You are on page 1of 12

0022-5347/01/1666-2472/0

THE JOURNAL OF UROLOGY® Vol. 166, 2472–2483, December 2001


Copyright © 2001 by AMERICAN UROLOGICAL ASSOCIATION, INC.® Printed in U.S.A.

Review Article

REACTIVE STROMA IN PROSTATE CANCER PROGRESSION


JENNIFER A. TUXHORN, GUSTAVO E. AYALA AND DAVID R. ROWLEY
From the Departments of Molecular and Cellular Biology and Pathology, Baylor College of Medicine, Houston, Texas

ABSTRACT

Purpose: The development of an altered stromal microenvironment in response to carcinoma is a


common feature of many tumors. We reviewed the literature describing characteristics of reactive
stroma, how reactive stroma affects cancer progression and how carcinoma regulates reactive stroma.
Moreover, we present a hypothesis of reactive stroma in prostate cancer and discuss how the biology
of reactive stroma may be used in novel diagnostic and therapeutic approaches.
Materials and Methods: An extensive literature search was performed to review reports of the
general features of wound repair stroma, general stromal responses to carcinoma, and stromal
biology of normal and prostate cancer tissues. These studies were analyzed and a reactive stroma
hypothesis in prostate cancer was developed.
Results: Modifications to the stroma of breast, colon and prostate tumors parallel the gener-
ation of granulation tissue in wound repair. These changes include stromal cell phenotypic
switching, extracellular matrix remodeling and angiogenesis induction. Therefore, it is predicted
that a modified wound healing response induces the formation of reactive stroma in cancer to
create a tumor promoting environment. Based on its role in wound repair and its over expression
in prostate cancer, transforming growth factor-␤ stands out as a potential regulator of reactive
stroma.
Conclusions: Reactive stroma in prostate cancer and granulation tissue in wound repair show
similar biological responses and processes that are predicted to promote cancer progression.
Further identification of specific functional and regulatory mechanisms in prostate cancer
reactive stroma may aid in the use of reactive stroma for novel diagnostic and therapeutic
approaches.
KEY WORDS: prostate, prostatic neoplasms, growth substances, granulation tissue, extracellular matrix

Cancer progression is a complex multistep process that may be an attractive target for additional diagnostic and
begins with the transformation of normal cells and continues therapeutic approaches to prostate cancer.
with tumor growth, invasion and metastasis. In addition to
transformed carcinoma cells, tumors also contain stromal
BIOLOGY OF THE STROMA
cells, extracellular matrix, newly formed blood vessels and
immune components. However, there is considerable evi- Structure and function. The stroma was once viewed as a
dence that tumor stroma is different than the stroma in passive support structure that contributed little to the over-
normal tissue. To maintain tissue homeostasis the host stro- all biological function of tissues. However, it is now under-
mal compartment reacts to carcinoma in a process similar to stood that the stroma is a dynamic environment that directly
the general wound repair pathway. The result of this re- influences epithelial cell behavior and performs tissue repair
sponse is the creation of a new stromal microenvironment, in response to injury. The stromal compartment is a complex
into which carcinoma grows and invades. Studies indicate arrangement of stromal cells and extracellular matrix plus
that this new reactive stroma environment enhances tumor- associated growth factors, regulatory molecules and remod-
igenesis by supporting cancer cell survival, proliferation and eling enzymes. Blood vessels, nerves and immune cells are
migration, and by inducing angiogenesis. Therefore, it is also integral parts of the stroma (fig. 1, A). These compo-
becoming more clear that cancer progression is not exclu- nents act in a coordinated manner to regulate cell function
sively regulated by the disruption of oncogene and tumor and maintain overall tissue homeostasis.
suppressor pathways in cancer cells. Much evidence now Fibroblasts and smooth muscle cells are the principal stro-
suggests that cancer progression also co-depends on the stro- mal cell types in the human prostate gland. A primary func-
mal compartment to create a more tumor promoting micro- tion of these cells is to synthesize the structural and regula-
environment. In this review we discuss the role of stroma in tory components of the extracellular matrix. The
maintaining tissue homeostasis and present mechanisms by extracellular matrix is a meshwork of fibrillar proteins, ad-
which carcinoma may induce the host programmed wound hesive glycoproteins and proteoglycans.1 Moreover, it is a
healing response. In addition, we consider how the genera- reservoir of active and latent growth factors.2 Structural
tion of this reactive stroma environment may promote cancer components, such as collagen and elastic fibers, provide me-
progression. We also propose that the stromal compartment chanical strength and flexibility to the tissue. These compo-
Supported by National Institutes of Health Grants RO1 CA58093, nents also serve as a substrate for cell attachment and mi-
RO1 DK45909, SPORE CA58204 and UO1 CA84296. gration, which are mediated by adhesive glycoproteins, such
2472
REACTIVE STROMA IN PROSTATE CANCER PROGRESSION 2473
growth factors and cytokines released by degranulating
platelets. Activated macrophages also secrete growth factors,
such as transforming growth factor-␤ (TGF-␤), platelet de-
rived growth factor (PDGF), fibroblast growth factor-2 and
vascular endothelial growth factor (VEGF), which induce
angiogenesis and fibroplasia. Quiescent fibroblasts from sur-
rounding tissue migrate to the wound site, where they pro-
liferate and produce extracellular matrix components, such
as collagen types I and III, fibronectin and proteoglycans.
This collection of macrophages, fibroblasts and new blood
FIG. 1. A, normal human prostate gland is composed of secretory vessels embedded within a loose matrix is called granulation
epithelium surrounded by stroma containing fibroblasts, smooth tissue. Epithelial cells are also stimulated to proliferate and
muscle cells (SMC), structural and regulatory components of extra- migrate through the granulation tissue to reestablish the
cellular matrix (ECM), blood vessels, nerves and immune cells. Re-
ciprocal stromal-epithelial interactions regulate normal tissue struc- epithelial layer. As the wound continues to heal, granulation
ture and function. B, prostate carcinoma cells proliferate and invade tissue fibroblasts differentiate into myofibroblasts, which de-
through basement membrane into stroma, which responds to repair velop characteristics of smooth muscle to assist in wound
disruption in tissue homeostasis, creating new stromal microenvi- closure.8 Fibroblasts and myofibroblasts continue to secrete
ronment termed reactive stroma. Reactive stroma shows stromal cell
phenotypic switching to myofibroblasts, extracellular matrix remod- large amounts of extracellular matrix, especially collagen
eling, elevated angiogenesis and influx of tumor associated macro- type I, as well as proteases, which degrade extracellular
phages (TAM). matrix components. The balance of extracellular matrix pro-
duction and degradation results in remodeling of the granu-
lation tissue to form a scar.3, 9 –11
as fibronectin and laminin. Proteoglycans regulate extracel- A similar repair process is observed in vascular injury.3, 12
lular matrix structure and permeability.3 They also bind to Platelets adhere to damaged endothelium in vessels and
and modulate the activity of growth factors, proteases and aggregate to form a clot, which recruits infiltration of inflam-
protease inhibitors.1, 2 matory cells. Growth factors and cytokines released from
Together stromal cells and the extracellular matrix create activated platelets and macrophages induce migration of
a microenvironment that regulates the growth and func- smooth muscle cells from the media of the vessel wall to the
tional differentiation of adjacent cells.4 For example, it is well intima. Smooth muscle cells that migrate to the vessel intima
established that prostate smooth muscle cells mediate appear to dedifferentiate from a quiescent contractile pheno-
stromal-epithelial interactions that are important for the type to a synthetic proliferative phenotype.13, 14 In the intima
development and maintenance of tissue differentiation.5 The the smooth muscle cells are induced to proliferate and secrete
inductive capability of extracellular matrix is also evident extracellular matrix components, including collagen, fi-
when studied in vitro. Culturing primary human prostate bronectin and glycosaminoglycans, to generate a modified
epithelium on reconstituted basement membrane extracellu- granulation tissue. During the final phase extracellular ma-
lar matrix induced cells to show a clustered morphology and trix remodeling occurs and the endothelium grows to cover
increase the secretion of prostate specific antigen and pros- the healed area.
tatic acid phosphatase.6 In contrast, when grown on tissue As these examples illustrate, the stromal compartment
culture plastic, these cells showed elevated proliferation and responds to injury by creating a microenvironment that is
reduced synthesis of prostate specific secretory proteins. It is fundamentally different from normal stroma. The granula-
clear that activation of growth factor receptors and extracel- tion tissue environment drives the repair process by recruit-
lular matrix interactions through integrins function to initi- ing immune cells, inducing stromal cell activation and secre-
ate intracellular signaling cascades that control cell motility, tion of extracellular matrix, growth factors and remodeling
proliferation and differentiation. Accordingly stromal regu- enzymes, stimulating angiogenesis and promoting epithelial
lation of cell behavior likely occurs by mobilizing extracellu- cell proliferation and migration. These growth promoting
lar matrix bound growth factors and by modulating direct conditions are necessary to repair the wound rapidly and
cell-extracellular matrix interactions.7 reestablish tissue homeostasis.
In addition to producing the extracellular matrix compo- Stromal cell phenotypic switching: the myofibroblast. As
nents and growth factors necessary to regulate normal tissue described, a key feature of the stromal response in wound
function, the stromal compartment is also responsible for repair is the ability of stromal cells to show plasticity or
tissue repair in response to injury. Thus, the stroma must be phenotypic switching.13, 15 Fibroblasts and smooth muscle
poised to respond rapidly to changes in homeostasis. Two key cells can reversibly regulate their proliferation rate and phe-
characteristics of the stromal compartment enable this to notype in response to the state of tissue homeostasis. Under
occur, namely stromal cell plasticity and extracellular matrix normal conditions these cells have a low proliferative index
organization. The storage of latent growth factors, proteases and likely secrete only the factors necessary to maintain
and protease inhibitors in the extracellular matrix allows for normal tissue function.13 However, in response to pathologi-
immediate mobilization of these factors to regulate extracel- cal situations stromal cells rapidly modulate to a reactive
lular matrix and tissue remodeling.2 Therefore, the stroma is phenotype, which proliferates and synthesizes extracellular
designed to effect a timely response to wounding and to matrix, growth factors and remodeling enzymes necessary
maintain tissue homeostasis. for tissue repair.
Stromal response in wound repair. The process of wound It appears that fibroblasts and vascular smooth muscle
healing exemplifies the complexity of stromal biology, in that cells modulate to a common myofibroblast phenotype during
it integrates all components of the stromal compartment in wound repair.16 The myofibroblast is described as an inter-
response to disruptions in tissue homeostasis. The wound mediate between fibroblasts and smooth muscle cells based
repair response is typically initiated when injury to the skin on cytoskeletal protein expression and ultrastructural fea-
or internal tissues results in the rupture of blood vessels. tures.8 Fibroblasts in granulation tissue switch to a myofi-
Spillage of blood into the interstitial matrix leads to platelet broblast phenotype, which is identified by vimentin and
aggregation and clot formation. The initial clot, which is smooth muscle ␣-actin expression. These cells are also char-
composed of fibrin, fibronectin and platelets, serves as a acterized by formation of contractile filaments, a well devel-
provisional matrix for cell migration. Neutrophils, macro- oped rough endoplasmic reticulum and Golgi apparatus, cell
phages and stromal cells are recruited to the wound site by attachments to the extracellular matrix, and gap junctions.15
2474 REACTIVE STROMA IN PROSTATE CANCER PROGRESSION

In response to vascular injury smooth muscle cells switch sis and an influx of inflammatory cells (fig. 1, B). It has been
from a quiescent contractile phenotype to a proliferative syn- suggested that the stromal response to cancer is a modified
thetic phenotype, which also shows the features of myofibro- wound repair response10, 23–25 and, therefore, the reactive
blasts.14 Cells with the myofibroblast phenotype increase stromal environment is likely to mimic granulation tissue.
vimentin expression and decrease expression of markers that These conditions would be predicted to promote tumorigen-
identify smooth muscle cells, such as calponin and smooth esis. Although the concept of reactive stroma in human pros-
muscle myosin heavy chain. In addition, a reduction in mi- tate cancer is just emerging,24 several studies to date have
crofilaments is accompanied by an increase in “synthetic described aspects of reactive stroma and shown that they are
cellular organelles, such as Golgi and rough endoplasmic associated with prostate cancer progression.
reticulum.”13 Gap junctions and focal tight junctions have Stromal cell phenotype. Perhaps the most common marker
also been reported.17 That these cells switch to a common of reactive stroma in cancer is the appearance of activated
phenotype is further supported by the observation that fibro- stromal cells with myofibroblastic characteristics. Cells with
blasts and arterial smooth muscle cells show myofibroblast the ultrastructural features of myofibroblasts have been ob-
characteristics when cultured in vitro.15, 18 served in the stroma of various types of invasive and meta-
Together these studies suggest that stromal cells show static carcinoma, including that of the breast, lung, colon,
plasticity via a continuum of phenotypes ranging from fibro- stomach and prostate.26 Smooth muscle ␣-actin has become
blast to myofibroblast to smooth muscle (fig. 2). Furthermore, the marker most often used to identify myofibroblasts by
stromal cells appear to modulate between the phenotypes immunohistochemistry.15 This test is effective in tissues in
based on physiological and pathological stimuli13, 15 with the which normal interstitial stromal cells do not express smooth
myofibroblast as the principal cell type in pathological con- muscle ␣-actin, as in the breast.27 However, smooth muscle
ditions. Cells characterized as myofibroblasts have been re- cells in normal prostate stroma express ␣-actin. Thus, it is
ported in numerous pathological processes, including wound necessary to examine multiple markers to distinguish myo-
healing granulation tissue, hypertrophic scars, fibromatoses fibroblasts from mature smooth muscle cells in the prostate.
and stromal response to neoplasia.19 Accordingly it is pre- We have previously examined the expression of vimentin,
dicted that the stromal phenotypic switch to an activated smooth muscle ␣-actin and calponin, a later stage marker of
myofibroblast cell type is a generic host response to disrup- smooth muscle differentiation,13 in the reactive stroma of
tions in tissue homeostasis that require repair. Therefore, Gleason 3 prostate cancer by immunohistochemistry. Our
myofibroblasts would be expected to be present in almost any preliminary results showed an increase in vimentin staining
physical or biological wound, ranging from a laceration to and a decrease in calponin staining, while the level of ␣-actin
carcinogenesis. Moreover, activation of stromal cells to the remained essentially the same.28 These observations suggest
reactive myofibroblast phenotype and their subsequent con- that the primary stromal cell type in prostate cancer reactive
tribution of extracellular matrix components, growth factors stroma is the myofibroblast. These data are consistent with
and proteases to the stromal microenvironment likely have observations by Hayward et al that “zones immediately sur-
an important role in the progression of these pathological rounding foci of prostatic carcinoma cells contained a fibro-
conditions. blastic stroma devoid of smooth muscle cells.”5 The loss of
smooth muscle in human prostate cancer is clearly illus-
ALTERED STROMAL BIOLOGY IN CANCER: REACTIVE STROMA trated in specimens stained with Masson’s trichrome. Fig-
ure 3 shows that normal prostate stroma is composed of
Carcinoma cell proliferation and invasion through the
red staining smooth muscle cells surrounded by blue stain-
basement membrane into the surrounding stroma would be
ing collagen fibers, while the reactive stoma surrounding
classified as a disruption in normal tissue homeostasis. Due
Gleason 3 prostate cancer is predominantly blue with rel-
to its primary role in wound repair it is expected that the
atively few scattered smooth muscle cells. Together these
stroma would respond in an effort to repair the damage.
data suggest that human prostate cancer reactive stroma
Modifications in stromal cell phenotype and extracellular
is characterized by a switch in stromal cell phenotype and
matrix composition have been reported in many types of
reactive stroma is composed primarily of myofibroblasts.
human cancer, particularly breast and colon cancer.20 –22
Since fibroblasts and vascular smooth muscle cells have
These changes have previously been referred to as desmopla-
the potential to switch to the myofibroblast phenotype during
sia (the formation of abundant collagenous stroma) or the
wound repair, it is possible that myofibroblasts in cancer
stromal response/stromal reaction. However, we use the term
reactive stroma may be derived from either cell type. A study
reactive stroma to describe the genesis of a new stromal
in which breast cancer cells were co-cultured with fibroblasts
microenvironment that is created in response to carcinoma
or vascular smooth muscle cells in 3-dimensional collagen
and accompanies tumor progression. The reactive stroma in
gels explored this issue.27 In fibroblast and cancer cell co-
cancer is typified by stromal cell phenotypic switching, ex-
tracellular matrix remodeling, increased growth factor bio- cultures the cells showed morphological organization similar
availability, elevated protease activity, increased angiogene- to that of in vivo tumors and the fibroblasts “readily con-
verted to myofibroblasts,”27 as assayed by ␣-actin expression.
Although vascular smooth muscle cell and cancer cell co-
cultures resulted in morphology similar to that of the fibro-
blast/cancer cell co-cultures, smooth muscle cells maintained
the expression of ␣-actin, calponin, and smooth muscle myo-
sin. These results indicate that fibroblasts are the primary
source of myofibroblasts in the reactive stroma of breast
tumors. To our knowledge it is not known whether myofibro-
blasts in human prostate tumors originate from fibroblasts,
smooth muscle cells or both cell types. In vitro studies have
FIG. 2. Stromal cells show phenotypic switching or plasticity.
shown that prostate fibroblasts can modulate to the myofibro-
Continuum of phenotypes from fibroblast to myofibroblast to smooth blast phenotype.29, 30 In addition, Hayward et al suggested that
muscle cells is observed based on physiological and pathological loss of appropriate paracrine signals from carcinoma cells may
conditions. Cell types may be identified histologically based on ex- result in dedifferentiation of prostate smooth muscle cells.5
pression pattern of vimentin, ␣-actin and late stage markers of
smooth muscle differentiation, such as calponin and smooth muscle Although it may potentially lead to the myofibroblast pheno-
myosin heavy chain (SM-MHC). Identifying specific cell types may type, direct evidence supporting carcinoma induced dedifferen-
be helpful for identifying reactive stroma in prostate cancer. tiation of smooth muscle is lacking. Therefore, it is predicted
REACTIVE STROMA IN PROSTATE CANCER PROGRESSION 2475

FIG. 3. Human prostate cancer specimens. A, normal human prostate epithelial acini surrounded by collagenous stroma (blue areas)
containing smooth muscle cells (red areas) and fibroblasts. B, Gleason 3 human prostate cancer in reactive stroma with some remaining
smooth muscle cells. C, Gleason 3 human prostate cancer in reactive stroma lacking smooth muscle cells. D, Gleason 4 human prostate
cancer progressing to stromal independent state with little stroma visible except around blood vessels. Reactive stroma composition in
prostate cancer may potentially be used to predict tumor progression rate. Masson’s trichrome stain, reduced from ⫻400.

that myofibroblasts in human prostate cancer reactive stroma collagen component of the extracellular matrix framework oc-
are derived from fibroblasts adjacent to carcinoma cells. cur during cancer progression.
In wound repair activated fibroblasts/myofibroblasts syn- Another important component of the extracellular matrix
thesize the key extracellular matrix components of granula- is fibronectin, which is a high molecular weight glycoprotein
tion tissue, namely fibronectin, collagen types I and III, and that mediates cell adhesion and migration.1 Alternative
many other glycoproteins and proteoglycans. They also re- splicing of pre-mRNA produces multiple isoforms of this pro-
lease growth factors that stimulate angiogenesis and pro- tein that differentially include cell type specific adhesion
teases that function to remodel the granulation tissue.9 domains.35 Fibronectins are widely expressed, especially at
Therefore, the presence of myofibroblasts in reactive stroma sites of active morphogenesis and inflammation, but the ex-
indicates that there may be increased production of extracel- pression of specific isoforms is regulated temporally and spa-
lular matrix components and overall extracellular matrix tially.1 Interestingly fibronectin molecules containing the
remodeling during prostate cancer progression. ED-A and ED-B domains are primarily found during devel-
Extracellular matrix synthesis and remodeling. Examination opment, although ED-A⫹ and ED-B⫹ fibronectin expression
of collagen fibers in human prostate stroma by scanning elec- has also been reported during wound healing and in the
tron microscopy indicated a disruption of the normal connective reactive stroma of many types of cancer.35, 36 Sites of wound
tissue framework in diseased states.31 In normal prostate it has repair have been shown to have increased local expression of
been reported that the “stromal network of collagen fibers is total fibronectin, including isoforms containing the ED-A and
loosely woven, fine and smooth in texture,” while in Gleason ED-B domains, which were expressed by macrophages and
score 7 adenocarcinoma the collagen fibers “appeared swollen in activated granulation tissue fibroblasts.37 Moreover, stromal
diameter” and there was “no regularity in the spatial relation- cells in human breast32 and colorectal38 tumors also showed
ship of the fibers.”31 Some areas of collagen destruction were increased expression of total fibronectin, and ED-A⫹ and
also observed. These data support the concept that remodeling ED-B⫹ isoforms. In addition, double staining experiments
of the extracellular matrix is a key feature of reactive stroma in using ED-B⫹ fibronectin in situ hybridization probes and
prostate cancer. Changes in collagen synthesis are not well smooth muscle ␣-actin antibodies identified myofibroblasts
characterized in prostate carcinoma but Masson’s trichrome as the source of ED-B⫹ fibronectin in oral squamous cell
staining suggests that there may be an increase in collagen carcinoma.39 In human prostate cancer the level of fibronec-
fibers in reactive stroma (fig. 3). Using an antibody to procolla- tin protein was elevated compared with that in normal tissue
gen I we observed collagen synthesis by reactive stromal cells in and benign prostatic hyperplasia,40 and reverse
human prostate cancer,28 which is consistent with breast can- transcriptase-polymerase chain reaction using mRNA iso-
cer. In situ hybridization studies showed that messenger lated from human prostate cancer tissue revealed increased
(m)RNA for types I and III procollagen are elevated in the expression of ED-B⫹ fibronectin.41 Also, these studies indi-
stromal cells of invasive breast carcinoma,32, 33 and an increase cated that fibronectin may regulate prostate cancer cell ad-
in collagen types I and III protein has been observed by immu- hesion and migration. Incubation with a polyclonal fibronec-
nohistochemistry.34 These data confirm that alterations in the tin antibody acted to disrupt adhesion of LNCaP human
2476 REACTIVE STROMA IN PROSTATE CANCER PROGRESSION

prostate cancer cells.41 Another study showed that the pres- pression of growth factors by reactive stromal cells recipro-
ence of plasma fibronectin induced invasion of DU145 human cally regulate carcinoma cells directly or indirectly. Foremost
prostate cancer cells through serum-free basement mem- in this list are members of the fibroblast growth factor fam-
branes in an in vitro invasion assay.42 It has been suggested ily. In human prostate cancer specimens fibroblast growth
that since ED-A⫹ and ED-B⫹ fibronectins are associated factor-2 expression has been shown to be significantly ele-
with cell migration during development, they may have a vated in stromal fibroblasts and in endothelial cells com-
similar function in wound healing.37 If it is correct, expres- pared with normal tissue.58 Fibroblast growth factor-2 stim-
sion of these embryonic fibronectins in reactive stroma may ulates fibroblast proliferation, extracellular matrix
create an extracellular matrix microenvironment that sup- production and protease secretion.11 It also functions as an
ports cancer cell migration and invasion. angiogenic factor that induces endothelial cell migration,
Myofibroblasts in the stroma of breast and colon carcinoma proliferation and differentiation into new blood vessels.59
were also shown to increase the expression of other extracel- Thus, fibroblast growth factor-2 may promote cancer progres-
lular matrix glycoproteins, such as tenascin-C and sion by inducing angiogenesis and reactive stroma formation.
thrombospondin-1.20, 22 These proteins are expressed at sites This growth factor may also directly regulate tumor cell
of tissue remodeling, where they act to influence cell adhe- proliferation and invasion. During cancer progression pros-
sion and migration.35 Thrombospondin-1 has been shown to tate carcinoma cells have been shown to up-regulate fibro-
stimulate tumor cell motility and may mediate tumor cell blast growth factor receptor isoforms with a high affinity for
adhesion to endothelial cells.43 The role of thrombospondin-1 fibroblast growth factor-2.58, 60 Accordingly, elevated sensi-
in prostate cancer is not well documented but expression in tivity to fibroblast growth factor-2 may stimulate cancer cell
reactive stroma is likely. proliferation and protease expression, thereby, supporting
Tenascin-C is closely associated with sites of stromal- tumor growth and invasion. Intense efforts are also under
epithelial interactions during development and tissue remod- way to examine the role of other fibroblast growth factor
eling.35 In the prostate tenascin was highly expressed around family members in prostate cancer.
developing glands but only weak expression was observed in Another stromal derived growth factor that is important in
normal adult tissue.44 Examination of high grade prostatic prostate cancer progression is hepatocyte growth factor. It is
intraepithelial neoplasia revealed periglandular expression produced by stromal cells and has been shown to stimulate
of tenascin-C and low/moderate grade human prostate cancer proliferation and migration of epithelial and endothelial cells
showed strong tenascin-C expression in the peritumoral stro- during development and tissue remodeling.61 Increased ex-
ma.45 Interestingly high molecular weight splice variants of pression of the hepatocyte growth factor receptor c-MET
tenascin-C, which contain additional fibronectin type III-like proto-oncogene has been associated with progression of sev-
repeats, have been reported in breast,46 colorectal47 and pros- eral types of carcinoma, including that of the prostate.62
tate44 cancer reactive stroma. It has been suggested that Moreover, hepatocyte growth factor acts to stimulate cell
expression of the high molecular weight isoform may favor growth, scattering and invasion of DU145 human prostate
cancer cell migration.47 cancer cells.62, 63 In addition, co-culture with DU145 or PC-3
Alterations in proteoglycan and glycosaminoglycan synthe- cells significantly increased hepatocyte growth factor expres-
sis have also been noted in wound repair and tumor reactive sion in human prostate stromal cells.63 These data provide
stroma.9, 48 An increase in proteoglycans with chondroitin direct evidence that human prostate cancer cells can induce
sulfate side chains has been reported in the stroma of colon47 surrounding stromal cells to secrete factors that promote
and breast cancer as well as in healing skin wounds.48 Ele- tumor growth and invasion. In addition, hepatocyte growth
vated levels of chondroitin sulfate in prostate cancer reactive factor may contribute to cancer progression by stimulating
stroma have been associated with prostate specific antigen angiogenesis.61
failure.49 In addition, expression of versican, a chondroitin Protease/protease inhibitor balance. Serine proteases and
sulfate proteoglycan, was shown to be higher in early stage matrix metalloproteinases (MMPs) have significant roles in
prostate cancer reactive stroma than in normal prostate tis- physiological and pathological tissue remodeling, including
sue.50 Increased versican expression was also reported in wound repair and tumorigenesis.64, 65 Furthermore, it is well
breast carcinoma reactive stroma51 and versican was shown established that elevated protease activity strongly corre-
to be expressed by smooth muscle cells at sites of vascular lates with metastatic potential in advanced cancer.66, 67 Al-
repair.52 though many cancer cell lines have been shown to express
Together these studies indicate that the extracellular ma- serine proteases and MMPs in vitro,68, 69 in vivo analysis of
trix surrounding prostate carcinoma is significantly modified various human tumors demonstrated that these proteases
in comparison to the extracellular matrix microenvironment were predominantly expressed by stromal cells.66 For exam-
of normal prostate tissue, and most extracellular matrix syn- ple, myofibroblasts in human breast cancer reactive stroma
thesis results from phenotypically switched reactive stromal expressed the serine protease urokinase plasminogen activa-
cells. It has been suggested that the alterations in extracel- tor,70 and MMP-2 and MMP-9 expression was localized to
lular matrix composition are induced by carcinoma cells.32, 53 stromal cells in human prostate cancer.71 Thus, reactive stro-
Co-culture of human fibroblasts with MCF-7 human breast mal cells may contribute to cancer progression by production
cancer cells provides data to support this theory. Production of proteases.
of collagen types I and III, and fibronectin was increased in Notably overall proteolytic activity is regulated by the bal-
human fibroblasts when co-cultured with MCF-7 cells.54 In ance of activated enzymes and their specific inhibitors.65
addition, MCF-7 cell conditioned media induced elevated ex- Examination of MMP and tissue inhibitor of metalloprotein-
pression of tenascin55 and glycosaminoglycans56 in fibro- ase (TIMP) expression in human prostate cancer by in situ
blasts. Similarly myofibroblasts isolated from granulation hybridization revealed that low grade (Gleason score 5) spec-
tissue exhibited increased collagen types I and III, and ver- imens showed elevated TIMP-1 and TIMP-2 expression in
sican mRNA expression.57 Therefore, it appears that cancer the stroma compared with MMP expression. However, high
cells are capable of inducing stromal cells to create an extra- grade (Gleason scores 8 to 10) specimens showed increased
cellular matrix similar to that of granulation tissue. Consid- levels of MMP-2 and MMP-9, while TIMPs were not ex-
ering how the extracellular matrix influences cell behavior it pressed.71 These observations indicate that the balance be-
follows that the reactive stromal microenvironment would be tween proteases and their inhibitors shifts during prostate
conducive to cancer cell survival, proliferation and migration. cancer progression. Similarly a study that measured total
Production of growth factors by reactive stroma. As carci- MMP enzyme activity and TIMP-1 expression in tissue ex-
noma derived growth factors regulate stroma, increased ex- tracts showed that the MMP-to-TIMP-1 ratio is higher in
REACTIVE STROMA IN PROSTATE CANCER PROGRESSION 2477
prostate cancer than in normal prostate tissues.72 Together tate cancer progression others have examined the effects of
these data suggest that the altered protease/protease inhib- anti-vascular endothelial growth factor neutralizing antibod-
itor balance in prostate tumors may promote cancer progres- ies on tumor growth in mice. In all cases treatment with the
sion. antibody suppressed angiogenesis, tumor growth and metas-
Studies which shift the protease/protease inhibitor bal- tasis even in well established tumors.87– 89
ance, indicate that proteases regulate prostate cancer pro- Although prostate cancer cells produce significant levels of
gression. For example, PC-3 human prostate carcinoma cells VEGF and other angiogenic factors, it is likely that surround-
transfected to over express the plasminogen activator ing stromal cells are also involved in angiogenesis. A recent
inhibitor-1 formed significantly smaller tumors in nude mice study used species specific anti-vascular endothelial growth
than in controls, and plasminogen activator inhibitor-1 ex- factor antibodies to treat a human rhabdomyosarcoma xeno-
pressing tumors showed reduced angiogenesis and metasta- graft in nude mice. Inhibition of VEGF produced by the host
sis.73 Similar results were obtained using the synthetic MMP stroma further reduced tumor growth compared with only
inhibitor A-177430. Mat Ly Lu rat prostate cancer cells blocking VEGF produced by the cancer cells.90 Stromal cells
formed large primary tumors and distant metastases when also produce several other growth factors that promote an-
injected into rats. However, treating the animals with giogenesis, such as PDGF, TGF-␤ and connective tissue
A-177430 resulted in decreased tumor growth, reduced an- growth factor.3 A 3-dimensional co-culture model using fibrin
giogenesis and prevention of metastasis.74 and collagen matrix showed that PC-3 human prostate can-
Since it has been shown that carcinoma cells affect the cer cells did not induce endothelial cell formation of capillary-
expression of extracellular matrix components and growth like structures unless fibroblasts were present.91 It is becom-
factors in adjacent stromal cells, it would be predicted that ing more clear that angiogenesis requires the cooperation of
cancer cells also influence stromal cell expression of pro- cancer cells and multiple cell types in reactive stroma.
teases. Co-culture of MCF-7 human breast cancer cells with In addition to providing the necessary blood supply, angio-
human fibroblasts induced expression of MMP-1, MMP-2, genesis may contribute to cancer progression in other ways.
MMP-3 and TIMP-1 in the fibroblasts.75 In contrast, co- For example, vascular endothelial cells secrete growth fac-
culture of normal mammary epithelial cells with fibroblasts tors and proteases that likely contribute to prostate cancer
only induced TIMP-1 expression. This study provides an- growth and invasion.66, 92 It has also been proposed that
other example of how stromal cell activity may be modulated tumor associated vasculature stimulates the generation of a
by tumor cells to promote cancer progression. granulation tissue-like environment. Blood vessels in tumors
Interestingly it is becoming clear that proteolytic degradation have enhanced permeability to circulating macromolecules,
of the extracellular matrix is more than a physical mechanism which is likely due to high levels of VEGF.93 Increased vas-
for tumor cell invasion through adjacent tissue.76 Extracellular cular permeability leads to the extravasation of plasma pro-
matrix remodeling by proteases alters extracellular matrix teins, including fibrinogen, fibronectin and clotting factors.
composition by releasing growth factors, generating bioactive Procoagulants produced by cancer cells, such as tissue factor,
extracellular matrix fragments and exposing cryptic or conceal- rapidly clot fibrinogen to form a fibrin matrix.10, 93 As in
ing existing cell adhesion sites.7, 77 These changes affect cell wound repair, the fibrin-fibronectin matrix recruits macro-
growth, survival and migration.76 Thus, the expression and phages, stimulates fibroblast proliferation and induces an-
activation of proteases further contribute to creating the reac- giogenesis. In addition, it has been shown that the provi-
tive stroma microenvironment that promotes tumor progres- sional matrix is remodeled into mature tumor stroma that
sion. resembles granulation tissue.10, 93 Fibrin deposits have been
Angiogenesis. Substantial evidence shows that angiogene- observed in many types of human cancer, including breast
sis is required for tumor growth and metastasis.78 Recruit- carcinoma.10 To our knowledge fibrin deposition has not been
ment of new blood vessel growth clearly illustrates the im- examined in prostate cancer, although elevated levels of tis-
portance of carcinoma-stroma interactions during cancer sue factor have been reported.94 Therefore, this process may
progression. Endothelial cells from microvessels in the sur- significantly contribute to the formation of reactive stroma in
rounding stroma must be induced to migrate into the tumor, prostate cancer.
where they proliferate and form new blood vessels. This Tumor-associated macrophages. Considering the similari-
complex process is regulated by the relative balance of an- ties of the cancer reactive stroma microenvironment and
giogenesis inducers and angiogenesis inhibitors in the extra- granulation tissue, the recruitment of macrophages to the
cellular milieu. These factors may be secreted by cancer or tumor would be expected. In fact, it has been reported that
stromal cells, or mobilized from the extracellular matrix. the macrophage density is significantly higher in malignant
Increased concentration of an activator or decreased levels of prostate tissue than in adjacent benign areas.95 The che-
an inhibitor alter the balance and lead to the growth of new moattraction of macrophages is likely due to fibrin deposits
blood vessels.79 Increased expression of the angiogenesis ac- and fragments of collagen and elastin in the extracellular
tivator, VEGF is observed in many types of cancer, including matrix.96 In addition, expression of macrophage recruitment
that of the prostate. factors, such as monocyte chemotactic protein-1, VEGF and
In normal human prostate tissue it has been reported that TGF-␤, have been observed in cancer and stromal cells.97, 98
VEGF is expressed at low levels and expression is restricted Tumor associated macrophages have multiple functions
to stromal cells.80, 81 However in high grade prostatic intra- that may inhibit or promote tumor growth. For example, they
epithelial neoplasia and prostate cancer elevated expression can release cytotoxic molecules and present tumor associated
was observed in cancer, stroma and vascular endothelial antigens to other components of the immune system to elim-
cells.80 – 83 Also, increased expression by human prostate can- inate tumor cells.97 However, macrophage functions associ-
cer cell lines correlated with tumor microvessel density and ated with wound repair may aid cancer progression. Tumor
metastatic potential when grown in nude mice.84 Additional associated macrophages have been shown to secrete numer-
studies have shown that VEGF expression in the prostate is ous growth factors that induce angiogenesis and may pro-
androgen regulated. Dihydrotestosterone stimulated expres- mote tumor growth.99 Also, it has been reported that tumor
sion in human fetal prostate fibroblasts,83 and testosterone associated macrophages release urokinase plasminogen acti-
replacement in castrated rats induced VEGF synthesis in the vator and MMPs, and show procoagulant activity to initiate
prostate epithelium.85 In addition, castrating mice bearing fibrin deposition. The balance of these activities determines
androgen responsive human prostate cancer xenografts re- how they influence tumor growth.98 As may be predicted,
sulted in a significant decrease in tumor VEGF expression evidence suggests that cancer cells secrete factors that redi-
and tumor volume.86 To determine the role of VEGF in pros- rect tumor associated macrophage activities to promote tu-
2478 REACTIVE STROMA IN PROSTATE CANCER PROGRESSION

mor progression.97 Thus, tumor associated macrophages are dent since co-injection of Matrigel extracellular matrix or
another key component of reactive stroma in cancer. stromal cells is required to form subcutaneous tumors in
Reactive stroma promotes cancer progression in vivo. It has nude mice. However, not all stromal cell lines support LN-
been proposed that a reactive stroma environment similar to CaP tumor growth. LNCaP cells formed tumors when co-
granulation tissue may favor tumorigenesis. To investigate injected with fibroblasts derived from a human osteogenic
this hypothesis experimentally, fragments of rat colon carci- sarcoma or with fibroblasts derived from rat fetal urogenital
noma tissue were implanted into undisturbed rat subcutane- sinus mesenchyme.103 Conversely co-injection of normal rat
ous tissue or into experimentally, induced granulation tis- prostate stromal cells with LNCaP cells rarely supported
sue.100 Carcinoma cells transplanted into undisturbed tumor growth, and normal adult human lung fibroblasts did
subcutaneous tissue “were arranged in compact groups with not support tumor growth at all.103 It may be that stromal
a strong tendency to form acini.”100 In contrast, carcinoma cells that supported tumor growth had an activated pheno-
cells transplanted into granulation tissue were widely dis- type independent of interaction with LNCaP cells. However,
persed and showed an invasive morphotype, including an it is also possible that stromal cells that failed to support
elongated shape with extensive tumor cell protrusions at the LNCaP tumor growth were unable to create the reactive
ultrastructural level. Tumor growth was not measured, al- stromal environment. Accordingly we predict that successful
though these results suggest that granulation tissue en- tumorigenesis requires the ability of carcinoma to induce
hances cancer cell invasion. phenotypic switching in adjacent stromal cells and the capa-
The synthesis of extracellular matrix components, growth bility of activated stromal cells to create the reactive stromal
factors and proteases by tumor associated stromal cells im- environment.
plies that these cells are important for creating the reactive Together these studies suggest that human prostate cancer
stroma environment. However, it does not directly demon- is accompanied by the induction of reactive stroma, charac-
strate their role in promoting tumor growth. Recent studies terized by stromal cell phenotypic switching, extracellular
have examined this issue by combining stromal cells with trans- matrix remodeling, altered growth factor and protease bio-
formed epithelium in models of tumorigenesis. Elenbaas et al availability, increased angiogenesis and an influx of tumor
reported that co-injection of primary human fibroblasts with associated macrophages. As in the stromal response de-
primary transformed human mammary epithelial cells en- scribed in breast and colon cancer, the reactive stroma envi-
hances tumor formation and accelerates tumor growth in nude ronment mimics that of granulation tissue in wound repair.
mice.101 Although primary human mammary epithelial cells These conditions likely support cancer cell proliferation, can-
transformed with the 3 oncogenes formed tumors when injected cer cell migration/invasion and stimulate angiogenesis to
subcutaneously, the incidence was only 52%. When these cells promote tumorigenesis. Furthermore, these studies indicate
were mixed with normal human mammary fibroblasts, tumors that cancer cells secrete factors that induce and regulate
formed at 100% of the injection sites, and average time to the reactive stroma.
development of palpable tumors was reduced from 52 to 20
days. These data suggest that while oncogene expression is
POTENTIAL REGULATORS OF REACTIVE STROMA
critical for tumor formation, the stromal microenvironment can
further promote tumorigenesis. Growth factors that regulate wound repair, namely TGF-␤,
Another study used carcinoma associated fibroblasts iso- fibroblast growth factor-2, PDGF and VEGF,9 are also likely
lated from human prostate cancer tissue and human prostate to modulate the reactive stroma environment in cancer. Ex-
epithelial cells immortalized with SV40 large T antigen.102 In pression of these growth factors is typically restricted to
vivo recombination of SV40 large T antigen with human stromal components but many cancer cells acquire the ability
prostate carcinoma associated fibroblasts in athymic rodent to express them with time. It follows that over expression of
hosts resulted in the rapid growth of tumors that resembled TGF-␤, PDGF, fibroblast growth factor-2 and VEGF by pros-
poorly differentiated adenocarcinoma. Interestingly normal tate carcinoma cells may function to induce reactive stroma
human prostate fibroblasts did not support SV40 large T and angiogenesis.
antigen tumor growth. This study indicates that normal Transforming growth factor-␤. Transforming growth
prostate stromal cells and stromal cells adjacent to prostate factor-␤ is a multifunctional growth factor that regulates
carcinoma show fundamental differences in the cell pheno- several biological processes, including development, wound
type. Notably carcinoma associated fibroblasts showed no healing and tumorigenesis.104, 105 Its release from degranu-
effect on normal epithelium, suggesting that epithelial cells lating platelets initiates the stromal response during wound
must sustain genetic alterations to initiate tumorigenicity. repair. In fact, it has been shown that TGF-␤1 is sufficient to
It seems likely that the ability of stromal cells to support induce the generation of granulation tissue and angiogenesis
tumor growth in vivo is related to their initial phenotype and in vivo.106, 107 Moreover, TGF-␤1 is the only growth factor
to the cancer cell ability to convert them to the activated known to induce differentiation of fibroblasts to the myofi-
myofibroblast phenotype. For example, the study of Olumi et broblast phenotype.107, 108 Transforming growth factor-␤ has
al implies that carcinoma associated fibroblasts show a reac- also been shown to attract immune cells by chemotaxis and
tive phenotype and, therefore, support the growth of SV40 stimulate extracellular matrix synthesis and remodeling.105
large T antigen tumors.102 However, SV40 large T antigen In many types of cancers TGF-␤ is over expressed by carci-
cells do not appear to induce normal human prostate fibro- noma cells.22, 23, 109 Although it inhibits epithelial cell growth in
blasts to switch to the reactive phenotype since tumors did vitro, studies show that TGF-␤ over expression by prostate
not form when these cells were recombined in vivo. In con- cancer cells promotes tumor growth in vivo.110 It has been
trast, transformed primary human mammary epithelial cells shown that prostate cancer cells avoid TGF-␤ growth inhibition
in the study of Elenbaas et al appear to have induced normal by down-regulating TGF-␤ receptor expression.111–114 However,
stromal cells to support tumor growth.101 Histological anal- the stroma maintains expression of these receptors and, there-
ysis of tumors generated by injecting transformed epithelium fore, it is still responsive to this growth factor.111 These obser-
revealed that mouse host stromal cells were recruited into vations suggest that TGF-␤ acts primarily on the stromal com-
the tumors. Furthermore, co-injection of transformed epithe- partment during prostate cancer progression.
lium with normal human mammary fibroblasts significantly Considering the role of TGF-␤ in wound repair and the sim-
promoted tumorigenesis. ilarities of granulation tissue and reactive stroma, it has been
This theory may also explain the differential support of proposed that TGF-␤ expression by carcinoma cells induces
LNCaP tumor growth that has been observed in different reactive stroma in cancer.22–24, 47 Transforming growth
fibroblast lines. LNCaP cells are considered stromal depen- factor-␤1 has been shown to modulate the myodifferentiation of
REACTIVE STROMA IN PROSTATE CANCER PROGRESSION 2479
prostate fibroblasts29, 30 and mammary gland fibroblasts108 to a in prostate cancer, but PDGF-A and PDGF receptor ␣ were
myofibroblast phenotype. It has also been shown to induce the expressed by epithelial and stromal cells in prostatic intra-
synthesis of many extracellular matrix components in reactive epithelial neoplasia.127 In addition, the human prostate can-
stroma, such as collagen types I and III, fibronectin, ED-A⫹ and cer cell lines DU145 and PC-3 express PDGF,128 and PDGF
ED-B⫹ fibronectin, tenascin, thrombospondin and versi- stimulated human prostate stromal cell proliferation in
can.35, 55, 57, 106, 115 In addition, TGF-␤ up-regulated stromal cell vitro.129 Thus, PDGF may be predicted to promote reactive
expression of other growth factors involved in tissue repair and stroma in the prostate.
angiogenesis, including fibroblast growth factor-2, VEGF and As discussed, the recent study of Giri et al indicated that
connective tissue growth factor.116 –119 Moreover, TGF-␤ may the elevated level of fibroblast growth factor-2 in human
function to suppress the immune system, possibly by inhibiting prostate cancer was “due to induction of stromal FGF-2 pro-
the production of cytotoxic molecules by tumor associated mac- duction.”58 However, it was also suggested that carcinoma
rophages.97, 109 cells in more advanced prostate cancers over express fibro-
Studies of in vivo tumor models further suggest that blast growth factor-2. For example, the human prostate can-
TGF-␤ over expression may support cancer progression by cer cell lines DU145 and PC-3 have been shown to express
acting through the stroma. For example, tumors formed by this growth factor, and it has been reported that prostate
human prostate cancer cells transfected to over express cancer cells that metastasize to bone also express it.59 Fur-
TGF-␤1 showed elevated angiogenesis and an increased rate thermore, studies of the Dunning rat model showed that
of metastasis.120 In another study CHO cells transfected to activation of fibroblast growth factor-2 expression accompa-
over express this growth factor formed significantly larger nied “progression of the epithelial cell to malignancy.”60 The
tumors at a more rapid rate in nude mice compared with ability of carcinoma cells to express fibroblast growth factor-2
control cells.121 Over production did not affect the prolifera- would be expected to stimulate angiogenesis and reactive
tion rate of CHO cells in vitro, although it significantly in- stroma, and may further promote cancer cell proliferation
creased angiogenesis in vivo. The specificity of TGF-␤1 was and invasion via an autocrine loop.
confirmed by co-injection of a neutralizing antibody that re- As described, over expression of VEGF by carcinoma cells
duced tumor growth and angiogenesis to control levels. Since has been reported in many tumor types, including prostate82
angiogenesis is a key component of reactive stroma, these and breast.32 It has been suggested that VEGF promotes
studies imply that TGF-␤1 promotes tumor progression by cancer progression in several ways.93 It has been shown to
inducing reactive stroma. increase microvascular permeability, which allows circulat-
Recent evidence suggests TGF-␤1 induction of reactive ing plasma proteins to leak into the surrounding extracellu-
stroma may be mediated by the expression of connective lar matrix. This leakage results in generation of a fibrin
tissue growth factor. Connective tissue growth factor expres- matrix that attracts fibroblasts, macrophages and endothe-
sion was shown to be induced in fibroblasts after activation lial cells to initiate the wound repair response and stimulate
with TGF-␤1117, and stimulated fibroblast proliferation, ex- angiogenesis. Also, VEGF stimulated endothelial cell prolif-
tracellular matrix synthesis and angiogenesis.122 In addition, eration and migration to drive angiogenesis. These activities
connective tissue growth factor antibodies or antisense oligo- suggest that VEGF has a significant role in the regulation of
nucleotides blocked TGF-␤ induced collagen type I synthesis reactive stroma in cancer.
in fibroblasts. Moreover, connective tissue growth factor and
TGF-␤ were found to be co-expressed at sites of wound repair,
PERSPECTIVES
fibrosis and in the reactive stroma of cancer.122, 123 In situ
hybridization for connective tissue growth factor and TGF-␤1 Reactive stroma hypothesis. Based on an understanding of
in mammary carcinomas showed that the former was ex- the stromal wound repair response we can begin to outline a
pressed in stromal cells, while the latter was expressed in working hypothesis describing the generation of reactive
cancer cells.123 These data further indicate that TGF-␤1 pro- stroma in response to prostate carcinoma. Initially carci-
duced by carcinoma cells acts on the surrounding stromal nogenesis at the prostatic intraepithelial neoplasia stage
cells to induce reactive stroma. results in altered epithelial proliferation, over expression
Together these studies provide evidence that TGF-␤ affects of growth factors and disruption of the basement mem-
several aspects of reactive stroma that are likely to promote brane extracellular matrix. These processes produce a dis-
cancer progression. As in its role in wound repair, TGF-␤ ruption in normal prostate tissue homeostasis, which is
regulates stromal cell differentiation, induces stromal cell considered a wound situation by the host. The stroma
expression of extracellular matrix components and growth reacts by activating fibroblasts to the myofibroblast phe-
factors, and promotes angiogenesis to create a granulation notype to repair the tissue disorder. In effect, an endoge-
tissue-like environment. Accordingly it is predicted to be a nous wound repair response has been initiated. The result
primary regulator of reactive stroma in cancer. However, it is in prostate cancer is the generation of a reactive stroma
known that the effects of TGF-␤ can be modulated by the that accompanies proliferative carcinoma cells. Accord-
context of other growth factors, such as PDGF, epidermal ingly this reactive stroma shows stromal cell phenotypic
growth factor and fibroblast growth factor-2.108, 124 Therefore, switching, extracellular matrix remodeling, increased bio-
the specific mechanisms by which TGF-␤ stimulates reactive availability of growth factors and induced angiogenesis to
stroma may be complex and likely result through the inte- drive the repair process. As in any wound repair situation,
grated actions of TGF-␤ and many other regulatory factors. the microenvironment would be expected to be growth
Other growth factors. PDGF has also been suggested as a promoting. Therefore, these alterations in the microenvi-
mediator of reactive stroma in cancer.22, 24 It is another com- ronment during early cancer progression would be pre-
ponent of the wound repair pathway that stimulates fibro- dicted to promote survival and proliferation of the carci-
blast and smooth muscle cell proliferation and migration. It noma cell, resulting in enhanced tumorigenesis.
also induces the production of extracellular matrix compo- Several potential factors may induce reactive stroma in
nents, growth factors and proteases.125 Human melanoma prostate cancer, although TGF-␤1 emerges as the most likely
cells transfected to over express platelet derived growth candidate. It is clear that it has a fundamental role in the
factor-BB produced tumors in nude mice that contained an wound repair response and it is over expressed by many
abundance of connective tissue and blood vessels.126 In con- carcinoma cells, including those of the prostate. Further-
trast, melanoma cells lacking PDGF expression formed tu- more, its action appears to be directed toward the stroma
mors that were poorly vascularized and necrotic with no since transforming growth factor-␤ receptor expression is
connective tissue. The role of PDGF is not well characterized present in stromal cells but down-regulated in prostate car-
2480 REACTIVE STROMA IN PROSTATE CANCER PROGRESSION

cinoma cells.111 The expected outcome of TGF-␤1 action in may be a potential site for prostate cancer therapy. For
these conditions would be the induction of reactive stroma, example, agents that block angiogenesis are receiving con-
while prostate cancer cells escape TGF-␤ growth inhibitory siderable attention as potential therapeutics against solid
effects. It is likely that fibroblasts immediately adjacent to tumors. Other therapies may target the reactive stromal
epithelial acini are activated to switch to the reactive myofi- myofibroblasts to alter the microenvironment. By inhibiting
broblast phenotype in response to carcinoma produced production of stromal derived factors that promote cancer
TGF-␤1 This hypothesis is consistent with observations in progression or by stimulating the synthesis of stromal de-
breast cancer reactive stroma27, 108 and indications that rived inhibitors, the microenvironment may be modulated to
transforming growth factor-␤1 regulates myodifferentiation inhibit tumor growth. A potential advantage of this indirect
of prostate fibroblasts.29, 30 Carcinoma derived TGF-␤1 would approach is that therapy would be directed toward a normal
also be expected to regulate the reactive stromal cell secre- host reactive stroma response rather than a carcinoma cell
tion of extracellular matrix components, growth factors and with genomic instability and less biological predictability.
proteases to create a tumor promoting environment that
resembles granulation tissue. Therefore, it is proposed that CONCLUSIONS
TGF-␤1 over expression by prostate cancer cells activates the
host wound repair pathway to generate reactive stroma, It is becoming more clear that reactive stroma is a general
which functions to promote tumorigenesis. This hypothesis feature of most types of carcinoma. Considerable evidence
would predict that the carcinoma cell does not simply invade suggests that stromal cells are activated by the carcinoma
the normal stroma. Rather it is likely that the developing cells and subsequently function to promote tumorigenesis.
tumor actively recruits fibroblasts and myofibroblasts to gen- Any disruption of tissue homeostasis, such as physical
erate its own stroma, which is a reactive stroma by nature of wounding, microorganism invasion or carcinogenesis of
the generic host response to wounding. epithelial cells, is predicted to induce a generic repair
Progression to stromal independence. Unlike early stages of response that leads to stromal cell activation. Thus, it is
prostate cancer there is an overall reduction in the amount of not surprising that the features of reactive stroma in can-
stroma associated with advanced prostate tumors. Figure 3 cer closely parallel the features of stromal granulation
shows that only thin strands of stroma are evident in poorly tissue in wound healing responses. Changes in reactive
(Gleason 4) compared with moderately differentiated (Glea- stroma include stromal cell phenotypic switching to acti-
son 3) carcinoma and normal tissue. These data suggest that vated myofibroblasts, altered extracellular matrix compo-
carcinoma cells have progressed to a stromal independent sition, increased growth factor bioavailability, elevated
stage. However, it may be that the tumor still requires com- protease activity, influx of tumor associated macrophages
ponents normally secreted by stroma for progression. In this and induction of angiogenesis. Together these alterations
scenario as the tumor overgrows the surrounding stroma, create a reactive stroma microenvironment that is permis-
data indicate that carcinoma cells begin to express factors sive or promotive to carcinoma cell growth or survival and,
typically produced by stromal cells. For example, prostate hence, supports tumor progression.
cancer cells over express VEGF and possibly fibroblast The concept of reactive stroma in prostate cancer is just
growth factor-2, presumably to induce angiogenesis. These now emerging and preliminary data suggest that this stroma is
factors are usually expressed in the stroma. Carcinoma cells similar to the reactive stroma characterized in breast and colon
also express proteases, such as urokinase plasminogen acti- cancer. Our current data suggest that the myofibroblast is the
vator and MMPs, which would enhance invasion and may key cell type in human prostate cancer reactive stroma, al-
activate growth factors in the extracellular matrix. Cancer though the specific mechanisms that generate reactive stroma
cells may even secrete extracellular matrix components nor- in prostate cancer are less clear. Due to its role in stromal
mally produced by stroma. Our reactive stroma hypothesis responses in wound healing and the fact that it is over ex-
suggests that as carcinoma acquires the ability to express pressed in prostate cancer TGF-␤ may be the principal mediator
these factors, reactive stromal cells are no longer required to of reactive stroma in prostate cancer. Over expression of other
support tumor growth. Accordingly, at this stage the tumor is growth factors that function in stromal wound repair, such as
considered stromal cell independent. Thus, reactive stroma fibroblast growth factor-2, VEGF, and PDGF, suggests that
may have key significance in affecting progression in early they are also potential regulators of the reactive stroma envi-
compared with more advanced prostate cancer. ronment in prostate cancer. Although reactive stroma is pre-
Diagnostics and therapeutics. Several studies have shown dicted to have a key role in early tumorigenesis, advanced
a correlation of cancer progression with markers of reactive stages of prostate cancer are likely to evolve to stromal inde-
stroma. For example, expression of extracellular matrix com- pendence. It is possible that the expression of certain markers of
ponents, such as tenascin-C45 and versican,50 have been as- reactive stroma may have clinical diagnostic value for providing
sociated with prostate cancer progression. It has also been information in addition to Gleason score for assessing the pro-
reported that angiogenesis correlates with prostate cancer gression potential of prostate cancer. Moreover, reactive stroma
metastasis.130 In addition, pronounced fibrosis in colon can- may emerge as an additional compartment of cells to target for
cer is associated with a poor prognosis.20, 131 We suggest that novel therapeutic approaches. Since the evidence suggests that
analysis of reactive stroma induction may be a valuable tool reactive stroma may have a role in early progression, a thera-
in prostate cancer diagnosis. Figure 3 shows that the reactive peutic approach toward altering the tumor promotive feature of
stroma environment may differ among specimens assigned reactive stroma may be a method of treating early prostate
the same Gleason score. Reactive stroma at some Gleason cancer. Currently little is understood about the biology of reac-
grade 3 cancer sites contains a significant smooth muscle tive stroma in prostate cancer. However, the many similarities
component, while reactive stroma at other Gleason grade 3 of reactive stroma in different types of carcinoma and reactive
sites has little smooth muscle (fig. 3, B and C). Thus, the stroma at wound repair sites suggest that the reactive stromal
extent of the stromal response may identify aggressive tu- environment has a fundamental and important role in promot-
mors and may potentially be used to predict the progression ing prostate cancer progression.
rate in patients with the same Gleason score. This technique
would have clinical benefit since it is currently difficult to REFERENCES
predict the outcome of moderately differentiated prostate 1. Kreis, T. and Vale, R.: Guidebook to the Extracellular Matrix,
tumors. Anchor, and Adhesion Proteins. New York: Oxford University
If reactive stroma promotes cancer progression as our hy- Press, 1999
pothesis predicts, it follows that the stromal compartment 2. Taipale, J. and Keski-Oja, J.: Growth factors in the extracellu-
REACTIVE STROMA IN PROSTATE CANCER PROGRESSION 2481
lar matrix. FASEB J, 11: 51, 1997 29. Gerdes, M. J., Larsen, M., Dang, T. D. et al: Regulation of
3. Cotran, R. S., Kumar, V., Collins, T. et al: Robbins Pathologic prostate stromal cell myodifferentiation by androgen and
Basis of Disease, 6th ed. Philadelphia: W. B. Saunders, 1999 TGF-␤1. Unpublished data, 2001
4. Lin, C. Q. and Bissell, M. J.: Multi-faceted regulation of cell 30. Peehl, D. M. and Sellers, R. G.: Induction of smooth muscle cell
differentiation by extracellular matrix. FASEB J, 7: 737, phenotype in cultured human prostatic stromal cells. Exp
1993 Cell Res, 232: 208, 1997
5. Hayward, S. W., Cunha, G. R. and Dahiya, R.: Normal devel- 31. Morrison, C., Thornhill, J. and Gaffney, E.: The connective
opment and carcinogenesis of the prostate. A unifying hy- tissue framework in the normal prostate, BPH and prostate
pothesis. Ann N Y Acad Sci, 784: 50, 1996 cancer: analysis by scanning electron microscopy after cellu-
6. Fong, C. J., Sherwood, E. R., Sutkowski, D. M. et al: Reconsti- lar digestion. Urol Res, 28: 304, 2000
tuted basement membrane promotes morphological and func- 32. Brown, L. F., Guidi, A. J., Schnitt, S. J. et al: Vascular stroma
tional differentiation of primary human prostatic epithelial formation in carcinoma in situ, invasive carcinoma, and met-
cells. Prostate, 19: 221, 1991 astatic carcinoma of the breast. Clin Cancer Res, 5: 1041,
7. Streuli, C.: Extracellular matrix remodelling and cellular dif- 1999
ferentiation. Curr Opin Cell Biol, 11: 634, 1999 33. Kauppila, S., Stenback, F., Risteli, J. et al: Aberrant type I and
8. Gabbiani, G., Hirschel, B. J., Ryan, G. B. et al: Granulation type III collagen gene expression in human breast cancer in
tissue as a contractile organ. A study of structure and func- vivo. J Pathol, 186: 262, 1998
tion. J Exp Med, 135: 719, 1972 34. Lagace, R., Grimaud, J. A., Schurch, W. et al: Myofibroblastic
9. Clark, R. A. F.: Wound repair. In: The Molecular and Cellular stromal reaction in carcinoma of the breast: variations of
Biology of Wound Repair, 2nd ed. Edited by R. A. F. Clark. collagenous matrix and structural glycoproteins. Virchows
New York: Plenum Press, p. 3, 1996 Arch A Pathol Anat Histopathol, 408: 49, 1985
10. Dvorak, H. F.: Tumors: wounds that do not heal. Similarities 35. Yamada, K. M. and Clark, R. A. F.: Provisional matrix. In: The
between tumor stroma generation and wound healing. Molecular and Cellular Biology of Wound Repair, 2nd ed.
N Engl J Med, 315: 1650, 1986 Edited by R. A. F. Clark. New York: Plenum Press, p. 51,
11. Mutsaers, S. E., Bishop, J. E., McGrouther, G. et al: Mecha- 1996
nisms of tissue repair: from wound healing to fibrosis. Int 36. Kosmehl, H., Berndt, A. and Katenkamp, D.: Molecular vari-
J Biochem Cell Biol, 29: 5, 1997 ants of fibronectin and laminin: structure, physiological oc-
12. Faxon, D. P., Coats, W. and Currier, J.: Remodeling of the currence and histopathological aspects. Virchows Arch, 429:
coronary artery after vascular injury. Prog Cardiovasc Dis, 311, 1996
40: 129, 1997 37. Brown, L. F., Dubin, D., Lavigne, L. et al: Macrophages and
13. Owens, G. K.: Regulation of differentiation of vascular smooth fibroblasts express embryonic fibronectins during cutaneous
muscle cells. Physiol Rev, 75: 487, 1995 wound healing. Am J Pathol, 142: 793, 1993
14. Thyberg, J.: Differentiated properties and proliferation of arte- 38. Pujuguet, P., Hammann, A., Moutet, M. et al: Expression of
rial smooth muscle cells in culture. Int Rev Cytol, 169: 183, fibronectin ED-A⫹ and ED-B⫹ isoforms by human and ex-
1996 perimental colorectal cancer. Contribution of cancer cells and
15. Schmitt-Graff, A., Desmouliere, A. and Gabbiani, G.: Heteroge- tumor-associated myofibroblasts. Am J Pathol, 148: 579,
neity of myofibroblast phenotypic features: an example of 1996
fibroblastic cell plasticity. Virchows Arch, 425: 3, 1994 39. Kosmehl, H., Berndt, A., Strassburger, S. et al: Distribution of
16. Gabbiani, G.: The cellular derivation and the life span of the laminin and fibronectin isoforms in oral mucosa and oral
myofibroblast. Pathol Res Pract, 192: 708, 1996 squamous cell carcinoma. Br J Cancer, 81: 1071, 1999
17. Kocher, O., Gabbiani, F., Gabbiani, G. et al: Phenotypic fea- 40. Sonmez, H., Suer, S., Karaarslan, I. et al: Tissue fibronectin
tures of smooth muscle cells during the evolution of experi- levels of human prostatic cancer, as a tumor marker. Cancer
mental carotid artery intimal thickening. Biochemical and Biochem Biophys, 15: 107, 1995
morphologic studies. Lab Invest, 65: 459, 1991 41. Albrecht, M., Renneberg, H., Wennemuth, G. et al: Fibronectin
18. Li, S., Sims, S., Jiao, Y. et al: Evidence from a novel human cell in human prostatic cells in vivo and in vitro: expression,
clone that adult vascular smooth muscle cells can convert distribution, and pathological significance. Histochem Cell
reversibly between noncontractile and contractile pheno- Biol, 112: 51, 1999
types. Circ Res, 85: 338, 1999 42. Livant, D. L., Brabec, R. K., Pienta, K. J. et al: Anti-invasive,
19. Seemayer, T. A., Lagace, R., Schurch, W. et al: The myofibro- antitumorigenic, and antimetastatic activities of the PHSCN
blast: biologic, pathologic, and theoretical considerations. sequence in prostate carcinoma. Cancer Res, 60: 309, 2000
Pathol Annu, 15: 443, 1980 43. Roberts, D. D.: Regulation of tumor growth and metastasis by
20. Martin, M., Pujuguet, P. and Martin, F.: Role of stromal myo- thrombospondin-1. FASEB J, 10: 1183, 1996
fibroblasts infiltrating colon cancer in tumor invasion. Pathol 44. Ibrahim, S. N., Lightner, V. A., Ventimiglia, J. B. et al: Tena-
Res Pract, 192: 712, 1996 scin expression in prostatic hyperplasia, intraepithelial neo-
21. Noel, A. and Foidart, J. M.: The role of stroma in breast carci- plasia, and carcinoma. Hum Pathol, 24: 982, 1993
noma growth in vivo. J Mammary Gland Biol Neoplasia, 3: 45. Xue, Y., Smedts, F., Latijnhouwers, M. A. et al: Tenascin-C
215, 1998 expression in prostatic intraepithelial neoplasia (PIN): a
22. Ronnov-Jessen, L., Petersen, O. W. and Bissell, M. J.: Cellular marker of progression? Anticancer Res, 18: 2679, 1998
changes involved in conversion of normal to malignant 46. Borsi, L., Carnemolla, B., Nicolo, G. et al: Expression of differ-
breast: importance of the stromal reaction. Physiol Rev, 76: ent tenascin isoforms in normal, hyperplastic and neoplastic
69, 1996 human breast tissues. Int J Cancer, 52: 688, 1992
23. Gregoire, M. and Lieubeau, B.: The role of fibroblasts in tumor 47. Hauptmann, S., Zardi, L., Siri, A. et al: Extracellular matrix
behavior. Cancer Metastasis Rev, 14: 339, 1995 proteins in colorectal carcinomas. Expression of tenascin and
24. Rowley, D. R.: What might a stromal response mean to prostate fibronectin isoforms. Lab Invest, 73: 172, 1995
cancer progression? Cancer Metastasis Rev, 17: 411, 1998 48. Yeo, T. K., Brown, L. and Dvorak, H. F.: Alterations in proteo-
25. Sieweke, M. H. and Bissell, M. J.: The tumor-promoting effect glycan synthesis common to healing wounds and tumors.
of wounding: a possible role for TGF-beta-induced stromal Am J Pathol, 138: 1437, 1991
alterations. Crit Rev Oncog, 5: 297, 1994 49. Ricciardelli, C., Quinn, D. I., Raymond, W. A. et al: Elevated
26. Seemayer, T. A., Lagace, R., Schurch, W. et al: Myofibroblasts levels of peritumoral chondroitin sulfate are predictive of
in the stroma of invasive and metastatic carcinoma: a possi- poor prognosis in patients treated by radical prostatectomy
ble host response to neoplasia. Am J Surg Pathol, 3: 525, 1979 for early-stage prostate cancer. Cancer Res, 59: 2324, 1999
27. Ronnov-Jessen, L., Petersen, O. W., Koteliansky, V. E. et al: 50. Ricciardelli, C., Mayne, K., Sykes, P. J. et al: Elevated levels of
The origin of the myofibroblasts in breast cancer. Recapitu- versican but not decorin predict disease progression in early-
lation of tumor environment in culture unravels diversity and stage prostate cancer. Clin Cancer Res, 4: 963, 1998
implicates converted fibroblasts and recruited smooth muscle 51. Nara, Y., Kato, Y., Torii, Y. et al: Immunohistochemical local-
cells. J Clin Invest, 95: 859, 1995 ization of extracellular matrix components in human breast
28. Tuxhorn, J. A., Ayala, G. and Rowley, D. R.: Markers of reactive tumours with special reference to PG-M/versican. Histochem
stroma in human prostate cancer. Unpublished data, 2001 J, 29: 21, 1997
2482 REACTIVE STROMA IN PROSTATE CANCER PROGRESSION

52. Merrilees, M., Beaumont, B., Scott, L. et al: Effect of TGF- carcinoma cells inhibits primary tumor growth, tumor-
beta(1) antisense S-oligonucleotide on synthesis and accumu- associated angiogenesis, and metastasis to lung and liver in
lation of matrix proteoglycans in balloon catheter-injured an athymic mouse model. J Clin Invest, 96: 2593, 1995
neointima of rabbit carotid arteries. J Vasc Res, 37: 50, 2000 74. Rabbani, S. A., Harakidas, P., Guo, Y. et al: Synthetic inhibitor
53. Iozzo, R. V.: Tumor stroma as a regulator of neoplastic behav- of matrix metalloproteases decreases tumor growth and me-
ior. Agonistic and antagonistic elements embedded in the tastases in a syngeneic model of rat prostate cancer in vivo.
same connective tissue. Lab Invest, 73: 157, 1995 Int J Cancer, 87: 276, 2000
54. Noel, A., Munaut, C., Boulvain, A. et al: Modulation of collagen 75. Ito, A., Nakajima, S., Sasaguri, Y. et al: Co-culture of human
and fibronectin synthesis in fibroblasts by normal and malig- breast adenocarcinoma MCF-7 cells and human dermal fibro-
nant cells. J Cell Biochem, 48: 150, 1992 blasts enhances the production of matrix metalloproteinases
55. Chiquet-Ehrismann, R., Kalla, P. and Pearson, C. A.: Partici- 1, 2 and 3 in fibroblasts. Br J Cancer, 71: 1039, 1995
pation of tenascin and transforming growth factor-beta in 76. DeClerck, Y. A.: Interactions between tumour cells and stromal
reciprocal epithelial-mesenchymal interactions of MCF7 cells cells and proteolytic modification of the extracellular matrix
and fibroblasts. Cancer Res, 49: 4322, 1989 by metalloproteinases in cancer. Eur J Cancer, 36: 1258, 2000
56. Merrilees, M. J. and Finlay, G. J.: Human tumor cells in culture 77. Vu, T. H. and Werb, Z.: Matrix metalloproteinases: effectors of
stimulate glycosaminoglycan synthesis by human skin fibro- development and normal physiology. Genes Dev, 14: 2123,
blasts. Lab Invest, 53: 30, 1985 2000
57. Hakkinen, L., Westermarck, J., Kahari, V. M. et al: Human 78. Folkman, J.: What is the evidence that tumors are angiogenesis
granulation-tissue fibroblasts show enhanced proteoglycan dependent? J Natl Cancer Inst, 82: 4, 1990
gene expression and altered response to TGF-beta 1. J Dent 79. Hanahan, D. and Folkman, J.: Patterns and emerging mecha-
Res, 75: 1767, 1996 nisms of the angiogenic switch during tumorigenesis. Cell,
58. Giri, D., Ropiquet, F. and Ittmann, M.: Alterations in expres- 86: 353, 1996
sion of basic fibroblast growth factor (FGF) 2 and its receptor 80. Ferrer, F. A., Miller, L. J., Andrawis, R. I. et al: Vascular
FGFR-1 in human prostate cancer. Clin Cancer Res, 5: 1063, endothelial growth factor (VEGF) expression in human pros-
1999 tate cancer: in situ and in vitro expression of VEGF by human
59. Dow, J. K. and deVere White, R. W.: Fibroblast growth factor 2: prostate cancer cells. J Urol, 157: 2329, 1997
its structure and property, paracrine function, tumor angio- 81. Mazzucchelli, R., Montironi, R., Santinelli, A. et al: Vascular
genesis, and prostate-related mitogenic and oncogenic func- endothelial growth factor expression and capillary architec-
tions. Urology, 55: 800, 2000 ture in high-grade PIN and prostate cancer in untreated and
60. Yan, G., Fukabori, Y., McBride, G. et al: Exon switching and androgen-ablated patients. Prostate, 45: 72, 2000
activation of stromal and embryonic fibroblast growth factor 82. Jackson, M. W., Bentel, J. M. and Tilley, W. D.: Vascular
(FGF)-FGF receptor genes in prostate epithelial cells accom- endothelial growth factor (VEGF) expression in prostate can-
pany stromal independence and malignancy. Mol Cell Biol, cer and benign prostatic hyperplasia. J Urol, 157: 2323, 1997
13: 4513, 1993 83. Levine, A. C., Liu, X. H., Greenberg, P. D. et al: Androgens
61. van der Voort, R., Taher, T. E., Derksen, P. W. et al: The induce the expression of vascular endothelial growth factor in
hepatocyte growth factor/Met pathway in development, tu- human fetal prostatic fibroblasts. Endocrinology, 139: 4672,
morigenesis, and B-cell differentiation. Adv Cancer Res, 79: 1998
39, 2000 84. Balbay, M. D., Pettaway, C. A., Kuniyasu, H. et al: Highly
62. Humphrey, P. A., Zhu, X., Zarnegar, R. et al: Hepatocyte metastatic human prostate cancer growing within the pros-
growth factor and its receptor (c-MET) in prostatic carci- tate of athymic mice overexpresses vascular endothelial
noma. Am J Pathol, 147: 386, 1995 growth factor. Clin Cancer Res, 5: 783, 1999
63. Nishimura, K., Kitamura, M., Miura, H. et al: Prostate stromal 85. Haggstrom, S., Lissbrant, I. F., Bergh, A. et al: Testosterone
cell-derived hepatocyte growth factor induces invasion of induces vascular endothelial growth factor synthesis in the
prostate cancer cell line DU145 through tumor-stromal inter- ventral prostate in castrated rats. J Urol, 161: 1620, 1999
action. Prostate, 41: 145, 1999 86. Joseph, I. B., Nelson, J. B., Denmeade, S. R. et al: Androgens
64. Johnsen, M., Lund, L. R., Romer, J. et al: Cancer invasion and regulate vascular endothelial growth factor content in normal
tissue remodeling: common themes in proteolytic matrix deg- and malignant prostatic tissue. Clin Cancer Res, 3: 2507,
radation. Curr Opin Cell Biol, 10: 667, 1998 1997
65. Mignatti, P., Rifkin, D. B., Welgus, H. G. et al: Proteinases and 87. Borgstrom, P., Bourdon, M. A., Hillan, K. J. et al: Neutralizing
tissue remodeling. In: The Molecular and Cellular Biology of anti-vascular endothelial growth factor antibody completely
Wound Repair, 2nd ed. Edited by R. A. F. Clark. New York: inhibits angiogenesis and growth of human prostate carci-
Plenum Press, p. 427, 1996 noma micro tumors in vivo. Prostate, 35: 1, 1998
66. MacDougall, J. R. and Matrisian, L. M.: Contributions of tumor 88. Kirschenbaum, A., Wang, J., Ren, M. et al: Inhibition of vascu-
and stromal matrix metalloproteinases to tumor progression, lar endothelial cell growth factor suppresses the in vivo
invasion and metastasis. Cancer Metastasis Rev, 14: 351, 1995 growth of human prostate tumors. Urol Oncol, 3: 3, 1997
67. Schmitt, M., Janicke, F., Moniwa, N. et al: Tumor-associated 89. Melnyk, O., Zimmerman, M., Kim, K. J. et al: Neutralizing
urokinase-type plasminogen activator: biological and clinical anti-vascular endothelial growth factor antibody inhibits fur-
significance. Biol Chem Hoppe Seyler, 373: 611, 1992 ther growth of established prostate cancer and metastases in
68. Hoosein, N. M., Boyd, D. D., Hollas, W. J. et al: Involvement of a pre-clinical model. J Urol, 161: 960, 1999
urokinase and its receptor in the invasiveness of human 90. Gerber, H. P., Kowalski, J., Sherman, D. et al: Complete inhi-
prostatic carcinoma cell lines. Cancer Commun, 3: 255, 1991 bition of rhabdomyosarcoma xenograft growth and neovascu-
69. Stearns, M. E., Fudge, K., Garcia, F. et al: IL-10 inhibition of larization requires blockade of both tumor and host vascular
human prostate PC-3 ML cell metastases in SCID mice: IL-10 endothelial growth factor. Cancer Res, 60: 6253, 2000
stimulation of TIMP-1 and inhibition of MMP-2/MMP-9 ex- 91. Janvier, R., Sourla, A., Koutsilieris, M. et al: Stromal fibro-
pression. Invasion Metastasis, 17: 62, 1997 blasts are required for PC-3 human prostate cancer cells to
70. Nielsen, B. S., Sehested, M., Timshel, S. et al: Messenger RNA produce capillary-like formation of endothelial cells in a
for urokinase plasminogen activator is expressed in myofi- three-dimensional co-culture system. Anticancer Res, 17:
broblasts adjacent to cancer cells in human breast cancer. 1551, 1997
Lab Invest, 74: 168, 1996 92. Pirtskhalaishvili, G. and Nelson, J. B.: Endothelium-derived
71. Wood, M., Fudge, K., Mohler, J. L. et al: In situ hybridization factors as paracrine mediators of prostate cancer progression.
studies of metalloproteinases 2 and 9 and TIMP-1 and Prostate, 44: 77, 2000
TIMP-2 expression in human prostate cancer. Clin Exp Me- 93. Senger, D. R., Brown, L. F., Claffey, K. P. et al: Vascular
tastasis, 15: 246, 1997 permeability factor, tumor angiogenesis and stroma genera-
72. Jung, K., Lein, M., Ulbrich, N. et al: Quantification of matrix tion. Invasion Metastasis, 14: 385, 1994
metalloproteinases and tissue inhibitors of metalloproteinase 94. Abdulkadir, S. A., Carvalhal, G. F., Kaleem, Z. et al: Tissue
in prostatic tissue: analytical aspects. Prostate, 34: 130, 1998 factor expression and angiogenesis in human prostate carci-
73. Soff, G. A., Sanderowitz, J., Gately, S. et al: Expression of noma. Hum Pathol, 31: 443, 2000
plasminogen activator inhibitor type 1 by human prostate 95. Shimura, S., Yang, G., Ebara, S. et al: Reduced infiltration of
REACTIVE STROMA IN PROSTATE CANCER PROGRESSION 2483
tumor-associated macrophages in human prostate cancer: as- 114. Williams, R. H., Stapleton, A. M., Yang, G. et al: Reduced levels
sociation with cancer progression. Cancer Res, 60: 5857, 2000 of transforming growth factor beta receptor type II in human
96. Riches, D. W. H.: Macrophage involvement in wound repair, prostate cancer: an immunohistochemical study. Clin Cancer
remodeling, and fibrosis. In: The Molecular and Cellular Bi- Res, 2: 635, 1996
ology of Wound Repair, 2nd ed. Edited by R. A. F. Clark. New 115. Borsi, L., Castellani, P., Risso, A. M. et al: Transforming growth
York: Plenum Press, p. 95, 1996 factor-beta regulates the splicing pattern of fibronectin mes-
97. Elgert, K. D., Alleva, D. G. and Mullins, D. W.: Tumor-induced senger RNA precursor. FEBS Lett, 261: 175, 1990
immune dysfunction: the macrophage connection. J Leukoc 116. Brogi, E., Wu, T., Namiki, A. et al: Indirect angiogenic cyto-
Biol, 64: 275, 1998 kines upregulate VEGF and bFGF gene expression in vascu-
98. Mantovani, A., Bottazzi, B., Colotta, F. et al: The origin and lar smooth muscle cells, whereas hypoxia upregulates VEGF
function of tumor-associated macrophages. Immunol Today, expression only. Circulation, 90: 649, 1994
13: 265, 1992 117. Igarashi, A., Okochi, H., Bradham, D. M. et al: Regulation of
99. Sunderkotter, C., Steinbrink, K., Goebeler, M. et al: Macro- connective tissue growth factor gene expression in human
phages and angiogenesis. J Leukoc Biol, 55: 410, 1994 skin fibroblasts and during wound repair. Mol Biol Cell, 4:
100. Dingemans, K. P., Zeeman-Boeschoten, I. M., Keep, R. F. et al: 637, 1993
Transplantation of colon carcinoma into granulation tissue 118. Pertovaara, L., Kaipainen, A., Mustonen, T. et al: Vascular
induces an invasive morphotype. Int J Cancer, 54: 1010, 1993 endothelial growth factor is induced in response to transform-
101. Elenbaas, B., Spirio, L., Koerner, F. et al: Human breast cancer ing growth factor-beta in fibroblastic and epithelial cells.
cells generated by oncogenic transformation of primary mam- J Biol Chem, 269: 6271, 1994
mary epithelial cells. Genes Dev, 15: 50, 2001 119. Story, M. T., Hopp, K. A. and Meier, D. A.: Regulation of basic
102. Olumi, A. F., Grossfeld, G. D., Hayward, S. W. et al: Carcinoma- fibroblast growth factor expression by transforming growth
associated fibroblasts direct tumor progression of initiated factor beta in cultured human prostate stromal cells. Pros-
human prostatic epithelium. Cancer Res, 59: 5002, 1999 tate, 28: 219, 1996
103. Gleave, M., Hsieh, J. T., Gao, C. A. et al: Acceleration of human 120. Stearns, M. E., Garcia, F. U., Fudge, K. et al: Role of interleukin
prostate cancer growth in vivo by factors produced by pros- 10 and transforming growth factor beta1 in the angiogenesis
tate and bone fibroblasts. Cancer Res, 51: 3753, 1991 and metastasis of human prostate primary tumor lines from
104. Massague, J.: The transforming growth factor-beta family. orthotopic implants in severe combined immunodeficiency
Annu Rev Cell Biol, 6: 597, 1990 mice. Clin Cancer Res, 5: 711, 1999
105. Roberts, A. B. and Sporn, M. B.: Transforming growth factor-␤.
121. Ueki, N., Nakazato, M., Ohkawa, T. et al: Excessive production
In: The Molecular and Cellular Biology of Wound Repair, 2nd
of transforming growth-factor beta 1 can play an important
ed. Edited by R. A. F. Clark. New York: Plenum Press, p. 275,
role in the development of tumorigenesis by its action for
1996
angiogenesis: validity of neutralizing antibodies to block tu-
106. Roberts, A. B., Sporn, M. B., Assoian, R. K. et al: Transforming
mor growth. Biochim Biophys Acta, 1137: 189, 1992
growth factor type beta: rapid induction of fibrosis and an-
122. Moussad, E. E. and Brigstock, D. R.: Connective tissue growth
giogenesis in vivo and stimulation of collagen formation in
vitro. Proc Natl Acad Sci USA, 83: 4167, 1986 factor: what’s in a name? Mol Genet Metab, 71: 276, 2000
107. Desmouliere, A., Geinoz, A., Gabbiani, F. et al: Transforming 123. Frazier, K. S. and Grotendorst, G. R.: Expression of connective
growth factor-beta 1 induces alpha-smooth muscle actin ex- tissue growth factor mRNA in the fibrous stroma of mam-
pression in granulation tissue myofibroblasts and in quies- mary tumors. Int J Biochem Cell Biol, 29: 153, 1997
cent and growing cultured fibroblasts. J Cell Biol, 122: 103, 124. Sporn, M. B. and Roberts, A. B.: Peptide growth factors are
1993 multifunctional. Nature, 332: 217, 1988
108. Ronnov-Jessen, L. and Petersen, O. W.: Induction of alpha- 125. Heldin, C. H. and Westermark, B.: Mechanism of action and in
smooth muscle actin by transforming growth factor-beta 1 in vivo role of platelet-derived growth factor. Physiol Rev, 79:
quiescent human breast gland fibroblasts. Implications for 1283, 1999
myofibroblast generation in breast neoplasia. Lab Invest, 68: 126. Forsberg, K., Valyi-Nagy, I., Heldin, C. H. et al: Platelet-
696, 1993 derived growth factor (PDGF) in oncogenesis: development of
109. Lee, C., Sintich, S. M., Mathews, E. P. et al: Transforming a vascular connective tissue stroma in xenotransplanted hu-
growth factor-beta in benign and malignant prostate. Pros- man melanoma producing PDGF-BB. Proc Natl Acad Sci
tate, 39: 285, 1999 USA, 90: 393, 1993
110. Steiner, M. S. and Barrack, E. R.: Transforming growth factor- 127. Fudge, K., Bostwick, D. G. and Stearns, M. E.: Platelet-derived
beta 1 overproduction in prostate cancer: effects on growth in growth factor A and B chains and the alpha and beta receptors
vivo and in vitro. Mol Endocrinol, 6: 15, 1992 in prostatic intraepithelial neoplasia. Prostate, 29: 282, 1996
111. Gerdes, M. J., Larsen, M., McBride, L. et al: Localization of 128. Sitaras, N. M., Sariban, E., Bravo, M. et al: Constitutive pro-
transforming growth factor-beta1 and type II receptor in duction of platelet-derived growth factor-like proteins by hu-
developing normal human prostate and carcinoma tissues. man prostate carcinoma cell lines. Cancer Res, 48: 1930, 1988
J Histochem Cytochem, 46: 379, 1998 129. Vlahos, C. J., Kriauciunas, T. D., Gleason, P. E. et al: Platelet-
112. Guo, Y., Jacobs, S. C. and Kyprianou, N.: Down-regulation of derived growth factor induces proliferation of hyperplastic
protein and mRNA expression for transforming growth human prostatic stromal cells. J Cell Biochem, 52: 404, 1993
factor-beta (TGF-beta1) type I and type II receptors in human 130. Weidner, N., Carroll, P. R., Flax, J. et al: Tumor angiogenesis
prostate cancer. Int J Cancer, 71: 573, 1997 correlates with metastasis in invasive prostate carcinoma.
113. Kim, I. Y., Ahn, H. J., Zelner, D. J. et al: Loss of expression of Am J Pathol, 143: 401, 1993
transforming growth factor beta type I and type II receptors 131. Halvorsen, T. B. and Seim, E.: Association between invasive-
correlates with tumor grade in human prostate cancer tis- ness, inflammatory reaction, desmoplasia and survival in
sues. Clin Cancer Res, 2: 1255, 1996 colorectal cancer. J Clin Pathol, 42: 162, 1989

You might also like