You are on page 1of 7

Human Pathology (2014) 45, 2430–2436

www.elsevier.com/locate/humpath

Original contribution

Immunohistochemical detection of ARID1A in


colorectal carcinoma: loss of staining is associated
with sporadic microsatellite unstable tumors with
medullary histology and high TNM stage☆,☆☆
Jiqing Ye MD a , Yi Zhou MD a , Martin R. Weiser MD b , Mithat Gönen PhD c ,
Liying Zhang MD a , Tushar Samdani MD b , Ruben Bacares BS a , Deborah DeLair MD a ,
Sinisa Ivelja MD a , Efsevia Vakiani MD, PhD a , David S. Klimstra MD a ,
Robert A. Soslow MD a , Jinru Shia MD a,⁎
a
Department of Pathology, Memorial Sloan–Kettering Cancer Center, 12705 York Ave, New York, NY, 10065
b
Department of Surgery, Memorial Sloan–Kettering Cancer Center, 12705 York Ave, New York, NY, 10065
c
Department of Biostatistics, Memorial Sloan–Kettering Cancer Center, 12705 York Ave, New York, NY, 10065

Received 3 June 2014; revised 8 August 2014; accepted 13 August 2014

Keywords:
Summary AT-rich interactive domain-containing protein 1A (ARID1A), a chromatin remodeling gene
Colon cancer;
recently discovered to be a tumor suppressor in ovarian cancers, has been found to be mutated at low
Microsatellite instability;
frequencies in many other tumors including colorectal carcinoma (CRC). An association between ARID1A
Immunohistochemistry;
alteration and DNA mismatch repair (MMR) deficiency has been implicated; understanding this association
MLH1 and PMS2;
may facilitate the understanding of the role of ARID1A in the various tumors. In this pilot study, we analyzed
MLH1 methylation
the immunohistochemical expression of ARID1A in a consecutive series of 257 CRCs that fulfilled a set of
relaxed criteria for Lynch syndrome screening; 59 (23%) were MMR deficient by immunohistochemistry
(44 MLH1/PMS2 deficient, 9 MSH2/MSH6 deficient, 4 MSH6 deficient, and 2 PMS2 deficient). ARID1A
loss was observed in 9% (22/257) of the cohort: 24% of MMR-deficient tumors (14/59, 13 of the 14 being
MLH1/PMS2 deficient) and 4% of MMR-normal tumors (8/198) (P b .05). MLH1 (mutL homolog 1)
promoter hypermethylation was observed in 10 of the 13 MLH1/PMS2-deficient/ARID1A-loss tumors,
indicating an association between ARID1A loss and sporadic microsatellite unstable CRCs. Among the
MMR-deficient cases, ARID1A loss correlated with old age (P = .04), poor tumor differentiation (P b .01),
medullary histology (P b .01), and an increased rate of nodal and distant metastasis (P = .03); these patients
also trended toward a worse 5-year overall survival. Among MMR-normal tumors, no differences in
clinicopathological features were detected between the groups stratified by ARID1A. In conclusion, our
results suggest that ARID1A loss may be linked to a specific subset of sporadic microsatellite unstable CRCs
that may be medullary but is more likely to present with metastatic disease, warranting further investigation.
© 2014 Elsevier Inc. All rights reserved.


This study was presented in part at the 102nd annual meeting of the United States and Canadian Academy of Pathology in Baltimore, MD, in March 2013.
☆☆
Disclosures: The authors declare no conflict of interest.
⁎ Corresponding author.
E-mail address: Shiaj@mskcc.org (J. Shia).

http://dx.doi.org/10.1016/j.humpath.2014.08.007
0046-8177/© 2014 Elsevier Inc. All rights reserved.
ARID1A in colorectal carcinoma 2431

1. Introduction criteria: (1) age 50 years or younger, (2) age older than
50 years and the tumor showed MSI-H histology as defined
AT-rich interactive domain-containing protein 1A by the Revised Bethesda guidelines [11], and (3) family and/or
(ARID1A) (also known as BAF250a), encoded by the AR- personal history meeting the Revised Bethesda guidelines as
ID1A gene, is a key component of the large adenosine determined by the treating physician. The patients were treated
triphosphate–dependent chromatin remodeling complex at Memorial Sloan–Kettering Cancer Center from 2007 to
SWItch/Sucrose NonFermentable (SWI/SNF) [1]. Owing 2009. During this period, colorectal cancers from patients
to the advanced sequencing techniques, this gene was first fulfilling any 1 of the above 3 criteria were routinely tested
found to be mutated frequently in certain carcinomas, with immunohistochemical stains for MLH1, MSH2, MSH6,
specifically, ovarian clear cell [2,3] and endometrioid [3] and PMS2. The patients' clinical information was retrieved
carcinomas and uterine endometrioid carcinomas [4]. In these from the hospital information system. The study was approved
tumors, ARID1A is believed to act as a tumor suppressor gene by the institutional board review.
[2-6]. Subsequently, mutations in this gene have been found to
occur in small subsets of carcinomas from a wide variety of 2.2. Tumor pathology
organ sites, including colon and rectum, stomach, pancreas,
liver, prostate, breast, and lung [7]. All study cases were systematically reviewed for the
Studies have suggested that, in at least some tumor types, following pathologic features: (1) tumor location (right
ARID1A mutation is associated with microsatellite instability colon referring to cecum, ascending and transverse colon,
(MSI) [7-9]. Jones et al [7] reported that, among gastric, colon, and left colon referring to descending and sigmoid colon)
prostate, and pancreatic carcinomas, the most commonly and (2) histologic grade (low grade referring to well or
observed mutations in ARID1A occurred in a coding moderately differentiated adenocarcinoma and high grade
microsatellite, specifically, a 7-base G tract in the coding referring to poorly differentiated adenocarcinoma with
region; and 12 of 12 MSI-high (MSI-H) cancers (6 colon, 5 b50% of the tumor showing gland formation) [12].
gastric, and 1 prostate) that had ARID1A mutation had their Medullary histology represented by solid growth with
mutations at mononucleotide tracts in the gene. Interestingly, a prominent intratumoral and stromal lymphocytes [13] was
recent study [6] indicated that, in the case of endometrial considered to be “high grade,” its presence in greater than or
carcinoma, loss of ARID1A protein was significantly more equal to 25% of the tumor was recorded as “medullary
prevalent in sporadic MSI-H cancers with MLH1 promoter component present,” and its presence in greater than or
hypermethylation than in Lynch-associated tumors (75% equal to 50% of the tumor qualified the tumor for
versus 14%, P b .0001). Such developments have prompted “medullary carcinoma”; (3) tumor-infiltrating lymphocytes
the speculation that ARID1A mutation may drive endometrial (TILs) (count per 10 high-power fields); and (4) tumor stage
carcinogenesis toward MSI through epigenetic alterations of (as defined by the seventh edition of American Joint
the MLH1 gene. Committee on Cancer Cancer Staging Manual [12]).
Mutation of ARID1A has been identified as a recurrent event
in colorectal carcinoma (CRC) in the recent Cancer Genome
2.3. Tissue microarray
Atlas project (TCGA) data [10]. Its specific role in colorectal
tumor development and progression, however, remains to be
Representative tumor blocks were selected from each of the
determined. Although it has been suggested that an association
study cases and used for construction of tissue microarray
between ARID1A alteration and MSI might exist in CRC as
(TMA). The ATA-27 automated arrayer (Beecher Instruments,
well [7,10], this has not been systematically studied. Thus, in this
Sun Prairie, WI, USA) was used to create the microarrays. The
pilot study, we evaluated the expression of ARID1A in a series
punch size was set at 0.6 mm. Three cores of different areas of
of CRCs stratified by the expression status of DNA mismatch
the tumor were sampled from each tumor specimen.
repair (MMR) proteins and MLH1 promoter methylation in
selected cases and explored the clinicopathological characteris-
tics associated with abnormal tumor expression of ARID1A. 2.4. Immunohistochemistry
Our study provided interesting findings that may serve to guide
further investigations that aim at understanding the biological Immunohistochemistry (IHC) for MLH1, PMS2, MSH2,
and clinical significance of ARID1A alteration in CRC. and MSH6 was performed on whole tumor sections of each
patient's resection specimen, using standard streptavidin-
biotin-peroxidase procedure. Primary monoclonal antibodies
2. Materials and methods used included MLH1 (clone G168-728, diluted 1:250; BD
PharMingen, San Diego, CA), MSH2 (clone FE11, diluted
2.1. Patient information 1:50; Oncogene Research Products, Cambridge, MA), MSH6
(clone GRBP.P1/2.D4, diluted 1:200; Serotec, Raleigh, NC),
Study cases were identified from a consecutive series of and PMS2 (clone A16-4, diluted 1:200; BD PharMingen).
CRC patients who fulfilled at least one of the following Nonneoplastic colonic mucosa and colorectal tumors known
2432 J. Ye et al.

to be deficient of MLH1, MSH2, MSH6, and PMS2 were the various subgroups were documented. The clinical and
used as external positive and negative controls, respectively. pathologic features were comparatively analyzed when
Abnormal MMR IHC was defined as complete absence of feasible. Continuous data were described as mean and SE
nuclear staining within tumor cells in the presence of or median and range as appropriate. Categorical data were
concurrent positive labeling in internal nonneoplastic tissues. described as numbers and percentages. The significance of
IHC for ARID1A was performed on the TMA slides using a differences in categorical variables between groups was
polyclonal antibody against ARID1A (HPA005456; Sigma- tested by the Fisher exact tests. Survival probabilities were
Aldrich, St. Louis, MO, USA) [4] and on the Ventana, Tucson, estimated by the method of Kaplan-Meier and compared
AZ, USA Discovery XT. The concentration of the polyclonal using the log-rank test. Statistical significance was defined
antibody was 0.2 mg/mL. The dilution factor of this antibody as P b .05.
was optimized by using liver parenchyma as negative control
and tonsil tissue as positive control as recommended. In addition
to antibody concentration, we also optimized target retrieval
buffer. After optimizing the IHC conditions, the TMA slides 3. Results
were deparaffinized. Target retrieval was performed in standard
CC1 slightly basic buffer. The primary antibody was diluted by A total of 302 CRCs from 301 patients were included in the
1:100 with a final concentration of 0.2 μg/mL. The incubation TMA. Of them, 45 cases (15%) were excluded due to tissue
time for the primary antibody was 60 minutes. The slides were loss, leaving 257 analyzable cases obtained from 256 patients.
washed, followed by incubation with the secondary antibody Of the 257 tumors, 59 (23%) had abnormal MMR IHC
and final development with the 3,3′-Diaminobenzidine Map Kit (Table 1). The abnormal patterns were as follows: 44 tumors
(Ventana, catalog no. 750-169) for 60 minutes. Abnormal losing MLH1/PMS2, 2 tumors losing PMS2 alone, 9 tumors
ARID1A IHC was defined as complete loss of nuclear staining in losing MSH2/MSH6, and 4 tumors losing MSH6 alone.
the tumor. Conversely, normal ARID1A IHC included cases The staining for ARID1A was easily interpretable (Fig.).
with positive nuclear staining, including cases with weak Normal staining was detected in 235 cases (235/257, 91%);
staining (intensity comparable with stromal cells) and strong 55% of which (129/235) showed a strong intensity, whereas
staining (intensity stronger than stromal cells). Cases with no the remaining 45% (106 cases) had a weaker but easily
analyzable core in the TMA were excluded from final analysis. discernible intensity. Abnormal ARID1A IHC defined as loss
The minimal requirement for a case to be regarded as analyzable of tumor cell staining was detected in 9% (22/257) of the
was the presence of at least 1 intact core. series; all abnormal cases had discernible positive internal
control. Some variation in staining intensity (moderate versus
2.5. MLH1 promoter methylation strong or moderate versus weak) was noted in a small subset of
positively stained cases. Overall, we did not encounter
significant staining heterogeneity among different cores.
For each case, a 5-micron section of formalin-fixed,
Abnormal ARID1A was significantly more prevalent in
paraffin-embedded tumor tissue was stained by hematoxylin
the MMR-abnormal group than in the MMR-normal group
and eosin to identify areas of tumor on the slide. Dissection
(14/59 or 24% versus 8/198 or 4%, P b .05). Conversely,
of the tumor area was carried out from the corresponding
abnormal MMR was significantly more common in the
unstained sections. The tumor tissue was deparaffinized, and
ARID1A-abnormal group than in the ARID1A-normal
DNA extraction was performed using the Qiagen DNeasy
group (14/22 or 64% versus 45/236 or 19%, P b .05).
Tissue Kit (cat no. 69504; Qiagen, Valencia, CA, USA).
Overall, a total of 14 cases (14/257, 5%) were abnormal in
Tumor DNA was amplified to test for the presence of
both MMR and ARID1A; of the 14 cases, 13 (93%) were
hypermethylation of 5 CpG sites within the MLH1 promoter
(in region −209 to −181 from the transcription start site). The
polymerase chain reaction product was subject to pyrose-
quencing on a Pyromark Q24 pyrosequencer. Because Table 1 Number of colorectal carcinoma cases showing
normal versus abnormal immunohistochemical expression of
bisulfite treatment converts unmethylated Cs to Ts, the
DNA MMR proteins and ARID1A a
degree of methylation for each CpG site was calculated as
follows: methylation % = peak height of methylated (C)/ ARID1A IHC b MMR IHC b
(peak height of methylated [C] + peak height nonmethylated Abnormal Normal Total
[T]) × 100. Methylation status was graded as “present,” when
Abnormal 14 (24%) 8 (4%) 22 (9%)
all 5 CpG sites were methylated above 10%. Normal 45 (76%) 190 (96%) 235 (91%)
Total 59 198 257
2.6. Statistical analysis a
P b.05.
b
For IHC for both MMR proteins and ARID1A, “abnormal”
Cases were categorized into subgroups based on staining indicates no nuclear staining in the tumor cells, and “normal” indicates
the presence of nuclear staining in the tumor cells.
results for MMR proteins and ARID1A. The frequencies of
ARID1A in colorectal carcinoma 2433

Fig. Photomicrographs of 2 colonic adenocarcinomas with different ARID1A immunohistochemical staining results. One is a moderately
differentiated adenocarcinoma (A) and shows positive nuclear labeling for ARID1A in both the tumor cells and the background nonneoplastic
cells (B). The second tumor is a medullary carcinoma (C) and shows loss of ARID1A staining in the tumor cells, whereas the background
nonneoplastic cells are positively stained (D) (original magnification, hematoxylin and eosin ×100 [A-D]).

abnormal in MLH1/PMS2 with only 1 case showing a ed promoter hypermethylation (as defined in the Methods
different abnormal pattern (loss of MSH6 alone). In contrast, section). All 10 cases had loss of both MLH1 and PMS2 by
31% (14/45) of the MMR-abnormal/ARID1A-normal cases IHC. Three MLH1/PMS2-abnormal cases (aged 36, 65, and 79
showed an MMR abnormal pattern different from loss of years, respectively) and 1 MSH6-abnormal case (age 50 years)
MLH1/PMS2. This is outlined in Table 2. did not exhibit MLH1 promoter methylation.
MLH1 promoter methylation analysis was performed on the The clinicopathological features of the various subgroups
14 ARID1A-abnormal/MMR-abnormal cases; 10 demonstrat- of the cases are summarized in Table 3.
Among MMR-abnormal cases, the group that also had
ARID1A abnormality showed an average age 11 years
older than the group with normal ARID1A (mean, 69.6
Table 2 Patterns of MMR-protein abnormality in colorectal
carcinomas with and without abnormal ARID1A a versus 58.4 years; median, 73.5 versus 62 years, P = .04).
When compared with the group with normal ARID1A, the
Patterns of MMR ARID1A b ARID1A-abnormal group tended to have tumors with
IHC abnormality b Abnormal Normal Total high histologic grade (11/14 or 79% versus 10/44 or 23%,
Loss of MLH1/PMS2 13 (93%) 31 (69%) 44 (75%) P b .01) and of medullary type, that is, having a
Other patterns medullary component in greater than or equal to 50% of
Loss of PMS2 0 2 2 the tumor (8/14 or 43% versus 5/45 or 11%, P b .01),
Loss of MSH2/MSH6 0 9 9 presented at later TNM stages (stages III/IV, 7/14 or 50%
Loss of MSH6 1 3 4 versus 8/45 or 18%, P = .03), and exhibited a trend (not
Subtotal 1 (7%) 14 (31%) 15 (25%) statistically significant) toward a worse 5-year overall
Total no. of cases 14 45 59 survival (68% versus 80%, P = .23) with a median
a
P = .08. follow-up time of 57 months (range, 1-138 months). TILs
b
For IHC for both MMR proteins and ARID1A, “abnormal” were increased in both ARID1A-normal and abnormal
indicates no nuclear staining in the tumor cells, and “normal” indicates
groups with no significant difference in the numbers of
the presence of nuclear staining in the tumor cells.
TILs/10 high-power fields.
2434 J. Ye et al.

Table 3 Correlation between immunohistochemical expression of MMR protein and ARID1A and clinicopathological features a
MMR IHC abnormal MMR IHC normal
ARID1A ARID1A
Total, Abnormal, Normal, P Total, Abnormal, Normal, P
n = 59 n = 14 n = 45 n = 198 a n=8 n = 190
Age (y)
Mean 61.0 69.6 58.4 .04 47.7 48.8 47.6 .77
Median 64.0 73.5 62 47 49.5 47
Range 29-89 36-89 29-85 18-86 44-53 18-86
Sex (n)
Male 27 5 22 .54 105 b 5 100 .72
Female 32 9 23 93 3 90
Tumor location (n)
Right colon 50 12 38 .69 55 0 55 .11
Left colon 7 1 6 58 5 53
Rectum 2 1 1 85 3 82
Histology grade (n)
Low 37 3 34 b.01 179 8 171 1.00
High 21 11 10 14 0 14
NA 1 0 1 5 0 5
Medullary carcinoma c (n) 13 8 5 b.01 1 0 1 1.00
TIL (count per 10 HPFs)
Mean 200 211 191 .44 14 6 14 .43
Median 198 231 173 4 1 4
Range 0-680 6-560 0-680 0-247 0-30 0-247
Tumor stage (n)
I and II 44 7 37 .03 84 3 81 1.00
III and IV 15 7 8 114 5 109
5-Year overall survival rates (%) 77% 68% 80% .23 66% 83% 65% .87
Abbreviations: NA, not available; TIL, tumor-infiltrating lymphocytes; HPFs, high-power fields.
a
For IHC for both MMR proteins and ARID1A, “abnormal” indicates no nuclear staining in the tumor cells and “normal” indicates the presence of
nuclear staining in the tumor cells.
b
One patient had 2 synchronous cancers.
c
Defined as having greater than or equal to 50% of the tumor showing medullary pattern.

In contrast, among the MMR-normal cases, no apparent ARID1A loss correlated with the following clinicopatholog-
difference was detected between the groups stratified by the ical features: old age (on average, approximately 10 years
ARID1A status in any of the features examined (Table 3). older than ARID1A-normal cases); higher tumor histologic
grade; medullary histology; and a more advanced TNM
stage, with a trend (not reaching statistical significance)
toward a worse 5-year overall survival.
4. Discussion The incidence of ARID1A loss in colorectal cancers in
general remains to be determined. Our study population was
This study analyzed a specific population of colorectal biased toward patients with increased risk for Lynch
cancer patients that was considered to be at increased risk for syndrome; thus, the observed rate of 9% may not represent
Lynch syndrome based on the Bethesda guidelines. The rate the general colorectal cancer patient population. Interesting-
of MMR protein loss by IHC was 23%. In such a population, ly, however, this rate is very close to the 10% of colorectal
we identified ARID1A loss (defined as loss of IHC staining cancer cases showing somatic mutations in ARID1A reported
in the tumor cells) in 9% of the cases. Our results further by Jones et al [7] and suggests that, although the subset of
indicated that (1) there existed a tight association between colorectal cancers that has ARID1A abnormality might be
ARID1A loss and MMR deficiency in CRC, with most small, it is not negligible. Current concepts hold that driver
ARID1A-abnormal tumors being MLH1/PMS2 deficient genes frequently present in 1 tumor entity often turn out to be
and showing MLH1 promoter hypermethylation, although significant in small subsets of other tumor types as well.
notably, ARID1A loss was observed in an MSH6-deficient Given that ARID1A is shown to be a driver gene that is
tumor as well; and (2) among the cases with abnormal MMR, frequently altered in ovarian carcinomas [2], the observation
ARID1A in colorectal carcinoma 2435

that it is abnormal in a small subset of colorectal cancers as carcinomas as well, a type of CRC first described by
well suggests that this gene also bears the potential to be Jessurun et al [16] and incorporated by the World Health
important in these colorectal cancers. This is of value and Organization classification. We observed that a significant
deserves our attention. subset (43%) of MMR-abnormal/ARID1A-abnormal tumors
Our finding of a significantly higher frequency of was of medullary type, with greater than 50% of the tumor
ARID1A loss in MMR-deficient CRCs than in MMR- having medullary histology, whereas only 11% of the MMR-
normal CRCs is in line with the published literature abnormal/ARID1A-normal tumors were of medullary type.
[7,10,14]. Adding to the existing literature, we further In general, although medullary carcinomas are morpholog-
observed that the MMR-deficient CRCs that show ARID1A ically similar to poorly differentiated/undifferentiated ade-
loss are primarily deficient in MLH1/PMS2 and harbor nocarcinoma, they are associated with lymphocytic
MLH1 promoter hypermethylation, that is, sporadic MSI infiltration and frequently associated with MSI [17,18].
cancers, and these patients, not surprisingly, are on average Clinically, both medullary carcinoma and MSI carcinomas of
approximately 10 years older than patients with MMR- other histologic subtypes are believed to have a better
abnormal/ARID1A-normal tumors. In contrast, of the MMR- prognosis [16,19,20] and be less likely to present with nodal
deficient/ARID1A-normal group, more than 30% of the involvement or advanced N or M stage [21]. For this reason,
tumors were MMR deficient in patterns other than concurrent the World Health Organization recommends that all MSI
loss of MLH1/PMS2, suggesting mechanisms other than CRCs be regarded as low grade, irrespective of their
MLH1 promoter hypermethylation. Unfortunately, however, histologic grade [22]. On the other hand, however, there
detailed genetic data were not available. are literature data showing that (1) there exist certain
A significant association between abnormal ARID1A and medullary carcinomas that fare worse than their poorly
sporadic MSI with MLH1 promoter methylation has indeed differentiated counterparts [23], and (2) some MSI cancers
been reported in endometrial cancers [6]. There, it has been behave more aggressively than others [24]. In this context,
speculated that ARID1A loss might have resulted in our finding of the MSI cancers that were ARID1A abnormal
epigenetic alterations by a deficient SWI/SNF complex showing overrepresentation of medullary histology yet
with subsequent MLH1 promoter methylation, or that the unexpected high TNM staging may serve to indicate that
loss of both ARID1A and MLH1 may be a reflection of ARID1A abnormality may be linked to a specific subset of
global genomic hypermethylation, that is, the CpG island sporadic MSI cancers that is biologically more virulent. As
methylator phenotype. This is a tenable hypothesis because such, ARID1A loss may bear prognostic significance.
ARID1A has been shown to harbor CpG islands in its Notably, ARID1A loss has been associated with tumor
promoter, and ARID1A promoter hypermethylation has indeed grade and stage in other organs as well. In a study of ovarian
been described in breast carcinoma in a recent report [15]. clear cell carcinoma, Katagiri et al [25] reported an
These hypotheses are likely applicable to colorectal tumors as association between ARID1A loss and advanced Interna-
well given the findings of our study. However, they remain to tional Federation of Gynecology and Obstetrics (FIGO) stage
be verified. To this end, it is particularly worth noting that loss and a shorter progression-free survival. Furthermore, their
of ARID1A protein has been observed to be associated with study suggested that tumors with loss of ARID1A were more
mutations of the gene [4], which would conflict with the likely to be resistant to platinum-based chemotherapy.
mechanism of promoter methylation. This, coupled with our Similar adverse prognostic implication of ARID1A loss
finding of abnormal ARID1A in an MSH6-deficient/MLH1 has also been observed in endometrial [26-28] and gastric
methylation–negative tumor, suggests that ARID1A alteration [29] cancers. Notably, however, inconsistent reports also
in tumors may be mediated by heterogeneous mechanisms. exist. For example, in an analysis of 111 uterine endome-
Very interestingly, our results indicated that, among trioid carcinomas, Rahman et al [30] failed to detect any
MMR-deficient CRCs, those that were also ARID1A significant association between ARID1A loss and the
abnormal not only had MLH1 promoter hypermethylation common clinicopathological features including FIGO stage
and were from significantly older patients, they also had and grade. In our study, we also failed to demonstrate a
other distinct pathologic features when compared with cases statistically significant difference in the 5-year overall
with normal ARID1A. First, these ARID1A-abnormal survival between ARID1A-normal and ARID1A-abnormal
tumors were of higher histologic grade and more frequently cases. The limited number of ARID1A-abnormal cases may
of medullary type. Second and more importantly, these cases have limited our survival analysis. We did indeed observe a
were more likely to present at an advanced TNM stage. trend toward abnormal ARID1A associating with a worse
These findings are of interest and engender new speculations outcome among the MMR-abnormal cases. In addition to the
with regard to the prognostic implications of medullary limitations caused by limited number of cases, the discrepant
histology, MSI, and ARID1A in CRC. results among the literature studies may also be due to
In this study, we defined histologic grade according to the differences in organ- and histology type–specific oncogenic
conventional criteria, that is, tumors with greater than 50% pathways. Attention toward such organ- and histology type–
showing solid growth or lacking gland formation were specific differences is warranted, as efforts on understanding
considered high grade. This was applied to medullary the role of ARID1A in various tumors are ongoing,
2436 J. Ye et al.

In conclusion, our study indicates that a small but [13] Shia J, Ellis NA, Paty PB, et al. Value of histopathology in predicting
significant subset of CRCs shows abnormal ARID1A, and microsatellite instability in hereditary nonpolyposis colorectal cancer
and sporadic colorectal cancer. Am J Surg Pathol 2003;27:1407-17.
most of these tumors are sporadic MSI cancers with MLH1 [14] Cajuso T, Hanninen UA, Kondelin J, et al. Exome sequencing reveals
promoter hypermethylation and loss of both MLH1and PMS2. frequent inactivating mutations in ARID1A, ARID1B, ARID2, and
When compared with ARID1A-normal MSI cancers, ARID4A in microsatellite unstable colorectal cancer. Int J Cancer
ARID1A-abnormal MSI cancers tend to present at a more 2013;135:611-23.
[15] Zhang X, Sun Q, Shan M, et al. Promoter hypermethylation of
advanced TNM stage, suggesting that ARID1A loss might
ARID1A gene is responsible for its low mRNA expression in many
identify a specific subset of MSI CRCs that is biologically invasive breast cancers. PLoS One 2013;8:e53931.
more aggressive. These findings warrant further investigation. [16] Jessurun J, Romero-Guadarrama M, Manivel JC. Medullary adeno-
Notably, it has been speculated that the role of ARID1A and carcinoma of the colon: clinicopathologic study of 11 cases. HUM
other related members of the SWI/SNF complex in tumors PATHOL 1999;30:843-8.
[17] Greenson JK, Huang SC, Herron C, et al. Pathologic predictors of
may be exploited for the development of novel therapeutic
microsatellite instability in colorectal cancer. Am J Surg Pathol 2009;
strategies [1]; as such strategies become available, detection of 33:126-33.
the ARID1A protein by IHC may also find application in [18] Shia J, Holck S, Depetris G, et al. Lynch syndrome-associated
predicting tumor response to target-selective agents. neoplasms: a discussion on histopathology and immunohistochemis-
try. Fam Cancer 2013;12:241-60.
[19] Thibodeau SN, Bren G, Schaid D. Microsatellite instability in cancer
of the proximal colon. Science 1993;260:816-9.
References [20] Guastadisegni C, Colafranceschi M, Ottini L, et al. Microsatellite
instability as a marker of prognosis and response to therapy: a meta-
[1] Oike T, Ogiwara H, Nakano T, et al. Inactivating mutations in SWI/ analysis of colorectal cancer survival data. Eur J Cancer 2010;46:
SNF chromatin remodeling genes in human cancer. Jpn J Clin Oncol 2788-98.
2013;43:849-55. [21] Lanza G, Gafa R, Matteuzzi M, et al. Medullary-type poorly
[2] Jones S, Wang TL, Shih Ie M, et al. Frequent mutations of chromatin differentiated adenocarcinoma of the large bowel: a distinct clinico-
remodeling gene ARID1A in ovarian clear cell carcinoma. Science pathologic entity characterized by microsatellite instability and
2010;330:228-31. improved survival. J Clin Oncol 1999;17:2429-38.
[3] Wiegand KC, Shah SP, Al-Agha OM, et al. ARID1A mutations in [22] Hamilton SR, Aaltonen LA. Pathology and Genetics of Tumours of the
endometriosis-associated ovarian carcinomas. N Engl J Med 2010; Digestive System. World Health Organization Classification of
363:1532-43. Tumors. Lyon: IARC Press; 2000.
[4] Guan B, Mao TL, Panuganti PK, et al. Mutation and loss of expression [23] Thirunavukarasu P, Sathaiah M, Singla S, et al. Medullary carcinoma
of ARID1A in uterine low-grade endometrioid carcinoma. Am J Surg of the large intestine: a population based analysis. Int J Oncol 2010;37:
Pathol 2011;35:625-32. 901-7.
[5] Guan B, Wang TL, Shih Ie M. ARID1A, a factor that promotes [24] Kim SH, Shin SJ, Lee KY, et al. Prognostic value of mucinous
formation of SWI/SNF-mediated chromatin remodeling, is a tumor histology depends on microsatellite instability status in patients with
suppressor in gynecologic cancers. Cancer Res 2011;71:6718-27. stage III colon cancer treated with adjuvant FOLFOX chemotherapy: a
[6] Bosse T, ter Haar NT, Seeber LM, et al. Loss of ARID1A expression retrospective cohort study. Ann Surg Oncol 2013;20:3407-13.
and its relationship with PI3K-Akt pathway alterations, TP53 and [25] Katagiri A, Nakayama K, Rahman MT, et al. Loss of ARID1A
microsatellite instability in endometrial cancer. Mod Pathol 2013;26: expression is related to shorter progression-free survival and chemore-
1525-35. sistance in ovarian clear cell carcinoma. Mod Pathol 2012;25:282-8.
[7] Jones S, Li M, Parsons DW, et al. Somatic mutations in the chromatin [26] Mao TL, Ardighieri L, Ayhan A, et al. Loss of ARID1A expression
remodeling gene ARID1A occur in several tumor types. Hum Mutat correlates with stages of tumor progression in uterine endometrioid
2012;33:100-3. carcinoma. Am J Surg Pathol 2013;37:1342-8.
[8] Allo G, Bernardini MQ, Wu RC, et al. ARID1A loss correlates with [27] Wiegand KC, Lee AF, Al-Agha OM, et al. Loss of BAF250a
mismatch repair deficiency and intact p53 expression in high-grade (ARID1A) is frequent in high-grade endometrial carcinomas. J Pathol
endometrial carcinomas. Mod Pathol 2013;135:611-23. 2011;224:328-33.
[9] Bosse T, Ter Haar NT, Seeber LM, et al. Loss of ARID1A expression and [28] Fadare O, Renshaw IL, Liang SX. Does the loss of ARID1A (BAF-
its relationship with PI3K-Akt pathway alterations, TP53 and microsat- 250a) expression in endometrial clear cell carcinomas have any
ellite instability in endometrial cancer. Mod Pathol 2013;135:611-23. clinicopathologic significance? A pilot assessment. J Cancer 2012;3:
[10] Cancer Genome Atlas Network. Comprehensive molecular character- 129-36.
ization of human colon and rectal cancer. Nature 2012;487:330-7. [29] Wang DD, Chen YB, Pan K, et al. Decreased expression of the
[11] Umar A, Boland CR, Terdiman JP, et al. Revised Bethesda guidelines ARID1A gene is associated with poor prognosis in primary gastric
for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and cancer. PLoS One 2012;7:e40364.
microsatellite instability. J Natl Cancer Inst 2004;96:261-8. [30] Rahman M, Nakayama K, Rahman MT, et al. Clinicopathologic
[12] Edge S, Byrd DR, Carducci MA, Compton CC, editors. AJCC Cancer analysis of loss of AT-rich interactive domain 1A expression in
Staging Manual. 7th ed. New York: Springer; 2009. endometrial cancer. HUM PATHOL 2013;44:103-9.

You might also like