Toll-like receptor (TLR) ligands may be a valuable tool to promote antitumor responses by reinforcing antitumor immunity. In addition to their expression in immune cells, functional TLRs are also expressed by many cancer cells, but their significance has been controversial. In this study, we examined the action of TLR ligands on tumor pathophysiology as a result of direct tumor cell effects. B16 murine melanoma cells were stimulated in vitro with a TLR4 ligand (LPS-B16) prior to inoculation into TLR4-deficient mice (Tlr4 lps-del). Under such conditions, B16 cells yielded smaller tumors than nonstimulated B16 cells. The apoptosis/proliferation balance of the cells was not modified by TLR ligand treatment, nor was this effect compromised in immunocompromised nude mice. Mechanistic investigations revealed that IFNβ was the critical factor produced by TLR4-activated tumor cells in mediating their in vivo outgrowth. Transcriptional analysis showed that TLR4 activation on B16 cells induced changes in the expression of type I IFN and type I IFN-related genes. Most importantly, culture supernatants from LPS-B16 cells improved the maturation of bone marrow–derived dendritic cells (BMDC) from TLR4-deficient mice, upregulating the expression of interleukin-12 and costimulatory molecules on those cells. BMDC maturation was blunted by addition of an IFNβ-neutralizing antibody. Moreover, tumor growth inhibition observed in LPS-B16 tumors was abrogated in IFNAR1-deficient mice lacking a functional type I IFN receptor for binding IFN. Together, our findings show that tumor cells can be induced through the TLR4 pathway to produce IFN and positively contribute to the antitumoral immune response. Cancer Res; 72(3); 592–603. ©2011 AACR.

Toll-like receptors (TLR) recognize molecules derived from pathogens as well as endogenous danger signals possessing similar chemical structures (1, 2). Recently, functional TLRs were found to be expressed in cancer cells, but their significance remains controversial (3–12). Whereas most of the therapeutic strategies using microbial products were designed with the idea of activating TLRs present on innate immune system cells (3–10), clear distinction about the separate contribution of immune and cancer cells to the immune response has yet to be done. Stimulation of TLR4, the main receptor of bacterial lipopolysaccharide (LPS), on tumor cells has been shown to have a positive role in tumorigenesis in in vivo (13–15), but mainly in in vitro settings (16–19). When tumor expression of TLR4 or the adaptor molecule MyD88 was inhibited in tumor cells themselves, opposite conclusions were observed. This suggests a more complex scenario and that the consequences of TLR4 triggering on tumor cells could depend on the type of tumor, the way TLR4 is activated, if such activation is sustained in time, as it would be in the case of chronic activation by endogenous ligands or if it is the result of an acute process (20–24).

Stimulating MAT-LU cells, a rat prostate adenocarcinoma cell line, with LPS in vitro, before inoculation, produced significant inhibition of tumor growth in Copenhagen rats (24, 25). The same effect was observed when B16 melanoma cells were stimulated for 48 hours in vitro with LPS or monophosphoryl lipid A prior to its inoculation into syngeneic mice. This in vivo inhibition of tumor growth depends exclusively on TLR4 present on tumor cell themselves and not on antigen-presenting cells from the host, as it was not observed in TLR4-deficient mice (Tlr4lps-del; ref. 24). When LPS-stimulated B16 or MAT-LU cells were inoculated into nude mice, the growth of tumors elicited did not significantly differ from tumors induced by nonstimulated B16 or MAT-LU cells, indicating that the T-cell compartment was required (24).

These findings prompted us to determine which molecular and cellular mechanisms might be involved in this phenomenon. We have identified IFNβ, produced by TLR4-activated tumor cells as an important mediator of these effects. Our work brings in a novel aspect to discuss: under specific conditions and, probably, at specific moments of tumor development, TLR4 triggering on tumor cells could positively help the cross-talk between tumor and immune cells, favoring an antitumoral immune response.

Reagents

LPS from Escherichia coli 055:B5 and non methylated deoxycytidyl-phosphate-deoxyguanosin (CpG) were from Sigma-Aldrich (St. Louis, MO). Ultra-pure LPS from E. coli K12 was from InvivoGen.

Animals

C57BL/6 mice were from UNLP, Argentina. C57BL/10ScNJ mice with a deletion of the Tlr4 gene (Tlr4lps-del) were from the Jackson Laboratories. Il12p40−/− mice (26) were provided by Dr. Silvia Di Genaro (UNSL, Argentina). Ifnar1−/− and Il28r−/− mice were provided by Dr. Claude Libert (Ghent University, Belgium). Animals were maintained at the Animal Resource Facility of the CIBICI-CONICET in accordance with the experimental ethics committee guidelines.

Cell lines

Murine B16-F0 melanoma and TRAMPC2 prostate adenocarcinoma cell lines were obtained from American Type Culture Collection and authenticated by isoenzymology and/or the Cytochrome C subunit I (COI) PCR assay. They were periodically cultured in our laboratory for the last 10 and 5 years, respectively. The MB49 bladder cancer cell line was gently provided by Dr. IC Summerhayes (Lahey Clinic, Burlington, MA) and was used after 5 passages of propagation in supplemented medium. All cell lines were free of Mycoplasma infection tested by PCR every 6 months. Cells were stimulated in vitro with LPS for 48 hours, washed exhaustively, and inoculated subcutaneously into Tlr4lps-del mice. To generate conditioned medium (CM), cells were kept in 2.5% FBS and stimulated or not with 1 μg/mL Ultrapure LPS (LPS-B16 CM or B16-CM, respectively). After 48 hours, supernatants were collected.

Generation of DCs

Dendritic cells (DC) were obtained from bone marrow of Tlr4lps-del mice as described by Inaba and colleagues (27): more than 80% of harvested cells expressed CD11c. CD11c+ cells (2.5 × 105/mL) were incubated with LPS-B16 CM or with B16-CM and exposed to 10 μg/mL of CpG for 24 hours to analyze cytokine secretion by ELISA. LPS-B16 CM and B16-CM was incubated for 1 hour with an IFNβ-blocking antibody (4 × 102NU/mL; PBL Interferon Source), and then used to stimulate bone marrow–derived DCs (BMDC).

In vivo tumor challenge

Melanomas were established in C57BL/6, Tlr4lps-del, Il12p40−/−, Ifnar1−/−, and Il28r−/− mice by subcutaneous injection of 1 × 106 cells into the right flank. Tumor development was monitored every day as described previously (24). To evaluate the therapeutic activity of LPS, C57BL/6 and Tlr4lps-del mice were inoculated with 1 × 106 B16 or 5 × 105 MB49 cells and, once tumors reached approximately 5 mm3, they were treated intratumorally with LPS (1 μg/200 μL) or with 200 μL of PBS, every other day for 6 consecutive times.

Obtention of tumor-infiltrating cells

Tumor-infiltrating cells were obtained as previously described (24).

Quantitative reverse transcriptase PCR

B16 cells (1 × 106 cells) were stimulated or not with Ultrapure LPS for 6 and 24 hours. In the case of DCs, they were previously incubated with LPS-B16 CM or B16-CM for 20 hours prior to the addition of CpG for 4 hours. mRNA expression was analyzed with mouse toll-like receptor signaling pathway, RT2Profiler PCR Array, SABiosciences according to the manufacturer's protocol. The PCR array was done in ABI Prism7500 Cycler Detection System. The following primers were used: IFNb1 Fw5′-TTACACTGCCTTTGCCATCC, Rev5′-ACTGTCTGCTGGTGGAGTTCAT; IL-6 Fw5′-GAGGATACCACTCCCAACAGACC-3′, Rev5′-AAGTGCATCATCGTTGTTCATACA-3′; GAPDH Fw5′-TCACCACCATGGAGAAGGC-3′, Rev5′-GCTAAGCAGTTGGTGGTGCA-3′. To analyze the data the 2(-Delta Delta C(T)) method was used (28).

ELISA

Cytokines were measured by ELISA Kits (e-Biosciences, BD-Bioscience, and PBL Interferon Source) according to the manufacturer's protocol. IFNβ levels were measured in 5-fold concentrated tumor supernatants (Vivaspin sample concentrator; GE Healthcare Life Science).

Flow cytometry

Monoclonal antibodies conjugated with their respective fluorochromes were from BD-Bioscience and e-Biosciences. Intracellular cytokine was detected after stimulating cells for 5 hours with PMA (10 ng/mL) and Ionomycin (1 μg/mL; Sigma-Aldrich). Brefeldin A (10 μg/mL; Sigma) was added for the last 4 hours of cell culture. Results were analyzed using FlowJo software (Tree Star, Inc.).

Statistics

Statistical analysis was done using the Tukey posttest to ANOVA analysis with the InfoStat software (National University of Córdoba). Values of P less than 0.05 were considered significant.

Triggering TLR4 on B16 cells induces the expression of important modulators of DC activity

To analyze the expression of genes related to TLR4-mediated signal transduction and effector molecules by a qPCR array, B16 cells were either nonstimulated or stimulated with 1 μg/mL LPS for 6 and 24 hours. Distinctive TLR4 activation of downstream genes in B16 cells is shown in the gene expression heat map in Fig. 1A. Whereas more than a 6-fold induction was observed for the NFkB mediator genes after 6 hours of stimulation (Nfkb1, Nfkb2, Nfkbia, Nfkbib, RelA, MyD88, and others), most of the effector molecules were induced after 24 hours poststimulation (150x IL-6, 6x CCL2, 6x Csf2, 13x TNFa). Interestingly, an important increase in the expression of genes from the IRF pathway (160x Cxcl10, 11x Irf1), mainly IFN beta (120x) was observed (Fig. 1A). This upregulation of IFN beta expression levels was confirmed by quantitative reverse transcriptase PCR using a different set of primers (Fig. 1B). The upregulation of IFNbeta mRNA expression levels was also observed in 2 other murine tumor cell lines expressing TLR4: the MB49 and TRAMPC2 cells (Supplementary Fig. S1A). Both cell lines generated smaller tumors once activated via TLR4, prior to their in vivo inoculation in Tlr4lps-del and C57BL/6 mice (Supplementary Fig. S1B and S1C). In the case of the TRAMPC2 prostate cancer model, the inhibition of tumor growth observed was not statistically significant, but the incidence of tumor-bearing animals was lower in the LPS group (Supplementary Fig. S1C). Low levels of secreted IFNβ could also be reliably measured by ELISA in LPS-activated tumor cells (Fig. 1B and Supplementary Fig. S1A). Interleukin (IL)-6 and TNFα secreted levels increase with time in LPS-stimulated B16 cells supernatants, whereas the levels of IL-10 drop after just 12 hours of stimulation of B16 cells with LPS (Fig. 1C). In contrast, the levels of TGFβ were not modified.

Figure 1.

Triggering TLR4 on B16 cells induces the expression of important modulators of DC activity. A, heat map of qPCR array analysis. Gene expression was compared with the expression in nonstimulated B16 cells. B, IFNb1 and IL-6 mRNA expression in B16 or LPS-B16 cells normalized to glyceraldehyde-3-phosphate dehydrogenase expression. IFNβ levels in 5-fold concentrated culture supernatants from B16 cells or LPS-stimulated B16 cells (LPS-B16) evaluated by ELISA. Results indicate means ± SD of triplicate wells. *, P < 0.05. C, cytokine levels evaluated by ELISA in B16-CM and LPS-B16 cells stimulated at the indicated times. Results indicate mean ± SD of triplicate wells. *, P < 0.05.

Figure 1.

Triggering TLR4 on B16 cells induces the expression of important modulators of DC activity. A, heat map of qPCR array analysis. Gene expression was compared with the expression in nonstimulated B16 cells. B, IFNb1 and IL-6 mRNA expression in B16 or LPS-B16 cells normalized to glyceraldehyde-3-phosphate dehydrogenase expression. IFNβ levels in 5-fold concentrated culture supernatants from B16 cells or LPS-stimulated B16 cells (LPS-B16) evaluated by ELISA. Results indicate means ± SD of triplicate wells. *, P < 0.05. C, cytokine levels evaluated by ELISA in B16-CM and LPS-B16 cells stimulated at the indicated times. Results indicate mean ± SD of triplicate wells. *, P < 0.05.

Close modal

LPS-stimulated B16 cell supernatants can partially reverse the suppressive effect of B16 cell–derived factors on Tlr4lps-del BMDC maturation

Tumor-derived factors significantly inhibit the generation of DCs from hematopoietic progenitors, increase the accumulation of myeloid suppressor cells (usually characterized as GR1+ CD11b+ cells), and inhibit DCs maturation (29–31). To see if factors secreted by LPS-stimulated B16 cells could somehow overcome this inhibitory effect, we collected supernatants from B16 cells stimulated for 48 hours with 1 μg/mL Ultrapure LPS (LPS-B16 CM) or from nonstimulated cells (B16-CM) and incubated with BMDCs from Tlr4lps-del mice for 20 hours before inducing their maturation with CpG. We used BMDCs from Tlr4lps-del mice to exclude any direct effect on the maturation state of the DCs by vestigial LPS in the CM. We hypothesize that factors present in B16-CM would interfere in this process, inhibiting the transcription of key effector molecules, whereas LPS-B16 CM would do it at a lesser extent or would not do it at all. After 4 hours of stimulation of Tlr4lps-del BMDCs with CpG, transcription of genes coding for effector molecules such as IL-12a (x8), IL-1a (x12), IL-1b (x7), IL-6 (x11), TNFa (x20), Csf2 (x15), Csf3 (x16) was increased (Fig. 2A). Transcription of genes related with the NFκB complex was not observed, although the expression of inhibitors of the pathway such as NFkBia (IkBalpha) showed a moderate raise (x11). When the same analysis was done in Tlr4lps-del BMDCs that had been incubated with B16-CM and LPS-B16 CM prior to maturation, some intriguing findings were detected. Transcription of genes coding for some effector molecules was not extremely altered (such as IL-1a and b, TNF and IL-6). Other genes were extremely downregulated in both experimental groups subjected to tumor supernatants (such as Csf2 and Csf3). In contrast, differences were found in the expression of certain genes among the 3 experimental groups. When transcription of genes of the NFkB complex was evaluated, only Tlr4lps-del BMDCs matured in the presence of LPS-B16 CM showed a significant increase (x380 NFkb1, x468 Rela), indicating a more sustained or stronger stimulation of the cells. Reasonably, a strong increase in transcription levels of both inhibitor molecules such as NFkBia (IkBalpha) and NFkBib (IkBbeta) was also observed (x80 and x1250 compared with x33 and x375 in B16-CM). Interestingly, the expression of IL-12a that was inhibited in DCs incubated with B16-CM (x1.8) was partially restored when LPS-B16 CM (x5) was present at the time of maturation.

Figure 2.

LPS-B16 CM can partially reverse the suppressive effect of B16-CM on TLR4lps-del BMDC maturation. A, heat map of qPCR array analysis. TLR4lps-del BMDCs incubated with medium, B16-CM, or LPS-B16 CM for 20 hours and then stimulated with CpG for 4 hours. Gene expression was compared with that from immature TLR4lps-del BMDCs. B, cytokine levels in culture supernatants of TLR4lps-del BMDCs under the different stimulating conditions. Cytokines present in B16 and in LPS-B16 supernatants (B16 and LPS-B16) are also shown. Results indicate means ± SD of triplicate wells. ND, not detectable, *, P < 0.05. C, tumors were induced with B16 or LPS-B16 cells and their volume was measured (n = 6; mean ± SEM). Results are representative of 3 independent experiments. *, P < 0.05.

Figure 2.

LPS-B16 CM can partially reverse the suppressive effect of B16-CM on TLR4lps-del BMDC maturation. A, heat map of qPCR array analysis. TLR4lps-del BMDCs incubated with medium, B16-CM, or LPS-B16 CM for 20 hours and then stimulated with CpG for 4 hours. Gene expression was compared with that from immature TLR4lps-del BMDCs. B, cytokine levels in culture supernatants of TLR4lps-del BMDCs under the different stimulating conditions. Cytokines present in B16 and in LPS-B16 supernatants (B16 and LPS-B16) are also shown. Results indicate means ± SD of triplicate wells. ND, not detectable, *, P < 0.05. C, tumors were induced with B16 or LPS-B16 cells and their volume was measured (n = 6; mean ± SEM). Results are representative of 3 independent experiments. *, P < 0.05.

Close modal

Next, we examined the ability of B16-CM and LPS-B16 CM to modulate cytokine secretion by DCs. As expected, immature DCs (iDCs) exposed to CpG showed a significant increase in IL-12p40 secretion (Fig. 2B). A 5-fold reduction in IL-12p40 secretion was observed when iDCs were simultaneously incubated with B16-CM and CpG for 24 hours. This reduction was not due to an expansion of Gr1+CD11c+ at the expense of CD11c+ cells, because the exposure of the iDCs for 24 hours to CM did not alter the final phenotype of the cells (Supplementary Fig. S2A and B). In contrast, IL-12p40 secretion levels were partially restored when the cells were incubated with LPS-B16 CM (3-fold reduction, P < 0.05; Fig. 2B). Similar results were observed when IL-12p70 levels were evaluated: even though CpG alone is a poor inducer of IL-12p70 (32), detectable levels of IL-12p70 could be reliably measured in Tlr4lps-del DCs exposed to CpG (Fig. 2B and Supplementary Fig. S2C). These levels were almost abrogated when Tlr4lps-del DCs were incubated with B16-CM, LPS-B16 CM, and B16-CM plus CpG. In contrast, IL-12p70 was partially restored when Tlr4lps-del DCs were matured with CpG in the presence of LPS-B16 CM (Fig. 2B and Supplementary Fig. S2C). When TNFα (Fig. 2B) and IL-6 (data not shown) were measured a similar effect was observed. The levels of these cytokines present in B16-CM or LPS-B16 CM did not significantly interfere with those secreted by DCs.

TGFβ is also secreted by the tumor itself as an escape mechanism (29, 30, 33). TGFβ levels in B16-CM and LPS-B16 CM were similar to those measured in Tlr4lps-del BMDCs in basal conditions. When Tlr4lps-del BMDCs were incubated with B16-CM for 20 hours, TGFβ levels raised approximately 1.5 times compared with those detected in the B16 cells supernatants, suggesting that under these experimental conditions, DCs are also a source of TGFβ. When BMDCs were incubated with LPS-B16 CM for 20 hours, TGFβ levels dropped significantly (Fig. 2B). Surprisingly, maturation of Tlr4lps-del BMDCs in the presence of LPS-B16 CM significantly reduces the levels of TGFβ secreted, when compared with those secreted by DCs matured in the presence of B16-CM, supporting our hypothesis that DC proactivating factors are increased in LPS-B16 CM.

In vitro stimulation of B16 cells with TLR4 agonists, such as LPS or monophosphoril lipid A, for 48 hours before inoculation produced significant inhibition of tumor growth in syngeneic C57BL/6 or in Tlr4lps-del mice. Because this effect was not observed in athymic nude mice, T cells must somehow be involved. DCs are crucial in initiating a Th1 response, mainly through the secretion of IL-12. To test whether cytokines secreted by DCs were crucial intermediaries in the inhibition of tumor growth observed in LPS-B16 tumor-bearing mice, we induced tumors with B16 or LPS-B16 cells in Tlr4lps-del and IL-12p40–deficient mice. As expected, LPS-B16 tumors were significantly smaller than B16 tumors in TLR4lps-del mice; in contrast, both experimental groups (B16 and LPS-B16) followed the same pattern of tumor growth in Il12p40−/− mice, indicating that either IL-12 or IL-23 (which share the IL-12p40 subunit) is required in the inhibition of tumor growth observed in wt animals bearing LPS-B16 tumors (Fig. 2C). This experiment also suggests that DCs, one of the most prolific producers of these cytokines could be involved in the inhibition of tumor growth observed in our in vivo model.

LPS-B16 tumor–bearing mice show spleen DCs with a more mature phenotype as well as increased frequencies of IL-12+ CD11c+ and IFNγ+ tumor-infiltrating cells

Then, Tlr4lps-del mice were subcutaneously injected with B16 or LPS-B16 cells, sacrificed on day 20 and CD11c+ and GR1+ CD11b+ cells from spleens and tumors were analyzed by flow cytometry. A higher percentage of CD11c+ cells and a lower percentage of GR1+ CD11b+ cells were found in the spleens of animals bearing LPS-B16 tumors, compared with those bearing B16 tumors or normal control (Fig. 3A and Supplementary Fig. S3). Spleen CD11c+ cells from all tumor-bearing animals showed an activated phenotype, with a higher percentage of cells expressing increased levels of MHC class II and costimulatory molecules (Fig. 3B). However, a further enhancement in the expression levels of CD40, CD80 and, in some experiments, of CD86 was observed in CD11c+ cells from animals bearing LPS-B16 tumors (Fig. 3B and C).

Figure 3.

LPS-B16 tumor–bearing TLR4lps-del mice show DCs with a more mature phenotype. A, percentages of CD11c+ cells in mice injected with B16 or LPS-B16 cells. Results show mean ± SD of 3 independent experiments. *, P < 0.05. B, costimulatory molecule expression on spleen CD11c+ cells from naive (without tumor) or B16 or LPS-B16 tumor–bearing mice (continuous line); isotype control (shaded histogram). A representative experiment is shown. C, mean fluorescence intensity (MFI) values of distinct activation markers determined on spleen CD11c+ cells from naive or tumor-bearing mice. Results show mean ± SD of 3 independent experiments *, P < 0.05. D, intracellular IL-12p40 staining on tumor-infiltrating CD11c+ cells. Numbers indicate the percentage of IL-12p40+ CD11c+ cells. Shaded histogram, isotype control; continuous line, IL-12p40+CD11c+ cells (right). E, ex vivo production of IFNγ and IL-10 by total tumor-infiltrating mononuclear cells, CD3+, CD11c+, and NK1.1+ cells from B16 (black histogram) or LPS-B16 (grey histogram) tumors. A representative experiment is shown. F, IFNγ and IL-10 in culture supernatants of tumor-infiltrating mononuclear cells activated ex vivo with PMA–Ionomycin evaluated by ELISA (left). *, P < 0.05. Percentages of IFNγ, IL-10 in CD3+cells and IL-12p70 in CD11c+cells in B16 or LPS-B16 tumors (right). Results show mean ± SD of 3 independent experiments.

Figure 3.

LPS-B16 tumor–bearing TLR4lps-del mice show DCs with a more mature phenotype. A, percentages of CD11c+ cells in mice injected with B16 or LPS-B16 cells. Results show mean ± SD of 3 independent experiments. *, P < 0.05. B, costimulatory molecule expression on spleen CD11c+ cells from naive (without tumor) or B16 or LPS-B16 tumor–bearing mice (continuous line); isotype control (shaded histogram). A representative experiment is shown. C, mean fluorescence intensity (MFI) values of distinct activation markers determined on spleen CD11c+ cells from naive or tumor-bearing mice. Results show mean ± SD of 3 independent experiments *, P < 0.05. D, intracellular IL-12p40 staining on tumor-infiltrating CD11c+ cells. Numbers indicate the percentage of IL-12p40+ CD11c+ cells. Shaded histogram, isotype control; continuous line, IL-12p40+CD11c+ cells (right). E, ex vivo production of IFNγ and IL-10 by total tumor-infiltrating mononuclear cells, CD3+, CD11c+, and NK1.1+ cells from B16 (black histogram) or LPS-B16 (grey histogram) tumors. A representative experiment is shown. F, IFNγ and IL-10 in culture supernatants of tumor-infiltrating mononuclear cells activated ex vivo with PMA–Ionomycin evaluated by ELISA (left). *, P < 0.05. Percentages of IFNγ, IL-10 in CD3+cells and IL-12p70 in CD11c+cells in B16 or LPS-B16 tumors (right). Results show mean ± SD of 3 independent experiments.

Close modal

Intratumoral infiltrating mononuclear cells were also analyzed. Although the percentage of CD11c+ cells in LPS-B16 tumors did not change (Fig. 3A), the frequency of IL-12+ CD11c+ cells (Fig. 3D) was increased. Moreover, when tumor-infiltrating mononuclear cells were stimulated ex vivo with PMA–ionomycin for 5 hours and IFNγ–IL-10 cytokine intracellular staining was done, an increased frequency of IFNγ+ CD3+ cells was observed in LPS-B16 tumors compared with B16 tumors. The opposite was observed when IL-10+ CD3+ cells were analyzed (Fig. 3E and F).

Thus, LPS-B16 cells inoculated into Tlr4lps-del mice promoted a better functionality of DCs in vivo, which could be associated with a more efficient Th1 response in situ and the reduced tumor growth observed.

IFNβ produced by TLR4-activated tumor cells is involved in reversing the suppressive effect of B16 cell–derived factors on Tlr4lps-del BMDC maturation

Induction of type I IFN during stimulation of DCs through innate receptors is essential for optimal production of the IL-12 p70 heterodimer (33). Thus, we investigated the putative role of IFNβ present in LPS-B16 CM in restoring the suppressive effect of B16-CM on Tlr4lps-del BMDC maturation. We looked at CD40 expression in Tlr4lps-del BMDCs incubated with LPS-B16 or B16- CMs for 20 hours and matured with CpG. A neutralizing anti-IFNβ was added to the CMs 1 hour before incubating them with DCs. As expected, the percentage of DCs expressing CD40 increases to an 80% after 4 hours of stimulation with CpG (Fig. 4A). This increase is not affected with the addition of anti-IFNβ to the culture medium. An inhibition in the percentage of CD40+ CD11c+ cells and in the levels of CD40 expression is observed when Tlr4lps-del BMDCs were incubated with B16-CM and then matured with CpG, which is restored when the cells are matured in the presence of LPS-B16 CM. This restitution of CD40 expression was abrogated when the neutralizing anti-IFNβ was added, indicating that IFNβ is participating in the improvement of DC maturation observed (Fig. 4A and Supplementary Fig. S4). Similar results were obtained when IL-12p70 secreted by the DCs under the different conditions were measured by ELISA (Fig. 4B).

Figure 4.

IFNβ derived from LPS-stimulated B16 cells induce TLR4lps-del dendritic cell activation. A, representative CD40 expression dot plot (left) and percentages of CD40+ cells (right) in CD11c+ BMDCs cultured with or without an anti-IFNβ–blocking antibody. B, IL-12p70 levels in CD11c+ BMDCs cultured as in Fig. 2B with or without an anti-IFNβ–blocking antibody. Results show mean ± SD of triplicate experiments. C, tumors were induced with B16 or LPS-B16 in IFNAR−/− mice (n = 9). Results are representative of 2 independent experiments. *, P < 0.05 for IFNAR+/+ LPS-B16 versus IFNAR−/− LPS-B16, IFNAR+/+ B16, and IFNAR−/−B16 (mean ± SEM). D, tumors were induced with B16 or LPS-B16 in IL-28R−/− mice (n = 10). *, P < 0.05 for IL-28R+/+ LPS-B16 versus IL-28R+/+ B16 and IL-28R−/− B16 (mean ± SEM).

Figure 4.

IFNβ derived from LPS-stimulated B16 cells induce TLR4lps-del dendritic cell activation. A, representative CD40 expression dot plot (left) and percentages of CD40+ cells (right) in CD11c+ BMDCs cultured with or without an anti-IFNβ–blocking antibody. B, IL-12p70 levels in CD11c+ BMDCs cultured as in Fig. 2B with or without an anti-IFNβ–blocking antibody. Results show mean ± SD of triplicate experiments. C, tumors were induced with B16 or LPS-B16 in IFNAR−/− mice (n = 9). Results are representative of 2 independent experiments. *, P < 0.05 for IFNAR+/+ LPS-B16 versus IFNAR−/− LPS-B16, IFNAR+/+ B16, and IFNAR−/−B16 (mean ± SEM). D, tumors were induced with B16 or LPS-B16 in IL-28R−/− mice (n = 10). *, P < 0.05 for IL-28R+/+ LPS-B16 versus IL-28R+/+ B16 and IL-28R−/− B16 (mean ± SEM).

Close modal

To confirm the role that type I IFN could be playing in our model, we inoculated B16 or LPS-B16 cells into mice lacking the IFNAR1 subunit of the type I IFN receptor. Inhibition of tumor growth was observed only in wild-type mice bearing LPS-B16 tumors, indicating that type I IFN signaling is involved in tumor growth inhibition induced by activation of TLR4 expressed on tumor cells (Fig. 4C). To observe whether IL-28 (a distinct category of type I–like IFN, also referred to as type III IFN; ref. 34) could be playing a role in tumor inhibition in our model, we inoculated B16 cells stimulated or not with LPS in Il28r KO mice. Inhibition of tumor growth was observed in wild-type mice, but only a partial and nonstatistically significant inhibition of tumor growth was seen in the Il28r KO mice, indicating that type III IFNs could also be playing a role in the phenomenon (Fig. 4D).

Local stimulation of TLR4 present on tumor cells retards temporally tumor growth in TLR4-deficient mice

To investigate whether TLR4 on tumor cells play a role in therapeutic settings, we carried out local TLR4 stimulation by injecting LPS intratumorally in B16 and MB49 tumors in C57BL/6 and Tlr4lps-del mice. In both models, a significant inhibition of tumor growth was observed in C57BL/6 animals that received LPS injections once tumors became visible (Fig. 5A). Interestingly, B16 and MB49 tumors in Tlr4lps-del mice also showed a significant but transient inhibition of tumor growth that was sustained as long as the LPS treatment was maintained. Immediately after the LPS treatment was finished, tumors began to grow (Fig. 5B). However, although nonstatistically different, tumor volumes kept being slightly smaller in LPS-treated mice.

Figure 5.

Local stimulation of TLR4 present on tumor cells retards temporally tumor growth in TLR4lps-del mice. LPS or PBS was injected in C57BL/6 (A) and TLR4lps-del (B) tumor-bearing mice for 6 days (arrows) at the tumor site (n = 8). *, P < 0.05.

Figure 5.

Local stimulation of TLR4 present on tumor cells retards temporally tumor growth in TLR4lps-del mice. LPS or PBS was injected in C57BL/6 (A) and TLR4lps-del (B) tumor-bearing mice for 6 days (arrows) at the tumor site (n = 8). *, P < 0.05.

Close modal

B16-derived soluble factors strongly suppressed the activation of BMDCs in response to CpG, downregulating the secretion levels of IL-12p40, IL-12, and TNFα and diminishing the percentage of CD40+ CD11c+cells. Interestingly, when BMDCs from Tlr4lps-del mice were matured with CpG in the presence of LPS-B16 CM, the inhibition observed in the different parameters was partially reversed. Although DCs obtained from spleens of B16 and LPS-B16 tumor–bearing mice both show an activated phenotype, it is improved in the latter group. Also, an increase in IL-12+ CD11c+ cells infiltrating tumors was observed, further providing evidence of their improved functional state in vivo. Initial CD4+ T-cell activation occurs in secondary lymph nodes, but to elicit effector functions, in lymph nodes or in nonlymphoid tissues such as tumors, they need to see again the specific MHC II–peptide complexes to produce effector cytokines, such as IFNγ (35). Tumor cells do not express MHC class II molecules; therefore, infiltrating DCs would sustain CD4 T-cell stimulation (36, 37). Tumor-infiltrating DCs have already been described in B16 melanomas, in numbers even higher than those found in normal skin, being poor producers of IL-12. The fact that tumor-infiltrating mononuclear cells produce higher levels of IFNγ and reduced levels of IL-10, argues in favor of a more efficient immune response taking place in LPS-B16 tumor–bearing animals, responsible for controlling tumor growth.

Thus, upon TLR4 activation, B16 cells could secrete a different pattern of soluble factors and cytokines, which would favor the balance to a friendlier environment for activating DCs. Transcriptional analysis of LPS-stimulated B16 cells confirms this idea. The expression of GM-CSF and G-CSF genes considerably increases as well as those involved in the type I IFN pathway. Also, the levels of IL-6 and TNFα are increased in LPS-stimulated B16 cell supernatants. IL-6 has been involved in activation of STAT3 in DCs, which in turn would contribute to the inhibition of their phenotypic and functional maturation (38). In contrast, type I IFN, TNFα, and granulocyte macrophage colony-stimulating factor (GM-CSF) have potent effect in inducing maturation of DCs and have been used as adjuvant therapy for advanced-stage melanoma in patients and in mice (39). Thus, cytokines reported to have modulatory effects on DC activation could simultaneously be present at augmented or diminished levels in LPS-B16 CM making a cocktail that would provide signals to DCs that, in turn, will be differentially integrated.

The involvement of DCs in our tumor model could also be indirectly analyzed by the lack of inhibition of tumor growth induced by inoculation of LPS-stimulated B16 cells in mice deficient for IL-12p40 subunit, arguing that antigen-presenting cells, main producers of IL-12 and IL-23 and presumably DCs, play a key role in our model. Both IL-12p40 and IL-12p70 secretion levels are partially restored when TLR4−/− DCs are matured with CpG in the presence of LPS-B16 CM. Type I IFNs was an obvious candidate to be playing a role in our model because type I IFNs play a crucial role in the induction of IL-12p70 (40, 41). Type I IFNs have an established role in regulating the innate and adaptive arms of the immune system (34, 42–44), and when given exogenously to tumor patients, retard tumor growth and inhibit angiogenesis (34). Mice challenged with tumor cells that produce type I IFN as recombinant protein do not develop tumors (45–47). B16 tumors grew faster in Ifnb1−/− mice and they reach larger sizes and higher weights compared with wt mice (45–47). Also, Ifnar1−/− mice are more susceptible to the development of sarcomas induced with a chemical carcinogen. As it has been reported previously, the levels of IFNβ secreted under our experimental conditions were very low and difficult to measure with commercial ELISA kits (45, 46). U'Ren L and colleagues have found that in vitro cultured tumor tissues spontaneously released low concentrations of IFN-α (in the range of 5 pg/mL) and that concentrations of IFN-β in cultured tumor supernatants were below the level of detection of the ELISA (<100 pg/mL; ref. 46). However, these low levels were enough to inhibit the generation of tumor-associated macrophages and to restrict tumor angiogenesis (45, 46).

Type III IFNs (also known as IL-28 and IL-29) could also have a role in the inhibition of tumor growth observed in LPS-B16 tumors. They share the same intracellular signaling with type I IFNs but use a cell surface receptor, IL-28R, mainly expressed in epithelial layers (34, 43). B16 cells express IL-28R (48). LPS-B16 tumors in Il28r KO mice are not significantly smaller but still show a reduced growth compared with B16 tumors. Therefore, we could hypothesize that IL-28 (produced by either LPS-activated tumor cells or host cells) could participate in this minor effect.

In our model, IFNβ produced by TLR4-activated tumor cells is involved in restoring the suppressive effect of B16 cell–derived factors on Tlr4lps-del BMDC maturation. Also, lack of inhibition of tumor growth is observed when LPS-stimulated B16 cells are inoculated in Ifnar1−/− mice. Thus, endogenous IFNβ, secreted by LPS-stimulated B16 cells could be enough to improve the maturation state of local DCs, promoting the secretion of IL-12 and then a more efficient antitumoral response (Fig. 6). Our findings show, for the first time, that tumor cells can be manipulated with classical adjuvants to contribute positively to the antitumoral immune response.

Figure 6.

Proposed model (1) LPS-B16 cells secrete IFNβ and other modulators of DC activity; (2) IFNβ can significantly restore the IL-12–secreting capacity of DCs at the site of inoculation, improving their maturation state; (3) DCs promote a Th1 response against the growing tumor (4).

Figure 6.

Proposed model (1) LPS-B16 cells secrete IFNβ and other modulators of DC activity; (2) IFNβ can significantly restore the IL-12–secreting capacity of DCs at the site of inoculation, improving their maturation state; (3) DCs promote a Th1 response against the growing tumor (4).

Close modal

Another aspect that should be discussed is whether this positive contribution could actually happen in more realistic scenarios such as therapeutic settings in which the adjuvant is administered once tumors are visible. Only a transitory halt of tumor growth was observed in both, B16 and MB49 tumors in Tlr4lps-del animals which were intratumorally treated with LPS, indicating that type I IFN is necessary but not sufficient. In both cases, it has to be highlighted that even in the absence of TLR4 on innate immune cells (which are the expected target of LPS), tumor growth is controlled by the LPS treatment (at least transiently) in a context in which it can only be recognized by tumor cells.

The role of TLRs and, particularly, TLR4 in tumorigenesis and tumor promotion is highly controversial. Further research in this topic will open up new avenues for understanding tumor biology and for identifying potential new therapy strategies for cancer.

No potential conflicts of interest were disclosed.

This work was supported by grants from ANPCYT-PICT 2007-0974 and PICT 2006-02369; FWO-MINCyT 0904, CONICET 2008-6437. V. Andreani and M.I. Crespo are PhD fellows of CONICET. N.G. Núñez and D.A. Nocera are PhD fellows of FONCyT. M. Maccioni, G. Morón, and V. Rivero are members of the Researcher Career of CONICET.

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.
Takeuchi
O
,
Akira
S
. 
Pattern recognition receptors and inflammation
.
Cell
2010
;
140
:
805
20
.
2.
Kawai
T
,
Akira
S
. 
The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors
.
Nat Immunol
2010
;
11
:
373
84
.
3.
Dubensky
TW
,
Reed
SG
. 
Adjuvants for cancer vaccines
.
Semin Immunol
2010
;
22
:
155
61
.
4.
Didierlaurent
AM
,
Morel
S
,
Lockman
L
,
Giannini
SL
,
Bisteau
M
,
Carlsen
H
, et al
AS04, an aluminum salt- and TLR4 agonist-based adjuvant system, induces a transient localized innate immune response leading to enhanced adaptive immunity
.
J Immunol
2009
;
183
:
6186
97
.
5.
Chicoine
MR
,
Zahner
M
,
Won
EK
,
Kalra
RR
,
Kitamura
T
,
Perry
A
, et al
The in vivo antitumoral effects of lipopolysaccharide against glioblastoma multiforme are mediated in part by Toll-like receptor 4
.
Neurosurgery
2007
;
60
:
372
80
.
6.
Garay
RP
,
Viens
P
,
Bauer
J
,
Normier
G
,
Bardou
M
,
Jeannin
JF
, et al
Cancer relapse under chemotherapy: why TLR2/4 receptor agonists can help
.
Eur J Pharmacol
2007
;
563
:
1
17
.
7.
de Bono
JS
,
Dalgleish
AG
,
Carmichael
J
,
Diffley
J
,
Lofts
FJ
,
Fyffe
D
, et al
Phase I study of ONO-4007, a synthetic analogue of the lipid A moiety of bacteria lipopolysaccharide
.
Clin Cancer Res
2000
;
6
:
397
405
.
8.
Okamoto
M
,
Oshikawa
T
,
Tano
T
,
Sasai
A
,
Kan
S
,
Moriya
Y
, et al
Mechanism of anticancer host response induced by OK-432, a streptococcal preparation, mediated by phagocytosis and Toll-like receptor 4 signaling
.
J Immunother
2006
;
29
:
78
86
.
9.
Engelhardt
R
,
Mackensen
A
,
Galanos
C
. 
Phase I trial of intravenously administered endotoxin (Salmonella abortus equi) in cancer patients
.
Cancer Res
1991
;
51
:
2524
30
.
10.
Lundin
JI
,
Checkoway
H
. 
Endotoxin and cancer
.
Environ Health Perspect
2009
;
117
:
1344
50
.
11.
Rakoff-Nahoum
S
,
Medzhitov
R
. 
Toll-like receptors and cancer
.
Nat Rev Cancer
2009
;
9
:
57
63
.
12.
Apetoh
L
,
Ghiringhelli
F
,
Tesniere
A
,
Obeid
M
,
Ortiz
C
,
Criollo
A
, et al
Toll like receptor 4-dependent contribution of the immune system to anticancer chemotherapy
.
Nature Med
2007
;
13
:
1050
9
.
13.
Earl
TM
,
Nicoud
IB
,
Pierce
JM
,
Wright
JP
,
Majoras
NE
,
Rubin
JE
, et al
Silencing of TLR4 decreases liver tumor burden in a murine model of colorectal metastasis and hepatic steatosis
.
Ann Surg Oncol
2009
;
16
:
1043
50
.
14.
Mittal
D
,
Saccheri
F
,
Vénéreau
E
,
Pusterla
T
,
Bianchi
ME
,
Rescigno
M
. 
TLR4-mediated skin carcinogenesis is dependent on immune and radioresistant cells
.
EMBO J
2010
;
29
:
2242
5
.
15.
Huang
B
,
Zhao
J
,
Li
H
,
He
KL
,
Chen
Y
,
Chen
SH
, et al
Toll-like receptors on tumor cells facilitate evasion of immune surveillance
.
Cancer Res
2005
;
65
:
5009
13
.
16.
Doan
HQ
,
Bowen
KA
,
Jackson
LA
,
Evers
BM
. 
Toll-like receptor 4 activation increases Akt phosphorylation in colon cancer cells
.
Anticancer Res
2009
;
29
:
2473
8
.
17.
Szczepanski
MJ
,
Czystowska
M
,
Szajnik
M
,
Harasymczuk
M
,
Boyiadzis
M
,
Kruk-Zagajewska
A
, et al
Triggering of Toll-like receptor 4 expressed on human head and neck squamous cell carcinoma promotes tumor development and protects the tumor from immune attack
.
Cancer Res
2009;
69
:
3105
13
.
18.
Fukata
M
,
Chen
A
,
Klepper
A
,
Krishnareddy
S
,
Vamadevan
AS
,
Thomas
LS
, et al
Cox-2 is regulated by toll-like receptor-4 (TLR4) signaling: Role in proliferation and apoptosis in the intestine
.
Gastroenterology
2006
;
131
:
862
77
.
19.
Szajnik
M
,
Szczepanski
MJ
,
Czystowska
M
,
Elishaev
E
,
Mandapathil
M
,
Nowak-Markwitz
E
, et al
TLR4 signaling induced by lipopolysaccharide or paclitaxel regulates tumor survival and chemoresistance in ovarian cancer
.
Oncogene
2009
;
28
:
4353
63
.
20.
Yang
HZ
,
Cui
B
,
Liu
HZ
,
Mi
S
,
Yan
J
,
Yan
HM
, et al
Blocking TLR2 activity attenuates pulmonary metastases of tumor
.
PLoS One
2009
;
4
:
6520
27
.
21.
Rhee
SH
,
Im
E
,
Pothoulakis
C
. 
Toll-like receptor 5 engagement modulates tumor development and growth in a mouse xenograft model of human colon cancer
.
Gastroenterology
2008
;
135
:
518
28
.
22.
Yusuf
N
,
Nasti
TH
,
Long
JA
,
Naseemuddin
M
,
Lucas
AP
,
Xu
H
, et al
Protective role of Toll-like receptor 4 during the initiation stage of cutaneous chemical carcinogenesis
.
Cancer Res
2008
;
68
:
615
22
.
23.
Nair
P
,
O'Donnell
CM
,
Janasek
K
,
Sajduk
MK
,
Smith
EA
,
Golden
JM
, et al
Lipopolysaccharide-treated mammary carcinomas secrete proinflammatory chemokines and exhibit reduced growth rates in vivo, but not in vitro
.
Immunol Invest
2009
;
38
:
730
48
.
24.
Andreani
V
,
Gatti
G
,
Simonella
L
,
Rivero
V
,
Maccioni
M
. 
Activation of Toll-like receptor 4 on tumor cells in vitro inhibits subsequent tumor growth in vivo
.
Cancer Res
2007
;
67
:
10519
27
.
25.
Gatti
G
,
Rivero
V
,
Motrich
RD
,
Maccioni
M
. 
Prostate epithelial cells can act as early sensors of infection by up-regulating TLR4 expression and proinflammatory mediators upon LPS stimulation
.
J Leukoc Biol
2006
;
79
:
989
98
.
26.
Magram
J
,
Connaughton
SE
,
Warrier
RR
,
Carvajal
DM
,
Wu
CY
,
Ferrante
J
, et al
IL-12-deficient mice are defective in IFN gamma production and type 1 cytokine responses
.
Immunity
1996
;
4
:
471
81
.
27.
Inaba
K
,
Swiggard
WJ
,
Steinman
RM
,
Romani
N
,
Schuler
G
,
Brinster
C
. 
Isolation of dendritic cells
.
Curr Protoc Immunol
2009
;
Chapter 3:Unit 3.7
.
28.
Livak
KJ
,
Schmittgen
TD
. 
Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T))
.
Methods
2001
;
25
:
402
8
.
29.
Rabinovich
GA
,
Gabrilovich
D
,
Sotomayor
EM
. 
Immunosuppressive strategies that are mediated by tumor cells
.
Annu Rev Immunol
2007
;
25
:
267
96
.
30.
Kusmartsev
S
,
Gabrilovich
DI
. 
Effect of tumor-derived cytokines and growth factors on differentiation and immune suppressive features of myeloid cells in cancer
.
Cancer Metastasis Rev
2006
;
25
:
323
31
.
31.
Idoyaga
J
,
Moreno
J
,
Bonifaz
L
. 
Tumor cells prevent mouse dendritic cell maturation induced by TLR ligands
.
Cancer Immunol Imunother
2007
;
56
:
1237
50
.
32.
Gautier
G
,
Humbert
M
,
Deauvieau
F
,
Scuiller
M
,
Hiscott
J
,
Bates
EE
, et al
A type I interferon autocrine-paracrine loop is involved in Toll-like receptor-induced interleukin-12p70 secretion by dendritic cells
.
J Exp Med
2005
;
201
:
1435
46
.
33.
Liu
VC
,
Wong
LY
,
Jang
T
,
Shah
AH
,
Park
I
,
Yang
X
, et al
Tumor evasion of the immune system by converting CD4+CD25- T cells into CD4+CD25+ T regulatory cells: role of tumor-derived TGF-beta
.
J Immunol
2007
;
178
:
2883
92
.
34.
Coccia
EM
. 
IFN regulation and functions in myeloid dendritic cells
.
Cytokine Growth Factor Rev
2008
;
19
:
21
32
.
35.
Reinhardt
RL
,
Bullard
DC
,
Weaver
CT
,
Jenkins
MK
. 
Preferential accumulation of antigen-specific effector CD4 T cells at an antigen injection site involves CD62E-dependent migration but not local proliferation
.
J Exp Med
2003
;
197
:
751
62
.
36.
Gerner
MY
,
Mescher
MF
. 
Antigen processing and MHC-II presentation by dermal and tumor-infiltrating dendritic cells
.
J Immunol
2009
;
182
:
2726
37
.
37.
Preynat-Seauve
O
,
Schuler
P
,
Contassot
E
,
Beermann
F
,
Huard
B
,
French
LE
. 
Tumor-infiltrating dendritic cells are potent antigen-presenting cells able to activate T cells and mediate tumor rejection
.
J Immunol
2006
;
176
:
61
7
.
38.
Alshamsan
A
,
Hamdy
S
,
Das
S
,
Lavasanifar
A
,
Samuel
J
,
El-Kadi
AO
. 
Validation of bone marrow derived dendritic cells as an appropriate model to study tumor-mediated suppression of DC maturation through STAT3 hyperactivation
.
J Pharm Pharm Sci
2010
;
13
:
21
6
.
39.
Kalaaji
AN
,
Lu
J
,
Markovic
SN
,
Celis
E
,
Pittelkow
MR
. 
Combination cytokine therapy inhibits tumor growth by generation of tumor-specific T-cell responses in a murine melanoma model
.
Cytokine
2010;
49
:
287
93
.
40.
Gerosa
F
,
Baldani-Guerra
B
,
Lyakh
LA
,
Batoni
G
,
Esin
S
,
Winkler-Pickett
RT
, et al
Differential regulation of interleukin 12 and interleukin 23 production in human dendritic cells
.
J Exp Med
2008
;
205
:
1447
61
.
41.
Trinchieri
G
,
Pflanz
S
,
Kastelein
RA
. 
The IL-12 family of heterodimeric cytokines: new players in the regulation of T cell responses
.
Immunity
2003
;
19
:
641
4
.
42.
Dunn
GP
,
Koebel
CM
,
Schreiber
RD
. 
Interferons, immunity and cancer immunoediting
.
Nat Rev Immunol
2006;
6
:
836
48
.
43.
Trinchieri
G
. 
Type I interferon: friend or foe?
J Exp Med
2010
;
207
:
2053
63
.
44.
Jonasch
E
,
Haluska
FG
. 
Interferon in oncological practice: review of interferon biology, clinical applications, and toxicities
.
Oncologist
2001;
6
:
34
55
45.
Jablonska
J
,
Leschner
S
,
Westphal
K
,
Lienenklaus
S
,
Weiss
S
. 
Neutrophils responsive to endogenous IFN-beta regulate tumor angiogenesis and growth in a mouse tumor model
.
J Clin Invest
2010
;
120
:
1151
64
.
46.
U'Ren
L
,
Guth
A
,
Kamstock
D
,
Dow
S
. 
Type I interferons inhibit the generation of tumor-associated macrophages
.
Cancer Immunol Immunother
2010
;
59
:
587
98
.
47.
Qin
XQ
,
Tao
N
,
Dergay
A
,
Moy
P
,
Fawell
S
,
Davis
A
, et al
Interferon-beta gene therapy inhibits tumor formation and causes regression of established tumors in immune-deficient mice
.
Proc Natl Acad Sci U S A
1998
;
95
:
14411
6
.
48.
Wongthida
P
,
Diaz
RM
,
Galivo
F
,
Kottke
T
,
Thompson
J
,
Pulido
J
, et al
Type III IFN interleukin-28 mediates the antitumor efficacy of oncolytic virus VSV in immune-competent mouse models of cancer
.
Cancer Res
2010
;
70
:
4539
49
.