Skip to main content
Cancer Science logoLink to Cancer Science
. 2025 Jun 25;116(9):2347–2361. doi: 10.1111/cas.70133

How Do Cancer Cells Create Cancer‐Associated Fibroblast Subtypes? Impacts of Extracellular Vesicles on Stromal Diversity

Yutaka Naito 1,2,
PMCID: PMC12400057  PMID: 40566852

ABSTRACT

Cancer‐associated fibroblasts (CAFs) are the major component of the tumor stroma. They mediate various attributes of tumor cells, such as cell growth, migration, invasion, angiogenesis, metabolic reprogramming, apoptosis, immune regulation, and extracellular matrix reconstitution, all related to cancer progression and treatment resistance. Although many researchers have recognized CAF heterogeneity, recent technological advances have emphasized the functional and phenotypic diversity of CAFs in cancer progression. Why are these CAF subtypes generated within tumor tissues? And how do cancer cells dictate such heterogeneous subtypes of CAFs? This review will highlight the CAF subtypes within the tumor microenvironment and their role in tumor progression. CAF subtype induction by extracellular vesicles (EVs) and their significance, which we reported previously, is also discussed.

Keywords: cancer‐associated fibroblasts, extracellular vesicles, heterogeneity, subtypes, tumor microenvironment


Cancer‐associated fibroblasts (CAFs) are the major component of the tumor stroma. This review will highlight the CAF subtypes within the tumor microenvironment and their role in tumor progression. CAF subtype induction by extracellular vesicles (EVs) and their significance, which we reported previously, are also discussed.

graphic file with name CAS-116-2347-g003.jpg


Abbreviations

ANTXR1

Anthrax Toxin Receptor 1

apCAFs

antigen‐presenting CAFs

BM‐MSCs

bone marrow‐derived mesenchymal stem cells

CAFs

cancer‐associated fibroblasts

CXCL12

CXC‐chemokine ligand 12

DPP4

dipeptidyl peptidase 4

Dpt

dermatopontin

ECM

extracellular matrix

EMT

epithelial‐mesenchymal transition

EVs

extracellular vesicles

FAP

fibroblast activation protein

FSP1

fibroblast‐specific protein‐1

GC

gastric cancer

HGF

hepatocyte growth factor

Hh

Hedgehog

Hmgb1

high‐mobility group box 1

iCAFs

inflammatory CAFs

ICC

intrahepatic cholangiocarcinoma

ICI

immune checkpoint inhibitor

IL

interleukin

IL1R1

IL‐1 receptors

Irf1

interferon regulatory factor 1

ITGA11

integrin subunit alpha 11

LIF

leukemia inhibitory factor

LRRC15

leucine‐rich repeat containing 15

MET

mesenchymal‐epithelial transition

miRNAs

microRNAs

MMPs

metalloproteinases

MVBs

multivesicular bodies

myCAFs

myofibroblastic CAFs

NG2

neuron‐glial antigen 2

PDAC

pancreatic ductal adenocarcinoma

PDGFR

platelet‐derived growth factor receptor

PD‐L1

programmed death ligand 1

PDPN

podoplanin

Pi16

peptidase inhibitor 16

Shh

Sonic hedgehog

TGFBR2

transforming growth factor‐beta receptor 2

TGF‐β

transforming growth factor‐beta

TME

tumor microenvironment

Tslp

thymic stromal lymphopoietin

YAP

yes‐associated protein

α‐SMA

alpha‐smooth muscle actin

1. Introduction

Over the past 3 decades, the tumor microenvironment (TME) has attracted considerable attention as the essential regulator of tumor progression. The TME comprises noncellular components, such as the extracellular matrix and various cellular components, including fibroblasts, endothelial cells, and immune cells. In healthy circumstances, such stromal cells sustain physiological homeostasis and prevent disease progression, such as carcinogenesis [1, 2, 3, 4, 5]. However, cancer cells that evade such normal homeostatic mechanisms can convert phenotypes of these noncellular and cellular components into TME characteristics favoring their progression [2, 6]. Of these TME components, cancer‐associated fibroblasts (CAFs) play an essential role in tumor progression by promoting tumor metastasis and the mechanical alteration of tumor tissues, influencing angiogenesis, immune cell infiltration and activation, and therapeutic resistance [7, 8]. Therefore, understanding the biological basis underlying the intercellular interactions between cancer cells and CAFs can provide new avenues to improve tumor treatment.

However, there is a challenge that needs to be addressed to target the TME, particularly CAFs, as a novel treatment modality. That is, the phenotypic and functional diversity of CAFs. Several researchers so far have recognized the heterogeneity of CAFs through various types of analyses, including immunohistochemical techniques [9, 10, 11, 12]. In addition, CAFs have historically been considered a tumor‐promoting factor, but some evidence has shown that some CAFs also retained tumor‐suppressive functions [13, 14, 15]. Consistent with these observations, current single‐cell resolution platforms have demonstrated that CAFs are heterogeneous populations with various subtypes. Multiple factors, including their cellular origin, humoral factors, extracellular vesicles (EVs), and physical stimuli, may be responsible for generating such CAF heterogeneity. CAF heterogeneity is thus engaged in the different aspects of cancer biology, making understanding the biology of the TME and its therapeutic applications more challenging. In this review, recent advances in understanding the mechanisms of creation of CAF heterogeneity, the functions of CAF subtypes in cancer progression, and the role of extracellular vesicles in CAF subtype induction and function are discussed.

2. What Are Fibroblasts? Their Functions Are in Normal Homeostasis

Fibroblasts are key players in maintaining tissue and organ homeostasis. One of their essential functions is the provision of molecules that form the extracellular matrix (ECM), the scaffolding of the cell, such as collagen, fibronectin, laminins, elastins, integrins, and metalloproteinases (MMPs), and other tissue‐specific ECM proteins [7, 8, 16]. By providing these proteins, fibroblasts sustain cellular scaffolds and the architectures of tissues and organs to promote correct cellular proliferation, differentiation, and tissue morphogenesis. For example, fibroblasts are temporarily activated during lung development to support alveolarization by producing elastin and tenascin frameworks [17]. The generation of mechanical force is also a typical function of fibroblasts. Physical contraction by activated fibroblasts appears to involve alveolar septum formation in alveolarization [18]. In particular, during the wound‐healing process, fibroblasts migrate into the wound and then enter the activated state, termed myofibroblasts, that are the primary source of ECM molecules to support epithelial cell migration into the wound [1, 19]. Along with these functions of fibroblasts, they also serve as a source of humoral factors, such as growth factors, hormones, and chemokines [1, 8, 16]. In the pancreas and liver, other types of fibroblasts, termed stellate cells, have lipid droplets containing retinoic acid [20]. The activated state of stellate cells is closely involved in the progressive pathogenesis in the liver and pancreas, such as fibrosis, carcinogenesis, and the chemoresistance of tumors [21, 22, 23]. Within the population of PDGFR‐expressing myofibroblasts at the crypt–villus junction of intestinal tissue, two distinct mesenchymal cell types have been identified. Telocytes, characterized by platelet‐derived growth factor receptor alpha (PDGFRα)‐high Foxl1‐positive, are located at the villus base and function as a reservoir for BMP ligands. In contrast, trophocytes, defined by PDGFRα‐low CD81‐positive, reside beneath crypts and express the BMP inhibitor Grem1, thereby supporting and maintaining intestinal stem cells [24]. Fibroblasts are, therefore, essential for the maintenance of proper biological functions.

Although fibroblasts are often considered to have homogeneous characteristics, their properties and cell lineages differ considerably among tissues and organs. Indeed, single‐cell resolution analysis showed the phenotypical and functional differences of fibroblast lineages among all mouse organs [25]. Stellate cells have unique features, such as lipid droplet storage, distinct from other fibroblasts within tissues other than the liver and pancreas. The origin of most fibroblasts is the primitive mesenchyme that develops from the mesoderm, and other mesenchymal cells, including adipocytes and osteoblasts, share the same embryonic origin [1, 7, 8]. Therefore, it is not surprising that the fibroblasts comprise heterogeneous subtypes. Importantly, no specific markers of fibroblasts have been found so far that are not expressed in other cell types. It is necessary to bear in mind the histological and embryological heterogeneity of the fibroblasts from which they originate to understand how the heterogeneity of CAFs comes about.

3. Definitions of CAFs

Generally, CAFs refer to the fibroblasts or stellate cells (in the pancreas and liver) that reside within the tumor stroma [26]. CAFs are similar in nature to activated fibroblasts during wound healing [1, 7]. The wound‐healing mechanism, an intrinsic function of fibroblasts, acts in response to tissue destruction by cancer cells. However, as Harold Dvorak postulated in 1986, “tumours are non‐healing wounds,” [27] and tissue destruction by cancer cells is persistent, unlike wound healing, creating a unique environment in which inflammatory and wound‐healing responses are prevalent. Such a “non‐healing wounds” response creates CAFs stimulated by variant signals from cancer cells, other cell types, and ECMs within the TME. Along with the heterogeneity of CAFs in their origin, the variance of extracellular signals can generate highly complex CAF subtypes within the tumor stroma.

The same issue of the lack of comprehensive fibroblast markers applies to CAFs. Therefore, defining the entire CAF population precisely and uniformly using simple marker expressions is challenging. So far, the definition likely depends on the study's design and purpose. For instance, single‐cell resolution analysis uses gene expressions of collagens, decolin, and complement C1r to isolate fibroblast lineages within tumor tissues [28]. For in vitro experimental use, fibroblasts that crawl out from tumor tissues in culture are exploited [29, 30]. Combining several antibodies or gene expressions is also used to isolate specific CAF subtypes [9, 31, 32, 33, 34, 35, 36, 37, 38]. Conversely, combining multiple gene or protein expressions for other cell lineage markers can separate CAFs that do not express these lineage markers [8, 39]. Another criterion of CAFs is cells that lack genetic mutation within a tumor to discriminate them from cancer cells showing epithelial–mesenchymal transition (EMT) [8]. A self‐developed fibroblast single‐cell atlas is also used to identify universal fibroblast lineages across the whole organ [25]. It should clarify and accumulate information on how CAFs are defined and separated and what CAF functions have been identified to understand CAF biology and develop novel therapeutic modalities.

4. Definition of CAF Subtypes and Their Functions for Tumor Progression

Recent advances in single‐cell resolution techniques have identified numerous CAF subtypes. The well‐known subtypes are myofibroblastic CAFs (myCAFs), inflammatory CAFs (or immunomodulatory CAFs: iCAFs), and antigen‐presenting CAFs (apCAFs) (Figure 1 and Table 1). However, finer subpopulations with specific gene expressions appear even within these subtypes. Some subtypes do not belong to these three groups (Other CAF subtypes in Figure 1 and Table 1). This review focuses on particular CAF subtypes whose molecular mechanisms have been investigated for their role in cancer progression.

FIGURE 1.

FIGURE 1

Functions and markers for each CAF subtype. Cancer‐associated fibroblasts consist of various subtypes. MyCAFs are similar to the myofibroblast phenotype and express ECM‐related or mechanical contraction‐related genes. They promote cancer cell growth, migration, invasion, and inhibition of T‐cell infiltration via mechanotransduction and humoral factors. However, contrary to the tumor‐promoting functions, myCAFs also include tumor‐suppressive subgroups via a mechanical ECM barrier to suppress cancer cell invasion. The iCAFs are a subtype that produces cytokines and chemokines to enhance tumor growth and immunosuppression. Distinct from myCAFs, they have no myofibroblast‐like features, such as ECM‐related genes. The apCAFs express MHC Class II molecules involved in antigen presentation by immune cells and act as a decoy receptor that differentiates naïve CD4‐positive T cells from Tregs. As with other CAF types, many subtypes have been elucidated, including vCAFs and FAP+ PDPN+ CAFs. “Steady‐state” CAFs are also found and likely include both the tumor‐promoter and the tumor‐suppressor subgroups.

TABLE 1.

Functions of CAF subtypes in each tumor type discussed in this review.

Tumor type CAF subtype (subpopulation) Functions Origin of CAF subtype Induction mechanisms Subtype markers References
Pancreatic ductal adenocarcinoma (PDAC) myCAF Not investigated Pancreatic Stellate Cells Jaxtacrine interactions α‐SMA [33]
iCAF Sustain the survival of PDAC organoids Pancreatic Stellate Cells Paracrine interactions IL‐6, IL‐11, LIF etc. [33]
myCAF Not investigated Not investigated Not investigated ACTA2, TAGLN, MMP11, MYL9, HOPX, POSTN, TPM1, TPM2 [31]
iCAF Not investigated Not investigated Not investigated IL6, PDGFRA, CXCL12, CFD, DPT, LMNA, AGTR1, HAS1, CXCL1, CXCL2, CCL2, IL8 [31]
apCAF Induce CD25 and CD69 in cocultured T cells Not investigated Not investigated COL1A1, CD74, HLA‐DRA, HLA‐DPA1, HLA‐DQA1, SLPI [31]
EGFR/ERBB2 signaling activated myCAF Promote PDAC metastasis Pancreatic Stellate Cells TGF‐β signaling EGFR/ERBB2 activation, AREG, Dusp6, ECM‐associated and TGF‐β dependent gene signatures, and KRAS signaling related genes etc. [40]
iCAF Not investigated Pancreatic Stellate Cells IL1 signaling JAK/STAT signaling and NF‐κB related genes [40]
apCAF Not investigated Not investigated Not investigated Ly6C‐ and MHCII+ [40]
LRRC15+ myCAF Induce therapeutic resistance of cancer cells against immune checkpoint inhibitors Pancreatic Stellate Cells TGF‐β signaling LRRC15 and PDPN [41]
iCAF Not investigated Pancreatic Stellate Cells May induced by IL1 signaling CXCL1, HAS1 etc. [41]
LRRC15+ myCAF Suppress CD8‐T cell functions to support the immune evasion of cancer cells Dermatopontin (DPT) + fibroblasts TGF‐β signaling LRRC15 and PDPN [42]
apCAF Act as a decoy receptor that differentiates naïve CD4‐positive T cells into regulatory T‐cells (Tregs) Mesothelial cells Both IL1 and TGF‐β signaling Cd74, H2‐As, H2‐Ab1, H2‐Eb1, Msln, Uok3b, Lrrn4, Krt19 [43]
FAP+ CAF Suppress antitumor immune effects via CXC‐chemokine ligand 12 (CXCL12) production Not investigated Not investigated FAP [44, 45]
Meflin+ CAF Suppress tumor progression and enhance the response to immune checkpoint inhibitors Mesenchymal stem cells Depending on Meflin expression Meflin [46, 47, 48]
Breast cancer CAF‐S1 Enriched in triple‐negative breast cancer and associated with an immunosuppressive microenvironment and promote Treg differentiation via CXCL12 Not investigated Not investigated CD29Med, FAPHi, FSP1Low‐Hi, α‐SMAHi, PDGFRbMed‐Hi, CAV1Low [34]
CAF‐S2 Enriched in Luminal A breast cancer Not investigated Not investigated CD29Low, FAPNeg, FSP1Neg‐Low, α‐SMANeg, PDGFRbNeg, CAV1Neg [34]
CAF‐S3 Enriched in Juxta‐tumor Not investigated Not investigated CD29Med, FAPNeg, FSP1Med‐Hi, α‐SMANeg‐Low, PDGFRbMed, CAV1Neg‐Low [34]
CAF‐S4 Enriched in HER2 breast cancer Not investigated Not investigated CD29Hi, FAPNeg, FSP1Low‐Med, α‐SMAHi, PDGFRbLow‐Med, CAV1Neg‐Low [34]
CAF‐S1 Stimulate cancer cell migration and invasion through CXCL12 and TGF‐β signallings to promote metastasis Not investigated Not investigated FAPHigh, CD29Med‐High, α‐SMAHigh, PDPNHigh, PDGFRβHigh [49]
CAF‐S4 High contractile capacity to support cancer cell invasion Not investigated Not investigated FAPLow‐Med, CD29High, α‐SMAHigh, PDPNLow, PDGFRβMed [49]
vCAF Associated with microvasculature signatures Perivascular cells (pericytes) Not investigated Vascular development and angiogenesis‐related genes [39]
matrix CAF Associated with invasion sigunatures of cancer cells Resident fibroblasts Not investigated ECM and EMT‐related genes [39]
cycling CAF May be proliferating segment of vCAFs vCAF subpopulation Not investigated Miki67, cell cycle related genes [39]
developmental CAF Not investigated May derived from cancer cells possessing EMT Not investigated Tissue development, cell differentiation, and ECM‐related genes; Similar expression pattern to tumor epithelium [39]
Colon cancer myCAF within liver metastatic sites Possess hyaluronan‐mediated tumor‐promotive function and may also have tumor‐suppressive function via creating ECM barrier May derived from hepatic stellate cells Not investigated Acta2 and several collagen isoforms [50]
iCAF within liver metastatic sites Secret HGF to enhance metastatic tumor growth May derived from hepatic stellate cells Not investigated Growth factor and inflammatory gene expressions [50]
mesothelial CAF (mesCAF) within liver metastatic sites Not investigated May derived from portal fibroblasts in liver Not investigated Mesothelial marker expressions (Msln, Upk1b, and Up3kb) [50]
myCAF May have tumor‐suppressive function but not investigated precisely Not investigated Not investigated ACTA2, RGS5, CSPG4, and AOC3 [51]
IL1R1+ iCAF Suppress CD8‐positive killer T cells and induce tumor‐promoting macrophages Resident fibroblasts IL1 signaling IL1R1, CXCL14, CXCL12, CXCL16 [51]
IL11+ CAF Promote tumor formation and growth Resident fibroblasts IL11 induced STAT3 signaling IL11 [51]
Gastric cancer myCAF‐like Not investigated Resident fibroblasts TGF‐β signaling ACTA2, COL4A1 [29]
iCAF‐like Create a favorable tumor microenvironment for cancer progression via chemokines Resident fibroblasts miRNAs (miR‐193b etc.) packaged into extracellular vesicles CXCL1, CXCL2, CXCL8 [29]
Intracolangio carcinoma (ICC) myCAF Promote tumor growth through hyaluronan synthase 2 Hepatic Stellate Cells Not investigated ECM‐related genes, Col1a1, and SERPINF1 [52]
iCAF Promote tumor growth through HGF‐MET signaling Hepatic Stellate Cells Not investigated Inflammatory and growth factor related genes and Rgs5 [52]
mesCAF Not investigated Portal fibroblasts Not investigated Msln, Upk1b, Upk3b, and Gpm6a as markers of portal fibroblasts/mesothelial cells [52]
vCAF Contribute tumorigenesis and cancer stemness through the IL‐6/IL‐6R signaling Not investigated Not investigated IL6, MYH11, CD146, CCL8, RGS5, ACTA2, COL1A2, PDGFRB [53]
matrix CAF Associated with ECM and stroma‐ related gene signatures Not investigated Not investigated POSTN, COL5A1, COL5A2, COL6A3, FN1, LUM, DCN, VCAN, ACTA2, COL1A2, PDGFRB [53]
iCAF Not investigated Not investigated Not investigated FBLN1, C3, IGF1, CXCL1, IGFBP6, SLP1, SAA1, ACTA2, COL1A2, PDGFRB [53]
apCAF Not investigated Not investigated Not investigated HLA‐DRB1, CD74, HLA‐DRA, ACTA2, COL1A2, PDGFRB [53]
Lung cancer PDPN+ CAF Increase EGFR‐TKI resistance in lung cancer cells Not investigated Not investigated PDPN [54]
apCAF Directly activate the TCRs of CD4 T cells and act as tumor suppressor. May originate from ATII cells Not investigated MHCII, SLP1, IL6, CFD, C1QA, C1QB [55]
myCAF Not investigated Not investigated Not investigated COL1A1, COL1A2, COL3A1, C1QTNF3, FAP, PDGFRA, CXCL12, ACTA2, MMP11, MMP13 [56]
iCAF May participate in brain metastasis of cancer cells through MET‐HGF signaling pathway Not investigated Not investigated FAP, PDGFRA, CXCL12, CXCL2, C7, APOD [56]
apCAF May participate in bone metastasis of cancer cells through SPP1‐CD44/SPP1‐PTGER4 signaling pathway Not investigated Not investigated CXCL2, CD74, HLA‐DRA, HLA‐DPB1 and HLA‐DPA1, RGS1, HLA‐DRB1, CCL4 [56]
myCAF Not investigated Resident fibroblasts Extracellular vesicles Proliferation‐ and ECM‐related genes [57]
Bladder cancer ITGA11+ PDGFRα+ myCAF Promote lymphangiogenesis and assist in intravasation to aid lymphatic metastasis Resident fibroblasts TGF‐β signaling ITGA11 and PDGFRα [58]
iCAF Induce EMT in cancer cells through IL‐6 signaling Resident fibroblasts Cancer‐derived extracellular vesicles IL6, ACTA2, FAP [59]
Ovarian cancer CAF‐S1 Increasing attraction, survival, and differentiation of T lymphocytes Not investigated Not investigated CD29Med‐Hi, FAPHi SMAMed‐Hi, FSP1Med‐Hi, PDGFRβMed‐Hi, CAV1Low [35]
CAF‐S2 Not investigated Not investigated Not investigated CD29Low FAP‐Neg SMANeg‐Low FSP1Neg‐Low PDGFRβNeg‐Low CAV1Neg [35]
CAF‐S3 Not investigated Not investigated Not investigated CD29Med FAPLow SMALow FSP1Med‐Hi PDGFRβMed CAV1‐Neg‐Low [35]
CAF‐S4 Not investigated (Could not be maintained in culture) Not investigated Not investigated CD29Hi FAPLow SMAHi FSP1Hi PDGFRβMed‐HiCAV1Neg‐Low [35]
ANTXR1+ ECM‐myCAF Associated with chemotherapy resistance and suppression of CD8‐positive T‐cell activation Not investigated Not investigated ANTXR1, FAP, ACTA2, CD29, FSP1, LRRC15, YAP1 [36]
Detox‐iCAF Enhance CD8‐positive T‐cell migration Not investigated Not investigated ANTXR1‐negative, lower expression of FAP and α‐SMA [36]
Skin cancer Matrix CAF Form ECM barrier to restrict T‐cell invasion into tumors May derived from resident fibroblasts Not investigated FAP, α‐SMA, COL1A1, COL1A2, COL3A1, Lumican, Periostin, Tenascin‐C [60]
iCAF Have immunomodulatory functions and may contribute to the induction of T‐cell exhaustion May derived from resident fibroblasts Not investigated IL6, IL8, IDO1, MMP1, MMP3, FAP, α‐SMA [60]
RGS5+ CAF Not investigated May derived from resident fibroblasts Not investigated FAP, α‐SMA, RGS5, TAGLN, DES [60]
iCAF‐like Enhance recruitment of neutrophil into the lung metastatic niche Resident fibroblasts in lung RNAs (Hmgb1, Tslp, Irf1) in extracellular vesicles Inflammatory genes including IL1α, IL1β, CCL2, CCL3, CCL5, CXCL10, CXCL12 [61]

Note: For some CAF subtype markers, classifications by expression level were used. In these cases, “Hi” for high expression, “Med” for medium expression, “Low” for low expression, “+” or “positive” for expressing marker and “‐” or “negative” for no marker expression were described. In addition to CAF subtypes discussed in this review, various other subtypes have been reported.

5. Myofibroblastic CAFs (myCAFs)

MyCAFs are similar to the myofibroblast phenotype described above, which arises during wound healing and expresses ECM‐related or mechanical contraction‐related genes, such as alpha‐smooth muscle actin and collagens (Figure 1 and Table 1). Thus, myCAFs are likely capable of cancer cell migration and invasion through ECM stiffening [62, 63]. In particular, myCAFs play a principal role in the mechanotransduction regulated by ECM hardening, leading to yes‐associated protein (YAP) activation and nuclear localization to induce survival and proliferation signals in cancer cells [64]. Paracrine signals derived from myCAFs, such as transforming growth factor‐beta (TGF‐β) signaling, mediate not only the activation of surrounding benign stromal fibroblasts but also the induction of EMT, cell migration, invasion, and immune evasion of cancer cells [8, 40, 65, 66]. Collagen Type I+ myCAFs are involved in intrahepatic cholangiocarcinoma (ICC) cell growth via hyaluronan production [52]. Thus, myCAFs can be involved in tumor progression via humoral factors and mechanical interactions.

Regarding other functions of myCAFs, alpha‐smooth muscle actin (α‐SMA)‐positive and leucine‐rich repeat containing 15 (LRRC15)‐positive podoplanin (PDPN)‐positive CAFs induce therapeutic resistance of cancer cells against immune checkpoint inhibitors (ICIs), such as antiprogrammed death ligand 1 (PD‐L1) antibodies [41], and they suppress CD8‐T cell functions to support the immune evasion of cancer cells [42]. Similarly, the Anthrax Toxin Receptor 1 (ANTXR1)‐positive fibroblast activation protein (FAP)‐positive CAF subtype in high‐grade serous ovarian cancer is identified as an ECM‐producing myCAF, which is associated with chemotherapy resistance and suppression of CD8‐positive T‐cell activation in a YAP1‐dependent manner [36]. The myCAFs expressing integrin subunit alpha 11 (ITGA11) and PDGFRα promote lymphangiogenesis of endothelial cells and assist in the intravasation of bladder cancer cells to aid lymphatic metastasis [58]. Various subpopulations in the myCAF subtype might have shared and distinct tumor‐promoting functions.

In contrast to the tumor‐promoting functions, tumor‐suppressive functions of myCAFs have been found in the pancreatic ductal adenocarcinoma (PDAC) mouse model. Ganciclovir‐induced depletion of α‐SMA+ CAFs in mice that spontaneously develop PDAC unexpectedly results in poorly differentiated tumors with enhanced hypoxia and EMT and increased regulatory T cells to suppress immune surveillance [13]. An experimental system that deleted the Collagen Type I gene specifically from α‐SMA+ CAFs has also been reported by the same research team, and tumor progression and induction of immunosuppression have been shown even in this case [15]. This tumor‐promotive effect of collagen depletion from myCAFs can be due to the loss of the collagen‐based mechanical ECM barrier surrounding the cancer cells [50]. In the model of ICC [52], the presence or absence of Collagen Type I expression in myCAFs did not impact tumor growth, suggesting that the mechanical actions of myCAFs do not universally assist cancer progression. These mechanotransductive effects may differ depending on the cancer cell type, CAF origin, and the surrounding tissue environment. The genetic engineering loss of sonic hedgehog (Shh) has also been reported. Shh ligand and Hedgehog (Hh) signaling contribute to desmoplastic stroma formation in many solid tumors. Intriguingly, loss of Hh signaling decreases α‐SMA+ stromal cells, but it extensively accelerates tumor progression in the PDAC model [14]. In this PDAC model, the α‐SMA+ CAFs with heterogeneous expression of the Hh signaling‐targeted gene are also found, suggesting that myCAFs comprise finer subpopulations with conflicting functions in cancer progression. Considering that the features of myCAFs resemble those of activated fibroblast features, they can result from the contradictory wound‐healing action of benign‐resembling subtypes and the tumor‐supporting function of other subtypes, although this remains unclear.

6. Inflammatory CAFs (iCAFs)

The iCAFs are a subtype that produces cytokines and growth factors, thereby inducing an inflammatory microenvironment and supporting tumor progression (Figure 1 and Table 1). They were first defined in the PDAC model [33] and then have also been reported in various cancer types, including breast, gastric, and colorectal cancers. The iCAFs in PDAC arise from pancreatic stellate cells cocultured with cancer organoids. Functionally, they produce inflammatory cytokines such as interleukin (IL)‐6, IL‐11, and leukemia inhibitory factor (LIF) to sustain the survival of PDAC organoids [33]. In the ICC, iCAF‐producing hepatocyte growth factor (HGF) enhances ICC cell proliferation via the cancer‐expressing HGF receptor MET [52]. Similarly, a study using liver metastasis models of colorectal cancer and PDAC reported that HGF derived from iCAFs within metastatic sites contributes to metastatic tumor growth [50]. Interestingly, this study also suggested that myCAFs derived from hepatic stellate cells, also the origin of iCAFs, possess a tumor suppressor aspect by producing Collagen Type I. Thus, iCAF subtypes have distinct functions from myCAFs but can arise from a common cell type.

As for the other functional role of iCAFs, several reports show the immunosuppressive function of iCAFs. For instance, iCAF‐expressing IL‐1 receptors (IL1R1) have been reported in CMS4 colorectal cancer. IL1R1+ iCAFs can suppress CD8‐positive killer T cells and induce tumor‐promoting macrophages [51]. The iCAFs in skin cancers also have immunomodulatory functions and may contribute to the induction of T‐cell exhaustion [60]. These findings suggest that iCAFs induce tumor growth and antitumor immune effects as principal functions through humoral factors such as inflammatory cytokines. Although myCAFs also mediate tumor growth and immune surveillance, iCAFs are partially distinct from myCAF functions with ECM stiffness and mechanotransduction.

7. Antigen‐Presenting CAFs (apCAFs)

The apCAF is a subtype identified by single‐cell RNA‐seq analysis using pancreatic cancer tissues and expresses MHC Class II molecules involved in antigen presentation by immune cells (Figure 1 and Table 1). Functionally, it can act as a decoy receptor that differentiates naïve CD4‐positive T cells into regulatory T cells (Tregs) [31, 43]. However, unlike apCAFs in pancreatic cancer, a subpopulation of apCAFs in lung cancer promotes MHC Class II immunity and acts to suppress tumors [55]. As for apCAFs in pancreatic cancer, they can originate from mesothelial cells, and indeed, a monoclonal antibody to the mesothelial cell marker mesothelin reduced the abundance of apCAFs [43]. Recently, it has also been reported that apCAFs in nonsmall cell lung cancer are involved in bone metastasis of cancer cells, whereas iCAFs contribute to brain metastasis [56]. Whether cancer cells with bone and brain metastasis capabilities have converted CAFs to their properties remains unclear, but CAF subtypes may directly manage the organ tropism in tumor metastasis.

8. Other CAF Subtypes

Several reports have identified other CAF subtypes besides the CAF subtypes discussed above (Figure 1 and Table 1). In 2006, immunohistochemical analysis using mouse tumor tissues visualized CAF phenotypic heterogeneity [9]. This study showed a fibroblast‐specific protein‐1 (FSP1/S100A4)‐positive CAF subtype and other subpopulations expressing α‐SMA, PDGFRβ, and neuron–glial antigen 2 (NG2). This finding suggests CAF heterogeneity due to differences in marker protein expression and cell lineage of origin [9]. A study using a mouse PDAC model that specifically eliminates FAP‐positive cancer stromal cells from primary tumors showed that FAP‐positive CAFs suppress antitumor immune effects via CXC‐chemokine ligand 12 (CXCL12) production [44, 45]. CAF‐secreted CXCL12 is also closely associated with tumor angiogenesis [67, 68]. Related to this, CAF subtypes with angiogenesis‐related gene expressions, termed vascular‐CAFs (vCAFs), are found [39, 53]. As for other CAF subtypes, PDPN‐positive CAFs have been reported to increase EGFR‐TKI resistance in lung cancer cells [54]. However, these FAP+ or PDPN+ CAFs may be “subgroups” subdivisions of CAF subtypes such as myCAF and iCAF described above. For instance, five CAF markers, including FAP and PDPN, are used to identify the CAF‐S1 subtype, a key modulator in breast and ovarian cancer progression [34, 35, 49]. The subsequent single‐cell analysis showed that the FAP‐high CAF‐S1 subtype defined in breast cancer includes five myCAF and three iCAF subgroups [37]. Whether subpopulations of FAP+ or PDPN+ CAF subtypes are common to all solid tumors remains unclear. Further investigation, including other cellular lineage markers, would aid in defining precise CAF subtypes.

Normal fibroblasts are known to function in an inherently cancer‐suppressive manner [69, 70]. Interestingly, such “steady‐state” normal fibroblast subtypes with peptidase inhibitor 16 (Pi16)‐positive and dermatopontin (Dpt)‐positive expressions are found in both healthy and diseased tissues [25]. Experimental depletion of LRRC15‐positive myCAFs has been reported to increase the number of Pi16+ subtypes within tumor tissues [42]. Moreover, synthetic retinoids may enhance the response to immune checkpoint inhibitors by artificially inducing the cancer‐suppressive Meflin+ CAF subtype [46, 47, 48]. Meflin is a marker of mesenchymal stem cells (MSCs) and fibroblasts in normal tissues [71]. These findings suggest that direct targeting of cancer‐promoting CAF subtypes induces tumor‐suppressing CAF subtypes within the TME. However, the CAF subtype with a secretome pattern similar to normal fibroblasts is also found in oral squamous cell carcinoma. It can promote cancer cell invasion in a hyaluronic acid‐dependent manner [72]. Therefore, not all “steady‐state” fibroblast subtypes likely behave as tumor suppressors. Determining whether they promote or suppress cancer may be complicated by subtyping on simple transcriptome and proteome profiling.

9. Why Do CAF Subtypes Arise?

9.1. Cellular Diversity and Cancer‐Derived Signals Determining CAF Subtypes

As discussed above, various factors are involved in the creation of CAF subtypes within the TME. One of these factors is the cellular lineage‐dependent mechanism. Although resident fibroblasts, stellate cells, or astrocytes within specific organs are the primary origin of CAFs, various cell lineages have been proposed as the origin of CAFs, including adipocytes, bone marrow‐derived mesenchymal stem cells (BM‐MSCs), vascular endothelial pericytes, epithelial cells, and mesothelial cells (cell lineage‐dependent, Figure 2) [6, 26]. For instance, a study with an in vivo model of gastric cancer (GC) showed that BM‐MSCs are recruited in dysplastic stomach and subsequently converted into CAFs [73]. Consistent with this study, BM‐MSC‐derived CAFs are responsible for cell proliferation and angiogenesis in the lung metastasis model of breast cancer. However, PDGFRα‐positive CAFs in metastatic foci are derived from resident fibroblasts and express Col1a, Acta2, and extracellular matrix‐related genes [74]. Mesothelial cells and pericytes can be the origin of apCAFs and vCAFs, respectively [39, 43, 53]. These findings show that cellular lineage‐dependent mechanisms can shape part of the functional and phenotypic heterogeneity of CAFs.

FIGURE 2.

FIGURE 2

How do cancer cells produce distinct CAF subtypes? Various factors are involved in CAF subtype creation within the tumor microenvironment (TME). One is the cellular lineage‐dependent mechanism. Many origin cells can convert into CAFs, so multiple features emerge as CAF subtypes. Second is the environmental signaling‐dependent mechanism. A single‐cell lineage can differentiate into different CAF subtypes via various signaling pathways within the TME. Interconversion between CAF subtypes is also found. Third is the epigenetic regulation‐dependent mechanism. Methylation patterns of histones are associated with CAF subtype marker gene coding regions, suggesting that epigenetic regulation also contributes to CAF heterogeneity.

As other factors involved in the creation of CAF subtypes, the features of cancer cells, such as genetic background and metastatic properties, also affect CAF subtype composition (environmental signaling‐dependent, Figure 2). Differential p53 mutation status induces different CAF phenotypes to create a premetastatic and chemoresistant microenvironment [75]. We also demonstrated that highly metastatic GC cells educate stromal fibroblast phenotypes in transcriptome and metabolomic levels [29, 76]. Thus, both intrinsic properties of cancer cells and intercellular signaling from the TME contribute to establishing CAF heterogeneity.

Besides the examples for creating CAF subtypes described above, inter‐subtype conversions of CAF subtypes have also been reported (environmental‐signaling dependent, Figure 2). In the PDAC model, the induction of myCAFs depends on TGF‐β signaling, but the iCAFs are determined by IL‐1B‐inducing NF‐κB/STAT3 signaling. A model of the antagonistic relationship of these signals that regulate the CAF subtype has been proposed [32, 33]. TGF‐β signaling and NF‐κB are well known as antagonists, suggesting that the balanced relationship between these signaling pathways creates CAF heterogeneity. Indeed, the induction of apCAFs from mesothelial cells in cancer tissue requires both TGF‐β and IL‐1α signals. Thus, apCAFs might have the properties of both myCAFs and iCAFs and represent an intermediate state in which both signals are received. Related to these findings, dipeptidyl peptidase‐4 (DPP4) and YAP1‐dependent signaling also mediate the conversion of iCAF subgroups, Detox‐iCAFs, to a myCAF subpopulation, ECM‐myCAFs [38]. The DPP4‐mediated pathway directly converts Detox‐iCAFs into ECM‐myCAFs, but the YAP1‐mediated pathway appears to induce other intermediate myCAF subgroups resembling features of activated fibroblasts during wound healing. TGF‐β signaling via transforming growth factor‐beta receptor 2 (TGFBR2) can also induce ECM‐myCAFs from Detox‐iCAFs [38]. Highly complex intercellular signaling may organize stromal heterogeneity in solid tumors.

Epigenetic changes have been proposed as the other way for CAF subtype regulation to be independent of cell‐derived signals and fibroblast lineages (epigenetic regulation‐dependent, Figure 2). Single‐cell resolution ATAC‐seq analysis shows that open chromatin status correlates with some CAF subtype‐related gene loci, suggesting that chromatin accessibility affects transcriptomic features and may determine some CAF subtypes [77]. Therefore, epigenetic changes within the tumor background tissue may mediate CAF subtype regulation. Further mechanistic analysis is needed to clarify CAF subtype creation.

9.2. Contribution of Extracellular Vesicles in CAF Subtype Induction

EVs are lipid bilayer membrane vesicles that contain various bioactive molecules, such as mRNAs, microRNAs (miRNAs), and proteins. These molecules in EVs are transferred to the recipient cells within the proximal tissues and distal organs and mediate numerous cellular phenotypes. In particular, many cancer cells crosstalk with stromal cells through EVs to regulate tumor initiation, growth, invasion, metastasis, and treatment resistance. In addition, EVs are detectable in various body fluids, such as blood, saliva, and urine, besides their striking functions in intercellular communications [78, 79]. The composition of these molecules in EVs varies according to the gene expression profiles of their origin cells. Thus, EVs are attractive targets as an avenue to improve cancer diagnosis if they package tumor‐specific molecules.

EVs include various subtypes with different components, sizes, and biogenesis processes. For example, exosomes (100–200 nm) originate from the exocytosis of multivesicular bodies (MVBs) [79, 80]. Ectosomes (150–1000 nm) are generated directly from the plasma membrane [80]. Apoptotic bodies (100–5000 nm) arise during programmed cell death [80]. However, the expert consensus in EV research encourages using the nomenclature “EVs” unless subcellular origin can be demonstrated. This review will use the term “EVs” for all subtypes of membrane vesicles in the extracellular space to avoid misleading readers.

So far, many researchers have demonstrated that cancer‐derived EVs convert different stromal cell types, including fibroblasts, macrophages, and mesenchymal stem cells, into CAFs [79, 81]. However, most of these reports regarding the induction of CAFs via EVs remain to clarify whether cancer EVs can create heterogeneous subtypes of CAFs. Given that cancer‐derived signals can reprogram a single‐cell lineage into two types of CAF subtypes [32, 33], it is highly plausible that cancer EVs are also responsible for CAF subtype creation. Indeed, our group found that gastric cancer (GC) cell‐secreting EVs induce chemokine expressions such as IL‐1 and IL‐8 in stomach fibroblasts, similar to the property of iCAFs [29]. As mechanisms, these GC‐derived EVs transport a series of miRNAs, including miR‐193b, into stomach fibroblasts to induce chemokine expressions. These GC cell‐derived EVs cannot regulate the expression of α‐SMA and collagen, which are part of the myCAF subtypes. Importantly, high‐metastatic GC cells are inducible in both α‐SMA and chemokine expressions in fibroblasts compared with low‐metastatic GC cells. These findings suggest that cancer‐derived EVs selectively confer iCAF‐like features in fibroblasts. IL‐8‐expressing CAFs within tumor stroma are closely associated with poor outcomes in patients with GC, suggesting that GC generates a microenvironment favorable for their progression. Consistent with our findings, other research groups also provided evidence regarding CAF subtype creation via cancer‐derived EVs. Bladder cancer cell‐derived EVs convert the primary bladder fibroblasts into iCAF features [59]. Compared with TGF‐β treatment, these cancer‐derived EVs have a lesser effect on the induction of α‐SMA in fibroblasts [59]. EV‐induced iCAFs express IL‐6 and activate the STAT3 signaling pathway that leads to the EMT in bladder cancer [59]. In addition, metastatic melanoma cell‐derived EVs activate proinflammatory signaling in the fibroblasts of metastatic niches [61]. These EVs contain RNAs capable of instigating inflammatory signaling in the fibroblasts, such as high‐mobility group box 1 (Hmgb1), thymic stromal lymphopoietin (Tslp), and interferon regulatory factor 1 (Irf1) [61]. Interestingly, EVs derived from lung adenocarcinoma cells induce ECM‐myCAF‐like features but decreased iCAF‐related gene expressions in lung fibroblasts [57]. These findings suggest that cancer cells may confer favorable features to surrounding stroma via EVs and change the CAF subtype balances within the TME (Figure 3).

FIGURE 3.

FIGURE 3

Impact of extracellular vesicles on CAF subtype construction. Extracellular vesicles (EVs) also contribute to CAF subtype construction. The composition of these molecules in EVs varies according to the gene expression profiles of their origin cells. Thus, the impact of cancer EVs on CAF subtypes also apparently depends on cancer cell features. EVs derived from GC and bladder cancer can induce iCAF subtypes through transferring miRNAs and lncRNAs. In contrast, lung cancer‐derived EVs have a capacity for myCAF induction, but not iCAF induction. Cancer cells may induce CAF subtypes that have adapted to their features via EVs.

10. Conclusion

Accumulating evidence has clarified the nature of CAFs and their function in tumor progression. However, many aspects of CAF biology, in particular, how CAF heterogeneity is created and how they mediate tumor progression and therapeutic resistance remain unclear. In addition, many CAF subtype classifications have emerged from single‐cell analyses and are likely not yet well organized. The classification of subtype markers and their precise function in tumor progression will need to be addressed in future studies. The fact that CAFs have both cancer‐promotive and inhibitory subtypes has led to the key concept that the balance between them determines the role of CAFs in the TME [46, 48]. However, given that different cell lineages and subtypes have been reported for each organ and tissue of origin, we believe a systematic understanding of CAF subtypes in all cancer types is required to prove this concept. Understanding the nature of the CAF subtypes and their function may facilitate the development of new therapeutic modalities and diagnostic methods by leveraging the profile of CAF subtypes.

Author Contributions

Yutaka Naito: conceptualization, funding acquisition, project administration, visualization, writing – review and editing.

Ethics Statement

The author has nothing to report.

Consent

The author has nothing to report.

Conflicts of Interest

The author declares no conflicts of interest.

Acknowledgments

The author would like to thank all members of the Department of Bioregulation, Nippon Medical School, for their support.

Funding: This work was supported by the JSPS KAKENHI Grant Number 22K15567 (Y.N.).

References

  • 1. Kalluri R. and Zeisberg M., “Fibroblasts in Cancer,” Nature Reviews. Cancer 6 (2006): 392–401. [DOI] [PubMed] [Google Scholar]
  • 2. Hanahan D. and Weinberg R. A., “Hallmarks of Cancer: The Next Generation,” Cell 144 (2011): 646–674. [DOI] [PubMed] [Google Scholar]
  • 3. Vinay D. S., Ryan E. P., Pawelec G., et al., “Immune Evasion in Cancer: Mechanistic Basis and Therapeutic Strategies,” Seminars in Cancer Biology 35, no. Suppl (2015): S185–S198. [DOI] [PubMed] [Google Scholar]
  • 4. Ribas A., “Adaptive Immune Resistance: How Cancer Protects From Immune Attack,” Cancer Discovery 5 (2015): 915–919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Tsujita K., Satow R., Asada S., et al., “Homeostatic Membrane Tension Constrains Cancer Cell Dissemination by Counteracting BAR Protein Assembly,” Nature Communications 12 (2021): 5930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Joyce J. A. and Pollard J. W., “Microenvironmental Regulation of Metastasis,” Nature Reviews. Cancer 9 (2009): 239–252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Kalluri R., “The Biology and Function of Fibroblasts in Cancer,” Nature Reviews. Cancer 16 (2016): 582–598. [DOI] [PubMed] [Google Scholar]
  • 8. Sahai E., Astsaturov I., Cukierman E., et al., “A Framework for Advancing Our Understanding of Cancer‐Associated Fibroblasts,” Nature Reviews. Cancer 20 (2020): 174–186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Sugimoto H., Mundel T. M., Kieran M. W., and Kalluri R., “Identification of Fibroblast Heterogeneity in the Tumor Microenvironment,” Cancer Biology & Therapy 5 (2006): 1640–1646. [DOI] [PubMed] [Google Scholar]
  • 10. Herrera M., Islam A. B., Herrera A., et al., “Functional Heterogeneity of Cancer‐Associated Fibroblasts From Human Colon Tumors Shows Specific Prognostic Gene Expression Signature,” Clinical Cancer Research 19 (2013): 5914–5926. [DOI] [PubMed] [Google Scholar]
  • 11. Franco O. E., Jiang M., Strand D. W., et al., “Altered TGF‐Beta Signaling in a Subpopulation of Human Stromal Cells Promotes Prostatic Carcinogenesis,” Cancer Research 71 (2011): 1272–1281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Prakash J., “Cancer‐Associated Fibroblasts: Perspectives in Cancer Therapy,” Trends Cancer 2 (2016): 277–279. [DOI] [PubMed] [Google Scholar]
  • 13. Ozdemir B. C., Pentcheva‐Hoang T., Carstens J. L., et al., “Depletion of Carcinoma‐Associated Fibroblasts and Fibrosis Induces Immunosuppression and Accelerates Pancreas Cancer With Reduced Survival,” Cancer Cell 25 (2014): 719–734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Rhim A. D., Oberstein P. E., Thomas D. H., et al., “Stromal Elements Act to Restrain, Rather Than Support, Pancreatic Ductal Adenocarcinoma,” Cancer Cell 25 (2014): 735–747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Chen Y., Kim J., Yang S., et al., “Type I Collagen Deletion in alphaSMA(+) Myofibroblasts Augments Immune Suppression and Accelerates Progression of Pancreatic Cancer,” Cancer Cell 39 (2021): 548–565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Plikus M. V., Wang X., Sinha S., et al., “Fibroblasts: Origins, Definitions, and Functions in Health and Disease,” Cell 184 (2021): 3852–3872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Ushakumary M. G., Riccetti M., and Perl A. T., “Resident Interstitial Lung Fibroblasts and Their Role in Alveolar Stem Cell Niche Development, Homeostasis, Injury, and Regeneration,” Stem Cells Translational Medicine 10 (2021): 1021–1032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Rippa A. L., Alpeeva E. V., Vasiliev A. V., and Vorotelyak E. A., “Alveologenesis: What Governs Secondary Septa Formation,” International Journal of Molecular Sciences 22 (2021): 12107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Moretti L., Stalfort J., Barker T. H., and Abebayehu D., “The Interplay of Fibroblasts, the Extracellular Matrix, and Inflammation in Scar Formation,” Journal of Biological Chemistry 298 (2022): 101530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Apte M. V., Pirola R. C., and Wilson J. S., “Pancreatic Stellate Cells: A Starring Role in Normal and Diseased Pancreas,” Frontiers in Physiology 3 (2012): 344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Sherman M. H., Yu R. T., Engle D. D., et al., “Vitamin D Receptor‐Mediated Stromal Reprogramming Suppresses Pancreatitis and Enhances Pancreatic Cancer Therapy,” Cell 159 (2014): 80–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Beilfuss A., Sowa J. P., Sydor S., et al., “Vitamin D Counteracts Fibrogenic TGF‐Beta Signalling in Human Hepatic Stellate Cells Both Receptor‐Dependently and Independently,” Gut 64 (2015): 791–799. [DOI] [PubMed] [Google Scholar]
  • 23. Duran A., Hernandez E. D., Reina‐Campos M., et al., “p62/SQSTM1 by Binding to Vitamin D Receptor Inhibits Hepatic Stellate Cell Activity, Fibrosis, and Liver Cancer,” Cancer Cell 30 (2016): 595–609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. McCarthy N., Manieri E., Storm E. E., et al., “Distinct Mesenchymal Cell Populations Generate the Essential Intestinal BMP Signaling Gradient,” Cell Stem Cell 26 (2020): 391–402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Buechler M. B., Pradhan R. N., Krishnamurty A. T., et al., “Cross‐Tissue Organization of the Fibroblast Lineage,” Nature 593 (2021): 575–579. [DOI] [PubMed] [Google Scholar]
  • 26. Park D., Sahai E., and Rullan A., “SnapShot: Cancer‐Associated Fibroblasts,” Cell 181 (2020): e481. [DOI] [PubMed] [Google Scholar]
  • 27. Dvorak H. F., “Tumors: Wounds That Do Not Heal. Similarities Between Tumor Stroma Generation and Wound Healing,” New England Journal of Medicine 315 (1986): 1650–1659. [DOI] [PubMed] [Google Scholar]
  • 28. Lambrechts D., Wauters E., Boeckx B., et al., “Phenotype Molding of Stromal Cells in the Lung Tumor Microenvironment,” Nature Medicine 24 (2018): 1277–1289. [DOI] [PubMed] [Google Scholar]
  • 29. Naito Y., Yamamoto Y., Sakamoto N., et al., “Cancer Extracellular Vesicles Contribute to Stromal Heterogeneity by Inducing Chemokines in Cancer‐Associated Fibroblasts,” Oncogene 38 (2019): 5566–5579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Arwert E. N., Milford E. L., Rullan A., et al., “STING and IRF3 in Stromal Fibroblasts Enable Sensing of Genomic Stress in Cancer Cells to Undermine Oncolytic Viral Therapy,” Nature Cell Biology 22 (2020): 758–766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Elyada E., Bolisetty M., Laise P., et al., “Cross‐Species Single‐Cell Analysis of Pancreatic Ductal Adenocarcinoma Reveals Antigen‐Presenting Cancer‐Associated Fibroblasts,” Cancer Discovery 9 (2019): 1102–1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Biffi G., Oni T. E., Spielman B., et al., “IL1‐Induced JAK/STAT Signaling Is Antagonized by TGFbeta to Shape CAF Heterogeneity in Pancreatic Ductal Adenocarcinoma,” Cancer Discovery 9 (2019): 282–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Ohlund D., Handly‐Santana A., Biffi G., et al., “Distinct Populations of Inflammatory Fibroblasts and Myofibroblasts in Pancreatic Cancer,” Journal of Experimental Medicine 214 (2017): 579–596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Costa A., Kieffer Y., Scholer‐Dahirel A., et al., “Fibroblast Heterogeneity and Immunosuppressive Environment in Human Breast Cancer,” Cancer Cell 33 (2018): 463–479. [DOI] [PubMed] [Google Scholar]
  • 35. Givel A. M., Kieffer Y., Scholer‐Dahirel A., et al., “miR200‐Regulated CXCL12beta Promotes Fibroblast Heterogeneity and Immunosuppression in Ovarian Cancers,” Nature Communications 9 (2018): 1056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Licaj M., Mhaidly R., Kieffer Y., et al., “Residual ANTXR1+ Myofibroblasts After Chemotherapy Inhibit Anti‐Tumor Immunity via YAP1 Signaling Pathway,” Nature Communications 15 (2024): 1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Kieffer Y., Hocine H. R., Gentric G., et al., “Single‐Cell Analysis Reveals Fibroblast Clusters Linked to Immunotherapy Resistance in Cancer,” Cancer Discovery 10 (2020): 1330–1351. [DOI] [PubMed] [Google Scholar]
  • 38. Croizer H., Mhaidly R., Kieffer Y., et al., “Deciphering the Spatial Landscape and Plasticity of Immunosuppressive Fibroblasts in Breast Cancer,” Nature Communications 15 (2024): 2806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Bartoschek M., Oskolkov N., Bocci M., et al., “Spatially and Functionally Distinct Subclasses of Breast Cancer‐Associated Fibroblasts Revealed by Single Cell RNA Sequencing,” Nature Communications 9 (2018): 5150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Mucciolo G., Araos Henriquez J., Jihad M., et al., “EGFR‐Activated Myofibroblasts Promote Metastasis of Pancreatic Cancer,” Cancer Cell 42 (2024): 101–118.e11. [DOI] [PubMed] [Google Scholar]
  • 41. Dominguez C. X., Muller S., Keerthivasan S., et al., “Single‐Cell RNA Sequencing Reveals Stromal Evolution Into LRRC15(+) Myofibroblasts as a Determinant of Patient Response to Cancer Immunotherapy,” Cancer Discovery 10 (2020): 232–253. [DOI] [PubMed] [Google Scholar]
  • 42. Krishnamurty A. T., Shyer J. A., Thai M., et al., “LRRC15(+) Myofibroblasts Dictate the Stromal Setpoint to Suppress Tumour Immunity,” Nature 611 (2022): 148–154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Huang H., Wang Z., Zhang Y., et al., “Mesothelial Cell‐Derived Antigen‐Presenting Cancer‐Associated Fibroblasts Induce Expansion of Regulatory T Cells in Pancreatic Cancer,” Cancer Cell 40 (2022): 656–673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Kraman M., Bambrough P. J., Arnold J. N., et al., “Suppression of Antitumor Immunity by Stromal Cells Expressing Fibroblast Activation Protein‐Alpha,” Science 330 (2010): 827–830. [DOI] [PubMed] [Google Scholar]
  • 45. Feig C., Jones J. O., Kraman M., et al., “Targeting CXCL12 From FAP‐Expressing Carcinoma‐Associated Fibroblasts Synergizes With Anti‐PD‐L1 Immunotherapy in Pancreatic Cancer,” Proceedings of the National Academy of Sciences of the United States of America 110 (2013): 20212–20217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Mizutani Y., Kobayashi H., Iida T., et al., “Meflin‐Positive Cancer‐Associated Fibroblasts Inhibit Pancreatic Carcinogenesis,” Cancer Research 79 (2019): 5367–5381. [DOI] [PubMed] [Google Scholar]
  • 47. Owaki T., Iida T., Miyai Y., et al., “Synthetic Retinoid‐Mediated Preconditioning of Cancer‐Associated Fibroblasts and Macrophages Improves Cancer Response to Immune Checkpoint Blockade,” British Journal of Cancer 131 (2024): 372–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Iida T., Mizutani Y., Esaki N., et al., “Pharmacologic Conversion of Cancer‐Associated Fibroblasts From a Protumor Phenotype to an Antitumor Phenotype Improves the Sensitivity of Pancreatic Cancer to Chemotherapeutics,” Oncogene 41 (2022): 2764–2777. [DOI] [PubMed] [Google Scholar]
  • 49. Pelon F., Bourachot B., Kieffer Y., et al., “Cancer‐Associated Fibroblast Heterogeneity in Axillary Lymph Nodes Drives Metastases in Breast Cancer Through Complementary Mechanisms,” Nature Communications 11 (2020): 404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Bhattacharjee S., Hamberger F., Ravichandra A., et al., “Tumor Restriction by Type I Collagen Opposes Tumor‐Promoting Effects of Cancer‐Associated Fibroblasts,” Journal of Clinical Investigation 131 (2021): e146987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Koncina E., Nurmik M., Pozdeev V. I., et al., “IL1R1(+) Cancer‐Associated Fibroblasts Drive Tumor Development and Immunosuppression in Colorectal Cancer,” Nature Communications 14 (2023): 4251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Affo S., Nair A., Brundu F., et al., “Promotion of Cholangiocarcinoma Growth by Diverse Cancer‐Associated Fibroblast Subpopulations,” Cancer Cell 39 (2021): 866–882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Zhang M., Yang H., Wan L., et al., “Single‐Cell Transcriptomic Architecture and Intercellular Crosstalk of Human Intrahepatic Cholangiocarcinoma,” Journal of Hepatology 73 (2020): 1118–1130. [DOI] [PubMed] [Google Scholar]
  • 54. Yoshida T., Ishii G., Goto K., et al., “Podoplanin‐Positive Cancer‐Associated Fibroblasts in the Tumor Microenvironment Induce Primary Resistance to EGFR‐TKIs in Lung Adenocarcinoma With EGFR Mutation,” Clinical Cancer Research 21 (2015): 642–651. [DOI] [PubMed] [Google Scholar]
  • 55. Kerdidani D., Aerakis E., Verrou K. M., et al., “Lung Tumor MHCII Immunity Depends on In Situ Antigen Presentation by Fibroblasts,” Journal of Experimental Medicine 219 (2022): e20210815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Xu K., Wang H., Zou Y. X., et al., “Distinct Fibroblast Subpopulations Associated With Bone, Brain or Intrapulmonary Metastasis in Advanced Non‐Small‐Cell Lung Cancer,” Clinical and Translational Medicine 14 (2024): e1605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Trejo Vazquez J. A., Towle R., Farnsworth D. A., Sarafan M., Lockwood W. W., and Garnis C., “Extracellular Vesicles From Lung Adenocarcinoma Cells Induce Activation of Different Cancer‐Associated Fibroblast Subtypes,” Biomedicine 12 (2024): 2523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Zheng H., An M., Luo Y., et al., “PDGFRalpha(+)ITGA11(+) Fibroblasts Foster Early‐Stage Cancer Lymphovascular Invasion and Lymphatic Metastasis via ITGA11‐SELE Interplay,” Cancer Cell 42 (2024): 682–700. [DOI] [PubMed] [Google Scholar]
  • 59. Goulet C. R., Champagne A., Bernard G., et al., “Cancer‐Associated Fibroblasts Induce Epithelial‐Mesenchymal Transition of Bladder Cancer Cells Through Paracrine IL‐6 Signalling,” BMC Cancer 19 (2019): 137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Forsthuber A., Aschenbrenner B., Korosec A., et al., “Cancer‐Associated Fibroblast Subtypes Modulate the Tumor‐Immune Microenvironment and Are Associated With Skin Cancer Malignancy,” Nature Communications 15 (2024): 9678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Gener Lahav T., Adler O., Zait Y., et al., “Melanoma‐Derived Extracellular Vesicles Instigate Proinflammatory Signaling in the Metastatic Microenvironment,” International Journal of Cancer 145 (2019): 2521–2534. [DOI] [PubMed] [Google Scholar]
  • 62. Caligiuri G. and Tuveson D. A., “Activated Fibroblasts in Cancer: Perspectives and Challenges,” Cancer Cell 41 (2023): 434–449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Lavie D., Ben‐Shmuel A., Erez N., and Scherz‐Shouval R., “Cancer‐Associated Fibroblasts in the Single‐Cell Era,” Nature Cancer 3 (2022): 793–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Panciera T., Azzolin L., Cordenonsi M., and Piccolo S., “Mechanobiology of YAP and TAZ in Physiology and Disease,” Nature Reviews. Molecular Cell Biology 18 (2017): 758–770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Matsumura Y., Ito Y., Mezawa Y., et al., “Stromal Fibroblasts Induce Metastatic Tumor Cell Clusters via Epithelial‐Mesenchymal Plasticity,” Life Science Alliance 2 (2019): e201900425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Flavell R. A., Sanjabi S., Wrzesinski S. H., and Licona‐Limon P., “The Polarization of Immune Cells in the Tumour Environment by TGFbeta,” Nature Reviews. Immunology 10 (2010): 554–567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Orimo A., Gupta P. B., Sgroi D. C., et al., “Stromal Fibroblasts Present in Invasive Human Breast Carcinomas Promote Tumor Growth and Angiogenesis Through Elevated SDF‐1/CXCL12 Secretion,” Cell 121 (2005): 335–348. [DOI] [PubMed] [Google Scholar]
  • 68. Yang Y., Li J., Lei W., et al., “CXCL12‐CXCR4/CXCR7 Axis in Cancer: From Mechanisms to Clinical Applications,” International Journal of Biological Sciences 19 (2023): 3341–3359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Stoker M. G., Shearer M., and O'Neill C., “Growth Inhibition of Polyoma‐Transformed Cells by Contact With Static Normal Fibroblasts,” Journal of Cell Science 1 (1966): 297–310. [DOI] [PubMed] [Google Scholar]
  • 70. Dotto G. P., Weinberg R. A., and Ariza A., “Malignant Transformation of Mouse Primary Keratinocytes by Harvey Sarcoma Virus and Its Modulation by Surrounding Normal Cells,” Proceedings of the National Academy of Sciences of the United States of America 85 (1988): 6389–6393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Maeda K., Enomoto A., Hara A., et al., “Identification of Meflin as a Potential Marker for Mesenchymal Stromal Cells,” Scientific Reports 6 (2016): 22288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Costea D. E., Hills A., Osman A. H., et al., “Identification of Two Distinct Carcinoma‐Associated Fibroblast Subtypes With Differential Tumor‐Promoting Abilities in Oral Squamous Cell Carcinoma,” Cancer Research 73 (2013): 3888–3901. [DOI] [PubMed] [Google Scholar]
  • 73. Quante M., Tu S. P., Tomita H., et al., “Bone Marrow‐Derived Myofibroblasts Contribute to the Mesenchymal Stem Cell Niche and Promote Tumor Growth,” Cancer Cell 19 (2011): 257–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Raz Y., Cohen N., Shani O., et al., “Bone Marrow‐Derived Fibroblasts Are a Functionally Distinct Stromal Cell Population in Breast Cancer,” Journal of Experimental Medicine 215 (2018): 3075–3093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Vennin C., Melenec P., Rouet R., et al., “CAF Hierarchy Driven by Pancreatic Cancer Cell p53‐Status Creates a Pro‐Metastatic and Chemoresistant Environment via Perlecan,” Nature Communications 10 (2019): 3637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Kogure A., Naito Y., Yamamoto Y., et al., “Cancer Cells With High‐Metastatic Potential Promote a Glycolytic Shift in Activated Fibroblasts,” PLoS One 15 (2020): e0234613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Foster D. S., Januszyk M., Delitto D., et al., “Multiomic Analysis Reveals Conservation of Cancer‐Associated Fibroblast Phenotypes Across Species and Tissue of Origin,” Cancer Cell 40 (2022): 1392–1406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Urabe F., Kosaka N., Ito K., Kimura T., Egawa S., and Ochiya T., “Extracellular Vesicles as Biomarkers and Therapeutic Targets for Cancer,” American Journal of Physiology. Cell Physiology 318 (2020): C29–C39. [DOI] [PubMed] [Google Scholar]
  • 79. Naito Y., Yoshioka Y., and Ochiya T., “Intercellular Crosstalk Between Cancer Cells and Cancer‐Associated Fibroblasts via Extracellular Vesicles,” Cancer Cell International 22 (2022): 367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Welsh J. A., Goberdhan D. C. I., O'Driscoll L., et al., “Minimal Information for Studies of Extracellular Vesicles (MISEV2023): From Basic to Advanced Approaches,” Journal of Extracellular Vesicles 13 (2024): e12404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Sohal I. S. and Kasinski A. L., “Emerging Diversity in Extracellular Vesicles and Their Roles in Cancer,” Frontiers in Oncology 13 (2023): 1167717. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cancer Science are provided here courtesy of Wiley

RESOURCES