Gemcitabine constitutes one of the backbones for chemotherapy treatment in pancreatic ductal adenocarcinoma (PDAC), but patients often respond poorly to this agent. Molecular markers downstream of gemcitabine treatment in preclinical models may provide an insight into resistance mechanisms. Using cytokine arrays, we identified potential secretory biomarkers of gemcitabine resistance (response) in the transgenic KRasG12D; Trp53R172H; Pdx-1 Cre (KPC) mouse model of PDAC. We verified the oncogenic role of the cytokine tissue inhibitor of matrix metalloproteinases 1 (TIMP1) in primary pancreatic tumors and metastases using both in vitro techniques and animal models. We identified potential pathways affected downstream of TIMP1 using the Illumina Human H12 array. Our findings were validated in both primary and metastatic models of pancreatic cancer. Gemcitabine increased inflammatory cytokines including TIMP1 in the KPC mouse model. TIMP1 was upregulated in patients with pancreatic intraepithelial neoplasias grade 3 and PDAC lesions relative to matched normal pancreatic tissue. In addition, TIMP1 played a role in tumor clonogenic survival and vascular density, while TIMP1 inhibition resensitized tumors to gemcitabine and radiotherapy. We observed a linear relationship between TIMP-1 expression, liver metastatic burden, and infiltration by CD11b+Gr1+ myeloid cells and CD4+CD25+FOXP3+ Tregs, whereas the presence of tumor cells was required for immune cell infiltration. Overall, our results identify TIMP1 upregulation as a resistance mechanism to gemcitabine and provide a rationale for combining chemo/radiotherapy with TIMP1 inhibitors in PDAC. Cancer Res; 77(21); 5952–62. ©2017 AACR.

Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive cancers with dismal prognosis for overall survival, due to late presentation and limited responses to therapy (1, 2). About 80% of cases are inoperable at diagnosis leaving chemotherapy and/or chemoradiotherapy as the main treatment options. Gemcitabine constitutes one of the chemotherapy backbones but patients commonly show either poor or even complete lack of response to this agent (1). Gemcitabine is a DNA nucleoside analogue that irreversibly inhibits ribonucleotide reductase activity, preventing the synthesis of dioxyribonucleotides required for DNA replication and repair (3). Treatment induces DNA damage and apoptosis of growing cells, thus is frequently employed in combinatorial chemotherapy regimens to treat ovarian, breast, and bladder cancers in addition to pancreatic cancer. While patients have been shown to benefit from gemcitabine, poor responses in pancreas imply that intrinsic resistance and poor patient response to this chemotherapeutic limits efficacy (4). In studies addressing gemcitabine sensitivity, it was recently shown that acute cytokine release following DNA damage may contribute to resistance acquired during the course of treatment as well as to treatment failure (5).

A number of cytokines have been associated with PDAC and have been investigated for potential as early diagnostic or predictive biomarkers, and are beginning to demonstrate some power in diagnosis (1, 4). TIMP1 was originally highlighted as having potential to distinguish patients with PDAC from controls; however, subsequent evidence demonstrated that although patients display elevated levels, TIMP1 does not identify early disease (6).

Tissue inhibitor of matrix metalloproteases (TIMP1) is known to inhibit metalloproteases (MMP) disintegrin and metalloproteinase domain-containing protein 10 and 17 (ADAM-10 and ADAM-17; ref. 7). TIMP1 has also been shown to activate prosurvival signaling, independent of its MMP inhibitory activity, through binding with its receptor CD63 and subsequent engagement of integrin β1–mediated activation of PI3K signaling (8–11).

TIMP1 can influence a number of tumorigenic biological processes, such as proliferation, apoptosis, and metastasis (7, 12, 13). Although TIMP1 expression is suggested as a prognostic marker in several malignancies (14–16), and its expression is associated with hyperproliferation of K-Ras (G12D)-mutated pancreatic cells (17), the functional role of TIMP1 within the PDAC microenvironment remains largely unexplored. Intriguingly, tumor-derived TIMP1 has been proposed to support neutrophil infiltration to the normal liver and the creation of a premetastatic niche to which circulating tumor cells adhere and seed metastasis in colorectal cancer (13). Here, we found that TIMP1 is directly elevated in response to gemcitabine treatment that was predominantly associated with a protumorigenic phenotype.

Cell lines

KPC cells derived from pancreatic tumors of KRasG12D; Trp53R172H; Pdx-1 Cre (KPC) mice and cultured in DMEM supplemented with 10% FCS and authenticated as previously reported (18, 19). The human PDAC cell line PANC1 was purchased from European Collection of Authenticated Cell Cultures and cultured in DMEM supplemented with 10% FCS. PANC1 cells were authenticated by STR profiling. To generate stable knockdowns (KD) of mouse TIMP1 and human TIMP1, KPC and PANC1 cells were transduced with lentivirus-mediated shRNA or the control vector (MISSION shRNA, Sigma). To generate stable overexpression, PANC1 cells were transduced with retroviral particles. The p6610 MSCV-IP N-HA only TIMP1 (retroviral plasmid) was purchased from Addgene (catalog number 35009) and used to generate live retroviral particles in the packaging cell line HEK 293T. The retroviral particles were then used to transduce PANC1 wild-type (WT) and knockdown (KD) cells, alongside an empty retroviral control vector. Immortalized pancreatic CAF-stellate cells were obtained from Neuromics, and cultured in VitroPlus III, low serum, complete (Neuromics). Pancreatic CAFS were phenotypically tested by Vitro Biopharma for a range of markers including CD105, CD90, CD44, CD326, CD133, FAP, GFAP, FSP1, α-SMA, and vimentin. KPC and PANC1 cells were selected with 5 μg/mL and 2.5 μg/mL of puromycin, respectively. All cell lines were obtained in 2015, and fresh aliquots were thawed and used up for a limited amount of time to the sixth passage, except gemcitabine-resistant cells that were maintained for two months in cell culture. All cell lines in our laboratory were tested for Mycoplasma every 4 weeks using the MycoAlert kit (Lonza).

Animal models

All animal experiments were carried out according to the UK Animal (Scientific Procedures) act of 1986. SCID and C57BL/6 mice were purchased from Charles River Laboratories. KPC transgenic mice (20) were bred at the premises of the University of Oxford (Oxford, United Kingdom). Genotyping was carried out at Transnetyx Inc. When KPC mice reached 12 weeks of age, palpation, sonography, and/or MRI was used to identify and measure the size of intra-abdominal pancreatic tumors in KPC mice.

For the subcutaneous models, human PANC1 and murine KPC cells (1 × 106 cells in 100 μL of PBS) were injected subcutaneously into SCID (immunosuppressive) and C57BL/6 (immunocompetent) mice, respectively. Mice were randomized when tumors reached 100 mm3. Where specified, mice were treated with gemcitabine (100 mg/kg, i.p.) or 8 Gy. Tumor volume was measured by calipers as described previously (21). Non-tumor–bearing C57BL/6 mice were also treated with gemcitabine.

For the liver metastasis model, SCID or C57BL/6 mice were injected intrasplenically with 5 × 105 PANC1 or KPC cells, respectively, in PBS as described previously (22). C57BL/6 mice were sacrificed at day 14 after inoculation, whereas SCID mice were imaged by MRI and sacrificed when liver metastasis was observed 21–26 days after inoculation. For all animal experiments, 5–7 mice were randomized in each experimental group.

MRI

The MRI method (23), including its technical characteristics, is described in detail in Supplementary Methods.

Cytokine arrays, phosphokinase, and ELISA

Proteome profiler antibody array and phosphokinase array (R&D Systems) were performed according to manufacturer's protocol. Human and mouse TIMP1 Quantikine and DuoSet ELISA kits (R&D Systems) were used according to manufacturer's protocol.

IHC/immunofluorescence, Western blot analysis, and flow cytometry

Tissue sections were stained for hematoxylin/eosin (H&E) and CD31 staining, and scored as described previously (21, 24). Immunofluorescence, Western blot analysis, and flow cytometry are described in Supplementary Methods.

Clonogenic survival, migration, and invasion assays

Clonogenic survival was carried out as reported previously (24). Wound scratches across a monolayer of KPC and PANC1 cells were conducted out using a sterile 200 μL pipette tip. Bright-field images were taken at 16 and 22 hours until wound closure. Boyden invasion chamber inserts (Costar, 12-μm pore size) were coated with 100 μg/cm2 of Matrigel. Cells were seeded onto the chamber insert and placed into a 24-well plate containing a cell line monolayer acting as an attractant in its respective media, and invasion was measured after 16 hours.

Microarray and oncomine analysis, RT-qPCR, and invasion toward CAFS

These assays are described in Supplementary Methods. In the microarray experiment, standard MIAME guidelines were followed. The full microarray data (GSE94891) are available on GEO.

Statistical analysis

GraphPad Prism 5 was used for all statistical analyses besides Kaplan–Meier survival curves for which IBM SPSS software Version 21 (SPSS Inc.) was used. By default, values were expressed as mean ± SD and statistical significance was measured using one-way ANOVA with Bonferroni posttest, or using unpaired t test. The log-rank test was used to compare Kaplan–Meier curves, while unpaired nonparametric comparisons were made using the Mann–Whitney test where appropriate. In all tests, P values <0.05 were considered significant.

Identification of TIMP1 as a gemcitabine response gene

To gain a better understanding of cytokine release following DNA damage, we treated transgenic KPC mice bearing tumor volumes between 70 and 120 mm3 with saline or gemcitabine (100 mg/kg, i.p.) at day 1, 4, and 8. To identify both early and sustained response genes following gemcitabine treatment, mice were divided into three groups; control mice were treated with saline and culled at their terminal endpoint. Gemcitabine “short-term” mice were sacrificed at day 9 from treatment initiation, that is, 1 day upon completion of gemcitabine, and gemcitabine “long-term” mice were allowed to progress and were sacrificed at their terminal endpoint (Fig. 1A). There was no clear separation, in the mice treated with gemcitabine relative to control treated mice, suggesting no survival advantage with gemcitabine (Fig. 1B; Supplementary Fig. S1A). We did not cull any mice at day 9 in the saline group but believe this is irrelevant to the current work as some of the transgenic KPC mice became terminally ill at this time point. However, the mean tumor volume was only marginally decreased in the gemcitabine short-term group and remained the same at the terminal endpoint compared with controls (Supplementary Fig. S1B and S1C, respectively). To identify potential secreted biomarkers that could reveal signaling mechanisms downstream of gemcitabine treatment, cytokine profiling was carried out on the serum from the three groups (Fig. 1C). Several cytokines including TIMP1, IL6, G-CSF, granulocyte–macrophage CSF (GM-CSF); and chemokine (C-X-C motif) ligand 13 (CXCL13), were upregulated early upon treatment with gemcitabine. The levels of TIMP1 were particularly high and its increased levels were sustained until mice reached their terminal endpoint. TIMP1 levels in serum of KPC mice after gemcitabine were confirmed by ELISA in n = 6 mice (Fig. 1D). IHC staining for TIMP1 also confirmed elevated expression of TIMP1 in PDAC tumors following gemcitabine treatment, irrespective of stromal content of the tumors (Fig. 1E), as quantified in (Supplementary Fig. S1D). In addition, TIMP1 mRNA levels were found to be higher in tumors following gemcitabine treatment (Supplementary Fig. S1E). Alternatively, there was no increase in TIMP1 levels in the serum of non-tumor–bearing mice following gemcitabine treatment (Supplementary Fig. S1F), indicating the tumor to be the source of TIMP1. Furthermore, to determine whether TIMP1 played a role in gemcitabine resistance, gemcitabine-resistant KPC cells were generated by culturing cells in progressively increasing concentrations of the drug over a period of 2 months, confirmed by clonogenic survival assay (Supplementary Fig. S1G). Compared with the parental sensitive cell line (KPCS), gemcitabine-resistant cells (KPCR) showed higher expression of TIMP1 in both conditioned media (Fig. 1F) and lysates (Fig. 1G). Moreover, TIMP1 levels further increased in conditioned media (Fig. 1F) following treatment with gemcitabine in both the sensitive and the resistant cell lines. In addition, online datamining confirmed the increased expression of TIMP1 in gemcitabine-resistant lung cancer cells, relative to gemcitabine-sensitive lung cancer cell lines (Fig. 1H; ref. 25). These data indicate that TIMP1 is induced by gemcitabine and that TIMP1 levels are intrinsically higher in gemcitabine-resistant cells compared with its sensitive counterparts, and may play a role in tumor cell resistance to gemcitabine.

Figure 1.

Identification of TIMP1 as a gemcitabine response gene. A, Schematic representation of gemcitabine treatment given to transgenic KPC mice when tumors reached 70–120 mm3. Mice were sacrificed either short-term (24 hours) or long-term (3–7 weeks; when tumors reached 500 mm3 or due to sickness) upon completion of gemcitabine treatment. B, Kaplan–Meier curve showing probability of survival following gemcitabine treatment versus the untreated control group. C, Cytokine arrays and quantification showing protein expression short-term and long-term upon completion of gemcitabine treatment in serum from representative KPC mice from A. D, ELISA showing TIMP1 levels in serum from all KPC mice (n = 6) shown in A. E, H&E and TIMP1 expression in control and gemcitabine-treated tumors (whole face section and selected regions). ELISA showing TIMP1 levels in conditioned media (F) and lysates (G) obtained from the parental KPC cell line (KPCS) and the gemcitabine-resistant KPC cell line (KPCR). H, Oncomine data showing significantly higher TIMP1 expression in gemcitabine-resistant compared with gemcitabine-sensitive lung cancer cells. Log-rank test used for B, unpaired two-tailed t-test in D–F, one-tailed Mann–Whitney used in H. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Figure 1.

Identification of TIMP1 as a gemcitabine response gene. A, Schematic representation of gemcitabine treatment given to transgenic KPC mice when tumors reached 70–120 mm3. Mice were sacrificed either short-term (24 hours) or long-term (3–7 weeks; when tumors reached 500 mm3 or due to sickness) upon completion of gemcitabine treatment. B, Kaplan–Meier curve showing probability of survival following gemcitabine treatment versus the untreated control group. C, Cytokine arrays and quantification showing protein expression short-term and long-term upon completion of gemcitabine treatment in serum from representative KPC mice from A. D, ELISA showing TIMP1 levels in serum from all KPC mice (n = 6) shown in A. E, H&E and TIMP1 expression in control and gemcitabine-treated tumors (whole face section and selected regions). ELISA showing TIMP1 levels in conditioned media (F) and lysates (G) obtained from the parental KPC cell line (KPCS) and the gemcitabine-resistant KPC cell line (KPCR). H, Oncomine data showing significantly higher TIMP1 expression in gemcitabine-resistant compared with gemcitabine-sensitive lung cancer cells. Log-rank test used for B, unpaired two-tailed t-test in D–F, one-tailed Mann–Whitney used in H. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Close modal

TIMP1 is a potential marker of PDAC

Interestingly, analysis of microarray data from 4 different PDAC cohorts revealed a significant upregulation of TIMP1 in human PDAC relative to normal matched pancreatic tissue. In the Iacobuzio-Donahue dataset (26) comprising 12 resected infiltrating PDACs compared with 5 samples of normal pancreas, TIMP1 is among the top 6% of genes upregulated in PDAC versus normal pancreas (P = 0.003; Fig. 2A). In the Segara dataset comprising multiple samples of pancreatic tissue from 11 surgical resections and six matched normal appearing pancreas tissue (27), TIMP1 is in the top 5% of genes upregulated in PDAC relative to normal tissue (P = 4.95E–4; Fig. 2B). In the Badea dataset (28) comparing samples of whole PDAC tissue and matched samples of normal pancreas (n = 39), TIMP1 was overexpressed in the top 1% of upregulated genes (P = 5.03E–15; Fig. 2C). Buchholz and colleagues (29) microdissected surgically resected tissue to remove normal tissue effects and in this dataset we observe TIMP1 is upregulated 3-fold and belongs to the top 2% of upregulated genes in both PanIN-3 lesions and PDAC compared with normal duct and acinar cells, PanIN-1b, and PanIN-2 lesions (Fig. 2D). This indicates that TIMP1 is upregulated at the later stages of PDAC progression, in agreement with recent reports recent that have clearly dismissed the TIMP1 as a marker of early disease (6).

Figure 2.

TIMP1 is a potential marker of PDAC. Log2 median-centered ratio of TIMP1 mRNA expression in publicly-available datasets using Oncomine in n = 12 human PDAC tissues and five samples from the normal pancreas (A); n = 11 surgical resections of human PDAC and six matched normal pancreatic tissue (B); n = 39 samples of human PDAC and matched normal pancreatic tissue (C); and surgically resected tissue, microdissected to separate out six normal pancreatic duct specimens, six PanIN-1b lesions, eight PanIN-2 lesions, ten PanIN-3 lesions, and eight human PDAC specimens (D). **, P < 0.01; ***, P < 0.001.

Figure 2.

TIMP1 is a potential marker of PDAC. Log2 median-centered ratio of TIMP1 mRNA expression in publicly-available datasets using Oncomine in n = 12 human PDAC tissues and five samples from the normal pancreas (A); n = 11 surgical resections of human PDAC and six matched normal pancreatic tissue (B); n = 39 samples of human PDAC and matched normal pancreatic tissue (C); and surgically resected tissue, microdissected to separate out six normal pancreatic duct specimens, six PanIN-1b lesions, eight PanIN-2 lesions, ten PanIN-3 lesions, and eight human PDAC specimens (D). **, P < 0.01; ***, P < 0.001.

Close modal

TIMP1 knockdown decreases clonogenic survival and enhances therapy response

To elucidate the role of TIMP1, we stably knocked down TIMP1 expression using five different lentiviral shRNA constructs each in the human PDAC cell line PANC1 and the murine PDAC cell line KPC (Supplementary Fig. S2A and S2B). The cell lines with the highest knockdowns were confirmed by qPCR (Supplementary Fig. S2C and S2D) as well as by Western blotting (Supplementary Fig. S2E) besides ELISA (Fig. 3A and B). TIMP1 knockdown resulted in a decrease in colony formation in both cell lines (Fig. 3C and D); quantified in (Supplementary Fig. S2F and S2G). In addition, the surviving fraction in TIMP1 knockdown sensitized both cell lines to gemcitabine and radiation compared with untreated/DMSO controls, (Fig. 3E–H). We overexpressed TIMP1 retrovirally in our WT and KD cell line alongside the appropriate empty retroviral vector (Fig. 3I). Overexpression of TIMP1 resulted in a rescue of cell growth and desensitized cells to radiation and gemcitabine (Fig. 3J–L). Representative images are shown in (Supplementary Fig. S2H–S2J). We injected PANC1 and KPC TIMP1 WT and KD subcutaneously into SCID and C57bl/6 mice, respectively, and noted that the time it took for the tumors to reach a volume of 300 mm3 was significantly longer in the TIMP1 KD group compared with TIMP1 WT cells (Fig. 3M). We found no differences in the amount of necrosis or stroma between the two groups in PANC1 and KPC tumors (Supplementary Fig. S2K–S2N). With the KPC cells, the time taken for tumors to reach maximum volume (700 mm3) was significantly longer in the TIMP1 KD group (Fig. 3N). To confirm whether our in vitro chemo/radiosensitization was replicated in vivo, we treated mice with 8 Gy or 100 mg/kg gemcitabine. In keeping with our in vitro data, tumor growth was significantly delayed (Fig. 3O and P). Taken together, these results show that TIMP1 facilitates primary tumor growth, associated with lower response to gemcitabine and radiation both in vitro and in our in vivo subcutaneous models.

Figure 3.

TIMP1 knockdown results in decreased clonogenic survival and sensitization of cancer cells to chemotherapy and radiotherapy. ELISA showing TIMP1 expression in PANC1 cells (A) and KPC cells (B) having a stable knockdown of TIMP1 and cells transfected with a control empty vector. C and D, Clonogenic surviving fractions in PANC1 and KPC cells described in A and B, respectively, relative to untreated/DMSO controls, respectively. E–H, Clonogenic surviving fractions in PANC1 and KPC cells after radiation and gemcitabine, relative to untreated/DMSO controls, respectively. I, Western blot analysis showing stable overexpression of TIMP1 in WT and KD cells. J–L, Clonogenic survival assay in PANC1 cells described in I. M, Time to maximum tumor volume and to 300 mm3 in PANC1 TIMP1 WT and KD cells injected subcutaneously into SCID mice. N–P, Time taken to 300 mm3 in KPC TIMP1 WT and KD cells injected into C57Bl/6 mice in untreated, irradiated 8 Gy, and gemcitabine-treated (100 mg/kg, i.p.) mice. Treatment was initiated when tumors reached 100 mm3. Unpaired two-tailed t test were performed for AL. Log-rank and Mann–Whitney (two-tailed) nonparametric tests were used for MP. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Figure 3.

TIMP1 knockdown results in decreased clonogenic survival and sensitization of cancer cells to chemotherapy and radiotherapy. ELISA showing TIMP1 expression in PANC1 cells (A) and KPC cells (B) having a stable knockdown of TIMP1 and cells transfected with a control empty vector. C and D, Clonogenic surviving fractions in PANC1 and KPC cells described in A and B, respectively, relative to untreated/DMSO controls, respectively. E–H, Clonogenic surviving fractions in PANC1 and KPC cells after radiation and gemcitabine, relative to untreated/DMSO controls, respectively. I, Western blot analysis showing stable overexpression of TIMP1 in WT and KD cells. J–L, Clonogenic survival assay in PANC1 cells described in I. M, Time to maximum tumor volume and to 300 mm3 in PANC1 TIMP1 WT and KD cells injected subcutaneously into SCID mice. N–P, Time taken to 300 mm3 in KPC TIMP1 WT and KD cells injected into C57Bl/6 mice in untreated, irradiated 8 Gy, and gemcitabine-treated (100 mg/kg, i.p.) mice. Treatment was initiated when tumors reached 100 mm3. Unpaired two-tailed t test were performed for AL. Log-rank and Mann–Whitney (two-tailed) nonparametric tests were used for MP. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Close modal

Cell adhesion, chemotaxis, and inflammatory pathways are altered by TIMP1 expression

To identify potential tumorigenic pathways affected by TIMP1 signaling, we carried out a microarray and analyzed gene expression in TIMP1 WT versus KD PANC1 cells. In total, 136 differentially regulated genes were identified (Fig. 4). Gene annotation studies indicated enriched expression of inflammatory transcripts, cell adhesion transcripts, and chemotaxis in TIMP1 KD cells (Table 1). Representative genes were validated by real-time qPCR (Supplementary Fig. S3A–S3D).

Figure 4.

Differential expression analysis reveals that expression of 136 genes is significantly altered in TIMP1 KD. Gene ontology enrichment analysis using GeneCodis identified enrichment of the biological processes cell adhesion, chemotaxis, and inflammatory response. A, Heatmap of the top 50 differently expressed genes in PANC1 TIMP1 WT versus TIMP1 KD cells on the Illumina Human H12 array.

Figure 4.

Differential expression analysis reveals that expression of 136 genes is significantly altered in TIMP1 KD. Gene ontology enrichment analysis using GeneCodis identified enrichment of the biological processes cell adhesion, chemotaxis, and inflammatory response. A, Heatmap of the top 50 differently expressed genes in PANC1 TIMP1 WT versus TIMP1 KD cells on the Illumina Human H12 array.

Close modal
Table 1.

Gene Ontology enrichment analysis (biological processes)

Inflammatory response (P = 0.00011)
BMP2 Bone morphogenetic protein 2 
OLR1 Oxidized low density lipoprotein (lectin-like) receptor 1 
S100A8 S100 calcium binding protein A8 
S100A9 S100 calcium binding protein A9 
AOX1 Aldehyde oxidase 1 
AOC3 Amine oxidase, copper containing 3 (vascular adhesion protein 1) 
CCR4 Chemokine (C-C motif) receptor 4 
TPST1 Tyrosylprotein sulfotransferase 1 
RIPK2 Receptor-interacting serine-threonine kinase 2 
Cell adhesion (P = 0.03002) 
FEZ1 Fasciculation and elongation protein zeta 1 (zygin I) 
OLR1 Oxidized low density lipoprotein (lectin-like) receptor 1 
SRPX Sushi-repeat containing protein, X-linked 
COL8A1 Collagen, type VIII, alpha 1 
AOC3 Amine oxidase, copper containing 3 (vascular adhesion protein 1) 
ITGA6 Integrin, alpha 6 
ITGA1 Integrin, alpha 1 
PTPRU Protein tyrosine phosphatase, receptor type, U 
Chemotaxis (P = 0.01593) 
CCR4 C-C motif chemokine receptor 4 
DEFB1 Defensin beta 1 
CXCL16 C-X-C motif chemokine ligand 16 
S100A8 S100 calcium binding protein A8 
S100A9 S100 calcium binding protein A9 
Inflammatory response (P = 0.00011)
BMP2 Bone morphogenetic protein 2 
OLR1 Oxidized low density lipoprotein (lectin-like) receptor 1 
S100A8 S100 calcium binding protein A8 
S100A9 S100 calcium binding protein A9 
AOX1 Aldehyde oxidase 1 
AOC3 Amine oxidase, copper containing 3 (vascular adhesion protein 1) 
CCR4 Chemokine (C-C motif) receptor 4 
TPST1 Tyrosylprotein sulfotransferase 1 
RIPK2 Receptor-interacting serine-threonine kinase 2 
Cell adhesion (P = 0.03002) 
FEZ1 Fasciculation and elongation protein zeta 1 (zygin I) 
OLR1 Oxidized low density lipoprotein (lectin-like) receptor 1 
SRPX Sushi-repeat containing protein, X-linked 
COL8A1 Collagen, type VIII, alpha 1 
AOC3 Amine oxidase, copper containing 3 (vascular adhesion protein 1) 
ITGA6 Integrin, alpha 6 
ITGA1 Integrin, alpha 1 
PTPRU Protein tyrosine phosphatase, receptor type, U 
Chemotaxis (P = 0.01593) 
CCR4 C-C motif chemokine receptor 4 
DEFB1 Defensin beta 1 
CXCL16 C-X-C motif chemokine ligand 16 
S100A8 S100 calcium binding protein A8 
S100A9 S100 calcium binding protein A9 

NOTE: Top three biological processes (inflammatory response, cell adhesion, and chemotaxis) identified to be enriched by the gene ontology enrichment analysis and their associated differentially expressed genes (HGNC symbol and description). The enrichment P value is corrected for multiple testing using false discovery rate.

TIMP1 knockdown results in decreased migration and liver metastasis

To verify whether the role of TIMP1 in chemotaxis translated to an increased migratory capacity, we compared TIMP1 WT and KD cells in a wound scratch assay. TIMP1 expression resulted in a stronger migratory phenotype in both PANC1 (Fig. 5A) and KPC cells (Fig. 5B) in the scratch wound assay quantified in (Supplementary Fig. S4A). Also, both PANC1 and KPC TIMP1 KD cells showed decreased invasive capacity towards CAF compared with their WT counterparts (Supplementary Fig. S4B). TIMP1 did not affect the expression of phosphokinases in PANC1 cells in vitro (Supplementary Fig. S4C). TIMP1 has been previously reported to enhance angiogenesis (30, 31). In our primary subcutaneous animal models, tumors derived from both PANC1 and KPC TIMP1 KD cells showed decreased blood vessel density staining compared with WT groups (Fig. 5C and D). As our gene enrichment analysis identified transcripts involved in cell adhesion and chemotaxis, we examined whether TIMP1 expression in PDAC cells could affect their metastatic potential in vivo. As the liver is the most common site of metastatic spread in PDAC, we injected PANC1 TIMP1 WT and KD cells intrasplenically into SCID mice as reported (32). MRI imaging was used to monitor liver metastasis (Supplementary Fig. S4D and S4E). TIMP1 KD resulted in significantly reduced hepatic metastatic burden (Fig. 5E and F) with lower vascular density within the metastatic regions (Fig. 5G). To verify our results in a different mouse model, we injected KPC TIMP1 WT and KD cells intrasplenically into C57Bl/6 mice. The hepatic metastatic burden was significantly lower following TIMP1 suppression (Fig. 5H). H&E staining confirmed the presence of metastasis in the liver (Fig. 5I) and blood vessel density was also significantly reduced (Fig. 5J). To address pathways that may be involved in the metastatic process, we performed a protein array chIP, but found that pathway activation was indistinguishable from controls (Supplementary Fig. S4C)

Figure 5.

TIMP1 knockdown results in decreased migration and metastasis migration wound scratch assays in PANC1 TIMP1 WT and KD cells (A), and KPC TIMP1 WT and KD cells (B). CD31 immunofluorescence images taken at ×10 magnification showing blood vessel density and quantification in PANC1 TIMP1 WT and KD xenografts in SCID mice (C), and KPC TIMP1 WT and KD allografts in C57Bl/6 mice (D). E, Representative whole liver images of SCID mice resected 14 days after intrasplenic injection of PANC1 TIMP1 WT and KD cells, together with the corresponding liver weights as a percentage of total body weight. Representative H&E staining at ×10 and ×20 magnification (F) and the corresponding CD31 images at ×10 and ×20 magnification of the matched areas for CD31 (G). H, Representative whole liver images of C57Bl/6 mice resected 14 days after intrasplenic injection of KPC TIMP1 WT and KD cells, with the corresponding liver weights as a percentage of total body weight. Representative H&E staining at ×10 magnification (I) and the corresponding CD31 images at ×10 and ×20 magnification (J) of the matched areas for CD31. Unpaired two-tailed t test was performed for C and D. Mann–Whitney (two-tailed) nonparametric tests used for E and H. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Figure 5.

TIMP1 knockdown results in decreased migration and metastasis migration wound scratch assays in PANC1 TIMP1 WT and KD cells (A), and KPC TIMP1 WT and KD cells (B). CD31 immunofluorescence images taken at ×10 magnification showing blood vessel density and quantification in PANC1 TIMP1 WT and KD xenografts in SCID mice (C), and KPC TIMP1 WT and KD allografts in C57Bl/6 mice (D). E, Representative whole liver images of SCID mice resected 14 days after intrasplenic injection of PANC1 TIMP1 WT and KD cells, together with the corresponding liver weights as a percentage of total body weight. Representative H&E staining at ×10 and ×20 magnification (F) and the corresponding CD31 images at ×10 and ×20 magnification of the matched areas for CD31 (G). H, Representative whole liver images of C57Bl/6 mice resected 14 days after intrasplenic injection of KPC TIMP1 WT and KD cells, with the corresponding liver weights as a percentage of total body weight. Representative H&E staining at ×10 magnification (I) and the corresponding CD31 images at ×10 and ×20 magnification (J) of the matched areas for CD31. Unpaired two-tailed t test was performed for C and D. Mann–Whitney (two-tailed) nonparametric tests used for E and H. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Close modal

TIMP1 level in blood has been considered as a prognostic factor in patients with liver metastasis (33). We harvested blood from C57Bl/6 mice at days 4, 7, and 14 postintrasplenic injection of KPC cells and found significantly increased circulating TIMP1 with increased metastatic burden (Fig. 6A). We conclude that TIMP1 expression correlates with enhanced vascular density and is essential for tumor cell migration and metastatic growth.

Figure 6.

TIMP1 enhances the recruitment of MDSCs and Tregs in liver metastases. A, Time-course of TIMP1 levels in serum of KPC parental cells injected intrasplenically into C57Bl/6. B, FACS analysis quantifications of immune marker populations in livers of mice described in A. FACS analysis quantifications of immune myeloid cell populations in livers of SCID mice injected intrasplenically with PANC1 TIMP1-WT and KD cells (C) and C57Bl/6 mice injected with KPC TIMP1 WT or KD cells (D). y-axis illustrates the percentage of gated cells as indicated. Unpaired two-tailed t test was performed for Fig. 6. *, P < 0.05; **, P < 0.01; ***, P < 0.001. ns, nonsignificant.

Figure 6.

TIMP1 enhances the recruitment of MDSCs and Tregs in liver metastases. A, Time-course of TIMP1 levels in serum of KPC parental cells injected intrasplenically into C57Bl/6. B, FACS analysis quantifications of immune marker populations in livers of mice described in A. FACS analysis quantifications of immune myeloid cell populations in livers of SCID mice injected intrasplenically with PANC1 TIMP1-WT and KD cells (C) and C57Bl/6 mice injected with KPC TIMP1 WT or KD cells (D). y-axis illustrates the percentage of gated cells as indicated. Unpaired two-tailed t test was performed for Fig. 6. *, P < 0.05; **, P < 0.01; ***, P < 0.001. ns, nonsignificant.

Close modal

TIMP1 promotes myeloid cell recruitment and Tregs in liver metastasis

TIMP1 can promote immunosuppression (13). Our microarray data identified inflammatory pathways altered by TIMP1. Hence, we queried whether there were any differences in immunosuppressive myeloid populations in the tumor microenvironment that are known to promote malignant growth. While we failed to identify significant changes in primary subcutaneous PANC1 and KPC mouse models following TIMP1 suppression (Supplementary Fig. S5A and S5B), we noted distinct changes in the immune profile of metastatic livers. When we examined the immune profile in livers harboring metastases at day 4, 7, and 14 following intrasplenic injection of KPC cells in C57Bl/6 mice, no macroscopic metastasis was observed at day 4 or day 7, but metastases were evident by day 14 and CD45+CXCR2+ neutrophils increased with progressive metastatic development (Fig. 6B). Taken together, these results suggest that increasing TIMP1 levels during metastasis formation are associated with increased liver infiltration by myeloid cells.

When we investigated the immune profile in liver metastases following intrasplenic injection of TIMP1 WT and KD PANC1 cells in SCID mice (Fig. 5E) we noted a significant decrease in CD11b+Gr1+ myeloid-derived suppressor cells (MDSC) and neutrophil infiltration following TIMP1 suppression (Fig. 6C). Similarly, MDSCs and CD4+CD25+FoxP3+ regulatory T cells were decreased in liver metastases of C57Bl/6 mice intrasplenically injected with TIMP1 KD KPC cells compared with WT controls (Fig. 6D). Taken together, our results suggest that the presence of TIMP1 expressing cells is associated with hepatic recruitment of immunosuppressive populations. To query whether the differences in immune cell infiltration was due to differences in metastatic burden, we compared immune cell infiltration in metastasis of equal size and found most of the MDSCs to indeed localize to the tumor and to be of equal percentage. This is indicative of a linear relationship between TIMP1 expression, liver metastatic burden, and immune cell infiltration (Supplementary Fig. S5C and S5D). This would imply that the presence of tumor cells themselves may be required for immune cell infiltration.

Gemcitabine constitutes one of the backbones in the treatment of advanced PDAC but patients often respond poorly to this agent. We identified TIMP1 as a gemcitabine response gene in serum taken from transgenic KPC mice. IHC staining revealed increased TIMP1 protein expression independently of the stromal content in tumor after gemcitabine treatment. This was also confirmed by transcriptional analysis showing an increase in TIMP1 mRNA levels in tumors from gemcitabine-treated mice. Although no direct evidence in PDAC, previous analysis of 264 breast cancer patients demonstrated longer overall survival after gemcitabine-incorporating chemotherapy in patients with TIMP1-negative tumors compared with TIMP1-positive cases (34). Additional studies have correlated high TIMP1 expression with rapid tumor progression after treatment with chemotherapy agents such as cyclophosphamide methotrexate, 5-fluorouracil, epirubicin, and doxorubicin in breast and colon cancers (35, 36). Furthermore, we found TIMP1 expression to be among the top 1% of upregulated genes in human PDAC cohorts and it was significantly upregulated in human PDAC compared with normal pancreatic tissue. Of note, treatment of nontumor–bearing mice with gemcitabine did not alter TIMP1 levels, suggesting that the presence of tumor cells is necessary for TIMP1 upregulation upon chemotherapy with gemcitabine. These data indicate that TIMP1 expression is correlated with tumor progression and decreased response to gemcitabine in PDAC.

We found that TIMP1 expression is important for tumor cell survival as evidenced from decreased colony formation following stable knockdown of TIMP1 in both human and murine PDAC cell lines. Also, overexpression of TIMP1 enhanced clonogenic survival in untreated cells, and also after gemcitabine and radiotherapy that further supports its protumorigenic role. TIMP1 can activate prosurvival and invasive signaling through engaging CD63, activating PI3K/Akt signaling (8–11). Besides this, TIMP1 can regulate cell proliferation by translocating to the nucleus and interacting with the zinc finger protein PLZF (37). Of note, TIMP1 expression occurs downstream of the K-Ras signaling in the transgenic KPC model (17). Inhibition of TIMP1 resulted in improved response of PDAC cells to ionizing radiation and gemcitabine. Accordingly, Oncomine database analysis revealed significant TIMP1 upregulation in gemcitabine-resistant lung cancer cell lines compared with gemcitabine-sensitive counterparts. This is in line with our findings and others demonstrating a correlation between TIMP1 and decreased sensitivity of cell lines to chemotherapy (38).

Previous studies have reported a link between TIMP1 and angiogenesis. TIMP1-overexpressing tumors displayed increased VEGF-A expression (39), augmented growth rates with increased vessel density (40), and metastatic colonization in murine models of lung cancer (41). Inhibition of secreted TIMP1 decreased myofibroblast-induced angiogenesis (42). Also, TIMP1-positive cells have been observed adjacent to CD34+ blood vessels in colorectal liver metastasis (43). In agreement, we now show that TIMP1 suppression results in decreased vessel density, both in human and murine PDAC xenograft and liver metastasis models. Furthermore, we identified alterations in chemotaxis pathways upon knockdown of TIMP1, thus supporting a protumorigenic and prometastatic role for TIMP1 via alteration of chemotaxis and the possible promotion of angiogenesis.

We also found that expression of genes playing a role in inflammatory pathways appeared most dependent on TIMP1 levels. Inflammation is a critical component of tumor progression (44). The protumorigenic role of TIMP1 has been previously attributed to its cytokine-like functions that form an inflammatory milieu (45). In addition to TIMP1 upregulation, we noted an increase in the inflammatory cytokine IL6 expression following gemcitabine treatment in KPC mice. Of note, IL6 promotes progression of PanINs into PDAC (46, 47), whereas TIMP1 expression is downstream of IL6 in rat hepatocytes (46). Also, the TIMP1 promoter is a direct target of the inflammatory mediator NFκB (46). Thus, administration of gemcitabine appears to induce an inflammatory response, including TIMP1 upregulation in PDAC.

TIMP1 can promote an immunosuppressive environment via SDF-1–dependent recruitment of neutrophils to the liver and creating a premetastatic niche (13). Suppression of TIMP1 reduced PDAC cell migration, and also invasion of PDAC cells toward CAFs (immortalized PSCs), indicating a prometastatic role for TIMP1. Furthermore, by using an intrasplenic injection model, we observe that circulating PDAC tumor cells require TIMP1 expression to support hepatic colonization associated with recruitment of CXCR2-positive neutrophils, CD11b+Gr1+ MDSCs and CD4+CD25+FOXP3+ regulatory T cells to the liver. Inhibition of CXCR2 abrogates metastatic seeding (19), whereas Tregs are often encountered in fibrotic tissue and can promote liver metastases (48, 49). Notably, examination of small liver metastases with comparable size between TIMP1 WT and KD groups failed to reveal a significant difference of MDSCs or Tregs between the two groups, indicative of a linear relationship between TIMP1 expression, liver metastatic burden, and immune cell infiltration. This is suggestive that the presence of tumor cells is essential for immune cell infiltration (1, 49, 50).

To summarize, treatment of PDAC with gemcitabine led to upregulation of the cytokine TIMP1. TIMP1 expression correlates with tumor progression in a number of patient cohorts analyzed. Suppression of TIMP1 enhanced tumor response to gemcitabine and radiotherapy, and significantly decreased clonogenic survival, migration, and invasion both in vitro and in vivo. Importantly, TIMP1 suppression was associated with decreased vascular density in the tumor xenografts and liver metastasis, and reduced liver infiltration by immunosuppressive cell populations, resulting in an overall reduction in liver metastasis formation. Our study therefore provides a rationale for the design and addition of TIMP1-specific inhibitors to chemo/radiotherapy.

No potential conflicts of interest were disclosed.

Conception and design: Z.D'Costa, W.G. McKenna, E. O'Neill, E. Fokas

Development of methodology: Z.D'Costa, R.V. Stiphout, S.Y. Lim, P. Kinchesh, S.C. Smart, W.G. McKenna, E. O'Neill, E. Fokas

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): Z.D'Costa, K. Jones, A. Azad, S.Y. Lim, A.L. Gomes, S.C. Smart, E. O'Neill

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): Z.D'Costa, K. Jones, A. Azad, R.V. Stiphout, S.Y. Lim, A.L. Gomes, S.C. Smart, W.G. McKenna, F.M. Buffa, O.J. Sansom, E. O'Neill, E. Fokas

Writing, review, and/or revision of the manuscript: Z.D'Costa, R.V. Stiphout, S.Y. Lim, A.L. Gomes, P. Kinchesh, S.C. Smart, W.G. McKenna, O.J. Sansom, E. O'Neill, E. Fokas

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): Z.D'Costa, E. Fokas

Study supervision: W.G. McKenna, R.J. Muschel, E. O'Neill, E. Fokas

We thank Mick Woodcock, Angela Diana, and Graham Brown for the technical support.

This work was funded by Cancer Research UK (CRUK C5255/A15935; CRUK A19277). Z.C. D'Costa, A.K. Azad, W.G. McKenna, and E. Fokas were also funded by the Kidani Memorial Trust.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Fokas
E
,
O'Neill
E
,
Gordon-Weeks
A
,
Mukherjee
S
,
McKenna
WG
,
Muschel
RJ
. 
Pancreatic ductal adenocarcinoma: from genetics to biology to radiobiology to oncoimmunology and all the way back to the clinic
.
Biochim Biophys Acta
2015
;
1855
:
61
82
.
2.
Hidalgo
M
. 
Pancreatic cancer
.
N Eng J Med
2010
;
362
:
1605
17
.
3.
Mini
E
,
Nobili
S
,
Caciagli
B
,
Landini
I
,
Mazzei
T
. 
Cellular pharmacology of gemcitabine
.
Ann Oncol
2006
;
17
:
v7
12
.
4.
Ryan
DP
,
Hong
TS
,
Bardeesy
N
. 
Pancreatic adenocarcinoma
.
N Eng J Med
2014
;
371
:
2140
1
.
5.
Gilbert
LA
,
Hemann
MT
. 
DNA damage-mediated induction of a chemoresistant niche
.
Cell
2010
;
143
:
355
66
.
6.
Jenkinson
C
,
Elliott
V
,
Menon
U
,
Apostolidou
S
,
Fourkala
OE
,
Gentry-Maharaj
A
, et al
Evaluation in pre-diagnosis samples discounts ICAM-1 and TIMP1 as biomarkers for earlier diagnosis of pancreatic cancer
.
J Proteomics
2015
;
113
:
400
2
.
7.
Stetler-Stevenson
WG
. 
Tissue inhibitors of metalloproteinases in cell signaling: metalloproteinase-independent biological activities
.
Sci Signal
2008
;
1
:
re6
.
8.
Jung
KK
,
Liu
XW
,
Chirco
R
,
Fridman
R
,
Kim
HR
. 
Identification of CD63 as a tissue inhibitor of metalloproteinase-1 interacting cell surface protein
.
EMBO J
2006
;
25
:
3934
42
.
9.
Toricelli
M
,
Melo
FH
,
Peres
GB
,
Silva
DC
,
Jasiulionis
MG
. 
Timp1 interacts with beta-1 integrin and CD63 along melanoma genesis and confers anoikis resistance by activating PI3-K signaling pathway independently of Akt phosphorylation
.
Mol Cancer
2013
;
12
:
22
.
10.
Lee
SY
,
Kim
JM
,
Cho
SY
,
Kim
HS
,
Shin
HS
,
Jeon
JY
, et al
TIMP1 modulates chemotaxis of human neural stem cells through CD63 and integrin signalling
.
Biochem J
2014
;
459
:
565
76
.
11.
D'Angelo
RC
,
Liu
XW
,
Najy
AJ
,
Jung
YS
,
Won
J
,
Chai
KX
, et al
TIMP1 via TWIST1 induces EMT phenotypes in human breast epithelial cells
.
Mol Cancer Res
2014
;
12
:
1324
33
.
12.
Brew
K
,
Nagase
H
. 
The tissue inhibitors of metalloproteinases (TIMPs): an ancient family with structural and functional diversity
.
Biochim Biophys Acta
2010
;
1803
:
55
71
.
13.
Seubert
B
,
Grunwald
B
,
Kobuch
J
,
Cui
H
,
Schelter
F
,
Schaten
S
, et al
Tissue inhibitor of metalloproteinases (TIMP)-1 creates a premetastatic niche in the liver through SDF-1/CXCR4-dependent neutrophil recruitment in mice
.
Hepatology
2015
;
61
:
238
48
.
14.
Schrohl
AS
,
Holten-Andersen
MN
,
Peters
HA
,
Look
MP
,
Meijer-van Gelder
ME
,
Klijn
JG
, et al
Tumor tissue levels of tissue inhibitor of metalloproteinase-1 as a prognostic marker in primary breast cancer
.
Clin Cancer Res
2004
;
10
:
2289
98
.
15.
Birgisson
H
,
Nielsen
HJ
,
Christensen
IJ
,
Glimelius
B
,
Brunner
N
. 
Preoperative plasma TIMP1 is an independent prognostic indicator in patients with primary colorectal cancer: a prospective validation study
.
Eur J Cancer
2010
;
46
:
3323
31
.
16.
Roy
R
,
Zurakowski
D
,
Wischhusen
J
,
Frauenhoffer
C
,
Hooshmand
S
,
Kulke
M
, et al
Urinary TIMP1 and MMP-2 levels detect the presence of pancreatic malignancies
.
Br J Cancer
2014
;
111
:
1772
9
.
17.
Botta
GP
,
Reichert
M
,
Reginato
MJ
,
Heeg
S
,
Rustgi
AK
,
Lelkes
PI
. 
ERK2-regulated TIMP1 induces hyperproliferation of K-Ras(G12D)-transformed pancreatic ductal cells
.
Neoplasia
2013
;
15
:
359
72
.
18.
Morton
JP
,
Timpson
P
,
Karim
SA
,
Ridgway
RA
,
Athineos
D
,
Doyle
B
, et al
Mutant p53 drives metastasis and overcomes growth arrest/senescence in pancreatic cancer
.
Proc Natl Acad Sci U S A
2010
;
107
:
246
51
.
19.
Steele
CW
,
Karim
SA
,
Leach
JD
,
Bailey
P
,
Upstill-Goddard
R
,
Rishi
L
, et al
CXCR2 inhibition profoundly suppresses metastases and augments immunotherapy in pancreatic ductal adenocarcinoma
.
Cancer Cell
2016
;
29
:
832
45
.
20.
Hingorani
SR
,
Wang
L
,
Multani
AS
,
Combs
C
,
Deramaudt
TB
,
Hruban
RH
, et al
Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice
.
Cancer Cell
2005
;
7
:
469
83
.
21.
Fokas
E
,
Prevo
R
,
Pollard
JR
,
Reaper
PM
,
Charlton
PA
,
Cornelissen
B
, et al
Targeting ATR in vivo using the novel inhibitor VE-822 results in selective sensitization of pancreatic tumors to radiation
.
Cell Death Dis
2012
;
3
:
e441
.
22.
Lim
SY
,
Yuzhalin
AE
,
Gordon-Weeks
AN
,
Muschel
RJ
. 
Tumor-infiltrating monocytes/macrophages promote tumor invasion and migration by upregulating S100A8 and S100A9 expression in cancer cells
.
Oncogene
2016
;
35
:
5735
45
.
23.
Kinchesh P
GS
,
Gomes
AL
,
Kersemans
V
,
Beech
J
,
Allen
D
,
Smart
S
. 
Accelerated imaging of the mouse body using k-space segmentation, cardio-respiratory synchronisation and short, constant TR: application to b-SSFP
.
Proc Intl Soc Mag Reson Med
2016
;
24
:
1825
.
24.
Prevo
R
,
Deutsch
E
,
Sampson
O
,
Diplexcito
J
,
Cengel
K
,
Harper
J
, et al
Class I PI3 kinase inhibition by the pyridinylfuranopyrimidine inhibitor PI-103 enhances tumor radiosensitivity
.
Cancer Res
2008
;
68
:
5915
23
.
25.
Gemma
A
,
Li
C
,
Sugiyama
Y
,
Matsuda
K
,
Seike
Y
,
Kosaihira
S
, et al
Anticancer drug clustering in lung cancer based on gene expression profiles and sensitivity database
.
BMC Cancer
2006
;
6
:
174
.
26.
Iacobuzio-Donahue
CA
,
Maitra
A
,
Olsen
M
,
Lowe
AW
,
van Heek
NT
,
Rosty
C
, et al
Exploration of global gene expression patterns in pancreatic adenocarcinoma using cDNA microarrays
.
Am J Pathol
2003
;
162
:
1151
62
.
27.
Segara
D
,
Biankin
AV
,
Kench
JG
,
Langusch
CC
,
Dawson
AC
,
Skalicky
DA
, et al
Expression of HOXB2, a retinoic acid signaling target in pancreatic cancer and pancreatic intraepithelial neoplasia
.
Clin Cancer Res
2005
;
11
:
3587
96
.
28.
Badea
L
,
Herlea
V
,
Dima
SO
,
Dumitrascu
T
,
Popescu
I
. 
Combined gene expression analysis of whole-tissue and microdissected pancreatic ductal adenocarcinoma identifies genes specifically overexpressed in tumor epithelia
.
Hepato-gastroenterology
2008
;
55
:
2016
27
.
29.
Buchholz
M
,
Braun
M
,
Heidenblut
A
,
Kestler
HA
,
Kloppel
G
,
Schmiegel
W
, et al
Transcriptome analysis of microdissected pancreatic intraepithelial neoplastic lesions
.
Oncogene
2005
;
24
:
6626
36
.
30.
Yamada
E
,
Tobe
T
,
Yamada
H
,
Okamoto
N
,
Zack
DJ
,
Werb
Z
, et al
TIMP1 promotes VEGF-induced neovascularization in the retina
.
Histol Histopathol
2001
;
16
:
87
97
.
31.
Liu
H
,
Chen
B
,
Lilly
B
. 
Fibroblasts potentiate blood vessel formation partially through secreted factor TIMP1
.
Angiogenesis
2008
;
11
:
223
34
.
32.
Zhao
L
,
Lim
SY
,
Gordon-Weeks
AN
,
Tapmeier
TT
,
Im
JH
,
Cao
Y
, et al
Recruitment of a myeloid cell subset (CD11b/Gr1 mid) via CCL2/CCR2 promotes the development of colorectal cancer liver metastasis
.
Hepatology
2013
;
57
:
829
39
.
33.
Bunatova
K
,
Pesta
M
,
Kulda
V
,
Topolcan
O
,
Vrzalova
J
,
Sutnar
A
, et al
Plasma TIMP1 level is a prognostic factor in patients with liver metastases
.
Anticancer Res
2012
;
32
:
4601
6
.
34.
Jorgensen
CL
,
Bjerre
C
,
Ejlertsen
B
,
Bjerre
KD
,
Balslev
E
,
Bartels
A
, et al
TIMP1 and responsiveness to gemcitabine in advanced breast cancer; results from a randomized phase III trial from the Danish breast cancer cooperative group
.
BMC Cancer
2014
;
14
:
360
.
35.
Schrohl
AS
,
Meijer-van Gelder
ME
,
Holten-Andersen
MN
,
Christensen
IJ
,
Look
MP
,
Mouridsen
HT
, et al
Primary tumor levels of tissue inhibitor of metalloproteinases-1 are predictive of resistance to chemotherapy in patients with metastatic breast cancer
.
Clin Cancer Res
2006
;
12
:
7054
8
.
36.
Sorensen
NM
,
Bystrom
P
,
Christensen
IJ
,
Berglund
A
,
Nielsen
HJ
,
Brunner
N
, et al
TIMP1 is significantly associated with objective response and survival in metastatic colorectal cancer patients receiving combination of irinotecan, 5-fluorouracil, and folinic acid
.
Clin Cancer Res
2007
;
13
:
4117
22
.
37.
Rho
SB
,
Chung
BM
,
Lee
JH
. 
TIMP1 regulates cell proliferation by interacting with the ninth zinc finger domain of PLZF
.
J Cell Biochem
2007
;
101
:
57
67
.
38.
Davidsen
ML
,
Wurtz
SO
,
Romer
MU
,
Sorensen
NM
,
Johansen
SK
,
Christensen
IJ
, et al
TIMP1 gene deficiency increases tumour cell sensitivity to chemotherapy-induced apoptosis
.
Br J Cancer
2006
;
95
:
1114
20
.
39.
Cui
H
,
Seubert
B
,
Stahl
E
,
Dietz
H
,
Reuning
U
,
Moreno-Leon
L
, et al
Tissue inhibitor of metalloproteinases-1 induces a pro-tumourigenic increase of miR-210 in lung adenocarcinoma cells and their exosomes
.
Oncogene
2015
;
34
:
3640
50
.
40.
Rojiani
MV
,
Ghoshal-Gupta
S
,
Kutiyanawalla
A
,
Mathur
S
,
Rojiani
AM
. 
TIMP1 overexpression in lung carcinoma enhances tumor kinetics and angiogenesis in brain metastasis
.
J Neuropathol Exp Neurol
2015
;
74
:
293
304
.
41.
Chang
YH
,
Chiu
YJ
,
Cheng
HC
,
Liu
FJ
,
Lai
WW
,
Chang
HJ
, et al
Down-regulation of TIMP1 inhibits cell migration, invasion, and metastatic colonization in lung adenocarcinoma
.
Tumour Biol
2015
;
36
:
3957
67
.
42.
Mayrand
D
,
Laforce-Lavoie
A
,
Larochelle
S
,
Langlois
A
,
Genest
H
,
Roy
M
, et al
Angiogenic properties of myofibroblasts isolated from normal human skin wounds
.
Angiogenesis
2012
;
15
:
199
212
.
43.
Illemann
M
,
Eefsen
RH
,
Bird
NC
,
Majeed
A
,
Osterlind
K
,
Laerum
OD
, et al
Tissue inhibitor of matrix metalloproteinase-1 expression in colorectal cancer liver metastases is associated with vascular structures
.
Mol Carcinogen
2016
;
55
:
193
208
.
44.
Coussens
LM
,
Zitvogel
L
,
Palucka
AK
. 
Neutralizing tumor-promoting chronic inflammation: a magic bullet?
Science
2013
;
339
:
286
91
.
45.
Rossi
L
,
Forte
D
,
Migliardi
G
,
Salvestrini
V
,
Buzzi
M
,
Ricciardi
MR
, et al
The tissue inhibitor of metalloproteinases 1 increases the clonogenic efficiency of human hematopoietic progenitor cells through CD63/PI3K/Akt signaling
.
Exp Hematol
2015
;
43
:
974
85.e1
.
46.
Roeb
E
,
Graeve
L
,
Mullberg
J
,
Matern
S
,
Rose-John
S
. 
TIMP1 protein expression is stimulated by IL-1 beta and IL-6 in primary rat hepatocytes
.
FEBS Lett
1994
;
349
:
45
9
.
47.
Lesina
M
,
Kurkowski
MU
,
Ludes
K
,
Rose-John
S
,
Treiber
M
,
Kloppel
G
, et al
Stat3/Socs3 activation by IL-6 transsignaling promotes progression of pancreatic intraepithelial neoplasia and development of pancreatic cancer
.
Cancer Cell
2011
;
19
:
456
69
.
48.
Langhans
B
,
Kramer
B
,
Louis
M
,
Nischalke
HD
,
Huneburg
R
,
Staratschek-Jox
A
, et al
Intrahepatic IL-8 producing Foxp3(+)CD4(+) regulatory T cells and fibrogenesis in chronic hepatitis C
.
J Hepatol
2013
;
59
:
229
35
.
49.
Zhang
X
,
Feng
M
,
Liu
X
,
Bai
L
,
Kong
M
,
Chen
Y
, et al
Persistence of cirrhosis is maintained by intrahepatic regulatory T cells that inhibit fibrosis resolution by regulating the balance of tissue inhibitors of metalloproteinases and matrix metalloproteinases
.
Translat Res
2016
;
169
:
67
79
.
50.
Neesse
A
,
Algul
H
,
Tuveson
DA
,
Gress
TM
. 
Stromal biology and therapy in pancreatic cancer: a changing paradigm
.
Gut
2015
;
64
:
1476
84
.