Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • Log out
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Cancer Research
Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Immunology

Transforming Growth Factor β Inhibits the Antigen-Presenting Functions and Antitumor Activity of Dendritic Cell Vaccines

James J. Kobie, Rita S. Wu, Robert A. Kurt, Sunming Lou, Miranda K. Adelman, Luke J. Whitesell, Lalitha V. Ramanathapuram, Carlos L. Arteaga and Emmanuel T. Akporiaye
James J. Kobie
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Rita S. Wu
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Robert A. Kurt
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sunming Lou
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Miranda K. Adelman
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Luke J. Whitesell
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lalitha V. Ramanathapuram
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Carlos L. Arteaga
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Emmanuel T. Akporiaye
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI:  Published April 2003
  • Article
  • Info & Metrics
  • PDF
Loading

Abstract

Dendritic cell (DC)-based vaccines have exhibited minimal effectiveness in treating established tumors, likely because of factors present in the tumor microenvironment. One such factor is transforming growth factor β (TGF-β), a cytokine that is produced by numerous tumor types and has been demonstrated to impair DC functions in vitro. We have evaluated the effect of TGF-β on the immunostimulatory activities of DCs. We demonstrate that TGF-β exposure inhibits the ability of DCs to present antigen, stimulate tumor-sensitized T lymphocytes, and migrate to draining lymph nodes. Neutralization of TGF-β using the TGF-β-neutralizing monoclonal antibody 2G7 enhanced the ability of DC vaccines to inhibit the growth of established 4T1 murine mammary tumors. Treatment of 4T1 tumors transduced with the antisense TGF-β transgene (4T1-asT) with the combination of DC and 2G7 monoclonal antibody inhibited tumor growth and resulted in complete regression of tumors in 40% of the mice. These results demonstrate that neutralization of TGF-β in tumor-bearing mice enhances the efficacy of DC-based vaccines.

INTRODUCTION

In recent years, DCs 4 have become popular candidates in cancer vaccine development because of their crucial role in inducing T-cell responses. Upon antigen uptake, DCs residing in peripheral tissues internalize and process antigen and migrate to secondary lymphoid organs where they stimulate naïve T lymphocytes in the context of class I and class II MHC antigens (1) . The effectiveness of DCs as antigen-presenting cells provides the rationale for their use as cancer vaccines with the objective of inducing durable antitumor immune responses. In numerous rodent models, vaccines consisting of tumor antigen-pulsed DCs are effective in inducing CTL responses and providing protection against subsequent tumor challenge (2, 3, 4, 5) . In contrast, DC vaccines have been less effective in abrogating established tumors in mice (2, 3, 4, 5) and human cancer patients (6) . The insensitivity of established tumors to DC therapy is likely because of factors present within the tumor microenvironment that are inimical to the optimal induction of an antitumor immune response (7) . Examination of circulating and tumor-infiltrating DCs in tumor-bearing animals and in cancer patients has revealed that DCs are functionally impaired in their ability to induce T-cell responses (8, 9, 10, 11) . These deficits have been associated with down-regulation of MHC and costimulatory molecules (10 , 11) and tumor-induced apoptosis of DCs (12) .

One of the factors produced within the tumor microenvironment that might interfere with DC functions is TGF-β. TGF-β is a pleiotropic cytokine produced by cancer cells of different histological types (13, 14, 15, 16) . Among the plethora of immunosuppressive effects of TGF-β (17) is the capacity to interfere with several DC functions. These include down-regulation of cell surface MHC antigens, costimulatory molecules, chemokine receptors, as well as impairment of in vitro chemotaxis (18, 19, 20) .

Although the in vitro effects of TGF-β on DCs are relatively well known, the impact of TGF-β on the ability of DCs to migrate to secondary lymphoid organs and induce specific antitumor T-cell responses in vivo remains to be determined. In this study, we demonstrate that TGF-β inhibits DC migration to DLN and diminishes their capacity to stimulate IFN-γ secretion by tumor-sensitized T lymphocytes. Most importantly, we show that the combined use of antisense TGF-β gene transfer plus TGF-β-neutralizing antibody increases the efficacy of DC vaccines in treating established TGF-β-secreting 4T1 mammary tumors.

MATERIALS AND METHODS

Animals.

Six-week-old female BALB/c and C57BL/6 (B6) mice were purchased from The Harlan Laboratory (Indianapolis, IN). Six-week-old female BALB/c-TgN (DO11.10) 10 Loh mice were purchased from The Jackson Laboratory (Bar Harbor, ME). All mice were housed at the University of Arizona Animal Facilities in accordance with the Principles of Animal Care (NIH publication no. 85-23, revised 1985).

Tumors.

4T1 murine mammary tumor cells were kindly provided by Dr. Fred Miller (Michigan Cancer Foundation, Detroit, MI) and maintained as described previously (21) .

TGF-β-neutralizing Antibody (2G7).

The 2G7 mouse IgG1 mAb was generated after immunization of BALB/c mice with recombinant human TGF-β1. 2G7 neutralized the growth inhibitory activity of TGF-β1, TGF-β2, and TGF-β3 on Mv1Lu epithelial cells (22) .

Generation of DCs and TGF-β Treatment.

Bone marrow cells were harvested from flushed marrow cavities of femurs and tibiae under aseptic conditions and cultured with 100 units/ml granulocyte macrophage colony-stimulating factor and 100 units/ml interleukin 4 (Peprotech, Rocky Hill, NJ) at 106 cells/ml in complete media (RPMI 1640 containing 10% heat-inactivated FBS, 0.1 mm nonessential amino acids, 1 μm sodium pyruvate, 2 mm l-glutamine, 100 μg/ml streptomycin, 100 units/ml penicillin, 0.5 μg/ml fungizone, and 5 × 10−5 m 2-mercaptoethanol). Cytokines were replenished on day 4. On day 6 of culture, DCs were collected and cultured at 106 cells/ml with granulocyte macrophage colony-stimulating factor and interleukin 4 with or without the addition of 10 ng/ml of recombinant human TGF-β1 (R&D Systems, Minneapolis, MN) for 6 days. DCs were matured with 200 units/ml of TNF-α (Peprotech) for 48 h.

FACS Analysis.

All antibodies used were purchased from Caltag Laboratories (Burlingame, CA) unless otherwise noted. For analysis of DCs, samples were stained with PE-conjugated anti-CD11c (BD PharMingen, San Diego, CA), FITC-conjugated anti-I-Ad (BD PharMingen), PE-conjugated anti-B7.1 (CD80), FITC-conjugated anti-B7.2 (CD86), or PE-conjugated anti-CD40. T cells were stained with PE-conjugated anti-CD3, FITC-conjugated anti-B220, PE-conjugated anti-CD8, or FITC-conjugated anti-CD4. Cells were analyzed using a FACStarPLUS flow cytometer (Becton Dickinson Immunocytometry Systems, San Jose, CA).

Induction of Allogeneic Mixed Lymphocyte Reactions.

Spleen cells from B6 mice were harvested and enriched for CD3-positive cells using a T-cell enrichment column (R&D Systems). Cells were 80–95% CD3 positive as determined by FACS analysis. Varying numbers of DCs were incubated with 2 × 105 T lymphocytes for 5 days in 96-well tissue culture plates (Sarstedt, Newton, NC) with the addition of 1 μCi of [3H]thymidine (Perkin-Elmer Life Sciences, Boston, MA) for the final 18 h of culture.

OVA Peptide Presentation Assay.

Spleen cells from DO11.10 OVA T cell receptor transgenic mice were enriched for CD3 positive cells as described above. Varying numbers of DCs were incubated with 2 × 105 T lymphocytes in the presence of 1 μm of OVA peptide (ISQAVHAAHAEINEAGR; United Biochemical Research, Seattle, WA) in 96-well tissue culture plates for 5 days with the addition of 1 μCi of [3H]thymidine for the final 18 h of culture.

Endocytosis and Phagocytosis Assays.

Endocytosis and phagocytosis assays were performed using modifications of previously described procedures (23) . Endocytosis was measured by incubating 2 × 105 DC with 400 μg of FITC-conjugated dextran beads, 40,000 MW (Molecular Probes, Eugene, OR) for 30 min at 4°C or 37°C. Phagocytosis was measured by incubating DCs with FITC-conjugated Escherichia coli (Molecular Probes) at a ratio of 100 E. coli particles to 1 DC for 60 min at 4°C or 37°C. After incubation, cells were washed extensively with PBS containing 0.5% bovine albumin and 0.1% sodium azide and analyzed by flow cytometry.

Stimulation of Tumor-sensitized T Lymphocytes.

Bone marrow-derived DCs were pulsed with 4T1 tumor cell lysate at a ratio of 3 tumor cell equivalents/DC for 24 h in the presence or absence of 10 ng/ml TGF-β1. DCs were then matured with 20 ng/ml TNF-α in the presence or absence of 10 ng/ml TGF-β1. Splenic T lymphocytes were purified from mice bearing 14 day 4T1 tumors as indicated above. Ten thousand DCs were incubated with 2 × 105 splenic T lymphocytes in 96-well tissue culture plates for 5 days with the addition of 1 μCi of [3H]thymidine for the final 18 h of culture. One million tumor DLN cells from mice bearing 14 day 4T1 tumors were incubated with 2.5 × 105 DCs for 48 h, and IFN-γ production was evaluated by ELISA (R&D Systems).

In Vivo Migration Assay.

Bone marrow-derived DCs were matured with 200 units/ml TNF-α with or without 10 ng/ml TGF-β1 for 48 h. DCs were labeled with 10 μm PKH-67L, green fluorescent dye (Sigma, St. Louis, MO) as described previously (20) . Naive mice received s.c. injections in the right flank with 5–8 × 106 DCs. Forty-eight h after injection, mice were sacrificed, and inguinal lymph nodes were harvested and disaggregated. Lymph node cells were centrifuged (Shandon, Pittsburgh, PA) onto glass slides at 700 rpm for 4 min. The slides were fixed with 4% paraformaldehyde and stained with a propidium iodide/RNase solution (Phoenix Flow Systems, San Diego, CA). Slides were analyzed using a laser scanning cytometer (CompuCyte, Cambridge, MA). Detection and contouring of cells was keyed by propidium iodide signal, whereas DCs were identified by their green fluorescence signal; 35,000 propidium iodide events were analyzed from each treatment group/experiment. The identity of each DC detected during scanning was confirmed visually by direct microscopic observation using the instrument’s relocation function.

In Vitro Chemotaxis Assay.

Bone marrow-derived DCs were matured with TNF-α in the presence or absence of TGF-β1 for 48 h as indicated above. An in vitro chemotaxis assay was performed as described previously (24) . SLC (Peprotech) and MIP-3β (R&D Systems) were diluted with serum-free media to a final volume of 600 μl of 100 ng/ml chemokine and added to 24-well tissue culture plates (Corning Costar, Cambridge, MA). Transwell culture inserts (Corning Costar) with 6.5-mm diameter and 5.0-μm pore-size were inserted into each well, and DCs (4 × 105 cells/each well) were added to the top chamber in serum-free media at a final volume of 100 μl. After the plates were incubated at 37°C in 5% CO2 for 4 h, the cells in the bottom chamber were recovered, the migrating cells were counted, and an aliquot was stained with anti-CD11c mAbs to be analyzed by FACS. Controls included wells with chemokine in both the top and bottom chambers. The number of migrated cells was determined by subtracting the number of migrated cells in control wells from the number of migrated cells in experimental wells.

Treatment of Established Tumors.

Six-week-old BALB/c mice were orthotopically injected with 104 4T1-N or 4T1-asT tumor cells into the mammary gland. DCs were pulsed with 4T1 tumor cell lysate at a ratio of 3 tumor cell equivalents/dendritic cell for 24 h. After pulsing, DCs were matured with 200 units/ml TNF-α for 48 h. Mice were injected i.t. with 1.5 × 106 tumor cell lysate-pulsed, matured DCs in 50 μl of PBS on day 15 when tumors were palpable. Vaccination was repeated on days 20 and 25. Two h before each vaccination, mice received i.p. injections of 300 μg of 2G7 mAb. In combination with DCs or alone, mice received 100 μg of 2G7 mAb i.t. Primary tumors were measured as reported previously (21) . Mice exhibiting complete tumor regression were challenged with 10-fold more 4T1 tumor cells (105) and monitored for tumor growth.

Statistical Analysis.

For all analyses, student t tests were performed using Prism software (GraphPad, San Diego, CA). Ps of <0.05 were considered to indicate significant differences between data sets.

RESULTS

Antigen Uptake and Presentation by DCs.

Before evaluating the effect of TGF-β in vivo, we evaluated its effects on several DC functions in vitro. Treatment of DCs with TGF-β caused a significant (P < 0.05) decrease in the levels of B7.1 and CD40 (Fig. 1A) ⇓ , as well as in the percentage of cells expressing CD40 (Fig. 1B) ⇓ . Next, we evaluated the ability of DCs to endocytose dextran particles and phagocytose E. coli particles after TGF-β treatment. TGF-β exposure inhibited the endoocytic and phagocytic capacities of DCs by 58% and 87%, respectively (Fig. 1C), a pattern that was observed in all three experiments conducted. To evaluate the effect of TGF-β exposure on antigen presentation, mixed lymphocyte reactions were performed. Stimulation of allogeneic T lymphocytes (Fig. 1D) ⇓ and presentation of OVA peptide (Fig. 1E) ⇓ by DCs was significantly (P < 0.05) suppressed after TGF-β exposure. These results demonstrate that TGF-β inhibits the uptake and presentation of antigens by DCs.

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

Effect of TGF-β exposure on DC functions in vitro. DCs were incubated in the absence or presence of 10 ng/ml TGF-β for 6 days. Cytokines were replenished every 2 days. A and B, DCs were stained with anti-CD11c, anti-I-Ad, anti-B7.1, anti-B7.2, and anti-CD40 antibodies and analyzed by flow cytometry. The results are mean ± SE of three independent experiments. C, DCs were incubated with FITC-conjugated dextran particles or FITC-conjugated E. coli particles at 4°C or 37°C, fixed, and analyzed by flow cytometry. Values represent mean fluorescence intensity (MFI) at 37°C minus 4°C. D, DCs were incubated with 2 × 105 splenic T cells isolated from C57/BL6 mice for 5 days with the addition of [3H]thymidine for the last 18 h of culture. Values represent mean ± SE of six replicates. E, DCs were collected and incubated with 2 × 105 splenic T cells isolated from BALB/c-TgN (DO11.10) 10 Loh mice in the presence of OVA peptide for 5 days with the addition of [3H]thymidine for the last 18 h of culture. Values represent mean ± SE of four replicates. Results are from one representative experiment of three independent experiments. ∗ refers to statistical significance between groups (P < 0.05).

Induction of Tumor-sensitized T Lymphocytes.

Observing that TGF-β inhibits the antigen uptake and presentation capacities of DCs, we evaluated its effect on the stimulation of tumor-sensitized T lymphocytes by tumor cell lysate-pulsed immature and mature DCs. TGF-β-treated mature DCs were significantly less effective (P = 0.0002) than untreated mature DCs at stimulating the proliferation of tumor-sensitized T lymphocytes (Fig. 2A) ⇓ . The ability of immature and mature DCs to stimulate IFN-γ production by tumor-sensitized lymphocytes was also significantly inhibited (P < 0.0001, P = 0.0003) by 60 and 22%, respectively, after TGF-β exposure. Taken together, these data demonstrate that TGF-β exposure reduces the ability of DCs to stimulate antitumor immune responses.

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

Stimulation of tumor-sensitized T lymphocytes. DCs were pulsed with tumor cell lysate for 48 h in the presence or absence of 10 ng/ml TGF-β (immature), then cultured in TNF-α in the presence or absence of TGF-β for 48 h (mature). A, ten thousand DCs were incubated with 2 × 105 splenic T cells isolated from mice bearing 4T1 tumors for 5 days, and [3H]thymidine was added for the last 18 h of culture. B, one million T cells isolated from lymph nodes draining 4T1 tumors were incubated with 2.5 × 105 DCs for 48 h. After incubation, supernatant was analyzed for IFN-γ production. Numbers are mean ± SE of triplicate samples. Results are from one representative experiment of two independent experiments. ∗ refers to statistical significance between groups (P < 0.05).

In Vivo Migration and In Vitro Chemotaxis.

Because migration of DCs to secondary lymphoid organs is necessary for T-cell priming (25) , we assessed the effect of TGF-β treatment on the ability of mature DC to migrate to DLNs. For this purpose, TGF-β-treated DCs were labeled with PKH-67 and injected s.c. into the hind flank of mice. Forty-eight h later, draining inguinal lymph nodes were harvested, and infiltrating DCs were enumerated by scanning laser cytometry. TGF-β treatment resulted in a decrease in migration of mature DCs to the DLNs (Table 1) ⇓ . This was a significant (P < 0.05) trend observed over four independent experiments. To determine the mechanism responsible for decreased migration of mature DCs, in vitro chemotaxis toward SLC and MIP-3β was evaluated. DCs migrating to the bottom chambers were recovered and analyzed by flow cytometry for CD11c expression. No significant difference in CD11c expression was observed between untreated and TGF-β-treated DCs (data not shown). TGF-β treatment caused reduced migration of DC toward both SLC and MIP-3β, however, only migration toward MIP-3β was significantly decreased (P < 0.05; Fig. 3 ⇓ ).

Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

Effect of TGF-β on in vitro chemotaxis. DCs were added to the upper chamber of a transwell migration chamber and evaluated for chemotactic migration. Cells migrating to the bottom chamber were recovered and analyzed by flow cytometry for CD11c expression. The results are mean ± SE of three independent experiments. ∗ refers to statistical significance between groups (P < 0.05).

View this table:
  • View inline
  • View popup
Table 1

Effect of TGF-β on in vivo migration

Treatment of Established Tumors with DCs and Neutralizing TGF-β Antibody.

The impairment of critical DC functions, including antigen uptake and antigen presentation, tumor-specific T lymphocyte stimulation, and in vivo migration by TGF-β, suggested that inhibition of TGF-β production in tumor-bearing animals may improve the efficacy of DC vaccines. To test this possibility, TGF-β production was suppressed by transfer of an antisense TGF-β transgene into 4T1 cells (4T1-asT) as described previously (21) . Expression of the transgene resulted in >90% inhibition of TGF-β production (0.083 ± 0.003 ng/ml in 4T1-asT compared with 1.244 ± 0.188 ng/ml in mock-transduced cells (4T1-N). Treatment of 4T1-asT tumors with 2G7 mAb alone or DCs plus 2G7 mAb significantly inhibited (P < 0.05) tumor growth compared with 4T1-asT tumors treated with DCs alone (Fig. 4) ⇓ . Furthermore, complete tumor regression occurred in 40% (two of five) of 4T1-asT-tumor-bearing mice that were treated with DCs plus 2G7 mAb (Fig. 4) ⇓ . Similarly, mock-transduced (4T1-N) tumors responded significantly better (P < 0.05) to treatment with DCs plus 2G7 mAb as compared with treatment with DCs alone; however, tumor growth inhibition in this group was inferior to that observed in animals bearing 4T1-asT tumors. (Fig. 4) ⇓ . Tumor growth was not affected by treatment with isotype control (IgG) antibody (data not shown). To determine whether the mice that exhibited complete tumor regression had developed long-term immunity, they were rechallenged with parental 4T1 tumor cells. These mice failed to develop tumors (zero of two); however, all control naive mice challenged with tumor cells developed tumors (three of three). These data suggest that neutralization of TGF-β in mice bearing TGF-β-secreting tumors enhances the effectiveness of DC vaccines in treating established tumors.

Fig. 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 4.

Treatment of established tumors. BALB/c mice with established 4T1-N (mock transfected) and 4T1-asT (transfected with antisense TGF-β gene) tumors were vaccinated intratumorally on days 15, 20, and 25 with 1.5 × 106 tumor cell lysate-pulsed mature DCs alone or in combination with 100 μg i.t and 300 μg i.p of 2G7, TGF-β-neutralizing antibody. Control mice were treated with 2G7 antibody alone. Mice were monitored for tumor growth. Graph represents mean tumor volume ± SE of 5 mice. ∗ indicates significant (P < 0.05) difference as compared with mice treated with DC alone on day 40.

DISCUSSION

In this study, we evaluated the impact of TGF-β on in vivo migration and immunostimulatory activities of DCs. Our results demonstrate that TGF-β treatment diminishes the ability of DC to migrate to secondary lymphoid organs and to induce T-cell responses. Most importantly, suppression of tumor-derived TGF-β by antisense TGF-β gene transfer plus neutralization of secreted TGF-β with anti-TGF-β mAb significantly improved the antitumor activity of intratumorally injected tumor cell lysate-pulsed DCs. Previous studies have demonstrated that suppression of TGF-β by antisense gene transfer (13 , 21 , 26) or abrogation of TGF-β using neutralizing antibody (27 , 28) increases tumor immunogenicity, leading to tumor growth inhibition or rejection. To our knowledge, this is the first study to evaluate the impact of both approaches simultaneously on DCs in controlling established tumors. The finding that the antitumor effect was most evident when antisense TGF-β-expressing tumors were treated with DCs plus anti-TGF-β mAb directly implicates tumor-derived TGF-β in tumor progression. In our study, TGF-β within the tumor milieu could be promoting tumor growth by interfering with the antigen-presenting and effector functions of DCs at the tumor site, as well as preventing the emigration of injected DCs to DLNs to activate naïve tumor-specific T lymphocytes. The former possibility is supported by the recent findings by Kirk et al. (29) who suggested that migration of i.t. injected DCs to DLNs is not required for the induction of an antitumor response. Using SLC gene-modified DCs, they demonstrated comparable tumor growth inhibition in normal mice and lymphotoxin a−/− mice lacking peripheral lymph nodes (29) . The latter possibility is supported by the decreased chemotactic response of TGF-β-treated DCs to MIP-3β and SLC observed in our study. These chemokines produced in the lymph nodes recruit DCs via their interaction with the chemokine receptor, CCR7 (20 , 30) . A possible explanation is that CCR7 gene expression is inhibited in DC by TGF-β treatment as has been reported by others (19 , 20) .

As with previously published studies (29 , 31 , 32) , only a minute fraction (<1%) of s.c. injected DCs in our study migrated to DLNs. It is yet to be determined if these lymph node-infiltrating DCs represent a unique subpopulation capable of singularly stimulating the antitumor response or require the participation of endogenous DCs to achieve this goal. In the setting of DC vaccination to prevent or treat inaccessible micrometastases, it is desirable that adoptively transferred DCs are able to migrate to secondary lymphoid organs to stimulate naïve T lymphocytes. Thus this LN-infiltrating, ex vivo manipulated DC population needs to be more actively studied.

In summary, this study demonstrates the potential usefulness of a combined therapeutic approach to eliminate immunosuppressive tumor-derived factors to improve the effectiveness of DC-based vaccines.

Acknowledgments

We thank Vivian Mack for technical assistance and Barbara Carolus for flow cytometric analysis.

Footnotes

  • 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 Supported by NIH Grants 1 RO1 CA9411-01 and DAMD 170010128 and DAMD 17010126 from the Department of Defense/United States Army.

  • ↵2 Present address: Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY 10021.

  • ↵3 To whom requests for reprints should be addressed, at The University of Arizona, Department of Microbiology and Immunology, 1501 North Campbell Avenue, Tucson, AZ 85724. E-mail: akporiay{at}u.arizona.edu

  • ↵4 The abbreviations used are: DC, dendritic cell; DLN, draining lymph node; mAb, monoclonal antibody; FBS, fetal bovine serum; TGF-β, transforming growth factor β; TNF-α, tumor necrosis factor α; FACS, fluorescence-activated cell sorter; PE, r-phycoerythrin; OVA, ovalbumin; SLC, secondary lymphoid chemokine; MIP-3β, macrophage inflammatory protein 3β.

  • Received May 28, 2002.
  • Accepted February 18, 2003.
  • ©2003 American Association for Cancer Research.

References

  1. ↵
    Steinman R. M. The dendritic cell system and its role in immunogenicity. Ann. Rev. Immunol., 9: 271-296, 1991.
    OpenUrlCrossRefPubMed
  2. ↵
    Gabrilovich D. I., Nadaf S., Corak J., Berzofsky J. A., Carbone D. P. Dendritic cells in antitumor immune responses. II. Dendritic cells grown from bone marrow precursors, but not mature DC from tumor-bearing mice, are effective antigen carriers in the therapy of established tumors. Cell Immunol., 170: 111-119, 1996.
    OpenUrlCrossRefPubMed
  3. ↵
    Coveney E., Wheatley G. H., III, Lyerly H. K. Active immunization using dendritic cells mixed with tumor cells inhibits the growth of primary breast cancer. Surgery (St. Louis), 122: 228-234, 1997.
    OpenUrlCrossRefPubMed
  4. ↵
    DeMatos P., Abdel-Wahab Z., Vervaert C., Hester D., Seigler H. Pulsing of dendritic cells with cell lysates from either B16 melanoma or MCA-106 fibrosarcoma yields equally effective vaccines against B16 tumors in mice. J. Surg., 68: 79-91, 1998.
    OpenUrl
  5. ↵
    Fields R. C., Shimizu K., Mule J. J. Murine dendritic cells pulsed with whole tumor lysates mediate potent antitumor immune responses in vitro and in vivo. Proc. Natl. Acad. Sci. USA, 95: 9482-9487, 1998.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    Morse M. A., Lyerly H. K. Clinical applications of dendritic cell vaccines. Curr. Opin. Mol. Ther., 2: 20-28, 2000.
    OpenUrlPubMed
  7. ↵
    Marincola F. M., Jaffe E. M., Hicklin D. J., Ferrone S. Escape of human solid tumors from T-cell recognition: molecular mechanisms and functional significance. Adv. Immunol., 74: 181-273, 2000.
    OpenUrlPubMed
  8. ↵
    Chaux P., Moutet M., Faivre J., Martin F., Martin M. Inflammatory cells infiltrating human colorectal carcinomas express HLA class II but not B7–1 and B7–2 costimulatory molecules of the T-cell activation. Lab. Investig., 74: 975-983, 1996.
    OpenUrlPubMed
  9. ↵
    Gabrilovich D. I., Ciernik I. F., Carbone D. P. Dendritic cells in antitumor immune responses. I. Defective antigen presentation in tumor-bearing hosts. Cell. Immunol., 170: 101-110, 1996.
    OpenUrlCrossRefPubMed
  10. ↵
    Enk A. H., Jonuleit H., Saloga J., Knop J. Dendritic cells as mediators of tumor-induced tolerance in metastatic melanoma. Int. J. Cancer, 73: 309-316, 1997.
    OpenUrlCrossRefPubMed
  11. ↵
    Gabrilovich D. I., Corak J., Ciernik I. F., Kavanaugh D., Carbone D. P. Decreased antigen presentation by dendritic cells in patients with breast cancer. Clin. Cancer Res., 3: 483-490, 1997.
    OpenUrlAbstract
  12. ↵
    Esche C., Lokshin A., Shurin G. V., Gastman B. R., Rabinowich H., Watkins S. C., Lotze M. T., Shurin M. R. Tumor’s other immune targets: dendritic cells. J. Leukoc. Biol., 66: 336-344, 1999.
    OpenUrlAbstract
  13. ↵
    Stampfer M. R., Yaswen P., Alhadeff M., Hosoda J. TGF-β induction of extracellular matrix associated proteins in normal and transformed human mammary epithelial cells in culture is independent of growth effects. J. Cell. Physiol., 155: 210-221, 1993.
    OpenUrlCrossRefPubMed
  14. ↵
    Fischer J. R., Darjes H., Lahm H., Schindel M., Drings P., Krammer P. H. Constitutive secretion of bioactive transforming growth factor β1 by small cell lung cancer cell lines. Eur. J. Cancer, 30A: 2125-2129, 1994.
    OpenUrlCrossRef
  15. ↵
    Rodeck U., Bossler A., Graeven U., Fox F. E., Nowell P. C., Knabbe C., Kari C. Transforming growth factor β production and responsiveness in normal human melanocytes and melanoma cells. Cancer Res., 2: 575-581, 1994.
    OpenUrl
  16. ↵
    Park J. A., Kurt R. A., Schluter S. F., Hersh E. M., Akporiaye E. T. Expression of an antisense transforming growth factor β1 transgene reduces tumorigenicity of EMT6 mammary tumor cells. Cancer Gene Ther., 4: 42-50, 1997.
    OpenUrlPubMed
  17. ↵
    De Visser K., Kast W. M. Effects of TGF-β on the immune system: implications for cancer immunotherapy. Leukemia (Baltimore), 13: 1188-1199, 1999.
    OpenUrlCrossRefPubMed
  18. ↵
    Ogata M., Zhang Y., Wang Y., Itakura M., Zhang Y. Y., Harada A., Hashimoto S., Matsushima K. Chemotactic response toward chemokines and its regulation by transforming growth factor β1 of murine bone marrow hematopoietic progenitor cell-derived different subsets of dendritic cells. Blood, 93: 3225-3232, 1999.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Sato K., Kawasaki H., Nagayama H., Enomoto M., Morimoto C., Tadokoro K., Juji T., Takahashi T. A. TGF-β1 reciprocally controls chemotaxis of human peripheral blood monocyte-derived dendritic cells via chemokine receptors. J. Immunol., 164: 2285-2295, 2000.
    OpenUrlAbstract/FREE Full Text
  20. ↵
    Takayama T., Morelli A. E., Onai N., Hirao M., Matsushima K., Tahara H., Thomson A. W. Mammalian and viral IL-10 enhance C-C chemokine receptor 5 but down-regulate C-C chemokine receptor 7 expression by myeloid dendritic cells: impact on chemotactic responses and in vivo homing ability. J. Immunol., 166: 7136-7143, 2001.
    OpenUrlAbstract/FREE Full Text
  21. ↵
    Wu R. S., Kobie J. J., Besselsen D. G., Fong T. C., Mack V. D., McEarchern J. A., Akporiaye E. T. Comparative analysis of IFN-γ, B7.1 and antisense TGF-β gene transfer on the tumorigenicity of a poorly immunogenic metastatic mammary carcinoma. Cancer Immunol. Immunother., 50: 229-240, 2001.
    OpenUrlCrossRefPubMed
  22. ↵
    Lucas C., Bald L. N., Fendly B. M., Mora-Worms M., Figari I. S., Patzer E. J., Palladino M. A. The autocrine production of transforming growth factor-β1 during lymphocyte activation. A study with a monoclonal antibody-based ELISA. J. Immunol., 145: 1415-1422, 1990.
    OpenUrlAbstract
  23. ↵
    Labeur M. S., Roters B., Pers B., Mehling A., Luger T. A., Schwarz T., Grabbe S. Generation of tumor immunity by bone marrow-derived dendritic cells correlates with dendritic cell maturation stage. J. Immunol., 162: 168-175, 1999.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    Hirao M., Onai N., Hiroishi K., Watkins S. C., Matsushima K., Robbins P. D., Lotze M. T., Tahara H. CC chemokine receptor-7 on dendritic cells is induced after interaction with apoptotic tumor cells: critical role in migration from the tumor site to draining lymph nodes. Cancer Res., 60: 2209-2217, 2000.
    OpenUrlAbstract/FREE Full Text
  25. ↵
    Banchereau J., Steinman R. Dendritic cells and the control of immunity. Nature (Lond.), 392: 245-252, 1998.
    OpenUrlCrossRefPubMed
  26. ↵
    Fakhrai H., Dorigo O., Shawler D. L., Lin H., Mercola D., Black K. L., Royston I., Sobol R. E. Eradication of established intracranial rat gliomas by transforming growth factor β antisense gene therapy. Proc. Natl. Acad. Sci. USA, 93: 2909-2914, 1996.
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Arteaga C. L., Hurd S. D., Winnier A. R., Johnson M. D., Fendly B. M., Forbes J. T. Anti-transforming growth factor (TGF)-β antibodies inhibit breast cancer cell tumorigenicity and increase mouse spleen natural killer cell activity. J. Clin. Investig., 92: 2568-2576, 1993.
  28. ↵
    Wojtowicz-Praga S., Verma U. N., Wakefield L., Esteban J. M., Hartmann D., Mazumder A., Verma U. M. Modulation of B16 melanoma growth and metastasis by anti-transforming growth factor β antibody and interleukin-2. J. Immunother. Emphasis Tumor Immunol., 19: 169-175, 1996.
    OpenUrlPubMed
  29. ↵
    Kirk C. J., Hartogan-O’Conner C. D., Mule J. J. The dynamics of the T-cell antitumor response: chemokine-secreting dendritic cells can prime tumor-reactive T cells extranodally. Cancer Res., 61: 8794-8802, 2001.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    Rossi D., Zlotnik A. The biology of chemokines and their receptors. Annu. Rev. Immunol., 18: 217-242, 2000.
    OpenUrlCrossRefPubMed
  31. ↵
    Eggert A. A., Schreurs M. W., Boerman O. C., Oyen W. J., de Boer A. J., Punt C. J., Figdor C. G., Adema G. J. Biodistribution and vaccine efficiency of murine dendritic cells are dependent on the route of administration. Cancer Res., 59: 3340-3345, 1999.
    OpenUrlAbstract/FREE Full Text
  32. ↵
    Lappin M. B., Weiss J. M., Delattre V., Mai B., Dittmar H., Maier C., Manke K., Grabbe S., Martin S., Simon J. C. Analysis of mouse dendritic cell migration in vivo upon subcutaneous and intravenous injection. Immunology, 98: 181-188, 1999.
    OpenUrlCrossRefPubMed
View Abstract
PreviousNext
Back to top
Cancer Research: 63 (8)
April 2003
Volume 63, Issue 8
  • Table of Contents
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Transforming Growth Factor β Inhibits the Antigen-Presenting Functions and Antitumor Activity of Dendritic Cell Vaccines
(Your Name) has forwarded a page to you from Cancer Research
(Your Name) thought you would be interested in this article in Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Transforming Growth Factor β Inhibits the Antigen-Presenting Functions and Antitumor Activity of Dendritic Cell Vaccines
James J. Kobie, Rita S. Wu, Robert A. Kurt, Sunming Lou, Miranda K. Adelman, Luke J. Whitesell, Lalitha V. Ramanathapuram, Carlos L. Arteaga and Emmanuel T. Akporiaye
Cancer Res April 15 2003 (63) (8) 1860-1864;

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Transforming Growth Factor β Inhibits the Antigen-Presenting Functions and Antitumor Activity of Dendritic Cell Vaccines
James J. Kobie, Rita S. Wu, Robert A. Kurt, Sunming Lou, Miranda K. Adelman, Luke J. Whitesell, Lalitha V. Ramanathapuram, Carlos L. Arteaga and Emmanuel T. Akporiaye
Cancer Res April 15 2003 (63) (8) 1860-1864;
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • INTRODUCTION
    • MATERIALS AND METHODS
    • RESULTS
    • DISCUSSION
    • Acknowledgments
    • Footnotes
    • References
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • Abstract 6699: CUE-102 Immuno-STATs for selective targeting and expansion of WT1-specific T cells for the treatment of HLA-A02+and/or HLA-A24+patients with WT1+ malignancies
  • Abstract 6606: Develop dual-targeted CAR-T cells to achieve RCC cures
  • Abstract 6647: Sensitizing microsatellite stable colorectal cancer to immune checkpoint therapy utilizing Wnt pathway inhibition
Show more Immunology
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Cancer Research Online ISSN: 1538-7445
Cancer Research Print ISSN: 0008-5472
Journal of Cancer Research ISSN: 0099-7013
American Journal of Cancer ISSN: 0099-7374

Advertisement