ArticlePDF Available

Abstract and Figures

The tight junction protein claudin-4 has been reported to be overexpressed in advanced ovarian cancer. We investigated the prognostic significance of claudin-4 overexpression and whether claudin-4 expression could predict platinum response in primary ovarian carcinoma (OC). Claudin-4 expression was evaluated by immunohistochemistry in a tissue microarray of 140 OCs. Multivariable Cox-regression models were used to assess the effect of claudin-4 overexpression on progression-free survival and overall survival (OS). Kaplan-Meier survival analyses and the logrank test were performed comparing claudin-4 high and low groups. The association between claudin-4 expression and platinum resistance was assessed using risk ratios and the Pearson χ test. A dataset of >1500 epithelial ovarian cancers was used to study the association between CLDN4 mRNA and survival. Of 140 evaluable cases, 71 (51%) displayed high claudin-4 expression. Claudin-4 overexpression predicted shorter 5-yr progression-free survival and OS in univariable analyses [hazard ratio (HR)=1.6 (1.1-2.5), P=0.020 and HR=1.6 (1.0-2.4), P=0.041, respectively]. Hazard of relapse was similar [HR=1.5 (1.0-2.4)] after adjustment for age, stage, type, and BRCA1/2 status in a multivariable analysis, but the evidence was slightly weaker (P=0.076). Validation in an external cohort confirmed the association between high expression of CLDN4 and poor 10-yr OS [HR=1.3 (1.1-1.5), P<0.001]. However, no confident association between claudin-4 and platinum sensitivity was found in our cohort [risk ratio=1.2 (0.7-2.0), P=0.3]. These findings suggest that high expression of claudin-4 may have a prognostic value in OC. The role of claudin-4 in the development of platinum resistance remains unclear.
Content may be subject to copyright.
International Journal of Gynecological Pathology
00:1–9, Lippincott Williams & Wilkins, Baltimore
Copyright r2017 by the International Society of Gynecological Pathologists
Original Article
Claudin-4 Expression is Associated With Survival in Ovarian
Cancer But Not With Chemotherapy Response
Laura Martı
´n de la Fuente, M.D., Susanne Malander, M.D., Ph.D., Linda Hartman, Ph.D.,
Jenny-Maria Jo
¨nsson, M.D., Ph.D., Anna Ebbesson, M.Sc., Mef Nilbert, M.D., Ph.D.,
Anna Ma
˚sba
¨ck, M.D., Ph.D., and Ingrid Hedenfalk, Ph.D.
Summary: The tight junction protein claudin-4 has been reported to be overexpressed in
advanced ovarian cancer. We investigated the prognostic significance of claudin-4 over-
expression and whether claudin-4 expression could predict platinum response in primary
ovarian carcinoma (OC). Claudin-4 expression was evaluated by immunohistochemistry in a
tissue microarray of 140 OCs. Multivariable Cox-regression models were used to assess the
effect of claudin-4 overexpression on progression-free survival and overall survival (OS).
Kaplan-Meier survival analyses and the logrank test were performed comparing claudin-4 high
and low groups. The association between claudin-4 expression and platinum resistance was
assessed using risk ratios and the Pearson w
2
test. A dataset of >1500 epithelial ovarian
cancers was used to study the association between CLDN4 mRNA and survival. Of 140
evaluable cases, 71 (51%) displayed high claudin-4 expression. Claudin-4 overexpression
predicted shorter 5-yr progression-free survival and OS in univariable analyses [hazard ratio
(HR) = 1.6 (1.1–2.5), P= 0.020 and HR = 1.6 (1.0–2.4), P= 0.041, respectively]. Hazard of
relapse was similar [HR = 1.5 (1.0–2.4)] after adjustment for age, stage, type, and BRCA1/2
status in a multivariable analysis, but the evidence was slightly weaker (P=0.076).Validation
in an external cohort confirmed the association between high expression of CLDN4 and poor
10-yr OS [HR = 1.3 (1.1–1.5), Po0.001]. However, no confident association between claudin-
4 and platinum sensitivity was found in our cohort [risk ratio = 1.2 (0.7–2.0), P=0.3]. These
findings suggest that high expression of claudin-4 may have a prognostic value in OC. The role
of claudin-4 in the development of platinum resistance remains unclear. Key Words: Claudin-
4—Ovarian carcinoma—Platinum resistance—Prognostic factor.
Ovarian cancer is one of the leading causes of
cancer death among women due to difficulties in both
diagnosis and therapy. Because of the insidious onset
of the disease and the lack of reliable screening
methods, two thirds of patients present with ad-
vanced stage disease upon diagnosis (1). The 5-yr
From the Department of Clinical Sciences, Division of Oncology and Pathology, Lund University and Ska
˚ne University Hospital
(L.M.d.l.F., S.M., L.H., J.-M.J., A.E., M.N., I.H.); Regional Cancer Center South Sweden (L.H., M.N.); Department of Surgical Pathology,
Division of Laboratory Medicine, Ska
˚ne University Hospital (A.M.); and CREATE Health Strategic Center for Translational Cancer
Research, Lund University (I.H.), Lund, Sweden.
I.H.: received grant support from the Swedish Cancer Society, the G Nilsson Cancer Foundation, the B Kamprad Foundation, the Cancer
and Allergy Foundation, King Gustaf V’s Jubilee Foundation, the Lund University Hospital Research Foundation, and governmental
funding of clinical research within the National Health Services (ALF). The remaining authors declare no conflict of interest.
Address correspondence and reprint requests to Laura Martı
´n de la Fuente, MD, Department of Clinical Sciences, Division of Oncology
and Pathology, Lund University Cancer Center/Medicon Village, Lund SE-223 81, Sweden. E-mail: laura.martin_de_la_fuente@med.lu.se.
Supplemental Digital Content is available for this article. Direct URL citations appear in the printed text and are provided in the HTML
and PDF versions of this article on the journal’s Website, www.intjgynpathology.com.
This is an open-access article distributed under the terms of the Creative Commons Attribution-Non Commercial-No Derivatives License 4.0
(CCBY-NC-ND), where it is permissible to download and share the work provided it is properly cited. The work cannot be changed in any
way or used commercially without permission from the journal.
1DOI: 10.1097/PGP.0000000000000394
relative survival rate is 27% for patients with
advanced disease (stage III-IV) (2). Despite efforts
aimed at early detection and new therapeutic
approaches, the poor survival persists and may be
explained by the heterogeneity and poorly under-
stood pathogenesis of ovarian carcinoma (OC).
Platinum compounds comprise the most active
chemotherapy available and constitute standard treat-
ment after cytoreductive surgery for the majority of
women diagnosed with ovarian cancer (3). First-line
chemotherapy with platinum-based chemotherapy yields
response rates of >80%. However, nearly all patients
relapse and eventually develop platinum resistance (4). A
biomarker capable of predicting platinum resistance at
primary surgery could make it possible to identify
patients who may not be suitable for platinum-based
therapy, and thus be spared its side effects, or who could
be eligible for other treatment options.
Claudins belong to a multigene family, with B24
members, and are considered one of the main tight
junction (TJ) forming proteins (5,6). A TJ acts as a
cellular barrier and is involved in paracellular perme-
ability and cell polarity (7,8). Since the discovery of
claudins in 1998, many studies have aimed to character-
ize the claudin family, revealing that the specific
combination and coexpression of different claudin
species determines the TJ barrier characteristics in a
tissue-specific manner (9,10). TJ structure and function
are often altered in human carcinomas, where TJ loss
can contribute to cancer progression (11). For example,
claudin-1 has been found to be downregulated in breast
cancer, and claudin-2 is downregulated in breast and
prostate cancer (12,13). In contrast, claudin-4 has been
reported to be upregulated in pancreatic, colorectal,
gastric, breast, prostate, and ovarian cancers (13–20).
Previous studies have reported increased expression of
claudins 3 and 4 in OC compared with normal ovarian
surface epithelium and benign ovarian tumors (20–22).
Claudin-4 has been proposed as a possible diagnostic
and prognostic biomarker in OC (3,23). However, there
are limited and contradictory data regarding the involve-
ment of claudins in chemotherapy resistance (24–26).
Therefore, in this study we investigated the role of
claudin-4 as a potential prognostic marker in 140 patients
with primary OC. The relationship between platinum
resistance and claudin-4 expression was also investigated.
MATERIALS AND METHODS
Patients
A total of 128 patients with OC were recruited
consecutively in the southern Swedish health care
region between 1998 and 2000 (27). In addition, 18
patients with OC were recruited at the oncogenetic
counseling at Lund University Hospital (Sweden)
between 1981 and 1997. Six patients with unknown
primary tumor or missing follow-up information
were excluded. Thus, a cohort of 140 OC patients was
assessed for claudin-4 protein expression.
The study was approved by the Ethics Committee
at Lund University, Sweden, waiving the requirement
for informed consent. The histologic subtype and
grade were determined according to WHO 2014 (28),
and all tumors were staged according to the Interna-
tional Federation of Gynecology and Obstetrics
(FIGO) criteria (29). Tumors were also classified into
types I and II on the basis of both histology and
grade (30). Platinum resistance was defined as
primary progressive disease or recurrence within
6 mo of completing platinum-based therapy, the
definition used in clinical practice today.
The majority of the tumors (70%) were of serous
histology, followed by endometrioid, mucinous, and
clear cell histologies. Approximately 75% of the
patients were diagnosed with advanced stage (III/IV)
disease, 64% were classified as type II, and 31/140
(22%) were carriers of a germline BRCA1 or BRCA2
mutation. Platinum-based chemotherapy was admin-
istered postoperatively to 132 patients: Carboplatin
(AUC5) combined with paclitaxel (175 mg/m
2
)to70
patients, Carboplatin (AUC5) combined with cyclo-
phosphamide (500 mg/m
2
) to 25 patients, and un-
specified platinum-based chemotherapy to 36
patients. Thirty-nine of the 132 patients who received
chemotherapy (30%) were platinum resistant. Seven
patients with early-stage (IA-B) disease and type I
tumors were excluded from the study of claudin-4
expression as a treatment predictive marker for
platinum response, as they had not received
platinum-based therapy, in accordance with current
guidelines. One patient was excluded from the same
analysis because she received non–platinum-based
chemotherapy postoperatively.
Data from a cohort of 1582 OC patients available
through the online tool Kaplan-Meier Plotter (31)
were used for validation.
Tissue Microarray (TMA) Construction and
Immunohistochemistry
We used a TMA with 0.6 mm triplicate core needle
biopsies from viable tumor areas (27). Sections, 3–4 mm
in thickness, were deparaffinized, rehydrated, and
stained. Antigen retrieval was performed using Dako
2 L. MARTI´N DE LA FUENTE ET AL.
Int J Gynecol Pathol Vol. 00, No. 00, ’’ 2017
Target retrieval solution of pH 6 (Dako A/S, Glostrup,
Denmark) in a pressure cooker 2100 Retriever
(Histolab Products AB, Gothenburg, Sweden). The
sections were incubated with a claudin-4 mouse
monoclonal antibody (Cat.No. 32-9400; Invitrogen
Corporation, Camarillo) at a 1:100 dilution for 30 min.
Immunohistochemical reactions were performed using
the Autostainer Plus with the Dako REAL EnVision
Detection system (Dako). Normal colon tissue was
included as a positive control. The negative controls
were ovarian stroma and follicle cells as well as smooth
muscle and adipose tissue, all reported as negative for
claudin-4 expression in The Human Protein Atlas (32).
We also stained whole-tissue sections with benign
ovary, fallopian tube, and endometrium, as well as 3
cases of serous cystadenomas.
A semiquantitative analysis of claudin-4 was
performed by 2 blinded investigators following
recommendations from a senior gynecologic pathol-
ogist (A.M.) and on the basis of the similar type of
expression pattern observed for HER2. Claudin-4
staining was distinct and predominantly confined to
the cell membrane. Two staining patterns were
observed: (i) punctate and partial staining around
the membrane; and (ii) complete membrane staining,
covering the circumference of the membrane surface.
Results from the core with the highest/strongest
positivity were recorded into 5 subgroups on the basis
of staining pattern and fraction of stained cancer cells
(0: no staining, 1: <50% of cells with punctate/
partial membrane staining, 2: 50%–75% of cells with
punctate/partial membrane staining, 3: 50%–75% of
cells with complete membrane staining, 4: >75% of
cells with complete membrane staining).
As claudin-4 was expressed in the majority of OCs
examined and no consensus for cutoff was available
in the literature, an exploratory cutoff was used in the
present study. Tumors with a Z3+ score were
considered to have high expression and the rest had
low expression. This cutoff was established on the
basis of the percentage of patients with primary
platinum-resistant disease and the 2 staining patterns
observed, before evaluating the effect on prognosis or
platinum resistance.
Statistical Analyses
The prognostic value of claudin-4 was investigated
using 5-yr progression-free survival (PFS) time and 5-yr
overall survival (OS) time as endpoints. PFS time was
defined as the time interval between date of diagnosis
and the first sign of disease recurrence (clinical and/or
radiologic) or death, whichever occurred first. OS time
was defined as the time interval between date of
diagnosis and death. Of the patients who died within
5 yr after diagnosis, all but 2 died of ovarian cancer.
Statistical analyses were performed with SPSS for
Windows version 22. To analyze the probability of a
patient being platinum resistant, we calculated the
relative risk (risk ratio) of high versus low claudin-4
expression with 95% confidence interval (95% CI).
Associations between claudin-4 expression and plat-
inum resistance and other clinical parameters were
assessed using the Pearson w
2
test or the Fisher exact
test, except for the ordinal variable stage, which was
compared using the Mann-Whitney Utest. Survival
analyses for PFS and OS were performed using the
Kaplan-Meier method, and differences between
groups were tested using the logrank test. The effect
of claudin-4 expression on survival was expressed
using hazard ratios (HR) with 95% CI, estimated
using univariable (crude effect) and multivariable
(adjusting for clinical factors known to influence
ovarian cancer survival) Cox regression. These
factors included age at diagnosis (Z70 y vs. <70),
tumor type (II vs. I), stage (III/IV vs. I/II), and
BRCA1/2 mutation status (wild-type vs. muta-
tion) (30,33–35) and were analyzed as binary factors.
The cutoff of 70 yr was chosen on the basis of
previous studies reporting an inferior outcome
among elderly women (33). The classification into
type I and II is made on the basis of both histology
and grade, and defines 2 fundamentally different
groups regarding tumorigenesis and prognosis (30). A
large meta-analysis showed better OS and PFS for
BRCA1 and BRCA2 mutation carriers compared
with noncarriers (35). The prognostic factor residual
disease following cytoreductive surgery was not
included in the analyses, as this information was
not documented. Cox regression was performed for
the type II tumors alone as a stability analysis. All P-
values are 2-sided.
RESULTS
Immunohistochemical Staining of Claudin-4
Seven of the 140 OCs displayed no staining, and
the distribution in subgroups 1–4 was as follows:
1: n = 32, 2: n = 30, 3: n = 18, 4: n = 53. Thus, 71 cases
(51%) displayed high claudin-4 expression. Figure 1
shows representative examples of high and low claudin-
4expressioninOC.Theexpressionofclaudin-4(lowvs.
high) in the different histologic subtypes, stages, and
types, as well as in relation to age, BRCA1/2 mutation
3CLAUDIN-4 EXPRESSION IN OVARIAN CARCINOMA
Int J Gynecol Pathol Vol. 00, No. 00, ’’ 2017
status, and platinum sensitivity, is shown in Table 1.
High expression of claudin-4 correlated with high-grade
serous histology compared with low-grade serous
histology (w
2
test, P= 0.037) and high age at diagnosis
(w
2
test, P= 0.049). A correlation between type II
tumors and high claudin-4 expression was also
observed, but this did not reach statistical significance
(w
2
test, P= 0.071). No correlation was found with
BRCA1/2 status (wild-type vs. mutant, w
2
test, P=0.6)
or stage (Mann-Whitney Utest, P=0.1) (Table 1).
Two whole-tissue sections from a benign ovary with
mesothelial hyperplasia revealed negative staining in
the mesothelium, including the hyperplastic areas,
whereas cortical serous inclusion cysts were strongly
positive (Fig. 1). Epithelial cells in the 3 cases of serous
cystadenomas also displayed strong claudin-4 expres-
sion. Other cell types in the ovary, including stromal
and smooth muscle cells, showed no claudin-4
expression in all evaluated cases, in line with the
Human Protein Atlas (32). Epithelium from normal
fallopian tube and normal endometrium displayed
high claudin-4 expression, also in line with the Human
Protein Atlas (5 and 10 cases, respectively) (32).
Claudin-4 Expression and Platinum Sensitivity
Among the 132 patients who had received
platinum-based chemotherapy, a patient with high
claudin-4 expression had a 1.2 (95% CI = 0.7–2.0,
P= 0.3) times higher risk (risk ratio) of being
platinum resistant compared with a patient with
low claudin-4 expression (Table 1). No associations
with platinum responsiveness were found in separate
analyses of type II tumors (n = 86, P= 0.6), type I
tumors (n = 42, Fisher Exact test, P= 0.4), the
serous subtype (n = 97, P= 0.7), late-stage (III-IV)
disease (n = 96, P= 0.8), early-stage (I-II) disease
(n = 36, Fisher Exact test, P= 0.2), or BRCA1/2
wild-type status (n = 100, P= 0.2).
Claudin-4 Expression and Prognosis of OC
Univariable analyses revealed an association be-
tween PFS and claudin-4 expression [HR = 1.7
(1.1–2.6), P= 0.020] (Table 2), with patients whose
tumors expressed high levels of claudin-4 displaying
an inferior outcome (Kaplan-Meier survival
analysis, Fig. 2). Hazard of relapse was similar
[HR = 1.5 (1.0–2.4)] after adjustment for age, stage,
type, and BRCA1/2 status in a multivariable analysis,
but the evidence was slightly weaker (P= 0.076)
(Table 2). A comparable association was observed for
OS in the univariable and multivariable analyses
[HR = 1.6 (1.0–2.4), P= 0.041 and HR = 1.5
(0.9–2.3), P= 0.1] (Table 3). Analyses of type II
tumors (n = 86) showed a similar HR in multivariable
analysis compared with all OCs [1.6 (0.9–2.8),
FIG. 1. Immunohistochemical evaluation of claudin-4 in ovarian tissues, including claudin-4 high (A) and low (B) serous ovarian cancer, and
claudin-4 high (D) and low (E) endometrioid ovarian cancer. Negative ovarian mesothelium and positive cortical inclusion cyst (C). Positive
control, healthy colon (F). Magnification: 40 .
4 L. MARTI´N DE LA FUENTE ET AL.
Int J Gynecol Pathol Vol. 00, No. 00, ’’ 2017
P= 0.078 for PFS and 1.6 (0.9–2.7), P= 0.086 for
OS].
To extend our findings, we investigated the role of
CLDN4 mRNA levels in a cohort of 1582 OC
patients available through the online tool Kaplan-
Meier Plotter (31). The online tool comprises gene
expression data and survival information from 13
independent public OC data sets (Gene Expression
Omnibus and The Cancer Genome Atlas, n = 28-565,
2015 version). The best performing threshold was
used as cutoff, where 1105 patients (70%) were
classified as CLDN4 low and 477 (30%) as CLDN4
high. These results were in line with our findings, as
CLDN4-high patients displayed worse 5-yr OS in a
univariable analysis [HR = 1.3 (1.1–1.5), Po0.001]
compared with CLDN4-low patients. Interestingly,
the results were consistent after 10 yr of follow-up [see
Fig. Supplemental Digital Content 1, http://link-
s.lww.com/IJGP/A57, HR = 1.3 (1. 2–1.5), Po0.001
for 10-yr OS]. However, a similar difference in 10-yr
PFS was not observed [HR = 1.2 (1.0–1.3),
P= 0.064].
DISCUSSION
A previous study has reported low or absent
claudin-4 expression in normal ovarian surface
epithelium and ovarian cystadenomas, in contrast
to the high expression reported in OC, which
according to the authors supports the involvement
of claudin-4 in malignant transformation (20). We
also found absence of expression in ovarian surface
mesothelium, and interestingly also in the hyper-
plastic changes. In contrast to the former study, we
found strong positive expression in cortical inclusion
cysts (CIC) and serous cystadenomas. Furthermore,
we also found strong positive expression in normal
fallopian tube epithelium, consistent with data
reported in the Human Protein Atlas (32).
TABLE 1. Claudin-4 expression and clinicopathologic parameters
N (%)
N (%) Claudin-4 low Claudin-4 high P*
All ECs 140 69 (49) 71 (51)
Age (yr)
Mean (range) 59 (26–83) 58 (26–83) 61 (26–83)
o70 110 (79) 59 (54) 51 (46) 0.049
Z70 30 (21) 10 (33) 20 (67)
BRCA1/2 status
Mutant 31(22) 14 (45) 17 (55) 0.6
Wild-type 109 (78) 55 (51) 54 (49)
Histology
High-grade serous 80 (70) 36 (45) 44 (55) 0.037w
Low-grade serous 18 (13) 13 (72) 5 (28)
Endometrioid FIGO 3 5 (3.6) 1 4
Endometrioid FIGO 1/2 16 (11.4) 7 9
Mucinous 10 (7) 4 6
Clear cell 5 (3.6) 5 0
Carcinosarcoma 1 0 1
Mixed and missing 5
Tumor type
I 49 (36) 29 (59) 20 (41) 0.071
II 86 (64) 37 (43) 49 (57)
Missing 5
Stage
I 27 (12) 17 (65) 10 (37) 0.1z
II 17 (13) 9 (53) 8 (47)
III 77 (60) 35 (45) 42 (55)
IV 19 (15) 8 (42) 11 (58)
Platinum responsiveness
Sensitive 91 (70) 43 (50) 44 (50) 0.3
Resistant 39 (30) 17 (44) 22 (56)
Missing 2
Bold values are statistically significant Pvalue o0.05.
*w
2
test between claudin-4 low and high groups.
ww
2
test comparing the high-grade and low-grade serous ovarian carcinomas.
zMann-Whitney Utest between claudin-4 low and high groups.
5CLAUDIN-4 EXPRESSION IN OVARIAN CARCINOMA
Int J Gynecol Pathol Vol. 00, No. 00, ’’ 2017
A new paradigm for the pathogenesis of ovarian
serous carcinoma, which questions the traditional
model of ovarian mesothelium as the cell of origin for
serous neoplasia, has been proposed. Cogent argu-
ments support the role of fallopian tube epithelium in
the genesis of both low and high-grade serous
OCs (36–43). Serous tubal intraepithelial carcinomas
found in prophylactic salpingo-oophorectomies in
BRCA mutation carriers and sporadic cases of pelvic
high-grade serous carcinoma are this far the best
evidence of this extraovarian hypothesis (40,42,43).
Furthermore, a new model of serous tumorigenesis
proposes that normal tubal epithelium may shed and
implant on the ovarian surface, thereby forming
CICs. CICs and serous cystadenomas follow a benign
course in most cases, but in some cases they can
eventually lead to low and sometimes high-grade
serous tumors (42). Thus, no conclusion regarding
the involvement of claudin-4 in malignant trans-
formation should be made on the basis of the
comparison with ovarian mesothelium (40).
In our study, fallopian tube epithelium, as well as
CICs and serous cystadenomas, displayed high
claudin-4 expression, arguing against the upregula-
tion of claudin-4 in malignant transformation.
Furthermore, we examined endometrial epithelium,
as endometrioid and clear cell tumors have been
associated with endometriosis and because their gene
expression profiles resemble those of endometrial
epithelium (40,43). High endometrial claudin-4 ex-
pression was observed, also consistent with the
Human Protein Atlas (32).
Decreased expression of claudins in cancer is in
agreement with the hypothesis that tumorigenesis is
accompanied by TJ disruption and loss of cell-cell
adhesion, leading to disease dissemination (11). In
contrast, overexpression of claudin-4 has been reported
in several cancer types, including OC (20–22,44).
Upregulation of CLDN3 and CLDN4 has been shown
to increase cell invasion and motility in OC cell lines,
and, conversely, knockdown caused the opposite
effects, suggesting that claudin-3 and 4 may indeed
promote ovarian tumorigenesis (45). In the present
study, we found that high levels of claudin-4 were
associated with decreased PFS and OS. The hazards of
relapse or death were similar but not significant when
TABLE 2. Univariable and multivariable analyses of progression-free survival
5-yr PFS univariable Cox 5-yr PFS multivariable Cox
N (events) HR (95% CI) PHR (95% CI) P
Claudin-4 expression
Low 64 (38) 1
High 70 (55) 1.7 (1.1–2.6) 0.020 1.5 (1.0–2.3) 0.076
Age at diagnosis (yr)
o70 107 (71) 1
Z70 27 (22) 1.5 (0.9–2.4) 0.1 1.1 (0.6–1.8) 0.7
BRCA1/2 status
Mutant 29 (18) 1
Wild-type 105 (75) 1.7 (1.0–3.1) 0.052 2.6 (1.5–4.6) 0.001
Stage
I/II 40 (11) 1
III/IV 94 (82) 6.2 (3.3–11.7) o0.001 4.4 (2.2–8.8) o0.001
Type
I 47 (20) 1
II 82 (68) 2.9 (1.8–4.8) o0.001 2.2 (1.2–3.8) 0.023
Bold values are statistically significant Pvalue o0.05.
CI indicates confidence interval; HR, hazard ratio; PFS, progression-free survival.
FIG. 2. Kaplan-Meier survival analysis. Association between
progression-free survival (PFS) and the level of claudin-4
expression. Patients with high claudin-4 expression (solid line,
n = 70) had shorter PFS than those with low expression (dotted
line, n = 64). Median PFS 18 versus 37 mo. Logrank P= 0.018.
6 L. MARTI´N DE LA FUENTE ET AL.
Int J Gynecol Pathol Vol. 00, No. 00, ’’ 2017
adjusting for known prognostic factors, probably due
to lack of power. The group with low claudin-4
expression had better prognosis and moreover dis-
played lower claudin-4 expression than the normal
tissue levels used for comparison. Thus, there is a
possibility that tumors that downregulate claudin-4
expression during malignant transformation become
less aggressive tumors with better prognosis. Our
findings also suggest that claudin-4 may have prog-
nostic value.
Currently, the development of resistance can only
be determined retrospectively, after patients have
experienced the burden and toxicity of ineffective
therapy (3). Clinical recurrences that occur within
6 mo after cessation of primary platinum-based
chemotherapy are considered platinum resistant. This
definition of platinum resistance is a strong predictor
of response to second-line therapy, with very low
response rates for patients defined as platinum
resistant (46,47). A biomarker capable of predicting
platinum resistance at primary surgery could enable
the identification of patients not suitable for
platinum-based therapy and who may thus be spared
its side effects. Furthermore, patients with over-
expression of claudin-4 and platinum-resistant dis-
ease may be candidates for clinical trials, including
claudin-4-targeted therapy, as claudin-4 is potentially
druggable using the C-terminal fragment of Clostri-
dium perfringens enterotoxin (24–26). Such a bio-
marker could therefore provide the basis for
individualization of therapy and improved outcome.
In the current study we attempted to establish
whether claudin-4 might be predictive of platinum
sensitivity. Only a few reports are available on the
association between platinum resistance and claudin-
4 expression, and the results are contradictory. A
proteomic study found 7-fold increased levels of
claudin-4 protein in a platinum-resistant cell line
compared with the sensitive parental line (3). Two
studies have reported associations between high levels
of claudin-4 protein and platinum resistance (24,26),
whereas a third study failed to detect any association
with platinum resistance or survival (25). These
studies are, however, small (n = 12–43) and may
not be representative of the whole spectrum of OC. In
fact, one of the studies reporting significantly higher
claudin-4 expression in chemoresistant compared
with chemosensitive cases had an overrepresentation
of clear cell histology, known to be inherently more
chemoresistant (10/43 cases, 23%), compared with
our cohort (4/130 cases, 3%) (26). The association
between platinum resistance and claudin-4 may hence
be confounded by the histologic type, emphasizing
the importance of stratification of histologic and
probably also molecular subtypes when investigating
potential treatment predictive biomarkers in OC. The
other study compared CLDN4 mRNA levels at
primary surgery from 6 patients with chemotherapy
naive serous OC with chemotherapy resistant tumors
from 6 patients with recurrent disease, showing
CLDN4 upregulation at recurrence (24). In contrast,
we investigated only tumor tissue from primary
surgery, as we were interested in investigating the
potential predictive value at first-line chemotherapy.
However, pairwise comparisons between primary
surgery and relapse may be interesting and could
potentially inform treatment decisions in recurrent
disease, as there may be a group of patients in whose
TABLE 3. Univariable and multivariable analyses of overall survival
5-yr OS univariable Cox 5-yr OS multivariable Cox
N (events) HR (95% CI) PHR (95% CI) P
Claudin-4 expression
Low 69 (36) 1
High 71 (48) 1.6 (1.0–2.4) 0.041 1.5 (0.9–2.3) 0.1
Age at diagnosis (yr)
o70 110 (60) 1
Z70 30 (24) 1.9 (1.2–2.9) 0.011 1.7 (1.0–2.9) 0.037
BRCA1/2 status
Mutant 31 (16) 1
Wild-type 109 (68) 1.4 (0.8–2.4) 0.2 2.1 (1.2–3.7) 0.013
Stage
I/II 44 (13) 1
III/IV 96 (71) 4.2 (2.3–7.6) o0.001 2.8 (1.5–5.3) 0.002
Type
I 49 (17) 1
II 86 (64) 3.0 (1.7–5.1) o0.001 2.1 (1.1–3.8) 0.020
Bold values are statistically significant Pvalue o0.05.
CI indicates confidence interval; HR, hazard ratio.
7CLAUDIN-4 EXPRESSION IN OVARIAN CARCINOMA
Int J Gynecol Pathol Vol. 00, No. 00, ’’ 2017
tumors claudin-4 is upregulated upon development of
platinum resistance. This line of research warrants
further attention in larger cohorts with matched primary
tumors and relapses. Nevertheless, we did not find a
confident association between claudin-4 expression and
platinum sensitivity. Our findings are, however, con-
sistent with a previous report showing that claudin-4
expression, determined by qRT-PCR and IHC, did not
differ between platinum-sensitive and platinum-resistant
tumors in a cohort of 36 patients with high-grade,
advanced-stage serous ovarian cancer (25). Further-
more, the definition of platinum resistance differed in
the previously reported studies. Taken together, we did
not find claudin-4 to be a useful predictive marker for
platinum sensitivity in the clinical situation, either in an
unselected cohort or within any other subgroup.
Of interest, claudin-4 has been suggested as a
potential therapeutic target. Suppression of claudin-4
expression in human serous ovarian cancer cell lines
by siRNA led to cellular accumulation of cisplatin
and a significant increase in cisplatin sensitivity (26),
suggesting that upregulation of claudin-4 expression
may contribute to platinum resistance by decreasing
drug uptake. Additional mechanisms of drug resist-
ance may nevertheless also be present (48), and it
remains to be confirmed whether claudin-4 over-
expression is involved. We hypothesize that the
heterogeneity and complexity of resistance mecha-
nisms may explain why no differences between
resistant and sensitive tumors were observed in the
present study.
In conclusion, we identified claudin-4 as a potential
prognostic biomarker in OC. Although previous studies
have suggested a relationship between claudin-4 and
platinum resistance, we were not able to validate this in
our larger cohort. The druggable nature of claudin-4
nevertheless makes it appealing to pursue further as a
target for circumventing platinum resistance. Further
investigations aimed at studying the relationship be-
tween claudin-4 and platinum resistance are needed, in
addition to validation studies to find application into
clinical practice.
Acknowledgment: The authors would like to acknowl-
edge Kristina Lo
¨vgren for assistance with TMAs and
stainings.
REFERENCES
1. Eisenhauer EL, Salani R, Copeland LJ. Epithelial ovarian
cancer. In: Di Saia PJ, Creasman WT, eds. Clinical Gynecologic
Oncology, Eighth edn. Philadelphia, PA: Anne Altepeter;
2012:285–328.
2. NNA Howlader, Krapcho M, Garshell J. SEER Cancer
Statistics Review, 1975-2011. Bethesda, MD: National Cancer
Institute; 2013.
3. Stewart JJ, White JT, Yan X, et al. Proteins associated with
cisplatin resistance in ovarian cancer cells identified by
quantitative proteomic technology and integrated with mRNA
expression levels. Mol Cell Proteomics 2006;5:433–43.
4. Sood AK, Buller RE. Drug resistance in ovarian cancer: from
the laboratory to the clinic. Obstet Gynecol 1998;92:312–9.
5. Morita K, Furuse M, Fujimoto K, et al. Claudin multigene
family encoding four-transmembrane domain protein compo-
nents of tight junction strands. Proc Natl Acad Sci U S A
1999;96:511–6.
6. Gonzalez-Mariscal L, Betanzos A, Nava P, et al. Tight
junction proteins. Prog Biophys Mol Biol 2003;81:1–44.
7. Cereijido M, Valdes J, Shoshani L, et al. Role of tight junctions
in establishing and maintaining cell polarity. Annu Rev Physiol
1998;60:161–77.
8. Schneeberger EE, Lynch RD. The tight junction: a multi-
functional complex. Am J Physiol Cell Physiol 2004;286:
C1213–28.
9. Furuse M, Fujita K, Hiiragi T, et al. Claudin-1 and -2: novel
integral membrane proteins localizing at tight junctions with
no sequence similarity to occludin. J Cell Biol 1998;141:
1539–50.
10. Tsukita S, Furuse M, Itoh M. Multifunctional strands in tight
junctions. Nat Rev Mol Cell Biol 2001;2:285–93.
11. Oliveira SS, Morgado-Diaz JA. Claudins: multifunctional
players in epithelial tight junctions and their role in cancer.
Cell Mol Life Sci 2007;64:17–28.
12. Tokes AM, Kulka J, Paku S, et al. Claudin-1, -3 and -4
proteins and mRNA expression in benign and malignant breast
lesions: a research study. Breast Cancer Res 2005;7:R296–305.
13. Soini Y. Expression of claudins 1, 2, 3, 4, 5 and 7 in various
types of tumours. Histopathology 2005;46:551–60.
14. Nichols LS, Ashfaq R, Iacobuzio-Donahue CA. Claudin 4
protein expression in primary and metastatic pancreatic cancer:
support for use as a therapeutic target. Am J Clin Pathol
2004;121:226–30.
15. Michl P, Buchholz M, Rolke M, et al. Claudin-4: a new target
for pancreatic cancer treatment using Clostridium perfringens
enterotoxin. Gastroenterology 2001;121:678–84.
16. de Oliveira SS, de Oliveira IM, De Souza W, et al. Claudins
upregulation in human colorectal cancer. FEBS Lett 2005;579:
6179–85.
17. Resnick MB, Gavilanez M, Newton E, et al. Claudin
expression in gastric adenocarcinomas: a tissue microarray
study with prognostic correlation. Hum Pathol 2005;36:
886–92.
18. Cunningham SC, Kamangar F, Kim MP, et al. Claudin-4,
mitogen-activated protein kinase kinase 4, and stratifin are
markers of gastric adenocarcinoma precursor lesions. Cancer
Epidemiol Biomarkers Prev 2006;15:281–7.
19. Kominsky SL, Vali M, Korz D, et al. Clostridium perfringens
enterotoxin elicits rapid and specific cytolysis of breast
carcinoma cells mediated through tight junction proteins
claudin 3 and 4. Am J Pathol 2004;164:1627–33.
20. Rangel LB, Agarwal R, D’Souza T, et al. Tight junction
proteins claudin-3 and claudin-4 are frequently overexpressed
in ovarian cancer but not in ovarian cystadenomas. Clin
Cancer Res 2003;9:2567–75.
21. Hough CD, Sherman-Baust CA, Pizer ES, et al. Large-scale
serial analysis of gene expression reveals genes differentially
expressed in ovarian cancer. Cancer Res 2000;60:6281–7.
22. Santin AD, Zhan F, Bellone S, et al. Gene expression profiles
in primary ovarian serous papillary tumors and normal
ovarian epithelium: identification of candidate molecular
markers for ovarian cancer diagnosis and therapy. Int J
Cancer 2004;112:14–25.
8 L. MARTI´N DE LA FUENTE ET AL.
Int J Gynecol Pathol Vol. 00, No. 00, ’’ 2017
23. Szabo I, Kiss A, Schaff Z, et al. Claudins as diagnostic and
prognostic markers in gynecological cancer. Histol Histopathol
2009;24:1607–15.
24. Santin AD, Cane S, Bellone S, et al. Treatment of
chemotherapy-resistant human ovarian cancer xenografts in
C.B-17/SCID mice by intraperitoneal administration of
Clostridium perfringens enterotoxin. Cancer Res 2005;65:
4334–42.
25. Litkouhi B, Kwong J, Lo CM, et al. Claudin-4 overexpression
in epithelial ovarian cancer is associated with hypomethylation
and is a potential target for modulation of tight junction
barrier function using a C-terminal fragment of Clostridium
perfringens enterotoxin. Neoplasia 2007;9:304–14.
26. Yoshida H, Sumi T, Zhi X, et al. Claudin-4: a potential
therapeutic target in chemotherapy-resistant ovarian cancer.
Anticancer Res 2011;31:1271–7.
27. Malander S, Rambech E, Kristoffersson U, et al. The
contribution of the hereditary nonpolyposis colorectal cancer
syndrome to the development of ovarian cancer. Gynecol Oncol
2006;101:238–43.
28. Kurman RJ, Carcangiu ML, Herrington CS, Young RH. eds.
World Health Organization Classification of Tumours of the
Female Reproductive Organs 4th ed. IARC: Lyon; 2014.
29. Heintz AP, Odicino F, Maisonneuve P, et al. Carcinoma of the
ovary. FIGO 26th Annual Report on the Results of Treatment
in Gynecological Cancer. Int J Gynaecol Obstet 2006;
95(suppl 1):S161–92.
30. Shih IeM, Kurman RJ. Ovarian tumorigenesis: a proposed
model based on morphological and molecular genetic analysis.
Am J Pathol 2004;164:1511–8.
31. Gyorffy B, Lanczky A, Szallasi Z. Implementing an online tool
for genome-wide validation of survival-associated biomarkers
in ovarian-cancer using microarray data from 1287 patients.
Endocr Relat Cancer 2012;19:197–208.
32. Uhlen M, Fagerberg L, Hallstrom BM, et al. Proteomics.
Tissue-based map of the human proteome. Science 2015;347:
1260419.
33. Sabatier R, Calderon B, Jr, Lambaudie E, et al. Prognostic
factors for ovarian epithelial cancer in the elderly: a case-
control study. Int J Gynecol Cancer 2015;25:815–22.
34. Swenerton KD, Hislop TG, Spinelli J, et al. Ovarian
carcinoma: a multivariate analysis of prognostic factors. Obstet
Gynecol 1985;65:264–70.
35. Zhong Q, Peng HL, Zhao X, et al. Effects of BRCA1- and
BRCA2-related mutations on ovarian and breast cancer
survival: a meta-analysis. Clin Cancer Res 2015;21:211–20.
36. Medeiros F, Muto MG, Lee Y, et al. The tubal fimbria is a
preferred site for early adenocarcinoma in women with familial
ovarian cancer syndrome. Am J Surg Pathol 2006;30:230–6.
37. Kindelberger DW, Lee Y, Miron A, et al. Intraepithelial
carcinoma of the fimbria and pelvic serous carcinoma: evidence
for a causal relationship. Am J Surg Pathol 2007;31:161–9.
38. Jarboe E, Folkins A, Nucci MR, et al. Serous carcinogenesis in
the fallopian tube: a descriptive classification. Int J Gynecol
Pathol 2008;27:1–9.
39. Gilks CB, Prat J. Ovarian carcinoma pathology and genetics:
recent advances. Hum Pathol 2009;40:1213–23.
40. Kurman RJ, Shih IeM. The origin and pathogenesis of
epithelial ovarian cancer: a proposed unifying theory. Am J
Surg Pathol 2010;34:433–43.
41. Jones PM, Drapkin R. Modeling high-grade serous carcinoma:
how converging insights into pathogenesis and genetics are
driving better experimental platforms. Front Oncol 2013;3:217.
42. Nik NN, Vang R, Shih IeM, et al. Origin and pathogenesis of
pelvic (ovarian, tubal, and primary peritoneal) serous carcino-
ma. Annu Rev Pathol 2014;9:27–45.
43. Dubeau L. The cell of origin of ovarian epithelial tumours.
Lancet Oncol 2008;9:1191–7.
44. Hibbs K, Skubitz KM, Pambuccian SE, et al. Differential gene
expression in ovarian carcinoma: identification of potential
biomarkers. Am J Pathol 2004;165:397–414.
45. Agarwal R, D’Souza T, Morin PJ. Claudin-3 and claudin-4
expression in ovarian epithelial cells enhances invasion and is
associated with increased matrix metalloproteinase-2 activity.
Cancer Res 2005;65:7378–85.
46. Blackledge G, Lawton F, Redman C, et al. Response of
patients in phase II studies of chemotherapy in ovarian cancer:
implications for patient treatment and the design of phase II
trials. Br J Cancer 1989;59:650–3.
47. Markman M, Markman J, Webster K, et al. Duration of
response to second-line, platinum-based chemotherapy for
ovarian cancer: implications for patient management and
clinical trial design. J Clin Oncol 2004;22:3120–5.
48. Vasey PA. Resistance to chemotherapy in advanced ovarian
cancer: mechanisms and current strategies. Br J Cancer
2003;89(suppl 3):S23–28.
9CLAUDIN-4 EXPRESSION IN OVARIAN CARCINOMA
Int J Gynecol Pathol Vol. 00, No. 00, ’’ 2017
... Claudin-4 is also used for differentiation of adenocarcinoma, as well as a predictor of overall survival in those with ovarian cancer. 4 Additionally, juvenile granulosa cell tumors may clinically manifest with hypercalcemia, which further complicates the diagnosis. 5 Dr Reyes: What would be your approach to counseling a patient and her family with this diagnosis? ...
... 5 Dr Reyes: What would be your recommendation at this time for treatment? When feasible and appropriate, upfront surgical tumor removal and chemotherapy serves as therapy for patients with small cell 4 Even so, survival is described to be only 33% in stage IA disease. 6 Complete and adequate treatment typically follows with chemotherapy, whereas radiotherapy has not been routinely used as part of treatment. ...
... The present study identified frequent expression of claudin-10 in HGSC, in agreement with previous studies of other claudin family members [7][8][9][10][11][12][13][21][22][23][24]. However, in contrast to the previously reported overexpression of claudins 1, 3 and 7 in tubo-ovarian carcinoma effusions compared to surgical specimens [13], expression of claudin-10 was significantly higher in the latter. ...
... Claudin-7 protein expression was significantly related to poor chemotherapy response and shorter PFS in another study of ovarian specimens of various histotypes [22]. Claudin-4 expression was associated with significantly shorter PFS and OS in another study of ovarian specimens of various histotypes, though neither retained significance in multivariate analysis [23]. Conversely, claudin-4 expression was unrelated to PFS or OS in analysis of a larger series of HGSC [24]. ...
Article
Full-text available
The objective of this study was to analyze the expression and prognostic role of the gap junction protein claudin-10 in high-grade serous carcinoma (HGSC). Claudin-10 protein expression by immunohistochemistry was analyzed in 588 HGSC (414 effusions, 174 surgical specimens). Expression in mesotheliomas (n = 97; 47 effusions, 50 surgical specimens) was studied for comparative purposes. CLDN10 mRNA expression by quantitative RT-PCR (qRT-PCR) was analyzed in 40 HGSC effusions. Claudin-10 protein expression was found in 360/588 (61%) HGSC vs. 19/97 (20%) mesotheliomas (p < 0.001), and was higher in HGSC surgical specimens compared to effusions (p < 0.001). qRT-PCR confirmed the presence of CLDN10 mRNA in HGSC effusions. High (> 25%) claudin-10 expression in HGSC effusions was significantly associated with shorter overall survival (OS; p = 0.036) and progression-free survival (PFS; p = 0.045) in univariate analysis, and was an independent prognosticator of OS in multivariate analysis (p = 0.045). In conclusion, claudin-10 protein expression is higher in HGSC compared to mesothelioma, although the diagnostic power of this marker appear to be lesser than other claudin family members. Claudin-10 expression in HGSC effusions is marker of more aggressive disease.
... Increased CLDN4 expression thus results in the loss of tight junction integrity 14 , and we hypothesized that the junction protein would be exposed to a radioligand in the presence of disease due to the loss of junctional integrity in metastatic cells 12,15,16 . Previous literature indicates that 1) CLDN4 expression is also enhanced in prostate 17 , breast 18 and ovarian 19 cancers and 2) as pancreatic precancers advance, the expression level of CLDN4 increases (p = 0.001) 15 . Intense positive CLDN4 immunolabeling has been noted within virtually all primary (71/72 [99%]) and metastatic (49/49 [100%]) PDAC tissue samples and in 10 of 11 precancerous PanIN specimens 20 . ...
Article
Full-text available
Molecular imaging using positron emission tomography (PET) provides sensitive detection and mapping of molecular targets. While cancer-associated fibroblasts and integrins have been proposed as targets for imaging of pancreatic ductal adenocarcinoma (PDAC), herein, spatial transcriptomics and proteomics of human surgical samples are applied to select PDAC targets. We find that selected cancer cell surface markers are spatially correlated and provide specific cancer localization, whereas the spatial correlation between cancer markers and immune-related or fibroblast markers is low. Claudin-4 expression increases ~16 fold in cancer as compared with normal pancreas, and tight junction localization confers low background for imaging in normal tissue. We develop a peptide-based molecular imaging agent targeted to claudin-4 with accumulation to ~25% injected activity per cubic centimeter (IA/cc) in metastases and ~18% IA/cc in tumors. Our work motivates a data-driven approach to selection of molecular targets.
... In addition, we also created a prognosis prediction model with 9 selected IOCE-related genes including CLDN4, AKT2, CSPG5, ALDOC, LTA4H, PSMA2, PSMA5, TCIRG1, and ANO6. For instance, CLDN4 has been reported to be overexpressed and is associated with a poor prognosis in OC [37], which is similar to our results. A down-regulation of CLDN4 makes ovarian cancer cells vulnerable to Taxol and Carboplatin [38]. ...
Article
Full-text available
Background Cancer-derived exosomes contribute significantly in intracellular communication, particularly during tumorigenesis. Here, we aimed to identify two immune-related ovarian cancer-derived exosomes (IOCEs) subgroups in ovarian cancer (OC) and establish a prognostic model for OC patients based on immune-related IOCEs. Methods The Cancer Genome Atlas (TCGA) database was used to obtain RNA-seq data, as well as clinical and prognostic information. Consensus clustering analysis was performed to identify two IOCEs-associated subgroups. Kaplan-Meier analysis was used to compare the overall survival (OS) between IOCEs-high and IOCEs-low subtype. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were conducted to investigate the mechanisms and biological effects of differentially expressed genes (DEGs) between the two subtypes. Besides, an IOCE-related prognostic model of OC was constructed by Lasso regression analysis, and the signature was validated using GSE140082 as the validation set. Results In total, we obtained 21 differentially expressed IOCEs in OC, and identified two IOCE-associated subgroups by consensus clustering. IOCE-low subgroup showed a favorable prognosis while IOCE-high subgroup had a higher level of immune cell infiltration and immune response. GSEA showed that pathways in cancer and immune response were mainly enriched in IOCE-high subgroup. Thus, IOCE-high subgroup may benefit more in immunotherapy treatment. In addition, we constructed a risk model based on nine IOCE-associated genes (CLDN4, AKT2, CSPG5, ALDOC, LTA4H, PSMA2, PSMA5, TCIRG1, ANO6). Conclusion We developed a novel stratification system for OV based on IOCE signature, which could be used to estimate the prognosis as well as immunotherapy for OC patient.
... Exploring the detailed effects of various mod-ulators on different types of ovarian cancer and achieving accurate treatment of drugs will be crucial for the treatment of ovarian cancer. Accordingly, we summarize the degree and source of benign and malignant ovarian cancer according to the type of ovarian cancer (Table 1, Ref. [17][18][19][20][21][22][23][24][25][26][27][28][29][30][31][32]), in an effort to supplement the content of this review. ...
Article
Full-text available
Objective This review aims to provide some theoretical guidance for the precise treatment of ovarian cancer and the development of estrogen-related drugs. Mechanism Ovarian cancer is one of the leading causes of death in gynecological cancer patients, mainly affecting middle-aged and elderly women. It has the characteristics of hidden location, strong heterogeneity and lack of specific symptoms in the early stage. Numerous studies have shown that estrogen receptor (ER) plays an important role in different types of cancer, including ovarian cancer. Accordingly, the study of ER signaling pathways and related regulatory factors in ovarian cancer cells should help us understand the pathogenesis of ovarian cancer. Findings in Brief The expression of estrogen receptor subtypes is related to ovarian cancer gene and leads to ovarian cancer. Estrogen receptor modulators appear to be an important factor in the prognosis of patients with ovarian cancer after hormone therapy. Conclusions This review summarizes the regulatory mechanism of ER in the occurrence and development of ovarian cancer and outlines the specific role of estrogen receptor modulators (SERMs) in the treatment and prevention of ovarian cancer.
... Claudin-4 is often overexpressed in epithelial cancers (Hewitt et al., 2006) including breast (Kolokytha et al., 2014, Szasz et al., 2011, prostate (Landers et al., 2008, Li, 2021, gastric (Soini et al., 2006, Liu andLi, 2020), pancreatic (Li, 2021, Torres et al., 2018, bladder (Kuwada et al., 2015), lung (Li, 2021), colorectal (Fujiwara-Tani et al., 2018, Holczbauer et al., 2013 and ovarian (Hewitt et al., 2006, Hough et al., 2000, Li, 2021. Survival time in ovarian cancer patients is decreased when tumors have high levels of claudin-4 (Yoshida et al., 2011, Martin de la Fuente et al., 2018. Knockdown of claudin-4 has recently been found to sensitize ovarian cancer cells to paclitaxel and PARP inhibitors (Breed et al., 2019, Yamamoto et al., 2022 and targeting of claudin-3 and claudin-4 has been proposed as a chemotherapeutic strategy in ovarian cancer (Uthayanan and El-Bahrawy, 2022). ...
Article
Full-text available
Claudin-4, a protein with the structure of classic claudins most often found in cell-cell junctions, is frequently overexpressed in epithelial cancers where its localization has not been studied. In this study we aimed to find out where this membrane protein is localized in an ovarian tumor model, OVCAR3 cells, that express high levels of the protein. Immunohistochemical studies showed claudin-4 staining in a perinuclear region, at most plasma membranes and in cytoplasmic puncta. Native claudin-4 did not overlap with phosphorylated claudin-4, which was partially located in focal adhesions. Using claudin-4 BioID technology identified more precisely we confirmed that large amounts of claudin-4 are localized to the Golgi compartment, including in dispersed Golgi in cells where claudin-4 is partially knocked down and in dividing cells. Claudin-4 appears to be present in the vicinity of several types of cell-cell junctions, but there is no evidence that it forms tight junctions in these tumor cells. Both claudin-4, the Golgi marker GM130, and the plasma membrane receptor Notch2 were found in dispersed Golgi in dividing cells. This definition of the cellular architecture of claudin-4 should provide a framework for better understanding of the function of claudin-4 in tumor cells and its molecular interactions.
Preprint
Full-text available
Soft tissue sarcomas (STS) histopathological classification system has several conceptual caveats, impacting prognostication and treatment. The clinical and molecular–based tools currently employed to estimate prognosis also have limitations. Clinically driven molecular profiling studies may cover these gaps. We performed DNA sequencing (DNAseq) and RNA sequencing (RNAseq), portraying the molecular profile of 102 samples of 3 of the most common STS subtypes. The RNAseq data was analyzed using unsupervised machine learning models, unravelling previously unknown molecular patterns and identifying 4 well–defined transcriptomic clusters. These transcriptomic clusters have a clear prognostic value, a finding that was externally validated. This transcriptomic cluster–based classification′s prognostic value is superior to the prognostic accuracy of currently used clinical–based (SARCULATOR nomograms) and molecular–based (CINSARC) prognostication tools. The analysis of DNAseq data from the same cohort of samples revealed a plethora of unique and, in some cases, never documented molecular targets for precision treatment across different transcriptomic clusters.
Article
Metastasis from one neoplasm to another is referred to as tumor‐to‐tumor metastasis (TTM). TTM is rarely observed. Here, we present a patient with TTM from a thymic carcinoma to an ovarian mature teratoma. A 25‐year‐old woman, diagnosed with unresectable thymic carcinoma, presented with a cyst with a solid tumor component in her right ovary. Laparoscopic cystectomy of the right ovary revealed that the solid tumor was a distant metastasis of the thymic carcinoma in an ovarian mature teratoma. The possibility of malignant transformation of the ovarian mature teratoma was ruled out, enabling accurate staging of the thymic carcinoma. This case emphasizes the need for clinicians to consider TTM and the importance of pathological confirmation of TTM when investigating potential distant metastases.
Article
Background Celastrol is a biologically active compound with potent anti-tumor properties. However, the mechanism of action of celastrol in gastric cancer (GC) has not been fully elucidated. Methods To explore the specific mechanism of the effect of celastrol on GC cells. GC cells were transfected with forkhead box A1 (FOXA1) or claudin 4 (CLDN4), or short hairpin RNA targeting FOXA1. The expressions of FOXA1 and CLDN4 in GC cells were determined by quantitative reverse transcription PCR and Western blot. GC cell proliferation, migration, and invasion were measured by MTT assay and Transwell assay, respectively. The interaction between CLDN4 and FOXA1 was examined by luciferase reporter assay. Resluts CLDN4 and FOXA1 were upregulated in GC cells. Celastrol prevented the proliferation, migration, and invasion of GC cells by downregulating FOXA1 expression. Overexpression of FOXA1 or CLDN4 accelerated GC progression. CLDN4 overexpression also induced the activation of the expressions of phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathway. FOXA1 enhanced the transcription of CLDN4. Conclusion Celastrol regulated GC progression via targeting the FOXA1/CLDN4 axis to impede the PI3K/AKT pathway. Our study proposed a new mechanism of how celastrol inhibited tumorigenesis in GC, which provided evidence for the potential use of celastrol for anti-GC treatment.
Article
Epithelial ovarian cancer (EOC) is the gynecological malignant tumor of poorest prognosis and higher mortality rate. Chemotherapy is the base of high-grade serous ovarian cancer (HGSOC) treatment; however, it favors the emergence of chemoresistance and metastasis. Thus, there is an urge to search for new therapeutic targets, such as proteins related to cellular proliferation and invasion. Herein, we investigated the expression profile of claudin-16 (CLDN16 protein and CLDN16 transcript) and its possible functions in EOC. In silico analysis of CLDN16 expression profile was performed using data extracted from GENT2 and GEPIA2 platforms. A retrospective study was carried out with 55 patients to evaluate the expression of CLDN16. The samples were evaluated by immunohistochemistry, immunofluorescence, qRT-PCR, molecular docking, sequencing, and immunoblotting assays. Statistical analyzes were performed using Kaplan-Meier curves, one-way ANOVA, Turkey posttest. Data were analyzed using GraphPad Prism 8.0. In silico experiments showed that CLDN16 is overexpressed in EOC. 80.0% of all EOC types overexpressed CLDN16, of which in 87% of the cases the protein is restricted to cellular cytoplasm. CLDN16 expression was not related to tumor stage, tumor cells differentiation status, tumor responsiveness to cisplatin, or patients' survival rate. When compared to data obtained from in silico analysis regarding EOC stage and degree of differentiation, differences were found in the former but not in the later, neither in survival curves. CLDN16 expression in HGSOC OVCAR-3 cells increased by 1.95-fold (p < 0.001), 2.32-fold (p < 0.001), and 6.57-fold (p < 0.001) via PKC, PI3K, and estrogen pathways, respectively. Altogether, our results suggest that despite the low number of samples included in our in vitro studies, adding to the expression profile findings, we provided a comprehensive study of CLDN16 expression in EOC. Therefore, we hypothesize that CLDN16 is a potential target in the diagnosis and treatment of the disease.
Article
Full-text available
Resolving the molecular details of proteome variation in the different tissues and organs of the human body will greatly increase our knowledge of human biology and disease. Here, we present a map of the human tissue proteome based on an integrated omics approach that involves quantitative transcriptomics at the tissue and organ level, combined with tissue microarray-based immunohistochemistry, to achieve spatial localization of proteins down to the single-cell level. Our tissue-based analysis detected more than 90% of the putative protein-coding genes. We used this approach to explore the human secretome, the membrane proteome, the druggable proteome, the cancer proteome, and the metabolic functions in 32 different tissues and organs. All the data are integrated in an interactive Web-based database that allows exploration of individual proteins, as well as navigation of global expression patterns, in all major tissues and organs in the human body. Copyright © 2015, American Association for the Advancement of Science.
Article
Full-text available
Recent developments in the study of epithelial ovarian cancer have called into question the traditional views regarding the site of tumor initiation. Histopathologic studies and genomic analyses suggest that extra-ovarian sites, like the fallopian tube, may harbor the coveted cell of origin and could therefore contribute significantly to the development of high-grade serous ovarian carcinoma (HG-SOC). Our ability to validate these emerging genomic and pathologic observations and characterize the early transformation events of HG-SOC hinges on the development of novel model systems. Currently, there are only a handful of new model systems that are addressing these concerns. This review will chronicle the convergent evolution of these ovarian cancer model systems in the context of the changing pathologic and genomic understanding of HG-SOC.
Article
Objective: To provide a review of the basic mechanisms of drug resistance in ovarian cancer and novel strategies to modulate drug resistance for the general obstetrician-gynecologist. Data Sources: Relevant articles published through December 1996 were identified using the MEDLINE data base. Additional sources were found by cross-referencing. Methods of Study Selection: Publications identified by the search were reviewed and evaluated critically for their relevance to drug resistance in ovarian cancer. Tabulation, Integration, and Results: Each reference was reviewed for its contribution to the knowledge regarding mechanisms of chemotherapy resistance in ovarian cancer or strategies to modulate resistance. Conclusion: Ovarian cancer patients have high response rates to initial chemotherapy after cytoreductive surgery. However, most will develop resistance to chemotherapy during the course of their treatment. There are multiple mechanisms resulting in drug resistance. Strategies to modulate drug resistance include dose intensity, various pharmacologic agents, and gene therapy.
Article
Ovarian cancer is the leading cause of mortality by gynecologic cancers in Western countries. Many publications have suggested that age may be an independent prognostic factor in ovarian carcinoma. There are only few data concerning the impact of treatments and geriatric features within the elderly population. We collected data of older (≥70 years old) patients treated in our institution for an invasive ovarian carcinoma between 1995 and 2011. First we described usual clinical and pathological features for these patients, as well as their outcome. We compared these parameters with that of young (<70 years old) patients treated during the same period. We then observed geriatric features in our set: Eastern Cooperative Oncology Group performance status, number of medications, Charlson index, body mass index, hemoglobin, and glomerular filtration rate. We finally looked for prognostic factors specific of the elderly population. One hundred nine elderly patients were identified and compared with 488 younger cases. There was no difference concerning clinicopathologic data. Surgery was more frequently complete in young women (58% vs 41.7%), and older patients received less chemotherapy courses and less taxanes (38.4% vs 67.1%). Young patients had a longer overall survival (median, 65.2 vs 26.2 months, P = 8.5E-10, log-rank test). Multivariate analyses confirmed that age was an independent prognostic factor and that within the elderly set the International Federation of Gynecology and Obstetrics stage, surgery results, number of chemotherapy cycles administered and performance status had a significant prognostic value. No clear correlation could be observed between geriatric characteristics and treatments administration. Ovarian cancer prognosis is poorer for older women, but they are more frequently suboptimally treated. No correlation could be observed between geriatric factors and surgery or chemotherapy achievement. Treatment decision should be based on objective geriatric assessment in order to improve outcome in this population.
Article
Purpose: To estimate the effects of BRCA1 and BRCA2 mutations on ovarian cancer and breast cancer survival. Experimental design: We searched PubMed and EMBASE for studies that evaluated the associations between BRCA mutations and ovarian or breast cancer survival. Meta-analysis was conducted to generate combined HRs with 95% confidence intervals (CI) for overall survival (OS) and progression-free survival (PFS). Results: From 1,201 unique citations, we identified 27 articles that compared prognosis between BRCA mutation carriers and noncarriers in patients with ovarian or breast cancer. Fourteen studies examined ovarian cancer survival and 13 studies examined breast cancer survival. For ovarian cancer, meta-analysis demonstrated that both BRCA1 and BRCA2 mutation carriers had better OS (HR, 0.76; 95% CI, 0.70-0.83 for BRCA1 mutation carriers; HR, 0.58; 95% CI, 0.50-0.66 for BRCA2 mutation carriers) and PFS (HR, 0.65; 95% CI, 0.52-0.81 for BRCA1 mutation carriers; HR, 0.61; 95% CI, 0.47-0.80 for BRCA2 mutation carriers) than noncarriers, regardless of tumor stage, grade, or histologic subtype. Among patients with breast cancer, BRCA1 mutation carriers had worse OS (HR, 1.50; 95% CI, 1.11-2.04) than noncarriers but were not significantly different from noncarriers in PFS. BRCA2 mutation was not associated with breast cancer prognosis. Conclusions: Our analyses suggest that BRCA mutations are robust predictors of outcomes in both ovarian and breast cancers and these mutations should be taken into account when devising appropriate therapeutic strategies.
Article
A new paradigm for the pathogenesis of female pelvic cancer helps explain persistent problems in describing the development and diverse morphology of these neoplasms. This paradigm incorporates recent advances in the molecular pathogenesis of epithelial ovarian cancer (EOC) with new insights into the origin of these tumors. Correlated clinicopathologic and molecular genetic studies gave rise to a dualistic model that divides the various histologic types of EOCs into two broad categories designated type I and type II. Because serous carcinomas are the most common EOC and account for the vast majority of deaths, they form the subject of this review. Recent studies indicate that the precursor of ovarian high-grade serous carcinoma appears to develop from an occult intraepithelial carcinoma in the fimbria of the fallopian tube and involves the ovary secondarily. Another possible mechanism is implantation of normal fimbrial epithelium on the denuded ovarian surface at the site of rupture when ovulation occurs, causing the development of cortical inclusion cysts. The dualistic model serves as a framework for the study of ovarian cancer and can help investigators organize this complex group of neoplasms. It also facilitates the development of novel approaches to prevention, screening, and treatment of this devastating disease. Expected final online publication date for the Annual Review of Pathology: Mechanisms of Disease Volume 9 is February 28, 2014. Please see http://www.annualreviews.org/catalog/pubdates.aspx for revised estimates.