19
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

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

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          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://clinicaltrials.gov/, identifier NCT03662815.

          Related collections

          Most cited references41

          • Record: found
          • Abstract: found
          • Article: not found

          Improved Survival with Ipilimumab in Patients with Metastatic Melanoma

          An improvement in overall survival among patients with metastatic melanoma has been an elusive goal. In this phase 3 study, ipilimumab--which blocks cytotoxic T-lymphocyte-associated antigen 4 to potentiate an antitumor T-cell response--administered with or without a glycoprotein 100 (gp100) peptide vaccine was compared with gp100 alone in patients with previously treated metastatic melanoma. A total of 676 HLA-A*0201-positive patients with unresectable stage III or IV melanoma, whose disease had progressed while they were receiving therapy for metastatic disease, were randomly assigned, in a 3:1:1 ratio, to receive ipilimumab plus gp100 (403 patients), ipilimumab alone (137), or gp100 alone (136). Ipilimumab, at a dose of 3 mg per kilogram of body weight, was administered with or without gp100 every 3 weeks for up to four treatments (induction). Eligible patients could receive reinduction therapy. The primary end point was overall survival. The median overall survival was 10.0 months among patients receiving ipilimumab plus gp100, as compared with 6.4 months among patients receiving gp100 alone (hazard ratio for death, 0.68; P<0.001). The median overall survival with ipilimumab alone was 10.1 months (hazard ratio for death in the comparison with gp100 alone, 0.66; P=0.003). No difference in overall survival was detected between the ipilimumab groups (hazard ratio with ipilimumab plus gp100, 1.04; P=0.76). Grade 3 or 4 immune-related adverse events occurred in 10 to 15% of patients treated with ipilimumab and in 3% treated with gp100 alone. There were 14 deaths related to the study drugs (2.1%), and 7 were associated with immune-related adverse events. Ipilimumab, with or without a gp100 peptide vaccine, as compared with gp100 alone, improved overall survival in patients with previously treated metastatic melanoma. Adverse events can be severe, long-lasting, or both, but most are reversible with appropriate treatment. (Funded by Medarex and Bristol-Myers Squibb; ClinicalTrials.gov number, NCT00094653.)
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            An immunogenic personal neoantigen vaccine for patients with melanoma

            Effective anti-tumour immunity in humans has been associated with the presence of T cells directed at cancer neoantigens, a class of HLA-bound peptides that arise from tumour-specific mutations. They are highly immunogenic because they are not present in normal tissues and hence bypass central thymic tolerance. Although neoantigens were long-envisioned as optimal targets for an anti-tumour immune response, their systematic discovery and evaluation only became feasible with the recent availability of massively parallel sequencing for detection of all coding mutations within tumours, and of machine learning approaches to reliably predict those mutated peptides with high-affinity binding of autologous human leukocyte antigen (HLA) molecules. We hypothesized that vaccination with neoantigens can both expand pre-existing neoantigen-specific T-cell populations and induce a broader repertoire of new T-cell specificities in cancer patients, tipping the intra-tumoural balance in favour of enhanced tumour control. Here we demonstrate the feasibility, safety, and immunogenicity of a vaccine that targets up to 20 predicted personal tumour neoantigens. Vaccine-induced polyfunctional CD4+ and CD8+ T cells targeted 58 (60%) and 15 (16%) of the 97 unique neoantigens used across patients, respectively. These T cells discriminated mutated from wild-type antigens, and in some cases directly recognized autologous tumour. Of six vaccinated patients, four had no recurrence at 25 months after vaccination, while two with recurrent disease were subsequently treated with anti-PD-1 (anti-programmed cell death-1) therapy and experienced complete tumour regression, with expansion of the repertoire of neoantigen-specific T cells. These data provide a strong rationale for further development of this approach, alone and in combination with checkpoint blockade or other immunotherapies.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Anti-CTLA-4 antibodies of IgG2a isotype enhance antitumor activity through reduction of intratumoral regulatory T cells.

              Antitumor activity of CTLA-4 antibody blockade is thought to be mediated by interfering with the negative regulation of T-effector cell (Teff) function resulting from CTLA-4 engagement by B7-ligands. In addition, a role for CTLA-4 on regulatory T cells (Treg), wherein CTLA-4 loss or inhibition results in reduced Treg function, may also contribute to antitumor responses by anti-CTLA-4 treatment. We have examined the role of the immunoglobulin constant region on the antitumor activity of anti-CTLA-4 to analyze in greater detail the mechanism of action of anti-CTLA-4 antibodies. Anti-CTLA-4 antibody containing the murine immunoglobulin G (IgG)2a constant region exhibits enhanced antitumor activity in subcutaneous established MC38 and CT26 colon adenocarcinoma tumor models compared with anti-CTLA-4 containing the IgG2b constant region. Interestingly, anti-CTLA-4 antibodies containing mouse IgG1 or a mutated mouse IgG1-D265A, which eliminates binding to all Fcγ receptors (FcγR), do not show antitumor activity in these models. Assessment of Teff and Treg populations at the tumor and in the periphery showed that anti-CTLA-4-IgG2a mediated a rapid and dramatic reduction of Tregs at the tumor site, whereas treatment with each of the isotypes expanded Tregs in the periphery. Expansion of CD8(+) Teffs is observed with both the IgG2a and IgG2b anti-CTLA-4 isotypes, resulting in a superior Teff to Treg ratio for the IgG2a isotype. These data suggest that anti-CTLA-4 promotes antitumor activity by a selective reduction of intratumoral Tregs along with concomitant activation of Teffs. ©2013 AACR.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Immunol
                Front Immunol
                Front. Immunol.
                Frontiers in Immunology
                Frontiers Media S.A.
                1664-3224
                29 September 2022
                2022
                : 13
                : 1000681
                Affiliations
                [1] 1 Sir Run Run Shaw Hospital, Zhejiang University School of Medicine , Hangzhou, China
                [2] 2 Hangzhou Neoantigen Therapeutics Co. , Ltd., Hangzhou, China
                [3] 3 College of Pharmaceutical Sciences, Zhejiang University , Hangzhou, China
                [4] 4 Vancouver Prostate Centre, University of British Columbia , Vancouver, BC, Canada
                [5] 5 Hangzhou AI-Force Therapeutics Co. , Ltd., Hangzhou, China
                [6] 6 Zhejiang California International Nanosystems Institute, Zhejiang University , Hangzhou, China
                [7] 7 School of Chemistry and Molecular Biosciences, The University of Queensland , Brisbane, QLD, Australia
                [8] 8 Hangzhou AI-Nano Therapeutics Co., Ltd. , Hangzhou, China
                [9] 9 Shulan (Hangzhou) Hospital , Hangzhou, China
                [10] 10 ZJU-Hangzhou Global Scientific and Technological Innovation Center , Hangzhou, China
                Author notes

                Edited by: Jai Rudra, Washington University in St. Louis, United States

                Reviewed by: Rupsa Basu, Humane Genomics, United States; Philippe Rochigneux, Institut Paoli-Calmettes (IPC), France; Wayne Robert Thomas, University of Western Australia, Australia

                *Correspondence: Hongming Pan, panhongming@ 123456zju.edu.cn ; Shuqing Chen, chenshuqing@ 123456zju.edu.cn ; Yong Fang, fangyong@ 123456zju.edu.cn

                †These authors share first authorship

                This article was submitted to Vaccines and Molecular Therapeutics, a section of the journal Frontiers in Immunology

                Article
                10.3389/fimmu.2022.1000681
                9559398
                36248865
                95cd750e-53c4-4a50-9269-c819f30ab511
                Copyright © 2022 Shou, Mo, Zhang, Lu, Han, Liu, Qiu, Li, Han, Ma, Guo, Guo, Huang, Zhang, Ye, Pan, Chen and Fang

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 22 July 2022
                : 07 September 2022
                Page count
                Figures: 5, Tables: 2, Equations: 0, References: 41, Pages: 16, Words: 8058
                Funding
                Funded by: National Natural Science Foundation of China , doi 10.13039/501100001809;
                Award ID: 81702809, 81872238
                Funded by: Medical Science and Technology Project of Zhejiang Province , doi 10.13039/501100017594;
                Award ID: 2016ZDB007, 2017ZD021
                Funded by: National Natural Science Foundation of China , doi 10.13039/501100001809;
                Award ID: U20A20409
                Funded by: Natural Science Foundation of Zhejiang Province , doi 10.13039/501100004731;
                Award ID: LY13H160013, LQ16H160003
                Funded by: Health Commission of Zhejiang Province , doi 10.13039/501100014996;
                Award ID: 2016KYA115
                Funded by: Medical Science and Technology Project of Zhejiang Province , doi 10.13039/501100017594;
                Award ID: 2017197380
                Funded by: Chinese Society of Clinical Oncology , doi 10.13039/501100009812;
                Award ID: Y-QL2019-0316, Y-MSD2020-0314, XS022
                Categories
                Immunology
                Original Research

                Immunology
                neoantigen vaccine,radiofrequency ablation,immune checkpoint inhibition,cancer,immunotherapy

                Comments

                Comment on this article