Skip to main content
Erschienen in: Annals of Surgical Oncology 13/2019

Open Access 11.10.2019 | Colorectal Cancer

The Relationship Between Tumor Budding, Tumor Microenvironment, and Survival in Patients with Primary Operable Colorectal Cancer

verfasst von: Hester C. van Wyk, PhD, Antonia Roseweir, PhD, Peter Alexander, MBChB, James H. Park, PhD, Paul G. Horgan, PhD, Donald C. McMillan, PhD, Joanne Edwards, PhD

Erschienen in: Annals of Surgical Oncology | Ausgabe 13/2019

Abstract

Background

Tumor budding is an independent prognostic factor in colorectal cancer (CRC) and has recently been well-defined by the International Tumour Budding Consensus Conference (ITBCC).

Objective

The aim of the present study was to use the ITBCC budding evaluation method to examine the relationship between tumor budding, tumor factors, tumor microenvironment, and survival in patients with primary operable CRC.

Methods

Hematoxylin and eosin-stained slides of 952 CRC patients diagnosed between 1997 and 2007 were evaluated for tumor budding according to the ITBCC criteria. The tumor microenvironment was evaluated using tumor stroma percentage (TSP) and Klintrup–Makinen (KM) grade to assess the tumor inflammatory cell infiltrate.

Results

High budding (n = 268, 28%) was significantly associated with TNM stage (p < 0.001), competent mismatch repair (MMR; p < 0.05), venous invasion (p < 0.001), weak KM grade (p < 0.001), high TSP (p < 0.001), and reduced cancer-specific survival (CSS) (hazard ratio 8.68, 95% confidence interval 6.30–11.97; p < 0.001). Tumor budding effectively stratifies CSS stage T1 through to T4 (all p < 0.05) independent of associated factors.

Conclusions

Tumor budding effectively stratifies patients’ survival in primary operable CRC independent of other phenotypic features. In particular, the combination of T stage and budding should form the basis of a new staging system for primary operable CRC.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Tumor budding has been defined as a single tumor cell or small cluster of four or fewer tumor cells at the invasive front12,18 and should be considered a promising and strong prognostic factor in colorectal cancer (CRC).19 Widespread reporting of tumor budding has not progressed to routine clinical practice due to a lack of consensus on scoring methods. However, routine reporting is now advocated by using the approach outlined by the International Tumour Budding Consensus Conference (ITBCC), with recommendations for the assessment and reporting of tumor budding in CRC.6
The ITBCC recommends that tumor budding should be included in future CRC guidelines and protocols and should be considered, along with other clinicopathological factors, in a multidisciplinary setting. The recent dataset for histopathological reporting of CRC by the royal pathologist stated that they would reconsider reporting tumor budding when new data become available.4
The tumor microenvironment also plays an important role in CRC outcomes. Marked peritumoral inflammation has been associated with favorable outcome,3,14 while the presence of a high tumor stroma percentage (TSP) has been validated as a stage-independent marker of reduced survival in patients with primary operable CRC.7,8. Both contribute to the development of a tumor microenvironment score that can potentially supplement the current TNM staging system.9
The aim of this study was to assess the proposed method by ITBCC in clinical practice and investigate the relationship between tumor budding and tumor factors, tumor microenvironment, and survival in primary operable CRC.

Patients and Methods

Patients

Patients were identified from a prospectively collected database of patients undergoing surgery for CRC between 1997 and 2007 at the Royal Infirmary, Western Infirmary and Stobhill Hospital, Glasgow. Any patient with a synchronous cancer, inflammatory bowel disease, and mortality within 30 days of surgery, who had received neoadjuvant therapy, was excluded. Furthermore, patients who had their disease managed entirely endoscopically, without formal colonic or rectal resection, were also excluded from the study.
Patients were staged according to the TNM criteria that was applicable at the time of surgery and to current practice in the UK.4 The West of Scotland Research Ethics Committee granted study approval. Patients were followed up for at least 5 years, and the date and cause of death were cross-checked with electronic case records. Cancer-specific survival (CSS) was measured from the date of surgery until the date of death from CRC.

Routine Stains: Hematoxylin and Eosin and Elastica

For evaluation, two consecutive cut samples from each specimen were stained; one was routinely stained with hematoxylin and eosin (H&E), and the other was stained with Miller’s elastic stain (BDH, Poole, Dorset, UK) and counterstained with H&E. Routine pathological elastica staining was used to assess the presence of venous invasion.13. Only venous invasion was evaluated and no special staining was utilized to improve the assessment of lymphatic or perineural invasion.

Assessment of Tumor Budding

Tumor budding has been assessed on scanned H&E-stained slides in a single hotspot as defined by ITBCC. Tumor budding was counted on H&E slides, and was assessed in one hotspot (in a field measuring 0.785 mm2) at the invasive front. A two-tier system was used along with the budding count to facilitate risk stratification in CRC. Cut-offs used were: low, 0–9 buds; and high, ≥ 10 buds (Fig. 1a).
Tumor budding was evaluated by two different observers to ensure reliability, and co-scoring of randomly selected cases was carried out by HvW and PA. The interobserver intraclass correlation coefficient (ICCC) for the scores was 0.794 (p < 0.001).

Evaluation of Tumor Inflammatory Cell Infiltrate and Tumor Stroma Percentage

Klintrup–Makinen grade was used to assess the generalized inflammatory infiltrate semi-quantitatively. Full H&E sections of the deepest point of tumor invasion were used, whereby inflammatory cell density at the invasive margin was graded using a 4-point scale; it was classified as low-grade (no increase or mild/patchy increase in inflammatory cells) or high-grade (prominent inflammatory reaction forming a band at the invasive margin, or florid cup-like infiltrate at the invasive edge with destruction of cancer cell islands), as previously described3,14 (Fig. 1b).
Tumor stroma percentage was assessed semi-quantitatively using full sections of the deepest point of tumor invasion; the proportion of stroma was calculated as a percentage of the visible field, excluding areas of mucin deposition or necrosis. Tumors were subsequently graded as low (50%) or high (> 50%) TSP as previously described8 (Fig. 1c).

Assessment of Ki-67, Mismatch Repair (MMR) and BRAF Status

The Ki-67 proliferation index was assessed cohort using a threshold of 50%. A subset of patients in the full cohort underwent evaluation of the mismatch repair (MMR) and BRAF statuses. Using immunohistochemistry, a previously constructed tissue microarray comprising cores of formalin-fixed paraffin-embedded cancer tissue was used to assess MMR and BRAF status. Immunohistochemistry for MMR status has been previously described.10 MMR protein expression was reported as MMR-competent or -deficient, by a single blinded observer. The assessment of BRAF status was performed as previously described by our group11 and therefore the results were incorporated in our study. BRAF V600E mutation was reported as absent or present by a single blinded observer.

Statistical Analysis

Interrelationships between tumor location, clinicopathological characteristics, and measures of the tumor microenvironment were analyzed using the Chi square test. Five-year CSS and overall survial was examined using Kaplan–Meier log-rank survival analysis and univariate Cox proportional hazards regression to calculate hazard ratios and 95% confidence intervals. Variables found to be statistically significant (p < 0.05) on univariate analysis were entered into a Cox regression multivariate model using a backward conditional method. A p value < 0.05 was significant. Analyses were performed using SPSS software version 24 (IBM SPSS, Armonk, NY, USA).

Results

The cohort consisted of 952 patients. Clinical and pathological features are shown in Table 1. Two-thirds of patients included were over the age of 65 years, and 52% were male. Overall, 131 (14%) patients had TNM stage I disease, 445 (47%) had stage II disease, 355 (37%) had stage III disease, and 21 (2%) had stage IV disease; 713 (75%) patients had colon tumors and 239 (25%) had rectal cancer. Venous invasion was present in 321 (34%) tumors, and tumor necrosis was present in 362 (38%) tumors. High-grade tumor budding was present in 28% of tumors, a low-grade inflammatory cell infiltrate was present in 69% of tumors, and 25% of tumors had a high TSP. MMR deficiency was identified in 17% of patients, and 20% of patients had BRAF V600E mutations. The median follow-up for patients was 11.7 years (range 6.4–16.3), with 226 cancer deaths and 271 non-cancer deaths. On univariate survival analysis, T stage (p < 0.001), N stage (p < 0.001), TNM stage (p < 0.001), Ki-67 proliferation index (p < 0.001), MMR (p ≤ 0.001), venous invasion (p < 0.001), Klintrup–Makinen (KM) grade (p < 0.001), TSP (p < 0.001), and tumor budding (p < 0.001) (Fig. 2) were significantly associated with CSS.
Table 1
Clinicopathological features of patients with primary operable colorectal cancer and cancer-specific survival
 
N (%)
[952 patients]
Univariate analysis
[CSS]
p value
Host characteristics
Age (years)
1.00 (0.76–1.32)
0.984
 < 65
297 (31)
 > 65
655 (69)
Sex
1.20 (0.92–1.56)
0.176
 Female
456 (48)
 Male
496 (52)
Adjuvant therapy
1.03 (0.68–1.28)
0.859
 Yes
105 (34)
 No
204 (64)
Tumor characteristics
Tumor site
1.18 (0.89–1.58)
0.247
 Colon
713 (75)
 Rectum
239 (25)
Tstage
1.97 (1.61–2.41)
< 0.001
 1
40 (4)
 2
117 (12)
 3
514 (54)
 4
281 (30)
N stage
2.09 (1.78–2.47)
< 0.001
 0
582 (61)
 1
253 (27)
 2
113 (12)
TNM stage
2.95 (2.38–3.66)
< 0.001
 1
131 (14)
 2
445 (47)
 3
355 (37)
 4
21 (2)
Ki-67 proliferation index
0.60 (0.46–0.79)
< 0.001
 Low
450 (48)
 High
491 (52)
Mismatch repair status
0.64 (0.50–0.82)
0.001
 Competent
763 (83)
 Deficient
162 (17)
BRAF status
0.91 (0.65–1.28)
0.589
 Low
735 (80)
 High
187 (20)
Tumor necrosis
1.08 (0.82–1.41)
0.579
 Low
581 (62)
 High
362 (38)
Venous invasion
2.62 (2.02–3.41)
< 0.001
 Absent
631 (66)
 Present
321 (34)
Klintrup–Makinen grade
0.33 (0.23–0.46)
<0.001
 Weak
651 (69)
 Strong
296 (31)
Tumor stroma percentage
2.07 (1.58–2.74)
< 0.001
 Low
704 (75)
 High
232 (25)
Tumor budding
10.41 (7.76–13.96)
< 0.001
 Low
684 (72)
 High
268 (28)
CSS cancer-specific survival
The relationship between tumor budding, clinicopathological features of primary operable CRC, and tumor budding are shown in Table 2. High-grade budding was associated with T stage (p < 0.001), N stage (p < 0.001), TNM stage (p < 0.001), MMR status (p < 0.05), venous invasion (p < 0.001), KM grade (p < 0.001), and TSP (p < 0.001).
Table 2
The relationship between tumor budding and clinicopathological features of patients with primary operable colorectal cancer
 
Tumor budding [N (%)]
p value
Low [684 (72%)]
High [268 (28%)]
Host characteristics
Age (years)
0.110
 < 65
205 (30)
92 (34)
 > 65
479 (70)
176 (66)
Sex
0.199
 Male
334 (49)
122 (46)
 Female
350 (51)
146 (54)
Tumor characteristics
 
Tumor site
0.451
 Colon
511 (75)
202 (75)
 Rectum
173 (25)
66 (25)
T stage
< 0.001
 I
38 (6)
2 (1)
 II
101 (15)
16 (6)
 III
393 (57)
121 (45)
 IV
152 (22)
129 (48)
N stage
< 0.001
 0
471 (69)
111 (42)
 1
154 (23)
99 (37)
 2
55 (8)
57 (21)
TNM stage
< 0.001
 1
120 (17)
11 (4)
 2
349 (51)
96 (36)
 3
209 (31)
146 (55)
 4
6 (1)
15 (5)
Proliferation index
0.285
 Low
318 (47)
131 (49)
 High
357 (53)
134 (51)
Mismatch repair status
0.012
 Competent
539 (81)
224 (87)
 Deficient
129 (19)
33 (13)
BRAF status
0.066
 Low
519 (78)
216 (83)
 High
143 (22)
44 (17)
Necrosis
0.499
 < 25%
417 (62)
164 (61)
 > 25%
259 (38)
103 (39)
Venous invasion
< 0.001
 Absent
493 (72)
138 (52)
 Present
191 (28)
130 (48)
Klintrup–Makinen
< 0.001
 Weak
432 (64)
219 (82)
 strong
248 (36)
48 (18)
Tumor stroma percentage
< 0.001
 ≤ 50
542 (81)
162 (61)
 > 50
127 (19)
105 (39)
The relationship between T stage (1–4), clinicopathological characteristics, and CSS in patients with primary operable CRC are shown in Table 3 and Fig. 3a–d. T1 and T2 stages were associated with tumor budding (p < 0.01 and p < 0.001, respectively), and T3 stage was associated with venous invasion (p < 0.01), Ki-69 proliferation index (p < 0.01), KM grade (p < 0.05), and tumor budding (p < 0.001). T4 stage was associated with N stage (p < 0.05), venous invasion (p < 0.05), MMR status (p < 0.05), and tumor budding (p < 0.001).
Table 3
Relationship between T stage, clinicopathological characteristics, and cancer-specific survival in patients with primary operable colorectal cancer
 T stage
Univariate analysis
p value
Multivariate analysis
p value
T1 [n = 40]
N stage (0/1/2)
1.55 (0.16–14.92)
0.704
  
Ki67 proliferation Index (low/high)
1.66 (0.17–15.98)
0.660
  
Mismatch repair status (competent/deficient)
1.44 (0.14–13.85)
0.753
  
BRAF status (low/high)
0.04 (0–308.53)
0.569
  
Tumor necrosis (low/high)
0.31 (0.00–375.83)
0.468
  
Venous invasion (no/yes)
0.46 (0.0–17,931)
0.760
  
Klintrup–Makinen grade (weak/strong)
0.142 (0.015–1.37)
0.091
0.97 (0.008–1.19)
0.068
Tumor stroma percentage (low/high)
0.043 (0.00–17,062)
0.685
  
Tumor budding (low/high)
8.99 (0.92–88.13)
0.059
8.44 (1.12–303.95)
0.042
T2 [n = 117]
N stage (0/1/2)
1.92 (0.88–4.20)
0.104
  
Ki67 proliferation index (low/high)
0.557 (0.26–2.02)
0.547
  
Mismatch repair status (competent/deficient)
0.75 (0.17–3.33)
0.704
  
BRAF status (low/high)
1.21 (0.34–4.33)
0.78
  
Tumor necrosis (low/high)
1.41 (0.48–4.13)
0.530
  
Venous invasion (no/yes)
3.18 (1.12–9.02)
0.030
2.78 (0.96–8.11)
0.060
Klintrup–Makinen grade (weak/strong)
0.60 (0.22–1.66)
0.328
  
Tumor stroma percentage (low/high)
1.41 (0.48–4.13)
0.530
  
Tumor budding (low/high)
8.39 (3.03.03–23.20)
< 0.001
7.86 (2.81–21.95)
< 0.001
T3 [n = 514]
N stage (0/1/2)
1.74 (1.36–2.22)
< 0.001
1.20 (0.92–1.56)
0.176
Ki67 proliferation index (low/high)
0.98 (0.97–0.99)
0.002
0.57 (0.38–0.87)
0.008
Mismatch repair status (competent/deficient)
0.87 (0.50–1.50)
0.610
  
BRAF status (low/high)
0.96 (0.60–1.56)
0.882
  
Tumor necrosis (low/high)
1.04 (0.70–1.54)
0.848
  
Venous invasion (no/yes)
1.78 (1.21–2.62)
0.003
1.74 (1.16–2.60)
0.007
Klintrup–Makinen grade (weak/strong)
0.37 (0.21–0.65)
0.001
0.55 (0.30–0.99)
0.049
Tumor stroma percentage (low/high)
1.75 (1.15–2.66)
0.008
0.96 (0.61–1.53)
0.877
Tumor budding (low/high)
8.98 (4.03–9.39)
< 0.001
9.11 (5.92–14.03)
< 0.001
T4 [n = 281]
N stage (0/1/2)
2.07 (1.62–2.65)
< 0.001
1.34 (1.02–1.76)
0.032
Ki67 proliferation index (low/high)
0.78 (0.57–1.18)
0.246
  
Mismatch repair status (competent/deficient)
0.21 (0.84–0.51
0.001
0.32 (0.13–0.80)
0.015
BRAF status (low/high)
0.84 (0.49–1.41)
0.504
  
Tumor necrosis (low/high)
0.93 (0.63–1.39)
0.736
  
Venous invasion (no/yes)
2.99 (1.98–4.52)
< 0.001
1.89 (1.22–2.95)
0.005
Klintrup–Makinen grade (weak/strong)
0.35 (0.19–0.63)
<0.001
0.62 (0.33–1.14)
0.124
Tumor stroma percentage (low/high)
0.99 (0.98–1.01)
0.505
  
Tumor budding (low/high)
10.36 (5.96–18.01)
< 0.001
8.23 (4.59–14.73)
< 0.001
The relationship between TNM stage, clinicopathological characteristics, and CSS in patients with primary operable CRC are shown in Table 4. TNM stages I and II were associated with venous invasion (p < 0.05), KM grade (p < 0.05), and tumor budding (p < 0.001). TNM stage III was associated with venous invasion (p < 0.05) and tumor budding (p < 0.001), and TNM stage IV was associated with tumor budding (p < 0.05).
Table 4
Relationship between TNM stage, clinicopathological characteristics, and cancer-specific survival in patients with primary operable colorectal cancer
TNM stage
Univariate analysis
p value
Multivariate analysis
p value
Stage 1 [n = 131]
Ki67 proliferation index (low/high)
0.65 (0.22–1.95)
0.445
  
Mismatch repair status (competent/deficient)
0.39 (0.0–21.82)
0.313
  
BRAF status (low/high)
0.57 (0.07–4.38)
0.585
  
Tumor necrosis (low/high)
1.69 (0.57–5.19)
0.353
  
Venous invasion (absent/present)
3.44 (1.05–11.22)
0.041
3.53 (1.02–12.17)
0.046
Klintrup–Makinen grade (weak/strong)
0.28 (0.08–0.89)
0.032
0.27 (0.08–0.94)
0.040
Tumor stroma percentage (low/high)
1.79 (0.49–6.50)
0.378
  
Tumor budding (low/high)
9.96 (3.34–29.72)
< 0.001
7.90 (2.63–23.74)
< 0.001
Stage 2 [n = 445]
Ki67 proliferation index (low/high)
0.55 (0.34–0.89)
0.014
  
Mismatch repair status (competent/deficient)
0.58 (0.29–1.14)
0.113
  
BRAF status (low/high)
0.97 (0.55–1.73)
0.927
  
Tumor necrosis (low/high)
0.87 (0.53–1.40)
0.555
  
Venous invasion (absent/present)
2.34 (1.47–3.74)
0.001
2.05 (1.27–3.31)
0.003
Klintrup–Makinen grade (weak/strong)
0.41 (0.22–0.77)
0.005
0.51 (0.26–0.97)
0.040
Tumor stroma percentage (low/high)
3.07 (1.89–4.98)
0.001
1.53 (0.92–2.54)
0.102
Tumor budding (low/high)
10.21 (6.26–16.64)
< 0.001
10.23 (6.12–17.07)
< 0.001
Stage 3 [n = 355]
Ki67 proliferation index (low/high)
0.66 (0.46–0.95)
0.025
0.99 (0.99–1.00)
0.296
Mismatch repair status (competent/deficient)
0.83 (0.45–1.50)
0.529
  
BRAF status (low/high)
0.83 (0.52–1.34)
0.453
  
Tumor necrosis (low/high)
1.29 (0.90–1.85)
0.166
  
Venous invasion (absent/present)
1.81 (1.27–2.59)
0.001
1.55 (1.08–2.23)
0.017
Klintrup–Makinen grade (weak/strong)
0.46 (0.28–0.75)
0.0502
0.62 (0.38–1.01)
0.054
Tumor stroma percentage (low/high)
1.27 (0.87–1.85)
0.218
  
Tumor budding (low/high)
7.46 (4.87–11.42)
< 0.001
9.11 (5.92–14.03)
< 0.001
Stage 4 [n = 21]
Ki67 proliferation index (low/high)
0.58 (0.21–1.61)
0.294
  
Mismatch repair status (competent/deficient)
4.27 (0.48–38.24)
0.195
  
BRAF status (low/high)
0.72 (0.23–2.21)
0.561
  
Tumor necrosis (low/high)
0.63 (0.18–2.23)
0.478
  
venous invasion (absent/present)
1.28 (0.47–3.47)
0.628
  
Klintrup–Makinen grade (weak/strong)
0.31 (0.0–13.67)
0.264
  
Tumor stroma percentage (low/high)
1.48 (0.55–3.99)
0.444
  
Tumor budding (low/high)
10.82 (1.38–85.02)
0.024
10.82 (1.38–85.02)
0.024
Tumor budding is an independent prognostic factor in all stages with venous invasion, independent of stages I–III, and KM grade, independent of stages I–II.

Discussion

The results of the present study show that tumor budding effectively stratifies CSS in patients with primary operable cancer. Furthermore, compared with other tumor characteristics, including T and nodal stage, tumor budding had the highest hazard ratio, approximately three times that of any other tumor characteristic. Therefore, tumor budding is a good candidate to form the basis of a new staging system for primary operable CRC.
It was also of interest that in T1 and T2 tumors, only tumor budding had significant prognostic value. This would indicate that tumor budding occurs early in tumor invasion and may represent aggressive characteristics of malignant tumors, such as loss of cell adhesion and local invasion.5,20 Indeed, it may be argued that given its association with other characteristics of the tumor and its microenvironment, tumor budding is the most important component of tumor invasion. Indeed, the prognostic importance of tumor budding in local excision specimens in predicting outcome and/or predictive of nodal metastatic disease in stage pT1 CRCs is increasingly being recognized.1,16,17,20
The Ki-67 antigen is widely used to evaluate tumor proliferative activity as autonomous cell proliferation is a main feature of neoplasia. However, despite the clear association of tumor budding with migration and invasion, paradoxically, tumor buds appear not to be associated with proliferation. However, the association between tumor budding intensity and proliferative activity is still poorly understood and it is speculated that host invasion, by budding tumor cells, might be activated only after the cell cycle has been switched off.2,15 Further work is required to elucidate the molecular basis of this relationship.
In the present study, it was of interest that other components of the tumor microenvironment had increasing prognostic value in patients with T3 and T4 disease. The basis of this observation is not clear, however it may be that in larger, more invasive tumors, the components of the tumor microenvironment, such as the tumor stroma and inflammatory infiltrate, become more important in determining the future of the primary tumor. For example, the good outcome associated with a pronounced tumor inflammatory cell infiltrate may be due to the effective elimination of the tumor. In contrast, the poor outcome associated with the pronounced tumor stroma may be due to the supportive environment for the tumor. Indeed, Ueno et al. reported an association between fibrotic immature stroma and the intensity of tumor budding as histological dedifferentiation, including dissociation of cancer cells and the first step of invasion. Therefore, the present findings further support a pertinent role of tumor stroma in facilitating tumor cell de-differentiation and dissemination. Further work is required to elucidate the molecular basis of this relationship.

Conclusions

The findings of the present study indicate that tumor budding effectively stratifies cancer survival in patients with primary operable CRC. This stratification is independent of recognized tumor factors, including TNM stage. Therefore, the ITBCC budding evaluation method should be used to assess tumor budding and may form the basis of a new staging system in patients with CRC.
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.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

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.

Literatur
1.
Zurück zum Zitat Brown IS, Bettington ML, Bettington A, Miller G, Rosty C. Adverse histological features in malignant colorectal polyps: a contemporary series of 239 cases. J Clin Pathol. 2016;69:292–9.CrossRef Brown IS, Bettington ML, Bettington A, Miller G, Rosty C. Adverse histological features in malignant colorectal polyps: a contemporary series of 239 cases. J Clin Pathol. 2016;69:292–9.CrossRef
2.
Zurück zum Zitat Dawson H, Koelzer VH, Karamitopoulou E, et al. The apoptotic and proliferation rate of tumour budding cells in colorectal cancer outlines a heterogeneous population of cells with various impacts on clinical outcome. Histopathology. 2014;64:577–84.CrossRef Dawson H, Koelzer VH, Karamitopoulou E, et al. The apoptotic and proliferation rate of tumour budding cells in colorectal cancer outlines a heterogeneous population of cells with various impacts on clinical outcome. Histopathology. 2014;64:577–84.CrossRef
3.
Zurück zum Zitat Klintrup K, Mäkinen JM, Kauppila S, et al. Inflammation and prognosis in colorectal cancer. Eur. J. Cancer. 2005;41:2645–54.CrossRef Klintrup K, Mäkinen JM, Kauppila S, et al. Inflammation and prognosis in colorectal cancer. Eur. J. Cancer. 2005;41:2645–54.CrossRef
5.
Zurück zum Zitat Lugli A, Karamitopoulou E, Panayiotides I, et al. CD8+ lymphocytes/tumour-budding index: an independent prognostic factor representing a ‘pro-/anti-tumour ‘approach to tumour host interaction in colorectal cancer. Br J Cancer. 2009;101:1382.CrossRef Lugli A, Karamitopoulou E, Panayiotides I, et al. CD8+ lymphocytes/tumour-budding index: an independent prognostic factor representing a ‘pro-/anti-tumour ‘approach to tumour host interaction in colorectal cancer. Br J Cancer. 2009;101:1382.CrossRef
6.
Zurück zum Zitat Lugli A, Kirsch R, Ajioka Y, et al. Recommendations for reporting tumor budding in colorectal cancer based on the International Tumor Budding Consensus Conference (ITBCC) 2016. Mod Pathol. 2017;30:1299.CrossRef Lugli A, Kirsch R, Ajioka Y, et al. Recommendations for reporting tumor budding in colorectal cancer based on the International Tumor Budding Consensus Conference (ITBCC) 2016. Mod Pathol. 2017;30:1299.CrossRef
7.
Zurück zum Zitat Mesker WE, Junggeburt J, Szuhai K, et al. The carcinoma–stromal ratio of colon carcinoma is an independent factor for survival compared to lymph node status and tumor stage. Anal. Cell. Pathol. 2007;29:387–98. Mesker WE, Junggeburt J, Szuhai K, et al. The carcinoma–stromal ratio of colon carcinoma is an independent factor for survival compared to lymph node status and tumor stage. Anal. Cell. Pathol. 2007;29:387–98.
8.
Zurück zum Zitat Park J, Richards C, McMillan D, Horgan P, Roxburgh C. The relationship between tumour stroma percentage, the tumour microenvironment and survival in patients with primary operable colorectal cancer. Ann. Oncol. 2014;25:644–51.CrossRef Park J, Richards C, McMillan D, Horgan P, Roxburgh C. The relationship between tumour stroma percentage, the tumour microenvironment and survival in patients with primary operable colorectal cancer. Ann. Oncol. 2014;25:644–51.CrossRef
9.
Zurück zum Zitat Park JH, McMillan DC, Powell AG, et al. Evaluation of a tumor microenvironment-based prognostic score in primary operable colorectal cancer. Clin. Cancer Res. 2015;21:882–8.CrossRef Park JH, McMillan DC, Powell AG, et al. Evaluation of a tumor microenvironment-based prognostic score in primary operable colorectal cancer. Clin. Cancer Res. 2015;21:882–8.CrossRef
10.
Zurück zum Zitat Park JH, Powell AG, Roxburgh CS, Horgan PG, McMillan DC, Edwards J. Mismatch repair status in patients with primary operable colorectal cancer: associations with the local and systemic tumour environment. Br J Cancer. 2016;114:562.CrossRef Park JH, Powell AG, Roxburgh CS, Horgan PG, McMillan DC, Edwards J. Mismatch repair status in patients with primary operable colorectal cancer: associations with the local and systemic tumour environment. Br J Cancer. 2016;114:562.CrossRef
11.
Zurück zum Zitat Patel M, McSorley ST, Park JH, et al. The relationship between right-sided tumour location, tumour microenvironment, systemic inflammation, adjuvant therapy and survival in patients undergoing surgery for colon and rectal cancer. Br J Cancer. 2018;118:705.CrossRef Patel M, McSorley ST, Park JH, et al. The relationship between right-sided tumour location, tumour microenvironment, systemic inflammation, adjuvant therapy and survival in patients undergoing surgery for colon and rectal cancer. Br J Cancer. 2018;118:705.CrossRef
12.
Zurück zum Zitat Prall F, Nizze H, Barten M. Tumour budding as prognostic factor in stage I/II colorectal carcinoma. Histopathology. 2005;47:17–24.CrossRef Prall F, Nizze H, Barten M. Tumour budding as prognostic factor in stage I/II colorectal carcinoma. Histopathology. 2005;47:17–24.CrossRef
13.
Zurück zum Zitat Roxburgh CS, McMillan DC, Anderson JH, McKee RF, Horgan PG, Foulis AK. Elastica staining for venous invasion results in superior prediction of cancer-specific survival in colorectal cancer. Ann. Surg. 2010;252:989–97.CrossRef Roxburgh CS, McMillan DC, Anderson JH, McKee RF, Horgan PG, Foulis AK. Elastica staining for venous invasion results in superior prediction of cancer-specific survival in colorectal cancer. Ann. Surg. 2010;252:989–97.CrossRef
14.
Zurück zum Zitat Roxburgh CS, Salmond JM, Horgan PG, Oien KA, McMillan DC. Tumour inflammatory infiltrate predicts survival following curative resection for node-negative colorectal cancer. Eur J Cancer. 2009;45:2138–45.CrossRef Roxburgh CS, Salmond JM, Horgan PG, Oien KA, McMillan DC. Tumour inflammatory infiltrate predicts survival following curative resection for node-negative colorectal cancer. Eur J Cancer. 2009;45:2138–45.CrossRef
15.
Zurück zum Zitat Rubio CA. Arrest of cell proliferation in budding tumor cells ahead of the invading edge of colonic carcinomas. A preliminary report. Anticancer Res. 2008;28:2417–20.PubMed Rubio CA. Arrest of cell proliferation in budding tumor cells ahead of the invading edge of colonic carcinomas. A preliminary report. Anticancer Res. 2008;28:2417–20.PubMed
16.
Zurück zum Zitat Ueno H, Jones AM, Wilkinson KH, Jass J, Talbot I. Histological categorisation of fibrotic cancer stroma in advanced rectal cancer. Gut. 2004;53:581–6.CrossRef Ueno H, Jones AM, Wilkinson KH, Jass J, Talbot I. Histological categorisation of fibrotic cancer stroma in advanced rectal cancer. Gut. 2004;53:581–6.CrossRef
17.
Zurück zum Zitat Ueno H, Mochizuki H, Hashiguchi Y, et al. Risk factors for an adverse outcome in early invasive colorectal carcinoma. Gastroenterology. 2004;127:385–94.CrossRef Ueno H, Mochizuki H, Hashiguchi Y, et al. Risk factors for an adverse outcome in early invasive colorectal carcinoma. Gastroenterology. 2004;127:385–94.CrossRef
18.
Zurück zum Zitat Ueno H, Murphy J, Jass J, Mochizuki H, Talbot I. Tumourbudding’as an index to estimate the potential of aggressiveness in rectal cancer. Histopathology. 2002;40:127–32.CrossRef Ueno H, Murphy J, Jass J, Mochizuki H, Talbot I. Tumourbudding’as an index to estimate the potential of aggressiveness in rectal cancer. Histopathology. 2002;40:127–32.CrossRef
19.
Zurück zum Zitat van Wyk HC, Park JH, Edwards J, Horgan PG, McMillan DC, Going JJ. The relationship between tumour budding, the tumour microenvironment and survival in patients with primary operable colorectal cancer. British J Cancer. 2016;115:156.CrossRef van Wyk HC, Park JH, Edwards J, Horgan PG, McMillan DC, Going JJ. The relationship between tumour budding, the tumour microenvironment and survival in patients with primary operable colorectal cancer. British J Cancer. 2016;115:156.CrossRef
20.
Zurück zum Zitat Wang H-S, Liang W-Y, Lin T-C, et al. Curative resection of T1 colorectal carcinoma: risk of lymph node metastasis and long-term prognosis. Dis Colon Rectum. 2005;48:1182–92.CrossRef Wang H-S, Liang W-Y, Lin T-C, et al. Curative resection of T1 colorectal carcinoma: risk of lymph node metastasis and long-term prognosis. Dis Colon Rectum. 2005;48:1182–92.CrossRef
Metadaten
Titel
The Relationship Between Tumor Budding, Tumor Microenvironment, and Survival in Patients with Primary Operable Colorectal Cancer
verfasst von
Hester C. van Wyk, PhD
Antonia Roseweir, PhD
Peter Alexander, MBChB
James H. Park, PhD
Paul G. Horgan, PhD
Donald C. McMillan, PhD
Joanne Edwards, PhD
Publikationsdatum
11.10.2019
Verlag
Springer International Publishing
Erschienen in
Annals of Surgical Oncology / Ausgabe 13/2019
Print ISSN: 1068-9265
Elektronische ISSN: 1534-4681
DOI
https://doi.org/10.1245/s10434-019-07931-6

Weitere Artikel der Ausgabe 13/2019

Annals of Surgical Oncology 13/2019 Zur Ausgabe

Update Chirurgie

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

S3-Leitlinie „Diagnostik und Therapie des Karpaltunnelsyndroms“

CME: 2 Punkte

Prof. Dr. med. Gregor Antoniadis 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“

CME: 2 Punkte

Dr. med. Benjamin Meyknecht, PD Dr. med. Oliver Pieske Das Webinar S2e-Leitlinie „Distale Radiusfraktur“ 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“

CME: 2 Punkte

Dr. med. Mihailo Andric
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.