Cancer Stem Cells 4

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Oncogene

https://doi.org/10.1038/s41388-019-1128-4

REVIEW ARTICLE

Tumor propagating cells: drivers of tumor plasticity, heterogeneity,


and recurrence
Alexandre Teixeira Vessoni1 Eduardo Cremonese Filippi-Chiela2 Guido Lenz3
● ● ●

Luis Francisco Zirnberger Batista1,4,5

Received: 15 August 2019 / Revised: 15 November 2019 / Accepted: 20 November 2019


© The Author(s), under exclusive licence to Springer Nature Limited 2019

Abstract
Tumorigenesis is associated with the development of a highly variable pattern of cellular diversity, consequence of genetic
and epigenetic diversification, followed by clonal selection and expansion. This process is shaped by the microenvironment
and leads to intratumoral heterogeneity, which is characterized by differences between cancer cells in terms of gene
expression, phenotypic markers, growth dynamics, and resistance to treatment. Another relevant aspect in intratumor
heterogeneity is cell plasticity—the ability of a cell to switch to new identities. In this review, we focus on the mechanisms
1234567890();,:
1234567890();,:

that regulate cancer cell plasticity within a tumor, and explore the concept of tumor propagating cells, or TPCs, a cancer cell
able to propagate/phenocopy the parental tumor and recapitulate tumor heterogeneity. We discuss the influence of the
microenvironment and driver mutations on TPCs formation and function, the existence of phenotypically distinct TPC
clones within a tumor, the evolution of TPCs with disease progression, and their implications for therapy.

Introduction microenvironment [1]. Different models have been pro-


posed to explain how intratumor heterogeneity arises from a
A tumor is a complex admixture of phenotypically and a single cancer-initiating cell (CIC). Solid evidence supports
genetically distinct populations of transformed cells whose a non-linear, branched pattern of evolution, where cancer
behavior, including growth and resistance to therapy, is cells share genetic alterations that occurred earlier in tumor
shaped by the multiple interactions between them, the history, followed by a gradual accumulation of new driver
extracellular matrix, and with other cells in the tumor mutations at subclonal populations [2–11]. Evidence also
supports a punctuated form of evolution, where a large
number of genomic aberrations occur in short bursts of
time, generating high intratumor heterogeneity at very early
* Alexandre Teixeira Vessoni stages of cancer progression [12–15]. Epigenetic differences
alexandre.vessoni@wustl.edu add an additional layer of complexity to intratumor het-
* Luis Francisco Zirnberger Batista erogeneity as they can induce significant alterations in the
lbatista@wustl.edu
gene expression landscape within a tumor [16–19].
1
Accordingly, gene expression profiles associated with
Department of Medicine, Washington University School of
positive or negative prognosis can be detected in different
Medicine, St. Louis, MO 63110, USA
2
regions of the same tumor [3]. In this sense, a single biopsy
Departmento de Ciências Morfológicas, Instituto de Ciências
is likely to underestimate tumor complexity, leading to
Básicas da Saúde, Universidade Federal do Rio Grande do Sul,
Porto Alegre, RS, Brazil difficulties in validating oncogenic markers and in predict-
3 ing disease relapse [3]. Comprehending intratumor hetero-
Departamento de Biofísica e Centro de Biotecnologia,
Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, geneity is thus critical to the design of more effective
Brazil therapies.
4
Department of Developmental Biology, Washington University In addition to subclonal differences, another critical
School of Medicine, St. Louis, MO 63110, USA contributor to intratumor heterogeneity is cell plasticity,
5
Center for Regenerative Medicine, Washington University School i.e., the ability of a given cell to change its identity and
of Medicine, St. Louis, MO 63110, USA acquire characteristics of other cell types. In this sense, two
A. T. Vessoni et al.

models have been proposed to explain how a single cancer patient and show biologically distinct growth properties
cell can give rise to heterogenous progenies: (A) the sto- [9, 24]. In addition, CSCs do not necessarily originate from
chastic (or clonal evolution) and (B) the cancer stem cell a tissue stem cell, and tumorigenicity is not exclusively
(or hierarchical) models. In the stochastic model, all tumor found in cancer cells that express stem cell markers [25–
cells are equipotent, i.e., they all can self-renew and give 30]. Therefore, the term “tumor propagating cells” (TPCs)
rise to distinct progenies, and their behavior is stochasti- [29, 31, 32] will be used hereafter to refer to cancer cells
cally determined by intrinsic and extrinsic variables. In the responsible for tumor development and able to propagate
cancer stem cell (CSC) model, only a fraction of the tumor cancer upon serial transplantation assays. Tumors formed
cells, termed CSCs, are endowed with self-renewal poten- by TPCs are histologically heterogeneous and recapitulate
tial and able to give rise to phenotypically diverse cells, critical histopathological features of the original tumor, such
thus sustaining tumor growth in a defined hierarchical as infiltration/migration and multilineage differentiation.
mode. In this model, only CSCs give rise to serially Although some authors also refer to these cells as CICs, we
transplantable tumors that phenocopy the parental tumor in reserve this term to the precancerous cell in which the
xenotransplantation assays using immunocompromised crucial hit that initiates cancer development occurred
animals [20, 21] (Fig. 1). [31, 32]. In this review, we consider CSCs a specific case in
However, as we will explore in detail in the following which TPCs display stem cell markers, as opposed to the
sections, the stochastic and CSC models are not necessarily existence of cancer cells able to propagate heterogeneous
mutually exclusive, since: (i) cancer cells can interconvert tumors and that lack the expression of such markers (more
between states with high and low expression of tumor- details in the following section). Please, refer to the glossary
igenicity markers [16, 22, 23]; and (ii) genetically distinct for more information regarding TPC, CSC, and CIC
CSCs that share a common ancestral may exist in the same definitions.

Fig. 1 The stochastic,


hierarchical, and dynamic
models of tumor propagation. In
the stochastic model, all cancer
cells are equipotent in their
ability to propagate a tumor that
phenocopies the heterogeneity
of the primary tumor. In the
hierarchical model, only a subset
of cancer cells (also known as
cancer stem cells) are endowed
with the ability to propagate the
tumor. In the dynamic model,
stochastic state transitions allow
cancer cells to interchange
between states with high or null/
low tumor propagation capacity.
These state transitions are
influenced by the
microenvironment, genetic/
epigenetic signature, and
metabolism
Tumor propagating cells: drivers of tumor plasticity, heterogeneity, and recurrence

Assessing tumorigenic potential relies on observing long-term maintenance and recurrence of different tumor
tumor formation in vivo. The inability of one given cancer types [51–53]. Those evidences provided support to the idea
cell to induce tumor upon animal transplantation does not that CSCs and TPCs were one and the same. However,
necessarily mean that it lacks tumorigenic potential. It can evidence shows that the early progeny of stem cells, the
result from an inability to adapt to the host and/or survive progenitor cells, can also act as TPCs and sustain tumor
upon certain conditions that do not match the original niche growth. For instance, during the late stage (blast crisis) of
where the tumor was initially formed. Thus, these caveats human chronic myeloid leukemia associated to BCR-ABL
pose a hurdle to provide a definitive proof for the lack of oncogenic protein, granulocytic/monocytic-restricted pro-
tumorigenicity in any given cancer cell. That said, the use of genitors can acquire self-renewal potential and the ability to
cell surface markers to segregate and compare tumorigenic propagate leukemia through increased β-catenin activation
potential between different cancer cells may be subject to [54]. Progenitor-like TPCs were also described in solid
these hurdles. As it will be discussed in the following tumors. For instance, in mouse models of medulloblastoma,
sections, under specific circumstances, changing transplan- the TPC population was restricted to immature granule
tation conditions revealed high tumorigenic potential in neuron precursor-like cells that do not express the stem cell
unappreciated cancer cell populations. Despite these con- marker CD133 and cannot form neurospheres (an in vitro
siderations, the use of cell surface markers to differentiate test commonly used to detect stem cell potential), but
TPCs from non-TPCs is still the most direct (and express the progenitor cell marker Math1 [27]. Likewise, in
approachable) method to compare the tumorigenicity a mouse model of high-grade glioma, Id1low tumor cells,
between different cancer cells derived from a tumor. which have progenitor cells properties and limited self-
Therefore, we consider the use of surface markers to dis- renewal capacity, display higher tumorigenicity than tumor
tinguish TPCs from non-TPCs in this review, although the cells with higher self-renewal capacity and elevated
true TPC character can only be assessed by the growth of a expression of the neural stem cell marker Id1 [28].
tumor, as stated previously. Collectively, these findings show that cancer cells with
TPCs display several strategies that increase their resis- high tumorigenic potential are not necessarily those that
tance to therapy [33, 34] including increased checkpoint display tissue stem cell markers and properties. In fact,
activation and DNA repair capacity [35], enhanced anti- under specific circumstances, high stem-cell marker
oxidant defenses [36], autocrine production of anti- expression can be a poor indicator of the tumor propagating
apoptotic molecules [37], and expression of multidrug- potential of a cancer cell [28]. Therefore, the term TPC can
resistance efflux pumps [38–40]. In addition, evidences be more generally used to describe cancer cells able to
suggest that the TPC phenotype is a dynamic state that propagate cancer regardless of the presence of stem cell
could be acquired by non-TPC tumor cells via state tran- markers.
sition [23, 41–43]. In this review, we discuss (i) the phe-
notypic heterogeneity of TPC clones in solid tumors and
leukemias; (ii) how TPCs evolve with disease progression; Reversible state transitions and the changes
(iii) evidence of state transition enabling acquisition of high in tumorigenic potential of cancer cells
tumorigenic potential; (iv) the role of the tumor micro-
environment in this process; (v) methodological challenges TPCs can give rise to tumors containing cancer cells with
associated to the study of TPCs, and finally, (vi) the lower/null tumorigenic potential [44, 47–50]. Interestingly,
implications of TPCs for the development of improved evidence also suggests the opposite way: that cancer cells
therapies. can interconvert between a low tumorigenic/non-TPC state
to a TPC state [23, 41, 42]. For instance, when basal,
luminal, and stem cells from breast cancer cell lines are
Tumorigenic potential in cancer cells lacking segregated and cultured separately in vitro, they all return to
expression of stem cell markers equilibrium cell-state proportion over time via stochastic
state transitions [41]. More recently, the use of single cell
TPCs were first identified in acute myeloid leukemia (AML) RNA sequencing and barcoding revealed the potential of
as a rare population of adult hematopoietic cells that one single glioblastoma cell to generate the four states
retained a stem cell signature profile (CD34+/CD38−) [44]. (neural-progenitor-, oligodendrocyte-progenitor-, astrocyte-
This initial finding was followed by the identification of , and mesenchymal-like) observed in this malignancy [55].
stem-like TPCs in several human solid tumors, including The ability of cells to transit between different states can be
breast [45], brain [46, 47], ovarian [48], and colon [49, 50]. significantly lowered via activation of oncogenes and/or
In vivo lineage-tracing assays later provided evidence that inactivation of tumor suppressor genes. For instance, pri-
TPCs with stem cell signatures were responsible for the mary human mammary epithelial cells immortalized with
A. T. Vessoni et al.

human telomerase can spontaneously convert from a relatively simple (i.e., presence of two to three clones that
CD44low (differentiated) to a CD44high (stem cell) pheno- evolved in a linear sequence) or very complex (presence of
type [23]. Upon transformation with SV40 and HRAS up to ten clones related via a branching evolutionary pat-
expression, the conversion of CD44low cells (low tumori- tern). In addition, the clonal architecture at relapse could
genic) to CD44high (highly tumorigenic) phenotype is vary greatly from that at diagnosis [9, 67, 69, 76, 77].
greatly increased [23]. Similarly, tumorigenic p53KO Intraclonal heterogeneity of TPCs also occurs in solid
fibroblasts expressing Kras-G12D can convert from a less tumors such as glioblastoma and colo-rectal cancer
tumorigenic/more differentiated (Thy1+) to a more [65, 66, 78, 79], and the phenotypic diversity among TPCs
tumorigenic/less mature (Thy1−/Sca1−) phenotype. An can also arise between genetically identical multiple TPCs
increase in oncogenic signaling with Myc overexpression lacking a hierarchical organization between them and dif-
accentuates this process [42]. fering in their cell cycle/proliferation abilities [68].
In addition, evidence linking oncogenic signaling to In the last few years, single cell analysis started to shed
dedifferentiation and tumorigenesis was also described in more light on the extremely diverse genetics and types of
mouse models of intestinal [30], breast [56], and squamous cells that cause cancer. For instance: single cell tran-
cell carcinoma [57]. In the intestine, the Wnt pathway and scriptomics revealed that subclasses of IDH-mutant glioma
oncogenic KRAS-G12D have been shown to cooperate to share an architecture composed of malignant cells that
dedifferentiate villus cells into crypt stem cells (LGR5+) and recapitulate glial and neural precursor programs but differ
initiate tumorigenesis [30]. In the skin, cancer cells can considerably in microglia and macrophages (tumor micro-
hijack a “lineage infidelity” mechanism that can take place environment cells) signature [80]. Single cell tran-
during normal processes of wound repair via HRAS/MAPK scriptomics also allowed the identitication of a
signaling [57]. In breast tissue, expression of the oncogenic subpopulation of cells (primitive oligodendrocyte pro-
mutant PIK3CA-H1047R conferred multipotency to uni- genitor cells) that drive gliomagenesis, and also revealed the
potent mammary cells and ability to induce tumors in mouse, role played by a RNA-binding protein implicated in oligo-
which could be further potentiated by P53 ablation [56]. dendrocyte fate specification (Zfp36l1) in this process [81].
At last, activation of Epithelial-to-Mesenchymal Transi- In addition, recent work with single cell lineage tracing
tion (EMT), a critical developmental program hijacked by using endogenous mitochondrial DNA variants in ATAC-
cancer cells that often allows them to disseminate and seq provides additional evidence that the human pre-
metastasize [1], can induce the acquisition of TPC traits by leukemic hematopoietic stem cell population is hetero-
non-TPC cancer cells [16, 22, 58], and BRCA1 may be genous, and that one subclone evolves to become the
involved in suppressing this process in mammary cells [59]. leukemia propagating cell in AML [73].
Combined, the evidence discussed above shows that cells Collectively, these data indicate that genetically distinct
with low tumorigenic potential can transit to a state of TPC clones with biologically distinct growth properties can
greater tumorigenicity in a dynamic manner. It also indi- exist in individual cancer samples, and that genetically
cates that the accumulation of genetic lesions that inactivate identical TPCs can interchange between phenotypically
tumor suppressor genes and/or that activate oncogene sig- diverse states. Use of single cell analysis will allow to
naling during disease progression can reduce the barrier for increase our understanding of this complexity, and its
a non-tumorigenic cancer cell to become tumorigenic, clinical role in limiting clinical effectiveness and con-
which could account for the higher frequency of TPCs in tributing to tumor relapse.
late-stage cancers [24, 60–62].

Microenvironment and cancer therapy


Phenotypically diverse clones of TPCs regulate TPCs generation and function
contribute to intratumor heterogeneity
Apart from genetic diversity, a plethora of microenviron-
Evidence supports the existence of multiple TPCs popula- mental stimuli contributes to intratumor heterogeneity by
tions in individual cancer samples [9, 24, 63–70]. In leu- regulating TPC generation and function. In this sense,
kemia, multiple -and genetically distinct- leukemia paracrine and juxtracrine signaling derived from stroma
propagating cell clones with different engraftment capacity cells, and changes in oxygen and nitric oxide levels are
were identified [9, 11, 24, 71–75]. Subclones display unique some of the key players in the microenvironment–cancer
genetic alterations, yet shared copy number alterations with interaction. Some of these relations are depicted below and
the dominant clone at diagnosis, showing that these sub- illustrated in Fig. 2.
clones evolved from the initial diagnostic clone [11, 24, 71]. Immune cells, which are known to play a large and
The genetic architecture of the TPC population can be central role in tumor progression and metastasis [82–85],
Tumor propagating cells: drivers of tumor plasticity, heterogeneity, and recurrence

Fig. 2 The influence of the microenvironment on the main pathways cells), and secretion of cytokines that maintain the TPC state. In lung
regulating acquisition and maintenance of tumor propagating potential and colon cancer, the TPC state is regulated by milk fat globule-EGF
in different cancer cells. Several pathways are depicted in this figure. factor 8 protein (MFG-E8) secreted by macrophages. In addition,
In gliomas, the TPC state is regulated by the Notch pathway (via nitric colon cancer TPCs can be sustained by the activation of β-catenin/Wnt
oxide secreted by endothelial cells); by the Protein Tyrosine Phos- pathway by hepatocyte growth factor (HGF) secreted by myofibro-
phatase Receptor Type Z1 (PTPRZ1), whose activation is driven by blasts. Colorectal cancer TPCs are sustained by NF-κB activation (via
Pleiotropin secreted by macrophages; and by signal transducer and Interleukin-17A (IL-17A) secreted by cancer associated fibroblasts, as
activator of transcription 3 (STAT3) and Hedgehog pathways. In well as by the STAT3 and Hedgehog pathways. In squamous cell
addition, glioma TPCs can differentiate into pericytes that support a carcinoma, the TPC state is regulated by transforming growth factor-β
pro-glioma vasculature. In breast cancer cells, the TPC state is regu- (TGF-β) secreted by myeloid cells, which binds to its receptor (TbRI/
lated by ADAM Metallopeptidase Domain 10 (ADAM10)-containing II) and activates Mothers Against DPP Homolog 2 (SMAD2) in the
exosomes, which are secreted by fibroblasts and activate the Notch and cancer cell. Illustrated in this figure is also the ability of TPCs to
RhoA (Ras homolog gene family, member A) pathway in the cancer activate regulatory T-cells (T-reg) via Interleukin 4 (IL4), C-C Motif
cells; and by monocytes/macrophages that physically interact with and Chemokine Ligand 5 (CCL5), and STAT3, thus inhibiting T-cells’
activate the EPH receptor A4 (EphA4), resulting in the activation of proliferation and activation, and promoting tumor immune tolerance
NF-κB (Nuclear Factor kappa-light-chain-enhancer of activated B

can engage in a complex crosstalk with TPCs. For instance, of mouse squamous cell carcinoma TPC via secretion of
tumor-associated macrophages can support glioma TPCs transforming growth factor-β (TGF- β) and triggering of
through activation of the Protein Tyrosine Phosphatase EMT features, while TPCs that are distant to the peri-
Receptor Type Z1 (PTPRZ1) receptor via secretion of vasculature adopt a faster cell cycle rate associated with
pleiotropin [86]. Monocytes/macrophages can also physi- tumor growth and differentiation [43]. TGF-β activation can
cally interact with and activate the EPH receptor A4 also boost the resistance of TPCs to cisplatin via activation
(EphA4) in breast TPCs [87], resulting in the activation of of glutathione metabolism genes [43]. Finally, TPCs display
Nuclear Factor kappa-light-chain-enhancer of activated B immunomodulatory features—for instance, TPCs can acti-
cells (NF-κB) and secretion of cytokines that maintain the vate regulatory T-cells (T-reg) and inhibit T-cells' pro-
TPC state [87]. Similarly, lung and colon TPCs can induce liferation and activation, thereby promoting tumor immune
the secretion of milk fat globule-EGF factor 8 protein tolerance [89–91].
(MFG-E8) by macrophages, which, in turn, increases the In addition to immune cells, cancer-associated fibroblasts
tumorigenicity and the resistance of TPCs to therapy [88]. can also regulate TPC function and their metastatic poten-
Myeloid cells at perivascular sites can enhance invasiveness tial. Exosomes (cell-derived vesicles that transfer DNA,
A. T. Vessoni et al.

RNA, and proteins to other cells; [92]) derived from (HIF1A) and HIF2A [103], while radiation induced repro-
fibroblasts containing ADAM Metallopeptidase Domain 10 gramming of breast cancer cells into breast TPCs via acti-
(ADAM10) activate Notch and Ras homolog gene family, vation of the Notch pathway [104].
member A (RhoA) in breast cancer cells, resulting in Finally, recent data showed a connection between neu-
increased cell motility and expression of TPC markers [93]. ronal cell activity and glioma growth and invasion as well as
Cancer-associated fibroblasts activated by chemotherapy breast cancer metastasis to the brain [105–107]. In these
can also secrete Interleukin-17A (IL-17A), which augments reports, glutamate released by neurons were able to activate
the tumorigenic potential, growth rates, migration, and glutamate receptors expressed in the membrane of the cancer
chemotherapy resistance of colorectal cancer TPCs in a NF- cells, supporting tumor cell proliferation and/or metastasis.
κB-dependent manner [94]. Hepatocyte growth factor Therefore, combined with other studies [27, 60, 68, 87],
derived from myofibroblasts can also induce a TPC phe- data described in this section reveal that TPCs regulation
notype in more differentiated colon cancer cells, causing heavily depends on their interaction with the micro-
these cells to re-express TPC-associated markers such as environment, which provides a plethora of inputs that are
activation of β-catenin/Wnt pathway [95]. Similarly, mye- critical to sustain their identity. Decoding the information
lodysplastic syndrome cells can remodel their micro- exchanged by TPCs and their surrounding microenviron-
environment by instructing normal mesenchymal stem cells ment might help develop new therapies aiming at differ-
to acquire a disease-associated phenotype that secrete fac- entiating and “locking” cancer cells in a non-TPC stage.
tors such as LIF, VEGFA, IGFBP2, and ANGPTL4, sup-
porting engraftment and expansion of the myelodysplastic
propagating cells [96]. Metabolism of TPCs
Another important component of the TPC regulation by
the microenvironment is the vasculature. Highly vascularized Cancer cells often display an altered energy metabolism that
brain tumors have higher incidence of CD133+/Nestin+ cells favors glycolysis over oxidative phosphorylation even
closely associated to the tumor capillaries [97], suggesting under aerobic conditions, the so-called Warburg effect
that the perivascular niche supports cells that have TPC [108, 109]. This is associated with a fast proliferation rate,
markers. In agreement, co-injection of primary human endo- as glycolytic intermediates can be diverted into amino acid,
thelial cells with medulloblastoma cells accelerated tumor nucleotide, and lipid biosynthesis routes required for cell
growth and increased the amount of TPCs in the resulting proliferation [108, 109]. While the Warburg effect is well-
tumor, while pharmacological ablation of tumor vasculature established in tumors, recent evidence shows that the
with Erlotinib and Bevacizumab caused tumor arrest and metabolism of cancer cells is often more complex. For
complete loss of CD133+/Nestin+ tumor cells [97]. The instance, lactate secreted by a population of cancer cells
regulation of TPCs by endothelial cells involves the secretion under the Warburg effect can be used by another population
of soluble factors such as nitric oxide [98, 99]. Interestingly, of cancer cells in oxygenated areas of the same tumor to
in a glioblastoma xenograft, endothelial cells were able to fuel their oxidative metabolism [110]. Significant metabolic
induce the differentiation of the glioma TPCs into pericytes to differences may also exist between primary and metastatic
support vasculature formation and tumor growth [100]. tumor sites, as in the case of pancreatic cancer, where dis-
Molecules released by dying cancer cells can also impact tant metastasis are dependent on the oxidative branch of the
the TPC population of the tumor. Upon chemotherapy, pentose phosphate pathway to support an epigenetic sig-
dying cancer cells of a xenograft bladder tumor release nature that drives their tumorigenicity [111]. In this sense, it
PGE2, inducing the mobilization of a small subpopulation is interesting to consider if the metabolism of TPCs differs
of quiescent cytokeratin 14-positive cells (CK14+), which from non-TPC cancer cells, and how relevant this is for
are highly tumorigenic and resistant to therapy. Blocking their maintenance and function (Fig. 3).
PGE2 release prevented the expansion of these TPCs and A growing body of evidence indicates that TPCs from
reduced lung metastasis [101]. Also, TPCs can protect the different cancer types display increased dependence on
non-TPC population from anoikis via a bystander effect mitochondria oxidative phosphorylation (OXPHOS), when
[102], revealing a cooperatively crosstalk between these compared with the more differentiated/non-TPCs cells of
different cancer cell types. the same tumor. This metabolic configuration is critical for
Evidence also shows that commonly used chemother- their maintenance and tumor growth [112–120]. However,
apeutic agents and radiotherapy can accelerate the inter- subpopulations of TPCs that are metabolically heterogenous
conversion between non-TPC to TPC state. Temozolomide, have also been described. For instance, a subset of TPCs
an alkylating agent used in the treatment of glioblastoma, (CD36+) were found to reside in the gonadal adipose tissue
drives the acquisition of TPC characteristics by non-TPC of a mouse model of blast-crisis chronic myeloid leukemia.
glioma cells, possibly via hypoxia-inducing factor 1-alpha These cells are able to co-opt neighboring adipocytes to
Tumor propagating cells: drivers of tumor plasticity, heterogeneity, and recurrence

Fig. 3 Metabolism in the control of the acquisition and maintenance of telangiectasia mutated (ATM), thus supporting tumorigenic potential
tumor propagating potential in cancer cells. Top: Cytochrome C and in different cancer cells. Reactive oxygen species produced by mito-
Endonuclease G (EndoG) released by mitochondria to the cytosol chondria oxidative phosphorylation (OXPHOS) can also sustain breast
cause activation of caspase-activated DNAse and formation of geno- cancer TPCs via activation of the hypoxia inducible factor 1α (HIF-1α)
mic DNA double strand breaks, which in turn activate Nuclear Factor pathway. Bottom: mitophagy sustains TPCs in liver and colorectal
kappa-light-chain-enhancer of activated B cells (NF-KB) and signal cancer cells via elimination of PTEN-induced kinase 1 (PINK1), thus
transducer and activator of transcription 3 (Stat3) pathways via ataxia preventing P53-mediated inhibition of NANOG transcription

release free fatty acids and support the TPCs’ metabolic preventing its translocation to the nucleus where it binds
demands and survival [121]. and represses activation of the NANOG promoter, a gene
Mitochondria can also be involved in an intricate reg- that sustains TPC phenotype in these cancer cells [123]. In
ulation of tumorigenic potential in different cancer cell lines ovarian TPCs, autophagy sustains tumorigenicity and pro-
that involves self-inflicting DNA damage [122]. In this tects these cells from chemotherapy-induced death, possibly
sense, mitochondria releases cytochrome C and Endonu- via elimination of reactive oxygen species-producing
clease G (EndoG) to the cytosol, causing activation of mitochondria and/or by supporting metabolism during
caspase-activated DNAse and formation of genomic DNA nutrient/oxygen starvation [125].
double strand breaks. In response to this damage, NF-KB Altogether, these studies highlight the important and the
and Signal transducer and activator of transcription 3 (Stat3) diverse role played by mitochondria and OXPHOS in the
pathways are activated (via ataxia telangiectasia mutated, or regulation of TPCs. Metabolic heterogeneity may also occur
ATM), supporting tumorigenic potential in cancer cells between TPCs of different niches from the same tumor, and
[122]. Reactive oxygen species produced by mitochondria these differences may account for therapy resistance and
OXPHOS can also sustain breast cancer TPCs via activation tumor relapse [112], highligthing the relevance of the
of the hypoxia inducible factor 1α (HIF-1α) pathway [116]. metabolic regulation of TPCs on cancer development and
Interestingly, while in breast cancer MYC positively reg- therapy response.
ulates OXPHOS and TPCs maintenance, in pancreatic
cancer MYC decreased OXPHOS and reduced stemness
properties of TPCs [112], revealing a complex regulation of Methodology challenges to study TPCs
metabolism by MYC in different cancers. in vivo and in in vitro
Intriguingly, elimination of mitochondria via mitophagy
(an autophagy-mediated pathway for mitochondria recy- Understanding methodology limitations and patient-to-
cling) sustains TPCs from liver and colorectal cancer patient variations in TPCs populations is critical. In this
[123, 124]. In the case of liver TPCs, mitophagy eliminates section, we highlight important studies that brought con-
the mitochondria-associated PTEN-induced kinase 1 siderable improvement in the methodologies used to study
(PINK1) that can phosphorylate and activate P53, thus TPCs, as well as important considerations regarding TPC
A. T. Vessoni et al.

heterogeneity that arises during disease progression, and metastasis) or in samples passaged in xenografts, virtually
during in vitro and in vivo propagation of these cells. every tumor cell, regardless of its immunophenotype, seems
Xenotransplantation assays are the most stringent to have tumorigenic potential [61, 62, 128]. These results
method to detect and test TPC potential [21]. However, it suggest a continuous malignant progression of tumor cells
comes with caveats, as to whether the assay selects a with disease progression and/or xenograft growth, arguing
population of human cancer cells able to readily engraft and caution in the comparison of samples obtained from dif-
grow in a foreign and immune-compromised environment ferent patients, at different stages of the disease, and/or
[126]. As mentioned before, the inability of a cancer cell to propagated in xenografts.
grow a tumor ex vivo may not be a consequence of lack of Isolation and propagation of cancer cells in vitro is an
tumorigenicity, but a failure of the cell to adapt to certain essential tool for disease modeling and drug discovery.
conditions that do not perfectly recapitulate the environment Still, cancer cells may adapt to these artificial conditions,
where the tumor was originally formed. For instance, with concomitant loss of characteristics relevant for the
xenotransplantation of human acute myeloid leukemia disease in vivo. For instance, fetal bovine serum (FBS),
samples into a severe combined immunodeficient (SCID) routinely used to grow cancer cell lines in vitro, can
mouse revealed very low TPC frequency (1 in every severely impair growth and maintenance of certain TPCs,
250.000 transplanted cells) [44], while the transplantation of such as those derived from human glioblastoma [130]. In
mouse lymphoma or mouse leukemia cells into non-irra- this case, the growth of freshly isolated cells in serum-free
diated, congenic animals, showed that as few as ten neural basal media, which is used for the propagation of
unsorted cells were enough to induce malignancy that neural stem cells, preserved the ability of these cultured
recapitulated features of the primary tumor [29]. cells to give rise to highliy infiltrative tumors that phe-
A second caveat refers to the methods used to isolate and nocopied key characteristics of the parental tumor. On the
transplant TPCs, and the degree of immunodeficiency of the other hand, cells grown in the presence of FBS acquired
host. For instance, anti-CD38 antibodies used to sort human several genetic and/or epigenetic alterations and could only
leukemia TPCs had an inhibitory effect on the engraftment form intracranial tumors with very low infiltrative capacity
of these cells as it targeted them to clearance by the innate [130]. The serum-free neural basal media, however, does
immune cells [127]. Combining the administration of not seem adequate to the in vitro growth of every type of
immunosuppressants with intrabone marrow injection of the glioma TPCs, as it compromised the tumorigenic potential
CD38-coated cells revealed the presence of TPCs in pre- of oligodendrocyte progenitor (OPC)-like mouse glioma
viously unappreciated CD34+/CD38+ populations in some cells [131, 132]. As different media containing single or a
cases of AML [127]. Another important development was few growth factors were traditionally designed based on the
the use of highly immunocompromised mice (IL2rg−/− putative cell of origin of gliomas, they are prone to select
versus NOD/SCID) and the co-injection of cancer cells with subpopulations which would otherwise be reduced either
matrigel in xenotransplantation assays, which increased, by in vivo or by a media containing a different combination of
orders of magnitude, the frequency of melanoma TPCs, growth factors [132].
reaching up to one in every every unsorted melanoma cells Although serum can exert a drastic effect on the main-
derived from patients [61, 128]. tenance of glioma TPCs in vitro, it does not affect TPCs
A third caveat stems from the fact that the TPC pheno- from other cancers in the same manner. In the case of
type is not necessarily uniform across patients [129]. melanoma, for instance, cell lines grown in media supple-
Moreover, TPC frequency increases with disease progres- mented with FBS were still able to engraft at 100% effi-
sion/malignancy [24, 60–62, 80]. For instance, in primary ciency when only very few unsorted cells were transplanted
serous ovarian cancer (SOC), not only TPC frequency [62]. Combined, this indicates that the in vitro growth of
varies up to 100 times between patients, but also their TPCs obtained from different tissues requires unique culture
immunophenotype is different: CD133+ marks TPCs in conditions.
some patient’s samples, while in other samples TPCs are
CD133−; in other cases, CD133 status does not seem to be
related to the TPC phenotype of cells [129]. Also, the Future perspectives: the therapeutic
growth of tumor samples in xenograft may change TPCs challenges to target TPCs
phenotype. In this sense, matched primary SOC tumors and
xenografts showed opposite TPC phenotypes regarding TPCs are influenced by a plethora of factors, including
CD133 expression [129]. In the same line, it was shown in interaction with the tumor microenvironment, acquisition of
freshly obtained samples of melanoma from different stages cancer-associated mutations, and disease stage [22, 23, 60].
of the disease that the CD271+ fraction of cells is enriched Understanding how these variables regulate TPCs dynamics
for TPCs whereas in late stage samples (lymph node may reveal new therapeutic opportunities.
Tumor propagating cells: drivers of tumor plasticity, heterogeneity, and recurrence

As evidence suggests that non-TPC cancer cells may single cell resolution will be critical for the success of such
change their phenotype and acquire tumorigenic potential therapeutic strategies.
[23, 41, 42], targeting only TPCs may not be fruitful ther-
apeutically. One potential strategy is to try to differentiate and
“lock” cancer cells in a non-TPC state incapable of propa- Glossary
gating the tumor [16, 80, 133]. Such strategies may be
achieved pharmacologically [134], genetically [81, 135–139], Tumor propagating cells (TPCs)
or by modulating microenvironment cues [17, 43, 68, 98,
133, 134]. This strategy, however, needs to take into con- Cancer cells responsible for tumor development and able to
sideration the genetic and functional heterogeneity between propagate cancer upon serial xenotransplantation assays.
distinct TPC subpopulations [9, 24, 65–67], as well as The tumor formed by TPCs is histologically heterogeneous
changes in clonal architecture during treatment and relapse and phenocopies critical histopathological features of the
[134]. In this sense, spatially resolved omics analysis original tumor, such as infiltration/migration and multi-
[140, 141] and RNA sequencing at the single-cell [80] level lineage differentiation (including into non-TPCs). TPCs
may provide personalized, deeper details regarding shared may or may not express markers that are characteristic of
and/or exclusive pathways that sustain tumorigenic potential normal stem cells.
in distinct TPC subpopulations.
Therapeutic approaches that exclusively target TPCs, Cancer stem cells (CSC)
but not normal stem/progenitor cells, would potentially
lower toxicity for patients. For instance, HIF2A is pre- A specific case in which TPCs share some characteristics
ferentially expressed in glioblastoma TPCs with stem cell with normal stem cells (such as surface markers that are
characteristics, but not in normal brain stem cells, and is widely used to purify these cells). CSCs may have origi-
important for TPC proliferation, survival and tumor- nated from normal stem cells or acquired expression of stem
igenicity [142]. Similarly, leukemia propagating cells were cell markers during cancer progression.
much more sensitive to the HIF1A inhibitor echinomycin
than normal hematopoietic progenitor cells [143]. Target- Cancer initiating cell (CIC)
ing CD44 can impair engraftment and induce differentia-
tion of some leukemia propagating cells, while sparing Also known as tumor initiating cell, it is the cell of origin of
normal hematopoietic stem cells [144, 145]. In the same cancer, meaning the precancerous cell in which the earliest
line, leukemia propagating cells have increased expression and crucial hit that lead to the growth of cancer occurred.
of CD47 in comparison to normal HSCs, and the use of
anti-CD47 antibody preferentially induces phagocytosis of Cell plasticity
the cancerous counterparts [146]. Leukemia propagating
cells also heavily rely on OXPHOS for ATP production, in The ability of one cell to change its functional
contrast to normal HSCs [120], making the use of characteristics.
OXPHOS inhibitors, such as metformin [147], an attractive
approach. At last, the R882H mutated version of DNA Stem cells
methyltransferase 3A (DNMT3A, detected in 20–30% of
human acute leukemia), but not the wild type version of Unspecialized cells characterized by two main properties:
this gene, was found to activate expression of Mesi1, Mu1 self-renewal and the ability to differentiate into specialized
and HOXA1 genes that block differentiation and induce cell types.
self-renewal; this mutated DNMT3A cooperates with
NRAS mutation to induce leukemogenesis, but renders the Progenitor cells
TPCs sensitive to Dot1 inhibition, causing them to differ-
entiate and lose self-renewal potential [148]. Nevertheless, Early progeny of stem cells, able to differentiate into one or
it is important to decipher if a determined target pathway is more cell types. Progenitor cells are not able to self-renew
required for the maintenance of all the phenotypically indefinitely.
diverse TPCs of a given tumor, and if (and how) the
dependency of the TPCs on those pathways change with Acknowledgements ATV was supported by the Philip Majerus Fel-
lowship Fund. LFZB is supported by the NHLBI (4R00HL114732;
disease progression. Targeting TPCs is an attractive ther-
1R01HL137793) and grants from the V Foundation for Cancer
apeutic approach, and comprehension of TPCs’ evolution Research, the Edward Mallinckrodt Jr. Foundation, the AA&MDS
with disease progression/relapse, intratumor clonal diver- International Foundation, the CONCERN Foundation, the Department
sity, and their regulation by the microenvironment from a of Defense Bone Marrow Failure Research Program (BM160054) and
A. T. Vessoni et al.

the American Cancer Society. We thank Sapiens Scientific Illustrations 15. Hicks J, Krasnitz A, Lakshmi B, Navin NE, Riggs M, Leibu E,
for the artistic work on Figs. 1–3. et al. Novel patterns of genome rearrangement and their asso-
ciation with survival in breast cancer. Genome Res. 2006;16:
1465–79.
Compliance with ethical standards 16. Chaffer CL, Marjanovic ND, Lee T, Bell G, Kleer CG, Reinhardt
F, et al. Poised chromatin at the ZEB1 promoter enables breast
Conflict of interest The authors declare that they have no conflict of cancer cell plasticity and enhances tumorigenicity. Cell.
interest. 2013;154:61–74.
17. Roesch A, Fukunaga-Kalabis M, Schmidt EC, Zabierowski SE,
Publisher’s note Springer Nature remains neutral with regard to Brafford PA, Vultur A, et al. A temporarily distinct subpopula-
jurisdictional claims in published maps and institutional affiliations. tion of slow-cycling melanoma cells is required for continuous
tumor growth. Cell. 2010;141:583–94.
18. Sharma SV, Lee DY, Li B, Quinlan MP, Takahashi F, Mahes-
waran S, et al. A chromatin-mediated reversible drug-tolerant
References state in cancer cell subpopulations. Cell. 2010;141:69–80.
19. Li S, Garrett-Bakelman FE, Chung SS, Sanders MA, Hricik T,
1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next gen- Rapaport F, et al. Distinct evolution and dynamics of epigenetic
eration. Cell. 2011;144:646–74. and genetic heterogeneity in acute myeloid leukemia. Nat Med.
2. Nik-Zainal S, Van Loo P, Wedge DC, Alexandrov LB, Green- 2016;22:792–9.
man CD, Lau KW, et al. The life history of 21 breast cancers. 20. Shackleton M, Quintana E, Fearon ER, Morrison SJ. Hetero-
Cell. 2012;149:994–1007. geneity in cancer: cancer stem cells versus clonal evolution. Cell.
3. Gerlinger M, Rowan AJ, Stuart Horswell MM, Larkin J, 2009;138:822–9.
Endesfelder D, Gronroos E, et al. Intratumor heterogeneity and 21. Kreso A, Dick JE. Evolution of the cancer stem cell model. Cell
branched evolution revealed by multiregion sequencing marco. Stem Cell. 2014;14:275–91.
N. Engl J Med. 2012;366:883–92. 22. Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY,
4. Harbst K, Lauss M, Cirenajwis H, Isaksson K, Rosengren F, et al. The epithelial-mesenchymal transition generates cells with
Törngren T, et al. Multiregion whole-exome sequencing properties of stem cells. Cell. 2008;133:704–15.
uncovers the genetic evolution and mutational heterogeneity of 23. Chaffer CL, Brueckmann I, Scheel C, Kaestli AJ, Wiggins PA,
early-stage metastatic melanoma. Cancer Res. 2016;76: Rodrigues LO, et al. Normal and neoplastic nonstem cells can
4765–74. spontaneously convert to a stem-like state. Proc Natl Acad Sci
5. Kim TM, Jung SH, An CH, Lee SHSH, Baek IP, Kim MS. et al. USA. 2011;108:7950–5.
Subclonal genomic architectures of primary and metastatic col- 24. Notta F, Mullighan CG, Wang JCY, Poeppl A, Doulatov S,
orectal cancer based on intratumoral genetic heterogeneity. Clin Phillips LA, et al. Evolution of human BCR–ABL1 lympho-
Cancer Res. 2015;21:4461–72. blastic leukaemia-initiating cells. Nature. 2011;469:362–7.
6. Gerlinger M, Horswell S, Larkin J, Rowan AJ, Salm MP, Varela 25. Visvader JE. Cells of origin in cancer. Nature. 2011;469:314–22.
I, et al. Genomic architecture and evolution of clear cell renal cell 26. Cozzio A, Passegué E, Cleary ML, Weissman IL. Similar MLL-
carcinomas defined by multiregion sequencing. Nat Genet. associated leukemias arising from self-renewing stem cells and
2014;46:225–33. short-lived myeloid progenitors. Genes Dev. 2003;17:3029–35.
7. Sottoriva A, Spiteri I, Piccirillo SGM, Touloumis A, Collins VP, 27. Read TA, Fogarty MP, Markant SL, McLendon RE, Wei Z,
Marioni JC, et al. Intratumor heterogeneity in human glio- Ellison DW, et al. Identification of CD15 as a marker for tumor-
blastoma reflects cancer evolutionary dynamics. Proc Natl Acad propagating cells in a mouse model of medulloblastoma. Cancer
Sci USA. 2013;110:4009–14. Cell. 2009;15:135–47.
8. de Bruin EC, McGranahan N, Mitter R, Salm M, Wedge DC, 28. Barrett LE, Granot Z, Coker C, Iavarone A, Hambardzumyan D,
Yates L, et al. Spatial and temporal diversity in genomic Holland EC, et al. Self-renewal does not predict tumor growth
instability processes defines lung cancer evolution. Science. potential in mouse models of high-grade glioma. Cancer Cell.
2014;346:251–6. 2012;21:11–24.
9. Anderson K, Lutz C, van Delft FW, Bateman CM, Guo Y, 29. Kelly PN, Dakic A, Adams JM, Nutt SL, Strasser A. Tumor
Colman SM, et al. Genetic variegation of clonal architecture and growth need not be driven by rare cancer stem cells. Science.
propagating cells in leukaemia. Nature. 2011;469:356–61. 2007;317:337.
10. Xue R, Li R, Guo H, Guo L, Su Z, Ni X, et al. Variable intra- 30. Schwitalla S, Fingerle AA, Cammareri P, Nebelsiek T, Göktuna
tumor genomic heterogeneity of multiple lesions in patients with SI, Ziegler PK, et al. Intestinal tumorigenesis initiated by ded-
hepatocellular carcinoma. Gastroenterology. 2016;150:998–1008. ifferentiation and acquisition of stem-cell-like properties. Cell.
11. Hughes AEO, Magrini V, Demeter R, Miller CA, Fulton R, 2013;152:25–38.
Fulton LL et al. Clonal architecture of secondary acute myeloid 31. Hong D, Gupta R, Ancliff P, Atzberger A, Brown J, Soneji S,
leukemia defined by single-cell sequencing. PLoS Genet. et al. Initiating and cancer-propagating cells in TEL-AML1-
2014;10. https://doi.org/10.1371/journal.pgen.1004462. associated childhood leukemia. Science. 2008;319:336–9.
12. Sottoriva A, Kang H, Ma Z, Graham TA, Salomon MP, Zhao J, 32. Maenhaut C, Dumont JE, Roger PP, van Staveren WCG. Cancer
et al. A Big Bang model of human colorectal tumor growth. Nat stem cells: a reality, a myth, a fuzzy concept or a misnomer? An
Genet. 2015;47:209–16. analysis. Carcinogenesis. 2010;31:149–58.
13. Baca SC, Prandi D, Lawrence MS, Mosquera JM, Romanel A, 33. Li X, Lewis MT, Huang J, Gutierrez C, Osborne CK, Wu MF,
Drier Y, et al. Punctuated evolution of prostate cancer genomes. et al. Intrinsic resistance of tumorigenic breast cancer cells to
Cell. 2013;153:666–77. chemotherapy. J Natl Cancer Inst. 2008;100:672–9.
14. Stephens PJ, Greenman CD, Fu B, Yang F, Bignell GR, Mudie 34. Blazek ER, Foutch JL, Maki G. Daoy medulloblastoma cells that
LJ, et al. Massive genomic rearrangement acquired in a single express CD133 are radioresistant relative to CD133- cells, and
catastrophic event during cancer development. Cell. 2011;144: the CD133+ sector is enlarged by hypoxia. Int J Radiat Oncol
27–40. Biol Phys. 2007;67:1–5.
Tumor propagating cells: drivers of tumor plasticity, heterogeneity, and recurrence

35. Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, 55. Neftel C, Laffy J, Filbin MG, Hara T, Shore ME, Rahme GJ,
et al. Glioma stem cells promote radioresistance by preferential et al. An integrative model of cellular states, plasticity, and
activation of the DNA damage response. Nature. 2006;444: genetics for glioblastoma. Cell. 2019;178:835–49.
756–60. 56. Van Keymeulen A, Lee MY, Ousset M, Brohée S, Rorive S,
36. Diehn M, Cho RW, Lobo NA, Kalisky T, Dorie MJ, Kulp AN, Giraddi RR, et al. Reactivation of multipotency by oncogenic
et al. Association of reactive oxygen species levels and radio- PIK3CA induces breast tumour heterogeneity. Nature. 2015;525:
resistance in cancer stem cells. Nature. 2009;458:780–3. 119–23.
37. Todaro M, Alea MP, Di Stefano AB, Cammareri P, Vermeulen 57. Ge Y, Gomez NC, Adam RC, Nikolova M, Yang H, Verma A,
L, Iovino F, et al. Colon cancer stem cells dictate tumor growth et al. Stem cell lineage infidelity drives wound repair and cancer.
and resist cell death by production of interleukin-4. Cell Stem Cell. 2017;169:636–42.e14.
Cell. 2007;1:389–402. 58. Yang Y, Wang G, Zhu D, Huang Y, Luo Y, Su P, et al.
38. Patrawala L, Calhoun T, Schneider-Broussard R, Zhou J, Clay- Epithelial-mesenchymal transition and cancer stem cell-like
pool K, Tang DG. Side population is enriched in tumorigenic, phenotype induced by Twist1 contribute to acquired resistance
stem-like cancer cells, whereas ABCG2+ and ABCG2− cancer to irinotecan in colon cancer. Int J Oncol. 2017;51:515–24.
cells are similarly tumorigenic. Cancer Res. 2005;65:6207–19. 59. Bai F, Chan HL, Scott A, Smith MD, Fan C, Herschkowitz JI,
39. Wulf GG, Wang RY, Kuehnle I, Weidner D, Marini F, Brenner et al. BRCA1 suppresses epithelial-to-mesenchymal transition
MK, et al. A leukemic stem cell with intrinsic drug efflux and stem cell dedifferentiation during mammary and tumor
capacity in acute myeloid leukemia. Blood. 2001;98:1166–73. development. Cancer Res. 2014;74:6161–72.
40. Tehranchi R, Woll PS, Anderson K, Buza-Vidas N, Mizukami T, 60. Lapouge G, Beck B, Nassar D, Dubois C, Dekoninck S, Blan-
Mead AJ, et al. Persistent malignant stem cells in del(5q) mye- pain C. Skin squamous cell carcinoma propagating cells increase
lodysplasia in remission. N. Engl J Med. 2010;363:1025–37. with tumour progression and invasiveness. EMBO J. 2012;31:
41. Gupta PB, Fillmore CM, Jiang G, Shapira SD, Tao K, Kuper- 4563–75.
wasser C, et al. Stochastic state transitions give rise to pheno- 61. Quintana E, Shackleton M, Sabel MS, Fullen DR, Johnson TM,
typic equilibrium in populations of cancer cells. Cell. Morrison SJ. Efficient tumour formation by single human mel-
2011;146:633–44. anoma cells. Nature. 2008;456:593–8.
42. Ischenko I, Zhi J, Moll UM, Nemajerova A, Petrenko O. Direct 62. Boiko AD, Razorenova OV, van de Rijn M, Swetter SM,
reprogramming by oncogenic Ras and Myc. Proc Natl Acad Sci Johnson DL, Ly DP, et al. Human melanoma-initiating cells
USA. 2013;110:3937–42. express neural crest nerve growth factor receptor CD271. Nature.
43. Oshimori N, Oristian D, Fuchs E. TGF-β promotes heterogeneity 2010;466:133–7.
and drug resistance in squamous cell carcinoma. Cell. 2015;160: 63. Chen R, Nishimura MC, Bumbaca SM, Kharbanda S, Forrest
963–76. WF, Kasman IM, et al. A hierarchy of self-renewing tumor-
44. Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres- initiating cell types in glioblastoma. Cancer Cell. 2010;17:
Cortes J, et al. A cell initiating human acute myeloid leukaemia 362–75.
after transplantation into SCID mice. Nature. 1994;367:645–8. 64. Taussig DC, Vargaftig J, Miraki-moud F, Griessinger E, Shar-
45. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, rock K, Gribben JG, et al. Leukemia-initiating cells from some
Clarke MF. Prospective identification of tumorigenic breast acute myeloid leukemia patients with mutated nucleophosmin
cancer cells. Proc Natl Acad Sci USA. 2003;100:3983–8. reside in the CD34- fraction. Blood. 2010;115:1976–85.
46. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, 65. Piccirillo SGM, Colman S, Potter NE, Van Delft FW, Lillis S,
et al. Identification of human brain tumour initiating cells. Nat- Carnicer MJ, et al. Genetic and functional diversity of propa-
ure. 2004;432:396–401. gating cells in glioblastoma. Stem Cell Rep. 2015;4:7–15.
47. Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire 66. Giessler KM, Kleinheinz K, Huebschmann D, Balasubramanian
J, et al. Identification of a cancer stem cell in human brain GP, Dubash TD, Dieter SM, et al. Genetic subclone architecture
tumors. Cancer Res. 2003;63:5821–8. of tumor clone-initiating cells in colorectal cancer. J Exp Med.
48. Bapat SA, Koppikar CB, Kurrey NK. Stem and progenitor-like 2017;214:2073–88.
cells contribute to the aggressive behavior of human epithelial 67. Walker BA, Wardell CP, Melchor L, Brioli A, Johnson DC,
ovarian cancer. Cancer Res. 2005;65:3025–9. Kaiser MF, et al. Intraclonal heterogeneity is a critical early event
49. O’Brien CA, Pollett A, Gallinger S, Dick JE. A human colon in the development of myeloma and precedes the development of
cancer cell capable of initiating tumour growth in immunodefi- clinical symptoms. Leukemia. 2014;28:384–90.
cient mice. Nature. 2007;445:106–10. 68. Schober M, Fuchs E. Tumor-initiating stem cells of squamous
50. Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, cell carcinomas and their control by TGF-β and integrin/focal
Peschle C, et al. Identification and expansion of human colon- adhesion kinase (FAK) signaling. Proc Natl Acad Sci USA.
cancer-initiating cells. Nature. 2007;445:111–5. 2011;108:10544–9.
51. Schepers AG, Snippert HJ, Stange DE, Born M, van den, Es JH, 69. Jacoby MA, Duncavage EJ, Chang GS, Miller CA, Shao J,
van, Wetering Mvande, et al. Lineage tracing reveals Lgr5+ Elliott K, et al. Subclones dominate at MDS progression fol-
stem cell activity in mouse intestinal adenomas. Science. 2012; lowing allogeneic hematopoietic cell transplant. JCI Insight.
337:730–5. 2018;3:1–12.
52. Driessens G, Beck B, Caauwe A, Simons BD, Blanpain C. 70. Wong TN, Miller CA, Jotte MRM, Bagegni N, Baty JD, Schmidt
Defining the mode of tumour growth by clonal analysis. Nature. AP, et al. Cellular stressors contribute to the expansion of
2012;488:527–30. hematopoietic clones of varying leukemic potential. Nat Com-
53. Chen J, Li Y, Yu T-S, McKay RM, Burns DK, Kernie SG, et al. mun. 2018;9:1–10.
A restricted cell population propagates glioblastoma growth after 71. Engle EK, Fisher DAC, Miller CA, McLellan MD, Fulton RS,
chemotherapy. Nature. 2012;488:522–6. Moore DM, et al. Clonal evolution revealed by whole genome
54. Jamieson CHM, Ailles LE, Dylla SJ, Muijtjens M, Jones C, sequencing in a case of primary myelofibrosis transformed to
Zehnder JL, et al. Granulocyte–macrophage progenitors as can- secondary acute myeloid leukemia. Leukemia. 2015;29:869–76.
didate leukemic stem cells in blast-crisis CML. N. Engl J Med. 72. Miller CA, White BS, Dees ND, Griffith M, Welch JS, Griffith
2004;351:657–67. OL, et al. SciClone: inferring clonal architecture and tracking the
A. T. Vessoni et al.

spatial and temporal patterns of tumor evolution. PLoS Comput CCL5 and regulatory T cells. Clin Exp Immunol. 2018;191:
Biol. 2014;10. https://doi.org/10.1371/journal.pcbi.1003665. 60–73.
73. Xu J, Nuno K, Litzenburger UM, Qi Y, Corces MR, Majeti R, 91. Volonte A, Di Tomaso T, Spinelli M, Todaro M, Sanvito F,
et al. Single-cell lineage tracing by endogenous mutations enri- Albarello L, et al. Cancer-initiating cells from colorectal cancer
ched in transposase accessible mitochondrial DNA. Elife. 2019; patients escape from T cell-mediated immunosurveillance
8:1–14. in vitro through membrane-bound IL-4. J Immunol. 2014;192:
74. Klco JM, Spencer DH, Miller CA, Griffith M, Lamprecht TL, 523–32.
O’Laughlin M, et al. Functional heterogeneity of genetically 92. van Niel G, D’Angelo G, Raposo G. Shedding light on the cell
defined subclones in acute myeloid leukemia. Cancer Cell. biology of extracellular vesicles. Nat Rev Mol Cell Biol.
2014;25:379–92. 2018;19:213–28.
75. Corces MR, Chang HY, Majeti R. Preleukemic hematopoietic 93. Shimoda M, Principe S, Jackson HW, Luga V, Fang H, Moly-
stem cells in human acute myeloid leukemia. Front Oncol. neux SD, et al. Loss of the Timp gene family is sufficient for the
2017;7:1–8. acquisition of the CAF-like cell state. Nat Cell Biol.
76. Ding L, Ley TJ, Larson DE, Miller CA, Koboldt DC, Welch JS, 2014;16:889–901.
et al. Clonal evolution in relapsed acute myeloid leukaemia 94. Lotti F, Jarrar AM, Pai RK, Hitomi M, Lathia J, Mace A, et al.
revealed by whole-genome sequencing. Nature. 2012;481:506–10. Chemotherapy activates cancer-associated fibroblasts to maintain
77. Egan JB, Shi CX, Tembe W, Christoforides A, Kurdoglu A, colorectal cancer-initiating cells by IL-17A. J Exp Med.
Sinari S, et al. Whole-genome sequencing of multiple myeloma 2013;210:2851–72.
from diagnosis to plasma cell leukemia reveals genomic initiat- 95. Vermeulen L, De Sousa E, Melo F, van der Heijden M, Cameron
ing events, evolution, and clonal tides. Blood. 2012;120:1060–6. K, de Jong JH, et al. Wnt activity defines colon cancer stem cells
78. Kreso A, O’Brien C, van Galen P, Gan O, Notta F, Brown A, and is regulated by the microenvironment. Nat Cell Biol. 2010;
et al. Variable clonal repopulation dynamics influence che- 12:468–76.
motherapy response in colorectal. Cancer Sci. 2013;339:543–8. 96. Medyouf H, Mossner M, Jann JC, Nolte F, Raffel S, Herrmann
79. Dieter SM, Ball CR, Hoffmann CM, Nowrouzi A, Herbst F, C, et al. Myelodysplastic cells in patients reprogram mesenchy-
Zavidij O, et al. Distinct types of tumor-initiating cells form mal stromal cells to establish a transplantable stem cell niche
human colon cancer tumors and metastases. Cell Stem Cell. disease unit. Cell Stem Cell. 2014;14:824–37.
2011;9:357–65. 97. Calabrese C, Poppleton H, Kocak M, Hogg TL, Fuller C,
80. Venteicher AS, Tirosh I, Hebert C, Yizhak K, Neftel C, Filbin Hamner B, et al. A perivascular niche for brain tumor stem cells.
MG, et al. Decoupling genetics, lineages, and microenvironment Cancer Cell. 2007;11:69–82.
in IDH-mutant gliomas by single-cell RNA-seq. Science. 98. Charles N, Ozawa T, Squatrito M, Bleau AM, Brennan CW,
2017;355:1–11. Hambardzumyan D, et al. Perivascular nitric oxide activates
81. Weng Q, Wang J, Wang J, He D, Cheng Z, Zhang F et al. Single- notch signaling and promotes stem-like character in PDGF-
cell transcriptomics uncovers glial progenitor diversity and cell induced glioma cells. Cell Stem Cell. 2010;6:141–52.
fate determinants during development and gliomagenesis. Cell 99. Jeon HM, Kim SH, Jin X, Park JB, Kim SH, Joshi K, et al.
Stem Cell. 2019;24:707–23. Crosstalk between glioma-initiating cells and endothelial cells
82. Costa-Silva B, Aiello NM, Ocean AJ, Singh S, Zhang H, Thakur drives tumor progression. Cancer Res. 2014;74:4482–92.
BK, et al. Pancreatic cancer exosomes initiate pre-metastatic 100. Cheng L, Huang Z, Zhou W, Wu Q, Donnola S, Liu JK, et al.
niche formation in the liver. Nat Cell Biol. 2015;17:816–26. Glioblastoma stem cells generate vascular pericytes to support
83. Elinav E, Nowarski R, Thaiss CA, Hu B, Jin C, Flavell RA. vessel function and tumor growth. Cell. 2013;153:139–52.
Inflammation-induced cancer: crosstalk between tumours, 101. Kurtova AV, Xiao J, Mo Q, Pazhanisamy S, Krasnow R, Lerner
immune cells and microorganisms. Nat Rev Cancer. 2013;13: SP, et al. Blocking PGE2-induced tumour repopulation abrogates
759–71. bladder cancer chemoresistance. Nature. 2014;517:209–13.
84. Zamarron BF, Chen W. Dual roles of immune cells and their 102. Kim SY, Hong SH, Basse PH, Wu C, Bartlett DL, Kwon YT,
factors in cancer development and progression. Int J Biol Sci. et al. Cancer stem cells protect non-stem cells from anoikis:
2011;7:651–8. Bystander effects. J Cell Biochem. 2016;13:2289–301.
85. Shalapour S, Karin M, Shalapour S, Karin M. Immunity, 103. Auffinger B, Tobias AL, Han Y, Lee G, Guo D, Dey M, et al.
inflammation, and cancer: an eternal fight between good and evil. Conversion of differentiated cancer cells into cancer stem-like
J Clin Investig. 2015;125:3347–55. cells in a glioblastoma model after primary chemotherapy. Cell
86. Shi Y, Ping YF, Zhou W, He ZC, Chen C, Bian BSJ, et al. Death Differ. 2014;21:1119–31.
Tumour-associated macrophages secrete pleiotrophin to promote 104. Lagadec C, Vlashi E, Della Donna L, Dekmezian C, Pajonk F.
PTPRZ1 signalling in glioblastoma stem cells for tumour Radiation-induced reprogramming of breast cancer cells. Stem
growth. Nat Commun. 2017;8:1–17. Cells. 2012;30:833–44.
87. Lu H, Clauser KR, Tam WL, Fröse J, Ye X, Eaton EN, et al. A 105. Venkatesh HS, Morishita W, Geraghty AC, Silverbush D, Gil-
breast cancer stem cell niche supported by juxtacrine signalling lespie SM, Arzt M, et al. Electrical and synaptic integration of
from monocytes and macrophages. Nat Cell Biol. 2014;16: glioma into neural circuits. Nature. 2019;573:539–45.
1105–17. 106. Venkataramani V, Tanev DI, Strahle C, Studier-fischer A, Fan-
88. Jinushi M, Chiba S, Yoshiyama H, Masutomi K, Kinoshita I, khauser L, Kessler T, et al. Glutamatergic synaptic input to
Dosaka-Akita H, et al. Tumor-associated macrophages regulate glioma cells drives brain tumour progression. Nature. 2019;573:
tumorigenicity and anticancer drug responses of cancer stem/ 532–8.
initiating cells. Proc Natl Acad Sci USA. 2011;108:12425–30. 107. Zeng Q, Michael IP, Zhang P, Saghafinia S, Knott G, Jiao W,
89. Wei J, Barr J, Kong L-Y, Wang Y, Wu A, Sharma AK, et al. et al. Synaptic proximity enables NMDAR signalling to promote
Glioblastoma cancer-initiating cells inhibit T-cell proliferation brain metastasis. Nature. 2019;573:526–31.
and effector responses by the signal transducers and activators of 108. Hsu PP, Sabatini DM. Cancer cell metabolism: Warburg and
transcription 3 pathway. Mol Cancer Ther. 2010;9:67–78. beyond. Cell. 2008;134:703–7.
90. You Y, Li Y, Li M, Lei M, Wu M, Qu Y, et al. Ovarian cancer 109. Liberti MV, Locasale JW. The Warburg Effect: how does it
stem cells promote tumour immune privilege and invasion via benefit cancer cells? Trends Biochem Sci. 2016;41:211–8.
Tumor propagating cells: drivers of tumor plasticity, heterogeneity, and recurrence

110. Sonveaux P, Végran F, Schroeder T, Wergin MC, Verrax J, human repopulating cells masks the heterogeneity of leukemia
Rabbani ZN, et al. Targeting lactate-fueled respiration selectively initiating cells. Blood. 2008;112:568–76.
kills hypoxic tumor cells in mice. J Clin Investig. 2008;118: 128. Quintana E, Shackleton M, Foster HR, Fullen DR, Sabel MS,
3930–42. Johnson TM, et al. Phenotypic heterogeneity among tumorigenic
111. McDonald OG, Li X, Saunders T, Tryggvadottir R, Mentch SJ, melanoma cells from patients that is reversible and not hier-
Warmoes MO, et al. Epigenomic reprogramming during pan- archically organized. Cancer Cell. 2010;18:510–23.
creatic cancer progression links anabolic glucose metabolism to 129. Stewart JM, Shaw PA, Gedye C, Bernardini MQ, Neel BG, Ailles
distant metastasis. Nat Genet. 2017;49:367–76. LE. Phenotypic heterogeneity and instability of human ovarian
112. Sancho P, Burgos-Ramos E, Tavera A, Bou Kheir T, Jagust P, tumor-initiating cells. Proc Natl Acad Sci. 2011;108:
Schoenhals M, et al. MYC/PGC-1α balance determines the 6468–73.
metabolic phenotype and plasticity of pancreatic cancer stem 130. Lee J, Kotliarova S, Kotliarov Y, Li A, Su Q, Donin NM, et al.
cells. Cell Metab. 2015;22:590–605. Tumor stem cells derived from glioblastomas cultured in bFGF
113. Vlashi E, Lagadec C, Vergnes L, Reue K, Frohnen P, Chan M, and EGF more closely mirror the phenotype and genotype of
et al. Metabolic differences in breast cancer stem cells and dif- primary tumors than do serum-cultured cell lines. Cancer Cell.
ferentiated progeny. Breast Cancer Res Treat. 2014;146:525–34. 2006;9:391–403.
114. De Luca A, Fiorillo M, Peiris-Pagès M, Ozsvari B, Smith DL, 131. Ledur PF, Liu C, He H, Harris AR, Minussi DC, Zhou HY, et al.
Sanchez-Alvarez R, et al. Mitochondrial biogenesis is required Culture conditions tailored to the cell of origin are critical for
for the anchorage-independent survival and propagation of stem- maintaining native properties and tumorigenicity of glioma cells.
like cancer cells. Oncotarget. 2015;6:14777–95. Neuro Oncol. 2016;18:1413–24.
115. Viale A, Pettazzoni P, Lyssiotis CA, Ying H, Sánchez N, 132. Ledur PF, Onzi GR, Zong H, Lenz G. Culture conditions
Marchesini M, et al. Oncogene ablation-resistant pancreatic defining glioblastoma cells behavior: what is the impact for novel
cancer cells depend on mitochondrial function. Nature. 2014; discoveries? Oncotarget. 2017;8:69185–97.
514:628–32. 133. McClellan JS, Dove C, Gentles AJ, Ryan CE, Majeti R. Repro-
116. Lee K, Giltnane JM, Balko JM, Rathmell JC, Fesik SW, Arteaga gramming of primary human Philadelphia chromosome-positive B
CL, et al. MYC and MCL1 cooperatively promote chemotherapy- cell acute lymphoblastic leukemia cells into nonleukemic macro-
resistant breast cancer stem cells via regulation of mitochondrial phages. Proc Natl Acad Sci USA. 2015;112:4074–9.
oxidative phosphorylation. Cell Metab. 2017;26:633–47. 134. Quek L, David MD, Kennedy A, Metzner M, Amatangelo M, Shih
117. Lamb R, Harrison H, Hulit J, Smith DL, Lisanti MP, Sotgia F. A, et al. Clonal heterogeneity of acute myeloid leukemia treated
Mitochondria as new therapeutic targets for eradicating cancer with the IDH2 inhibitor enasidenib. Nat Med. 2018;24:
stem cells: Quantitative proteomics and functional validation via 1167–77.
MCT1/2 inhibition. Oncotarget. 2014;5:11029–37. 135. Shih AH, Jiang Y, Meydan C, Shank K, Pandey S, Barreyro L,
118. Vlashi E, Lagadec C, Vergnes L, Matsutani T, Masui K, Poulou et al. Mutational cooperativity linked to combinatorial epigenetic
M, et al. Metabolic state of glioma stem cells and non- gain of function in acute myeloid leukemia. Cancer Cell.
tumorigenic cells. Proc Natl Acad Sci USA. 2011;108:16062–7. 2015;27:502–15.
119. Denise C, Paoli P, Calvani M, Taddei ML, Giannoni E, Kopetz 136. Park SM, Gönen M, Vu L, Minuesa G, Tivnan P, Barlowe TS,
S, et al. 5-Fluorouracil resistant colon cancer cells are addicted to et al. Musashi2 sustains the mixed-lineage leukemia’driven stem
OXPHOS to survive and enhance stem-like traits. Oncotarget cell regulatory program. J Clin Investig. 2015;125:1286–98.
2015;6. https://doi.org/10.18632/oncotarget.5991. 137. Mazumdar C, Shen Y, Xavy S, Zhao F, Reinisch A, Li R, et al.
120. Lagadinou ED, Sach A, Callahan K, Rossi RM, Neering SJ, Leukemia-associated cohesin mutants dominantly enforce stem
Minhajuddin M, et al. BCL-2 inhibition targets oxidative phos- cell programs and impair human hematopoietic progenitor dif-
phorylation and selectively eradicates quiescent human leukemia ferentiation. Cell Stem Cell. 2015;17:675–88.
stem cells. Cell Stem Cell. 2013;12:329–41. 138. Sun Y, Zhou B, Mao F, Xu J, Miao H, Zou Z, et al. HOXA9
121. Ye H, Adane B, Khan N, Stranahan AW, Park CY, Jordan CT, reprograms the enhancer landscape to promote leukemogenesis.
et al. Leukemic stem cells evade chemotherapy by metabolic Cancer Cell. 2018;34:643–58.e5.
adaptation to an adipose tissue niche article leukemic stem cells 139. Trissal MC, Wong TN, Yao JC, Ramaswamy R, Kuo I, Baty J,
evade chemotherapy by metabolic adaptation to an adipose tissue et al. MIR142 loss-of-function mutations derepress ASH1L to
niche. Stem Cell. 2016;19:23–37. increase HOXA gene expression and promote leukemogenesis.
122. Liu X, Li F, Huang Q, Zhang Z, Zhou L, Deng Y, et al. Self- Cancer Res. 2018;78:3510–21.
inflicted DNA double-strand breaks sustain tumorigenicity and 140. Schapiro D, Jackson HW, Raghuraman S, Fischer JR, Zanotelli
stemness of cancer cells. Cell Res. 2017;27:764–83. VRT, Schulz D, et al. HistoCAT: Analysis of cell phenotypes
123. Liu K, Lee J, Kim JY, Wang L, Tian Y, Chan ST, et al. Mito- and interactions in multiplex image cytometry data. Nat Meth-
phagy controls the activities of tumor suppressor p53 to regulate ods. 2017;14:873–6.
hepatic cancer stem cells. Mol Cell. 2017;68:281–92.e5. 141. Giesen C, Wang HAO, Schapiro D, Zivanovic N, Jacobs A,
124. Yan C, Luo L, Guo CY, Goto S, Urata Y, Shao JH, et al. Hattendorf B, et al. Highly multiplexed imaging of tumor tissues
Doxorubicin-induced mitophagy contributes to drug resistance in with subcellular resolution by mass cytometry. Nat Methods.
cancer stem cells from HCT8 human colorectal cancer cells. 2014;11:417–22.
Cancer Lett. 2017;388:34–42. 142. Li Z, Bao S, Wu Q, Wang H, Eyler C, Sathornsumetee S, et al.
125. Pagotto A, Pilotto G, Mazzoldi EL, Nicoletto MO, Frezzini S, Hypoxia-inducible factors regulate tumorigenic capacity of
Pastò A, et al. Autophagy inhibition reduces chemoresistance glioma stem cells. Cancer Cell. 2009;15:501–13.
and tumorigenic potential of human ovarian cancer stem cells. 143. Wang Y, Liu Y, Malek SN, Zheng P, Liu Y. Targeting HIF1α
Cell Death Dis Dis. 2017;8:1–10. eliminates cancer stem cells in hematological malignancies. Cell
126. Rycaj K, Tang DG. Cell-of-origin of cancer versus cancer stem Stem Cell. 2011;8:399–411.
cells: assays and interpretations. Cancer Res. 2015;75:4003–11. 144. Jin L, Hope KJ, Zhai Q, Smadja-Joffe F, Dick JE. Targeting of
127. Taussig DC, Miraki-moud F, Anjos-afonso F, Pearce DJ, Allen CD44 eradicates human acute myeloid leukemic stem cells. Nat
K, Ridler C, et al. Anti-CD38 antibody mediated clearance of Med. 2006;12:1167–74.
A. T. Vessoni et al.

145. Krause DS, Lazarides K, von Andrian UH, Van Etten RA. 147. Bridges HR, Jones AJ, Pollak MN, Hirst J. Effects of metformin
Requirement for CD44 in homing and engraftment of BCR- and other biguanides on oxidative phosphorylation in mito-
ABL–expressing leukemic stem cells. Nat Med. 2006;12:1175–80. chondria. Biochem J. 2014;462:475–87.
146. Majeti R, Chao MP, Alizadeh AA, Pang WW, Jaiswal S, Gibbs 148. Lu R, Wang P, Parton T, Zhou Y, Chrysovergis K, Rockowitz S,
KD, et al. CD47 is an adverse prognostic factor and therapeutic et al. Epigenetic perturbations by Arg882-mutated DNMT3A
antibody target on human acute myeloid leukemia stem cells. potentiate aberrant stem cell gene-expression program and acute
Cell. 2009;138:286–99. leukemia development. Cancer Cell. 2016;30:92–107.

You might also like