Skip to main content
Erschienen in: BMC Cancer 1/2015

Open Access 01.12.2015 | Research article

Il-6 signaling between ductal carcinoma in situ cells and carcinoma-associated fibroblasts mediates tumor cell growth and migration

verfasst von: Kingsley O. Osuala, Mansoureh Sameni, Seema Shah, Neha Aggarwal, Michelle L. Simonait, Omar E. Franco, Yan Hong, Simon W. Hayward, Fariba Behbod, Raymond R. Mattingly, Bonnie F. Sloane

Erschienen in: BMC Cancer | Ausgabe 1/2015

Abstract

Background

Ductal carcinoma in situ (DCIS) is a non-obligate precursor lesion of invasive breast cancer in which approximately half the patients will progress to invasive cancer. Gaining a better understanding of DCIS progression may reduce overtreatment of patients. Expression of the pro-inflammatory cytokine interleukin-6 increases with pathological stage and grade, and is associated with poorer prognosis in breast cancer patients. Carcinoma associated fibroblasts (CAFs), which are present in the stroma of DCIS patients are known to secrete pro-inflammatory cytokines and promote tumor progression.

Methods

We hypothesized that IL-6 paracrine signaling between DCIS cells and CAFs mediates DCIS proliferation and migration. To test this hypothesis, we utilized the mammary architecture and microenvironment engineering or MAME model to study the interactions between human breast CAFs and human DCIS cells in 3D over time. We specifically inhibited autocrine and paracrine IL-6 signaling to determine its contribution to early stage tumor progression.

Results

Here, DCIS cells formed multicellular structures that exhibited increased proliferation and migration when cultured with CAFs. Treatment with an IL-6 neutralizing antibody inhibited growth and migration of the multicellular structures. Moreover, selective knockdown of IL-6 in CAFs, but not in DCIS cells, abrogated the migratory phenotype.

Conclusion

Our results suggest that paracrine IL-6 signaling between preinvasive DCIS cells and stromal CAFs represent an important factor in the initiation of DCIS progression to invasive breast carcinoma.
Begleitmaterial
Additional file 4: Figure S2. Oxymatrine treatment. (A-B) MCF10.DCIS cells were grown in the presence of DMSO (A) or 1 mg/ml oxymatrine (B). Oxymatrine or DMSO was added 1 day after initial seeding. Scale bar, 200 μm. MCF10.DCIS cells grown in DMSO form large structures similar to untreated cultures (cf. Fig. 2a). (C) Measurement of structure diameters in three tiled images from three independent experiments. (D) Cell viability was evaluated using an ATP-based luminescence assay 24 h after drug treatment. Arbitrary units (A.U.). **P < 0.01, not significant (NS). Student’s t-test; mean ± SD. (TIFF 141 kb)
Additional file 12: Video S4. Video clip shows the transition through optical slices of a MCF10.DCIS structure in co-culture with CAF40TKi fibroblasts (Full 3D image shown in Fig. 4e and f). Cultures were fixed and immunostained for IL-6 (red), with DAPI labeled nuclei. MCF10.DCIS cells formed large nodular-like structures that branched out towards CFSE-labeled CAFs (pseudo-colored white). Images were obtained on a Zeiss LSM 780 and converted to a movie file using Volocity® software. (MOV 1 mb)
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12885-015-1576-3) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

KOO and BFS designed research plan; KOO made substantial contributions to conception, acquisition, analysis and interpretation of data; MS conducted immunohistochemistry and made substantial contribution to data analysis; NA, and SS made substantial contributions to data analysis; MLS performed video editing and contributed to data analysis; KOO, BFS, RRM, SWH, OEF, YH, and FB drafted the manuscript and participated in revising it for intellectual content. All authors read and approved the final manuscript.
Abkürzungen
DCIS
Ductal carcinoma in situ
CAFs
Carcinoma-associated fibroblasts
IL-6
Interleukin 6
IL-6TS
Interleukin 6– trans-signaling
MAME
Mammary architecture and microenvironment engineering
CFSE
Carboxyfluorescein diacetate succinimidyl ester
dDQ-col
Degraded dye-quenched collagen
3D
Three-dimensional
EMT
Epithelial to mesenchymal transition
ELISA
Enzyme-linked immunosorbent assay
qRT-PCR
Quantative Real-time polymerase chain reaction
shRNA
Short hairpin ribonucleic acid
LSM
Laser scanning microscope
DIC
Differential interference contrast
Oxy
Oxymatrine
DMSO
Dimethyl sulfoxide
DNA
Deoxyribonucleic acid
FDA
Federal Drug Administration

Background

Ductal carcinoma in situ (DCIS) of the breast is a pre-invasive lesion and a risk factor for subsequent invasive ductal carcinoma (IDC) [1]. DCIS represents about 20 % of newly diagnosed breast cancers in the United States [2]. If left untreated, approximately half of DCIS tumors will progress to IDC while half will remain indolent [3, 4]. Although there are many subtypes of DCIS, it is not currently possible to identify which will progress. This has led to aggressive treatments, specifically radiation with either lumpectomy or mastectomy [5].
Components of the tumor microenvironment are increasingly implicated in the progression of many cancers. Early morphological and physiological changes in breast epithelium are minimal, and compounding factors such as tumor-suppressive paracrine signaling from myoepithelial cells [6] or the extracellular matrix [7] may hide early indicators of ductal cell aberration. Such changes may include, but are not limited to: gene expression modulation, epigenetic alterations, and loss of genomic stability in both the epithelial and stromal compartments. In the tumor microenvironment, carcinoma-associated fibroblasts (CAFs) represent a fibroblast population or mixture of sub-populations that can promote tumor progression [813]. Although this mechanism is not fully understood, it is known that CAFs secrete numerous cytokines and growth factors [14].
Interleukin 6 (IL-6) is a pro-inflammatory cytokine shown to alter cell morphology, modulate cell migration and the epithelial to mesenchymal transition [1517]. Many of these processes occur upon IL-6 activation of the transmembrane IL-6 receptor (IL-6R), which heterodimerizes with the ubiquitously expressed cell surface receptor glycoprotein 130 (gp130). Downstream activation of the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway initiates IL-6 target gene transcription [18, 19]. Alternatively, secreted IL-6 can bind to the soluble IL-6 receptor (sIL-6R), which then binds gp130 at the cell surface and initiates intracellular signaling. This form of IL-6 signaling has been coined “IL-6 trans-signaling” (IL-6TS) [20]. IL-6 has been linked to the upregulation of proteases such as cysteine cathepsins and matrix metalloproteinases that are known to play a role in cancer progression [21, 22]. The IL-6 signaling axis is commonly upregulated in invasive cancers, suggesting that IL-6 may be an important mediator of events involved in tumor cell invasion [2325].
The effects of IL-6 signaling inhibitors on breast cancer cell morphology and proliferation have been evaluated in monotypic cultures. Two studies have shown that inhibiting autocrine IL-6 signaling in either triple negative breast cancer cell lines [26], or the ER-positive MCF7 cell line [27] significantly inhibited cell growth. Additionally, Leslie et al. show that knockdown of IL-6 in an invasive variant of the MCF10A cells, MCF10A-H-RasV12, inhibited cell migration in a transwell assay, and inhibited growth in a xenograft mouse model [28]. Although these studies evaluated paracrine signaling, cells were treated with exogenous recombinant protein rather than co-culturing different cell types. Therefore the authors were unable to evaluate in 3D the dynamic cell:cell interactions between two separate human cell types or the cell:microenvironment interactions.
Our 3D mammary architecture and microenvironment engineering (MAME) culture model mimics in vivo architecture, providing a suitable setting to study cell:cell interactions and, notably, physiologically relevant cell:cell signaling over time. Additionally, 3D in vitro cell culture models better represent in vivo tumor drug response, which would facilitate efficacious therapy development at the preclinical stage [29]. Here we examine the role of IL-6 in progression of pre-invasive breast DCIS to an invasive phenotype, and show how co-culture of DCIS cells with CAFs changes DCIS growth and invasive potential.

Methods

Cell lines

MCF10A human breast non-transformed epithelial cells, MCF10.DCIS and SUM102 human breast DCIS cells, and WS-12Ti human breast carcinoma-associated fibroblasts were provided by Dr. Bonnie F. Sloane. All primary fibroblasts were derived from human breast tissue. CAF40T were derived from biopsy tissue diagnosed as invasive carcinoma. NAF98 were derived from benign tissue. Both CAF40T and NAF98 fibroblast cell lines were provided by Dr. Simon W. Hayward. These fibroblasts were immortalized in Dr. Sloane’s lab and are designated CAF40TKi and NAF98i, respectively. The FB-NF, NAF-FB, and FB-CAF primary fibroblasts were derived from patient biopsies diagnosed as: benign (FB-NF, NAF-FB), or invasive carcinoma with accompanying DCIS (FB-CAF) and provided by Dr. Fariba Behbod. The FB-NF fibroblasts were immortalized in Dr. Sloane’s lab and designated FB-NF-i. The FB-CAF and NAF-FB fibroblasts were not immortalized. All patient derived cells were received de-identified and therefore are exempt from IRB oversight.

Cell culture

In this study we utilized non-tumor forming MCF10A human breast epithelial cells [30] and the human DCIS cell lines MCF10.DCIS and SUM102, which were maintained as previously described [31]. See supplemental methods for more detailed information (Additional file 1). All 3D MAME cultures were performed using Cultrex (3433-005-01, Trevigen, Gaithersburg, MD) similar to previously described [32]. Briefly, cell culture dishes were coated with 100 % Cultrex. Cells were added on top of solidified Cultrex and allowed to adhere for 30–45 min before being overlaid with 2 % Cultrex in phenol red-free DMEM F12 media containing 2.5 % fetal bovine serum (Additional file 2: Figure S1). In co-culture experiments, fibroblasts were added first and allowed to adhere before adding tumor cells. Once tumor cells had adhered to the matrix, an overlay of 2 % Cultrex was added. Media were changed every 4 days.

Measurement of multicellular structures

Differential interference contrast (DIC) images of three random fields at 20X magnification were used to measure multicellular structures. Three individuals of whom two were study-blinded measured the diameter and perimeter of structures and the number and length of interconnections between structures. All three data sets were used in the quantification. This analysis was performed using Zen imaging software (Zeiss, Thornwood, NY). Volume measurements were obtained using 3D fluorescent images and quantified using Volocity software (Perkin-Elmer, Waltham, Mass).

Gene expression

RNA was isolated from cells grown in either 2D monolayer or 3D MAME cultures. For 2D culture, cells were washed in phosphate buffered saline and harvested using 0.5 % Trypsin/EDTA (Life Technologies, Foster City, CA) and pelleted. The cell pellets were resuspended in TRIzol® Reagent (Life Technologies, Foster City, CA) for RNA extraction. All qRT-PCR reactions were performed using Taqman Assays (Life Technologies, Foster City, CA). See supplemental list of Taqman Assays (Additional file 3: Table S1).

ELISA

ELISA kits (human IL6R-ab46029, human IL6-ab46044, and human GAPDH-ab119627) were purchased from Abcam® (San Francisco, CA). Aliquots of lysates were collected for ELISA assays and measurement of total DNA.

Immunohistochemistry

A breast tissue microarray (BR8011) was purchased from US Biomax® (Rockville, MD). Dr. Fariba Behbod provided patient tissue microarrays, and biopsy sections were purchased from ProSci Incorporated (cat # 10–010 and 10–003, Poway, CA). The thickness of all tissue sections immunostained were 10-microns.

Immunofluorescence

Nuclei were labeled with Hoechst (33342, Thermo Scientific) or EDU (Life Technologies, Foster City, CA). Polyclonal antibodies to human IL-6 (AF-206-NA, R&D Systems, Minneapolis, MN) were used at a concentration of 1 μg/ml. Mono-specific antibodies to human cathepsin B have been previously isolated and characterized [33]. Cathepsin B immunostaining was performed as previously described with the exception that 1 % Tween 20 replaced the 0.01 % saponin [34]. For some studies, CAF40TKi were pre-labeled, prior to seeding in MAME co-cultures, utilizing CellTrace CFSE (carboxyfluorescein diacetate succinimidyl ester; Life Technologies, Foster City, CA) according to the manufacturer’s protocol.

Drug treatments

For treatment of MAME cultures with IL-6 nAb (R&D Systems, AF-206-NA), we added 1 μg/ml of IL-6 nAb in the 2 % Cultrex overlay to 3D cultures on the first day of culture and refreshed with IL-6 nAb and 2 % overlay every 4 days. The antibody concentration was selected based on preliminary studies in which we determined the lowest concentration needed to inhibit growth of tumor structures. Oxymatrine (Sigma, St. Louis, MO) at 1-mg/ml (3.7 mM) was added 24 hours after cell seeding and was replaced with fresh drug every 4 days. The oxymatrine concentration was determined empirically based on the concentration at which proliferation was inhibited to 50 % of control. The protease inhibitors CA074Me and E64d (Sigma, St. Louis, MO) were used at a concentration of 10 μM [35].

Live cell proteolysis assay

Dye-quenched collagen IV (DQ-collagen IV, Life Technologies, Foster City, CA) was used admixed in Cultrex as previously described [32]. MAME cultures in optical glass bottom cell culture dishes were imaged live for a period of 10 to 60 min under 5 % CO2 at 37 °C.

Confocal microscopy

Confocal microscopy was performed on either a Zeiss LSM 510 or LSM 780 upright confocal microscope (Zeiss, Thornwood, NY). All cell cultures used for imaging were seeded on 40 mm optical glass bottom culture dishes at a density of 45 cells/mm2 (~5000 cells/dish).

Statistics

Data were statistically analyzed using Student’s t-test on GraphPad Prism 6.0 (GraphPad Software Inc., La Jolla, CA).

Ethics statement

All human subject materials and experiments in this study have been reviewed by the Wayne State University Institutional Review Board and deemed exempt according to the definition codified in the common rule at 45 CFR 46.102(d)(f).

Results

Human breast DCIS cells express pro-inflammatory cytokines

Increased levels of pro-inflammatory cytokines, including IL-6, in tumors and serum of breast cancer patients correlate with poor prognosis [26, 3638]. Immunohistochemistry on breast tissue from female patients, age 30 to 81 with an average age of 49.5, confirmed IL-6 protein expression in 65 % of patient samples diagnosed with DCIS (Fig. 1a and b, cf. 1c and d).
We utilized a 3D MAME culture model (Additional file 2: Figure S1), to study the role of IL-6 signaling between human breast DCIS cells and human breast CAFs. The MAME model allows for co-culture of multiple cell types in the context of a three dimensional microenvironment. Our model is advantageous over commercially available 3D platforms as it can be utilized for live cell microscopy, and imaging live cell functional assays (such as proteolysis) [32]. MAME cultures are tractable and can be altered to grow multiple cell types at various ratios or at various relative positions within the matrix. Additionally, the reduced density overlay allows for real-time collection and analysis of cytokine secretion without disrupting the longitudinal growth of the culture.
Here we examined the expression of IL-6 and the associated pro-inflammatory genes interleukin 1β (IL1β) and nuclear factor kappa light chain enhancer of activated B cells 1 (NFκB1) in human breast MCF10.DCIS and SUM102 cell lines. Our data show higher IL-6 mRNA levels in both MCF10.DCIS and SUM102 cell lines, as compared to the MCF10A breast epithelial cell line (Fig. 1e). Levels of IL-6 protein in lysates of the three cell lines were below the level of detection. When we measured secreted IL-6 in conditioned media from MCF10.DCIS or SUM102 cultures, we found that both DCIS cell lines secreted 10 to 30-fold more IL-6 than the MCF10A cells (Fig. 1f).

Blocking IL-6 autocrine signaling inhibits MCF10.DCIS growth

To test whether blocking IL-6 autocrine signaling affected DCIS cell growth, we treated MCF10.DCIS MAME cultures for 8 days with either an IL-6 neutralizing antibody (nAb) or an equivalent concentration of a species and isotype matched antibody (Fig. 2). IL-6 nAb treatment resulted in a reduction in diameter of the multicellular 3D structures (Fig. 2b, cf. 2a, quantified in 2d). This was reversible as replacement of media on day 8, with media lacking IL-6 nAb, resulted in a significant increase in diameter of the MCF10.DCIS structures after an additional 48 h (Fig. 2c, cf. 2b, quantified in 2d).
To determine the effects of IL-6 nAb on MCF10.DCIS structures, we examined the expression of a panel of candidate genes that have been associated with tumor growth and invasion [3943]. The expression of IL-6 was upregulated 2-fold in the treated cultures. The expression of TWIST1, vimentin, and Fibroblast Growth Factor 2 was downregulated greater than 2-fold, while minimal changes were observed in the expression of E-cadherin, N-cadherin, and NFκB1 (Fig. 2e). To test whether pharmacological suppression of IL-6 could reproduce IL-6 nAb mediated growth inhibition, we treated cells with oxymatrine, a naturally occurring inhibitor of IL-6 gene expression. Oxymatrine has been shown to prevent nuclear translocation of NFκB-p65 thereby inhibiting transcriptional activation of its target genes, which include IL-6 [44]. Oxymatrine treatment was able to replicate the growth inhibitory effects observed with IL-6 nAb (Additional file 4: Figure S2B, cf. S2A, quantified in S2C). Neither oxymatrine nor IL-6 nAb treatment resulted in marked cell death as cytotoxicity assays showed no difference in cell viability after 48-hour drug treatment (Additional file 4: Figure S2D).

Carcinoma-associated fibroblasts express IL-6 and promote DCIS cell proliferation and motility

CAFs represent a population or group of populations of stromal cells that can promote tumor cell growth [14, 4547]. The mechanism of supported tumor growth is likely through stromal-epithelial paracrine signaling. Therefore, we next evaluated human breast CAFs to determine their contribution of IL-6 in the tumor microenvironment. Additionally, we examined the role that CAFs play in MCF10.DCIS cell proliferation and motility in the 3D MAME model.
We examined the expression of IL-6 mRNA in normal human fibroblasts and CAFs grown in 3D. Here we found that CAFs exhibited elevated expression of IL-6 mRNA compared to normal fibroblasts (Fig. 3a). Protein levels of IL-6 in FB-NF-i normal fibroblast lysates were near the lower limit of detection and undetectable in NAF-FB or NAF98i lysates. IL-6 levels in CAF40TKi lysates were significantly higher than in FB-NF-i lysates (Fig. 3b). Levels of IL-6 in CAF-conditioned media were higher than in normal fibroblast-conditioned media (Fig. 3c).
We next co-cultured MCF10.DCIS cells with CAFs in 3D, at a seeding ratio of five tumor cells to one CAF [32], to evaluate the effect of CAF-secreted cytokines on DCIS cell proliferation and related changes in morphology of multicellular structures. We found that in MCF10.DCIS:CAF40TKi co-cultures there was an increase in the average diameter and volume of the multicellular structures and a prominent formation of branch-like interconnections between the structures (Fig. 3e, cf. 3d, quantified in Additional file 5: Figure S3A). Fluorescent imaging of MCF10.DCIS:CAF40TKi 3D co-cultures revealed that the branch-like multicellular connections between structures were primarily composed of tumor cells, yet contained some CAFs (Additional file 5: Figure S3B and Additional file 6: Video S1).
We also observed that CAFs induced an increased rate of proliferation in MCF10.DCIS cells. Using a thymidine analog to evaluate the rate of DNA synthesis, we observed that co-cultures had a consistently higher rate of DNA synthesis than CAFs alone or DCIS cells alone (Additional file 7: Figure S4A-D). CAF and DCIS co-culture using a slower growing DCIS cell line, i.e., SUM102, also resulted in changes in multicellular structure formation. We observed the presence of multiple invasive processes in SUM102:CAF co-cultures that were completely absent in cultures of the SUM102 cells alone (Additional file 8: Figure S5C and D, arrow, cf. S5A and B).

DCIS cells migrate with CAFs at the invasive front

CAFs express and secrete a number of proteases, which enhance their ability to migrate and remodel extracellular matrices [4851]. In CAF:MCF10.DCIS co-cultures, we evaluated cell:cell interactions and motility. Here we found that MCF10.DCIS spheroids formed attachments to CAFs and remained associated with them throughout an 8-day culture. On day 1 (24 h after seeding) of a co-culture we observed many single DCIS cells and a few CSFE labeled CAFs (Fig. 4a). After 3 days in culture DCIS cells had formed small spheroids that were in contact with one or more CAFs (Fig. 4b). On day 5 we observed CAFs at the invasive edges of DCIS tumor spheroids. A representative image shows a CAF (Fig. 4c, arrow) in contact with a DCIS spheroid. A time-lapse video shows the CAF leading the DCIS spheroid (Additional file 9: Video S2). High magnification imaging shows heterocellular contact between a tumor structure and a single CAF (Additional file 10: Figure S6). We did not observe these invasive characteristics in DCIS cell grown alone, as the tumor structures tended to roll in the extracellular matrix (Additional file 11: Video S3). By day 7 CAFs were seen at the invasive edge of most tumor structures (Fig. 4d).
Using confocal microscopy we examined the 3D spatial organization of the DCIS cells in relation to CAFs. A top view of a 3D reconstruction shows an MCF10.DCIS structure (only nuclei labeled) branching out toward CFSE labeled CAFs (pseudo-colored white) (Fig. 4e, Additional file 12: Video S4). The core of this structure (dashed circle) formed shortly after seeding and grew in size before the two branching outgrowths formed. These outgrowths are reminiscent of the “strand” multicellular migration pattern previously described [52]. A side view of the same structure shows that DCIS cells migrated downward from the core structure toward the CAFs (Fig. 4f).
IL-6 protein expression in these co-cultures was detected using immunofluorescent staining (Additional file 13: Figure S7). A CFSE-labeled CAF showed high expression of IL-6, whereas the leading edge of the DCIS multicellular structure showed a gradient of IL-6 that was strongest near the attachment to the CAF (Additional file 13: Figure S7A, arrow). We also showed that MCF10.DCIS cells that do not migrate to serum-free media (Additional file 13: Figure S7D) migrated toward CAF-conditioned serum-free media (Additional file 13: Figure S7E, S7F).

CAFs and MCF10.DCIS cells utilize cathepsin B to degrade matrix

IL-6 has previously been shown to upregulate cathepsin B [21], a protease associated with breast cancer progression. Therefore, we examined the expression and localization of cathepsin B in relation to active proteolysis in our 3D MAME co-cultures. Our active proteolysis assays were performed during live cell imaging [33]. Fluorescent imaging of live cells in MAME co-culture revealed regions of proteolytic activity (green) around several multicellular structures (Fig. 5a). The cells were then fixed for cathepsin B immunofluorescent staining. Visualizing the same structures revealed cathepsin B expression at the interface between the multicellular structures and the matrix (Fig. 5b). Fluorescent overlay shows co-localization between DQ-collagen IV degradation and areas with high cathepsin B expression (Fig. 5c); however, matrix degradation also occurred in areas that did not stain for cathepsin B (arrow). Upon visualization using light microscopy, the cells in these areas had morphological features consistent with CAFs, which would be consistent with current knowledge of fibroblast migration and their secretion of many proteases in addition to cathepsin B [53, 54].
We then used protease inhibitors to determine the contribution of all cysteine proteases and/or cathepsin B and L, to the overall proteolysis in the MAME culture. We treated cultures with E64d, a cell permeable pan-cysteine protease inhibitor, and found a 30 – 40 % reduction of matrix degradation (Fig. 5d). We also treated cultures with the cell permeable cathepsin B/L inhibitor, CA074Me, and found a 60 – 70 % reduction in matrix degradation (Fig. 5e). CA074Me has previously been shown to be a more efficacious inhibitor against cathepsins B and L than is E64d [35, 55]. We did not observe complete inhibition of proteolysis with the cysteine protease inhibitors, a finding in accord with previous findings that DCIS cells and CAFs secrete many non-cysteine proteases [31, 56, 57].

Blocking IL-6 inhibits CAF-stimulated effects on human breast DCIS cells

We have shown that CAFs promote the proliferation and migration of human breast DCIS cells. To determine whether CAF-stimulated migration and interaction with MCF10.DCIS cells could be inhibited by blocking IL-6, we co-cultured MCF10.DCIS cells with two human breast carcinoma-associated fibroblast lines WS-12Ti or CAF40TKi, in the presence of IL-6 nAb or an isotype control antibody. IL-6 nAb reduced the size of MCF10.DCIS:CAF multicellular structures when grown with either CAF cell line (Fig. 6a-e). Multicellular structure volume measurements confirmed a significant reduction when treated with IL-6 nAb (Fig. 6f-h).
To confirm the role of IL-6 as a key player in the formation of the multicellular structure interconnections, we utilized shRNA targeting IL-6 expression. In CAF40TKi and MCF10.DCIS cells, we achieved greater than 50 % reduction in secreted IL-6 (Additional file 14: Figure S8A). When we co-cultured CAF40TKi-shRNA control fibroblasts with MCF10.DCIS cells, we saw a phenotype similar to non-shRNA transduced cultures (Additional file 14: Figure S8B, cf. 3E). Knocking down CAF40TKi IL-6 expression in co-culture resulted in the formation of multicellular structures with uniform borders and few invasive processes (Additional file 14: Figure S8C). Co-culture with non-shRNA transduced CAF40TKi fibroblasts and shRNA-IL-6 transduced MCF10.DCIS cells showed greater MCF10.DCIS:CAF40TKi interaction and multicellular structure branching (Additional file 14: Figure S8D).
IL-6 signaling is propagated through either direct cell membrane receptor signaling or soluble receptor trans-signaling (TS). In the DCIS cell lines, we found that IL-6R expression was low and sometimes undetectable by qRT-PCR. Additionally, we detected very low levels of sIL-6R secreted from MCF10.DCIS cells and none from SUM102 cells. In contrast, CAFs had a higher level of IL-6R gene expression and high secretion of sIL-6R as verified by ELISA. These findings suggest that IL-6TS is a likely mechanism of IL-6 action in these DCIS cells [58] (Additional file 15: Figure S9).

Discussion

A number of factors produced by CAFs have been shown to be involved in promoting malignant transformation in epithelial cells, these include TGFß [5961] and CXCL12 (SDF-1) [59, 61, 62]. These factors are involved in eliciting a range of responses that are context-dependent and that benefit the tumor in various ways. The present study describes a novel addition to these known interactions and a new mechanism by which CAF can influence tumor progression. There is a direct correlation between serum IL-6 levels and poor prognosis in breast cancer patients [36, 6365]. Studies have shown that CAFs [66], and various immune/inflammatory cells secrete pro-inflammatory cytokines including IL-6 and contribute to tumorigenicity [6773].
In the current study, we show that IL-6 expression can be found in both the tumor and stromal compartments. In our IHC data we found approximately 65 % of patient samples had positive IL-6 staining; however, when we examined IL-6 expression in our DCIS tumor cell lines, we found the expression to be near the lower threshold of our assay. This discrepancy may be due to differences in gene expression between tissue and cells [74], differences in IL-6 expression with tumor grade/invasiveness [65], degree of “stemness” in cell lines vs. tissue [75], and differences between the assays and/or sample collection.
We next examined the interaction between human breast DCIS epithelial cells and human breast CAFs in the context of an in vitro 3D microenvironment. We hypothesized that CAFs promoted the migration of breast DCIS cells via paracrine signaling within the tumor microenvironment. Here we observed that DCIS cells migrated towards CAFs and upon attachment to CAFs, DCIS cells remained attached and migrated through the matrix following the lead of the CAFs. Similar findings of fibroblast-led migration have been reported for co-cultures of invasive squamous carcinoma cells and CAFs [76]. It is also likely that CAFs migrated to DCIS cells, as some fibroblasts moved more easily and quickly through the matrix as seen in time-lapse videos; therefore it is possible that some fibroblasts migrated to and attached to DCIS cells thereupon leading migration of the DCIS cells.
An increase in DCIS cell proliferation and a change in multicellular structure morphology was observed in all of our co-culture experiments. DCIS multicellular structures showed invasive characteristics, having lost their uniform circular structure and had developed single or multiple protruding edges. An underlying mechanism for DCIS:CAF interaction, enhanced tumor cell proliferation, and migration was IL-6 signaling in the tumor microenvironment. We [31] as well as Sung et al. [77] have shown in 3D co-culture systems that paracrine HGF and its receptor cMet can drive the invasive potential of the MCF10.DCIS cells. We confirmed this in xenografts formed by orthotopic implantation of HGF-secreting fibroblasts and MCF10.DCIS cells in SCID mice [31]. HGF and IL-6 have been shown to cooperatively enhance lung cancer cell invasion by upregulation of their corresponding cell surface receptors [78]. Also of interest is the stem cell-like properties of the proliferating DCIS cell population as Krishnamurthy et al. have shown that endothelial IL-6 enhances self-renewal of cancer stem-like cells. Whether or not this is the case for CAF IL-6 in regard to proliferation of DCIS cells has yet to be determined [79].
Treatment of DCIS:CAF co-cultures with an IL-6 nAb abrogated the proliferation and migratory phenotype acquired by DCIS cells. This phenotype was primarily produced by the inhibition of CAF secreted IL-6 as shRNA knockdown of IL-6 in CAFs, but not in DCIS cells was able to replicate the phenotype. We also showed that MCF10.DCIS cells treated with IL-6 nAb had a down regulation of genes associated with EMT. A recent study suggests that tumor cell EMT is mediated through factors secreted from CAFs and that selective inhibition of TGFβ1 is sufficient to reverse EMT associated gene expression [80]. Other studies show cross talk between IL-6 and TGFβ signaling consistent with IL-6 and TGFβ both acting as drivers of EMT [81].
A Federal Drug Administration (FDA) approved humanized anti-IL-6R antibody, tocilizumab (Actemra), has shown promise in the treatment of inflammatory diseases particularly rheumatoid arthritis, Crohn’s disease, and Castleman’s disease. A major caveat of Actemra is that serum IL-6 levels are increased in patients after drug administration [82]. Since breast cancer patients with elevated serum IL-6 have poorer prognosis, Actemra may not be a practical therapy in these patients, however alternative approaches to reduce IL-6 signaling may prove efficacious.
Siltuximab, a monoclonal antibody against IL-6, is in clinical trials for therapies including; combinatorial treatment of metastatic renal cell carcinoma, multiple myeloma, and prostate cancer [8386]. The FDA recently approved siltuximab for the treatment of multicentric Castleman’s disease [87]. Studies have shown that siltuximab significantly inhibits the growth of non-small-cell lung cancer in primary xenografts [88], and ovarian cancer cell xenografts [89]. Therapeutic use of siltuximab for the treatment of breast cancer has not been fully evaluated, although preliminary studies with the estrogen receptor alpha positive MCF-7 breast cancer cell line suggests potential efficacy [90].
A limitation of the current study is that we did not have complete histories or demographic data to perform correlation studies to determine how IL-6 expression in DCIS relates to tumor prognosis. Another limitation is there is a limited number of commercially available DCIS cell lines. Here we used two cell lines that are commercially available. A third DCIS cell line that is commercially available is the SUM-225 DCIS cell line; however, this line comes from a metastatic recurrence so we chose not to use it. Another limitation is that our study only followed the progression and interaction of DCIS cells with CAFs, but not inflammatory cells associated with breast tumors. Additionally, we are investigating the probability of increasing the number of cell types grown together in our 3D MAME culture, for example: macrophages, adipocytes and lymphocytes.

Conclusion

In conclusion, we have shown that IL-6 paracrine signaling between DCIS cells and CAFs is a key mediator of early stage breast cancer cell proliferation and migration. Furthermore, IL-6 from CAFs facilitated the transition of DCIS cells from preinvasive to an invasive phenotype. This study highlights the necessity to explore paracrine signaling within the context of the tumor microenvironment and components therein.

Acknowledgments

This work was funded in part by a Department of Defense Breast Cancer Research Program Postdoctoral Fellowship Award (W81XWH-12-1-0024; KO) and a multi-PI R01 CA131990 from the National Institutes of Health (BFS and RRM). The Microscopy, Imaging and Cytometry Resources Core is supported, in part, by NIH Center grant P30 CA022453 to the Karmanos Cancer Institute at Wayne State University, and the Perinatology Research Branch of the National Institutes of Child Health and Development at Wayne State University.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

KOO and BFS designed research plan; KOO made substantial contributions to conception, acquisition, analysis and interpretation of data; MS conducted immunohistochemistry and made substantial contribution to data analysis; NA, and SS made substantial contributions to data analysis; MLS performed video editing and contributed to data analysis; KOO, BFS, RRM, SWH, OEF, YH, and FB drafted the manuscript and participated in revising it for intellectual content. All authors read and approved the final manuscript.
Anhänge

Additional files

Additional file 4: Figure S2. Oxymatrine treatment. (A-B) MCF10.DCIS cells were grown in the presence of DMSO (A) or 1 mg/ml oxymatrine (B). Oxymatrine or DMSO was added 1 day after initial seeding. Scale bar, 200 μm. MCF10.DCIS cells grown in DMSO form large structures similar to untreated cultures (cf. Fig. 2a). (C) Measurement of structure diameters in three tiled images from three independent experiments. (D) Cell viability was evaluated using an ATP-based luminescence assay 24 h after drug treatment. Arbitrary units (A.U.). **P < 0.01, not significant (NS). Student’s t-test; mean ± SD. (TIFF 141 kb)
Additional file 12: Video S4. Video clip shows the transition through optical slices of a MCF10.DCIS structure in co-culture with CAF40TKi fibroblasts (Full 3D image shown in Fig. 4e and f). Cultures were fixed and immunostained for IL-6 (red), with DAPI labeled nuclei. MCF10.DCIS cells formed large nodular-like structures that branched out towards CFSE-labeled CAFs (pseudo-colored white). Images were obtained on a Zeiss LSM 780 and converted to a movie file using Volocity® software. (MOV 1 mb)
Literatur
1.
Zurück zum Zitat Virnig BA, Tuttle TM, Shamliyan T, Kane RL. Ductal carcinoma in situ of the breast: a systematic review of incidence, treatment, and outcomes. J Natl Cancer Inst. 2010;102(3):170–8.CrossRefPubMed Virnig BA, Tuttle TM, Shamliyan T, Kane RL. Ductal carcinoma in situ of the breast: a systematic review of incidence, treatment, and outcomes. J Natl Cancer Inst. 2010;102(3):170–8.CrossRefPubMed
2.
Zurück zum Zitat Society AC. Cancer Facts & Figures 2014. American Cancer Society. Cancer.org. 2014;8–10. Society AC. Cancer Facts & Figures 2014. American Cancer Society. Cancer.org. 2014;8–10.
3.
Zurück zum Zitat Sanders ME, Schuyler PA, Dupont WD, Page DL. The natural history of low-grade ductal carcinoma in situ of the breast in women treated by biopsy only revealed over 30 years of long-term follow-up. Cancer. 2005;103(12):2481–4.CrossRefPubMed Sanders ME, Schuyler PA, Dupont WD, Page DL. The natural history of low-grade ductal carcinoma in situ of the breast in women treated by biopsy only revealed over 30 years of long-term follow-up. Cancer. 2005;103(12):2481–4.CrossRefPubMed
4.
Zurück zum Zitat Page DL, Dupont WD, Rogers LW, Jensen RA, Schuyler PA. Continued local recurrence of carcinoma 15–25 years after a diagnosis of low grade ductal carcinoma in situ of the breast treated only by biopsy. Cancer. 1995;76(7):1197–200.CrossRefPubMed Page DL, Dupont WD, Rogers LW, Jensen RA, Schuyler PA. Continued local recurrence of carcinoma 15–25 years after a diagnosis of low grade ductal carcinoma in situ of the breast treated only by biopsy. Cancer. 1995;76(7):1197–200.CrossRefPubMed
5.
Zurück zum Zitat Network NCC. Stage 0 Breast Cancer. NCCN Guidlines for Patients. 2014;1(2014):17–28. Network NCC. Stage 0 Breast Cancer. NCCN Guidlines for Patients. 2014;1(2014):17–28.
6.
Zurück zum Zitat Bissell MJ, Radisky DC, Rizki A, Weaver VM, Petersen OW. The organizing principle: microenvironmental influences in the normal and malignant breast. Differentiation. 2002;70(9–10):537–46.CrossRefPubMedPubMedCentral Bissell MJ, Radisky DC, Rizki A, Weaver VM, Petersen OW. The organizing principle: microenvironmental influences in the normal and malignant breast. Differentiation. 2002;70(9–10):537–46.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Lee GY, Kenny PA, Lee EH, Bissell MJ. Three-dimensional culture models of normal and malignant breast epithelial cells. Nat Methods. 2007;4(4):359–65.CrossRefPubMedPubMedCentral Lee GY, Kenny PA, Lee EH, Bissell MJ. Three-dimensional culture models of normal and malignant breast epithelial cells. Nat Methods. 2007;4(4):359–65.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Ma XJ, Dahiya S, Richardson E, Erlander M, Sgroi DC. Gene expression profiling of the tumor microenvironment during breast cancer progression. Breast Cancer Res. 2009;11(1):R7.CrossRefPubMedPubMedCentral Ma XJ, Dahiya S, Richardson E, Erlander M, Sgroi DC. Gene expression profiling of the tumor microenvironment during breast cancer progression. Breast Cancer Res. 2009;11(1):R7.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Hu M, Yao J, Cai L, Bachman KE, van den Brule F, Velculescu V, et al. Distinct epigenetic changes in the stromal cells of breast cancers. Nat Genet. 2005;37(8):899–905. Hu M, Yao J, Cai L, Bachman KE, van den Brule F, Velculescu V, et al. Distinct epigenetic changes in the stromal cells of breast cancers. Nat Genet. 2005;37(8):899–905.
10.
Zurück zum Zitat Sung KE, Yang N, Pehlke C, Keely PJ, Eliceiri KW, Friedl A, et al. Transition to invasion in breast cancer: a microfluidic in vitro model enables examination of spatial and temporal effects. Integr Biol. 2011;3(4):439–50. Sung KE, Yang N, Pehlke C, Keely PJ, Eliceiri KW, Friedl A, et al. Transition to invasion in breast cancer: a microfluidic in vitro model enables examination of spatial and temporal effects. Integr Biol. 2011;3(4):439–50.
11.
Zurück zum Zitat Madar S, Goldstein I, Rotter V. Cancer associated fibroblasts’--more than meets the eye. Trends Mol Med. 2013;19(8):447–53.CrossRefPubMed Madar S, Goldstein I, Rotter V. Cancer associated fibroblasts’--more than meets the eye. Trends Mol Med. 2013;19(8):447–53.CrossRefPubMed
12.
Zurück zum Zitat Campbell I, Polyak K, Haviv I. Clonal mutations in the cancer-associated fibroblasts: the case against genetic coevolution. Cancer Res. 2009;69(17):6765–8. discussion 6769.CrossRefPubMed Campbell I, Polyak K, Haviv I. Clonal mutations in the cancer-associated fibroblasts: the case against genetic coevolution. Cancer Res. 2009;69(17):6765–8. discussion 6769.CrossRefPubMed
13.
Zurück zum Zitat Qiu W, Hu M, Sridhar A, Opeskin K, Fox S, Shipitsin M, et al. No evidence of clonal somatic genetic alterations in cancer-associated fibroblasts from human breast and ovarian carcinomas. Nat Genet. 2008;40(5):650–5. Qiu W, Hu M, Sridhar A, Opeskin K, Fox S, Shipitsin M, et al. No evidence of clonal somatic genetic alterations in cancer-associated fibroblasts from human breast and ovarian carcinomas. Nat Genet. 2008;40(5):650–5.
15.
Zurück zum Zitat Tamm I, Cardinale I, Murphy JS. Decreased adherence of interleukin 6-treated breast carcinoma cells can lead to separation from neighbors after mitosis. Proc Natl Acad Sci U S A. 1991;88(10):4414–8.CrossRefPubMedPubMedCentral Tamm I, Cardinale I, Murphy JS. Decreased adherence of interleukin 6-treated breast carcinoma cells can lead to separation from neighbors after mitosis. Proc Natl Acad Sci U S A. 1991;88(10):4414–8.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Tamm I, Cardinale I, Kikuchi T, Krueger JG. E-cadherin distribution in interleukin 6-induced cell-cell separation of ductal breast carcinoma cells. Proc Natl Acad Sci U S A. 1994;91(10):4338–42.CrossRefPubMedPubMedCentral Tamm I, Cardinale I, Kikuchi T, Krueger JG. E-cadherin distribution in interleukin 6-induced cell-cell separation of ductal breast carcinoma cells. Proc Natl Acad Sci U S A. 1994;91(10):4338–42.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Krueger J, Ray A, Tamm I, Sehgal PB. Expression and function of interleukin-6 in epithelial cells. J Cell Biochem. 1991;45(4):327–34.CrossRefPubMed Krueger J, Ray A, Tamm I, Sehgal PB. Expression and function of interleukin-6 in epithelial cells. J Cell Biochem. 1991;45(4):327–34.CrossRefPubMed
18.
Zurück zum Zitat Cheng GZ, Zhang WZ, Sun M, Wang Q, Coppola D, Mansour M, et al. Twist is transcriptionally induced by activation of STAT3 and mediates STAT3 oncogenic function. J Biol Chem. 2008;283(21):14665–73. Cheng GZ, Zhang WZ, Sun M, Wang Q, Coppola D, Mansour M, et al. Twist is transcriptionally induced by activation of STAT3 and mediates STAT3 oncogenic function. J Biol Chem. 2008;283(21):14665–73.
19.
Zurück zum Zitat Sullivan NJ, Sasser AK, Axel AE, Vesuna F, Raman V, Ramirez N, et al. Interleukin-6 induces an epithelial-mesenchymal transition phenotype in human breast cancer cells. Oncogene. 2009;28(33):2940–7. Sullivan NJ, Sasser AK, Axel AE, Vesuna F, Raman V, Ramirez N, et al. Interleukin-6 induces an epithelial-mesenchymal transition phenotype in human breast cancer cells. Oncogene. 2009;28(33):2940–7.
20.
Zurück zum Zitat Rose-John S, Heinrich PC. Soluble receptors for cytokines and growth factors: generation and biological function. Biochem J. 1994;300(Pt 2):281–90.CrossRefPubMedPubMedCentral Rose-John S, Heinrich PC. Soluble receptors for cytokines and growth factors: generation and biological function. Biochem J. 1994;300(Pt 2):281–90.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Mohamed MM, Cavallo-Medved D, Rudy D, Anbalagan A, Moin K, Sloane BF. Interleukin-6 increases expression and secretion of cathepsin B by breast tumor-associated monocytes. Cell Physiol Biochem. 2010;25(2–3):315–24.CrossRefPubMedPubMedCentral Mohamed MM, Cavallo-Medved D, Rudy D, Anbalagan A, Moin K, Sloane BF. Interleukin-6 increases expression and secretion of cathepsin B by breast tumor-associated monocytes. Cell Physiol Biochem. 2010;25(2–3):315–24.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Bengsch F, Buck A, Gunther SC, Seiz JR, Tacke M, Pfeifer D, et al. Cell type-dependent pathogenic functions of overexpressed human cathepsin B in murine breast cancer progression. Oncogene. 2013. Bengsch F, Buck A, Gunther SC, Seiz JR, Tacke M, Pfeifer D, et al. Cell type-dependent pathogenic functions of overexpressed human cathepsin B in murine breast cancer progression. Oncogene. 2013.
23.
Zurück zum Zitat Hobisch A, Rogatsch H, Hittmair A, Fuchs D, Bartsch Jr G, Klocker H, et al. Immunohistochemical localization of interleukin-6 and its receptor in benign, premalignant and malignant prostate tissue. J Pathol. 2000;191(3):239–44. Hobisch A, Rogatsch H, Hittmair A, Fuchs D, Bartsch Jr G, Klocker H, et al. Immunohistochemical localization of interleukin-6 and its receptor in benign, premalignant and malignant prostate tissue. J Pathol. 2000;191(3):239–44.
24.
Zurück zum Zitat Kinoshita T, Ito H, Miki C. Serum interleukin-6 level reflects the tumor proliferative activity in patients with colorectal carcinoma. Cancer. 1999;85(12):2526–31.CrossRefPubMed Kinoshita T, Ito H, Miki C. Serum interleukin-6 level reflects the tumor proliferative activity in patients with colorectal carcinoma. Cancer. 1999;85(12):2526–31.CrossRefPubMed
25.
Zurück zum Zitat Guo Y, Xu F, Lu T, Duan Z, Zhang Z. Interleukin-6 signaling pathway in targeted therapy for cancer. Cancer Treat Rev. 2012;38(7):904–10.CrossRefPubMed Guo Y, Xu F, Lu T, Duan Z, Zhang Z. Interleukin-6 signaling pathway in targeted therapy for cancer. Cancer Treat Rev. 2012;38(7):904–10.CrossRefPubMed
26.
Zurück zum Zitat Hartman ZC, Poage GM, den Hollander P, Tsimelzon A, Hill J, Panupinthu N, et al. Growth of triple-negative breast cancer cells relies upon coordinate autocrine expression of the proinflammatory cytokines IL-6 and IL-8. Cancer Res. 2013;73(11):3470–80. Hartman ZC, Poage GM, den Hollander P, Tsimelzon A, Hill J, Panupinthu N, et al. Growth of triple-negative breast cancer cells relies upon coordinate autocrine expression of the proinflammatory cytokines IL-6 and IL-8. Cancer Res. 2013;73(11):3470–80.
27.
Zurück zum Zitat Jiang XP, Yang DC, Elliott RL, Head JF. Down-regulation of expression of interleukin-6 and its receptor results in growth inhibition of MCF-7 breast cancer cells. Anticancer Res. 2011;31(9):2899–906.PubMed Jiang XP, Yang DC, Elliott RL, Head JF. Down-regulation of expression of interleukin-6 and its receptor results in growth inhibition of MCF-7 breast cancer cells. Anticancer Res. 2011;31(9):2899–906.PubMed
28.
Zurück zum Zitat Leslie K, Gao SP, Berishaj M, Podsypanina K, Ho H, Ivashkiv L, et al. Differential interleukin-6/Stat3 signaling as a function of cellular context mediates Ras-induced transformation. Breast Cancer Res. 2010;12(5):R80. Leslie K, Gao SP, Berishaj M, Podsypanina K, Ho H, Ivashkiv L, et al. Differential interleukin-6/Stat3 signaling as a function of cellular context mediates Ras-induced transformation. Breast Cancer Res. 2010;12(5):R80.
29.
Zurück zum Zitat Lovitt CJ, Shelper TB, Avery VM. Evaluation of chemotherapeutics in a three-dimensional breast cancer model. J Cancer Res Clin Oncol. 2015;141(5):951–9.CrossRefPubMed Lovitt CJ, Shelper TB, Avery VM. Evaluation of chemotherapeutics in a three-dimensional breast cancer model. J Cancer Res Clin Oncol. 2015;141(5):951–9.CrossRefPubMed
30.
Zurück zum Zitat Dawson PJ, Wolman SR, Tait L, Heppner GH, Miller FR. MCF10AT: a model for the evolution of cancer from proliferative breast disease. Am J Pathol. 1996;148(1):313–9.PubMedPubMedCentral Dawson PJ, Wolman SR, Tait L, Heppner GH, Miller FR. MCF10AT: a model for the evolution of cancer from proliferative breast disease. Am J Pathol. 1996;148(1):313–9.PubMedPubMedCentral
31.
Zurück zum Zitat Jedeszko C, Victor BC, Podgorski I, Sloane BF. Fibroblast hepatocyte growth factor promotes invasion of human mammary ductal carcinoma in situ. Cancer Res. 2009;69(23):9148–55.CrossRefPubMedPubMedCentral Jedeszko C, Victor BC, Podgorski I, Sloane BF. Fibroblast hepatocyte growth factor promotes invasion of human mammary ductal carcinoma in situ. Cancer Res. 2009;69(23):9148–55.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Sameni M, Anbalagan A, Olive MB, Moin K, Mattingly RR, Sloane BF. MAME models for 4D live-cell imaging of tumor: microenvironment interactions that impact malignant progression. J Vis Exp. 2012;(60). doi: 10.3791/3661. Sameni M, Anbalagan A, Olive MB, Moin K, Mattingly RR, Sloane BF. MAME models for 4D live-cell imaging of tumor: microenvironment interactions that impact malignant progression. J Vis Exp. 2012;(60). doi: 10.3791/3661.
33.
Zurück zum Zitat Cavallo-Medved D, Rudy D, Blum G, Bogyo M, Caglic D, Sloane BF. Live-cell imaging demonstrates extracellular matrix degradation in association with active cathepsin B in caveolae of endothelial cells during tube formation. Exp Cell Res. 2009;315(7):1234–46.CrossRefPubMedPubMedCentral Cavallo-Medved D, Rudy D, Blum G, Bogyo M, Caglic D, Sloane BF. Live-cell imaging demonstrates extracellular matrix degradation in association with active cathepsin B in caveolae of endothelial cells during tube formation. Exp Cell Res. 2009;315(7):1234–46.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Sloane BF, Moin K, Sameni M, Tait LR, Rozhin J, Ziegler G. Membrane association of cathepsin B can be induced by transfection of human breast epithelial cells with c-Ha-ras oncogene. J Cell Sci. 1994;107(Pt 2):373–84.PubMed Sloane BF, Moin K, Sameni M, Tait LR, Rozhin J, Ziegler G. Membrane association of cathepsin B can be induced by transfection of human breast epithelial cells with c-Ha-ras oncogene. J Cell Sci. 1994;107(Pt 2):373–84.PubMed
35.
Zurück zum Zitat Mullins SR, Sameni M, Blum G, Bogyo M, Sloane BF, Moin K. Three-dimensional cultures modeling premalignant progression of human breast epithelial cells: role of cysteine cathepsins. Biol Chem. 2012;393(12):1405–16.CrossRefPubMedPubMedCentral Mullins SR, Sameni M, Blum G, Bogyo M, Sloane BF, Moin K. Three-dimensional cultures modeling premalignant progression of human breast epithelial cells: role of cysteine cathepsins. Biol Chem. 2012;393(12):1405–16.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Dethlefsen C, Hojfeldt G, Hojman P. The role of intratumoral and systemic IL-6 in breast cancer. Breast Cancer Res Treat. 2013;138(3):657–64.CrossRefPubMed Dethlefsen C, Hojfeldt G, Hojman P. The role of intratumoral and systemic IL-6 in breast cancer. Breast Cancer Res Treat. 2013;138(3):657–64.CrossRefPubMed
37.
Zurück zum Zitat Bachelot T, Ray-Coquard I, Menetrier-Caux C, Rastkha M, Duc A, Blay JY. Prognostic value of serum levels of interleukin 6 and of serum and plasma levels of vascular endothelial growth factor in hormone-refractory metastatic breast cancer patients. Br J Cancer. 2003;88(11):1721–6.CrossRefPubMedPubMedCentral Bachelot T, Ray-Coquard I, Menetrier-Caux C, Rastkha M, Duc A, Blay JY. Prognostic value of serum levels of interleukin 6 and of serum and plasma levels of vascular endothelial growth factor in hormone-refractory metastatic breast cancer patients. Br J Cancer. 2003;88(11):1721–6.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Salgado R, Junius S, Benoy I, Van Dam P, Vermeulen P, Van Marck E, et al. Circulating interleukin-6 predicts survival in patients with metastatic breast cancer. Int J Cancer. 2003;103(5):642–6. Salgado R, Junius S, Benoy I, Van Dam P, Vermeulen P, Van Marck E, et al. Circulating interleukin-6 predicts survival in patients with metastatic breast cancer. Int J Cancer. 2003;103(5):642–6.
39.
Zurück zum Zitat Yang L, Han S, Sun Y. An IL6-STAT3 loop mediates resistance to PI3K inhibitors by inducing epithelial-mesenchymal transition and cancer stem cell expansion in human breast cancer cells. Biochem Biophys Res Commun. 2014;453(3):582–7.CrossRefPubMed Yang L, Han S, Sun Y. An IL6-STAT3 loop mediates resistance to PI3K inhibitors by inducing epithelial-mesenchymal transition and cancer stem cell expansion in human breast cancer cells. Biochem Biophys Res Commun. 2014;453(3):582–7.CrossRefPubMed
40.
Zurück zum Zitat Hung SY, Shih YP, Chen M, Lo SH. Up-regulated cten by FGF2 contributes to FGF2-mediated cell migration. Mol Carcinog. 2014;53(10):787–92.CrossRefPubMed Hung SY, Shih YP, Chen M, Lo SH. Up-regulated cten by FGF2 contributes to FGF2-mediated cell migration. Mol Carcinog. 2014;53(10):787–92.CrossRefPubMed
41.
Zurück zum Zitat Patel NA, Patel PS, Vora HH. Role of PRL-3, Snail, Cytokeratin and Vimentin expression in epithelial mesenchymal transition in breast carcinoma. Breast Dis. 2014;35(2):113–127. Patel NA, Patel PS, Vora HH. Role of PRL-3, Snail, Cytokeratin and Vimentin expression in epithelial mesenchymal transition in breast carcinoma. Breast Dis. 2014;35(2):113–127.
42.
Zurück zum Zitat D’Angelo RC, Liu XW, Najy AJ, Jung YS, Won J, Chai KX, et al. TIMP-1 via TWIST1 induces EMT phenotypes in human breast epithelial cells. Mol Cancer Res. 2014;12(9):1324–33. D’Angelo RC, Liu XW, Najy AJ, Jung YS, Won J, Chai KX, et al. TIMP-1 via TWIST1 induces EMT phenotypes in human breast epithelial cells. Mol Cancer Res. 2014;12(9):1324–33.
43.
Zurück zum Zitat Ibrahim SA, Hassan H, Vilardo L, Kumar SK, Kumar AV, Kelsch R, et al. Syndecan-1 (CD138) modulates triple-negative breast cancer stem cell properties via regulation of LRP-6 and IL-6-mediated STAT3 signaling. PLoS One. 2013;8(12), e85737. Ibrahim SA, Hassan H, Vilardo L, Kumar SK, Kumar AV, Kelsch R, et al. Syndecan-1 (CD138) modulates triple-negative breast cancer stem cell properties via regulation of LRP-6 and IL-6-mediated STAT3 signaling. PLoS One. 2013;8(12), e85737.
44.
Zurück zum Zitat Guzman JR, Koo JS, Goldsmith JR, Muhlbauer M, Narula A, Jobin C. Oxymatrine prevents NF-kappaB nuclear translocation and ameliorates acute intestinal inflammation. Sci Rep. 2013;3:1629.CrossRefPubMedPubMedCentral Guzman JR, Koo JS, Goldsmith JR, Muhlbauer M, Narula A, Jobin C. Oxymatrine prevents NF-kappaB nuclear translocation and ameliorates acute intestinal inflammation. Sci Rep. 2013;3:1629.CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Cirri P, Chiarugi P. Cancer associated fibroblasts: the dark side of the coin. Am J Cancer Res. 2011;1(4):482–97.PubMedPubMedCentral Cirri P, Chiarugi P. Cancer associated fibroblasts: the dark side of the coin. Am J Cancer Res. 2011;1(4):482–97.PubMedPubMedCentral
46.
Zurück zum Zitat Orimo A, Weinberg RA. Stromal fibroblasts in cancer: a novel tumor-promoting cell type. Cell Cycle. 2006;5(15):1597–601.CrossRefPubMed Orimo A, Weinberg RA. Stromal fibroblasts in cancer: a novel tumor-promoting cell type. Cell Cycle. 2006;5(15):1597–601.CrossRefPubMed
47.
Zurück zum Zitat Olumi AF, Grossfeld GD, Hayward SW, Carroll PR, Tlsty TD, Cunha GR. Carcinoma-associated fibroblasts direct tumor progression of initiated human prostatic epithelium. Cancer Res. 1999;59(19):5002–11.PubMed Olumi AF, Grossfeld GD, Hayward SW, Carroll PR, Tlsty TD, Cunha GR. Carcinoma-associated fibroblasts direct tumor progression of initiated human prostatic epithelium. Cancer Res. 1999;59(19):5002–11.PubMed
48.
Zurück zum Zitat Chang HY, Chi JT, Dudoit S, Bondre C, van de Rijn M, Botstein D, et al. Diversity, topographic differentiation, and positional memory in human fibroblasts. Proc Natl Acad Sci U S A. 2002;99(20):12877–82. Chang HY, Chi JT, Dudoit S, Bondre C, van de Rijn M, Botstein D, et al. Diversity, topographic differentiation, and positional memory in human fibroblasts. Proc Natl Acad Sci U S A. 2002;99(20):12877–82.
49.
Zurück zum Zitat Rowe RG, Keena D, Sabeh F, Willis AL, Weiss SJ. Pulmonary fibroblasts mobilize the membrane-tethered matrix metalloprotease, MT1-MMP, to destructively remodel and invade interstitial type I collagen barriers. Am J Physiol Lung Cell Mol Physiol. 2011;301(5):L683–92.CrossRefPubMed Rowe RG, Keena D, Sabeh F, Willis AL, Weiss SJ. Pulmonary fibroblasts mobilize the membrane-tethered matrix metalloprotease, MT1-MMP, to destructively remodel and invade interstitial type I collagen barriers. Am J Physiol Lung Cell Mol Physiol. 2011;301(5):L683–92.CrossRefPubMed
50.
Zurück zum Zitat Holliday DL, Hughes S, Shaw JA, Walker RA, Jones JL. Intrinsic genetic characteristics determine tumor-modifying capacity of fibroblasts: matrix metalloproteinase-3 5A/5A genotype enhances breast cancer cell invasion. Breast Cancer Res. 2007;9(5):R67.CrossRefPubMedPubMedCentral Holliday DL, Hughes S, Shaw JA, Walker RA, Jones JL. Intrinsic genetic characteristics determine tumor-modifying capacity of fibroblasts: matrix metalloproteinase-3 5A/5A genotype enhances breast cancer cell invasion. Breast Cancer Res. 2007;9(5):R67.CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat Casey T, Bond J, Tighe S, Hunter T, Lintault L, Patel O, et al. Molecular signatures suggest a major role for stromal cells in development of invasive breast cancer. Breast Cancer Res Treat. 2009;114(1):47–62. Casey T, Bond J, Tighe S, Hunter T, Lintault L, Patel O, et al. Molecular signatures suggest a major role for stromal cells in development of invasive breast cancer. Breast Cancer Res Treat. 2009;114(1):47–62.
52.
Zurück zum Zitat Friedl P, Alexander S. Cancer invasion and the microenvironment: plasticity and reciprocity. Cell. 2011;147(5):992–1009.CrossRefPubMed Friedl P, Alexander S. Cancer invasion and the microenvironment: plasticity and reciprocity. Cell. 2011;147(5):992–1009.CrossRefPubMed
53.
Zurück zum Zitat Egeblad M, Werb Z. New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2002;2(3):161–74.CrossRefPubMed Egeblad M, Werb Z. New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2002;2(3):161–74.CrossRefPubMed
54.
Zurück zum Zitat O’Brien P, O’Connor BF. Seprase: an overview of an important matrix serine protease. Biochim Biophys Acta. 2008;1784(9):1130–45.CrossRefPubMed O’Brien P, O’Connor BF. Seprase: an overview of an important matrix serine protease. Biochim Biophys Acta. 2008;1784(9):1130–45.CrossRefPubMed
55.
Zurück zum Zitat Linebaugh BE, Sameni M, Day NA, Sloane BF, Keppler D. Exocytosis of active cathepsin B enzyme activity at pH 7.0, inhibition and molecular mass. Eur J Biochem. 1999;264(1):100–9.CrossRefPubMed Linebaugh BE, Sameni M, Day NA, Sloane BF, Keppler D. Exocytosis of active cathepsin B enzyme activity at pH 7.0, inhibition and molecular mass. Eur J Biochem. 1999;264(1):100–9.CrossRefPubMed
56.
Zurück zum Zitat Nielsen BS, Rank F, Lopez JM, Balbin M, Vizoso F, Lund LR, et al. Collagenase-3 expression in breast myofibroblasts as a molecular marker of transition of ductal carcinoma in situ lesions to invasive ductal carcinomas. Cancer Res. 2001;61(19):7091–100. Nielsen BS, Rank F, Lopez JM, Balbin M, Vizoso F, Lund LR, et al. Collagenase-3 expression in breast myofibroblasts as a molecular marker of transition of ductal carcinoma in situ lesions to invasive ductal carcinomas. Cancer Res. 2001;61(19):7091–100.
58.
Zurück zum Zitat Peters M, Muller AM, Rose-John S. Interleukin-6 and soluble interleukin-6 receptor: direct stimulation of gp130 and hematopoiesis. Blood. 1998;92(10):3495–504.PubMed Peters M, Muller AM, Rose-John S. Interleukin-6 and soluble interleukin-6 receptor: direct stimulation of gp130 and hematopoiesis. Blood. 1998;92(10):3495–504.PubMed
59.
Zurück zum Zitat Ao M, Franco OE, Park D, Raman D, Williams K, Hayward SW. Cross-talk between paracrine-acting cytokine and chemokine pathways promotes malignancy in benign human prostatic epithelium. Cancer Res. 2007;67(9):4244–53.CrossRefPubMed Ao M, Franco OE, Park D, Raman D, Williams K, Hayward SW. Cross-talk between paracrine-acting cytokine and chemokine pathways promotes malignancy in benign human prostatic epithelium. Cancer Res. 2007;67(9):4244–53.CrossRefPubMed
60.
Zurück zum Zitat Franco OE, Jiang M, Strand DW, Peacock J, Fernandez S, Jackson 2nd RS, et al. Altered TGF-beta signaling in a subpopulation of human stromal cells promotes prostatic carcinogenesis. Cancer Res. 2011;71(4):1272–81. Franco OE, Jiang M, Strand DW, Peacock J, Fernandez S, Jackson 2nd RS, et al. Altered TGF-beta signaling in a subpopulation of human stromal cells promotes prostatic carcinogenesis. Cancer Res. 2011;71(4):1272–81.
61.
Zurück zum Zitat Hendrayani SF, Al-Khalaf HH, Aboussekhra A. The Cytokine IL-6 Reactivates Breast Stromal Fibroblasts through Transcription Factor STAT3-dependent Up-regulation of the RNA Binding Protein AUF1. J Biol Chem. 2014. Hendrayani SF, Al-Khalaf HH, Aboussekhra A. The Cytokine IL-6 Reactivates Breast Stromal Fibroblasts through Transcription Factor STAT3-dependent Up-regulation of the RNA Binding Protein AUF1. J Biol Chem. 2014.
62.
Zurück zum Zitat Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005;121(3):335–48. Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005;121(3):335–48.
63.
Zurück zum Zitat Cho YA, Sung MK, Yeon JY, Ro J, Kim J. Prognostic role of interleukin-6, interleukin-8, and leptin levels according to breast cancer subtype. Cancer Res Treat. 2013;45(3):210–9.CrossRefPubMedPubMedCentral Cho YA, Sung MK, Yeon JY, Ro J, Kim J. Prognostic role of interleukin-6, interleukin-8, and leptin levels according to breast cancer subtype. Cancer Res Treat. 2013;45(3):210–9.CrossRefPubMedPubMedCentral
64.
Zurück zum Zitat Narita D, Seclaman E, Ursoniu S, Ilina R, Cireap N, Anghel A. Expression of CCL18 and interleukin-6 in the plasma of breast cancer patients as compared with benign tumor patients and healthy controls. Rom J Morphol Embryol. 2011;52(4):1261–7.PubMed Narita D, Seclaman E, Ursoniu S, Ilina R, Cireap N, Anghel A. Expression of CCL18 and interleukin-6 in the plasma of breast cancer patients as compared with benign tumor patients and healthy controls. Rom J Morphol Embryol. 2011;52(4):1261–7.PubMed
65.
Zurück zum Zitat Ravishankaran P, Karunanithi R. Clinical significance of preoperative serum interleukin-6 and C-reactive protein level in breast cancer patients. World J Surg Oncol. 2011;9:18.CrossRefPubMedPubMedCentral Ravishankaran P, Karunanithi R. Clinical significance of preoperative serum interleukin-6 and C-reactive protein level in breast cancer patients. World J Surg Oncol. 2011;9:18.CrossRefPubMedPubMedCentral
66.
Zurück zum Zitat Franco OE, Shaw AK, Strand DW, Hayward SW. Cancer associated fibroblasts in cancer pathogenesis. Semin Cell Dev Biol. 2010;21(1):33–9.CrossRefPubMed Franco OE, Shaw AK, Strand DW, Hayward SW. Cancer associated fibroblasts in cancer pathogenesis. Semin Cell Dev Biol. 2010;21(1):33–9.CrossRefPubMed
67.
Zurück zum Zitat Laoui D, Movahedi K, Van Overmeire E, Van den Bossche J, Schouppe E, Mommer C, et al. Tumor-associated macrophages in breast cancer: distinct subsets, distinct functions. Int J Dev Biol. 2011;55(7–9):861–7. Laoui D, Movahedi K, Van Overmeire E, Van den Bossche J, Schouppe E, Mommer C, et al. Tumor-associated macrophages in breast cancer: distinct subsets, distinct functions. Int J Dev Biol. 2011;55(7–9):861–7.
68.
Zurück zum Zitat Medrek C, Ponten F, Jirstrom K, Leandersson K. The presence of tumor associated macrophages in tumor stroma as a prognostic marker for breast cancer patients. BMC Cancer. 2012;12:306.CrossRefPubMedPubMedCentral Medrek C, Ponten F, Jirstrom K, Leandersson K. The presence of tumor associated macrophages in tumor stroma as a prognostic marker for breast cancer patients. BMC Cancer. 2012;12:306.CrossRefPubMedPubMedCentral
69.
Zurück zum Zitat Rudnick JA, Kuperwasser C. Stromal biomarkers in breast cancer development and progression. Clin Exp Metastasis. 2012;29(7):663–72.CrossRefPubMed Rudnick JA, Kuperwasser C. Stromal biomarkers in breast cancer development and progression. Clin Exp Metastasis. 2012;29(7):663–72.CrossRefPubMed
70.
Zurück zum Zitat Palucka K, Coussens LM, O’Shaughnessy J. Dendritic cells, inflammation, and breast cancer. Cancer J. 2013;19(6):511–6.CrossRefPubMed Palucka K, Coussens LM, O’Shaughnessy J. Dendritic cells, inflammation, and breast cancer. Cancer J. 2013;19(6):511–6.CrossRefPubMed
71.
Zurück zum Zitat Kees T, Egeblad M. Innate immune cells in breast cancer--from villains to heroes? J Mammary Gland Biol Neoplasia. 2011;16(3):189–203.CrossRefPubMed Kees T, Egeblad M. Innate immune cells in breast cancer--from villains to heroes? J Mammary Gland Biol Neoplasia. 2011;16(3):189–203.CrossRefPubMed
72.
Zurück zum Zitat Fridman WH, Galon J, Pages F, Tartour E, Sautes-Fridman C, Kroemer G. Prognostic and predictive impact of intra- and peritumoral immune infiltrates. Cancer Res. 2011;71(17):5601–5.CrossRefPubMed Fridman WH, Galon J, Pages F, Tartour E, Sautes-Fridman C, Kroemer G. Prognostic and predictive impact of intra- and peritumoral immune infiltrates. Cancer Res. 2011;71(17):5601–5.CrossRefPubMed
73.
Zurück zum Zitat DeNardo DG, Coussens LM. Inflammation and breast cancer. Balancing immune response: crosstalk between adaptive and innate immune cells during breast cancer progression. Breast Cancer Res. 2007;9(4):212.CrossRefPubMedPubMedCentral DeNardo DG, Coussens LM. Inflammation and breast cancer. Balancing immune response: crosstalk between adaptive and innate immune cells during breast cancer progression. Breast Cancer Res. 2007;9(4):212.CrossRefPubMedPubMedCentral
74.
Zurück zum Zitat Wilson KS, Roberts H, Leek R, Harris AL, Geradts J. Differential gene expression patterns in HER2/neu-positive and -negative breast cancer cell lines and tissues. Am J Pathol. 2002;161(4):1171–85.CrossRefPubMedPubMedCentral Wilson KS, Roberts H, Leek R, Harris AL, Geradts J. Differential gene expression patterns in HER2/neu-positive and -negative breast cancer cell lines and tissues. Am J Pathol. 2002;161(4):1171–85.CrossRefPubMedPubMedCentral
75.
Zurück zum Zitat Kim G, Ouzounova M, Quraishi AA, Davis A, Tawakkol N, Clouthier SG, et al. SOCS3-mediated regulation of inflammatory cytokines in PTEN and p53 inactivated triple negative breast cancer model. Oncogene. 2015;34(6):671–80. Kim G, Ouzounova M, Quraishi AA, Davis A, Tawakkol N, Clouthier SG, et al. SOCS3-mediated regulation of inflammatory cytokines in PTEN and p53 inactivated triple negative breast cancer model. Oncogene. 2015;34(6):671–80.
76.
Zurück zum Zitat Gaggioli C, Hooper S, Hidalgo-Carcedo C, Grosse R, Marshall JF, Harrington K, et al. Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nat Cell Biol. 2007;9(12):1392–400. Gaggioli C, Hooper S, Hidalgo-Carcedo C, Grosse R, Marshall JF, Harrington K, et al. Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nat Cell Biol. 2007;9(12):1392–400.
77.
Zurück zum Zitat Sung KE, Su X, Berthier E, Pehlke C, Friedl A, Beebe DJ. Understanding the impact of 2D and 3D fibroblast cultures on in vitro breast cancer models. PLoS One. 2013;8(10), e76373.CrossRefPubMedPubMedCentral Sung KE, Su X, Berthier E, Pehlke C, Friedl A, Beebe DJ. Understanding the impact of 2D and 3D fibroblast cultures on in vitro breast cancer models. PLoS One. 2013;8(10), e76373.CrossRefPubMedPubMedCentral
78.
Zurück zum Zitat To Y, Dohi M, Matsumoto K, Tanaka R, Sato A, Nakagome K, et al. A two-way interaction between hepatocyte growth factor and interleukin-6 in tissue invasion of lung cancer cell line. Am J Respir Cell Mol Biol. 2002;27(2):220–6. To Y, Dohi M, Matsumoto K, Tanaka R, Sato A, Nakagome K, et al. A two-way interaction between hepatocyte growth factor and interleukin-6 in tissue invasion of lung cancer cell line. Am J Respir Cell Mol Biol. 2002;27(2):220–6.
79.
Zurück zum Zitat Krishnamurthy S, Warner KA, Dong Z, Imai A, Nor C, Ward BB, et al. Endothelial interleukin-6 defines the tumorigenic potential of primary human cancer stem cells. Stem Cells. 2014;32(11):2845–57. Krishnamurthy S, Warner KA, Dong Z, Imai A, Nor C, Ward BB, et al. Endothelial interleukin-6 defines the tumorigenic potential of primary human cancer stem cells. Stem Cells. 2014;32(11):2845–57.
80.
Zurück zum Zitat Yu Y, Xiao CH, Tan LD, Wang QS, Li XQ, Feng YM. Cancer-associated fibroblasts induce epithelial-mesenchymal transition of breast cancer cells through paracrine TGF-beta signalling. Br J Cancer. 2014;110(3):724–32.CrossRefPubMed Yu Y, Xiao CH, Tan LD, Wang QS, Li XQ, Feng YM. Cancer-associated fibroblasts induce epithelial-mesenchymal transition of breast cancer cells through paracrine TGF-beta signalling. Br J Cancer. 2014;110(3):724–32.CrossRefPubMed
81.
Zurück zum Zitat Zhang XL, Topley N, Ito T, Phillips A. Interleukin-6 regulation of transforming growth factor (TGF)-beta receptor compartmentalization and turnover enhances TGF-beta1 signaling. J Biol Chem. 2005;280(13):12239–45.CrossRefPubMed Zhang XL, Topley N, Ito T, Phillips A. Interleukin-6 regulation of transforming growth factor (TGF)-beta receptor compartmentalization and turnover enhances TGF-beta1 signaling. J Biol Chem. 2005;280(13):12239–45.CrossRefPubMed
82.
Zurück zum Zitat Nishimoto N, Terao K, Mima T, Nakahara H, Takagi N, Kakehi T. Mechanisms and pathologic significances in increase in serum interleukin-6 (IL-6) and soluble IL-6 receptor after administration of an anti-IL-6 receptor antibody, tocilizumab, in patients with rheumatoid arthritis and Castleman disease. Blood. 2008;112(10):3959–64.CrossRefPubMed Nishimoto N, Terao K, Mima T, Nakahara H, Takagi N, Kakehi T. Mechanisms and pathologic significances in increase in serum interleukin-6 (IL-6) and soluble IL-6 receptor after administration of an anti-IL-6 receptor antibody, tocilizumab, in patients with rheumatoid arthritis and Castleman disease. Blood. 2008;112(10):3959–64.CrossRefPubMed
83.
Zurück zum Zitat Voorhees PM, Manges RF, Sonneveld P, Jagannath S, Somlo G, Krishnan A, et al. A phase 2 multicentre study of siltuximab, an anti-interleukin-6 monoclonal antibody, in patients with relapsed or refractory multiple myeloma. Br J Haematol. 2013;161(3):357–66. Voorhees PM, Manges RF, Sonneveld P, Jagannath S, Somlo G, Krishnan A, et al. A phase 2 multicentre study of siltuximab, an anti-interleukin-6 monoclonal antibody, in patients with relapsed or refractory multiple myeloma. Br J Haematol. 2013;161(3):357–66.
84.
Zurück zum Zitat Rossi JF, Negrier S, James ND, Kocak I, Hawkins R, Davis H, et al. A phase I/II study of siltuximab (CNTO 328), an anti-interleukin-6 monoclonal antibody, in metastatic renal cell cancer. Br J Cancer. 2010;103(8):1154–62. Rossi JF, Negrier S, James ND, Kocak I, Hawkins R, Davis H, et al. A phase I/II study of siltuximab (CNTO 328), an anti-interleukin-6 monoclonal antibody, in metastatic renal cell cancer. Br J Cancer. 2010;103(8):1154–62.
85.
Zurück zum Zitat Hunsucker SA, Magarotto V, Kuhn DJ, Kornblau SM, Wang M, Weber DM, et al. Blockade of interleukin-6 signalling with siltuximab enhances melphalan cytotoxicity in preclinical models of multiple myeloma. Br J Haematol. 2011;152(5):579–92. Hunsucker SA, Magarotto V, Kuhn DJ, Kornblau SM, Wang M, Weber DM, et al. Blockade of interleukin-6 signalling with siltuximab enhances melphalan cytotoxicity in preclinical models of multiple myeloma. Br J Haematol. 2011;152(5):579–92.
86.
Zurück zum Zitat Dorff TB, Goldman B, Pinski JK, Mack PC, Lara Jr PN, Van Veldhuizen Jr PJ, et al. Clinical and correlative results of SWOG S0354: a phase II trial of CNTO328 (siltuximab), a monoclonal antibody against interleukin-6, in chemotherapy-pretreated patients with castration-resistant prostate cancer. Clin Cancer Re. 2010;16(11):3028–34. Dorff TB, Goldman B, Pinski JK, Mack PC, Lara Jr PN, Van Veldhuizen Jr PJ, et al. Clinical and correlative results of SWOG S0354: a phase II trial of CNTO328 (siltuximab), a monoclonal antibody against interleukin-6, in chemotherapy-pretreated patients with castration-resistant prostate cancer. Clin Cancer Re. 2010;16(11):3028–34.
88.
Zurück zum Zitat Song L, Smith MA, Doshi P, Sasser K, Fulp W, Altiok S, et al. Antitumor efficacy of the anti-interleukin-6 (IL-6) antibody siltuximab in mouse xenograft models of lung cancer. J Thorac Oncol. 2014;9(7):974–82. Song L, Smith MA, Doshi P, Sasser K, Fulp W, Altiok S, et al. Antitumor efficacy of the anti-interleukin-6 (IL-6) antibody siltuximab in mouse xenograft models of lung cancer. J Thorac Oncol. 2014;9(7):974–82.
89.
Zurück zum Zitat Coward J, Kulbe H, Chakravarty P, Leader D, Vassileva V, Leinster DA, et al. Interleukin-6 as a therapeutic target in human ovarian cancer. Clin Cancer Res. 2011;17(18):6083–96. Coward J, Kulbe H, Chakravarty P, Leader D, Vassileva V, Leinster DA, et al. Interleukin-6 as a therapeutic target in human ovarian cancer. Clin Cancer Res. 2011;17(18):6083–96.
90.
Zurück zum Zitat Axel A, Casneuf T, King P, Alvarez J, Hall B, Sasser K. Abstract 3530: The role of IL-6 in ER + breast cancer and potential use for Siltuximab, an anti-IL-6 antibody, in ER + breast cancer treatment. Cancer Res. 2013;73(8 Supplement):3530–0. Axel A, Casneuf T, King P, Alvarez J, Hall B, Sasser K. Abstract 3530: The role of IL-6 in ER + breast cancer and potential use for Siltuximab, an anti-IL-6 antibody, in ER + breast cancer treatment. Cancer Res. 2013;73(8 Supplement):3530–0.
Metadaten
Titel
Il-6 signaling between ductal carcinoma in situ cells and carcinoma-associated fibroblasts mediates tumor cell growth and migration
verfasst von
Kingsley O. Osuala
Mansoureh Sameni
Seema Shah
Neha Aggarwal
Michelle L. Simonait
Omar E. Franco
Yan Hong
Simon W. Hayward
Fariba Behbod
Raymond R. Mattingly
Bonnie F. Sloane
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2015
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-015-1576-3

Weitere Artikel der Ausgabe 1/2015

BMC Cancer 1/2015 Zur Ausgabe

Hodgkin Lymphom: BrECADD-Regime übertrifft die Erwartungen

05.06.2024 ASCO 2024 Kongressbericht

Das Kombinationsregime BrECADD mit Brentuximab vedotin ermöglichte in der Studie HD21 beim fortgeschrittenen klassischen Hodgkin-Lymphom eine unerwartet hohe progressionsfreie Überlebensrate von 94,3% nach vier Jahren. Gleichzeitig war das Regime besser tolerabel als der bisherige Standard eBEACOPP.

Antikörper-Drug-Konjugat verdoppelt PFS bei Multiplem Myelom

05.06.2024 ASCO 2024 Nachrichten

Zwei Phase-3-Studien deuten auf erhebliche Vorteile des Antikörper-Wirkstoff-Konjugats Belantamab-Mafodotin bei vorbehandelten Personen mit Multiplem Myelom: Im Vergleich mit einer Standard-Tripeltherapie wurde das progressionsfreie Überleben teilweise mehr als verdoppelt.

Neuer TKI gegen CML: Höhere Wirksamkeit, seltener Nebenwirkungen

05.06.2024 Chronische myeloische Leukämie Nachrichten

Der Tyrosinkinasehemmer (TKI) Asciminib ist älteren Vertretern dieser Gruppe bei CML offenbar überlegen: Personen mit frisch diagnostizierter CML entwickelten damit in einer Phase-3-Studie häufiger eine gut molekulare Response, aber seltener ernste Nebenwirkungen.

Brustkrebs-Prävention wird neu gedacht

04.06.2024 ASCO 2024 Kongressbericht

Zurzeit untersuchen Forschende verschiedene neue Ansätze zur Prävention von Brustkrebs bei Personen mit hohem Risiko. Darunter Denosumab, die prophylaktische Bestrahlung der Brust – und Impfungen.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.