Invariant natural killer T cells regulate breast cancer response to radiation and CTLA-4 blockade

Karsten A Pilones, Noriko Kawashima, Anne Marie Yang, James S Babb, Silvia C Formenti, Sandra Demaria, Karsten A Pilones, Noriko Kawashima, Anne Marie Yang, James S Babb, Silvia C Formenti, Sandra Demaria

Abstract

Purpose: Immunoregulatory and suppressive mechanisms represent major obstacles to the success of immunotherapy in cancer patients. We have shown that the combination of radiotherapy to the primary tumor and CTL-associated protein 4 (CTLA-4) blockade induces antitumor immunity, inhibiting metastases and extending the survival of mice bearing the poorly immunogenic and highly metastatic 4T1 mammary carcinoma. Similarly to patients with metastatic cancer, however, mice were seldom cured. Here we tested the hypothesis that invariant natural killer T (iNKT) cells, a subset with unique regulatory functions, can regulate the response to radiotherapy and CTLA-4 blockade.

Experimental design: The growth of 4T1 primary tumors and lung metastases was compared in wild-type and iNKT cell-deficient (iNKT-/-) mice. Treatment was started on day 13 when the primary tumors were palpable. Mice received radiotherapy to the primary tumor in two doses of 12 Gy in combination or not with 9H10 monoclonal antibody against CTLA-4. Response to treatment was assessed by measuring primary tumor growth delay/regression, survival, and number of lung metastases.

Results: The response to radiotherapy plus 9H10 was markedly enhanced in the absence of iNKT cells, with 50% of iNKT-/- versus 0% of wild-type mice showing complete tumor regression, long-term survival, and resistance to a challenge with 4T1 cells. Administration of the iNKT cell activator alpha-galactosylceramide did not enhance the response of wild-type mice to radiotherapy plus 9H10. Tumor-infiltrating iNKT cells were markedly reduced in wild-type mice treated with radiotherapy plus 9H10.

Conclusions: iNKT cells play a major role in regulating the response to treatment with local radiotherapy and CTLA-4 blockade.

Figures

Figure 1. CD8 T cell-mediated inhibition of…
Figure 1. CD8 T cell-mediated inhibition of metastases in untreated tumor-bearing mice lacking iNKT cells
(A) Mice received 5×104 4T1 cells s.c. on Day 0. The number of metastatic cells in the lungs at Day 34 was significantly lower for the iNKT−/− than for the WT mice (p=0.0009). Each symbol represents one animal. (B, C) mAb-mediated CD4 and CD8 T-cell depletion was started on Day −1, and maintained up to the day of sacrifice. Mice were sacrificed on Day 33 to determine the tumor weigh (B) and number of metastatic cells in the lungs (C). Bars represent the mean ± SE of 8 to 10 animals/group. (B) There was no significant tumor weight difference (p>0.6) between WT and iNKT−/− mice non-depleted or depleted of CD8 or CD4 T cells. However, tumor weight was significantly increased in mice depleted of both T cell subsets (p<0.01 when compared to all other groups). (C) The number of metastases in iNKT−/− mice depleted of CD8 T cells (CD8) or CD4 and CD8 T cells (CD4+CD8) was significantly higher than in non-depleted iNKT−/− mice (p=0.004 and 0.001, respectively). In contrast, CD4 depletion did not significantly increase the number of metastases (p=0.94). Whereas there was a significant difference between WT and non-depleted iNKT−/− mice in the number of metastases (p=0.043), no significant difference was seen between WT and iNKT−/− mice after CD8 T cell depletion by itself or in combination with CD4 T cell depletion. (D) 4T1 cells are not intrinsically immunogenic in iNKT−/− mice. WT and iNKT−/− mice (N=5/group) were vaccinated weekly for 3 weeks with 106 irradiated (100 Gy) 4T1 cells s.c. in the left flank (+) or mock vaccinated with DMEM (−). Seven days after the last vaccination mice were challenged with 5×104 viable 4T1 cells and followed for tumor development. All mice developed tumors and there was significant difference in tumor weight in vaccinated WT (p=0.94) or iNKT (p=0.62) mice on Day 26.
Figure 2. Therapeutic response of iNKT−/− mice…
Figure 2. Therapeutic response of iNKT−/− mice with established 4T1 carcinoma to treatment with local RT and CTLA-4 blockade
Treatment was started on Day 13 post-s.c. inoculation with 4T1 cells. RT was delivered in two fractions of 12 Gy to the s.c. tumors on Day 13 and 14. Ab were given i.p. 1, 4 and 7 days post-RT. (A) Tumor growth delay in iNKT−/− mice treated with control hamster IgG (IgG) (open traingles, N=8), 9H10 (closed triangles, N=9), RT + IgG (open circles, N=9), or RT + 9H10 (closed circles, N=9). Tumor volume is shown as the mean ± SE for animals with tumors in each treatment group up to Day 32 when all animals were alive. The number of mice with complete tumor regression over the total number of mice per group is indicated. Tumor volume differences between treated and control (IgG) mice were statistically significant (p

Figure 3. Comparison between WT and iNKT−/−…

Figure 3. Comparison between WT and iNKT−/− mice with established 4T1 carcinoma in the response…

Figure 3. Comparison between WT and iNKT−/− mice with established 4T1 carcinoma in the response to treatment with local RT and CTLA-4 blockade
Comparison of response to treatment with RT and 9H10 (solid lines) or IgG (broken lines) in WT (black) and iNKT−/− (red) 4T1-tumor bearing mice. Treatment was started on Day 13 post-s.c. inoculation in the flank. RT was delivered in two fractions of 12 Gy to the s.c. tumors on Day 13 and 14. Ab were given i.p. 1, 4 and 7 days post-RT. (A) Tumor volume is shown as the mean ± SE for animals with tumors in each treatment group up to Day 29 when all animals were alive. The number of mice with complete tumor regression by Day 29 over the total number of mice per group is indicated. Two additional iNKT−/− mice had complete tumor regression after Day 29. Primary tumor growth was not significantly different in WT and iNKT−/− mice receiving the control IgG (p>0.05) at all time points. In contrast, iNKT−/− mice receiving RT+9H10 had a significantly lower tumor volume than WT mice receiving RT+9H10 (p

Figure 4. Administration of αGC does not…

Figure 4. Administration of αGC does not enhance the response of 4T1 tumor-bearing WT mice…

Figure 4. Administration of αGC does not enhance the response of 4T1 tumor-bearing WT mice to local RT and CTLA-4 blockade
Treatment was started on Day 13 post-s.c. inoculation in the flank. RT was delivered in two fractions of 12 Gy to the s.c. tumors on Day 13 and 14. 9H10 was given i.p. 1, 4 and 7 days post-RT. αGC (100 ng) was given i.p. twice per week starting on Day 1 post-RT. WT mice (N=5/group) were treated with (A) vehicle (closed circles), 9H10 + vehicle (closed diamonds), RT + vehicle (closed squares), RT + 9H10 + vehicle (closed triangles), (B) αGC (open circles), 9H10 + αGC (open diamonds), RT + αGC (open squares), or RT + 9H10 + αGC (open triangles). Tumor volume is shown as the mean ± SEM for animals with tumors in each treatment group up to Day 27 when all animals were alive. RT caused a significant (p

Figure 5. iNKT cells infiltrating 4T1 tumors…

Figure 5. iNKT cells infiltrating 4T1 tumors are reduced by treatment with local RT and…

Figure 5. iNKT cells infiltrating 4T1 tumors are reduced by treatment with local RT and CTLA-4 blockade
WT mice were injected with 4T1 cells on Day 0 and left untreated (None) or treated with local RT and 9H10 (RT+9H10) as described in the legend of Figure 3. Tumors and lungs were harvested on Day 29, and obtained single cell suspensions stained FITC-anti-CD3 mAb and CD1d-PBS-57 PE or control CD1d-Vehicle PE tetramers to identify iNKT cells, followed by flow cytometry analysis. The lymphocyte gate was set based on the scattered plots in the spleen. (A) Histograms showing tumor-derived cells from untreated mice stained with CD3 and CD1d-PBS-57 or control tetramers, as indicated, and gated on CD3+ cells. Numbers indicate the percentage of cells in the square gate. (B) The percentage of cells in the lymphocyte gate positive for CD3 and CD1d-PBS-57 was multiplied for the percentage of cells in the lymphocyte gate and for the total number of viable cells isolated from the tumors, and divided for the tumor weight to obtain the number of cells per mg of tumor in treated (RT+9H10, white bar) and untreated (None, black bar) mice. (C) Percentage of CD3+ T cells that bind the CD1d-PBS-57 tetramers in tumor and lungs of treated (RT+9H10, white bars) and untreated (None, black bars) mice, as indicated. All data are from 4 to 5 mice of each group. Errors bars are absent because pooling of tumors and lungs within each group was necessary to obtain sufficient cells for analysis.

Figure 6. Accumulation of MDSC and TGFβ…

Figure 6. Accumulation of MDSC and TGFβ production in WT and iNKT−/− mice bearing 4T1…

Figure 6. Accumulation of MDSC and TGFβ production in WT and iNKT−/− mice bearing 4T1 tumors
WT and NKT−/− mice were inoculated s.c. with 5×104 4T1 cells on Day 0 (N=5/group). 3 non-tumor-bearing mice of each genotype served as controls. On Day 14 primary tumors and spleens were harvested and obtained cell suspensions were stained with PE-anti-CD11b and Cy-anti-Gr-1 mAbs to detect MDSC. (A) Representative histograms of spleen from one healthy (top row) and one tumor-bearing (middle row) mouse showing the CD11b+Gr-1+ MDSC. Histograms from pooled tumors isolated from 5 WT and 5 iNKT−/− mice showing the CD11b+Gr-1+ MDSC (bottom row). Numbers indicate the percentage of MDSC. (B) Percentage of spleen MDSC in relation to the tumor weight in WT (closed triangles) and iNKT−/− (open circles) mice. Each symbol represents an individual mouse. For control healthy mice, one symbol representing the mean of 3 animals is shown. (C) Splenocytes isolated from tumor-free (−) (N=3) and tumor-bearing (+) (N=5) mice were cultured o.n. and the concentration of TGFβ in the culture supernatants was determined by ELISA. Data are the mean ± SE. In both WT and iNKT−/− mice spleen cells derived from tumor-bearing mice produced significantly more TGFβ than spleen cells derived from healthy mice (p<0.05). Although the baseline production of TGFβ did not differ significantly between WT and iNKT−/− healthy mice (p=0.35), tumor-bearing iNKT−/− mice produced more TGFβ than tumor-bearing WT mice (p<0.01).
Similar articles
Cited by
References
    1. Rosenberg SA. Progress in human tumour immunology and immunotherapy. Nature. 2001;411:380–384. - PubMed
    1. Herber DL, Nagaraj S, Djeu JY, Gabrilovich DI. Mechanism and therapeutic reversal of immune suppression in cancer. Cancer Res. 2007;67:5067–5069. - PMC - PubMed
    1. Finn OJ. Cancer vaccines: between the idea and the reality. Nat Rev Immunol. 2003;3:630–641. - PubMed
    1. Gabrilovich DI, Bronte V, Chen SH, et al. The terminology issue for myeloid-derived suppressor cells. Cancer Res. 2007;67:425. - PMC - PubMed
    1. Salvadori S, Martinelli G, Zier K. Resection of solid tumors reverses T cell defects and restores protective immunity. J Immunol. 2000;164:2214–2220. - PubMed
Show all 50 references
Publication types
MeSH terms
Related information
[x]
Cite
Copy Download .nbib .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 3. Comparison between WT and iNKT−/−…
Figure 3. Comparison between WT and iNKT−/− mice with established 4T1 carcinoma in the response to treatment with local RT and CTLA-4 blockade
Comparison of response to treatment with RT and 9H10 (solid lines) or IgG (broken lines) in WT (black) and iNKT−/− (red) 4T1-tumor bearing mice. Treatment was started on Day 13 post-s.c. inoculation in the flank. RT was delivered in two fractions of 12 Gy to the s.c. tumors on Day 13 and 14. Ab were given i.p. 1, 4 and 7 days post-RT. (A) Tumor volume is shown as the mean ± SE for animals with tumors in each treatment group up to Day 29 when all animals were alive. The number of mice with complete tumor regression by Day 29 over the total number of mice per group is indicated. Two additional iNKT−/− mice had complete tumor regression after Day 29. Primary tumor growth was not significantly different in WT and iNKT−/− mice receiving the control IgG (p>0.05) at all time points. In contrast, iNKT−/− mice receiving RT+9H10 had a significantly lower tumor volume than WT mice receiving RT+9H10 (p

Figure 4. Administration of αGC does not…

Figure 4. Administration of αGC does not enhance the response of 4T1 tumor-bearing WT mice…

Figure 4. Administration of αGC does not enhance the response of 4T1 tumor-bearing WT mice to local RT and CTLA-4 blockade
Treatment was started on Day 13 post-s.c. inoculation in the flank. RT was delivered in two fractions of 12 Gy to the s.c. tumors on Day 13 and 14. 9H10 was given i.p. 1, 4 and 7 days post-RT. αGC (100 ng) was given i.p. twice per week starting on Day 1 post-RT. WT mice (N=5/group) were treated with (A) vehicle (closed circles), 9H10 + vehicle (closed diamonds), RT + vehicle (closed squares), RT + 9H10 + vehicle (closed triangles), (B) αGC (open circles), 9H10 + αGC (open diamonds), RT + αGC (open squares), or RT + 9H10 + αGC (open triangles). Tumor volume is shown as the mean ± SEM for animals with tumors in each treatment group up to Day 27 when all animals were alive. RT caused a significant (p

Figure 5. iNKT cells infiltrating 4T1 tumors…

Figure 5. iNKT cells infiltrating 4T1 tumors are reduced by treatment with local RT and…

Figure 5. iNKT cells infiltrating 4T1 tumors are reduced by treatment with local RT and CTLA-4 blockade
WT mice were injected with 4T1 cells on Day 0 and left untreated (None) or treated with local RT and 9H10 (RT+9H10) as described in the legend of Figure 3. Tumors and lungs were harvested on Day 29, and obtained single cell suspensions stained FITC-anti-CD3 mAb and CD1d-PBS-57 PE or control CD1d-Vehicle PE tetramers to identify iNKT cells, followed by flow cytometry analysis. The lymphocyte gate was set based on the scattered plots in the spleen. (A) Histograms showing tumor-derived cells from untreated mice stained with CD3 and CD1d-PBS-57 or control tetramers, as indicated, and gated on CD3+ cells. Numbers indicate the percentage of cells in the square gate. (B) The percentage of cells in the lymphocyte gate positive for CD3 and CD1d-PBS-57 was multiplied for the percentage of cells in the lymphocyte gate and for the total number of viable cells isolated from the tumors, and divided for the tumor weight to obtain the number of cells per mg of tumor in treated (RT+9H10, white bar) and untreated (None, black bar) mice. (C) Percentage of CD3+ T cells that bind the CD1d-PBS-57 tetramers in tumor and lungs of treated (RT+9H10, white bars) and untreated (None, black bars) mice, as indicated. All data are from 4 to 5 mice of each group. Errors bars are absent because pooling of tumors and lungs within each group was necessary to obtain sufficient cells for analysis.

Figure 6. Accumulation of MDSC and TGFβ…

Figure 6. Accumulation of MDSC and TGFβ production in WT and iNKT−/− mice bearing 4T1…

Figure 6. Accumulation of MDSC and TGFβ production in WT and iNKT−/− mice bearing 4T1 tumors
WT and NKT−/− mice were inoculated s.c. with 5×104 4T1 cells on Day 0 (N=5/group). 3 non-tumor-bearing mice of each genotype served as controls. On Day 14 primary tumors and spleens were harvested and obtained cell suspensions were stained with PE-anti-CD11b and Cy-anti-Gr-1 mAbs to detect MDSC. (A) Representative histograms of spleen from one healthy (top row) and one tumor-bearing (middle row) mouse showing the CD11b+Gr-1+ MDSC. Histograms from pooled tumors isolated from 5 WT and 5 iNKT−/− mice showing the CD11b+Gr-1+ MDSC (bottom row). Numbers indicate the percentage of MDSC. (B) Percentage of spleen MDSC in relation to the tumor weight in WT (closed triangles) and iNKT−/− (open circles) mice. Each symbol represents an individual mouse. For control healthy mice, one symbol representing the mean of 3 animals is shown. (C) Splenocytes isolated from tumor-free (−) (N=3) and tumor-bearing (+) (N=5) mice were cultured o.n. and the concentration of TGFβ in the culture supernatants was determined by ELISA. Data are the mean ± SE. In both WT and iNKT−/− mice spleen cells derived from tumor-bearing mice produced significantly more TGFβ than spleen cells derived from healthy mice (p<0.05). Although the baseline production of TGFβ did not differ significantly between WT and iNKT−/− healthy mice (p=0.35), tumor-bearing iNKT−/− mice produced more TGFβ than tumor-bearing WT mice (p<0.01).
Similar articles
Cited by
References
    1. Rosenberg SA. Progress in human tumour immunology and immunotherapy. Nature. 2001;411:380–384. - PubMed
    1. Herber DL, Nagaraj S, Djeu JY, Gabrilovich DI. Mechanism and therapeutic reversal of immune suppression in cancer. Cancer Res. 2007;67:5067–5069. - PMC - PubMed
    1. Finn OJ. Cancer vaccines: between the idea and the reality. Nat Rev Immunol. 2003;3:630–641. - PubMed
    1. Gabrilovich DI, Bronte V, Chen SH, et al. The terminology issue for myeloid-derived suppressor cells. Cancer Res. 2007;67:425. - PMC - PubMed
    1. Salvadori S, Martinelli G, Zier K. Resection of solid tumors reverses T cell defects and restores protective immunity. J Immunol. 2000;164:2214–2220. - PubMed
Show all 50 references
Publication types
MeSH terms
Related information
[x]
Cite
Copy Download .nbib .nbib
Format: AMA APA MLA NLM
Figure 4. Administration of αGC does not…
Figure 4. Administration of αGC does not enhance the response of 4T1 tumor-bearing WT mice to local RT and CTLA-4 blockade
Treatment was started on Day 13 post-s.c. inoculation in the flank. RT was delivered in two fractions of 12 Gy to the s.c. tumors on Day 13 and 14. 9H10 was given i.p. 1, 4 and 7 days post-RT. αGC (100 ng) was given i.p. twice per week starting on Day 1 post-RT. WT mice (N=5/group) were treated with (A) vehicle (closed circles), 9H10 + vehicle (closed diamonds), RT + vehicle (closed squares), RT + 9H10 + vehicle (closed triangles), (B) αGC (open circles), 9H10 + αGC (open diamonds), RT + αGC (open squares), or RT + 9H10 + αGC (open triangles). Tumor volume is shown as the mean ± SEM for animals with tumors in each treatment group up to Day 27 when all animals were alive. RT caused a significant (p

Figure 5. iNKT cells infiltrating 4T1 tumors…

Figure 5. iNKT cells infiltrating 4T1 tumors are reduced by treatment with local RT and…

Figure 5. iNKT cells infiltrating 4T1 tumors are reduced by treatment with local RT and CTLA-4 blockade
WT mice were injected with 4T1 cells on Day 0 and left untreated (None) or treated with local RT and 9H10 (RT+9H10) as described in the legend of Figure 3. Tumors and lungs were harvested on Day 29, and obtained single cell suspensions stained FITC-anti-CD3 mAb and CD1d-PBS-57 PE or control CD1d-Vehicle PE tetramers to identify iNKT cells, followed by flow cytometry analysis. The lymphocyte gate was set based on the scattered plots in the spleen. (A) Histograms showing tumor-derived cells from untreated mice stained with CD3 and CD1d-PBS-57 or control tetramers, as indicated, and gated on CD3+ cells. Numbers indicate the percentage of cells in the square gate. (B) The percentage of cells in the lymphocyte gate positive for CD3 and CD1d-PBS-57 was multiplied for the percentage of cells in the lymphocyte gate and for the total number of viable cells isolated from the tumors, and divided for the tumor weight to obtain the number of cells per mg of tumor in treated (RT+9H10, white bar) and untreated (None, black bar) mice. (C) Percentage of CD3+ T cells that bind the CD1d-PBS-57 tetramers in tumor and lungs of treated (RT+9H10, white bars) and untreated (None, black bars) mice, as indicated. All data are from 4 to 5 mice of each group. Errors bars are absent because pooling of tumors and lungs within each group was necessary to obtain sufficient cells for analysis.

Figure 6. Accumulation of MDSC and TGFβ…

Figure 6. Accumulation of MDSC and TGFβ production in WT and iNKT−/− mice bearing 4T1…

Figure 6. Accumulation of MDSC and TGFβ production in WT and iNKT−/− mice bearing 4T1 tumors
WT and NKT−/− mice were inoculated s.c. with 5×104 4T1 cells on Day 0 (N=5/group). 3 non-tumor-bearing mice of each genotype served as controls. On Day 14 primary tumors and spleens were harvested and obtained cell suspensions were stained with PE-anti-CD11b and Cy-anti-Gr-1 mAbs to detect MDSC. (A) Representative histograms of spleen from one healthy (top row) and one tumor-bearing (middle row) mouse showing the CD11b+Gr-1+ MDSC. Histograms from pooled tumors isolated from 5 WT and 5 iNKT−/− mice showing the CD11b+Gr-1+ MDSC (bottom row). Numbers indicate the percentage of MDSC. (B) Percentage of spleen MDSC in relation to the tumor weight in WT (closed triangles) and iNKT−/− (open circles) mice. Each symbol represents an individual mouse. For control healthy mice, one symbol representing the mean of 3 animals is shown. (C) Splenocytes isolated from tumor-free (−) (N=3) and tumor-bearing (+) (N=5) mice were cultured o.n. and the concentration of TGFβ in the culture supernatants was determined by ELISA. Data are the mean ± SE. In both WT and iNKT−/− mice spleen cells derived from tumor-bearing mice produced significantly more TGFβ than spleen cells derived from healthy mice (p<0.05). Although the baseline production of TGFβ did not differ significantly between WT and iNKT−/− healthy mice (p=0.35), tumor-bearing iNKT−/− mice produced more TGFβ than tumor-bearing WT mice (p<0.01).
Figure 5. iNKT cells infiltrating 4T1 tumors…
Figure 5. iNKT cells infiltrating 4T1 tumors are reduced by treatment with local RT and CTLA-4 blockade
WT mice were injected with 4T1 cells on Day 0 and left untreated (None) or treated with local RT and 9H10 (RT+9H10) as described in the legend of Figure 3. Tumors and lungs were harvested on Day 29, and obtained single cell suspensions stained FITC-anti-CD3 mAb and CD1d-PBS-57 PE or control CD1d-Vehicle PE tetramers to identify iNKT cells, followed by flow cytometry analysis. The lymphocyte gate was set based on the scattered plots in the spleen. (A) Histograms showing tumor-derived cells from untreated mice stained with CD3 and CD1d-PBS-57 or control tetramers, as indicated, and gated on CD3+ cells. Numbers indicate the percentage of cells in the square gate. (B) The percentage of cells in the lymphocyte gate positive for CD3 and CD1d-PBS-57 was multiplied for the percentage of cells in the lymphocyte gate and for the total number of viable cells isolated from the tumors, and divided for the tumor weight to obtain the number of cells per mg of tumor in treated (RT+9H10, white bar) and untreated (None, black bar) mice. (C) Percentage of CD3+ T cells that bind the CD1d-PBS-57 tetramers in tumor and lungs of treated (RT+9H10, white bars) and untreated (None, black bars) mice, as indicated. All data are from 4 to 5 mice of each group. Errors bars are absent because pooling of tumors and lungs within each group was necessary to obtain sufficient cells for analysis.
Figure 6. Accumulation of MDSC and TGFβ…
Figure 6. Accumulation of MDSC and TGFβ production in WT and iNKT−/− mice bearing 4T1 tumors
WT and NKT−/− mice were inoculated s.c. with 5×104 4T1 cells on Day 0 (N=5/group). 3 non-tumor-bearing mice of each genotype served as controls. On Day 14 primary tumors and spleens were harvested and obtained cell suspensions were stained with PE-anti-CD11b and Cy-anti-Gr-1 mAbs to detect MDSC. (A) Representative histograms of spleen from one healthy (top row) and one tumor-bearing (middle row) mouse showing the CD11b+Gr-1+ MDSC. Histograms from pooled tumors isolated from 5 WT and 5 iNKT−/− mice showing the CD11b+Gr-1+ MDSC (bottom row). Numbers indicate the percentage of MDSC. (B) Percentage of spleen MDSC in relation to the tumor weight in WT (closed triangles) and iNKT−/− (open circles) mice. Each symbol represents an individual mouse. For control healthy mice, one symbol representing the mean of 3 animals is shown. (C) Splenocytes isolated from tumor-free (−) (N=3) and tumor-bearing (+) (N=5) mice were cultured o.n. and the concentration of TGFβ in the culture supernatants was determined by ELISA. Data are the mean ± SE. In both WT and iNKT−/− mice spleen cells derived from tumor-bearing mice produced significantly more TGFβ than spleen cells derived from healthy mice (p<0.05). Although the baseline production of TGFβ did not differ significantly between WT and iNKT−/− healthy mice (p=0.35), tumor-bearing iNKT−/− mice produced more TGFβ than tumor-bearing WT mice (p<0.01).

References

    1. Rosenberg SA. Progress in human tumour immunology and immunotherapy. Nature. 2001;411:380–384.
    1. Herber DL, Nagaraj S, Djeu JY, Gabrilovich DI. Mechanism and therapeutic reversal of immune suppression in cancer. Cancer Res. 2007;67:5067–5069.
    1. Finn OJ. Cancer vaccines: between the idea and the reality. Nat Rev Immunol. 2003;3:630–641.
    1. Gabrilovich DI, Bronte V, Chen SH, et al. The terminology issue for myeloid-derived suppressor cells. Cancer Res. 2007;67:425.
    1. Salvadori S, Martinelli G, Zier K. Resection of solid tumors reverses T cell defects and restores protective immunity. J Immunol. 2000;164:2214–2220.
    1. Shimizu J, Yamazaki S, Sakaguchi S. Induction of tumor immunity by removing CD25+CD4+ T cells: a common basis between tumor immunity and autoimmunity. J Immunol. 1999;163:5211–5218.
    1. Bluestone JA, Abbas AK. Natural versus adaptive regulatory T cells. Nat Rev Immunol. 2003;3:253–257.
    1. Liu VC, Wong LY, Jang T, 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-b. J Immunol. 2007;178:2883–2892.
    1. Valzasina B, Piconese S, Guiducci C, Colombo MP. Tumor-induced expansion of regulatory T cells by conversion of CD4+CD25− lymphocytes is thymus and proliferation independent. Cancer Res. 2006;66:4488–4495.
    1. Godfrey DI, Kronenberg M. Going both ways: immune regulation via CD1d-dependent NKT cells. J Clin Invest. 2004;114:1379–1388.
    1. Van Kaer L. Alpha-Galactosylceramide therapy for autoimmune diseases: prospects and obstacles. Nat Rev Immunol. 2005;5:31–42.
    1. Palmer JL, Tulley JM, Kovacs EJ, Gamelli RL, Taniguchi M, Faunce DE. Injury-induced suppression of effector T cell immunity requires CD1d-positive APCs and CD1d-restricted NKT cells. J Immunol. 2006;177:92–99.
    1. Godfrey DI, MacDonald HR, Kronenberg M, Smyth MJ, Van Kaer L. NKT cells: what's in a name? Nat Rev Immunol. 2004;4:231–237.
    1. Smyth MJ, Thia KY, Street SE, et al. Differential tumor surveillance by natural killer (NK) and NKT cells. J Exp Med. 2000;191:661–668.
    1. Crowe NY, Smyth MJ, Godfrey DI. A critical role for natural killer T cells in immunosurveillance of methylcholanthrene-induced sarcomas. J Exp Med. 2002;196:119–127.
    1. Terabe M, Matsui S, Noben-Trauth N, et al. NKT cell-mediated repression of tumor immunosurveillance by IL-13 and the IL-4R-STAT6 pathway. Nat Immunol. 2000;1:515–520.
    1. Moodycliffe AM, Nghiem D, Clydesdale G, Ullrich SE. Immune suppression and skin cancer development: regulation by NKT cells. Nat Immunol. 2000;1:521–525.
    1. Terabe M, Swann J, Ambrosino E, et al. A nonclassical non-V〈14J〈18 CD1d-restricted (type II) NKT cell is sufficient for down-regulation of tumor immunosurveillance. J Exp Med. 2005;202:1627–1633.
    1. Terabe M, Matsui S, Park JM, et al. Transforming growth factor-β production and myeloid cells are an effector mechanism through which CD1d-restricted T cells block cytotoxic T lymphocyte-mediated tumor immunosurveillance: abrogation prevents tumor recurrence. J Exp Med. 2003;198:1741–1752.
    1. Park JM, Terabe M, van den Broeke LT, Donaldson DD, Berzofsky JA. Unmasking immunosurveillance against a syngeneic colon cancer by elimination of CD4+ NKT regulatory cells and IL-13. Int J Cancer. 2005;114:80–87.
    1. Ostrand-Rosenberg S, Clements VK, Terabe M, Park JM, Berzofsky JA, Dissanayake SK. Resistance to metastatic disease in STAT6-deficient mice requires hemopoietic and nonhemopoietic cells and is IFN-© dependent. J Immunol. 2002;169:5796–5804.
    1. Terabe M, Khanna C, Bose S, et al. CD1d-restricted natural killer T cells can down-regulate tumor immunosurveillance independent of interleukin-4 receptor-signal transducer and activator of transcription 6 or transforming growth factor-beta. Cancer Res. 2006;66:3869–3875.
    1. Hayakawa Y, Godfrey DI, Smyth MJ. Alpha-galactosylceramide: potential immunomodulatory activity and future application. Curr Med Chem. 2004;11:241–252.
    1. Swann JB, Coquet JM, Smyth MJ, Godfrey DI. CD1-restricted T cells and tumor immunity. Curr Top Microbiol Immunol. 2007;314:293–323.
    1. Demaria S, Bhardwaj N, McBride WH, Formenti SC. Combining radiotherapy and immunotherapy: a revived partnership. Int J Radiat Oncol Biol Phys. 2005;63:655–666.
    1. Demaria S, Formenti SC. Sensors of ionizing radiation effects on the immunological microenvironment of cancer. Int J Radiat Biol. 2007;83:819–825.
    1. Aslakson CJ, Miller FR. Selective events in the metastatic process defined by analysis of the sequential dissemination of subpopulations of a mouse mammary tumor. Cancer Res. 1992;52:1399–1405.
    1. Demaria S, Kawashima N, Yang AM, et al. Immune-mediated inhibition of metastases following treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin Cancer Res. 2005;11:728–734.
    1. Cui J, Shin T, Kawano T, et al. Requirement for Vα14 NKT cells in IL-12-mediated rejection of tumors. Science. 1997;278:1623–1626.
    1. Kim KJ, Kanellopoulos-Langevin C, Merwin RM, Sachs DH, Asofsky R. Establishment and characterization of BALB/c lymphoma lines with B cell properties. J Immunol. 1979;122:549–554.
    1. Liu Y, Goff RD, Zhou D, et al. A modified alpha-galactosyl ceramide for staining and stimulating natural killer T cells. J Immunol Methods. 2006;312:34–39.
    1. Matsumura S, Wang B, Kawashima N, et al. Radiation-induced CXCL16 release by breast cancer cells attracts effector T cells. J Immunol. 2008;181:3099–3107.
    1. Tulley JM, Palmer JL, Gamelli RL, Faunce DE. Prevention of injury-induced suppression of T-cell immunity by the CD1d/NKT cell-specific ligand alpha-galactosylceramide. Shock. 2008;29:269–277.
    1. Gallina G, Dolcetti L, Serafini P, et al. Tumors induce a subset of inflammatory monocytes with immunosuppressive activity on CD8+ T cells. J Clin Invest. 2006;116:2777–2790.
    1. Gorelik E. Concomitant tumor immunity and the resistance to a second tumor challenge. Adv Cancer Res. 1983;39:71–120.
    1. Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer immunoediting: from immunosurveillance to tumor escape. Nature Immunol. 2002;3:991–998.
    1. Turk MJ, Guevara-Patiño JA, Rizzuto GA, Engelhorn ME, Sakaguchi S, Houghton AN. Concomitant tumor immunity to a poorly immunogenic melanoma is prevented by regulatory T cells. J Exp Med. 2004;200:771–782.
    1. Matzinger P. Tolerance, danger, and the extended family. Annu Rev Immunol. 1994;12:991–1045.
    1. Frey AB, Monu N. Effector-phase tolerance: another mechanism of how cancer escapes antitumor immune response. J Leukoc Biol. 2006;79:652–662.
    1. Zhang B, Bowerman NA, Salama JK, et al. Induced sensitization of tumor stroma leads to eradication of established cancer by T cells. J Exp Med. 2007;204:49–55.
    1. Egen JG, Kuhns MS, Allison JP. CTLA-4: new insights into its biological function and use in tumor immunotherapy. Nat Immunol. 2002;3:611–618.
    1. Peggs KS, Quezada SA, Korman AJ, Allison JP. Principles and use of anti-CTLA4 antibody in human cancer immunotherapy. Curr Opin Immunol. 2006;18:206–213.
    1. Sinha P, Clements VK, Ostrand-Rosenberg S. Reduction of myeloid-derived suppressor cells and induction of M1 macrophages facilitate the rejection of established metastatic disease. J Immunol. 2005;174:636–645.
    1. Kusmartsev S, Gabrilovich DI. STAT1 signaling regulates tumor-associated macrophage-mediated T cell deletion. J Immunol. 2005;174:4880–4891.
    1. Teng MW, Westwood JA, Darcy PK, et al. Combined natural killer T-cell based immunotherapy eradicates established tumors in mice. Cancer Res. 2007;67:7495–7504.
    1. Wu DY, Segal NH, Sidobre S, Kronenberg M, Chapman PB. Cross-presentation of disialoganglioside GD3 to natural killer T cells. J Exp Med. 2003;198:173–181.
    1. Scott-Browne JP, Matsuda JL, Mallevaey T, et al. Germline-encoded recognition of diverse glycolipids by natural killer T cells. Nat Immunol. 2007;8:1105–1113.
    1. Shimizu K, Goto A, Fukui M, Taniguchi M, Fujii S. Tumor cells loaded with alpha-galactosylceramide induce innate NKT and NK cell-dependent resistance to tumor implantation in mice. J Immunol. 2007;178:2853–2861.
    1. Jensen SM, Meijer SL, Kurt RA, Urba WJ, Hu HM, Fox BA. Regression of a mammary adenocarcinoma in STAT6−/− mice is dependent on the presence of STAT6-reactive T cells. J Immunol. 2003;170:2014–2021.
    1. Speiser DE, Rimoldi D, Batard P, et al. Disease-driven T cell activation predicts immune responses to vaccination against melanoma. Cancer Immun. 2003;3:12.

Source: PubMed

3
Suscribir