Clinical response of advanced cancer patients to cellular immunotherapy and intensity-modulated radiation therapy

Kenichiro Hasumi, Yukimasa Aoki, Ryuko Wantanabe, Dean L Mann, Kenichiro Hasumi, Yukimasa Aoki, Ryuko Wantanabe, Dean L Mann

Abstract

Patients afflicted with advanced cancers were treated with the intratumoral injection of autologous immature dendritic cells (iDCs) followed by activated T-cell infusion and intensity-modulated radiation therapy (IMRT). A second round of iDCs and activated T cells was then administered to patients after the last radiation cycle. This complete regimen was repeated for new and recurring lesions after 6 weeks of follow-up. One year post therapy, outcome analyses were performed to evaluate treatment efficacy. Patients were grouped according to both the number and size of tumors and clinical parameters at treatment initiation, including recurrent disease after standard cancer therapy, Stage IV disease, and no prior therapy. Irrespective of prior treatment status, 23/37 patients with ≤ 5 neoplastic lesions that were ≤ 3 cm in diameter achieved complete responses (CRs), and 5/37 exhibited partial responses (PRs). Among 130 individuals harboring larger and more numerous lesions, CRs were observed in 7/74 patients that had received prior SCT and in 2/56 previously untreated patients. Some patients manifested immune responses including an increase in CD8+CD56+ lymphocytes among circulating mononuclear cells in the course of treatment. To prospectively explore the therapeutic use of these cells, CD8+ cells were isolated from patients that had been treated with cellular immunotherapy and IMRT, expanded in vitro, and injected into recurrent metastatic sites in 13 individuals who underwent the same immunoradiotherapeutic regimens but failed to respond. CRs were achieved in 34 of 58 of such recurrent lesions while PRs in 17 of 58. These data support the expanded use of immunoradiotherapy in advanced cancer patients exhibiting progressive disease.

Keywords: activated T cells; cancer vaccine; cytotoxic T lymphocytes; immature dendritic cells; intratumoral injection.

Figures

https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3858384/bin/onci-2-e26381-g1.jpg
Figure 1. Immunoradiotherapeutic protocol employed in this study. (A) DC-IMRT protocol: sequence of administration of immature dendritic cells (iDCs), activated T cells and intensity modulated radiation therapy (IMRT) followed by the re-administration of the iDCs and activated T cells. (B) CTL protocol: isolation of cells for the generation of cytotoxic T lymphocytes (CTLs) and timing of subsequent intratumoral injection (blue arrows).
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3858384/bin/onci-2-e26381-g2.jpg
Figure 2. Expression of CD3, CD8 and CD56 among peripheral blood mononuclear cells isolated before and after immunoradiotherapy. Representative dot plots of peripheral blood mononuclear cells (PBMCs) obtained from a patient with metastatic gastric cancer before (first panel in each row) and after (1st round, 2nd round, and 1-y follow-up, as indicated) the immunoradiotherapeutic regimen described in Figure 1A, upon staining with anti-CD3, anti-CD8 and anti-CD56 antibodies.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3858384/bin/onci-2-e26381-g3.jpg
Figure 3. Therapeutic responses to the intratumoral injection of therapy-primed cytotoxic T lymphocytes. Representative PET-CT images of metastatic lung cancer lesions (arrows) injected with autologous cytotoxic T lymphocytes (CTLs) generated ex vivo from CD8+ T cells isolated from the peripheral blood mononuclear cells upon the failure of immunoradiotherapy. The chronological progression is indicated by date and demonstrates tumor regression.

References

    1. Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392:245–52. doi: 10.1038/32588.
    1. Ilett EJ, Prestwich RJ, Melcher AA. The evolving role of dendritic cells in cancer therapy. Expert Opin Biol Ther. 2010;10:369–79. doi: 10.1517/14712590903559830.
    1. Galluzzi L, Senovilla L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology. 2012;1:1111–34. doi: 10.4161/onci.21494.
    1. van den Broek M, von Boehmer L, Knuth A. Developments in cancer immunotherapy. Dig Dis. 2010;28:51–6. doi: 10.1159/000282064.
    1. Small EJ, Schellhammer PF, Higano CS, Redfern CH, Nemunaitis JJ, Valone FH, Verjee SS, Jones LA, Hershberg RM. Placebo-controlled phase III trial of immunologic therapy with sipuleucel-T (APC8015) in patients with metastatic, asymptomatic hormone refractory prostate cancer. J Clin Oncol. 2006;24:3089–94. doi: 10.1200/JCO.2005.04.5252.
    1. Brower V. Approval of provenge seen as first step for cancer treatment vaccines. J Natl Cancer Inst. 2010;102:1108–10. doi: 10.1093/jnci/djq295.
    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. Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9:162–74. doi: 10.1038/nri2506.
    1. Ménétrier-Caux C, Gobert M, Caux C. Differences in tumor regulatory T-cell localization and activation status impact patient outcome. Cancer Res. 2009;69:7895–8. doi: 10.1158/0008-5472.CAN-09-1642.
    1. Nishikawa H, Sakaguchi S. Regulatory T cells in tumor immunity. Int J Cancer. 2010;127:759–67.
    1. Chen Z, Xia D, Bi X, Saxena A, Sidhu N, El-Gayed A, Xiang J. Combined radiation therapy and dendritic cell vaccine for treating solid tumors with liver micro-metastasis. J Gene Med. 2005;7:506–17. doi: 10.1002/jgm.692.
    1. Chi KH, Liu SJ, Li CP, Kuo HP, Wang YS, Chao Y, Hsieh SL. Combination of conformal radiotherapy and intratumoral injection of adoptive dendritic cell immunotherapy in refractory hepatoma. J Immunother. 2005;28:129–35. doi: 10.1097/01.cji.0000154248.74383.5e.
    1. Liu WM, Meyer B, Dalgleish AG. How immunotherapy can enhance the response to other modalities and improve outcome and quality of life. J BUON. 2009;14(Suppl 1):S103–9.
    1. Moschella F, Proietti E, Capone I, Belardelli F. Combination strategies for enhancing the efficacy of immunotherapy in cancer patients. Ann N Y Acad Sci. 2010;1194:169–78. doi: 10.1111/j.1749-6632.2010.05464.x.
    1. Demaria S, Bhardwaj N, McBride WH, Formenti SC. Combining radiotherapy and immunotherapy: a revived partnership. Int J Radiat Oncol Biol Phys. 2005;63:655–66. doi: 10.1016/j.ijrobp.2005.06.032.
    1. Ferrara TA, Hodge JW, Gulley JL. Combining radiation and immunotherapy for synergistic antitumor therapy. Curr Opin Mol Ther. 2009;11:37–42.
    1. Friedman EJ. Immune modulation by ionizing radiation and its implications for cancer immunotherapy. Curr Pharm Des. 2002;8:1765–80. doi: 10.2174/1381612023394089.
    1. Hannani D, Sistigu A, Kepp O, Galluzzi L, Kroemer G, Zitvogel L. Prerequisites for the antitumor vaccine-like effect of chemotherapy and radiotherapy. Cancer J. 2011;17:351–8. doi: 10.1097/PPO.0b013e3182325d4d.
    1. Teitz-Tennenbaum S, Li Q, Okuyama R, Davis MA, Sun R, Whitfield J, Knibbs RN, Stoolman LM, Chang AE. Mechanisms involved in radiation enhancement of intratumoral dendritic cell therapy. J Immunother. 2008;31:345–58. doi: 10.1097/CJI.0b013e318163628c.
    1. Teitz-Tennenbaum S, Li Q, Davis MA, Wilder-Romans K, Hoff J, Li M, Chang AE. Radiotherapy combined with intratumoral dendritic cell vaccination enhances the therapeutic efficacy of adoptive T-cell transfer. J Immunother. 2009;32:602–12. doi: 10.1097/CJI.0b013e3181a95165.
    1. Hasumi KAY, Watanabe R, Hankey KG, Mann DL. Therapeutic response in patients with advanced malignancies treated with combined dendritic cell-activated T cell based immunotherapy and intensity-modulated radiotherapy. Cancers. 2011;3:2223–42. doi: 10.3390/cancers3022223.
    1. Hongeng S, Petvises S, Worapongpaiboon S, Rerkamnuaychoke B, Pakakasama S, Jootar S. Generation of CD3+ CD56+ cytokine-induced killer cells and their in vitro cytotoxicity against pediatric cancer cells. Int J Hematol. 2003;77:175–9. doi: 10.1007/BF02983217.
    1. Pievani A, Borleri G, Pende D, Moretta L, Rambaldi A, Golay J, Introna M. Dual-functional capability of CD3+CD56+ CIK cells, a T-cell subset that acquires NK function and retains TCR-mediated specific cytotoxicity. Blood. 2011;118:3301–10. doi: 10.1182/blood-2011-02-336321.
    1. Mann DL, Celluzzi CM, Hankey KG, Harris KM, Watanabe R, Hasumi K. Combining conventional therapies with intratumoral injection of autologous dendritic cells and activated T cells to treat patients with advanced cancers. Ann N Y Acad Sci. 2009;1174:41–50. doi: 10.1111/j.1749-6632.2009.04934.x.
    1. Domschke C, Schuetz F, Ge Y, Seibel T, Falk C, Brors B, Vlodavsky I, Sommerfeldt N, Sinn HP, Kühnle MC, et al. Intratumoral cytokines and tumor cell biology determine spontaneous breast cancer-specific immune responses and their correlation to prognosis. Cancer Res. 2009;69:8420–8. doi: 10.1158/0008-5472.CAN-09-1627.
    1. Ferrantini M, Capone I, Belardelli F. Dendritic cells and cytokines in immune rejection of cancer. Cytokine Growth Factor Rev. 2008;19:93–107. doi: 10.1016/j.cytogfr.2007.10.003.
    1. Franciszkiewicz K, Boissonnas A, Boutet M, Combadière C, Mami-Chouaib F. Role of chemokines and chemokine receptors in shaping the effector phase of the antitumor immune response. Cancer Res. 2012;72:6325–32. doi: 10.1158/0008-5472.CAN-12-2027.
    1. Skalova K, Mollova K, Michalek J. Human myeloid dendritic cells for cancer therapy: Does maturation matter? Vaccine. 2010;28:5153–60. doi: 10.1016/j.vaccine.2010.05.042.
    1. Harris KM, Lenz P, Hankey KG, MacVittie T, Farese A, Nakajima K, Hasumi K, Mann DL. Products of anti-CD3/anti-CD28 activated lymphocytes induce differentiation and maturation of dendritic cells and have adjuvant-like activity in vitro and in vivo. Clin Immunol. 2008;129:58–68. doi: 10.1016/j.clim.2008.06.003.
    1. Bhide SA, Nutting CM. Recent advances in radiotherapy. BMC Med. 2010;8:25. doi: 10.1186/1741-7015-8-25.
    1. Yovino S, Poppe M, Jabbour S, David V, Garofalo M, Pandya N, Alexander R, Hanna N, Regine WF. Intensity-modulated radiation therapy significantly improves acute gastrointestinal toxicity in pancreatic and ampullary cancers. Int J Radiat Oncol Biol Phys. 2011;79:158–62. doi: 10.1016/j.ijrobp.2009.10.043.
    1. Linn YC, Lau LC, Hui KM. Generation of cytokine-induced killer cells from leukaemic samples with in vitro cytotoxicity against autologous and allogeneic leukaemic blasts. Br J Haematol. 2002;116:78–86. doi: 10.1046/j.1365-2141.2002.03247.x.
    1. Wang FS, Liu MX, Zhang B, Shi M, Lei ZY, Sun WB, Du QY, Chen JM. Antitumor activities of human autologous cytokine-induced killer (CIK) cells against hepatocellular carcinoma cells in vitro and in vivo. World J Gastroenterol. 2002;8:464–8.
    1. Jäkel CE, Hauser S, Rogenhofer S, Müller SC, Brossart P, Schmidt-Wolf IG. Clinical studies applying cytokine-induced killer cells for the treatment of renal cell carcinoma. Clin Dev Immunol. 2012;2012:473245. doi: 10.1155/2012/473245.

Source: PubMed

3
Abonnere