Cancer associated fibroblasts

2 downloads 0 Views 2MB Size Report
Oct 25, 2011 - Allrightsreserved.http://dx.doi.org/10.1016/j.molmed.2013.05.004. Corresponding authors: Madar, S. ([email protected]);. Rotter, V.
Opinion

‘Cancer associated fibroblasts’ – more than meets the eye Shalom Madar*, Ido Goldstein*, and Varda Rotter Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, 76100, Israel

Cancer associated fibroblasts (CAFs) are a subpopulation of cells that reside within the tumor microenvironment and promotes the transformation process by encouraging tumor growth, angiogenesis, inflammation, and metastasis. CAF-specific proteins serve as both prognostic markers and targets for anticancer drugs. With the growing interest in CAFs, several controversial issues have been raised, including the genomic landscape of these cells, the identity of specific markers, and their cell of origin. Here, we tackle these debated issues and put forward a new definition for ‘CAF’ as a cell ‘state’ rather than a cell type. We hope this conceptualization can resolve the ongoing discrepancies revolving around CAF research and aid in designing better anti-cancer treatment strategies. The tumor microenvironment and cancer associated fibroblasts The tumor microenvironment (see Glossary) can be crudely defined as the sum of all the non-transformed elements residing within or in the vicinity of a tumor, including, for example, nearby tissue, vasculature, and immune cells. One well-studied and populous component of the tumor microenvironment is cancer associated fibroblasts (CAFs), a subpopulation of the tumor–stroma milieu that in many cases dictates tumor outcome [1]. Normal fibroblasts are elongated cells residing in connective tissues and are responsible for the synthesis and turnover of the extracellular matrix (ECM). Fibroblasts control and support normal tissue homeostasis and participate in a plethora of biological processes such as wound healing and senescence. Unlike normal fibroblasts, CAFs either reside within the tumor margins or infiltrate the tumor mass, and facilitate the transformation process [2]. Other than their location, the exact definition of CAFs is elusive for several reasons. First, given that these cells are genetically stable and not the cause of disease, their definition seems to depend on the presence of the adjacent tumor. Moreover, CAFs share several markers with other inhabitants of the stroma, such as myofibroblasts, myoepithelial cells, muscle cells, mesenchymal stem cells (MSCs), and endothelia [3]. Thus, identifying and isolating the CAF population has proven to be an arduous task. Another debated issue is the origin Corresponding authors: Madar, S. ([email protected]); Rotter, V. ([email protected]) * These authors are equal contributors. Keywords: stroma; microenvironment; CAFs; inflammation; mutation; stem cells. 1471-4914/$ – see front matter ß 2013 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.molmed.2013.05.004

of CAFs; many origins have been proposed, ranging from normal fibroblasts and MSCs to trans-differentiated epithelial and endothelial cells [1,4]. These challenges are discussed in greater detail below, and by exploring the facts gathered to date, we reconstruct and present a novel definition of CAFs – ‘CAF state’ – which we believe will help to resolve some of the debate surrounding these cells. CAF phenotype Intrinsic CAF phenotype The phenotype of CAFs isolated from malignant tissues is distinct from that of normal fibroblasts, including a more rapid proliferation rate, enhanced collagen production, secretion of growth factors and other ECM modulators, and the activation of unique expression programs [5–7]. In multiple instances, CAFs have been shown to proliferate faster than normal fibroblasts. For example, CAFs extracted from oral carcinoma exhibit an increased proliferation rate and mitosis ability compared with normal fibroblasts [8], and similarly, CAFs from cutaneous squamous cell carcinoma display enhanced proliferation and migration compared with their normal counterparts [9]. Extrinsic CAF phenotype – tumor enhancing effects Apart from an increased division rate, the gold standard for identifying CAFs is their capacity to promote cancer progression in vivo, usually when coinjected with tumor cells or when recruited to the tumor site [10–12]. Interestingly, irradiated fibroblasts, or even the media in which they were grown, are sufficient to enhance the tumorigenicity of breast cancer grafts, suggesting that a transient effect induced by CAF-secreted factors is at play [13]. Among the secreted factors that mediate the tumor promoting effect of CAFs is CXCL12 (SDF1-a), a chemokine that can induce angiogenesis and enhance the proliferative capacity of cancer cells [14]. Tumor growth factor b (TGF-b) signaling in fibroblasts has also been reported to modulate Glossary Cancer associated fibroblasts (CAFs): fibroblasts residing within the tumor mass with the ability to promote tumorigenic features. Epithelial-to-mesenchymal transition (EMT): a differentiation process which converts cells of epithelial origin into mesenchymal cells. EMT occurs in normal development and wound healing and is harnessed by cancer cells mainly as a means to acquire migratory capacity – a vital step towards metastasis. Microenvironment/tumor stroma: all the non-transformed components in the vicinity of the tumor. These elements include cells of the immune system (such as macrophages and lymphocytes), blood vessel cells, fibroblasts, myofibroblasts, mesenchymal stem cells, adipocytes, and the ECM. Trends in Molecular Medicine, August 2013, Vol. 19, No. 8

447

Opinion the oncogenic potential of adjacent epithelia [15], a connection between these two secreted factors has been observed: CXCL12 secreted from CAFs can act via TGF-bregulated CXCR4 in adjacent epithelial cells [16]. The epithelial cells, in turn, activate the AKT pathway and elicit tumorigenesis. Furthermore, overexpression of TGFb and hepatocyte growth factor (HGF) in stromal cells, followed by their coinjection with breast organoids, results in an increased tumor incidence compared with coinjection of organoids and normal fibroblasts [17]. The ECM modulators matrix metalloproteinases (MMPs) are also governed by the tumor stroma. CAFs obtained from breast cancer patients display elevated levels of MMP1 compared with normal fibroblasts, whereas the latter upregulate MMP1 expression when subjected to soluble breast cancer factors [18]. Moreover, MMP1 originating from the stroma has been reported to augment the migration and invasion of breast cancer cells [19]. CAFs can promote tumor drug resistance, for example, CAFs bypass anti-vascular endothelial growth factor (antiVEGF) treatment by activating the platelet-derived growth factor C (PDGF-C) pathway [20]. CAFs were also shown to confer various cancer cell lines with resistance to RAF inhibitors through HGF secretion [21]. Ablation of fibroblast activation protein (FAP)+ stromal cells in mice hampers tumor resistance to therapeutic vaccination [22]. Finally, inhibition of hedgehog signaling in the stroma of mice enabled gemcitabine antitumor effect in a pancreatic cancer model [23]. Recently Luga et al. revealed a novel form of communication between CAFs and tumor cells, whereas CAFs exosomes mobilize autocrine Wnt–PCP signaling to promote breast cancer invasiveness [24]. On a broader scope, by altering the ECM composition CAFs can influence tumor metastasis (Box 1). Finally, CAFs secrete proinflammatory cytokines as they partake in cancer-related inflammation (Box 2). The origin of CAFs Intuitively, CAFs are grasped as normal fibroblasts that have been altered as a result of continuous exposure to cancer cells. Indeed, CAFs often share several similarities with normal fibroblasts [25]. ‘Cell plasticity’ is a term describing the ability of cells to alter their differentiation status [26], and numerous studies suggest that several cell types of both mesenchymal and epithelial origin display some degree of plasticity in the vicinity of tumors and provide an alternative source for CAFs. In general, MSCs assume several roles during cancer progression, such as differentiating into blood vessel supporting cells [27] or recruiting endothelial precursor cells [14]. Recent reports support the notion that MSCs are also a possible source for CAFs. In gastrointestinal cancer female patients who received bone marrow transplants from male donors, a significant proportion of a smooth muscle actin (aSMA) expressing CAFs were of donor origin, most likely recruited from the donor bone marrow [28]. Using a mouse model, Ishii et al. reported that transplanted bone marrow cells can be detected in the margins of a grafted tumor, where they differentiate into endothelial cells and myofibroblasts [29]. Similar mouse models 448

Trends in Molecular Medicine August 2013, Vol. 19, No. 8

Box 1. The involvement of CAFs in metastasis formation The tumor–stroma interface marks a physical barrier that contains the tumor and prevents its spread. During the course of malignancy this barrier is breached by cancer cells that escape the tumor mass and are now free to roam the bloodstream and colonize in a distant site. Several reports allude to the contribution of CAFs to the metastatic process. Olaso et al. documented the presence of intrametastatic aSMA-expressing cells appearing at the early stages of hepatic metastasis derived from B-16 melanoma cells in a mouse model [90]. Labeled CAFs were also reported to be recruited by liver metastases in vivo [10]. The elevated presence of CAFs in metastatic human specimens was also evident by immunostaining in several types of cancer [91]. In another study, melanoma cells obtained from different stages of the disease were co-cultured with normal fibroblasts. The growth rate of early stage cells was attenuated by fibroblasts, whereas metastatic cells were not affected by their presence, indicating a selective pressure imposed by fibroblasts, one that selects for outgrowing metastatic clones [92]. Both tumor cells and hematopoietic cells ‘educate’ CAFs in a secondary site to form a niche for metastasis [93,94]. In these sites, CAFs ‘feed’ the secondary tumor with secreted factors which support its growth [95]. CAFs are inherently equipped with motility and migratory capacities that can be exploited by tumor cells. In a 3D ‘organotypic’ invasion assay, carcinoma cells utilized CAF characteristics in order to invade without the need to undergo EMT [96]. Another study suggests that under harsh hypoxic conditions, tumor cells elevate CXCR4 which allows them to migrate towards a gradient of CAFinduced CXCL12 and escape to a normoxic environment at a distant site [97]. Overall, CAFs seem to be abundant at metastatic tumor sites and promote the transition of in situ tumors towards malignancy by affecting the rate-limiting steps of the process.

revealed that labeled bone marrow cells introduced into tumor-bearing mice constitute 20–25% of the stromal fibroblasts in the vicinity of the tumor [30,31]. CAFs expressing high levels of CD90 (a common MSC marker) were isolated and, despite being tumor-supportive, failed to express other known MSC markers, except for CD24 [32]. Nevertheless, it is plausible that the markers CD90 and CD24 are remnants of formerly undifferentiated MSCs. Finally, human MSCs treated with conditioned media of breast cancer cells exhibit CAF-like behavior including elevated expression of aSMA and CXCL12 and tumor-supportive capabilities in vitro and in vivo [33]. Another plausible source of CAFs is smooth muscle cells (SMCs). Normal prostate stroma is enriched with SMCs expressing high levels of aSMA [34]. This population is thought to disappear during prostate cancer progression and be replaced by CAFs [35]. Prostatic SMCs express high levels of androgen receptor (AR) and are androgen-responsive. By contrast, Wikstrom et al. documented the downregulation of AR in the prostate tumor stroma and therefore hypothesized that SMCs might be the source of CAFs [36]. Myofibroblasts (a term that is sometimes used interchangeably with CAFs) are considered to be ‘activated’ fibroblasts that assume an active role in wound healing as well as in scaring and fibrotic lesions. SchmittGra¨ff et al. suggested that myofibroblasts might acquire a CAF phenotype when the proper stimuli are present [37]. CAFs may originate from recruited and differentiated circulating fibrocytes. Invasive ductal carcinomas of the breast were found to contain fewer fibrocytes but more differentiated aSMA+ fibroblasts than in situ lesions, indicating a possible differentiation of fibrocytes into CAFs within the breast stroma [38].

Opinion Box 2. CAFs and cancer-related inflammation The link between cancer and inflammation was first postulated by Virchow in 1863 who maintained that inflammation predisposes individuals to cancer. Indeed, studies revealed that persistent inflammation lasting over a decade is a major risk factor in various cancers [98]. Recently, several reports have documented a link between CAFs and cancer-related inflammation in which a common pattern emerges. First, the tumor cells secrete CAF-activating factors such as IL-1 and TNF-a [25,73–76]. Following this activation, CAFs initiate a proinflammatory response that includes, among others, IL6 and IL-8 [75,76]. These secreted factors may affect tumor growth in a direct manner or induce inflammation by recruiting components of the immune system [30,73,74]. Interestingly, both normal fibroblasts and CAFs are capable of evoking a proinflammatory response [99]. Accordingly, CAFs produced from liver metastases and normal liver fibroblasts are both able to induce IL-6 [100]. Combined, these results suggest that normal fibroblasts possess the capacity to induce a proinflammatory response, and this capacity is retained after their transition to CAFs, as it becomes beneficial for the evolving tumor.

Cells of epithelial origin are also candidates as suppliers for the CAF population. Carcinomas are the most frequent type of cancer, and although they originate from an epithelial lineage, they might also contribute to the CAF pool. For example, breast cancer cells from a biopsy specimen that were suspected to have undergone an epithelial-tomesenchymal transition (EMT) were extracted and further characterized in vitro. These cells share a specific chromosomal aberration with the adjacent tumor cells and express residual levels of keratin, confirming their epithelial origin. By contrast, they exhibit fibroblast morphology, express high levels of aSMA, and although they cannot form tumors, they increase the size of tumors from a coinjected breast cancer cell line in nude mice [39]. In another study, GFP-labeled cancer cells were shown to provide 25% of the adjacent CAF population [40]. Endothelial cells are yet another emerging source for CAFs. Zeisberg et al. reported that endothelial cells treated with TGF-b exhibit CAF morphology and elevated fibroblast-specific protein-1 (FSP1) expression, coupled with decreased expression of the endothelial marker CD31. This phenomenon is also evident in a mouse model [41]. Two recent studies have disclosed that pericytes give rise to scar-forming stromal cells at injury sites [42,43]. This implies that perivascular cells, often found in the vicinity of tumor sites, are yet another source for CAF precursors; however, future studies should test this hypothesis. Recently, adipose tissue derived stem cells were reported to differentiate into CAF-like cells following exposure to conditioned media from breast cancer cell lines [44]. Common CAF markers and gene signatures In general, normal fibroblasts display vast heterogeneity manifested by differences in appearance, behavior, and gene expression (reviewed in [45]). On top of the heterogeneity found within the fibroblasts, other stromal residents resemble fibroblasts, especially with regard to the expression of ‘specific’ markers. Thus, the heterogeneity of the stromal compartment, combined with the presence of

Trends in Molecular Medicine August 2013, Vol. 19, No. 8

multiple subpopulations of fibroblasts, make it difficult to properly isolate and define CAFs based on the expression of specific markers. Nevertheless, CAF markers are widely described in the literature. Among the downregulated markers are caveolin-1 [46] and laminin [47], whereas the CAF markers most frequently reported to be upregulated include aSMA [48], vimentin [8], and FSP1 [49]. By contrast, Sugimoto et al. used a mouse model of cancer to compare six markers belonging to several mesenchymal groups that inhabit the stroma. aSMA, plateletderived growth factor receptor b (PDGFRb), and chondroitin sulfate proteoglycan 4 overlapped with each other in identifying fibroblasts, myofibroblasts, and vascular related cells; vimentin and type I collagen were also not specific to fibroblasts [49]. This work questions the specificity of these commonly accepted CAF markers. Moreover, aSMA was reported to be downregulated rather than upregulated in the stromal compartment of prostate cancer [50]. These findings emphasize the heterogeneity of the stromal compartment and lead us to surmise there is no known unequivocal marker to distinguish between mesenchymal subpopulations. Despite the findings mentioned above indicating aSMA elevation is both cancer-type specific and a general marker for mesenchymal cells, it is wrongfully accepted as the definitive marker for all CAFs [3,51]. Several studies have identified a ‘CAF gene expression profile’ [52–54] and even ‘CAF signatures’ that are predictive of tumor outcome [55,56]. Closer examination of these studies reveals that on the specific gene level, the differences outnumber the commonalities. However, if we zoom out of the gene level and examine gene families, different types and stages of cancers yield similar groups of genes – cell adhesion, immune response, and ECM modulation [52–54]. This is exactly what we would expect to find from different cell types under similar conditions – converging into performing the same tasks in different ways (i.e., genes). A novel definition for the term CAFs The inconsistency of CAF markers/gene signatures, which represents the heterogeneity of the stromal compartment and the ample reservoir for their cell of origin, suggest that the tumor and its microenvironment exhibit a considerable degree of plasticity that is not restricted to a specific germ layer. This situation warrants a new definition for the term ‘CAFs’. In the field of stem cell research, a similar controversy arose as to the exact definition of ‘stem cell’. An elegant solution for this dispute defined ‘stemness’ as a state in the life cycle of a cell [26]. By the same token, ‘CAFs’ should be defined as the dynamic state of fibroblast-like cells found in the vicinity of the tumor that promote its progression – for brevity a ‘CAF state’. But how do cells maintain their ‘CAF state’ for long periods of time? If we perceive the tumor microenvironment as an evolving element, it might be useful to borrow evolution based concepts to resolve this conundrum. Accordingly, three possible mechanisms come to mind: genetic mutations, epigenetic alterations, and persistent environmental effects. 449

Opinion Genetic mutations – the genomic landscape of CAFs A huge debate is being held over the genomic landscape of stromal cells. Generally speaking, the stromal compartment surrounding tumors is perceived as genetically stable, especially when compared with the rapidly dividing tumor cells that constitute most of the tumor mass. Nevertheless, some studies have described genomic modifications of cancer associated stroma, including amplifications, loss of heterozygosity (LOH), and smaller mutations. After injecting human prostate cancer cells into nude mice, Pathak et al. found mouse stromal cells harboring multiple copies of mouse chromosome 15 in the vicinity of the tumors [57]. Microdissection of stromal and cancer cells from prostate tumors, followed by PCR, revealed LOH of chromosome 8p in all of the tested tumors and in three out of nine mesenchymal samples [58]. LOH of p53 was also documented in microdissected stromal cells of breast and colon cancers [59]. Furthermore, microdissected stromal and epithelial tissues of 11 breast samples exhibited LOH in specific DNA loci, both in tumor and stromal cells. Importantly, several such incidents of LOH were exclusively detected in the stroma and, strikingly, some were evident in distant stromal cells [60], suggesting that the stromal cell population is heterogeneous and distinct from cancer cells and may consist of genetically unstable clones. Finally, additional studies have described p53 and PTEN mutations in the stromal compartment of breast cancer patients [59,61,62]. In overwhelming contrast to this, other reports portray the stromal population as genetically stable. Accordingly, no karyotypic abnormalities were found in the CAFs of oral carcinoma [8]. CAFs isolated from pancreatic cancer do not exhibit somatic copy variation or immunohistological evidence for p53 mutations [63]. Allinen et al. thoroughly examined breast CAFs and discovered that despite dramatic gene expression changes in CAFs compared with normal fibroblasts, no genetic alterations are evident [64]. In another study, breast CAFs extracted from 25 tumors produced only one sample that exhibited copy number variation and p53 mutation [65]. Similar results have been obtained in ovarian cancer samples [66]. What could be the reason for such an enormous discrepancy arising in the literature regarding the genomic landscape of CAFs? Bearing in mind the ‘CAF state’ hypothesis presented above, we should consider the possibility that the CAF population does not necessarily descend from normal fibroblasts. The genetically abnormal stroma might represent a subpopulation of carcinoma cells that underwent EMT. For example, 38 genes showed similar genetic alterations (either gain or loss of genetic material) in stromal and epithelial compartments of 11 ovarian tumor pairs [67], suggesting that these two cell populations originated from the same mother cell despite the different morphological appearance. Moreover, chromogenic in situ hybridization of breast cancer samples revealed that the genetic features detected in tumor cells were also present in a minority of fibroblast-like cells near the epithelial clusters, indicating that some of these cells are clonal and derived from the epithelial tumor cells [68]. Fukino et al. found more correlations between clinicopathological features and the LOH/allelic imbalance in the stroma than 450

Trends in Molecular Medicine August 2013, Vol. 19, No. 8

in the epithelium of breast cancer samples [69], yet no ‘stromal-like’ tumors have been reported in this study. The fact that stromal cells multiply in a much lower rate render them less prone to the major selective forces imposed on highly prolific cancer cells. If, indeed, stromal cells were equally genetically instable, we would expect to find aggressive CAF clones giving rise to sarcoma-like tumors alongside carcinomas. As this is clearly not the case, we presume that the genetic alterations in CAFs are more sparse and subtle. In our opinion, it is more plausible that the reported ‘stromal’ populations that exhibit distinct mutations are merely subgroups of cancer cells which underwent EMT. Epigenetic alterations An alternative mechanism to CAF genomic aberrations is epigenetic alterations. A recent interesting study from the Witte group revealed that overexpression of an epigenetic regulator in the stroma leads to increased Wnt/b-catenin signaling which results in neoplastic lesions of adjacent epithelial cells [70]. These findings coincide with previous reports on aberrant Wnt/b-catenin signaling in CAFs [71]. Further evidence supporting the CAF epigenetic regulation notion are thoroughly reviewed by Rudnick and Kuperwasser [72]. Persistent environmental effects Another way by which CAFs might sustain their ‘CAF state’ is the constant presence of signals originating from tumor cells. As mentioned in Box 2, tumors are known to secrete CAF-activating factors such as interleukin-1 (IL-1) and tumor necrosis factor a (TNF-a) [25,73–76]. Epidermal growth factor (EGF) released from breast cancer cells ‘educate’ CAFs to enhance the production and secretion of leptin, eventually leading to tumor progression [77]. A recent study from our laboratory revealed that tumor cells induce ATF3 elevation in adjacent stromal cells [5]. Moreover, when ATF3 was overexpressed in three distinct types of CAFs (produced from prostate, breast, and lung cancers), the result was a very similar gene expression program, and a capacity to promote tumor growth. Abundance of a tumor signal could be the result of a specific mutation in the tumor itself [78]. The concept of ‘extended phenotype’ was first put forward by Richard Dawkins – suggesting that a gene in an organism might alter its environment [79]. Thus, if we accept the notion that CAFs are genetically stable, we can attribute the ‘CAF state’ to an ‘extended phenotype’ of tumor mutations. Box 3. Outstanding questions  Does a stromal subpopulation consisting of a high mutation rate exist, or is it a cancerous subpopulation with a mesenchymal appearance?  Would stromal-targeting therapies be sufficient to cure cancer or would they need to be coadministered with tumor-targeting drugs?  Are there any other stromal subpopulations which exist as ‘states’ and not as cell types?  If CAFs are indeed genetically stable, what are the epigenetic mechanisms that dictate their unique gene expression and behavior?  What cell surface elements govern the tumor–stroma interaction?

Opinion

Trends in Molecular Medicine August 2013, Vol. 19, No. 8

Although the secreted factors involved in the tumor– stroma interaction are widely studied and mentioned throughout this review, the cell surface determinants that govern this interaction are an interesting yet currently understudied angle (Box 3). A recent study reported on the intercellular transfer of Ras oncoproteins from melanoma to adjacent T cells [80]. Future studies should be dedicated to exploring this type of interaction. Concluding remarks and future perspectives The tumor microenvironment has become an important pillar in the cancer research field with research on CAFs being one of its main focuses. However, several persistent discrepancies have accumulated and warrant taking a new perspective. In this review, we suggest to conceive CAF traits (namely, mesenchymal appearance and tumor promoting ability) as a ‘state’ rather than a cell type. ‘CAF state’ might be exhibited by cells of different origins. But how could this type of information be translated into clinical practice? In recent years, the targets of several drugs are stromal components rather than the tumor itself [51]. These drugs target blood vessel growth, fibroblast-specific proteins, and secreted ECM residents. Now let us consider a tumor containing ‘CAF state’ cells (depicted in Figure 1). To tailor the proper stromal-related drugs to the right patient, we first need to establish the identity of these cells. This could be done based on the expression of a specific marker (note that a tumor might consist of several stromal subpopulations). In the next step, we should administer drug(s) that target the predominant cell type or rather a combination of drugs targeting more than one population in the stromal compartment.

For example, FAP is an integral membrane serine protease, often upregulated in fibroblasts [81]. Several FAP-related drugs have been developed, among them anti-FAP antibodies [82,83] and FAP activity inhibitors [84,85]. Endothelial cells might transform into CAF-resembling cells, as described above. These cells often express VEGF and thus could be targeted with VEGF-antagonizing agents [51]. MSCs are another considerable source for CAFs. Although MSCs share specific markers with other cell types, some markers such as GD2 are considered unique identifiers of human MSCs [86]. Recently, it was reported that GD3S is highly expressed in GD2 positive stem cells and that GD3S inhibition reduces tumor formation in vivo [87]. Although the authors referred to cancer initiating stem cells in their report, these findings could also be valid for stromal MSCs. Finally, tumor cells resembling CAFs are the outcome of an EMT process and therefore would most likely respond to anticancer drugs targeting features such as a high mutation rate occurring in tumor suppressor genes (e.g., p53) and oncogenes (e.g., Myc). Should that be the case, the proper drugs to be administered are tumor suppressor reactivators or oncogene inhibitors. On a different note, several studies and clinical trials have used modified MSCs and CAFs for antitumor drug delivery with various degrees of success and safety [88,89]. It would be interesting to investigate whether the use of other ‘CAF state’ cells coming from different origins could yield improved results. By combining our knowledge regarding stromal cell markers and the recent advancements in stromal-based anticancer therapy, the personalized therapy concept could also be extended to the tumor microenvironment, thus providing a more comprehensive way to treat cancer. Acknowledgments

α

in GD hi 3 bi s to r

b Pa FA

F

GD 2, GD 3S

AP

References

Fibroblasts MSCs

CAFs

Endothelial cells VE αV E

GF

ab

GF

We wish to thank Professor Dov Zipori for inspiration and guidance. His ‘stem state’ concept has led to the birth of this review. We acknowledge and appreciate further work that was done in this field but could not be included owing to space limitations.

Tumor cells (EMT) c cifi pe n s ) s o or  c sor m ta My Tu mu 53, pres r, or (p sup ato ibit or v inh m ac e Tu re gen co on TRENDS in Molecular Medicine

Figure 1. Tailor-made therapy based on cancer associated fibroblast (CAF) cell of origin. CAF-resembling cells (inner circle) could be the descendants of several cell types (outer circle). These cells are identified by specific markers (purple trapezoids) and eventually might be targeted by anticancer drugs (pink trapezoids).

1 Franco, O.E. et al. (2010) Cancer associated fibroblasts in cancer pathogenesis. Semin. Cell Dev. Biol. 21, 33–39 2 Koontongkaew, S. (2013) The tumor microenvironment contribution to development, growth, invasion and metastasis of head and neck squamous cell carcinomas. J. Cancer 4, 66–83 3 Kalluri, R. and Zeisberg, M. (2006) Fibroblasts in cancer. Nat. Rev. Cancer 6, 392–401 4 Xing, F. et al. (2010) Cancer associated fibroblasts (CAFs) in tumor microenvironment. Front. Biosci. 15, 166–179 5 Buganim, Y. et al. (2011) Transcriptional activity of ATF3 in the stromal compartment of tumors promotes cancer progression. Carcinogenesis 32, 1749–1757 6 Madar, S. et al. (2009) Modulated expression of WFDC1 during carcinogenesis and cellular senescence. Carcinogenesis 30, 20–27 7 Rasmussen, A.A. and Cullen, K.J. (1998) Paracrine/autocrine regulation of breast cancer by the insulin-like growth factors. Breast Cancer Res. Treat. 47, 219–233 8 Liu, Y. et al. (2006) Separation, cultivation and biological characteristics of oral carcinoma-associated fibroblasts. Oral Dis. 12, 375–380 9 Commandeur, S. et al. (2011) Functional characterization of cancerassociated fibroblasts of human cutaneous squamous cell carcinoma. Exp. Dermatol. 20, 737–742 451

Opinion 10 Suetsugu, A. et al. (2011) Imaging the recruitment of cancerassociated fibroblasts by liver-metastatic colon cancer. J. Cell. Biochem. 112, 949–953 11 Musumeci, M. et al. (2011) Control of tumor and microenvironment cross-talk by miR-15a and miR-16 in prostate cancer. Oncogene 30, 4231–4242 12 Olumi, A.F. et al. (1999) Carcinoma-associated fibroblasts direct tumor progression of initiated human prostatic epithelium. Cancer Res. 59, 5002–5011 13 Noel, A. et al. (1993) Enhancement of tumorigenicity of human breast adenocarcinoma cells in nude mice by matrigel and fibroblasts. Br. J. Cancer 68, 909–915 14 Orimo, A. 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 15 Bhowmick, N.A. et al. (2004) TGF-b signaling in fibroblasts modulates the oncogenic potential of adjacent epithelia. Science 303, 848–851 16 Ao, M. et al. (2007) Cross-talk between paracrine-acting cytokine and chemokine pathways promotes malignancy in benign human prostatic epithelium. Cancer Res. 67, 4244–4253 17 Kuperwasser, C. et al. (2004) Reconstruction of functionally normal and malignant human breast tissues in mice. Proc. Natl. Acad. Sci. U.S.A. 101, 4966–4971 18 Eck, S.M. et al. (2009) CXCR4 and matrix metalloproteinase-1 are elevated in breast carcinoma-associated fibroblasts and in normal mammary fibroblasts exposed to factors secreted by breast cancer cells. Mol. Cancer Res. 7, 1033–1044 19 Boire, A. et al. (2005) PAR1 is a matrix metalloprotease-1 receptor that promotes invasion and tumorigenesis of breast cancer cells. Cell 120, 303–313 20 Crawford, Y. et al. (2009) PDGF-C mediates the angiogenic and tumorigenic properties of fibroblasts associated with tumors refractory to anti-VEGF treatment. Cancer Cell 15, 21–34 21 Straussman, R. et al. (2012) Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature 487, 500–504 22 Kraman, M. et al. (2010) Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-a. Science 330, 827–830 23 Olive, K.P. et al. (2009) Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 324, 1457–1461 24 Luga, V. et al. (2012) Exosomes mediate stromal mobilization of autocrine Wnt–PCP signaling in breast cancer cell migration. Cell 151, 1542–1556 25 Mueller, L. et al. (2007) Stromal fibroblasts in colorectal liver metastases originate from resident fibroblasts and generate an inflammatory microenvironment. Am. J. Pathol. 171, 1608–1618 26 Zipori, D. (2009) Biology of Stem Cells and the Molecular Basis of the Stem State. Springer 27 Song, S. et al. (2005) PDGFRb+ perivascular progenitor cells in tumours regulate pericyte differentiation and vascular survival. Nat. Cell Biol. 7, 870–879 28 Worthley, D.L. et al. (2009) Human gastrointestinal neoplasiaassociated myofibroblasts can develop from bone marrow-derived cells following allogeneic stem cell transplantation. Stem Cells 27, 1463–1468 29 Ishii, G. et al. (2003) Bone-marrow-derived myofibroblasts contribute to the cancer-induced stromal reaction. Biochem. Biophys. Res. Commun. 309, 232–240 30 Quante, M. et al. (2011) Bone marrow-derived myofibroblasts contribute to the mesenchymal stem cell niche and promote tumor growth. Cancer Cell 19, 257–272 31 Direkze, N.C. et al. (2004) Bone marrow contribution to tumorassociated myofibroblasts and fibroblasts. Cancer Res. 64, 8492–8495 32 Zhao, H. and Peehl, D.M. (2009) Tumor-promoting phenotype of CD90hi prostate cancer-associated fibroblasts. Prostate 69, 991–1000 33 Mishra, P.J. et al. (2008) Carcinoma-associated fibroblast-like differentiation of human mesenchymal stem cells. Cancer Res. 68, 4331–4339 34 Cunha, G.R. et al. (1992) Role of epithelial–mesenchymal interactions in the differentiation and spatial organization of visceral smooth muscle. Epithelial Cell Biol. 1, 76–83 452

Trends in Molecular Medicine August 2013, Vol. 19, No. 8

35 Cunha, G.R. et al. (2003) Role of the stromal microenvironment in carcinogenesis of the prostate. Int. J. Cancer 107, 1–10 36 Wikstrom, P. et al. (2009) Low stroma androgen receptor level in normal and tumor prostate tissue is related to poor outcome in prostate cancer patients. Prostate 69, 799–809 37 Schmitt-Gra¨ff, A. et al. (1994) Heterogeneity of myofibroblast phenotypic features: an example of fibroblastic cell plasticity. Virchows Arch. 425, 3–24 38 Barth, P.J. et al. (2002) CD34+ fibrocytes in invasive ductal carcinoma, ductal carcinoma in situ, and benign breast lesions. Virchows Arch. 440, 298–303 39 Petersen, O.W. et al. (2003) Epithelial to mesenchymal transition in human breast cancer can provide a nonmalignant stroma. Am. J. Pathol. 162, 391–402 40 Mink, S.R. et al. (2010) Cancer-associated fibroblasts derived from EGFR–TKI-resistant tumors reverse EGFR pathway inhibition by EGFR–TKIs. Mol. Cancer Res. 8, 809–820 41 Zeisberg, E.M. et al. (2007) Discovery of endothelial to mesenchymal transition as a source for carcinoma-associated fibroblasts. Cancer Res. 67, 10123–10128 42 Goritz, C. et al. (2011) A pericyte origin of spinal cord scar tissue. Science 333, 238–242 43 Dulauroy, S. et al. (2012) Lineage tracing and genetic ablation of ADAM12+ perivascular cells identify a major source of profibrotic cells during acute tissue injury. Nat. Med. http://dx.doi.org/10.1038/ nm.2848 44 Jotzu, C. et al. (2011) Adipose tissue derived stem cells differentiate into carcinoma-associated fibroblast-like cells under the influence of tumor derived factors. Cell. Oncol. (Dordr.) 34, 55–67 45 Baglole, C.J. et al. (2000) Functional assessment of fibroblast heterogeneity by the cell-surface glycoprotein Thy-1. In Madame Curie Biosci. Database [Internet]. Landes Bioscience, (Austin (TX)) Available from: http://www.ncbi.nlm.nih.gov/books/NBK6131/ 46 Mercier, I. et al. (2008) Human breast cancer-associated fibroblasts (CAFs) show caveolin-1 downregulation and RB tumor suppressor functional inactivation: implications for the response to hormonal therapy. Cancer Biol. Ther. 7, 1212–1225 47 Tlsty, T.D. (2001) Stromal cells can contribute oncogenic signals. Semin. Cancer Biol. 11, 97–104 48 Huang, M. et al. (2010) Breast cancer stromal fibroblasts promote the generation of CD44+CD24– cells through SDF-1/CXCR4 interaction. J. Exp. Clin. Cancer Res. 29, 80 49 Sugimoto, H. et al. (2006) Identification of fibroblast heterogeneity in the tumor microenvironment. Cancer Biol. Ther. 5, 1640–1646 50 Ayala, G. et al. (2003) Reactive stroma as a predictor of biochemical-free recurrence in prostate cancer. Clin. Cancer Res. 9, 4792–4801 51 Gonda, T.A. et al. (2010) Molecular biology of cancer-associated fibroblasts: can these cells be targeted in anti-cancer therapy? Semin. Cell Dev. Biol. 21, 2–10 52 Casey, T. et al. (2009) Molecular signatures suggest a major role for stromal cells in development of invasive breast cancer. Breast Cancer Res. Treat. 114, 47–62 53 Ma, X.J. et al. (2009) Gene expression profiling of the tumor microenvironment during breast cancer progression. Breast Cancer Res. 11, R7 54 Saadi, A. 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 55 Chang, H.Y. et al. (2005) Robustness, scalability, and integration of a wound-response gene expression signature in predicting breast cancer survival. Proc. Natl. Acad. Sci. U.S.A. 102, 3738–3743 56 Finak, G. et al. (2008) Stromal gene expression predicts clinical outcome in breast cancer. Nat. Med. 14, 518–527 57 Pathak, S. et al. (1997) Can cancer cells transform normal host cells into malignant cells? Br. J. Cancer 76, 1134–1138 58 Macintosh, C.A. et al. (1998) Precise microdissection of human prostate cancers reveals genotypic heterogeneity. Cancer Res. 58, 23–28 59 Wernert, N. et al. (2001) Presence of genetic alterations in microdissected stroma of human colon and breast cancers. Anticancer Res. 21, 2259–2264

Opinion 60 Moinfar, F. et al. (2000) Concurrent and independent genetic alterations in the stromal and epithelial cells of mammary carcinoma: implications for tumorigenesis. Cancer Res. 60, 2562–2566 61 Kurose, K. et al. (2002) Frequent somatic mutations in PTEN and TP53 are mutually exclusive in the stroma of breast carcinomas. Nat. Genet. 32, 355–357 62 Patocs, A. et al. (2007) Breast-cancer stromal cells with TP53 mutations and nodal metastases. N. Engl. J. Med. 357, 2543–2551 63 Walter, K. et al. (2008) Pancreatic cancer associated fibroblasts display normal allelotypes. Cancer Biol. Ther. 7, 882–888 64 Allinen, M. et al. (2004) Molecular characterization of the tumor microenvironment in breast cancer. Cancer Cell 6, 17–32 65 Hosein, A.N. et al. (2010) Breast carcinoma-associated fibroblasts rarely contain p53 mutations or chromosomal aberrations. Cancer Res. 70, 5770–5777 66 Akahane, T. et al. (2013) The origin of stroma surrounding epithelial ovarian cancer cells. Int. J. Gynecol. Pathol. 32, 26–30 67 Tuhkanen, H. et al. (2006) Frequent gene dosage alterations in stromal cells of epithelial ovarian carcinomas. Int. J. Cancer 119, 1345–1353 68 Geyer, F.C. et al. (2010) Genomic and immunohistochemical analysis of adenosquamous carcinoma of the breast. Mod. Pathol. 23, 951–960 69 Fukino, K. et al. (2007) Genomic instability within tumor stroma and clinicopathological characteristics of sporadic primary invasive breast carcinoma. JAMA 297, 2103–2111 70 Zong, Y. et al. (2012) Stromal epigenetic dysregulation is sufficient to initiate mouse prostate cancer via paracrine Wnt signaling. Proc. Natl. Acad. Sci. U.S.A. 109, E3395–E3404 71 Fu, L. et al. (2011) Wnt2 secreted by tumour fibroblasts promotes tumour progression in oesophageal cancer by activation of the Wnt/bcatenin signalling pathway. Gut 60, 1635–1643 72 Rudnick, J.A. and Kuperwasser, C. (2012) Stromal biomarkers in breast cancer development and progression. Clin. Exp. Metastasis 29, 663–672 73 De Monte, L. et al. (2011) Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer. J. Exp. Med. 208, 469–478 74 Erez, N. et al. (2010) Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kB-dependent manner. Cancer Cell 17, 135–147 75 Kogan-Sakin, I. et al. (2009) Prostate stromal cells produce CXCL-1, CXCL-2, CXCL-3 and IL-8 in response to epithelia-secreted IL-1. Carcinogenesis 30, 698–705 76 Tjomsland, V. et al. (2011) Interleukin 1a sustains the expression of inflammatory factors in human pancreatic cancer microenvironment by targeting cancer-associated fibroblasts. Neoplasia 13, 664–675 77 Barone, I. et al. (2012) Leptin mediates tumor–stromal interactions that promote the invasive growth of breast cancer cells. Cancer Res. 72, 1416–1427 78 Madar, S. et al. (2013) Mutant p53 attenuates the anti-tumorigenic activity of fibroblasts-secreted interferon b. PLoS ONE 8, e61353 79 Dawkins, R. (1982) The Extended Phenotype. Oxford University Press 80 Vernitsky, H. et al. (2012) Ras oncoproteins transfer from melanoma cells to T cells and modulate their effector functions. J. Immunol. 189, 4361–4370

Trends in Molecular Medicine August 2013, Vol. 19, No. 8

81 Hua, X. et al. (2011) Expression and role of fibroblast activation protein-a in microinvasive breast carcinoma. Diagn. Pathol. 6, 111 82 Samel, D. et al. (2003) Generation of a FasL-based proapoptotic fusion protein devoid of systemic toxicity due to cell-surface antigenrestricted activation. J. Biol. Chem. 278, 32077–32082 83 Scott, A.M. et al. (2003) A Phase I dose-escalation study of sibrotuzumab in patients with advanced or metastatic fibroblast activation protein-positive cancer. Clin. Cancer Res. 9, 1639–1647 84 Ryabtsova, O. et al. (2012) Acylated Gly-(2-cyano)pyrrolidines as inhibitors of fibroblast activation protein (FAP) and the issue of FAP/prolyl oligopeptidase (PREP)-selectivity. Bioorg. Med. Chem. Lett. 22, 3412–3417 85 Adams, S. et al. (2004) PT-100, a small molecule dipeptidyl peptidase inhibitor, has potent antitumor effects and augments antibodymediated cytotoxicity via a novel immune mechanism. Cancer Res. 64, 5471–5480 86 Martinez, C. et al. (2007) Human bone marrow mesenchymal stromal cells express the neural ganglioside GD2: a novel surface marker for the identification of MSCs. Blood 109, 4245–4248 87 Battula, V.L. et al. (2012) Ganglioside GD2 identifies breast cancer stem cells and promotes tumorigenesis. J. Clin. Invest. 122, 2066–2078 88 Greco, S.J. and Rameshwar, P. (2012) Mesenchymal stem cells in drug/ gene delivery: implications for cell therapy. Ther. Deliv. 3, 997–1004 89 Al-Zoubi, M. et al. (2013) Creating a tumor-resistant microenvironment: cell-mediated delivery of TNFa completely prevents breast cancer tumor formation in vivo. Cell Cycle 12, 480–490 90 Olaso, E. et al. (1997) Tumor-dependent activation of rodent hepatic stellate cells during experimental melanoma metastasis. Hepatology 26, 634–642 91 Vered, M. et al. (2010) Cancer-associated fibroblasts and epithelial– mesenchymal transition in metastatic oral tongue squamous cell carcinoma. Int. J. Cancer 127, 1356–1362 92 Cornil, I. et al. (1991) Fibroblast cell interactions with human melanoma cells affect tumor cell growth as a function of tumor progression. Proc. Natl. Acad. Sci. U.S.A. 88, 6028–6032 93 Malanchi, I. et al. (2012) Interactions between cancer stem cells and their niche govern metastatic colonization. Nature 481, 85–89 94 Elkabets, M. et al. (2011) Human tumors instigate granulinexpressing hematopoietic cells that promote malignancy by activating stromal fibroblasts in mice. J. Clin. Invest. 121, 784–799 95 Calon, A. et al. (2012) Dependency of colorectal cancer on a TGF-bdriven program in stromal cells for metastasis initiation. Cancer Cell 22, 571–584 96 Gaggioli, C. (2008) Collective invasion of carcinoma cells: when the fibroblasts take the lead. Cell Adh. Migr. 2, 45–47 97 Raman, D. et al. (2007) Role of chemokines in tumor growth. Cancer Lett. 256, 137–165 98 Fitzpatrick, F.A. (2001) Inflammation, carcinogenesis and cancer. Int. Immunopharmacol. 1, 1651–1667 99 Rasanen, K. et al. (2009) Differences in the nemosis response of normal and cancer-associated fibroblasts from patients with oral squamous cell carcinoma. PLoS ONE 4, e6879 100 Mueller, L. et al. (2010) TNF-a similarly induces IL-6 and MCP-1 in fibroblasts from colorectal liver metastases and normal liver fibroblasts. Biochem. Biophys. Res. Commun. 397, 586–591

453

Journal of Visualized Experiments

www.jove.com

Video Article

Experimental Generation of Carcinoma-Associated Fibroblasts (CAFs) from Human Mammary Fibroblasts Urszula M. Polanska1,* , Ahmet Acar1,* , Akira Orimo1,2 1CR-UK 2Atopy

Stromal-Tumour Interaction Group, Paterson Institute for Cancer Research, University of Manchester Research Center, Juntendo University

*These authors contributed equally

Correspondence to: Akira Orimo at [email protected] URL: http://www.jove.com/details.php?id=3201 DOI: 10.3791/3201 Keywords: Medicine, Issue 56, cancer, stromal myofibroblasts, experimentally generated carcinoma-associated fibroblasts (exp-CAFs), fibroblast, human mammary carcinomas, tumour xenografts, Date Published: 10/25/2011 Citation: Polanska, U.M., Acar, A., Orimo, A. Experimental Generation of Carcinoma-Associated Fibroblasts (CAFs) from Human Mammary Fibroblasts. J. Vis. Exp. (56), e3201, DOI : 10.3791/3201 (2011).

Abstract Carcinomas are complex tissues comprised of neoplastic cells and a non-cancerous compartment referred to as the 'stroma'. The stroma consists of extracellular matrix (ECM) and a variety of mesenchymal cells, including fibroblasts, myofibroblasts, endothelial cells, pericytes and leukocytes 1-3. The tumour-associated stroma is responsive to substantial paracrine signals released by neighbouring carcinoma cells. During the disease process, the stroma often becomes populated by carcinoma-associated fibroblasts (CAFs) including large numbers of myofibroblasts. These cells have previously been extracted from many different types of human carcinomas for their in vitro culture. A subpopulation of CAFs is distinguishable through their up-regulation of α-smooth muscle actin (α-SMA) expression4,5. These cells are a hallmark of 'activated fibroblasts' that share similar properties with myofibroblasts commonly observed in injured and fibrotic tissues 6. The presence of this myofibroblastic CAF subset is highly related to high-grade malignancies and associated with poor prognoses in patients. Many laboratories, including our own, have shown that CAFs, when injected with carcinoma cells into immunodeficient mice, are capable of substantially promoting tumourigenesis 7-10. CAFs prepared from carcinoma patients, however, frequently undergo senescence during propagation in culture limiting the extensiveness of their use throughout ongoing experimentation. To overcome this difficulty, we developed a novel technique to experimentally generate immortalised human mammary CAF cell lines (exp-CAFs) from human mammary fibroblasts, using a coimplantation breast tumour xenograft model. In order to generate exp-CAFs, parental human mammary fibroblasts, obtained from the reduction mammoplasty tissue, were first immortalised with hTERT, the catalytic subunit of the telomerase holoenzyme, and engineered to express GFP and a puromycin resistance gene. These cells were coimplanted with MCF-7 human breast carcinoma cells expressing an activated ras oncogene (MCF-7-ras cells) into a mouse xenograft. After a period of incubation in vivo, the initially injected human mammary fibroblasts were extracted from the tumour xenografts on the basis of their puromycin resistance 11. We observed that the resident human mammary fibroblasts have differentiated, adopting a myofibroblastic phenotype and acquired tumour-promoting properties during the course of tumour progression. Importantly, these cells, defined as exp-CAFs, closely mimic the tumour-promoting myofibroblastic phenotype of CAFs isolated from breast carcinomas dissected from patients. Our tumour xenograft-derived exp-CAFs therefore provide an effective model to study the biology of CAFs in human breast carcinomas. The described protocol may also be extended for generating and characterising various CAF populations derived from other types of human carcinomas.

Video Link The video component of this article can be found at http://www.jove.com/details.php?id=3201

Protocol

1. Isolation of primary cultured human normal mammary fibroblasts Experimental procedures for isolating primary cultured human normal mammary fibroblasts are outlined in Fig. 1A. 1. Prepare the cell dissociation buffer, as described previously12: Dulbecco's Modified Eagle's Medium (DMEM) with 10% fetal calf serum (FCS), penicillin-streptomycin (200 units/ml), collagenase type I (1 mg/ml) and hyaluronidase (125 units/ml). 2. Wash the breast tissue dissected from a reduction mammoplasty (~0.5 gram) several times in phosphate buffered saline (PBS). Mince the tissue into small fragments (