You are on page 1of 11

PERSpECTIVES

for future therapeutic development efforts.


Cancer stem cell–immune cell Given the rapid development in each of
these seemingly independent areas, CSC

crosstalk in tumour progression biology and immunotherapy, exploring the


interface between these fields may provide
a new viewpoint and additional insight into
Defne Bayik and Justin D. Lathia each individual field as well as identify key
overlapping areas that can be leveraged
Abstract | Cellular heterogeneity and an immunosuppressive tumour for additional mechanistic insight and
microenvironment are independent yet synergistic drivers of tumour progression therapeutic development efforts.
and underlie therapeutic resistance. Recent studies have highlighted the complex To assess CSC–immune cell interactions,
we focus on signalling programmes that
interaction between these cell-​intrinsic and cell-​extrinsic mechanisms. The
are common for self-​renewal and immune
reciprocal communication between cancer stem cells (CSCs) and infiltrating reprogramming and that facilitate
immune cell populations in the tumour microenvironment is a paradigm for these immune evasion, tumour growth and
interactions. In this Perspective, we discuss the signalling programmes that metastasis, and therapeutic resistance.
simultaneously induce CSCs and reprogramme the immune response to facilitate In addition, we examine these programmes in
tumour immune evasion, metastasis and recurrence. We further highlight the context of individual immune cell
lineages that are resident in or infiltrate
biological factors that can impact the nature of CSC–immune cell communication. the tumour microenvironment. We also
Finally, we discuss targeting opportunities for simultaneous regulation of the CSC provide examples of cellular and molecular
niche and immunosurveillance. mechanisms that reciprocally amplify CSC
maintenance and immunosuppression.
Cellular heterogeneity has been a cells has transitioned from a theoretical Finally, we integrate these themes
long-​appreciated hallmark of advanced possibility to a viable therapeutic option for to highlight targeting opportunities to
cancers and serves as a framework to many advanced cancers4. Although many concomitantly attenuate CSC maintenance
understand aggressive and therapeutically of these strategies are based on the systemic and reduce immunosuppression in the
resistant cancers. One aspect of this targeting of protumorigenic immune cells or tumour microenvironment.
framework is the cancer stem cell the activation of an antitumoural immune
(CSC) hypothesis, which refers to response, the molecular mechanisms altering Myeloid cells
treatment-​refractory, tumour-​initiating cell the immune cell function within the tumour Tumour-​infiltrating myeloid cells are
populations1. While defining a CSC has microenvironment are not fully elucidated. a heterogeneous lineage consisting of
been challenging for a variety of reasons, Furthermore, instructive interactions monocytes, myeloid-​derived suppressor
including CSC population heterogeneity in the tumour microenvironment may cells (MDSCs), granulocytes, macrophages
and patient-​to-​patient variability, CSC underlie acquired resistance to these and dendritic cells (DCs) originating from
populations have been functionally validated therapeutic strategies, as the tumour the common myeloid progenitors in the
across multiple cancers and have revealed microenvironment is generally considered bone marrow, as well as tissue-​resident
a series of cell-​intrinsic and cell-​extrinsic to be immunosuppressive5. An additional macrophages derived from yolk sac or
molecular mechanisms that underlie confounding variable is how immune fetal liver monocytes7. Despite differences
tumour growth and therapeutic resistance cell lineages interact with CSCs, as both in their ontogeny and activation status,
(Box 1). CSC signalling has been the focus immune evasion and CSCs are recognized these diverse cell types interact with CSCs,
of therapeutic development efforts that are as integral parts of tumour growth and albeit the relative contribution of each
in the initial stages of clinical evaluation2,3. metastatic spread. Moreover, it was recently myeloid population to cancer progression is
Another aspect of CSC studies has focused demonstrated that a high-​stemness signature tumour type dependent. There is growing
on cell-​extrinsic interactions with the correlates with a poor immunogenic appreciation of the protumorigenic role
surrounding tumour microenvironment response across 21 solid malignancies, of this reciprocal communication axis.
that rely on a series of bidirectional cellular highlighting a potential interaction between These interactions, summarized in Fig. 1,
mechanisms, such as direct cell contact and these two protumorigenic pathways6. constitute potential therapeutic targets
ligand–receptor interactions, to drive tumour On the basis of these associations, the for the reprogramming of the tumour
growth through interaction with resident aim of this Perspective is to evaluate our microenvironment across multiple cancers.
and infiltrating non-​neoplastic tumour cells. current understanding of CSC–immune
These interactions are also being considered cell interactions in cancer to determine Monocytes and macrophages. CSC–
in therapeutic development2. how these interactions drive tumour immune cell interactions are best defined
Harnessing the immune system to growth and to identify novel dual CSC in the context of tumour-​associated
recognize and effectively eliminate tumour targeting and immunotherapy strategies macrophages (TAMs), which can arise

526 | August 2021 | volume 21 www.nature.com/nrc

0123456789();:
Perspectives

from tissue-​resident macrophages or can


Box 1 | Hallmarks of cancer stem cells
differentiate from immature monocytes.
Tissue-​resident macrophages populate First defined in haematological malignancies, cancer stem cells (CSCs) have since been identified
organs during embryonic development, across many solid tumours. While there are no universal markers of CSCs, surface CD133, CD34,
diversify to respond to a specific set of CD44, epithelial cell adhesion molecule (EPCAM) and CD24 expression, aldehyde dehydrogenase
(ALDH) activity and exclusion of Hoechst 33342 dye have been widely used to isolate these cells in
environmental cues by establishing distinct
different tumour types1. However, marker expression infers only the CSC state, which needs to be
expression programmes and maintain organ
rigorously defined by functional properties of self-​renewal and tumour initiation. Self-​renewal can
homeostasis by performing specialized be determined in many ways, including lineage tracing, but is generally observed using in vitro and
functions depending on their anatomical in vivo limiting-​dilution analysis via sphere formation (in vitro) and tumour initiation (in vivo) assays.
location8,9. Several studies have identified Generation of heterogeneous tumours upon implantation in immunocompromised mice and serial
tissue-​resident macrophages as an integral xenograft transplantation are frequently used to assess tumour-​initiation capacity140. CSCs are
component of adult stem cell niches (Box 2), maintained through a combination of cell-​intrinsic signalling pathways and environmental factors147.
raising the possibility that this homeostatic Collectively, these interactions maintain a CSC state that is defined by self-​renewal and tumour
mechanism may be leveraged by CSCs for initiation and may have enhanced therapeutic resistance and an epithelial–mesenchymal
their maintenance10,11. transition programme1,2,140.
While in vitro studies have classified
macrophages into dichotomous pro­ function in vitro, suggesting that CSCs may mechanisms that warrant testing of
inflammatory M1 and immunosuppressive impact the polarization state of TAMs10,19–24. combinational approaches. WISP1 and
M2 phenotypes8, TAMs can exist in a Reciprocally, TAMs can foster CSC periostin are among those targets, as their
spectrum of activation states, which is phenotypes through soluble mediators, knockdown reduced tumour-​infiltrating
further complicated by spatial and temporal such as IL-6, TGFβ, WNT ligands and myeloid cell frequency and delayed
variation12–14. To a certain degree, this pleiotrophin, or via juxtacrine signalling as tumour growth in preclinical glioblastoma
variation is determined by the evolving determined by co-​culture experiments25–30 models19,20, but they lack specific inhibitors
needs of the tumour microenvironment as (Box 3; Fig. 1a). Macrophage signalling to for clinical use. Pertinently, CSCs not only
well as by the source of the protumorigenic CSCs through receptor-​type tyrosine-​protein polarize macrophages to a protumorigenic
macrophage population. TAMs consist phosphatase-​ζ (PTPRZ1) and ephrin state but also use protective mechanisms to
of both prenatally derived macrophages, type A receptor 4 (EPHA4), among other avoid phagocytosis by macrophages. The
which are retained in some organs during receptors that have not yet been elucidated, CD47 ‘don’t eat me’ signal is upregulated
adulthood through local proliferation, increased self-​renewal (as read out by sphere in leukaemia33, HCC34, pancreatic ductal
and blood-​derived macrophages, which formation and tumour-​initiation capacity adenocarcinoma (PDAC)35 and lung cancer36
contribute to or replace the pool of tissue-​ of CSCs) via activation of nuclear factor-​κB cells expressing phenotypic markers of
resident macrophages15–17. Among the (NF-​κB), AKT and signal transducer and CSCs. Preclinical studies revealed that
multitude of factors produced by tumour activator of transcription 3 (STAT3)25,27–29 blockade of the CD47–signal regulatory
and stromal cells that govern monocyte and (Box 3; Fig. 1a). Importantly, the ability protein-​α (SIRPα) axis can trigger tumour
macrophage infiltration and survival, of CSCs to recruit TAMs through the phagocytosis across disease models33–37, and
C-​C motif chemokine 2 (CCL2) and expression of immunomodulatory factors a recent phase I clinical trial determined
macro­phage colony-​stimulating factor 1 is intertwined with stemness epigenetic that a humanized anti-​CD47 antibody is
(CSF1) are the main regulators of this programming and CSC transcriptional well tolerated in patients with advanced
behaviour. Therefore, influx of monocytes activity. For example, in human metastatic cancers38, suggesting that inhibition of
in early stage mouse mammary tumours, prostate cell lines, Polycomb repressive CD47 has translational potential. These
an essential step for subsequent metastatic complex 1 (PRC1)-​dependent chromatin observations highlight the complex interplay
spread, is facilitated by the CCL2–C-​C remodelling was necessary for downstream with functional consequences of enhanced
chemokine receptor type 2 (CCR2) CCL2 expression by CSCs31. In HCC TAM recruitment and polarization as well as
signalling axis18. CSCs can be imperative to mouse models, CCL2 and CSF1 expression increased CSC activity and survival. While
monocyte recruitment across tumour types, was driven by the activation of the Hippo the unique set of mediators contributing to
as supernatants from patient-​derived or pathway effector Yes-​associated protein this pathological axis might differ depending
mouse cholangiocarcinoma, hepatocellular (YAP), which is also essential for hepatocyte on the tumour type (Table 1), interference
carcinoma (HCC) or glioblastoma cells dedifferentiation and CSC proliferation21. with this CSC–TAM communication
grown in CSC-​enriching conditions These observations highlight the along with activation of tumour lysis
(sphere culture) had elevated levels of complexity of CSC–macrophage properties of antitumoural macrophages
protumorigenic macrophage factors, crosstalk and underscore its implications provides the conceptual framework for
including CCL2, CCL5, CSF1, growth for simultaneous targeting of added therapeutic value.
differentiation factor 15 (GDF15), immunosuppression and stemness
interleukin-13 (IL-13), transforming phenotypes. However, inhibition of the Dendritic cells. DCs play an essential role
growth factor-​β (TGFβ), periostin or CCL2–CCR2 axis alone was ineffective in priming the T cell-​mediated antitumour
WNT‐induced signalling protein 1 in reducing CSC frequency in the immune response by cross-​presenting
(WISP1; also known as CCN4) than their aforementioned HCC model21, and CSF1 tumour antigens. However, tumours can
non-​CSC counterparts10,19–24 (Box 3; Fig. 1a). receptor (CSF1R) inhibitors have had restrict the antitumorigenic properties
Subsequent treatment of macrophages variable clinical success32. These limitations of DCs by limiting their trafficking,
with conditioned media from these sphere are potentially due to the non-​specific preventing their maturation and inducing
cultures informed expression of markers elimination of antitumorigenic myeloid differentiation of tolerogenic subtypes7.
associated with immunosuppressive cells and the presence of compensatory These DCs are not devoid of function and

NAtuRe RevIeWs | CAnceR volume 21 | August 2021 | 527

0123456789();:
Perspectives

can actively support cancer growth as well as histocompatibility complex class I (MHC-​I) As CXCL1 also contributes to premetastatic
metastatic spread39. This is in part mediated antigen G (MHC-​G)41 (Fig. 1b). While the niche formation of colon cancer by
by reprogramming of regulatory CD4+ precise mechanism of action was not recruiting CXCR2+ MDSCs, mediators
T cell populations by monocyte-​derived investigated in that study, MHC-​G serves as secreted by dysfunctional or tolerogenic DCs
DCs, suggesting that tumours can inform a ligand to immunoglobulin-​like transcript might drive tumour progression through
downstream interactions of DCs with other (ILT) inhibitory receptors and can arrest multiple downstream effector functions44.
immune cell populations39,40. DC differentiation by signalling through However, it is plausible that the receptor–
Although there is very limited knowledge the IL-6–STAT3 axis42 (Box 3). ligand interaction is tumour specific, as
of the specific role of CSCs in this process, Tumorigenic DCs can also provide the CXCL12–CXCR4 axis was germane
several recent observations support a prosurvival signals for the maintenance to support provided by follicular DCs to
reciprocal relationship between CSCs and of CSCs in solid and haematological B cell lymphoma stem cells, which were
tumorigenic DCs. For example, cells from malignancies as a result of an altered characterized by exclusion of Hoechst 33342
human renal cancers expressing CD105 secretory phenotype. DCs isolated from dye, also known as the side population, and
(also known as endoglin), a surrogate patients with colon cancer or conditioned increased sphere-​forming capacity45 (Fig. 1b).
marker for CSCs, blocked maturation of with culture medium from a human colon These early findings highlight the
monocyte-​derived DCs in vitro at a higher cancer cell line are characterized by lower necessity of educating DCs against CSCs for
rate than CD105− tumour cells, on the IL-12 and higher C-​X-​C motif chemokine 1 the success of DC vaccine strategies that are
basis of the expression of co-​stimulatory (CXCL1) production43. CXCL1 increases currently in clinical development. To this
molecules41. This phenotypic change was expression of the stem cell signalling end, DCs pulsed with the lysates of mouse
attributed to extracellular vesicle-​associated network proteins NANOG, OCT4, SOX2 cancer cell lines grown in CSC-​enriching
expression of the inhibitory receptor major and MYC and drives cell migration43 (Fig. 1b). sphere conditions or selected for ALDH

a CSC b c d
CD47
P P P • NANOG P P P P P P
STAT3 Additional • OCT4 STAT3 STAT3
receptors • SOX2
NF-κB AKT • MYC NF-κB

IL-6R CXCR4 IL-6R

• CCL2
EPHA4

• CSF1 MIF CXCL5


• CCL5 PTPRZ1 G-CSF
MHC-G Notch
• IL-13
• TGFβ
• GDF15 S100A9
• WISP or PGE2
• Pleiotrophin
• Periostin • TGFβ
• IL-6 ILT
Ephrin A

TGFβ
CSF1R • WNT NO

CCR2 CXCL12 CXCL1 CD74


IL-6
Arginase G-CSFR
P P P
STAT3 Additional
receptors P P P CXCR2
AKT STAT3

Macrophage DC M-MDSC PMN-MDSC and TAN

Fig. 1 | A unique set of mediators informs the communication between maturation through major histocompatibility complex class I antigen G
cancer stem cells and innate immune cell populations. Reciprocal commu- (MHC-​G)–immunoglobulin-​like transcript (ILT) inhibitory receptor interaction.
nication between cancer stem cells (CSCs) and myeloid cells through soluble Tolerogenic DCs release C-​X-​C motif chemokine 12 (CXCL12), which serves
mediators or juxtacrine signalling promotes immunosuppression and stem- as a ligand to C-​X-​C chemokine receptor type 4 (CXCR4) and CXCL1 to pro-
ness. a | CSCs drive monocytes and macrophages via molecules including C-​C mote CSCs. These interactions lead to the activation of the stem cell tran-
motif chemokine 2 (CCL2), CCL5, colony-​stimulating factor 1 (CSF1), trans- scription factors NANOG, OCT4, SOX2 and MYC. c | Nitric oxide (NO)
forming growth factor-​β (TGFβ), growth/differentiation factor 15 (GDF15), produced by monocytic myeloid-​derived suppressor cells (M-​MDSCs) acti-
WNT-​induced signalling protein 1 (WISP1) and periostin that bind to surface vates Notch on CSCs, and IL-6 signalling increases STAT3 phosphorylation.
receptors, including CSF1 receptor (CSF1R) and C-​C chemokine receptor CSCs recruit M-​MDSCs via macrophage migration inhibitory factor (MIF)
type 2 (CCR2). Macrophages, in return, express factors including interleukin-6 through CD74. CSCs also promote arginase 1 and STAT3 signalling in
(IL-6), IL-13, pleiotrophin and TGFβ that signal through receptors such as M-​MDSCs. d | The CXCL5–CXCR2 axis and granulocyte colony-​stimulating
receptor-​type tyrosine-​protein phosphatase-​ζ (PTPRZ1) and ephrin type A factor (G-​CSF) recruit tumour-​associated neutrophils (TANs) and polymor-
receptor 4 (EPHA4) to support CSCs. These interactions activate the down- phonuclear MDSCs (PMN-​MDSCs). In return, prostaglandin E2 (PGE2) and
stream signal transducer and activator of transcription 3 (STAT3), AKT and S100A9 support stemness. TGFβ has a bidirectional role in this communica-
nuclear factor-​κB (NF-​κB) pathways. CSCs also evade macrophage phago­ tion axis. Dashed arrows indicate indirect associations. G-​CSFR, granulocyte
cytosis through CD47 expression. b | CSCs interfere with dendritic cell (DC) colony-​stimulating factor receptor; IL-6R, interleukin-6 receptor.

528 | August 2021 | volume 21 www.nature.com/nrc

0123456789();:
Perspectives

expression elicited a better immune Box 2 | Interactions between adult stem cells and the immune system
response than those pulsed with bulk
tumour cells in glioblastoma, melanoma and Several studies have indicated that macrophages and regulatory T cells (Treg cells) are essential
head and neck squamous cell carcinoma components of the stem cell niches across organ systems148. Targeting of macrophages through
(HNSCC) mouse models46,47. Nonetheless, CSF1R blockade was associated with reduced numbers of LGR5+ stem cells in the intestinal
crypt149, while depletion of CD169+ macrophages was shown to interfere with retention of
the presence of CSCs in mouse tumour
haematopoietic stem cells by acting on mesenchymal stem cells within the bone marrow niche150.
cell lines is a contested subject, and high Similarly, mammary gland stem cells failed to repopulate fat pads of mice with the Csf1-​null
immunogenicity of these cell lines48 can osteopetrotic (op) mutation (Csf1op/op), which have reduced levels of macrophages151. This is a
further complicate accurate representation functional stem cell–macrophage interaction with broader implications for organ development,
of human tumours. Thus, identification of as stem cells isolated from mammary pads of Csf1op/op mice also had decreased clonogenic activity151,
additional mechanisms of DC dysregulation and inhibition of CSF1 was previously shown to interfere with mammary gland development152.
in tumours and neoantigens selectively While this particular crosstalk was mediated through WNT ligand production by NOTCH1+,
expressed in CSCs as opposed to normal NOTCH3+ and NOTCH4+ macrophages as a result of interaction with Delta-​like 1 (DLL1) expressed
stem cells are essential to improve the on the surface of stem cells153, there is still limited knowledge of the niche factors maintaining the
success and manage the toxicity of this stem cell–macrophage dialogue during homeostasis and injury. Recruitment of macrophages by
CCL2 during hair follicle regeneration raises the possibility that this is a dynamic process impacted
treatment approach in patients.
by environmental stimuli154. Treg cells have also been identified in bone marrow, intestinal and hair
follicle stem cell niches155–158. Self-​renewal of intestinal stem cells and an immune-​privileged status
Myeloid-​derived suppressor cells and of transplanted haematopoietic stem cells are maintained through IL-10 production by Treg cells,
tumour-​associated neutrophils. MDSCs are and depletion of Treg cells decreased the frequency of stem cells155,157,158. In addition, regeneration
immature bone marrow-​derived cells that of hair follicles required accumulation of Treg cells and Jagged 1 expression, pointing to a potent
expand in patients with malignancies and role for Treg cells in regulating stem cell activity156. These observations also suggest the possibility
infiltrate tumours. In addition to serving as that CSCs manipulate a host mechanism to form a favourable environment and rely on pre-​existing
markers of malignancy, MDSCs suppress interactions such as Notch or WNT signalling3, especially given the evidence that adult stem cells
the antitumour immune response and constitute precursors of CSCs in some cancers116.
limit the efficacy of anticancer therapies49.
Monocytic MDSCs (M-​MDSCs) and
polymorphonuclear MDSCs (PMN-​MDSCs) models50,54. Mechanistic analysis indicated and TGFβ, respectively57–59 (Box 3; Fig. 1d).
are two subsets of MDSCs that are involved that nitric oxide synthase 2 gene (Nos2) Subcutaneous G-​CSF-​overexpressing
in different steps of tumorigenic growth expression and nitric oxide production human cervical tumours in nude mice were
and metastatic spread50. Tumour-​associated by M-​MDSCs promoted CSC phenotypes characterized by more MDSCs and ALDH+
neutrophils (TANs) are a potential third via activation of Notch signalling and tumour cells than were tumours initiated
cell type closely linked to PMN-​MDSCs. sustained STAT3 phosphorylation in with the parental lines59. In a genetically
Although PMN-​MDSCs are widely accepted cancer cells50,54 (Box 3; Fig. 1c). In mouse engineered mouse model of prostate
as a population separate from inflammatory breast cancer cell lines, this effect was cancer, CXCL5-​dependent mobilization
neutrophils on the basis of their functional observed as increased levels of vimentin and of CXCR2+ PMN-​MDSCs was linked to
annotation and transcriptome profile51, phosphorylated STAT3, which was reversed YAP hyperactivation57. An increased TGFβ
the distinction between TANs and PMN-​ by pharmaceutical inhibition of NOS2, when signature in CD133+ mouse melanoma
MDSCs is not well defined. The variation they were co-​cultured with M-​MDSCs50. cells (CD133 is also known as PROM1)
between MDSC subtypes and TANs is linked Similarly, culturing of human breast cancer was associated with increased infiltration
to their complex and distinct roles in the cell lines with MDSCs resulted in an increase of PMN-​MDSCs and TAMs in tumours
tumour microenvironment but can also in the ALDH+ fraction, and self-​renewal, as initiated with the CD133+ fraction compared
depend on the cancer type. read out by sphere formation, was blocked with CD133− counterparts58. These
Initial observations suggested that by the combination of an NOS2 inhibitor observations point to conserved signalling
MDSCs co-​cultured with patient-​derived and an anti-​IL-6 antibody54. Importantly, mechanisms, such as the Hippo–YAP
primary ovarian cancer tissue promote this is not a unidirectional relationship, pathway and miR-92 (refs21,57,58), that drive
sphere formation and ALDH expression as CSCs recruit MDSCs to constrain T cell immunosuppressive myeloid cells into the
by stimulating miR-101 and blocking activity and create a favourable environment tumour microenvironment.
carboxy-​terminal-​binding protein 2 for tumour growth. This can be achieved Reciprocally, PMN-​MDSCs contribute
(CTBP2) in tumour cells52. Subsequent through a CSC-​dependent increase in the to the stemness of cancer cells via multiple
studies identified M-​MDSCs as the major levels of STAT3 and arginase, markers of mechanisms. Co-​culture of human
driver of CSC phenotype in pancreatic suppressive function, in M-​MDSCs through multiple myeloma cells with PMN-​MDSCs
and breast cancers53. M-​MDSCs, which mediators that are not well defined53,55. increased self-​renewal as assessed by
were detected at high levels in human However, macrophage migration inhibitory tumour sphere formation and increased
pancreatic cancer tissue, were capable of factor (MIF) is one such factor secreted by relative expression of NANOG, OCT4
increasing the frequency of the fraction CSCs to promote M-​MDSC accumulation and SOX2, which was reversed by the
of ALDH+ stem cells in mouse tumour cell and survival in the glioblastoma PIWI-​interacting RNA 823 antagomir or
lines in culture and increasing the in vivo microenvironment55,56. a DNA methyltransferase 3β (DNMT3B)
tumour growth rate following implantation In mouse cervical, prostate and inhibitor60. Similarly, PMN-​MDSCs
of these cells subcutaneously53. Similarly, melanoma tumour models, where PMN-​ increase STAT3 phosphorylation, CD133
M-​MDSCs constituted the majority of MDSCs play a more dominant role, MDSC and CD44 expression and sphere formation
tumour-​infiltrating MDSCs in human breast accumulation is driven by granulocyte of mouse and human colorectal cancer
tumour tissue and in mouse breast cancer colony-​stimulating factor (G-​CSF), CXCL5 cell lines in vitro via transfer of exosomal

NAtuRe RevIeWs | CAnceR volume 21 | August 2021 | 529

0123456789();:
Perspectives

S100A9 (ref.61). Furthermore, prostaglandin Lymphocytes. T cells and B cells are essential potentially point to variations in NK cell
E2 (PGE2) expression by MDSCs was linked components of immunosurveillance and interactions on the basis of tumour type,
to expansion of the ALDH+ fraction in have a vital role in long-​term antigen-​ with the presence of additional factors
human cervical cancer cell lines in vitro, specific immune responses. Natural killer informing the nature of communication or
as this effect was reversed by a PGE2 (NK) cells and innate lymphoid cells selective enrichment of NK cell-​resistant
inhibitor59. (ILCs) also arise from common lymphoid CSC populations in some cancers. To further
TAN–CSC interactions have been progenitors but lack antigen-​specific add to this complexity, CSCs can inform
reported in the context of lung cancer and receptors. There is limited understanding NK cell activity. CD133+ patient-​derived
HCC. In multiple mouse models of lung of how CSCs interact with lymphoid cell glioblastoma stem cells secrete TGFβ to
cancer, tumour-​derived TGFβ is ascribed populations except that CSCs use multiple restrict NKG2D expression on peripheral
as a regulator of protumorigenic TAN mechanisms to escape immunosurveillance. blood mononuclear cells71 (Box 3). Given
polarization62. CSC-​derived CXCL5 was Tumour sphere-​forming glioblastoma64, the accumulating evidence for B cell
also linked to chemotaxis of TANs in mouse colon cancer65, lung cancer66, ABCB5+ and ILC dysregulation in cancer and its
models of HCC63 (Fig. 1d). In vitro studies melanoma67 and CD34+ leukaemia68 stem ramifications for antitumour immunity72,73,
indicated that TANs can also secrete TGFβ cells are characterized by reduced expression it is likely that there is underappreciated
along with bone morphogenetic protein 2 of MHC-​I and/or NK cell receptor D crosstalk between CSCs and these immune
(BMP2) to reprogramme cancer cell lines to (NKG2D) ligand. While lower levels of cell populations. The possibility of a direct
acquire functional and phenotypical traits NKG2D ligand in CD34+ leukaemia stem line of communication between CSCs
of stem cells, including colony formation cells and MHC-​I in CD133+ glioblastoma and ILCs is further suggested by group 3
capacity, through upregulation of NF-​κB63, stem cells provided protection against ILC-​dependent STAT3 phosphorylation
affirming the bidirectional role of TGFβ in NK cell lysis in vitro68,69, in phenotypically in intestinal stem cells during tissue
the TAN–CSC communication axis (Box 3). defined lung cancer and colon cancer regeneration74. While these mechanisms
Collectively, these observations reveal that stem cells, decreased MHC-​I expression wait to be elucidated, the complex interplay
MDSCs and TANs support CSCs and are correlated with susceptibility to in vitro between CSCs and T cell subsets is better
putative targets for cancer immunotherapy. cytotoxic killing66,70. These differences described (summarized in Fig. 2).

Box 3 | TGFβ and STAT3 as master regulators


Transforming growth factor-​β (TGFβ) is a pleiotropic cytokine that lies immune phenotypes and CSC behaviour25,29,54,61,145,146,159. Activation
at the intersection of the pathological immune response and cancer stem of STAT3 can inform co-​inhibitory receptor expression by both
cell (CSC) modulation159 (see the figure, part a). As it is produced by and macrophages98 and CSCs97, and its inhibition in CSCs further interferes
acts on many of the cells involved in CSC–immune cell crosstalk, TGFβ with their ability to promote macrophages and Treg cells and suppress T cell
can amplify the impact of the pathogenic signal to drive tumour growth proliferation22,99 (see the figure, part b). Conversely, blockade of STAT3 in
in an autocrine or paracrine manner. TGFβ is an important regulator of immune cells hampers their ability to propagate CSCs53. Thereby, STAT3
the dedifferentiation process that reinforces stem signatures and immune is a key intracellular pathway that is a therapeutic target for simultaneous
evasion by signalling through SMADs26,30,58,63,71,82,93. The immune effects of reversal of immunosuppression and reprogramming of CSCs90,128,143–145.
CSC-​derived TGFβ span the inhibition of natural killer (NK) and cytotoxic In this context, TGFβ and STAT3 emerge as master regulators that shape
T cell activation71, promotion of TANs63, polarization of immunosuppressive the tumour architecture, warranting testing of combinational approaches
macrophages22, and differentiation and recruitment of regulatory T cells targeting these pathways.
(Treg cells)58,99. The close association of TGFβ with other communication DC, dendritic cell; M-​MDSC, monocytic myeloid-​derived suppressor
axes in the tumour microenvironment, most notably IL-6–Janus kinase 2 cell; NKG2D, natural killer cell receptor D; PDL1, programmed cell
(JAK2), can also potentiate downstream signal transducer and activator death 1 ligand 1; TAN, tumour-​associated neutrophil; VTCN1, V-​set
of transcription 3 (STAT3) activation, further amplifying protumorigenic domain-​containing T cell activation inhibitor 1.
a ↓ NKG2D b

P P P
STAT3

TAN M-MDSC ↑ PDL1


Macrophage NK cell

P P P
STAT3 P P P
TGFβ STAT3
SMAD2
P P P ↑ VTCN1
Regulatory CSC
B cell

P P P
STAT3 P P P
STAT3

Treg cell DC
TH17 cell T cell

530 | August 2021 | volume 21 www.nature.com/nrc

0123456789();:
Perspectives

Regulatory T cells and T helper 17 cells. and CCL5 to actively recruit Treg cells24,31,75–77 CCL5 expression, induced migration of and
Regulatory T cells (Treg cells) have a central (Fig. 2). In a mouse glioblastoma model, IL-10 production in Treg cells in a CCR5-​
role in tumour immunobiology as the CCL2-​mediated Treg cell trafficking was dependent manner75. By contrast, Treg cell
main suppressive T cell subset, and were dependent on expression of CCR4, as migration to SOX2+ mouse breast cancer
previously recognized as constituents of tumour infiltration was limited in Ccr4-​ cell lines was induced by CCL1 in transwell
adult stem cell niches (Box 2). In general, knockout mice77. CD133+ human ovarian assays76, indicating an abundance of
CSCs secrete the chemokines CCL1, CCL2 cancer cell lines, which had higher levels of chemokines that can recruit Treg cells to the
tumour microenvironment. CCL1, CCL2
and CCL5 are also crucial for the trafficking
Table 1 | Cellular mechanisms underlying CSC–immune cell communication of myeloid cells that are capable of
Signalling axis Mechanism of action Tumour type Refs promoting the infiltration of Treg cells31,78–80,
CCL2–CCR2 Recruitment of macrophages, Glioblastoma 22,77 suggesting that CSCs drive Treg cell
MDSCs and Treg cells by CSCs accumulation through a combination
HCC 21
of direct and indirect mechanisms.
Pancreas 143
Additionally, self-​renewing CSCs from a
Prostate 31 variety of cancer cell lines exhibit elevated
CSF1–CSF1R Polarization of TAMs by CSCs Breast 141 expression of indoleamine 2,3-​dioxygenase 1
(IDO1) and TGFβ, two important inducers
Cholangiocarcinoma 10
of Treg cell recruitment and generation58,81–84
Colorectal (Box 3; Fig. 2). Through production of these
141

Glioblastoma 22,77 mediators, CSCs can also stimulate Treg cells


HCC 21 to enhance CSC marker expression and
self-​renewal as assessed by increased sphere
Pancreas 143
formation in mouse breast cancer cell lines76.
Prostate 31
Treg cells share precursors with T
IDO1 CSC-​mediated accumulation of Breast 84 helper 17 (TH17) cells, and these two cell
Treg cells types can transdifferentiate in response
Glioblastoma 81,83
to environmental cues85,86. This balance
Prostate 84
between the two CD4+ T cell subsets is
IL-1β CSC phenotype promotion by Breast 114
essential for the regulation of self-​tolerance
macrophagesa versus inflammation. Because of such
Colorectal 120

plasticity, it is not surprising that IDO1


Pancreas 124
and TGFβ can also impact the pathogenic
IL-6 Upregulation of CSC phenotype, Breast 54
TH17 cell response in the tumour
perpetuation of TAM and MDSC
function, TH17 cell polarization Colorectal 88 microenvironment. Whereas IDO1 blocks
Glioblastoma 98 the conversion of Treg cells to TH17 cells,
TGFβ can trigger such a phenotypic switch
HCC 29
in collaboration with the IL-6–STAT3
Multiple myeloma 60
pathway85. These observations further
NSCLC 25 support the composite role of TGFβ in
Ovarian 144 modulation of the antitumour immune
response (Box 3). Although the role of TH17
TGFβ Stimulation of CSC phenotype, Colorectal 82
cells in cancer remains a contested issue87,
promotion of immunosuppressive
myeloid cell activity, T cell and NK Glioblastoma 22,71,99
IL-17 secreted by TH17 cells was shown
cell inhibition, Treg cell and TH17 cell HCC 26,63 to augment the self-​renewal capacity of
polarization
Melanoma 58 CSCs in multiple tumour models, and
IL-22, another cytokine produced by
Pancreas 30
these cells, induced the phosphorylation
STAT3 CSC maintenance, TAM polarization Breast 54
of STAT3 in tumour cells88–92 (Fig. 2). All
and function, MDSC polarization things considered, the versatile nature of
and function, cytotoxic T cell Colorectal 61

suppression, Treg cell generation


Treg cells and TH17 cells should be extensively
Glioblastoma 11,22,98,99
interrogated to decipher their means of
HCC 29,128
intercommunication with CSCs and other
HNSCC 97
immune cell populations for effective
NSCLC 25,90 therapeutic targeting.
Pancreas 53,143
Cytotoxic T cells. Communication of
CCL2, C-​C motif chemokine 2; CCR2, C-​C chemokine receptor type 2; CSC, cancer stem cell; CSF1,
colony-​stimulating factor 1; CSF1R, colony-​stimulating factor 1 receptor; HCC, hepatocellular carcinoma;
CSCs with CD8+ T cells can be categorized
HNSCC, head and neck squamous cell carcinoma; IDO1, indoleamine 2,3-​dioxygenase 1; IL-1β, into two areas: evasion of CSCs from
interleukin-1β; IL-6, interleukin-6; MDSC, myeloid-​derived suppressor cell; NK, natural killer; NSCLC, T cell-​mediated death and inhibition of
non-​small-​cell lung carcinoma; STAT3, signal transducer and activator of transcription 3; TAM, tumour
associated macrophage; TGFβ, transforming growth factor-​β; TH17 cell, T helper 17 cell; Treg cell, regulatory antitumorigenic properties of T cells. From
T cell. aParticular consideration in the context of ageing. the perspective of CSCs, effective execution

NAtuRe RevIeWs | CAnceR volume 21 | August 2021 | 531

0123456789();:
Perspectives

↓ T cell in CSC-​enriching conditions can account


TGFβ or
proliferation IL-10 for some of the activity99, but this is not
T cells Macrophage
necessarily achieved as a result of direct
communication between CSCs and T cells.
It is noteworthy that most of these
↓ AKT P P P
and ERK α5β1 integrin STAT3
studies are conducted ex vivo and using
artificial culture conditions, which limit
TNC
their translational potential. Moreover,
PD1 EV a portion of the observed effect can be
PDL1 attributed to the presence of myeloid cells that
• CCL1 • CCL2 serve as intermediaries. As such, impaired
↓ MHC-I • CCL5 • IL-4
• IL-6 • IL-10 T cell activation is in part mediated by the
CSC uptake of extracellular vesicles derived
P P P from self-​renewing CSCs by myeloid cells
TGFβ
STAT3
in peripheral blood mononuclear cells
• CCL1 and subsequent IL-6, IL-10 and IL-1β
P P P P P P
STAT3 • CCL2 STAT3 production100. CSC supernatants are also
• CCL5 M-MDSC enriched in IL-4, IL-6 and IL-10, all of
IL-17 • TGFβ TGFβ
or IL-22 • IDO1 which can activate STAT3 signalling in
TH17 cell Treg cell
tumour-​associated myeloid cells, with
outcomes such as induced expression of the
Fig. 2 | CSCs interfere with T cell activity directly or through immunosuppressive myeloid cells. checkpoint molecules VTCN1 and PDL1, and
Cancer stem cells (CSCs) suppress or evade antitumorigenic T cells in part by releasing extracellular production of the immunosuppressive factors
vesicle (EV)-​associated or free tenascin C (TNC) and reducing AKT and ERK signalling. CSCs also IL-10 and TGFβ65,94,98,99,101 (Box 3; Fig. 2).
induce tumour-​promoting regulatory T cells (Treg cells) and T helper 17 (TH17) cells. CSCs downregulate Still, the CSC secretome can directly
major histocompatibility complex class I (MHC-​I) and overexpress checkpoint regulators, including interfere with T cell effector function
programmed cell death 1 ligand 1 (PDL1). CSCs further drive recruitment and polarization of TH17 cells in the absence of myeloid cells. One
and Treg cells by the combination of C-​C motif chemokine 1 (CCL1), CCL2, CCL5, transforming growth
of the best documented examples is
factor-​β (TGFβ) and indoleamine 2,3-​dioxygenase 1 (IDO1). An additional layer of regulation of T cell
tenascin C (TNC). Engagement of
activity is mediated indirectly by immunosuppressive myeloid cells, including macrophages and mono-
cytic myeloid-​derived suppressor cells (M-​MDSCs). This effect partially depends on CCL1, CCL2, CCL5, extracellular vesicle-​associated or free
interleukin-4 (IL-4), IL-6 and IL-10 secreted by CSCs. Myeloid cells produce additional suppressive TNC with the integrin receptor α5β1
molecules, such as the checkpoint inhibitor ligand PDL1, to block T cell activation in a contact-​ on T cells downregulates AKT and ERK
dependent manner or by promoting Treg cells. Collectively, these interactions reshape the tumour phosphorylation to impair proliferation
microenvironment and create a habitat where Treg cells and TH17 cells support CSCs, the latter via IL-17 in co-​culture conditions102,103 (Fig. 2).
production. STAT3, signal transducer and activator of transcription 3. Nevertheless, inhibition of TNC was not
sufficient to fully reverse CSC-​mediated
T cell suppression, warranting further
of the first area can occur via limiting the array of co-​inhibitory receptors expressed investigation of CSC-​derived soluble
ability of CSCs to present neoantigens by CSCs has not been investigated, several mediators that can directly impact cytotoxic
to T cells via downregulation of MHC-​I studies revealed that CSCs, which were T cell activity. Importantly, emerging
expression in the CSCs66,67. Furthermore, defined on the basis of expression of evidence suggests that dysfunctional T cells
single-​cell RNA sequencing analysis of phenotypic markers, have higher levels incapable of lysing CSCs can promote
mouse epidermal skin squamous cell of inhibitory checkpoint receptors. For functional and phenotypic stemness
carcinomas defined an increased stemness example, expression of programmed cell traits in a breast cancer cell line through
signature in tumour cells resistant to death 1 ligand 1 (PDL1) was elevated in an undefined mechanism104. Gaining
adoptive T cell therapy . Mechanistic
93
CD44 breast cancer stem cells , CD133
+ 94,95 +
mechanistic insight into these interactions
studies revealed that these cells expressing colon cancer stem cells94,96, and CD44+ will be essential for the design of chimeric
markers for CSCs escaped cytotoxic T cell HNSCC stem cells97 and expression of antigen receptor T cell strategies directed
killing by upregulating CD80 in a TGFβ-​ V-​set domain-​containing T cell activation against CSC antigens105.
dependent manner . While these studies
93
inhibitor 1 (VTCN1) was elevated in
indicate the presence of multiple avoidance CD133+ glioblastoma stem cells98 compared Environmental regulators
mechanisms, our knowledge of therapeutic with non-​cancer stem cells (Fig. 2). In Although there is accumulating evidence
utility is currently limited. addition, sphere-​forming patient-​derived that CSCs and immune cells interact in
In contrast, the regulation of T cell glioblastoma cells or human glioblastoma various types of malignancies, factors
activity by CSCs is a more elaborate cell lines64,99,100 and patient-​derived colon impacting the nature of this communication
and better defined process that involves cancer cells65 and ABCB5+ patient-​derived are less well described. A complex interplay
multiple pathways through both direct melanoma cells67 have been shown to within the tumour microenvironment can
and indirect contact. One mechanism suppress the proliferation of T cells and limit be influenced by host variation induced by
by which CSCs impair cytotoxic T cell the expression of co-​stimulatory molecules factors including (but not limited to) sex,
activity is through selective enrichment of in vitro. Upregulation of phosphorylated age and metabolic state. Understanding
inhibitory checkpoint ligands compared STAT3 in T cells co-​cultured with CD133+ the contribution of these biological
with non-​CSC tumour cells. While the full primary human glioblastoma cells grown determinants should provide unique

532 | August 2021 | volume 21 www.nature.com/nrc

0123456789();:
Perspectives

insights into altered pathways and additional infidelity characterized by combinational associated with a chronic low-​grade
regulators that sustain the pathological expression of key identity genes inflammatory state126 that could not only
conversation between CSCs and the immune distinguishes CSCs from restricted stem shape the antitumour immune response,
system. These additional factors, while cell lineages116, suggesting that deregulation such as MDSC accumulation49, but that also
potentially confounding, are likely to inform of multipotent cells can give rise to CSCs. has the potential to reshape CSC niches.
future therapeutic efforts in the emerging By proxy, this observation also indicates Studies established that healthy stem cells
era of personalized medicine. that CSCs arising from a pre-​existing stem can change their chromatin accessibility in
cell pool can inherit age-​induced cellular response to acute inflammatory stimuli to
Sex as a biological variable. characteristics that provide a survival acquire a distinct memory phenotype, which
Epidemiologically, male sex is associated advantage. With ageing, stem cells acquire a can increase susceptibility to stress-​induced
with an increased rate of non-​reproductive more quiescent state that is associated with lineage infidelity116,127. These observations
cancers, worse prognosis and higher downregulation of antigen presentation raise the possibility that stem cells acquire
mortality106. While the impact of biological and enhanced ability to escape from a pro-​oncogenic phenotype as a result of a
sex on cancer treatment is under intensive T cell killing117,118, indicating that similar sustained inflammatory response or as
investigation mainly through the use of functional changes in CSCs could form part of tissue adaptation mechanisms128.
adjuvant hormone blockers, mechanistic the basis of cancer immune evasion and Accordingly, HFD consumption enhanced
studies focusing on the relative contribution therapeutic resistance. stem cell function and oncogenic
of genetic factors versus hormonal cues are An opposing point of view is that CSCs transformation in experimental mouse
still in their infancy. Early reports indicate emerge from dedifferentiating tumour models of colon cancer as a consequence of
that both differences in cellular programming cells119. Notably, co-​existence of these altered niche signalling pathways directed by
and sex-​specific immune interactions can two mechanisms could be the source fatty acids129,130.
contribute to cancer progression107. As an of CSC heterogeneity observed within Alternatively, altered stem cell behaviour
example, enhanced susceptibility of mouse tumours. Ageing of the immune system could be a secondary effect to diet-​induced
astrocytes to malignant transformation is another factor that could contribute changes in microbiome composition131. In
following inactivation of the tumour to CSC heterogeneity and abundance support of this notion, HFD consumption
suppressors RB1 and p53 has been implicated since co-​culturing cancer cell lines with shifted the commitment of haematopoietic
in the male predominance of glioblastoma108. macrophages, MDSCs or TANs in vitro stem cells towards common myeloid
Importantly, these cells had an increased can trigger tumour cell dedifferentiation progenitors, leading to decreased numbers
colony-​forming ability, suggesting that as demonstrated by the upregulation of of common lymphoid progenitors in
they acquire a more stem-​like phenotype stem cell markers and the acquisition healthy mice through modulation of the
as compared with female astrocytes lacking of self-​renewal capacity26,29,30,52–54,59,63,120. microbiota132. A microbial component,
RB1 and p53. In parallel, male glioblastoma Importantly, advanced age is accompanied β-​glucan, and the associated IL-1β
tumours are infiltrated at higher rates by a heightened inflammation signature. inflammatory response were central to
by immunosuppressive myeloid cells, in Changes in the frequency of immune cell this process132,133. In addition, microbial
particular by M-​MDSCs109. Although it has populations originate from myeloid-​biased metabolites are also capable of regulating
been shown that CSCs recruit M-MDSCs lineage commitment of aged haematopoietic stem cell behaviour134 and the antitumour
to the tumour microenvironment through stem cells121, and epigenetic modification immune response135. While microbiome
MIF55,56, any potential correlation between of human immune cells also determines dysbiosis is especially well documented
increased SOX2+CD133+ CSC and age-​dependent immunological phenotypes, in gastrointestinal malignancies, it has
M-MDSC abundance in male tumours such as upregulation of IL-1β122. As a central far-​reaching consequences for antitumour
and sexual dimorphism in the expression regulator of the proinflammatory response, immunity, tumour growth dynamics
of immunomodulatory factors by CSCs IL-1β can affect numerous physiological and immunotherapy response across
remains to be investigated. These types of pathways, including CSC maintenance, malignancies136–139. However, how microbial
investigations are of particular importance, especially in inflammation-​driven instructions define the pathological
as sex hormones are capable of altering gastrointestinal malignancies. Colon communication between CSCs and immune
both immune cell function and CSC and pancreatic cancers can potentiate cells remains an outstanding question.
behaviour110–114. macrophages to secrete IL-1β, which
upregulates expression of stemness Therapeutic resistance
Ageing. Ageing is another risk factor linked markers in tumour cells and augments Many immunotherapies and CSC
to higher cancer prevalence115. Increased sphere-​forming ability via activation of pathway inhibitors are currently in clinical
accumulation of genetic mutations, reduced NF-​κB120,123,124. However, the dynamic role development or are being evaluated for
immune activity, altered cellular metabolism of IL-1β in age-​related cancer differences solid and non-​solid tumours. While these
and decline in sex hormone levels are among awaits to be elucidated, and understanding therapeutic strategies (Table 1) are likely to
the multitude of factors connecting ageing the influences of ageing on CSC–immune improve treatment outcomes, CSCs, which
with multiple pathophysiological conditions, cell communication axes could inform are more resistant to anticancer therapies
including adult cancers. However, there therapeutic strategies. generally2,140 than the tumour bulk, can
is limited knowledge of how ageing of alter their intrinsic programming to rewire
the tumour microenvironment impacts Obesity and diet. Obesity is a risk factor their mode of interaction with immune
protumorigenic pathways. for cancer occurrence125, although the cells, and this could hinder therapeutic
Ontogeny of CSCs is an important extent of altered metabolic pathways and targeting. For example, chemoresistant
consideration for the understanding of their their ramifications are not fully defined. CSCs from human breast and colon
behaviour in the context of ageing. Lineage Obesity and a high-​fat diet (HFD) are cancer cell lines were shown to upregulate

NAtuRe RevIeWs | CAnceR volume 21 | August 2021 | 533

0123456789();:
Perspectives

interferon regulatory factor 5 (IRF5) in cancer145. Similarly, a multimodal role 1. Batlle, E. & Clevers, H. Cancer stem cells revisited.
Nat. Med. 23, 1124–1134 (2017).
expression to boost CSF1 production141. for TGFβ (Box 3) makes it an attractive 2. Saygin, C., Matei, D., Majeti, R., Reizes, O. &
These drug-​resistant self-​renewing therapeutic candidate. Therefore, Lathia, J. D. Targeting cancer stemness in the clinic:
from hype to hope. Cell Stem Cell 24, 25–40 (2019).
cells can enhance the polarization of neutralizing antibodies and small molecules 3. Clara, J. A., Monge, C., Yang, Y. & Takebe, N.
phenotypically immunosuppressive are under clinical investigation, but the Targeting signalling pathways and the immune
microenvironment of cancer stem cells - a clinical
macrophages from CD14+ peripheral central role of TGFβ in tissue remodelling update. Nat. Rev. Clin. Oncol. 17, 204–232 (2020).
monocytes in a co-​transplantation setting, remains a chief safety concern146. Hence, 4. Galluzzi, L., Chan, T. A., Kroemer, G., Wolchok, J. D. &
Lopez-​Soto, A. The hallmarks of successful anticancer
which results in accelerated tumour growth the outcomes of these clinical trials and immunotherapy. Sci. Transl. Med. 10, eaat7807 (2018).
in immunocompromised mice141. As a accompanying biomarker and mechanistic 5. Hinshaw, D. C. & Shevde, L. A. The tumor
microenvironment innately modulates cancer
separate study demonstrated that CSCs studies will be essential to identify resistance progression. Cancer Res. 79, 4557–4566 (2019).
undergo chromatin remodelling to acquire mechanisms and combinational approaches, 6. Miranda, A. et al. Cancer stemness, intratumoral
heterogeneity, and immune response across cancers.
tolerance to receptor tyrosine kinase including simultaneous targeting of TGFβ Proc. Natl Acad. Sci. USA 116, 9020–9029 (2019).
inhibitors and that inhibition of histone and STAT3. 7. Engblom, C., Pfirschke, C. & Pittet, M. J. The role of
myeloid cells in cancer therapies. Nat. Rev. Cancer 16,
deacetylases interferes with CSC sphere 447–462 (2016).
formation by overcoming drug resistance, Conclusions and perspectives 8. Davies, L. C., Jenkins, S. J., Allen, J. E. & Taylor, P. R.
Tissue-​resident macrophages. Nat. Immunol. 14,
epigenetic modifications emerge as a likely In this Perspective, we have provided 986–995 (2013).
mechanism altering their communication examples of communication of CSCs with 9. Mass, E. et al. Specification of tissue-​resident
macrophages during organogenesis. Science 353,
with immune cells142. individual immune cell lineages, as well aaf4238 (2016).
TAMs can also drive drug resistance as the mechanisms of crosstalk across a 10. Raggi, C. et al. Cholangiocarcinoma stem-​like subset
shapes tumor-​initiating niche by educating associated
of CSCs in a paracrine fashion. CSCs variety of cancers. Moving forward, we macrophages. J. Hepatol. 66, 102–115 (2017).
from colon and lung cancer cell lines anticipate that this knowledge will help 11. Hide, T. et al. Oligodendrocyte progenitor cells and
macrophages/microglia produce glioma stem cell
prime macrophages to induce secretion guide the next generation of therapeutic niches at the tumor border. EBioMedicine 30,
of lactadherin and IL-6, which in return strategies for simultaneous targeting of 94–104 (2018).
12. Huang, Y. K. et al. Macrophage spatial
activates STAT3 signalling25. In the context CSCs and pathological immune response. heterogeneity in gastric cancer defined by multiplex
of colon cancer, this is accompanied However, rapid realization of this will immunohistochemistry. Nat. Commun. 10, 3928
(2019).
by acquired resistance to cisplatin require investment in the interrogation of 13. Bowman, R. L. et al. Macrophage ontogeny underlies
chemotherapy in vivo. Similar observations CSC programmes in immunocompetent differences in tumor-​specific education in brain
malignancies. Cell Rep. 17, 2445–2459 (2016).
were made in mouse PDAC and human mouse models, a topic that is generally 14. Cassetta, L. et al. Human tumor-​associated
ovarian cancer models, where gemcitabine overlooked, as the initial CSC observations macrophage and monocyte transcriptional landscapes
reveal cancer-​specific reprogramming, biomarkers,
or carboplatin chemotherapy results in and models used human tumour tissue in and therapeutic targets. Cancer Cell 35, 588–602
an increased tumour-​associated myeloid immunocompromised mice. New models e510 (2019).
cell frequency in mouse PDAC tumours leveraging bona fide cell surface markers 15. Hashimoto, D. et al. Tissue-​resident macrophages
self-​maintain locally throughout adult life with minimal
in vivo and human ovarian heterospheroid and/or reporter systems rigorously validated contribution from circulating monocytes. Immunity 38,
cultures in vitro143,144. Inhibition of CSF1R for functional CSC activity are likely to 792–804 (2013).
16. Ginhoux, F. & Guilliams, M. Tissue-​resident macrophage
to target myeloid cells was accompanied stimulate a new series of in vivo studies ontogeny and homeostasis. Immunity 44, 439–449
by reduced ALDH+ PDAC stem cell investigating the interaction of these cells (2016).
17. Laviron, M. & Boissonnas, A. Ontogeny of tumor-​
frequency in vivo and, in combination with with the immune system. In parallel, associated macrophages. Front. Immunol. 10, 1799
gemcitabine, limited tumour growth in part preclinical and clinical immunotherapy (2019).
18. Qian, B. Z. et al. CCL2 recruits inflammatory
by downregulating STAT3 activation143. studies would benefit from additional monocytes to facilitate breast-​tumour metastasis.
Similarly, knockdown of WNT5B in assessment of CSCs using a series of Nature 475, 222–225 (2011).
19. Tao, W. et al. Dual role of WISP1 in maintaining glioma
macrophages reduced ALDH activity in functional assays including tumour-​sphere stem cells and tumor-​supportive macrophages in
ovarian cancer stem cells grown as sphere formation and an in vivo limiting-​dilution glioblastoma. Nat. Commun. 11, 3015 (2020).
20. Zhou, W. et al. Periostin secreted by glioblastoma
cultures144. This effect is likely not restricted assay140. With the rapid advancement in stem cells recruits M2 tumour-​associated
to the innate immune lineage, and Treg cells single-​cell and spatial analysis techniques, macrophages and promotes malignant growth.
Nat. Cell Biol. 17, 170–182 (2015).
can also potentiate chemoresistance of these lines of investigations have tremendous 21. Guo, X. et al. Single tumor-​initiating cells evade
CSCs76. Collectively, these studies suggest potential to define complex interactions immune clearance by recruiting type II macrophages.
Genes Dev. 31, 247–259 (2017).
that successful treatment strategies will involving multiple immune cell lineages and 22. Wu, A. et al. Glioma cancer stem cells induce
likely need to concomitantly overcome CSCs. Finally, consideration of biological immunosuppressive macrophages/microglia.
Neuro Oncol. 12, 1113–1125 (2010).
both CSC-​dependent and immune-​driven sex, ageing, and metabolic or microbiome 23. Yi, L. et al. Glioma-​initiating cells: a predominant
resistance mechanisms and that shared interactions is poised to reveal additional role in microglia/macrophages tropism to glioma.
J. Neuroimmunol. 232, 75–82 (2011).
signalling pathways can comprise therapeutic opportunities, as these factors 24. Guo, X., Pan, Y. & Gutmann, D. H. Genetic and
therapeutic vulnerabilities. individually impact tumour growth and genomic alterations differentially dictate low-​grade
glioma growth through cancer stem cell-​specific
Given the cardinal role for STAT3 immune response. chemokine recruitment of T cells and microglia.
signalling in CSC maintenance and Neuro Oncol. 21, 1250–1262 (2019).
Defne Bayik 1,2
and Justin D. Lathia 1,2 ✉ 25. Jinushi, M. et al. Tumor-​associated macrophages
immunosuppressive cell behaviour (Box 3), regulate tumorigenicity and anticancer drug responses
it emerges as a promising candidate for 1
Lerner Research Institute, Cleveland Clinic, of cancer stem/initiating cells. Proc. Natl Acad.
Cleveland, OH, USA. Sci. USA 108, 12425–12430 (2011).
therapeutic targeting. Consequently, 26. Fan, Q. M. et al. Tumor-​associated macrophages
there are many ongoing clinical trials Case Comprehensive Cancer Center, Cleveland,
2
promote cancer stem cell-​like properties via
OH, USA. transforming growth factor-​beta1-induced epithelial-​
testing STAT3 small-​molecule inhibitors, mesenchymal transition in hepatocellular carcinoma.
✉e-​mail: lathiaj@ccf.org
oligonucleotides and indirect pathway Cancer Lett. 352, 160–168 (2014).
27. Lu, H. et al. A breast cancer stem cell niche supported
antagonists as a single agent or in https://doi.org/10.1038/s41568-021-00366-​w
by juxtacrine signalling from monocytes and
combination with other immunotherapies Published online 8 June 2021 macrophages. Nat. Cell Biol. 16, 1105–1117 (2014).

534 | August 2021 | volume 21 www.nature.com/nrc

0123456789();:
Perspectives

28. Shi, Y. et al. Tumour-​associated macrophages interaction with stromal cells. Semin. Immunol. 35, regulatory T cells. Clin. Exp. Immunol. 191, 60–73
secrete pleiotrophin to promote PTPRZ1 signalling 19–28 (2018). (2018).
in glioblastoma stem cells for tumour growth. 52. Cui, T. X. et al. Myeloid-​derived suppressor cells 76. Xu, Y. et al. Sox2 communicates with Tregs through
Nat. Commun. 8, 15080 (2017). enhance stemness of cancer cells by inducing CCL1 to promote the stemness property of breast
29. Wan, S. et al. Tumor-​associated macrophages produce microRNA101 and suppressing the corepressor cancer cells. Stem Cell 35, 2351–2365 (2017).
interleukin 6 and signal via STAT3 to promote CtBP2. Immunity 39, 611–621 (2013). 77. Chang, A. L. et al. CCL2 produced by the glioma
expansion of human hepatocellular carcinoma stem 53. Panni, R. Z. et al. Tumor-​induced STAT3 activation in microenvironment is essential for the recruitment of
cells. Gastroenterology 147, 1393–1404 (2014). monocytic myeloid-​derived suppressor cells enhances regulatory T cells and myeloid-​derived suppressor
30. Zhang, B. et al. Macrophage-​expressed CD51 stemness and mesenchymal properties in human cells. Cancer Res. 76, 5671–5682 (2016).
promotes cancer stem cell properties via the pancreatic cancer. Cancer Immunol. Immunother. 63, 78. Ban, Y. et al. Targeting autocrine CCL5-CCR5 axis
TGF-​beta1/smad2/3 axis in pancreatic cancer. 513–528 (2014). reprograms immunosuppressive myeloid cells and
Cancer Lett. 459, 204–215 (2019). 54. Peng, D. et al. Myeloid-​derived suppressor cells reinvigorates antitumor immunity. Cancer Res. 77,
31. Su, W. et al. The Polycomb repressor complex 1 endow stem-​like qualities to breast cancer cells 2857–2868 (2017).
drives double-​negative prostate cancer metastasis through IL6/STAT3 and NO/NOTCH cross-​talk 79. Eruslanov, E. et al. Expansion of CCR8+ inflammatory
by coordinating stemness and immune suppression. signaling. Cancer Res. 76, 3156–3165 (2016). myeloid cells in cancer patients with urothelial and
Cancer Cell 36, 139–155 e110 (2019). 55. Otvos, B. et al. Cancer stem cell-​secreted macrophage renal carcinomas. Clin. Cancer Res. 19, 1670–1680
32. Cannarile, M. A. et al. Colony-​stimulating factor 1 migration inhibitory factor stimulates myeloid derived (2013).
receptor (CSF1R) inhibitors in cancer therapy. suppressor cell function and facilitates glioblastoma 80. Curiel, T. J. et al. Specific recruitment of regulatory
J. Immunother. Cancer 5, 53 (2017). immune evasion. Stem Cell 34, 2026–2039 (2016). T cells in ovarian carcinoma fosters immune privilege
33. Theocharides, A. P. et al. Disruption of SIRPalpha 56. Alban, T. J. et al. Glioblastoma myeloid-​derived and predicts reduced survival. Nat. Med. 10,
signaling in macrophages eliminates human acute suppressor cell subsets express differential 942–949 (2004).
myeloid leukemia stem cells in xenografts. J. Exp. macrophage migration inhibitory factor receptor 81. Wainwright, D. A. et al. IDO expression in brain
Med. 209, 1883–1899 (2012). profiles that can be targeted to reduce immune tumors increases the recruitment of regulatory T cells
34. Lee, T. K. et al. Blockade of CD47-mediated cathepsin suppression. Front. Immunol. 11, 1191 (2020). and negatively impacts survival. Clin. Cancer Res. 18,
S/protease-​activated receptor 2 signaling provides a 57. Wang, G. et al. Targeting YAP-​dependent MDSC 6110–6121 (2012).
therapeutic target for hepatocellular carcinoma. infiltration impairs tumor progression. Cancer Discov. 82. Nakano, M. et al. Dedifferentiation process driven by
Hepatology 60, 179–191 (2014). 6, 80–95 (2016). TGF-​beta signaling enhances stem cell properties in
35. Cioffi, M. et al. Inhibition of CD47 effectively targets 58. Shidal, C., Singh, N. P., Nagarkatti, P. & Nagarkatti, M. human colorectal cancer. Oncogene 38, 780–793
pancreatic cancer stem cells via dual mechanisms. MicroRNA-92 expression in CD133+ melanoma stem (2019).
Clin. Cancer Res. 21, 2325–2337 (2015). cells regulates immunosuppression in the tumor 83. Ozawa, Y. et al. Indoleamine 2,3-dioxygenase 1
36. Liu, L. et al. Anti-​CD47 antibody as a targeted microenvironment via integrin-​dependent activation is highly expressed in glioma stem cells. Biochem.
therapeutic agent for human lung cancer and cancer of TGFbeta. Cancer Res. 79, 3622–3635 (2019). Biophys. Res. Commun. 524, 723–729 (2020).
stem cells. Front. Immunol. 8, 404 (2017). 59. Kuroda, H. et al. Prostaglandin E2 produced by 84. Stapelberg, M. et al. Indoleamine-2,3-dioxygenase
37. Hutter, G. et al. Microglia are effector cells of CD47- myeloid-​derived suppressive cells induces cancer elevated in tumor-​initiating cells is suppressed by
SIRPalpha antiphagocytic axis disruption against stem cells in uterine cervical cancer. Oncotarget 9, mitocans. Free Radic. Biol. Med. 67, 41–50 (2014).
glioblastoma. Proc. Natl Acad. Sci. USA 116, 36317–36330 (2018). 85. Sharma, M. D. et al. Indoleamine 2,3-dioxygenase
997–1006 (2019). 60. Ai, L. et al. Myeloid-​derived suppressor cells endow controls conversion of Foxp3+ Tregs to TH17-like
38. Sikic, B. I. et al. First-​in-human, first-​in-class phase I stem-​like qualities to multiple myeloma cells by cells in tumor-​draining lymph nodes. Blood 113,
trial of the anti-​CD47 antibody Hu5F9-G4 in patients inducing piRNA-823 expression and DNMT3B 6102–6111 (2009).
with advanced cancers. J. Clin. Oncol. 37, 946–953 activation. Mol. Cancer 18, 88 (2019). 86. Gagliani, N. et al. Th17 cells transdifferentiate into
(2019). 61. Wang, Y. et al. Granulocytic myeloid-​derived regulatory T cells during resolution of inflammation.
39. Kenkel, J. A. et al. An immunosuppressive dendritic suppressor cells promote the stemness of colorectal Nature 523, 221–225 (2015).
cell subset accumulates at secondary sites and cancer cells through exosomal S100A9. Adv. Sci. 6, 87. Martin, F., Apetoh, L. & Ghiringhelli, F. Controversies
promotes metastasis in pancreatic cancer. Cancer Res. 1901278 (2019). on the role of Th17 in cancer: a TGF-​beta-dependent
77, 4158–4170 (2017). 62. Fridlender, Z. G. et al. Polarization of tumor-​associated immunosuppressive activity? Trends Mol. Med. 18,
40. Barilla, R. M. et al. Specialized dendritic cells induce neutrophil phenotype by TGF-​beta: “N1” versus “N2” 742–749 (2012).
tumor-​promoting IL-10+IL-17+ FoxP3neg regulatory TAN. Cancer Cell 16, 183–194 (2009). 88. Yang, S. et al. Foxp3+IL-17+ T cells promote
CD4+ T cells in pancreatic carcinoma. Nat. Commun. 63. Zhou, S. L. et al. A positive feedback loop between development of cancer-​initiating cells in colorectal
10, 1424 (2019). cancer stem-​like cells and tumor-​associated cancer. J. Leukoc. Biol. 89, 85–91 (2011).
41. Grange, C. et al. Role of HLA-​G and extracellular neutrophils controls hepatocellular carcinoma 89. Zhang, Y. et al. Immune cell production of interleukin 17
vesicles in renal cancer stem cell-​induced inhibition of progression. Hepatology 70, 1214–1230 (2019). induces stem cell features of pancreatic intraepithelial
dendritic cell differentiation. BMC Cancer 15, 1009 64. Di Tomaso, T. et al. Immunobiological characterization neoplasia cells. Gastroenterology 155, 210–223 e213
(2015). of cancer stem cells isolated from glioblastoma (2018).
42. Liang, S. et al. Modulation of dendritic cell patients. Clin. Cancer Res. 16, 800–813 (2010). 90. Wang, R. et al. Th17 cell-​derived IL-17A promoted
differentiation by HLA-​G and ILT4 requires the IL-6– 65. Volonte, A. et al. Cancer-​initiating cells from tumor progression via STAT3/NF-​kappaB/Notch1
STAT3 signaling pathway. Proc. Natl Acad. Sci. USA colorectal cancer patients escape from T cell-​mediated signaling in non-​small cell lung cancer.
105, 8357–8362 (2008). immunosurveillance in vitro through membrane-​bound Oncoimmunology 7, e1461303 (2018).
43. Hsu, Y. L. et al. Interaction between tumor-​associated IL-4. J. Immunol. 192, 523–532 (2014). 91. He, W. et al. IL22RA1/STAT3 signaling promotes
dendritic cells and colon cancer cells contributes 66. Morrison, B. J., Steel, J. C. & Morris, J. C. Reduction stemness and tumorigenicity in pancreatic cancer.
to tumor progression via CXCL1. Int. J. Mol. Sci. of MHC-​I expression limits T-​lymphocyte-mediated Cancer Res. 78, 3293–3305 (2018).
https://doi.org/10.3390/ijms19082427 (2018). killing of cancer-​initiating cells. BMC Cancer 18, 469 92. Jiang, R. et al. IL-22 is related to development
44. Wang, D., Sun, H., Wei, J., Cen, B. & DuBois, R. N. (2018). of human colon cancer by activation of STAT3.
CXCL1 is critical for premetastatic niche formation 67. Schatton, T. et al. Modulation of T-​cell activation by BMC Cancer 13, 59 (2013).
and metastasis in colorectal cancer. Cancer Res. 77, malignant melanoma initiating cells. Cancer Res. 70, 93. Miao, Y. et al. Adaptive immune resistance emerges
3655–3665 (2017). 697–708 (2010). from tumor-​initiating stem cells. Cell 177, 1172–1186
45. Lee, C. G. et al. A rare fraction of drug-​resistant 68. Paczulla, A. M. et al. Absence of NKG2D ligands e1114 (2019).
follicular lymphoma cancer stem cells interacts with defines leukaemia stem cells and mediates their 94. Wu, Y. et al. Increased PD-​L1 expression in breast and
follicular dendritic cells to maintain tumourigenic immune evasion. Nature 572, 254–259 (2019). colon cancer stem cells. Clin. Exp. Pharmacol. Physiol.
potential. Br. J. Haematol. 158, 79–90 (2012). 69. Wu, A. et al. Expression of MHC I and NK ligands 44, 602–604 (2017).
46. Pellegatta, S. et al. Neurospheres enriched in cancer on human CD133+ glioma cells: possible targets 95. Hsu, J. M. et al. STT3-dependent PD-​L1 accumulation
stem-​like cells are highly effective in eliciting a of immunotherapy. J. Neurooncol. 83, 121–131 on cancer stem cells promotes immune evasion.
dendritic cell-​mediated immune response against (2007). Nat. Commun. 9, 1908 (2018).
malignant gliomas. Cancer Res. 66, 10247–10252 70. Tallerico, R. et al. Human NK cells selective targeting 96. Zhi, Y. et al. B7H1 expression and epithelial-​
(2006). of colon cancer-​initiating cells: a role for natural to-mesenchymal transition phenotypes on colorectal
47. Ning, N. et al. Cancer stem cell vaccination confers cytotoxicity receptors and MHC class I molecules. cancer stem-​like cells. PLoS ONE 10, e0135528
significant antitumor immunity. Cancer Res. 72, J. Immunol. 190, 2381–2390 (2013). (2015).
1853–1864 (2012). 71. Beier, C. P. et al. The cancer stem cell subtype 97. Lee, Y. et al. CD44+ cells in head and neck squamous
48. Lechner, M. G. et al. Immunogenicity of murine solid determines immune infiltration of glioblastoma. cell carcinoma suppress T-​cell-mediated immunity
tumor models as a defining feature of in vivo behavior Stem Cell Dev. 21, 2753–2761 (2012). by selective constitutive and inducible expression
and response to immunotherapy. J. Immunother. 36, 72. Sharonov, G. V., Serebrovskaya, E. O., Yuzhakova, D. V., of PD-​L1. Clin. Cancer Res. 22, 3571–3581 (2016).
477–489 (2013). Britanova, O. V. & Chudakov, D. M. B cells, plasma 98. Yao, Y. et al. B7-H4(B7x)-mediated cross-​talk between
49. Veglia, F., Sanseviero, E. & Gabrilovich, D. I. cells and antibody repertoires in the tumour glioma-​initiating cells and macrophages via the IL6/
Myeloid-​derived suppressor cells in the era of microenvironment. Nat. Rev. Immunol. 20, 294–307 JAK/STAT3 pathway lead to poor prognosis in glioma
increasing myeloid cell diversity. Nat. Rev. Immunol. (2020). patients. Clin. Cancer Res. 22, 2778–2790 (2016).
https://doi.org/10.1038/s41577-020-00490-y 73. Bruchard, M. & Ghiringhelli, F. Deciphering the roles 99. Wei, J. et al. Glioblastoma cancer-​initiating cells inhibit
(2021). of innate lymphoid cells in cancer. Front. Immunol. 10, T-​cell proliferation and effector responses by the signal
50. Ouzounova, M. et al. Monocytic and granulocytic 656 (2019). transducers and activators of transcription 3 pathway.
myeloid derived suppressor cells differentially regulate 74. Lindemans, C. A. et al. Interleukin-22 promotes Mol. Cancer Ther. 9, 67–78 (2010).
spatiotemporal tumour plasticity during metastatic intestinal-​stem-cell-​mediated epithelial regeneration. 100. Domenis, R. et al. Systemic T cells immunosuppression
cascade. Nat. Commun. 8, 14979 (2017). Nature 528, 560–564 (2015). of glioma stem cell-​derived exosomes is mediated
51. Zhou, J., Nefedova, Y., Lei, A. & Gabrilovich, D. 75. You, Y. et al. Ovarian cancer stem cells promote by monocytic myeloid-​derived suppressor cells.
Neutrophils and PMN-​MDSC: their biological role and tumour immune privilege and invasion via CCL5 and PLoS ONE 12, e0169932 (2017).

NAtuRe RevIeWs | CAnceR volume 21 | August 2021 | 535

0123456789();:
Perspectives

101. Gabrusiewicz, K. et al. Glioblastoma stem cell-​derived of colon cancer cells through Zeb1 activation. current knowledge and future perspectives.
exosomes induce M2 macrophages and PD-​L1 Mol. Cancer 11, 87 (2012). Ann. Oncol. 31, 1336–1349 (2020).
expression on human monocytes. Oncoimmunology 7, 124. Nomura, A. et al. NFkappaB-​mediated invasiveness in 147. Laplane, L. & Solary, E. Towards a classification of
e1412909 (2018). CD133+ pancreatic TICs is regulated by autocrine and stem cells. eLife https://doi.org/10.7554/eLife.46563
102. Mirzaei, R. et al. Brain tumor-​initiating cells export paracrine activation of IL1 signaling. Mol. Cancer Res. (2019).
tenascin-​C associated with exosomes to suppress 16, 162–172 (2018). 148. Naik, S., Larsen, S. B., Cowley, C. J. & Fuchs, E.
T cell activity. Oncoimmunology 7, e1478647 (2018). 125. Lauby-​Secretan, B. et al. Body fatness and cancer– Two to Tango: dialog between immunity and stem
103. Jachetti, E. et al. Tenascin-​C protects cancer stem-​like viewpoint of the IARC Working Group. N. Engl. J. Med. cells in health and disease. Cell 175, 908–920
cells from immune surveillance by arresting T-​cell 375, 794–798 (2016). (2018).
activation. Cancer Res. 75, 2095–2108 (2015). 126. Duan, Y. et al. Inflammatory links between high fat 149. Sehgal, A. et al. The role of CSF1R-​dependent
104. Stein, R. G. et al. Cognate nonlytic interactions diets and diseases. Front. Immunol. 9, 2649 (2018). macrophages in control of the intestinal stem-​cell
between CD8+ T cells and breast cancer cells induce 127. Naik, S. et al. Inflammatory memory sensitizes skin niche. Nat. Commun. 9, 1272 (2018).
cancer stem cell-​like properties. Cancer Res. 79, epithelial stem cells to tissue damage. Nature 550, 150. Chow, A. et al. Bone marrow CD169+ macrophages
1507–1519 (2019). 475–480 (2017). promote the retention of hematopoietic stem and
105. Wang, D. et al. CRISPR screening of CAR T cells and 128. Li, X. F. et al. Chronic inflammation-​elicited liver progenitor cells in the mesenchymal stem cell niche.
cancer stem cells reveals critical dependencies for progenitor cell conversion to liver cancer stem cell with J. Exp. Med. 208, 261–271 (2011).
cell-​based therapies. Cancer Discov. https://doi.org/ clinical significance. Hepatology 66, 1934–1951 151. Gyorki, D. E., Asselin-​Labat, M. L., van Rooijen, N.,
10.1158/2159-8290.CD-20-1243 (2020). (2017). Lindeman, G. J. & Visvader, J. E. Resident
106. Clocchiatti, A., Cora, E., Zhang, Y. & Dotto, G. P. 129. Beyaz, S. et al. High-​fat diet enhances stemness and macrophages influence stem cell activity in the
Sexual dimorphism in cancer. Nat. Rev. Cancer 16, tumorigenicity of intestinal progenitors. Nature 531, mammary gland. Breast Cancer Res. 11, R62 (2009).
330–339 (2016). 53–58 (2016). 152. Van Nguyen, A. & Pollard, J. W. Colony stimulating
107. Klein, S. L. & Flanagan, K. L. Sex differences in 130. Yoshimoto, S. et al. Obesity-​induced gut microbial factor-1 is required to recruit macrophages into the
immune responses. Nat. Rev. Immunol. 16, 626–638 metabolite promotes liver cancer through senescence mammary gland to facilitate mammary ductal
(2016). secretome. Nature 499, 97–101 (2013). outgrowth. Dev. Biol. 247, 11–25 (2002).
108. Sun, T. et al. Sexually dimorphic RB inactivation 131. David, L. A. et al. Diet rapidly and reproducibly alters 153. Chakrabarti, R. et al. Notch ligand Dll1 mediates
underlies mesenchymal glioblastoma prevalence in the human gut microbiome. Nature 505, 559–563 cross-​talk between mammary stem cells and the
males. J. Clin. Invest. 124, 4123–4133 (2014). (2014). macrophageal niche. Science https://doi.org/10.1126/
109. Bayik, D. et al. Myeloid-​derived suppressor cell 132. Luo, Y. et al. Microbiota from obese mice regulate science.aan4153 (2018).
subsets drive glioblastoma growth in a sex-​specific hematopoietic stem cell differentiation by altering the 154. Chen, C. C. et al. Organ-​level quorum sensing directs
manner. Cancer Discov. 10, 1210–1225 (2020). bone niche. Cell Metab. 22, 886–894 (2015). regeneration in hair stem cell populations. Cell 161,
110. Fillmore, C. M. et al. Estrogen expands breast cancer 133. Mitroulis, I. et al. Modulation of myelopoiesis 277–290 (2015).
stem-​like cells through paracrine FGF/Tbx3 signaling. progenitors is an integral component of trained 155. Fujisaki, J. et al. In vivo imaging of Treg cells providing
Proc. Natl Acad. Sci. USA 107, 21737–21742 immunity. Cell 172, 147–161 e112 (2018). immune privilege to the haematopoietic stem-​cell
(2010). 134. Kaiko, G. E. et al. The colonic crypt protects stem niche. Nature 474, 216–219 (2011).
111. Sun, Y. et al. Estrogen promotes stemness and cells from microbiota-​derived metabolites. Cell 165, 156. Ali, N. et al. Regulatory T cells in skin facilitate
invasiveness of ER-​positive breast cancer cells through 1708–1720 (2016). epithelial stem cell differentiation. Cell 169,
Gli1 activation. Mol. Cancer 13, 137 (2014). 135. Mager, L. F. et al. Microbiome-​derived inosine 1119–1129 e1111 (2017).
112. Svoronos, N. et al. Tumor cell-​independent estrogen modulates response to checkpoint inhibitor 157. Hirata, Y. et al. CD150high bone marrow Tregs maintain
signaling drives disease progression through immunotherapy. Science 369, 1481–1489 (2020). hematopoietic stem cell quiescence and immune
mobilization of myeloid-​derived suppressor cells. 136. Gopalakrishnan, V. et al. Gut microbiome modulates privilege via adenosine. Cell Stem Cell 22, 445–453
Cancer Discov. 7, 72–85 (2017). response to anti-​PD-1 immunotherapy in melanoma e445 (2018).
113. Generali, D. et al. Immunomodulation of FOXP3+ patients. Science 359, 97–103 (2018). 158. Biton, M. et al. T helper cell cytokines modulate
regulatory T cells by the aromatase inhibitor letrozole 137. Matson, V. et al. The commensal microbiome is intestinal stem cell renewal and differentiation. Cell
in breast cancer patients. Clin. Cancer Res. 15, associated with anti-​PD-1 efficacy in metastatic 175, 1307–1320 e1322 (2018).
1046–1051 (2009). melanoma patients. Science 359, 104–108 (2018). 159. Bellomo, C., Caja, L. & Moustakas, A. Transforming
114. Sarmiento-​Castro, A. et al. Increased expression of 138. Routy, B. et al. Gut microbiome influences efficacy of growth factor beta as regulator of cancer stemness
interleukin-1 receptor characterizes anti-​estrogen- PD-1-based immunotherapy against epithelial tumors. and metastasis. Br. J. Cancer 115, 761–769 (2016).
resistant ALDH+ breast cancer stem cells. Stem Cell Science 359, 91–97 (2018).
Rep. 15, 307–316 (2020). 139. Riquelme, E. et al. Tumor microbiome diversity and Acknowledgements
115. White, M. C. et al. Age and cancer risk: a potentially composition influence pancreatic cancer outcomes. The authors thank E. Mulkearns-​Hubert for editorial assis-
modifiable relationship. Am. J. Prev. Med. 46, S7–15 Cell 178, 795–806 e712 (2019). tance. Work in the Lathia laboratory is supported by the
(2014). 140. Lathia, J. D., Mack, S. C., Mulkearns-​Hubert, E. E., Cleveland Clinic, Case Comprehensive Cancer Center,
116. Ge, Y. et al. Stem cell lineage infidelity drives wound Valentim, C. L. & Rich, J. N. Cancer stem cells in the American Brain Tumor Association, the US National Brain
repair and cancer. Cell 169, 636–650 e614 (2017). glioblastoma. Genes Dev. 29, 1203–1217 (2015). Tumor Society and the NIH (R01 NS109742, R01 NS117104
117. Kalamakis, G. et al. Quiescence modulates stem cell 141. Yamashina, T. et al. Cancer stem-​like cells derived from and P01 CA245705 (J.L.) and F32CA243314 and
maintenance and regenerative capacity in the aging chemoresistant tumors have a unique capacity to K99CA248611 (D.B.)).
brain. Cell 176, 1407–1419 e1414 (2019). prime tumorigenic myeloid cells. Cancer Res. 74,
118. Agudo, J. et al. Quiescent tissue stem cells evade 2698–2709 (2014). Author contributions
immune surveillance. Immunity 48, 271–285 e275 142. Sharma, S. V. et al. A chromatin-​mediated reversible Both authors researched data for the article, substantially
(2018). drug-​tolerant state in cancer cell subpopulations. Cell contributed to discussion of the content, and wrote, reviewed
119. Bocci, F. et al. Toward understanding cancer stem 141, 69–80 (2010). and edited the manuscript before submission.
cell heterogeneity in the tumor microenvironment. 143. Mitchem, J. B. et al. Targeting tumor-​infiltrating
Proc. Natl Acad. Sci. USA 116, 148–157 (2019). macrophages decreases tumor-​initiating cells, relieves Competing interests
120. Kaler, P., Godasi, B. N., Augenlicht, L. & Klampfer, L. immunosuppression, and improves chemotherapeutic The authors declare no competing interests.
The NF-​kappaB/AKT-​dependent induction of Wnt responses. Cancer Res. 73, 1128–1141 (2013).
signaling in colon cancer cells by macrophages and 144. Raghavan, S., Mehta, P., Xie, Y., Lei, Y. L. & Mehta, G. Peer review information
IL-1beta. Cancer Microenviron. 2, 69–80 (2009). Ovarian cancer stem cells and macrophages Nature Reviews Cancer thanks R. Clarke, C. Petritsch and the
121. Pang, W. W. et al. Human bone marrow hematopoietic reciprocally interact through the WNT pathway to other, anonymous, reviewer(s) for their contribution to
stem cells are increased in frequency and myeloid-​ promote pro-​tumoral and malignant phenotypes in the peer review of this work.
biased with age. Proc. Natl Acad. Sci. USA 108, 3D engineered microenvironments. J. Immunother.
20012–20017 (2011). Cancer 7, 190 (2019). Publisher’s note
122. Marquez, E. J. et al. Sexual-​dimorphism in human 145. Zou, S. et al. Targeting STAT3 in cancer Springer Nature remains neutral with regard to jurisdictional
immune system aging. Nat. Commun. 11, 751 (2020). immunotherapy. Mol. Cancer 19, 145 (2020). claims in published maps and institutional affiliations.
123. Li, Y., Wang, L., Pappan, L., Galliher-​Beckley, A. & 146. Ciardiello, D., Elez, E., Tabernero, J. & Seoane, J.
Shi, J. IL-1beta promotes stemness and invasiveness Clinical development of therapies targeting TGFbeta: © Springer Nature Limited 2021

536 | August 2021 | volume 21 www.nature.com/nrc

0123456789();:

You might also like