xmlns="http://www.w3.org/TR/REC-html40"> Ken’s Pathology Guide

 

Adenocarcinoma of the Lung

 

 

Epidemiology:

    • Risk factors:
      • Smoking (75% in smokers)
      • High-dose ionizing radiation (uranium)
      • Asbestos (particularly in smokers)
      • Radon
    • Most common type of lung cancer in women and nonsmokers
      • Although 75% of adenocarcinomas are found in smokers

 

Common sites:

    • Peripheral location in the lung
    • Involve the pleura commonly

 

Gross features:

    • Gray-yellow
    • Cavitation is rare
    • Broncholoalveolar carcinoma (now AIS and invasive mucinous carcinoma):
      • Almost always in periphery of lung
      • Mucinous type is more likely to be multicentric and may grossly show mucin
      • Often multiple diffuse, soft, gray nodules coalescing to form a pneumonia-like consolidation
      • May have a mucinous, gray translucence

 

Histologic features:

    • glandular differentiation
    • mucin production (80%)
    • most show mixed patterns (formerly called “mixed subtype”)
    • classification of invasive ADC by predominant pattern:
      • lepidic (formerly mixed subtype with nonmucinous BAC)
      • acinar
      • papillary
      • solid (poor prognosis)
        • poorly differentiated
        • If 100% solid, need to see intracellular mucin
          • should be present within at least 5 cells in at least 2 high-power fields
          • mucicarmine or PASD positive
      • micropapillary (poor prognosis)
    • variants:
      • invasive mucinous adenocarcinoma (formerly mucinous BAC)
      • colloid adenocarcinoma
      • fetal adenocarcinoma
      • enteric adenocarcinoma
    • types / growth patterns:
      • acinar
      • often bronchioloalveolar pattern of spread at the periphery of the tumor
      • papillary type shows papillary architecture similar to other papillary carcinomas
        • >25% papillary architecture is an unfavorable prognostic factor
      • mixed
    • Adenocarcinoma in situ (AIS) (<= 3 cm by definition) (formerly bronchioloalveolar BAC type):
      • pure bronchioloalveolar growth pattern with no evidence of stromal, vascular, or pleural invasion
      • lepidic pattern – growth along preexisting structures with preservation of alveolar architecture
      • often grows around the edge of a focal scar
        • smaller scar is better prognosis
      • usually non-mucinous (rarely mucinous)
      • mucinous subtype (rare – most are now classified as invasive mucinous adenocarcinoma):
        • tend to be multicentric (?now invasive mucinous adenocarcinoma)
        • tall columnar cells
        • abundant apical cytoplasmic mucin
        • small basally oriented nuclei
        • airspaces often filled with mucin
      • non-mucinous subtype:
        • tend to be solitary
        • Clara cells or tye II pneumocytes
        • cuboidal cells with a hobnail or saw-toothed appearance often
        • nuclear inclusions (50% of cases)
          • PAS positive
          • Surfactant apoprotein positive immunohistochemically
          • EM – network of 40nm branching microtubules thought to arise from inner nuclear membrane
      • Mixed mucinous and non-mucinous type
      • note that metastatic adenocarcinomas to the lung can mimic this pattern, particularly mucinous subtype
    • minimally invasive adenocarcinoma (MIA) (<= 3 cm by definition)
      • lepidic pattern predominantly, with <= 5mm invasion
      • non-mucinous predomninantly (rarely mucinous)
    • invasive mucinous adenocarcinoma (formerly mucinous BAC)
      • goblet or columnar cells
      • abundant intracellular mucin
        • note that mucin production does not make the diagnosis – needs to be goblet or columnar cells
      • patterns may be seen similar to non-mucinous adenocarcinoma, but not solid:
        • lepidic
        • acinar
        • papillary
        • micropapillary
      • criteria to differentiate from mucinous AIS or mucinous MIA (only need one of these criteria to call invasive mucinous adenocarcinoma):
        • size > 3cm
        • invasion > 0.5cm
        • multiple nodules
          • or lack of a circumscribed border with military spread into adjacent lung parenchyma
      • strong tendency to multicentricity involving multiple lobes and bilateral lungs (?aerogenous spread)
      • strong association with KRAS mutations (~76%)
    • mixed mucinous and non-mucinous adenocarcinoma
      • needs at least 10 % of each component
    • fetal adenocarcinoma:
      • tubules composed of glycogen-rich, nonciliated cells resembling fetal lung tubues
      • subnuclear vacuoles are common and characteristic
      • squamoid morules may be seen
      • most are low grade with a favourable outcome
      • high-grade tumours occur
      • may be mixed, should be classified according to the predominant component
      • beta-catenin gene mutations (detected by nuclear and cytoplasmic IHC staining
    • mucinous (“colloid”) adenocarcinoma
      • extracellular mucin in abundant pools
      • distend alveolar spaces with destruction of their walls
      • mucin pools contain clusters of mucin-secreting tumour cells (may be inconspicuous)
      • more often seen along with other histologic patterns, rather than as a pure pattern
        • colloid adenocarcinoma classification is used if it is the predominant component
      •  
    • mucinous cystadenocarcinoma
      • rare
      • should be classified as colloid adenocarcinoma with cystic changes
    • signet ring adenocarcinoma
    • clear cell adenocarcinoma

 

 

Immunophenotype:

Marker:

Sensitivity:

Specificity:

PAS pos & PASD neg

80%

 

AE1/AE3

 

 

CAM 5.2

 

 

EMA

 

 

CEA

 

 

CK7

Most

Positive in up to 30% of squamous cell carcinomas

TTF-1*

75% of invasive

(Negative in most invasive mucinous adenocarcinomas)

(less common in solid pattern)

Also positive in:

Small cell CA

Large cell NEC

Carcinoids

Thyroid CA

Napsin A

Comparable to TTF-1

Sometimes expressed in RCC

CK20 (neg)*

Most

Positive in ~54% of invasive mucinous adenocarcinoma

 

Thyroglobulin (neg)

 

 

P63 (neg)

May be positive in up to 30% of adenoCA

(Focal / weak)

 

P40 (neg)

More specific than p63, but can be seen in adenoCA (focal / weak)

 

34Beta12 (CK903) (neg)

Frequently positive in solid pattern adenoCA

 

EGFR mutation specific antibodies

(not recommended)

76% for L858R

60% or less for exon 19 deletions

High

ALK

(5A4 or D5F3 clones)

FDA approved Ventana ALK (D5F3) CDx Assay as a companion diagnostic for crizotinib

May be used to screen for ALK rearrangement with confirmation by FISH before initiating ALK-targeted therapy

 

 

ROS1

“Robust screening tool”

Suboptimal

Positive results need to be confirmed by another technique

BRAF (V600E)

Variable

Variable

RET

Not well evaluated yet

 

 

PD-L1

Preliminary studies show an encouraging predictive association with response to PD1 antibody therapy.

Further assay validation is needed.

Antibody and scoring cutoffs have not been established.

Currently no validated, commercially available assay for PD-L1 expression that is predictive of outcome

 

 

·       *mucinousBAC is typically CK20 pos and TTF-1 neg

·       Biopsies:

·       Cases positive for an adenocarcinoma marker (i.e. TTF1 or napsin A) and/or mucin, with a negative squamous marker (i.e. p40 or p63) should be classified as NSCC, favour adenocarcinoma

·       If an adenocarcinoma marker such as TTF-1 is positive, the tumour should be classified as NSCC, favour adenocarcinoma, regardless of any expression of squamous markers.

·       If intracytoplasmic mucin can be demonstrated in a poorly-differentiated NSCC with a mucin stain in at least two tumour cells in the biopsy (and in the absence of IHC markers for adenocarcinoma or squamous cell carcinoma) the diagnosis of adenocarcinoma is appropriate

·       If ADC marker (i.e. TTF-1) and/or mucin +ve, and SqCC marker neg, can call NSCC, favour ADC

·       Resections:

·       Undifferentiated carcinomas that express pneumocyte immunohistochemical markers, and/or mucin expression, are classified as adenocarcinoma (previously large cell carcinomas)

·       Although primary lung adenoCA can be TTF-1 negative, additional IHC studies may be helpful to exclude metastasis (i.e. CDX2, CK20, ER, or PR)

 

Molecular features:

    • > 50% contain an identifiable genetic alteration, some of which can be targeted by a specific therapeutic inhibitor
    • KRAS mutations (20-40%) (extremely rare in other histologic types)
      • Mutations at codon 12
      • Mutually exclusive with EGFR and ALK alterations typically
      • Methylation often present in early stages of cancer
      • RAS is negatively regulated by the catalytic reaction of RAS GTPase-activating proteins (RAS-GAPs), which enhances RAS GTPase activity [193]
      • Epidemiology:
        • More frequent in smokers (30%) than non-smokers (5%)
        • smokers186, 192,194
        • the incidenceof KRAS mutations increased as smoke exposure increased193
        • Recently, we evaluated the frequency of KRAS mutations inlung adenocarcinomas from nearly 500 patients, of whom 17%had never smoked cigarettes (30). We noted that KRAS mutations occurred in 22% of the overall population andin15% oflung adenocarcinomas from never-smokers.194
        • KRAS transition mutations (G/A) were more common in patients who hadnever smoked cigarettes. In contrast, transversion mutations(G/T or G/C) were more common in former/current smokers194
        • Thus, unlike EGFR mutations, which occur more frequently in tumors from never-smokers (31), KRAS tumorstatus cannot be easily predicted on the basis of smoking historyalone.194
        • KRAS mutations occur predominantlyin Caucasian patients rather than in East Asians; theincidence of KRAS mutation is 30% in Caucasian patients and 10% in East Asian patients with adenocarcinoma193
        • African Americans aresignificantly less likely to harbor EGFR mutations (2%),whereas the frequency of KRAS mutations (23%) does notdiffer from that in Caucasians [55].
      • Gross:
        • Hilar location frequently
      • Histology:
        • Invasive mucinous adenocarcinoma (formerly mucinous BAC) show a very strong correlation with KRAS mutations
        • Overall histology cannot be relied upon for predicting KRAS mutation
      • Prognosis:
        • Their impact on overall survival remains controversial.
        • Soh et al. found that gene dosage is associated with prognostic impact [43].193
        • of 237 lung adenocarcinoma patients,six patients who harbored both a KRAS mutation and CNGhad significantly shortened survival (P=0.04) [43].193
        • KRAS mutations could be associated with Progressive Disease status.200
      • Prediction of response to therapy:
        • No demonstrably efficacious treatments
        • Lack of response to EGFR inhibitors
          • the decision to treat with an EGFR TKI can no longer be made without an EGFR result, and the role of KRAS testing in this context has diminished.
            • because KRAS and EGFR mutations are mutually exclusive, a rapid and inexpensive KRAS assay may be performed initially to exclude KRAS-mutated tumors from EGFR mutation testing as part of an algorithm designed to maximize testing efficiency, provided that the sample is sufficient to perform the KRAS test without sacrificing EGFR and ALK testing, and that the totality of clinically relevant molecular results can be obtained within the target TAT.
          • absence of a KRAS mutation does not add clinically useful information to the EGFR mutation result and should not be used as a determinant of EGFR TKI therapy.
          • it is notclear whether the response to EGFR-TKIs differs betweentumors harboring KRAS mutations and those harboringneither KRAS nor EGFR mutations193
          • Objective response to EGFR TKI can be seen in 0% to 3% of patients with KRAS mutations and 26% of patients with KRAS wild type.
          • In 2005, we examined the tumor KRASstatus in patients with NSCLC who had been treated with eitherdrug as a single agent (2). Collectively, none of 21 patients whosedisease responded radiographically had KRASmutations, while 9of 38 patients with refractory disease had KRAS mutations (P 50.02). Multiple other groups have reported similar ndings (Table1)194
          • KRAS mutations are mutually exclusive to EGFR mutations
        • Of note, the response rate for patients treated with carboplatin and paclitaxel did not differ signicantly by KRASmutation status (26% versus 23%)194
      •  
    • EGFR mutations (20%)
      • The EGFR gene is located on the short arm of chromosome 7 (7p11.2) and encodes a 170-kDa type I transmembrane growth factor receptor with TK activity (10)218
      • Strongly associated with clinical response to EGFR tyrosine kinase inhibitors such as gefitinib, erlotinib, afatinib, small-molecule (tyrosine) kinase inhibitors
        • Exon 18 Gly719#
      • Epidemiologic associations:
        • never smokers or light smokers (never smoked, 47% compared with ever smoked, 7%),
        • predominantly in female
          • (male, 10% compared with female,38%)
        • ethnicity:
          • 10-20% for caucasians, 50% for East Asians
          • East Asians, 32% compared with Caucasians, 7%, African Americans aresignificantly less likely to harbor EGFR mutations (2%),whereas the frequency of KRAS mutations (23%) does notdiffer from that in Caucasians [55].
      • Histologic associations:
        • TTF-1 positive (close relationship)
        • much more common in adenocarcinoma than other histologic types
        • lepidic, papillary, or acinar histology (more likely)
          • less likely in poorly-differentiated, mucinous, or solid histology (but still found with significant frequeny in all grades)
          • histologic subtype should not be used to determine which samples should be tested for EGFR
        • very infrequent in squamous cell carcinoma (but some are reported)
        • very infrequent in invasive mucinous adenocarcinoma (~3%)
        • not detected in small cell carcinoma nor in carcinoids
        • rare reports in salivary gland-type tumours, large cell carcinomas, sarcomatoid carcinomas, large cell neuroendocrine carcinoma
      • EGFR Mutation correlates with copy number changes
      • Prognosis
        • Several studies have reported that patients with untreated NSCLC with EGFR mutations have a more favourableprognosis than patients with wild-type EGFR.201
        • In the BR.21 trial, untreated patients in the placebo group with EGFRmutant tumours had a median survival of 8·3 months(range 3·3–11·1) compared with 3·3 months (2·5–6·8)for patients with wild-type EGFR.1(201)
        • in the TRIBUTE trial: Patients with mutated tumors (29/228=13%) had signicantly better clinicaloutcomes in all assessed end points, including survival, compared with patients with wildtype EGFR tumors, regardlessof the therapygiven218
        • treatment with erlotinib pluschemotherapy versus chemotherapy alone inadvanced NSCLC,218
        • EGFR mutations were detected in 13% of tumors and were associated with longer survival,irrespective of treatment (P .001).220
        • We analyzed 397 patients with lung adenocarcinoma who underwent potentially curative pulmonary resection. Univariate analysis showed that patients with EGFR mutations survived for a longer period than those without mutations (p 0.0046).221
        • Multivariate analysis using the Cox proportional hazards model revealed that smoking status (p 0.0310) and disease stage (p 0.0001) were independent prognostic factors. However,none of the gene mutations was independent prognostic factors(EGFR, p 0.3225; KRAS, p 0.8500; TP53, p 0.3191).221
      • Prediction of response to therapy:
        • Benefits for first-line treatment with gefitinib, erlotinib, and afatinib
        • Gefitinib, afatinib, and erlotinib response:
          • EGFR molecular testing should be used to select patients for EGFR-targeted TKI therapy
            • Testing should not be chosen based on clinical characteristics
          • RR 68% (response rate)
            • these patients almost invariably experience recurrence or progression while on treatment after a median of 8 to 16 months, a clinical phenomenon termed acquired resistance
          • PFS 12 months
            • Significantly longer than those receiving platinum-based chemotherapy
          • OS not improved compared to chemotherapy so far
          • EGFR wild-type tumours respond better to conventional chemotherapy than to EGFR TKI
        • Platinum-based chemotherapy:
          • EGFR wild-type tumours respond better to conventional chemotherapy than to EGFR TKI
        • In 2004, the results of the National Cancer Institute of Canada Clinical Trials Group BR.21study led to the global approval of erlotinib asthe rst targeted therapy for advanced NSCLC(6, 7).218
        • At approximately the same time, two groups (8, 9) reported the discovery of mutations in the tyrosine kinase (TK) domain ofthe EGFR gene in NSCLC. Furthermore, thepresence of these mutations appeared to correlate with sensitivity to the EGFR inhibitor ge-tinib.218
        • which mutations are responsive / non-responsive?
          • L858R and exon 19 deletion mutations show the greatest sensitivity to small-molecule epidermal growth factor receptor (EGFR) inhibitors
          • Among all EGFR mutations, four types arestrongly correlated with TKI sensitivity in vitroand in vivo. These are point mutations in exons 18 (G719A/C) and 21 (L858R andL861Q) and in-frame deletions in exon 19(218)
            • Exon 18 Gly719 is sensitive for example
          • Exon 20 EGFR activating mutations are generally associated with resistance to EGFR tyrosine kinase inhibitors such as erlotinib, afatinib, and gefitinib, although insertions at or before position 768 can be associated with sensitivity
            • E.g. A763_Y764insFQEA mutant, which is sensitive to erlotinib and gefitinib.
          • Recently, a rare exon 22 mutation (E884K) that may confer differential sensitivity to different EGFR small-molecule inhibitors was reported (23)218
          • There is limited data on response to EGFR tyrosine kinase inhibitors for many of the uncommon EGFR activating mutations.
          • Whereas V689M, N700D,L718P, V765A, V783A, A839T, and K846R areassociated with response to getinib, E709Q/L,A763V, N826S, and V752I are associated withlack of response (62).218
        • mutations conferring resistance during treatment
          • biopsy at time of relapse may be required to determine the best course of therapy
          • The most common secondary mutation is the T790M substitution of methionine for threonine on codon790 (85–87).218 (60-70% using sensitive methods)
            • caused by a single base substitution, C to T, at nucleotide 2369
            • This mutation occurs in cis on the same allele as the original activating mutations; it may be either an exon 19 deletion oran exon 21 L858R mutation218
            • studies showed that the T790M mutation preferentially increasesthe afnity of the receptor for ATP, therebyreducing the effectiveness of the TKI (25)218
            • Although T790M mutation induces resistance to getinib, it can still be inhibited by some irreversible EGFR inhibitors (88)218
            • accounts for approximately 50% to 60% of acquired resistance to EGFR TKI therapy (51)218
            • If seen in untreated/pretreated patients, may be present in the germline and indicate a hereditary cancer syndrome, in which case genetic counseling is suggested.
              • Most studies have only rarely detected T790M in pretreatment samples.
              • Notably,some studies have detected the T790M mutation at low levels in a few patients prior to initiation of EGFR TKI therapy,suggesting thatsubclones bearing these mutations are preexistent (83), as is true in other cancers, such aschronic myelogenous leukemia, that respond totargeted TKIs.218
            • Resistance mutations in the drug target markedly diminish the potency of the inhibitor againstthe kinase. Examples include EGFR T790M and BCR-ABL T315I.223
            • EGFR T790M is essentially the sole resistance mutation observed in the clinic.223
            • Recent data suggest that AR patients with the T790M mutation can derive continued clinical benefit from the first-line EGFR TKI
          • L747S, D761Y, and T854A
            • owing to their relatively low prevalence, there is not much clinical experience with these
          • MET amplication may account for approximately 10% to 20% of acquired resistance to getinib and erlotinib (110).218
            • there is currently a lack of a precise definition of clinically significant MET amplification in this setting and more research is needed before guidelines can be formulated
          • ERBB2 amplification has been reported in a subset
          • SCLC histology and associated ‘‘SCLCtype’’ radiosensitivity and chemosensitivity have been observed in some AR cases
      • EGFR Mutation details
        • TK domain of EGFR; the mutations are characterized by short deletions in exon 19 and point mutations (G719S, L858R,and L861Q) in exons19 and 21 (Figure 2)218
        • According to their nucleotide changes,the mutations have been classified into three types.
          • Class I mutations include short in-frame deletions that result in the loss of four to six amino acids (E746 to S752) encoded by exon 19.
            • Small deletions in the LREA motif of exon 19
          • Class II mutations are single-nucleotide substitutions that may occur throughout exons 18 to 21.
            • By far most common is L858R (leucine to arginine) (exon 21)
          • Class III mutations are in-frame duplications and/or insertions that occur mostly in exon 20 (20, 21).218
        • About 3% of EGFR mutations occur at codon 719 and cause substitution of glycine to cysteine, alanine, or serine (G719X) [54]. In addition, about 3% are in-frame insertion mutations in exon 20 [193]
      • Molecular biology:
        • EGFR belongs to theHER/erbB family of receptor tyrosine kinases (RTKs), which includes HER1 (EGFR/erbB1),HER2 (neu, erbB2), HER3 (erbB3), and HER4(erbB4).218
        • Intracellular signaling is mediated mainly through the RAS-RAF-MEK-MAPK pathway, the PI3KPTEN-AKT pathway, and the signal transducer and activator of transcription (STAT)pathway (13). Downstream EGFR signaling ultimately leads to increased proliferation, angiogenesis, metastasis, anddecreased apoptosis(Figure 1) (14).218
        • EGFR mutation has been shown to activate the antiapoptotic Akt and signal transducers and activators of transcription pathways
      • EGFR mutation testing guidelines (CAP/IASLC/AMP/ASCO)
    • ALK gene rearrangement (2-7% in US):
      • inv(2)(p21p23)
        • Inversion on chromosome 2p resulting in EML4-ALK fusion gene
        • fusion gene encoding the amino-terminal portion of EML4 (2p21) and the intracellular region of ALK (2p23), genes that are normally approximately 13 Mb apart.8
      • other less common variant fusions have been reported
        • translocations with other chromosomes (KIF5B-ALK, TFG-ALK)
          • KIF5B-ALK
          • TFG-ALK
          • KLC1-ALK
        • NOT the NPM-ALK transloation characterized in ALCL
      • Epidemiology:
        • Never smokers
        • Younger age
        • No sharp differences in prevalence according to sex and ethnic origin
      • Histology:
        • Adenocarcinoma, adenosquamous
          • Very infrequent in squamous cell carcinoma
        • Subtype is not strongly predictive
          • Solid histology maybe
          • Signet ring histology maybe
      • Inevitable acquired resistance to ALK-targeted inhibitors:
        • ALK kinase domain mutation (often)
          • ALK mutations that confer acquired resistance to crizotinib
            • L1152R, C1156Y, F1174L, L1196M, L1198P, D1203N, and G1269A have been reported
            • Testing for secondary mutations in ALK associated with acquired resistance to ALK inhibitors is not currently required for clinical management (the mechanism of resistance does not clearly predict response to next line therapy)
            • Ceritinib has shown efficacy in these relapsed patients and has been approved for those who developed resistance to, or could not tolerate, crizotinib
      • Testing guidelines (CAP/IASLC/AMP/ASCO)
      • Prediction of response to therapy:
        • ALK rearrangement predicts response to therapy with a targeted ALK inhibitor, such as crizotinib or ceritinib.
        • Some evidence suggests the type of FISH pattern (breakapart versus 5' probe deletion) may have implications for treatment response and outcomes.
        • Polysomy involving the ALK locus confirms that fluorescence in situ hybridization (FISH) scoring was carried out in tumor cells but does not predict response to therapy with targeted ALK inhibitors.
        • ALK IHC:
          • Absence of ALK protein expression in cancer cells suggests that this tumor is unlikely to harbor ALK rearrangement and to respond to treatment with a targeted inhibitor, such as crizotinib and ceritinib.
          • ALK protein expression in cancer cells (based on platform criteria) predicts the presence of ALK rearrangement and response to therapy with a targeted inhibitor, such as crizotinib and ceritinib.
          • Tumors with faint cytoplasmic labeling should be designated as equivocal. This result can rarely occur both with and without mutation.
        • mean response rate to EGFR TKI for patients with EGFR mutations is 68%
        • crizotinib (ALK/MET/ROS1 inhibitor):
          • RR 57%
          • PFS 6 months or greater in 72%
          • FDA approved for advanced-stage, ALK-positive lung cancer
          • ALK molecular testing should be used to select patients for ALK-targeted TKI therapy
            • Testing should not be chosen based on clinical characteristics
        •  
    • BRAF mutation (5%)
      • V600E (half of BRAF mutations in lung cancer)
        • Proportion of non-V600E mutations is higher in lung cancer than other cancers
      • dabrafenib (BRAF inhibitor) combined with trametinib (MEK inhibitor) showed efficacy in Phase II clinical trials (63% overall response rate in V600E)
      • smokers
    • ROS1 gene rearrangement (1-2% of NSCLC)
      • ROS1 rearrangement predicts a high response rate to therapy with a targeted inhibitor, such as crizotinib.
      • Fusion partners: SLC34A2, CD74, TPM3, GOPC (FIG), SDC4, EZR, LRIG3, KDELR2, CCDC6
      • FISH, IHC, PCR, NGS may be used (no “gold standard” method defined yet)
      • Polysomy involving the ROS1 locus confirms that fluorescence in situ hybridization (FISH) scoring was carried out in tumor cells but does not predict response to therapy with targeted inhibitors.
      • ROS1 IHC:
        • Absence of ROS1 protein expression in cancer cells suggests that this tumor is unlikely to harbor ROS1rearrangement and to respond to treatment with a targeted inhibitor, such as crizotinib.
        • ROS1 protein expression in cancer cells is highly sensitive for a rearrangement involving ROS1 but is not entirely specific. Therefore, confirmatory molecular methods should be used when ROS1 protein expression is detected.
        • Tumors with faint cytoplasmic labeling should be designated as equivocal. This result can rarely occur both with and without mutation.
    • MET mutations (3%)
      • Splicing variants
      • Insertion-deletion mutations resulting in exon 14 deletion
        • May be associated with MET amplification
      • Associated with response to crizotinib and cabozantinib
    • RET mutations and gene fusions (1-2%)
      • RET rearrangement is associated with response to targeted RET inhibitor therapies, such as cabozantinib and vandetinib
      • Absence of RET rearrangement in cancer cells suggests that this tumor is unlikely to respond to treatment with a targeted RET inhibitor.
      • Never smokers (almost exclusively)
      • Cabozantinib (multi-targeted inhibitor) shows promise in phase II trials
        • Phase 3 trials not conducted yet
      • FISH
        • The most common rearrangement, KIF5B-RET, is subtle intrachromosomal inversion and may be difficult to detect practically by FISH
      • PCR, NGS
    • ERBB2 mutation
      • exon 20 insertion mutation
      • phase I promising for HER-inhibitors
    • MAP2K1 (MEK1) (rare)
      • May predict sensitivity to MEK inhibitors
    • PIK3CA mutation (rare)
    • AKT1 mutation (rare)
    • PTEN loss
    • NRG1/NTRK1 (TRKA) fusion (rare)
    • beta-catenin gene mutations
      • fetal adenocarcinoma histology
    • MET amplification
      • Rare cases of de novo MET amplification have been associated with profound responses to crizotinib
      • May be associated with exon 14 deletion (see above)
    • EGFR copy number gain / amplification
      • Increased EGFR gene copy number (polysomy or amplification) is observed in about 40% of cases, with a range of 8% to 66%
      • EGFR TKI response rates for patients with EGFR polysomy/amplification is 30%
    •  
    • Methylation rates of APC, CDH13, and RARb (significantly higher in ADC than in SCC)
    • Changes common to all lung CA

 

Other features:

    • Metastasize widely and earlier than SCC
    • Atypical adenomatous hyperplasia is precursor lesion for some
    • Prognostic factors:
      • AIS and MIA – 100% or near 100% survival with complete resection
      • Size of central scar – smaller is better prognosis
      • Vascular invasion is worse prognosis
      • Papillary pattern >25% is worse prognosis
      • Fetal adenocarcinoma – most are low grade and favorable outcome
    • Predictive factors:
      • EGFR mutation – predictive of responsiveness to EGFR tyrosine kinase inhibitors
      • ADC histology is a strong predictor for improved outcome with pemetrexed therapy compared with SCC
      •  

 

References:

    • Robbins 2005
    • Travis WD.  Pathology of Lung Cancer.  Clinics in Chest Medicine 2002; 23(1): 65-81.
    • WHO blue book 2004
    • Travis et al.  International association for the study of lung cancer / American Thoracic Society / European Respitratory Society International Multidisciplinary Classification of Lung Adenocarcinoma.  Journal of Thoracic Oncology 2011;6(2):244-285.
    • Thomas RK, Weir B, Meyerson M.  Genomic approaches to lung cancer.  Clin Cancer Res (2006); 12(14 Suppl): 4384s-91s.
    • Travis et al.  WHO Classification of Tumours of the Lung, Pleura, Thymus, and Heart, 4th ed. (2015)
    • CAP – Template for Reporting Results of Biomarker Testing of specimens from patients with non-small cell carcinoma of the lung (June 2016)
      • CAP draft lung biomarker template protocol (Sep. 2015)