Low-dose cyclophosphamide synergizes with dendritic cell-based immunotherapy in antitumor activity

Joris D Veltman, Margaretha E H Lambers, Menno van Nimwegen, Sanne de Jong, Rudi W Hendriks, Henk C Hoogsteden, Joachim G J V Aerts, Joost P J J Hegmans, Joris D Veltman, Margaretha E H Lambers, Menno van Nimwegen, Sanne de Jong, Rudi W Hendriks, Henk C Hoogsteden, Joachim G J V Aerts, Joost P J J Hegmans

Abstract

Clinical immunotherapy trials like dendritic cell-based vaccinations are hampered by the tumor's offensive repertoire that suppresses the incoming effector cells. Regulatory T cells are instrumental in suppressing the function of cytotoxic T cells. We studied the effect of low-dose cyclophosphamide on the suppressive function of regulatory T cells and investigated if the success rate of dendritic cell immunotherapy could be improved. For this, mesothelioma tumor-bearing mice were treated with dendritic cell-based immunotherapy alone or in combination with low-dose of cyclophosphamide. Proportions of regulatory T cells and the cytotoxic T cell functions at different stages of disease were analyzed. We found that low-dose cyclophosphamide induced beneficial immunomodulatory effects by preventing the induction of Tregs, and as a consequence, cytotoxic T cell function was no longer affected. Addition of cyclophosphamide improved immunotherapy leading to an increased median and overall survival. Future studies are needed to address the usefulness of this combination treatment for mesothelioma patients.

Figures

Figure 1
Figure 1
Increase in the proportion of regulatory T cells in a murine model for mesothelioma. (a) Left: The transcription factor Foxp3, a hallmark of naturally arising CD4+CD25+ Tregs, was expressed inside the tumor (red staining). Cells were counterstained with haematoxylin. [Magnification 200×] Right: A higher magnification (400×) shows the appearance of Foxp3+ cells as indicated by arrows. (b) Tregs (blue staining) were occasionally present in the near vicinity of CD8+ cells (red staining) [Magnification 1000×]. (c) and (d) Percentage of Tregs (defined as CD4+CD25+Foxp3+ cells) from total CD4+ T cells in the peripheral blood and draining lymph nodes is significantly higher at day 10 in tumor-bearing mice compared to healthy mice as observed by flow cytometry. (e) A positive correlation was found between the total tumor weight and the percentage of Tregs, measured in the lymph nodes of tumor-bearing mice. Five mice were euthanized at day 10 (tumor weight varied from 0.1 gram to 1 gram) and three mice at stage with ill health (tumor weight 1.5 to 2.1 gram). Correlation coefficient of the trend line (R-squared) is 0.780 (P  value is  .0039).
Figure 2
Figure 2
Tregs inhibit CTL function in vitro. (a) Activated splenocytes from DC-treated mice were cocultured with CD4+CD25+Foxp3+ cells (purified from tumor-bearing mice) in different ratios. The percentage of intracellular IFN-γ of CD8+ cells was measured using flow cytometry. And (b) the percentage of intracellular IFN-γ and granzyme B expression was measured using FACS. A reduction in the IFN-γ and granzyme B expression was found especially in those fractions where increasing doses of Treg were added to the CD8+ T cells.
Figure 3
Figure 3
CTX influences lymphocyte subsets in peripheral blood and draining lymph nodes of tumor-bearing mice. (a) The percentage of CD19+ cells was significantly decreased (P < .05) in CTX-treated tumor-bearing mice. CD3+ cells were significantly increased (P = .0325) in the peripheral blood of CTX-treated tumor-bearing mice compared to untreated tumor-bearing mice. (b) CD19+ cells were significantly decreased (P < .05) while CD3+ cells were significantly increased (P < .05), while the percentage of CD4+CD25+Foxp3+ cells was significantly decreased (P < .05) in the splenocytes of CTX-treated mice compared to untreated tumor-bearing mice.
Figure 4
Figure 4
CTX combined with DC-based immunotherapy prolongs survival. Kaplan-Meier survival plot showing the effect of the combination of CTX and DC-based immunotherapy. Mice were given drinking water supplemented with 0.13 mg/ml CTX from day 3 till day 10 and day 14 till day 21. Tumor-lysate-pulsed DCs were given at day 12. Survival significantly improved when CTX and DC-based immunotherapy were combined compared to DC-based immunotherapy (P = .0035) or CTX administration alone (P = .0056). Each group contained 8 mice.
Figure 5
Figure 5
Long-period administration of CTX improves survival compared to pre- or post-immunotherapy treatment. Mice were divided into four groups (n = 6 per group) and inoculated with a lethal dose of AB1 tumor cells on day 0. Mice received low-dose CTX before (day 3 till 10) or after (day 14 till 21) immunotherapy or metronomic dosed CTX (day 3 till 10 and day 14 till 21). Groups 2, 3, and 4 were treated with DC-immunotherapy on day 12. Group 1 functioned as a tumor control group and did not receive any treatment. Administration of metronomic dosed CTX was not significantly better than CTX treatment before (P = .840) or after (P = .454) immunotherapy. However, the combination of CTX and immunotherapy was significantly better than no treatment (CTX before immunotherapy compared to untreated P = .0081, CTX after immunotherapy compared to untreated P = .0147, metronomic dosed CTX and immunotherapy compared to untreated P = .0018).

References

    1. Robinson BWS, Musk AW, Lake RA. Malignant mesothelioma. The Lancet. 2005;366(9483):397–408.
    1. Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392(6673):245–252.
    1. Banchereau J, Palucka AK. Dendritic cells as therapeutic vaccines against cancer. Nature Reviews Immunology. 2005;5(4):296–306.
    1. Steinman RM, Dhodapkar M. Active immunization against cancer with dendritic cells: the near future. International Journal of Cancer. 2001;94(4):459–473.
    1. Hegmans JPJJ, Hemmes A, Aerts JG, Hoogsteden HC, Lambrecht BN. Immunotherapy of murine malignant mesothelioma using tumor lysate-pulsed dendritic cells. American Journal of Respiratory and Critical Care Medicine. 2005;171(10):1168–1177.
    1. Figdor CG, de Vries IJM, Lesterhuis WJ, Melief CJM. Dendritic cell immunotherapy: mapping the way. Nature Medicine. 2004;10(5):475–480.
    1. Sakaguchi S, Sakaguchi N, Shimizu J, et al. Immunologic tolerance maintained by CD25+CD4+ regulatory T cells: their common role in controlling autoimmunity, tumor immunity, and transplantation tolerance. Immunological Reviews. 2001;182:18–32.
    1. Hegmans JPJJ, Hemmes A, Hammad H, Boon L, Hoogsteden HC, Lambrecht BN. Mesothelioma environment comprises cytokines and T-regulatory cells that suppress immune responses. European Respiratory Journal. 2006;27(6):1086–1095.
    1. Meloni F, Morosini M, Solari N, et al. Foxp3 expressing CD4+CD25+ and CD8+CD8− T regulatory cells in the peripheral blood of patients with lung cancer and pleural mesothelioma. Human Immunology. 2006;67(1-2):1–12.
    1. DeLong P, Carroll RG, Henry AC, et al. Regulatory T cells and cytokines in malignant pleural effusions secondary to mesothelioma and carcinoma. Cancer Biology and Therapy. 2005;4(3):342–346.
    1. Berd D, Mastrangelo MJ. Active immunotherapy of human melanoma exploiting the immunopotentiating effects of cyclophosphamide. Cancer Investigation. 1988;6(3):337–349.
    1. Mescher MF, Rogers JD. Immunotherapy of established murine tumors with large multivalent immunogen and cyclophosphamide. Journal of Immunotherapy. 1996;19(2):102–112.
    1. Liu J-Y, Wu Y, Zhang X-S, et al. Single administration of low dose cyclophosphamide augments the antitumor effect of dendritic cell vaccine. Cancer Immunology, Immunotherapy. 2007;56(10):1597–1604.
    1. Taieb J, Chaput N, Schartz N, et al. Chemoimmunotherapy of tumors: cyclophosphamide synergizes with exosome based vaccines. Journal of Immunology. 2006;176(5):2722–2729.
    1. Goldfarb RH, Ohashi M, Brunson KW, et al. Augmentation of IL-2 activated natural killer cell adoptive immunotherapy with cyclophosphamide. Anticancer Research. 1998;18(3):1441–1446.
    1. Proietti E, Greco G, Garrone B, et al. Importance of cyclophosphamide-induced bystander effect on T cells for a successful tumor eradication in response to adoptive immunotherapy in mice. Journal of Clinical Investigation. 1998;101(2):429–441.
    1. Mihalyo MA, Doody ADH, McAleer JP, et al. In vivo cyclophosphamide and IL-2 treatment impedes self-antigen-induced effector CD4 cell tolerization: implications for adoptive immunotherapy. Journal of Immunology. 2004;172(9):5338–5345.
    1. Li L, Okino T, Sugie T, et al. Cyclophosphamide given after active specific immunization augments antitumor immunity by modulation of Th1 commitment of CD4+ T cells. Journal of Surgical Oncology. 1998;67(4):221–227.
    1. Hermans IF, Chong TW, Palmowski MJ, Harris AL, Cerundolo V. Synergistic effect of metronomic dosing of cyclophosphamide combined with specific antitumor immunotherapy in a murine melanoma model. Cancer Research. 2003;63(23):8408–8413.
    1. North RJ. Cyclophosphamide-facilitated adoptive immunotherapy of an established tumor depends on elimination of tumor-induced suppressor T cells. Journal of Experimental Medicine. 1982;155(4):1063–1074.
    1. Salem ML, Kadima AN, El-Naggar SA, et al. Defining the ability of cyclophosphamide preconditioning to enhance the antigen-specific CD8+ T-cell response to peptide vaccination: creation of a beneficial host microenvironment involving type I IFNs and myeloid cells. Journal of Immunotherapy. 2007;30(1):40–53.
    1. Nowak AK, Lake RA, Robinson BWS. Combined chemoimmunotherapy of solid tumours: improving vaccines? Advanced Drug Delivery Reviews. 2006;58(8):975–990.
    1. Ghiringhelli F, Menard C, Puig PE, et al. Metronomic cyclophosphamide regimen selectively depletes CD4+CD25+ regulatory T cells and restores T and NK effector functions in end stage cancer patients. Cancer Immunology, Immunotherapy. 2007;56(5):641–648.
    1. Motoyoshi Y, Kaminoda K, Saitoh O, et al. Different mechanisms for anti-tumor effects of low- and high-dose cyclophosphamide. Oncology Reports. 2006;16(1):141–146.
    1. Ikezawa Y, Nakazawa M, Tamura C, Takahashi K, Minami M, Ikezawa Z. Cyclophosphamide decreases the number, percentage and the function of CD25+CD4+ regulatory T cells, which suppress induction of contact hypersensitivity. Journal of Dermatological Science. 2005;39(2):105–112.
    1. Lutsiak MEC, Semnani RT, De Pascalis R, Kashmiri SVS, Schlom J, Sabzevari H. Inhibition of CD4+25+ T regulatory cell function implicated in enhanced immune response by low-dose cyclophosphamide. Blood. 2005;105(7):2862–2868.
    1. Ghiringhelli F, Larmonier N, Schmitt E, et al. CD4+CD25+ regulatory T cells suppress tumor immunity but are sensitive to cyclophosphamide which allows immunotherapy of established tumors to be curative. European Journal of Immunology. 2004;34(2):336–344.
    1. van der Most RG, Currie AJ, Mahendran S, et al. Tumor eradication after cyclophosphamide depends on concurrent depletion of regulatory T cells: a role for cycling TNFR2-expressing effector-suppressor T cells in limiting effective chemotherapy. Cancer Immunology, Immunotherapy. 2009;58(8):1219–1228.
    1. van der Most RG, Currie AJ, Mahendran S, et al. Tumor eradication after cyclophosphamide depends on concurrent depletion of regulatory T cells: a role for cycling TNFR2-expressing effector-suppressor T cells in limiting effective chemotherapy. Cancer Immunology, Immunotherapy. 2009;58(8):1219–1228.
    1. Jackaman C, Cornwall S, Lew AM, Zhan Y, Robinson BWS, Nelson DJ. Local effector failure in mesothelioma is not mediated by CD4+CD25+ T-regulator cells. European Respiratory Journal. 2009;34(1):162–175.
    1. Davis MR, Manning LS, Whitaker D, Garlepp MJ, Robinson BWS. Establishment of a murine model of malignant mesothelioma. International Journal of Cancer. 1992;52(6):881–886.
    1. Lutz MB, Kukutsch N, Ogilvie ALJ, et al. An advanced culture method for generating large quantities of highly pure dendritic cells from mouse bone marrow. Journal of Immunological Methods. 1999;223(1):77–92.
    1. Miyara M, Sakaguchi S. Natural regulatory T cells: mechanisms of suppression. Trends in Molecular Medicine. 2007;13(3):108–116.
    1. Sakaguchi S. Regulatory T cells. Springer Seminars in Immunopathology. 2006;28(1):1–2.
    1. Yamaguchi T, Sakaguchi S. Regulatory T cells in immune surveillance and treatment of cancer. Seminars in Cancer Biology. 2006;16(2):115–123.
    1. Nishikawa H, Kato T, Tawara I, et al. Definition of target antigens for naturally occurring CD4+CD25+ regulatory T cells. Journal of Experimental Medicine. 2005;201(5):681–686.
    1. Sakaguchi S. Immunologic tolerance maintained by regulatory T cells: implications for autoimmunity, tumor immunity and transplantation tolerance. Vox Sanguinis. 2002;83(supplement 1):151–153.
    1. Machiels J-PH, Reilly RT, Emens LA, et al. Cyclophosphamide, doxorubicin, and paclitaxel enhance the antitumor immune response of granulocyte/macrophage-colony stimulating factor-secreting whole-cell vaccines in HER-2/neu tolerized mice. Cancer Research. 2001;61(9):3689–3697.
    1. Wada S, Yoshimura K, Hipkiss EL, et al. Cyclophosphamide augments antitumor immunity: studies in an autochthonous prostate cancer model. Cancer Research. 2009;69(10):4309–4318.
    1. Maesen F, Willighagen R. Regression of a malignant tumour of the pleura. European Journal of Respiratory Diseases. 1987;71(2):135–138.
    1. Robinson BWS, Robinson C, Lake RA. Localised spontaneous regression in mesothelioma—possible immunological mechanism. Lung Cancer. 2001;32(2):197–201.
    1. Schwartz E, Maayan C, Mouallem M, Engelberg S, Friedman E. Malignant peritoneal mesothelioma: long-term spontaneous clinical remission. Medical and Pediatric Oncology. 1991;19(4):325–328.
    1. Pilling JE, Nicholson AG, Harmer C, Goldstraw P. Prolonged survival due to spontaneous regression and surgical excision of malignant mesothelioma. Annals of Thoracic Surgery. 2007;83(1):314–315.
    1. Yanagawa H, Sone S, Fukuta K, Nishioka Y, Ogura T. Local adoptive immunotherapy using lymphokine-activated killer cells and interleukin-2 against malignant pleural mesothelioma: report of two cases. Japanese Journal of Clinical Oncology. 1991;21(5):377–383.
    1. Bielefeldt-Ohmann H, Marzo AL, Himbeck RP, Jarnicki AG, Robinson BWS, Fitzpatrick DR. Interleukin-6 involvement in mesothelioma pathobiology: inhibition by interferon α immunotherapy. Cancer Immunology Immunotherapy. 1995;40(4):241–250.
    1. Astoul P, Picat-Joossen D, Viallat J-R, Boutin C. Intrapleural administration of interleukin-2 for the treatment of patients with malignant pleural mesothelioma: a phase II study. Cancer. 1998;83(10):2099–2104.
    1. Caminschi I, Venetsanakos E, Leong CC, Garlepp MJ, Robinson BWS, Scott B. Cytokine gene therapy of mesothelioma: immune and antitumor effects of transfected interleukin-12. American Journal of Respiratory Cell and Molecular Biology. 1999;21(3):347–356.
    1. Kruklitis RJ, Singhal S, Delong P, et al. Immuno-gene therapy with interferon-β before surgical debulking delays recurrence and improves survival in a murine model of malignant mesothelioma. Journal of Thoracic and Cardiovascular Surgery. 2004;127(1):123–130.
    1. Robinson C, Callow M, Stevenson S, Scott B, Robinson BWS, Lake RA. Serologic responses in patients with malignant mesothelioma. Evidence for both public and private specificities. American Journal of Respiratory Cell and Molecular Biology. 2000;22(5):550–556.
    1. Banchereau J, Briere F, Caux C, et al. Immunobiology of dendritic cells. Annual Review of Immunology. 2000;18:767–811.
    1. Nagaraj S, Gabrilovich DI. Tumor escape mechanism governed by myeloid-derived suppressor cells. Cancer Research. 2008;68(8):2561–2563.
    1. Grossman WJ, Verbsky JW, Barchet W, Colonna M, Atkinson JP, Ley TJ. Human T regulatory cells can use the perforin pathway to cause autologous target cell death. Immunity. 2004;21(4):589–601.
    1. Cao X, Cai SF, Fehniger TA, et al. Granzyme B and perforin are important for regulatory T cell-mediated suppression of tumor clearance. Immunity. 2007;27(4):635–646.
    1. Darrasse-Jèze G, Bergot A-S, Durgeau A, et al. Tumor emergence is sensed by self-specific CD44hi memory Tregs that create a dominant tolerogenic environment for tumors in mice. Journal of Clinical Investigation. 2009;119(9):2648–2662.
    1. Awwad M, North RJ. Cyclophosphamide-induced immunologically mediated regression of a cyclophosphamide-resistant murine tumor: a consequence of eliminating precursor L3T4+ suppressor T-cells. Cancer Research. 1989;49(7):1649–1654.
    1. Ercolini AM, Ladle BH, Manning EA, et al. Recruitment of latent pools of high-avidity CD8+ T cells to the antitumor immune response. Journal of Experimental Medicine. 2005;201(10):1591–1602.
    1. Xu L, Xu W, Jiang Z, Zhang F, Chu Y, Xiong S. Depletion of CD4+CD25high regulatory T cells from tumor infiltrating lymphocytes predominantly induces Th1 type immune response in vivo which inhibits tumor growth in adoptive immunotherapy. Cancer Biology and Therapy. 2009;8(1):66–72.
    1. Hori S, Sakaguchi S. Foxp3: a critical regulator of the development and function of regulatory T cells. Microbes and Infection. 2004;6(8):745–751.

Source: PubMed

3
Se inscrever