Clinical study of a survivin long peptide vaccine (SurVaxM) in patients with recurrent malignant glioma

Robert A Fenstermaker, Michael J Ciesielski, Jingxin Qiu, Nuo Yang, Cheryl L Frank, Kelvin P Lee, Laszlo R Mechtler, Ahmed Belal, Manmeet S Ahluwalia, Alan D Hutson, Robert A Fenstermaker, Michael J Ciesielski, Jingxin Qiu, Nuo Yang, Cheryl L Frank, Kelvin P Lee, Laszlo R Mechtler, Ahmed Belal, Manmeet S Ahluwalia, Alan D Hutson

Abstract

Survivin is an anti-apoptotic protein that is highly expressed in many cancers, including malignant gliomas. Preclinical studies established that the conjugated survivin peptide mimic SurVaxM (SVN53-67/M57-KLH) could stimulate an anti-tumor immune response against murine glioma in vivo, as well as human glioma cells ex vivo. The current clinical study was conducted to test safety, immunogenicity and clinical effects of the vaccine. Recurrent malignant glioma patients whose tumors were survivin-positive, and who had either HLA-A*02 or HLA-A*03 MHC class I allele-positivity, were given subcutaneous injections of SurVaxM (500 μg) in Montanide ISA 51 with sargramostim (100 μg) at 2-week intervals. SurVaxM was well tolerated with mostly grade one adverse events (AE) and no serious adverse events (SAE) attributable to the study drug. Six patients experienced local injection site reactions; three patients reported fatigue (grades 1 and 2), and 2 patients experienced myalgia (grade 1). Six of eight immunologically evaluable patients developed both cellular and humoral immune responses to vaccine. The vaccine also stimulated HLA-A*02, HLA-A*03 and HLA-A*24 restricted T cell responses. Three patients maintained a partial clinical response or stable disease for more than 6 months. Median progression-free survival was 17.6 weeks, and median overall survival was 86.6 weeks from study entry with seven of nine patients surviving more than 12 months.

Keywords: Apoptosis; Glioma; Immunotherapy; Peptide; Survivin; Vaccine.

Conflict of interest statement

R.A. Fenstermaker and M.J. Ciesielski are co-inventors on patents regarding SVN53-67/M57-KLH and are co-founders of MimiVax, LLC, which has licensed such patents from Roswell Park Cancer Institute. No additional potential conflicts of interest were disclosed by the other authors.

Figures

Fig. 1
Fig. 1
Long survivin peptide contained in SVN53-67/M57-KLH is shown with brackets indicating empirically confirmed immuno-reactive HLA-A*02, HLA-A*03, HLA-A*11 and HLA-A*24 epitopes within it. The position of the cysteine-to-methionine substitution (mimic) within the peptide is indicated (dark gray)
Fig. 2
Fig. 2
a FACS analysis of representative MHC class I dextramer assays of PBMC from a HLA-A*02/24 patient. Multimer binding of CD8+ T cells specific for survivin epitopes Pep1 and Pep6 (see Fig. 1) is shown. The upper right quadrant of each panel shows survivin multimer +/CD8+ gated T cells. Week 0 is pre-immunization blood sampling. Control represents results with a nonsense dextramer. b Binding of multimers to CD8+ T cell receptors in patients measured at weeks 8–150 following study entry. Data acquisition was performed by FACS analysis as revealed in a. Patient samples registering ≥1 % over baseline survivin-specific CD8+ T cells are shown as (filled circle). Patients not mounting an immune response to a particular peptide are shown as (open circle). Weakly positive patients registering ≥0.75 % and <1 % above baseline are (circled times). Custom-designed multimers included: Pep1, HLA-A*0201: QMFFCFKEL; Pep2, HLA-A*0201: AQMFFCFKEL; Pep3, HLA-A*0301: DLAQMFFCFK; Pep4, HLA-A*0301: LAQMFFCFK; Pep5, HLA-A*0301: AQMFFCFK; and Pep6; HLA-A*24: DLAQMFFCF. A negative control nonsense multimer was used to assess nonspecific binding. MHC Dextramer-PE (Immudex, Copenhagen, Denmark) to A*0201:QMFFCFKEL; A*0301:AQMFFCFK; A*0301:DLAQMFFCFK; A*0301:LAQMFFCFK; as well as iTag-PE Tetramer (BD Coulter) to A*0201:AQMFFCFKEL; and A*24:DLAQMFFCF
Fig. 3
Fig. 3
Antibodies produced in response to vaccination were measured as a potential biomarker. a IgG antibodies to SVN53-67/M57, and b IgG antibodies to SVN53-67 were measured in the serum of each patient by ELISA following prime-boost doses, and in those patients who entered an extended dosing phase. Seven patients produced significant levels (>1 O.D.) of SVN53-67/M57 (a) and six patients developed high titers (>1 O.D.) of SVN53-67 antibodies (b). One patient (#4) was not evaluable due to progression before all four prime-boost doses could be administered. c IFNγ mRNA levels in isolated CD4+ (left) and CD8+ (right) cells from individual vaccinated patients. Cells were stimulated in culture for 2 h with pooled overlapping survivin peptides (a.a. 53–67) at the indicated concentrations. qPCR of IFNγ mRNA was performed as described. Relative quantitation was performed using the ∆∆Ct method, and data were standardized to GAPDH mRNA expression and normalized to IFNγ mRNA levels in unstimulated cells. CD4+ and CD8+ cell populations were verified to be 94 and 98 % pure, respectively, by FACS analysis (data not shown)
Fig. 4
Fig. 4
FACS analysis of CD8+ and CD4+ proliferation in CFSE dye-loaded PBMC obtained from two patients (patient #1; ad) and (patient #7; eh) 12 weeks following vaccination. Proliferation was measured in response to stimulation of cells using either the long (15 amino acid) survivin peptide mimic (SVN53-67/M57) or a 14 amino acid unrelated control peptide (seq: LEEKKQNYVVTDHC). Cell division is indicated by left-shifted peaks
Fig. 5
Fig. 5
Immunohistochemistry of T and B cell markers (×200) in tissue sections of one patient (#8) with recurrent disease 5.6 months following protocol entry. a CD4+ and b CD8+ T cells are shown in representative fields of the patient’s glioblastoma prior to vaccine treatment. c CD4+, d CD8+, e CD20+ and f PD-L1+ cells within contiguous histologic sections of tumor after vaccine treatment and subsequent tumor recurrence
Fig. 6
Fig. 6
Progression-free survival (filled triangle) and overall survival (filled square) of patients. Median PFS was 17.6 weeks, and median OS was 86.6 weeks following study entry. MRI showing tumor at diagnosis (left), first recurrence (center) and 42-month follow-up (right)

References

    1. Li F, Ambrosini G, Chu EY, Plescia J, Tognin S, Marchisio PC, Altieri DC. Control of apoptosis and mitotic spindle checkpoint by survivin. Nature. 1998;396:580–584. doi: 10.1038/25141.
    1. Ambrosini G, Adida C, Altieri DC. A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma. Nat Med. 1997;3:917–921. doi: 10.1038/nm0897-917.
    1. Satoh K, Kaneko K, Hirota M, Masamune A, Satoh A, Shimosegawa T. Expression of survivin is correlated with cancer cell apoptosis and is involved in the development of human pancreatic duct cell tumors. Cancer. 2001;92:271–278. doi: 10.1002/1097-0142(20010715)92:2<271::AID-CNCR1319>;2-0.
    1. Kajiwara Y, Yamasaki F, Hama S, Yahara K, Yoshioka H, Sugiyama K, Arita K, Kurisu K. Expression of survivin in astrocytic tumors: correlation with malignant grade and prognosis. Cancer. 2003;97:1077–1083. doi: 10.1002/cncr.11122.
    1. Verhaak RG, Hoadley KA, Purdom E, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell. 2010;17:98–110. doi: 10.1016/j.ccr.2009.12.020.
    1. Adida C, Crotty PL, McGrath J, Berrebi D, Diebold J, Altieri DC. Developmentally regulated expression of the novel cancer anti-apoptosis gene survivin in human and mouse differentiation. Am J Pathol. 1998;152:43–49.
    1. Uematsu M, Ohsawa I, Aokage T, Nishimaki K, Matsumoto K, Takahashi H, Asoh S, Teramoto A, Ohta S. Prognostic significance of the immunohistochemical index of survivin in glioma: a comparative study with the MIB-1 index. J Neurooncol. 2005;72:231–238. doi: 10.1007/s11060-004-2353-3.
    1. Muller L, Muller-Haegele S, Mitsuhashi M, Gooding W, Okada H, Whiteside TL. Exosomes isolated from plasma of glioma patients enrolled in a vaccination trial reflect antitumor immune activity and might predict survival. Oncoimmunology. 2015;4:e1008347. doi: 10.1080/2162402X.2015.1008347.
    1. Andersen MH, Pedersen LO, Capeller B, Brocker EB, Becker JC, thor Straten P. Spontaneous cytotoxic T-cell responses against survivin-derived MHC class I-restricted T-cell epitopes in situ as well as ex vivo in cancer patients. Cancer Res. 2001;61:5964–5968.
    1. Altieri DC. Survivin and apoptosis control. Adv Cancer Res. 2003;88:31–52. doi: 10.1016/S0065-230X(03)88303-3.
    1. Ciesielski MJ, Ahluwalia MS, Munich SA, Orton M, Barone T, Chanan-Khan A, Fenstermaker RA. Antitumor cytotoxic T-cell response induced by a survivin peptide mimic. Cancer Immunol Immunother. 2010;59:1211–1221. doi: 10.1007/s00262-010-0845-x.
    1. Ciesielski MJ, Kozbor D, Castanaro CA, Barone TA, Fenstermaker RA. Therapeutic effect of a T helper cell supported CTL response induced by a survivin peptide vaccine against murine cerebral glioma. Cancer Immunol Immunother. 2008;57:1827–1835. doi: 10.1007/s00262-008-0510-9.
    1. Hung K, Hayashi R, Lafond-Walker A, Lowenstein C, Pardoll D, Levitsky H. The central role of CD4(+) T cells in the antitumor immune response. J Exp Med. 1998;188:2357–2368. doi: 10.1084/jem.188.12.2357.
    1. Pardoll DM. Inducing autoimmune disease to treat cancer. Proc Natl Acad Sci USA. 1999;96:5340–5342. doi: 10.1073/pnas.96.10.5340.
    1. Fenstermaker RA, Ciesielski MJ. Challenges in the development of a survivin vaccine (SurVaxM) for malignant glioma. Expert Rev Vaccines. 2014;13:377–385. doi: 10.1586/14760584.2014.881255.
    1. Rohayem J, Diestelkoetter P, Weigle B, Oehmichen A, Schmitz M, Mehlhorn J, Conrad K, Rieber EP. Antibody response to the tumor-associated inhibitor of apoptosis protein survivin in cancer patients. Cancer Res. 2000;60:1815–1817.
    1. Hadrup SR, Gehl J, Sorensen RB, Geertsen PF, Straten PT, Andersen MH. Persistence of survivin specific T cells for seven years in a melanoma patient during complete remission. Cancer Biol Ther. 2006;5:480–482. doi: 10.4161/cbt.5.5.2652.
    1. Yagihashi A, Asanuma K, Nakamura M, Araya J, Mano Y, Torigoe T, Kobayashi D, Watanabe N. Detection of anti-survivin antibody in gastrointestinal cancer patients. Clin Chem. 2001;47:1729–1731.
    1. Reker S, Meier A, Holten-Andersen L, Svane IM, Becker JC, thor Straten P, Andersen MH. Identification of novel survivin-derived CTL epitopes. Cancer Biol Ther. 2004;3:173–179. doi: 10.4161/cbt.3.2.611.
    1. Ciesielski MJ, Apfel L, Barone TA, Castro CA, Weiss TC, Fenstermaker RA. Antitumor effects of a xenogeneic survivin bone marrow derived dendritic cell vaccine against murine GL261 gliomas. Cancer Immunol Immunother. 2006;55:1491–1503. doi: 10.1007/s00262-006-0138-6.
    1. Ciesielski MJ, Qiu J, Fenstermaker RA. Survivin as a Cancer Vaccine Target. J Vaccines Vaccin. 2014;5:230.
    1. Overwijk WW, Restifo NP. Autoimmunity and the immunotherapy of cancer: targeting the “self” to destroy the “other”. Crit Rev Immunol. 2000;20:433–450. doi: 10.1615/CritRevImmunol.v20.i6.10.
    1. Fangusaro JR, Jiang Y, Holloway MP, Caldas H, Singh V, Boue DR, Hayes J, Altura RA. Survivin, Survivin-2B, and Survivin-deItaEx3 expression in medulloblastoma: biologic markers of tumour morphology and clinical outcome. Br J Cancer. 2005;92:359–365.
    1. Nakano J, Huang C, Liu D, Masuya D, Yokomise H, Ueno M, Haba R, Sumitomo S. The clinical significance of splice variants and subcellular localisation of survivin in non-small cell lung cancers. Br J Cancer. 2008;98:1109–1117. doi: 10.1038/sj.bjc.6604253.
    1. Castro MG, Baker GJ, Lowenstein PR. Blocking immunosuppressive checkpoints for glioma therapy: the more the merrier! Clin Cancer Res. 2014;20:5147–5149. doi: 10.1158/1078-0432.CCR-14-0820.
    1. Widenmeyer M, Griesemann H, Stevanovic S, et al. Promiscuous survivin peptide induces robust CD4+ T-cell responses in the majority of vaccinated cancer patients. Int J Cancer. 2012;131:140–149. doi: 10.1002/ijc.26365.
    1. Kim EK, Cho HI, Yoon SH, Park MJ, Sohn HJ, Kim HJ, Oh ST, Kim TG. Efficient generation of survivin-specific cytotoxic T lymphocytes from healthy persons in vitro: quantitative and qualitative effects of CD4+ T cells. Vaccine. 2008;26:3987–3997. doi: 10.1016/j.vaccine.2008.05.036.
    1. Moeller M, Kershaw MH, Cameron R, Westwood JA, Trapani JA, Smyth MJ, Darcy PK. Sustained antigen-specific antitumor recall response mediated by gene-modified CD4+ T helper-1 and CD8+ T cells. Cancer Res. 2007;67:11428–11437. doi: 10.1158/0008-5472.CAN-07-1141.
    1. Shen L, Sundstedt A, Ciesielski M, et al. Tasquinimod modulates suppressive myeloid cells and enhances cancer immunotherapies in murine models. Cancer Immunol Res. 2015;3:136–148. doi: 10.1158/2326-6066.CIR-14-0036.
    1. Wong ET, Hess KR, Gleason MJ, Jaeckle KA, Kyritsis AP, Prados MD, Levin VA, Yung WK. Outcomes and prognostic factors in recurrent glioma patients enrolled onto phase II clinical trials. J Clin Oncol. 1999;17:2572–2578.
    1. Stupp R, Wong ET, Kanner AA, et al. NovoTTF-100A versus physician’s choice chemotherapy in recurrent glioblastoma: a randomised phase III trial of a novel treatment modality. Eur J Cancer. 2012;48:2192–2202. doi: 10.1016/j.ejca.2012.04.011.

Source: PubMed

3
S'abonner