ArticlePDF Available

Architecture of Cancer-Associated Fibroblasts in Tumor Microenvironment: Mapping Their Origins, Heterogeneity, and Role in Cancer Therapy Resistance

Authors:

Abstract and Figures

The tumor stroma, a key component of the tumor microenvironment (TME), is a key determinant of response and resistance to cancer treatment. The stromal cells, extracellular matrix (ECM), and blood vessels influence cancer cell response to therapy and play key roles in tumor relapse and therapeutic outcomes. Of the stromal cells present in the TME, much attention has been given to cancer-associated fibroblasts (CAFs) as they are the most abundant and important in cancer initiation, progression, and therapy resistance. Besides releasing several factors, CAFs also synthesize the ECM, a key component of the tumor stroma. In this expert review, we examine the role of CAFs in the regulation of tumor cell behavior and reveal how CAF-derived factors and signaling influence tumor cell heterogeneity and development of novel strategies to combat cancer. Importantly, CAFs display both phenotypic and functional heterogeneity, with significant ramifications on CAF-directed therapies. Principal anti-cancer therapies targeting CAFs take the form of: (1) CAFs' ablation through use of immunotherapies, (2) re-education of CAFs to normalize the cells, (3) cellular therapies involving CAFs delivering drugs such as oncolytic adenoviruses, and (4) stromal depletion via targeting the ECM and its related signaling. The CAFs' heterogeneity could be a result of different cellular origins and the cancer-specific tumor microenvironmental effects, underscoring the need for further multiomics and biochemical studies on CAFs and the subsets. Lastly, we present recent advances in therapeutic targeting of CAFs and the success of such endeavors or their lack thereof. We recommend that to advance global public health and personalized medicine, treatments in the oncology clinic should be combinatorial in nature, strategically targeting both cancer cells and stromal cells, and their interactions.
Content may be subject to copyright.
Review Article
Architecture of Cancer-Associated Fibroblasts
in Tumor Microenvironment:
Mapping Their Origins, Heterogeneity,
and Role in Cancer Therapy Resistance
Kevin Dzobo
1,2
and Collet Dandara
3
Abstract
The tumor stroma, a key component of the tumor microenvironment (TME), is a key determinant of response
and resistance to cancer treatment. The stromal cells, extracellular matrix (ECM), and blood vessels influence
cancer cell response to therapy and play key roles in tumor relapse and therapeutic outcomes. Of the stromal
cells present in the TME, much attention has been given to cancer-associated fibroblasts (CAFs) as they are the
most abundant and important in cancer initiation, progression, and therapy resistance. Besides releasing several
factors, CAFs also synthesize the ECM, a key component of the tumor stroma. In this expert review, we
examine the role of CAFs in the regulation of tumor cell behavior and reveal how CAF-derived factors and
signaling influence tumor cell heterogeneity and development of novel strategies to combat cancer. Importantly,
CAFs display both phenotypic and functional heterogeneity, with significant ramifications on CAF-directed
therapies. Principal anti-cancer therapies targeting CAFs take the form of: (1) CAFs’ ablation through use of
immunotherapies, (2) re-education of CAFs to normalize the cells, (3) cellular therapies involving CAFs
delivering drugs such as oncolytic adenoviruses, and (4) stromal depletion via targeting the ECM and its related
signaling. The CAFs’ heterogeneity could be a result of different cellular origins and the cancer-specific tumor
microenvironmental effects, underscoring the need for further multiomics and biochemical studies on CAFs and
the subsets. Lastly, we present recent advances in therapeutic targeting of CAFs and the success of such
endeavors or their lack thereof. We recommend that to advance global public health and personalized medicine,
treatments in the oncology clinic should be combinatorial in nature, strategically targeting both cancer cells and
stromal cells, and their interactions.
Keywords: cancer biology, tumor microenvironment, tumor stroma, cancer-associated fibroblasts, biomarkers,
gene expression, personalized medicine
Introduction
Recent cancer incidence and mortality statistics
indicate an always increasing burden of cancer in the
coming years, with most new cases occurring in countries of
low and middle income (Bray et al., 2018; Ferlay et al., 2019;
Fidler and Bray, 2018). Based on GLOBOCAN estimates and
others, there is a need for a new drive to find and develop new
strategies to reduce this burden worldwide ( Bray, 2016; Bray
and Soerjomataram, 2015; Bray et al., 2012, 2018; Ferlay et al.,
2019; Fidler et al., 2016). The cancer incidence and mortality
statistics provided are likely lower than the actual figures given
the limited and strained surveillance systems found in many
low- and medium-income countries (Bray et al., 2018; Ferlay
et al., 2019; Znaor et al., 2018). It is important to note that
progress has been made in raising awareness to cancer and
several cancer prevention strategies are being adopted; how-
ever, the picture still looks gloomy going forward.
Although great success has been achieved in treating
cancer via different therapeutic strategies directed at cancer
1
International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa.
2
Division of Medical Biochemistry, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious
Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.
3
Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Disease and Molecular
Medicine, University of Cape Town, Cape Town, South Africa.
OMICS A Journal of Integrative Biology
Volume 24, Number 6, 2020
ªMary Ann Liebert, Inc.
DOI: 10.1089/omi.2020.0023
314
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
cells, reports show that cancer deaths are likely to increase
globally (Bray and Soerjomataram, 2015; Bray et al., 2018;
Ferlay et al., 2019; Fidler et al., 2016; Moten et al., 2014).
Significant progress made in understanding the underlying
causes and molecular mechanisms involved in tumor initia-
tion and development has led to better patients’ outcomes
(Lesina et al., 2011; Mongiat et al., 2016; Senthebane et al.,
2017, 2018). Two major contributors to cancer deaths still
requiring better understanding are cancer relapse and me-
tastasis (Crunkhorn, 2018; Li et al., 2015; Yates et al., 2017).
Research into tumor relapse and metastasis is ongoing, with
several studies reporting novel drugs and effective strategies
aimed at limiting these processes (Mitra et al., 2015; Zhou
et al., 2009).
Further, studies have revealed the tumor microenviron-
ment (TME) as key to tumor initiation, response to therapy,
relapse, and metastasis, ultimately influencing patients’
management in the clinic (Dzobo et al., 2016a; Kalluri, 2016;
Senthebane et al., 2017, 2018). Specifically, the TME has
been shown to provide some form of protection to cancer
cells, reduce cancer cell response to therapy, and, ultimately,
promote therapy resistance (Quail and Joyce, 2013; Senthe-
bane et al., 2017, 2018). The TME can also modify cancer
cells, resulting in cancer cell heterogeneity (Dzobo et al.,
2018b; Quail and Joyce, 2013).
Studies have shown that subpopulations of cells within the
TME, including cancer-associated fibroblasts (CAFs), can
affect cancer cells differently (Ishii et al., 2016; O
¨hlund et al.,
2014, 2017). In addition, several studies have shown that the
extracellular matrix (ECM) can influence cancer cell prolif-
eration, migration, and response to therapy (Noguera et al.,
2012; Senthebane et al., 2018). This has resulted in increased
attention being given to the role of tumor-associated cells and
ECM as well as the development of therapies directed at the
tumor stroma. Recently, studies also demonstrated the gut
microbiota’s influence on cancer response to therapy through
modulation of TME (Iida et al., 2013; Ve
´tizou et al., 2015).
While underscoring the importance of microbiota in dis-
ease treatment and outcome, these studies show that modu-
lation of TME play a huge part in cancer cell response to
treatment. In addition, the activation of the immune system
has been shown to influence cancer cell sensitivity to radio-
therapy (Strom et al., 2017). Thus, the targeting or modula-
tion of the TME has real therapeutic value with the potential
to improve patients’ outcomes. A deeper understanding of the
TME and its contribution to cancer cell behavior is pertinent.
This comprehensive review presents advances in our
knowledge on CAFs and the role played by these cells in
disease initiation and progression. This review brings to the
fore the strategies being adopted to include TME-directed
therapies in the clinic, with the aim of reducing fatal disease.
The TME is used to describe the cells, ECM, and blood
vessels found within the vicinity of cancer cells in a tumor.
Included in this definition are cells such as fibroblasts, mac-
rophages, endothelial cells, lymphocytes, myeloid-derived
cells, and the ECM. Several studies have shown that de-
pending on the stage of tumor development, the TME can
provide both an inhibitory and promoting environment to
cancer cell growth (Senthebane et al., 2017, 2018). Cells
within the TME can be from the local vicinity or migrated
from distant environments (Friedl and Alexander, 2011;
Wels et al., 2008).
The CAFs are the most abundant cells within the TME and
play key roles in tumor initiation and progression (Kalluri,
2016; Su et al., 2018). Immune cells within the TME are
mostly macrophages (Hao et al., 2012; Kim et al., 2012).
Endothelial cells mostly function to form new blood vessels
that are necessary for the supply of nutrients to the growing
tumor and removal of toxic waste (De Palma et al., 2017). In
addition, endothelial cells also secrete platelet-derived
growth factor (PDGF), which attracts pericytes to tumor
blood vessels, and these pericytes are involved in the stabi-
lization of newly formed blood vessels (Armulik et al., 2011).
Stromal cells such as fibroblasts and macrophages are also
involved in the synthesis and maintenance of the ECM
(Bonnans et al., 2014; Riches, 1988). By releasing enzymes
such as matrix metalloproteinases, and growth factors, stro-
mal cells contribute to tumor ECM remodeling (Bonnans
et al., 2014). ECM remodeling enables cancer cells to migrate
and invade surrounding tissues (Gaggioli et al., 2007). As the
tumor develops, both tumor cells and the TME components
co-evolve and are transformed through the release of various
growth factors and other biomolecules, with the TME ini-
tially being tumor-restrictive but tumor-promoting at later
stages (Senthebane et al., 2017, 2018).
Several studies have demonstrated stromal cell heteroge-
neity, especially of CAFs and macrophages (Bauer et al.,
2010; O
¨hlund et al., 2017; Su et al., 2018). This heterogeneity
manifests as either tumor-restrictive or tumor-promoting
activities of the cells (O
¨zdemir et al., 2014; Rhim et al.,
2014). Elaborate studies by Su et al. as well as by O
¨hlund
et al. demonstrated the presence of distinct populations of
fibroblasts within tumors that have specific functions and that
they influence cancer cell response to therapy (Hwang et al.,
2008; O
¨hlund et al., 2017; Su et al., 2018).
Lately, the use of cell surface markers to isolate and
characterize these distinct population of fibroblasts has al-
lowed a deeper analysis of their behavior and functions (O
¨h-
lund et al., 2017; Su et al., 2018). The identification and use of
specific cell surface markers can allow targeted manipulation
of specific fibroblasts populations to achieve a specific goal
during cancer treatment. Under normal physiological condi-
tions, fibroblasts are dormant/quiescent or inactive but can be
activated by various growth factors, cytokines, and chemo-
kines (Gaggioli et al., 2007; Mueller and Fusenig, 2004).
Currently, very few clinical trials have been conducted that
target CAFs to treat cancer. This is partly because the translation
of laboratory scientific evidence into clinical use requires more
resources. It is, therefore, imperative that a deeper understand-
ing of stromal cell behavior and biology is obtained to improve
strategies targeting such cells. This review describes in detail
the involvement of CAFs in cancer pathogenesis.
Mapping CAFs’ Origins and Heterogeneity
In a landmark publication in 1858, Virchow described fi-
broblasts as cells that were spindle shaped and were re-
sponsible for the synthesis of collagen (Virchow, 1871).
Further studies showed that fibroblasts are mostly dormant
cells within the ECM and are transformed under conditions
such as inflammation, fibrosis, and wound healing (Darby
and Hewitson, 2007; Kalluri and Zeisberg, 2006). With both
inflammation and fibrosis being associated with cancer ini-
tiation and development, fibroblasts are, therefore, activated
CAFSIN TUMOR MICROENVIRONMENT 315
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
during these processes (Dvorak, 1986; Hanahan and Wein-
berg, 2011; Wynn and Ramalingam, 2012). These activated
fibroblasts associated with cancer are referred to as CAFs or
tumor-associated fibroblasts (Kalluri and Zeisberg, 2006;
Mueller and Fusenig, 2004).
Although initially having an anti-tumorigenic pheno-
type as normal fibroblasts, CAFs eventually become pro-
tumorigenic through mechanisms that are still under intense
investigations (O
¨hlund et al., 2014; Senthebane et al., 2017).
The CAFs will eventually become the dominant stromal cell
type within the TME and promote tumor progression via
release of several factors and the synthesis of the ECM
(Hanahan and Weinberg, 2011; Kalluri, 2016; Senthebane
et al., 2017, 2018).
Irrespective of their cell of origin, CAFs are large spindle-
shaped cells showing increased stress fibers and well-
developed cellular-ECM connections (De Wever et al.,
2008). Although their shape appears identical to normal fi-
broblasts, CAFs show increased numbers of ribosomes and a
rough endoplasmic reticulum (De Wever et al., 2008). Early
markers used for positive identification of CAFs include
alpha-smooth muscle actin (a-SMA), vimentin, and desmin
(Lazard et al., 1993; Znaor et al., 2018). Unlike their normal
equivalent, CAFs demonstrate increased proliferation and
migration capacity and show increased expression of ECM
proteins and ECM degrading enzymes (Ma et al., 2009; Saadi
et al., 2010). The resulting remodeling of the TME, referred
to as desmoplasia, causes fibrosis and stiffening of the tissue
(Kalli and Stylianopoulos, 2018; Poltavets et al., 2018).
The presence of CAFs and tissue stiffening have been
associated with cancer relapse, implying that CAFs and
desmoplasia contribute to cancer progression and therapy
resistance (Laklai et al., 2016; Tsujino et al., 2007). Calvo
et al. (2013) demonstrated that ECM remodeling is a re-
quirement for CAFs’ continuous presence within the TME.
Thus, there is a feedback loop whereby CAFs build and
maintain the TME and remodeling of the ECM whereas the
remodeling is needed for both the generation and mainte-
nance of CAFs (Calvo et al., 2013; Maller et al., 2013).
The balance between ECM synthesis and degradation is
needed for homeostasis maintenance. ECM proteins such as
collagens and fibronectin can block immune cells from in-
filtrating into the tumor (Cukierman and Bassi, 2010). In
addition, the ECM provides the ‘‘theater’’ in which all the
cellular interactions take place; blood vessels are formed; as
well as cancer cells are allowed to escape immune detection
(Cukierman and Bassi, 2010; Gorski et al., 1994).
The CAFs promote angiogenesis through the release of
MMPs, which degrade the ECM and allow the release of
vascular endothelial growth factor-A (VEGF-A) sequestered
by the ECM (Baeriswyl and Christofori, 2009). The release of
VEGF-A promotes the formation of the vascular system,
allowing the tumor to grow large with enhanced exchange of
nutrients and toxic substances (Baeriswyl and Christofori,
2009; Raica et al., 2009). The CAFs are known to release
several growth factors and cytokines that are known to pro-
mote inflammation and to assist in the evasion of the immune
system (Flavell et al., 2010; Yang et al., 2016). In short, CAFs
are the builders and are involved in the maintenance of the
TME via synthesis and release of ECM proteins and protein
factors (De Wever et al., 2008; Hwang et al., 2008; Kalluri,
2016; Kalluri and Zeisberg, 2006; Senthebane et al., 2017, 2018).
In turn, the TME promotes tumor initiation, progression,
and metastasis (De Wever et al., 2008; Hanahan and Wein-
berg, 2011; Kalluri, 2016; Kalluri and Zeisberg, 2006; O
¨h-
lund et al., 2014). Increased knowledge on the origins and
role of CAFs within the TME may allow the development of
new anti-cancer strategies (Brechbuhl et al., 2017; Bussard
et al., 2016; Casey et al., 2008; Glentis et al., 2017).
Several studies show that CAFs originate from different
cell types and this has been suggested to cause the hetero-
geneity observed in these cells (De Wever et al., 2008; Kalluri
and Zeisberg, 2006; O
¨hlund et al., 2014; Senthebane et al.,
2017). Besides the activation of fibroblasts within the vicinity
of the cancer cells, CAFs can also originate from bone
marrow-derived fibrocytes and mesenchymal stem/stromal
cells (MSCs) that are recruited to the tumor; the transdiffer-
entiation of pericytes, smooth muscle cells, and adipocytes;
epithelial cells that underwent epithelial to mesenchymal
transition (EMT); endothelial cells that underwent endothe-
lial to mesenchymal transition (EndMT); and, finally, the
activation of quiescent stellate cells (Fig. 1) (O
¨hlund et al.,
2017; Omary et al., 2007; Yin et al., 2013).
As demonstrated in breast cancer, bone marrow-derived
fibrocytes that are usually inactive and present in circulation
can be recruited to the tumor and become CAFs over time
(Barth et al., 2002).
Our recent work showed that MSCs can be converted to
‘‘CAFs’’ over time through interaction with cancer cells
(Senthebane et al., 2017). We demonstrated that transfor-
mation of MSCs into CAFs was dependent on the release of
transforming growth factor-beta (TGF-b) by both cancer
cells and the MSCs (Senthebane et al., 2017). Several other
studies have also shown that MSCs can transform/differen-
tiate into CAFs in several cancers ( Jung et al., 2013; Zhu
et al., 2014). For example, Weber et al. (2015) demonstrated
that osteopontin mediates TGF-b-dependent transformation
of MSCs into CAFs in breast cancer.
Tissue-resident MSCs and those recruited from distant
tissues and organs can interact with cancer cells and be
transformed to CAFs. Quiescent stellate cells can also
transform into CAFs when activated and contribute to the
CAF population in the liver and the pancreas (Omary et al.,
2007; Yin et al., 2013). Epithelial cells within the vicinity of
cancer cells can undergo EMT and become CAFs under fi-
brotic conditions (Iwano et al., 2002). This means that
epithelial-derived cancers can end up with a huge population
of CAFs driving tumor progression. Endothelial cells can
also undergo EndMT and become CAFs (Zeisberg et al.,
2007). Both transformed epithelial cells and endothelial cells
express CAF markers, including S100A4 (Iwano et al., 2002;
Zeisberg et al., 2007).
Through a transdifferentiation process, cells such as adi-
pocytes and pericytes can become CAFs (Dulauroy et al.,
2012; Jotzu et al., 2011). It is, therefore, plausible to speculate
that all cells that are within the vicinity of cancer cells and
those that end up at the tumor site via circulation can poten-
tially be transformed into cells that will eventually promote
tumor growth such as CAFs. We are just beginning to get a
clear picture of the components of the TME and functions in
tumor initiation and progression, and detailed mechanistic
studies are likely to reveal interesting information.
It is now accepted that CAFs show heterogeneity with
distinct subsets of cells displaying different phenotypes and
316 DZOBO AND DANDARA
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
functions within the TME (O
¨hlund et al., 2014, 2017; Orimo
and Weinberg, 2007; Su et al., 2018). Further, CAFs’ het-
erogeneity is also dependent on the stage of tumor develop-
ment (Huelsken and Hanahan, 2018). Normal fibroblasts are
generally considered genetically stable, whereas the same
cannot be said about CAFs. The process of transformation,
the generation of reactive oxygen species (ROS), and the
hypoxic conditions within the tumor can result in DNA al-
terations in CAFs as well, with CAFs co-evolving with
cancer cells as the tumor develops (Campbell et al., 2011;
O
¨hlund et al., 2014).
Although rare, alterations to the genetic material of CAFs
or stromal cells have been found in different cancers
(Moinfar et al., 2000; Patocs et al., 2007). Several available
techniques, including the use of CRISPR-Cas9 technology
and tracing experiments, can be used to determine whether
genetic alterations occur in CAFs as they do in cancer.
It is plausible to speculate that as the tumor evolves so do
the cells associated with it, such as CAFs. Our study clearly
demonstrates that previously anti-cancer activity of MSCs is
diminished over time and ‘‘transformed’’ cells eventually
become pro-tumorigenic (Senthebane et al., 2017). In addi-
tion, the synthesis of the ECM over time by both CAFs and
cancer cells results in combinatorial ECM, which is pro-
tumorigenic than normal ECM (Senthebane et al., 2018).
Clearly, more research and focused analysis of the TME
components is needed, especially the tumor-associated cells.
Given their possible cellular origin, it is not surprising that
CAFs display great phenotypic heterogeneity. Different
CAFs’ subsets have been identified within the TME and are
spatially distributed throughout the tumor (O
¨hlund et al.,
2014; Patocs et al., 2007). Recent characterization of CAFs
has identified specific biological markers for specific subsets
of CAFs (Kalluri, 2016; Vennin et al., 2019). In addition, no
CAF marker is expressed by all CAFs. Early studies identi-
fied markers such as a-SMA, fibroblast activation protein-
alpha (FAP-aor just FAP), and PDGF receptor-b(PDGFR-b)
to be expressed by different subsets of CAFs, with none being
expressed by all CAFs (Akrish et al., 2016; Kalluri, 2016). To
be useful, a combination of these markers is usually used
during CAFs’ characterization.
Among the markers, a-SMA has proven useful in the
identification of CAFs as well as a marker for smooth muscle
cells and pericytes (O
¨hlund et al., 2014). Senthebane et al.
(2017) used both vimentin and a-SMA as markers to study
cancer cell-derived TGF-b-mediated transformation of MSCs
to CAFs in vitro. Over time, the expression of both markers
was significantly increased in MSCs co-cultured with cancer
cells. Cancers in which CAFs express elevated levels of a-
SMA include breast, pancreatic, and liver cancers (Ayala
FIG. 1. CAFs can be derived from different cellular sources. Fibroblasts and stellate cells can be activated through various
processes to become CAFs; mesenchymal stem cells and fibrocytes are recruited to the tumor via circulation; epithelial cells
and endothelial cells undergo epithelial to mesenchymal transition and endothelial to mesenchymal transition, respectively,
to become CAFs. Further, pericytes and adipocytes are known to undergo transdifferentiation to become CAFs. CAFs,
cancer-associated fibroblasts; MSC, mesenchymal stem cell. Figure is adapted from our earlier publication, Senthebane
et al. (2017).
CAFSIN TUMOR MICROENVIRONMENT 317
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
et al., 2003; Yin et al., 2013). However, several other cells,
including normal fibroblasts, smooth muscle cells, cardio-
myocytes, and pericytes, also express a-SMA. Mostly in-
volved in the maintenance of cellular cell structure, a-SMA is
also involved in the migration and contraction of cells.
Another CAF marker vimentin is highly expressed by
CAFs in breast and prostate cancers (Kalluri and Zeisberg,
2006; Vuoriluoto et al., 2011). We recently showed that vi-
mentin can be induced via TGF-b-mediated transformation
of MSCs (Senthebane et al., 2017). Biological functions of
vimentin include promotion of migration and maintenance of
cellular structure and integrity (Guo et al., 2013; Toivola
et al., 2005; Wang and Stamenovic, 2002). In contrast to a-
SMA and vimentin, desmin is mostly downregulated in CAFs
and is also expressed by fibroblasts, muscle cells, and peri-
cytes (Armulik et al., 2011; Eyden, 2008).
Caveolin-1 is a scaffolding protein expressed in CAF
subpopulations as well as in cells such as endothelial cells,
fibroblasts, and adipocytes. Low expression of caveolin-1 is
used as a marker of a CAF subpopulation undergoing meta-
bolic reprogramming and promoting tumorigenesis (Guido
et al., 2012). In contrast, elevated levels of caveolin-1 are
observed in CAFs with the propensity to promote metastasis
(Cohen et al., 2004; Goetz et al., 2011). This clearly dem-
onstrates the challenges involved in sorting and isolating of
CAFs, a necessary step to their characterization, with huge
ramifications to their therapeutic targeting.
Recently, CD10 and G-protein-coupled receptor 77
(GPR77) have been shown to be highly expressed in CAFs
and are involved in promoting cancer stemness and resistance
to chemotherapy in breast cancer cells (Su et al., 2018).
Illustrating the challenges faced by any CAFs-targeted ther-
apies, CD10 and GPR77 are also expressed in bone marrow-
derived stromal cells and polymorphonuclear neutrophils,
respectively (Karnoub et al., 2007; Su et al., 2018).
S100A4, sometimes referred to as fibroblast-specific pro-
tein, is expressed by fibroblasts within tumors and displays
serine protease activity, allowing it to remodel the ECM
(Zhang et al., 2013a). Fibroblasts expressing S100A4 are
known to protect cancer cells via ECM production. Cells ex-
pressing S100A4 are known to be highly malignant and dis-
play a propensity for migration (Wang et al., 2005); for
example, S100A4 is highly expressed in CAFs in breast can-
cers (Orimo et al., 2005).
This marker is also expressed by other cells, including cells
undergoing EMT, macrophages, and normal fibroblasts
(Li et al., 2010; Orimo et al., 2005; Zhang et al., 2013a).
PDGFR-bis a CAF marker that has been targeted with kinase
inhibitors (Pietras et al., 2008). PDGF receptor signaling
inhibition with Imatinib was shown to abrogate malignant
progression of cervical lesions (Pietras et al., 2008). CAFs
found in colorectal and cervical cancers express high
levels of PDGFR-b(Pen
˜a et al., 2013; Pietras et al., 2008).
Macrophages have been shown to cause immune sup-
pression through the expression of FAP (Arnold et al.,
2014). The FAP is expressed in many human cancers and
is highly expressed in CAFs (Arnold et al., 2014; Huber
et al., 2003).
In summary, CAFs are a heterogeneous population of cells
clearly demonstrating their diverse cellular origin and have
been shown to have many functions within the solid tumor.
A major challenge facing scientists today is the identification
of reliable cell surface markers that can be used for thera-
peutic targeting. New data demonstrating that CAFs’ func-
tion is dependent on the location within the tumor amplify the
challenge faced (O
¨hlund et al., 2014, 2017). Irrespective of
cell of origin, tumor stage, and location within the tumor,
recent studies emphasize the need to identify cancer-specific
CAF markers or CAF subset markers for use in diagnosis and
anti-cancer targeting.
CAFs and Carcinogenesis
Data from several studies illustrate the supporting role of
TME components in tumor progression, metastasis, and for-
mation of new tumors (Bonnans et al., 2014; Cukierman and
Bassi, 2010; Senthebane et al., 2017). Although initially be-
ing prohibitive of tumor growth, we demonstrated that over
time cells within the TME promote tumor growth via the
release of growth factors and deposition of ECM (Nicosia
et al., 1993; Senthebane et al., 2017, 2018).
Several other studies confirmed that normal fibroblasts
show inhibitory effects on cancer cell growth in vitro (Stoker
et al., 1966). The expression of phosphatase and tensin ho-
molog in stromal cells was shown to be necessary for this
inhibition effect on epithelial tumors (Trimboli et al., 2009).
Several growth factors and cytokines, including vascular
endothelial growth factor (VEGF), stromal derived factor 1
(SDF-1), TGF-b, and interleukin-6 (IL-6), have been shown
to be necessary in the transformation of normal stromal cells
into cancer-supporting cells (Giannoni et al., 2010; Senthe-
bane et al., 2017).
In addition, conditions within the TME such as hypoxia
and ROS within the TME milieu also contribute to the con-
version of normal cells to pro-tumorigenic cells (Maxwell
et al., 1997; Ryan et al., 2000). Through the expression and
secretion of growth factors and cytokines and their receptors,
CAFs impact cancer cell-associated processes such as in-
flammation, angiogenesis, migration, invasion, chemoresis-
tance, and immune evasion (Fig. 2) (Casey et al., 2008; San
Francisco et al., 2004; Wang et al., 2017). Erez et al. (2010)
demonstrated that normal fibroblasts can be induced to ex-
press inflammatory genes by carcinoma cells.
microRNAs (miRNAs) are also believed to be involved in
the transformation of normal fibroblasts into CAFs. miRNAs
are delivered to their target cells via exosomes, allowing
miRNAs to induce and transform local and distant fibroblasts
(Kahlert and Kalluri, 2013; Webber et al., 2010). A recent
study by Fang et al. (2018) demonstrated that exosomal
delivered miRNA-1247 from cancer cells induces the trans-
formation of fibroblasts within lung premetastatic environ-
ments, thereby promoting metastasis.
Mitra et al. (2012) also demonstrated that cancer cells re-
program fibroblasts to CAFs via the release of miRNAs in
ovarian cancer. Pang et al. (2015) also showed that cancer
cell-derived miRNA-155 was involved in the transformation
of normal fibroblasts into CAFs in pancreatic cancer. It is
important to note that the involvement of miRNAs is bi-
directional, with stromal cells also affecting cancer cells via
the release of micro-RNAs (Musumeci et al., 2011; Nouraee
et al., 2013). The hypoxic conditions of the TME result in the
generation of ROS. Toullec et al. (2010) demonstrated that
ROS within the TME induce the expression of hypoxia in-
ducible factor 1-a(HIF1-a) and CXC-chemokine ligand 12
318 DZOBO AND DANDARA
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
by stromal fibroblasts, allowing the cells to have enhanced
metabolism and migratory capabilities.
Albrengues et al. (2015) showed that there is an epigenetic
switch that drives the transformation of normal fibroblasts into
CAFs. Leukemia inhibitory factor, a member of the IL-6 su-
perfamily, was shown to initiate the epigenetic switch, leading
to the activation of the Janus kinase–signal transducer and
activator of transcription (JAK-STAT) signaling (Albrengues
et al., 2015). The continuous activation of the JAK-STAT
signaling leads to CAFs promoting cancer cell invasive be-
havior. Several other studies have shown the importance of the
JAK-STAT signaling in the maintenance of the CAF pheno-
type (Gao etal., 2009; Huynh et al., 2017; Tan et al., 2019). The
continuous interaction between CAFs and cancer cells means
that during tumor progression CAFs also change, with CAFs
demonstrating heterogeneity at both phenotype and function
level (Bruzzese et al., 2014; Scherz-Shouval et al., 2014).
The growth and transformation of normal epithelial cells is
driven partly by CAFs (Olumi et al., 1999). Olumi et al.
(1999) demonstrated that CAFs and not normal fibroblasts
were responsible for the transformation as well as cancer
initiation and growth of prostatic epithelial cells. Ku-
perwasser et al. (2004) also demonstrated that TGF-bfrom
CAFs was necessary for tumor formation from epithelial
cells, a result that was in contrast to the use of normal fi-
broblasts. In addition, Shekhar et al. (2001) demonstrated that
normal fibroblasts inhibited tumor growth and the tumori-
genic transformation of epithelial cells.
Implantation of tumor cells and CAFs together allowed
enhanced tumor growth compared with co-implantation of
tumor cells and normal fibroblasts (Picard et al., 1986).
Several studies have shown the involvement of both cancer
cell-derived and CAF-derived TGF-band heat shock factor-1
in driving tumor cell growth and metastasis (Hawinkels et al.,
2014; Scherz-Shouval et al., 2014; Senthebane et al., 2017).
Using both esophageal and breast cancer cells, we demon-
strated that both CAFs and cancer cells released TGF-b,
which promoted the transformation of fibroblasts in addition
to promoting tumor growth (Senthebane et al., 2017). Several
cancer cells have been shown to express TGF-breceptors on
their cell surfaces, supporting the notion that cancer cells
respond to both autocrine and paracrine TGF-bsignaling
(Calon et al., 2014; Li et al., 2005).
Besides releasing growth factors, CAFs also influence tu-
mor growth through the release of enzymes such as MMPs
and urokinase-type plasminogen activator (Danø et al., 2005;
Deryugina and Quigley, 2015; Egeblad and Werb, 2002;
Martens et al., 2003; Taguchi et al., 2014).
Although further research into the role of CAFs into tumor-
igenesis is required, there is some evidence that CAFs induce
epithelial cell transformation (Wang et al., 2018). To sustain
tumor growth, new blood vessels are formed and these supply
all the nutrients required by cancer cells as well as remove toxic
substances from the tumor (Carmeliet and Jain, 2011).
CAFs release SDF-1, which has been shown to recruit
endothelial progenitor cells, leading to formation of new
blood vessels within the TME (Liu et al., 2019a; Orimo et al.,
2005). CAFs are known to release several other angiogenic
factors such as PDGF-C, VEGF-A, and secreted frizzled-
related protein-2 (SFRP2) (De Palma et al., 2017; Liekens
FIG. 2. Effect of CAFs on tumor cells, other stromal cells, and the ECM within the TM. The CAFs synthesize and secrete
the ECM, MMPs, and various factors, impacting cancer cell processes. Most of the secreted factors are growth factors and
cytokines, including HGF, FGF, TGF-b, IL-6, IL-1, COX-2, VEGF, HIF1-a, PDGF, and SDF-1, and they impact tumor
processes such as angiogenesis (1), chemoresistance (2), proliferation (3), and ECM remodeling (4). ECM, extracellular
matrix; FGF, fibroblast growth factor; HGF, hepatocyte growth factor; HIF1-a, hypoxia inducible factor 1-a; IL-6,
interleukin-6; MMPs, matrix metalloproteases; PDGF, platelet-derived growth factor; SDF-1, stromal-derived factor 1;
TGF-b, transforming growth factor-beta; TIMP, tissue inhibitor of metalloproteinases; TME, tumor microenvironment;
VEGF, vascular endothelial growth factor.
CAFSIN TUMOR MICROENVIRONMENT 319
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
et al., 2010). Through the synthesis of ECM proteins and
generation of ECM stiffness, CAFs influence blood vessel
formation and also the flow of blood through the tumor
(Egeblad et al., 2010; Senthebane et al., 2018; Twardowski
et al., 2007).
Most early-stage epithelial tumors have a basal lamina that
acts as a barrier between the tumor and the vascular system
(Bluff et al., 2009; Bossi et al., 1995). This results in early-
stage tumors lacking proper supply of nutrients and removal
of toxic substances. Migration and invasion of tumor cells
into the surrounding tissues induce angiogenesis as well
(Bergers and Benjamin, 2003). The induction of angiogenesis
is coupled to infiltration of leukocytes, proliferation and ac-
tivation of fibroblasts, as well as increased deposition of
ECM proteins (Dai et al., 2009; De Palma et al., 2017; Neve
et al., 2014; Wang et al., 2011; Zhou et al., 2006). Depending
on the tumor type, stromal components, and site in the human
body, vascularization can take different forms and patterns
(Bluff et al., 2009; Fukumura et al., 1997; Jubb et al., 2011;
Monsky et al., 2002).
In addition, the balance of expression of anti-angiogenic
factors and pro-angiogenic factors will ultimately play a key
role in determining both formation and pattern of blood
vessels within the tumor (Bussolino et al., 1991; Monsky et al.,
2002; Morrissey et al., 2008; Murakami and Simons, 2008).
Ultimately, pro-angiogenic factors and signaling are
eventually upregulated during tumor formation, leading to
aberrant and dysregulated formation of blood vessels (Ber-
gers and Benjamin, 2003; Castello et al., 2017). CAFs to-
gether with infiltrating leukocytes, macrophages, and other
stromal cells such as pericytes are known to be the major
sources of pro-angiogenic factors such as VEGFA, PDGFC,
fibroblast growth factor 2 (FGF2), Osteopontin, and MMPs
(Table 1, below) (Anderberg et al., 2009; Crawford et al.,
2009; Deroanne et al., 1997; Fukumura et al., 1998; Gerber
et al., 2000; Morikawa et al., 2002; Zhang et al., 2017; Zhao
et al., 2014).
For blood vessels to be formed there has to be the right
tumor or TME elasticity and stiffness (Egeblad et al., 2010;
Senthebane et al., 2017). The CAFs synthesize the TME
ECM; whereas enzymes released by CAFs such as MMPs,
hydroxylases, and lysyl oxidases influence both ECM syn-
thesis and degradation and, therefore, control biophysical
properties of the TME (De Palma et al., 2017; Dzobo et al.,
2012, 2014, 2018a; Liu et al., 2019b; Taguchi et al., 2014;
Twardowski et al., 2007). Growth factors such as PDGFC act
on CAFs in an autocrine manner to induce the secretion of
FGF2 as well as osteopontin (Anderberg et al., 2009; Liu
et al., 2019a; Pietras et al., 2008).
As the tumor grows in size, cancer cells migrate and invade
surrounding tissues. When cancer cells breach the basement
membrane, they enter into circulation, travel to distant tissues
and organs, and, eventually, extravasate into new tissues, a
process called metastasis. It is important to note that only a
few cancer cells survive the arduous journey to new tissues
and organs, but once there, these cancer cells colonize and
form new tumors, metastases (Chaffer and Weinberg, 2011;
Gupta and Massague
´, 2006). Grum-Schwensen et al. (2005)
demonstrated that tumors without stromal cell-derived mts1
protein do not metastasize, demonstrating the importance of
stromal cells in tumor growth and spread.
Several studies have shown that besides stromal-derived
growth factors, several cytokines are also released by stromal
cells and aid cancer cell metastasis (Chow and Luster, 2014;
Guo and Deng, 2018; Valkenburg et al., 2018). IL-6 has re-
ceived a lot of attention and has been shown to help cancer
Table 1. Several Angiogenic Factors Are Produced by Cancer-Associated Fibroblasts, Cancer-Associated
Macrophages, and Cancer-Associated Neutrophils and Influence Vascularization Within
the Tumor Microenvironment
Angiogenic factor Cellular source Effect on vascular system Effect on vascular system
VEGFA CAFs, CAMs, CANs Pro-angiogenic, recruits other
cells such as myeloid cells,
ECM production
Fukumura et al. (1998); Gerber
et al. (2000); Kalluri (2016)
PDGFC CAFs Pro-angiogenic Anderberg et al. (2009); Crawford
et al. (2009); Pietras et al.
(2008)
FGF CAFs, CAMs, CANs Pro-angiogenic Deroanne et al. (1997); Murakami
and Simons (2008)
CXCL12 CAFs, CAMs Pro-angiogenic, recruits
monocytes, endothelial cells
Liekens et al., 2010; Orimo et al.
(2005); Zhang et al. (2017)
CSF3 CAFs Pro-angiogenic Bussolino et al. (1991); Zhao et al.
(2014)
Collagen CAFs Pro-angiogenic Neve et al. (2014); Twardowski
et al. (2007); Zhou et al., 2006)
Fibronectin CAFs Pro-angiogenic Mongiat et al. (2016); Nicosia
et al. (1993)
Osteopontin CAFs Pro-angiogenic Castello et al. (2017); Dai et al.
(2009); Wang et al. (2011)
MMPs CAFs, CAMs, CANs Pro-angiogenic, creates space
for blood vessel formation
Deryugina and Quigley (2015);
Liu et al. (2019a); Taguchi et al.
(2014)
CAFs, cancer-associated fibroblasts; CAMs, cancer-associated macrophages; CANs, cancer-associated neutrophils; ECM, extracellular
matrix; FGF, fibroblast growth factor; MMPs, matrix metalloproteases; PDGFC, platelet-derived growth factor C; VEGFA, vascular
endothelial growth factor-A.
320 DZOBO AND DANDARA
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
cells metastasize to the bone (Tawara et al., 2011). Chang
et al. (2013) showed that IL-6 and its downstream signaling
cascades such as JAK-STAT are involved in breast cancer
tumorigenesis and metastasis.
The CAF-derived SDF1 has been shown to aid breast
cancer cells home to the bone and aid in adaptation to this
new environment (Al-Ansari et al., 2013; Zhang et al.,
2013b). The release of stanniocalcin-1 (STC1) by CAFs
drives colon cancer metastasis (Pen
˜a et al., 2013). TGF-bhas
been implicated in many cellular processes, and Yu et al.
(2014) demonstrated that TGF-bcan induce EMT in cancer
cells. Both TGF-band hepatocyte growth factor (HGF) have
been shown to promote invasiveness in esophageal cancer
(Grugan et al., 2010; Wang et al., 2016).
By releasing huge amounts of MMPs as well as synthesizing
the ECM, CAFs can remodel the TME and allow cancer cells to
migrate and invade surrounding tissues. Increase in collagen
levels within the TME has been shown to promote cancer cell
invasiveness (Provenzano et al., 2008; Vellinga et al., 2016).
Degradation of the ECM by CAF-derived MMPs can create
highways through which cancer cells can migrate to other tis-
sues and organs (Glentis et al., 2017; Senthebane et al., 2018).
In addition, ECM stiffening is enhanced via activation of YAP1
in CAFs and has been shown to allow cancer cell invasion and
formation of tumor blood vessels (Calvo et al., 2013).
Goetz et al. (2011) demonstrated that CAF-driven bio-
mechanical remodeling of the TME can allow migration and
invasion of cancer cells. Once cancer cells reach their new
environment, it has to be remodeled to suit their needs or the
cancer cells adapt to the new conditions. As postulated by
Paget in the ‘‘seed and soil’’ theory, the new microenviron-
ment must allow cancer cells to flourish (Paget, 1889).
Kaplan et al. (2005) suggested that a premetastatic envi-
ronment must be rich in ECM proteins such as fibroblast-
derived fibronectin. In addition, stromal cell-derived factors
including TGF-band SDF1 within the premetastatic regions
may act as attractants to cancer cells (Calon et al., 2012;
Kaplan et al., 2005). Resident fibroblasts within the pre-
metastatic regions promote blood vessel formation, allowing
tumors to grow (Calon et al., 2012; O’Connell et al., 2011). It
is plausible to suggest that these fibroblasts and any other
stromal cells associated with metastatic cancer cells are res-
ident cells within the colonized regions.
The Role of CAFs in Therapy Resistance
There are two possible ways through which cancer cells
can be resistant to therapy. First, therapy resistance can be
intrinsic. The expression of several transporter proteins such
as the ABC proteins and other genetic alterations can influ-
ence drug uptake and export at the cellular levels. Second,
tumors are always evolving and this can result in previously
responsive tumors to become irresponsive. The TME has
emerged as a contributor to therapy resistance via the actions
of stromal cells and the ECM. Excessive synthesis of the
ECM by CAFs can limit drugs’ access to cancer cells and
other stromal cells. The ECM around cancer cells can form a
hindrance to drugs and can limit blood vessel formation
within the tumor (Olive et al., 2009; Zhao et al., 2018).
The binding of cancer cells to CAF-derived ECM proteins
such as fibronectin, a process called cell-adhesion-mediated
drug resistance, also aids cancer cells in avoiding drugs, thus
causing drug resistance (Damiano et al., 2001; Hazlehurst
et al., 2000). The versatile IL-6 cytokine has been implicated
in resistance to chemotherapy in lung cancer (Shintani et al.,
2016). Although it is understandable that cancer cells can
develop therapy resistance, CAFs have also been reported to
be resistant to several drugs, including gemcitabine in pan-
creatic cancer (Richards et al., 2017). Communication be-
tween CAFs and cancer cells also includes exosomes, which
transport several factors and miRNAs from CAFs to cancer
cells (Richards et al., 2017).
Most chemotherapeutic drugs target rapidly growing
cancer cells, but they cannot effectively clear all cancer cells
during treatment. In addition, chemotherapy induces DNA
damage in stromal cells, resulting in the release of several
factors that promote cancer cell survival (Huber et al., 2015).
Further, several studies demonstrated that chemotherapy can
induce fibroblasts to become CAF-like cells that promote
stem cell behavior in cancer cells (Peiris-Page
`s et al., 2015a,
2015b). Radiotherapy is known to induce the synthesis of
large amounts of ECM proteins, resulting in the survival of
cancer cells (Barker et al., 2015; Eke and Cordes, 2011;
Erkan et al., 2007). When irradiated, stromal cells can in-
crease the secretion of factors involved in chemoresistance
(Chargari et al., 2013).
The EMT process is used by cancer cells to transform and
reduce the expression of many cellular membrane proteins
such as drug transporters (Zheng et al., 2015). The net effect
of reduced drug transporter expression is reduced drug in-
take, ultimately leading to therapy resistance. The EMT is a
process that is activated by several signaling pathways and
occurs in normal events such as embryonic development.
Unfortunately, it also occurs during neoplastic conversion
of cells as well as fibrosis (Arumugam et al., 2009; Hotz et al.,
2007). Cells undergoing EMT gradually lose epithelial cell
junction proteins and begin to synthesize more vimentin, be-
coming more migratory, invasive, and chemoresistant (Ar-
umugam et al., 2009; Hotz et al., 2007). Several studies have
shown enhanced vimentin, Snail, and ZEB1expression in cells
that are chemoresistant (Guaita et al., 2002; Taube et al., 2010;
Wellner et al., 2009). Zheng et al. (2015) demonstrated that
although EMT is not necessary for metastasis, it induces the
development of chemoresistance in pancreatic cancer.
In pancreatic cancer, resistance to gemcitabine is asso-
ciated with EMT (Vega et al., 2004; Wellner et al., 2009;
Zhang et al., 2010a). Poor survival of pancreatic patients
has been associated with an increased EMT program that
causes an impaired response to chemotherapy (Hidalgo,
2010; Kleger et al., 2014). Even more sinister is the seem-
ingly ability of CAFs to uptake and accumulate high con-
centrations of drugs within the TME, resulting in a reduced
amount of drugs eventually reaching cancer cells (Hess-
mann et al., 2018). Chemotherapy can kill cancer cells via
production of ROS. The CAFs have been shown to abrogate
ROS production and therefore prevent death of cancer cells
when exposed to chemotherapy (Cheteh et al., 2017).
Several survival pathways such as MEK-ERK and PI3K-
Akt are known to be activated in cancer cells via the action of
CAF-derived HGF and cause cancer cell resistance to RAF
inhibitors (Straussman et al., 2012). In elaborate experiments,
Senthebane et al. (2017) demonstrated that CAF-derived
TGF-bplays a role in the resistance of breast and esophageal
cancer cells to paclitaxel and cisplatin. Thus, the CAF
CAFSIN TUMOR MICROENVIRONMENT 321
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
secretome plays a huge role in the development of therapy
resistance in cancer cells.
The presence of cancer stem cells (CSCs) within the TME
is still debatable, but several studies have shown that these
stem-like cells are able to resist therapies (Codd et al., 2018;
Dawood et al., 2014; Nassar and Blanpain, 2016; Steinbichler
et al., 2018; Yu et al., 2007a). Several studies have demon-
strated that CAFs subsets express drug transporter proteins
such as ABC transporters and also provide a protective en-
vironment to CSCs within the TME (Begicevic and Falasca,
2017; Domenichini et al., 2019; Su et al., 2018). CAF-derived
cytokines such as IL-6 are responsible for maintenance of the
CSC phenotype (Ebbing et al., 2019).
Microvesicles from CAF also deliver micro-RNAs to
generate CSCs in breast cancer (Sansone et al., 2017).
Overall, studies have demonstrated that CAFs or their sub-
sets promote therapy resistance in cancer cells via various
mechanisms from accumulating large amounts of drugs, re-
lease of protein factors, and mi-RNAs-laden exosomes to
providing physical barriers to therapy. The combined tar-
geting of these CAFs or their subsets together with cancer
cells may offer durable treatment solutions for many cancers.
CAFs and Other Stromal Cell Interactions
The interaction between TME components is through cell-
to-cell adhesions, release of growth factors, and cytokines
and indirectly through exosomes. In addition, cells interact
with the ECM via surface receptors such as integrins.
Crosstalk between stromal cells is inevitable given the
compact nature of most solid tumors. The most abundant
immune cells within the TME are the macrophages, referred
to as cancer-associated macrophages (CAMs) or tumor-
associated macrophages (TAMs).
As reported by Comito et al. (2014), CAFs and TAMs
work together to promote tumor progression in prostate
cancer. Communication between the two types of cells occurs
via the release of CXCL14 by CAFs, resulting in the re-
cruitment of TAMs to the tumor site and subsequent differ-
entiation (Comito et al., 2014). In turn, M2 TAMs are known
to activate CAFs and therefore further promote tumorigenic
growth. Hashimoto et al. (2016) demonstrated that CAFs
derived from bone marrow MSCs promote macrophage in-
vasiveness and transformation and, in turn, the resulting
TAMs can prompt the proliferation of CAFs. By working
together, these CAFs and TAMs can promote tumorigenic
growth of neuroblastoma (Hashimoto et al., 2016).
Myeloid cells within tumors can increase the expression of
S100A8 in response to IL-6 released by CAFs in colon cancer
(Kim et al., 2012). Myeloid cells within tumors can also
differentiate into suppressor cells that are linked to suppres-
sion of immunity (Kim et al., 2012). For example, circulating
myeloid-derived suppressor cells are recruited to tumors via
the release of FAP by CAFs and these cause the suppression
of immunity in hepatic cancer (Yang et al., 2016).
The CAFs are known to express the FAS ligand and this is
known to cause apoptotic death of CD8-positive T cells
(Lakins et al., 2018). Further, CAFs express programmed
cell death 1 ligand 2 (PD-L2), which causes the functional
inactivation of T cells (Lakins et al., 2018). Our study and
several others have shown that CAFs secrete huge amounts
of TGF-b, which has been suggested to cause immunosup-
pression and can induce transformation of immune cells into
immune-suppressing cells (Flavell et al., 2010; Gutcher
et al., 2011; Senthebane et al., 2017). Recently, Mariathasan
et al. (2018) provided further evidence of the involvement of
TGF-bin abrogating antitumor immunity in bladder cancer.
The CAFs attract and sequester CD8-positive T cells and
this prevents T cells from binding to and killing cancer cells
(Ene–Obong et al., 2013).
The net effect of all this is the suppression of anti-tumor
activity of immune cells. The removal of FAP-positive
CAFs was shown to reactivate the anti-tumor activity of
immune cells of T cells (Kraman et al., 2010). Overall, CAFs
are more likely to promote tumorigenic growth through their
effect on immune cells within the TME. Several growth
factors are known to be sequestered by the ECM and these
include VEGFA and TGF-b. Through production of matrix
metalloproteases (MMPs), CAFs can degrade the ECM and
cause enhanced levels of free growth factors within the TME
(Bonnans et al., 2014). Thus, through recruiting endothelial
cells in addition to releasing VEGFA, CAFs promote tumor
vascularization (Bonnans et al., 2014). The interaction of
CAFs and other cells, therefore, creates a tumor environ-
ment that promotes immune evasion, inflammation, and
vascularization.
As reported by Senthebane et al. (2017), CAFs and other
TME components can also display anti-tumorigenic effects,
especially during the early stages of tumor development. Our
data revealed that normal fibroblast-derived ECM limits
cancer cell proliferation and migration compared with an
ECM from fibroblasts co-cultured with cancer cells (Sen-
thebane et al., 2018). Further evidence of the involvement of
CAFs or their normal counterparts in anti-tumorigenic be-
haviour comes from studies of breast cancer. Brechbuhl et al.
(2017) reported two subsets of CAFs in breast cancer, with
one subset expressing CD146 and conferring sensitivity to
tamoxifen, whereas the CD146- subset confers tamoxifen
resistance. The removal of CAFs expressing a-SMA from
pancreatic ductal adenocarcinoma (PDAC) results in the
acceleration of cancer and reduced survival (O
¨zdemir et al.,
2014; Rhim et al., 2014).
With this in mind, novel therapeutic strategies aimed at
CAFs have to be selective and cognizant of the CAFs’ phe-
notypic and functional heterogeneity. Mere targeting of
CAFs for removal based on presumed pro-tumorigenic be-
havior will be detrimental and can lead to fatal disease. The
CAF subsets must be identified and only targeted therapy
should be used. In addition, it might be helpful to re-educate
or re-direct the pro-tumorigenic CAFs into anti-tumorigenic
CAFs. To be able to do this, it is important that specific
markers for anti-tumorigenic and pro-tumorigenic CAFs be
identified to distinguish the two.
Advances in Therapeutic Targeting of CAFs
CAF-directed therapies
It has long been recognized that manipulation of CAFs’
functions, especially the tumorigenic promotion part, have
therapeutic value in cancer treatment. In theory, targeting
CAFs appear easy since they are the major stromal compo-
nents of the tumor. In addition, CAFs have been described as
genetically stable, making them a better target than cancer cells
in cancer immunotherapy (Ostermann et al., 2008). In practice,
322 DZOBO AND DANDARA
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
however, major challenges need to be overcome before this
promising avenue is utilized in cancer treatment.
First, targeting CAFs will have a negative effect on normal
tissue and other components of the TME. For example, the
depletion of CAFs can result in less ECM production, cre-
ating highways through which cancer cells escape and me-
tastasize to other tissues and organs. Second, the removal of
CAFs itself suffers from the lack of specific markers that can
be used to achieve that. Third, and probably most challenging
is the existence of CAFs subsets displaying phenotypic and
functional heterogeneity. To overcome this requires the
identification of more than one specific markers of each
subset that can be used for targeting that subset.
Several approaches have been devised to target CAFs
within the TME. First, the activation and trans-differentiation
of stromal cells into CAFs can be targeted and inhibited.
Since CAFs influence both tumor cells and stromal cells via
synthesis and secretion of growth factors and cytokines, in-
hibition of this synthesis and secretion is under intense
scrutiny, with the aim of inhibiting CAF function. Second,
CAFs’ normalization through the use of several molecules
has been tried.
For example, all-trans-retinoic acid (ATRA) can be used to
induce quiescence in CAFs or activated fibroblasts/stromal
cells, preventing aberrant secretion of growth factors and
cytokines. Although CAFs are pro-tumorigenic, they can be
used as carriers of drugs to kill cancer cells. Together with
other cells such as CAMs, MSCs, and pericytes, CAFs can
carry anti-cancer viruses and apoptosis-inducing ligands.
Several studies, including our recent publication, have
demonstrated that MSCs can be transformed into CAFs
(Borriello et al., 2017; Chan et al., 2019; Ridge et al., 2018;
Senthebane et al., 2017; Tan et al., 2019). Senthebane et al.
(2018) demonstrated the influence of the ECM in tumor
growth and chemoresistance.
Importantly, the presence of ECM proteins, including type
I collagen and fibronectin, was shown to affect drug delivery
to cancer cells, with the knockdown of both ECM proteins
showing increased drug-induced cancer cell death (Senthe-
bane et al., 2018). Previous studies implicated signaling
pathways such as the MEK-ERK and MMPs in regulating
ECM synthesis (Dzobo et al., 2012, 2014). The ablation of
stromal ECM can, therefore, be used to increase cancer cell
sensitivity to anti-cancer drugs. Direct targeting of CAFs or
their subsets is still challenging, with difficulties in the
identification of markers to be used to identify the CAFs.
Currently, immunotherapy, use of immunotoxins, and DNA
vaccines can be used to target CAFs, with the duly still out on
the effectiveness of such therapy.
Senthebane et al. (2017) recently utilized both vimentin
and a-SMA as markers to identify MSC-derived CAFs. Other
studies have shown that a-SMA only identifies a specific
subset of CAFs (Choi et al., 2013; Ding et al., 2014; O
¨zdemir
et al., 2014). O
¨zdemir et al. (2014) described that the removal
of a-SMA-positive CAFs in pancreatic ductal adenocarci-
noma (PDAC) was linked to decreased angiogenesis. The
same authors observed that angiogenesis decreased whereas
there was increased hypoxia within solid tumors leading to
increased EMT and the presence of CSCs (O
¨zdemir et al.,
2014). Several lung and colon cancer animal models have
shown that removal of FAP from within solid tumors is as-
sociated with decreased tumor growth (Santos et al., 2009).
Santos et al. (2009) demonstrated that a preclinical FAP
inhibitor, PT630, can inhibit the growth of tumors as well as
stromagenesis in lung and colon cancers. Similarly, the re-
moval of FAP in PDAC also reduced tumor growth (Feig
et al., 2013). Ostermann et al. (2008) developed a monoclonal
antibody against FAP that they conjugated to maytansinoid
and this treatment has been used in pancreatic, head, and neck
cancers where it has shown its effectiveness.
In addition, extensive experiments by Loeffler et al. (2006)
using a mouse DNA vaccine against FAP resulted in ablation
of CAFs and enhanced drug uptake by tumors. The DNA
vaccine induced a CD8
+
T cells to kill a large part of the
CAFs expressing FAP. Further evidence for the utility of the
DNA vaccine against FAP-positive CAFs was provided by
Reisfeld (Reisfeld, 2007). The utility of chimeric antigen
receptor T (CAR T) cell treatment for solid tumors is still to
be proven.
Several reports, however, show that targeting FAP-positive
CAFs using CAR T cells results in antitumor activity (Kakarla
et al., 2013; Lo et al., 2015; Wang et al., 2014). However,
CAFs are not the only cells expressing FAP. Bone marrow-
derived MSCs are also known to express FAP (Chung et al.,
2014). Thus, it is possible that anti-tumor CAR T cells will also
target other cells. Although reports show that CAR T cells
targeting FAP-positive CAFs are able to kill cancer cells in
lung and pancreatic cancers (Lo et al., 2015), Roberts et al.
(2013) demonstrated that CAR T cells killed bone marrow-
derived stem cells and can cause cachexia and anemia.
Increased knowledge and analytical tools have enabled
scientists to identify new and better surface markers for
CAFs. For example, Su et al. (2018) were able to characterize
tumorigenic CAF subsets based on the expression of CD10
and GPR77. Used together with FAP and a-SMA, these new
surface markers are likely to define specific CAF subsets,
leading to their killing.
The same authors reported reduced cancer cell stemness
when GPR77 was blocked (Su et al., 2018). Several strategies
are being developed to target cellular sources of CAFs.
Senthebane et al. (2017) demonstrated that it is possible that
MSCs are transformed into CAFs through their interaction
with cancer cells. Thus, targeting MSCs may prevent the
accumulation of CAFs in tumors, reducing pro-tumorigenic
cells within the tumor. The removal of pro-tumorigenic
support is likely to reduce tumor growth and metastasis.
Several clinical trials have been undertaken to target CAFs
sources with the duly still out on their effectiveness.
The re-education of activated CAFs has been suggested as
a strategy to direct CAFs from being pro-tumorigenic to anti-
tumorigenic. The inactivation of CAFs can induce quies-
cence, with the resulting CAFs or fibroblasts dividing slowly
and releasing normal levels of growth factors and other fac-
tors. Akin to quiescent satellite cells in muscle, these re-
educated CAFs can attain a tumor-suppressive phenotype.
The induction of quiescence in pancreatic stellate cells in
PDAC through the use of ATRA reverts the activated cells to
normal fibroblasts and arrests tumor growth (Froeling et al.,
2011). The mechanism of action of ATRA involves inhibition
of the versatile Wnt-b-catenin signaling cascade (Froeling
et al., 2011).
Inactivated pancreatic stellate cells also release seques-
tered CD8
+
T cells, allowing infiltration into PDAC and re-
sulting in tumor growth arrest (Ene–Obong et al., 2013).
CAFSIN TUMOR MICROENVIRONMENT 323
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
Carapuca et al. (2016) showed that coupling ATRA, a vita-
min A analogue, and gemcitabine can be effective at treating
PDAC in animal models through inhibition of several path-
ways, including Wnt-b-catenin and Hedgehog signaling.
Sherman et al. (2014) demonstrated that global reprogram-
ming of the stroma through the use of calcipotriol, a vitamin
D receptor ligand, and gemcitabine resulted in reduced in-
flammation and ECM synthesis in PDAC tumors. Reduced
ECM synthesis allowed better delivery of gemcitabine into
the tumor, leading to tumor regression (Sherman et al., 2014).
Both ATRA and calcipotriol are at the preclinical stage of
investigations. Thus, depending on the tumor type, different
strategies from ablation of CAFs to normalization of acti-
vated CAFs may be undertaken to achieve better cancer
treatment. This provides a new avenue of cancer treatment
that focuses not just on cancer cells but on the stromal
component as well. Combinations of strategies focused on
cancer cells and stromal cells may offer a durable cure for
cancer treatment.
Targeting signaling pathways activated in CAFs
and their downstream effectors
One of the remaining challenges to CAFs’ ablation and
normalization is the huge heterogeneity observed in many
tumors, meaning some CAFs subsets will remain unaltered
by treatment. Instead of targeting the CAFs, several ap-
proaches have been developed to target signaling pathways
activated in CAFs and factors released by tumor and stromal
cells. Tumor cells and stromal cells release huge amounts of
factors during tumor initiation and progression (Senthebane
et al., 2017, 2018; Sullivan et al., 2010). Senthebane et al.
(2017) demonstrated that TGF-breleased by tumor cells and
transforming MSCs can be involved in the transformation of
stromal cells into potential CAFs. The same study also
showed that once transformed, activated CAFs release in-
creased levels of the same growth factor, TGF-b, which is
involved in tumor growth and development of chemoresis-
tance (Senthebane et al., 2017).
One signaling cascade that has been studied in detail in
different cellular processes is the IL-6-JAK-STAT pathway
(Giannoni et al., 2010; Sansone and Bromberg, 2012; Steyn
et al., 2019). Sanz-Moreno et al. (2011) showed that On-
costatin M, a member of the IL-6 superfamily, activates
STAT3 signaling and drives ECM remodeling. ECM re-
modeling allows cancer cells to invade surrounding tissues,
escape, and metastasize to other sites (Sanz-Moreno et al.,
2011). A detailed description of inhibition of the IL-6-JAK-
STAT signaling pathway as well as the TGF-bsignaling is
given next.
Pietras et al. (2008) demonstrated that the reduction of
FGF2 and fibroblasts growth factor-7 (FGF7) in animal
models of cervical cancer via Imatinib-mediated inhibition of
PDGF receptor slowed cancer cell division and disrupted
angiogenesis. The authors showed that targeting CAFs can
act in complement to conventional therapeutic strategies and
improve the management of cancers that are difficult to cure.
Research by Anderberg et al. (2009) further showed that
PDGF-CC induces the expression of osteopontin in CAFs,
leading to tumor growth acceleration.
The chemokine SDF1, exclusively produced by FAP-
positive CAFs, binds to its receptor CXCR4, resulting in the
suppression of immunity within the TME through the pre-
vention of CD8
+
T cell infiltration (Fearon, 2015; Feig et al.,
2013; Kraman et al., 2010). Several inhibitors of SDF1-
CXCR4 interactions have been developed and these can
reactivate the anti-tumor immunity by enhancing the infil-
tration of CD8
+
T cells into the TME (Feig et al., 2013).
Inhibition of SDF1 and CXCR4 interactions include the use
of inhibitors such as AMD3100 and may enhance the anti-
tumor effects of monoclonal antibodies against PDI/PDL1 in
pancreatic duct adenocarcinoma (Feig et al., 2013).
Another strategy under consideration to control CAF-
derived factors includes the inhibition of protein synthe-
sis. The inhibition of protein synthesis through the use of
SOM230, an inhibitor of protein synthesis in a-SMA-positive
CAFs, resulted in reduced levels of several molecules (Duluc
et al., 2015). Indeed, the inhibition of protein synthesis in
CAFs led to less ability of cancer cells to resist chemotherapy
in a PDAC model (Duluc et al., 2015).
Targeting the IL-6-JAK-STAT signaling in cancer
It has been shown that the IL-6-JAK-STAT3 is abnor-
mally activated in several cancers, with enhanced activation
of the pathway linked to poor patients’ survival (Chen et al.,
2013b; Kusaba et al., 2006; Macha et al., 2011). Within the
TME, the IL-6-JAK-STAT3 signaling has been shown to be
responsible for cancer cell invasive and metastatic behavior.
In addition, the IL-6-JAK-STAT3 signaling represses im-
munological response to tumors. Anti-cancer agents or in-
hibitors against members of the IL-6-JAK-STAT3 have
been approved by the FDA and the EMA, whereas others are
still under investigation.
Several pathological conditions display high levels of
IL-6 and increased activation of the IL-6-JAK-STA3, in-
cluding rheumatoid arthritis as well as many cancers (Ku-
mari et al., 2016; Masjedi et al., 2018). Mutations in genes
of several JAK-STAT pathway members activate the path-
way in many neoplasms. Increased IL-6 in circulation and
within the TME is a result of the JAK-STAT pathway being
constitutively activated (Kusaba et al., 2006; Taniguchi and
Karin, 2014; Wang et al., 2013).
Besides CAFs, multiple other cells have been shown to
produce IL-6 within the TME, from pericytes, immune cells,
and even tumor cells (Bournazou and Bromberg, 2013; Na-
gasaki et al., 2014; Nozawa et al., 2006). The activation of
STAT3 by IL-6 in tumor cells results in the expression of
proliferation and survival genes, including cyclin D1 and
BCL2-like protein 1 (BCL-xL), respectively (Fisher et al.,
2014). Since both CAFs and tumor cells show activation of
IL-6-JAK-STAT signaling, the description given next and the
targeting of the IL-6-JAK-STAT signaling can therefore be
applied to both CAFs and tumor cells to achieve durable
cancer treatment. IL-6 activated STAT3 can induce VEGF,
MMPs, and TGF-bexpression, promoting angiogenesis, in-
vasion, and development of therapy resistance, respectively
(Fisher et al., 2014; Yu and Jove, 2004; Yu et al., 2009).
Besides direct effects on cancer cells, IL-6 and JAK-STAT
signaling influences the behavior of stromal cells, including
CAFs, CAMs, TANs, effector T cells, and killer cells (Harris
et al., 2007; Iwata-Kajihara et al., 2011; Kortylewski et al.,
2005; Kujawski et al., 2010; Lee et al., 2010; Yu et al.,
2007b). By affecting both CAFs and immune cells, the
324 DZOBO AND DANDARA
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
IL-6-JAK-STAT signaling contributes to the suppression of
the immune response in the TME.
The importance of IL-6 in TME is underscored by its
various effects on tumor cell and stromal cell survival, pro-
liferation, and invasiveness (Kortylewski et al., 2005; Le-
derle et al., 2011). Two IL-6 signaling pathways are known:
the classical IL-6 signaling pathway, which involves IL-6
binding to the IL-6 receptor on the cell surface and interacts
with transmembrane protein gp130; the trans-signaling
pathway whereby IL-6 binds to a soluble form of the IL-6
receptor and then interacts with gp130. Importantly, although
similarities exist in the ways in which both signaling path-
ways regulate cell behavior, the trans-signaling pathway
controls the recruitment and activation of stromal cells
(Campbell et al., 2014; Hunter and Jones, 2015). Activation
of the IL-6 signaling results in activation of mostly JAK1 and
JAK2, which then phosphorylate members of the STATs
family ( Johnson et al., 2018; Steyn et al., 2019).
Of the STATs proteins, STAT3 is the most studied and has
been linked with tumor progression and suppression of the
immune system (Bromberg, 2002; Yu et al., 2009). Activa-
tion of STATs proteins is regulated by PIAS proteins, sup-
pressor of cytokine signaling proteins, phosphatases, and
miRNAs (Dorritie et al., 2014; Kim et al., 2010; Sekine et al.,
2006; Zhang et al., 2007).
The IL-6 levels are increased in cancers, including breast,
lung, colorectal, esophageal, ovarian, and prostate cancers
(Chen et al., 2013a; Chung and Chang, 2003; Culig and Puhr,
2012; Dethlefsen et al., 2013; Maccio
`and Madeddu, 2013).
Several preclinical studies utilizing models and patient
samples have demonstrated the critical roles of circulating
IL-6 in tumor development (Becker et al., 2004; Lesina et al.,
2011; Sansone et al., 2007). For example, IL-6 enhances CSC
proliferation in breast cancer (Sansone et al., 2007). In ad-
dition, IL-6 is known to promote EMT and metastasis in
breast cancer (Chang et al., 2013; Sullivan et al., 2009).
Consequently, IL-6 levels in circulation have been used as
prognostic indicators in patients’ survival and can predict
response to therapy (Gao et al., 2016; Knupfer and Preiss,
2010; Maccio and Madeddu, 2013; Sanguinete et al., 2017).
Several studies utilizing models and patients’ samples
have demonstrated the critical role played by CAF- and
tumor-derived IL-6 cancers, including breast, colorectal,
esophageal, lung, and skin cancers (Gao et al., 2007; Gri-
vennikov et al., 2009; Lederle et al., 2011; Sansone et al.,
2007). Aberrant levels of STAT3 activation have been shown
in many cancers (Frank, 2007; Roeser et al., 2015). Besides
being involved in tumor progression, several studies have
shown that STAT3 can promote therapy resistance (Sen et al.,
2012; Spitzner et al., 2014).
Several strategies have been developed to target the IL-6-
JAK-STAT signaling pathway (Fig. 3). Siltuximab, for
example, targets IL-6 directly and Tocilizumab targets the
IL-6 receptor. Siltuximab has shown efficacy against sev-
eral solid tumors, including lung and prostate cancers (Ca-
varretta et al., 2008; Song et al., 2014). Siltuximab have
been shown to reduce the levels of STAT3 and other sig-
naling cascades such as MEK-ERK in tumors (Fizazi et al.,
2012; Karkera et al., 2011). Tocilizumab is used in patients
with rheumatoid arthritis and has efficacy against colorectal
and pancreatic cancers (Dijkgraaf et al., 2015; Grivennikov
et al., 2009).
FIG. 3. Some of the strategies targeting both CAF- and tumor cell-derived IL-6-JAK-STAT signaling pathways are
indicated. JAK, Janus kinase; STAT, signal transducer and activator of transcription; X, blocking of signaling step.
CAFSIN TUMOR MICROENVIRONMENT 325
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
Inhibitors of JAKs include Pacritinib, Ruxolitinib, and
Tofacitinib and these have shown varied efficacy against
cancers such as colitis, myelofibrosis, liver, pancreatic, and
ovarian cancers (Hedvat et al., 2009; Komrokji et al., 2015;
Neubauer et al., 1998; Sandborn et al., 2017; Tavallai et al.,
2016; Vannucchi et al., 2015). The STAT3 inhibitors dem-
onstrated antitumor activities and promoted apoptosis against
several cancer cells (Chen et al., 2007; Fuh et al., 2009; Pan
et al., 2013; Schust et al., 2006; Siddiquee et al., 2007; Zhang
et al., 2010b).
Targeting the CAF-derived TGF-bsignaling
pathway in cancer
Several reports document the lack of cancer patients’ re-
sponse to therapy being associated with TGF-bsignaling in
CAFs and CAMs (Brunen et al., 2013; Colak and Ten Dijke,
2017; Ganesh and Massague, 2018; Hao et al., 2019; Ja-
kowlew, 2006; Mariathasan et al., 2018; Reiss, 1999). In
normal cells and during early stages of tumor development,
the TGF-bsignaling displays anti-tumor functions, with re-
ports of cancer cell cycle arrest and induction of apoptosis
(Colak and Ten Dijke, 2017; Gu and Feng, 2018; Senthebane
et al., 2017).
Our study demonstrated that as tumors develop, TGF-b
from both stromal cells and tumor cells participates in the
transformation of normal fibroblasts and MSCs into CAFs
and thus contributes to further tumor development, metasta-
sis, and therapy resistance (Fig. 4) (Bussard et al., 2016;
Castells et al., 2012; Senthebane et al., 2017). Thus, de-
pending on the stage of tumor development, TGF-bsignaling
can have both tumor-promoting and tumor-suppressive
functions. Targeting this pleiotropic signaling cascade is
therefore difficult, with further research required to determine
the specific members of the pathway to target as well as the
right doses. Importantly, the effects of inhibition of this sig-
naling cascade on both cancer cells and stromal components
must be further investigated. The elucidation of possible
biomarkers that can point to the usefulness of potential in-
hibitors during and after treatment must be determined.
In their elaborate study, Mariathasan et al. (2018) dem-
onstrated that a combination of TGF-bsignaling blockade
and anti-PD-L1 repressed TGF-bsignaling in stromal cells,
leading to enhanced tumor suppression. Importantly, the
authors demonstrated the need to combine treatment strate-
gies to attain durable cure in cancer. Several drugs targeting
TGF-bare under trial and these include fresolimumab
(NCT02581787).
Targeting CAF-derived ECM proteins
and associated signaling
The CAFs are the principal cells responsible for synthe-
sizing the ECM (Barbazan and Matic Vignjevic, 2019; Er-
dogan and Webb, 2017; Houthuijzen and Jonkers, 2018;
Khan et al., 2018; Liu et al., 2019b). In normal tissue, fibrillar
collagens, fibronectin, hyaluronan, and tenascin C are de-
posited in large amounts to make the ECM. Increased syn-
thesis of ECM proteins results in desmoplastic reactions and
these are known to drive tumor growth (Ronnov-Jessen et al.,
1996). Besides inducing desmoplastic reactions, increased
ECM synthesis creates a barrier for the delivery of drugs to
cancer cells (Laklai et al., 2016; Senthebane et al., 2017). In
detailed in vitro experiments, we demonstrated that the
presence of an ECM increased the resistance of breast and
esophageal cancer cells to several chemotherapeutic agents
(Senthebane et al., 2018).
Although it is plausible that a reduction in ECM synthesis
in CAFs and other stromal cells such as macrophages is an
appealing strategy, improper ECM synthesis can create
FIG. 4. Both tumor and stromal cells secrete TGF-binto the TM milieu. TGF-band other growth factors can be
sequestered by the ECM. Increased TGF-bwithin the TM leads to transformation of MSCs and fibroblasts into CAFs.
Increased CAF- and tumor-derived TGF-blevels cause increased tumor growth, chemoresistance, and metastasis. Figure
adapted from Senthebane et al. (2017).
326 DZOBO AND DANDARA
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
‘‘highways’’ through which cancer cells may metastasize
( Jabłon
´ska-Trypuc
´et al., 2016; Senthebane et al., 2017,
2018). Increased amounts of both collagen and hyaluronan in
tumors can result in compressed vascular networks, inad-
vertently reducing the flow of drugs to cancer cells (Chauhan
et al., 2013; Dzobo et al., 2014). Targeting both collagen and
hyaluronan production by both CAFs and macrophages can
allow drugs to reach cancer cells (Chauhan et al., 2013). The
inhibition of collagen through the use of halofuginone has
been shown to reduce desmoplasia, impacting tumor pro-
gression ( Juarez et al., 2012).
Enzymatic degradation of ECM proteins, such as collagen
via MMPs occurs during ECM remodeling. The use of MMPs
as an anti-cancer therapy has largely failed to materialize
(Vandenbroucke and Libert, 2014). For a start, there is so
much overlap in MMP activities and indiscriminate degra-
dation of ECM proteins can have a negative effect during
treatment (Vandenbroucke and Libert, 2014).
Currently, several MMPs inhibitors are under investigation
in many cancers (Chiappori et al., 2007). Deciphering the
exact MMPs activities and their specific targets can aid in the
development of novel use of MMPs in cancer treatment
(Coussens et al., 2002; Dzobo et al., 2012; Wojtowicz-Praga
et al., 1997). Hyaluronan can be depleted through the use of
recombinant hyaluronidase enzyme to allow drugs to reach
cancer cells ( Jacobetz et al., 2013; Provenzano et al., 2012).
Targeting hyaluronic acid via the use of PEGPH20 is still
under investigation (NCT01453153). New clinical trial data
(HALO-109-301) show that when used together with gem-
citabine and nab-paclitaxel in patients with advanced pan-
creatic cancer, PEGPH20 is promising (Doherty et al., 2018).
Caution, however, must be heeded as the resulting expan-
sion of the vascular system can result in invasion and metas-
tasis of cancer cells. A combination of the removal of ECM
proteins during treatment can enhance patients’ survival,
demonstrated in PDAC by Hingorani et al. (2016). In vitro
work by Senthebane et al. (2018) demonstrated that the re-
moval of both collagen and fibronectin from ECM resulted in
increased apoptosis in esophageal cancer cells. Antibodies
have been used to target fibronectin to reduce vascularization
and tenascin C to prevent metastasis (Ebbinghaus et al., 2004;
Reardon et al., 2006). Targeting Tenascin C together with
chemotherapy improves the survival of glioma patients
(Reardon et al., 2006). The inhibition of signaling pathways
perturbed in TME such as the IL-6-JAK-STAT and sonic
hedgehog cascade can aid in reducing CAFs and their secreted
factors, resulting in better therapy response (Olive et al., 2009).
For example, IPI-926, an inhibitor of the Hedgehog pathway,
together with Gemcitabine has been shown to influence resis-
tance to chemotherapy in PDAC (Olive et al., 2009). Currently,
most stromal-directed therapy is still in its infancy and removal
of any stromal elements must be studied carefully. Stromal
cells and their products such as the ECM are needed for normal
tissue architecture and homeostasis (Dzobo et al., 2012;
Senthebane et al., 2017, 2018; Sherman et al., 2014). Any
stromal-directed therapy will have to work in combination with
tumor-directed therapy to achieve durable cancer treatment.
Targeting CAF- or stromal-induced angiogenesis
Given the diverse cells and mechanisms through which
tumor vasculature is induced and develops, the inhibition of
tumor angiogenesis is challenging. Although angiogenesis
inhibition has been successful in some cases, partial to total
failure has also been reported in some studies (Bergers and
Hanahan, 2008; Ferrara and Adamis, 2016; Ferrara, 2016;
Jayson et al., 2016; Okaji et al., 2008; Patrikidou et al., 2014;
Saharinen et al., 2017; Schmittnaegel et al., 2017). Studies
demonstrated that tumor angiogenesis is regulated by several
factors and mechanisms and does not solely dependent on
VEGF-A signaling (Bergers and Hanahan, 2008; Ferrara,
2016; Potente et al., 2011). Several studies have also shown
the existence of tumor growth requiring no angiogenesis
(Pa
`ez-Ribes et al., 2009; Rubenstein et al., 2000).
Angiogenesis-independent tumor growth is also achieved
through the action of immune cells such as macrophages and
neutrophils (De Palma and Lewis, 2013; Gabrusiewicz et al.,
2014; Liang and Ferrara, 2016). Coupling anti-angiogenesis
specific therapy with strategies that disrupt any compensatory
factors and signaling from cells such as macrophages and
neutrophils offers better potential at inhibiting angiogenesis
in tumors (Baer et al., 2016; Biswas and Mantovani, 2010;
Lewis et al., 2016). Blockage or elimination of both macro-
phages and neutrophils that confer VEGF-A-independent
angiogenesis to tumors has been shown to enhance tumor
response to VEGF-A specific therapy (Rivera et al., 2015).
Kaneda et al. (2016) demonstrated that the inhibition of
PI3K signaling, highly expressed in myeloid cells, success-
fully inhibited angiogenesis in cancer treatment. Caution
must be taken, however, as several studies have shown that
the inhibition of angiogenesis may enhance the ability of
tumor-associated cells to promote tumor growth (Bergers and
Hanahan, 2008; De Palma and Lewis, 2013; Ferrara and
Adamis, 2016). Crawford et al. (2009) demonstrated that
some tumors can overcome angiogenesis inhibition through
upregulation of PDGF-C
Currently, Bevacizumab (anti-VEGF) is under clinical
trials for the treatment of different cancers from colorectal
cancer (NCT02885753), lung cancer (NCT03100955) to
cholangiocarcinoma cancer (NCT03251443) in combination
with 5-fluorouracil and panitumumab. Ramucirumab, also
known as LY3009806, is under clinical trial in combination
with paclitaxel for the treatment of gastric adenocarcinoma
(NCT02898077). RAD001 is also under clinical trial for the
treatment of renal cell carcinoma (NCT01206764).
CAFs as Delivery Vehicles of Therapeutic Agents
As described by Miao et al. (2017), CAFs can be used to
deliver anti-tumor drugs or nanoparticles and cause cancer
cell death via apoptosis. It has also been shown that fibro-
blasts within a tumor can scavenge and accumulate thera-
peutic drugs, resulting in increased drug concentrations
within tumors (Hessmann et al., 2018). Senthebane et al.
(2017) demonstrated that MSCs can be transformed into
CAFs and thus are a major source of CAFs found in tumors.
The authors and others demonstrated that although MSCs
initially demonstrate anti-tumor behavior they are easily
transformed into tumor-supporting cells (CAFs) when in
contact with cancer cells (Dzobo et al., 2016b; Shi et al., 2017).
Due to their likelihood of being found within tumors,
MSCs have been suggested as drug-delivery vehicles or can
be manipulated to secrete anti-tumor factors (Shi et al., 2017).
Amniotic MSCs have been suggested to deliver therapeutic
CAFSIN TUMOR MICROENVIRONMENT 327
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
agents to tumors (Bonomi et al., 2015). Several clinical trials
on the use of MSCs in gene therapies for different cancers are
underway (Abrate et al., 2014; Jobst et al., 2017; Niess et al.,
2015). Overall, the use of MSC-based gene therapies requires
further investigations as Senthebane et al. (2017) demon-
strated. The authors’ results show that MSCs can be pro-
tumorigenic or anti-tumorigenic depending on tumor stage
(Dzobo et al., 2016b; Senthebane et al., 2017). Other studies
support this dual effect of MSCs (Clarke et al., 2015; Hong
et al., 2014; Park et al., 2008).
Of late, CAFs are now being used as a stromal indicator of
the disease stage during diagnosis and as an indicator of tu-
mor response to treatment strategies. Tsujino et al. (2007)
demonstrated that the number of myofibroblasts within TME
can be used as a clinical biomarker of disease relapse in co-
lorectal cancer. Surowiak et al. (2007) showed that the pres-
ence of CAFs within breast cancer tissue is a poor prognostic
factor. Contrasting data on the prognostic value of CAFs in
tumors may be due to CAFs’ heterogeneity as well as the co-
evolution of CAFs with tumor cells during tumor progres-
sion (Brechbuhl et al., 2017; Paulsson and Micke, 2014).
Outlook and Conclusion
To summarize, we provide the definitions of the key con-
cepts in the present analysis in Box 1. Several lines of re-
search are being initiated to evaluate the effectiveness of
many therapeutic drugs on CAFs. Epigenetic drugs, includ-
ing histone deacetylase inhibitors, are under investigation
for their effect on several signaling pathways such as the
JAK-STAT pathway in both cancer cells and CAFs (Dzobo,
2019). These epigenetic drugs are being evaluated on their
potential to prevent CAFs’ generation and increase in abun-
dance within tumors (Younes et al., 2016). Another appealing
strategy includes the use of normal fibroblasts in cancer
treatment or the reversion of activated CAFs to normal fi-
broblasts (Kirsner et al., 2012).
Many questions remain to be answered regarding the
origin of CAFs or their subsets, as this can influence CAF-
directed therapeutic strategies adopted during cancer treat-
ment. It is hoped that as new information becomes available,
new markers and signaling pathways specific only to CAFs
or their subsets can be identified.
The identification that MSCs, besides fibroblasts, can be a
source of CAFs might explain CAFs’ heterogeneity and
could be the reason why several CAFs subsets are observed in
tumors (Senthebane et al., 2017). If different cells contribute
to the CAFs’ subsets observed in tumors, which cell(s) of
origin gives rise to pro-tumorigenic CAFs and which cell(s)
gives rise to anti-tumorigenic CAFs? Based on data revealed
by Senthebane et al. (2017), it appears that despite different
cell(s) of origin, stromal cells may start as anti-tumorigenic
but are transformed through several secreted factors to be-
come pro-tumorigenic. Whether different cell types con-
tribute to different subsets requires further investigation.
In addition, the identification of specific markers for
different subsets can allow retrospective determination of
cells of origin as well as the development of specific anti-
tumorigenic therapies against these CAFs’ subsets. Stromal
cells can also co-evolve with tumor cells. Is tumor cell het-
erogeneity also partly driven by different CAF subsets?
Epigenetic regulation of CAFs must also be investigated to
allow the development of drugs targeting epigenetics of
CAFs (Dzobo, 2019). Many clinical trials conducted for
different candidate drugs have focused on targeting cancer
cells and up to now no trial has been done on candidate drugs
targeting CAFs.
One major challenge faced by scientists during the de-
velopment of novel cancer therapies has been the absence of
adequate cancer models that recapitulate the TMEs as seen
in vivo to use in preclinical studies. This has prevented the
proper understanding of tumor cellstromal cell interactions
as well as cellmatrix interactions. For example, very few
available cancer models include cells such as CAFs, CAMs,
and cancer-associated neutrophils (Dzobo et al., 2018a). As
shown by recent reports, the addition of ECM proteins to
cancer models as well as the development of tumor spheroids
and organoids promises to reveal more about tumor initiation
and development than the use of cancer cells in in vitro ex-
periments (Dzobo et al., 2019).
New advances, including microfluidic technology, will
allow the development of what is known as ‘‘human organs
on chips.’’ With the ability to change parameters as required
during tumor development as well as potential additions of
Box 1. Definitions of the Key Concepts
in the Present Expert Review
Endothelial to mesenchymal transition: The process
through which endothelial cells are transformed into
mesenchymal cells. This is achieved through the loss
of endothelial properties and the gaining of
mesenchymal properties.
Epithelial to mesenchymal transition: The process
through which epithelial cells are transformed into
mesenchymal cells. This is achieved through the loss
of epithelial properties and the gaining of
mesenchymal properties.
Extracellular matrix (ECM): Fibrous proteins secreted by
stromal cells including fibroblasts, mesenchymal stem
cells, and pericytes and its main function is to provide
structural support and biochemical cues to surrounding
cells. The ECM accumulates growth factors,
cytokines, and chemokines and these, in turn, influence
tumor and stromal cell behavior.
Desmoplasia: The resulting state of tissue after a
response to an injury and includes fibrosis as stromal
cells synthesize huge amounts of ECM proteins.
Matrix metalloproteases: A large family of zinc-
containing protease enzymes referred to as
endopeptidases and are responsible for degrading
ECM proteins such as fibronectin, collagens, and
laminins.
Tumor microenvironment: Includes all cellular and
noncellular components of the environment
surrounding/within a tumor and includes all stromal
cells, blood vessels, and the ECM. Stromal cells
include cancer-associated fibroblasts, cancer-
associated macrophages, cancer-associated
neutrophils, pericytes, and other immune cells. The
tumor microenvironment (TME) provides the
‘‘theater’’ within which tumor cells can survive and
tumors develop and eventually spread.
Stromal cells: Includes all cells found within the TME
and act to support tumor cells by secreting tumor-
promoting growth factors, cytokines, chemokines, and
ECM proteins.
328 DZOBO AND DANDARA
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
several cells, including CAFs, these new cancer models will
potentially reveal accurate information about tumor initiation
and development. In addition, microfluidic technology as
applied to tumor development will also allow collection of
samples at specific times with ease.
Author Disclosure Statement
The authors declare they have no conflicting financial
interests.
Funding Information
The funding for this research was provided by the National
Research Foundation (NRF) of South Africa (Grant no.
91457: RCA13101656402), the International Centre for
Genetic Engineering and Biotechnology (ICGEB) (Grant no.
2015/0001), and the University of Cape Town.
References
Abrate A, Buono R, Canu T, et al. (2014). Mesenchymal stem
cells expressing therapeutic genes induce autochthonous
prostate tumour regression. Eur J Cancer 50, 2478–2488.
Akrish SJ, Rachmiel A, Sabo E, Vered M, and Ben-Izhak O.
(2016). Cancer associated fibroblasts are an infrequent finding
in the microenvironment of proliferative verrucous leuko-
plakia associated squamous cell carcinoma. J Oral Pathol
Med 46, 353–358.
Al-Ansari M, Hendrayani S, Shehata A, and Aboussekhra A.
(2013). p16 INK4A Represses the paracrine tumor-promoting
effects of breast stromal fibroblasts. Oncogene 32, 2356.
Albrengues J, Bertero T, Grasset E, et al. (2015). Epigenetic
switch drives the conversion of fibroblasts into proinvasive
cancer-associated fibroblasts. Nat Commun 6, 10204.
Anderberg C, Li H, Fredriksson L, et al. (2009). Paracrine
signaling by platelet-derived growth factor-CC promotes tu-
mor growth by recruitment of cancer-associated fibroblasts.
Cancer Res 69, 369–378.
Armulik A, Genove
´G, and Betsholtz C. (2011). Pericytes:
Developmental, physiological, and pathological perspectives,
problems, and promises. Dev Cell 21, 193–215.
Arnold JN, Magiera L, Kraman M, and Fearon DT. (2014).
Tumoral immune suppression by macrophages expressing
fibroblast activation protein-aand heme oxygenase-1. Cancer
Immunol Res 2, 121–126.
Arumugam T, Ramachandran V, Fournier KF, et al. (2009).
Epithelial to mesenchymal transition contributes to drug re-
sistance in pancreatic cancer. Cancer Res 69, 5820–5828.
Ayala G, Tuxhorn JA, Wheeler TM, et al. (2003). Reactive
stroma as a predictor of biochemical-free recurrence in
prostate cancer. Clin Cancer Res 9, 4792–4801.
Baer C, Squadrito ML, Laoui D, et al. (2016). Suppression of
microRNA activity amplifies IFN-c-induced macrophage ac-
tivation and promotes anti-tumour immunity. Nat Cell Biol
18, 790.
Baeriswyl V, and Christofori G. (2009). The angiogenic switch
in carcinogenesis. Semin Cancer Biol 19, 329–337.
Barbazan J, and Matic Vignjevic D. (2019). Cancer associated
fibroblasts: Is the force the path to the dark side? Curr Opin
Cell Biol 56, 71–79.
Barker HE, Paget JT, Khan AA, and Harrington KJ. (2015). The
tumour microenvironment after radiotherapy: Mechanisms of
resistance and recurrence. Nat Rev Cancer 15, 409–425.
Barth PJ, Ebrahimsade S, Ramaswamy A, and Moll R. (2002).
CD34+fibrocytes in invasive ductal carcinoma, ductal car-
cinoma in situ, and benign breast lesions. Virchows Arch 440,
298–303.
Bauer M, Su G, Casper C, He R, Rehrauer W, and Friedl A.
(2010). Heterogeneity of gene expression in stromal fibro-
blasts of human breast carcinomas and normal breast. On-
cogene 29, 1732.
Becker C, Fantini MC, Schramm C, et al. (2004). TGF-beta
suppresses tumor progression in colon cancer by inhibition of
IL-6 trans-signaling. Immunity 21, 491–501.
Begicevic R-R, and Falasca M. (2017). ABC transporters in
cancer stem cells: Beyond chemoresistance. Int J Mol Sci 18,
2362.
Bergers G, and Benjamin LE. (2003). Angiogenesis: Tumor-
igenesis and the angiogenic switch. Nat Rev Cancer 3, 401.
Bergers G, and Hanahan D. (2008). Modes of resistance to anti-
angiogenic therapy. Nat Rev Cancer 8, 592–603.
Biswas SK, and Mantovani A. (2010). Macrophage plasticity
and interaction with lymphocyte subsets: Cancer as a para-
digm. Nat Immunol 11, 889.
Bluff J, Menakuru S, Cross S, et al. (2009). Angiogenesis is
associated with the onset of hyperplasia in human ductal
breast disease. Br J Cancer 101, 666.
Bonnans C, Chou J, and Werb Z. (2014). Remodelling the ex-
tracellular matrix in development and disease. Nat Rev Mol
Cell Biol 15, 786–801.
Bonomi A, Silini A, Vertua E, et al. (2015). Human amniotic
mesenchymal stromal cells (hAMSCs) as potential vehicles
for drug delivery in cancer therapy: An in vitro study. Stem
Cell Res Ther 6, 155.
Borriello L, Nakata R, Sheard MA, et al. (2017). Cancer-
associated fibroblasts share characteristics and protumori-
genic activity with mesenchymal stromal cells. Cancer Res
77, 5142–5157.
Bossi P, Viale G, Lee AK, Alfano R, Coggi G, and Bosari S.
(1995). Angiogenesis in colorectal tumors: Microvessel
quantitation in adenomas and carcinomas with clinicopatho-
logical correlations. Cancer Res 55, 5049–5053.
Bournazou E, and Bromberg J. (2013). Targeting the tumor
microenvironment: JAK-STAT3 signaling. JAK-STAT 2,
e23828.
Bray F. (2016). The evolving scale and profile of cancer
worldwide: Much Ado About Everything. Cancer Epidemiol
Biomarkers Prev 25, 3–5.
Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, and
Jemal A. (2018). Global cancer statistics 2018: GLOBOCAN
estimates of incidence and mortality worldwide for 36 can-
cers in 185 countries. CA Cancer J Clin 68, 394–424.
Bray F, Jemal A, Grey N, Ferlay J, and Forman D. (2012).
Global cancer transitions according to the Human Develop-
ment Index (2008–2030): A population-based study. Lancet
Oncol 13, 790–801.
Bray F, and Soerjomataram I. (2015). The Changing Global
Burden of Cancer: Transitions in Human Development and
Implications for Cancer Prevention and Control. In: Cancer:
Disease Control Priorities, Third Edition (Volume 3). Gel-
band H, Jha P, Sankaranarayanan R, and Horton S, eds.
Washington, DC: The International Bank for Reconstruction
and Development/The World Bank (c) 2015 International
Bank for Reconstruction and Development/The World Bank.
Brechbuhl HM, Finlay-Schultz J, Yamamoto TM, et al. (2017).
Fibroblast subtypes regulate responsiveness of luminal breast
cancer to estrogen. Clin Cancer Res 23, 1710–1721.
CAFSIN TUMOR MICROENVIRONMENT 329
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
Bromberg J. (2002). Stat proteins and oncogenesis. J Clin Invest
109, 1139–1142.
Brunen D, Willems SM, Kellner U, Midgley R, Simon I, and
Bernards R. (2013). TGF-beta: An emerging player in drug
resistance. Cell Cycle 12, 2960–2968.
Bruzzese F, Ha
¨gglo
¨f C, Leone A, et al. (2014). Local and
systemic protumorigenic effects of cancer-associated
fibroblast-derived GDF15. Cancer Res 74, 3408–3417.
Bussard KM, Mutkus L, Stumpf K, Gomez-Manzano C, and
Marini FC. (2016). Tumor-associated stromal cells as key
contributors to the tumor microenvironment. Breast Cancer
Res 18, 84.
Bussolino F, Ziche M, Wang JM, et al. (1991). In vitro and
in vivo activation of endothelial cells by colony-stimulating
factors. J Clin Invest 87, 986–995.
Calon A, Espinet E, Palomo-Ponce S, et al. (2012). Dependency
of colorectal cancer on a TGF-b-driven program in stromal
cells for metastasis initiation. Cancer Cell 22, 571–584.
Calon A, Tauriello D, and Batlle E. (2014). TGF-beta in CAF-
mediated tumor growth and metastasis. Semin Cancer Biol
25, 15–22.
Calvo F, Ege N, Grande-Garcia A, et al. (2013). Mechan-
otransduction and YAP-dependent matrix remodelling is re-
quired for the generation and maintenance of cancer-
associated fibroblasts. Nat Cell Biol 15, 637.
Campbell I, Qiu W, and Haviv I. (2011). Genetic changes in
tumour microenvironments. J Pathol 223, 450–458.
Campbell IL, Erta M, Lim SL, et al. (2014). Trans-signaling is a
dominant mechanism for the pathogenic actions of
interleukin-6 in the brain. J Neurosci 34, 2503–2513.
Carapuc¸a EF, Gemenetzidis E, Feig C, et al. (2016). Anti-
stromal treatment together with chemotherapy targets multi-
ple signalling pathways in pancreatic adenocarcinoma.
J Pathol 239, 286–296.
Carmeliet P, and Jain RK. (2011). Molecular mechanisms and
clinical applications of angiogenesis. Nature 473, 298.
Casey TM, Eneman J, Crocker A, et al. (2008). Cancer asso-
ciated fibroblasts stimulated by transforming growth factor
beta1 (TGF-b1) increase invasion rate of tumor cells: A
population study. Breast Cancer Res Treat 110, 39–49.
Castello LM, Raineri D, Salmi L, et al. (2017). Osteopontin at
the crossroads of inflammation and tumor progression.
Mediators Inflamm 2017, 4049098.
Castells M, Thibault B, Delord JP, and Couderc B. (2012).
Implication of tumor microenvironment in chemoresistance:
Tumor-associated stromal cells protect tumor cells from cell
death. Int J Mol Sci 13, 9545–9571.
Cavarretta IT, Neuwirt H, Zaki MH, et al. (2008). Mcl-1 is
regulated by IL-6 and mediates the survival activity of the
cytokine in a model of late stage prostate carcinoma. Adv Exp
Med Biol 617, 547–555.
Chaffer CL, and Weinberg RA. (2011). A perspective on cancer
cell metastasis. Science 331, 1559–1564.
Chan TS, Shaked Y, and Tsai KK. (2019). Targeting the in-
terplay between cancer fibroblasts, mesenchymal stem cells,
and cancer stem cells in desmoplastic cancers. Front Oncol 9,
688.
Chang Q, Bournazou E, Sansone P, et al. (2013). The IL-6/JAK/
Stat3 feed-forward loop drives tumorigenesis and metastasis.
Neoplasia 15, 848–862.
Chargari C, Clemenson C, Martins I, Perfettini J-L, and Deutsch
E. (2013). Understanding the functions of tumor stroma in
resistance to ionizing radiation: Emerging targets for phar-
macological modulation. Drug Resist Updat 16, 10–21.
Chauhan VP, Martin JD, Liu H, et al. (2013). Angiotensin in-
hibition enhances drug delivery and potentiates chemotherapy
by decompressing tumour blood vessels. Nat Commun 4,
2516.
Chen CL, Loy A, Cen L, et al. (2007). Signal transducer and
activator of transcription 3 is involved in cell growth and
survival of human rhabdomyosarcoma and osteosarcoma
cells. BMC Cancer 7, 111.
Chen MF, Chen PT, Lu MS, Lin PY, Chen WC, and Lee KD.
(2013a). IL-6 expression predicts treatment response and
outcome in squamous cell carcinoma of the esophagus. Mol
Cancer 12, 26.
Chen Y, Wang J, Wang X, et al. (2013b). STAT3, a poor sur-
vival predicator, is associated with lymph node metastasis
from breast cancer. J Breast Cancer 16, 40–49.
Cheteh EH, Augsten M, Rundqvist H, et al. (2017). Human
cancer-associated fibroblasts enhance glutathione levels and
antagonize drug-induced prostate cancer cell death. Cell
Death Dis 8, e2848.
Chiappori AA, Eckhardt SG, Bukowski R, et al. (2007).
A phase I pharmacokinetic and pharmacodynamic study of
s-3304, a novel matrix metalloproteinase inhibitor, in patients
with advanced and refractory solid tumors. Clin Cancer Res
13, 2091–2099.
Choi S-Y, Sung R, Lee S-J, et al. (2013). Podoplanin, a-smooth
muscle actin or S100A4 expressing cancer-associated fibro-
blasts are associated with different prognosis in colorectal
cancers. J Korean Med Sci 28, 1293–1301.
Chow MT, and Luster AD. (2014). Chemokines in cancer.
Cancer Immunol Res 2, 1125–1131.
Chung K-M, Hsu S-C, Chu Y-R, et al. (2014). Fibroblast acti-
vation protein (FAP) is essential for the migration of bone
marrow mesenchymal stem cells through RhoA activation.
PLoS One 9, e88772.
Chung YC, and Chang YF. (2003). Serum interleukin-6 levels
reflect the disease status of colorectal cancer. J Surg Oncol
83, 222–226.
Clarke MR, Imhoff FM, and Baird SK. (2015). Mesenchymal
stem cells inhibit breast cancer cell migration and invasion
through secretion of tissue inhibitor of metalloproteinase-1
and-2. Mol Carcinog 54, 1214–1219.
Codd AS, Kanaseki T, Torigo T, and Tabi Z. (2018). Cancer
stem cells as targets for immunotherapy. Immunology 153,
304–314.
Cohen AW, Hnasko R, Schubert W, and Lisanti MP. (2004).
Role of caveolae and caveolins in health and disease. Physiol
Rev 84, 1341–1379.
Colak S, and Ten Dijke P. (2017). Targeting TGF-beta signal-
ing in cancer. Trends Cancer 3, 56–71.
Comito G, Giannoni E, Segura C, et al. (2014). Cancer-associated
fibroblasts and M2-polarized macrophages synergize during
prostate carcinoma progression. Oncogene 33, 2423.
Coussens LM, Fingleton B, and Matrisian LM. (2002). Matrix
metalloproteinase inhibitors and cancer—Trials and tribula-
tions. Science 295, 2387–2392.
Crawford Y, Kasman I, Yu L, et al. (2009). PDGF-C mediates
the angiogenic and tumorigenic properties of fibroblasts as-
sociated with tumors refractory to anti-VEGF treatment.
Cancer Cell 15, 21–34.
Crunkhorn S. (2018). Preventing metastatic relapse. Nat Rev
Drug Discov 17, 394.
Cukierman E, and Bassi DE. (2010). Physico-mechanical as-
pects of extracellular matrix influences on tumorigenic be-
haviors. Semin Cancer Biol 20, 139–145.
330 DZOBO AND DANDARA
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
Culig Z, and Puhr M. (2012). Interleukin-6: A multifunctional
targetable cytokine in human prostate cancer. Mol Cell En-
docrinol 360, 52–58.
Dai J, Peng L, Fan K, et al. (2009). Osteopontin induces an-
giogenesis through activation of PI3K/AKT and ERK1/2 in
endothelial cells. Oncogene 28, 3412–3422.
Damiano JS, Hazlehurst LA, and Dalton WS. (2001). Cell
adhesion-mediated drug resistance (CAM-DR) protects the
K562 chronic myelogenous leukemia cell line from apoptosis
induced by BCR/ABL inhibition, cytotoxic drugs, and
gamma-irradiation. Leukemia 15, 1232–1239.
Danø K, Behrendt N, Høyer-Hansen G, et al. (2005). Plasmi-
nogen activation and cancer. Thromb Haemost 93, 676–681.
Darby IA, and Hewitson TD. (2007). Fibroblast differentiation
in wound healing and fibrosis. Int Rev Cytol 257, 143–179.
Dawood S, Austin L, and Cristofanilli M. (2014). Cancer stem
cells: Implications for cancer therapy. Oncology (Williston
Park) 28, 1101–1107, 1110.
De Palma M, Biziato D, and Petrova TV. (2017). Micro-
environmental regulation of tumour angiogenesis. Nat Rev
Cancer 17, 457–474.
De Palma M, and Lewis CE. (2013). Macrophage regulation of
tumor responses to anticancer therapies. Cancer Cell 23,
277–286.
De Wever O, Demetter P, Mareel M, and Bracke M. (2008).
Stromal myofibroblasts are drivers of invasive cancer growth.
Int J Cancer 123, 2229–2238.
Deroanne CF, Hajitou A, Calberg-Bacq CM, Nusgens BV, and
Lapiere CM. (1997). Angiogenesis by fibroblast growth factor
4 is mediated through an autocrine up-regulation of vascular
endothelial growth factor expression. Cancer Res 57, 5590–
5597.
Deryugina EI, and Quigley JP. (2015). Tumor angiogenesis:
MMP-mediated induction of intravasation- and metastasis-
sustaining neovasculature. Matrix Biol 44–46, 94–112.
Dethlefsen C, Højfeldt G, and Hojman P. (2013). The role of
intratumoral and systemic IL-6 in breast cancer. Breast
Cancer Res Treat 138, 657–664.
Dijkgraaf EM, Santegoets SJ, Reyners AK, et al. (2015).
A phase I trial combining carboplatin/doxorubicin with to-
cilizumab, an anti-IL-6R monoclonal antibody, and
interferon-alpha2b in patients with recurrent epithelial ovar-
ian cancer. Ann Oncol 26, 2141–2149.
Ding L, Zhang Z, Shang D, et al. (2014). a-Smooth muscle
actin-positive myofibroblasts, in association with epithelial–
mesenchymal transition and lymphogenesis, is a critical
prognostic parameter in patients with oral tongue squamous
cell carcinoma. J Oral Pathol Med 43, 335–343.
Doherty GJ, Tempero M, and Corrie PG. (2018). HALO-109-
301: A Phase III trial of PEGPH20 (with gemcitabine and
nab-paclitaxel) in hyaluronic acid-high stage IV pancreatic
cancer. Future Oncol 14, 13–22.
Domenichini A, Adamska A, and Falasca M. (2019). ABC
transporters as cancer drivers: Potential functions in cancer
development. Biochim Biophys Acta 1863, 52–60.
Dorritie K, Mccubrey J, and Johnson D. (2014). STAT tran-
scription factors in hematopoiesis and leukemogenesis: Op-
portunities for therapeutic intervention. Leukemia 28, 248.
Dulauroy S, Di Carlo SE, Langa F, Eberl G, and Peduto L.
(2012). Lineage tracing and genetic ablation of ADAM12+
perivascular cells identify a major source of profibrotic cells
during acute tissue injury. Nat Med 18, 1262.
Duluc C, Moatassim-Billah S, Chalabi-Dchar M, et al. (2015).
Pharmacological targeting of the protein synthesis mTOR/
4E-BP1 pathway in cancer-associated fibroblasts abrogates
pancreatic tumour chemoresistance. EMBO Mol Med 7,
735–753.
Dvorak HF. (1986). Tumors: Wounds that do not heal. N Engl J
Med 315, 1650–1659.
Dzobo K. (2019). Epigenomics-guided drug development: Re-
cent advances in solving the cancer treatment ‘‘jigsaw puz-
zle’’. OMICS 23, 70–85.
Dzobo K, Leaner VD, and Parker MI. (2012). Feedback regu-
lation of the alpha2(1) collagen gene via the Mek-Erk sig-
naling pathway. IUBMB Life 64, 87–98.
Dzobo K, Leaner VD, and Parker MI. (2014). Absence of
feedback regulation in the synthesis of COL1A1. Life Sci
103, 25–33.
Dzobo K, Motaung K, and Adesida A. (2019). Recent trends in
decellularized extracellular matrix bioinks for 3D printing:
An updated review. Int J Mol Sci 20, pii: E4628.
Dzobo K, Rowe A, Senthebane DA, Almazyadi MAM, Patten
V, and Parker MI. (2018a). Three-dimensional organoids in
cancer research: The search for the holy grail of preclinical
cancer modeling. OMICS 22, 733–748.
Dzobo K, Senthebane DA, Rowe A, et al. (2016a). Cancer stem
cell hypothesis for therapeutic innovation in clinical oncolo-
gy? Taking the root out, not chopping the leaf. OMICS 20,
681–691.
Dzobo K, Senthebane DA, Thomford NE, Rowe A, Dandara C,
and Parker MI. (2018b). Not everyone fits the mold: in-
tratumor and intertumor heterogeneity and innovative cancer
drug design and development. OMICS 22, 17–34.
Dzobo K, Vogelsang M, Thomford NE, et al. (2016b).
Wharton’s jelly-derived mesenchymal stromal cells and
fibroblast-derived extracellular matrix synergistically acti-
vate apoptosis in a p21-dependent mechanism in WHCO1
and MDA MB 231 cancer cells in vitro. Stem Cells Int 2016,
4842134.
Ebbing EA, Van Der Zalm AP, Steins A, et al. (2019). Stromal-
derived interleukin 6 drives epithelial-to-mesenchymal tran-
sition and therapy resistance in esophageal adenocarcinoma.
Proc Natl Acad Sci U S A 116, 2237–2242.
Ebbinghaus C, Scheuermann J, Neri D, and Elia G. (2004).
Diagnostic and therapeutic applications of recombinant anti-
bodies: Targeting the extra-domain B of fibronectin, a marker
of tumor angiogenesis. Curr Pharm Des 10, 1537–1549.
Egeblad M, Rasch MG, and Weaver VM. (2010). Dynamic
interplay between the collagen scaffold and tumor evolution.
Curr Opin Cell Biol 22, 697–706.
Egeblad M, and Werb Z. (2002). New functions for the matrix
metalloproteinases in cancer progression. Nat Rev Cancer
2, 161.
Eke I, and Cordes N. (2011). Radiobiology goes 3D: How ECM
and cell morphology impact on cell survival after irradiation.
Radiother Oncol 99, 271–278.
Ene–Obong A, Clear AJ, Watt J, et al. (2013). Activated pan-
creatic stellate cells sequester CD8+T cells to reduce their
infiltration of the juxtatumoral compartment of pancreatic
ductal adenocarcinoma. Gastroenterology 145, 1121–1132.
Erdogan B, and Webb DJ. (2017). Cancer-associated fibroblasts
modulate growth factor signaling and extracellular matrix
remodeling to regulate tumor metastasis. Biochem Soc Trans
45, 229–236.
Erez N, Truitt M, Olson P, and Hanahan D. (2010). Cancer-
associated fibroblasts are activated in incipient neoplasia to
orchestrate tumor-promoting inflammation in an NF-jB-
dependent manner. Cancer Cell 17, 135–147.
CAFSIN TUMOR MICROENVIRONMENT 331
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
Erkan M, Kleeff J, Gorbachevski A, et al. (2007). Periostin
creates a tumor-supportive microenvironment in the pancreas
by sustaining fibrogenic stellate cell activity. Gastro-
enterology 132, 1447–1464.
Eyden B. (2008). The myofibroblast: Phenotypic characteriza-
tion as a prerequisite to understanding its functions in trans-
lational medicine. J Cell Mol Med 12, 22–37.
Fang T, Lv H, Lv G, et al. (2018). Tumor-derived exosomal
miR-1247-3p induces cancer-associated fibroblast activation
to foster lung metastasis of liver cancer. Nat Commun 9, 191.
Fearon D. (2015). Inhibition of cxcr4 signaling in cancer
immunotherapy. Patent Application Publication. US 2015/
0216843A1.
Feig C, Jones JO, Kraman M, et al. (2013). Targeting CXCL12
from FAP-expressing carcinoma-associated fibroblasts sy-
nergizes with anti–PD-L1 immunotherapy in pancreatic
cancer. Proc Natl Acad Sci U S A 110, 20212–20217.
Ferlay J, Colombet M, Soerjomataram I, et al. (2019). Esti-
mating the global cancer incidence and mortality in 2018:
GLOBOCAN sources and methods. Int J Cancer 144, 1941–
1953.
Ferrara N. (2016). VEGF and intraocular neovascularization:
From discovery to therapy. Transl Vis Sci Technol 5, 10.
Ferrara N, and Adamis AP. (2016). Ten years of anti-vascular
endothelial growth factor therapy. Nat Rev Drug Discov 15,
385.
Fidler MM, and Bray F. (2018). Global cancer inequalities.
Front Oncol 8, 293.
Fidler MM, Soerjomataram I, and Bray F. (2016). A global
view on cancer incidence and national levels of the human
development index. Int J Cancer 139, 2436–2446.
Fisher DT, Appenheimer MM, and Evans SS. (2014). The two
faces of IL-6 in the tumor microenvironment. Semin Im-
munol 26, 38–47.
Fizazi K, De Bono JS, Flechon A, et al. (2012). Randomised
phase II study of siltuximab (CNTO 328), an anti-IL-6
monoclonal antibody, in combination with mitoxantrone/
prednisone versus mitoxantrone/prednisone alone in meta-
static castration-resistant prostate cancer. Eur J Cancer 48,
85–93.
Flavell RA, Sanjabi S, Wrzesinski SH, and Licona-Limo
´n
P. (2010). The polarization of immune cells in the tumour
environment by TGFb. Nat Rev Immunol 10, 554.
Frank DA. (2007). STAT3 as a central mediator of neoplastic
cellular transformation. Cancer Lett 251, 199–210.
Friedl P, and Alexander S. (2011). Cancer invasion and the
microenvironment: Plasticity and reciprocity. Cell 147,
992–1009.
Froeling FE, Feig C, Chelala C, et al. (2011). Retinoic acid–
induced pancreatic stellate cell quiescence reduces paracrine
Wnt–b-catenin signaling to slow tumor progression. Gastro-
enterology 141, 1486–1497.e1414.
Fuh B, Sobo M, Cen L, et al. (2009). LLL-3 inhibits STAT3
activity, suppresses glioblastoma cell growth and prolongs
survival in a mouse glioblastoma model. Br J Cancer 100,
106–112.
Fukumura D, Xavier R, Sugiura T, et al. (1998). Tumor in-
duction of VEGF promoter activity in stromal cells. Cell 94,
715–725.
Fukumura D, Yuan F, Monsky WL, Chen Y, and Jain RK.
(1997). Effect of host microenvironment on the microcircula-
tion of human colon adenocarcinoma. Am J Pathol 151, 679.
Gabrusiewicz K, Liu D, Cortes-Santiago N, et al. (2014). Anti-
vascular endothelial growth factor therapy-induced glioma
invasion is associated with accumulation of Tie2-expressing
monocytes. Oncotarget 5, 2208.
Gaggioli C, Hooper S, Hidalgo-Carcedo C, et al. (2007).
Fibroblast-led collective invasion of carcinoma cells with
differing roles for RhoGTPases in leading and following
cells. Nat Cell Biol 9, 1392.
Ganesh K, and Massague J. (2018). TGF-beta inhibition and
immunotherapy: Checkmate. Immunity 48, 626–628.
Gao H, Priebe W, Glod J, and Banerjee D. (2009). Activation of
signal transducers and activators of transcription 3 and focal
adhesion kinase by stromal cell-derived factor 1 is required
for migration of human mesenchymal stem cells in response
to tumor cell-conditioned medium. Stem Cells 27, 857–865.
Gao J, Zhao S, and Halstensen TS. (2016). Increased
interleukin-6 expression is associated with poor prognosis
and acquired cisplatin resistance in head and neck squamous
cell carcinoma. Oncol Rep 35, 3265–3274.
Gao SP, Mark KG, Leslie K, et al. (2007). Mutations in the
EGFR kinase domain mediate STAT3 activation via IL-6
production in human lung adenocarcinomas. J Clin Invest
117, 3846–3856.
Gerber H-P, Kowalski J, Sherman D, Eberhard DA, and Ferrara
N. (2000). Complete inhibition of rhabdomyosarcoma xeno-
graft growth and neovascularization requires blockade of both
tumor and host vascular endothelial growth factor. Cancer
Res 60, 6253–6258.
Giannoni E, Bianchini F, Masieri L, et al. (2010). Reciprocal
activation of prostate cancer cells and cancer-associated fi-
broblasts stimulates epithelial-mesenchymal transition and
cancer stemness. Cancer Res 70, 6945–6956.
Glentis A, Oertle P, Mariani P, et al. (2017). Cancer-associated
fibroblasts induce metalloprotease-independent cancer cell
invasion of the basement membrane. Nat Commun 8, 924.
Goetz JG, Minguet S, Navarro-Le
´rida I, et al. (2011). Bio-
mechanical remodeling of the microenvironment by stromal
caveolin-1 favors tumor invasion and metastasis. Cell 146,
148–163.
Gorski A, Castronovo V, Stepien-Sopniewska B, et al. (1994).
Depressed immune surveillance against cancer: Role of de-
ficient T cell: Extracellular matrix interactions. Cell Adhes
Commun 2, 225–233.
Grivennikov S, Karin E, Terzic J, et al. (2009). IL-6 and Stat3
are required for survival of intestinal epithelial cells and
development of colitis-associated cancer. Cancer Cell 15,
103–113.
Grugan KD, Miller CG, Yao Y, et al. (2010). Fibroblast-
secreted hepatocyte growth factor plays a functional role in
esophageal squamous cell carcinoma invasion. Proc Natl
Acad Sci U S A 107, 11026–11031.
Grum-Schwensen B, Klingelhofer J, Berg CH, et al. (2005).
Suppression of tumor development and metastasis formation
in mice lacking the S100A4 (mts1) gene. Cancer Res 65,
3772–3780.
Gu S, and Feng XH. (2018). TGF-beta signaling in cancer. Acta
Biochim Biophys Sin (Shanghai) 50, 941–949.
Guaita S, Puig I, Francı
´C, et al. (2002). Snail induction of
epithelial to mesenchymal transition in tumor cells is ac-
companied by MUC1 repression andZEB1 expression. J Biol
Chem 277, 39209–39216.
Guido C, Whitaker-Menezes D, Capparelli C, et al. (2012).
Metabolic reprogramming of cancer-associated fibroblasts by
TGF-bdrives tumor growth: Connecting TGF-bsignaling
with ‘‘Warburg-like’’ cancer metabolism and L-lactate pro-
duction. Cell Cycle 11, 3019–3035.
332 DZOBO AND DANDARA
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
Guo M, Ehrlicher AJ, Mahammad S, et al. (2013). The role of
vimentin intermediate filaments in cortical and cytoplasmic
mechanics. Biophys J 105, 1562–1568.
Guo S, and Deng C-X. (2018). Effect of stromal cells in tumor
microenvironment on metastasis initiation. Int J Biol Sci 14,
2083–2093.
Gupta GP, and Massague
´J. (2006). Cancer metastasis: Building
a framework. Cell 127, 679–695.
Gutcher I, Donkor MK, Ma Q, Rudensky AY, Flavell RA, and
Li MO. (2011). Autocrine transforming growth factor-b1
promotes in vivo Th17 cell differentiation. Immunity 34,
396–408.
Hanahan D, and Weinberg RA. (2011). Hallmarks of cancer:
The next generation. Cell 144, 646–674.
Hao N-B, Lu
¨M-H, Fan Y-H, Cao Y-L, Zhang Z-R, and Yang
S-M. (2012). Macrophages in tumor microenvironments and
the progression of tumors. Clin Dev Immunol 2012, 948098.
Hao Y, Baker D, and Ten Dijke P. (2019). TGF-beta-mediated
epithelial-mesenchymal transition and cancer metastasis. Int J
Mol Sci 20, pii: E2767.
Harris TJ, Grosso JF, Yen H-R, et al. (2007). Cutting edge: An
in vivo requirement for STAT3 signaling in TH17 develop-
ment and TH17-dependent autoimmunity. J Immunol 179,
4313–4317.
Hashimoto O, Yoshida M, Koma YI, et al. (2016). Collabora-
tion of cancer-associated fibroblasts and tumour-associated
macrophages for neuroblastoma development. J Pathol 240,
211–223.
Hawinkels L, Paauwe M, Verspaget H, et al. (2014). Interaction
with colon cancer cells hyperactivates TGF-bsignaling in
cancer-associated fibroblasts. Oncogene 33, 97.
Hazlehurst LA, Damiano JS, Buyuksal I, Pledger WJ, and
Dalton WS. (2000). Adhesion to fibronectin via beta1 in-
tegrins regulates p27kip1 levels and contributes to cell ad-
hesion mediated drug resistance (CAM-DR). Oncogene 19,
4319–4327.
Hedvat M, Huszar D, Herrmann A, et al. (2009). The JAK2
inhibitor AZD1480 potently blocks Stat3 signaling and on-
cogenesis in solid tumors. Cancer Cell 16, 487–497.
Hessmann E, Patzak M, Klein L, et al. (2018). Fibroblast drug
scavenging increases intratumoural gemcitabine accumula-
tion in murine pancreas cancer. Gut 67, 497–507.
Hidalgo M. (2010). Pancreatic cancer. N Engl J Med 362,
1605–1617.
Hingorani SR, Harris WP, Beck JT, et al. (2016). Phase Ib study
of PEGylated recombinant human hyaluronidase and gemci-
tabine in patients with advanced pancreatic cancer. Clin
Cancer Res 22, 2848–2854.
Hong I-S, Lee H-Y, and Kang K-S. (2014). Mesenchymal stem
cells and cancer: Friends or enemies? Mutat Res 768, 98–106.
Hotz B, Arndt M, Dullat S, Bhargava S, Buhr H-J, and Hotz
HG. (2007). Epithelial to mesenchymal transition: Expression
of the regulators snail, slug, and twist in pancreatic cancer.
Clin Cancer Res 13, 4769–4776.
Houthuijzen JM, and Jonkers J. (2018). Cancer-associated fi-
broblasts as key regulators of the breast cancer tumor mi-
croenvironment. Cancer Metastasis Rev 37, 577–597.
Huber MA, Schubert RD, Peter RU, et al. (2003). Fibroblast
activation protein: Differential expression and serine protease
activity in reactive stromal fibroblasts of melanocytic skin
tumors. J Invest Dermatol 120, 182–188.
Huber RM, Lucas JM, Gomez-Sarosi LA, et al. (2015). DNA
damage induces GDNF secretion in the tumor microenvi-
ronment with paracrine effects promoting prostate cancer
treatment resistance. Oncotarget 6, 2134–2147.
Huelsken J, and Hanahan D. (2018). A subset of cancer-
associated fibroblasts determines therapy resistance. Cell 172,
643–644.
Hunter CA, and Jones SA. (2015). IL-6 as a keystone cytokine
in health and disease. Nat Immunol 16, 448.
Huynh J, Etemadi N, Hollande F, Ernst M, and Buchert M.
(2017). The JAK/STAT3 axis: A comprehensive drug target
for solid malignancies. Semin Cancer Biol 45, 13–22.
Hwang RF, Moore T, Arumugam T, et al. (2008). Cancer-
associated stromal fibroblasts promote pancreatic tumor
progression. Cancer Res 68, 918–926.
Iida N, Dzutsev A, Stewart CA, et al. (2013). Commensal
bacteria control cancer response to therapy by modulating the
tumor microenvironment. Science 342, 967–970.
Ishii G, Ochiai A, and Neri S. (2016). Phenotypic and functional
heterogeneity of cancer-associated fibroblast within the tumor
microenvironment. Adv Drug Deliv Rev 99, 186–196.
Iwano M, Plieth D, Danoff TM, Xue C, Okada H, and Neilson
EG. (2002). Evidence that fibroblasts derive from epithelium
during tissue fibrosis. J Clin Invest 110, 341–350.
Iwata-Kajihara T, Sumimoto H, Kawamura N, et al. (2011).
Enhanced cancer immunotherapy using STAT3-depleted
dendritic cells with high Th1-inducing ability and resistance
to cancer cell-derived inhibitory factors. J Immunol 187,
27–36.
Jabłon
´ska-Trypuc
´A,MatejczykM,andRosochackiS.
(2016). Matrix metalloproteinases (MMPs), the main ex-
tracellular matrix (ECM) enzymes in collagen degradation,
as a target for anticancer drugs. J Enzyme Inhib Med Chem
31, 177–183.
Jacobetz MA, Chan DS, Neesse A, et al. (2013). Hyaluronan
impairs vascular function and drug delivery in a mouse model
of pancreatic cancer. Gut 62, 112–120.
Jakowlew SB. (2006). Transforming growth factor-beta in
cancer and metastasis. Cancer Metastasis Rev 25, 435–457.
Jayson GC, Kerbel R, Ellis LM, and Harris AL. (2016). Anti-
angiogenic therapy in oncology: Current status and future
directions. Lancet 388, 518–529.
Jobst C, Peter S, Gu
¨nther C, et al. (2017). Treatment of ad-
vanced gastrointestinal cancer with genetically modified au-
tologous mesenchymal stem cells-TREAT-ME-1-a phase I,
first in human, first in class trial. Oncotarget 8, 80156.
Johnson DE, O’keefe RA, and Grandis JR. (2018). Targeting
the IL-6/JAK/STAT3 signalling axis in cancer. Nat Rev Clin
Oncol 15, 234–248.
Jotzu C, Alt E, Welte G, et al. (2011). Adipose tissue derived
stem cells differentiate into carcinoma-associated fibroblast-
like cells under the influence of tumor derived factors. Cell
Oncol 34, 55–67.
Juarez P, Mohammad KS, Yin JJ, et al. (2012). Halofuginone
inhibits the establishment and progression of melanoma bone
metastases. Cancer Res 72, 6247–6256.
Jubb A, Cesario A, Ferguson M, et al. (2011). Vascular phe-
notypes in primary non-small cell lung carcinomas and
matched brain metastases. Br J Cancer 104, 1877.
Jung Y, Kim JK, Shiozawa Y, et al. (2013). Recruitment of
mesenchymal stem cells into prostate tumours promotes
metastasis. Nat Commun 4, 1795.
Kahlert C, and Kalluri R. (2013). Exosomes in tumor micro-
environment influence cancer progression and metastasis.
J Mol Med 91, 431–437.
CAFSIN TUMOR MICROENVIRONMENT 333
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
Kakarla S, Chow KK, Mata M, et al. (2013). Antitumor effects
of chimeric receptor engineered human T cells directed to
tumor stroma. Mol Ther 21, 1611–1620.
Kalli M, and Stylianopoulos T. (2018). Defining the role of
solid stress and matrix stiffness in cancer cell proliferation
and metastasis. Front Oncol 8, 55.
Kalluri R. (2016). The biology and function of fibroblasts in
cancer. Nat Rev Cancer 16, 582.
Kalluri R, and Zeisberg M. (2006). Fibroblasts in cancer. Nat
Rev Cancer 6, 392.
Kaneda MM, Messer KS, Ralainirina N, et al. (2016). PI3Kcis
a molecular switch that controls immune suppression. Nature
539, 437.
Kaplan RN, Riba RD, Zacharoulis S, et al. (2005). VEGFR1-
positive haematopoietic bone marrow progenitors initiate the
pre-metastatic niche. Nature 438, 820.
Karkera J, Steiner H, Li W, et al. (2011). The anti-interleukin-6
antibody siltuximab down-regulates genes implicated in tu-
morigenesis in prostate cancer patients from a phase I study.
Prostate 71, 1455–1465.
Karnoub AE, Dash AB, Vo AP, et al. (2007). Mesenchymal
stem cells within tumour stroma promote breast cancer me-
tastasis. Nature 449, 557.
Khan GJ, Sun L, Khan S, Yuan S, and Nongyue H. (2018).
Versatility of cancer associated fibroblasts: Commendable
targets for anti-tumor therapy. Curr Drug Targets 19, 1573–
1588.
Kim DJ, Tremblay ML, and Digiovanni J. (2010). Protein ty-
rosine phosphatases, TC-PTP, SHP1, and SHP2, cooperate in
rapid dephosphorylation of Stat3 in keratinocytes following
UVB irradiation. PLoS One 5, e10290.
Kim JH, Sun-Hee O, Kim E-J, et al. (2012). The role of
myofibroblasts in upregulation of S100A8 and S100A9 and
the differentiation of myeloid cells in the colorectal cancer
microenvironment. Biochem Biophys Res Commun 423,
60–66.
Kirsner RS, Marston WA, Snyder RJ, Lee TD, Cargill DI, and
Slade HB. (2012). Spray-applied cell therapy with human
allogeneic fibroblasts and keratinocytes for the treatment of
chronic venous leg ulcers: A phase 2, multicentre, double-
blind, randomised, placebo-controlled trial. Lancet 380,
977–985.
Kleger A, Perkhofer L, and Seufferlein T. (2014). Smarter drugs
emerging in pancreatic cancer therapy. Ann Oncol 25, 1260–
1270.
Knupfer H, and Preiss R. (2010). Serum interleukin-6 levels in
colorectal cancer patients—A summary of published results.
Int J Colorectal Dis 25, 135–140.
Komrokji RS, Seymour JF, Roberts AW, et al. (2015). Results
of a phase 2 study of pacritinib (SB1518), a JAK2/
JAK2(V617F) inhibitor, in patients with myelofibrosis. Blood
125, 2649–2655.
Kortylewski M, Kujawski M, Wang T, et al. (2005). Inhibiting
Stat3 signaling in the hematopoietic system elicits multi-
component antitumor immunity. Nat Med 11, 1314.
Kraman M, Bambrough PJ, Arnold JN, et al. (2010). Suppres-
sion of antitumor immunity by stromal cells expressing fi-
broblast activation protein–a. Science 330, 827–830.
Kujawski M, Zhang C, Herrmann A, et al. (2010). Targeting
STAT3 in adoptively transferred T cells promotes their
in vivo expansion and antitumor effects. Cancer Res 70,
9599–9610.
Kumari N, Dwarakanath B, Das A, and Bhatt AN. (2016). Role
of interleukin-6 in cancer progression and therapeutic resis-
tance. Tumor Biol 37, 11553–11572.
Kuperwasser C, Chavarria T, Wu M, et al. (2004). Re-
construction of functionally normal and malignant human
breast tissues in mice. Proc Natl Acad Sci U S A 101, 4966–
4971.
Kusaba T, Nakayama T, Yamazumi K, et al. (2006). Activation
of STAT3 is a marker of poor prognosis in human colorectal
cancer. Oncol Rep 15, 1445–1451.
Lakins MA, Ghorani E, Munir H, Martins CP, and Shields JD.
(2018). Cancer-associated fibroblasts induce antigen-specific
deletion of CD8+T Cells to protect tumour cells. Nat Com-
mun 9, 948.
Laklai H, Miroshnikova YA, Pickup MW, et al. (2016). Gen-
otype tunes pancreatic ductal adenocarcinoma tissue tension
to induce matricellular fibrosis and tumor progression. Nat
Med 22, 497.
Lazard D, Sastre X, Frid MG, Glukhova MA, Thiery J-P, and
Koteliansky VE. (1993). Expression of smooth muscle-
specific proteins in myoepithelium and stromal myofibro-
blasts of normal and malignant human breast tissue. Proc Natl
Acad Sci U S A 90, 999–1003.
Lederle W, Depner S, Schnur S, et al. (2011). IL-6 promotes
malignant growth of skin SCCs by regulating a network of
autocrine and paracrine cytokines. Int J Cancer 128, 2803–
2814.
Lee H, Pal SK, Reckamp K, Figlin RA, and Yu H. (2010).
STAT3: A target to enhance antitumor immune response.
Cancer Immunol Immunother 344, 41–59.
Lesina M, Kurkowski MU, Ludes K, et al. (2011). Stat3/Socs3
activation by IL-6 transsignaling promotes progression of
pancreatic intraepithelial neoplasia and development of pan-
creatic cancer. Cancer Cell 19, 456–469.
Lewis CE, Harney AS, and Pollard JW. (2016). The multifac-
eted role of perivascular macrophages in tumors. Cancer Cell
30, 18–25.
Li F, Cao Y, Townsend CM, and Ko TC. (2005). TGF-bsig-
naling in colon cancer cells. World J Surg 29, 306–311.
Li Y, Rogoff HA, Keates S, et al. (2015). Suppression of cancer
relapse and metastasis by inhibiting cancer stemness. Proc
Natl Acad Sci U S A 112, 1839–1844.
Li Z-H, Dulyaninova NG, House RP, Almo SC, and Bresnick
AR. (2010). S100A4 regulates macrophage chemotaxis. Mol
Biol Cell 21, 2598–2610.
Liang W, and Ferrara N. (2016). The complex role of neutro-
phils in tumor angiogenesis and metastasis. Cancer Immunol
Res 4, 83–91.
Liekens S, Schols D, and Hatse S. (2010). CXCL12-CXCR4
axis in angiogenesis, metastasis and stem cell mobilization.
Curr Pharm Des 16, 3903–3920.
Liu T, Han C, Wang S, et al. (2019a). Cancer-associated fi-
broblasts: An emerging target of anti-cancer immunotherapy.
J Hematol Oncol 12, 86.
Liu T, Zhou L, Li D, Andl T, and Zhang Y. (2019b). Cancer-
associated fibroblasts build and secure the tumor microenvi-
ronment. Front Cell Dev Biol 7, 60.
Lo A, Wang L-CS, Scholler J, et al. (2015). Tumor-promoting
desmoplasia is disrupted by depleting FAP-expressing stro-
mal cells. Cancer Res 75, 2800–2810.
Loeffler M, Kru
¨ger JA, Niethammer AG, and Reisfeld RA.
(2006). Targeting tumor-associated fibroblasts improves
334 DZOBO AND DANDARA
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
cancer chemotherapy by increasing intratumoral drug uptake.
J Clin Invest 116, 1955–1962.
Ma X-J, Dahiya S, Richardson E, Erlander M, and Sgroi DC.
(2009). Gene expression profiling of the tumor microenvi-
ronment during breast cancer progression. Breast Cancer Res
11, R7.
Maccio A, and Madeddu C. (2013). The role of interleukin-6 in
the evolution of ovarian cancer: Clinical and prognostic im-
plications—A review. J Mol Med (Berl) 91, 1355–1368.
Macha MA, Matta A, Kaur J, et al. (2011). Prognostic sig-
nificance of nuclear pSTAT3 in oral cancer. Head Neck 33,
482–489.
Maller O, Dufort CC, and Weaver VM. (2013). YAP forces
fibroblasts to feel the tension. Nat Cell Biol 15, 570.
Mariathasan S, Turley SJ, Nickles D, et al. (2018). TGFbeta
attenuates tumour response to PD-L1 blockade by contribut-
ing to exclusion of T cells. Nature 554, 544–548.
Martens JW, Sieuwerts AM, Bolt-De Vries J, et al. (2003).
Aging of stromal-derived human breast fibroblasts might
contribute to breast cancer progression. Thromb Haemost 89,
393–404.
Masjedi A, Hashemi V, Hojjat-Farsangi M, et al. (2018). The
significant role of interleukin-6 and its signaling pathway in
the immunopathogenesis and treatment of breast cancer.
Biomed Pharmacother 108, 1415–1424.
Maxwell P, Dachs G, Gleadle J, et al. (1997). Hypoxia-
inducible factor-1 modulates gene expression in solid tumors
and influences both angiogenesis and tumor growth. Proc Natl
Acad Sci U S A 94, 8104–8109.
Miao L, Liu Q, Lin CM, et al. (2017). Targeting tumor-
associated fibroblasts for therapeutic delivery in desmoplastic
tumors. Cancer Res 77, 719–731.
Mitra A, Mishra L, and Li S. (2015). EMT, CTCs and CSCs in
tumor relapse and drug-resistance. Oncotarget 6, 10697.
Mitra AK, Zillhardt M, Hua Y, et al. (2012). MicroRNAs re-
program normal fibroblasts into cancer-associated fibroblasts
in ovarian cancer. Cancer Discovery 2, 1100–1108.
Moinfar F, Man YG, Arnould L, Bratthauer GL, Ratschek M,
and Tavassoli FA. (2000). Concurrent and independent ge-
netic alterations in the stromal and epithelial cells of mam-
mary carcinoma: Implications for tumorigenesis. Cancer Res
60, 2562–2566.
Mongiat M, Andreuzzi E, Tarticchio G, and Paulitti A. (2016).
Extracellular matrix, a hard player in angiogenesis. Int J Mol
Sci 17, pii: E1822.
Monsky WL, Carreira CM, Tsuzuki Y, Gohongi T, Fukumura
D, and Jain RK. (2002). Role of host microenvironment in
angiogenesis and microvascular functions in human breast
cancer xenografts: Mammary fat pad versus cranial tumors.
Clin Cancer Res 8, 1008–1013.
Morikawa S, Baluk P, Kaidoh T, Haskell A, Jain RK, and
Mcdonald DM. (2002). Abnormalities in pericytes on blood
vessels and endothelial sprouts in tumors. Am J Pathol 160,
985–1000.
Morrissey C, True LD, Roudier MP, et al. (2008). Differential
expression of angiogenesis associated genes in prostate can-
cer bone, liver and lymph node metastases. Clin Exp Me-
tastasis 25, 377–388.
Moten A, Schafer D, Farmer P, Kim J, and Ferrari M. (2014).
Redefining global health priorities: Improving cancer care in
developing settings. J Glob Health 4, 010304.
Mueller MM, and Fusenig NE. (2004). Friends or foes—Bipolar
effects of the tumour stroma in cancer. Nat Rev Cancer 4,
839.
Murakami M, and Simons M. (2008). Fibroblast growth factor
regulation of neovascularization. Curr Opin Hematol 15,
215–220.
Musumeci M, Coppola V, Addario A, et al. (2011). Control of
tumor and microenvironment cross-talk by miR-15a and miR-
16 in prostate cancer. Oncogene 30, 4231–4242.
Nagasaki T, Hara M, Nakanishi H, Takahashi H, Sato M, and
Takeyama H. (2014). Interleukin-6 released by colon cancer-
associated fibroblasts is critical for tumour angiogenesis:
Anti-interleukin-6 receptor antibody suppressed angiogenesis
and inhibited tumour–stroma interaction. Br J Cancer 110,
469.
Nassar D, and Blanpain C. (2016). Cancer stem cells: Basic
concepts and therapeutic implications. Annu Rev Pathol 11,
47–76.
Neubauer H, Cumano A, Muller M, Wu H, Huffstadt U, and
Pfeffer K. (1998). Jak2 deficiency defines an essential de-
velopmental checkpoint in definitive hematopoiesis. Cell 93,
397–409.
Neve A, Cantatore FP, Maruotti N, Corrado A, and Ribatti D.
(2014). Extracellular matrix modulates angiogenesis in
physiological and pathological conditions. Biomed Res Int
2014, 756078.
Nicosia RF, Bonanno E, and Smith M. (1993). Fibronectin
promotes the elongation of microvessels during angiogenesis
in vitro. J Cell Physiol 154, 654–661.
Niess H, Von Einem JC, Thomas MN, et al. (2015). Treatment
of advanced gastrointestinal tumors with genetically modi-
fied autologous mesenchymal stromal cells (TREAT-ME1):
Study protocol of a phase I/II clinical trial. BMC Cancer 15,
237.
Noguera R, Nieto OA, Tadeo I, Farin
˜as F, and A
´lvaro T.
(2012). Extracellular matrix, biotensegrity and tumor mi-
croenvironment. An update and overview. Histol Histo-
pathol 27, 693–705.
Nouraee N, Van Roosbroeck K, Vasei M, et al. (2013). Ex-
pression, tissue distribution and function of miR-21 in
esophageal squamous cell carcinoma. PLoS One 8, e73009.
Nozawa H, Chiu C, and Hanahan D. (2006). Infiltrating neu-
trophils mediate the initial angiogenic switch in a mouse
model of multistage carcinogenesis. Proc Natl Acad Sci U S
A 103, 12493–12498.
O’connell JT, Sugimoto H, Cooke VG, et al. (2011). VEGF-A
and Tenascin-C produced by S100A4+stromal cells are im-
portant for metastatic colonization. Proc Natl Acad Sci U S A
108, 16002–16007.
O
¨hlund D, Elyada E, and Tuveson D. (2014). Fibroblast
heterogeneity in the cancer wound. J Exp Med 211, 1503–
1523.
O
¨hlund D, Handly-Santana A, Biffi G, et al. (2017). Distinct
populations of inflammatory fibroblasts and myofibroblasts in
pancreatic cancer. J Exp Med 214, 579–596.
Okaji Y, Tsuno NH, Tanaka M, et al. (2008). Pilot study of anti-
angiogenic vaccine using fixed whole endothelium in patients
with progressive malignancy after failure of conventional
therapy. Eur J Cancer 44, 383–390.
Olive KP, Jacobetz MA, Davidson CJ, et al. (2009). Inhibition
of Hedgehog signaling enhances delivery of chemotherapy in
a mouse model of pancreatic cancer. Science 324, 1457–
1461.
Olumi AF, Grossfeld GD, Hayward SW, Carroll PR, Tlsty TD,
and Cunha GR. (1999). Carcinoma-associated fibroblasts di-
rect tumor progression of initiated human prostatic epithe-
lium. Cancer Res 59, 5002–5011.
CAFSIN TUMOR MICROENVIRONMENT 335
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
Omary MB, Lugea A, Lowe AW, and Pandol SJ. (2007). The
pancreatic stellate cell: A star on the rise in pancreatic dis-
eases. J Clin Invest 117, 50–59.
Orimo A, Gupta PB, Sgroi DC, et al. (2005). Stromal fibroblasts
present in invasive human breast carcinomas promote tumor
growth and angiogenesis through elevated SDF-1/CXCL12
secretion. Cell 121, 335–348.
Orimo A, and Weinberg RA. (2007). Heterogeneity of stromal
fibroblasts in tumor. Cancer Biol Ther 6, 618–619.
Ostermann E, Garin-Chesa P, Heider KH, et al. (2008). Effec-
tive immunoconjugate therapy in cancer models targeting a
serine protease of tumor fibroblasts. Clin Cancer Res 14,
4584–4592.
O
¨zdemir BC, Pentcheva-Hoang T, Carstens JL, et al. (2014).
Depletion of carcinoma-associated fibroblasts and fibrosis
induces immunosuppression and accelerates pancreas cancer
with reduced survival. Cancer Cell 25, 719–734.
Pa
`ez-Ribes M, Allen E, Hudock J, et al. (2009). Antiangiogenic
therapy elicits malignant progression of tumors to increased
local invasion and distant metastasis. Cancer Cell 15, 220–
231.
Paget S. (1889). The distribution of secondary growths in
cancer of the breast. Lancet 133, 571–573.
Pan Y, Zhou F, Zhang R, and Claret FX. (2013). Stat3 inhibitor
Stattic exhibits potent antitumor activity and induces chemo-
and radio-sensitivity in nasopharyngeal carcinoma. PLoS One
8, e54565.
Pang W, Su J, Wang Y, et al. (2015). Pancreatic cancer-
secreted miR-155 implicates in the conversion from normal
fibroblasts to cancer-associated fibroblasts. Cancer Sci 106,
1362–1369.
Park H, Cho J-A, Kim S-K, Kim J-H, and Lee S-H. (2008).
Hyperthermia on mesenchymal stem cells (MSCs) can sen-
sitize tumor cells to undergo cell death. Int J Hyperthermia
24, 638–648.
Patocs A, Zhang L, Xu Y, et al. (2007). Breast-cancer stromal
cells with TP53 mutations and nodal metastases. N Engl J
Med 357, 2543–2551.
Patrikidou A, Sinapi I, Regnault H, et al. (2014). Gemcitabine
and oxaliplatin chemotherapy for advanced hepatocellular
carcinoma after failure of anti-angiogenic therapies. Invest
New Drugs 32, 1028–1035.
Paulsson J, and Micke P. (2014). Prognostic relevance of
cancer-associated fibroblasts in human cancer. Semin Cancer
Biol 25, 61–68.
Peiris-Page
`s M, Smith DL, Gy}
orffy B, Sotgia F, and Lisanti
MP. (2015a). Proteomic identification of prognostic tumour
biomarkers, using chemotherapy-induced cancer-associated
fibroblasts. Aging (Albany NY) 7, 816.
Peiris-Page
`s M, Sotgia F, and Lisanti MP. (2015b). Che-
motherapy induces the cancer-associated fibroblast pheno-
type, activating paracrine Hedgehog-GLI signalling in breast
cancer cells. Oncotarget 6, 10728–10745.
Pen
˜aC,Ce
´spedes MV, Lindh MB, et al. (2013). STC1 ex-
pression by cancer-associated fibroblasts drives metastasis of
colorectal cancer. Cancer Res 73, 1287–1297.
Picard O, Rolland Y, and Poupon MF. (1986). Fibroblast-
dependent tumorigenicity of cells in nude mice: Implication
for implantation of metastases. Cancer Res 46, 3290–
3294.
Pietras K, Pahler J, Bergers G, and Hanahan D. (2008). Func-
tions of paracrine PDGF signaling in the proangiogenic tumor
stroma revealed by pharmacological targeting. PLoS Med 5,
e19.
Poltavets V, Kochetkova M, Pitson SM, and Samuel MS.
(2018). The role of the extracellular matrix and its molecular
and cellular regulators in cancer cell plasticity. Front Oncol 8,
431.
Potente M, Gerhardt H, and Carmeliet P. (2011). Basic and
therapeutic aspects of angiogenesis. Cell 146, 873–887.
Provenzano PP, Cuevas C, Chang AE, Goel VK, Von Hoff DD,
and Hingorani SR. (2012). Enzymatic targeting of the stroma
ablates physical barriers to treatment of pancreatic ductal
adenocarcinoma. Cancer Cell 21, 418–429.
Provenzano PP, Inman DR, Eliceiri KW, et al. (2008). Collagen
density promotes mammary tumor initiation and progression.
BMC Med 6, 11.
Quail DF, and Joyce JA. (2013). Microenvironmental regulation
of tumor progression and metastasis. Nat Med 19, 1423.
Raica M, Cimpean AM, and Ribatti D. (2009). Angiogenesis in
pre-malignant conditions. Eur J Cancer 45, 1924–1934.
Reardon DA, Akabani G, Coleman RE, et al. (2006). Salvage
radioimmunotherapy with murine iodine-131–labeled anti-
tenascin monoclonal antibody 81C6 for patients with recur-
rent primary and metastatic malignant brain tumors: Phase II
study results. J Clin Oncol 24, 115–122.
Reisfeld RA. (2007). Immunotherapy targeting tumor stromal
fibroblasts improves chemotherapy of breast cancer. Scripps
Research Inst, La Jolla, CA.
Reiss M. (1999). TGF-beta and cancer. Microbes Infect 1,
1327–1347.
Rhim AD, Oberstein PE, Thomas DH, et al. (2014). Stromal
elements act to restrain, rather than support, pancreatic ductal
adenocarcinoma. Cancer Cell 25, 735–747.
Richards KE, Zeleniak AE, Fishel ML, Wu J, Littlepage LE,
and Hill R. (2017). Cancer-associated fibroblast exosomes
regulate survival and proliferation of pancreatic cancer cells.
Oncogene 36, 1770.
Riches DW. (1988). The multiple roles of macrophages in
wound healing. In: The Molecular and Cellular Biology of
Wound Repair. Eds: Clark RAF, and Henson PM. Plenum
Press, New York, NY, USA. 213–239.
Ridge SM, Bhattacharyya D, Dervan E, et al. (2018). Secreted
factors from metastatic prostate cancer cells stimulate mes-
enchymal stem cell transition to a pro-tumourigenic ‘acti-
vated’ state that enhances prostate cancer cell migration. Int J
Cancer 142, 2056–2067.
Rivera LB, Meyronet D, Hervieu V, Frederick MJ, Bergsland E,
and Bergers G. (2015). Intratumoral myeloid cells regulate
responsiveness and resistance to antiangiogenic therapy. Cell
Rep 11, 577–591.
Roberts EW, Deonarine A, Jones JO, et al. (2013). Depletion of
stromal cells expressing fibroblast activation protein-afrom
skeletal muscle and bone marrow results in cachexia and
anemia. J Exp Med 210, 1137–1151.
Roeser JC, Leach SD, and Mcallister F. (2015). Emerging
strategies for cancer immunoprevention. Oncogene 34, 6029–
6039.
Ronnov-Jessen L, Petersen OW, and Bissell MJ. (1996). Cel-
lular changes involved in conversion of normal to malignant
breast: Importance of the stromal reaction. Physiol Rev 76,
69–125.
Rubenstein JL, Kim J, Ozawa T, et al. (2000). Anti-VEGF
antibody treatment of glioblastoma prolongs survival but re-
sults in increased vascular cooption. Neoplasia 2, 306–314.
Ryan HE, Poloni M, Mcnulty W, et al. (2000). Hypoxia-
inducible factor-1ais a positive factor in solid tumor growth.
Cancer Res 60, 4010–4015.
336 DZOBO AND DANDARA
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
Saadi A, Shannon NB, Lao-Sirieix P, et al. (2010). Stromal
genes discriminate preinvasive from invasive disease, predict
outcome, and highlight inflammatory pathways in digestive
cancers. Proc Natl Acad Sci U S A 107, 2177–2182.
Saharinen P, Eklund L, and Alitalo K. (2017). Therapeutic
targeting of the angiopoietin–TIE pathway. Nat Rev Drug
Discov 16, 635.
San Francisco IF, Dewolf WC, Peehl DM, and Olumi AF.
(2004). Expression of transforming growth factor-beta 1 and
growth in soft agar differentiate prostate carcinoma-
associated fibroblasts from normal prostate fibroblasts. Int J
Cancer 112, 213–218.
Sandborn WJ, Su C, Sands BE, et al. (2017). Tofacitinib as
induction and maintenance therapy for ulcerative colitis.
N Engl J Med 376, 1723–1736.
Sanguinete MMM, Oliveira PH, Martins-Filho A, et al. (2017).
Serum IL-6 and IL-8 correlate with prognostic factors in
ovarian cancer. Immunol Invest 46, 677–688.
Sansone P, Berishaj M, Rajasekhar VK, et al. (2017). Evolution
of cancer stem-like cells in endocrine-resistant metastatic
breast cancers is mediated by stromal microvesicles. Cancer
Res 77, 1927–1941.
Sansone P, and Bromberg J. (2012). Targeting the interleukin-6/
Jak/stat pathway in human malignancies. J Clin Oncol 30,
1005.
Sansone P, Storci G, Tavolari S, et al. (2007). IL-6 triggers
malignant features in mammospheres from human ductal
breast carcinoma and normal mammary gland. J Clin Invest
117, 3988–4002.
Santos AM, Jung J, Aziz N, Kissil JL, and Pure E. (2009).
Targeting fibroblast activation protein inhibits tumor stro-
magenesis and growth in mice. J Clin Invest 119, 3613–
3625.
Sanz-Moreno V, Gaggioli C, Yeo M, et al. (2011). ROCK and
JAK1 signaling cooperate to control actomyosin contractility
in tumor cells and stroma. Cancer Cell 20, 229–245.
Scherz-Shouval R, Santagata S, Mendillo ML, et al. (2014). The
reprogramming of tumor stroma by HSF1 is a potent enabler
of malignancy. Cell 158, 564–578.
Schmittnaegel M, Rigamonti N, Kadioglu E, et al. (2017). Dual
angiopoietin-2 and VEGFA inhibition elicits antitumor im-
munity that is enhanced by PD-1 checkpoint blockade. Sci
Transl Med 9, eaak9670.
Schust J, Sperl B, Hollis A, Mayer TU, and Berg T. (2006).
Stattic: A small-molecule inhibitor of STAT3 activation and
dimerization. Chem Biol 13, 1235–1242.
Sekine Y, Tsuji S, Ikeda O, et al. (2006). Regulation of STAT3-
mediated signaling by LMW-DSP2. Oncogene 25, 5801.
Sen M, Joyce S, Panahandeh M, et al. (2012). Targeting Stat3
abrogates EGFR inhibitor resistance in cancer. Clin Cancer
Res 18, 4986–4996.
Senthebane D, Jonker T, Rowe A, et al. (2018). The role of
tumor microenvironment in chemoresistance: 3D extracellu-
lar matrices as accomplices. Int J Mol Sci 19, 2861.
Senthebane DA, Rowe A, Thomford NE, et al. (2017). The role
of tumor microenvironment in chemoresistance: To survive,
keep your enemies closer. Int J Mol Sci 18, 1586.
Shekhar MP, Werdell J, Santner SJ, Pauley RJ, and Tait L.
(2001). Breast stroma plays a dominant regulatory role in
breast epithelial growth and differentiation: Implications
for tumor development and progression. Cancer Res 61,
1320–1326.
Sherman MH, Ruth TY, Engle DD, et al. (2014). Vitamin D
receptor-mediated stromal reprogramming suppresses pan-
creatitis and enhances pancreatic cancer therapy. Cell 159,
80–93.
Shi Y, Du L, Lin L, and Wang Y. (2017). Tumour-associated
mesenchymal stem/stromal cells: Emerging therapeutic tar-
gets. Nat Rev Drug Discov 16, 35.
Shintani Y, Fujiwara A, Kimura T, et al. (2016). IL-6 secreted
from cancer-associated fibroblasts mediates chemoresistance
in NSCLC by increasing epithelial-mesenchymal transition
signaling. J Thorac Oncol 11, 1482–1492.
Siddiquee K, Zhang S, Guida WC, et al. (2007). Selective
chemical probe inhibitor of Stat3, identified through
structure-based virtual screening, induces antitumor activity.
Proc Natl Acad Sci U S A 104, 7391–7396.
Song L, Smith MA, Doshi P, et al. (2014). Antitumor efficacy
of the anti-interleukin-6 (IL-6) antibody siltuximab in
mouse xenograft models of lung cancer. J Thorac Oncol 9,
974–982.
Spitzner M, Ebner R, Wolff HA, Ghadimi BM, Wienands J, and
Grade M. (2014). STAT3: A novel molecular mediator of
resistance to chemoradiotherapy. Cancers (Basel) 6, 1986–
2011.
Steinbichler TB, Dudas J, Skvortsov S, Ganswindt U, Rie-
chelmann H, and Skvortsova II. (2018). Therapy resistance
mediated by cancer stem cells. Semin Cancer Biol 53, 156–
167.
SteynPJ,DzoboK,SmithRI,andMyburghKH.
(2019). Interleukin-6 induces myogenic differentiation via
JAK2-STAT3 signaling in mouse C2C12 myoblast cell line
and primary human myoblasts. Int J Mol Sci 20, pii:
E5273.
Stoker M, Shearer M, and O’neill C. (1966). Growth inhibition
of polyoma-transformed cells by contact with static normal
fibroblasts. J Cell Sci 1, 297–310.
Straussman R, Morikawa T, Shee K, et al. (2012). Tumour
micro-environment elicits innate resistance to RAF inhibitors
through HGF secretion. Nature 487, 500.
Strom T, Harrison LB, Giuliano AR, et al. (2017). Tumour
radiosensitivity is associated with immune activation in solid
tumours. Eur J Cancer 84, 304–314.
Su S, Chen J, Yao H, et al. (2018). CD10+GPR77+cancer-
associated fibroblasts promote cancer formation and che-
moresistance by sustaining cancer stemness. Cell 172,
841–856.e816.
Sullivan JP, Minna JD, and Shay JW. (2010). Evidence for self-
renewing lung cancer stem cells and their implications in
tumor initiation, progression, and targeted therapy. Cancer
Metastasis Rev 29, 61–72.
Sullivan NJ, Sasser AK, Axel AE, et al. (2009). Interleukin-6
induces an epithelial-mesenchymal transition phenotype in
human breast cancer cells. Oncogene 28, 2940–2947.
Surowiak P, Murawa D, Materna V, et al. (2007). Occurence of
stromal myofibroblasts in the invasive ductal breast cancer
tissue is an unfavourable prognostic factor. Anticancer Res
27, 2917–2924.
Taguchi A, Kawana K, Tomio K, et al. (2014). Matrix me-
talloproteinase (MMP)-9 in cancer-associated fibroblasts
(CAFs) is suppressed by omega-3 polyunsaturated fatty acids
in vitro and in vivo. PLoS One 9, e89605.
Tan H-X, Cao Z-B, He T-T, Huang T, Xiang C-L, and Liu Y.
(2019). TGFb1 is essential for MSCs-CAFs differentiation
CAFSIN TUMOR MICROENVIRONMENT 337
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
and promotes HCT116 cells migration and invasion via
JAK/STAT3 signaling. Onco Targets Ther 12, 5323–5334.
Taniguchi K, and Karin M. (2014). IL-6 and related cytokines
as the critical lynchpins between inflammation and cancer.
Semin Immunol 26, 54–74.
Taube JH, Herschkowitz JI, Komurov K, et al. (2010). Core
epithelial-to-mesenchymal transition interactome gene-
expression signature is associated with claudin-low and
metaplastic breast cancer subtypes. Proc Natl Acad Sci U S A
107, 15449–15454.
Tavallai M, Booth L, Roberts JL, Poklepovic A, and Dent
P. (2016). Rationally repurposing ruxolitinib ( Jakafi (
)) as a
solid tumor therapeutic. Front Oncol 6, 142.
Tawara K, Oxford JT, and Jorcyk CL. (2011). Clinical signifi-
cance of interleukin (IL)-6 in cancer metastasis to bone:
Potential of anti-IL-6 therapies. Cancer Manag Res 3, 177.
Toivola DM, Tao G-Z, Habtezion A, Liao J, and Omary MB.
(2005). Cellular integrity plus: Organelle-related and protein-
targeting functions of intermediate filaments. Trends Cell
Biol 15, 608–617.
Toullec A, Gerald D, Despouy G, et al. (2010). Oxidative stress
promotes myofibroblast differentiation and tumour spreading.
EMBO Mol Med 2, 211–230.
Trimboli AJ, Cantemir-Stone CZ, Li F, et al. (2009). Pten in
stromal fibroblasts suppresses mammary epithelial tumours.
Nature 461, 1084.
Tsujino T, Seshimo I, Yamamoto H, et al. (2007). Stromal
myofibroblasts predict disease recurrence for colorectal can-
cer. Clin Cancer Res 13, 2082–2090.
Twardowski T, Fertala A, Orgel JP, and San Antonio JD. (2007).
Type I collagen and collagen mimetics as angiogenesis pro-
moting superpolymers. Curr Pharm Des 13, 3608–3621.
Valkenburg KC, De Groot AE, and Pienta KJ. (2018). Targeting
the tumour stroma to improve cancer therapy. Nat Rev Clin
Oncol 15, 366–381.
Vandenbroucke RE, and Libert C. (2014). Is there new hope for
therapeutic matrix metalloproteinase inhibition? Nat Rev
Drug Discov 13, 904–927.
Vannucchi AM, Kiladjian JJ, Griesshammer M, et al. (2015).
Ruxolitinib versus standard therapy for the treatment of
polycythemia vera. N Engl J Med 372, 426–435.
Vega S, Morales AV, Ocan
˜a OH, Valde
´s F, Fabregat I, and
Nieto MA. (2004). Snail blocks the cell cycle and confers
resistance to cell death. Genes Dev 18, 1131–1143.
Vellinga T, Den Uil S, Rinkes I, et al. (2016). Collagen-rich
stroma in aggressive colon tumors induces mesenchymal
gene expression and tumor cell invasion. Oncogene 35, 5263.
Vennin C, Me
´le
´nec P, Rouet R, et al. (2019). CAF hierarchy
driven by pancreatic cancer cell p53-status creates a pro-
metastatic and chemoresistant environment via perlecan. Nat
Commun 10, 3637.
Ve
´tizou M, Pitt JM, Daille
`re R, et al. (2015). Anticancer im-
munotherapy by CTLA-4 blockade relies on the gut micro-
biota. Science 350, 1079–1084.
Virchow R. (1871). Die Cellularpathologie in ihrer Be-
gru
¨ndung auf physiologische und pathologische Gewebe-
lehre. Hirschwald. Berlin, Germany, 4th ed., ref no: 43203.
Vuoriluoto K, Haugen H, Kiviluoto S, et al. (2011). Vimentin
regulates EMT induction by Slug and oncogenic H-Ras and
migration by governing Axl expression in breast cancer.
Oncogene 30, 1436.
Wang J, Zhang G, Wang J, Wang L, Huang X, and Cheng Y.
(2016). The role of cancer-associated fibroblasts in esopha-
geal cancer. J Transl Med 14, 30.
Wang L, Cao L, Wang H, et al. (2017). Cancer-associated fi-
broblasts enhance metastatic potential of lung cancer cells
through IL-6/STAT3 signaling pathway. Oncotarget 8, 76116.
Wang L-CS, Lo A, Scholler J, et al. (2014). Targeting fibroblast
activation protein in tumor stroma with chimeric antigen re-
ceptor T cells can inhibit tumor growth and augment host
immunity without severe toxicity. Cancer Immunol Res 2,
154–166.
Wang N, and Stamenovic D. (2002). Mechanics of vimentin
intermediate filaments. J Muscle Res Cell Motil 23, 535–540.
Wang X, Zhang W, Sun X, Lin Y, and Chen W. (2018). Cancer-
associated fibroblasts induce epithelial-mesenchymal transi-
tion through secreted cytokines in endometrial cancer cells.
Oncol Lett 15, 5694–5702.
Wang XM, Yu DMT, Mccaughan GW, and Gorrell MD.
(2005). Fibroblast activation protein increases apoptosis, cell
adhesion, and migration by the LX-2 human stellate cell line.
Hepatology 42, 935–945.
Wang Y, Yan W, Lu X, et al. (2011). Overexpression of os-
teopontin induces angiogenesis of endothelial progenitor cells
via the avbeta3/PI3K/AKT/eNOS/NO signaling pathway in
glioma cells. Eur J Cell Biol 90, 642–648.
Wang Z, Si X, Xu A, et al. (2013). Activation of STAT3 in
human gastric cancer cells via interleukin (IL)-6-type cyto-
kine signaling correlates with clinical implications. PLoS One
8, e75788.
Webber J, Steadman R, Mason MD, Tabi Z, and Clayton A.
(2010). Cancer exosomes trigger fibroblast to myofibroblast
differentiation. Cancer Res 70, 9621–9630.
Weber CE, Kothari AN, Wai PY, et al. (2015). Osteopontin
mediates an MZF1-TGF-beta1-dependent transformation of
mesenchymal stem cells into cancer-associated fibroblasts in
breast cancer. Oncogene 34, 4821–4833.
Wellner U, Schubert J, Burk UC, et al. (2009). The EMT-
activator ZEB1 promotes tumorigenicity by repressing
stemness-inhibiting microRNAs. Nat Cell Biol 11, 1487.
Wels J, Kaplan RN, Rafii S, and Lyden D. (2008). Migratory
neighbors and distant invaders: Tumor-associated niche cells.
Genes Dev 22, 559–574.
Wojtowicz-Praga SM, Dickson RB, and Hawkins MJ. (1997).
Matrix metalloproteinase inhibitors. Invest New Drugs 15,
61–75.
Wynn TA, and Ramalingam TR. (2012). Mechanisms of fi-
brosis: Therapeutic translation for fibrotic disease. Nat Med
18, 1028.
Yang X, Lin Y, Shi Y, et al. (2016). FAP promotes immuno-
suppression by cancer-associated fibroblasts in the tumor
microenvironment via STAT3–CCL2 signaling. Cancer Res
76, 4124–4135.
Yates LR, Knappskog S, Wedge D, et al. (2017). Genomic
evolution of breast cancer metastasis and relapse. Cancer Cell
32, 169–184.e167.
Yin C, Evason KJ, Asahina K, and Stainier DY. (2013). Hepatic
stellate cells in liver development, regeneration, and cancer.
J Clin Invest 123, 1902–1910.
Younes A, Berdeja JG, Patel MR, et al. (2016). Safety, tolera-
bility, and preliminary activity of CUDC-907, a first-in-class,
oral, dual inhibitor of HDAC and PI3K, in patients with re-
lapsed or refractory lymphoma or multiple myeloma: An
open-label, dose-escalation, phase 1 trial. Lancet Oncol 17,
622–631.
Yu F, Yao H, Zhu P, et al. (2007a). let-7 regulates self renewal
and tumorigenicity of breast cancer cells. Cell 131, 1109–
1123.
338 DZOBO AND DANDARA
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
Yu H, and Jove R. (2004). The STATs of cancer—New mo-
lecular targets come of age. Nat Rev Cancer 4, 97.
Yu H, Kortylewski M, and Pardoll D. (2007b). Tumour im-
munology: Crosstalk between cancer and immune cells: Role
of STAT3 in the tumour microenvironment. Nat Rev Im-
munol 7, 41.
Yu H, Pardoll D, and Jove R. (2009). STATs in cancer in-
flammation and immunity: A leading role for STAT3. Nat
Rev Cancer 9, 798.
Yu Y, Xiao C, Tan L, Wang Q, Li X, and Feng Y. (2014).
Cancer-associated fibroblasts induce epithelial–mesenchymal
transition of breast cancer cells through paracrine TGF-b
signalling. Br J Cancer 110, 724.
Zeisberg EM, Potenta S, Xie L, Zeisberg M, and Kalluri R.
(2007). Discovery of endothelial to mesenchymal transition
as a source for carcinoma-associated fibroblasts. Cancer Res
67, 10123–10128.
Zhang J, Chen L, Liu X, Kammertoens T, Blankenstein T, and
Qin Z. (2013a). Fibroblast-specific protein 1/S100A4–pos-
itive cells prevent carcinoma through collagen production and
encapsulation of carcinogens. Cancer Res 73, 2770–2781.
Zhang K, Jiao X, Liu X, et al. (2010a). Knockdown of snail
sensitizes pancreatic cancer cells to chemotherapeutic agents
and irradiation. Int J Mol Sci 11, 4891–4904.
Zhang L, Zhou Y, Sun X, Zhou J, and Yang P. (2017). CXCL12
overexpression promotes the angiogenesis potential of peri-
odontal ligament stem cells. Sci Rep 7, 10286.
Zhang X, Guo A, Yu J, et al. (2007). Identification of STAT3 as
a substrate of receptor protein tyrosine phosphatase T. Proc
Natl Acad Sci U S A 104, 4060–4064.
Zhang X, Yue P, Fletcher S, Zhao W, Gunning PT, and Turkson
J. (2010b). A novel small-molecule disrupts Stat3 SH2
domain-phosphotyrosine interactions and Stat3-dependent
tumor processes. Biochem Pharmacol 79, 1398–1409.
Zhang XH-F, Jin X, Malladi S, et al. (2013b). Selection of bone
metastasis seeds by mesenchymal signals in the primary tu-
mor stroma. Cell 154, 1060–1073.
Zhao J, Chen L, Shu B, et al. (2014). Granulocyte/macrophage
colony-stimulating factor influences angiogenesis by regu-
lating the coordinated expression of VEGF and the Ang/Tie
system. PLoS One 9, e92691.
Zhao J, Wang H, Hsiao CH, et al. (2018). Simultaneous inhi-
bition of hedgehog signaling and tumor proliferation re-
models stroma and enhances pancreatic cancer therapy.
Biomaterials 159, 215–228.
Zheng X, Carstens JL, Kim J, et al. (2015). Epithelial-to-
mesenchymal transition is dispensable for metastasis but
induces chemoresistance in pancreatic cancer. Nature 527,
525–530.
Zhou B-BS, Zhang H, Damelin M, Geles KG, Grindley JC, and
Dirks PB. (2009). Tumour-initiating cells: Challenges and
opportunities for anticancer drug discovery. Nat Rev Drug
Discov 8, 806.
Zhou L, Isenberg JS, Cao Z, and Roberts DD. (2006). Type I
collagen is a molecular target for inhibition of angiogenesis
by endogenous thrombospondin-1. Oncogene 25, 536–545.
Zhu Q, Zhang X, Zhang L, et al. (2014). The IL-6–STAT3 axis
mediates a reciprocal crosstalk between cancer-derived
mesenchymal stem cells and neutrophils to synergistically
prompt gastric cancer progression. Cell Death Dis 5, e1295.
Znaor A, Eser S, Anton-Culver H, et al. (2018). Cancer sur-
veillance in northern Africa, and central and western Asia:
Challenges and strategies in support of developing cancer
registries. Lancet Oncol 19, e85–e92.
Address correspondence to:
Kevin Dzobo, PhD
International Centre for Genetic Engineering
and Biotechnology (ICGEB)
Cape Town Component
Wernher and Beit Building (South)
Anzio Road
Observatory
Cape Town 7925
South Africa
E-mail: kdzobosnr@yahoo.com
Collet Dandara, PhD
Division of Human Genetics
Pharmacogenetics Research Group
Division of Human Genetics
Department of Pathology
Faculty of Health Sciences
Institute of Infectious Diseases and Molecular Medicine
University of Cape Town
Anzio Road
Observatory
Cape Town 7925
South Africa
E-mail: collet.dandara@uct.ac.za
Abbreviations Used
a-SMA ¼alpha-smooth muscle actin
ATRA ¼all-trans-retinoic acid
CAFs ¼cancer-associated fibroblasts
CAMs ¼cancer-associated macrophages
CANs ¼cancer-associated neutrophils
CAR T ¼chimeric antigen receptor T
CSCs ¼cancer stem cells
ECM ¼extracellular matrix
EMT ¼epithelial to mesenchymal transition
EndMT ¼endothelial to mesenchymal transition
FAP ¼fibroblast activation protein
FGF ¼fibroblast growth factor
GPR77 ¼G-protein-coupled receptor 77
HGF ¼hepatocyte growth factor
HIF1-a¼hypoxia inducible factor 1-a
IL-6 ¼interleukin-6
JAK ¼Janus kinase
miRNAs ¼microRNAs
MMPs ¼matrix metalloproteases
MSCs ¼mesenchymal stem cells
PDAC ¼pancreatic ductal adenocarcinoma
PDGF ¼platelet-derived growth factor
PDGFR-b¼PDGF receptor-b
ROS ¼reactive oxygen species
SDF-1 ¼stromal-derived factor 1
SFRP2 ¼secreted frizzled-related protein-2
STAT ¼signal transducer and activator of transcription
STC1 ¼stanniocalcin-1
TAMs ¼tumor-associated macrophages
TGF-b¼transforming growth factor-beta
TIMP ¼tissue inhibitor of metallo proteinases
TME ¼tumor microenvironment
VEGF ¼vascular endothelial growth factor
VEGFA ¼vascular endothelial growth factor-A
CAFSIN TUMOR MICROENVIRONMENT 339
Downloaded by Swinburne University Of Technology from www.liebertpub.com at 06/05/20. For personal use only.
... Frontiers in Cell and Developmental Biology frontiersin.org (LUM), decorin (DCN), microfibril associated protein 5 (MFAP5), fibronectin, tenacin-C, periostin, and remodeling enzymes: lysyl oxidase (LOX), lysyl oxidase-like 1 (LOXL 1), and matrix metalloproteinase (MMP); and (4) growth factors and cytokines: TGF-β, vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), paternally expressed gene 2 (PEG2), connective tissue growth factor (CTGF), stromal cell-derived factor-1 (SDF-1), and WNT, and so on (Dzobo and Dandara, 2020;Aden et al., 2023;Xu et al., 2023). Additional research stemming from the diversity of CAFs includes whether the development of cancer or mutations in tumor suppressor genes within tumor cells can lead to the transformation of one subtype into another or the emergence of new subtypes. ...
... In recent years, exosomes have garnered increasing attention in tumor research due to their role in transporting different active compounds from cells, influencing immune escape, tumorigenesis, and TME reconstruction (Dzobo and Dandara, 2020). Other types of cells can secrete exosomes, which are extracellular vesicles with a diameter ranging from 30 to 150 nm. ...
Article
Full-text available
Cancer-associated fibroblasts (CAFs), a class of stromal cells in the tumor microenvironment (TME), play a key role in controlling cancer cell invasion and metastasis, immune evasion, angiogenesis, and resistance to chemotherapy. CAFs mediate their activities by secreting soluble chemicals, releasing exosomes, and altering the extracellular matrix (ECM). Exosomes contain various biomolecules, such as nucleic acids, lipids, and proteins. microRNA (miRNA), a 22–26 nucleotide non-coding RNA, can regulate the cellular transcription processes. Studies have shown that miRNA-loaded exosomes secreted by CAFs engage in various regulatory communication networks with other TME constituents. This study focused on the roles of CAF-derived exosomal miRNAs in generating cancer malignant characteristics, including immune modulation, tumor growth, migration and invasion, epithelial-mesenchymal transition (EMT), and treatment resistance. This study thoroughly examines miRNA’s dual regulatory roles in promoting and suppressing cancer. Thus, changes in the CAF-derived exosomal miRNAs can be used as biomarkers for the diagnosis and prognosis of patients, and their specificity can be used to develop newer therapies. This review also discusses the pressing problems that require immediate attention, aiming to inspire researchers to explore more novel avenues in this field.
... The CAFs seemed to have a different population of their subsets that rose from the heterogenic signaling from the TME based on the local sources [19]. The activation and the differentiation strongly depend on chemical regulators such as platelet-derived growth factor (PDGF), TGF-β, epidermal growth factor (EGF), and fibroblast growth factor 2 (FGF-2). ...
... Thus, CAFs offer a successful opportunity for drug discovery programs. Several studies have shown that CAFs are very stable and have fewer tendencies to develop resistance than cancer cells [12][13][14][15][16][17][18][19][20]. Moreover, they occupy spatially defined surroundings in TME and have higher proximity to the blood vessels. ...
Article
Full-text available
Tumor epithelial development and chemoresistance are highly promoted by the tumor microenvironment (TME), which is mostly made up of the cancer stroma. This is due to several causes. Cancer-associated fibroblasts (CAFs) stand out among them as being essential for the promotion of tumors. Understanding the fibroblastic population within a single tumor is made more challenging by the undeniable heterogeneity within it, even though particular stromal alterations are still up for debate. Numerous chemical signals released by tumors improve the connections between heterotypic fibroblasts and CAFs, promoting the spread of cancer. It becomes essential to have a thorough understanding of this complex microenvironment to effectively prevent solid tumor growth. Important new insights into the role of CAFs in the TME have been revealed by recent studies. The objective of this review is to carefully investigate the relationship between CAFs in tumors and plant secondary metabolites, with a focus on thymoquinone (TQ). The literature published between 2010 and 2023 was searched in PubMed and Google Scholar with keywords such as TQ, TME, cancer-associated fibroblasts, mechanism of action, and flavonoids. The results showed a wealth of data substantiating the activity of plant secondary metabolites, particularly TQ's involvement in blocking CAF operations. Scrutinized research also clarified the wider effect of flavonoids on pathways related to cancer. The present study highlights the complex dynamics of the TME and emphasizes the critical role of CAFs. It also examines the possible interventions provided by secondary metabolites found in plants, with TQ playing a vital role in regulating CAF function based on recent literature.
... Research has demonstrated that fibroblasts undergo a metamorphosis in circumstances of inflammation, fibrosis, and wound healing (8)(9)(10). Given that both inflammation and fibrosis are linked to the initiation and progression of cancer, fibroblasts are stimulated during these processes (11). These fibroblasts activated in the context of cancer are commonly referred to as CAFs (11). ...
... Given that both inflammation and fibrosis are linked to the initiation and progression of cancer, fibroblasts are stimulated during these processes (11). These fibroblasts activated in the context of cancer are commonly referred to as CAFs (11). Kojima et al. (12) demonstrated that autocrine transforming growth factor-beta (TGF-b) and stromal cell-derived factor-1 signaling induce the transformation of normal mammary fibroblasts into CAFs. ...
Article
Full-text available
Cancer-associated fibroblasts (CAFs) play a substantial role in the tumor microenvironment, exhibiting a strong association with the advancement of various types of cancer, including breast, pancreatic, and prostate cancer. CAFs represent the most abundant mesenchymal cell population in breast cancer. Through diverse mechanisms, including the release of cytokines and exosomes, CAFs contribute to the progression of breast cancer by influencing tumor energy metabolism, promoting angiogenesis, impairing immune cell function, and remodeling the extracellular matrix. Moreover, CAFs considerably impact the response to treatment in breast cancer. Consequently, the development of interventions targeting CAFs has emerged as a promising therapeutic approach in the management of breast cancer. This article provides an analysis of the role of CAFs in breast cancer, specifically in relation to diagnosis, treatment, drug resistance, and prognosis. The paper succinctly outlines the diverse mechanisms through which CAFs contribute to the malignant behavior of breast cancer cells, including proliferation, invasion, metastasis, and drug resistance. Furthermore, the article emphasizes the potential of CAFs as valuable tools for early diagnosis, targeted therapy, treatment resistance, and prognosis assessment in breast cancer, thereby offering novel approaches for targeted therapy and overcoming treatment resistance in this disease.
... Tumor pathogenesis and progression are modulated by both the cancer cells themselves and the tumor microenvironment (TME) [4]. The tumor stroma plays a critical role in enhancing tumor aggressiveness by modulating the deposition of the extracellular matrix (ECM) as well as cellular metabolism [5]. Of all TME stromal cells, cancer-associated fibroblasts (CAFs) have been the most widely examined. ...
Article
Full-text available
BACKGROUND Breast cancer (BC), a leading malignant disease, affects women all over the world. Cancer associated fibroblasts (CAFs) stimulate epithelial-mesenchymal transition, and induce chemoresistance and immunosuppression. AIM To establish a CAFs-associated prognostic signature to improve BC patient outcome estimation. METHODS We retrieved the transcript profile and clinical data of 1072 BC samples from The Cancer Genome Atlas (TCGA) databases, and 3661 BC samples from the The Gene Expression Omnibus. CAFs and immune cell infiltrations were quantified using CIBERSORT algorithm. CAF-associated gene identification was done by weighted gene co-expression network analysis. A CAF risk signature was established via univariate, least absolute shrinkage and selection operator regression, and multivariate Cox regression analyses. The receiver operating characteristic (ROC) and Kaplan-Meier curves were employed to evaluate the predictability of the model. Subsequently, a nomogram was developed with the risk score and patient clinical signature. Using Spearman's correlations analysis, the relationship between CAF risk score and gene set enrichment scores were examined. Patient samples were collected to validate gene expression by quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS Employing an 8-gene (IL18, MYD88, GLIPR1, TNN, BHLHE41, DNAJB5, FKBP14, and XG) signature, we attempted to estimate BC patient prognosis. Based on our analysis, high-risk patients exhibited worse outcomes than low-risk patients. Multivariate analysis revealed the risk score as an independent indicator of BC patient prognosis. ROC analysis exhibited satisfactory nomogram predictability. The area under the curve showed 0.805 at 3 years, and 0.801 at 5 years in the TCGA cohort. We also demonstrated that a reduced CAF risk score was strongly associated with enhanced chemotherapeutic outcomes. CAF risk score was significantly correlated with most hallmark gene sets. Finally, the prognostic signature were further validated by qRT-PCR. CONCLUSION We introduced a newly-discovered CAFs-associated gene signature, which can be employed to estimate BC patient outcomes conveniently and accurately.
... Notably α-smooth muscle actin (α-SMA)-rich cancer-associated fibroblasts (CAFs) govern the various stroma biological processes [8]. Following, α-SMA-expressing CAFs demonstrate a different gene expression pattern to normal fibroblasts and may confer CSC/oncogenic phenotypes on normal epithelial cells [9][10][11]. They enhance chemoresistance and cancer progression [12,13] through multiple processes, such as compositing and remodelling of the ECM, as well as secretion of several soluble factors that likely can interact with the cancer cells, and disruption of immunity [14][15][16]. ...
Article
Full-text available
Background: The tumor microenvironment (TME) plays a vital role in tumor progression through intricate molecular interactions. Cancer-associated fibroblasts (CAFs), notably those expressing alpha-smooth muscle actin (α-SMA) or myofibroblasts, are instrumental in this context and correlate with unfavorable outcomes in colorectal cancer (CRC). While several transcription factors influence TME, the exact regulator causing CAF dysregulation in CRC remains elusive. Prospero Homeobox 1 (PROX1) stands out, as its inhibition reduces α-SMA-rich CAF activity. However, the therapeutic role of PROX1 is debated due to inconsistent study findings. Methods: Using the ULCAN portal, we noted an elevated PROX1 level in advanced colon adenocarcinoma, linking to a poor prognosis. Assays determined the impact of PROX1 overexpression on CRC cell properties, while co-culture experiments spotlighted the PROX1-CAF relationship. Molecular expressions were validated by qRT-PCR and Western blots, with in vivo studies further solidifying the observations. Results: Our study emphasized the connection between PROX1 and α-SMA in CAFs. Elevated PROX1 in CRC samples correlated with increased α-SMA in tumors. PROX1 modulation influenced the behavior of specific CRC cells, with its overexpression fostering invasiveness. Kaplan-Meier evaluations demonstrated a link between PROX1 or α-SMA and survival outcomes. Consequently, PROX1, alone or with α-SMA, emerges as a CRC prognostic marker. Co-culture and animal experiments further highlighted this relationship. Conclusion: PROX1 appears crucial in modulating CRC behavior and therapeutic resistance within the TME by influencing CAFs, signifying the combined PROX1/α-SMA gene as a potential CRC prognostic marker. The concept of developing inhibitors targeting this gene set emerges as a prospective therapeutic strategy. However, this study is bound by limitations, including potential challenges in clinical translation, a focused exploration on PROX1/α-SMA potentially overlooking other significant molecular contributors, and the preliminary nature of the inhibitor development proposition.
... TME consists of diverse cellular components, including normal fibroblasts, cancer-associated fibroblasts (CAFs), immune cells, endothelial cells, etc., soluble molecules like chemokines, enzymes, cytokines, etc., and extracellular matrix (ECM). Notably, the interaction between fibroblasts and cancer cells not only promotes tumor development but also triggers the activation of fibroblasts [13]. The study by Krzysiek-Maczka and co-authors demonstrated that the infection with H. pylori induced the activation of gastric fibroblasts, leading to the induction of epithelial-to-mesenchymal transition in gastric RGM-1 cells through the secretion of transforming growth factor [14]. ...
Article
Full-text available
Background Fibroblasts, especially cancer-associated fibroblasts (CAFs), represent the predominant stromal cell population in the tumor microenvironment and have an important function in tumorigenesis by interacting with tumor cells. However, their interaction remains elusive in an inflammatory tumor microenvironment induced by Helicobacter pylori (H. pylori). Methods The expression of Serpin family E member 1 (Serpin E1) was measured in fibroblasts with or without H. pylori infection, and primary gastric cancer (GC) cells. Serpin E1 knockdown and overexpression fibroblasts were generated using Serpin E1 siRNA or lentivirus carrying Serpin E1. Co-culture models of fibroblasts and GC cells or human umbilical vein endothelial cells (HUVECs) were established with direct contact or the Transwell system. In vitro functional experiments and in vivo tumorigenesis assay were employed to study the malignant behaviors of GC cells interacting with fibroblasts. ELISA was used for quantifying the levels of Serpin E1 and VEGFA in the culture supernatant. The tube formation capacity of HUVECs was assessed using a tube formation assay. Recombinant human Serpin E1 (recSerpin E1), anti-Serpin E1 antibody, and a MAPK pathway inhibitor were utilized to treat HUVECs for elucidating the underlying molecular mechanisms. Results Serpin E1 was predominantly expressed in gastric CAFs. H. pylori infection significantly enhanced the expression and secretion of Serpin E1 by CAFs. Both fibroblast-derived Serpin E1 and recSerpin E1 enhanced the growth, invasion, and migration of GC cells, along with increased VEGFA expression and tube formation in HUVECs. Furthermore, the co-inoculation of GC cells and fibroblasts overexpressing Serpin E1 triggered the expression of Serpin E1 in cancer cells, which facilitated together xenograft tumor growth and peritoneal dissemination of GC cells in nude mice, with an increased expression of Ki67, Serpin E1, CD31 and/or VEGFA. These processes may be mediated by Serpin E1-induced migration and p38 MAPK/VEGFA-mediated angiogenesis of HUVECs. Conclusion H. pylori infection induces Serpin E1 expression in fibroblasts, subsequently triggering its expression in GC cells through their interaction. Serpin E1 derived from these cells promotes the migration and p38 MAPK/VEGFA-mediated angiogenesis of HUVECs, thereby facilitating GC growth and peritoneal metastasis. Targeting Serpin E1 signaling is a potential therapy strategy for H. pylori-induced GC.
... The tumor microenvironment consists of many cell types that regulate tumor growth and progression [4]. The role of various subpopulations of T cells [5], myeloid suppressor cells [6], tumor-associated macrophages [7], neutrophils [8], and fibroblasts [9] in the regulation of antitumor immune responses was established after many years of research. It was shown that a population of erythroid progenitor cells with immunoregulatory properties could also be involved in the regulation of the local and systemic antitumor immune response [10]. ...
Article
Full-text available
The tumor microenvironment is an important factor that can determine the success or failure of antitumor therapy. Cells of hematopoietic origin are one of the most important mediators of the tumor–host interaction and, depending on the cell type and functional state, exert pro- or antitumor effects in the tumor microenvironment or in adjacent tissues. Erythroid cells can be full members of the tumor microenvironment and exhibit immunoregulatory properties. Tumor growth is accompanied by the need to obtain growth factors and oxygen, which stimulates the appearance of the foci of extramedullary erythropoiesis. Tumor cells create conditions to maintain the long-term proliferation and viability of erythroid cells. In turn, tumor erythroid cells have a number of mechanisms to suppress the antitumor immune response. This review considers current data on the existence of erythroid cells in the tumor microenvironment, formation of angiogenic clusters, and creation of optimal conditions for tumor growth. Despite being the most important life-support function of the body, erythroid cells support tumor growth and do not work against it. The study of various signaling mechanisms linking tumor growth with the mobilization of erythroid cells and the phenotypic and functional differences between erythroid cells of different origin allows us to identify potential targets for immunotherapy.
Article
Full-text available
Background Osteosarcoma (OS) stands out as the most common bone tumor, with approximately 20% of the patients receiving a diagnosis of metastatic OS at their initial assessment. A significant challenge lies in the frequent existence of undetected metastases during the initial diagnosis. Mesenchymal stem cells (MSCs) possess unique abilities that facilitate tumor growth, and their interaction with OS cells is crucial for metastatic spread. Methods and results We demonstrated that, in vitro, MSCs exhibited a heightened migration response toward the secretome of non-metastatic OS cells. When challenged to a secretome derived from lungs preloaded with OS cells, MSCs exhibited greater migration toward lungs colonized with metastatic OS cells. Moreover, in vivo, MSCs displayed preferential migratory and homing behavior toward lungs colonized by metastatic OS cells. Metastatic OS cells, in turn, demonstrated an increased migratory response to the MSCs’ secretome. This behavior was associated with heightened cathepsin D (CTSD) expression and the release of active metalloproteinase 2 (MMP2) by metastatic OS cells. Conclusions Our assessment focused on two complementary tumor capabilities crucial to metastatic spread, emphasizing the significance of inherent cell features. The findings underscore the pivotal role of signaling integration within the niche, with a complex interplay of migratory responses among established OS cells in the lungs, prometastatic OS cells in the primary tumor, and circulating MSCs. Pulmonary metastases continue to be a significant factor contributing to OS mortality. Understanding these mechanisms and identifying differentially expressed genes is essential for pinpointing markers and targets to manage metastatic spread and improve outcomes for patients with OS.
Article
Full-text available
Extracellular biophysical properties have particular implications for a wide spectrum of cellular behaviors and functions, including growth, motility, differentiation, apoptosis, gene expression, cell–matrix and cell–cell adhesion, and signal transduction including mechanotransduction. Cells not only react to unambiguously mechanical cues from the extracellular matrix (ECM), but can occasionally manipulate the mechanical features of the matrix in parallel with biological characteristics, thus interfering with downstream matrix-based cues in both physiological and pathological processes. Bidirectional interactions between cells and (bio)materials in vitro can alter cell phenotype and mechanotransduction, as well as ECM structure, intentionally or unintentionally. Interactions between cell and matrix mechanics in vivo are of particular importance in a variety of diseases, including primarily cancer. Stiffness values between normal and cancerous tissue can range between 500 Pa (soft) and 48 kPa (stiff), respectively. Even the shear flow can increase from 0.1–1 dyn/cm2 (normal tissue) to 1–10 dyn/cm2 (cancerous tissue). There are currently many new areas of activity in tumor research on various biological length scales, which are highlighted in this review. Moreover, the complexity of interactions between ECM and cancer cells is reduced to common features of different tumors and the characteristics are highlighted to identify the main pathways of interaction. This all contributes to the standardization of mechanotransduction models and approaches, which, ultimately, increases the understanding of the complex interaction. Finally, both the in vitro and in vivo effects of this mechanics–biology pairing have key insights and implications for clinical practice in tumor treatment and, consequently, clinical translation.
Article
Thoracic SMARCA4-deficient undifferentiated tumor (SMARCA4-UT) is a rare malignant disease. We present the case of a 56-year-old woman with thoracic SMARCA4-UT presenting as a mediastinal mass who underwent 68 Ga-DOTA-FAPI-04 PET/CT imaging. Intense 68 Ga-DOTA-FAPI-04 uptake was observed in the primary tumor and lymph node metastases. After 7 cycles of immune checkpoint inhibitor plus chemotherapy, the patient underwent mediastinal mass resection, and postoperative pathology confirmed a complete pathologic response. This case may provide valuable insights into the diagnosis and monitoring of the treatment response of thoracic SMARCA4-UT.
Article
Full-text available
Transforming growth factor β (TGF-β) is a secreted cytokine that regulates cell proliferation, migration, and the differentiation of a plethora of different cell types. Consistent with these findings, TGF-β plays a key role in controlling embryogenic development, inflammation, and tissue repair, as well as in maintaining adult tissue homeostasis. TGF-β elicits a broad range of context-dependent cellular responses, and consequently, alterations in TGF-β signaling have been implicated in many diseases, including cancer. During the early stages of tumorigenesis, TGF-β acts as a tumor suppressor by inducing cytostasis and the apoptosis of normal and premalignant cells. However, at later stages, when cancer cells have acquired oncogenic mutations and/or have lost tumor suppressor gene function, cells are resistant to TGF-β-induced growth arrest, and TGF-β functions as a tumor promotor by stimulating tumor cells to undergo the so-called epithelial-mesenchymal transition (EMT). The latter leads to metastasis and chemotherapy resistance. TGF-β further supports cancer growth and progression by activating tumor angiogenesis and cancer-associated fibroblasts and enabling the tumor to evade inhibitory immune responses. In this review, we will consider the role of TGF-β signaling in cell cycle arrest, apoptosis, EMT and cancer cell metastasis. In particular, we will highlight recent insights into the multistep and dynamically controlled process of TGF-β-induced EMT and the functions of miRNAs and long noncoding RNAs in this process. Finally, we will discuss how these new mechanistic insights might be exploited to develop novel therapeutic interventions.
Article
Full-text available
Postnatal muscle growth and exercise-or injury-induced regeneration are facilitated by myoblasts. Myoblasts respond to a variety of proteins such as cytokines that activate various signaling cascades. Cytokines belonging to the interleukin 6 superfamily (IL-6) influence myoblasts' proliferation but their effect on differentiation is still being researched. The Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway is one of the key signaling pathways identified to be activated by IL-6. The aim of this study was to investigate myoblast fate as well as activation of JAK-STAT pathway at different physiologically relevant IL-6 concentrations (10 pg/mL; 100 pg/mL; 10 ng/mL) in the C2C12 mouse myoblast cell line and primary human myoblasts, isolated from eight young healthy male volunteers. Myoblasts' cell cycle progression, proliferation and differentiation in vitro were assessed. Low IL-6 concentrations facilitated cell cycle transition from the quiescence/Gap1 (G0/G1) to the synthesis (S-) phases. Low and medium IL-6 concentrations decreased the expression of myoblast determination protein 1 (MyoD) and myogenin and increased proliferating cell nuclear antigen (PCNA) expression. In contrast, high IL-6 concentration shifted a larger proportion of cells to the pro-differentiation G0/G1 phase of the cell cycle, substantiated by significant increases of both MyoD and myogenin expression and decreased PCNA expression. Low IL-6 concentration was responsible for prolonged JAK1 activation and increased suppressor of cytokine signaling 1 (SOCS1) protein expression. JAK-STAT inhibition abrogated IL-6-mediated C2C12 cell proliferation. In contrast, high IL-6 initially increased JAK1 activation but resulted in prolonged JAK2 activation and elevated SOCS3 protein expression. High IL-6 concentration decreased interleukin-6 receptor (IL-6R) expression 24 h after treatment whilst low IL-6 concentration increased IL-6 receptor (IL-6R) expression at the same time point. In conclusion, this study demonstrated that IL-6 has concentration-and time-dependent effects on both C2C12 mouse myoblasts and primary human myoblasts. Low IL-6 concentration induces proliferation whilst high IL-6 concentration induces differentiation. These effects are mediated by specific components of the JAK/STAT/SOCS pathway.
Article
Full-text available
The promise of regenerative medicine and tissue engineering is founded on the ability to regenerate diseased or damaged tissues and organs into functional tissues and organs or the creation of new tissues and organs altogether. In theory, damaged and diseased tissues and organs can be regenerated or created using different configurations and combinations of extracellular matrix (ECM), cells, and inductive biomolecules. Regenerative medicine and tissue engineering can allow the improvement of patients' quality of life through availing novel treatment options. The coupling of regenerative medicine and tissue engineering with 3D printing, big data, and computational algorithms is revolutionizing the treatment of patients in a huge way. 3D bioprinting allows the proper placement of cells and ECMs, allowing the recapitulation of native microenvironments of tissues and organs. 3D bioprinting utilizes different bioinks made up of different formulations of ECM/biomaterials, biomolecules, and even cells. The choice of the bioink used during 3D bioprinting is very important as properties such as printability, compatibility, and physical strength influence the final construct printed. The extracellular matrix (ECM) provides both physical and mechanical microenvironment needed by cells to survive and proliferate. Decellularized ECM bioink contains biochemical cues from the original native ECM and also the right proportions of ECM proteins. Different techniques and characterization methods are used to derive bioinks from several tissues and organs and to evaluate their quality. This review discusses the uses of decellularized ECM bioinks and argues that they represent the most biomimetic bioinks available. In addition, we briefly discuss some polymer-based bioinks utilized in 3D bioprinting.
Article
Full-text available
Among all the stromal cells that present in the tumor microenvironment, cancer-associated fibroblasts (CAFs) are one of the most abundant and critical components of the tumor mesenchyme, which not only provide physical support for tumor cells but also play a key role in promoting and retarding tumorigenesis in a context-dependent manner. CAFs have also been involved in the modulation of many components of the immune system, and recent studies have revealed their roles in immune evasion and poor responses to cancer immunotherapy. In this review, we describe our current understanding of the tumorigenic significance, origin, and heterogeneity of CAFs, as well as the roles of different CAFs subtypes in distinct immune cell types. More importantly, we highlight potential therapeutic strategies that target CAFs to unleash the immune system against the tumor.
Article
Full-text available
Heterogeneous subtypes of cancer-associated fibroblasts (CAFs) coexist within pancreatic cancer tissues and can both promote and restrain disease progression. Here, we interrogate how cancer cells harboring distinct alterations in p53 manipulate CAFs. We reveal the existence of a p53-driven hierarchy, where cancer cells with a gain-of-function (GOF) mutant p53 educate a dominant population of CAFs that establish a pro-metastatic environment for GOF and null p53 cancer cells alike. We also demonstrate that CAFs educated by null p53 cancer cells may be reprogrammed by either GOF mutant p53 cells or their CAFs. We identify perlecan as a key component of this pro-metastatic environment. Using intravital imaging, we observe that these dominant CAFs delay cancer cell response to chemotherapy. Lastly, we reveal that depleting perlecan in the stroma combined with chemotherapy prolongs mouse survival, supporting it as a potential target for anti-stromal therapies in pancreatic cancer.
Article
Full-text available
Malignant tumors are highly heterogeneous and likely contain a subset of cancer cells termed cancer stem cells (CSCs). CSCs exist in a dynamic equilibrium with their microenvironments and the CSC phenotype is tightly regulated by both cell-intrinsic and cell-extrinsic factors including those derived from their surrounding cells or stroma. Many human solid tumors like breast, lung, colorectal and pancreatic cancers are characterized by a pronounced stromal reaction termed “the desmoplastic response.” Carcinoma-associated fibroblasts (CAFs) derived either from resident fibroblasts or tumor-infiltrating mesenchymal stem cells (MSCs) are a major component of the stroma in desmoplastic cancers. Recent studies identified subpopulations of CAFs proficient in secreting a plethora of factors to foster CSCs, tumor growth, and invasion. In addition, cytotoxic therapy can lead to the enrichment of functionally perturbed CAFs, which are endowed with additional capabilities to enhance cancer stemness, leading to treatment resistance and tumor aggressiveness. When recruited into the tumor stroma, bone-marrow-derived MSCs can promote cancer stemness by secreting a specific set of paracrine factors or converting into pro-stemness CAFs. Thus, blockade of the crosstalk of pro-stemness CAFs and MSCs with CSCs may provide a new avenue to improving the therapeutic outcome of desmoplastic tumors. This up-to-date, in-depth and balanced review describes the recent progress in understanding the pro-stemness roles of CAFs and tumor-infiltrating MSCs and the associated paracrine signaling processes. We emphasize the effects of systemic chemotherapy on the CAF/MSC–CSC interplay. We summarize various promising and novel approaches in mitigating the stimulatory effect of CAFs or MSCs on CSCs that have shown efficacies in preclinical models of desmoplastic tumors and highlight the unique advantages of CAF- or MSC-targeted therapies. We also discuss potential challenges in the clinical development of CSC- or MSC-targeted therapies and propose CAF-related biomarkers that can guide the next-generation clinical studies.
Article
Full-text available
Background and purpose: Colorectal cancer (CRC) frequently metastasizes to the liver, which involves the participation of multiple cytokines. Tumor microenvironment (TME) composed of cancer-associated fibroblasts (CAFs) and tumor cells acts as an essential factor in cancer metastasis. Transforming growth factor β1 (TGFβ1) is a vital cytokine involved in migration and invasion of cancer cells. However, the underlying mechanisms remain unclear. In the present study, we aimed to investigate the role and molecular mechanisms of TGFβ1 in TME. Methods: The conditioned medium prepared from colorectal cancer HCT116 and HT29 cells was used to culture mesenchymal stem cells (MSCs). The differentiation of MSCs to CAFs was detected by flow cytometry. The role of TGFβ1 in colorectal cancer cells metastasis was examined by wound-healing assay and transwell assay. And the activation of the Janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) signaling pathway was measured by Western blot assay. Results: TGFβ1 induced the differentiation of MSCs to CAFs and improved HCT116 and HT29 cells migration and invasion. Meanwhile, TGFβ1 also upregulated the phosphorylation of STAT3 and enhanced the nuclear localization of p-STAT3, which activated JAK/STAT3 signaling pathway. Conclusion: TGFβ1 induced the differentiation of MSCs into CAFs and promoted the migration and invasion of HCT116 and HT29 cells, which depended on the activation of JAK/STAT3 signaling pathway.
Article
Full-text available
Tumor cells reside in a highly complex and heterogeneous tumor microenvironment (TME), which is composed of a myriad of genetically stable non-cancer cells, including fibroblasts, immune cells, endothelial cells, and epithelial cells, and a tumor-specific extracellular matrix (ECM). Cancer-associated fibroblasts (CAFs), as an abundant and active stromal cell population in the TME, function as the signaling center and remodeling machine to aid the creation of a desmoplastic tumor niche. Although there is no denial that the TME and CAFs may have anti-tumor effects as well, a great deal of findings reported in recent years have convincingly revealed the tumor-promoting effects of CAFs and CAF-derived ECM proteins, enzymes, chemical factors and other downstream effectors. While there is growing enthusiasm for the development of CAF-targeting therapies, a better understanding of the complexities of CAF-ECM and CAF-cancer cell interactions is necessary before novel therapeutic strategies targeting the malignant tumor “soil” can be successfully implemented in the clinic.
Article
Full-text available
The human epigenome plays a key role in determining cellular identity and eventually function. Drug discovery undertakings have focused mainly on the role of genomics in carcinogenesis, with the focus turning to the epigenome recently. Drugs targeting DNA and histone modifications are under development with some such as 5-azacytidine, decitabine, vorinostat, and panobinostat already approved by the Food and Drug Administration (FDA) and the European Medicines Agency (EMA). This expert review offers a critical analysis of the epigenomics-guided drug discovery and development and the opportunities and challenges for the next decade. Importantly, the coupling of epigenetic editing techniques, such as clustered regularly interspersed short pal-indromic repeat (CRISPR)-CRISPR-associated protein-9 (Cas9) and APOBEC-coupled epigenetic sequencing (ACE-seq) with epigenetic drug screens, will allow the identification of small-molecule inhibitors or drugs able to reverse epigenetic changes responsible for many diseases. In addition, concrete and sustainable innovation in cancer treatment ought to integrate epigenome targeting drugs with classic therapies such as chemotherapy and immunotherapy.
Article
The most abundant cell type in the tumor microenvironment are cancer-associated fibroblasts (CAFs). CAFs play an important role in tumor growth and progression. Besides direct communication with cancer cells via secreted molecules or cell-cell adhesions, CAFs also indirectly affect cancer cell behavior by remodeling the extracellular matrix (ECM). Here, we summarize recent findings on the distinct mechanisms that CAFs use to modify ECM, specifically, their proteolytic versus force-dependent activity. We then review the consequences of CAF force transmission on the physico-chemical properties of the matrix, focusing on the deposition of new matrix components, and the alteration of the organization and stiffness of the ECM. CAFs promote tumor invasion by creating the roads cancer cells use to escape the tumor mass. However, there is also evidence that CAFs can prevent invasion, possibly by forming a physical barrier around the tumor edge. We discuss the controversial role of CAFs in tumor progression.