Lung and Mediastinal Tumors


Gene

Alteration

Frequency (%)

Histology

Smoking history frequent patterns

EGFR

Mutation

10–35

Adenocarcinoma, few adenosquamous carcinoma

Frequent in never smokers

KRAS

Mutation

15–25

Mostly in adenocarcinomas, frequently mucinous morphology

Former/current smokers, rarely never smokers

FGFR1

Amplification

20

More frequent in squamous cell carcinoma than adenocarcinoma

Former/current smokers

MET

Amplification

5–20

Acquired resistance to EGFR TKI—adenocarcinoma

Frequent in never smokers

MET

Amplification

2–4

Previously untreated—patients—adenocarcinoma

Frequent in never smokers

PTEN

Mutation

4–8

Squamous cell carcinoma

Ever smokers

DDR2

Mutation

~4

Adenocarcinoma and squamous cell carcinoma

No significant association yet described

ALK

Rearrangement

3–7

Adenocarcinoma, acinar histology, or signet-ring cells

Light smokers (<10 pack years) and/or never smokers

BRAF

Mutation

1–3

Mostly in adenocarcinomas

Former/current smokers

PIK3CA

Mutation

1–3

More frequent in squamous cell carcinoma than adenocarcinoma

Ever smokers and never smokers

NTRK1

Rearrangement

3

Adenocarcinoma

Never smokers

ERBB2 (HER2)

Mutation

2–4

Adenocarcinoma

Never smokers

ROS1

Rearrangement

2

Adenocarcinoma

Light smokers (<10 pack years) and/or never-smokers

AKT1 (AKT)

Mutation

1

Adenocarcinoma and squamous cell carcinoma

No significant association yet described

MAP2K1 (MEK1)

Mutation

1

Mostly in adenocarcinomas than squamous cell carcinoma

No significant association yet described

NRAS

Mutation

1

Adenocarcinoma

Former/current smokers

RET

Rearrangement

1

Adenocarcinoma

Frequent in never smokers



The first randomized trial involving East Asian patients with advanced lung adenocarcinoma (stage IIIB/IV), the Iressa Pan-Asia Study [IPASS], and subsequent five additional trials showed that for patients with an activating EGFR mutation, initial treatment with an EGFR TKI (gefitinib and/or erlotinib) was superior to standard platinum-based chemotherapy [12]. These findings have been a stepping stone for progress in selection of targeted therapy based on molecular genetic alterations in the tumors.

There are certain patterns noted in distribution of recurrent mutations. Never smokers with adenocarcinoma have the highest incidence of EGFR, HER2, ALK, RET, and ROS1 mutations while in smokers, adenocarcinomas with mucinous histology tend to have frequent KRAS mutations [6].

The clinical practice guidelines (CPG) were recently published for Lung Cancer Biomarkers to identify predictive biomarkers of molecular therapy for EGFR and ALK genes by collaborative efforts by College of American Pathologists (CAP), International Association for the Study of Lung Cancer (IASLC), and Association for Molecular Pathologists (AMP). Molecular testing for EGFR and ALK mutations is discussed below in detail. Other mutations that show promise for targeted therapies are currently in various stages of clinical trials and these will be discussed subsequently in this chapter. Next-generation sequencing (NGS) as testing platform is increasingly being used for detecting multiple mutations across many genes with a focused custom panel or a whole exome/whole genome approach. The NGS testing protocols and details are discussed elsewhere in this book. Each assay has their pros and cons and the summary of most widely used molecular testing methods is listed in Table 8.2.


Table 8.2
Types of frequently used molecular methods, mutations detected, and assay performance





























































































































































Methods

Types of mutations

Assay performance

Point mutation (SNVs)

Small insertions, deletions, duplications

Copy number changes (large deletion, insertions, amplification)

Structural alterations—translocations, inversions, etc.

Chemistry—detection

Sensitivity to detect % of mutant tumora

Specificitya

Turnaround timea

Sanger sequencing (traditional or enrichment: LNA, PNA)

XX

XX
 
XX (RNA)

Dideoxy PCR—CE

Low-average

High

2–3 days

Allele-specific PCR

XX
     
Real-time PCR or CE

High

High

1–2 days

Pyrosequencing

XX

X
   
Pyrophosphate detection upon base incorporation

High

High

1–2 days

Single-base extension PCR

XX

X
   
Dideoxy PCR—CE

Average-high

High

2–3 days

Single-base extension PCR-MS

XX

X
   
MALDI-TOF

Average-high

High

2–3 days

MLPA

XX
 
XX
 
Ligation-PCR-CE

Average-low

High

2–3 days

In situ hybridization (FISH/CISH)
   
XX

XX

In situ hybridization—fluorescence microscopy

Low

Low

2–3 days

NGS—Targeted sequencing-amplicon capture

XX

XX
   
Dependent on platform used

Highb

Highb

Days to weeks

NGS—Targeted sequencing-hybridization capture

XX

XX

XX

XX

Dependent on platform used

Highb

Highb

Days to weeks

NGS—whole-exome sequencing

XX

XX

XX

X

Dependent on platform used

Highb

Highb

Weeks to months

NGS—whole-genome sequencing

XX

XX

XX

XX

Dependent on platform used

Highb

Highb

Weeks to months

Array CGH
   
XX
 
Competitive hybridization (FISH), scanningc

Average

Average

1 week to 1 month

Genome-wide SNP array

XX
 
XX
 
Competitive hybridization (FISH), scanningc

Low

Low

Less than 1 week


XX mutations detected reliably, X mutations detected with difficulty

aThe assignments in these columns are subjective and are dependent on the specimen type, tumor content, tests ordered, methodology, the laboratory conducting the tests, etc. The “low (<80 %),” “average (80–95 %),” and “high (>95 %)” are presented to simplify and to compare platforms generally

bDependent on platform used

cOther platforms/methodology available

CE capillary electrophoresis, CGH comparative genomic hybridization, CNV copy number variation, CISH chromogenic in situ hybridization, FISH fluorescence in situ hybridization, LNA locked nucleic acid, MLPA multiplex ligation-dependent probe amplification, MS mass spectrometry, MALDITOF matrix-assisted laser desorption ionization time-of-flight, NGS next-generation sequencing, PCR polymerase chain reaction, PNA peptide nucleic acid, SNP single-nucleotide polymorphism, SNVs single-nucleotide variants

Table 8.2 Adapted from Vnencak-Jones CL, Berger M, Pao W. Types of molecular tumor testing. My Cancer Genome. 2014. http://​www.​mycancergenome.​org/​content/​other/​molecular-medicine/​types-of-molecular-tumor-testing/​ [30] and Katsanis SH, Katsanis N. Molecular genetic testing and the future of clinical genomics. Nat Rev Genet. 2013;14(6):415-26. doi: 10.1038/nrg3493. Review. PubMed PMID: 23681062 [31]

Any applicable drugs currently approved or not approved and in different stages of clinical trials are listed at the end of this chapter in Appendix, in a table generated from Web site http://​www.​mycancergenome.​org/​content/​disease/​lung-cancer/​.


Epidermal Growth Factor Receptor (EGFR)


EGFR (ERBB1) gene, located on chromosome 7p12, encodes for a protein which is one of the four receptor tyrosine kinases belonging to ERBB family receptor tyrosine kinases. Receptor binding with ligands such as epidermal growth factor (EGF), transforming growth factor, alpha (TGFA), amphiregulin (AREG), epithelial mitogen/epigen (EPGN), betacellulin (BTC), epiregulin (EREG), and heparin-binding EGF-like growth factor (HBEGF), triggers homo- and/or heterodimerization of the receptors leading to autophosphorylation on key cytoplasmic residues. This in turn activates complex downstream signaling cascades in at least four major downstream signaling pathways including the RAS-RAF-MEK-ERK, PI3K-AKT, PLCG1-PRRT2 (PLCgamma-PKC), and STATs modules and sometimes, G protein-coupled receptor signaling and NFKB1 (NF-kappa-B) signaling cascade. These activated signaling pathways drive many cellular responses such as changes in gene expression, cytoskeletal rearrangement, and anti-apoptosis and increased cell proliferation. Due to activating mutations in cancer cells, there is a constitutive or sustained active state that leads to pro-survival signaling which increases potential for invasiveness, proliferation, metastasis, and angiogenesis [14].


Mutations


In the USA about 10 % of patients with NSCLC tumors harbor EGFR mutations while the frequency is much higher in East Asia, about 35 % [1517]. The clinicopathologic characteristics of EGFR mutations are as follows: they are more frequently found in tumors from females, and never smokers (defined as less than 100 cigarettes in a patient’s lifetime) with adenocarcinoma histology with particularly lepidic growth pattern. Other histological patterns of adenocarcinoma and some adenosquamous carcinomas may contain EGFR mutations. In addition, they are observed in other subsets of NSCLC, including in former and current smokers [1517]. These mutations in lung adenocarcinoma are mutually exclusive with many other oncogenic mutations like KRAS, BRAF, and ALK gene rearrangement. The mutations occur within EGFR exons 18–21 encoding a portion of the EGFR kinase domain. EGFR mutations are usually heterozygous with frequent gene amplification of the mutant allele [18, 19]. Approximately 90 % of these mutations involve exon 19 (deletions) or exon 21 point mutation (L858R) [20]. These mutations increase the kinase activity of EGFR, leading to hyperactivation of downstream pro-survival signaling pathways [21].

Most of the EGFR mutations in exon 18–21 are predictive of sensitivity towards EGFR TKI such as gefitinib or erlotinib (see Table 8.2). EGFR exon 20 T790M mutation is associated with acquired and in rare cases primary resistance to EGFR TKI. Most exon 20 insertions have been shown to have reduced EGFR TKI response. However, the EGFR A763_Y764insFQEA mutation is an exception and has been associated in retrospective studies with increased EGFR TKI sensitivity [22]. Data remain very limited for some types of insertions. G719 and L861 mutations are considered somewhat less sensitive compared to major exon 19 and 21 mutations. Mutations at E709 and S768 often occur in combination with another of the listed mutations [2329].


Role of the Molecular Tests


The EGFR mutation testing is not used as a diagnostic test for lung adenocarcinoma. Tumors with EGFR mutations in exons 18–21 lead to constitutive activation of the downstream signaling and this is thought to yield more aggressive tumor phenotypes. EGFR mutation testing is recommended for patients being considered TKI therapy. To that effect CPG for EGFR testing were published in 2013.

Briefly, after a systematic review of extensive published data on EGFR-mutated lung cancer and ALK rearrangements in lung cancers, CAP/IASLC/AMP with interest in the diagnosis and management of lung cancer published evidence-based recommendations for the molecular testing of lung cancers for these two predictive biomarkers in a CPG in 2013 [13].


Which Mutations Should be Tested?


Since close to 90 % of the mutations in lung adenocarcinomas have deletions in exon 19 or point mutation in exon 21 (L858R) of EGFR gene, most laboratories offer molecular tests designed to detect these alterations. However, after the publication of EGFR mutations and ALK rearrangements testing guidelines for selecting patients for TKI therapy, the current recommendation for clinical EGFR mutation testing includes ability to detect all individual mutations that have been reported with a frequency of at least 1 % of EGFR-mutated lung adenocarcinomas (Table 8.3 from guidelines). This is not a concern for laboratories offering Sanger sequencing, which provides information on mutations sequenced throughout the exons 18–21. But it is an important consideration in the selection or design of mutation-specific assays.


Table 8.3
EGFR mutations recommended to be tested for selecting patients for EGFR-TKI therapy

































































EGFR exon

EGFR codon

Mutations (amino acid)

Nucleotide substitutions

Approximate % of all EGFR mutations

18

E709

E709K

E709A

E709G E709V E709D E709Q

c.2125G>A

c.2126A>C

c.2126A>G

c.2126A>T

c.2127A>C, c.2127A>T

c.2125G>C

1

G719

G719S G719A

G719C G719D

c.2155G>A

c.2156G>C

c.2155G>T

c.2156G>A

2–5

19

K739

I740

P741

V742

A743

I744

Insertions

18 bp ins
 
1

E746

L747

R748

E749

A750

T751

S752

P753

Deletions

15 bp del

18 bp del

9 bp del

24 bp del

12 bp del
 
45

20

S768

V769

D770

N771

P772

H773

V774

Insertions
 
4–10

S768

S768I

c.2303G>T

1–2

T790

T790M

c.2369C>T

2

21

L858

L858R

L858M

c.2573T>G

c.2572C>A (rare)

40

L861

L861Q

L861R

c.2582T>A,

c.2582T>G

2–5


Table 8.3 adapted from Lindeman NI 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 Mol Diagn. 2013;15(4):415-53. doi: 10.1016/j.jmoldx.2013.03.001. Epub 2013 Apr 4. Erratum in: J Mol Diagn. 2013 Sep;15(5):730. PubMed PMID: 23562183 [13]


When to Test


EGFR molecular testing should be used to select patients for EGFR-targeted TKI therapy. Patients with lung adenocarcinoma should not be excluded from testing on the basis of clinical characteristics. All patients with advanced lung adenocarcinomas at the time of diagnosis irrespective of morphology and grade and/or at the time of recurrence or progression in patients who originally presented with lower stage disease but were not previously tested are eligible for molecular testing. Lower stage tumors may be tested for mutations but the decision to do so should be made locally by each laboratory, in collaboration with its oncology team. Current summary of guideline recommendation is listed in Table 8.4 [13].


Table 8.4
Summary of guideline recommendations [13]





















































































































Section I: When should molecular testing of lung cancers be performed?

Question 1: Which patients should be tested for EGFR mutations and ALK rearrangements?

1.1a: Recommendation: EGFR molecular testing should be used to select patients for EGFR-targeted TKI therapy, and patients with lung adenocarcinoma should not be excluded from testing on the basis of clinical characteristics

1.1b: Recommendation: ALK molecular testing should be used to select patients for ALK-targeted TKI therapy, and patients with lung adenocarcinoma should not be excluded from testing on the basis of clinical characteristics

1.2: Recommendation: In the setting of lung cancer resection specimens, EGFR and ALK testing is recommended for adenocarcinomas and mixed lung cancers with an adenocarcinoma component, regardless of histologic grade. In the setting of fully excised lung cancer specimens, EGFR and ALK testing is not recommended in lung cancers that lack any adenocarcinoma component, such as pure squamous cell carcinomas, pure small-cell carcinomas, or large-cell carcinomas lacking any immunohistochemistry (IHC) evidence of adenocarcinoma differentiation

1.3: Recommendation: In the setting of more limited lung cancer specimens (biopsies, cytology) where an adenocarcinoma component cannot be completely excluded, EGFR and ALK testing may be performed in cases showing squamous or small-cell histology but clinical criteria (e.g., young age, lack of smoking history) may be useful in selecting a subset of these samples for testing

1.4: Recommendation: To determine EGFR and ALK status for initial treatment selection, primary tumors or metastatic lesions are equally suitable for testing

1.5: Expert consensus opinion: For patients with multiple, apparently separate, primary lung adenocarcinomas, each tumor may be tested but testing of multiple different areas within a single tumor is not necessary

Question 2: When should a patient specimen be tested for EGFR mutation or ALK rearrangement?

2.1a: Recommendation: EGFR mutation testing should be ordered at the time of diagnosis for patients presenting with advanced-stage disease (stage IV according to the 7th edition TNM staging system) who are suitable for therapy or at the time of recurrence or progression in patients who originally presented with lower stage disease but were not previously tested

2.1b: Suggestion: ALK rearrangement testing should be ordered at the time of diagnosis for patients presenting with advanced-stage disease (stage IV according to the 7th edition TNM staging system) who are suitable for therapy or at the time of recurrence or progression in patients who originally presented with lower stage disease but were not previously tested

2.2a: Expert consensus opinion: EGFR testing of tumors at diagnosis from patients presenting with stage I, II, or III disease is encouraged but the decision to do so should be made locally by each laboratory, in collaboration with its oncology team

2.2b: Expert consensus opinion: ALK testing of tumors at diagnosis from patients presenting with stage I, II, or III disease is encouraged, but the decision to do so should be made locally by each laboratory, in collaboration with its oncology team

2.3: Recommendation: Tissue should be prioritized for EGFR and ALK testing

Question 3: How rapidly should test results be available?

3.1: Expert consensus opinion: EGFR and ALK results should be available within 2 weeks (10 working days) of receiving the specimen in the testing laboratory

3.2: Expert consensus opinion: Laboratories with average turnaround times beyond 2 weeks need to make available a more rapid test—either in-house or through a reference laboratory—in instances of clinical urgency

3.3: Expert consensus opinion: Laboratory departments should establish processes to ensure that specimens that have a final histopathologic diagnosis are sent to outside molecular pathology laboratories within 3 working days of receiving requests and to intramural molecular pathology laboratories within 24 h

Section II: How should EGFR testing be performed?

Question 4: How should specimens be processed for EGFR mutation testing?

4.1: Expert consensus opinion: Pathologists should use formalin-fixed, paraffin-embedded (FFPE) specimens or fresh, frozen, or alcohol-fixed specimens for PCR-based EGFR mutation tests. Other tissue treatments (e.g., acidic or heavy metal fixatives, or decalcifying solutions) should be avoided in specimens destined for EGFR testing

4.2: Expert consensus opinion: Cytologic samples are also suitable for EGFR and ALK testing, with cell blocks being preferred over smear preparations

Question 5: What are the specimen requirements for EGFR testing?

5.1: Expert consensus opinion: Pathologists should determine the adequacy of specimens for EGFR testing by assessing cancer cell content and DNA quantity and quality

5.2: Expert consensus opinion: Each laboratory should establish the minimum proportion and number of cancer cells needed for mutation detection during validation

5.3: Expert consensus opinion: A pathologist should assess the tumor content of each specimen and either perform or guide a trained technologist to perform microdissection for tumor cell enrichment as needed

Question 6: How should EGFR testing be performed?

6.1: Recommendation: Laboratories may use any validated EGFR testing method with sufficient performance characteristics

6.2: Expert consensus opinion: Laboratories should use EGFR test methods that are able to detect mutations in specimens with at least 50 % cancer cell content, although laboratories are strongly encouraged to use (or have available at an external reference laboratory) more sensitive tests that are able to detect mutations in specimens with as little as 10 % cancer cells

6.3: Expert consensus opinion: Clinical EGFR mutation testing should be able to detect all individual mutations that have been reported with a frequency of at least 1% of EGFR-mutated lung adenocarcinomas

6.4: Recommendation: Immunohistochemistry for total EGFR is not recommended for selection of EGFR TKI therapy

6.5: Recommendation: EGFR copy number analysis (i.e., FISH or CISH) is not recommended for selection of EGFR TKI therapy

Question 7: What Is the role of KRAS analysis in selecting patients for targeted therapy with EGFR TKIs?

7.1: Recommendation: KRAS mutation testing is not recommended as a sole determinant of EGFR TKI therapy

Question 8: What additional testing considerations are important in the setting of secondary or acquired EGFR TKI resistance?

8.1: Recommendation: If a laboratory performs testing on specimens from patients with acquired resistance to EGFR kinase inhibitors, such tests should be able to detect the secondary EGFR T790M mutation in as few as 5% of cells

Section III: How should ALK testing be performed?

Question 9: What methods should be used for ALK testing?

9.1: Recommendation: Laboratories should use an ALK FISH assay using dual-labeled break-apart probes for selecting patients for ALK TKI therapy; ALK immunohistochemistry, if carefully validated, may be considered as a screening methodology to select specimens for ALK FISH testing

9.2: Recommendation: RT-PCR is not recommended as an alternative to FISH for selecting patients for ALK inhibitor therapy

9.3: Expert consensus opinion: A pathologist should be involved in the selection of sections for ALK FISH testing, by assessing tumor architecture, cytology, and specimen quality

9.4: Expert consensus opinion: A pathologist should participate in the interpretation of ALK FISH slides, either by performing the analysis directly or by reviewing the interpretations of cytogeneticists or technologists with specialized training in solid tumor FISH analysis

9.5: Expert consensus opinion: Testing for secondary mutations in ALK associated with acquired resistance to ALK inhibitors is not currently required for clinical management

Section IV: Should other genes be routinely tested in lung adenocarcinoma?

Question 10: Are other molecular markers suitable for testing in lung cancer?

10.1a: Recommendation: Testing for EGFR should be prioritized over other molecular markers in lung adenocarcinoma

10.1b: Suggestion: After EGFR testing, testing for ALK should be prioritized over other proposed molecular markers in lung adenocarcinoma, for which published evidence is insufficient to support testing guideline development at the present time

Section V: How should molecular testing of lung adenocarcinomas be implemented and operationalized?

Question 11: Must all adenocarcinomas be tested for both EGFR and ALK?

11.1: Expert consensus opinion: Laboratories may implement testing algorithms to enhance the efficiency of molecular testing of lung adenocarcinomas, provided the overall turnaround time requirements are met

Question 12: How should EGFR and ALK results be reported?

12.1: Expert consensus opinion: EGFR mutation testing reports and ALK FISH reports should include a results and interpretation section readily understandable by oncologists and by nonspecialist pathologists

Question 13: How should EGFR and ALK testing be validated?

13.1: Expert consensus opinion: EGFR and ALK testing validation should follow the same guidelines as for other molecular diagnostics and FISH tests

Question 14: How should quality assurance be maintained?

14.1: Expert consensus opinion: Laboratories should follow similar quality control and quality assurance policies and procedures for EGFR and ALK testing in lung cancers as for other clinical laboratory assays. In particular, laboratories performing EGFR and ALK testing for TKI therapy should enroll in proficiency testing, if available


Table 8.4 adapted from Lindeman NI 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 Mol Diagn. 2013;15(4):415-53. doi: 10.1016/j.jmoldx.2013.03.001. Epub 2013 Apr 4. Erratum in: J Mol Diagn. 2013 Sep;15(5):730. PubMed PMID: 23562183

CISH chromogenic in situ hybridization, EGFR, epidermal growth factor receptor, FISH fluorescence in situ hybridization, PCR polymerase chain reaction, RT-PCR reverse transcription-polymerase chain reaction, TKI tyrosine kinase inhibitor, TNM tumor, node, metastasis


Sample Requirements


The effects of routine histology processing on any molecular diagnostics applications that rely on PCR are not different for EGFR mutation analysis. EGFR testing can be performed on fresh, frozen, FFPE, or alcohol-fixed specimens. The selection of the specimen to be tested should be performed by pathologists who also determine the adequacy of specimens for EGFR testing by assessing cancer cell content and DNA quantity and quality. Tumor content may be enriched by directing technologists to microdissect the tumor. Every CLIA-certified laboratory offering molecular tests should validate all types of specimen types and processing variations likely to be encountered (i.e., FFPE, fresh, frozen, alcohol fixed), and testing should be performed and reported only on validated specimen types. Other specimen types including tumor samples with inadequate tumor content should be rejected as inadequate or tested at the discretion of the laboratory director, with clear communication on the report that the test was performed on a nonvalidated specimen type.


Brief Overview of Common Platforms Currently Used for EGFR Testing


Laboratories may use any testing method that is validated for EGFR mutation detection with sufficient performance characteristics. These include ability to detect mutations in specimens with at least 50 % cancer cell content, but are encouraged to be able to detect mutations in specimens with as little as 10 % cancer cells. Different methodologies exist in the published literature, each having different advantages and disadvantages. They include Sanger sequencing with and without mutated allele enrichment strategies [e.g., locked nucleic acid (LNA), peptide nucleic acid clamps (PNA)], the amplification refractory mutation system (ARMS-PCR), length analysis, restriction fragment length polymorphism, real-time PCR, high-resolution melting curve analysis, single-base extension genotyping (including mass spectrometry-based genotyping), denaturing high-performance liquid chromatography, and more recently next-generation sequencing platforms. Common testing methodologies are described below.


Length Analysis and Restriction Fragment Length Polymorphism


Deletions in exon 19 of EGFR : Most commonly used assay to detect deletions in exon 19 of EGFR is length analysis of fluorescently labeled PCR products. If there is presence of mutation, the mutant allele is of smaller size than the wild type and therefore can be easily detected by capillary electrophoresis (Fig. 8.1). Different size deletions can be discriminated from wild-type allele by the difference in size of the length of PCR product.

A322102_1_En_8_Fig1_HTML.gif


Fig. 8.1
EGFR exon 19 deletion (15 bp deletion) mutant peak being the shorter allele

EGFR exon 21 point mutation: The 2573 T>G (L858R) mutation creates a new Sau96I restriction site, GGNCC. Incubation and digestion of PCR product with restriction enzyme Sau96I generate an additional shorter fragment, the basis for a PCR restriction fragment length polymorphism (PCR-RFLP) assay design. The digested fluorescently labeled PCR products are then analyzed by capillary electrophoresis [32] (Fig. 8.2). The sensitivity or limit of detection for both the assays is estimated between 3 and 6 %.

A322102_1_En_8_Fig2_HTML.gif


Fig. 8.2
EGFR exon 21 point mutation (L858R) with PCR-RFLP-based assay


Amplification Refractory Mutation System-PCR


ARMS-PCR uses the ability of Taq DNA polymerase effectively distinguishing between a match and a mismatch at the 3′ end of a PCR primer. Specific mutated sequences are selectively amplified with maximum efficiency only when the primer is fully matched, in a background of the sequences that do not carry the mutation including wild-type sequences. Only a low-level background amplification occurs when the 3′ base is mismatched. The fluorescently labeled probes are called scorpions which have fluorophore and quencher in close proximity leading to reduced fluorescence [fluorescence resonance energy transfer (FRET)]. Upon successful PCR, the fluorophore separates leading to increased fluorescence. A commercially available assay detecting 29 mutations in EGFR gene in exons 18–21 is available using this method (Fig. 8.3). The sensitivity for all the mutations except T790M is about 1 %. For T790M, the sensitivity is about 10 %.

A322102_1_En_8_Fig3_HTML.gif


Fig. 8.3
(a) Qiagen RGQ mutation assay: designed to detect 28 mutations associated with TKI sensitivity and one resistance mutation in exon 20 (T790M). (b) ARMS PCR plots showing examples of exon 19 mutation


Sanger Sequencing (SS)


Direct Sanger sequencing has been the traditional method to detect mutations in different genes including EGFR exons 18–21. It is generally accepted that direct sequencing is likely to miss mutations when the tumor cell content is less than 25 % of the sample. Therefore, although a gold standard, SS may not be sensitive enough to detect mutations in a limited tissue sample such as needle biopsies or fine-needle aspiration cytology material. In a limited sample, enrichment strategies for mutant allele using LNA or PNA probe may be necessary for acceptable clinical sensitivity. An example of deletion in exon 19 of EGFR is shown in Fig. 8.4.

A322102_1_En_8_Fig4_HTML.gif


Fig. 8.4
EGFR exon 19 deletion mutation: arrow showing boundary of the deletion displaying overlapping sequences of the shorter allele


Mass Spectrometry: Matrix-Assisted Laser Desorption/Ionization Time-of-Flight Mass Spectrometry


Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) MassARRAY system (Sequenom), originally designed to analyze single-nucleotide polymorphisms in amplified DNA fragments, is also used to detect known somatic point mutations [33]. Genotyping is done by single-allele base extension reaction products that discriminate mutant and wild-type alleles for a given point mutation based on mass differences that are specific to the sequence of the wild-type product and the mutant products as resolved by MALDI-TOF MS [34].

The sensitivity of detection of mutation is generally accepted between 1 and 10 % depending upon plex level of the assay. Example of positive exon 21 L858R mutation is shown in Fig. 8.5. The wild-type and mutant peaks are discriminated based on differences in the masses of the single-base extension with mass-modified dNTPs.

A322102_1_En_8_Fig5_HTML.gif


Fig. 8.5
EGFR exon 21 2573 T>G (L858R) wild-type and mutant alleles highlighted in blue dotted lines. Courtesy: Laetitia Borsu, Ph.D. Assistant Attending, Genomics Project Manager, MSKCC, New York, NY


Significance of Negative Results


Negative results for EGFR indicate lack of clinically significant response to EGFR TKI therapy.


Is the Test Adaptable to Immunohistochemistry?


Immunohistochemistry (IHC) for total EGFR is not recommended for selection of EGFR TKI therapy due to poor correlation between total EGFR expression and mutation status. In addition, response to EGFR TKI does not correlate with total EGFR expression detected by using immunohistochemistry [19, 35, 36]. IHC for phosphorylated EGFR is limited as the stability of phosphorylation status in FFPE material is variable. EGFR mutation-specific monoclonal antibodies against mutant protein, viz. L858R point mutation in exon 21 and 15 bp deletion in exon 19 (most common deletion), have been shown to be sensitive and specific to detect these mutations in the published literature [3739]. But for exon 19 deletions, the sensitivity is reduced to detect other sized deletions. These mutation-specific antibodies could be used to identify patients eligible for EGFR TKI therapy. However, mutation-specific IHC is very insensitive to be used as a stand-alone assay. Also, for negative results, currently there is no algorithm that exists to test for mutations and molecular testing is still needed. In addition, no prospective clinical experience is available for validation. IHC may be an option in small low-cellularity diagnostic specimens with marginal tumor cellularity for DNA-based analysis, but again negative results do not replace mutation testing [40]. Per published guidelines for EGFR testing for selecting patients eligible for EGFR TKI, the body of published data is insufficient to make an evidence-based recommendation regarding the use of EGFR mutation-specific IHC at this time [13].


FISH or CISH Testing for EGFR Amplification


EGFR amplification or polysomy is frequently encountered in patients with lung adenocarcinomas but the response rate with EGFR TKI for these patients is well below the response rate in patients with EGFR mutations (30 vs. 68 %) [35, 36]. Amplification of mutant allele is noted commonly [19] and that is strongly associated with EGFR polysomy/amplification. This association is thought to be related to EGFR TKI response. But in cases where there is discrepancy between EGFR mutation and copy number, the mutation status is better predictor response to EGFR TKI [36]. Therefore, EGFR copy number analysis (FISH or chromogenic in situ hybridization) is not recommended for selection of EGFR TKI therapy [13].


ALK (Anaplastic Lymphoma Receptor Tyrosine Kinase)


ALK gene, located on chromosome 2p23, encodes a receptor tyrosine kinase, which belongs to the insulin receptor superfamily [41]. ALK plays an important role in the development of the brain and exerts its effects on specific neurons in the nervous system. This gene is frequently rearranged with different genes with novel fusions, mutated, or amplified in many tumors including anaplastic large-cell lymphomas, neuroblastoma, and NSCLC [41].


ALK Mutations


In 2007, Soda and co-workers reported an interstitial deletion and inversion within the short arm of chromosome arm 2 that resulted in the formation of a novel fusion gene comprising portions of the echinoderm microtubule-associated protein-like 4 (EML4) gene and the ALK gene in a subset of lung adenocarcinoma [42]. The chimeric fusion of EML4-ALK results in constitutive dimerization leading to oncogenic activity of ALK gene [43]. The original report from Soda et al. showed 7 % positive rate in NSCLC in Japanese patients (5 of 75). Subsequent studies in the USA have shown the gene fusion positive rate in about 2–7 % of all NSCLCs. This gene fusion is more frequent in adenocarcinomas in never smokers or light smokers.

Patients with tumors containing EML4-ALK mutation respond effectively with ALK kinase inhibitors alone or in combination [44, 45]. In a large series ALK rearrangement-positive (about 5 % of 1,500 NSCLC patients screened) patients treated with EGFR inhibitor, crizotinib, showed an overall response rate of 57 %, with 72 % having a PFS of 6 months or greater [46]. The US Food and Drug Administration (FDA) approved crizotinib for advanced-stage, ALK-positive lung cancer in November 2013. This targeted therapy is also recommended by guidelines from professional organizations, including the American Society of Clinical Oncology (ASCO), European Society for Medical Oncology, and National Comprehensive Cancer Network (NCCN). To that effect as described above CAP/IASLC/AMP published evidence-based recommendations for the molecular testing of lung cancers for ALK as second predictive biomarkers in a CPG in 2013 [13]


Molecular Testing for ALK Mutations


ALK gene rearrangement can be detected using RT-PCR, direct Sanger sequencing, fluorescence in situ hybridization (FISH), and immunohistochemistry.


RT-PCR


To detect all fusion products, RT-PCR has limitations as there are many fusion combinations between different exons on EML4 and ALK genes and each needs a separate primer pair. Also, rare other fusion partners with ALK (KIF5BALK, TFGALK [47, 48]) will be missed by RT-PCR as the primers are designed for specific breakpoints in different exons [49]. Thus there are concerns for a higher failure rate of an RNA-based assay in routine FFPE pathology material and RT-PCR is currently not recommended as a first-line diagnostic method for determining ALK fusion status


Immunohistochemistry


IHC is an attractive and cheaper way to detect ALK overexpression as most anatomic pathology departments have IHC laboratory and pathologists use IHC extensively in their routine practise. However, due to low expression of ALK protein in ALK-rearranged lung adenocarcinomas, IHC is not reliable using the same antibody clone compared to anaplastic large-cell lymphoma (mouse monoclonal anti-human CD246, clone ALK1). Recently two rabbit monoclonal anti-human ALK antibodies (clones D5F3 and D9E4) have shown high sensitivity, specificity, and reproducibility compared to ALK FISH. These reports show promising results and IHC assays may potentially facilitate the routine identification of ALK-rearranged lung adenocarcinoma [50]. The published view by CAP/IASLC/AMP based on current literature is that the data is still limited and more studies are needed to recommend developing a specific recommendation on the use of ALK IHC as a sole determinant of ALK TKI therapy [13, 124].


ALK FISH


The commercial availability of a dual-probe “break-apart” (FISH) assay for ALK fusions/rearrangements is the most efficient and consistent way to detect ALK gene rearrangements for clinical use. This is the same ALK FISH dual probe used to detect anaplastic large-cell lymphoma [51]. This commercially available ALK FISH assay is FDA approved to be used as a companion diagnostic assay for selecting patients for crizotinib therapy.


Specimen Requirements


The consensus for specimen adequacy for FISH testing is similar to EGFR testing (see above) and other FISH assays used in molecular genetic laboratories using FFPE tissue sections. Each laboratory is required to validate the FISH assay on internal samples which are expected in routine testing before offering the test for clinical use. These include different fixatives (FFPE, alcohol) and specimen types (surgical biopsy, cytology specimens, etc.). DNA-damaging fixatives, decalcified tissues in general, are not suitable for any molecular assay. A pathologist should assess the adequacy of the tumor proportion in a given specimen and mark the most cellular area for scoring. The minimum tumor cell content/requirement, although similar to EGFR testing, is less stringent as FISH is a direct visualization and scoring assay. Interpretations should be done by a trained pathologist either by directly screening and scoring the FISH slide or by reviewing the results of cytogeneticists or technologist with specialized training in solid tumor FISH analysis.


Assay Interpretation


Negative results have a yellow fusion signal or narrowly split orange/red and green signals using break-apart dual-labeled FISH assay (Fig. 8.6). The most common positive result in lung cancer will result in one separate orange/red and one separate green signal (Fig. 8.7). The separation of orange/red and green signals should be a gap larger than two signal diameters, to be indicative of an ALK gene rearrangement. The germline unaltered ALK region servers as a built-in negative control which stays as a yellow fusion signal or appears as two narrowly split orange/red and green signals. The second positive result is loss of the green 5′ probe with a remaining unpaired 3′ orange/red probe, indicating an unbalanced rearrangement (Fig. 8.8). Minimum of 50 tumor nuclei in a tumor-rich area are recommended for adequate scoring. The tumors are considered positive if 15 % or more of 50 nuclei assessed show the classic split-signal pattern or loss of the green 5′ probe with remaining red 3′ probe [51, 52]. This cutoff is part of the labeling of the FDA-approved commercial FISH assay.

A322102_1_En_8_Fig6_HTML.jpg


Fig. 8.6
Negative for ALK rearrangement. Arrows: Fusion yellow signal. Open arrows: Narrowly split orange/red and green signals interpreted as negative for ALK rearrangement. Note aneuploidy of chromosome 2p (original magnification × 1,000)


A322102_1_En_8_Fig7_HTML.jpg


Fig. 8.7
Positive for ALK rearrangement. Arrows: One separate orange/red and one separate green signal. Open arrows: Fusion yellow signal built in negative control (wild type) (original magnification × 1,000)


A322102_1_En_8_Fig8_HTML.jpg


Fig. 8.8
Positive for ALK rearrangement (single 3′ ALK). Arrows: Loss of the green 5′ probe with remaining red 3′ probe. Open arrows: Fusion signal serving as built in negative control (wild type) (original magnification × 1,000)


Other Gene Mutations That Alter Response to Current Targeted Therapy With EGFR inhibitors or Show Potential For Targeted Therapy in Lung Cancer



KRAS (Kirsten Rat Sarcoma Viral Oncogene Homolog)


KRAS gene, located on chromosome 12p12.1, encodes a protein which is member of the small GTPase superfamily [53]. Ras proteins bind GDP/GTP and possess intrinsic GTPase activity. In many malignancies a single amino acid substitution leads to an activating mutation that results in constitutive activation of the protein leading up-regulation of downstream signaling cascade including MAPK kinase, PI3 kinase signaling [53], etc.


KRAS Mutations


KRAS mutations are seen in 15–25 % of lung adenocarcinomas and are uncommon in squamous cell carcinomas (SCC) [54]. Most mutations are point mutations producing an amino acid substitution at codons 12, 13, or 61 leading to constitutive activation of downstream signaling pathways. KRAS, EGFR, and ALK mutations are mutually exclusive in NSCLC. KRAS mutations are more frequently encountered in adenocarcinoma with mucinous morphology compared to other morphologies and in patients with history of smoking, both former/current smokers and never smokers [55, 56]. Five percent of never smokers with lung adenocarcinoma have been shown to harbor KRAS mutations [57]. In contrast to colorectal adenocarcinomas, in advanced NSCLC, the role of KRAS as a biomarker either as a prognostic or predictive marker is uncertain at this time as very few prospective randomized trials have been completed [55, 58].

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Oct 9, 2016 | Posted by in ONCOLOGY | Comments Off on Lung and Mediastinal Tumors

Full access? Get Clinical Tree

Get Clinical Tree app for offline access