1
|
Radzevičiūtė-Valčiukė E, Mickevičiūtė E, Želvys A, Lekešytė B, Malakauskaitė P, Gečaitė J, Malyško-Ptašinskė V, German N, Žalnėravičius R, Kašėta V, Kulbacka J, Novickij V. Improving bleomycin electrochemotherapy with gold nanoparticles: first in vivo study on intra-tumoral field amplification. Bioelectrochemistry 2025; 165:108999. [PMID: 40345058 DOI: 10.1016/j.bioelechem.2025.108999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/14/2025] [Accepted: 05/04/2025] [Indexed: 05/11/2025]
Abstract
Electrochemotherapy (ECT) is a cancer treatment approach that utilizes the application of electroporation (EP) with standard chemotherapeutic drugs, resulting in a locally enhanced chemotherapy effect due to enhanced intracellular drug delivery. The aim of this study was to demonstrate (for the first time) that microsecond-range bleomycin electrochemotherapy (1.5 kV/cm × 100 μs × 8 pulses, 1 Hz), when combined with gold nanoparticles (AuNPs, 13 and 46 nm), can be efficiently utilized for in vivo carcinoma tumor treatment. It was anticipated that AuNPs would promote a better treatment response due to local electric field amplification within the tumor as predicted by available in vitro research. We focus the attenuation of tumor progression and reduction of the frequency of metastasis incl. the immune response in the murine BALB/C and 4T1 cancer model. It is shown that the application of 13 nm AuNPs hardly influenced the dynamics of tumor progression (when compared to ECT alone), the synergistic effects are not statistically significant by the end of experiment, which is not the case in vitro. However, the application of 46 nm AuNPs significantly potentiated the efficacy of ECT, which confirms the promising alliance of conductive nanoparticles for local intra-tumoral electric field amplification and ECT.
Collapse
Affiliation(s)
- Eivina Radzevičiūtė-Valčiukė
- State Research Institute Centre for Innovative Medicine, Department of Immunology and Bioelectrochemistry, Vilnius, Lithuania; Faculty of Electronics, Vilnius Gediminas Technical University, Vilnius, Lithuania.
| | - Eglė Mickevičiūtė
- State Research Institute Centre for Innovative Medicine, Department of Immunology and Bioelectrochemistry, Vilnius, Lithuania; Faculty of Electronics, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Augustinas Želvys
- State Research Institute Centre for Innovative Medicine, Department of Immunology and Bioelectrochemistry, Vilnius, Lithuania; Faculty of Electronics, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Barbora Lekešytė
- State Research Institute Centre for Innovative Medicine, Department of Immunology and Bioelectrochemistry, Vilnius, Lithuania; Faculty of Electronics, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Paulina Malakauskaitė
- State Research Institute Centre for Innovative Medicine, Department of Immunology and Bioelectrochemistry, Vilnius, Lithuania; Faculty of Electronics, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Jovita Gečaitė
- State Research Institute Centre for Innovative Medicine, Department of Immunology and Bioelectrochemistry, Vilnius, Lithuania
| | - Veronika Malyško-Ptašinskė
- State Research Institute Centre for Innovative Medicine, Department of Immunology and Bioelectrochemistry, Vilnius, Lithuania; Faculty of Electronics, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Natalija German
- State Research Institute Centre for Innovative Medicine, Department of Immunology and Bioelectrochemistry, Vilnius, Lithuania
| | - Rokas Žalnėravičius
- Centre for Physical Science and Technology (FTMC), Department of Electrochemical Materials Science, Vilnius, Lithuania
| | - Vytautas Kašėta
- State Research Institute Centre for Innovative Medicine, Department of Stem Cell Biology, Vilnius, Lithuania
| | - Julita Kulbacka
- State Research Institute Centre for Innovative Medicine, Department of Immunology and Bioelectrochemistry, Vilnius, Lithuania; Wroclaw Medical University, Faculty of Pharmacy, Department of Molecular and Cellular Biology, Wroclaw, Poland
| | - Vitalij Novickij
- State Research Institute Centre for Innovative Medicine, Department of Immunology and Bioelectrochemistry, Vilnius, Lithuania; Faculty of Electronics, Vilnius Gediminas Technical University, Vilnius, Lithuania.
| |
Collapse
|
2
|
Park H, Kim G, Kim N, Ha S, Yim H. Efficacy and safety of natural killer cell therapy in patients with solid tumors: a systematic review and meta-analysis. Front Immunol 2024; 15:1454427. [PMID: 39478866 PMCID: PMC11522797 DOI: 10.3389/fimmu.2024.1454427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024] Open
Abstract
Introduction In 2020, global cancer statistics reported 19.3 million new cases and 10 million deaths annually, highlighting the urgent need for effective treatments. Current therapies, such as surgery, radiation, and chemotherapy, have limitations in comprehensively addressing solid tumor. Recent advances in cancer biology and immuno-oncology, including CAR-T cell therapy, show promise but face efficacy challenges against solid tumors. Methods This meta-analysis systematically reviewed studies from PubMed, Embase, Cochrane, and ClinicalTrials.gov databases up to May 2024 to evaluate the clinical efficacy and safety of unmodified NK cell therapies in solid tumors. The included trials focused on reporting objective response rates (ORR). Results Thirty-one trials involving 600 patients across various cancers (e.g., NSCLC, HCC, breast, ovarian) were analyzed. NK cell therapies demonstrated promising ORRs, particularly 72.3% in hepatocellular carcinoma, often in combination with local therapies. Safety profiles were favorable, with fatigue being the most common adverse event. Discussion NK cell therapies represent a promising treatment option for solid tumors, offering a viable alternative to genetically modified cell therapies like CAR-T. Further research is needed to optimize the clinical utility of NK cell therapy and integrate it effectively into standard cancer treatment regimens. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42023438410, identifier CRD42023438410.
Collapse
Affiliation(s)
- Heesook Park
- Department of Public Health, The Catholic University of Korea, Seoul, Republic of Korea
| | - Gyurin Kim
- Department of Public Health, The Catholic University of Korea, Seoul, Republic of Korea
| | - Najin Kim
- Medical Library, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sungyoen Ha
- Department of Statistics, Sungkyunkwan University of Korea, Seoul, Republic of Korea
| | - Hyeonwoo Yim
- Department of Preventive Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
3
|
Wang K, Wang L, Wang Y, Xiao L, Wei J, Hu Y, Wang D, Huang H. Reprogramming natural killer cells for cancer therapy. Mol Ther 2024; 32:2835-2855. [PMID: 38273655 PMCID: PMC11403237 DOI: 10.1016/j.ymthe.2024.01.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/05/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
The last decade has seen rapid development in the field of cellular immunotherapy, particularly in regard to chimeric antigen receptor (CAR)-modified T cells. However, challenges, such as severe treatment-related toxicities and inconsistent quality of autologous products, have hindered the broader use of CAR-T cell therapy, highlighting the need to explore alternative immune cells for cancer targeting. In this regard, natural killer (NK) cells have been extensively studied in cellular immunotherapy and were found to exert cytotoxic effects without being restricted by human leukocyte antigen and have a lower risk of causing graft-versus-host disease; making them favorable for the development of readily available "off-the-shelf" products. Clinical trials utilizing unedited NK cells or reprogrammed NK cells have shown early signs of their effectiveness against tumors. However, limitations, including limited in vivo persistence and expansion potential, remained. To enhance the antitumor function of NK cells, advanced gene-editing technologies and combination approaches have been explored. In this review, we summarize current clinical trials of antitumor NK cell therapy, provide an overview of innovative strategies for reprogramming NK cells, which include improvements in persistence, cytotoxicity, trafficking and the ability to counteract the immunosuppressive tumor microenvironment, and also discuss some potential combination therapies.
Collapse
Affiliation(s)
- Kexin Wang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Liangzhu Laboratory, Hangzhou, Zhejiang Province, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, China; Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, Zhejiang Province, China
| | - Linqin Wang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Liangzhu Laboratory, Hangzhou, Zhejiang Province, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, China; Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, Zhejiang Province, China
| | - Yiyun Wang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Liangzhu Laboratory, Hangzhou, Zhejiang Province, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, China; Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, Zhejiang Province, China
| | - Lu Xiao
- Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jieping Wei
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Liangzhu Laboratory, Hangzhou, Zhejiang Province, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, China; Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, Zhejiang Province, China
| | - Yongxian Hu
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Liangzhu Laboratory, Hangzhou, Zhejiang Province, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, China; Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, Zhejiang Province, China.
| | - Dongrui Wang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Liangzhu Laboratory, Hangzhou, Zhejiang Province, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, China; Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, Zhejiang Province, China.
| | - He Huang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Liangzhu Laboratory, Hangzhou, Zhejiang Province, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, China; Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
4
|
Holtermann A, Gislon M, Angele M, Subklewe M, von Bergwelt-Baildon M, Lauber K, Kobold S. Prospects of Synergy: Local Interventions and CAR T Cell Therapy in Solid Tumors. BioDrugs 2024; 38:611-637. [PMID: 39080180 PMCID: PMC11358237 DOI: 10.1007/s40259-024-00669-y] [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] [Accepted: 06/17/2024] [Indexed: 08/30/2024]
Abstract
Chimeric antigen receptor T cell therapy has been established in the treatment of various B cell malignancies. However, translating this therapeutic effect to treat solid tumors has been challenging because of their inter-tumoral as well as intratumoral heterogeneity and immunosuppressive microenvironment. Local interventions, such as surgery, radiotherapy, local ablation, and locoregional drug delivery, can enhance chimeric antigen receptor T cell therapy in solid tumors by improving tumor infiltration and reducing systemic toxicities. Additionally, ablation and radiotherapy have proven to (re-)activate systemic immune responses via abscopal effects and reprogram the tumor microenvironment on a physical, cellular, and chemical level. This review highlights the potential synergy of the combined approaches to overcome barriers of chimeric antigen receptor T cell therapy and summarizes recent studies that may pave the way for new treatment regimens.
Collapse
Affiliation(s)
- Anne Holtermann
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80336, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Mila Gislon
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80336, Munich, Germany
| | - Martin Angele
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Marion Subklewe
- Department of Medicine III, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80336, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany.
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München-German Research Center for Environmental Health Neuherberg, Munich, Germany.
| |
Collapse
|
5
|
Zeng J, Liu B, Lang X, Wang Z, Fan Y, Gao C, Fu D. Clinical efficacy and safety of irreversible electroporation combined with chemotherapy in stage IV pancreatic cancer treatment. J Cancer Res Ther 2024; 20:1357-1361. [PMID: 39206999 DOI: 10.4103/jcrt.jcrt_73_24] [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: 01/10/2024] [Accepted: 06/22/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE This study evaluates the clinical efficacy and safety of irreversible electroporation (IRE) therapy combined with chemotherapy in patients with stage IV pancreatic cancer. METHODS Between September 2021 and November 2023, we enrolled 38 patients with stage IV pancreatic cancer, with 20 receiving IRE plus chemotherapy and 18 receiving only chemotherapy. We recorded the general information of the patients and regularly followed up postoperative IRE-related adverse reactions. Progression-free survival (PFS) and overall survival (OS) were evaluated during follow-up. RESULTS Median OS was longer in the IRE group than in the chemotherapy group. Median PFS was slightly extended with IRE compared to chemotherapy alone. The mean hospital stay for the IRE group was 5.90 ± 0.75 days. Four serious adverse events occurred after IRE. Postoperative pain scores were significantly lower than preoperative scores. CONCLUSION IRE combined with chemotherapy showed clinical effectiveness in stage IV pancreatic cancer treatment, offering potential pain reduction benefits with fewer adverse effects and shorter hospital stays.
Collapse
Affiliation(s)
- Jia Zeng
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Boyu Liu
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xu Lang
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhe Wang
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yong Fan
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Chuntao Gao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Dianxun Fu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
6
|
Su YY, Chiang NJ, Chiu TJ, Huang CJ, Hsu SJ, Lin HC, Yang SH, Yang Y, Chou WC, Chen YY, Bai LY, Li CP, Chen JS. Systemic treatments in pancreatic cancer: Taiwan pancreas society recommendation. Biomed J 2024; 47:100696. [PMID: 38169173 PMCID: PMC11332987 DOI: 10.1016/j.bj.2023.100696] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/05/2023] [Accepted: 12/23/2023] [Indexed: 01/05/2024] Open
Abstract
Pancreatic cancer is a highly aggressive malignancy with a poor prognosis. Over the past decade, significant therapeutic advancements have improved the survival rates of patients with pancreatic cancer. One of the primary factors contributing to these positive outcomes is the evolution of chemotherapy, from monotherapy to doublet or triplet regimens, and the integration of multimodal approaches. Additionally, targeted agents tailored to patients with specific genetic alterations and the development of cell therapies show promise in benefiting certain subpopulations. This article focuses on examining pivotal studies that explore the role of chemotherapy in neoadjuvant, adjuvant, maintenance, and salvage settings; highlights interesting findings related to cell therapy; and provides an overview of ongoing trials concerning metastatic settings. This review primarily aimed to offer recommendations based on therapeutic evidence, recent advancements in new treatment combinations, and the most innovative approaches. A unique aspect of this review is the inclusion of published papers on clinical trials and real-world data in Taiwan, thus adding a valuable perspective to the overall analysis.
Collapse
Affiliation(s)
- Yung-Yeh Su
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan; Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Internal Medicine, Kaohsiung Medical University Hospital, and Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Nai-Jung Chiang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan; Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Therapeutic and Research Center of Pancreatic Cancer, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tai-Jan Chiu
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chien-Jui Huang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shao-Jung Hsu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Therapeutic and Research Center of Pancreatic Cancer, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hsin-Chen Lin
- Division of Medical Oncology, Department of Oncology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Shih-Hung Yang
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Youngsen Yang
- Division of Cancer Prevention and Control, Department of Oncology, Taichung Veterans General Hospital, Taichung, Taiwan; College of Medicine, School of Medicine, China Medical University, Taichung, Taiwan
| | - Wen-Chi Chou
- Chang Gung University College of Medicine, Taoyuan, Taiwan; Department of Hematology-Oncology, Linkou Chang Gung Memorial Hospital, Taiwan
| | - Yen-Yang Chen
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Li-Yuan Bai
- College of Medicine, School of Medicine, China Medical University, Taichung, Taiwan; Division of Hematology and Oncology, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Chung-Pin Li
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Therapeutic and Research Center of Pancreatic Cancer, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Clinical Skills Training, Department of Medical Education, Taipei Veterans General Hospital, Taipei, Taiwan.
| | - Jen-Shi Chen
- Chang Gung University College of Medicine, Taoyuan, Taiwan; Department of Hematology-Oncology, Linkou Chang Gung Memorial Hospital, Taiwan.
| |
Collapse
|
7
|
Cao Z, Guan M, Cheng C, Wang F, Jing Y, Zhang K, Jiao J, Ruan L, Chen Z. KIF20B and MET, hub genes of DIAPHs, predict poor prognosis and promote pancreatic cancer progression. Pathol Res Pract 2024; 254:155046. [PMID: 38266456 DOI: 10.1016/j.prp.2023.155046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 01/26/2024]
Abstract
BACKGROUND The DIAPHs (DIAPH1, DIAPH2, and DIAPH3) are members of the diaphanous subfamily of the formin family. KIF20B and MET, hub genes of DIAPHs, play crucial roles in cytoskeletal remodeling, cell migration, and adhesion. However, their combined prognostic and treatment value in pancreatic adenocarcinoma (PC) warrants further investigation. METHODS Multiomics analysis tools were used to comprehensively assess the genomic expression and prognostic value of KIF20B and MET in PC. Immune cell infiltration, functional enrichment, single-cell RNA-seq (scRNA) analysis, potential therapeutic drugs, and nomograms were established and analyzed. CCK-8 levels, transwell assay, Co-IP assay, mass spectrometry, and western blotting were performed to assess the role of KIF20B and MET as modulators of β-catenin and Lactate Dehydrogenase A (LDHA) in vitro. Xenograft tumor models were used to evaluate the anti-tumor effects in vivo. RESULTS DIAPHs, KIF20B, and MET were overexpressed and functioned as poor prognostic markers of PC. Immunoinfiltration analysis revealed that pDC and NK cells were enriched with low expression levels of KIF20B and MET, whereas Th2 cells were enriched with high expression levels of these two genes. The copy number variations (CNVs) in KIF20B and MET were positively correlated with B cell and CD4 + T cell infiltration. Immunological checkpoints NT5E and CD44 were positively correlated with KIF20B and MET expression. Moreover, the nomogram constructed based on KIF20B and MET demonstrated predictive value for overall survival. scRNA-Seq analysis indicated that KIF20B and MET were enriched in endothelial, malignant, B, T, and CD8 + T cells, which correlated with glycolysis and the epithelial-mesenchymal transition (EMT). The interactions of KIF20B and MET with β-catenin and LDHA were verified by Co-IP assay and mass spectrometry. Knockdown of KIF20B and MET downregulates β-catenin and LDHA in vitro. Furthermore, dual knockdown of KIF20B and MET exhibited a synergistic suppressive effect on PC progression in vitro and in vivo. CONCLUSION DIAPHs, KIF20B, and MET are promising candidates for the prognosis and treatment of PC. More importantly, downregulation of KIF20B and MET inhibited pancreatic cancer progression by regulating LDHA and EMT.
Collapse
Affiliation(s)
- Zhangqi Cao
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Mingwei Guan
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chienshan Cheng
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Fengjiao Wang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yanhua Jing
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ke Zhang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Juying Jiao
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Linjie Ruan
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhen Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
8
|
Huang L, Lyu Z, Yang H, Gu M, Jiao Y, Shi Y. Acute toxicities of intravenous, intraperitoneal, or intratumoral injection of natural killer cells in human pancreatic adenocarcinoma-bearing mice: Randomized study. Int Immunopharmacol 2023; 124:110881. [PMID: 37666066 DOI: 10.1016/j.intimp.2023.110881] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 08/10/2023] [Accepted: 08/28/2023] [Indexed: 09/06/2023]
Abstract
AIMS To investigate the possible acute toxicities and pathological changes associated with intravenous, intraperitoneal, or intratumoral injection of natural killer (NK) cells in mice subcutaneously bearing human pancreatic adenocarcinoma (PaC). METHODS 100 NPG tumor-bearing mice (50/sex) were engrafted subcutaneously with human PaC BXPC-3 cells 9 days before administration. They were randomly divided into 10 groups with 5 males and 5 females in each group. Mice in Group 1 were given sodium chloride intravenously as vehicle control, and mice in Groups 2-4 human peripheral blood-derived NK cells intravenously at doses of 2 × 107, 1 × 108, and 5 × 108 cells/kg, respectively; mice in Groups 5-7 were injected with NK cells intraperitoneally at doses of 2 × 107, 1 × 108, and 5 × 108 cells/kg, respectively, and mice in Groups 8-10 with NK cells intratumorally at doses of 4 × 103, 2 × 104, and 1 × 105 cells/mm3, respectively. Each group was given a single dose; the mice were observed clinically, and body weight, food intake, blood biochemistry, and tumor volume were measured. On Day 15, the mice were euthanized for gross anatomy and histopathology. RESULTS On planned euthanasia, in Groups 2-4 no gross or microscopic pathological changes related to cells injection were found; in Groups 5-7 mice of both sexes showed a decrease in extramedullary hematopoiesis of spleen, and at the dose of 5 × 108 cells/kg, mice of both sexes showed an increase in the composition of spleen white pulp cells. In Groups 8-10, mice of both sexes at doses of 4 × 103 and 1 × 105 cells/mm3 and female mice at the dose of 2 × 104 cells/mm3 showed a decrease in extramedullary hematopoiesis of spleen, and female mice at a dose of 4 × 103 cells/mm3 and mice of both sexes at doses of ≥ 2 × 104 cells/mm3 showed an increase in the composition of spleen white pulp cells; perivascular/peribronchiolar inflammatory cell infiltration in lung and bronchus was observed in mice of both sexes at doses of ≥ 2 × 104 cells/mm3, and inflammatory cell infiltration in liver was observed in mice of both sexes at a dose of 1 × 105 cells/mm3. No other abnormal changes with toxicological significance in clinical observation, body weight, food intake, or blood biochemistry were observed in each group. CONCLUSIONS In our study intravenous injection appears the safest way to give NK cells to human PaC-bearing mice. Using intraperitoneal or intratumoral administration, spleen, liver, and lung were the most often affected organs, albeit with mostly mild pathological changes.
Collapse
Affiliation(s)
- Lei Huang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Medical Center on Aging of Ruijin Hospital, MCARJH, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Zhaojie Lyu
- Peking University Shenzhen Hospital, Shenzhen, China
| | - Hui Yang
- Department of General Surgery, Shanghai Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mancang Gu
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China; Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Yang Jiao
- Jiangsu RE-STEM Biotechnology Co., Ltd., Soochow, China.
| | - Yan Shi
- Department of General Surgery, Shanghai Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
9
|
Koh EK, Lee HR, Son WC, Park GY, Kim J, Bae JH, Park YS. Combinatorial immunotherapy with gemcitabine and ex vivo-expanded NK cells induces anti-tumor effects in pancreatic cancer. Sci Rep 2023; 13:7656. [PMID: 37169953 PMCID: PMC10175562 DOI: 10.1038/s41598-023-34827-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/09/2023] [Indexed: 05/13/2023] Open
Abstract
Pancreatic cancer is difficult to diagnose at the initial stage and is often discovered after metastasis to nearby organs. Gemcitabine is currently used as a standard treatment for pancreatic cancer. However, since chemotherapy for pancreatic cancer has not yet reached satisfactory therapeutic results, adjuvant chemotherapy methods are attempted. It can be expected that combining immune cell therapy with existing anticancer drug combination treatment will prevent cancer recurrence and increase survival rates. We isolated natural killer (NK) cells and co-cultured them with strongly activated autologous peripheral blood mononuclear cells (PBMCs) as feeder cells, activated using CD3 antibody, IFN-r, IL-2, and γ-radiation. NK cells expanded in this method showed greater cytotoxicity than resting NK cells, when co-cultured with pancreatic cancer cell lines. Tumor growth was effectively inhibited in a pancreatic cancer mouse xenograft model. Therapeutic efficacy was increased by using gemcitabine and erlotinib in combination. These findings suggest that NK cells cultured by the method proposed here have excellent anti-tumor activity. We demonstrate that activated NK cells can efficiently inhibit pancreatic tumors when used in combination with gemcitabine-based therapy.
Collapse
Affiliation(s)
- Eun-Kyoung Koh
- Department of Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan, 46033, South Korea
- Department of Biochemistry, Pusan National University School of Medicine, Yangsan, 50612, South Korea
| | - Hong-Rae Lee
- Department of Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan, 46033, South Korea
| | - Woo-Chang Son
- Department of Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan, 46033, South Korea
| | - Ga-Young Park
- Department of Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan, 46033, South Korea
| | - Juhee Kim
- Department of Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan, 46033, South Korea
| | - Jae-Ho Bae
- Department of Biochemistry, Pusan National University School of Medicine, Yangsan, 50612, South Korea.
| | - You-Soo Park
- Department of Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan, 46033, South Korea.
| |
Collapse
|
10
|
Li Y, Xiang S, Pan W, Wang J, Zhan H, Liu S. Targeting tumor immunosuppressive microenvironment for pancreatic cancer immunotherapy: Current research and future perspective. Front Oncol 2023; 13:1166860. [PMID: 37064113 PMCID: PMC10090519 DOI: 10.3389/fonc.2023.1166860] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/20/2023] [Indexed: 03/31/2023] Open
Abstract
Pancreatic cancer is one of the most malignant tumors with increased incidence rate. The effect of surgery combined with chemoradiotherapy on survival of patients is unsatisfactory. New treatment strategy such as immunotherapy need to be investigated. The accumulation of desmoplastic stroma, infiltration of immunosuppressive cells including myeloid derived suppressor cells (MDSCs), tumor associated macrophages (TAMs), cancer‐associated fibroblasts (CAFs), and regulatory T cells (Tregs), as well as tumor associated cytokine such as TGF-β, IL-10, IL-35, CCL5 and CXCL12 construct an immunosuppressive microenvironment of pancreatic cancer, which presents challenges for immunotherapy. In this review article, we explore the roles and mechanism of immunosuppressive cells and lymphocytes in establishing an immunosuppressive tumor microenvironment in pancreatic cancer. In addition, immunotherapy strategies for pancreatic cancer based on tumor microenvironment including immune checkpoint inhibitors, targeting extracellular matrix (ECM), interfering with stromal cells or cytokines in TME, cancer vaccines and extracellular vesicles (EVs) are also discussed. It is necessary to identify an approach of immunotherapy in combination with other modalities to produce a synergistic effect with increased response rates in pancreatic cancer therapy.
Collapse
Affiliation(s)
- Ying Li
- Department of Blood Transfusion, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shuai Xiang
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenjun Pan
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Wang
- Department of Operating Room, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hanxiang Zhan
- Department of General Surgery, Qilu hospital, Shandong University, Jinan, Shandong, China
- *Correspondence: Shanglong Liu, ; Hanxiang Zhan,
| | - Shanglong Liu
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Shanglong Liu, ; Hanxiang Zhan,
| |
Collapse
|
11
|
Tasu JP, Tougeron D, Rols MP. Irreversible electroporation and electrochemotherapy in oncology: State of the art. Diagn Interv Imaging 2022; 103:499-509. [PMID: 36266192 DOI: 10.1016/j.diii.2022.09.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 01/10/2023]
Abstract
Thermal tumor ablation techniques including radiofrequency, microwave, LASER, high-intensity focused ultrasound and cryoablation are routinely used to treated liver, kidney, bone, or lung tumors. However, all these techniques are thermal and can therefore be affected by heat sink effect, which can lead to incomplete ablation, and thermal injuries of non-targeted tissues are possible. Under certain conditions, high voltage pulsed electric field can induce formation of pores in the cell membrane. This phenomenon, called electropermeabilization, is also known as "electroporation". Under certain conditions, electroporation can be irreversible, leading to cell death. Irreversible electroporation has demonstrated efficacy for the treatment of liver and prostate cancers, whereas data are scarce regarding pancreatic and renal cancers. During reversible electroporation, transient cell permeability can be used to introduce cytotoxic drugs into tumor cells (commonly bleomycin or cisplatin). Reversible electroporation used in conjunction with cytotoxic drugs shows promise in terms of oncological response, particularly for solid cutaneous and subcutaneous tumors such as melanoma. Irreversible and reversible electroporation are both not thermal ablation techniques and therefore open a new promising horizon for tumor ablation.
Collapse
Affiliation(s)
- Jean-Pierre Tasu
- Department of Diagnosis and interventional radiology, University Hospital of Poitiers, 86021 Poitiers, France; LaTim, UBO and INSERM 1101, University of Brest, 29000 Brest, France.
| | - David Tougeron
- Department of Hepatogastroenterology, University Hospital of Poitiers, 86000 Poitiers, France
| | - Marie-Pierre Rols
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, 31000 Toulouse, France
| |
Collapse
|
12
|
NK and cells with NK-like activities in cancer immunotherapy-clinical perspectives. Med Oncol 2022; 39:131. [PMID: 35716327 DOI: 10.1007/s12032-022-01735-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/13/2022] [Indexed: 01/10/2023]
Abstract
Natural killer (NK) cells are lymphoid cells of innate immunity that take important roles in immune surveillance. NK cells are considered as a bridge between innate and adaptive immunity, and their infiltration into tumor area is related positively with prolonged patient survival. They are defined as CD16+ CD56+ CD3- cells in clinic. NK cells promote cytolytic effects on target cells and induce their apoptosis. Loss of NK cell cytotoxic activity and reduction in the number of activating receptors are the current issues for application of such cells in cellular immunotherapy, which resulted in the diminished long-term effects. The focus of this review is to discuss about the activity of NK cells and cells with NK-like activity including natural killer T (NKT), cytokine-induced killer (CIK) and lymphokine-activated killer (LAK) cells in immunotherapy of human solid cancers.
Collapse
|
13
|
Li Y, Wang J, Wang H, Zhang S, Wei Y, Liu S. The Interplay Between Inflammation and Stromal Components in Pancreatic Cancer. Front Immunol 2022; 13:850093. [PMID: 35493517 PMCID: PMC9046560 DOI: 10.3389/fimmu.2022.850093] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/24/2022] [Indexed: 01/18/2023] Open
Abstract
Inflammation involves interactions between various immune cells, inflammatory cells, chemokines and cytokines in pancreatic cancer. Cancer cells as well as surrounding stromal and inflammatory cells establish an inflammatory tumor microenvironment (TME). Inflammation is closely associated with immunity. Meanwhile, immune cells are involved in both inflammation and immune response. Tumor-promoting inflammation and tumor-suppressive immunity are two main characteristics of the tumor microenvironment in pancreatic cancer. Yet, the mechanism of inflammation and immune response in pancreatic cancer development is still unclear due to the dual role of some cytokines and the complicated crosstalk between tumor and stromal components in TME. In this review, we outline the principal cytokines and stromal cells in the pancreatic TME that are involved in the tumor-promoting and immunosuppressive effects of inflammation, and discuss the interaction between inflammation and stromal components in pancreatic cancer progression. Moreover, the clinical approaches based on targeting TME in pancreatic cancer are also summarized. Defining the mechanisms of interplay between inflammation and stromal components will be essential for further development of anti-cancer therapies.
Collapse
Affiliation(s)
- Ying Li
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Wang
- Department of Operating Room, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Haiyan Wang
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shaoqiang Zhang
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yingxin Wei
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Shanglong Liu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
14
|
Lambin T, Lafon C, Drainville RA, Pioche M, Prat F. Locoregional therapies and their effects on the tumoral microenvironment of pancreatic ductal adenocarcinoma. World J Gastroenterol 2022; 28:1288-1303. [PMID: 35645539 PMCID: PMC9099187 DOI: 10.3748/wjg.v28.i13.1288] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/10/2022] [Accepted: 02/27/2022] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is expected to become the second leading cause of death from cancer by 2030. Despite intensive research in the field of therapeutics, the 5-year overall survival is approximately 8%, with only 20% of patients eligible for surgery at the time of diagnosis. The tumoral microenvironment (TME) of the PDAC is one of the main causes for resistance to antitumoral treatments due to the presence of tumor vasculature, stroma, and a modified immune response. The TME of PDAC is characterized by high stiffness due to fibrosis, with hypo microvascular perfusion, along with an immunosuppressive environment that constitutes a barrier to effective antitumoral treatment. While systemic therapies often produce severe side effects that can alter patients' quality of life, locoregional therapies have gained attention since their action is localized to the pancreas and can thus alleviate some of the barriers to effective antitumoral treatment due to their physical effects. Local hyperthermia using radiofrequency ablation and radiation therapy - most commonly using a local high single dose - are the two main modalities holding promise for clinical efficacy. Recently, irreversible electroporation and focused ultrasound-derived cavitation have gained increasing attention. To date, most of the data are limited to preclinical studies, but ongoing clinical trials may help better define the role of these locoregional therapies in the management of PDAC patients.
Collapse
Affiliation(s)
- Thomas Lambin
- LabTAU, INSERM, Centre Léon Bérard, Université Lyon 1, Univ Lyon, Lyon 69003, France
- Department of Gastroenterology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon 69008, France
| | - Cyril Lafon
- LabTAU, INSERM, Centre Léon Bérard, Université Lyon 1, Univ Lyon, Lyon 69003, France
| | | | - Mathieu Pioche
- Department of Gastroenterology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon 69008, France
| | - Frédéric Prat
- Service d’Endoscopie Digestive, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, Clichy 92110, France
- INSERM U1016, Institut Cochin, Université de Paris, Paris 75014, France
| |
Collapse
|
15
|
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
|
16
|
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
|
17
|
Current Limitations and Novel Perspectives in Pancreatic Cancer Treatment. Cancers (Basel) 2022; 14:cancers14040985. [PMID: 35205732 PMCID: PMC8870068 DOI: 10.3390/cancers14040985] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/03/2022] [Accepted: 02/14/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary This review article presents a synopsis of the key clinical developments, their limitations, and future perspectives in the treatment of pancreatic cancer. In the first part, we summarize the available treatments for pancreatic cancer patients according to tumor stage, as well as the most relevant clinical trials over the past two decades. Despite this progress, there is still much to be improved in terms of patient survival. Therefore, in the second part, we consider various components of the tumor microenvironment in pancreatic cancer, looking for the key drivers of therapy resistance and tumor progression, which may lead to the discovery of new potential targets. We also discuss the most prominent molecules targeting the stroma and immune compartment that are being investigated in either preclinical or clinical trials. Finally, we also outline interesting venues for further research, such as possible combinations of therapies that may have the potential for clinical application. Abstract Pancreatic cancer is one of the deadliest cancers worldwide, largely due to its aggressive development. Consequently, treatment options are often palliative, as only one-fifth of patients present with potentially curable tumors. The only available treatment with curative intent is surgery followed by adjuvant chemotherapy. However, even for patients that are eligible for surgery, the 5-year OS remains below 10%. Hence, there is an urgent need to find new therapeutic regimens. In the first part of this review, we discuss the tumor staging method and its impact on the corresponding current standard-of-care treatments for PDAC. We also consider the key clinical trials over the last 20 years that have improved patient survival. In the second part, we provide an overview of the major components and cell types involved in PDAC, as well as their respective roles and interactions with each other. A deeper knowledge of the interactions taking place in the TME may lead to the discovery of potential new therapeutic targets. Finally, we discuss promising treatment strategies targeting specific components of the TME and potential combinations thereof. Overall, this review provides an overview of the current challenges and future perspectives in the treatment of pancreatic cancer.
Collapse
|
18
|
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: 41] [Impact Index Per Article: 13.7] [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
|
19
|
Tian G, Guan J, Chu Y, Zhao Q, Jiang T. Immunomodulatory Effect of Irreversible Electroporation Alone and Its Cooperating With Immunotherapy in Pancreatic Cancer. Front Oncol 2021; 11:712042. [PMID: 34568040 PMCID: PMC8462269 DOI: 10.3389/fonc.2021.712042] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/19/2021] [Indexed: 01/05/2023] Open
Abstract
Emerging studies have showed irreversible electroporation (IRE) focused on pancreatic cancer (PC). However, the effects of IRE treatment on the immune response of PC remain unknown. Moreover, there are few studies on the therapeutic effect of IRE combining with immunotherapy on PC. Thus, we review recent advances in our understanding of IRE alone and its working with immunotherapy towards the immune response of PC, discussing potential opportunities for exploring future treatment strategies.
Collapse
Affiliation(s)
- Guo Tian
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Biomedicine, Key Laboratory of Pulsed Power Translational Medicine of Zhejiang Province, Hangzhou, China
| | - Jiajia Guan
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanhua Chu
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiyu Zhao
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Biomedicine, Key Laboratory of Pulsed Power Translational Medicine of Zhejiang Province, Hangzhou, China
| | - Tian'an Jiang
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Biomedicine, Key Laboratory of Pulsed Power Translational Medicine of Zhejiang Province, Hangzhou, China
| |
Collapse
|
20
|
Zhu H, Liu X. Advances of Tumorigenesis, Diagnosis at Early Stage, and Cellular Immunotherapy in Gastrointestinal Malignancies. Front Oncol 2021; 11:666340. [PMID: 34434889 PMCID: PMC8381364 DOI: 10.3389/fonc.2021.666340] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 07/19/2021] [Indexed: 01/10/2023] Open
Abstract
Globally, in 2018, 4.8 million new patients have a diagnosis of gastrointestinal (GI) cancers, while 3.4 million people died of such disorders. GI malignancies are tightly relevant to 26% of the world-wide cancer incidence and occupies 35% of all cancer-associated deaths. In this article, we principally investigated molecular and cellular mechanisms of tumorigenesis in five major GI cancers occurring at esophagus, stomach, liver, pancreas, and colorectal region that illustrate high morbidity in Eastern and Western countries. Moreover, through this investigation, we not only emphasize importance of the tumor microenvironment in development and treatment of malignant tumors but also identify significance of M2PK, miRNAs, ctDNAs, circRNAs, and CTCs in early detection of GI cancers, as well as systematically evaluate contribution of personalized precision medicine including cellular immunotherapy, new antigen and vaccine therapy, and oncolytic virotherapy in treatment of GI cancers.
Collapse
Affiliation(s)
- Haipeng Zhu
- Precision and Personalized Cancer Treatment Center, Division of Cancer Diagnosis & Therapy, Ciming Boao International Hospital, Boao Lecheng International Medical Tourism Pilot Zone, Qionghai, China.,Stem Cell and Biotherapy Technology Research Center, Xinxiang Medical College, Xinxiang, China
| | - Xiaojun Liu
- Division of Cellular & Biomedical Science, Ciming Boao International Hospital, Boao Lecheng International Medical Tourism Pilot Zone, Qionghai, China
| |
Collapse
|
21
|
Pancreatic Cancer and Immunotherapy: A Clinical Overview. Cancers (Basel) 2021; 13:cancers13164138. [PMID: 34439292 PMCID: PMC8393975 DOI: 10.3390/cancers13164138] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [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
|
22
|
Peng X, Chen L, Jiao Y, Wang Y, Hao Z, Zhan X. Application of natural killer cells in pancreatic cancer. Oncol Lett 2021; 22:647. [PMID: 34386069 DOI: 10.3892/ol.2021.12908] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/30/2021] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer, a highly malignant disease, is characterized by rapid progression and early metastasis. Although the integrative treatment of pancreatic cancer has made great progress, the prognosis of patients with advanced pancreatic cancer remains extremely poor. In recent years, with the advancements in tumor immunology, immunotherapy has become a promising remedy for pancreatic cancer. Natural killer (NK) cells are the key lymphocytes in the innate immune system. NK cell function does not require antigen pre-sensitization and is not major histocompatibility complex restricted. By targeting tumors or virus-infected cells, the cells play a key role in immune surveillance. Although several questions about NK cells in pancreatic cancer still need to be further studied, there are extensive theories supporting the clinical application prospects of NK cell immunotherapy in pancreatic cancer. Since very few studies have evaluated the role of NK cells in pancreatic cancer, this review provides a comprehensive update of the role of NK cells in pancreatic cancer immunotherapy.
Collapse
Affiliation(s)
- Xiaobo Peng
- Department of Oncology, Changhai Hospital Affiliated to Naval Military Medical University, Shanghai 200433, P.R. China
| | - Ling Chen
- Department of Oncology, Changhai Hospital Affiliated to Naval Military Medical University, Shanghai 200433, P.R. China
| | - Yuan Jiao
- Department of Oncology, Changhai Hospital Affiliated to Naval Military Medical University, Shanghai 200433, P.R. China
| | - Yujie Wang
- Department of Oncology, Changhai Hospital Affiliated to Naval Military Medical University, Shanghai 200433, P.R. China
| | - Zhibin Hao
- Department of Oncology, Changhai Hospital Affiliated to Naval Military Medical University, Shanghai 200433, P.R. China
| | - Xianbao Zhan
- Department of Oncology, Changhai Hospital Affiliated to Naval Military Medical University, Shanghai 200433, P.R. China
| |
Collapse
|
23
|
Stem cells-derived natural killer cells for cancer immunotherapy: current protocols, feasibility, and benefits of ex vivo generated natural killer cells in treatment of advanced solid tumors. Cancer Immunol Immunother 2021; 70:3369-3395. [PMID: 34218295 DOI: 10.1007/s00262-021-02975-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 05/26/2021] [Indexed: 12/13/2022]
Abstract
Nowadays, natural killer (NK) cell-based immunotherapy provides a practical therapeutic strategy for patients with advanced solid tumors (STs). This approach is adaptively conducted by the autologous and identical NK cells after in vitro expansion and overnight activation. However, the NK cell-based cancer immunotherapy has been faced with some fundamental and technical limitations. Moreover, the desirable outcomes of the NK cell therapy may not be achieved due to the complex tumor microenvironment by inhibition of intra-tumoral polarization and cytotoxicity of implanted NK cells. Currently, stem cells (SCs) technology provides a powerful opportunity to generate more effective and universal sources of the NK cells. Till now, several strategies have been developed to differentiate types of the pluripotent and adult SCs into the mature NK cells, with both feeder layer-dependent and/or feeder laye-free strategies. Higher cytokine production and intra-tumoral polarization capabilities as well as stronger anti-tumor properties are the main features of these SCs-derived NK cells. The present review article focuses on the principal barriers through the conventional NK cell immunotherapies for patients with advanced STs. It also provides a comprehensive resource of protocols regarding the generation of SCs-derived NK cells in an ex vivo condition.
Collapse
|
24
|
Fincham REA, Delvecchio FR, Goulart MR, Yeong JPS, Kocher HM. Natural killer cells in pancreatic cancer stroma. World J Gastroenterol 2021; 27:3483-3501. [PMID: 34239264 PMCID: PMC8240050 DOI: 10.3748/wjg.v27.i24.3483] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/06/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer remains one of medicine's largest areas of unmet need. With five-year survival rates of < 8%, little improvement has been made in the last 50 years. Typically presenting with advance stage disease, treatment options are limited. To date, surgery remains the only potentially curative option, however, with such late disease presentation, the majority of patients are unresectable. Thus, new therapeutic options and a greater understanding of the complex stromal interactions within the tumour microenvironment are sorely needed to revise the dismal outlook for pancreatic cancer patients. Natural killer (NK) cells are crucial effector units in cancer immunosurveillance. Often used as a prognostic biomarker in a range of malignancies, NK cells have received much attention as an attractive target for immunotherapies, both as cell therapy and as a pharmaceutical target. Despite this interest, the role of NK cells in pancreatic cancer remains poorly defined. Nevertheless, increasing evidence of the importance of NK cells in this dismal prognosis disease is beginning to come to light. Here, we review the role of NK cells in pancreatic cancer, examine the complex interactions of these crucial effector units within pancreatic cancer stroma and shed light on the increasingly attractive use of NK cells as therapy.
Collapse
Affiliation(s)
- Rachel Elizabeth Ann Fincham
- Barts Cancer Institute-CRUK Centre of Excellence, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Francesca Romana Delvecchio
- Barts Cancer Institute-CRUK Centre of Excellence, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Michelle R Goulart
- Barts Cancer Institute-CRUK Centre of Excellence, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Joe Poe Sheng Yeong
- Institute of Molecular and Cellular Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore
| | - Hemant M Kocher
- Centre for Tumour Biology, Barts Cancer Institute-CRUK Centre of Excellence, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| |
Collapse
|
25
|
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
|
26
|
Lin M, Luo H, Liang S, Chen J, Liu A, Niu L, Jiang Y. Pembrolizumab plus allogeneic NK cells in advanced non-small cell lung cancer patients. J Clin Invest 2021; 130:2560-2569. [PMID: 32027620 DOI: 10.1172/jci132712] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 01/30/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUNDThe anti-programmed cell death 1 (anti-PD-1) antibody pembrolizumab is clinically active against non-small cell lung cancer (NSCLC). In addition to T cells, human natural killer (NK) cells, reported to have the potential to prolong the survival of patients with advanced NSCLC, also express PD-1. This study aimed to investigate the safety and efficacy of pembrolizumab plus allogeneic NK cells in patients with previously treated advanced NSCLC.METHODSIn total, 109 enrolled patients with a programmed death ligand 1 (PD-L1) tumor proportion score (TPS) of 1% or higher were randomly allocated to group A (n = 55 patients given pembrolizumab plus NK cells) or group B (n = 54 patients given pembrolizumab alone). The patients received i.v. pembrolizumab (10 mg/kg) once every 3 weeks and continued treatment until the occurrence of tumor progression or unacceptable toxicity. The patients in group A continuously received 2 cycles of NK cell therapy as 1 course of treatment.RESULTSIn our study, patients in group A had longer survival than did patients in group B (median overall survival [OS]: 15.5 months vs. 13.3 months; median progression-free survival [PFS]: 6.5 months vs. 4.3 months; P < 0.05). In group A patients with a TPS of 50% or higher, the median OS and PFS was significantly longer. Moreover, the patients in group A treated with multiple courses of NK cell infusion had better OS (18.5 months) than did those who received a single course of NK cell infusion (13.5 months).CONCLUSIONPembrolizumab plus NK cell therapy yielded improved survival benefits in patients with previously treated PD-L1+ advanced NSCLC.TRIAL REGISTRATIONClinicalTrials.gov NCT02843204.FUNDINGThis work was supported by grants from the National Natural Science Foundation of China (NSFC) - Guangdong Joint Foundation of China (no. U1601225); the NSFC (no. 81671965); the Guangdong Provincial Key Laboratory Construction Project of China (no. 2017B030314034); and the Key Scientific and Technological Program of Guangzhou City (no. 201607020016).
Collapse
Affiliation(s)
- Mao Lin
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Biological Treatment Center, Fuda Cancer Hospital, Jinan University, Guangzhou, China
| | - Haihua Luo
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shuzhen Liang
- Medical Research Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jibing Chen
- Biological Treatment Center, Fuda Cancer Hospital, Jinan University, Guangzhou, China
| | - Aihua Liu
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lizhi Niu
- Biological Treatment Center, Fuda Cancer Hospital, Jinan University, Guangzhou, China.,Cancer Center, Fuda Cancer Hospital, Jinan University, Guangzhou, China
| | - Yong Jiang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
27
|
Wang L, Xu J, Yu J, Liang P. Review of clinical tumor ablation advance in Asia. Int J Hyperthermia 2021; 38:1639-1649. [PMID: 34781824 DOI: 10.1080/02656736.2021.1983037] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 09/12/2021] [Accepted: 09/15/2021] [Indexed: 02/08/2023] Open
Abstract
Tumor ablation has been widely applied in Asia, accounting for 44.65% of clinical studies worldwide. We reviewed 5853 clinical studies to provide insight on the advance of tumor ablation in Asia chronologically and geographically among different techniques and organs. Since 1998, tumor ablation application has dramatically evolved in Asia. All kinds of ablation techniques, including percutaneous ethanol injection (PEI), radiofrequency ablation (RFA), microwave ablation (MWA), laser ablation (LA), cryoablation (CA), high-intensity focused ultrasound (HIFU), and irreversible electroporation (IRE), have been applied, with the first application of PEI and the most popular application of RFA. Twenty-five countries and one district in Asia have applied tumor ablation in various organs, including liver, lung, uterus, thyroid, kidney, pancreas, bone, prostate, breast, adrenal gland, lymph node parathyroid, esophagus, etc. Due to the high incidence of tumors as well as advanced economy and technology, East Asia accounted for 93.87% of studies, led by China (45.00%), Japan (32.72%), South Korea (12.10%), and Taiwan (4.03%). With the enrichment of evidence from large-scale multicenter and randomized control studies, China and South Korea have issued several guidelines on tumor ablation for liver, lung, and thyroid, which provided recommendations for global standardization of tumor ablation techniques. Therefore, Asia has made active contribution to global tumor ablation therapy.KeypointsKey point 1: Asia accounted for 44.65% of clinical studies worldwide on tumor ablation.Key point 2: Twenty-five countries and one district in Asia have used tumor ablation in various organs, and East Asia accounted for 93.87% of studies, led by China (45.00%), Japan (32.72%), South Korea (12.10%), and Taiwan (4.03%).Key point 3: China and South Korea have issued several guidelines on tumor ablation for liver, lung, and thyroid, which provided recommendations for global standardization of tumor ablation techniques.
Collapse
Affiliation(s)
- Luo Wang
- Department of Interventional Ultrasound, Chinese PLA General Hospital, Beijing, China
| | - Jinshun Xu
- Department of Ultrasound, Laboratory of Ultrasound Imaging Drug, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Yu
- Department of Interventional Ultrasound, Chinese PLA General Hospital, Beijing, China
| | - Ping Liang
- Department of Interventional Ultrasound, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
28
|
Lin M, Zhang X, Liang S, Luo H, Alnaggar M, Liu A, Yin Z, Chen J, Niu L, Jiang Y. Irreversible electroporation plus allogenic Vγ9Vδ2 T cells enhances antitumor effect for locally advanced pancreatic cancer patients. Signal Transduct Target Ther 2020; 5:215. [PMID: 33093457 PMCID: PMC7582168 DOI: 10.1038/s41392-020-00260-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/10/2020] [Accepted: 07/15/2020] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy has limited efficacy against locally advanced pancreatic cancer (LAPC) due to the presence of an immunosuppressive microenvironment (ISM). Irreversible electroporation (IRE) can not only induce immunogenic cell death, but also alleviate immunosuppression. This study aimed to investigate the antitumor efficacy of IRE plus allogeneic γδ T cells in LAPC patients. A total of 62 patients who met the eligibility criteria were enrolled in this trial, then randomized into two groups (A: n = 30 and B: n = 32). All patients received IRE therapy and after receiving IRE, the group A patients received at least two cycles of γδ T-cell infusion as one course continuously. Group A patients had better survival than group B patients (median OS: 14.5 months vs. 11 months; median PFS: 11 months vs. 8.5 months). Moreover, the group A patients treated with multiple courses of γδ T-cell infusion had longer OS (17 months) than those who received a single course (13.5 months). IRE combined with allogeneic γδ T-cell infusion is a promising strategy to enhance the antitumor efficacy in LAPC patients, yielding extended survival benefits. ClinicalTrials.gov ID: NCT03180437.
Collapse
Affiliation(s)
- Mao Lin
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Biological Treatment Center, Fuda Cancer Hospital, Jinan University, Guangzhou, 510665, China
| | - Xiaoyan Zhang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shuzhen Liang
- Medical Research Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Haihua Luo
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Mohammed Alnaggar
- Department of Oncology, Tongji Chibi Hospital, Tongji Medical College, Huazhong University of Science and Technology, Chibi, 437300, China
| | - Aihua Liu
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhinan Yin
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632, China
| | - Jibing Chen
- Biological Treatment Center, Fuda Cancer Hospital, Jinan University, Guangzhou, 510665, China.
| | - Lizhi Niu
- Biological Treatment Center, Fuda Cancer Hospital, Jinan University, Guangzhou, 510665, China. .,Department of Oncology, Fuda Cancer Hospital, Jinan University, Guangzhou, 510665, China.
| | - Yong Jiang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
29
|
Huber M, Brehm CU, Gress TM, Buchholz M, Alashkar Alhamwe B, Pogge von Strandmann E, Slater EP, Bartsch JW, Bauer C, Lauth M. The Immune Microenvironment in Pancreatic Cancer. Int J Mol Sci 2020; 21:E7307. [PMID: 33022971 PMCID: PMC7583843 DOI: 10.3390/ijms21197307] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 02/07/2023] Open
Abstract
The biology of solid tumors is strongly determined by the interactions of cancer cells with their surrounding microenvironment. In this regard, pancreatic cancer (pancreatic ductal adenocarcinoma, PDAC) represents a paradigmatic example for the multitude of possible tumor-stroma interactions. PDAC has proven particularly refractory to novel immunotherapies, which is a fact that is mediated by a unique assemblage of various immune cells creating a strongly immunosuppressive environment in which this cancer type thrives. In this review, we outline currently available knowledge on the cross-talk between tumor cells and the cellular immune microenvironment, highlighting the physiological and pathological cellular interactions, as well as the resulting therapeutic approaches derived thereof. Hopefully a better understanding of the complex tumor-stroma interactions will one day lead to a significant advancement in patient care.
Collapse
Affiliation(s)
- Magdalena Huber
- Institute for Medical Microbiology and Hospital Hygiene, Philipps University Marburg, 35043 Marburg, Germany;
| | - Corinna U. Brehm
- Institute of Pathology, University Hospital Giessen-Marburg, 35043 Marburg, Germany;
| | - Thomas M. Gress
- Department of Gastroenterology, Endocrinology, Metabolism and Infectiology, Center for Tumor- and Immunology (ZTI), Philipps University Marburg, 35043 Marburg, Germany; (T.M.G.); (M.B.); (C.B.)
| | - Malte Buchholz
- Department of Gastroenterology, Endocrinology, Metabolism and Infectiology, Center for Tumor- and Immunology (ZTI), Philipps University Marburg, 35043 Marburg, Germany; (T.M.G.); (M.B.); (C.B.)
| | - Bilal Alashkar Alhamwe
- Institute for Tumor Immunology, Clinic for Hematology, Oncology and Immunology, Center for Tumor Biology and Immunology (ZTI), Philipps University Marburg, 35043 Marburg, Germany; (E.P.v.S.); (B.A.A.)
| | - Elke Pogge von Strandmann
- Institute for Tumor Immunology, Clinic for Hematology, Oncology and Immunology, Center for Tumor Biology and Immunology (ZTI), Philipps University Marburg, 35043 Marburg, Germany; (E.P.v.S.); (B.A.A.)
| | - Emily P. Slater
- Department of Visceral-, Thoracic- and Vascular Surgery, Philipps University Marburg, Baldingerstrasse, 35043 Marburg, Germany;
| | - Jörg W. Bartsch
- Department of Neurosurgery, Philipps University Marburg, Baldingerstrasse, 35043 Marburg, Germany;
| | - Christian Bauer
- Department of Gastroenterology, Endocrinology, Metabolism and Infectiology, Center for Tumor- and Immunology (ZTI), Philipps University Marburg, 35043 Marburg, Germany; (T.M.G.); (M.B.); (C.B.)
| | - Matthias Lauth
- Department of Gastroenterology, Endocrinology, Metabolism and Infectiology, Center for Tumor- and Immunology (ZTI), Philipps University Marburg, 35043 Marburg, Germany; (T.M.G.); (M.B.); (C.B.)
| |
Collapse
|
30
|
Ponath V, Frech M, Bittermann M, Al Khayer R, Neubauer A, Brendel C, Pogge von Strandmann E. The Oncoprotein SKI Acts as A Suppressor of NK Cell-Mediated Immunosurveillance in PDAC. Cancers (Basel) 2020; 12:E2857. [PMID: 33023028 PMCID: PMC7601115 DOI: 10.3390/cancers12102857] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/23/2020] [Accepted: 09/29/2020] [Indexed: 12/14/2022] Open
Abstract
Drugs targeting epigenetic mechanisms such as histone deacetylase inhibitors (HDACi) suppress tumor growth. HDACi also induce the expression of ligands for the cytotoxicity receptor NKG2D rendering tumors more susceptible to natural killer (NK) cell-dependent killing. The major acetylases responsible for the expression of NKG2D ligands (NKG2D-L) are CBP and p300. The role of the oncogene and transcriptional repressor SKI, an essential part of an HDAC-recruiting co-repressor complex, which competes with CBP/p300 for binding to SMAD3 in TGFβ signaling, is unknown. Here we show that the siRNA-mediated downregulation of SKI in the pancreatic cancer cell lines Panc-1 and Patu8988t leads to an increased target cell killing by primary NK cells. However, the higher cytotoxicity of NK cells did not correlate with the induction of NKG2D-L. Of note, the expression of NKG2D-L and consequently NK cell-dependent killing could be induced upon LBH589 (LBH, panobinostat) or valproic acid (VPA) treatment irrespective of the SKI expression level but was significantly higher in pancreatic cancer cells upon genetic ablation of SKI. These data suggest that SKI represses the inducible expression of NKG2D-L. The combination of HDACi with NK cell-based immunotherapy is an attractive treatment option for pancreatic tumors, specifically for patients with high SKI protein levels.
Collapse
Affiliation(s)
- Viviane Ponath
- Institute for Tumor Immunology, Clinic for Hematology, Oncology and Immunology, Philipps University of Marburg, Hans-Meerwein-Strasse 3, 35043 Marburg, Germany; (V.P.); (M.B.); (R.A.K.)
| | - Miriam Frech
- Clinic for Hematology, Oncology, Immunology and Center for Tumor Biology and Immunology, Philipps University of Marburg, Baldingerstrasse, 35037 Marburg, Germany; (M.F.); (A.N.); (C.B.)
| | - Mathis Bittermann
- Institute for Tumor Immunology, Clinic for Hematology, Oncology and Immunology, Philipps University of Marburg, Hans-Meerwein-Strasse 3, 35043 Marburg, Germany; (V.P.); (M.B.); (R.A.K.)
| | - Reem Al Khayer
- Institute for Tumor Immunology, Clinic for Hematology, Oncology and Immunology, Philipps University of Marburg, Hans-Meerwein-Strasse 3, 35043 Marburg, Germany; (V.P.); (M.B.); (R.A.K.)
| | - Andreas Neubauer
- Clinic for Hematology, Oncology, Immunology and Center for Tumor Biology and Immunology, Philipps University of Marburg, Baldingerstrasse, 35037 Marburg, Germany; (M.F.); (A.N.); (C.B.)
| | - Cornelia Brendel
- Clinic for Hematology, Oncology, Immunology and Center for Tumor Biology and Immunology, Philipps University of Marburg, Baldingerstrasse, 35037 Marburg, Germany; (M.F.); (A.N.); (C.B.)
| | - Elke Pogge von Strandmann
- Institute for Tumor Immunology, Clinic for Hematology, Oncology and Immunology, Philipps University of Marburg, Hans-Meerwein-Strasse 3, 35043 Marburg, Germany; (V.P.); (M.B.); (R.A.K.)
| |
Collapse
|
31
|
Regulation and modulation of antitumor immunity in pancreatic cancer. Nat Immunol 2020; 21:1152-1159. [PMID: 32807942 DOI: 10.1038/s41590-020-0761-y] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 07/15/2020] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma carries a dismal prognosis, and outcomes have improved little with modern therapeutics. Checkpoint-based immunotherapy has failed to elicit responses in the vast majority of patients with pancreatic cancer. Alongside tumor cell-intrinsic mechanisms associated with oncogenic KRAS-induced inflammation, the tolerogenic myeloid cell infiltrate has emerged as a critical impediment to adaptive antitumor immune responses. Furthermore, the discovery of an intratumoral microbiome and the elucidation of host-microbe interactions that curtail antitumor immunity also present opportunities for intervention. Here we review the mechanisms of immunotherapy resistance in pancreatic ductal adenocarcinoma and discuss strategies to directly augment T cell responses in parallel with myeloid cell- and microbiome-targeted approaches that may enable immune-mediated control of this malignancy.
Collapse
|
32
|
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
|
33
|
He C, Huang X, Zhang Y, Lin X, Li S. A Novel Prediction Tool Based on Large Cohorts to Determine the Cancer-Specific Survival Probability of Patients With Locally Advanced Pancreatic Cancer After Irreversible Electroporation Treatment. Front Oncol 2020; 10:952. [PMID: 32695671 PMCID: PMC7339950 DOI: 10.3389/fonc.2020.00952] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 05/14/2020] [Indexed: 12/14/2022] Open
Abstract
Irreversible electroporation (IRE) is a novel method which was especially suitable for the treatment of locally advanced pancreatic cancer (LAPC). The purpose of this study was to evaluate probabilities of overall survival (OS) and cancer-specific survival (CSS) in patients with LAPC after IRE treatment and to construct nomograms to predict survival for these patients. Data of patients were retrospectively collected from the Surveillance, Epidemiology, and End Results (SEER) database and medical records of Sun Yat-sen University Cancer Center (SYSUCC). A total of 312 LAPC patients after IRE treatment were included into this study. The 3-year cumulative incidence of cancer-specific mortality for patients with LAPC after IRE treatment was 74.3%. Nomograms for predicting probabilities of OS, CSS, and non-cancer-specific survival (NCSS) were built and calibrated with the concordance index (C-index) and the area under receiver operating characteristic (ROC) curve (AUC). The established nomograms were well-calibrated, with C-indexes of 0.782 for OS prediction, 0.729 for CSS prediction, and 0.730 for NCSS prediction. Compared with the TNM stage system, the established nomograms displayed higher values of AUC and showed better discriminatory power for predicting OS, CSS, and NCSS. These nomograms were well-calibrated and could serve to guide management of LAPC patients after IRE treatment.
Collapse
Affiliation(s)
- Chaobin He
- State Key Laboratory of Oncology in South China, Department of Pancreatobiliary Surgery, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xin Huang
- State Key Laboratory of Oncology in South China, Department of Pancreatobiliary Surgery, 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, China
| | - Xiaojun Lin
- State Key Laboratory of Oncology in South China, Department of Pancreatobiliary Surgery, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shengping Li
- State Key Laboratory of Oncology in South China, Department of Pancreatobiliary Surgery, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
34
|
Analysis of ex vivo expanded and activated clinical-grade human NK cells after cryopreservation. Cytotherapy 2020; 22:450-457. [PMID: 32536506 DOI: 10.1016/j.jcyt.2020.05.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 04/16/2020] [Accepted: 05/02/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND AIMS Several methods to expand and activate (EA) NK cells ex vivo have been developed for the treatment of relapsed or refractory cancers. Infusion of fresh NK cells is generally preferred to the infusion of cryopreserved/thawed (C/T) NK cells because of concern that cryopreservation diminishes NK cell activity. However, there has been little head-to-head comparison of the functionality of fresh versus C/T NK cell products. METHODS We evaluated activity of fresh and C/T EA NK cells generated by interleukin (IL)-15, IL-2 and CD137L expansion. RESULTS Analysis of C/T NK cell products demonstrated decreased recovery of viable CD56+ cells, but the proportion of NK cells in the C/T EA NK cell product did not decrease compared with the fresh EA NK cell product. Fresh and C/T EA NK cells demonstrated increased granzyme B compared with NK cells pre-expansion, but only fresh EA NK cells showed increased NKG2D. Compared with fresh EA NK cells, cytotoxic ability of C/T EA NK cells was reduced, but C/T EA NK cells remained potently cytotoxic against tumor cells via both antibody-independent and antibody-dependent mechanisms within 4 h post-thaw. Fresh EA NK cells generated high levels of gamma interferon (IFN-γ), which was abrogated by JAK1/JAK2 inhibition with ruxolitinib, but C/T EA NK cells showed lower IFN-γ unaffected by JAK1/JAK2 inhibition. DISCUSSION Usage of C/T EA NK cells may be an option to provide serial "boost" NK cell infusions from a single apheresis to maximize NK cell persistence and potentially improve NK-induced responses to refractory cancer.
Collapse
|
35
|
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
|
36
|
Wu Y, Li J, Jabbarzadeh Kaboli P, Shen J, Wu X, Zhao Y, Ji H, Du F, Zhou Y, Wang Y, Zhang H, Yin J, Wen Q, Cho CH, Li M, Xiao Z. Natural killer cells as a double-edged sword in cancer immunotherapy: A comprehensive review from cytokine therapy to adoptive cell immunotherapy. Pharmacol Res 2020; 155:104691. [DOI: 10.1016/j.phrs.2020.104691] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/06/2020] [Accepted: 02/10/2020] [Indexed: 02/08/2023]
|
37
|
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: 197] [Impact Index Per Article: 39.4] [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
|
38
|
Hong G, Chen X, Sun X, Zhou M, Liu B, Li Z, Yu Z, Gao W, Liu T. Effect of autologous NK cell immunotherapy on advanced lung adenocarcinoma with EGFR mutations. PRECISION CLINICAL MEDICINE 2019; 2:235-245. [PMID: 35693880 PMCID: PMC8985770 DOI: 10.1093/pcmedi/pbz023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 11/25/2022] Open
Abstract
This study investigated the efficiency of natural killer (NK) cell immunotherapy on non-small cell lung cancer with and without EGFR mutations in order to evaluate the response rate (RR) and progression-free survival (PFS). Among the 48 patients recruited, 24 were clinically confirmed to be EGFR mutation positive. The study group was treated with autologous NK cell immunotherapy. Comparisons of the lymphocyte number, serum tumour-related biomarkers, circulating tumour cells (CTC), Karnofsky Performance Status (KPS) and survival curves were carried out before and after NK cell immunotherapy. The safety and short-term effects were evaluated, followed by median PFS and RR assessments. The serum CEA and CA125 values were found lower in the NK cell therapy group than that of the non-NK treatment group (p < 0.05). The χ2 test showed a 75% RR of the study group A, significantly higher than that of the control group B (16.7%; p < 0.01). The RR of groups C (58.3%) and D (41.7%) were not statistically significant. The p values of the 4 groups were 0.012, 0.012, 0.166 and 1 from group A to group D, respectively. The median PFS was 9 months in EGFR mutation positive group undergoing NK cell infusion interference. By evaluating the changes in immune function, tumour biomarkers, CTC, KPS and PFS, we demonstrated that NK cell therapy had better clinical therapeutic effects on EGFR mutation-positive lung adenocarcinoma.
Collapse
Affiliation(s)
- Guodai Hong
- Department of Biotherapy and Oncology, Shenzhen Luohu People’s Hospital, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen 518001, China
| | - Xuemei Chen
- The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| | - Xizhuo Sun
- Department of Biotherapy and Oncology, Shenzhen Luohu People’s Hospital, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen 518001, China
| | - Meiling Zhou
- Department of Biotherapy and Oncology, Shenzhen Luohu People’s Hospital, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen 518001, China
- Public Service Platform for Cell Quality Testing and Evaluation of Shenzhen, Shenzhen 518001, China
| | - Bing Liu
- Department of Biotherapy and Oncology, Shenzhen Luohu People’s Hospital, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen 518001, China
- Public Service Platform for Cell Quality Testing and Evaluation of Shenzhen, Shenzhen 518001, China
| | - Zhu Li
- Department of Biotherapy and Oncology, Shenzhen Luohu People’s Hospital, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen 518001, China
- Public Service Platform for Cell Quality Testing and Evaluation of Shenzhen, Shenzhen 518001, China
| | - Zhendong Yu
- Central Laboratory, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Wenbin Gao
- Department of Biotherapy and Oncology, Shenzhen Luohu People’s Hospital, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen 518001, China
| | - Tao Liu
- Department of Biotherapy and Oncology, Shenzhen Luohu People’s Hospital, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen 518001, China
- Public Service Platform for Cell Quality Testing and Evaluation of Shenzhen, Shenzhen 518001, China
| |
Collapse
|
39
|
Lyu C, Lopez-Ichikawa M, Rubinsky B, Chang TT. Normal and fibrotic liver parenchyma respond differently to irreversible electroporation. HPB (Oxford) 2019; 21:1344-1353. [PMID: 30879992 PMCID: PMC7170179 DOI: 10.1016/j.hpb.2019.01.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/30/2018] [Accepted: 01/31/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND The safety and efficacy of irreversible electroporation (IRE) in treating hepatic, biliary, and pancreatic malignancies are active areas of clinical investigation. In addition, recent studies have shown that IRE may enable regenerative surgery and in vivo tissue engineering. To use IRE effectively in these clinical applications, it is important to understand how different tissue microenvironments impact the response to IRE. In this study, we characterize the electrical and histological properties of non-fibrotic and fibrotic liver parenchyma before and after IRE treatment. METHODS Electrical resistivity and histology of fibrotic liver from C57BL/6 mice fed a 0.1% 3,5-diethylcarbonyl-1,4-dihydrocollidine (DDC) diet were compared to those of non-fibrotic liver from matched control mice before and after IRE treatment. RESULTS At baseline, the electrical resistivity of fibrotic liver was lower than that of non-fibrotic liver. Post-IRE, resistivity of non-fibrotic liver declined and then recovered back to baseline with time, correlating with hepatocyte repopulation of the ablated parenchyma without deposition of fibrotic scar. In contrast, resistivity of fibrotic liver remained depressed after IRE treatment, correlating with persistent inflammation. CONCLUSION Non-fibrotic and fibrotic liver respond to IRE differently. The underlying tissue microenvironment is an important modifying factor to consider when designing IRE protocols for tissue ablation.
Collapse
Affiliation(s)
- Chenang Lyu
- Department of Mechanical Engineering, University of California, Berkeley, CA 94720
| | | | - Boris Rubinsky
- Department of Mechanical Engineering, University of California, Berkeley, CA 94720
| | - Tammy T. Chang
- Department of Surgery, University of California, San Francisco, CA 94143
| |
Collapse
|
40
|
Wang L, Cheng CS, Chen L, Chen Z. Benefit from the inclusion of surgery in the treatment of patients with stage III pancreatic cancer: a propensity-adjusted, population-based SEER analysis. Cancer Manag Res 2018; 10:1907-1918. [PMID: 30013397 PMCID: PMC6038853 DOI: 10.2147/cmar.s167103] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose In the past 20 years, surgical resection has been a secure and applicable procedure for pancreatic cancer (PC), but it remains controversial for stage III PC with data evaluating its efficacy mostly derived from small randomized trials. Hence, we designed this study to further evaluate its benefit using surveillance, epidemiology, and end results dataset. Patients and methods Patients with stage III PC were identified in the surveillance, epidemiology, and end results registries from 2004 to 2014. The effect of surgery on cancer-specific survival was assessed by risk-adjusted Cox proportional hazard regression modeling and propensity score matching. Results Overall, 6,138 patients with stage III PC were included. Of these, 608 patients underwent primary tumor surgery. On multivariable analyses, surgery was independently associated with improved cancer-specific survival (HR=0.580; 95% CI=0.523–0.643, p<0.001). The survival benefit with surgery was also observed in the propensity score-matched cohort (HR=0.501; 95% CI=0.438–0.573, p<0.001). Conclusion Primary tumor surgery is associated with improved survival in stage III PC. Prospective randomized trials are needed to confirm these results, and further efforts are required to address patient selection.
Collapse
Affiliation(s)
- Lai Wang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China, .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China,
| | - Chien-Shan Cheng
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China, .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China,
| | - Lianyu Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China, .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China,
| | - Zhen Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China, .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China,
| |
Collapse
|
41
|
Abstract
Pancreatic cancer is among the three deadliest cancers worldwide with the lowest 5-year survival of all cancers. Despite all efforts, therapeutic improvements have barely been made over the last decade. Even recent highly promising targeted and immunotherapeutic approaches did not live up to their expectations. Therefore, other horizons have to be explored. Natural Killer (NK) cells are gaining more and more interest as a highly attractive target for cancer immunotherapies, both as pharmaceutical target and for cell therapies. In this systematic review we summarise the pathophysiological adaptions of NK cells in pancreatic cancer and highlight possible (future) therapeutic NK cell-related targets. Furthermore, an extensive overview of recent therapeutic approaches with an effect on NK cells is given, including cytokine-based, viro- and bacteriotherapy and cell therapy. We also discuss ongoing clinical trials that might influence NK cells. In conclusion, although several issues regarding NK cells in pancreatic cancer remain unsolved and need further investigation, extensive evidence is already provided that support NK cell oriented approaches in pancreatic cancer.
Collapse
|