Skip to main content
Erschienen in: Annals of Surgical Oncology 6/2016

Open Access 02.02.2016 | Thoracic Oncology

Minor Components of Micropapillary and Solid Subtypes in Lung Adenocarcinoma are Predictors of Lymph Node Metastasis and Poor Prognosis

verfasst von: Yue Zhao, MD, Rui Wang, MD, Xuxia Shen, MD, Yunjian Pan, MD, Chao Cheng, MD, Yuan Li, MD, Lei Shen, MD, Yang Zhang, MD, Hang Li, MD, Difan Zheng, Ting Ye, MD, Shanbo Zheng, MD, Yihua Sun, MD, Haiquan Chen, MD

Erschienen in: Annals of Surgical Oncology | Ausgabe 6/2016

Abstract

Background

Lung adenocarcinoma with micropapillary and solid predominant subtypes was reported to be associated with poor prognosis; however, whether minor components (non-predominant) of micropapillary and solid subtypes predict poor prognosis remains unknown. In this study, we investigated the predictive and prognostic value of lymph node metastasis of minor micropapillary and solid components.

Methods

Specimens of resected tumors of 1244 patients were reclassified to determine the predominant subtype and minor components (>5 %, but not predominant). Of these specimens, 105 contained a micropapillary component and 210 contained a solid component. The correlation between each subtype and lymph node metastasis was analyzed, and survival analyses were used to determine the association between each subtype and patient survival.

Results

Adenocarcinomas harboring micropapillary and/or solid components held higher rates of metastatic lymph node stations (25.2 % vs. 15.6 %, p = 0.002; and 24.0 % vs. 14.9 %, p < 0.001, respectively) and lymph nodes (17.3 % vs. 10.1 %, p = 0.004; and 15.5 % vs. 9.7 %, p = 0.001, respectively). Patients with micropapillary and solid components in their tumors showed a shorter median recurrence-free survival (15.8 vs. 62.8 months, p < 0.001; and 20.8 months vs. not reached, p < 0.001) and overall survival (47.0 months vs. not reached, p < 0.001; and 69.0 months vs. not reached, p < 0.001).

Conclusions

Minor components of micropapillary and/or solid subtypes of lung adenocarcinoma are correlated with lymph node metastasis and poor prognosis. Thus, it is beneficial to focus not only on predominant subtypes but also minor components to predict prognoses and make therapeutic strategies more comprehensively.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1245/​s10434-015-5043-9) contains supplementary material, which is available to authorized users.
Yue Zhao, Rui Wang and Xuxia Shen have contributed equally to this work.
Yihua Sun and Haiquan Chen have contributed equally to this work.
Adenocarcinoma has long been an independent histological class of lung cancer and has been broadly studied for therapeutic efficacy and toxicities.15 In 2011, a new classification system of subtypes of lung adenocarcinoma was recommended by the International Association for the Study of Lung Cancer (IASLC), American Thoracic Society (ATS), and European Respiratory Society (ERS) to further study and advance the field.6 Since then, a number of investigations have provided evidence for the relationship between different subtypes and treatment outcomes.711
Several studies have reported that micropapillary- and solid-predominant subtypes of lung adenocarcinoma were associated with poor prognoses;1216 however, lung adenocarcinomas usually contain complex mixtures of different subtypes.17 Whether minor components of each subtype are associated with lymph node metastasis and poor prognosis still remains unknown and needs further clarification.
In this study, we comprehensively analyzed 1244 consecutive patients who were diagnosed with stage I–IV invasive lung adenocarcinoma and who underwent surgical resection between August 2006 and May 2013. Our aim was to provide clinical evidence for the predictive and prognostic value of minor components of lung adenocarcinoma.

Patients and Methods

Patients and Tissues

Overall, 1244 consecutive patients who were diagnosed with invasive lung adenocarcinoma and who underwent surgical resection between August 2006 and May 2013 were included in this study. R0 resection was achieved in 1240 of the 1244 patients. Patients with no or insufficient archived tumor specimens were excluded, and no patient underwent neoadjuvant therapy. To ensure an accurate assessment, tumors were reclassified by three pathologists (XS, LS, and YL) and categorized into the following subtypes: lepidic (L), acinar (A), papillary (P), micropapillary (M), and solid (S) predominant subtypes, as well as invasive mucinous adenocarcinoma (IMA), according to the newly announced IASLC/ATS/ERS lung adenocarcinoma classification system.6 Each of the 1244 slides was reviewed by these three pathologists separately. Discordant results were reconsidered together by the three pathologists until a consensus was reached. Specimens that did not belong to any one of these categories were marked as ‘others’. For those specimens that were mixed by more than one subtype, the subtype that occupied most of the tumor (even if <50 %) was defined as the predominant subtype, and subtype(s) that occupied no less than 5 % but were not predominant were defined as minor components. We put them in a sequence from the largest to the smallest amount.
This study was approved by the Committee for Ethical Review of Research (Fudan University Shanghai Cancer Center IRB# 090977-1).

Statistical Analysis

Clinical and pathological characteristics, such as sex, age, smoking status, lymphovascular invasion status, tumor location, and nodal status (N0, N1, or N2), together with predominant subtypes and minor components, were analyzed to determine the correlation with lymph node metastasis. p values, hazard ratios and 95 % confidence intervals (CI) were calculated using Pearson’s Chi square test or Fisher’s exact test, and the two-tailed significance level was set at 0.05. Metastatic rates of lymph node stations and lymph nodes of each predominant subtype and minor component were calculated to evaluate the correlation between lymph node metastasis and each subtype. p values were calculated using Student’s t test. Moreover, Kaplan–Meier survival curves were used to compare patients’ recurrence-free survival (RFS) and overall survival (OS). Finally, a multivariate Cox model adjusted for sex, age, smoking status, lymphovascular invasion status, tumor location, and nodal status (N0, N1, or N2) was used to determine the odds ratio (OR) and 95 % CI for each minor component. All statistical analyses were performed using SPSS version 20.0 (IBM Corporation, Armonk, NY, USA).

Results

For the 1244 patients with pathologically validated lung adenocarcinoma included in this study, a reclassification by three pathologists manifested that there were 158 lepidic-predominant, 598 acinar-predominant, 170 papillary-predominant, 68 micropapillary-predominant, 171 solid-predominant, and 72 invasive mucinous adenocarcinoma. Clinical and pathological characteristics are shown in Table 1. Of the 1244 patients, 109 had a tumor containing a minor component of lepidic subtype, 196 a minor acinar component, 178 a minor papillary component, 105 a minor micropapillary component, 210 a minor solid component, and 62 had a tumor containing a minor invasive mucinous component. Patients were aged 60 years (range 22–110), 560 were males and 684 were females (Table 1). The pathologic stage was 0 in 6 (0.5 %) patients, IA in 517 (41.6 %) patients, IB in 178 (14.3 %) patients, IIA in 120 (9.6 %) patients, IIB in 46 (3.7 %) patients, IIIA in 338 (27.2 %) patients, IIIB in 15 (1.2 %) patients, and IV in 24 (1.9 %) patients.
Table 1
Clinicopathological characteristics of different subtypes of lung adenocarcinoma (n = 1244)
 
Total
N0 (n = 789)
N1/N2 (n = 455)
p value
HR (95 % CI)
Gender
 Male
560 (45.0 %)
327 (41.4 %)
233 (51.2 %)
0.001
1.282 (1.108–1.483)
 Female
684 (55.0 %)
462 (58.6 %)
222 (48.8 %)
  
Age
 < Average
598 (48.1 %)
360 (45.6 %)
238 (52.3 %)
0.023
1.185 (1.023–1.372)
 ≥ Average
646 (51.9 %)
429 (54.4 %)
217 (47.7 %)
  
Smoking status
 Former/current
403 (32.4 %)
225 (28.5 %)
178 (39.1 %)
<0.001
1.340 (1.159–1.553)
 Never
841 (67.6 %)
564 (71.5 %)
277 (60.9 %)
  
Tumor locationa
 LUL
330 (26.5 %)
193 (24.5 %)
137 (30.1 %)
0.033
0.838 (0.717–0.979)
 LLL
200 (16.1 %)
119 (15.1 %)
81 (17.8 %)
0.208
0.885 (0.734–1.066)
 RUL
406 (32.6 %)
274 (34.7 %)
132 (29.0 %)
0.038
1.186 (1.006–1.397)
 RML
103 (8.3 %)
64 (8.1 %)
39 (8.6 %)
0.777
0.963 (0.743–1.248)
 RLL
231 (18.6 %)
150 (19.0 %)
81 (17.8 %)
0.597
1.053 (0.868–1.277)
LVI
 Yes
203 (16.3 %)
35 (4.4 %)
168 (36.9 %)
<0.001
3.003 (2.674–3.378)
 No
1041 (83.7 %)
754 (95.6 %)
287 (63.1 %)
  
Differentiation
 Well
169 (13.6 %)
153 (19.4 %)
16 (3.5 %)
<0.001
0.232 (0.145–0.372)
 Moderate
714 (57.4 %)
492 (62.4 %)
222 (48.8 %)
<0.001
0.707 (0.612–0.818)
 Poor
361 (29.0 %)
144 (18.3 %)
217 (47.7 %)
<0.001
2.232 (1.946–2.558)
Tumor size (cm)
 < 3
756 (60.8 %)
572 (72.5 %)
184 (40.4 %)
<0.001
2.283 (1.965–2.646)
 ≥ 3
488 (39.2 %)
217 (27.5 %)
271 (59.6 %)
  
Predominant subtype
 L
158 (12.7 %)
154 (19.5 %)
4 (0.9 %)
<0.001
0.061 (0.023–0.161)
 A
598 (48.1 %)
211 (46.4 %)
387 (49.0 %)
0.363
0.935 (0.806–1.082)
 P
170 (13.7 %)
98 (12.4 %)
72 (15.8 %)
0.092
1.188 (0.979–1.441)
 M
68 (5.5 %)
35 (4.4 %)
33 (7.3 %)
0.035
1.353 (1.046–1.748)
 S
171 (13.7 %)
62 (7.9 %)
109 (24.0 %)
<0.001
1.976 (1.715–2.278)
 IMA
72 (5.8 %)
48 (6.1 %)
24 (5.3 %)
0.556
0.907 (0.648–1.267)
Minor components
 L
109 (8.8 %)
94 (11.9 %)
15 (3.3 %)
<0.001
0.355 (0.221–0.571)
 A
196 (15.8 %)
123 (15.6 %)
73 (16.0 %)
0.832
1.021 (0.838–1.247)
 P
178 (14.3 %)
97 (12.3 %)
81 (17.8 %)
0.008
1.297 (1.083–1.553)
 M
105 (8.4 %)
49 (6.2 %)
56 (12.3 %)
<0.001
1.522 (1.252–1.852)
 S
210 (16.9 %)
103 (13.1 %)
107 (23.5 %)
<0.001
1.513 (1.292–1.773)
 IMA
62 (5.0 %)
34 (4.3 %)
28 (6.2 %)
0.150
1.250 (0.941–1.661)
p value, HR, and 95 % CI were calculated using the Chi square test and Fisher’s exact test
HR hazard ratio, CI confidence interval, LUL left upper lobe, LLL left lower lobe, RUL right upper lobe, RML right middle lobe, RLL right lower lobe, LVI lymphovascular invasion, L lepidic, A acinar, P papillary, M micropapillary, S solid, IMA invasive mucinous adenocarcinoma
aWhen adding the percentage of this category together, a number more than 100 % may be obtained as some tumors occurred in more than one lobe

The Predictive Value of Lymph Node Metastasis

Of the 1244 patients, 789 (63.4 %) had pathologically validated N0 disease and 455 (36.6 %) had either pathologically validated N1 or N2 disease (Table 1). Our data showed that lymph node metastasis had a correlation with male sex (p < 0.001), younger age at initial diagnosis (p = 0.023), ever-smoker (p < 0.001), lymphovascular invasion (p < 0.001), poor differentiation (p < 0.001), and larger tumor size (p < 0.001). The micropapillary-predominant subtype (p = 0.035; HR 1.353; 95 % CI 1.046–1.748) and solid-predominant subtype (p < 0.001; HR 1.976; 95 % CI 1.715–2.278) were both associated with lymph node metastasis, along with the papillary minor component (p = 0.008; HR 1.297; 95 % CI 1.083–1.553), micropapillary minor component (p < 0.001; HR 1.522; 95 % CI 1.252–1.852), and solid minor component (p < 0.001; HR 1.513; 95 % CI 1.292–1.773). On the other hand, tumors presenting as well-differentiated (p < 0.001) or moderately differentiated (p < 0.001), lepidic-predominant subtype (p < 0.001), and lepidic minor component (p < 0.001) showed lower probabilities of having lymph node metastasis (Table 1).
To further investigate the correlation between different subtypes and lymph node metastasis, we also recorded the number of metastatic lymph node stations/lymph nodes and resected lymph node stations/lymph nodes, and made a calculation of metastatic rates (Tables 2, 3). Regional lymph node stations were defined as per the recommendations made by Mountain and Dresler in 1997.18 According to our data, tumors harboring the micropapillary minor component and solid minor component had higher metastatic rates of lymph node station (25.2 vs. 15.6 %, p = 0.002; and 24.0 vs. 14.9 %, p < 0.001, respectively) and lymph node (17.3 vs. 10.1 %, p = 0.004; and 15.5 vs. 9.7 %, p = 0.001, respectively). A similar trend was observed in tumors with micropapillary-predominant and solid-predominant subtypes (Tables 2, 3). Additionally, we found that patients with lepidic-predominant or minor component tumors had a lower probability of experiencing lymph node metastasis (also shown in Tables 2, 3). Furthermore, we also analyzed the relationship between the second predominant subtype and lymph node metastasis. All 606 patients with two or more presenting subtypes were included (see electronic supplementary Table 1), and this analysis showed a similar result as previous ones (see electronic supplementary Tables 2 and 3).
Table 2
Relationship between different subtypes of lung adenocarcinoma and metastatic rate of lymph node station (n = 1244)
Subtype
Negative (%)a
Minor component (%)
p value
Predominant (%)
p value
L
17.5
5.2
<0.001
8.2
<0.001
A
16.6
15.6
0.650
15.5
0.233
P
15.9
19.5
0.106
20.1
0.081
M
15.6
25.2
0.002
23.8
0.066
S
14.9
24.0
<0.001
28.7
<0.001
IMA
16.2
21.4
0.138
14.7
0.580
Metastatic rate of lymph node station = (number of metastatic lymph node stations/number of totally resected lymph node stations) × 100 %
L lepidic, A acinar, P papillary, M micropapillary, S solid, IMA invasive mucinous adenocarcinoma
aPercentage of patients with subtype of interest not observed or less than 5 %
Table 3
Relationship between different subtypes of lung adenocarcinoma and metastatic rate of lymph node (n = 1244)
Subtype
Negative (%)a
Minor component (%)
p value
Predominant (%)
p value
L
11.4
3.1
<0.001
6.3
<0.001
A
10.8
10.1
0.663
9.8
0.176
P
10.5
11.9
0.397
13.5
0.081
M
10.1
17.3
0.004
17.5
0.056
S
9.7
15.5
0.001
18.0
<0.001
IMA
10.6
12.7
0.441
9.7
0.687
Metastatic rate of lymph node = (number of metastatic lymph nodes/number of totally resected lymph nodes) × 100 %
L lepidic, A acinar, P papillary, M micropapillary, S solid, IMA invasive mucinous adenocarcinoma
aPercentage of patients with subtype of interest not observed or less than 5 %

Does Each Subtype Predict a Different Prognosis?

Of the 1244 patients, 288 lacked sufficient follow-up data; therefore, we included the remaining 956 patients in subsequent survival analysis. Seventy-eight of the 956 patients had a tumor containing a minor micropapillary component, and 124 had a tumor containing a minor solid component, along with 19 micropapillary-predominant subtypes and 161 solid-predominant subtypes (Fig. 1).
In univariate analysis, patients with tumors containing a minor component of micropapillary subtype had a significantly shorter RFS (p < 0.001) and OS (p < 0.001). A similar observation was made among patients with tumors containing a minor component of solid subtype, which also showed a significantly shorter RFS (p < 0.001) and OS (p < 0.001). Meanwhile, patients with tumors of micropapillary- or solid-predominant subtype also had significantly worse RFS (p = 0.006 and p < 0.001, respectively) and OS (p = 0.007 and p < 0.001, respectively).
In a multivariate analysis using the Cox model adjusted for sex, age, smoking status, lymphovascular invasion status, tumor location, and nodal status (N0, N1, or N2), we found that tumors with a minor component of micropapillary subtype were correlated with shorter RFS (p < 0.001; OR 0.524; 95 % CI 0.388–0.708) and OS (p = 0.012; OR 0.585; 95 % CI 0.385–0.890) (Table 4). Tumors with a minor component of solid subtype were also related to a worse RFS (p = 0.014; OR 0.728; 95 % CI 0.567–0.936) and OS (p = 0.039; OR 0.690; 95 % CI 0.484–0.982) (Table 4). No significant difference was observed between other minor components and patient survival.
Table 4
Multivariate analysis of recurrence-free survival and overall survival for patients with minor components of different subtypes of lung adenocarcinoma
Subtype
OR
95 % CI
p value
Recurrence-free survival
 Lepidic
1.361
0.905–2.041
0.139
 Acinar
1.104
0.83–1.453
0.485
 Papillary
0.978
0.752–1.271
0.866
 Micropapillary
0.524
0.388–0.708
<0.001
 Solid
0.728
0.567–0.936
0.014
 Invasive mucinous carcinoma
1.351
0.883–2.066
0.165
Overall survival
 Lepidic
1.730
0.878–3.413
0.113
 Acinar
1.122
0.754–1.669
0.571
 Papillary
1.284
0.861–1.912
0.220
 Micropapillary
0.585
0.385–0.890
0.012
 Solid
0.690
0.484–0.982
0.039
 Invasive mucinous carcinoma
0.870
0.519–1.456
0.594
Multivariate Cox model was adjusted for sex, age, smoking status, lymphovascular invasion or no lymphovascular invasion, tumor location, and nodal status (N0, N1, or N2). A two-tailed p < 0.05 was regarded as statistically different
OR odds ratio, CI confidence interval

Discussion

Lymph node metastasis is a major way for cancer cells to migrate from the primary tumor to distant organs, which promises a poor prognosis for lung cancer patients. With the classification of subtyping lung adenocarcinoma by the IASLC/ATS/ERS,6 several studies have reported that micropapillary- and solid-predominant subtypes were related to poor prognoses;9,1214,1921 however, lung adenocarcinomas usually contain a mixture of different subtypes. Thus, it is necessary for clinicians and researchers to understand whether minor components influence patients’ prognoses to help predict their prognoses and make therapeutic strategies. Yeh et al. pointed out that the presence of micropapillary or solid patterns were of predictive value with increased risk of occult lymph node metastasis;22 however, studies focusing on minor components are limited. In this study, our aim was to determine whether there was a correlation between minor components of lung adenocarcinoma and lymph node metastasis. According to our study of 1244 patients with pathologically proven lung adenocarcinoma that was initially diagnosed between August 2006 and May 2013, we found that patients with lung adenocarcinoma of lepidic (p < 0.001) subtype tended to have a lower rate of lymph node metastasis, while patients with lung adenocarcinoma of papillary, micropapillary and solid subtypes were more likely to have lymph node metastasis (Table 1). These results are consistent with several previous studies1921,23,24 but none reported the results of patients with lung adenocarcinoma of the papillary subtype. In addition, our data showed that patients with lung adenocarcinoma predominated by different subtypes, as well as patients presenting with different minor components, experience different possibilities of presenting with lymph node metastasis.
To further understand the relationship between different subtypes and lymph node metastasis, we recorded the number of metastatic lymph node stations/lymph nodes and resected lymph node stations/lymph nodes for each predominant subtype and minor component, and calculated the metastatic rate. According to our study, tumors with lepidic components had a lower rate of metastatic lymph node stations and lymph nodes, while tumors with micropapillary and solid components had a higher metastatic rate of lymph node stations and lymph nodes. Similar conclusions can also be made for patients with lepidic-, micropapillary- and solid-predominant and second predominant tumors (Tables 2, 3; electronic supplementary Tables 2, 3), with only one exception—the correlation between micropapillary second predominant subtype and metastatic rate of lymph node station, probably due to the sample size. That is to say, tumors containing minor components of micropapillary and solid subtypes are aggressive enough and have the potential for lymph node metastasis.
In univariate analysis, patients presenting with micropapillary and solid subtypes had shorter RFS and OS (Fig. 1). Interestingly, whether tumors contained a minor or predominant component of micropapillary and solid subtypes was not significantly different in RFS (p = 0.973 and p = 0.635, respectively, data not shown) or OS (p = 0.692 and p = 0.291, respectively, data not shown). This finding suggests that minor components of micropapillary and solid subtypes are as valuable as micropapillary- and solid-predominant subtypes in predicting patients’ prognosis. In multivariate analysis, minor components of micropapillary and solid subtypes were both independent predictive factors of a poor prognosis (Table 4). These results support the fact that micropapillary and solid subtypes are poor indicators for patients’ prognosis, even if they are minor components of a specific tumor.

Conclusions

Minor components, as well as predominant subtypes of micropapillary and solid subtypes of lung adenocarcinoma, are independent poor indicators of lymph node metastasis and prognosis. Our study concentrates not only on the predominant subtype but also on minor components of lung adenocarcinoma. It is believed that more data are needed to better clarify this issue, and we hope that minor components of lung adenocarcinoma are taken into consideration by clinicians when predicting patients’ prognosis and designing comprehensive therapeutic strategies in order to benefit more patients.

Acknowledgments

This study was approved by the Committee for Ethical Review of Research (Fudan University Shanghai Cancer Center IRB# 090977-1). It identifies that not only micropapillary- and solid-predominant subtypes but also minor micropapillary and solid components of lung adenocarcinoma are poor predictors for lymph node metastasis and poor prognosis.

Disclosure

Yue Zhao, Rui Wang, Xuxia Shen, Yunjian Pan, Chao Cheng, Yuan Li, Lei Shen, Yang Zhang, Hang Li, Difan Zheng, Ting Ye, Shanbo Zheng, Yihua Sun, and Haiquan Chen declare no potential conflicts of interest exist with any companies/organizations whose products or services may be discussed in this article.

Funding

This work was funded by the National Natural Science Foundation of China (81330056, 81401886, 81401891, 81422029, 81472173, and 81372525) and the Key Project of Science and Technology Commission of Shanghai Municipality (JGGG1302).
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

Die Chirurgie

Print-Titel

Das Abo mit mehr Tiefe

Mit der Zeitschrift Die Chirurgie erhalten Sie zusätzlich Online-Zugriff auf weitere 43 chirurgische Fachzeitschriften, CME-Fortbildungen, Webinare, Vorbereitungskursen zur Facharztprüfung und die digitale Enzyklopädie e.Medpedia.

Bis 30. April 2024 bestellen und im ersten Jahr nur 199 € zahlen!

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Anhänge

Electronic supplementary material

Literatur
1.
Zurück zum Zitat Scagliotti GV, Park K, Patil S, et al. Survival without toxicity for cisplatin plus pemetrexed versus cisplatin plus gemcitabine in chemonaive patients with advanced non-small cell lung cancer: a risk-benefit analysis of a large phase III study. Eur J Cancer. 2009;45:2298–303.CrossRefPubMed Scagliotti GV, Park K, Patil S, et al. Survival without toxicity for cisplatin plus pemetrexed versus cisplatin plus gemcitabine in chemonaive patients with advanced non-small cell lung cancer: a risk-benefit analysis of a large phase III study. Eur J Cancer. 2009;45:2298–303.CrossRefPubMed
2.
Zurück zum Zitat Scagliotti GV, Parikh P, von Pawel J, et al. Phase III study comparing cisplatin plus gemcitabine with cisplatin plus pemetrexed in chemotherapy-naive patients with advanced-stage non-small-cell lung cancer. J Clin Oncol. 2008;26:3543–51.CrossRefPubMed Scagliotti GV, Parikh P, von Pawel J, et al. Phase III study comparing cisplatin plus gemcitabine with cisplatin plus pemetrexed in chemotherapy-naive patients with advanced-stage non-small-cell lung cancer. J Clin Oncol. 2008;26:3543–51.CrossRefPubMed
3.
Zurück zum Zitat Ciuleanu T, Brodowicz T, Zielinski C, et al. Maintenance pemetrexed plus best supportive care versus placebo plus best supportive care for non-small-cell lung cancer: a randomised, double-blind, phase 3 study. Lancet. 2009;374:1432–40.CrossRefPubMed Ciuleanu T, Brodowicz T, Zielinski C, et al. Maintenance pemetrexed plus best supportive care versus placebo plus best supportive care for non-small-cell lung cancer: a randomised, double-blind, phase 3 study. Lancet. 2009;374:1432–40.CrossRefPubMed
4.
Zurück zum Zitat Scagliotti G, Hanna N, Fossella F, et al. The differential efficacy of pemetrexed according to NSCLC histology: a review of two phase III studies. Oncologist. 2009;14(3):253–63.CrossRefPubMed Scagliotti G, Hanna N, Fossella F, et al. The differential efficacy of pemetrexed according to NSCLC histology: a review of two phase III studies. Oncologist. 2009;14(3):253–63.CrossRefPubMed
5.
Zurück zum Zitat Johnson DH, Fehrenbacher L, Novotny WF, et al. Randomized phase II trial comparing bevacizumab plus carboplatin and paclitaxel with carboplatin and paclitaxel alone in previously untreated locally advanced or metastatic non-small-cell lung cancer. J Clin Oncol. 2004;22:2184–91.CrossRefPubMed Johnson DH, Fehrenbacher L, Novotny WF, et al. Randomized phase II trial comparing bevacizumab plus carboplatin and paclitaxel with carboplatin and paclitaxel alone in previously untreated locally advanced or metastatic non-small-cell lung cancer. J Clin Oncol. 2004;22:2184–91.CrossRefPubMed
6.
Zurück zum Zitat Travis WD, Brambilla E, Noguchi M, 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(2):244–85.CrossRefPubMedPubMedCentral Travis WD, Brambilla E, Noguchi M, 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(2):244–85.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Hung JJ, Yeh YC, Jeng WJ, et al. Predictive value of the International Association for the Study of Lung Cancer/American Thoracic Society/European Respiratory Society classification of lung adenocarcinoma in tumor recurrence and patient survival. J Clin Oncol. 2014;32:2357–64.CrossRefPubMed Hung JJ, Yeh YC, Jeng WJ, et al. Predictive value of the International Association for the Study of Lung Cancer/American Thoracic Society/European Respiratory Society classification of lung adenocarcinoma in tumor recurrence and patient survival. J Clin Oncol. 2014;32:2357–64.CrossRefPubMed
8.
Zurück zum Zitat Campos-Parra AD, Aviles A, Contreras-Reyes S, et al. Relevance of the novel IASLC/ATS/ERS classification of lung adenocarcinoma in advanced disease. Eur Respir J. 2014;43:1439–47.CrossRefPubMed Campos-Parra AD, Aviles A, Contreras-Reyes S, et al. Relevance of the novel IASLC/ATS/ERS classification of lung adenocarcinoma in advanced disease. Eur Respir J. 2014;43:1439–47.CrossRefPubMed
9.
Zurück zum Zitat Russell PA, Barnett SA, Walkiewicz M, et al. Correlation of mutation status and survival with predominant histologic subtype according to the new IASLC/ATS/ERS lung adenocarcinoma classification in stage III (N2) patients. J Thorac Oncol. 2013;8:461–8.CrossRefPubMed Russell PA, Barnett SA, Walkiewicz M, et al. Correlation of mutation status and survival with predominant histologic subtype according to the new IASLC/ATS/ERS lung adenocarcinoma classification in stage III (N2) patients. J Thorac Oncol. 2013;8:461–8.CrossRefPubMed
10.
Zurück zum Zitat Warth A, Muley T, Meister M, et al. The novel histologic International Association for the Study of Lung Cancer/American Thoracic Society/European Respiratory Society classification system of lung adenocarcinoma is a stage-independent predictor of survival. J Clin Oncol. 2012;30:1438–46.CrossRefPubMed Warth A, Muley T, Meister M, et al. The novel histologic International Association for the Study of Lung Cancer/American Thoracic Society/European Respiratory Society classification system of lung adenocarcinoma is a stage-independent predictor of survival. J Clin Oncol. 2012;30:1438–46.CrossRefPubMed
11.
Zurück zum Zitat Yoshizawa A, Motoi N, Riely GJ, et al. 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;24:653–64.CrossRefPubMed Yoshizawa A, Motoi N, Riely GJ, et al. 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;24:653–64.CrossRefPubMed
12.
Zurück zum Zitat Zhang Y, Wang R, Cai D, et al. A comprehensive investigation of molecular features and prognosis of lung adenocarcinoma with micropapillary component. J Thorac Oncol. 2014;9:1772–8.CrossRefPubMed Zhang Y, Wang R, Cai D, et al. A comprehensive investigation of molecular features and prognosis of lung adenocarcinoma with micropapillary component. J Thorac Oncol. 2014;9:1772–8.CrossRefPubMed
13.
Zurück zum Zitat Ujiie H, Kadota K, Chaft JE, et al. Solid predominant histologic subtype in resected stage I lung adenocarcinoma is an independent predictor of early, extrathoracic, multisite recurrence and of poor postrecurrence survival. J Clin Oncol. 2015;33(26):2877–84.CrossRefPubMed Ujiie H, Kadota K, Chaft JE, et al. Solid predominant histologic subtype in resected stage I lung adenocarcinoma is an independent predictor of early, extrathoracic, multisite recurrence and of poor postrecurrence survival. J Clin Oncol. 2015;33(26):2877–84.CrossRefPubMed
14.
Zurück zum Zitat Xu S, Xi J, Jiang W, et al. Solid component and tumor size correlate with prognosis of stage IB lung adenocarcinoma. Ann Thorac Surg. 2015;99:961–7.CrossRefPubMed Xu S, Xi J, Jiang W, et al. Solid component and tumor size correlate with prognosis of stage IB lung adenocarcinoma. Ann Thorac Surg. 2015;99:961–7.CrossRefPubMed
15.
Zurück zum Zitat Cha MJ, Lee HY, Lee KS, et al. Micropapillary and solid subtypes of invasive lung adenocarcinoma: clinical predictors of histopathology and outcome. J Thorac Cardiovasc Surg. 2014;147:921–8.CrossRefPubMed Cha MJ, Lee HY, Lee KS, et al. Micropapillary and solid subtypes of invasive lung adenocarcinoma: clinical predictors of histopathology and outcome. J Thorac Cardiovasc Surg. 2014;147:921–8.CrossRefPubMed
16.
Zurück zum Zitat Yang X, Liu Y, Lian F, et al. Lepidic and micropapillary growth pattern and expression of Napsin A can stratify patients of stage I lung adenocarcinoma into different prognostic subgroup. Int J Clin Exp Pathol. 2014;7:1459–68.PubMedPubMedCentral Yang X, Liu Y, Lian F, et al. Lepidic and micropapillary growth pattern and expression of Napsin A can stratify patients of stage I lung adenocarcinoma into different prognostic subgroup. Int J Clin Exp Pathol. 2014;7:1459–68.PubMedPubMedCentral
17.
Zurück zum Zitat Travis WD, Brambilla E, Nicholson AG, et al. The 2015 World Health Organization classification of lung tumors: impact of genetic, clinical and radiologic advances since the 2004 classification. J Thorac Oncol. 2015;10:1243–60.CrossRefPubMed Travis WD, Brambilla E, Nicholson AG, et al. The 2015 World Health Organization classification of lung tumors: impact of genetic, clinical and radiologic advances since the 2004 classification. J Thorac Oncol. 2015;10:1243–60.CrossRefPubMed
18.
Zurück zum Zitat Mountain CF, Dresler CM. Regional lymph node classification for lung cancer staging. Chest. 1997;111:1718–23.CrossRefPubMed Mountain CF, Dresler CM. Regional lymph node classification for lung cancer staging. Chest. 1997;111:1718–23.CrossRefPubMed
19.
20.
Zurück zum Zitat Lee G, Lee HY, Jeong JY, et al. Clinical impact of minimal micropapillary pattern in invasive lung adenocarcinoma: prognostic significance and survival outcomes. Am J Surg Pathol. 2015;39:660–6.CrossRefPubMed Lee G, Lee HY, Jeong JY, et al. Clinical impact of minimal micropapillary pattern in invasive lung adenocarcinoma: prognostic significance and survival outcomes. Am J Surg Pathol. 2015;39:660–6.CrossRefPubMed
21.
Zurück zum Zitat Nitadori J, Bograd AJ, Kadota K, et al. Impact of micropapillary histologic subtype in selecting limited resection vs lobectomy for lung adenocarcinoma of 2 cm or smaller. J Natl Cancer Inst. 2013;105:1212–20.CrossRefPubMedPubMedCentral Nitadori J, Bograd AJ, Kadota K, et al. Impact of micropapillary histologic subtype in selecting limited resection vs lobectomy for lung adenocarcinoma of 2 cm or smaller. J Natl Cancer Inst. 2013;105:1212–20.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Yeh YC, Kadota K, Nitadori JI, et al. International Association for the Study of Lung Cancer/American Thoracic Society/European Respiratory Society classification predicts occult lymph node metastasis in clinically mediastinal node-negative lung adenocarcinoma. Eur J Cardiothorac Surg. 2016;49(1):e9–15.CrossRefPubMed Yeh YC, Kadota K, Nitadori JI, et al. International Association for the Study of Lung Cancer/American Thoracic Society/European Respiratory Society classification predicts occult lymph node metastasis in clinically mediastinal node-negative lung adenocarcinoma. Eur J Cardiothorac Surg. 2016;49(1):e9–15.CrossRefPubMed
23.
Zurück zum Zitat Araki K, Kidokoro Y, Hosoya K, et al. Excellent prognosis of lepidic-predominant lung adenocarcinoma: low incidence of lymphatic vessel invasion as a key factor. Anticancer Res. 2014;34:3153–6.PubMed Araki K, Kidokoro Y, Hosoya K, et al. Excellent prognosis of lepidic-predominant lung adenocarcinoma: low incidence of lymphatic vessel invasion as a key factor. Anticancer Res. 2014;34:3153–6.PubMed
24.
Zurück zum Zitat Tsubokawa N, Mimae T, Sasada S, et al. Negative prognostic influence of micropapillary pattern in stage IA lung adenocarcinoma. Eur J Cardiothorac Surg. 2016;49(1):293–9.CrossRefPubMed Tsubokawa N, Mimae T, Sasada S, et al. Negative prognostic influence of micropapillary pattern in stage IA lung adenocarcinoma. Eur J Cardiothorac Surg. 2016;49(1):293–9.CrossRefPubMed
Metadaten
Titel
Minor Components of Micropapillary and Solid Subtypes in Lung Adenocarcinoma are Predictors of Lymph Node Metastasis and Poor Prognosis
verfasst von
Yue Zhao, MD
Rui Wang, MD
Xuxia Shen, MD
Yunjian Pan, MD
Chao Cheng, MD
Yuan Li, MD
Lei Shen, MD
Yang Zhang, MD
Hang Li, MD
Difan Zheng
Ting Ye, MD
Shanbo Zheng, MD
Yihua Sun, MD
Haiquan Chen, MD
Publikationsdatum
02.02.2016
Verlag
Springer International Publishing
Erschienen in
Annals of Surgical Oncology / Ausgabe 6/2016
Print ISSN: 1068-9265
Elektronische ISSN: 1534-4681
DOI
https://doi.org/10.1245/s10434-015-5043-9

Weitere Artikel der Ausgabe 6/2016

Annals of Surgical Oncology 6/2016 Zur Ausgabe

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Wie sieht der OP der Zukunft aus?

04.05.2024 DCK 2024 Kongressbericht

Der OP in der Zukunft wird mit weniger Personal auskommen – nicht, weil die Technik das medizinische Fachpersonal verdrängt, sondern weil der Personalmangel es nötig macht.

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Recycling im OP – möglich, aber teuer

02.05.2024 DCK 2024 Kongressbericht

Auch wenn sich Krankenhäuser nachhaltig und grün geben – sie tragen aktuell erheblich zu den CO2-Emissionen bei und produzieren jede Menge Müll. Ein Pilotprojekt aus Bonn zeigt, dass viele Op.-Abfälle wiederverwertet werden können.

Update Chirurgie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.

S3-Leitlinie „Diagnostik und Therapie des Karpaltunnelsyndroms“

Karpaltunnelsyndrom BDC Leitlinien Webinare
CME: 2 Punkte

Das Karpaltunnelsyndrom ist die häufigste Kompressionsneuropathie peripherer Nerven. Obwohl die Anamnese mit dem nächtlichen Einschlafen der Hand (Brachialgia parästhetica nocturna) sehr typisch ist, ist eine klinisch-neurologische Untersuchung und Elektroneurografie in manchen Fällen auch eine Neurosonografie erforderlich. Im Anfangsstadium sind konservative Maßnahmen (Handgelenksschiene, Ergotherapie) empfehlenswert. Bei nicht Ansprechen der konservativen Therapie oder Auftreten von neurologischen Ausfällen ist eine Dekompression des N. medianus am Karpaltunnel indiziert.

Prof. Dr. med. Gregor Antoniadis
Berufsverband der Deutschen Chirurgie e.V.

S2e-Leitlinie „Distale Radiusfraktur“

Radiusfraktur BDC Leitlinien Webinare
CME: 2 Punkte

Das Webinar beschäftigt sich mit Fragen und Antworten zu Diagnostik und Klassifikation sowie Möglichkeiten des Ausschlusses von Zusatzverletzungen. Die Referenten erläutern, welche Frakturen konservativ behandelt werden können und wie. Das Webinar beantwortet die Frage nach aktuellen operativen Therapiekonzepten: Welcher Zugang, welches Osteosynthesematerial? Auf was muss bei der Nachbehandlung der distalen Radiusfraktur geachtet werden?

PD Dr. med. Oliver Pieske
Dr. med. Benjamin Meyknecht
Berufsverband der Deutschen Chirurgie e.V.

S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“

Appendizitis BDC Leitlinien Webinare
CME: 2 Punkte

Inhalte des Webinars zur S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“ sind die Darstellung des Projektes und des Erstellungswegs zur S1-Leitlinie, die Erläuterung der klinischen Relevanz der Klassifikation EAES 2015, die wissenschaftliche Begründung der wichtigsten Empfehlungen und die Darstellung stadiengerechter Therapieoptionen.

Dr. med. Mihailo Andric
Berufsverband der Deutschen Chirurgie e.V.