Efficacy of intracellular immune checkpoint-silenced DC vaccine

Danhong Wang, Xue F Huang, Bangxing Hong, Xiao-Tong Song, Liangding Hu, Min Jiang, Bin Zhang, Hongmei Ning, Yuhang Li, Chen Xu, Xiao Lou, Botao Li, Zhiyong Yu, Jiangwei Hu, Jianlin Chen, Fan Yang, Haiyan Gao, Guoliang Ding, Lianming Liao, Lisa Rollins, Lindsey Jones, Si-Yi Chen, Hu Chen, Danhong Wang, Xue F Huang, Bangxing Hong, Xiao-Tong Song, Liangding Hu, Min Jiang, Bin Zhang, Hongmei Ning, Yuhang Li, Chen Xu, Xiao Lou, Botao Li, Zhiyong Yu, Jiangwei Hu, Jianlin Chen, Fan Yang, Haiyan Gao, Guoliang Ding, Lianming Liao, Lisa Rollins, Lindsey Jones, Si-Yi Chen, Hu Chen

Abstract

Background: DC-based tumor vaccines have had limited clinical success thus far. SOCS1, a key inhibitor of inflammatory cytokine signaling, is an immune checkpoint regulator that limits DC immunopotency.

Methods: We generated a genetically modified DC (gmDC) vaccine to perform immunotherapy. The adenovirus (Ad-siSSF) delivers two tumor-associated antigens (TAAs), survivin and MUC1; secretory bacterial flagellin for DC maturation; and an RNA interference moiety to suppress SOCS1. A 2-stage phase I trial was performed for patients with relapsed acute leukemia after allogenic hematopoietic stem cell transplantation: in stage 1, we compared the safety and efficacy between gmDC treatment (23 patients) and standard donor lymphocyte infusion (25 patients); in stage 2, we tested the efficacy of the gmDC vaccine for 12 acute myeloid leukemia (AML) patients with early molecular relapse.

Results: gmDCs elicited potent TAA-specific CTL responses in vitro, and the immunostimulatory activity of gmDC vaccination was demonstrated in rhesus monkeys. A stage 1 study established that this combinatory gmDC vaccine is safe in acute leukemia patients and yielded improved survival rate. In stage 2, we observed a complete remission rate of 83% in 12 relapsed AML patients. Overall, no grade 3 or grade 4 graft-versus-host disease incidence was detected in any of the 35 patients enrolled.

Conclusions: This study, with combinatory modifications in DCs, demonstrates the safety and efficacy of SOCS1-silenced DCs in treating relapsed acute leukemia.

Trial registration: ClinicalTrials.gov NCT01956630.

Funding: National Institute of Health (R01CA90427); the Key New Drug Development and Manufacturing Program of the "Twelfth Five-Year Plan" of China (2011ZX09102-001-29); and Clinical Application Research of Beijing (Z131107002213148).

Keywords: Dendritic cells; Hematology; Immunology; Immunotherapy; Leukemias.

Conflict of interest statement

Conflict of interest: The authors have declared that no conflict of interest exists.

Figures

Figure 1. Enhanced maturation of Ad-siSSF–transduced hDCs.
Figure 1. Enhanced maturation of Ad-siSSF–transduced hDCs.
(A) Expression levels of a panel of surface markers on human PBMC DCs at 48 hours after transduction with Ad-siSSF, control Ad-GFP virus (MOI = 25,000 vp), or PBS-treated (mock) (isotype controls: brown line). With the exception of OX40L and TLR5, all surface markers are upregulated after Ad-siSSF transduction compared with mock or Ad-GFP controls. (B) The levels of representative proinflammatory cytokines and chemokines in the culture media of human PBMC DCs 48 hours after transduction with different adenoviruses at a MOI of 25,000 vp, as analyzed by ELISA. The hDCs have significantly enhanced levels of these cytokines and chemokines after Ad-siSSF transduction compared with controls. (C) Migration rates of transduced human PBMC DCs in response to recombinant CCL21 (100 ng/ml) show the enhanced migratory activity of Ad-siSSF–transduced DCs compared with controls. Results are presented from 1 of 3 repeated assays (mean ± SEM). *P < 0.05, Ad-siSSF vs. Ad-SM DCs, as determined by Student’s 2-tailed t test.
Figure 2. Enhanced priming of TAA-specific T…
Figure 2. Enhanced priming of TAA-specific T cells by Ad-siSSF-DCs and enhanced TAA-specific CTL cytolytic activity against human tumor cells.
Human autologous T cells were cocultured with Ad-transduced DCs or PBS-treated hDCs (mock) (20:1) for 2 weeks. (A) Intracellular IFNG staining of cocultured CD4+ and CD8+ T cells. FACS analysis shows an increase of IFNG expression after coculture with Ad-siSSF-DCs compared with controls. (B) Increased IFNG-producing T cells after coculture with Ad-siSSF-DCs compared with coculture with controls is shown by ELISpot. Representative data from 1 of 5 HLA-A2+ donors are shown. (C) Cytolytic activities of HLA-A2+ T cells against various human tumor cell lines after 2 weeks of in vitro sensitization with different adenovirus-transfected autologous DCs were determined by standard 51Cr release assays. T cells sensitized by Ad-siSSF-DCs exhibited increased killing compared with those sensitized by controls. Cytolytic percentages are presented from 1 of 3 repeated experiments. Error bars represent mean ± SEM. *P < 0.05, Ad-siSSF vs. Ad-SM; **P < 0.01, Ad-siSSF vs. Ad-GFP or mock, as determined by Student’s 2-tailed t test.
Figure 3. ELISpot assay of immune responses…
Figure 3. ELISpot assay of immune responses induced by a single injection of Ad-siSSF vector in monkeys.
Groups of monkeys (n = 3 per group) were injected with Ad-siSSF or control PBS, and toxicities and immune responses were measured 14 days later. Frequencies of antigen-specific T cells against MUC1 (hMUC1 20mer), survivin (hSur protein), and against the MUC1-survivin fusion protein (hMUC1+Sur) were significantly increased in monkeys given Ad-siSSF compared with the mock group (PBS). P < 0.00001 for responses against designated peptides in the Ad-siSSF group vs. mock group, as determined by Student’s 2-tailed t test.
Figure 4. Consolidated Standards of Reporting Trials…
Figure 4. Consolidated Standards of Reporting Trials diagram.
Figure 5. Kaplan-Meier estimates of overall survival.
Figure 5. Kaplan-Meier estimates of overall survival.
The 3-year probability of OS was significantly higher in the Ad-siSSF-DC group (n = 23) than in the DLI group (n = 25) (48.9 % vs. 27.5%, respectively). P = 0.028, as determined by log-rank test.
Figure 6. Comparing WT1 expression in AL…
Figure 6. Comparing WT1 expression in AL patients before and after treatment.
For the majority of the patients (n = 12), WT1 expression decreased drastically after infusion of Ad-siSSF-DCs. Across all patients, the pretreatment mean of WT1 copies was 1.92 per 1,000 ABL copies versus 0.18 WT1 copies detected after treatment. The complete remission rate was 83%. The GVHD rate was 50%, and no cases of grade 3 or grade 4 GVHD were observed.

References

    1. Henkes M, van der Kuip H, Aulitzky WE. Therapeutic options for chronic myeloid leukemia: focus on imatinib (Glivec, Gleevectrade mark) Ther Clin Risk Manag. 2008;4(1):163–187.
    1. Van Tendeloo VF, et al. Induction of complete and molecular remissions in acute myeloid leukemia by Wilms’ tumor 1 antigen-targeted dendritic cell vaccination. Proc Natl Acad Sci USA. 2010;107(31):13824–13829. doi: 10.1073/pnas.1008051107.
    1. Ravandi F, Burnett AK, Agura ED, Kantarjian HM. Progress in the treatment of acute myeloid leukemia. Cancer. 2007;110(9):1900–1910. doi: 10.1002/cncr.23000.
    1. Jabbour EJ, Estey E, Kantarjian HM. Adult acute myeloid leukemia. Mayo Clin Proc. 2006;81(2):247–260. doi: 10.4065/81.2.247.
    1. Deol A, Lum LG. Role of donor lymphocyte infusions in relapsed hematological malignancies after stem cell transplantation revisited. Cancer Treat Rev. 2010;36(7):528–538. doi: 10.1016/j.ctrv.2010.03.004.
    1. Sekeres MA. Treatment of older adults with acute myeloid leukemia: state of the art and current perspectives. Haematologica. 2008;93(12):1769–1772. doi: 10.3324/haematol.2008.000497.
    1. Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392(6673):245–252. doi: 10.1038/32588.
    1. Gilboa E. DC-based cancer vaccines. J Clin Invest. 2007;117(5):1195–1203. doi: 10.1172/JCI31205.
    1. Rosenberg SA, et al. Immunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients with metastatic melanoma. Nat Med. 1998;4(3):321–327. doi: 10.1038/nm0398-321.
    1. Song W, et al. Dendritic cells genetically modified with an adenovirus vector encoding the cDNA for a model antigen induce protective and therapeutic antitumor immunity. J Exp Med. 1997;186(8):1247–1256. doi: 10.1084/jem.186.8.1247.
    1. Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nat Med. 2004;10(9):909–915. doi: 10.1038/nm1100.
    1. Lesterhuis WJ, Haanen JB, Punt CJ. Cancer immunotherapy--revisited. Nat Rev Drug Discov. 2011;10(8):591–600. doi: 10.1038/nrd3500.
    1. Small EJ, et al. 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(19):3089–3094. doi: 10.1200/JCO.2005.04.5252.
    1. Evel-Kabler K, Chen SY. Dendritic cell-based tumor vaccines and antigen presentation attenuators. Mol Ther. 2006;13(5):850–858. doi: 10.1016/j.ymthe.2006.02.009.
    1. Liew FY, Xu D, Brint EK, O’Neill LA. Negative regulation of toll-like receptor-mediated immune responses. Nat Rev Immunol. 2005;5(6):446–458. doi: 10.1038/nri1630.
    1. Shen L, Evel-Kabler K, Strube R, Chen SY. Silencing of SOCS1 enhances antigen presentation by dendritic cells and antigen-specific anti-tumor immunity. Nat Biotechnol. 2004;22(12):1546–1553. doi: 10.1038/nbt1035.
    1. Evel-Kabler K, Song XT, Aldrich M, Huang XF, Chen SY. SOCS1 restricts dendritic cells’ ability to break self tolerance and induce antitumor immunity by regulating IL-12 production and signaling. J Clin Invest. 2006;116(1):90–100. doi: 10.1172/JCI26169.
    1. Kubo M, Hanada T, Yoshimura A. Suppressors of cytokine signaling and immunity. Nat Immunol. 2003;4(12):1169–1176. doi: 10.1038/ni1012.
    1. Alexander WS, Hilton DJ. The role of suppressors of cytokine signaling (SOCS) proteins in regulation of the immune response. Annu Rev Immunol. 2004;22:503–529. doi: 10.1146/annurev.immunol.22.091003.090312.
    1. Mansell A, et al. Suppressor of cytokine signaling 1 negatively regulates Toll-like receptor signaling by mediating Mal degradation. Nat Immunol. 2006;7(2):148–155. doi: 10.1038/ni1299.
    1. Hong B, et al. A super TLR agonist to improve efficacy of dendritic cell vaccine in induction of anti-HCV immunity. PLoS One. 2012;7(11):e48614. doi: 10.1371/journal.pone.0048614.
    1. Hayashi F, et al. The innate immune response to bacterial flagellin is mediated by Toll-like receptor 5. Nature. 2001;410(6832):1099–1103. doi: 10.1038/35074106.
    1. Tsujimoto H, et al. Flagellin enhances NK cell proliferation and activation directly and through dendritic cell-NK cell interactions. J Leukoc Biol. 2005;78(4):888–897. doi: 10.1189/jlb.0105051.
    1. Caron G, et al. Direct stimulation of human T cells via TLR5 and TLR7/8: flagellin and R-848 up-regulate proliferation and IFN-gamma production by memory CD4+ T cells. J Immunol. 2005;175(3):1551–1557. doi: 10.4049/jimmunol.175.3.1551.
    1. Gelman AE, Zhang J, Choi Y, Turka LA. Toll-like receptor ligands directly promote activated CD4+ T cell survival. J Immunol. 2004;172(10):6065–6073. doi: 10.4049/jimmunol.172.10.6065.
    1. Means TK, Hayashi F, Smith KD, Aderem A, Luster AD. The Toll-like receptor 5 stimulus bacterial flagellin induces maturation and chemokine production in human dendritic cells. J Immunol. 2003;170(10):5165–5175. doi: 10.4049/jimmunol.170.10.5165.
    1. Didierlaurent A, et al. Flagellin promotes myeloid differentiation factor 88-dependent development of Th2-type response. J Immunol. 2004;172(11):6922–6930. doi: 10.4049/jimmunol.172.11.6922.
    1. Cunningham AF, et al. Responses to the soluble flagellar protein FliC are Th2, while those to FliC on Salmonella are Th1. Eur J Immunol. 2004;34(11):2986–2995. doi: 10.1002/eji.200425403.
    1. McSorley SJ, Ehst BD, Yu Y, Gewirtz AT. Bacterial flagellin is an effective adjuvant for CD4+ T cells in vivo. J Immunol. 2002;169(7):3914–3919. doi: 10.4049/jimmunol.169.7.3914.
    1. Strindelius L, Filler M, Sjöholm I. Mucosal immunization with purified flagellin from Salmonella induces systemic and mucosal immune responses in C3H/HeJ mice. Vaccine. 2004;22(27-28):3797–3808. doi: 10.1016/j.vaccine.2003.12.035.
    1. Huleatt JW, et al. Vaccination with recombinant fusion proteins incorporating Toll-like receptor ligands induces rapid cellular and humoral immunity. Vaccine. 2007;25(4):763–775. doi: 10.1016/j.vaccine.2006.08.013.
    1. Cuadros C, Lopez-Hernandez FJ, Dominguez AL, McClelland M, Lustgarten J. Flagellin fusion proteins as adjuvants or vaccines induce specific immune responses. Infect Immun. 2004;72(5):2810–2816. doi: 10.1128/IAI.72.5.2810-2816.2004.
    1. Lee SE, et al. A bacterial flagellin, Vibrio vulnificus FlaB, has a strong mucosal adjuvant activity to induce protective immunity. Infect Immun. 2006;74(1):694–702. doi: 10.1128/IAI.74.1.694-702.2006.
    1. Applequist SE, et al. Activation of innate immunity, inflammation, and potentiation of DNA vaccination through mammalian expression of the TLR5 agonist flagellin. J Immunol. 2005;175(6):3882–3891. doi: 10.4049/jimmunol.175.6.3882.
    1. Tamm I, et al. IAP-family protein survivin inhibits caspase activity and apoptosis induced by Fas (CD95), Bax, caspases, and anticancer drugs. Cancer Res. 1998;58(23):5315–5320.
    1. Velculescu VE, et al. Analysis of human transcriptomes. Nat Genet. 1999;23(4):387–388.
    1. Chawla-Sarkar M, Bae SI, Reu FJ, Jacobs BS, Lindner DJ, Borden EC. Downregulation of Bcl-2, FLIP or IAPs (XIAP and survivin) by siRNAs sensitizes resistant melanoma cells to Apo2L/TRAIL-induced apoptosis. Cell Death Differ. 2004;11(8):915–923. doi: 10.1038/sj.cdd.4401416.
    1. Kappler M, et al. Elevated expression level of survivin protein in soft-tissue sarcomas is a strong independent predictor of survival. Clin Cancer Res. 2003;9(3):1098–1104.
    1. Kim HJ, Choi EJ, Sohn HJ, Park SH, Min WS, Kim TG. Combinatorial molecular marker assays of WT1, survivin, and TERT at initial diagnosis of adult acute myeloid leukemia. Eur J Haematol. 2013;91(5):411–422. doi: 10.1111/ejh.12167.
    1. Altieri DC. The molecular basis and potential role of survivin in cancer diagnosis and therapy. Trends Mol Med. 2001;7(12):542–547. doi: 10.1016/S1471-4914(01)02243-2.
    1. Gendler SJ. MUC1, the renaissance molecule. J Mammary Gland Biol Neoplasia. 2001;6(3):339–353. doi: 10.1023/A:1011379725811.
    1. Li GW, et al. [Expression of MUC1 gene and MDR1 gene in non-M3 subtype acute leukemia and their correlations to clinical treatment efficacy] Ai Zheng. 2005;24(8):1011–1014.
    1. Randolph GJ, Angeli V, Swartz MA. Dendritic-cell trafficking to lymph nodes through lymphatic vessels. Nat Rev Immunol. 2005;5(8):617–628. doi: 10.1038/nri1670.
    1. Yan L, Woodham AW, Da Silva DM, Kast WM. Functional analysis of HPV-like particle-activated Langerhans cells in vitro. Methods Mol Biol. 2015;1249:333–350. doi: 10.1007/978-1-4939-2013-6_25.
    1. Cilloni D, et al. Real-time quantitative polymerase chain reaction detection of minimal residual disease by standardized WT1 assay to enhance risk stratification in acute myeloid leukemia: a European LeukemiaNet study. J Clin Oncol. 2009;27(31):5195–5201. doi: 10.1200/JCO.2009.22.4865.
    1. Marine JC, et al. SOCS1 deficiency causes a lymphocyte-dependent perinatal lethality. Cell. 1999;98(5):609–616. doi: 10.1016/S0092-8674(00)80048-3.
    1. Alexander WS, et al. SOCS1 is a critical inhibitor of interferon gamma signaling and prevents the potentially fatal neonatal actions of this cytokine. Cell. 1999;98(5):597–608.
    1. Metcalf D, Mifsud S, Di Rago L, Alexander WS. The lethal effects of transplantation of Socs1-/- bone marrow cells into irradiated adult syngeneic recipients. Proc Natl Acad Sci USA. 2003;100(14):8436–8441. doi: 10.1073/pnas.1032925100.
    1. Janeway CA, Medzhitov R. Innate immune recognition. Annu Rev Immunol. 2002;20:197–216. doi: 10.1146/annurev.immunol.20.083001.084359.
    1. Beutler B, Rietschel ET. Innate immune sensing and its roots: the story of endotoxin. Nat Rev Immunol. 2003;3(2):169–176. doi: 10.1038/nri1004.
    1. Akira S, Takeda K. Toll-like receptor signalling. Nat Rev Immunol. 2004;4(7):499–511. doi: 10.1038/nri1391.
    1. Altieri DC. Validating survivin as a cancer therapeutic target. Nat Rev Cancer. 2003;3(1):46–54. doi: 10.1038/nrc968.
    1. Mori A, Wada H, Nishimura Y, Okamoto T, Takemoto Y, Kakishita E. Expression of the antiapoptosis gene survivin in human leukemia. Int J Hematol. 2002;75(2):161–165. doi: 10.1007/BF02982021.
    1. Schmidt SM, et al. Survivin is a shared tumor-associated antigen expressed in a broad variety of malignancies and recognized by specific cytotoxic T cells. Blood. 2003;102(2):571–576. doi: 10.1182/blood-2002-08-2554.
    1. Charalambous A, Oks M, Nchinda G, Yamazaki S, Steinman RM. Dendritic cell targeting of survivin protein in a xenogeneic form elicits strong CD4+ T cell immunity to mouse survivin. J Immunol. 2006;177(12):8410–8421. doi: 10.4049/jimmunol.177.12.8410.
    1. Idenoue S, et al. A potent immunogenic general cancer vaccine that targets survivin, an inhibitor of apoptosis proteins. Clin Cancer Res. 2005;11(4):1474–1482. doi: 10.1158/1078-0432.CCR-03-0817.
    1. Vlad AM, Kettel JC, Alajez NM, Carlos CA, Finn OJ. MUC1 immunobiology: from discovery to clinical applications. Adv Immunol. 2004;82:249–293. doi: 10.1016/S0065-2776(04)82006-6.
    1. Brossart P, et al. The epithelial tumor antigen MUC1 is expressed in hematological malignancies and is recognized by MUC1-specific cytotoxic T-lymphocytes. Cancer Res. 2001;61(18):6846–6850.
    1. Tsang KY, Palena C, Gulley J, Arlen P, Schlom J. A human cytotoxic T-lymphocyte epitope and its agonist epitope from the nonvariable number of tandem repeat sequence of MUC-1. Clin Cancer Res. 2004;10(6):2139–2149. doi: 10.1158/1078-0432.CCR-1011-03.
    1. Barnd DL, Lan MS, Metzgar RS, Finn OJ. Specific, major histocompatibility complex-unrestricted recognition of tumor-associated mucins by human cytotoxic T cells. Proc Natl Acad Sci USA. 1989;86(18):7159–7163. doi: 10.1073/pnas.86.18.7159.
    1. Agrawal B, Reddish MA, Longenecker BM. In vitro induction of MUC-1 peptide-specific type 1 T lymphocyte and cytotoxic T lymphocyte responses from healthy multiparous donors. J Immunol. 1996;157(5):2089–2095.
    1. Turner MS, Cohen PA, Finn OJ. Lack of effective MUC1 tumor antigen-specific immunity in MUC1-transgenic mice results from a Th/T regulatory cell imbalance that can be corrected by adoptive transfer of wild-type Th cells. J Immunol. 2007;178(5):2787–2793. doi: 10.4049/jimmunol.178.5.2787.
    1. Ramanathan RK, et al. Phase I study of a MUC1 vaccine composed of different doses of MUC1 peptide with SB-AS2 adjuvant in resected and locally advanced pancreatic cancer. Cancer Immunol Immunother. 2005;54(3):254–264. doi: 10.1007/s00262-004-0581-1.
    1. Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer. 2012;12(4):265–277. doi: 10.1038/nrc3258.
    1. Weissman D, et al. HIV gag mRNA transfection of dendritic cells (DC) delivers encoded antigen to MHC class I and II molecules, causes DC maturation, and induces a potent human in vitro primary immune response. J Immunol. 2000;165(8):4710–4717. doi: 10.4049/jimmunol.165.8.4710.
    1. zum Büschenfelde CM, Metzger J, Hermann C, Nicklisch N, Peschel C, Bernhard H. The generation of both T killer and Th cell clones specific for the tumor-associated antigen HER2 using retrovirally transduced dendritic cells. J Immunol. 2001;167(3):1712–1719. doi: 10.4049/jimmunol.167.3.1712.
    1. Ferrara F, Palmieri S, Mele G. Prognostic factors and therapeutic options for relapsed or refractory acute myeloid leukemia. Haematologica. 2004;89(8):998–1008.
    1. Liu L, et al. Randomized study of autologous cytokine-induced killer cell immunotherapy in metastatic renal carcinoma. Clin Cancer Res. 2012;18(6):1751–1759. doi: 10.1158/1078-0432.CCR-11-2442.
    1. Przepiorka D, et al. 1994 Consensus conference on acute GVHD grading. Bone Marrow Transplant. 1995;15(6):825–828.
    1. Ren W, Strube R, Zhang X, Chen SY, Huang XF. Potent tumor-specific immunity induced by an in vivo heat shock protein-suicide gene-based tumor vaccine. Cancer Res. 2004;64(18):6645–6651. doi: 10.1158/0008-5472.CAN-04-1084.

Source: PubMed

3
Subskrybuj