Metastatic Non–Small Cell Lung Cancer Management: Novel Targets and Recent Clinical Advances

Jonathan W. Riess, MD, and Heather A. Wakelee, MD

Jonathan W. Riess, MD, and Heather A. Wakelee, MD

Dr. Riess is a Fellow and Dr. Wakelee is an Assistant Professor in the Department of Medicine, Division of Oncology, at Stanford Cancer Institute, Stanford University School of Medicine, in Stanford, California.

Address correspondence to: Heather A. Wakelee, MD, Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford Cancer Institute, 875 Blake Wilbur Dr., Stanford, CA 94305; E-mail: hwakelee@stanford.edu

Abstract: Lung cancer continues to be the most common cause of cancer-related mortality in the United States and other developed countries. The most common subtype is non–small cell lung cancer (NSCLC). Within NSCLC, we are discovering remarkable molecular heterogeneity. Most current actionable mutations have been identified in patients with adenocarcinoma histology, but now new mutations are being discovered in squamous cell histology patients as well. This molecular heterogeneity provides an opportunity for clinical trials to exploit various candidate oncogene-addicted pathways in NSCLC. This article focuses on 2 shifting paradigms in NSCLC management: the recent advances in targeted therapy and maintenance treatment.

Introduction

We are in the midst of an exciting time in the treatment of non–small cell lung cancer (NSCLC), with remarkable recent advances in management. Treatment is increasingly being driven by molecular characteristics. Drugs targeting driver mutations have greatly improved outcomes in the selected patients who harbor these mutations, such as erlotinib (Tarceva, Genentech/OSI) in NSCLC with epidermal growth factor receptor (EGFR) activating mutations and, most recently, crizotinib (PF-02341066, Pfizer) in NSCLC with an anaplastic lymphoma kinase (ALK) translocation. The search for other important driver mutations and drugs to target these dysregulated pathways is ongoing (Table 1).

The ability to link molecular markers to targeted treatments, both in the trial setting and as standard of care for patients with ALK and EGFR activating mutations, has radically altered our approach to therapy (Table 2). Molecular profiling is important for current treatment and for future advances in lung cancer. For example, the Lung Cancer Mutation Consortium, a group of 14 institutions, recently presented data profiling tissue from NSCLC adenocarcinoma for specific mutations and linked them to relevant targeted treatment with either approved drugs or clinical trials.1 A similar analysis of a smaller group of squamous cell histology patients indicates that identification of driver mutations is also feasible in that subset of NSCLC patients.2 The BATTLE (Biomarker-Integrated Approaches of Targeted Therapy for Lung Cancer Elimination) trial illustrates an exciting new approach to NSCLC clinical trials. This study demonstrated the feasibility of obtaining repeat biopsies after initial progression in heavily pretreated patients to guide therapy. The study adaptively randomized patients based on biomarker results from pretreatment biopsies in an attempt to determine subsets of patients who may benefit from specific targeted treatments.3

In addition to targeted treatments, maintenance treatment of NSCLC has also gained increasing popularity as a new treatment paradigm in NSCLC. This review is intended as a summary of the most relevant recent advances in metastatic NSCLC treatment, with a focus on maintenance therapy, targeted agents, and novel immunotherapeutics.

Targeting EGFR in Patients With EGFR Activating Mutations

EGFR activating mutations are found in approximately 15–20% of lung adenocarcinoma patients, with a higher frequency in never-smokers, Asians, and women.1,4 However, lung adenocarcinomas from former and current smokers may also contain an EGFR activating mutation, with recent data showing that approximately 6% of adenocarcinomas from current smokers and 15% from former smokers harbor an EGFR activating mutation.5

EGFR tyrosine kinase inhibitors (EGFR-TKIs) are now the standard of care for first-line treatment of patients whose tumors have an EGFR activating mutation. A prespecified subset analysis of the landmark IPASS (Iressa Pan-Asia Study) first showed that the EGFR-TKI gefitinib (Iressa, AstraZeneca) prolongs progression-free survival (PFS) compared to first-line chemotherapy in Asian patients who harbor an activating EGFR mutation.4 Results from a Japanese trial confirmed the PFS benefit of EGFR-TKIs compared to platinum doublet therapy.6 The EURTAC (European Erlotinib Versus Chemotherapy) trial confirmed this PFS benefit in a predominantly European population.7 The lack of overall survival (OS) benefit seen throughout these trials, despite prolongation of PFS, is most likely secondary to patient crossover, with most patients eventually going on to receive an EGFR-TKI.

Unfortunately, resistance develops in most EGFR mutant tumors, with a median PFS of 10.8 months in the Japanese study, 9.5 months in the IPASS study, and 9.7 months in the European study. The 2 main mechanisms of resistance to EGFR-TKIs include the T790M mutation, which represents about 50% of all resistance mutations, and MET amplification, which may represent up to 20%.8 The T790M mutation changes the conformation of the EGFR receptor and sterically hinders the binding of the EGFR-TKI.9 MET amplification leads to EGFR independent activation of the downstream PI3K-AKT pathway.10 Discovery of other resistance mechanisms is ongoing.

Numerous efforts to rationally combine other targeted agents to overcome resistance to EGFR-TKI are under way. Currently, there is promising phase II data combining the irreversible EGFR-TKI afatinib and the EGFR antibody cetuximab (Erbitux, ImClone/Bristol-Myers Squibb). In a phase II trial, afatinib combined with cetuximab in 61 patients with acquired EGFR resistance produced a 100% disease control rate and a high partial response (PR) rate, which was achieved by approximately one-third of patients with a T790M mutation.11

Aside from novel therapeutics, patients who become resistant to EGFR-TKI may benefit in the future from re-exposure to an EGFR-TKI. After time off an EGFR-TKI, the patient may be resensitized to EGFR inhibition from regrowth of a fraction of nonresistant EGFR-mutated tumor cells. A small retrospective study showed a 73% disease control rate in 11 patients retreated with gefitinib after initial treatment failure.12

Debate exists over whether EGFR-TKIs should be combined with chemotherapy, and whether they should be continued beyond progression. Early clinical trials combining EGFR-TKIs with chemotherapy were negative in an unselected population.13,14 The case for continuing a TKI in patients with an EGFR mutation is based on a more rapid progression noted in some patients taken off EGFR-TKIs after initial progression.15 If an EGFR activating mutation is discovered during first-line chemotherapy, the most recent National Comprehensive Cancer Network (NCCN) guidelines suggest adding erlotinib as a category 2B recommendation. However, there is no consensus on this point and many providers will continue the chemotherapy until progression, then switch to the EGFR-TKI or add it as maintenance therapy. The CALGB (Cancer and Leukemia Group B) trial, which looked at combined chemotherapy and erlotinib, did not demonstrate a survival benefit for the combination, even in patients with EGFR activating mutations.16

Targeting EGFR in an Unselected NSCLC Population

In contrast to EGFR-TKIs, which function through intracellular inhibition of the EGFR tyrosine kinase domain, cetuximab is a monoclonal antibody that binds to the extracellular domain of EGFR. The FLEX (First-Line Erbitux in Lung Cancer) study assigned 1,125 patients with NSCLC to either cisplatin and vinorelbine or cisplatin, vinorelbine, and cetuximab.17 Median overall survival was 1.2 months longer in the patients receiving cetuximab (P=.044). Based on a subset analysis, benefit seemed greatest in squamous cell histology patients (hazard ratio [HR], 0.80). However, when added to carboplatin and paclitaxel, cetuximab did not increase progression-free survival (PFS) or overall survival (OS), despite a higher overall response rate (ORR).18

With mixed data regarding the benefit of cetuximab and OS, identifying biomarkers to select patients who preferentially benefit from cetuximab is of great interest. A subgroup analysis of the FLEX study suggested that patients who had a first-cycle rash might benefit more from cetuximab, with an OS of 15 months versus 8.8 months in patients who did not have a first-cycle rash.19 Another potential biomarker is EGFR expression assayed by immunohistochemistry (IHC). In a retrospective analysis, an EGFR IHC score was generated from a percentage of cancer cells that expressed EGFR with a range of 0–300. Dichotomizing patients into IHC scores greater or less than 200, the median OS in the high-expression group treated with cetuximab plus chemotherapy was 12 months, compared with 9.6 months in high-expression patients treated with chemotherapy alone (HR, 0.73; P=.011).20 Although cetuximab is not approved by the US Food and Drug Administration (FDA) for the treatment of NSCLC, these selection criteria may lead to a role for the drug in subsets of NSCLC patients.

ALK

Since the ALK oncogenic fusion protein was identified in a NSCLC patient in 2007,21 there has been rapid development of agents targeting ALK. In unselected NSCLC patients, 2–7% harbor an ALK gene rearrangement. In lung adenocarcinoma, ALK’s fusion partner, EML4, lies upstream on chromosome 2. Nine different inversions of EML4-ALK that cause constitutive tyrosine kinase activation have been identified.22

Crizotinib (Xalkori, Pfizer), an oral tyrosine kinase inhibitor, was initially developed as a MET inhibitor, but activity in a NSCLC patient who was later found to have an ALK gene rearrangement spurred its development in ALK-positive NSCLC. In a phase I/II trial of these patients, crizotinib as second-line treatment led to a response in 57% of patients and disease stability in 33% of patients.23 Resistance eventually developed, with a mean PFS of 14 weeks and a 6-month PFS achieved by 27.2% of patients. Mechanisms of resistance to ALK inhibition are an area of active investigation. Like the T790M EGFR mutation, some resistance to ALK inhibition is mediated by gatekeeper mutations that alter the ATP binding site.24 Crizotinib has recently been given accelerated approval by the FDA in metastatic NSCLC patients harboring an ALK translocation along with a companion fluorescent in situ hybridization (FISH) diagnostic test. Approval is contingent upon the results of the ongoing phase III PROFILE 1007 (A Phase III Trial of Crizotinib Versus Standard of Care in Patients With Advanced Non–Small-Cell Lung Cancer With a Specific Alteration of the Anaplastic Lymphoma Kinase Gene) trial of crizotinib versus physician’s choice of pemetrexed (Alimta, Eli Lilly) or docetaxel in the second-line setting and beyond.

NSCLC patients with nonsquamous histology preferentially benefit from pemetrexed over gemcitabine. The inverse is true for patients with squamous cell histology who do not appear to derive benefit from pemetrexed. This was shown in a phase III study, where nonsquamous histology patients who received cisplatin and pemetrexed had improved OS compared with cisplatin and gemcitabine in the first-line setting.25 Within NSCLC-adenocarcinoma patients, data are emerging that patients who harbor an ALK translocation may have an improved response to pemetrexed. In a retrospective, exploratory analysis, patients with an ALK gene rearrangement had a prolonged PFS (9 months) compared with patients with a K-ras mutation (7 months), EGFR activating mutation (5.5 months), or neither of these mutations (4 months).26 Another retrospective analysis showed increased time to progression (TTP) and ORR in patients whose tumors harbored an ALK translocation, compared with EGFR mutant and wild-type tumors (TTP 9.4 months vs 1.4 months vs 2.9 months, respectively).27 A hypothesis for the differential effect of pemetrexed in ALK-positive tumors includes increased response to lower thymidylate synthase levels as well as differential expression of other pathways involved in DNA base biosynthesis.28 These intriguing results await further preclinical studies and clinical validation in a prospective fashion.

Efforts to overcome resistance to crizotinib are ongoing, and irreversible ALK inhibitors are currently in development. Another promising class of drugs for ALK-positive patients is heat-shock protein-90 (HSP90) inhibitors. In a crizotinib-resistant ALK-positive cell line, HSP90 inhibition overcomes crizotinib resistance and suppresses phosphorylation of ALK, AKT, and ERK, inducing marked apoptosis.29 In an open-label, phase II study of the HSP90 inhibitor retaspimycin (IPI-504, Infinity/MedImmune), 96 patients were enrolled with a primary endpoint of 16-week PFS. Among the 76 evaluable patients, the PFS rate at 16 weeks was 24.1%, with a 5.3% ORR and 54% disease stability rate. All objective responders were patients with ALK gene rearrangements.30 Ganetespib (STA-9090) is another HSP90 inhibitor with promising early phase data. A phase IIb/III trial of ganetespib and docetaxel is currently ongoing.

K-ras

K-ras is the most frequent mutation found among NSCLC patients with adenocarcinoma. Despite being the first driver mutation discovered in NSCLC, therapies targeting K-ras have been less successful than those targeting EGFR and ALK thus far. ARQ197 (tivantinib), a noncompetitive inhibitor of c-MET, may have some activity in patients with K-ras mutations. In a randomized phase II trial of patients treated with erlotinib plus ARQ197 versus erlotinib alone, PFS was 3.7 months versus 2.2 months, just approaching statistical significance. In a subset analysis, K-ras mutant patients derived the most benefit (HR, 0.18), but the total number of patients studied with K-ras mutations was small. The phase III MARQUEE (Met Inhibitor ARQ 197 Plus Erlotinib vs Erlotinib Plus Placebo in NSCLC) trial is comparing erlotinib plus ARQ197 to chemotherapy in nonsquamous metastatic patients, and is stratified by EGFR and K-ras mutation status.

Although it is being combined with ARQ197, erlotinib alone may be less effective as a single-agent, second-line treatment of NSCLC with K-ras mutations. A recent molecular marker analysis of the SATURN (Sequential Tarceva in Unresectable NSCLC) trial, which showed a modest OS benefit of erlotinib maintenance in unselected patients, indicated that K-ras mutation status predicted decreased benefit from erlotinib treatment.31 The recently published BATTLE trial also showed that patients with K-ras mutations had a poorer 8-week disease control rate with erlotinib than with sorafenib (79% vs 14%; P=.016). A trend towards increased 8-week disease control rate in patients with K-ras mutant tumors treated with sorafenib compared with the other treatments in the BATTLE study (61% vs 32%; P=.11) may also signal preferential benefit of sorafenib in K-ras tumors.

MET

MET is a tyrosine kinase receptor that binds hepatocyte growth factor leading to prosurvival, motility, and proliferation signals.32 Overexpression is associated with worse prognosis in several tumor types, including NSCLC. Promising data were presented at the 2011 American Society of Clinical Oncology (ASCO) meeting on a phase II trial combining erlotinib with onartuzumab (MetMab, Genentech), a monovalent antibody that inhibits hepatocyte growth factor–mediated stimulation of the MET receptor.33

In a phase II trial, 137 patients were randomized to receive erlotinib plus placebo or erlotinib plus onartuzumab, with crossover allowed upon progression. No difference in PFS or OS was noted in the intent-to-treat population. However, significant improvement in PFS and OS was noted in the MET IHC-positive patients. Trends toward harm were noted in the MET IHC-negative patients. Benefit in MET IHC-positive patients was noted even in EGFR wild-type patients treated with erlotinib and onartuzumab. Based on these phase II results, a phase III trial is under way in MET IHC-positive patients, as is a phase II trial combining chemotherapy with onartuzumab.

PI3-Kinase (PI3K)/BRAF

There are several less common mutations in NSCLC patients. PI3K mutations have been identified in several tumor types, but only approximately 2% of NSCLC patients.34 Both adenocarcinoma and squamous cell histology can harbor these mutations, and PI3K mutations can coexist with EGFR mutations.35 BKM120 is a competitive inhibitor of PI3K that is currently being studied in clinical trials. BEZ235 is a dual competitive inhibitor of PI3K and mTOR that recently completed phase I trials.

Approximately 2% of lung adenocarcinoma patients harbor a BRAF mutation,1 and they are less likely to harbor the V600E mutation than melanoma patients.36-38 Ongoing trials are looking at BRAF inhibitors in NSCLC patients with BRAF mutations. BRAF mutations have also been shown to predict sensitivity to MEK inhibition.39 There are several other mutations that occur at lower frequency in NSCLC patients, including HER2, MEK, and AKT. However, significant benefit from targeting these mutations has yet to be seen, and is the subject of ongoing clinical trials.

ROS1

ROS1 translocations are a newly discovered driver mutation in NSCLC. They are estimated to occur in approximately 2% of lung adenocarcinoma, and patient characteristics are similar to ALK and EGFR (nonsmokers, lung adenocarcinoma histology, and younger patients).40 Recent preclinical data indicate sensitivity to ALK inhibitors, including crizotinib, and crizotinib was shown to dramatically shrink tumor size in at least 1 patient harboring an ROS1 translocation.40

Squamous Cell Histology

Development of effective targeted treatments for squamous cell histology has lagged behind its adenocarcinoma counterpart, but is now the subject of active investigation. The fibroblast growth factor receptor (FGFR) has been identified as a potential target in squamous cell histology lung cancer. FGFRs are a class of membrane-bound tyrosine kinase receptors that bind to FGF, leading to activation of downstream growth and survival signals. FGFR1 is amplified in approximately 3% of lung adenocarcinoma patients and in 21% of squamous cell lung cancer patients, with inhibition of FGFR1 leading to cell death in preclinical studies.41,42 Early phase trials targeting FGFR with monoclonal antibodies and tyrosine kinase inhibitors are in progress.

The discoidin domain receptor 2 (DDR2) is another promising target in squamous cell histology lung cancer. When activated, DDR2 promotes cell migration, proliferation, and survival.43 It is mutated in approximately 4% of squamous cell lung cancer patients, with preclinical and early clinical data indicating sensitivity to dasatinib (Sprycel, Bristol-Myers Squibb).44 A phase II trial with dasatinib in advanced squamous cell histology lung cancer patients is currently recruiting participants.

Nonvaccine Immunologic Therapies in Metastatic NSCLC

Exciting developments are occurring using immunotherapeutic agents in NSCLC. There are several vaccine studies, mainly in nonmetastatic patients, that have been expertly covered in several recent reviews.45

Ipilumumab (Yervoy, Bristol-Myers Squibb), a fully humanized IgG1 monoclonal antibody, augments T-cell activation, and proliferation by blocking cytotoxic T-lymphocyte associated antigen 4 (CTLA-4), a negative regulator of T-cells.46 It is the first agent to show an overall survival benefit in metastatic melanoma.47 Based on the success of ipilumumab in melanoma, trials in other tumor types, including NSCLC, are in progress. A recent phase II trial showed improved immune-related PFS (irPFS) when ipilumumab was combined with carboplatin and paclitaxel in first-line treatment of NSCLC.48 Statistically significant improvement in irPFS was seen in the sequential arm (HR, 0.686; P=.025) when ipilumumab was added to chemotherapy after the second cycle and not in the concurrent arm. The potential benefit of sequential ipilumumab suggests that T-cell exposure to tumor antigens before ipilumumab administration may improve response.

Another immunotherapeutic agent with an interesting mechanism of action is talactoferrin. Talactoferrin is a recombinant form of human lactoferrin, and is identical to human lactoferrin, except in its glycosylation.49 Interestingly, oral talactoferrin is not systemically absorbed.50 In preclinical studies, following oral administration, talactoferrin is transported into intestinal Peyer’s Patches (aggregated lymphoid nodules), where it recruits circulating dendritic cells,51 inducing a strong, systemic, innate and adaptive immune response, which leads to immune-cell infiltration of distant tumors.52 In a single-agent, phase II trial, oral talactoferrin extended OS in patients with refractory NSCLC by 2.3 months (P<.05, one-tailed log-rank test).53,54 To confirm the phase II data, the trial known as FORTIS-M (A Phase 3, Randomized, Double-blind, Placebo-controlled Study of Oral Talactoferrin in Addition to Best Supportive Care in Patients With Non-small Cell Lung Cancer Who Have Failed Two or More Prior Treatment Regimens) has completed accrual.

Maintenance Therapy

Historically, the standard of care for first-line treatment of metastatic NSCLC has been a platinum doublet for 4–6 cycles, followed by observation until progression. New data with continuation or switch maintenance therapy are challenging that paradigm. The goal of continuation maintenance is to give the same therapy past 4–6 cycles, with the hope that the additional administration of the same therapy will continue to suppress disease (Table 3). Switch maintenance changes therapy to a different second-line agent, with the hope of suppressing disease by delaying resistance to treatment (Table 4).

Continuation Maintenance

In all of the phase III trials of bevacizumab (Avastin, Genentech/Roche) in NSCLC, the anti-VEGF monoclonal antibody has been continued until progression or unacceptable toxicity, even after completion of 4–6 cycles of a platinum doublet. The AvaALL (MO22097) study is a randomized, phase IIIB trial that plans on continuing bevacizumab even after progression. The rationale for continuing bevacizumab beyond progression is based on a large observational cohort study in metastatic colorectal cancer, in which a significant OS benefit was seen in patients who received bevacizumab with chemotherapy beyond progression versus chemotherapy alone.55 The reasons for this benefit are unclear, but this trial seeks to test the benefit of bevacizumab beyond progression in a prospective, randomized fashion in metastatic nonsquamous NSCLC.56

Regarding continuation maintenance for standard chemotherapy, both gemcitabine and pemetrexed have been studied, with mixed data for continuing gemcitabine. In 1 trial, continuing gemcitabine after response or disease stabilization with 4 cycles of cisplatin and gemcitabine showed increased TTP (6.6 months vs 5 months; 3.6 months vs 2 months for the maintenance period; P<.001) but no difference in OS (13 months vs 10 months).57 Another phase III trial of gemcitabine as continuation maintenance after 4 cycles of carboplatin and gemcitabine failed to show a PFS or OS benefit in a phase III trial, but contained a high number of patients with poor performance status.58

The PARAMOUNT (Phase III Study of Maintenance Pemetrexed [pem] Plus Best Supportive Care [bsc] Versus Placebo Plus bsc Immediately Following Induction Treatment With Pem Plus Cisplatin for Advanced Nonsquamous Non-Small Cell Lung Cancer [NSCLC]) trial examined the benefit of continuing pemetrexed after 4 cycles of cisplatin and pemetrexed induction therapy in nonsquamous NSCLC patients. PFS from initiation of maintenance (4.1 months vs 2.8 months; P<.0001) and from start of induction therapy (6.9 months vs 5.59 months; P<.0001) was significantly improved.59 Maintenance pemetrexed was generally well tolerated, and no difference was noted in health-related quality of life between the groups. The OS data have not yet been presented, but were included in the label for pemetrexed in Europe, and an OS benefit was demonstrated with pemetrexed maintenance. The study known as AVAPERL (A Study of Avastin [Bevacizumab] With or Without Pemetrexed as Maintenance Therapy After Avastin in First Line in Patients With Non-Squamous Non-Small Cell Lung Cancer) randomized responding or stable disease patients who received cisplatin, pemetrexed, and bevacizumab to either pemetrexed plus bevacizumab or bevacizumab alone. PFS in patients who received continuation maintenance with pemetrexed and bevacizumab was prolonged compared to bevacizumab alone (10.2 months vs 6.6 months; P<.001), and there was an OS benefit with the combination as well.60 Similarly, the phase III PointBreak (JMHD) trial has completed accrual. This study randomized nonsquamous NSCLC patients to continuation maintenance with either bevacizumab alone or both pemetrexed and bevacizumab after 4 cycles of carboplatin, bevacizumab, and pemetrexed.

Switch Maintenance

Pemetrexed has also been studied in the switch-maintenance setting. Pemetrexed maintenance after 4 cycles of a nonpemetrexed, platinum-based doublet improves PFS (4.3 months vs 2.6 months; P≤.0001) and OS (13.4 months vs 10.6 months; P=.012) compared with placebo.61 Mainly adenocarcinoma histology patients benefited, and no OS benefit was seen in squamous histology patients. A criticism of the data is that 33% of patients randomized to placebo plus best supportive care did not go on to receive second-line treatment upon progression. A phase III Eastern Cooperative Oncology Group (ECOG) trial known as E5508 is ongoing. The trial is randomizing bevacizumab-eligible NSCLC patients to switch maintenance with either bevacizumab or pemetrexed monotherapy versus combined pemetrexed and bevacizumab after completing 4 cycles of carboplatin/paclitaxel/bevacizumab.

Docetaxel, gemcitabine, and erlotinib have also been studied in the switch maintenance setting. Immediate docetaxel compared to delayed docetaxel after frontline carboplatin and gemcitabine resulted in increased PFS of 3 months (5.7 months vs 2.7 months; P=.0001) and a trend towards increased OS (12.3 months vs 9.7 months; P=.085).62 Quality of life scores were similar in both groups. A large portion in the delayed docetaxel group never received second-line therapy (37.2%) compared with the immediate docetaxel group (5.2%). The trend toward an OS benefit is explained by the inferior outcomes of patients who were not able to receive second-line docetaxel upon progression, as OS was nearly identical when comparing only patients who actually received docetaxel in both the immediate and delayed groups.

The benefit of erlotinib in relapsed/refractory disease was shown in the OS improvement in an unselected patient population seen in the BR21 trial that eventually led to the FDA approval of erlotinib in the third-line treatment setting.63 The SATURN trial looked at erlotinib as maintenance therapy in unselected patients after completion of 4 cycles of platinum-based chemotherapy.64 PFS (12.3 weeks vs 11.1 weeks; P<.0001) and OS (12 months vs 11 months; P=.0088) were prolonged in patients who received maintenance erlotinib. Although the benefit of erlotinib was most pronounced in patients who harbored EGFR-activating mutations, a subset analysis revealed a modest benefit in OS that extended to EGFR wild-type patients (HR, 0.77; confidence interval [CI], 0.61–0.97). In a subsequent preplanned analysis, the OS benefit in the SATURN trial was prolonged only in patients with stable disease after first-line chemotherapy (median OS, 11.9 months vs 9.6 months; HR, 0.72; P=.0019), and extended to patients whose tumors were EGFR wild-type.65

Conclusion

Targeted therapy has improved clinical outcomes for patients harboring EGFR activating mutations or ALK translocations. With the identification of substantial molecular heterogeneity in NSCLC, the clinical development of specific therapies against other molecular targets holds great promise. More progress has been made in lung adenocarcinoma histology, but new potential targets in squamous cell lung cancer have emerged. The keys to successful, targeted treatment are determining whether the targeted genetic alterations are driving tumor growth or are passive bystanders, and the continued development of therapies that specifically inhibit oncogene-addicted pathways. The development of new immunotherapies with intriguing mechanisms of action is also under way. Maintenance therapy has become a new treatment paradigm after first-line therapy, and is an appropriate option in selected patients.

References

1. Kris MG, Johnson BE, Kwiatkowski DJ, et al. Identification of driver mutations in tumor specimens from 1,000 patients with lung adenocarcinoma: the NCI’s Lung Cancer Mutation Consortium (LCMC). J Clin Oncol (ASCO Annual Meeting Abstracts). 2011;29: Abstract CRA7506.

2. Sivachenko A, Hammerman Pho N, Cherniak A, Ramos A, Getz G, Meyerson M. Genomic characterization and targeted therapeutics in squamous cell lung cancer. Paper presented at: 14th World Conference on Lung Cancer. July 3-7, 2011; Amsterdam, the Netherlands: Abstract 11.

3. Kim ES, Herbst RS, Wistuba II, et al. The BATTLE trial: personalizing therapy for lung cancer. Cancer Discovery. 2011;1:44-53.

4. Mok TS, Wu YL, Thongprasert S, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 2009;361:947-957.

5. D’Angelo SP, Pietanza MC, Johnson ML, et al. Incidence of EGFR exon 19 deletions and L858R in tumor specimens from men and cigarette smokers with lung adenocarcinomas. J Clin Oncol. 2011;29:2066-2070.

6. 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;362:2380-2388.

7. Rosell R, Carcereny E, Gervais R, et al. 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.

8. Cataldo VD, Gibbons DL, Perez-Soler R, Quintas-Cardama A. Treatment of non-small-cell lung cancer with erlotinib or gefitinib. N Engl J Med. 2011;364:947-955.

9. 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;352:786-792.

10. Engelman JA, Zejnullahu K, Mitsudomi T, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316:1039-1043.

11. Janjigian YY, Groen HJ, Horn L, et al. Activity and tolerability of afatinib (BIBW 2992) and cetuximab in NSCLC patients with acquired resistance to erlotinib or gefitinib. J Clin Oncol (ASCO Annual Meeting Abstracts). 2011;29: Abstract 7525. 

12. Watanabe S, Tanaka J, Ota T, et al. Clinical responses to EGFR-tyrosine kinase inhibitor retreatment in non-small cell lung cancer patients who benefited from prior effective gefitinib therapy: a retrospective analysis. BMC Cancer. 2011;11:1.

13. Herbst RS, Giaccone G, Schiller JH, et al. Gefitinib in combination with paclitaxel and carboplatin in advanced non-small-cell lung cancer: a phase III trial—INTACT 2. J Clin Oncol. 2004;22:785-794.

14. Herbst RS, Prager D, Hermann R, et al. TRIBUTE: a phase III trial of erlotinib hydrochloride (OSI-774) combined with carboplatin and paclitaxel chemotherapy in advanced non-small-cell lung cancer. J Clin Oncol. 2005;23:5892-5899.

15. Chaft JE, Oxnard GR, Sima CS, Kris MG, Miller VA, Riely GJ. Disease flare after tyrosine kinase inhibitor discontinuation in patients with EGFR-mutant lung cancer and acquired resistance to erlotinib or gefitinib: implications for clinical trial design. Clin Cancer Res. 2011;17:6298-6303.

16. Janne PA, Wang XF, Socinski MA, et al. Randomized phase II trial of erlotinib (E) alone or in combination with carboplatin/paclitaxel (CP) in never or light former smokers with advanced lung adenocarcinoma: CALGB 30406. J Clin Oncol (ASCO Annual Meeting Abstracts). 2010;28: Abstract 7503.

17. Pirker R, Pereira JR, Szczesna A, et al. Cetuximab plus chemotherapy in patients with advanced non-small-cell lung cancer (FLEX): an open-label randomised phase III trial. Lancet. 2009;373:1525-1531.

18. Lynch TJ, Patel T, Dreisbach L, et al. Cetuximab and first-line taxane/carboplatin chemotherapy in advanced non-small-cell lung cancer: results of the randomized multicenter phase III trial BMS099. J Clin Oncol. 2010;28:911-917.

19. Gatzemeier U, von Pawel J, Vynnychenko I, et al. First-cycle rash and survival in patients with advanced non-small-cell lung cancer receiving cetuximab in combination with first-line chemotherapy: a subgroup analysis of data from the FLEX phase 3 study. Lancet Oncol. 2011;12:30-37.

20. Pirker R, Pereira JR, von Pawel J, et al. EGFR expression as a predictor of survival for first-line chemotherapy plus cetuximab in patients with advanced non-small-cell lung cancer: analysis of data from the phase 3 FLEX study. Lancet Oncol. 2012;13:33-42.

21. Soda M, Choi YL, Enomoto M, et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature. 2007;448:561-566.

22. Horn L, Pao W. EML4-ALK: honing in on a new target in non-small-cell lung cancer. J Clin Oncol. 2009;27:4232-4235.

23. Kwak EL, Bang YJ, Camidge DR, et al. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med. 2010;363:1693-1703.

24. 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;363:1734-1739.

25. Scagliotti GV, Parikh P, von Pawel J, et al. 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.

26. Camidge DR, Kono SA, Lu X, et al. Anaplastic lymphoma kinase gene rearrangements in non-small cell lung cancer are associated with prolonged progression-free survival on pemetrexed. J Thorac Oncol. 2011;6:774-780.

27. Lee JO, Kim TM, Lee SH, et al. Anaplastic lymphoma kinase translocation: a predictive biomarker of pemetrexed in patients with non-small cell lung cancer. J Thorac Oncol. 2011;6:1474-1480.

28. Boccalatte FE, Voena C, Riganti C, et al. The enzymatic activity of 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/IMP cyclohydrolase is enhanced by NPM-ALK: new insights in ALK-mediated pathogenesis and the treatment of ALCL. Blood. 2009;113:2776-2790.

29. Katayama R, Khan TM, Benes C, et al. Therapeutic strategies to overcome crizotinib resistance in non-small cell lung cancers harboring the fusion oncogene EML4-ALK. Proc Natl Acad Sci U S A. 2011;108:7535-7540.

30. Sequist LV, Gettinger S, Senzer NN, et al. Activity of IPI-504, a novel heat-shock protein 90 inhibitor, in patients with molecularly defined non-small-cell lung cancer. J Clin Oncol. 2010;28:4953-4960.

31. Brugger W, Triller N, Blasinska-Morawiec M, et al. Prospective molecular marker analyses of EGFR and KRAS from a randomized, placebo-controlled study of erlotinib maintenance therapy in advanced non-small-cell lung cancer. J Clin Oncol. 2011;29:4113-4120.

32. Cecchi F, Rabe DC, Bottaro DP. Targeting the HGF/Met signalling pathway in cancer. Eur J Cancer. 2010;46:1260-1270.

33. Spigel DR, Ervin TJ, Ramlau R, et al. Final efficacy results from OAM4558g, a randomized phase II study evaluating MetMAb or placebo in combination with erlotinib in advanced NSCLC. J Clin Oncol (ASCO Annual Meeting Abstracts). 2011;29: Abstract 7505.

34. Pao W, Girard N. New driver mutations in non-small-cell lung cancer. Lancet Oncol. 2011;12:175-180.

35. Kawano O, Sasaki H, Endo K, et al. PIK3CA mutation status in Japanese lung cancer patients. Lung Cancer. 2006;54:209-215.

36. Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949-954.

37. Brose MS, Volpe P, Feldman M, et al. BRAF and RAS mutations in human lung cancer and melanoma. Cancer Res. 2002;62:6997-7000.

38. Marchetti A, Felicioni L, Malatesta S, et al. Clinical features and outcome of patients with non-small-cell lung cancer harboring BRAF mutations. J Clin Oncol. 2011;29:3574-3579.

39. Solit DB, Garraway LA, Pratilas CA, et al. BRAF mutation predicts sensitivity to MEK inhibition. Nature. 2006;439:358-362.

40. Bergethon K, Shaw AT, Ignatius Ou SH, et al. ROS1 Rearrangements Define a Unique Molecular Class of Lung Cancers. J Clin Oncol. 2012;30:863-870.

41. Dutt A, Ramos AH, Hammerman PS, et al. Inhibitor-sensitive FGFR1 amplification in human non-small cell lung cancer. PLoS One. 2011;6:e20351.

42. Weiss J, Sos ML, Seidel D, et al. Frequent and focal FGFR1 amplification associates with therapeutically tractable FGFR1 dependency in squamous cell lung cancer. Sci Transl Med. 2010;2:62ra93.

43. Labrador JP, Azcoitia V, Tuckermann J, et al. The collagen receptor DDR2 regulates proliferation and its elimination leads to dwarfism. EMBO Rep. 2001;2:446-452.

44. Hammerman PS. Mutations in the DDR2 kinase gene identify a novel therapeutic target in squamous cell lung cancer. Cancer Discovery. 2011;1:79-89.

45. Decoster L, Wauters I, Vansteenkiste JF. Vaccination therapy for non-small-cell lung cancer: review of agents in phase III development. Ann Oncol. Dec 12 2011 [epub ahead of print].

46. Fong L, Small EJ. Anti-cytotoxic T-lymphocyte antigen-4 antibody: the first in an emerging class of immunomodulatory antibodies for cancer treatment. J Clin Oncol. 2008;26:5275-5283.

47. Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711-723.

48. Lynch TJ, Bondarenko IN, Luft A, et al. Phase II trial of ipilimumab (IPI) and paclitaxel/carboplatin (P/C) in first-line stage IIIb/IV non-small cell lung cancer (NSCLC). J Clin Oncol (ASCO Annual Meeting Abstracts). 2010;28: Abstract 7531.

49. Ward PP, Piddington CS, Cunningham GA, Zhou X, Wyatt RD, Conneely OM. A system for production of commercial quantities of human lactoferrin: a broad spectrum natural antibiotic. Biotechnology (N Y). 1995;13:498-503.

50. Hayes TG, Falchook GF, Varadhachary GR, et al. Phase I trial of oral talactoferrin alfa in refractory solid tumors. Invest New Drugs. 2006;24:233-240.

51. Wang WP, Iigo M, Sato J, Sekine K, Adachi I, Tsuda H. Activation of intestinal mucosal immunity in tumor-bearing mice by lactoferrin. Jpn J Cancer Res. 2000;91:1022-1027.

52. Kelly RJ, Giaccone G. The role of talactoferrin alpha in the treatment of non-small cell lung cancer. Expert Opin Biol Ther. 2010;10:1379-1386.

53. Parikh PM, Ranade A, Vaid AK, et al. The effect of talactoferrin on overall survival in prognostically important NSCLC subsets in a randomized, placebo-controlled phase II trial. J Clin Oncol (ASCO Annual Meeting Abstracts). 2011;29: Abstract 7569.

54. Parikh PM, Vaid A, Advani SH, et al. Randomized, double-blind, placebo-controlled phase II study of single-agent oral talactoferrin in patients with locally advanced or metastatic non-small-cell lung cancer that progressed after chemotherapy. J Clin Oncol. 2011;29:4129-4136.

55. Grothey A, Sugrue MM, Purdie DM, et al. Bevacizumab beyond first progression is associated with prolonged overall survival in metastatic colorectal cancer: results from a large observational cohort study (BRiTE). J Clin Oncol. 2008;26:5326-5334.

56. Gridelli C, Bennouna J, de Castro J, et al. Randomized phase IIIb trial evaluating the continuation of bevacizumab beyond disease progression in patients with advanced non-squamous non-small-cell lung cancer after first-line treatment with bevacizumab plus platinum-based chemotherapy: treatment rationale and protocol dynamics of the AvaALL (MO22097) trial. Clin Lung Cancer. 2011;12:407-411.

57. Brodowicz T, Krzakowski M, Zwitter M, et al. Cisplatin and gemcitabine first-line chemotherapy followed by maintenance gemcitabine or best supportive care in advanced non-small cell lung cancer: a phase III trial. Lung Cancer. 2006;52:155-163.

58. Belani CP, Waterhouse DM, Ghazal H, et al. Phase III study of maintenance gemcitabine (G) and best supportive care (BSC) versus BSC, following standard combination therapy with gemcitabine-carboplatin (G-Cb) for patients with advanced non-small cell lung cancer (NSCLC). J Clin Oncol (ASCO Annual Meeting Abstracts). 2010;28: abstract 7506.

59. Paz-Ares L, de Marinis F, Dediu M, et al. Maintenance therapy with pemetrexed plus best supportive care versus placebo plus best supportive care after induction therapy with pemetrexed plus cisplatin for advanced non-squamous non-small-cell lung cancer (PARAMOUNT): a double-blind, phase 3, randomised controlled trial. Lancet Oncol. 2012;13:247-255.

60. Barlesi F, de Castro J, Dvornichenko V, et al. Final efficacy outcomes for patients with advanced nonsquamous nonsmall cell lung cancer randomized to continuation maintenance with bevacizumab or bevacizumab plus pemetrexed after first-line bevacizumab-cisplatin-pemetrexed treatment. Paper presented at: the 2011 European Multidisciplinary Cancer Congress; September 23-27, 2011; Stockholm, Sweden: Abstract LBA34.

61. Ciuleanu T, Brodowicz T, Zielinski C, et al. Maintenance pemetrexed plus best supportive care versus placebo plus best supportive care for non-small-cell lung cancer: a randomised, double-blind, phase 3 study. Lancet. 2009;374:1432-1440.

62. Fidias PM, Dakhil SR, Lyss AP, et al. Phase III study of immediate compared with delayed docetaxel after front-line therapy with gemcitabine plus carboplatin in advanced non-small-cell lung cancer. J Clin Oncol. 2009;27:591-598.

63. Shepherd FA, Rodrigues Pereira J, Ciuleanu T, et al. Erlotinib in previously treated non-small-cell lung cancer. N Engl J Med. 2005;353:123-132.

64. Cappuzzo F, Ciuleanu T, Stelmakh L, et al. Erlotinib as maintenance treatment in advanced non-small-cell lung cancer: a multicentre, randomised, placebo-controlled phase 3 study. Lancet Oncol. 2010;11:521-529.

65. Coudert B, Ciuleanu T, Park K, et al. Survival benefit with erlotinib maintenance therapy in patients with advanced non-small-cell lung cancer (NSCLC) according to response to first-line chemotherapy. Ann Oncol. 2012;23:388-394.

66. Sandler A, Gray R, Perry MC, et al. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med. 2006;355:2542-2550.

67. Reck M, von Pawel J, Zatloukal P, et al. Phase III trial of cisplatin plus gemcitabine with either placebo or bevacizumab as first-line therapy for nonsquamous non-small-cell lung cancer: AVAil. J Clin Oncol. 2009;27:1227-1234.