1
|
Duncan JL, Ahmad RN, Danesi H, Slade DJ, Davalos RV, Verbridge SS. Electro-antibacterial therapy (EAT) to enhance intracellular bacteria clearance in pancreatic cancer cells. Bioelectrochemistry 2024; 157:108669. [PMID: 38377890 PMCID: PMC11648442 DOI: 10.1016/j.bioelechem.2024.108669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 02/08/2024] [Accepted: 02/10/2024] [Indexed: 02/22/2024]
Abstract
Intratumoral bacteria have been implicated in driving tumor progression, yet effective treatments to modulate the tumor microbiome remain limited. In this study, we investigate the use of electroporation in combination with metronidazole to enhance the clearance of intracellular Fusobacterium nucleatum within pancreatic cancer cells. We explore various parameters, including electric field strength, pulse width, and pulse number to assess the permeability of pancreatic cancer cells infected with F. nucleatum, compared to non-infected cells of the same type. We subsequently quantify the clearance of intracellular bacteria when these pulsing schemes are applied to a suspension of infected pancreatic cancer cells in the presence of metronidazole. Our results reveal distinct differences in cell permeability between infected and non-infected cells, identifying a unique biophysical marker for host cells infected with F. nucleatum. We demonstrate that the combinatorial use of electroporation and metronidazole significantly enhances the delivery of metronidazole into host cells, leading to more effective clearance of intracellular F. nucleatum compared to independent treatments; we term this novel approach Electro-Antibacterial Therapy (EAT). EAT holds promise as an innovative strategy for addressing intratumoral bacteria in pancreatic cancer, other malignancies, and potentially treatment-resistant infections, offering new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Josie L Duncan
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech - Emory University, Atlanta, GA, USA
| | - Raffae N Ahmad
- Virginia Tech - Wake Forest University School of Biomedical Engineering and Sciences, Blacksburg, VA, USA
| | - Hunter Danesi
- Virginia Tech - Wake Forest University School of Biomedical Engineering and Sciences, Blacksburg, VA, USA
| | - Daniel J Slade
- Department of Biochemistry, Virginia Tech, Blacksburg, VA, USA
| | - Rafael V Davalos
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech - Emory University, Atlanta, GA, USA; Virginia Tech - Wake Forest University School of Biomedical Engineering and Sciences, Blacksburg, VA, USA.
| | - Scott S Verbridge
- Virginia Tech - Wake Forest University School of Biomedical Engineering and Sciences, Blacksburg, VA, USA
| |
Collapse
|
2
|
Kulbacka J, Rembiałkowska N, Radzevičiūtė-Valčiukė E, Szewczyk A, Novickij V. Cardiomyocytes Permeabilization and Electrotransfection by Unipolar and Bipolar Asymmetric Electric Field Pulses. Bioelectricity 2024; 6:91-96. [PMID: 39119571 PMCID: PMC11304875 DOI: 10.1089/bioe.2024.0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024] Open
Abstract
Short electric field pulses represent a novel potential approach for achieving uniform electroporation within tissue containing elongated cells oriented in various directions, such as electroporation-based cardiac ablation procedures. In this study, we investigated how electroporation with nanosecond pulses with respect to different pulse shapes (unipolar, bipolar, and asymmetric) influences cardiomyocyte permeabilization and gene transfer. For this purpose, rat cardiomyocytes (H9c2) were used. The efficacy of the pulsed electric field protocols was assessed by flow cytometry and electrogene transfer by fluorescent and holotomographic microscopy. The response of the cells was assessed by the metabolic activity (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide [MTT] assay), F-actin distribution in cells by confocal microscopy, and muscle atrophy F-box (MAFbx) marker. We show nano- and microsecond pulse protocols, which are not cytotoxic for cardiac muscle cells and can be efficiently used for gene electrotransfection. Asymmetric nanosecond pulsed electric fields were similarly efficient in plasmid delivery as microsecond and millisecond protocols. However, the millisecond protocol induced a higher MAFbx expression in H9c2 cells.
Collapse
Affiliation(s)
- Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Nina Rembiałkowska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Eivina Radzevičiūtė-Valčiukė
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Institute of High Magnetic Fields, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Anna Szewczyk
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Vitalij Novickij
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Institute of High Magnetic Fields, Vilnius Gediminas Technical University, Vilnius, Lithuania
| |
Collapse
|
3
|
Szlasa W, Sauer N, Baczyńska D, Ziętek M, Haczkiewicz-Leśniak K, Karpiński P, Fleszar M, Fortuna P, Kulus MJ, Piotrowska A, Kmiecik A, Barańska A, Michel O, Novickij V, Tarek M, Kasperkiewicz P, Dzięgiel P, Podhorska-Okołów M, Saczko J, Kulbacka J. Pulsed electric field induces exocytosis and overexpression of MAGE antigens in melanoma. Sci Rep 2024; 14:12546. [PMID: 38822068 PMCID: PMC11143327 DOI: 10.1038/s41598-024-63181-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 05/27/2024] [Indexed: 06/02/2024] Open
Abstract
Nanosecond pulsed electric field (nsPEF) has emerged as a promising approach for inducing cell death in melanoma, either as a standalone treatment or in combination with chemotherapeutics. However, to date, there has been a shortage of studies exploring the impact of nsPEF on the expression of cancer-specific molecules. In this investigation, we sought to assess the effects of nsPEF on melanoma-specific MAGE (Melanoma Antigen Gene Protein Family) expression. To achieve this, melanoma cells were exposed to nsPEF with parameters set at 8 kV/cm, 200 ns duration, 100 pulses, and a frequency of 10 kHz. We also aimed to comprehensively describe the consequences of this electric field on melanoma cells' invasion and proliferation potential. Our findings reveal that following exposure to nsPEF, melanoma cells release microvesicles containing MAGE antigens, leading to a simultaneous increase in the expression and mRNA content of membrane-associated antigens such as MAGE-A1. Notably, we observed an unexpected increase in the expression of PD-1 as well. While we did not observe significant differences in the cells' proliferation or invasion potential, a remarkable alteration in the cells' metabolomic and lipidomic profiles towards a less aggressive phenotype was evident. Furthermore, we validated these results using ex vivo tissue cultures and 3D melanoma culture models. Our study demonstrates that nsPEF can elevate the expression of membrane-associated proteins, including melanoma-specific antigens. The mechanism underlying the overexpression of MAGE antigens involves the initial release of microvesicles containing MAGE antigens, followed by a gradual increase in mRNA levels, ultimately resulting in elevated expression of MAGE antigens post-experiment. These findings shed light on a novel method for modulating cancer cells to overexpress cancer-specific molecules, thereby potentially enhancing their sensitivity to targeted anticancer therapy.
Collapse
Affiliation(s)
- Wojciech Szlasa
- Medical University Hospital, Borowska 213, 50-556, Wrocław, Poland.
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland.
| | - Natalia Sauer
- Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Dagmara Baczyńska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Marcin Ziętek
- Department of Surgical Oncology, Wroclaw Comprehensive Cancer Center, Wroclaw, Poland
| | | | - Paweł Karpiński
- Department of Genetics, Wroclaw Medical University, Wroclaw, Poland
| | - Mariusz Fleszar
- Department of Medical Biochemistry, Wroclaw Medical University, Wroclaw, Poland
- Omics Research Center, Wroclaw Medical University, Wrocław, Poland
| | - Paulina Fortuna
- Department of Medical Biochemistry, Wroclaw Medical University, Wroclaw, Poland
- Omics Research Center, Wroclaw Medical University, Wrocław, Poland
| | - Michał J Kulus
- Division of Ultrastructural Research, Faculty of Medicine, Wroclaw Medical University, 50-368, Wroclaw, Poland
| | - Aleksandra Piotrowska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw, Poland
| | - Alicja Kmiecik
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw, Poland
| | - Agnieszka Barańska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw, Poland
| | - Olga Michel
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Vitalij Novickij
- Faculty of Electronics, Vilnius Gediminas Technical University, 03227, Vilnius, Lithuania
- Department of Immunology, State Research Institute Centre for Innovative Medicine, Santariškių 5, 08410, Vilnius, Lithuania
| | - Mounir Tarek
- Université de Lorraine, CNRS, LPCT, 54000, Nancy, France
| | - Paulina Kasperkiewicz
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw, Poland
| | - Marzenna Podhorska-Okołów
- Division of Ultrastructural Research, Faculty of Medicine, Wroclaw Medical University, 50-368, Wroclaw, Poland
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
- Department of Immunology, State Research Institute Centre for Innovative Medicine, Santariškių 5, 08410, Vilnius, Lithuania
| |
Collapse
|
4
|
Lee KE, Xu Y, Geng B, Kim J, Kellon N, He M, Zhang Z, Li D, Gouchoe DA, Zhu H. Prime editing-mediated correction of the leptin receptor in muscle cells of db/db mice. Biotechnol J 2024; 19:e2300676. [PMID: 38730523 PMCID: PMC11090457 DOI: 10.1002/biot.202300676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/22/2024] [Accepted: 04/01/2024] [Indexed: 05/13/2024]
Abstract
Genetic diseases can be caused by monogenic diseases, which result from a single gene mutation in the DNA sequence. Many innovative approaches have been developed to cure monogenic genetic diseases, namely by genome editing. A specific type of genomic editing, prime editing, has the potential advantage to edit the human genome without requiring double-strand breaks or donor DNA templates for editing. Additionally, prime editing does not require a precisely positioned protospacer adjacent motif (PAM) sequence, which offers flexible target and more precise genomic editing. Here we detail a novel construction of a prime editing extended guide RNA (pegRNA) to target mutated leptin receptors in B6.BKS(D)-Leprdb/J mice (db/db mice). The pegRNA was then injected into the flexor digitorum brevis (FDB) muscle of db/db mice to demonstrate in vivo efficacy, which resulted in pegRNA mediated base transversion at endogenous base transversion. Genomic DNA sequencing confirmed that prime editing could correct the mutation of leptin receptor gene in db/db mice. Furthermore, prime editing treated skeletal muscle exhibited enhanced leptin receptor signals. Thus, the current study showed in vivo efficacy of prime editing to correct mutant protein and rescue the physiology associated with functional protein.
Collapse
Affiliation(s)
- Kyung Eun Lee
- Department of Surgery, Davis Heart and Lung Research Institute, the Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Yanping Xu
- Department of Surgery, Davis Heart and Lung Research Institute, the Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Bingchuan Geng
- Department of Surgery, Davis Heart and Lung Research Institute, the Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Jongsoo Kim
- Department of Surgery, Davis Heart and Lung Research Institute, the Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Natalie Kellon
- Department of Surgery, Davis Heart and Lung Research Institute, the Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Michele He
- Department of Surgery, Davis Heart and Lung Research Institute, the Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Zhentao Zhang
- Department of Surgery, Davis Heart and Lung Research Institute, the Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Deqiang Li
- The Center for Cardiovascular Research at Nationwide Children’s Hospital, Columbus, OH 43205
| | - Doug A. Gouchoe
- COPPER Laboratory, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus OH 43210, USA
| | - Hua Zhu
- Department of Surgery, Davis Heart and Lung Research Institute, the Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
5
|
Chandarana C, Tiwari A. A Review of Clinical Trials of Cancer and Its Treatment as a Vaccine. Rev Recent Clin Trials 2024; 19:7-33. [PMID: 37953617 DOI: 10.2174/0115748871260733231031081921] [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: 06/03/2023] [Revised: 08/20/2023] [Accepted: 09/11/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND Cancer and infectious diseases are one of the greatest challenges of modern medicine. An unhealthy lifestyle, poor drug use, or drug misuse contribute to the rise in morbidity and mortality brought on by these illnesses. The inadequacies of the medications now being used to treat these disorders, along with the growing issue of drug resistance, have compelled researchers to look for novel compounds with therapeutic promise. The number of infections and diseases has significantly abated due to vaccine development and use over time, which is described in detail. Several novel vaccines can now be produced by manipulating Deoxyribonucleic acid (DNA), Ribonucleic acid (RNA), Messenger Ribonucleic acid (mRNA), proteins, viral vector Recombinant, and other molecules due to advances in genetic engineering and our understanding of the immune defense. OBJECTIVE The main topic of discussion is cancer-based vaccinations, which were developed less than a decade ago but have already been used to treat a wide range of both life-threatening and deadly diseases. It contains clinical studies for cancer vaccines against kidney, liver, prostate, cervix, and certain RNA-based cancer vaccines against breast and bladder cancer. RESULTS Numerous studies using various DNA and RNA-based methods have been conducted on the basis of cancer, with 9-10 diseases related to DNA and 8-9 diseases associated with RNA. Some of these studies have been completed, while others have been eliminated due to a lack of research; further studies are ongoing regarding the same. CONCLUSION This brief discussion of vaccines and their varieties with examples also discusses vaccine clinical trials in relation to cancer diseases in this DNA and RNA-based cancer vaccine that has had successful clinical trials like the cervical cancer drug VGX-3100, the kidney cancer drug Pembrolizumab, MGN-1601, the prostate cancer drug pTVG-HP with rhGM-CSF, the melanoma cancer drug proteasome siRNA, and the lung cancer drug FRAME-001.
Collapse
Affiliation(s)
- Chandani Chandarana
- Department of Quality Assurance, SSR College of Pharmacy, Sayli Road, Silvassa, U.T of Dadra Nagar and Haveli- 396230, India
| | - Anuradha Tiwari
- Department of Quality Assurance, SSR College of Pharmacy, Sayli Road, Silvassa, U.T of Dadra Nagar and Haveli- 396230, India
| |
Collapse
|
6
|
Endo Y. Editorial: Application of innovative techniques in genetic and cellular therapies. Front Bioeng Biotechnol 2023; 11:1336900. [PMID: 38164404 PMCID: PMC10757950 DOI: 10.3389/fbioe.2023.1336900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 12/08/2023] [Indexed: 01/03/2024] Open
Affiliation(s)
- Yori Endo
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
7
|
Yang M, Wang PY, Hao YL, Liang M, Yu ZY, Li XC, Li YP. A real-world case-control study on the efficacy and safety of pulsed field ablation for atrial fibrillation. Eur J Med Res 2023; 28:519. [PMID: 37968748 PMCID: PMC10648358 DOI: 10.1186/s40001-023-01509-5] [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: 09/13/2023] [Accepted: 11/05/2023] [Indexed: 11/17/2023] Open
Abstract
OBJECTIVE The primary objective of this study was to evaluate the efficacy and safety of pulsed field ablation in individuals diagnosed with atrial fibrillation. METHODS A total of 36 patients diagnosed with atrial fibrillation were enrolled in the pulsed field ablation group, while another 36 patients diagnosed with atrial fibrillation were included in the radiofrequency ablation group. Among the study participants, 15 patients in the pulsed field ablation group and 17 patients in the radiofrequency ablation group had persistent atrial fibrillation. Comprehensive comparisons were made between the two groups, including baseline data, underlying diseases, medication usage, intraoperative parameters, and atrial fibrillation recurrence rates at 1, 3, and 6 months during the postoperative follow-up period. RESULTS (1) There were no significant differences observed between the two groups concerning baseline data and antiarrhythmic drug usage (P > 0.05); (2) the effective ablation time for both left and right pulmonary veins in the pulsed field ablation group was markedly shorter compared to the radiofrequency ablation group (P < 0.001 for each vein); (3) within the pulsed field ablation group, the number of discharges, catheter operation time, and effective ablation time for the left pulmonary vein were significantly higher than those for the right pulmonary vein (P < 0.05). Conversely, in the radiofrequency ablation group, the number of discharges for the left pulmonary vein was significantly higher than that for the right pulmonary vein (P < 0.05); and (4) when comparing sinus rhythm maintenance at 1, 3, and 6 months postoperatively, no statistically significant differences were noted between the two groups for paroxysmal, persistent, and paroxysmal + persistent atrial fibrillation cases (P > 0.05). CONCLUSION During the 6-month follow-up period, pulsed field ablation demonstrated comparable efficacy to radiofrequency ablation with respect to recurrence rates for both paroxysmal and persistent atrial fibrillation. Moreover, pulsed field ablation exhibited high safety levels, excellent surgical efficiency, and a notably brief learning curve, affirming its viability as a therapeutic option for these conditions.
Collapse
Affiliation(s)
- Ming Yang
- Department of Cardiology, The People's Hospital of Yuxi City, The 6th Affiliated Hospital of Kunming Medical University, No.21 of Nieer Road, Hongta District, Yuxi, 653100, China.
| | - Peng-Yu Wang
- Department of Cardiology, The People's Hospital of Yuxi City, The 6th Affiliated Hospital of Kunming Medical University, No.21 of Nieer Road, Hongta District, Yuxi, 653100, China
| | - Ying-Lu Hao
- Department of Cardiology, The People's Hospital of Yuxi City, The 6th Affiliated Hospital of Kunming Medical University, No.21 of Nieer Road, Hongta District, Yuxi, 653100, China
| | - Mei Liang
- Department of Cardiology, The People's Hospital of Yuxi City, The 6th Affiliated Hospital of Kunming Medical University, No.21 of Nieer Road, Hongta District, Yuxi, 653100, China
| | - Zi-Yang Yu
- Department of Cardiology, The People's Hospital of Yuxi City, The 6th Affiliated Hospital of Kunming Medical University, No.21 of Nieer Road, Hongta District, Yuxi, 653100, China
| | - Xi-Chen Li
- Department of Cardiology, The People's Hospital of Yuxi City, The 6th Affiliated Hospital of Kunming Medical University, No.21 of Nieer Road, Hongta District, Yuxi, 653100, China
| | - Yan-Ping Li
- Department of Cardiology, The People's Hospital of Yuxi City, The 6th Affiliated Hospital of Kunming Medical University, No.21 of Nieer Road, Hongta District, Yuxi, 653100, China
| |
Collapse
|
8
|
Aryana A, Ji SY, Hata C, de la Rama A, Nguyen K, Panescu D. Preclinical evaluation of a novel single-shot pulsed field ablation system for pulmonary vein and atrial ablation. J Cardiovasc Electrophysiol 2023; 34:2203-2212. [PMID: 37464948 DOI: 10.1111/jce.16010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 06/06/2023] [Accepted: 07/10/2023] [Indexed: 07/20/2023]
Abstract
INTRODUCTION Pulsed field ablation (PFA) is a nonthermal ablative strategy that achieves cell death via electroporation. Herein, we investigated the preclinical safety and efficacy of PFA using two novel 8-French, 16-electrode spiral PFA/mapping catheters (ElePulse; CRC EP, Inc.). METHODS Bipolar PFA (>1.8 kV) was performed using 30 s, single-shot, QRS-gated applications. Altogether, 94 atrial structures were ablated in 23 swine, one canine, and one ovine, including right and left atria and atrial appendages, pulmonary veins, and superior and inferior (IVC) vena cavae. We also examined the impact of PFA on the phrenic nerve (14 swine) and on a deviated esophagus after delivery of PFA from inside the IVC (five swine). RESULTS All applications were single-shot without catheter repositioning. Minimal microbubbling was observed without significant skeletal muscle twitching/activation (mean acceleration: 0.05 m/s2 ). There was a marked reduction in post-PFA versus pre-PFA atrial electrogram amplitude (0.17 ± 0.21 vs. 1.18 ± 1.08 mV; p < .0001). Lesion durability was demonstrated up to 3 months in all targeted tissues. Histologically, lesions were contiguous and transmural, except in the atrial appendage, and without any thermal effects. Magnetic resonance, gross, and histologic examinations of the brain, rete mirabile, and kidneys revealed no thromboembolism. No acute/long-term phrenic nerve dysfunction was encountered. Although within 2 h of ablation, histologic examinations of the esophagus revealed acute PFA-related changes in the muscular layer, these completely resolved by 21 ± 5 days. CONCLUSION A novel, single-shot, spiral PFA system is capable of safely creating large, durable atrial lesions without significant adverse effects on the phrenic nerve or the esophagus.
Collapse
Affiliation(s)
- Arash Aryana
- Cardiac Arrhythmia Service, Mercy General Hospital and Dignity Health Heart and Vascular Institute, Sacramento, California, USA
| | | | - Cary Hata
- CRC EP, Inc., Tustin, California, USA
| | | | | | | |
Collapse
|
9
|
Komel T, Bosnjak M, Sersa G, Cemazar M. Expression of GFP and DsRed fluorescent proteins after gene electrotransfer of tumour cells in vitro. Bioelectrochemistry 2023; 153:108490. [PMID: 37356264 DOI: 10.1016/j.bioelechem.2023.108490] [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: 02/22/2023] [Revised: 06/15/2023] [Accepted: 06/17/2023] [Indexed: 06/27/2023]
Abstract
Fluorescent reporter genes are widely used to study the transfection of various types of primary cells and cell lines. The aim of our research was to investigate the expression dynamics of GFP and DsRed reporter genes individually and combined after gene electrotransfer of plasmids with two different electroporation protocols in B16F10 and CT26 cells in vitro. The cytotoxicity after gene electrotransfer of both plasmids was first determined. Second, the intensity of fluorescence and the percentage of cells transfected with both plasmids individually and in combination were monitored in real time. The results show that the percentage of viability after gene electrotransfer of plasmids using the EP2 pulses was significantly higher compared to the EP1 pulses. In contrast, the percentage of transfected cells and fluorescence intensity were higher after gene electrotransfer with the EP1 pulse protocol. Moreover, the percentage of transfected cells was higher and started earlier in the B16F10 cell line than in the CT26 cell line. However, fluorescence intensity was higher in CT26 cells. Co-expression of fluorescent proteins was achieved only in a small number of cells. In conclusion, this study elucidated some of the dynamics of reporter gene expression in cancer cell lines after gene electrotransfer.
Collapse
Affiliation(s)
- Tilen Komel
- Institute of Oncology Ljubljana, Department of Experimental Oncology, Zaloska 2, SI-1000 Ljubljana, Slovenia; University of Ljubljana, Faculty of Medicine, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| | - Masa Bosnjak
- Institute of Oncology Ljubljana, Department of Experimental Oncology, Zaloska 2, SI-1000 Ljubljana, Slovenia
| | - Gregor Sersa
- Institute of Oncology Ljubljana, Department of Experimental Oncology, Zaloska 2, SI-1000 Ljubljana, Slovenia; University of Ljubljana, Faculty of Health Sciences, Zdravstvena pot 5, SI - 1000 Ljubljana, Slovenia
| | - Maja Cemazar
- Institute of Oncology Ljubljana, Department of Experimental Oncology, Zaloska 2, SI-1000 Ljubljana, Slovenia; University of Primorska, Faculty of Health Sciences, Polje 42, SI - 6310 Izola, Slovenia.
| |
Collapse
|
10
|
Chehelgerdi M, Chehelgerdi M. The use of RNA-based treatments in the field of cancer immunotherapy. Mol Cancer 2023; 22:106. [PMID: 37420174 PMCID: PMC10401791 DOI: 10.1186/s12943-023-01807-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/13/2023] [Indexed: 07/09/2023] Open
Abstract
Over the past several decades, mRNA vaccines have evolved from a theoretical concept to a clinical reality. These vaccines offer several advantages over traditional vaccine techniques, including their high potency, rapid development, low-cost manufacturing, and safe administration. However, until recently, concerns over the instability and inefficient distribution of mRNA in vivo have limited their utility. Fortunately, recent technological advancements have mostly resolved these concerns, resulting in the development of numerous mRNA vaccination platforms for infectious diseases and various types of cancer. These platforms have shown promising outcomes in both animal models and humans. This study highlights the potential of mRNA vaccines as a promising alternative approach to conventional vaccine techniques and cancer treatment. This review article aims to provide a thorough and detailed examination of mRNA vaccines, including their mechanisms of action and potential applications in cancer immunotherapy. Additionally, the article will analyze the current state of mRNA vaccine technology and highlight future directions for the development and implementation of this promising vaccine platform as a mainstream therapeutic option. The review will also discuss potential challenges and limitations of mRNA vaccines, such as their stability and in vivo distribution, and suggest ways to overcome these issues. By providing a comprehensive overview and critical analysis of mRNA vaccines, this review aims to contribute to the advancement of this innovative approach to cancer treatment.
Collapse
Affiliation(s)
- Mohammad Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran.
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Matin Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| |
Collapse
|
11
|
Campana LG, Daud A, Lancellotti F, Arroyo JP, Davalos RV, Di Prata C, Gehl J. Pulsed Electric Fields in Oncology: A Snapshot of Current Clinical Practices and Research Directions from the 4th World Congress of Electroporation. Cancers (Basel) 2023; 15:3340. [PMID: 37444450 PMCID: PMC10340685 DOI: 10.3390/cancers15133340] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/29/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
The 4th World Congress of Electroporation (Copenhagen, 9-13 October 2022) provided a unique opportunity to convene leading experts in pulsed electric fields (PEF). PEF-based therapies harness electric fields to produce therapeutically useful effects on cancers and represent a valuable option for a variety of patients. As such, irreversible electroporation (IRE), gene electrotransfer (GET), electrochemotherapy (ECT), calcium electroporation (Ca-EP), and tumour-treating fields (TTF) are on the rise. Still, their full therapeutic potential remains underappreciated, and the field faces fragmentation, as shown by parallel maturation and differences in the stages of development and regulatory approval worldwide. This narrative review provides a glimpse of PEF-based techniques, including key mechanisms, clinical indications, and advances in therapy; finally, it offers insights into current research directions. By highlighting a common ground, the authors aim to break silos, strengthen cross-functional collaboration, and pave the way to novel possibilities for intervention. Intriguingly, beyond their peculiar mechanism of action, PEF-based therapies share technical interconnections and multifaceted biological effects (e.g., vascular, immunological) worth exploiting in combinatorial strategies.
Collapse
Affiliation(s)
- Luca G. Campana
- Department of Surgery, Manchester University NHS Foundation Trust, Oxford Rd., Manchester M13 9WL, UK;
| | - Adil Daud
- Department of Medicine, University of California, 550 16 Street, San Francisco, CA 94158, USA;
| | - Francesco Lancellotti
- Department of Surgery, Manchester University NHS Foundation Trust, Oxford Rd., Manchester M13 9WL, UK;
| | - Julio P. Arroyo
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA; (J.P.A.); (R.V.D.)
| | - Rafael V. Davalos
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA; (J.P.A.); (R.V.D.)
- Institute for Critical Technology and Applied Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Claudia Di Prata
- Department of Surgery, San Martino Hospital, 32100 Belluno, Italy;
| | - Julie Gehl
- Department of Clinical Oncology and Palliative Care, Zealand University Hospital, 4000 Roskilde, Denmark;
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 1165 Copenhagen, Denmark
| |
Collapse
|
12
|
Jiang M, Fiering S, Shao Q. Combining energy-based focal ablation and immune checkpoint inhibitors: preclinical research and clinical trials. Front Oncol 2023; 13:1153066. [PMID: 37251920 PMCID: PMC10211342 DOI: 10.3389/fonc.2023.1153066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 04/12/2023] [Indexed: 05/31/2023] Open
Abstract
Energy-based focal therapy (FT) uses targeted, minimally invasive procedures to destroy tumors while preserving normal tissue and function. There is strong emerging interest in understanding how systemic immunity against the tumor can occur with cancer immunotherapy, most notably immune checkpoint inhibitors (ICI). The motivation for combining FT and ICI in cancer management relies on the synergy between the two different therapies: FT complements ICI by reducing tumor burden, increasing objective response rate, and reducing side effects of ICI; ICI supplements FT by reducing local recurrence, controlling distal metastases, and providing long-term protection. This combinatorial strategy has shown promising results in preclinical study (since 2004) and the clinical trials (since 2011). Understanding the synergy calls for understanding the physics and biology behind the two different therapies with distinctive mechanisms of action. In this review, we introduce different types of energy-based FT by covering the biophysics of tissue-energy interaction and present the immunomodulatory properties of FT. We discuss the basis of cancer immunotherapy with the emphasis on ICI. We examine the approaches researchers have been using and the results from both preclinical models and clinical trials from our exhaustive literature research. Finally, the challenges of the combinatory strategy and opportunities of future research is discussed extensively.
Collapse
Affiliation(s)
- Minhan Jiang
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Steven Fiering
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
- Dartmouth Cancer Center, Dartmouth Geisel School of Medicine and Dartmouth Health, Lebanon, NH, United States
| | - Qi Shao
- Department of Radiology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
13
|
Cuypers ML, Geukens N, Hollevoet K, Declerck P, Dewilde M. Exploring the Fate of Antibody-Encoding pDNA after Intramuscular Electroporation in Mice. Pharmaceutics 2023; 15:pharmaceutics15041160. [PMID: 37111645 PMCID: PMC10146361 DOI: 10.3390/pharmaceutics15041160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/29/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023] Open
Abstract
DNA-based antibody therapy seeks to administer the encoding nucleotide sequence rather than the antibody protein. To further improve the in vivo monoclonal antibody (mAb) expression, a better understanding of what happens after the administration of the encoding plasmid DNA (pDNA) is required. This study reports the quantitative evaluation and localization of the administered pDNA over time and its association with corresponding mRNA levels and systemic protein concentrations. pDNA encoding the murine anti-HER2 4D5 mAb was administered to BALB/c mice via intramuscular injection followed by electroporation. Muscle biopsies and blood samples were taken at different time points (up to 3 months). In muscle, pDNA levels decreased 90% between 24 h and one week post treatment (p < 0.0001). In contrast, mRNA levels remained stable over time. The 4D5 antibody plasma concentrations reached peak levels at week two followed by a slow decrease (50% after 12 weeks, p < 0.0001). Evaluation of pDNA localization revealed that extranuclear pDNA was cleared fast, whereas the nuclear fraction remained relatively stable. This is in line with the observed mRNA and protein levels over time and indicates that only a minor fraction of the administered pDNA is ultimately responsible for the observed systemic mAb levels. In conclusion, this study demonstrates that durable expression is dependent on the nuclear uptake of the pDNA. Therefore, efforts to increase the protein levels upon pDNA-based gene therapy should focus on strategies to increase both cellular entry and migration of the pDNA into the nucleus. The currently applied methodology can be used to guide the design and evaluation of novel plasmid-based vectors or alternative delivery methods in order to achieve a robust and prolonged protein expression.
Collapse
Affiliation(s)
- Marie-Lynn Cuypers
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven-University of Leuven, O&N II Herestraat 49 Box 820, 3000 Leuven, Belgium
| | - Nick Geukens
- PharmAbs-The KU Leuven Antibody Center, KU Leuven-University of Leuven, O&N II Herestraat 49 Box 820, 3000 Leuven, Belgium
| | - Kevin Hollevoet
- PharmAbs-The KU Leuven Antibody Center, KU Leuven-University of Leuven, O&N II Herestraat 49 Box 820, 3000 Leuven, Belgium
| | - Paul Declerck
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven-University of Leuven, O&N II Herestraat 49 Box 820, 3000 Leuven, Belgium
- PharmAbs-The KU Leuven Antibody Center, KU Leuven-University of Leuven, O&N II Herestraat 49 Box 820, 3000 Leuven, Belgium
| | - Maarten Dewilde
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven-University of Leuven, O&N II Herestraat 49 Box 820, 3000 Leuven, Belgium
- PharmAbs-The KU Leuven Antibody Center, KU Leuven-University of Leuven, O&N II Herestraat 49 Box 820, 3000 Leuven, Belgium
| |
Collapse
|
14
|
Lambracht-Washington D, Fu M, Wight-Carter M, Riegel M, Hynan LS, Rosenberg RN. DNA Aβ42 immunization via needle-less Jet injection in mice and rabbits as potential immunotherapy for Alzheimer's disease. J Neurol Sci 2023; 446:120564. [PMID: 36731358 DOI: 10.1016/j.jns.2023.120564] [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: 05/13/2022] [Revised: 01/11/2023] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia found in the elderly and disease progression is associated with accumulation of Amyloid beta 1-42 (Aβ42) in brain. An immune-mediated approach as a preventive intervention to reduce amyloid plaques without causing brain inflammation is highly desirable for future clinical use. Genetic immunization, in which the immunizing agent is DNA encoding Aβ42, has great potential because the immune response to DNA delivered into the skin is generally non-inflammatory, and thus differs quantitatively and qualitatively from immune responses elicited by peptides, which are inflammatory with production of IFNγ and IL-17 cytokines by activated T cells. DNA immunization has historically been proven difficult to apply to larger mammals. A potential barrier to use DNA immunization in large mammals is the method for delivery of the DNA antigen. We tested jet injection in mice and rabbits and found good antibody production and safe immune responses (no inflammatory cytokines). We found significant reduction of amyloid plaques and Aβ peptides in brains of the DNA Aβ42 immunized 3xTg-AD mouse model. This study was designed to optimize DNA delivery for possible testing of the DNA Aβ42 vaccine for AD prevention in a clinical trial.
Collapse
Affiliation(s)
| | - Min Fu
- Department of Neurology, UT Southwestern Medical Center Dallas, TX, USA.
| | - Mary Wight-Carter
- Animal Resource Center, UT Southwestern Medical Center Dallas, TX, USA.
| | - Matthew Riegel
- Animal Resource Center, UT Southwestern Medical Center Dallas, TX, USA; University of Kansas, Lawrence, KS, USA.
| | - Linda S Hynan
- Departments of Population and Data Sciences (Biostatistics) & Psychiatry, UT Southwestern Medical Center Dallas, TX, USA.
| | - Roger N Rosenberg
- Department of Neurology, UT Southwestern Medical Center Dallas, TX, USA.
| |
Collapse
|
15
|
Tumor Radiosensitization by Gene Electrotransfer-Mediated Double Targeting of Tumor Vasculature. Int J Mol Sci 2023; 24:ijms24032755. [PMID: 36769077 PMCID: PMC9917180 DOI: 10.3390/ijms24032755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
Targeting the tumor vasculature through specific endothelial cell markers involved in different signaling pathways represents a promising tool for tumor radiosensitization. Two prominent targets are endoglin (CD105), a transforming growth factor β co-receptor, and the melanoma cell adhesion molecule (CD1046), present also on many tumors. In our recent in vitro study, we constructed and evaluated a plasmid for simultaneous silencing of these two targets. In the current study, our aim was to explore the therapeutic potential of gene electrotransfer-mediated delivery of this new plasmid in vivo, and to elucidate the effects of combined therapy with tumor irradiation. The antitumor effect was evaluated by determination of tumor growth delay and proportion of tumor free mice in the syngeneic murine mammary adenocarcinoma tumor model TS/A. Histological analysis of tumors (vascularization, proliferation, hypoxia, necrosis, apoptosis and infiltration of immune cells) was performed to evaluate the therapeutic mechanisms. Additionally, potential activation of the immune response was evaluated by determining the induction of DNA sensor STING and selected pro-inflammatory cytokines using qRT-PCR. The results point to a significant radiosensitization and a good therapeutic potential of this gene therapy approach in an otherwise radioresistant and immunologically cold TS/A tumor model, making it a promising novel treatment modality for a wide range of tumors.
Collapse
|
16
|
Loo JH, Wang Z, Chong RS. Caveolin-1 in vascular health and glaucoma: A critical vascular regulator and potential therapeutic target. Front Med (Lausanne) 2023; 10:1087123. [PMID: 36760400 PMCID: PMC9902660 DOI: 10.3389/fmed.2023.1087123] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/04/2023] [Indexed: 01/25/2023] Open
Abstract
Caveolin-1 (Cav-1) is an integral scaffolding membrane protein found in most cell types. Cav-1 has been found to contribute significantly to ocular function, with mutations of Cav-1 being associated with a genetic risk of glaucoma development. Raised intraocular pressure (IOP) is a major modifiable risk factor for glaucoma. Cav-1 may be involved in both IOP-dependent and independent mechanisms involving vascular dysregulation. Systemic vascular diseases including hypertension, diabetes and hyperlipidaemia, have been shown to be associated with glaucoma development. Cav-1 is closely interlinked with endothelial nitric oxide synthase pathways that mediate vascular function and prevent cardiovascular diseases. Endothelial nitric oxide synthase and endothelin-1 are key vasoactive molecules expressed in retinal blood vessels that function to autoregulate ocular blood flow (OBF). Disruptions in the homeostasis of OBF have led to a growing concept of impaired neurovascular coupling in glaucoma. The imbalance between perfusion and neuronal stimulation arising from Cav-1 depletion may result in relative ischemia of the optic nerve head and glaucomatous injury. OBF is also governed by circadian variation in IOP and systemic blood pressure (BP). Cav-1 has been shown to influence central BP variability and other circadian rhythms such as the diurnal phagolysosomal digestion of photoreceptor fragments and toxic substrates to maintain ocular health. Overall, the vast implications of Cav-1 on various ocular mechanisms leading to glaucoma suggest a potential for new therapeutics to enhance Cav-1 expression, which has seen success in other neurodegenerative diseases.
Collapse
Affiliation(s)
- Jing Hong Loo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Rachel S. Chong
- Glaucoma Department, Singapore National Eye Center, Singapore, Singapore,Ocular Imaging Department, Singapore Eye Research Institute, Singapore, Singapore,*Correspondence: Rachel S. Chong ✉
| |
Collapse
|
17
|
Malyško-Ptašinskė V, Staigvila G, Novickij V. Invasive and non-invasive electrodes for successful drug and gene delivery in electroporation-based treatments. Front Bioeng Biotechnol 2023; 10:1094968. [PMID: 36727038 PMCID: PMC9885012 DOI: 10.3389/fbioe.2022.1094968] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/28/2022] [Indexed: 01/17/2023] Open
Abstract
Electroporation is an effective physical method for irreversible or reversible permeabilization of plasma membranes of biological cells and is typically used for tissue ablation or targeted drug/DNA delivery into living cells. In the context of cancer treatment, full recovery from an electroporation-based procedure is frequently dependent on the spatial distribution/homogeneity of the electric field in the tissue; therefore, the structure of electrodes/applicators plays an important role. This review focuses on the analysis of electrodes and in silico models used for electroporation in cancer treatment and gene therapy. We have reviewed various invasive and non-invasive electrodes; analyzed the spatial electric field distribution using finite element method analysis; evaluated parametric compatibility, and the pros and cons of application; and summarized options for improvement. Additionally, this review highlights the importance of tissue bioimpedance for accurate treatment planning using numerical modeling and the effects of pulse frequency on tissue conductivity and relative permittivity values.
Collapse
Affiliation(s)
| | - Gediminas Staigvila
- Faculty of Electronics, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Vitalij Novickij
- Faculty of Electronics, Vilnius Gediminas Technical University, Vilnius, Lithuania
- Department of Immunology, State Research Institute Centre of Innovative Medicine, Vilnius, Lithuania
| |
Collapse
|
18
|
Bhandary M, Sales Conniff A, Miranda K, Heller LC. Acute Effects of Intratumor DNA Electrotransfer. Pharmaceutics 2022; 14:pharmaceutics14102097. [PMID: 36297532 PMCID: PMC9611921 DOI: 10.3390/pharmaceutics14102097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/14/2022] Open
Abstract
Intratumor therapeutic DNA electroporation or electrotransfer is in clinical trials in the United States and is under development in many other countries. Acute changes in endogenous gene expression in response to DNA or to pulse application may significantly modulate the therapeutic efficacy of the expressed proteins. Oligonucleotide arrays were used in this study to quantify changes in mRNA expression in B16-F10 mouse melanoma tumors four hours after DNA electrotransfer. The data were subjected to the DAVID v6.8 web server for functional annotation to reveal regulated genes and genetic pathways. Gene ontology analysis revealed several molecular functions related to cytoskeletal remodeling and inflammatory signaling. In B16-F10 cells, F-actin remodeling was confirmed by phalloidin staining in cells that received pulse application alone or in the presence of DNA. Chemokine secretion was confirmed in cells receiving DNA electrotransfer. These results indicate that pulse application alone or in the presence of DNA may modulate the therapeutic efficacy of therapeutic DNA electrotransfer.
Collapse
|
19
|
Jacobs L, Yshii L, Junius S, Geukens N, Liston A, Hollevoet K, Declerck P. Intratumoral DNA-based delivery of checkpoint-inhibiting antibodies and interleukin 12 triggers T cell infiltration and anti-tumor response. Cancer Gene Ther 2022; 29:984-992. [PMID: 34754076 DOI: 10.1038/s41417-021-00403-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 10/08/2021] [Accepted: 10/26/2021] [Indexed: 01/22/2023]
Abstract
To improve the anti-tumor efficacy of immune checkpoint inhibitors, numerous combination therapies are under clinical evaluation, including with IL-12 gene therapy. The current study evaluated the simultaneous delivery of the cytokine and checkpoint-inhibiting antibodies by intratumoral DNA electroporation in mice. In the MC38 tumor model, combined administration of plasmids encoding IL-12 and an anti-PD-1 antibody induced significant anti-tumor responses, yet similar to the monotherapies. When treatment was expanded with a DNA-based anti-CTLA-4 antibody, this triple combination significantly delayed tumor growth compared to IL-12 alone and the combination of anti-PD-1 and anti-CTLA-4 antibodies. Despite low drug plasma concentrations, the triple combination enabled significant abscopal effects in contralateral tumors, which was not the case for the other treatments. The DNA-based immunotherapies increased T cell infiltration in electroporated tumors, especially of CD8+ T cells, and upregulated the expression of CD8+ effector markers. No general immune activation was detected in spleens following either intratumoral treatment. In B16F10 tumors, evaluation of the triple combination was hampered by a high sensitivity to control plasmids. In conclusion, intratumoral gene electrotransfer allowed effective combined delivery of multiple immunotherapeutics. This approach induced responses in treated and contralateral tumors, while limiting systemic drug exposure and potentially detrimental systemic immunological effects.
Collapse
Affiliation(s)
- Liesl Jacobs
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven - University of Leuven, Leuven, Belgium
| | - Lidia Yshii
- Department of Microbiology, Immunology and Transplantation, KU Leuven - University of Leuven, Leuven, Belgium.,VIB Center for Brain and Disease Research, VIB, Leuven, Belgium
| | - Steffie Junius
- Department of Microbiology, Immunology and Transplantation, KU Leuven - University of Leuven, Leuven, Belgium.,VIB Center for Brain and Disease Research, VIB, Leuven, Belgium
| | - Nick Geukens
- PharmAbs - the KU Leuven Antibody Center, KU Leuven - University of Leuven, Leuven, Belgium
| | - Adrian Liston
- Department of Microbiology, Immunology and Transplantation, KU Leuven - University of Leuven, Leuven, Belgium.,VIB Center for Brain and Disease Research, VIB, Leuven, Belgium.,Immunology Programme, Babraham Institute, Cambridge, United Kingdom
| | - Kevin Hollevoet
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven - University of Leuven, Leuven, Belgium. .,PharmAbs - the KU Leuven Antibody Center, KU Leuven - University of Leuven, Leuven, Belgium.
| | - Paul Declerck
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven - University of Leuven, Leuven, Belgium. .,PharmAbs - the KU Leuven Antibody Center, KU Leuven - University of Leuven, Leuven, Belgium.
| |
Collapse
|
20
|
Conforti A, Salvatori E, Lione L, Compagnone M, Pinto E, Shorrock C, Hayward JA, Sun Y, Liang BM, Palombo F, Viscount B, Aurisicchio L. Linear DNA amplicons as a novel cancer vaccine strategy. J Exp Clin Cancer Res 2022; 41:195. [PMID: 35668533 PMCID: PMC9169303 DOI: 10.1186/s13046-022-02402-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/20/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND DNA-based vaccines represent a simple, safe and promising strategy for harnessing the immune system to fight infectious diseases as well as various forms of cancer and thus are considered an important tool in the cancer immunotherapy toolbox. Nonetheless, the manufacture of plasmid DNA vaccines has several drawbacks, including long lead times and the need to remove impurities from bacterial cultures. Here we report the development of polymerase chain reaction (PCR)-produced amplicon expression vectors as DNA vaccines and their in vivo application to elicit antigen-specific immune responses in animal cancer models. METHODS Plasmid DNA and amplicon expression was assessed both in vitro, by Hela cells transfection, and in vivo, by evaluating luciferase expression in wild-type mice through optical imaging. Immunogenicity induced by DNA amplicons was assessed by vaccinating wild-type mice against a tumor-associated antigen, whereas the antitumoral effect of DNA amplicons was evaluated in a murine cancer model in combination with immune-checkpoint inhibitors (ICIs). RESULTS Amplicons encoding tumor-associated-antigens, such as telomerase reverse transcriptase or neoantigens expressed by murine tumor cell lines, were able to elicit antigen-specific immune responses and proved to significantly impact tumor growth when administered in combination with ICIs. CONCLUSIONS These results strongly support the further exploration of the use of PCR-based amplicons as an innovative immunotherapeutic approach to cancer treatment.
Collapse
Affiliation(s)
- Antonella Conforti
- Takis, Via Castel Romano 100, 00128 Rome, Italy
- Evvivax, Via Castel Romano 100, 00128 Rome, Italy
| | | | - Lucia Lione
- Takis, Via Castel Romano 100, 00128 Rome, Italy
| | | | | | - Clay Shorrock
- Applied DNA Sciences, 50 Health Sciences Drive, Stony Brook, NY 11790 USA
| | - James A. Hayward
- Applied DNA Sciences, 50 Health Sciences Drive, Stony Brook, NY 11790 USA
| | - Yuhua Sun
- Applied DNA Sciences, 50 Health Sciences Drive, Stony Brook, NY 11790 USA
| | - Ben Minghwa Liang
- Applied DNA Sciences, 50 Health Sciences Drive, Stony Brook, NY 11790 USA
| | - Fabio Palombo
- Takis, Via Castel Romano 100, 00128 Rome, Italy
- Neomatrix, Via Castel Romano 100, 00128 Rome, Italy
| | - Brian Viscount
- Applied DNA Sciences, 50 Health Sciences Drive, Stony Brook, NY 11790 USA
| | - Luigi Aurisicchio
- Takis, Via Castel Romano 100, 00128 Rome, Italy
- Evvivax, Via Castel Romano 100, 00128 Rome, Italy
- Neomatrix, Via Castel Romano 100, 00128 Rome, Italy
| |
Collapse
|
21
|
Sachdev S, Potočnik T, Rems L, Miklavčič D. Revisiting the role of pulsed electric fields in overcoming the barriers to in vivo gene electrotransfer. Bioelectrochemistry 2022; 144:107994. [PMID: 34930678 DOI: 10.1016/j.bioelechem.2021.107994] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/15/2021] [Accepted: 11/02/2021] [Indexed: 12/21/2022]
Abstract
Gene therapies are revolutionizing medicine by providing a way to cure hitherto incurable diseases. The scientific and technological advances have enabled the first gene therapies to become clinically approved. In addition, with the ongoing COVID-19 pandemic, we are witnessing record speeds in the development and distribution of gene-based vaccines. For gene therapy to take effect, the therapeutic nucleic acids (RNA or DNA) need to overcome several barriers before they can execute their function of producing a protein or silencing a defective or overexpressing gene. This includes the barriers of the interstitium, the cell membrane, the cytoplasmic barriers and (in case of DNA) the nuclear envelope. Gene electrotransfer (GET), i.e., transfection by means of pulsed electric fields, is a non-viral technique that can overcome these barriers in a safe and effective manner. GET has reached the clinical stage of investigations where it is currently being evaluated for its therapeutic benefits across a wide variety of indications. In this review, we formalize our current understanding of GET from a biophysical perspective and critically discuss the mechanisms by which electric field can aid in overcoming the barriers. We also identify the gaps in knowledge that are hindering optimization of GET in vivo.
Collapse
Affiliation(s)
- Shaurya Sachdev
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, 1000 Ljubljana, Slovenia
| | - Tjaša Potočnik
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, 1000 Ljubljana, Slovenia
| | - Lea Rems
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, 1000 Ljubljana, Slovenia
| | - Damijan Miklavčič
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, 1000 Ljubljana, Slovenia.
| |
Collapse
|
22
|
Kardani K, Milani A, Bolhassani A. Gene delivery in adherent and suspension cells using the combined physical methods. Cytotechnology 2022; 74:245-257. [PMID: 35464169 PMCID: PMC8975990 DOI: 10.1007/s10616-022-00524-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 01/25/2022] [Indexed: 11/24/2022] Open
Abstract
Physical methods are widely utilized to deliver nucleic acids into cells such as electro-transfection or heat shock. An efficient gene electro-transfection requires the best conditions including voltage, the pulse length or number, buffer, incubation time and DNA form. In this study, the delivery of pEGFP-N1 vector into two adherent cell lines (HEK-293 T and COS-7) with the same origin (epithelial cells), and also mouse bone marrow-derived dendritic cells (DCs) was evaluated using electroporation under different conditions alone and along with heat treatment. Our data showed that the highest green fluorescent protein (GFP) expression in HEK-293 T and COS-7 cells was observed in serum-free RPMI cell culture medium as electroporation buffer, voltage (200 V), the pulse number (2), the pulse length (15 ms), the circular form of DNA, and 48 h after electro-transfection. In addition, the highest GFP expression in DCs was detected in serum-free RPMI, voltage (300 V), the pulse number (1), the pulse length (5 ms), and 48 h after electro-transfection. The use of sucrose as electroporation buffer, the pulse number (2), and the pulse length (25 ms) led to further cytotoxicity and lower transfection in HEK293T and COS-7 cells than other conditions. Moreover, the high voltage (700 V) increased the cell cytotoxicity, and decreased electro-transfection efficiency in DCs. On the other hand, the best conditions of electroporation along with heat treatment could significantly augment the transfection efficiency in all the cells. These data will be useful for gene delivery in other cells with the same properties using physical methods. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-022-00524-4.
Collapse
Affiliation(s)
- Kimia Kardani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Alireza Milani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
23
|
Sales Conniff A, Encalada G, Patel S, Bhandary M, Al-Takrouri F, Heller L. Poly(I:C) transfection induces a pro-inflammatory cascade in murine mammary carcinoma and fibrosarcoma cells. RNA Biol 2022; 19:841-851. [PMID: 35737804 PMCID: PMC9235898 DOI: 10.1080/15476286.2022.2084861] [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] [Indexed: 11/12/2022] Open
Abstract
Germline-encoded pattern recognition receptors [PRRs] in mammalian cells function in the detection of molecular patterns associated with pathogen invasion or cellular damage. A PRR subset is activated by the atypical presence and location of double-stranded RNA [dsRNA] or its synthetic analogue polyinosinic-polycytidylic acid [poly(I:C)], triggering pro-inflammatory signalling and death in many cell types. Poly(I:C) has been tested as a sole or combination cancer therapy in preclinical studies and clinical trials. The purpose of this study was to evaluate the effects of poly(I:C) transfection via electroporation on cell lines from a cancer of epithelial origin, 4T1 mammary carcinoma, and a cancer of mesenchymal origin, WEHI 164 fibrosarcoma. The effects of the poly(I:C) delivery on cell metabolism implicate the induction of cell death. A pro-inflammatory response was demonstrated by mRNA upregulation and the secretion of Type I interferon and several cytokines and chemokines. The mRNAs of dsRNA sensor DExD/H-box helicase 58/retinoic acid-inducible gene I protein [Ddx58/RIG-I] and sensor/co-sensor DEAH-box helicase 9 [Dhx9] were not regulated, but the mRNAs of RNA sensors toll-like receptor 3 [TLR3], interferon-induced with helicase C domain 1/melanoma differentiation-associated protein 5 [Ifih1/MDA5] and Z-DNA binding protein 1 [Zbp1] and co-sensors DEAD (Asp-Glu-Ala-Asp) box polypeptide 60 [Ddx60] and interferon-inducible protein 204 [Ifi204] were upregulated in both cell lines. The mRNAs encoding signalling pathways components were present or upregulated in both cell types. These data demonstrate that RNA sensing effects can be amplified by electroporation delivery, potentially expanding the practicality of this immunotherapeutic approach.
Collapse
Affiliation(s)
- A Sales Conniff
- Department of Medical Engineering, University of South Florida, Tampa, FL, USA
| | - G Encalada
- Department of Medical Engineering, University of South Florida, Tampa, FL, USA
| | - S Patel
- Department of Medical Engineering, University of South Florida, Tampa, FL, USA
| | - M Bhandary
- Department of Medical Engineering, University of South Florida, Tampa, FL, USA
| | - F Al-Takrouri
- Department of Medical Engineering, University of South Florida, Tampa, FL, USA
| | - L Heller
- Department of Medical Engineering, University of South Florida, Tampa, FL, USA
| |
Collapse
|
24
|
Arora S, Sharma D, Layek B, Singh J. A Review of Brain-Targeted Nonviral Gene-Based Therapies for the Treatment of Alzheimer's Disease. Mol Pharm 2021; 18:4237-4255. [PMID: 34705472 DOI: 10.1021/acs.molpharmaceut.1c00611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diseases of the central nervous system (CNS) are difficult to treat owing to the complexity of the brain and the presence of a natural blood-brain-barrier (BBB). Alzheimer's disease (AD) is one of the major progressive and currently incurable neurodegenerative disorders of the CNS, which accounts for 60-80% of cases of dementia. The pathophysiology of AD involves the accumulation of amyloid beta (Aβ) plaques and neurofibrillary tangles (NFTs) in the brain. Additionally, synaptic loss and imbalance of neuronal signaling molecules are characterized as important markers of AD. Existing treatments of AD help in the management of its symptoms and aim toward the maintenance of cognitive functions, behavior, and attenuation of gradual memory loss. Over the past decade, nonviral gene therapy has attracted increasing interest due to its various advantages over its viral counterparts. Moreover, advancements in nonviral gene technology have led to their increasing contributions in clinical trials. However, brain-targeted nonviral gene delivery vectors come across various extracellular and intracellular barriers, limiting their ability to transfer the therapeutic gene into the target cells. Chief barriers to nonviral gene therapy have been discussed briefly in this review. We have also highlighted the rapid advancement of several nonviral gene therapies for AD, which are broadly categorized into physical and chemical methods. These methods aim to modulate Aβ, beta-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1), apolipoprotein E, or neurotrophic factors' expression in the CNS. Overall, this review discusses challenges and recent advancements of nonviral gene therapy for AD.
Collapse
Affiliation(s)
- Sanjay Arora
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Divya Sharma
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Buddhadev Layek
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| |
Collapse
|
25
|
Vermeire G, De Smidt E, Geukens N, Williams JA, Declerck P, Hollevoet K. Improved Potency and Safety of DNA-Encoded Antibody Therapeutics Through Plasmid Backbone and Expression Cassette Engineering. Hum Gene Ther 2021; 32:1200-1209. [PMID: 34482757 DOI: 10.1089/hum.2021.105] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
DNA-encoded delivery of antibodies presents a labor- and cost-effective alternative to conventional antibody therapeutics. This study aims to improve the potency and safety of this approach by evaluating various plasmid backbones and expression cassettes. In vitro, antibody levels consistently improved with decreasing sizes of backbone, ranging from conventional to minimal. In vivo, following intramuscular electrotransfer in mice, the correlation was less consistent. While the largest conventional plasmid (10.2 kb) gave the lowest monoclonal antibody (mAb) levels, a regular conventional plasmid (8.6 kb) demonstrated similar levels as a minimal Nanoplasmid (6.8 kb). A reduction in size beyond a standard conventional backbone thus did not improve mAb levels in vivo. Cassette modifications, such as swapping antibody chain order or use of two versus a single encoding plasmid, significantly increased antibody expression in vitro, but failed to translate in vivo. Conversely, a significant improvement in vivo but not in vitro was found with a set of muscle-specific promoters, of which a newly engineered variant gave roughly 1.5- to 2-fold higher plasma antibody concentrations than the ubiquitous CAG promoter. In conclusion, despite the limited translation between in vitro and in vivo, we identified various clinically relevant improvements to our DNA-based antibody platform, both in potency and biosafety.
Collapse
Affiliation(s)
- Giles Vermeire
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven-University of Leuven, Leuven, Belgium
| | - Elien De Smidt
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven-University of Leuven, Leuven, Belgium.,PharmAbs, the KU Leuven Antibody Center-University of Leuven, Leuven, Belgium
| | - Nick Geukens
- PharmAbs, the KU Leuven Antibody Center-University of Leuven, Leuven, Belgium
| | | | - Paul Declerck
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven-University of Leuven, Leuven, Belgium.,PharmAbs, the KU Leuven Antibody Center-University of Leuven, Leuven, Belgium
| | - Kevin Hollevoet
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven-University of Leuven, Leuven, Belgium.,PharmAbs, the KU Leuven Antibody Center-University of Leuven, Leuven, Belgium
| |
Collapse
|
26
|
Tesse A, André FM, Ragot T. Aluminum particles generated during millisecond electric pulse application enhance adenovirus-mediated gene transfer in L929 cells. Sci Rep 2021; 11:17725. [PMID: 34489497 PMCID: PMC8421418 DOI: 10.1038/s41598-021-96781-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 08/04/2021] [Indexed: 11/20/2022] Open
Abstract
Gene electrotransfer is an attractive method of non-viral gene delivery. However, the mechanism of DNA penetration across the plasma membrane is widely discussed. To explore this process for even larger structures, like viruses, we applied various combinations of short/long and high/low-amplitude electric pulses to L929 cells, mixed with a human adenovirus vector expressing GFP. We observed a transgene expression increase, both in the number of GFP-converted cells and GFP levels, when we added a low-voltage/millisecond-pulse treatment to the adenovirus/cell mixture. This increase, reflecting enhanced virus penetration, was proportional to the applied electric field amplitude and pulse number, but was not associated with membrane permeabilization, nor to direct cell modifications. We demonstrated that this effect is mainly due to adenovirus particle interactions with aggregated aluminum particles released from energized electrodes. Indeed, after centrifugation of the pulsed viral suspension and later on addition to cells, the activity was found mainly associated with the aluminum aggregates concentrated in the lower fraction and was proportional to generated quantities. Overall, this work focused on the use of electrotransfer to facilitate the adenovirus entry into cell, demonstrating that modifications of the penetrating agent can be more important than modifications of the target cell for transfer efficacy.
Collapse
Affiliation(s)
- Angela Tesse
- Université de Nantes, CNRS, INSERM, l'institut du thorax, 8 quai Moncousu, F-44000, Nantes, France
| | - Franck M André
- CNRS, Institut Gustave Roussy, Université Paris-Saclay, Aspects métaboliques et systémiques de l'oncogenèse pour de nouvelles approches thérapeutiques, UMR 9018, 114 rue Edouard Vaillant, F-94805, Villejuif, France
| | - Thierry Ragot
- CNRS, Institut Gustave Roussy, Université Paris-Saclay, Aspects métaboliques et systémiques de l'oncogenèse pour de nouvelles approches thérapeutiques, UMR 9018, 114 rue Edouard Vaillant, F-94805, Villejuif, France.
| |
Collapse
|
27
|
Zamponi M, Petrella R, Mollica PA. Picosecond Pulsed Electric Fields and Promise in Neurodegeneration Research. Bioelectricity 2021. [DOI: 10.1089/bioe.2021.0005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Martina Zamponi
- School of Medical Diagnostic and Translational Sciences, Old Dominion University, Norfolk, Virginia, USA
- Biomedical Engineering Institute, Old Dominion University, Norfolk, Virginia, USA
| | - Ross Petrella
- Joint Department of Biomedical Engineering at the University of North Carolina and North Carolina State University, Raleigh, North Carolina, USA
| | - Peter A. Mollica
- School of Medical Diagnostic and Translational Sciences, Old Dominion University, Norfolk, Virginia, USA
- Molecular Diagnostics Laboratory, Sentara Norfolk General Hospital, Norfolk, Virginia, USA
| |
Collapse
|
28
|
Mulia GE, Picanço-Castro V, Stavrou EF, Athanassiadou A, Figueiredo ML. Advances in the Development and the Applications of Non-viral, Episomal Vectors for Gene Therapy. Hum Gene Ther 2021; 32:1076-1095. [PMID: 34348480 PMCID: PMC8819515 DOI: 10.1089/hum.2020.310] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Nonviral and nonintegrating episomal vectors are reemerging as a valid, alternative technology to integrating viral vectors for gene therapy, due to their more favorable safety profile, significantly lower risk for insertional mutagenesis, and a lesser potential for innate immune reactions, in addition to their low production cost. Over the past few years, attempts have been made to generate highly functional nonviral vectors that display long-term maintenance within cells and promote more sustained gene expression relative to conventional plasmids. Extensive research into the parameters that stabilize the episomal DNA within dividing and nondividing cells has shed light into the genetic and epigenetic mechanisms that govern replication and transcription of episomal DNA within a mammalian nucleus in long-term cell culture. Episomal vectors based on scaffold/matrix attachment regions (S/MARs) do not integrate into the genomic DNA and address the serious problem of plasmid loss during mitosis by providing mitotic stability to established plasmids, which results in long-term transfection and transgene expression. The inclusion, in such vectors, of an origin of replication—initiation region—from the human genome has greatly enhanced their performance in primary cell culture. A number of vectors that function as episomes have arisen, which are either devoid or depleted of harmful CpG sequences and bacterial genes, and their effectiveness, as well as that of nonintegrating viral episomes, is enhanced when combined with S/MAR elements. As a result of these advances, an “S/MAR technology” has emerged for the production of efficient episomal vectors. Significant research continues in this field and innovations, in combination with promising systems based on nanoparticles and potentially combined with physical delivery methods, will enable the generation of optimized systems with scale-up and clinical application suitability utilizing episomal vectors.
Collapse
Affiliation(s)
- Grace E Mulia
- Purdue University, Basic Medical Sciences, West Lafayette, Indiana, United States;
| | - Virginia Picanço-Castro
- University of Sao Paulo Faculty of Medicine of Ribeirao Preto, 54539, Center for Cell-based Therapy, Ribeirao Preto, São Paulo, Brazil;
| | - Eleana F Stavrou
- University of Patras, Department of General Biology, Patras, Greece;
| | - Aglaia- Athanassiadou
- University of Patras Medical School, General Biology, Asklepiou str, University Campus, Rion Patras, Greece, 26504;
| | - Marxa L Figueiredo
- Purdue University, Basic Medical Sciences, 625 Harrison St., LYNN 2177, West Lafayette, Indiana, United States, 47907;
| |
Collapse
|
29
|
Tranberg KG. Local Destruction of Tumors and Systemic Immune Effects. Front Oncol 2021; 11:708810. [PMID: 34307177 PMCID: PMC8298109 DOI: 10.3389/fonc.2021.708810] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/23/2021] [Indexed: 12/22/2022] Open
Abstract
Current immune-based therapies signify a major advancement in cancer therapy; yet, they are not effective in the majority of patients. Physically based local destruction techniques have been shown to induce immunologic effects and are increasingly used in order to improve the outcome of immunotherapies. The various local destruction methods have different modes of action and there is considerable variation between the different techniques with respect to the ability and frequency to create a systemic anti-tumor immunologic effect. Since the abscopal effect is considered to be the best indicator of a relevant immunologic effect, the present review focused on the tissue changes associated with this effect in order to find determinants for a strong immunologic response, both when local destruction is used alone and combined with immunotherapy. In addition to the T cell-inflammation that was induced by all methods, the analysis indicated that it was important for an optimal outcome that the released antigens were not destroyed, tumor cell death was necrotic and tumor tissue perfusion was at least partially preserved allowing for antigen presentation, immune cell trafficking and reduction of hypoxia. Local treatment with controlled low level hyperthermia met these requisites and was especially prone to result in abscopal immune activity on its own.
Collapse
|
30
|
Eusébio D, Neves AR, Costa D, Biswas S, Alves G, Cui Z, Sousa Â. Methods to improve the immunogenicity of plasmid DNA vaccines. Drug Discov Today 2021; 26:2575-2592. [PMID: 34214667 DOI: 10.1016/j.drudis.2021.06.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/31/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023]
Abstract
DNA vaccines have emerged as innovative approaches that have great potential to overcome the limitations of current conventional vaccines. Plasmid DNA vaccines are often safer than other vaccines because they carry only antigen genetic information, are more stable and easier to produce, and can stimulate both humoral and cellular immune responses. Although the results of ongoing clinical trials are very promising, some limitations compromise the immunogenicity of these vaccines. Thus, this review describes different strategies that can be explored to improve the immunogenicity of plasmid DNA vaccines, including the optimization of the plasmid vector backbone, the use of different methods for vaccine delivery, the use of alternative administration routes and the inclusion of adjuvants. In combination, these improvements could lead to the successful clinical use of plasmid DNA vaccines.
Collapse
Affiliation(s)
- Dalinda Eusébio
- CICS-UBI - Health Science Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ana R Neves
- CICS-UBI - Health Science Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Diana Costa
- CICS-UBI - Health Science Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Swati Biswas
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Shameerpet, Hyderabad 500078, Telangana, India
| | - Gilberto Alves
- CICS-UBI - Health Science Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Zhengrong Cui
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX 78712, USA
| | - Ângela Sousa
- CICS-UBI - Health Science Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal.
| |
Collapse
|
31
|
Brloznik M, Boc N, Cemazar M, Sersa G, Bosnjak M, Brezar SK, Pavlin D. Tumor perfusion evaluation using dynamic contrast-enhanced ultrasound after electrochemotherapy and IL-12 plasmid electrotransfer in murine melanoma. Sci Rep 2021; 11:13446. [PMID: 34188103 PMCID: PMC8242003 DOI: 10.1038/s41598-021-92820-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 05/24/2021] [Indexed: 11/09/2022] Open
Abstract
Electrochemotherapy with bleomycin (ECT BLM) is an effective antitumor treatment already used in clinical oncology. However, ECT alone is still considered a local antitumor therapy because it cannot induce systemic immunity. When combined with adjuvant gene electrotransfer of plasmid DNA encoding IL-12 (GET pIL-12), the combined therapy leads to a systemic effect on untreated tumors and distant metastases. Although the antitumor efficacy of both therapies alone or in combination has been demonstrated at both preclinical and clinical levels, data on the predictors of efficacy of the treatments are still lacking. Herein, we evaluated the results of dynamic contrast-enhanced ultrasound (DCE-US) as a predictive factor for ECT BLM and GET pIL-12 in murine melanoma. Melanoma B16F10 tumors grown in female C57Bl/6NCrl mice were treated with GET pIL-12 and ECT BLM. Immediately after therapy, 6 h and 1, 3, 7 and 10 days later, tumors were examined by DCE-US. Statistical analysis was performed to inspect the correlation between tumor doubling time (DT) and DCE-US measurements using semilinear regression models and Bland-Altman plots. Therapeutic groups in which DCE-US showed reduced tumor perfusion had longer tumor DTs. It was confirmed that the DCE-US parameter peak enhancement (PE), reflecting relative blood volume, had predictive value for the outcome of therapy: larger PE correlated with shorter DT. In addition, perfusion heterogeneity was also associated with outcome: tumors that had more heterogeneous perfusion had faster growth, i.e., shorter DTs. This study demonstrates that DCE-US can be used as a method to predict the efficacy of electroporation-based treatment.
Collapse
Affiliation(s)
- Maja Brloznik
- Clinic for Small Animals, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, Ljubljana, Slovenia
| | - Nina Boc
- Institute of Oncology Ljubljana, Zaloška 2, Ljubljana, Slovenia
| | - Maja Cemazar
- Institute of Oncology Ljubljana, Zaloška 2, Ljubljana, Slovenia.,Faculty of Health Sciences, University of Primorska, Polje 42, Izola, Slovenia
| | - Gregor Sersa
- Institute of Oncology Ljubljana, Zaloška 2, Ljubljana, Slovenia.,Faculty of Health Sciences, University of Ljubljana, Zdravstvena 5, Ljubljana, Slovenia
| | - Masa Bosnjak
- Institute of Oncology Ljubljana, Zaloška 2, Ljubljana, Slovenia
| | - Simona Kranjc Brezar
- Institute of Oncology Ljubljana, Zaloška 2, Ljubljana, Slovenia. .,Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana, Slovenia.
| | - Darja Pavlin
- Clinic for Small Animals, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, Ljubljana, Slovenia.
| |
Collapse
|
32
|
Kranjc M, Kranjc Brezar S, Serša G, Miklavčič D. Contactless delivery of plasmid encoding EGFP in vivo by high-intensity pulsed electromagnetic field. Bioelectrochemistry 2021; 141:107847. [PMID: 34058542 DOI: 10.1016/j.bioelechem.2021.107847] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 05/14/2021] [Accepted: 05/18/2021] [Indexed: 11/28/2022]
Abstract
High-Intensity Pulsed Electromagnetic Fields (HI-PEMF) treatment is an emerging noninvasive and contactless alternative to conventional electroporation, since the electric field inside the tissue is induced remotely by external pulsed magnetic field. Recently, HI-PEMF was applied for delivering siRNA molecules to silence enhanced green fluorescent protein (EGFP) in tumors in vivo. Still, delivered siRNA molecules were 21 base pairs long, which is 200-times smaller compared to nucleic acids such as plasmid DNA (pDNA) that are delivered in gene therapies to various targets to generate therapeutic effect. In our study, we demonstrate the use HI-PEMF treatment as a feasible noninvasive approach to achieve in vivo transfection by enabling the transport of larger molecules such as pDNA encoding EGFP into muscle and skin. We obtained a long-term expression of EGFP in the muscle and skin after HI-PEMF, in some mice even up to 230 days and up to 190 days, respectively. Histological analysis showed significantly less infiltration of inflammatory mononuclear cells in muscle tissue after the delivery of pEGFP using HI-PEMF compared to conventional gene electrotransfer. Furthermore, the antitumor effectiveness using HI-PEMF for electrotransfer of therapeutic plasmid, i.e., silencing MCAM was demonstrated. In conclusion, feasibility of HI-PEMF was demonstrated for transfection of different tissues (muscle, skin, tumor) and could have great potential in gene therapy and in DNA vaccination.
Collapse
Affiliation(s)
- Matej Kranjc
- University of Ljubljana, Faculty of Electrical Engineering, Trzaska 25, SI-1000 Ljubljana, Slovenia
| | - Simona Kranjc Brezar
- Institute of Oncology Ljubljana, Department of Experimental Oncology, Zaloska 2, SI-1000 Ljubljana, Slovenia; University of Ljubljana, Faculty of Medicine, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| | - Gregor Serša
- Institute of Oncology Ljubljana, Department of Experimental Oncology, Zaloska 2, SI-1000 Ljubljana, Slovenia; University of Ljubljana, Faculty of Health Sciences, Zdravstvena pot 5, SI - 1000 Ljubljana, Slovenia
| | - Damijan Miklavčič
- University of Ljubljana, Faculty of Electrical Engineering, Trzaska 25, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
33
|
Savarin M, Kamensek U, Znidar K, Todorovic V, Sersa G, Cemazar M. Evaluation of a Novel Plasmid for Simultaneous Gene Electrotransfer-Mediated Silencing of CD105 and CD146 in Combination with Irradiation. Int J Mol Sci 2021; 22:ijms22063069. [PMID: 33802812 PMCID: PMC8002395 DOI: 10.3390/ijms22063069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/12/2021] [Accepted: 03/13/2021] [Indexed: 12/12/2022] Open
Abstract
Targeting tumor vasculature through specific endothelial cell markers represents a promising approach for cancer treatment. Here our aim was to construct an antibiotic resistance gene-free plasmid encoding shRNAs to simultaneously target two endothelial cell markers, CD105 and CD146, and to test its functionality and therapeutic potential in vitro when delivered by gene electrotransfer (GET) and combined with irradiation (IR). Functionality of the plasmid was evaluated by determining the silencing of the targeted genes using qRT-PCR. Antiproliferative and antiangiogenic effects were determined by the cytotoxicity assay tube formation assay and wound healing assay in murine endothelial cells 2H-11. The functionality of the plasmid construct was also evaluated in malignant melanoma tumor cell line B16F10. Additionally, potential activation of immune response was measured by induction of DNA sensor STING and proinflammatory cytokines by qRT-PCR in endothelial cells 2H-11. We demonstrated that the plasmid construction was successful and can efficiently silence the expression of the two targeted genes. As a consequence of silencing, reduced migration rate and angiogenic potential was confirmed in 2H-11 endothelial cells. Furthermore, induction of DNA sensor STING and proinflammatory cytokines were determined, which could add to the therapeutic effectiveness when used in vivo. To conclude, we successfully constructed a novel plasmid DNA with two shRNAs, which holds a great promise for further in vivo testing.
Collapse
Affiliation(s)
- Monika Savarin
- Department of Experimental Oncology, Institute of Oncology Ljubljana, 1000 Ljubljana, Slovenia; (U.K.); (K.Z.); (V.T.); (G.S.)
- Correspondence: (M.S.); (M.C.)
| | - Urska Kamensek
- Department of Experimental Oncology, Institute of Oncology Ljubljana, 1000 Ljubljana, Slovenia; (U.K.); (K.Z.); (V.T.); (G.S.)
- Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Katarina Znidar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, 1000 Ljubljana, Slovenia; (U.K.); (K.Z.); (V.T.); (G.S.)
| | - Vesna Todorovic
- Department of Experimental Oncology, Institute of Oncology Ljubljana, 1000 Ljubljana, Slovenia; (U.K.); (K.Z.); (V.T.); (G.S.)
| | - Gregor Sersa
- Department of Experimental Oncology, Institute of Oncology Ljubljana, 1000 Ljubljana, Slovenia; (U.K.); (K.Z.); (V.T.); (G.S.)
- Faculty of Health Sciences, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, 1000 Ljubljana, Slovenia; (U.K.); (K.Z.); (V.T.); (G.S.)
- Faculty of Health Sciences, University of Primorska, 6310 Izola, Slovenia
- Correspondence: (M.S.); (M.C.)
| |
Collapse
|
34
|
Eliseeva SI, Knowlden ZA, Lester GMS, Dean DA, Georas SN, Chapman TJ. Changes in lung immune cell infiltrates after electric field treatment in mice. Sci Rep 2021; 11:1453. [PMID: 33446928 PMCID: PMC7809414 DOI: 10.1038/s41598-021-81174-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 12/23/2020] [Indexed: 01/13/2023] Open
Abstract
Exogenous electric fields are currently used in human therapy in a number of contexts. Interestingly, electric fields have also been shown to alter migration and function of immune cells, suggesting the potential for electric field-based immune therapy. Little is known as to the effect of electric field treatment (EFT) on the lung. To determine if EFT associates with changes in lung immune cell infiltration, we used a mouse model with varying methods of EFT application and measured cells and soluble mediators using flow cytometry and cytokine/chemokine multiplex. EFT was associated with a transient increase in lung neutrophils and decrease in eosinophils in naïve mice within 2 h of treatment, accompanied by an increase in IL-6 levels. In order to test whether EFT could alter eosinophil/neutrophil recruitment in a relevant disease model, a mouse model of allergic airway inflammation was used. Four EFT doses in allergen-sensitized mice resulted in increased neutrophil and reduced eosinophil infiltrates following allergen challenge, suggesting a durable change in inflammation by EFT. Mice with allergic inflammation were analyzed by flexiVent for measures of lung function. EFT-treated mice had increased inspiratory capacity and other measures of lung function were not diminished. These data suggest EFT may be used to manipulate immune cell infiltration in the lung without affecting lung function.
Collapse
Affiliation(s)
- Sophia I Eliseeva
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Zackery A Knowlden
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Gillian MSchiralli Lester
- Departments of Pediatrics, Biomedical Engineering, and Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - David A Dean
- Departments of Pediatrics, Biomedical Engineering, and Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Steve N Georas
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Timothy J Chapman
- Center for Infectious Diseases and Immunity, Rochester General Hospital Research Institute, Rochester Regional Health, 1425 Portland Ave, Room 403, Rochester, NY, 14621, USA.
| |
Collapse
|
35
|
Heller R, Shi G. Controlled Delivery of Plasmid DNA to Melanoma Tumors by Gene Electrotransfer. Methods Mol Biol 2021; 2265:635-644. [PMID: 33704744 DOI: 10.1007/978-1-0716-1205-7_43] [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] [Indexed: 03/24/2023]
Abstract
Gene electrotransfer (GET) is a reliable and effective physical method for in vivo delivery of plasmid DNA (pDNA). Several preclinical and clinical studies have utilized GET to deliver plasmids encoding immune stimulating genes for treatment of melanoma and other tumor types. Intratumor delivery of plasmids encoding cytokines directly to tumors can induce not only a local immune response, but a systemic one as well. To obtain an effective immune response, it is critical to achieve the appropriate expression pattern of the delivered transgene. Expression pattern (levels and kinetics) can be modified by manipulating the electrotransfer parameters. These parameters include the tissue target and the electric pulse parameters of pulse width, electric field, and pulse number. We have found that to induce a robust immune response, we needed only low to moderately elevated expression levels compared to controls. When developing a therapeutic protocol, it is important to establish what expression profile will enable the appropriate response. In this chapter we describe how to determine the appropriate GET protocol to achieve the expression profile that can result in the desired clinical response.
Collapse
Affiliation(s)
- Richard Heller
- Department of Medical Engineering, Colleges of Medicine and Engineering, University of South Florida, Tampa, FL, USA.
| | - Guilan Shi
- Department of Medical Engineering, Colleges of Medicine and Engineering, University of South Florida, Tampa, FL, USA
| |
Collapse
|
36
|
Heller LC, Heller R. Gene Electrotransfer. ELECTROPORATION IN VETERINARY ONCOLOGY PRACTICE 2021:219-234. [DOI: 10.1007/978-3-030-80668-2_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
37
|
Oxidative Effects during Irreversible Electroporation of Melanoma Cells-In Vitro Study. Molecules 2020; 26:molecules26010154. [PMID: 33396317 PMCID: PMC7796376 DOI: 10.3390/molecules26010154] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/15/2020] [Accepted: 12/28/2020] [Indexed: 12/14/2022] Open
Abstract
Irreversible electroporation (IRE) is today used as an alternative to surgery for the excision of cancer lesions. This study aimed to investigate the oxidative and cytotoxic effects the cells undergo during irreversible electroporation using IRE protocols. To do so, we used IRE-inducing pulsed electric fields (PEFs) (eight pulses of 0.1 ms duration and 2-4 kV/cm intensity) and compared their effects to those of PEFs of intensities below the electroporation threshold (eight pulses, 0.1 ms, 0.2-0.4 kV/cm) and the PEFs involving elongated pulses (eight pulses, 10 ms, 0.2-0.4 kV/cm). Next, to follow the morphology of the melanoma cell membranes after treatment with the PEFs, we analyzed the permeability and integrity of their membranes and analyzed the radical oxygen species (ROS) bursts and the membrane lipids' oxidation. Our data showed that IRE-induced high cytotoxic effect is associated both with irreversible cell membrane disruption and ROS-associated oxidation, which is occurrent also in the low electric field range. It was shown that the viability of melanoma cells characterized by similar ROS content and lipid membrane oxidation after PEF treatment depends on the integrity of the membrane system. Namely, when the effects of the PEF on the membrane are reversible, aside from the high level of ROS and membrane oxidation, the cell does not undergo cell death.
Collapse
|
38
|
Wang L, Chang CC, Sylvers J, Yuan F. A statistical framework for determination of minimal plasmid copy number required for transgene expression in mammalian cells. Bioelectrochemistry 2020; 138:107731. [PMID: 33434786 DOI: 10.1016/j.bioelechem.2020.107731] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023]
Abstract
Plasmid DNA (pDNA) has been widely used for non-viral gene delivery. After pDNA molecules enter a mammalian cell, they may be trapped in subcellular structures or degraded by nucleases. Only a fraction of them can function as templates for transcription in the nucleus. Thus, an important question is, what is the minimal amount of pDNA molecules that need to be delivered into a cell for transgene expression? At present, it is technically a challenge to experimentally answer the question. To this end, we developed a statistical framework to establish the relationship between two experimentally quantifiable factors - average copy number of pDNA per cell among a group of cells after transfection and percent of the cells with transgene expression. The framework was applied to the analysis of electrotransfection under different experimental conditions in vitro. We experimentally varied the average copy number per cell and the electrotransfection efficiency through changes in extracellular pDNA dose, electric field strength, and pulse number. The experimental data could be explained or predicted quantitatively by the statistical framework. Based on the data and the framework, we could predict that the minimal number of pDNA molecules in the nucleus for transgene expression was on the order of 10. Although the prediction was dependent on the cell and experimental conditions used in the study, the framework may be generally applied to analysis of non-viral gene delivery.
Collapse
Affiliation(s)
- Liangli Wang
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Chun-Chi Chang
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Justin Sylvers
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Fan Yuan
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA.
| |
Collapse
|
39
|
Electroporation outperforms in vivo-jetPEI for intratumoral DNA-based reporter gene transfer. Sci Rep 2020; 10:19532. [PMID: 33177564 PMCID: PMC7659317 DOI: 10.1038/s41598-020-75206-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/30/2020] [Indexed: 01/29/2023] Open
Abstract
Intratumoral delivery of drug-encoding plasmid DNA (pDNA) enables localised in vivo expression of biological drugs, offering an attractive alternative to conventional protein treatment. However, this requires physical or chemical methods to enhance the low transfection efficiency of naked pDNA. Electroporation and complexation with the polycation in vivo-jetPEI are both evaluated in the clinic for intratumoral pDNA delivery, but lack head-to-head comparison. This study therefore compared both methods for intratumoral DNA-based reporter gene transfer in a subcutaneous mouse tumour model. Intratumoral electroporation resulted in strong reporter expression that was restricted to the tumour area and persisted for at least ten days. Intratumoral expression after injection of pDNA-jetPEI complexes was two to three logs lower, did not exceed the background in most mice, and lasted less than five days even with repeated dosing. Remarkably, reporter expression was primarily detected in the lungs, presumably due to leakage of pDNA-jetPEI complexes into the systemic circulation. In conclusion, electroporation enabled more efficient, prolonged and tumour-specific reporter expression compared to intratumoral injection of pDNA complexed with in vivo-jetPEI. These results favour the use of electroporation for intratumoral DNA-based gene transfer, and suggest further optimisation of pDNA-jetPEI complexes is needed to improve their efficacy and biosafety.
Collapse
|
40
|
Shirley SA, Lundberg CG, Heller R. Electrotransfer of IL-15/IL-15Rα Complex for the Treatment of Established Melanoma. Cancers (Basel) 2020; 12:cancers12103072. [PMID: 33096755 PMCID: PMC7589551 DOI: 10.3390/cancers12103072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/12/2020] [Accepted: 10/19/2020] [Indexed: 01/20/2023] Open
Abstract
Simple Summary The stimulation of the immune system through the administration of immunomodulatory agents such as cytokines has the potential to be an effective anti-cancer therapy. Obtaining the correct dose is an important aspect with respect to minimizing toxicity and obtaining the desired effect. A method to decrease the toxicity of this type of treatment is to replace the high-dose recombinant protein injections by using DNA expressing genes for one or more of these anti-cancer agents. In this current study, we have evaluated the delivery of interleukin-15 and its receptor in the form of plasmid DNA in a mouse melanoma model. We utilize a delivery approach that can deliver plasmid DNA in a manner that results in the desired level of expression being produced and induces a potent anti-tumor response as well as an immune memory response. Abstract Gene electrotransfer (GET) is a safe, reliable, and effective method of delivering plasmid DNA (pDNA) to solid tumors. GET has been previously used to deliver interleukin-15 (IL-15) to mouse melanoma, resulting in long-term tumor regression and the survival of a percentage of treated animals after challenge. To enhance this effect, we evaluated modulating the expression levels of IL-15 and co-expressing its receptor, IL-15Rα. GET was used to deliver plasmids encoding IL-15 and IL-15Rα to established B16.F10 tumors on days 0, 4, and 7. Two delivery protocols that yielded different expression profiles were utilized. Mice that were tumor-free for 50 days were then challenged with B16.F10 cells on the opposite flank and monitored for an additional 50 days. The amount of IL-15 expressed and the presence or absence of IL-15Rα in the treated tumors did not significantly affect the tumor regression and long-term survival. Upon challenge, however, low levels of IL-15 were more protective and resulted in a greater production of anti-tumor cytokines such as IFN-γ and MIP-1β and a greater amount of CD11b+ and CD3e+ cells infiltrating tumors. While mice with high levels of IL-15 showed CD11b+ and CD3e+ cell infiltrate, there was a substantial presence of NK cells that was absent in other treated groups. We can conclude that the level of IL-15 expressed in tumors after GET is an important determinant of the therapeutic outcome, a finding that will help us finetune this type of therapy.
Collapse
Affiliation(s)
- Shawna A. Shirley
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA; (S.A.S.); (C.G.L.)
| | - Cathryn G. Lundberg
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA; (S.A.S.); (C.G.L.)
| | - Richard Heller
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA; (S.A.S.); (C.G.L.)
- Department of Medical Engineering, University of South Florida, Tampa, FL 33512, USA
- Correspondence: ; Tel.: +01-813-974-1221
| |
Collapse
|
41
|
Zhang Y, Yan Z, Xia X, Lin Y. A Novel Electroporation System for Living Cell Staining and Membrane Dynamics Interrogation. MICROMACHINES 2020; 11:E767. [PMID: 32796554 PMCID: PMC7466103 DOI: 10.3390/mi11080767] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/07/2020] [Accepted: 08/10/2020] [Indexed: 12/11/2022]
Abstract
A novel electroporation system was developed to introduce transient membrane pores to cells in a spatially and temporally controlled manner, allowing us to achieve fast electrotransfection and live cell staining as well as to systematically interrogate the dynamics of the cell membrane. Specifically, using this platform, we showed that both reversible and irreversible electroporation could be induced in the cell population, with nano-sized membrane pores in the former case being able to self-reseal in ~10 min. In addition, green fluorescent protein(GFP)-vinculin plasmid and 543 phalloidin have been delivered successively into fibroblast cells, which enables us to monitor the distinct roles of vinculin and F-actin in cell adhesion and migration as well as their possible interplay during these processes. Compared to conventional bulk electroporation and staining methods, the new system offers advantages such as low-voltage operation, cellular level manipulation and testing, fast and adjustable transfection/staining and real-time monitoring; the new system therefore could be useful in different biophysical studies in the future.
Collapse
Affiliation(s)
- Yuanjun Zhang
- HKU-Shenzhen Institute of Research and Innovation (HKU-SIRI), Guangdong 518057, China; (Y.Z.); (Z.Y.); (X.X.)
| | - Zishen Yan
- HKU-Shenzhen Institute of Research and Innovation (HKU-SIRI), Guangdong 518057, China; (Y.Z.); (Z.Y.); (X.X.)
| | - Xingyu Xia
- HKU-Shenzhen Institute of Research and Innovation (HKU-SIRI), Guangdong 518057, China; (Y.Z.); (Z.Y.); (X.X.)
| | - Yuan Lin
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
42
|
Kiełbik A, Szlasa W, Saczko J, Kulbacka J. Electroporation-Based Treatments in Urology. Cancers (Basel) 2020; 12:E2208. [PMID: 32784598 PMCID: PMC7465806 DOI: 10.3390/cancers12082208] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 02/06/2023] Open
Abstract
The observation that an application of a pulsed electric field (PEF) resulted in an increased permeability of the cell membrane has led to the discovery of the phenomenon called electroporation (EP). Depending on the parameters of the electric current and cell features, electroporation can be either reversible or irreversible. The irreversible electroporation (IRE) found its use in urology as a non-thermal ablative method of prostate and renal cancer. As its mechanism is based on the permeabilization of cell membrane phospholipids, IRE (as well as other treatments based on EP) provides selectivity sparing extracellular proteins and matrix. Reversible EP enables the transfer of genes, drugs, and small exogenous proteins. In clinical practice, reversible EP can locally increase the uptake of cytotoxic drugs such as cisplatin and bleomycin. This approach is known as electrochemotherapy (ECT). Few in vivo and in vitro trials of ECT have been performed on urological cancers. EP provides the possibility of transmission of genes across the cell membrane. As the protocols of gene electrotransfer (GET) over the last few years have improved, EP has become a well-known technique for non-viral cell transfection. GET involves DNA transfection directly to the cancer or the host skin and muscle tissue. Among urological cancers, the GET of several plasmids encoding prostate cancer antigens has been investigated in clinical trials. This review brings into discussion the underlying mechanism of EP and an overview of the latest progress and development perspectives of EP-based treatments in urology.
Collapse
Affiliation(s)
- Aleksander Kiełbik
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (A.K.); (W.S.)
| | - Wojciech Szlasa
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (A.K.); (W.S.)
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| |
Collapse
|
43
|
DNA-based delivery of anti-DR5 Nanobodies improves exposure and anti-tumor efficacy over protein-based administration. Cancer Gene Ther 2020; 28:828-838. [PMID: 32733055 DOI: 10.1038/s41417-020-0204-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/16/2020] [Accepted: 07/22/2020] [Indexed: 11/09/2022]
Abstract
Nanobodies present an appealing class of potential cancer therapeutics. The current study explores the in vivo expression of these molecules through DNA-encoded delivery. We hypothesized that this approach could address the rapid clearance of Nanobodies and, through half-life modulation, increase the produced levels in circulation. We therefore evaluated pharmacokinetics and efficacy of variants of an anti-death receptor 5 Nanobody (NbDR5), either monovalent or multivalent with half-life extension properties, after DNA-based administration. Intramuscular electrotransfer of a monovalent NbDR5-encoding plasmid (pNbDR5) did not result in detectable plasma levels in BALB/c mice. A tetravalent NbDR5-encoding plasmid (pNbDR54) provided peak concentrations of 54 ng/mL, which remained above 24 ng/mL during a 12-week follow-up. DNA-based delivery of these Nanobody formats fused to a Nanobody binding to serum albumin (NbSA), pNbDR5-NbSA and pNbDR54-NbSA, resulted in significantly higher plasma levels, with peak titers of 5.2 and 7.7 µg/mL, respectively. In an athymic-nude mice COLO 205 colon-cancer model, a quadrupled intramuscular DNA dose led to peak plasma levels of 270 ng/mL for pNbDR54 and 38 µg/mL for pNbDR54-NbSA. Potent anti-tumor responses were only observed for pNbDR54, following either intramuscular or intratumoral delivery. Despite comparable in vitro activity and superior plasma exposure, NbDR54-NbSA was less effective than NbDR54 in vivo, regardless of whether delivered as DNA or protein. Overall, DNA-based Nanobody delivery resulted in more potent and durable anti-tumor responses than protein-based Nanobody delivery. In conclusion, this study demonstrates pre-clinical proof of concept for DNA-based Nanobodies in oncology and highlights the improved outcome over conventional protein administration.
Collapse
|
44
|
Reddy OL, Savani BN, Stroncek DF, Panch SR. Advances in gene therapy for hematologic disease and considerations for transfusion medicine. Semin Hematol 2020; 57:83-91. [PMID: 32892847 DOI: 10.1053/j.seminhematol.2020.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Indexed: 12/26/2022]
Abstract
As the list of regulatory agency-approved gene therapies grows, these products are now in the therapeutic spotlight with the potential to cure or dramatically alleviate several benign and malignant hematologic diseases. The mechanisms for gene manipulation are diverse, and include the use of a variety of cell sources and both viral vector- and nuclease-based targeted approaches. Gene editing has also reached the realm of blood component therapy and testing, where cultured products are being developed to improve transfusion support for individuals with rare blood types. In this review, we summarize the milestones in the development of gene therapies for hematologic diseases, mechanisms for gene manipulation, and implications for transfusion medicine and blood centers as these therapies continue to advance and grow.
Collapse
Affiliation(s)
- Opal L Reddy
- Center for Cellular Engineering, National institutes of Health, Clinical Center, Bethesda, Maryland
| | - Bipin N Savani
- Vanderbilt University Medical Center, Nashville, Tennessee
| | - David F Stroncek
- Center for Cellular Engineering, National institutes of Health, Clinical Center, Bethesda, Maryland
| | - Sandhya R Panch
- Center for Cellular Engineering, National institutes of Health, Clinical Center, Bethesda, Maryland.
| |
Collapse
|
45
|
Induction of a local muscular dystrophy using electroporation in vivo: an easy tool for screening therapeutics. Sci Rep 2020; 10:11301. [PMID: 32647247 PMCID: PMC7347864 DOI: 10.1038/s41598-020-68135-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 06/09/2020] [Indexed: 01/19/2023] Open
Abstract
Intramuscular injection and electroporation of naked plasmid DNA (IMEP) has emerged as a potential alternative to viral vector injection for transgene expression into skeletal muscles. In this study, IMEP was used to express the DUX4 gene into mouse tibialis anterior muscle. DUX4 is normally expressed in germ cells and early embryo, and silenced in adult muscle cells where its pathological reactivation leads to Facioscapulohumeral muscular dystrophy. DUX4 encodes a potent transcription factor causing a large deregulation cascade. Its high toxicity but sporadic expression constitutes major issues for testing emerging therapeutics. The IMEP method appeared as a convenient technique to locally express DUX4 in mouse muscles. Histological analyses revealed well delineated muscle lesions 1-week after DUX4 IMEP. We have therefore developed a convenient outcome measure by quantification of the damaged muscle area using color thresholding. This method was used to characterize lesion distribution and to assess plasmid recirculation and dose–response. DUX4 expression and activity were confirmed at the mRNA and protein levels and through a quantification of target gene expression. Finally, this study gives a proof of concept of IMEP model usefulness for the rapid screening of therapeutic strategies, as demonstrated using antisense oligonucleotides against DUX4 mRNA.
Collapse
|
46
|
Wang Y, Chang CC, Wang L, Yuan F. Enhancing Cell Viability and Efficiency of Plasmid DNA Electrotransfer Through Reducing Plasma Membrane Permeabilization. Bioelectricity 2020; 2:251-257. [PMID: 33344914 DOI: 10.1089/bioe.2020.0007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Pulsed electric field has been widely used to facilitate molecular cargo transfer into cells. However, the cell viability is often decreased when trying to increase the electrotransfer efficiency. We hypothesize that the decrease is due to electropermeabilization of cell membrane that disrupts homeostasis of intracellular microenvironment. Thus, a reduction in the membrane permeabilization may increase the cell viability. Materials and Methods Different compounds were supplemented into the pulsing buffer prior to electrotransfer for reduction of cell membrane damage. Extent of the damage was quantified by leakiness of the membrane to a fluorescent dye, calcein, preloaded into cells. At 24 hours post electrotransfer, cell viability and electrotransfer efficiency were quantified with flow cytometry. Results The cell viability could be substantially increased by supplementation of either type B gelatin or bovine serum albumin (BSA), without compromising the electrotransfer efficiency. The supplementation also decreased the amount of calcein leaking out of the cells, suggesting that the improvement in cell viability was due to the reduction in electrotransfer-induced membrane damage. Conclusion Data from the study demonstrate that type B gelatin and BSA can be used as inexpensive supplements for improving cell viability in electrotransfer.
Collapse
Affiliation(s)
- Yanhua Wang
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Chun-Chi Chang
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Liangli Wang
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Fan Yuan
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| |
Collapse
|
47
|
Dermol-Černe J, Batista Napotnik T, Reberšek M, Miklavčič D. Short microsecond pulses achieve homogeneous electroporation of elongated biological cells irrespective of their orientation in electric field. Sci Rep 2020; 10:9149. [PMID: 32499601 PMCID: PMC7272635 DOI: 10.1038/s41598-020-65830-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 04/27/2020] [Indexed: 12/21/2022] Open
Abstract
In gene electrotransfer and cardiac ablation with irreversible electroporation, treated muscle cells are typically of elongated shape and their orientation may vary. Orientation of cells in electric field has been reported to affect electroporation, and hence electrodes placement and pulse parameters choice in treatments for achieving homogeneous effect in tissue is important. We investigated how cell orientation influences electroporation with respect to different pulse durations (ns to ms range), both experimentally and numerically. Experimentally detected electroporation (evaluated separately for cells parallel and perpendicular to electric field) via Ca2+ uptake in H9c2 and AC16 cardiomyocytes was numerically modeled using the asymptotic pore equation. Results showed that cell orientation affects electroporation extent: using short, nanosecond pulses, cells perpendicular to electric field are significantly more electroporated than parallel (up to 100-times more pores formed), and with long, millisecond pulses, cells parallel to electric field are more electroporated than perpendicular (up to 1000-times more pores formed). In the range of a few microseconds, cells of both orientations were electroporated to the same extent. Using pulses of a few microseconds lends itself as a new possible strategy in achieving homogeneous electroporation in tissue with elongated cells of different orientation (e.g. electroporation-based cardiac ablation).
Collapse
Affiliation(s)
- Janja Dermol-Černe
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, 1000, Ljubljana, Slovenia
| | - Tina Batista Napotnik
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, 1000, Ljubljana, Slovenia
| | - Matej Reberšek
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, 1000, Ljubljana, Slovenia
| | - Damijan Miklavčič
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, 1000, Ljubljana, Slovenia.
| |
Collapse
|
48
|
Sieni E, Dettin M, De Robertis M, Bazzolo B, Conconi MT, Zamuner A, Marino R, Keller F, Campana LG, Signori E. The Efficiency of Gene Electrotransfer in Breast-Cancer Cell Lines Cultured on a Novel Collagen-Free 3D Scaffold. Cancers (Basel) 2020; 12:cancers12041043. [PMID: 32340405 PMCID: PMC7226458 DOI: 10.3390/cancers12041043] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/08/2020] [Accepted: 04/21/2020] [Indexed: 12/15/2022] Open
Abstract
Gene Electro-Transfer (GET) is a powerful method of DNA delivery with great potential for medical applications. Although GET has been extensively studied in vitro and in vivo, the optimal parameters remain controversial. 2D cell cultures have been widely used to investigate GET protocols, but have intrinsic limitations, whereas 3D cultures may represent a more reliable model thanks to the capacity of reproducing the tumor architecture. Here we applied two GET protocols, using a plate or linear electrode, on 3D-cultured HCC1954 and MDA-MB231 breast cancer cell lines grown on a novel collagen-free 3D scaffold and compared results with conventional 2D cultures. To evaluate the electrotransfer efficiency, we used the plasmid pEGFP-C3 encoding the enhanced green fluorescent protein (EGFP) reporter gene. The novel 3D scaffold promoted extracellular matrix deposition, which particularly influences cell behavior in both in vitro cell cultures and in vivo tumor tissue. While the transfection efficiency was similar in the 2D-cultures, we observed significant differences in the 3D-model. The transfection efficiency in the 3D vs 2D model was 44% versus 15% (p < 0.01) and 24% versus 17% (p < 0.01) in HCC1954 and MDA-MB231 cell cultures, respectively. These findings suggest that the novel 3D scaffold allows reproducing, at least partially, the peculiar morphology of the original tumor tissues, thus allowing us to detect meaningful differences between the two cell lines. Following GET with plate electrodes, cell viability was higher in 3D-cultured HCC1954 (66%) and MDA-MB231 (96%) cell lines compared to their 2D counterpart (53% and 63%, respectively, p < 0.001). Based on these results, we propose the novel 3D scaffold as a reliable support for the preparation of cell cultures in GET studies. It may increase the reliability of in vitro assays and allow the optimization of GET parameters of in vivo protocols.
Collapse
Affiliation(s)
- Elisabetta Sieni
- Department of Theoretical and Applied Sciences, University of Insubria, 21100 Varese, Italy
- Correspondence: (E.S.); (E.S.); Tel.: +39-0332-421405 (E.S.); Tel.: +39-0-649-934-232 (E.S.)
| | - Monica Dettin
- Department of Industrial Engineering, University of Padova, 35131 Padova, Italy; (M.D.); (A.Z.)
| | - Mariangela De Robertis
- CNR-Institute of Biomembrane, Bioenergetics and Molecular Biotechnology, 70126 Bari, Italy;
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari, 70126 Bari, Itay
| | - Bianca Bazzolo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (B.B.); (M.T.C.)
| | - Maria Teresa Conconi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (B.B.); (M.T.C.)
| | - Annj Zamuner
- Department of Industrial Engineering, University of Padova, 35131 Padova, Italy; (M.D.); (A.Z.)
| | - Ramona Marino
- Campus Bio-Medico University of Rome, 00128 Roma, Italy; (R.M.); (F.K.)
| | - Flavio Keller
- Campus Bio-Medico University of Rome, 00128 Roma, Italy; (R.M.); (F.K.)
| | - Luca Giovanni Campana
- Department of Surgical Oncological and Gastroenterological Sciences DISCOG, University of Padova, 35124 Padova, Italy;
| | - Emanuela Signori
- Campus Bio-Medico University of Rome, 00128 Roma, Italy; (R.M.); (F.K.)
- CNR-Institute of Translational Pharmacology, 00133 Roma, Italy
- Correspondence: (E.S.); (E.S.); Tel.: +39-0332-421405 (E.S.); Tel.: +39-0-649-934-232 (E.S.)
| |
Collapse
|
49
|
Jacobs L, De Smidt E, Geukens N, Declerck P, Hollevoet K. DNA-Based Delivery of Checkpoint Inhibitors in Muscle and Tumor Enables Long-Term Responses with Distinct Exposure. Mol Ther 2020; 28:1068-1077. [PMID: 32101701 DOI: 10.1016/j.ymthe.2020.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 02/08/2020] [Indexed: 12/17/2022] Open
Abstract
Checkpoint-inhibiting antibodies elicit impressive clinical responses, but still face several issues. The current study evaluated whether DNA-based delivery can broaden the application of checkpoint inhibitors, specifically by pursuing cost-efficient in vivo production, facilitating combination therapies, and exploring administration routes that lower immune-related toxicity risks. We therefore optimized plasmid-encoded anti-CTLA-4 and anti-PD-1 antibodies, and studied their pharmacokinetics and pharmacodynamics when delivered alone and in combination via intramuscular or intratumoral electroporation in mice. Intramuscular electrotransfer of these DNA-based antibodies induced complete regressions in a subcutaneous MC38 tumor model, with plasma concentrations up to 4 and 14 μg/mL for anti-CTLA-4 and anti-PD-1 antibodies, respectively, and antibody detection for at least 6 months. Intratumoral antibody gene electrotransfer gave similar anti-tumor responses as the intramuscular approach. Antibody plasma levels, however, were up to 70-fold lower and substantially more transient, potentially improving biosafety of the expressed checkpoint inhibitors. Intratumoral delivery also generated a systemic anti-tumor response, illustrated by moderate abscopal effects and prolonged protection of cured mice against a tumor rechallenge. In conclusion, intramuscular and intratumoral DNA-based delivery of checkpoint inhibitors both enabled long-term anti-tumor responses despite distinct systemic antibody exposure, highlighting the potential of the tumor as delivery site for DNA-based therapeutics.
Collapse
Affiliation(s)
- Liesl Jacobs
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven - University of Leuven, Leuven, Belgium
| | - Elien De Smidt
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven - University of Leuven, Leuven, Belgium; PharmAbs - The KU Leuven Antibody Center, KU Leuven - University of Leuven, Leuven, Belgium
| | - Nick Geukens
- PharmAbs - The KU Leuven Antibody Center, KU Leuven - University of Leuven, Leuven, Belgium
| | - Paul Declerck
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven - University of Leuven, Leuven, Belgium; PharmAbs - The KU Leuven Antibody Center, KU Leuven - University of Leuven, Leuven, Belgium.
| | - Kevin Hollevoet
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven - University of Leuven, Leuven, Belgium.
| |
Collapse
|
50
|
Sachdev S, Feijoo Moreira S, Keehnen Y, Rems L, Kreutzer MT, Boukany PE. DNA-membrane complex formation during electroporation is DNA size-dependent. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183089. [DOI: 10.1016/j.bbamem.2019.183089] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 09/11/2019] [Accepted: 10/22/2019] [Indexed: 01/09/2023]
|