You are on page 1of 16

Human Pathology (2015) xx, xxx–xxx

www.elsevier.com/locate/humpath

Progress in pathology

Tumor budding in colorectal cancer—ready for


diagnostic practice?☆,☆☆
Viktor H. Koelzer MD a,b,⁎, Inti Zlobec PhD b , Alessandro Lugli MD a,b
a
Clinical Pathology Division, Institute of Pathology, University of Bern, CH-3010 Bern, Switzerland
b
Translational Research Unit, Institute of Pathology, University of Bern, CH-3010 Bern, Switzerland

Received 30 April 2015; revised 30 July 2015; accepted 13 August 2015

Keywords:
Summary Tumor budding is an important additional prognostic factor for patients with colorectal cancer
Colorectal cancer;
(CRC). Defined as the presence of single tumor cells or small clusters of up to 5 cells in the tumor stroma, tumor
Tumor budding;
budding has been linked to epithelial-mesenchymal transition. Based on well-designed retrospective studies,
Prognostic factor;
tumor budding has been linked to adverse outcome of CRC patients in 3 clinical scenarios: (1) in malignant
Epithelial-mesenchymal
polyps, detection of tumor buds is a risk factor for lymph node metastasis indicating the need for colorectal
transition;
surgery; (2) tumor budding in stage II CRC is a highly adverse prognostic indicator and may aid patient
Metastasis
selection for adjuvant therapy; (3) in the preoperative setting, presence of tumor budding in biopsy material may
help to identify high-risk rectal cancer patients for neoadjuvant therapy. However, lack of consensus guidelines
for standardized assessment still limits reporting in daily diagnostic practice. This article provides a practical and
comprehensive overview on tumor budding aimed at the practicing pathologist. First, we review the prognostic
value of tumor budding for the management of colon and rectal cancer patients. Second, we outline a practical,
evidence-based proposal for the assessment of tumor budding in the daily sign-out. Last, we summarize the
current knowledge of the molecular characteristics of high-grade budding tumors in the context of personalized
treatment approaches and biomarker discovery.
© 2015 Elsevier Inc. All rights reserved.

1. Introduction (CRC) has been termed tumor budding [1] (Fig. 1). Tumor
budding has received increasing attention by gastrointestinal
The presence of de-differentiated single cells or small pathologists as a valuable prognostic factor. It is an indicator of
clusters of up to 5 cells at the invasive front of colorectal cancer aggressive tumor biology that may be driven by an
epithelial-mesenchymal transition (EMT)–like process. The
characterization of the clinical and biological implications of
☆ tumor budding has proceeded farthest in colon and rectal
Competing interest: The coauthors have no relevant affiliations or financial
involvement with any organization or entity with a financial interest in or cancers and is rapidly advancing in carcinomas of the upper
financial conflict with the subject matter or materials discussed in the manuscript. gastrointestinal tract [2], pancreas [3,4], breast [5,6], head and
This includes employment, consultancies, honoraria, stock ownership or options, neck [7], and lung [8–10]. Substantial evidence from
expert testimony, royalties, or patents received or pending. retrospective studies suggests that tumor budding is ready to
☆☆
Funding/Support: This project received no grant from any funding
take center stage as an additional prognostic factor for CRC
agency in the public, commercial, or not-for-profit sectors.
⁎ Corresponding author at: Institute of Pathology, University of Bern, patients in distinct clinical settings [11]. Specifically, detection
CH-3010 Bern, Switzerland. of tumor budding in malignant polyps may allow the
E-mail address: viktor.koelzer@pathology.unibe.ch (V. H. Koelzer). identification of patients at high risk for nodal metastasis as

http://dx.doi.org/10.1016/j.humpath.2015.08.007
0046-8177/© 2015 Elsevier Inc. All rights reserved.
2 V. H. Koelzer et al.

TUMOR BUDDING (H&E) Control (UICC) [16], the Association of Directors of Anatomic
and Surgical Pathology [17], and inclusion in the guidelines for
CRC screening, diagnosis, and treatment in Europe and Japan
[18–21].In the UpToDate information resource, tumor budding
is currently listed as a category IIB prognostic factor [22].
Convincing evidence suggests that tumor budding may be
the visible correlate of an EMT-related process in the tumor
microenvironment of CRC [23]. The detection of tumor buds
at the invasive front may therefore represent a morphologic
* link between tumor progression, vascular invasion, the spread
of circulating tumor cells, and the establishment of metastatic
disease. Because metastasis is the leading cause of death for
100µm CRC patients, the characterization of signaling aberrations
leading to tumor budding may represent an important area of
Fig. 1 Tumor budding. High-power image (×40) of tumor budding
research to discover new options for precision therapy.
cells (→) at the invasive front of CRC in a standard H&E stain. Tumor
budding is defined as the presence of single cells or small clusters of Tumor budding is frequently observed in daily diagnostic
cancer cells in the tumor stroma and is frequently found in tumors with practice but infrequently reported. In particular, standardized
an invasive growth pattern and desmoplastic stromal response. reporting is currently held back by disagreement concerning
Detection of tumor budding is frequently associated with lympho- the most reproducible scoring method, the selection of the best
vascular invasion (*) in the tumor microenvironment. block for budding assessment, and the use of immunohisto-
chemistry. The purpose of this review is therefore to present a
candidates for surgical resection [12]. In stage II CRC tumor practical and comprehensive overview on tumor budding
budding may indicate a high chance for metastatic relapse and aimed at the practicing pathologist.
may complement clinical decision making on adjuvant therapy
and follow-up [13]. Last, detection of tumor budding in
preoperative biopsies may allow the identification of rectal 2. Materials and methods
cancer patients for for neoadjuvant treatment and risk-adapted
surgery to reduce the risk of local recurrence [14,15] (Fig. 2). For review of the primary and secondary literature
The importance of tumor budding additional prognostic factor is concerning the prognostic impact of tumor budding in CRC
reflected in publications by the Union for International Cancer (stage I, stage II, preoperative biopsies), electronic keyword

TUMOR BUDDING
Potential clinical scenarios
Malignant polyps

Tumor budding as a predictor of lymph node metastases

Clinical implication: surgical resection

Stage II CRC

Tumor budding as an adverse prognostic factor

Clinical implication: risk adapted follow up and adjuvant therapy

Pre-operative biopsies of colon and rectal cancer

Tumor budding as an adverse prognostic factor and predictor of lymph node and distant metastasis

Clinical implication: neo-adjuvant therapy and risk adapted surgery

Fig. 2 Potential clinical scenarios to report tumor budding in CRC. Assessment of tumor budding can provide valuable prognostic information in
distinct clinical settings. In malignant polyps, presence of tumor budding as reported by the gastrointestinal pathologist indicates a significantly
elevated risk for nodal metastasis. Surgical resection may be considered. In stage II disease, tumor budding is an independent adverse prognostic
factor that can contribute to the identification of high-risk patients for neoadjuvant therapy and follow-up. Detected in preoperative biopsy material
of colon and rectal cancer patients, tumor budding is an adverse prognostic indicator and correlates with an elevated risk for metastatic spread to
lymph nodes and distant organs. Risk-adapted surgery and neoadjuvant treatment may be a valid option for these patients.
Tumor budding in colorectal cancer 3

searches using Boolean operators were performed in MED- Importantly, it is already considered good practice to
LINE, MEDLINE In Process, Scopus, Web of Science, report tumor budding in early stage CRC according to the
EMBASE, Google Scholar, and ISI Proceedings. Reference European guidelines for quality assurance in CRC screening
lists were searched manually to locate additional items. Search and diagnosis [18]. The importance of including a comment
queries were not limited by date and include all literature on tumor budding in early stage lesions is further highlighted
published up to the 30th of April 2015 (date of submission). by the European Society for Medical Oncology recommen-
Case reports or notes were excluded. All items that met the dations for clinical management of patients with colon and
inclusion criteria were reviewed and assessed for relevance. rectal cancers: the 2012 guidelines advocate standard
Relevant studies were retrieved, catalogued, and categorized surgical resection for early stage CRC if high-risk features
according to the clinical setting of interest. Data tables were including grading greater than 2, invasion of submucosa,
reviewed by all authors of this article. lymphatic or venous invasion, resection margins less than 1
mm, tumor budding, or invasive carcinoma in a sessile polyp
are detected, even after definite R0 removal [19].
3. Tumor budding as a histomorphologic
prognostic factor in colon and rectal cancers 3.2. Stage II CRC

3.1. Early stage CRC AJCC/UICC stage II CRC (pT3/4, pN0, M0) encompass a
heterogeneous group of patients with an observed 5-year
Early stage CRC (American Joint Commitee on Cancer survival ranging from 66.5% (stage IIA) to 37.3% (stage IIC)
(AJCC) and UICC stage I; pT1/2, pN0, M0) is commonly [28]. This contrasts with an observed 5-year survival of 73.1%
identified in the histopathology laboratory after endoscopic (stage IIIA) and 46.3% (stage IIIC) for node-positive patients
polyp removal. Although most patients fare well after the [28]. Clearly, some node-negative, stage II patients will face
complete endoscopic removal of a malignant polyp, up to 17% early relapse of disease after resection of the primary tumor due
already have clinically unapparent micrometastasis in to the outgrowth of clinically unapparent micrometastasis. The
locoregional lymph nodes at the time of resection [24,25]. identification of these stage II patients with an elevated risk of
Additional histomorphologic factors are therefore needed to recurrence through the use of additional pathological risk
identify patients that may be candidates for colorectal surgery. factors is therefore important to support clinical decision
Based on retrospective cohort studies, the assessment of tumor making on follow-up and adjuvant therapy. For high-risk
budding can provide important additional information on the patients, adjuvant therapy can be considered based on a careful
risk of nodal metastasis in this setting (Table 1). assessment of potential risks and benefits including prognosis
These results have been well confirmed by recent [29–31]. According to the 2015 NCCN guidelines, high-risk
meta-analyses using robust statistical methods: in a system- features include “poorly differentiated histology (exclusive of
atic review of 23 cohort studies including 4510 pT1 patients, those cancers that are high microsatellite instability), lymphat-
Beaton and colleagues [24] report high-grade tumor budding ic/vascular invasion, bowel obstruction, b12 lymph nodes
to be significantly associated with nodal metastasis (odds examined, perineural invasion, localized perforation, or close,
ratio [OR], 7.74; 95% confidence interval [CI], 4.47-13.39; indeterminate, or positive margins” [31].
P b .001) exceeding the relative risk (RR) of other Importantly, the presence of tumor budding is increas-
established adverse prognostic indicators including greater ingly recognized as an additional high-risk feature for
than 1-mm depth of submucosal invasion (OR, 3.87; 95% CI, AJCC/UICC stage II CRC patients. Based on a series of
1.50-10.00; P = .005), lymphatic invasion (OR, 4.81; 95% well-designed retrospective cohort studies, high-grade
CI, 3.14-7.37; P b .00001), and poor tumor differentiation tumor budding is significantly linked to poor overall and
(OR, 5.60; 95% CI, 2.90-10.82; P b .00001). These findings disease-free survival of curatively resected stage II CRC
are underlined by separate studies from Japan [26] and the [13,32–37] (Table 2). In fact, survival rates of stage II
Netherlands [27]: in a systematic review of 3556 pT1 patients with high-grade tumor budding were found not to
carcinomas from 30 different Japanese institutions, Ueno and differ significantly in a direct comparison with a matched
colleagues [26] report the presence of tumor budding, poorly cohort of stage III patients in a retrospective analysis
differentiated clusters, poor tumor differentiation, vascular including 446 CRC patients [13]. Furthermore, presence of
invasion, and submucosal invasion depth as significant risk tumor budding was consistently related to aggressive
factors for lymph node metastasis (all, P b .0001). Based on clinicopathological features including lymphatic and ve-
a meta-analysis of 17 published studies including 3621 nous invasion, higher tumor grade, and an infiltrative tumor
patients, Bosch and associates [27] confirm lymphatic margin [13,32–37]. Taken together, tumor budding is an
invasion (RR, 5.2; 95% CI, 4.0-6.8), submucosal invasion important additional prognostic indicator in stage II disease
at least 1 mm (RR, 5.2; 95% CI, 1.8-15.4), budding (RR, 5.1; and may aid identification of high-risk patients for adjuvant
95% CI, 3.6-7.3), and poor histologic differentiation (RR, therapy. Tumor budding is therefore a promising candidate
4.8; 95% CI, 3.3-6.9) as significant pathological risk factors for inclusion into reporting guidelines of stage II disease and
for nodal metastasis in pT1 CRC. validation in prospective clinical trials.
4
Table 1 Tumor budding as a risk factor for lymph node metastasis in stage I CRC
Study Year n (no. of EMR) Assessment method High-grade TB % Clinicopathological features Clinicopathological features significantly
analyzed for correlation with associated with lymph node metastasis (P b .05)
lymph node metastasis
Okuyama et al [80] 2002 101 (0) Morodomi et al [81] Any TB found Age, sex, SMd, L, V, TB, location, size TB, L1, V1, location in rectum
H&E in 42%
Egashira et al [82] 2004 140 (0) Morodomi et al [81] Any TB found SMd, SMw, L, V, TB, LInf, differentiation L1, V1, depth of invasion, cribriform-type
H&E in 60% of invasive component, structural atypia structural atypia, low LInf
of invasive component, border
Ueno et al [63] 2004 285 (80) Ueno et al [63] H&E 15.1% Age, sex, G, SMd, SMw, L, TB, G, LVI, cribriform growth pattern
V, TB, location, size, TuConf
Kitajima et al [83] 2004 865 Hase et al [52] H&E 42.3% G, SMd, L, V, TB, status of TB, SMd, L1, V1, status of muscularis mucosae,
muscularis mucosae, Histological histological type at deepest portion
type at deepest portion
Wang et al [84] 2005 159 (0) Morodomi et al [81] 15.1% Age, G, SMd, L, V, Pn, TB, TB, G, L1, low LInf
H&E border, location, CEA level,
Borrmann type, mucinous
component, no. of lymph nodes
sampled, LInf, MSI
Kazama et al [85] 2006 56 (0) Morodomi et al [81] 75.0% TB TB
PanCK
Masaki et al [86] 2006 76 (21) Ono et al [87] H&E Continuous score G, SMd, SMw, L, V, TB, size, TuConf TB, G
Kawaura et al [88] 2007 122 Ueno et al [63] H&E 18.8% G, SMd, L, V, TB, size, location, TB, SMd, L1, V1, G (univariate)
LInf, TuConf, histologic type at L1, V1 (multivariate)
deepest portion, lymphatic density,
adenomatous component
Kaneko et al [89] 2007 214 (N.S.) Ueno et al [63] H&E 18.2% G, SMd, L, V, TB, size, location, TB, TuConf, G, L1
TuConf, no. of lymph nodes sampled
Sohn et al [90] 2007 48 (0) Ueno et al [63] H&E 20.8% G, SMd, LVI, TB, size TB, LVI
Yasuda et al [91] 2007 86 (0) Morodomi et al [81] 46.5% Age, sex, G, SMd, LVI, TB, location, TB, LVI
H&E size, TuConf
Ishikawa et al [92] 2008 71 (0) Morodomi et al [81] 50.7% (cutoff=4) Age, sex, G, SMd, L, V, TB, location, TB (multivariate), L1
H&E and PanCK size, TuConf,
Yamauchi et al [93] 2008 164 (62) Ueno et al [63] H&E 14.6% G, SMd, L, V, TB TB, G
Choi et al [94] 2009 168 (0) Ueno et al [46] H&E 18.4% G, SMd, TB, LVI, V, resection status TB (multivariate)
Ogawa et al [95] 2009 98 (15) Ueno et al [46] H&E 48.9% Age, sex, G, SMd, L, V, TB, location, TB, G (multivariate)
size, TuConf

V. H. Koelzer et al.
Suzuki et al [47] 2009 124 (0) Ueno et al [46] H&E, 7.3% (H&E) L, V, TB TB, V1 (using elastic stain)
PanCK 17.7% (PanCK)
Kajiwara et al [96] 2010 244 (0) Ueno et al [46] H&E 44.7% Age, sex, G, SMd, L, V, TB, location, TB, L1, poorly differentiated component, myxoid
size, border TuConf, annularity, poorly cancer stroma, border (univariate)
differentiated component, myxoid Poorly differentiated component, L1, myxoid
cancer stroma, infiltration level in cancer stroma (multivariate)
muscularis propria,
Tumor budding in colorectal cancer
Komori et al [97] 2010 111 (0) Ueno et al [46] H&E SMd, SMw, L, V, TB, area of SM TB, Histological type
invasion, histologic type at deepest
invasion
Tateishi et al [98] 2010 322 (0) Ueno et al [46] H&E 32.9% G, SMd, L, V, TB, disruption of TB (multivariate), L1, G
muscularis mucosae
Ueno et al [99] 2010 296 (67) Ueno et al [63] H&E 11.8% G, SMd, SMw, L, V, TB, size, TB, SMd, SMw, LVI, cribriform pattern, extent
TuConf, growth pattern, adenoma of poorly differentiated component, extent of
component, extent of poorly mucin-producing component, grade of least-
differentiated component, cribriform differentiated component
pattern, extent of mucin-producing
component
Akishima-Fukasawa 2011 111 (0) Ueno et al [63] H&E 50.5% G, SMd, SMw, L, V, TB, TuConf, TB, SMd, G, L1, border, disruption of muscularis
et al [100] border, disruption of muscularis mucosa, neutrophil infiltration in cancer cells,
mucosae, neutrophil infiltration in fibrotic cancer-type stroma, Crohn-like reaction,
cancer cells, fibrotic cancer-type microscopic abscess formation
stroma, LInf, Crohn-like reaction,
microscopic abscess formation
Keum et al [101] 2012 434 (0) Ueno et al [46] H&E 40% in patients Age, sex, G, L, V, TB, location, TB found in all 20 patients with recurrence
with recurrence size, border, TuConf, annularity,
poorly differentiated component,
myxoid cancer stroma, infiltration
level in muscularis propria
Kye et al [102] 2012 55 (0) Ueno et al [46] H&E 36% Age, sex, SMd, LVI, TB, location, TB (multivariate)
size, TuConf, circumference ratio,
presence of microacinar structures
Nakadoi et al [103] 2012 499 (150) Ueno et al [63] H&E Colon 9.2%; G, SMd, LVI, TB TB (multivariate), G, SMd, LVI
rectum 15.3%
Suh et al [104] 2012 435 (111) Ueno et al [46] H&E 17.6% G, SMd, LVI, TB, background TB, G, LVI, No background adenoma
adenoma
Oka et al [105] 2013 118 Modified Ueno et al 15.3% G, SMd, LVI, TB TB
[63] H&E
Umemura et al [106] 2013 142 Ueno et al [63] H&E 11.2% H&E, Age, sex, SMd, L, V, TB, location, TB (as assessed by IHC only), SMd (multivariate)
and β-catenin 9.8% β-catenin TuConf, size, β-catenin expression,
operation method
Beaton et al [24] 2013 Meta-analysis N.A. N.A. G, SMd, LVI, TB TB (OR, 7.74; 95% CI, 4.47-13.39; P b .001),
of 23 studies G (OR, 5.60; 95% CI, 2.90-10.82; P b .00001),
(n = 4510) LVI (OR, 4.81; 95% CI, 3.14-7.37; P b .00001),
SMd ≥1 mm (OR, 3.87; 95% CI, 1.50-10.00,
P = .005)
Ueno et al [107] 2014 3556 patients from Ueno et al [63] H&E 19.4% G, SMd, LVI, TB, PDC G (OR, 7.2; 95% CI, 4.4-11.8; P b .0001),
30 institutions LVI (OR, 4.8; 95% CI, 3.8-6.0; P b .0001),
SMd ≥1mm (OR, 4.0; 95% CI, 2.5-6.5;
P b .0001)
TB (OR, 3.8; 95% CI, 3.0-4.7; P b .0001)
PDC (OR, 3.3; 95% CI, 2.6-4.1; P b .0001)

5
6
Table 1 (continued)
Study Year n (no. of EMR) Assessment method High-grade TB % Clinicopathological features Clinicopathological features significantly
analyzed for correlation with associated with lymph node metastasis (P b .05)
lymph node metastasis
Bosch et al [27] 2013 Meta-analysis N.A. N.A. G, SMd (based on SM levels, cutoff LVI (RR, 5.2; 95% CI, 4.0-6.8),
of 17 studies value 1 mm and cutoff value 2 mm), SMd ≥1mm (RR, 5.2; 95% CI, 1.8-15.4),
(n = 3621) SMw, L, V, LVI, TB, Linf, border, TB (RR, 5.1; 95% CI, 3.6-7.3),
Tu Conf, poor differentiation at G (RR, 4.8; 95% CI, 3.3-6.9)
invasion front, size, histologic type,
cribriform subtype, microvessel density
Suh et al [108] 2013 150 (75) Ueno et al [46] H&E 29.3% in surgically G, SMd, LVI, TB, tumor size, TB, SMd, LVI (univariate)
treated patients TuConf, margin status TB, SMd (multivariate)
Ryu et al [109] 2014 179 Ueno et al [46] H&E 18.9% Age, sex, G, SMd, LVI, TB, TuConf, TB, G, L1, SMd, border, tumor depth, presence of
Pn, border, tumor depth, preexisting cribriform structures, desmoplasia, micropapillary
adenoma, presence of cribriform structures, mucinous differentiation, absence of
structures, desmoplasia, micropapillary Crohn-like reaction
structures, mucinous differentiation,
Crohn-like reaction, intraglandular
necrosis, clear cell change, signet ring
differentiation, solid differentiation
Caputo et al [110] 2014 48 Ueno et al [46] H&E 12.5% G, SMd, LVI, TB G
Macias-Garcia 2015 97 Ueno et al [63] H&E 13.4% Age, sex, G, SMd, L, V, TB, LInf, TB, G, L1, absence of LInf, border, sessile
et al [111] border, TuConf, size, location, stalk morphology (univariate)
invasion, presence of background G, border, absence of LInf, sessile morphology
adenoma (multivariate)
Kawachi et al [112] 2015 806 Ueno et al [63] H&E 28.9% Age, sex, G, SMd, L, V, TB, size, TB, SMd, G, L1, V1, nonpedunculated TuConf
location, TuConf (univariate)
TB, SMd (multivariate)
Wada et al [113] 2015 Meta-analysis All Ueno et al [63] N.A. G, SMd, LVI, TB, LInf, differential TB (OR, 7.45; 95 % CI, 4.27-13.02; P = .0077),
of 5 studies H&E/IHC subtype at the deepest portion, L1 (OR, 5.19; 95 % CI, 3.31-8.15; P = .01)
(n = 770) lymphatic vessel density,
adenomatous component
Abbreviations: Border, tumor border configuration; CEA, carcinoembryonic antigen; CI, confidence interval; CRC, colorectal carcinoma; EMR: endoscopic mucosal resection; G, tumor grade; H&E,
hematoxylin and eosin; IHC, immunohistochemistry; L1, lymphatic invasion; LInf, lymphoid cell infiltrates; LVI, lymphovascular invasion; MSI, microsatellite instability; N.A., not applicable; N.S., not
significant; OR, odds ratio; PanCK, pan-cytokeratin; PDC, poorly differentiated clusters; Pn, perineural invasion; SMd, depth of submucosal invasion; SMw, width of submucosal invasion; TB, tumor budding;
TuConf, macroscopic tumor configuration; V1, venous invasion.

V. H. Koelzer et al.
Tumor budding in colorectal cancer
Table 2 Tumor budding as a histopathologic prognostic factor in stage II CRC
Study Year n Method High-grade TB % Clinicopathological End point Outcome
features correlated
with TB (P b .05)
Tanaka et al 2003 138 Hase et al [52] H&E 19.6% Disease recurrence Disease-specific 5-year 5-y survival rates with low-grade TB: 98%;
[33] pT3 survival rate high-grade TB: 74% (P b .0001; univariate)
Okuyama et al 2003 83 Morodomi et al [81] H&E 57.9% L1, pN1, local recurrence, Cumulative disease-specific 5-y survival rates with low-grade TB: 85.0%;
[114] (rectal) liver metastasis 5-y survival rate high-grade TB: 51.8% (P b .002)
pT3
Masaki et al 2005 72 Raw number of buds; H&E N.A. pN1 pN stage N.A.
[115] (rectal)
pT2
Nakamura et al 2008 200 Nakamura et al [13] 34.5% Disease recurrence Cumulative 5-y and 5-y survival rates with low-grade TB: 93.9%;
[13] pT3-4 H&E (peritoneal and 10-y survival rates high-grade TB: 73.9% (P b .001).
liver metastasis) 10-y survival rates with low-grade TB: 90.6%;
high-grade TB: 67.8% (P b .001).
Wang et al [34] 2009 128 Wang et al [34] H&E 45% LVI, infiltrative tumor Disease-specific 5-y 5-y survival rates with low-grade TB: 91%;
pT3 margin survival rate high-grade TB: 63% (P b .0001).
Kevans et al 2011 258 Wang et al [34] H&E 44% TB more frequent in 5-y survival rate TB independently associated with survival
[116] (122 cases microsatellite stable (HR, 7.9; 95% CI, 3-21; P b .001)
analyzed) tumors
Betge et al [35] 2012 120 Modified Ueno et al [46] H&E; 30% LVI, G 5-y progression-free 5-y progression-free survival rates with
pT3-4 area 0.95 mm2; high-grade TB and disease-specific low-grade TB: 91%; high-grade TB:
defined as ≥10 buds/HPF survival rates 65% (P = .009).
5-y disease-specific survival rates with
low-grade TB: 94%; high-grade TB:
72% (P b .001)
Canney et al 2012 205 Wang et al [34] H&E pT3 44%, pT4a TB most frequent in 5-y survival rate Not significant
[36] pT3-4b 73%, pT4b 25% pT4a disease
Horcic et al 2013 105 Interobserver study (PanCK): Hase et al: 60%, Infiltrating tumor Disease-specific 1-HPF and 10-HPF methods (1 HPF,
[37] pT3-4 Hase et al [52], Nakamura Nakamura et al 59.1%, border configuration 5-y survival rate P = .0007; 10 HPF, P = .0067) and
et al [13], Wang et al [34], Ueno et al 83% (except Wang et al) Wang (P = .0459) significantly linked
1 HPF [57], 10 HPFs [50] Wang et al: 41.9% to patient outcome based on a single or
multiple observers (univariate).
1 HPF and 10 HPFs significantly linked
to survival outcome in multivariate analysis.
Lai et al [32] 2014 135 Ueno et al [46] H&E 27 N.A. Progression-free survival 5-y progression-free survival rates with
pT3-4 disease-specific survival low-grade TB: 89%; high-grade TB:
57.6% (P b .001).
5-y disease-specific survival rates with
low-grade TB: 92%; high-grade TB:
66.7% (P b .001)
Abbreviations: Border, tumor border configuration; HR, hazard ratio; L1, lymphatic invasion; LVI, lymphovascular invasion; N.A., not applicable; PanCK, pan-cytokeratin; pN, pathological nodal stage; pT,
pathological tumor stage; TB, tumor budding; 1 HPF, 1-high-power field method; 10 HPF, 10-high-power field method.

7
8 V. H. Koelzer et al.

3.3. Tumor budding in preoperative biopsies of good) [32,34,45–51]. However, interobserver concordance
colon and rectal cancers was frequently assessed using a limited subset of cases and
scores from a single academic center, which may artificially
The preoperative biopsy of primary CRC is an essential step reduce statistical divergence. Systematic interobserver
for confirmation of diagnosis but currently fails to provide studies comparing different scoring methods with observers
prognostic information to the clinician beyond a rough from different institutions are rare: In a virtual microscopy
estimation of tumor grade, assessment of mismatch-repair study including 10 investigators from different centers, the
protein expression, and molecular pathology studies. Recent scoring methods by Hase et al [52], Nakamura et al [53],
studies indicate that preoperative biopsy material may contain Ueno et al [54,55], and Wang et al [34] were systematically
significant prognostic information based on the assessment of compared using both hematoxylin and eosin (H&E)– and
tumor budding and immune cell infiltrates [14,15,38,39] cytokeratin-stained sections. For these methods, overall
(Table 3). In particular, presence of tumor budding in colon agreement was fair and primarily dependent on the
and rectal cancers biopsies may be a strong indicator of nodal participants' experience with the assessment of tumor
and distant metastasis at the time of resection [15,40] and has budding and training in gastrointestinal pathology [56].
been associated with nonresponse to neoadjuvant chemora- Use of immunohistochemistry led to an increase in the
diotherapy and poor survival outcome in rectal cancer patients reported tumor budding counts but did not improve
[14]. As the infiltrative margin is rarely sampled on biopsy interobserver reproducibility.
specimens of CRC, intratumoral budding is assessed for In a subsequent study using cytokeratin-stained sections
prognostication [40]. A cutoff of 6 tumor buds in 1 high-power scored by 3 observers, tumor budding as assessed according
field (HPF; ×40) has been proposed to identify high-risk to Hase, Nakamura, Ueno, Wang (conventional and rapid
patients, but assessment of tumor budding on a continuous method), the densest HPF (1-HPF method [57]) and the 10
scale may allow for a more precise risk estimate to complement densest HPFs (10-HPF method [50]) were systematically
clinical decision making in the preoperative setting [15,41]. In compared in respect to prognostic impact and interobserver
addition to radiology, endoscopic imaging, and ultrasound for reproducibility [37]. In this analysis, the 1-HPF and 10-HPF
local staging, risk factors present in the preoperative biopsy methods (intraclass correlation coefficient [ICC] = 0.83 and
may thereby contribute important prognostic and possibly 0.91, respectively) significantly outperformed the scoring
predictive information for the multidisciplinary management methods according to Hase (κ = 0.29-0.51), Nakamura (κ =
of colon and rectal cancers patients. 0.41-0.52), Ueno (κ = 0.39-0.56), and Wang (conventional
method, κ = 0.46-0.62, and rapid method, κ = 0.46-0.58
respectively).
Notably, only tumor budding as evaluated in 1-HPF and
4. Tumor budding: scoring systems and 10-HPF methods had significant prognostic effects on
interobserver variability patient survival in a cohort of 105 well-characterized stage
II CRC patients [37]. The 1-HPF and 10-HPF methods were
Ease of assessment and interobserver reproducibility are subsequently tested in a multicenter interobserver study
central prerequisites for the establishment of any histomorpho- involving 6 independent community and academic centers
logic prognostic indicator in daily diagnostic practice. In using both H&E- and cytokeratin-stained slides [41]. In this
retrospective analyses, commonly used additional prognostic analysis, 3 to 4 times more tumor buds were detected in
factors such as tumor grade [42,43] and vascular invasion [44] cytokeratin as compared with H&E-stained slides. Importantly,
have been shown to be only moderately reproducible between excellent interobserver correlations for tumor budding scores
observers. It is therefore essential to identify the most effective (10 HPFs: ICC= 0.83 and 1 HPF: ICC= 0.85) were reached
and reproducible method of assessment for tumor budding based using cytokeratin stains, whereas interobserver agreement
on well-designed interobserver studies and to agree on for H&E slides was only moderate (10 HPFs: ICC= 0.58 and
transparent guidelines for routine application. This includes 1 HPF: ICC= 0.49) [41].
testing the methodological approach in clearly defined clinical
scenarios (eg, scoring of buds in biopsy material, malignant
polyps, and resection specimens [Fig. 2]) to provide reproduc- 5. Practical, evidence-based proposal for the
ible prognostic data for the interdisciplinary management of assessment of tumor budding in daily
CRC patients. diagnostic practice
4.1. Diagnostic reproducibility of tumor budding Agreement on the optimal method for the assessment of
tumor budding is of key importance to transfer the available
Several investigators have included an assessment of evidence regarding interobserver reproducibility, practica-
interobserver variation in prognostic studies. Based on bility, and prognostic impact into diagnostic practice. Central
scores from 2 or more observers, reported κ values for tumor aims are as follows: definition of clear parameters for
budding scores range from 0.41 (moderate) to 0.938 (very selection of the tissue block, optimal visualization of tumor
Tumor budding in colorectal cancer
Table 3 Tumor budding in preoperative biopsies as an indicator of adverse prognosis and elevated risk of metastasis
Study Year n Method High-grade TB % Clinicopathological features End point Outcome
correlated with TB (P b .05)
Morodomi 1989 112 rectal cancer Morodomi et al 46% (ITB, biopsy) ITB in biopsy associated with N.A. N.A.
et al [81] patients (all with [81] H&E presence of pN1 and L1 in the
preoperative biopsies, resection specimen
40 with matched
resection specimens)
Giger et al 2012 135 colon cancer Modified Nakamura 17% (ITB, biopsy), ITB in biopsy associated with N.A. N.A.
[40] patients (72 with et al [13] H&E 57% (PTB, resection presence of pN1 and higher
preoperative biopsies) specimen) tumor grade in the resection
specimen. More ITB in biopsy
in patients with distant metastasis
Rogers et al 2013 185 rectal cancer Modified Nakamura 20% (ITB, biopsy) ITB in biopsy associated with Disease-Free survival, High-grade ITB in the preoperative
[14] patients (89 with et al [13] H&E more-advanced ypT stage, ypN1, cancer-specific death, biopsy was associated with a lower
preoperative biopsies, residual poorly differentiated chemotherapy response disease-free 5-y survival rate (33% vs
all received neoadjuvant tumors and LVI in resection 78%, P = .001), lower cancer-specific
treatment) specimen 5-y survival rate (61% vs 87%,
P = .021), and predicted cancer-
specific death (HR, 3.51; 95% CI,
1.03-11.93; P = .040) and
nonresponse to neoadjuvant
chemoradiotherapy
Zlobec et al 2014 346 CRC patients 1-HPF method 0-10 buds (n = 97), ITB in biopsy associated with more N.A. N.A.
[15] (185 with preoperative [57] PanCK 11-20 buds (n = 19), advanced pT stage, L1, and V1 in
biopsies, patients who 21-30 buds (n = 7), resection specimen. More ITB in
received neoadjuvant or 31-50 buds (n = 6), biopsy in patients with distant
preoperative treatment N50 buds (n = 4) metastasis
excluded)
Abbreviations: CI, confidence interval; H&E, hematoxylin and eosin; HPF, high-power field; HR, hazard ratio; ITB, intratumoral budding; L1, lymphatic invasion; LVI, lymphovascular invasion; N.A., not
applicable; PanCK, pan-cytokeratin; PTB, peritumoral budding; pN, pathological nodal stage; pT, pathological tumor stage; TB, tumor budding; V1, venous invasion.

9
10 V. H. Koelzer et al.

budding cells, and standardized scoring criteria for the STANDARD HISTOLOGY (H&E)
relevant clinical scenarios (stage II CRC, preoperative
biopsies, malignant polyps).
A

• Selecting the optimal tissue block: the assessment of


resection specimens needs to account for tumor
heterogeneity. This is reached by selecting the block
with the highest grade of tumor budding for further
analysis during sign-out of the H&E slides. Because
tumor budding is most often found in association with
an infiltrative tumor margin [1,58], this can be used as a
surrogate criterion to select the optimal tissue block.

• Optimal visualization of tumor budding cells: although IMMUNOHISTOCHEMISTRY (AE1-AE3/CD8)


tumor budding can be assessed in H&E in unproblematic
cases, tumor buds can be obscured by peritumoral B
inflammation and reactive stromal fibroblasts and may
be confused with ruptured glands (Fig. 3). This
contributes to suboptimal interobserver reproducibility
and the failure of standard histology to reflect the true
amount of tumor budding cells in an individual
case [41,56,59,60]. Immunohistochemistry facilitates the
detection of tumor budding cells and may significantly
improve interobserver reproducibility [12,41]. Many
studies have so far failed to provide the exact features
used for the identification of tumor budding cells in 50µm
cytokeratin stains. To address this problem, we recently
implemented standardized features for the identification Fig. 3 Optimal visualization of tumor buds. Comparative
of tumor buds in CRC resection specimens by cytokeratin high-power image (×40) of tumor budding in a CRC with strong
peritumoral inflammation, stromal activation, and glandular
immunohistochemistry [41]:
destruction. Serial sections were stained with standard H&E and
- Tumor buds are single cells or clusters of up to
an immunohistochemical double stain (pan-cytokeratin AE1/AE3,
5 cells (≤5 cells) present in the peritumoral stroma brown; CD8, red) to visualize tumor budding cells and peritumoral
- Tumor buds show cytoplasmic reactivity to cytoker- CD8+ lymphocytes. In cases with strong inflammation, the
atin stains and a clearly identifiable nucleus. differentiation of tumor budding cells from reactive stromal
- Cytoplasmic pseudofragments, ruptured glands, fibroblasts and activated histiocytes can be challenging using
mucin pools, and necrosis are excluded. standard H&E stains. Cytokeratin facilitates detection of tumor
budding cells and allows for the scoring of tumor buds with strong
• Standardized scoring criteria: importantly, tumor budding reproducibility and specificity.
cells can be manually quantified under the microscope and
by using digital analysis methods [61,62]. Just as mitotic reproducibility [37,41,50]. To assess tumor budding in the
counts in breast cancer, tumor budding is a continuous 10-HPF method, the invasive front is first scanned at low
feature and can be assessed as such. Quantitative magnification (×4-×10) to identify areas of highest budding
assessment of tumor buds allows for the combination of density [37]. Tumor buds are then counted under high
stringently defined histomorphologic criteria with a defined magnification (×40) and the tumor budding count is reported
visual field size. Importantly, numeric tumor budding (Fig. 4A). Based on previous studies, patients with an
counts are highly reproducible based on interobserver average of more than 10 buds per HPF (100 buds is total)
studies [37,41,50], whereas qualitative assessment may have a highly adverse prognosis [50]. However, raw tumor
suffer from poor interobserver reproducibility [37]. budding counts can be used to directly calculate the probability
of clinically relevant outcomes [37,41]. We therefore advise to
Two central scenarios are met in daily diagnostic practice additionally report the actual tumor budding count to aid
when signing out CRC specimens. Most commonly, tumor clinical decision making.
budding will be assessed in CRC resection specimens. In this In contrast to resection specimens, limited material will be
setting, tumor budding as assessed in the 10 HPPs of highest available for the assessment of tumor budding in preoperative
density along the tumor invasion front (10-HPF method) has biopsies and malignant polyps. Importantly, tumor budding
been shown to reliably capture associations with adverse counts as assessed in a single hotspot strongly correlate with
clinicopathological features [37,50] with superior interobserver prognosis as shown by using the 1-HPF [15,37,57], Giger
Tumor budding in colorectal cancer 11
RESECTION SPECIMENS – 10HPF METHOD MALIGNANT POLYPS – 1HPF METHOD
Field selection on low power Field selection on low power

A B

4x-10x 4x-10x

Counting buds in 10 high power fields (10HPF) Counting buds in 1 high power field (1HPF)

40x 40x

BIOPSY FRAGMENTS – 1HPF METHOD


Field selection on low power

4x-10x

Counting buds in 1 high power field (1HPF)

40x

Fig. 4 A, For assessment of tumor budding in CRC resection specimens using the 10-HPF method, the invasive front is first scanned at low power
(×5-×10) to identify areas of the highest budding density (top panel). Tumor buds are then counted in a total of 10 HPFs (×40, field area 0.49 mm2)
along the invasion front (bottom panel). B and C, For assessment of tumor budding in malignant polyps or biopsy material, the specimens are first
scanned at low power (×5-×10) to identify the hotspot with the most tumor buds (top panel). Tumor buds are then counted in 1 HPF (×40, field area 0.49
mm2) in the area of highest density (bottom panel).
12 V. H. Koelzer et al.

[14,40], and Ueno methods [46,63]. Based on the proven suggests that tumor budding cells at the invasive front may
reproducibility of using pan-cytokeratin stains for the 1-HPF be formed in a process likened to EMT. In support of this
method in CRC resection specimens [37,41,56], we currently theory, tumor budding is particularly frequent in CRC with
favor this approach when assessing tumor budding in biopsy constitutive activation of WNT signaling, characterized by
material and malignant polyps (Fig. 4B and C). However, loss of membranous E-cadherin and nuclear translocation of
interobserver reproducibility of the 1-HPF method still needs β-catenin [65–68]. On a molecular level, APC mutations are
to be independently confirmed in this setting. frequently found in high-grade budding cancers, providing
additional evidence for WNT signaling as a driver of
invasive growth [65,69]. TGF-β signaling may additionally
contribute to stromal remodeling and activation of EMT
5.1. Relation of budding to grade, tumor border
[70,71]. Furthermore, activation of tyrosine-kinase receptor
configuration, and special morphologic variants pathways including RAS, RAF, and several mitogen-
of CRC activated protein kinases may add to the activation of EMT
and the generation of the tumor budding phenotype
Tumor budding is a conceptually independent histomor- (reviewed by Dawson and Lugli [72]). Indeed, presence of
phologic feature of CRC and does not count toward tumor activating BRAF and KRAS mutations has been previously
grade [17]. In fact, tumor budding is also frequently observed associated with high-grade tumor budding in CRC [73,74].
at the invasive front of tumors with a predominantly glandular Once formed, tumor buds display an armamentarium of
growth pattern that are graded as well or moderately enzymes allowing for efficient migration in the tumor stroma.
differentiated according to World Health Organization In particular, expression of metalloproteinases and cathepsin
recommendations [64]. Furthermore, the presence of tumor allows for the breakdown of stromal collagen and efficient
budding does not influence the assessment of tumor border invasion of host tissue [75]. Tumor budding cells have also
configuration [1,58]. Although tumor budding is most been assigned an increased resistance to apoptotic stimuli,
common in cancers with an invasive-type margin as assessed chemotherapeutics, and immunogenic cell death based on a
under low power, tumor budding is an independent, super- reduced expression of caspases [76] and Raf-kinase inhibitory
imposed feature observed at high magnification and should be protein (RKIP), an important checkpoint inhibitor of the
separately reported [1]. RAF-kinase-signaling pathway [77]. Indeed, residual tumor
Two specific, but infrequent histologic subtypes of CRC budding cells can be observed in fibrotic tumor beds after
can pose a challenge when assessing tumor budding cells at the neoadjuvant treatment of rectal cancer, adding morphologic
invasive front. First, carcinomas with a primary dissociative evidence to these correlative analyses [78].
growth pattern or signetring cell differentiation do not allow Recent studies suggest that tumor budding cells also
the differentiation of “tumor buds” from the main tumor body. interact with the host immune system. In patients with a
It has been previously suggested that primary CRCs with a strong peritumoral infiltration by CD8+ cytotoxic T cells,
diffuse growth pattern should be classified as high-grade tumor tumor budding is infrequently encountered [57]. As CD8+
budding by definition [1]. Second, mucinous carcinomas T cells represent an important antitumoral effector mecha-
frequently form pools containing single cancer cells or small nism, a destruction of tumor budding cells in patients with a
tumor cell clusters. Because these cell clusters frequently lie in functional immune response at the tumor-host interface may
mucin pools and are not surrounded by tumor stroma, they do be postulated. As further evidence of tumor-host interaction,
not qualify as bona fide tumor buds. tumor budding cells may down-regulate components of the
major histocompatibility complex I under selective pressure
of the antitumoral host response [79]. As a consequence,
6. Molecular characteristics of tumor budding tumor budding cells and CD8+ T cells can be seen as 2
in CRC opposing sides of an attacker-defender model. This offers an
integrative basis to further refine prognostication of CRC
patients based on specific properties of the tumor microen-
Molecular features of CRC impact the morphologic vironment [57].
appearance on the histologic slide. Specific genomic and Based on the present evidence at the molecular and protein
proteomic changes in the mutational landscape of CRC may level, tumor budding cells may represent an important stage of
therefore cause the formation of tumor budding cells. The CRC progression—they are the first morphologic evidence of
identification of pathogenic signaling alterations driving the dissociative tumor growth and echo detrimental changes at the
tumor budding phenotype may allow not only a better tumor/host interface. It seems likely that tumor budding cells are
understanding of the malignant progression of colorectal indeed the source for circulating tumor cells in CRC patients and
tumors but also the development of new precision therapeu- the seed for nodal and distant metastatic disease. Consequently,
tics targeting the earliest phase of metastatic dissemination. targeting tumor budding cells may inhibit metastatic spread of
CRC with high-grade tumor budding share aggressive CRC at the earliest possible time point. Based on our current
molecular and biological features. Correlative evidence knowledge, WNT, RAS, and TGF-β signaling pathways as well
Tumor budding in colorectal cancer 13

as proteins involved in stromal degradation and the antitumoral [4] Kohler I, Bronsert P, Timme S, et al. Detailed analysis of epithelial-
immune response may provide valid options for the develop- mesenchymal transition and tumor budding identifies predictors of
long-term survival in pancreatic ductal adenocarcinoma. J Gastro-
ment of these therapeutic strategies. enterol Hepatol 2015;30(Suppl. 1):78-84.
[5] Salhia B, Trippel M, Pfaltz K, et al. High tumor budding stratifies
breast cancer with metastatic properties. Breast Cancer Res Treat
7. Conclusions and future perspectives 2015;150:363-71.
[6] Liang F, Cao W, Wang Y, Li L, Zhang G, Wang Z. The prognostic
Tumor budding is a valuable prognostic indicator for CRC value of tumor budding in invasive breast cancer. Pathol Res Pract
2013;209:269-75.
patients. Detection of tumor budding in malignant polyps [7] Almangush A, Salo T, Hagstrom J, Leivo I. Tumour budding in head and
indicates a significantly elevated probability of nodal neck squamous cell carcinoma—a systematic review. Histopathology
metastasis. After an endoscopic polyp removal, colorectal 2014;65:587-94.
surgery may therefore be indicated in a risk-adapted approach. [8] Masuda R, Kijima H, Imamura N, et al. Tumor budding is a
In stage II disease, presence of tumor budding is an significant indicator of a poor prognosis in lung squamous cell
carcinoma patients. Mole Med Rep 2012;6:937-43.
independent adverse prognostic indicator. For these patients, [9] Yamaguchi Y, Ishii G, Kojima M, et al. Histopathologic features of the
surgery may not be curative and adjuvant therapy considered. tumor budding in adenocarcinoma of the lung: tumor budding as an index
Detection of tumor budding in preoperative biopsies of colon to predict the potential aggressiveness. J Thorac Oncol 2010;5:1361-8.
and rectal cancers may indicate an increased probability for [10] Kadota K, Yeh YC, Villena-Vargas J, et al. Tumor budding correlates
metastatic dissemination to lymph nodes and distant organs. with protumor immune microenvironment and is an independent
prognostic factor for recurrence of stage I lung adenocarcinoma. Chest
Neoadjuvant treatment and risk-adapted tumor surgery can be 2015;148:711-21.
considered in this patient population. [11] Koelzer VH, Zlobec I, Lugli A. Tumor budding in the clinical
To facilitate the application of tumor budding in daily management of colon and rectal cancer. Colorectal Cancer 2014;3:
diagnostic practice, standardized guidelines for reporting and 387-403.
[12] van Wyk HC, Park J, Roxburgh C, Horgan P, Foulis A, McMillan
assessment are essential. Much progress has been made in this
DC. The role of tumour budding in predicting survival in patients
respect over recent years. Based on meaningful retrospective with primary operable colorectal cancer: a systematic review. Cancer
data and a strong evidence base, tumor budding has been Treat Rev 2015;41:151-9.
included in several reporting recommendations in the United [13] Nakamura T, Mitomi H, Kanazawa H, Ohkura Y, Watanabe M.
States, Europe, and Asia. Systematic interobserver studies Tumor budding as an index to identify high-risk patients with stage II
have contributed to the identification of the most reproducible colon cancer. Dis Colon Rectum 2008;51:568-72.
[14] Rogers AC, Gibbons D, Hanly AM, et al. Prognostic significance of
assessment methods for tumor budding. The advantages of tumor budding in rectal cancer biopsies before neoadjuvant therapy.
using immunohistochemistry to detect tumor buds of CRC Mod Pathol 2014;27:156-62.
have been demonstrated but must be weighed against the costs. [15] Zlobec I, Hadrich M, Dawson H, et al. Intratumoural budding (ITB)
Together these data form an important base to reach a working in preoperative biopsies predicts the presence of lymph node and
distant metastases in colon and rectal cancer patients. Br J Cancer
model. Taking the next step toward a unified approach will be
2014;110:1008-13.
essential to avoid that one of the potentially most valuable [16] Gospodarowicz MK, O'Sullivan B, Sobin LH. Prognostic factors in
prognostic factors available to gastrointestinal pathologists and cancer. 3rd ed. New York, NY, USA: Wiley; 2006. p. 267-79.
CRC patients is lost in translation. [17] Jass JR, O'Brien J, Riddell RH, Snover DC, Association of Directors
To attain this goal, specific questions will need to be of Anatomic and Surgical Pathology. Recommendations for the
reporting of surgically resected specimens of colorectal carcinoma:
addressed: first and foremost, an international, evidence-based
Association of Directors of Anatomic and Surgical Pathology. Am J
consensus on standardized reporting of tumor budding in CRC Clin Pathol 2008;129:13-23.
has to be found. In particular, decisions on the optimal scoring [18] Vieth M, Quirke P, Lambert R, von Karsa L, Risio M, International
method and whether cutoffs or continuous budding counts Agency for Research on Cancer. European guidelines for quality
should be included in histopathology reports have to be made. assurance in colorectal cancer screening and diagnosis. First edition—
annotations of colorectal lesions. Endoscopy 2012;44(Suppl 3):SE131-9.
Second, the normalization of tumor budding scores to visual
[19] Schmoll HJ, Van Cutsem E, Stein A, et al. ESMO consensus guidelines
field size needs to be systematically pursued for application in for management of patients with colon and rectal cancer. a personalized
diagnostic practice. Last, the inclusion of tumor budding into approach to clinical decision making. Ann Oncol 2012;23:2479-516.
clinical trials should be pursued to confirm the influence on [20] Watanabe T, Itabashi M, Shimada Y, et al. Japanese Society for
prognosis in the prospective setting. Cancer of the Colon and Rectum (JSCCR) guidelines 2010 for the
treatment of colorectal cancer. Int J Clin Oncol 2012;17:1-29.
[21] van de Velde CJ, Boelens PG, Borras JM, et al. EURECCA
References colorectal: multidisciplinary management: European consensus
conference colon & rectum. Eur J Cancer 2014;50:1.e1-1.e34.
[1] Prall F. Tumour budding in colorectal carcinoma. Histopathology [22] Compton C, Tanabe K, Savarese D. UptoDate: pathology and
2007;50:151-62. prognostic determinants of colorectal cancer; 2015.
[2] Koelzer VH, Langer R, Zlobec I, Lugli A. Tumor budding in upper [23] De Craene B, Berx G. Regulatory networks defining EMT during
gastrointestinal carcinomas. Front Oncol 2014;4:216. cancer initiation and progression. Nat Rev Cancer 2013;13:97-110.
[3] Karamitopoulou E, Zlobec I, Born D, et al. Tumour budding is a [24] Beaton C, Twine CP, Williams GL, Radcliffe AG. Systematic review and
strong and independent prognostic factor in pancreatic cancer. Eur J meta-analysis of histopathological factors influencing the risk of lymph
Cancer 2013;49:1032-9. node metastasis in early colorectal cancer. Colorectal Dis 2013;15:788-97.
14 V. H. Koelzer et al.

[25] Coverlizza S, Risio M, Ferrari A, Fenoglio-Preiser CM, Rossini FP. of the Japanese Society for Cancer of the Colon and Rectum. J Clin
Colorectal adenomas containing invasive carcinoma. Pathologic assess- Pathol 2013;66:551-8.
ment of lymph node metastatic potential. Cancer 1989;64:1937-47. [45] Choi HJ, Park KJ, Shin JS, Roh MS, Kwon HC, Lee HS. Tumor
[26] Ueno H, Hase K, Hashiguchi Y, et al. Novel risk factors for lymph budding as a prognostic marker in stage-III rectal carcinoma. Int J
node metastasis in early invasive colorectal cancer: a multi-institution Color Dis 2007;22:863-8.
pathology review. J Gastroenterol 2014;49:1314-23. [46] Ueno H, Murphy J, Jass JR, Mochizuki H, Talbot IC. Tumour
[27] Bosch SL, Teerenstra S, de Wilt JH, Cunningham C, Nagtegaal ID. ‘budding’ as an index to estimate the potential of aggressiveness in
Predicting lymph node metastasis in pT1 colorectal cancer: a rectal cancer. Histopathology 2002;40:127-32.
systematic review of risk factors providing rationale for therapy [47] Suzuki A, Togashi K, Nokubi M, et al. Evaluation of venous invasion
decisions. Endoscopy 2013;45:827-34. by Elastica van Gieson stain and tumor budding predicts local and
[28] Rubin P, Hansen JT, Rubin P. TNM staging atlas with oncoanatomy. distant metastases in patients with T1 stage colorectal cancer. Am J
Philadelphia: Wolters Kluwer Health/Lippincott Williams & Wilkins; Surg Pathol 2009;33:1601-7.
2012. [48] Okuyama T, Nakamura T, Yamaguchi M. Budding is useful to select
[29] Benson III AB, Schrag D, Somerfield MR, et al. American Society of high-risk patients in stage II well-differentiated or moderately
Clinical Oncology recommendations on adjuvant chemotherapy for differentiated colon adenocarcinoma. Dis Colon Rectum 2003;46:
stage II colon cancer. J Clin Oncol 2004;22:3408-19. 1400-6.
[30] Gill S, Loprinzi CL, Sargent DJ, et al. Pooled analysis of fluorouracil- [49] Prall F, Nizze H, Barten M. Tumour budding as prognostic factor in
based adjuvant therapy for stage II and III colon cancer: who benefits stage I/II colorectal carcinoma. Histopathology 2005;47:17-24.
and by how much? J Clin Oncol 2004;22:1797-806. [50] Karamitopoulou E, Zlobec I, Kölzer V, et al. Proposal for a 10-high-
[31] National Comprehensive Cancer Network (NCCN). NCCN Clinical power-fields scoring method for the assessment of tumor budding in
Practice Guidelines in Oncology: Colon Cancer. Version 2. The colorectal cancer. Mod Pathol 2013;26:295-301.
National Comprehensive Cancer Network website; 2015 [Updated [51] Zlobec I, Molinari F, Martin V, et al. Tumor budding predicts
March 2014. Accessed March 12th, 2015]. response to anti-EGFR therapies in metastatic colorectal cancer
[32] Lai YH, Wu LC, Li PS, et al. Tumour budding is a reproducible index patients. World J Gastroenterol 2010;16:4823-31.
for risk stratification of patients with stage II colon cancer. Colorectal [52] Hase K, Shatney C, Johnson D, Trollope M, Vierra M. Prognostic
Dis 2014;16:259-64. value of tumor “budding” in patients with colorectal cancer. Dis
[33] Tanaka M, Hashiguchi Y, Ueno H, Hase K, Mochizuki H. Tumor Colon Rectum 1993;36:627-35.
budding at the invasive margin can predict patients at high risk of [53] Nakamura T, Mitomi H, Kikuchi S, Ohtani Y, Sato K. Evaluation of
recurrence after curative surgery for stage II, T3 colon cancer. Dis the usefulness of tumor budding on the prediction of metastasis to the
Colon Rectum 2003;46:1054-9. lung and liver after curative excision of colorectal cancer. Hepato-
[34] Wang LM, Kevans D, Mulcahy H, et al. Tumor budding is a strong Gastroenterology 2005;52:1432-5.
and reproducible prognostic marker in T3N0 colorectal cancer. Am J [54] Ueno H, Mochizuki H, Shinto E, Hashiguchi Y, Hase K, Talbot IC.
Surg Pathol 2009;33:134-41. Histologic indices in biopsy specimens for estimating the probability
[35] Betge J, Kornprat P, Pollheimer MJ, et al. Tumor budding is an of extended local spread in patients with rectal carcinoma. Cancer
independent predictor of outcome in AJCC/UICC stage II colorectal 2002;94:2882-91.
cancer. Ann Surg Oncol 2012;19:3706-12. [55] Ueno H, Price AB, Wilkinson KH, Jass JR, Mochizuki H, Talbot IC.
[36] Canney AL, Kevans D, Wang LM, et al. Stage II colonic A new prognostic staging system for rectal cancer. Ann Surg 2004;
adenocarcinoma: a detailed study of pT4N0 with emphasis on 240:832-9.
peritoneal involvement and the role of tumour budding. Histopathology [56] Puppa G, Senore C, Sheahan K, et al. Diagnostic reproducibility of
2012;61:488-96. tumour budding in colorectal cancer: a multicentre, multinational
[37] Horcic M, Koelzer VH, Karamitopoulou E, et al. Tumor budding study using virtual microscopy. Histopathology 2012;61:562-75.
score based on 10 high-power fields is a promising basis for a [57] Lugli A, Karamitopoulou E, Panayiotides I, et al. CD8+ lymphocytes/
standardized prognostic scoring system in stage II colorectal cancer. tumour-budding index: an independent prognostic factor representing
HUM PATHOL 2013;44:697-705. a ‘pro-/anti-tumour’ approach to tumour host interaction in colorectal
[38] Koelzer VH, Lugli A, Dawson H, et al. CD8/CD45RO T-cell cancer. Br J Cancer 2009;101:1382-92.
infiltration in endoscopic biopsies of colorectal cancer predicts nodal [58] Koelzer VH, Lugli A. The tumor border configuration of colorectal cancer
metastasis and survival. J Transl Med 2014;12:81. as a histomorphological prognostic indicator. Front Oncol 2014;4:29.
[39] Anitei MG, Zeitoun G, Mlecnik B, et al. Prognostic and predictive [59] Ohtsuki K, Koyama F, Tamura T, et al. Prognostic value of
values of the immunoscore in patients with rectal cancer. Clin Cancer immunohistochemical analysis of tumor budding in colorectal
Res 2014;20:1891-9. carcinoma. Anticancer Res 2008;28:1831-6.
[40] Giger OT, Comtesse SC, Lugli A, Zlobec I, Kurrer MO. Intra-tumoral [60] Rubio CA. Arrest of cell proliferation in budding tumor cells ahead of
budding in preoperative biopsy specimens predicts lymph node and the invading edge of colonic carcinomas. A preliminary report.
distant metastasis in patients with colorectal cancer. Mod Pathol Anticancer Res 2008;28:2417-20.
2012;25:1048-53. [61] Caie PD, Turnbull AK, Farrington SM, Oniscu A, Harrison DJ.
[41] Koelzer VH, Zlobec I, Berger MD, et al. Tumor budding in colorectal Quantification of tumour budding, lymphatic vessel density and invasion
cancer revisited: results of a multicenter interobserver study. through image analysis in colorectal cancer. J Transl Med 2014;12:156.
Virchows Arch 2015;466:485-93. [62] Lugli A, Karamitopoulou E, Zlobec I. Tumour budding: a promising
[42] Thomas GD, Dixon MF, Smeeton NC, Williams NS. Observer parameter in colorectal cancer. Br J Cancer 2012;106:1713-7.
variation in the histological grading of rectal carcinoma. J Clin Pathol [63] Ueno H, Mochizuki H, Hashiguchi Y, et al. Risk factors for an adverse
1983;36:385-91. outcome in early invasive colorectal carcinoma. Gastroenterology 2004;
[43] Chandler I, Houlston RS. Interobserver agreement in grading of 127:385-94.
colorectal cancers—findings from a nationwide Web-based survey of [64] Bosman FT, World Health Organization. International Agency for
histopathologists. Histopathology 2008;52:494-9. Research on Cancer. WHO classification of tumours of the digestive
[44] Kojima M, Shimazaki H, Iwaya K, et al. Pathological diagnostic system. Lyon: International Agency for Research on Cancer; 2010.
criterion of blood and lymphatic vessel invasion in colorectal cancer: [65] Prall F, Weirich V, Ostwald C. Phenotypes of invasion in sporadic
a framework for developing an objective pathological diagnostic colorectal carcinomas related to aberrations of the adenomatous
system using the Delphi method, from the Pathology Working Group polyposis coli (APC) gene. Histopathology 2007;50:318-30.
Tumor budding in colorectal cancer 15

[66] Garcia-Solano J, Conesa-Zamora P, Trujillo-Santos J, Torres-Moreno D, metastasis in submucosal colorectal cancer detected by anticytokeratin
Makinen MJ, Perez-Guillermo M. Immunohistochemical expression antibody CAM5.2. Br J Cancer 2006;94:293-8.
profile of beta-catenin, E-cadherin, P-cadherin, laminin-5gamma2 chain, [86] Masaki T, Matsuoka H, Sugiyama M, Abe N, Sakamoto A, Atomi Y. Actual
and SMAD4 in colorectal serrated adenocarcinoma. HUM PATHOL 2012; number of tumor budding as a new tool for the individualization of treatment
43:1094-102. of T1 colorectal carcinomas. J Gastroenterol Hepatol 2006;21:1115-21.
[67] Brabletz T, Hlubek F, Spaderna S, et al. Invasion and metastasis in [87] Ono M, Sakamoto M, Ino Y, et al. Cancer cell morphology at the
colorectal cancer: epithelial-mesenchymal transition, mesenchymal- invasive front and expression of cell adhesion-related carbohydrate in
epithelial transition, stem cells and beta-catenin. Cells Tissues Organs the primary lesion of patients with colorectal carcinoma with liver
2005;179:56-65. metastasis. Cancer 1996;78:1179-86.
[68] Karamitopoulou E, Lugli A, Panayiotides I, et al. Systematic assessment [88] Kawaura K, Fujii S, Murata Y, et al. The lymphatic infiltration
of protein phenotypes characterizing high-grade tumour budding in identified by D2-40 monoclonal antibody predicts lymph node
mismatch repair-proficient colorectal cancer. Histopathology 2010;57: metastasis in submucosal invasive colorectal cancer. Pathobiology
233-43. 2007;74:328-35.
[69] Jass JR, Barker M, Fraser L, et al. APC mutation and tumour budding [89] Kaneko I, Tanaka S, Oka S, et al. Immunohistochemical molecular
in colorectal cancer. J Clin Pathol 2003;56:69-73. markers as predictors of curability of endoscopically resected
[70] Guzinska-Ustymowicz K, Kemona A. Transforming growth factor submucosal colorectal cancer. World J Gastroenterol 2007;13:3829-35.
beta can be a parameter of aggressiveness of pT1 colorectal cancer. [90] Sohn DK, Chang HJ, Park JW, et al. Histopathological risk factors for
World J Gastroenterol 2005;11:1193-5. lymph node metastasis in submucosal invasive colorectal carcinoma of
[71] Bhowmick NA, Ghiassi M, Bakin A, et al. Transforming growth factor- pedunculated or semipedunculated type. J Clin Pathol 2007;60:912-5.
beta1 mediates epithelial to mesenchymal transdifferentiation through a [91] Yasuda K, Inomata M, Shiromizu A, Shiraishi N, Higashi H, Kitano S.
RhoA-dependent mechanism. Mol Biol Cell 2001;12:27-36. Risk factors for occult lymph node metastasis of colorectal cancer
[72] Dawson H, Lugli A. Molecular and pathogenetic aspects of tumor invading the submucosa and indications for endoscopic mucosal
budding in colorectal cancer. Front Med (Lausanne) 2015;2:11. resection. Dis Colon Rectum 2007;50:1370-6.
[73] Prall F, Ostwald C. High-degree tumor budding and podia-formation [92] Ishikawa Y, Akishima-Fukasawa Y, Ito K, et al. Histopathologic
in sporadic colorectal carcinomas with K-ras gene mutations. HUM determinants of regional lymph node metastasis in early colorectal
PATHOL 2007;38:1696-702. cancer. Cancer 2008;112:924-33.
[74] Zhao Q, Yang S, Ponchiardi C, O'Brien M. BRAF/KRAS mutation, [93] Yamauchi H, Togashi K, Kawamura YJ, et al. Pathological predictors
MSI/MSS status and tumor budding in colorectal carcinoma. Lab for lymph node metastasis in T1 colorectal cancer. Surg Today 2008;
Invest 2007;95(Suppl S1):201A-2A. 38:905-10.
[75] Guzinska-Ustymowicz K. MMP-9 and cathepsin B expression in [94] Choi DH, Sohn DK, Chang HJ, Lim SB, Choi HS, Jeong SY.
tumor budding as an indicator of a more aggressive phenotype of Indications for subsequent surgery after endoscopic resection of
colorectal cancer (CRC). Anticancer Res 2006;26:1589-94. submucosally invasive colorectal carcinomas: a prospective cohort
[76] Dawson H, Koelzer VH, Karamitopoulou E, et al. The apoptotic and study. Dis Colon Rectum 2009;52:438-45.
proliferation rate of tumour budding cells in colorectal cancer outlines [95] Ogawa T, Yoshida T, Tsuruta T, et al. Tumor budding is predictive of
a heterogeneous population of cells with various impacts on clinical lymphatic involvement and lymph node metastases in submucosal
outcome. Histopathology 2014;64:577-84. invasive colorectal adenocarcinomas and in non-polypoid compared
[77] Koelzer VH, Karamitopoulou E, Dawson H, Kondi-Pafiti A, Zlobec I, with polypoid growths. Scand J Gastroenterol 2009;44:605-14.
Lugli A. Geographic analysis of RKIP expression and its clinical [96] Kajiwara Y, Ueno H, Hashiguchi Y, Mochizuki H, Hase K. Risk
relevance in colorectal cancer. Br J Cancer 2013;108:2088-96. factors of nodal involvement in T2 colorectal cancer. Dis Colon
[78] Sannier A, Lefevre JH, Panis Y, Cazals-Hatem D, Bedossa P, Guedj N. Rectum 2010;53:1393-9.
Pathological prognostic factors in locally advanced rectal carcinoma [97] Komori K, Hirai T, Kanemitsu Y, et al. Is “depth of submucosal
after neoadjuvant radiochemotherapy: analysis of 113 cases. Histopa- invasion N or = 1,000 microm” an important predictive factor for
thology 2014;65:623-30. lymph node metastases in early invasive colorectal cancer (pT1)?
[79] Koelzer VH, Dawson H, Andersson E, et al. Active immunosurveillance Hepato-Gastroenterology 2010;57:1123-7.
in the tumor microenvironment of colorectal cancer is associated with low [98] Tateishi Y, Nakanishi Y, Taniguchi H, Shimoda T, Umemura S.
frequency tumor budding and improved outcome. Transl Res 2015;166: Pathological prognostic factors predicting lymph node metastasis in
207-17. submucosal invasive (T1) colorectal carcinoma. Mod Pathol 2010;23:
[80] Okuyama T, Oya M, Ishikawa H. Budding as a risk factor for lymph 1068-72.
node metastasis in pT1 or pT2 well-differentiated colorectal [99] Ueno H, Hashiguchi Y, Kajiwara Y, et al. Proposed objective criteria
adenocarcinoma. Dis Colon Rectum 2002;45:628-34. for “grade 3” in early invasive colorectal cancer. Am J Clin Pathol
[81] Morodomi T, Isomoto H, Shirouzu K, Kakegawa K, Irie K, 2010;134:312-22.
Morimatsu M. An index for estimating the probability of lymph [100] Akishima-Fukasawa Y, Ishikawa Y, Akasaka Y, et al. Histopatho-
node metastasis in rectal cancers. Lymph node metastasis and the logical predictors of regional lymph node metastasis at the invasive
histopathology of actively invasive regions of cancer. Cancer 1989; front in early colorectal cancer. Histopathology 2011;59:470-81.
63:539-43. [101] Keum MA, Lim SB, Kim SA, et al. Clinicopathologic factors
[82] Egashira Y, Yoshida T, Hirata I, et al. Analysis of pathological risk affecting recurrence after curative surgery for stage I colorectal
factors for lymph node metastasis of submucosal invasive colon cancer. J Korean Soc Coloproctol 2012;28:49-55.
cancer. Mod Pathol 2004;17:503-11. [102] Kye BH, Jung JH, Kim HJ, Kang SG, Cho HM, Kim JG. Tumor budding
[83] Kitajima K, Fujimori T, Fujii S, et al. Correlations between lymph as a risk factor of lymph node metastasis in submucosal invasive T1
node metastasis and depth of submucosal invasion in submucosal colorectal carcinoma: a retrospective study. BMC Surg 2012;12:16.
invasive colorectal carcinoma: a Japanese collaborative study. [103] Nakadoi K, Tanaka S, Kanao H, et al. Management of T1 colorectal
J Gastroenterol 2004;39:534-43. carcinoma with special reference to criteria for curative endoscopic
[84] Wang HS, Liang WY, Lin TC, et al. Curative resection of T1 resection. J Gastroenterol Hepatol 2012;27:1057-62.
colorectal carcinoma: risk of lymph node metastasis and long-term [104] Suh JH, Han KS, Kim BC, et al. Predictors for lymph node metastasis
prognosis. Dis Colon Rectum 2005;48:1182-92. in T1 colorectal cancer. Endoscopy 2012;44:590-5.
[85] Kazama S, Watanabe T, Ajioka Y, Kanazawa T, Nagawa H. Tumour [105] Oka S, Tanaka S, Nakadoi K, Kanao H, Chayama K. Risk analysis of
budding at the deepest invasive margin correlates with lymph node submucosal invasive rectal carcinomas for lymph node metastasis to
16 V. H. Koelzer et al.

expand indication criteria for endoscopic resection. Dig Endosc 2013; submucosal invasive (T1) colorectal cancer. Int J Color Dis
25(Suppl 2):21-5. 2015;30:761-8.
[106] Umemura K, Takagi S, Shimada T, et al. Prognostic and diagnostic [112] Kawachi H, Eishi Y, Ueno H, et al. A three-tier classification system
significance of tumor budding associated with beta-catenin expression in based on the depth of submucosal invasion and budding/sprouting
submucosal invasive colorectal carcinoma. Tohoku J Exp Med 2013; can improve the treatment strategy for T1 colorectal cancer: a
229:53-9. retrospective multicenter study. Mod Pathol 2015;28:872-9.
[107] Ueno H, Hase K, Hashiguchi Y, et al. Novel risk factors for lymph [113] Wada H, Shiozawa M, Katayama K, et al. Systematic review and
node metastasis in early invasive colorectal cancer: a multi-institution meta-analysis of histopathological predictive factors for lymph
pathology review. J Gastroenterol 2014;49:1314-23. node metastasis in T1 colorectal cancer. J Gastroenterol 2015;50:
[108] Suh JP, Youk EG, Lee EJ, et al. Endoscopic submucosal dissection 727-34.
for nonpedunculated submucosal invasive colorectal cancer: is it [114] Okuyama T, Oya M, Ishikawa H. Budding as a useful prognostic marker
feasible? Eur J Gastroenterol Hepatol 2013;25:1051-9. in pT3 well- or moderately-differentiated rectal adenocarcinoma. J Surg
[109] Ryu HS, Kim WH, Ahn S, et al. Combined morphologic and molecular Oncol 2003;83:42-7.
classification for predicting lymph node metastasis in early-stage [115] Masaki T, Matsuoka H, Sugiyama M, et al. Tumor budding and
colorectal adenocarcinoma. Ann Surg Oncol 2014;21:1809-16. evidence-based treatment of T2 rectal carcinomas. J Surg Oncol
[110] Caputo D, Caricato M, La Vaccara V, Taffon C, Capolupo GT, 2005;92:59-63.
Coppola R. T1 colorectal cancer: poor histological grading is [116] Kevans D, Wang LM, Sheahan K, et al. Epithelial-mesenchymal
predictive of lymph-node metastases. Int J Surg 2014;12:209-12. transition (EMT) protein expression in a cohort of stage II colorectal
[111] Macias-Garcia F, Celeiro-Munoz C, Lesquereux-Martinez L, et al. cancer patients with characterized tumor budding and mismatch
A clinical model for predicting lymph node metastasis in repair protein status. Int J Surg Pathol 2011;19:751-60.

You might also like