Randomized trial of neoadjuvant vaccination with tumor-cell lysate induces T cell response in low-grade gliomas

Hirokazu Ogino, Jennie W Taylor, Takahide Nejo, David Gibson, Payal B Watchmaker, Kaori Okada, Atsuro Saijo, Meghan R Tedesco, Anny Shai, Cynthia M Wong, Jane E Rabbitt, Michael R Olin, Christopher L Moertel, Yasuhiko Nishioka, Andres M Salazar, Annette M Molinaro, Joanna J Phillips, Nicholas A Butowski, Jennifer L Clarke, Nancy Ann Oberheim Bush, Shawn L Hervey-Jumper, Philip Theodosopoulos, Susan M Chang, Mitchel S Berger, Hideho Okada, Hirokazu Ogino, Jennie W Taylor, Takahide Nejo, David Gibson, Payal B Watchmaker, Kaori Okada, Atsuro Saijo, Meghan R Tedesco, Anny Shai, Cynthia M Wong, Jane E Rabbitt, Michael R Olin, Christopher L Moertel, Yasuhiko Nishioka, Andres M Salazar, Annette M Molinaro, Joanna J Phillips, Nicholas A Butowski, Jennifer L Clarke, Nancy Ann Oberheim Bush, Shawn L Hervey-Jumper, Philip Theodosopoulos, Susan M Chang, Mitchel S Berger, Hideho Okada

Abstract

BACKGROUNDLong-term prognosis of WHO grade II low-grade gliomas (LGGs) is poor, with a high risk of recurrence and malignant transformation into high-grade gliomas. Given the relatively intact immune system of patients with LGGs and the slow tumor growth rate, vaccines are an attractive treatment strategy.METHODSWe conducted a pilot study to evaluate the safety and immunological effects of vaccination with GBM6-AD, lysate of an allogeneic glioblastoma stem cell line, with poly-ICLC in patients with LGGs. Patients were randomized to receive the vaccines before surgery (arm 1) or not (arm 2) and all patients received adjuvant vaccines. Coprimary outcomes were to evaluate safety and immune response in the tumor.RESULTSA total of 17 eligible patients were enrolled - 9 in arm 1 and 8 in arm 2. This regimen was well tolerated with no regimen-limiting toxicity. Neoadjuvant vaccination induced upregulation of type-1 cytokines and chemokines and increased activated CD8+ T cells in peripheral blood. Single-cell RNA/T cell receptor sequencing detected CD8+ T cell clones that expanded with effector phenotype and migrated into the tumor microenvironment (TME) in response to neoadjuvant vaccination. Mass cytometric analyses detected increased tissue resident-like CD8+ T cells with effector memory phenotype in the TME after the neoadjuvant vaccination.CONCLUSIONThe regimen induced effector CD8+ T cell response in peripheral blood and enabled vaccine-reactive CD8+ T cells to migrate into the TME. Further refinements of the regimen may have to be integrated into future strategies.TRIAL REGISTRATIONClinicalTrials.gov NCT02549833.FUNDINGNIH (1R35NS105068, 1R21CA233856), Dabbiere Foundation, Parker Institute for Cancer Immunotherapy, and Daiichi Sankyo Foundation of Life Science.

Keywords: Brain cancer; Cancer immunotherapy; Oncology; T cells; Vaccines.

Figures

Figure 1. Study schema.
Figure 1. Study schema.
Patients were randomized to arm 1 or 2. Patients in arm 1 received GBM6-AD lysate and poly-ICLC on days –23 ± 2, –16 ± 2, –9 ± 2, and –2 relative to the scheduled surgery. At least 2 weeks after the postoperative steroid was tapered, but within 10 weeks after surgery, patients in arm 1 and arm 2 started receiving the GBM6-AD/poly-ICLC vaccines every 3 weeks for 5 doses (weeks A1, A4, A7, A10, and A13; defined as the weeks from first adjuvant vaccine dose) followed by booster vaccines at weeks A32 and A48.
Figure 2. Neoadjuvant vaccinations with GBM6-AD lysate…
Figure 2. Neoadjuvant vaccinations with GBM6-AD lysate and poly-ICLC induced the upregulation of type-1 chemokines and cytokines in peripheral blood.
Serum concentrations of multiple chemokines and cytokines were measured by Luminex multiplex assay. The type-1 chemokine CXCL10 was elevated in arm 1 samples on the day of surgery, within 48 hours of the last neoadjuvant vaccination. Effector cytokines, such as IFN-γ, TNF-α, and IL-10, also demonstrated significant upregulation after the neoadjuvant vaccines. *P < 0.05 (calculated by paired Wilcoxon test) and **P < 0.05 (calculated by nonpaired Wilcoxon test).
Figure 3. Mass cytometric analyses detected increases…
Figure 3. Mass cytometric analyses detected increases of PD-1+GZMBhiTbethi effector memory and GZMBhiTbethi effector CD8+ T cells after the neoadjuvant vaccines.
(A) T-distributed stochastic neighbor embedding (t-SNE) plot of CD8+ T cells. To evaluate the vaccine-induced changes of phenotype in peripheral blood, mass cytometric analyses were conducted. CD8+ T cells were subjected to dimension reductional algorithm t-SNE for visualization in 2D space and clustered by FlowSOM based on the expression status of 7 differentiation markers (CD62L, CD27, CD127, CCR7, CD45RO, CD45RA, and PD-1). (B) Heatmap visualizing the relative expression (z score) of T cell–relevant markers in each subpopulation. Each cluster was annotated based on the expression status of differentiation markers as listed above. (C) The longitudinal analyses of proportions of each subpopulation in arm 1 patients. Neoadjuvant vaccination with GBM6-AD and poly-ICLC increased PD-1+GZMBhiTbethi effector memory and GZMBhiTbethi effector CD8+ T cells while decreasing naive CD8+ T cells. *P < 0.05 (paired Wilcoxon test). (D) The expression levels of activation markers, such as CD38, Tbet, and PD-1, on the PD-1+GZMBhiTbethi effector memory cells were enhanced in the samples obtained after the neoadjuvant vaccines. *P < 0.05 (nonpaired, 2-tailed t test).
Figure 4. scRNA-Seq analyses revealed the increases…
Figure 4. scRNA-Seq analyses revealed the increases of effector CD4+ and CD8+ and decreases of naive CD4+ and CD8+ T cell populations after the neoadjuvant vaccinations.
ScRNA-Seq and scTCR-Seq analyses on the 10x Genomics platform were conducted in PBMCs obtained from the 4 immunological responders (patients 103-018, -26, -29, -51) at baseline and after neoadjuvant vaccines. (A) UMAP of pooled PBMCs from all 4 patients at baseline and after neoadjuvant vaccines. Clusters were annotated based on the expression of known marker genes. Mono, monocyte; pDC, plasmacytoid DC; MK, megakaryocyte; B, B cells. (B) UMAP was colored by TCR detection. TCRs were mainly detected in 5 clusters that represent T cell and NKT cell populations (pink). (C) UMAP of T cells and NKT cells. T cell and NKT cell populations were reclustered and grouped into 9 subpopulations. EM, effector memory. (D) UMAP of T cells and NKT cells was colored by treatment status (either pre- or post-vaccination). Cytotoxic T cells, such as effector CD8+ T cells and NKT cells, were enriched in postvaccinated samples (light blue). (E) The bar plot showing the proportion of each cell cluster in each sample. (F) Quantification of each cell cluster in prevaccinated and postvaccinated samples. The proportion of effector T cells showed a trend toward an increase in postvaccinated samples while that of naive T cells showed a trend toward a decrease. P values were calculated by paired Wilcoxon test.
Figure 5. Vaccine-reactive CD8 + T cell…
Figure 5. Vaccine-reactive CD8+ T cell clones with an effector phenotype migrated into the tumor microenvironment.
(A) The top 15 frequent clonotypes in postvaccinated samples were extracted, and their frequencies were compared. Most of these clonotypes showed higher frequencies in postvaccinated samples than at baseline. (B) The TCR clonotypes that were enriched in postvaccinated PBMCs were extracted with an adjusted P value less than 0.15. Patients 103-018, -26, -29, and -51 were found to have 26, 5, 13, and 32 enriched TCR-b clonotypes, respectively, in their PBMCs. Some of these clonotypes were also found in the TCR repertoire of corresponding tumors (determined by bulk TCR-Seq). (C) The T cell clones that had these overlapped clonotypes mostly belonged to the effector CD8 cluster in PBMCs in all cases. (D and E) The expression of GZMB was upregulated by neoadjuvant vaccinations in these T cell clones. Log-normalized (count) on x axis was calculated as log (count/[total count of the cell] × 10,000 + 1). *P < 0.05 (nonpaired, 2-tailed t test).
Figure 6. The proportion of tissue resident–like…
Figure 6. The proportion of tissue resident–like CD8+ T cells with effector memory phenotype was significantly higher in the vaccinated tumor microenvironment.
Single-cell suspensions dissociated from tumor samples from arm 1 (4 cases) and arm 2 (6 cases) were analyzed by mass cytometry. (A) CD3+ T cells were subjected to dimension reductional algorithm t-SNE and clustered by FlowSOM based on the expression status of 10 differentiation markers (CD4, CD8a, CD62L, CD27, CD127, CCR7, CD45RO, CD45RA, CD25, and PD-1). (B) Heatmap visualizing the relative expression (z score) of T cell–relevant markers in each subpopulation. Each cluster was annotated based on the expression status of differentiation markers as listed above. (C) The proportion of tissue resident–like CD8+ T cells with effector memory phenotype (CD103+, PD-1+, CXCR3hi, CCR7–, CD45RO+, GZMBhi) was significantly higher in arm 1 samples. *P < 0.05 (nonpaired Wilcoxon test). The proportion of Tregs in arm 1 showed a trend toward a higher percentage than arm 2 but without statistical significance. (D) TILs in this tissue resident–like CD8+ T cell cluster in arm 1 tumors demonstrated significantly higher expression levels for the CXCL10 receptor CXCR3, GZMB, and Tbet than those in arm 2 tumors.

References

    1. Ostrom QT, et al. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2013-2017. Neuro Oncol. 2020;22(12 suppl 2):iv1–iv96.
    1. Louis DN, et al. The 2016 World Health Organization classification of tumors of the central nervous system: a summary. Acta Neuropathol. 2016;131(6):803–820. doi: 10.1007/s00401-016-1545-1.
    1. Sanai N, et al. Low-grade gliomas in adults. J Neurosurg. 2011;115(5):948–965. doi: 10.3171/2011.7.JNS101238.
    1. Brown PD, et al. Clinical Radiation Oncology. Churchill-Livingstone; 2006:493–514.
    1. van den Bent MJ, et al. Long-term efficacy of early versus delayed radiotherapy for low-grade astrocytoma and oligodendroglioma in adults: the EORTC 22845 randomised trial. Lancet. 2005;366(9490):985–990. doi: 10.1016/S0140-6736(05)67070-5.
    1. Papagikos MA, et al. Lessons learned from randomised clinical trials in adult low grade glioma. Lancet Oncol. 2005;6(4):240–244. doi: 10.1016/S1470-2045(05)70095-4.
    1. Shaw EG, et al. Recurrence following neurosurgeon-determined gross-total resection of adult supratentorial low-grade glioma: results of a prospective clinical trial. J Neurosurg. 2008;109(5):835–841. doi: 10.3171/JNS/2008/109/11/0835.
    1. Parsons DW, et al. An integrated genomic analysis of human glioblastoma multiforme. Science. 2008;321(5897):1807–1812. doi: 10.1126/science.1164382.
    1. Yan H, et al. IDH1 and IDH2 mutations in gliomas. N Engl J Med. 2009;360(8):765–773. doi: 10.1056/NEJMoa0808710.
    1. Bunse L, et al. Suppression of antitumor T cell immunity by the oncometabolite (R)-2-hydroxyglutarate. Nat Med. 2018;24(8):1192–1203. doi: 10.1038/s41591-018-0095-6.
    1. Kohanbash G, et al. Isocitrate dehydrogenase mutations suppress STAT1 and CD8+ T cell accumulation in gliomas. J Clin Invest. 2017;127(4):1425–1437. doi: 10.1172/JCI90644.
    1. Okada H, et al. Induction of robust type-I CD8+ T-cell responses in WHO grade 2 low-grade glioma patients receiving peptide-based vaccines in combination with poly-ICLC. Clin Cancer Res. 2015;21(2):286–294. doi: 10.1158/1078-0432.CCR-14-1790.
    1. Olin MR, et al. Vaccination with dendritic cells loaded with allogeneic brain tumor cells for recurrent malignant brain tumors induces a CD4(+)IL17(+) response. J Immunother Cancer. 2014;2:4. doi: 10.1186/2051-1426-2-4.
    1. Zhu X, et al. Toll like receptor-3 ligand poly-ICLC promotes the efficacy of peripheral vaccinations with tumor antigen-derived peptide epitopes in murine CNS tumor models. J Transl Med. 2007;5:10. doi: 10.1186/1479-5876-5-10.
    1. Newman AM, et al. Determining cell type abundance and expression from bulk tissues with digital cytometry. Nat Biotechnol. 2019;37(7):773–782. doi: 10.1038/s41587-019-0114-2.
    1. van den Bent MJ, et al. Response assessment in neuro-oncology (a report of the RANO group): assessment of outcome in trials of diffuse low-grade gliomas. Lancet Oncol. 2011;12(6):583–593. doi: 10.1016/S1470-2045(11)70057-2.
    1. Okada H, et al. Induction of CD8+ T-cell responses against novel glioma-associated antigen peptides and clinical activity by vaccinations with {alpha}-type 1 polarized dendritic cells and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in patients with recurrent malignant glioma. J Clin Oncol. 2011;29(3):330–336. doi: 10.1200/JCO.2010.30.7744.
    1. Zhu X, et al. Poly-ICLC promotes the infiltration of effector T cells into intracranial gliomas via induction of CXCL10 in IFN-alpha and IFN-gamma dependent manners. Cancer Immunol Immunother. 2010;59(9):1401–1409. doi: 10.1007/s00262-010-0876-3.
    1. Smolders J, et al. Tissue-resident memory T cells populate the human brain. Nat Commun. 2018;9(1):4593. doi: 10.1038/s41467-018-07053-9.
    1. Thorsson V, et al. The immune landscape of cancer. Immunity. 2018;48(4):812–830. doi: 10.1016/j.immuni.2018.03.023.
    1. Nduom EK, et al. Characterization of the blood-brain barrier of metastatic and primary malignant neoplasms. J Neurosurg. 2013;119(2):427–433. doi: 10.3171/2013.3.JNS122226.
    1. González FE, et al. Tumor cell lysates as immunogenic sources for cancer vaccine design. Hum Vaccin Immunother. 2014;10(11):3261–3269. doi: 10.4161/21645515.2014.982996.
    1. Cloughesy TF, et al. Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma. Nat Med. 2019;25(3):477–486. doi: 10.1038/s41591-018-0337-7.
    1. Schalper KA, et al. Neoadjuvant nivolumab modifies the tumor immune microenvironment in resectable glioblastoma. Nat Med. 2019;25(3):470–476. doi: 10.1038/s41591-018-0339-5.
    1. Friebel E, et al. Single-cell mapping of human brain cancer reveals tumor-specific instruction of tissue-invading leukocytes. Cell. 2020;181(7):1626–1642. doi: 10.1016/j.cell.2020.04.055.
    1. Klemm F, et al. Interrogation of the microenvironmental landscape in brain tumors reveals disease-specific alterations of immune cells. Cell. 2020;181(7):1643–1660. doi: 10.1016/j.cell.2020.05.007.
    1. Eguchi J, et al. Identification of interleukin-13 receptor alpha2 peptide analogues capable of inducing improved antiglioma CTL responses. Cancer Res. 2006;66(11):5883–5891. doi: 10.1158/0008-5472.CAN-06-0363.
    1. Hatano M, et al. EphA2 as a glioma-associated antigen: a novel target for glioma vaccines. Neoplasia. 2005;7(8):717–722. doi: 10.1593/neo.05277.
    1. Okano F, et al. Identification of a novel HLA-A*0201-restricted, cytotoxic T lymphocyte epitope in a human glioma-associated antigen, interleukin 13 receptor alpha2 chain. Clin Cancer Res. 2002;8(9):2851–2855.
    1. Xiong Z, et al. Tumor-derived vaccines containing CD200 inhibit immune activation: implications for immunotherapy. Immunotherapy. 2016;8(9):1059–1071. doi: 10.2217/imt-2016-0033.
    1. Gielis S, et al. Detection of enriched T cell epitope specificity in full T cell receptor sequence repertoires. Front Immunol. 2019;10:2820. doi: 10.3389/fimmu.2019.02820.
    1. Springer I, et al. Prediction of specific TCR-peptide binding from large dictionaries of TCR-peptide pairs. Front Immunol. 2020;11:1803. doi: 10.3389/fimmu.2020.01803.
    1. Cancer Genome Atlas Research Network. et al. Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas. N Engl J Med. 2015;372(26):2481–2498. doi: 10.1056/NEJMoa1402121.
    1. Palladini A, et al. Cancer immunoprevention: from mice to early clinical trials. BMC Immunol. 2018;19(1):16. doi: 10.1186/s12865-018-0253-0.
    1. Kimura T, et al. MUC1 vaccine for individuals with advanced adenoma of the colon: a cancer immunoprevention feasibility study. Cancer Prev Res (Phila) 2013;6(1):18–26. doi: 10.1158/1940-6207.CAPR-12-0275.
    1. Ge C, et al. Phase I clinical trial of a novel autologous modified-DC vaccine in patients with resected NSCLC. BMC Cancer. 2017;17(1):884. doi: 10.1186/s12885-017-3859-3.
    1. Chen H, et al. Cytofkit: a bioconductor package for an integrated mass cytometry data analysis pipeline. PLoS Comput Biol. 2016;12(9):e1005112. doi: 10.1371/journal.pcbi.1005112.
    1. Butler A, et al. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat Biotechnol. 2018;36(5):411–420. doi: 10.1038/nbt.4096.
    1. Yost KE, et al. Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat Med. 2019;25(8):1251–1259. doi: 10.1038/s41591-019-0522-3.
    1. Chen S, et al. fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics. 2018;34(17):i884–i890. doi: 10.1093/bioinformatics/bty560.
    1. Pertea M, et al. StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat Biotechnol. 2015;33(3):290–295. doi: 10.1038/nbt.3122.
    1. Vivian J, et al. Toil enables reproducible, open source, big biomedical data analyses. Nat Biotechnol. 2017;35(4):314–316. doi: 10.1038/nbt.3772.
    1. Ceccarelli M, et al. Molecular profiling reveals biologically discrete subsets and pathways of progression in diffuse glioma. Cell. 2016;164(3):550–563. doi: 10.1016/j.cell.2015.12.028.

Source: PubMed

3
Sottoscrivi