Single dose denileukin diftitox does not enhance vaccine-induced T cell responses or effectively deplete Tregs in advanced melanoma: immune monitoring and clinical results of a randomized phase II trial

Jason J Luke, Yuanyuan Zha, Karen Matijevich, Thomas F Gajewski, Jason J Luke, Yuanyuan Zha, Karen Matijevich, Thomas F Gajewski

Abstract

Background: Depletion of CD25(+) Tregs improves anti-tumor immunity in preclinical models. Denileukin diftitox is a recombinant fusion protein of human IL-2 and diptheria toxin fragment that also can kill CD25(+) T cells. Prior clinical trials of denileukin diftitox suggested reduction of FoxP3(+) Tregs and some clinical responses.

Method: To investigate the immunologic effects of denileukin difitox on vaccine-specific immune responses in melanoma, a randomized clinical trial of single dose denileukin diftitox prior to vaccination versus vaccination alone in subjects with HLA-A2(+) metastatic melanoma was performed. Treatment included randomization to a 4-peptide vaccine (Melan-A, gp100, MAGE3 and NA17 with GM-CSF emulsified in Montanide) alone or after single dose of denileukin diftitox (18 mcg/kg). Vaccine was given every 2 weeks for 3 doses and, absent clinical progression, continued every 2 weeks. Blood and tumor biopsies were obtained pretreatment and after 3 vaccinations for immunologic assessments.

Results: In 17 treated subjects there were no drug-related G3-4 adverse events. One partial response and 8 stable disease were observed in 9 subjects (4 DD: 5 vaccine only) with no impact of denileukin diftitox on time to progression. Total peripheral Tregs were not significantly altered, and in 1 patient biopsy intra-tumoral FoxP3 transcripts were not reduced following denileukin diftitox. ELISA for IL2R-α demonstrated no impact on outcomes by soluble CD25 level. Immune monitoring suggested the development of modest vaccine-specific CD8(+) T cell responses in the control group, however immunization efficacy was actually reduced in the denileukin diftitox group.

Conclusion: Our results indicate that denileukin diftitox did not effectively deplete Tregs, augment T cell responses, or improve clinical activity in melanoma. Clinicaltrials.gov ID: NCT00515528; Registered August 9, 2007.

Keywords: Denileukin difitox; Immunotherapy; Melanoma; Tregs; Vaccine.

Figures

Fig. 1
Fig. 1
Representative flow cytometric gating for CD4+CD25+FoxP3+ cells. Peripheral blood mononuclear cells were isolated using density gradient centrifugation before and after patient received denileukin diftitox treatment versus no treatment. Cells were labeled with anti-CD4, anti-CD25, and anti-FoxP3 Abs from the Treg staining kit (Miltenyi Biotech, Inc.) and then analyzed using a MACSQuant analyzer
Fig. 2
Fig. 2
CD4+CD25hiFoxP3+ Treg in peripheral blood before and after denileukin diftitox treatment. Peripheral blood mononuclear cells were isolated using density gradient centrifugation before and after patient received denileukin diftitox treatment versus no treatment. In the denileukin difitox arm, peripheral blood mononuclear cells were isolated at the time of consent (pre) and before the first vaccination (post). Tregs were first labeled with anti-CD4, anti-CD25, and anti-FoxP3 Abs from the Treg staining kit (Miltenyi Biotech, Inc.) and then analyzed using a MACSQuant analyzer. Paired student t-test was used to determine whether there was a statistical difference between the pre- and post-treatment samples in each cohort
Fig. 3
Fig. 3
Intratumoral FoxP3 mRNA transcripts before and after treatment with and without denileukin diftitox. Fresh biopsies was collected before and after treatment when feasible and snap frozen in liquid nitrogen. RNA samples were isolated using the Trizol regent, and cDNA was synthesized using the High Fidelity cDNA Synthesis Kit (Roche Applied Science). qRT-PCR was then performed to analyze the FoxP3 transcripts levels on ABI-7300 qPCR instrument (Applied Biosystem). β-actin were used as an internal control
Fig. 4
Fig. 4
Soluble interleukin-2 receptor-α in patients treated or untreated with denileukin diftitox. Blood was drawn before and after subjects were treated on the trial. Serum was aliquoted and stored at -80 °C until analysis. Soluble interleukin-2 receptor (soluble CD25) levels were analyzed by ELISA. Student t-test (assuming different variance in each group) was used to determine whether there was a statistical difference between the pre and post samples in each cohort and between the two cohorts
Fig. 5
Fig. 5
Circulating antigen-specific CD8+ T cells in patients treated or untreated with denileukin diftitox. Peripheral blood mononuclear cells were seeded on anti-IFN-γ Ab-coated ELISPOT plates. 5 μM of peptide was then added to each well and incubated overnight. The plates were then incubated with the biotin-labeled anti-IFN-γ Ab and developed according to manufacturer’s procedure. The plates were read and analyzed on CTL-ImmunoSpot S6 Core Analyzer from Cellular Technology Ltd. Each point represents the numerical difference of Ag-specific CD8+ T cells in PBMCs post-vaccine and pre-treatment for each patient. Each point represents an individual patient. Paired student t-test was used to determine whether there was a difference between the pre- and post- samples in each cohort

References

    1. Valmori D, Dutoit V, Ayyoub M, et al. Simultaneous CD8+ T cell responses to multiple tumor antigen epitopes in a multipeptide melanoma vaccine. Cancer Immun. 2003;3:15.
    1. Gajewski TF, Fallarino F, Ashikari A, Sherman M. Immunization of HLA-A2+ melanoma patients with MAGE-3 or MelanA peptide-pulsed autologous peripheral blood mononuclear cells plus recombinant human interleukin 12. Clin Cancer Res. 2001;7:895s–901.
    1. Ahlers JD, Dunlop N, Alling DW, Nara PL, Berzofsky JA. Cytokine-in-adjuvant steering of the immune response phenotype to HIV-1 vaccine constructs: granulocyte-macrophage colony-stimulating factor and TNF-alpha synergize with IL-12 to enhance induction of cytotoxic T lymphocytes. J Immunol. 1997;158:3947–58.
    1. Slingluff CL, Jr, Petroni GR, Yamshchikov GV, et al. Clinical and immunologic results of a randomized phase II trial of vaccination using four melanoma peptides either administered in granulocyte-macrophage colony-stimulating factor in adjuvant or pulsed on dendritic cells. J Clin Oncol. 2003;21:4016–26. doi: 10.1200/JCO.2003.10.005.
    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;163:5211–8.
    1. Viguier M, Lemaitre F, Verola O, et al. Foxp3 expressing CD4 + CD25(high) regulatory T cells are overrepresented in human metastatic melanoma lymph nodes and inhibit the function of infiltrating T cells. J Immunol. 2004;173:1444–53. doi: 10.4049/jimmunol.173.2.1444.
    1. Curiel TJ, Coukos G, Zou L, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med. 2004;10:942–9. doi: 10.1038/nm1093.
    1. Kuzel TM. DAB389IL-2 (Denileukin Diftitox, ONTAK): Review of Clinical Trials to Date. Clinical Lymphoma. 2000;1(Supplement 1):S33–6. doi: 10.3816/CLM.2000.s.006.
    1. Foss FM. Interleukin-2 fusion toxin: targeted therapy for cutaneous T cell lymphoma. Ann N Y Acad Sci. 2001;941:166–76. doi: 10.1111/j.1749-6632.2001.tb03720.x.
    1. Dannull J, Su Z, Rizzieri D, et al. Enhancement of vaccine-mediated antitumor immunity in cancer patients after depletion of regulatory T cells. J Clin Invest. 2005;115:3623–33. doi: 10.1172/JCI25947.
    1. Attia P, Maker AV, Haworth LR, Rogers-Freezer L, Rosenberg SA. Inability of a fusion protein of IL-2 and diphtheria toxin (Denileukin Diftitox, DAB389IL-2, ONTAK) to eliminate regulatory T lymphocytes in patients with melanoma. J Immunother. 2005;28:582–92. doi: 10.1097/01.cji.0000175468.19742.10.
    1. Spranger S, Spaapen RM, Zha Y, et al. Up-regulation of PD-L1, IDO, and T(regs) in the melanoma tumor microenvironment is driven by CD8(+) T cells. Sci Transl Med. 2013;5:200ra116. doi: 10.1126/scitranslmed.3006504.
    1. Peterson AC, Harlin H, Gajewski TF. Immunization with Melan-A peptide-pulsed peripheral blood mononuclear cells plus recombinant human interleukin-12 induces clinical activity and T-cell responses in advanced melanoma. J Clin Oncol. 2003;21:2342–8. doi: 10.1200/JCO.2003.12.144.
    1. Geynisman DM, Zha Y, Kunnavakkam R, et al. A randomized pilot phase I study of modified carcinoembryonic antigen (CEA) peptide (CAP1-6D)/montanide/GM-CSF-vaccine in patients with pancreatic adenocarcinoma. J Immunother Cancer. 2013;1:8. doi: 10.1186/2051-1426-1-8.
    1. Kline J, Brown IE, Zha YY, et al. Homeostatic proliferation plus regulatory T-cell depletion promotes potent rejection of B16 melanoma. Clin Cancer Res. 2008;14:3156–67. doi: 10.1158/1078-0432.CCR-07-4696.
    1. Sutmuller RP, van Duivenvoorde LM, van Elsas A, et al. Synergism of cytotoxic T lymphocyte-associated antigen 4 blockade and depletion of CD25(+) regulatory T cells in antitumor therapy reveals alternative pathways for suppression of autoreactive cytotoxic T lymphocyte responses. J Exp Med. 2001;194:823–32. doi: 10.1084/jem.194.6.823.
    1. Ko K, Yamazaki S, Nakamura K, et al. Treatment of advanced tumors with agonistic anti-GITR mAb and its effects on tumor-infiltrating Foxp3 + CD25 + CD4+ regulatory T cells. J Exp Med. 2005;202:885–91. doi: 10.1084/jem.20050940.
    1. Telang S, Rasku MA, Clem AL, et al. Phase II trial of the regulatory T cell-depleting agent, denileukin diftitox, in patients with unresectable stage IV melanoma. BMC Cancer. 2011;11:515. doi: 10.1186/1471-2407-11-515.
    1. Powell DJ, Jr, Felipe-Silva A, Merino MJ, et al. Administration of a CD25-directed immunotoxin, LMB-2, to patients with metastatic melanoma induces a selective partial reduction in regulatory T cells in vivo. J Immunol. 2007;179:4919–28. doi: 10.4049/jimmunol.179.7.4919.
    1. Powell DJ, Jr, Attia P, Ghetie V, Schindler J, Vitetta ES, Rosenberg SA. Partial reduction of human FOXP3+ CD4 T cells in vivo after CD25-directed recombinant immunotoxin administration. J Immunother. 2008;31:189–98. doi: 10.1097/CJI.0b013e31815dc0e8.
    1. Rech AJ, Mick R, Martin S, et al. CD25 blockade depletes and selectively reprograms regulatory T cells in concert with immunotherapy in cancer patients. Sci Transl Med. 2012;4:134ra62. doi: 10.1126/scitranslmed.3003330.
    1. Rech AJ, Vonderheide RH. Clinical use of anti-CD25 antibody daclizumab to enhance immune responses to tumor antigen vaccination by targeting regulatory T cells. Ann N Y Acad Sci. 2009;1174:99–106. doi: 10.1111/j.1749-6632.2009.04939.x.
    1. Ogura M, Ishida T, Hatake K, et al. Multicenter phase II study of mogamulizumab (KW-0761), a defucosylated anti-cc chemokine receptor 4 antibody, in patients with relapsed peripheral T-cell lymphoma and cutaneous T-cell lymphoma. J Clin Oncol. 2014;32:1157–63. doi: 10.1200/JCO.2013.52.0924.

Source: PubMed

3
Abonnere