcinoma patients37-39; these EGFR mutants were not sensitive to gefitinib or erlotinib and thus comprised a class of primary resistance mutations in lung adenocarcinoma.36,40

There was some early controversy regarding whether EGFR mutations were truly predictive of gefitinib and erlotinib response, possibly in part because of the confounding effect of the difficulty of somatic mutation detection in stromally contaminated tumors as well as the shortage of evaluable tissue in some trials.41 However, a recent series of phase III clinical trials in Asian patients confirmed a survival benefit of gefitinib over standard chemotherapy as a first-line agent for lung cancer patients who harbor EGFR mutations.42-44 Mutant EGFR is thus a proven therapeutic target in lung adenocarcinoma.

Although patients harboring EGFR mutations in exons 18, 19, and 21 respond well to gefitinib and erlotinib, the response is not durable, and patients relapse after about a year of treatment.41 The most frequent mechanism by which patients develop resistance to gefitinib or erlotinib treatment is acquisition of a second-site resistance mutation in exon 20 of EGFR, encoding T790M, which occurs in about 50% of relapsed patients.45,46 This mutant is analogous to the ABL T315I “gatekeeper” residue substitution that occurs in chronic myeloid leukemia patients in blast crisis phase following an initial response to imatinib treatment.47 The EGFR T790M mutation in particular has also been shown to decrease EGFR affinity for gefitinib in the context of L858R mutation via increased affinity for ATP.48 Interestingly, rare germline mutations encoding EGFR T790M appear to cause inherited susceptibility to lung cancer, often accompanied by activating somatic mutations in EGFR exons 18, 19, and 21.49,50

Gefitinib and erlotinib are thus ineffective against the T790M acquired resistance mutation. However, a second generation of irreversible EGFR inhibitors that covalently modify the protein has recently been developed. Preclinical activity of several of these compounds in L858R-T790M model systems looked promising, especially in combination with rapamycin,51,52 but clinical benefit has yet to be demonstrated. The recent discovery of an anilinopyrimidine-based small molecule that preferentially binds and inhibits EGFR T790M over wild-type has also generated much excitement.53

A second mechanism for the development of resistance to gefitinib, amplification of the receptor tyrosine kinase MET, has been identified in approximately 20% of patients54 but can pre-exist prior to treatment and is not mutually exclusive with T790M mutation.55,56 Resistant cells harboring MET amplification maintained upregulated PI3K signaling in an ERBB3-dependent manner even in the presence of gefitinib.54 In vitro studies indicate that treatment with a combination of gefitinib and a MET inhibitor may circumvent resistance to gefitinib mediated by MET amplification, with the caveat that additional alterations resistant to the combination of both inhibitors, such as MET Y1230H, may also arise.54,57

ALK

Translocations between the receptor tyrosine kinase gene ALK and echinoderm microtubule-associated protein 4, EML4, resulting in the fusion protein EML4-ALK, were described in lung adenocarcinoma in 2007.58 Although originally reported in 7% of NSCLC patient samples tested, the actual frequency may be closer to 4%. NIH-3T3 cells expressing the EML4-ALK variant formed tumors when injected into nude mice, confirming the oncogenic nature of the translocation.58

Because recurring NPM-ALK translocations had already been described in anaplastic large cell lymphoma,59 efforts were under way to develop ALK inhibitors for this disease, facilitating rapid testing of ALK inhibitors in lung adenocarcinoma preclinical models and clinical trials. Although the ALK inhibitor TAE684 was cytotoxic in only 1 of 3 lung adenocarcinoma cell lines harboring an EML4-ALK translocation, the same small molecule efficiently caused tumor regression in transgenic mouse models of EML4-ALK–driven disease.60,61 Importantly, data from an early clinical trial of crizotinib, a dual ALK and MET inhibitor, in NSCLC patients with EML4-ALK translocations look promising.62

Similar to EGFR mutant lung adenocarcinoma patients treated with gefitinib or erlotinib, patients who develop resistance to crizotinib treatment have been identified. Reported acquired resistance alleles of EML4-ALK encode ALK C1156Y, L1196M, and F1174L63,64; interestingly, ALK F1174L was also found to be a driver oncoprotein in neuroblastoma patients naïve of ALK inhibitor treatment.65-68

ERBB2

Somatic mutations of ERBB2 in lung adenocarcinoma were first described in the same year as the EGFR mutations, albeit at lower frequency, approximately 2% to 4%.69,70 These mutations are typically small in-frame insertions in exon 20 of the kinase domain, analogous to the primary resistance mutations of EGFR in the paralogous exon 20. ERBB2 is a receptor tyrosine kinase that does not bind any known ligand but homodimerizes or heterodimerizes with the highly related EGFR and other members of the ERBB family, ERBB3 and ERBB4, to activate downstream signaling pathways.71 These mutations are activating and oncogenic in cell-based transformation assays and respond in vitro to the irreversible inhibitors of EGFR that also bind and inhibit ERBB2.51,72-74 Again, whether these inhibitors are clinically effective against kinase domain mutants of ERBB2 found in lung adenocarcinoma remains to be demonstrated. The therapeutic antibody trastuzumab, developed against the wild-type receptor for use in wild-type ERBB2-amplified breast cancer, does not look promising in preclinical models of mutant ERBB2.74-76

Oncogenic and drug-sensitive mutations of the extracellular domain of EGFR have been described in glioblastoma,77,78 raising the possibility that extracellular domain mutations of ERBB2 may also be found in cancer patients. In fact, a mutation encoding ERBB2 S310F was reported in the lung adenocarcinoma TSP.8 This mutation, although not frequent in lung adenocarcinoma, has been found in other cancers as well and is oncogenic and sensitive to irreversible inhibitors of EGFR/ERBB2 in vitro (H. Greulich, unpublished data), raising the necessity of looking beyond the kinase domain of ERBB2 for clinically relevant activating somatic mutations.

BRAF

Mutations of the serine/threonine kinase gene BRAF have been found at low frequency, about 2%, in lung adenocarcinoma. These mutations, first reported in 2002,79,80 tend to occur in exons 11 and 15 of the kinase domain; however, the V600E mutations frequently found in melanoma and other cancers are rare in lung adenocarcinoma. Although the precise role of these mutations in the development of lung adenocarcinoma remains somewhat enigmatic, there is evidence of gain of function for at least some of the observed alleles.81

Although V600E mutant melanoma has recently been successfully targeted with an inhibitor of BRAF, PLX4032, this inhibitor does not have activity against other BRAF mutants, including those more commonly found in lung adenocarcinoma.82,83 Testing of other BRAF inhibitors in BRAF mutant lung cancer has not been reported. A more promising therapeutic avenue may be MEK inhibition, which inhibits growth of lung adenocarcinoma cell lines harboring BRAF mutations.84 Several such agents have failed to show efficacy in unselected NSCLC patients,19-21 but given the low frequency of BRAF mutation in lung adenocarcinoma, a trial targeting only lung cancer patients with BRAF mutations may be required to uncover any possible therapeutic effect.

Nonmutually Exclusive Oncogenic Alterations

Two other oncogenes, NRAS and PIK3CA, exhibit recurring mutations in lung adenocarcinoma but are not mutually exclusive with the 5 described above. These 2 lung cancer genes will be discussed below along with rare known oncogenic mutations in other genes uncovered by the lung adenocarcinoma TSP and other sequencing efforts.

PIK3CA

A somatic mutation of PIK3CA, encoding the p110α catalytic subunit of phosphatidylinositol 3-kinase (PI3K), was first reported in lung cancer in 2004, along with similar mutations at a much higher frequency in colorectal carcinoma.85 In light of subsequent reports, the overall mutation frequency appears to be 1% to 2%, and mutations cluster in the helical and kinase domains as is the case for PIK3CA mutations in other cancers.86-88 Many of the observed mutations have been shown to be activating and oncogenic in transformation assays and to increase invasiveness in xenograft models,89,90 and PI3K inhibition did reverse lung tumorigenesis in transgenic mouse models driven by PIK3CA H1047R,18 but RNA interference and inhibitor experiments have not yet demonstrated a convincing response in adenocarcinoma cell lines. This indicates that inhibition of PI3K may not be sufficient for tumor therapy, even in patients harboring activating mutations, possibly because of the coexpression of additional mutationally activated oncogenes.

NRAS

Like KRAS, NRAS encodes a low molecular weight GTPase that is similarly C-terminally farnesylated and, when in the GTP-bound state, binds and activates Raf.14-16 Isolated reports of NRAS mutations at codons 12 or 61 occur in the literature as early as 1991,12,91 but it was the lung adenocarcinoma TSP that first detected the significance of a recurring mutation, Q61L, in a single experiment.8 It is difficult to accurately calculate the mutation rate with so few observations, but it is likely between 1% to 2%. The NRAS Q61L mutation is oncogenic12; however, little has been done with regard to validating mutant NRAS as a therapeutic target in lung adenocarcinoma.

CTNNB1

In the APC pathway frequently inactivated in colorectal carcinoma, the APC-AXIN-GSK3β tumor suppressor complex acts to phosphorylate β-catenin and target it for ubiquitin-mediated degradation.92,93 Mutations of the gene encoding β-catenin, CTNNB1, are frequent in colorectal carcinoma and endometrial carcinoma and tend to impact or even eliminate APC-dependent serine and threonine phosphorylation sites, resulting in oncogenic stabilization of β-catenin.94,95 Recurring somatic mutations encoding CTNNB1 G34E, S37C, and S37F were initially described in lung adenocarcinoma in 2001 and confirmed in the TSP experiment.8,96 Although the number of observations is still too low to define the overall mutation frequency in lung adenocarcinoma, the current best estimate is between 1% to 4%. The observed mutation profile mirrors the previously described pattern of mutations in endometrial carcinoma rather than colorectal carcinomas, also characterized by a high frequency of microsatellite instability; the reasons for this remain unclear.94

Exploratory preclinical experiments inhibiting β-catenin signaling in colorectal carcinoma with RNA interference against CTNNB1, KRAS, and transcription factor ITF2 or small molecules that stabilize the APC-AXIN-GSK3β destruction complex look promising,97,98 but these approaches may not easily translate to lung adenocarcinoma therapies for patients harboring stabilizing mutations of CTNNB1. Thus, no real progress has been made in targeting oncogenic mutant forms of CTNNB1 in lung cancer.

Other Rare Activating Mutations

Rare activating mutations of additional known oncogenes have also been detected in lung cancer. Because they have been observed so infrequently, it is not clear whether these mutated genes would have any value as therapeutic targets in lung adenocarcinoma, unless the appropriate inhibitors were developed due to indications of utility in a different tumor type. For example, a mutation of the serine/threonine kinase gene AKT1, which acts downstream of PIK3CA to promote cell proliferation, motility, and viability, was identified in the lung adenocarcinoma TSP experiment.8,99 This mutation, E17K, was previously found in 8% of breast cancer samples, 6% of colorectal cancer samples, and 5% of bladder cancer samples and was demonstrated to be activating and oncogenic.100,101 Intriguingly, AKT1 E17K has also been reported in 2 lung squamous cell carcinomas.102 It is possible that the development of inhibitors for activated AKT in other tumor types could benefit the few lung adenocarcinoma patients who express the E17K substitution as well.

A somatic activating mutation of the dual-specificity kinase MEK1, encoding K57N, has also been reported in 2 lung adenocarcinoma samples.103 MEK1 functions downstream of RAS and RAF proteins to activate ERK1 and ERK2. Unlike expression of wild-type MEK1, expression of MEK1 K57N supports IL-3–independent proliferation of Ba/F3 cells, indicating that the somatic allele encoding K57N is oncogenic.103 Although MEK inhibitors did not show efficacy in unselected NSCLC patients in phase II clinical trials, it possible that such inhibitors might yet be effective in the small population of lung adenocarcinoma patients who harbor activating MEK1 mutations.19-21

The PTPN11 gene, which encodes the nonreceptor tyrosine phosphatase SHP2, has pleiotropic effects in the cell. SHP2 enhances RAS-ERK signaling and, under certain circumstances, can also affect PI3K-AKT signaling and RHO activity; however, the precise mechanism of these effects is not completely understood.104 Activating somatic mutations of PTPN11 have been reported in several childhood hematopoietic cancers105 and more recently in lung adenocarcinoma.8,106-108 There is some in vitro evidence that a combination of MEK inhibition and inhibition of mTOR, a kinase downstream of PI3K and AKT, might be effective against tumor cells harboring activating mutations of PTPN11.109

It is difficult to judge the importance of these rare activating mutations without a better understanding of their frequency in lung adenocarcinoma patients. The TSP experiment, which included 188 samples, was insufficiently powered to reliably detect genes mutated in less than 5% of samples. Sequencing of larger numbers of samples will likely permit a more accurate estimation of the frequency of these mutations in lung cancer patients. Even at 1%, these rare mutations could occur in thousands of patients, justifying efforts in targeted therapies.

Significantly Mutated Tumor Suppressor Genes

Inactivation of tumor suppressor genes also plays a role in the development of lung adenocarcinoma. Although the design of targeted therapies for tumor suppressor genes is not as straightforward as for oncogenes, approaches such as targeting activated genes downstream of an inactivated tumor suppressor gene or identification of synthetic lethal interactions are being examined. Several tumor suppressors were significantly mutated in the TSP experiment (Fig. 1), including those already known to be involved in lung adenocarcinoma, such as TP53, STK11, and CDKN2A, and those not well characterized in lung adenocarcinoma, including NF1, ATM, APC, and RB1. Interestingly, many of these tumor suppressor genes lie in the same pathways as the mutated oncogenes described above.

TP53

TP53 is the most frequently mutated gene in lung adenocarcinoma, with somatic mutations found in close to 70% of patient samples (Fig. 1). Its protein product, p53, activates transcriptional programs that induce cell cycle arrest, apoptosis, or senescence in response to diverse cellular stresses.110 Recurring mutations of TP53 in lung adenocarcinoma were described in 1989 and include missense mutations, frameshift insertions and deletions, splice site mutations, and nonsense mutations.8,111,112 These mutations can result in simple loss of protein function, dominant negative activity by virtue of dimerization with wild-type p53, and even neomorphic gain of function, consistent with oncoprotein activity.113 Indicating a possible essential function, homozygous deletions of TP53 are rare in cancer.114,115 However, MDM2, an oncogene encoding an E3 ubiquitin ligase that targets p53 for degradation, is a target of focal amplification in lung adenocarcinoma.115,116

Several approaches have been taken to targeting mutant p53.116,117 Gene therapy with adenovirally delivered wild-type TP53 was approved for the treatment of head and neck cancer in China (Gendicine, Shenzhen SiBiono GeneTech, Shenzhen, China)118 and is in phase III clinical trials for head and neck cancer in the United States (Advexin, Introgen Therapeutics, Austin, TX). Some mutations adversely affect the stability of the core domain of p53119 yet are still expressed or even overexpressed; for this subset of mutants, compounds that stabilize the native protein conformation appear to restore its tumor suppressor activity.120,121 Such compounds have not yet advanced beyond preclinical studies. A third intriguing approach to targeting mutant p53 involves abrogating the G2 checkpoint of the cell cycle in the presence of traditional cytotoxic chemotherapeutics that cause DNA damage; because loss of p53 activity abolishes the G1 checkpoint, treatment with G2 checkpoint inhibitors forces the tumor cells into mitosis with irreparable DNA damage.122 Several inhibitors of the G2 checkpoint protein CHK1 are in phase I clinical trials in combination with cytotoxic agents.122 It remains to be seen which of these approaches will result in clinical benefit, if any.

STK11

STK11 encodes the serine/threonine protein kinase also known as LKB1 (liver kinase B1), which phosphorylates and activates AMP-activated protein kinase (AMPK) under conditions of low intracellular ATP levels; activated AMPK in turn inhibits mTOR in a TSC2- and RHEB-dependent manner.123 Truncating germline mutations of STK11 were identified in patients with Peutz-Jeghers syndrome, a rare hereditary disease characterized by predisposition to several types of malignancies.124 More recently, similar truncating but somatic mutations were found in lung adenocarcinoma patients.125 In the TSP experiment, 34 patients (18%) harbored somatic mutations of STK11, including 10 nonsense mutations, 9 frameshift insertions or deletions, and 6 splice site mutations as well as 9 missense mutations.8 STK11 knockout exacerbated KRAS G12D tumorigenesis in a mouse model of lung cancer, and these mice were used to show that a combination of MEK, PI3K, and SRC inhibition caused tumor regression in this model.126,127 Again, whether this approach has any clinical efficacy remains to be determined.

CDKN2A

Two tumor suppressor proteins are encoded by the human CDKN2A locus: p16 and p14ARF.128 Interestingly, these 2 transcripts utilize distinct first exons and alternative reading frames in the remaining exons. p16 functions to inhibit activity of the cyclin-dependent kinases CDK4 and CDK6, inducing arrest in G1 of the cell cycle by blocking phosphorylation of the RB protein. In contrast, p14 binds and inhibits MDM2-mediated ubiquitination and degradation of the tumor suppressor p53. Somatic mutations of CDKN2A were first described in lung adenocarcinoma in 1994.129 Subsequently, many mechanisms for CDKN2A inactivation were uncovered in addition to nonsynonymous point mutations, including homozygous deletions, frameshift and nonsense events that result in protein truncation, and promoter methylation.130-132 Only 9 samples in the TSP experiment (5%) were demonstrated to harbor mutant CDKN2A; however, the small size of this gene was a major factor contributing to statistical significance.8 Specifically, if we imagine one driver event per gene, then the driver mutation rate per MB will be higher for small genes, which allows us to better distinguish drivers from the background (M. Lawrence et al., unpublished data).

Importantly, whereas CDKN2A and TP53 mutations frequently coexist in lung adenocarcinoma, RB and p16 inactivation appear to be largely mutually exclusive.8,133,134 This is consistent with the idea that release of CDK inhibition is the required effect of CDKN2A somatic alterations. Several CDK inhibitors are in clinical trials, but none has yet been developed into an effective therapy for lung cancer.

NF1

The most significantly mutated gene in the TSP experiment that was not previously appreciated in lung cancer is the tumor suppressor gene NF1. NF1 was originally cloned as the tumor suppressor gene disrupted in the germline of patients with neurofibromatosis, characterized by benign Schwann cell tumors called neurofibromas, as well as an increased risk of malignancies.135-138 The NF1 protein product is a GTPase activating protein for RAS, stimulating hydrolysis of bound GTP to GDP on RAS, resulting in downregulation of RAS protein activity.136 In the TSP experiment, 7% of patients harbored somatic mutations of NF1, including 4 nonsense mutations, 5 splice site mutations, and 1 frameshift deletion as well as 6 missense mutations.8 Furthermore, 3 patients harbored 2 NF1 mutations each. Although it is not known if these mutation pairs are in cis or trans, this observation is potentially consistent with Knudsen’s 2-hit hypothesis for tumor suppressor gene inactivation.139 Because the primary effect of NF1 inactivation appears to be upregulation of RAS pathway signaling, the therapeutic approaches under investigation for tumors harboring KRAS mutations described above may also be useful in patients with somatic mutations of NF1.

ATM

Of 188 lung adenocarcinoma patients whose DNA was sequenced in the TSP experiment, 7% were found to harbor somatic mutations of the serine/threonine protein kinase gene ATM.8 These mutations included 10 missense mutations, 2 frameshift deletions, a splice site mutation, and a nonsense mutation, consistent with loss of function. Germline mutations of ATM, or “ataxia telangiectasia mutated,” are found in patients affected with the familial disease ataxia telangiectasia (AT), characterized by pleiotropic symptoms including sensitivity to ionizing radiation and predisposition to hematopoietic malignancies.140 ATM was subsequently shown to induce the G1 and G2 cell cycle checkpoints in response to DNA damage by phosphorylation of p53 and CHK2, respectively, although it is now known that ATM has a large number of substrates that contribute to the DNA damage response.141,142

Inactivating somatic mutations of ATM have been identified in several hematopoietic cancers, including T cell prolymphocytic leukemia, B cell chronic lymphocytic leukemia, and mantle cell lymphoma.143 However, recurring mutations of ATM had not been previously recognized in solid tumors prior to the TSP experiment. Inhibition of ATM has been shown to enhance cellular sensitivity to ionizing radiation, and there is some anecdotal evidence that the radiosensitivity of mantle cell lymphoma tumors can be in part attributed to ATM inactivation.144-147 To my knowledge, possible radiosensitization of lung adenocarcinoma tumor cells harboring mutations of ATM has not yet been explored.

APC

The familial adenomatous polyposis coli gene, APC, was identified by positional cloning in patients displaying a characteristic hereditary predisposition to colorectal tumors.92,148 APC mutations, primarily nonsense mutations and frameshift insertions and deletions encoding truncated proteins, were subsequently identified in a majority of sporadic colorectal adenomas and carcinomas.149 Somatic APC mutations have furthermore been described in several other solid tumors, most prominently in gastric and pancreatic cancers.150,151 In the lung adenocarcinoma TSP experiment, 13 somatic mutations of APC were detected in 11 patients, for a frequency of 6%.8 Mutations included 3 nonsense mutations, 4 frameshift insertions and deletions, a splice site mutation, and 5 missense mutations, again consistent with loss of function of a tumor suppressor gene.

As described above, APC is a scaffolding protein that forms a tumor suppressor complex with AXIN and GSK3β that phosphorylates β-catenin and targets it for proteasomal degradation.92 Because APC and KRAS mutations frequently co-occur in lung adenocarcinoma, the aforementioned therapeutic approach involving simultaneous inactivation of KRAS, CTNNB1, and ITF2 may be applicable to lung adenocarcinoma patients harboring inactivating mutations of APC.97 Small molecules that stabilize the APC-AXIN-GSK3β destruction complex or destabilize interaction of β-catenin with TCF/LEF transcription factor family members may likewise constitute a reasonable therapeutic approach for such lung adenocarcinoma patients.98,152 Because of the well-characterized frequency of inactivating APC mutations in colorectal carcinoma, these approaches are primarily being explored in the context of colon cancer; it remains to be seen whether any evidence will be produced supporting these approaches in lung adenocarcinoma cells harboring mutations of APC.

RB1

Although alterations of RB1 in small cell lung cancer have been known for many years,153,154 recurring and statistically significant RB1 mutations were only recently found in lung adenocarcinoma.8 Mutations of the tumor suppressor gene RB1 were identified in only 7 patients in the TSP experiment, for a frequency of 4%; however, only nonsense mutations, frameshift deletions, and splice site mutations were observed, all of which would be expected to result in a truncated protein product and occur infrequently, thus increasing statistical significance.8

RB associates with and modulates activity of the E2F family of transcription factors. In the classic paradigm, RB binds and sequesters E2Fs, thus inhibiting transcription of E2F target genes involved in cell cycle progression and growth promotion; phosphorylation by activated CDKs relieves this repression and permits transcription of the E2F target genes.155 Layers of complexity of RB function have since been uncovered.156 Rb1+/− mice develop pituitary and thyroid tumors; E2F-1 and E2F-4 losses ameliorate tumorigenesis in this model, whereas Skp2 loss causes overt synthetic lethality of aberrant pituitary melanotroph cells.157-159 However, there are currently no data to support these or any other putative therapeutic approaches for lung adenocarcinoma cells harboring RB1 mutations.

Next-Generation Sequencing Data

Much has thus been learned from exon-directed Sanger sequencing of lung adenocarcinoma samples. Several issues affect these experiments, however, including loss of power to detect tumor-specific somatic mutations in the presence of stromal contamination and limiting mutation detection to a subset of coding sequences. The falling cost of sequencing has recently enabled whole-exome and whole-genome cancer sequencing projects, which provide mutation analysis in a completely unbiased manner. Data collection for next-generation sequencing methods is moreover “digital,” collected for individual molecules, such that power to detect low-abundance alleles, whether because of stromal contamination or tumor heterogeneity, is dependent only on the depth of coverage. The whole-genome sequence from a single lung adenocarcinoma patient has already been reported.160 As whole-exome and whole-genome data from large numbers of samples are generated, a more global snapshot of somatic mutation in lung adenocarcinoma will emerge, providing a comprehensive view of the lung adenocarcinoma genome and putative therapeutic targets.

Acknowledgments

The author thanks Marcin Imielinski for critical reading of the article and Gad Getz and Michael Lawrence for discussions on power calculations in somatic mutation detection. This review is dedicated to the memory of Hidesaburo Hanafusa.

Footnotes

The author(s) declared no potential conflicts of interest with respect to the authorship and/or publication of this article.

This work was supported in part by a grant from Uniting Against Lung Cancer.

References

  • 1. Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA Cancer J Clin. 2010;60:277-300 [DOI] [PubMed] [Google Scholar]
  • 2. Travis WD, Travis LB, Devesa SS. Lung cancer. Cancer. 1995;75:191-202 [DOI] [PubMed] [Google Scholar]
  • 3. Subramanian J, Govindan R. Lung cancer in never smokers: a review. J Clin Oncol. 2007; 25:561-70 [DOI] [PubMed] [Google Scholar]
  • 4. Sharma SV, Settleman J. Exploiting the balance between life and death: targeted cancer therapy and “oncogenic shock.” Biochem Pharmacol. 2010;80:666-73 [DOI] [PubMed] [Google Scholar]
  • 5. Weinstein IB. Cancer. Addiction to oncogenes: the Achilles heal of cancer. Science. 2002;297:63-4 [DOI] [PubMed] [Google Scholar]
  • 6. Druker BJ, Sawyers CL, Kantarjian H, et al. Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with the Philadelphia chromosome. N Engl J Med. 2001;344:1038-42 [DOI] [PubMed] [Google Scholar]
  • 7. Joensuu H, Roberts PJ, Sarlomo-Rikala M, et al. Effect of the tyrosine kinase inhibitor STI571 in a patient with a metastatic gastrointestinal stromal tumor. N Engl J Med. 2001;344:1052-6 [DOI] [PubMed] [Google Scholar]
  • 8. Ding L, Getz G, Wheeler DA, et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature. 2008;455:1069-75 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Gandhi J, Zhang J, Xie Y, et al. Alterations in genes of the EGFR signaling pathway and their relationship to EGFR tyrosine kinase inhibitor sensitivity in lung cancer cell lines. PLoS ONE. 2009;4:e4576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Ladanyi M, Pao W. Lung adenocarcinoma: guiding EGFR-targeted therapy and beyond. Mod Pathol. 2008;21 Suppl 2:S16-22 [DOI] [PubMed] [Google Scholar]
  • 11. Sun Y, Ren Y, Fang Z, et al. Lung adenocarcinoma from East Asian never-smokers is a disease largely defined by targetable oncogenic mutant kinases. J Clin Oncol. 2010;28:4616-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Reynolds SH, Anna CK, Brown KC, et al. Activated protooncogenes in human lung tumors from smokers. Proc Natl Acad Sci U S A. 1991;88:1085-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Rodenhuis S, Slebos RJ, Boot AJ, et al. Incidence and possible clinical significance of K-ras oncogene activation in adenocarcinoma of the human lung. Cancer Res. 1988;48:5738-41 [PubMed] [Google Scholar]
  • 14. Barbacid M. ras genes. Annu Rev Biochem. 1987;56:779-827 [DOI] [PubMed] [Google Scholar]
  • 15. Campbell SL, Khosravi-Far R, Rossman KL, Clark GJ, Der CJ. Increasing complexity of Ras signaling. Oncogene. 1998;17:1395-413 [DOI] [PubMed] [Google Scholar]
  • 16. Brunner TB, Hahn SM, Gupta AK, Muschel RJ, McKenna WG, Bernhard EJ. Farnesyltransferase inhibitors: an overview of the results of preclinical and clinical investigations. Cancer Res. 2003;63:5656-68 [PubMed] [Google Scholar]
  • 17. Whyte DB, Kirschmeier P, Hockenberry TN, et al. K- and N-Ras are geranylgeranylated in cells treated with farnesyl protein transferase inhibitors. J Biol Chem. 1997;272:14459-64 [DOI] [PubMed] [Google Scholar]
  • 18. Engelman JA, Chen L, Tan X, et al. Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers. Nat Med. 2008;14:1351-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Hainsworth JD, Cebotaru CL, Kanarev V, et al. A phase II, open-label, randomized study to assess the efficacy and safety of AZD6244 (ARRY-142886) versus pemetrexed in patients with non-small cell lung cancer who have failed one or two prior chemotherapeutic regimens. J Thorac Oncol. 2010;5:1630-6 [DOI] [PubMed] [Google Scholar]
  • 20. Haura EB, Ricart AD, Larson TG, et al. A phase II study of PD-0325901, an oral MEK inhibitor, in previously treated patients with advanced non-small cell lung cancer. Clin Cancer Res. 2010;16:2450-7 [DOI] [PubMed] [Google Scholar]
  • 21. Rinehart J, Adjei AA, Lorusso PM, et al. Multicenter phase II study of the oral MEK inhibitor, CI-1040, in patients with advanced non-small-cell lung, breast, colon, and pancreatic cancer. J Clin Oncol. 2004;22:4456-62 [DOI] [PubMed] [Google Scholar]
  • 22. Blumenschein GR, Jr., Gatzemeier U, Fossella F, et al. Phase II, multicenter, uncontrolled trial of single-agent sorafenib in patients with relapsed or refractory, advanced non-small-cell lung cancer. J Clin Oncol. 2009;27:4274-80 [DOI] [PubMed] [Google Scholar]
  • 23. Hatzivassiliou G, Song K, Yen I, et al. RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth. Nature. 2010;464:431-5 [DOI] [PubMed] [Google Scholar]
  • 24. Heidorn SJ, Milagre C, Whittaker S, et al. Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAF. Cell. 2010;140:209-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Poulikakos PI, Zhang C, Bollag G, Shokat KM, Rosen N. RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF. Nature. 2010;464:427-30 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Kaelin WG., Jr. The concept of synthetic lethality in the context of anticancer therapy. Nat Rev Cancer. 2005;5:689-98 [DOI] [PubMed] [Google Scholar]
  • 27. Barbie DA, Tamayo P, Boehm JS, et al. Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1. Nature. 2009;462:108-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Luo J, Emanuele MJ, Li D, et al. A genome-wide RNAi screen identifies multiple synthetic lethal interactions with the Ras oncogene. Cell. 2009;137:835-48 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Scholl C, Frohling S, Dunn IF, et al. Synthetic lethal interaction between oncogenic KRAS dependency and STK33 suppression in human cancer cells. Cell. 2009;137:821-34 [DOI] [PubMed] [Google Scholar]
  • 30. Meylan E, Dooley AL, Feldser DM, et al. Requirement for NF-kappaB signalling in a mouse model of lung adenocarcinoma. Nature. 2009;462:104-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Puyol M, Martin A, Dubus P, et al. A synthetic lethal interaction between K-Ras oncogenes and Cdk4 unveils a therapeutic strategy for non-small cell lung carcinoma. Cancer Cell. 2010;18:63-73 [DOI] [PubMed] [Google Scholar]
  • 32. Singh A, Greninger P, Rhodes D, et al. A gene expression signature associated with “K-Ras addiction” reveals regulators of EMT and tumor cell survival. Cancer Cell. 2009;15:489-500 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Lynch TJ, Bell DW, Sordella R, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 2004;350:2129-39 [DOI] [PubMed] [Google Scholar]
  • 34. Paez JG, Janne PA, Lee JC, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science. 2004;304:1497-500 [DOI] [PubMed] [Google Scholar]
  • 35. Pao W, Miller V, Zakowski M, et al. EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib. Proc Natl Acad Sci U S A. 2004;101:13306-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Greulich H, Chen TH, Feng W, et al. Oncogenic transformation by inhibitor-sensitive and -resistant EGFR mutants. PLoS Med. 2005;2:e313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Huang SF, Liu HP, Li LH, et al. High frequency of epidermal growth factor receptor mutations with complex patterns in non-small cell lung cancers related to gefitinib responsiveness in Taiwan. Clin Cancer Res. 2004;10:8195-203 [DOI] [PubMed] [Google Scholar]
  • 38. Kosaka T, Yatabe Y, Endoh H, Kuwano H, Takahashi T, Mitsudomi T. Mutations of the epidermal growth factor receptor gene in lung cancer: biological and clinical implications. Cancer Res. 2004;64:8919-23 [DOI] [PubMed] [Google Scholar]
  • 39. Shigematsu H, Lin L, Takahashi T, et al. Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancers. J Natl Cancer Inst. 2005;97:339-46 [DOI] [PubMed] [Google Scholar]
  • 40. Wu JY, Wu SG, Yang CH, et al. Lung cancer with epidermal growth factor receptor exon 20 mutations is associated with poor gefitinib treatment response. Clin Cancer Res. 2008;14:4877-82 [DOI] [PubMed] [Google Scholar]
  • 41. Pao W, Chmielecki J. Rational, biologically based treatment of EGFR-mutant non-small-cell lung cancer. Nat Rev Cancer. 2010;10:760-74 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Maemondo M, Inoue A, Kobayashi K, et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med. 2010;362:2380-8 [DOI] [PubMed] [Google Scholar]
  • 43. Mitsudomi T, Morita S, Yatabe Y, et al. Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol. 2010;11:121-8 [DOI] [PubMed] [Google Scholar]
  • 44. Mok TS, Wu YL, Thongprasert S, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 2009;361:947-57 [DOI] [PubMed] [Google Scholar]
  • 45. Kobayashi S, Boggon TJ, Dayaram T, et al. EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N Engl J Med. 2005;352:786-92 [DOI] [PubMed] [Google Scholar]
  • 46. Pao W, Miller VA, Politi KA, et al. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med. 2005;2:e73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Gorre ME, Mohammed M, Ellwood K, et al. Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification. Science. 2001;293:876-80 [DOI] [PubMed] [Google Scholar]
  • 48. Yun CH, Mengwasser KE, Toms AV, et al. The T790M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP. Proc Natl Acad Sci U S A. 2008;105:2070-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Bell DW, Gore I, Okimoto RA, et al. Inherited susceptibility to lung cancer may be associated with the T790M drug resistance mutation in EGFR. Nat Genet. 2005;37:1315-6 [DOI] [PubMed] [Google Scholar]
  • 50. Vikis H, Sato M, James M, et al. EGFR-T790M is a rare lung cancer susceptibility allele with enhanced kinase activity. Cancer Res. 2007;67:4665-70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Li D, Ambrogio L, Shimamura T, et al. BIBW2992, an irreversible EGFR/HER2 inhibitor highly effective in preclinical lung cancer models. Oncogene. 2008;27:4702-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Li D, Shimamura T, Ji H, et al. Bronchial and peripheral murine lung carcinomas induced by T790M-L858R mutant EGFR respond to HKI-272 and rapamycin combination therapy. Cancer Cell. 2007;12:81-93 [DOI] [PubMed] [Google Scholar]
  • 53. Zhou W, Ercan D, Chen L, et al. Novel mutant-selective EGFR kinase inhibitors against EGFR T790M. Nature. 2009;462:1070-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Engelman JA, Zejnullahu K, Mitsudomi T, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316:1039-43 [DOI] [PubMed] [Google Scholar]
  • 55. Bean J, Brennan C, Shih JY, et al. MET amplification occurs with or without T790M mutations in EGFR mutant lung tumors with acquired resistance to gefitinib or erlotinib. Proc Natl Acad Sci U S A. 2007;104:20932-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Turke AB, Zejnullahu K, Wu YL, et al. Preexistence and clonal selection of MET amplification in EGFR mutant NSCLC. Cancer Cell. 2010;17:77-88 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Qi J, McTigue MA, Rogers A, et al. Multiple mutations and bypass mechanisms can contribute to development of acquired resistance to MET inhibitors. Cancer Res. 2011;71:1081-91 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Soda M, Choi YL, Enomoto M, et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature. 2007;448:561-6 [DOI] [PubMed] [Google Scholar]
  • 59. Morris SW, Kirstein MN, Valentine MB, et al. Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non-Hodgkin’s lymphoma. Science. 1994;263:1281-4 [DOI] [PubMed] [Google Scholar]
  • 60. Chen Z, Sasaki T, Tan X, et al. Inhibition of ALK, PI3K/MEK, and HSP90 in murine lung adenocarcinoma induced by EML4-ALK fusion oncogene. Cancer Res. 2010;70:9827-36 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Koivunen JP, Mermel C, Zejnullahu K, et al. EML4-ALK fusion gene and efficacy of an ALK kinase inhibitor in lung cancer. Clin Cancer Res. 2008;14:4275-83 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Kwak EL, Bang YJ, Camidge DR, et al. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med. 2010;363:1693-703 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Choi YL, Soda M, Yamashita Y, et al. EML4-ALK mutations in lung cancer that confer resistance to ALK inhibitors. N Engl J Med. 2010;363:1734-9 [DOI] [PubMed] [Google Scholar]
  • 64. Sasaki T, Okuda K, Zheng W, et al. The neuroblastoma-associated F1174L ALK mutation causes resistance to an ALK kinase inhibitor in ALK-translocated cancers. Cancer Res. 2010;70:10038-43 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Chen Y, Takita J, Choi YL, et al. Oncogenic mutations of ALK kinase in neuroblastoma. Nature. 2008;455:971-4 [DOI] [PubMed] [Google Scholar]
  • 66. George RE, Sanda T, Hanna M, et al. Activating mutations in ALK provide a therapeutic target in neuroblastoma. Nature. 2008;455:975-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Janoueix-Lerosey I, Lequin D, Brugieres L, et al. Somatic and germline activating mutations of the ALK kinase receptor in neuroblastoma. Nature. 2008;455:967-70 [DOI] [PubMed] [Google Scholar]
  • 68. Mosse YP, Laudenslager M, Longo L, et al. Identification of ALK as a major familial neuroblastoma predisposition gene. Nature. 2008;455:930-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Shigematsu H, Takahashi T, Nomura M, et al. Somatic mutations of the HER2 kinase domain in lung adenocarcinomas. Cancer Res. 2005;65:1642-6 [DOI] [PubMed] [Google Scholar]
  • 70. Stephens P, Hunter C, Bignell G, et al. Lung cancer: intragenic ERBB2 kinase mutations in tumours. Nature. 2004;431:525-6 [DOI] [PubMed] [Google Scholar]
  • 71. Hynes NE, Lane HA. ERBB receptors and cancer: the complexity of targeted inhibitors. Nat Rev Cancer. 2005;5:341-54 [DOI] [PubMed] [Google Scholar]
  • 72. Minami Y, Shimamura T, Shah K, et al. The major lung cancer-derived mutants of ERBB2 are oncogenic and are associated with sensitivity to the irreversible EGFR/ERBB2 inhibitor HKI-272. Oncogene. 2007;26:5023-7 [DOI] [PubMed] [Google Scholar]
  • 73. Shimamura T, Ji H, Minami Y, et al. Non-small-cell lung cancer and Ba/F3 transformed cells harboring the ERBB2 G776insV_G/C mutation are sensitive to the dual-specific epidermal growth factor receptor and ERBB2 inhibitor HKI-272. Cancer Res. 2006;66:6487-91 [DOI] [PubMed] [Google Scholar]
  • 74. Wang SE, Narasanna A, Perez-Torres M, et al. HER2 kinase domain mutation results in constitutive phosphorylation and activation of HER2 and EGFR and resistance to EGFR tyrosine kinase inhibitors. Cancer Cell. 2006;10:25-38 [DOI] [PubMed] [Google Scholar]
  • 75. Baselga J, Swain SM. Novel anticancer targets: revisiting ERBB2 and discovering ERBB3. Nat Rev Cancer. 2009;9:463-75 [DOI] [PubMed] [Google Scholar]
  • 76. Perera SA, Li D, Shimamura T, et al. HER2YVMA drives rapid development of adenosquamous lung tumors in mice that are sensitive to BIBW2992 and rapamycin combination therapy. Proc Natl Acad Sci U S A. 2009;106:474-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Lee JC, Vivanco I, Beroukhim R, et al. Epidermal growth factor receptor activation in glioblastoma through novel missense mutations in the extracellular domain. PLoS Med. 2006;3:e485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Network CGAR Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455:1061-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Brose MS, Volpe P, Feldman M, et al. BRAF and RAS mutations in human lung cancer and melanoma. Cancer Res. 2002;62:6997-7000 [PubMed] [Google Scholar]
  • 80. Naoki K, Chen TH, Richards WG, Sugarbaker DJ, Meyerson M. Missense mutations of the BRAF gene in human lung adenocarcinoma. Cancer Res. 2002;62:7001-3 [PubMed] [Google Scholar]
  • 81. Wan PT, Garnett MJ, Roe SM, et al. Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell. 2004;116:855-67 [DOI] [PubMed] [Google Scholar]
  • 82. Bollag G, Hirth P, Tsai J, et al. Clinical efficacy of a RAF inhibitor needs broad target blockade in BRAF-mutant melanoma. Nature. 2010;467:596-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Yang H, Higgins B, Kolinsky K, et al. RG7204 (PLX4032), a selective BRAFV600E inhibitor, displays potent antitumor activity in preclinical melanoma models. Cancer Res. 2010;70:5518-27 [DOI] [PubMed] [Google Scholar]
  • 84. Pratilas CA, Hanrahan AJ, Halilovic E, et al. Genetic predictors of MEK dependence in non-small cell lung cancer. Cancer Res. 2008;68:9375-83 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Samuels Y, Wang Z, Bardelli A, et al. High frequency of mutations of the PIK3CA gene in human cancers. Science. 2004;304:554. [DOI] [PubMed] [Google Scholar]
  • 86. Lee JW, Soung YH, Kim SY, et al. PIK3CA gene is frequently mutated in breast carcinomas and hepatocellular carcinomas. Oncogene. 2005;24:1477-80 [DOI] [PubMed] [Google Scholar]
  • 87. Marks JL, McLellan MD, Zakowski MF, et al. Mutational analysis of EGFR and related signaling pathway genes in lung adenocarcinomas identifies a novel somatic kinase domain mutation in FGFR4. PLoS ONE. 2007;2:e426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Yamamoto H, Shigematsu H, Nomura M, et al. PIK3CA mutations and copy number gains in human lung cancers. Cancer Res. 2008;68:6913-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Kang S, Bader AG, Vogt PK. Phosphatidylinositol 3-kinase mutations identified in human cancer are oncogenic. Proc Natl Acad Sci U S A. 2005;102:802-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Samuels Y, Diaz LA, Jr., Schmidt-Kittler O, et al. Mutant PIK3CA promotes cell growth and invasion of human cancer cells. Cancer Cell. 2005;7:561-73 [DOI] [PubMed] [Google Scholar]
  • 91. Sasaki H, Okuda K, Kawano O, et al. Nras and Kras mutation in Japanese lung cancer patients: genotyping analysis using LightCycler. Oncol Rep. 2007;18:623-8 [PubMed] [Google Scholar]
  • 92. Fodde R, Smits R, Clevers H. APC, signal transduction and genetic instability in colorectal cancer. Nat Rev Cancer. 2001;1:55-67 [DOI] [PubMed] [Google Scholar]
  • 93. Polakis P. The many ways of Wnt in cancer. Curr Opin Genet Dev. 2007;17:45-51 [DOI] [PubMed] [Google Scholar]
  • 94. Mirabelli-Primdahl L, Gryfe R, Kim H, et al. Beta-catenin mutations are specific for colorectal carcinomas with microsatellite instability but occur in endometrial carcinomas irrespective of mutator pathway. Cancer Res. 1999;59:3346-51 [PubMed] [Google Scholar]
  • 95. Morin PJ, Sparks AB, Korinek V, et al. Activation of beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC. Science. 1997;275:1787-90 [DOI] [PubMed] [Google Scholar]
  • 96. Sunaga N, Kohno T, Kolligs FT, Fearon ER, Saito R, Yokota J. Constitutive activation of the Wnt signaling pathway by CTNNB1 (beta-catenin) mutations in a subset of human lung adenocarcinoma. Genes Chromosomes Cancer. 2001;30:316-21 [DOI] [PubMed] [Google Scholar]
  • 97. Mologni L, Dekhil H, Ceccon M, et al. Colorectal tumors are effectively eradicated by combined inhibition of {beta}-catenin, KRAS, and the oncogenic transcription factor ITF2. Cancer Res. 2010;70:7253-63 [DOI] [PubMed] [Google Scholar]
  • 98. Waaler J, Machon O, von Kries JP, et al. Novel synthetic antagonists of canonical Wnt signaling inhibit colorectal cancer cell growth. Cancer Res. 2011;71:197-205 [DOI] [PubMed] [Google Scholar]
  • 99. Franke TF. Intracellular signaling by Akt: bound to be specific. Sci Signal. 2008;1:pe29. [DOI] [PubMed] [Google Scholar]
  • 100. Askham JM, Platt F, Chambers PA, Snowden H, Taylor CF, Knowles MA. AKT1 mutations in bladder cancer: identification of a novel oncogenic mutation that can co-operate with E17K. Oncogene. 2010;29:150-5 [DOI] [PubMed] [Google Scholar]
  • 101. Carpten JD, Faber AL, Horn C, et al. A transforming mutation in the pleckstrin homology domain of AKT1 in cancer. Nature. 2007;448:439-44 [DOI] [PubMed] [Google Scholar]
  • 102. Malanga D, Scrima M, De Marco C, et al. Activating E17K mutation in the gene encoding the protein kinase AKT1 in a subset of squamous cell carcinoma of the lung. Cell Cycle. 2008;7:665-9 [DOI] [PubMed] [Google Scholar]
  • 103. Marks JL, Gong Y, Chitale D, et al. Novel MEK1 mutation identified by mutational analysis of epidermal growth factor receptor signaling pathway genes in lung adenocarcinoma. Cancer Res. 2008;68:5524-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Mohi MG, Neel BG. The role of Shp2 (PTPN11) in cancer. Curr Opin Genet Dev. 2007;17:23-30 [DOI] [PubMed] [Google Scholar]
  • 105. Tartaglia M, Niemeyer CM, Fragale A, et al. Somatic mutations in PTPN11 in juvenile myelomonocytic leukemia, myelodysplastic syndromes and acute myeloid leukemia. Nat Genet. 2003;34:148-50 [DOI] [PubMed] [Google Scholar]
  • 106. Bentires-Alj M, Paez JG, David FS, et al. Activating mutations of the noonan syndrome-associated SHP2/PTPN11 gene in human solid tumors and adult acute myelogenous leukemia. Cancer Res. 2004;64:8816-20 [DOI] [PubMed] [Google Scholar]
  • 107. Keilhack H, David FS, McGregor M, Cantley LC, Neel BG. Diverse biochemical properties of Shp2 mutants: implications for disease phenotypes. J Biol Chem. 2005;280:30984-93 [DOI] [PubMed] [Google Scholar]
  • 108. Niihori T, Aoki Y, Ohashi H, et al. Functional analysis of PTPN11/SHP-2 mutants identified in Noonan syndrome and childhood leukemia. J Hum Genet. 2005;50:192-202 [DOI] [PubMed] [Google Scholar]
  • 109. Mohi MG, Williams IR, Dearolf CR, et al. Prognostic, therapeutic, and mechanistic implications of a mouse model of leukemia evoked by Shp2 (PTPN11) mutations. Cancer Cell. 2005;7:179-91 [DOI] [PubMed] [Google Scholar]
  • 110. Vazquez A, Bond EE, Levine AJ, Bond GL. The genetics of the p53 pathway, apoptosis and cancer therapy. Nat Rev Drug Discov. 2008;7:979-87 [DOI] [PubMed] [Google Scholar]
  • 111. Nigro JM, Baker SJ, Preisinger AC, et al. Mutations in the p53 gene occur in diverse human tumour types. Nature. 1989;342:705-8 [DOI] [PubMed] [Google Scholar]
  • 112. Takahashi T, Nau MM, Chiba I, et al. p53: a frequent target for genetic abnormalities in lung cancer. Science. 1989;246:491-4 [DOI] [PubMed] [Google Scholar]
  • 113. Brosh R, Rotter V. When mutants gain new powers: news from the mutant p53 field. Nat Rev Cancer. 2009;9:701-13 [DOI] [PubMed] [Google Scholar]
  • 114. Beroukhim R, Mermel CH, Porter D, et al. The landscape of somatic copy-number alteration across human cancers. Nature. 2010;463:899-905 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Weir BA, Woo MS, Getz G, et al. Characterizing the cancer genome in lung adenocarcinoma. Nature. 2007;450:893-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Levine AJ, Oren M. The first 30 years of p53: growing ever more complex. Nat Rev Cancer. 2009;9:749-58 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Mandinova A, Lee SW. The p53 pathway as a target in cancer therapeutics: obstacles and promise. Sci Transl Med. 2011;3:64rv1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Peng Z. Current status of gendicine in China: recombinant human Ad-p53 agent for treatment of cancers. Hum Gene Ther. 2005;16:1016-27 [DOI] [PubMed] [Google Scholar]
  • 119. Friedler A, Veprintsev DB, Hansson LO, Fersht AR. Kinetic instability of p53 core domain mutants: implications for rescue by small molecules. J Biol Chem. 2003;278:24108-12 [DOI] [PubMed] [Google Scholar]
  • 120. Demma M, Maxwell E, Ramos R, et al. SCH529074, a small molecule activator of mutant p53, which binds p53 DNA binding domain (DBD), restores growth-suppressive function to mutant p53 and interrupts HDM2-mediated ubiquitination of wild type p53. J Biol Chem. 2010;285:10198-212 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Lambert JM, Gorzov P, Veprintsev DB, et al. PRIMA-1 reactivates mutant p53 by covalent binding to the core domain. Cancer Cell. 2009;15:376-88 [DOI] [PubMed] [Google Scholar]
  • 122. Ma CX, Janetka JW, Piwnica-Worms H. Death by releasing the breaks: CHK1 inhibitors as cancer therapeutics. Trends Mol Med. 2011;17:88-96 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Shackelford DB, Shaw RJ. The LKB1-AMPK pathway: metabolism and growth control in tumour suppression. Nat Rev Cancer. 2009;9:563-75 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Hemminki A, Markie D, Tomlinson I, et al. A serine/threonine kinase gene defective in Peutz-Jeghers syndrome. Nature. 1998;391:184-7 [DOI] [PubMed] [Google Scholar]
  • 125. Sanchez-Cespedes M, Parrella P, Esteller M, et al. Inactivation of LKB1/STK11 is a common event in adenocarcinomas of the lung. Cancer Res. 2002;62:3659-62 [PubMed] [Google Scholar]
  • 126. Carretero J, Shimamura T, Rikova K, et al. Integrative genomic and proteomic analyses identify targets for Lkb1-deficient metastatic lung tumors. Cancer Cell. 2010;17:547-59 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Ji H, Ramsey MR, Hayes DN, et al. LKB1 modulates lung cancer differentiation and metastasis. Nature. 2007;448:807-10 [DOI] [PubMed] [Google Scholar]
  • 128. Chin L, Pomerantz J, DePinho RA. The INK4a/ARF tumor suppressor: one gene—two products—two pathways. Trends Biochem Sci. 1998;23:291-6 [DOI] [PubMed] [Google Scholar]
  • 129. Hayashi N, Sugimoto Y, Tsuchiya E, Ogawa M, Nakamura Y. Somatic mutations of the MTS (multiple tumor suppressor) 1/CDK4l (cyclin-dependent kinase-4 inhibitor) gene in human primary non-small cell lung carcinomas. Biochem Biophys Res Commun. 1994;202:1426-30 [DOI] [PubMed] [Google Scholar]
  • 130. Merlo A, Herman JG, Mao L, et al. 5′ CpG island methylation is associated with transcriptional silencing of the tumour suppressor p16/CDKN2/MTS1 in human cancers. Nat Med. 1995;1:686-92 [DOI] [PubMed] [Google Scholar]
  • 131. Packenham JP, Taylor JA, White CM, Anna CH, Barrett JC, Devereux TR. Homozygous deletions at chromosome 9p21 and mutation analysis of p16 and p15 in microdissected primary non-small cell lung cancers. Clin Cancer Res. 1995;1:687-90 [PubMed] [Google Scholar]
  • 132. Shapiro GI, Park JE, Edwards CD, et al. Multiple mechanisms of p16INK4A inactivation in non-small cell lung cancer cell lines. Cancer Res. 1995;55:6200-9 [PubMed] [Google Scholar]
  • 133. Sanchez-Cespedes M, Reed AL, Buta M, et al. Inactivation of the INK4A/ARF locus frequently coexists with TP53 mutations in non-small cell lung cancer. Oncogene. 1999;18:5843-9 [DOI] [PubMed] [Google Scholar]
  • 134. Shapiro GI, Edwards CD, Kobzik L, et al. Reciprocal Rb inactivation and p16INK4 expression in primary lung cancers and cell lines. Cancer Res. 1995;55:505-9 [PubMed] [Google Scholar]
  • 135. Cawthon RM, Weiss R, Xu GF, et al. A major segment of the neurofibromatosis type 1 gene: cDNA sequence, genomic structure, and point mutations. Cell. 1990;62:193-201 [DOI] [PubMed] [Google Scholar]
  • 136. Lee MJ, Stephenson DA. Recent developments in neurofibromatosis type 1. Curr Opin Neurol. 2007;20:135-41 [DOI] [PubMed] [Google Scholar]
  • 137. Viskochil D, Buchberg AM, Xu G, et al. Deletions and a translocation interrupt a cloned gene at the neurofibromatosis type 1 locus. Cell. 1990;62:187-92 [DOI] [PubMed] [Google Scholar]
  • 138. Wallace MR, Marchuk DA, Andersen LB, et al. Type 1 neurofibromatosis gene: identification of a large transcript disrupted in three NF1 patients. Science. 1990;249:181-6 [DOI] [PubMed] [Google Scholar]
  • 139. Knudson AG., Jr. Mutation and cancer: statistical study of retinoblastoma. Proc Natl Acad Sci U S A. 1971;68:820-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Savitsky K, Bar-Shira A, Gilad S, et al. A single ataxia telangiectasia gene with a product similar to PI-3 kinase. Science. 1995;268:1749-53 [DOI] [PubMed] [Google Scholar]
  • 141. Abraham RT. Cell cycle checkpoint signaling through the ATM and ATR kinases. Genes Dev. 2001;15:2177-96 [DOI] [PubMed] [Google Scholar]
  • 142. Lavin MF. Ataxia-telangiectasia: from a rare disorder to a paradigm for cell signalling and cancer. Nat Rev Mol Cell Biol. 2008;9:759-69 [DOI] [PubMed] [Google Scholar]
  • 143. Boultwood J. Ataxia telangiectasia gene mutations in leukaemia and lymphoma. J Clin Pathol. 2001;54:512-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Filippi AR, Franco P, Galliano M, Ricardi U. Peripheral blood complete remission after splenic irradiation in mantle-cell lymphoma with 11q22-23 deletion and ATM inactivation. Radiat Oncol. 2006;1:35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. M’Kacher R, Bennaceur A, Farace F, et al. Multiple molecular mechanisms contribute to radiation sensitivity in mantle cell lymphoma. Oncogene. 2003;22:7905-12 [DOI] [PubMed] [Google Scholar]
  • 146. Rainey MD, Charlton ME, Stanton RV, Kastan MB. Transient inhibition of ATM kinase is sufficient to enhance cellular sensitivity to ionizing radiation. Cancer Res. 2008;68:7466-74 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. White JS, Choi S, Bakkenist CJ. Transient ATM kinase inhibition disrupts DNA damage-induced sister chromatid exchange. Sci Signal. 2010;3:ra44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Groden J, Thliveris A, Samowitz W, et al. Identification and characterization of the familial adenomatous polyposis coli gene. Cell. 1991;66:589-600 [DOI] [PubMed] [Google Scholar]
  • 149. Powell SM, Zilz N, Beazer-Barclay Y, et al. APC mutations occur early during colorectal tumorigenesis. Nature. 1992;359:235-7 [DOI] [PubMed] [Google Scholar]
  • 150. Horii A, Nakatsuru S, Miyoshi Y, et al. Frequent somatic mutations of the APC gene in human pancreatic cancer. Cancer Res. 1992;52:6696-8 [PubMed] [Google Scholar]
  • 151. Horii A, Nakatsuru S, Miyoshi Y, et al. The APC gene, responsible for familial adenomatous polyposis, is mutated in human gastric cancer. Cancer Res. 1992;52:3231-3 [PubMed] [Google Scholar]
  • 152. Lepourcelet M, Chen YN, France DS, et al. Small-molecule antagonists of the oncogenic Tcf/beta-catenin protein complex. Cancer Cell. 2004;5:91-102 [DOI] [PubMed] [Google Scholar]
  • 153. Harbour JW, Lai SL, Whang-Peng J, Gazdar AF, Minna JD, Kaye FJ. Abnormalities in structure and expression of the human retinoblastoma gene in SCLC. Science. 1988;241:353-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Yokota J, Akiyama T, Fung YK, et al. Altered expression of the retinoblastoma (RB) gene in small-cell carcinoma of the lung. Oncogene. 1988;3:471-5 [PubMed] [Google Scholar]
  • 155. Taya Y. RB kinases and RB-binding proteins: new points of view. Trends Biochem Sci. 1997;22:14-7 [DOI] [PubMed] [Google Scholar]
  • 156. Chen HZ, Tsai SY, Leone G. Emerging roles of E2Fs in cancer: an exit from cell cycle control. Nat Rev Cancer. 2009;9:785-97 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Lee EY, Cam H, Ziebold U, Rayman JB, Lees JA, Dynlacht BD. E2F4 loss suppresses tumorigenesis in Rb mutant mice. Cancer Cell. 2002;2:463-72 [DOI] [PubMed] [Google Scholar]
  • 158. Wang H, Bauzon F, Ji P, et al. Skp2 is required for survival of aberrantly proliferating Rb1-deficient cells and for tumorigenesis in Rb1+/− mice. Nat Genet. 2010;42:83-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Yamasaki L, Bronson R, Williams BO, Dyson NJ, Harlow E, Jacks T. Loss of E2F-1 reduces tumorigenesis and extends the lifespan of Rb1(+/−)mice. Nat Genet. 1998;18:360-4 [DOI] [PubMed] [Google Scholar]
  • 160. Lee W, Jiang Z, Liu J, et al. The mutation spectrum revealed by paired genome sequences from a lung cancer patient. Nature. 2010;465:473-77 [DOI] [PubMed] [Google Scholar]

Articles from Genes & Cancer are provided here courtesy of Impact Journals, LLC

RESOURCES