A Paracrine Role for IL6 in Prostate Cancer Patients: Lack of Production by Primary or Metastatic Tumor Cells.
Journal: 2016/July - Cancer immunology research
ISSN: 2326-6074
Abstract:
Correlative human studies suggest that the pleiotropic cytokine IL6 contributes to the development and/or progression of prostate cancer. However, the source of IL6 production in the prostate microenvironment in patients has yet to be determined. The cellular origin of IL6 in primary and metastatic prostate cancer was examined in formalin-fixed, paraffin-embedded tissues using a highly sensitive and specific chromogenic in situ hybridization (CISH) assay that underwent extensive analytical validation. Quantitative RT-PCR showed that benign prostate tissues often had higher expression of IL6 mRNA than matched tumor specimens. CISH analysis further indicated that both primary and metastatic prostate adenocarcinoma cells do not express IL6 mRNA. IL6 expression was highly heterogeneous across specimens and was nearly exclusively restricted to the prostate stromal compartment--including endothelial cells and macrophages, among other cell types. The number of IL6-expressing cells correlated positively with the presence of acute inflammation. In metastatic disease, tumor cells were negative in all lesions examined, and IL6 expression was restricted to endothelial cells within the vasculature of bone metastases. Finally, IL6 was not detected in any cells in soft tissue metastases. These data suggest that, in prostate cancer patients, paracrine rather than autocrine IL6 production is likely associated with any role for the cytokine in disease progression.
Relations:
Content
Citations
(10)
References
(35)
Grants
(153)
Diseases
(1)
Conditions
(2)
Chemicals
(1)
Genes
(1)
Organisms
(1)
Processes
(4)
Anatomy
(2)
Affiliates
(3)
Similar articles
Articles by the same authors
Discussion board
Cancer Immunol Res 3(10): 1175-1184

A Paracrine Role for IL6 in Prostate Cancer Patients: Lack of Production by Primary or Metastatic Tumor Cells

+2 authors

Introduction

Interleukin 6 (IL6) is a pleiotropic cytokine produced by an array of cell types and affects diverse physiologic processes including immune responses, hematopoiesis, and cellular proliferation and differentiation (1). Under normal conditions IL6 levels in cells are typically low, although a number of stimuli result in induction of IL6 expression and secretion. For example, during acute inflammatory responses to infections, cellular production of IL6 is essential to the induction of acute phase proteins. While the normal homeostatic response to inflammation is resolution and reversion of IL6 production to normal low levels, unrestrained production of IL6 drives chronic inflammation and increased systemic levels of IL6 have been associated with diseases such as autoimmune disorders, arthritis, hepatitis, inflammatory bowel disease, pancreatitis, and cancer (2).

Early evidence for a role for IL6 in advanced prostate cancer came from studies examining serum levels of IL6 in relation to metastatic or hormone-refractory prostate cancer (35). These data showed that serum levels of IL6 are significantly elevated in prostate cancer patients with hormone-refractory disease compared to normal controls or men with prostatitis, benign prostatic hyperplasia (BPH), and localized and recurrent disease (3). Likewise, serum IL6 levels were correlated to patients with clinically evident metastases (4) or with extent of bone metastasis (5). Subsequent studies have consistently shown that elevated systemic IL6 levels confer poor prognosis (68), and may also serve as a marker of prostate cancer morbidity, including cachexia (9, 10). A key clinical question is precisely when during disease development and progression IL6 is expressed and what specific cell types are responsible for its production (e.g., prostate tumor cells or another cellular source). A secondary question is whether the elevated systemic levels of this cytokine actually drive disease progression, or whether the elevated levels of IL6 are a surrogate for tumor burden.

Along these lines, multiple studies showed that some prostate cancer cell lines can secrete IL6 in vitro. The androgen-independent prostate cancer cell lines DU145 and PC3 have been shown to secrete IL6, whereas androgen-sensitive LNCaP cells do not (9, 11, 12). In primary prostate cancer, protein extracts prepared from prostate cancer tissues showed elevated IL6 levels compared to benign tissues in approximately 50% of cases when analyzed by enzyme-linked immunosorbent assay (ELISA) (12). Furthermore, studies using immunohistochemistry (IHC) to detect IL6 in prostate tissues have reported IL6 production by both benign and malignant prostate epithelium (13, 14). An additional study utilizing IHC to detect IL6 in a series of metastatic tissues from 26 prostate cancer patients reported that IL6 is produced in the majority of prostate cancer bone metastases and to a lesser extent in prostate cancer soft tissue metastases (15). Another series of studies proposed that IL6 along with a related member of the IL6 family of cytokines, oncostatin M (OSM), may activate androgen receptor (AR) in the absence of androgen, providing a potential mechanism whereby IL-6 contributes to recurrent prostate cancer growth following androgen deprivation therapy (reviewed in (16)). Cumulatively, these studies have led to the hypothesis that IL6 serves as an autocrine growth factor in both primary and metastatic prostate cancer (12, 13, 16).

Additional studies have suggested a role for paracrine IL6 signaling in prostate cancer progression. Recent work using a human prostate dissociation and tissue recombination system identified a role for paracrine expression of IL6 or OSM specifically in the stromal compartment in concert with cell-autonomous oncogenic events, such as PTEN loss of function, in the promotion of an aggressive prostate cancer phenotype (17). Another recent study identified a role for paracrine signaling from IL6 up-regulation in mesenchymal stem cells and promotion of adipogenesis and prostate cancer cell migration and invasion (18). Again, this study indicated a specific role for IL6 production from the stromal compartment in facilitating prostate cancer progression (18).

Cumulatively, the evidence to date indicates that IL6 may act as a key mediator in several steps in prostate carcinogenesis including initiation, progression, metastases, and the development of castration resistance and/or resistance to chemotherapy. What is less well understood is what cell type(s) are responsible for production of the cytokine in the tumor microenvironment in patients, and by extension, whether IL6 in prostate cancer patients functions through autocrine or paracrine mechanisms.

Materials and Methods

Patient population and clinical samples

All specimens were acquired under Institutional Review Board (IRB)-approved protocols at the respective institutions. RNA samples from matched tumor and benign tissues were obtained from 10 radical prostatectomy specimens using the standard operating procedure (SOP) protocols for the Prostate Cancer Biorepository Network (PCBN) as previously described (19). Each case consisted of fresh frozen tumor and matched benign peripheral zone tissues obtained at radical prostatectomy. For RNA isolation, tissues containing cancer were dissected such that they contained at least 70–90% tumor cells. Recently collected formalin-fixed paraffin-embedded (FFPE) primary clinical prostate cancer tissues (<1 year old) were obtained from 21 prostatectomy specimens in addition to 12 biopsy or autopsy metastatic tissue samples from 9 cases at Johns Hopkins Hospital and 20 bone metastatic tissue samples from 10 cases at the University of Washington Medical Center for use in chromogenic in situ hybridization (CISH) assays. One block containing the highest grade/index cancer and adjacent benign tissue was chosen for CISH analysis from each prostatectomy case. The clinical and pathologic details of the patient samples are listed in Supplementary Table S1. Tissue microarrays (TMA) containing metastatic tissues (bone and soft tissue metastases) from 21 cases (University of Washington Medical Center) and 15 cases (Johns Hopkins Hospital) were used in IL6 IHC experiments.

Cell lines

LNCaP, VCaP and CWR22Rv1 were obtained from the American Type Culture Collection (ATCC). PC3, DU145, MCF7, and NCI-H460 cells were obtained from the NCI-Frederick. PrEC and PrSC cells were obtained from Lonza (Basel, Switzerland). LAPC4, RWPE-1, and C4-2B cells were obtained from J.T. Isaacs (Johns Hopkins University) and LNCaP-abl cells were obtained from Z. Culig (University of Innsbruck). All cell lines used were authenticated via short tandem repeat (STR) profiling of 9 genomic loci with the Powerplex 1.2 system (Promega) before use.

Quantitative real-time reverse transcription PCR (q-RT-PCR)

RNA was treated with DNase I (RNase-free, Ambion) followed by cDNA synthesis using the SuperScript First Strand Synthesis System for RT-PCR (Invitrogen) following standard protocol for ‘First-Strand Synthesis Using Random Primers’. Quantitative PCR was performed with SYBR Green Supermix (Bio-Rad) and 0.4 µM IL6 primers (IL6-F 5’-GGTACATCCTCGACGGCATCT-3’ and IL6-R 5’-GTGCCTCTTTGCTGCTTTCAC-3’) or 1.0 µM GAPDH primers (GAPDH-F 5’-CGCTCTCTGCTCCTCCTGTT-3’ and GAPDH-R 5’-CCATGGTGTCTGAGCGATGT-3’) in a real-time detection system. PCR conditions were as follows: 2 min at 94 °C, 40 cycles of 30 sec at 94 °C, 30 sec at 60 °C, and 30 sec at 72 °C, followed by a melt curve analysis. GAPDH was used as a housekeeping gene for normalization. The fold-differences in expression levels of IL6 in tumor samples were determined using the 2T method, relative to GAPDH and to the matched benign tissue.

Chromogenic in situ hybridization (CISH)

CISH was performed using the RNAscope® 2.0 FFPE Brown Reagent Kit or RNAscope® 2-plex assay kit (Advanced Cell Diagnostics, Inc.). Briefly, FFPE tissues were first baked at 60°C for 1 hr followed by deparaffinization in two changes of 100% xylene for 5 min each and two changes of 100% alcohol for 3 min each. Next, the slides were treated with endogenous peroxidase blocking pretreatment reagent for 10 min at room temperature. The slides were then added to boiling buffer for 30 min at 99–104°C in a water bath and then treated with protease digestion buffer for 30 min at 40°C. The slides were incubated with a custom RNAscope target probe designed against IL6 mRNA (probe region 27–876, NCBI reference sequence Accession #{"type":"entrez-nucleotide","attrs":{"text":"NM_000600.3","term_id":"224831235","term_text":"NM_000600.3"}}NM_000600.3) or peptidyl prolyl isomerase B (PPIB), also known as cyclophilin B, as a positive control mRNA (probe region 139–989, NCBI reference sequence Accession #{"type":"entrez-nucleotide","attrs":{"text":"NM_000942.4","term_id":"44890060"}}NM_000942.4) for 2 hr at 40°C, followed by signal amplification. DAB was used for colorimetric detection for 10 min at room temperature.

Immunohistochemistry (IHC)

IHC was performed using the Power Vision+ Poly-HRP IHC kit (Leica Biosystems). Slides were steamed for 45 min in antigen retrieval solution (Dako #S1700) and incubated with rabbit polyclonal anti-IL6 antibody (#6672; Abcam, lot #GR106735-5 at 1:1000 dilution) for 45 minutes at room temperature. Poly-HRP-conjugated anti-rabbit IgG antibody was used as secondary antibody. Staining was visualized using 3,3′-diaminobenzidine (Sigma), and slides were counterstained with hematoxylin.

Western blot

Cells were lysed in radioimmunoprecipitation assay (RIPA) buffer containing protease inhibitors (Sigma) and phosphatase inhibitors (Cell Signaling). Lysates were then centrifuged at 14,000 X g for 10 min at 4°C. Proteins were electrophoresed and transferred to nitrocellulose membranes for immunoblotting. Membranes were probed with anti-IL6 antibody (#6672 1:500 dilution; Abcam, lot #GR106735-5). The blots were visualized using the Odyssey Infrared Imaging System (LI-COR Biosciences). Recombinant mouse IL6 (R&amp;D Systems) was used as a positive control for Western blot.

Controls for CISH, Western blot, and IHC

MCF7 (breast cancer cell line) cells were transfected with the IL6 cDNA clone expression vector (Origene, SC125236) using lipofectamine (Life Technologies). NCI-H460 cells were treated with monensin (Golgi-Stop™, BD Biosciences) at a dilution of 1:1000 for 4 hrs.

Patient population and clinical samples

All specimens were acquired under Institutional Review Board (IRB)-approved protocols at the respective institutions. RNA samples from matched tumor and benign tissues were obtained from 10 radical prostatectomy specimens using the standard operating procedure (SOP) protocols for the Prostate Cancer Biorepository Network (PCBN) as previously described (19). Each case consisted of fresh frozen tumor and matched benign peripheral zone tissues obtained at radical prostatectomy. For RNA isolation, tissues containing cancer were dissected such that they contained at least 70–90% tumor cells. Recently collected formalin-fixed paraffin-embedded (FFPE) primary clinical prostate cancer tissues (<1 year old) were obtained from 21 prostatectomy specimens in addition to 12 biopsy or autopsy metastatic tissue samples from 9 cases at Johns Hopkins Hospital and 20 bone metastatic tissue samples from 10 cases at the University of Washington Medical Center for use in chromogenic in situ hybridization (CISH) assays. One block containing the highest grade/index cancer and adjacent benign tissue was chosen for CISH analysis from each prostatectomy case. The clinical and pathologic details of the patient samples are listed in Supplementary Table S1. Tissue microarrays (TMA) containing metastatic tissues (bone and soft tissue metastases) from 21 cases (University of Washington Medical Center) and 15 cases (Johns Hopkins Hospital) were used in IL6 IHC experiments.

Cell lines

LNCaP, VCaP and CWR22Rv1 were obtained from the American Type Culture Collection (ATCC). PC3, DU145, MCF7, and NCI-H460 cells were obtained from the NCI-Frederick. PrEC and PrSC cells were obtained from Lonza (Basel, Switzerland). LAPC4, RWPE-1, and C4-2B cells were obtained from J.T. Isaacs (Johns Hopkins University) and LNCaP-abl cells were obtained from Z. Culig (University of Innsbruck). All cell lines used were authenticated via short tandem repeat (STR) profiling of 9 genomic loci with the Powerplex 1.2 system (Promega) before use.

Quantitative real-time reverse transcription PCR (q-RT-PCR)

RNA was treated with DNase I (RNase-free, Ambion) followed by cDNA synthesis using the SuperScript First Strand Synthesis System for RT-PCR (Invitrogen) following standard protocol for ‘First-Strand Synthesis Using Random Primers’. Quantitative PCR was performed with SYBR Green Supermix (Bio-Rad) and 0.4 µM IL6 primers (IL6-F 5’-GGTACATCCTCGACGGCATCT-3’ and IL6-R 5’-GTGCCTCTTTGCTGCTTTCAC-3’) or 1.0 µM GAPDH primers (GAPDH-F 5’-CGCTCTCTGCTCCTCCTGTT-3’ and GAPDH-R 5’-CCATGGTGTCTGAGCGATGT-3’) in a real-time detection system. PCR conditions were as follows: 2 min at 94 °C, 40 cycles of 30 sec at 94 °C, 30 sec at 60 °C, and 30 sec at 72 °C, followed by a melt curve analysis. GAPDH was used as a housekeeping gene for normalization. The fold-differences in expression levels of IL6 in tumor samples were determined using the 2T method, relative to GAPDH and to the matched benign tissue.

Chromogenic in situ hybridization (CISH)

CISH was performed using the RNAscope® 2.0 FFPE Brown Reagent Kit or RNAscope® 2-plex assay kit (Advanced Cell Diagnostics, Inc.). Briefly, FFPE tissues were first baked at 60°C for 1 hr followed by deparaffinization in two changes of 100% xylene for 5 min each and two changes of 100% alcohol for 3 min each. Next, the slides were treated with endogenous peroxidase blocking pretreatment reagent for 10 min at room temperature. The slides were then added to boiling buffer for 30 min at 99–104°C in a water bath and then treated with protease digestion buffer for 30 min at 40°C. The slides were incubated with a custom RNAscope target probe designed against IL6 mRNA (probe region 27–876, NCBI reference sequence Accession #{"type":"entrez-nucleotide","attrs":{"text":"NM_000600.3","term_id":"224831235","term_text":"NM_000600.3"}}NM_000600.3) or peptidyl prolyl isomerase B (PPIB), also known as cyclophilin B, as a positive control mRNA (probe region 139–989, NCBI reference sequence Accession #{"type":"entrez-nucleotide","attrs":{"text":"NM_000942.4","term_id":"44890060"}}NM_000942.4) for 2 hr at 40°C, followed by signal amplification. DAB was used for colorimetric detection for 10 min at room temperature.

Immunohistochemistry (IHC)

IHC was performed using the Power Vision+ Poly-HRP IHC kit (Leica Biosystems). Slides were steamed for 45 min in antigen retrieval solution (Dako #S1700) and incubated with rabbit polyclonal anti-IL6 antibody (#6672; Abcam, lot #GR106735-5 at 1:1000 dilution) for 45 minutes at room temperature. Poly-HRP-conjugated anti-rabbit IgG antibody was used as secondary antibody. Staining was visualized using 3,3′-diaminobenzidine (Sigma), and slides were counterstained with hematoxylin.

Western blot

Cells were lysed in radioimmunoprecipitation assay (RIPA) buffer containing protease inhibitors (Sigma) and phosphatase inhibitors (Cell Signaling). Lysates were then centrifuged at 14,000 X g for 10 min at 4°C. Proteins were electrophoresed and transferred to nitrocellulose membranes for immunoblotting. Membranes were probed with anti-IL6 antibody (#6672 1:500 dilution; Abcam, lot #GR106735-5). The blots were visualized using the Odyssey Infrared Imaging System (LI-COR Biosciences). Recombinant mouse IL6 (R&amp;D Systems) was used as a positive control for Western blot.

Controls for CISH, Western blot, and IHC

MCF7 (breast cancer cell line) cells were transfected with the IL6 cDNA clone expression vector (Origene, SC125236) using lipofectamine (Life Technologies). NCI-H460 cells were treated with monensin (Golgi-Stop™, BD Biosciences) at a dilution of 1:1000 for 4 hrs.

Results

Variable expression levels of IL6 transcript are present in primary prostate adenocarcinoma when compared to matched benign tissues as assessed by q-RT-PCR

To determine if IL6 expression levels in primary prostate cancer are associated with prognostic factors, we obtained a series of RNA samples from fresh-frozen cancer and matched benign tissues from 10 radical prostatectomy specimens. This series was enriched for cases of higher grade (Gleason ≥ 7) cancer (Supplementary Table S1). IL6 message levels were not significantly elevated in tumors compared to matched benign tissues, as only 3 of 10 prostatectomy samples were observed to have higher expression of IL6 in tumor compared to benign tissue (Fig. 1). Conversely, there were more cases that showed higher levels of IL6 in the benign regions than in those containing cancer (Fig. 1). Based on this apparent discrepancy with published data (12, 13), we queried the Oncomine database (20) for datasets of prostate carcinoma vs. normal tissues and an analysis of 17 such datasets showed only one dataset with a greater than 2-fold increase in cancer versus benign tissues and a p-value ≤ 0.05 (Supplementary Table S2). Due to our observation of a low frequency of IL6 overexpression in tumor compared to benign tissues as well as the elevated IL6 levels observed in benign tissues in our q-RT-PCR studies (Fig. 1B) we next sought to determine the cellular origin of IL6 in prostate specimens.

An external file that holds a picture, illustration, etc.
Object name is nihms698369f1.jpg
IL6 mRNA expression in benign and malignant prostate tissues as assessed by q-RT-PCR

RNA extracts were prepared from matched tumor and benign tissues from radical prostatectomy specimens. IL6 mRNA expression was then determined by q-RT-PCR. (A) Log2 IL6 mRNA expression in tumor relative to matched benign tissue in 10 prostatectomy specimens. IL6 mRNA expression levels were normalized to GAPDH and then tumor was compared to benign using the 2T method followed by log2 transformation. (B) Log2 relative IL6 mRNA expression in benign samples versus tumor samples.

Development of a highly sensitive and specific IL6 chromogenic in situ hybridization (CISH) assay

For the development of an IL6 CISH assay, we used the highly sensitive RNAscope® 2.0 assay from Advanced Cell Diagnostics (ACD). This assay consists of a hybridization probe set complimentary to a length of the protein coding region of the IL6 mRNA (see methods) in which the hybridization event is subjected to signal amplification and chromogenic detection. To establish the specificity of the CISH approach, we transfected MCF-7 cells (which do not express IL6 mRNA) with an IL6 cDNA clone expression vector (Origene, SC125236). As shown in Fig. 2A, the transfected cells stained strongly positive using the CISH assay and thus served as a genetically defined positive control. To further verify the specificity of the probe set, we next quantified IL6 mRNA levels in a panel of prostate cancer cell lines (LAPC4, CWR22Rv1, PC3, LNCaP, C4-2B, VCaP, DU145, and LNCaP-abl), benign prostate cell lines (PrSC, PrEC) and an HPV-transformed prostate cell line (RWPE-1) via q-RT-PCR. Providing further evidence for the specificity of the CISH assay, we found that there was complete concordance between the two assays; i.e. all lines that were positive for IL6 by q-RT-PCR were positive by CISH for IL6 mRNA expression (Fig. 2B, C). Of note, although previous studies reported that the PC3 cell line secretes IL6 (9, 11, 12), we did not find the line to be positive for IL6 mRNA expression in the present study (Fig. 2B, C). We hypothesize that this could be due to differences in cell culture conditions for this highly inducible cytokine. The CISH assay has been reported to have a detection limit of a single mRNA molecule (21).

An external file that holds a picture, illustration, etc.
Object name is nihms698369f2.jpg
Validation of IL6 chromogenic in situ hybridization (CISH) assay

(A) IL6 transfected (positive control) or non-transfected (negative control) FFPE MCF7 cells assayed by IL6 CISH. (B) IL6 q-RT-PCR on 11 prostate cell lines. IL6 mRNA expression levels were normalized to GAPDH and then to PrEC cell expression level using the 2T method. (C) IL6 CISH was performed on the same 11 prostate cell lines and representative examples are shown. Brown staining (arrowheads) represent positive IL6 mRNA expression. Single dots in the nucleus of cells are interpreted as the genomic copy of the gene (PC3 cells, arrow). Both q-RT-PCR and CISH analyses indicated that three of the prostate cell lines (PrSC, RWPE-1, DU145) were positive for IL6 mRNA, indicating complete concordance between the two assays.

Prostate adenocarcinoma cells in prostatectomy specimens do not express IL6 mRNA

We next utilized the IL6 CISH mRNA assay on FFPE tissues from a series of 21 radical prostatectomy specimens of varying Gleason grades and tumor stages (see Supplementary Table S1). In each case, we examined full tissue sections containing the highest grade/index cancer and adjacent benign tissue. We verified RNA integrity in the tissue sections used in this study by using a positive control probe set against peptidyl prolyl isomerase B (PPIB) on adjacent sections. PPIB hybridization signals demonstrated expression in virtually all cells present on all slides that were used in this study (Fig. 3A), showing that the RNA in these specimens was intact. We did not detect positive IL6 mRNA expression in tumor cells in any of the cases examined, regardless of tumor grade (Fig. 3A, Table 1). Instead, IL6 mRNA expression in areas containing prostate cancer was restricted to cells within the stromal compartment of the tumor and primarily in tumor-associated endothelial cells (Fig. 3B). The detection of IL6 mRNA in stromal cells, in combination with positive control staining of transfected MCF-7 cells (Fig. 2A) and prostate cell lines (Fig. 2C), confirms that the IL6 CISH probe was functioning as expected and that prostate adenocarcinoma cells in primary tumors do not express IL6 mRNA.

An external file that holds a picture, illustration, etc.
Object name is nihms698369f3.jpg
IL6 mRNA is not detected in prostate adenocarcinoma cells in primary tumors

Twenty one prostatectomy specimens were selected for IL6 CISH (see Table 1). PPIB (housekeeping gene) is used as a positive control for the CISH assay. (A) Shown are representative examples of low grade (Gleason pattern 3) and higher grade (Gleason patterns 4 and 3) prostate cancer with positive staining for PPIB (positive control) and negative staining for IL6. (B) IL6-positive cells in the endothelium (arrow) area of cancer.

Table 1

Assessment of Levels and Distribution of IL-6 mRNA Expression in Prostatectomy Tissues

PATIENTIL-6 EPITHELIUMIL-6 STROMAAcute
Inflammation
Present?

Patient
#
Gleason
Score
Clinical
Stage
TumorNormalAtrophyNon-
inflamed
Inflamed
13+3=6T2N0MX--++++++++Yes
23+4=7T2N0MX----+++Yes
34+5=9T2N0MX--+--No
43+4=7T2N0MX--+-+No
53+3=6T2N0MX----+No
63+4=7**T2N0MX----++No
73+3=6*T2N0MX--++-++No
83+4=7T2N0MX----++++Yes
95+4=9T2N0MX----++No
103+3=6*T2N0MX-----No
113+4=7**T2N0MX-----No
123+4=7T2N0MX--+-++No
133+4=7T3AN0MX-----No
143+4=7T2N0MX--++-++No
153+3=6T2N0MX--+-++No
163+4=7T2N0MX--+++++++++Yes
173+3=6T2N0MX----+++Yes
184+3=7**T3AN0MX--+-+No
193+4=7T3AN0MX-----No
204+3=7T3BN0MX--++-+No
213+4=7**T2N0MX--+-++No

- no cells positive for IL6 expression; + areas with 5–10 positive cells per 20X field; ++ areas with 11–25 positive cells per 20X field; +++ areas with 26–50 positive cells per 20X field; ++++ areas of >50 positive cells per 20X field.

Tertiary pattern 4;
Tertiary pattern 5.

IL6 mRNA expression is highly up-regulated in areas of acute inflammation and prostatic atrophy

As shown in Table 1, an assessment of the distribution of IL6 mRNA expression in the prostatectomy tissues as analyzed by IL6 CISH demonstrated that, overall, the expression was highly heterogeneous from case-to-case and by region within a given case. As shown in Fig. 4A–B, the number of IL6-positive cells was highly increased in the stroma in areas of acute inflammation (as evidenced by accumulation of neutrophils within glandular lumens), although the neutrophils were not positive. The cases with the highest numbers of IL6 mRNA expressing cells were those in which acute inflammation was present (see Table 1). Increases in IL6-positive cells were also seen in areas of proliferative inflammatory atrophy (PIA, Fig. 4C–D). Positive cells in areas of prostatic atrophy were generally confined to the stroma surrounding atrophy. In some instances, positive IL6 mRNA expression was observed in select epithelial cells in atrophic glands (Table 1, Fig. 4D). Morphologically these appeared to be epithelial cells; however we did not perform a double label with an epithelial cell marker. In a number of cases the IL6 mRNA-expressing cells could be identified as endothelial cells lining small blood vessels (presumably venules or lymphatics, Fig. 4C) as well as prostate smooth muscle cells. Other positive staining cells had an appearance consistent with macrophages. To more definitively identify IL6 mRNA-expressing cells, we performed co-staining (2-plex) for IL6 and either CD68 (a macrophage marker) or CD31 (an endothelial cell marker). The results of this 2-plex staining clearly indicated that some of the IL6-positive cells are CD68-positive macrophages and some are CD31-positive endothelial cells (Supplementary Fig. S1). Interestingly, the majority of IL6-positive cells in areas of acute inflammation did not co-stain for CD68 or CD31, suggesting that most of the IL6 mRNA-expressing cells in these areas are of a different stromal cell type, such as fibroblasts, smooth muscle cells, other types of inflammatory cells, etc.

An external file that holds a picture, illustration, etc.
Object name is nihms698369f4.jpg
IL6 mRNA expression is nearly exclusively restricted to the prostate stromal compartment

(A,B) IL6 mRNA-expressing cells (brown staining) are highly enriched in the stroma in areas of acute inflammation as indicated by the presence of neutrophils in glandular lumens (red arrows). (C) IL6-positive cells in the stroma (arrows) and endothelium (arrowheads) surrounding prostate atrophy. (D) Positive epithelial staining was rare and restricted to prostatic atrophy (arrowheads).

Prostate adenocarcinoma cells in metastatic lesions do not express IL6 mRNA

Previous studies suggested that production of IL6 by metastatic prostate cancer cells may facilitate invasion and metastasis to bone and/or promote resistance to prostate cancer therapies (15, 22). Therefore, we next examined IL6 mRNA expression in biopsy samples from patients with castration-resistant metastatic prostate cancer (samples evaluated included 4 lymph node specimens and 3 liver biopsy samples) and 25 autopsy samples which included metastases to the liver, lung, bone, and lymph node (see Supplementary Table S1). Consistent with data from primary prostatectomy specimens, IL6 mRNA expression was not observed in prostate cancer cells in any of the metastatic tissues examined. Also consistent with the primary prostatectomy specimens, the metastatic tissues all showed strong hybridization signals with PPIB as a positive control (Fig. 5A), confirming RNA integrity. Interestingly, IL6 mRNA staining was observed in endothelial cells in blood vessels in 9 of 21 bone metastases (42.9%, Fig. 5B, Supplementary Fig. S2, Supplementary Table S3) but not in any of the 11 soft tissue metastases analyzed (p = 0.013, Fisher’s exact test).

An external file that holds a picture, illustration, etc.
Object name is nihms698369f5.jpg
IL6 mRNA is not detected in metastatic prostate cancer cells

In this study, metastatic prostate cancer in lymph node and liver biopsy samples and autopsy samples (liver, lung, bone, and lymph node) were assayed by IL6 CISH. No IL6-positive mRNA expression was observed in prostate cancer cells in any of the metastatic tissues. (A) Shown are representative examples of lymph node biopsy and autopsy liver, lung, and bone metastases with positive staining for PPIB (positive control) and negative staining for IL6. (B) An example of IL6 mRNA-positive blood vessels (arrowheads) and IL6-negative tumor cells (arrows) in bone metastases.

IL6 IHC is only successful when Golgi export is blocked

Finally, we performed a series of analyses to examine IL6 protein production in cell lines and tissues using IHC. To perform these studies, we prepared FFPE blocks from a lung cancer cell line (NCI-H460) that was strongly positive for IL6 mRNA as assessed by CISH (Fig. 6A) and produces physiologic levels (e.g., not transfection levels) of IL6 protein as assessed by Western blot (Supplementary Fig. S3). Of interest, there appears to be heterogeneity in cells that are positive for IL6 expression (i.e., not all cultured cells appear to be positive) via CISH (Fig. 6A) and this was consistent with what we observed for the IL6 mRNA-positive prostate cell lines as well (Fig. 2C). IHC on the NCI-H460 cells using the polyclonal anti-IL6 antibody (#6672; Abcam, lot #GR106735-5) showed no detectable signal above background levels (as established by negative control cell lines, Fig. 6 B,C). In one sense, these data are not particularly surprising, since IL6 is a secreted protein and secreted proteins are generally only detectable by flow cytometry analysis when protein export from the Golgi apparatus is blocked using protein transport inhibitors such as brefeldin A or monensin (23). As such, we next treated NCI-H460 cells with monensin prior to formalin fixation and preparation of FFPE blocks for use with IHC. IHC of “stopped” cell lines treated with monensin showed that NCI-H460 cells now stained positive for IL6 protein (Fig. 6D). We then performed IL6 IHC on a series of prostatectomy specimens including cases that were strongly positive in areas of acute inflammation with IL6 CISH (Fig. 6E, F). We did not detect IL6 protein above background levels in the acutely inflamed areas, either in epithelial or stromal cells using IHC, and likewise we did not observe IL6 protein in prostate tumor cells using IHC (Fig. 6G). Similarly, we did not detect any IL6 protein in tumor cells via IHC on metastatic lesions from standard slides from cases used for CISH assays or in two separate metastatic prostate cancer TMAs, including one of bone metastases (Fig. 6H). The IHC signal in metastases was restricted to extracellular spaces between tumor cell nests (Fig. 6H) and no specific signal was detectable above background levels in the tumor cells in any of the cases. The lack of positive signal for IL6 protein using IHC in areas that were positive for IL6 mRNA using CISH is likely not due to lack of IL6 protein production by the IL6 mRNA-positive cells. Rather, these results, coupled with our experiments using monensin in the NCI-H460 cell line, suggest that detection of this particular cytokine in tissue sections using IHC could be severely limited without being able to block protein export prior to fixation.

An external file that holds a picture, illustration, etc.
Object name is nihms698369f6.jpg
A requirement for protein transport inhibition for IL6 IHC

(A) IL6 CISH performed on the IL6-positive cell line NCI-H460. IL6 IHC performed on (B) PC3 cells (IL6 negative as assessed by q-RT-PCR and CISH) and (C) NCI-H460 cells. Marked difference in IHC results when NCI-H460 cells are treated with monensin prior to fixation (D). Arrows point to IL6-positive cells. The same prostatectomy case containing acute inflammation assayed for IL6 by CISH (E) and IHC (F). Example of IL6 IHC on primary prostate cancer (G) and prostate cancer bone metastasis (H). All tumor cells were negative in all samples analyzed. Positive staining observed in extracellular spaces (arrows in H) was considered to be non-specific.

To further demonstrate the sensitivity of the CISH assay over IHC, we used qPCR to generate a standard curve using estimated copies of the IL6 cDNA vector (Supplementary Fig. S4A). q-RT-PCR was then performed with RNA extracted from a known number of cells of the DU145 cell line (the lowest positive IL6-expressing cell line by q-RT-PCR/CISH in Fig. 2 B,C) and PC3 (determined to be negative in our hands by CISH). Comparison of the results from DU145 and PC3 to the standard curve indicated values of approximately 2 copies of IL6 mRNA per cell in the DU145 cell line and 0.017 copies per cell for PC3 (roughly 1 copy of IL6 mRNA per 59 cells). Cells from the same flasks of DU145 and PC3 used for RNA extraction were made into FFPE cell blocks. CISH analysis revealed (as also seen in Fig. 2C) positive detection of IL6 mRNA within the cytoplasm of DU145 cells (Supplementary Fig. S4B). Conversely, in PC3 cells we see no cytoplasmic staining and generally one or two single dots in the nucleus. Since we have observed such signals at times in cells that have no corresponding mRNA (A.M. De Marzo, Q. Zheng, A.K. Meeker unpublished results), and since it is clear that the CISH assay can at times detect genomic DNA as one or two dots in the nucleus (21), we do not consider 1–2 dots in the nucleus to be a bona fide positive signal and consider it below the limit of detection. IHC on adjacent slides were negative for IL6 protein (Supplementary Fig. S4B). We do not interpret this to mean that IL6 mRNA-expressing cells do not make IL6 protein, but that the IL6 protein cannot be detected using IHC without protein transport inhibition and, therefore, the CISH assay is more sensitive than our standard IHC assay.

Variable expression levels of IL6 transcript are present in primary prostate adenocarcinoma when compared to matched benign tissues as assessed by q-RT-PCR

To determine if IL6 expression levels in primary prostate cancer are associated with prognostic factors, we obtained a series of RNA samples from fresh-frozen cancer and matched benign tissues from 10 radical prostatectomy specimens. This series was enriched for cases of higher grade (Gleason ≥ 7) cancer (Supplementary Table S1). IL6 message levels were not significantly elevated in tumors compared to matched benign tissues, as only 3 of 10 prostatectomy samples were observed to have higher expression of IL6 in tumor compared to benign tissue (Fig. 1). Conversely, there were more cases that showed higher levels of IL6 in the benign regions than in those containing cancer (Fig. 1). Based on this apparent discrepancy with published data (12, 13), we queried the Oncomine database (20) for datasets of prostate carcinoma vs. normal tissues and an analysis of 17 such datasets showed only one dataset with a greater than 2-fold increase in cancer versus benign tissues and a p-value ≤ 0.05 (Supplementary Table S2). Due to our observation of a low frequency of IL6 overexpression in tumor compared to benign tissues as well as the elevated IL6 levels observed in benign tissues in our q-RT-PCR studies (Fig. 1B) we next sought to determine the cellular origin of IL6 in prostate specimens.

An external file that holds a picture, illustration, etc.
Object name is nihms698369f1.jpg
IL6 mRNA expression in benign and malignant prostate tissues as assessed by q-RT-PCR

RNA extracts were prepared from matched tumor and benign tissues from radical prostatectomy specimens. IL6 mRNA expression was then determined by q-RT-PCR. (A) Log2 IL6 mRNA expression in tumor relative to matched benign tissue in 10 prostatectomy specimens. IL6 mRNA expression levels were normalized to GAPDH and then tumor was compared to benign using the 2T method followed by log2 transformation. (B) Log2 relative IL6 mRNA expression in benign samples versus tumor samples.

Development of a highly sensitive and specific IL6 chromogenic in situ hybridization (CISH) assay

For the development of an IL6 CISH assay, we used the highly sensitive RNAscope® 2.0 assay from Advanced Cell Diagnostics (ACD). This assay consists of a hybridization probe set complimentary to a length of the protein coding region of the IL6 mRNA (see methods) in which the hybridization event is subjected to signal amplification and chromogenic detection. To establish the specificity of the CISH approach, we transfected MCF-7 cells (which do not express IL6 mRNA) with an IL6 cDNA clone expression vector (Origene, SC125236). As shown in Fig. 2A, the transfected cells stained strongly positive using the CISH assay and thus served as a genetically defined positive control. To further verify the specificity of the probe set, we next quantified IL6 mRNA levels in a panel of prostate cancer cell lines (LAPC4, CWR22Rv1, PC3, LNCaP, C4-2B, VCaP, DU145, and LNCaP-abl), benign prostate cell lines (PrSC, PrEC) and an HPV-transformed prostate cell line (RWPE-1) via q-RT-PCR. Providing further evidence for the specificity of the CISH assay, we found that there was complete concordance between the two assays; i.e. all lines that were positive for IL6 by q-RT-PCR were positive by CISH for IL6 mRNA expression (Fig. 2B, C). Of note, although previous studies reported that the PC3 cell line secretes IL6 (9, 11, 12), we did not find the line to be positive for IL6 mRNA expression in the present study (Fig. 2B, C). We hypothesize that this could be due to differences in cell culture conditions for this highly inducible cytokine. The CISH assay has been reported to have a detection limit of a single mRNA molecule (21).

An external file that holds a picture, illustration, etc.
Object name is nihms698369f2.jpg
Validation of IL6 chromogenic in situ hybridization (CISH) assay

(A) IL6 transfected (positive control) or non-transfected (negative control) FFPE MCF7 cells assayed by IL6 CISH. (B) IL6 q-RT-PCR on 11 prostate cell lines. IL6 mRNA expression levels were normalized to GAPDH and then to PrEC cell expression level using the 2T method. (C) IL6 CISH was performed on the same 11 prostate cell lines and representative examples are shown. Brown staining (arrowheads) represent positive IL6 mRNA expression. Single dots in the nucleus of cells are interpreted as the genomic copy of the gene (PC3 cells, arrow). Both q-RT-PCR and CISH analyses indicated that three of the prostate cell lines (PrSC, RWPE-1, DU145) were positive for IL6 mRNA, indicating complete concordance between the two assays.

Prostate adenocarcinoma cells in prostatectomy specimens do not express IL6 mRNA

We next utilized the IL6 CISH mRNA assay on FFPE tissues from a series of 21 radical prostatectomy specimens of varying Gleason grades and tumor stages (see Supplementary Table S1). In each case, we examined full tissue sections containing the highest grade/index cancer and adjacent benign tissue. We verified RNA integrity in the tissue sections used in this study by using a positive control probe set against peptidyl prolyl isomerase B (PPIB) on adjacent sections. PPIB hybridization signals demonstrated expression in virtually all cells present on all slides that were used in this study (Fig. 3A), showing that the RNA in these specimens was intact. We did not detect positive IL6 mRNA expression in tumor cells in any of the cases examined, regardless of tumor grade (Fig. 3A, Table 1). Instead, IL6 mRNA expression in areas containing prostate cancer was restricted to cells within the stromal compartment of the tumor and primarily in tumor-associated endothelial cells (Fig. 3B). The detection of IL6 mRNA in stromal cells, in combination with positive control staining of transfected MCF-7 cells (Fig. 2A) and prostate cell lines (Fig. 2C), confirms that the IL6 CISH probe was functioning as expected and that prostate adenocarcinoma cells in primary tumors do not express IL6 mRNA.

An external file that holds a picture, illustration, etc.
Object name is nihms698369f3.jpg
IL6 mRNA is not detected in prostate adenocarcinoma cells in primary tumors

Twenty one prostatectomy specimens were selected for IL6 CISH (see Table 1). PPIB (housekeeping gene) is used as a positive control for the CISH assay. (A) Shown are representative examples of low grade (Gleason pattern 3) and higher grade (Gleason patterns 4 and 3) prostate cancer with positive staining for PPIB (positive control) and negative staining for IL6. (B) IL6-positive cells in the endothelium (arrow) area of cancer.

Table 1

Assessment of Levels and Distribution of IL-6 mRNA Expression in Prostatectomy Tissues

PATIENTIL-6 EPITHELIUMIL-6 STROMAAcute
Inflammation
Present?

Patient
#
Gleason
Score
Clinical
Stage
TumorNormalAtrophyNon-
inflamed
Inflamed
13+3=6T2N0MX--++++++++Yes
23+4=7T2N0MX----+++Yes
34+5=9T2N0MX--+--No
43+4=7T2N0MX--+-+No
53+3=6T2N0MX----+No
63+4=7**T2N0MX----++No
73+3=6*T2N0MX--++-++No
83+4=7T2N0MX----++++Yes
95+4=9T2N0MX----++No
103+3=6*T2N0MX-----No
113+4=7**T2N0MX-----No
123+4=7T2N0MX--+-++No
133+4=7T3AN0MX-----No
143+4=7T2N0MX--++-++No
153+3=6T2N0MX--+-++No
163+4=7T2N0MX--+++++++++Yes
173+3=6T2N0MX----+++Yes
184+3=7**T3AN0MX--+-+No
193+4=7T3AN0MX-----No
204+3=7T3BN0MX--++-+No
213+4=7**T2N0MX--+-++No

- no cells positive for IL6 expression; + areas with 5–10 positive cells per 20X field; ++ areas with 11–25 positive cells per 20X field; +++ areas with 26–50 positive cells per 20X field; ++++ areas of >50 positive cells per 20X field.

Tertiary pattern 4;
Tertiary pattern 5.

IL6 mRNA expression is highly up-regulated in areas of acute inflammation and prostatic atrophy

As shown in Table 1, an assessment of the distribution of IL6 mRNA expression in the prostatectomy tissues as analyzed by IL6 CISH demonstrated that, overall, the expression was highly heterogeneous from case-to-case and by region within a given case. As shown in Fig. 4A–B, the number of IL6-positive cells was highly increased in the stroma in areas of acute inflammation (as evidenced by accumulation of neutrophils within glandular lumens), although the neutrophils were not positive. The cases with the highest numbers of IL6 mRNA expressing cells were those in which acute inflammation was present (see Table 1). Increases in IL6-positive cells were also seen in areas of proliferative inflammatory atrophy (PIA, Fig. 4C–D). Positive cells in areas of prostatic atrophy were generally confined to the stroma surrounding atrophy. In some instances, positive IL6 mRNA expression was observed in select epithelial cells in atrophic glands (Table 1, Fig. 4D). Morphologically these appeared to be epithelial cells; however we did not perform a double label with an epithelial cell marker. In a number of cases the IL6 mRNA-expressing cells could be identified as endothelial cells lining small blood vessels (presumably venules or lymphatics, Fig. 4C) as well as prostate smooth muscle cells. Other positive staining cells had an appearance consistent with macrophages. To more definitively identify IL6 mRNA-expressing cells, we performed co-staining (2-plex) for IL6 and either CD68 (a macrophage marker) or CD31 (an endothelial cell marker). The results of this 2-plex staining clearly indicated that some of the IL6-positive cells are CD68-positive macrophages and some are CD31-positive endothelial cells (Supplementary Fig. S1). Interestingly, the majority of IL6-positive cells in areas of acute inflammation did not co-stain for CD68 or CD31, suggesting that most of the IL6 mRNA-expressing cells in these areas are of a different stromal cell type, such as fibroblasts, smooth muscle cells, other types of inflammatory cells, etc.

An external file that holds a picture, illustration, etc.
Object name is nihms698369f4.jpg
IL6 mRNA expression is nearly exclusively restricted to the prostate stromal compartment

(A,B) IL6 mRNA-expressing cells (brown staining) are highly enriched in the stroma in areas of acute inflammation as indicated by the presence of neutrophils in glandular lumens (red arrows). (C) IL6-positive cells in the stroma (arrows) and endothelium (arrowheads) surrounding prostate atrophy. (D) Positive epithelial staining was rare and restricted to prostatic atrophy (arrowheads).

Prostate adenocarcinoma cells in metastatic lesions do not express IL6 mRNA

Previous studies suggested that production of IL6 by metastatic prostate cancer cells may facilitate invasion and metastasis to bone and/or promote resistance to prostate cancer therapies (15, 22). Therefore, we next examined IL6 mRNA expression in biopsy samples from patients with castration-resistant metastatic prostate cancer (samples evaluated included 4 lymph node specimens and 3 liver biopsy samples) and 25 autopsy samples which included metastases to the liver, lung, bone, and lymph node (see Supplementary Table S1). Consistent with data from primary prostatectomy specimens, IL6 mRNA expression was not observed in prostate cancer cells in any of the metastatic tissues examined. Also consistent with the primary prostatectomy specimens, the metastatic tissues all showed strong hybridization signals with PPIB as a positive control (Fig. 5A), confirming RNA integrity. Interestingly, IL6 mRNA staining was observed in endothelial cells in blood vessels in 9 of 21 bone metastases (42.9%, Fig. 5B, Supplementary Fig. S2, Supplementary Table S3) but not in any of the 11 soft tissue metastases analyzed (p = 0.013, Fisher’s exact test).

An external file that holds a picture, illustration, etc.
Object name is nihms698369f5.jpg
IL6 mRNA is not detected in metastatic prostate cancer cells

In this study, metastatic prostate cancer in lymph node and liver biopsy samples and autopsy samples (liver, lung, bone, and lymph node) were assayed by IL6 CISH. No IL6-positive mRNA expression was observed in prostate cancer cells in any of the metastatic tissues. (A) Shown are representative examples of lymph node biopsy and autopsy liver, lung, and bone metastases with positive staining for PPIB (positive control) and negative staining for IL6. (B) An example of IL6 mRNA-positive blood vessels (arrowheads) and IL6-negative tumor cells (arrows) in bone metastases.

IL6 IHC is only successful when Golgi export is blocked

Finally, we performed a series of analyses to examine IL6 protein production in cell lines and tissues using IHC. To perform these studies, we prepared FFPE blocks from a lung cancer cell line (NCI-H460) that was strongly positive for IL6 mRNA as assessed by CISH (Fig. 6A) and produces physiologic levels (e.g., not transfection levels) of IL6 protein as assessed by Western blot (Supplementary Fig. S3). Of interest, there appears to be heterogeneity in cells that are positive for IL6 expression (i.e., not all cultured cells appear to be positive) via CISH (Fig. 6A) and this was consistent with what we observed for the IL6 mRNA-positive prostate cell lines as well (Fig. 2C). IHC on the NCI-H460 cells using the polyclonal anti-IL6 antibody (#6672; Abcam, lot #GR106735-5) showed no detectable signal above background levels (as established by negative control cell lines, Fig. 6 B,C). In one sense, these data are not particularly surprising, since IL6 is a secreted protein and secreted proteins are generally only detectable by flow cytometry analysis when protein export from the Golgi apparatus is blocked using protein transport inhibitors such as brefeldin A or monensin (23). As such, we next treated NCI-H460 cells with monensin prior to formalin fixation and preparation of FFPE blocks for use with IHC. IHC of “stopped” cell lines treated with monensin showed that NCI-H460 cells now stained positive for IL6 protein (Fig. 6D). We then performed IL6 IHC on a series of prostatectomy specimens including cases that were strongly positive in areas of acute inflammation with IL6 CISH (Fig. 6E, F). We did not detect IL6 protein above background levels in the acutely inflamed areas, either in epithelial or stromal cells using IHC, and likewise we did not observe IL6 protein in prostate tumor cells using IHC (Fig. 6G). Similarly, we did not detect any IL6 protein in tumor cells via IHC on metastatic lesions from standard slides from cases used for CISH assays or in two separate metastatic prostate cancer TMAs, including one of bone metastases (Fig. 6H). The IHC signal in metastases was restricted to extracellular spaces between tumor cell nests (Fig. 6H) and no specific signal was detectable above background levels in the tumor cells in any of the cases. The lack of positive signal for IL6 protein using IHC in areas that were positive for IL6 mRNA using CISH is likely not due to lack of IL6 protein production by the IL6 mRNA-positive cells. Rather, these results, coupled with our experiments using monensin in the NCI-H460 cell line, suggest that detection of this particular cytokine in tissue sections using IHC could be severely limited without being able to block protein export prior to fixation.

An external file that holds a picture, illustration, etc.
Object name is nihms698369f6.jpg
A requirement for protein transport inhibition for IL6 IHC

(A) IL6 CISH performed on the IL6-positive cell line NCI-H460. IL6 IHC performed on (B) PC3 cells (IL6 negative as assessed by q-RT-PCR and CISH) and (C) NCI-H460 cells. Marked difference in IHC results when NCI-H460 cells are treated with monensin prior to fixation (D). Arrows point to IL6-positive cells. The same prostatectomy case containing acute inflammation assayed for IL6 by CISH (E) and IHC (F). Example of IL6 IHC on primary prostate cancer (G) and prostate cancer bone metastasis (H). All tumor cells were negative in all samples analyzed. Positive staining observed in extracellular spaces (arrows in H) was considered to be non-specific.

To further demonstrate the sensitivity of the CISH assay over IHC, we used qPCR to generate a standard curve using estimated copies of the IL6 cDNA vector (Supplementary Fig. S4A). q-RT-PCR was then performed with RNA extracted from a known number of cells of the DU145 cell line (the lowest positive IL6-expressing cell line by q-RT-PCR/CISH in Fig. 2 B,C) and PC3 (determined to be negative in our hands by CISH). Comparison of the results from DU145 and PC3 to the standard curve indicated values of approximately 2 copies of IL6 mRNA per cell in the DU145 cell line and 0.017 copies per cell for PC3 (roughly 1 copy of IL6 mRNA per 59 cells). Cells from the same flasks of DU145 and PC3 used for RNA extraction were made into FFPE cell blocks. CISH analysis revealed (as also seen in Fig. 2C) positive detection of IL6 mRNA within the cytoplasm of DU145 cells (Supplementary Fig. S4B). Conversely, in PC3 cells we see no cytoplasmic staining and generally one or two single dots in the nucleus. Since we have observed such signals at times in cells that have no corresponding mRNA (A.M. De Marzo, Q. Zheng, A.K. Meeker unpublished results), and since it is clear that the CISH assay can at times detect genomic DNA as one or two dots in the nucleus (21), we do not consider 1–2 dots in the nucleus to be a bona fide positive signal and consider it below the limit of detection. IHC on adjacent slides were negative for IL6 protein (Supplementary Fig. S4B). We do not interpret this to mean that IL6 mRNA-expressing cells do not make IL6 protein, but that the IL6 protein cannot be detected using IHC without protein transport inhibition and, therefore, the CISH assay is more sensitive than our standard IHC assay.

Conclusions

The cellular origin of IL6 in prostate cancer: a focus on the stromal compartment

In the present study, we analytically validated and used a chromogenic in situ hybridization assay to detect IL6 mRNA in tissue sections to sensitively and specifically determine the cellular origin of IL6 in the prostate primary and metastatic tumor microenvironment. The results of our studies indicate that prostatic adenocarcinoma cells do not express IL6 mRNA. Rather, IL6 mRNA expression is restricted nearly exclusively to the cells in the stromal compartment including endothelium, and is highly up-regulated in areas of acute inflammation and prostatic atrophy.

Although prostate adenocarcinoma cells do not express IL6 mRNA as evidenced by the results of our study, this does not eliminate a potential contributory role for IL6 signaling in prostate cancer development and/or progression. IL6 production by cells in the stromal compartment may signal in a paracrine fashion through the transmembrane IL6 receptor (IL6R) mediated by glycoprotein 130 (gp130) or via a soluble IL6 receptor (sIL6R) that signals through membrane-bound gp130. Therefore, IL6 potentially can signal through any cell that produces IL6R or gp130. In this respect, multiple studies have demonstrated the presence of IL6R and/or gp130 in prostate cancer cells (reviewed in (24)).

Interestingly, of the 11 prostate cell lines derived from cancer or benign tissues that we examined for IL6 mRNA expression levels in the present study, the cell line that was found to express the highest levels of IL6 mRNA via qRT-PCR was PrSC cells (Fig. 2B). PrSC cells are a prostate stromal cell line; therefore, the high levels of IL6 mRNA expression in this cell line of stromal cell origin would correlate to our IL6 CISH results in prostate tissue sections where we found that IL6 mRNA expression was restricted almost exclusively to the stromal compartment. Of note, we also detected IL6 mRNA expression in RWPE-1 cells, which are HPV-immortalized benign prostate epithelial cells. It is possible that IL6 expression is induced in this line by the HPV-infection (25). Whereas dual stains for IL6 and CD68 or CD31 confirmed that some of the IL6 mRNA-expressing cells in the stromal compartment are prostate-infiltrating macrophages or endothelial cells, respectively (Supplementary Fig. S1), this represented the minority of IL6-positive stromal cells in the highly positive areas surrounding acute inflammation (Fig. 4A–B). We predict that additional cells in the stromal compartment that express IL6 mRNA may include fibroblasts/myofibroblasts and smooth muscle cells. In this respect, Hobisch and colleagues detected IL6 secretion into the supernatant of ex vivo cultured prostatic fibroblasts and smooth muscle cells, although no positive staining of these types of stromal cells was detected using IHC (14). It should be noted that those data are quite consistent with the results of the present study.

In areas of acute inflammation in some cases, a large proportion of cells in the stroma were positive for IL6 mRNA expression (Fig. 4A–B), arguing that a number of cell types in these areas may express IL6 mRNA in what may be part of a positive feedback loop as has been previously described (26). The stimulus for acute inflammation that is frequently observed on radical prostatectomy specimens (albeit to a lesser degree than chronic inflammation) is unknown (27), but may be caused in part by bacterial infections (2729). A recent study also identified a role for IL6 up-regulation by bone marrow-derived mesenchymal stem cells (MSC) in promotion of adipogenesis and prostate cancer progression (18), and it is possible that some of the IL6-positive cells identified in the present study may represent MSCs. This would be difficult to assess using the current CISH technologies that are limited to 1–2 markers per assay, as MSCs are typically identified using a number of surface markers such as CD105, CD166, CD44 and CD29 (18). Future studies utilizing techniques such as flow cytometry may help to further verify this possibility. Another recent study using a human prostate dissociation and tissue recombination system identified a role for paracrine expression of IL6 in the stromal compartment in concert with cell-autonomous oncogenic events in the promotion of an aggressive prostate cancer phenotype (17). In all, these studies in parallel with the results of the present study set the precedence for a potentially important role for paracrine IL6 signaling originating from the stromal compartment in the prostate tumor microenvironment.

The role of IL6 in metastatic disease

Studies have consistently shown that serum levels of IL6 are elevated in metastatic prostate cancer patients (3, 4, 69, 30), and that these levels may correlate to tumor burden (4, 5, 31) and/or may serve as a surrogate marker for morbidity associated with advanced prostate cancer including cachexia (9, 10). Interestingly, our studies of IL6 CISH in a series of metastatic prostate cancer biopsy or autopsy samples indicated that metastatic prostate cancer cells do not express IL6 mRNA. While prostate tumor cells do not express IL6, we did observe a significant difference between IL6-positive blood vessels in bone metastases versus soft tissue metastases (Supplementary Table S3). Our data strongly suggest that the increased systemic levels of IL6 observed in advanced prostate cancer patients is not from production of the cytokine by metastatic tumor cells. Rather, elevated IL6 levels may be due at least in part to increased IL6 production by the tumor vasculature. Since the presence of bone metastases is also associated with morbidity, it is plausible that the presence of IL6 in vasculature in bone metastases could be a source of the elevated serum IL6 in patients with a high metastatic burden. The lack of IL6 mRNA expression by primary and metastatic prostate cancer cells observed in our study may help explain why minimal to no clinical activity has been observed to date when a monoclonal antibody therapy targeting IL6 (siltuximab) has been tested in clinical trials (32, 33).

IL6 and IHC

IHC-based studies on IL6 production in prostate tissue sections have reported varying degrees of basal cell staining in benign epithelium as well as more pronounced staining of tumor epithelium (13, 14) that increases in intensity with increasing pathologic Gleason grade (13). Likewise, a previous study using IHC to detect IL6 production in prostate cancer metastases reported over twice as many bone metastases samples are positive for IL6 than soft tissue metastases, and with much stronger staining intensity (15). Unfortunately, our data do not support those results. With a well-validated, positively controlled IL6 CISH assay, we did not detect IL6 mRNA in any of the primary or metastatic prostate cancer cells in our study. We were able to detect IL6 using IHC in positive control cell line specimens, but only when IL6 accumulation was augmented by blocking Golgi export using a protein transport inhibitor.

To help determine whether we could account for the discrepant results between a past study (15) and the present one, we compared by IHC the staining obtained with the polyclonal anti-IL6 antibody batch (#6672; Abcam, lot # 385304) that was used previously to the currently commercially available batch (lot # GR169214-2). These analyses indicated that under identical conditions, the newer version of the antibody does not stain tumor cells in bone metastases, yet, the older antibody batch gave similar results to that reported previously (15) with strong tumor cell staining (C. Morrissey, unpublished data). Taken together with the present study, it would appear that prior results showing high level expression in tumor cells is related to antibody lot variability and it no longer appears with the newer currently available antibody from the same vendor. It should also be noted that traditional decalcification/fixation methods used for bone can have a significant impact on RNA integrity; although formic acid treatment as was used for the metastatic bone samples in the present study has been shown to result in improved nucleic acid recovery and quality over stronger acid treatment (34, 35). We acknowledge this potential limitation to the present study, and addressed this concern by including the positive control (PPIB) in the CISH assay for each sample analyzed. Likewise, we detected IL6 mRNA in the metastatic bone samples (albeit not in the tumor cells but in the blood vessels), indicating that at least there did not appear to be any generalized issues with detection of IL6 mRNA in the decalcified bone samples.

In conclusion, our results suggest that, in prostate cancer patients, paracrine rather than autocrine IL6 production is likely associated with any role for the cytokine in disease development and/or progression.

The cellular origin of IL6 in prostate cancer: a focus on the stromal compartment

In the present study, we analytically validated and used a chromogenic in situ hybridization assay to detect IL6 mRNA in tissue sections to sensitively and specifically determine the cellular origin of IL6 in the prostate primary and metastatic tumor microenvironment. The results of our studies indicate that prostatic adenocarcinoma cells do not express IL6 mRNA. Rather, IL6 mRNA expression is restricted nearly exclusively to the cells in the stromal compartment including endothelium, and is highly up-regulated in areas of acute inflammation and prostatic atrophy.

Although prostate adenocarcinoma cells do not express IL6 mRNA as evidenced by the results of our study, this does not eliminate a potential contributory role for IL6 signaling in prostate cancer development and/or progression. IL6 production by cells in the stromal compartment may signal in a paracrine fashion through the transmembrane IL6 receptor (IL6R) mediated by glycoprotein 130 (gp130) or via a soluble IL6 receptor (sIL6R) that signals through membrane-bound gp130. Therefore, IL6 potentially can signal through any cell that produces IL6R or gp130. In this respect, multiple studies have demonstrated the presence of IL6R and/or gp130 in prostate cancer cells (reviewed in (24)).

Interestingly, of the 11 prostate cell lines derived from cancer or benign tissues that we examined for IL6 mRNA expression levels in the present study, the cell line that was found to express the highest levels of IL6 mRNA via qRT-PCR was PrSC cells (Fig. 2B). PrSC cells are a prostate stromal cell line; therefore, the high levels of IL6 mRNA expression in this cell line of stromal cell origin would correlate to our IL6 CISH results in prostate tissue sections where we found that IL6 mRNA expression was restricted almost exclusively to the stromal compartment. Of note, we also detected IL6 mRNA expression in RWPE-1 cells, which are HPV-immortalized benign prostate epithelial cells. It is possible that IL6 expression is induced in this line by the HPV-infection (25). Whereas dual stains for IL6 and CD68 or CD31 confirmed that some of the IL6 mRNA-expressing cells in the stromal compartment are prostate-infiltrating macrophages or endothelial cells, respectively (Supplementary Fig. S1), this represented the minority of IL6-positive stromal cells in the highly positive areas surrounding acute inflammation (Fig. 4A–B). We predict that additional cells in the stromal compartment that express IL6 mRNA may include fibroblasts/myofibroblasts and smooth muscle cells. In this respect, Hobisch and colleagues detected IL6 secretion into the supernatant of ex vivo cultured prostatic fibroblasts and smooth muscle cells, although no positive staining of these types of stromal cells was detected using IHC (14). It should be noted that those data are quite consistent with the results of the present study.

In areas of acute inflammation in some cases, a large proportion of cells in the stroma were positive for IL6 mRNA expression (Fig. 4A–B), arguing that a number of cell types in these areas may express IL6 mRNA in what may be part of a positive feedback loop as has been previously described (26). The stimulus for acute inflammation that is frequently observed on radical prostatectomy specimens (albeit to a lesser degree than chronic inflammation) is unknown (27), but may be caused in part by bacterial infections (2729). A recent study also identified a role for IL6 up-regulation by bone marrow-derived mesenchymal stem cells (MSC) in promotion of adipogenesis and prostate cancer progression (18), and it is possible that some of the IL6-positive cells identified in the present study may represent MSCs. This would be difficult to assess using the current CISH technologies that are limited to 1–2 markers per assay, as MSCs are typically identified using a number of surface markers such as CD105, CD166, CD44 and CD29 (18). Future studies utilizing techniques such as flow cytometry may help to further verify this possibility. Another recent study using a human prostate dissociation and tissue recombination system identified a role for paracrine expression of IL6 in the stromal compartment in concert with cell-autonomous oncogenic events in the promotion of an aggressive prostate cancer phenotype (17). In all, these studies in parallel with the results of the present study set the precedence for a potentially important role for paracrine IL6 signaling originating from the stromal compartment in the prostate tumor microenvironment.

The role of IL6 in metastatic disease

Studies have consistently shown that serum levels of IL6 are elevated in metastatic prostate cancer patients (3, 4, 69, 30), and that these levels may correlate to tumor burden (4, 5, 31) and/or may serve as a surrogate marker for morbidity associated with advanced prostate cancer including cachexia (9, 10). Interestingly, our studies of IL6 CISH in a series of metastatic prostate cancer biopsy or autopsy samples indicated that metastatic prostate cancer cells do not express IL6 mRNA. While prostate tumor cells do not express IL6, we did observe a significant difference between IL6-positive blood vessels in bone metastases versus soft tissue metastases (Supplementary Table S3). Our data strongly suggest that the increased systemic levels of IL6 observed in advanced prostate cancer patients is not from production of the cytokine by metastatic tumor cells. Rather, elevated IL6 levels may be due at least in part to increased IL6 production by the tumor vasculature. Since the presence of bone metastases is also associated with morbidity, it is plausible that the presence of IL6 in vasculature in bone metastases could be a source of the elevated serum IL6 in patients with a high metastatic burden. The lack of IL6 mRNA expression by primary and metastatic prostate cancer cells observed in our study may help explain why minimal to no clinical activity has been observed to date when a monoclonal antibody therapy targeting IL6 (siltuximab) has been tested in clinical trials (32, 33).

IL6 and IHC

IHC-based studies on IL6 production in prostate tissue sections have reported varying degrees of basal cell staining in benign epithelium as well as more pronounced staining of tumor epithelium (13, 14) that increases in intensity with increasing pathologic Gleason grade (13). Likewise, a previous study using IHC to detect IL6 production in prostate cancer metastases reported over twice as many bone metastases samples are positive for IL6 than soft tissue metastases, and with much stronger staining intensity (15). Unfortunately, our data do not support those results. With a well-validated, positively controlled IL6 CISH assay, we did not detect IL6 mRNA in any of the primary or metastatic prostate cancer cells in our study. We were able to detect IL6 using IHC in positive control cell line specimens, but only when IL6 accumulation was augmented by blocking Golgi export using a protein transport inhibitor.

To help determine whether we could account for the discrepant results between a past study (15) and the present one, we compared by IHC the staining obtained with the polyclonal anti-IL6 antibody batch (#6672; Abcam, lot # 385304) that was used previously to the currently commercially available batch (lot # GR169214-2). These analyses indicated that under identical conditions, the newer version of the antibody does not stain tumor cells in bone metastases, yet, the older antibody batch gave similar results to that reported previously (15) with strong tumor cell staining (C. Morrissey, unpublished data). Taken together with the present study, it would appear that prior results showing high level expression in tumor cells is related to antibody lot variability and it no longer appears with the newer currently available antibody from the same vendor. It should also be noted that traditional decalcification/fixation methods used for bone can have a significant impact on RNA integrity; although formic acid treatment as was used for the metastatic bone samples in the present study has been shown to result in improved nucleic acid recovery and quality over stronger acid treatment (34, 35). We acknowledge this potential limitation to the present study, and addressed this concern by including the positive control (PPIB) in the CISH assay for each sample analyzed. Likewise, we detected IL6 mRNA in the metastatic bone samples (albeit not in the tumor cells but in the blood vessels), indicating that at least there did not appear to be any generalized issues with detection of IL6 mRNA in the decalcified bone samples.

In conclusion, our results suggest that, in prostate cancer patients, paracrine rather than autocrine IL6 production is likely associated with any role for the cytokine in disease development and/or progression.

Supplementary Material

1

2

3

4

5

6

1

Click here to view.(22M, tif)

2

Click here to view.(17M, tif)

3

Click here to view.(475K, tif)

4

Click here to view.(1.8M, tif)

5

Click here to view.(13K, docx)

6

Click here to view.(95K, pdf)

Acknowledgements

We thank the patients and their families who participated in the studies at Johns Hopkins and the Prostate Cancer Donor Program at the University of Washington. We acknowledge the investigators Drs. Celestia Higano, Paul Lange, Bruce Montgomery, Evan Yu, Martine Roudier, Peter Nelson, and Lawrence True for their contributions to the University of Washington Medical Center Prostate Cancer Donor Rapid Autopsy Program and Belinda Nghiem for technical help.

Financial Support: K.S.S. is supported as the Chris and Felicia Evensen Prostate Cancer Foundation (PCF) Young Investigator. K.S.S. and A.M.D. are supported through The Patrick C. Walsh Prostate Cancer Research Fund as The Beth W. and A. Ross Myers Scholar and The Virginia and Warren Schwerin Scholar, respectively. NIH/NCI prostate SPORE pathology core (Award No 5P50CA058236), Prostate Cancer Biorepository Network (PCBN) (Department of Defense Award No W81XWH-10-2-0056 and W81XWH-10-2-0046), Pacific Northwest Prostate Cancer SPORE (P50CA97186), PO1 NIH grant (PO1CA085859), Richard M. LUCAS Foundation.

Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287
Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287
Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287
Department of Urology, University of Washington Medical Center, Seattle, WA, 98195
Corresponding Author: Karen S. Sfanos, Department of Pathology, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB II Rm. 153, Baltimore, MD 21231. Phone: 443-287-4592. Fax: 410-502-9817. ude.imhj@sonafsk

Abstract

Correlative human studies suggest that the pleiotropic cytokine interleukin-6 (IL6) contributes to the development and/or progression of prostate cancer. However, the source of IL6 production in the prostate microenvironment in patients has yet to be determined. The cellular origin of IL6 in primary and metastatic prostate cancer was examined in formalin-fixed, paraffin-embedded (FFPE) tissues using a highly sensitive and specific chromogenic in situ hybridization (CISH) assay that underwent extensive analytical validation. Quantitative RT-PCR (q-RT-PCR) showed that benign prostate tissues often had higher expression of IL6 mRNA than matched tumor specimens. CISH analysis further indicated that both primary and metastatic prostate adenocarcinoma cells do not express IL6 mRNA. IL6 expression was highly heterogeneous across specimens and was nearly exclusively restricted to the prostate stromal compartment – including endothelial cells and macrophages among other cell types. The number of IL6-expressing cells correlated positively with the presence of acute inflammation. In metastatic disease, tumor cells were negative in all lesions examined and IL6 expression was restricted to endothelial cells within the vasculature of bone metastases. Finally, IL6 was not detected in any cells in soft tissue metastases. These data suggest that, in prostate cancer patients, paracrine rather than autocrine IL6 production is likely associated with any role for the cytokine in disease progression.

Keywords: IL6, prostate cancer, neoangiogenesis, metastasis, microenvironment
Abstract

Footnotes

Conflicts of Interest: None

Footnotes

References

  • 1. Keller ET, Wanagat J, Ershler WBMolecular and cellular biology of Interleukin-6 and its receptor. Front Biosci. 1996;1:d340–d357.[PubMed][Google Scholar]
  • 2. Hodge DR, Hurt EM, Farrar WLThe role of IL-6 and STAT3 in inflammation and cancer. Eur J Cancer. 2005;41:2502–2512.[PubMed][Google Scholar]
  • 3. Drachenberg DE, Elgamal A-AA, Rowbotham R, Peterson M, Murphy GPCirculating levels of interleukin-6 in patients with hormone refractory prostate cancer. Prostate. 1999;41:127–133.[PubMed][Google Scholar]
  • 4. Adler HL, McCurdy MA, Kattan MW, Timme TL, Scardino PT, Thompson TCElevated levels of circulating interleukin-6 and transforming growth factor-beta 1 in patients with metastatic prostatic carcinoma. J Urol. 1999;161:182–187.[PubMed][Google Scholar]
  • 5. Akimoto S, Okumura A, Fuse HRelationship between serum levels of Interleukin-6, tumor necrosis factor-alpha; and bone turnover markers in prostate cancer patients. Endocr J. 1998;45:183–189.[PubMed][Google Scholar]
  • 6. Tumminello F, Badalamenti G, Incorvaia L, Fulfaro F, D’Amico C, Leto GSerum interleukin-6 in patients with metastatic bone disease: correlation with cystatin C. Med Oncol. 2009;26:10–15.[PubMed][Google Scholar]
  • 7. George DJ, Halabi S, Shepard TF, Sanford B, Vogelzang NJ, Small EJ, et al The prognostic significance of plasma interleukin-6 levels in patients with metastatic hormone-refractory prostate cancer: Results from cancer and leukemia group B 9480. Clin Cancer Res. 2005;11:1815–1820.[PubMed][Google Scholar]
  • 8. Shariat SF, Andrews B, Kattan MW, Kim J, Wheeler TM, Slawin KMPlasma levels of interleukin-6 and its soluble receptor are associated with prostate cancer progression and metastasis. Urology. 2001;58:1008–1015.[PubMed][Google Scholar]
  • 9. Twillie DA, Eisenberger MA, Carducci MA, Hseih W-S, Kim WY, Simons JWInterleukin-6: A candidate mediator of human prostate cancer morbidity. Urology. 1995;45:542–549.[PubMed][Google Scholar]
  • 10. Kuroda K, Nakashima J, Kanao K, Kikuchi E, Miyajima A, Horiguchi Y, et al Interleukin 6 is associated with cachexia in patients with prostate cancer. Urology. 2007;69:113–117.[PubMed][Google Scholar]
  • 11. Chung TDK, Yu JJ, Spiotto MT, Bartkowski M, Simons JWCharacterization of the role of IL-6 in the progression of prostate cancer. Prostate. 1999;38:199–207.[PubMed][Google Scholar]
  • 12. Giri D, Ozen M, Ittmann MInterleukin-6 is an autocrine growth factor in human prostate cancer. Am J Pathol. 2001;159:2159–2165.[Google Scholar]
  • 13. Royuela M, Ricote M, Parsons MS, García-Tuñón I, Paniagua R, De Miguel MPImmunohistochemical analysis of the IL-6 family of cytokines and their receptors in benign, hyperplasic, and malignant human prostate. J Pathol. 2004;202:41–49.[PubMed][Google Scholar]
  • 14. Hobisch A, Rogatsch H, Hittmair A, Fuchs D, Bartsch G, Klocker H, et al Immunohistochemical localization of interleukin-6 and its receptor in benign, premalignant and malignant prostate tissue. J Pathol. 2000;191:239–244.[PubMed][Google Scholar]
  • 15. Morrissey C, Lai JS, Brown LG, Wang Y-C, Roudier MP, Coleman IM, et al The expression of osteoclastogenesis-associated factors and osteoblast response to osteolytic prostate cancer cells. Prostate. 2010;70:412–424.[Google Scholar]
  • 16. Culig Z, Steiner H, Bartsch G, Hobisch AInterleukin-6 regulation of prostate cancer cell growth. J Cell Biochem. 2005;95:497–505.[PubMed][Google Scholar]
  • 17. Smith DA, Kiba A, Zong Y, Witte ONInterleukin-6 and oncostatin-M synergize with the PI3K/AKT pathway to promote aggressive prostate malignancy in mouse and human tissues. Mol Cancer Res. 2013;11:1159–1165.[Google Scholar]
  • 18. Sung S-Y, Liao C-H, Wu H-P, Hsiao W-C, Wu IH, Jinpu, et al Loss of Let-7 microRNA upregulates IL-6 in bone marrow-derived mesenchymal stem cells triggering a reactive stromal response to prostate cancer. PLoS ONE. 2013;8:e71637.[Google Scholar]
  • 19. Darshan M, Zheng Q, Fedor HL, Wyhs N, Yegnasubramanian S, Lee P, et al Biobanking of derivatives from radical retropubic and robot-assisted laparoscopic prostatectomy tissues as part of the prostate cancer biorepository network. Prostate. 2014;74:61–69.[Google Scholar]
  • 20. Rhodes D, Yu J, Shanker K, Deshpande N, Varambally R, Ghosh D, et al ONCOMINE: a cancer microarray database and integrated data-mining platform. Neoplasia. 2004;6:1–6.[Google Scholar]
  • 21. Player AN, Shen L-P, Kenny D, Antao VP, Kolberg JASingle-copy gene detection using branched DNA (bDNA) in situ hybridization. J Histochem Cytochem. 2001;49:603–611.[PubMed][Google Scholar]
  • 22. Tawara K, Oxford J, Jorcyk CClinical significance of interleukin (IL)-6 in cancer metastasis to bone: potential of anti-IL-6 therapies. Cancer Manag Res. 2011;3:177–189.[Google Scholar]
  • 23. Jung T, Schauer U, Heusser C, Neumann C, Rieger CDetection of intracellular cytokines by flow cytometry. J Immunol Methods. 1993;159:197–207.[PubMed][Google Scholar]
  • 24. Smith PC, Hobisch A, Lin D-L, Culig Z, Keller ETInterleukin-6 and prostate cancer progression. Cytokine Growth Factor Rev. 2001;12:33–40.[PubMed][Google Scholar]
  • 25. Ren C, Cheng X, Lu B, Yang GActivation of interleukin-6/signal transducer and activator of transcription 3 by human papillomavirus early proteins 6 induces fibroblast senescence to promote cervical tumourigenesis through autocrine and paracrine pathways in tumour microenvironment. Eur J Cancer. 2013;49:3889–3899.[PubMed][Google Scholar]
  • 26. Ogura H, Murakami M, Okuyama Y, Tsuruoka M, Kitabayashi C, Kanamoto M, et al Interleukin-17 promotes autoimmunity by triggering a positive-feedback loop via Interleukin-6 induction. Immunity. 2008;29:628–636.[PubMed][Google Scholar]
  • 27. De Marzo AM, Platz EA, Sutcliffe S, Xu J, Gronberg H, Drake CG, et al Inflammation in prostate carcinogenesis. Nat Rev Cancer. 2007;7:256–269.[Google Scholar]
  • 28. Sfanos KS, De Marzo AMProstate cancer and inflammation: the evidence. Histopathology. 2012;60:199–215.[Google Scholar]
  • 29. Sfanos KS, Wilson BA, De Marzo AM, Isaacs WBAcute inflammatory proteins constitute the organic matrix of prostatic corpora amylacea and calculi in men with prostate cancer. Proc Natl Acad Sci U S A. 2009;106:3443–3448.[Google Scholar]
  • 30. Hoosein N, Abdul M, McCabe R, Gero E, Deftos L, Banks M, et al Clinical significance of elevation in neuroendocrine factors and interleukin-6 in metastatic prostate cancer. Urol Oncol. 1995;1:246–251.[PubMed][Google Scholar]
  • 31. Michalaki V, Syrigos K, Charles P, Waxman JSerum levels of IL-6 and TNF-[alpha] correlate with clinicopathological features and patient survival in patients with prostate cancer. Br J Cancer. 2004;90:2312–2316.[Google Scholar]
  • 32. Fizazi K, De Bono JS, Flechon A, Heidenreich A, Voog E, Davis NB, et al Randomised phase II study of siltuximab (CNTO 328), an anti-IL-6 monoclonal antibody, in combination with mitoxantrone/prednisone versus mitoxantrone/prednisone alone in metastatic castration-resistant prostate cancer. Eur J Cancer. 2012;48:85–93.[PubMed][Google Scholar]
  • 33. Dorff TB, Goldman B, Pinski JK, Mack PC, Lara PN, Van Veldhuizen 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 Res. 2010;16:3028–3034.[Google Scholar]
  • 34. Singh VM, Salunga RC, Huang VJ, Tran Y, Erlander M, Plumlee P, et al Analysis of the effect of various decalcification agents on the quantity and quality of nucleic acid (DNA and RNA) recovered from bone biopsies. Ann Diagn Pathol. 2013;17:322–326.[PubMed][Google Scholar]
  • 35. Brown RSD, Edwards J, Bartlett JW, Jones C, Dogan ARoutine acid decalcification of bone marrow samples can preserve DNA for FISH and CGH studies in metastatic prostate cancer. J Histochem Cytochem. 2002;50:113–115.[PubMed][Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.