Combination treatment of radiofrequency ablation and peptide neoantigen vaccination: Promising modality for future cancer immunotherapy

Jiawei Shou, Fan Mo, Shanshan Zhang, Lantian Lu, Ning Han, Liang Liu, Min Qiu, Hongseng Li, Weidong Han, Dongying Ma, Xiaojie Guo, Qianpeng Guo, Qinxue Huang, Xiaomeng Zhang, Shengli Ye, Hongming Pan, Shuqing Chen, Yong Fang, Jiawei Shou, Fan Mo, Shanshan Zhang, Lantian Lu, Ning Han, Liang Liu, Min Qiu, Hongseng Li, Weidong Han, Dongying Ma, Xiaojie Guo, Qianpeng Guo, Qinxue Huang, Xiaomeng Zhang, Shengli Ye, Hongming Pan, Shuqing Chen, Yong Fang

Abstract

Background: The safety and immunogenicity of a personalized neoantigen-based peptide vaccine, iNeo-Vac-P01, was reported previously in patients with a variety of cancer types. The current study investigated the synergistic effects of radiofrequency ablation (RFA) and neoantigen vaccination in cancer patients and tumor-bearing mice.

Methods: Twenty-eight cancer patients were enrolled in this study, including 10 patients who had received RFA treatment within 6 months before vaccination (Cohort 1), and 18 patients who had not (Cohort 2). Individualized neoantigen peptide vaccines were designed, manufactured, and subcutaneously administrated with GM-CSF as an adjuvant for all patients. Mouse models were employed to validate the synergistic efficacy of combination treatment of RFA and neoantigen vaccination.

Results: Longer median progression free survival (mPFS) and median overall survival (mOS) were observed in patients in Cohort 1 compared to patients in Cohort 2 (4.42 and 20.18 months vs. 2.82 and 10.94 months). The results of ex vivo IFN-γ ELISpot assay showed that patients in Cohort 1 had stronger neoantigen-specific immune responses at baseline and post vaccination. Mice receiving combination treatment of RFA and neoantigen vaccines displayed higher antitumor immune responses than mice receiving single modality. The combination of PD-1 blockage with RFA and neoantigen vaccines further enhanced the antitumor response in mice.

Conclusion: Neoantigen vaccination after local RFA treatment could improve the clinical and immune response among patients of different cancer types. The synergistic antitumor potentials of these two modalities were also validated in mice, and might be further enhanced by immune checkpoint inhibition. The mechanisms of their synergies require further investigation.

Clinical trial registration: https://ichgcp.net/clinical-trials-registry/NCT03662815" title="See in ClinicalTrials.gov">NCT03662815.

Keywords: cancer; immune checkpoint inhibition; immunotherapy; neoantigen vaccine; radiofrequency ablation.

Conflict of interest statement

FM, SZ, LLu, NH, LLiu, MQ, DM, XG, QH, XZ, and SC are employed by Hangzhou Neoantigen Therapeutics Co., Ltd. FM is employed by company Hangzhou AI-Force Therapeutics Co., Ltd. NH is employed by company Hangzhou AI-Nano 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 © 2022 Shou, Mo, Zhang, Lu, Han, Liu, Qiu, Li, Han, Ma, Guo, Guo, Huang, Zhang, Ye, Pan, Chen and Fang.

Figures

Figure 1
Figure 1
Better clinical response in RFA+Vac patients upon receiving iNeo-Vac. (A) Swimmer plots of RFA+Vac patients (with yellow background) and Vac patients (with blue background); (B) Waterfall plots of RFA+Vac patients (shown in yellow) and Vac patients (shown in blue). Dashed line above 20% or below -30% indicate 20% increase (≥ 20% is considered as progressive disease) or 30% reduction (≤ -30% is considered as partial response) of the sum of the longest diameters of the tumors, respectively. # suggested that new lesion(s) was found; (C) Spider plots of RFA+Vac patients and Vac patients. The “best clinical response” in panel b was defined as the largest reduction of target lesion in size during treatment.
Figure 2
Figure 2
RFA+Vac patients showed better progression-free survival (PFS) and overall survival (OS). (A) Progression-free survival of Vac patients (shown in blue) and RFA+Vac patients (shown in yellow). (B) Overall survival of Vac patients (blue) and RFA+Vac patients (yellow). (C) Characteristics of patients in RFA+Vac colorectal group, in comparison to that of patients in RFA colorectal group. (D) Overall survival of RFA-pretreated colorectal cancer patients (RFA+Vac colorectal) (yellow) in this study, in comparison to RFA-treated colorectal cancer patients (RFA colorectal) (green) during the same period at Sir Run Run Shaw Hospital. Longer OS was observed for patients receiving additional neoantigen vaccination (p = 0.037). Survival data were compared by logrank test.
Figure 3
Figure 3
RFA+Vac patients showed better immune responses upon receiving iNeo-Vac-P01. (A)Ex vivo ELISpot assay results of each patient. Green triangle and red diamond represent the relative response rate, calculated per the ratios of peptides with positive ELISpot reads to all peptides immunized before and after vaccination, respectively. The bar chart shows the spot numbers of the peptide or peptide pool with best reads per 105 PBMCs. # Indicated that peptide pools were used for ELISpot assay for this patient; + indicated that relatively high abundant novel clones of peripheral T cells were detected for this patient after vaccination. (B, C) Data shown in Box and Whiskers (with minimum to maximum, showing all points and median). (B) Baseline and post-vaccination ELISpot spot counts per 105 PBMCs of RFA+Vac (yellow) and Vac (blue) patients. c) ELISpot spot counts per 105 PBMCs at baseline and post vaccination of the peak response for RFA+Vac patients with tint < 3 months, or 3 ≤ tint < 6 months. No significant difference was found between the two groups at baseline (blue and yellow); however, the difference was intensified post vaccination, as patients with tint < 3 months showed stronger peak response (red). Baseline response and peak response of all RFA+Vac patients showed statistically significant difference (p < 0.0051). (D) Flow cytometric analyses of RFA+Vac and Vac patients on T cell subsets. RFA+Vac and Vac patients were further divided into two subgroups: patients with good response (OS ≥ 12 months) and poor response (OS < 12 months); number of active CD4+ T cells (# per μL), and proportion of CD4+ or CD8+ T cells that expressed CTLA4 (%) at baseline, prime phase and boost phase were analyzed, respectively. (E) Flow cytometric analyses of RFA+Vac and Vac patients on cytokines including IFN-γ, IL-6 and TNF-α at baseline, prime phase, and boost phase. A t-test analysis was applied to indicate the significance in data difference with a two-tailed p value. ns, not significant (p > 0.05); *p ≤ 0.05; **p ≤ 0.01.
Figure 4
Figure 4
Case report of patient P015, who had multiple RFA treatments prior to vaccination. (A) The treatment scheme of patient P015. Two batches of iNeo-Vac-P01 were scheduled for this patient. (B) CT scan images of the tumor at different time points during the treatment. The red arrows indicated the corresponding tumor lesion. SLDs: sum of longest diameters. SLDs: sum of the longest diameters. (C, D)Ex vivo ELISpot assay of IFN-γ+ PBMCs during vaccination. (E) Increased abundance of peripheral neoantigen-specific T cell clones post vaccination as detected by TCR sequencing. (F) Multiplexed IF images of FFPE samples obtained from patient P015 pre- and post-vaccination. Signals of CD8, CD4 and Granzyme B are shown in fuchsias, green and yellow. Double stranded DNA is shown in blue. Shown on the right are the proportion (%) and density (counts per mm2) of CD4+ Granzyme B+ and CD8+ Granzyme B+ cells.
Figure 5
Figure 5
Combination treatment of RFA and neoantigen vaccination inhibited tumor growth and elicited robust local and systemic antitumor immune responses in mice. (A) Schematic illustration of the treatment mice received. Mouse models with unilateral or bilateral tumor(s) were established; mice with bilateral tumors received RFA treatment on the left flank tumor. (B, C) Tumor growth of mice receiving different treatments. Ablation of an irrelevant tumor (4T1) did not help inhibit the growth of CT26 tumor. Combinational use of RFA and iNeo-Vac-P01 (RFA+Vac) significantly slowed down the growth of CT26 tumor, and the inclusion of anti-PD-1 further enhanced this antitumor effect. (D) Flow cytometric analyses of PD-1 expression in tumor tissues (n=3). (E) Flow cytometric analyses of multifunctional T cells, gating on CD8+ perforin+ cells (n=3). (F) Flow cytometric analyses of cytotoxic T cells, gating on CD8+ IFN-γ+ cells (n=3). (G)Ex vivo ELISpot assay. Spleen cells were collected and re-stimulated by neoantigen peptides. Data shown in Mean ± SD; two-tailed p value was obtained through unpaired t test. ns, not significant (p > 0.05); **p ≤ 0.01; ***p ≤ 0.001; ****p ≤ 0.0001.

References

    1. Sun C, Xu S. Advances in personalized neoantigen vaccines for cancer immunotherapy. BioSci Trends (2020) 14(5):349–53. doi: 10.5582/bst.2020.03267
    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. doi: 10.1038/nmat4822
    1. Srivastava RM, Purohit TA, Chan TA. Diverse neoantigens and the development of cancer therapies. Semin Radiat Oncol (2020) 30(2):113–28. doi: 10.1016/j.semradonc.2019.12.001
    1. Ni Q, Zhang F, Liu Y, Wang Z, Yu G, Liang B, et al. . A bi-adjuvant nanovaccine that potentiates immunogenicity of neoantigen for combination immunotherapy of colorectal cancer. Sci Adv (2020) 6(12):eaaw6071. doi: 10.1126/sciadv.aaw6071
    1. Shae D, Baljon JJ, Wenbe M, Christov PP, Becker KW, Kumar A, et al. . Co-Delivery of peptide neoantigens and stimulator of interferon genes agonists enhances response to cancer vaccines. ACS Nano (2020) 14(8):9904–16. doi: 10.1021/acsnano.0c02765
    1. Baharom F, Ramirez-Valdez RA, Tobin KKS, Yamane H, Dutertre CA, Khalilnezhad A, et al. . Intravenous nanoparticle vaccination generates stem-like TCF1(+) neoantigen-specific CD8(+) T cells. Nat Immunol (2020) 22(1):41–52. doi: 10.1038/s41590-020-00810-3
    1. Lynn GM, Sedlik C, Baharom F, Zhu Y, Ramirez-Valdez RA, Coble VL, et al. . Peptide-TLR-7/8a conjugate vaccines chemically programmed for nanoparticle self-assembly enhance CD8 T-cell immunity to tumor antigens. Nat Biotechnol (2020) 38(3):320–32. doi: 10.1038/s41587-019-0390-x
    1. Qiu F, Becker JW, Knight FC, Baljon JJ, Sevimli S, Shae D, et al. . Poly(propylacrylic acid)-peptide nanoplexes as a platform for enhancing the immunogenicity of neoantigen cancer vaccines. Biomaterials (2018) 182:82–91. doi: 10.1016/j.biomaterials.2018.07.052
    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. doi: 10.1038/nature22991
    1. Fang Y, Mo F, Shou J, Wang H, Luo K, Zhang S, et al. . A pan-cancer clinical study of personalized neoantigen vaccine monotherapy in treating patients with various types of advanced solid tumors. Clin Cancer Res (2020) 26(17):4511. doi: 10.1158/1078-0432.CCR-19-2881
    1. Yin J, Dong J, Gao W, Wang Y. A case report of remarkable response to association of radiofrequency ablation with subsequent atezolizumab in stage IV nonsmall cell lung cancer. Med (Baltimore) (2018) 97(44):e13112. doi: 10.1097/MD.0000000000013112
    1. Zeng P, Shen D, Zeng CH, Chang XF, Teng GJ. Emerging opportunities for combining locoregional therapy with immune checkpoint inhibitors in hepatocellular carcinoma. Curr Oncol Rep (2020) 22(8):76. doi: 10.1007/s11912-020-00943-6
    1. Slovak R, Ludwig JM, Gettinger SN, Herbst RS, Kim SH. Immuno-thermal ablations - boosting the anticancer immune response. J ImmunoTher Cancer (2017) 5(1):78. doi: 10.1186/s40425-017-0284-8
    1. Fei Q, Pan Y, Lin W, Zhou Y, Yu X, Hou Z, et al. . High-dimensional single-cell analysis delineates radiofrequency ablation induced immune microenvironmental remodeling in pancreatic cancer. Cell Death Dis (2020) 11(7):589. doi: 10.1038/s41419-020-02787-1
    1. Shi L, Chen L, Wu C, Zhu Y, Xu B, Zheng X, et al. . PD-1 blockade boosts radiofrequency ablation-elicited adaptive immune responses against tumor. Clin Cancer Res (2016) 22(5):1173–84. doi: 10.1158/1078-0432.CCR-15-1352
    1. Duffy AG, Ulahannan SV, Makrova-Rusher O, Rahma O, Wedemeyer H, Pratt D, et al. . Tremelimumab in combination with ablation in patients with advanced hepatocellular carcinoma. J Hepatol (2017) 66(3):545–51. doi: 10.1016/j.jhep.2016.10.029
    1. Löffler MW, Nussbaum B, Jäger G, Jurmeister PS, Budczies J, Pereira PL, et al. . A non-interventional clinical trial assessing immune responses after radiofrequency ablation of liver metastases from colorectal cancer. Front Immunol (2019) 10:2526. doi: 10.3389/fimmu.2019.02526
    1. Heikkilä K, Ebrahim S, Lawlor DA. Systematic review of the association between circulating interleukin-6 (IL-6) and cancer. Eur J Cancer (2008) 44(7):937–45. doi: 10.1016/j.ejca.2008.02.047
    1. Silva EM, Mariano VS, Pastrez PRA, Pinto MC, Gastro AG, Syrjanen KJ, et al. . High systemic IL-6 is associated with worse prognosis in patients with non-small cell lung cancer. PloS One (2017) 12(7):e0181125. doi: 10.1371/journal.pone.0181125
    1. Kumari N, Dwarakanath BS, Das A, Bhatt AN. Role of interleukin-6 in cancer progression and therapeutic resistance. Tumor Biol (2016) 37(9):11553–72. doi: 10.1007/s13277-016-5098-7
    1. Shao C, Yang M, Pan Y, Xie D, Chen B, Ren S, et al. . Case report: Abscopal effect of microwave ablation in a patient with advanced squamous NSCLC and resistance to immunotherapy. Front Immunol (2021) 12:696749. doi: 10.3389/fimmu.2021.696749
    1. Iwai T, Oebisu N, Hoshi M, Orita K, Yamamoto A, Hamamoto S, et al. . Promising abscopal effect of combination therapy with thermal tumour ablation and intratumoural OK-432 injection in the rat osteosarcoma model. Sci Rep (2020) 10(1):9679. doi: 10.1038/s41598-020-66934-6
    1. Huang S, Li T, Chen Y, Liu J, Wang Y, Yang C, et al. . Microwave ablation combined with anti-PD-1 therapy enhances systemic antitumor immunity in a multitumor murine model of Hepa1-6. Int J Hyperther (2022) 39(1):278–86. doi: 10.1080/02656736.2022.2032406
    1. Selby MJ, Engelhardt JJ, Quigley M, Henning KA, Chen T, Srinivasan M, et al. . Anti-CTLA-4 antibodies of IgG2a isotype enhance antitumor activity through reduction of intratumoral regulatory T cells. Cancer Immunol Res (2013) 1(1):32–42. doi: 10.1158/2326-6066.CIR-13-0013
    1. Simon S, Labarriere N. PD-1 expression on tumor-specific T cells: Friend or foe for immunotherapy? Oncoimmunology (2017) 7(1):e1364828. doi: 10.1080/2162402X.2017.1364828
    1. Gros A, Parkhurst MR, Tran E, Pasetto A, Robbins PF, Ilyas S, et al. . Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nat Med (2016) 22(4):433–8. doi: 10.1038/nm.4051
    1. Cros A, Robbins PF, Yao X, Li YF, Turcotte S, Tran E, et al. . PD-1 identifies the patient-specific CD8+tumor-reactive repertoire infiltrating human tumors. J Clin Invest (2014) 124(5):2246–59. doi: 10.1172/JCI73639
    1. Voskoboinik I, Whisstock JC, Trapani JA. Perforin and granzymes: function, dysfunction and human pathology. Nat Rev Immunol (2015) 15(6):388–400. doi: 10.1038/nri3839
    1. Shiina S, Tateishi R, Arano T, Uchino K, Enoku K, Nakagawa H, et al. . Radiofrequency ablation for hepatocellular carcinoma: 10-year outcome and prognostic factors. Am J Gastroenterol (2012) 107(4):569–77; quiz 578. doi: 10.1038/ajg.2011.425
    1. Ahmed M, Kumar G, Moussa M, Wang Y, Rozenblum N, Galun E , et al. . Hepatic radiofrequency ablation-induced stimulation of distant tumor growth is suppressed by c-met inhibition. Radiology (2016) 279(1):103–17. doi: 10.1148/radiol.2015150080
    1. Beham B, Fazio PD, Michl P, Neureiter D, Kemmerling R, Hahn EG, et al. . Additive antitumour response to the rabbit VX2 hepatoma by combined radio frequency ablation and toll like receptor 9 stimulation. Gut (2016) 65(1):134–43. doi: 10.1136/gutjnl-2014-308286
    1. Salomon N, Vascotto F, Selmi A, Vormehr M, Quinkhardt J, Bukur T, et al. . A liposomal RNA vaccine inducing neoantigen-specific CD4+ T cells augments the antitumor activity of local radiotherapy in mice. OncoImmunology (2020) 9(1):1771925. doi: 10.1080/2162402X.2020.1771925
    1. Cutsem EV, Martinelli E, Cascinu S, Sobrero A, Banzi M, Seitz J, et al. . Regorafenib for patients with metastatic colorectal cancer who progressed after standard therapy: Results of the Large, single-arm, open-label phase IIIb CONSIGN study. Oncologist (2019) 24(2):185–92. doi: 10.1634/theoncologist.2018-0072
    1. Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. . Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med (2010) 363(8):711–23. doi: 10.1056/NEJMoa1003466
    1. Lu S. A Randomized, double-blind, placebo-controlled, multicenter study of fruquintinib in Chinese patients with advanced nonsquamous non-small-cell lung cancer - the FALUCA study. Lung Cancer (2020) 146:252–62. doi: 10.1016/j.lungcan.2020.06.016
    1. Wang-Gillam A, Li C, Bodoky G, Dean A, Shan Y, Jameson G, et al. . Nanoliposomal irinotecan with fluorouracil and folinic acid in metastatic pancreatic cancer after previous gemcitabine-based therapy (NAPOLI-1): a global, randomised, open-label, phase 3 trial. Lancet (2016) 387(10018):545–57. doi: 10.1016/S0140-6736(15)00986-1
    1. Lamarca A, Palmer DH, Wasan HS, Ross PJ, Ma YT, Arora A, et al. . Second-line FOLFOX chemotherapy versus active symptom control for advanced biliary tract cancer (ABC-06): a phase 3, open-label, randomised, controlled trial. Lancet Oncol (2021) 22(5):690–701. doi: 10.1016/S1470-2045(21)00027-9
    1. Park IH, Im S, Jung KH, Sohn JH, Park YH, Lee KS, et al. . Randomized open label phase III trial of irinotecan plus capecitabine versus capecitabine monotherapy in patients with metastatic breast cancer previously treated with anthracycline and taxane: PROCEED trial (KCSG BR 11-01). Cancer Res Treat (2019) 51(1):43–52. doi: 10.4143/crt.2017.562
    1. Hamanishi J, Takeshima N, Katsumata N, Ushijima K, Kimura T, Takeuchi S, et al. . Nivolumab versus gemcitabine or pegylated liposomal doxorubicin for patients with platinum-resistant ovarian cancer: Open-label, randomized trial in Japan. (NINJA) J Clin Oncol (2021) 39(33):3671–81. doi: 10.1200/JCO.21.00334
    1. Bellone G, Smirne C, Mauri FA, Tonel E, Carbone A, Buffolino A, et al. . Cytokine expression profile in human pancreatic carcinoma cells and in surgical specimens: implications for survival. Cancer Immunol Immunother (2006) 55(6):684–98. doi: 10.1007/s00262-005-0047-0
    1. Chambers CA, Kuhns MS, Egen JG, Allison JP. CTLA-4-Mediated inhibition in regulation of T cell responses: Mechanisms and manipulation in tumor immunotherapy. Annu Rev Immunol (2001) 19(1):565–94. doi: 10.1146/annurev.immunol.19.1.565

Source: PubMed

3
S'abonner