Effects of conventional therapeutic interventions on the number and function of regulatory T cells

Mario Roselli, Vittore Cereda, Maria Giovanna di Bari, Vincenzo Formica, Antonella Spila, Caroline Jochems, Benedetto Farsaci, Renee Donahue, James L Gulley, Jeffrey Schlom, Fiorella Guadagni, Mario Roselli, Vittore Cereda, Maria Giovanna di Bari, Vincenzo Formica, Antonella Spila, Caroline Jochems, Benedetto Farsaci, Renee Donahue, James L Gulley, Jeffrey Schlom, Fiorella Guadagni

Abstract

Several lines of investigation have revealed the apparent interplay between the immune system of the host and many conventional, "standard-of-care" anticancer therapies, including chemotherapy and small molecule targeted therapeutics. In particular, preclinical and clinical studies have demonstrated the important role of regulatory T cells (Tregs) in inhibiting immune responses elicited by immunotherapeutic regimens such as those based on anticancer vaccines or checkpoint inhibitors. However, how the number and immunosuppressive function of Tregs change in cancer patients undergoing treatment with non-immune anticancer therapies remains to be precisely elucidated. To determine whether immunostimulatory therapies can be employed successfully in combination with conventional anticancer regimens, we have investigated both the number and function of Tregs obtained from the peripheral blood of carcinoma patients before the initiation and during the course of chemotherapeutic and targeted agent regimens. Our studies show that the treatment of breast cancer patients with tamoxifen plus leuprolide, a gonadotropin releasing hormone agonist, has minimal effects on Tregs, while sunitinib appears to exert differential effects on Tregs among patients with metastatic renal carcinoma. However, the administration of docetaxel to patients with metastatic prostate or breast cancer, as well as that of cisplatin plus vinorelbine to non-small cell lung cancer patients, appears to significantly increase the ratio between effector T cells and Tregs and to reduce the immunosuppressive activity of the latter in the majority of patients. These studies provide the rationale for the selective use of active immunotherapy regimens in combination with specific standard-of-care therapies to achieve the most beneficial clinical outcome among carcinoma patients.

Keywords: T lymphocytes; Tregs; carcinoma; chemotherapy; drug therapy.

Figures

https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3862634/bin/onci-2-e27025-g1.jpg
Figure 1. Circulating immune cell subsets in patients with prostate cancer or non-small cell lung cancer before and during the course of chemotherapy. (A–C) Patients (n = 15) with hormone refractory metastatic prostate cancer were treated with docetaxel and evaluated before and during therapy for the number of peripheral blood mononuclear cells (PBMCs) (A), the frequency of CD4+ T cells among PBMCs (B), and the frequency of regulatory T cells (Tregs) among CD4+ T cells (C). Peripheral blood samples were collected at the indicated time points. Statistical analyses were performed using Friedman and Dunn’s multiple comparison tests and revealed a significant decrease in the frequency of Tregs at post-cycle I (**P < 0.01) and pre-cycle II (***P < 0.001). (D–F) Patients (n = 14) with stage IB, II, and IIIA non-small cell lung cancer (NSCLC) were evaluated before and during therapy with cisplatin plus vinorelbine for the number of PBMCs (D), the frequency of CD4+ T cells among PBMCs (E), and the frequency of Tregs among CD4+ T cells (F). Peripheral blood samples were collected at the indicated time points. Statistical analyses were performed using Friedman and Dunn’s multiple comparison tests; * P < 0.05, *** P < 0.001. In all dot plots, the median and interquartile range are shown.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3862634/bin/onci-2-e27025-g2.jpg
Figure 2. Docetaxel-induced changes in the ratio of effector to regulatory T cells and in the immunosuppressive activity of the latter in hormone-refractory prostate cancer patients. (A) Waterfall plot of the change in the ratio between effector T cells and regulatory T cells (Teff:Treg ratio) in the course of docetaxel therapy in patients with hormone-refractory prostate cancer (n = 15). Patients were treated with docetaxel weekly for 3 weeks followed by 1 week of rest; this comprised a 4-week cycle of therapy. Peripheral blood samples were collected prior to therapy (baseline, day 0) and at the end of the week of rest before starting cycle II. (B) Waterfall plot of the change in suppressive activity of Tregs in the course of docetaxel therapy in patients with hormone-refractory prostate cancer (n = 11).
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3862634/bin/onci-2-e27025-g3.jpg
Figure 3. Docetaxel treatment of metastatic breast cancer patients increased the ratio of CD4+ or CD8+ T lymphocytes vs. regulatory T cells. (A–F) Cytofluorometric analysis of peripheral blood mononuclear cells (PBMCs) from patients (n = 8) with metastatic breast cancer treated with docetaxel. PBMCs were analyzed before treatment (baseline) and at the time of first re-staging near day 91 (at the second week of cycle III). (A) Percentage of CD4+ T lymphocytes among PBMCs. (B) Percentage of CD8+ T lymphocytes among PBMCs. (C) Percentage of CD4+CD25highCD127–FoxP3+ regulatory T cells (Tregs) among PBMCs. (D) Ratio of CD4+ effector T cells to Tregs (CD4+ Teff:Treg ratio). (E) Ratio of CD8+ effector T cells to Tregs (CD8+ Teff:Treg ratio). (F) Percentage of CD3–CD56+ natural killer (NK) cells among PBMCs. Medians at baseline and cycle III are shown. Statistical analyses were performed by Wilcoxon matched-pairs signed rank test and P < 0.05 was considered statistically significant.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3862634/bin/onci-2-e27025-g4.jpg
Figure 4. Cisplatin plus vinorelbine-induced changes in the ratio of effector T cells to regulatory T cells and in the immunosuppressive activity of the latter in non-small cell lung cancer patients. (A) Waterfall plot of the change in the ratio between CD4+ effector T cells and regulatory T cells (Teff:Treg ratio) in non-small cell lung cancer (NSCLC) patients before and during therapy with cisplatin plus vinorelbine (n = 14). Patients with NSCLC (stages IB, II, and IIIA) were treated in the adjuvant setting, post-surgery, with cisplatin plus vinorelbine at day 1 and vinorelbine at day 8. This was followed by 14 days of rest, which comprised the 3-week cycle of therapy. PBMC were collected from peripheral blood at baseline and post-cycle III. (B) Waterfall plot of the change in immunosuppressive activity of Tregs from NSCLC patients before and during therapy with cisplatin plus vinorelbine (n = 10). The Treg suppressive activity was evaluated baseline and post-cycle III.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3862634/bin/onci-2-e27025-g5.jpg
Figure 5. Circulating immune cell subsets in patients with breast cancer and metastatic renal cell carcinoma before and during therapy. (A–C) Premenopausal (< 50 y) breast cancer patients (n = 8) were treated upon surgery with tamoxifen plus a gonadotropin-releasing hormone (GnRH) agonist, and evaluated before and during therapy for the number of circulating peripheral blood mononuclear cells (PBMCs; A), the frequency of CD4+ T cells among PBMCs (B), and the frequency of regulatory T cells (Tregs) among CD4+ T cells (C). Peripheral blood samples were collected at the indicated time points. (D–F) Metastatic renal cell carcinoma patients on sunitinib monotherapy (n = 10) were evaluated before and during treatment for the frequency of CD4+ T cells among PBMCs (D), the frequency of Tregs among CD4+ T cells (E), and the absolute number of Tregs (F). Peripheral blood was collected at the indicated time points. Statistical analyses were performed using Friedman and Dunn’s multiple comparison test; * P < 0.05, ** P < 0.01, *** P < 0.001. In all dot plots, the median and interquartile ranges are shown.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3862634/bin/onci-2-e27025-g6.jpg
Figure 6. Effects of targeted anticancer therapeutics on the ratio between effector and regulatory T cells and on the immunosuppressive activity of the latter in patients with breast cancer or metastatic renal cell carcinoma. (A–B) Waterfall plots of the change in the ratio between CD4+ effector T cells and regulatory T cells (Teff:Treg ratio) (A), and in the immunosuppressive activity of Tregs (B), in breast cancer patients receiving tamoxifen plus a gonadotropin-releasing hormone (GnRH) agonist upon surgery (n = 8). Patients of premenopausal status were treated in the adjuvant setting post-surgery with tamoxifen daily and GnRH agonist every 3 months. Peripheral blood samples were collected prior to therapy (baseline, day 0) and at post 3 months. (C–D) Waterfall plots of the change in CD4+ Teff:Treg ratio (C), and Treg-mediated immunosuppression (D), in metastatic renal cell carcinoma patients receiving sunitinib monotherapy (n = 10). Patients with metastatic renal cell carcinoma were treated with sunitinib monotherapy daily for 4 weeks followed by 2 weeks of rest; this comprised a 6-week cycle of therapy. Peripheral blood was collected prior to therapy, and at the end of the second round of chemotherapy in cycle II.

References

    1. Fridman WH, Pagès F, Sautès-Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer. 2012;12:298–306. doi: 10.1038/nrc3245.
    1. Galluzzi L, Senovilla L, Zitvogel L, Kroemer G. The secret ally: immunostimulation by anticancer drugs. Nat Rev Drug Discov. 2012;11:215–33. doi: 10.1038/nrd3626.
    1. Galon J, Pagès F, Marincola FM, Thurin M, Trinchieri G, Fox BA, Gajewski TF, Ascierto PA. The immune score as a new possible approach for the classification of cancer. J Transl Med. 2012;10:1. doi: 10.1186/1479-5876-10-1.
    1. Jochems C, Schlom J. Tumor-infiltrating immune cells and prognosis: the potential link between conventional cancer therapy and immunity. Exp Biol Med (Maywood) 2011;236:567–79. doi: 10.1258/ebm.2011.011007.
    1. Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini JL, Castedo M, Mignot G, Panaretakis T, Casares N, et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med. 2007;13:54–61. doi: 10.1038/nm1523.
    1. Tesniere A, Schlemmer F, Boige V, Kepp O, Martins I, Ghiringhelli F, Aymeric L, Michaud M, Apetoh L, Barault L, et al. Immunogenic death of colon cancer cells treated with oxaliplatin. Oncogene. 2010;29:482–91. doi: 10.1038/onc.2009.356.
    1. Garnett CT, Palena C, Chakraborty M, Tsang KY, Schlom J, Hodge JW. Sublethal irradiation of human tumor cells modulates phenotype resulting in enhanced killing by cytotoxic T lymphocytes. Cancer Res. 2004;64:7985–94. doi: 10.1158/0008-5472.CAN-04-1525.
    1. Hodge JW, Ardiani A, Farsaci B, Kwilas AR, Gameiro SR. The tipping point for combination therapy: cancer vaccines with radiation, chemotherapy, or targeted small molecule inhibitors. Semin Oncol. 2012;39:323–39. doi: 10.1053/j.seminoncol.2012.02.006.
    1. Reits EA, Hodge JW, Herberts CA, Groothuis TA, Chakraborty M, Wansley EK, Camphausen K, Luiten RM, de Ru AH, Neijssen J, et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med. 2006;203:1259–71. doi: 10.1084/jem.20052494.
    1. Adotevi O, Pere H, Ravel P, Haicheur N, Badoual C, Merillon N, Medioni J, Peyrard S, Roncelin S, Verkarre V, et al. A decrease of regulatory T cells correlates with overall survival after sunitinib-based antiangiogenic therapy in metastatic renal cancer patients. J Immunother. 2010;33:991–8. doi: 10.1097/CJI.0b013e3181f4c208.
    1. Emens LA, Asquith JM, Leatherman JM, Kobrin BJ, Petrik S, Laiko M, Levi J, Daphtary MM, Biedrzycki B, Wolff AC, et al. Timed sequential treatment with cyclophosphamide, doxorubicin, and an allogeneic granulocyte-macrophage colony-stimulating factor-secreting breast tumor vaccine: a chemotherapy dose-ranging factorial study of safety and immune activation. J Clin Oncol. 2009;27:5911–8. doi: 10.1200/JCO.2009.23.3494.
    1. Finke JH, Rini B, Ireland J, Rayman P, Richmond A, Golshayan A, Wood L, Elson P, Garcia J, Dreicer R, et al. Sunitinib reverses type-1 immune suppression and decreases T-regulatory cells in renal cell carcinoma patients. Clin Cancer Res. 2008;14:6674–82. doi: 10.1158/1078-0432.CCR-07-5212.
    1. Ko JS, Rayman P, Ireland J, Swaidani S, Li G, Bunting KD, Rini B, Finke JH, Cohen PA. Direct and differential suppression of myeloid-derived suppressor cell subsets by sunitinib is compartmentally constrained. Cancer Res. 2010;70:3526–36. doi: 10.1158/0008-5472.CAN-09-3278.
    1. Ko JS, Zea AH, Rini BI, Ireland JL, Elson P, Cohen P, Golshayan A, Rayman PA, Wood L, Garcia J, et al. Sunitinib mediates reversal of myeloid-derived suppressor cell accumulation in renal cell carcinoma patients. Clin Cancer Res. 2009;15:2148–57. doi: 10.1158/1078-0432.CCR-08-1332.
    1. Vanneman M, Dranoff G. Combining immunotherapy and targeted therapies in cancer treatment. Nat Rev Cancer. 2012;12:237–51. doi: 10.1038/nrc3237.
    1. Schlom J. Therapeutic cancer vaccines: current status and moving forward. J Natl Cancer Inst. 2012;104:599–613. doi: 10.1093/jnci/djs033.
    1. Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, Redfern CH, Ferrari AC, Dreicer R, Sims RB, et al. IMPACT Study Investigators Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363:411–22. doi: 10.1056/NEJMoa1001294.
    1. von Mehren M, Arlen P, Gulley J, Rogatko A, Cooper HS, Meropol NJ, Alpaugh RK, Davey M, McLaughlin S, Beard MT, et al. The influence of granulocyte macrophage colony-stimulating factor and prior chemotherapy on the immunological response to a vaccine (ALVAC-CEA B7.1) in patients with metastatic carcinoma. Clin Cancer Res. 2001;7:1181–91.
    1. von Mehren M, Arlen P, Tsang KY, Rogatko A, Meropol N, Cooper HS, Davey M, McLaughlin S, Schlom J, Weiner LM. Pilot study of a dual gene recombinant avipox vaccine containing both carcinoembryonic antigen (CEA) and B7.1 transgenes in patients with recurrent CEA-expressing adenocarcinomas. Clin Cancer Res. 2000;6:2219–28.
    1. Gajewski TF, Meng Y, Blank C, Brown I, Kacha A, Kline J, Harlin H. Immune resistance orchestrated by the tumor microenvironment. Immunol Rev. 2006;213:131–45. doi: 10.1111/j.1600-065X.2006.00442.x.
    1. Gajewski TF, Meng Y, Harlin H. Immune suppression in the tumor microenvironment. J Immunother. 2006;29:233–40. doi: 10.1097/01.cji.0000199193.29048.56.
    1. Zitvogel L, Kepp O, Galluzzi L, Kroemer G. Inflammasomes in carcinogenesis and anticancer immune responses. Nat Immunol. 2012;13:343–51. doi: 10.1038/ni.2224.
    1. deLeeuw RJ, Kost SE, Kakal JA, Nelson BH. The prognostic value of FoxP3+ tumor-infiltrating lymphocytes in cancer: a critical review of the literature. Clin Cancer Res. 2012;18:3022–9. doi: 10.1158/1078-0432.CCR-11-3216.
    1. Ménétrier-Caux C, Curiel T, Faget J, Manuel M, Caux C, Zou W. Targeting regulatory T cells. Target Oncol 2012; 7:15-28;; 10.1007/s11523-012-0208-y
    1. Cesana GC, DeRaffele G, Cohen S, Moroziewicz D, Mitcham J, Stoutenburg J, Cheung K, Hesdorffer C, Kim-Schulze S, Kaufman HL. Characterization of CD4+CD25+ regulatory T cells in patients treated with high-dose interleukin-2 for metastatic melanoma or renal cell carcinoma. J Clin Oncol. 2006;24:1169–77. doi: 10.1200/JCO.2005.03.6830.
    1. Condamine T, Gabrilovich DI. Molecular mechanisms regulating myeloid-derived suppressor cell differentiation and function. Trends Immunol. 2011;32:19–25. doi: 10.1016/j.it.2010.10.002.
    1. Gajewski TF. Failure at the effector phase: immune barriers at the level of the melanoma tumor microenvironment. Clin Cancer Res. 2007;13:5256–61. doi: 10.1158/1078-0432.CCR-07-0892.
    1. Lindenberg JJ, Fehres CM, van Cruijsen H, Oosterhoff D, de Gruijl TD. Cross-talk between tumor and myeloid cells: how to tip the balance in favor of antitumor immunity. Immunotherapy 2011; 3:77-96;; 10.2217/imt.10.95
    1. Lutsiak ME, Tagaya Y, Adams AJ, Schlom J, Sabzevari H. Tumor-induced impairment of TCR signaling results in compromised functionality of tumor-infiltrating regulatory T cells. J Immunol. 2008;180:5871–81.
    1. Mocellin S, Wang E, Marincola FM. Cytokines and immune response in the tumor microenvironment. J Immunother. 2001;24:392–407. doi: 10.1097/00002371-200109000-00002.
    1. Nagaraj S, Gupta K, Pisarev V, Kinarsky L, Sherman S, Kang L, Herber DL, Schneck J, Gabrilovich DI. Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer. Nat Med. 2007;13:828–35. doi: 10.1038/nm1609.
    1. Nagaraj S, Schrum AG, Cho HI, Celis E, Gabrilovich DI. Mechanism of T cell tolerance induced by myeloid-derived suppressor cells. J Immunol. 2010;184:3106–16. doi: 10.4049/jimmunol.0902661.
    1. Shevach EM. The resurrection of T cell-mediated suppression. J Immunol. 2011;186:3805–7. doi: 10.4049/jimmunol.1100364.
    1. Vergati M, Cereda V, Madan RA, Gulley JL, Huen NY, Rogers CJ, Hance KW, Arlen PM, Schlom J, Tsang KY. Analysis of circulating regulatory T cells in patients with metastatic prostate cancer pre- versus post-vaccination. Cancer Immunol Immunother. 2011;60:197–206. doi: 10.1007/s00262-010-0927-9.
    1. Gulley JL, Arlen PM, Madan RA, Tsang KY, Pazdur MP, Skarupa L, Jones JL, Poole DJ, Higgins JP, Hodge JW, et al. Immunologic and prognostic factors associated with overall survival employing a poxviral-based PSA vaccine in metastatic castrate-resistant prostate cancer. Cancer Immunol Immunother. 2010;59:663–74. doi: 10.1007/s00262-009-0782-8.
    1. Halabi S, Small EJ, Kantoff PW, Kattan MW, Kaplan EB, Dawson NA, Levine EG, Blumenstein BA, Vogelzang NJ. Prognostic model for predicting survival in men with hormone-refractory metastatic prostate cancer. J Clin Oncol. 2003;21:1232–7. doi: 10.1200/JCO.2003.06.100.
    1. Farsaci B, Higgins JP, Hodge JW. Consequence of dose scheduling of sunitinib on host immune response elements and vaccine combination therapy. Int J Cancer. 2012;130:1948–59. doi: 10.1002/ijc.26219.
    1. Huen NY, Pang AL, Tucker JA, Lee TL, Vergati M, Jochems C, Intrivici C, Cereda V, Chan WY, Rennert OM, et al. Up-regulation of proliferative and migratory genes in regulatory T cells from patients with metastatic castration-resistant prostate cancer. Int J Cancer. 2013;133:373–82. doi: 10.1002/ijc.28026.
    1. Gameiro SR, Caballero JA, Higgins JP, Apelian D, Hodge JW. Exploitation of differential homeostatic proliferation of T-cell subsets following chemotherapy to enhance the efficacy of vaccine-mediated antitumor responses. Cancer Immunol Immunother. 2011;60:1227–42. doi: 10.1007/s00262-011-1020-8.
    1. Ito T, Hanabuchi S, Wang YH, Park WR, Arima K, Bover L, Qin FX, Gilliet M, Liu YJ. Two functional subsets of FOXP3+ regulatory T cells in human thymus and periphery. Immunity. 2008;28:870–80. doi: 10.1016/j.immuni.2008.03.018.
    1. Schmetterer KG, Neunkirchner A, Pickl WF. Naturally occurring regulatory T cells: markers, mechanisms, and manipulation. FASEB J. 2012;26:2253–76. doi: 10.1096/fj.11-193672.
    1. Docetaxel Alone or in Combination With Vaccine to Treat Breast Cancer [NCI-6977].
    1. Yokokawa J, Cereda V, Remondo C, Gulley JL, Arlen PM, Schlom J, Tsang KY. Enhanced functionality of CD4+CD25(high)FoxP3+ regulatory T cells in the peripheral blood of patients with prostate cancer. Clin Cancer Res. 2008;14:1032–40. doi: 10.1158/1078-0432.CCR-07-2056.

Source: PubMed

3
订阅