You are on page 1of 11

Modern Pathology (2012) 25, 1315–1325

& 2012 USCAP, Inc. All rights reserved 0893-3952/12 $32.00 1315

Tumor budding in colorectal carcinoma:


time to take notice
Bojana Mitrovic, David F Schaeffer, Robert H Riddell and Richard Kirsch

Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada

Tumor ‘budding’, loosely defined by the presence of individual cells and small clusters of tumor cells at the
invasive front of carcinomas, has received much recent attention, particularly in the setting of colorectal
carcinoma. It has been postulated to represent an epithelial–mesenchymal transition. Tumor budding is a well-
established independent adverse prognostic factor in colorectal carcinoma that may allow for stratification of
patients into risk categories more meaningful than those defined by TNM staging, and also potentially guide
treatment decisions, especially in T1 and T3 N0 (Stage II, Dukes’ B) colorectal carcinoma. Unfortunately, its
universal acceptance as a reportable factor has been held back by a lack of definitional uniformity with respect
to both qualitative and quantitative aspects of tumor budding. The purpose of this review is fourfold: (1) to
describe the morphology of tumor budding and its relationship to other potentially important features of the
invasive front; (2) to summarize current knowledge regarding the prognostic significance and potential clinical
implications of this histomorphological feature; (3) to highlight the challenges posed by a lack of data to allow
standardization with respect to the qualitative and quantitative criteria used to define budding; and (4) to
present a practical approach to the assessment of tumor budding in everyday practice.
Modern Pathology (2012) 25, 1315–1325; doi:10.1038/modpathol.2012.94; published online 13 July 2012

Keywords: colorectal carcinoma; epithelial–mesenchymal transition; prognostic factors; tumor budding

Colorectal carcinoma is one of the commonest larly in node-negative disease. Thus, its assessment
human cancers, and one of the leading causes has the potential to enhance prognostic accuracy
of cancer-related death.1 Prognosis and treatment and influence treatment algorithms. When exam-
decisions are based primarily on the extent of the ined carefully, the majority of colorectal carcinomas
disease, as codified by the TNM staging system. display some degree of budding; hence, attempts
Unfortunately, a substantial number of tumors have been made at developing scoring systems
behave poorly despite being categorized as low risk to identify a prognostically significant degree of
based on their TNM stage.2 Thus, the search for budding, commonly termed ‘high-grade’ budding.
additional prognostic factors in the assessment of Definitions of high-grade budding vary substantially
colorectal carcinoma has been a major research among different authors and even among different
focus. Of the histopathological factors studied to studies by the same authors.
date, the most promising include extramural venous Although tumor budding only entered the main-
invasion, the nature of the advancing front (pushing stream pathology literature in the past decade, it
vs infiltrative), an inflammatory infiltrate, micro- was first described in the 1950s by Imai,3 who
satellite instability, and tumor budding—the pre- postulated that the presence of ‘sprouting’ at the
sence of small discrete clusters of tumor cells at the invasive edge of carcinomas reflected a more rapid
invasive edge (Figure 1). There is now overwhelm- tumor growth rate (or ‘Schub’ phase of growth).
ing evidence that tumor budding is an independent Budding has been described in association with
prognostic factor in colorectal carcinoma, particu- carcinomas at a number of different sites, but it
has been most extensively studied in colorectal
carcinoma.
Correspondence: Dr B Mitrovic, MD, Department of Pathology The biology of tumor budding is not understood
and Laboratory Medicine, Mount Sinai Hospital, 600 University but the prevailing theory is that at least some types
Avenue, Toronto, Ontario, Canada M5G 1X5.
E-mail: BMitrovic@mtsinai.on.ca
of budding represent an example of epithelial–
Received 15 December 2011; revised 30 March 2012; accepted 1 mesenchymal transition, a process thought to occur
April 2012; published online 13 July 2012 physiologically during embryological development,

www.modernpathology.org
Tumor budding
1316 B Mitrovic et al

Figure 1 Tumor budding in colorectal carcinoma (a) Illustration of a colorectal carcinoma with an infiltrative border but no budding.
Tumor buds, defined as individual cells or small clusters (o5 cells) of tumor cells at the invasive front (arrows), are illustrated in these
examples from (b) a malignant polyp and (c and d) a colorectal cancer resection specimen. Note the blurring of the tumor–stroma
interface (c), which corresponds with tumor budding at higher magnification (d). (Original magnification: a—  100; b—  200; c—  20;
d—  400).

and pathologically during fibrosis and tumor inva- Finally, we will present a practical approach
sion.4,5 The epithelial–mesenchymal transition is to the assessment of tumor budding. Although
characterized by loss of cell adhesion molecules, performing a tumor bud count is easy in theory,
cytoskeletal alterations, increased production of deciding what is or is not a bud can prove
extracellular matrix components, resistance to apop- surprisingly difficult in practice. Buds are often
tosis, and ability to degrade basement membrane, obscured in the setting of a prominent inflammatory
resulting in a phenotype with increased migratory reaction; fibroblasts and stromal cells may occasion-
capacity and invasiveness.6,7 Current theories ally be mistaken for buds; and single-cell buds are
relating to the epithelial–mesenchymal transition usually inconspicuous and often missed on H&E
and tumor budding have been comprehensively evaluation (Figure 2).
reviewed elsewhere, most recently by Zlobec et al,8
and will not be discussed here.
The purpose of this review is to examine Morphological features
practical issues related to tumor budding that
may be of interest to the practicing pathologist. Although tumor budding (‘sprouting’) was origin-
We will discuss the morphological features ally described by Imai in the 1950s, the first detailed
and prognostic significance of tumor budding description of tumor buds in the English language
and summarize the various scoring systems for its literature was put forth by Gabbert et al9 who used
assessment in order to highlight problems related light and electron microscopy to characterize what
to definitional heterogeneity in tumor-budding they termed ‘tumor dedifferentiation’ at the invasive
studies. edge of colorectal carcinomas. They identified a

Modern Pathology (2012) 25, 1315–1325


Tumor budding
B Mitrovic et al 1317

Figure 2 ‘Challenging scenarios’. (a) Peritumoral inflammatory cells, including histiocytes, can be difficult to differentiate from tumor
buds, and may sometimes obscure the underlying budding. (b) Immunohistochemistry for anti-cytokeratin may help to highlight tumor
buds in this setting. (c) Given that tumor budding may represent an epithelial–mesenchymal transformation, their separation from
stromal cells can also be challenging and (d) anti-cytokeratin stains may be very useful in identifying tumor buds. (e) Fragmentation of
neoplastic glandular structures (arrow) can sometimes give the impression of extensive tumor budding on H&E and on
immunohistochemistry with (f) anti-cytokeratin. In this example, the small fragments/individual tumor cells highlighted by anti-
cytokeratin in the region of the fragmented gland (arrow between fragments) may be a consequence of fragmentation rather than true
budding. (Original magnification: a—  40; b—  40; c—  100; d—  100; e—  200; f—  200).

subset of colorectal carcinomas with invasive with well-developed glandular structures giving
fronts that displayed a strikingly different architec- way to isolated tumor cells and small cell groups,
ture compared with the central area of the tumor, as illustrated in Figure 1. At the ultrastructural level,

Modern Pathology (2012) 25, 1315–1325


Tumor budding
1318 B Mitrovic et al

these small clusters and isolated tumor cells invasion: Kazama et al22 found no relationship
appeared poorly differentiated, with large nuclei between budding and vascular invasion, whereas
and a loss of microvilli and basement membrane, three other studies have reported a statistically
and poorly developed or absent junctional com- significant correlation between budding and venous
plexes and desmosomes. invasion, though the association was not as pro-
When seen in a single section, tumor buds appear nounced as the relationship with lymphatic inva-
as clusters of cells that have broken off from the sion and lymph node metastases.11,12,32
main tumor mass. By examining serial sections of The tumor–host interaction at the invasive front
high budding tumors stained with anti-cytokeratin may be of prognostic importance in the setting of
antibodies, Prall et al10 demonstrated that most buds tumor budding. Several studies have shown peri-
that appear to represent discrete clusters of cells are tumoral lymphocytic infiltration to be an indepen-
in fact connected to adjacent larger glands. Indeed, dent prognostic factor in colorectal carcinoma.
Morodomi et al11 coined the term ‘budding’ because Lugli et al23 have demonstrated that a peritumoral
the undifferentiated single cells and tubular tumor lymphocytic reaction is associated with improved
nests that they counted as buds both appeared to be prognosis in the setting of tumor budding, suggest-
budding from larger neoplastic glands. ing that the immune response might target the tumor
Shinto et al12,13 used immunohistochemistry with buds. The nature of the stroma at the invasive
anti-cytokeratin antibodies to highlight another margin may provide further prognostic information,
feature of buds, termed cytoplasmic pseudofragmen- with myxoid stroma being associated with worse
tation, whereby cytoplasmic fragments, visible only prognosis,20,39 but further studies are needed to
by immunohistochemistry, are seen in the immedi- confirm the reproducibility and prognostic impor-
ate vicinity of tumor buds. When examined on serial tance of these findings.
sections, some of the fragments were shown to be
connected to the buds (hence the term ‘pseudofrag-
ments’). The presence of pseudofragments is asso- Clinical significance of tumor budding
ciated with high-grade budding, but was shown to
Tumor budding is associated with other histopatho-
be an independent prognostic factor on multivariate
logical factors known to portend a worse prognosis,
analysis, suggesting that its presence signifies an
namely higher tumor grade, infiltrating tumor border,
aggressive budding phenotype.
the presence of lymphovascular and perineural
Although tumor buds are usually most prominent
invasion, and lymph node metastases.11,12,15–32 On
at the invasive front, intratumoral budding has been
multivariate analysis, budding has consistently
described as well. Only one study has attempted to
emerged as an independent adverse prognostic factor,
assess the significance of this finding: Lugli et al14
associated with local tumor recurrence and distant
reported that the presence of intratumoral budding
metastases, and significantly worse overall and
was strongly correlated with peritumoral budding,
disease-free survival.12,15,16,21,27,28,31,32,34–36,38,40,41
but was found to be an independent prognostic
The adverse prognostic impact of high-grade
factor on multivariate analysis.
tumor budding is seen in both early and advanced
There is a strong association between budding and
colorectal carcinoma, and there are several scenarios
the presence of lymph node metastases and lym-
in which this feature might influence clinical
phovascular invasion,11,12,15–32 defined by the pre-
decision making, particularly in early colorectal
sence of tumor cells within an endothelium-lined
carcinoma.
space, and it has been suggested that buds represent
the part of the tumor that has gained the ability to
invade lymphatics and vascular channels. This idea Tumor Budding in Early Colorectal Carcinoma
is supported by two intriguing morphological
studies: Morodomi et al33 examined serial sections Submucosally invasive colorectal carcinoma is
of high-budding areas to demonstrate that budding associated with excellent outcomes and low rates
nests are often found adjacent to areas of lympho- of lymph node metastases, and a subset of patients
vascular space invasion, and, in a more recent can be successfully managed with endoscopic
study, Ohtsuki et al31 performed double staining mucosal resection or polypectomy,42 thus avoiding
for anti-cytokeratin antibodies and anti-lymphatic the risks associated with segmental resection.
antibodies, finding that a number of ‘buds’ at the Identification of candidates for endoscopic resection
invasive edge of a tumor are in fact located in small rests mainly on the absence of certain high-risk
lymphatic spaces. Similarly, the presence of bud- histopathological features, namely high tumor
ding has been associated with increased risk of grade, lymphovascular invasion, and tumor bud-
distant metastases,34–36 suggesting that budding may ding.26,42 Tumor extending to the cauterized margin
also be associated with vascular invasion. A few can often be managed by endoscopic follow up.
tumor-budding studies have used vascular mar- Several large studies have shown tumor budding
kers and/or elastic stains to assess vascular inva- in submucosally invasive colorectal carcinoma to
sion,11,12,19,22,26,32,37,38 but only four have analyzed be an independent prognostic factor associated
the relationship between budding and vascular with lymph node metastases, local recurrence, and

Modern Pathology (2012) 25, 1315–1325


Tumor budding
B Mitrovic et al 1319

cancer-related death.16,19,22,25,26,29,30,37,43,44 In the the costs and side effects of chemotherapy in all
largest of these studies, Ueno et al26 demonstrated patients.46 The significantly worse outcome experi-
that, in submucosally invasive colorectal carcinoma, enced by Stage II patients with tumor budding
high tumor grade, lymphovascular invasion, and has prompted some authors to suggest that adju-
tumor budding are the three factors independently vant chemotherapy should be considered in these
associated with lymph node metastases. Patients patients,18,34–36,41 but there are no published data
without any of these three features showed excep- assessing the effectiveness of chemotherapy in Stage
tionally low rates of lymph node metastases (1%, II colorectal carcinoma with tumor budding.
1/138); in the presence of one risk factor, the rate of
nodal metastases increased substantially to 21%
(12/58), and when two or three factors were present, Tumor Budding in Stage III (T1–4 N þ ) Colorectal
the risk was 36% (20/55), suggesting that in the Carcinoma
absence of these factors, polypectomy alone is Data regarding budding in Stage III colorectal
sufficient treatment for early colorectal carcinoma carcinoma are limited, and the two studies reporting
provided the resection margins are clear. In this on the prognostic significance of budding in this
study, Ueno et al further showed that in the absence subgroup of patients have produced conflicting
of extensive submucosal invasion (defined as results. Choi et al15 studied tumor budding in 103
r4 mm wide and r2 mm deep), the risk of nodal patients with Stage III rectal carcinoma; budding
metastases (including isolated tumor cells, identifi- was an independent poor prognostic factor: when
able only with anti-cytokeratin antibodies) in added to N stage as a prognostic variable, there was
tumors lacking the three above-mentioned features better prognostic stratification with respect to 5-year
was 0; thus, extent of submucosal invasion may disease-free survival compared with the Americal
need to be included in the histopathological assess- Joint Comittee on Cancer nodal staging alone. On the
ment of T1 tumors if strict criteria are to be applied other hand, in a series of 477 Stage III colorectal
for the identification of patients that can safely be carcinomas reported on by Sy et al,47 although there
managed without surgical resection. was a survival difference between high- and low-
budding groups, on multivariate analysis budding
was not found to be an independent prognostic
Tumor Budding in Stage II (T3–4 N0) Colorectal factor. Although more studies are needed to define
Carcinoma the prognostic significance of tumor budding in this
group, it is not likely that the reporting of this factor
As patients with Stage II colorectal carcinoma will impact the clinical management of these
have highly variable outcomes, tumor budding patients.
may be particularly useful in identifying high-risk
subgroups within this population. In one of the
earlier studies of tumor budding, Hase et al18 Tumor Budding in Metastatic Colorectal Carcinoma
demonstrated that 5-year survival rates of Dukes B
(stage II, T3–4 N0) patients with high-grade budding The significance of tumor budding in metastatic
are significantly worse than those of Dukes C (N þ ) colorectal carcinoma has received little attention.
patients without budding (29 percent vs 66 percent; However, one study showed that the presence of
Po0.001). A number of more recent studies have tumor budding predicts poor response to anti-EGFR
confirmed that patients with Stage II colorectal therapy in patients with metastatic colorectal carci-
carcinoma do significantly worse when high-grade noma: Zlobec et al48 studied a series of 43 patients
budding is present,34–36,38,41,45 and several studies with metastatic colorectal carcinoma treated with
have shown that survival rates of patients with Stage cetuximab or penitumumab, and found all patients
II colorectal carcinoma with high-grade budding are with a KRAS mutation (n ¼ 7) and/or high-grade
equivalent to survival rates of patients with Stage III tumor budding (n ¼ 11, including 4 with KRAS
colorectal carcinoma.34–36 In their studies of Stage II mutation) to be nonresponsive to anti-EGFR therapy.
and III pT3 tumors, Okuyama et al34,35 found that Although this finding needs to be confirmed in
tumor budding was the only factor on multivariate larger cohorts, it raises the interesting possibility
analysis to be associated with decreased survival that tumor budding in addition to K-RAS analysis
and was more prognostically significant than lymph may more accurately determine eligibility for anti-
node metastases. EGFR therapy.
Currently, the indications for chemotherapy in
Stage II colorectal carcinoma are unclear, and adju- Tumor Budding in Microsatellite Unstable Tumors
vant therapy is considered only in subgroups with
adverse prognostic features, eg, high-histological The association between tumor budding and poor
grade, pT4 disease, and extramural venous invasion. prognosis in sporadic microsatellite stable colorectal
The QUASAR trial demonstrated that chemotherapy carcinoma is well established, but whether this can
does result in increased survival in Stage II patients, be extrapolated to sporadic and Lynch syndrome-
but the improvement was small and may not justify related high frequency microsatellite unstable

Modern Pathology (2012) 25, 1315–1325


Tumor budding
1320 B Mitrovic et al

colorectal carcinoma is unclear. Many studies of calculating an average bud count,11 counting the
tumor budding have excluded Lynch syndrome- number of buds along the entire invasive
related cases, while others have not attempted to front19,24,52,53 or calculating the proportion of inva-
analyze the groups separately. Only a few studies sive front with tumor budding.21,36,40 Such metho-
have compared the frequency of tumor budding in dological heterogeneity is illustrated in Table 1,
these different types of colorectal carcinomas: in which summarizes the bud counting methods used
these studies, the frequency of tumor budding was in a sample of tumor-budding studies.
approximately 50% in sporadic microsatellite stable
and low-frequency microsatellite unstable tumors;
lower rates of tumor budding were seen in Lynch Establishing a Prognostically Significant Cutoff for
syndrome-associated tumors (B20%); whereas ‘High-Grade’ Budding
tumor budding was found to be virtually absent
Although Ueno’s definition of ‘high-grade’ budding
in sporadic high-frequency microsatellite unstable
(ie, 10 buds in a 25  field) is the most widely
colorectal carcinoma.23,49,50 It has been postulated
applied in the literature, there is no evidence that
that the relative infrequency of budding in micro-
this is the optimal cutoff for evaluating budding.
satellite unstable tumors may, at least in part,
Only a handful of studies have attempted to analyze
explain the relatively better prognosis of these
their data so as to generate a clinically useful
tumors.49 Interestingly, compared with mismatch
threshold for the assessment of high-grade budding.
repair-proficient tumors, mismatch repair-deficient
The results and methodologies of these studies are
tumors have been shown to display less cytoplasmic
summarized in Table 2. In addition, Masaki et al24,53
pseudofragmentation in the context of high-grade
have proposed a formula based on the total number
budding,51 and to be associated with less intratu-
of buds counted along the entire invasive margin to
moral budding,14 both of which are associated with a
predict the probability of lymph node metastases
worse prognosis. Given the differing biological
in T1 and T2 colorectal carcinoma: although this
pathways of microsatellite stable, sporadic high-
is an interesting idea, it is not practical for use in
frequency microsatellite unstable and Lynch syn-
everyday practice.
drome-related tumors, it seems likely that the
frequency and clinical significance of tumor budding
will vary significantly among these three groups, but Subjectivity in the Qualitative Assessment of Tumour
awaits confirmation in controlled studies. Budding
In addition to the quantitative issues of inconsistent
Scoring systems cutoffs and differing field diameters, there are a
number of qualitative considerations contributing to
Although a large number of studies have shown subjectivity in defining a tumour bud. Although
tumor budding to be an independent adverse only a few authors have specifically commented on
prognostic factor in colorectal carcinoma, study this problem, the identification of tumor buds can
methodologies have varied widely. Different authors prove very difficult in several circumstances, eg, (1)
have used different scoring systems as well as stromal cells or histiocytes masquerading as buds,
differing definitions as to what constitutes a bud. (2) marked inflammation obscuring buds, (3) diffi-
The most widely cited scoring system, developed by culties in determining whether a small cluster of
Ueno et al,27 defines ‘high-grade budding’ as 410 cells represents a true bud or mechanical fragmenta-
buds composed of fewer than 5 cells in a 25  field tion of a larger gland, etc (Figure 2). Most studies of
(field area ¼ 0.385 mm2). Other authors have defined tumor budding have not provided much detail
buds as being composed of r5 cells,38 while some regarding qualitative criteria used to include or
have not set an upper cell limit and instead count exclude a potential ‘bud’. In addition, there appear
both single cells and bundles of five or more cells as to be discrepancies in the qualitative assessment of
buds, as proposed by Morodomi et al11 and budding, even among studies ostensibly using the
commonly applied in the Japanese literature. This same objective criteria of clusters of o5 cells: eg,
heterogeneity in defining budding is compounded while Jass et al49 counted only ‘discrete clusters’ of
by the use of varying field diameters and cutoffs for cells as buds, Ha et al17 defined buds as clusters
defining ‘high-grade’ budding, especially when a ‘appearing to bud from a larger gland’, suggesting
score is given as #/hpf rather than/mm2. Although that they may have also counted buds that were not
most authors have used Ueno’s definition of ‘high- completely separate from adjacent glands. Although
grade budding’ (410 buds of fewer than 5 cells), such variability may be reduced to some degree by
all have used a 20  objective, rather than the establishing consensus criteria, there will likely
25  objective used by Ueno et al, resulting in remain a subset of cases in which budding will be
significantly greater field areas than that originally difficult to evaluate owing to the resemblance of
reported by Ueno. Furthermore, while the majority buds to surrounding stromal cells and/or the
of studies have evaluated only areas with the concealment of buds in the setting of a prominent
greatest amount of budding, others have proposed peritumoral inflammatory reaction (Figure 2).

Modern Pathology (2012) 25, 1315–1325


Table 1 Summary of scoring systems for tumor budding evaluation in a selected sample of relevant studies

First author Patient population Evaluation Bud definition Cutoff for ‘high-grade’ budding Proportion of cases classified as high
method grade

Morodomi11 40 advanced rectal H&E Isolated cancer cells Count performed at four locations Mildly positive, 37.5%;
carcinomas and bundles of 5cells (1.25 mm2 field area), and average taken Strongly positive, 27.5%
Three categories:
0–4, negative;
4–14, mildly positive;
414, strongly positive

Hase18 663 colorectal carcinomas H&E Not specified N/A: classified according to subjective 26%
(subjective evaluation) impression

Ueno27 638 rectal carcinomas H&E o5 cells 10 buds in 25  field (0.385 mm2) 30.1%

Okuyama35 196 pT3 well to moderately H&E Up to 5 cells N/A: classified according to subjective 43.3%
differentiated colorectal impression
carcinomas

Jass49 95 colorectal carcinomas, H&E Up to 5 cells 5 buds in 40  field (area not specified) 32%
stratified according to MSI
status

Guzinska-Ustymowicz16 24 T1 rectal carcinomas H&E Up to 5 cells Any budding considered positive 35%

B Mitrovic et al
Tumor budding
Ha17 90 well to moderately H&E o5cells 47 buds in 20  field (area not specified) N/A (mean bud count selected as
differentiated rectal Cutoff selected based on mean intensity cutoff such that 50% of cases would
carcinomas of budding (7.5) be classified as ‘high grade’)

Kanazawa22 159 colorectal carcinomas H&E ‘Small cluster’ Divided into three groups based on 29% marked
of cells proportion of invasive front with budding: 37% moderate
0–1/3, mild;
1/3–2/3, moderate;
42/3, marked

Losi44 295 Stage I colorectal Immunohisto- o5 cells 45 buds in 20  field (area not specified) 6%
carcinomas chemistry

Ishikawa43 71 submucosally invasive Immunohisto- o5 cells 44 buds in 40  field (area not specified) 51%
colorectal carcinomas chemistry
Modern Pathology (2012) 25, 1315–1325

Yamauchi29 164 T1 colorectal Immunohisto- o5 cells 5 buds in 20  field (area not specified) 15%
carcinomas chemistry

Wang41 128 pT3 N0 colorectal Immunohisto- o5 cells Budding was counted in 5 fields 45%
carcinomas chemistry (20  0.95 mm2); a median count
of 1 buds considered positive

1321
Tumor budding
1322 B Mitrovic et al

Table 2 Summary of studies proposing evidence-based cutoffs for ‘high-grade’ budding

First Method of Bud Method for determination of optimal cutoff Resulting definition of
author evaluation definition ‘high-grade’ budding

Choi15 H&E o5 cells Patients were divided semiquantitatively into four budding groups 410 buds in 20  field
based on quartiles. The cutoff considered the best discriminator of (area not specified)
disease-free survival was between groups 3 and 4.
Sy47 H&E o5 cells Patients were divided arbitrarily into five budding groups. 49 buds in 20  field
As there were no statistically significant differences among (0.95 mm2)
the first four groups, while the highest budding group showed
significantly worse survival, the cutoff between groups 4 and 5
was deemed the most prognostically significant.
Prall38 Immunohisto- Up to ROC curves were constructed to define the budding cutoff with 25 buds in 25  field
chemistry 5 cells the greatest sensitivity and specificity for predicting the (0.785 mm2)
development of metastases.
Lugli23 Immunohisto- Up to ROC curves were constructed to define the budding cutoff with 16 buds in 40  field
chemistry 5 cells the greatest sensitivity and specificity for discriminating (area not specified)
between survivors and nonsurvivors.
Zlobec48 Immunohisto- o5 cells ROC curves were constructed to define the budding cutoff 414 buds in 40  field
chemistry with the best predictive ability for anti-EGFR therapy (area not specified)
nonresponse in a population of metastatic CRC.

Role of Immunohistochemistry in the Evaluation of suffer from suboptimal interobserver variability.


Budding A few studies on tumor budding have incorporated
an assessment of interobserver variability, with
Some authors have attempted to eliminate the sub- reported kappa values ranging from 0.41 to
jectivity inherent in bud counting by using immuno- 0.938.15,27,32,34,38,41,48 Kappa values, a commonly used
histochemistry with anti-cytokeratin antibodies to measure of interobserver agreement, can be interpreted
highlight buds and distinguish them from surrounding as follows: o0.20, poor; 0.21–0.40, fair; 0.41–0.60,
stromal cells.12,13,22,23,31,32,38,40,43,44,48,51,54,55 The use of moderate; 0.61–0.80, good; and 40.80, very good.56
immunohistochemistry in the assessment of budding Intuitively, one would expect the use of
is somewhat controversial: some authors argue that the immunohistochemistry to improve interobserver
evaluation of budding should be limited to H&E- variability, but studies assessing interobserver varia-
stained slides because of the cost and impracticality of bility on immunohistochemically stained slides
performing immunohistochemistry in routine cases, have reported kappa values ranging from 0.53 to
while others argue that immunohistochemistry should 0.874,32,38,48 similar to those reported by authors
be used routinely to improve the accuracy and assessing tumour budding on H&E.15,27,32,34,41 Only
reproducibility of bud counts. In addition to cost one study has directly compared interobserver
considerations, the use of immunohistochemistry may variability between assessments performed on H&E
also require the establishment of a separate cutoff for and immunohistochemically stained slides among
defining positive budding. One study comparing the the same group of pathologists: when Suzuki et al32
bud counts obtained on H&E to those obtained with evaluated budding with both H&E and immuno-
immunohistochemistry, not surprisingly, found signif- histochemistry, they showed only a modest
icantly higher counts with the latter.31 Indeed, the few improvement in interobserver variability when
studies that have attempted to define an optimal anti-cytokeratin antibodies were used (k ¼ 0.41 with
budding threshold on anti-cytokeratin-stained slides H&E, and k ¼ 0.53 with immunohistochemistry).
have reported higher cutoffs than those found to be
prognostically significant on H&E (summarized in
Table 2).
Currently, the role for immunohistochemistry in the Practical considerations in the assess-
evaluation of tumor budding is unclear, and further ment and reporting of tumor budding
studies are needed to assess the relationship between Opinions are divided as to whether tumor budding
bud counts obtained on H&E vs those obtained with should be recorded in the pathological assessment
anti-cytokeratin antibodies, and to determine the most of colorectal carcinoma. Although the College of
prognostically useful cutoff for bud counting with both American Pathologists has not included budding in
H&E and anti-cytokeratin antibodies. its checklist of reportable features, both the Associa-
tion of Directors of Anatomic and Surgical Pathology
Interobserver Variability in the Assessment of Tumor and the Union for International Cancer Control
Budding recommend its inclusion in routine reporting.57,58
Our institutional policy is to report the presence
In light of these myriad sources of subjectivity, or absence of tumor budding in all malignant
one would expect the evaluation of budding to polyps and colorectal cancer resection specimens.

Modern Pathology (2012) 25, 1315–1325


Tumor budding
B Mitrovic et al 1323

Although we recognize the limitations posed by a only on its presence, but on its associated risk of
lack of definitional uniformity, we feel that its nodal metastasis (as we do for lymphovascular
assessment is important both for the purposes of invasion and a poorly differentiated component).
data gathering, and, in the setting of malignant Our approach will undoubtedly need to be refined
polyps, for guiding treatment decisions. once optimal evidence-based guidelines are estab-
Until universally accepted criteria are established, lished. However, based on the available evidence,
we apply those of Ueno et al which are not only of and in an area lacking standardization, this strategy
proven prognostic significance, but are also easily provides institutional guidelines and consistency
applied and are the most widely used criteria in the for reporting of tumour budding.
budding literature. We report tumor budding as
present if Z10 groups of o5 cells are counted in
a 20  objective field (ie, Ueno’s so-called ‘high- Conclusions and future directions
grade budding’).27 In borderline cases, where only
5–10 definite buds/20  field are identified but there There is strong evidence to suggest that tumor bud-
are additional possible tumor cells that cannot be ding is an adverse prognostic factor that can help to
confidently differentiated from stroma or histio- stratify patients into more meaningful risk groups
cytes, a cytokeratin stain is performed. If this than TNM staging alone, and, even more impor-
confirms the impression of additional tumor cells, tantly, has the potential to guide decision making.
bringing the count to Z10 buds, we report as Its presence in T1 colorectal carcinoma, when
positive for tumor budding. However, we caution evaluated in conjunction with other prognostically
against the routine use of cytokeratin stains in cases significant clinical and histopathological features,
where bud counts on H&E do not approach 10/ can identify a subset of high-risk patients requiring
20  objective. In our experience, counts by cyto- segmental resection including nodes. In the setting
keratin immunohistochemistry are substantially of Stage II colorectal carcinoma, the presence of
higher than those on H&E and the limited data budding has been associated with significantly
suggest that much higher cutoffs are needed to reach worse outcomes, and if this subset of patients is
prognostic significance.23,38,48 As tumor budding shown to derive a survival benefit from chemo-
is most prominent at the invasive tumor front, we therapy, the evaluation of budding could potentially
concentrate on, but do not restrict our assessment to, assume an important role in treatment algorithms in
this area. Stage II colorectal carcinoma.
At scanning power, clues to the presence of tumor In order for the considerable prognostic power
budding include: of tumor budding to be fully realized, consensus
criteria for its evaluation must be established, both
(a) infiltrating growth pattern; to guide further research in this area and to provide
(b) marked irregularity at invasive front; and
the practicing pathologist with guidelines for its
(c) blurring of the interface between tumor and
reporting. With respect to setting these criteria,
underlying stroma.
studies focusing on budding should be designed to
The presence of these features prompts close determine, if possible, the optimal quantitative
evaluation at medium and high magnification. cutoff for defining clinically significant tumor
In our experience, there are several scenarios in budding. Tumor budding is a continuous variable;
which tumor-budding assessment is challenging: thus, any cutoff is bound to be at least somewhat
arbitrary, but an attempt must be made at defining
(a) abundance of histiocytes and activated lympho-
the budding threshold that will provide the most
cytes at invasive front (Figure 2a);
clinically relevant prognostic and predictive infor-
(b) prominent stromal reaction (Figure 2c);
mation. Furthermore, authors reporting on tumor
(c) glandular fragmentation associated with a
budding should provide more detailed information
marked acute inflammatory infiltrate; regarding the qualitative and quantitative criteria
(d) tumour fragments floating in mucin pools; and
used to evaluate budding in order to allow for
(e) fragmented glands with surrounding retraction
meaningful comparisons among different studies.
artifact (Figure 2e)
Finally, the role of immunohistochemistry in the
In scenarios (a) and (b), a cytokeratin stain is used evaluation of budding needs to be clarified.
to distinguish between tumor cells and stromal/ Although it is probably unnecessary and impractical
inflammatory cells (Figures 2b and d). In scenarios to perform immunohistochemistry on all colorectal
(c) to (e), the fragmented cells are excluded from bud carcinoma cases, there may be certain situations,
counts. particularly in the context of a prominent peritu-
We believe it is particularly important to report moral inflammatory reaction, where a cytokeratin
tumour budding in the setting of malignant polyps, stain may reveal buds that are obscured on H&E.
as this finding increases the risk of nodal metastasis Criteria for immunohistochemical evaluation there-
to around 15–20%,26 and should prompt considera- fore also need to be established.
tion of a segmental resection with lymphadenec- The greatest obstacle to the adoption of budding
tomy. As such, we make a point of commenting not as a routinely reportable feature is the lack of

Modern Pathology (2012) 25, 1315–1325


Tumor budding
1324 B Mitrovic et al

well-defined, evidence-based criteria for its assess- 14 Lugli A, Vlajnic T, Giger O, et al. Intratumoral budding
ment. Given the overwhelming evidence that tumor as a potential parameter of tumor progression in
budding is one of the most powerful prognostic mismatch repair-proficient and mismatch repair-defi-
factors at our disposal, it is incumbent on the GI cient colorectal cancer patients. Hum Pathol 2011;
42:1833–1840.
pathology community to move swiftly to address 15 Choi HJ, Park KJ, Shin JS, et al. Tumor budding as a
and remove the barriers to its universal reporting. prognostic marker in stage-III rectal carcinoma. Int J
Colorectal Dis 2007;22:863–868.
16 Guzinska-Ustymowicz K. The role of tumour budding
Acknowledgement at the front of invasion and recurrence of rectal
carcinoma. Anticancer Res 2005;25:1269–1272.
We thank Dr Masanori Tanaka (Department of 17 Ha SS, Choi HJ, Park KJ, et al. Intensity of tumor
Pathology, City Hospital, Hirosaki, Japan) for assis- budding as an index for the malignant potential
tance in obtaining and translating Japanese articles. in invasive rectal carcinoma. Cancer Res Treat 2005;
37:177–182.
18 Hase K, Shatney C, Johnson D, et al. Prognostic value
Disclosure/conflict of interest of tumor ‘budding’ in patients with colorectal cancer.
Dis Colon Rectum 1993;36:627–635.
The authors declare no conflict of interest. 19 Homma Y, Hamano T, Otsuki Y, et al. Severe tumor
budding is a risk factor for lateral lymph node meta-
stasis in early rectal cancers. J Surg Oncol 2010;102:
230–234.
References 20 Kajiwara Y, Ueno H, Hashiguchi Y, et al. Risk factors of
nodal involvement in T2 colorectal cancer. Dis Colon
1 Greenlee RT, Hill-Harmon MB, Murray T, et al. Cancer Rectum 2010;53:1393–1399.
statistics, 2001. CA Cancer J Clin 2001;51:15–36. 21 Kanazawa H, Mitomi H, Nishiyama Y, et al. Tumour
2 Puppa G, Sonzogni A, Colombari R, et al. TNM staging budding at invasive margins and outcome in colorectal
system of colorectal carcinoma: a critical appraisal cancer. Colorectal Dis 2008;10:41–47.
of challenging issues. Arch Pathol Lab Med 2010;134: 22 Kazama S, Watanabe T, Ajioka Y, et al. Tumour
837–852. budding at the deepest invasive margin correlates with
3 Imai T. The growth of human carcinoma: a morpho- lymph node metastasis in submucosal colorectal
logical analysis. Fukuoka Igaku Zasshi 1954;45:30. cancer detected by anticytokeratin antibody CAM5.2.
4 Chaffer CL, Weinberg RA. A perspective on cancer cell Br J Cancer 2006;94:293–298.
metastasis. Science 2011;331:1559–1564. 23 Lugli A, Karamitopoulou E, Panayiotides I, et al. CD8+
5 Kalluri R. EMT: when epithelial cells decide to become lymphocytes/tumour-budding index: an independent
mesenchymal-like cells. J Clin Invest 2009;119: prognostic factor representing a ‘pro-/anti-tumour’
1417–1419. approach to tumour host interaction in colorectal
6 Kalluri R, Weinberg RA. The basics of epithelial- cancer. Br J Cancer 2009;101:1382–1392.
mesenchymal transition. J Clin Invest 2009;119: 24 Masaki T, Matsuoka H, Sugiyama M, et al. Tumor
1420–1428. budding and evidence-based treatment of T2 rectal
7 Acloque H, Adams MS, Fishwick K, et al. Epithelial- carcinomas. J Surg Oncol 2005;92:59–63.
mesenchymal transitions: the importance of changing 25 Sohn DK, Chang HJ, Park JW, et al. Histopathological
cell state in development and disease. J Clin Invest risk factors for lymph node metastasis in submucosal
2009;119:1438–1449. invasive colorectal carcinoma of pedunculated or
8 Zlobec I, Lugli A. Epithelial mesenchymal transition semipedunculated type. J Clin Pathol 2007;60:
and tumor budding in aggressive colorectal cancer: 912–915.
tumor budding as oncotarget. Oncotarget 2010;1: 26 Ueno H, Mochizuki H, Hashiguchi Y, et al. Risk factors
651–661. for an adverse outcome in early invasive colorectal
9 Gabbert H, Wagner R, Moll R, et al. Tumor dedifferen- carcinoma. Gastroenterology 2004;127:385–394.
tiation: an important step in tumor invasion. Clin Exp 27 Ueno H, Murphy J, Jass JR, et al. Tumour ‘budding’ as
Metastasis 1985;3:257–279. an index to estimate the potential of aggressiveness
10 Prall F, Ostwald C, Linnebacher M. Tubular invasion in rectal cancer. Histopathology 2002;40:127–132.
and the morphogenesis of tumor budding in colorectal 28 Ueno H, Price AB, Wilkinson KH, et al. A new
carcinoma. Hum Pathol 2009;40:1510–1512. prognostic staging system for rectal cancer. Ann Surg
11 Morodomi T, Isomoto H, Shirouzu K, et al. An index 2004;240:832–839.
for estimating the probability of lymph node metas- 29 Yamauchi H, Togashi K, Kawamura YJ, et al. Patholo-
tasis in rectal cancers. Lymph node metastasis and the gical predictors for lymph node metastasis in T1
histopathology of actively invasive regions of cancer. colorectal cancer. Surg Today 2008;38:905–910.
Cancer 1989;63:539–543. 30 Wang HS, Liang WY, Lin TC, et al. Curative resection
12 Shinto E, Jass JR, Tsuda H, et al. Differential prognostic of T1 colorectal carcinoma: risk of lymph node
significance of morphologic invasive markers in color- metastasis and long-term prognosis. Dis Colon Rectum
ectal cancer: tumor budding and cytoplasmic podia. 2005;48:1182–1192.
Dis Colon Rectum 2006;49:1422–1430. 31 Ohtsuki K, Koyama F, Tamura T, et al. Prognostic value
13 Shinto E, Mochizuki H, Ueno H, et al. A novel classi- of immunohistochemical analysis of tumor budding in
fication of tumour budding in colorectal cancer based colorectal carcinoma. Anticancer Res 2008;28:1831–1836.
on the presence of cytoplasmic pseudo-fragments 32 Suzuki A, Togashi K, Nokubi M, et al. Evaluation of
around budding foci. Histopathology 2005;47:25–31. venous invasion by Elastica van Gieson stain and

Modern Pathology (2012) 25, 1315–1325


Tumor budding
B Mitrovic et al 1325

tumor budding predicts local and distant metastases 46 Gray R, Barnwell J, McConkey C, et al. Adjuvant
in patients with T1 stage colorectal cancer. Am J Surg chemotherapy versus observation in patients with
Pathol 2009;33:1601–1607. colorectal cancer: a randomised study. Lancet 2007;
33 Morodomi T. Clinicopathological studies of advanced 370:2020–2029.
rectal cancers—prediction of the degree of lymph 47 Sy J, Fung CL, Dent OF, et al. Tumor budding and
node metastasis from histopathological finding of pre- survival after potentially curative resection of node-
operative biopsy specimens. Nippon Geka Gakkai positive colon cancer. Dis Colon Rectum 2010;53:
Zasshi 1988;89:352–364. 301–307.
34 Okuyama T, Oya M, Ishikawa H. Budding as a useful 48 Zlobec I, Molinari F, Martin V, et al. Tumor budding
prognostic marker in pT3 well- or moderately-differ- predicts response to anti-EGFR therapies in metastatic
entiated rectal adenocarcinoma. J Surg Oncol 2003;83: colorectal cancer patients. World J Gastroenterol
42–47. 2010;16:4823–4831.
35 Okuyama T, Nakamura T, Yamaguchi M. Budding is 49 Jass JR, Barker M, Fraser L, et al. APC mutation and
useful to select high-risk patients in stage II well- tumour budding in colorectal cancer. J Clin Pathol
differentiated or moderately differentiated colon ade- 2003;56:69–73.
nocarcinoma. Dis Colon Rectum 2003;46:1400–1406. 50 Kevans D, Wang LM, Sheahan K, et al. Epithelial-
36 Nakamura T, Mitomi H, Kanazawa H, et al. Tumor Mesenchymal Transition (EMT) protein expression in
budding as an index to identify high-risk patients with a cohort of stage II colorectal cancer patients with
stage II colon cancer. Dis Colon Rectum 2008;51: characterized tumor budding and mismatch repair
568–572. protein status. Int J Surg Pathol 2011;19:751–760.
37 Yasuda K, Inomata M, Shiromizu A, et al. Risk factors 51 Shinto E, Baker K, Tsuda H, et al. Tumor buds show
for occult lymph node metastasis of colorectal cancer reduced expression of laminin-5 gamma 2 chain in
invading the submucosa and indications for endo- DNA mismatch repair deficient colorectal cancer. Dis
scopic mucosal resection. Dis Colon Rectum 2007;50: Colon Rectum 2006;49:1193–1202.
1370–1376. 52 Mikami T, Yoshida T, Numata Y, et al. Invasive
38 Prall F, Nizze H, Barten M. Tumour budding as behavior of ulcerative colitis-associated carcinoma is
prognostic factor in stage I/II colorectal carcinoma. related to reduced expression of CD44 extracellular
Histopathology 2005;47:17–24. domain: comparison with sporadic colon carcinoma.
39 Ueno H, Jones AM, Wilkinson KH, et al. Histological Diagn Pathol 2011;6:30.
categorisation of fibrotic cancer stroma in advanced 53 Masaki T, Matsuoka H, Sugiyama M, et al. Actual
rectal cancer. Gut 2004;53:581–586. number of tumor budding as a new tool for the
40 Syk E, Lenander C, Nilsson PJ, et al. Tumour budding individualization of treatment of T1 colorectal carci-
correlates with local recurrence of rectal cancer. nomas. J Gastroenterol Hepatol 2006;21:1115–1121.
Colorectal Dis 2011;13:255–262. 54 Ogawa T, Yoshida T, Tsuruta T, et al. Tumor budding
41 Wang LM, Kevans D, Mulcahy H, et al. Tumor budding is predictive of lymphatic involvement and lymph
is a strong and reproducible prognostic marker in T3N0 node metastases in submucosal invasive colorectal
colorectal cancer. Am J Surg Pathol 2009;33:134–141. adenocarcinomas and in non-polypoid compared with
42 Tytherleigh MG, Warren BF, Mortensen NJ. Manage- polypoid growths. Scand J Gastroenterol 2009;44:
ment of early rectal cancer. Br J Surg 2008;95:409–423. 605–614.
43 Ishikawa Y, Akishima-Fukasawa Y, Ito K, et al. Histo- 55 Prall F, Ostwald C. High-degree tumor budding and
pathologic determinants of regional lymph node meta- podia-formation in sporadic colorectal carcinomas
stasis in early colorectal cancer. Cancer 2008;112: with K-ras gene mutations. Hum Pathol 2007;38:
924–933. 1696–1702.
44 Losi L, Ponti G, Gregorio CD, et al. Prognostic 56 Altman DG. Practical Statistics for Medical Research,
significance of histological features and biological Vol, Chapman and Hall: London, 1991.
parameters in stage I (pT1 and pT2) colorectal adeno- 57 Turner RR, Li C, Compton CC. Newer pathologic
carcinoma. Pathol Res Pract 2006;202:663–670. assessment techniques for colorectal carcinoma. Clin
45 Tanaka M, Hashiguchi Y, Ueno H, et al. Tumor Cancer Res 2007;13:6871s–6876s.
budding at the invasive margin can predict patients 58 Jass JR, O’Brien MJ, Riddell RH, et al. Recommenda-
at high risk of recurrence after curative surgery for tions for the reporting of surgically resected specimens
stage II, T3 colon cancer. Dis Colon Rectum 2003; of colorectal carcinoma. Virchows Arch 2007;450:
46:1054–1059. 1–13.

Modern Pathology (2012) 25, 1315–1325

You might also like