Download Cancer stem cells: AMLs show the way

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Extracellular matrix wikipedia , lookup

Cell cycle wikipedia , lookup

Cell growth wikipedia , lookup

Mitosis wikipedia , lookup

Tissue engineering wikipedia , lookup

Cell culture wikipedia , lookup

Cell encapsulation wikipedia , lookup

Organ-on-a-chip wikipedia , lookup

List of types of proteins wikipedia , lookup

SULF1 wikipedia , lookup

Cellular differentiation wikipedia , lookup

JADE1 wikipedia , lookup

Amitosis wikipedia , lookup

Hematopoietic stem cell wikipedia , lookup

Transcript
Stem Cells and Development
Cancer stem cells: AMLs show the way
D. Bonnet1
Hematopoietic Stem Cell Laboratory, Cancer Research U.K., London Research Institute, 44 Lincoln’s Inn Fields, London WC2A 3PX, U.K.
Abstract
The blood-related cancer leukaemia was the first disease where human CSCs (cancer stem cells), or LSCs
(leukaemic stem cells), were isolated. The haematopoietic system is one of the best tissues for investigating
CSCs, since the developmental hierarchy of normal blood formation is well defined. Leukaemia can now
be viewed as aberrant haematopoietic processes initiated by rare LSCs that have maintained or reacquired
the capacity for indefinite proliferation through accumulated mutations and/or epigenetic changes. Yet,
despite their critical importance, much remains to be learned about the developmental origin of LSCs and
the mechanisms responsible for their emergence in the course of the disease. This report will review our
current knowledge on LSC development and finally demonstrate how these discoveries provide a paradigm
for identification of CSCs from solid tumours.
The hallmark properties of HSCs (haematopoietic stem cells)
are the ability to balance self-renewal versus differentiation
cell fate decisions to provide sufficient primitive cells to sustain haematopoieisis, while generating more mature cells with
specialized capacities. Through the process of asymmetric
cell division, a single division can result in the formation
of both an identical stem cell and a more mature cell [1]. In
order to ensure a persistent pool of regenerating cells without
outgrowth of immature cell types, tight regulation of HSC
division is required. Unchecked growth of immature cells is
thought to represent a paradigm for malignant outgrowth,
at least for AML (acute myeloid leukaemia) and chronic
myeloid leukaemia [2,3]. Thus determining the composition
and relationship of the cell types that constitute the human
stem cell compartment may help both to identify the cellular
and molecular factors that govern normal and LSC (leukaemic
stem cell) development, and to advance clinical applications of
transplantation, gene therapy, stem cell expansion and tumour
cell purging. This review will introduce the notion of LSCs,
discuss the potential origin of these cells with an emphasis
on myeloid leukaemia and finally examine the impacts these
discoveries may have clinically and in understanding the
organization of cancer of other tissues.
Functional heterogeneity in tumours
The development of quantitative assays enabling measurement of the clonogenicity of malignant haematopoietic cells
led to the first demonstrations that only a small subset of cancer cells is capable of extensive proliferation in vitro [4]. Such
studies revealed the existence of functional heterogeneity
Key words: cancer stem cell (CSC), haematopoietic stem cell (HSC), leukaemic stem cell (LSC),
self-renewal, tumour, xenotransplantation model.
Abbreviations used: AML, acute myeloid leukaemia; CMP, common myeloid progenitor; CSC,
cancer stem cell; GMP, granulocyte/monocyte progenitor; HSC, haematopoietic stem cell; LSC,
leukaemic stem cell; MLL, mixed lineage leukaemia; NOD, non-obese diabetic; SCID, severe
immunodeficient mice; SL-IC, SCID leukaemia-initiating cell.
1
To whom correspondence should be addressed (email dominique.bonnet@cancer.
org.uk).
within tumours, and introduced the concept of tumour stem
cells. Subsequently, studies in AML have been key in elucidating the biological basis of tumour heterogeneity. AML
is a clonal disorder of aberrant haematopoiesis characterized
by an accumulation of functionally immature blasts, which
fail to differentiate normally. Despite their morphological
homogeneity, the blast cell population is biologically
heterogeneous. Only a minority of proliferative leukaemic
blasts (AML-CFU) is able to give rise to colonies in vitro.
This observation suggested that, as in normal haematopoiesis, the leukaemic clone in AML is organized as a
hierarchy, in which a small number of proliferating progenitors continuously replenish the bulk population of noncycling leukaemic blasts.
Concept of LSCs
Emerging evidence has provided new insights into cancer
biology by emphasizing the relationship between stem cells
and tumour cells and by proposing the notion that cancer
cells might contain some CSCs (cancer stem cells), which are
rare cells with indefinite self-renewal potential that drive the
formation and growth of the tumours. The existence of CSCs
was revealed first in leukaemia [5–7] but has now extended
to other cancer types [8–10]. Transplantation of primary
AML cells into SCID (severe immunodeficient) [6] or NOD
(non-obese diabetic)/SCID [7] mice led to the finding that
only rare cells, termed SL-ICs (SCID leukaemia-initiating
cells), are capable of initiating and sustaining growth of the
leukaemic clone in vivo. In addition to their ability to differentiate and proliferate, serial transplantation experiments
showed that SL-ICs possess high self-renewal capacity, and
thus can be considered to be AML stem cells. Importantly,
SL-ICs can be prospectively identified and purified as
CD34+ CD38− cells in AML patient samples, regardless of
the phenotype of the bulk blast population, and are the
only cells capable of self-renewal, as demonstrated by serial
transplantation [7]. These findings show that, like the normal
C 2005
Biochemical Society
1531
1532
Biochemical Society Transactions (2005) Volume 33, part 6
haematopoietic system, AML is organized as a hierarchy of
distinct, functionally heterogeneous classes of cells that is
ultimately sustained by a small number of LSCs. These studies
provided the first direct evidence for the CSC hypothesis.
Comparison between normal HSCs and LSCs
While LSCs appear to share similar cell-surface markers
previously identified for normal HSCs, such as CD34, CD38,
HLA-DR and CD71, several groups have reported that some
markers are differentially expressed between the two, such
as CD90, Thy.1, c-kit and IL-3 (interleukin 3) receptor
[11–14]. Despite these few phenotypic differences between
normal HSCs and LSCs, a recent work has reported similar
heterogeneity in the normal and LSC compartment based
on self-renewal and proliferation capacities. Using clonal
tracking of retroviral-transduced normal and leukaemic cells
in NOD/SCID mice, it was demonstrated that both normal
and LSC compartments were composed of individual stem
cell classes that differ in their repopulating and self-renewal
capacities [15,16]. Overall, these findings suggest that the
pathways that regulate normal commitment/differentiation
and self-renewal processes in haematopoietic cells are not
completely abolished in LSC. Rather, the effects of transforming mutations are layered on to the normal developmental framework of HSC, resulting in the leukaemic clone
having an aberrant developmental hierarchy that retains
aspects of its normal counterpart. This concept is supported
by a correlation between genes required for normal haematopoietic development and those perturbed in leukaemia [17],
and by the recent demonstration that Bmi-1 plays a key role
in self-renewal determination in both normal and leukaemic
murine stem cells [18,19].
Gene expression pattern of LSCs versus
normal HSCs
The phenotypic description of LSCs now enables their
purification and will facilitate identification of genes that
are preferentially expressed in these cells compared with
normal HSCs. However, gene expression profiling is
usually conducted on mononuclear cells of AML patients
from either peripheral blood and/or bone marrow. Gene
expression profiling of highly purified LSCs would allow
the identification of genes that reflect the biology of the cells
that are actually driving the leukaemia. Hence, in addition to
being a more efficient way to further understand the biology
of LSCs, this should also provide a more efficient way of
identifying new therapeutics and diagnostic targets.
The cell of origin in cancer: studies in AML
A focus of much cancer research is identification of the
normal cell within which cancer initiates. The target of cell
transforming mutations is still unknown. Because normal
stem cells and LSCs share the ability to self-renew, as well
as various developmental pathways, it has been postulated
that LSCs are HSCs that have become leukaemic as the result
of accumulated mutations. Conversely, LSCs could derive
C 2005
Biochemical Society
from more committed progenitors or even a differentiated
mature cell, which would first have to reacquire the selfrenewal capacity before accumulating additional mutations.
There are two reasons to think that normal HSCs themselves are the target of leukaemic transformation. First, HSCs
have the machinery for self-renewal already activated; thus
maintaining this activation may be simpler than turning it
on de novo in a more differentiated cell. Secondly, stem
cells persist for long period of time and thus have a greater
opportunity to accumulate mutations than more mature
short-lived cell types.
There is now evidence that most subtypes of human AML
arise from mutations that accumulate in HSCs. For most
AML subtypes, except for promyelocytic leukaemia (AMLM3) subtype, the only cells capable of transplanting leukaemia in NOD/SCID mice have a CD34+ CD38− phenotype, similar to that of normal HSCs, whereas more mature
CD34+ CD38+ leukaemic blasts cannot transfer the disease
to mice [5,7].
On the other hand, evidence indicating that cells devoid
of self-renewal activity, such as committed progenitors and
mature cells, can also be the targets for leukaemic transformation comes from analyses of leukaemia-associated genes
in the mouse. Indeed, using promoter elements of several
myeloid-specific human genes [like those encoding MRP8
(multidrug resistance protein 8), CD11b and cathepsin G] to
target transgene expression specifically to committed myeloid
cells allowed the generation of multiple accurate transgenic
mouse models of human leukaemias [20,21]. More recently,
Cozzio et al. [22] have shown that the potent leukaemic fusion
gene MLL-ENL (mixed lineage leukaemia-eleven nineteen
leukaemia), which results from the t(11; 19) translocation,
can induce the exact same leukaemia when transduced into
HSCs as well as into CMPs (common myeloid progenitors)
and GMPs (granulocyte/monocyte progenitors). Another
fusion gene MOZ-TIF2 has also recently been shown to contribute to the transformation of both HSCs and more committed myeloid progenitors [23]. These results imply that
myeloid leukaemias induced by these oncogenes can be initiated in committed progenitors due to their intrinsic capacities to confer leukaemogenic self-renewal potential. Using
a different fusion partner of MLL, GAS7 (growth arrest specific gene 7), So et al. [24] showed that only the transduction
of murine HSCs but not CMP, and GMP resulted in the
production of mixed lineage leukaemias in transplanted mice.
Although the mouse system provides a valuable tool to study
leukaemogenesis, the results obtained for mice do not imply
that committed myeloid progenitors will necessarily be the
target of transformation in the corresponding human disease.
Nevertheless, it appears highly probable that human AML
might arise from cells at both HSC and myeloid stages,
depending mostly on the nature of the associated fusion gene.
CSCs in solid tumours
Recent studies in solid tumours indicate that the concept of
cancer as a hierarchy that is initiated and maintained by a
Stem Cells and Development
rare population of stem cells may have broader implications
beyond the field of haematopoiesis. Al-Hajj et al. [10] were
able to prospectively isolate a minor phenotypically distinct
subset of breast cancer cells that was able to recapitulate the
tumours when transplanted into NOD/SCID mice. Thus
like AML, breast cancer appears to be driven by a rare
subpopulation of cells that demonstrate self-renewal and
produce differentiated non-tumorigenic progenies. A recent
report has also suggested the existence of brain CSCs, which
are able to generate new tumours in vivo that exhibit both
self-renewal and differentiation [8,9].
Based on these recent studies, the paradigm of cancer as
a hierarchical disease whose growth is sustained by a rare
population of stem cells is emerging. Implicit in this model of
cancer development is the notion that CSCs are biologically
distinct from other cells in the tumour, and are able to initiate
and sustain tumour growth in vivo, whereas the bulk cells are
not.
Conclusions
The identification of CSCs has important implications for
future research as well as for the development of novel
therapies. In order to learn more about the nature of the events
involved in cancer, research should focus more on CSCs and
not on the bulk cells that makes up the majority of the tumour.
Existing therapies have been developed largely against the
bulk population. The lack of durable response in most cases
suggests that the treatment used may not effectively target
the CSC population. Indeed, the failure of the current
therapeutic regimens is likely to be related to the resistance
and persistence of CSCs. Future studies must focus instead
on identifying and characterizing the rare cancer-initiating
cells, and cancer treatments must be designed to specifically
target these CSCs if they are to effectively cure and prevent
disease relapse.
References
1 Ho, A.D. (2005) Exp. Hematol. 33, 1–8
2 Reya, T., Morrison, S.J., Clarke, M.F. and Weissman, I.L. (2001)
Nature (London) 414, 105–111
3 Passegue, E., Jamieson, C.H.M., Ailles, L.E. and Weissman, I.L. (2003)
Proc. Natl. Acad. Sci. U.S.A. 100, 11842–11849
4 Griffin, J.D. and Lowenberg, B. (1986) Blood 68, 1185–1195
5 Sutherland, H.J., Blair, A. and Zapf, R.W. (1996) Blood 87, 4754–4761
6 Lapidot, T., Sirard, C., Vormoor, J., Murdoch, B., Hoang, T.,
Caceres-Cortes, J., Minden, M., Paterson, B., Caligiuri, M.A. and Dick, J.E.
(1994) Nature (London) 367, 645–648
7 Bonnet, D. and Dick, J.E. (1997) Nat. Med. 3, 730–737
8 Singh, S.K., Clarke, I.D., Terasaki, M., Bonn, V.E., Hawkins, C., Squire, J.
and Dirks, P.B. (2003) Cancer Res. 63, 5821–5828
9 Singh, S.K., Hawkins, C., Clarke, I.D., Squire, J.A., Bayani, J., Hide, T.,
Henkelman, R.M., Cusimano, M.D. and Dirks, P.B. (2004)
Nature (London) 432, 396–401
10 Al-Hajj, M., Wicha, M.S., Benito-Hernandez, A., Morrison, S.J. and
Clarke, M.F. (2003) Proc. Natl. Acad. Sci. U.S.A. 100, 3983–3988
11 Blair, A., Hogge, D.E., Ailles, L.E., Lansdorp, P.M. and Sutherland, H.J.
(1997) Blood 89, 3104–3112
12 Blair, A., Hogge, D.E. and Sutherland, H.J. (1998) Blood 92, 4325–4335
13 Blair, A. and Sutherland, H.J. (2000) Exp. Hematol. 28, 660–671
14 Jordan, C.T., Upchurch, D., Szilvassy, S.J., Guzman, M.L., Howard, D.S.,
Pettigrew, A.L., Meyerrose, T., Rossi, R., Grimes, B., Rizzieri, D.A. et al.
(2000) Leukemia 10, 1777–1784
15 Guenechea, G., Gan, O.I., Dorrell, C. and Dick, J.E. (2001) Nat. Immunol.
1, 75–82
16 Hope, K.J., Jin, L. and Dick, J.E. (2004) Nat. Immunol. 7, 738–743
17 Tenen, D.G. (2003) Nat. Rev. Cancer 3, 89–101
18 Lessard, J. and Sauvageau, G. (2003) Nature (London) 423, 255–260
19 Park, I.K., Qian, D., Kiel, M., Becker, M.W., Pihalja, M., Weissman, I.L.,
Morrison, S.J. and Clarke, M.F. (2003) Nature (London) 423, 302–305
20 Jaiswal, S., Traver, D., Miyamoto, T., Akashi, K., Lagasse, E. and
Weissman, I.L. (2003) Proc. Natl. Acad. Sci. U.S.A. 100, 10002–10007
21 Brown, D., Kogan, S., Lagasse, E., Weissman, I., Alcalay, M., Pelicci, P.G.,
Atwater, S. and Bishop, J.M. (1997) Proc. Natl. Acad. Sci. U.S.A. 94,
2551–2556
22 Cozzio, A., Passegue, E., Ayton, P.M., Karsunky, H., Cleary, M.L. and
Weissman, I.L. (2003) Genes Dev. 24, 3029–3035
23 Huntly, B.J., Shigenatsu, H., Deguchi, K., Lee, B.H., Mizuno, S., Duclos, N.,
Rowan, R., Amaral, S., Curley, D., Williams, I.R. et al. (2004) Cancer Cell
6, 587–596
24 So, C.W., Karsunky, H., Passegue, E., Cozzio, A., Weissman, I.L. and
Cleary, M.L. (2003) Cancer Cell 3, 161–171
Received 17 June 2005
C 2005
Biochemical Society
1533