ArticlePDF Available

Immunohistochemical Evidence of Loss of PTEN Expression in Primary Ductal Adenocarcinomas of the Breast

Authors:

Abstract and Figures

Germline mutations in PTEN, encoding a dual-specificity phosphatase on 10q23.3, cause Cowden syndrome (CS), which is characterized by a high risk of breast and thyroid cancers. Loss of heterozygosity of 10q22-24 markers and somatic PTEN mutations have been found to a greater or lesser extent in a variety of sporadic component and noncomponent cancers of CS. Among several series of sporadic breast carcinomas, the frequency of loss of flanking markers around PTEN is approximately 30 to 40%, and the somatic intragenic PTEN mutation frequency is <5%. In this study, we analyzed PTEN expression in 33 sporadic primary breast carcinoma samples using immunohistochemistry and correlated this to structural studies at the molecular level. Normal mammary tissue had a distinctive pattern of expression: myoepithelial cells uniformly showed strong PTEN expression. The PTEN protein level in mammary epithelial cells was variable. Ductal hyperplasia with and without atypia exhibited higher PTEN protein levels than normal mammary epithelial cells. Among the 33 carcinoma samples, 5 (15%) were immunohistochemically PTEN-negative; 6 (18%) had reduced staining, and the rest were PTEN-positive. In the PTEN-positive tumors as well as in normal epithelium, the protein was localized in the cytoplasm and in the nucleus (or nuclear membrane). Among the immunostain negative group, all had hemizygous PTEN deletion but no structural alteration of the remaining allele. Thus, in these cases, an epigenetic phenomenon such as hypermethylation, -ecreased protein synthesis or increased protein degradation may be involved. In the cases with reduced staining, 5 of 6 had hemizygous PTEN deletion and 1 did not have any structural abnormality. Finally, clinicopathological features were analyzed against PTEN protein expression. Three of the 5 PTEN immunostain-negative carcinomas were also both estrogen and progesterone receptor-negative, whereas only 5 of 22 of the PTEN-positive group were double receptor-negative. The significance of this last observation requires further study.
Content may be subject to copyright.
Immunohistochemical Evidence of Loss of PTEN
Expression in Primary Ductal Adenocarcinomas of
the Breast
Aurel Perren,*
†‡
Liang-Ping Weng,*
Alexander H. Boag,
§
Ulricke Ziebold,
i
Kosha Thakore,*
Patricia L. M. Dahia,*
Paul Komminoth,
Jacqueline A. Lees,
i
Lois M. Mulligan,
§
George L. Mutter,** and
Charis Eng*
††
From the Clinical Cancer Genetics and Human Cancer Genetics
Programs,*Ohio State University Comprehensive Cancer Center,
Columbus, Ohio; the Dana-Farber Cancer Institute and Harvard
Medical School,
Boston, Massachusetts; the Department of
Pathology,
University of Zu¨ rich, Zu¨ rich, Switzerland; the
Departments of Pathology
§
and Paediatrics,
Queen’s University,
Kingston, Ontario, Canada; the Department of Biology,
i
Cancer
Research Center, Massachusetts Institute of Technology,
Cambridge, Massachusetts; the Department of Pathology,**
Brigham and Women’s Hospital and Harvard Medical School,
Boston, Massachusetts; and the CRC Human Cancer Genetics
Research Group,
††
University of Cambridge, Cambridge,
United Kingdom
Germline mutations in PTEN, encoding a dual-speci-
ficity phosphatase on 10q23.3, cause Cowden syn-
drome (CS), which is characterized by a high risk of
breast and thyroid cancers. Loss of heterozygosity of
10q22–24 markers and somatic PTEN mutations have
been found to a greater or lesser extent in a variety of
sporadic component and noncomponent cancers of
CS. Among several series of sporadic breast carcino-
mas, the frequency of loss of flanking markers
around PTEN is approximately 30 to 40%, and the
somatic intragenic PTEN mutation frequency is <5%.
In this study, we analyzed PTEN expression in 33
sporadic primary breast carcinoma samples using im-
munohistochemistry and correlated this to structural
studies at the molecular level. Normal mammary tis-
sue had a distinctive pattern of expression: myoepi-
thelial cells uniformly showed strong PTEN expres-
sion. The PTEN protein level in mammary epithelial
cells was variable. Ductal hyperplasia with and with-
out atypia exhibited higher PTEN protein levels than
normal mammary epithelial cells. Among the 33 car-
cinoma samples, 5 (15%) were immunohistochemi-
cally PTEN-negative; 6 (18%) had reduced staining,
and the rest were PTEN-positive. In the PTEN-positive
tumors as well as in normal epithelium, the protein
was localized in the cytoplasm and in the nucleus (or
nuclear membrane). Among the immunostain nega-
tive group, all had hemizygous PTEN deletion but no
structural alteration of the remaining allele. Thus, in
these cases, an epigenetic phenomenon such as hy-
permethylation, decreased protein synthesis or in-
creased protein degradation may be involved. In the
cases with reduced staining, 5 of 6 had hemizygous
PTEN deletion and 1 did not have any structural ab-
normality. Finally, clinicopathological features were
analyzed against PTEN protein expression. Three of
the 5 PTEN immunostain-negative carcinomas were
also both estrogen and progesterone receptor-nega-
tive, whereas only 5 of 22 of the PTEN-positive group
were double receptor-negative. The significance of
this last observation requires further study. (Am J
Pathol 1999, 155:1253–1260)
The tumor suppressor gene PTEN, encoding a dual-
specificity phosphatase, has been cloned and mapped
to chromosome 10q23.3.
1–3
Germline PTEN mutations
are found in the autosomal dominant Cowden syndrome
(CS), which is characterized by multiple hamartomas in-
volving many organ systems as well as an increased risk
of developing breast and thyroid cancers.
4,5
Loss of het-
erozygosity (LOH) of markers at 10q23–25 is a frequent
event (30 –50%) in endometrial cancer,
6–9
glioblas-
toma,
10
and breast cancer.
11–13
Somatic intragenic mu-
tations of PTEN are a frequent event in endometrial car-
cinomas,
6–9
malignant gliomas,
14–17
and melanomas.
18
However, unlike endometrial carcinoma and glioblas-
toma, only a very small fraction (,5%) of the 40% of
primary breast cancers showing allelic loss in this region
also have mutations in the remaining allele,
11–13,19
de-
Supported in part by grants from the American Cancer Society (RPG-97-
064-01-VM and RPG98-211-01-CCE), the Department of Defense Breast
Cancer Research Program (DAMD17-98-1-8058), the Concert for the
Cure (to C. E.), the National Cancer Institute (P30CA16058, Comprehen-
sive Cancer Center), and the Canadian Breast Cancer Research Initiative
(to L. M. M.). P. L. M. D. is a Postdoctoral Fellow of the Susan G. Komen
Breast Cancer Research Foundation (to C. E.) and A. P. is a Fellow of the
Lydia Hochstrasser-Stiftung, Zu¨ rich, Switzerland (to P. K.).
Accepted for publication June 15, 1999.
Address reprint requests to Charis Eng, Human Cancer Genetics Pro-
gram, Ohio State University Comprehensive Cancer Center, 690C Medi-
cal Research Facility, 420 W. 12th Avenue, Columbus, Ohio 43210. E-
mail: eng-1@medctr.osu.edu.
American Journal of Pathology, Vol. 155, No. 4, October 1999
Copyright © American Society for Investigative Pathology
1253
spite the fact that females with CS have a #50% lifetime
risk of developing breast cancer.
5,20,21
In contrast to
these analyses based on primary breast carcinomas,
initial studies using breast cancer cell lines seemed to
show that a large proportion have biallelic loss of
PTEN.
1,3
Investigators, therefore, questioned whether
loss of one PTEN allele (haploinsufficiency) is sufficient
for tumorigenesis or whether inactivation of the second
allele might occur through epigenetic rather than muta-
tional events.
We report a study of PTEN expression using immuno-
histochemical methods in a series of 33 primary human
breast tumors. This is a powerful method because it
provides an internal control comparing the staining of
tumor tissue to that of the adjacent normal breast tissue.
We also began to explore the association of PTEN ex-
pression with genomic PTEN status and clinicopatholog-
ical features.
Materials and Methods
Breast Carcinoma Samples
Paraffin blocks of 33 unselected sporadic primary ductal
breast carcinomas were drawn from the files of the King-
ston General Hospital (Kingston, ON, Canada). LOH
analysis with seven microsatellite markers known to map
to the 10q23 interval and flanking PTEN as well as PTEN
mutation analysis have been performed previously.
13
Of
the 33 women diagnosed with primary mammary adeno-
carcinomas, 4 were diagnosed before the age of 50. The
tumors ranged in size from 1 to 6 cm. There were 2 well
differentiated, 13 moderately differentiated, and 18
poorly differentiated tumors. Seven of the 13 women had
regional lymph node involvement at presentation.
Immunohistochemistry
The monoclonal antibody 6H2.1 raised against the last
100 C-terminal amino acids of PTEN (Ziebold and Lees,
unpublished) was used in all immunohistochemical
analyses.
The tissue samples were fixed by immersion in 10%
buffered formalin and embedded in paraffin according to
standard procedures.
22
Four-millimeter sections were cut
and mounted on Superfrost Plus slides (Fisher Scientific,
Pittsburgh, PA). Immunostaining was performed essen-
tially as described.
22–24
In summary, the sections were
deparaffinized and hydrated by passing through xylene
and a graded series of ethanol. Antigen retrieval was
performed for 20 minutes at 98°C in 0.01 mol/L sodium
citrate buffer, pH 6.4, in a microwave oven. Incubating
the sections in 0.3% hydrogen peroxide for 30 minutes
blocked endogenous peroxidase activity. After blocking
for 30 minutes in 0.75% normal horse serum the sections
were incubated with 6H2.1 (dilution 1:100) for 1 hour at
room temperature. The sections were washed in Tris-
buffered saline and then incubated with biotinylated
horse anti-mouse IgG followed by avidin peroxidase us-
ing the Vectastain ABC elite kit (Vector Laboratories,
Burlingame, CA). The chromogenic reaction was carried
out with 3–39diaminobenzidine using nickel cobalt am-
plification,
25
which gives a black product. After counter-
staining with Nuclear Fast Red (Rowley Biochemical,
Danvers, MA) and mounting, the slides were evaluated
under a light microscope. According to the amount of
staining, the tumors were divided in three groups: the
group assigned 11 showed increased or equal staining
intensity compared to the corresponding normal tissue;
the group assigned 1had decreased staining intensity;
and the group assigned 2had no trace of staining.
A series of commercially available cell lines with known
PTEN status was used as positive and negative controls
to prove antibody specificity by immunohistochemistry:
Balb C/3T3, Nalm6, DU145, MDA-MB-468, A172, and
PC3 (see Results). In addition, the U2OS osteosarcoma
cell line was transfected with full length PTEN cDNA
expression construct as a further positive control.
Incubating the sections in the absence of antibody as
well as preincubation during 2 hours at 37°C of the anti-
body with recombinant PTEN protein led to negative re-
sults (data not shown).
Western Blot Analysis
As biochemical proof of antibody specificity for PTEN,
total protein lysates were obtained
26
(Dahia, 1999 #955)
from a series of commercially available cell lines (Amer-
ican Type Culture Collection, Manassas, VA), for which
PTEN status is known: MCF-7, T47D, MDA-MB-435S,
ZR-75-1, BT-549, and MDA-MB-468 (see Results). In ad-
dition, as an additional positive control, the wild-type full
length human PTEN cDNA sequence was cloned into the
mammalian expression vector pUHD10-3, which con-
tains a tetracycline-suppressible promoter (Gossen,
1992 #1065), and stably transfected into the MCF7/T-off
(Clontech, Palo Alto, CA) breast cancer cell line. After 24
hours of tetracycline withdrawal for purposes of PTEN
induction, protein lysates were collected for Western blot
analysis as well. Western blot analysis was performed as
previously described,
26
except that 6H2.1 was used at a
1:250 dilution. Control antibody was against
a
-tubulin
and used at 1:1000 dilution.
LOH Analysis
All breast carcinoma samples have been previously eval-
uated for LOH with markers closely flanking, but not
within, PTEN.
13
In the event that our immunohistochemi-
cal results seemed to be discordant with the molecular
analyses, further LOH analysis was performed using
markers within PTEN itself as previously described.
26,27
Potential hemizygosity at the PTEN locus was assessed
by screening for a T/G polymorphism within PTEN intron
8 detected by differential digestion with the restriction
endonuclease HincII as previously described
27
and
the intragenic markers D10S2491, AFM086wg9, and
D10S2492.
1254 Perren et al
AJP October 1999, Vol. 155, No. 4
Results
Specificity of Monoclonal Antibody 6H2.1
Because this study relied on a monoclonal antibody,
6H2.1, specific recognition of PTEN by this antibody is
crucial. Western blot analysis using a series of breast
cancer lines with known PTEN status and the 6H2.1 anti-
PTEN monoclonal antibody demonstrated the specificity
of this antibody (Figure 1). Western analysis of three
PTEN1/1lines, MCF-7, T-47D, and MDA-MB-435S, re-
vealed a single band at the molecular weight predicted
for PTEN. After induction of MCF-7/PTEN, increased ex-
pression of PTEN was evidenced by an increased band
intensity (Figure 1). In contrast, ZR-75-1, with a hemizy-
gous deletion of PTEN and a missense mutation in the
remaining allele, yielded a weak band of the expected
size. BT-549 and MDA-MB-468, which are null for PTEN,
had no signal. No nonspecific bands were noted. Control
blot with anti-
a
-tubulin antibody revealed signals for all
lines.
To test the suitability of the antibody for immunohisto-
chemistry, we used PTEN-transfected U2OS cells as well
as a series of cell lines expressing PTEN (Balb C/3T3,
Nalm6, DU145) as positive controls. MDA-MB-468, a breast
cancer cell line with a hemizygous deletion of PTEN and a
truncating mutation of the remaining allele, A172, a glioblas-
toma cell line with loss of one PTEN allele and a truncating
mutation in exon 2 of the remaining allele and PC3, a
prostate cell line with homozygous deletion of PTEN,
were used as negative controls (data not shown).
PTEN Immunohistochemistry in Primary
Breast Carcinomas
Samples from 33 sporadic primary breast carcinomas,
which had been examined previously for LOH of markers
flanking PTEN as well as somatic PTEN mutations,
13
were
subjected to immunohistochemical analysis using a
monoclonal antibody, 6H2.1, raised against the terminal
100 amino acids of human PTEN. Of the 33 total cancers,
29 had accompanying normal tissue; in each of the 29
samples, the normal glandular epithelium showed immu-
noreactivity to 6H2.1. Interestingly, there was a distinctive
staining pattern in the normal tissue. The myoepithelial
cells of the normal ducts showed the strongest signal with
a nuclear predominance (Figure 2B). In contrast, the
amount of staining in the epithelial cell layer was variable.
Areas of epithelial ductal hyperplasia with and without
atypia stained more strongly than the normal epithelia
(Figure 2A). Endothelial cells, especially within neovas-
cular capillaries, and nerves showed strong PTEN ex-
pression and were useful as internal positive controls.
Of 33 breast carcinoma samples, 5 (15%) lost all PTEN
immunoreactivity and showed negative immunostaining,
graded 2(Table 1 and Figure 2, E and F). In each of
these 5 cases, adjacent non-neoplastic glands (Figure
2F) as well as enclosed non-neoplastic ducts (Figure 2 E)
stained positively. Interestingly, the cells within the des-
moplastic reaction surrounding each of these 5 carcino-
mas had high PTEN expression. Six of the 33 (18%)
breast cancer specimens stained weakly, graded 1,in
Figure 1. Western blot of 7 breast cancer cell lines using the anti-PTEN monoclonal antibody 6H2.1 (left panel) and using the anti-
a
-tubulin antibody as a control
(right panel). MCF-7, T-47D, and MDA-MB-435S have endogenous PTEN. BT-549 and MDA-MB-468 are PTEN-null. ZR-75-1 has monoallelic PTEN deletion and
a missense mutation on the remaining allele. MCF-7/PTEN is the MCF-7 line transfected with a wild-type PTEN construct and a tetracycline-inducible promoter
after withdrawal of tetracycline and, hence, induced expression of PTEN.
PTEN Immunohistochemistry in Breast Carcinoma 1255
AJP October 1999, Vol. 155, No. 4
comparison to the normal tissue (Table 1 and Figure 3).
One of these tumors (Sample 40, Table 1) showed pos-
itive immunostaining in the intraductal component,
whereas the adjacent invasive component lost almost all
PTEN protein expression (Figure 3A). The remaining 22
(66%) tumors stained positively, graded 11 (increased
staining compared to normal glands). All these tumors
showed homogeneous PTEN immunoreactivity through-
out the examined section. PTEN immunoreactivity in
these 22 tumors as well as their corresponding normal
and hyperplastic breast tissue involved the cytoplasmic
and nuclear (most likely nuclear membrane) compart-
ment of the cells.
Comparison of Immunohistochemical and
Structural Mutation Data
Immunohistochemical evidence of PTEN expression was
absent or weak in a total of 11 (33%) of 33 breast carci-
nomas. These breast carcinomas had been previously
examined for LOH of markers flanking PTEN and also for
intragenic PTEN mutations;
13
40% demonstrated LOH
but there were no intragenic PTEN mutations or biallelic
deletion. Whether there is a one-to-one concordance be-
tween molecular and immunohistochemical observations
is further explored in this report.
LOH analysis for markers in the 10q22–24 interval was
previously performed using seven microsatellite markers
(centromeric to telomeric): D10S579, D10S215,
D10S1765, D10S541, D10S1735, D10S1739, and
D10S564.
13
PTEN lies between D10S1765 and D10S541,
a genetic distance of 1 cM but a physical distance of only
several hundred kilobasepairs. For purposes of this
study, to compare the immunohistochemical data to the
LOH data, PTEN was considered to be physically deleted
only when one or more immediately flanking (informative)
markers centromeric and telomeric of PTEN showed
LOH. Using this strict and conservative interpretation for
monoallelic PTEN deletion, 6 of the tumors were shown to
have a loss of one allele of the PTEN gene, another 7 were
shown to have a loss flanking one side of (which may or
Figure 2. A: Ductal hyperplasia ( case 58) with increased staining in the epithelial layer (original magnification, 360). B: Normal breast glands (case 46) with
predominantly nuclear staining in the myoepithelial layer (original magnification, 360). C(case 48) and D(case 43): Ductal carcinoma with strong PTEN staining
(11, original magnification, 330). E: Ductal PTEN-negative carcinoma (arrowhead, case 58) and surrounding normal duct (arrow). Original magnification, 330.
F: Ductal PTEN-negative carcinoma (arrowhead, case 46) with non-neoplastic normal duct (arrow). Original magnification, 330.
Table 1. Correlation between PTEN Immunostaining and
PTEN and/or 10q22-23 LOH
PTEN
Immuno
11
PTEN
Immuno
1
PTEN
Immuno
2
LOH 59Markers 4* 2 4
LOH 39Markers 1* 2 5
ROH Flank Markers 18 2* 0
Total Tumors 22 6 5
Correlation between PTEN immunostaining and LOH of 59and/or 39
flanking markers.
Concordance 82%.
LOH, loss of heterozygosity; ROH, retention of heterozygosity.
*Apparent discordance 18%.
1256 Perren et al
AJP October 1999, Vol. 155, No. 4
may not include) PTEN. For these latter 7 tumors, poten-
tial hemizygosity at the PTEN locus was further assessed
by screening for a T/G polymorphism within PTEN intron
8 (IVS8132T/G), detected by differential digestion with
the restriction endonuclease HincII, and the intragenic
polymorphic markers AFM086wg9, D10S2491, and
D10S2492. AFM086wg9 lies in intron 2 of PTEN. The likely
intragenic marker order is centromere 2D10S2491 2
AFM086wg9 2D10S2492/IVS8132T/G 2telomere
(Marsh and Eng, unpublished).
Of the 5 breast carcinomas that exhibited no immuno-
histochemical evidence of PTEN expression (graded 2),
4 showed extensive LOH of markers flanking PTEN and
hence, PTEN itself (Table 1, Column 3 and Table 3). The
fifth carcinoma had LOH on the telomeric side (D10S541)
of PTEN. Further molecular analysis revealed retention of
heterozygosity at AFM086wg9 but LOH at the IVS8132T/G
polymorphism, suggesting hemizygous deletion of the 39
end of PTEN. Therefore, all 5 breast carcinomas that had
negative PTEN immunostaining also had hemizygous
PTEN deletion (Table 3). None of these 5 had biallelic
deletion of PTEN nor did they have a second intragenic
PTEN hit, ie, mutation of the remaining allele.
Of the 6 carcinomas that had weak PTEN immunostain-
ing, graded 1, 4 had been previously shown to have LOH
of markers flanking one side or the other of PTEN and 2
Figure 3. Cases with weak staining (arrows in Cand F, non-neoplastic duct; arrow in E, blood vessel). A: Ductal carcinoma (case 40) showing no staining
(graded 2) in the invasive component (top) adjacent to immunostain-positive intraductal component (bottom). B: Case 66. C: Case 59. D: Case 57. E: Case 45.
Original magnification, 330.
PTEN Immunohistochemistry in Breast Carcinoma 1257
AJP October 1999, Vol. 155, No. 4
showed no LOH of flanking markers (Tables 2 and 3).
Further LOH analysis within PTEN revealed that the 4
carcinomas with LOH of markers flanking one side of the
gene also had LOH of at least one of the intragenic
markers (Table 2). Thus, these 4 tumors with decreased
immunostaining seemed to have hemizygous deletion of
PTEN or at least part of it. In the remaining two carcino-
mas without LOH of markers immediately flanking the
gene, further analyses within the gene were uninformative
or showed retention of heterozygosity (Tumors 45 and 57,
Tables 2 and 3). In all likelihood, PTEN might not be
altered at the structural level in that particular tumor.
Among the remaining 22 carcinomas that showed im-
munohistochemical evidence of strong PTEN expression
(increased staining compared to normal mammary
glands), 18 (82%) showed no LOH and biallelic presence
of PTEN was demonstrated (Table 1). There were 4 tu-
mors that seemed to be immunostained (grade 11), yet
showed LOH flanking PTEN (Tables 2 and 3). However, it
should be noted that 3 of these 4 tumors had LOH of
D10S1765 immediately centromeric of PTEN but with ei-
ther retention of heterozygosity or noninformativeness at
D10S541 immediately 39of the gene. Further LOH anal-
ysis within PTEN corroborates the previous observations
(Table 2): in tumor 6, 39markers within the gene showed
retention of heterozygosity and a 59marker (AFM086wg9)
showed LOH; in tumor 5, where D10S1765 showed LOH,
markers within the gene (AFM086wg9 and D10S2492)
and 39of the gene (D10S541) all showed retention of
heterozygosity. Similarly, tumor 9, which had LOH at
D10S1765, had 3 of 4 intragenic markers with retention of
heterozygosity. Tumor 53 was unusual in that both
D10S1765 and D10S541 had LOH, although molecular
analysis demonstrated all 4 intragenic markers with re-
tained heterozygosity.
Correlation of PTEN Immunohistochemistry and
Clinicopathological Parameters
PTEN immunostaining status was compared with such
clinicopathological parameters as age at diagnosis, size
of primary tumor, tumor grade, lymph node status, and
estrogen receptor and progesterone receptor status. Be-
cause of the relatively small numbers, especially in the
context of subset analyses, no conclusions could be
drawn with confidence from our observed correlations.
The most interesting association seemed to be that be-
tween PTEN expression and hormone receptor status
(Table 4). Three of the 5 carcinomas (67%) that had no
PTEN protein were estrogen and progesterone receptor-
negative compared to 5 of 22 (23%; P,0.05 Fisher’s
exact test) in the PTEN-immunopositive samples. All 6
carcinomas that had weak PTEN staining were estrogen
and progesterone receptor-positive. Other trends are
also noteworthy. Although there were only 2 grade I tu-
mors, both had high PTEN expression. All 5 tumors that
were 1.5 cm or smaller had high levels of PTEN protein.
Table 2. Analysis of Correlation between PTEN Immunostaining and PTEN Intragenic LOH in Cases with Decreased
Immunostaining and Apparently Discordant Tumors
Tumor
Immuno-
staining score
PTEN
10q22-23 Markers
S1765 S2491 AFM086 S2492 IVS8 S541
41 11 LOH NI LOH ROH ROH NI
52 11 LOH NI ROH ROH N/A ROH
53 11 LOH ROH ROH ROH ROH LOH
50 11 LOH ROH ROH NI ROH NI
40 1LOH N/A NI N/A LOH ROH
59 1LOH N/A LOH N/A LOH ROH
66 1ROH NI LOH N/A N/A LOH
57 1ROH LOH LOH N/A N/A ROH
55 1ROH NI NI N/A LOH LOH
45 1ROH NI ROH NI ROH ROH
Tumor numbers correspond to those of Feilotter et al.
13
LOH, loss of heterozygosity; ROH, retention of heterozygosity; NI, not informative (germline homozygosity at marker); N/A, not applicable or not
done.
Table 3. Summary of PTEN Expression by
Immunohistochemistry Compared to Molecular
Analysis
PTEN Expression LOH* ROH
PTEN250
PTEN151
PTEN11 121
*LOH of both flanking markers or a minimum of LOH of one
intragenic marker.
Table 4. Estrogen/Progesterone Receptor Status of Breast
Carcinomas by PTEN Immunostaining Status
PTEN IHC status ER/PR 2ER/PR 1
Negative (2)3 2
Decreased (1)0 7
Positive (11)5 16
ER, estrogen receptor; PR, progesterone receptor.
An equivocal positive receptor status (n55) was scored as a
positive.
1258 Perren et al
AJP October 1999, Vol. 155, No. 4
Discussion
In this first report of immunohistochemical analyses of
PTEN expression in sporadic primary breast carcinomas,
we found that 33% of these tumors had either no or
decreased expression of PTEN, which generally ap-
peared to correlate with structural monoallelic deletion of
the gene. Although it is understandable that tumors with
monoallelic loss of PTEN have decreased PTEN expres-
sion at the protein level, one must explain the 5 samples
with no immunoreactivity and structural PTEN hemizygos-
ity. None of these samples was found to have intragenic
PTEN mutations in the remaining allele, either. It is more
than plausible, therefore, that an epigenetic phenome-
non, such as hypermethylation of the promoter region
28
and decreased protein synthesis or increased protein
turnover,
26
might be inactivating the remaining allele.
Similarly, for the tumor (case 45) with decreased staining
but no structural PTEN deletion, similar hypotheses may
be raised. Other explanations include point mutations in
the putative promoter of the remaining allele or normal
tissue contamination of the breast samples, thus giving
pseudo-hemizygosity in the face of real homozygous de-
letion. The latter can be discarded because very careful
microdissection of the carcinoma components was per-
formed by a pathologist with extensive experience in
microdissection. Further, since the pattern of all positive
and negative tumors was homogeneous, regional PTEN
deletions in tumor subclones are very unlikely. Con-
versely, the observation of rare immunopositive tumors
(n54) which appear to have LOH of flanking markers
can be plausibly explained as well: at least in 3 informa-
tive tumors, no deletion of the gene proper or no deletion
of most of the 39end of the gene has occurred. Hence,
the monoclonal antibody, which is raised against the C
terminus of PTEN, would still immunostain these samples
positively. In this situation, therefore, incomplete 59dele-
tion of PTEN might still be associated with translation of a
truncated immunocompetent PTEN protein. In summary,
while structural deletion or mutation of PTEN can lead to
decreased PTEN protein levels, other mechanisms which
lead to complete loss of PTEN expression seem to be
prominent as well, at least in the breast carcinoma model.
Whether loss of PTEN expression is an early or late
event in breast carcinogenesis is still controversial, al-
though preliminary reports suggest that it is a late
event.
11
The observation that loss of PTEN expression is
correlated with a negative estrogen and progesterone
status and that both grade I tumors had strong PTEN
expression also strengthen this hypothesis. There is no
doubt that these latter clinicopathological observations
need to be investigated further. Nonetheless, these data
in toto argue that despite the observation that germline
PTEN mutations cause Cowden syndrome,
4
somatic
PTEN mutation or functional loss of PTEN expression is
associated with tumor progression and not tumor initia-
tion, at least in the breast cancer model. It is also clear
from our and other data that breast carcinogenesis does
not rely uniformly on the involvement of the PTEN path-
way, although how PTEN plays a role in various aspects
of normal development and in the pathogenesis of breast
carcinoma is not straightforward.
Acknowledgments
We thank Jeff FitzGerald for technical assistance, Dr.
Oliver Gimm for expert administrative assistance and
many members of CE’s laboratory for critical review of the
manuscript.
References
1. Li J, Yen C, Liaw D, Podsypanina K, Bose S, Wang S, Puc J, Miliaresis
C, Rodgers L, McCombie R, Bigner SH, Giovanella BC, Ittman M,
Tycko B, Hibshoosh H, Wigler MH, Parsons R: PTEN, a putative
protein tyrosine phosphatase gene mutated in human brain, breast,
and prostate cancer. Science 1997, 275:1943–1947
2. Li D-M, Sun H: TEP1, encoded by a candidate tumor suppressor
locus, is a novel protein tyrosine phosphatase regulated by transform-
ing growth factor B. Cancer Res 1997, 57:2124 –2129
3. Steck PA, Pershouse MA, Jasser SA, Yung WKA, Lin H, Ligon AH,
Langford LA, Baumgard ML, Hattier T, Davis T, Frye C, Hu R, Swed-
lund B, Teng DHF, Tavtigian SV: Identification of a candidate tumor
suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in
multiple advanced cancers. Nat Genet 1997, 15:356 –362
4. Liaw D, Marsh DJ, Li J, Dahia PLM, Wang SI, Zheng Z, Bose S, Call
KM, Tsou HC, Peacocke M, Eng C, Parsons R: Germline mutations of
the PTEN gene in Cowden disease, an inherited breast and thyroid
cancer syndrome. Nat Genet 1997, 16:64 –67
5. Eng C, Peacocke M: PTEN and inherited hamartoma-cancer syn-
dromes. Nat Genet 1998, 19:223
6. Levine RL, Cargile CB, Blazes MS, van Rees B, Kurman RJ, Ellenson
LH: PTEN mutations and microsatellite instability in complex atypical
hyperplasia, a precursor lesion to uterine endometrioid carcinoma.
Cancer Res 1998, 58:3254 –3258
7. Maxwell GL, Risinger JL, Gumbs C, Shaw H, Bentley RC, Barrett JC,
Berchuck A, Futreal PA: Mutation of the PTEN tumor suppressor gene
in endometrial hyperplasias. Cancer Res 1998, 58:2500 –2503
8. Tashiro H, Blazes MS, Wu R, Cho KR, Bose S, Wang SI, Li J, Parsons
R, Ellenson LH: Mutations in PTEN are frequent in endometrial carci-
noma but rare in other common gynecological malignancies. Cancer
Res 1997, 57:3935–3940
9. Kong D, Suzuki A, Zou T-T, Sakurada A, Kemp LW, Wakatsuki S,
Yokohama T, Yamakawa H, Furukawa T, Sato M, Ohuchi N, Sato S,
Yin J, Want S, Abraham JM, Souza RF, Smolinksi KN, Meltzer SJ, Horii
A: PTEN1 is frequently mutated in primary endometrial carcinomas.
Nat Genet 1997, 17:143–144
10. Maier D, Zhang ZW, Taylor E, Hamou MF, Gratzl O, van Meir EG,
Scott RJ, Merlo A: Somatic deletion mapping on chromosome 10 and
sequence analysis of PTEN/MMAC1 point to the 10q25–26 region as
the primary target in low-grade and high-grade gliomas. Oncogene
1998, 16:3331–3335
11. Bose S, Wang SI, Terry MB, Hibshoosh H, Parsons R: Allelic loss of
chromosome 10q23 is associated with tumor progression in breast
carcinomas. Oncogene 1998, 17:123–127
12. Singh B, Ittman MM, Krolewski JJ: Sporadic breast cancers exhibit
loss of heterozygosity on chromosome segment 10q23 close to the
Cowden disease locus. Genes Chromosomes Cancer 1998, 21:166
171
13. Feilotter HE, Coulon V, McVeigh JL, Boag AH, Dorion-Bonnet F,
Duboue´ B, Latham WCW, Eng C, Mulligan LM, Longy M: Analysis of
the 10q23 chromosomal region and the PTEN gene in human spo-
radic breast carcinoma. Br J Cancer 1999, 79:718 –723
14. Du¨ rr E-M, Rollbrocker B, Hayashi Y, Peters N, Meyer-Puttlitz B, Louis
DN, Schramm J, Wiestler OD, Parsons R, Eng C, von Deimling A:
PTEN mutations in gliomas and glioneuronal tumors. Oncogene 1998,
16:2259 –2264
15. Wang SI, Puc J, Li J, Bruce JN, Cairns P, Sidransky D, Parsons R:
Somatic mutations of PTEN in glioblastoma multiforme. Cancer Res
1997, 57:4183–4186
PTEN Immunohistochemistry in Breast Carcinoma 1259
AJP October 1999, Vol. 155, No. 4
16. Rasheed BKA, Stenzel TT, McLendon RE, Parsons R, Friedman AH,
Friedman HS, Bigner DD, Bigner SH: PTEN gene mutations are seen
in high-grade but not in low-grade gliomas. Cancer Res 1997, 37:
4187– 4190
17. Bostro¨ m J, LudwigCobbers JMJ, Wolter M, Tabatabai G, Weber RG,
Lichter P, Collins VP, Reifenberger G: Mutation of the PTEN (MMAC1)
tumor suppressor gene in a subset of glioblastomas but not in me-
ningiomas with loss of chromosome arm 10q. Cancer Res 1998,
58:29 –33
18. Tsao HS, Zhang X, Benoit E, Haluska FG: Identification of PTEN/
MMAC1 alterations in uncultured melanomas and melanoma cell
lines. Oncogene 1998, 16:3397–3402
19. Rhei E, Kang L, Bogomoliniy F, Federici MG, Borgen PI, Boyd J:
Mutation analysis of the putative tumor suppressor gene PTEN/
MMAC1 in primary breast carcinomas. Cancer Res 1997, 57:3657–
3659
20. Starink TM, van der Veen JPW, Arwert F, de Waal LP, de Lange GG,
Gille JJP, Eriksson AW: The Cowden syndrome: a clinical and genetic
study in 21 patients. Clin Genet 1986, 29:222–233
21. Eng C: Genetics of Cowden syndrome: through the looking glass of
oncology. Int J Oncol 1998, 12:701–710
22. Eng C, Thomas GA, Neuberg DS, Mulligan LM, Healey CS, Houghton
C, Frilling A, Raue F, Williams ED, Ponder BAJ: Mutation of the RET
proto-oncogene is correlated with RET immunostaining in subpopu-
lations of cells in sporadic medullary thyroid carcinoma. J Clin Endo-
crinol Metab 1998, 83:4310 –4313
23. Komminoth P, Roth J, Schroder S, Saremaiani P, Heitz PU: Overlap-
ping expression of immunohistochemical markers and synaptophysin
mRNA in pheochromocytomas and adenocortical carcinomas. Lab
Invest 1995, 72:424– 431
24. Heitz PU, Kasper M, Polak JM, Kloppel G: Pancreatic endocrine
tumors. Hum Pathol 1982, 13:263–271
25. Werner M, von Wasiekewski R, Komminoth P: Antigen retrieval, signal
amplification and intensification in immunohistochemistry. Histochem
Cell Biol 1996, 105:253–260
26. Dahia PLM, Aguiar RCT, Alberta J, Kum J, Caron S, Sills H, Marsh
DJM, Freedman A, Ritz J, Stiles C, Eng C: PTEN is inversely corre-
lated with the cell survival factor PKB/Akt and is inactivated by di-
verse mechanisms in haematologic malignancies. Hum Mol Genet
1999, in press
27. Dahia PLM, Marsh DJ, Zheng Z, Zedenius J, Komminoth P, Frisk T,
Wallin G, Parsons R, Longy M, Larsson C, Eng C: Somatic deletions
and mutations in the Cowden disease gene, PTEN, in sporadic thy-
roid tumors. Cancer Res 1997, 57:4710 –4713
28. Whang YE, Wu X, Suzuki H, Reiter RE, Tran C, Vessella RL, Said JW,
Isaacs WB, Sawyers CL: Inactivation of the tumor suppressor PTEN/
MMAC1 in advanced human prostate cancer through loss of expres-
sion. Proc Natl Acad Sci USA 1998, 95:5246 –5250
1260 Perren et al
AJP October 1999, Vol. 155, No. 4
... While total laryngectomy was performed in 100 (71.4%) cases, the remaining cases were partial laryngectomy. Of the cases, 38 The median follow-up was 38 months (1-126 months). The overall survival rate of the cases was 74.7%, the disease-free survival rate was 78.7%. ...
... In their evaluation with the semi-quantification method used in many studies in the literature, they accepted lower staining or no staining as low expression compared to the surrounding epithelium and stroma, and high PTEN expression if staining with equal or more intensity. [34,[36][37][38][39][40][41] Low PTEN expression was detected in 59.9% of all cases and in 62.2% of laryngeal carcinomas. They stated that low PTEN expression is associated with an aggressive clinical course. ...
Article
Full-text available
The prognosis of laryngeal cancer is affected by clinicopathological factors. Because of that, an effective prognostic marker is very valuable in managing the clinical process. The p53 evaluation method, used in the literature recently, was used for the first time in laryngeal cancer. We evaluated PTEN with 2 methods with the highest significance in the literature on laryngeal cancer. All demographic and histopathological data from 140 laryngeal cancers were compared with p53 and PTEN expressions and survival. p53 staining patterns were classified as wild and mutant. PTEN expression was evaluated according to the staining intensity named PTEN1 and according to the proportion of stained cells named PTEN2. In the series, 93.6% were males, and the mean survival was 38 months. 69.3% of cases were p53 mutants. PTEN loss was found to be 85.7% and 57.9%, respectively. Tumor size and thyroid cartilage invasion for PTEN1 and age for p53 were identified as independent predictive factors (P < .01). Advanced age, total laryngectomy, and extranodal spread were independent poor prognostic factors for overall survival and the presence of subglottic involvement, perineural invasion, and extranodal spread were for disease-free survival (P < .01). This is the first study in which the new p53 classification was used in laryngeal cancer, and will contribute significantly to the literature with differences from the previous evaluation patterns. Evaluation of PTEN based on staining intensity is more appropriate compared to the percentage of stained cells.
... In addition, salient metabolic differences among ZR-75 (more phosphorylative) and MCF-7 (highly glycolytic) cells have been reported, and the use of specific genes and energy pathways probably impart more aggressiveness to ZR-75 compared to MCF-7 [37]. Nonetheless, such different behavior might also depend on the fact that, different from MCF-7 and T-47D, ZR-75 are PTEN-mutant cells [38], showing basal hyperactive AKT signaling [39], further emphasized by the condition of Tam resistance, which results, in turn, in very low levels of FoxO3a [19]. In this scenario, FoxO3a transient expression might not be sufficient to counteract such a sustained PI3K/AKT pathway. ...
Article
Full-text available
Tamoxifen-resistant breast cancer cells (TamR-BCCs) are characterized by an enhanced metabolic phenotype compared to tamoxifen-sensitive cells. FoxO3a is an important modulator of cell metabolism, and its deregulation has been involved in the acquisition of tamoxifen resistance. Therefore, tetracycline-inducible FoxO3a was overexpressed in TamR-BCCs (TamR/TetOn-AAA), which, together with their control cell line (TamR/TetOn-V), were subjected to seahorse metabolic assays and proteomic analysis. FoxO3a was able to counteract the increased oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) observed in TamR by reducing their energetic activity and glycolytic rate. FoxO3a caused glucose accumulation, very likely by reducing LDH activity and mitigated TamR biosynthetic needs by reducing G6PDH activity and hindering NADPH production via the pentose phosphate pathway (PPP). Proteomic analysis revealed a FoxO3a-dependent marked decrease in the expression of LDH as well as of several enzymes involved in carbohydrate metabolism (e.g., Aldolase A, LDHA and phosphofructokinase) and the analysis of cBioPortal datasets of BC patients evidenced a significant inverse correlation of these proteins and FoxO3a. Interestingly, FoxO3a also increased mitochondrial biogenesis despite reducing mitochondrial functionality by triggering ROS production. Based on these findings, FoxO3a inducing/activating drugs could represent promising tools to be exploited in the management of patients who are refractory to antiestrogen therapy.
... As a lipid phosphatase, PTEN is responsible for the dephosphorylation of phosphatidylinositol 3,4,5-triphosphate (PIP 3 ) in PI3Ks/AKT pathway to regulate cell proliferation, regulation of metabolism, neuronal function and genome instability [2][3][4][5][6][7][8][9]. Pathogenic variation in PTEN damages PTEN function and causes high cancer risk that the carriers have up to 80% lifetime risk of developing breast, thyroid, kidney and brain cancer [10][11][12][13][14][15]. Therefore, revealing the function of the genetic variants in PTEN is crucial in understanding the mechanisms of the diseases caused by the mutated PTEN and for clinical applications. ...
Article
Full-text available
Phosphatase and tensin homolog (PTEN), a tumor suppressor with dual phosphatase properties, is a key factor in PI3K/AKT signaling pathway. Pathogenic germline variation in PTEN can abrogate its ability to dephosphorylate, causing high cancer risk. Lack of functional evidence lets numerous PTEN variants be classified as variants of uncertain significance (VUS). Utilizing Molecular Dynamics (MD) simulations, we performed a thorough evaluation for 147 PTEN missense VUS, sorting them into 66 deleterious and 81 tolerated variants. Utilizing replica exchange molecular dynamic (REMD) simulations, we further assessed the variants situated in the catalytic core of PTEN’s phosphatase domain and uncovered conformational alterations influencing the structural stability of the phosphatase domain. There was a high degree of agreement between our results and the variants classified by Variant Abundance by Massively Parallel Sequencing, saturation mutagenesis, multiplexed functional data and experimental assays. Our extensive analysis of PTEN missense VUS should benefit their clinical applications in PTEN-related cancer. Significance statement Classification of PTEN variants affecting its lipid phosphatase activity is important for understanding the roles of PTEN variation in the pathogenesis of hereditary and sporadic malignancies. Of the 3000 variants identified in PTEN, 1296 (43%) were assigned as VUS. Here, we applied MD and REMD simulations to investigate the effects of PTEN missense VUS on the structural integrity of the PTEN phosphatase domain consisting the WPD, P and TI active sites. We classified a total of 147 missense VUS into 66 deleterious and 81 tolerated variants by referring to the control group comprising 54 pathogenic and 12 benign variants. The classification was largely in concordance with these classified by experimental approaches.
... Loss of PTEN protein expression has not been reported to correlate with stage, tumor grade, (21). In addition, a significant relationship was found with PR loss (22). Contrary to this findings, in our study, there was a significant and strong correlation between the downregulation of the PTEN gene expression and the positivity of ER and PR values in both blood and tumor tissues. ...
Article
Objectives Cowden syndrome (CS) is a multisystem disease with an elevated lifetime risk of internal malignancy. We aim to assess the role of PTEN immunostain as a screening test for CS in a variety of common CS-associated neoplasms, with a particular focus on cutaneous tumors. Methods We retrospectively searched for patients meeting criteria for CS and/or demonstrating germline PTEN mutation from 2008 to 2022. We then performed PTEN immunostains on tumors of these patients as well as control cases. Results Our study included 30 patients with CS who had a total of 25 CS-associated malignancies (13 thyroid, 8 breast, and 4 endometrial carcinomas). Specifically, there were 11 patients with biopsy-confirmed CS-associated cutaneous neoplasms, including 1 patient with multiple trichilemmomas and 3 with multiple sclerotic fibromas. In total, 45 CS-associated tumors (6 trichilemmomas, 7 sclerotic fibromas, 5 thyroid carcinomas, 18 adenomatous thyroid nodules, 6 breast carcinomas, and 3 endometrial carcinomas) and 31 non-CS cases (9 trichilemmomas, 5 sclerotic fibromas, 8 adenomatous thyroid nodules, and 3 thyroid, 3 breast, and 3 endometrial carcinomas) were available for PTEN immunohistochemical staining. PTEN expression was lost in 43 (96%) of 45 CS-associated lesions and retained in 30 (97%) of 31 sporadic tumors. The overall sensitivity and specificity of PTEN loss of expression as a screening test for CS were 96% and 97%, respectively. Conclusions PTEN immunohistochemistry on CS-associated tumors, especially trichilemmomas, can serve as a readily accessible and cost-effective screening test for CS.
Article
Full-text available
PTEN is a major tumor suppressor gene frequently mutated in human tumors, and germline PTEN gene mutations are the molecular diagnostic of PTEN Hamartoma Tumor Syndrome (PHTS), a heterogeneous disorder that manifests with multiple hamartomas, cancer predisposition, and neurodevelopmental alterations. A diversity of translational and splicing PTEN isoforms exist, as well as PTEN C-terminal truncated variants generated by disease-associated nonsense mutations. However, most of the available anti-PTEN monoclonal antibodies (mAb) recognize epitopes at the PTEN C-terminal tail, which may introduce a bias in the analysis of the expression of PTEN isoforms and variants. We here describe the generation and precise characterization of anti-PTEN mAb recognizing the PTEN C2-domain, and their use to monitor the expression and function of PTEN isoforms and PTEN missense and nonsense mutations associated to disease. These anti-PTEN C2 domain mAb are suitable to study the pathogenicity of PTEN C-terminal truncations that retain stability and function but have lost the PTEN C-terminal epitopes. The use of well-defined anti-PTEN mAb recognizing distinct PTEN regions, as the ones here described, will help to understand the deleterious effects of specific PTEN mutations in human disease.
Article
Full-text available
Background Survival rates of head and neck squamous cell carcinoma (HNSCC) have only marginally improved in the last decades. Hence there is a need for predictive biomarkers for long-time survival that can help to guide treatment decisions and might lead to the development of new therapies. The phosphatidylinositol 3-kinase (PI3K)/AKT/mTOR signaling pathway is the most frequently altered pathway in HNSCC, genes are often mutated, amplificated and overexpressed causing aberrant signaling affecting cell growth and differentiation. Numerous genetic alterations of upstream and downstream factors have currently been clarified. However, their predictive value has yet to be established. Therefore we assess the predictive value of p-mTOR, p-ERK and PTEN expression. Methods Tissue microarrays (TMA’s) of HPV-negative patients with oropharyngeal (n = 48), hypopharyngeal (n = 16) or laryngeal (n = 13) SCC, treated with primary chemoradiation (cisplatin/carboplatin/cetuximab and radiotherapy), were histologically stained for p-mTOR, PTEN and p-ERK. Expression was correlated to overall survival (OS), disease free survival (DFS) and locoregional control (LRC). Also p-mTOR was histologically stained in a separate cohort of HNSCC organoids (n = 8) and correlated to mTOR-inhibitor everolimus response. Results High p-mTOR expression correlated significantly with worse OS in multivariate analysis in the whole patient cohort [Hazar Ratio (HR) 1.06, 95%CI 1.01–1.11, p = 0.03] and in the cisplatin/carboplatin group with both worse OS (HR 1.09, 95%CI 1.02–1.16, p = 0.02) and DFS (HR 1.06, 95%CI 1.00–1.12, p = 0,04). p-ERK expression correlated significantly with DFS in univariate analysis in the whole patient cohort (HR 1.03, 95%CI 1.00–1.05, p = 0.04) and cisplatin/carboplatin group (HR 1.03, 95%CI 1.00–1.07, p = 0.04). PTEN-expression did not correlate with OS/DFS/LRC. Better organoid response to everolimus correlated significantly to higher p-mTOR expression (Rs = − 0.731, p = 0.04). Conclusions High p-mTOR expression predicts and high p-ERK expression tends to predict worse treatment outcome in HPV negative HNSCC patients treated with chemoradiation, providing additional evidence that these markers are candidate prognostic biomarkers for survival in this patient population. Also this study shows that the use of HNSCC organoids for biomarker research has potential. The role of PTEN expression as prognostic biomarker remains unclear, as consistent evidence on its prognostic and predictive value is lacking.
Article
Phosphatase and tensin homolog (PTEN) is a tumour suppressor gene and has a role in inhibiting the oncogenic AKT signalling pathway by dephosphorylating phosphatidylinositol 3,4,5-triphosphate (PIP3) into phosphatidylinositol 4,5-bisphosphate (PIP2). The function of PTEN is regulated by different mechanisms and inactive PTEN results in aggressive tumour phenotype and tumorigenesis. Identifying targeted therapies for inactive tumour suppressor genes such as PTEN has been challenging as it is difficult to restore the tumour suppressor functions. Therefore, focusing on the downstream signalling pathways to discover a targeted therapy for inactive tumour suppressor genes has highlighted the importance of synthetic lethality studies. This review focuses on the potential synthetic lethality genes discovered in PTEN-inactive cancer types. These discovered genes could be potential targeted therapies for PTEN-inactive cancer types and may improve the treatment response rates for aggressive types of cancer.
Article
Full-text available
Mapping of homozygous deletions on human chromosome 10q23 has led to the isolation of a candidate tumor suppressor gene, PTEN, that appears to be mutated at considerable frequency in human cancers. In preliminary screens, mutations of PTEN were detected in 31% (13/42) of glioblastoma cell lines and xenografts, 100% (4/4) of prostate cancer cell lines, 6% (4/65) of breast cancer cell lines and xenografts, and 17% (3/18) of primary glioblastomas. The predicted PTEN product has a protein tyrosine phosphatase domain and extensive homology to tensin, a protein that interacts with actin filaments at focal adhesions. These homologies suggest that PTEN may suppress tumor cell growth by antagonizing protein tyrosine kinases and may regulate tumor cell invasion and metastasis through interactions at focal adhesions.
Article
The PTEN gene, located on 10q23, has recently been implicated as a candidate tumor suppressor gene in brain, breast and prostate tumors. In the present study, 123 brain tumors, including various grades and histological types of gliomas occurring in children and adults, were analyzed for PTEN mutations by SSCP assay and sequencing. Mutations in the PTEN gene were found in 13 of 42 adult glioblastomas and 3 of 13 adult anaplastic astrocytomas, whereas none of the 21 low-grade adult gliomas or the 22 childhood gliomas of all grades showed mutations. The single medulloblastoma with a mutation was a recurrent tumor that also possessed a p53 mutation. High-grade adult gliomas with PTEN mutations included cases that also contained gene amplification or p53 gene mutations, as well as cases that did not contain either of these abnormalities. There was no obvious relationship between presence of PTEN mutation and survival; however, there was a tendency for PTEN mutations to occur in older age group patients. This analysis suggest that PTEN gene mutations are restricted to high-grade adult gliomas and that this abnormality is independent of the presence or absence of gene amplification or p53 gene mutation in these tumors.
Article
One hundred twenty-five pancreatic endocrine tumors were analyzed by immunocytochemistry using various antisera. Twenty-three of 27 insulinomas, 10 of 10 PP-omas (PP: pancreatic polypeptide) and 15 of 30 “nonsecreting” tumors were benign, whereas 8 of 13 glucagonomas, 16 of 24 gastrinomas, and 16 of 21 VIP-omas (VIP: vasoactive intestinal polypeptide) were malignant. As a rule, the hormone secreted by the tumor and causing clinical symptoms could be localized by immunocytochemistry. Fifty of 95 active tumors were found to contain cells immunoreactive to peptide(s) not causing clinical symptoms, and 15 of 30 “nonsecreting” tumors were shown to be multicellular. By electron microscopy more than one cell type could be identified in 12 tumors. Histologically, the growth pattern of the tumors was very variable and distribution of immunoreactive cells was distinctly patchy. Radioimmunoassay on extracts of 20 of 27 tumors confirmed the presence of peptides visualized by immunocytochemistry. In 17 of 22 specimens, groups of endocrine cells in close contact with ductules were found in the pancreatic parenchyma distant from the tumor. Pancreatic endocrine tumors probably arise from the pancreatic ductular epithelium. They are often multicellular, producing and sometimes secreting more than one hormone or hormone-like substance. They represent highly complex biologic systems in which the interrelationship of various gastrointestinal-pancreatic hormones can be studied.
Article
An analysis of the findings in 21 patients with the Cowden syndrome or the multiple hamartoma syndrome is presented. The Cowden syndrome is a cancer-associated genodermatosis with characteristic mucocutaneous findings and a wide array of associated abnormalities including a high incidence of breast cancer in female patients. Genetic studies confirmed autosomal dominant inheritance with a high penetrance in both sexes and moderate interfamilial and intrafamilial differences in the expressivity of a number of symptoms. Familial occurrence was present in 4 of the 7 families. There was a strong predominance of female patients (6:1), which may be fortuitous. Mucocutaneous changes were the most constant (100% incidence) and characteristic findings; they almost invariably became manifest in the second decade. Four of our 18 female patients (22%) were treated for breast cancer, a lower incidence than reported previously. No increased incidence of other types of malignancies was found. Craniomegaly (high head circumference) was found to be the most common extracutaneous manifestation (80% incidence); craniomegaly appears to be an important early marker. We also found high incidences of gastrointestinal polyps (approximately 60%) and cutaneous fibromas (76%), while the incidence of thyroid abnormalities, thus far regarded as the most common extracutaneous finding, was similar to that reported previously (62%). G-banded karyotype and preliminary DNA-repair studies revealed no clear abnormalities. No linkage with the loci of HLA, and immunoglobulin haplotypes was found.
Article
One hundred twenty-five pancreatic endocrine tumors were analyzed by immunocytochemistry using various antisera. Twenty-three of 27 insulinomas, 10 of 10 PP-omas (PP: pancreatic polypeptide) and 15 of 30 "nonsecreting" tumors were benign, whereas 8 of 13 glucagonomas, 16 of 24 gastrinomas, and 16 of 21 VIP-omas (VIP: vasoactive intestinal polypeptide) were malignant. As a rule, the hormone secreted by the tumor and causing clinical symptoms could be localized by immunocytochemistry. Fifty of 95 active tumors were found to contain cells immunoreactive to peptide(s) not causing clinical symptoms, and 54 of 30 "nonsecreting" tumors were shown to be multicellular. By electron microscopy more than one cell type could be identified in 12 tumors. Histologically, the growth pattern of the tumors was very variable and distribution of immunoreactive cells was distinctly patchy. Radioimmunoassay on extracts of 20 of 27 tumors confirmed the presence of peptides visualized by immunocytochemistry. In 17 of 22 specimens, groups of endocrine cells in close contact with ductules were found in the pancreatic parenchyma distant from the tumor. Pancreatic endocrine tumors probably arise from the pancreatic ductular epithelium. They are often multicellular, producing and sometimes secreting more than one hormone or hormone-like substance. They represent highly complex biologic systems in which the interrelationship of various gastrointestinal-pancreatic hormones can be studied.
Article
The differential diagnosis of cortical versus medullary tumors of the adrenal gland may be a problem in diagnostic pathology. Conflicting results have been reported about the distribution of various immunohistochemical markers in the normal as well as neoplastic adrenal cortex and medulla. Archival, formaldehyde-fixed, and paraffin-embedded material comprising 27 adrenocortical carcinomas (ACC, meeting Weiss' histologic criteria), 28 pheochromocytomas (PCC), and adjacent nontumorous tissue (13 glands) were analyzed by immunogold-silver staining for the expression of polysialic acid (poly Sia), cytokeratins (CK), synaptophysin (SYN), chromogranin A (CrgA), somatostatin (SOM), calcitonin (CT), and the "adrenocortical marker" D11. Further, SYN mRNA expression was studied by nonradioactive in situ hybridization using digoxigenin-labeled oligonucleotide probes. In the normal adrenal gland, poly Sia was exclusively detected in the medulla and in cortical nerves. In ACCs, SYN immunoreactivity was present in 23 of 27 tumors (85%), D11 was present in 22 of 27 (81%), poly Sia was present focally in 8 of 27 (29%), and CK was present in 7 of 27 (25%). Synthesis of SYN in ACCS was demonstrated by mRNA in situ hybridization. Immunoreactivity for CrgA, SOM, or CT was not detectable. No difference of the marker profiles was seen in the nine clinically hormone-producing (three androgen, five corticosteroid, one mineralocorticoid) ACCs compared with the clinically silent tumors. In PCCs, all 28 tumors were immunoreactive for poly Sia and SYN; CrgA was detectable in 26 of 28 (93%), CT was detectable in 6 of 28 (21%), and SOM was detectable in 2 of 28 (7%) tumors. Staining for D11 or CK was undetectable. By immunohistochemistry, no distinction was possible between the 4 clinically malignant, the 6 multiple endocrine neoplasia type 2A-associated, and the 18 sporadic benign PCCs. Poly Sia of the neural cell adhesion molecule is consistently detected in normal adrenal medullary cells as well as in PCCs, and is occasionally focally expressed in ACCs. CrgA, CK, and D11 are reliable markers to immunohistochemically distinguish ACC from PCC. Immunoreactivity for SYN and detection of its mRNA in the majority of ACCs indicates the existence of tumor cells sharing both cortical and medullary features. Such focal neuroendocrine differentiation in ACCs can lead to confusion with PCCs.
Article
In this overview we emphasize new methods of improving immunohistochemical results in formaldehyde-fixed tissue samples. The benefit of heat-induced antigen retrieval in demasking of concealed epitopes is demonstrated. We provide guidance on the influence of heat-induced antigen retrieval in commonly applied monoclonal and polyclonal antibodies. Moreover, we show the promising methods of signal amplification using biotinylated tyramine and signal intensification of diaminobenzidine reaction products by metallic ions.
Article
Deletions involving regions of chromosome 10 occur in the vast majority (> 90%) of human glioblastoma multiformes. A region at chromosome 10q23-24 was implicated to contain a tumour suppressor gene and the identification of homozygous deletions in four glioma cell lines further refined the location. We have identified a gene, designated MMAC1, that spans these deletions and encodes a widely expressed 5.5-kb mRNA. The predicted MMAC1 protein contains sequence motifs with significant homology to the catalytic domain of protein phosphatases and to the cytoskeletal proteins, tensin and auxilin. MMAC1 coding-region mutations were observed in a number of glioma, prostate, kidney and breast carcinoma cell lines or tumour specimens. Our results identify a strong candidate tumour suppressor gene at chromosome 10q23.3, whose loss of function appears to be associated with the oncogenesis of multiple human cancers.
Article
Cowden disease (CD) is an autosomal dominant cancer predisposition syndrome associated with an elevated risk for tumours of the breast, thyroid and skin. Lhermitte-Duclos disease (LDD) cosegregates with a subset of CD families and is associated with macrocephaly, ataxia and dysplastic cerebellar gangliocytomatosis. The common feature of these diseases is a predisposition to hamartomas, benign tumours containing differentiated but disorganized cells indigenous to the tissue of origin. Linkage analysis has determined that a single locus within chromosome 10q23 is likely to be responsible for both of these diseases. A candidate tumour suppressor gene (PTEN) within this region is mutated in sporadic brain, breast and prostate cancer. Another group has independently isolated the same gene, termed MMAC1, and also found somatic mutations throughout the gene in advanced sporadic cancers. Mutational analysis of PTEN in CD kindreds has identified germline mutations in four of five families. We found nonsense and missense mutations that are predicted to disrupt the protein tyrosine/dual-specificity phosphatase domain of this gene. Thus, PTEN appears to behave as a tumour suppressor gene in the germline. Our data also imply that PTEN may play a role in organizing the relationship of different cell types within an organ during development.