1
|
Hoch CC, Hachani K, Han Y, Schmidl B, Wirth M, Multhoff G, Bashiri Dezfouli A, Wollenberg B. The future of interleukin gene therapy in head and neck cancers. Expert Opin Biol Ther 2024; 24:1057-1073. [PMID: 39291462 DOI: 10.1080/14712598.2024.2405568] [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: 06/26/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024]
Abstract
INTRODUCTION Head and neck cancer (HNC), primarily head and neck squamous cell carcinomas, originates from the squamous epithelium in areas like the oral cavity, lip, larynx, and oropharynx. With high morbidity impacting critical functions, combined treatments like surgery, radiation, and chemotherapy often fall short in advanced stages, highlighting the need for innovative therapies. AREAS COVERED This review critically evaluates interleukin (IL) gene therapy for treating HNC. The discussion extends to key ILs in HNC, various gene therapy techniques and delivery methods. We particularly focus on the application of IL-2, IL-12, and IL-24 gene therapies, examining their mechanisms and outcomes in preclinical studies and clinical trials. The final sections address IL gene therapy challenges in HNC, exploring solutions and critically assessing future therapeutic directions. EXPERT OPINION Despite advancements in genomic and immunotherapy, significant challenges in HNC treatment persist, primarily due to the immunosuppressive nature of the tumor microenvironment and the adverse effects of current therapies. The therapeutic efficacy of IL gene therapy hinges on overcoming these hurdles through refined delivery methods that ensure targeted, tumor-specific gene expression. Future strategies should focus on refining gene delivery methods and combining IL gene therapy with other treatments to optimize efficacy and minimize toxicity.
Collapse
Affiliation(s)
- Cosima C Hoch
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Khouloud Hachani
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Yu Han
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Benedikt Schmidl
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Markus Wirth
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Gabriele Multhoff
- Central Institute for Translational Cancer Research, Technical University of Munich (TranslaTUM), Munich, Germany
- Department of Radiation Oncology, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Ali Bashiri Dezfouli
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
- Central Institute for Translational Cancer Research, Technical University of Munich (TranslaTUM), Munich, Germany
- Department of Radiation Oncology, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Barbara Wollenberg
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| |
Collapse
|
2
|
Batista Napotnik T, Polajžer T, Miklavčič D. Cell death due to electroporation - A review. Bioelectrochemistry 2021; 141:107871. [PMID: 34147013 DOI: 10.1016/j.bioelechem.2021.107871] [Citation(s) in RCA: 232] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/12/2021] [Accepted: 06/03/2021] [Indexed: 12/15/2022]
Abstract
Exposure of cells to high voltage electric pulses increases transiently membrane permeability through membrane electroporation. Electroporation can be reversible and is used in gene transfer and enhanced drug delivery but can also lead to cell death. Electroporation resulting in cell death (termed as irreversible electroporation) has been successfully used as a new non-thermal ablation method of soft tissue such as tumours or arrhythmogenic heart tissue. Even though the mechanisms of cell death can influence the outcome of electroporation-based treatments due to use of different electric pulse parameters and conditions, these are not elucidated yet. We review the mechanisms of cell death after electroporation reported in literature, cell injuries that may lead to cell death after electroporation and membrane repair mechanisms involved. The knowledge of membrane repair and cell death mechanisms after cell exposure to electric pulses, targets of electric field in cells need to be identified to optimize existing and develop of new electroporation-based techniques used in medicine, biotechnology, and food technology.
Collapse
Affiliation(s)
- Tina Batista Napotnik
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, 1000 Ljubljana, Slovenia
| | - Tamara Polajžer
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, 1000 Ljubljana, Slovenia
| | - Damijan Miklavčič
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, 1000 Ljubljana, Slovenia.
| |
Collapse
|
3
|
Nguyen KG, Vrabel MR, Mantooth SM, Hopkins JJ, Wagner ES, Gabaldon TA, Zaharoff DA. Localized Interleukin-12 for Cancer Immunotherapy. Front Immunol 2020; 11:575597. [PMID: 33178203 PMCID: PMC7593768 DOI: 10.3389/fimmu.2020.575597] [Citation(s) in RCA: 268] [Impact Index Per Article: 53.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/08/2020] [Indexed: 12/30/2022] Open
Abstract
Interleukin-12 (IL-12) is a potent, pro-inflammatory type 1 cytokine that has long been studied as a potential immunotherapy for cancer. Unfortunately, IL-12's remarkable antitumor efficacy in preclinical models has yet to be replicated in humans. Early clinical trials in the mid-1990's showed that systemic delivery of IL-12 incurred dose-limiting toxicities. Nevertheless, IL-12's pleiotropic activity, i.e., its ability to engage multiple effector mechanisms and reverse tumor-induced immunosuppression, continues to entice cancer researchers. The development of strategies which maximize IL-12 delivery to the tumor microenvironment while minimizing systemic exposure are of increasing interest. Diverse IL-12 delivery systems, from immunocytokine fusions to polymeric nanoparticles, have demonstrated robust antitumor immunity with reduced adverse events in preclinical studies. Several localized IL-12 delivery approaches have recently reached the clinical stage with several more at the precipice of translation. Taken together, localized delivery systems are supporting an IL-12 renaissance which may finally allow this potent cytokine to fulfill its considerable clinical potential. This review begins with a brief historical account of cytokine monotherapies and describes how IL-12 went from promising new cure to ostracized black sheep following multiple on-study deaths. The bulk of this comprehensive review focuses on developments in diverse localized delivery strategies for IL-12-based cancer immunotherapies. Advantages and limitations of different delivery technologies are highlighted. Finally, perspectives on how IL-12-based immunotherapies may be utilized for widespread clinical application in the very near future are offered.
Collapse
Affiliation(s)
- Khue G Nguyen
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Maura R Vrabel
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Siena M Mantooth
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Jared J Hopkins
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Ethan S Wagner
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Taylor A Gabaldon
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - David A Zaharoff
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
4
|
Nemec A, Milevoj N, Lampreht Tratar U, Serša G, Čemažar M, Tozon N. Electroporation-Based Treatments in Small Animal Veterinary Oral and Maxillofacial Oncology. Front Vet Sci 2020; 7:575911. [PMID: 33134356 PMCID: PMC7550461 DOI: 10.3389/fvets.2020.575911] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/27/2020] [Indexed: 12/18/2022] Open
Abstract
Electroporation is a method of inducing an increase in permeability of the cell membrane through the application of an electric field and can be used as a delivery method for introducing molecules of interest (e.g., chemotherapeutics or plasmid DNA) into cells. Electroporation-based treatments (i.e., electrochemotherapy, gene electrotransfer, and their combinations) have been shown to be safe and effective in veterinary oncology, but they are currently mostly recommended for the treatment of those solid tumors for which clients have declined surgery and/or radiotherapy. Published data show that electroporation-based treatments are also safe, simple, fast and cost-effective treatment alternatives for selected oral and maxillofacial tumors, especially small squamous cell carcinoma and malignant melanoma tumors not involving the bone in dogs. In these patients, a good local response to treatment is expected to result in increased survival time with good quality of life. Despite emerging evidence of the clinical efficacy of electroporation-based treatments for oral and maxillofacial tumors, further investigation is needed to optimize treatment protocols, improve clinical data reporting and better understand the mechanisms of patients' response to the treatment.
Collapse
Affiliation(s)
- Ana Nemec
- Small Animal Clinic, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Nina Milevoj
- Small Animal Clinic, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia
| | | | - Gregor Serša
- Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Maja Čemažar
- Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Nataša Tozon
- Small Animal Clinic, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
5
|
Nath S, Mondal S, Butti R, Prasanna Gunasekaran V, Chatterjee U, Halder A, Kundu GC, Mandal C. Desialylation of Sonic-Hedgehog by Neu2 Inhibits Its Association with Patched1 Reducing Stemness-Like Properties in Pancreatic Cancer Sphere-forming Cells. Cells 2020; 9:cells9061512. [PMID: 32575925 PMCID: PMC7349614 DOI: 10.3390/cells9061512] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/06/2020] [Accepted: 04/08/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer stem cells (CSCs) are crucial regulators of tumor recurrence/progression. The maintenance of CSCs is dependent on aberrant activation of various pathways, including Hedgehog. Prevalent sialylations contribute to aggressiveness in CSCs. Here, we have addressed the role of sialylation in regulating stemness-like properties of pancreatic cancer sphere-forming cells (PCS) through modulation of the Hedgehog (Hh) pathway. The status of CD133/CD44/surface-sialylation was checked by flow cytometry and effects of Neu2 overexpression in PCS were compared using qPCR, immunoblotting, co-immunoprecipitation and also by colony-formation assays. The work was also validated in a xenograft model after Neu2 overexpression. Neu2 and Shh status in patient tissues were examined by immunohistochemistry. PCS showed higher Hh-pathway activity and sialylation with reduced cytosolic-sialidase (Neu2). Neu2 overexpression caused desialylation of Shh, thereby reducing Shh-Patched1 binding thus causing decreased Hh-pathway activity with lower expression of Snail/Slug/CyclinD1 leading to reduction of stemness-like properties. Neu2-overexpression also induced apoptosis in PCS. Additionally, Neu2-overexpressed PCS demonstrated lower mTORC2 formation and inhibitory-phosphorylation of Gsk3β, reflecting a close relationship with reduced Hh pathway. Moreover, both Neu2 and Rictor (a major component of mTORC2) co-transfection reduced stem cell markers and Hh-pathway activity in PCS. Neu2-overexpressed tumors showed reduction in tumor mass with downregulation of stem cell markers/Shh/mTOR and upregulation of Bax/Caspase8/Caspase3. Thus, we established that reduced sialylation by Neu2 overexpression leads to decreased stemness-like properties by desialylation of Shh, which impaired its association with Patched1 thereby inhibiting the Hh pathway. All these may be responsible for enhanced apoptosis in Neu2-overexpressed PCS.
Collapse
Affiliation(s)
- Shalini Nath
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4, Raja S.C. Mallick Road, Kolkata 700032, India; (S.N.); (S.M.)
| | - Susmita Mondal
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4, Raja S.C. Mallick Road, Kolkata 700032, India; (S.N.); (S.M.)
| | - Ramesh Butti
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science (NCCS), Pune 411007, India; (R.B.); (V.P.G.); (G.C.K.)
| | - Vinoth Prasanna Gunasekaran
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science (NCCS), Pune 411007, India; (R.B.); (V.P.G.); (G.C.K.)
| | - Uttara Chatterjee
- Department of Pathology, Institute of Post-Graduate Medical Education and Research Hospital, Kolkata, West Bengal 700020, India;
| | - Aniket Halder
- School of Digestive & Liver Diseases, Institute of Post-Graduate Medical Education and Research Hospital, Kolkata, West Bengal 700020, India;
| | - Gopal C. Kundu
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science (NCCS), Pune 411007, India; (R.B.); (V.P.G.); (G.C.K.)
| | - Chitra Mandal
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4, Raja S.C. Mallick Road, Kolkata 700032, India; (S.N.); (S.M.)
- Correspondence: or ; Tel.: +91-33-2499-5717
| |
Collapse
|
6
|
Pasquet L, Bellard E, Chabot S, Markelc B, Rols MP, Teissie J, Golzio M. Pre-clinical investigation of the synergy effect of interleukin-12 gene-electro-transfer during partially irreversible electropermeabilization against melanoma. J Immunother Cancer 2019; 7:161. [PMID: 31242938 PMCID: PMC6595571 DOI: 10.1186/s40425-019-0638-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 06/13/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Melanoma is a very aggressive skin tumor that can be cured when diagnosed and treated in its early stages. However, at the time of identification, the tumor is frequently in a metastatic stage. Intensive research is currently ongoing to improve the efficacy of the immune system in eliminating cancer cells. One approach is to boost the activation of cytotoxic T cells by IL-12 cytokine that plays a central role in the activation of the immune system. In parallel, physical methods such as electropermeabilization-based treatments are currently under investigation and show promising results. METHODS In this study, we set electrical parameters to induce a partial-irreversible electropermeabilization (pIRE) of melanoma to induce a sufficient cell death and potential release of tumor antigens able to activate immune cells. This protocol mimics the situation where irreversible electropermeabilization is not fully completed. Then, a peritumoral plasmid IL-12 electrotransfer was combined with pIRE treatment. Evaluation of the tumor growth and survival was performed in mouse strains having a different immunological background (C57Bl/6 (WT), nude and C57Bl6 (TLR9-/-)). RESULTS pIRE treatment induced apoptotic cell death and a temporary tumor growth delay in all mouse strains. In C57Bl/6 mice, we showed that peritumoral plasmid IL-12 electrotransfer combined with tumor pIRE treatment induced tumor regression correlating with a local secretion of IL-12 and IFN-γ. This combined treatment induced a growth delay of distant tumors and prevented the emergence of a second tumor in 50% of immunocompetent mice. CONCLUSIONS The combination of pIL-12 GET and pIRE not only enhanced survival but could bring a curative effect in wild type mice. This two-step treatment, named Immune-Gene Electro-Therapy (IGET), led to a systemic activation of the adaptive immune system and the development of an anti-tumor immune memory.
Collapse
Affiliation(s)
- Lise Pasquet
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, BP 64182, UMR 5089, 205 Route de Narbonne, F-31077, Toulouse Cedex, France
| | - Elisabeth Bellard
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, BP 64182, UMR 5089, 205 Route de Narbonne, F-31077, Toulouse Cedex, France
| | - Sophie Chabot
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, BP 64182, UMR 5089, 205 Route de Narbonne, F-31077, Toulouse Cedex, France
| | - Bostjan Markelc
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, BP 64182, UMR 5089, 205 Route de Narbonne, F-31077, Toulouse Cedex, France
| | - Marie-Pierre Rols
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, BP 64182, UMR 5089, 205 Route de Narbonne, F-31077, Toulouse Cedex, France
| | - Justin Teissie
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, BP 64182, UMR 5089, 205 Route de Narbonne, F-31077, Toulouse Cedex, France.
| | - Muriel Golzio
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, BP 64182, UMR 5089, 205 Route de Narbonne, F-31077, Toulouse Cedex, France.
| |
Collapse
|
7
|
Plaschke CC, Johannesen HH, Hansen RH, Hendel HW, Kiss K, Gehl J, Wessel I. The DAHANCA 32 study: Electrochemotherapy for recurrent mucosal head and neck cancer. Head Neck 2018; 41:329-339. [PMID: 30552847 DOI: 10.1002/hed.25454] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 05/30/2018] [Accepted: 08/15/2018] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Electrochemotherapy is an established treatment for cutaneous tumors. This study aimed at determining efficacy of electrochemotherapy in recurrent head and neck cancer. METHODS Phase II clinical trial in patients with recurrent head and neck carcinomas with no curative treatment options. Electrochemotherapy was performed under general anesthesia. Primary endpoint was tumor response (CT scanning) evaluated at week 8. Secondary endpoints included biopsy results, MRI and fluorodeoxyglucose-positron emission tomography scanning, safety, toxicity, pain score, and quality-of-life questionnaires. RESULTS Of 26 patients treated, 5 (19%) achieved complete response, 10 (39%) partial response, resulting in an objective response of 58%. Two responders remain without recurrence. No serious adverse events occurred during treatment. Four events occurred posttreatment: one bleeding episode, two episodes with mucosal swelling, and one patient died due to disease progression. CONCLUSION Electrochemotherapy is efficient against local recurrence of head and neck cancer with an overall response rate of 58%.
Collapse
Affiliation(s)
- Christina Caroline Plaschke
- Department of Otorhinolaryngology, Head & Neck Surgery and Audiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Helle Hjorth Johannesen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Rasmus Hvass Hansen
- Department of Radiology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | | | - Katalin Kiss
- Department of Pathology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Julie Gehl
- Center for Experimental Drug and Gene Electrotransfer (C*EDGE), Department of Clinical Oncology and Palliative Care, Zealand University Hospital, Roskilde, Denmark.,Faculty of Health and Medical Sciences, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.,Department of Oncology, Herlev and Gentofte Hospital, University of Copenhagen, Herlev, Denmark
| | - Irene Wessel
- Department of Otorhinolaryngology, Head & Neck Surgery and Audiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
8
|
Hu J, Sun C, Bernatchez C, Xia X, Hwu P, Dotti G, Li S. T-cell Homing Therapy for Reducing Regulatory T Cells and Preserving Effector T-cell Function in Large Solid Tumors. Clin Cancer Res 2018; 24:2920-2934. [PMID: 29391351 PMCID: PMC6004229 DOI: 10.1158/1078-0432.ccr-17-1365] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 11/20/2017] [Accepted: 01/25/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Infused autologous tumor-infiltrating lymphocytes (TIL) and tumor-targeted chimeric antigen receptor (CAR) T cells typically surround malignant lesions or penetrate small tumor nodules but fail to penetrate large solid tumors, significantly compromising their antitumor impact. Strategies to overcome this primary challenge are largely required.Experimental Design: We tested the effects of IL12 plus doxorubicin on T-cell penetration and efficacy in solid tumors in a murine lung cancer model, a murine breast carcinoma lung metastasis model, and two human xenograft tumor models bearing large tumors (>10 mm).Results: Intriguingly, this simple approach increased the numbers, the distribution, and the depth of penetration of infused CD8+ T cells in these tumors, including both TILs and CAR T cells. This combined treatment halted tumor progression and significantly extended survival time. Studies of the underlying mechanism revealed multiple effects. First, the combined treatment maintained the high ratios of immune-stimulatory receptors to immune-inhibitory receptors on infiltrated CD8+ T cells, reduced the accumulation of immunosuppressive regulatory T cells, and enhanced the numbers of T-bet+ effector T cells in the tumors. Second, doxorubicin induced chemokines CXCL9 and CXCL10, which may attract NKG2D+CD8+ T cells to tumors, and this effect was boosted by IL12-induced IFNγ accumulation in tumors, promoting the penetration of NKG2D+CD8+ T cells.Conclusions: The deep penetration of infused T cells associated with combined IL12 plus doxorubicin yielded striking therapeutic effects in murine and human xenograft solid tumors. This approach might broaden the application of T-cell therapy to a wider range of solid tumors. Clin Cancer Res; 24(12); 2920-34. ©2018 AACRSee related commentary by Berraondo et al., p. 2716.
Collapse
Affiliation(s)
- Jiemiao Hu
- Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chuang Sun
- Department of Microbiology and Immunology, University of North Carolina, School of Medicine, Chapel Hill, North Carolina
| | - Chantale Bernatchez
- Department of Melanoma Medical Oncology, Center for Cancer Immunology Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xueqing Xia
- Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Patrick Hwu
- Department of Melanoma Medical Oncology, Center for Cancer Immunology Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gianpietro Dotti
- Department of Microbiology and Immunology, University of North Carolina, School of Medicine, Chapel Hill, North Carolina
| | - Shulin Li
- Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
9
|
Canton DA, Shirley S, Wright J, Connolly R, Burkart C, Mukhopadhyay A, Twitty C, Qattan KE, Campbell JS, Le MH, Pierce RH, Gargosky S, Daud A, Algazi A. Melanoma treatment with intratumoral electroporation of tavokinogene telseplasmid (pIL-12, tavokinogene telseplasmid). Immunotherapy 2017; 9:1309-1321. [PMID: 29064334 DOI: 10.2217/imt-2017-0096] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Tumors evade detection and/or clearance by the immune system via multiple mechanisms. IL-12 is a potent immunomodulatory cytokine that plays a central role in immune priming. However, systemic delivery of IL-12 can result in life-threatening toxicity and therefore has shown limited efficacy at doses that can be safely administered. We developed an electroporation technique to produce highly localized IL-12 expression within tumors leading to regression of both treated and untreated lesions in animal models and in patients with a favorable safety profile. Furthermore, intratumoral tavokinogene telseplasmid electroporation can drive cellular immune responses, converting 'cold' tumors into 'hot' tumors. Clinical trials are ongoing to determine whether intratumoral tavokinogene telseplasmid electroporation synergizes with checkpoint blockade therapy in immunologically cold tumors predicted not to respond to PD-1 antibody monotherapy.
Collapse
Affiliation(s)
- David A Canton
- OncoSec Medical Incorporated, 5820 Nancy Ridge Dr, San Diego, CA 92121, USA
| | - Shawna Shirley
- OncoSec Medical Incorporated, 5820 Nancy Ridge Dr, San Diego, CA 92121, USA
| | - Jocelyn Wright
- OncoSec Medical Incorporated, 5820 Nancy Ridge Dr, San Diego, CA 92121, USA
| | - Richard Connolly
- OncoSec Medical Incorporated, 5820 Nancy Ridge Dr, San Diego, CA 92121, USA.,Fred Hutchinson Cancer Research Center, Clinical Research Division, 1100 Fairview Ave. N. Seattle, WA 98109, USA
| | - Christoph Burkart
- OncoSec Medical Incorporated, 5820 Nancy Ridge Dr, San Diego, CA 92121, USA
| | | | - Chris Twitty
- OncoSec Medical Incorporated, 5820 Nancy Ridge Dr, San Diego, CA 92121, USA
| | - Kristen E Qattan
- OncoSec Medical Incorporated, 5820 Nancy Ridge Dr, San Diego, CA 92121, USA
| | - Jean S Campbell
- Fred Hutchinson Cancer Research Center, Clinical Research Division, 1100 Fairview Ave. N. Seattle, WA 98109, USA
| | - Mai H Le
- Fred Hutchinson Cancer Research Center, Clinical Research Division, 1100 Fairview Ave. N. Seattle, WA 98109, USA
| | - Robert H Pierce
- Fred Hutchinson Cancer Research Center, Clinical Research Division, 1100 Fairview Ave. N. Seattle, WA 98109, USA
| | - Sharron Gargosky
- OncoSec Medical Incorporated, 5820 Nancy Ridge Dr, San Diego, CA 92121, USA
| | - Adil Daud
- UCSF Helen Diller Family Comprehensive Cancer Center, 1600 Divisadero St, San Francisco, CA 94115, USA
| | - Alain Algazi
- UCSF Helen Diller Family Comprehensive Cancer Center, 1600 Divisadero St, San Francisco, CA 94115, USA
| |
Collapse
|
10
|
Gene Electrotransfer of Plasmid-Encoding IL-12 Recruits the M1 Macrophages and Antigen-Presenting Cells Inducing the Eradication of Aggressive B16F10 Murine Melanoma. Mediators Inflamm 2017; 2017:5285890. [PMID: 28596641 PMCID: PMC5449735 DOI: 10.1155/2017/5285890] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/03/2017] [Accepted: 03/12/2017] [Indexed: 12/30/2022] Open
Abstract
Cancer immunotherapy is currently one of the leading approaches in cancer treatment. Gene electrotransfer of plasmids encoding interleukin 12 (IL-12) into the cells leads to the production of IL-12, which drives immune cell polarization to an antitumoral response. One of the cell types that shows great promise in targeting tumor cells under the influence of IL-12 cytokine milieu is that of macrophages. Therefore, the aim of this study was to evaluate gene electrotransfer of antibiotic resistance-free plasmid DNA-encoding murine IL-12 (mIL-12) in mice bearing aggressive B16F10 murine melanoma. IL-12 electrotransfer resulted in the complete long-term eradication of the tumors. Serum mIL-12 and murine interferon γ (mIFNγ) were increased after IL-12 gene electrotransfer. Further on, hematoxylin and eosin (HE) staining showed increased infiltration of immune cells that lasted from day 4 until day 14. Immunohistochemistry (IHC) staining of F4/80, MHCII, and CD11c showed higher positive staining in the IL-12 gene electrotransfer group than in the control groups. Immune cell infiltration into the tumors and the high density of MHCII- and CD11c-positive cells suggest an antitumor polarization of macrophages and the presence of antigen-presenting cells that contributes to the important antitumor effectiveness of IL-12.
Collapse
|
11
|
Xiao Y, Yang Y, Wu Y, Wang C, Cheng H, Zhao W, Li Y, Liu B, Long J, Guo W, Gao G, Gou M. Nanoparticles co-delivering pVSVMP and pIL12 for synergistic gene therapy of colon cancer. RSC Adv 2017. [DOI: 10.1039/c7ra03727a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Nanoparticles delivering therapeutic genes have promising applications in cancer treatments.
Collapse
|
12
|
Kostrzak A, Caval V, Escande M, Pliquet E, Thalmensi J, Bestetti T, Julithe M, Fiette L, Huet T, Wain-Hobson S, Langlade-Demoyen P. APOBEC3A intratumoral DNA electroporation in mice. Gene Ther 2016; 24:74-83. [PMID: 27858943 DOI: 10.1038/gt.2016.77] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 10/26/2016] [Accepted: 11/11/2016] [Indexed: 12/21/2022]
Abstract
Human APOBEC3A (A3A) cytidine deaminase shows pro-apoptotic properties resulting from hypermutation of genomic DNA, induction of double-stranded DNA breaks (DSBs) and G1 cell cycle arrest. Given this, we evaluated the antitumor efficacy of A3A by intratumoral electroporation of an A3A expression plasmid. DNA was repeatedly electroporated into B16OVA, B16Luc tumors of C57BL/6J mice as well as the aggressive fibrosarcoma Sarc2 tumor of HLA-A*0201/DRB1*0101 transgenic mice using noninvasive plate electrodes. Intratumoral electroporation of A3A plasmid DNA resulted in regression of ~50% of small B16OVA melanoma tumors that did not rebound in the following 2 months without treatment. Larger or more aggressive tumors escaped regression when so treated. As APOBEC3A was much less efficient in provoking hypermutation and DSBs in B16OVA cells compared with human or quail cells, it is likely that APOBEC3A would be more efficient in a human setting than in a mouse model.
Collapse
Affiliation(s)
- A Kostrzak
- Invectys, Pepinière Paris Biotech Santé Cochin, Paris, France
| | - V Caval
- Molecular Retrovirology Unit, Institut Pasteur, Paris, France
| | - M Escande
- Invectys, Pepinière Paris Biotech Santé Cochin, Paris, France
| | - E Pliquet
- Invectys, Pepinière Paris Biotech Santé Cochin, Paris, France
| | - J Thalmensi
- Invectys, Pepinière Paris Biotech Santé Cochin, Paris, France
| | - T Bestetti
- Invectys, Pepinière Paris Biotech Santé Cochin, Paris, France
| | - M Julithe
- Invectys, Pepinière Paris Biotech Santé Cochin, Paris, France
| | - L Fiette
- Human Histopathology and Animal Models, Infection & Epidemiology Department, Institut Pasteur, Paris, France.,Université Paris Descartes, Sorbonne Paris-Cité, Paris, France
| | - T Huet
- Invectys, Pepinière Paris Biotech Santé Cochin, Paris, France
| | - S Wain-Hobson
- Invectys, Pepinière Paris Biotech Santé Cochin, Paris, France.,Molecular Retrovirology Unit, Institut Pasteur, Paris, France
| | - P Langlade-Demoyen
- Invectys, Pepinière Paris Biotech Santé Cochin, Paris, France.,Molecular Retrovirology Unit, Institut Pasteur, Paris, France
| |
Collapse
|
13
|
Wen S, Zhang J, Zhou P, Luo C, Liu Y, Xu Z, Chen X, Ma H. The anti-tumour effect of a DNA vaccine carrying a fusion gene of human VEGFR2 and IL-12. BIOTECHNOL BIOTEC EQ 2016. [DOI: 10.1080/13102818.2016.1207488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Sha Wen
- Department of Geriatrics, The First Affiliated Hospital, Chongqing Medical University, Chongqing, P.R. China
| | - Jia Zhang
- Department of Geriatrics, The First Affiliated Hospital, Chongqing Medical University, Chongqing, P.R. China
| | - Ping Zhou
- Department of Geriatrics, The First Affiliated Hospital, Chongqing Medical University, Chongqing, P.R. China
| | - Cheng Luo
- Department of Geriatrics, The First Affiliated Hospital, Chongqing Medical University, Chongqing, P.R. China
| | - Yingfu Liu
- Department of Cell Biology, Logistics University of China People's Armed Police Forces, Tianjin, P.R. China
| | - Zhongwei Xu
- Central Laboratory, Logistics University of China People's Armed Police Forces, Tianjin, P.R. China
| | - Xiaoyi Chen
- Department of Cell Biology, Logistics University of China People's Armed Police Forces, Tianjin, P.R. China
| | - Houxun Ma
- Department of Geriatrics, The First Affiliated Hospital, Chongqing Medical University, Chongqing, P.R. China
| |
Collapse
|
14
|
Cicchelero L, Denies S, Haers H, Vanderperren K, Stock E, Van Brantegem L, de Rooster H, Sanders NN. Intratumoural interleukin 12 gene therapy stimulates the immune system and decreases angiogenesis in dogs with spontaneous cancer. Vet Comp Oncol 2016; 15:1187-1205. [PMID: 27506827 DOI: 10.1111/vco.12255] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 06/22/2016] [Accepted: 07/03/2016] [Indexed: 12/13/2022]
Abstract
Interleukin 12 (IL-12) is a powerful immunostimulatory cytokine with a strong antitumoural activity. In this work, the immunological, anti-angiogenic and clinical effects of three consecutive intratumoural IL-12 electrogene therapy (EGT) treatments were evaluated in nine dogs with spontaneous cancer. In all the dogs, tumour biopsies and blood samples were taken prior, during and after the intratumoural IL-12 EGT (on days 1, 8, 35 and 1, 3, 8, 15, 35, respectively). An initial decrease in immune cells was followed by an increase above baseline 1-3 weeks after treatment initiation. Interestingly, the decrease in peripheral leukocytes 2 days after the first intratumoural IL-12 EGT coincided with erythema and tumour swelling. Transient increases of IL-12 and interferon γ were measured in the serum and the tumour tissue, whereas IL-10 transiently increased only in the serum. The effect of intratumoural IL-12 EGT on the levels of IL-24 and vascular endothelial growth factor in the sera and tumour biopsies differed per dog. Via contrast-enhanced ultrasound (US) (on days 1, 8 and 35), we demonstrated that intratumoural IL-12 EGT resulted in a significant decrease of the relative blood volume and blood flow speed in the tumour compared with baseline. Metastases were present in two dogs. In one of these dogs, IL-12 EGT of the primary tumour caused a transient partial regression of the metastases, but not of the primary tumour. The second dog with metastases did not survive long enough to complete the entire treatment cycle. Despite encouraging immunostimulatory and anti-angiogenic effects after intratumoural IL-12 EGT, no clinically relevant outcomes were observed in this study, as persistent tumour regression could not be obtained. On the other hand, the laboratory and US results hold great promise for combinatorial strategies of intratumoural IL-12 EGT with conventional antitumour (immuno)therapies.
Collapse
Affiliation(s)
- L Cicchelero
- Faculty of Veterinary Medicine, Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Ghent University, Merelbeke, Belgium
| | - S Denies
- Faculty of Veterinary Medicine, Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Ghent University, Merelbeke, Belgium
| | - H Haers
- Faculty of Veterinary Medicine, Department of Medical Imaging of Domestic Animals, Ghent University, Merelbeke, Belgium
| | - K Vanderperren
- Faculty of Veterinary Medicine, Department of Medical Imaging of Domestic Animals, Ghent University, Merelbeke, Belgium
| | - E Stock
- Faculty of Veterinary Medicine, Department of Medical Imaging of Domestic Animals, Ghent University, Merelbeke, Belgium
| | - L Van Brantegem
- Faculty of Veterinary Medicine, Department of Pathology, Bacteriology and Poultry Diseases, Ghent University, Merelbeke, Belgium
| | - H de Rooster
- Small Animal Hospital, Faculty of Veterinary Medicine, Department of Medicine and Clinical Biology of Small Animals, Ghent University, Merelbeke, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - N N Sanders
- Faculty of Veterinary Medicine, Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Ghent University, Merelbeke, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
15
|
Gao M, Zhu X, Wu L, Qiu L. Cationic Polyphosphazene Vesicles for Cancer Immunotherapy by Efficient in Vivo Cytokine IL-12 Plasmid Delivery. Biomacromolecules 2016; 17:2199-209. [DOI: 10.1021/acs.biomac.6b00433] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Menghua Gao
- College
of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Xiumei Zhu
- College
of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Liping Wu
- College
of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Liyan Qiu
- Ministry
of Education (MOE) Key Laboratory of Synthesis and Functionalization,
Department of Polymer Science and Engineering, Zhejiang University, 38 Zheda Road, Hangzhou 310027, China
| |
Collapse
|
16
|
Hu J, Zhu S, Xia X, Zhang L, Kleinerman ES, Li S. CD8+T cell-specific induction of NKG2D receptor by doxorubicin plus interleukin-12 and its contribution to CD8+T cell accumulation in tumors. Mol Cancer 2014; 13:34. [PMID: 24565056 PMCID: PMC3938086 DOI: 10.1186/1476-4598-13-34] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 02/03/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Increased infiltration of CD8+T cells into tumors has a positive impact on survival. Our previous study showed that doxorubicin (Dox) plus interleukin-12 (IL-12) boosted the accumulation of CD8+T cells in tumors and had a greater antitumor effect than did either agent alone. The purpose of this study was to determine the impact of NKG2D expression on CD8+T cell infiltration and antitumor efficacy. METHODS Tumor-bearing mice were administered Dox, IL-12 plasmid DNA, or both via intraperitoneal injection or intramuscular electroporation. The induction of NKG2D on CD8+T cells and other lymphocytes was analyzed via flow cytometry, and NKG2D-positive CD8+T cell-specific localization in tumors was determined by using immunofluorescence staining in various types of immune cell-depleted mice. RESULTS The combination of Dox plus IL-12 specifically increased expression of NKG2D in CD8+T cells but not in other types of immune cells, including NK cells, which naturally express NKG2D. This induced NKG2D expression in CD8+T cells was associated with increased accumulation of CD8+T cells in murine tumors. Administration of NKG2D-blocking antibody or CD8+T cell-depletion antibody abrogated the NKG2D+CD8+T cell detection in tumors, whereas administration of NK cell-depletion antibody had no effect. Increased NKG2D expression in CD8+T cells was associated with increased antitumor efficacy in vivo. CONCLUSION We conclude that Dox plus IL-12 induces NKG2D in CD8+T cells in vivo and boosts NKG2D+CD8+T-dependent antitumor immune surveillance. This discovery reveals a novel mechanism for how chemoimmunotherapy synergistically promotes T cell-mediated antitumor immune surveillance.
Collapse
Affiliation(s)
| | | | | | | | | | - Shulin Li
- Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
17
|
Ribatti D, Moschetta M, Vacca A. Macrophages in multiple myeloma. Immunol Lett 2013; 161:241-4. [PMID: 24370642 DOI: 10.1016/j.imlet.2013.12.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 12/13/2013] [Indexed: 12/30/2022]
Abstract
Tumor associated macrophages (TAMs) are a rich source of pro-angiogenic cytokines and growth factors, and a relationship between the TAMs content, the rate of tumor growth and the extent of vascularization has been shown in several tumors. In this article, we have summarized the literature and our data concerning the involvement of TAMs in angiogenesis occurring in multiple myeloma. Finally, therapeutic aspects concerning the potential role of molecules which inhibit macrophage recruitment in the tumor side are also discussed.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy; National Cancer Institute "Giovanni Paolo II", Bari, Italy.
| | - Michele Moschetta
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Bari, Italy
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Bari, Italy.
| |
Collapse
|
18
|
Beebe SJ, Sain NM, Ren W. Induction of Cell Death Mechanisms and Apoptosis by Nanosecond Pulsed Electric Fields (nsPEFs). Cells 2013; 2:136-62. [PMID: 24709649 PMCID: PMC3972658 DOI: 10.3390/cells2010136] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Revised: 02/05/2013] [Accepted: 02/21/2013] [Indexed: 12/21/2022] Open
Abstract
Pulse power technology using nanosecond pulsed electric fields (nsPEFs) offers a new stimulus to modulate cell functions or induce cell death for cancer cell ablation. New data and a literature review demonstrate fundamental and basic cellular mechanisms when nsPEFs interact with cellular targets. NsPEFs supra-electroporate cells creating large numbers of nanopores in all cell membranes. While nsPEFs have multiple cellular targets, these studies show that nsPEF-induced dissipation of ΔΨm closely parallels deterioration in cell viability. Increases in intracellular Ca2+ alone were not sufficient for cell death; however, cell death depended of the presence of Ca2+. When both events occur, cell death ensues. Further, direct evidence supports the hypothesis that pulse rise-fall times or high frequency components of nsPEFs are important for decreasing ΔΨm and cell viability. Evidence indicates in Jurkat cells that cytochrome c release from mitochondria is caspase-independent indicating an absence of extrinsic apoptosis and that cell death can be caspase-dependent and -independent. The Ca2+ dependence of nsPEF-induced dissipation of ΔΨm suggests that nanoporation of inner mitochondria membranes is less likely and effects on a Ca2+-dependent protein(s) or the membrane in which it is embedded are more likely a target for nsPEF-induced cell death. The mitochondria permeability transition pore (mPTP) complex is a likely candidate. Data demonstrate that nsPEFs can bypass cancer mutations that evade apoptosis through mechanisms at either the DISC or the apoptosome.
Collapse
Affiliation(s)
- Stephen J Beebe
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, 4211 Monarch Way, IRP2, Norfolk, Virginia, 23508, USA.
| | - Nova M Sain
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, 4211 Monarch Way, IRP2, Norfolk, Virginia, 23508, USA.
| | - Wei Ren
- Division of Molecular Carcinogenesis and Targeted Therapy for Cancer, Chinese Academy of Sciences, 1 Beichen West Road, Beijing 100101, China.
| |
Collapse
|
19
|
Multiple myeloma macrophages: pivotal players in the tumor microenvironment. JOURNAL OF ONCOLOGY 2013; 2013:183602. [PMID: 23431298 PMCID: PMC3570938 DOI: 10.1155/2013/183602] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 01/03/2013] [Indexed: 01/20/2023]
Abstract
Tumor microenvironment is essential for multiple myeloma (MM) growth, progression, and drug resistance through provision of survival signals and secretion of growth and proangiogenic factors. This paper examines the importance of macrophages within MM bone marrow (BM) microenvironment, referred to as MM-associated macrophages, as a potential niche component that supports tumor plasma cells. These macrophages are derived from peripheral blood monocytes recruited into the tumor. Upon activation by MM plasma cells and mesenchymal stromal cells, macrophages can release growth factors, proteolytic enzymes, cytokines, and inflammatory mediators that promote plasma cell growth and survival. Macrophages promote tumor progression through several mechanisms including angiogenesis, growth, and drug resistance. Indeed, these macrophages are essential for the induction of an angiogenic response through vasculogenic mimicry, and this ability proceeds in step with progression of the plasma cell tumors. Data suggest that macrophages play an important role in the biology and survival of patients with MM, and they may be a target for the MM antivascular management.
Collapse
|
20
|
Abstract
Intratumoral gene electroporation uses electric charges to facilitate entry of plasmid DNA into cells in a reproducible and highly efficient manner, especially to accessible sites such as cutaneous and subcutaneous melanomas. Effective for locally treated disease, electroporation of plasmid DNA encoding interleukin-12 can also induce responses in untreated distant disease, suggesting that adaptive immune responses are being elicited that can target melanoma-associated antigens. In vivo electroporation with immunomodulatory cytokine DNA is a promising approach that can trigger systemic anti-tumor immune responses without the systemic toxicity associated with intravenous cytokine delivery and potentially offer complete long-term tumor regression.
Collapse
Affiliation(s)
- Edward Cha
- Department of Medicine; University of California-San Francisco, CA, USA.
| | | |
Collapse
|
21
|
Kim J, Denu RA, Dollar BA, Escalante LE, Kuether JP, Callander NS, Asimakopoulos F, Hematti P. Macrophages and mesenchymal stromal cells support survival and proliferation of multiple myeloma cells. Br J Haematol 2012; 158:336-46. [PMID: 22583117 DOI: 10.1111/j.1365-2141.2012.09154.x] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 03/27/2012] [Indexed: 02/05/2023]
Abstract
Multiple myeloma (MM) is characterized by almost exclusive tropism of malignant cells for the bone marrow (BM) milieu. The survival and proliferation of malignant plasma cells have been shown to rely on interactions with nonmalignant stromal cells, in particular mesenchymal stromal cells (MSCs), in the BM microenvironment. However, the BM microenvironment is composed of a diverse array of cell types. This study examined the role of macrophages, an abundant component of BM stroma, as a potential niche component that supports malignant plasma cells. We investigated the proliferation of MM tumour cell lines when cultured alone or together with MSCs, macrophages, or a combination of MSCs and macrophages, using the carboxyfluorescein succinimidyl ester assay. Consistently, we observed increased proliferation of MM cell lines in the presence of either MSCs or macrophages compared to cell line-only control. Furthermore, the combined co-culture of MSCs plus macrophages induced the greatest degree of proliferation of myeloma cells. In addition to increased proliferation, MSCs and macrophages decreased the rate of apoptosis of myeloma cells. Our in vitro studies provide evidence that highlights the role of macrophages as a key component of the BM microenvironment facilitating the growth of malignant plasma cells in MM.
Collapse
Affiliation(s)
- Jaehyup Kim
- Department of Medicine, University of Wisconsin-Madison, School of Medicine and Public Health, USA.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Prijic S, Prosen L, Cemazar M, Scancar J, Romih R, Lavrencak J, Bregar VB, Coer A, Krzan M, Znidarsic A, Sersa G. Surface modified magnetic nanoparticles for immuno-gene therapy of murine mammary adenocarcinoma. Biomaterials 2012; 33:4379-91. [PMID: 22429983 DOI: 10.1016/j.biomaterials.2012.02.061] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 02/16/2012] [Indexed: 11/27/2022]
Abstract
Cancer immuno-gene therapy is an introduction of nucleic acids encoding immunostimulatory proteins, such as cytokine interleukin 12 (IL-12), into somatic cells to stimulate an immune response against a tumor. Various methods can be used for the introduction of nucleic acids into cells; magnetofection involves binding of nucleic acids to magnetic nanoparticles with subsequent exposure to an external magnetic field. Here we show that surface modified superparamagnetic iron oxide nanoparticles (SPIONs) with a combination of polyacrylic acid (PAA) and polyethylenimine (PEI) (SPIONs-PAA-PEI) proved to be safe and effective for magnetofection of cells and tumors in mice. Magnetofection of cells with plasmid DNA encoding reporter gene using SPIONs-PAA-PEI was superior in transfection efficiency to commercially available SPIONs. Magnetofection of murine mammary adenocarcinoma with plasmid DNA encoding IL-12 using SPIONs-PAA-PEI resulted in significant antitumor effect and could be further refined for cancer immuno-gene therapy.
Collapse
Affiliation(s)
- Sara Prijic
- Kolektor Group, Nanotesla Institute, Ljubljana, Slovenia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Pre-clinical toxicity assessment of tumor-targeted interleukin-12 low-intensity electrogenetherapy. Cancer Gene Ther 2011; 18:265-74. [PMID: 21233859 DOI: 10.1038/cgt.2010.77] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
This study's goal was to assess the safety of tumor-targeted interleukin-12 (ttIL-12) when administered by electrogenetherapy in C3H/HeJ mice by identifying an initial safe dose for human dose escalation schemes, toxicity target organs, markers of toxicity, and toxicity reversibility. Tumor-free mice receiving two doses of 0.45% NaCl, 1 μg ttIL-12 DNA in 0.45% NaCl or 5 μg ttIL-12 DNA in 0.45% NaCl, 10 days apart combined with low-intensity electroporation were compared with non-treatment controls over time. All mice had blood cell counts, serum chemistry profiles, plasma interleukin-12 and IFNγ determinations, necropsy and multi-organ histopathology. Mild treatment-associated changes included electroporation-associated muscle changes that resolved by 30 days; decreased total white blood cell counts and infectious disease in the 5 μg ttIL-12 group, but not in the 1 μg group, and liver changes in ttIL-12 groups that correlated with alanine transaminase levels and resolved by 30 days. Dystrophic cardiac calcification seen in older, 5 μg ttIL-12-treated mice was the only serious toxicity. Based on these results and the lack of any effect on wound healing when combined with surgery, low-intensity electrogenetherapy with ttIL-12 appears to be safe and well tolerated.
Collapse
|
24
|
Heller LC, Cruz YL, Ferraro B, Yang H, Heller R. Plasmid injection and application of electric pulses alter endogenous mRNA and protein expression in B16.F10 mouse melanomas. Cancer Gene Ther 2010; 17:864-71. [PMID: 20706286 PMCID: PMC2981654 DOI: 10.1038/cgt.2010.43] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Revised: 02/22/2010] [Accepted: 04/23/2010] [Indexed: 11/08/2022]
Abstract
The application of electric pulses to tissues causes cell membrane destabilization, allowing exogenous molecules to enter the cells. This delivery technique can be used for plasmid gene therapy. Reporter gene expression after plasmid delivery with eight representative published protocols was compared in B16.F10 mouse melanoma tumors. This expression varied significantly based on the pulse parameters utilized for delivery. To observe the possible influence of plasmid injection and/or pulse application on endogenous gene expression, levels of stress-related mRNAs 4 and 24 h after delivery were determined by PCR array. Increases in mRNA levels for several inflammatory chemokines and cytokines were observed in response to plasmid injection, electric pulses alone or the combination. This upregulation was confirmed by individual real-time reverse transcription TaqMan PCR assays. Proteins were extracted at the same time points from identically treated tumors and inflammatory protein levels were assayed by enzyme-linked immunosorbent assay and by a custom multiplex bead array. Increases in inflammatory protein levels generally paralleled mRNA levels. Some differences were observed, which may have been due to differing expression kinetics. The observed upregulated expression of these cytokines and chemokines may aid or inhibit the therapeutic effectiveness of immune-based cancer gene therapies.
Collapse
Affiliation(s)
- L C Heller
- Department of Medical Laboratory and Radiation Sciences, Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA.
| | | | | | | | | |
Collapse
|
25
|
Zhu S, Lee DA, Li S. IL-12 and IL-27 sequential gene therapy via intramuscular electroporation delivery for eliminating distal aggressive tumors. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:2348-54. [PMID: 20139275 PMCID: PMC2824785 DOI: 10.4049/jimmunol.0902371] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Eradication of residual malignancies and metastatic tumors via a systemic approach is the key for successfully treating cancer and increasing cancer patient survival. Systemic administration of IL-12 protein in an acute large dose is effective but toxic. Systemic administration of IL-12 gene by persistently expressing a low level of IL-12 protein may reduce the systemic toxicity but only eradicates IL-12-sensitive tumors. In this study, we discovered that sequential administration of IL-12- and IL-27-encoding DNA, referred to as sequential IL-12-->IL-27 (IL-12 administration followed by IL-27 administration 10 d after) gene therapy, not only eradicated IL-12-sensitive CT26 tumors from 100% of mice but also eradicated the highly malignant 4T1 tumors from 33% of treated mice in multiple independent experiments. This IL-12-->IL-27 sequential gene therapy is not only superior to IL-12-encoding plasmid DNA given a total of two times at a 10-d interval sequential gene therapy for eliminating tumors but also for inducing CTL activity, increasing T cell infiltration into tumors, and yielding a large number of tumor-specific IFN-gamma-positive CD8 T cells. Notably, depletion of either T or NK cells during the IL-27 treatment phase reverses tumor eradication, suggesting an NK cell requirement for this sequential gene therapy-mediated tumor eradication. Both reversal of the administration sequence and coadministration of IL-12 and IL-27 impaired tumor eradication in 4T1 tumor-bearing mice. This IL-12-->IL-27 sequential gene therapy, via sequential administration of IL-12- and IL-27-encoding plasmid DNA into tumor-bearing mice through i.m. electroporation, provides a simple but effective approach for eliminating inaccessible residual tumors.
Collapse
Affiliation(s)
- Shiguo Zhu
- School of Medicine & Institute of Medical Science, Shanghai Jiao Tong University, 280 Chongqing South Road, Shanghai 200025, PR China
| | - Dean Anthony Lee
- The University of Texas MD Anderson Cancer Center, Division of Pediatrics, Houston, TX 77030
| | - Shulin Li
- Department of Comparative Biomedical Sciences, Louisiana State University, Skip Bertman Drive, Baton Rouge, LA 70803
| |
Collapse
|
26
|
Fewell JG, Matar MM, Rice JS, Brunhoeber E, Slobodkin G, Pence C, Worker M, Lewis DH, Anwer K. Treatment of disseminated ovarian cancer using nonviral interleukin-12 gene therapy delivered intraperitoneally. J Gene Med 2009; 11:718-28. [PMID: 19507172 DOI: 10.1002/jgm.1356] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND The poor prognosis associated with ovarian cancer is primarily the result of delayed diagnosis and the lack of an effective treatment for advanced disease. Use of novel immunotherapy strategies are being evaluated that work to enhance local and systemic immune responses against cancer cells and can possibly work together with traditional cytotoxic chemotherapy regimens to produce more effective treatment options. METHODS In the present study, we describe a gene-based therapy whereby the anticancer cytokine interleukin-12 gene (pmIL-12) is formulated with a synthetic polymeric delivery vehicle (PPC) and administered intraperitoneally into a mouse model of disseminated ovarian cancer. RESULTS The administration of pmIL-12/PPC in tumor-bearing mice was associated with a shift towards a Th1 immune state, including significant increases in murine IL-12 (mIL-12) and interferon (IFN)-gamma (mIFN-gamma) in ascites fluid, with little change in systemic levels of these proteins. The mIL-12 protein was detectable for several days and could be reintroduced with subsequent injections. We show that treatment delayed the onset of ascites formation and improved survival in a dose-dependent manner. A significant decrease in vascular endothelial growth factor was associated with pmIL-12/PPC delivery and believed to play a predominant role in inhibiting ascites accumulation. Administration of pmIL-12/PPC was associated with minimal toxicity and, when combined with standard chemotherapies, resulted in additive improvement in survival. CONCLUSIONS Taken together, these results suggest that pmIL-12/PPC may be an effective strategy for inhibiting progression of disseminated ovarian cancer and may offer a new option for treatment of advanced disease that can be used to complement standard therapies.
Collapse
|
27
|
Enhancement of reporter gene detection sensitivity by insertion of specific mini-peptide-coding sequences. Cancer Gene Ther 2009; 17:131-40. [PMID: 19713998 DOI: 10.1038/cgt.2009.54] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Two important aspects of gene therapy are to increase the level of gene expression and track the gene delivery site and expression, and a sensitive reporter gene may be one of the options for preclinical studies and possibly for human clinical trials. We report the novel concept of increasing the activity of the gene products. With the insertion of the mini-peptide-coding sequence CWDDWLC into the plasmid DNA of a SEAP reporter gene, we observed vast increases in the enzyme activity in vitro in all murine and human cell lines used. In addition, in vivo injection of this CWDDWLC-SEAP-encoding gene resulted in the same increases in reporter gene activity, but these increases did not correspond to alterations in the level of the gene products in the serum. Minor sequence changes in this mini-peptide negate the activity increase of the reporter gene. We report the novel concept of increasing the activity of gene products as another method to improve the reporting sensitivity of reporter genes. This improved reporter gene could complement any improved vector for maximizing the reporter sensitivity. Moreover, this strategy has the potential to be used to discover peptides that improve the activity of therapeutic genes.
Collapse
|
28
|
Tsai YS, Shiau AL, Chen YF, Tsai HT, Lee HL, Tzai TS, Wu CL. Enhancement of antitumor immune response by targeted interleukin-12 electrogene transfer through antiHER2 single-chain antibody in a murine bladder tumor model. Vaccine 2009; 27:5383-92. [DOI: 10.1016/j.vaccine.2009.06.079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2009] [Revised: 06/21/2009] [Accepted: 06/22/2009] [Indexed: 01/29/2023]
|
29
|
Policastro LL, Ibañez IL, Durán HA, Soria G, Gottifredi V, Podhajcer OL. Suppression of cancer growth by nonviral gene therapy based on a novel reactive oxygen species-responsive promoter. Mol Ther 2009; 17:1355-64. [PMID: 19436270 DOI: 10.1038/mt.2009.103] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Increased reactive oxygen species (ROS) production has been reported as a distinctive feature of different pathologies including cancer. Therefore, we assessed whether increased ROS production in the cancer microenvironment could be selectively exploited to develop a selective anticancer therapy. For this purpose, we constructed a novel chimeric promoter, based on a ROS-response motif located in the VEGF gene promoter placed, in turn, downstream of a second ROS-response motif obtained from the early growth response 1 (Egr-1) gene promoter. The activity of the chimeric promoter was largely dependent on variations in intracellular ROS levels and showed a high inducible response to exogenous H(2)O(2). Transient expression of the thymidine kinase (TK) gene driven by the chimeric promoter, followed by gancyclovir (GCV) administration, inhibited human colorectal cancer and melanoma cell growth in vitro and in vivo. Moreover, electrotransfer of the TK gene followed by GCV administration exerted a potent therapeutic effect on established tumors. This response was improved when combined with chemotherapeutic drugs. Thus, we show for the first time that a distinctive pro-oxidant state can be used to develop new selective gene therapeutics for cancer.
Collapse
Affiliation(s)
- Lucía L Policastro
- Department of Radiobiology, National Atomic Energy Commission-CONICET, Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
30
|
Matsui M, Kishida T, Nakano H, Yoshimoto K, Shin-Ya M, Shimada T, Nakai S, Imanishi J, Yoshimoto T, Hisa Y, Mazda O. Interleukin-27 Activates Natural Killer Cells and Suppresses NK-Resistant Head and Neck Squamous Cell Carcinoma through Inducing Antibody-Dependent Cellular Cytotoxicity. Cancer Res 2009; 69:2523-30. [DOI: 10.1158/0008-5472.can-08-2793] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
31
|
Daud AI, DeConti RC, Andrews S, Urbas P, Riker AI, Sondak VK, Munster PN, Sullivan DM, Ugen KE, Messina JL, Heller R. Phase I trial of interleukin-12 plasmid electroporation in patients with metastatic melanoma. J Clin Oncol 2008; 26:5896-903. [PMID: 19029422 DOI: 10.1200/jco.2007.15.6794] [Citation(s) in RCA: 448] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Gene-based immunotherapy for cancer is limited by the lack of safe, efficient, reproducible, and titratable delivery methods. Direct injection of DNA into tissue, although safer than viral vectors, suffers from low gene transfer efficiency. In vivo electroporation, in preclinical models, significantly enhances gene transfer efficiency while retaining the safety advantages of plasmid DNA. PATIENTS AND METHODS A phase I dose escalation trial of plasmid interleukin (IL)-12 electroporation was carried out in patients with metastatic melanoma. Patients received electroporation on days 1, 5, and 8 during a single 39-day cycle, into metastatic melanoma lesions with six 100-mus pulses at a 1,300-V/cm electric field through a penetrating six-electrode array immediately after DNA injection. Pre- and post-treatment biopsies were obtained at defined time points for detailed histologic evaluation and determination of IL-12 protein levels. RESULTS Twenty-four patients were treated at seven dose levels, with minimal systemic toxicity. Transient pain after electroporation was the major adverse effect. Post-treatment biopsies showed plasmid dose proportional increases in IL-12 protein levels as well as marked tumor necrosis and lymphocytic infiltrate. Two (10%) of 19 patients with nonelectroporated distant lesions and no other systemic therapy showed complete regression of all metastases, whereas eight additional patients (42%) showed disease stabilization or partial response. CONCLUSION This report describes the first human trial, to our knowledge, of gene transfer utilizing in vivo DNA electroporation. The results indicated this modality to be safe, effective, reproducible, and titratable.
Collapse
Affiliation(s)
- Adil I Daud
- Cutaneous Oncology and Experimental Therapeutics Programs, H. Lee Moffitt Cancer Center, University of South Florida, Tampa, FL, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Administering Plasmid DNA Encoding Tumor Vessel–anchored IFN-α for Localizing Gene Product Within or Into Tumors. Mol Ther 2008; 16:901-906. [DOI: 10.1038/mt.2008.40] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2007] [Accepted: 02/14/2008] [Indexed: 11/08/2022] Open
|
33
|
Pavlin D, Tozon N, Sersa G, Pogacnik A, Cemazar M. Efficient electrotransfection into canine muscle. Technol Cancer Res Treat 2008; 7:45-54. [PMID: 18198924 DOI: 10.1177/153303460800700106] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Two different types of electroporation protocols have been developed for efficient electrotransfer of plasmid DNA into skeletal muscle of experimental animals. At first, only low voltage electric pulses have been used, but lately, a combination of high and low voltage pulses has been suggested as more efficient. Up to date, in dogs, this type of electroporation protocol has never been used for muscle targeted plasmid DNA electrotransfection. In this study, we used two different DNA plasmids, one encoding green fluorescent protein and one encoding human interleukin-12. Five different electroporation protocols were evaluated. Three of them featured different combinations of high and low voltage pulses, and two were performed with delivery of low voltage pulses only. Our study shows that combination of 1 high voltage pulse (600 V/cm, 100 mus), followed by 4 low voltage pulses (80 V/cm, 100 ms, 1 Hz) yielded in the same transfection efficiency as the standard trains of low voltage pulses. However, this protocol is performed quicker and, thus, more suitable for potential use in clinical practice. In addition, it yielded in detectable systemic expression of human interleukin-12. Electrotransfer of either of the plasmids was associated with only mild and transitory local side effects, without clinically detectable systemic side effects. The results indicate that electrotransfection is a feasible, effective, and safe method for muscle targeted gene therapy in dogs, which could have potential for clinical applications in veterinary medicine of small animals.
Collapse
Affiliation(s)
- D Pavlin
- University of Ljubljana, Veterinary Faculty Ljubljana, Gerbiceva 60, SI-1000 Ljubljana, Slovenia
| | | | | | | | | |
Collapse
|
34
|
Systemic IL-12 gene therapy for treating malignancy via intramuscular electroporation. Methods Mol Biol 2008. [PMID: 18370211 DOI: 10.1007/978-1-59745-194-9_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Interleukin 12 (IL-12) is effective in treating systemic microscopic malignancies by inducing T helper 1 (T(H)1) response, inhibiting angiogenesis, and triggering secondary cytokine production. Unfortunately, daily systemic administration of an acute dose of IL-12 protein is very costly and severely toxic. Here, a simple, economic, and less toxic approach, intramuscular administration of IL-12 gene, is provided for treating tumors in three tumor models. The results indicate that intramuscular administration of IL-12 encoding plasmid DNA via electroporation is a promising technology for treating systemic residual malignancies (less than 3-5 mm in diameter), as illustrated by the inhibition of tumor growth and lung metastases as well as the extension of survival rate. This approach is not effective in treating tumors larger than 3-5 mm in diameter.
Collapse
|
35
|
Abstract
Delivery of plasmid DNA encoding therapeutic genes into tumors is one of the main applications of electroporation. This chapter summarizes various investigators' electroporation parameters for intratumoral gene delivery. In addition to electroporation parameters, injection volume is also critical for achieving a high level of gene expression via electroporation. In this study, we attempt to provide a strategy for determining the optimal injection volume for intratumoral injection via electroporation. Unlike muscle tissues, the optimal volume for gene delivery into tumors via electroporation may vary greatly based on the tumor size and the electroporation parameters. More efforts in defining the optimal injection volume should be made to further advance intratumoral electroporation gene therapy for treating tumors.
Collapse
Affiliation(s)
- Shulin Li
- Department of Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, LA, USA
| |
Collapse
|
36
|
Zhu S, Waguespack M, Barker SA, Li S. Doxorubicin Directs the Accumulation of Interleukin-12–Induced IFNγ into Tumors for Enhancing STAT1–Dependent Antitumor Effect. Clin Cancer Res 2007; 13:4252-60. [PMID: 17634555 DOI: 10.1158/1078-0432.ccr-06-2894] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To examine the mechanism by which doxorubicin plus interleukin-12 (IL-12) gene transfer induces enhanced therapeutic efficacy against tumors. EXPERIMENTAL DESIGN Tumor-bearing mice were treated with doxorubicin, IL-12-encoding plasmid DNA, doxorubicin plus IL-12-encoding plasmid DNA, or plasmid DNA control. Doxorubicin was systemically given via i.p. injection, and IL-12 was systemically expressed via i.m. injection. To show that doxorubicin enhances the accumulation of IL-12-induced IFN gamma into tumors and the signal transducer and activator of transcription 1 (Stat1)-dependent antitumor efficacy, the distribution of IFN gamma and the therapeutic end points, such as T-cell infiltration, inhibition of tumor vessel density, tumor growth inhibition, and inhibition of spontaneous tumor metastasis in wild-type and Stat1(-/-) host and tumors were determined after the treatment at the indicated time points. RESULTS In this study, a novel mechanism was unveiled. We discovered that doxorubicin enhances the accumulation of IL-12-induced IFN gamma in tumors. The doxorubicin-mediated accumulation of IFN gamma in tumors is caused by an increased accumulation of IFN gamma-secreting immune cells and not by a direct translocation of IFN gamma protein into tumors. Depletion of immune cells reverses the doxorubicin-mediated accumulation of IFN gamma into tumors and reverses the inhibition of tumor vessel density induced by coadministration of doxorubicin and IL-12 DNA. Knocking out IFN gamma signaling in the tumor host reverses the significant inhibition of tumor growth by coadministration of doxorubicin and IL-12. CONCLUSIONS The enhanced antitumor efficacy by coadministration of doxorubicin and IL-12 is dependent on the accumulation of IFN gamma in tumors. This discovery provides a possible strategy to reduce side effects caused by IL-12.
Collapse
Affiliation(s)
- Shiguo Zhu
- Department of Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, Louisiana 70803, USA
| | | | | | | |
Collapse
|
37
|
Affiliation(s)
- Loree C Heller
- Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | | |
Collapse
|
38
|
Liu J, Xia X, Torrero M, Barrett R, Shillitoe EJ, Li S. The mechanism of exogenous B7.1-enhanced IL-12-mediated complete regression of tumors by a single electroporation delivery. Int J Cancer 2006; 119:2113-8. [PMID: 16823840 DOI: 10.1002/ijc.22100] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Electroporation-based mono-gene therapy has received great interest in recent years but coadministration of different therapeutic genes for treatment of tumors has not been well explored. We hypothesize that electroporation is capable of delivering multiple genes that induce an additive or synergistic antitumor effect. To test this hypothesis, we used mice that were bearing SCCVII or TRAMP tumors. Established tumors with a diameter of 4-5 mm were injected with control plasmid DNA or plasmid DNA encoding B7.1, IL-12 or both via electroporation. Tumor regression, CTL activity and the level of B7.1, IL-12 and Stat1 expression were determined in both wild-type mice and in mice with a knock-out of the Stat1 gene. Remarkably, a single coadministration of the plasmids that encoded IL-12 and B7.1 eradicated tumors in 80% of mice. The therapeutic effect was associated with high levels of endogenous B7.1 expression, activity of cytotoxic lymphocytes, and activation of Stat1. Both exogenous B7.1 and IL-12 were required for inducing a high level of Stat1 activation in tumors, which occurred through a mechanism that was independent of the host Stat1. Both stimulators were also required for inducing the strong cytotoxic lymphocyte activity and for increasing the level and extending the duration of endogenous B7.1 expression. We therefore propose a 2-signal stimulation model to explain the synergistic effect of the coadministration of IL-12 and B7.1 on the regression of the tumors.
Collapse
Affiliation(s)
- Jianguo Liu
- Department of Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | | | | | | | | | | |
Collapse
|
39
|
Abstract
The utilisation of nonviral gene delivery methods has been increasing steadily, however, a drawback has been the relative low efficiency of gene transfer with naked DNA compared with viral delivery methods. In vivo electroporation, which has previously been used clinically to deliver chemotherapeutic agents, also enhances the delivery of plasmid DNA and has been used to deliver plasmids to several tissue types, particularly muscle and tumour. Recently, a large number of preclinical studies for a variety of therapeutic modalities have demonstrated the potential of electrically mediated gene transfer. Although clinical trials using gene transfer with in vivo electroporation have not as yet been realised, the tremendous growth of this technology suggests that the first trials will soon be initiated.
Collapse
MESH Headings
- Animals
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/therapy
- Electroporation
- Erythropoietin/administration & dosage
- Erythropoietin/genetics
- Erythropoietin/metabolism
- Forecasting
- Gene Expression Regulation
- Gene Transfer Techniques
- Genetic Therapy/methods
- Genetic Therapy/trends
- Growth Substances/administration & dosage
- Growth Substances/genetics
- Growth Substances/metabolism
- Hematologic Diseases/genetics
- Hematologic Diseases/metabolism
- Hematologic Diseases/therapy
- Humans
- Injections, Intramuscular
- Interleukin-12/administration & dosage
- Interleukin-12/genetics
- Interleukin-12/metabolism
- Melanoma/genetics
- Melanoma/metabolism
- Melanoma/therapy
- Plasmids/administration & dosage
- Plasmids/genetics
- Plasmids/metabolism
- Protein Deficiency/genetics
- Protein Deficiency/metabolism
- Protein Deficiency/therapy
- Toxins, Biological/administration & dosage
- Toxins, Biological/genetics
- Toxins, Biological/metabolism
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Vaccines, DNA/metabolism
Collapse
Affiliation(s)
- Loree C Heller
- University of South Florida, Center of Molecular Delivery, Department of Medical Microbiology and Immunology, Center for Molecular Delivery, College of Medicine, Tampa, 33612-4799, USA.
| | | | | |
Collapse
|
40
|
Salem ML, Gillanders WE, Kadima AN, El-Naggar S, Rubinstein MP, Demcheva M, Vournakis JN, Cole DJ. Review: novel nonviral delivery approaches for interleukin-12 protein and gene systems: curbing toxicity and enhancing adjuvant activity. J Interferon Cytokine Res 2006; 26:593-608. [PMID: 16978064 DOI: 10.1089/jir.2006.26.593] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
It has become increasingly apparent that the ability to generate an optimal host immune response requires effective cross talk between the innate and adaptive components of the immune system. Pro-inflammatory cytokines, in particular those that can induce a danger signal, often called signal 3, are crucial in this role of initiating and augmenting the presentation of exogenous antigen to T cells by dendritic cells. Interleukin-12 (IL-12) in particular has been defined as a "signal 3" cytokine required for the antigen cross priming. Given this unique interactive function, a significant amount of work has been performed to define possible therapeutic applications for IL-12. Systemic IL-12 administration can clearly act as a potent adjuvant for postvaccination T cell responses in a variety of diseases. As an example, in the cancer setting, systemic IL-12 is capable of suppressing tumor growth, metastasis, and angiogenesis in vivo. IL-12, however, has been associated with significant dose- and schedule-dependent toxicity in early clinical trials, results that have proven to be a major obstacle to its clinical application. Recent research has focused on decreasing the toxicity of IL-12 using different delivery approaches, including virus-based and gene-modified cell-based delivery. Although effective, these approaches also have limitations, including the generation of neutralizing antibodies, in addition to lacking the simplicity and versatility required for universal clinical application. Thus, there is a significant interest in the development of alternative delivery approaches for IL-12 administration that can overcome these issues. Several nonviral delivery approaches for IL-12 protein or gene expression vectors are being defined, including alum, liposomes, and polymer-based delivery. These developing approaches have shown promising adjuvant effects with significantly lessened systemic toxicity. This article discusses the potential capabilities of these nonvirus-based IL-12 delivery systems in different disease settings, including allergy, infection, and cancer.
Collapse
Affiliation(s)
- Mohamed Labib Salem
- Department of Surgery, Section of Surgical Oncology, Medical University of South Carolina, Charleston, SC 29425, USA.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
|
42
|
Torrero MN, Xia X, Henk W, Yu S, Li S. Stat1 deficiency in the host enhances interleukin-12-mediated tumor regression. Cancer Res 2006; 66:4461-7. [PMID: 16618773 DOI: 10.1158/0008-5472.can-05-3554] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Signal transducer and activator of transcription 1 (Stat1) is considered a key transcription factor that inhibits tumorigenesis, and Stat1 activation in the host is required for interleukin-12 (IL-12)-mediated generation of CTL activity. Using syngeneic Stat1-/- C3H mice bearing SCCVII tumors in this study, we discovered opposite results. Stat1 deficiency in the host significantly enhances IL-12-mediated tumor regression, resulting in tumor eradication from 60% of SCCVII tumor-bearing mice and significant inhibition of tumor growth when compared with control treatment (P < 0.01). This effect is independent of both Stat1-activating cytokine IFN-gamma and Stat1-downstream effector molecule FasL because neither neutralization of IFN-gamma nor knocking out of FasL enhances or inhibits IL-12-mediated tumor regression. IL-12 induces a high intensity of tumor-specific CTL activity in Stat1-deficient mice (P < 0.01), increases the CD8 T-cell density in tumor bearing Stat1-/- mice, and induces a T-cell-dependent tumor regression. The increased CTL activity and the high-intensity infiltration of T cells into the tumors in IL-12-treated Stat1-/- mice are likely due to the longer survival than the same cells from wild-type mice. Together, the data show that inhibition of Stat1 expression in the host enhances tumor-local IL-12 gene therapy for regressing tumors. This conclusion provides a new concept for designing an effective treatment strategy.
Collapse
Affiliation(s)
- Marina N Torrero
- Department of Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, Louisiana 70803, USA
| | | | | | | | | |
Collapse
|
43
|
Li S, Zhang L, Torrero M, Cannon M, Barret R. Administration route- and immune cell activation-dependent tumor eradication by IL12 electrotransfer. Mol Ther 2005; 12:942-9. [PMID: 15953768 DOI: 10.1016/j.ymthe.2005.03.037] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2005] [Revised: 03/03/2005] [Accepted: 03/15/2005] [Indexed: 11/20/2022] Open
Abstract
Injection of DNA via electric pulses into targeted tissues is referred to as electrotransfer. Intratumoral electrotransfer of the IL12 gene is more effective than intramuscular electrotransfer of the same gene in the eradication of established tumors. To understand the underlying immunological mechanisms, T cell infiltration, CTL activity, inhibition of angiogenesis, and transgene expression were analyzed using immunohistochemistry, fluorescence-based CTL analysis, Northern blot, and ELISA. In addition, the therapeutic effects of IL12 gene therapy were determined in immunocompetent, immune-cell-depleted, and immunodeficient mice. We found that intratumoral, but not intramuscular, electrotransfer of the IL12 gene induces CD8+ T cell infiltration, CTL activity, and tumor eradication. Tumor eradication by intratumoral IL12 gene electrotransfer requires both NK and T cells. The absence of either cell type will abrogate the intratumoral IL12 gene therapy-induced tumor eradication. Such a requirement explains why tumors cannot be eradicated by intramuscular electrotransfer of the IL12 gene. Only NK-cell-dependent, and not T-cell-dependent, anti-tumor effects are induced by intramuscular administration. Together, these results suggest that NK cells play an important role in both administration routes, mediating tumor growth inhibition, but T cells are specifically activated by intratumoral IL12 gene electrotransfer and not by the intramuscular route.
Collapse
Affiliation(s)
- Shulin Li
- Department of Comparative Biomedical Sciences, Louisiana State University, Skip Bertman Drive, Baton Rouge, LA 70803, USA.
| | | | | | | | | |
Collapse
|
44
|
McKechnie A, Robins RA, Eremin O. Immunological aspects of head and neck cancer: biology, pathophysiology and therapeutic mechanisms. Surgeon 2005; 2:187-207. [PMID: 15570827 DOI: 10.1016/s1479-666x(04)80001-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Advanced cancer and head and neck cancer, in particular, remains a major clinical challenge with its associated morbidity and inevitable mortality. Local control of early disease is achievable in many solid tumours with current surgical and radiotherapeutic techniques but metastatic disease is associated with poor outcome and prognosis. It is known that, by the time of presentation, many patients will already have occult microscopic metastatic disease, and surgery and radiotherapy will not result in long-term survival. What little effect modern chemotherapeutic agents have on microscopic disease is, however, limited by systemic toxicity and multi-drug resistance. Immune surveillance is postulated to be operative in man. There is evidence, however, that patients with progressive tumour growth have failure of host defences both locally and systemically. Various possible defects and tumour escape mechanisms are discussed in the review. Immunotherapy and, in particular adoptive T cell therapy and DC therapy, show promise as putative tumour-specific therapy with clinical benefits. These techniques are undergoing development and evaluation in phase 1 clinical trials. Preliminary data suggest that the treatments are well tolerated. Unfortunately, there is limited evidence of significant and prolonged improvements in clinical outcome. Further developments of beneficial protocols (adjuvants, mode and frequency of vaccination etc) and multicentre studies of the use of immunotherapy in cancer are now required.
Collapse
Affiliation(s)
- A McKechnie
- Department of Surgery, University of Nottingham, Nottingham, UK
| | | | | |
Collapse
|
45
|
Li S, Wilkinson M, Xia X, David M, Xu L, Purkel-Sutton A, Bhardwaj A. Induction of IFN-regulated factors and antitumoral surveillance by transfected placebo plasmid DNA. Mol Ther 2005; 11:112-9. [PMID: 15585412 DOI: 10.1016/j.ymthe.2004.09.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2004] [Revised: 08/27/2004] [Accepted: 09/10/2004] [Indexed: 11/24/2022] Open
Abstract
Delivery of DNA encoding therapeutic genes in vivo has great potential for treating malignancy as well as genetic diseases. Delivery of placebo DNA without a transgene is used as a control in gene therapy studies. It is tacitly assumed by most investigators that the protein expressed from the transfected DNA has phenotypic consequences, but that the consequences are not from the DNA itself. Here, we demonstrate that transfection of control plasmid DNA (that does not express a gene product) into tumor cell lines induces a dramatic (>10-fold) increase in the expression of the interferon (IFN)-regulated genes IRF7, STAT1, MIG (approved gene symbol CXCL9), MHCI (MICA), and CD11a (ITGAL) in tumor cell lines. Induction of these genes inhibits tumor development and tumor growth in immunocompetent mice that are immunized with apoptotic tumor cells. The antibody depletion study indicates that the underlying mechanism by which transfection of control DNA induces IFN-regulated genes is the induction of a secreting factor(s) such as IFN-beta. Three lines of evidence indicate that DNA transfection-mediated induction of IFN-regulatory genes is independent of TLR9. The three lines of evidence are: (1) TLR9 is not expressed in either SCCVII or 4T1 cell line, (2) activation of TLR9 downstream signaling molecules is not associated with the induction of gene expression, and (3) the secretion factor(s) obtained from the conditioned medium of DNA-transfected SCCVII tumor cells induces the same type of gene expression in the 4T1 tumor cell line, which is refractory to the gene induction by DNA transfection. Our finding indicates that the 4T1 tumor cell line, which is resistant to the DNA transfection-mediated induction of IFN-regulated genes, can be used to determine the real therapeutic gene function.
Collapse
Affiliation(s)
- Shulin Li
- Department of CBS, SVM, Louisiana State University, Skip Bertman Drive, Baton Rouge, LA 70803, USA.
| | | | | | | | | | | | | |
Collapse
|
46
|
Mir LM, Moller PH, André F, Gehl J. Electric pulse-mediated gene delivery to various animal tissues. ADVANCES IN GENETICS 2005; 54:83-114. [PMID: 16096009 DOI: 10.1016/s0065-2660(05)54005-7] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Electroporation designates the use of electric pulses to transiently permeabilize the cell membrane. It has been shown that DNA can be transferred to cells through a combined effect of electric pulses causing (1) permeabilization of the cell membrane and (2) an electrophoretic effect on DNA, leading the polyanionic molecule to move toward or across the destabilized membrane. This process is now referred to as DNA electrotransfer or electro gene transfer (EGT). Several studies have shown that EGT can be highly efficient, with low variability both in vitro and in vivo. Furthermore, the area transfected is restricted by the placement of the electrodes, and is thus highly controllable. This has led to an increasing use of the technology to transfer reporter or therapeutic genes to various tissues, as evidenced from the large amount of data accumulated on this new approach for non-viral gene therapy, termed electrogenetherapy (EGT as well). By transfecting cells with a long lifetime, such as muscle fibers, a very long-term expression of genes can be obtained. A great variety of tissues have been transfected successfully, from muscle as the most extensively used, to both soft (e.g., spleen) and hard tissue (e.g., cartilage). It has been shown that therapeutic levels of systemically circulating proteins can be obtained, opening possibilities for using EGT therapeutically. This chapter describes the various aspects of in vivo gene delivery by means of electric pulses, from important issues in methodology to updated results concerning the electrotransfer of reporter and therapeutic genes to different tissues.
Collapse
Affiliation(s)
- Lluis M Mir
- Laboratory of Vectorology and Gene Transfer, UMR 8121 CNRS Institut Gustave-Roussy, F-94805 Villejuif Cédex, France
| | | | | | | |
Collapse
|
47
|
Nakano H, Kishida T, Asada H, Shin-Ya M, Shinomiya T, Imanishi J, Shimada T, Nakai S, Takeuchi M, Hisa Y, Mazda O. Interleukin-21 triggers both cellular and humoral immune responses leading to therapeutic antitumor effects against head and neck squamous cell carcinoma. J Gene Med 2005; 8:90-9. [PMID: 16097036 DOI: 10.1002/jgm.817] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Interleukin-21 (IL-21) plays important roles in the regulation of T, B, and natural killer (NK) cells. We hypothesized that the cytokine may provide a novel immunotherapy strategy for cancer by stimulating both Th1 and Th2 immune responses. In this context, antitumor immunity induced by IL-21 was examined in mice bearing subcutaneous head and neck squamous cell carcinomas (HNSCC). METHODS A plasmid vector encoding murine IL-21 was injected intravenously into mice with pre-established HNSCC tumors, either alone or in combination with a vector construct expressing IL-15. Cytotoxic T lymphocyte (CTL) and NK killing activities were evaluated by chrome release assays, while HNSCC-specific antibody was examined by flow cytometry and ELISA. RESULTS Significant antitumor effects were obtained by repeated transfection with either the IL-21 or the IL-15 gene. Co-administration of both cytokine genes resulted in increased suppression of tumor growth, significantly prolonging the survival periods of the animals. Thirty percent of the tumor-bearing mice that received the combination therapy survived for more than 300 days, completely rejecting rechallenge with the tumor at a distant site. IL-21 induced significant elevation of HNSCC-specific CTL activity, while IL-21 and IL-15 augmented NK activity in an additive manner. IL-21 gene transfer also promoted the production of tumor-specific IgG. CONCLUSIONS In vivo transduction of the IL-21 gene elicits powerful antitumor immunity, including both humoral and cellular arms of the immune response, and results in significant suppression of pre-established HNSCC. Co-transfer of the IL-15 gene further improved the therapeutic outcome, mainly by augmenting NK tumoricidal activity. The biological effects of IL-21 may be in sharp contrast to those of conventional Th1 and Th2 cytokines, suggesting intriguing implications of this cytokine for the classical concept of Th1 vs. Th2 paradigm.
Collapse
Affiliation(s)
- Hiroshi Nakano
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
André F, Mir LM. DNA electrotransfer: its principles and an updated review of its therapeutic applications. Gene Ther 2004; 11 Suppl 1:S33-42. [PMID: 15454955 DOI: 10.1038/sj.gt.3302367] [Citation(s) in RCA: 181] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The use of electric pulses to transfect all types of cells is well known and regularly used in vitro for bacteria and eukaryotic cells transformation. Electric pulses can also be delivered in vivo either transcutaneously or with electrodes in direct contact with the tissues. After injection of naked DNA in a tissue, appropriate local electric pulses can result in a very high expression of the transferred genes. This manuscript describes the evolution in the concepts and the various optimization steps that have led to the use of combinations of pulses that fit with the known roles of the electric pulses in DNA electrotransfer, namely cell electropermeabilization and DNA electrophoresis. A summary of the main applications published until now is also reported, restricted to the in vivo preclinical trials using therapeutic genes.
Collapse
Affiliation(s)
- F André
- Laboratory of Vectorology and Gene Transfer, UMR 8121 CNRS - Institut Gustave-Roussy, Villejuif Cedex, France
| | | |
Collapse
|
49
|
Goto T, Nishi T, Kobayashi O, Tamura T, Dev SB, Takeshima H, Kochi M, Kuratsu JI, Sakata T, Ushio Y. Combination electro-gene therapy using herpes virus thymidine kinase and interleukin-12 expression plasmids is highly efficient against murine carcinomas in vivo. Mol Ther 2004; 10:929-37. [PMID: 15509510 DOI: 10.1016/j.ymthe.2004.07.028] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2004] [Accepted: 07/27/2004] [Indexed: 10/26/2022] Open
Abstract
We report the use of plasmid DNA-mediated combination gene therapy for tumor-bearing mice using in vivo electroporation, also called electro-gene therapy (EGT), that resulted in uncomplicated and complete cures in more than 90% of the mice. Subcutaneously inoculated CT26 tumors in syngeneic BALB/c mice were subjected to repeated EGT treatments consisting of intratumoral co-injection of naked plasmids encoding the cytokine interleukin-12 (IL-12) (p35 and p40 subunits) and the suicide gene herpes simplex virus thymidine kinase (HSV-tk), followed by in vivo electroporation. The early anti-tumor effect was always stronger, and the rate of cure, as seen in the long-term follow-up, was always greater in the groups treated with combination EGT than in those treated with IL-12 or HSV-tk EGT alone. Systemic levels of IL-12 and IFN-gamma increased in both combination and IL-12-alone EGT-treated groups. Moreover, combination EGT for established subcutaneous tumors strongly reduced hematogenous lung metastases and increased survival time when live CT26 tumor cells were injected through the tail vein. Limited experiments on C57/B16 mice with murine melanoma also showed very similar trends. These results suggest that this simple and safe method of plasmid-mediated combination EGT may provide a potentially effective gene therapy for cancer.
Collapse
Affiliation(s)
- Tomoaki Goto
- Department of Neurosurgery, Saiseikai Kumamoto Hospital, 5-3-1 Chikami, Kumamoto 861-4193, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Nagai H, Horikawa T, Hara I, Fukunaga A, Oniki S, Oka M, Nishigori C, Ichihashi M. In vivo elimination of CD25+ regulatory T cells leads to tumor rejection of B16F10 melanoma, when combined with interleukin-12 gene transfer. Exp Dermatol 2004; 13:613-20. [PMID: 15447721 DOI: 10.1111/j.0906-6705.2004.00198.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
CD4(+)CD25(+) T cells are an important population that plays a crucial role in the maintenance of peripheral self-tolerance. Recently, it was shown that the elimination of these cells by in vivo administration of anti-CD25 monoclonal antibody (mAb) caused the regression of highly immunogenic tumors in syngeneic mice. In this study, we examined whether B16F10 melanoma cells regressed with the elimination of CD25(+) regulatory T cells. We found the melanoma cells were not affected at all by in vivo anti-CD25 mAb administration alone but tumor rejection resulted in all mice when the administration was combined with IL-12 gene transfer to tumor cells. In vivo, depletion of natural killer (NK) cells or CD8(+) T cells cancelled the tumor rejection. NK-cell depletion allowed IL-12-transfected B16F10 melanoma (B16/IL-12) to grow from an early stage and resulted in a more rapid tumor growth of B16/IL-12 than that in mice without administration of anti-CD25 mAb. On the other hand, CD8(+) T-cell depletion did not affect the tumor growth in the early phase but allowed B16/IL-12 to grow in rather a late phase and resulted in almost the same degree of tumor growth as in mice without administration of anti-CD25 mAb. In a previous study, we showed that the elimination of CD4(+) T cells enhanced the antitumor effect of B16/IL-12 and induced vitiligo-like coat color alteration. Therefore, we also examined the frequency of the change to a vitiligo-like coat color in mice showing tumor rejection caused by CD25(+) T-cell elimination to compare with the mechanism enhancing the antitumor effects by cell elimination. The elimination of CD25(+) T cells did not induce vitiligo-like coat color changes, though that of CD4(+) T cells induced the change in 60% of mice. Furthermore, we confirmed that elimination of CD25(+) T cells did not affect the T-helper (Th) 1/Th2 cytokine profile, while that of CD4(+)T cells abrogated the Th2 cytokines (IL-4 and IL-10) and resulted in a Th1-dominant cytokine profile in the tumor-draining lymph nodes (TDLNs) of B16/IL-12-bearing mice. These results indicate that in vivo depletion of CD25(+) regulatory T cells is a potent useful adjuvant in immunotherapy of B16F10 melanoma, when combined with IL-12 gene transfer and that the enhancement of the antitumor effect by CD25(+) T-cell depletion is mediated through CD8(+) T cells and may differ from the enhancing mechanism caused by CD4(+) T-cell depletion.
Collapse
Affiliation(s)
- Hiroshi Nagai
- Division of Dermatology, Department of Clinical Molecular Medicine Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan.
| | | | | | | | | | | | | | | |
Collapse
|