ArticlePDF Available

Most Early Disseminated Cancer Cells Detected in Bone Marrow of Breast Cancer Patients Have a Putative Breast Cancer Stem Cell Phenotype

Authors:

Abstract and Figures

The presence of disseminated tumor cells (DTC) in the bone marrow of breast cancer patients is an acknowledged independent prognostic factor. The biological metastatic potential of these cells has not yet been shown. The presence of putative breast cancer stem cells is shown both in primary tumors and distant metastases. These cells with a CD44(+)CD24(-/low) phenotype represent a minor population in primary breast cancer and are associated with self-renewal and tumorigenic potential. Recognizing the potential effect of prevalence of putative stem cells among DTC, we evaluated the bone marrow DTC. We employed the double/triple-staining immunohistochemistry protocol and modified the established bone marrow cytokeratin (CK) staining protocol by adding steps for additional antigens, CD44 and/or CD24. We evaluated 50 bone marrow specimens, previously categorized as CK(+) from early breast cancer patients. CK(+) cells were examined for CD44 and CD24 expression by light microscopy, fluorescence microscopy, and spectral imaging. We detected the putative stem cell-like phenotype in all CK(+) specimens. The mean prevalence of putative stem/progenitor cells was 72% and median prevalence was 65% (range, 33-100%) among the overall DTC per patient, compared with primary tumors where this phenotype is reported in <10% of cells. This is the first evidence of the existence of the putative stem-like phenotype within the DTC in bone marrow in early breast cancer patients. All patients had a putative stem cell phenotype among the DTC and most individual DTC showed such phenotype. Future molecular characterization of these cells is warranted.
A and B, double staining immunohistochemistry results for breast cancer bone marrow samples. The bone marrow samples show presence of distinct stem-like and non-stem tumor cells (magnification, Â40). A, two tumor cells with brown cytoplasmic staining for CK (DAB) and red membranous staining for CD24 (Vector Red). B, an example of a CK + CD24 À cell (putative breast cancer stem cell). C to F, the applicability of spectral imaging analysis for immunohistochemistry double-stained samples, where the chromogens are not easily distinguishable with the naked eye. Here, the cytospun bone marrow aspirates were assessed for CK (red ; stained with Fast Red) and CD44 (brown ; stained with DAB). The cells were counterstained with hematoxylin. C, transmitted light image, showing overall cell population subjected to double-staining for CK and CD44. Arrow, two CK positive cells that are difficult to assess for their CD44 positivity. D and E, spectrally unmixed images with pseudocolors assigned to hematoxylin (D) and DAB (CD44; E) using SpectraView software, showing that all the cells in the field show nuclear hematoxylin as well as CD44 staining, including the two cells denoted by arrow. F, fluorescence image obtained for CK with Cy3 filter for Fast Red. G to J, applicability of spectral imaging analysis for immunohistochemistry double-stained samples for CK and CD24. In this case, the expression of CK was assessed using DAB (brown) and that of CD24 with Fast red (red). The cells were counterstained with hematoxylin. G, transmitted light image, showing overall cell population subjected to double-staining for CK and CD24. Arrow, a CK-positive cell that appears as CD24 À/low . To confirm that this cell is indeed CD24 À/low , we did further spectral imaging analysis. Spectrally unmixed images with pseudocolors assigned to hematoxylin (H) and DAB (CK; I). These images show that, whereas all cells in the field show nuclear hematoxylin, only the cell denoted is positive for CK (DAB). J, fluorescence image obtained for CD24 staining with Cy3 filter for Fast Red. Spectral imaging analysis of the cell denoted in (G) shows that the cell at this location expresses no/low CD24 (red). K, another CK-positive cell; L, the same cell with specific membranous fluorescence demarking the CD44 positivity; note the surrounding CD44 + lymphocytes.
… 
Content may be subject to copyright.
Most Early Disseminated Cancer Cells Detected in Bone
Marrow of Breast Cancer Patients Have a Putative
Breast Cancer Stem Cell Phenotype
Marija Balic,
1
Henr y Lin,
1
LillianYoung,
1
Debra Hawes,
1
Armando Giuliano,
2
George McNamara,
3,4
Ram H. Datar,
1
and Richard J. C ote
1
Abstract Purpose: The presence of disseminated tumor cells (DTC) in the bone marrow of breast cancer
patients is an acknowledged independent prognostic factor.The biological metastatic potential of
these cells has not yet been shown. The presence of putative breast cancer stem cells is shown
both in primary tumors and distant metastases. These cells with a CD44
+
CD24
/low
phenotype
represent a minor population in primary breast cancer and are associated with self-renewal and
tumorigenic potential. Recognizing the potential effect of prevalence of putative stem cells among
DTC, we evaluated the bone marrow DTC.
Experimental Design: We employed the double/triple-staining immunohistochemistry
protocol and modified the established bone marrow cytokeratin (CK) staining protocol by
adding steps for additional antigens, CD44 and/or CD24.We evaluated 50 bone marrow speci-
mens, previously categorized as CK
+
from early breast cancer patients. CK
+
cells were examined
for CD44 and CD24 expression by light microscopy, fluorescence microscopy, and spectral
imaging.
Result s: We detected the putative stem cell ^ like phenotype in all CK
+
specimens. The mean
prevalence of putative stem/progenitor cells was 72% and median prevalence was 65 % (range,
33-100%) among the overall DTC per patient, compared with primary tumors where this pheno-
type is reported in <10 % o f c el ls .
Conclusions: This is the first evidence of the existence of the putative stem-like phenotype
within the DTC in bone marrow in early breast cancer patients. All patients had a putative
stem cell phenotype among the DTC and most individual DTC showed such phenotype. Future
molecular characterization of these cells is warranted.
The most important factor influencing the outcome of
patients with an invasive cancer is whether the tumor has
spread regionally or systemically (1). Metastases start out as
single cells that detach from the primary tumor and travel to
distal locations; these cells in transit, however, are not
detectable by routine histologic or radiologic methods. Tumor
cell dissemination to the bone marrow is an early event in
progression of breast cancer (1) and is associated with poor
prognosis (24). Disseminated tumor cells (DTC) are
commonly detected by immunohistochemistry for epithelial
antigens, such as cytokeratins (CK). The biology of these cells
is not well understood and is critical for therapeutic improve-
ments. Recent evidence suggests that only a certain type of
tumor cells, cancer stem cells or cancer initiating cells, harbor
tumorigenic potential (5). The hypothesis that the initiation
of malignancy has to take place in cancer stem cells derives
from the observation that it often takes many months or
years for the promotion stage of carcinogenesis to occur,
suggesting that the cancer stem cells must stay viable over a
long period (6). Such cells have been identified for hema-
tologic malignancies (7), brain tumors (8), and breast cancers.
For breast cancers, such cells have been identified to have a
CD44
+
CD24
/low
phenotype (9) and represent a minor
population (10-20%) within primary tumors (10). Cells with
this phenotype have been shown to be tumorigenic and
multipotent, capable of generating cells with all the different
lineages. One recently published study further characterized
these cells with additional neoangiogenic and cytoprotective
markers (11). These cells may be responsible for therapeutic
failure (12).
CD44 is a cell adhesion molecule known to be expressed in
most cell types (13, 14) and has been associated with stem cells
Human Cancer Biology
Authors’ Affiliations:
1
Depart ment of Pathology, Keck School of Medicine,
University of Southern California;
2
Joyce Eisenberg-Keefer Breast Center, John
Wayne Cancer Institute, Santa Monica, California;
3
Congressman Julian Dixon
Cellular Image Core, Childrens Hospital Los Angeles Saban Research Institute, Los
Angeles, C alifornia; and
4
Division of Cancer Immunothe rapeutics a nd Tumor
Immunology, City of Hope National Medical Center, Duarte, California
Received 1/25/06; revised 4/27/06; accepted 5/12/06.
Grant support: National Cancer Institute grants 5 R011 CA 585 40 and U10
CA76001and Doheny Eye InstituteSpecialized Imaging Core grant EY03040.
The costs of publication of this article were defrayed in part by the paymentof page
charges. This article must therefore be hereby marked advertisement in accordance
with18 U.S.C. Section 1734 solely to indicate this fact.
Requests for reprints: Richard J. Cote, Department of Pathology, University of
Southern California, and Norris Comprehensive Cancer Center, Keck School of
Medicine, 2011Zonal Avenue, Los Angeles, CA 90033. Phone: 323-865 -0212;
Fax: 323-865-0077; E-mail: cote_r
@
ccnt.hsc.usc.edu.
F2006 American Association for Cancer Research.
doi:10.1158/1078-0432.CCR-06-0169
www.aacrjournals.org Clin Cancer Res 200 6;12(19) October 1, 200 65615
Research.
on July 3, 2021. © 2006 American Association for Cancerclincancerres.aacrjournals.org Downloaded from
in normal breast tissue (15). CD24 is expressed in the early
stages of B-cell development and is highly expressed on neutro-
phils, but is absent in normal T cells or monocytes (16).
Whereas CD24 is not present in adult human tissues, it has
been shown to be expressed in human carcinomas (17, 18).
Although considered in one study as a prognostic marker in
breast cancer, where higher CD24 expression as seen by
immunohistochemistry was considered to favor the worse
prognosis (19), recent data suggest a different role for CD24 in
the carcinogenesis of breast cancer, such that decreased
expression or loss of CD24 seems to be the characteristic of
the stemness of a tumor cell (9, 11). CD24 reduces stromal
cellderived factor-1mediated migration and signaling via
CXCR4 in breast cancer cell lines with enhanced CD24
expression, suppressing their metastatic potential. On the
contrary, the metastatic potential of CD24
cells is increased
as evidenced by small interfering RNA inhibition (20). This role
of CD24 is also supported by in vivo analysis of nonobese
diabetic severe combined immunodeficient mouse models.
Taking into account the significance of DTC in breast cancer
(24), a reasonable next step is that we analyze the com-
partments known to be customarily associated with such
cells—the bone marrow and the lymph nodes—to identify the
potential cancer stem cells and characterize them. The
underlying hypothesis, therefore, is that cancer stem cells
comprise a subpopulation of DTC, and that immunohisto-
chemical identification of this subpopulation may provide a
tool to assess the malignant potential of specific cells and thus
may help to present the clinical course of the malignancy. In
the present study, we show for the first time an application in
bone marrow of a newly developed immunohistochemical
protocol to stratify DTC into subpopulations, and further
show that, surprisingly, even in the early-stage breast cancer
bone marrow samples studied, a vast majority of the DTC
match the phenotype criterion of putative cancer stem/
progenitor cells.
Materials and Methods
Patients and specimen collection
Bone marrow specimens were obtained from early breast cancer
patients, participating in the American College of Surgeons Oncology
Group Z-0010 clinical trial for detection and molecular characteriza-
tion of DTC. Following patient informed consent, unilateral bone
marrow aspirates from upper iliac crest were processed as described
below. For the present analysis, frozen cytospin slides from early
breast cancer patients previously categorized as CK
+
were used.
Because the analysis of patient samples is still ongoing, the study
remains blinded for clinical details and follow-up data are not yet
available. The study was approved by local Institutional Review
Boards.
Preparation of cultured cell slides
Breast cancer cell lines MCF-7, MDA-MB-231, SK-BR-3, HCC-38,
HCC-1395, and HCC-70 were purchased from American Type Culture
Collection (Manassas, VA). Cells were grown in appropriate growth
medium as suggested by American Type Culture Collection and
harvested by trypsinization before reaching confluence. The cells were
subjected to cytocentrifugation (at a density of f1,000/mm
2
) onto
positively charged slides by centrifugation at 1,000 rpm for 10 minutes.
Slides were air-dried overnight, followed by acetone fixation for
10 minutes, and stored at 20jC until use for immunohistochemical
analysis.
Bone marrow preparation
The procedure for processing the bone marrow aspirates has
previously been published (21). Briefly, the mononuclear cell
fraction containing any DTC was enriched by Ficoll-Hypaque density
gradient centrifugation, using Beckman GS-6 centrifuge, at 400 g
for 35 minutes and washed twice with PBS before transferring
the cells onto microscope slides by cytocentrifugation at a density
of 5 10
5
/mm
2
. Subsequently, the cells were fixed in acetone for
10 minutes and stored at 20jC.
Immunohistochemistry
Antibodies . A cocktail of two different mouse monoclonal
antibodies against CK was used as primary immunohistochemistry
detection reagent for circulating tumor cells: AE-1 (Signet, Dedham,
MA) against low and intermediate type I acidic keratins and
CAM5.2 (Becton Dickinson, San Jose, CA) against CK8 and CK18.
Further, mouse monoclonal primary antibodies anti-CD44 (clone
156-3C11, LabVision, Fremont, CA), binding to standard isoform
of CD44, and anti-CD24 (clone SN3b, LabVision) were used to
detect the putative breast cancer stem/progenitor cell markers
(10, 19).
Single-marker immunohistochemistry
Antibodies were titered until optimal concentration was deter-
mined by two independent trained personnel (M.B. and D.H.),
viewing under 40and 100objective lenses. The following
procedure was used for evaluating the expressions of the three
markers (CK, CD44, and CD24) on different cell lines and bone
marrow specimens. Slides were brought to room temperature before
blocking with normal horse serum for 30 minutes and incubated
with primary antibody diluted in blocking buffer for 1 hour.
Subsequently, the slides were washed thrice with PBS and incubated
with biotinylated secondary antibody (antimouse). After washing in
PBS thrice to remove the unbound secondary antibody, the immune
complexes were made visible by 30-minute incubation with avidin-
biotin-horseradish peroxidase complexes (Vector Laboratories, Bur-
lingame, CA) and 3,3-diaminobenzidine (DAB; Biogenex, San
Ramon, CA) as substrate. Cultured MCF7 and MDA-MB-231 were
used as positive controls for each run in single-, double-, and triple-
marker immunohistochemistry.
Double-marker immunohistochemistry
We optimized the double marker immunohistochemistry proce-
dure using slides with MDA-MB-231 cell line following the
completion of the single marker immunohistochemistry. The
optimized protocol is briefly described below. Slides were brought
to room temperature and incubated with normal horse serum
blocking solution for 30 minutes, with Universal block solution for
10 minutes, and with Tris-HCl for 5 minutes. Subsequently, the
slides were incubated with a cocktail of first primary antibodies for
2 hours (AE1 at 1:200 dilution and CAM5.2 at 1:100 dilution from
stock solution) following similar detection system with DAB as
described above in single-marker immunohistochemistry. The
second primary antibody was incubated either overnight (CD24
antibody at 1:100 dilution from stock solution) or with similar
procedure with a different detection system, alkaline phosphatase
with Vector Red as substrate (Vector Laboratories) and the addition
of a 30-minute blocking step of endogenous alkaline phosphatase
activity with 0.1 mol/L levamisole along with PBS with 5 mmol/L
levamisole for all the washes before the application of the avidin-
biotin alkaline phosphatase complexes. When the slides were
assessed for CK and CD44 staining, the procedure was slightly
modified. The slides were first incubated with anti-CD44 antibody
(at 1:300 dilution from stock solution) for 1 hour, followed by
detection using horseradish peroxidase/DAB system, and then with
the cocktail for anti-CK antibodies as described above, followed by
detection with alkaline phosphatase-Vector Red system.
Human Cancer Biology
www.aacrjournals.orgClin Cancer Res 20 06;12(19) October 1, 20 06 5616
Research.
on July 3, 2021. © 2006 American Association for Cancerclincancerres.aacrjournals.org Downloaded from
Triple-marker immunohistochemistry with immunoflu-
orescence
Additional CD44 assessment was done on the slides where the
immunodetection was already completed for CK and CD24. The slides
were first soaked in warm water for a few minutes to remove the
coverslips. After washing with PBS thrice, additional blocking was done
with normal horse serum for 30 minutes and incubated with primary
antibody for 2 hours (CD44 at dilution 1:200 from stock solution).
After washing in PBS thrice, Alexa Fluor 488 goat anti-mouse
immunoglobulin G (Molecular Probes, Invitrogen, Carlsbad, CA) was
added at 1:100 dilution from the stock solution and incubated for
1 hour. Following nuclear staining with 4,6-diamidino-2-phenylindole
(Vecatshield mounting medium containing 4,6-diamidino-2-phenyl-
indole), the slides were coverslipped and sealed with nail polish. The
protocol optimized for triple staining in cultured cell line slides was
used to analyze the clinical bone marrow samples.
DTC identification
All preparations were analyzed by two independent trained
personnel (M.B. and D.H.) using 40objective lens and 100
objective lens with oil immersion.
When slides were analyzed for CK and CD44 staining, CK was
represented by red cytoplasmic staining and CD44 by brown
membranous staining. When slides were assessed for CK and CD24,
cells stained for CK, which was brown cytoplasmic staining, and
CD24, which was membranous red staining, were classified as
CK
+
CD24
+
cells (see Fig. 1A), and cells dominant with only brown
cytoplasmic staining were classified as CK
+
CD24
(putative breast
cancer stem/progenitor cells; Fig. 1B). By changing the focal plane of
the objective, some cells previously considered negative for membrane
CD24 staining did show some membrane staining, and were
characterized as CK
+
CD24
+
.
Slides assessed for all the three antigens, CK, CD24, and CD44,
were first analyzed with light microscopy. CK
+
cells were marked and
subsequently examined for CD44 expression using a Leica DM LB2
microscope equipped with Diagnostic Instruments 7.3 3 shot color
camera viewed with a Chroma filter set consisting of an excitation
filter of 480/40 nm, dichroic filter of 505-nm long pass, and an
emission filter of 5,350/50 nm (Diagnostic Instruments, Sterling
Heights, MI).
Fig. 1. Aand B, double staining immunohistochemistry results for breast cancer
bone marrow samples. Thebone marrow samples show presence of distinct
stem-like and non-stem tumor cells (magnification, 40). A, two tumor cells with
brown cytoplasmic staining for CK (DAB) and red membranous staining for CD24
(Vector Red). B, an example of a CK
+
CD2 4
cell (putative breast cancer stem cell).
Cto F, the applicability of spectral imaging analysis for immunohistochemistry
double-stained samples, where the chromogens are not easily distinguishable with
the naked eye. Here, the cytospun bone marrowaspirates were assessed for CK
(red; stained with Fast Red) and CD44 (brown ; stained with DAB).The cells were
counterstained with hematoxylin. C, transmittedlight image, showing overall cell
population subjected to double-staining for CK and CD44. Arrow, two CK positive
cells that are difficult to assess for their CD44 positivity. Dand E, spectrally
unmixed images with pseudocolors assigned to hematoxylin (D)andDAB(CD44;
E) using SpectraView software, showing that all the cells in the field show nuclear
hema toxylin as well as CD4 4 staining, including the t wo cells d enoted by a rrow.
F, fluorescence image obtained for CK with Cy3 filter for Fast Red. Gto J,
applicability of spectral imaging analysis for immunohistochemistry double-stained
samples for CK and CD24. In this case, the expression of CK was assessed using
DAB (brown) and that of CD24 with Fast red (red ). The cells were counterstained
with hematoxylin. G, transmitted light image, showing overall cell population
subjected to double-staining for CK and CD24. Arrow, a CK-positive cell that
appears as CD24
/low
. To confirm that this c ell is indeed CD24
/low
,wedidfurther
spectralimaging analysis. Spectrally unmixed images with pseudocolors assigned
to hematoxylin (H)andDAB(CK;I). These images show that, whereas all cells in
the field show nuclear hematoxylin, only the cell denoted is positive for CK (DAB).
J, fluorescence image obtained for CD24 staining with Cy3 filter for Fast Red.
Spectral imaging analysis of the cell denoted in (G) shows that the cell at this
location expresses no/low CD24 (red). K, another CK-positive cell; L, the same c ell
with specific membranous fluorescence demarking the CD44 positivity; note the
surrounding CD44
+
lymphocy tes.
Stem Cells Are Majority of Disseminated Cancer Cells
www.aacrjournals.org Clin Cancer Res 200 6;12(19) October 1, 200 65617
Research.
on July 3, 2021. © 2006 American Association for Cancerclincancerres.aacrjournals.org Downloaded from
Spectral imaging
When it was not possible to distinguish with the naked eye if the cell
is single or double stained, spectral imaging was done. Images were
acquired at the CHLARI Congressman Julian Dixon Cellular Image Core
with a Leica DM RXA microscope using a HC Plan 20/0.70 numerical
aperture Ph2 circular differential interference contrast microscopy
objective lens, 1 optovar, and Koehler illumination (Leica Micro-
systems, Inc., Bannockburn, IL). A SKY/SD-300/VDS-1300 spectral
imager with Spectral Imaging 2.5 and SpectraView 1.1 software
(Applied Spectral Imaging, Inc., Carlsbad, CA) was used. Transmitted
light spectral images were acquired at maximum tungsten-halogen lamp
power (12 V) through a CB11 color balancing filter and ND4 neutral
density filter. Spectral image exposure time was 20 ms per frame to
avoid saturating the charge-coupled device detector. Spectral image
wavelength range was 400 to 750 nm. Vector Red fluorescence was
viewed through a Chroma Technology HQ Cy3 filter set (Rockingham,
VT) with a Sutter Instruments LS300/LLG380 Xenon arc lamp/1-m
liquid light guide (Novato, CA). DAB is not fluorescent and did not
attenuate the red fluorescence from these dyes. Fluorescence images
were acquired in SD-300/VDS-1300 direct view mode as 12-bit
monochrome images and saved as 16-bit TIFF images. Spectral color
images were saved from SpectraView and merged with monochrome
images using Adobe Photoshop 6.0 (San Jose, CA). Figure 1C to F
shows the applicability of spectral imaging analysis for immunohisto-
chemistry double-stained samples for analysis of CK
+
cell for CD44
expression, and Fig. 1Gto Jfor analysis of CK
+
cell for CD24
expression.
Result s
We evaluated bone marrow samples for the presence and
prevalence of putative cancer stem/progenitor cells. We
accomplished this by using pan-CK expression to first identify
tumor cells, followed by characterization of these DTC into
CD44
+
CD24
(cancer stem/progenitor cell subpopulation) and
CD44
+
CD24
+
(non-stem population). We first assessed six
different breast cancer cell lines for CD44, which were all found
to be immunohistochemistry positive. Furthermore, as proof of
principle, we stained 10 slides, selected from the 50 breast
cancer cases, for CK and CD44. In these 10 cases, we detected 22
CK
+
cells, and all of them were CD44
+
. In three cases where
CD44
+
reactivity was difficult to distinguish with the naked
eye, we did spectral imaging analysis to evaluate the cells for
the expression of CD44 (as shown in an example in Fig. 1C-F).
These observations, coupled with the evidence that most cells
(breast as well as hematopoietic; refs. 13, 14) and breast cancer
cell lines are CD44
+
, led us to reason that most of the breast
cancer DTC are both CK and CD44 positive. We employed the
double immunohistochemistry staining protocol optimized for
cell lines on cytospin preparations from bone marrow, which
were previously characterized as CK
+
.
Of 100 slides (from 50 clinical samples, 2 slides each), 2 were
excluded from the analysis due to poor sample quality. The
remaining 98 slides were evaluated for the presence and
prevalence of CK
+
CD24
+
or CK
+
CD24
cells.
Only 3 of 50 (6%) clinical bone marrow specimens have not
been confirmed for being positive on repeat immunohisto-
chemistry because no CK
+
cells could be detected in either slide.
This finding is attributable to sampling variability during
cytospin preparations, an acknowledged clinical reality. In all
47 (100%) clinical bone marrow specimens with CK
+
cells,
CK
+
CD24
phenotype was identified (Fig. 1B, G-J). The mean
percentage of cells with CK
+
CD24
phenotype detected in DTC
per clinical sample was 74.87% (range, 33-100%). The median
percentage of such cells per clinical sample was 71%. Table 1
provides the numbers of detected cells with either of the two
phenotypes and percentages of CK
+
CD24
cells in each bone
marrow specimen. On analyzing a total of 332 CK
+
cells, 232
(70.59%) cells showed CK
+
CD24
phenotype.
To further ascertain the prevalence of CD44 expression across
CK
+
cells, we have additionally done reevaluation of the bone
marrow specimens where possible. Of the 100 slides that were
assessed for CK and CD24, 71 slides underwent the additional
analysis. We used the cultured cell line MDA-MB-231 as
positive control, and the lymphocytes as internal positive
control, where the majority are positive for CD44. When
specific membranous fluorescence was undetectable in a slide,
it was regarded as insufficient immunohistochemical reaction.
Six of 71 slides showed insufficient staining for CD44. Two
hundred-eighteen cells could be identified in the remaining 65
slides as CK
+
. Of these, 19 (9%) were categorized as CD44
;the
rest of the cells (91%) showed specific positive membranous
reaction (see Fig. 1L). The CD44
cells were evenly distributed
between the CK
+
CD24
(4%) and CK
+
CD24
+
(5%) cell
populations. In none of the specimens could we identify only
CK
+
CD44
cells. From 199 CD44
+
cells, 141 (71%) were
CD24
and 58 (29%) were CD24
+
.CK
+
CD44
+
CD24
cells
represented 65% of the total pool of 218 cells, whereas
CK
+
CD44
+
CD24
+
cells represented 26%.
Discussion
Whereas identifying the rare tumor cells in a background
of numerous hematopoietic cells from bone marrow is itself
a labor-intensive task requiring much expertise, distinguishing
the tumor cells further between different subpopulations
(stem/non-stem) poses a new challenge. Using an optimized
immunohistochemistry procedure, we show here that the
majority of CK
+
cells (DTC) have the putative stem cell
phenotype (CD44
+
CD24
+
; refs. 9, 11). Further, we show here
for the first time the usefulness of spectral imaging for
analysis of single DTC and expression of additional markers.
Potential enhancement of the above assay can come through
application of newer techniques such as quantum dot
labeling, which would allow the labeling of cells with up
to six different markers and simultaneous analysis of their
expression.
To our surprise, all the patients with CK
+
DTC had cells with
putative breast cancer stem cell phenotype, and in fact, even
when the assessment for CD44 was completed on the sub-
stantial portion of the slides, the majority (71%) of DTC were
found to have a putative breast cancer stem cell like pheno-
type. The majority of DTC isolated from bone marrow have
previously been shown to be viable tumor cells with
proliferative potential under specific culturing conditions
(22); our results may be attributable to this finding because
the vast majority of the detected cells have a stem cell like
phenotype. Somewhat unexpected, the high percentage of CK
+
cells having CD44 expression in bone marrow may be
attributable to the previous findings in cell lines that, as an
adhesion factor, CD44 also represents a homing factor of breast
cancer cells to settle down in bone marrow (23).
Although DTC are regarded as the prerequisite for relapse
and metastasis (24), no studies have yet examined these cells
Human Cancer Biology
www.aacrjournals.orgClin Cancer Res 20 06;12(19) October 1, 20 06 5618
Research.
on July 3, 2021. © 2006 American Association for Cancerclincancerres.aacrjournals.org Downloaded from
for the existence of putative stem cell phenotype. Abraham
et al. (10) have recently analyzed primary breast tumors for
prevalence of CD44
+
CD24
/low
cells and their effect on
clinical outcome. They found that 70% of patients had
<10% of cells with CD44
+
CD24
/low
phenotype and that
higher prevalence of these cells correlated with distant
metastases, in particular, bone metastases. However, in their
study, the finding had no effect on overall survival of breast
cancer patients. Kristiansen et al. (19) considered higher CD24
expression as a prognostic marker in breast cancer to favor the
worse prognosis. Their immunohistochemistry data, however,
show differential expression of CD24 in tumor cells, with a
high number of CD24
+
cells, but cell nests can be recognized
with low or no expression of CD24
. These data can be
explained given the recent evidence that CD24
stem cells
give rise to both CD24
+
and CD24
cells (9) and that the
Table 1. Presence and distribution of putative stem cells across all the bone marrow specimens analyzed
Patient no. No. CK
+
CD24
cells No. CK
+
CD24
+
cells Total no. CK
+
cells % CK
+
CD24
phenotype
1 10 6 16 62.5
2 9 5 14 64.3
32 24 50
40 00
5 4 2 6 66.7
6 5 3 8 62.5
7628 75
87 29 77.8
96 28 75
10 3 0 3 100
11 11 1 12 91.6
12 5 6 11 45.5
13 5 1 6 83.3
14 7 4 11 63.6
15 9 3 12 75
16 9 5 14 64.2
17 3 2 5 60
18 6 3 9 66.7
19 11 5 16 68.8
20 6 2 8 75
21 2 0 2 100
22 1 2 3 33
23 7 2 9 77.8
24 1 0 1 100
25 7 5 11 58.3
26 14 6 20 70
27 1 0 1 100
28 2 0 2 100
29 2 0 2 100
30 7 2 9 77.8
31 2 0 2 100
32 4 3 7 57.1
33 5 3 8 62.5
34 0 0 0
35 5 3 8 62.5
36 6 0 6 100
37 8 3 11 72.7
38 7 3 10 70
39 1 0 1 100
40 4 0 4 100
41 3 0 3 100
42 4 2 6 66.7
43 5 2 7 71.4
44 3 0 3 100
45 4 1 5 80
46 3 2 5 60
47 5 2 7 71.4
48 0 0 0
49 1 1 3 50
50 2 2 4 50
Average of CK
+
CD24
phenotype/total DTC per patient 74.87
%CK
+
CD24
phenotype in overall total DTC 70.59 (240 of 340)
NOTE: The table provides the total number of CK
+
CD24
,CK
+
CD24
+
,andCK
+
cells, analyzed from two cytospun slides from bone marrow of
each patient. For each patient the percentage of CK
+
cells with CK
+
CD24
phenotype is presented along with the mean percentage per patient
and the overall percentage of all CK
+
cells with CK
+
CD24
phenotype.
Stem Cells Are Majority of Disseminated Cancer Cells
www.aacrjournals.org Clin Cancer Res 200 6;12(19) October 1, 200 65619
Research.
on July 3, 2021. © 2006 American Association for Cancerclincancerres.aacrjournals.org Downloaded from
larger proportion of cells in tumors is represented by CD24
+
cells. In addition, it is still not clear if the number of cancer
stem cells—or simply their presence—in the primary tumor is
critical for further disease development.
We are well aware of the fact that our present study as well as
previous studies has analyzed these cells for a predefined
phenotype, and that this is intended only as a preliminary
morphologic characterization of these cells. Whereas the
finding of DTC with putative stem/progenitor cell like
phenotype in majority of breast cancer patient bone marrow
is somewhat unexpected, it must be noted that this phenotypic
characterization needs to be supplemented with functional
analysis of these cells, which would inform on their capability
of self-renewal. Therefore, further molecular characterization
and biological analysis of the putative stem cells in prospec-
tively collected patient samples must be done. Nonetheless,
considering the present evidence, which suggests the effect of
such cells on the progression of breast cancer (9, 11, 25) and
other solid tumors (8, 24), we strongly believe that our assay
permits an initial identification of putative stem-like cell
subpopulation among the DTC, which can then permit further
molecular and functional characterization.
Using the assay we present here as a basic morphologic
assay, it should also be possible to better characterize DTC
for additional stem celllike markers. Some of the molecular
features of putative stem cells are emerging in the recent
literature (11, 26). Their data suggest that only a small
proportion of the CD44
+
CD24
/low
cells may have the self-
renewal capability. However, even if only a proportion of the
putative stem-like cells detected in our study have an ability
to perpetuate indefinitely, this finding indicates that at least
a proportion of the cancer cells detected in the bone marrow
of patients with early-stage breast cancer are capable of
forming metastases. Further, the majority of the DTC
detected in the bone marrow have the putative stem cell
phenotype, as compared with what has been reported in the
primary and metastatic tumor formations (where only a
minority of cells have this phenotype; refs. 9, 10), clearly
suggesting that there is a selection for these stem-like cells
and that DTC detected in the bone marrow of patients with
early-stage breast cancer are not merely shed from the
primary tumor.
Breast cancer patient population has a lifetime enhanced
risk of death from cancer when compared with similar normal
population group (27). This may be attributable to the
potential effect of the existence of cancer stem cells among the
DTC in patients. Recent pooled analysis by Braun et al. (3) in
a large number of patients analyzed shows that although the
presence of DTC is associated with a poor prognosis, a
significant proportion of patients still do well even after
10 years. The reason for this variability could be that a
proportion of these cells may be capable of remaining
‘‘dormant,’’ settled in bone marrow and/or distant organs,
which are driven towards the recurrent tumorigenesis only
after unknown initiation events occur in these cells. In
addition, besides their presence, the environment of DTC
may be as crucial in determining the destiny of the cells and
final patient outcome (24).
This is the first evidence of the existence of the putative
stem/progenitor cell (CD44
+
CD24
) subpopulation within the
DTC component in bone marrow. Because we could identify
the putative breast cancer stem cell phenotype in all patients
where we detected CK
+
cells, our data suggest that the majority
of the patients with DTC may have a lifetime risk for relapse,
an idea that has been suggested previously (28). Existence of
putative stem cells will be a clinically relevant issue that we
must address. Recognizing that it is desirable to correlate these
findings of existence of putative stem cells among the DTC
population with the actual clinical progression of breast cancer
patients, we will in the future examine the clinical correlates
for these patients once the clinical outcome data become
available from American College of Surgeons Oncology Group
clinical trial.
Whereas the possible presence of early tumor dissemination
is the rationale behind the use of systemic adjuvant chemo-
therapy in patients who have undergone definitive local
treatment of the primary tumor (29, 30), the presence of
cancer stem cells may explain the failure of adjuvant
chemotherapy in a proportion of early-stage breast cancer
patients. Further research aimed at enrichment and expression
profiling of CD44
+
CD24
population of DTC in a larger set of
prospectively collected bone marrow specimens can help
identify additional, novel potential therapeutic targets and
further define the biological potential of these cells.
Acknowledgments
We thankJustinTsai (LabVision, Fremont, CA) for the kind technical support.
Human Cancer Biology
www.aacrjournals.orgClin Cancer Res 20 06;12(19) October 1, 20 06 5620
Reference s
1. LugoTG, Braun S, Cote RJ, Pantel K, Rusch V.Detec-
tion and measurement of occult disease for the prog-
nosisofsolidtumors.JClinOncol2003;21:2609^15.
2. Cote RJ, Rosen PP, Lesser ML, et al. Prediction of
early relapse in patients with operable breast cancer
by detection ofoccult bone marrow micrometastases.
J Clin Oncol 1991;9:1749 ^ 56.
3. BraunS,VoglFD,NaumeB,etal.Apooledanalysis
of bone marrow micrometastasis in breast cancer.
N Engl J Med 20 05;35 3:793 ^ 80 2.
4. Coombes RC, Berger U, Mansi J, et al. Prognostic
significance of micrometastases in bone marrow in
patients with primary breast cancer. NCI Monogr
198 6;(1):51 ^ 3.
5. Al-Hajj M, Clarke MF. Self-renewal and solid tumor
stem cells. Oncogene 20 04;23:7274 ^ 82.
6. ReyaT, Morrison SJ, Clarke MF, Weissman IL. Stem
cells, cancer, and cancer stem cells. Nature 2001;414:
10 5 ^ 11.
7. Huntly BJ, Gilliland DG. Leuka emia stem cells and the
evolution of cancer-stem-cell research. Nat Rev Can-
cer 20 05;5:311 ^ 21.
8. Singh SK, Hawkins C, Clarke ID, et al. Identification of
human brain tumour initiating cells. Nature 2004;432:
396 ^ 401.
9. Al-Hajj M, Wicha MS, Benito-Hernandez A , Morrison
SJ, Clarke MF. Prospective identification of tumori-
genic breast cancer cells. Proc Natl Acad Sci U S A
2003;100:39 83 ^ 8.
10. Abraham BK, Fritz P, McClellan M, Hauptvogel P,
Athelogou M, Brauch H. Prevalence of CD44
+
/
CD24
/low
cells in breast cancer may not be associat-
ed with clinical outcome but may favor distant metas-
tasis. Clin Cancer Res 200 5;11:1154 ^ 9.
11. Ponti D, Costa A, Zaffaroni N, et al. Isolation and
in vitro propagation of tumorigenic breast cancer cells
with s tem/progenitor cell prop ertie s. Cancer Res
2005;65:5506 ^ 11.
12 . Al-HajjM,BeckerMW,WichaM,WeissmanI,Clarke
MF.Therapeutic implications of cancer stem cells. Curr
Opin Genet Dev 20 04; 14:4 3 ^ 7.
13 . Goldsby RA, KindtTJ, Osborne BA. CD antigens. In:
Kuby Immunology. 4th ed. New York: W.H. Freeman
and Company ; 200 0. p. 593 ^6 07.
14. Goodison S, UrquidiV,Tarin D. CD44 cell adhesion
molecules. Mol Pathol 1999;52:189^ 9 6.
15. Hebbard L, Steffen A, Zawadzki V, et al. CD44
expression and regulation during mammary gland
development and function. J Cell Sci 2 000; 113:
2619 ^ 30.
16. Aigner S, Sthoeger ZM, Fogel M, et al. CD24,
Research.
on July 3, 2021. © 2006 American Association for Cancerclincancerres.aacrjournals.org Downloaded from
a mucin-type glycoprotein, is a ligand for P-selectin
on human tumor cells. Blood 1997;89:3385^ 95.
17. Jackson D, Waibel R, Weber E , Bell J, Stahel RA.
CD24, a signal-transducing molecule expressed on
human B cells, is a major surface antigen on small
cell lung carcinomas. Cancer Res 1992;52:5264^ 70.
18. Kris tiansen G, Schluns K , Yongwei Y, Denkert C,
Dietel M, Petersen I. CD24 is an independent prog-
nostic marker of survival in nonsmall cell lung cancer
patients. Br J Cancer 20 03;88:231^ 6.
19. Kristiansen G,Winzer KJ, Mayordomo E, etal. CD24
expression is a new prognostic marker in breast can-
cer. Clin Cancer Res 2003;9:4906 ^ 13.
20. Schabath H, Runz S, Joumaa S, Altevogt P. CD24
affects CXCR4 function in pre-B lymphocytes and
breast carcinoma cells. JCell Sci 2006;119:314 ^25.
21. BauerKD,delaTorre-BuenoJ,DielIJ,etal.Reliable
and sensi tive analysis of occult bone marrow met asta-
ses using automated cellular imaging. Clin Cancer Res
2000;6:3552 ^ 9.
22. SolakogluO,MaierhoferC,LahrG,etal.Heteroge-
neous proliferative potential of occult metastatic cells
in bone marrow of patients with solid epithelial
tumors. Proc Natl Aca d Sci U S A 2002;9 9:224 6 ^51.
23. Draffin JE, McFarlane S, Hill A, Johnst on PG,
Waugh DJ. CD44 potentiates the adherence of meta-
staticprostate and breast cancer cells to bone marrow
endothelial cells. Cancer Res 2004;64:5702^ 11.
24. Brabletz T, Jung A, Spaderna S, Hlubek F, Kirchner
T. Opinion: migrating cancer stemcellsanintegrated
concept of malignant tumour progression. Nat Rev
Cancer 2005;5:744 ^ 9.
25. Dontu G, Al-Hajj M, Ab dallah WM, Clarke MF,
Wicha MS. Stem cells in normal breast development
and breast cancer. Cell Prolif 2003;36:59^ 72.
26. Glinsky GV. Death-from-cancer signature s and
stem cell contribution to metastatic cancer. Cell Cycle
2005;4:1171^ 5.
27. Brinkley D, Haybrittle JL. The curability of breast
cancer. Lancet 1975;2:95 ^ 7.
28. Dearnaley DP, OrmerodMG, Sloane JP, et al.
Micrometastases in breast cancer: long-term follow-
up of the firs t patient cohort. Eur J Cancer 1991;27:
236 ^ 9.
29. Pantel K, Brakenhoff RH. Dissecting the metastatic
cascade. Nat Rev Cancer 2004;4:448 ^56.
30.Schabel FM, Jr. Rationale for adjuvant chemothera-
py. Cancer1977;39:2875^ 82.
Stem Cells Are Majority of Disseminated Cancer Cells
www.aacrjournals.org Clin Cancer Res 200 6;12(19) October 1, 200 65621
Research.
on July 3, 2021. © 2006 American Association for Cancerclincancerres.aacrjournals.org Downloaded from
2006;12:5615-5621. Clin Cancer Res
Marija Balic, Henry Lin, Lillian Young, et al.
Cancer Stem Cell Phenotype
Marrow of Breast Cancer Patients Have a Putative Breast
Most Early Disseminated Cancer Cells Detected in Bone
Updated version
http://clincancerres.aacrjournals.org/content/12/19/5615
Access the most recent version of this article at:
Cited articles
http://clincancerres.aacrjournals.org/content/12/19/5615.full#ref-list-1
This article cites 28 articles, 14 of which you can access for free at:
Citing articles
http://clincancerres.aacrjournals.org/content/12/19/5615.full#related-urls
This article has been cited by 40 HighWire-hosted articles. Access the articles at:
E-mail alerts related to this article or journal.Sign up to receive free email-alerts
Subscriptions
Reprints and
.pubs@aacr.orgDepartment at
To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Permissions
Rightslink site.
(CCC)
Click on "Request Permissions" which will take you to the Copyright Clearance Center's
.http://clincancerres.aacrjournals.org/content/12/19/5615
To request permission to re-use all or part of this article, use this link
Research.
on July 3, 2021. © 2006 American Association for Cancerclincancerres.aacrjournals.org Downloaded from
... Extirpar el tejido necrótico y tumoral elimina los tejidos quimioresistentes, restaura la inmunocompetencia del huésped y reduce el crecimiento de las metástasis [24,25], y resulta en un aumento en la supervivencia de los pacientes [26]. Aunque, por otro lado, existe la hipótesis de que la cirugía en este grupo de pacientes puede estimular la progresión de la enfermedad por la mayor liberación de factores de crecimiento locales [27], estos factores de crecimiento a su vez pueden acelerar la proliferación de las células tumorales circulantes en sangre periférica y afectar la SG y la SLP [24,25,[28][29][30]. ...
... Esto explicaría el efecto benéfico reportado en SG y SLP en las pacientes que recibieron TC. Estudios retrospectivos mostraron que las pacientes operadas tenían mejores características pronósticas [30][31][32][33] y algunas habían presentado respuesta al tratamiento sistémico, estas pudieron ser entonces las causas de la mejor sobrevida y no el procedimiento quirúrgico [12]. Si bien en este estudio las pacientes llevadas a cirugía tenían una mayor carga tumoral, también se observó que al agrupar las indicaciones quirúrgicas la mayoría había presentado una respuesta completa o parcial a la terapia sistémica, lo que llevó a plantear la posibilidad de que el beneficio se asociara realmente a esta terapia. ...
Article
Full-text available
Introducción: El manejo sistémico es el pilar del tratamiento en las pacientes con cáncer de mama metastásico al debut. Sin embargo, la terapia conjunta (sistémica con cirugía local/locorregional) es objeto de investigación para determinar si ofrece un beneficio adicional en los resultados oncológicos. Los ensayos clínicos aleatorizados tienen reportes contradictorios en cuanto a supervivencia global, mientras que los estudios retrospectivos muestran un impacto favorable. Esta investigación tuvo como objetivo describir la supervivencia global y la supervivencia libre de progresión de pacientes con carcinoma de mama metastásico al debut, tratadas con terapia sistémica exclusiva o terapia conjunta. Materiales y método: Estudio retrospectivo de una cohorte de pacientes con carcinoma de mama metastásico al debut de una clínica de referencia oncológica. Se evaluaron dos grupos de manejo: con terapia sistémica exclusiva vs. terapia conjunta. Los resultados principales evaluados fueron la supervivencia libre de progresión y la supervivencia global, calculados mediante las funciones de supervivencia de Kaplan-Meier y ajustados a las variables confusoras con modelos de Cox. Resultados: Recibieron terapia sistémica exclusiva 174 pacientes y 88 pacientes, terapia conjunta. La mediana de seguimiento fue de 58,38 meses; la supervivencia libre de progresión fue de 38,56 meses en el grupo de terapia sistémica exclusiva vs. 72,25 meses para el grupo de terapia conjunta. La supervivencia global fue de 42,4 meses (IC 95 % 33,23-51,56) en terapia sistémica exclusiva vs. 82,33 (IC 95 % 62,1-102,55) en terapia conjunta, ambos resultados estadísticamente significativos para el grupo quirúrgico. Conclusión: En pacientes con carcinoma de mama metastásico al debut, la supervivencia global y la supervivencia libre de progresión fueron mejores en los tratados con terapia conjunta que en los manejados con terapia sistémica exclusiva.
... Interestingly, apart from CTCs, other cellular subsets like disseminated tumor cells were also found to display stem cell characteristics. According to certain reports, disseminated tumor cells found within the bone marrow of cancer patients depicted strong cancer stem cell characteristics, this is indeed indicative of the strong potential of CCSCs to initiate micrometastasis in the bone marrow (Balic et al., 2006;Giordano et al., 2012). Interestingly, although micrometastases may be driven by CCSCs, hence portraying stem-cell like characteristics, macrometastases is found to possess a close resemblance to the primary tumor cells. ...
Chapter
Full-text available
Circulating Cancer Stem Cells (CCSCs) constitute a distinctive category of cancer cells that play pivotal roles in shaping multiple facets of the cancer continuum, spanning from the initiation and progression of oncogenesis and tumorigenesis to the intricate processes of metastasis and cancer relapse. Demonstrating a unique intermediate phenotype, these specialized cells exhibit characteristics that lie at the crossroads between epithelial and mesenchymal phenotypes. CCSCs actively represent a rare population in the blood circulation, where their predictive and prognostic potential has often been harnessed to gain valuable insights into the diagnosis, metastatic behavior, and recurrence patterns of cancer. Moreover, CCSCs serve as critical indicators of a patient’s response to treatment, resistance mechanisms, and overall clinical outcomes. This comprehensive exploration delves into various intriguing aspects related to CCSCs, providing an in-depth analysis of their roles and implications. The work extensively discusses the significance of CCSCs in the intricate landscape of cancer biology, shedding light on their unique attributes and contributions to disease progression. Emphasis is placed on unraveling the complexities surrounding CCSCs, offering a thorough and unbiased overview of the challenges associated with the analysis of these specialized cells. In addition, a noteworthy focus of this work is directed towards the escalating importance of CCSC-targeted cancer therapies. The implications stemming from existing research reports are thoroughly examined, presenting a nuanced perspective on the current state of CCSC-targeted therapeutic approaches. Furthermore, the inherent challenges in these therapeutic techniques, have also been discussed, with an effort to offer insights into future prospects and potential advancements in the field. Overall, this work aims to contribute to the evolving landscape of cancer research by providing a comprehensive exploration of CCSCs and their potential as targets for innovative therapeutic strategies.
... CTSK has been shown to promote invasion and metastasis of breast cancer through direct degradation of the extracellular matrix (ECM) and promotion of angiogenesis or enhancing paracrine interactions [20,21]. High expression of CTSK enhances the invasiveness of prostate cancer cells and provides anchor points for cancer cells, playing a key role in lymph node metastasis and bone metastasis in prostate cancer [22][23][24]. CTSK expression could be detected in lung tissues such as bronchial and alveolar epithelium and alveolar macrophages [25], however, CTSK was significantly elevated in TAM in advanced NSCLC [26], which may be related to the fact that CTSK could promote the proliferation of A549 cells by directly activating the mTOR signaling pathway, thereby promoting the progression of NSCLC [27]. In colorectal cancer (CRC), CTSK can act as a mediator of intestinal microbiota imbalance leading to metastasis of CRC [28]. ...
Article
Full-text available
Background Cathepsin-K (CTSK) is overexpressed in Gastric cancer (GC) and the mechanism of its overexpression in GC is still unclear. The present work found CTSK as a potential predictive biomarker and immunotherapeutic target for GC based on the tumor microenvironment (TME). Methods From public databases, gene expression profiles and clinical data of GC were downloaded to analyze the distribution of stromal and immune cells and tumor abundance in TME. Differentially expressed genes (DEGs) associated with TME were obtained by differential analysis, followed by cross-screening to obtain CTSK as a gene associated with TME. Next, a series of methods and tools were employed to explore the relationships between clinicopathological features of GC and CTSK expression as well as prognosis, tumor immune microenvironment, immune checkpoints and drug sensitivity. And GSEA was used to investigate the potential role of CTSK in the tumor microenvironment of GC. Results From the dataset, we obtained a total of 656 DEGs associated with TME and the stromal component of TME was found to be closely involved in GC prognosis. CTSK was cross-screened as the key gene associated with TME by the PPI network and univariate Cox regression analysis. Pan-cancer analysis revealed significant high expression of CTSK in a variety of cancers. Subsequently, we hypothesized that high-expressed CTSK was closely correlated with poor prognosis and lymph node metastasis of tumors, and that CTSK, a GC TME-related gene, was largely involved in a range of biological behaviors of tumors, with a significant correlation between several immune cells. Conclusion CTSK was validated as a potential prognostic biomarker related to TME of GC and could be a promising next-generation immunotherapeutic target for GC. Supplementary Information The online version contains supplementary material available at 10.1007/s12672-023-00821-0.
... However, not all patients with detactable DTCs will suffer from relapse, supporting the hypothesis of metastatic inefficiency of these cells [27]. Therefore, efforts have been made to further characterize the metastatic potential of DTCs and their role as a predictive therapeutic marker [30][31][32][33]. ...
Article
Full-text available
Purpose Androgen receptor (AR) can serve as a new therapeutic target since it was shown to play a proliferative role in several breast cancer (BC) subtypes. Moreover, AR positivity has been suggested to reflect the metastatic potential of tumor cells in some BC subtypes. The aim of this study was to determine the AR expression on disseminated tumor cells (DTCs) as a surrogate marker of minimal residual disease (MRD) and potential precursor of metastasis in early BC. Methods Bone marrow (BM) aspirates from 62 DTC-positive early BC patients were included into this study and analyzed by immunofluorescence staining for the presence of AR-positive DTCs. CK-positive, CD45-negative cells containing an intact nucleus (DAPI positive) were identified as DTCs. AR expression of the primary tumor (PT) was assessed by immunohistochemistry on formalin-fixed, paraffin-embedded (FFPE) tumor sections from core biopsies and surgical specimens. Results AR status of DTCs could be determined in 21 patients. We detected AR-positive DTCs in nine samples (43%). AR expression of DTCs and corresponding PT showed a concordance rate of 33%. The DTC-AR status did not correlate with clinicopathological factors, nor did we observe a significant correlation between the AR status of the PT and other established prognostic factors for BC. Conclusion AR-positive DTCs can be detected in BM of early BC patients with a marked discordance of the AR status between DTCs and corresponding PTs. The clinical significance of these findings needs further investigation.
... Metastases start from single cells or cell clusters that separate from the original tumor and migrate to distant areas. In most cases of BC, the dissemination of tumor cells to other tissues occurs early on, concomitant with the progression of the primary tumor [87,88]. Despite this, it is possible for DTCs to remain inactive for many years or even decades before visible metastases emerge. ...
Article
Full-text available
This paper is a cross fertilization of ideas about the importance of molecular aspects of breast cancer metastasis by basic scientists, a pathologist, and clinical oncologists at the Henry Ford Health symposium. We address four major topics: (i) the complex roles of lymphatic endothelial cells and the molecules that stimulate them to enhance lymph node and systemic metastasis and influence the anti-tumor immunity that might inhibit metastasis; (ii) the interaction of molecules and cells when breast cancer spreads to bone, and how bone metastases may themselves spread to internal viscera; (iii) how molecular expression and morphologic subtypes of breast cancer assist clinicians in determining which patients to treat with more or less aggressive therapies; (iv) how the outcomes of patients with oligometastases in breast cancer are different from those with multiple metastases and how that could justify the aggressive treatment of these patients with the hope of cure.
Article
Cancer metastasis is a biologically complex process that remains a major challenge in the oncology clinic, accounting for nearly all of the mortality associated with malignant neoplasms. To establish metastatic growths, carcinoma cells must disseminate from the primary tumour, survive in unfamiliar tissue microenvironments, re-activate programs of proliferation, and escape innate and adaptive immunosurveillance. The entire process is extremely inefficient and can occur over protracted timescales, yielding only a vanishingly small number of carcinoma cells that are able to complete all of the required steps. Here we review both the cancer-cell-intrinsic mechanisms and microenvironmental interactions that enable metastatic colonization. In particular, we highlight recent work on the behaviour of already-disseminated tumour cells, since meaningful progress in treating metastatic disease will clearly require a better understanding of the cells that spawn metastases, which generally have disseminated by the time of initial diagnosis.
Chapter
3d metal-based chemotherapeutic agents are considered as the promising viable options for the treatment of solid malignancies as compared to cisplatin which exhibited high systemic toxicity resulting in adverse reactions and further were found inactive against many resistant cancer cells and secondary metastatic conditions. Since these metal ions have a tendency to coordinate to diverse ligand scaffolds, a huge number of structurally well-characterized complexes that could act as potent anticancer agents have emerged, wherein the ligand imparts a distinct functionality viz., facilitating uptake in cells, solubility in water, bioisosteric replacements, and directing to the specific pathway of cell death apoptosis vs necrosis, binding to a specific domain at the target sites, overcoming considerably the shortcomings of platinate class of anticancer drugs. The potential chemotherapeutic agents utilizing metals for anticancer treatment have been categorized according to the structure of the ligand framework incorporated in the metal complexes as given below.
Article
Full-text available
Stem cell biology has come of age. Unequivocal proof that stem cells exist in the haematopoietic system has given way to the prospective isolation of several tissue-specific stem and progenitor cells, the initial delineation of their properties and expressed genetic programmes, and the beginnings of their utility in regenerative medicine. Perhaps the most important and useful property of stem cells is that of self-renewal. Through this property, striking parallels can be found between stem cells and cancer cells: tumours may often originate from the transformation of normal stem cells, similar signalling pathways may regulate self-renewal in stem cells and cancer cells, and cancer cells may include 'cancer stem cells' — rare cells with indefinite potential for self-renewal that drive tumorigenesis.
Article
Full-text available
The main focus of this review is the role of mammary stem cells in normal breast development and carcinogenesis. We have developed a new in vitro culture system that permits, for the first time, the propagation of mammary stem and progenitor cells in an undifferentiated state, which should facilitate the elucidation of pathways that regulate normal mammary stem-cell self-renewal and differentiation. Furthermore, we propose a model in which transformation of stem cells, or early progenitor cells, results in carcinogenesis. A key event in this process is the deregulation of normal self-renewal in these cells. Transformed mammary stem or progenitor cells undergo aberrant differentiation processes that result in generation of the phenotypic heterogeneity found in human and rodent breast cancers. This phenotypic diversity is driven by a small subset of mammary tumour stem cells. We will discuss the important implications of this mammary tumour stem-cell model.
Article
Full-text available
Breast cancer is the most common malignancy in United States women, accounting for >40,000 deaths each year. These breast tumors are comprised of phenotypically diverse populations of breast cancer cells. Using a model in which human breast cancer cells were grown in immunocompromised mice, we found that only a minority of breast cancer cells had the ability to form new tumors. We were able to distinguish the tumorigenic (tumor initiating) from the nontumorigenic cancer cells based on cell surface marker expression. We prospectively identified and isolated the tumorigenic cells as CD44(+)CD24(-/low)Lineage(-) in eight of nine patients. As few as 100 cells with this phenotype were able to form tumors in mice, whereas tens of thousands of cells with alternate phenotypes failed to form tumors. The tumorigenic subpopulation could be serially passaged: each time cells within this population generated new tumors containing additional CD44(+)CD24(-/low)Lineage(-) tumorigenic cells as well as the phenotypicall
Article
Purpose: Breast cancer is composed of phenotypically diverse populations of cancer cells. The ability to form breast tumors has been shown by in vitro/in vivo studies to be restricted to epithelial tumor cells with CD44+/CD24−/low characteristics. Validation of these findings with respect to detection in clinical samples, prognosis, and clinical relevance is in demand. Experimental Design: We investigated breast cancer tissues for the prevalence of CD44+/CD24−/low tumor cells and their prognostic value. The study included paraffin-embedded tissues of 136 patients with and without recurrences. In addition, a breast cancer progression array with normal, carcinoma in situ, and carcinoma tissues was analyzed. We applied double-staining immunohistochemistry for the detection of CD44+/CD24−/low cells. Evaluation was by microscopic pathologic inspection and automated image analysis. Results: CD44+/CD24−/low cells ranged from 0% to 40% in normal breast and from 0% to 80% in breast tumor tissues. The prevalence of CD44+/CD24−/low tumor cells in 122 tumors was ≤10% in the majority (78%) of cases and >10% in the remainder. There was no significant correlation between CD44+/CD24−/low tumor cell prevalence and tumor progression. Although recurrences of tumors with high percentages of CD44+/CD24−/low tumor cells were mainly distant, preferably osseous metastasis, there was no correlation with the event-free and overall survival. There was no influence on the response to different treatment modalities. Conclusions: Our findings suggest that the prevalence of CD44+/CD24−/low tumor cells in breast cancer may not be associated with clinical outcome and survival but may favor distant metastasis.
Article
Results are presented of a long-term follow-up of patients with breast cancer seen in the Cambridge area from 1947 to 1950. After 21 years the survival curve of the breast-cancer patients runs parallel to the expected survival curve of a similar normal population. This suggests that 18% of all the breast-cancer patients may be regarded as cured of their disease in the sense defined by Easson and Russell. An analysis of the causes of death in the study group after 20 years' follow-up shows an excess of deaths from breast cancer. The number is 16 times more than would be expected in the normal population, although the overall death-rates in the two groups are the same. This questions the concept of cure.
Article
Surgery or radiation therapy fails to cure clinically evident human cancer, in the main, if the disease is systemic (has metastasized) when first recognized, because neither modality can effectively remove or kill distant and/or unrecognized metastases. Starting drug treatment of a tumor when it is first clinically recognized, or when it re-presents following noncurative surgical or radiological treatment, fails to cure over 90% of cancer in man because the body burden of tumor cells exceeds the tumor cell kill potential of nearly all of the most effective drugs or drug combinations against most tumors. The indicated approach to improving cure rates under these circumstances is to use chemotherapy as an adjuvant to effective (life-prolonging) but noncurative surgical and/or radiological treatment. Surgical adjuvant chemotherapy of carcinoma of the breast and osteogenic sarcoma has already been shown to increase the disease-free interval over that obtained with surgery alone, and the probability of having achieved significant increases in long-term cure rates is high. Laboratory studies with transplantable metastatic lung, breast, and colon carcinomas and melanotic melanoma, and with a spontaneous breast carcinoma of mice, all of which are uniformly fatal if untreated, have shown that: 1) the incidence of metastatic disease is directly related to tumor mass, 2) surgical cure rates drop as tumor mass at surgery increases, 3) grossly evident primary tumors are generally not curable by drug treatment, and 4) surgical adjuvant chemotherapy increases the long-term cure rates with all of these tumors and significantly increases the life span of treatment failures. Effective surgical adjuvant chemotherapy is both dose-responsive and related to the body burden of metastatic tumor at time of drug treatment. The effectiveness of surgical adjuvant chemotherapy decreases 1) as the tumor staging is advanced prior to surgery, 2) as the interval from surgery to start of effective chemotherapy is increased, and 3) as the drug doses are reduced. Additionally, and of critical importance to treatment planning, some drugs that are marginally effective or ineffective against the presurgical total body burden of tumor cells are curative in some to all mice with metastatic disease if given shortly after surgical removal of the primary tumor.
Article
Cell lines derived from human small cell carcinoma of the lung express high levels of a surface polypeptide termed the cluster-w4 antigen, which was previously identified as a potential target for toxin-based immunotherapy of lung cancer. We have cloned a complementary DNA encoding the cluster-w4 antigen from COS-1 fibroblasts transfected with a SW2 small cell carcinoma library, by panning with a mixture of the cluster-w4-specific monoclonal antibodies SWA11, SWA21, and SWA22. The sequence of the cluster-w4 complementary DNA encodes an unusually short (80-amino acid) protein identical to that recently reported for the leukocyte activation molecule CD24 except for a single valine-alanine substitution due to a single-base polymorphism within the region of the gene coding for the extracellular domain. Biochemical analyses of the cloned cluster-w4 antigen confirmed both the presence of the phosphatidylinositol tail and the extensive glycosylation reported for the CD24 molecule. Furthermore, the cloned cluster-w4 antigen expressed on COS cells was shown to react with a comprehensive panel of CD24-specific monoclonal antibodies, as assessed by indirect immunofluorescence staining. Northern blot hybridization indicated the presence of several transcript sizes for the cluster-w4 antigen that were greatly overexpressed in small cell carcinoma cell lines, compared with normal hemopoietic cells and CD24-positive cell lines. Southern blot hybridization of restriction digests of genomic DNA identified a complex pattern of bands consistent with either a complex gene structure containing many exons or the presence of a family of closely related genes.
Article
"Micrometastases" can be identified in the bone marrow of patients with apparently localised breast cancer using an immunocytochemical stain for epithelial membrane antigen (EMA). Of 39 women who had marrow samples examined at the time of initial presentation (37), or with locally recurrent disease (2), 13 (33%) had samples which contained small numbers of EMA positive cells. 10 out of 23 (44%) lymph-node positive patients were marrow positive, compared to 1 out of 14 (7%) lymph node negative cases (P = 0.03). Long-term follow-up (median 9.5 years) has shown that 11 out of 13 (85%) patients with micrometastases have developed metastatic disease compared to 8 out of 26 (31%) with negative bone marrow aspirates (P less than 0.05). The small number of EMA positive cells detected in bone marrow samples probably reflects the high metastatic potential of primary or recurrent cancers rather than established microscopic deposits; it is not yet clear whether the finding of such micrometastases will act as an independent variable compared to established prognostic factors.
Article
We used monoclonal antibodies to identify occult micrometastases in the bone marrow of 49 patients with operable (stage I and II) breast carcinoma. Follow-up (mean, 29 months; median, 30 months) revealed that 12 patients recurred. The presence of bone marrow micrometastases (BMM) was significantly associated with early recurrence (P less than .04). The estimated 2-year recurrence rate for patients with no BMM detected (BMM-) was 3%; in patients with BMM, the 2-year recurrence rate was 33%. When BMM and axillary lymph node (LN) status were combined, groups of patients at low risk (LN-, BMM-; 2-year recurrence rate, 0%) and high risk (LN+, BMM+; 2-year recurrence rate, 42%) for early recurrence were identified. Bone marrow tumor burden was related to early recurrence. Among patients with BMM, those who did not recur had on average fewer extrinsic cells in their marrow than those who recurred (15 v 43 cells, respectively). Multivariate analysis comparing BMM, LN+ versus LN-, and tumor size (less than or equal to 2 cm v greater than 2 cm) revealed no factor independently associated with early recurrence. Peripheral tumor burden of BMM (0 or less than 10 extrinsic cells v greater than or equal to 10 extrinsic cells) was the only independent predictor of early recurrence (P less than .003). In conjunction with conventional prognostic factors, particularly axillary LN status, evaluation for BMM might be used to stratify patients for adjuvant treatment programs. Because this pilot study involved few patients with short-term follow-up, the results should be interpreted with caution. The examination of bone marrow for micrometastases remains an experimental procedure; the clinical usefulness of the test will be established through larger studies with long-term follow-up.