ArticlePDF AvailableLiterature Review

Tumor deposits in colorectal cancer: Refining their definition in the TNM system

Wiley
Annals of Gastroenterological Surgery
Authors:

Abstract and Figures

Tumor deposits (TDs) are discontinuous tumor spread in the mesocolon/mesorectum which is found in approximately 20% of colorectal cancer (CRC) and negatively affects survival. We have a history of repeated revisions on TD definition and categorization in the tumor-node-metastasis (TNM) system leading to stage migration. Since 1997, TDs have been categorized as T or N factors depending on their size (TNM5) or contour (TNM6). In 2009, TNM7 provided the category of N1c for TDs in a case without positive lymph nodes (LNs), which is also used in TNM8. However, increasing evidence suggests that these revisions are suboptimal and only “partially” successful. Specifically, the N1c rule is certainly useful for oncologists who are having difficulty with TDs in a case with no positive LNs. However, it has failed to maximize the value of the TNM system because of the underused prognostic information of individual TDs. Recently, the potential value of an alternative staging method has been highlighted in several studies using the “counting method.” For this method, all nodular type TDs are individually counted together with positive LNs to derive the final pN, yielding a prognostic and diagnostic value that is superior to existing TNM systems. The TNM system has long stuck to the origin of TDs in providing its categorization, but it is time to make way for alternative options and initiate an international discussion on optimal treatment of TDs in tumor staging; otherwise, a proportion of patients end up missing an opportunity to receive the optimal adjuvant treatment.
This content is subject to copyright. Terms and conditions apply.
Ann Gastroenterol Surg. 2023;7:225–235.
|
225www.AGSjournal.com
1 |INTRODUCTION
Tumor staging in conventional pathological investigation is still at
the center of clinical reporting on colorectal cancer (CRC) even in the
precision medicine era . A recent milestone s tudy by the International
Duration of Adjuvant Chemotherapy (IDEA) collaboration high-
lighted the importance of the sub- staging of stages II and stage III
in determining an adjuvant chemotherapy regimen for individual pa-
tients.1,2 Currently, T and N stages are unprecedentedly important
as they determine the selection of patients for the chemotherapy
regimen, thereby determining patient clinical outcomes.3
Tumor deposits (TDs) were isolated tumor lesions in the regional
lymphatic area other than those in the lymph nodes, which were first
noticed in the 1980s by some pathologists, such as Jass and Morson.
Received: 30 Octobe r 2022 
|
Revised: 23 Nove mber 2022 
|
Accepted: 22 December 202 2
DOI: 10.1002/ags3 .12652
REVIEW ARTICLE
Tumor deposits in colorectal cancer: Refining their definition in
the TNM system
Hideki Ueno1| Iris D. Nagtegaal2| Philip Quirke3| Kenichi Sugihara4| Yoichi Ajioka5
This is an op en access ar ticle under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium,
provide d the original work is properly cited.
© 2023 The Authors. Annals of Gastroenterological Surgery published by John Wiley & S ons Australia, Ltd on behalf of The Japa nese Society of
Gastroenterological Surgery.
1Department of Surgery, National Defense
Medical College, Saitama, Japan
2Depar tment of Pathology, Radb oud
University Medical Centre, Nijmegen, The
Netherlands
3Divisio n of Patholog y and Data Analytic s,
University of Leeds, St James's Uni versity
Hospit al, Leeds, UK
4Depar tment of Surgical Oncology, Tokyo
Medical and Dental Universit y, Graduate
School of Medical and Dental Sci ences,
Tokyo, Japan
5Divisio n of Molecular and Diagnos tic
Pathology, Niigata University Graduate
School of Medical and Dental Sci ences,
Niigata, Japan
Correspondence
Hideki Ueno, Department of Surgery,
National Defense Medical College, 3- 2
Namiki, Tokorozawa, Saitama 359- 8513,
Japan.
Email: ueno_surg1@ndmc.ac.jp
Abstract
Tumor deposits (TDs) are discontinuous tumor spread in the mesocolon/mesorectum
which is found in approximately 20% of colorectal cancer (CRC) and negatively affects
survival. We have a history of repeated revisions on TD definition and categoriza-
tion in the tumor- node- metastasis (TNM) system leading to stage migration. Since
1997, TDs have been categorized as T or N factors depending on their size (TNM5)
or contour (TNM6). In 2009, TNM7 provided the category of N1c for TDs in a case
without positive lymph nodes (LNs), which is also used in TNM8. However, increasing
evidence suggests that these revisions are suboptimal and only “partially” successful.
Specifically, the N1c rule is certainly useful for oncologists who are having difficulty
with TDs in a case with no positive LNs. However, it has failed to maximize the value
of the TNM system because of the underused prognostic information of individual
TDs. Recently, the potential value of an alternative staging method has been high-
lighted in several studies using the “counting method.” For this method, all nodular
type TDs are individually counted together with positive LNs to derive the final pN,
yielding a prognostic and diagnostic value that is superior to existing TNM systems.
The TNM system has long stuck to the origin of TDs in providing its categorization,
but it is time to make way for alternative options and initiate an international discus-
sion on optimal treatment of TDs in tumor staging; otherwise, a proportion of patients
end up missing an opportunity to receive the optimal adjuvant treatment.
KEYWORDS
Extramural cancer deposits without lymph node structure (EX ), Lymph node metastasis, Tumor
deposits (TDs), tumor stage, Tumor- node- metastasis (TNM) system
226 
|
    UENO et al.
They described that “the clinical importance of this observation is
unknown at present.4 The TDs were first adopted in the Union for
International Cancer Control (UICC)'s TNM classification 5th edition
in 1997 as a staging factor (Table 1).5 Since then, pathological prac-
tice for TDs has been revised in every revision of the TNM staging
system until its 8th edition (2017). However, uncer tainty remained in
TD definition and categorization in the tumor staging system. Over
time, considerable new data around TDs has accumulated, which
should now guide us in the optimization of the TNM staging system.
2 |PROGNOSTIC IMPACT OF TDS
Early studies of the prognostic impact of TDs date back to the 1990s.
Harrison et al. investigated metastatic tumor nodules in perirectal or
pericolic fat with the definition of “discrete aggregates of carcinoma
within fibroadipose tissue unassociated with recognizable lymph
node (LN) structure and not contiguous with the mural component
of invasive carcinoma” and showed that such lesions adversely af-
fected the prognosis in rectal cancer (1994)6 and right- sided colon
cancer (1995).7 In 1997, Ueno and Mochizuki first r eported the ac tual
status of discontinuous cancer- spread lesions based on the system-
atic investigation of the lymph drainage area of a primar y carcinoma,
i.e., not only extramural adipose tissue attached to the bowel but
also adipose tissue of the regional lymphatic area that was postop-
eratively harvested for pathologic examination of LNs (Figure 1).8
Their study reported a 26% incidence rate of such lesions in patients
with curative resection and 66% in those with non- curative resec-
tion. They categorized the discontinuous lesions into four patterns:
scattering, vessel invasion, neural invasion, and nodular type, and all
types notably exer ted an adverse impact on survival.
Since the first decade of the 2000s, an increasing number of
papers has been published on the adverse prognostic impact of
TDs in CRC. The meta- analysis by Nagtegaal et al. revealed an av-
erage 22% incidence of TDs (range, 5%– 42%) in a total of 10 106
patients in 17 studies, which variously included stages I– IV colon
or rectal cancer, depending on the study.9 The hazard ratio (HR)
on the prognostic magnitude of TDs was 2. 2 (1.6– 3.0) on disease-
free sur vival (DFS) (five studies, 1246 patients), 3.3 (2.2– 4.7) on
disease- specific survival (five studies, 4446 patients), and 2.9 (2.2–
3.8) on overall survival (OS) (three studies, 814 patients) in univar-
iate analyses. Importantly, the inclusion of additional variance did
not diminish the significance of the HR for TD even in the multi-
variable models.9
Goldstein and Turner repor ted that the presence of TDs is an in-
dependent poor prognostic factor and insisted that TDs are distinct
from LN metastases (LNM) and should not be considered their prog-
nostic equivalent.10 Actually, the background of the tumor is differ-
ent between patients with positive LNM and TD, where TD is more
likely to appear in advanced tumors, with a lower 5- year survival rate
in patient s positive for TD than LNM.11 However, the HR between
positive LNM and TD were not statistically different, demonstrating
4.1– 4.5 for LNM and 4.05.3 for TD, respectively.11
3 |HISTOLOGICAL DEFINITIONS OF TDS
Varying terminology has been used in the literature to describe the
lesions associated with TDs, such as metastatic tumor nodules,6,7
extra- bowel skipped cancer infiltration,8 mesorectal microfoci,12
non- nodal metastatic foci,13 soft tissue implants of tumors,13 and
extramural discontinuous cancer spread.11 Over time, these terms
have gradually unified as TDs. However, the definition of TDs has
not yet been standardized effectively even in the UICC/American
Joint Committee on Cancer (AJCC) staging system, where patho-
logical definition of TDs has been changing at every revision from
TNM5 to TNM8 (Table 1). In its latest edition, some uncertainties
remained in TD definition, causing a diagnostic disparity of TDs, fol-
lowed by stage migration in a certain proportion of patients with
CRC (Table 2).
3.1  | Uncertainty in the diagnostic criteria for TDs
3.1.1  |  Intravascular or perineural TDs
TDs exist in various forms but can pathologically be categorized into
two types, i.e., tumor nodules (ND) (Figure 2) and relatively small de-
posits of cancer predominantly confined to the vascular (lymphatic
or venous vessel) or perineural spaces (intravascular or perineural
TDs) (Figure 3). In 1997, TDs first appeared in TNM5 with the term
“tumor nodule,” which was also adopted by TNM6. The term “TDs
(satellites)” was alternatively used in TNM7, although without speci-
fied reason, in which TDs were defined as “macro- or microscopic
nests or nodules, in the lymph drainage area of a primary carcinoma
without histological evidence of residual LN in the nodule.” It is un-
clear whether TDs under TNM5, TNM6, and TNM7 included non-
nodular TDs, such as intravascular or perineural TDs (Figure 3) that
reportedly exist in approximately 3% in the peritumoral adipose tis-
sue located at >5 mm from the primary tumor and 1%– 2% in the “LN”
specimens (specimens postoperatively harvested for pathological
examination of LN metastasis).14
A single- center study focused on the distinction between nodu-
lar and intravascular TDs and revealed HRs of 4.7 (3.5– 6.2) and 2.5
(1.6– 3.8), respectively, and the AIC value for T staging was improved
when intravascular TDs were treated as T category.15 Additionally,
a multicenter study conducted by the Japanese Society for Cancer
of the Colon and Rectum (JSCCR), which included two cohorts com-
prising 1716 patient s and 2242 patients, respectively, revealed that
the statistic figures representing the performance of tumor staging
were improved when intravascular or perineural TDs were treated
as a T- factor.14
TNM7 and TNM 8 have provided a distin ction between p erineural
and lymphovascular invasions in terms of being treated as a T- factor
and V (venous invasion) or L (lymphatic invasion) classification, re-
spectively.16,17 However, the tumor stage, rather than V and L classi-
fication, is currently considered mostly as a treatment decision factor
in clinical practice; thus, we may better revisit the concept adopted
   
|
227
UENO et al.
TABLE 1 Definition and categorization of tumor deposits (TDs) in staging systems
Staging system
(publication year) Terminology Definition Categorization criteria for tumor staging
TNM5 (1997) Tumor nodule A nodule in perirect al or
pericolic adipose tissue
without histological
evidence of a residual
lymph node in the nodule.
A tumor nodule of >3 mm in diameter is classified as regional
LNM; a tumor nodule up to 3 mm in diameter is classified in
the T category as a discontinuous extension, i.e., T3.
TNM6 (2002) Tumor nodule A tumor nodule in the pericolic
or perirectal adipose
tissue without histological
evidence of residual lymph
node in the nodule.
If the nodule has the form and smooth contour of a lymph
node, it is classified in the pN categor y as a regional LNM;
if the nodule has an irregular contour, it should be classified
in the T category and also coded as V1 (microscopic venous
invasion) or V2, if it was grossly evident, because there is a
strong likelihood that it represents venous invasion.
TNM7 (2009) Tumor deposit s
(satellites)
Macroscopic or microscopic
nests or nodules in the
pericolorectal adipose
tissue's lymph drainage
area of a primary
carcinoma without
histological evidence of
residual lymph node in the
nodule.
If tumor deposits are observed with lesions that would
otherwise be classified as T1 or T2, then the T
classification is not changed, but the nodule(s) is recorded
as N1c. If a nodule is considered by the pathologist as a
totally replaced lymph node (generally having a smooth
contour), it should be recorded as a positive lymph node
and not as a satellite, and each nodule should be separately
counted as a lymph node in the final pN determination.
TNM8 (2017) Tumor deposit s
(satellites)
Discrete macroscopic or
microscopic nodules of
cancer in the pericolorectal
adipose tissue's lymph
drainage area of a
primar y carcinoma that
are discontinuous from
the primary and without
histological evidence of
residual lymph node or
identifiable vascular or
neural structure.
If a vessel wall is identifiable on H&E, elastic, or other stains,
it should be classified as a venous invasion (V1/2) or
lymphatic invasion (L1). Similarly, the lesion should be
classified as a perineural invasion (Pn1) if neural structures
are identifiable.
The presence of tumor deposits does not change the primary
tumor T category but changes the node status (N) to pN1c
if all regional lymph nodes are negative on pathological
examination.
JSCCR8 (2013)
JSCCR9 (2018)
Extr amural cancer
deposits
without lymph
node structure
(EX)
Extramural cancer deposits
with no lymph node
struc ture within the
regional lymph node area.
EX includes localized
lesions comprising
lymphatic invasion, venous
invasion, perineural
invasion (vascular/
perineural invasion lesions),
and other lesions (tumor
nodule: [ND]).
All tumor deposits located
in the extramural f atty
tissue are regarded as
EX in tumors in which
continuous spread is
confined within the SM
or MP. Tumor deposits
located ≥5 mm from the
leading edge of the primary
tumor are designated as
EX for tumors that directly
penetrate the MP.
ND is treated as LNM and each ND is separately counted as
a lymph node in the final pN determination. Vascular/
perineural invasion lesions are treated as T- factor, thereby
changing the final pT determination (i.e., T3) in tumors that
would otherwise be classified as T1 or T2.
ND with his tological evidence of venous invasion or perineural
invasion in the nodule is recorded with a symbol of ND(V+)
or ND(Pn+) be cause it represents a strong likelihood of
getting a poor prognosis.
Abbreviation: EX, extramural cancer deposit without lymph node structure; H&E, hematoxylin and eosin staining; JSCCR , Japanese Society for
Cancer of the Colon and Rectum; LNM, lymph node metastasis; MP, muscularis propria; ND(Pn+), tumor nodule with histological evidence of
perineural invasion in the nodule; ND(V+), tumor nodule with histological evidence of venous invasion in the nodule; ND, tumor nodule without
histological evidence of residual lymph node structure; SM, submucosal layer.
228 
|
    UENO et al.
in TNM5 and TNM6 in which prognostic information of all TD types
was effectively reflected in either the T or N stage. In Japan, intra-
vascular or perineural TDs are treated differently from nodular TDs
in determining the tumor stage. Specifically, intravascular or perineu-
ral TDs are regarded as a T- factor, thereby making a tumor upstage
to pT3 if observed in otherwise pT1 or pT2 tumors. This may be com-
parable to the concept of the TD treatment adopted in TNM5 where
a pericolic or perirectal tumor nodule of up to 3 mm in diameter was
treated as a T category, i.e., a T3- determinant factor.5
3.1.2  |  TDs with identifiable vascular or
neural structure
TD definition has increased in complexity in TNM7, which demands
pathologists to distinguish whether the origin of the TD is LNM or
not. Furthermore, the lesions with histological evidence of identifi-
able vascular or neural structure are not regarded as TDs by defini-
tion in TNM8. Conceivably, this is based on the concept that TDs
should only be applied to lesions having no identifiable origin,18 but
this is becoming a cause of st age migration (Figure 4). All isolated
tumor lesions in the mesocolon or mesorectum, including LNM, have
originated from migrated tumor cells that travel via the preexisting
anatomical structures, such as the lymphatic or venous systems, or
less frequently, the neural system, regardless of whether these are
pathologically evident or not. Differentiating the treatment of TDs
based on their assumed origin in the absence of clear evidence that
such pathological practice benefits patients in planning postopera-
tive treatment would not be logical.
Currently, approximately one in five patients with nodular TDs
also show signs of venous or perineural invasion in the nodule, and
such lesions have valuable prognostic information.11 The estimated
5- year survival rate of patients with such lesions was as low as 30%–
45% based on a single- center study and two cohorts in the JSCCR's
multicenter study.11,15 In Japan, since 2013, these no dules have been
recorded in pathological report s with the symbols ND(V+), ND(Pn+),
or ND(V&Pn+) and are incorporated in tumor staging, similar to
other nodular TDs.19
3.2  | What should be the distance of TDs from the
primary tumor?
Since TNM5, uncertainties were obser ved in the UICC/AJCC
definition concerning the area of peritumoral TD evaluation.
Specifically, the distance from the primary tumor or the bowel
wall for an isolated tumor lesion to be diagnosed as a TD has no
consensus.16,17 Approximately 16% of T3/T4 tumors have peritu-
moral deposits discontinuously located at >2 mm from the body
of the primary tumor and the muscularis propria. 20 TNM8 recom-
mended the lesions to be “discontinuous” from the primar y tumor
to be classified as TDs, but without specific criteria for judging
this “discontinuity,” resulting in a great deal of inconsistency in TD
diagnoses (Figure 5). Nagtegaal and Quirke brought up the dif-
ficulties that arise when determining whether a TD is a deposit
or just a continuous growth of tumor, causing TD misdiagnoses.21
Certainly, such a difficulty may well be understood in pathological
images of TDs used in some studies, in which the TD is so close
FIGURE 1 Area of adipose tissue harboring tumor deposit (TD). TDs exist in extramural adipose tissue attached to the bowel wall with
the primary tumor (A) and in a lump of adipose tissue postoperatively harvested for pathologic examination of LN metastasis (B). Regarding
the length of discontinuity to define peritumoral TD (two- headed arrow in [A]), a yardstick of 5- mm discontinuit y is used as a criterion for
judging a peritumoral TD in the Japanese classification of colorec tal, appendiceal, and anal carcinoma (third English edition), i.e., only a
deposit discontinuously located at ≥5 mm from the main body of the primar y tumor is attributed to the final pathological stage (see Figure 5).
LN, Lymph node; MP, Muscularis propria; TD, Tumor deposit
(A)
(B)
   
|
229
UENO et al.
to the main body of the primary tumor that it is located within the
connective tissue extending directly from the primary tumor.22– 24
Since 2013, guidance for this issue was provided in the
Japanese Classification of Colorectal Carcinoma (8th Japanese
edition),25 wherein all deposits located in the extramural fatty tis-
sue are regarded as TDs in a tumor that is other wise diagnosed as
T1 or T2. A yardstick of 5 mm of discontinuity is used as a crite-
rion for judging a peritumoral TD for T3 or T4 tumors, i.e., only a
deposit discontinuously located ≥5 mm from the main body of the
primar y tumor can be attributed to the final pathological stage
(Figure 1). The 5- mm criterion for peritumoral deposits had been
arbitrarily determined based on the consensus of the committee
in the JSCCR , who emphasized the results of a multicenter study
in which the prognostic value of tumor stage had been improved
by the pre- planned assessment criteria for TDs, including the
5- mm criterion.11,14,26
Similarly, other pathologists attempted to make a yardstick for
the term “discontinuously.” For example, Gopal et al. determined
that at least 1 cm from the advancing edge was needed to diagnose
TDs.23 Conversely, Frankel and Jin considered the tumor nodule a
TD irresp ective of the dist ance of the tumor nod ule from the leading
edge of the tumor, when there is no clear connection and the nod-
ule appears discrete.27 However, this manner requires demanding
costly deeper sectioning to ascertain whether there is truly no con-
nection from the leading edge of the primary tumor body.
4 |TD CATEGORIZATION IN THE TNM
CLASSIFICATION
The TNM classification has categorized TDs based on the “size rule”
in TNM5, the “contour rule” in TNM6, and placed them into the “N1c
category” in TNM7 and TNM8. More specifically, a tumor nodule of
>3 mm is classified as the N category and up to 3 mm as the T cate-
gory in TNM5. The size rule might be introduced in the presumption
that the larger the TD was, the more likely that it originated in LNM,
although the rationale for the cut- off was not disclosed. In TNM6,
TDs were classified in the N category if the nodule had the form and
smooth contour of an LN.
Size or contour criteria are not presented in TNM7 or TNM8. TDs
were no longer treated as a T category,28 although there is a confusing
description in the 4th (2012)16 and 5th editions (2019)17 of the TNM
Supplement that discontinuous extramural extension becomes a rea-
son for a “pT3” diagno sis if there is regiona l LNM. TDs were classif ied as
N1c in tumors that would otherwise be classified as N0 in TNM7. The
number of TDs does not affect the N category although TDs should
TABLE 2 Issues to be solved for future international tumor staging systems regarding the definition and categorization of tumor
deposits (TDs)
Points at issue in the TNM8 Evidence to date associated with the issue
1. Uncertainty regarding the
definition of the “discontinuity” in
peritumoral TD diagnosis
1. There is no international consensus as to the definition of the distance from the advancing edge
of the main body of the primary tumor or bowel wall for peritumoral TD. Some criteria have been
proposed such as 1) ≥5 mm,11, 26 2) ≥1 cm,23 and 3) no clear connection.27
2. The “5- mm” is the only criterion for peritumoral TD that had been predetermined in a multicenter
st udy,11,26 and this criterion has been used across Japan since 2013.19
2. Uncer tainty regarding the
appropriateness of isolated
intravascular or perineural TDs
not being t aken into account in
tumor staging
1. Intravascular or perineural TDs have b een included in many studies to analyze the prognostic impact
of TDs and there is no evidence that the prognostic value of tumor st aging is improved by excluding
such lesions from staging factors.
2. The prognostic power of the T stage was improved by treating isolated intravascular/perineural
deposit s as a pT3- determining factor in tumors otherwise diagnosed as pT1 or pT2.14,15
3. Lack of meaningful rationale
of distinguishing TDs with
identifiable vascular or neural
struc tures from other TDs in
tumor staging
1. None of the studies that investigated the prognostic value of TDs have excluded TDs with
identif iable vascular or neural structure from the analyses and there is no evidence that the
prognos tic value of tumor staging is improved by excluding such TDs from staging factors.
2. Nodular TD accompanied by the finding of venous/perineural invasion in the nodule has a prognostic
value that is greater than other types of TDs.11,15
3. Impaired diagnostic reproducibility of tumor staging is inevitable due to interobserver disagreement
regarding the judgment for vascular or neural st ructure in the lesion. K appa statistic was rep ortedly
0.61 between nodular TD with venous/perineural invasion and other types of extramural
discontinuous lesion.11
4. Lack of evidence for the value of
the N1c categor y
1. A multicenter study showed that TNM7 adopting the N1c category is superior to TNM6
charac terized by the contour rule, but not to TNM5 characterized by the size rule in terms of its
prognostic power.26
2. Under the N1c rule, the tumor s tage does not change according to the number of existing TDs in the
resected specimens, but the number of TD has prognostic information.10,11,36,37
3. Impaired diagnostic reproducibility of tumor staging is inevitable due to interobserver disagreement
on the distinction between TDs and LNM. Kappa statistic was 0.74 between LNM and TDs,14 and
0.38 between “nodal” or “non- nodal” origin.18
4. An increasing number of studies indicate the value of a modified s taging system in which TDs are
counted individually same as lymph nodes in the f inal pN determination (see Ta bl e 3).
Abbreviations: CI, confidence interval; HR , hazard ratio; TD, tumor deposit.
230 
|
    UENO et al.
be separately counted and recorded in pathological reports.16,17 Also
in the TNM8 , the N1c category is used for all tumors with any T stage,
as long as all regional LNs are negative on pathological examination.
4.1  | Scientific evidence for TNM
classification revisions
After the publication of TNM6 that introduced the “contour rule,”
by which TDs were classified as LNM or venous invasion depend-
ing on whether the contour was smooth or irregular, the weak sci-
entific background in the process of revising the TNM system was
criticized.21 Already for TNM5 there were issues as the size rule
had been established based on a study that was not subsequently
published.29 Similarly, the contour rule was introduced into TNM6
based on three small studies,29 in which only 348,6 344,7 and 40 0
single- center patients10 were analyzed, respectively. None of
these studies were intended to assess the prognostic relevance
of TD in terms of it s shape, and only the prognostic impact of TDs
was repor ted according to their TD criteria. Quirke et al. ques-
tioned the validity of the TNM6 criteria for TDs because of the
contour rule.30
After the revision from the “size rule” to the “contour rule,” we
experienced further TNM classification system revisions, but the
process is not substantiated by any clear scientific evidence, thereby
inviting criticism that TNM should be restructured on a basis equiv-
alent to evidence- based guidelines.31
4.2  | TD categorization and its relevance to the
prognostic value of tumor staging
4.2.1  |  Advantages and disadvantages of the
N1c category
The category of N1c was reportedly created to make a prognostic
subgroup for oncologist s who were in a quandar y about how to treat
patients who had TDs but lacked positive LN in terms of adjuvant
therapy administration.32 Additionally, the name of the category
“N1c” was selected because the letter c was the subsequent letter
in the alphabet and not necessarily to suggest prognosis.27 N1c is
repeatedly shown to not indicate poorer survival outcome than N1b
according to propensity score matching analyses on the Surveillan ce,
Epidemiology, and End Results (SEER) database.33,34
FIGURE 2 Tumor nodules in the pericolorectal adipose tissue lymph drainage area of a primary carcinoma. In TNM6, tumor nodules
without histological evidence of residual lymph node in the nodule are classified in the pN category as a regional lymph node metastasis
if the nodule has a smooth contour (A). A nodule with an irregular contour (B) is classified in the T category and also coded as V1 or V2.
In TNM7 and TNM8, tumor nodules are no longer treated as a T category. A nodule considered by the pathologist s as a totally replaced
lymph node is regarded as a positive lymph node, and otherwise, it may change the node status to pN1c depending on some conditions
defined differently in TNM7 and TNM8. No specific criteria for a nodule that should be diagnosed as a totally replaced lymph node are
being provided other than a short explanatory note that it is “generally having a smooth contour”. (A and B), hematoxylin and eosin staining;
Bar, 1 mm
   
|
231
UENO et al.
Putting all evidence for the prognostic value of TDs into context,
a new category of N1c would be regarded as “partially” successful in
terms of prognostic stratification. Specifically, the TNM7 system has
successfully achieved its purpose because the evidence is suggesting
that N1c identifies a group of patients with poor survival outcomes,
thereby indicating the necessity of adjuvant therapy in patients with-
out LNM.35,36 Conversely, the current definition of N1c is an obsta-
cle in effectively utilizing the full prognostic information of individual
TDs. TDs are only affecting the tumor stage in patients with no pos-
itive LNs in the TNM system; furthermore, the number of TD is not
considered in deriving the final tumor stage. We can hardly agree that
the current TNM system successfully maximizes its performance of
prognostic prediction26 because of the substantial prognostic infor-
mation in the number of TDs regardless of LN positivity.10,11,36,37
4.2.2  |  Tumor staging with the “counting” principle
In 2007, a first reported single- center study aimed to clarify
how TDs should be treated in tumor staging and revealed that N
FIGURE 3 Non- nodular type tumor
deposits in the pericolorectal adipose
tissue lymph drainage area of a primary
carcinoma. An intravascular tumor deposit
located near a non- met astatic lymph node
(A) and a perineural tumor deposit (B) in
the regional lymph node area. (A and B),
hematoxylin and eosin stain; Bar, 500 mm
FIGURE 4 Stage migration caused by different categorizations of a tumor deposit ( TD) depending on staging systems. The picture in the
upper- left panel indicates a peritumoral TD with a diameter of approximately 3.5 mm with an irregular contour and an identifiable vascular
structure. Under TNM5, this nodule is classified as an LN because it is >3 mm in diameter. On the contrary, this nodule is considered a lesion
of the T category because of its contour and is also coded as venous invasion under TNM6. The category N1c is used for this nodule in the
absence of regional LN metastasis under TNM7, whereas under TNM8, the tumor stage does not change by this nodule which is regarded
as venous invasion because the vascular structure is evident (arrow). Since 2013, this nodule has been invariably treated the same as LN
metast asis to derive the final N stage in Japan. Picture, hematoxylin and eosin staining; bar, 1 mm. The inset illustrates the magnification
of the part of the nodule that is indicated with an arrow (Victoria blue– hematoxylin and eosin staining). LN, Lymph node; MP, Muscularis
propria; TD: Tumor deposit
232 
|
    UENO et al.
staging was capable of more accurately predicting survival out-
comes than TNM5 or TNM6 when the number of nodular TD
was added to that of positive LN to derive the final pN stage ir-
respective of the size, contour, or estimated original structure (the
“counting” principle).15 The validity of the “counting” principle was
strongly validated in two multicenter cohorts in a study projec ted
by the JSCCR.26 An impor tant result obtained in the JSCCR study
was that increasing numbers of nodular TDs were associated with
adverse survival outcome. More importantly, statistic indexes for
tumor staging were in favor of a revised staging system based on
the “counting” principle compared to the TNM7 system in both N
and TNM stages.26
Recently, the prognostic value of the “counting” principle was
validated by an increasing number of studies (Table 3). Song et al.
analyzed 513 patients with CRC to compare the alternative staging
system based on the “counting” principle to the TNM7 system and
revealed the superiority of the “counting” principle in terms of prog-
nostic prediction.38 Similarly, the multicenter database of Lie et al.
with 4121 patients with stage II and III CRC revealed that the revised
pN categor y based on the “counting” principle was superior to the
TNM7 pN category for predicting DFS and OS.39 Additionally, two
reports from Pei et al. analyzed the SEER database of patients with
stage III CRC (21 290 patients treated between 1975 and 201640 and
9198 patients between 2010 and 201641 ), both of which demon-
strated that the “counting” principle improved the prognostic per-
formance of pN and TNM stages.
Two post hoc analyses of a phase III study for stage III colon can-
cer repor ted the value of the “counting” principle, the IDEA France
study (1942 patients)42 and the CALGB/SWAG 80702 study (2028
patients).43 The proportion of patients having TD was reported quite
differently as 10% in the IDEA France42 and 26% in the CALGB/
SWAG 0702,43 presumably due to variation in diagnostic criteria for
TDs between France and the United States and Canada. However,
the prognostic outcomes of patients who were restaged from pN1
to pN2 by the “counting” principle were similarly shown in two stud-
ies, i.e., their DFS rate was significantly lower than that of patients
confirmed with pN1 and was comparable to that of patients initially
staged as pN2.42,43 Additionally, Pyo et al. estimated the prognos-
tic power of modified staging based on the “counting” principle in
patients who completed 6 months of CAPOX treatment.44 Patients
upstaged to N2 from an initial stage of N1 experienced significantly
worse 3- year DFS compared to those remaining in the N1 stage (73%
vs. 89%), which was comparable to patients initially staged as N2.
A limited number of studies reported that the concept of the
N1c category could be reasonably accepted, ignoring the number
of TDs. A study from China concluded that the number of TDs was
not a prognostically significant parameter in the TNM staging sys-
tem because they found no difference in survival outcome between
patients having one TD and those with >1 TDs in any examined sub-
stage except for T3N1c.35 However, the number of patients included
in each substage was as small as only 3– 42. Some pathologists in
the United States have endorsed the current TNM staging system
and argued that the number of TDs should not be added to the total
number of positive LNs,27 but the evidence is lacking for this argu-
ment in terms of whether the current TNM sys tem truly achieves the
optimal prognostic grouping in patients with CRC.
FIGURE 5 The distance of peritumoral TDs to be located from the body of the primary tumor. The UICC defines tumor deposits ( TDs)
as discrete macroscopic or microscopic nodules of cancer in the pericolorectal adipose tissue's lymph drainage area of a primary carcinoma
that are discontinuous from the primar y, but the objective judgment is difficult for the discontinuity. (A) A nodule located at 7.5 mm from
the body of the primary tumor; (B) a nodule that is located just below the body of the primary tumor and some streaks of fibrous tissue
connecting them; (C) a nodule that is connected to the body of the primary tumor with cancerous tissue. In Japan, among these nodules,
only the nodule (A) is regarded as a TD that should be recorded and treated as an N factor according to the “5- mm” rule for the discontinuity
of TDs (Japanese classification of colorectal, Appendiceal, and anal carcinoma, third English edition). (A– C), hematoxylin and eosin staining;
Bar, 1 mm
   
|
233
UENO et al.
TABLE 3 Scientific literature reporting the value of new st aging based on the “counting” principle for categorizing tumor deposits (TDs) in colorectal cancer
Authors (publication
year) Study design Patients examined Summar y of the results
Ueno et al. (2007)15 Retrospective, single-
center study
1027 patients with T2– T4 CRC (1980– 1999) pNnew was superior to pNTNM5/TNM6 in AIC for CSS, when TDs other than
intravascular TD were added to the LNM count.
Ueno et al. (2012)26 Retrospective, multicenter
study
1716 (1994– 1998: first cohor t) and 2242 (1999– 20 03:
second cohort) patients with stage I– III CRC
pNnew and pTNMnew were superior to pNTNM5/TNM6/TNM7 and pTNNTNM7,
respec tively, in AIC and C- index for DSS, when nodular TDs were added to
the LNM count.
Song et al. (2012)38 Retrospective, single-
center study
513 patients with stage III CRC (1994– 2007 ) pNnew and pTNMnew were superior to pNTNM7 and pTNMTNM7, respectively, in
C- index for CSS, when TDs were added to the LNM count .
Li et al. (2016)39 Retrospective, multicenter
study
4121 patients with stage II and III CRC (200 4– 2011) pNnew was superior to pNTNM7 in C- index for DFS and OS, when TDs were
added to the LNM count.
Nagtegaal et al. (2017)9Systematic review and
meta- analysis
17 studies comprised 10 106 patients with CRC
(1964 – 2013)
An increasing number of TD was associated with poor outcomes. The
combination of TD and LNM was associated with a signific antly higher risk
of liver met astasis than LNM alone.
Delattre et al. (2020)42 Post hoc analysis of a
clinical trial
1942 patients with stage III colon cancer (2009– 2014) in
the IDEA France study
Patient s upstaged from pN1 to pN2new by the addition of TD to LNM count had
a significantly worse DFS than those with pN1new, and it was comparable to
pN2.
Pei et al. (2020)40 Retrospective database
analysis
21 290 patients with stage III CRC from the SEER database
(1975 to 2016)
pNnew and pTNMnew were superior to pNTNM8 and pTNMTNM8, respectively, in
AUC and AIC for OS, when TDs were added to the LNM count.
Pei et al. (2020)41 Retrospective database
analysis
9198 patients with stage III CRC from the SEER database
(2010– 2016)
pNnew was superior to pN in AUC and AIC for OS, when TDs were added to the
LNM count.
Cohen et al. (2021)43 Post hoc analysis of a
clinical trial
2028 patients with stage III CRC (2010– 2015) included in
the CALGB/SWOG 80702 study
Patient s upstaged from pN1 to pN2new by the addition of TD to LNM count
had significantly worse DFS and OS than those with pN1new, and they were
comparable to pN2.
Pyo et al. (2021)44 Retrospective, single-
center study
2446 patients with stage III CRC (2010– 2019) Among patients who completed 6 months of adjuvant chemotherapy, those
upstaged from pN1 to pN2new by the addition of TD to LNM count had a
significantly worse DFS than those pN1new, and it was comparable to pN2.
Abbreviations: AIC, Akaike's information criterion; AUC, area under the receiver- operating characteristic curve; C- index, Harrell's concordance index; CRC, colorectal cancer; CSS, cancer- specific
survival; DFS, disease- free survival; DSS, disease- specific survival; OS, overall survival; pNnew and pTNMnew, revised pN and pTNM based on the “counting” principle, respectively, i.e., a new method of
categorization with adding the number of tumor deposits (TDs) to the number of LNMs to derive a final N stage; pNTNM5/TNM6/TNM7, pN according to the definition of TNM5, TNM6, or TNM7; pTNMTNM7,
pTNM according to the definition of TNM7; SEER, Sur veillance, Epidemiology, and End Results.
234 
|
    UENO et al.
4.3  | Diagnostic reproducibility of tumor staging
Under the current UICC/AJCC definition for TNM staging, patholo-
gists have to distinguish a TD from some other isolated tumor lesions,
such as (1) an intravascular or perineural deposit, (2) a totally replaced
LN, and (3) a tumor nodule accompanied by venous or perineural inva-
sion, to derive the final TNM stage. Three multi- institutional studies
addressed the issue of judgment reproducibility of TDs. The JSCCR
study which involved 11 hospitals revealed a 0.74 kappa coef ficient
for distinguishing LNM from TDs and 0.51 for distinguishing between
smooth- contour nodules as a tot ally replaced LN and other types of
discontinuous lesions.14 Similarly, Brouwer et al. reported that the
kappa value for the distinction between “nodal” or “non- nodal” origin
was only 0.38 when evaluated by eight experienced gastrointestinal
pathologists.18 Rock et al. indicated that the complete agreement on
the distinction bet ween LNM and TDs was less than half under the
definition of the AJCC 7th edition even among pathologists with a
specific interest in gastrointestinal pathology.45 All these results high-
light the difficulties of achieving sufficient interobserver agreement in
distinguishing different types of discontinuous cancer spread lesions.
Consequently, at present, there is substantial interobserver inconsist-
ency in the tumor staging of individual patients caused by pathologi-
cal practice for TDs at the moment. The “counting” principle, wherein
an individual nodular TD is to be equally treated as positive LNs ir-
respective of the size, contour, or estimated original structure, would
be a promising, one- size- fits- all solution for this challenging situation.
5 |INTERNATIONAL CONSENSUS
NEEDED FOR FUTURE REVISIONS OF TNM
CLASSIFICATION
Uncertainty and confusion still remain regarding the role of TDs in
tumor staging as listed in Table 2. The definition of peritumoral TDs
in terms of the distance from the main tumor is an important issue,
and an international consensus on the definition of “discontinuity” for
TD is warranted. In Japan, the 5- mm criterion to define a peritumoral
TD is already employed across the country. The wide- ranging prac-
tices found among the literature also highlight the need for interna-
tional consensus on how to handle pathological specimens, such as
the number of sections needed for the diagnosis of TDs, how to count
the number of TDs, and whether to use immunohistochemical staining
as an adjunct of diagnostic tool. Furthermore, we have to accumulate
clinical data for establishing how we treat intravascular or perineural
TD, wherein t he lesions are less frequently observed than nodular TDs
but give a cer tain degree of prognostic information about the patient.
The UICC/AJCC has generated the definition and categorization
of TDs in the TNM staging system in which the origin of the TDs
plays a crucial part. However, no scientific evidence was presented
for the prognostic value of the origin of TDs. Furthermore, accurate
identification of the origin is impossible rather than challenging,46
and it is highly concerning that the diagnostic reproducibility of the
TNM classification is impaired due to the diagnostic inaccuracy of
TDs.18,30 Recent evidence suggests a promising solution, which is
to incorporate individual nodular TDs in the N staging based on the
“counting” principle. Certainly, we recognize the differences be-
tween TDs and LNM on some points, such as anatomical distribu-
tion, biological aggressiveness of the primary tumor, and prognostic
impact11; however, we have to bear in mind that the most import ant
role of tumor staging is, after all, accurate prognostic prediction.
In conclusion, the treatment of TDs in tumor staging should be
determined in terms of how it will maximize the prognostic value
of TNM classification and its reproducibility. All international initia-
tives to untangle the issues around TDs need to bring us to the op-
timal TNM staging by which patients would benefit from the most
evidence- based treatment. We should all aim for an evidence- based,
reproducible, and robust TNM staging system and considering our
suggestions should improve the current situation.
CONFLICT OF INTEREST
Hideki Ueno is a current Associate Editor of the Annals of
Gastroenterological Surgery.
ORCID
Hideki Ueno https://orcid.org/0000-0002-8600-1199
REFERENCES
1. Grothey A , Sobrero AF, Shields AF, Yoshino T, Paul J, Taieb J, et al.
Duration of adjuvant chemotherapy for stage III colon cancer. N
Engl J Med. 2018;378:1177– 88.
2. Iveson TJ, Sobrero AF, Yoshino T, Souglakos I, Ou FS, Meyers JP, et al.
Duration of adjuvant doublet chemotherapy (3 or 6 months) in patients
with high- risk stage II colorectal cancer. J Clin Oncol. 2021;39:631– 41.
3. Shinto E, Ike H, Hida J, Kobayashi H, Hashiguchi Y, Hase K, et al.
Proposal of a modified subclassification system for stage III col-
orect al cancer: a multi- institutional retrospective analysis. Ann
Gastroenterol Surg. 2020;4:667– 75.
4. Jass JR, Morson BC . Reporting colorectal cancer. J Clin Pathol.
1987;40:1016– 23.
5. Sobin LH, Wittekind C . UICC TNM classification of malignant tu-
mours. 5th ed. New York, NY: John Wiley & Sons; 1997.
6. Harrison JC , Dean PJ, el- Zek y F, Vander Zwaag R. From dukes
through Jass: pathological prognostic indicators in rec tal cancer.
Hum Pathol. 1994;25:498– 505.
7. Harrison JC, Dean PJ, el- Zek y F, Vander Zwaag R. Impact of the
Crohn's- like lymphoid reaction on s taging of right- sided colon can-
cer: results of multivariate analysis. Hum Pathol. 1995;26:31– 8.
8. Ueno H, Mochizuki H. Clinical significance of ex trabowel skipped
cancer infiltration in rectal cancer. Surg Today. 1997;27:617– 22.
9. Nagtegaal ID, Knijn N, Hugen N, Mar shall HC, Sugihara K, tot T,
et al. Tumor deposits in colorectal cancer: improving the value
of modern staging- a sys tematic review and meta- analysis. J Clin
Onc ol. 2 017;35:1119– 27.
10. Goldstein NS, Turner JR. Pericolonic tumor deposits in patients
with T3N+M0 colon adenocarcinomas. Cancer. 2000;88:2228– 38.
11. Ueno H, Mochizuki H, Shirouzu K, Kusumi T, Yamada K, Ikegami
M, et al. Ac tual status of distribution and prognostic impact of ex-
tramural discontinuous cancer spread in colorectal c ancer. J Clin
Oncol. 2011;29:2550– 6.
12. Ratto C , Ricci R, Rossi C, Morelli U, Vecchio F, Doglietto G.
Mesorectal microfoci adversely affect the prognosis of patients
with rec tal cancer. Dis Colon Rectum. 2002;45:733– 43.
   
|
235
UENO et al.
13. Singh AK, Myerson RJ, Birnbaum EH, Fleshman JW, Kodner IJ,
Lockett MA, et al. Outcome of patients with rec tal adenocarcinoma
and localized pelvic non- nodal metas tatic foci. Dis Colon Rectum.
200 0;43:1217– 21.
14. Ueno H, Mochizuki H, Shirouzu K, Kusumi T, Yamada K, Ikegami
M, et al. Multicenter study for optimal categorization of extr a-
mural tumor deposit s for colorectal cancer staging. Ann Surg.
2012;255:739– 46 .
15. Ueno H, Mochizuki H, Hashiguchi Y, Ishiguro M, Miyoshi M,
Kajiwara Y, et al. Extramural cancer deposits without nodal struc-
ture in colorectal c ancer. Optimal c ategorization for prognostic
staging. Am J Clin Pathol. 2007;127:287– 94.
16. Union for International Cancer Control. TNM supplement. 4th ed.
Oxford: John Wiley & Sons, Ltd; 2012.
17. Union for International Cancer Control. TNM supplement. 5th ed.
Oxford: John Wiley & Sons, Ltd; 2019.
18. Brouwer NPM, Lord AC, Terlizzo M, Bateman AC, Wes t NP, Goldin
R, et al. Interobser ver variation in the classification of tumor depos-
its in rectal cancer— is the use of histopathological characteristics
the way to go? Virchows Arch. 2021;479:11118.
19. Japanese Society for Cancer of the Colon and Rectum. Japanese
classification of colorectal, appendiceal, and anal carcinoma. 3rd
English ed . Tokyo: Kanehara & Co; 2019.
20. Ueno H, Hashiguchi Y, Shimazaki H, Shinto E, Kajiwara Y, Nakanishi
K, et al. Peritumoral deposits as an adverse prognostic indicator of
colorec tal cancer. Am J Surg. 2014;207:70– 7.
21. Nag tegaal ID, Quirke P. Colorectal tumour deposits in the
mesorectum and pericolon; a critical review. Histopathology.
20 07 ;51 :141– 9.
22. Song JS, Chang HJ, Kim DY, Kim SY, Baek JY, Park JW, et al. Is the
N1c category of the new American joint committee on cancer stag-
ing system applicable to patient s with rect al cancer who receive
preoperative chemoradiotherapy? Cancer. 2011;117:3917 24.
23. Gopal P, Lu P, Ayers GD, Herline AJ, Washington MK. Tumor
deposits in rectal adenocarcinoma after neoadjuvant chemo-
radiation are associated with poor prognosis. Mod Pathol.
2014;27:1281 7.
24. Zhang L- N, Xiao W- W, Xi S- Y, OuYang PY, You KY, Zeng ZF, et al.
Tumor deposit s: markers of poor prognosis in patients with locally
advanced rectal cancer following neoadjuvant chemoradiotherapy.
Oncotarget. 2016;7:6335– 44.
25. Japanese Society for Cancer of the Colon and Rectum. Japanese
classification of colorectal carcinoma. 8th Japanese ed. Tokyo:
Kanehara & Co; 2013.
26. Ueno H, Mochizuki H, Akagi Y, Kusumi T, Yamada K, Ikegami M,
et al. Optimal colorec tal cancer staging criteria in TNM classifica-
tion. J Clin Oncol. 2012;30:1519– 26.
27. Frankel WL, Jin M . Serosal surfaces, mucin pools, and deposit s,
oh my: challenges in staging colorectal carcinoma. Mod Pathol.
2015;Suppl 1:S95– S108.
28. Union for International Cancer Control. TNM classific ation of malig-
nant tumours. 8th ed. West Sussex: John Wiley & Sons, Ltd; 2017.
29. Sobin LH, Compton CA, Gospodarowicz M, Greene FL, Gunderson
LL, Jessup JM, et al. ' Evidence- based medicine: the time has come
to set standards for staging'. Is a radical overhaul really needed? J
Pathol. 2010;221:361– 2.
30. Quirke P, Williams G T, Ectors N, Ensari A, Piard F, Nagtegaal I. The
future of the TNM staging system in colorectal cancer: time for a
debate? Lancet Oncol. 2007;8:651 7.
31. Quirke P, Cuvelier C, Ensari A, Glimelius B, Laurberg S, Ortiz H,
et al. Evidence- based medicine: the time has come to set standards
for staging. J Pathol. 2010;221:35760.
32. Chen VW, Hsieh M- C, Charlton ME, et al. Analysis of stage and
clinical/prognostic factors for colon and rec tal cancer from SEER
registries: AJCC and collaborative stage data collection system .
Cancer. 2014;120(Supple 23):3793– 806.
33. Shen F, Hong X. Prognostic value of N1c in colorectal c ancer: a
large population- based study using propensit y score matching. Int
J Colorectal Dis. 2019;34:1375– 83.
34. Liu C , Tian M, Pei H, Tan F, Li Y. Prognostic value of the N1c in stage
III and IV colorectal cancer: a propensity score matching study
based on the surveillance, epidemiology, and end results (SEER) da-
tabase. J Invest Surg. 2022;35:850– 9.
35. Tong L, Gao P, Wang Z, Song YX, Xu YY, Sun Z, et al. Is the sev-
enth edition of the UICC/AJCC TNM staging system reasonable
for patients with tumor deposits in colorectal cancer? Ann Surg.
2012;255:208– 13.
36. Jin M , Roth R, Rock JB, Washington MK, Lehman A, Frankel WL .
The impact of tumor deposits on colonic adenocarcinoma AJCC
TNM staging and outcome. Am J Surg Pathol. 2015;39:109– 15.
37. Shi M, Zhang H, Yao G, Wu J, Zhu C , Zhang X, et al. The role of
tumor deposits in predicting the efficacy of chemotherapy in stage
III colon c ancer. Front Oncol. 2020;10:586603.
38. Song Y- X, Gao P, Wang Z- N, Liang JW, Sun Z, Wang MX , et al.
Can the tumor deposits be counted as metastatic lymph nodes in
the UICC TNM staging system for colorectal cancer? PLoS One.
2012;7:e34 087.
39. Li J, Yang S, Hu J, Liu H, du F, Yin J, et al. Tumor deposits counted
as positive lymph nodes in TNM staging for advanced col-
orectal cancer: a retrospective multicenter study. Oncotarget.
2016;7:18269– 79.
40. Pei J- P, Zhang C- D, Fu X, Ba Y, Yue S, Zhao ZM , et al. A modified
tumor- node- metastasis classification for stage III colorectal can-
cers based on treating tumor deposits as positive lymph nodes.
Front Med. 2020;7:571154.
41. Pei J- P, Zhang C- D, Liang Y, Zhang C, Wu KZ, Li YZ, et al. A modified
pathol ogical N stage in cluding stat us of tumor deposi ts in colorec tal
cancer with nodal metastasis. Front. Oncologia. 2020;10:548692.
42. Delattre J- F, Cohen R, Henriques J, Falcoz A, Emile JF, Fratte S,
et al. Prognostic value of tumor deposits for disease- free survival
in patients with stage III colon cancer: a post hoc analysis of the
IDEA France phase III trial (PRODIGE- GERCOR). J Clin Oncol.
2020;38:1702 10.
43. Cohen R, Shi Q, Meyers J, Jin Z, Svrcek M, Fuchs C, et al. Combining
tumor deposits with the number of lymph node metastases to im-
prove the prognostic accuracy in stage III colon cancer: a post hoc
analysis of the CALGB/SWOG 80702 phase III study (Alliance). Ann
Oncol. 2021;32:1267 75.
4 4. Pyo DH, Kim SH, Ha SY, Yun SH, Cho YB, Huh JW, et al. Revised
nodal staging integrating tumor deposit counts with positive lymph
nodes in patients wit h stage III colon cancer. Ann Surg. 2021.
https://doi.org/10.1097/SLA.00000 00000 005355. Online ahead
of print.
45. Rock JB, Washington MK, Adsay NV, Greenson JK, Montgomery
EA, Robert ME, et al. Debating deposits: an interobserver variabil-
ity study of lymph nodes and pericolonic tumor deposits in colonic
adenocarcinoma. Arch Pathol Lab Med. 2014;138:636– 42.
46. Wünsch K, Müller J, Jähnig H, Herrmann RA , Arnholdt HM, Märkl
M. Shape is not associated with the origin of pericolonic tumor de-
posits . Am J Clin Pathol. 2010;133:388– 94.
How to cite this article: Ueno H, Nagtegaal ID, Quirke P,
Sugihara K, Ajioka Y. Tumor deposits in colorectal cancer:
Refining their definition in the TNM system. Ann
Gastroenterol Surg. 2023;7:225–235. https://doi.org/10.1002/
ags 3.12652
... TDs are de ned as tumor foci in the pericolic or perirectal fat, distant from the tumor invasion front, in the lymphatic drainage territory of the tumor, without recognizable residual lymph node tissue [7,10]. TDs are detected in approximately 20% of CRC, and have important prognostic impact [11,12]. In the 8th AJCC, TDs, whatever their number, are subcategorized as N1c in pathologic staging only in the absence of LNM [7]. ...
... Therefore, TDs have no impact on pathological staging in the presence of LNM. Hence, the current AJCC de nition may underestimate the clinical impact of TDs in CRC [11,12]. Ueno et al. stated that the potential value of an alternative staging method has been highlighted in several studies using the "counting method", in which all nodular type of TDs are individually counted together with positive LNs to derive the nal pN category, yielding a prognostic and diagnostic value that is superior to existing TNM systems [11]. ...
... Hence, the current AJCC de nition may underestimate the clinical impact of TDs in CRC [11,12]. Ueno et al. stated that the potential value of an alternative staging method has been highlighted in several studies using the "counting method", in which all nodular type of TDs are individually counted together with positive LNs to derive the nal pN category, yielding a prognostic and diagnostic value that is superior to existing TNM systems [11]. ...
Preprint
Full-text available
Background Although previous studies have demonstrated that tumor deposits (TDs) are associated with worse prognosis in colon cancer, their clinical significance in rectal cancer has not been fully elucidated, especially in the lateral pelvic lymph node (LPLN) area. This study aimed to clarify the clinical significance of TDs, focusing on the number of metastatic foci, including lymph node metastasis (LNM) and TD, in the LPLN area. Methods This retrospective study involved 226 consecutive patients with cStage II/III low rectal cancer who underwent LPLN dissection. Metastatic foci, including LNM and TD, in the LPLN area were defined as lateral pelvic metastasis (LP-M). LP-M was evaluated according to LP-M status: presence (absence vs presence), histopathological classification (LNM vs TD), and number (1 to 3 vs 4 or more). We evaluated the relapse-free survival of each model and compared them using the Akaike information criterion (AIC) and Harrell’s concordance index (c-index). Results Forty-nine of 226 patients (22%) had LP-M, and 15 patients (7%) had TDs. The median number of LP-M per patient was one (range, 1-9). The best risk stratification power was observed for number (AIC, 758; c-index, 0.668) compared with presence (AIC, 759; c-index, 0.665) or histopathological classification (AIC, 761; c-index, 0.664). The number of LP-M was an independent prognostic factor for both relapse-free and overall survival, and was significantly associated with cumulative local recurrence. Conclusion The number of metastatic foci, including LNM and TD, in the LPLN area is useful for risk stratification of patients with low rectal cancer.
... In CRC patients, the presence of TD without lymph node metastasis is defined as stage N1c [14]. There is controversy in the medical community regarding the prognostic value of TD, as it is believed to be considered only in the absence of lymph node metastasis, potentially underestimating the severity of the disease [19,20]. Goldstein and Turner reported that TD as an independent adverse prognostic factor in CRC, should be distinguished from lymph node metastasis (LNM). ...
Article
Full-text available
Background Colorectal signet-ring cell carcinoma (SRCC) is a rare cancer with a bleak prognosis. The relationship between its clinicopathological features and survival remains incompletely elucidated. Tumor deposits (TD) have been utilized to guide the N staging in the 8th edition of American Joint Committee on Cancer (AJCC) staging manual, but their prognostic significance remains to be established in colorectal SRCC. Patients and methods The subjects of this study were patients with stage III/IV colorectal SRCC who underwent surgical treatment. The research comprised two cohorts: a training cohort and a validation cohort. The training cohort consisted of 631 qualified patients from the SEER database, while the validation cohort included 135 eligible patients from four independent hospitals in China. The study assessed the impact of TD on Cancer-Specific Survival (CSS) and Overall Survival (OS) using Kaplan-Meier survival curves and Cox regression models. Additionally, a prognostic nomogram model was constructed for further evaluation. Results In both cohorts, TD-positive patients were typically in the stage IV and exhibited the presence of perineural invasion (PNI) (P < 0.05). Compared to the TD-negative group, the TD-positive group showed significantly poorer CSS (the training cohort: HR, 1.87; 95% CI, 1.52–2.31; the validation cohort: HR, 2.43; 95% CI, 1.55–3.81; all P values < 0.001). This association was significant in stage III but not in stage IV. In the multivariate model, after adjusting for covariates, TD maintained an independent prognostic value (P < 0.05). A nomogram model including TD, N stage, T stage, TNM stage, CEA, and chemotherapy was constructed. Through internal and external validation, the model demonstrated good calibration and accuracy. Further survival curve analysis based on individual scores from the model showed good discrimination. Conclusion TD positivity is an independent factor of poor prognosis in colorectal SRCC patients, and it is more effective to predict the prognosis of colorectal SRCC by building a model with TD and other clinically related variables.
... Since the 5th edition, the definition of pT and pN has not changed substantially [23]. Changes in TNM stage groupings in CRC mainly affected the evaluation of tumour deposits, which occur only in a comparably small subset of CRCs (around 20%) [24]. ...
Article
Full-text available
Background Stroma AReactive Invasion Front Areas (SARIFA) is a novel prognostic histopathologic biomarker measured at the invasive front in haematoxylin & eosin (H&E) stained colon and gastric cancer resection specimens. The aim of the current study was to validate the prognostic relevance of SARIFA-status in colorectal cancer (CRC) patients and investigate its association with the luminal proportion of tumour (PoT). Methods We established the SARIFA-status in 164 CRC resection specimens. The relationship between SARIFA-status, clinicopathological characteristics, recurrence-free survival (RFS), cancer-specific survival (CSS), and PoT was investigated. Results SARIFA-status was positive in 22.6% of all CRCs. SARIFA-positivity was related to higher pT, pN, pTNM stage and high grade of differentiation. SARIFA-positivity was associated with shorter RFS independent of known prognostic factors analysing all CRCs (RFS: hazard ratio (HR) 2.6, p = 0.032, CSS: HR 2.4, p = 0.05) and shorter RFS and CSS analysing only rectal cancers. SARIFA-positivity, which was measured at the invasive front, was associated with PoT-low (p = 0.009), e.g., higher stroma content, and lower vessel density (p = 0.0059) measured at the luminal tumour surface. Conclusion Here, we validated the relationship between SARIFA-status and prognosis in CRC patients and provided first evidence for a potential prognostic relevance in the subgroup of rectal cancer patients. Interestingly, CRCs with different SARIFA-status also showed histological differences measurable at the luminal tumour surface. Further studies to better understand the relationship between high luminal intratumoural stroma content and absence of a stroma reaction at the invasive front (SARIFA-positivity) are warranted and may inform future treatment decisions in CRC patients.
... TDs refer to the discontinuous spread of tumors in the mesocolon/mesorectum. Studies in large cohorts have shown that TDs in combination with MLNs are the strongest predictor of PM (Lord et al. 2021;Nagtegaal et al. 2017;Ueno et al. 2023). To date, TDs without regional lymph node metastasis are classified as N1c, which does not utilize the metastatic and prognostic information provided by TDs. ...
Article
Full-text available
Background: Early detection and intervention could significantly improve the prognosis of patients with peritoneal metastasis (PM). Our main purpose was to develop a model to predict the risk of PM in patients with colorectal cancer (CRC). Methods: Patients from the Surveillance, Epidemiology, and End Results (SEER) database with CRC classified according to the AJCC 8th TNM staging system were selected for the study. After data pre-processing, the dataset was divided into a training set and a validation set. In the training set, univariate logistic analysis and stepwise multivariate logistic regression analysis were utilized to screen clinical features and construct a risk prediction model. Then, we validated the model using the confusion matrix, receiver operating characteristic (ROC) curves, decision curve analysis (DCA), and calibration curves to examine its performance. Results: The model constructed using stepwise multivariate logistic regression analysis incorporated the following eight clinical features: age, tumor location, histological type, T stage, carcinoembryonic antigen (CEA) level, tumor deposits (TDs), log odds (LODDS) of metastatic lymph nodes, and extraperitoneal metastasis (EM). The areas under the curve (AUCs) of the model in the training and validation sets were 0.924 and 0.912, respectively. The accuracy and the recall ratio were higher than 0.8 in both cohorts. DCA and the calibration curves also confirmed its excellent predictive power. Conclusions: Our model can effectively predict the risk of PM in CRC patients, which is of great significance for the timely identification of patients at high risk of PM and further clinical decision-making.
Article
The significance of discontinuous growth (DG) of the tumor to include tumor deposits and intramural metastasis in esophageal adenocarcinoma (EAC) is unclear. Esophagectomy specimens from 151 treatment-naïve and 121 treated patients with EAC were reviewed. DG was defined as discrete (≥2 mm away) tumor foci identified at the periphery of the main tumor in the submucosa, muscularis propria, and/or periadventitial tissue. Patients' demographics, clinicopathologic parameters, and oncologic outcomes were compared between tumors with DG versus without DG. DGs were identified in 16% of treatment-naïve and 29% of treated cases (P=0.01). Age, gender, and tumor location were comparable in DG+ and DG- groups. For the treatment-naïve group, DG+ tumors were larger with higher tumor grade and stage and more frequent extranodal extension, lymphovascular/perineural invasion, and positive margin. Patients with treated tumors presented at higher disease stages with higher rates of recurrence and metastasis compared with treatment-naïve patients. In this group, DG was also associated with TNM stage and more frequent lymphovascular/perineural spread and positive margin, but not with tumor size, grade, or extranodal extension. In multivariate analysis, in all patients adjusted for tumor size, lymphovascular involvement, margin, T and N stage, metastasis, neoadjuvant therapy status, treatment year, and DG, DG was found to be an independent adverse predictor of survival outcomes in EAC. DG in EAC is associated with adverse clinicopathologic features and worse patient outcomes. DG should be considered throughout the entire clinicopathologic evaluation of treatment-naïve and treated tumors as well as in future staging systems.
Article
Full-text available
Background: Based on the clinical data of colorectal cancer (CRC) patients who underwent surgery at our institution, a model for predicting the formation of tumor deposits (TDs) in this patient population was established. Aim: To establish an effective model for predicting TD formation, thus enabling clinicians to identify CRC patients at high risk for TDs and implement personalized treatment strategies. Methods: CRC patients (n = 645) who met the inclusion criteria were randomly divided into training (n = 452) and validation (n = 193) cohorts using a 7:3 ratio in this retrospective analysis. Least absolute shrinkage and selection operator regression was employed to screen potential risk factors, and multivariable logistic regression analysis was used to identify independent risk factors. Subsequently, a predictive model for TD formation in CRC patients was constructed based on the independent risk factors. The discrimination ability of the model, its consistency with actual results, and its clinical applicability were evaluated using receiver-operating characteristic curves, area under the curve (AUC), calibration curves, and decision curve analysis (DCA). Results: Thirty-four (7.5%) patients with TDs were identified in the training cohort based on postoperative pathological specimens. Multivariate logistic regression analysis identified female sex, preoperative intestinal obstruction, left-sided CRC, and lymph node metastasis as independent risk factors for TD formation. The AUCs of the nomogram models constructed using these variables were 0.839 and 0.853 in the training and validation cohorts, respectively. The calibration curve demonstrated good consistency, and the training cohort DCA yielded a threshold probability of 7%-78%. Conclusion: This study developed and validated a nomogram with good predictive performance for identifying TDs in CRC patients. Our predictive model can assist surgeons in making optimal treatment decisions.
Article
Full-text available
Backgraund. Unified terminology is a necessary condition for successful interdisciplinary communication in oncology. The variety of anatomical, pathomorphological and clinical terms used in rectal cancer (RC) is often accompanied by their ambiguous interpretation both in domestic and foreign scientific literature. This not only complicates the interaction between specialists, but also makes it difficult to compare the results of RC treatment obtained in different medical institutions. Aim. Develop domestic recommendations on a unified terminology of RC for diagnostic purposes and provide radiologists with a consensus methodological resource on the terminology, description and interpretation of RC imaging results. Materials and methods. Based on the analysis of recent domestic and international scientific and methodological literature on RC, the key terms used in the diagnosis and treatment planning of RC were selected, followed by a two-time online discussion of their interpretations by experts from the Russian Society of Radiologists and Therapeutic Radiation Oncologists, the Association of Oncologists of Russia and the Russian Association of Therapeutic Radiation Oncologists until reaching consensus (80%) of experts on all items. Terms for which there was no consensus were not included in the final list. Results. A list of anatomical, pathomorphological and clinical terms used in the diagnosis, staging and treatment planning of RC has been compiled and, based on expert consensus, their interpretation has been determined. Conclusion. A lexicon recommended in the description and formulation of the conclusion of diagnostic studies in patients with RC is proposed.
Article
Background: Although previous studies have demonstrated that tumor deposits (TDs) are associated with worse prognosis in colon cancer, their clinical significance in rectal cancer has not been fully elucidated, especially in the lateral pelvic lymph node (LPLN) area. This study aimed to clarify the clinical significance of TDs, focusing on the number of metastatic foci, including lymph node metastases (LNMs) and TDs, in the LPLN area. Methods: This retrospective study involved 226 consecutive patients with cStage II/III low rectal cancer who underwent LPLN dissection. Metastatic foci, including LNM and TD, in the LPLN area were defined as lateral pelvic metastases (LP-M) and were evaluated according to LP-M status: presence (absence vs. presence), histopathological classification (LNM vs. TD), and number (one to three vs. four or more). We evaluated the relapse-free survival of each model and compared them using the Akaike information criterion (AIC) and Harrell's concordance index (c-index). Results: Forty-nine of 226 patients (22%) had LP-M, and 15 patients (7%) had TDs. The median number of LP-M per patient was one (range, 1-9). The best risk stratification power was observed for number (AIC, 758; c-index, 0.668) compared with presence (AIC, 759; c-index, 0.665) and histopathological classification (AIC, 761; c-index, 0.664). The number of LP-M was an independent prognostic factor for both relapse-free and overall survival, and was significantly associated with cumulative local recurrence. Conclusion: The number of metastatic foci, including LNMs and TDs, in the LPLN area is useful for risk stratification of patients with low rectal cancer.
Article
Full-text available
The focus on lymph node metastases (LNM) as the most important prognostic marker in colorectal cancer (CRC) has been challenged by the finding that other types of locoregional spread, including tumor deposits (TDs), extramural venous invasion (EMVI), and perineural invasion (PNI), also have significant impact. However, there are concerns about interobserver variation when differentiating between these features. Therefore, this study analyzed interobserver agreement between pathologists when assessing routine tumor nodules based on TNM 8. Electronic slides of 50 tumor nodules that were not treated with neoadjuvant therapy were reviewed by 8 gastrointestinal pathologists. They were asked to classify each nodule as TD, LNM, EMVI, or PNI, and to list which histological discriminatory features were present. There was overall agreement of 73.5% (κ 0.38, 95%-CI 0.33–0.43) if a nodal versus non-nodal classification was used, and 52.2% (κ 0.27, 95%-CI 0.23–0.31) if EMVI and PNI were classified separately. The interobserver agreement varied significantly between discriminatory features from κ 0.64 (95%-CI 0.58–0.70) for roundness to κ 0.26 (95%-CI 0.12–0.41) for a lone arteriole sign, and the presence of discriminatory features did not always correlate with the final classification. Since extranodal pathways of spread are prognostically relevant , classification of tumor nodules is important. There is currently no evidence for the prognostic relevance of the origin of TD, and although some histopathological characteristics showed good interobserver agreement, these are often non-specific. To optimize interobserver agreement, we recommend a binary classification of nodal versus extranodal tumor nodules which is based on prognostic evidence and yields good overall agreement.
Article
Full-text available
PURPOSE As oxaliplatin results in cumulative neurotoxicity, reducing treatment duration without loss of efficacy would benefit patients and healthcare providers. PATIENTS AND METHODS Four of the six studies in the International Duration of Adjuvant Chemotherapy (IDEA) collaboration included patients with high-risk stage II colon and rectal cancers. Patients were treated (clinician and/or patient choice) with either fluorouracil, leucovorin, and oxaliplatin (FOLFOX) or capecitabine and oxaliplatin (CAPOX) and randomly assigned to receive 3- or 6-month treatment. The primary end point is disease-free survival (DFS), and noninferiority of 3-month treatment was defined as a hazard ratio (HR) of < 1.2- v 6-month arm. To detect this with 80% power at a one-sided type one error rate of 0.10, a total of 542 DFS events were required. RESULTS 3,273 eligible patients were randomly assigned to either 3- or 6-month treatment with 62% receiving CAPOX and 38% FOLFOX. There were 553 DFS events. Five-year DFS was 80.7% and 83.9% for 3-month and 6-month treatment, respectively (HR, 1.17; 80% CI, 1.05 to 1.31; P [for noninferiority] .39). This crossed the noninferiority limit of 1.2. As in the IDEA stage III analysis, the duration effect appeared dependent on the chemotherapy regimen although a test of interaction was negative. HR for CAPOX was 1.02 (80% CI, 0.88 to 1.17), and HR for FOLFOX was 1.41 (80% CI, 1.18 to 1.68). CONCLUSION Although noninferiority has not been demonstrated in the overall population, the convenience, reduced toxicity, and cost of 3-month adjuvant CAPOX suggest it as a potential option for high-risk stage II colon cancer if oxaliplatin-based chemotherapy is suitable. The relative contribution of the factors used to define high-risk stage II disease needs better understanding.
Article
Full-text available
Background The American Joint Committee on Cancer 8th classification states that colorectal cancer (CRC) is classified as N1c stage when regional lymph nodes (LNs) are negative and tumor deposits (TDs) are positive. However, how to classify TDs when regional LNs are positive remains unclear. The current study aimed to investigate the possibility of combining positive LNs and positive TDs to develop a modified pathological N (mpN) stage for CRC. Methods We retrospectively analyzed 9,198 patients with stage III CRC from the Surveillance, Epidemiology, and End Results program who underwent surgery (6,440 in the training cohort and 2,758 the validation cohort). The combination of positive LNs and TD status was defined as mpN stage. Overall survival (OS) according to mpN and pathological N (pN) stages was analyzed by the Kaplan–Meier method. The area under the curves (AUCs) and Akaike’s information criterion (AIC) were applied to assess the predictive discrimination abilities and goodness-of-fit of the model. The clinical benefits were measured using decision curve analyses. The validation cohort was used to validate the results. Results AUC analysis showed that the prognostic discrimination of mpN stage (AUC = 0.628, 95% confidence interval (CI), 0.616–0.640) was better than that of pN stage (AUC = 0.618, 95% CI, 0.606–0.630, p = 0.006) for OS. The AIC demonstrated that mpN stage (AIC = 30,217) also showed superior model-fitting compared with pN stage (AIC = 30,257) and decision curve analyses revealed that mpN stage had better clinical benefits than pN stage. Similar results were found in the validation cohort. Conclusions Among patients with CRC and LN metastasis, mpN stage might be superior to pN stage for assessing prognosis and survival, suggesting that TD status should be included in the pN stage.
Article
Full-text available
Background: The tumor-node-metastasis classification of the American Joint Committee on Cancer classified tumor deposits (TDs) in patients with colorectal cancer (CRC) without lymph node (LN) metastasis as N1c, but the classification of TDs in patients with LN metastases remains controversial. This study investigated the probability of regarding TDs as positive LNs (pLNs) in pN stage and estimated its prognostic ability in CRC. Methods: We used the Surveillance, Epidemiology, and End Results program to analyze CRC patients who underwent surgical therapy (14,906 training cohort, 6,384 validation cohort). A modified pN stage (mpN) was identified using the number of pLNs plus TDs. Overall survival (OS) was analyzed using the Kaplan–Meier survival curves, and significant prognostic factors were identified by univariate and multivariate analyses. Prognostic ability was estimated using the area under the curve (AUC), calibration curve, and the Akaike's information criterion (AIC). Clinical benefit was measured by the decision curve analyses (DCA). The results were validated using the validation cohort. Results: Both the pN and mpN stages were independent prognostic factors in CRC according to univariate and multivariate analyses. The AUC analysis showed that the mpN stage had better prognostic discrimination for OS than the pN stage (0.612 vs. 0.605, P < 0.001). The AIC demonstrated that the mpN stage also showed superior model-fitting compared with the pN stage (49,756 vs. 49,841). The DCA further revealed that the mpN stage had better clinical benefits than the pN stage. The validation cohort showed similar findings. Conclusions: We concluded that counting TDs as pLNs may be superior to the pN stage when assessing the prognosis of CRC patients.
Article
Full-text available
Purpose: To evaluate the role of tumor deposits (TDs) in predicting the efficacy of chemotherapy in stage III colon cancer. Methods: Using the SEER∗Stat software Version 8.3.6, we started with a national cohort of colon cancer cases diagnosed between 2004 and 2016. We used the χ2 (Chi-square) test to compare differences between different categorical variables according to the number of TDs. The Cox proportional hazards regression model was used to determine the independent association of different clinical and pathological variables with CSS, which were adjusted for other significant prognostic factors. Results: We have identified 29,017 patients diagnosed with stage III colon cancer from the SEER database. The results of multivariate analyses showed that patients with the receipt of chemotherapy had 54.7% decreased risk of cancer-specific mortality compared with those not (HR = 0.453, 95% CI = 0.425-0.483, P < 0.0001) in the no-TD group; In the 1-2-TD group, patients with the receipt of chemotherapy had 56.8% decreased risk of cancer-specific mortality compared with those not (HR = 0.432, 95% CI = 0.364-0.512, P < 0.0001); In the ≥3-TD group, patients with the receipt of chemotherapy had 51.8% decreased risk of cancer-specific mortality compared with those not (HR = 0.482, 95% CI = 0.389-0.597, P < 0.0001). Conclusions: Our study demonstrated that the presence of TDs was associated with a dismal prognosis and high number of TDs would also contribute to the worse survival of colon cancer. High number of TDs did not affect the survival benefit of chemotherapy in stage III colon cancer.
Article
Full-text available
Aim The prognostic value of the stage III subclassification system based on the Japanese Classification of Colorectal, Appendiceal, and Anal Carcinoma has not yet been clarified. This study aimed to develop a modified system with optimal risk stratification and compare its performance with the current staging systems. Methods Clinicopathological data from 6855 patients with stage III colorectal cancers who underwent D3 dissection were collected from a nationwide multicenter database. After determining patient survival rates across 13 divisions based on pathological N stage (N1, N2a, and N2b/N3) and tumor depth (T1, T2, T3, T4a, and T4b), except for T1N2a and T1N2b/N3 due to the small number, we categorized patients into three groups and developed a trisection staging system according to the Akaike information criterion. We then compared the Akaike information criterion of the developed system with those of the current staging systems. Results The T1N1[rank, 1] division (98.5%) had the most favorable prognosis in terms of 5‐year cancer‐specific survival, followed by T2N1[2] (93.9%), T2N2a[3] (92.0%), T3N1[4] (87.0%), T3N2a[5] (78.8%), T4aN1[6] (78.7%), T2N2b/N3[7] (77.8%), T4aN2a[8] (75.2%), T4bN1[9] (73.5%), T3N2b/N3[10] (64.7%), T4aN2b/N3[11] (61.5%), T4bN2b/N3[12] (43.0%), and T4bN2a[13] (42.5%). Compared to the categorizations of the Japanese and tumor‐node‐metastasis systems (Akaike information criterion, 22 684.6 and 22 727.1, respectively), the following stage categorizations were proven to be the most clinically efficacious: T1N1[1]‐T3N1[4], T3N2a[5]‐T4bN1[9], and T3N2b/N3[10]‐T4bN2a[13] (Akaike information criterion, 22 649.2). Conclusion The proposed modified system may be useful in the risk stratification of patients with stage III colorectal cancer who had undergone D3 dissection.
Article
Objective: We evaluated the prognostic value of TD counts and incorporated them with the number of positive lymph nodes to develop a revised nodal staging. Summary background data: The current American Joint Committee on Cancer (AJCC) staging on colon cancer includes the tumor deposits (TDs) only for node-negative patients, as N1c, and their counts are not considered. Methods: We included consecutive patients with stage III colorectal cancer who underwent curative resections between January 2010 and December 2019. The patients were grouped as TD 0, TD 1, TD 2, or TD ≥ 3 based on their TD counts. Disease-free survival and overall survival were compared. Results: Out of 2,446 eligible stage III patients, 658 (26.9%) had TDs. Among them, 500 (76.0%) patients concurrently had positive lymph nodes (LNs). TD counts were significantly related to worse DFS and OS regardless of pT stages or the number of positive LNs. The patients were restaged based on the integrated number of TD counts and LNs. The N3 stage, which had ten or more integrated TDs and positive LNs, was newly classified. Among the patients who completed six months of adjuvant chemotherapy, those upstaged to N2 from an initial stage of N1 experienced significantly worse DFS than those confirmed as N1, and it was comparable to N2 patients. The newly N3-staged patients showed significantly worse DFS than the patients initial staged as N2. Conclusions: Revised N staging using the integrated number of TD counts and positive LNs could predict DFS more accurately than current staging. It would also draw greater attention to high-risk stage III colon cancer staged as N3.
Article
Background: In colon cancer, tumor deposits (TD) are considered in assigning prognosis and staging only in the absence of lymph node metastasis (i.e., stage III pN1c tumors). We aimed at evaluating the prognostic value of the presence and the number of TD in patients with stage III, node-positive colon cancer. Patients and methods: All participants from the CALGB/SWOG 80702 phase III trial were included in this post hoc analysis. Pathology reports were reviewed for the presence and the number of TD, lymphovascular and perineural invasion. Associations with disease-free survival (DFS) and overall survival (OS) were evaluated by multivariable Cox models adjusting for gender, treatment arm, T-stage, N-stage, lymphovascular invasion, perineural invasion and lymph node ratio. Results: Overall, 2028 patients were included, with 524 (26%) TD-positive and 1504 (74%) TD-negative tumors. Of the TD-positive patients, 80 (15.4%) were node negative (i.e., pN1c), 239 (46.1%) were pN1a/b (<4 positive lymph nodes) and 200 (38.6%) were pN2 (≥4 positive lymph nodes). The presence of TD was associated with poorer DFS (adjusted hazard ratio (aHR)= 1.63, 95%CI 1.33-1.98) and OS (aHR = 1.59, 95%CI 1.24-2.04). The negative effect of TD was observed for both pN1a/b and pN2 groups. Among TD-positive patients, the number of TD had a linear negative effect on DFS and OS. Combining TD and the number of lymph node metastases, 104 of 1470 (7.1%) pN1 patients were re-staged as pN2, with worse outcomes than patients confirmed as pN1 (3-year DFS rate: 80.5% versus 65.4%, P=.0003; 5-year OS rate: 87.9% versus 69.1%, P=<0.0001). DFS was not different between patients re-staged as pN2 and those initially staged as pN2 (3-year DFS rate: 62.3% versus 65.4%, P=.4895). Conclusions: Combining the number of TD and the number of lymph node metastases improved the prognostication accuracy of TNM staging.
Article
Introduction The prognostic role of the N1c remains unclear in colorectal cancer (CRC). Our study aimed to determine the prognostic value of N1c. Methods Patients diagnosed in 2010-2015 were accessed from the Surveillance, Epidemiology, and End Results (SEER) database. COX univariate and multivariate regression analysis and the Kaplan-Meier method were used to assess the impact of the N1c stage on the cause-specific (CSS) and overall survival (OS). Propensity score matching (PSM) was used to construct a matched group with similar propensity scores. Results Kaplan-Meier analysis showed that the CSS and OS rates in N1a were significantly better than N1c in stage III and IV CRCs after reducing selection bias (CSS: P < 0.001 in stage III, P = 0.041 in stage IV; OS: P < 0.001 in stage III, P = 0.0079 in stage IV). There were no statistical differences in CSS and OS between N1b and N1c (CSS: P = 0.500 in stage III, P = 0.270 in stage IV; OS: P = 0.390 in stage III, P = 0.600 in stage IV). Further, the prognostic value of N1c with only one tumor deposit (TD) is equivalent to N1a based on the comparison of CSS and OS rates (CSS: P = 0.420; OS: P = 0.310). Whereas N1c with only one TD had significantly better CSS and OS than N1b (CSS: P = 0.039; OS: P = 0.037). Conclusion The CSS and OS rates of N1c do not achieve a statistical difference with N1b in both stage III and IV CRCs. Significantly, higher CSS and OS rates were found in N1c with only one TD versus N1b stage in stage III CRC.
Article
PURPOSE Tumor deposits (TDs) seem to affect the prognosis of patients with colon cancer (CC). In the seventh edition of the American Joint Committee on Cancer TNM staging system for CC, the presence of TDs is only considered in the absence of lymph node metastases (LNMs). In the era of personalized duration of histopathologic criteria-based adjuvant therapy, this could potentially lead to a decrease in the prognostic prediction accuracy. PATIENTS AND METHODS A post hoc analysis of all pathologic reports from patients with stage III CC included in the IDEA France phase III study (ClinicalTrials.gov identifier: NCT00958737 ) investigating the duration of adjuvant fluorouracil, leucovorin, and oxaliplatin or capecitabine and oxaliplatin therapy (3 v 6 months) was performed. The primary objective was to determine the prognostic impact of TD on disease-free survival (DFS). The effect of the addition of TD to LNM count on pN restaging was also evaluated. A multivariable analysis was performed to establish the association between TD and DFS. RESULTS Of 1,942 patients, 184 (9.5%) had TDs. The pN1a/b and pN1c populations showed similar DFS. TD-positive patients had worse prognosis compared with TD-negative patients, with 3-year DFS rates of 65.6% (95% CI, 58.0% to 72.1%) and 74.7% (95% CI, 72.6% to 76.7%; P = .0079), respectively. On multivariable analysis, TDs were associated with a higher risk of recurrence or death (hazard ratio [HR], 1.36; P = .0201). Other adverse factors included pT4 and/or pN2 disease (HR, 2.21; P < .001), the 3 months of adjuvant treatment (HR, 1.29; P = .0029), tumor obstruction (HR, 1.28; P = .0233), and male sex (HR, 1.24; P = .0151). Patients restaged as having pN2 disease (n = 35, 2.3%) had similar DFS as patients initially classified as pN2. CONCLUSION The presence of TDs is an independent prognostic factor for DFS in patients with stage III CC. The addition of TD to LNM may help to better define the duration of adjuvant therapy.