Abstract
Introduction
Tumor genotyping using single gene assays (SGAs) is standard practice in advanced non-small-cell lung cancer (NSCLC). We evaluated how the introduction of next generation sequencing (NGS) into day-to-day clinical practice altered therapeutic decision-making.
Methods
Clinicopathologic data, tumor genotype, and clinical decisions were retrospectively compiled over 6 months following introduction of NGS assay use at our institution in 82 patient-tumor samples (7 by primary NGS, 22 by sequential SGAs followed by NGS, and 53 by SGAs).
Results
SGAs identified abnormalities in 34 samples, and all patients with advanced EGFR-mutated or ALK-rearranged tumors received approved tyrosine kinase inhibitors (TKIs) or were consented for clinical trials. NGS was more commonly requested for EGFR, ALK, and KRAS-negative tumors (p<0.0001). NGS was successful in 24/29 (82.7%) tumors. Of 17 adenocarcinomas (ACs), 11 (7 from patients with ≤15 pack-years of smoking) had abnormalities in a known driver oncogene. This led to a change in decision-making in 8 patients, trial consideration in 6, and off-label TKI use in 2. Of 7 squamous cell (SC) carcinomas, 1 had a driver aberration (FGFR1); 6 had other genomic events (all with TP53 mutations). In no cases were clinical decisions altered (p=0.0538 when compared to ACs).
Conclusions
Targeted NGS can identify a significant number of therapeutically-relevant driver events in lung ACs; particularly in never or light smokers. For SC lung cancers, NGS is less likely to alter current practice. Further research into the cost effectiveness and optimal use of NGS and improved provider training in genomic oncology are warranted.
Keywords: Non-small-cell lung cancer, next generation sequencing, genotype, EGFR, ALK, driver oncogene, MET, ROS1, MET, adenocarcinoma, squamous cell carcinomas
1. INTRODUCTION
The management of advanced non-small-cell lung cancer (NSCLC) is increasingly directed by knowledge of tumor genotype. Expert groups like the College of American Pathologists (CAP), the International Association for the Study of Lung Cancer (IASLC), the Association for Molecular Pathology (AMP), and the American Society of Clinical Oncology (ASCO) now endorse routine testing for epidermal growth factor receptor (EGFR) mutations or anaplastic lymphoma kinase (ALK) rearrangements using rapid single gene assays (SGAs) (1-3). Knowledge of these predictive biomarkers has permitted selective application of tyrosine kinase inhibitors (TKIs) (1-3), with EGFR and ALK TKIs having gained approval from the U.S. Food and Drug Administration (FDA) on the basis of these genomic features (4-9).
The genetic landscape of NSCLC is complex. Oncogenic and/or therapeutically-relevant genomic aberrations include: mutations, amplifications, deletions, and rearrangements/fusions. It is now well established that a significant proportion of lung adenocarcinomas (ACs) harbor mutations in driver oncogenes that can augment “sustained proliferative signaling” - a hallmark feature of tumorigenesis. These include mutations in: v-ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS), Raf murine sarcoma viral oncogene homolog B1 (BRAF), V-erb-b2 erythroblastic leukemia viral oncogene homolog 2 (ERBB2), rearranged during transfection (RET), c-ros oncogene 1 (ROS1), and neurotrophic tyrosine kinase receptor type 1 (NTRK1), among others (10). Squamous cell (SC) lung cancers may instead harbor genomic changes involving: fibroblast growth factor receptor (FGFR) 1/2/3/4, phosphatidylinositol-3-kinase catalytic subunit alpha1 (PIK3CA), and discoidin domain-containing receptor 2 (DDR2), among others (10, 11).
Technological advances have led to the introduction of next generation sequencing (NGS) platforms into the thoracic oncology clinic (10). As opposed to SGAs, NGS permits massive parallel sequencing that affords maximal tumor genomic assessment while using precious tumor samples sparingly (12). These NGS assays offered by Clinical Laboratory Improvement Amendments (CLIA)-certified commercial or academic vendors are usually only feasible when based on a targeted panel of genes (i.e. targeted NGS) that select for the most readily targetable alterations. Many of these NGS assays, especially whole genome and whole exome, require substantial nucleic acid input (250ng-1ug), though some have been optimized to allow results from lower concentrations of deoxyribonucleic acid (DNA) and more recently ribonucleic acid (RNA) from formalin-fixed paraffin-embedded (FFPE) specimens or cytology specimens (12-14).
The feasibility and applicability of NGS in day-to-day clinical practice—as opposed to use in research settings alone—has not been well vetted in the literature to date. We therefore evaluated how the introduction of NGS assays into daily practice altered therapeutic decision-making for a cohort of NSCLCs treated by a multidisciplinary Thoracic Oncology team at this National Cancer Institute (NCI)-affiliated cancer center. In this cohort, NGS was applied as part of routine practice and not in parallel with other academic/commercial efforts, such as assay development or clinical trial screening.
2. METHODS
2.1 Cohort selection and data collection
Patients seen at Beth Israel Deaconess Medical Center (BIDMC, a member of the Dana-Farber/Harvard Cancer Center) with a diagnosis of NSCLC and whose tumors were submitted for either SGA or NGS were identified through an ongoing Institutional Review Board-approved study (15, 16). Patient inclusion was restricted from May 1st to October 31st, 2014 (the 6-month interval since introduction of NGS in clinical NSCLC specimens). Clinical, pathologic, radiographic, and tumor genotyping parameters were collected by retrospective chart extraction and managed using REDCap electronic data capture tools hosted at BIDMC. A review of clinical documentation, clinical trial screening/consent, and anti-cancer therapies administered allowed for determination of clinical decision-making.
2.2 Tumor genotyping
Following routine pathologic diagnosis of NSCLC, tumor material (from surgical specimens, core needle biopsies or cell aspirates/concentrates) in FFPE tissue blocks was submitted for genomic analysis. SGAs (EGFR exon 18-21 mutation analysis, KRAS codon 12 and 13 mutation analysis, ALK fluorescence in situ hybridization [FISH], ROS1 FISH) and NGS were performed as previously described (13-17). Three different NGS assays were used. The first two were performed by an academic medical center (Massachusetts General Hospital; Boston, MA) using an anchored multiplex polymerase chain reaction (AMP) assay that employs a targeted sequencing strategy (13). The first AMP assay (SNaPshot-NGS-V1) evaluates single nucleotide variants (SNV) and insertions/deletions (indels) in genomic DNA using NGS targeting 39 putative oncogenes and tumor suppressor genes (13); this assay was used in 22 of the study cases. The second AMP assay (ALK, RET, ROS1 NGS Gene Fusion Assay) evaluates fusion transcript detection for ALK, ROS1 and RET using genomic RNA (13); this assay was used in 6 of the study cases. The third NGS assay (FoundationOne, Foundation Medicine; Cambridge, MA) interrogates 315 genes as well as introns of 28 genes involved in rearrangements using massively parallel DNA sequencing to characterize base substitutions, short indels, copy number alterations, and selected fusions (14); this assay was used in 2 of the study cases. A CLIA-certified single gene FGFR1 FISH test (Massachusetts General Hospital; Boston, MA) to evaluate copy number of FGFR1 (17) was used in addition to NGS in SC carcinomas; this assay was used in 5 of the study cases.
2.3 Statistical methods
Fisher’s exact test was used to compare categorical variables. All p-values reported were two-sided.
3. RESULTS
3.1 Patient and tumor characteristics
Table 1 illustrates baseline patient and tumor characteristics. The cohort comprised 82 patients, most of whom had stage IV/recurrent disease (72.0%) and AC histology (90.2%).
Table 1.
Baseline characteristics of patients and tumors genotyped over a 6-month period.
| Age at time of tissue acquisition Median (range) |
67 (34-92) |
| Women n (%) Men n (%) |
49 (59.7) 33 (40.3) |
| Race n (%) White Asian Black Other |
66 (80.5) 8 (9.8) 5 (6.1) 3 (3.6) |
| Smoking status n (%) Current smoker Former smoker Never smoker |
27 (32.9) 41 (50.0) 14 (17.1) |
| Stage n (%) I-III IV/recurrent |
23 (28.0) 59 (72.0) |
| Histology n (%) Adenocarcinoma Squamous cell carcinoma NSCLC (NOS) |
74 (90.2) 7 (8.5) 1 (1.2) |
| Type of tissue n (%) Surgical specimen Small biopsy Cytology block from aspirate/fluid |
22 (26.8) 17 (20.7) 43 (52.4) |
| Anatomic site of tissue acquisition n (%) Bone Brain Extra-thoracic lymph node Lung Mediastinal/hilar lymph node Pleura Other |
4 (4.9) 3 (3.6) 5 (6.1) 27 (32.9) 24 (29.3) 12 (14.6) 7 (8.5) |
NSCLC, non-small-cell lung cancer; NOS, not otherwise specified
3.2 SGAs for EGFR/ALK/KRAS/ROS1 and clinical decisions
Figure 1 depicts the clinical use and outcomes of genomic analyses in the 82 patient-tumor samples. SGAs were ordered in 75 tumors. Analyses for abnormalities in EGFR, ALK, KRAS, and ROS1 were successful in: 94.6% (71/75), 96% (72/75), 94.4% (68/72), and 79.7% (55/69), respectively. The increased failure rate of ROS1 testing is noted, raising the possibility of technical issues with the FISH test (rather than inherent lack of tumor material). Of successfully genotyped tumors, abnormalities in EGFR, ALK, KRAS, and ROS1 were found in: 14.0% (10/71), 4.1% (3/72), 30.8% (21/68), and 0% (0/55), respectively. Abnormalities in EGFR, KRAS, and ALK were mutually exclusive—except in one case with a concomitant EGFR exon 19 deletion and positive ALK FISH identified on the same sample.
Figure 1.
Flow chart of genomic analyses over a 6-month period. A.) Single gene assays: results, clinical decisions and trial evaluation. B.) Next generation sequencing (NGS) assays: results, clinical decisions and trial evaluation.
A total of 10 patients with tumors harboring EGFR mutations were identified. Of these, 8 with metastatic AC received an FDA-approved EGFR TKI (erlotinib), with 3 of the 8 consenting for a clinical trial of erlotinib (www.clinicaltrials.gov: NCT00997334). One patient with stage III disease consented for a clinical trial of adjuvant afatinib (NCT01746251). One patient with stage III disease received concurrent chemoradiation, as per evidence-based guidelines.
All 3 patients with ALK-rearranged ACs received an FDA-approved ALK TKI: one received crizotinib, and 2 were consented for a clinical trial evaluating crizotinib versus alectinib (NCT02075840).
The 21 patients with KRAS-mutated ACs received standard therapies as per expert guidelines (3). Of the 16 patients with stage IV disease, 2 were eligible for and were offered a clinical trial (NCT01933932) following progression on standard platinum-doublet therapy.
35 patients had tumors that were negative for abnormalities in EGFR, ALK, and KRAS. None of these patients were offered off-label TKIs, but 3 of these patients consented for clinical trials using cytotoxic chemotherapy as a backbone.
3.3 Use of NGS assays and clinical decisions
NGS assays were ordered in 29 tumors, corresponding to 35.3% of the original cohort. 22 NGS requests (21 ACs, 1 SC carcinoma) were made for tumors that had previously undergone first-pass SGAs. For the remaining 7 NGS requests (1 AC, 6 SC carcinomas), NGS was the lone genomic test requested (Figure 1B). Of the 22 NGS assays that were requested following initial SGAs, nearly all (21/22, 95.4%) came from tumors that had previously tested negative for abnormalities in EGFR, ALK, and KRAS. Conversely, the majority (21/35, 60%) of tumors testing negative for EGFR, ALK, and KRAS on initial SGAs were subsequently sent for NGS (Figure 1A). There were no observed discrepancies between NGS and SGA results. Of the 29 total tumors submitted for NGS, 24 (82.7%) had successful results. The failure rate was higher if the sample had been previously submitted for SGAs (5/22, 22.7%), as compared to a failure rate of 0% (0/7) when NGS was the initial genomic test (p=0.2965).
Of the 24 successful samples undergoing NGS, most (17/24, 70.8%) were of AC histology, the majority of which (11/17, 64.7%) harbored a well-established driver oncogene: 1 EGFR-mutated, 2 ERBB2-mutated, 1 ROS1-rearranged, 1 RET-rearranged, 2 KRAS-mutated, 1 MET-amplified tumor, 2 MET-exon 14 splicing mutated, and 1 MAP2K1-mutated tumor; concurrent TP53 mutations were seen in 40% of cases. Of the 6 tumors without an identifiable principal oncogene mutation, TP53 mutations were most common (66.6%). Details of individual tumor genotyping results and clinical decisions are listed in Table 2.
Table 2.
Clinicopathologic characteristics, next generation sequencing (NGS) results and changes in clinical decisions for cases with successful assays
| Patient no. |
Stage/Sex/Race Smoking/pack- years |
Single gene assay/results |
Prior therapy | NGS assay | “Major” driver(s) | “Other” change(s) | Change in clinical decision |
Clinical decision from NGS results |
|---|---|---|---|---|---|---|---|---|
| Adenocarcinomas | ||||||||
| 1 | IV/F/Asian never/0 |
No/ NA |
Palliative radiotherapy |
SNaPshot NGS V1 + gene fusion assay |
EGFR p.delE746_A750 |
none | Yes | Consented first line erlotinib (clinical trial NCT00997334) |
| 2 | IV/F/Black never/0 |
Yes/ EGFR, ALK, KRAS, ROS1 negative |
Palliative radiotherapy |
SNaPshot NGS V1 |
ERBB2 p.E740_A741insAYV M |
TP53 p. R342* | Yes | Consented clinical trial neratinib (NCT01827267) |
| 3 | IV/F/White former/5 |
Yes/ EGFR, ALK, KRAS negative; ROS1 failed |
Palliative radiotherapy 1st line platinum- pemetrexed |
SNaPshot NGS V1 + gene fusion assay |
ERBB2 p.V747_G748insGSP |
TP53 p.Y220C | Yes | Off label use afatinib |
| 4 | recurrent/M/White former/4 |
Yes/ EGFR, ALK, KRAS negative; ROS1 failed |
Surgery Adjuvant platinum- docetaxel Palliative radiotherapy 1st line platinum- pemetrexed |
SNaPshot NGS V1 + ALK, RET, ROS1 NGS gene fusion assay |
EZH-ROS1 fusion | none | Yes | Recommended clinical trial crizotinib (NCT00585195) at progression; not consented |
| 5 | IV/F/Asian never/0 |
Yes/ EGFR, ALK, KRAS negative; ROS1 not performed |
1st line platinum- paclitaxel 2nd line docetaxel |
Foundation One |
KIF5B-RET fusion | PTEN_D326fs*18 | Yes | Consented clinical trial sunitinib (NCT01829217) |
| 6 | IV/F/White former/12 |
Yes/ EGFR, ALK, KRAS, ROS1 negative |
1st line platinum- pemetrexed |
Foundation One |
MET amplification | GRIN2A p.F183I KDM5C p.P380fs*50 PBRM p.R1010* NKX2-1 amplification |
Yes | Consented clinical trial crizotinib (NCT00585195) |
| 7 | IV/F/White former/24 |
Yes/ EGFR, ALK, KRAS, ROS1 negative |
Palliative radiotherapy 1st line platinum- pemetrexed |
SNaPshot NGS V1 + gene fusion assay |
MET p.D1010_splice exon 14 PIK3CA p.E542K |
TP53 p.R337L | Yes | No immediate change in therapy. Consideration of off label crizotinib at progression |
| 8 | IV/M/White former/16 |
Yes/ EGFR, ALK, KRAS negative; ROS1 failed |
Palliative radiotherapy 1st line platinum- pemetrexed |
SNaPshot NGS V1 |
KRAS p.Q61H | none | Yes | No immediate change in therapy. Consideration of clinical trial selumetinib (NCT01933932) at progression |
| 9 | IV/F/White former/30 |
Yes/ EGFR, ALK, negative; KRAS p. G12C; ROS1 not performed |
Palliative radiotherapy 1st line platinum- paclitaxel 2nd line pemetrexed |
SNaPshot NGS V1 |
KRAS p.G12C | TP53 p.A159P | No | No clinical recommendation |
| 10 | IV/F/White former/25 |
Yes/ EGFR, ALK, KRAS, ROS1 negative |
1st line platinum- pemetrexed 2nd line docetaxel |
SNaPshot NGS V1 |
MAP2K1 p.K57N | STK11 p.T308C | No | No clinical recommendation |
| 11 | II/F/White never/0 |
Yes/ EGFR, ALK, KRAS, ROS1 negative |
Surgery Adjuvant platinum- pemetrexed |
SNaPshot NGS V1 + gene fusion assay |
MET p.D1010N _splice exon 14 |
none | No | No clinical recommendation |
| 12 | IV/F/White never/0 |
Yes/ EGFR, ALK, KRAS, ROS1 negative |
1st line platinum- paclitaxel 2nd line docetaxel 3rd line pemetrexed |
SNaPshot NGS V1 |
none | MET p.D981V GNA11 p.L229F |
No | No clinical recommendation |
| 13 | IV/F/White current/40 |
Yes/ EGFR, ALK, KRAS, ROS1 negative |
Best supportive measures |
SNaPshot NGS V1 |
none | TP53 p.G226* | No | No clinical recommendation |
| 14 | recurrent/M/White former/40 |
Yes/ EGFR, ALK, KRAS, ROS1 negative |
Surgery | SNaPshot NGS V1 |
none | CDH1 p.D805N CDKN2A p.H123Q |
No | No clinical recommendation |
| 15 | IV/M/White current/50 |
Yes/ EGFR, ALK, KRAS negative; ROS1 failed |
1st line platinum- paclitaxel 2nd line docetaxel |
SNaPshot NGS V1 |
none | TP53 p.F134L MET p.T992I SMAD4_ splice |
No | No clinical recommendation |
| 16 | II/M/Asian current/10 |
Yes/ EGFR, ALK, KRAS, ROS1 negative |
Surgery | SNaPshot NGS V1 |
none | TP53 p.R273C | No | No clinical recommendation |
| 17 | II/M/White former/40 |
Yes/ EGFR, ALK, KRAS, ROS1 negative |
Surgery | SNaPshot NGS V1 |
none | TP53 p.R273L STK11_splice FGFR1 p.D132A (SNP) |
No | No clinical recommendation |
| Squamous cell carcinomas | ||||||||
| 18 | III/F/White/ former/40 |
No/ NA |
Definitive radiotherapy concurrent platinum- paclitaxel |
SNaPshot NGS V1 + FGFR1 FISH |
FGFR1 amplification (borderline) |
TP53 p.R283P | No | No clinical recommendation |
| 19 | Recurrent/M/White former/60 |
No/ NA |
Surgery Palliative radiotherapy |
SNaPshot NGS V1 + FGFR1 FISH |
none | TP53 p.R181P STK11 p.G187Vfs* |
No | No clinical recommendation |
| 20 | IV/M/White former/80 |
No/ NA |
Palliative radiotherapy 1st line platinum- paditaxel |
SNaPshot NGS V1 + FGFR1 FISH |
none | TP53 p.L130V | No | No clinical recommendation Subsequently consented clinical trial docetaxel (NCT01750281) |
| 21 | IV/M/White current/50 |
No/ NA |
Palliative radiotherapy 1st line platinum- paclitaxel |
SNaPshot NGS V1 + FGFR1 FISH |
none | TP53 p.P36Afs*7 APC p.T1218M NOTCH1 p.Q1476S |
No | No clinical recommendation |
| 22 | IV/M/White current/90 |
No/ NA |
1st line platinum- paclitaxel |
SNaPshot NGS V1 + FGFR1 FISH |
none | TP53 p.L257G | No | No clinical recommendation |
| 23 | IV/M/White never/0 |
Yes/ EGFR, ALKnegative; KRAS, ROS1 not performed |
1st line platinum- gemcitabine 2nd line docetaxel |
SNaPshot NGS V1 + gene fusion assay |
none | TP53 p.delV157_ M160 |
No | No clinical recommendation |
| 24 | I/M/White former/30 |
No/ NA |
Surgery | SNaPshot NGS V1 |
none | TP53 p. H178D | No | No clinical recommendation |
Clinical decisions were impacted by NGS results in 8/17 (47.0%) ACs. A total of 6 patients were considered for clinical trials available at our institution on the basis of findings from NGS: 4 consented for a clinical trial of an oncogene-specific TKI, and 2 were offered a genotype-specific trial upon subsequent progression. One patient with an ERBB2-mutated AC received off-label afatinib (a clinical trial for this genotype had closed to accrual). Another patient with a MET exon 14 splicing mutation was offered off-label crizotinib at first progression. While not FDA-approved for the latter two indications, these TKIs are acknowledged as targeted therapies for consideration as per the National Comprehensive Cancer Network (NCCN) guidelines for NSCLC (3).
The results of NGS assays were of lesser therapeutic consequence in the 7 SC carcinomas, where all tumors harbored TP53 mutations but lacked the prevalent/principal driver oncogenes observed in ACs (1/7 or 14.2% vs. 10/17 or 58.8% in SC vs. AC, respectively; p=0.0778). Only 1 case of borderline FGFR1 amplification was identified, but this did not lead to a change in clinical decision-making as the clinical trial of an FGFR inhibitor (NCT01703481) available at our institution at the time was not applicable for this patient; this patient received concurrent chemoradiation as per expert guidelines. Therefore, in none of the cases of SC carcinoma were clinical decisions altered by NGS results—a proportion that was notably lower than that observed in those patients with AC (0% or 0/7 vs. 47.0% or 8/17 for SC vs. AC, respectively; p=0.0538). The therapeutic utility of tumor genotyping stratified by histology is indicated in Figure 2 and Table 2.
Figure 2.
Genomic abnormalities stratified by histology and patient smoking status. A.) Genomic findings and targeted therapies in adenocarcinomas, as identified by next generation sequencing (NGS). B.) Genomic findings and targeted therapies in squamous cell carcinomas, as identified by NGS. C.) Genomic findings and targeted therapies in adenocarcinomas from never-/light- smokers [≤ 15 pack-years smoking]. D.) Genomic findings and targeted therapies in adenocarcinomas from patients with > 15 pack-years smoking.
3.4 Alterations in driver oncogenes in lung ACs from never or light smokers
Driver oncogenes - for which TKIs are approved or have some level of clinical evidence (i.e. EGFR, ALK, ROS1, RET, ERBB2, MET) - identified by combined SGAs and/or NGS (Figure 2) were significantly more frequent in patients with a ≤ 15 pack-year tobacco history (18/27, 66.6%) than in those with > 15 pack-years of tobacco use (3/42, 7.1%; p<0.0001). KRAS mutations were more frequent in the group with > 15 pack-years of tobacco use (17/42 or 40.4% vs. 5/27 or 18.5%, respectively; p=0.0682).
4. DISCUSSION
The care of advanced NSCLC has evolved dramatically in the last decade, owing to the evolution of targeted, biologically pertinent therapies offered on the basis of vetted genomic biomarkers. When used in these genotype-specific settings, targeted therapies have afforded consistent improvements in progression-free survival and quality of life, as compared to conventional cytotoxic agents (4-10). Unsurprisingly, oncology patients and providers in academic and community settings alike are increasingly aware of, desire, and are incumbent to obtain timely and accurate tumor genomic testing to help guide therapeutic decision-making (18, 19). It is also now apparent that the genomic landscape of NSCLC extends beyond EGFR and ALK alone, with potentially relevant aberrations also found in: KRAS, BRAF, ERBB2, RET, ROS1, NTRK1, FGFR 1/2/3/4, PIK3CA, and DDR2, among others (10, 11).
4.1 Outcomes of NGS use in NSCLC by other groups
The clinical utility of comprehensive genomic analyses of lung ACs has been recently highlighted by the Lung Cancer Mutation Consortium (LCMC), which analyzed 10 potential genomic drivers from tumor specimens obtained from academic centers nationwide (20). An oncogenic driver was found in 64% of tumors; these results were subsequently used to select a precision therapy or clinical trial in 28% of cases. Notably, median survival was improved for those patients whose tumors harbored an abnormality amenable to a genotype-directed therapy (20). The Washington University lung cancer program recently published their experience with an internally-developed 23-gene NGS assay (21). A 45% assay failure rate was noted; however, NGS afforded actionable information in 46% of successfully assayed NSCLCs, and a targeted therapy was instituted in 11% of genotyped tumors (21). Similarly, colleagues at Memorial Sloan Kettering used hybrid-capture-based NGS testing (FoundationOne) following serial negative SGAs in patients with AC and light/no (≤15 pack-year) tobacco use (22). 34% of patients were excluded on the basis of tissue exhaustion from prior testing. Actionable genomic changes were identified in 39% of patients (using targeted therapies on/off study) (22). We are unaware of other peer-reviewed publications detailing the clinical application of NGS in day-to-day practice in a multidisciplinary NSCLC clinic.
4.2 NGS in EGFR, ALK, KRAS-wild type tumors
In the cohort presented here, NGS was used in greater than one-third of cases and in a rationally selected subset with tumors most likely to harbor changes that may render genotype-directed therapy of use, i.e.: patients without other common driver mutations (EGFR, ALK, and KRAS) and/or those with light/no tobacco use (≤15 pack-years). This cohort highlights the application of NGS in routine clinical practice and not in parallel with other efforts (i.e. assay development or clinical trial screening) (20-22). Therefore, it serves as an instructive platform for assessing how this technology is being adopted, implemented, and applied in daily care.
4.3 Pearls and pitfalls of NGS use
Important limitations of this analysis include: its retrospective nature, the limited sample size, utilization of archival tissue, and absence of direct involvement in NGS analysis/interpretation. Moreover, we currently have no pre-specified algorithm to direct obtaining and integrating genomic results into patient care, beyond application of approved targeted therapies in selected populations.
NGS was successfully executed in the vast majority of submitted samples (>80%). As observed by our group and others, driver oncogenes with known therapeutic potential were more readily identified in those with AC vs. SC histology. NGS of ACs impacted real-world decision-making in nearly half of cases, enabling genotype-specific therapies. In most ACs in which a driver mutation was identified and a relevant clinical trial available/appropriate, enrollment was offered. We favor this strategy for integrating genomic data into patient care, so as to drive participation in genotype-directed trials and facilitate rational and rigorous approval of novel therapies and targeted applications. During our data collection period, our center had active protocols for tumors with aberrations in EGFR, ALK, ROS1, MET, KRAS and ERBB2.
4.4 NGS use in SC carcinomas
Genomic results (NGS or SGAs) for SC carcinomas did not lead to practice changes in our cohort. The number of cases analyzed was small, and additional numbers would have likely revealed tumors with PI3KCA, DDR2, and FGFR aberrations; these have been described by other groups as potentially targetable in preclinical models (10, 11). However, the absence of known bona fide driver oncogenes in the majority of SC carcinomas (10) has made the evolution of targeted therapies for this histologic subtype much slower. To date, no TKI is approved by regulatory agencies or recommended by the NCCN for SC carcinomas (3). This unmet clinical need has led to development of novel biomarker-driven trials. The National Clinical Trials Network (NCTN) has recently started the LUNG-Map/S1400 (NCT02154490) trial to address this need (http://www.lung-map.org/). This is a multi-armed randomized phase II/III trial emphasizing matched gene-drug strategies using the FoundationOne platform for NGS. If LUNG-Map is successful, then NGS and novel targeted drugs may become part of the standard approach to managing SC lung cancers, as well.
4.5 Optimizing NGS use in day-to-day practice
It is speculative to consider whether if used as a sole method, NGS may have allowed for detection of all clinically-relevant genomic alterations, with a higher success rate, and without the time, effort, cost, and tissue exhaustion associated with sequential testing. As have others, we note that there was a trend toward more frequent NGS failure rates in samples where SGAs preceded. These factors underscore the critical importance of timely and effective collaboration between members of the multidisciplinary team to obtain, select, and retain optimal specimen(s) for effective testing. Enthusiasm for the routine use of up-front NGS should be tempered by consideration of the financial, medical, and psychosocial costs and cost effectiveness of this approach. This includes consideration of “costs” associated with the assay itself, subsequent anti-cancer medication(s), additional tissue acquisition procedures, therapeutic delays pending tissue testing, and inadequately known benefits/risks for the patient. Such efforts should occur in parallel with the development and application of these genomic platforms. It is an urgent need to establish the optimal use of genomic testing (i.e. sequential SGAs versus NGS) in the day-to-day care of NSCLC patients.
The evolution of genotype-directed cancer care also has implications for the education of medical professionals. A recent publication testifies to this unmet need: in a study of 160 faculty members at a well-known academic cancer center, higher “genomic confidence” was associated with a higher likelihood that a provider would obtain and apply genomic data to patient care; 22% of surveyed cancer providers indicated low confidence in their genomic knowledge (19). Targeted areas for genomic oncology curricula should include: methodologies for genomic assessment (i.e. understanding what methods are specifically utilized by commercial/institutional genomic assay vendors); optimal/adequate specimen selection; utilization of validated resources for interpretation of results; understanding the therapeutic implications—or lack thereof—of a given finding; obtaining informed consent for testing from patients; and discussing results of testing with patients—including findings of uncertain significance. In recognition of this evolving and increasingly apparent educational need, colleagues have already started to take systematic approaches to educate trainees in this important domain (23-25).
5. CONCLUSIONS
In summary, we report the patterns of use and implications of NGS testing in an academic thoracic oncology clinic. Sequential single gene and targeted NGS assays can identify a significant number of confirmed driver events in lung ACs for which targeted therapies are either already available or in development. NGS is particularly impactful in never/light smokers and in whom first pass SGAs do not identify any targetable abnormalities, as this subset is likely to be enriched with oncogenic targets that are of therapeutic relevance. In SC lung cancers, however, the impact of NGS in standard practice remains unclear, and innovative biomarker-driven trials have commenced to address this unmet need. The growth of platforms that permit rapid tumor genotyping has fueled a furtive interest in the development and application of personalized approaches in cancer therapeutics. Comprehensive first line genotyping of NSCLCs using NGS is therefore an attractive extension of these efforts. However, future research into the cost and relative impact/effectiveness of this approach, as well as improved training of medical professionals in the practice of genomic oncology, are warranted to maximize this strategy of care.
CLINICAL PRACTICE POINTS.
- Tumor genotyping using single gene assays for EGFR mutation and ALK rearrangementare standard practice in advanced non-small-cell lung cancer;
- Targeted next generation sequencing can identify a significant number of therapeutically-relevant driver events in lung adenocarcinoma; particularly in never or light smokers;
- The introduction of next generation sequencing/comprehensive molecular profiling into day-to-day clinical practice altered therapeutic decision-making and increased the number of cases that can benefit from precision oncology;
- Further research into the cost effectiveness and optimal use of next generation sequencing and improved provider training in genomic oncology are warranted.
ACKNOLWEDGMENTS
Funding/Grant Support: This work was funded in part through a fellowship from the American Society of Clinical Oncology Conquer Cancer Foundation (DBC), an American Cancer Society grant (RSG 11-186 to DBC), a Lung Cancer Foundation of America-International Association for the Study of Lung Cancer grant (to DBC), and National Institutes of Health (NIH) grant CA090578 (to DBC).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
CONFLICT OF INTEREST STATEMENT
Daniel B. Costa has received consulting fees from Pfizer and honoraria fromBoehringerIngelheim.
Deepa Rangachari, Paul A. VanderLaan, Xiuning Le, Erik Folch, Michael S. Kent, Sidharta P. Gangadharan, Adnan Majid, Richard L. Haspel, Loren J. Joseph and Mark S. Huberman have no conflicts to disclose.
No other conflict of interest is stated.
REFERENCES
- 1.Lindeman NI, Cagle PT, Beasley MB, et al. Molecular Testing Guideline for Selection of Lung Cancer Patients for EGFR and ALK Tyrosine Kinase Inhibitors: Guideline from the College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology. J Thorac Oncol. 2013;8:823–859. doi: 10.1097/JTO.0b013e318290868f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Leighl NB, Rekhtman N, Biermann WA, et al. Molecular testing for selection of patients with lung cancer for epidermal growth factor receptor and anaplastic lymphoma kinase tyrosine kinase inhibitors: american society of clinical oncology endorsement of the college of american pathologists/international association for the study of lung cancer/association for molecular pathology guideline. J Clin Oncol. 2014;32:3673–3679. doi: 10.1200/JCO.2014.57.3055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Ettinger DS, Akerley W, Borghaei H, et al. Non-small cell lung cancer, version 2.2013. J Natl Compr Canc Netw. 2013;11:645–653. doi: 10.6004/jnccn.2013.0084. [DOI] [PubMed] [Google Scholar]
- 4.Jorge SE, Kobayashi SS, Costa DB. Epidermal growth factor receptor (EGFR) mutations in lung cancer: preclinical and clinical data. Braz J Med Biol Res. 2014;47:929–939. doi: 10.1590/1414-431X20144099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.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. doi: 10.1016/S1470-2045(11)70393-X. [DOI] [PubMed] [Google Scholar]
- 6.Sequist LV, Yang JC, Yamamoto N, et al. Phase III Study of Afatinib or Cisplatin Plus Pemetrexed in Patients With Metastatic Lung Adenocarcinoma With EGFR Mutations. J Clin Oncol. 2013;31:3327–3334. doi: 10.1200/JCO.2012.44.2806. [DOI] [PubMed] [Google Scholar]
- 7.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. doi: 10.1056/NEJMoa1006448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Shaw AT, Kim DW, Nakagawa K, et al. Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med. 2013;368:2385–2394. doi: 10.1056/NEJMoa1214886. [DOI] [PubMed] [Google Scholar]
- 9.Shaw AT, Kim DW, Mehra R, et al. Ceritinib in ALK-rearranged non-small-cell lung cancer. N Engl J Med. 2014;370:1189–1197. doi: 10.1056/NEJMoa1311107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Gerber DE, Gandhi L, Costa DB. Management and future directions in non-small cell lung cancer with known activating mutations. Am Soc Clin Oncol Educ Book. 2014:e353–e365. doi: 10.14694/EdBook_AM.2014.34.e353. [DOI] [PubMed] [Google Scholar]
- 11.Hammerman PS. Cancer Genome Atlas Research Network: Comprehensive genomic characterization of squamous cell lung cancers. Nature. 2012;489:519–525. doi: 10.1038/nature11404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Yatabe Y, Thomas RK. Era of Comprehensive Cancer Genome Analyses. J Clin Oncol. 2014 doi: 10.1200/JCO.2014.58.4375. [DOI] [PubMed] [Google Scholar]
- 13.Zheng Z, Liebers M, Zhelyazkova B, et al. Anchored multiplex PCR for targeted next-generation sequencing. Nat Med. 2014 doi: 10.1038/nm.3729. [DOI] [PubMed] [Google Scholar]
- 14.Frampton GM, Fichtenholtz A, Otto GA, et al. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol. 2013;31:1023–1031. doi: 10.1038/nbt.2696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.VanderLaan PA, Yamaguchi N, Folch E, et al. Success and failure rates of tumor genotyping techniques in routine pathological samples with non-small-cell lung cancer. Lung Cancer. 2014;84:39–44. doi: 10.1016/j.lungcan.2014.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Yamaguchi N, VanderLaan PA, Folch E, et al. Smoking status and self-reported race affect the frequency of clinically relevant oncogenic alterations in non-small-cell lung cancers at a United States-based academic medical practice. Lung Cancer. 2013;82:31–37. doi: 10.1016/j.lungcan.2013.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Heist RS, Mino-Kenudson M, Sequist LV, et al. FGFR1 amplification in squamous cell carcinoma of the lung. J Thorac Oncol. 2012;7:1775–1780. doi: 10.1097/JTO.0b013e31826aed28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Gray SW, Hicks-Courant K, Lathan CS, et al. Attitudes of patients with cancer about personalized medicine and somatic genetic testing. J Oncol Pract. 2012;8:329–35. doi: 10.1200/JOP.2012.000626. 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Gray SW, Hicks-Courant K, Cronin A, et al. Physicians' attitudes about multiplex tumor genomic testing. J Clin Oncol. 2014;32:1317–1323. doi: 10.1200/JCO.2013.52.4298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Kris MG, Johnson BE, Berry LD, et al. Using multiplexed assays of oncogenic drivers in lung cancers to select targeted drugs. JAMA. 2014;311:1998–2006. doi: 10.1001/jama.2014.3741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Hagemann IS, Devarakonda S, Lockwood CM, et al. Clinical next-generation sequencing in patients with non-small cell lung cancer. Cancer. 2014 doi: 10.1002/cncr.29089. [DOI] [PubMed] [Google Scholar]
- 22.Drilon A, Wang L, Arcila ME, et al. Broad, hybrid capture-based next-generation sequencing identifies actionable genomic alterations in "driver-negative" lung adenocarcinomas. Clin Cancer Res. 2015 doi: 10.1158/1078-0432.CCR-14-2683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Haspel RL, Saffitz JE. Genomic oncology education: an urgent need, a new approach. Cancer J. 2014;20:91–95. doi: 10.1097/PPO.0000000000000015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Haspel RL, Olsen RJ, Berry A, et al. Progress and potential: training in genomic pathology. Arch Pathol Lab Med. 2014;138:498–504. doi: 10.5858/arpa.2013-0359-SA. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Haspel RL, Rinder HM, Frank KM, et al. The current state of resident training in genomic pathology: a comprehensive analysis using the resident in-service examination. Am J Clin Pathol. 2014;142:445–451. doi: 10.1309/AJCPH2A4XTXJUKDZ. [DOI] [PMC free article] [PubMed] [Google Scholar]


