ArticlePDF Available

Distinct Chromosomal Profiles in Metastasizing and Non-Metastasizing Colorectal Carcinomas

Authors:

Abstract and Figures

The prognosis of colorectal cancer patients is to a considerable extent determined by the metastatic potency of the primary tumor. However, despite the fact that liver metastases are the leading cause of death for cancer patients, the molecular basis still remains poorly understood and independent prognostic markers have not been established. Comparative genomic hybridization (CGH) was used to screen colorectal carcinomas without distant metastases (n=18) and carcinomas synchronously metastatic to the liver (n=18). We aimed to detect distinct chromosomal aberrations indicating a metastatic phenotype. Metastatic tumors exhibited a significantly (P=0.03) higher ANCA value (13.8) if compared with non-metastatic cancers (10.0). Furthermore, we observed that losses of chromosomal regions 1p32-ter and 9q33-ter were present at much higher frequencies in metastatic than in non-metastatic cancers, respectively (P=0.02 and 0.04). These data indicate that metastatic tumors may be separated from non-metastatic colorectal cancers based on their genomic profile.
Content may be subject to copyright.
Cellular Oncology 28 (2006) 273–281 273
IOS Press
Distinct chromosomal profiles in
metastasizing and non-metastasizing
colorectal carcinomas
B. Michael Ghadimi
a,
,MarianGrade
a
, Carsten Mönkemeyer
a
, Bettina Kulle
b
, Jochen Gaedcke
a
,
Bastian Gunawan
c
, Claus Langer
a
, Torsten Liersch
a
and Heinz Becker
a
a
Department of General Surgery, University Medical Center, Göttingen, Germany
b
Department of Genetic Epidemiology, University Medical Center, Göttingen, Germany
c
Department of Pathology, University Medical Center, Göttingen, Germany
Abstract. Background: The prognosis of colorectal cancer patients is to a considerable extent determined by the metastatic
potency of the primary tumor. However, despite the fact that liver metastases are the leading cause of death for cancer patients,
the molecular basis still remains poorly understood and independent prognostic markers have not been established. Materials
and methods: Comparative genomic hybridization (CGH) was used to screen colorectal carcinomas without distant metastases
(n = 18) and carcinomas synchronously metastatic to the liver (n = 18). We aimed to detect distinct chromosomal aberrations
indicating a metastatic phenotype. Results and discussion: Metastatic tumors exhibited a significantly (P = 0.03) higher ANCA
value (13.8) if compared with non-metastatic cancers (10.0). Furthermore, we observed that losses of chromosomal regions 1p32-
ter and 9q33-ter were present at much higher frequencies in metastatic than in non-metastatic cancers, respectively (P = 0.02
and 0.04). Conclusion: These data indicate that metastatic tumors may be separated from non-metastatic colorectal cancers based
on their genomic profile.
Keywords: Colorectal cancer, comparative genomic hybridization, liver metastases, predictive marker, prognosis
Abbreviations: UICC, International Union against
Cancer; CGH, comparative genomic hybridization;
ANCA, average number of chromosomal copy alter-
ations.
1. Introduction
The stepwise progression of colorectal carcinomas
is accompanied by gains of chromosomes 7, 8q, 13
and 20, as well as losses of chromosomes 4, 8p and
18q [9,20,28,36]. In a previous study we demonstrated
that gains of chromosome 8q23-24 are associated with
lymph node positivity in non-metastatic colorectal car-
cinomas [16]. Whereas this aberration was detected in
the vast majority of lymph node positive tumors, it was
only rarely present in lymph node negative carcinomas
*
Corresponding author: B. Michael Ghadimi, MD, Department
of General Surgery, Georg-August-University Göttingen, Robert-
Koch-Str. 40, 37099 Göttingen, Germany. E-mail: mghadim@
uni-goettingen.de.
suggesting that genes located at 8q23-24 might favor
the development of lymphatic metastases in colorectal
cancers. Regarding metastatic disease, i.e. tumors with
systemic spread, several investigators evaluated the un-
derlying genomic changes of advanced colorectal can-
cers. But the results remain contradictory, and a chro-
mosomal aberration based metastatic genotype has not
been established [2–4,6,10,24,25,31,33].
From the clinical point of view, a significant num-
ber of colorectal cancer patients develop distant metas-
tases, preferably to the liver, even though approxi-
mately 60% to 70% of these patients will undergo po-
tential curative surgery at the time of cancer diagno-
sis [39]. Therefore, adjuvant chemotherapy is widely
considered as the gold standard for patients with UICC
stage III colon cancer, who are at high risk of recur-
rence [1,8]. Nevertheless, even stage II tumors have
the potential to form distant metastases, and approxi-
mately 20% of these patients die of recurrent disease
[29]. Prospective randomized trials have therefore been
initiated to evaluate the potential benefit of adjuvant
1570-5870/06/$17.00 2006 – IOS Press and the authors. All rights reserved
274 B.M. Ghadimi et al. / Distinct chromosomal profiles in colorectal carcinomas
chemotherapy for stage II colon cancer patients. Ac-
cordingly, accurate predictive biomarkers would help
to establish an individualized, metastatic phenotype-
based therapy [18].
The aim of the present study was to investigate
distinct patterns of copy number alterations in ad-
vanced colorectal cancers. We therefore used compar-
ative genomic hybridization (CGH) to determine ge-
nomic differences between non-metastatic colorectal
cancers (mainly UICC III) and cancers synchronously
metastatic to the liver (UICC stage IV) in an attempt to
identify chromosomal aberrations that might serve as
genetic markers for the risk of liver metastases.
2. Materials and methods
2.1. Materials
In the present study, we prospectively collected sur-
gical specimens from 36 patients diagnosed with a
colorectal cancer between 2000 and 2002. Only fresh
frozen tumor samples with a tumor cell content of at
least 70% (established on hematoxylin–eosin-stained
tissue sections) were analyzed. The histopathological
classification was based on the WHO histological typ-
ing of colorectal cancers [38]. The clinical data are
summarized in Table 1. All tumors were adenocarcino-
mas and have been classified as either non-metastatic
carcinomas (group 1; pT2-4 pN0-2 M0; n = 18) or as
cancers with hepatic metastases (group 2; pT2-4 pN0-
2M1;n = 18). We only selected cancer patients ex-
hibiting liver metastases synchronously, defined as di-
agnosed within 6 months following diagnosis of the
primary tumor.
2.2. Comparative genomic hybridization
CGH experiments and analysis were performed
as previously described [15]. Briefly, CGH was per-
formed on normal, sex-matched metaphase chromo-
somes prepared according to standard procedures fol-
lowing the criteria by du Manoir and colleagues
[13]. Normal DNA was labeled in a standard nick-
translation reaction substituting dTTP with digoxi-
genin-12-dUTP (Roche; Mannheim, Germany). Tumor
DNA was extracted using a commercially available
DNA-isolation KIT from Qiagen (Hilden, Germany)
and labeled by substituting dTTP with biotin-16-dUTP
(Roche; Mannheim, Germany).
For CGH, 300 ng of normal digoxigenin-labeled and
300 ng of biotin-labeled tumor DNA were ethanol pre-
cipitated in the presence of 30 µg of the Cot-1 frac-
tion of human DNA (Roche; Mannheim, Germany).
The probe DNA was dried and resuspended in 10 µlof
hybridization solution (50% formamide, 2× SSC, 10%
dextran sulfate), denatured (5 minutes at 75
C), and
preannealed for 1 hour at 37
C. The normal metaphase
chromosomes were denatured separately (70% for-
mamide, 2× SSC) for 2 minutes at 75
C. Hybridiza-
tion took place under a coverslip for 3 days at 37
C.
Posthybridization steps were performed as described in
detail elsewhere [15]. Biotin-labeled tumor sequences
were detected with FITC conjugated to avidin (Vector
laboratories; Burlingame, CA), and the digoxigenin-
labeled reference DNA was visualized with antidigoxi-
genin Fab fragments conjugated to rhodamine (Roche;
Mannheim, Germany). The chromosomes were coun-
terstained with 4,6-diamidino-2-phenylindole (DAPI;
Vector Laboratories; Burlingame, CA) and embed-
ded in an antifade solution containing paraphenylene-
diamine (Sigma; St. Louis, USA).
Gray-level images were acquired for each fluo-
rochrome using a CCD camera (Sensys, Photomet-
rics, Munich, Germany) coupled to an epifluorescence
microscope (Axiovert 25, Zeiss, Jena, Germany) us-
ing sequential exposure through fluorochrome spe-
cific filters. For automated karyotyping and analy-
sis a software package was used (Quips Karyotyp-
ing/CGH; Vysis; Downer’s Grove, USA). At least
12–15 metaphases have been evaluated according to
the guidelines suggested in the ISCN 1995 [30]. The
karyograms (Figs 1 and 2) summarize the individual
CGH experiments. The lines to the left of the chromo-
somal ideograms indicate chromosomal losses (ratio of
0.8), the lines to the right chromosomal gains (ratio
of 1.2). Bold lines indicate high-level copy number in-
creases, exceeding a threshold of 1.5 (amplifications).
Genomic instability was estimated as the average
number of copy alterations (ANCA), which is deduced
by dividing the total number of chromosomal copy al-
terations in a karyogram (see Figs 1 and 2) by the num-
ber of tumors analyzed (for details see [37]).
2.3. Data analysis
We first compared the average number of chromoso-
mal aberrations (ANCA values) of the non-metastatic
and the metastatic tumors with a two-sided t-test for in-
dependent samples. In addition, Fisher’s exact test was
used to determine the potential significant influence of
B.M. Ghadimi et al. / Distinct chromosomal profiles in colorectal carcinomas 275
Table 1
Clinical data of 36 patients with colorectal cancer
Age (yrs) Sex TNM staging UICC stage Grading Localization
Group 1
28 63 F pT2 pN0 (0/23) M0 I 2 Ascending colon
27 54 M pT3 pN0 (0/25) M0 II 2 Cecum
21 60 F pT3 pN0 (0/22) M0 II 2 Rectum
20 53 M pT3 pN0 (0/14) M0 II 2 Sigmoid colon
19 67 M pT3 pN0 (0/26) M0 II 2 Sigmoid colon
23 73 M pT2 pN1 (2/16) M0 III 2 Sigmoid colon
25 81 M pT2 pN1 (1/21) M0 III 2 Sigmoid colon
31 52 M pT3 pN1 (1/33) M0 III 2 Ascending colon
35 73 M pT3 pN1 (1/32) M0 III 2 Cecum
32 54 M pT3 pN2 (19/21) M0 III 2 Rectum
24 71 M pT3 pN2 (21/55) M0 III 2 Ascending colon
30 60 M pT3 pN2 (6/13) M0 III 2 Rectum
29 66 F pT3 pN2 (5/21) M0 III 2 Ascending colon
34 68 F pT3 pN2 (14/15) M0 III 2 Rectum
33 65 F pT3 pN2 (12/12) M0 III 2 Ascending colon
26 71 M pT4 pN2 (9/19) M0 III 2 Cecum
36 65 M pT4 pN2 (4/22) M0 III 2 Rectum
22 69 F pT4 pN2 (9/21) M0 III 2 Cecum
Group 2
18 63 M pT2 pN0 (0/25) M1 IV 2 Rectum
7 67 F pT3 pN0 (0/19) M1 IV 2 Descending colon
2 65 M pT3 pN0 (0/10) M1 IV 2 Descending colon
5 66 M pT3 pN0 (0/17) M1 IV 2 Sigmoid colon
16 60 M pT3 pN0 (0/17) M1 IV 2 Rectum
15 68 M pT2 pN1 (1/20) M1 IV 2 Rectum
8 49 F pT2 pN2 (5/21) M1 IV 2 Rectum
13 65 M pT3 pN1 (3/19) M1 IV 2 Rectum
12 53 M pT3 pN1 (3/21) M1 IV 2 Rectum
6 78 M pT3 pN2 (16/27) M1 IV 2 Sigmoid colon
11 78 F pT3 pN2 (20/24) M1 IV 2 Transverse colon
3 61 F pT3 pN2 (22/40) M1 IV 2 Ascending colon
14 64 M pT3 pN2 (6/17) M1 IV 2 Rectum
10 78 F pT3 pN2 (4/30) M1 IV 2 Ascending colon
17 60 M pT3 pN2 (18/21) M1 IV 2 Rectum
9 75 F pT4 pN2 (21/26) M1 IV 2 Rectum
1 49 F pT4 pN2 (29/30) M1 IV 2 Sigmoid colon
4 50 M pT4 pN2 (9/23) M1 IV 2 Sigmoid colon
276 B.M. Ghadimi et al. / Distinct chromosomal profiles in colorectal carcinomas
Fig. 1. Karyogram of chromosomal gains and losses in 18 colorectal carcinomas without liver metastases (group 1).
B.M. Ghadimi et al. / Distinct chromosomal profiles in colorectal carcinomas 277
Fig. 2. Karyogram of chromosomal gains and losses in 18 colorectal carcinomas with synchronous liver metastases (group 2).
278 B.M. Ghadimi et al. / Distinct chromosomal profiles in colorectal carcinomas
chromosomal gains and losses on the metastatic phe-
notype. Differences with a P<0.05 were considered
statistically significant.
3. Results
CGH was used to screen for copy number changes
in 36 patients with colorectal cancer. The clinical data
for all patients are presented in Table 1.
3.1. Colorectal carcinomas without liver metastases
(group 1)
All 18 tumors displayed chromosomal imbalances.
Overall, we detected 90 gains and 90 losses, resulting
in an ANCA value of 10. Frequent gains affected re-
gions on chromosomes 7 (50%), 8q (50%), 13 (61%),
20 (83%) and X (50%). Losses of chromosomal ma-
terial frequently mapped to 8p (33%), 14 (39%), 15
(33%), 17p (33%) and 18q (72%). Amplifications lo-
calized exclusively to chromosome arm 20q. Figure 1
summarizes the chromosomal aberrations in the ana-
lyzed non-metastatic tumors.
3.2. Colorectal carcinomas with synchronous liver
metastases (group 2)
In this group of carcinomas (n = 18), we observed
an ANCA value of 13.8 (104 gains and 144 losses;
Fig. 2). DNA gains affected regions of chromosomes 7
(56%), 8q (56%), 13 (72%) and 20 (83%). High-level
copy number increases could be mapped to chromo-
somal band 5p13 and chromosome arms 7p and 20q.
Decreased values were observed for chromosomal re-
gions of 1p (61%), 4q (33%), 8p (67%), 9q (39%), 15
(39%), 16p (44%), 17p (67%), 17q (44%), 18q (67%),
19 (50%), 20p (33) and 22 (50%).
3.3. Comparison of non-metastatic and metastatic
colorectal carcinomas
The metastatic tumors displayed a higher degree of
chromosomal instability than the non-metastatic tu-
mors, which is reflected by a mean ANCA value of
13.8 in group 2 and 9.9 in group 1, respectively (P =
0.03). Additionally, UICC stage IV cancers showed
significantly more chromosomal losses (P = 0.01),
whereas no significant difference could be detected
when comparing the chromosomal gains in the two
groups (P = 0.33). Furthermore, we identified two
Fig. 3. Frequencies of sub-chromosomal imbalances in metastatic
(group 2) and non-metastatic (group 1) colorectal cancers.
distinct chromosomal losses which were present at
much higher frequencies in metastatic than in non-
metastatic cancers, losses of 1p32-ter (P = 0.02)
and 9q33-ter (P = 0.04), respectively (Fig. 3). Of
note, chromosomal gains with higher frequencies in
metastatic cancers could only be mapped to chromo-
some 5p (P = 0.05).
4. Discussion
Several investigators evaluated the genomic changes
underlying metastases formation of colorectal can-
cers using CGH, but the results remain contradic-
tory (recently reviewed in [11,19]). Analyzing primary
Dukes’ stage C and D carcinomas and correspond-
ing metastases, Al-Mulla and colleagues found that
gains of chromosome arms 17q and 6p were signif-
icantly associated with metastatic disease [3]. Nakao
and colleagues reported that primary metastatic tu-
mors showed higher frequencies of gains of 6q, 7q,
8q, 13q and 20q [31]. Some of these findings have
B.M. Ghadimi et al. / Distinct chromosomal profiles in colorectal carcinomas 279
been particularly confirmed by other groups, looking
at either primary tumors or metastases [4,10,33]. How-
ever, many authors stated that the primary tumors in-
vestigated neither showed identical nor totally differ-
ent genomic aberrations when compared with the cor-
responding metastases. Usually, the metastases con-
tained additional gains and losses [3,4,21,24,25]. This
is supported by findings from Alcock and colleagues,
who analyzed microdissected sub-regions from pri-
mary tumors and corresponding hepatic metastases.
They reported that no two samples from one case were
identical, although common changes like gains of X
and 12q as well as losses of 8p, 16p, 9p, 1q, 18q and
10q were identified [2]. Interestingly, de Angelis and
colleagues did not find any chromosomal aberration
correlated with clinical stage [9].
Accordingly, defined chromosomal aberrations that
clearly distinguish metastatic colorectal tumors from
non-metastatic tumors remain to be established. How-
ever, it is important to note that some groups analyzed
DNA from primary tumors, whereas others examined
metastases. Since tumors with a potential metastatic
phenotype should already be identified at the time of
diagnosis, i.e. at the time a biopsy is taken, we fa-
vor to analyze the genomic features of the primary tu-
mor. One could also argue that, depending on the time
of resection, metastases could potentially have accu-
mulated additional DNA changes. These aberrations
would then not mirror the genomic features of the pri-
mary tumor, but instead just be the manifestation of a
longer growth process in a selective environment.
In the present investigation, we used compara-
tive genomic hybridization (CGH) to compare non-
metastatic colorectal cancers with cancers synchro-
nously metastatic to the liver. Our analysis revealed
that both groups have certain chromosomal aberrations
in common that have been previously identified, for ex-
ample gains of 7, 8q, 13 and 20 and losses of 4, 8p,
14, 15, 17p and 18. In particular, since 62% of the
UICC stage III cancers revealed gains of chromosome
8q23-24, we could confirm our previously reported re-
sult that lymph node positive cancers show high fre-
quencies of this chromosomal aberration [16]. Addi-
tionally, this study demonstrates that metastatic tumors
showed significantly more chromosomal losses than
non-metastatic tumors (P = 0.01). Furthermore, we
identified distinct chromosomal aberrations that were
present at much higher frequencies in metastatic than
in non-metastatic cancers, such as losses of 1p32-ter
and 9q33-ter, respectively (P = 0.02 and 0.04). Most
interestingly, these chromosomal losses were present
in small metastatic tumors (pT2) as well as in locally
advanced metastatic tumors (pT4), and in lymph node
negative metastatic tumors as well as in lymph node
positive metastatic tumors. Possibly, the incidence of
these chromosomal aberrations reflects a highly ag-
gressive metastatic genotype of colorectal cancers.
Even though it is well known that aberrations of
chromosome 1p need to be interpreted with care [28],
we think it is rather unlikely that they represent techni-
cal artifacts. Firstly, in the present investigation, these
chromosomal aberrations have been predominantly de-
tected in metastatic tumors, whereas loss of 1p and
9q was a rather rare event in non-metastatic tumors.
Secondly, several studies have previously shown that
chromosome arm 1p is commonly affected in colorec-
tal adenocarcinomas. Deletions of 1p have been identi-
fied in colorectal adenocarcinomas using conventional
cytogenetics, fluorescence in situ hybridization or loss
of heterozygosity analyses, and the most frequently af-
fected region has been shown to be 1p32-p36 [5,12,
14,17,27,34,35,40]. Furthermore, deletions in specific
sub-regions of 1p have been associated with a poor
prognosis [23,32].
In summary, this analysis identified distinct chro-
mosomal losses that might separate metastatic from
non-metastatic colorectal carcinomas, losses of chro-
mosomal regions 1p32-ter and 9q33-ter, respectively.
This indicates that it might be possible to establish re-
liable markers for prediction of the metastatic pheno-
type. Furthermore, one can speculate that tumor sup-
pressor genes, which are predominantly inactivated via
allelic loss, might be more important for the develop-
ment of metastatic disease than oncogenes. CDC2L1
[7], CDC2L2 [26] and TP73 [22] represent potential
target genes on chromosome arm 1p. Obviously, these
hypotheses need to be validated in larger prospective
analyses.
Acknowledgement
This manuscript is part of the doctoral thesis of C.
Mönkemeyer.
References
[1] A.M. Abulafi and N.S. Williams, Local recurrence of colorectal
cancer: the problem, mechanisms, management and adjuvant
therapy, Br.J.Surg.81 (1994), 7–19.
280 B.M. Ghadimi et al. / Distinct chromosomal profiles in colorectal carcinomas
[2] H.E. Alcock, T.J. Stephenson, J.A. Royds and D.W. Hammond,
Analysis of colorectal tumor progression by microdissection
and comparative genomic hybridization, Genes Chromosomes
Cancer 37 (2003), 369–380.
[3] F. Al-Mulla, W.N. Keith, I.R. Pickford, J.J. Going and G.D.
Birnie, Comparative genomic hybridization analysis of primary
colorectal carcinomas and their synchronous metastases, Genes
Chromosomes Cancer 24 (1999), 306–314.
[4] H. Aragane, C. Sakakura, M. Nakanishi, R. Yasuoka, Y. Fujita,
H. Taniguchi, A. Hagiwara, T. Yamaguchi, T. Abe, J. Inazawa
and H. Yamagishi, Chromosomal aberrations in colorectal can-
cers and liver metastases analyzed by comparative genomic hy-
bridization, Int. J. Cancer 94 (2001), 623–629.
[5] G. Bardi, T. Sukhikh, N. Pandis, C. Fenger, O. Kronborg and
S. Heim, Karyotypic characterization of colorectal adenocarci-
nomas, Genes Chromosomes Cancer 12 (1995), 97–109.
[6] T.E. Buffart, J. Coffa, M.A. Hermsen, B. Carvalho, J.R. van der
Sijp, B. Ylstra, G. Pals, J.P. Schouten and G.A. Meijer, DNA
copy number changes at 8q11-24 in metastasized colorectal
cancer, Cell. Oncol. 27 (2005), 57–65.
[7] B. Bunnell, L.S. Heath, D.E. Adams, J.M. Lahti and V.J. Kidd,
Elevated expression of a p58 protein kinase leads to changes
in the CHO cell cycle, Proc. Nat. Acad. Sci. U.S.A. 87 (1990),
7467–7471.
[8] A.M. Cohen, Surgical considerations in patients with cancer of
the colon and rectum, Semin. Oncol. 18 (1991), 381–387.
[9] P.M. De Angelis, O.P. Clausen, A. Schjolberg and T. Stokke,
Chromosomal gains and losses in primary colorectal carcino-
mas detected by CGH and their associations with tumour DNA
ploidy, genotypes and phenotypes, Br.J.Cancer80 (1999),
526–535.
[10] C.B. Diep, L.A. Parada, M.R. Teixeira, M. Eknaes, J.M. Nes-
land, B. Johansson and R.A. Lothe, Genetic profiling of col-
orectal cancer liver metastases by combined comparative ge-
nomic hybridization and G-banding analysis, Genes Chromo-
somes Cancer 36 (2003), 189–197.
[11] C.B. Diep, K. Kleivi, F.R. Ribeiro, M.R. Teixeira, O.C. Lindg-
jaerde and R.A. Lothe, The order of genetic events associ-
ated with colorectal cancer progression inferred from meta-
analysis of copy number changes, Genes Chromosomes Cancer
45 (2006), 31–41.
[12] A. Di Vinci, E. Infusini, C. Peveri, A. Sciutto, R. Orecchia, E.
Geido, R. Monaco and W. Giaretti, Intratumor heterogeneity
of chromosome 1, 7, 17, and 18 aneusomies obtained by FISH
and association with flow cytometric DNA index in human col-
orectal adenocarcinomas, Cytometry 35 (1999), 369–375.
[13] S. du Manoir, O.P. Kallioniemi, P. Lichter, J. Piper, P.A.
Benedetti, A.D. Carothers, J.A. Fantes, J.M. Garcia-Sagredo, T.
Gerdes, M. Giollant et al., Hardware and software requirements
for quantitative analysis of comparative genomic hybridization,
Cytometry 19 (1995), 4–9.
[14] H. Gerdes, Q. Chen, A.H. Elahi, A. Sircar, E. Goldberg, D.
Winawer, C. Urmacher, S.J. Winawer and S.C. Jhanwar, Re-
current deletions involving chromosomes 1, 5, 17, and 18 in
colorectal carcinoma: possible role in biological and clinical
behavior of tumors, Anticancer Res. 15 (1995), 13–24.
[15] B.M. Ghadimi, D.L. Sackett, M.J. Difilippantonio, E. Schrock,
T. Neumann, A. Jauho, G. Auer and T. Ried, Centrosome
amplification and instability occurs exclusively in aneuploid,
but not in diploid colorectal cancer cell lines, and correlates
with numerical chromosomal aberrations, Genes Chromosomes
Cancer 27 (2000), 183–190.
[16] B.M. Ghadimi, M. Grade, T. Liersch, C. Langer, A. Siemer, L.
Fuzesi and H. Becker, Gain of chromosome 8q23-24 is a pre-
dictive marker for lymph node positivity in colorectal cancer,
Clin. Cancer Res. 9 (2003), 1808–1814.
[17] W. Giaretti, S. Molinu, J. Ceccarelli and C. Prevosto, Chro-
mosomal instability, aneuploidy, and gene mutations in human
sporadic colorectal adenomas, Cell. Oncol. 26 (2004), 301–
305.
[18] M. Grade, H. Becker and B.M. Ghadimi, The impact of mole-
cular pathology in oncology: The clinician’s perspective, Cell.
Oncol. 26 (2004), 275–278.
[19] M. Grade, H. Becker, T. Liersch, T. Ried and B.M. Ghadimi,
Molecular cytogenetics: Genomic imbalances in colorectal
cancer and their clinical impact, Cell. Oncol. 28 (2006), 71–84.
[20] M.A. Hermsen, C. Postma, J.P. Baak, M. Weiss, A. Rapallo, A.
Sciutto, G. Roemen, J.W. Arends, R. Williams, W. Giaretti, A.
De Goeij and G.A. Meijer, Colorectal adenoma to carcinoma
progression follows multiple pathways of chromosomal insta-
bility, Gastroenterology 123 (2002), 1109–1119.
[21] M. Iwamoto, D. Banerjee, L.G. Menon, A. Jurkiewicz, P.H.
Rao, N.E. Kemeny, Y. Fong, S.C. Jhanwar, R. Gorlick and J.R.
Bertino, Overexpression of E2F-1 in lung and liver metastases
of human colon cancer is associated with gene amplification,
Cancer Biol. Ther. 3 (2004), 395–399.
[22] C.A. Jost, M.C. Marin and W.G. Kaelin Jr., p73 is a human p53-
related protein that can induce apoptosis, Nature 389 (1997),
191–194.
[23] T. Kambara, G.B. Sharp, T. Nagasaka, M. Takeda, H.
Sasamoto, H. Nakagawa, H. Isozaki, D.G. MacPhee, J.R. Jass,
N. Tanaka and N. Matsubara, Allelic loss of a common mi-
crosatellite marker MYCL1: a useful prognostic factor of poor
outcomes in colorectal cancer, Clin. Cancer Res. 10 (2004),
1758–1763.
[24] T. Knösel, S. Petersen, H. Schwabe, K. Schluns, U. Stein, P.M.
Schlag, M. Dietel and I. Petersen, Incidence of chromosomal
imbalances in advanced colorectal carcinomas and their metas-
tases, Virchows Arch. 440 (2002), 187–194.
[25] W.M. Korn, T. Yasutake, W.L. Kuo, R.S. Warren, C. Collins,
M. Tomita, J. Gray and F.M. Waldman, Chromosome arm
20q gains and other genomic alterations in colorectal cancer
metastatic to liver, as analyzed by comparative genomic hy-
bridization and fluorescence in situ hybridization, Genes Chro-
mosomes Cancer 25 (1999), 82–90.
[26] J.M. Lahti, M. Valentine, J. Xiang, B. Jones, J. Amann, J.
Grenet, G. Richmond, A.T. Look and V.J. Kidd, Alterations
in the PITSLRE protein kinase gene complex on chromosome
1p36 in childhood neuroblastoma, Nature Genet. 7 (1994),
370–375.
[27] I. Leister, A. Weith, S. Bruderlein, C. Cziepluch, D. Kangwan-
pong, P. Schlag and M. Schwab, Human colorectal cancer: high
frequency of deletions at chromosome 1p35, Cancer Res. 50
(1990), 7232–7235.
[28] G.A. Meijer, M.A. Hermsen, J.P. Baak, P.J. van Diest, S.G.
Meuwissen, J.A. Belien, J.M. Hoovers, H. Joenje, P.J. Sni-
B.M. Ghadimi et al. / Distinct chromosomal profiles in colorectal carcinomas 281
jders and J.M. Walboomers, Progression from colorectal ade-
noma to carcinoma is associated with non-random chromoso-
mal gains as detected by comparative genomic hybridization,
J. Clin. Pathol. 51 (1998), 901–909.
[29] R. Midgley and D. Kerr, Colorectal cancer, Lancet 353 (1999),
391–399.
[30] F. Mitelman, ISCN. An International System for Human Cyto-
genetic Nomenclature, S. Karger, Basel, 1995.
[31] K. Nakao, M. Shibusawa, A. Ishihara, H. Yoshizawa, A. Tsun-
oda, M. Kusano, A. Kurose, T. Makita and K. Sasaki, Genetic
changes in colorectal carcinoma tumors with liver metastases
analyzed by comparative genomic hybridization and DNA
ploidy, Cancer 91 (2001), 721–726.
[32] O.A. Ogunbiyi, P.J. Goodfellow, G. Gagliardi, P.E. Swanson,
E.H. Birnbaum, J.W. Fleshman, I.J. Kodner and J.F. Moley,
Prognostic value of chromosome 1p allelic loss in colon cancer,
Gastroenterology 113 (1997), 761–766.
[33] A. Paredes-Zaglul, J.J. Kang, Y.P. Essig, W. Mao, R. Irby, M.
Wloch and T.J. Yeatman, Analysis of colorectal cancer by com-
parative genomic hybridization: evidence for induction of the
metastatic phenotype by loss of tumor suppressor genes, Clin.
Cancer Res. 4 (1998), 879–886.
[34] C. Praml, L.H. Finke, C. Herfarth, P. Schlag, M. Schwab and L.
Amler, Deletion mapping defines different regions in 1p34.2-
pter that may harbor genetic information related to human col-
orectal cancer, Oncogene 11 (1995), 1357–1362.
[35] G. Ragnarsson, G. Eiriksdottir, J.T. Johannsdottir, J.G. Jonas-
son, V. Egilsson and S. Ingvarsson, Loss of heterozygosity at
chromosome 1p in different solid human tumours: association
with survival, Br.J.Cancer79 (1999), 1468–1474.
[36] T. Ried, R. Knutzen, R. Steinbeck, H. Blegen, E. Schröck, K.
Heselmeyer, S. du Manoir and G. Auer, Comparative genomic
hybridization reveals a specific pattern of chromosomal gains
and losses during the genesis of colorectal tumors, Genes Chro-
mosomes Cancer 15 (1996), 234–245.
[37] T. Ried, K. Heselmeyer-Haddad, H. Blegen, E. Schrock and
G. Auer, Genomic changes defining the genesis, progression,
and malignancy potential in solid human tumors: a pheno-
type/genotype correlation, Genes Chromosomes Cancer 25
(1999), 195–204.
[38] L.H. Sobin and C. Wittekind, UICC: TNM Classification of
Malignant Tumors, 5th edn, John Wiley & Sons, New York,
1997.
[39] G. Steele Jr. and T.S. Ravikumar, Resection of hepatic metas-
tases from colorectal cancer. Biologic perspective, Ann. Surg.
210 (1989), 127–138.
[40] L. Thorstensen, H. Qvist, S. Heim, G.J. Liefers, J.M. Nesland,
K.E. Giercksky and R.A. Lothe, Evaluation of 1p losses in pri-
mary carcinomas, local recurrences and peripheral metastases
from colorectal cancer patients, Neoplasia 2 (2000), 514–522.
... It is established that CRC develops through several pathways resulting in different cytogenetic and molecular characteristics of the tumour [9]. The largest subset of CRCs evolves through chromosomal instability (CIN), which results in common chromosomal aberrations including losses at 8p, 17p, and 18q as well as gains at 7p, 7q, 8q, 13q, and 20q, and more variably losses at 1p, 4p, 4q, 5q, 14q, 15q, and 18p as well as gains at 1q, and 20p [10][11][12][13][14][15][16][17]. In contrast, CRC with a high-degree of microsatellite (MS) instability (MSI-H) [18][19][20] often present with a stable, near-diploid karyotype, although some MSI-H tumours show chromosomal imbalances as well [21][22][23][24][25]. MSI-H is characterised by disruption of the DNA mismatch repair (MMR) system that maintains DNA sequence fidelity [26]. ...
... Furthermore, our oncogenetic tree modeling attributed -1p, -4, and -5q, amongst others, as late events in tumour evolution. Hepatic metastasis were previously shown to be enriched in losses at 1p [17], and loss of chromosome 4 has been linked to advanced stages and metastatic events in patients with CRCs [65]. Loss of 5q was shown to represent an aberration acquired in brain and pulmonary metastasis of CRCS, while -5q was only rarely observed in the corresponding primary tumours [12,13], which independently provides evidence for -5q as late event. ...
Article
Full-text available
Background and aim: Colorectal carcinomas (CRCs) progress through heterogeneous pathways. The aim of this study was to analyse whether or not the cytogenetic evolution of CRC is linked to tumour site, level of chromosomal imbalances and metastasis. Method: A set of therapy-naïve pT3 CRCs comprising 26 proximal and 49 distal pT3 CRCs was studied by combining immunohistochemistry of mismatch repair (MMR) proteins, microsatellite analyses, molecular karyotyping as well as clinical parameters. Results: An MMR deficient/microsatellite-instabile (dMMR/MSI-H) status was associated with a location of the primary tumour proximal to the splenic flexure, and dMMR/MSI-H tumours presented with significantly lower levels of chromosomal imbalances in comparison to MMR proficient/microsatellite-stable (pMMR/MSS) tumours. Oncogenetic tree modeling suggested two evolutionary clusters characterised by dMMR/MSI-H and chromosomal instability (CIN), respectively, for both, the proximal and distal CRCs. In CIN cases, +13q, -18q, and +20q were predicted as preferentially early events, and -1p, -4, -and -5q as late events. Separate oncogenetic tree models of proximal and distal cases indicated similar early events independent of tumour site. However, in cases with high CIN defined by more than 10 copy number aberrations, loss of 17p occurred earlier in cytogenetic evolution than in cases showing low to moderate CIN. Differences in the oncogenetic trees were observed for CRCs with lymph node and distant metastasis. Loss of 8p was modeled as early event in node-positive CRC, while +7p and +8q comprised early events in CRC with distant metastasis. Conclusion: CRCs characterised by CIN follow multiple, interconnected genetic pathways in line with the basic 'Vogelgram' concept proposed for the progression of CRC that places the accumulation of genetic changes into the center of tumour evolution. However, the timing of specific genetic events may favour the metastatic potential.
... These chromosomal alterations have been extensively described in the literature in sCRC [57,68,109,117,118]. In turn, they found a genetic profile more specific for liver metastatic sCRC that included losses at the 1p36-p33, 17p11 and 18q12-23 chromosome regions, together with gains of chromosome 7 and 13q22-34, detected with higher frequency in primary tumors from liver metastatic sCRC [123], in line with data from other groups reported in the literature [122][123][124]. Furthermore, the presence of alterations at most of these chromosome regions was also associated with a poorer OS, among their sCRC patients, supporting the (potential) clinical and functional relevance of genes encoded in these regions in the migration and homing of tumor cells to the liver. ...
... For example, loss of chromosome 1p33, where a potential tumor suppressor gene (i.e. ELAVL4) is encoded [125], has been recurrently associated in the literature with aggressive phenotypes and poor sCRC patient outcome [124,126,127]. Similarly, deletion of 17p is a well-known chromosomal alteration that is present in sCRC, particularly among patients with more advanced disease stages, that has also been associated with disease progression [32,55,128]. ...
Article
Sporadic colorectal cancer (sCRC) is the third leading cause of cancer death in the Western world. Approximately, a quarter of sCRC patients present metastatic dissemination at the moment of diagnosis, the liver being the most frequently affected organ. Additionally, this group of CRC patients is characterized by a worse prognosis. In the last decades, significant technological developments for genome analysis have fostered the identification and characterization of genetic alterations involved in the pathogenesis of sCRC. However, genetic alterations involved in the metastatic process through which tumor cells are able to colonize other tissues with a different microenvironment, still remain to be fully identified. Here, we review current knowledge about the most relevant genomic alterations involved in the liver metastatic process of sCRC, including detailed information about the genetic profile of primary colorectal tumors vs. their paired liver metastases.
... The presence of LOH in chromosomal regions bearing tumor suppressors is key for events of tumor evolution. Previous reports of LOH incidence in CRC shows that 1p36 deletion is a common event (Ragnarsson et al., 1999;Lin Thorstensen et al., 2000) related to a bad prognosis (Ogunbiyi et al., 1997) and metastasis (Thorstensen et al., 2000;Ghadimi et al., 2006). Also, deletions in this chromosomal area could affect morphology and behavior of the CRC tumor itself (Fijneman et al., 2007). ...
Article
Full-text available
High incidence of Rho Cdc42-GTPase overexpression has been found in Colorectal Cancer (CRC) samples, suggesting its potential role in tumor development. However, no conclusive studies have shown the lack of mutations and/or copy number of Cdc42 gene in this type of samples. To understand mutation/deletion and copy number status of Cdc42 gene, CRC patients were evaluated for both parameters. More than Cdc42 mutants, single-nucleotide variants were found. Analysis of regions flanking the Cdc42 gene showed allelic imbalance; 58.7% were loss of heterozygosity (LOH) positive and 14.8% presented microsatellite instability. The highest LOH percentage was located between microsatellite markers D1S199 and D1S2674, where the Cdc42 gene is located. No association between gender, age, and tumor stage was found. LOH validation through gene dosage analysis showed most CRC patients with allelic imbalance also presented a low gene dosage of Cdc42, although equal amounts of Cdc42 mRNA were detected in all samples. Although changes in Cdc42 expression were not found in any condition, Cdc42 activation was different between high and normal gene dosage samples, but not between samples with normal and low copy number. Low dosage of Cdc42 was also not related to changes in methylation status at the Cdc42 promoter region. Results suggest that low copy of Cdc42 gene is not associated with Cdc42 protein dysfunction in CRC patients.
... 50,54 In line with our findings, Ghadimi et al observed a greater frequency of chromosome 1p32ter loss in metastatic versus nonmetastatic cancer (61% vs 11% of patients) using comparative genomic hybridization. 53 Our results indicate a particularly high frequency of losses involving the 1p33 chromosomal locus, which may harbor relevant tumor suppressor genes like the embryonic lethal, abnormal vision (ELAV)-like neuron specific RNA binding protein 4 (ELAVL4) gene. Of note, Stawski et al recently reported significantly decreased expression of ELAVL4 among male patients with meningiomas who carried deletions at D1S162, supporting the Abbreviations: bp, base pairs; HR, hazard ratio; NS, statistically nonsignificant (P > .05); ...
Article
Metastatic dissemination is the most frequent cause of death in patients with sporadic colorectal cancer (sCRC). It is believed that the metastatic process is related at least in part to a specific background of genetic alterations accumulated in cells from primary tumors, and the ability to detect such alterations is critical for the identification of patients with sCRC who are at risk of developing metastases. The authors used high-resolution, 500-K single nucleotide polymorphism arrays to identify copy number alteration profiles present at diagnosis in primary tumors from patients with metastatic (n = 23) versus nonmetastatic (n = 26) sCRC. The results revealed a characteristic pattern of copy number alterations in metastatic sCRC tumors that involved losses of 23 regions at chromosomes 1p, 17p, and 18q, together with gains of 35 regions at chromosomes 7 and 13q. In line with expectations, the copy number profile investigated involved multiple genes that were associated previously with sCRC (ie, SMAD2) and/or the metastatic process (ie, podocalyxin-like [PODXL]), and it also was associated with a poorer outcome. Cancer 2014. © 2014 American Cancer Society.
... Loss of heterozygosity events at 14q12-13 and 14q32 had earlier been associated with metastatic recurrence of early-stage CRC [25] but is not linked to CNAs at the initial stage. Loss of 1p has been associated with metastatic CRC with an increased frequency and was reported in non-metastatic CRC as well [26]. Other reports of copy number losses have implicated chromosome 5 [5] and 15 [27]. ...
Article
Full-text available
Genomic abnormalities leading to colorectal cancer (CRC) include somatic events causing copy number aberrations (CNAs) as well as copy neutral manifestations such as loss of heterozygosity (LOH) and uniparental disomy (UPD). We studied the causal effect of these events by analyzing high resolution cytogenetic microarray data of 15 tumor-normal paired samples. We detected 144 genes affected by CNAs. A subset of 91 genes are known to be CRC related yet high GISTIC scores indicate 24 genes on chromosomes 7, 8, 18 and 20 to be strongly relevant. Combining GISTIC ranking with functional analyses and degree of loss/gain we identify three genes in regions of significant loss (ATP8B1, NARS, and ATP5A1) and eight in regions of gain (CTCFL, SPO11, ZNF217, PLEKHA8, HOXA3, GPNMB, IGF2BP3 and PCAT1) as novel in their association with CRC. Pathway and target prediction analysis of CNA affected genes and microRNAs, respectively indicates TGF-β signaling pathway to be involved in causing CRC. Finally, LOH and UPD collectively affected nine cancer related genes. Transcription factor binding sites on regions of >35% copy number loss/gain influenced 16 CRC genes. Our analysis shows patient specific CRC manifestations at the genomic level and that these different events affect individual CRC patients differently.
Article
Chromosomal instability together with microsatellite and epigenetic instability is a defined characteristic of a variety of human cancers. Studies on chromosomal instability are first line examinations that allow the location of critical regions or genes crucial for cancer development and progression. Sporadic colorectal cancer is one of the most common cancers in the world. According to its biology it can be divided into two groups: microsatellite instable and chromosomal instable. Chromosomal instability is characteristic for about 85% of sporadic colorectal cancer; therefore a correlation between a pattern of chromosomal aberrations and histopathological as well as clinical features of colorectal cancer is widely investigated. Unbalanced aberrations can be found using the CGH technique without the step of cell culture. The presence of specific aberrations has been reported for colorectal cancer, which can be interpreted as the presence of a stable cancer genotype. This information may be useful for clinical therapy.
Article
A genomic analysis (Comparative Genomic Hybridization) evidenced, among other chromosomic alterations, a microdeletion at 1p36.12 locus in 23% and 47% of colon adenomas and carcinomas, respectively. Among the 15 genes located in the deleted region, we focused on E2F2 gene involvedin various cellular processes. The Kaplan-Meier curve analysis showed that E2F2 deletion is associated with a better progression-free survival. To better understand the involvement of genes targeted by the microdeletion in colon tumors, we assessed the functional impact of the underexpression of E2F2 and of its direct neighbor gene ID3, coding for a dominant-negative repressor involved in cell differentiation. Our results indicated that E2F2 loss favors tumor growth and prevent metastatic spread. In collaboration with the biochemical team directed by the Pr Céline TARNUS, we started a pilotstudy to prove the anti-tumor potential of new chemical inhibitors highly selective of the aminopeptidase N.
Article
The mouse secretory phospholipase A2 group IIA (sPLA2-IIA) gene Pla2g2a has been identified as a susceptibility gene for cancer of the small and large intestine. Interestingly, unlike most previously identified tumor susceptibility genes, Pla2g2a does not behave like a classical oncogene or tumor suppressor gene. Hence, identification of its biological functions in tumor development may shed new light on general mechanisms that modulate colon cancer risk. So far, sPLA2-IIA has been proposed to play a role in anti-bacterial defense, inflammation and eicosanoid generation, in clearance of apoptotic cells, and in the Wnt signaling pathway. More recently, comparison of RNA expression profiles of colon from Pla2g2a-transgenic to Pla2g2a-deficient mice confirmed and even extended sPLA2-IIA's diverse biological effects. In this review we aim to summarize current knowledge about the various links of sPLA2-IIA to cancer of the gastro-intestinal tract, and propose several models to illustrate its putative biological effects on tumor development.
Article
At present no objective parameters to identify the risk of liver metastasis after surgery have been established in rectal cancer. To identify the chromosomal aberrations that are correlated with liver metastasis of rectal cancer. Primary tumor tissues of rectal carcinoma were analyzed by array-based comparative genomic hybridization (array-CGH). Genomic aberrations were identified by Genomic Workbench and MD-SeeGH. The most frequent gains in rectal cancer were at 20q11.21-q13.33, 8q11.21-q24.3, 13q12.11-q14.2 and losses in 5q13.2, 8p23.3-p22, 17p13.3-p13.2 and 18q11.2-q23. Seven amplifications at 6p21.1, 8q24.21, 8q24.3, 13q13.2 and 20q13.2-q13.32 and nine homozygous deletions at 1q31.3, 4q12-q13.1, 4q32.3-q33, 5q13.2, 8p23.2, 8q11.23, 16p13.2, 19p13.11 and 19q13.41 were identified. Both frequency plot comparison and SAM (Significance analysis of microarray) methods indicated that losses at 1p35.3, 4p14, 14q23.1-q32.11 and 18p11.32-p11.21 were more frequent in patients without liver metastasis. These liver metastasis associated genomic changes may be useful to reveal the mechanism of metastasis and identify candidate biomarkers.
Article
Intestinal homeostasis is dependent on the proper host/microbiota interaction via pattern recognition receptors. Toll-like receptors are a specialised group of membrane receptors which detect pathogen-associated conserved structures. They are present in the intestinal tract and are required for intestinal homeostasis. Dysregulation in the Toll-like receptor signalling can conceivably result in a dysregulated immune response which could contribute to major intestinal pathologies including colorectal cancer. Evidence for the role of microbiota and toll-like receptors in colorectal cancer is emerging. In this report the evidence for the contribution of toll-like receptors to the pathogenesis of colorectal cancer; potential mechanisms affecting toll-like receptor signalling; and their therapeutic targeting in colorectal cancer are reviewed.
Article
Full-text available
Measurement of the nuclear DNA content allows classification of human cancers as either diploid or aneuploid. To gain further insight into mechanisms of aneuploidy, we compared the cytogenetic profile of mismatch-repair–deficient diploid versus mismatch-repair–proficient aneuploid colorectal carcinoma cell lines using comparative genomic hybridization and spectral karyotyping. Aneuploid carcinomas revealed an average of 19 chromosomal imbalances per cell line. Such numerical aberrations were exceedingly scarce in the diploid tumors. This pattern of chromosomal aberrations is consistent with a mechanism involving the impairment of chromosome segregation fidelity during mitotic cell division. In support of this idea, we demonstrate the exclusive occurrence of centrosome amplification and instability in all of the aneuploid tumor cell lines analyzed. All diploid tumors contained centrosomes that were functionally and structurally indistinguishable from those in normal human fibroblasts. Due to the observed differences in centrosomes between these two classes of tumors, we incubated the cells with the microtubule depolymerizing drugs nocodazole and griseofulvin. Our results indicate that the aneuploid tumor cell lines have an increased sensitivity to these reagents and a delay in aster formation and microtubule regrowth. However, microtubule nucleation was initiated from one or two centers in both the diploid and aneuploid cells. These observations support the notion that the integrity of the centrosome plays a central role in the development of aneuploidy. Genes Chromosomes Cancer 27:183–190, 2000. Published 2000 Wiley-Liss, Inc.
Article
The transition of normal epithelium to invasive carcinoma occurs sequentially. In colorectal and cervical carcinogenesis, this transition is reflected by histomorphologically defined grades of increasing dysplasia that untreated may progress to invasive disease. In an attempt to understand the role of chromosomal aberrations during tumorigenesis we have applied comparative genomic hybridization using DNA extracted from defined stages of colorectal and cervical tumors, from low‐ and high‐grade astrocytic tumors and from diploid and aneuploid breast carcinomas. Genetic instability, as measured by the number of chromosomal copy alterations per case, increases significantly at the transition from precursor lesions to invasive carcinomas and continues to increase with tumor stage. Aggressive tumors have a higher number of copy alterations per case. High‐level copy number changes (amplifications) become more prevalent in advanced‐stage disease. Subtractive karyograms of chromosomal gains and losses were used to map tumor stage‐specific chromosomal aberrations and clearly showed that nonrandom chromosomal aberrations occur during disease progression. In colorectal and cervical tumors, chromosomal copy number changes were correlated with nuclear DNA content, proliferative activity, expression levels of the tumor suppressor gene TP53, and the cyclin‐dependent kinase inhibitor p21/WAF1, as well as the presence of viral genomes. Here we summarize and review the results of this comprehensive phenotype/genotype correlation and discuss the relevance of stage‐specific chromosomal aberrations with respect to diagnostic applications. Genes Chromosomes Cancer 25:195–204, 1999. Published 1999 Wiley‐Liss, Inc.
Article
Liver metastases are found in 10% of primary colorectal malignancies, and they affects the prognosis of patients with colorectal carcinoma. The authors investigated DNA copy number aberrations by using comparative genomic hybridization (CGH) and DNA ploidy alterations by using flow cytometry (FCM) in patients with primary colorectal carcinoma (primary tumors). To determine whether there are characteristic DNA copy number alterations that contribute to liver metastasis, cytogenetic aberrations were examined by CGH and FCM. The authors analyzed 35 primary tumors, including 16 primary tumors with liver metastasis, by using CGH and FCM. Increases in DNA copy numbers were detected in 6q (5 of 16 tumors), 7q (6 of 16 tumors), 8q (7 of 16 tumors), 9p (5 of 16 tumors), 13q (8 of 16 tumors), 20p (9 of 16 tumors), and 20q (15 of 16 tumors) in primary tumors with liver metastases. Decreases in DNA copy numbers were found in 17p (5 of 16 tumors), 18p (6 of 19 tumors), 18q (8 of 16 tumors), and 22q (5 of 16 tumors). In contrast, primary tumors without liver metastasis showed gains in chromosome arms 8q (2 of 19 tumors), 13q (2 of 19 tumors), 20p (6 of 19 tumors), and 20q (5 of 19 tumors); however, they showed no gains in 6q or 7q and showed losses in chromosome arms 17p (2 of 19 tumors), 18p (4 of 19 tumors), 18q (6 of 19 tumors), and 22q (5 of 19 tumors). There was a significant difference in the frequency of DNA copy number gains and losses in 6q (P < 0.05), 7q (P < 0.01), 8q (P < 0.05), 13q (P < 0.05), and 20q (P < 0.01), respectively, between primary tumors with and without liver metastases. The differences in the DNA index were not significant between the two groups of primary tumors. In liver metastases of primary tumors from patients with colorectal carcinoma, a correlation between DNA copy number aberrations and gains of chromosome arms 6q, 7q, 8q, 13q, and 20q is suggested. Cancer 2001;91:721–6.
Article
Comparative genomic hybridization (CGH) was used to screen for changes in the number of DNA sequence copies in 30 primary colorectal cancers and 16 liver metastases, to identify regions that contain genes important for the development and progression of colorectal cancer. In primary colorectal cancer, we found frequent gains at 7p21 (36.7%), 7q31-36 (30%), 8q23-24 (43.0%), 12p (30%), 14q24-32 (33.3%), 16p (40.0%), 20p (33.3%), 20q (63.3%) and 21q (36.3%), while loss was often noted at 18q12-23 (36.7%). In metastatic tumors, there were significantly more gains and losses of DNA sequences than in primary tumors, with gains at 8q23-24 (found in 62.5% of recurrences vs. 43.0% of primary tumors), 15q21-26 (37.5% vs. 20.0%), 19p (43.8% vs. 20.0%) and 20q (81.3% vs. 63.3%) and losses at 18q12-23 (50.0% vs. 36.7%). The pattern of genetic changes seen in metastatic tumors, with frequent gains at 8q23-24 and 20q and loss at 18q12-23, suggests the progression of colorectal cancer. We investigated a clinical follow-up study for all patients examined by CGH and directed our attention to the genetic changes consisting of gains at 8q and 20q. The incidence of liver metastases was higher in patients with primary colorectal cancer with these genetic changes. Gains at 8q and 20q might be useful to identify patients at high risk for developing liver metastases. © 2001 Wiley-Liss, Inc.
Article
BACKGROUND Liver metastases are found in 10% of primary colorectal malignancies, and they affects the prognosis of patients with colorectal carcinoma. The authors investigated DNA copy number aberrations by using comparative genomic hybridization (CGH) and DNA ploidy alterations by using flow cytometry (FCM) in patients with primary colorectal carcinoma (primary tumors). To determine whether there are characteristic DNA copy number alterations that contribute to liver metastasis, cytogenetic aberrations were examined by CGH and FCM.METHODS The authors analyzed 35 primary tumors, including 16 primary tumors with liver metastasis, by using CGH and FCM.RESULTSIncreases in DNA copy numbers were detected in 6q (5 of 16 tumors), 7q (6 of 16 tumors), 8q (7 of 16 tumors), 9p (5 of 16 tumors), 13q (8 of 16 tumors), 20p (9 of 16 tumors), and 20q (15 of 16 tumors) in primary tumors with liver metastases. Decreases in DNA copy numbers were found in 17p (5 of 16 tumors), 18p (6 of 19 tumors), 18q (8 of 16 tumors), and 22q (5 of 16 tumors). In contrast, primary tumors without liver metastasis showed gains in chromosome arms 8q (2 of 19 tumors), 13q (2 of 19 tumors), 20p (6 of 19 tumors), and 20q (5 of 19 tumors); however, they showed no gains in 6q or 7q and showed losses in chromosome arms 17p (2 of 19 tumors), 18p (4 of 19 tumors), 18q (6 of 19 tumors), and 22q (5 of 19 tumors). There was a significant difference in the frequency of DNA copy number gains and losses in 6q (P < 0.05), 7q (P < 0.01), 8q (P < 0.05), 13q (P < 0.05), and 20q (P < 0.01), respectively, between primary tumors with and without liver metastases. The differences in the DNA index were not significant between the two groups of primary tumors.CONCLUSIONS In liver metastases of primary tumors from patients with colorectal carcinoma, a correlation between DNA copy number aberrations and gains of chromosome arms 6q, 7q, 8q, 13q, and 20q is suggested. Cancer 2001;91:721–6. © 2001 American Cancer Society.
Article
Recommendations are made for hardware and software capabilities that will permit a level of performance of comparative genomic hybridization (CGH) analysis on metaphase chromosomes that is comparable to the best current practice. Guidelines for interpreting the results of CGH analysis in terms of chromosomal gains or losses are also presented. © 1995 Wiley-Liss, Inc.