Skip to main content
Erschienen in: Diagnostic Pathology 1/2012

Open Access 01.12.2012 | Research

Stromal micropapillary component as a novel unfavorable prognostic factor of lung adenocarcinoma

verfasst von: Miki Ohe, Tomoyuki Yokose, Yuji Sakuma, Yohei Miyagi, Naoyuki Okamoto, Sachie Osanai, Chikako Hasegawa, Haruhiko Nakayama, Yoichi Kameda, Kouzo Yamada, Takeshi Isobe

Erschienen in: Diagnostic Pathology | Ausgabe 1/2012

Abstract

Background

Pulmonary adenocarcinomas with a micropapillary component having small papillary tufts and lacking a central fibrovascular core are thought to result in poor prognosis. However, the component consists of tumor cells often floating within alveolar spaces (aerogenous micropapillary component [AMPC]) rather than invading fibrotic stroma observed in other organs like breast (stromal invasive micropapillary component [SMPC]). We previously observed cases of lung adenocarcinoma with predominant SMPC that was associated with micropapillary growth of tumors in fibrotic stroma observed in other organs. We evaluated the incidence and clinicopathological characteristics of SMPC in lung adenocarcinoma cases.

Patients and Methods

We investigated the clinicopathological characteristics and prognostic significance of SMPC in lung adenocarcinoma cases by reviewing 559 patients who had undergone surgical resection. We examined the SMPC by performing immunohistochemical analysis with 17 antibodies and by genetic analysis with epidermal growth factor receptor (EGFR) and KRAS mutations.

Results

SMPC-positive (SMPC(+)) tumors were observed in 19 cases (3.4%). The presence of SMPC was significantly associated with tumor size, advanced-stage disease, lymph node metastasis, pleural invasion, lymphatic invasion, and vascular invasion. Patients with SMPC(+) tumors had significantly poorer outcomes than those with SMPC-negative tumors. Multivariate analysis revealed that SMPC was a significant independent prognostic factor of lung adenocarcinoma, especially for disease-free survival of pathological stage I patients (p = 0.035). SMPC showed significantly higher expression of E-cadherin and lower expression of CD44 than the corresponding expression levels shown by AMPC and showed lower surfactant apoprotein A and phospho-c-Met expression level than corresponding expression levels shown by tumor cell components without a micropapillary component. Fourteen cases with SMPC(+) tumors (74%) showed EGFR mutations, and none of them showed KRAS mutations.

Conclusions

SMPC(+) tumors are rare, but they may be associated with a poor prognosis and have different phenotypic and genotypic characteristics from those of AMPC(+) tumors.

Virtual Slides

The virtual slide(s) for this article can be found here: http://​www.​diagnosticpathol​ogy.​diagnomx.​eu/​vs/​9433341526290040​.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1746-1596-7-3) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

MO and TY designed the study, performed clinical and pathological investigation, and wrote the drafts. YS and YM participated in pathological and genetical investigation. NO participated in statistical investigation. SO performed the histological and immunohistochemical evaluation. CH assisted the clinical investigation. HN participated in managing and operating the patients. YK assisted the pathological investigation. KY participated in collecting clinical data and images. TI participated in its design and coordination and helped to draft the manuscript. All authors read and approved the final manuscript.
Abkürzungen
AMPC
aerogenous micropapillary component
SMPC
stromal micropapillary component
MPC
micropapillary component
TMA
tumor tissue microarray
DFS
disease-free survival.

Background

A new lung adenocarcinoma classification system has been proposed by the International Association for the Study of Lung Cancer, American Thoracic Society, and European Respiratory Society (IASLC/ATS/ERS) [1]. In this classification, the micropapillary component (MPC) was recommended as a new subtype of lung adenocarcinoma in addition to the lepidic, acinar, papillary, and solid subtypes defined in the 2004 World Health Organization (WHO) classification [2]. MPC was defined as tumor cells growing in papillary tufts lacking fibrovascular cores and may float within alveolar spaces. MPC-predominant lung adenocarcinoma shows a high incidence of nodal metastasis and a poor prognosis [38]. MPC-predominant carcinomas developing in various other organs, such as the breast and urinary bladder, known as invasive micropapillary carcinoma, also have a poor prognosis. However, localization of MPC in the lungs is significantly different from that in the other organs; MPC in lung adenocarcinoma is distinguished by floating tumor cells within alveolar spaces (aerogenous micropapillary component, AMPC), while MPC in other organs has been observed primarily in the stroma as invasive components (stromal invasive micropapillary component, SMPC) [3, 4].
Few studies have examined lung adenocarcinoma with SMPC [9, 10]. Recently, we reported 2 cases of SMPC-predominant lung adenocarcinoma [9]. The proportion of SMPC in both tumors was greater than 50% in area. We observed that SMPC had a strong association with vascular invasion, similar to the cases of SMPC-predominant carcinoma in other organs. However, a large-scale investigation on pulmonary SMPC has not been conducted.
The aims of this study included: (1) clarifying the incidence of SMPC in lung adenocarcinoma; (2) elucidating the clinicopathological characteristics of the tumor; and (3) determining the prognoses of the SMPC-positive (SMPC(+)) tumors and comparing them with those of SMPC-negative (SMPC(-)) tumors. We reviewed 559 resected lung adenocarcinomas for this study with performing immunohistochemical and genetic analysis.

Methods

Patients

We analyzed 565 consecutive cases of primary lung adenocarcinoma treated by surgical resection at the Kanagawa Cancer Center between February 2007 and December 2010. Formalin fixation of the resected lung tissue was performed within 48 hours to reduce the loss of immunohistochemical antigen expression and degeneration of DNA. Six patients who had received preoperative chemotherapy were excluded. A total of 559 cases were enrolled in the study. The median follow-up time was 634.5 days (range, 28-1512 days). All patients provided informed consent, and the studies were performed according to the requirements of the institutional review board of Kanagawa Cancer Center.

Pathological review

Excised specimens were fixed in a solution of 10% buffered formaldehyde, and the sections were embedded in paraffin. Next, 4-μm-thick sections, including the largest cut surface of the tumor, were prepared and stained using hematoxylin and eosin (HE) as well as alcian blue and elastica-van-Gieson (AB-EVG) to detect cytoplasmic mucin production and the elastic fiber framework. Lymphatic invasion and pulmonary metastasis were evaluated on HE sections. Vascular and pleural invasion was evaluated in AB-EVG sections. Sections were reviewed by 2 observers (M.O. and T.Y.) who were unaware of the clinical data. Tumor size was measured as the maximal diameter on the cut sections of the lung. Pathological stage was determined based on the criteria of the 7th TNM classification of Union of International Cancer Control [11].

Histological definition of micropapillary components

Histopathological diagnosis of lung adenocarcinoma was determined according to the IASLC/ATS/ERS international multidisciplinary classification of lung adenocarcinoma [1]. Comprehensive histological subtyping was performed on the primary tumor and divided by percentage into 5 distinctive subtypes: lepidic, acinar, papillary, micropapillary, and solid, totaling 100% per tumor. We defined the subtype as positive when it occupied at least 1% of the entire tumor. We classified a micropapillary subtype into 2 components, AMPC and SMPC, using the following criteria: AMPC is widely recognized in the lungs as tumor cells floating within alveolar spaces, and SMPC includes papillary components consisting of tufts lacking central fibrovascular cores, surrounded by lacunar spaces and identified as invasive components in the stroma as previously described [9] (Figure 1A and 1B). Additionally, a tumor area without micropapillary components was defined as a non-micropapillary component (nMPC).

Tumor tissue microarray (TMA) synthesis

TMAs were constructed using a manual tissue-arraying instrument (KIN-4; Azumaya, Tokyo, Japan) as previously described [12], and specimens were punched using a stylet 3 mm in diameter.

Immunohistochemistry

The 17 antibodies used for immunohistochemical characterization of tumor cells in TMA in this study are listed in Table 1. Immunohistochemical staining was performed as follows. TMA recipient blocks were cut into 4-μm-thick sections and mounted on silane-coated slides. HE staining was performed on initial sections to verify histology. The remaining sections were deparaffinized in xylene and dehydrated in a graded alcohol series, and endogenous peroxidase was blocked using 3% hydrogen peroxide in absolute methyl alcohol. Heat-induced epitope retrieval was performed for 20 min at 95°C in 0.02 mol/L citrate buffer (pH 6.0) in samples fixed with 10% formalin if necessary. The slides were rinsed using deionized water and incubated with primary antibodies. They were then washed 3 times in phosphate-buffered saline and incubated with EnVision+ System-HRP (DAKO, Glostrup, Denmark). The reaction products were visualized using 3-3'-diaminobenzidine tetrahydrochloride, and sections were counterstained using hematoxylin. Additionally, a similar staining method was used for anti-podoplanin antibody (clone D2-40, pre-diluted; Ventana, Tucson, AZ, USA) to evaluate lymphatic permeation.
Table 1
Antibodies
Classification/Antibody
Clone
Dilution
Source
Cellular adhesion molecules
  
   E-cadherin
NCH-38
1:100
DakoCytomation, Carpinteria, CA, USA
   CD44
DF1485
1:400
Novocastra, Newcastle upon Tyne, UK
   Laminin5γ2
4G1
1:50
DakoCytomation, Glostrup, Denmark
Growth factor
  
   VEGF-C
Polyclonal
1:50
Abcam, Cambridge, UK
Apoptosis-associated proteins
  
   bcl2
124
1:50
DakoCytomation, Glostrup, Denmark
   p53
DO-7
Pre-diluted
Nichirei, Tokyo, Japan
   cleaved caspase-3
Polyclonal
1:400
Cell signaling, Danvers, MA, USA
Mucin-related proteins
  
   MUC1
Ma695
1:100
Novocastra, Newcastle upon Tyne, UK
   MUC6
CLH5
1:100
Novocastra, Newcastle upon Tyne, UK
Hypoxia induced protein
  
   HIF-1α
EP1215Y
1:500
Abcam, Cambridge, UK
Others
   
   TTF-1
8G7G3/1
1:100
DakoCytomation, Carpinteria, CA, USA
   SP-A
PE10
1:100
Dako, Kyoto, Japan
   Vimentin
V9
Pre-diluted
DakoCytomation, Carpinteria, CA, USA
   Ki-67
MIB-1
1:50
Dako, Glostrup, Denmark
   LYVE1
15A5B2
1:400
Oriental Yeast, Tokyo, Japan
   c-Met
EP1454Y
1:200
Abcam, Cambridge, UK
   Phospho-c-Met
Polyclonal
1:800
Stressgen, Ann Arbor, MI, USA
VEGF-C, vascular endothelial growth factor-C; HIF-1α, hypoxia induced factor 1-α; TTF-1, thyroid transcription factor-1; SP-A, surfactant apoprotein A; LYVE1, lymphatic vessel endothelial hyaluronan receptor 1.

Calculation of staining scores

Immunostaining was scored based on staining intensity and percentage of positively stained cells, with 2 observers evaluating immunostained samples independently. When the observers gave different scores to immunostained samples, the slides were reviewed together under a multiheaded microscope until a consensus was reached. Sections were classified by staining intensity as negative (total absence of staining), 1+ (weak staining), 2+ (moderate staining), or 3+ (strong staining). Staining scores were calculated by multiplying the percentage of positive tumor cells per section (0-100%) by the staining intensity; scores obtained ranged from 0 to 300. Expression of p53, cleaved caspase-3, and Ki-67 were determined by counting 300 tumor cells under a high power field (×400) and results are shown as the percentage of positive cells.

Mutation analysis

Mutation analyses of EGFR gene exons 19 and 21 and KRAS gene codons 12 and 13 were performed using loop-hybrid mobility shift assays and gene sequencing procedures described elsewhere [13].

Statistical analysis

All calculations were performed using SPSS software (Dr. SPSS II for Windows Standard version 11.0; SPSS Inc., Chicago, IL, USA). The Chi-square for independence or Fisher's exact probability test was performed to analyze differences in patient characteristics between the 2 groups. The Fisher's exact probability test was performed if there were 5 or fewer observations in a group. For univariate analysis, all cumulative survival was estimated using the Kaplan-Meier method, and differences in variables were calculated using the log-rank test. Multivariate regression analysis was conducted according to the Cox proportional hazard model. The Mann-Whitney U test was used to compare staining scores. Differences were considered significant when the P value was less than 0.05.

Results

Clinicopathological characteristics of patients with SMPC

Figure 2 shows a Venn diagram of the relationship between the micropapillary component sets in the 559 patients examined in this study. SMPC was observed in 19 patients (3.4%) and AMPC in 99 (17.7%) patients. A mixture of SMPC and AMPC was observed in 14 patients, pure SMPC without AMPC in 5 patients, and pure AMPC without SMPC in 85 patients. A micropapillary pattern was observed in 50-100% in 2 SMPC tumor and less than 50% in 17 SMPC tumors. No SMPC(+) tumors were completely replaced by SMPC. Clinicopathological characteristics of patients with SMPC(+) and SMPC(-) tumors are summarized in Table 2. Patients with SMPC(+) tumors were significantly found to be at a more advanced stage, larger than 30 mm in diameter, and have more frequent lymph node metastasis compared to those with SMPC(-) tumors. Pleural, lymphatic, and vascular invasion were observed more often in patients with SMPC(+) tumors than in those with SMPC(-) tumors. (68% vs. 17%, P < 0.001; 74% vs. 15%, P < 0.001; 74% vs. 22%, P < 0.001, respectively). No significant differences in age, gender, or smoking status were observed between patients with SMPC(+) and SMPC(-) tumors.
Table 2
Clinicopathological characteristics of patients with SMPC
 
all
%
SMPC
P value
   
(-)
%
(+)
%
 
No.
559
 
540
97
19
3
 
Age
       
   Median
67
 
67
 
67
 
0.219*
   Range
23-87
 
23-87
 
40-76
  
Gender
       
   Female
288
52
282
52
6
32
0.077**
   Male
271
48
258
48
13
68
 
Smoking status
       
   Nonsmoker
284
51
276
51
8
42
0.596**
   Smoker
275
49
264
49
11
58
 
   BI Average
369
 
364
 
502
  
Tumor size
       
   Average(mm)
25
 
25
 
35
  
   Range(mm)
5-140
 
5-140
 
15-75
  
   < 30 mm
396
71
388
72
9
47
< 0.001*
   ≥ 30 mm
163
29
152
28
10
53
 
Pathological stage
      
   IA
363
65
360
67
4
21
< 0.001**
   IB
95
17
88
16
6
32
 
   IIA
36
6
31
6
5
26
 
   IIB
13
2
13
2
0
0
 
   IIIA
42
8
39
7
3
16
 
   ≥ IIIB
10
2
9
2
1
5
 
Lymph node metastasis
      
   NX
69
12
68
13
1
5
 
   N0
420
75
409
75
11
58
0.002**
   ≥ N1
70
13
63
12
7
21
 
Pleural invasion
       
   Negative
452
80
446
83
6
32
< 0.001**
   Positive
107
20
94
17
13
68
 
Lymphatic invasion
      
   Negative
466
83
461
85
5
26
< 0.001**
   Positive
93
17
79
15
14
74
 
Vascular invasion
      
   Negative
427
76
422
78
5
26
< 0.001**
   Positive
132
24
118
22
14
74
 
* Mann-Whitney's U test
** Chi-square for independence test
No., number of patients; BI, Brinkman index = (number of cigarettes per day) × (duration of years); SMPC, stromal micropapillary component; AMPC, aerogeneous micropapillary component

Survival analysis

Among all stage patients, median follow-up time was 654 days (range, 33-1512 days) in SMPC(-) tumors, 240 days (range, 28-661 days) in SMPC(+) tumors, 664 days (range, 28-1512 days) in AMPC(-) tumors, and 467 days (range, 36-1412 days) in AMPC(+) tumors. Among the stage I patients, median follow-up time was 767 days (range, 59-1343 days) in SMPC(-) tumors, 192 days (range, 227-485 days) in SMPC(+) tumors, 767 days (range, 59-1343 days) in AMPC(-) tumors, and 836 days (range, 140-1233 days) in AMPC(+) tumors. Recurrence occurred in 28 of 559 cases. SMPC(+) tumors recurred in 4 of 19 in all stage and in 2 of 10 in p-stage I, and AMPC(+) tumors recurred in 8 of 99 cases and 4 of 69 cases, respectively. In all stage, disease-free survival (DFS) of patients with SMPC(+) tumors was significantly poorer than that in patients with SMPC(-) tumors (Figure 3A, P < 0.001); the same result was observed in patients with AMPC(+) and AMPC(-) tumors (Figure 3B, P = 0.045,). In p-stage I patients, DFS of those with SMPC(+) tumors showed significantly poorer outcome than that of patients with SMPC(-) tumors (Figure 3C, P < 0.001); the same result was observed between patients with AMPC(+) and AMPC(-) tumors (Figure 3D, P = 0.023).
In univariate analysis, high pathological stage (P < 0.001), pleural invasion (P < 0.001), lymphatic invasion (P < 0.001), vascular invasion (P < 0.001), SMPC(+) (P < 0.001), and AMPC(+) tumors (P = 0.045) showed an unfavorable influence on survival for all stage, and pleural invasion (P < 0.001), lymphatic invasion (P < 0.001), vascular invasion (P < 0.001), SMPC(+) (P < 0.001), and AMPC(+) tumors (P = 0.023) showed an unfavorable influence on survival for p-stage I (Table 3, 4). In multivariate analysis, pathological stage (P = 0.028), lymphatic invasion (P = 0.009), and vascular invasion (P = 0.011) were identified as significant independent prognostic factors for all stage (Table 3). Though not observed for all stage, the presence of SMPC(+) tumors (P = 0.035) was identified as a significant independent prognostic factor for p-stage I, as well as lymphatic invasion (P = 0.020) and vascular invasion (P = 0.049) (Table 4). The presence of AMPC(+) tumors was not a significant prognostic factor for all stage or p-stage I.
Table 3
Impact of potential prognostic factors on DFS of patients of lung adenocarcinoma in all stage by univariate and multivariate analysis
 
No.
%
Univariate
analysis
Multivariate analysis
   
P value
Hazard ratio
95% CI
P value
Total
559
     
Age
      
   < 65
213
38
0.388
1.000
  
   ≥ 65
346
62
 
1.933
0.849-4.402
0.116
Gender
      
   Female
288
52
0.768
1.000
  
   Male
271
48
 
0.807
0.232-2.803
0.735
Smoking status
      
   Non-smoker
284
49
0.560
1.000
  
   Smoker
275
51
 
1.164
0.342-3.956
0.808
Tumor size
      
   < 30 mm
396
71
0.059
1.000
  
   ≥ 30 mm
163
29
 
0.819
0.338-1.985
0.658
Pathological stage
      
   I
458
82
<0.001
1.000
  
   II, III, IV
101
18
 
2.768
1.113-6.884
0.028
Pleural invasion
      
   Negative
452
81
<0.001
1.000
  
   Positive
107
19
 
0.848
0.345-2.083
0.719
Lymphatic invasion
      
   Negative
466
83
<0.001
1.000
  
   Positive
93
17
 
3.430
1.363-8.634
0.009
Vascular invasion
      
   Negative
427
76
<0.001
1.000
  
   Positive
132
24
 
3.309
1.312-8.350
0.011
SMPC
      
   Negative
540
97
<0.001
1.000
  
   Positive
19
3
 
1.871
0.528-6.630
0.332
AMPC
      
   Negative
460
83
0.045
1.000
  
   Positive
99
17
 
1.132
0.450-2.845
0.792
DFS, disease free survival; No., number of patients; SMPC, stromal micropapillary component; AMPC, aerogeneous micropapillary component; CI, confidence interval.
Table 4
Impact of potential prognostic factors on DFS of patients of lung adenocarcinoma in p-stage I by univariate and multivariate analysis
 
No.
%
Univariate
Analysis
Multivariate analysis
   
P value
Hazard ratio
95% CI
P value
Total
458
     
Age
      
   < 65
172
38
0.394
1.000
  
   ≥ 65
286
62
 
2.191
0.474-10.131
0.316
Gender
      
   Female
249
54
0.063
1.000
  
   Male
209
46
 
0.157
0.014-1.787
0.136
Smoking status
      
   Non-smoker
248
54
0.204
1.000
  
   Smoker
210
46
 
0.768
0.117-5.052
0.784
Tumor size
      
   < 30 mm
358
78
0.264
1.000
  
   ≥ 30 mm
100
22
 
0.304
0.037-2.504
0.268
Pleural invasion
      
   Negative
402
88
< 0.001
1.000
  
   Positive
56
12
 
1.519
0.328-7.040
0.593
Lymphatic invasion
      
   Negative
415
91
< 0.001
1.000
  
   Positive
43
9
 
5.016
1.295-19.434
0.020
Vascular invasion
      
   Negative
390
85
< 0.001
1.000
  
   Positive
68
15
 
4.494
1.006-20.081
0.049
SMPC
      
   Negative
448
98
< 0.001
1.000
  
   Positive
10
2
 
9.028
1.164-70.031
0.035
AMPC
      
   Negative
389
98
0.023
1.000
  
   Positive
69
2
 
1.825
0.378-8.808
0.454
DFS, disease free survival; No., number of patients; SMPC, stromal micropapillary component; AMPC, aerogeneous micropapillary component; CI, confidence interval.

Immunohistochemical findings

We evaluated immunohistochemical profiles of SMPC, AMPC, and nMPC. These lesions were evaluated in TMAs for 33 cases, including 19 SMPC(+) tumors and 14 pure AMPC tumors. The latter 14 tumors were selected from 85 pure AMPC tumors according to operation date, patient age, gender, and smoking status to match clinical background factors between SMPC and AMPC. nMPC was generally included in TMA cores of SMPC and AMPC. The total number of TMA was 19 SMPC and 28 AMPC. Staining scores are summarized in Table 5.
Table 5
Staining Scores in SMPC, AMPC and nMPC lesions
Classification/Antibody
SMPC
AMPC
nMPC
Cellular adhesion molecules
   
   E-cadherin
215.3*
143.9
187.1
   CD44
60.8
205.9
141.3
   Laminin5γ2
69.4
36.9
60.3
Growth factor
  
   VEGF-C
294.4
296.4
282.1
Apoptosis-associated proteins
  
   bcl2
13.2
11.1
21.8
   p53§
36.4
26.1
45.0
   cleaved caspase-3§
0.3
0.2
0.4
Mucin-related proteins
   
   MUC1
169.7
182.5
202.1
   MUC6
0.0
0.0
0.0
Hypoxia induced protein
   
   HIF-1α
1.8
2.4
2.9
Others
   
   TTF-1
267.9
289.3
248.6
   SP-A
45.2
82.6
123.2
   Vimentin
112.1
117.9
72.1
   Ki-67§
22.6
16.9
16.2
   LYVE1
98.9
107.2
101.9
   c-Met
217.2
253.8
211.4
   Phospho-c-Met
34.2
50.0
88.0
SMPC, stromal micropapillary component; AMPC, aerogeneous micropapillary component; nMPC, non-micropapillary component; VEGF-C, vascular endothelial growth factor-C; HIF-1α, hypoxia induced factor 1-α; TTF-1, thyroid transcription factor-1; SP-A, surfactant apoprotein A; LYVE1, lymphatic vessel endothelial hyaluronan receptor 1.
§ Positivity rate of positive tumor cells in 300 tumor cells (percentage).
* The difference in staining scores between SMPC and AMPC is statistically significant (P = 0.020).
‡ The differences in staining scores between SMPC and nMPC are statistically significant for CD44 (P = 0.011), and SP-A (P = 0.024) and phospho-c-Met (P = 0.011) expression.
¶ The difference in staining scores between AMPC and nMPC is statistically significant (P = 0.015).
In cellular adhesion molecules, E-cadherin staining scores in patients with SMPC, AMPC, and nMPC were 215.3, 143.9, and 187.1, respectively, and although the differences were not significant between patients with SMPC or nMPC and between patients with AMPC or nMPC (P = 0.312, 0.127, respectively), staining scores of SMPC were significantly higher than those for patients with AMPC (P = 0.020) (Figure 4A-C). CD44 staining scores in SMPC, AMPC, and nMPC were 60.8, 205.9, and 141.3, respectively. The CD44 expression level in SMPC was significantly lower than in AMPC (P < 0.001) and significantly higher than that in nMPC lesions (P = 0.015) (Figure 4D-F).
For other antibodies, staining scores of surfactant apoprotein A (SP-A) in the SMPC, AMPC, and nMPC were 45.2, 82.6, and 123.2, respectively, and although the difference was not significant between AMPC and nMPC (P = 0.203), the staining score in SMPC was significantly lower than those in nMPC (P = 0.024) (Figure 4G-I). Similarly, staining scores of phospho-c-Met in SMPC, AMPC, and nMPC were 34.2, 50.5, 88.0, respectively, and staining scores in SMPC were significantly lower than those in nMPC (Figure 4J-L).

Mutation analysis

Mutation analysis was performed in 33 patients for whom TMAs were constructed for immunohistochemical analysis. Table 6 summarizes the results of the mutation analysis. Although no cases examined possessed the KRAS mutations, EGFR mutations were detected in 20 cases (61%): 14 in patients with SMPC(+) tumors (74%) and 6 in patients with SMPC(-) tumors (43%). There was no significant association between the existence of SMPC and EGFR mutations. Among the 20 cases with EGFR mutations, 7 had deletions at exon 19, 13 had a point mutation at exon 21, and there were no multiple mutations. Among the 13 cases with a point mutation at exon 21, 12 had an L858R mutation and one had an L861Q mutation.
Table 6
Mutation analysis
 
total
%
SMPC(+) cases
%
SMPC(-) cases
%
P value
No.
33
 
19
 
14
  
EGFR mutation
       
   Negative
13
39
5
26
8
57
0.076*
   Positive
20
61
14
74
6
43
 
   ex19
7
35§
5
36§
2
33§
0.664*
   ex21
13
65§
9
64§
4
67§
 
KRAS mutation
       
   Negative
33
100
19
100
14
100
-
   Positive
0
0
0
 
0
  
No., number of patients; EGFR, epidermal growth factor receptor; SMPC, stromal micropapillary component
* Fisher's exact probability test
§ Rate of positive cases at ex19 and 21 in EGFR mutation positive cases, respectively.

Discussion

The present study revealed the incidence of SMPC(+) lung adenocarcinoma in consecutive surgical cases to be 3.4%, which is lower than that of AMPC(+) lung adenocarcinoma (17.7%). In non-pulmonary organs, the incidence of invasive micropapillary carcinoma was reported to be 7% in breast carcinoma [14], 0.9% in urinary bladder cancer [15], and 9.4% in colon cancer [16]. Generally, invasive micropapillary carcinomas occur infrequently in any organ.
Prognosis of lung adenocarcinoma with MPC has been reported to be worse and have the potential for high malignancy [17, 18], but no studies have separately evaluated SMPC and AMPC. We showed that SMPC(+) tumors as well as AMPC(+) tumors are associated with several biological factors including tumor size, lymph node metastasis, advanced stage disease, and pleural and lymphovascular invasion. Univariate analysis also revealed the presence of SMPC and AMPC as a significant predictor of unfavorable outcome. However, the most remarkable finding was observed in multivariate analysis: among the patients in p-stage I, patients with not AMPC but SMPC showed a significantly poorer DFS than those without MPC. We used immunohistochemistry with monoclonal antibody D2-40 against lymphatic endothelium in TMA specimens and found that lymphatic vessels are involved within SMPC areas in 4 (21%) of 19 SMPC(+) tumors (data not shown). When compared with AMPC(+) tumors, SMPC(+) tumors significantly more often showed pleural, lymphatic, and vascular invasion than AMPC(+) tumors (68% vs. 33%, P = 0.004; 74% vs. 30%, P < 0.001; 74% vs. 41%, P = 0.010, respectively). Therefore, these data suggest that a strong association between SMPC(+) tumors and pleural and lymphovascular invasion may in part explain their aggressive behavior.
Moreover, we investigated the immunohistochemical differences between SMPC and AMPC. In the study, we observed high E-cadherin expression and low CD44 expression in SMPC. Phospho-c-Met expression generally decreases in SMPC to a greater extent than in AMPC. Recently, it has been suggested that E-cadherin repression and CD44 expression are associated with the epithelial-mesenchymal transition (EMT), which was thought to lead to tumor invasion [19, 20]. Additionally, Elliot et al. reported that hepatocyte growth factor (HGF) and c-Met signaling promotes EMT in breast cancer [21], and Orian-Rousseau et al. reported that CD44 is strictly required for c-Met activation by HGF in human carcinoma [22]. Consistent with these data, EMT may not occur in SMPC despite its existence in the stroma, or invasion of SMPC may occur through a different invasion mechanism from EMT. Our immunohistochemical findings of SMPC showed lower expression of SP-A than that of nMPC. Many studies have reported that SP-A deletion is correlated with patient survival, and reduced SP-A in MPC may be an excellent indicator for poor prognosis in small-size lung adenocarcinoma [23, 24]. Reduced SP-A may contribute to an unfavorable outcome of SMPC(+) tumors.
Some studies have reported a significant association between the presence of MPC and EGFR mutations and effectiveness of EGFR tyrosine kinase inhibitor (EGFR-TKI) for MPC(+) tumors [2528]. Since SMPC of lung adenocarcinoma may be associated with a high incidence of EGFR mutations, EGFR-TKI may be effective against SMPC(+) tumors. Patients with these pathological features of lung adenocarcinoma may benefit from EGFR-TKI as postoperative chemotherapy or first-line chemotherapy of relapsed lung adenocarcinoma.
In conclusion, we observed SMPC(+) adenocarcinoma. The incidence of SMPC(+) tumors is low, and SMPC(+) tumors have a different prognostic impact compared to AMPC(+) tumors. Particularly for the early stage tumors, SMPC(+) tumors have different pathobiological characteristics from AMPC(+) tumors, and SMPC(+) tumors frequently contain the EGFR mutation. Therefore, it is important to determine the presence of SMPC in lung adenocarcinoma, particularly p-stage I tumors, and the presence of SMPC should be noted in a pathology report to alert the clinician to the possibility of poor prognosis.

Acknowledgements

The authors thank Ms. Yoshihara for TMA synthesis.
This article was supported by Kanagawa Cancer Research Fund and Grant for Collaboration between Hospital and Research Institute, Kanagawa Cancer Center.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

MO and TY designed the study, performed clinical and pathological investigation, and wrote the drafts. YS and YM participated in pathological and genetical investigation. NO participated in statistical investigation. SO performed the histological and immunohistochemical evaluation. CH assisted the clinical investigation. HN participated in managing and operating the patients. YK assisted the pathological investigation. KY participated in collecting clinical data and images. TI participated in its design and coordination and helped to draft the manuscript. All authors read and approved the final manuscript.
Anhänge

Authors’ original submitted files for images

Literatur
1.
Zurück zum Zitat Travis WD, et al.: International Association for the Study of Lung Cancer/American Thoracic Society/European Respiratory Society international multidisciplinary classification of lung adenocarcinoma. J Thorac Oncol. 2011, 6: 244-285. 10.1097/JTO.0b013e318206a221.PubMedCentralCrossRefPubMed Travis WD, et al.: International Association for the Study of Lung Cancer/American Thoracic Society/European Respiratory Society international multidisciplinary classification of lung adenocarcinoma. J Thorac Oncol. 2011, 6: 244-285. 10.1097/JTO.0b013e318206a221.PubMedCentralCrossRefPubMed
2.
Zurück zum Zitat Yoshizawa A, Motoi N, Riely GJ, Sima CS, Gerald WL, Kris MK, Park BJ, Rusch VW, Travis WD: Impact of proposed IASLC/ATS/ERS classification of lung adenocarcinoma: prognostic subgroups and implications for further revision of staging based on analysis of 514 stage I cases. Mod Pathol. 2011, 24: 653-664. 10.1038/modpathol.2010.232.CrossRefPubMed Yoshizawa A, Motoi N, Riely GJ, Sima CS, Gerald WL, Kris MK, Park BJ, Rusch VW, Travis WD: Impact of proposed IASLC/ATS/ERS classification of lung adenocarcinoma: prognostic subgroups and implications for further revision of staging based on analysis of 514 stage I cases. Mod Pathol. 2011, 24: 653-664. 10.1038/modpathol.2010.232.CrossRefPubMed
3.
Zurück zum Zitat Amin MB, Tamboli P, Merchant SH, Ordóñez NG, Ro J, Ayala AG, Ro JY: Micropapillary component in lung adenocarcinoma: a distinctive histologic feature with possible prognostic significance. Am J Surg Pathol. 2002, 26: 358-364. 10.1097/00000478-200203000-00010.CrossRefPubMed Amin MB, Tamboli P, Merchant SH, Ordóñez NG, Ro J, Ayala AG, Ro JY: Micropapillary component in lung adenocarcinoma: a distinctive histologic feature with possible prognostic significance. Am J Surg Pathol. 2002, 26: 358-364. 10.1097/00000478-200203000-00010.CrossRefPubMed
4.
Zurück zum Zitat Miyoshi T, Satoh Y, Okumura S, Nakagawa K, Shirakusa T, Tsuchiya E, Ishikawa Y: Early-stage lung adenocarcinoma with micropapillary pattern, a distinct pathologic marker for a significantly poor prognosis. Am J Surg Pathol. 2003, 27: 101-109. 10.1097/00000478-200301000-00011.CrossRefPubMed Miyoshi T, Satoh Y, Okumura S, Nakagawa K, Shirakusa T, Tsuchiya E, Ishikawa Y: Early-stage lung adenocarcinoma with micropapillary pattern, a distinct pathologic marker for a significantly poor prognosis. Am J Surg Pathol. 2003, 27: 101-109. 10.1097/00000478-200301000-00011.CrossRefPubMed
5.
Zurück zum Zitat Maeda R, Isowa N, Onuma H, Miura H, Harada T, Touge H, Tokuyasu H, Kawasaki Y: Lung adenocarcinomas with micropapillary components. Gen Thorac Cardiovasc Surg. 2009, 57: 534-539. 10.1007/s11748-009-0436-y.CrossRefPubMed Maeda R, Isowa N, Onuma H, Miura H, Harada T, Touge H, Tokuyasu H, Kawasaki Y: Lung adenocarcinomas with micropapillary components. Gen Thorac Cardiovasc Surg. 2009, 57: 534-539. 10.1007/s11748-009-0436-y.CrossRefPubMed
6.
Zurück zum Zitat Nagano T, Ishii G, Nagai K, Ito T, Kawase A, Takahashi K, Nishimura Y, Noshiwaki Y, Ochiai A: Structural and biological properties of a papillary component generating a micropapillary component in lung adenocarcinoma. Lung Cancer. 2010, 67: 282-289. 10.1016/j.lungcan.2009.04.014.CrossRefPubMed Nagano T, Ishii G, Nagai K, Ito T, Kawase A, Takahashi K, Nishimura Y, Noshiwaki Y, Ochiai A: Structural and biological properties of a papillary component generating a micropapillary component in lung adenocarcinoma. Lung Cancer. 2010, 67: 282-289. 10.1016/j.lungcan.2009.04.014.CrossRefPubMed
7.
Zurück zum Zitat Roh MS, Lee JI, Choi PJ, Hong YS: Relationship between micropapillary component and micrometastasis in the regional lymph nodes of patients with stage I lung adenocarcinoma. Histopathology. 2004, 45: 580-586. 10.1111/j.1365-2559.2004.01953.x.CrossRefPubMed Roh MS, Lee JI, Choi PJ, Hong YS: Relationship between micropapillary component and micrometastasis in the regional lymph nodes of patients with stage I lung adenocarcinoma. Histopathology. 2004, 45: 580-586. 10.1111/j.1365-2559.2004.01953.x.CrossRefPubMed
8.
Zurück zum Zitat Kamiya K, Hayashi Y, Douguchi J, Hashiguchi A, Yamada T, Izumi Y, Watanabe M, Kawamura M, Horinouchi H, Shimada N, Kobayashi K, Sakamoto M: Histopathological features and prognostic significance of the micropapillary pattern in lung adenocarcinoma. Mod Pathol. 2008, 21: 992-1001. 10.1038/modpathol.2008.79.CrossRefPubMed Kamiya K, Hayashi Y, Douguchi J, Hashiguchi A, Yamada T, Izumi Y, Watanabe M, Kawamura M, Horinouchi H, Shimada N, Kobayashi K, Sakamoto M: Histopathological features and prognostic significance of the micropapillary pattern in lung adenocarcinoma. Mod Pathol. 2008, 21: 992-1001. 10.1038/modpathol.2008.79.CrossRefPubMed
9.
Zurück zum Zitat Ohe M, Yokose T, Sakuma Y, Osanai S, Hasegawa C, Washimi K, Nawa K, Woo T, Hamanaka R, Nakayama H, Kameda K, Yamada K, Isobe T: Stromal micropapillary pattern predominant lung adenocarcinoma - A report of two cases. Diagn Pathol. 2011, 6: 92-10.1186/1746-1596-6-92.PubMedCentralCrossRefPubMed Ohe M, Yokose T, Sakuma Y, Osanai S, Hasegawa C, Washimi K, Nawa K, Woo T, Hamanaka R, Nakayama H, Kameda K, Yamada K, Isobe T: Stromal micropapillary pattern predominant lung adenocarcinoma - A report of two cases. Diagn Pathol. 2011, 6: 92-10.1186/1746-1596-6-92.PubMedCentralCrossRefPubMed
10.
Zurück zum Zitat Kuroda N, Hamaguchi N, Takeuchi E, Ohara M, Hirouchi T, Mizuno K: Lung adenocarcinoma with a micropapillary pattern: a clinicopathological study of 25 cases. APMIS. 2006, 114: 381-385. 10.1111/j.1600-0463.2006.apm_340.x.CrossRefPubMed Kuroda N, Hamaguchi N, Takeuchi E, Ohara M, Hirouchi T, Mizuno K: Lung adenocarcinoma with a micropapillary pattern: a clinicopathological study of 25 cases. APMIS. 2006, 114: 381-385. 10.1111/j.1600-0463.2006.apm_340.x.CrossRefPubMed
11.
Zurück zum Zitat TNM classification of malignant tumors. Edited by: Sobin LH, Gospodarowicz MK, Wittekind C. 2009, Wiley-Blackwell, seventh TNM classification of malignant tumors. Edited by: Sobin LH, Gospodarowicz MK, Wittekind C. 2009, Wiley-Blackwell, seventh
12.
Zurück zum Zitat Tsuta K, Ishii G, Nitadori J, Murata Y, Kodama T, Nagai K, Ochiai A: Comparison of the immunophenotypes of signet-ring cell carcinoma, solid adenocarcinoma with mucin production, and mucinous bronchioloalveolar carcinoma of the lung characterized by the presence of cytoplasmic mucin. J Pathol. 2006, 209: 78-87. 10.1002/path.1947.CrossRefPubMed Tsuta K, Ishii G, Nitadori J, Murata Y, Kodama T, Nagai K, Ochiai A: Comparison of the immunophenotypes of signet-ring cell carcinoma, solid adenocarcinoma with mucin production, and mucinous bronchioloalveolar carcinoma of the lung characterized by the presence of cytoplasmic mucin. J Pathol. 2006, 209: 78-87. 10.1002/path.1947.CrossRefPubMed
13.
Zurück zum Zitat Matsukuma S, Yoshihara M, Kasai F, Kato A, Yoshida A, Akaike M, Kobayashi O, Nakayama H, Sakuma Y, Yoshida T, Kameda Y, Ysuchiya E, Miyagi Y: Rapid and simple detection of hot spot point mutations of epidermal growth factor receptor, BRAF, and NRAS in cancers using the loop-hybrid mobility shift assay. J Mol Diagn. 2006, 8: 504-512. 10.2353/jmoldx.2006.060030.PubMedCentralCrossRefPubMed Matsukuma S, Yoshihara M, Kasai F, Kato A, Yoshida A, Akaike M, Kobayashi O, Nakayama H, Sakuma Y, Yoshida T, Kameda Y, Ysuchiya E, Miyagi Y: Rapid and simple detection of hot spot point mutations of epidermal growth factor receptor, BRAF, and NRAS in cancers using the loop-hybrid mobility shift assay. J Mol Diagn. 2006, 8: 504-512. 10.2353/jmoldx.2006.060030.PubMedCentralCrossRefPubMed
14.
Zurück zum Zitat Walsh MM, Bleiweiss IJ: Invasive micropapillary carcinoma of the breast: eighty cases of an under-recognized entity. Hum Pathol. 2001, 32: 583-589. 10.1053/hupa.2001.24988.CrossRefPubMed Walsh MM, Bleiweiss IJ: Invasive micropapillary carcinoma of the breast: eighty cases of an under-recognized entity. Hum Pathol. 2001, 32: 583-589. 10.1053/hupa.2001.24988.CrossRefPubMed
15.
Zurück zum Zitat Johansson SL, Borghede G, Holmang S: Micropapillary bladder carcinoma: a clinicopathological study of 20 cases. J Urol. 1999, 161: 1798-1802. 10.1016/S0022-5347(05)68807-6.CrossRefPubMed Johansson SL, Borghede G, Holmang S: Micropapillary bladder carcinoma: a clinicopathological study of 20 cases. J Urol. 1999, 161: 1798-1802. 10.1016/S0022-5347(05)68807-6.CrossRefPubMed
16.
Zurück zum Zitat Kim M, Hong S, Jang S, Yu E, Kim J, Kim K, Gong G, Ro J: Invasive colorectal micropapillary carcinoma: an aggressive variant of adenocarcinoma. Hum Pathol. 2006, 37: 809-815. 10.1016/j.humpath.2005.10.018.CrossRefPubMed Kim M, Hong S, Jang S, Yu E, Kim J, Kim K, Gong G, Ro J: Invasive colorectal micropapillary carcinoma: an aggressive variant of adenocarcinoma. Hum Pathol. 2006, 37: 809-815. 10.1016/j.humpath.2005.10.018.CrossRefPubMed
17.
Zurück zum Zitat Nora S, Presmanes MC, Monroy V, Alandro MH, Alvarez-Fernandez E: Micropapillary lung adenocarcinoma: a distinctive histologic subtype with prognostic significance. Cases series. Hum Pathol. 2008, 39: 324-330. 10.1016/j.humpath.2007.05.029.CrossRef Nora S, Presmanes MC, Monroy V, Alandro MH, Alvarez-Fernandez E: Micropapillary lung adenocarcinoma: a distinctive histologic subtype with prognostic significance. Cases series. Hum Pathol. 2008, 39: 324-330. 10.1016/j.humpath.2007.05.029.CrossRef
18.
Zurück zum Zitat Kamiya K, Hayashi Y, Douguchi J, Hashiguchi A, Yamada T, Izumi Y, Watanabe M, Kawamura M, Horinouchi H, Shimada N, Kobayashi K, Sakamoto M: Histopathological features and prognostic significance of the micropapillary pattern in lung adenocarcinoma. Mod Pathol. 2008, 21: 992-1001. 10.1038/modpathol.2008.79.CrossRefPubMed Kamiya K, Hayashi Y, Douguchi J, Hashiguchi A, Yamada T, Izumi Y, Watanabe M, Kawamura M, Horinouchi H, Shimada N, Kobayashi K, Sakamoto M: Histopathological features and prognostic significance of the micropapillary pattern in lung adenocarcinoma. Mod Pathol. 2008, 21: 992-1001. 10.1038/modpathol.2008.79.CrossRefPubMed
19.
Zurück zum Zitat Nieto MA: Epithelial-mesenchymal transitions in development and disease: old views and new perspectives. Int J Dev Biol. 2009, 53: 1541-1547. 10.1387/ijdb.072410mn.CrossRefPubMed Nieto MA: Epithelial-mesenchymal transitions in development and disease: old views and new perspectives. Int J Dev Biol. 2009, 53: 1541-1547. 10.1387/ijdb.072410mn.CrossRefPubMed
20.
Zurück zum Zitat Ponta H, Sherman L, Herrlich PA: CD44: from adhesion molecules to signaling regulators. Nat Rev Mol Cell Biol. 2003, 4: 33-45. 10.1038/nrm1004.CrossRefPubMed Ponta H, Sherman L, Herrlich PA: CD44: from adhesion molecules to signaling regulators. Nat Rev Mol Cell Biol. 2003, 4: 33-45. 10.1038/nrm1004.CrossRefPubMed
21.
Zurück zum Zitat Elliot BE, Hung WL, Boag AH, Tuck AB: The role of hepatocyte growth factor (scatter factor) in epithelial-mesenchymal transition and breast cancer. Can J Physiol Pharmacol. 2002, 80: 91-102. 10.1139/y02-010.CrossRef Elliot BE, Hung WL, Boag AH, Tuck AB: The role of hepatocyte growth factor (scatter factor) in epithelial-mesenchymal transition and breast cancer. Can J Physiol Pharmacol. 2002, 80: 91-102. 10.1139/y02-010.CrossRef
22.
Zurück zum Zitat Orian-Rousseau V, Chen L, Sleeman JP, Herrlich P, Ponta H: CD44 is required for two consecutive steps in HGF/c-Met signaling. Genes Dev. 2002, 16: 3074-3086. 10.1101/gad.242602.PubMedCentralCrossRefPubMed Orian-Rousseau V, Chen L, Sleeman JP, Herrlich P, Ponta H: CD44 is required for two consecutive steps in HGF/c-Met signaling. Genes Dev. 2002, 16: 3074-3086. 10.1101/gad.242602.PubMedCentralCrossRefPubMed
23.
Zurück zum Zitat Jiang F, Caraway NP, Bekele BN, Zhang HZ, Khanna A, Wang H, Li R, Fernandez RL, Zaidi TM, Johnston DA, Katz RL: Surfactant protein A gene deletion and prognostics for patients with stage I non-small cell lung cancer. Clin Cancer Res. 2005, 11: 5417-5424. 10.1158/1078-0432.CCR-04-2087.CrossRefPubMed Jiang F, Caraway NP, Bekele BN, Zhang HZ, Khanna A, Wang H, Li R, Fernandez RL, Zaidi TM, Johnston DA, Katz RL: Surfactant protein A gene deletion and prognostics for patients with stage I non-small cell lung cancer. Clin Cancer Res. 2005, 11: 5417-5424. 10.1158/1078-0432.CCR-04-2087.CrossRefPubMed
24.
Zurück zum Zitat Tsutsumida H, Nomoto M, Goto M, Kitajima S, Kubota I, Hirotsu Y, Wakimoto J, Hollingsworth MA, Yonezawa S: A micropapillary pattern is predictive of a poor prognosis in lung adenocarcinoma, and reduced surfactant apoprotein A expression in the micropapillary pattern is an excellent indicator of a poor prognosis. Modern Pathol. 2007, 20: 638-647. 10.1038/modpathol.3800780.CrossRef Tsutsumida H, Nomoto M, Goto M, Kitajima S, Kubota I, Hirotsu Y, Wakimoto J, Hollingsworth MA, Yonezawa S: A micropapillary pattern is predictive of a poor prognosis in lung adenocarcinoma, and reduced surfactant apoprotein A expression in the micropapillary pattern is an excellent indicator of a poor prognosis. Modern Pathol. 2007, 20: 638-647. 10.1038/modpathol.3800780.CrossRef
25.
Zurück zum Zitat Ninomiya H, Hiramatsu M, Inamura K, Nomura K, Okui M, Miyoshi T, Okumura S, Satoh Y, Nakagawa K, Nishio M, Horai T, Miyata S, Tsuchiya E, Fukuyama E, Ishikawa Y: Correlation between morphology and EGFR mutations in lung adenocarcinomas Significance of the micropapillary pattern and the hobnail cell type. Lung Cancer. 2009, 63: 235-240. 10.1016/j.lungcan.2008.04.017.CrossRefPubMed Ninomiya H, Hiramatsu M, Inamura K, Nomura K, Okui M, Miyoshi T, Okumura S, Satoh Y, Nakagawa K, Nishio M, Horai T, Miyata S, Tsuchiya E, Fukuyama E, Ishikawa Y: Correlation between morphology and EGFR mutations in lung adenocarcinomas Significance of the micropapillary pattern and the hobnail cell type. Lung Cancer. 2009, 63: 235-240. 10.1016/j.lungcan.2008.04.017.CrossRefPubMed
26.
Zurück zum Zitat Inamura K, Ninomiya H, Ishikawa Y, Matsubara O: Is the epidermal growth factor receptor status in lung cancers reflected in clinicopathologic features?. Arch Pathol Lab Med. 2010, 134: 66-72.PubMed Inamura K, Ninomiya H, Ishikawa Y, Matsubara O: Is the epidermal growth factor receptor status in lung cancers reflected in clinicopathologic features?. Arch Pathol Lab Med. 2010, 134: 66-72.PubMed
27.
Zurück zum Zitat Motoi N, Szoke J, Riely GJ, Seshan VE, Kris MG, Rusch VW, Gerald WL, Travis WD: Lung adenocarcinoma: modification of the 2004 WHO mixed subtype to include the major histologic subtype suggests correlations between papillary and micropapillary adenocarcinoma subtypes, EGFR mutations and gene expression analysis. Am J Surg Pathol. 2008, 32: 810-827. 10.1097/PAS.0b013e31815cb162.CrossRefPubMed Motoi N, Szoke J, Riely GJ, Seshan VE, Kris MG, Rusch VW, Gerald WL, Travis WD: Lung adenocarcinoma: modification of the 2004 WHO mixed subtype to include the major histologic subtype suggests correlations between papillary and micropapillary adenocarcinoma subtypes, EGFR mutations and gene expression analysis. Am J Surg Pathol. 2008, 32: 810-827. 10.1097/PAS.0b013e31815cb162.CrossRefPubMed
28.
Zurück zum Zitat Kim YH, Ishii G, Goto K, Nagai K, Tsuta K, Shiono S, Nitadori J, Kodama T, Nishiwaki Y, Ochiai A: Dominant papillary subtype is a significant predictor of the response to gefitinib in adenocarcinoma of the lung. Clin Cancer Res. 2004, 10: 7311-7317. 10.1158/1078-0432.CCR-04-0811.CrossRefPubMed Kim YH, Ishii G, Goto K, Nagai K, Tsuta K, Shiono S, Nitadori J, Kodama T, Nishiwaki Y, Ochiai A: Dominant papillary subtype is a significant predictor of the response to gefitinib in adenocarcinoma of the lung. Clin Cancer Res. 2004, 10: 7311-7317. 10.1158/1078-0432.CCR-04-0811.CrossRefPubMed
Metadaten
Titel
Stromal micropapillary component as a novel unfavorable prognostic factor of lung adenocarcinoma
verfasst von
Miki Ohe
Tomoyuki Yokose
Yuji Sakuma
Yohei Miyagi
Naoyuki Okamoto
Sachie Osanai
Chikako Hasegawa
Haruhiko Nakayama
Yoichi Kameda
Kouzo Yamada
Takeshi Isobe
Publikationsdatum
01.12.2012
Verlag
BioMed Central
Erschienen in
Diagnostic Pathology / Ausgabe 1/2012
Elektronische ISSN: 1746-1596
DOI
https://doi.org/10.1186/1746-1596-7-3

Weitere Artikel der Ausgabe 1/2012

Diagnostic Pathology 1/2012 Zur Ausgabe

Neu im Fachgebiet Pathologie

Molekularpathologische Untersuchungen im Wandel der Zeit

Open Access Biomarker Leitthema

Um auch an kleinen Gewebeproben zuverlässige und reproduzierbare Ergebnisse zu gewährleisten ist eine strenge Qualitätskontrolle in jedem Schritt des Arbeitsablaufs erforderlich. Eine nicht ordnungsgemäße Prüfung oder Behandlung des …

Vergleichende Pathologie in der onkologischen Forschung

Pathologie Leitthema

Die vergleichende experimentelle Pathologie („comparative experimental pathology“) ist ein Fachbereich an der Schnittstelle von Human- und Veterinärmedizin. Sie widmet sich der vergleichenden Erforschung von Gemeinsamkeiten und Unterschieden von …

Gastrointestinale Stromatumoren

Open Access GIST CME-Artikel

Gastrointestinale Stromatumoren (GIST) stellen seit über 20 Jahren ein Paradigma für die zielgerichtete Therapie mit Tyrosinkinaseinhibitoren dar. Eine elementare Voraussetzung für eine mögliche neoadjuvante oder adjuvante Behandlung bei …

Personalisierte Medizin in der Onkologie

Aufgrund des erheblichen technologischen Fortschritts in der molekularen und genetischen Diagnostik sowie zunehmender Erkenntnisse über die molekulare Pathogenese von Krankheiten hat in den letzten zwei Jahrzehnten ein grundlegender …