1
|
Li Y, Lam SSK, Gao Y, Shore E, Anderson DW, Hode T, Martin RC. Enhancing the Immunotherapeutic Effect by IP-001 and Irreversible Electroporation in Mouse Oligometastatic Models of Pancreatic Adenocarcinoma. Ann Surg Oncol 2025; 32:2786-2798. [PMID: 39739260 DOI: 10.1245/s10434-024-16742-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/10/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND This study aimed to evaluate the immunotherapeutic effect of irreversible electroporation (IRE) and IP-001 in pancreatic adenocarcinoma with metastasis. METHODS Orthotopic models of pancreatic adenocarcinoma with hepatic oligometastasis were established by implantation of tumor tissues (derived from Pan02 or KPC cells) size 2 mm3 into the pancreas and left liver lobe in C57BL/6J mice. One week after implantation, the tumor-burden mice were subjected to saline control, IRE, IP-001, and IRE+IP-001. For IRE therapy (1000 V, 0.1 ms, 10 pulses administered 10 times), the pancreas tumor was treated, whereas the oligometastasis was untreated as the IRE off-target tumor. Intratumoral administration of IP-001(0.4 ml/kg) was performed. RESULTS In the KPC oligometastatic model, IRE+IP-001 therapy significantly suppressed the growth of oligometastatic tumor. Flow cytometry showed significantly increased tumor-infiltrating lymphocytes (TILs) (e.g., CD8+ cytotoxic T lymphocytes) and significantly increased monocytes/macrophages in the oligometastatic tumor tissues from IRE+IP-001 treatment compared with the sham control. Significantly decreased Treg cells and tumor-associated macrophages (TAMs) also were found in the oligometastatic tumor tissues from IRE+IP-001 treatment compared with the sham control. In the Pan02 oligometastatic model, both IRE+IP-001 therapy and IRE+anti-PD-L1 immunotherapy significantly suppressed the growth of oligometastatic tumor, which was associated with the increased CD8+ cytotoxic T lymphocytes. However, increased monocytes/macrophages were found in the mice that had IRE+IP-001 therapy, but not in the mice that had IRE+anti-PD-L1 immunotherapy. CONCLUSION The study provided compelling evidence for the efficacy of IRE&IP-001 therapy in suppressing pancreatic tumors, including off-target oligometastatic lesions. The observed off-target effect underscores the importance of systemic immune activation in achieving effective tumor control.
Collapse
Affiliation(s)
- Yan Li
- Division of Surgical Oncology, Department of Surgery, School of Medicine, University of Louisville, Louisville, KY, USA.
| | | | - Yonglin Gao
- Division of Surgical Oncology, Department of Surgery, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Emily Shore
- Division of Surgical Oncology, Department of Surgery, School of Medicine, University of Louisville, Louisville, KY, USA
| | | | | | - Robert C Martin
- Division of Surgical Oncology, Department of Surgery, School of Medicine, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
2
|
Zeng C, Hua S, Zhou J, Zeng T, Chen J, Su L, Jiang A, Zhou M, Tang Z. Oral Microalgae-Based Biosystem to Enhance Irreversible Electroporation Immunotherapy in Hepatocellular Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409381. [PMID: 39874200 PMCID: PMC12005737 DOI: 10.1002/advs.202409381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/22/2024] [Indexed: 01/30/2025]
Abstract
Irreversible electroporation (IRE) is a novel local tumor ablation technique that can potentially stimulate immune responses. However, IRE alone cannot effectively activate the immune system or prevent distant metastases. Therefore, this study utilized the biocompatibility of Chlorella vulgaris (C. vulgaris) and polydopamine (PDA) adhesive properties to encapsulate a PD-1 inhibitor (PI). The PDA coating protects the drug from degradation by stomach acid and enhances its intestinal absorption. This carrier demonstrates excellent in vivo drug release control and biodistribution, significantly increasing the oral bioavailability of PI. Combining IRE with this natural carrier significantly improves the therapeutic efficacy, which increases the local drug concentration and activates the immune system. This system demonstrates significantly improved therapeutic efficacy against local tumors compared with PI or IRE alone and significantly reduces PI-associated side effects. A convenient oral delivery system is developed using this readily available natural micro-carrier that not only improves the therapeutic effect of IRE but also mitigates its adverse effects, indicating significant potential for clinical applications. This discovery offers a new strategy for hepatocellular carcinoma treatment with the potential to improve patient outcomes.
Collapse
Affiliation(s)
- Cheng Zeng
- Department of SurgeryCenter for Cancer Medicinethe Fourth Affiliated Hospital of School of MedicineInternational School of MedicineInternational Institutes of MedicineZhejiang UniversityYiwu322000China
- Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancerthe Fourth Affiliated Hospital, Zhejiang University School of MedicineYiwu322000China
| | - Shiyuan Hua
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute)Zhejiang UniversityHaining314400China
- Institute of Translational MedicineZhejiang UniversityHangzhou310029China
- Zhejiang University‐Ordos City Etuoke Banner Joint Research CenterZhejiang UniversityHaining314400China
- The National Key Laboratory of Biobased Transportation Fuel TechnologyZhejiang UniversityHangzhou310027China
| | - Jiayu Zhou
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute)Zhejiang UniversityHaining314400China
- School of MedicineShihezi UniversityShiheziXinjiang832002China
| | - Tangye Zeng
- Department of SurgeryCenter for Cancer Medicinethe Fourth Affiliated Hospital of School of MedicineInternational School of MedicineInternational Institutes of MedicineZhejiang UniversityYiwu322000China
- Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancerthe Fourth Affiliated Hospital, Zhejiang University School of MedicineYiwu322000China
| | - Jianke Chen
- Department of SurgeryCenter for Cancer Medicinethe Fourth Affiliated Hospital of School of MedicineInternational School of MedicineInternational Institutes of MedicineZhejiang UniversityYiwu322000China
| | - Lijian Su
- Department of SurgeryCenter for Cancer Medicinethe Fourth Affiliated Hospital of School of MedicineInternational School of MedicineInternational Institutes of MedicineZhejiang UniversityYiwu322000China
| | - Angfeng Jiang
- Department of SurgeryCenter for Cancer Medicinethe Fourth Affiliated Hospital of School of MedicineInternational School of MedicineInternational Institutes of MedicineZhejiang UniversityYiwu322000China
- Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancerthe Fourth Affiliated Hospital, Zhejiang University School of MedicineYiwu322000China
| | - Min Zhou
- Department of SurgeryCenter for Cancer Medicinethe Fourth Affiliated Hospital of School of MedicineInternational School of MedicineInternational Institutes of MedicineZhejiang UniversityYiwu322000China
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute)Zhejiang UniversityHaining314400China
- Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancerthe Fourth Affiliated Hospital, Zhejiang University School of MedicineYiwu322000China
- Institute of Translational MedicineZhejiang UniversityHangzhou310029China
- Zhejiang University‐Ordos City Etuoke Banner Joint Research CenterZhejiang UniversityHaining314400China
- The National Key Laboratory of Biobased Transportation Fuel TechnologyZhejiang UniversityHangzhou310027China
| | - Zhe Tang
- Department of SurgeryCenter for Cancer Medicinethe Fourth Affiliated Hospital of School of MedicineInternational School of MedicineInternational Institutes of MedicineZhejiang UniversityYiwu322000China
- Department of SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhou310000China
- Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancerthe Fourth Affiliated Hospital, Zhejiang University School of MedicineYiwu322000China
| |
Collapse
|
3
|
Martin RC, Li Y, Shore EA, Malik DA, Li H, Hu X, Hayat T, Tan M, McMasters KM, Yan J. Irreversible Electroporation and Beta-Glucan-Induced Trained Innate Immunity for Treatment of Pancreatic Ductal Adenocarcinoma: A Phase II Study. J Am Coll Surg 2025; 240:351-361. [PMID: 39840846 PMCID: PMC11928255 DOI: 10.1097/xcs.0000000000001291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
BACKGROUND Irreversible electroporation (IRE) has augmented the effects of certain immunotherapies in pancreatic ductal adenocarcinoma (PDA). Yeast-derived particulate beta-glucan induces trained innate immunity and successfully reduced murine pancreatic cancer burden. This is a phase II study to test the hypothesis that IRE may augment beta-glucan-induced trained immunity in patients with PDA. STUDY DESIGN In this phase II clinical trial (NCT03080974), surgical ablative IRE was performed on clinical stage III PDA followed by oral beta-glucan administration for 12 months or until disease recurrence. Peripheral blood was taken preoperative, 14 days, and every 3 months and was evaluated by mass cytometry and compared with patients who received IRE alone. RESULTS Thirty consecutive patients with preoperative clinical stage III PDA were treated with IRE and then initiated on oral beta-glucan postoperatively were compared with 20 patients treated with IRE alone. There were no dose-limiting toxicities with oral beta-glucan, and compliance with therapy was 96% in all patients. Seven patients (23%) developed grade 3 or 4 treatment-related adverse events at 90 days; none required a dose modification of oral beta-glucan. A median disease-free interval (DFI) was 18 months (range 6 to 48 months), with a median overall survival (OS) of 32.5 months (range 4 to 53 months). At 12 months post-IRE, immunophenotyping was demonstrated a significant effect with improvement in the IRE-beta-glucan-treated group. This also resulted in a significant decrease on naive CD4 and CD8 T cells with increased CD4 and CD8 terminal effector cells in the IRE-beta-glucan-treated group, which correlated with a significant improvement in DFI and OS (p = 0.001). CONCLUSIONS Combined beta-glucan with IRE-ablated PDA tumor cells elicited a potent trained response and augmented antitumor functionality at 12 months post-IRE, which translated into an improved DFI and OS.
Collapse
Affiliation(s)
- Robert Cg Martin
- From the Divisions of Surgical Oncology (Martin, Y Li, Shore, Malik, Hayat, Tan, McMasters), The Hiram C Polk Jr, MD, Department of Surgery, University of Louisville School of Medicine, Louisville, KY
| | - Yan Li
- From the Divisions of Surgical Oncology (Martin, Y Li, Shore, Malik, Hayat, Tan, McMasters), The Hiram C Polk Jr, MD, Department of Surgery, University of Louisville School of Medicine, Louisville, KY
| | - Emily A Shore
- From the Divisions of Surgical Oncology (Martin, Y Li, Shore, Malik, Hayat, Tan, McMasters), The Hiram C Polk Jr, MD, Department of Surgery, University of Louisville School of Medicine, Louisville, KY
| | - Danial A Malik
- From the Divisions of Surgical Oncology (Martin, Y Li, Shore, Malik, Hayat, Tan, McMasters), The Hiram C Polk Jr, MD, Department of Surgery, University of Louisville School of Medicine, Louisville, KY
| | - Hong Li
- Functional Immunomics Core (H Li), Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY
| | - Xiaoling Hu
- Immunotherapy (Hu, Yan), The Hiram C Polk Jr, MD, Department of Surgery, University of Louisville School of Medicine, Louisville, KY
- Immuno-Oncology Program (Hu, Yan), Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY
| | - Traci Hayat
- From the Divisions of Surgical Oncology (Martin, Y Li, Shore, Malik, Hayat, Tan, McMasters), The Hiram C Polk Jr, MD, Department of Surgery, University of Louisville School of Medicine, Louisville, KY
| | - Min Tan
- From the Divisions of Surgical Oncology (Martin, Y Li, Shore, Malik, Hayat, Tan, McMasters), The Hiram C Polk Jr, MD, Department of Surgery, University of Louisville School of Medicine, Louisville, KY
| | - Kelly M McMasters
- From the Divisions of Surgical Oncology (Martin, Y Li, Shore, Malik, Hayat, Tan, McMasters), The Hiram C Polk Jr, MD, Department of Surgery, University of Louisville School of Medicine, Louisville, KY
| | - Jun Yan
- Immunotherapy (Hu, Yan), The Hiram C Polk Jr, MD, Department of Surgery, University of Louisville School of Medicine, Louisville, KY
- Immuno-Oncology Program (Hu, Yan), Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY (Yan)
| |
Collapse
|
4
|
Geboers B, Timmer F, Vos D, Scheffer H, Bakker J, Ruarus A, Vroomen L, Stam A, Lougheed S, Schouten E, Puijk R, van den Tol P, Lagerwaard F, de Vries J, Bruynzeel A, Meijerink M, de Gruijl T. Systemic immunomodulation by irreversible electroporation versus stereotactic ablative body radiotherapy in locally advanced pancreatic cancer: the CROSSFIRE trial. J Immunother Cancer 2025; 13:e010222. [PMID: 40139834 PMCID: PMC11950998 DOI: 10.1136/jitc-2024-010222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/28/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Irreversible electroporation (IRE) and stereotactic ablative body radiotherapy (SABR) are cytoreductive therapies for locally advanced pancreatic cancer (LAPC). Both may signify immunogenic cell death. We aimed to compare systemic immune responses between the treatments. METHODS As part of the randomized phase II CROSSFIRE trial (NCT02791503), comparing the oncological efficacy of IRE to SABR in patients with LAPC, pre- and post-treatment (2 weeks and 3 months) peripheral blood samples were collected. Frequency and activation status of lymphocytic and myeloid subsets were determined using flow cytometry. T cell responses to pancreatic cancer associated with Wilms tumor-1 (WT-1) and survivin tumor antigens were determined by interferon-γ enzyme-linked immunospot assay. RESULTS In total, 20 IRE and 20 SABR-treated participants were analyzed (20 men; median age 65 (IQR 55-70)). IRE induced immediate decreases in systemic regulatory T cell (Treg) and conventional type-1 dendritic cell rates, coinciding with CD4+/CD8+ T cell activation by upregulation of PD-1, which was associated with improved overall survival (OS). SABR similarly induced immediate CD4+/CD8+ T cell activation by upregulation of Ki67 and CD25 but resulted in asynchronously delayed Treg downregulation. SABR also induced a durable increase in CD4+ EM T cells, associated with improved OS. Ablation-induced WT-1 or survivin-specific T cell responses were observed in 9/16 (56%) immune competent participants (IRE n=5, SABR n=4) and were associated with longer OS. CONCLUSION Distinct immune stimulatory responses associated with improved OS, suggest that SABR might benefit from combined Treg depletion strategies while IRE could benefit from PD-1 checkpoint inhibition. TRIAL REGISTRATION NUMBER The trial was registered on clinical trials.gov (NCT02791503).
Collapse
Affiliation(s)
- Bart Geboers
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
- Cancer Centre Amsterdam, Amsterdam, The Netherlands
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Department of Medical Imaging, Prince of Wales Hospital, Sydney, New South Wales, Australia
| | - Florentine Timmer
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
- Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Danielle Vos
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
- Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Hester Scheffer
- Department of Radiology, Noord West Ziekenhuis Groep, Alkmaar, The Netherlands
| | - Joyce Bakker
- Cancer Centre Amsterdam, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
| | - Alette Ruarus
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
- Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Laurien Vroomen
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
- Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | | | | | - Evelien Schouten
- Department of Radiotherapy, Antoni van Leeuwenhoek Hospital - Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Robbert Puijk
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, OLVG, Amsterdam, The Netherlands
| | | | - Frank Lagerwaard
- Department of Radiation Oncology, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
| | - Jan de Vries
- Department of Radiology and Nuclear Medicine, OLVG, Amsterdam, The Netherlands
| | - Anna Bruynzeel
- Department of Radiation Oncology, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
| | - Martijn Meijerink
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
- Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Tanja de Gruijl
- Cancer Centre Amsterdam, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
| |
Collapse
|
5
|
Cereda V, D’Andrea MR. Pancreatic cancer: failures and hopes-a review of new promising treatment approaches. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002299. [PMID: 40124650 PMCID: PMC11926728 DOI: 10.37349/etat.2025.1002299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/22/2025] [Indexed: 03/25/2025] Open
Abstract
Pancreatic cancer is a challenging disease with limited treatment options and a high mortality rate. Just few therapy advances have been made in recent years. Tumor microenvironment, immunosuppressive features and mutational status represent important obstacles in the improvement of survival outcomes. Up to now, first-line therapy did achieve a median overall survival of less than 12 months and this discouraging data lead clinicians all over the world to focus their efforts on various fields of investigation: 1) sequential cycling of different systemic therapy in order to overcome mechanisms of resistance; 2) discovery of new predictive bio-markers, in order to target specific patient population; 3) combination treatment, in order to modulate the tumor microenvironment of pancreatic cancer; 4) new modalities of the delivery of drugs in order to pass the physical barrier of desmoplasia and tumor stroma. This review shows future directions of treatment strategies in advanced pancreatic cancer through a deep analysis of these recent macro areas of research.
Collapse
Affiliation(s)
- Vittore Cereda
- Asl Roma 4, Hospital S. Paolo Civitavecchia, 00053 Civitavecchia, Italy
| | | |
Collapse
|
6
|
Geboers B, Scheltema MJ, Jung J, Bakker J, Timmer FE, Cerutti X, Katelaris A, Doan P, Gondoputro W, Blazevski A, Agrawal S, Matthews J, Haynes A, Robertson T, Thompson JE, Meijerink MR, Clark SJ, de Gruijl TD, Stricker PD. Irreversible electroporation of localised prostate cancer downregulates immune suppression and induces systemic anti-tumour T-cell activation - IRE-IMMUNO study. BJU Int 2025; 135:319-328. [PMID: 39101639 PMCID: PMC11745989 DOI: 10.1111/bju.16496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/06/2024]
Abstract
OBJECTIVES To prospectively compare systemic anti-tumour immune responses induced by irreversible electroporation (IRE) and robot-assisted radical prostatectomy (RARP) in patients with localised intermediate-risk prostate cancer (PCa). PATIENTS AND METHODS Between February 2021 and June 2022, before and after treatment (at 5, 14 and 30 days) peripheral blood samples of 30 patients with localised PCa were prospectively collected. Patient inclusion criteria were: International Society of Urological Pathologists Grade 2-3, clinical cancer stage ≤T2c, prostate-specific antigen level <20 ng/mL). Patients were treated with IRE (n = 20) or RARP (n = 10). Frequency and activation status of lymphocytic and myeloid immune cell subsets were determined using flow cytometry. PCa-specific T-cell responses to prostatic acid phosphatase (PSAP) and cancer testis antigen (New York oesophageal squamous cell carcinoma 1 [NY-ESO-1]) were determined by interferon-γ enzyme-linked immunospot assay (ELISpot). Repeated-measures analysis of variance and two-sided Student's t-tests were used to compare immune responses over time and between treatment cohorts. RESULTS Patient and tumour characteristics were similar between the cohorts except for age (median 68 years [IRE] and 62 years [RARP], P = 0.01). IRE induced depletion of systemic regulatory T cells (P = 0.0001) and a simultaneous increase in activated cytotoxic T-lymphocyte antigen 4 (CTLA-4)+ cluster of differentiation (CD)4+ (P < 0.001) and CD8+ (P = 0.032) T cells, consistent with reduction of systemic immune suppression allowing for effector T-cell activation, peaking 14 days after IRE. Effects were positively correlated with tumour volume/ablation size. Accordingly, IRE induced expansion of PSAP and/or NY-ESO-1 specific T-cell responses in four of the eight immune competent patients. Temporarily increased activated myeloid derived suppressor cell frequencies (P = 0.047) were consistent with transient immunosuppression after RARP. CONCLUSIONS Irreversible electroporation induces a PCa-specific systemic immune response in patients with localised PCa, aiding conversion of the tumour microenvironment into a more immune permissive state. Therapeutic efficacy might be further enhanced by combination with CTLA-4 checkpoint inhibition, potentially opening up a new synergistic treatment paradigm for high-risk localised or (oligo)metastatic disease.
Collapse
|
7
|
Imran KM, Brock RM, Beitel-White N, Powar M, Orr K, Aycock KN, Alinezhadbalalami N, Salameh ZS, Eversole P, Tintera B, Markov Madanick J, Hendricks-Wenger A, Coutermarsh-Ott S, Davalos RV, Allen IC. Irreversible electroporation promotes a pro-inflammatory tumor microenvironment and anti-tumor immunity in a mouse pancreatic cancer model. Front Immunol 2024; 15:1352821. [PMID: 38711517 PMCID: PMC11070574 DOI: 10.3389/fimmu.2024.1352821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 04/09/2024] [Indexed: 05/08/2024] Open
Abstract
Pancreatic cancer is a significant cause of cancer-related mortality and often presents with limited treatment options. Pancreatic tumors are also notorious for their immunosuppressive microenvironment. Irreversible electroporation (IRE) is a non-thermal tumor ablation modality that employs high-voltage microsecond pulses to transiently permeabilize cell membranes, ultimately inducing cell death. However, the understanding of IRE's impact beyond the initiation of focal cell death in tumor tissue remains limited. In this study, we demonstrate that IRE triggers a unique mix of cell death pathways and orchestrates a shift in the local tumor microenvironment driven, in part, by reducing the myeloid-derived suppressor cell (MDSC) and regulatory T cell populations and increasing cytotoxic T lymphocytes and neutrophils. We further show that IRE drives induce cell cycle arrest at the G0/G1 phase in vitro and promote inflammatory cell death pathways consistent with pyroptosis and programmed necrosis in vivo. IRE-treated mice exhibited a substantial extension in progression-free survival. However, within a span of 14 days, the tumor immune cell populations reverted to their pre-treatment composition, which resulted in an attenuation of the systemic immune response targeting contralateral tumors and ultimately resulting in tumor regrowth. Mechanistically, we show that IRE augments IFN- γ signaling, resulting in the up-regulation of the PD-L1 checkpoint in pancreatic cancer cells. Together, these findings shed light on potential mechanisms of tumor regrowth following IRE treatment and offer insights into co-therapeutic targets to improve treatment strategies.
Collapse
Affiliation(s)
- Khan Mohammad Imran
- Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA, United States
| | - Rebecca M. Brock
- Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA, United States
| | - Natalie Beitel-White
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Manali Powar
- Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA, United States
| | - Katie Orr
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, United States
| | - Kenneth N. Aycock
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Nastaran Alinezhadbalalami
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Zaid S. Salameh
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Paige Eversole
- Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Benjamin Tintera
- Department of Surgery, Carilion Clinic and Virginia Tech Carilion School of Medicine, Roanoke, VA, United States
| | - Justin Markov Madanick
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, United States
| | - Alissa Hendricks-Wenger
- Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA, United States
| | - Sheryl Coutermarsh-Ott
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, United States
| | - Rafael V. Davalos
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Irving C. Allen
- Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA, United States
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, United States
- Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA, United States
| |
Collapse
|
8
|
Qiu L, Liu C, Li H. Successful immunotherapy with PD-1 Iinhibitor for advanced pancreatic cancer: report of two cases and review of literature. Anticancer Drugs 2024; 35:263-270. [PMID: 38194502 DOI: 10.1097/cad.0000000000001546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Pancreatic cancer is a highly malignant tumor, and most patients are diagnosed at an advanced stage. Unfortunately, due to the immunosuppressive tumor microenvironment of pancreatic cancer, the benefits of immunotherapy for patients with advanced pancreatic cancer are still unclear. Here, we present two cases of advanced pancreatic cancer being controlled by immunotherapy, with pathological diagnoses of ductal adenocarcinoma and acinar cell carcinoma, respectively. Next-generation sequencing (NGS) of both patients is high tumor mutation burden (tumor mutation burden-High) and microsatellite stable. The patient with pancreatic ductal adenocarcinoma was diagnosed as a locally advanced disease (stage III). She received irreversible electroporation, used the programmed death receptor-1 (PD-1) inhibitor (pembrolizumab) combined with chemotherapy (S-1), and then used only the PD-1 inhibitor as a maintenance treatment. As a result, the patient's lesion was significantly reduced, with a partial response time of up to 31 months. The patient with acinar cell carcinoma was diagnosed as a metastatic disease (stage IV), next-generation sequencing revealed mutations in SMAD4 and KMT2D, and two chemotherapy regimens were used unsuccessfully. Then, the combination of chemotherapy with PD-1 (tislelizumab) and vascular endothelial growth factor/vascular endothelial growth factor receptor (anlotinib) inhibitors were used, and the lesions of the patient were significantly reduced, and the progression-free survival after immunotherapy was 19 months. In advanced pancreatic cancer, a prognosis of this magnitude is rare. Our cases reveal the potential of immunotherapy as a cornerstone treatment in the management of advanced pancreatic cancer.
Collapse
Affiliation(s)
- Lijie Qiu
- Department of Oncology, Sun Yat-sen University First Affiliated Hospital
- Department of Radiology, Sun Yat-sen University Sixth Affiliated Hospital
| | - Chen Liu
- Department of Radiology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Heping Li
- Department of Oncology, Sun Yat-sen University First Affiliated Hospital
| |
Collapse
|
9
|
Dabiri R, Rashid MU, Khan OS, Jehanzeb S, Alomari M, Zafar H, Zahid E, Rahman AU, Karam A, Ahmad S. Immune modulators for pancreatic ductal adenocarcinoma therapy. IMMUNE LANDSCAPE OF PANCREATIC CANCER DEVELOPMENT AND DRUG RESISTANCE 2024:103-129. [DOI: 10.1016/b978-0-443-23523-8.00021-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
10
|
Timmer FEF, Geboers B, Scheffer HJ, Bakker J, Ruarus AH, Dijkstra M, van der Lei S, Boon R, Nieuwenhuizen S, van den Bemd BAT, Schouten EAC, van den Tol PM, Puijk RS, de Vries JJJ, de Gruijl TD, Meijerink MR. Tissue Resistance Decrease during Irreversible Electroporation of Pancreatic Cancer as a Biomarker for the Adaptive Immune Response and Survival. J Vasc Interv Radiol 2023; 34:1777-1784.e4. [PMID: 37391072 DOI: 10.1016/j.jvir.2023.06.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 06/07/2023] [Accepted: 06/20/2023] [Indexed: 07/02/2023] Open
Abstract
PURPOSE To correlate irreversible electroporation (IRE) procedural resistance changes with survival outcomes and the IRE-induced systemic immune response in patients with locally advanced pancreatic cancer (LAPC). MATERIALS AND METHODS Data on IRE procedural tissue resistance (R) features and survival outcomes were collected from patients with LAPC treated within the context of 2 prospective clinical trials in a single tertiary center. Preprocedural and postprocedural peripheral blood samples were prospectively collected for immune monitoring. The change (ie, decrease) in R during the first 10 test pulses (ΔR10p) and during the total procedure (ΔRtotal) were calculated. Patients were divided in 2 groups on the basis of the median change in R (large ΔR vs small ΔR) and compared for differences in overall survival (OS) and progression-free survival and immune cell subsets. RESULTS A total of 54 patients were included; of these, 20 underwent immune monitoring. Linear regression modeling showed that the first 10 test pulses reflected the change in tissue resistance during the total procedure appropriately (P < .001; R2 = 0.91). A large change in tissue resistance significantly correlated with a better OS (P = .026) and longer time to disease progression (P = .045). Furthermore, a large change in tissue resistance was associated with CD8+ T cell activation through significant upregulation of Ki-67+ (P = .02) and PD-1+ (P = .047). Additionally, this subgroup demonstrated significantly increased expression of CD80 on conventional dendritic cells (cDC1; P = .027) and PD-L1 on immunosuppressive myeloid-derived suppressor cells (P = .039). CONCLUSIONS IRE procedural resistance changes may serve as a biomarker for survival and IRE-induced systemic CD8+ T cell and cDC1 activation.
Collapse
Affiliation(s)
- Florentine E F Timmer
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers (UMC), location Vrije Universiteit, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Bart Geboers
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers (UMC), location Vrije Universiteit, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands.
| | - Hester J Scheffer
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers (UMC), location Vrije Universiteit, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Joyce Bakker
- Department of Medical Oncology, Amsterdam UMC, location Vrije Universiteit, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Alette H Ruarus
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers (UMC), location Vrije Universiteit, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Madelon Dijkstra
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers (UMC), location Vrije Universiteit, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Susan van der Lei
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers (UMC), location Vrije Universiteit, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Rianne Boon
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers (UMC), location Vrije Universiteit, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Sanne Nieuwenhuizen
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers (UMC), location Vrije Universiteit, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Bente A T van den Bemd
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers (UMC), location Vrije Universiteit, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Evelien A C Schouten
- Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | | | - Robbert S Puijk
- Cancer Center Amsterdam, Amsterdam, the Netherlands; Department of Radiology and Nuclear Medicine, Onze Lieve Vrouwen Gasthuis, Amsterdam, the Netherlands
| | - Jan J J de Vries
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers (UMC), location Vrije Universiteit, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, Amsterdam UMC, location Vrije Universiteit, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Martijn R Meijerink
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers (UMC), location Vrije Universiteit, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
11
|
Ma Y, Xing Y, Li H, Yuan T, Liang B, Li R, Li J, Li Z, Li S, Niu L. Irreversible electroporation combined with chemotherapy and PD-1/PD-L1 blockade enhanced antitumor immunity for locally advanced pancreatic cancer. Front Immunol 2023; 14:1193040. [PMID: 37691923 PMCID: PMC10485610 DOI: 10.3389/fimmu.2023.1193040] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 08/09/2023] [Indexed: 09/12/2023] Open
Abstract
Background Irreversible electroporation (IRE) is a novel local tumor ablation approach with the potential to stimulate an antitumor immune response. However, it is not effective in preventing distant metastasis in isolation. This study aimed to compare the potential of augmenting the antitumor immune response in patients with locally advanced pancreatic cancer (LAPC) who underwent IRE combined with chemotherapy and PD-1/PD-L1 blockade with those who underwent IRE combined with chemotherapy. Methods A retrospective review was conducted on LAPC patients treated either with IRE in combination with chemotherapy and PD-1/PD-L1 blockade (group A) or with IRE with chemotherapy alone (group B) from July 2015 to June 2021. The primary outcomes were overall survival (OS) and progression-free survival (PFS), with immune responses and adverse events serving as secondary endpoints. Risk factors for OS and PFS were identified using univariate and multivariate analyses. Results A total of 103 patients were included in the final analysis, comprising 25 in group A and 78 in group B. The median duration of follow-up was 18.2 months (3.0-38.6 months). Group A patients demonstrated improved survival compared to group B (median OS: 23.6 vs. 19.4 months, p = 0.001; median PFS: 18.2 vs. 14.7 months, p = 0.022). The data suggest a robust immune response in group A, while adverse events related to the treatment were similar in both groups. The multivariate analysis identified the combination of IRE, chemotherapy, and PD-1/PD-L1 blockade as an independent prognostic factor for OS and PFS. Conclusion The addition of PD-1/PD-L1 blockade to the regimen of IRE combined with chemotherapy enhanced antitumor immunity and extended survival in LAPC patients.
Collapse
Affiliation(s)
- Yangyang Ma
- Central Laboratory, Affiliated Fuda Cancer Hospital, Jinan University, Guangzhou, China
| | - Yanli Xing
- Department of Oncology, Affiliated Fuda Cancer Hospital, Jinan University, Guangzhou, China
| | - Hongmei Li
- Department of Oncology, Affiliated Fuda Cancer Hospital, Jinan University, Guangzhou, China
| | - Ting Yuan
- Department of Oncology, Affiliated Fuda Cancer Hospital, Jinan University, Guangzhou, China
| | - Bing Liang
- Department of Surgery and Anesthesia, Affiliated Fuda Cancer Hospital, Jinan University, Guangzhou, China
| | - Rongrong Li
- Department of Ultrasound, Affiliated Fuda Cancer Hospital, Jinan University, Guangzhou, China
| | - Jianyu Li
- Department of Surgery and Anesthesia, Affiliated Fuda Cancer Hospital, Jinan University, Guangzhou, China
| | - Zhonghai Li
- Department of Radiology, Affiliated Fuda Cancer Hospital, Jinan University, Guangzhou, China
| | - Shuying Li
- Department of Surgery and Anesthesia, Affiliated Fuda Cancer Hospital, Jinan University, Guangzhou, China
| | - Lizhi Niu
- Department of Oncology, Affiliated Fuda Cancer Hospital, Jinan University, Guangzhou, China
| |
Collapse
|
12
|
Campana LG, Daud A, Lancellotti F, Arroyo JP, Davalos RV, Di Prata C, Gehl J. Pulsed Electric Fields in Oncology: A Snapshot of Current Clinical Practices and Research Directions from the 4th World Congress of Electroporation. Cancers (Basel) 2023; 15:3340. [PMID: 37444450 PMCID: PMC10340685 DOI: 10.3390/cancers15133340] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/29/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
The 4th World Congress of Electroporation (Copenhagen, 9-13 October 2022) provided a unique opportunity to convene leading experts in pulsed electric fields (PEF). PEF-based therapies harness electric fields to produce therapeutically useful effects on cancers and represent a valuable option for a variety of patients. As such, irreversible electroporation (IRE), gene electrotransfer (GET), electrochemotherapy (ECT), calcium electroporation (Ca-EP), and tumour-treating fields (TTF) are on the rise. Still, their full therapeutic potential remains underappreciated, and the field faces fragmentation, as shown by parallel maturation and differences in the stages of development and regulatory approval worldwide. This narrative review provides a glimpse of PEF-based techniques, including key mechanisms, clinical indications, and advances in therapy; finally, it offers insights into current research directions. By highlighting a common ground, the authors aim to break silos, strengthen cross-functional collaboration, and pave the way to novel possibilities for intervention. Intriguingly, beyond their peculiar mechanism of action, PEF-based therapies share technical interconnections and multifaceted biological effects (e.g., vascular, immunological) worth exploiting in combinatorial strategies.
Collapse
Affiliation(s)
- Luca G. Campana
- Department of Surgery, Manchester University NHS Foundation Trust, Oxford Rd., Manchester M13 9WL, UK;
| | - Adil Daud
- Department of Medicine, University of California, 550 16 Street, San Francisco, CA 94158, USA;
| | - Francesco Lancellotti
- Department of Surgery, Manchester University NHS Foundation Trust, Oxford Rd., Manchester M13 9WL, UK;
| | - Julio P. Arroyo
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA; (J.P.A.); (R.V.D.)
| | - Rafael V. Davalos
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA; (J.P.A.); (R.V.D.)
- Institute for Critical Technology and Applied Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Claudia Di Prata
- Department of Surgery, San Martino Hospital, 32100 Belluno, Italy;
| | - Julie Gehl
- Department of Clinical Oncology and Palliative Care, Zealand University Hospital, 4000 Roskilde, Denmark;
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 1165 Copenhagen, Denmark
| |
Collapse
|
13
|
Shen J, Pan P, Hu X, Zhao J, Wu H. Safety and Efficacy of Irreversible Electroporation in Locally Advanced Pancreatic Cancer: An Evaluation from a Surgeon's Perspective. Cancers (Basel) 2022; 14:cancers14225677. [PMID: 36428767 PMCID: PMC9688427 DOI: 10.3390/cancers14225677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022] Open
Abstract
Irreversible electroporation (IRE) has emerged as a promising treatment for patients with locally advanced pancreatic cancer (LAPC). Therefore, in this study, we evaluate the safety and efficacy of IRE against LAPC, as well as exploring its impact on anti-tumor immunity. A retrospective analysis was conducted in consecutive patients at a single institution. Eligible patients were assigned to IRE, palliative surgery (PS), or vascular resection (VR) groups, according to their respective treatments. The IRE group consisted of LAPC patients. One-to-one propensity score matching was performed, in order to compare the incidence of complications and median overall survival (mOS). Serum and intratumoral cytokines, as well as intratumoral immune cells, were analyzed in order to identify changes in immunity after IRE. A total of 210 patients were included. After matching, the rate of major complications (Clavien−Dindo III−V), intra-abdominal hemorrhage, and re-intervention in the IRE group were similar to those in the VR group (p > 0.05). The mOS of the IRE group (13.0 months) was shorter than that of the VR group (15.0 months), but longer than that of the PS group (8.0 months) (p < 0.05). Patients in the IRE group had elevated serum levels of immunogenic cytokines, including IL-2, IL-6, and TNF-α, which were related to anti-tumor immunity. The survival advantage in IRE-treated patients was attributed to tumor ablation and immune modulation effects. Overall, IRE can be considered a feasible treatment for patients with LAPC.
Collapse
Affiliation(s)
- Jian Shen
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Penglin Pan
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaoli Hu
- Department of Radiology, Wuhan Asia Heart Hospital, Wuhan 430022, China
| | - Jun Zhao
- Department of Anatomy, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan 430030, China
- Correspondence: (J.Z.); (H.W.); Tel.: +86-027-8525-1631 (H.W.)
| | - Heshui Wu
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Correspondence: (J.Z.); (H.W.); Tel.: +86-027-8525-1631 (H.W.)
| |
Collapse
|
14
|
Image-guided in situ cancer vaccination with combination of multi-functional nano-adjuvant and an irreversible electroporation technique. Biomaterials 2022; 289:121762. [DOI: 10.1016/j.biomaterials.2022.121762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 08/16/2022] [Accepted: 08/21/2022] [Indexed: 11/19/2022]
|
15
|
Thomas AS, Kwon W, Horowitz DP, Bates SE, Fojo AT, Manji GA, Schreibman S, Schrope BA, Chabot JA, Kluger MD. Long-term follow-up experience with adjuvant therapy after irreversible electroporation of locally advanced pancreatic cancer. J Surg Oncol 2022; 126:1442-1450. [PMID: 36048146 DOI: 10.1002/jso.27085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/09/2022] [Accepted: 08/24/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Irreversible electroporation (IRE) expands the surgical options for patients with unresectable pancreatic cancer. This study evaluated for differences in survival stratified by type of IRE and receipt of adjuvant chemotherapy. METHODS Patients with locally advanced pancreatic cancer treated by IRE (2012-2020) were retrospectively included. Overall survival (OS) and recurrence-free survival (RFS) were compared by type of IRE (in situ for local tumor control or IRE of potentially positive margins with resection) and by receipt of adjuvant chemotherapy. RESULTS Thirty-nine patients had IRE in situ, 61 had IRE for margin extension, and 19 received adjuvant chemotherapy. Most (97.00%) underwent induction chemotherapy. OS was 28.71 months (interquartile range [IQR] 19.17, 51.19) from diagnosis, with no difference by IRE type (hazard ratio [HR] 1.05 for margin extension [p = 0.85]) or adjuvant chemotherapy (HR 1.14 [p = 0.639]). RFS was 8.51 months (IQR 4.95, 20.17) with no difference by IRE type (HR 0.90 for margin extension [p = 0.694]) or adjuvant chemotherapy (HR 0.90 [p = 0.711]). CONCLUSION These findings suggest that adjuvant therapy may have limited benefit for patients treated with induction chemotherapy followed by local control with IRE for unresectable pancreatic cancer. Further study of the duration and timing of systemic therapy is warranted to maximize benefit and limit toxicity.
Collapse
Affiliation(s)
- Alexander S Thomas
- Department of Surgery, Division of Gastrointestinal and Endocrine Surgery, Columbia University Irving Medical Center, New York, New York, USA
| | - Wooil Kwon
- Department of Surgery, Division of Gastrointestinal and Endocrine Surgery, Columbia University Irving Medical Center, New York, New York, USA.,Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - David P Horowitz
- Department of Radiation Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical New York, New York, New York, USA
| | - Susan E Bates
- Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, New York, USA
| | - Antonio T Fojo
- Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, New York, USA
| | - Gulam A Manji
- Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, New York, USA
| | - Stephen Schreibman
- Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, New York, USA
| | - Beth A Schrope
- Department of Surgery, Division of Gastrointestinal and Endocrine Surgery, Columbia University Irving Medical Center, New York, New York, USA
| | - John A Chabot
- Department of Surgery, Division of Gastrointestinal and Endocrine Surgery, Columbia University Irving Medical Center, New York, New York, USA
| | - Michael D Kluger
- Department of Surgery, Division of Gastrointestinal and Endocrine Surgery, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
16
|
Vidal-Jove J, Serres X, Vlaisavljevich E, Cannata J, Duryea A, Miller R, Merino X, Velat M, Kam Y, Bolduan R, Amaral J, Hall T, Xu Z, Lee FT, Ziemlewicz TJ. First-in-man histotripsy of hepatic tumors: the THERESA trial, a feasibility study. Int J Hyperthermia 2022; 39:1115-1123. [PMID: 36002243 DOI: 10.1080/02656736.2022.2112309] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022] Open
Abstract
Rationale Current hepatic locoregional therapies are limited in terms of effectiveness and toxicities. Given promising pre-clinical results, a first in-human trial was designed to assess the technical effectiveness and safety profile of histotripsy, a noninvasive, non-thermal, non-ionizing focused ultrasound therapy that creates precise, predictable tissue destruction, in patients with primary and secondary liver tumors.Methods A multicenter phase I trial (Theresa Study) was performed in a single country with 8 weeks of planned follow-up. Eight of fourteen recruited patients were deemed eligible and enrolled in the study. Hepatic histotripsy, was performed with a prototype system (HistoSonics, Inc., Ann Arbor, MI). Eleven tumors were targeted in the 8 patients who all had unresectable end-stage multifocal liver tumors: colorectal liver metastases (CRLM) in 5 patients (7 tumors), breast cancer metastases in 1 (1 tumor), cholangiocarcinoma metastases in 1 (2 tumors), and hepatocellular carcinoma (HCC) in 1 (1 tumor). The primary endpoint was acute technical success, defined as creating a zone of tissue destruction per planned volume assessed by MRI 1-day post-procedure. Safety (device-related adverse events) through 2 months was a secondary endpoint.Results The 8 patients had a median age of 60.4 years with an average targeted tumor diameter of 1.4 cm. The primary endpoint was achieved in all procedures. The secondary safety profile endpoint identified no device-related adverse events. Two patients experienced a continuous decline in tumor markers during the eight weeks following the procedure.Conclusions This first-in-human trial demonstrates that hepatic histotripsy effectively destroys liver tissue in a predictable manner, correlating very well with the planned histotripsy volume, and has a high safety profile without any device-related adverse events. Based on these results, the need for more definitive clinical trials is warranted. Trial Registration: Study to Evaluate VORTX Rx (Theresa). NCT03741088. https://clinicaltrials.gov/ct2/show/NCT03741088 KEY POINTSHistotripsy, a new noninvasive, non-thermal, non-ionizing focused ultrasound therapy, safely created a zone of tissue destruction in the liver that correlated very well with the pre-defined planned tissue destruction volume.In this first human trial histotripsy was well tolerated with no histotripsy device-related adverse events and its primary endpoint of acute technical success was achieved in all 8 enrolled patients with primary or secondary liver tumors.This new locoregional therapy for patients with liver tumors is safe and effective, warranting further trials.
Collapse
Affiliation(s)
- Joan Vidal-Jove
- Institute Khuab for Interventional Oncology, Comprehensive Tumor Center, Barcelona, Spain
| | - Xavier Serres
- Department of Interventional Ultrasound, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Eli Vlaisavljevich
- Department of Biomedical Engineering, Virginia Polytechnic University, Blacksburg, Virginia
| | - Jon Cannata
- Research and Development, Histosonics, Inc, Ann Arbor, MI, USA
| | - Alex Duryea
- Research and Development, Histosonics, Inc, Ann Arbor, MI, USA
| | - Ryan Miller
- Research and Development, Histosonics, Inc, Ann Arbor, MI, USA
| | - Xavier Merino
- Department of Interventional Ultrasound, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Manuela Velat
- Institute Khuab for Interventional Oncology, Comprehensive Tumor Center, Barcelona, Spain
| | - Yossi Kam
- R&D Clinical Excellence, Philips Healthcare, Haifa, Israel
| | - Ryan Bolduan
- Clinical Research and Medical Affairs, Histosonics, Inc, Ann Arbor, MI, USA
| | - Joseph Amaral
- Clinical Research and Medical Affairs, Histosonics, Inc, Ann Arbor, MI, USA
| | - Timothy Hall
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Zhen Xu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Fred T Lee
- Department of Radiology, University of Wisconsin, Madison, WI, USA
| | | |
Collapse
|
17
|
Wang Y, Jiang T, Xie L, Wang H, Zhao J, Xu L, Fang C. Effect of pulsed field ablation on solid tumor cells and microenvironment. Front Oncol 2022; 12:899722. [PMID: 36081554 PMCID: PMC9447365 DOI: 10.3389/fonc.2022.899722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 07/22/2022] [Indexed: 11/13/2022] Open
Abstract
Pulsed field ablation can increase membrane permeability and is an emerging non-thermal ablation. While ablating tumor tissues, electrical pulses not only act on the membrane structure of cells to cause irreversible electroporation, but also convert tumors into an immune active state, increase the permeability of microvessels, inhibit the proliferation of pathological blood vessels, and soften the extracellular matrix thereby inhibiting infiltrative tumor growth. Electrical pulses can alter the tumor microenvironment, making the inhibitory effect on the tumor not limited to short-term killing, but mobilizing the collective immune system to inhibit tumor growth and invasion together.
Collapse
Affiliation(s)
- Yujue Wang
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tian’an Jiang
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Pulsed Power Translational Medicine of Zhejiang Province, Hangzhou, China
- Zhejiang University Cancer Center, Hangzhou, China
- *Correspondence: Tian’an Jiang,
| | - Liting Xie
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Pulsed Power Translational Medicine of Zhejiang Province, Hangzhou, China
| | - Huiyang Wang
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Pulsed Power Translational Medicine of Zhejiang Province, Hangzhou, China
| | - Jing Zhao
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lei Xu
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chengyu Fang
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
18
|
Justesen TF, Orhan A, Raskov H, Nolsoe C, Gögenur I. Electroporation and Immunotherapy-Unleashing the Abscopal Effect. Cancers (Basel) 2022; 14:cancers14122876. [PMID: 35740542 PMCID: PMC9221311 DOI: 10.3390/cancers14122876] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/09/2022] [Accepted: 06/09/2022] [Indexed: 01/10/2023] Open
Abstract
Simple Summary Electrochemotherapy and irreversible electroporation are primarily used for treating patients with cutaneous and subcutaneous tumors and pancreatic cancer, respectively. Increasing numbers of studies have shown that the treatments may elicit an immune response in addition to eliminating the tumor cells. The purpose of this review is to give an in-depth introduction to the electroporation-induced immune response and the local and peripheral immune systems, and to describe the various studies investigating the combination of electroporation and immunotherapy. The review may help guide and inspire the design of future clinical trials investigating the potential synergy of electroporation and immunotherapy in cancer treatment. Abstract The discovery of electroporation in 1968 has led to the development of electrochemotherapy (ECT) and irreversible electroporation (IRE). ECT and IRE have been established as treatments of cutaneous and subcutaneous tumors and locally advanced pancreatic cancer, respectively. Interestingly, the treatment modalities have been shown to elicit immunogenic cell death, which in turn can induce an immune response towards the tumor cells. With the dawn of the immunotherapy era, the potential of combining ECT and IRE with immunotherapy has led to the launch of numerous studies. Data from the first clinical trials are promising, and new combination regimes might change the way we treat tumors characterized by low immunogenicity and high levels of immunosuppression, such as melanoma and pancreatic cancer. In this review we will give an introduction to ECT and IRE and discuss the impact on the immune system. Additionally, we will present the results of clinical and preclinical trials, investigating the combination of electroporation modalities and immunotherapy.
Collapse
Affiliation(s)
- Tobias Freyberg Justesen
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark; (A.O.); (H.R.); (I.G.)
- Correspondence:
| | - Adile Orhan
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark; (A.O.); (H.R.); (I.G.)
| | - Hans Raskov
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark; (A.O.); (H.R.); (I.G.)
| | - Christian Nolsoe
- Center for Surgical Ultrasound, Department of Surgery, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark;
- Copenhagen Academy for Medical Education and Simulation (CAMES), University of Copenhagen and the Capital Region of Denmark, Ryesgade 53B, 2100 Copenhagen, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark; (A.O.); (H.R.); (I.G.)
- Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| |
Collapse
|
19
|
Rai ZL, Ranieri V, Palmer DH, Littler P, Ghaneh P, Gurusamy K, Manas D, Pizzo E, Psarelli EE, Gilmore R, Peddu P, Bartlett DC, de Liguori Carino N, Davidson BR. Treatment of unresectable locally advanced pancreatic cancer with percutaneous irreversible electroporation (IRE) following initial systemic chemotherapy (LAP-PIE) trial: study protocol for a feasibility randomised controlled trial. BMJ Open 2022; 12:e050166. [PMID: 35551086 PMCID: PMC9109032 DOI: 10.1136/bmjopen-2021-050166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 01/17/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Approximately 30% of patients with pancreas cancer have unresectable locally advanced disease, which is currently treated with systemic chemotherapy. A new treatment option of irreversible electroporation (IRE) has been investigated for these patients since 2005. Cohort studies suggest that IRE confers a survival advantage, but with associated, procedure-related complications. Selection bias may account for improved survival and there have been no prospective randomised trials evaluating the harms and benefits of therapy. The aim of this trial is to evaluate the feasibility of a randomised comparison of IRE therapy with chemotherapy versus chemotherapy alone in patients with locally advanced pancreatic cancer (LAPC). METHODS AND ANALYSIS Eligible patients with LAPC who have undergone first-line 5-FluoroUracil, Leucovorin, Irinotecan and Oxaliplatin chemotherapy will be randomised to receive either a single session of IRE followed by (if indicated) further chemotherapy or to chemotherapy alone (standard of care). Fifty patients from up to seven specialist pancreas centres in the UK will be recruited over a period of 15 months. Trial follow-up will be 12 months. The primary outcome measure is ability to recruit. Secondary objectives include practicality and technical success of treatment, acceptability of treatment to patients and clinicians and safety of treatment. A qualitative study has been incorporated to evaluate the patient and clinician perspective of the locally advanced pancreatic cancer with percutaneous irreversible electroporation trial. It is likely that the data obtained will guide the structure, the primary outcome measure, the power and the duration of a subsequent multicentre randomised controlled trial aimed at establishing the clinical efficiency of pancreas IRE therapy. Indicative procedure-related costings will be collected in this feasibility trial, which will inform the cost evaluation in the subsequent study on efficiency. ETHICS AND DISSEMINATION The protocol has received approval by London-Brent Research Ethics Committee reference number 21/LO/0077.Results will be analysed following completion of trial recruitment and follow-up. Results will be presented to international conferences with an interest in oncology, hepatopancreaticobiliary surgery and interventional radiology and be published in a peer-reviewed journal. TRIAL REGISTRATION NUMBER ISRCTN14986389.
Collapse
Affiliation(s)
- Zainab L Rai
- Division of Surgery and Interventional Science, University College London, London, UK
- Wellcome/EPSRC Centre for Interventional and Surgical Sciences (WEISS), London, UK
| | - Veronica Ranieri
- Research & Development, Tavistock and Portman NHS Foundation Trust, London, UK
- Science & Technology Studies, University College London, London, UK
| | - Daniel H Palmer
- Hepato-Pancreatco-Biliary Disease, University of Liverpool, Liverpool, Merseyside, UK
| | - Peter Littler
- Hepato-Pancreatico-Biliary Surgery, Freeman Hospital, Newcastle Upon Tyne, UK
| | - Pauleh Ghaneh
- Hepato-Pancreatco-Biliary Disease, University of Liverpool, Liverpool, Merseyside, UK
| | - Kurinchi Gurusamy
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Derek Manas
- Hepato-Pancreatico-Biliary Surgery, Newcastle Upon Tyne Hospitals NHS Trust, Newcastle Upon Tyne, UK
| | - Elena Pizzo
- Applied Health Research, University College London, London, UK
| | | | - Roopinder Gilmore
- Division of Surgery and Interventional Science, University College London, London, UK
- Oncology, Royal Free London NHS Foundation Trust, London, UK
| | - Praveen Peddu
- Department of Radiology, King's College Hospital NHS Trust, London, UK
| | - David C Bartlett
- Hepato-Pancreatico-Biliary Surgery, University Hospitals Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK
| | | | | |
Collapse
|
20
|
Murphy KR, Aycock KN, Hay AN, Rossmeisl JH, Davalos RV, Dervisis NG. High-frequency irreversible electroporation brain tumor ablation: exploring the dynamics of cell death and recovery. Bioelectrochemistry 2022; 144:108001. [PMID: 34844040 PMCID: PMC8792323 DOI: 10.1016/j.bioelechem.2021.108001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/12/2021] [Accepted: 11/14/2021] [Indexed: 11/02/2022]
Abstract
Improved therapeutics for malignant brain tumors are urgently needed. High-frequency irreversible electroporation (H-FIRE) is a minimally invasive, nonthermal tissue ablation technique, which utilizes high-frequency, bipolar electric pulses to precisely kill tumor cells. The mechanisms of H-FIRE-induced tumor cell death and potential for cellular recovery are incompletely characterized. We hypothesized that tumor cells treated with specific H-FIRE electric field doses can survive and retain proliferative capacity. F98 glioma and LL/2 Lewis lung carcinoma cell suspensions were treated with H-FIRE to model primary and metastatic brain cancer, respectively. Cell membrane permeability, apoptosis, metabolic viability, and proliferative capacity were temporally measured using exclusion dyes, condensed chromatin staining, WST-8 fluorescence, and clonogenic assays, respectively. Both tumor cell lines exhibited dose-dependent permeabilization, with 1,500 V/cm permitting and 3,000 V/cm inhibiting membrane recovery 24 h post-treatment. Cells treated with 1,500 V/cm demonstrated significant and progressive recovery of apoptosis and metabolic activity, in contrast to cells treated with higher H-FIRE doses. Cancer cells treated with recovery-permitting doses of H-FIRE maintained while those treated with recovery-inhibiting doses lost proliferative capacity. Taken together, our data suggest that H-FIRE induces reversible and irreversible cellular damage in a dose-dependent manner, and the presence of dose-dependent recovery mechanisms permits tumor cell proliferation.
Collapse
Affiliation(s)
- Kelsey R Murphy
- Department of Biomedical and Veterinary Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, United States.
| | - Kenneth N Aycock
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, United States.
| | - Alayna N Hay
- Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, United States.
| | - John H Rossmeisl
- Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, United States.
| | - Rafael V Davalos
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, United States; ICTAS Center for Engineered Health, Virginia Tech, Kelly Hall, Blacksburg, VA 24061, United States.
| | - Nikolaos G Dervisis
- Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, United States; Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, United States.
| |
Collapse
|
21
|
Garnier J, Turrini O, Chretien AS, Olive D. Local Ablative Therapy Associated with Immunotherapy in Locally Advanced Pancreatic Cancer: A Solution to Overcome the Double Trouble?-A Comprehensive Review. J Clin Med 2022; 11:1948. [PMID: 35407555 PMCID: PMC8999652 DOI: 10.3390/jcm11071948] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/15/2022] [Accepted: 03/24/2022] [Indexed: 02/04/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a major killer and is a challenging clinical research issue with abysmal survival due to unsatisfactory therapeutic efficacy. Two major issues thwart the treatment of locally advanced nonresectable pancreatic cancer (LAPC): high micrometastasis rate and surgical inaccessibility. Local ablative therapies induce a systemic antitumor response (i.e., abscopal effect) in addition to local effects. Thus, the incorporation of additional therapies could be key to improving immunotherapy's clinical efficacy. In this systematic review, we explore recent applications of local ablative therapies combined with immunotherapy to overcome immune resistance in PDAC and discuss future perspectives and challenges. Particularly, we describe four chemoradiation studies and nine reports on irreversible electroporation (IRE). Clinically, IRE is the ablative therapy of choice, utilized in all but two clinical trials, and may create a favorable microenvironment for immunotherapy. Various immunotherapies have been used in combination with IRE, such as NK cell- or γδ T cell-based therapy, as well as immune checkpoint inhibitors. The results of the clinical trials presented in this review and the advancement potential of these therapies to phase II/III trials remain unknown. A multiple treatment approach involving chemotherapy, local ablation, and immunotherapy holds promise in overcoming the double trouble of LAPC.
Collapse
Affiliation(s)
- Jonathan Garnier
- Departement of Surgical Oncology, Institut Paoli-Calmettes, 232 Boulevard de Sainte Marguerite, 13009 Marseille, France
| | - Olivier Turrini
- Departement of Surgical Oncology, Institut Paoli-Calmettes, 232 Boulevard de Sainte Marguerite, 13009 Marseille, France
- Centre de Recherche en Cancérologie de Marseille, Aix-Marseille University, 58 Boulevard Charles Livon, 13007 Marseille, France
| | - Anne-Sophie Chretien
- Centre de Recherche en Cancérologie de Marseille, Aix-Marseille University, 58 Boulevard Charles Livon, 13007 Marseille, France
- Team Immunity and Cancer, U1068 Inserm, UMR7258 Centre National de la Recherche Scientifique, 13009 Marseille, France
- Departement of Immunomonitoring, Institut Paoli-Calmettes, 232 Boulevard de Sainte Marguerite, 13009 Marseille, France
| | - Daniel Olive
- Centre de Recherche en Cancérologie de Marseille, Aix-Marseille University, 58 Boulevard Charles Livon, 13007 Marseille, France
- Team Immunity and Cancer, U1068 Inserm, UMR7258 Centre National de la Recherche Scientifique, 13009 Marseille, France
- Departement of Immunomonitoring, Institut Paoli-Calmettes, 232 Boulevard de Sainte Marguerite, 13009 Marseille, France
| |
Collapse
|
22
|
Senders ZJ, Martin RCG. Intratumoral Immunotherapy and Tumor Ablation: A Local Approach with Broad Potential. Cancers (Basel) 2022; 14:cancers14071754. [PMID: 35406525 PMCID: PMC8996835 DOI: 10.3390/cancers14071754] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/11/2022] [Accepted: 03/17/2022] [Indexed: 12/22/2022] Open
Abstract
Several intratumoral immunotherapeutic agents have shown efficacy in controlling local disease; however, their ability to induce a durable systemic immune response is limited. Likewise, tumor ablation is well-established due to its role in local disease control but generally produces only a modest immunogenic effect. It has recently been recognized, however, that there is potential synergy between these two modalities and their distinct mechanisms of immune modulation. The aim of this review is to evaluate the existing data regarding multimodality therapy with intratumoral immunotherapy and tumor ablation. We discuss the rationale for this therapeutic approach, highlight novel combinations, and address the challenges to their clinical utility. There is substantial evidence that combination therapy with intratumoral immunotherapy and tumor ablation can potentiate durable systemic immune responses and should be further evaluated in the clinical setting.
Collapse
|
23
|
Zhang N, Li Z, Han X, Zhu Z, Li Z, Zhao Y, Liu Z, Lv Y. Irreversible Electroporation: An Emerging Immunomodulatory Therapy on Solid Tumors. Front Immunol 2022; 12:811726. [PMID: 35069599 PMCID: PMC8777104 DOI: 10.3389/fimmu.2021.811726] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/13/2021] [Indexed: 01/10/2023] Open
Abstract
Irreversible electroporation (IRE), a novel non-thermal ablation technique, is utilized to ablate unresectable solid tumors and demonstrates favorable safety and efficacy in the clinic. IRE applies electric pulses to alter the cell transmembrane voltage and causes nanometer-sized membrane defects or pores in the cells, which leads to loss of cell homeostasis and ultimately results in cell death. The major drawbacks of IRE are incomplete ablation and susceptibility to recurrence, which limit its clinical application. Recent studies have shown that IRE promotes the massive release of intracellular concealed tumor antigens that become an "in-situ tumor vaccine," inducing a potential antitumor immune response to kill residual tumor cells after ablation and inhibiting local recurrence and distant metastasis. Therefore, IRE can be regarded as a potential immunomodulatory therapy, and combined with immunotherapy, it can exhibit synergistic treatment effects on malignant tumors, which provides broad application prospects for tumor treatment. This work reviewed the current status of the clinical efficacy of IRE in tumor treatment, summarized the characteristics of local and systemic immune responses induced by IRE in tumor-bearing organisms, and analyzed the specific mechanisms of the IRE-induced immune response. Moreover, we reviewed the current research progress of IRE combined with immunotherapy in the treatment of solid tumors. Based on the findings, we present deficiencies of current preclinical studies of animal models and analyze possible reasons and solutions. We also propose possible demands for clinical research. This review aimed to provide theoretical and practical guidance for the combination of IRE with immunotherapy in the treatment of malignant tumors.
Collapse
Affiliation(s)
- Nana Zhang
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Zhuoqun Li
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xuan Han
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ziyu Zhu
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Zhujun Li
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yan Zhao
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Zhijun Liu
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yi Lv
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
24
|
Mills BN, Qiu H, Drage MG, Chen C, Mathew JS, Garrett-Larsen J, Ye J, Uccello TP, Murphy JD, Belt BA, Lord EM, Katz AW, Linehan DC, Gerber SA. Modulation of the Human Pancreatic Ductal Adenocarcinoma Immune Microenvironment by Stereotactic Body Radiotherapy. Clin Cancer Res 2021; 28:150-162. [PMID: 34862242 DOI: 10.1158/1078-0432.ccr-21-2495] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/23/2021] [Accepted: 10/13/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Stereotactic body radiotherapy (SBRT) is an emerging treatment modality for pancreatic ductal adenocarcinoma (PDAC), which can effectively prime cytotoxic T cells by inducing immunogenic tumor cell death in preclinical models. SBRT effects on human PDAC have yet to be thoroughly investigated; therefore, this study aimed to characterize immunomodulation in the human PDAC tumor microenvironment following therapy. EXPERIMENTAL DESIGN Tumor samples were obtained from patients with resectable PDAC. Radiotherapy was delivered a median of 7 days prior to surgical resection, and sections were analyzed by multiplex IHC (mIHC), RNA sequencing, and T-cell receptor sequencing (TCR-seq). RESULTS Analysis of SBRT-treated tumor tissue indicated reduced tumor cell density and increased immunogenic cell death relative to untreated controls. Radiotherapy promoted collagen deposition; however, vasculature was unaffected and spatial analyses lacked evidence of T-cell sequestration. Conversely, SBRT resulted in fewer tertiary lymphoid structures and failed to lessen or reprogram abundant immune suppressor populations. Higher percentages of PD-1+ T cells were observed following SBRT, and a subset of tumors displayed more clonal T-cell repertoires. CONCLUSIONS These findings suggest that SBRT augmentation of antitumor immunogenicity may be dampened by an overabundance of refractory immunosuppressive populations, and support the continued development of SBRT/immunotherapy combination for human PDAC.
Collapse
Affiliation(s)
- Bradley N Mills
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
| | - Haoming Qiu
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York.,Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Michael G Drage
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York
| | - Chunmo Chen
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York
| | - Jocelyn S Mathew
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
| | - Jesse Garrett-Larsen
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
| | - Jian Ye
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
| | - Taylor P Uccello
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
| | - Joseph D Murphy
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
| | - Brian A Belt
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
| | - Edith M Lord
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York.,Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
| | - Alan W Katz
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York.,Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - David C Linehan
- Department of Surgery, University of Rochester Medical Center, Rochester, New York.,Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Scott A Gerber
- Department of Surgery, University of Rochester Medical Center, Rochester, New York. .,Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York.,Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
25
|
Heger U, Hackert T. Can local ablative techniques replace surgery for locally advanced pancreatic cancer? J Gastrointest Oncol 2021; 12:2536-2546. [PMID: 34790414 DOI: 10.21037/jgo-20-379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 11/04/2020] [Indexed: 12/25/2022] Open
Abstract
In the treatment of pancreatic ductal adenocarcinoma (PDAC) the best chance at long term survival or cure has to date always included the complete surgical removal of the tumor. However, locally advanced pancreatic cancer (LAPC), about 25% of all newly diagnosed PDAC, is defined by its primary technical unresectability due to infiltration of visceral arteries and absence of metastasis. Induction therapies, especially FOLFIRINOX treatment, together with technical surgical advancement have increased the numbers for conversion to secondary resectability. Recent data on resections after induction therapy show promising, almost doubled survival compared to palliative treatment. Yet, around 70% of LAPC remain unresectable after induction therapy, often due to persistent local invasion. As locally ablative techniques are becoming more widely available this review examines their possible applicability to substitute for surgery in these cases which we propose to group under the new term "Inconvertible LAPC". The need for defining this novel subgroup who might benefit from ablative treatment is based on the findings in our review that high-level evidence on ablative techniques for PDAC is largely lacking and the latest effective, harmonized treatment guidelines for LAPC are not often incorporated in these studies. The "inconvertible LAPC" label requires persistent unresectability after staging and induction therapy of LAPC according to current guidelines followed by liberal indication for aggressive surgical exploration at a center equipped for extended pancreatic resections. Ideally, this specification of a new, distinct patient group will also put it in the spotlight more, hopefully prompt more trials designed to generate robust evidence and optimize transferability of study results.
Collapse
Affiliation(s)
- Ulrike Heger
- Department of Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Thilo Hackert
- Department of Surgery, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
26
|
He C, Sun S, Zhang Y, Li S. Irreversible Electroporation Plus Anti-PD-1 Antibody versus Irreversible Electroporation Alone for Patients with Locally Advanced Pancreatic Cancer. J Inflamm Res 2021; 14:4795-4807. [PMID: 34584438 PMCID: PMC8464362 DOI: 10.2147/jir.s331023] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/09/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Irreversible electroporation (IRE) is shown to not only improve the prognosis of patients with locally advanced pancreatic cancer (LAPC) but also activate the immune system. Considering the immune-activating function of IRE, IRE may enhance the effect of immune checkpoint inhibitors in the treatment of LAPC. We aimed to compare the effect and safety of IRE combined with toripalimab versus IRE alone for LAPC. METHODS We retrospectively collected data from LAPC patients treated with IRE plus toripalimab (240mg, 7 days after IRE) or IRE alone at Sun Yat‑sen University Cancer Center. Overall and progression-free survival and treatment-related adverse events were evaluated and compared. RESULTS From August 2015 to June 2020, a total of 85 patients were collected and analyzed in this study: 70 in the IRE group and 15 in the IRE plus toripalimab group. The IRE plus toripalimab group showed longer OS [44.33 months (95% CI 17.39-71.27) versus 23.37 months (95% CI 21.20-25.54), P=0.010] and PFS [27.5 months (95% CI not reached) versus 10.6 months (95% CI 7.79-13.42), P=0.036], compared with IRE group. There were no treatment-related deaths in all patients of this study. Although pancreatic fistula, biliary fistula, abscess, vomiting and gastroparesis were a little more common in IRE plus toripalimab group, no significant differences in the rates of all adverse events between these two groups were observed. CONCLUSION IRE plus toripalimab had acceptable toxic effects and might improve survival in LAPC compared with IRE alone.
Collapse
Affiliation(s)
- Chaobin He
- Department of Pancreatobiliary Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China
| | - Shuxin Sun
- Department of General Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, People’s Republic of China
| | - Yu Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, People’s Republic of China
| | - Shengping Li
- Department of Pancreatobiliary Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China
| |
Collapse
|
27
|
Pancreatic Cancer and Immunotherapy: A Clinical Overview. Cancers (Basel) 2021; 13:cancers13164138. [PMID: 34439292 PMCID: PMC8393975 DOI: 10.3390/cancers13164138] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with high mortality. The vast majority of patients present with unresectable, advanced stage disease, for whom standard of care chemo(radio)therapy may improve survival by several months. Immunotherapy has led to a fundamental shift in the treatment of several advanced cancers. However, its efficacy in PDAC in terms of clinical benefit is limited, possibly owing to the immunosuppressive, inaccessible tumor microenvironment. Still, various immunotherapies have demonstrated the capacity to initiate local and systemic immune responses, suggesting an immune potentiating effect. In this review, we address PDAC's immunosuppressive tumor microenvironment and immune evasion methods and discuss a wide range of immunotherapies, including immunomodulators (i.e., immune checkpoint inhibitors, immune stimulatory agonists, cytokines and adjuvants), oncolytic viruses, adoptive cell therapies (i.e., T cells and natural killer cells) and cancer vaccines. We provide a general introduction to their working mechanism as well as evidence of their clinical efficacy and immune potentiating abilities in PDAC. The key to successful implementation of immunotherapy in this disease may rely on exploitation of synergistic effects between treatment combinations. Accordingly, future treatment approaches should aim to incorporate diverse and novel immunotherapeutic strategies coupled with cytotoxic drugs and/or local ablative treatment, targeting a wide array of tumor-induced immune escape mechanisms.
Collapse
|
28
|
Geboers B, Timmer FEF, Ruarus AH, Pouw JEE, Schouten EAC, Bakker J, Puijk RS, Nieuwenhuizen S, Dijkstra M, van den Tol MP, de Vries JJJ, Oprea-Lager DE, Menke-van der Houven van Oordt CW, van der Vliet HJ, Wilmink JW, Scheffer HJ, de Gruijl TD, Meijerink MR. Irreversible Electroporation and Nivolumab Combined with Intratumoral Administration of a Toll-Like Receptor Ligand, as a Means of In Vivo Vaccination for Metastatic Pancreatic Ductal Adenocarcinoma (PANFIRE-III). A Phase-I Study Protocol. Cancers (Basel) 2021; 13:cancers13153902. [PMID: 34359801 PMCID: PMC8345515 DOI: 10.3390/cancers13153902] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/24/2021] [Accepted: 07/28/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Metastatic pancreatic ductal adenocarcinoma has a dismal prognosis, and to date no curative treatment options exist. The image guided tumor ablation technique irreversible electroporation (IRE) employs high-voltage electrical pulses through the application of several needle electrodes in and around the tumor in order to induce cell death. IRE ablation of the primary tumor has the ability to reduce pancreatic tumor induced immune suppression while allowing the expansion of tumor specific effector T cells, hereby possibly shifting the pancreatic tumor microenvironment into a more immune permissive state. The addition of immune enhancing therapies to IRE might work synergistically and could potentially induce a clinically significant treatment effect. This study protocol describes the rationale and design of the PANFIRE-III trial that aims to assess the safety of the combination of IRE with IMO-2125 (toll-like receptor 9 ligand) and/or nivolumab in patients with metastatic pancreatic ductal adenocarcinoma. Abstract Irreversible electroporation (IRE) is a novel image-guided tumor ablation technique with the ability to generate a window for the establishment of systemic antitumor immunity. IRE transiently alters the tumor’s immunosuppressive microenvironment while simultaneously generating antigen release, thereby instigating an adaptive immune response. Combining IRE with immunotherapeutic drugs, i.e., electroimmunotherapy, has synergistic potential and might induce a durable antitumor response. The primary objective of this study is to assess the safety of the combination of IRE with IMO-2125 (a toll-like receptor 9 ligand) and/or nivolumab in patients with metastatic pancreatic ductal adenocarcinoma (mPDAC). In this randomized controlled phase I clinical trial, 18 patients with mPDAC pretreated with chemotherapy will be enrolled in one of three study arms: A (control): nivolumab monotherapy; B: percutaneous IRE of the primary tumor followed by nivolumab; or C: intratumoral injection of IMO-2125 followed by percutaneous IRE of the primary tumor and nivolumab. Assessments include contrast enhanced computed tomography (ceCT), 18F-FDG and 18F-BMS-986192 (PD-L1) positron emission tomography (PET)-CT, biopsies of the primary tumor and metastases, peripheral blood samples, and quality of life and pain questionnaires. There is no curative treatment option for patients with mPDAC, and palliative chemotherapy regimens only moderately improve survival. Consequently, there is an urgent need for innovative and radically different treatment approaches. Should electroimmunotherapy establish an effective and durable anti-tumor response, it may ultimately improve PDAC’s dismal prognosis.
Collapse
Affiliation(s)
- Bart Geboers
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (F.E.F.T.); (A.H.R.); (E.A.C.S.); (R.S.P.); (S.N.); (M.D.); (J.J.J.d.V.); (D.E.O.-L.); (H.J.S.); (M.R.M.)
- Correspondence:
| | - Florentine E. F. Timmer
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (F.E.F.T.); (A.H.R.); (E.A.C.S.); (R.S.P.); (S.N.); (M.D.); (J.J.J.d.V.); (D.E.O.-L.); (H.J.S.); (M.R.M.)
| | - Alette H. Ruarus
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (F.E.F.T.); (A.H.R.); (E.A.C.S.); (R.S.P.); (S.N.); (M.D.); (J.J.J.d.V.); (D.E.O.-L.); (H.J.S.); (M.R.M.)
| | - Johanna E. E. Pouw
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (J.E.E.P.); (J.B.); (C.W.M.-v.d.H.v.O.); (H.J.v.d.V.); (J.W.W.); (T.D.d.G.)
| | - Evelien A. C. Schouten
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (F.E.F.T.); (A.H.R.); (E.A.C.S.); (R.S.P.); (S.N.); (M.D.); (J.J.J.d.V.); (D.E.O.-L.); (H.J.S.); (M.R.M.)
| | - Joyce Bakker
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (J.E.E.P.); (J.B.); (C.W.M.-v.d.H.v.O.); (H.J.v.d.V.); (J.W.W.); (T.D.d.G.)
| | - Robbert S. Puijk
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (F.E.F.T.); (A.H.R.); (E.A.C.S.); (R.S.P.); (S.N.); (M.D.); (J.J.J.d.V.); (D.E.O.-L.); (H.J.S.); (M.R.M.)
| | - Sanne Nieuwenhuizen
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (F.E.F.T.); (A.H.R.); (E.A.C.S.); (R.S.P.); (S.N.); (M.D.); (J.J.J.d.V.); (D.E.O.-L.); (H.J.S.); (M.R.M.)
| | - Madelon Dijkstra
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (F.E.F.T.); (A.H.R.); (E.A.C.S.); (R.S.P.); (S.N.); (M.D.); (J.J.J.d.V.); (D.E.O.-L.); (H.J.S.); (M.R.M.)
| | - M. Petrousjka van den Tol
- Department of Surgery, Amsterdam University Medical Centers, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands;
| | - Jan J. J. de Vries
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (F.E.F.T.); (A.H.R.); (E.A.C.S.); (R.S.P.); (S.N.); (M.D.); (J.J.J.d.V.); (D.E.O.-L.); (H.J.S.); (M.R.M.)
| | - Daniela E. Oprea-Lager
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (F.E.F.T.); (A.H.R.); (E.A.C.S.); (R.S.P.); (S.N.); (M.D.); (J.J.J.d.V.); (D.E.O.-L.); (H.J.S.); (M.R.M.)
| | - C. Willemien Menke-van der Houven van Oordt
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (J.E.E.P.); (J.B.); (C.W.M.-v.d.H.v.O.); (H.J.v.d.V.); (J.W.W.); (T.D.d.G.)
| | - Hans J. van der Vliet
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (J.E.E.P.); (J.B.); (C.W.M.-v.d.H.v.O.); (H.J.v.d.V.); (J.W.W.); (T.D.d.G.)
- Lava Therapeutics, Yalelaan 60, 3584 CM Utrecht, The Netherlands
| | - Johanna W. Wilmink
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (J.E.E.P.); (J.B.); (C.W.M.-v.d.H.v.O.); (H.J.v.d.V.); (J.W.W.); (T.D.d.G.)
| | - Hester J. Scheffer
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (F.E.F.T.); (A.H.R.); (E.A.C.S.); (R.S.P.); (S.N.); (M.D.); (J.J.J.d.V.); (D.E.O.-L.); (H.J.S.); (M.R.M.)
| | - Tanja D. de Gruijl
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (J.E.E.P.); (J.B.); (C.W.M.-v.d.H.v.O.); (H.J.v.d.V.); (J.W.W.); (T.D.d.G.)
| | - Martijn R. Meijerink
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Centers, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (F.E.F.T.); (A.H.R.); (E.A.C.S.); (R.S.P.); (S.N.); (M.D.); (J.J.J.d.V.); (D.E.O.-L.); (H.J.S.); (M.R.M.)
| | | |
Collapse
|
29
|
Burbach BJ, O'Flanagan SD, Shao Q, Young KM, Slaughter JR, Rollins MR, Street TJL, Granger VE, Beura LK, Azarin SM, Ramadhyani S, Forsyth BR, Bischof JC, Shimizu Y. Irreversible electroporation augments checkpoint immunotherapy in prostate cancer and promotes tumor antigen-specific tissue-resident memory CD8+ T cells. Nat Commun 2021; 12:3862. [PMID: 34162858 PMCID: PMC8222297 DOI: 10.1038/s41467-021-24132-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 06/02/2021] [Indexed: 01/04/2023] Open
Abstract
Memory CD8+ T cells populate non-lymphoid tissues (NLTs) following pathogen infection, but little is known about the establishment of endogenous tumor-specific tissue-resident memory T cells (TRM) during cancer immunotherapy. Using a transplantable mouse model of prostate carcinoma, here we report that tumor challenge leads to expansion of naïve neoantigen-specific CD8+ T cells and formation of a small population of non-recirculating TRM in several NLTs. Primary tumor destruction by irreversible electroporation (IRE), followed by anti-CTLA-4 immune checkpoint inhibitor (ICI), promotes robust expansion of tumor-specific CD8+ T cells in blood, tumor, and NLTs. Parabiosis studies confirm that TRM establishment following dual therapy is associated with tumor remission in a subset of cases and protection from subsequent tumor challenge. Addition of anti-PD-1 following dual IRE + anti-CTLA-4 treatment blocks tumor growth in non-responsive cases. This work indicates that focal tumor destruction using IRE combined with ICI is a potent in situ tumor vaccination strategy that generates protective tumor-specific TRM.
Collapse
Affiliation(s)
- Brandon J Burbach
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA.
- Center for Immunology, University of Minnesota, Minneapolis, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, USA.
- Institute for Engineering in Medicine, University of Minnesota, Minneapolis, USA.
| | - Stephen D O'Flanagan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, USA
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, USA
| | - Qi Shao
- Masonic Cancer Center, University of Minnesota, Minneapolis, USA
- Institute for Engineering in Medicine, University of Minnesota, Minneapolis, USA
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, USA
| | - Katharine M Young
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, USA
| | - Joseph R Slaughter
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, USA
| | - Meagan R Rollins
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, USA
- Boston Scientific Corporation, Maple Grove, MN, USA
| | - Tami Jo L Street
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, USA
| | - Victoria E Granger
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, USA
| | - Lalit K Beura
- Center for Immunology, University of Minnesota, Minneapolis, USA
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, USA
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Samira M Azarin
- Masonic Cancer Center, University of Minnesota, Minneapolis, USA
- Institute for Engineering in Medicine, University of Minnesota, Minneapolis, USA
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, USA
| | | | | | - John C Bischof
- Masonic Cancer Center, University of Minnesota, Minneapolis, USA
- Institute for Engineering in Medicine, University of Minnesota, Minneapolis, USA
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, USA
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, USA
| | - Yoji Shimizu
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA.
- Center for Immunology, University of Minnesota, Minneapolis, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, USA.
- Institute for Engineering in Medicine, University of Minnesota, Minneapolis, USA.
| |
Collapse
|
30
|
Batista Napotnik T, Polajžer T, Miklavčič D. Cell death due to electroporation - A review. Bioelectrochemistry 2021; 141:107871. [PMID: 34147013 DOI: 10.1016/j.bioelechem.2021.107871] [Citation(s) in RCA: 217] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/12/2021] [Accepted: 06/03/2021] [Indexed: 12/15/2022]
Abstract
Exposure of cells to high voltage electric pulses increases transiently membrane permeability through membrane electroporation. Electroporation can be reversible and is used in gene transfer and enhanced drug delivery but can also lead to cell death. Electroporation resulting in cell death (termed as irreversible electroporation) has been successfully used as a new non-thermal ablation method of soft tissue such as tumours or arrhythmogenic heart tissue. Even though the mechanisms of cell death can influence the outcome of electroporation-based treatments due to use of different electric pulse parameters and conditions, these are not elucidated yet. We review the mechanisms of cell death after electroporation reported in literature, cell injuries that may lead to cell death after electroporation and membrane repair mechanisms involved. The knowledge of membrane repair and cell death mechanisms after cell exposure to electric pulses, targets of electric field in cells need to be identified to optimize existing and develop of new electroporation-based techniques used in medicine, biotechnology, and food technology.
Collapse
Affiliation(s)
- Tina Batista Napotnik
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, 1000 Ljubljana, Slovenia
| | - Tamara Polajžer
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, 1000 Ljubljana, Slovenia
| | - Damijan Miklavčič
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, 1000 Ljubljana, Slovenia.
| |
Collapse
|
31
|
Hendricks-Wenger A, Hutchison R, Vlaisavljevich E, Allen IC. Immunological Effects of Histotripsy for Cancer Therapy. Front Oncol 2021; 11:681629. [PMID: 34136405 PMCID: PMC8200675 DOI: 10.3389/fonc.2021.681629] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/10/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer is the second leading cause of death worldwide despite major advancements in diagnosis and therapy over the past century. One of the most debilitating aspects of cancer is the burden brought on by metastatic disease. Therefore, an ideal treatment protocol would address not only debulking larger primary tumors but also circulating tumor cells and distant metastases. To address this need, the use of immune modulating therapies has become a pillar in the oncology armamentarium. A therapeutic option that has recently emerged is the use of focal ablation therapies that can destroy a tumor through various physical or mechanical mechanisms and release a cellular lysate with the potential to stimulate an immune response. Histotripsy is a non-invasive, non-ionizing, non-thermal, ultrasound guided ablation technology that has shown promise over the past decade as a debulking therapy. As histotripsy therapies have developed, the full picture of the accompanying immune response has revealed a wide range of immunogenic mechanisms that include DAMP and anti-tumor mediator release, changes in local cellular immune populations, development of a systemic immune response, and therapeutic synergism with the inclusion of checkpoint inhibitor therapies. These studies also suggest that there is an immune effect from histotripsy therapies across multiple murine tumor types that may be reproducible. Overall, the effects of histotripsy on tumors show a positive effect on immunomodulation.
Collapse
Affiliation(s)
- Alissa Hendricks-Wenger
- Graduate Program in Translational Biology, Medicine and Health, Virginia Tech, Roanoke, VA, United States
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, United States
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, United States
| | - Ruby Hutchison
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, United States
| | - Eli Vlaisavljevich
- Graduate Program in Translational Biology, Medicine and Health, Virginia Tech, Roanoke, VA, United States
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, United States
- Institute for Critical Technology and Applied Sciences Center for Engineered Health, Virginia Tech, Blacksburg, VA, United States
| | - Irving Coy Allen
- Graduate Program in Translational Biology, Medicine and Health, Virginia Tech, Roanoke, VA, United States
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, United States
- Institute for Critical Technology and Applied Sciences Center for Engineered Health, Virginia Tech, Blacksburg, VA, United States
- Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA, United States
| |
Collapse
|
32
|
He C, Sun S, Zhang Y, Li S. Identification of Circulating Biomarkers and Construction of a Prognostic Signature for Survival Prediction in Locally Advanced Pancreatic Cancer After Irreversible Electroporation. J Inflamm Res 2021; 14:1689-1699. [PMID: 33953596 PMCID: PMC8091593 DOI: 10.2147/jir.s307884] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/14/2021] [Indexed: 12/14/2022] Open
Abstract
Background Irreversible electroporation (IRE) is a novel treatment for locally advanced pancreatic cancer (LAPC), but the predictive factors, based on cytokines and immunocytes of survival, are still lacking. This study aimed to establish a risk model based on cytokines and immunocytes for LAPC patients undergoing IRE treatment. Patients and Methods Peripheral blood samples were obtained from 31 LAPC patients and 8 healthy control subjects before IRE. The phenotypes of lymphocytes were analyzed by flow cytometry, and the cytokines were evaluated with Luminex microarray assay. Least absolute shrinkage and selection operator (LASSO) and Cox regression were applied to assess the prognostic factors for overall survival (OS) and progression-free survival (PFS). A receiver operating characteristic (ROC) curve and a concordance index (C-index) were used to compare the abilities to predict survival rates. Results The relationship between multiple cytokines and clinical factors was evaluated and their prognostic value was compared. The five best predictors for OS and PFS, including CA19-9, CD3+CD4+ T cells, CD3+CD8+ T cells, IL-17A, and TNF-α were selected and incorporated into a new immune panel. A risk model based on this immune panel was established and exhibited significantly higher values of C-indexes and AUC for OS and PFS prediction as compared with tumor marker score and TNM stage system. Conclusion We presented a risk model based on a microarray assay of cytokines and lymphocytes for LAPC patients after receiving IRE treatment for the first time. The established risk model showed relatively good performance in survival prediction and was able to facilitate tailed patient management in clinical practice.
Collapse
Affiliation(s)
- Chaobin He
- Department of Pancreatobiliary Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Shuxin Sun
- Department of Pancreatobiliary Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Yu Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Shengping Li
- Department of Pancreatobiliary Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| |
Collapse
|
33
|
Guo X, Du F, Liu Q, Guo Y, Wang Q, Huang W, Wang Z, Ding X, Wu Z. Immunological effect of irreversible electroporation on hepatocellular carcinoma. BMC Cancer 2021; 21:443. [PMID: 33882892 PMCID: PMC8061072 DOI: 10.1186/s12885-021-08176-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 04/12/2021] [Indexed: 02/08/2023] Open
Abstract
Background This study intends to investigate the immunological effects of tumor ablation with irreversible electroporation (IRE). Methods We evaluated the systemic immune response in patients with hepatocellular carcinoma (HCC) after IRE treatment. Furthermore, we analyzed the tumor infiltrating T lymphocytes and the level of serum cytokines in IRE and control groups of tumor-bearing mice. Results We observed that IRE induced an increase in WBC, neutrophil and monocyte counts and a decrease in lymphocyte count 1 day post-IRE and returned to baseline values within 7 days in the patients. Meanwhile, circulating CD4+ T cell subsets, but not CD8+, decreased 1 day post-IRE. The activated T cells and natural killer (NK) cells increased, and regulatory T (Treg) cells decreased. Furthermore, a significant increase in cytotoxic CD8+ T cells infiltration was observed on ablative tumors in mice. The level of serum IFN-γ also significantly increased in the IRE group. Conclusions Our study demonstrated that IRE upregulated activated T cells and downregulated Tregs in the peripheral blood of patients. Meanwhile, the results from the animal model indicated that IRE could induce antitumor adaptive immunity dominated by the infiltration of cytotoxic CD8+ T cells into the tumors, accompanied by reduced Tregs.
Collapse
Affiliation(s)
- Xiaoxia Guo
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Fang Du
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qin Liu
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yan Guo
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qingbing Wang
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wei Huang
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhongmin Wang
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaoyi Ding
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Zhiyuan Wu
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
34
|
Timmer FE, Geboers B, Nieuwenhuizen S, Schouten EA, Dijkstra M, de Vries JJ, van den Tol MP, de Gruijl TD, Scheffer HJ, Meijerink MR. Locally Advanced Pancreatic Cancer: Percutaneous Management Using Ablation, Brachytherapy, Intra-arterial Chemotherapy, and Intra-tumoral Immunotherapy. Curr Oncol Rep 2021; 23:68. [PMID: 33864144 PMCID: PMC8052234 DOI: 10.1007/s11912-021-01057-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive neoplasms, bearing a terrible prognosis. Stage III tumors, also known as locally advanced pancreatic cancer (LAPC), are unresectable, and current palliative chemotherapy regimens have only modestly improved survival in these patients. At this stage of disease, interventional techniques may be of value and further prolong life. The aim of this review was to explore current literature on locoregional percutaneous management for LAPC. RECENT FINDINGS Locoregional percutaneous interventional techniques such as ablation, brachytherapy, and intra-arterial chemotherapy possess cytoreductive abilities and have the potential to increase survival. In addition, recent research demonstrates the immunomodulatory capacities of these treatments. This immune response may be leveraged by combining the interventional techniques with intra-tumoral immunotherapy, possibly creating a durable anti-tumor effect. This multimodality treatment approach is currently being examined in several ongoing clinical trials. The use of certain interventional techniques appears to improve survival in LAPC patients and may work synergistically when combined with immunotherapy. However, definitive conclusions can only be made when large prospective (randomized controlled) trials confirm these results.
Collapse
Affiliation(s)
- Florentine E.F. Timmer
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Bart Geboers
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Sanne Nieuwenhuizen
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Evelien A.C. Schouten
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Madelon Dijkstra
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Jan J.J. de Vries
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - M. Petrousjka van den Tol
- Department of Surgical Oncology, Amsterdam UMC (location VUmc), De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Tanja D. de Gruijl
- Department of Medical Oncology, Amsterdam UMC (location VUmc)-Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Hester J. Scheffer
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Martijn R. Meijerink
- Department of Radiology and Nuclear Medicine, Amsterdam UMC (location VUmc), De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
35
|
Rai ZL, Feakins R, Pallett LJ, Manas D, Davidson BR. Irreversible Electroporation (IRE) in Locally Advanced Pancreatic Cancer: A Review of Current Clinical Outcomes, Mechanism of Action and Opportunities for Synergistic Therapy. J Clin Med 2021; 10:1609. [PMID: 33920118 PMCID: PMC8068938 DOI: 10.3390/jcm10081609] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/04/2021] [Accepted: 04/07/2021] [Indexed: 12/11/2022] Open
Abstract
Locally advanced pancreatic cancer (LAPC) accounts for 30% of patients with pancreatic cancer. Irreversible electroporation (IRE) is a novel cancer treatment that may improve survival and quality of life in LAPC. This narrative review will provide a perspective on the clinical experience of pancreas IRE therapy, explore the evidence for the mode of action, assess treatment complications, and propose strategies for augmenting IRE response. A systematic search was performed using PubMed regarding the clinical use and safety profile of IRE on pancreatic cancer, post-IRE sequential histological changes, associated immune response, and synergistic therapies. Animal data demonstrate that IRE induces both apoptosis and necrosis followed by fibrosis. Major complications may result from IRE; procedure related mortality is up to 2%, with an average morbidity as high as 36%. Nevertheless, prospective and retrospective studies suggest that IRE treatment may increase median overall survival of LAPC to as much as 30 months and provide preliminary data justifying the well-designed trials currently underway, comparing IRE to the standard of care treatment. The mechanism of action of IRE remains unknown, and there is a lack of data on treatment variables and efficiency in humans. There is emerging data suggesting that IRE can be augmented with synergistic therapies such as immunotherapy.
Collapse
Affiliation(s)
- Zainab L. Rai
- Centre of Surgical Innovation, Organ Regeneration and Transplantation, University College London (UCL), London NW3 2QG, UK;
- Wellcome/EPSRC Center for Interventional and Surgical Sciences (WEISS), London W1W 7TY, UK
- Royal Free NHS Foundation Trust, London NW3 2QG, UK;
| | - Roger Feakins
- Royal Free NHS Foundation Trust, London NW3 2QG, UK;
| | - Laura J. Pallett
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London WC1E 6BT, UK;
| | - Derek Manas
- Newcastle Upon Tyne NHS Foundation Trust, Newcastle-Upon-Tyne NE7 7DN, UK;
| | - Brian R. Davidson
- Centre of Surgical Innovation, Organ Regeneration and Transplantation, University College London (UCL), London NW3 2QG, UK;
- Royal Free NHS Foundation Trust, London NW3 2QG, UK;
| |
Collapse
|
36
|
He C, Sun S, Zhang Y, Xie F, Li S. The role of irreversible electroporation in promoting M1 macrophage polarization via regulating the HMGB1-RAGE-MAPK axis in pancreatic cancer. Oncoimmunology 2021; 10:1897295. [PMID: 33763295 PMCID: PMC7954423 DOI: 10.1080/2162402x.2021.1897295] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/01/2021] [Accepted: 02/24/2021] [Indexed: 12/13/2022] Open
Abstract
Irreversible electroporation (IRE) is an effective method for treating pancreatic ductal adenocarcinoma (PDAC). It remains unclear whether IRE can induce a specific immune response by stimulating macrophages. Here, the associated markers of macrophages were analyzed after exposure to tumor culture supernatant (TSN) of tumor cells treated with electroporation. Subcutaneous and orthotopic PDAC models were also used to evaluate the effect of macrophage polarization induced by IRE. Aside from its direct killing effect, IRE could induce the immunogenic cell death of tumor cells by increasing the synthesis and secretion of damage associated molecular patterns. Moreover, IRE could increase the release of HMGB1, which activates the MAPK-p38 pathway and leads to the increased expression of M1 markers in macrophages, through binding to the receptor of the advanced glycation end-product (RAGE) receptor. M1 polarization was inhibited by the inhibitors of HMGB1 release, the RAGE receptor, and the MAPK-p38 signaling pathway, but it was activated by rHMGB1 or the stimulator of MAPK-p38. In addition, the promotion of M1 macrophage polarization was enhanced by the positive-feedback release or expression of HMGB1 and RAGE through the MAPK-ERK pathway in macrophages. The promotion of M1 macrophage polarization induced by IRE provided a specific rationale for the combination of IRE and immune therapy in treating PDAC.
Collapse
Affiliation(s)
- Chaobin He
- Department of Pancreatobiliary Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Shuxin Sun
- Department of Pancreatobiliary Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Yu Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Fengxiao Xie
- Department of Pancreatobiliary Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Shengping Li
- Department of Pancreatobiliary Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| |
Collapse
|
37
|
Kwon W, Thomas A, Kluger MD. Irreversible electroporation of locally advanced pancreatic cancer. Semin Oncol 2021; 48:84-94. [PMID: 33648735 DOI: 10.1053/j.seminoncol.2021.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/18/2021] [Accepted: 02/02/2021] [Indexed: 12/14/2022]
Abstract
Locally advanced pancreatic cancer (LAPC) constitutes approximately one-third of all pancreatic cancer, with standard of care inconsistently defined and achieving modest outcomes at best. While resection after downstaging offers the chance for cure, only a fraction of patients with LAPC become candidates for resection. Chemotherapy remains the mainstay of treatment for the remainder. In these patients, ablative therapy may be given for local control of the tumor. Irreversible electroporation (IRE) is an attractive ablative technique. IRE changes the permeability of tumor cell membranes to induce apoptosis. Unlike other ablative therapies, IRE causes little thermal injury to the target area, making it ideal for LAPC involving major vessels. Compared to systemic chemotherapy alone, IRE seems to offer some survival benefit. Although early studies reported notable morbidity and mortality rates, IRE presents opportunities for those who cannot undergo resection and who otherwise have limited options. Another role of IRE is to extend the margins of resected tumors when there is a concern for R1 resection. Perhaps most exciting, IRE is thought to have effects beyond local ablation. IRE has immunomodulatory effects, which may induce in vivo vaccination and may potentially synergize with immunotherapy. Through electrochemotherapy, IRE may enhance drug delivery to residual tumor cells. Ultimately the role of IRE in the treatment of LAPC still needs to be validated through well designed randomized trials. Investigations of its future possibilities are in the early stages. IRE offers the potential to provide more options to LAPC patients.
Collapse
Affiliation(s)
- Wooil Kwon
- Division of GI/Endocrine Surgery, Department of Surgery, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA; Division of Hepatobiliary & Pancreatic Surgery, Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Alexander Thomas
- Division of GI/Endocrine Surgery, Department of Surgery, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Michael D Kluger
- Division of GI/Endocrine Surgery, Department of Surgery, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
38
|
Dai Z, Wang Z, Lei K, Liao J, Peng Z, Lin M, Liang P, Yu J, Peng S, Chen S, Kuang M. Irreversible electroporation induces CD8 + T cell immune response against post-ablation hepatocellular carcinoma growth. Cancer Lett 2021; 503:1-10. [PMID: 33444692 DOI: 10.1016/j.canlet.2021.01.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/06/2020] [Accepted: 01/01/2021] [Indexed: 12/18/2022]
Abstract
Ablative treatment evokes antitumor immunity, but knowledge on the emerging irreversible electroporation (IRE)-induced immunity in hepatocellular carcinoma (HCC) is limited. To investigate the immune effects induced by IRE and its role in preventing post-ablation HCC progression, a C57BL/6J mouse model bearing subcutaneous H22 hepatoma was employed. IRE treatment significantly suppresses HCC growth, and treated mice are tumor-free after secondary tumor injection and show increased splenic interferon-gamma (IFN-γ)+CD8+ T cells. Additionally, more CD8+ T and dendritic cells, but not CD4+ T, B or NK cells, infiltrate into peri-ablation zones after IRE at day 7. Depletion of CD8+ T cells induces local tumor regrowth and distant metastasis after IRE. Vaccination using IRE-processed H22 lysates prevents tumorigenesis in mice, suggesting a protective immune response. IRE also alleviates immunosuppression by reducing local and splenic Treg and PD-1+ T cells. Regarding mechanism, IRE induces cell necrosis and significant release of danger-associated molecular patterns including ATP, high mobility group box 1 and calreticulin that are pivotal to CD8+ T cell immunity. Together, IRE is a promising approach to evoke CD8+ T cell immunity, which help prevent post-ablation HCC progression.
Collapse
Affiliation(s)
- Zihao Dai
- Department of Liver Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zongren Wang
- Department of Urology Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Kai Lei
- Department of Liver Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Junbin Liao
- Department of Liver Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhenwei Peng
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Manxia Lin
- Division of Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ping Liang
- Division of Interventional Ultrasound, The Chinese PLA General Hospital, Beijing, China
| | - Jie Yu
- Division of Interventional Ultrasound, The Chinese PLA General Hospital, Beijing, China
| | - Sui Peng
- Clinical Trial Unit, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shuling Chen
- Division of Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Ming Kuang
- Department of Liver Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Division of Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
39
|
Sodergren MH, Mangal N, Wasan H, Sadanandam A, Balachandran VP, Jiao LR, Habib N. Immunological combination treatment holds the key to improving survival in pancreatic cancer. J Cancer Res Clin Oncol 2020; 146:2897-2911. [PMID: 32748119 PMCID: PMC7519893 DOI: 10.1007/s00432-020-03332-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/22/2020] [Indexed: 12/18/2022]
Abstract
Advances in surgery, peri-operative care and systemic chemotherapy have not significantly improved the prognosis of pancreatic cancer for several decades. Early clinical trials of immunotherapy have yielded disappointing results proposing other means by which the tumour microenvironment serves to decrease the immune response. Additionally, the emergence of various subtypes of pancreatic cancer has emerged as a factor for treatment responses with immunogenic subtypes carrying a better prognosis. Herein we discuss the reasons for the poor response to checkpoint inhibitors and outline a rationale why combination treatments are likely to be most effective. We review the therapies which could provide optimal synergistic effects to immunotherapy including chemotherapy, agents targeting the stroma, co-stimulatory molecules, vaccinations and methods of immunogenic tumour priming including radiofrequency ablation. Finally, we discuss reasons why peri-operative and in particular neoadjuvant combination treatments are likely to be most effective and should be considered for early clinical trials.
Collapse
Affiliation(s)
- M H Sodergren
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, DuCane Road, London, W12 0HS, UK.
| | - N Mangal
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, DuCane Road, London, W12 0HS, UK
| | - H Wasan
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, DuCane Road, London, W12 0HS, UK
| | - A Sadanandam
- Division of Molecular Pathology, Institute for Cancer Research, London, UK
- Centre for Molecular Pathology, Royal Marsden Hospital, London, UK
| | - V P Balachandran
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, USA
| | - L R Jiao
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, DuCane Road, London, W12 0HS, UK
| | - N Habib
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, DuCane Road, London, W12 0HS, UK
| |
Collapse
|
40
|
O'Neill C, Hayat T, Hamm J, Healey M, Zheng Q, Li Y, Martin RCG. A phase 1b trial of concurrent immunotherapy and irreversible electroporation in the treatment of locally advanced pancreatic adenocarcinoma. Surgery 2020; 168:610-616. [PMID: 32631655 DOI: 10.1016/j.surg.2020.04.057] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/16/2020] [Accepted: 04/25/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Combining immune checkpoint blockade therapy with operative disruptive immunomodulation using irreversible electroporation may overcome the resistance to systemic therapy found in patients with locally advanced, unresectable pancreatic cancer. We describe the safety profile and efficacy of IRE with nivolumab. METHODS In the preclinical phase of study, human pancreatic cell lines were cultured with interferon-γ (10 ng/mL) and murine models of pancreatic cancer were treated with irreversible electroporation and programmed death ligand-1 (PD-L1) expression was measured. In this phase 1b clinical trial (NCT03080974), surgical ablative irreversible electroporation was performed followed by nivolumab. The primary end point was dose-limiting toxicity. RESULTS Human pancreatic cells express PD-L1 when cultured with interferon-γ: quantitative polymerase chain reaction MiaPaca (15.2 rel. fold ± 0.5; P < .01) and S20-13 (31.0 rel. fold ± 4.4; P < .01). Murine orthotopic tumors treated by irreversible electroporation had an increase in signal intensity score for the expression of PD-L1 in residual tumor (P < .01). Ten patients were included in the safety analysis with a 12-month median follow-up (interquartile range 6.0, 15.8). No dose-limiting toxicities occurred. Seven patients developed grade 3/4 treatment-related adverse events; none required a dose modification of nivolumab; nivolumab-related adverse events occurred in 1 patient. Mean time to progression was 6.3 months (confidence interval 3.5-10.0) with current median overall survival of 18.0 months (confidence interval 9.2-26.8). CONCLUSION Irreversible electroporation induces expression of PD-L1 in vitro. Combination therapy with concurrent nivolumab is well tolerated. A multicenter, phase 2 adjuvant trial is underway using irreversible electroporation and nivolumab in patients with locally advanced pancreatic cancer.
Collapse
Affiliation(s)
- Conor O'Neill
- Hiram Polk, Jr. MD Department of Surgery, University of Louisville, KY
| | - Traci Hayat
- Hiram Polk, Jr. MD Department of Surgery, University of Louisville, KY
| | - John Hamm
- Hiram Polk, Jr. MD Department of Surgery, University of Louisville, KY
| | - Mary Healey
- Hiram Polk, Jr. MD Department of Surgery, University of Louisville, KY
| | - Qianqian Zheng
- Hiram Polk, Jr. MD Department of Surgery, University of Louisville, KY
| | - Yan Li
- Hiram Polk, Jr. MD Department of Surgery, University of Louisville, KY
| | - Robert C G Martin
- Hiram Polk, Jr. MD Department of Surgery, University of Louisville, KY.
| |
Collapse
|
41
|
Kiełbik A, Szlasa W, Saczko J, Kulbacka J. Electroporation-Based Treatments in Urology. Cancers (Basel) 2020; 12:E2208. [PMID: 32784598 PMCID: PMC7465806 DOI: 10.3390/cancers12082208] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 02/06/2023] Open
Abstract
The observation that an application of a pulsed electric field (PEF) resulted in an increased permeability of the cell membrane has led to the discovery of the phenomenon called electroporation (EP). Depending on the parameters of the electric current and cell features, electroporation can be either reversible or irreversible. The irreversible electroporation (IRE) found its use in urology as a non-thermal ablative method of prostate and renal cancer. As its mechanism is based on the permeabilization of cell membrane phospholipids, IRE (as well as other treatments based on EP) provides selectivity sparing extracellular proteins and matrix. Reversible EP enables the transfer of genes, drugs, and small exogenous proteins. In clinical practice, reversible EP can locally increase the uptake of cytotoxic drugs such as cisplatin and bleomycin. This approach is known as electrochemotherapy (ECT). Few in vivo and in vitro trials of ECT have been performed on urological cancers. EP provides the possibility of transmission of genes across the cell membrane. As the protocols of gene electrotransfer (GET) over the last few years have improved, EP has become a well-known technique for non-viral cell transfection. GET involves DNA transfection directly to the cancer or the host skin and muscle tissue. Among urological cancers, the GET of several plasmids encoding prostate cancer antigens has been investigated in clinical trials. This review brings into discussion the underlying mechanism of EP and an overview of the latest progress and development perspectives of EP-based treatments in urology.
Collapse
Affiliation(s)
- Aleksander Kiełbik
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (A.K.); (W.S.)
| | - Wojciech Szlasa
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (A.K.); (W.S.)
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| |
Collapse
|
42
|
Analysis of damage-associated molecular pattern molecules due to electroporation of cells in vitro. Radiol Oncol 2020; 54:317-328. [PMID: 32726295 PMCID: PMC7409611 DOI: 10.2478/raon-2020-0047] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 07/07/2020] [Indexed: 01/10/2023] Open
Abstract
Background Tumor cells can die via immunogenic cell death pathway, in which damage-associated molecular pattern molecules (DAMPs) are released from the cells. These molecules activate cells involved in the immune response. Both innate and adaptive immune response can be activated, causing a destruction of the remaining infected cells. Activation of immune response is also an important component of tumor treatment with electrochemotherapy (ECT) and irreversible electroporation (IRE). We thus explored, if and when specific DAMPs are released as a consequence of electroporation in vitro. Materials and methods In this in vitro study, 100 μs long electric pulses were applied to a suspension of Chinese hamster ovary cells. The release of DAMPs - specifically: adenosine triphosphate (ATP), calreticulin, nucleic acids and uric acid was investigated at different time points after exposing the cells to electric pulses of different amplitudes. The release of DAMPs was statistically correlated with cell permeabilization and cell survival, e.g. reversible and irreversible electroporation. Results In general, the release of DAMPs increases with increasing pulse amplitude. Concentration of DAMPs depend on the time interval between exposure of the cells to pulses and the analysis. Concentrations of most DAMPs correlate strongly with cell death. However, we detected no uric acid in the investigated samples. Conclusions Release of DAMPs can serve as a marker for prediction of cell death. Since the stability of certain DAMPs is time dependent, this should be considered when designing protocols for detecting DAMPs after electric pulse treatment.
Collapse
|
43
|
He C, Huang X, Zhang Y, Lin X, Li S. T-cell activation and immune memory enhancement induced by irreversible electroporation in pancreatic cancer. Clin Transl Med 2020; 10:e39. [PMID: 32508058 PMCID: PMC7403705 DOI: 10.1002/ctm2.39] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 12/16/2022] Open
Abstract
Background Irreversible electroporation is shown to induce immune changes in pancreatic cancer while the histology evidences are still lacking. The aim of this study is to show the immune changes in histology and explore whether irreversible electroporation (IRE) can induce immunogenic cell death (ICD) of tumor cells and activate specific immune responses. Methods Subcutaneous and orthotopic pancreatic cancer models were established and used to evaluate the effect of immune modulation of IRE. The infiltration of T cells was assessed in several tissue samples before and after IRE. Abscopal effect was then assessed by comparing the tumor growth of subcutaneous tumors after in situ ablation with IRE or exposure to tumor culture supernatant (TSN) of IRE‐treated Pan02. The expression of damage‐associated molecular patterns (DAMPs) of tumor cells after IRE was detected in vitro. Results IRE could significantly suppress the tumor growth and increase the infiltration of CD8+ T cells. After ablation with IRE or stimulation with TSN of Pan02 treated by IRE, the growth of untreated tumor was suppressed and the effector CD8+ T cells and memory T cells increased significantly in mice. Additionally, the inhibition effect of tumor growth increased along with the increasing strength levels of electroporation. IRE induced ICD of tumor cells by increasing the synthesis and secretion of DAMPs. Conclusions IRE induced local immunomodulation by increasing specific T cells infiltration. Through enhancing specific immune memory, IRE not only led a complete tumor regression in suit, but also induced abscopal effect, suppressing the growth of the latent lesions.
Collapse
Affiliation(s)
- Chaobin He
- Department of Pancreatobiliary Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Xin Huang
- Department of Pancreatobiliary Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Yu Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Xiaojun Lin
- Department of Pancreatobiliary Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Shengping Li
- Department of Pancreatobiliary Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| |
Collapse
|
44
|
Timmer FEF, Geboers B, Ruarus AH, Schouten EAC, Nieuwenhuizen S, Puijk RS, de Vries JJJ, Meijerink MR, Scheffer HJ. Irreversible Electroporation for Locally Advanced Pancreatic Cancer. Tech Vasc Interv Radiol 2020; 23:100675. [PMID: 32591191 DOI: 10.1016/j.tvir.2020.100675] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Several minimally invasive image guided tumor ablation techniques have been added to the treatment spectrum for locally advanced pancreatic cancer (LAPC). Irreversible electroporation (IRE) might have a significant additive value in the management of this difficult-to-treat disease. As opposed to thermal ablative techniques, IRE induces cell death by the delivery of high-voltage electrical pulses. The electrical energy disrupts the cellular membrane integrity, causes loss of cellular homeostasis and ultimately results in cell death. The extracellular matrix of connective tissue in surrounding delicate structures such as bile ducts, bowel wall, and larger blood vessels is spared. The preservation of these structures makes IRE attractive for the treatment of pancreatic cancers that are unresectable due to their anatomical location (ie, LAPC and local recurrence after surgical resection). In addition to its cytoreductive abilities, evidence is emerging on IRE's capability to induce systemic immunomodulation through active in vivo vaccination against pancreatic cancer cells. These effects in combination with immunotherapy may offer a new treatment paradigm for tumors with low immunogenic potential like pancreatic ductal adenocarcinoma (PDAC). This review discusses several practical and technical issues of IRE for LAPC: clinical evaluation, indications, patient preparations, procedural steps, imaging characteristics, clinical results, and "tricks of the trade" used to improve the safety and efficacy of the treatment. Future directions such as the combination of IRE with immunotherapy will be shortly addressed.
Collapse
Affiliation(s)
- Florentine E F Timmer
- Department of Radiology and Nuclear Medicine at the Amsterdam University Medical Center, Vrije Universiteit-Cancer Center Amsterdam in Amsterdam, The Netherlands
| | - Bart Geboers
- Department of Radiology and Nuclear Medicine at the Amsterdam University Medical Center, Vrije Universiteit-Cancer Center Amsterdam in Amsterdam, The Netherlands.
| | - Alette H Ruarus
- Department of Radiology and Nuclear Medicine at the Amsterdam University Medical Center, Vrije Universiteit-Cancer Center Amsterdam in Amsterdam, The Netherlands
| | - Evelien A C Schouten
- Department of Radiology and Nuclear Medicine at the Amsterdam University Medical Center, Vrije Universiteit-Cancer Center Amsterdam in Amsterdam, The Netherlands
| | - Sanne Nieuwenhuizen
- Department of Radiology and Nuclear Medicine at the Amsterdam University Medical Center, Vrije Universiteit-Cancer Center Amsterdam in Amsterdam, The Netherlands
| | - Robbert S Puijk
- Department of Radiology and Nuclear Medicine at the Amsterdam University Medical Center, Vrije Universiteit-Cancer Center Amsterdam in Amsterdam, The Netherlands
| | - Jan J J de Vries
- Department of Radiology and Nuclear Medicine at the Amsterdam University Medical Center, Vrije Universiteit-Cancer Center Amsterdam in Amsterdam, The Netherlands
| | - Martijn R Meijerink
- Department of Radiology and Nuclear Medicine at the Amsterdam University Medical Center, Vrije Universiteit-Cancer Center Amsterdam in Amsterdam, The Netherlands
| | - Hester J Scheffer
- Department of Radiology and Nuclear Medicine at the Amsterdam University Medical Center, Vrije Universiteit-Cancer Center Amsterdam in Amsterdam, The Netherlands
| |
Collapse
|
45
|
He C, Wang J, Zhang Y, Cai Z, Lin X, Li S. Comparison of combination therapies in the management of locally advanced pancreatic cancer: Induction chemotherapy followed by irreversible electroporation vs radiofrequency ablation. Cancer Med 2020; 9:4699-4710. [PMID: 32410380 PMCID: PMC7333834 DOI: 10.1002/cam4.3119] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/23/2020] [Accepted: 04/23/2020] [Indexed: 12/17/2022] Open
Abstract
Background Locally advanced pancreatic cancer (LAPC) remains a challenge for current treatments. Local destructive therapies, such as irreversible electroporation (IRE) and radiofrequency ablation (RFA), were used more and more frequently in the treatment of LAPC. Objective This study aimed to compare the efficacy of IRE with RFA in patients with LAPC. Methods From August 2015 to August 2017, 58 LAPC patients after IRE or RFA therapy, which was performed through open approach, were retrospectively reviewed. The survival outcomes after IRE (36 patients) and RFA (18 patients) were compared after propensity score matching (PSM) analysis. Results Before PSM analysis, IRE after the induction chemotherapy resulted in significant higher overall survival (OS) rates and progression‐free survival (PFS) rates to RFA (2‐year OS, 53.5% vs 30.8%, P = .013; 2‐year PFS, 28.4% vs 12.1%, P = .043). After PSM analysis, compared with RFA, the survival benefit of IRE was even more obvious, (2‐year OS, 53.5% vs 27.0%, P = .010; 2‐year PFS, 28.4% vs 6.4%, P = .018). For patients with tumor larger than 4 cm, IRE resulted in comparable OS and PFS between RFA and IRE while IRE also achieved better long‐term OS to RFA for those with tumor smaller than 4 cm. Multivariate analysis illustrated that IRE was a favorable prognostic factor in terms of OS and PFS in patients with LAPC. Conclusions IRE after induction chemotherapy is superior to RFA after induction chemotherapy for treating LAPC patients while these two therapies have comparable efficacy for tumors which were larger than 4 cm.
Collapse
Affiliation(s)
- Chaobin He
- Department of Pancreatobiliary Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jun Wang
- Department of Pancreatobiliary Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yu Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, P.R. China
| | - Zhiyuan Cai
- Department of Pancreatobiliary Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaojun Lin
- Department of Pancreatobiliary Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shengping Li
- Department of Pancreatobiliary Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
46
|
Geboers B, Scheffer HJ, Graybill PM, Ruarus AH, Nieuwenhuizen S, Puijk RS, van den Tol PM, Davalos RV, Rubinsky B, de Gruijl TD, Miklavčič D, Meijerink MR. High-Voltage Electrical Pulses in Oncology: Irreversible Electroporation, Electrochemotherapy, Gene Electrotransfer, Electrofusion, and Electroimmunotherapy. Radiology 2020; 295:254-272. [PMID: 32208094 DOI: 10.1148/radiol.2020192190] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
This review summarizes the use of high-voltage electrical pulses (HVEPs) in clinical oncology to treat solid tumors with irreversible electroporation (IRE) and electrochemotherapy (ECT). HVEPs increase the membrane permeability of cells, a phenomenon known as electroporation. Unlike alternative ablative therapies, electroporation does not affect the structural integrity of surrounding tissue, thereby enabling tumors in the vicinity of vital structures to be treated. IRE uses HVEPs to cause cell death by inducing membrane disruption, and it is primarily used as a radical ablative therapy in the treatment of soft-tissue tumors in the liver, kidney, prostate, and pancreas. ECT uses HVEPs to transiently increase membrane permeability, enhancing cellular cytotoxic drug uptake in tumors. IRE and ECT show immunogenic effects that could be augmented when combined with immunomodulatory drugs, a combination therapy the authors term electroimmunotherapy. Additional electroporation-based technologies that may reach clinical importance, such as gene electrotransfer, electrofusion, and electroimmunotherapy, are concisely reviewed. HVEPs represent a substantial advancement in cancer research, and continued improvement and implementation of these presented technologies will require close collaboration between engineers, interventional radiologists, medical oncologists, and immuno-oncologists.
Collapse
Affiliation(s)
- Bart Geboers
- From the Departments of Radiology and Nuclear Medicine (B.G., H.J.S., A.H.R., S.N., R.S.P., M.R.M.), Surgery (P.M.v.d.T.), and Medical Oncology (T.D.d.G.), Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands; Bioelectromechanical Systems Laboratory, Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest University, Blacksburg, Va (P.M.G., R.V.D.); Department of Bioengineering and Department of Mechanical Engineering, University of California, Berkeley, Berkeley, Calif (B.R.); and Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia (D.M.)
| | - Hester J Scheffer
- From the Departments of Radiology and Nuclear Medicine (B.G., H.J.S., A.H.R., S.N., R.S.P., M.R.M.), Surgery (P.M.v.d.T.), and Medical Oncology (T.D.d.G.), Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands; Bioelectromechanical Systems Laboratory, Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest University, Blacksburg, Va (P.M.G., R.V.D.); Department of Bioengineering and Department of Mechanical Engineering, University of California, Berkeley, Berkeley, Calif (B.R.); and Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia (D.M.)
| | - Philip M Graybill
- From the Departments of Radiology and Nuclear Medicine (B.G., H.J.S., A.H.R., S.N., R.S.P., M.R.M.), Surgery (P.M.v.d.T.), and Medical Oncology (T.D.d.G.), Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands; Bioelectromechanical Systems Laboratory, Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest University, Blacksburg, Va (P.M.G., R.V.D.); Department of Bioengineering and Department of Mechanical Engineering, University of California, Berkeley, Berkeley, Calif (B.R.); and Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia (D.M.)
| | - Alette H Ruarus
- From the Departments of Radiology and Nuclear Medicine (B.G., H.J.S., A.H.R., S.N., R.S.P., M.R.M.), Surgery (P.M.v.d.T.), and Medical Oncology (T.D.d.G.), Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands; Bioelectromechanical Systems Laboratory, Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest University, Blacksburg, Va (P.M.G., R.V.D.); Department of Bioengineering and Department of Mechanical Engineering, University of California, Berkeley, Berkeley, Calif (B.R.); and Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia (D.M.)
| | - Sanne Nieuwenhuizen
- From the Departments of Radiology and Nuclear Medicine (B.G., H.J.S., A.H.R., S.N., R.S.P., M.R.M.), Surgery (P.M.v.d.T.), and Medical Oncology (T.D.d.G.), Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands; Bioelectromechanical Systems Laboratory, Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest University, Blacksburg, Va (P.M.G., R.V.D.); Department of Bioengineering and Department of Mechanical Engineering, University of California, Berkeley, Berkeley, Calif (B.R.); and Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia (D.M.)
| | - Robbert S Puijk
- From the Departments of Radiology and Nuclear Medicine (B.G., H.J.S., A.H.R., S.N., R.S.P., M.R.M.), Surgery (P.M.v.d.T.), and Medical Oncology (T.D.d.G.), Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands; Bioelectromechanical Systems Laboratory, Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest University, Blacksburg, Va (P.M.G., R.V.D.); Department of Bioengineering and Department of Mechanical Engineering, University of California, Berkeley, Berkeley, Calif (B.R.); and Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia (D.M.)
| | - Petrousjka M van den Tol
- From the Departments of Radiology and Nuclear Medicine (B.G., H.J.S., A.H.R., S.N., R.S.P., M.R.M.), Surgery (P.M.v.d.T.), and Medical Oncology (T.D.d.G.), Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands; Bioelectromechanical Systems Laboratory, Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest University, Blacksburg, Va (P.M.G., R.V.D.); Department of Bioengineering and Department of Mechanical Engineering, University of California, Berkeley, Berkeley, Calif (B.R.); and Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia (D.M.)
| | - Rafael V Davalos
- From the Departments of Radiology and Nuclear Medicine (B.G., H.J.S., A.H.R., S.N., R.S.P., M.R.M.), Surgery (P.M.v.d.T.), and Medical Oncology (T.D.d.G.), Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands; Bioelectromechanical Systems Laboratory, Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest University, Blacksburg, Va (P.M.G., R.V.D.); Department of Bioengineering and Department of Mechanical Engineering, University of California, Berkeley, Berkeley, Calif (B.R.); and Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia (D.M.)
| | - Boris Rubinsky
- From the Departments of Radiology and Nuclear Medicine (B.G., H.J.S., A.H.R., S.N., R.S.P., M.R.M.), Surgery (P.M.v.d.T.), and Medical Oncology (T.D.d.G.), Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands; Bioelectromechanical Systems Laboratory, Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest University, Blacksburg, Va (P.M.G., R.V.D.); Department of Bioengineering and Department of Mechanical Engineering, University of California, Berkeley, Berkeley, Calif (B.R.); and Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia (D.M.)
| | - Tanja D de Gruijl
- From the Departments of Radiology and Nuclear Medicine (B.G., H.J.S., A.H.R., S.N., R.S.P., M.R.M.), Surgery (P.M.v.d.T.), and Medical Oncology (T.D.d.G.), Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands; Bioelectromechanical Systems Laboratory, Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest University, Blacksburg, Va (P.M.G., R.V.D.); Department of Bioengineering and Department of Mechanical Engineering, University of California, Berkeley, Berkeley, Calif (B.R.); and Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia (D.M.)
| | - Damijan Miklavčič
- From the Departments of Radiology and Nuclear Medicine (B.G., H.J.S., A.H.R., S.N., R.S.P., M.R.M.), Surgery (P.M.v.d.T.), and Medical Oncology (T.D.d.G.), Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands; Bioelectromechanical Systems Laboratory, Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest University, Blacksburg, Va (P.M.G., R.V.D.); Department of Bioengineering and Department of Mechanical Engineering, University of California, Berkeley, Berkeley, Calif (B.R.); and Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia (D.M.)
| | - Martijn R Meijerink
- From the Departments of Radiology and Nuclear Medicine (B.G., H.J.S., A.H.R., S.N., R.S.P., M.R.M.), Surgery (P.M.v.d.T.), and Medical Oncology (T.D.d.G.), Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands; Bioelectromechanical Systems Laboratory, Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest University, Blacksburg, Va (P.M.G., R.V.D.); Department of Bioengineering and Department of Mechanical Engineering, University of California, Berkeley, Berkeley, Calif (B.R.); and Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia (D.M.)
| |
Collapse
|
47
|
Real-time prediction of patient immune cell modulation during irreversible electroporation therapy. Sci Rep 2019; 9:17739. [PMID: 31780711 PMCID: PMC6882846 DOI: 10.1038/s41598-019-53974-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 11/05/2019] [Indexed: 12/18/2022] Open
Abstract
Immunotherapies have demonstrated limited efficacy in pancreatic ductal adenocarcinoma (PDAC) patients despite their success in treating other tumor types. This limitation is largely due to the relatively immunosuppressive environment surrounding the tumor. A focal ablative technique called irreversible electroporation (IRE) has been shown to modulate this environment, enhancing the efficacy of immunotherapy. One enhancing factor related to improved prognosis is a decrease in regulatory T cells (Treg). This decrease has been previously unpredictable for clinicians using IRE, who currently have limited real-time metrics for determining the activation of the patient’s immune response. Here, we report that larger overall changes in output current are correlated with larger decreases in T cell populations 24 hours post-treatment. This result suggests that clinicians can make real-time decisions regarding optimal follow-up therapy based on the range of output current delivered during treatment. This capability could maximize the immunomodulating effect of IRE in synergy with follow-up immunotherapy. Additionally, these results suggest that feedback from a preliminary IRE treatment of the local tumor may help inform clinicians regarding the timing and choice of subsequent therapies, such as resection, immunotherapy, chemotherapy, or follow-up thermal or non-thermal ablation.
Collapse
|
48
|
Oncolysis without viruses — inducing systemic anticancer immune responses with local therapies. Nat Rev Clin Oncol 2019; 17:49-64. [PMID: 31595049 DOI: 10.1038/s41571-019-0272-7] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2019] [Indexed: 02/06/2023]
|
49
|
Scheffer HJ, Stam AGM, Geboers B, Vroomen LGPH, Ruarus A, de Bruijn B, van den Tol MP, Kazemier G, Meijerink MR, de Gruijl TD. Irreversible electroporation of locally advanced pancreatic cancer transiently alleviates immune suppression and creates a window for antitumor T cell activation. Oncoimmunology 2019; 8:1652532. [PMID: 31646081 PMCID: PMC6791414 DOI: 10.1080/2162402x.2019.1652532] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 07/26/2019] [Accepted: 07/31/2019] [Indexed: 12/18/2022] Open
Abstract
Purpose: Local tumor ablation through irreversible electroporation (IRE) may offer a novel therapeutic option for locally advanced pancreatic cancer (LAPC). It may also serve as a means of in vivo vaccination. To obtain evidence of the induction of systemic antitumor immunity following local IRE-mediated ablation, we performed an explorative immune monitoring study. Methods: In ten patients enrolled in a clinical trial exploring the safety, feasibility, and efficacy of percutaneous image-guided IRE in LAPC, we determined the frequency and activation state of lymphocytic and myeloid subsets in pre- and post-treatment peripheral blood samples using flow cytometry. Tumor-specific systemic T cell responses to the pancreatic cancer associated antigen Wilms Tumor (WT)1 were determined after in vitro stimulation in an interferon-y enzyme-linked immunospot assay (Elispot), at baseline and at 2 weeks and 3 months after IRE. Results: Our data showed a transient decrease in systemic regulatory T cells (Treg) and a simultaneous transient increase in activated PD-1+ T cells, consistent with the temporary reduction of tumor-related immune suppression after the IRE procedure. Accordingly, we found post-IRE boosting of a pre-existing WT1 specific T cell response in two out of three patients as well as the de novo induction of these responses in another two patients. There was a trend for these WT1 T cell responses to be related to longer overall survival (p = .055). Conclusions: These findings are consistent with a systemic and tumor-specific immune stimulatory effect of IRE and support the combination of percutaneous IRE with therapeutic immune modulation.
Collapse
Affiliation(s)
- Hester J Scheffer
- Departments of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit-Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Anita G M Stam
- Departments of Medical Oncology, Amsterdam University Medical Center, Vrije Universiteit-Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Bart Geboers
- Departments of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit-Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Laurien G P H Vroomen
- Departments of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit-Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Alette Ruarus
- Departments of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit-Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Beaunelle de Bruijn
- Departments of Medical Oncology, Amsterdam University Medical Center, Vrije Universiteit-Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - M Petrousjka van den Tol
- Departments of Surgery, Amsterdam University Medical Center, Vrije Universiteit-Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Geert Kazemier
- Departments of Surgery, Amsterdam University Medical Center, Vrije Universiteit-Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Martijn R Meijerink
- Departments of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit-Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Tanja D de Gruijl
- Departments of Medical Oncology, Amsterdam University Medical Center, Vrije Universiteit-Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
50
|
Immunomodulatory Effect after Irreversible Electroporation in Patients with Locally Advanced Pancreatic Cancer. JOURNAL OF ONCOLOGY 2019; 2019:9346017. [PMID: 31214261 PMCID: PMC6535893 DOI: 10.1155/2019/9346017] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 12/24/2022]
Abstract
Purpose Irreversible electroporation (IRE) has been demonstrated to be a safe and effective method for locally advanced pancreatic cancer (LAPC). The aim of this study was to evaluate the immunomodulatory effect after IRE and to evaluate the prognostic value of variations of the immune parameters in LAPC patients after IRE. Methods Peripheral blood samples of 34 patients were obtained preoperatively and on the third day (D3) and seventh day (D7) after IRE, respectively. The phenotypes of lymphocytes were analyzed by flow cytometry, and dynamic changes of serum levels of cytokines, complement, and immunoglobulin were assayed by enzyme-linked immunosorbent assay. Receiver operating characteristic (ROC) curve and concordance index (C-index) were used to compare the survival predictive ability. Results There was a transitory decrease followed by a steady increase for CD4+ T cell, CD8+ T cell, NK cell, IL-2, C3, C4, and IgG while a reverse trend was detected for Treg cell, IL-6, and IL10 after IRE. The alteration of CD8+ T cell between D3 and D7 was identified as a prognostic factor for both overall survival (OS) and progression-free survival (PFS). The values of ROC curve (AUC) and C-indexes of the alteration of CD8+ T cell for OS and PFS were 0.816 and 0.773 and 0.816 and 0.639, respectively, which were larger than those of other immune or inflammation-based indexes. Conclusions This study presented the first evidence of IRE-based immunomodulatory in patients with LAPC. The alteration of CD8+ T cell between D3 and D7 showed relatively good performance and could be used as an effective tool for prognostic evaluation for LAPC patients after IRE.
Collapse
|