You are on page 1of 8

Review Cancer

Prevention
Research
Heterogeneity in Colorectal Cancer Stem Cells
Akihiro Hirata1, Yuichiro Hatano2, Masayuki Niwa3, Akira Hara2, and
Hiroyuki Tomita2

Abstract
Cancer stem cells (CSC) have attracted a great deal of CSCs no longer should be viewed as a fixed target
interest for their clinical relevance in a range of cancers, population, and we should note that their heteroge-
including colorectal cancer. CSCs were initially consid- neous and dynamic nature presents a serious problem
ered to be cell populations with homogeneous, well- for the development and implementation of specific

Downloaded from http://aacrjournals.org/cancerpreventionresearch/article-pdf/12/7/413/2244191/413.pdf by guest on 28 March 2022


defined phenotypic and molecular characteristics. How- therapeutic strategies. This review summarizes past and
ever, accumulating evidence suggests that CSCs repre- current literature related to the heterogeneity and
sent phenotypically and functionally heterogeneous dynamics of colorectal CSC populations, focusing on
populations. Recent studies demonstrate colorectal evidence for distinct subpopulations, and signaling
CSCs to be dynamic rather than static, and continuously pathways, and intra- and extratumoral factors involved
altered by multiple extrinsic and intrinsic factors. Thus, in their regulation in cancer tissues.

Introduction teristics with their corresponding normal tissue-resident


stem cells, and intestinal stem cells (ISC) play a crucial
It has long been recognized that malignant cells
role in maintenance of homeostatic epithelial renewals
within a single tumor display significant heterogeneity
in the normal colonic tissues (9), colorectal cancer pro-
in morphology, proliferative activity, and function (1).
vides a compelling model of a hierarchically organized
The cancer stem cell (CSC) concept provides a con-
solid tumor, with CSCs at the apex. Furthermore, data
vincing explanation for the mechanisms underlying
from ISCs should offer clues to understanding CSCs in
this heterogeneity. CSCs, also called tumor-initiating
colorectal cancer.
cells, are defined by their capacity to self-renew and
In this review, we summarize the past and current
generate diverse cells that comprise the tumor (2). They
evidence related to heterogeneity within colorectal CSC
are thought to initiate and continually sustain tumor
populations and our understanding of the potential
growth.
mechanisms underlying this heterogeneity.
The first evidence for the existence of CSCs came from
studies on acute myelogenous leukemia in the 1990s, in
which a rare subset of leukemic cells with a CD34þ/ Heterogeneity within Colorectal CSC
CD38 phenotype was shown to induce leukemia in
Populations
immunocompromised mice (3, 4). A decade following
the identification of leukemic stem cells, the discovery of Markers for colorectal CSCs
a CSC subset in breast cancers (5) expanded the concept Since the initial publication on brain tumors (10),
to solid tumors and thereafter CSCs were identified in CD133 (also known as prominin-1) has been established
multiple solid tumors, including colorectal cancer (6–8). as a marker for CSC populations in a variety of cancers.
Because CSCs share phenotypic and molecular charac- Human colorectal CSCs were first identified by CD133
expression (7, 8). CD133þ cells were consistently capable
of generating tumors that resembled the original patient
1
Division of Animal Experiment, Life Science Research Center, Gifu University, tumor when serially transplanted in immunocompro-
Gifu, Japan. 2Department of Tumor Pathology, Gifu University Graduate mised mice, while their CD133 counterparts failed to
School of Medicine, Gifu, Japan. 3Medical Science Division, United Graduate
School of Drug Discovery and Medical Information Sciences, Gifu University, give rise to xenografts. To date, other markers have been
Gifu, Japan. reported to identify CSCs in human colorectal cancers
Corresponding Author: Hiroyuki Tomita, 1-1 Yanagido, Gifu City, Gifu 501-1194, (Table 1), including CD44 (6), CD44v6 (11), and alde-
Japan. Phone: 81-58-230-6225; Fax: 81-58-230-6226; E-mail: hyde dehydrogenase 1 (ALDH1) (12).
h_tomita@gifu-u.ac.jp
Cancer Prev Res 2019;12:413–20 Overlap and nonoverlap among CSC markers
doi: 10.1158/1940-6207.CAPR-18-0482 Flow-cytometric analysis has revealed overlaps between
2019 American Association for Cancer Research. populations of CD133þ colorectal cancer cells and

www.aacrjournals.org 413
Hirata et al.

Table 1. Markers for colorectal cancer stem cells


Samples from which CSC candidates Methods used to demonstrate cancer
Phenotype were isolated initiating and/or metastatic capacitya Reference
CSC markers
CD133 Human CRC tissues (primary lesions) Xenotransplantation (SCID mouse, subcutaneous 8
transplantation)
Human CRC tissues (primary or metastatic Xenotransplantation (NOD-SCID mouse, renal capsule 7
liver lesions) transplantation)
CD44 (EpCAMhigh/CD44þ) Human CRC xenografts Xenotransplantation (NOD-SCID mouse, subcutaneous 6
transplantation)
high þ
CD166 (EpCAM /CD166 ) Human CRC xenografts Xenotransplantation (NOD-SCID mouse, subcutaneous 6
transplantation)
ALDH1 Human CRC tissues (primary lesions) Xenotransplantation (NOD-SCID mouse, subcutaneous 12
transplantation)
Human CRC xenografts
CD44v6 Human CRC tissues Xenotransplantation (NOD-SCID mouse, orthotopic 11
transplantation)
Human CRC sphere
CD26 (CD26þ/CD133/CD44)

Downloaded from http://aacrjournals.org/cancerpreventionresearch/article-pdf/12/7/413/2244191/413.pdf by guest on 28 March 2022


Human CRC tissues (primary lesions) Xenotransplantation (SCID mouse, subcutaneous and 21
orthotopic transplantation)
LGR5 Spheroids derived from human CRC tissues Xenotransplantation (nude mouse, subcutaneous 27
(primary lesions) transplantation)
Organoids derived from human CRC tissues Lineage tracing in human CRC orgaoids xenografted in 28
NOG mouse (renal capsule transplantation)b
CSC with metastatic capacity
CD133þ/CXCR4þ Human CRC cell line (HCT116) Xenotransplantation (nude mouse, tail-vein injection) 20
CD26þ (CD26þ/CD133þ or /CD44þ or ) Human CRC tissues (primary lesions of a Xenotransplantation (SCID mouse, orthotopic 21
patient with hepatic metastasis) transplantation)
CD44v6 Human CRC tissues Xenotransplantation (NOD-SCID mouse, orthotopic 11
transplantation)
Human CRC spheres
CSC with organ-specific metastatic potential
CD133þ/CD110 Human CRC tissues (primary lesions of Xenotransplantation (NOG mouse, orthotopic 25
CD133þ/CDCP1 patients with CRC with liver and lung transplantation)
metastases)
Abbreviations: ALDH1, aldehyde dehydrogenase 1; CRC, colorectal cancer; CSC, cancer stem cell; SCID, severe combined immunodeficient; NOD-SCID, nonobese
diabetic; NOG, Nod/Shi-SCID,IL-2Rg null.
a
Only the most reliable/conventional assays are listed for each study, although multiple assays were used to assess CSC properties in many studies.
b
CSC functions in tumor tissues, rather than cancer-initiating capacity, were examined in the xenografts.

populations expressing CD44, CD29, CD24, and CD166, CD133þ/CD44þ cells induced tumors in immunocom-
all of which have been described as enriched for colorectal promised mice under conditions in which the same num-
CSCs (13). This finding suggests that CD133 expression is ber of CD133þ/CD44 cells failed to engraft, indicating
the most broadly distributed marker for colorectal CSCs. that CD44 provides further enrichment of CSCs in the
However, as has been reported in human glioblasto- CD133þ subset (18).
mas (14–16), colorectal cancer cells with CSC properties
do not necessarily express CD133. It has been shown CSCs with metastatic capacity
that both CD133þ and CD133 subsets from hepatic Distant metastasis is the primary cause of lethality in
metastases of colorectal tumors are capable of reconstitut- patients with colorectal cancer. Previous studies have
ing tumors when subcutaneously injected into immuno- suggested that only certain subsets of CSCs are capable
compromised mice; moreover, tumors derived from of distant metastasis (Table 1). Experiments showing
CD133 cells grew at a more rapid rate (17). Further differences in metastatic capacity between CSC subsets
analysis revealed that CD133 cells frequently expressed are the most convincing demonstrations of functional
CD44, another CSC marker (6), in colonospheres heterogeneity.
derived from CD133 cells (17). The following finding Hermann and colleagues were the first to identify a
is also indicative of the existence of CSCs phenotypically specific subset of CSCs responsible for metastasis in human
distinct from CD133þ cells in colorectal tumors: CD44þ/ pancreatic cancer cell lines, based on the cell-surface
EpCAMhigh cells with CSC properties were found even in expression of CD133 and CXCR4, a receptor for the che-
colorectal tumors in which CD133þ cells were not mokine CXCL12 (19). When orthotopically injected into
contained (6). nude mice, both CD133þ/CXCR4þ and CD133þ/CXCR4
Complicating matters further, it has been shown that cells could produce tumors at the injection site, but only
CD133 and CD44 can be nonmutually exclusive markers, CD133þ/CXCR4þ cells produced liver metastases (19). A
as a partial overlap between the two cell subsets in colo- subsequent study showed that the colorectal cancer cell
rectal cancers has been repeatedly reported (6, 13, 18). line HCT116 also contains CD133þ/CXCR4þ cells, which

414 Cancer Prev Res; 12(7) July 2019 Cancer Prevention Research
Colorectal Cancer Stem Cells

have a significantly higher metastatic capacity in nude mice static CSC subsets can be contained within CD133þ CSC
than CD133þ/CXCR4 cells (20). populations.
Pang and colleagues demonstrated that a subpopula-
tion of colorectal CSCs expressing CD26 has both tumor-
initiating and metastatic capacities (21). Orthotopic
Dynamics of Colorectal CSCs
implantation into mice of CD133þ/CD26þ cells isolated Recent studies demonstrate colorectal CSCs to be
from the primary tumor of a colorectal cancer patient dynamic rather than static populations that are continu-
with hepatic metastasis produced liver metastases follow- ously altered by multiple factors including genetic and
ing tumor formation in the cecal wall, while their CD26 epigenetic alterations, interactions between tumor cells,
counterparts induced tumor growth only at the site microenvironmental factors, hormone action, and cancer
of injection (21). They also showed that circulating therapy. Any or all of these factors could contribute to
CD133þ/CD26þ/CD44þ cells could be detected in port- producing heterogeneity in colorectal CSC populations.
al blood after cecal-wall injection and that intraportal This section summarizes past and current findings related
injection of CD133þ/CD26þ/CD44þ cells, but not to the dynamics of colorectal CSCs, particularly the sig-
CD133þ/CD26/CD44þ cells, led to liver metastasis

Downloaded from http://aacrjournals.org/cancerpreventionresearch/article-pdf/12/7/413/2244191/413.pdf by guest on 28 March 2022


naling pathways and intra- and extratumoral factors
(21). In vitro evaluations revealed that CD26 knockdown involved in their regulation. While ISCs are not the primary
by small interfering RNA (siRNA) reduced the migratory focus of this section, we have provided a brief description
and invasive capacities of the CD26þ cells, with a down- of them, focusing on ISC findings that offer clues to
regulation of epithelial–mesenchymal transition (EMT) understanding CSC dynamics in colorectal cancer.
markers (21). Consistent with these findings, clinical
studies have reported that CD26 expression in colorectal Relationship between ISCs and colorectal CSCs
cancer is correlated with poor prognoses (22–24). Inter- CSCs share phenotypic and molecular characteristics
estingly, CD26þ/CD326 circulating cancer cells have with their normal counterparts, tissue-resident, adult stem
been proposed as prognostic markers for the recurrence cells. Notably, the colorectal CSC markers identified to date
of colorectal cancer (24). are also expressed by normal ISCs (12, 26). Conversely, the
In a recent study, Todaro and colleagues pheno- human homolog of the best-documented marker for
typically identified colorectal CSCs with metastatic murine ISCs, leucine-rich repeat-containing G-protein–
capacity based on their expression of the variant form coupled receptor 5 (LGR5), could also serve as a marker
6 of CD44 (CD44v6; ref. 11). CD44v6þ cells were able for CSCs in human colorectal cancers (27). A recent
to induce tumor growth in gut, lung, and liver after study showed that LGR5-expressing cells possess CSC
orthotopic injection into mice, while their CD44v6 properties in xenografted organoids derived from human
counterparts were not metastatic (11). Interestingly, colorectal cancer (28). CSCs do not necessarily originate
while there was a substantial overlap between CD44v6þ from the transformation of normal stem cells, but in
and CD26þ populations, CD44v6þ/CD26 cells show- the case of colorectal cancer, the similarity could be attrib-
ed considerable metastatic potential in the orthotopic utable, at least in part, to the origin of CSCs. Colorectal
model (11), suggesting phenotypic heterogeneity even cancer develops and progresses by the sequential accumu-
within metastatic CSCs. lation of genetic alterations, in which activation of the
Interestingly, a recent study suggested that preferential Wnt/b-catenin signaling pathway, via an APC or CTNNB1
metastasis to particular target organs could be attributable (b-catenin) mutation, marks the first step in tumor forma-
in part to phenotypic/functional diversity within metastat- tion. Given that intestinal epithelial cells are consistently
ic CSCs. Gao and colleagues reported that only colorectal renewed for a shorter period than that required to accu-
CSCs expressing CD110, a specific receptor for thrombo- mulate causative genetic alterations, it is reasonable to
poietin, were able to colonize the liver after orthotopic hypothesize that CSCs arise from long-lived ISCs. The
implantation in immunocompromised mice, while CSCs discovery of reliable murine ISC markers, including
expressing CUB-domain-containing protein 1 (CDCP1) Lgr5 (9) and B lymphoma Mo-MLV insertion region 1 (Bmi1;
produced lung metastases (25). They showed that knock- ref. 29), enables the testing of this hypothesis. Actually, it
down of either CD110 or CDCP1 by siRNA reduced liver or has been demonstrated that specific activation of Wnt/
lung metastasis, respectively, but neither had a discernible b-catenin signaling in ISCs expressing Lgr5, Bmi1, or Cd133
effect on primary tumor growth. In addition, they showed results in adenoma formation in mice (29–31).
that CD110 and CDCP1 functioned in extravasation into These findings do not necessarily preclude the
the liver parenchyma and adhesion on the pulmonary possibility that intestinal epithelial cells other than
endothelium, respectively (25). It is notable that both ISCs can be the founders of intestinal tumors under
CD110þ and CDCP1þ CSCs were exclusively contained specific conditions. In fact, activation of Wnt and Notch
within the CD133þ population with no overlap between signaling in basic helix–loop–helix family member
these subpopulations (25), indicating that distinct meta- a15 (Bhlha15)-positive progenitor cells resulted in the

www.aacrjournals.org Cancer Prev Res; 12(7) July 2019 415


Hirata et al.

rapid development of intestinal tumors in mice (32). similarly, Delta-like 1 (Dll1)high secretory progenitor cells
Unlike observations from ISCs (29–31), Wnt activation regenerate stem cell compartments that contain Lgr5þ cells
alone in Bhlha15þ cells was insufficient to drive colonic following irradiation damage (43). In the colon, atonal
carcinogenesis (32), suggesting that ISCs are at higher homolog-1 (Atoh1)þ or Bhlha15þ secretory progenitor cells
risk for tumorigenic transformation. Similarly, activation significantly contribute to colonic crypt regeneration after
of Wnt signaling in doublecortin-like kinase 1 protein mucosal damage (32, 44–46). These findings indicate the
(Dckl1)-positive tuft cells led to colorectal tumor devel- dedifferentiation capacity of the intestinal epithelium;
opment in mice under inflammatory conditions, but not however, this capacity seems limited to fated progenitors
under normal conditions (33). rather than to terminally differentiated cells.
Recent studies have demonstrated the reversion of
differentiated non-CSC populations to CSCs in novel
Convertibility of ISCs and colorectal CSCs
colorectal cancer models using genetically engineered
ISCs are one of the most intensively studied subjects in
organoids (28, 47). By xenotransplantation of organoids
stem cell biology. While previous works based on label-
derived from human colorectal cancer cells, Shimokawa
retaining cells (34) hypothesized the existence of a single
and colleagues demonstrated that KRT20þ differentiated

Downloaded from http://aacrjournals.org/cancerpreventionresearch/article-pdf/12/7/413/2244191/413.pdf by guest on 28 March 2022


quiescent population of stem cells residing in a specific
tumor cells revert to LGR5þ CSCs and contribute
location of the intestinal crypt, it is currently accepted that
to tumor growth after selective ablation of LGR5þ
both quiescent and active ISCs coexist in distinct
CSCs (28). Likewise, the recovery of functional Lgr5þ
niches (35). Both populations possess the capacity to
CSCs was also observed in transplanted mouse colon
self-renew and give rise to all differentiated intestinal
cancer organoids, in which Lgr5 cells rapidly replen-
epithelial cell types, despite having entirely different pro-
ished Lgr5þ CSC populations after the ablation of Lgr5þ
liferative activities. In the small intestine of mice, Lgr5
cells (47).
is the best-known marker for active ISCs (9), while quies-
cent ISCs have multiple markers including Bmi1 (29),
homeodomain-only protein (Hopx; ref. 36), telomerase reverse Regulatory factors for colorectal CSCs
transcriptase (Tert; ref. 37), and leucine-rich repeats and Consistent with the importance of Wnt/b-catenin sig-
immunoglobulin-like domains protein 1 (Lrig1; ref. 38). Active naling in both intestinal stemness and colon carcinogen-
and quiescent ISCs represent functionally distinct ISC esis, Wnt signaling plays a central role in the regulation of
populations: actively cycling Lgr5þ ISCs contribute to colorectal CSCs. Although other signaling pathways have
homeostatic epithelial renewal, while slow-cycling Bmi1þ been implicated in the control of colorectal CSCs, here we
ISCs are considered to be a reserve stem cell pool, based on focus on Wnt/b-catenin signaling. Vermeulen and collea-
their active contribution to regeneration after radiation gues demonstrated that colorectal cancer cells show vari-
damage (39). Consistently, it has been reported that Bmi1þ able levels of Wnt activation and that only those with the
ISCs give rise to Lgr5þ ISCs in the small intestine of mice highest levels possess CSC properties (48). A study from
after the selective ablation of Lgr5þ cells (40). Conversely, our group also supports the notion that the Wnt activity
Takeda and colleagues showed that Lgr5þ cells can give rise levels define colorectal CSCs. We investigated the dose-
to Hopxþ cells in mice and in organoid cultures (36). These dependent effect of Wnt activation in the mouse colonic
results indicate that active and quiescent ISCs can inter- epithelium by regulating the expression levels of S33Y
convert and/or replenish each other. In the colon of mice, mutant b-catenin (49). Higher levels of mutant b-catenin
Lgr5þ cells also reside at the crypt base and serve as active expression induced the amplification of Lgr5þ cells in
ISCs (9), while Lrig1 marks predominately quiescent stem colonic crypts with de novo crypt formation, whereas lower
cells (38). Asfaha and colleagues identified Lgr5 cells with levels of expression only enhanced cell proliferation (49).
ISC properties within a Krt19þ population distributed Consistent with these findings, Ordonez-Moran and col-
above the crypt base in the colon of mice (41). Genetic leagues showed that HOXA5 induction counteracted CSC
lineage-tracing experiments revealed that Krt19þ/Lgr5 traits, preventing tumor growth and metastasis by inhibit-
colonic cells generated entire colonic crypts, including ing Wnt signaling activity in colorectal cancers (50). Colo-
Lgr5þ cells at the colonic crypt base (41). rectal CSCs is regulated through the interaction between
The differentiation of intestinal epithelial cells normally Wnt and other signaling pathways, as is the normal ISCs.
proceeds in a unidirectional manner, from ISCs to the For example, bone morphogenetic protein 4 (BMP4) pro-
terminally differentiated cells via lineage-committed pro- motes differentiation and apoptosis by antagonizing Wnt
genitor cells, but recent studies have demonstrated the signaling in colorectal CSCs (51). In a recent study,
dedifferentiation capacity of the intestinal epithelial cells. Whissell and colleagues identified the transcription factor
In the small intestine of mice, Alkaline phosphatase intestinal GATA-binding factor 6 (GATA6) as a key regulator of Wnt
(AlPi)-expressing enterocyte progenitors can dedifferenti- and BMP signaling in colorectal cancers (52). GATA6
ate into Lgr5þ ISCs upon the depletion of Lgr5þ cells (42); enabled CSC self-renewal through the repression of BMP

416 Cancer Prev Res; 12(7) July 2019 Cancer Prevention Research
Colorectal Cancer Stem Cells

gene expression, competing with b-catenin/Tcf4 to bind to Multiple reports have shown that colorectal CSCs
a regulatory region of the BMP4 locus (52). are more resistant to chemotherapy (60, 61) and there-
Microenvironmental factors are important in the regu- fore likely play an essential role in recurrence follow-
lation of colorectal CSCs as well as of their normal ing conventional anticancer treatments. Enrichment
counterparts, ISCs (53). Tumor stroma undergoes dra- of colorectal CSCs in xenografts after chemotherapy
matic changes during the process of tumor progression clearly showed the limitation of conventional therapies
and therefore could produce more complex effects than for colorectal cancers (28, 60). Therefore, CSCs repre-
it does in normal homeostatic conditions. Hepatocyte sent an attractive target for more effective therapies,
growth factor (HGF) secreted from myofibroblasts and several potential CSC-targeted drugs or strategies
induced CSC properties in WntLow colorectal cancer cells have been actually proposed (62). However, consider-
by enhancing Wnt signaling activity (48). When cocul- ing the heterogeneity and dynamism of colorectal
tured with colorectal cancer cell lines, mesenchymal CSC populations, CSCs no longer represent a fixed tar-
stem cells increased the number of cancer cells with get population, making it necessary to establish improv-
tumor-initiating capacity by producing prostaglandin ed strategies, designed to counteract to their dynamic

Downloaded from http://aacrjournals.org/cancerpreventionresearch/article-pdf/12/7/413/2244191/413.pdf by guest on 28 March 2022


E2 and cytokines that activated Wnt/b-catenin signal- nature.
ing (54). It is possible that CSCs acquire a metastatic Recent studies have indicated that the selective ablation
capacity by the actions of cytokines secreted from cancer- of colorectal CSCs is not sufficient to produce complete
associated fibroblasts (CAF) during tumor progression. tumor regression (28, 47). In allografts of mouse colorectal
As described above, colorectal CSCs with metastatic cancer organoids, continuous ablation of Lgr5þ CSCs
capacity are phenotypically identified by CD44v6 ex- restricted tumor growth but did not produce tumor regres-
pression (11). Cytokines secreted from CAFs, including sion, because proliferative Lgr5 cells maintained the
HGF, osteopontin, and stromal-derived factor 1a, tumors even with depletion of Lgr5þ CSCs (47). Further-
enhanced CD44v6 expression by activating Wnt signal- more, Lgr5 cells can rapidly replenish the Lgr5þ popula-
ing, transforming nonmetastatic progenitors into meta- tion to reinitiate tumor growth after cessation of Lgr5þ
static CSCs (11). In addition to stromal cells, Paneth cell ablation (47). A similar phenomenon was observed
cells and cKitþ secretory cells constitute niches for Lgr5þ in xenografted human colorectal cancer organoids
ISCs in the murine small intestine and colon, respec- (28). Upon ablation of LGR5þ CSCs, tumor size was
tively, where they provide essential signals for stem cell reduced (28). However, compensatory proliferation of
maintenance, including the Notch ligand Dll4 (55, 56). KRT20þ non-CSC cells reduced the effectiveness of the
As Notch signaling is elevated in colorectal CSCs (57) ablation and the tumor eventually regrew in parallel with
and antibody blockade of DLL4 reduces CSC frequency the reappearance of LGR5þ cells (28).
in colorectal cancers (57), the CSC state might also be
regulated through interactions between tumor cells in
colorectal cancers. Conclusion
The regulation of Wnt and BMP4 signaling pathways Although CSCs were initially considered to be cell
by activated thyroid hormones has been reported in populations with well-defined phenotypic and molec-
colorectal CSCs (58). Type 3 deiodinase 3 (D3), a chief ular characteristics, CSCs have more recently been
thyroid hormone T3–inactivating enzyme, has been shown to be phenotypically/functionally heterogeneous
shown to be expressed at high levels in CD133þ and and highly dynamic populations. These characteristics
Wnthigh cell populations (58). T3 treatment induced present a serious problem in establishing therapeutic
CSC differentiation, decreasing the tumorigenic poten- strategies targeting CSCs. Considering that the stemness
tial in CSCs, accompanied by the upregulation of BMP4 of colorectal cancer cells is a dynamic state that is
and attenuation of Wnt signaling (58). These findings constantly altered by multiple extrinsic factors, in addi-
suggest that colorectal cancers may be regulated not only tion to intrinsic cellular factors (genetic and epigenetic
by local factors within or surrounding a lesion, but also alterations), a better understanding of the tumor envi-
by hormonal action. ronment should lead to improved strategies for the
eradication of colorectal CSC by regulating their CSC
Clinical Implications of Colorectal CSCs state.

The CSC concept has attracted a great deal of interest for


its potential clinical implications. Colorectal cancer Disclosure of Potential Conflicts of Interest
No potential conflicts of interest were disclosed.
remains the fourth leading cause of cancer-related deaths
worldwide. Although its occurrence is declining in devel-
oped countries, its incidence is still rapidly rising in many Received December 12, 2018; revised April 7, 2019; accepted May
developing countries (59). 14, 2019; published first May 17, 2019.

www.aacrjournals.org Cancer Prev Res; 12(7) July 2019 417


Hirata et al.

References
1. Heppner GH, Miller BE. Tumor heterogeneity: biological impli- growth and metastatic activity in human pancreatic cancer.
cations and therapeutic consequences. Cancer Metastasis Rev Cell Stem Cell 2007;1:313–23.
1983;2:5–23. 20. Zhang SS, Han ZP, Jing YY, Tao SF, Li TJ, Wang H, et al. CD133(þ)
2. Kreso A, Dick JE. Evolution of the cancer stem cell model. CXCR4(þ) colon cancer cells exhibit metastatic potential and
Cell Stem Cell 2014;14:275–91. predict poor prognosis of patients. BMC Med 2012;10:85.
3. Bonnet D, Dick JE. Human acute myeloid leukemia is organized 21. Pang R, Law WL, Chu AC, Poon JT, Lam CS, Chow AK, et al. A
as a hierarchy that originates from a primitive hematopoietic cell. subpopulation of CD26þ cancer stem cells with metastatic
Nat Med 1997;3:730–7. capacity in human colorectal cancer. Cell Stem Cell 2010;6:
4. Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres- 603–15.
Cortes J, et al. A cell initiating human acute myeloid leukaemia 22. de la Haba-Rodriguez J, Macho A, Calzado MA, Blazquez MV,
after transplantation into SCID mice. Nature 1994;367:645–8. Gomez MA, Munoz EE, et al. Soluble dipeptidyl peptidase IV
5. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke (CD-26) in serum of patients with colorectal carcinoma.
MF. Prospective identification of tumorigenic breast cancer cells. Neoplasma 2002;49:307–11.
Proc Natl Acad Sci U S A 2003;100:3983–8. 23. Lam CS, Cheung AH, Wong SK, Wan TM, Ng L, Chow AK, et al.
6. Dalerba P, Dylla SJ, Park IK, Liu R, Wang X, Cho RW, et al. Prognostic significance of CD26 in patients with colorectal can-

Downloaded from http://aacrjournals.org/cancerpreventionresearch/article-pdf/12/7/413/2244191/413.pdf by guest on 28 March 2022


Phenotypic characterization of human colorectal cancer stem cer. PLoS One 2014;9:e98582.
cells. Proc Natl Acad Sci U S A 2007;104:10158–63. 24. Lieto E, Galizia G, Orditura M, Romano C, Zamboli A, Castellano
7. O'Brien CA, Pollett A, Gallinger S, Dick JE. A human colon cancer P, et al. CD26-positive/CD326-negative circulating cancer cells as
cell capable of initiating tumour growth in immunodeficient prognostic markers for colorectal cancer recurrence. Oncol Lett
mice. Nature 2007;445:106–10. 2015;9:542–50.
8. Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, 25. Gao W, Chen L, Ma Z, Du Z, Zhao Z, Hu Z, et al. Isolation and
Peschle C, et al. Identification and expansion of human colon- phenotypic characterization of colorectal cancer stem cells with
cancer-initiating cells. Nature 2007;445:111–5. organ-specific metastatic potential. Gastroenterology 2013;145:
9. Barker N, van Es JH, Kuipers J, Kujala P, van den Born M, 636–46.
Cozijnsen M, et al. Identification of stem cells in small intestine 26. Snippert HJ, van Es JH, van den Born M, Begthel H, Stange DE,
and colon by marker gene Lgr5. Nature 2007;449:1003–7. Barker N, et al. Prominin-1/CD133 marks stem cells and early
10. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, et al. progenitors in mouse small intestine. Gastroenterology 2009;
Identification of human brain tumour initiating cells. Nature 136:2187–94.
2004;432:396–401. 27. Kemper K, Prasetyanti PR, De Lau W, Rodermond H, Clevers H,
11. Todaro M, Gaggianesi M, Catalano V, Benfante A, Iovino F, Medema JP. Monoclonal antibodies against Lgr5 identify
Biffoni M, et al. CD44v6 is a marker of constitutive and repro- human colorectal cancer stem cells. Stem Cells 2012;30:
grammed cancer stem cells driving colon cancer metastasis. 2378–86.
Cell Stem Cell 2014;14:342–56. 28. Shimokawa M, Ohta Y, Nishikori S, Matano M, Takano A, Fujii M,
12. Huang EH, Hynes MJ, Zhang T, Ginestier C, Dontu G, Appelman et al. Visualization and targeting of LGR5(þ) human colon cancer
H, et al. Aldehyde dehydrogenase 1 is a marker for normal and stem cells. Nature 2017;545:187–92.
malignant human colonic stem cells (SC) and tracks SC over- 29. Sangiorgi E, Capecchi MR. Bmi1 is expressed in vivo in intestinal
population during colon tumorigenesis. Cancer Res 2009;69: stem cells. Nat Genet 2008;40:915–20.
3382–9. 30. Barker N, Ridgway RA, van Es JH, van de Wetering M, Begthel H,
13. Vermeulen L, Todaro M, de Sousa Mello F, Sprick MR, Kemper K, van den Born M, et al. Crypt stem cells as the cells-of-origin of
Perez Alea M, et al. Single-cell cloning of colon cancer stem cells intestinal cancer. Nature 2009;457:608–11.
reveals a multi-lineage differentiation capacity. Proc Natl Acad Sci 31. Zhu L, Gibson P, Currle DS, Tong Y, Richardson RJ, Bayazitov IT,
U S A 2008;105:13427–32. et al. Prominin 1 marks intestinal stem cells that are susceptible to
14. Beier D, Hau P, Proescholdt M, Lohmeier A, Wischhusen J, Oefner neoplastic transformation. Nature 2009;457:603–7.
PJ, et al. CD133(þ) and CD133(-) glioblastoma-derived cancer 32. Hayakawa Y, Tsuboi M, Asfaha S, Kinoshita H, Niikura R, Konishi
stem cells show differential growth characteristics and molecular M, et al. BHLHA15-positive secretory precursor cells can give rise
profiles. Cancer Res 2007;67:4010–5. to tumors in intestine and colon in mice. Gastroenterology 2019;
15. Joo KM, Kim SY, Jin X, Song SY, Kong DS, Lee JI, et al. Clinical and 156:1066–81.
biological implications of CD133-positive and CD133-negative 33. Westphalen CB, Asfaha S, Hayakawa Y, Takemoto Y, Lukin DJ,
cells in glioblastomas. Lab Invest 2008;88:808–15. Nuber AH, et al. Long-lived intestinal tuft cells serve as colon
16. Wang J, Sakariassen PO, Tsinkalovsky O, Immervoll H, Boe SO, cancer-initiating cells. J Clin Invest 2014;124:1283–95.
Svendsen A, et al. CD133 negative glioma cells form tumors in 34. Booth C, Potten CS. Gut instincts: thoughts on intestinal epithe-
nude rats and give rise to CD133 positive cells. Int J Cancer 2008; lial stem cells. J Clin Invest 2000;105:1493–9.
122:761–8. 35. Li L, Clevers H. Coexistence of quiescent and active adult stem
17. Shmelkov SV, Butler JM, Hooper AT, Hormigo A, Kushner J, Milde cells in mammals. Science 2010;327:542–5.
T, et al. CD133 expression is not restricted to stem cells, and both 36. Takeda N, Jain R, LeBoeuf MR, Wang Q, Lu MM, Epstein JA.
CD133þ and CD133- metastatic colon cancer cells initiate Interconversion between intestinal stem cell populations in dis-
tumors. J Clin Invest 2008;118:2111–20. tinct niches. Science 2011;334:1420–4.
18. Haraguchi N, Ohkuma M, Sakashita H, Matsuzaki S, Tanaka F, 37. Montgomery RK, Carlone DL, Richmond CA, Farilla L,
Mimori K, et al. CD133þCD44þ population efficiently enriches Kranendonk ME, Henderson DE, et al. Mouse telomerase
colon cancer initiating cells. Ann Surg Oncol 2008;15:2927–33. reverse transcriptase (mTert) expression marks slowly cycling
19. Hermann PC, Huber SL, Herrler T, Aicher A, Ellwart JW, Guba M, intestinal stem cells. Proc Natl Acad Sci U S A 2011;108:
et al. Distinct populations of cancer stem cells determine tumor 179–84.

418 Cancer Prev Res; 12(7) July 2019 Cancer Prevention Research
Colorectal Cancer Stem Cells

38. Powell AE, Wang Y, Li Y, Poulin EJ, Means AL, Washington MK, 50. Ordonez-Moran P, Dafflon C, Imajo M, Nishida E, Huelsken J.
et al. The pan-ErbB negative regulator Lrig1 is an intestinal stem HOXA5 counteracts stem cell traits by inhibiting wnt signaling in
cell marker that functions as a tumor suppressor. Cell 2012;149: colorectal cancer. Cancer Cell 2015;28:815–29.
146–58. 51. Lombardo Y, Scopelliti A, Cammareri P, Todaro M, Iovino F,
39. Yan KS, Chia LA, Li X, Ootani A, Su J, Lee JY, et al. The intestinal Ricci-Vitiani L, et al. Bone morphogenetic protein 4 induces
stem cell markers Bmi1 and Lgr5 identify two functionally differentiation of colorectal cancer stem cells and increases their
distinct populations. Proc Natl Acad Sci U S A 2012;109: response to chemotherapy in mice. Gastroenterology 2011;140:
466–71. 297–309.
40. Tian H, Biehs B, Warming S, Leong KG, Rangell L, Klein OD, et al. 52. Whissell G, Montagni E, Martinelli P, Hernando-Momblona X,
A reserve stem cell population in small intestine renders Lgr5- Sevillano M, Jung P, et al. The transcription factor GATA6 enables
positive cells dispensable. Nature 2011;478:255–9. self-renewal of colon adenoma stem cells by repressing BMP gene
41. Asfaha S, Hayakawa Y, Muley A, Stokes S, Graham TA, Ericksen expression. Nat Cell Biol 2014;16:695–707.
RE, et al. Krt19(þ)/Lgr5(-) cells are radioresistant cancer-initiat- 53. Medema JP, Vermeulen L. Microenvironmental regulation of
ing stem cells in the colon and intestine. Cell Stem Cell 2015;16: stem cells in intestinal homeostasis and cancer. Nature 2011;
627–38. 474:318–26.
42. Tetteh PW, Basak O, Farin HF, Wiebrands K, Kretzschmar K, 54. Li HJ, Reinhardt F, Herschman HR, Weinberg RA. Cancer-

Downloaded from http://aacrjournals.org/cancerpreventionresearch/article-pdf/12/7/413/2244191/413.pdf by guest on 28 March 2022


Begthel H, et al. Replacement of lost Lgr5-positive stem cells stimulated mesenchymal stem cells create a carcinoma stem
through plasticity of their enterocyte-lineage daughters. Cell Stem cell niche via prostaglandin E2 signaling. Cancer Discov 2012;
Cell 2016;18:203–13. 2:840–55.
43. van Es JH, Sato T, van de Wetering M, Lyubimova A, Yee Nee 55. Rothenberg ME, Nusse Y, Kalisky T, Lee JJ, Dalerba P, Scheeren F,
AN, Gregorieff A, et al. Dll1þ secretory progenitor cells revert et al. Identification of a cKit(þ) colonic crypt base secretory cell
to stem cells upon crypt damage. Nat Cell Biol 2012;14: that supports Lgr5(þ) stem cells in mice. Gastroenterology 2012;
1099–104. 142:1195–205.
44. Castillo-Azofeifa D, Fazio EN, Nattiv R, Good HJ, Wald T, Pest 56. Sato T, van Es JH, Snippert HJ, Stange DE, Vries RG, van den Born
MA, et al. Atoh1(þ) secretory progenitors possess renewal capac- M, et al. Paneth cells constitute the niche for Lgr5 stem cells in
ity independent of Lgr5(þ) cells during colonic regeneration. intestinal crypts. Nature 2011;469:415–8.
EMBO J 2019;38. pii: e99984. 57. Hoey T, Yen WC, Axelrod F, Basi J, Donigian L, Dylla S, et al. DLL4
45. Ishibashi F, Shimizu H, Nakata T, Fujii S, Suzuki K, Kawamoto A, blockade inhibits tumor growth and reduces tumor-initiating cell
et al. Contribution of ATOH1(þ) cells to the homeostasis, repair, frequency. Cell Stem Cell 2009;5:168–77.
and tumorigenesis of the colonic epithelium. Stem Cell Rep 2018; 58. Catalano V, Dentice M, Ambrosio R, Luongo C, Carollo R,
10:27–42. Benfante A, et al. Activated thyroid hormone promotes differen-
46. Tomic G, Morrissey E, Kozar S, Ben-Moshe S, Hoyle A, Azzarelli R, tiation and chemotherapeutic sensitization of colorectal cancer
et al. Phospho-regulation of ATOH1 is required for plasticity of stem cells by regulating Wnt and BMP4 Signaling. Cancer Res
secretory progenitors and tissue regeneration. Cell Stem Cell 2016;76:1237–44.
2018;23:436–43. 59. International Agency for Research on Cancer. World Cancer
47. de Sousa e Melo F, Kurtova AV, Harnoss JM, Kljavin N, Hoeck JD, Report 2014. Lyon, France: World Health Organization; 2014.
Hung J, et al. A distinct role for Lgr5(þ) stem cells in primary and 60. Dylla SJ, Beviglia L, Park IK, Chartier C, Raval J, Ngan L, et al.
metastatic colon cancer. Nature 2017;543:676–80. Colorectal cancer stem cells are enriched in xenogeneic tumors
48. Vermeulen L, De Sousa EMF, van der Heijden M, Cameron K, de following chemotherapy. PLoS One 2008;3:e2428.
Jong JH, Borovski T, et al. Wnt activity defines colon cancer stem 61. Todaro M, Alea MP, Di Stefano AB, Cammareri P, Vermeulen L,
cells and is regulated by the microenvironment. Nat Cell Biol Iovino F, et al. Colon cancer stem cells dictate tumor growth and
2010;12:468–76. resist cell death by production of interleukin-4. Cell Stem Cell
49. Hirata A, Utikal J, Yamashita S, Aoki H, Watanabe A, Yamamoto 2007;1:389–402.
T, et al. Dose-dependent roles for canonical Wnt signalling in de 62. Saygin C, Matei D, Majeti R, Reizes O, Lathia JD. Targeting cancer
novo crypt formation and cell cycle properties of the colonic stemness in the clinic: from hype to hope. Cell Stem Cell 2019;24:
epithelium. Development 2013;140:66–75. 25–40.

www.aacrjournals.org Cancer Prev Res; 12(7) July 2019 419


Downloaded from http://aacrjournals.org/cancerpreventionresearch/article-pdf/12/7/413/2244191/413.pdf by guest on 28 March 2022

You might also like