telomerase elevation in pancreatic ductal carcinoma ...creatic ductal carcinomas were 100 and 80%,...

7
Vol. 3, 993-998, June 1997 Clinical Cancer Research 993 Telomerase Elevation in Pancreatic Ductal Carcinoma Compared to Nonmalignant Pathological States Nobuhiro Suehara, Kazuhiro Mizumoto,’ Tsuyoshi Muta, Yohei Tominaga, Hideo Shimura, Shigetaka Kitajima, Naotaka Hamasaki, Masazumi Tsuneyoshi, and Masao Tanaka Departments of Surgery I [N. S., K. M., Y. T., H. S., M. Ta.], Clinical Chemistry and Laboratory Medicine [T. M., S. K., N. H.], and Pathology 2 [M. Ts.], Kyushu University Faculty of Medicine, Fukuoka 812-82, Japan ABSTRACT Telomerase activity was measured in surgically re- sected tissues of 20 human pancreatic ductal carcinomas, 12 adenomas, 5 pancreatitis tissues, 14 normal pancreatic ducts, and 13 normal pancreatic tissues (primarily made up of acinar cells) using a PCR-based telomerase assay. Rela- tive telomerase activity was expressed as the equivalent telomerase intensity of the number of cells of a human pancreatic cancer cell line, MIA PaCa-2, per microgram of protein in the tissue samples. The median value (25th per- centile, 75th percentile) of relative telomerase activity in pancreatic carcinomas was 13.2 (3.58, 244), which was sig- nificantly higher relative to normal tissues, normal ducts, pancreatitis tissues, and adenomas (P < 0.0001). When the cutoff value of relative telomerase activity was set at 1.00 and 3.00, the positivity rates of telomerase activity in pan- creatic ductal carcinomas were 100 and 80%, respectively. Some of the adenoma samples displayed a weak telomerase ladder. However, when semiquantitatively analyzed, the rel- ative telomerase activity of all adenoma tissues was less than 1.00 equivalent cells per microgram protein of the tissues, which was equivalent to the values encountered in normal ducts. Thus, our results indicate that reactivation of telo- merase may occur at a late stage of pancreatic ductal car- cinogenesis. Therefore, telomerase may be a specific marker for distinguishing pancreatic cancer from pancreatitis and adenomas. INTRODUCTION Telomerase is an enzyme ribonucleoprotein responsible for cell immortality, and it synthesizes a six-nucleotide sequence designated as TTAGGG of telomeric DNA onto the chrorno- somal ends in germ-line cells and in immortal cells (1, 2). Received 10/29/96; revised 2/7/97; accepted 2/25/97. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. I To whom requests for reprints should be addressed. Phone: 81-92- 641-1 151, Ext. 5442; Fax: 8 1-92-642-5458. Although germ-line cells expressing telomerase activity main- tam telomeric repeats, somatic cells lose telomeres progres- sively as the cells undergo cell division (3, 4), which may lead to cellular senescence due to the repression of telomerase ac- tivity (5, 6). Cancer cells appear to attain immortality with the reactivation of telomerase (7). The conventional assay for telomerase activity requires a large quantity of cells or tissue (8), which in itself is a limitation in the investigation of the role of telomerase in carcinogenesis. Recently, a highly sensitive PCR-based assay was developed to detect telomerase activity in tissue extracts derived from a very small number of immortal cells (7). By this method, telomerase activity was detected in approximately 85-90% of various car- cinoma tissues such as lung cancer (9), colorectal cancer (10), hepatocellullar carcinoma (1 1 ), gastric cancer (1 2), and breast cancer (13). In addition, in cultured cells, 98 of 100 immortal cell populations and none of 22 mortal cell populations cx- pressed telomerase activity (7). Thus, it is apparent that telo- merase is repressed in normal somatic cells and tissues but is reactivated in most immortal cells and human cancers. The results of these studies suggest that telomerase may ultimately be required to maintain cell proliferation indefinitely. Therefore, detection of telomerase activity in clinically available specimens may be of value in the diagnosis of malignant tumors. Pancreatic cancer is one of the most aggressive malignant tumors and generally has an extremely poor prognosis (14, 15). Diagnosis of pancreatic cancer in the early stage may contribute to drastic improvement of the prognosis. Human pancreatic ductal carcinomas display variable but consistent genetic changes, including mutations of K-ras (16, 17), p53 (18), APC (19), andpl6 (20). Ifsuch genetic changes can be detected using a small number of cells, they can serve as a clinical marker to the diagnosis of cancer. K-ras mutations have been detected in the pancreatic juice obtained by endoscopic retrograde pancre- atography in 55 to 100% of patients with pancreatic cancer (21-24). However, K-ras mutations are present in noncancerous tissues such as ductal hyperplasia (25) and adenoma (26, 27) and, therefore, are of limited value as a clue to the diagnosis of pancreatic cancer. We reported previously that telomerase is highly activated in human pancreatic ductal carcinoma when investigated by a semiquantitative modified PCR-based assay (28). Apparently, telomerase is a new prevalent marker for human pancreatic ductal carcinoma. However, it is not known at which step of pancreatic carcinogenesis telornerase is reactivated. If telomer- ase is activated exclusively in cancer and repressed in preneo- plastic lesions of pancreatitis or adenoma, telomerase could well be a specific marker of pancreatic cancer. In the present study, relative values of telomerase activity were measured in surgi- cally resected tissues of pancreatic carcinoma, adenoma, pan- creatitis, normal pancreatic ducts, and normal pancreatic tissues to evaluate the putative role of telomerase in the diagnosis of pancreatic cancer. Research. on June 6, 2021. © 1997 American Association for Cancer clincancerres.aacrjournals.org Downloaded from

Upload: others

Post on 27-Jan-2021

1 views

Category:

Documents


0 download

TRANSCRIPT

  • Vol. 3, 993-998, June 1997 Clinical Cancer Research 993

    Telomerase Elevation in Pancreatic Ductal Carcinoma Compared to

    Nonmalignant Pathological States

    Nobuhiro Suehara, Kazuhiro Mizumoto,’

    Tsuyoshi Muta, Yohei Tominaga, Hideo Shimura,Shigetaka Kitajima, Naotaka Hamasaki,Masazumi Tsuneyoshi, and Masao TanakaDepartments of Surgery I [N. S., K. M., Y. T., H. S., M. Ta.], Clinical

    Chemistry and Laboratory Medicine [T. M., S. K., N. H.], and

    Pathology 2 [M. Ts.], Kyushu University Faculty of Medicine,Fukuoka 812-82, Japan

    ABSTRACT

    Telomerase activity was measured in surgically re-

    sected tissues of 20 human pancreatic ductal carcinomas, 12

    adenomas, 5 pancreatitis tissues, 14 normal pancreatic

    ducts, and 13 normal pancreatic tissues (primarily made up

    of acinar cells) using a PCR-based telomerase assay. Rela-

    tive telomerase activity was expressed as the equivalent

    telomerase intensity of the number of cells of a human

    pancreatic cancer cell line, MIA PaCa-2, per microgram of

    protein in the tissue samples. The median value (25th per-

    centile, 75th percentile) of relative telomerase activity inpancreatic carcinomas was 13.2 (3.58, 244), which was sig-

    nificantly higher relative to normal tissues, normal ducts,

    pancreatitis tissues, and adenomas (P < 0.0001). When thecutoff value of relative telomerase activity was set at 1.00

    and 3.00, the positivity rates of telomerase activity in pan-

    creatic ductal carcinomas were 100 and 80%, respectively.

    Some of the adenoma samples displayed a weak telomerase

    ladder. However, when semiquantitatively analyzed, the rel-

    ative telomerase activity of all adenoma tissues was less than1.00 equivalent cells per microgram protein of the tissues,

    which was equivalent to the values encountered in normal

    ducts. Thus, our results indicate that reactivation of telo-

    merase may occur at a late stage of pancreatic ductal car-

    cinogenesis. Therefore, telomerase may be a specific marker

    for distinguishing pancreatic cancer from pancreatitis and

    adenomas.

    INTRODUCTION

    Telomerase is an enzyme ribonucleoprotein responsible for

    cell immortality, and it synthesizes a six-nucleotide sequence

    designated as TTAGGG of telomeric DNA onto the chrorno-

    somal ends in germ-line cells and in immortal cells (1, 2).

    Received 10/29/96; revised 2/7/97; accepted 2/25/97.The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby marked

    advertisement in accordance with 1 8 U.S.C. Section 1734 solely to

    indicate this fact.I To whom requests for reprints should be addressed. Phone: 81-92-641-1 151, Ext. 5442; Fax: 8 1-92-642-5458.

    Although germ-line cells expressing telomerase activity main-

    tam telomeric repeats, somatic cells lose telomeres progres-

    sively as the cells undergo cell division (3, 4), which may lead

    to cellular senescence due to the repression of telomerase ac-

    tivity (5, 6). Cancer cells appear to attain immortality with the

    reactivation of telomerase (7).

    The conventional assay for telomerase activity requires a

    large quantity of cells or tissue (8), which in itself is a limitation

    in the investigation of the role of telomerase in carcinogenesis.

    Recently, a highly sensitive PCR-based assay was developed to

    detect telomerase activity in tissue extracts derived from a very

    small number of immortal cells (7). By this method, telomerase

    activity was detected in approximately 85-90% of various car-

    cinoma tissues such as lung cancer (9), colorectal cancer (10),

    hepatocellullar carcinoma (1 1 ), gastric cancer (1 2), and breast

    cancer (13). In addition, in cultured cells, 98 of 100 immortal

    cell populations and none of 22 mortal cell populations cx-

    pressed telomerase activity (7). Thus, it is apparent that telo-

    merase is repressed in normal somatic cells and tissues but is

    reactivated in most immortal cells and human cancers. The

    results of these studies suggest that telomerase may ultimately

    be required to maintain cell proliferation indefinitely. Therefore,

    detection of telomerase activity in clinically available specimens

    may be of value in the diagnosis of malignant tumors.

    Pancreatic cancer is one of the most aggressive malignant

    tumors and generally has an extremely poor prognosis (14, 15).

    Diagnosis of pancreatic cancer in the early stage may contribute

    to drastic improvement of the prognosis. Human pancreatic

    ductal carcinomas display variable but consistent genetic

    changes, including mutations of K-ras (16, 17), p53 (18), APC

    (19), andpl6 (20). Ifsuch genetic changes can be detected using

    a small number of cells, they can serve as a clinical marker to

    the diagnosis of cancer. K-ras mutations have been detected in

    the pancreatic juice obtained by endoscopic retrograde pancre-

    atography in 55 to 100% of patients with pancreatic cancer

    (21-24). However, K-ras mutations are present in noncancerous

    tissues such as ductal hyperplasia (25) and adenoma (26, 27)

    and, therefore, are of limited value as a clue to the diagnosis of

    pancreatic cancer.

    We reported previously that telomerase is highly activated

    in human pancreatic ductal carcinoma when investigated by a

    semiquantitative modified PCR-based assay (28). Apparently,

    telomerase is a new prevalent marker for human pancreatic

    ductal carcinoma. However, it is not known at which step of

    pancreatic carcinogenesis telornerase is reactivated. If telomer-

    ase is activated exclusively in cancer and repressed in preneo-

    plastic lesions of pancreatitis or adenoma, telomerase could well

    be a specific marker of pancreatic cancer. In the present study,

    relative values of telomerase activity were measured in surgi-

    cally resected tissues of pancreatic carcinoma, adenoma, pan-

    creatitis, normal pancreatic ducts, and normal pancreatic tissues

    to evaluate the putative role of telomerase in the diagnosis of

    pancreatic cancer.

    Research. on June 6, 2021. © 1997 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • 994 Telomerase Elevation in Pancreatic Ductal Carcinoma

    Table I Relative values of telo merase a ctivity i n patients w ith pancreatic aden omas and carcinomas

    Case Histology Age Sex” Staging” Differentiation Location Rd ative tclomerase activity’-Adenomas

    25 Serous cystadenoma 82 F Pt 0.44

    38 Serous cystadenoma 72 F Pb 0.25

    100 Serous cystadenoma 32 M Pt 0.21I 10 Serous cystadenoma 53 F Pb 0.58

    62 Mucinous cystadenoma 54 M Pb-t 0.56

    1 15 Mucinous cystadenoma 75 F Pb-t 0.447 Intraductal papillary adenoma 67 M Ph 0.71

    94 Intraductal papillary adenoma 81 F Ph 0.30

    102 Intraductal papillary adenoma 46 F Pb-t 0.76

    104 Intraductal papillary adenoma 68 F Ph 0.76108 Intraductal papillary adcnoma 53 F Ph 0.30

    I 30 lntraductal papillary adenoma 78 M Pb 0.34

    Carcinomas

    5 Intraductal papillary adenocarcinoma 72 F TI�,N�M() Ph 2750

    35 lntraductal papillary adenocarcinoma 61 F T15NJM() Ph 196

    6 Tubular adenocarcinoma 77 M T,N1M1 Well Ph 518,000

    59 Tubular adenocarcinoma 72 M T2N,M0 Well Pb 57.5

    65 Tubular adenocarcinoma 75 F T3N1M1 Well Ph 5.27

    66 Tubular adenocarcinoma 77 F T3N0M#{216} Well Ph 26067 Tubular adenocarcinoma 59 M T2N1M#{216} Well Ph 3.50

    81 Tubular adenocarcinoma 73 M T,N0M0 Well Ph 295

    I I I Tubular adenocarcinoma 68 M T3N)M() Well Ph 4.70

    I Tubular adenocarcinoma 50 F T3N1M, Moderate Pb-t 8.93

    69 Tubular adenocarcinoma 40 M T3N,M0 Moderate Pb-t 2.3576 Tubular adenocarcinoma 69 M T3N0M0 Moderate Ph 1.98

    113 Tubular adenocarcinoma 66 F T,N0M0 Moderate Ph 5.19

    121 Tubular adenocarcinoma 52 M T1N1MJ Moderate Ph 3.83

    131 Tubular adenocarcinoma 53 F T,N0M0 Moderate Ph 2.46

    I 2 Tubular adenocarcinoma 55 M T1N0M0 Poor Ph 34.7

    83 Tubular adenocarcinoma 59 M T�N)M() Poor Ph 2.17

    70 Tubular adenocarcinoma 74 F T,N0M0 Poor Pb-t 970

    93 Mucinous cystadenocarcinoma 66 F T3N1M0 Ph 4.98

    41 Anaplastic ductal carcinoma 74 F T3N1M0 Pb-t 17.4

    ,1 M. male; F, female; Ph, head of pancreas: Pb, body of pancreas; Pt, tail of pancreas; Pb-t, body and tail of pancreas.

    “ Tumor stage is assessed according to UICC-TNM staging (29).‘ Relative values of telomerase activity are expressed as the equivalent telomerase intensity of the number of cells of MIA PaCa-2 per microgram

    of protein in the tissue samples.

    MATERIALS AND METHODSCell Lines and Tissue Samples. The human pancreatic

    cancer cell line MIA PaCa-2, generously provided by Japanese

    Cancer Resources Bank (Tokyo, Japan), was cultured in DMEM

    (Nissui Pharmaceutical Co., Ltd., Tokyo, Japan) supplemented

    with penicillin (1000 units/mI), streptomycin sulfate (100 ji.g/

    ml), and 10% heat-inactivated fetal bovine serum.

    Tissue samples were obtained at the time of surgery either

    at Kyushu University Hospital (Fukuoka, Japan) or at affiliated

    hospitals. Specimens were from 20 primary pancreatic ductal

    carcinomas, including 2 intraductal papillary adenocarcinomas,

    16 tubular adenocarcinomas (7 well, 6 moderately, and 3 poorly

    differentiated adenocarcinomas), a mucinous cystadenocarci-

    noma, and an anaplastic adenocarcinoma; 12 pancreatic adeno-

    mas, including 4 serous cystadenomas, 2 mucinous cystadeno-

    mas, and 6 intraductal papillary adenomas; 5 pancreatitis

    tissues: 14 normal main pancreatic ducts; and 13 normal pan-

    creatic tissues (primarily made up of acinar cells). They were

    removed as soon as possible after resection and were stored at

    -80#{176}C until use. Normal pancreatic ducts were taken from the

    main pancreatic duct in patients with pancreatitis and from the

    main ducts apart from the tumor in patients with neoplasms.

    Normal pancreatic tissue samples were collected from a periph-

    eral soft part of the pancreas in the same manner. The tissue

    samples in pancreatitis were primarily from a hard portion of the

    pancreatic mass including the stenotic ducts. The samples of

    serous cystadenomas or mucinous cystadenomas were obtained

    from the cyst wall. In patients with intraductal papillary adeno-

    mas, the affected ducts were opened, and a sample was taken

    from a macroscopically distinct tumor. The tissues of pancreatic

    carcinoma were excised from a hard portion of the tumors. All

    tissues adjacent to the specimens were histologically examined,

    and the diagnosis was confirmed. In cases of unresectable pan-

    creatic carcinomas such as were found in patients 12, 69, 93, and

    121 (Table 1), needle biopsies were performed at laparotomy,

    and a portion of the same sample was diagnosed histologically.

    The definitions of stage grouping, histological classification,

    and lymphatic metastasis of pancreatic cancers were made ac-

    cording to the UICC-TNM2 classification (29).

    2 The abbreviation used is: UICC-TNM, International Union against

    Cancer, Tumor-Node-Metastasis.

    Research. on June 6, 2021. © 1997 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • Clinical Cancer Research 995

    Telomerase Assay. Telomerase activity was measured

    as described previously (28). Briefly, the cell pellets from

    MIA PaCa-2 were suspended with CHAPS lysis buffer [10 m�i

    Tris-HC1 (pH 7.5), 1 mM MgC1.,, 1 m�i EGTA, 0.1 mrvi

    4-(2-aminoethyl)-benzenesulfonylfluoride hydrochloride, 5 ms�

    �3-mercaptoethanol, 0.5% CHAPS, ]0% glycerol, and protease

    inhibitors. The frozen pancreatic tissues were washed once in

    ice-cold PBS and also homogenized with CHAPS lysis buffer in

    microtubes with matching pestles. The supernatants (CHAPS

    cell extract) were stored at -80#{176}C until use. Protein concentra-

    tion of the tissue extract was measured by Bradford assay (30),

    and 6 or 0.6 �ig extract was used for each telomerase assay.

    Telomerase activity was assayed by the modification of the

    PCR-based telomerase assay as described by Kim et a!. (7) and

    Tahara et a!. (3 1). The CHAPS cell extracts were incubated with

    reaction buffer containing 50 �1M deoxynucleotide triphos-

    phates, 0.3 p.Ci of [a-32P]dCTP. 2 units of Taq DNA polymer-

    ase (Promega Corp., Madison, WI), and 0.1 p.g of TS primer

    (5’-AATCCGTCGAGCAGAGTF-3’). During this step, 0.1 �g

    of CX primer (5’-CCCTTACCCTTACCCTTACCCTAA-3’)

    was added, and the reaction mixture was subjected to 3 1 PCR

    cycles. Telomerase activity was detected as a 6-base ladder

    signal that disappeared with RNase (Boehringer-Manheim

    Corp., Manheim, Germany) pretreatment, indicating that the

    reaction was specific for telomerase activity (see Fig. 1). MIA

    PaCa-2 extracts equivalent to 1, 10, 102, and ]0� cells were

    always measured as a standard per assay, and relative values of

    telomerase activity were expressed as the equivalent telomerase

    intensity of the number of MIA PaCa-2 cells per microgram

    protein in each sample as described previously (28). Signal

    intensity of the 6-base ladder was measured by NIH image,

    version 1 .59 (NTIS, Springfield, VA). A 36-bp internal standard

    (Oncor, Inc., Gaithersburg. MD) was used as an internal control.

    Logarithmic values of the relative telomerase activity were

    statistically analyzed by one-way ANOVA and Fisher’s test

    because they showed a normal distribution.

    RESULTSEfficiency of the PCR reaction was confirmed with the

    detection of a 36-bp internal standard (Fig. I). Telomerase

    activity in the human pancreatic cancer cell line MIA PaCa-2

    was detected as a six-nucleotide repeat ladder, and the signal

    intensity was reduced according to the decrease in the number of

    cancer cells by dilution. Regression analysis was performed

    between logarithmic values of the cell numbers and telomerase

    intensity, and the correlation coefficient always exceeded 0.9.

    A definite telomerase ladder was not observed in all normal

    pancreatic tissue and duct specimens (Fig. la), and the median

    values (25th percentile, 75th percentile) of relative telomerase

    activity expressed by the equivalent telomerase intensity of the

    number of MIA PaCa-2 cells were 0.25 (0.20, 0.45) and 0.31

    (0.14, 0.50), respectively. In all pancreatitis sample extracts, the

    telomerase ladder was also undetectable, and the median value

    (25th percentile, 75th percentile) of relative telomerase activity

    was 0. 17 (0.09, 0.46). All relative values of telomerase activities

    of normal pancreas, normal ducts, and pancreatitis were under

    I .00 equivalent cells per microgram of tissue protein.

    A telomerase ladder was indistinct in most pancreatic ad-

    enomas. However, some of the adenomas presented weak te-

    lornerase ladder signals, which would have been regarded as

    positive had they been assessed by simple qualitative telomerase

    analysis. Semiquantitatively analyzed, relative telomerase activ-

    ities of each adenoma are listed in Table I . All values of

    adenomas were under 1 .00 equivalent cells per microgram of

    tissue protein. The median value (25th percentile, 75th percen-

    tile) in adenomas was 0.44 (0.30, 0.68), which was slightly

    higher than that encountered in normal tissues and ducts but not

    significantly different from these two groups. Mucinous cysta-

    denomas and intraductal adenomas are generally believed to

    have a greater malignant potential than serous cystadenomas.

    However, telomerase activity was not significantly different

    between these two groups (Table 2).

    A telomerase ladder was clearly detected in almost all

    pancreatic ductal carcinomas (Fig. la). All of the values of

    relative telomerase activity in carcinomas were over 1 .00 equiv-

    alent cells per microgram of tissue protein and varied from I .98

    to 518,000. The median value (25th percentile, 75th percentile)

    of relative telomerase activities of ductal carcinomas was 13.2

    (3.58, 244), which was significantly higher than that encoun-

    tered in normal tissues, normal ducts, pancreatitis, and adeno-

    mas (P < 0.0001). Relative values of telomerase activity of all

    samples are summarized in Fig. 2 by means of a box-and-

    whisker plot analysis, which clearly demonstrates exclusive

    activation of telomerase in pancreatic ductal carcinomas. With

    regard to the histological differentiation in pancreatic ductal

    carcinomas, there were no significant differences among intra-

    ductal papillary, well, moderately, and poorly differentiated

    adenocarcinomas. However, relative telomerase activity in the

    group of intraductal papillary and well-differentiated adenocar-

    cinomas was significant when compared to the group of mod-

    erately to poorly differentiated adenocarcinomas (Table 2; P <

    0.05). In terms of staging of the disease, the median value of

    telomerase activity in pancreatic ductal carcinomas in the T1-T2

    group tended to be higher than that in the T3-T4 group, although

    the probability value of the difference was 0.0544. There was no

    significant difference between the locations of the carcinomas,

    i.e., pancreatic head versus pancreatic body and tail.

    Telomerase activity was measured mixing carcinoma tis-

    sues with normal ducts to evaluate the effect of normal cells on

    the telomerase ladder. The intensity of the telomerase ladder of

    carcinoma was reduced to some extent, consistent with the

    inclusion of normal ducts (Fig. lb).

    DISCUSSION

    The present study has revealed that telomerase was defi-

    nitely activated in almost all pancreatic ductal carcinomas. In

    addition to the repression of telomerase in normal pancreatic

    tissues, normal ducts that are precursor cells of ductal carcino-

    mas did not show any telomerase ladder. When the cutoff value

    of relative telomerasc activity was set at I .00 and 3.00, the

    positivity rate of pancreatic ductal carcinomas was 100 and

    80%, respectively. Furthermore, relative values of telomerase

    activity in pancreatitis or adenomas are comparable to those of

    normal ducts. These results suggest that the possibility that

    telomerase could be a new prevalent marker for pancreatic

    ductal carcinoma.

    Research. on June 6, 2021. © 1997 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • a

    LLg tissue

    extractRNase

    b

    I � I 1 II II

    6 6 6 6 6 6 6 6660.6 0.6 0.6

    --+--+--+- +- +--

    6 6

    if

    12

    996 Telomerase Elevation in Pancreatic Ductal Carcinoma

    Internal

    (36bp) � � �..control’-#{248}

    1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20Fig. I �i. telomerase activity signals in pancreatic ductal carcinoma. adenoma, pancreatitis tissue. normal duct. and normal tissue samples. From each

    tissue sample. an aliquot of the extract containing 6 or 0.6 p.g of protein with ( + ) or without ( - ) RNase pretreatment was used in each assay. Aliquotsof the extracts containing I . 10. 102. and l0� cells of the human pancreatic cancer cell line MIA PaCa-2 having telomerase activity were used as a

    standard. Lysis buffer was used as a negative control. Telomerase activity was detected after electrophoresis of the enzyme reaction products andautoradiography as a six-nucleotide repeat ladder. A 36-bp internal standard was used as an internal control. b: Lane 1. mixing extracts from pancreatic

    carcinoma patient 70 (3 �i.g of protein) with 3 p.g of albumin. Lane 2. mixing extracts from pancreatic carcinoma patient 70 (3 p.g of protein) withextracts from normal pancreatic duct patient 70 (3 p.g of protein).

    Relative values of telomerase activity in pancreatic ductal

    carcinomas varied from 1.98 to 518,000. The wide variation of

    the results does not mean instability of the assay per se. Radio-

    activity of �2P is unstable and. as such, difficult to maintain at

    the same level in each analysis. However, determination of

    telomerase activity in a human pancreatic cancer cell line can

    standardize the results of each assay. Nearly the same activity

    value was obtained when one sample was analyzed several

    times. Pancreatic cancer tissues include variable numbers of

    cancer cells along with abundant stromas with fibroblasts, which

    may vary from sample to sample. The study involving mixing of

    carcinoma tissues and normal ducts revealed that normal cells

    could reduce the intensity of the telomerase ladder of carcino-

    mas cells (Fig. lh). In addition to the difference in the number

    of carcinoma cells, the difference in the proportion of malignant

    cells to normal cells in the tissue could be one of the possible

    explanations for the wide variation in the telomerase activity in

    pancreatic ductal carcinomas.

    Adenoma-carcinoma sequence can be applied in pancreatic

    carcinogenesis as indicated in hamster models of pancreatic

    carcinogenesis (32). Clinically, mucinous cystadenoma and in-

    traductal papillary adenoma are presumed to have malignant

    potential. Some adenomas showed a clear 6-base ladder, but the

    intensity of the signal was much weaker when compared to any

    of pancreatic ductal carcinomas. Recently, it was reported that

    weak telomerase activity was detected in precancerous lesions,

    for example. in 23% of gastric intestinal metaplasias and 50% of

    gastric adenomas (3 1 ). In colorectal lesions, telomerase activity

    was detected in 0% (10) or 100% (31) in adenomas by qualita-

    tive analysis. There is still a possibility that weak telomerase

    activity may also be detected in pancreatic adenomas as more

    patients with adenoma are evaluated. However, the notable

    point is that there is a clear difference in telomerase activity

    expressed between adenomas and carcinomas.

    Of practical clinical importance is the distinction between

    chronic pancreatitis and pancreatic cancer. It has been pointed

    out that mass-forming pancreatitis masquerades as pancreatic

    cancer (33). A preoperative diagnostic tool is sorely needed to

    Research. on June 6, 2021. © 1997 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • 10000

    1000

    100

    10

    0.1

    0.01

    Normal Tissues Normal Ducts Pancreatitis Tissues Adenomas Carcinomas

    Clinical Cancer Research 997

    Table 2 Relative telomerase activity in patients with normal pancreatic ducts . adenomas, and carcinomas

    Relative tclomcrasc activity” Median

    Lesion No. of samples (25th percentile. 75th percentile)

    Normal ducts 14 0.31 (0.14, 0.50)

    Adenomas 12 0.44 (0.30, 0.68)Scrous cystadenomas 4 0.35 (0.22, 0.55)Mucinous cystadenomas and intraductal papillary adenomas 8 0.50 (0.31. 0.75)

    Carcinomas 20 13.2 (3.58. 244)”Differentiation

    intraductal papillary and well-differentiated adenocarcinomas 9 196 ( 13.5. 1520)

    moderately and poorly differentiated adenocarcinomas 9 3.83 (2.26, 21.8)’

    T classification”

    T,-1’, 9 196(4.35, 1860)

    T3-T4 II 5.27 (2.35. 21.7)

    LocationHead 15 5.27 (3.50, 260)Body-tail S 17.5 (5.64. 514)

    a Relative values of telomerase activity are expressed as the equivalent telomerase intensity of the number of cells of MIA PaCa-2 per microgram

    of protein in the tissue samples.“ Significantly different from groups of normal ducts and adenomas (P < 0.0001).‘ Significantly different from a group of intraductal papillary and well-differentiated adenocarcinomas (P < 0.05).“ Tumor stage is assessed according to UICC-TNM staging (29).

    Fig. 2 Relative values of telomerase activity.Relative values of telomerase activity arc cx-pressed as the equivalent telomerase intensity of

    the number of cells of MIA PaCa-2 per micro-

    gram protein of samples. The results are cx-

    pressed by means of a box-and-whisker plot anal-

    ysis. The botto�n and top edges of the box arelocated at the sample 25th and 75th percentiles.respectively. The center horizontal line is drawn

    at the sample median. The center vertical lines

    from the box extend to a distance of the 10th or90th percentiles. The relative values of telomeraseactivity in carcinoma samples arc significantlyhigher than those in other tissue samples (P <0.0001).

    aE

    C

    C

    >

    a:

    100000

    discriminate pancreatic cancer from chronic pancreatitis. The

    present study demonstrated that telomerase was exclusively

    activated in the tissues of pancreatic carcinomas while being

    completely repressed in chronic pancreatitis. The future study of

    telomerase activity on cell samples in pancreatic juice obtained

    preoperatively by endoscopic retrograde pancreatography is

    warranted.

    In regard to the staging of pancreatic cancer, relative te-

    lomerase activity tended to be higher in the T1-T, group than in

    the T3-T4 group. In the T3-T4 group, the unique composition of

    the far-advanced pancreatic carcinoma, consisting of a large

    amount of fibrous stroma, necrotic tissue, and mucin in some

    cases, along with a relatively small number of cancer cells, may

    account for the lower telomerase activity. Telomerase activity

    was significantly higher in the group of intraductal papillary and

    well-differentiated adenocarcinomas when compared to the

    group of moderately to poorly differentiated adenocarcinomas.

    Considering that telomerase activity correlates to cellular im-

    mortality, a patient with pancreatic cancer presenting with high

    telomerase activity will have a poor prognosis. It has been

    reported that the survival rate of gastric cancer tumors with

    telomerase activity is significantly shorter compared to gastric

    cancer tumors without telomerase activity (12). Similarly, neu-

    roblastoma patients presenting with high telomerase activity

    have an unfavorable prognosis compared to cases of neuroblas-

    toma with low telomerase activity (34). To our knowledge. there

    have been no reports of a clear relationship between histological

    differentiation and outcome of pancreatic cancer. Furthermore,

    no previous study has demonstrated an unfavorable prognosis

    for intraductal papillary and well-differentiated adenocarcino-

    mas. The group of intraductal papillary and well-differentiated

    adenocarcinomas consisted of a relatively dense cellular corn-

    ponent of carcinoma. On the other hand, the group of moder-

    ately to poorly differentiated adenocarcinomas consisted of rel-

    atively abundant stromas. The difference in telomerase activity

    among cancer cell populations may indeed be affected by such

    proportional differences between malignant cells and normal

    cells.

    Research. on June 6, 2021. © 1997 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • 998 Telomerase Elevation in Pancreatic Ductal Carcinoma

    ACKNOWLEDGMENTSWe are grateful to N. Kinukawa, Department of Medical Informat-

    ics, Kyushu University, for advice on the statistical analysis. We thank

    Professor Sheshadri Narayanan, New York Medical College. for assist-

    ance in the preparation of the manuscript.

    REFERENCES

    I . Greider, C. W., and Blackburn, E. H. Identification of a specific

    telomere terminal transferase activity in Tetrahymena extracts. Cell, 43:405-413, 1985.

    2. Morin, G. B. The human telomere terminal transfcrasc enzyme is a

    ribonucleoprotcin that synthesizes TTAGGG repeats. Cell, 59: 521-

    529, 1989.

    3. Harley, C. B. Tclomcrc loss: mitotic clock or genetic time bomb?

    Mutat. Res., 256: 271-282, 1991.

    4. Shay, J. W., Werbin, H., and Wright, W. E. Tclomcrc shortening may

    contribute to aging and cancer: a perspective. Mol. Cell. Differ., 2:1-21, 1994.

    5. Harley. C. B., Futcher, A. B.. and Greidcr, C. W. Telomcrcs shorten

    during aging of human fibroblasts. Nature (Lond.), 345: 458-460,

    1990.

    6. Allsopp. R. C., Vaziri, H.. Patterson, C., Goldstein, S., Younglai,E. V., Futchcr. A. B., Grcidcr, C. W., and Harley. C. B. Telomere lengthpredicts replicative capacity of human fibroblasts. Proc. Natl. Acad. Sci.

    USA,89: 10114-10118, 1992.

    7. Kim, N. W., Piatyszek, M. A., Prowse, K. R., Harley, C. B., West,

    M. D., Ho, P. L. C., Covicllo, G. M., Wright. W. E., Weinrich, S. L., and

    Shay. J. W. Specific association of human telomerase activity withimmortal cells and cancer. Science (Washington DC), 266: 201 1-2015,1994.

    8. Counter, C. M., Hirte, H. W., Bacchctti, S., and Harley, C. B.

    Telomerase activity in human ovarian carcinoma. Proc. Natl. Acad. Sci.USA, 91: 2900-2904, 1994.

    9. Hiyama, K., Hiyama, E., Ishioka, S., Yamakido, M., Inai, K., Gazdar,A. F., Piatyszck. M. A.. and Shay. J. W. Tclomcrasc activity in small-

    cell and non-small-cell lung cancers. J. NatI. Cancer Inst., 87: 895-903,

    1995.

    10. Chadencau, C., Hay, K.. Hirte, H. W., Gallinger, S., and Bacchctti,S. Tclomcrase activity associated with acquisition of malignancy inhuman colorectal cancer. Cancer Rcs., 55: 2533-2536, 1995.

    I I . Tahara, H., Nakanishi, 1., Kitamoto, M., Nakashio, R., Shay, J. W.,Tahara, E., Kajiyama, G., and Ide, 1. Telomerase activity in human livertissues: comparison between chronic liver disease and hepatocellular

    carcinomas. Cancer Res., 55: 2734-2736, 1995.

    12. Hiyama, E., Yokoyama, T., Tatsumoto, N., Hiyama, K., Imamura,

    Y.. Murakami, Y., Kodama, T., Piatyszek, M. A., Shay, J. W., and

    Matsuura, Y. Telomcrasc activity in gastric cancer. Cancer Res., 55:

    3258-3262, 1995.

    13. Hiyama. E., Gollahon, L., Kataoka, T., Kuroi, K., Yokoyama, 1..Gazdar, A. F., Hiyama, K., Piatyszck, M. A.. and Shay, J. W. Telo-merase activity in human breast tumors. J. NatI. Cancer Inst., 88:

    116-122, 1996.

    14. Warshaw, A. L., and Fernandez-del Castillo, C. Pancreatic carci-noma. N. EngI. J. Med., 326: 455-465, 1992.

    IS. Niederhuber, J. E., Brennan, M. F., and Mcnck, H. R. The national

    cancer data base report on pancreatic cancer. Cancer (Phila.), 76:

    1671-1677, 1995.

    16. Almoguera, C., Shibata, D., Forrester, K., Martin, J., Arnheim, N.,and Perucho, M. Most human carcinomas of the exocrine pancreascontain mutant c-K-ras genes. Cell, 53: 549-554, 1988.

    17. Hruban, R. H., van Mansfcld, A. D. M., Offerhaus, G. J. A., van

    Wecring. D. H. J., Allison, D. C., Goodman, S. 1., Kensler, T. W.. Bose,K. K., Cameron, J. L., and Bos, J. L. K-ras oncogene activation in

    adenocarcinoma of the human pancreas. Am. J. Pathol., 143: 545-554,

    1993.

    18. Redston, M. S., Caldas, C., Seymour, A. B., Hruban, R. H.,

    da Costa, L., Yco, C. J., and Kern, S. E. p53 mutations in pancreaticcarcinoma and evidence of common involvement of homocopolymcr

    tracts in DNA microdclctions. Cancer Res., 54: 3025-3033, 1994.

    19. Hon. A., Nakatsuru, S., Miyoshi, Y., Ichii, S., Nagase, H., Ando,

    H., Yanagisawa, A., Tsuchiya, E., Kato, Y., and Nakamura, Y. Frequent

    somatic mutations of the APC gene in human pancreatic cancer. CancerRes., 52: 6696-6698, 1992.

    20. Caldas, C.. Hahn, S. A., da Costa, L. T., Rcdston, M. S., Schutte,

    M., Seymour, A. B., Weinstein, C. L., Hruban, R. H., Yco, C. J., and

    Kern, S. E. Frequent somatic mutations and homozygous deletions ofthe p16 (MTSI) gene in pancreatic adenocarcinoma. Nat. Gcnct., 8:27-32, 1994.

    21. Tada, M., Omata, M., Kawai, S.. Saisho, H., Ohto, M.. Saiki, R. K.,

    and Sninsky. J. J. Detection of ras gene mutations in pancreatic juice

    and peripheral blood of patients with pancreatic adenocarcinoma. Can-cer Res., 53: 2472-2474, 1993.

    22. Watanabe, H., Sawabu, N., Ohta, H., Satomura, Y., Yamakawa, 0.,

    Motoo, Y., Okai, T., Takahashi, H., and Wakabayashi, T. Identificationof K-ras oncogene mutations in the pure pancreatic juice of patients

    with ductal pancreatic cancers. Jpn. J. Cancer Res., 84: 961-965, 1993.

    23. Berth#{233}lemy, P., Bouisson, M., Escourrou, J., Vaysse. N., Rumeau,

    J. L.. and Pradayrol. L. Identification of K-ras mutations in pancreatic

    juice in the early diagnosis of pancreatic cancer. Ann. Intern. Mcd.. 123:

    188-191, 1995.

    24. Watanabe, H.. Sawabu, N., Songur. Y.. Yamaguchi, Y., Yamakawa,

    0., Satomura, Y., Ohta, H., Motoo, Y., Okai, 1., and Wakabayashi, T.

    Detection of K-ras point mutations at codon I 2 in pure pancreatic juicefor the diagnosis of pancreatic cancer by PCR-RFLP analysis. Pancreas,12: 18-24, 1996.

    25. Yanagisawa, A., Ohtake, K., Ohashi, K., Hon. M., Kitagawa, T.,

    Sugano. H., and Kato, Y. Frequent c-Ki-ras oncogene activation inmucous cell hyperplasias of pancreas suffering from chronic inflamma-tion. Cancer Res., 53: 953-956, 1993.

    26. Tada, M., Omata. M., and Ohto, M. Ras gene mutations in intra-ductal papillary neoplasms of the pancreas. Cancer (Phila.), 67: 634-

    637. 1991.

    27. Yanagisawa, A., Kato, Y., Ohtake, K., Kitagawa, T., Ohashi, K.,

    Hori, M., Takagi, K., and Sugano, H. c-K-ras point mutations inductectatic-type mucinous cystic neoplasms of the pancreas. Jpn. J.

    Cancer Res., 82: 1057-1060, 1991.

    28. Mizumoto, K., Suehara, N., Muta, T., Kitajima, S.. Hamasaki, N.,

    Tominaga, Y., Shimura, H., and Tanaka, M. Semi-quantitative analysis

    of telomerase in pancreatic ductal adenocarcinoma. J. Gastroenterol.,

    31: 894-897, 1996.

    29. Bcahrs, 0. H., Henson, D. E., Hutter, R. V. P., and Kennedy, B. J.

    (eds.). Pancreas. Manual for Staging of Cancer, Ed. 4. American Joint

    Committee on Cancer. Philadelphia: J. B. Lippincott Co., 1992.

    30. Bradford, M. M. A raid and sensitive method for the quantitation ofmicrogram quantities of protein utilizing the principle of protein-dye

    binding. Anal. Biochem., 72: 248-254, 1976.

    31. Tahara, H., Kuniyasu. H., Yokozaki, H., Yasui, W., Shay. J. W.,

    Ide, T., and Tahara, E. Tclomcrase activity in prencoplastic and nco-

    plastic gastric and colorcctal lesions. Clin. Cancer Rcs., 1: 1245-1251.1995.

    32. Mizumoto, K., Tsutsumi, M., Denda, A., and Konishi, Y. Rapid

    production of pancreatic carcinoma by initiation with N-nitroso-bis

    (2-oxopropyl)aminc and repeated augmentation pressure in hamsters.J. Nail Cancer Inst., 80: 1564-1567, 1988.

    33. Yamaguchi, K., Chijiiwa, K., Saiki, S., Nakatsuka, A., and Tanaka,

    M. “Mass-forming” pancreatitis masquerades as pancreatic carcinoma.

    Int. J. Pancrcatol., 20: 27-35, 1996.

    34. Hiyama, E., Hiyama, K., Yokoyama, T., Matsuura, Y., Piatyszck,

    M. A., and Shay, J. W. Correlating telomerase activity levels withhuman neuroblastoma outcomes. Nat. Mcd., 1: 249-255, 1995.

    Research. on June 6, 2021. © 1997 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • 1997;3:993-998. Clin Cancer Res N Suehara, K Mizumoto, T Muta, et al. to nonmalignant pathological states.Telomerase elevation in pancreatic ductal carcinoma compared

    Updated version

    http://clincancerres.aacrjournals.org/content/3/6/993

    Access the most recent version of this article at:

    E-mail alerts related to this article or journal.Sign up to receive free email-alerts

    Subscriptions

    Reprints and

    [email protected] at

    To order reprints of this article or to subscribe to the journal, contact the AACR Publications

    Permissions

    Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

    .http://clincancerres.aacrjournals.org/content/3/6/993To request permission to re-use all or part of this article, use this link

    Research. on June 6, 2021. © 1997 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/content/3/6/993http://clincancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://clincancerres.aacrjournals.org/content/3/6/993http://clincancerres.aacrjournals.org/