A Neoantigen-Based Peptide Vaccine for Patients With Advanced Pancreatic Cancer Refractory to Standard Treatment

Zheling Chen, Shanshan Zhang, Ning Han, Jiahong Jiang, Yunyun Xu, Dongying Ma, Lantian Lu, Xiaojie Guo, Min Qiu, Qinxue Huang, Huimin Wang, Fan Mo, Shuqing Chen, Liu Yang, Zheling Chen, Shanshan Zhang, Ning Han, Jiahong Jiang, Yunyun Xu, Dongying Ma, Lantian Lu, Xiaojie Guo, Min Qiu, Qinxue Huang, Huimin Wang, Fan Mo, Shuqing Chen, Liu Yang

Abstract

Background: Neoantigens are critical targets to elicit robust antitumor T-cell responses. Personalized cancer vaccines developed based on neoantigens have shown promising results by prolonging cancer patients' overall survival (OS) for several cancer types. However, the safety and efficacy of these vaccine modalities remains unclear in pancreatic cancer patients.

Methods: This retrospective study enrolled 7 advanced pancreatic cancer patients. Up to 20 neoantigen peptides per patient identified by our in-house pipeline iNeo-Suite were selected, manufactured and administered to these patients with low tumor mutation burden (TMB) (less than 10 mutations/Mb). Each patient received multiple doses of vaccine depending on the progression of the disease. Peripheral blood samples of each patient were collected pre- and post-vaccination for the analysis of the immunogenicity of iNeo-Vac-P01 through ELISpot assay and flow cytometry.

Results: No severe vaccine-related adverse effects were witnessed in patients enrolled in this study. The mean OS, OS associated with vaccine treatment and progression free survival (PFS) were reported to be 24.1, 8.3 and 3.1 months, respectively. Higher peripheral IFN-γ titer and CD4+ or CD8+ effector memory T cells count post vaccination were found in patients with relatively long overall survival. Remarkably, for patient P01 who had a 21-month OS associated with vaccine treatment, the abundance of antigen-specific TCR clone drastically increased from 0% to nearly 100%, indicating the potential of iNeo-Vac-P01 in inducing the activation of a specific subset of T cells to kill cancer cells.

Conclusions: Neoantigen identification and selection were successfully applied to advanced pancreatic cancer patients with low TMB. As one of the earliest studies that addressed an issue in treating pancreatic cancer with personalized vaccines, it has been demonstrated that iNeo-Vac-P01, a personalized neoantigen-based peptide vaccine, could improve the currently limited clinical efficacy of pancreatic cancer.

Clinical trial registration: ClinicalTrials.gov, identifier (NCT03645148).Registered August 24, 2018 - Retrospectively registered.

Keywords: immunotherapy; neoantigen; pancreatic cancer; peptide; vaccine.

Conflict of interest statement

FM is an employee for Hangzhou Neoantigen Therapeutics Co., Ltd and Hangzhou Al-Force Therapeutics. SZ, NH, DM, HW, XG, KL, MQ, QH, LL, and SC are employees for Hangzhou Neoantigen Therapeutics Co., Ltd. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Chen, Zhang, Han, Jiang, Xu, Ma, Lu, Guo, Qiu, Huang, Wang, Mo, Chen and Yang.

Figures

Figure 1
Figure 1
Clinical treatment process for each patient from surgery or primary visit until the end of follow-up.
Figure 2
Figure 2
The clinical response and prognosis of treated patients. (A) The overall survival curve of each patient. (B) The percentage changes of tumor lesion in size from baseline. The changes in lesions between a positive value of 25% and a negative value of 50% are considered stable disease.
Figure 3
Figure 3
A case report of Patient P01. (A) Treatment timeline of P01. (B) Comparison of lymph node before & after vaccination by imaging. (C, D)Ex vivo IFN-γ ELISpot of PBMCs was performed with peptides at different time points. The dimethyl sulfoxide (DMSO) group was used as the negative control and mixed peptides from CEF (including peptides of cytomegalovirus, Epstein–Barr virus and influenza virus) were used as the positive control. (E) Increased abundance of peripheral T cell clones after vaccination was detected by TCR sequencing.
Figure 4
Figure 4
Immune Response induced by iNeo-Vac-P01. (A) iNeo-Vac-P01 induced specific T cell response. For each patient marked in X-axis, green triangle and red diamond represent the response rates pre- and post-vaccination, defined as the ratios of the numbers of peptides (or peptide pools) with positive ELISpot results before and after vaccination to the total number of peptides (or peptide pools) in vaccine, respectively. The bar chart with secondary Y-axis represented the IFN-γ spots per 105 PBMCs of the peptide with best response for each patient. (B) Change in T cell subsets count at baseline and 22 days post iNeo-Vac-P01 vaccination. Blue and yellow dots represent the cell counts at baseline and 22 days post iNeo-Vac-P01, respectively. Grey dots represent the fold change of corresponding cell count, which can be read at the right Y-axis. Counts of T cell subsets including CD4+ central memory, CD4+ effector memory, CD8+ central memory and CD8+ effector memory T cells were compared in long OS patients (shown in dark grey) and short OS patients (shown in light grey). *: significant, p=0.01 to 0.05; **: very significant, p=0.001 to 0.01; ns: not significant.
Figure 5
Figure 5
The proportion of CTLA4+ T cells in peripheral blood increased after vaccination. The proportions of CD8+ CTLA-4+ T cells and CD4+ CTLA-4+ T cells ratio to total T cells in peripheral blood were determined by flow cytometry. Fold changes of post vaccination compared with baseline were calculated.

References

    1. Idachaba S, Dada O, Abimbola O, Olayinka O, Uma A, Olunu E, et al. . A Review of Pancreatic Cancer: Epidemiology, Genetics, Screening, and Management. Open Access Macedonian J Med Sci (2019) 7(4):663–71. 10.3889/oamjms.2019.104
    1. Bliss LA, Witkowski ER, Yang CJ, Tseng JF. Outcomes in Operative Management of Pancreatic Cancer. J Surg Oncol (2014) 110(5):592–8. 10.1002/jso.23744
    1. Vincent A, Herman J, Schulick R, Hruban RH, Goggins M. Pancreatic Cancer. Lancet (2011) 378(9791):607–20. 10.1016/S0140-6736(10)62307-0
    1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: Cancer J Clin (2018) 68(6):394–424. 10.3322/caac.21492
    1. Nair R, Westin J. CAR T-Cells. Adv Exp Med Biol (2020) 1244:215–33. 10.1007/978-3-030-41008-7_10
    1. Han Y, Liu D, Li L. PD-1/PD-L1 Pathway: Current Researches in Cancer. Am J Cancer Res (2020) 10(3):727–42.
    1. Weiss GJ, Waypa J, Blaydorn L, Coats J, McGahey K, Sangal A, et al. . A Phase Ib Study of Pembrolizumab Plus Chemotherapy in Patients With Advanced Cancer (PembroPlus). Br J Cancer (2017) 117(1):33–40. 10.1038/bjc.2017.145
    1. Aglietta M, Barone C, Sawyer MB, Moore MJ, Miller WH, Jr, Bagala C, et al. . A Phase I Dose Escalation Trial of Tremelimumab (CP-675,206) in Combination With Gemcitabine in Chemotherapy-Naive Patients With Metastatic Pancreatic Cancer. Ann Oncol (2014) 25(9):1750–5. 10.1093/annonc/mdu205
    1. Bansal D, Beck R, Arora V, Knoche EM, Picus J, Reimers MA, et al. . A Pilot Trial of Neoantigen DNA Vaccine in Combination With Nivolumab/Ipilimumab and Prostvac in Metastatic Hormone-Sensitive Prostate Cancer (mHSPC). J Clin Oncol (2021) 39(6_suppl):TPS192–TPS. 10.1200/JCO.2021.39.6_suppl.TPS192
    1. Ding Z, Li Q, Zhang R, Xie L, Shu Y, Gao S, et al. . Personalized Neoantigen Pulsed Dendritic Cell Vaccine for Advanced Lung Cancer. Signal Transduct Targeted Ther (2021) 6(1):26. 10.1038/s41392-020-00448-5
    1. Ott PA, Hu-Lieskovan S, Chmielowski B, Govindan R, Naing A, Bhardwaj N, et al. . A Phase Ib Trial of Personalized Neoantigen Therapy Plus Anti-PD-1 in Patients With Advanced Melanoma, Non-Small Cell Lung Cancer, or Bladder Cancer. Cell (2020) 183(2):347–62 e24. 10.1016/j.cell.2020.08.053
    1. Yadav M, Jhunjhunwala S, Phung QT, Lupardus P, Tanguay J, Bumbaca S, et al. . Predicting Immunogenic Tumour Mutations by Combining Mass Spectrometry and Exome Sequencing. Nature (2014) 515(7528):572–6. 10.1038/nature14001
    1. Balachandran VP, Łuksza M, Zhao JN, Makarov V, Moral JA, Remark R, et al. . Identification of Unique Neoantigen Qualities in Long-Term Survivors of Pancreatic Cancer. Nature (2017) 551(7681):512–6. 10.1038/nature24462
    1. Ott PA, Hu Z, Keskin DB, Shukla SA, Sun J, Bozym DJ, et al. . An Immunogenic Personal Neoantigen Vaccine for Patients With Melanoma. Nature (2017) 547(7662):217–21. 10.1038/nature22991
    1. Sahin U, Derhovanessian E, Miller M, Kloke BP, Simon P, Lower M, et al. . Personalized RNA Mutanome Vaccines Mobilize Poly-Specific Therapeutic Immunity Against Cancer. Nature (2017) 547(7662):222–6. 10.1038/nature23003
    1. Keskin DB, Anandappa AJ, Sun J, Tirosh I, Mathewson ND, Li S, et al. . Neoantigen Vaccine Generates Intratumoral T Cell Responses in Phase Ib Glioblastoma Trial. Nature (2019) 565(7738):234–9. 10.1038/s41586-018-0792-9
    1. Forghanifard MM, Gholamin M, Moaven O, Farshchian M, Ghahraman M, Aledavood A, et al. . Neoantigen in Esophageal Squamous Cell Carcinoma for Dendritic Cell-Based Cancer Vaccine Development. Med Oncol (2014) 31(10):191. 10.1007/s12032-014-0191-5
    1. Kuai R, Ochyl LJ, Bahjat KS, Schwendeman A, Moon JJ. Designer Vaccine Nanodiscs for Personalized Cancer Immunotherapy. Nat Mater (2017) 16(4):489–96. 10.1038/nmat4822
    1. Middleton G, Silcocks P, Cox T, Valle J, Wadsley J, Propper D, et al. . Gemcitabine and Capecitabine With or Without Telomerase Peptide Vaccine GV1001 in Patients With Locally Advanced or Metastatic Pancreatic Cancer (TeloVac): An Open-Label, Randomised, Phase 3 Trial. Lancet Oncol (2014) 15(8):829–40. 10.1016/S1470-2045(14)70236-0
    1. Shima H, Kutomi G, Satomi F, Imamura M, Kimura Y, Mizuguchi T, et al. . Case Report: Long-Term Survival of a Pancreatic Cancer Patient Immunized With an SVN-2B Peptide Vaccine. Cancer Immunol Immunother: CII (2018) 67(10):1603–9. 10.1007/s00262-018-2217-x
    1. Suzuki N, Hazama S, Iguchi H, Uesugi K, Tanaka H, Hirakawa K, et al. . Phase II Clinical Trial of Peptide Cocktail Therapy for Patients With Advanced Pancreatic Cancer: VENUS-PC Study. Cancer Sci (2017) 108(1):73–80. 10.1111/cas.13113
    1. Weden S, Klemp M, Gladhaug IP, Moller M, Eriksen JA, Gaudernack G, et al. . Long-Term Follow-Up of Patients With Resected Pancreatic Cancer Following Vaccination Against Mutant K-Ras. Int J Cancer (2011) 128(5):1120–8. 10.1002/ijc.25449
    1. Gjertsen MK, Buanes T, Rosseland AR, Bakka A, Gladhaug I, Soreide O, et al. . Intradermal Ras Peptide Vaccination With Granulocyte-Macrophage Colony-Stimulating Factor as Adjuvant: Clinical and Immunological Responses in Patients With Pancreatic Adenocarcinoma. Int J Cancer (2001) 92(3):441–50. 10.1002/ijc.1205
    1. Kirner A, Mayer-Mokler A, Reinhardt C. IMA901: A Multi-Peptide Cancer Vaccine for Treatment of Renal Cell Cancer. Hum Vaccin Immunother (2014) 10(11):3179–89. 10.4161/21645515.2014.983857
    1. Bais P, Namburi S, Gatti DM, Zhang X, Chuang JH. CloudNeo: A Cloud Pipeline for Identifying Patient-Specific Tumor Neoantigens. Bioinformatics (2017) 33(19):3110–2. 10.1093/bioinformatics/btx375
    1. Hundal J, Miller CA, Griffith M, Griffith OL, Walker J, Kiwala S, et al. . Cancer Immunogenomics: Computational Neoantigen Identification and Vaccine Design. Cold Spring Harb Symp Quant Biol (2016) 81:105–11. 10.1101/sqb.2016.81.030726
    1. Schenck RO, Lakatos E, Gatenbee C, Graham TA, Anderson ARA. NeoPredPipe: High-Throughput Neoantigen Prediction and Recognition Potential Pipeline. BMC Bioinf (2019) 20(1):264. 10.1186/s12859-019-2876-4
    1. Tappeiner E, Finotello F, Charoentong P, Mayer C, Rieder D, Trajanoski Z. TIminer: NGS Data Mining Pipeline for Cancer Immunology and Immunotherapy. Bioinformatics (2017) 33(19):3140–1. 10.1093/bioinformatics/btx377
    1. Torres-Garcia W, Zheng S, Sivachenko A, Vegesna R, Wang Q, Yao R, et al. . PRADA: Pipeline for RNA Sequencing Data Analysis. Bioinformatics (2014) 30(15):2224–6. 10.1093/bioinformatics/btu169
    1. Sallusto F, Geginat J, Lanzavecchia A. Central Memory and Effector Memory T Cell Subsets: Function, Generation, and Maintenance. Annu Rev Immunol (2004) 22:745–63. 10.1146/annurev.immunol.22.012703.104702
    1. Ryan DP, Hong TS, Bardeesy N. Pancreatic Adenocarcinoma. New Engl J Med (2014) 371(22):2140–1. 10.1056/NEJMc1412266
    1. Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, et al. . FOLFIRINOX Versus Gemcitabine for Metastatic Pancreatic Cancer. New Engl J Med (2011) 364(19):1817–25. 10.1056/NEJMoa1011923
    1. Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, et al. . Increased Survival in Pancreatic Cancer With Nab-Paclitaxel Plus Gemcitabine. New Engl J Med (2013) 369(18):1691–703. 10.1056/NEJMoa1304369
    1. Vienot A, Beinse G, Louvet C, de Mestier L, Meurisse A, Fein F, et al. . Overall Survival Prediction and Usefulness of Second-Line Chemotherapy in Advanced Pancreatic Adenocarcinoma. J Natl Cancer Inst (2017) 109(10). 10.1093/jnci/djx037
    1. Rahma OE, Hamilton JM, Wojtowicz M, Dakheel O, Bernstein S, Liewehr DJ, et al. . The Immunological and Clinical Effects of Mutated Ras Peptide Vaccine in Combination With IL-2, GM-CSF, or Both in Patients With Solid Tumors. J Trans Med (2014) 12:55. 10.1186/1479-5876-12-55

Source: PubMed

3
Abonner