Irreversible electroporation plus allogenic Vγ9Vδ2 T cells enhances antitumor effect for locally advanced pancreatic cancer patients

Mao Lin, Xiaoyan Zhang, Shuzhen Liang, Haihua Luo, Mohammed Alnaggar, Aihua Liu, Zhinan Yin, Jibing Chen, Lizhi Niu, Yong Jiang, Mao Lin, Xiaoyan Zhang, Shuzhen Liang, Haihua Luo, Mohammed Alnaggar, Aihua Liu, Zhinan Yin, Jibing Chen, Lizhi Niu, Yong Jiang

Abstract

Immunotherapy has limited efficacy against locally advanced pancreatic cancer (LAPC) due to the presence of an immunosuppressive microenvironment (ISM). Irreversible electroporation (IRE) can not only induce immunogenic cell death, but also alleviate immunosuppression. This study aimed to investigate the antitumor efficacy of IRE plus allogeneic γδ T cells in LAPC patients. A total of 62 patients who met the eligibility criteria were enrolled in this trial, then randomized into two groups (A: n = 30 and B: n = 32). All patients received IRE therapy and after receiving IRE, the group A patients received at least two cycles of γδ T-cell infusion as one course continuously. Group A patients had better survival than group B patients (median OS: 14.5 months vs. 11 months; median PFS: 11 months vs. 8.5 months). Moreover, the group A patients treated with multiple courses of γδ T-cell infusion had longer OS (17 months) than those who received a single course (13.5 months). IRE combined with allogeneic γδ T-cell infusion is a promising strategy to enhance the antitumor efficacy in LAPC patients, yielding extended survival benefits.ClinicalTrials.gov ID: NCT03180437.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
The clinical trial profile and treatment schedule. a The clinical trial profile. A total of 176 PDAC patients from 12 countries were screened for enrollment, of which 70 (39.8%) were assessed for LAPC. During screening, five participants were excluded because of compromised liver function (n = 2), severe coronary disease (n = 1), allergy to contrast media (n = 1), and chemotherapy or radiation therapy ≤6 week (n = 1). After systemic therapy, three participants were excluded because of downstaging to resection (n = 2) and withdrew consent (n = 1). Overall, the LAPC patients were screened for the eligibility criteria, and 62 (35.2%) were finally enrolled in the study and assigned randomly to group A (n = 30) or group B (n = 32). b Clinical treatment schedule. Patients in group A received IRE plus one to three courses of allogeneic γδ T cells; one γδ T-cell treatment course was designed to contain two cycles, totaling six γδ T-cell infusions in 28 days, i.e., days 12, 13, and 14 for the first cycle and days 26, 27, and 28 for the second cycle. Patients in group B just received IRE (n = 62)
Fig. 2
Fig. 2
All-cause adverse events in the safety population. a All-cause adverse events with a difference of no <5% between the study groups. b Proportions of patients with treatment-related adverse events presented by grade. There was no significant difference between the two groups. n = 62. P > 0.05. Chi-square test
Fig. 3
Fig. 3
Evaluation of tumor markers before treatment and 90 days after treatment. a The level of CA19-9 was quantitated by a chemiluminescent immunoassay. n = 62. Data are shown as individual data points with Box and Whiskers graph (bottom: 25%; top: 75%; line: median; whiskers: min to max). Comparison within groups: ***P < 0.001; comparison between groups: ###P < 0.001. Mann–Whitney test or Wilcoxon test. b The number of CD45−CK+CD326+cells (CTCs) was acquired with a FACSCantoTM II. Data are shown as individual data points with Box and Whiskers graph (bottom: 25%; top: 75%; line: median; whiskers: min to max). Comparison within groups: ***P < 0.001; comparison between groups: #P < 0.05. Two-tailed Student’s t test (n = 62)
Fig. 4
Fig. 4
Detection of immune parameters before treatment and 90 days after treatment. ac Flow cytometry was performed with the 6-color TBNK reagent to detect lymphocytes in the blood. df Flow cytometry was performed with the Cytometric Bead Array Human Th1/Th2 Cytokine Kit II to detect cytokines in the blood. Data are shown as individual data points with Box and Whiskers graph (bottom: 25%; top: 75%; line: median; whiskers: min to max). Comparison within groups: *P < 0.05; **P < 0.01; ***P < 0.001; comparison between groups: #P < 0.05; ##P < 0.01; ###P < 0.001. Two-tailed Student’s t test (n = 62)
Fig. 5
Fig. 5
Expression of NKG2D, PD-1, and CD44 before treatment and 90 days after treatment. Surface receptors NKG2D, PD-1, and CD44 on γδ T cells were stained with the monoclonal antibodies of mouse anti-human NKG2D, PD-1, and CD44, then analyzed by flow cytometry with a FACSCantoTM II. MFI mean fluorescence intensity. Data are shown as individual data points with Scatter dot plot (line at: mean with SD). Comparison within groups: **P < 0.01; ***P < 0.001; comparison between groups: #P < 0.05; ##P < 0.01. Two-tailed Student’s t test (n = 62)
Fig. 6
Fig. 6
Changes of PS score and TMTD. a PS scores were calculated by medical staffs’ observation before and 3 months after treatment. b TMTD was evaluated by tumor imaging system before and 3 months after treatment. Data are shown as individual data points with Scatter dot plot (line at: mean with SD). Comparison within groups: *P < 0.05; ***P < 0.001; comparison between groups: ###P < 0.001. Mann–Whitney test or Wilcoxon test (n = 62)
Fig. 7
Fig. 7
Kaplan–Meier survival curves. a Graph showed OS from the time of treatment in total population (n = 62). b Graph showed OS from the time of treatment in group A patients who received a single course of γδ T-cell infusion or multiple courses of γδ T-cell infusions (n = 30). c Graph showed PFS from the time of treatment in total population (n = 62). d Graph showed PFS from the time of treatment in group A patients who received a single course of γδ T-cell infusion or multiple courses of γδ T-cell infusions (n = 30)

References

    1. Balaban EP, et al. Locally advanced, unresectable pancreatic cancer: american society of clinical oncology clinical practice guideline. J. Clin. Oncol. 2016;34:2654–2668.
    1. He C, et al. Immunomodulatory effect after irreversible electroporation in patients with locally advanced pancreatic cancer. J. Oncol. 2019;2019:9346017.
    1. Ruarus AH, et al. Percutaneous irreversible electroporation in locally advanced and recurrent pancreatic cancer (PANFIRE-2): a multicenter, prospective, single-arm, phase II study. Radiology. 2020;294:212–220.
    1. Narayanan JSS, et al. Irreversible electroporation combined with checkpoint blockade and TLR7 stimulation induces antitumor immunity in a murine pancreatic cancer model. Cancer Immunol. Res. 2019;7:1714–1726.
    1. Zhao J, et al. Irreversible electroporation reverses resistance to immune checkpoint blockade in pancreatic cancer. Nat. Commun. 2019;10:899.
    1. Clark CE, et al. Dynamics of the immune reaction to pancreatic cancer from inception to invasion. Cancer Res. 2007;67:9518–9527.
    1. Feig C, et al. The pancreas cancer microenvironment. Clin. Cancer Res. 2012;18:4266–4276.
    1. Martinez-Bosch N, Vinaixa J, Navarro P. Immune evasion in pancreatic cancer: from mechanisms to therapy. Cancers. 2018;10:6.
    1. Martin RC, II, McFarland K, Ellis S, Velanovich V. Irreversible electroporation in locally advanced pancreatic cancer: potential improved overall survival. Ann. Surg. Oncol. 2013;20(Suppl 3):S443–449.
    1. Varshney S, et al. Radiofrequency ablation of unresectable pancreatic carcinoma: feasibility, efficacy and safety. JOP. 2006;7:74–78.
    1. Moir J, et al. Systematic review of irreversible electroporation in the treatment of advanced pancreatic cancer. Eur. J. Surg. Oncol. 2014;40:1598–1604.
    1. Martin RC, II, et al. Treatment of 200 locally advanced (stage III) pancreatic adenocarcinoma patients with irreversible electroporation: safety and efficacy. Ann. Surg. 2015;262:486–494.
    1. Rubinsky B, Onik G, Mikus P. Irreversible electroporation: a new ablation modality–clinical implications. Technol. Cancer Res. Treat. 2007;6:37–48.
    1. Lin M, et al. Percutaneous irreversible electroporation combined with allogeneic natural killer cell immunotherapy for patients with unresectable (stage III/IV) pancreatic cancer: a promising treatment. J. Cancer Res. Clin. Oncol. 2017;143:2607–2618.
    1. Lin M, et al. Short-term clinical efficacy of percutaneous irreversible electroporation combined with allogeneic natural killer cell for treating metastatic pancreatic cancer. Immunol. Lett. 2017;186:20–27.
    1. Scheffer HJ, et al. Ablation of locally advanced pancreatic cancer with percutaneous irreversible electroporation: results of the phase I/II PANFIRE study. Radiology. 2017;282:585–597.
    1. Bhutiani N, et al. Irreversible electroporation enhances delivery of gemcitabine to pancreatic adenocarcinoma. J. Surg. Oncol. 2016;114:181–186.
    1. Bulvik BE, et al. Irreversible electroporation versus radiofrequency ablation: a comparison of local and systemic effects in a small-animal model. Radiology. 2016;280:413–424.
    1. Scheffer HJ, et al. Irreversible electroporation of locally advanced pancreatic cancer transiently alleviates immune suppression and creates a window for antitumor T cell activation. Oncoimmunology. 2019;8:1652532.
    1. Hu Y, et al. Selenium nanoparticles as new strategy to potentiate gammadelta T cell anti-tumor cytotoxicity through upregulation of tubulin-alpha acetylation. Biomaterials. 2019;222:119397.
    1. Neelapu SS, et al. Chimeric antigen receptor T-cell therapy—assessment and management of toxicities. Nat. Rev. Clin. Oncol. 2018;15:47–62.
    1. Silva-Santos B, Mensurado S, Coffelt SB. Gammadelta T cells: pleiotropic immune effectors with therapeutic potential in cancer. Nat. Rev. Cancer. 2019;19:392–404.
    1. Zajdel A, Kalucka M, Chodurek E, Wilczok A. DHA but not AA enhances cisplatin cytotoxicity in ovarian cancer cells. Nutr. Cancer. 2018;70:1118–1125.
    1. Dudley ME, et al. Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J. Clin. Oncol. 2005;23:2346–2357.
    1. Bouet-Toussaint F, et al. Vgamma9Vdelta2 T cell-mediated recognition of human solid tumors. Potential for immunotherapy of hepatocellular and colorectal carcinomas. Cancer Immunol. Immunother. 2008;57:531–539.
    1. Dieli F, et al. Targeting human {gamma}delta} T cells with zoledronate and interleukin-2 for immunotherapy of hormone-refractory prostate cancer. Cancer Res. 2007;67:7450–7457.
    1. Meraviglia S, et al. In vivo manipulation of Vgamma9Vdelta2 T cells with zoledronate and low-dose interleukin-2 for immunotherapy of advanced breast cancer patients. Clin. Exp. Immunol. 2010;161:290–297.
    1. Alnaggar M, et al. Allogenic vgamma9vdelta2 T cell as new potential immunotherapy drug for solid tumor: a case study for cholangiocarcinoma. J. Immunother. Cancer. 2019;7:36.
    1. Xiang Z, Tu W. Dual face of vgamma9vdelta2-T cells in tumor immunology: anti- versus pro-tumoral activities. Front. Immunol. 2017;8:1041.
    1. Lin M, et al. Pembrolizumab plus allogeneic NK cells in advanced non-small cell lung cancer patients. J. Clin. Investig. 2020;130:2560–2569.
    1. Okazaki T, et al. A rheostat for immune responses: the unique properties of PD-1 and their advantages for clinical application. Nat. Immunol. 2013;14:1212–1218.
    1. Mansson C, et al. Percutaneous irreversible electroporation for treatment of locally advanced pancreatic cancer following chemotherapy or radiochemotherapy. Eur. J. Surg. Oncol. 2016;42:1401–1406.
    1. Narayanan G, et al. Percutaneous image-guided irreversible electroporation for the treatment of unresectable, locally advanced pancreatic adenocarcinoma. J. Vasc. Interv. Radiol. 2017;28:342–348.
    1. Brudno JN, Kochenderfer JN. Chimeric antigen receptor T-cell therapies for lymphoma. Nat. Rev. Clin. Oncol. 2018;15:31–46.
    1. Thind K, Padrnos LJ, Ramanathan RK, Borad MJ. Immunotherapy in pancreatic cancer treatment: a new frontier. Ther. Adv. Gastroenterol. 2017;10:168–194.
    1. Yang Y, et al. Safety and short-term efficacy of irreversible electroporation and allogenic natural killer cell immunotherapy combination in the treatment of patients with unresectable primary liver cancer. Cardiovasc. Interv. Radiol. 2019;42:48–59.
    1. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 2015;348:62–68.
    1. Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 2018;8:1069–1086.
    1. Willcox BE, Willcox CR. Gammadelta TCR ligands: the quest to solve a 500-million-year-old mystery. Nat. Immunol. 2019;20:121–128.
    1. Geller MA, Miller JS. Use of allogeneic NK cells for cancer immunotherapy. Immunotherapy. 2011;3:1445–1459.
    1. Field W, Rostas JW, Martin RCG. Quality of life assessment for patients undergoing irreversible electroporation (IRE) for treatment of locally advanced pancreatic cancer (LAPC) Am. J. Surg. 2019;218:571–578.
    1. Rossi C, et al. Boosting gammadelta T cell-mediated antibody-dependent cellular cytotoxicity by PD-1 blockade in follicular lymphoma. Oncoimmunology. 2019;8:1554175.
    1. Benyamine A, et al. BTN3A is a prognosis marker and a promising target for Vgamma9Vdelta2 T cells based-immunotherapy in pancreatic ductal adenocarcinoma (PDAC) Oncoimmunology. 2017;7:e1372080.
    1. Chauvin C, et al. NKG2D controls natural reactivity of vgamma9vdelta2 T lymphocytes against mesenchymal glioblastoma cells. Clin. Cancer Res. 2019;25:7218–7228.
    1. Capietto AH, Martinet L, Fournie JJ. Stimulated gammadelta T cells increase the in vivo efficacy of trastuzumab in HER-2+ breast cancer. J. Immunol. 2011;187:1031–1038.
    1. Zeng J, et al. The safety and efficacy of irreversible electroporation for large hepatocellular carcinoma. Technol. Cancer Res. Treat. 2017;16:120–124.

Source: PubMed

3
订阅