Download as pdf or txt
Download as pdf or txt
You are on page 1of 30

ll

Leading Edge

Review
Fibroblasts in cancer: Unity in heterogeneity
Yash Chhabra1,* and Ashani T. Weeraratna1,*
1Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Department of Oncology, Sidney Kimmel Cancer

Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA


*Correspondence: ychhabr1@jhmi.edu (Y.C.), aweerar1@jhu.edu (A.T.W.)
https://doi.org/10.1016/j.cell.2023.03.016

SUMMARY

Tumor cells do not exist in isolation in vivo, and carcinogenesis depends on the surrounding tumor microen-
vironment (TME), composed of a myriad of cell types and biophysical and biochemical components. Fibro-
blasts are integral in maintaining tissue homeostasis. However, even before a tumor develops, pro-tumori-
genic fibroblasts in close proximity can provide the fertile ‘soil’ to the cancer ‘seed’ and are known as
cancer-associated fibroblasts (CAFs). In response to intrinsic and extrinsic stressors, CAFs reorganize the
TME enabling metastasis, therapeutic resistance, dormancy and reactivation by secreting cellular and acel-
lular factors. In this review, we summarize the recent discoveries on CAF-mediated cancer progression with a
particular focus on fibroblast heterogeneity and plasticity.

INTRODUCTION An Achilles’ heel of current chemo- and immunotherapeutic


approaches is that a majority of current therapies primarily target
The last three decades have generated tremendous knowledge the fast-growing tumor ‘‘seeds’’ but largely ignore the contribu-
that has led to defining the ‘‘hallmarks of cancer’’ and ushered tion of fibroblasts or the fertilizing tumor ‘‘soil.’’14,15 Our current
a shift in our approach to understanding tumor biology. Much understanding of the impact of fibroblasts in supporting tumor
of the early research focused on tumor cells and the genetic growth, metastasis, and therapeutic resistance is insufficient to
and environmental processes promoting their transformation or guide us in establishing treatment modalities and targeting stra-
driving their malignancy. With breakthroughs in biochemical, tegies.16 Therefore, it will be necessary to understand the contri-
biophysical, and engineering methodologies and technological bution of the surrounding healthy stroma in malignancy and how
explosion in ‘‘-omics’’ fields—and especially with advances in its modification by cancer cells affects the course of the disease.
immunologic approaches to cancer therapy—we have begun Advances in transcriptomics and proteomics at single-cell reso-
to understand the tumor in its entirety. It is critical to appreciate lution, where a specific CAF population can be associated with a
the cellular diversity, complex interactions, and turnover rate of particular cancer subtype,17 may aid in promoting this under-
not just the tumor itself but its surrounding microenvironment. standing and predicting responses, stratifying patients, and
An important and abundant component of both the stromal tis- personalizing therapy.
sue microenvironment and TME are the fibroblasts that are not This review aims to summarize the premise and promise made
completely characterized. Within a tissue, healthy fibroblasts from the recent developments in the field of fibroblast—and in
play an important role in maintaining homeostasis, such as dur- particular CAF—biology, highlighting commonalities of fibro-
ing wound healing and regeneration and in promoting fibrotic blasts present across different cancer types and factors promot-
conditions during excessive fibrosis and scarring, as well as can- ing their differentiation to CAF and other cell types. Finally, we
cer progression, respectively.1–3 Fibroblasts appear to have a will focus on the molecular and functional heterogeneity between
bimodal effect on cancerous cells, in that they initially prevent CAFs that makes tumors evasive to therapy and highlight the role
malignant transformation and progression,4–7 but as the disease of intrinsic processes such as aging and senescence in promot-
progresses, they become misdirected and promote tumor ing the transition to CAFs.
growth. These tumor-surrounding fibroblasts are known as can-
cer-associated fibroblasts (CAFs). CAFs form a major compo- NORMAL FIBROBLASTS
nent of the tumor stroma and are primarily associated with tumor
maintenance and promotion via diverse mechanisms,8 including Fibroblasts are active in intercellular signaling and are critical in
supporting tumor cell growth by secreting growth factors, che- many developmental processes, physiologic functions, and pa-
mokines, extracellular vesicles (EVs), and extracellular matrix thologies.18,19 They are responsible for not only providing struc-
(ECM) remodeling that regulates tumor proliferation, metastasis, tural integrity but also in regulating tissue homeostasis. Fibro-
therapy resistance, immune evasion,1,2,9 and more recently re- blasts are the most abundant cell type in all the connective
activation from tumor dormancy.10,11 Clinically, activated CAFs tissues and are responsible for generating distinct microarchi-
have been associated with worse prognosis, resistance to ther- tecture and providing biochemical cues and biophysical sup-
apies, and disease recurrence in multiple cancers.12,13 port.20 Fibroblasts show heterogeneity based on anatomical

1580 Cell 186, April 13, 2023 ª 2023 Elsevier Inc.


ll
Review

origin, and their types and roles can vary due to tissue composi- actin (aSMA), stress fibers, and fibronectin splice variants,38
tion (e.g., vascularization, innervation, abundance of fat or mus- which allows ECM synthesis and remodeling at the site of injury.
cle), developmental origin (e.g., mesoderm/neural crest in the This deposition of reactive stroma is referred to as desmoplastic
skin, epicardium/endothelium in the heart), and the requirement stroma. The induction of aSMA alters cytoskeletal organization,
for tissue-specific functions (e.g., supporting hair follicle forma- which increases the contractility between activated fibroblasts—
tion in the skin or bone resorption in the synovium).21 These dif- at this point called myofibroblasts—and the ECM to close the
ferences in the behavior of fibroblasts from different body sites wound. After wound closure, the activated fibroblasts secrete
are a culmination of intrinsic differences and highlights their abil- matrix proteins and attract epithelial cells to complete the heal-
ity to respond to stress (mechanical, oxidative) that differs be- ing process before undergoing programmed cell death and ulti-
tween regions in the body.22 mately a reduction in myofibroblast populations.39,40 As a result,
only quiescent, non-contractile fibroblasts are left at the resolved
Fibroblasts and the extracellular matrix wound site. Several parallels have been drawn between wound
Among other cell types, an important function of fibroblasts is the healing and tumorigenesis where tumors have been likened to
production of a tissue-specific basement membrane that serves ‘‘wounds that never heal.’’30,41 This is apparent because, unlike
as a protective barrier, thus imparting specificity and functionality the activated fibroblasts in a normal tissue that undergo
to the surrounding epithelium such as in skin, pancreatic islets, apoptosis, the myofibroblasts in a tumor continue to persist. In
and heart.23–25 This involves synthesizing, as well as degrading, addition, the ability of fibroblasts to significantly influence other
ECM components such as collagens, fibronectins, laminins, cell types in their vicinity has shed light on their role in inflamma-
elastins, proteoglycans, integrins, matrix metalloproteinases tory processes. Fibroblasts can modulate leukocyte activity by
(MMPs), tissue inhibitors of metalloproteinases (TIMPs), and a secreting certain signaling molecules at inflammation sites and
host of other ECM proteins in an organ-dependent manner.26,27 can either inhibit immune responses under physiological condi-
ECM by nature is a complex molecular milieu and, although tions by removing dead or redundant immune cells or create a
non-cellular itself, provides cells with the tensile scaffold vital chronic inflammatory environment during fibrosis and cancer
for appropriate structural assembly into macroscopic structures. by secreting cytokines that attract immune cells.42–44
It also serves as a reservoir of growth factors, hormones, and Studies have also shown that multiple, functionally distinct
signaling molecules that integrate positional cues and translate fibroblast subtypes can arise from a single tissue. This is prom-
them into fundamental cellular fates, underlying growth, differen- inent in the dermis, where an unwounded mouse skin was shown
tiation, survival, and motility.28,29 ECM is present within all tis- to comprise two distinct fibroblast lineages: papillary (superficial)
sues and organs and is essential for tissue morphogenesis, dermis and reticular (deeper) dermis that are regulated by
differentiation, and maintaining homeostasis.30 Fundamentally, age.19,45 Although papillary fibroblasts undergo rapid prolifera-
ECM is composed of water, protein, and polysaccharides; how- tion, reticular fibroblasts actively produce ECM and are respon-
ever, the physical, topological, and biochemical composition sible for dermal wound repair.46 Other studies have established
of the ECM is markedly heterogeneous and tissue specific, striking heterogeneity in the fibroblasts residing in both murine
whereas adhesion, communication, and differentiation remain and human dermis using single-cell RNA sequencing (scRNA-
its common cellular functions.31 The importance of the fibro- seq).47 Further investigation comparing fibroblasts in different
blast-secreted ECM is also evident in a wide range of syndromes wound healing outcomes have established cell-intrinsic differ-
arising directly from the genetic abnormalities in matrix proteins ences that contribute to distinct healing outcomes between
in monogenic disorders such as osteogenesis imperfecta,32 these sites.46,48 This is reflected in the profibrotic dermal fibro-
Marfan’s syndrome,33 and other genetic disorders of the con- blasts that promote healing via scarring, whereas those in the
nective tissue such as Ehlers-Danlos syndrome.34,35 In addition, oral mucosa heal with minimal scarring.49 Reciprocal transplan-
a large diversity of acquired human diseases, such as coronary tation of these fibroblasts also confirmed that the fibroblasts
heart disease, hypertension, chronic obstructive pulmonary dis- responsible for tissue repair in the mouse oral mucosa (defined
ease, asthma, chronic kidney disease, and neurodegenerative by Wnt1 expression) were intrinsically non-fibrotic, whereas
and autoimmune disorders are directly linked with abnormal En1-positive fibroblasts from the dorsal dermis were scar pro-
ECM function in their pathogenesis.36 ducing.49,50 This full diversity of fibroblasts within the same organ
is being investigated beyond the skin, where it is also likely to
Fibroblast role in wound healing and inflammation exist. For example, fibroblasts derived from kidney cortical and
Normal fibroblasts play crucial roles during the process of wound medullary regions exhibit differences in cell culture and in their
healing and inflammation. Upon tissue damage, the resident fi- responses to mitogenic stimulation.51,52
broblasts proliferate, differentiate, and invade the site of injury. Fibroblasts across the body also show temporal heterogene-
This occurs in response to platelet clotting, following vessel ity,19,53–57 which is the direct outcome of the interaction with their
injury, resulting in secretion of cytokines (e.g., transforming immediate microenvironment, including the ECM, crosstalk with
growth factor b [TGFb], platelet-derived growth factor [PDGF], other cell types via paracrine and autocrine signals and response
interleukin-1b [IL1b], MMPs, and TIMPs) in the microenviron- to biomechanical cues (e.g., tissue stiffness, shear force). Finally,
ment that degrade the basement membrane, induce cell prolifer- the fibroblast heterogeneity reflected in a range of phenotypic
ation and migration, and recruit inflammatory cells and fibro- states such as quiescence, proliferation, senescence, activa-
blasts.27,37 These fibroblasts are referred to as ‘‘activated’’ and tion, migration, differentiation, or aging adds another level of
are characterized by de novo expression of a-smooth muscle complexity to their function.

Cell 186, April 13, 2023 1581


ll
Review

CANCER-ASSOCIATED FIBROBLASTS mately serve as reservoirs that yield specialized and activated
fibroblasts in diseased states including cancer.75 Both universal
CAFs are found in almost all solid tumors; however, their abun- and specialized fibroblast subtypes were similar between human
dance varies between different types of cancers. Multiplex imag- and mouse and the heterogeneity was promoted by tissue
ing techniques, such as antibody-based staining and nucleic type and disease context.75 CAFs can be quiescent or activated
acid in situ hybridization, have been informative in establishing based on their cellular states. The quiescent CAFs often
spatial location of CAFs within a tumor,58,59 but their origins resemble normal fibroblasts with regards to reduced prolifera-
remain unclear. Inherent limitations associated with the longitu- tion and metabolic capacity.76 On the other hand, activated
dinal sampling of the same human biopsy samples have made CAFs, termed myofibroblasts, are characterized by attributes
it nigh impossible to determine the spatiotemporal regulation not limited to increase in proliferation, ability to migrate, meta-
of fibroblasts conversion into CAFs. Therefore, cell-line-based bolic activity, production of ECM, and growth factors and sus-
2D and 3D spheroid or organoid models in vitro have been estab- ceptible to epigenetic modifications.9 Although it is difficult to
lished for their ease of use. These models have shed light on few ascertain when the CAFs arise, it is plausible that kinetics of
facets of CAF biology, such as cell communication and crosstalk such a change will be dependent on the anatomical site of origin
with other cell types and matrices, but they fail to recapitulate the since resident fibroblasts exhibit organ-specific transcriptomics
heterogeneous and complex interactions of CAFs.60–62 Other profile77 as well as their positional memory within the tissue,
state-of-the-art methods like liquid-air interface models,63,64 which imparts a unique gene expression.78,79 This is in addition
microfluidics technology,65,66 and bio-printed tissues65,66 have to the other surrounding stromal cells that might be in close prox-
been designed and are being implemented to emulate certain imity and can transdifferentiate into CAFs (Figure 2).
cancer types. Ultimately, an extensive amount of research in ro- The development of immunohistochemical methodologies
dent models, utilizing irreversible labeling of cell types in a tissue- and scRNA-seq have redefined classical CAF markers, such
restricted manner, has permitted cell tracing to model disease as PDGF receptor-a/b (PDGFRa/b), aSMA, fibroblast-associ-
progression. Although this is a step in the right direction, manip- ated protein (FAP), and fibroblast-specific protein-1 (FSP1), as
ulating specific fibroblast types in vivo has been technically chal- subtype-specific.56,58,79–85 For example, elevated expression
lenging due to a lack of specific drivers or owing to overlapping of aSMA was reported to be a distinctive marker in activated
gene expression or off-target effects of inducing agents.67 More CAFs compared to normal fibroblasts, which were later shown
recently, intravital microscopy in mouse models has aided in lon- to be present in low abundance in other cancer types. Similarly,
gitudinal assessment of CAFs during tumor progression,68,69 as the broadly used marker for both normal and activated fibro-
well as the interaction between the CAFs and stroma.70,71 The blasts is FSP1, also called S100A4. However, this protein is
implementation of scRNA-seq techniques has revealed a previ- also expressed in several types of immune cells and certain can-
ously unappreciated level of CAF heterogeneity demonstrating cer cells.80,84–86 Another technical challenge posed during CAF
the presence of transcriptionally distinct CAF populations but isolation is the lack of a specific marker since most markers
also highlighting fibroblast signatures common across different are shared with other cell types. For example, PDPN is highly ex-
solid cancers.1 However, a key limitation of this technique is pressed in lymphatic endothelial cells,87 FAP is also expressed
that there is a potential to induce ectopic gene expression during by macrophages,88 and aSMA is mainly expressed by smooth
cell isolation from tissues, leading to mischaracterization of muscle cells89 and to a lesser extent in pericytes.90 This would
certain cell subpopulations.72 Spatial transcriptomics can avoid suggest that a set of subtype-specific markers vs. normal fibro-
this technical artifact by assaying cells in their native tissue blast markers are needed to isolate specific CAF population. In
context. Applications of this technique in glioblastoma and addition to the structural proteins, certain functional markers
another preliminary study in colorectal cancer have already have been described for CAFs. Numerous transcriptional fac-
started to emerge.73,74 Integrating single-cell and spatial tran- tors, including heat shock transcription factor (HSF1), STAT3,
scriptomics in the future holds promise in determining discrete, MYC, and yes-associated protein (YAP) have also been reported
dynamic, and transient cell subpopulations within various tumor to be involved in defining CAF signature and reprogram-
stages. This will allow a more accurate assessment of CAF ming.91–94 It is therefore critical that any interpretation of CAF
origin, their function as well spatial location within a tumor, which function within a tumor takes into consideration their associated
can indicate new therapeutic strategies in the future. Although heterogeneity to avoid any reproducibility issues, possibly
the molecular definition of CAFs is still a debated issue, field arising from the different stages of activation, sources of origin,
experts have suggested a framework that has increased our un- tissue location, and molecular characteristics.
derstanding of CAF biology;3 nevertheless, the CAFs like normal
fibroblasts are heterogeneous and governed by the same under- Phenotypic heterogeneity
lying conditions (Figure 1). Below, we highlight key characteris- Similar to the normal fibroblasts, CAF identification and isolation
tics and dynamics of CAFs, with emphasis on their heterogeneity requires a combination of morphological features, biomarkers,
and plasticity across different cancer types. and genetic mutations. As a result, cell populations within a tumor
that lack cancer cell mutations; exhibit an elongated spindle
Heterogeneity in cancer-associated fibroblasts morphology; are negative for the expression of endothelial,
Single-cell studies in both human and mouse species undertak- immune, and epithelial cell markers; and are positive for mesen-
ing pan-tissue analysis of fibroblasts has suggested the occur- chymal markers (VIM, aSMA, FAP etc.) are defined as CAFs.3,95
rence of universal fibroblast states in normal tissues, which ulti- However, none of these biomarkers are specific for CAFs, as

1582 Cell 186, April 13, 2023


ll
Review
Figure 1. Heterogeneity in fibroblast func-
tion during homeostasis and cancer devel-
opment
The heterogeneity principles are common to both
normal fibroblasts and cancer-associated fibro-
blasts (CAFs) and occur at multiple levels; cellular
level depending on the state/function of the fibro-
blast, microenvironment level depending on the
communication with other cellular and acellular
components, regional level depending on the
residing tissue and its function and lastly at the
organismal level encompassing the attributes of
demographics, physiological status, and inter-
sectionality. During normal homeostasis and heal-
ing after an acute injury, the quiescent fibroblasts
become activated (activated fibroblasts) to
mediate repair and eventually perish. In certain
conditions such as desmoplasia, the activated
fibroblasts can establish self-activating and
feedback loop, which gives rise to their pro-
tumorigenic capacities. Intrinsic or extrinsic
stressors enables their transition into CAFs
providing greater proliferative capacity. The prin-
ciples above dictate if the quiescent fibroblast
present in the normal tissues transition into acti-
vated fibroblasts, a phenomenon common to both
wound repair in healthy tissues and transition to
CAFs in a tumor microenvironment. Due to the
inherent plasticity, the proliferative CAFs can qui-
esce and become tumor-restraining, but it remains
unknown if they can completely revert back to their
normal non-tumorigenic state. Within the tumor
microenvironment (TME), CAFs communicate with
each other (via autocrine signaling) and other stro-
mal and tumor cells (in a paracrine manner) through
growth factors, metabolites, extracellular vesicles
(EVs) and chemokines while constantly shaping
and reorganizing the extracellular matrix (ECM) to
promote a tumor permissive environment.

blast markers itself.3 This heterogeneity is


universally observed across tumor types.

Lineage-dependent heterogeneity
Resident fibroblasts within a tumor
mass, by definition, are the most com-
they do not exclude for mesenchymal cells (adipocytes or peri- mon source of CAFs.79,103,104 Owing to their high degree of
cytes). Although few reports have identified p53 mutations in plasticity, pro-tumorigenic fibroblasts and myofibroblasts get
CAFs,96–98 they were later discredited owing to experimental arti- molded by their surrounding microenvironment, which dic-
facts.99 Subsequent studies have confirmed that CAFs indeed lack tates their function. In response to intrinsic cues and extrinsic
any detectable genetic alterations.99–102 Further, relying on cellular stressors these resident fibroblasts can get activated and
morphology is also not reliable since the tumor cells undergoing revert from their quiescent state to a proliferative state,
dynamic states of epithelial to mesenchymal transition (EMT) or referred to as ‘stromagenesis’, as observed in some tu-
phenotype switching (proliferative to invasive state) can also mors.105,106 In that regard, a vast array of cytokines secreted
show elongated morphology similar to fibroblasts. More impor- by the growing tumor and infiltrating immune cells such as
tantly, isolation of CAFs from peri-lesional or tumor-surrounding TGFb, several receptor tyrosine kinase (RTK) ligands; PDGF,
regions remains challenging since numerous intracellular, extra- fibroblast growth factor (FGF), epidermal growth factor (EGF)
cellular and cell surface proteins are also common to normal fibro- and other inflammatory chemokines, such as interleukin-6
blasts. This is also confounded by the fact that ex vivo cultures of (IL6), IL1b enable this transition to CAFs via nuclear factor
isolated tumor and stromal cells including CAFs can undergo kappa B (NF-kB) and janus kinase (JAK)/signal transducer
changes based on the substrate, media and environmental condi- and activator of transcription (STAT) pathways, respec-
tions and therefore may not remain an accurate representation of tively.91,107–109 Contribution from environmental stressors in
the cell type in vivo. Expectedly, no universal marker of CAFs exists the form of ECM stiffness, reactive oxygen species and DNA
that suggests significant heterogeneity in the expression of fibro- damage from therapeutic interventions110,111 and other

Cell 186, April 13, 2023 1583


ll
Review
Figure 2. Heterogeneity in cancer-associ-
ated fibroblasts occurs at multiple levels
Cancer-associated fibroblasts (CAFs) comprise a
complex and heterogeneous group of cells as a
consequence of its diverse sources of origin.
These cells respond to intrinsic and extrinsic cues
in the tumor microenvironment (TME) that can be
grouped into defined mechanisms permitting CAF
activation. Multiple cell types within the TME can
answer the cancer hijack call in initiating transition
to CAFs through direct activation of resident
fibroblast and stellate cells. Transdifferentiation of
adipocytes, pericytes and smooth muscle cells;
endothelial to mesenchymal transition (EndMT) of
endothelial cells; epithelial to mesenchymal tran-
sition (EMT) of epithelial cells and recruitment and
activation of mesenchymal stem cells are other
mechanisms that can give rise to CAFs. CAFs
show heterogeneity of molecular expression
based on the differentiating markers of its pro-
genitor state. Markers indicated in purple are
shared by both CAF and the cell type of origin,
whereas those indicated in green are unique to the
cell type of origin. In addition, CAFs also show
heterogeneity in their function and features based
on their anatomical localization within the body.

expressing myofibroblasts.125,126 These


myofibroblasts were initially detected
during wound healing and fibrosis and ul-
timately responsible for the concept that
‘tumors are wounds that do not heal’.41
Since then, myofibroblasts that do not
express aSMA (termed proto-myofibro-
blasts) were also reported with function
in specialized normal connective tissue
and early in wound healing.127,128 Overall,
the existence of both myofibroblast CAFs
and non-myofibroblast CAFs appears to
sources (UV irradiation) can also give rise to pro-tumorigenic be the most consistent observation across different cancer
fibroblasts.112–114 The inherent plasticity of epithelial and types. However, it is the coexistence of different CAF subtypes
mesenchymal cell types during pathological processes has within a tumor evident from opposing capabilities such as tumor
become a topic of intense investigation. Numerous cell types restraining vs. tumor promoting that has made it essential to
found in the TME or in proximal tissue have been proposed characterize the CAFs based on their functional heterogeneity.
as candidates for the CAF compartment via processes of This has been made possible in recent years by the utilization
transdifferentiation, endothelial to mesenchymal transition of scRNA-seq, which has enabled the redefinition of classical
(EndMT) and epithelial to mesenchymal transition (EMT) markers as CAF subtype-specific.
(Figure 2). Some examples of CAF precursors include but CAFs, like normal fibroblasts are often characterized by
are not limited to bone marrow or adipose-derived mesen- the presence of markers such as vimentin (VIM), but many
chymal stem cells (Ad-MSCs), activated pancreatic and he- other markers have been identified to more specifically identify
patic stellate cells, fibrocytes, pericytes, adipocytes, epithelial activated CAFs. These include aSMA, FAP, FSP1, periostin
cells as well as peripheral nerves and endothelial cells.115–124 (POSTN), PDGFRa/b, podoplanin (PDPN), and integrin-b1
Spatial location and origin of CAFs therefore serve as an (CD29). However, it should be noted that these markers identify
important driver of phenotype leading to considerable intra- different cell populations with distinct expression profiles, and
and inter-heterogeneity across the diverse CAF subpopula- some overlapping, indicating a high level of heterogeneity
tions, both at the functional and gene expression level. among CAFs in the TME.1,3,56,79,108 In addition, the anatomical
Consequently, adding to the difficulty in distinguishing CAFs site of origin—i.e., cancer type—can have a profound impact
from other cell types expressing similar markers. on the CAF functions and ultimately the CAF subtypes since resi-
dent fibroblasts, the main precursors of CAFs, exhibit organ-
Functional and molecular heterogeneity specific transcriptomics profile.77 Since numerous local TMEs
The most well recognized of the CAFs reported predominantly can exist in the same tumor, activated fibroblasts can interact
in desmoplastic stroma across different tumors was aSMA with tumor cells and other stromal cells at several interfaces,

1584 Cell 186, April 13, 2023


ll
Review

which can regulate their plasticity in a spatial and temporal ing genes involved the recruitment and regulation of immune
manner. cells; CAF-S2, which is similar to myCAFs and shows upregu-
Two distinct subpopulations of CAFs with unique transcrip- lated genes encoding ECM components that creates a desmo-
tomic signature, referred to as myofibroblastic CAFs (myCAFs) plastic matrix and a contractile stromal subset and CAF-S3,
and inflammatory CAFs (iCAFs), have been identified in pancre- which may represent both pericytes associated with the vascu-
atic cancer. The myCAFs are primarily located close to the tumor lature (vCAFs) or fibroblasts populations but their precise cellular
mass and have a matrix-producing myofibroblastic phenotype identity remains elusive.145 Research on colorectal cancer has
with high expression of aSMA and low expression of IL6, shown that CAFs can be divided into two main subtypes known
whereas iCAFs are located more distally to the tumor edges as CAF-As and CAF-Bs based on TGFb induced gene expres-
and distributed throughout and defined by expression of immu- sion. These subtypes can be identified by specific markers,
nomodulatory molecules such as IL6 and CXCL12.79 These two with CAF-As expressing high levels of MMP2, decorin,
subsets were confirmed in a subsequent mouse pancreatic can- collagen-1a2, and FAP, whereas CAF-Bs express markers
cer study100 and shown to dynamically change from one subset commonly found in myofibroblasts such as aSMA, transgelin,
to the another through interplay of TGFb and IL1 induced and PDGFa.83 These observations suggest that CAFs are a
signaling, supporting the existence of different CAFs subtypes highly diverse group of cells, with varying gene signatures and
within a tumor at the same time.91 The iCAFs were reported protein expressions depending on the type of cancer and even
to share similar transcriptional profiles and signaling pathway among patients with the same type of cancer. This variability in
activation with senescent fibroblasts and secretory pheno- CAF signatures may be useful in developing prognostic factors
type.129–131 Single-cell transcriptomic analysis has also shown and new cancer diagnosis and treatment strategies.
that myCAFs and iCAFs are driven by different signaling path- In breast cancer, scRNA-seq identified different CAF subtypes
ways and reported in other types of cancer originating from that were designated based on their gene expression as develop-
breast, lung, colon or rectum and ovarian tissues.132–136 Subse- ment-associated CAFs (dCAFs) with a signature associated with
quently, another study further identified a novel population of differentiation, development and morphogenesis of tissues; ma-
CAFs in late-stage pancreatic cancer with MHC-class II trix-associated CAFs (mCAFs) with a signature associated with
(MHCII)-related genes and CD74 expression termed antigen- matrisome (ECM) gene set; vascular-associated CAFs (vCAFs)
presenting CAFs (apCAFs). The apCAFs lack the classical co- with a signature associated with angiogenesis and vascular devel-
stimulatory molecules (e.g., CD40, CD80, and CD86) that are opment and cycling CAFs (cCAFs) with a signature associated with
required to induce activation and clonal expansion of CD4+ proliferation.81 Histological characterization of mammary tumors
T cells after T cell receptor (TCR) ligation, confirming a putative revealed distinct cellular sources of CAFs, as vCAFs, mCAFs,
immune-modulatory function of these CAFs.137 A lack of co- and dCAFs originated from a perivascular location, resident fibro-
stimulatory molecules on antigen-presenting cells has been re- blasts, and malignant cells having undergone an EMT, respec-
ported to cause T cell anergy and induction of regulatory tively, whereas cCAFs were essentially designated as the prolifer-
T cells (Tregs).138,139 Consistent with these reports, apCAFs acti- ating versions of vCAFs.81 Other studies in breast cancer have
vated CD4+ T cells and promoted their differentiation into Tregs, shown the presence of other distinct CAF subpopulations accu-
directly contributing to tumor immunosuppression.1,137 As mulating differentially with regards to breast cancer subtypes
opposed to pancreatic stellate cells being the cell of origin for and tumor localization. By analyzing multiple markers, including
myCAFs and iCAFs, apCAFs were shown to be a derivate of FAP, aSMA, and CD29, four different CAF populations (referred
mesothelial cells.133,140 Similarly, existence of apCAFs have to as CAF-S1 to CAF-S4) were identified.58 These populations
also been reported in breast cancer in addition to the iCAFs included normal fibroblasts (CAF-S2) devoid of any markers and
and myCAFs subsets.141 Further analysis of pancreatic cancer similar to those found in healthy tissues; myofibroblasts (CAF-S1
on the basis of stroma density resulted in the discovery of a novel and CAF-S4) based on aSMA expression that are restricted within
CAF subtype within the low desmoplastic tumor characterized the tumor; CAF-S3 was significantly associated with healthy tissue
by a highly activated glycolytic metabolic state (termed meCAFs) outside the tumor and CAF-S2 was equally distributed in the two
and associated with increased metastasis but better response to compartments.58 Genes upregulated in CAF-S1 subset were
immunotherapy.142 involved in cell adhesion, ECM organization, and immune suppres-
Single-cell transcriptomics on lung cancer revealed the pres- sion, whereas CAF-S4 fibroblasts were characterized by muscle
ence of 5 distinct clusters where cluster 1 was enriched within tu- contraction, regulation of actin cytoskeleton and oxidative meta-
mors and associated with EMT and TGFb signature and had des- bolism58 suggesting that CAF-S1 supports both myCAFs- and
moplastic function similar to myCAFs, whereas cluster 2 was iCAFs-like functions, whereas CAF-S4 is restricted to myCAFs-
localized as co-clusters with pericytes and expressed higher like functions. Similar CAF subsets were also reported in ovarian
levels of aSMA with a more angiogenic function143 similar to cancer study where both CAF-S1 and CAF-S4 were myofibroblas-
vCAFs. However, contrary to the immunosuppression function tic owing to aSMA expression but only CAF-S1 correlated with
reported earlier, apCAFs in lung cancer are unique in their ability iCAFs with regards to immunosuppressive function.146
to activate effector T cells and are tumor-suppressive in their The increasing complexity in CAF subtypes has been further
function.144 brought to light within the same CAF subtype in breast cancer.135
Other scRNA-seq studies in melanoma have uncovered the The fibroblast-activated CAFs (CAF-S1) in breast cancer can be
existence of three main CAF subsets; CAF-S1, which is similar divided into 8 different clusters, with three of them belonging to
to iCAFs and likely engages in immune crosstalk and upregulat- the iCAFs subgroup and five of them belonging to the myCAF

Cell 186, April 13, 2023 1585


ll
Review

subgroup. Using specific pathways and gene signatures for each mammary glands,176 and following exposure to low dose radia-
cluster, the three subgroups of iCAFs have been named IL-iCAFs tion.177 As discussed later, intrinsically aged fibroblasts can also
(relating to interleukin signaling), IFNg-iCAF (relating to the Inter- behave like CAFs and can contribute to tumorigenesis. In addi-
feron g-related pathway), and detox-iCAFs (relating to detoxifi- tion, a landmark study showed that CAFs alone were sufficient
cation pathways). The five subgroups of myCAFs have been in driving tumor progression of initiated non-tumorigenic pros-
named ecm-myCAFs (relating to ECM proteins), TGFb-myCAFs tate epithelial cells both in vitro and in vivo.178 This study sup-
(relating to the TGFb-dependent pathway), wound-myCAFs ports that CAFs and therefore TME could essentially provide
(relating to wound healing signaling), IFNa/b-myCAFs (relating the equivalent of a subsequent mutational hit that is prerequisite
to the IFNa/b-related pathway), and acto-myCAFs (relating to for tumor promotion.179 Further evidence also comes from
acto-myosin signaling).135 The CAF subtypes across distinct occurrence of an altered ECM signature with distinct structural,
cancer types are listed in Table 1 and four main CAF subtypes mechanical and compositional characteristics prior to any gross
are described in Figure 3. signs of inflammation.180 Presence of pre-symptomatic patho-
Although CAFs remain heterogeneous, few studies aimed at logical state can make the surrounding tissue more vulnerable
determining therapy resistance CAF signatures have identified to cancer and other disease states (Figure 4).
molecular characteristics of CAF subtypes that are shared across Considering that tumor initiation, which is the outcome of mul-
several cancer types. This has also shed light on how the tissue/ tiple genetic and epigenetic mutations, occurs directly in the
cell of tumor origin affects the functions of CAFs within distinct epithelial cells, it is likely that fibroblasts get activated through
TMEs. In that regard, scRNA-seq across ten solid cancers has stromagenesis as described earlier. However, it is important to
revealed six distinct clusters associated with CAFs signatures; my- note that the fibroblast activation within a TME does not imply
ofibroblasts CAFs (CAFmyo), inflammatory CAFs (CAFinfla), and adi- that these activated CAFs become cancer promoters from the
pogenic CAFs (CAFadi) were the most predominant subtypes along onset. The varying degree of CAF activation is evident from the
with three minor components identified based on their origin as occurrence of quiescent CAFs, tumor-restraining CAFs, and tu-
endothelial-to-mesenchymal transition CAF (CAFEndMT), periph- mor-promoting CAFs.151 In the early stages of cancer develop-
eral nerve-like CAF (CAFPN), and antigen-presenting CAF ment, the tumor-restraining CAFs act as sentinels in close prox-
(CAFap).122 In another study, six CAF subtypes were defined, imity to transformed cells.86,156 This is also bolstered by the fact
shared across melanoma, lung and head and neck cancers, with that blockade of sonic hedgehog (Shh) signaling or selective
characteristics associated with immunosuppressive and stem- deletion of aSMA expressing CAFs resulted in tumor progression
ness attributes.173 Similarly, a multiomics approach comparing in PDAC mouse models.168,169,171 Occurrence of tumor-restrain-
pancreatic, skin and breast cancers has identified three conserved ing CAFs have also been reported in breast, bladder, colon, and
CAF superclusters across species termed steady state-like (PI16 + intestinal cancers167,170,172,181 suggesting that these CAFs may
DPP4+), mechanoresponsive (POSTN+MGP+), and immunomod- be universally present to inhibit tumor initiation.
ulatory CAFs (IL1R1+CXCL12+).174 Interestingly, although steady
state-like and immunomodulatory CAFs originate from normal Cancer progression
mammary fibroblasts, mechanoresponsive CAFs arise during the CAFs play a complex and multifaceted role during the entire
malignancy stage and selective disruption of underlying mechan- course of tumor development, from the pre-neoplastic state until
ical force or immune checkpoint inhibition therapy resulted in dy- the terminal stage of metastasis (Figure 4). Studies have shown
namic changes in CAF distribution that affected tumor growth.174 that tumor cells can educate and recruit subsets of CAFs
A similar observation was also reported where CAF subtypes were residing in close proximity to generate a pro-metastatic
associated with different clinical outcomes allowing identification niche.79,182 This is in addition to the effect of tumor genotype
of molecular pathways involved in tumor progression and therapy in regulating the functional heterogeneity of both normal fibro-
resistance to immune checkpoint blockade.173 This molecular blasts and CAFs.71,183–185 Other examples highlighting the effect
level understanding of the pan-cancer attributes as well as hetero- of genetically altered fibroblasts in tumor promotion come from
geneity and plasticity within CAF subpopulations is still devel- mammary epithelial cells that undergo transformation when
oping, but recent progress being made in defining and character- co-cultured with fibroblasts overexpressing Wingless-type
izing CAFs will revolutionize the way for CAF-targeting therapeutic MMTV integration site family member 1 (Wnt1).186 Similarly,
options in combination with immunotherapies. inactivation of Phosphatase and tensin homolog (PTEN) in mam-
mary fibroblasts significantly enhanced the malignant transfor-
FIBROBLASTS IN CANCER INITIATION AND mation and growth of mammary adenocarcinoma in mice.187
PROGRESSION These Pten-inactivated murine fibroblasts also showed a strong
correlation with CAFs in breast cancer patients. Moreover, TGFb
Cancer initiation receptor II gene knock-out in FSP1-positive cells promotes pros-
The effect of the stromal fibroblasts in supporting and promoting tate intraepithelial neoplasia and fore-stomach squamous cell
cancer development is well documented and although CAFs are carcinoma.188 Further support comes from in vivo experiments
an outcome of the hijacking signals from tumor cells, it has been where highly metastatic mouse mammary cancer cells when in-
established that fibroblast acquisition of a pro-tumorigenic state jected into Fsp1-deficient mouse model exhibit considerably de-
can occur well before a tumor can arise. This is best evident in layed and decreased tumor initiation, whereas the co-injection of
the case of mammary fibroblasts in high mammographic breast FSP1-positive fibroblasts restored tumor formation and pro-
density environment,175 during weaning-induced involution of moted metastasis.189 Earlier studies have also shed light on

1586 Cell 186, April 13, 2023


Review
Table 1. Functional heterogeneity of CAFs in distinct tumor types
Characteristic markers
Cancer type CAF subtypes (Gene expression) Functions Reference
Pancreatic Cancer myCAFs aSMA, VIM, CTGF, COL1A1, Myofibroblast-like, matrix deposition Öhlunde et al.79
(Pancreatic Ductal COL5A1, COL6A1
Adenocarcinoma) iCAFs IL6, IL1, IL11, LIF Inflammatory infiltration, chemokines and
cytokines secretion, tumor-promoting
CAF-A POSTN Tumor proliferation, metastasis Neuzillet et al.147
CAF-B MYH11 Lymph node metastasis
CAF-C PDPN Immune promotion
myCAFs FAP Reduction of T cell infiltration Fearon, D.T.148
Desmoplastic stroma
myCAFs FAP Tumor aggressiveness Kawase et al. and
Looi et al.149,150
myCAFs aSMA Prevent tumor dissemination Miyai et al.151
CAF-FB1 Cxcl14, Ptn, Igf1, Igfbp4/7, Similar to iCAFs Hosein et al.100
Il6, Ccl2/7, Cxcl12, Pdgfra
CAF-FB2 Ly6a, Ly6c1, Nov, Pi16 Physiological functions of fibroblasts
CAF-FB3 Lrrn4, Gas6, Cav1, Cdh11, Similar to myCAFs
Gpm6a, Msln, Lgals7, Nkain4
iCAFs IL6 Reduction of oxidative stress Elyada et al.137
Secretory-CAFs CXCR2 Pro-tumorigenic secretion Awaji et al.152
iCAFs IL6 Inflammation Biffi et al. and Helms
et al.91,153
apCAFs PDGFRa, SAA3 Tumor growth Djurec et al.6
apCAFs HLA-DRA (MHC II genes), Deactivating CD4 T cell Elyada et al.137
CD74, SAA3, SLPI
meCAFs Increased glycolysis, Higher risk of metastasis and poor prognosis Wang et al.142
PLA2G2A, CRABP2 but better response to immunotherapy
Prrx1-CAFs PRRX1 CAF plasticity Feldmann et al.154
LRRC15-CAFs LRRC15 Poor response to immunotherapy Dominguez et al.133
NetG1-CAFs NETG1 Nutritional support (glutamate/glutamine Francescone et al.155
metabolism), immunosuppression
Cell 186, April 13, 2023 1587

Meflin1-CAFs ISLR Tumor suppression Mizutani et al.156


(Continued on next page)

ll
1588 Cell 186, April 13, 2023

Table 1. Continued
Characteristic markers
Cancer type CAF subtypes (Gene expression) Functions Reference

ll
Breast Cancer CAF-S1 CD29, FAP, aSMA, PDGFRb, F Tumor proliferation, EMT promotion, migration, Costa et al. and
SP1 and CXCL12 Lymph node metastasis, immune suppression Pelon et al.58,157
CAF-S2 Not reported Not reported
CAF-S3 CD29, FSP1, PDGFRb Unknown (resemble normal fibroblast)
CAF-S4 CD29, FSP1, PDGFRb and aSMA Tumor invasion, Lymph node metastasis
myCAFs aSMA, TAGLN, MYL9, IGFBP3, TNC Tumor proliferation, Migration, Invasion, Sebastian et al.141
Angiogenesis and EMT
iCAFs Ly6c1, CLEC3B, HAS1, DPT, Tumor proliferation, Metastasis, Angiogenesis,
COL14A1 Immune evasion and Chemoresistance
apCAFs CD74, MHC-II genes, KRT18, FSP1 Antigen-present, Immune modulation
CAFs CD10, GPR77 Promote cancer stemness and Su et al.158
chemoresistance
dCAFs SCRG1, SOX9, SOX10 Not reported Bartoschek et al.81
mCAFs FBLN1, PDGFRa, CXCL14 Immune regulation
vCAFs NID2, NOTCH3, EPAS1, COL18A1, Angiogenesis
NR2F2
CAF-S1 FAPhi CD29med-hi SMAhi ECM and Inflammation signature Kieffer et al.135
MCAMlo Restricted to tumor and Lymph nodes
CAF-S2 FAPneg CD29lo SMAneg Normal fibroblast signature
Tumor and normal tissue
CAF-S3 FAPneg CD29med SMAneg Normal fibroblast signature
Tumor and normal tissue
CAF-S4 FAPneg SMAhi CD29hi Perivascular signature
MCAMhi Restricted to tumor and Lymph nodes
detox-iCAF FAPhi CD29med, ANTXR1( ), Detoxification and inflammatory response
GPC3(+), ADH1B, GPX3
IL-iCAF FAPhi CD29med, ANTXR1( ) Growth factor, TNF signaling, and IL pathway
DLK1(+), RGMA, SCARA5
IFNy-iCAF FAPhi CD29med, ANTXR1( ), Response to IFNg and cytokine-mediated
CCL19, CCL5 signaling pathway,
Similar to apCAFs
ecm-myCAF FAPhi CD29med, ANTXR1(+), ECM secretion
acto-myCAF FAPhi CD29med, ANTXR1(+), Acto-myosin contractility
SDC1(+), GGH, PLP2
IFNab-myCAF FAPhi CD29med, ANTXR1(+), IFNa/b signaling
IFIT3, IRF7

Review
TGFb-myCAF FAPhi CD29med, ANTXR1(+), TGFb signaling pathway and matrisome,
LAMP5(+), CST1, TGFB1 immunoevasion
Wound-myCAF FAPhi CD29med, ANTXR1(+), Assembly of collagen fibrils and wound healing,
CD9(+), SEMA3C, SFRP4 anti-PD1 response
(Continued on next page)
Review
Table 1. Continued
Characteristic markers
Cancer type CAF subtypes (Gene expression) Functions Reference
Colorectal Cancer CAF-A MMP2, DCN, FAP, PDPN ECM remodeling Li et al.83
and COL1A2
CAF-B aSMA, TAGLN, PDGFa, LUM Not reported
Oral Cancer (Oral Squamous CAF-D TGFb EMT, tumor invasion Costea et al.159
Cell Carcinoma) CAF-N Hyaluronan, MMPs Tumor invasion, immunosuppression
Ovarian cancer (High grade pan-CAFs CD49e Not reported Hussain et al.160
serous ovarian carcinoma) Aggressive-CAFs CD49e high Proliferation, migration,
FAP high Lymph node metastasis and therapy resistance
CAF-S1 CD29, FAP, aSMA, FSP1, Tumor proliferation, Immune suppression Givel et al.146
PDGFRb and CXCL12b
CAF-S2 (non-activated) Not reported Not reported
CAF-S3 (non-activated) CD29, FSP1 and PDGFRb Not reported
CAF-S4 CD29, aSMA, FSP1 and Tumor proliferation
PDGFRb
iCAFs IL6, IL10, C1QA/B/C, CFB, Immunomodulation Izar et al.161
CXCL1/2/10/12
Lung Cancer Cluster1 COL10A1 EMT signature Lambrechts et al.143
Cluster2 aSMA Not reported
Cluster4 PLA2G2A Found in the leading edge of tumor
Cluster5 MMP3 Lower myogenesis
Cluster7 CCL2 High mTOR signature
CAF-1 FAP(+) Higher-stage tumors Grout et al.162
CAF-2 FAP(+), aSMA(+) Higher-stage tumors, ECM and contractility,
Immune cell exclusion
CAF-3 MYH11(+), aSMA(+), FAP( ), Low-stage tumors, ECM and contractility,
ADH1B( ) Immune cell exclusion
CAF-4 ADH1B(+) Low-stage tumors, T cell enrichment
HD-CAFs ST8SIA2, WNT7B, SLITRK6, higher matrix reorganizing ability and Hao et al.163
and IGFL3 promotion of tumor cell invasion and growth
LD-CAFs Negative for HD-CAFs markers Low desmoplastic
Cell 186, April 13, 2023 1589

CAF7 FAP(+), PDGFRa( ) Immune suppressive Pellinen et al.164


CAF13 FAP( ), PDGFRa(+) Good prognosis
CAF-I HGFhi, FGF7hi/lo, pSMAD2lo Support tumor cells therapy resistance Hu et al.165
lo hi lo
CAF-II HGF , FGF7 , pSMAD2 Support tumor cells therapy resistance
CAF-III HGFlo, FGF7lo, pSMAD2hi Better clinical response

ll
CAFs CD10, GPR77 Chemoresistance and poor survival Su et al.158
apCAFs MHCII(+), FAP(+), CD45( ) Tumor-suppressive Kerdidani et al.144
(Continued on next page)
1590 Cell 186, April 13, 2023

ll
Table 1. Continued
Characteristic markers
Cancer type CAF subtypes (Gene expression) Functions Reference
Head and Neck Squamous CAF-1 FAP, PDPN, COL1A1, VIM, Lymph node metastasis, promote EMT Puram et al.61
Cell Carcinoma THY1, MMP11, CAV1
CAF-2 FAP, PDPN, JUN, FOS, Not reported
FGF7, TGFbR
CAF3 Not reported Normal fibroblasts
Intrahepatic Cholangiocarcinoma EMT-like CAFs KRT19 Epithelial-like Zhang et al.166
(ICC) myCAFs POSTN, COL5A1 ECM modulation
vCAFs MCAM, IL6 Angiogenesis
apCAFs MHC-II genes, CD74 Antigen presenting, immunomodulation
Oral/pancreatic/Colon/Bladder Restraining-CAFs BMP4, Anti-tumoral effect Gerling et al.; Lee et al.;
Hedgehog signaling Özdemir et al.; Pallangyo
Intestinal cancers IKKb signaling et al.; Rhim et al. and
Shin et al.167–172
VIM vimentin, CTGF connective tissue growth factor, FAPa fibroblast activation protein-a, BMP4 bone morphogenetic protein-4, MCAM melanoma cell adhesion molecule, KRT keratin, POSTN
periostin, COL collagen, MHC major histocompatibility complex, FGF fibroblast growth factor , PDPN podoplanin, MMP matrix metalloprotease, EMT epithelial to mesenchymal transition,
GPR77 complement component 5a receptor 2, HGF hepatocyte growth factor, aSMA a smooth muscle actin, Prrx paired related homeobox, TAGLN transgelin, IL interleukin, TGF transforming
growth factor, PDGF platelet-derived growth factor, PDGFR PDGF receptor, THY1 CD90 cell surface antigen, FSP-1 fibroblast specific protein-1, CXCL chemokine (C-X-C motif) ligand, ADH
alcohol dehydrogenase, CFB complement factor B, MYH11 myosin heavy chain-11, ST8SIA2 ST8 alpha-N-acetyl-neuraminide alpha-2,8-sialyltransferase 2, WNT7B Wnt family member 7B,
SLITRK6 SLIT and NTRK like family member-6, IGF insulin-like growth factor (IGF), IGFL3 like family member-3, CAV1 caveolin-1, LUM lumican, SEMA3C semaphorin 3C, SFRP4 secreted friz-
zled related protein-4, C1Q complement component 1 Q subcomponent, DCN decorin, LAMP5 lysosomal-associated membrane protein family, member 5, ANTXR1 ANTXR cell adhesion
molecule-1, MYL9 myosin light chain-9, IGFBP IGF binding protein, TNC tenascin, GGH gamma-glutamyl hydrolase, PLP2 proteolipid protein-2, IFIT3 interferon induced protein with tetratri-
copeptide repeats-3, IRF7 interferon regulatory factor-7, CST1 cystatin SN, SDC1 syndecan-1, GPX3 glutathione peroxidase 3, HAS1 hyaluronan synthase-1, DPT dermatopontin, SCRG1 stim-
ulator of chondrogenesis-1, SOX SRY (sex determining region Y)-box, FBLN1 fibulin-1, NR2F2 nuclear receptor subfamily 2, group F, member 2, NID2 nidogen-2, EPAS1 endothelial PAS domain
protein-1, PI16 peptidase inhibitor-16, SLPI Calcineurin-like metallo-phosphoesterase superfamily protein, SAA3 serum amyloid A3, LIF leukemia inhibitory factor, Ptn pleiotrophin, Lrr4 leucine
rich repeat neuronal-4, Gas6 growth arrest specific-6, Cdh11 cadherin-11, Gpm6a glycoprotein M6A, Msln mesothelin, Lgals7 galectin-7, Nkain4 Na+/K+ transporting ATPase interacting-4,
NETG1 netrinG1.

Review
ll
Review
Figure 3. Functional heterogeneity in can-
cer-associated fibroblasts
Cancer-associated fibroblasts (CAFs) represent
a heterogeneous cellular population within
the tumor microenvironment. Single-cell RNA
sequencing and multiplex imaging have revealed
different CAF subtypes across multiple cancer
types. These CAFs are divided into 4 main groups:
iCAFs (inflammatory CAFs), myCAFs (myofibro-
blastic CAFs), apCAFs (antigen presenting CAFs)
and vCAFs (vascular CAFs). The cellular function,
plasticity and the heterogeneity in the molecular
marker expression associated with these CAFs
are indicated.

and IL22), MAPK (IL6, TGFb, and FGF),


or Hippo (EVs).1,193 These factors also
increase the survival of tumor cells via
upregulation of anti-apoptotic proteins
like B-cell lymphoma-2 (BCL2) and
myeloid cell leukemia-1 (MCL1) or down-
regulation of pro-apoptotic proteins like
BCL2 Associated X protein (BAX).194 In
other situations, fibroblasts succumb to
epigenetic changes (aging, senescence-
induced) and external stressors that ulti-
mately transform them to affect tumor
cell behavior. For example, generation
of reactive oxygen species (ROS) in
an inflammatory TME promotes fibro-
blast activation toward tumor-promoting
functions.3,53

Cancer metastasis
Cancer is characterized by uncontrolled
cellular growth giving rise to a tumor at
the site of origin. The tumor is considered
benign if it lacks the ability to invade other
the signaling cascades that operate as a consequence to the neighboring. However, when the tumor resists the constraints of
body’s response to cancer, giving way to a tumor-supportive space it starts to spread and infiltrate neighboring normal tissues
local microenvironment at distal and metastatic sites.190,191 becoming malignant.195 This spread to distant sites via angio-
Further support comes from the role of senescent fibroblasts in genic, or lymphatic system is called metastasis and is the prom-
abetting cancer progression from premalignant cells as reported inent cause of death from cancer. This multistep process requires
in skin cancer model where senescent fibroblast-mediated os- the migration of tumor cells from the primary site, their seeding in
teopontin (OPN) secretion promoted tumorigenesis.192 a distal organ, and ultimately outgrowth of those seeded cells. To
do that the tumor cells, must first move through tissues via the
Cancer growth and stemness meshwork of matrix.10,196 Moreover, migrating tumor cells have
In addition to modulating ECM components, CAFs promote a to maintain survival while penetrating layers of tightly connected
conducive environment for tumor proliferation as a result of para- epithelial and endothelial cells and breach several structural and
crine interactions between tumor cells and CAFs. CAFs secrete a blood flow barriers.197 During the dissemination across lymphatic
range of growth factors and cytokines that influence the stem- and vascular boundaries, tumor cells make and break contacts
ness behavior of the epithelium, including hepatocyte growth with different proteins and navigate their way before finally
factor (HGF), EGF, insulin-like growth factor (IGFs), IGF-binding settling at a new site where they either undergo dormancy or
protein (IGFBPs), FGF2, OPN, and TGFb. CAFs, either directly continue to grow and proliferate with decisions dependent on
or through EV secretion, additionally provide lipids and metabo- the age, metabolic health, environmental stressors, and co-mor-
lites that are taken up by cancer cells for effective prolifera- bidities of the host.53,198,199 This progression from an isolated tu-
tion3,53 in vitro and tumor growth in vivo via activation of their mor to disseminated metastatic disease is a multistep process,
respective signaling pathways, i.e., integrin/FAK-Src (POSTN), with each step involving intricate cross talk between the cancer
WNT/b-catenin (HGF and OPN), PI3K/mTOR (CXCL12, HGF, cells and the surrounding tumor microenvironment (TME) and

Cell 186, April 13, 2023 1591


ll
Review

Figure 4. Role of fibroblasts in carcinogenesis


The phenotype and functions of fibroblasts are in dynamic evolution along with tumor initiation and progression. During early stages of carcinogenesis, pro-
tumorigenic fibroblasts can drive tumor initiation and promote proliferation of mutated cancer cells. These activated fibroblasts in close proximity to the tumor
cells can then transition to cancer-associated fibroblasts (CAFs). During this initial phase of cancer development, the heterogeneous cancer-associated
fibroblast (CAF) subpopulations, referred to as tumor-restraining CAFs, can potentially protect normal tissue against cancer invasiveness through the various
indicated mechanisms. However, in advanced-stage disease, tumor cells and other stromal cells reprogram the CAFs via continuous crosstalk to promote a
tumor permissive niche. This allows tumor cells to proliferate and grow through dynamic processes that includes ECM remodeling, growth factors, cytokines and
extracellular vesicles secretion, metabolic reprogramming and creating an immunosuppressive tumor microenvironment (TME). The mechanical coupling be-
tween CAFs and cancer cells, permits local invasion and regional dissemination of tumor cells from the primary tumor site. As the tumor evolves, CAFs reorganize
the TME by promoting angiogenesis and epithelial-to-mesenchymal transition of tumor cells enabling distal metastasis (shown here with lung as an example)
following intra- and extravasation. These disseminated tumor cells also overcome immune surveillance by resident immune cells, and direct immune sup-
pression. At the metastatic site, the disseminated tumor cells, in the face of contextual signals, either enter or exit dormancy. This is governed by a plethora of
microenvironmental cues, encompassing the cellular, molecular, and physical factors converge to induce either stress-related or mitogenic signals to tumor cells.
Upon reawakening, proliferating cancer cells then colonize the metastatic niche forming macrometastases, co-opting and recruiting local stromal cells to further
support cell growth by indicated mechanisms. Subsequently, fibroblasts and CAFs continue to play important roles across the continuum of tumor progression
and metastasis to visceral sites.

acellular extracellular matrix (ECM).200–202 Numerous studies mary tumor and facilitates metastasis to distant organs. Further,
have now shown that TME regulates this tumor behavior from recent studies have shown that CAFs like cancer cells can also
initiation to progression by influencing cancer cell invasion in a disseminate via circulation95 to distant metastatic sites as indi-
qualitative and quantitative manner. vidual CAFs or in clusters with cancer cells, suggesting that
In addition to the local tumor growth, CAFs have been shown they have even more complex roles in metastasis.203 Crosstalk
to play an integral role in cancer invasion across the epithelial between CAFs and cancer cells causes changes in the ECM
and endothelial basement membranes, which typically involves components and structural scaffolding within the TME as well
coordinated adhesive and proteolytic activities.3 CAF-mediated as induces EMT in cancer cells imparting cell motility and
stromagenesis drives increased cancer invasiveness at the pri- increasing invasive capacity.204 Beyond tumor invasion, CAFs

1592 Cell 186, April 13, 2023


ll
Review

can also play a vital role in the colonization of distant organs in of endothelial cells and pericytes tumor to drive invasion and
metastatic disease, by creating a niche for tumor cells. CAFs metastasis.218–222 Autocrine signaling through PDGF and VEGF
have been shown to provide stromal support for disseminated further promotes the production of pro-angiogenic molecules
cancer cells through a combination of environmental factors, IL6, and IL8.223–226 Elevated levels of WNT2 selectively in colon
including POSTN and tenascin-C (TNC) proteins.205 In addition, CAFs has been shown to increase angiogenesis by secreting
TGFb is an established inducer of EMT through paracrine pro-angiogenic molecules and ECM.227 Additionally, chemokine
signaling, allowing premalignant cells to acquire mesenchymal production from CAFs has also been shown to induce vasculo-
properties for invasion and metastasis.197 Further, CAF expres- genic mimicry by actions of TGFb and CXCL12.228 CAFs ex-
sion of caveolin-1 was shown to promote matrix remodeling, pressing connective tissue growth factor (CTGF) were shown to
invasiveness, and increased metastases in mice injected with increase the micro-vessel density and tumor growth activity in
breast adenocarcinoma cells.206 Further, anisotropic fibronectin a prostate cancer xenograft model.229 CAFs mediated ECM syn-
organization orchestrated by CAFs also provides directions for thesis and remodeling also generates solid stress resulting in the
cell migration.207 Additionally, in some cases CAFs have been vascular and lymphatic vessel compression, reduction in perfu-
shown to migrate with metastasizing cells and remodel the sion rates and increased hypoxia. This generation of hypoxic
ECM, as well as the metastatic site’s milieu, to sustain growth TME by CAFs gives rise to aggressive clones of tumor cells by
and survival of the tumor cells54 (Figure 4). formation of new vasculature and mediating an angiogenic
switch in addition to generating an immunosuppressive environ-
CAF regulation of ECM ment.230–232 However, chronic hypoxia has been shown to
In the early stages of tumor growth, the host tissue architecture dampen the ECM remodeling by head and neck and vulval
gets distorted from the aberrant accumulation of ECM compo- CAFs and revert their phenotype.233 In addition to the growth fac-
nents. CAFs are the most prominent ECM-producing cells in tors, angiogenesis is also promoted by degradation of ECM com-
TME that together with immune cells contribute to desmoplasia ponents and basement membrane driven by CAFs secreted
and matrix-remodeling via MMPs. In PDAC for instance, the MMPs.234,235 Finally, CAFs can enhance vascular growth addi-
accumulation of thick desmoplastic stroma that also contains tionally through biomechanical forces and involving the Rho/
high amounts of hyaluronan, promotes tumor growth in mice ROCK pathway and YAP signaling.236
and correlates significantly with poor prognosis in patients.28,208
In addition to altered biochemical signals provided by the des- CAF regulation of anti-tumor immunity
moplastic TME, the production and re-assembly of ECM, The role of CAFs in immunomodulation of cancer tissue serves
including linear collagen structures, alters the physical proper- two primary purposes: CAFs facilitate tumorigenesis by inducing
ties and biomechanical activity of the microenvironment.209 a chronic inflammatory state for cancer cells, and by establishing
Desmoplasia induces tumor stiffness via processes closely an immunosuppressive TME to maintain survival. This is made
attributed to the over-activation of lysyl oxidase (LOX), an possible by secretion of immunomodulating cytokines such as
enzyme that crosslinks collagen and other ECM compo- TGFb, IL6, CXCL1, and CXCL12.134–137 Additionally, CAFs can
nents.210,211 The increased tumor stiffness promotes integrin- express molecules such as netrinG1,155 and signal to immuno-
based focal adhesion assembly and increases the formation of suppressive populations, such as neutrophils and myeloid-
adhesion complexes in both cancer and stromal cells, thus derived suppressor cells (MDSCs), which can further promote
creating an increasingly pro-tumorigenic microenvironment. conversion into CAFs.237–239 A direct outcome of crosstalk be-
Increased integrin activity in CAFs transduces mechanical forces tween tumor cells and CAFs through TGFb signaling is the reduc-
that further change the orientation of collagen and fibronectin tion in cytotoxic T cells but increase in infiltration of Tregs as well
fibers, promoting cancer growth and invasion.200,207,212 In gen- as the recruitment and enhanced differentiation of macrophages
eral, the generation of physical forces and stiffness-dependent into tumor-associated macrophages (TAMs) resulting in immuno-
cytoskeletal rearrangements are tightly linked to the dysregula- therapy failure.240–242 CAFs work closely with TAMs in providing
tion of YAP1 transcriptional co-activator in fibroblasts and can- metastatic niches for tumor cells, by facilitating the uptake of tu-
cer cells, which leads to transcriptional programs to further mor derived EVs by TAMs and migrating to the metastatic site to
potentiate CAF activation and cancer cell growth.92 Excessive prepare for tumor growth.243–245 Finally, CAFs may directly inter-
stroma acts as a barrier to drug delivery and results in excessive fere with anti-tumoral role of natural killer (NK) cells through pros-
interstitial fluid pressure caused by compression of the blood taglandin E2 (PGE2) secretion or MMPs-mediated decrease in
and lymphatic vessels.213 Further, ECM-altering properties of NK activating receptor ligands in tumor cells, thus reducing NK
CAFs allow prolonged secretion of inflammatory cytokines receptor-dependent cytotoxic activity.246,247
such as IL8, IL6 and leukemia inhibitory factor (LIF) by tumor
cells, which in turn sustains the pulsatile TGFb and STAT3 CAF regulation of cancer cell metabolism
signaling as reported in the pancreatic cancer TME.214–217 These Metabolic adaptations in tumors require access to amino acids,
events further accelerate proliferation and phenotypic switch of nucleotides, lipids to assist with their growth and metastatic
CAFs into a tumor-promoting phenotype. needs. This is also reflected in metabolic heterogeneity within
the tumor owing to dynamic EMT (or phenotype switch) pro-
CAF regulation of angiogenesis cesses, as some tumor cells demand higher nutrients and switch
CAFs secrete several angiogenic molecules like VEGF, PDGF, to alternative carbon sources such as glutamine.248 CAFs also
CXCL12, or HGF that can promote proliferation and recruitment contribute to metabolic heterogeneity based on their spatial

Cell 186, April 13, 2023 1593


ll
Review

location within the tumor as reported in CAFs from melanoma, reprogramming by sequestering TGFb in an inactive state.269,270
breast, colon, and pancreatic cancers.76,249–251 This also en- Further evidence of tumor reawakening comes from normal aged
compasses the synergistic crosstalk between CAFs and other lung fibroblasts271 and activated fibroblast -mediated produc-
cell types within the TME, to meet constant energy requirements tion of POSTN at the micrometastatic site, which promotes not
for tumor growth, invasion, and metastasis, can also be met by just colonization but also maintenance of stem-cell like proper-
autophagy, lipid secretion and ECM remodeling. ties in the cancer cells, necessary for tumor initiation and
growth.11 However, considering only a few invasive cancer cells
CAF regulation of therapeutic resistance reach distal sites, a question remains regarding the phenotypic
One of the most intriguing aspects of CAFs is their contribu- and secretory behavior of these CAFs at distal sites: are they
tion to therapeutic resistance in several different cancers. programmed by cancer cells or an outcome of epigenetic
Several studies have indicated that CAFs promote chemoresist- changes in the surrounding fibroblasts at the metastatic niche.
ance via different mechanisms such as secretion of cytokine Interestingly, an earlier study has suggested the possibility that
and miRNAs, increase in cancer stem cells (CSCs), and tumor cells can carry stromal cells during metastasis.203
decreased drug bioavailability via production of a stromal
barrier.71,110,111,216,252–256 In breast, colorectal and pancreatic tu- AGING AND FIBROBLASTS
mors, CAFs promote the resistance to chemotherapeutic agents
like gemcitabine and doxorubicin and to combination chemo- Genomic instability is a shared hallmark of aging and cancer272
therapy like doxorubicin, 5-fluorouracil and cisplatin through the and age-mediated accumulation of alterations in normal epithe-
production of IL6, IL17A, PDGF, and insulin like growth factor lium can initiate carcinogenesis and induce malignancy.273,274
(IGF) that activate the NF-kB and ERK1/2 pathways in tumor Whereas several somatic mutations are detected in cancer cells,
cells.194,257–260 CAFs are also a source of extracellular vesicles no somatic genetic alterations have been reported in fibro-
(EVs) that contain regulatory microRNAs, which can in turn pro- blasts,99–102 suggesting a strong contribution from epigenetic
mote resistance to cisplatin and paclitaxel in cancer cells by changes. More importantly, recognizing the role of intrinsic/
downregulating p21 expression and ferroptosis.261,262 healthy aging in fibroblast biology is especially critical now
more than ever, considering aging itself has important implica-
FIBROBLASTS IN CANCER DORMANCY AND tions across every stage of tumorigenesis; from initiation to
REAWAKENING metastasis, anti-tumor immunity, chemoresistance and cancer
dormancy to reawakening.1,53 It is therefore not surprising that
Subsequent to metastatic dissemination, cancer cells can aging in addition to the individual’s physiological state can expe-
remain latent and stay undetected for years before emerging dite the transition of normal fibroblasts to an activated pro-
as a proliferative and aggressive disease.263 There is now tumorigenic state and eventually to CAFs. In that regard, aging
emerging evidence that connects fibroblasts to tumor dormancy is enforced by the accumulation of senescent fibroblasts at the
but precedent from pancreatic cancer studies supports that tissue level, which ultimately results in the functional impairment
depletion of CAFs results in a more aggressive pheno- and eventually predisposition to emergence of malignancies
type.168,169,171 It is therefore reasonable to assume that there among other diseases. This is evident across most cancer types
also exists a subpopulation of CAFs associated with the entry where intrinsic aging in conjunction with the individual’s genetic
into or awakening from cancer dormancy. CAFs by nature background and extrinsic stressors, like UV irradiation, smoking
secrete a milieu of soluble and insoluble factors, the same can history, and socioeconomic background increases susceptibility
be responsible for initiating dormancy or their reawakening in a to cancer21,275 (Figure 5).
tissue-dependent manner. In that regard, several cytokines Increasing age is inversely proportional to fibroblast abun-
TGFb, IFNb, IL8, have been documented to contribute to main- dance as well as ECM integrity.276–278 Analyses of dermal fibro-
tenance of cancer cells’ quiescence at distal metastatic sites.1,11 blasts from young and old mice has revealed age-mediated
In addition, owing to their high capacity of ECM synthesis and variability in wound healing, also evident in elderly patients.279
remodeling, tumor stiffness can also regulate cancer cell In addition, numerous studies have highlighted that age is asso-
dormancy by constricting growth of tumor cells especially as ciated with characteristic cellular attributes associated with ge-
they invade from tissues of variable or contrasting stiffness.264 netic damage, telomere attrition, and inflammation.1,272,280 In
This has been shown using a 3D fibrin gel system in melanoma that regard, scRNA-seq analysis of mouse dermis has revealed
cells265 and an ECM-mimicking semi-solid Matrigel in lung that aged fibroblasts exhibit transcriptional noise while partially
adenocarcinoma cells266 resulting in an induced dormancy losing their identity and gaining adipogenic traits.281 This was
state. Part of the dormancy program is the gain of drug-resis- also supported in human dermal fibroblasts where aged fibro-
tance mechanisms as they await further signals from the sur- blasts showed partial loss in their cell identity while exhibiting
rounding microenvironment before switching to a proliferative differential secretory, mesenchymal, and pro-inflammatory func-
state and spreading to distal sites. In addition to ECM remodel- tional annotations as compared to young fibroblasts.282 Impor-
ing, its composition can also determine the fate of the dormant tantly, secreted factors and ECM modulation mediated by these
cancer cells as both type I collagen and fibronectin have been intrinsically aged dermal fibroblasts supported melanoma
shown to promote proliferation via b1-integrin induced PI3K/ progression.255,276,283
AKT and FAK activation.267,268 Similarly, TNC, which is increas- Previous evidence has indicated that senescent non-malig-
ingly in the metastatic niche, could inhibit pro-dormancy cellular nant cells exhibit SASP acquisition following a wide range of

1594 Cell 186, April 13, 2023


ll
Review

Figure 5. Aging, cancer and fibroblasts


Fibroblasts have been shown to play tumor-promoting roles in cancer during aging. This can occur when fibroblasts become senescent, or prematurely aged, due
to replicative stress or oxidative stress and DNA damage, or in response to therapeutic interventions or when fibroblasts age normally (epigenetic changes).
Secreted changes that occur during senescence such as the acquisition of senescence-associated secretory phenotype (SASP) as well as the aged fibroblasts
have been shown to promote tumor progression and therapy resistance by modulating the immune microenvironment, driving multiple changes including pro-
angiogenesis, ECM remodeling, metabolic changes that also impact the outgrowth of tumor cells at metastatic sites. However, an important distinguishing factor
between senescent fibroblasts and aged CAFs is that, whereas senescence promotes growth arrest, CAFs continue to proliferate. It remains unknown if aged
CAFs resemble quiescent senescent CAFs or can proliferate via crosstalk with the surrounding tumor cells.

cellular stressors, including persistent DNA damage, epigenomic igenic.287,288 It is also important to note that SASP factors are not
perturbations, therapy-induced toxicity, and oxidative stress.284 unique to senescent cells but also expressed in non-senescent,
Although senescence is a context-dependent phenomenon, it is proliferating cells such as CAFs.284 This would suggest that aging
characterized by cell-cycle arrest, a flattened morphology induced senescence and SASP may serve as a precursor in at-
in vitro, heterochromatin changes, and expression of senes- taining the CAF phenotype (aged CAFs) earlier in aged vs. young
cence-associated b-galactosidase (SA b-gal).284–286 An impor- TME and that these aged CAFs would also lack any tumor-re-
tant aspect of senescence is the acquisition of senescence-asso- straining function since they are primed to be pro-tumorigenic.
ciated secretory phenotype (SASP) resulting in production of a Hence, these aged-CAFs can expedite tumor progression mainly
range of secreted factors. In both young and healthy tissues, by the secretion of SASP factors, which can cause chronic
SASP plays a critical role in directing and recruiting immune cells inflammation, promote angiogenesis, and enhance immunosup-
for clearance of senescent fibroblast cells and terminating inflam- pressive activity,289,290 features of an aged TME53 (Figure 5).
mation.53,272 However, with age the senescent fibroblasts ac- However, contrary to their dividing counterparts in a young
quire SASP, which disrupts this balance making them pro-tumor- TME, it is unknown if aged CAFs retain sufficient plasticity to

Cell 186, April 13, 2023 1595


ll
Review

regain the ability to proliferate due to crosstalk with tumor cells explored to target CAFs alone or in combination with existing
and surrounding ECM. But it is highly likely that the aged CAFs chemo- or immuno-therapies. These strategies (Table 2) can
by virtue can act as quiescent CAFs and maintain and propagate be stratified as: (a) using cell surface markers to eliminate or limit
senescence (and SASP) as well awaken dormant tumor cells at the presence of CAFs, (b) switching CAFs to a dormant state,
distal sites and impart therapy resistance, an attribute particularly (c) antagonizing or sequestering the CAF secretome
common in aged patients.271 A recent study has identified two (d) targeting the signaling pathways that activate CAFs,
aging CAF gene signatures in low grade glioma associated with (e) targeting the downstream elements that are crucial for CAF
distinct prognosis and TME characteristics.291 It remains to be function, and (f) targeting ECM proteins. Although various pre-
assessed if a shared aged CAFs signature exists, which is appli- clinical and clinical studies in targeting CAFs are ongoing, the re-
cable to other cancer types. Future research and technological sults have been inconsistent, suggesting that caution should be
advances will make it plausible to elucidate biomarkers for pre- exercised when using CAF-directed therapies.
dicting prognosis and therapy response.
SUMMARY AND FUTURE OUTLOOK
FIBROBLAST PLASTICITY
The ability of tumor cells to defy the basic cellular principles and
It is established that CAFs show distinct functional states sug- norms that apply to tissue homeostasis but grow uncontrollably is
gesting that the CAF subtypes are cell states rather than end- well recognized. However, as numerous therapeutic modalities
points in differentiation.1 More importantly, this plasticity and regimens have demonstrated, approaches to eliminate the
observed in CAFs is not unique, but an extension of the same tumor "seed’’, or discard the ECM ‘‘soil’’ have been discouraging.
behavior evident in normal fibroblasts.79,91,137,292 This has It has become apparent that the CAF component within the TME
been evident in dermal fibroblasts that change from reticular to is actively involved in tumor initiation, dissemination, drug resis-
papillary permitting ECM production or proliferation, respec- tance and even extending to tumor dormancy and reawakening.
tively and eventually tumor invasion.46,293 This plasticity has This requires a better understanding of the heterogeneous nature
also been reported during lung fibrosis294 and is subjected to and complexity within the CAF populations. The identity and pro-
epigenetic regulation and dynamic state transitions.57,295,296 portion of CAF subtypes also differ across normal, inflammatory,
There is also remarkable diversity in the tissue microenviron- premalignant, and malignant states; however, it is important to
ments that contain fibroblasts, and these niches can change note that both normal fibroblasts and CAFs are governed by
dramatically following injury, as is the case during reprogram- the same set of principles regarding the tissue state, local and
ming of myofibroblasts, into adipocytes,.297 In addition to these regional variation and most importantly the host attributes of in-
parameters, the plasticity in CAFs also occurs in response to tersectionality (age, sex, ethnicity to name a few).
crosstalk with tumor cells or as collateral during therapeutic in- Although elegant lineage tracing studies have discovered the
terventions.298 This was shown in PDAC mouse model where origin of fibroblasts in normal or fibrotic tissues, not enough is
CAF reprogramming in the presence of JAK inhibitor shifted known about the origin of CAFs. Technological advances in sin-
the iCAFs to myCAFs subtype.91 Other studies have shown gle-cell resolution and bio-fabricated 3D models have unveiled
that KRAS mutation and p53 mutational status in PDAC can incredible insights into cellular heterogeneity and interactions
also influence CAFs resulting in enhanced desmoplastic stroma with other cell types, particularly an unexpected diversity
and ECM remodeling.171,299 amongst the fibroblasts at the level of molecular marker expres-
It is increasingly recognized that sufficient plasticity also exists sion. New strategies to stratify fibroblasts that are guided by their
among some CAF subsets and is even more pronounced during functional diversity rather than the expression of specific marker
tumor progression.122,300,301 For example, in vitro experiments genes are essential to understand and compare fibroblast het-
have shown that iCAFs can be converted to myCAFs when erogeneity in different organ systems and across different spe-
cultured in a monolayer.79 Independent of the switch mecha- cies. We are only beginning to understand the functional rele-
nism, the induction of iCAF leads to elevated production of cyto- vance of this dynamic and spatiotemporal heterogeneity, for
kines and chemokines, including VEGFA, PDGF, IL6, tumor organ function in homeostasis and cancer biology, and the chal-
necrosis factor (TNF), NF-kB, HGF, CXCL12.1 Furthermore, the lenges are numerous. Finally, translating the discoveries in animal
responsiveness of matrix production by fibroblasts and the models to the human setting is a difficult hurdle. Rodent models
aSMA promoter to a range of extracellular cues, including sub- are impressively establishing the trajectory of certain cell types
strate stiffness, supports the idea that the aSMA-high, matrix- toward a pathological phenotype, but this is difficult to replicate
producing-high state is reversible.302,303 Considering the hetero- in human tissues. Together, CAFs have emerged as dynamic
geneity and plasticity of CAFs, it remains to be seen alteration of players in the TME. Although challenges remain, the field of
this behavior can be made beneficial by converting their tumor- CAF biology is on the cusp of the next big discovery that will facil-
promotion behavior to tumor restraining. itate tailored targeting of cancer types or stages, if not alone, then
as combination strategy to optimize clinical benefit.
THERAPEUTIC TARGETING OF CAFs
ACKNOWLEDGMENTS
Recent advances in CAF biology have led to the development of
new therapeutic strategies targeting the tumor stroma, particu- We apologize for omitting relevant works and citations due to space con-
larly the CAFs.16,296,304 Novel avenues are actively being straints. A.T.W. is also supported by P01CA114046 (NCI), U01CA227550

1596 Cell 186, April 13, 2023


Review
Table 2. Strategies directed against CAFs
Drug Cancer type Clinical Stage Mode of action (pathways/molecules) Reference
aFAP-PE38 Breast Preclinical Blocks FAPa Fang et al.305
Sibrotuzumab Phase I
Pan-FGFR inhibitors Urothelial, Phase 1 Inhibits CAF activation Formisano et al.; Guagnano et al.;
(Erdafitinib, BAY1163877, NSCLC, esophageal, Phase 2 Kim et al.; Palakurthi et al. and
BGJ398) cholangiocarcinoma Perera et al.306–310
Breast, HCC,
Navitoclax Cholangiocarcinoma Preclinical BCL2 inhibitor to initiate cell death Mertens et al.311
PT630 Lung Preclinical FAP deletion Santos et al.312
ProAgio Breast, Preclinical Integrin avb3 (reduces collagen, decreases Sharma et al. and Turaga et al.313,314
Pancreatic CAF secretome)
HA-PTX Breast, solid tumors Preclinical MMP (suppresses angiogenesis, ECM Chiappori et al.; Heo et al. and Yu
S-3304 Phase I degradation) et al.315–317
Astragaloside IV Gastric Preclinical miR-214 and miR-301a (reduce oncogenic Wang et al.318
SOX2, NANOG)
Fasudil PDAC Preclinical Rho/ROCK inhibitor Huo et al. and Whatcott et al.319,320
SCLC
NT157 Colorectal Preclinical IGF1R and STAT3 inhibitor Sanchez-Lopez et al.321
sTRAIL LPD Solid tumors Preclinical Induces fibroblast reprogramming Miao et al.322
Dasatinib Lung Preclinical PDGFR inhibitor Haubeiss et al.323
Vitamin D receptor ligand PDAC Preclinical Reprograms pancreatic stellate cells to Sherman et al.324
(Paricalcitol) quiescence
EB1089, LE135 Squamous cell carcinoma Preclinical Increases apoptosis by blocking Vitamin D Chan et al.325
Receptor/RARb
AC1MMYR2 Breast, gastric, glioblastoma Preclinical microRNA-21 maturation inhibitor Ren et al.326
Ruxolitinib Head and neck, lung, breast Phase 2 JAK/STAT pathway and DNA Albrengues et al.295
methyltransferase activity inhibitor
Minnelide PDAC Preclinical TGFb signaling inhibitor Dauer et al.327
All-trans-retinoic acid (ATRA) Breast Phase 1 Downregulates actomyosin contractility via Chronopoulos et al.328
PDAC, Prostate MLC2
Reprograms CAFs to quiescence
HA-PTX/MATT LY2157299 Breast, ovary Preclinical Blocks CAF activation by suppressing Lv et al. and Zhang et al.329,330
TGFb signaling and inhibiting TGFb-R1
Cell 186, April 13, 2023 1597

GKT137831 Head and neck, colorectal Preclinical Transdifferentiation to CAFs by Hanley et al.331
inhibiting NOX4
Artemisinin derivative Breast Preclinical Suppresses TGFb pathway Yao et al.332
Tranilast Lung Preclinical Suppresses proliferation and TGFb release Ohshio et al.333
Pirfenidone Pancreatic, NSCLC Phase 1 Suppresses proliferation and downregulate Mediavilla-Varela et al.334

ll
LY210976 Breast, liver Preclinical TGFb, PDGF and collagen synthesis
Bevacizumab Pleural mesothelioma Phase 3 Inhibits angiogenesis via VEGF Zalcman et al.335
(Continued on next page)
1598 Cell 186, April 13, 2023

ll
Table 2. Continued
Drug Cancer type Clinical Stage Mode of action (pathways/molecules) Reference
Metformin Ovarian Preclinical NF-kB inhibitor and inhibit IL6 secretion Xu et al.336
Imatinib Cervical Preclinical PDGFR inhbitor Pietras et al.226
Sonidegib TNBC Phase I Hedgehog pathway inhibitor Cazet et al.214
Talabostat Colorectal Phase 2 FAP activity inhibitor Narra et al.337
Pasireotide PDAC Phase 1 MTOR/4E-BP1 protein synthesis inhibitor Duluc et al.338
Losartan Breast, stomach Preclinical Collagen 1 synthesis inhibitor Diop-Frimpong et al.339
AMD3100 Gastric Preclinical CXCL12/CXCR4 signaling inhibitor Izumi et al.340
5-Fluorouracil Lung Blocks CAF recruitment Ma et al.341
JNJ-40346527 Lung, breast, colon Preclinical Blocks PMN-MDSC recruitment via CSF1R Kumar et al.342
SB225002 and CXCR2 respectively
Disulfiram/Copper Nasopharynx Preclinical Induces cytotoxic effects in CAFs through Li et al.343
ROS/MAPK and ferroptosis pathway
MORAb-004 Colorectal Phase 2 Blocks CD248 receptor-ligand interaction Diaz et al.344
131
I-m81C6 Brain Phase 2 Delays tumor growth by blocking tenascin Reardon et al.345
Anti-GPR77 Breast Preclinical Improve chemosensitivity Su et al.158
IPI-926 + Gemcitabine Pancreatic Preclinical Depletes CAF by reducing stroma Ko et al. and Olive et al.346,347
Phase 1 (discontinued)
Tocilizumab Gastric Preclinical Reduces IL6 Ham et al.348
PEGPH20 Pancreatic Phase 2 Increases drug delivery by targeting Hingorani et al. and Thompson et al.349,350
hyaluronan
ATRA all-trans-retinoic acid, CXCL12 CXC motif ligand 12, CXCR4 CXC motif chemokine receptor 4, IGF1R insulin-like growth factor type 1 receptor, IL6 interleukin-6, FAP fibroblast activation
protein, JAK Janus kinase, MLC2 myosin light chain 2, mTOR mammalian target of rapamycin, NF-kB nuclear factor kB, NSCLC non-small-cell lung cancer, PDAC pancreatic ductal adenocar-
cinoma, PDGF platelet-derived growth factor, PDGFR PDGF receptor, ROCK Rho kinase, TNBC triple negative breast cancer, STAT3 signal transducer and activator of transcription 3, TGF-b
transforming growth factor-b, BCL2 B-cell lymphoma 2, CSF1R Colony Stimulating Factor 1 Receptor, CXCR2 C-X-C Motif Chemokine Receptor 2, PMN-MDSC Polymorphonuclear Myeloid-
derived suppressor cells.

Review
ll
Review
(NCI), R01CA232256 (NCI), R01CA207935 (NCI), a Bloomberg Distinguished 17. Lavie, D., Ben-Shmuel, A., Erez, N., and Scherz-Shouval, R. (2022). Can-
Professorship and the EV McCollum Endowed Chair. cer-associated fibroblasts in the single-cell era. Nat. Cancer 3, 793–807.
18. Marsh, T., Pietras, K., and McAllister, S.S. (2013). Fibroblasts as archi-
DECLARATION OF INTERESTS tects of cancer pathogenesis. Biochim. Biophys. Acta 1832, 1070–1078.
19. Sorrell, J.M., and Caplan, A.I. (2004). Fibroblast heterogeneity: more than
The authors declare no competing interest. skin deep. J. Cell Sci. 117, 667–675.
20. Plikus, M.V., Wang, X., Sinha, S., Forte, E., Thompson, S.M., Herzog,
REFERENCES E.L., Driskell, R.R., Rosenthal, N., Biernaskie, J., and Horsley, V.
(2021). Fibroblasts: origins, definitions, and functions in health and dis-
1. Biffi, G., and Tuveson, D.A. (2021). Diversity and biology of cancer-asso- ease. Cell 184, 3852–3872.
ciated fibroblasts. Physiol. Rev. 101, 147–176. 21. Shaw, T.J., and Rognoni, E. (2020). Dissecting fibroblast heterogeneity in
2. Kochetkova, M., and Samuel, M.S. (2022). Differentiation of the tumor health and fibrotic disease. Curr. Rheumatol. Rep. 22, 33.
microenvironment: are CAFs the Organizer? Trends Cell Biol. 32, 22. Lynch, M.D., and Watt, F.M. (2018). Fibroblast heterogeneity: implica-
285–294. tions for human disease. J. Clin. Invest. 128, 26–35.
3. Sahai, E., Astsaturov, I., Cukierman, E., DeNardo, D.G., Egeblad, M., 23. Kuwabara, J.T., Hara, A., Bhutada, S., Gojanovich, G.S., Chen, J., Hoku-
Evans, R.M., Fearon, D., Greten, F.R., Hingorani, S.R., Hunter, T., et al. tan, K., Shettigar, V., Lee, A.Y., DeAngelo, L.P., Heckl, J.R., et al. (2022).
(2020). A framework for advancing our understanding of cancer-associ- Consequences of PDGFRalpha(+) fibroblast reduction in adult murine
ated fibroblasts. Nat. Rev. Cancer 20, 174–186. hearts. Elife 11, e69854.
4. Alkasalias, T., Flaberg, E., Kashuba, V., Alexeyenko, A., Pavlova, T., Sav- 24. Nilsson, J., Fardoos, R., Hansen, L., Lövkvist, H., Pietras, K., Holmberg,
chenko, A., Szekely, L., Klein, G., and Guven, H. (2014). Inhibition of tu- D., and Schmidt-Christensen, A. (2020). Recruited fibroblasts reconsti-
mor cell proliferation and motility by fibroblasts is both contact and sol- tute the peri-islet membrane: a longitudinal imaging study of human islet
uble factor dependent. Proc. Natl. Acad. Sci. USA 111, 17188–17193. grafting and revascularisation. Diabetologia 63, 137–148.

5. Chang, P.H., Hwang-Verslues, W.W., Chang, Y.C., Chen, C.C., Hsiao, 25. Tsutsui, K., Machida, H., Nakagawa, A., Ahn, K., Morita, R., Sekiguchi,
M., Jeng, Y.M., Chang, K.J., Lee, E.Y.H.P., Shew, J.Y., and Lee, W.H. K., Miner, J.H., and Fujiwara, H. (2021). Mapping the molecular and
(2012). Activation of Robo1 signaling of breast cancer cells by Slit2 structural specialization of the skin basement membrane for inter-tissue
from stromal fibroblast restrains tumorigenesis via blocking PI3K/Akt/ interactions. Nat. Commun. 12, 2577.
beta-catenin pathway. Cancer Res. 72, 4652–4661. 26. Driskell, R.R., and Watt, F.M. (2015). Understanding fibroblast heteroge-
6. Djurec, M., Graña, O., Lee, A., Troulé, K., Espinet, E., Cabras, L., Navas, neity in the skin. Trends Cell Biol. 25, 92–99.
C., Blasco, M.T., Martı́n-Dı́az, L., Burdiel, M., et al. (2018). Saa3 is a key 27. Moretti, L., Stalfort, J., Barker, T.H., and Abebayehu, D. (2022). The inter-
mediator of the protumorigenic properties of cancer-associated fibro- play of fibroblasts, the extracellular matrix, and inflammation in scar for-
blasts in pancreatic tumors. Proc. Natl. Acad. Sci. USA 115, mation. J. Biol. Chem. 298, 101530.
E1147–E1156. 28. Alexander, J., and Cukierman, E. (2020). Cancer associated fibroblast:
7. Kaukonen, R., Mai, A., Georgiadou, M., Saari, M., De Franceschi, N., mediators of tumorigenesis. Matrix Biol. 91–92, 19–34.
Betz, T., Sihto, H., Ventelä, S., Elo, L., Jokitalo, E., et al. (2016). Normal 29. Birch, H.L. (2018). Extracellular matrix and ageing. Subcell. Biochem. 90,
stroma suppresses cancer cell proliferation via mechanosensitive regu- 169–190.
lation of JMJD1a-mediated transcription. Nat. Commun. 7, 12237. 30. Bonnans, C., Chou, J., and Werb, Z. (2014). Remodelling the extracellular
8. Liotta, L.A., and Kohn, E.C. (2001). The microenvironment of the tumour- matrix in development and disease. Nat. Rev. Mol. Cell Biol. 15, 786–801.
host interface. Nature 411, 375–379. 31. Frantz, C., Stewart, K.M., and Weaver, V.M. (2010). The extracellular ma-
9. Fiori, M.E., Di Franco, S., Villanova, L., Bianca, P., Stassi, G., and De Ma- trix at a glance. J. Cell Sci. 123, 4195–4200.
ria, R. (2019). Cancer-associated fibroblasts as abettors of tumor pro- 32. Glorieux, F.H. (2008). Osteogenesis imperfecta. Best Pract. Res. Clin.
gression at the crossroads of EMT and therapy resistance. Mol. Cancer Rheumatol. 22, 85–100.
18, 70.
33. Keane, M.G., and Pyeritz, R.E. (2008). Medical management of Marfan
10. Kai, F., Drain, A.P., and Weaver, V.M. (2019). The extracellular matrix syndrome. Circulation 117, 2802–2813.
modulates the metastatic journey. Dev. Cell 49, 332–346. 34. Mao, J.R., and Bristow, J. (2001). The Ehlers-Danlos syndrome: on
11. Parker, A.L., and Cox, T.R. (2020). The role of the ECM in lung cancer beyond collagens. J. Clin. Invest. 107, 1063–1069.
dormancy and outgrowth. Front. Oncol. 10, 1766. 35. Parapia, L.A., and Jackson, C. (2008). Ehlers-Danlos syndrome–a histor-
12. Calon, A., Lonardo, E., Berenguer-Llergo, A., Espinet, E., Hernando- ical review. Br. J. Haematol. 141, 32–35.
Momblona, X., Iglesias, M., Sevillano, M., Palomo-Ponce, S., Tauriello, 36. Järveläinen, H., Sainio, A., Koulu, M., Wight, T.N., and Penttinen, R.
D.V.F., Byrom, D., et al. (2015). Stromal gene expression defines poor- (2009). Extracellular matrix molecules: potential targets in pharmaco-
prognosis subtypes in colorectal cancer. Nat. Genet. 47, 320–329. therapy. Pharmacol. Rev. 61, 198–223.
13. Liu, L., Liu, L., Yao, H.H., Zhu, Z.Q., Ning, Z.L., and Huang, Q. (2016). 37. Nurden, A.T. (2011). Platelets, inflammation and tissue regeneration.
Stromal myofibroblasts are associated with poor prognosis in solid can- Thromb. Haemost. 105 (Suppl 1 ), S13–S33.
cers: a meta-analysis of published studies. PLoS One 11, e0159947.
38. Tomasek, J.J., Gabbiani, G., Hinz, B., Chaponnier, C., and Brown, R.A.
14. de Groot, A.E., Roy, S., Brown, J.S., Pienta, K.J., and Amend, S.R. (2002). Myofibroblasts and mechano-regulation of connective tissue re-
(2017). Revisiting seed and soil: examining the primary tumor and cancer modelling. Nat. Rev. Mol. Cell Biol. 3, 349–363.
cell foraging in metastasis. Mol. Cancer Res. 15, 361–370. 39. Bizik, J., Kankuri, E., Ristimäki, A., Taı̈eb, A., Vapaatalo, H., Lubitz, W.,
15. LeBleu, V.S., and Kalluri, R. (2018). A peek into cancer-associated fibro- and Vaheri, A. (2004). Cell-cell contacts trigger programmed necrosis
blasts: origins, functions and translational impact. Dis. Model. Mech. 11, and induce cyclooxygenase-2 expression. Cell Death Differ. 11,
dmm029447. 183–195.
16. Valkenburg, K.C., de Groot, A.E., and Pienta, K.J. (2018). Targeting the 40. Eming, S.A., Martin, P., and Tomic-Canic, M. (2014). Wound repair and
tumour stroma to improve cancer therapy. Nat. Rev. Clin. Oncol. 15, regeneration: mechanisms, signaling, and translation. Sci. Transl. Med.
366–381. 6, 265sr6.

Cell 186, April 13, 2023 1599


ll
Review
41. Dvorak, H.F. (1986). Tumors: wounds that do not heal. Similarities be- 60. Franco-Barraza, J., Beacham, D.A., Amatangelo, M.D., and Cukierman,
tween tumor stroma generation and wound healing. N. Engl. J. Med. E. (2016). Preparation of extracellular matrices produced by cultured and
315, 1650–1659. primary fibroblasts. Curr. Protoc. Cell Biol. 71, 10.9.1–10.9.34.
42. Brown, F.D., and Turley, S.J. (2015). Fibroblastic reticular cells: organiza- 61. Puram, S.V., Tirosh, I., Parikh, A.S., Patel, A.P., Yizhak, K., Gillespie, S.,
tion and regulation of the T lymphocyte life cycle. J. Immunol. 194, Rodman, C., Luo, C.L., Mroz, E.A., Emerick, K.S., et al. (2017). Single-cell
1389–1394. transcriptomic analysis of primary and metastatic tumor ecosystems in
43. Fletcher, A.L., Malhotra, D., and Turley, S.J. (2011). Lymph node stroma head and neck cancer. Cell 171, 1611–1624.e24.
broaden the peripheral tolerance paradigm. Trends Immunol. 32, 12–18. 62. Waise, S., Parker, R., Rose-Zerilli, M.J.J., Layfield, D.M., Wood, O., West,
44. Mescher, A.L. (2017). Macrophages and fibroblasts during inflammation J., Ottensmeier, C.H., Thomas, G.J., and Hanley, C.J. (2019). An opti-
and tissue repair in models of organ regeneration. Regeneration (Oxf) 4, mised tissue disaggregation and data processing pipeline for character-
39–53. ising fibroblast phenotypes using single-cell RNA sequencing. Sci. Rep.
9, 9580.
45. Mine, S., Fortunel, N.O., Pageon, H., and Asselineau, D. (2008). Aging al-
ters functionally human dermal papillary fibroblasts but not reticular fi- 63. Neal, J.T., Li, X., Zhu, J., Giangarra, V., Grzeskowiak, C.L., Ju, J., Liu, I.H.,
broblasts: a new view of skin morphogenesis and aging. PLoS One Chiou, S.H., Salahudeen, A.A., Smith, A.R., et al. (2018). Organoid
3, e4066. modeling of the tumor immune microenvironment. Cell 175, 1972–
1988.e16.
46. Rognoni, E., Pisco, A.O., Hiratsuka, T., Sipilä, K.H., Belmonte, J.M., Mo-
basseri, S.A., Philippeos, C., Dilão, R., and Watt, F.M. (2018). Fibroblast 64. Ootani, A., Li, X., Sangiorgi, E., Ho, Q.T., Ueno, H., Toda, S., Sugihara, H.,
state switching orchestrates dermal maturation and wound healing. Mol. Fujimoto, K., Weissman, I.L., Capecchi, M.R., and Kuo, C.J. (2009). Sus-
Syst. Biol. 14, e8174. tained in vitro intestinal epithelial culture within a Wnt-dependent stem
cell niche. Nat. Med. 15, 701–706.
47. Muhl, L., Genové, G., Leptidis, S., Liu, J., He, L., Mocci, G., Sun, Y., Gus-
tafsson, S., Buyandelger, B., Chivukula, I.V., et al. (2020). Single-cell 65. Langer, E.M., Allen-Petersen, B.L., King, S.M., Kendsersky, N.D., Turn-
analysis uncovers fibroblast heterogeneity and criteria for fibroblast idge, M.A., Kuziel, G.M., Riggers, R., Samatham, R., Amery, T.S., Jac-
and mural cell identification and discrimination. Nat. Commun. 11, 3953. ques, S.L., et al. (2019). Modeling Tumor Phenotypes In Vitro with
Three-Dimensional Bioprinting. Cell Rep. 26, 608–623.e6.
48. Rognoni, E., and Watt, F.M. (2018). Skin cell heterogeneity in develop-
ment, wound healing, and cancer. Trends Cell Biol. 28, 709–722. 66. Madden, L.R., Nguyen, T.V., Garcia-Mojica, S., Shah, V., Le, A.V., Peier,
A., Visconti, R., Parker, E.M., Presnell, S.C., Nguyen, D.G., and Retting,
49. Rinkevich, Y., Walmsley, G.G., Hu, M.S., Maan, Z.N., Newman, A.M.,
K.N. (2018). Bioprinted 3D primary human intestinal tissues model as-
Drukker, M., Januszyk, M., Krampitz, G.W., Gurtner, G.C., Lorenz,
pects of native physiology and ADME/Tox functions. iScience 2,
H.P., et al. (2015). Skin fibrosis. Identification and isolation of a dermal
156–167.
lineage with intrinsic fibrogenic potential. Science 348, aaa2151.
67. Cortes, E., Lachowski, D., Robinson, B., Sarper, M., Teppo, J.S., Thorpe,
50. Leavitt, T., Hu, M.S., Marshall, C.D., Barnes, L.A., Lorenz, H.P., and S.D., Lieberthal, T.J., Iwamoto, K., Lee, D.A., Okada-Hatakeyama, M.,
Longaker, M.T. (2016). Scarless wound healing: finding the right cells et al. (2019). Tamoxifen mechanically reprograms the tumor microenvi-
and signals. Cell Tissue Res. 365, 483–493. ronment via HIF-1A and reduces cancer cell survival. EMBO Rep. 20,
51. Knecht, A., Fine, L.G., Kleinman, K.S., Rodemann, H.P., Müller, G.A., e46557.
Woo, D.D., and Norman, J.T. (1991). Fibroblasts of rabbit kidney in cul- 68. Entenberg, D., Oktay, M.H., and Condeelis, J.S. (2023). Intravital imaging
ture. II. Paracrine stimulation of papillary fibroblasts by PDGF. Am. J. to study cancer progression and metastasis. Nat. Rev. Cancer 23, 25–42.
Physiol. 261, F292–F299.
69. Ewald, A.J., Werb, Z., and Egeblad, M. (2011). Dynamic, long-term in vivo
52. Rodemann, H.P., Müller, G.A., Knecht, A., Norman, J.T., and Fine, L.G. imaging of tumor-stroma interactions in mouse models of breast cancer
(1991). Fibroblasts of rabbit kidney in culture. I. Characterization and using spinning-disk confocal microscopy. Cold Spring Harb. Protoc.
identification of cell-specific markers. Am. J. Physiol. 261, F283–F291. 2011, pdb.top97.
53. Fane, M., and Weeraratna, A.T. (2020). How the ageing microenviron- 70. Murphy, K.J., Reed, D.A., Trpceski, M., Herrmann, D., and Timpson, P.
ment influences tumour progression. Nat. Rev. Cancer 20, 89–106. (2021). Quantifying and visualising the nuances of cellular dynamics
54. Ligorio, M., Sil, S., Malagon-Lopez, J., Nieman, L.T., Misale, S., Di Pilato, in vivo using intravital imaging. Curr. Opin. Cell Biol. 72, 41–53.
M., Ebright, R.Y., Karabacak, M.N., Kulkarni, A.S., Liu, A., et al. (2019). 71. Vennin, C., Mélénec, P., Rouet, R., Nobis, M., Cazet, A.S., Murphy, K.J.,
Stromal microenvironment shapes the intratumoral architecture of Herrmann, D., Reed, D.A., Lucas, M.C., Warren, S.C., et al. (2019). CAF
pancreatic cancer. Cell 178, 160–175.e27. hierarchy driven by pancreatic cancer cell p53-status creates a pro-met-
55. Monteran, L., and Erez, N. (2019). The dark side of fibroblasts: cancer- astatic and chemoresistant environment via perlecan. Nat. Commun.
associated fibroblasts as mediators of immunosuppression in the tumor 10, 3637.
microenvironment. Front. Immunol. 10, 1835. 72. van den Brink, S.C., Sage, F., Vértesy, Á., Spanjaard, B., Peterson-Ma-
56. Sugimoto, H., Mundel, T.M., Kieran, M.W., and Kalluri, R. (2006). Identi- duro, J., Baron, C.S., Robin, C., and van Oudenaarden, A. (2017). Sin-
fication of fibroblast heterogeneity in the tumor microenvironment. Can- gle-cell sequencing reveals dissociation-induced gene expression in tis-
cer Biol. Ther. 5, 1640–1646. sue subpopulations. Nat. Methods 14, 935–936.
57. Zabransky, D.J., Jaffee, E.M., and Weeraratna, A.T. (2022). Shared ge- 73. Jain, S., Rick, J.W., Joshi, R.S., Beniwal, A., Spatz, J., Gill, S., Chang,
netic and epigenetic changes link aging and cancer. Trends Cell Biol. A.C.C., Choudhary, N., Nguyen, A.T., Sudhir, S., et al. (2023). Single-
32, 338–350. cell RNA sequencing and spatial transcriptomics reveal cancer-associ-
58. Costa, A., Kieffer, Y., Scholer-Dahirel, A., Pelon, F., Bourachot, B., Car- ated fibroblasts in glioblastoma with protumoral effects. J. Clin. Invest.
don, M., Sirven, P., Magagna, I., Fuhrmann, L., Bernard, C., et al. 133, e147087.
(2018). Fibroblast heterogeneity and immunosuppressive environment 74. Peng, Z., Ye, M., Ding, H., Feng, Z., and Hu, K. (2022). Spatial transcrip-
in human breast cancer. Cancer Cell 33, 463–479.e10. tomics atlas reveals the crosstalk between cancer-associated fibroblasts
59. Jackson, H.W., Fischer, J.R., Zanotelli, V.R.T., Ali, H.R., Mechera, R., and tumor microenvironment components in colorectal cancer. J. Transl.
Soysal, S.D., Moch, H., Muenst, S., Varga, Z., Weber, W.P., and Boden- Med. 20, 302.
miller, B. (2020). The single-cell pathology landscape of breast cancer. 75. Buechler, M.B., Pradhan, R.N., Krishnamurty, A.T., Cox, C., Calviello,
Nature 578, 615–620. A.K., Wang, A.W., Yang, Y.A., Tam, L., Caothien, R., Roose-Girma, M.,

1600 Cell 186, April 13, 2023


ll
Review
et al. (2021). Cross-tissue organization of the fibroblast lineage. Nature 91. Biffi, G., Oni, T.E., Spielman, B., Hao, Y., Elyada, E., Park, Y., Preall, J.,
593, 575–579. and Tuveson, D.A. (2019). IL1-induced JAK/STAT signaling is antago-
76. Sanford-Crane, H., Abrego, J., and Sherman, M.H. (2019). Fibroblasts as nized by tgfbeta to shape CAF heterogeneity in pancreatic ductal adeno-
Modulators of Local and Systemic Cancer Metabolism. Cancers 11, 619. carcinoma. Cancer Discov. 9, 282–301.
92. Calvo, F., Ege, N., Grande-Garcia, A., Hooper, S., Jenkins, R.P.,
77. Rinn, J.L., Bondre, C., Gladstone, H.B., Brown, P.O., and Chang, H.Y.
Chaudhry, S.I., Harrington, K., Williamson, P., Moeendarbary, E., Char-
(2006). Anatomic demarcation by positional variation in fibroblast gene
ras, G., and Sahai, E. (2013). Mechanotransduction and YAP-dependent
expression programs. PLoS Genet. 2, e119.
matrix remodelling is required for the generation and maintenance of
78. Chang, H.Y., Chi, J.T., Dudoit, S., Bondre, C., van de Rijn, M., Botstein, cancer-associated fibroblasts. Nat. Cell Biol. 15, 637–646.
D., and Brown, P.O. (2002). Diversity, topographic differentiation, and
93. Scherz-Shouval, R., Santagata, S., Mendillo, M.L., Sholl, L.M., Ben-
positional memory in human fibroblasts. Proc. Natl. Acad. Sci. USA 99,
Aharon, I., Beck, A.H., Dias-Santagata, D., Koeva, M., Stemmer, S.M.,
12877–12882.
Whitesell, L., and Lindquist, S. (2014). The reprogramming of tumor
79. Öhlund, D., Handly-Santana, A., Biffi, G., Elyada, E., Almeida, A.S., Ponz- stroma by HSF1 is a potent enabler of malignancy. Cell 158, 564–578.
Sarvise, M., Corbo, V., Oni, T.E., Hearn, S.A., Lee, E.J., et al. (2017).
94. Shani, O., Raz, Y., Monteran, L., Scharff, Y., Levi-Galibov, O., Megides,
Distinct populations of inflammatory fibroblasts and myofibroblasts in
O., Shacham, H., Cohen, N., Silverbush, D., Avivi, C., et al. (2021). Evo-
pancreatic cancer. J. Exp. Med. 214, 579–596.
lution of fibroblasts in the lung metastatic microenvironment is driven
80. Anderberg, C., Li, H., Fredriksson, L., Andrae, J., Betsholtz, C., Li, X., by stage-specific transcriptional plasticity. Elife 10, e60745.
Eriksson, U., and Pietras, K. (2009). Paracrine signaling by platelet-
95. Ao, Z., Shah, S.H., Machlin, L.M., Parajuli, R., Miller, P.C., Rawal, S., Wil-
derived growth factor-CC promotes tumor growth by recruitment of can-
liams, A.J., Cote, R.J., Lippman, M.E., Datar, R.H., and El-Ashry, D.
cer-associated fibroblasts. Cancer Res. 69, 369–378.
(2015). Identification of cancer-associated fibroblasts in circulating blood
81. Bartoschek, M., Oskolkov, N., Bocci, M., Lövrot, J., Larsson, C., Som- from patients with metastatic breast cancer. Cancer Res. 75, 4681–4687.
marin, M., Madsen, C.D., Lindgren, D., Pekar, G., Karlsson, G., et al.
96. Hill, R., Song, Y., Cardiff, R.D., and Van Dyke, T. (2005). Selective evolu-
(2018). Spatially and functionally distinct subclasses of breast cancer-
tion of stromal mesenchyme with p53 loss in response to epithelial
associated fibroblasts revealed by single cell RNA sequencing. Nat.
tumorigenesis. Cell 123, 1001–1011.
Commun. 9, 5150.
97. Kurose, K., Gilley, K., Matsumoto, S., Watson, P.H., Zhou, X.P., and Eng,
82. Bernard, V., Semaan, A., Huang, J., San Lucas, F.A., Mulu, F.C., Ste-
C. (2002). Frequent somatic mutations in PTEN and TP53 are mutually
phens, B.M., Guerrero, P.A., Huang, Y., Zhao, J., Kamyabi, N., et al.
exclusive in the stroma of breast carcinomas. Nat. Genet. 32, 355–357.
(2019). Single-cell transcriptomics of pancreatic cancer precursors dem-
onstrates epithelial and microenvironmental heterogeneity as an early 98. Patocs, A., Zhang, L., Xu, Y., Weber, F., Caldes, T., Mutter, G.L., Platzer,
event in neoplastic progression. Clin. Cancer Res. 25, 2194–2205. P., and Eng, C. (2007). Breast-cancer stromal cells with TP53 mutations
and nodal metastases. N. Engl. J. Med. 357, 2543–2551.
83. Li, H., Courtois, E.T., Sengupta, D., Tan, Y., Chen, K.H., Goh, J.J.L.,
Kong, S.L., Chua, C., Hon, L.K., Tan, W.S., et al. (2017). Reference 99. Campbell, I., Polyak, K., and Haviv, I. (2009). Clonal mutations in the can-
component analysis of single-cell transcriptomes elucidates cellular het- cer-associated fibroblasts: the case against genetic coevolution. Cancer
erogeneity in human colorectal tumors. Nat. Genet. 49, 708–718. Res. 69, 6765–6768.

84. Micke, P., and Ostman, A. (2004). Tumour-stroma interaction: cancer- 100. Hosein, A.N., Huang, H., Wang, Z., Parmar, K., Du, W., Huang, J., Maitra,
associated fibroblasts as novel targets in anti-cancer therapy? Lung A., Olson, E., Verma, U., and Brekken, R.A. (2019). Cellular heterogeneity
Cancer 45 (Suppl 2 ), S163–S175. during mouse pancreatic ductal adenocarcinoma progression at single-
cell resolution. JCI Insight 5, e129212.
85. Paulsson, J., Sjöblom, T., Micke, P., Pontén, F., Landberg, G., Heldin,
101. Qiu, W., Hu, M., Sridhar, A., Opeskin, K., Fox, S., Shipitsin, M., Trivett, M.,
C.H., Bergh, J., Brennan, D.J., Jirström, K., and Ostman, A. (2009). Prog-
Thompson, E.R., Ramakrishna, M., Gorringe, K.L., et al. (2008). No evi-
nostic significance of stromal platelet-derived growth factor beta-recep-
dence of clonal somatic genetic alterations in cancer-associated fibro-
tor expression in human breast cancer. Am. J. Pathol. 175, 334–341.
blasts from human breast and ovarian carcinomas. Nat. Genet. 40,
86. McAndrews, K.M., Chen, Y., Darpolor, J.K., Zheng, X., Yang, S., Cars- 650–655.
tens, J.L., Li, B., Wang, H., Miyake, T., Correa de Sampaio, P., et al.
102. Walter, K., Omura, N., Hong, S.M., Griffith, M., and Goggins, M. (2008).
(2022). Identification of functional heterogeneity of carcinoma-associ-
Pancreatic cancer associated fibroblasts display normal allelotypes.
ated fibroblasts with distinct IL6-mediated therapy resistance in pancre-
Cancer Biol. Ther. 7, 882–888.
atic cancer. Cancer Discov. 12, 1580–1597.
103. Arina, A., Idel, C., Hyjek, E.M., Alegre, M.L., Wang, Y., Bindokas, V.P.,
87. Breiteneder-Geleff, S., Soleiman, A., Kowalski, H., Horvat, R., Amann, G.,
Weichselbaum, R.R., and Schreiber, H. (2016). Tumor-associated fibro-
Kriehuber, E., Diem, K., Weninger, W., Tschachler, E., Alitalo, K., and Ker-
blasts predominantly come from local and not circulating precursors.
jaschki, D. (1999). Angiosarcomas express mixed endothelial pheno-
Proc. Natl. Acad. Sci. USA 113, 7551–7556.
types of blood and lymphatic capillaries: podoplanin as a specific marker
for lymphatic endothelium. Am. J. Pathol. 154, 385–394. 104. Paulsson, J., and Micke, P. (2014). Prognostic relevance of cancer-asso-
ciated fibroblasts in human cancer. Semin. Cancer Biol. 25, 61–68.
88. Arnold, J.N., Magiera, L., Kraman, M., and Fearon, D.T. (2014). Tumoral
immune suppression by macrophages expressing fibroblast activation 105. Beacham, D.A., and Cukierman, E. (2005). Stromagenesis: the changing
protein-alpha and heme oxygenase-1. Cancer Immunol. Res. 2, face of fibroblastic microenvironments during tumor progression. Semin.
121–126. Cancer Biol. 15, 329–341.

89. Rockey, D.C., Weymouth, N., and Shi, Z. (2013). Smooth muscle alpha 106. Ruiter, D., Bogenrieder, T., Elder, D., and Herlyn, M. (2002). Melanoma-
actin (Acta2) and myofibroblast function during hepatic wound healing. stroma interactions: structural and functional aspects. Lancet Oncol. 3,
PLoS One 8, e77166. 35–43.

90. Rønnov-Jessen, L., Petersen, O.W., Koteliansky, V.E., and Bissell, M.J. 107. Elenbaas, B., and Weinberg, R.A. (2001). Heterotypic signaling between
(1995). The origin of the myofibroblasts in breast cancer. Recapitulation epithelial tumor cells and fibroblasts in carcinoma formation. Exp. Cell
of tumor environment in culture unravels diversity and implicates con- Res. 264, 169–184.
verted fibroblasts and recruited smooth muscle cells. J. Clin. Invest. 108. Erez, N., Truitt, M., Olson, P., Arron, S.T., and Hanahan, D. (2010). Can-
95, 859–873. cer-associated fibroblasts are activated in incipient neoplasia to

Cell 186, April 13, 2023 1601


ll
Review
orchestrate tumor-promoting inflammation in an NF-kappaB-dependent cer cell proliferation and progression to an invasive phenotype. Stem
manner. Cancer Cell 17, 135–147. Cells Int. 2017, 9216502.
109. Löhr, M., Schmidt, C., Ringel, J., Kluth, M., Müller, P., Nizze, H., and Jes- 125. Sappino, A.P., Skalli, O., Jackson, B., Schürch, W., and Gabbiani, G.
nowski, R. (2001). Transforming growth factor-beta1 induces desmopla- (1988). Smooth-muscle differentiation in stromal cells of malignant and
sia in an experimental model of human pancreatic carcinoma. Cancer non-malignant breast tissues. Int. J. Cancer 41, 707–712.
Res. 61, 550–555.
126. Tsukamoto, H., Mishima, Y., Hayashibe, K., and Sasase, A. (1992).
110. Straub, J.M., New, J., Hamilton, C.D., Lominska, C., Shnayder, Y., and Alpha-smooth muscle actin expression in tumor and stromal cells of
Thomas, S.M. (2015). Radiation-induced fibrosis: mechanisms and impli- benign and malignant human pigment cell tumors. J. Invest. Dermatol.
cations for therapy. J. Cancer Res. Clin. Oncol. 141, 1985–1994. 98, 116–120.
111. Sun, Y., Campisi, J., Higano, C., Beer, T.M., Porter, P., Coleman, I., True, 127. Hinz, B., Mastrangelo, D., Iselin, C.E., Chaponnier, C., and Gabbiani, G.
L., and Nelson, P.S. (2012). Treatment-induced damage to the tumor (2001). Mechanical tension controls granulation tissue contractile activity
microenvironment promotes prostate cancer therapy resistance through and myofibroblast differentiation. Am. J. Pathol. 159, 1009–1020.
WNT16B. Nat. Med. 18, 1359–1368.
128. Kapanci, Y., Ribaux, C., Chaponnier, C., and Gabbiani, G. (1992). Cyto-
112. Ke, Y., and Wang, X.J. (2021). TGFbeta Signaling in Photoaging and UV- skeletal features of alveolar myofibroblasts and pericytes in normal hu-
Induced Skin Cancer. J. Invest. Dermatol. 141, 1104–1110. man and rat lung. J. Histochem. Cytochem. 40, 1955–1963.
113. Martinez-Outschoorn, U.E., Balliet, R.M., Rivadeneira, D.B., Chiavarina,
129. Chien, Y., Scuoppo, C., Wang, X., Fang, X., Balgley, B., Bolden, J.E., Pre-
B., Pavlides, S., Wang, C., Whitaker-Menezes, D., Daumer, K.M., Lin,
msrirut, P., Luo, W., Chicas, A., Lee, C.S., et al. (2011). Control of the
Z., Witkiewicz, A.K., et al. (2010). Oxidative stress in cancer associated
senescence-associated secretory phenotype by NF-kappaB promotes
fibroblasts drives tumor-stroma co-evolution: A new paradigm for under-
senescence and enhances chemosensitivity. Genes Dev. 25, 2125–2136.
standing tumor metabolism, the field effect and genomic instability in
cancer cells. Cell Cycle 9, 3256–3276. 130. Hoare, M., Ito, Y., Kang, T.W., Weekes, M.P., Matheson, N.J., Patten,
D.A., Shetty, S., Parry, A.J., Menon, S., Salama, R., et al. (2016). NOTCH1
114. Toullec, A., Gerald, D., Despouy, G., Bourachot, B., Cardon, M., Lefort,
mediates a switch between two distinct secretomes during senescence.
S., Richardson, M., Rigaill, G., Parrini, M.C., Lucchesi, C., et al. (2010).
Nat. Cell Biol. 18, 979–992.
Oxidative stress promotes myofibroblast differentiation and tumour
spreading. EMBO Mol. Med. 2, 211–230. 131. Kuilman, T., Michaloglou, C., Vredeveld, L.C.W., Douma, S., van Doorn,
R., Desmet, C.J., Aarden, L.A., Mooi, W.J., and Peeper, D.S. (2008).
115. Direkze, N.C., Forbes, S.J., Brittan, M., Hunt, T., Jeffery, R., Preston, S.L.,
Oncogene-induced senescence relayed by an interleukin-dependent in-
Poulsom, R., Hodivala-Dilke, K., Alison, M.R., and Wright, N.A. (2003).
flammatory network. Cell 133, 1019–1031.
Multiple organ engraftment by bone-marrow-derived myofibroblasts
and fibroblasts in bone-marrow-transplanted mice. Stem Cell. 21, 132. Bhattacharjee, S., Hamberger, F., Ravichandra, A., Miller, M., Nair, A.,
514–520. Affo, S., Filliol, A., Chin, L., Savage, T.M., Yin, D., et al. (2021). Tumor re-
116. Gao, Z., Daquinag, A.C., Su, F., Snyder, B., and Kolonin, M.G. (2018). striction by type I collagen opposes tumor-promoting effects of cancer-
PDGFRalpha/PDGFRbeta signaling balance modulates progenitor cell associated fibroblasts. J. Clin. Invest. 131, e146987.
differentiation into white and beige adipocytes. Development 145, 133. Dominguez, C.X., Müller, S., Keerthivasan, S., Koeppen, H., Hung, J.,
dev155861. Gierke, S., Breart, B., Foreman, O., Bainbridge, T.W., Castiglioni, A.,
117. Hosaka, K., Yang, Y., Seki, T., Fischer, C., Dubey, O., Fredlund, E., Hart- et al. (2020). Single-cell RNA sequencing reveals stromal evolution into
man, J., Religa, P., Morikawa, H., Ishii, Y., et al. (2016). Pericyte-fibroblast LRRC15(+) myofibroblasts as a determinant of patient response to can-
transition promotes tumor growth and metastasis. Proc. Natl. Acad. Sci. cer immunotherapy. Cancer Discov. 10, 232–253.
USA 113, E5618–E5627. 134. Hornburg, M., Desbois, M., Lu, S., Guan, Y., Lo, A.A., Kaufman, S., Elrod,
118. Iwano, M., Plieth, D., Danoff, T.M., Xue, C., Okada, H., and Neilson, E.G. A., Lotstein, A., DesRochers, T.M., Munoz-Rodriguez, J.L., et al. (2021).
(2002). Evidence that fibroblasts derive from epithelium during tissue Single-cell dissection of cellular components and interactions shaping
fibrosis. J. Clin. Invest. 110, 341–350. the tumor immune phenotypes in ovarian cancer. Cancer Cell 39,
928–944.e6.
119. Jung, Y., Kim, J.K., Shiozawa, Y., Wang, J., Mishra, A., Joseph, J., Berry,
J.E., McGee, S., Lee, E., Sun, H., et al. (2013). Recruitment of mesen- 135. Kieffer, Y., Hocine, H.R., Gentric, G., Pelon, F., Bernard, C., Bourachot,
chymal stem cells into prostate tumours promotes metastasis. Nat. Com- B., Lameiras, S., Albergante, L., Bonneau, C., Guyard, A., et al. (2020).
mun. 4, 1795. Single-cell analysis reveals fibroblast clusters linked to immunotherapy
120. Karnoub, A.E., Dash, A.B., Vo, A.P., Sullivan, A., Brooks, M.W., Bell, resistance in cancer. Cancer Discov. 10, 1330–1351.
G.W., Richardson, A.L., Polyak, K., Tubo, R., and Weinberg, R.A. 136. Wu, S.Z., Roden, D.L., Wang, C., Holliday, H., Harvey, K., Cazet, A.S.,
(2007). Mesenchymal stem cells within tumour stroma promote breast Murphy, K.J., Pereira, B., Al-Eryani, G., Bartonicek, N., et al. (2020). Stro-
cancer metastasis. Nature 449, 557–563. mal cell diversity associated with immune evasion in human triple-nega-
121. Kidd, S., Spaeth, E., Watson, K., Burks, J., Lu, H., Klopp, A., Andreeff, M., tive breast cancer. EMBO J. 39, e104063.
and Marini, F.C. (2012). Origins of the tumor microenvironment: quantita- 137. Elyada, E., Bolisetty, M., Laise, P., Flynn, W.F., Courtois, E.T., Burkhart,
tive assessment of adipose-derived and bone marrow-derived stroma. R.A., Teinor, J.A., Belleau, P., Biffi, G., Lucito, M.S., et al. (2019). Cross-
PLoS One 7, e30563. species single-cell analysis of pancreatic ductal adenocarcinoma reveals
122. Luo, H., Xia, X., Huang, L.B., An, H., Cao, M., Kim, G.D., Chen, H.N., antigen-presenting cancer-associated fibroblasts. Cancer Discov. 9,
Zhang, W.H., Shu, Y., Kong, X., et al. (2022). Pan-cancer single-cell anal- 1102–1123.
ysis reveals the heterogeneity and plasticity of cancer-associated fibro- 138. Ferrer, I.R., Wagener, M.E., Song, M., Kirk, A.D., Larsen, C.P., and Ford,
blasts in the tumor microenvironment. Nat. Commun. 13, 6619. M.L. (2011). Antigen-specific induced Foxp3+ regulatory T cells are
123. Ogawa, M., LaRue, A.C., and Drake, C.J. (2006). Hematopoietic origin of generated following CD40/CD154 blockade. Proc. Natl. Acad. Sci. USA
fibroblasts/myofibroblasts: Its pathophysiologic implications. Blood 108, 108, 20701–20706.
2893–2896. 139. Mikami, N., Kawakami, R., Chen, K.Y., Sugimoto, A., Ohkura, N., and Sa-
124. Strong, A.L., Pei, D.T., Hurst, C.G., Gimble, J.M., Burow, M.E., and Bun- kaguchi, S. (2020). Epigenetic conversion of conventional T cells into reg-
nell, B.A. (2017). Obesity enhances the conversion of adipose-derived ulatory T cells by CD28 signal deprivation. Proc. Natl. Acad. Sci. USA
stromal/stem cells into carcinoma-associated fibroblast leading to can- 117, 12258–12268.

1602 Cell 186, April 13, 2023


ll
Review
140. Huang, H., Wang, Z., Zhang, Y., Pradhan, R.N., Ganguly, D., Chandra, R., cancer-associated fibroblast-driven nutritional support and immunosup-
Murimwa, G., Wright, S., Gu, X., Maddipati, R., et al. (2022). Mesothelial pression. Cancer Discov. 11, 446–479.
cell-derived antigen-presenting cancer-associated fibroblasts induce 156. Mizutani, Y., Kobayashi, H., Iida, T., Asai, N., Masamune, A., Hara, A.,
expansion of regulatory T cells in pancreatic cancer. Cancer Cell 40, Esaki, N., Ushida, K., Mii, S., Shiraki, Y., et al. (2019). Meflin-positive can-
656–673.e7. cer-associated fibroblasts inhibit pancreatic carcinogenesis. Cancer
141. Sebastian, A., Hum, N.R., Martin, K.A., Gilmore, S.F., Peran, I., Byers, Res. 79, 5367–5381.
S.W., Wheeler, E.K., Coleman, M.A., and Loots, G.G. (2020). Single-
157. Pelon, F., Bourachot, B., Kieffer, Y., Magagna, I., Mermet-Meillon, F.,
cell transcriptomic analysis of tumor-derived fibroblasts and normal tis-
Bonnet, I., Costa, A., Givel, A.M., Attieh, Y., Barbazan, J., et al. (2020).
sue-resident fibroblasts reveals fibroblast heterogeneity in breast can-
Cancer-associated fibroblast heterogeneity in axillary lymph nodes
cer. Cancers 12, 1307.
drives metastases in breast cancer through complementary mecha-
142. Wang, Y., Liang, Y., Xu, H., Zhang, X., Mao, T., Cui, J., Yao, J., Wang, Y., nisms. Nat. Commun. 11, 404.
Jiao, F., Xiao, X., et al. (2021). Single-cell analysis of pancreatic ductal
158. Su, S., Chen, J., Yao, H., Liu, J., Yu, S., Lao, L., Wang, M., Luo, M., Xing,
adenocarcinoma identifies a novel fibroblast subtype associated with
Y., Chen, F., et al. (2018). CD10(+)GPR77(+) cancer-associated fibro-
poor prognosis but better immunotherapy response. Cell Discov. 7, 36.
blasts promote cancer formation and chemoresistance by sustaining
143. Lambrechts, D., Wauters, E., Boeckx, B., Aibar, S., Nittner, D., Burton, cancer stemness. Cell 172, 841–856.e16.
O., Bassez, A., Decaluwé, H., Pircher, A., Van den Eynde, K., et al.
(2018). Phenotype molding of stromal cells in the lung tumor microenvi- 159. Costea, D.E., Hills, A., Osman, A.H., Thurlow, J., Kalna, G., Huang, X.,
ronment. Nat. Med. 24, 1277–1289. Pena Murillo, C., Parajuli, H., Suliman, S., Kulasekara, K.K., et al.
(2013). Identification of two distinct carcinoma-associated fibroblast
144. Kerdidani, D., Aerakis, E., Verrou, K.M., Angelidis, I., Douka, K., Maniou, subtypes with differential tumor-promoting abilities in oral squamous
M.A., Stamoulis, P., Goudevenou, K., Prados, A., Tzaferis, C., et al. cell carcinoma. Cancer Res. 73, 3888–3901.
(2022). Lung tumor MHCII immunity depends on in situ antigen presenta-
tion by fibroblasts. J. Exp. Med. 219, e20210815. 160. Hussain, A., Voisin, V., Poon, S., Karamboulas, C., Bui, N.H.B., Meens, J.,
Dmytryshyn, J., Ho, V.W., Tang, K.H., Paterson, J., et al. (2020). Distinct
145. Davidson, S., Efremova, M., Riedel, A., Mahata, B., Pramanik, J., Huuh-
fibroblast functional states drive clinical outcomes in ovarian cancer and
tanen, J., Kar, G., Vento-Tormo, R., Hagai, T., Chen, X., et al. (2020). Sin-
are regulated by TCF21. J. Exp. Med. 217, e20191094.
gle-cell RNA sequencing reveals a dynamic stromal niche that supports
tumor growth. Cell Rep. 31, 107628. 161. Izar, B., Tirosh, I., Stover, E.H., Wakiro, I., Cuoco, M.S., Alter, I., Rodman,
C., Leeson, R., Su, M.J., Shah, P., et al. (2020). A single-cell landscape of
146. Givel, A.M., Kieffer, Y., Scholer-Dahirel, A., Sirven, P., Cardon, M., Pelon,
high-grade serous ovarian cancer. Nat. Med. 26, 1271–1279.
F., Magagna, I., Gentric, G., Costa, A., Bonneau, C., et al. (2018). miR200-
regulated CXCL12beta promotes fibroblast heterogeneity and immuno- 162. Grout, J.A., Sirven, P., Leader, A.M., Maskey, S., Hector, E., Puisieux, I.,
suppression in ovarian cancers. Nat. Commun. 9, 1056. Steffan, F., Cheng, E., Tung, N., Maurin, M., et al. (2022). Spatial posi-
147. Neuzillet, C., Tijeras-Raballand, A., Ragulan, C., Cros, J., Patil, Y., tioning and matrix programs of cancer-associated fibroblasts promote
Martinet, M., Erkan, M., Kleeff, J., Wilson, J., Apte, M., et al. (2019). Inter- T-cell exclusion in human lung tumors. Cancer Discov. 12, 2606–2625.
and intra-tumoural heterogeneity in cancer-associated fibroblasts of hu- 163. Hao, J., Zeltz, C., Pintilie, M., Li, Q., Sakashita, S., Wang, T., Cabanero,
man pancreatic ductal adenocarcinoma. J. Pathol. 248, 51–65. M., Martins-Filho, S.N., Wang, D.Y., Pasko, E., et al. (2019). Characteriza-
148. Fearon, D.T. (2014). The carcinoma-associated fibroblast expressing tion of distinct populations of carcinoma-associated fibroblasts from
fibroblast activation protein and escape from immune surveillance. Can- non-small cell lung carcinoma reveals a role for ST8SIA2 in cancer cell in-
cer Immunol. Res. 2, 187–193. vasion. Neoplasia 21, 482–493.

149. Kawase, T., Yasui, Y., Nishina, S., Hara, Y., Yanatori, I., Tomiyama, Y., 164. Pellinen, T., Paavolainen, L., Martı́n-Bernabé, A., Papatella Araujo, R.,
Nakashima, Y., Yoshida, K., Kishi, F., Nakamura, M., and Hino, K. Strell, C., Mezheyeuski, A., Backman, M., La Fleur, L., Brück, O., Sjölund,
(2015). Fibroblast activation protein-alpha-expressing fibroblasts pro- J., et al. (2023). Fibroblast subsets in non-small cell lung cancer: associ-
mote the progression of pancreatic ductal adenocarcinoma. BMC Gas- ations with survival, mutations, and immune features. J. Natl. Cancer Inst.
troenterol. 15, 109. 115, 71–82.
150. Looi, C.K., Chung, F.F.L., Leong, C.O., Wong, S.F., Rosli, R., and Mai, 165. Hu, H., Piotrowska, Z., Hare, P.J., Chen, H., Mulvey, H.E., Mayfield, A.,
C.W. (2019). Therapeutic challenges and current immunomodulatory Noeen, S., Kattermann, K., Greenberg, M., Williams, A., et al. (2021).
strategies in targeting the immunosuppressive pancreatic tumor micro- Three subtypes of lung cancer fibroblasts define distinct therapeutic par-
environment. J. Exp. Clin. Cancer Res. 38, 162. adigms. Cancer Cell 39, 1531–1547.e10.
151. Miyai, Y., Esaki, N., Takahashi, M., and Enomoto, A. (2020). Cancer- 166. Zhang, M., Yang, H., Wan, L., Wang, Z., Wang, H., Ge, C., Liu, Y., Hao, Y.,
associated fibroblasts that restrain cancer progression: hypotheses Zhang, D., Shi, G., et al. (2020b). Single-cell transcriptomic architecture
and perspectives. Cancer Sci. 111, 1047–1057. and intercellular crosstalk of human intrahepatic cholangiocarcinoma.
152. Awaji, M., Saxena, S., Wu, L., Prajapati, D.R., Purohit, A., Varney, M.L., J. Hepatol. 73, 1118–1130.
Kumar, S., Rachagani, S., Ly, Q.P., Jain, M., et al. (2020). CXCR2 167. Gerling, M., Büller, N.V.J.A., Kirn, L.M., Joost, S., Frings, O., Englert, B.,
signaling promotes secretory cancer-associated fibroblasts in pancre- Bergström, Å., Kuiper, R.V., Blaas, L., Wielenga, M.C.B., et al. (2016).
atic ductal adenocarcinoma. FASEB J 34, 9405–9418. Stromal Hedgehog signalling is downregulated in colon cancer and its
153. Helms, E., Onate, M.K., and Sherman, M.H. (2020). Fibroblast heteroge- restoration restrains tumour growth. Nat. Commun. 7, 12321.
neity in the pancreatic tumor microenvironment. Cancer Discov. 10, 168. Lee, J.J., Perera, R.M., Wang, H., Wu, D.C., Liu, X.S., Han, S., Fitamant,
648–656. J., Jones, P.D., Ghanta, K.S., Kawano, S., et al. (2014). Stromal response
154. Feldmann, K., Maurer, C., Peschke, K., Teller, S., Schuck, K., Steiger, K., to Hedgehog signaling restrains pancreatic cancer progression. Proc.
Engleitner, T., Öllinger, R., Nomura, A., Wirges, N., et al. (2021). Mesen- Natl. Acad. Sci. USA 111, E3091–E3100.
chymal plasticity regulated by Prrx1 drives aggressive pancreatic cancer 169. Özdemir, B.C., Pentcheva-Hoang, T., Carstens, J.L., Zheng, X., Wu,
biology. Gastroenterology 160, 346–361.e24. C.C., Simpson, T.R., Laklai, H., Sugimoto, H., Kahlert, C., Novitskiy,
155. Francescone, R., Barbosa Vendramini-Costa, D., Franco-Barraza, J., S.V., et al. (2014). Depletion of carcinoma-associated fibroblasts and
Wagner, J., Muir, A., Lau, A.N., Gabitova, L., Pazina, T., Gupta, S., Luong, fibrosis induces immunosuppression and accelerates pancreas cancer
T., et al. (2021). Netrin G1 promotes pancreatic tumorigenesis through with reduced survival. Cancer Cell 25, 719–734.

Cell 186, April 13, 2023 1603


ll
Review
170. Pallangyo, C.K., Ziegler, P.K., and Greten, F.R. (2015). IKKbeta acts as a tance in mice and is associated with patient survival. Gastroenterology
tumor suppressor in cancer-associated fibroblasts during intestinal 151, 180–193.e12.
tumorigenesis. J. Exp. Med. 212, 2253–2266. 186. Jue, S.F., Bradley, R.S., Rudnicki, J.A., Varmus, H.E., and Brown, A.M.
171. Rhim, A.D., Oberstein, P.E., Thomas, D.H., Mirek, E.T., Palermo, C.F., (1992). The mouse Wnt-1 gene can act via a paracrine mechanism in
Sastra, S.A., Dekleva, E.N., Saunders, T., Becerra, C.P., Tattersall, transformation of mammary epithelial cells. Mol. Cell Biol. 12, 321–328.
I.W., et al. (2014). Stromal elements act to restrain, rather than support, 187. Trimboli, A.J., Cantemir-Stone, C.Z., Li, F., Wallace, J.A., Merchant, A.,
pancreatic ductal adenocarcinoma. Cancer Cell 25, 735–747. Creasap, N., Thompson, J.C., Caserta, E., Wang, H., Chong, J.L., et al.
172. Shin, K., Lim, A., Zhao, C., Sahoo, D., Pan, Y., Spiekerkoetter, E., Liao, (2009). Pten in stromal fibroblasts suppresses mammary epithelial tu-
J.C., and Beachy, P.A. (2014). Hedgehog signaling restrains bladder can- mours. Nature 461, 1084–1091.
cer progression by eliciting stromal production of urothelial differentiation 188. Bhowmick, N.A., Chytil, A., Plieth, D., Gorska, A.E., Dumont, N., Shap-
factors. Cancer Cell 26, 521–533. pell, S., Washington, M.K., Neilson, E.G., and Moses, H.L. (2004). TGF-
173. Galbo, P.M., Jr., Zang, X., and Zheng, D. (2021). Molecular features of beta signaling in fibroblasts modulates the oncogenic potential of adja-
cancer-associated fibroblast subtypes and their implication on cancer cent epithelia. Science 303, 848–851.
pathogenesis, prognosis, and immunotherapy resistance. Clin. Cancer 189. Grum-Schwensen, B., Klingelhofer, J., Berg, C.H., El-Naaman, C., Gri-
Res. 27, 2636–2647. gorian, M., Lukanidin, E., and Ambartsumian, N. (2005). Suppression of
174. Foster, D.S., Januszyk, M., Delitto, D., Yost, K.E., Griffin, M., Guo, J., tumor development and metastasis formation in mice lacking the
Guardino, N., Delitto, A.E., Chinta, M., Burcham, A.R., et al. (2022). Mul- S100A4(mts1) gene. Cancer Res. 65, 3772–3780.
tiomic analysis reveals conservation of cancer-associated fibroblast 190. Elkabets, M., Gifford, A.M., Scheel, C., Nilsson, B., Reinhardt, F., Bray,
phenotypes across species and tissue of origin. Cancer Cell 40, 1392– M.A., Carpenter, A.E., Jirström, K., Magnusson, K., Ebert, B.L., et al.
1406.e7. (2011). Human tumors instigate granulin-expressing hematopoietic cells
175. Esbona, K., Inman, D., Saha, S., Jeffery, J., Schedin, P., Wilke, L., and that promote malignancy by activating stromal fibroblasts in mice. J. Clin.
Keely, P. (2016). COX-2 modulates mammary tumor progression in Invest. 121, 784–799.
response to collagen density. Breast Cancer Res. 18, 35. 191. Kuznetsov, H.S., Marsh, T., Markens, B.A., Castaño, Z., Greene-Colozzi,
176. Guo, Q., Minnier, J., Burchard, J., Chiotti, K., Spellman, P., and Schedin, A., Hay, S.A., Brown, V.E., Richardson, A.L., Signoretti, S., Battinelli,
P. (2017). Physiologically activated mammary fibroblasts promote post- E.M., and McAllister, S.S. (2012). Identification of luminal breast cancers
partum mammary cancer. JCI Insight 2, e89206. that establish a tumor-supportive macroenvironment defined by proan-
177. Tsai, K.K.C., Chuang, E.Y.Y., Little, J.B., and Yuan, Z.M. (2005). Cellular giogenic platelets and bone marrow-derived cells. Cancer Discov. 2,
mechanisms for low-dose ionizing radiation-induced perturbation of the 1150–1165.
breast tissue microenvironment. Cancer Res. 65, 6734–6744. 192. Pazolli, E., Luo, X., Brehm, S., Carbery, K., Chung, J.J., Prior, J.L., Doh-
178. Olumi, A.F., Grossfeld, G.D., Hayward, S.W., Carroll, P.R., Tlsty, T.D., erty, J., Demehri, S., Salavaggione, L., Piwnica-Worms, D., and Stewart,
and Cunha, G.R. (1999). Carcinoma-associated fibroblasts direct tumor S.A. (2009). Senescent stromal-derived osteopontin promotes preneo-
progression of initiated human prostatic epithelium. Cancer Res. 59, plastic cell growth. Cancer Res. 69, 1230–1239.
5002–5011. 193. Louault, K., Li, R.R., and DeClerck, Y.A. (2020). Cancer-associated fibro-
179. Cukierman, E. (2021). A reflection on how carcinoma-associated fibro- blasts: understanding their heterogeneity. Cancers 12, 3108.
blasts were recognized as active participants of epithelial tumorigenesis. 194. Louault, K., Bonneaud, T.L., Séveno, C., Gomez-Bougie, P., Nguyen, F.,
Cancer Res. 81, 4668–4670. Gautier, F., Bourgeois, N., Loussouarn, D., Kerdraon, O., Barillé-Nion, S.,
180. Shimshoni, E., Adir, I., Afik, R., Solomonov, I., Shenoy, A., Adler, M., Pu- et al. (2019). Interactions between cancer-associated fibroblasts and tu-
ricelli, L., Sabino, F., Savickas, S., Mouhadeb, O., et al. (2021). Distinct mor cells promote MCL-1 dependency in estrogen receptor-positive
extracellular-matrix remodeling events precede symptoms of inflamma- breast cancers. Oncogene 38, 3261–3273.
tion. Matrix Biol. 96, 47–68. 195. Hanahan, D., and Weinberg, R.A. (2011). Hallmarks of cancer: the next
181. Maris, P., Blomme, A., Palacios, A.P., Costanza, B., Bellahcène, A., Bian- generation. Cell 144, 646–674.
chi, E., Gofflot, S., Drion, P., Trombino, G.E., Di Valentin, E., et al. (2015). 196. Staneva, R., El Marjou, F., Barbazan, J., Krndija, D., Richon, S., Clark,
Asporin is a fibroblast-derived TGF-beta1 inhibitor and a tumor suppres- A.G., and Vignjevic, D.M. (2019). Cancer cells in the tumor core exhibit
sor associated with good prognosis in breast cancer. PLoS Med. 12, spatially coordinated migration patterns. J. Cell Sci. 132, jcs220277.
e1001871.
197. Massagué, J., and Ganesh, K. (2021). Metastasis-initiating cells and eco-
182. Boyle, S.T., Poltavets, V., Kular, J., Pyne, N.T., Sandow, J.J., Lewis, A.C., systems. Cancer Discov. 11, 971–994.
Murphy, K.J., Kolesnikoff, N., Moretti, P.A.B., Tea, M.N., et al. (2020).
198. Smith, C.J., Minas, T.Z., and Ambs, S. (2018). Analysis of tumor biology
ROCK-mediated selective activation of PERK signalling causes fibro-
to advance cancer health disparity research. Am. J. Pathol. 188,
blast reprogramming and tumour progression through a CRELD2-
304–316.
dependent mechanism. Nat. Cell Biol. 22, 882–895.
199. Yeh, A.C., and Ramaswamy, S. (2015). Mechanisms of cancer cell
183. Laklai, H., Miroshnikova, Y.A., Pickup, M.W., Collisson, E.A., Kim, G.E.,
dormancy–another hallmark of cancer? Cancer Res. 75, 5014–5022.
Barrett, A.S., Hill, R.C., Lakins, J.N., Schlaepfer, D.D., Mouw, J.K.,
et al. (2016). Genotype tunes pancreatic ductal adenocarcinoma tissue 200. Malik, R., Lelkes, P.I., and Cukierman, E. (2015). Biomechanical and
tension to induce matricellular fibrosis and tumor progression. Nat. biochemical remodeling of stromal extracellular matrix in cancer. Trends
Med. 22, 497–505. Biotechnol. 33, 230–236.

184. Novo, D., Heath, N., Mitchell, L., Caligiuri, G., MacFarlane, A., Reijmer, 201. Nazemi, M., and Rainero, E. (2020). Cross-talk between the tumor micro-
D., Charlton, L., Knight, J., Calka, M., McGhee, E., et al. (2018). Mutant environment, extracellular matrix, and cell metabolism in cancer. Front.
p53s generate pro-invasive niches by influencing exosome podocalyxin Oncol. 10, 239.
levels. Nat. Commun. 9, 5069. 202. Poltavets, V., Kochetkova, M., Pitson, S.M., and Samuel, M.S. (2018).
185. Wörmann, S.M., Song, L., Ai, J., Diakopoulos, K.N., Kurkowski, M.U., The Role of the extracellular matrix and its molecular and cellular regula-
Görgülü, K., Ruess, D., Campbell, A., Doglioni, C., Jodrell, D., et al. tors in cancer cell plasticity. Front. Oncol. 8, 431.
(2016). Loss of P53 function activates JAK2-STAT3 signaling to promote 203. Duda, D.G., Duyverman, A.M.M.J., Kohno, M., Snuderl, M., Steller,
pancreatic tumor growth, stroma modification, and gemcitabine resis- E.J.A., Fukumura, D., and Jain, R.K. (2010). Malignant cells facilitate

1604 Cell 186, April 13, 2023


ll
Review
lung metastasis by bringing their own soil. Proc. Natl. Acad. Sci. USA 220. Jedeszko, C., Victor, B.C., Podgorski, I., and Sloane, B.F. (2009). Fibro-
107, 21677–21682. blast hepatocyte growth factor promotes invasion of human mammary
ductal carcinoma in situ. Cancer Res. 69, 9148–9155.
204. Aggarwal, V., Montoya, C.A., Donnenberg, V.S., and Sant, S. (2021).
Interplay between tumor microenvironment and partial EMT as the driver 221. Orimo, A., Gupta, P.B., Sgroi, D.C., Arenzana-Seisdedos, F., Delaunay,
of tumor progression. iScience 24, 102113. T., Naeem, R., Carey, V.J., Richardson, A.L., and Weinberg, R.A.
(2005). Stromal fibroblasts present in invasive human breast carcinomas
205. Soikkeli, J., Podlasz, P., Yin, M., Nummela, P., Jahkola, T., Virolainen, S.,
promote tumor growth and angiogenesis through elevated SDF-1/
Krogerus, L., Heikkilä, P., von Smitten, K., Saksela, O., and Hölttä, E.
CXCL12 secretion. Cell 121, 335–348.
(2010). Metastatic outgrowth encompasses COL-I, FN1, and POSTN
up-regulation and assembly to fibrillar networks regulating cell adhesion, 222. Wang, F.T., Sun, W., Zhang, J.T., and Fan, Y.Z. (2019). Cancer-associ-
migration, and growth. Am. J. Pathol. 177, 387–403. ated fibroblast regulation of tumor neo-angiogenesis as a therapeutic
target in cancer. Oncol. Lett. 17, 3055–3065.
206. Goetz, J.G., Minguet, S., Navarro-Lérida, I., Lazcano, J.J., Samaniego,
R., Calvo, E., Tello, M., Osteso-Ibáñez, T., Pellinen, T., Echarri, A., et al. 223. Ji, Z., Tian, W., Gao, W., Zang, R., Wang, H., and Yang, G. (2021). Can-
(2011). Biomechanical remodeling of the microenvironment by stromal cer-associated fibroblast-derived interleukin-8 promotes ovarian cancer
caveolin-1 favors tumor invasion and metastasis. Cell 146, 148–163. cell stemness and malignancy through the Notch3-mediated signaling.
Front. Cell Dev. Biol. 9, 684505.
207. Erdogan, B., Ao, M., White, L.M., Means, A.L., Brewer, B.M., Yang, L.,
Washington, M.K., Shi, C., Franco, O.E., Weaver, A.M., et al. (2017). Can- 224. Nagasaki, T., Hara, M., Nakanishi, H., Takahashi, H., Sato, M., and Take-
cer-associated fibroblasts promote directional cancer cell migration by yama, H. (2014). Interleukin-6 released by colon cancer-associated fibro-
aligning fibronectin. J. Cell Biol. 216, 3799–3816. blasts is critical for tumour angiogenesis: anti-interleukin-6 receptor anti-
body suppressed angiogenesis and inhibited tumour-stroma interaction.
208. Sato, N., Kohi, S., Hirata, K., and Goggins, M. (2016). Role of hyaluronan
Br. J. Cancer 110, 469–478.
in pancreatic cancer biology and therapy: Once again in the spotlight.
Cancer Sci. 107, 569–575. 225. Nilsson, M.B., Langley, R.R., and Fidler, I.J. (2005). Interleukin-6,
secreted by human ovarian carcinoma cells, is a potent proangiogenic
209. Cox, T.R., and Erler, J.T. (2011). Remodeling and homeostasis of the
cytokine. Cancer Res. 65, 10794–10800.
extracellular matrix: implications for fibrotic diseases and cancer. Dis.
Model. Mech. 4, 165–178. 226. Pietras, K., Pahler, J., Bergers, G., and Hanahan, D. (2008). Functions of
paracrine PDGF signaling in the proangiogenic tumor stroma revealed by
210. Levental, K.R., Yu, H., Kass, L., Lakins, J.N., Egeblad, M., Erler, J.T.,
pharmacological targeting. PLoS Med. 5, e19.
Fong, S.F.T., Csiszar, K., Giaccia, A., Weninger, W., et al. (2009). Matrix
crosslinking forces tumor progression by enhancing integrin signaling. 227. Unterleuthner, D., Neuhold, P., Schwarz, K., Janker, L., Neuditschko, B.,
Cell 139, 891–906. Nivarthi, H., Crncec, I., Kramer, N., Unger, C., Hengstschläger, M., et al.
(2020). Cancer-associated fibroblast-derived WNT2 increases tumor
211. Lu, P., Weaver, V.M., and Werb, Z. (2012). The extracellular matrix: a dy- angiogenesis in colon cancer. Angiogenesis 23, 159–177.
namic niche in cancer progression. J. Cell Biol. 196, 395–406.
228. Yang, J., Lu, Y., Lin, Y.Y., Zheng, Z.Y., Fang, J.H., He, S., and Zhuang,
212. Navab, R., Strumpf, D., To, C., Pasko, E., Kim, K.S., Park, C.J., Hai, J., S.M. (2016). Vascular mimicry formation is promoted by paracrine
Liu, J., Jonkman, J., Barczyk, M., et al. (2016). Integrin alpha11beta1 reg- TGF-beta and SDF1 of cancer-associated fibroblasts and inhibited by
ulates cancer stromal stiffness and promotes tumorigenicity and metas- miR-101 in hepatocellular carcinoma. Cancer Lett. 383, 18–27.
tasis in non-small cell lung cancer. Oncogene 35, 1899–1908.
229. Yang, F., Tuxhorn, J.A., Ressler, S.J., McAlhany, S.J., Dang, T.D., and
213. Ferrara, B., Pignatelli, C., Cossutta, M., Citro, A., Courty, J., and Pie- Rowley, D.R. (2005). Stromal expression of connective tissue growth fac-
monti, L. (2021). The extracellular matrix in pancreatic cancer: descrip- tor promotes angiogenesis and prostate cancer tumorigenesis. Cancer
tion of a complex network and promising therapeutic options. Cancers Res. 65, 8887–8895.
13, 4442.
230. Chiavarina, B., Whitaker-Menezes, D., Migneco, G., Martinez-Out-
214. Cazet, A.S., Hui, M.N., Elsworth, B.L., Wu, S.Z., Roden, D., Chan, C.L., schoorn, U.E., Pavlides, S., Howell, A., Tanowitz, H.B., Casimiro, M.C.,
Skhinas, J.N., Collot, R., Yang, J., Harvey, K., et al. (2018). Targeting stro- Wang, C., Pestell, R.G., et al. (2010). HIF1-alpha functions as a tumor
mal remodeling and cancer stem cell plasticity overcomes chemoresist- promoter in cancer associated fibroblasts, and as a tumor suppressor
ance in triple negative breast cancer. Nat. Commun. 9, 2897. in breast cancer cells: Autophagy drives compartment-specific onco-
215. Shi, Y., Gao, W., Lytle, N.K., Huang, P., Yuan, X., Dann, A.M., Ridinger- genesis. Cell Cycle 9, 3534–3551.
Saison, M., DelGiorno, K.E., Antal, C.E., Liang, G., et al. (2019). Targeting 231. Ide, T., Kitajima, Y., Miyoshi, A., Ohtsuka, T., Mitsuno, M., Ohtaka, K.,
LIF-mediated paracrine interaction for pancreatic cancer therapy and Koga, Y., and Miyazaki, K. (2006). Tumor-stromal cell interaction under
monitoring. Nature 569, 131–135. hypoxia increases the invasiveness of pancreatic cancer cells through
216. Straussman, R., Morikawa, T., Shee, K., Barzily-Rokni, M., Qian, Z.R., the hepatocyte growth factor/c-Met pathway. Int. J. Cancer 119,
Du, J., Davis, A., Mongare, M.M., Gould, J., Frederick, D.T., et al. 2750–2759.
(2012). Tumour micro-environment elicits innate resistance to RAF inhib- 232. Ziani, L., Buart, S., Chouaib, S., and Thiery, J. (2021). Hypoxia increases
itors through HGF secretion. Nature 487, 500–504. melanoma-associated fibroblasts immunosuppressive potential and
217. Tape, C.J., Ling, S., Dimitriadi, M., McMahon, K.M., Worboys, J.D., inhibitory effect on T cell-mediated cytotoxicity. OncoImmunology 10,
Leong, H.S., Norrie, I.C., Miller, C.J., Poulogiannis, G., Lauffenburger, 1950953.
D.A., and Jørgensen, C. (2016). Oncogenic KRAS Regulates Tumor 233. Madsen, C.D., Pedersen, J.T., Venning, F.A., Singh, L.B., Moeendarbary,
Cell Signaling via Stromal Reciprocation. Cell 165, 1818–1920. E., Charras, G., Cox, T.R., Sahai, E., and Erler, J.T. (2015). Hypoxia and
218. De Francesco, E.M., Lappano, R., Santolla, M.F., Marsico, S., Caruso, A., loss of PHD2 inactivate stromal fibroblasts to decrease tumour stiffness
and Maggiolini, M. (2013). HIF-1alpha/GPER signaling mediates the and metastasis. EMBO Rep. 16, 1394–1408.
expression of VEGF induced by hypoxia in breast cancer associated fi- 234. Ben Baruch, B., Mantsur, E., Franco-Barraza, J., Blacher, E., Cukierman,
broblasts (CAFs). Breast Cancer Res. 15, R64. E., and Stein, R. (2020). CD38 in cancer-associated fibroblasts promotes
219. Fukumura, D., Xavier, R., Sugiura, T., Chen, Y., Park, E.C., Lu, N., Selig, pro-tumoral activity. Lab. Invest. 100, 1517–1531.
M., Nielsen, G., Taksir, T., Jain, R.K., and Seed, B. (1998). Tumor induc- 235. Zigrino, P., Kuhn, I., Bäuerle, T., Zamek, J., Fox, J.W., Neumann, S.,
tion of VEGF promoter activity in stromal cells. Cell 94, 715–725. Licht, A., Schorpp-Kistner, M., Angel, P., and Mauch, C. (2009). Stromal

Cell 186, April 13, 2023 1605


ll
Review
expression of MMP-13 is required for melanoma invasion and metas- blasts through p62-mTORC1 signaling promotes inflammation and
tasis. J. Invest. Dermatol. 129, 2686–2693. tumorigenesis. Cancer Cell 26, 121–135.
236. Sewell-Loftin, M.K., Bayer, S.V.H., Crist, E., Hughes, T., Joison, S.M., 252. Hessmann, E., Patzak, M.S., Klein, L., Chen, N., Kari, V., Ramu, I., Bapiro,
Longmore, G.D., and George, S.C. (2017). Cancer-associated fibroblasts T.E., Frese, K.K., Gopinathan, A., Richards, F.M., et al. (2018). Fibroblast
support vascular growth through mechanical force. Sci. Rep. 7, 12574. drug scavenging increases intratumoural gemcitabine accumulation in
237. Deng, Y., Cheng, J., Fu, B., Liu, W., Chen, G., Zhang, Q., and Yang, Y. murine pancreas cancer. Gut 67, 497–507.
(2017). Hepatic carcinoma-associated fibroblasts enhance immune sup- 253. Hirata, E., Girotti, M.R., Viros, A., Hooper, S., Spencer-Dene, B., Mat-
pression by facilitating the generation of myeloid-derived suppressor suda, M., Larkin, J., Marais, R., and Sahai, E. (2015). Intravital imaging re-
cells. Oncogene 36, 1090–1101. veals how BRAF inhibition generates drug-tolerant microenvironments
238. Gabrilovich, D.I., and Nagaraj, S. (2009). Myeloid-derived suppressor with high integrin beta1/FAK signaling. Cancer Cell 27, 574–588.
cells as regulators of the immune system. Nat. Rev. Immunol. 9, 162–174. 254. Ireland, L., Santos, A., Ahmed, M.S., Rainer, C., Nielsen, S.R., Quaranta,
V., Weyer-Czernilofsky, U., Engle, D.D., Perez-Mancera, P.A., Coupland,
239. Mao, X., Xu, J., Wang, W., Liang, C., Hua, J., Liu, J., Zhang, B., Meng, Q.,
S.E., et al. (2016). Chemoresistance in pancreatic cancer is driven by
Yu, X., and Shi, S. (2021). Crosstalk between cancer-associated fibro-
stroma-derived insulin-like growth factors. Cancer Res. 76, 6851–6863.
blasts and immune cells in the tumor microenvironment: new findings
and future perspectives. Mol. Cancer 20, 131. 255. Kaur, A., Webster, M.R., Marchbank, K., Behera, R., Ndoye, A., Kugel,
C.H., 3rd, Dang, V.M., Appleton, J., O’Connell, M.P., Cheng, P., et al.
240. Chakravarthy, A., Khan, L., Bensler, N.P., Bose, P., and De Carvalho,
(2016). sFRP2 in the aged microenvironment drives melanoma metas-
D.D. (2018). TGF-beta-associated extracellular matrix genes link can-
tasis and therapy resistance. Nature 532, 250–254.
cer-associated fibroblasts to immune evasion and immunotherapy fail-
ure. Nat. Commun. 9, 4692. 256. Mantoni, T.S., Lunardi, S., Al-Assar, O., Masamune, A., and Brunner, T.B.
(2011). Pancreatic stellate cells radioprotect pancreatic cancer cells
241. Kato, T., Noma, K., Ohara, T., Kashima, H., Katsura, Y., Sato, H., Komoto,
through beta1-integrin signaling. Cancer Res. 71, 3453–3458.
S., Katsube, R., Ninomiya, T., Tazawa, H., et al. (2018). Cancer-associ-
ated fibroblasts affect intratumoral CD8(+) and FoxP3(+) T cells via IL6 257. Borriello, L., Nakata, R., Sheard, M.A., Fernandez, G.E., Sposto, R., Mal-
in the tumor microenvironment. Clin. Cancer Res. 24, 4820–4833. var, J., Blavier, L., Shimada, H., Asgharzadeh, S., Seeger, R.C., and De-
Clerck, Y.A. (2017). Cancer-associated fibroblasts share characteristics
242. Mariathasan, S., Turley, S.J., Nickles, D., Castiglioni, A., Yuen, K., Wang,
and protumorigenic activity with mesenchymal stromal cells. Cancer
Y., Kadel, E.E., III, Koeppen, H., Astarita, J.L., Cubas, R., et al. (2018).
Res. 77, 5142–5157.
TGFbeta attenuates tumour response to PD-L1 blockade by contributing
to exclusion of T cells. Nature 554, 544–548. 258. Lotti, F., Jarrar, A.M., Pai, R.K., Hitomi, M., Lathia, J., Mace, A., Gantt,
G.A., Jr., Sukhdeo, K., DeVecchio, J., Vasanji, A., et al. (2013). Chemo-
243. Azambuja, J.H., Ludwig, N., Yerneni, S.S., Braganhol, E., and Whiteside,
therapy activates cancer-associated fibroblasts to maintain colorectal
T.L. (2020). Arginase-1+ exosomes from reprogrammed macrophages
cancer-initiating cells by IL-17A. J. Exp. Med. 210, 2851–2872.
promote glioblastoma progression. Int. J. Mol. Sci. 21, 3990.
259. Mutgan, A.C., Besikcioglu, H.E., Wang, S., Friess, H., Ceyhan, G.O., and
244. Nielsen, S.R., Quaranta, V., Linford, A., Emeagi, P., Rainer, C., Santos, A.,
Demir, I.E. (2018). Insulin/IGF-driven cancer cell-stroma crosstalk as a
Ireland, L., Sakai, T., Sakai, K., Kim, Y.S., et al. (2016). Macrophage-
novel therapeutic target in pancreatic cancer. Mol. Cancer 17, 66.
secreted granulin supports pancreatic cancer metastasis by inducing
liver fibrosis. Nat. Cell Biol. 18, 549–560. 260. Zhang, L., Yao, J., Li, W., and Zhang, C. (2018a). Micro-RNA-21 regulates
cancer-associated fibroblast-mediated drug resistance in pancreatic
245. Suzuki, J., Aokage, K., Neri, S., Sakai, T., Hashimoto, H., Su, Y., Yama-
cancer. Oncol. Res. 26, 827–835.
zaki, S., Nakamura, H., Tane, K., Miyoshi, T., et al. (2021). Relationship
between podoplanin-expressing cancer-associated fibroblasts and the 261. Shoucair, I., Weber Mello, F., Jabalee, J., Maleki, S., and Garnis, C.
immune microenvironment of early lung squamous cell carcinoma. (2020). The role of cancer-associated fibroblasts and extracellular vesi-
Lung Cancer 153, 1–10. cles in tumorigenesis. Int. J. Mol. Sci. 21, 6837.

246. Balsamo, M., Scordamaglia, F., Pietra, G., Manzini, C., Cantoni, C., Boi- 262. Zhang, H., Deng, T., Liu, R., Ning, T., Yang, H., Liu, D., Zhang, Q., Lin, D.,
tano, M., Queirolo, P., Vermi, W., Facchetti, F., Moretta, A., et al. (2009). Ge, S., Bai, M., et al. (2020a). CAF secreted miR-522 suppresses ferrop-
Melanoma-associated fibroblasts modulate NK cell phenotype and anti- tosis and promotes acquired chemo-resistance in gastric cancer. Mol.
tumor cytotoxicity. Proc. Natl. Acad. Sci. USA 106, 20847–20852. Cancer 19, 43.

247. Ziani, L., Safta-Saadoun, T.B., Gourbeix, J., Cavalcanti, A., Robert, C., 263. Aguirre-Ghiso, J.A. (2021). Translating the science of cancer dormancy
Favre, G., Chouaib, S., and Thiery, J. (2017). Melanoma-associated fibro- to the clinic. Cancer Res. 81, 4673–4675.
blasts decrease tumor cell susceptibility to NK cell-mediated killing 264. Bakhshandeh, S., Werner, C., Fratzl, P., and Cipitria, A. (2022). Microen-
through matrix-metalloproteinases secretion. Oncotarget 8, vironment-mediated cancer dormancy: Insights from metastability the-
19780–19794. ory. Proc. Natl. Acad. Sci. USA 119. e2111046118.
248. Altea-Manzano, P., Cuadros, A.M., Broadfield, L.A., and Fendt, S.M. 265. Liu, J., Tan, Y., Zhang, H., Zhang, Y., Xu, P., Chen, J., Poh, Y.C., Tang, K.,
(2020). Nutrient metabolism and cancer in the in vivo context: a metabolic Wang, N., and Huang, B. (2012). Soft fibrin gels promote selection and
game of give and take. EMBO Rep. 21, e50635. growth of tumorigenic cells. Nat. Mater. 11, 734–741.
249. Bertero, T., Oldham, W.M., Grasset, E.M., Bourget, I., Boulter, E., Pisano, 266. Keeratichamroen, S., Lirdprapamongkol, K., and Svasti, J. (2018). Mech-
S., Hofman, P., Bellvert, F., Meneguzzi, G., Bulavin, D.V., et al. (2019). Tu- anism of ECM-induced dormancy and chemoresistance in A549 human
mor-stroma mechanics coordinate amino acid availability to sustain tu- lung carcinoma cells. Oncol. Rep. 39, 1765–1774.
mor growth and malignancy. Cell Metab. 29, 124–140.e10. 267. Barkan, D., El Touny, L.H., Michalowski, A.M., Smith, J.A., Chu, I., Davis,
250. Chang, C.H., Qiu, J., O’Sullivan, D., Buck, M.D., Noguchi, T., Curtis, J.D., A.S., Webster, J.D., Hoover, S., Simpson, R.M., Gauldie, J., and Green,
Chen, Q., Gindin, M., Gubin, M.M., van der Windt, G.J.W., et al. (2015). J.E. (2010). Metastatic growth from dormant cells induced by a col-I-en-
Metabolic competition in the tumor microenvironment is a driver of can- riched fibrotic environment. Cancer Res. 70, 5706–5716.
cer progression. Cell 162, 1229–1241. 268. Barkan, D., Kleinman, H., Simmons, J.L., Asmussen, H., Kamaraju, A.K.,
251. Valencia, T., Kim, J.Y., Abu-Baker, S., Moscat-Pardos, J., Ahn, C.S., Re- Hoenorhoff, M.J., Liu, Z.Y., Costes, S.V., Cho, E.H., Lockett, S., et al.
ina-Campos, M., Duran, A., Castilla, E.A., Metallo, C.M., Diaz-Meco, (2008). Inhibition of metastatic outgrowth from single dormant tumor cells
M.T., and Moscat, J. (2014). Metabolic reprogramming of stromal fibro- by targeting the cytoskeleton. Cancer Res. 68, 6241–6250.

1606 Cell 186, April 13, 2023


ll
Review
269. Massagué, J. (2012). TGFbeta signalling in context. Nat. Rev. Mol. Cell (2015). Superoxide anion radicals induce IGF-1 resistance through
Biol. 13, 616–630. concomitant activation of PTP1B and PTEN. EMBO Mol. Med. 7, 59–77.
270. Oskarsson, T., Acharyya, S., Zhang, X.H.F., Vanharanta, S., Tavazoie, 288. Wlaschek, M., Maity, P., Makrantonaki, E., and Scharffetter-Kochanek,
S.F., Morris, P.G., Downey, R.J., Manova-Todorova, K., Brogi, E., and K. (2021). Connective tissue and fibroblast senescence in skin aging.
Massagué, J. (2011). Breast cancer cells produce tenascin C as a meta- J. Invest. Dermatol. 141, 985–992.
static niche component to colonize the lungs. Nat. Med. 17, 867–874. 289. Yasuda, T., Baba, H., and Ishimoto, T. (2023). Cellular senescence in the
271. Fane, M.E., Chhabra, Y., Alicea, G.M., Maranto, D.A., Douglass, S.M., tumor microenvironment and context-specific cancer treatment strate-
Webster, M.R., Rebecca, V.W., Marino, G.E., Almeida, F., Ecker, B.L., gies. FEBS J. 290, 1290–1302.
et al. (2022). Stromal changes in the aged lung induce an emergence
290. Yasuda, T., Koiwa, M., Yonemura, A., Miyake, K., Kariya, R., Kubota, S.,
from melanoma dormancy. Nature 606, 396–405.
Yokomizo-Nakano, T., Yasuda-Yoshihara, N., Uchihara, T., Itoyama, R.,
272. López-Otı́n, C., Blasco, M.A., Partridge, L., Serrano, M., and Kroemer, G. et al. (2021b). Inflammation-driven senescence-associated secretory
(2023). Hallmarks of aging: An expanding universe. Cell 186, 243–278. phenotype in cancer-associated fibroblasts enhances peritoneal
273. Cagan, A., Baez-Ortega, A., Brzozowska, N., Abascal, F., Coorens, dissemination. Cell Rep. 34, 108779.
T.H.H., Sanders, M.A., Lawson, A.R.J., Harvey, L.M.R., Bhosle, S., 291. Zhou, Z., Wei, J., Kuang, L., Zhang, K., Liu, Y., He, Z., Li, L., and Lu, B.
Jones, D., et al. (2022). Somatic mutation rates scale with lifespan across (2022). Characterization of aging cancer-associated fibroblasts draws
mammals. Nature 604, 517–524. implications in prognosis and immunotherapy response in low-grade gli-
274. Martincorena, I., Roshan, A., Gerstung, M., Ellis, P., Van Loo, P., McLa- omas. Front. Genet. 13, 897083.
ren, S., Wedge, D.C., Fullam, A., Alexandrov, L.B., Tubio, J.M., et al. 292. Friedman, G., Levi-Galibov, O., David, E., Bornstein, C., Giladi, A., Da-
(2015). Tumor evolution. High burden and pervasive positive selection diani, M., Mayo, A., Halperin, C., Pevsner-Fischer, M., Lavon, H., et al.
of somatic mutations in normal human skin. Science 348, 880–886. (2020). Cancer-associated fibroblast compositions change with breast
275. Wu, S., Zhu, W., Thompson, P., and Hannun, Y.A. (2018). Evaluating cancer progression linking the ratio of S100A4(+) and PDPN(+) CAFs to
intrinsic and non-intrinsic cancer risk factors. Nat. Commun. 9, 3490. clinical outcome. Nat. Cancer 1, 692–708.
276. Kaur, A., Ecker, B.L., Douglass, S.M., Kugel, C.H.,, 3rd, Webster, M.R., 293. Hogervorst, M., Rietveld, M., de Gruijl, F., and El Ghalbzouri, A. (2018). A
Almeida, F.V., Somasundaram, R., Hayden, J., Ban, E., Ahmadzadeh, shift from papillary to reticular fibroblasts enables tumour-stroma inter-
H., et al. (2019). Remodeling of the collagen matrix in aging skin promotes action and invasion. Br. J. Cancer 118, 1089–1097.
melanoma metastasis and affects immune cell motility. Cancer Discov. 9, 294. El Agha, E., Moiseenko, A., Kheirollahi, V., De Langhe, S., Crnkovic, S.,
64–81. Kwapiszewska, G., Szibor, M., Kosanovic, D., Schwind, F., Schermuly,
277. Marsh, E., Gonzalez, D.G., Lathrop, E.A., Boucher, J., and Greco, V. R.T., et al. (2017). Two-way conversion between lipogenic and myogenic
(2018). Positional stability and membrane occupancy define skin fibro- fibroblastic phenotypes marks the progression and resolution of lung
blast homeostasis in vivo. Cell 175, 1620–1633.e13. fibrosis. Cell Stem Cell 20, 261–273.e263.
278. Panwar, P., Lamour, G., Mackenzie, N.C.W., Yang, H., Ko, F., Li, H., and 295. Albrengues, J., Bertero, T., Grasset, E., Bonan, S., Maiel, M., Bourget, I.,
Brömme, D. (2015). Changes in structural-mechanical properties and de- Philippe, C., Herraiz Serrano, C., Benamar, S., Croce, O., et al. (2015).
gradability of collagen during aging-associated modifications. J. Biol. Epigenetic switch drives the conversion of fibroblasts into proinvasive
Chem. 290, 23291–23306. cancer-associated fibroblasts. Nat. Commun. 6, 10204.
279. Ding, X., Kakanj, P., Leptin, M., and Eming, S.A. (2021). Regulation of the 296. Hah, N., Sherman, M.H., Yu, R.T., Downes, M., and Evans, R.M. (2015).
wound healing response during aging. J. Invest. Dermatol. 141, Targeting transcriptional and epigenetic reprogramming in stromal cells
1063–1070. in fibrosis and cancer. Cold Spring Harb. Symp. Quant. Biol. 80, 249–255.
280. Moskalev, A.A., Shaposhnikov, M.V., Plyusnina, E.N., Zhavoronkov, A., 297. Plikus, M.V., Guerrero-Juarez, C.F., Ito, M., Li, Y.R., Dedhia, P.H., Zheng,
Budovsky, A., Yanai, H., and Fraifeld, V.E. (2013). The role of DNA dam- Y., Shao, M., Gay, D.L., Ramos, R., Hsi, T.C., et al. (2017). Regeneration
age and repair in aging through the prism of Koch-like criteria. Ageing of fat cells from myofibroblasts during wound healing. Science 355,
Res. Rev. 12, 661–684. 748–752.
281. Salzer, M.C., Lafzi, A., Berenguer-Llergo, A., Youssif, C., Castellanos, A., 298. Mezawa, Y., and Orimo, A. (2022). Phenotypic heterogeneity, stability
Solanas, G., Peixoto, F.O., Stephan-Otto Attolini, C., Prats, N., Aguilera, and plasticity in tumor-promoting carcinoma-associated fibroblasts.
M., et al. (2018). Identity noise and adipogenic traits characterize dermal FEBS J. 289, 2429–2447.
fibroblast aging. Cell 175, 1575–1590.e22.
299. Maddalena, M., Mallel, G., Nataraj, N.B., Shreberk-Shaked, M., Hassin,
282. Solé-Boldo, L., Raddatz, G., Schütz, S., Mallm, J.P., Rippe, K., Lonsdorf, O., Mukherjee, S., Arandkar, S., Rotkopf, R., Kapsack, A., Lambiase,
A.S., Rodrı́guez-Paredes, M., and Lyko, F. (2020). Single-cell transcrip- G., et al. (2021). TP53 missense mutations in PDAC are associated with
tomes of the human skin reveal age-related loss of fibroblast priming. enhanced fibrosis and an immunosuppressive microenvironment. Proc.
Commun. Biol. 3, 188. Natl. Acad. Sci. USA 118. e2025631118.
283. Marino, G.E., and Weeraratna, A.T. (2020). A glitch in the matrix: Age- 300. Elwakeel, E., Brüggemann, M., Fink, A.F., Schulz, M.H., Schmid, T., Sa-
dependent changes in the extracellular matrix facilitate common sites vai, R., Brüne, B., Zarnack, K., and Weigert, A. (2019). Phenotypic plas-
of metastasis. Aging Cancer 1, 19–29. ticity of fibroblasts during mammary carcinoma development. Int. J.
284. Faget, D.V., Ren, Q., and Stewart, S.A. (2019). Unmasking senescence: Mol. Sci. 20, 4438.
context-dependent effects of SASP in cancer. Nat. Rev. Cancer 19, 301. Grauel, A.L., Nguyen, B., Ruddy, D., Laszewski, T., Schwartz, S., Chang,
439–453. J., Chen, J., Piquet, M., Pelletier, M., Yan, Z., et al. (2020). TGFbeta-
285. Campisi, J. (2013). Aging, cellular senescence, and cancer. Annu. Rev. blockade uncovers stromal plasticity in tumors by revealing the existence
Physiol. 75, 685–705. of a subset of interferon-licensed fibroblasts. Nat. Commun. 11, 6315.
286. Ruhland, M.K., Coussens, L.M., and Stewart, S.A. (2016). Senescence 302. Avery, D., Govindaraju, P., Jacob, M., Todd, L., Monslow, J., and Puré, E.
and cancer: an evolving inflammatory paradox. Biochim. Biophys. Acta (2018). Extracellular matrix directs phenotypic heterogeneity of activated
1865, 14–22. fibroblasts. Matrix Biol. 67, 90–106.
287. Singh, K., Maity, P., Krug, L., Meyer, P., Treiber, N., Lucas, T., Basu, A., 303. Malik, R., Luong, T., Cao, X., Han, B., Shah, N., Franco-Barraza, J., Han,
Kochanek, S., Wlaschek, M., Geiger, H., and Scharffetter-Kochanek, K. L., Shenoy, V.B., Lelkes, P.I., and Cukierman, E. (2019). Rigidity controls

Cell 186, April 13, 2023 1607


ll
Review
human desmoplastic matrix anisotropy to enable pancreatic cancer cell hibits pathological functions of gastric cancer-associated fibroblasts.
spread via extracellular signal-regulated kinase 2. Matrix Biol. 81, 50–69. World J. Gastroenterol. 23, 8512–8525.
304. Chen, X., and Song, E. (2019). Turning foes to friends: targeting cancer- 319. Huo, Z., Su, Y., Dong, Y., Zheng, Y., Zhang, Q., Duan, Y., and Wang, G.
associated fibroblasts. Nat. Rev. Drug Discov. 18, 99–115. (2020). Rho-kinase inhibition by Fasudil promotes tumor maturation and
305. Fang, J., Xiao, L., Joo, K.I., Liu, Y., Zhang, C., Liu, S., Conti, P.S., Li, Z., apoptosis in small-cell lung cancer. Am. J. Transl. Res. 12, 4354–4370.
and Wang, P. (2016). A potent immunotoxin targeting fibroblast activa- 320. Whatcott, C.J., Ng, S., Barrett, M.T., Hostetter, G., Von Hoff, D.D., and
tion protein for treatment of breast cancer in mice. Int. J. Cancer 138, Han, H. (2017). Inhibition of ROCK1 kinase modulates both tumor cells
1013–1023. and stromal fibroblasts in pancreatic cancer. PLoS One 12, e0183871.
306. Formisano, L., Lu, Y., Servetto, A., Hanker, A.B., Jansen, V.M., Bauer, 321. Sanchez-Lopez, E., Flashner-Abramson, E., Shalapour, S., Zhong, Z.,
J.A., Sudhan, D.R., Guerrero-Zotano, A.L., Croessmann, S., Guo, Y., Taniguchi, K., Levitzki, A., and Karin, M. (2016). Targeting colorectal can-
et al. (2019). Aberrant FGFR signaling mediates resistance to CDK4/6 in- cer via its microenvironment by inhibiting IGF-1 receptor-insulin receptor
hibitors in ER+ breast cancer. Nat. Commun. 10, 1373. substrate and STAT3 signaling. Oncogene 35, 2634–2644.
307. Guagnano, V., Furet, P., Spanka, C., Bordas, V., Le Douget, M., Stamm, C., 322. Miao, L., Liu, Q., Lin, C.M., Luo, C., Wang, Y., Liu, L., Yin, W., Hu, S., Kim,
Brueggen, J., Jensen, M.R., Schnell, C., Schmid, H., et al. (2011). Discovery W.Y., and Huang, L. (2017). Targeting tumor-associated fibroblasts for
of 3-(2,6-dichloro-3,5-dimethoxy-phenyl)-1-6-[4-(4-ethyl-piperazin-1- therapeutic delivery in desmoplastic tumors. Cancer Res. 77, 719–731.
yl)-phenylamino]-pyrimidin-4-yl-1-methyl-urea (NVP-BGJ398), a potent 323. Haubeiss, S., Schmid, J.O., Mürdter, T.E., Sonnenberg, M., Friedel, G.,
and selective inhibitor of the fibroblast growth factor receptor family of re- van der Kuip, H., and Aulitzky, W.E. (2010). Dasatinib reverses cancer-
ceptor tyrosine kinase. J. Med. Chem. 54, 7066–7083. associated fibroblasts (CAFs) from primary lung carcinomas to a pheno-
308. Kim, S.M., Kim, H., Yun, M.R., Kang, H.N., Pyo, K.H., Park, H.J., Lee, type comparable to that of normal fibroblasts. Mol. Cancer 9, 168.
J.M., Choi, H.M., Ellinghaus, P., Ocker, M., et al. (2016). Activation of 324. Sherman, M.H., Yu, R.T., Engle, D.D., Ding, N., Atkins, A.R., Tiriac, H.,
the Met kinase confers acquired drug resistance in FGFR-targeted lung Collisson, E.A., Connor, F., Van Dyke, T., Kozlov, S., et al. (2014). Vitamin
cancer therapy. Oncogenesis 5, e241. D receptor-mediated stromal reprogramming suppresses pancreatitis
309. Palakurthi, S., Kuraguchi, M., Zacharek, S.J., Zudaire, E., Huang, W., Bo- and enhances pancreatic cancer therapy. Cell 159, 80–93.
nal, D.M., Liu, J., Dhaneshwar, A., DePeaux, K., Gowaski, M.R., et al. 325. Chan, J.S.K., Sng, M.K., Teo, Z.Q., Chong, H.C., Twang, J.S., and Tan,
(2019). The combined effect of FGFR inhibition and PD-1 blockade pro- N.S. (2018). Targeting nuclear receptors in cancer-associated fibroblasts
motes tumor-intrinsic induction of antitumor immunity. Cancer Immunol. as concurrent therapy to inhibit development of chemoresistant tumors.
Res. 7, 1457–1471. Oncogene 37, 160–173.
310. Perera, T.P.S., Jovcheva, E., Mevellec, L., Vialard, J., De Lange, D., Ver- 326. Ren, Y., Zhou, X., Liu, X., Jia, H.H., Zhao, X.H., Wang, Q.X., Han, L., Song,
hulst, T., Paulussen, C., Van De Ven, K., King, P., Freyne, E., et al. (2017). X., Zhu, Z.Y., Sun, T., et al. (2016). Reprogramming carcinoma associ-
Discovery and pharmacological characterization of JNJ-42756493 (Er- ated fibroblasts by AC1MMYR2 impedes tumor metastasis and improves
dafitinib), a functionally selective small-molecule FGFR family inhibitor. chemotherapy efficacy. Cancer Lett. 374, 96–106.
Mol. Cancer Ther. 16, 1010–1020.
327. Dauer, P., Zhao, X., Gupta, V.K., Sharma, N., Kesh, K., Gnamlin, P., Du-
311. Mertens, J.C., Fingas, C.D., Christensen, J.D., Smoot, R.L., Bronk, S.F., deja, V., Vickers, S.M., Banerjee, S., and Saluja, A. (2018). Inactivation of
Werneburg, N.W., Gustafson, M.P., Dietz, A.B., Roberts, L.R., Sirica, cancer-associated-fibroblasts disrupts oncogenic signaling in pancre-
A.E., and Gores, G.J. (2013). Therapeutic effects of deleting cancer- atic cancer cells and promotes its regression. Cancer Res. 78,
associated fibroblasts in cholangiocarcinoma. Cancer Res. 73, 897–907. 1321–1333.
312. Santos, A.M., Jung, J., Aziz, N., Kissil, J.L., and Puré, E. (2009). Targeting 328. Chronopoulos, A., Robinson, B., Sarper, M., Cortes, E., Auernheimer, V.,
fibroblast activation protein inhibits tumor stromagenesis and growth in Lachowski, D., Attwood, S., Garcı́a, R., Ghassemi, S., Fabry, B., and Del
mice. J. Clin. Invest. 119, 3613–3625. Rı́o Hernández, A. (2016). ATRA mechanically reprograms pancreatic
313. Sharma, M., Turaga, R.C., Yuan, Y., Satyanarayana, G., Mishra, F., Bian, stellate cells to suppress matrix remodelling and inhibit cancer cell inva-
Z., Liu, W., Sun, L., Yang, J., and Liu, Z.R. (2021). Simultaneously target- sion. Nat. Commun. 7, 12630.
ing cancer-associated fibroblasts and angiogenic vessel as a treatment 329. Lv, Y., Xu, C., Zhao, X., Lin, C., Yang, X., Xin, X., Zhang, L., Qin, C., Han,
for TNBC. J. Exp. Med. 218, e20200712. X., Yang, L., et al. (2018). Nanoplatform assembled from a CD44-targeted
314. Turaga, R.C., Sharma, M., Mishra, F., Krasinskas, A., Yuan, Y., Yang, prodrug and smart liposomes for dual targeting of tumor microenviron-
J.J., Wang, S., Liu, C., Li, S., and Liu, Z.R. (2021). Modulation of can- ment and cancer cells. ACS Nano 12, 1519–1536.
cer-associated fibrotic stroma by an integrin alpha(v)beta(3) targeting 330. Zhang, Q., Hou, X., Evans, B.J., VanBlaricom, J.L., Weroha, S.J., and
protein for pancreatic cancer treatment. Cell. Mol. Gastroenterol. Hepa- Cliby, W.A. (2018b). LY2157299 monohydrate, a TGF-betaR1 inhibitor,
tol. 11, 161–179. suppresses tumor growth and ascites development in ovarian cancer.
315. Chiappori, A.A., Eckhardt, S.G., Bukowski, R., Sullivan, D.M., Ikeda, M., Cancers 10, 260.
Yano, Y., Yamada-Sawada, T., Kambayashi, Y., Tanaka, K., Javle, M.M., 331. Hanley, C.J., Mellone, M., Ford, K., Thirdborough, S.M., Mellows, T.,
et al. (2007). A phase I pharmacokinetic and pharmacodynamic study of Frampton, S.J., Smith, D.M., Harden, E., Szyndralewiez, C., Bullock,
s-3304, a novel matrix metalloproteinase inhibitor, in patients with M., et al. (2018). Targeting the myofibroblastic cancer-associated fibro-
advanced and refractory solid tumors. Clin. Cancer Res. 13, 2091–2099. blast phenotype through inhibition of NOX4. J. Natl. Cancer Inst. 110,
316. Heo, M.B., Kim, S.Y., Yun, W.S., and Lim, Y.T. (2015). Sequential delivery 109–120.
of an anticancer drug and combined immunomodulatory nanoparticles 332. Yao, Y., Guo, Q., Cao, Y., Qiu, Y., Tan, R., Yu, Z., Zhou, Y., and Lu, N.
for efficient chemoimmunotherapy. Int. J. Nanomedicine 10, 5981–5992. (2018). Artemisinin derivatives inactivate cancer-associated fibroblasts
317. Yu, D., Li, W., Zhang, Y., and Zhang, B. (2016). Anti-tumor efficiency of through suppressing TGF-beta signaling in breast cancer. J. Exp. Clin.
paclitaxel and DNA when co-delivered by pH responsive ligand modified Cancer Res. 37, 282.
nanocarriers for breast cancer treatment. Biomed. Pharmacother. 83, 333. Ohshio, Y., Hanaoka, J., Kontani, K., and Teramoto, K. (2014). Tranilast
1428–1435. inhibits the function of cancer-associated fibroblasts responsible for
318. Wang, Z.F., Ma, D.G., Zhu, Z., Mu, Y.P., Yang, Y.Y., Feng, L., Yang, H., the induction of immune suppressor cell types. Scand. J. Immunol. 80,
Liang, J.Q., Liu, Y.Y., Liu, L., and Lu, H.W. (2017). Astragaloside IV in- 408–416.

1608 Cell 186, April 13, 2023


ll
Review
334. Mediavilla-Varela, M., Boateng, K., Noyes, D., and Antonia, S.J. (2016). 343. Li, Y., Chen, F., Chen, J., Chan, S., He, Y., Liu, W., and Zhang, G. (2020).
The anti-fibrotic agent pirfenidone synergizes with cisplatin in killing tu- Disulfiram/copper induces antitumor activity against both nasopharyn-
mor cells and cancer-associated fibroblasts. BMC Cancer 16, 176. geal cancer cells and cancer-associated fibroblasts through ROS/
335. Zalcman, G., Mazieres, J., Margery, J., Greillier, L., Audigier-Valette, C., MAPK and ferroptosis Pathways. Cancers 12, 138.
Moro-Sibilot, D., Molinier, O., Corre, R., Monnet, I., Gounant, V., et al. 344. Diaz, L.A., Jr., Coughlin, C.M., Weil, S.C., Fishel, J., Gounder, M.M., Law-
(2016). Bevacizumab for newly diagnosed pleural mesothelioma in the rence, S., Azad, N., O’Shannessy, D.J., Grasso, L., Wustner, J., et al.
Mesothelioma Avastin Cisplatin Pemetrexed Study (MAPS): a rando- (2015). A first-in-human phase I study of MORAb-004, a monoclonal anti-
mised, controlled, open-label, phase 3 trial. Lancet 387, 1405–1414. body to endosialin in patients with advanced solid tumors. Clin. Cancer
336. Xu, S., Yang, Z., Jin, P., Yang, X., Li, X., Wei, X., Wang, Y., Long, S., Res. 21, 1281–1288.
Zhang, T., Chen, G., et al. (2018). Metformin suppresses tumor progres- 345. Reardon, D.A., Akabani, G., Coleman, R.E., Friedman, A.H., Friedman,
sion by inactivating stromal fibroblasts in ovarian cancer. Mol. Cancer H.S., Herndon, J.E., 2nd, McLendon, R.E., Pegram, C.N., Provenzale,
Ther. 17, 1291–1302. J.M., Quinn, J.A., et al. (2006). Salvage radioimmunotherapy with murine
337. Narra, K., Mullins, S.R., Lee, H.O., Strzemkowski-Brun, B., Magalong, K., iodine-131-labeled antitenascin monoclonal antibody 81C6 for patients
Christiansen, V.J., McKee, P.A., Egleston, B., Cohen, S.J., Weiner, L.M., with recurrent primary and metastatic malignant brain tumors: phase II
et al. (2007). Phase II trial of single agent Val-boroPro (Talabostat) inhib- study results. J. Clin. Oncol. 24, 115–122.
iting Fibroblast Activation Protein in patients with metastatic colorectal
346. Ko, A.H., LoConte, N., Tempero, M.A., Walker, E.J., Kate Kelley, R.,
cancer. Cancer Biol. Ther. 6, 1691–1699.
Lewis, S., Chang, W.C., Kantoff, E., Vannier, M.W., Catenacci, D.V.,
338. Duluc, C., Moatassim-Billah, S., Chalabi-Dchar, M., Perraud, A., Samain, et al. (2016). A Phase I Study of FOLFIRINOX Plus IPI-926, a hedgehog
R., Breibach, F., Gayral, M., Cordelier, P., Delisle, M.B., Bousquet-Du- pathway inhibitor, for advanced pancreatic adenocarcinoma. Pancreas
bouch, M.P., et al. (2015). Pharmacological targeting of the protein syn- 45, 370–375.
thesis mTOR/4E-BP1 pathway in cancer-associated fibroblasts abro-
gates pancreatic tumour chemoresistance. EMBO Mol. Med. 7, 735–753. 347. Olive, K.P., Jacobetz, M.A., Davidson, C.J., Gopinathan, A., McIntyre, D.,
Honess, D., Madhu, B., Goldgraben, M.A., Caldwell, M.E., Allard, D.,
339. Diop-Frimpong, B., Chauhan, V.P., Krane, S., Boucher, Y., and Jain, R.K.
et al. (2009). Inhibition of Hedgehog signaling enhances delivery of
(2011). Losartan inhibits collagen I synthesis and improves the distribu-
chemotherapy in a mouse model of pancreatic cancer. Science 324,
tion and efficacy of nanotherapeutics in tumors. Proc. Natl. Acad. Sci.
1457–1461.
USA 108, 2909–2914.
340. Izumi, D., Ishimoto, T., Miyake, K., Sugihara, H., Eto, K., Sawayama, H., 348. Ham, I.H., Oh, H.J., Jin, H., Bae, C.A., Jeon, S.M., Choi, K.S., Son, S.Y.,
Yasuda, T., Kiyozumi, Y., Kaida, T., Kurashige, J., et al. (2016). CXCL12/ Han, S.U., Brekken, R.A., Lee, D., and Hur, H. (2019). Targeting
CXCR4 activation by cancer-associated fibroblasts promotes integrin interleukin-6 as a strategy to overcome stroma-induced resistance to
beta1 clustering and invasiveness in gastric cancer. Int. J. Cancer 138, chemotherapy in gastric cancer. Mol. Cancer 18, 68.
1207–1219. 349. Hingorani, S.R., Zheng, L., Bullock, A.J., Seery, T.E., Harris, W.P., Sigal,
341. Ma, Y., Wang, Y., Xu, Z., Wang, Y., Fallon, J.K., and Liu, F. (2017). D.S., Braiteh, F., Ritch, P.S., Zalupski, M.M., Bahary, N., et al. (2018).
Extreme low dose of 5-fluorouracil reverses MDR in cancer by sensitizing HALO 202: randomized phase II study of PEGPH20 plus Nab-Pacli-
cancer associated fibroblasts and down-regulating P-gp. PLoS One 12, taxel/gemcitabine versus nab-paclitaxel/gemcitabine in patients with un-
e0180023. treated, metastatic pancreatic ductal adenocarcinoma. J. Clin. Oncol.
36, 359–366.
342. Kumar, V., Donthireddy, L., Marvel, D., Condamine, T., Wang, F., Lavilla-
Alonso, S., Hashimoto, A., Vonteddu, P., Behera, R., Goins, M.A., et al. 350. Thompson, C.B., Shepard, H.M., O’Connor, P.M., Kadhim, S., Jiang, P.,
(2017). Cancer-associated fibroblasts neutralize the anti-tumor effect of Osgood, R.J., Bookbinder, L.H., Li, X., Sugarman, B.J., Connor, R.J.,
CSF1 receptor blockade by inducing PMN-MDSC infiltration of tumors. et al. (2010). Enzymatic depletion of tumor hyaluronan induces antitumor
Cancer Cell 32, 654–668.e5. responses in preclinical animal models. Mol. Cancer Ther. 9, 3052–3064.

Cell 186, April 13, 2023 1609

You might also like