1
|
Zhang Z, Yu G, Eresen A, Chen Z, Yu Z, Yaghmai V, Zhang Z. Dendritic cell vaccination combined with irreversible electroporation for treating pancreatic cancer-a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2024; 12:77. [PMID: 39118942 PMCID: PMC11304422 DOI: 10.21037/atm-23-1882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 02/25/2024] [Indexed: 08/10/2024]
Abstract
Background and Objective Pancreatic ductal adenocarcinoma (PDAC) is 3rd most lethal cancer in the USA leading to a median survival of six months and less than 5% 5-year overall survival (OS). As the only potentially curative treatment, surgical resection is not suitable for up to 90% of the patients with PDAC due to late diagnosis. Highly fibrotic PDAC with an immunosuppressive tumor microenvironment restricts cytotoxic T lymphocyte (CTL) infiltration and functions causing limited success with systemic therapies like dendritic cell (DC)-based immunotherapy. In this study, we investigated the potential benefits of irreversible electroporation (IRE) ablation therapy in combination with DC vaccine therapy against PDAC. Methods We performed a literature search to identify studies focused on DC vaccine therapy and IRE ablation to boost therapeutic response against PDAC indexed in PubMed, Web of Science, and Scopus until February 20th, 2023. Key Content and Findings IRE ablation destructs tumor structure while preserving extracellular matrix and blood vessels facilitating local inflammation. The studies demonstrated IRE ablation reduces tumor fibrosis and promotes CTL tumor infiltration to PDAC tumors in addition to boosting immune response in rodent models. The administration of the DC vaccine following IRE ablation synergistically enhances therapeutic response and extends OS rates compared to the use of DC vaccination or IRE alone. Moreover, the implementation of data-driven approaches further allows dynamic and longitudinal monitoring of therapeutic response and OS following IRE plus DC vaccine immunoablation. Conclusions The combination of IRE ablation and DC vaccine immunotherapy is a potent strategy to enhance the therapeutic outcomes in patients with PDAC.
Collapse
Affiliation(s)
- Zigeng Zhang
- Department of Radiological Sciences, University of California Irvine, Irvine, CA, USA
| | - Guangbo Yu
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Aydin Eresen
- Department of Radiological Sciences, University of California Irvine, Irvine, CA, USA
| | - Zhilin Chen
- Department of Human Biology and Business Administration, University of Southern California, Los Angeles, CA, USA
| | - Zeyang Yu
- Information School, University of Washington, Seattle, WA, USA
| | - Vahid Yaghmai
- Department of Radiological Sciences, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | - Zhuoli Zhang
- Department of Radiological Sciences, University of California Irvine, Irvine, CA, USA
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
2
|
Zhu X, Shi Z, Mao Y, Lächelt U, Huang R. Cell Membrane Perforation: Patterns, Mechanisms and Functions. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310605. [PMID: 38344881 DOI: 10.1002/smll.202310605] [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: 11/18/2023] [Revised: 12/21/2023] [Indexed: 02/21/2024]
Abstract
Cell membrane is crucial for the cellular activities, and any disruption to it may affect the cells. It is demonstrated that cell membrane perforation is associated with some biological processes like programmed cell death (PCD) and infection of pathogens. Specific developments make it a promising technique to perforate the cell membrane controllably and precisely. The pores on the cell membrane provide direct pathways for the entry and exit of substances, and can also cause cell death, which means reasonable utilization of cell membrane perforation is able to assist intracellular delivery, eliminate diseased or cancerous cells, and bring about other benefits. This review classifies the patterns of cell membrane perforation based on the mechanisms into 1) physical patterns, 2) biological patterns, and 3) chemical patterns, introduces the characterization methods and then summarizes the functions according to the characteristics of reversible and irreversible pores, with the aim of providing a comprehensive summary of the knowledge related to cell membrane perforation and enlightening broad applications in biomedical science.
Collapse
Affiliation(s)
- Xinran Zhu
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Huashan Hospital, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Zhifeng Shi
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 201203, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 201203, China
| | - Ulrich Lächelt
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, 1090, Austria
| | - Rongqin Huang
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Huashan Hospital, School of Pharmacy, Fudan University, Shanghai, 201203, China
| |
Collapse
|
3
|
Roy Barman S, Jhunjhunwala S. Electrical Stimulation for Immunomodulation. ACS OMEGA 2024; 9:52-66. [PMID: 38222551 PMCID: PMC10785302 DOI: 10.1021/acsomega.3c06696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/29/2023] [Accepted: 12/04/2023] [Indexed: 01/16/2024]
Abstract
The immune system plays a key role in the development and progression of numerous diseases such as chronic wounds, autoimmune diseases, and various forms of cancer. Hence, controlling the behavior of immune cells has emerged as a promising approach for treating these diseases. Current modalities for immunomodulation focus on chemical based approaches, which while effective have the limitations of nonspecific systemic side effects or requiring invasive delivery approaches to reduce the systemic side effects. Recent advances have unraveled the significance of electrical stimulation as an attractive noninvasive approach to modulate immune cell phenotype and activity. This review provides insights on electrical stimulation strategies employed for regulating the behavior of macrophages, T and B cells, and neutrophils. For obtaining a better understanding, two major types of electrical stimulation sources, conventional and self-powered sources, that have been used for immunomodulation are extensively discussed. Next, the strategies of electrical stimulation that may be applied to cells in vitro and in vivo are discussed, with a focus on conventional and stimuli-responsive self-powered sources. A description of how these strategies influence the polarization, phagocytosis, migration, and differentiation of immune cells is also provided. Finally, recent developments in the use of highly localized and efficient platforms for electrical stimulation based immunomodulation are also highlighted.
Collapse
Affiliation(s)
- Snigdha Roy Barman
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India 560012
| | | |
Collapse
|
4
|
Salameh ZS, Aycock KN, Alinezhadbalalami N, Imran KM, McKillop IH, Allen IC, Davalos RV. Harnessing the Electrochemical Effects of Electroporation-Based Therapies to Enhance Anti-tumor Immune Responses. Ann Biomed Eng 2024; 52:48-56. [PMID: 37989902 PMCID: PMC10781785 DOI: 10.1007/s10439-023-03403-x] [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: 05/25/2023] [Accepted: 10/31/2023] [Indexed: 11/23/2023]
Abstract
This study introduces a new method of targeting acidosis (low pH) within the tumor microenvironment (TME) through the use of cathodic electrochemical reactions (CER). Low pH is oncogenic by supporting immunosuppression. Electrochemical reactions create local pH effects when a current passes through an electrolytic substrate such as biological tissue. Electrolysis has been used with electroporation (destabilization of the lipid bilayer via an applied electric potential) to increase cell death areas. However, the regulated increase of pH through only the cathode electrode has been ignored as a possible method to alleviate TME acidosis, which could provide substantial immunotherapeutic benefits. Here, we show through ex vivo modeling that CERs can intentionally elevate pH to an anti-tumor level and that increased alkalinity promotes activation of naïve macrophages. This study shows the potential of CERs to improve acidity within the TME and that it has the potential to be paired with existing electric field-based cancer therapies or as a stand-alone therapy.
Collapse
Affiliation(s)
- Zaid S Salameh
- Department of Biomedical Engineering and Mechanics, Virginia Tech, 325 Stanger St, Blacksburg, VA, 24061, USA
| | - Kenneth N Aycock
- Department of Biomedical Engineering and Mechanics, Virginia Tech, 325 Stanger St, Blacksburg, VA, 24061, USA
| | - Nastaran Alinezhadbalalami
- Department of Biomedical Engineering and Mechanics, Virginia Tech, 325 Stanger St, Blacksburg, VA, 24061, USA
| | - Khan Mohammad Imran
- Department of Biomedical Sciences and Pathobiology, VA-MD College of Veterinary Medicine, Virginia Tech, 205 Duck Pond Drive, Blacksburg, VA, 24061, USA
| | - Iain H McKillop
- Department of Surgery, Atrium Health Wake Forest Baptist Medical Center, 1000 Blythe Blvd, Charlotte, NC, 28203, USA
| | - Irving C Allen
- Department of Biomedical Sciences and Pathobiology, VA-MD College of Veterinary Medicine, Virginia Tech, 205 Duck Pond Drive, Blacksburg, VA, 24061, USA
| | - Rafael V Davalos
- Department of Biomedical Engineering and Mechanics, Virginia Tech, 325 Stanger St, Blacksburg, VA, 24061, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech - Emory, 313 Ferst Dr, Atlanta, GA, 30308, USA.
| |
Collapse
|
5
|
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: 0] [Impact Index Per Article: 0] [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
|
6
|
Woeste MR, Shrestha R, Geller AE, Li S, Montoya-Durango D, Ding C, Hu X, Li H, Puckett A, Mitchell RA, Hayat T, Tan M, Li Y, McMasters KM, Martin RCG, Yan J. Irreversible electroporation augments β-glucan induced trained innate immunity for the treatment of pancreatic ductal adenocarcinoma. J Immunother Cancer 2023; 11:e006221. [PMID: 37072351 PMCID: PMC10124260 DOI: 10.1136/jitc-2022-006221] [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] [Accepted: 03/23/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND Pancreatic cancer (PC) is a challenging diagnosis that is yet to benefit from the advancements in immuno-oncologic treatments. Irreversible electroporation (IRE), a non-thermal method of tumor ablation, is used in treatment of select patients with locally-advanced unresectable PC and has potentiated the effect of certain immunotherapies. Yeast-derived particulate β-glucan induces trained innate immunity and successfully reduces murine PC tumor burden. This study tests the hypothesis that IRE may augment β-glucan induced trained immunity in the treatment of PC. METHODS β-Glucan-trained pancreatic myeloid cells were evaluated ex vivo for trained responses and antitumor function after exposure to ablated and unablated tumor-conditioned media. β-Glucan and IRE combination therapy was tested in an orthotopic murine PC model in wild-type and Rag-/- mice. Tumor immune phenotypes were assessed by flow cytometry. Effect of oral β-glucan in the murine pancreas was evaluated and used in combination with IRE to treat PC. The peripheral blood of patients with PC taking oral β-glucan after IRE was evaluated by mass cytometry. RESULTS IRE-ablated tumor cells elicited a potent trained response ex vivo and augmented antitumor functionality. In vivo, β-glucan in combination with IRE reduced local and distant tumor burden prolonging survival in a murine orthotopic PC model. This combination augmented immune cell infiltration to the PC tumor microenvironment and potentiated the trained response from tumor-infiltrating myeloid cells. The antitumor effect of this dual therapy occurred independent of the adaptive immune response. Further, orally administered β-glucan was identified as an alternative route to induce trained immunity in the murine pancreas and prolonged PC survival in combination with IRE. β-Glucan in vitro treatment also induced trained immunity in peripheral blood monocytes obtained from patients with treatment-naïve PC. Finally, orally administered β-glucan was found to significantly alter the innate cell landscape within the peripheral blood of five patients with stage III locally-advanced PC who had undergone IRE. CONCLUSIONS These data highlight a relevant and novel application of trained immunity within the setting of surgical ablation that may stand to benefit patients with PC.
Collapse
Affiliation(s)
- Matthew R Woeste
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Division of Immunotherapy, The Hiram C. Polk Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Rejeena Shrestha
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Division of Immunotherapy, The Hiram C. Polk Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Anne E Geller
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Division of Immunotherapy, The Hiram C. Polk Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Shu Li
- Division of Immunotherapy, The Hiram C. Polk Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Diego Montoya-Durango
- Division of Immunotherapy, The Hiram C. Polk Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Chuanlin Ding
- Division of Immunotherapy, The Hiram C. Polk Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Xiaoling Hu
- Division of Immunotherapy, The Hiram C. Polk Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Hong Li
- Functional Immunomics Core, Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Aaron Puckett
- Functional Immunomics Core, Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Robert A Mitchell
- Division of Immunotherapy, The Hiram C. Polk Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Traci Hayat
- Division of Surgical Oncology, The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Min Tan
- Division of Surgical Oncology, The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Yan Li
- Division of Surgical Oncology, The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Kelly M McMasters
- Division of Surgical Oncology, The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Robert C G Martin
- Division of Surgical Oncology, The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Jun Yan
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Division of Immunotherapy, The Hiram C. Polk Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
7
|
Szlasa W, Janicka N, Sauer N, Michel O, Nowak B, Saczko J, Kulbacka J. Chemotherapy and Physical Therapeutics Modulate Antigens on Cancer Cells. Front Immunol 2022; 13:889950. [PMID: 35874714 PMCID: PMC9299262 DOI: 10.3389/fimmu.2022.889950] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/06/2022] [Indexed: 12/29/2022] Open
Abstract
Cancer cells possess specific properties, such as multidrug resistance or unlimited proliferation potential, due to the presence of specific proteins on their cell membranes. The release of proliferation-related proteins from the membrane can evoke a loss of adaptive ability in cancer cells and thus enhance the effects of anticancer therapy. The upregulation of cancer-specific membrane antigens results in a better outcome of immunotherapy. Moreover, cytotoxic T-cells may also become more effective when stimulated ex-vivo toward the anticancer response. Therefore, the modulation of membrane proteins may serve as an interesting attempt in anticancer therapy. The presence of membrane antigens relies on various physical factors such as temperature, exposure to radiation, or drugs. Therefore, changing the tumor microenvironment conditions may lead to cancer cells becoming sensitized to subsequent therapy. This paper focuses on the therapeutic approaches modulating membrane antigens and enzymes in anticancer therapy. It aims to analyze the possible methods for modulating the antigens, such as pharmacological treatment, electric field treatment, photodynamic reaction, treatment with magnetic field or X-ray radiation. Besides, an overview of the effects of chemotherapy and immunotherapy on the immunophenotype of cancer cells is presented. Finally, the authors review the clinical trials that involved the modulation of cell immunophenotype in anticancer therapy.
Collapse
Affiliation(s)
- Wojciech Szlasa
- Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Natalia Janicka
- Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Natalia Sauer
- Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Olga Michel
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Bernadetta Nowak
- Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
8
|
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: 11.0] [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
|
9
|
Imran KM, Nagai-Singer MA, Brock RM, Alinezhadbalalami N, Davalos RV, Allen IC. Exploration of Novel Pathways Underlying Irreversible Electroporation Induced Anti-Tumor Immunity in Pancreatic Cancer. Front Oncol 2022; 12:853779. [PMID: 35372046 PMCID: PMC8972192 DOI: 10.3389/fonc.2022.853779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/28/2022] [Indexed: 12/04/2022] Open
Abstract
Advancements in medical sciences and technologies have significantly improved the survival of many cancers; however, pancreatic cancer remains a deadly diagnosis. This malignancy is often diagnosed late in the disease when metastases have already occurred. Additionally, the location of the pancreas near vital organs limits surgical candidacy, the tumor's immunosuppressive environment limits immunotherapy success, and it is highly resistant to radiation and chemotherapy. Hence, clinicians and patients alike need a treatment paradigm that reduces primary tumor burden, activates systemic anti-tumor immunity, and reverses the local immunosuppressive microenvironment to eventually clear distant metastases. Irreversible electroporation (IRE), a novel non-thermal tumor ablation technique, applies high-voltage ultra-short pulses to permeabilize targeted cell membranes and induce cell death. Progression with IRE technology and an array of research studies have shown that beyond tumor debulking, IRE can induce anti-tumor immune responses possibly through tumor neo-antigen release. However, the success of IRE treatment (i.e. full ablation and tumor recurrence) is variable. We believe that IRE treatment induces IFNγ expression, which then modulates immune checkpoint molecules and thus leads to tumor recurrence. This indicates a co-therapeutic use of IRE and immune checkpoint inhibitors as a promising treatment for pancreatic cancer patients. Here, we review the well-defined and speculated pathways involved in the immunostimulatory effects of IRE treatment for pancreatic cancer, as well as the regulatory pathways that may negate these anti-tumor responses. By defining these underlying mechanisms, future studies may identify improvements to systemic immune system engagement following local tumor ablation with IRE and beyond.
Collapse
Affiliation(s)
- Khan Mohammad Imran
- Graduate Program in Translational Biology, Medicine and Health, Virginia Tech, Roanoke, VA, United States
| | - Margaret A. Nagai-Singer
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, United States
- Institute for Critical Technology and Applied Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Rebecca M. Brock
- Graduate Program in Translational Biology, Medicine and Health, Virginia Tech, Roanoke, VA, United States
| | - Nastaran Alinezhadbalalami
- Institute for Critical Technology and Applied Sciences, Virginia Tech, Blacksburg, VA, United States
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, United States
| | - Rafael V. Davalos
- Institute for Critical Technology and Applied Sciences, Virginia Tech, Blacksburg, VA, United States
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, United States
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA, United States
| | - Irving Coy Allen
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, United States
- Institute for Critical Technology and Applied Sciences, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
10
|
Aycock KN, Campelo SN, Davalos RV. A Comparative Modeling Study of Thermal Mitigation Strategies in Irreversible Electroporation Treatments. JOURNAL OF HEAT TRANSFER 2022; 144:031206. [PMID: 35833151 PMCID: PMC8823459 DOI: 10.1115/1.4053199] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/03/2021] [Indexed: 05/09/2023]
Abstract
Irreversible electroporation (IRE), also referred to as nonthermal pulsed field ablation (PFA), is an attractive focal ablation modality for solid tumors and cardiac tissue due to its ability to destroy aberrant cells with limited disruption of the underlying tissue architecture. Despite its nonthermal cell death mechanism, application of electrical energy results in Joule heating that, if ignored, can cause undesired thermal injury. Engineered thermal mitigation (TM) technologies including phase change materials (PCMs) and active cooling (AC) have been reported and tested as a potential means to limit thermal damage. However, several variables affect TM performance including the pulsing paradigm, electrode geometry, PCM composition, and chosen active cooling parameters, meaning direct comparisons between approaches are lacking. In this study, we developed a computational model of conventional bipolar and monopolar probes with solid, PCM-filled, or actively cooled cores to simulate clinical IRE treatments in pancreatic tissue. This approach reveals that probes with integrated PCM cores can be tuned to drastically limit thermal damage compared to existing solid probes. Furthermore, actively cooled probes provide additional control over thermal effects within the probe vicinity and can altogether abrogate thermal damage. In practice, such differences in performance must be weighed against the increased time, expense, and effort required for modified probes compared to existing solid probes.
Collapse
Affiliation(s)
- Kenneth N. Aycock
- Bioelectromechanical Systems Lab, Virginia Tech—Wake Forest School of Biomedical Engineering and Sciences, Virginia Tech Department of Biomedical Engineering and Mechanics, 320 Kelly Hall, 325 Stanger Street, Blacksburg, VA 24061
| | - Sabrina N. Campelo
- Bioelectromechanical Systems Lab, Virginia Tech—Wake Forest School of Biomedical Engineering and Sciences, Virginia Tech Department of Biomedical Engineering and Mechanics, 320 Kelly Hall, 325 Stanger Street, Blacksburg, VA 24061
| | - Rafael V. Davalos
- Bioelectromechanical Systems Lab, Virginia Tech—Wake Forest School of Biomedical Engineering and Sciences, Virginia Tech Department of Biomedical Engineering and Mechanics, 320 Kelly Hall, 325 Stanger Street, Blacksburg, VA 24061
| |
Collapse
|
11
|
Choi B, Kim DH. Multifunctional Nanocarriers-Mediated Synergistic Combination of Immune Checkpoint Inhibitor Cancer Immunotherapy and Interventional Oncology Therapy. ADVANCED NANOBIOMED RESEARCH 2021; 1:2100010. [PMID: 35663354 PMCID: PMC9162439 DOI: 10.1002/anbr.202100010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Immune checkpoint inhibitor (ICI) cancer immunotherapies are becoming one of the standard therapies for cancer patients. However, ICI cancer immunotherapy's overall response rate is still moderate and even combinational ICI cancer immunotherapies are not showing significant improvement in therapeutic outcomes. Only a subset of patients responds to the therapy due to the resistance and ignorance to the ICI cancer immunotherapy. Following immune-related adverse events (irAEs) are also limiting the whole therapeutic regimens. New approaches that can increase the immunotherapeutic efficacy and reduce systemic irAEs are required. Recently, multifunctional nanocarriers, which can extend the half-life of ICIs and modulate tumor microenvironment (TME), have shown a substantial opportunity to enhance ICI cancer immunotherapies. Interventional oncology (IO) allowing simultaneous diagnosis, immunogenic loco-regional therapeutic delivery, and real-time monitoring of the treatment efficacy have advanced to demonstrate the effective conversion of TME. The use of multifunctional nanocarriers with the IO therapies amplify the image guidance capability and immunogenic therapeutic localization for the potential combinational ICI cancer immunotherapy. This article will discuss the emerging opportunity of multifunctional nanocarriers mediated synergistic combination of ICI cancer immunotherapy and IO local therapy.
Collapse
Affiliation(s)
- Bongseo Choi
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Dong-Hyun Kim
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
- Department of Biomedical Engineering, McCormick School of Engineering, Evanston, IL 60208, USA
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL 60611, USA
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
12
|
Hendricks-Wenger A, Sereno J, Gannon J, Zeher A, Brock RM, Beitel-White N, Simon A, Davalos RV, Coutermarsh-Ott S, Vlaisavljevich E, Allen IC. Histotripsy Ablation Alters the Tumor Microenvironment and Promotes Immune System Activation in a Subcutaneous Model of Pancreatic Cancer. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:2987-3000. [PMID: 33956631 PMCID: PMC9295194 DOI: 10.1109/tuffc.2021.3078094] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Pancreatic cancer is a significant cause of cancer-related deaths in the United States with an abysmal five-year overall survival rate that is under 9%. Reasons for this mortality include the lack of late-stage treatment options and the immunosuppressive tumor microenvironment. Histotripsy is an ultrasound-guided, noninvasive, nonthermal tumor ablation therapy that mechanically lyses targeted cells. To study the effects of histotripsy on pancreatic cancer, we utilized an in vitro model of pancreatic adenocarcinoma and compared the release of potential antigens following histotripsy treatment to other ablation modalities. Histotripsy was found to release immune-stimulating molecules at magnitudes similar to other nonthermal ablation modalities and superior to thermal ablation modalities, which corresponded to increased innate immune system activation in vivo. In subsequent in vivo studies, murine Pan02 tumors were grown in mice and treated with histotripsy. Flow cytometry and rtPCR were used to determine changes in the tumor microenvironment over time compared to untreated animals. In mice with pancreatic tumors, we observed significantly increased tumor-progression-free and general survival, with increased activation of the innate immune system 24 h posttreatment and decreased tumor-associated immune cell populations within 14 days of treatment. This study demonstrates the feasibility of using histotripsy for pancreatic cancer ablation and provides mechanistic insight into the initial innate immune system activation following treatment. Further work is needed to establish the mechanisms behind the immunomodulation of the tumor microenvironment and immune effects.
Collapse
|
13
|
Alinezhadbalalami N, Graybill PM, Imran KM, Verbridge SS, Allen IC, Davalos RV. Generation of Tumor-activated T cells using electroporation. Bioelectrochemistry 2021; 142:107886. [PMID: 34303065 DOI: 10.1016/j.bioelechem.2021.107886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/30/2021] [Accepted: 07/09/2021] [Indexed: 12/26/2022]
Abstract
Expansion of cytotoxic T lymphocytes (CTLs) is a crucial step in almost all cancer immunotherapeutic methods. Current techniques for expansion of tumor-reactive CTLs present major limitations. This study introduces a novel method to effectively produce and expand tumor-activated CTLs using high-voltage pulsed electric fields. We hypothesize that utilizing high-voltage pulsed electric fields may be an ideal method to activate and expand CTLs due to their non-thermal celldeath mechanism. Tumor cells were subjected to high-frequency irreversible electroporation (HFIRE) with various electric field magnitudes (1250, 2500 V/cm) and pulse widths (1, 5, and 10 µs), or irreversible electroporation (IRE) at 1250 V/cm. The treated tumor cells were subsequently cocultured with CD4+ and CD8+ T cells along with antigen-presenting cells. We show that tumor-activated CTLs can be produced and expanded when exposed to treated tumor cells. Our results suggest that CTLs are more effectively expanded when pulsed with HFIRE conditions that induce significant cell death (longer pulse widths and higher voltages). Activated CD8+ T cells demonstrate cytotoxicity to untreated tumor cells suggesting effector function of the activated CTLs. The activated CTLs produced with our technique could be used for clinical applications with the goal of targeting and eliminating the tumor.
Collapse
Affiliation(s)
- Nastaran Alinezhadbalalami
- Department of Biomedical Engineering and Mechanics, Virginia Tech, 325 Stanger Street, Blacksburg, VA 24061, USA; Institute for Critical Technology and Applied Sciences, Virginia Tech, Kelly Hall, Blacksburg, VA 24061, USA.
| | - Philip M Graybill
- Department of Mechanical Engineering, Virginia Tech, Goodwin Hall, 635 Prices Fork Road, Blacksburg, VA 24061, USA; Institute for Critical Technology and Applied Sciences, Virginia Tech, Kelly Hall, Blacksburg, VA 24061, USA.
| | - Khan Mohammad Imran
- Graduate Program in Translational Biology, Medicine and Health, Virginia Tech, 1 Riverside Circle, Roanoke, VA 24016, USA; Institute for Critical Technology and Applied Sciences, Virginia Tech, Kelly Hall, Blacksburg, VA 24061, USA.
| | - Scott S Verbridge
- Department of Biomedical Engineering and Mechanics, Virginia Tech, 325 Stanger Street, Blacksburg, VA 24061, USA; Institute for Critical Technology and Applied Sciences, Virginia Tech, Kelly Hall, Blacksburg, VA 24061, USA.
| | - Irving C Allen
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, 205 Duck Pond Drive, Blacksburg, VA 24061, USA; Institute for Critical Technology and Applied Sciences, Virginia Tech, Kelly Hall, Blacksburg, VA 24061, USA.
| | - Rafael V Davalos
- Department of Biomedical Engineering and Mechanics, Virginia Tech, 325 Stanger Street, Blacksburg, VA 24061, USA; Department of Mechanical Engineering, Virginia Tech, Goodwin Hall, 635 Prices Fork Road, Blacksburg, VA 24061, USA; Institute for Critical Technology and Applied Sciences, Virginia Tech, Kelly Hall, Blacksburg, VA 24061, USA.
| |
Collapse
|
14
|
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: 35] [Impact Index Per Article: 11.7] [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
|
15
|
Lorenzo MF, Bhonsle SP, Arena CB, Davalos RV. Rapid Impedance Spectroscopy for Monitoring Tissue Impedance, Temperature, and Treatment Outcome During Electroporation-Based Therapies. IEEE Trans Biomed Eng 2021; 68:1536-1546. [PMID: 33156779 PMCID: PMC8127872 DOI: 10.1109/tbme.2020.3036535] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Electroporation-based therapies (EBTs) employ high voltage pulsed electric fields (PEFs) to permeabilize tumor tissue; this results in changes in electrical properties detectable using electrical impedance spectroscopy (EIS). Currently, commercial potentiostats for EIS are limited by impedance spectrum acquisition time ( ∼ 10 s); this timeframe is much larger than pulse periods used with EBTs ( ∼ 1 s). In this study, we utilize rapid EIS techniques to develop a methodology for characterizing electroporation (EP) and thermal effects associated with high-frequency irreversible EP (H-FIRE) in real-time by monitoring inter-burst impedance changes. METHODS A charge-balanced, bipolar rectangular chirp signal is proposed for rapid EIS. Validation of rapid EIS measurements against a commercial potentiostat was conducted in potato tissue using flat-plate electrodes and thereafter for the measurement of impedance changes throughout IRE treatment. Flat-plate electrodes were then utilized to uniformly heat potato tissue; throughout high-voltage H-FIRE treatment, low-voltage inter-burst impedance measurements were used to continually monitor impedance change and to identify a frequency at which thermal effects are delineated from EP effects. RESULTS Inter-burst impedance measurements (1.8 kHz - 4.93 MHz) were accomplished at 216 discrete frequencies. Impedance measurements at frequencies above ∼ 1 MHz served to delineate thermal and EP effects in measured impedance. CONCLUSION We demonstrate rapid-capture ( 1 s) EIS which enables monitoring of inter-burst impedance in real-time. For the first time, we show impedance analysis at high frequencies can delineate thermal effects from EP effects in measured impedance. SIGNIFICANCE The proposed waveform demonstrates the potential to perform inter-burst EIS using PEFs compatible with existing pulse generator topologies.
Collapse
|
16
|
Wang Z, Lu J, Huang W, Wu Z, Gong J, Wang Q, Liu Q, Wang C, Zhu Y, Ding X, Wang Z. A retrospective study of CT-guided percutaneous irreversible electroporation (IRE) ablation: clinical efficacy and safety. BMC Cancer 2021; 21:124. [PMID: 33546635 PMCID: PMC7866876 DOI: 10.1186/s12885-021-07820-w] [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: 10/02/2020] [Accepted: 01/19/2021] [Indexed: 12/31/2022] Open
Abstract
Background To evaluate the clinical efficacy and safety of ablating renal cell carcinoma (RCC) by irreversible electroporation (IRE). Methods Fifteen patients (19 lesions) with RCC who underwent IRE were retrospectively reviewed. Seven patients had solitary kidneys. Two lesions were located in the renal hilus. One patient had chronic renal insufficiency. Percutaneous biopsy for histopathology was performed. The best puncture path plan was evaluated before CT-guided IRE. The estimated glomerular filtration rate (eGFR) was compared vs baseline at 1–2 months after the ablation. Contrast-enhanced computed tomography imaging changes were evaluated immediately after IRE. Contrast-enhanced computed tomography/magnetic resonance was performed 1 month, 3 months, 6 months, 12 months and every year thereafter. The complications after treatment were also reviewed. Results The success rate of the procedure was 100%. The median tumor size was 2.4 (IQR 1.3–2.9) cm, with an median score of 6 (IQR 5.5–8) per R.E.N.A.L. criteria (radius, exophytic/endophytic, nearness to collecting system or sinus, anterior/posterior, and location relative to polar lines). Two cases (3 lesions) were punctured through the liver. In other cases, puncture was performed through the perirenal space. There were no severecomplications in interventional therapy. Transient gross hematuria occurred in 2 patients (centrally located). Self-limiting perinephric hematomas occurred in 1 patient. Needle puncture path metastasis was found in 1 patient 2.5 years after IRE. The subcutaneous metastasis was surgically removed, and there was no evidence of recurrence. There was no significant change in eGFR levels in terms of short- term clinical outcomes (t = 0.348, P = 0.733). At 6 months, all 15 patients with imaging studies available had no evidence of recurrence. At 1 year, 1 patient (1 of 15) was noted to have experienced needle tract metastasis and accepted salvage radiofrequency ablation (RFA) therapy. Conclusions IRE appears to be a safe and effective treatment for RCC that may offer a tissue-sparing method and complete ablation as an alternative therapy for RCC.
Collapse
Affiliation(s)
- Ziyin Wang
- Department of Radiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 197#, Rui Jin Er Road, Shanghai, 200025, People's Republic of China
| | - Jian Lu
- Department of Radiology, Ruijin Hospital Luwan Branch, School of Medicine, Shanghai Jiao Tong University, 149#, South Chongqing Road, Shanghai, 200020, People's Republic of China
| | - Wei Huang
- Department of Radiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 197#, Rui Jin Er Road, Shanghai, 200025, People's Republic of China
| | - Zhiyuan Wu
- Department of Radiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 197#, Rui Jin Er Road, Shanghai, 200025, People's Republic of China
| | - Ju Gong
- Department of Radiology, Ruijin Hospital Luwan Branch, School of Medicine, Shanghai Jiao Tong University, 149#, South Chongqing Road, Shanghai, 200020, People's Republic of China
| | - Qingbing Wang
- Department of Radiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 197#, Rui Jin Er Road, Shanghai, 200025, People's Republic of China
| | - Qin Liu
- Department of Radiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 197#, Rui Jin Er Road, Shanghai, 200025, People's Republic of China
| | - Cangyi Wang
- Department of Radiology, Ruijin Hospital Luwan Branch, School of Medicine, Shanghai Jiao Tong University, 149#, South Chongqing Road, Shanghai, 200020, People's Republic of China
| | - Yu Zhu
- Department of Radiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 197#, Rui Jin Er Road, Shanghai, 200025, People's Republic of China.
| | - Xiaoyi Ding
- Department of Radiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 197#, Rui Jin Er Road, Shanghai, 200025, People's Republic of China.
| | - Zhongmin Wang
- Department of Radiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 197#, Rui Jin Er Road, Shanghai, 200025, People's Republic of China. .,Department of Radiology, Ruijin Hospital Luwan Branch, School of Medicine, Shanghai Jiao Tong University, 149#, South Chongqing Road, Shanghai, 200020, People's Republic of China.
| |
Collapse
|
17
|
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: 38] [Impact Index Per Article: 12.7] [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
|
18
|
Massaro EK, Goswami I, Verbridge SS, von Spakovsky MR. Electro-chemo-mechanical model to investigate multi-pulse electric-field-driven integrin clustering. Bioelectrochemistry 2020; 137:107638. [PMID: 33160180 DOI: 10.1016/j.bioelechem.2020.107638] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 08/18/2020] [Accepted: 08/18/2020] [Indexed: 11/28/2022]
Abstract
The effect of pulsed electric fields (PEFs) on transmembrane proteins is not fully understood; how do chemo-mechanical cues in the microenvironment mediate the electric field sensing by these proteins? To answer this key gap in knowledge, we have developed a kinetic Monte Carlo statistical model of the integrin proteins that integrates three components of the morphogenetic field (i.e., chemical, mechanical, and electrical cues). Specifically, the model incorporates the mechanical stiffness of the cell membrane, the ligand density of the extracellular environment, the glycocalyx stiffness, thermal Brownian motion, and electric field induced diffusion. The effects of both steady-state electric fields and transient PEF pulse trains on integrin clustering are studied. Our results reveal that electric-field-driven integrin clustering is mediated by membrane stiffness and ligand density. In addition, we explore the effects of PEF pulse-train parameters (amplitude, polarity, and pulse-width) on integrin clustering. In summary, we demonstrate a computational methodology to incorporate experimental data and simulate integrin clustering when exposed to PEFs for time-scales comparable to experiments (seconds-minutes). Thus, we propose a blueprint for understanding PEF/electric field effects on protein induced signaling and highlight key impediments to incorporating experimental values into computational models such as the kinetic Monte Carlo method.
Collapse
Affiliation(s)
- Evan K Massaro
- Center for Computational Science and Engineering, Massachusetts Institute of Technology, MA, USA
| | - Ishan Goswami
- California Institute for Quantitative Biosciences, University of California Berkeley, CA, USA.
| | - Scott S Verbridge
- Department of Biomedical Engineering and Applied Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Michael R von Spakovsky
- Department of Mechanical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| |
Collapse
|
19
|
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: 3.8] [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
|
20
|
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: 45] [Impact Index Per Article: 11.3] [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
|
21
|
Brock RM, Beitel-White N, Coutermarsh-Ott S, Grider DJ, Lorenzo MF, Ringel-Scaia VM, Manuchehrabadi N, Martin RCG, Davalos RV, Allen IC. Patient Derived Xenografts Expand Human Primary Pancreatic Tumor Tissue Availability for ex vivo Irreversible Electroporation Testing. Front Oncol 2020; 10:843. [PMID: 32528898 PMCID: PMC7257557 DOI: 10.3389/fonc.2020.00843] [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: 01/16/2020] [Accepted: 04/28/2020] [Indexed: 12/13/2022] Open
Abstract
New methods of tumor ablation have shown exciting efficacy in pre-clinical models but often demonstrate limited success in the clinic. Due to a lack of quality or quantity in primary malignant tissue specimens, therapeutic development and optimization studies are typically conducted on healthy tissue or cell-line derived rodent tumors that don't allow for high resolution modeling of mechanical, chemical, and biological properties. These surrogates do not accurately recapitulate many critical components of the tumor microenvironment that can impact in situ treatment success. Here, we propose utilizing patient-derived xenograft (PDX) models to propagate clinically relevant tumor specimens for the optimization and development of novel tumor ablation modalities. Specimens from three individual pancreatic ductal adenocarcinoma (PDAC) patients were utilized to generate PDX models. This process generated 15-18 tumors that were allowed to expand to 1.5 cm in diameter over the course of 50-70 days. The PDX tumors were morphologically and pathologically identical to primary tumor tissue. Likewise, the PDX tumors were also found to be physiologically superior to other in vitro and ex vivo models based on immortalized cell lines. We utilized the PDX tumors to refine and optimize irreversible electroporation (IRE) treatment parameters. IRE, a novel, non-thermal tumor ablation modality, is being evaluated in a diverse range of cancer clinical trials including pancreatic cancer. The PDX tumors were compared against either Pan02 mouse derived tumors or resected tissue from human PDAC patients. The PDX tumors demonstrated similar changes in electrical conductivity and Joule heating following IRE treatment. Computational modeling revealed a high similarity in the predicted ablation size of the PDX tumors that closely correlate with the data generated with the primary human pancreatic tumor tissue. Gene expression analysis revealed that IRE treatment resulted in an increase in biological pathway signaling associated with interferon gamma signaling, necrosis and mitochondria dysfunction, suggesting potential co-therapy targets. Together, these findings highlight the utility of the PDX system in tumor ablation modeling for IRE and increasing clinical application efficacy. It is also feasible that the use of PDX models will significantly benefit other ablation modality testing beyond IRE.
Collapse
Affiliation(s)
- Rebecca M Brock
- Graduate Program in Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA, United States
| | - Natalie Beitel-White
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States.,Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Sheryl Coutermarsh-Ott
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, United States
| | - Douglas J Grider
- Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Virginia Polytechnic Institute and State University, Roanoke, VA, United States
| | - Melvin F Lorenzo
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Veronica M Ringel-Scaia
- Graduate Program in Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA, United States
| | | | - Robert C G Martin
- Division of Surgical Oncology, Department of Surgery, School of Medicine, University of Louisville, Louisville, KY, United States
| | - Rafael V Davalos
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Irving C Allen
- Graduate Program in Translational Biology, 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, Virginia Polytechnic Institute and State University, Roanoke, VA, United States
| |
Collapse
|
22
|
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: 1.5] [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
|