1
|
Mödl B, Moritsch S, Zwolanek D, Eferl R. Type I and II interferon signaling in colorectal cancer liver metastasis. Cytokine 2023; 161:156075. [PMID: 36323190 DOI: 10.1016/j.cyto.2022.156075] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 09/26/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022]
Abstract
Metastatic colorectal cancer is one of the leading causes of cancer-related deaths worldwide. Traditional chemotherapy extended the lifespan of cancer patients by only a few months, but targeted therapies and immunotherapy prolonged survival and led to long-term remissions in some cases. Type I and II interferons have direct pro-apoptotic and anti-proliferative effects on cancer cells and stimulate anti-cancer immunity. As a result, interferon production by cells in the tumor microenvironment is in the spotlight of immunotherapies as it affects the responses of anti-cancer immune cells. However, promoting effects of interferons on colorectal cancer metastasis have also been reported. Here we summarize our knowledge about pro- and anti-metastatic effects of type I and II interferons in colorectal cancer liver metastasis and discuss possible therapeutic implications.
Collapse
Affiliation(s)
- Bernadette Mödl
- Center for Cancer Research, Medical University of Vienna & Comprehensive Cancer Center, 1090 Vienna, Austria
| | - Stefan Moritsch
- Center for Cancer Research, Medical University of Vienna & Comprehensive Cancer Center, 1090 Vienna, Austria
| | - Daniela Zwolanek
- Center for Cancer Research, Medical University of Vienna & Comprehensive Cancer Center, 1090 Vienna, Austria
| | - Robert Eferl
- Center for Cancer Research, Medical University of Vienna & Comprehensive Cancer Center, 1090 Vienna, Austria.
| |
Collapse
|
2
|
Chen Y, Du Q, Zou Y, Guo Q, Huang J, Tao L, Shen X, Peng J. Co-delivery of doxorubicin and epacadostat via heparin coated pH-sensitive liposomes to suppress the lung metastasis of melanoma. Int J Pharm 2020; 584:119446. [PMID: 32439586 DOI: 10.1016/j.ijpharm.2020.119446] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/22/2020] [Accepted: 05/16/2020] [Indexed: 12/14/2022]
Abstract
High metastasis is responsible for the failure in the treatment of melanoma. Chemoimmunotherapy has shown conspicuous inhibition effects not only on the growth of tumor in situ, but also on the metastasis to distant organs. Given that the indoleamine-2,3 dioxygenase (IDO) overexpressed in the microenvironment of tumor leads to the immune escape, the combination of chemotherapeutic drug and IDO inhibitor might be a promising chemoimmunotherapy. Besides, the hematogenous metastasis mediated by platelets was supposed to be blocked by the heparin (HP). Therefore, a drug delivery system with all these elements involved might be a potential treatment for melanoma. Here, we developed a pH-sensitive liposomal dual-delivery system for doxorubicin (DOX) and epacadostat (EPA) with HP coated (HP/LDE). It was confirmed to enhance cytotoxicity and apoptosis, reverse the platelets-activated epithelial mesenchymal transformation (EMT) and prevent the invasion and migration in vitro. After systemic administration, HP/LDE provided the optimum anti-metastasis effect on the melanoma. The results of evaluation on DC maturation, CD8+ cytotoxic T lymphocytes (CTLs) activation and T cell mediated cytotoxicity were consistent in vitro and in vivo. Taken together, our study established a functional liposomal dual-delivery system with ideal anti-metastasis efficacy on melanoma.
Collapse
Affiliation(s)
- Yi Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, ChinaThe High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Qianming Du
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; Department of Clinical Pharmacy, School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ying Zou
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, ChinaThe High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Qianqian Guo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, ChinaThe High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Jing Huang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, ChinaThe High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Ling Tao
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, ChinaThe High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Xiangchun Shen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, ChinaThe High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China.
| | - Jianqing Peng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, ChinaThe High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China.
| |
Collapse
|
3
|
Synthetic Poly(L-Glutamic Acid)-conjugated CpG Exhibits Antitumor Efficacy With Increased Retention in Tumor and Draining Lymph Nodes After Intratumoral Injection in a Mouse Model of Melanoma. J Immunother 2018; 40:11-20. [PMID: 27681378 DOI: 10.1097/cji.0000000000000145] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
There is an urgent need for new clinically applicable drug-delivery methods to enhance accumulation of immune-activating drugs in tumors. We synthesized a poly(L-glutamic acid)-CpG ODN2216 conjugate (PG-CpG) and injected it intratumorally into C57BL/6 mice bearing subcutaneous B16-ovalbumin melanoma. PG-CpG elicited the same potent antitumoral activity as CpG with respect to reducing tumor growth and triggering antigen-specific CD8 T-cell responses in this well-established solid tumor model. Moreover, PG-CpG was retained significantly longer in both tumor and draining lymph nodes than was free CpG after intratumoral injection. Specifically, 48 hours after injection, 26.5%±16.9% of the injected PG-CpG dose versus 4.72%±2.61% of free CpG remained at the tumor, and 1.53%±1.22% of the injected PG-CpG versus 0.37%±0.33% of free CpG was retained in the draining inguinal lymph nodes. These findings indicate that PG is an effective synthetic polymeric carrier for delivery of immunostimulatory agents to tumors and lymph nodes.
Collapse
|
4
|
Ahmad Z, Kratzke RA. Novel oncolytic viral therapies in patients with thoracic malignancies. Oncolytic Virother 2016; 6:1-9. [PMID: 28053943 PMCID: PMC5189707 DOI: 10.2147/ov.s116012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Oncolytic virotherapy is the use of replication-competent viruses to treat malignancies. The potential of oncolytic virotherapy as an approach to cancer therapy is based on historical evidence that certain viral infections can cause spontaneous remission of both hematologic and solid tumor malignancies. Oncolytic virotherapy may eliminate cancer cells through either direct oncolysis of infected tumor cells or indirect immune-mediated oncolysis of uninfected tumor cells. Recent advances in oncolytic virotherapy include the development of a wide variety of genetically attenuated RNA viruses with precise cellular tropism and the identification of cell-surface receptors that facilitate viral transfer to the tissue of interest. Current research is also focused on targeting metastatic disease by sustaining the release of progeny viruses from infected tumor cells and understanding indirect tumor cell killing through immune-mediated mechanisms of virotherapy. The purpose of this review is to critically evaluate recent evidence on the clinical development of tissue-specific viruses capable of targeting tumor cells and eliciting secondary immune responses in lung cancers and mesothelioma.
Collapse
Affiliation(s)
- Zeeshan Ahmad
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Robert A Kratzke
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
5
|
Merry HE, Phelan P, Hwang B, Mulligan MS. Validating the use of short interfering RNA as a novel technique for cell-specific target gene knockdown in lung ischemia-reperfusion injury. J Thorac Cardiovasc Surg 2015; 151:499-506. [PMID: 26687890 DOI: 10.1016/j.jtcvs.2015.09.125] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 09/04/2015] [Accepted: 09/05/2015] [Indexed: 01/26/2023]
Abstract
OBJECTIVE Short interfering RNA is an effective method for target gene knockdown. However, concerns surround the design, administration, efficacy, specificity, and immunostimulatory potential. Although uptake by alveolar macrophages has been demonstrated, studies have not examined its use in lung ischemia-reperfusion injury. We describe the validation of short interference RNA as a novel technique for cell-specific target gene knockdown in our model of lung ischemia-reperfusion injury. METHODS Dose-response experiments were performed, and 3 distinct sequences of toll-like receptor-4, toll-like receptor-2, and myeloid differentiation factor-88 short interference RNA were tested for efficacy of knockdown. Saline, lipid vector, and noncoding short interference RNA controls were used. Similar experiments were performed in primary cultures of resident pulmonary cells. Target protein knockdown was assessed by Western blot. Rat serum and cell culture media were assessed for interferon and cytokine production. Biotin labeling was used to assess short interference RNA uptake. RESULTS Target protein expression was significantly reduced using short interference RNA. However, toll-like receptor-4 knockdown was isolated to alveolar macrophages, and biotin labeling confirmed toll-like receptor-4 short interference RNA localization to alveolar macrophages. There was significant knockdown of toll-like receptor-4 expression in cultured cells treated with toll-like receptor-4 short interference RNA. There was no significant change in interferon production after short interference RNA treatment. There was effective target protein knockdown with each sequence used. CONCLUSIONS Short interference RNA is a valid method for achieving target protein knockdown in alveolar macrophages and is an important tool in the evaluation of its role in the development of lung ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Heather E Merry
- Division of Thoracic Surgery, University of Washington, Seattle, Wash
| | - Patrick Phelan
- Division of Thoracic Surgery, University of Washington, Seattle, Wash
| | - Billanna Hwang
- Division of Thoracic Surgery, University of Washington, Seattle, Wash; University of Washington, Center for Lung Biology, Seattle, Wash.
| | - Michael S Mulligan
- Division of Thoracic Surgery, University of Washington, Seattle, Wash; University of Washington, Center for Lung Biology, Seattle, Wash
| |
Collapse
|
6
|
Wilson KD, Tam YK. Lipid-based delivery of CpG oligodeoxynucleotides for cancer immunotherapy. Expert Rev Clin Pharmacol 2014; 2:181-93. [DOI: 10.1586/17512433.2.2.181] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
7
|
Engel AL, Holt GE, Lu H. The pharmacokinetics of Toll-like receptor agonists and the impact on the immune system. Expert Rev Clin Pharmacol 2012; 4:275-89. [PMID: 21643519 DOI: 10.1586/ecp.11.5] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Toll-like receptor (TLR) ligation activates both the innate and adaptive immune systems, and plays an important role in antiviral and anti-tumor immunity. Therefore, a significant amount of effort has been devoted to exploit the therapeutic potential of TLR agonists. Depending on the therapeutic purpose, either as adjuvants to vaccine, chemotherapy or standalone therapy, TLR agonists have been administered via different routes. Both preclinical and clinical studies have suggested that the route of administration has significant effects on pharmacokinetics, and that understanding these effects is critical to the success of TLR agonist drug development. This article will summarize the pharmacokinetics of TLR agonists with different administration routes, with an emphasis on clinical studies of TLR ligands in oncologic applications.
Collapse
|
8
|
Gjetting T, Arildsen NS, Christensen CL, Poulsen TT, Roth JA, Handlos VN, Poulsen HS. In vitro and in vivo effects of polyethylene glycol (PEG)-modified lipid in DOTAP/cholesterol-mediated gene transfection. Int J Nanomedicine 2010; 5:371-83. [PMID: 20957159 PMCID: PMC2950395 DOI: 10.2147/ijn.s10462] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Indexed: 11/23/2022] Open
Abstract
Background: DOTAP/cholesterol-based lipoplexes are successfully used for delivery of plasmid DNA in vivo especially to the lungs, although low systemic stability and circulation have been reported. To achieve the aim of discovering the best method for systemic delivery of DNA to disseminated tumors we evaluated the potential of formulating DOTAP/cholesterol lipoplexes with a polyethylene glycol (PEG)-modified lipid, giving the benefit of the shielding and stabilizing properties of PEG in the bloodstream. Method: A direct comparison of properties in vitro and in vivo of 4 different DOTAP/cholesterol-based lipoplexes containing 0%, 2%, 4%, and 10% PEG was performed using reporter gene activity and radioactive tracer lipid markers to monitor biodistribution. Results: We found that 10% PEGylation of lipoplexes caused reduced retention in lung and heart tissues of nude mice compared to nonPEGylated lipoplexes, however no significant delivery to xenograft flank tumors was observed. Although PEGylated and nonPEGylated lipoplexes were delivered to cells the ability to mediate successful transfection is hampered upon PEGylation, presumably due to a changed uptake mechanism and intracellular processing. Conclusion: The eminent in vivo transfection potency of DOTAP/cholesterol-based lipoplexes is well established for expression in lung tumors, but it is unsuitable for expression in non first pass organs such as xenograft flank tumors in mice even after addition of a PEG-lipid in the formulation.
Collapse
Affiliation(s)
- Torben Gjetting
- Department of Radiation Biology, Finsen Center, University Hospital, Copenhagen, Denmark
| | | | | | | | | | | | | |
Collapse
|
9
|
Schaffert D, Kiss M, Rödl W, Shir A, Levitzki A, Ogris M, Wagner E. Poly(I:C)-mediated tumor growth suppression in EGF-receptor overexpressing tumors using EGF-polyethylene glycol-linear polyethylenimine as carrier. Pharm Res 2010; 28:731-41. [PMID: 20694527 DOI: 10.1007/s11095-010-0225-4] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Accepted: 07/19/2010] [Indexed: 11/28/2022]
Abstract
PURPOSE To develop a novel polyethylenimine (PEI)-based polymeric carrier for tumor-targeted delivery of cytotoxic double-stranded RNA polyinosinic:polycytidylic acid, poly(I:C). The novel carrier should be chemically less complex but at least as effective as a previously developed tetra-conjugate containing epidermal growth factor (EGF) as targeting ligand, polyethylene glycol (PEG) as shielding spacer, 25 kDa branched PEI as RNA binding and endosomal buffering agent, and melittin as endosomal escape agent. METHODS Novel conjugates were designed employing a simplified synthetic strategy based on 22 kDa linear polyethylenimine (LPEI), PEG spacers, and recombinant EGF. The efficacy of various conjugates (different PEG spacers, with and without targeting EGF) in poly(I:C)-mediated cell killing was evaluated in vitro using two human U87MG glioma cell lines. The most effective polyplex was tested for in vivo activity in A431 tumor xenografts. RESULTS Targeting conjugate LPEI-PEG2 kDa-EGF was found as most effective in poly(I:C)-triggered killing of tumor cells in vitro. The efficacy correlated with glioma cell EGFR levels. Repeated intravenous administration of poly(I:C) polypexes strongly retarded growth of A431 human tumor xenograft in mice. CONCLUSIONS The optimized LPEI-PEG2 kDa-EGF conjugate displays reduced chemical complexity and efficient poly(I:C)-mediated killing of EGFR overexpressing tumors in vitro and in vivo.
Collapse
Affiliation(s)
- David Schaffert
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität Munich, Butenandtstr. 5-13, 81377, Munich, Germany.
| | | | | | | | | | | | | |
Collapse
|
10
|
Oncolytic measles viruses encoding interferon beta and the thyroidal sodium iodide symporter gene for mesothelioma virotherapy. Cancer Gene Ther 2010; 17:550-8. [PMID: 20379224 PMCID: PMC2907639 DOI: 10.1038/cgt.2010.10] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mesothelioma usually leads to death within 8–14 months of diagnosis. To increase the potency of oncolytic measles viruses (MVs) for mesothelioma therapy, we inserted the interferon β (IFNβ) gene alone or with the human thyroidal sodium iodide symporter (NIS) gene into attenuated MV of the Edmonston lineage. The corresponding mouse IFNβ (mIFNβ) viruses, MV-mIFNβ and MV-mIFNβ-NIS, successfully propagated in human mesothelioma cells, leading to intercellular fusion and cell death. High levels of mIFNβ were detected in the supernatants of the infected cells, and radioiodine uptake was substantial in the cells infected with MV-mIFNβ-NIS. MV with mIFNβ expression triggered CD68-positive immune cell infiltration 2–4 times higher than MV-GFP injected into the tumor site. The numbers of CD31-positive vascular endothelial cells within the tumor were decreased at day 7 after intratumoral injection of MV-mIFNβ or MV-mIFNβ-NIS, but not after MV-GFP and PBS administration. Immunohistochemical analysis showed that MV-mIFNβ changed the microenvironment of the mesothelioma by increasing innate immune cell infiltration and inhibiting tumor angiogenesis. Oncolytic MVs coding for IFNβ effectively retarded growth of human mesotheliomas and prolonged survival time in several mesothelioma tumor models. The results suggest that oncolytic MVs that code for IFNβ and NIS will be potent and versatile agents for the treatment of human mesothelioma.
Collapse
|
11
|
Sun X, Zhang HW, Zhang ZR. Growth inhibition of the pulmonary metastatic tumors by systemic delivery of the p27 kip1 gene using lyophilized lipid-polycation-DNA complexes. J Gene Med 2009; 11:535-44. [PMID: 19326369 DOI: 10.1002/jgm.1322] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND p27(kip1) (p27), a cyclin-dependent kinase inhibitor (CDKI), is an important regulator of cell cycle progression and a putative tumor suppressor gene, and plays an important role in the inhibition of genesis and progression of several kinds of cancers. The present study aimed to evaluate the anti-tumor effects of p27 gene therapy by a nonviral gene delivery strategy on pulmonary metastatic tumors. METHODS A recombinant plasmid composed of a p27 sequence was constructed and identified; it was then formulated with condensing agent protamine sulfate and entrapped into cationic liposomes. The resulting lipid-polycation-DNA complexes (LPD) were prepared into lyophilized forms. 5 x 10(5) of CT26 colorectal adenocarcinoma cells were inoculated into female Balb/c mice via the tail vein to establish lung tumor models. On the second day, mice were randomly divided into six groups for different intravenous treatments: phosphate-buffered saline, empty liposomes, naked pDNA, LPD-p27 kip1, Cisplatin (DPP), and LPD-p27 kip1 plus DPP, respectively. RESULTS The growth curve of tumor and the growth inhibition rate of tumor showed that p27-LPDs could prolong the lifespan of the mice significantly, whereas the combination of p27-LPDs and DPP could further prolong the lifespan of the tumor-bearing animals. The histology of tumors examined by hematoxylin and eosin staining indicated that p27-LPDs had a stronger inhibition effect. Significant expression of p27 was detected in tumors using an immunohistochemical technique. CONCLUSIONS Lyophilized LPD could be used as a potential in vivo gene delivery carrier for lung cancer gene therapy.
Collapse
Affiliation(s)
- Xun Sun
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Sichuan, China
| | | | | |
Collapse
|
12
|
Lipid-based systemic delivery of siRNA. Adv Drug Deliv Rev 2009; 61:721-31. [PMID: 19328215 DOI: 10.1016/j.addr.2009.03.003] [Citation(s) in RCA: 352] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2009] [Accepted: 03/10/2009] [Indexed: 01/13/2023]
Abstract
RNAi technology has brought a new category of treatments for various diseases including genetic diseases, viral diseases, and cancer. Despite the great versatility of RNAi that can down regulate almost any protein in the cells, the delicate and precise machinery used for silencing is the same. The major challenge indeed for RNAi-based therapy is the delivery system. In this review, we start with the uniqueness and mechanism of RNAi machinery and the utility of RNAi in therapeutics. Then we discuss the challenges in systemic siRNA delivery by dividing them into two categories-kinetic and physical barriers. At the end, we discuss different strategies to overcome these barriers, especially focusing on the step of endosome escape. Toxicity issues and current successful examples for lipid-based delivery are also included in the review.
Collapse
|
13
|
Yang R, Yang SG, Shim WS, Cui F, Cheng G, Kim IW, Kim DD, Chung SJ, Shim CK. Lung-specific delivery of paclitaxel by chitosan-modified PLGA nanoparticles via transient formation of microaggregates. J Pharm Sci 2009; 98:970-84. [PMID: 18661542 DOI: 10.1002/jps.21487] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Chitosan-modified paclitaxel-loaded poly lactic-co-glycolic acid (PLGA) nanoparticles with a mean diameter of 200-300 nm in distilled water were prepared by a solvent evaporation method. The mean diameter increased dramatically in contact with the mouse (CDF(1)) plasma, as a function of chitosan concentration in the modification solution (e.g., 2670.5 nm for 0.7% chitosan-modified nanoparticles, NP(3)), but reverted to almost its original size (i.e., 350.7 nm for NP(3)) following 5 min of gentle agitation. The zeta potential of PLGA nanoparticles was changed to positive by the chitosan modification. The in vitro uptake into, and cytotoxicity of the nanoparticles against, a lung cancer cell line (A549) were significantly increased by the modification. Most importantly, a lung-specific increase in the distribution index of paclitaxel (i.e., AUC(lung)/AUC(plasma)) was observed for chitosan-modified nanoparticles (e.g., 99.9 for NP(3) vs. 5.4 for Taxol) when nanoparticles were administered to lung-metastasized mice via the tail vein at a paclitaxel dose of 10 mg/kg. Transient formation of aggregates in the blood stream followed by enhanced trapping in the lung capillaries, and electrical interaction-mediated enhanced uptake across the endothelial cells of the lung tumor capillary appear to be responsible for the lung-tumor-specific distribution of the chitosan modified nanoparticles.
Collapse
Affiliation(s)
- Rui Yang
- National Research Laboratory for Transporters Targeted Drug Design, College of Pharmacy, Seoul National University, Seoul 151-742, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Wilson KD, de Jong SD, Tam YK. Lipid-based delivery of CpG oligonucleotides enhances immunotherapeutic efficacy. Adv Drug Deliv Rev 2009; 61:233-42. [PMID: 19232375 DOI: 10.1016/j.addr.2008.12.014] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2008] [Accepted: 12/15/2008] [Indexed: 11/26/2022]
Abstract
There has been significant interest in the potential of cytosine-guanine (CpG) containing oligodeoxynucleotides (ODN) as an immunotherapy for malignant, infectious and allergic diseases. While human trials have yielded promising results, clinical use of free CpG ODN still faces several challenges which limit their effectiveness. These include suboptimal in vivo stability, toxicity, unfavorable pharmacokinetic/biodistribution characteristics, lack of specificity for target cells and the requirement for intracellular uptake. To overcome these challenges, optimized lipid-based delivery systems have been developed to protect the CpG ODN payload, modify their circulation/distribution so as to enhance immune cell targeting and facilitate intracellular uptake. Ultimately, lipid-mediated delivery has the capacity to increase the immunopotency of CpG ODN and enhance their prophylactic or therapeutic efficacy in a range of diseases. Lipid-encapsulation provides a feasible strategy to optimize the immunostimulatory activity and immunotherapeutic efficacy of CpG ODN, thereby allowing their full clinical potential to be realized.
Collapse
|
15
|
Saito Y, Higuchi Y, Kawakami S, Yamashita F, Hashida M. Immunostimulatory Characteristics Induced by Linear Polyethyleneimine–Plasmid DNA Complexes in Cultured Macrophages. Hum Gene Ther 2009; 20:137-45. [DOI: 10.1089/hum.2008.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Yasunori Saito
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yuriko Higuchi
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shigeru Kawakami
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Fumiyoshi Yamashita
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Mitsuru Hashida
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- Institute for Integrated Cell-Material Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
16
|
Enhanced electrostatic interaction between chitosan-modified PLGA nanoparticle and tumor. Int J Pharm 2008; 371:142-7. [PMID: 19118614 DOI: 10.1016/j.ijpharm.2008.12.007] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2008] [Revised: 11/10/2008] [Accepted: 12/04/2008] [Indexed: 11/21/2022]
Abstract
In our previous study, lung tumor-specific targeting of paclitaxel was achieved in mice by intravenous administration of chitosan-modified paclitaxel-loaded PLGA nanoparticles (C-NPs-paclitaxel). Transient formation of aggregates in the blood stream followed by enhanced trapping in the capillaries was proposed as a mechanism of the lung-specific accumulation of paclitaxel. In the present study, the mechanism of tumor lung preferential accumulation of paclitaxel from C-NPs-paclitaxel was investigated. Zeta potential and in vitro cellular cytotoxicity (A549 cells and CT-26 cells) of C-NPs-paclitaxel, and in vitro uptake of coumarin 6 to these cells from chitosan-modified coumarin 6 containing PLGA nanoparticles (C-NPs-coumarin 6) were examined as a function of pH (6.8, 7.4 and 8.0). The zeta potential of C-NPs-paclitaxel increased as the medium pH became more acidic. In vitro uptake of coumarin 6 by A549 cells and CT-26 cells was enhanced at lower pH for C-NPs-coumarin 6. In vitro cytotoxicity experiment with C-NPs-paclitaxel demonstrated enhanced cytotoxicity as the pH became more acidic. Therefore, enhanced electrostatic interaction between chitosan-modified PLGA nanoparticles and acidic microenvironment of tumor cells appears to be an underlying mechanism of lung tumor-specific accumulation of paclitaxel from C-NPs-paclitaxel.
Collapse
|
17
|
Abstract
Cationic liposome-nucleic acid complexes, which were originally developed for use as non-viral gene delivery vectors, may now have an equally important application as immunotherapeutic drugs. Recent studies have highlighted the ability of cationic liposomes to potently activate the innate immune system when used to deliver certain Toll-like receptor (TLR) agonists. The immune-enhancing properties of cationic liposomes have been most clearly demonstrated when combined with nucleic acid agonists for endosomally located TLRs, including TLR3, TLR7/8 and TLR9. Immune potentiation by cationic liposomes likely results from the combined effects of endosomal targeting, protection of nucleic acids from extracellular degradation, and from signaling via newly identified cytoplasmic receptors for nucleic acids. The potent innate immune stimulatory properties of liposome-nucleic acid complexes make them particularly attractive as non-specific immunotherapeutics and as vaccine adjuvants. Liposome-nucleic acid complexes have demonstrated impressive anticancer activity in a number of different animal tumor models. Moreover, liposome-nucleic acid complexes have also been shown to be effective for immunotherapy of acute viral and bacterial infections, as well as chronic fungal infections. When used as vaccine adjuvants, liposome-nucleic acid complexes target antigens for efficient uptake by dendritic cells and are particularly effective in eliciting CD8(+) T-cell responses to protein antigens. Thus, liposome-nucleic acid complexes form a potent and versatile immunotherapeutic platform.
Collapse
Affiliation(s)
- Steven Dow
- Department of Microbiology, Colorado State University, Ft. Collins, CO 80523, USA.
| |
Collapse
|
18
|
Pope-Harman A, Cheng MMC, Robertson F, Sakamoto J, Ferrari M. Biomedical nanotechnology for cancer. Med Clin North Am 2007; 91:899-927. [PMID: 17826110 DOI: 10.1016/j.mcna.2007.05.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Nanotechnology may hold the key to controlling many devastating diseases. In the fight against the pain, suffering, and death due to cancer, nanotechnology will allow earlier diagnosis and even prevention of malignancy at premalignant stages, in addition to providing multimodality treatment not possible with current conventional techniques. This review discusses nanotechnology already used in diagnostic and therapeutic applications for cancer. Also addressed are theoretic and evolving uses of nanotechnology, including multifunctional nanoparticles for imaging and therapy, nanochannel implants for controlled release of drugs, nanoscale devices for evaluation of proteomics and genomics, and diagnostic techniques that take advantage of physical changes in diseased tissue.
Collapse
Affiliation(s)
- Amy Pope-Harman
- Dorothy M. Davis Heart and Lung Research Institute, Department of Internal Medicine, The Ohio State University College of Medicine and Public Health, Columbus, OH 43210, USA.
| | | | | | | | | |
Collapse
|
19
|
Yonenaga Y, Mori A, Fujimoto A, Nagayama S, Tachibana T, Onodera H, Uemoto S. The administration of naked plasmid DNA into the liver induces antitumor innate immunity in a murine liver metastasis model. J Gene Med 2007; 9:299-307. [PMID: 17397091 DOI: 10.1002/jgm.1013] [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: 01/19/2023] Open
Abstract
BACKGROUND Gene therapy is a promising strategy against advanced cancer; however, the safety of viral vectors and the effectiveness of non-viral vectors have not yet been established. Recently, a hydrodynamics-based procedure was reported to be an effective and safe method to deliver and transduce DNA into the liver. Herein, we propose a strategy for liver metastasis by a hydrodynamics-based procedure to deliver naked non-coding plasmid DNA (pDNA) into the liver as an immunocompetent organ. METHODS AND RESULTS Mice received a rapid intravenous (i.v.) injection of naked pDNA in a large volume of saline (0.1 ml/g body weight). The single administration of a naked non-coding pDNA by the hydrodynamics-based procedure before tumor cell inoculation strongly suppressed liver metastasis formation. However, the usual i.v. injection (200 microl/body) of the same dose of naked pDNA could not suppress liver metastasis formation. Following the methylation of CpG sequences within the pDNA using CpG methylase, injection of the methylated pDNA by the hydrodynamics-based procedure could not suppress liver metastasis formation. Gadolinium chloride pretreatment did not interfere with this antitumor effect, but anti-asialo GM1 antiserum treatment did. These findings indicated that natural killer (NK) cells, not Kupffer cells, were involved in this antitumor effect. The NK cytotoxic activities of liver mononuclear cells were strongly enhanced after receiving a naked pDNA by the hydrodynamics-based procedure. CONCLUSIONS These observations suggest that unmethylated CpG motifs in pDNA stimulated immune cells, resulting in the activation of NK cells in the liver to suppress liver metastases in a murine model.
Collapse
Affiliation(s)
- Yoshikuni Yonenaga
- Department of Surgery, Graduate School of Medicine Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan.
| | | | | | | | | | | | | |
Collapse
|
20
|
Sakurai H, Kawabata K, Sakurai F, Nakagawa S, Mizuguchi H. Innate immune response induced by gene delivery vectors. Int J Pharm 2007; 354:9-15. [PMID: 17640834 DOI: 10.1016/j.ijpharm.2007.06.012] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2007] [Revised: 06/06/2007] [Accepted: 06/11/2007] [Indexed: 11/17/2022]
Abstract
Gene therapy is a clinical strategy that has the potential to treat an array of genetic and nongenetic diseases. Vectors for gene transfer are the essential tools of gene therapy. For gene therapy to be successful, an appropriate amount of the therapeutic gene must be delivered into the target cells without substantial toxicity. A major limitation of the use of gene therapy vectors is the innate immune responses triggered by systemic administration of such vectors. It is essential to overcome vector-mediated innate immune responses, such as production of inflammatory cytokines, the maturation of antigen-presenting cells and tissue damage, because the induction of these responses not only shortens the period of gene expression but also leads to serious side effects. Viral vectors (for example, adenovirus (Ad) vectors) have been assumed to be more potent in inducing innate immune responses in spite of their high transduction efficiency since they contain pathogenic proteins. However, recent studies have demonstrated that not only viral vectors but also nonviral vectors, such as lipoplex (liposome/plasmid DNA complex), can induce innate immune responses. Indeed, nonviral vectors including lipoplex induce comparable or larger levels of innate immune response than viral vectors. In this review, we present an overview of the innate immune responses induced by Ad vector and lipoplex, which are used primarily for in vivo gene transfer.
Collapse
Affiliation(s)
- Haruna Sakurai
- Laboratory of Gene Transfer and Regulation, National Institute of Biomedical Innovation, Osaka, Japan
| | | | | | | | | |
Collapse
|
21
|
Russ V, Wagner E. Cell and tissue targeting of nucleic acids for cancer gene therapy. Pharm Res 2007; 24:1047-57. [PMID: 17387604 DOI: 10.1007/s11095-006-9233-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2006] [Accepted: 12/26/2006] [Indexed: 12/11/2022]
Abstract
Tumor targeting--per definition--includes any strategy to improve the specificity of the therapeutic nucleic acid towards the tumor site, while highest biological activity should be maintained. Targeting has been successfully achieved at the transcriptional, transductional or delivery level. For tumor-specific delivery, physical targeting methods like electroporation, hyperthermia, magnetofection, photochemical internalization or ultrasound, and biological targeting systems, including active and passive tumor targeting, have been developed. Therapeutic effects could be demonstrated with various targeted nucleic acid formulations, such as tumor-targeted DNA plasmids expressing p53 or tumor necrosis factor alpha, small interfering RNAs knocking down gene expression from tumor specific chromosomal translocations or gene expression of tumor neoangiogenic processes, as well as double stranded RNA poly inosine-cytosine which triggers apoptosis in targeted tumor cells.
Collapse
Affiliation(s)
- Verena Russ
- Pharmaceutical Biology-Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universitaet, Munich, Germany
| | | |
Collapse
|
22
|
Meyer M, Wagner E. Recent developments in the application of plasmid DNA-based vectors and small interfering RNA therapeutics for cancer. Hum Gene Ther 2007; 17:1062-76. [PMID: 17032153 DOI: 10.1089/hum.2006.17.1062] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Increased understanding of the molecular pathological mechanisms of cancer, the advent of novel molecular tools such as synthetic small interfering RNA (siRNA) or plasmid DNA-based vectors (pDNA), and technology for the in vivo delivery of such biomolecular therapeutics have provided an encouraging perspective for cancer therapy. Numerous pDNAs and siRNAs have been tested in preclinical cancer models, and these first approaches have reached clinical evaluation. The therapeutic effector mechanisms include interference with neoangiogenesis, blockage of cell division, promotion of apoptosis and sensitization to chemotherapy, delivery of cytotoxic genes, and activation of anticancer immune responses. Physical methods have been developed for highly effective regional delivery. A series of innovative "smart" formulations directs the current development toward safe and effective systemic tumor-targeted delivery of pDNA and siRNA.
Collapse
Affiliation(s)
- Martin Meyer
- Department of Pharmacy, Center of Drug Research, Pharmaceutical Biology-Biotechnology, Ludwig Maximilian University, Munich 81377, Germany
| | | |
Collapse
|
23
|
Goding S, Yang Q, Mi Z, Robbins PD, Basse PH. Targeting of products of genes to tumor sites using adoptively transferred A-NK and T-LAK cells. Cancer Gene Ther 2007; 14:441-50. [PMID: 17273184 DOI: 10.1038/sj.cgt.7701019] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Despite successes in animals, cytokine gene expression selectively in human tumors is difficult to achieve owing to lack of efficient delivery methods. Since interleukin (IL)-2-activated natural killer (A-NK) and phytohemagglutinin and IL-2 activated killer T (T-LAK) cells, as previously demonstrated, localize and accumulate in murine lung tumor metastases following adoptive transfer, we transduced them to test their ability to deliver products of genes selectively to tumors. Assessments of transduction efficiency in vitro demonstrated that adenoviral transduction consistently resulted in high (>60%) transduction rates and substantial expression of transgenes such as GFP, Red2, luciferase, beta-galactosidase and mIL-12 for at least 4 days. In vivo experiments illustrated that Ad-GFP transduced A-NK and Ad-Red2 (RFP) transduced T-LAK or mIL-12 transduced A-NK cells localized 10-50-fold more or survived significantly better than mock transduced cells, respectively, within lung metastases than in the surrounding normal lung tissue. Most importantly, mIL-12 transduced A-NK cells provided a significantly greater antitumor response than non-transduced A-NK cells. Thus, adoptive transfer of A-NK and T-LAK cells represents an efficient method for targeting products of genes to tumor sites.
Collapse
Affiliation(s)
- S Goding
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | |
Collapse
|
24
|
Meyer M, Wagner E. Recent Developments in the Application of Plasmid DNA-Based Vectors and Small Interfering RNA Therapeutics for Cancer. Hum Gene Ther 2006. [DOI: 10.1089/hum.2006.17.ft-251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
25
|
Suzuki S, Kawakami S, Chansri N, Yamashita F, Hashida M. Inhibition of pulmonary metastasis in mice by all-trans retinoic acid incorporated in cationic liposomes. J Control Release 2006; 116:58-63. [PMID: 17067713 DOI: 10.1016/j.jconrel.2006.08.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2006] [Revised: 08/22/2006] [Accepted: 08/31/2006] [Indexed: 12/12/2022]
Abstract
The purpose of this study was to investigate whether all-trans retinoic acid (ATRA), an active metabolite of retinal, incorporated in cationic liposomes composed of 1,2 dioleoyl-3-trimethylammonium propane (DOTAP)/cholesterol could inhibit established metastatic lung tumors by delivery to the pulmonary tumor site after intravenous injection. After intravenous injection in mice, the highest lung accumulation of [(3)H]ATRA was observed by the DOTAP/cholesterol liposomes formulation, while other formulations including [(3)H]ATRA dissolved in serum or [(3)H]ATRA incorporated in distearoyl-l-phosphatidylcholine (DSPC)/cholesterol liposomes produced little accumulation in the lung. In mice used as a model of lung cancer metastasis, ATRA incorporated in DOTAP/cholesterol liposomes, injected intravenously, reduced the number of tumor nodules compared with free ATRA or ATRA incorporated in DSPC/cholesterol liposomes. These results suggest that ATRA incorporated in cationic liposomes would be an effective strategy for differentiation therapy of lung cancer metastasis.
Collapse
Affiliation(s)
- Sachiko Suzuki
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | | | | | |
Collapse
|
26
|
Kornbluth RS, Stone GW. Immunostimulatory combinations: designing the next generation of vaccine adjuvants. J Leukoc Biol 2006; 80:1084-102. [PMID: 16931603 DOI: 10.1189/jlb.0306147] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Agents that activate dendritic cells are essential components for vaccines and can be conceptualized as molecular adjuvants. Other molecular adjuvants affect downstream factors that shape the resulting immune response. This review provides a compendium of recently studied molecular adjuvants, focusing on CD8+ T cell responses, which have important roles in HIV vaccines. Reference is also made to CD8+ T cell antitumor responses, where parallel studies of molecular adjuvants are being pursued. Molecular adjuvants can be considered in the following groups: TNF superfamily molecules such as CD40 ligand; agonists for TLRs; agonists for NAIP, CIITA, HET-E, TP-1-leucine-rich repeat pathway receptors, such as nucleotide-binding and oligomerization domain (NOD)1, NOD2, and cryopyrin; chemokines; ILs; CSFs; IFNs; alarmins; and purinergic P2X7 receptor agonists. Complementing these positively acting agents are strategies to reduce the immunosuppressive effects of CD4+CD25+ regulatory T cells and negatively acting factors such as TGF-beta, IL-10, suppressor of cytokine signaling 1, and programmed cell death-1 using neutralizing antibodies, antisense, and small interfering RNA. Especially effective are combinations of molecular adjuvants, which can elicit a massive expansion of antigen-specific CD8+ T cells and show unprecedented efficacy in vaccine and tumor models. Taken together, these new approaches provide significant incremental progress in the development of vaccines to elicit cell-mediated immunity against HIV and other pathogens.
Collapse
Affiliation(s)
- Richard S Kornbluth
- Department of Medicine, University of California San Diego, 9500 Gilman Dr., #0679, La Jolla, CA 92093-0679, USA.
| | | |
Collapse
|
27
|
Wilderman MJ, Kim S, Gillespie CT, Sun J, Kapoor V, Vachani A, Sterman DH, Kaiser LR, Albelda SM. Blockade of TNF-alpha decreases both inflammation and efficacy of intrapulmonary Ad.IFNbeta immunotherapy in an orthotopic model of bronchogenic lung cancer. Mol Ther 2006; 13:910-7. [PMID: 16488193 DOI: 10.1016/j.ymthe.2005.12.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2005] [Revised: 12/27/2005] [Accepted: 12/27/2005] [Indexed: 01/22/2023] Open
Abstract
Adenoviral immuno-gene therapy using interferon-beta has been effective in an orthotopic model of lung cancer. However, pulmonary inflammation induced by adenoviral (Ad) vectors will almost certainly limit the maximally tolerated dose. On the other hand, the strong innate immune response generated by the vector may be helpful in initiating the adaptive immune response required for efficacy. The goals of this study were to develop an effective approach to inhibit Ad.IFNbeta-mediated acute pulmonary inflammation and to determine whether this reduction of Ad-mediated inflammation decreased the therapeutic efficacy of Ad.IFNbeta in a mouse model of bronchioloalveolar cancer. Our data show that anti-TNF-alpha antibodies can blunt the innate pulmonary immune response induced by Ad vectors, even in sensitized animals. However, this effect also inhibited the ability of the animal to generate anti-tumor immune responses and reduced survival in an orthotopic lung cancer model responsive to Ad.IFNbeta treatment. Interestingly, in a flank model of tumor using a cell line derived from the lung tumor, TNF-alpha blockade did not inhibit efficacy. These data suggest that the innate immune response to adenovirus in the lung may be important in immuno-gene therapy of lung cancer. Therapeutic application of anti-inflammatory therapy in immuno-gene therapy strategies should thus be undertaken with caution.
Collapse
Affiliation(s)
- Michael J Wilderman
- Thoracic Oncology Research Laboratory, University of Pennsylvania Medical Center, BRB II/III, 421 Curie Boulevard, Philadelphia, PA 19104-6160, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Kawakami S, Suzuki S, Yamashita F, Hashida M. Induction of apoptosis in A549 human lung cancer cells by all-trans retinoic acid incorporated in DOTAP/cholesterol liposomes. J Control Release 2006; 110:514-21. [PMID: 16360957 DOI: 10.1016/j.jconrel.2005.10.030] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2005] [Revised: 10/25/2005] [Accepted: 10/31/2005] [Indexed: 01/29/2023]
Abstract
All-trans retinoic acid (ATRA) has been shown to exert anti-cancer activities in a number of types of cancer cells. However, it has been reported that many NSCLC exhibited resistance to ATRA treatment. In the present study, we hypothesized that intracellular delivery of ATRA would overcome the ATRA resistance in A549 cells. Here, we investigated the induction of apoptosis by ATRA incorporated in cationic liposomes composed of DOTAP/cholesterol in A549 human lung cancer cells, which are insensitive (resistant) to the growth inhibitory effects of ATRA. The zeta potentials of DOTAP/cholesterol liposomes and DSPC/cholesterol liposomes were about +50 and -3 mV. In A549 cells, [(3)H]ATRA incorporated in DOTAP liposomes showed increased cellular association compared with [(3)H]ATRA or [(3)H]ATRA incorporated in DSPC/cholesterol liposomes. ATRA incorporated in DOTAP/cholesterol liposomes showed much higher cytotoxic effects and apoptosis-inducing activity compared with ATRA or ATRA incorporated in DSPC/cholesterol liposomes. The enhanced expression of TIG3 mRNA tumor suppressor gene by ATRA incorporation into DOTAP/cholesterol liposomes might partly explain the mechanism of enhanced cytotoxicity and/or apoptosis. These observations provide valuable information to help in the design of differentiation therapy by ATRA in non-small cell lung carcinoma.
Collapse
Affiliation(s)
- Shigeru Kawakami
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | | | | | | |
Collapse
|
29
|
Tam YK. Liposomal encapsulation enhances the activity of immunostimulatory oligonucleotides. ACTA ACUST UNITED AC 2006. [DOI: 10.2217/17460875.1.1.35] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
30
|
Wilderman MJ, Sun J, Jassar AS, Kapoor V, Khan M, Vachani A, Suzuki E, Kinniry PA, Sterman DH, Kaiser LR, Albelda SM. Intrapulmonary IFN-beta gene therapy using an adenoviral vector is highly effective in a murine orthotopic model of bronchogenic adenocarcinoma of the lung. Cancer Res 2005; 65:8379-87. [PMID: 16166316 DOI: 10.1158/0008-5472.can-05-0920] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Given previous work showing that an adenoviral vector expressing IFN-beta (Ad.IFNbeta) was highly effective in eradicating i.p. mesothelioma tumors, the antitumor efficacy of this agent was evaluated in an orthotopic model of bronchogenic adenocarcinoma of the lung. These transgenic mice have a conditionally expressed, oncogenic K-rasG12D allele that can be activated by intratracheal administration of an adenovirus expressing Cre recombinase (Ad.Cre). K-rasG12D mutant mice were given Ad.Cre intranasally to activate the oncogene. Mice were then given 10(9) plaque-forming units of a control vector (Ad.LacZ) or Ad.IFNbeta intranasally 3 and 4 weeks later, a time when lung tumors had been established. Cells derived from K-ras-mutated lung tumors were also grown in the flanks of mice to study mechanisms of therapeutic responses. In two separate experiments, untreated tumor-bearing mice all died by day 57 (median survival, 49 days). Ad.LacZ-treated mice all died by day 71 (median survival, 65 days). In contrast, 90% to 100% of mice treated with Ad.IFNbeta were long-term survivors (>120 days; P < 0.001). In addition, immunity to re-challenge with tumor cells was induced. In vitro and flank tumor studies showed that Ad.IFNbeta induced direct tumor cell killing and that depleting natural killer or CD8+ T cells, but not CD4+ T cells, with antibodies attenuated the effect of Ad.IFNbeta. These studies, showing remarkable antitumor activity in this orthotopic lung cancer model, provide strong preclinical support for a trial of Ad.IFNbeta to treat human non-small cell lung cancer.
Collapse
Affiliation(s)
- Michael J Wilderman
- Thoracic Oncology Research Laboratory, University of Pennsylvania Medical School, Philadelphia, Pennsylvania 19014-6160, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Dow S, Elmslie R, Kurzman I, MacEwen G, Pericle F, Liggitt D. Phase I study of liposome-DNA complexes encoding the interleukin-2 gene in dogs with osteosarcoma lung metastases. Hum Gene Ther 2005; 16:937-46. [PMID: 16076252 DOI: 10.1089/hum.2005.16.937] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Systemic gene delivery using cationic liposome-DNA complexes (LDCs) has been shown to elicit potent antitumor activity in mice with tumor metastases to the lungs. However, intravenous gene delivery for treatment of established cancer has not been evaluated previously in a spontaneous, large animal model. We therefore evaluated the safety, toxicity, and efficacy of intravenous gene delivery, using LDCs in dogs with established tumor metastases. Twenty dogs with chemotherapy-resistant osteosarcoma metastases to the lungs received a series of intravenous infusions of cationic liposomes and plasmid DNA encoding the canine interleukin-2 (IL-2) cDNA. Effects of intravenous gene delivery on immune activation, clinical and hematologic parameters, tumor responses, and survival times were assessed. We found that slow intravenous administration of IL-2 LDCs resulted in detectable IL-2 transgene expression in lung tissues of dogs. Repeated intravenous infusions of LDCs were well tolerated by dogs with lung tumor metastases and elicited systemic immune activation, as reflected by fever, leukogram changes, monocyte activation, and increased natural killer cell activity. Three of 20 dogs experienced partial or complete regression of lung metastases after infusion of IL-2 LDCs. Overall survival times were significantly increased in treated dogs compared with historical control animals with the same stage of disease. We conclude that repeated intravenous infusion of LDCs in cancerbearing dogs is safe and well tolerated at low doses and may be capable of eliciting antitumor activity in some animals with advanced tumor metastases.
Collapse
Affiliation(s)
- Steven Dow
- Department of Clinical Sciences, Colorado State University, Fort Collins, 80523, USA
| | | | | | | | | | | |
Collapse
|
32
|
Dow S, Elmslie R, Kurzman I, MacEwen G, Pericle F, Liggitt D. Phase I Study of Liposome-DNA Complexes Encoding the Interleukin-2 Gene in Dogs with Osteosarcoma Lung Metastases. Hum Gene Ther 2005. [DOI: 10.1089/hum.2005.16.ft-94] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
33
|
Salem ML. Systemic treatment with n-6 polyunsaturated fatty acids attenuates EL4 thymoma growth and metastasis through enhancing specific and non-specific anti-tumor cytolytic activities and production of TH1 cytokines. Int Immunopharmacol 2005; 5:947-60. [PMID: 15829411 DOI: 10.1016/j.intimp.2004.12.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2004] [Revised: 12/27/2004] [Accepted: 12/27/2004] [Indexed: 02/07/2023]
Abstract
Recently, there has been a great interest in the effects of different types of n-6 polyunsaturated acids (n-6 PUFAs) upon the immune system and cancer development. However, the effects of n-6 PUFAs are still controversial and as yet undefined. The present study aimed to investigate the anti-tumor effects of n-6 PUFAs against EL4 thymoma and the associated immune mechanisms. To this, sesame oil, a vegetable oil enriched with n-6 PUFAs, or free linoleic acid (LA) were administered intraperitoneally into C57BL/6 mice before and after challenge with EL4 lymphoma cells. Treatment with either sesame oil or LA attenuated the growth and metastasis of EL4 lymphoma. The anti-tumor effect of LA was superior to that of sesame oil, and associated with an increase in the survival rate of the tumor-bearing mice. In addition, both sesame oil and LA showed dose-dependent anti-lymphoma growth in vitro. Treatment with LA generated significant increases in the anti-lymphoma cytolytic and cytostatic activities of T cells and macrophages, respectively, and enhanced production of IL-2 and IFN-gamma while decreased production of IL-4, IL-6 and IL-10. In summation, the results suggest that n-6 PUFAs, represented by LA, can attenuate EL4 lymphoma growth and metastasis through enhancing the specific and non-specific anti-tumor cytolytic activities and production of TH1 cytokines. These findings might be of great importance for a proper design of systemic nourishment with PUFAs emulsions for cancer patients.
Collapse
Affiliation(s)
- Mohamed Labib Salem
- Department of Surgery, Section of Surgical Oncology, Holling Cancer Center R313, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
34
|
Sakakima Y, Hayashi S, Yagi Y, Hayakawa A, Tachibana K, Nakao A. Gene therapy for hepatocellular carcinoma using sonoporation enhanced by contrast agents. Cancer Gene Ther 2005; 12:884-9. [PMID: 15891773 DOI: 10.1038/sj.cgt.7700850] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We examined whether sonoporation enhanced by a contrast agent (BR14) was effective in gene therapy for hepatocelluar carcinoma (HCC). Human hepatic cancer cells (SK-Hep1) and plasmid cDNAs expressing green fluorescent protein (GFP), interferonbeta (IFNbeta), and LacZ were used. In vitro, SK-Hep1 cell suspensions with DNA and BR14 were sonoporated. Expressions of every plasmid cDNA and the antitumor effect of IFNbeta were analyzed. In vivo, GFP and IFNbeta genes with BR14 were directly injected into subcutaneous tumors using SK-Hep1 in nude mice, and transcutaneous sonoporation of the tumors was performed. GFP gene transfections and tumor diameters after IFNbeta gene transfection were examined. In vitro, no SK-Hep1 cells were transfected without sonication, whereas transfections were successful after sonication with BR14. Antitumor effect of IFNbeta gene transfection by ultrasound (US) and with BR14 was revealed. In vivo, the SK-Hep1 cells expressed GFP, and the IFNbeta gene transfection by US with BR14 reduced tumor size significantly. In conclusion, gene therapy with sonoporation enhanced by a contrast agent may become a new treatment option for HCC.
Collapse
Affiliation(s)
- Yoshikazu Sakakima
- Department of Surgery II, Nagoya University School of Medicine, Nagoya 466-8550, Japan.
| | | | | | | | | | | |
Collapse
|
35
|
Salem ML, Kadima AN, Cole DJ, Gillanders WE. Defining the antigen-specific T-cell response to vaccination and poly(I:C)/TLR3 signaling: evidence of enhanced primary and memory CD8 T-cell responses and antitumor immunity. J Immunother 2005; 28:220-8. [PMID: 15838378 DOI: 10.1097/01.cji.0000156828.75196.0d] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Poly(I:C), a synthetic double-stranded RNA polymer and a TLR3 agonist, can be used as a vaccine adjuvant to enhance adaptive immunity. However, the antigen-specific CD8 T-cell response to peptide vaccination and poly(I:C) has not been clearly defined. Here, the authors characterized the antigen-specific CD8 T-cell response to peptide vaccination and poly(I:C) and specifically addressed the hypothesis that poly(I:C) can enhance antitumor immunity. To define the antigen-specific T-cell response, the authors established a model based on the adoptive transfer of T cells from the OT-1 T-cell receptor transgenic mouse. In this model, vaccination with peptide alone resulted in a limited, transient expansion of antigen-specific CD8 T cells. In contrast, peptide vaccination with concomitant administration of poly(I:C) resulted in a dramatic sustained increase in the number of antigen-specific CD8 T cells. This increase in cell number was associated with an increase in CD8 T-cell function, as defined by specific IFN-gamma and TNF-alpha production, and protection from tumor challenge. The adjuvant effects of poly(I:C) appear to be at least partially dependent on an increase in the transcription of the anti-apoptotic molecules Bcl-3 and Bcl-xL and a concomitant decrease in apoptosis during the contraction phase of the primary T-cell response. In addition, administration of poly(I:C) enhanced the response to a nonimmunogenic self-antigen in a dendritic cell vaccine-based vaccine strategy. Collectively, these results confirm the potential of poly(I:C) as a vaccine adjuvant and suggest that targeting of TLR3 is likely to be a valuable addition to peptide-based vaccination strategies.
Collapse
Affiliation(s)
- Mohamed L Salem
- Department of Surgery, Section of Surgical Oncology, Medical University of South Carolina, Charleston, South Carolina 29425, USA.
| | | | | | | |
Collapse
|
36
|
Hashida M, Kawakami S, Yamashita F. Lipid Carrier Systems for Targeted Drug and Gene Delivery. Chem Pharm Bull (Tokyo) 2005; 53:871-80. [PMID: 16079512 DOI: 10.1248/cpb.53.871] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
For effective chemotherapy, it is necessary to deliver therapeutic agents selectively to their target sites, since most drugs are associated with both beneficial effects and side effects. The use of lipid dispersion carrier systems, such as lipid emulsions and liposomes, as carriers of lipophilic drugs has attracted particular interest. A drug delivery system can be defined as a methodology for manipulating drug distribution in the body. Since drug distribution depends on the carrier, administration route, particle size of the carrier, lipid composition of the carrier, electric charge of the carrier and ligand density of the targeting carrier, these factors must be optimized. Recently, the lipid carrier system has also been applied to gene delivery systems for gene therapy. However, in both drug and gene medicine cases, a lack of cell-selectivity limits the wide application of this kind of drug and/or gene therapy. Therefore, lipid carrier systems for targeted drug and gene delivery must be developed for the rational therapy. In this review, we shall focus on the progress of research into lipid carrier systems for drug and gene delivery following systemic or local injection.
Collapse
Affiliation(s)
- Mitsuru Hashida
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan.
| | | | | |
Collapse
|
37
|
Kawakami S, Ito Y, Fumoto S, Yamashita F, Hashida M. Enhanced gene expression in lung by a stabilized lipoplex using sodium chloride for complex formation. J Gene Med 2005; 7:1526-33. [PMID: 16170832 DOI: 10.1002/jgm.813] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In this study, we investigated the in vivo gene transfection efficacy of a 'surface charge regulated' (SCR) lipoplex, dispersed in the presence of an essential amount of NaCl during lipoplex formation. METHODS SCR lipoplexes were prepared and their physicochemical properties were analyzed. After intravenous (i.v.) administration, transfection efficacy, distribution characteristics, and liver toxicity were evaluated in mice. RESULTS At NaCl concentrations of 10 mM, the particle sizes of the SCR lipoplexes were about 120 nm and were compatible with a conventional lipoplex. However, fluorescent resonance energy transfer analysis revealed that cationic liposomes in the SCR lipoplexes increased fusion. After i.v. administration, the transfection activity in the lung of the SCR lipoplex (10 mM NaCl solution in the lipoplex) was approximately 10-fold higher than that of the conventional lipoplex. Pharmacokinetic studies demonstrated a higher distribution in lung by the SCR lipoplex. When the gene expression levels of the SCR lipoplex and conventional lipoplex were compared, the SCR lipoplex at a dose of 30 microg was compatible with that of the conventional lipoplex at a dose of 50 microg. A significantly higher serum alanine aminotransferase (ALT) activity and TNFalpha concentration was observed by the conventional lipoplex (pDNA dose; 50 microg), but this was not the case for the SCR lipoplex (pDNA dose; 30 microg). CONCLUSIONS We demonstrated that the SCR lipoplex could enhance the transfection efficacy in the lung without increasing the liver toxicity. Hence, the information will be valuable for the future use, design, and development of lipoplexes for in vivo applications.
Collapse
Affiliation(s)
- Shigeru Kawakami
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | | | | | |
Collapse
|
38
|
Weiss JM, Shivakumar R, Feller S, Li LH, Hanson A, Fogler WE, Fratantoni JC, Liu LN. Rapid, in vivo, evaluation of antiangiogenic and antineoplastic gene products by nonviral transfection of tumor cells. Cancer Gene Ther 2004; 11:346-53. [PMID: 15031722 DOI: 10.1038/sj.cgt.7700686] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Using a nonviral, electroporation-based gene transfection approach, we demonstrate the efficient and consistent transfection of two poorly immunogenic tumor cell lines: B16F10 melanoma and renal carcinoma (RENCA). Three genes, IL-12, angiostatin (AS), and an endostatin:angiostatin fusion protein (ES:AS) were subcloned into a DNA plasmid containing EBNA1-OriP, which was then transfected into B16F10 and RENCA cells. Significant levels of protein were secreted into the culture supernatants of transfected cells in vitro. Transfected tumor cells were injected subcutaneously into mice. All the three transgenes were capable of significantly delaying and reducing the formation of primary B16F10 and RENCA tumors, as well as B16F10 lung metastases. By day 11 post-injection, all control mice that received either mock-transfected or empty vector DNA-transfected B16F10 tumor cells had developed large primary tumors. In contrast, mice that received IL-12-transfected B16F10 cells did not develop appreciable tumors until day 17, and these were significantly smaller than controls. Similar results were observed for the RENCA model, in which only one of the IL-12 mice had developed tumors out to day 31. Expression of AS or ES:AS also significantly delayed and reduced primary tumors. Overall, ES:AS was more effective than AS alone. Furthermore, 25% of the AS mice and 33% of the ES:AS mice remained tumor-free at day 17, by which point all control mice had significant tumors. Mouse survival rates also correlated with the extent of tumor burden. Importantly, no lung metastases were detected in the lungs of mice that had received either AS or ES:AS-transfected B16F10 tumor cells and significantly fewer metastases were found in the IL-12 group. The consistency of our transfection results highlight the feasibility of directly electroporating tumor cells as a means to screen, identify, and validate in vivo potentially novel antiangiogenic and/or antineoplastic genes.
Collapse
MESH Headings
- Angiostatins/biosynthesis
- Angiostatins/genetics
- Animals
- Carcinoma, Renal Cell/blood supply
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Cell Division/genetics
- Cell Line, Tumor
- Cloning, Molecular
- Electroporation
- Endostatins/biosynthesis
- Endostatins/genetics
- Epstein-Barr Virus Nuclear Antigens/biosynthesis
- Epstein-Barr Virus Nuclear Antigens/genetics
- Gene Expression Regulation, Neoplastic
- Genetic Therapy
- Genetic Vectors
- Interleukin-12/biosynthesis
- Interleukin-12/genetics
- Kidney Neoplasms/blood supply
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Lung Neoplasms/secondary
- Male
- Melanoma/blood supply
- Melanoma/genetics
- Melanoma/metabolism
- Melanoma/pathology
- Mice
- Mice, Inbred BALB C
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Recombinant Fusion Proteins/biosynthesis
- Recombinant Fusion Proteins/genetics
- Transfection
- Viruses/genetics
Collapse
Affiliation(s)
- Jonathan M Weiss
- MaxCyte, Inc., 9640 Medical Center Drive, Rockville, Maryland 20850, USA
| | | | | | | | | | | | | | | |
Collapse
|