throbber
American Journal of Pathology, Vol. 157, No. 4, October 2000
`Copyright © American Society for Investigative Pathology
`
`Short Communication
`
`Mutation and Expression Analyses Reveal
`Differential Subcellular Compartmentalization of
`PTEN in Endocrine Pancreatic Tumors Compared to
`Normal Islet Cells
`
`Aurel Perren,*† Paul Komminoth,*
`Parvin Saremaslani,* Claudia Matter,*
`Seraina Feurer,* Jacqueline A. Lees,‡
`Philipp U. Heitz,* and Charis Eng†§
`From the Department of Pathology,* University of Zu¨ rich, Zurich,
`Switzerland; the Clinical Cancer Genetics and Human Cancer
`Genetics Programs,† Comprehensive Cancer Center, and Division
`of Human Genetics, Department of Internal Medicine, The Ohio
`State University, Columbus, Ohio; the Department of Cancer
`Biology,‡ Massachusetts Institute of Technology, Cambridge,
`Massachusetts; and the Cancer Research Campaign Human
`Cancer Genetics Research Group,§ University of Cambridge,
`Cambridge, United Kingdom
`
`The pathogenesis of sporadic endocrine pancreatic
`tumors (EPTs) is still primarily unknown. Compara-
`tive genomic hybridization studies revealed loss of
`10q in a significant number (nine of 31) of EPTs. The
`tumor suppressor gene PTEN lies on 10q23, and so, is
`a candidate to play some role in EPT pathogenesis.
`Germline PTEN mutations are found in Cowden and
`Bannayan-Riley-Ruvalcaba syndromes, whereas so-
`matic mutations and deletions are found in a variety
`of sporadic cancers. The mutation and expression
`status of PTEN in EPTs has not yet been examined.
`Mutation analysis of the entire coding region of PTEN
`including splice sites was performed in 33 tumors,
`revealing one tumor with somatic L182F (exon 6).
`Loss of heterozygosity of the 10q23 region was de-
`tected in eight of 15 informative malignant (53%) and
`in none of seven benign EPTs. PTEN expression was
`assessed in 24 available EPTs by immunohistochem-
`istry using a monoclonal anti-PTEN antibody. Of these
`24, 23 tumors showed strong immunoreactivity for
`PTEN. Only the EPTs with PTEN mutation lacked PTEN
`protein expression. Although normal islet cells always
`exhibited predominantly nuclear PTEN immunostain-
`ing, 19 of 23 EPTs had a predominantly cytoplasmic
`PTEN expression pattern. Exocrine pancreatic tissue
`
`was PTEN-negative throughout. PTEN mutation is a rare
`event in malignant EPTs and PTEN protein is ex-
`pressed in most (23 of 24) EPTs. Thus, intragenic
`mutation or another means of physical loss of PTEN
`is rarely involved in the pathogenesis of EPTs. In-
`stead, either an impaired transport system of PTEN to
`the nucleus or some other means of differential com-
`partmentalization could account for impaired PTEN
`function. Loss of heterozygosity of the 10q23 region is
`a frequent event in malignant EPTs and might suggest
`several hypotheses: a different tumor suppressor
`gene in the vicinity of PTEN might be principally
`involved in EPT formation; alternatively, 10q loss,
`including PTEN, seems to be associated with malig-
`nant transformation, but the first step toward neopla-
`sia might involve altered subcellular localization of
`(Am J Pathol 2000, 157:1097–1103)
`PTEN.
`
`The etiology and pathogenesis of sporadic endocrine
`pancreatic tumors (EPTs) remain primarily unknown. Mu-
`tations of the MEN1 gene responsible for the autosomal
`dominantly inherited MEN 1 syndrome are found only in
`;15 to 30% of sporadic EPTs.1,2 Oncogenes (such as
`FOS, C-MYC, M-MYC, and SIS) or tumor suppressor
`genes (such as TP53 or RB1) which are frequently acti-
`vated or mutated in other human tumors seem not to be
`involved in the neoplastic transformation of EPTs.3,4
`Comparative genomic hybridization analysis of EPTs re-
`vealed losses of Y, 6q, 11q, 3p, 3q, 11p, 6p, 10q, and Xq.
`The frequency of 10q loss was 25% of all EPTs with a
`
`Supported in part by the American Cancer Society RPG98 –211-01CCE
`(to C. E.) and the National Cancer Institute (P30 CA16058 to The Ohio
`State University Comprehensive Cancer Center).
`Accepted for publication July 1, 2000.
`Address reprint requests to Charis Eng, Human Cancer Genetics Pro-
`gram, The Ohio State University Comprehensive Cancer Center, 420 W.
`12th Avenue, Room 690C Medical Research Facility, Columbus, OH
`43210. E-mail: eng-1@medctr.osu.edu.
`
`1097
`
`Roxane Labs., Inc.
`Exhibit 1020
`Page 001
`
`

`
`Perren et al
`1098
`AJP October 2000, Vol. 157, No. 4
`
`frequency as high as seven of nine of nonfunctioning
`EPTs.5 The tumor suppressor gene PTEN maps to
`10q23.3.6 – 8
`Germline PTEN mutations are responsible for the au-
`tosomal dominantly inherited Cowden and Bannayan-
`Riley Ruvalcaba syndromes as well as a Proteus-like
`syndrome.9 –13 Relatively high frequencies of somatic in-
`tragenic PTEN mutations and deletions are found in non-
`cultured endometrial carcinomas14 –17 and malignant gli-
`omas.18,19 Despite the frequency of structural PTEN
`mutations in noncultured endometrial carcinomas, we
`have shown that epigenetic silencing of PTEN not only
`plays a prominent role in its pathogenesis, but also in the
`pathogenesis of the earliest endometrial precancers.17
`Further, we have demonstrated that loss of PTEN protein
`expression in the absence of mutations occurs in breast
`carcinogenesis.20 To examine whether 10q loss in EPTs
`points to involvement of the tumor suppressor PTEN, we
`analyzed a series of 33 EPTs for intragenic mutations and
`deletions of PTEN and PTEN protein expression.
`
`MaterialsandMethods
`
`Tumor Samples
`
`Thirty-three EPTs were drawn from the files of the Depart-
`ment of Pathology, University Hospital Zu¨rich, Switzer-
`land. The tumors were classified according to the most
`recent World Health Organization classification.21 They
`comprised 19 insulinomas (six malignant, 12 benign, one
`MEN 1-associated, and one of uncertain clinical behav-
`ior), two malignant glucagonomas, three malignant VIP-
`omas, three malignant gastrinomas (one MEN 1-associ-
`ated), and six nonfunctioning (five malignant, one
`benign) EPTs. Except for one gastrinoma and one insu-
`linoma as noted, all of the EPTs were sporadic and not
`associated with MEN 1 or von Hippel-Lindau (VHL) syn-
`drome. Comparative genomic hybridization analysis of
`these tumors has been performed previously.5
`Fresh-frozen tissue was snap-frozen in liquid nitrogen
`and stored at 280°C. Paraffin samples were fixed by
`immersion in 4% buffered formalin and embedded in
`paraffin according to standard procedures.
`
`DNA Extraction
`
`Genomic DNA from fresh-frozen tissue was isolated us-
`ing the D-5000 Purgene DNA Isolation Kit (Gentra Sys-
`tems, Minneapolis, MN) according to the manufacturer’s
`instructions. DNA from these fresh-frozen tissues was
`used for mutation analysis. Where no nonneoplastic
`fresh-frozen tissue was available, DNA was extracted
`from paraffin blocks for loss of heterozygosity (LOH) analy-
`sis. For this purpose, 10-mm sections of formalin-fixed par-
`affin-embedded tumor specimens were microdissected
`and DNA extraction was performed as described.2,22
`
`LOH Analysis
`
`To assess LOH of the PTEN region at 10q23, we used the
`centromeric marker D10S579,
`the intragenic markers
`AFMa086wg9 and D10S2491, and the telomeric marker
`D10S1735. Polymerase chain reaction (PCR) was per-
`formed according to standard procedures and the prod-
`ucts were electrophoresed through polyacrylamide gels
`containing 7 mol/L urea followed by silver staining as
`previously described.2 LOH was defined as a complete
`absence or reduced signal of one of the constitutional
`alleles in the tumor tissue compared to the corresponding
`nonneoplastic tissue.
`
`Mutation Analysis
`
`PCR amplification was performed in a 50-ml mixture 13
`PCR buffer (Perkin Elmer Europe, Rotkreuz, Switzerland)
`containing 400 ng of template DNA, 200 mmol/L of dNTP
`(Roche Diagnostics, Rotkreuz, Switzerland), 1 mmol/L
`each of intronic-based primers flanking each exon (Table
`1), and 0.2 ml ofTaq polymerase (AmpliTaq Gold; Perkin
`Elmer Europe). A touchdown PCR was performed with
`denaturation at 95°C for 1 minute, annealing at 55 to 48°C
`(with 1°C decrements per cycle) for 1 minute, and exten-
`sion at 72°C for 1 minute followed by additional 30 cycles
`at 48° annealing temperature and a final extension at 72°
`for 10 minutes. Varying concentrations of MgCl2 and
`dimethylsulfoxide were used (Table 1).
`For the single-strand conformation polymorphism anal-
`ysis, 10 ml of denatured PCR products in stop buffer (95%
`formamide, 20 mmol/L ethylenediaminetetraacetic acid,
`0.05% xylene cyanol, 0.05% bromophenol blue) were
`loaded onto nondenaturing polyacrylamide gels. Electro-
`phoresis was performed at 40 W for 5 hours at room
`temperature. After electrophoresis, the DNA was visual-
`ized by silver staining as described.23
`
`Immunohistochemistry
`
`The monoclonal anti-human PTEN antibody 6H2.1 raised
`against the last 100 C-terminal amino acids was used in
`immunohistochemical analysis.20,24 Specificity and
`all
`characterization of 6H2.1 has previously been demon-
`strated by Western blot, immunohistochemistry on cell
`lines with known PTEN expression status as well as the
`ability of cold peptide to compete off immunostaining on
`paraffin-embedded sections.17,20,25
`Twenty-four cases where paraffin blocks were avail-
`able were subjected to immunohistochemistry (Table 2).
`Four-mm sections were cut and mounted on Superfrost
`Plus slides (Fischer Scientific, Pittsburgh, PA). Immuno-
`staining was performed as described.20 A semiquantita-
`tive score was given to the nuclear and cytoplasmic
`staining of tumor and normal tissue: 2, 1, and 11.
`
`Roxane Labs., Inc.
`Exhibit 1020
`Page 002
`
`

`
`1099
`PTEN Expression in Endocrine Pancreatic Tumors
`AJP October 2000, Vol. 157, No. 4
`
`Table 1. Primers and PCR Conditions
`
`Primer
`
`PTEN 1
`
`PTEN 2
`PTEN 3
`
`PTEN 4
`
`PTEN 5a
`
`PTEN 5b
`
`PTEN 6
`
`PTEN 7
`
`PTEN 8b
`
`PTEN 8b
`
`PTEN 9
`
`Position
`AH007803
`
`2229–2379
`
`10242–10412
`18382–18528
`
`24099–24248
`
`26126–26283
`
`26232–26417
`
`32123–32324
`
`37865–38093
`
`40894–41137
`
`41084–41200
`
`45308–45549
`
`Sequence
`
`Length
`(bp)
`
`TCTGCCATCTCTCTCTCCT
`AGAGGAGCAGCCGCAGAAATG
`TTTCAGATATTTCTTTCCTTA
`TAATTTCAAATGTTAGCTCAT
`AAGATATTTGCAAGCATACAA
`GTTTGTTAGTATTAGTACTTT
`ACAACATAGTACAGTACATTC
`TATTCTGAGGTTATCTTTTA
`CTTTCCAGCTTTACAGTGAA
`GCTAAGTGAAGATGACAATCA
`AGGAAAAACATCAAAAAATAA
`TTGGCTTCTCTTTTTTTTCTG
`ACATGGAAGGATGAGAATTTC
`CCTGTGAAATAATACTGGTATG
`CTCCCAATGAAAGTAAAGTACA
`TTAAATATGTCATTTCATTTCTTTTTC
`CTTTGTCTTTATTTGCTTTGT
`GTGCAGATAATGACAAGGAATA
`ACACATCACATACATACAAGTC
`TTCATTTTAAATTTTCTTTCT
`TGGTGTTTTATCCCTCTTGAT
`
`151
`
`171
`147
`
`150
`
`158
`
`186
`
`202
`
`229
`
`244
`
`117
`
`242
`
`Condition
`
`2.0 mmol/L MgCl2, 10% DMSO
`
`2.5 mmol/L MgCl2
`2.5 mmol/L MgCl2
`
`2.5 mmol/L MgCl2
`
`2.5 mmol/L MgCl2, 10% DMSO
`
`2.5 mmol/L MgCl2, 10% DMSO
`
`2.5 mmol/L MgCl2
`
`—
`
`—
`
`—
`
`2 mmol/L MgCl2
`
`DMSO, dimethylsulfoxide.
`
`Table 2. Clinical Data and Results
`
`Clinical data
`No.
`Sex
`Age
`
`Tumor type
`
`CGH†
`
`ICH‡
`
`D10S579
`
`LOH - Analysis§
`AFM86
`D10S2491
`
`D10S1735
`
`Mutation analysis
`
`1
`2
`3
`4
`5
`6
`7
`8
`9
`10
`11
`12
`13
`14
`15
`16
`17
`18
`19
`20
`21
`22
`23
`24
`25
`26
`27
`28
`29
`30
`31
`32
`33
`
`M
`M
`M
`F
`F
`?
`M
`F
`F
`F
`F
`M
`F
`M
`M
`M
`M
`M
`F
`F
`F
`M
`F
`M
`F
`M
`F
`F
`F
`M
`M
`M
`M
`
`93
`36
`58
`81
`26
`?
`62
`22
`59
`35
`42
`53
`52
`61
`62
`52
`58
`56
`64
`56
`48
`44
`33
`51
`34
`53
`46
`59
`36
`57
`63
`20
`41
`
`ins
`ins
`ins
`ins
`ins
`Ins
`ins
`ins
`ins
`ins
`ins
`ins
`non f
`ins, mal
`ins, mal
`ins, mal
`ins, mal
`ins, mal
`ins, mal
`gluc, mal
`gluc, mal
`non f, mal
`non f, mal
`non f, mal
`non f, mal
`non f, mal
`vip, mal
`vip, mal
`vip, mal
`vip, mal
`gast, mal
`ins, MEN1
`gast, MEN1
`
`*
`ni
`
`ni
`ni
`*
`
`ni
`ni
`
`*
`ni
`*
`ni
`*
`ni
`ni
`f
`*
`ni
`ni
`
`*
`f
`
`*
`M
`
`C
`
`C
`N/C
`C
`C
`
`C/M
`N/C
`C
`C/M
`C
`N/C
`C
`C
`C
`neg
`N/C
`C/M
`C
`C
`C
`C
`C/M
`C
`
`C
`
`10q-
`
`10q rg-
`
`10q-
`10q-
`10q rg-
`10q rg-
`10q-
`10q-
`10q-
`10q-
`
`ni
`ni
`
`ni
`ni
`ni
`
`ni
`ni
`
`ni
`M
`ni
`M
`ni
`ni
`f
`ni
`ni
`*
`ni
`
`ni
`f
`
`ni
`M
`
`M
`M
`
`M
`M
`*
`
`M
`M
`
`ni
`M
`ni
`f
`f
`ni
`f
`f
`f
`ni
`ni
`
`*
`f
`
`ni
`M
`
`ni
`ni
`
`ni
`M
`ni
`
`ni
`ni
`
`f
`M
`ni
`ni
`ni
`*
`ni
`ni
`f
`f
`ni
`
`ni
`f
`
`ni
`ni
`
`neg
`neg
`neg
`neg
`neg
`neg
`neg
`neg
`neg
`neg
`neg
`neg
`neg
`neg
`neg
`neg
`neg
`neg
`neg
`neg
`neg
`neg
`Exon 6 L182F
`neg
`neg
`neg
`neg
`neg
`neg
`neg
`neg
`neg
`neg
`
`ins, insulinoma; gluc, glucogonoma; vip, VIPoma; gast, gastrinoma; non F, non-functioning; mal, malignant
`† 10q rg-, partial 10q deletion by CGH (5)
`‡ C, cytoplasmic expression; N, nuclear expression; M, cytoplasmic membrane staining; neg, no expression; blanks, no paraffin blocks available
`§ f LOH; h ROH; ni, not informative; *, no PCR amplification; blanks, no germline tissue available.
`
`Roxane Labs., Inc.
`Exhibit 1020
`Page 003
`
`

`
`Perren et al
`1100
`AJP October 2000, Vol. 157, No. 4
`
`Figure 1. LOH analysis of EPT-nontumor DNA from cases 28 and 31 using microsatellite markers within and flanking PTEN. f, LOH; M, ROH; ni, not informative.
`
`Results
`
`LOH Analysis
`
`For 22 tumor samples, paired normal tissue was available
`and therefore, could be used for LOH analysis (Table 2).
`Sixteen of these EPTs were informative for at least one of
`four markers within and flanking PTEN. Among these 16,
`eight tumors exhibited loss of one allele at the 10q23
`region, and eight retained heterozygosity for all of the
`informative markers (Table 2 and Figure 1). Case 17
`showed retention of heterozygosity at AFMa086wg9 and
`LOH of the adjacent marker D10S2491 indicating partial
`loss of one PTEN allele. LOH at 10q23 seemed to be
`associated with malignant phenotype: whereas eight of
`10 (80%) informative malignant and none of six informa-
`tive benign EPTs showed evidence of loss of one allele,
`two of 10 (20%) malignant and six of six (100%) informa-
`
`tive-benign EPTs retained both 10q23 alleles (P , 0.05,
`Fischer’s exact test).
`Of the eight informative tumors with LOH, five have
`been shown by comparative genomic hybridization to
`have large 10q losses.
`
`Mutation Analysis
`
`Single-strand conformation polymorphism analysis re-
`vealed an additional band in exon 6 in one tumor (case
`23, Figure 2). Sequencing of exon 6 confirmed that the
`aberrant single-strand conformation polymorphism band
`reflected a sequence variant (546A.T) which was absent
`in the corresponding germline DNA. This 546 A.T tran-
`sition represents a somatic missense mutation resulting
`in an amino acid change at codon 182, L182F. This tumor
`
`Roxane Labs., Inc.
`Exhibit 1020
`Page 004
`
`

`
`1101
`PTEN Expression in Endocrine Pancreatic Tumors
`AJP October 2000, Vol. 157, No. 4
`
`immunostaining (Figure 2). This malignant, nonfunction-
`ing EPT had a somatic PTEN mutation (as described
`above) and loss of the remaining allele (Table 2, case
`23). In the remaining 23 EPTs, all showed strong PTEN
`expression, graded 1 to 11. Nineteen of these 23 (83%)
`tumors exhibited a predominantly cytoplasmic PTEN
`staining (Figure 3b and Table 2); the staining pattern of
`four of these 19 tumors was suggestive of PTEN localiza-
`tion at the cell membrane (Figure 3c). In addition, four of
`these 19 tumors were found to have cytoplasmic staining
`as well as strong nuclear PTEN staining (case 10, 15, 19,
`and 24; Figure 3d). We found no association between
`staining pattern and clinical behavior in this relatively
`small series.
`
`Discussion
`
`We have detected a novel somatic mutation in exon 6 of
`PTEN. The nucleotide change 546A.T results in an
`amino acid change at codon 182, L182F. As expected for
`a tumor suppressor gene, this mutation was accompa-
`nied by LOH of the wild-type allele. The structural two-hits
`in a malignant nonfunctioning EPT (case 23) resulted in
`loss of immunoreactivity to the monoclonal anti-PTEN
`antibody recognizing a C-terminal epitope. One would
`have expected that loss of one allele with a missense mu-
`tation in the remaining allele would result in a full-length
`protein, with subsequent decreased PTEN immunoreactiv-
`ity. Our observations of complete loss of immunoreactivity
`may be explained by decreased transcription or translation,
`increased degradation of the mutant protein, or confor-
`mational change in the C-terminal 100 amino acids in-
`duced by the missense mutation. Most likely, the mis-
`sense mutation results in decreased transcript stability. In
`sum, somatic intragenic PTEN mutations do occur in a
`small percentage, one of 33 (3%), of EPTs. The patient
`with this widely invasive nonfunctioning EPT underwent a
`Whipple procedure and remains disease-free 11 years
`after the surgery.
`Although somatic intragenic PTEN mutation is infre-
`quent in EPT, we have shown that half of all informative
`EPTs harbor deletions of the 10q23 region, specifically
`involving PTEN. On the one hand, LOH analysis con-
`firmed the large losses of 10q detected by comparative
`genomic hybridization analysis; on the other, we de-
`tected three additional malignant EPTs (cases 14, 18,
`and 28) with loss of the 10q23 region solely detected by
`PCR-based analysis of microsatellite markers. Interest-
`ingly, all of the samples that had LOH were malignant
`EPTs. This finding suggests that allelic loss of this region
`could be associated with malignant behavior. Apart from
`the single malignant EPT with two structural hits and no
`PTEN expression, all of the EPTs with LOH remained
`PTEN-immunopositive. These observations are in con-
`trast to those made in breast cancer, thyroid neoplasia,
`and endometrial cancer, where loss of one PTEN allele is
`strongly associated with decreased PTEN protein level or
`complete loss of PTEN expression (XP Zhou and C Eng,
`unpublished observations).17,20,25 In these tumors, there-
`fore, either one or both inactivational events can be epi-
`
`Figure 2. Case 23 EPT with somatic PTEN mutation, 546A.T and loss of
`wild-type allele, resulting in negative PTEN immunoreactivity. Top: Negative
`PTEN immunostaining; note internal positive control of strongly staining
`neo-vessels. Single-strand conformation polymorphism and sequencing of
`exon 6 reveals a somatic mutation (546A.T). Note that the chromatogram
`shows the mutation as an apparent heterozygote although by both immu-
`nohistochemical and LOH analysis, this tumor has loss one allele and is
`mutant in the remaining. This pseudo-heterozygote appearance is because of
`some admixture with DNA from contaminating normal cells.
`
`also showed LOH at 10q23, at least involving the 39 part
`of PTEN (Table 1).
`
`Immunohistochemistry
`
`Twenty-four paraffin-embedded tumor samples were
`available for immunohistochemical analysis. Fifteen of
`these had adjacent normal pancreatic tissue containing
`the islets of Langerhans, the normal counterpart of EPTs.
`All of the islets showed strong immunoreactivity (11) to
`the antibody 6H2.1. With the exception of one case which
`was fixed in Bouin’s solution (case 33), all normal islets
`exhibited homogenous predominant nuclear PTEN ex-
`pression (Figure 3a). As internal positive controls, we
`used predominantly nuclear staining of endothelial cells
`(especially within neo-vessels) and cytoplasmic staining
`of Schwann cells in peripheral nerves (graded 11 stain-
`ing). Exocrine pancreatic acini were PTEN immunostain-
`negative throughout and were used as internal negative
`controls.
`Among 24 EPTs that were subjected to immunohisto-
`chemistry, one (4%) was completely negative for PTEN
`
`Roxane Labs., Inc.
`Exhibit 1020
`Page 005
`
`

`
`Perren et al
`1102
`AJP October 2000, Vol. 157, No. 4
`
`Figure 3. Immunohistochemistry with anti-PTEN antibody. Original magnification, 3100. a: Normal islet (case 11). b: Cytoplasmic staining of tumor cells (case
`11). c: Cytoplasmic staining of tumor cells with membrane pattern (case 25). d: Nuclear and cytoplasmic staining (case 24).
`
`genetic. In EPTs, however, hemizygous loss of 10q and
`PTEN do not seem to be associated with decreased
`expression, an observation that may generate several
`hypotheses. It may be argued that PTEN is not the pri-
`mary target of 10q23 deletion and that other tumor sup-
`pressor genes in the region are the major targets. Fine
`deletion mapping of the 10q22–24 region in thyroid adeno-
`mas and carcinomas, for example, pointed to two distinct
`critical intervals of LOH in this region.26 An alternative pos-
`tulate might be supported by our immunohistochemical
`data. Although PTEN protein is localized mainly in the
`nucleus in nonneoplastic islets, it is localized predomi-
`nantly in the cytoplasm and cell membrane in 19 of 24
`(80%) EPTs. Differential subcellular localization of PTEN
`has been observed previously in a large series ranging
`from normal thyroid to anaplastic thyroid carcinoma.25 In
`the thyroid series, exclusion of nuclear staining was as-
`sociated with increasing malignant potential. The per-
`centage of tumors with decreased nuclear PTEN was
`lowest in breast cancer (Perren and Eng, unpublished
`observations), up to 50% in thyroid carcinomas25 and is
`highest in EPTs (19 of 24, 80%). In contrast to the thyroid
`tumors, however, this shifting of PTEN from nucleus to
`cytoplasm is not associated with increasing malignant
`behavior in EPTs but instead, is associated with the neo-
`plastic state in general.
`The observation of nuclear staining remains a puzzle
`but clearly has been observed by others.27,28 PTEN lacks
`a clear nuclear localization signal, and so, if it does traffic
`
`in and out of the nucleus, a shuttle must be involved. Our
`observations in vivo together with circumstantial prelimi-
`nary evidence demonstrating positive PTEN signal by
`Western blot analysis of nuclear fractions28 (L-P Weng
`and C Eng, unpublished observations) argue that this
`phenomenon is not an artifact. Although PTENs major
`substrate PtdIns(3,4,5)P3 as well as its antagonist phos-
`phatidyl
`inositol 3-kinase (PI3-K) normally interact with
`PTEN in the cytoplasm, more specifically, at the cytoplas-
`mic membrane, the phospholipids and PI3-K have been
`found in the nucleus as well although their role in the
`nucleus is still unknown.29 –31 Whether PTENs nuclear
`localization is significant for its interaction with the phos-
`pholipids or whether it best controls proper cell cycling in
`that location are still speculative. Therefore, based on our
`series of observations20,25 (this report; LP Weng and C
`Eng, unpublished observations), and independent data
`from other groups,27,28 we hypothesize that inappropriate
`qualitative or quantitative subcellular compartmentaliza-
`tion of PTEN could be a frequent initiating event in EPTs,
`which results in neoplasia, whereas physical loss of 10q
`leads to progression to malignancy.
`
`References
`
`1. Zhuang Z, Vortmeyer AO, Pack S, Huang S, Pham TA, Wang C, Park
`WS, Agarwal SK, Debelenko LV, Kester M, Guru SC, Manickam P,
`Olufemi SE, Yu F, Heppner C, Crabtree JS, Skarulis MC, Venzon DJ,
`
`Roxane Labs., Inc.
`Exhibit 1020
`Page 006
`
`

`
`Emmert-Buck MR, Spiegel AM, Chandrasekharappa SC, Collins FS,
`Burns AL, Marx SJ, Lubensky IA: Somatic mutations of the MEN1
`tumor suppressor gene in sporadic gastrinomas and insulinomas.
`Cancer Res 1997, 57:4682– 4686
`2. Gortz B, Roth J, Krahenmann A, de Krijger RR, Muletta-Feurer S,
`Rutimann K, Saremaslani P, Speel EJ, Heitz PU, Komminoth P: Mu-
`tations and allelic deletions of the MEN1 gene are associated with a
`subset of sporadic endocrine pancreatic and neuroendocrine tumors
`and not restricted to foregut neoplasms. Am J Pathol 1999, 154:429 –
`436
`3. Lohmann DR, Funk A, Niedermeyer HP, Haupel S, Hofler H: Identifi-
`cation of p53 gene mutations in gastrointestinal and pancreatic car-
`cinoids by nonradioisotopic SSCA. Virchows Arch B Cell Pathol 1993,
`64:293–296
`4. Hofler H, Ruhri C, Putz B, Wirnsberger G, Hauser H: Oncogene
`expression in endocrine pancreatic tumors. Virchows Arch B Cell
`Pathol 1988, 55:355–361
`5. Speel EJ, Richter J, Moch H, Egenter C, Saremaslani P, Rutimann K,
`Zhao J, Barghorn A, Roth J, Heitz PU, Komminoth P: Genetic differ-
`ences in endocrine pancreatic tumor subtypes detected by compar-
`ative genomic hybridization. Am J Pathol 1999, 155:1787–1794
`6. Nelen MR, Padberg GW, Peeters EAJ, Lin AY, van den Helm B, Frants
`RR, Coulon V, Goldstein AM, van Reen MMM, Easton DF, Eeles RA,
`Hodgson S, Mulvihill JJ, Murday VA, Tucker MA, Mariman ECM,
`Starink TM, Ponder BAJ, Ropers HH, Kremer H, Longy M, Eng C:
`Localization of the gene for Cowden disease to 10q22–23. Nat Genet
`1996, 13:114 –116
`7. 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
`8. 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 tumour
`suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in
`multiple advanced cancers. Nat Genet 1997, 15:356 –362
`9. 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
`10. Marsh DJ, Dahia PLM, Zheng Z, Liaw D, Parsons R, Gorlin RJ, Eng C:
`Germline mutations in PTEN are present in Bannayan-Zonana syn-
`drome. Nat Genet 1997, 16:333–334
`11. Marsh DJ, Coulon V, Lunetta KL, Rocca-Serra P, Dahia PLM, Zheng
`Z, Liaw D, Caron S, Duboue´ B, Lin AY, Richardson A-L, Bonnetblanc
`J-M, Bressieux J-M, Cabarrot-Moreau A, Chompret A, Demange L,
`Eeles RA, Yahanda AM, Fearon ER, Fricker J-P, Gorlin RJ, Hodgson
`SV, Huson S, Lacombe D, LePrat F, Odent S, Toulouse C, Olopade
`OI, Sobol H, Tishler S, Woods CG, Robinson BG, Weber HC, Parsons
`R, Peacocke M, Longy M, Eng C: Mutation spectrum and genotype-
`phenotype analyses in Cowden disease and Bannayan-Zonana syn-
`drome, two hamartoma syndromes with germline PTEN mutation.
`Hum Mol Genet 1998, 7:507–515
`12. Marsh DJ, Kum JB, Lunetta KL, Bennett MJ, Gorlin RJ, Ahmed SF,
`Bodurtha J, Crowe C, Curtis MA, Dazouki M, Dunn T, Feit H, Geraghty
`MT, Graham JM, Hodgson SV, Hunter A, Korf BR, Manchester D,
`Miesfeldt S, Murday VA, Nathanson KA, Parisi M, Pober B, Romano C,
`Tolmie JL, Trembath R, Winter RM, Zackai EH, Zori RT, Weng LP,
`Dahia PLM, Eng C: PTEN mutation spectrum and genotype-pheno-
`type correlations in Bannayan-Riley-Ruvalcaba syndrome suggest a
`single entity with Cowden syndrome. Hum Mol Genet 1999, 8:1461–
`1472
`13. Zhou XP, Marsh DJ, Hampel H, Mulliken JB, Gimm O, Eng C: Germ-
`line and germline mosaic mutations associated with a Proteus-like
`syndrome of hemihypertrophy, lower limb asymmetry, arterio-venous
`malformations and lipomatosis. Hum Mol Genet 2000, 9:765–768
`14. 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,
`
`1103
`PTEN Expression in Endocrine Pancreatic Tumors
`AJP October 2000, Vol. 157, No. 4
`
`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
`15. 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
`16. 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
`17. Mutter GL, Lin M-C, Fitzgerald JT, Kum JB, Baak JPA, Lees JA, Weng
`L-P, Eng C: Altered PTEN expression as a diagnostic marker for the
`earliest endometrial precancers. J Natl Cancer Inst 2000, 92:924 –931
`18. 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 tumours. Oncogene
`1998, 16:2259 –2264
`19. Bostro¨m J, Ludwig-Cobbers JMJ, Wolter M, Tabatabai G, Weber RG,
`Lichter P, Collins VP, Reifenberger G: Mutation of the PTEN (MMAC1)
`tumour suppressor gene in a subset of glioblastomas but not in
`meningiomas with loss of chromosome arm 10q. Cancer Res 1998,
`58:29 –33
`20. Perren A, Weng LP, Boag AH, Ziebold U, Thakore K, Dahia PLM,
`Komminoth P, Less JA, Mulligan LM, Mutter GL, Eng C: Immunohis-
`tochemical evidence of loss of PTEN expression in primary ductal
`adenocarcinomas of the breast. Am J Pathol 1999, 155:1253–1260
`21. Solcia E, Klo¨ppel G, Sobin LH, (Eds): Histological Typing of Endo-
`crine Tumours. Heidelberg, Springer, 2000
`22. Perren A, Roth J, Muletta-Feurer S, Saramaslani P, Speel EJ, Heitz
`PU, Komminoth P: Clonal analysis of sporadic pancreatic endocrine
`tumours. J Pathol 1998, 186:363–371
`23. Komminoth P, Kunz E, Hiort O, Scho¨der S, Matias-Guiu X, Chris-
`tiansen G, Roth J, Heitz PU: Detection of RET proto-oncogene point
`mutations in paraffin-embedded pheochromocytoma specimens by
`nonradioactive single-strand conformation polymorphism analysis
`and direct sequencing. Am J Pathol 1994, 145:922–929
`24. Weng L-P, Smith WM, Dahia PLM, Ziebold U, Gil E, Lees JA, Eng C:
`PTEN suppresses breast cancer cell growth by phosphatase func-
`tion-dependent G1 arrest followed by apoptosis. Cancer Res 1999,
`59:5808 –5814
`25. Gimm O, Perren A, Weng LP, Marsh DJ, Yeh JJ, Ziebold U, Gil E,
`Hinze R, Delbridge L, Lees JA, Robinson BG, Komminoth P, Dralle H,
`Eng C: Differential nuclear and cytoplasmic expression of PTEN in
`normal thyroid tissue, and benign and malignant epithelial thyroid
`tumors. Am J Pathol 2000, 156:1693–1700
`26. Yeh JJ, Marsh DJ, Zedenius J, Dwight T, Delbridge L, Robinson BG,
`Eng C: Fine structure deletion analysis of 10q22–24 demonstrates
`novel regions of loss and suggests that sporadic follicular thyroid
`adenomas and follicular thyroid carcinomas develop along distinct
`parallel neoplastic pathways. Gene Chromsom Cancer 1999, 26:322–
`328
`27. Tamura M, Gu J, Matsumoto K, Aota S-I, Parsons R, Yamada KM:
`Inhibition of cell migration, spreading and focal adhesions by tumor
`suppressor PTEN. Science 1998, 280:1614 –1617
`28. Lachyankar MB, Sultana N, Schonhoff CM, Mitra P, Poluha W, Lam-
`bert S, Quesenberry PJ, Litofsky NS, Recht LD, Nabi R, Miller SJ,
`Ohta S, Neel BG, Ross AH: A role for nuclear PTEN in neuronal
`differentiation. J Neurosci 2000, 20:1404 –1413
`29. D’Santos CS, Clarke JH, Divecha N: Phospholipid signalling in the
`nucleus. Biochim Biophys Acta 1998, 1436:201–232
`30. Metjian A, Roll RL, Ma AD, Abrams CS: Agonists cause nuclear
`translocation of phosphotidylinositol 3-kinase gamma. A Gbeta-gam-
`ma-dependent pathway that requires p110-gamma amino terminus.
`J Biol Chem 1999, 274:27943–27947
`31. Marchisio M, Bertagnolo V, Colamussi ML, Capitani S, Neri LM:
`Phosphotidylinositol 3-kinase in HL-60 nuclei is bound to the nuclear
`matrix and increases during granulocyte differentiation. Biochem Bio-
`phys Res Commun 1998, 253:346 –351
`
`Roxane Labs., Inc.
`Exhibit 1020
`Page 007

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket