Substantially modified ratios of effector to regulatory T cells during chemotherapy in ovarian cancer patients return to pre-treatment levels at completion: implications for immunotherapy

Anthony Park, Chindu Govindaraj, Sue D Xiang, Julene Halo, Michael Quinn, Karen Scalzo-Inguanti, Magdalena Plebanski, Anthony Park, Chindu Govindaraj, Sue D Xiang, Julene Halo, Michael Quinn, Karen Scalzo-Inguanti, Magdalena Plebanski

Abstract

Ovarian cancer is the leading cause of death from gynaecological malignancy. Despite improved detection and treatment options, relapse rates remain high. Combining immunotherapy with the current standard treatments may provide an improved prognosis, however, little is known about how standard chemotherapy affects immune potential (particularly T cells) over time, and hence, when to optimally combine it with immunotherapy (e.g., vaccines). Herein, we assess the frequency and ratio of CD8+ central memory and effector T cells as well as CD4+ effector and regulatory T cells (Tregs) during the first 18 weeks of standard chemotherapy for ovarian cancer patients. In this pilot study, we observed increased levels of recently activated Tregs with tumor migrating ability (CD4+CD25hiFoxp3+CD127-CCR4+CD38+ cells) in patients when compared to controls. Although frequency changes of Tregs as well as the ratio of effector T cells to Tregs were observed during treatment, the Tregs consistently returned to pre-chemotherapy levels at the end of treatment. These results indicate T cell subset distributions associated with recurrence may be largely resistant to being "re-set" to healthy control homeostatic levels following standard treatments. However, it may be possible to enhance T effector to Treg ratios transiently during chemotherapy. These results suggest personalized immune monitoring maybe beneficial when combining novel immuno-therapeutics with standard treatment for ovarian cancer patients.

Figures

Figure 1
Figure 1
Major subsets within the PBMCs of ovarian cancer patients over the course of standard chemotherapy. Peripheral blood from five ovarian cancer patients was collected over the course of the standard chemotherapeutic treatment. PBMCs of patients and controls (n = 10) were isolated and flow cytometry was performed to determine (A) the T cell (CD3+ CD56−) frequency within PBMCs; (B) the CD4+ T cell frequency within the T cell fraction (C) CD8+ T cell frequency within the T cell fraction. Comparison of the frequency of cell populations between patients and controls: ** p < 0.01.
Figure 2
Figure 2
Frequencies of CD8+ T cell subsets present within the peripheral blood of ovarian cancer patients over the course of chemotherapy. Flow cytometry was performed on PBMCS isolated from patients (n = 5) and controls (n = 6). (A) The gating strategy employed to identify effector and central memory CD8+ T cells; (B) Comparison of the CD8+ T cell subset frequencies between controls and patients; pre-chemo representing 0 rounds while post-chemo represents 6 rounds of chemotherapy; (C) Frequency fluctuations of CD8+ T cell subsets of patients over the course of chemotherapy. The dotted line represents the mean frequency of the healthy controls. Comparison of CD8+ T cell subset frequencies between controls and patients: not significant.
Figure 3
Figure 3
Frequency of recently activated CD8+ T cell subsets present within the PBMCs of patients during the course of chemotherapy. Flow cytometry was performed on PBMCS isolated from controls (n = 6) and patients undergoing chemotherapy (n = 5). (A) The gating strategy employed to identify recently activated effector and central memory CD8+ T cells present within the PBMCs; (B) Comparison of the CD8+ T cell subsets between controls and patients: pre-chemo representing 0 rounds while post-chemo represents 6 rounds of chemotherapy; (C) Frequency fluctuations of CD8+ T cell subsets of patients over the course of chemotherapy. The dotted line represents the mean frequency of the healthy controls. Comparison of CD8+ T cell subset frequencies between controls and patients: not significant.
Figure 4
Figure 4
Frequencies of regulatory cell subsets present within the peripheral blood of ovarian cancer patients. (A) The gating strategy employed to identify regulatory T cells (CD4+CD25hiFOXP3+CD127−) present in the PBMCs; (B) Comparison of regulatory T cell subset frequencies between healthy controls (n = 5) and ovarian cancer patients undergoing chemotherapy (n = 5); (C) Frequency fluctuations of regulatory T cell subsets of patients over the course of chemotherapy. The dotted line represents the mean frequency of the healthy controls. Comparison of regulatory T cell subset frequencies between controls and patients: * p < 0.05.
Figure 5
Figure 5
Frequency of CD4+ effector T cell subsets present within the PBMCs of patients. (A) The gating strategy employed to identify the different CD4+ T cell subsets present in the PBMCs; (B) Comparison of CD4+ T cell subset frequencies between healthy controls (n = 5) and ovarian cancer patients undergoing chemotherapy (n = 5); (C) Frequency fluctuations of CD4+ T cell subsets of patients over the course of chemotherapy. The dotted line represents the mean frequency of the healthy controls. Comparison of T cell subset frequencies between controls and patients: * p < 0.05.
Figure 6
Figure 6
Ratio of CD4+ and CD8+ T cell subsets to Treg subsets identified within PBMCs of cancer patients over standard chemotherapeutic treatment. The ratios of (A) CD4+ T cell subsets to their corresponding Treg subsets and (B) CD8+ T cell subset to conventional Tregs (CD4+CD25hiFOXP3+CD127−) for the individual patients before each round of chemotherapy are represented here. The ratios of all five cancer patients before the first and during the last round of chemotherapy for their corresponding subsets are also shown in concurrent bar graphs (not significant).

References

    1. Bristow R.E., Berek J.S. Surgery for ovarian cancer: How to improve survival. Lancet. 2006;367:1558–1560. doi: 10.1016/S0140-6736(06)68671-6.
    1. Mastropaolo D., Camerman A., Luos Y., Brayers G., Camerman N. Crystal and molecular structure of paclitaxel (taxol) Proc. Natl. Acad. Sci. USA. 1995;92:6920–6924.
    1. Geney R., Sun L., Pera P., Bernacki R., Xia S., Horwitz S., Simmerling C., Ojima I. Use of the tubulin bound paclitaxel conformation for structure-based rational drug design. Chem. Biol. 2005;12:339–348. doi: 10.1016/j.chembiol.2005.01.004.
    1. Rustin G.J.S., Bast R.C., Jr., Kelloff G.J., Barrett J.C., Carter S.K., Nisen P.D., Sigman C.C., Parkinson D.R., Ruddon R.W. Use of CA-125 in clinical trial evaluation of new therapeutic drugs for ovarian cancer. Clin. Cancer Res. 2004;10:3919–3926.
    1. Bali A., Reynolds K. The current management of primary ovarian cancer: A review. Cancer Ther. 2004;2:305–316.
    1. Bookman M.A., Brady M.F., McGuire W.P., Harper P.G., Alberts D.S., Friedlander M., Colombo N., Fowler J.M., Argenta P.A., de Geest K., et al. Evaluation of new platinum-based treatment regimens in advanced-stage ovarian cancer: A Phase III Trial of the Gynecologic Cancer Intergroup. J. Clin. Oncol. 2009;27:1419–1425.
    1. Australian Institute of Health and Welfare. Cancer Survival and Prevalence in Australia—Cancers Diagnosed from 1982 to 2004. Australian Institute of Health and Welfare; Canberra ACT, Austrilia: 2008.
    1. Ozols R.F., Bundy B.N., Greer B.E., Fowler J.M., Clarke-Pearson D., Burger R.A., Mannel R.S., DeGeest K., Hartenbach E.M., Baergen R. Phase III trial of carboplatin and paclitaxel compared with cisplatin and paclitaxel in patients with optimally resected stage III ovarian cancer: A gynecologic oncology group study. J. Clin. Oncol. 2003;21:3194–3200.
    1. Curiel T.J. Tregs and rethinking cancer immunotherapy. J. Clin. Invest. 2007;117:1167–1174. doi: 10.1172/JCI31202.
    1. Ichihara F., Kono K., Takahashi A., Kawaida H., Sugai H., Fujii H. Increased populations of regulatory T cells in peripheral blood and tumor-infiltrating lymphocytes in patients with gastric and esophageal cancers. Clin. Cancer Res. 2003;9:4404–4408.
    1. Ormandy L.A., Hillemann T., Wedemeyer H., Manns M.P., Greten T.F., Korangy F. Increased populations of regulatory T cells in peripheral blood of patients with hepatocellular carcinoma. Cancer Res. 2005;65:2457–2464.
    1. Wolf A.M., Wolf D., Steurer M., Gastl G., Gunsilius E., Grubeck-Loebenstein B. Increase of regulatory T cells in the peripheral blood of cancer patients. Clin. Cancer Res. 2003;9:606–612.
    1. Liyanage U.K., Moore T.T., Joo H.-G., Tanaka Y., Herrmann V., Doherty G., Drebin J.A., Strasberg S.M., Eberlein T.J., Goedegebuure P.S., et al. Prevalence of regulatory T cells is increased in peripheral blood and tumor microenvironment of patients with pancreas or breast adenocarcinoma. J. Immunol. 2002;169:2756–2761.
    1. Takahashi T., Tagami T., Yamazaki S., Uede T., Shimizu J., Sakaguchi N., Mak T.W., Sakaguchi S. Immunologic self-tolerance maintained by CD25+CD4+ regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. J. Exp. Med. 2000;192:303–310. doi: 10.1084/jem.192.2.303.
    1. Tang Q., Bluestone J.A. The Foxp3+ regulatory T cell: A jack of all trades, master of regulation. Nat. Immunol. 2008;9:239–244.
    1. Ghiringhelli F., Menard C., Puig P., Ladoire S., Roux S., Martin F., Solary E., Le Cesne A., Zitvogel L., Chauffert B. Metronomic cyclophosphamide regimen selectively depletes CD4+CD25+ regulatory T cells and restores T and NK effector functions in end stage cancer patients. Cancer Immunol. Immunother. 2007;56:641–648. doi: 10.1007/s00262-006-0225-8.
    1. Curiel T.J., Coukos G., Zou L., Alvarez X., Cheng P., Mottram P., Evdemon-Hogan M., Conejo-Garcia J.R., Zhang L., Burow M., et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat. Med. 2004;10:942–949.
    1. Wolf D., Wolf A.M., Rumpold H., Fiegl H., Zeimet A.G., Muller-Holzner E., Deibl M., Gastl G., Gunsilius E., Marth C. The expression of the regulatory T cell specific forkhead box transcription factor FoxP3 is associated with poor prognosis in ovarian cancer. Clin. Cancer Res. 2005;11:8326–8331.
    1. Dannull J., Su Z., Rizzieri D., Yang B.K., Coleman D., Yancey D., Zhang A., Dahm P., Chao N., Gilboa E., et al. Enhancement of vaccine-mediated antitumor immunity in cancer patients after depletion of regulatory T cells. J. Clin. Invest. 2005;115:3623–3633.
    1. Gnjatic S., Altorki N.K., Tang D.N., Tu S.-M., Kundra V., Ritter G., Old L.J., Logothetis C.J., Sharma P. NY-ESO-1 DNA vaccine induces T-cell responses that are suppressed by Regulatory T cells. Clin. Cancer Res. 2009;15:2130–2139.
    1. Nicholaou T., Ebert L.M., Davis I.D., McArthur G.A., Jackson H., Dimopoulos N., Tan B., Maraskovsky E., Miloradovic L., Hopkins W., et al. Regulatory T-cell mediated attenuation of T-cell responses to the NY-ESO-1 ISCOMATRIX vaccine in patients with advanced malignant melanoma. Clin. Cancer Res. 2009;15:2166–2173.
    1. Curiel T.J., Barnett B., Kryczek I., Cheng P., Zou W. Regulatory T cells in ovarian cancer: Biology and therapeutic potential. Cancer Immun. 2006;6:20.
    1. Stahnke K., Fulda S., Friesen C., Strau G., Debatin K.-M. Activation of apoptosis pathways in peripheral blood lymphocytes by in vivo chemotherapy. Blood. 2001;98:3066–3073. doi: 10.1182/blood.V98.10.3066.
    1. Zou W. Regulatory T cells, tumour immunity and immunotherapy. Nat. Rev. Immunol. 2006;6:295–307. doi: 10.1038/nri1806.
    1. Vicari A., Luu R., Zhang N., Patel S., Makinen S., Hanson D., Weeratna R., Krieg A. Paclitaxel reduces regulatory T cell numbers and inhibitory function and enhances the anti-tumor effects of the TLR9 agonist PF-3512676 in the mouse. Cancer Immunol. Immunother. 2009;58:615–628. doi: 10.1007/s00262-008-0586-2.
    1. Zhang L., Dermawan K., Jin M., Liu R., Zheng H., Xu L., Zhang Y., Cai Y., Chu Y., Xiong S. Differential impairment of regulatory T cells rather than effector T cells by paclitaxel-based chemotherapy. Clin. Immunol. 2008;129:219–229. doi: 10.1016/j.clim.2008.07.013.
    1. Malmberg K.J. Effective immunotherapy against cancer: A question of overcoming immune suppression and immune escape? Cancer Immunol. Immunother. 2004;53:879–892.
    1. Wu X., Feng Q.-M., Wang Y., Shi J., Ge H.-L., Di W. The immunologic aspects in advanced ovarian cancer patients treated with paclitaxel and carboplatin chemotherapy. Cancer Immunol. Immunother. 2010;59:279–291. doi: 10.1007/s00262-009-0749-9.
    1. Coleman S., Clayton A., Mason M.D., Jasani B., Adams M., Tabi Z. Recovery of CD8+ T cell function during systemic chemotherapy in advanced ovarian cancer. Cancer Res. 2005;65:7000–7006.
    1. Appay V., Dunbar P.R., Callan M., Klenerman P., Gillespie G.M., Papagno L., Ogg G.S., King A., Lechner F., Spina C.A., et al. Memory CD8+ T cells vary in differentiation phenotype in different persistent virus infections. Nat. Med. 2002;8:379–385.
    1. Sato E., Olson S.H., Ahn J., Bundy B., Nishikawa H., Qian F., Jungbluth A.A., Frosina D., Gnjatic S., Ambrosone C., et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc. Natl. Acad. Sci. USA. 2005;102:18538–18543.
    1. Curiel T.J., Coukos G., Zou L., Alvarez X., Cheng P., Mottram P., Evdemon-Hogan M., Conejo-Garcia J.R., Zhang L., Burow M., et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat. Med. 2004;10:942–949.
    1. Mempel T.R., Bauer C.A. Intravital imaging of CD8+ T cell function in cancer. Clin. Exp. Metastasis. 2009;26:311–327. doi: 10.1007/s10585-008-9196-9.
    1. Gajewski T.F., Meng Y., Blank C., Brown I., Kacha A., Kline J., Harlin H. Immune resistance orchestrated by the tumor microenvironment. Immunol. Rev. 2006;213:131–145. doi: 10.1111/j.1600-065X.2006.00442.x.
    1. Sato E., Olson S.H., Ahn J., Bundy B., Nishikawa H., Qian F., Jungbluth A.A., Frosina D., Gnjatic S., Ambrosone C., et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc. Natl. Acad. Sci. USA. 2005;102:18538–18543.
    1. Klebanoff C.A., Gattinoni L., Torabi-Parizi P., Kerstann K., Cardones A.R., Finkelstein S.E., Palmer D.C., Antony P.A., Hwang S.T., Rosenberg S.A., et al. Central memory self/tumor-reactive CD8+ T cells confer superior antitumor immunity compared with effector memory T cells. Proc. Natl. Acad. Sci. USA. 2005;102:9571–9576.
    1. Seki I., Suzuki M., Miyasaka N., Kohsaka H. Expression of CD45 isoforms correlates with differential proliferative responses of peripheral CD4+ and CD8+ T cells. Immunol. Lett. 2010;129:39–46. doi: 10.1016/j.imlet.2009.12.020.
    1. Froebel K.S., Doherty K.V., Whitelaw J.A., Hague R.A., Mok J.Y., Bird A.G. Increased expression of the CD45RO (memory) antigen on T cells in HIV-infected children. AIDS. 1991;5:97–99.
    1. Mackall C.L., Fleisher T.A., Brown M.R., Magrath I.T., Shad A.T., Horowitz M.E., Wexler L.H., Adde M.A., McClure L.L., Gress R.E. Lymphocyte depletion during treatment with intensive chemotherapy for cancer. Blood. 1994;84:2221–2228.
    1. Liu Z., Cumberland W.G., Hultin L.E., Prince H.E., Detels R., Giorgi J.V. Elevated CD38 antigen expression on CD8+ T cells is a stronger marker for the risk of chronic HIV disease progression to AIDS and death in the Multicenter AIDS Cohort Study than CD4+ cell count, soluble immune activation markers, or combinations of HLA-DR and CD38 expression. J. Acquir. Immune Defic. Syndr. Hum. Retrovirol. 1997;16:83–92. doi: 10.1097/00042560-199710010-00003.
    1. Mackay C.R. Dual personality of memory T cells. Nature. 1999;401:659–660. doi: 10.1038/44309.
    1. Galgani M., di Giacomo A., Matarese G., La Cava A. The Yin and Yang of CD4(+) regulatory T cells in autoimmunity and cancer. Curr. Med. Chem. 2009;16:4626–4631. doi: 10.2174/092986709789878201.
    1. Curiel T.J., Coukos G., Zou L., Alvarez X., Cheng P., Mottram P., Evdemon-Hogan M., Conejo-Garcia J.R., Zhang L., Burow M., et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat. Med. 2004;10:942–949.
    1. Baecher-Allan C., Brown J.A., Freeman G.J., Hafler D.A. CD4+CD25 high regulatory cells in human peripheral blood. J. Immunol. 2001;167:1245–1253.
    1. Milne K., Alexander C., Webb J.R., Sun W., Dillon K., Kalloger S.E., Gilks C.B., Clarke B., Kobel M., Nelson B.H. Absolute lymphocyte count is associated with survival in ovarian cancer independent of tumor-infiltrating lymphocytes. J. Transl. Med. 2012;10:33. doi: 10.1186/1479-5876-10-33.
    1. Sakaguchi S. Regulatory T cells: Key controllers of immunologic self-tolerance. Cell. 2000;101:455–458. doi: 10.1016/S0092-8674(00)80856-9.
    1. Takahashi T., Tagami T., Yamazaki S., Uede T., Shimizu J., Sakaguchi N., Mak T.W., Sakaguchi S. Immunologic self-tolerance maintained by CD25(+)CD4(+) regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. J. Exp. Med. 2000;192:303–310. doi: 10.1084/jem.192.2.303.
    1. Wu X., Feng Q.M., Wang Y., Shi J., Ge H.L., Di W. The immunologic aspects in advanced ovarian cancer patients treated with paclitaxel and carboplatin chemotherapy. Cancer Immunol. Immunother. 2010;59:279–291. doi: 10.1007/s00262-009-0749-9.
    1. Rech A.J., Mick R., Kaplan D.E., Chang K.M., Domchek S.M., Vonderheide R.H. Homeostasis of peripheral FoxP3(+) CD4(+) regulatory T cells in patients with early and late stage breast cancer. Cancer Immunol. Immunother. 2010;59:599–607. doi: 10.1007/s00262-009-0780-x.
    1. Chen X., Oppenheim J.J. TNF-alpha: An activator of CD4+FoxP3+TNFR2+ regulatory T cells. Curr. Dir. Autoimmun. 2010;11:119–134. doi: 10.1159/000289201.
    1. Fu S., Zhang N., Yopp A.C., Chen D., Mao M., Zhang H., Ding Y., Bromberg J.S. TGF-beta induces Foxp3 + T-regulatory cells from CD4 + CD25—Precursors. Am. J. Transplant. 2004;4:1614–1627. doi: 10.1111/j.1600-6143.2004.00566.x.
    1. Lake R.A., Robinson B.W. Immunotherapy and chemotherapy—A practical partnership. Nat. Rev. Cancer. 2005;5:397–405. doi: 10.1038/nrc1613.
    1. Ghiringhelli F., Menard C., Puig P.E., Ladoire S., Roux S., Martin F., Solary E., Le Cesne A., Zitvogel L., Chauffert B. Metronomic cyclophosphamide regimen selectively depletes CD4+CD25+ regulatory T cells and restores T and NK effector functions in end stage cancer patients. Cancer Immunol. Immunother. 2007;56:641–648. doi: 10.1007/s00262-006-0225-8.
    1. Ghiringhelli F., Larmonier N., Schmitt E., Parcellier A., Cathelin D., Garrido C., Chauffert B., Solary E., Bonnotte B., Martin F. CD4+CD25+ regulatory T cells suppress tumor immunity but are sensitive to cyclophosphamide which allows immunotherapy of established tumors to be curative. Eur. J. Immunol. 2004;34:336–344.
    1. Barnett B., Kryczek I., Cheng P., Zou W., Curiel T.J. Regulatory T cells in ovarian cancer: Biology and therapeutic potential. Am. J. Reprod. Immunol. 2005;54:369–377. doi: 10.1111/j.1600-0897.2005.00330.x.
    1. Tredan O., Geay J.F., Touzet S., Delva R., Weber B., Cretin J., Provencal J., Martin J., Stefani L., Pujade-Lauraine E., et al. Carboplatin/cyclophosphamide or carboplatin/paclitaxel in elderly patients with advanced ovarian cancer? Analysis of two consecutive trials from the Groupe d’Investigateurs Nationaux pour l’Etude des Cancers Ovariens. Ann. Oncol. 2007;18:256–262.
    1. Stumpf M., Hasenburg A., Riener M.O., Jutting U., Wang C., Shen Y., Orlowska-Volk M., Fisch P., Wang Z., Gitsch G., et al. Intraepithelial CD8-positive T lymphocytes predict survival for patients with serous stage III ovarian carcinomas: Relevance of clonal selection of T lymphocytes. Br. J. Cancer. 2009;101:1513–1521.

Source: PubMed

3
Se inscrever