Purpose:

The aim of the study is blocking the recruitment of a protective stroma by altering the crosstalk between normal stromal cells and tumor cells for stripping tumors of the protection conferred by the microenvironment.

Experimental Design:

A transcriptomic analysis of cocultured normal colonic fibroblasts and colorectal tumor cells was performed. We focused on the study of molecules that mediate the communication between both compartments and that entail fibroblasts’ activation and the alteration of the sensitivity to chemotherapy. We identified targets for the blocking of the tumor–stroma interaction. Finally, we tested, in vivo, the blockade of the tumor–stroma interaction in orthotopic models derived from patients and in models of acquired resistance to oxaliplatin.

Results:

IL1β/TGFβ1 are the triggers for fibroblasts’ recruitment and conversion into carcinoma-associated fibroblasts (CAF) in colorectal cancer. CAFs then secrete proinflammatory factors that alter sensitivity in tumor cells, activating JAK/STAT and PI3KCA/AKT pathways. Blocking such crosstalk with a neutralizing IL1β antibody and a TGFBR1 inhibitor is relieved by the TAK1-mediated activation of the noncanonical TGFβ pathway, which induces a change in the cytokine/chemokine repertoire that maintains a sustained activation of AKT in tumor cells. TAK1 plus TGFBR1 inhibition blocks IL1β/TGFβ1-mediated fibroblast activation, decreasing the secretion of proinflammatory cytokines. In turn, tumor cells became more sensitive to chemotherapy. In vivo, the combination of a TAK1 inhibitor plus TGFBR1 inhibitor reduced the metastatic capacity of tumor cells and the recruitment of fibroblasts.

Conclusions:

Our findings provide a translational rationale for the inhibition of TAK1 and TGFBR1 to remove the chemoprotection conferred by CAFs.

Translational Relevance

For the development of a full-blown cancer, the cooption of the surrounding normal tissue is a crucial step. Although this process may go unnoticed until tumors acquire a certain entity and appearance of the first symptoms in the patient, it is very relevant in local or distant recurrences after surgery, where there is already a medical follow-up of the disease. Thus, avoiding the activation and recruitment of stroma in new relapses is a strategy for treating cancer when still lacking the protection conferred by the microenvironment. Therefore, hampering the crosstalk between normal resident fibroblasts and malignant cells will interfere with the development of a tumor with a relevant clinical entity and consequently generating more accessible tumors for the chemotherapy. Our findings put in value the concomitant blocking of TAK1 and TGFBR1 to diminish stroma recruitment and activation, providing rationale for a novel therapeutic approach to target metastatic desmoplastic colorectal cancers.

It is well known that the communication between tumor cells and CAFs has far-reaching consequences for the progression and spread of cancer. However, during early stages of tumorigenesis, the crosstalk between the incipient neoplasia and the surrounding normal environment is controlled by the tissue-specific homeostatic balance (1). A finely tuned repertoire of cytokines and their receptors maintains this balance, which is particularly important in the colonic and rectal epithelia, where newly transformed tumor cells are in close contact with pericryptal colorectal fibroblasts. Once the balance is lost due to selective pressures, pericryptal or normal colorectal fibroblasts (NCF) are modified to become CAFs (2) in a process in which the microenvironment evolves to support tumorigenesis (3). Once altered, CAFs can help hiding tumor cells from the immune system or protect them from chemotherapeutic drugs. In these processes, a dialogue between cytokines/growth factors and their corresponding receptors is established, in which TGFβ plays a fundamental and paradoxical role, because it is one of the most important factors for boosting the prometastatic capacity of tumors (4). Other molecular dialogues are established between tumor and stroma sometimes in a context-dependent manner (5–7).

Targeting of such bidirectional crosstalk could form the basis of a strategy for fighting cancer. In fact, a lot of information and experience has been obtained from inflammatory diseases like arthritis, Crohn disease and asthma, where hindering the binding of ligand-receptor is a well-established approach (8). Pharmacologic targeting of TGFβ1 enhances the intratumoral spread of chemotherapy and reverts the stroma's activated status (9). Similarly, inhibition of Sonic Hedgehog signaling facilitates drug delivery in PDAC (10). Another archetypal example is stromal CXCL12 and its receptor CXCR4 expressed in tumor cells, where inhibitor AMD3465 blocks the action of the chemokine and consequently decreases AKT activation (11). Dozens of similar examples have identified, coinciding with the explosion of development of targeted therapies.

However, the main problem is that tumors still adapt quickly and develop resistant phenotypes because redundant signaling mechanisms overcome such blocking strategies. While most tumor types are kinase-addicted, the blocking of such ligand-dependent kinases quite often results in the activation of the pathway, although not by the reactivation of the kinase (12). This arises from the redundancy in signaling cascades in tumor cells (13), because different kinases operate in the same pathway or have a similar function, and different ligands act on the same receptor. Furthermore, soluble stromal or tumoral receptors can rescue tumor cells from therapies that hinder the crosstalk between stromal and malignant cells (14). For this reason, the design of therapeutic strategies must take into consideration relevant “hubs” of intracellular signaling. This is a difficult task, because these strategies are often accompanied by very severe side effects.

The TGFβ-Activated Kinase 1 (TAK1) protein, encoded by MAP3K7 gene, is a serine/threonine kinase that mediates signaling by IL1β (15), TNFα (16), LPS (17), TLR (18), although it was discovered because of its role in the noncanonical TGFβ pathway (19). In fact, TAK1 is a proinflammatory effector that contributes to the IKK complex activation and consequently activates NFκβ pathway (20). However, it can also activate other downstream MAPKs like JNK (21), P38, and ERK (22). TAK1 is also involved in tumorigenesis, and its inhibition induces apoptosis (23) and chemosensitivity (24). All these observations suggest that TAK1 is a point of convergence for signaling pathways that are activated by a broad spectrum of stimuli. In our particular context, TAK1 plays a central role in the proinflammatory response of the microenvironment.

Here we report that cotargeting of TAK1 and TGFBR1 in fibroblasts and CAFs is an interesting approach for avoiding the secretion of protective soluble factors from activated fibroblasts through crosstalk with tumor cells in colorectal cancer. In turn, malignant cells are rendered more sensitive to chemotherapy due to the absence of the protective environment normally provided by fibroblasts. Interestingly, in vivo experiments of orthotopic patient-derived xenografts and splenic cell line injection show that the combined inhibition had a deep impact in the dissemination and settlement of distant metastases, inhibiting the recruitment of a supportive stroma.

Cell culture and cell lines

Primary NCFs, normal hepatic fibroblasts (NHF), and CAFs used in the study were isolated in our laboratory from surgical specimens after explicit patient's written consent had been obtained. NCFs and NHFs were isolated from at least 5 cm from the surgical margin of excised primary or metastatic tumors. Colorectal cell lines used are DLD1 (MSI-CMS1), HCT116 (MSI-CMS4), and HT29 (MSS-CMS3). Oxaliplatin-resistant derivatives (HTOXAR3 and DLDOXAR) were kindly provided by Dr. Martínez-Balibrea (Institut Germans Tries i Pujol, Badalona, Spain). All cells were cultured in DMEMF12 medium supplemented with 10% FBS, glutamine, and penicillin/streptomycin (Gibco) at 37°C in 5% CO2. Cells were periodically tested for Mycoplasma contamination and were authenticated by short tandem repeat profiling.

Eight different isolated NCFs (clinical details in Supplementary Table online) were cocultured with DLD1 cells (coming from a stage III colorectal adenocarcinoma) in 3-μm pore size Transwell inserts for 96 hours in 2% FBS DMEMF12. Monocultures of same NCFs and DLD1 cells in the same culture conditions were used as corresponding controls.

Reagents and inhibitors

IL1β and TGFβ1 were purchased from PeproTech. Neutralizing IL1β antibody was purchased from R&D Systems (reference AB-201-NA), diluted in water; TGFBR1 inhibitor III-CAS 356559-13-2 was purchased from Calbiochem, (5Z)-7-oxozeaenol (OXO) was purchased from Tocris, both diluted in DMSO. Oxaliplatin (L-OHP) was diluted in water and 5-fluorouracyl in saline buffer. For the in vivo studies, TGFBR1 inhibitor used was galunisertib, kindly provided by Eli Lilly & Co and was freshly suspended in 1% carboxymethylcellulose sodium salt, 0.5% SDS, 0.085% povidone, and 0.05% antifoam Y-30 emulsion.

Microarray analyses

RNA from eight paired primary NCFs, tumor cell–cocultured NCFs, paired CAFs, DLD1 monocultures, and cocultures of DLD1 cells with the corresponding eight NCFs, was extracted as described elsewhere (25).

Total mRNA was hybridized in an Affymetrix GeneChip Human Gene 1.0 ST Array. All computations and statistical analyses were performed using the R language. Microarray data were processed using the justRMA function in the simpleaffy package (26). The resulting data were used to look for genes that were differentially expressed between groups (monocultures vs. cocultures) using the Significance Analysis of Microarrays (SAM) test, available in the samr package (27). To obtain a reduced list of genes, we considered those with a false discovery rate (FDR) < 0.05 and a >2-fold change.

Gene-set enrichment analysis

Gene-set enrichment analysis (GSEA; ref. 28) was carried out on a preranked list of genes determined by the SAM d-statistic to be differentially expressed in cocultured and monocultured NCFs and tumor cells (blue and red phenotypes, respectively, in GSEA plots). We used the H.HALLMARKS; C2.CP.ALL and C5.BP gene sets.

Cell proliferation and viability

The WST-1 assay was performed following the manufacturer's instructions. Previously, six replicates of 2,000 cells/well (tumor cell lines) had been plated in 96-well plates and allowed to attach, then grown in DMEM/F12 at 37°C overnight. Next day, CM from treated cocultures were added accordingly. Cells were incubated for 5 days, treated with a range of doses against oxaliplatin (L-OHP), and the drug effect on tumor cells was calculated by normalizing the number of cells after 5 days of continuous treatment relative to the maximum number of cells in each CM. WST-1 assays were also tested directly on cells cocultured in 24-well Transwell inserts, seeding tumor cells in the bottom well and fibroblasts (either NCFs, NHFs, or CAFs) in the insert. Treatments were added when cells were attached to the culture surfaces and viability was assayed for 5 days.

Flow cytometry

Acquisition of CAF markers was assessed in 5-day cocultures by flow cytometry using anti-human CD90-APC (Miltenyi Biotec) to discriminate between fibroblasts (positive cells) from tumor cells (negative), and mouse anti-human CD49b-FITC (integrin A2) and mouse anti-human CD127-BV421 (IL7R). NCF and CAF monocultures were used as controls.

Animal models

The patient gave written consent to donate a piece of tumor to generate the orthoxenograft model. The Institutional Ethics Committees approved the study protocol. Experimental design was approved by the IDIBELL animal facility committee (AAALAC–Unit 1155). All experiments were performed in accordance with the guidelines for Ethical Conduct in the Care and Use of Animals as stated in the International Guiding Principles for Biomedical Research Involving Animals, developed by the Council for International Organizations of Medical Sciences.

A patient-derived orthotopic colorectal cancer xenograft model (PDOX), generated from a locoregional relapse 5 years after completing adjuvant treatment, was orthotopically implanted into the cecum of Crl:NU-Foxn1nu mice. PDOXs were inspected every other day and monitored for the presence of breathing and behavioral problems. Tumor volume was monitored every 2 days by palpation. After reaching approximately 200 mm3, animals were randomized into control arm (n = 9, FUOX treatment, 5-fluorouracyl plus oxaliplatin) and test arm (n = 9, FUOX plus galunisertib plus OXO) and treated for 30 days. Metastatic dissemination was determined in macroscopic and microscopic examination (hematoxylin–eosin and Masson trichrome) in peritoneum, liver, lung, adrenal glands, kidneys, diaphragm, and brain. Survival was recorded following strict animal welfare criteria, supervised by a veterinarian, and normally did not coincide with the death of the animal but with criteria of euthanasia as a result of veterinary supervision.

The capacity for blocking the supportive stroma to promote distant metastases was assessed by injecting 8 × 105 cells (HT29 and its L-OHP–resistant derivative HTOXAR3, and DLD1 and its L-OHP–resistant derivative DLDOXAR) into the spleen of Crl:NU-Foxn1nu mice. Two days after injection, the spleen was removed and a period of one week was allowed to pass for micrometastases to form. Next, animals were randomized into L-OHP (control) or L-OHP plus galunisertib plus OXO groups. The treatment lasted for 15 days, and after 30 days the animals were sacrificed to determine the presence of visceral and peritoneal metastases as described above.

Transcriptomic differences between monocultures and cocultures with prognostic value

To elucidate the contribution of tumor–stroma interactions for tumor growth and chemoresistance, we used a normal colonic fibroblast (NCF) coculture model from various patients affected by colorectal cancer and tumor cells (CRC DLD1 cell line). After 5 days of indirect contact through the mesh of a 3-μm Transwell insert, mRNA was obtained from each cell type. mRNA from monocultures of both types was also obtained for use as a reference for transcriptomic profiling. We hybridized mRNA in Affymetrix GeneChip Human Gene 1.0 ST expression arrays. Using a >2 (or −2) fold change (FC) and a false discovery rate (FDR) of q < 0.01, we obtained 886 differentially expressed genes (DEG) in the cocultivated NCFs with respect to monocultures (Supplementary Fig. S1; Supplementary Table S1). When we performed the same analysis on cocultured tumor cells under the same conditions of stringency, we obtained 1,066 DEGs in cocultured tumor cells compared with monocultured controls (Supplementary Table S2). In both cases, the large number of DEGs with low FDR values gives an idea of the profound transcriptomic changes induced by the crosstalk between these cell types.

To evaluate the biological importance of the coordinated variation in the change of expression observed in the two cocultured cell types, we performed GSEA on the complete list of ranked genes. The most notable pathways and biological processes enriched in cocultured fibroblasts are depicted in Fig. 1A and Supplementary Table S3 (Tumor cell GSEA in Supplementary Fig. S2). In addition, GSEA results revealed that the cocultured stroma and tumor cells acquired a transcriptomic profile enriched in pathways and biological processes, with considerable overlap with those of rectal cancer patients classified as nonresponders to neoadjuvant treatment according to our group's previous results obtained from microdissected endoscopic pretreatment biopsies (29).

Figure 1.

A, Gene set enrichment analysis representative pathways and processes between cocultured (red phenotype) and monocultured (blue phenotype) NCFs. We explored datasets H (Hallmark gene sets), C2 (curated gene sets), and C5 (BP GO biological processes). B, Heatmap of genes with a > 2-fold or <−2-fold change between NCF and cocultured NCF simultaneously with a > 2-fold or <−2-fold change between NCF and paired CAFs (from the same patient). As illustrated in the dendrogram, cocultured NCFs clustered with CAFs (with the exception of two CAFs that clustered with NCFs). The clinical data of the patients from which the fibroblasts were obtained are in Supplementary Table S5. C, Flow cytometry check of some genes coding for cell-surface markers (IL7R-CD127 and ITGA2-CD49) from the list of genes common to cocultured NCFs and CAFs. Cocultured NCFs acquired similar values to monocultured CAFs of CD127 and CD49 after 5 days in culture with tumor cells (black dots). To distinguish between tumor cells and fibroblasts we used a CD90-APC antibody, which expressed only nonfibroblast cells (red dots). By means of flow cytometry, we showed that CAFs and NCFs present a similar degree of expression of CD90, although values were somewhat lower in NCFs. After 5 days of coculture with tumor cells (black dots). After 5 days in coculture, cocultured NCFs expressed both markers in 80%. These values are similar to those of the CAFs, implying a simultaneous coexpression in approximately 70% of the cocultured NCFs. D, We wanted to determine whether the most differentially expressed genes in the cocultured tumor cells (> 4-fold change, FDR q < 0.01) were associated with worse prognosis in patients with colorectal carcinoma, taking disease-free survival time as a prognostic variable. To do this we used datasets GSE33113, which consists of 90 untreated stage II patients, and GSE14333, comprising 139 untreated stage I, II, and III patients. We used normalized ssGSEA (single-sample GSEA; ref. 47) to assign patients to the high- or low-risk of recurrence groups. The Cox regression model shows that patients with a score greater than zero had a higher risk of recurrence: HR = 2.78, 95% CI = 1.0–7.7, P = 0.05 for GSE33113, upper panel; and HR = 3.05, 95% CI = 1.13–8.7, P = 0.028 for GSE14333 (corrected for stage; high-risk group n = 68, 17 stage I, 33 stage II, 18 stage III; bottom); therefore, they had a worse prognosis, from which it could be concluded that the phenotype acquired by the tumor cells after having been cocultured with normal colonic fibroblasts for 5 days conferred aggressiveness on cancer cells. The clinical description of the series GSE33113 and GSE14333 used is in Supplementary Table S6. CI, confidence interval.

Figure 1.

A, Gene set enrichment analysis representative pathways and processes between cocultured (red phenotype) and monocultured (blue phenotype) NCFs. We explored datasets H (Hallmark gene sets), C2 (curated gene sets), and C5 (BP GO biological processes). B, Heatmap of genes with a > 2-fold or <−2-fold change between NCF and cocultured NCF simultaneously with a > 2-fold or <−2-fold change between NCF and paired CAFs (from the same patient). As illustrated in the dendrogram, cocultured NCFs clustered with CAFs (with the exception of two CAFs that clustered with NCFs). The clinical data of the patients from which the fibroblasts were obtained are in Supplementary Table S5. C, Flow cytometry check of some genes coding for cell-surface markers (IL7R-CD127 and ITGA2-CD49) from the list of genes common to cocultured NCFs and CAFs. Cocultured NCFs acquired similar values to monocultured CAFs of CD127 and CD49 after 5 days in culture with tumor cells (black dots). To distinguish between tumor cells and fibroblasts we used a CD90-APC antibody, which expressed only nonfibroblast cells (red dots). By means of flow cytometry, we showed that CAFs and NCFs present a similar degree of expression of CD90, although values were somewhat lower in NCFs. After 5 days of coculture with tumor cells (black dots). After 5 days in coculture, cocultured NCFs expressed both markers in 80%. These values are similar to those of the CAFs, implying a simultaneous coexpression in approximately 70% of the cocultured NCFs. D, We wanted to determine whether the most differentially expressed genes in the cocultured tumor cells (> 4-fold change, FDR q < 0.01) were associated with worse prognosis in patients with colorectal carcinoma, taking disease-free survival time as a prognostic variable. To do this we used datasets GSE33113, which consists of 90 untreated stage II patients, and GSE14333, comprising 139 untreated stage I, II, and III patients. We used normalized ssGSEA (single-sample GSEA; ref. 47) to assign patients to the high- or low-risk of recurrence groups. The Cox regression model shows that patients with a score greater than zero had a higher risk of recurrence: HR = 2.78, 95% CI = 1.0–7.7, P = 0.05 for GSE33113, upper panel; and HR = 3.05, 95% CI = 1.13–8.7, P = 0.028 for GSE14333 (corrected for stage; high-risk group n = 68, 17 stage I, 33 stage II, 18 stage III; bottom); therefore, they had a worse prognosis, from which it could be concluded that the phenotype acquired by the tumor cells after having been cocultured with normal colonic fibroblasts for 5 days conferred aggressiveness on cancer cells. The clinical description of the series GSE33113 and GSE14333 used is in Supplementary Table S6. CI, confidence interval.

Close modal

Cocultured NCFs acquired a CAF phenotype

We set out to determine whether the transcriptional changes suffered by the NCF fibroblasts when in paracrine communication with tumor cells were associated with the acquisition of a transcriptional pattern similar to that of their corresponding CAFs obtained from the same patient. In this way, this interaction could be used as a model for studying the participation and recruitment of fibroblasts during the early stages of tumor expansion and growth. To check this, we selected genes with a > 2-fold (or <−2-fold) change between NCFs and cocultured NCFs, with a simultaneous >2-fold (or <−2-fold) between NCF and paired CAFs. As observed in Fig. 1B, NCFs cocultured with tumor cells acquired a transcriptional profile largely similar to that of their corresponding CAFs. Two distinct clusters, CAFs and co-NCF in one, and monocultured NCF in the other, could be clearly identified. We also performed a principal component analysis (PCA) analysis in which genes of the first component generated two clusters, NCF in one, cocultured NCF and CAFs in the other, with a single misclassified sample (Supplementary Fig. S3).

We experimentally corroborated the overexpression of some of these markers in NCFs cocultured with tumor cells, in particular cell surface molecules such as CD49B (ITGA2) and CD127 (IL7R; Fig. 1C).

These results suggest that cocultured NCFs acquired a CAF-like phenotype.

Prognostic value of changes acquired by tumor cells

Using single-sample GSEA (ssGSEA), we determined that DEGs in cocultured tumor cells conferred a bad prognosis on two different datasets of not treated patients with colorectal cancer [GSE33113, 90 patients stage II, HR = 2.78 (95% CI 1–7.76), Cox P = 0.05; GSE14333, 139 patients stages I–III, HR = 3.05 (95% CI 1.13–8.7), Cox P = 0.028, corrected for stage; Fig. 1D).

In summary, the coculture of tumor cells and NCFs produces radical transcriptional changes in both cell types. Consequently, the acquisition of these changes has an important repercussion on the prognosis of patients affected by colorectal cancer.

IL1β and TGFβ1 are the triggers for the conversion of NCFs into CAFs

Our aim was to define the main mediating factors of the paracrine communication between NCF and epithelial tumor cells. For this purpose, an analysis of protein–protein interaction (PPI) was carried out, selecting from the transcriptomic data of cocultured cells (> 2-fold change and FDR q < 0.1 relative to corresponding monocultured cells) those mRNAs that encode proteins secreted in fibroblasts with a transcript that is in turn overexpressed and encodes a membrane receptor in epithelial cells, and vice versa. Detailed information is presented in the Supplementary Data and Supplementary Results (Supplementary Figs. S4–S6; Supplementary Table S4).

After integrating our transcriptomic/PPI data, we established our working hypothesis that cytokines released by NCFs stimulate tumor cells to secrete IL1β and TGFβ1. These soluble factors are responsible for the activation of NCF to convert them into CAFs. In turn, activated NCFs produce large amounts of IL1β, which potentiates the pathway, in an autocrine manner, as well as a large number of IL1β/TGFβ1 target proteins, mainly chemokines, cytokines, EGFR ligands and PAI-1 (SERPINE1), soluble factors with high affinity for receptors IL6R, IL11RA, (via JAK-STAT), and CCR6, CXCR3, EGFR, CCR2, FGFR, and cMET (via PI3K-AKT). These are all pathways involved in chemoresistance in colorectal cancer.

Blocking IL1β and TGFBR1 in cocultured fibroblasts creates a compensatory feedback loop mediated by the noncanonical TGFβ pathway, inducing sustained AKT activation in cocultured tumor cells

Transwell coculture experiments of NCFs with colorectal cancer cells were performed to evaluate the effects of soluble factors produced by the heterotypic crosstalk (Fig. 2A, left). Cocultures were intervened with agents blocking the action of soluble triggers according to our hypothesis (neutralizing IL1β antibody and a TGFBR1 inhibitor, or in combination). CM of these cocultures were used to test proliferation (Fig. 2A, second panel), migration (Fig. 2A, third panel), and sensitivity to drugs (Fig. 2A, fourth panel) in cells not previously exposed under these conditions. In the cells treated with CM in the presence of the two blocking agents, we observed an increase in proliferation, a decrease in migration and greater sensitivity to oxaliplatin (L-OHP), although the latter finding seems to be due mainly to the action of the neutralizing IL1β antibody.

Figure 2.

A, We obtained conditioned medium (CM) from Transwell cocultured NCFs and tumor cells (DLD1) in the absence of FBS under the following conditions: control conditioned medium (in black), with addition of neutralizing IL1β antibody (1 μg/mL; in blue), a TGFBR1 inhibitor (0.1 μmol/L; in red), or the combination of the neutralizing antibody and inhibitor (in yellow). After 24 hours of culture, conditioned medium was collected and FBS was reconstituted (10%) for subsequent analyses. As illustrated in the first left bar plot, proliferation was enhanced in the yellow condition, while migration was significantly decreased in the presence of the TGFBR1 inhibitor alone (red condition) or in combination with the neutralizing antibody (yellow condition; Kruskal–Wallis test with correction for multiple comparison). However, the conditioned medium significantly shifted the dose–response curve to oxaliplatin (L-OHP), decreasing the IC50 values when the neutralizing IL1β antibody was added to the conditioned medium (mean values of four independent experiments). B, We directly explored the blockade of the NCF–tumor cell crosstalk in cells in coculture, adding the neutralizing IL1β antibody (1 μg/mL) and the TGFBR1 inhibitor (0.1 μmol/L) in the bottom and top compartments of the Boyden chamber. There was no benefit from the treatments, either with respect to the absence of L-OHP or with four different concentrations of the drug. However, when we used CAFs instead of NCFs (right plot), there was a significant decrease in proliferation with the combination of the two blocking agents (solid yellow bar), but no differences when we added L-OHP (means of four independent experiments). C, Western blots of cocultured DLD1 and CAFs at 8 and 24 hours in Transwell mesh (3 μm) contact. There was a decrease in STAT3 signaling in tumor cells, but sustained AKT activation when blocking the crosstalk with the two agents. Interestingly, CAFs seems to switch signaling from P38 and SMAD to JNK and NFκβ when blocking IL1β and TGFBR1.

Figure 2.

A, We obtained conditioned medium (CM) from Transwell cocultured NCFs and tumor cells (DLD1) in the absence of FBS under the following conditions: control conditioned medium (in black), with addition of neutralizing IL1β antibody (1 μg/mL; in blue), a TGFBR1 inhibitor (0.1 μmol/L; in red), or the combination of the neutralizing antibody and inhibitor (in yellow). After 24 hours of culture, conditioned medium was collected and FBS was reconstituted (10%) for subsequent analyses. As illustrated in the first left bar plot, proliferation was enhanced in the yellow condition, while migration was significantly decreased in the presence of the TGFBR1 inhibitor alone (red condition) or in combination with the neutralizing antibody (yellow condition; Kruskal–Wallis test with correction for multiple comparison). However, the conditioned medium significantly shifted the dose–response curve to oxaliplatin (L-OHP), decreasing the IC50 values when the neutralizing IL1β antibody was added to the conditioned medium (mean values of four independent experiments). B, We directly explored the blockade of the NCF–tumor cell crosstalk in cells in coculture, adding the neutralizing IL1β antibody (1 μg/mL) and the TGFBR1 inhibitor (0.1 μmol/L) in the bottom and top compartments of the Boyden chamber. There was no benefit from the treatments, either with respect to the absence of L-OHP or with four different concentrations of the drug. However, when we used CAFs instead of NCFs (right plot), there was a significant decrease in proliferation with the combination of the two blocking agents (solid yellow bar), but no differences when we added L-OHP (means of four independent experiments). C, Western blots of cocultured DLD1 and CAFs at 8 and 24 hours in Transwell mesh (3 μm) contact. There was a decrease in STAT3 signaling in tumor cells, but sustained AKT activation when blocking the crosstalk with the two agents. Interestingly, CAFs seems to switch signaling from P38 and SMAD to JNK and NFκβ when blocking IL1β and TGFBR1.

Close modal

DLD1 proliferation and survival experiments were also performed directly on the Transwell inserts, coculturing tumor cells with NCFs or CAFs. However, as shown in Fig. 2B, we did not find any benefit in terms of proliferation from any of the treatments (Fig. 2B, left panel solid bars), or from the addition of oxaliplatin at different doses (5, 8, 10, and 40 μmol/L; Fig. 2B, left, dashed bars). We only observed a trend toward increased survival when using the neutralizing IL1β antibody in cocultures with NCFs. Likewise, the cocultures with CAFs did not indicate any benefit of the various treatments in terms of oxaliplatin sensitivity (Fig. 2B, right).

Then, we wanted to check the signaling pathways altered by the blockade of IL1β and TGFBR1. As illustrated in Fig. 2C, tumor cells displayed a progressive decrease in levels of STAT3 activation. However, the levels of AKT phosphorylation changed in the opposite direction, increasing as we added the blocking agents to alter the crosstalk between cell types. We observed a similar compensatory feedback loop in fibroblasts, switching from the activation of P38 and SMAD2/3 pathways in control cocultures to the activation of NFκβ pathway as we added IL1β and TGFBR1 blocking agents (Fig. 2C, right). Next, we attempted to characterize the soluble factors present in the CM of 5-day cocultures of cells under the various treatment conditions to understand the results obtained from the Western blot analyses. To this end, we used a cytokine array, interrogating 174 soluble factors. Eighty of the 174 soluble factors appeared to be modulated by the combination of treatments, 31 decreasing in concentration with the addition of the blocking agents, particularly when the combination of the neutralizing IL1β antibody plus the TGFBR1 inhibitor was used (Fig. 3). Conversely, the concentration of 49 cytokines increased with the change in crosstalk. Such cytokine and growth factor switches might be responsible for the differential signaling observed in fibroblasts and the sustained AKT activation in tumor cells.

Figure 3.

Cytokine array (Raybiotech) from cocultures in conditioned media that interrogates 174 soluble factors and cytokines. Dot blot for the 80 cytokines out of 174 than have a decreasing or increasing concentration in a monotonic trend (increasing or decreasing sequentially from control group to neutralizing antibody + TGFBR1 inhibitor group) across treatments in relation to cocultures control. Thirty-one cytokines have decreasing concentrations than cocultures control in intervened cocultures, particularly when using the two blocking agents. Values are the mean of two cytokine arrays (n = 2) consisting each one in a pool of three independent experiments. Values for each cytokine were then normalized according to control group (no treatment) and transformed in log scale. On the other hand, 49 cytokines showed a trend toward increasing sequentially from control cocultures to neutralizing antibody + TGFBR1 inhibitor. In this group, protumoral cytokines like CCL5, IL11, IL1β, HGF, CXCL12, or IL6 (in bold). Values depicted are log fold change relative to those of control cocultures. Dashed line depicted values in coculture control. Values above 0 means overexpression and below 0 infraexpression. Inside the red box, cytokines and soluble factors that are downregulated with the different treatments in relation to control (no treatment) are shown.

Figure 3.

Cytokine array (Raybiotech) from cocultures in conditioned media that interrogates 174 soluble factors and cytokines. Dot blot for the 80 cytokines out of 174 than have a decreasing or increasing concentration in a monotonic trend (increasing or decreasing sequentially from control group to neutralizing antibody + TGFBR1 inhibitor group) across treatments in relation to cocultures control. Thirty-one cytokines have decreasing concentrations than cocultures control in intervened cocultures, particularly when using the two blocking agents. Values are the mean of two cytokine arrays (n = 2) consisting each one in a pool of three independent experiments. Values for each cytokine were then normalized according to control group (no treatment) and transformed in log scale. On the other hand, 49 cytokines showed a trend toward increasing sequentially from control cocultures to neutralizing antibody + TGFBR1 inhibitor. In this group, protumoral cytokines like CCL5, IL11, IL1β, HGF, CXCL12, or IL6 (in bold). Values depicted are log fold change relative to those of control cocultures. Dashed line depicted values in coculture control. Values above 0 means overexpression and below 0 infraexpression. Inside the red box, cytokines and soluble factors that are downregulated with the different treatments in relation to control (no treatment) are shown.

Close modal

These results suggest that as we block the crosstalk between both cell types, compensatory mechanisms are produced where NFκβ pathway activation arises in fibroblasts through the noncanonical TGFβ pathway and TAK1, producing a cytokine turnover, which would exert redundant functions to those observed in the control cocultures.

Simultaneous TAK1 and TGFBR1 inhibition in fibroblasts overcomes stroma-mediated drug resistance

The results obtained to this point suggested that an alternative mechanism was activating NFκβ. The TGFBR1 inhibitor was inhibiting the canonical TGFβ1 pathway and the neutralizing IL1β antibody that controls the IL1β-mediated inflammatory processes. In any case, NFκβ pathway was activated when the two main ligands responsible for fibroblast activation in our model were blocked. Thus, we reasoned that the inhibition of a central signaling node converging from the IL1β pathway and the noncanonical TGFβ1 pathway, the TAK1 kinase (MAP3K7) might be an interesting approach. Therefore, the simultaneous blockade of TAK1 and TGFBR1 would allow us to inhibit signaling mediated by IL1β and TGFβ1 (via a canonical pathway involving a TGFBR1 inhibitor, and a noncanonical pathway involving a TAK1 inhibitor) and counteract the activation of NFĸβ, maintaining the inhibition of the MAPK- and SMAD-dependent pathways.

We first examined the effect of TAK1 inhibitor OXO on the proliferation of cells in monoculture (Fig. 4A). OXO significantly reduced the proliferation of CAFs, while no statistically significant effect was observed in tumor cells. We assayed the effect of coculture CM, either control, OXO-treated or (OXO + TGFBR1 inhibitor)-treated, on the sensitivity of tumor cells to L-OHP. The CM from OXO + TGFBR1 inhibitor–treated cocultures clearly conferred less protection from L-OHP than cocultures with control-CM (Fig. 4B). In addition, the combined treatment of OXO + TGFBR1 inhibitor sensitized tumor cells to L-OHP in all cell lines tested in cocultures with either NCFs, NHFs, or CAFs (Fig. 4C; dashed colored bars). The combination also diminished the degree of proliferation of tumor cells in the absence of L-OHP (solid bars in Fig. 4C). Moreover, OXO inhibited the activation of P38 and P65 in CAFs after short (Fig. 4D left) or sustained (Fig. 4D middle) IL1β stimulation. The combination of OXO + TGFBR1 inhibitor was the most effective at inhibiting P38 and P65 phosphorylation in (IL1β + TGFβ1)-treated NCFs (Fig. 4D, right), and rendered those treated NCFs without acquired CAF markers, like FAP and IL7R (Fig. 4E). Nevertheless, when we assayed signaling directly in cells in coculture, we observed sustained AKT activation in tumor cells (Fig. 4F, middle), although we did succeed in inhibiting the activation of P65 and P38 in cocultured fibroblasts (Fig. 4E, right). Likewise, the concentration of some of the cytokines that we had previously found to be increased after 5 days in culture, such as IL11 and IL6, were significantly reduced with the new treatment strategy (Fig. 4G). Even so, the sustained activation of AKT seemed to indicate a new cytokine turnover. A second cytokine array revealed that some cytokines and growth factors were secreted into the culture medium with an opposite tendency to that previously noted with the treatments with neutralizing IL1β antibody and the TGFBR1 inhibitor (Fig. 5).

Figure 4.

A, Effect of the TAK1 inhibitor (5z)-7 oxozeaenol (OXO) on CAFs (left plot) and DLD1 cells (right plot). A significant decrease in the number of CAFs was noted at 500 nmol/L OXO (means of three independent experiments). The DLD1 cell line is Kras-independent, so TAK1 inhibition does not induce apoptosis in these cells (23). B, We obtained CM from Transwell cocultures of CAFs and tumor cells (DLD1) in the absence of FBS under the following conditions: control conditioned medium (in black), with the addition of OXO (400 nmol/L; in green) and the combination of (400 nmol/L) OXO, 0.1 μmol/L TGFBR1 inhibitor (in purple). After 24 hours of culture, CM was collected and FBS was reconstituted (10%) for subsequent analyses. When we tested the reduced protective capacities of the intervened cocultures’ CM, we observed a substantial decrease in the IC50 values against L-OHP when using the combination of both agents OXO and TGFBR1 inhibitor. C, Proliferation and viability assay in Transwell cocultures using different colorectal cell lines and fibroblasts (NCF, NHF, and CAFs). There was a significant decrease in proliferation and viability against L-OHP [(10 μmol/L) for DLD1 cells and (2 μmol/L) for HT29 and HCT116 cells] for all combinations when combining the TAK1 inhibitor OXO and the TGFBR1 inhibitor (purple bars). Dunn multiple comparison test showed the group that displayed differences in relation to control. Despite the remarkably lower values compared with the controls, the differences between cocultures receiving only the TAK1 inhibitor (green) were not statistically significant (means of four independent experiments). The combination of OXO plus TGFBR1 inhibitor was the most effective decreasing the viability of different tumor cell lines, either using NCFs or NHFs, as surrogates of starting tumorigenesis in a primary site or in a liver metastasis, as well as using CAFs isolated from primary tumors. D, Western blot assays to check the short-term (left) and sustained (middle) effectiveness of the TAK1 inhibitor OXO at blocking TAK1 signaling and the key downstream proteins P38 and P65 in IL1β-stimulated CAFs. In addition, the combination of OXO plus TGFBR1 inhibitor was the most effective at limiting the activation of TAK1, P38, and P65 in IL1β-stimulated (10 ng/mL) and TGFβ1-stimulated (40 ng/mL) CAFs (right). E, The combination of OXO plus TGFBR1 inhibitor inhibited the acquisition of CAF markers (FAP) in NCFs treated with IL1β and TGFβ1, while the myofibroblastic αSMA marker was not affected. F, Western blots of cocultured DLD1 and CAFs at 8 and 24 hours in Transwell cocultures. We observed a decrease in STAT3 signaling in tumor cells but, again, sustained AKT activation when blocking the crosstalk with the two agents in tumor cells. However, P65 was inactivated in cocultured CAFs with the combination of the two inhibitors, unlike in previous treatments with the neutralizing antibody and the TGFBR1 inhibitor (right). G, ELISA levels of proinflammatory cytokine IL11 were determined in conditioned media from cocultured cells with different treatments assayed after 48 hours of cell contact. In OXO and OXO plus TGFBR1 inhibitor, the concentration of IL11 was lower relative to coculture control conditioned media, but only in the latter treatment was the decrease statistically significantly different from the control (left). In addition, after 6 days in coculture, the levels of IL11 and IL6 were still significantly lower in OXO plus TGFBR1 inhibitor–treated cocultures (middle and right).

Figure 4.

A, Effect of the TAK1 inhibitor (5z)-7 oxozeaenol (OXO) on CAFs (left plot) and DLD1 cells (right plot). A significant decrease in the number of CAFs was noted at 500 nmol/L OXO (means of three independent experiments). The DLD1 cell line is Kras-independent, so TAK1 inhibition does not induce apoptosis in these cells (23). B, We obtained CM from Transwell cocultures of CAFs and tumor cells (DLD1) in the absence of FBS under the following conditions: control conditioned medium (in black), with the addition of OXO (400 nmol/L; in green) and the combination of (400 nmol/L) OXO, 0.1 μmol/L TGFBR1 inhibitor (in purple). After 24 hours of culture, CM was collected and FBS was reconstituted (10%) for subsequent analyses. When we tested the reduced protective capacities of the intervened cocultures’ CM, we observed a substantial decrease in the IC50 values against L-OHP when using the combination of both agents OXO and TGFBR1 inhibitor. C, Proliferation and viability assay in Transwell cocultures using different colorectal cell lines and fibroblasts (NCF, NHF, and CAFs). There was a significant decrease in proliferation and viability against L-OHP [(10 μmol/L) for DLD1 cells and (2 μmol/L) for HT29 and HCT116 cells] for all combinations when combining the TAK1 inhibitor OXO and the TGFBR1 inhibitor (purple bars). Dunn multiple comparison test showed the group that displayed differences in relation to control. Despite the remarkably lower values compared with the controls, the differences between cocultures receiving only the TAK1 inhibitor (green) were not statistically significant (means of four independent experiments). The combination of OXO plus TGFBR1 inhibitor was the most effective decreasing the viability of different tumor cell lines, either using NCFs or NHFs, as surrogates of starting tumorigenesis in a primary site or in a liver metastasis, as well as using CAFs isolated from primary tumors. D, Western blot assays to check the short-term (left) and sustained (middle) effectiveness of the TAK1 inhibitor OXO at blocking TAK1 signaling and the key downstream proteins P38 and P65 in IL1β-stimulated CAFs. In addition, the combination of OXO plus TGFBR1 inhibitor was the most effective at limiting the activation of TAK1, P38, and P65 in IL1β-stimulated (10 ng/mL) and TGFβ1-stimulated (40 ng/mL) CAFs (right). E, The combination of OXO plus TGFBR1 inhibitor inhibited the acquisition of CAF markers (FAP) in NCFs treated with IL1β and TGFβ1, while the myofibroblastic αSMA marker was not affected. F, Western blots of cocultured DLD1 and CAFs at 8 and 24 hours in Transwell cocultures. We observed a decrease in STAT3 signaling in tumor cells but, again, sustained AKT activation when blocking the crosstalk with the two agents in tumor cells. However, P65 was inactivated in cocultured CAFs with the combination of the two inhibitors, unlike in previous treatments with the neutralizing antibody and the TGFBR1 inhibitor (right). G, ELISA levels of proinflammatory cytokine IL11 were determined in conditioned media from cocultured cells with different treatments assayed after 48 hours of cell contact. In OXO and OXO plus TGFBR1 inhibitor, the concentration of IL11 was lower relative to coculture control conditioned media, but only in the latter treatment was the decrease statistically significantly different from the control (left). In addition, after 6 days in coculture, the levels of IL11 and IL6 were still significantly lower in OXO plus TGFBR1 inhibitor–treated cocultures (middle and right).

Close modal
Figure 5.

Cytokine expression (Raybiotech cytokine array) in CM from cocultures treated with OXO (dark green), OXO plus TGFBR1 inhibitor (pink). Values are the mean of two cytokine arrays (n = 2) consisting each one in a pool of three independent experiments. Values for each cytokine were then normalized according to control group (no treatment) and transformed in log scale. Dashed line depicted values in coculture control. Values above 0 means overexpression and below 0 infraexpression. Interestingly, several cytokines have an opposite secretion trend compared with the use of the neutralizing antibody and the TGFBR1 inhibitor. As an example, protumoral factors like CCL5, IL6, or HGF, and antitumoral factors like GM-CSF, IL2, or IL7. Depicted in bold are those cytokines that most significantly changed the trend versus to control in relation to values obtained in Fig. 3. Interestingly, many of the cytokines in bold in the left part of the graph are depicted inside the red box in Fig. 3, showing an opposite trend of secretion with the treatments with OXO and OXO + TGFBR1 inhibitor.

Figure 5.

Cytokine expression (Raybiotech cytokine array) in CM from cocultures treated with OXO (dark green), OXO plus TGFBR1 inhibitor (pink). Values are the mean of two cytokine arrays (n = 2) consisting each one in a pool of three independent experiments. Values for each cytokine were then normalized according to control group (no treatment) and transformed in log scale. Dashed line depicted values in coculture control. Values above 0 means overexpression and below 0 infraexpression. Interestingly, several cytokines have an opposite secretion trend compared with the use of the neutralizing antibody and the TGFBR1 inhibitor. As an example, protumoral factors like CCL5, IL6, or HGF, and antitumoral factors like GM-CSF, IL2, or IL7. Depicted in bold are those cytokines that most significantly changed the trend versus to control in relation to values obtained in Fig. 3. Interestingly, many of the cytokines in bold in the left part of the graph are depicted inside the red box in Fig. 3, showing an opposite trend of secretion with the treatments with OXO and OXO + TGFBR1 inhibitor.

Close modal

Thus, the combination treatment appeared to counteract the NFκβ activation in fibroblasts, reducing in turn the secretion of IL6, IL11 (ELISA), sensitizing tumor cells to oxaliplatin.

TAK1 and TGFBR1 inhibition reduces metastatic potential of tumor cells

Once we had defined best in vitro treatment combination of those tested, we checked the in vivo effect of TAK1 and TGFBR1 inhibition in xenograft models on hindering the crosstalk between fibroblasts and tumor cells. First, we checked the efficacy of TAK1 inhibitor plus TGFBR1 inhibitor (galunisertib for all the in vivo experiments) in an orthotopic patient-derived xenograft model from a FOLFOX-resistant colorectal tumor with papillary histology (Fig. 6A). At the end of the experiment, three of the 9 animals were still alive with the FUOX + OXO + galunisertib treatment while only one of the animals was alive in the FUOX-treated arm. However, no statistically significant differences in survival were determined (Fig. 6B). However, there were differences in relation to the number of animals that had distant dissemination in the liver and peritoneum, significantly less in the group treated with OXO + galunisertib (Fig. 6C), as well as a trend (P = 0.07) toward a lower peritoneal tumor burden (Fig. 6D).

Figure 6.

A, Pictogram summarizing the experiment with the orthoxenograft colorectal model. Oxaliplatin L-OHP was administered in a single dose (10 mg/kg) intraperitoneally, and 5-fluorouracil 5-FU at (100 mg/kg) was provided at a weekly dose of (25 mg/kg). TAK1 inhibitor OXO was administered at 7.5 mg/kg every 3 days intraperitoneally. TGFBR1 inhibitor galunisertib was administered twice daily at 75 mg/kg by gastric instillation. B, Kaplan–Meier plot showing no differences in survival between FUOX (5-FU and L-OHP) or FUOX plus OXO plus galunisertib treatment in patient-derived xenografts (PDXs, orthotopic). Treatment lasted 30 days and was withdrawn 72 days after the orthotopic engraftment. C, There were significantly fewer animals with liver (χ2 test, P = 0.045) and peritoneal (χ2 test, P = 0.018) metastases in the group receiving FUOX plus OXO and galunisertib (purple bars; FUOX + G + OXO). The metastatic dissemination was first annotated by macroscopic examination of the peritoneal cavity, kidneys, adrenal glands, liver, and thoracic cavity (diaphragm and lungs). When the presence of metastasis was not observed macroscopically initially, histologic section was taken in sequential tissue depths and observation under a microscope by H&E staining. All the metastatic lesions (macroscopic and microscopic) were analyzed under the microscope by H&E staining and Masson trichrome. D, Moreover, the total peritoneal tumor burden in mice receiving FUOX plus OXO plus galunisertib was smaller (although not quite significantly so) than that in animals treated with FUOX (Mann–Whitney U test, P = 0.07). E, Pictogram summarizing the experiment with intrasplenic injection of tumor cells with different backgrounds and acquired resistance to L-OHP. Spleens were removed 48-hour postinjection, and animals were randomly assigned to treatment groups (L-OHP or L-OHP plus OXO plus galunisertib). After 15 days, treatment was withdrawn, and animals were euthanized 30 days later. F, In the experiment injecting HT29 cells, there were fewer animals with liver (χ2 test, P = 0.06) and lung (χ2 test, P = 0.05) metastases in the group receiving L-OHP plus OXO plus galunisertib (L-OHP + G + OXO). No differences were observed in the HT29 oxaliplatin–resistant derivative HTOXAR3. However, in experiments involving DLD1 and the oxaliplatin-resistant derivative DLDOXAR, fewer animals had liver (χ2 test P = 0.027 and P = 0.026 for DLD1 and DLDOXAR, respectively) and peritoneal (χ2 test P = 0.016 and P = 0.002, respectively) metastases in the arm treated with L-OHP plus OXO plus galunisertib (L-OHP + G + OXO). Metastatic dissemination was assessed as described above. G, The total peritoneal tumor burden in mice receiving L-OHP plus OXO plus galunisertib was significantly smaller only in the experiments with DLD1 and DLDOXAR (Mann–Whitney U test, P = 0.009 for DLD1 cells and P = 0.006 for DLDOXAR cells). H, Masson staining (intensity and extension) showing a decrease in collagen deposition in orthotopic PDX receiving FUOX plus OXO plus galunisertib (Mann–Whitney U test, P = 0.001). In the same vein, αSMA (I) and fibronectin (J) displayed less staining in tumors treated with OXO plus galunisertib in all models except for those of DLD1 and DLDOXAR. Representative images of HT29 metastases.

Figure 6.

A, Pictogram summarizing the experiment with the orthoxenograft colorectal model. Oxaliplatin L-OHP was administered in a single dose (10 mg/kg) intraperitoneally, and 5-fluorouracil 5-FU at (100 mg/kg) was provided at a weekly dose of (25 mg/kg). TAK1 inhibitor OXO was administered at 7.5 mg/kg every 3 days intraperitoneally. TGFBR1 inhibitor galunisertib was administered twice daily at 75 mg/kg by gastric instillation. B, Kaplan–Meier plot showing no differences in survival between FUOX (5-FU and L-OHP) or FUOX plus OXO plus galunisertib treatment in patient-derived xenografts (PDXs, orthotopic). Treatment lasted 30 days and was withdrawn 72 days after the orthotopic engraftment. C, There were significantly fewer animals with liver (χ2 test, P = 0.045) and peritoneal (χ2 test, P = 0.018) metastases in the group receiving FUOX plus OXO and galunisertib (purple bars; FUOX + G + OXO). The metastatic dissemination was first annotated by macroscopic examination of the peritoneal cavity, kidneys, adrenal glands, liver, and thoracic cavity (diaphragm and lungs). When the presence of metastasis was not observed macroscopically initially, histologic section was taken in sequential tissue depths and observation under a microscope by H&E staining. All the metastatic lesions (macroscopic and microscopic) were analyzed under the microscope by H&E staining and Masson trichrome. D, Moreover, the total peritoneal tumor burden in mice receiving FUOX plus OXO plus galunisertib was smaller (although not quite significantly so) than that in animals treated with FUOX (Mann–Whitney U test, P = 0.07). E, Pictogram summarizing the experiment with intrasplenic injection of tumor cells with different backgrounds and acquired resistance to L-OHP. Spleens were removed 48-hour postinjection, and animals were randomly assigned to treatment groups (L-OHP or L-OHP plus OXO plus galunisertib). After 15 days, treatment was withdrawn, and animals were euthanized 30 days later. F, In the experiment injecting HT29 cells, there were fewer animals with liver (χ2 test, P = 0.06) and lung (χ2 test, P = 0.05) metastases in the group receiving L-OHP plus OXO plus galunisertib (L-OHP + G + OXO). No differences were observed in the HT29 oxaliplatin–resistant derivative HTOXAR3. However, in experiments involving DLD1 and the oxaliplatin-resistant derivative DLDOXAR, fewer animals had liver (χ2 test P = 0.027 and P = 0.026 for DLD1 and DLDOXAR, respectively) and peritoneal (χ2 test P = 0.016 and P = 0.002, respectively) metastases in the arm treated with L-OHP plus OXO plus galunisertib (L-OHP + G + OXO). Metastatic dissemination was assessed as described above. G, The total peritoneal tumor burden in mice receiving L-OHP plus OXO plus galunisertib was significantly smaller only in the experiments with DLD1 and DLDOXAR (Mann–Whitney U test, P = 0.009 for DLD1 cells and P = 0.006 for DLDOXAR cells). H, Masson staining (intensity and extension) showing a decrease in collagen deposition in orthotopic PDX receiving FUOX plus OXO plus galunisertib (Mann–Whitney U test, P = 0.001). In the same vein, αSMA (I) and fibronectin (J) displayed less staining in tumors treated with OXO plus galunisertib in all models except for those of DLD1 and DLDOXAR. Representative images of HT29 metastases.

Close modal

We also checked the inhibition of the metastatic potential in intrasplenic injection models of colorectal cancer cell lines (HT29 and its oxaliplatin-resistant derivative HTOXAR3, and DLD1 and its oxaliplatin-resistant derivative DLDOXAR). Following the design illustrated in Fig. 6E, animals were euthanized 30 days after treatment withdrawal. Macroscopic and microscopic examination revealed that animals receiving oxaliplatin plus OXO and galunisertib exhibited lower metastatic spreading than animals receiving oxaliplatin alone, the differences being statistically significant in HT29, DLD1, and DLDOXAR cells (Fig. 6F). Moreover, the peritoneal tumor burden was also lower in OXO plus galunisertib-treated animals in DLD1 and DLDOXAR cells (Fig. 6G).

Interestingly, when we characterized the xenografts grown in mice (particularly the PDX model and the HT29 and HTOXAR models), we noticed that mice treated with the combination of OXO plus galunisertib presented tumors with less infiltration by CAFs and less collagen and extracellular matrix deposition, as shown in Fig. 6H–J. Tumors grown in DLD1 and DLDOXAR experiments recruited few fibroblasts.

In summary, the combination of TAK1 and TGFBR1 inhibition diminishes the metastatic settlement of tumor cells and the recruitment of resident fibroblasts in the TME.

The crosstalk between malignant cells and their environment, particularly CAFs in cancers with a high tumor–stroma ratio, is a fundamental event in the development of the tumorigenic process, not only for molecular signaling (30), but also for providing the necessary fuel for a demanding cancer (31, 32). Here we report the combined use of a TAK1 inhibitor plus a TGFBR1 inhibitor to avoid the IL1β and TGFβ1-mediated conversion of resident fibroblasts into CAFs, either colonic or hepatic, by inhibiting their protumorigenic protective soluble factor secretion, decreasing extracellular matrix deposition and, consequently, rendering tumor cells more sensitive to chemotherapy and inhibiting the metastatic potential and CAFs recruitment.

Tumor cells use different soluble factors and cell adhesion molecules to recruit a favorable stroma to grow and evade chemotherapy (33, 34). Our protein–protein interaction analysis identifies tumoral IL1β and TGFβ1 as the most significant soluble factors responsible for activating resident fibroblasts and their conversion into CAFs. These, in turn, trigger a cascade of molecular signaling initially mediated by P38 and SMADs that induces a massive secretion of activating ligands of JAK/STAT and AKT in tumor cells. The blocking of IL1β with a neutralizing antibody and the TGFBR1 by means of a specific inhibitor, the first hypothesis we tested, is relieved by a SMAD-independent and TAK1-mediated transient JNK activation and a sustained NFκβ-mediated switch in the cytokine repertoire, while maintaining the silencing of the SMAD-dependent pathway (according to the nonphosphorylation of SMAD2 and 3, and the high levels of BMP-4 and BMP-7, which are markers of inactivation of the canonical TGFβ pathway). In fact, the crosstalk between effector kinases has been described in various cell models (35, 36), particularly in the case of P38 and JNK (37). In addition, in the absence of TGFBR1, either by genetic deletion of pharmacologic inhibition, TGFBR2 can activate the noncanonical TGFβ pathway (38). This alternative mechanism seems to be cell-dependent, being observed in cells expressing high levels of TGFBR2, such as fibroblasts. Thus, TAK1-mediated activation of the noncanonical TGFβ pathway induces a change in the soluble factors, causing, in turn, a decrease in the activation of the JAK/STAT pathway in tumor cells. However, malignant cells sustain the AKT activation, compensating for the blocking of molecular signaling in other connected pathways, like STAT3. In fact, STAT3 inhibition has been reported to be compensated by AKT as a stress response in peripartum cardiomyopathy (39). In addition, it has been previously proposed that this AKT feedback loop might be the consequence of the capacity of different ligands to activate the same receptors tyrosine kinase (RTK). RTK redundancy has already been described (12, 40, 41), because tumor cells are addicted to cytokine/chemokine–mediated RTK signaling. In fact, some of the cytokines/chemokines that appear in the CM after blocking cocultures with the neutralizing antibody plus TGFBR1 inhibitor are molecules that activate the PI3KCA/AKT pathway. Thus, such redundancy raises the problem of acting therapeutically on convergence hubs from different combinations of RTK and RTK/cytokine pairs, rather than acting directly on ligands and membrane receptors. In this way, we may circumvent the addiction of tumor cells to RTK signaling and the plasticity of stromal cells to produce a wide repertoire of soluble factors, depending on the microenvironmental conditions. Nevertheless, toxicity arises in many cases adopting such an approach. In this regard, TAK1 is a MAPK activated by proinflammatory cytokines, chemokines, and the growth factors IL1β and TGFβ1, among others. Although TGFβ1 operates in the SMAD-dependent canonical pathway, under several circumstances it might also signal through TAK1 activating ERK (38) NFκβ transcriptional program (42), as mentioned above. However, in recent years, the role of TAK1 in cancer has been circumscribed in malignant cells (23, 43). In the context of the crosstalk between fibroblasts and tumor cells, either at the primary site or in metastasis, TAK1 is a convergence hub for IL1β and noncanonical TGFβ pathways. Thus, the second hypothesis we tested was the blocking of both IL1β-mediated and TGFβ SMAD–independent responses with a TAK1 inhibitor, and the TGFβ SMAD-dependent response with the TGFBR1 inhibitor. Very recently, it was reported that the inhibition of stromal TAK1 diminishes the senescence-associated secretory phenotype (SASP) in vitro and in vivo (44). In contrast, from our coculture experiments between fibroblasts and tumor cells, inhibiting only TAK1 was not sufficient to sensitize as many tumor cells to chemotherapy as possible or to decrease the levels of protumoral cytokines to inhibit the relevant survival pathways in tumor cells. The main reason is that the TGFβ−canonical pathway can maintain the crosstalk between fibroblasts and tumor cells. The simultaneous blockade of TAK1 and TGFBR1 manages to inhibit proinflammatory signals mediated by IL1β- and TGFβ-dependent processes, of the canonical SMAD-dependent and noncanonical SMAD-independent types. Consequently, the levels of proinflammatory cytokines were reduced along with the in vitro viability of colorectal cell lines, irrespective of using NCFs or NHFs (as coculture models of the initial steps of tumorigenesis in a primary site or in a liver metastasis) or CAFs, either with chemotherapy (L-OHP) or without. Furthermore, this combination of inhibitors seems to alter the acquisition of the proinflammatory traits of the CAFs in coculture, as determined by the inhibition of the expression of FAP and IL7R, and the maintenance of a myofibroblast marker like αSMA. Therefore, the combination of TAK1 inhibitor plus TGFBR1 inhibitor was tested in vivo, where we observed a significant decrease in metastatic growth in a patient-derived xenograft (PDX) model and in various models of oxaliplatin-sensitive, resistant derivative pairs. Moreover, when we analyzed the generated tumors histologically and IHC, we found significant alteration of the stroma in animals receiving the combination of both inhibitors. The low level of Masson staining observed in these tumors might be an indication of TGFβ pathway impairment, while the decrease in fibronectin staining is a surrogate marker for NFκβ inhibition. In some cases, the tumor size was not reduced but the stroma recruitment. This could be an opportunity to treat tumors with chemotherapy without the protection conferred by the stroma at that precise point in the tumorigenic process, rather than only at the beginning of the treatment, when they have not yet been deprived of this protective stroma.

TAK1 inhibition induces apoptosis in Kras-dependent colorectal cancer cell lines (23). In our experiments, all the cell lines used were Kras-independent. Although some are MSI, we consider that this fact does not interfere with the results obtained because the absence of an immune system, both in the in vitro and in vivo experiments, does not recapitulate the supposed differential immunosuppressive response of MSI tumors. It has also been shown that tumors of subtypes with a better prognosis, such as CMS1 (MSI) if they present high expression of CAFs genes, are clustered as tumors with a very poor prognosis (45). Also, HCT116 (MSI) is cataloged as CMS4 (46). Thus, our results are attributable to the hampering of the crosstalk rather than to the direct effect of the TAK1 inhibitor on the tumor cells.

In conclusion, the blockade of the crosstalk between fibroblasts and colorectal tumor cells by means of a TAK1 inhibitor and a TGFBR1 inhibitor hinders the acquisition of protumoral and proinflammatory traits of fibroblasts and consequently renders tumor cells more sensitive to chemotherapy. In addition, the in vivo combination of both inhibitors impaired metastatic growth and the recruitment of stroma by tumor cells.

G. Capella has ownership interests (including patents) at, is a consultant/advisory board member for, and reports receiving commercial research support from VCN Biosciences. V. Moreno is a consultant/advisory board member for Ferrer, and reports receiving commercial research support from Universal DX and Bioiberica. A. Villanueva has ownership interests (including patents) at Xenopat SL. No potential conflicts of interest were disclosed by the other authors.

Conception and design: R. Sanz-Pamplona, D.G. Molleví

Development of methodology: N.G. Díaz-Maroto, R. Sanz-Pamplona, F.J. Cimas, E. García, S. Gonçalves-Ribeiro, N. Albert, A. Villanueva, D.G. Molleví

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): N.G. Díaz-Maroto, M. Berdiel-Acer, G. Garcia-Vicién, V. Moreno, R. Salazar, A. Villanueva, D.G. Molleví

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): R. Sanz-Pamplona, E. García, V. Moreno, D.G. Molleví

Writing, review, and/or revision of the manuscript: N.G. Díaz-Maroto, R. Sanz-Pamplona, F.J. Cimas, V. Moreno, R. Salazar, D.G. Molleví

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): E. García

Study supervision: D.G. Molleví

Other (partial funding): G. Capella

This work has been supported by grant PI10/1604 from the Fondo de Investigaciones Sanitarias of the Spanish Government, Fondo Europeo de Desarrollo Regional (FEDER) “Una manera de hacer Europa”/“A way of shaping Europe,” and AGAUR grant number SGR771. This work has been supported by grants PI10/1604 and PI15/0232. In addition, D.G. Molleví and E. García are recipients of the SLT002/16/00399 grant, funded by the Department of Health of the Generalitat de Catalunya by the call “Acció Instrumental d'incorporació de Científics i Tecnòlegs.”

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.
Bissell
MJ
,
Hines
WC
. 
Why don't we get more cancer? A proposed role of the microenvironment in restraining cancer progression
.
Nat Med
2011
;
17
:
320
9
.
2.
Tao
L
,
Huang
G
,
Song
H
,
Chen
Y
,
Chen
L
. 
Cancer associated fibroblasts: an essential role in the tumor microenvironment
.
Oncol Lett
2017
;
14
:
2611
20
.
3.
Quail
DF
,
Joyce
JA
. 
Microenvironmental regulation of tumor progression and metastasis
.
Nat Med
2013
;
19
:
1423
37
.
4.
Calon
A
,
Tauriello
DVF
,
Batlle
E
. 
TGF-beta in CAF-mediated tumor growth and metastasis
.
Semin Cancer Biol
2014
;
25
:
15
22
.
5.
Yang
S
,
Pham
LK
,
Liao
CP
,
Frenkel
B
,
Reddi
AH
,
Roy-Burman
P
. 
A novel bone morphogenetic protein signaling in heterotypic cell interactions in prostate cancer
.
Cancer Res
2008
;
68
:
198
205
.
6.
Jung
DW
,
Che
ZM
,
Kim
J
,
Kim
K
,
Kim
KY
,
Williams
D
, et al
Tumor-stromal crosstalk in invasion of oral squamous cell carcinoma: a pivotal role of CCL7
.
Int J Cancer
2010
;
127
:
332
44
.
7.
Giannoni
E
,
Bianchini
F
,
Masieri
L
,
Serni
S
,
Torre
E
,
Calorini
L
, et al
Reciprocal activation of prostate cancer cells and cancer-associated fibroblasts stimulates epithelial-mesenchymal transition and cancer stemness
.
Cancer Res
2010
;
70
:
6945
56
.
8.
Lazennec
G
,
Richmond
A
. 
Chemokines and chemokine receptors: new insights into cancer-related inflammation
.
Trends Mol Med
2010
;
16
:
133
44
.
9.
Liu
J
,
Liao
S
,
Diop-Frimpong
B
,
Chen
W
,
Goel
S
,
Naxerova
K
, et al
TGF- blockade improves the distribution and efficacy of therapeutics in breast carcinoma by normalizing the tumor stroma
.
Proc Natl Acad Sci U S A
2012
;
109
:
16618
23
.
10.
Olive
KP
,
Jacobetz
MA
,
Davidson
CJ
,
Gopinathan
A
,
McIntyre
D
,
Honess
D
, et al
Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer
.
Science
2009
;
324
:
1457
61
.
11.
Zeng
Z
,
Shi
YX
,
Samudio
IJ
,
Wang
RY
,
Ling
X
,
Frolova
O
, et al
Targeting the leukemia microenvironment by CXCR4 inhibition overcomes resistance to kinase inhibitors and chemotherapy in AML
.
Blood
2009
;
113
:
6215
24
.
12.
Wilson
TR
,
Fridlyand
J
,
Yan
Y
,
Penuel
E
,
Burton
L
,
Chan
E
, et al
Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors
.
Nature
2012
;
487
:
505
9
.
13.
Klemm
F
,
Joyce
JA
. 
Microenvironmental regulation of therapeutic response in cancer
.
Trends Cell Biol
2015
;
25
:
198
213
.
14.
Jones
VS
,
Huang
RY
,
Chen
LP
,
Chen
ZS
,
Fu
L
,
Huang
RP
. 
Cytokines in cancer drug resistance: cues to new therapeutic strategies
.
Biochim Biophys Acta
2016
;
1865
:
255
65
.
15.
Ninomiya-Tsuji
J
,
Kishimoto
K
,
Hiyama
a
,
Inoue
J
,
Cao
Z
,
Matsumoto
K
. 
The kinase TAK1 can activate the NIK-I kappaB as well as the MAP kinase cascade in the IL-1 signalling pathway
.
Nature
1999
;
398
:
252
6
.
16.
Thiefes
A
,
Wolter
S
,
Mushinski
JF
,
Hoffmann
E
,
Dittrich-Breiholz
O
,
Graue
N
, et al
Simultaneous blockade of NFκB, JNK, and p38 MAPK by a kinase-inactive mutant of the protein kinase TAK1 sensitizes cells to apoptosis and affects a distinct spectrum of tumor necrosis target genes
.
J Biol Chem
2005
;
280
:
27728
41
.
17.
Irie
T
,
Muta
T
,
Takeshige
K
. 
TAK1 mediates an activation signal from toll-like receptor(s) to nuclear factor-kappaB in lipopolysaccharide-stimulated macrophages
.
FEBS Lett
2000
;
467
:
160
4
.
18.
Fechtner
S
,
Fox
DA
,
Ahmed
S
. 
Transforming growth factor beta activated kinase 1: a potential therapeutic target for rheumatic diseases
.
Rheumatology
2017
;
56
:
1060
8
.
19.
Yamaguchi
K
,
Shirakabe
K
,
Shibuya
H
,
Irie
K
,
Oishi
I
,
Ueno
N
, et al
Identification of a member of the MAPKKK family as a potential mediator of TGF-β signal transduction
.
Science
1995
;
270
:
2008
11
.
20.
Sakurai
H
,
Miyoshi
H
,
Toriumi
W
,
Sugita
T
. 
Functional interactions of transforming growth factor beta-activated kinase 1 with IkappaB kinases to stimulate NF-kappaB activation
.
J Biol Chem
1999
;
274
:
10641
8
.
21.
Lee
J
,
Mira-Arbibe
L
,
Ulevitch
RJ
. 
TAK1 regulates multiple protein kinase cascades activated by bacterial lipopolysaccharide
.
J Leukoc Biol
2000
;
68
:
909
15
.
22.
Wang
C
,
Deng
L
,
Hong
M
,
Akkaraju
GR
,
Inoue
JI
,
Chen
ZJ
. 
TAK1 is a ubiquitin-dependent kinase of MKK and IKK
.
Nature
2001
;
412
:
346
51
.
23.
Singh
A
,
Sweeney
MF
,
Yu
M
,
Burger
A
,
Greninger
P
,
Benes
C
, et al
TAK1 inhibition promotes apoptosis in KRAS-dependent colon cancers
.
Cell
2012
;
148
:
639
50
.
24.
Melisi
D
,
Xia
Q
,
Paradiso
G
,
Ling
J
,
Moccia
T
,
Carbone
C
, et al
Modulation of pancreatic cancer chemoresistance by inhibition of TAK1
.
J Natl Cancer Inst
2011
;
103
:
1190
204
.
25.
Berdiel-Acer
M
,
Sanz-Pamplona
R
,
Calon
A
,
Cuadras
D
,
Berenguer
A
,
Sanjuan
X
, et al
Differences between CAFs and their paired NCF from adjacent colonic mucosa reveal functional heterogeneity of CAFs, providing prognostic information
.
Mol Oncol
2014
;
8
:
1290
305
.
26.
Wilson
CL
,
Miller
CJ
. 
Simpleaffy: a BioConductor package for Affymetrix Quality Control and data analysis
.
Bioinformatics
2005
;
21
:
3683
5
.
27.
Tusher
VG
,
Tibshirani
R
,
Chu
G
. 
Significance analysis of microarrays applied to the ionizing radiation response
.
Proc Natl Acad Sci U S A
2001
;
98
:
5116
21
.
28.
Subramanian
A
,
Tamayo
P
,
Mootha
VK
,
Mukherjee
S
,
Ebert
BL
,
Gillette
MA
, et al
Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles
.
Proc Natl Acad Sci U S A
2005
;
102
:
15545
50
.
29.
Gonçalves-Ribeiro
S
,
Sanz-Pamplona
R
,
Vidal
A
,
Sanjuan
X
,
Guillen Díaz-Maroto
N
,
Soriano
A
, et al
Prediction of pathological response to neoadjuvant treatment in rectal cancer with a two-protein immunohistochemical score derived from stromal gene-profiling
.
Ann Oncol
2017
;
28
:
2160
8
.
30.
Kalluri
R
,
Zeisberg
M
. 
Fibroblasts in cancer
.
Nat Rev Cancer
2006
;
6
:
392
401
.
31.
Lyssiotis
CA
,
Kimmelman
AC
. 
Metabolic interactions in the tumor microenvironment
.
Trends Cell Biol
2017
;
27
:
863
75
.
32.
Avagliano
A
,
Granato
G
,
Ruocco
MR
,
Romano
V
,
Belviso
I
,
Carfora
A
, et al
Metabolic reprogramming of cancer associated fibroblasts: the slavery of stromal fibroblasts
.
Biomed Res Int
2018
;
2018
:
6075403
.
33.
Meads
MB
,
Gatenby
RA
,
Dalton
WS
. 
Environment-mediated drug resistance: a major contributor to minimal residual disease
.
Nat Rev Cancer
2009
;
9
:
665
74
.
34.
Sun
Y
. 
Tumor microenvironment and cancer therapy resistance
.
Cancer Lett
2016
;
380
:
205
15
.
35.
Mendoza
MC
,
Er
EE
,
Blenis
J
. 
The Ras-ERK and PI3K-mTOR pathways: cross-talk and compensation
.
Trends Biochem Sci
2011
;
36
:
320
8
.
36.
Britschgi
A
,
Andraos
R
,
Brinkhaus
H
,
Klebba
I
,
Romanet
V
,
Müller
U
, et al
JAK2/STAT5 inhibition circumvents resistance to PI3K/mTOR blockade: a rationale for cotargeting these pathways in metastatic breast cancer
.
Cancer Cell
2012
;
22
:
796
811
.
37.
Fey
D
,
Croucher
DR
,
Kolch
W
,
Kholodenko
BN
. 
Crosstalk and signaling switches in mitogen-activated protein kinase cascades
.
Front Physiol
2012
;
3
:
355
.
38.
Bandyopadhyay
B
,
Han
A
,
Dai
J
,
Fan
J
,
Li
Y
,
Chen
M
, et al
TbetaRI/Alk5-independent TbetaRII signaling to ERK1/2 in human skin cells according to distinct levels of TbetaRII expression
.
J Cell Sci
2011
;
124
:
19
24
.
39.
Ricke-Hoch
M
,
Bultmann
I
,
Stapel
B
,
Condorelli
G
,
Rinas
U
,
Sliwa
K
, et al
Opposing roles of Akt and STAT3 in the protection of the maternal heart from peripartum stress
.
Cardiovasc Res
2014
;
101
:
587
96
.
40.
Ozaki
K
,
Leonard
WJ
. 
Cytokine and cytokine receptor pleiotropy and redundancy
.
J Biol Chem
2002
;
277
:
29355
8
.
41.
Sun
C
,
Bernards
R
. 
Feedback and redundancy in receptor tyrosine kinase signaling: relevance to cancer therapies
.
Trends Biochem Sci
2014
;
39
:
465
74
.
42.
Zhang
YE
. 
Non-Smad pathways in TGF-beta signaling
.
Cell Res
2009
;
19
:
128
39
.
43.
McNew
KL
,
Whipple
WJ
,
Mehta
AK
,
Grant
TJ
,
Ray
L
,
Kenny
C
, et al
MEK and TAK1 regulate apoptosis in colon cancer cells with KRAS-dependent activation of proinflammatory signaling
.
Mol Cancer Res
2016
;
14
:
1204
16
.
44.
Zhang
B
,
Fu
D
,
Xu
Q
,
Cong
X
,
Wu
C
,
Zhong
X
, et al
The senescence-associated secretory phenotype is potentiated by feedforward regulatory mechanisms involving Zscan4 and TAK1
.
Nat Commun
2018
;
9
:
1723
.
45.
Calon
A
,
Lonardo
E
,
Berenguer-Llergo
A
,
Espinet
E
,
Hernando-Momblona
X
,
Iglesias
M
, et al
Stromal gene expression defines poor-prognosis subtypes in colorectal cancer
.
Nat Genet
2015
;
47
:
320
9
.
46.
Berg
KCG
,
Eide
PW
,
Eilertsen
IA
,
Johannessen
B
,
Bruun
J
,
Danielsen
SA
, et al
Multi-omics of 34 colorectal cancer cell lines - a resource for biomedical studies
.
Mol Cancer
2017
;
16
:
116
.
47.
Barbie
DA
,
Tamayo
P
,
Boehm
JS
,
Kim
SA
,
Moody
SE
,
Dunn
IF
, et al
Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1
.
Nature
2009
;
462
:
108
12
.