Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cancer Res Commun ; 3(7): 1248-1259, 2023 07.
Article in English | MEDLINE | ID: mdl-37465593

ABSTRACT

Siglec-15 (Sig15) has been implicated as an immune checkpoint expressed in solid tumor-infiltrating macrophages and is being targeted in clinical trials with mAbs to normalize the tumor immune microenvironment and stimulate antitumor immunity. However, the role of Sig15 in hematologic malignancies remains undefined. Sig15 mRNA and protein expression levels in hematologic malignancies were determined from publicly available databases, cell lines, and primary patient samples. Human B-cell acute lymphoblastic leukemia (B-ALL) cell lines were used to identify signaling pathways involved in the regulation of Sig15 expression. Secreted/soluble Sig15 and cytokine levels were measured from the plasma of children with leukemia and healthy controls. Knockdown and knockout of Siglec15 in a murine model of B-ALL was used to evaluate the effect of leukemia-derived Sig15 on the immune response to leukemia. We observed pathologic overexpression of Sig15 in a variety of hematologic malignancies, including primary B-ALL samples. This overexpression was driven by NFκB activation, which also increased the surface localization of Sig15. Secreted/soluble Sig15 was found to circulate at elevated levels in the plasma of children with B-ALL and correlated with an immune-suppressive cytokine milieu. Genetic inhibition of Sig15 in murine B-ALL promoted clearance of the leukemia by the immune system and a marked reversal of the immune-privileged leukemia bone marrow niche, including expanded early effector CD8+ T cells and reduction of immunosuppressive cytokines. Thus, Sig15 is a novel, potent immunosuppressive molecule active in leukemia that may be targeted therapeutically to activate T lymphocytes against leukemia cells. Significance: We demonstrate that Sig15 is overexpressed in hematologic malignancies driven by NFκB, is required for immune evasion in a mouse model of leukemia, and, for the first time, that it circulates at high levels in the plasma of children with leukemia.


Subject(s)
Burkitt Lymphoma , Hematologic Neoplasms , Leukemia , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Animals , Child , Humans , Mice , Adaptive Immunity , CD8-Positive T-Lymphocytes , Cytokines , Immunoglobulins , Membrane Proteins , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Sialic Acid Binding Immunoglobulin-like Lectins , Tumor Microenvironment/genetics
2.
Lab Invest ; 102(10): 1143-1149, 2022 10.
Article in English | MEDLINE | ID: mdl-35581307

ABSTRACT

Immune checkpoint blockade with programmed cell death (PD-1)/programmed death-ligand 1 (PD-L1) inhibitors has resulted in significant progress in the treatment of various cancer types. However, not all patients respond to PD-1/PD-L1 blockade, underscoring the importance of identifying new potential targets for immunotherapy. One promising target is the immune system modulator Siglec-15. In this study, we assess Siglec-15 expression in solid tumors, with a focus on lung, breast, head and neck squamous and bladder cancers. Using quantitative immunofluorescence (QIF) with a previously validated antibody, we found increased Siglec-15 expression in both tumor and immune cells in all the four cancer types. Siglec-15 was seen to be predominantly expressed by the stromal immune cells (83% in lung, 70.1% in breast, 95.2% in head and neck squamous cell and 89% in bladder cancers). Considerable intra-tumoral heterogeneity was noted across cancer types. As previously described for non-small cell lung cancer (NSCLC), Siglec-15 expression was seen to be mutually exclusive to PD-L1 in all the four cancer types, although this differential expression was maintained but somewhat diminished in head and neck squamous cell carcinoma (HNSCC). Siglec-15 was not prognostic either for overall survival (OS) or progression-free survival (PFS). In summary, we show broad expression of this potential immune modulatory target in a wide range of cancer types. These data suggest potential future clinical trials in these tumor types.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Head and Neck Neoplasms , Lung Neoplasms , Urinary Bladder Neoplasms , B7-H1 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Immune Checkpoint Inhibitors , Lung/metabolism , Lung Neoplasms/pathology , Programmed Cell Death 1 Receptor , Sialic Acid Binding Immunoglobulin-like Lectins/therapeutic use , Squamous Cell Carcinoma of Head and Neck , Urinary Bladder Neoplasms/drug therapy
3.
Lab Invest ; 102(7): 771-778, 2022 07.
Article in English | MEDLINE | ID: mdl-35459795

ABSTRACT

Siglec-15, a member of sialic-acid binding immunoglobulin type lectins, is normally expressed by myeloid cells and upregulated in some human cancers and represents a promising new target for immunotherapy. While PD-L1 blockade is an important strategy for immunotherapy, its effectiveness is limited. The expression of Siglec-15 has been demonstrated to be predominantly mutually exclusive to PD-L1 in certain cancer histologies. Thus, there is significant opportunity for Siglec-15 as an immunotherapeutic target for patients that do not respond to PD-1/PD-L1 inhibition. The aim of this study was to prospectively develop an immunohistochemical (IHC) assay for Siglec-15 to be used as a companion diagnostic for future clinical trials. Here, we create and validate an IHC assay with a novel recombinant antibody to the cytoplasmic domain of Siglec-15. To find an enriched target, this antibody was first used in a quantitative fluorescence (QIF) assay to screen a broad range of tumor histologies to determine tumor types where Siglec-15 demonstrated high expression. Based on this and previous data, we focused on development of a chromogenic IHC assay for lung cancer. Then we developed a scoring system for this assay that has high concordance amongst pathologist readers. We then use this chromogenic IHC assay to test the expression of Siglec-15 in two cohorts of NSCLC. We found that this assay shows a higher level of staining in both tumor and immune cells compared to previous QIF assays utilizing a polyclonal antibody. However, similar to that study, only a small percentage of positive Siglec-15 cases showed high expression for PD-L1. This validated assay for Siglec-15 expression may support development of a companion diagnostic assay to enrich for patients expressing the Siglec-15 target for therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Antibodies, Monoclonal/therapeutic use , B7-H1 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Immunohistochemistry , Lung Neoplasms/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/therapeutic use
4.
Bone Res ; 9(1): 47, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34719673

ABSTRACT

Osteoporosis (OP) is a common age-related disease characterized by a deterioration of bone mass and structure that predisposes patients to fragility fractures. Pharmaceutical therapies that promote anabolic bone formation in OP patients and OP-induced fracture are needed. We investigated whether a neutralizing antibody against Siglec-15 can simultaneously inhibit bone resorption and stimulate bone formation. We found that the multinucleation of osteoclasts was inhibited in SIGLEC-15 conditional knockout mice and mice undergoing Siglec-15 neutralizing antibody treatment. The secretion of platelet-derived growth factor-BB (PDGF-BB), the number of tartrate-resistant acid phosphatase-positive (TRAP+) mononuclear cells, and bone formation were significantly increased in the SIGLEC-15 conditional knockout mice and antibody-treated mice. The anabolic effect of the Siglec-15 neutralizing antibody on bone formation was blunted in mice with Pdgfb deleted in TRAP+ cells. These findings showed that the anabolic effect of the Siglec-15 neutralizing antibody was mediated by elevating PDGF-BB production of TRAP+ mononuclear cells. To test the therapeutic potential of the Siglec-15 neutralizing antibody, we injected the antibody in an ovariectomy-induced osteoporotic mouse model, which mimics postmenopausal osteoporosis in women, and in two fracture healing models because fracture is the most serious health consequence of osteoporosis. The Siglec-15 neutralizing antibody effectively reduced bone resorption and stimulated bone formation in estrogen deficiency-induced osteoporosis. Of note, the Siglec-15 neutralizing antibody promoted intramembranous and endochondral ossification at the damaged area of cortical bone in fracture healing mouse models. Thus, the Siglec-15 neutralizing antibody shows significant translational potential as a novel therapy for OP and bone fracture.

5.
Nat Med ; 25(4): 656-666, 2019 04.
Article in English | MEDLINE | ID: mdl-30833750

ABSTRACT

Overexpression of the B7-H1 (PD-L1) molecule in the tumor microenvironment (TME) is a major immune evasion mechanism in some patients with cancer, and antibody blockade of the B7-H1/PD-1 interaction can normalize compromised immunity without excessive side-effects. Using a genome-scale T cell activity array, we identified Siglec-15 as a critical immune suppressor. While only expressed on some myeloid cells normally, Siglec-15 is broadly upregulated on human cancer cells and tumor-infiltrating myeloid cells, and its expression is mutually exclusive to B7-H1, partially due to its induction by macrophage colony-stimulating factor and downregulation by IFN-γ. We demonstrate that Siglec-15 suppresses antigen-specific T cell responses in vitro and in vivo. Genetic ablation or antibody blockade of Siglec-15 amplifies anti-tumor immunity in the TME and inhibits tumor growth in some mouse models. Taken together, our results support Siglec-15 as a potential target for normalization cancer immunotherapy.


Subject(s)
Immunoglobulins/metabolism , Immunotherapy , Membrane Proteins/metabolism , Neoplasms/immunology , Neoplasms/therapy , Animals , Cell Line, Tumor , Cell Proliferation , Epitopes , Humans , Macrophages/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Cells/metabolism , Neoplasms/pathology , Proteome/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Lymphocytes/immunology
6.
J Immunol ; 201(3): 897-907, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29898965

ABSTRACT

The potent immune regulatory function of an agonistic B7-H4-Ig fusion protein (B7-H4Ig) has been demonstrated in multiple experimental autoimmune models; however, the identity of a functional B7-H4 receptor remained unknown. The biological activity of B7-H4 is associated with decreased inflammatory CD4+ T cell responses as supported by a correlation between B7-H4-expressing tumor-associated macrophages and Foxp3+ T cells within the tumor microenvironment. Recent data indicate that members of the semaphorin (Sema)/plexin/neuropilin (Nrp) family of proteins both positively and negatively modulate immune cell function. In this study, we show that B7-H4 binds the soluble Sema family member Sema3a. Additionally, B7-H4Ig-induced inhibition of inflammatory CD4+ T cell responses is lost in both Sema3a functional mutant mice and mice lacking Nrp-1 expression in Foxp3+ T cells. These findings indicate that B7-H4Ig binds to Sema3a, which acts as a functional bridge to stimulate an Nrp-1/Plexin A4 heterodimer to form a functional immunoregulatory receptor complex resulting in increased levels of phosphorylated PTEN and enhanced regulatory CD4+ T cell number and function.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Neuropilin-1/metabolism , Receptors, Cell Surface/metabolism , Semaphorin-3A/metabolism , V-Set Domain-Containing T-Cell Activation Inhibitor 1/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Female , Forkhead Transcription Factors/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , PTEN Phosphohydrolase/metabolism , Tumor Microenvironment/immunology
7.
Cancer Res ; 76(2): 227-38, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26637667

ABSTRACT

Alterations in EGFR, KRAS, and ALK are oncogenic drivers in lung cancer, but how oncogenic signaling influences immunity in the tumor microenvironment is just beginning to be understood. Immunosuppression likely contributes to lung cancer, because drugs that inhibit immune checkpoints like PD-1 and PD-L1 have clinical benefit. Here, we show that activation of the AKT-mTOR pathway tightly regulates PD-L1 expression in vitro and in vivo. Both oncogenic and IFNγ-mediated induction of PD-L1 was dependent on mTOR. In human lung adenocarcinomas and squamous cell carcinomas, membranous expression of PD-L1 was significantly associated with mTOR activation. These data suggest that oncogenic activation of the AKT-mTOR pathway promotes immune escape by driving expression of PD-L1, which was confirmed in syngeneic and genetically engineered mouse models of lung cancer where an mTOR inhibitor combined with a PD-1 antibody decreased tumor growth, increased tumor-infiltrating T cells, and decreased regulatory T cells.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Oncogene Protein v-akt/metabolism , Programmed Cell Death 1 Receptor/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Lung Neoplasms/pathology , Mice , Transfection
8.
J Autoimmun ; 44: 71-81, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23683881

ABSTRACT

We evaluated the therapeutic efficacy and mechanisms of action of both mouse and human B7-H4 Immunoglobulin fusion proteins (mB7-H4Ig; hB7-H4Ig) in treating EAE. The present data show that mB7-H4Ig both directly and indirectly (via increasing Treg function) inhibited CD4⁺ T-cell proliferation and differentiation in both Th1- and Th17-cell promoting conditions while inducing production of IL-10. B7-H4Ig treatment effectively ameliorated progression of both relapsing (R-EAE) and chronic EAE correlating with decreased numbers of activated CD4⁺ T-cells within the CNS and spleen, and a concurrent increase in number and function of Tregs. The functional requirement for Treg activation in treating EAE was demonstrated by a loss of therapeutic efficacy of hB7-H4Ig in R-EAE following inactivation of Treg function either by anti-CD25 treatment or blockade of IL-10. Significant to the eventual translation of this treatment into clinical practice, hB7-H4Ig similarly inhibited the in vitro differentiation of naïve human CD4⁺ T-cells in both Th1- and Th17-promoting conditions, while promoting the production of IL-10. B7-H4Ig thus regulates pro-inflammatory T-cell responses by a unique dual mechanism of action and demonstrates significant promise as a therapeutic for autoimmune diseases, including MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Immunoglobulins/pharmacology , Interleukin-10/immunology , T-Lymphocytes/drug effects , V-Set Domain-Containing T-Cell Activation Inhibitor 1/pharmacology , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Proliferation/drug effects , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Humans , Immunoglobulins/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , T-Lymphocytes/immunology , V-Set Domain-Containing T-Cell Activation Inhibitor 1/immunology
9.
Stem Cells Transl Med ; 2(5): 362-75, 2013 May.
Article in English | MEDLINE | ID: mdl-23592838

ABSTRACT

Rheumatoid arthritis (RA) is a systemic autoimmune disease with unknown etiology where tumor necrosis factor-α (TNFα) plays a critical role. Etanercept, a recombinant fusion protein of human soluble tumor necrosis factor receptor II (hsTNFR) linked to the Fc portion of human IgG1, is used to treat RA based on the rationale that sTNFR binds TNFα and blocks TNFα-mediated inflammation. We compared hsTNFR protein delivery from genetically engineered human mesenchymal stem cells (hMSCs) with etanercept. Blocking TNFα-dependent intercellular adhesion molecule-1 expression on transduced hMSCs and inhibition of nitric oxide production from TNFα-treated bovine chondrocytes by conditioned culture media from transduced hMSCs demonstrated the functionality of the hsTNFR construction. Implanted hsTNFR-transduced mesenchymal stem cells (MSCs) reduced mouse serum circulating TNFα generated from either implanted TNFα-expressing cells or lipopolysaccharide induction more effectively than etanercept (TNFα, 100%; interleukin [IL]-1α, 90%; and IL-6, 60% within 6 hours), suggesting faster clearance of the soluble tumor necrosis factor receptor (sTNFR)-TNFα complex from the animals. In vivo efficacy of sTNFR-transduced MSCs was illustrated in two (immune-deficient and immune-competent) arthritic rodent models. In the antibody-induced arthritis BalbC/SCID mouse model, intramuscular injection of hsTNFR-transduced hMSCs reduced joint inflammation by 90% compared with untransduced hMSCs; in the collagen-induced arthritis Fischer rat model, both sTNFR-transduced rat MSCs and etanercept inhibited joint inflammation by 30%. In vitro chondrogenesis assays showed the ability of TNFα and IL1α, but not interferon γ, to inhibit hMSC differentiation to chondrocytes, illustrating an additional negative role for inflammatory cytokines in joint repair. The data support the utility of hMSCs as therapeutic gene delivery vehicles and their potential to be used in alleviating inflammation within the arthritic joint.


Subject(s)
Adult Stem Cells/cytology , Arthritis, Experimental/drug therapy , Arthritis, Experimental/prevention & control , Genetic Engineering , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Receptors, Tumor Necrosis Factor, Type II/therapeutic use , Adult Stem Cells/metabolism , Animals , Arthritis, Experimental/pathology , Cattle , Cell Differentiation , Chondrogenesis , Cytokines/metabolism , Disease Models, Animal , Humans , Inflammation/complications , Inflammation/pathology , Inflammation/therapy , Inflammation Mediators/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Rats , Solubility , Transduction, Genetic , Treatment Outcome
10.
Int Immunopharmacol ; 15(3): 488-97, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23474736

ABSTRACT

Recent commercial approval of cancer vaccine, demonstrating statistically significant improvement in overall survival of prostate cancer patients has spurred renewed interest in active immunotherapies; specifically, strategies that lead to enhanced biological activity and robust efficacy for dendritic cell vaccines. A simple, widely used approach to generating multivalent cancer vaccines is to load tumor whole cell lysates into dendritic cells (DCs). Current DC vaccine manufacturing processes require co-incubation of tumor lysate antigens with immature DCs and their subsequent maturation. We compared electroloading of tumor cell lysates directly into mature DCs with the traditional method of lysate co-incubation with immature DCs. Electroloaded mature DCs were more potent in vitro, as judged by their ability to elicit significantly (p < 0.05) greater expansion of peptide antigen-specific CD8(+) T cells, than either lysate-electroloaded immature DCs or lysate-co-incubated immature DCs, both of which must be subsequently matured. Expanded CD8(+) T cells were functional as judged by their ability to produce IFN-γ upon antigen-specific re-stimulation. The electroloading technology used herein is an automated, scalable, functionally closed cGMP-compliant manufacturing technology supported by a Master File at CBER, FDA and represents an opportunity for translation of enhanced potency DC vaccines at clinical/commercial scale.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/metabolism , Electroporation/methods , Immunotherapy, Adoptive/methods , Melanoma/immunology , T-Lymphocytes, Cytotoxic/immunology , Antigen Presentation , Antigens, Neoplasm/immunology , CD8 Antigens/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Coculture Techniques , Dendritic Cells/cytology , Feasibility Studies , Humans , Interferon-gamma/metabolism , Lymphocyte Activation , Melanoma/therapy
11.
Methods Mol Biol ; 423: 139-53, 2008.
Article in English | MEDLINE | ID: mdl-18370195

ABSTRACT

Results from multiple human studies have continued to spur the development of dendritic cells (DCs) as therapeutic vaccines for the treatment of cancer, chronic viral infections, and autoimmune diseases. The antigen-specific activity of DCs is dependent on the ability of the DCs to take up and process tumor-associated antigens for presentation to the immune system. Although immature DCs have been shown to naturally take up tumor-associated antigens by phagocytosis, approaches that significantly affect antigen delivery need further evaluation, especially if such methodologies can be demonstrated to result in the elicitation of more robust and comprehensive immune responses. We have developed a rapid, robust, scalable, and regulatory-compliant process for loading DCs with whole tumor lysate. The use of whole tumor lysate facilitates the generation of a more robust immune response targeting multiple unique antigenic determinants in patient's tumors and likely reduces the tumor's potential of immune escape. We demonstrate that DCs electroloaded with tumor lysate elicit significantly stronger antitumor responses both in a tumor challenge model and in a therapeutic vaccination model for preexisting metastasic disease. These effects are observed in a processing scheme that requires 20- to 40-fold lower amounts of tumor lysate when compared with the standard coincubation/coculture methods employed in loading DCs.


Subject(s)
Cancer Vaccines/administration & dosage , Dendritic Cells/immunology , Animals , Antigens, Neoplasm/administration & dosage , Cancer Vaccines/isolation & purification , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/secondary , Carcinoma, Lewis Lung/therapy , Cell Line, Tumor , Cytotoxicity, Immunologic , Electrochemotherapy/methods , Humans , Immunotherapy/methods , Kidney Neoplasms/immunology , Kidney Neoplasms/therapy , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/therapy
12.
J Immunother ; 28(6): 542-50, 2005.
Article in English | MEDLINE | ID: mdl-16224271

ABSTRACT

Electroporation of dendritic cells (DCs) with tumor lysate elicited greater antitumor responses in vitro and in vivo, using less lysate than standard coincubation. Electroloaded DCs had normal surface marker expression and matured into competent antigen-presenting cells. In a renal carcinoma (RENCA) model, mice were pretreated with lysate-loaded DCs before tumor challenge. Mice that received DCs electroloaded with RENCA lysate had significantly smaller tumors (9+/-6 mm2) than mice given DCs coincubated with the same lysate (23+/-5 mm2). To evaluate a metastatic therapeutic tumor model, mice were first injected with Lewis lung carcinoma (LLC) and then given 2 doses of cryopreserved LLC lysate-loaded DCs. Mice treated with electroloaded DCs had a 50% reduction in lung metastases compared with control mice that received no DCs or DCs loaded with liver lysate. In contrast, DCs coincubated with LLC lysate were indistinguishable from controls. Tumor lysate-electroloaded but not-coincubated DCs also primed syngeneic mouse splenocytes in vitro to produce interferon-gamma and, specifically, lyse tumor cells. The electroloaded DCs elicited specific T-cell responses with less lysate than the amount reported in standard coincubation procedures. This approach may be particularly useful when small amounts of tumor material are available.


Subject(s)
Antigens, Neoplasm/immunology , Carcinoma, Renal Cell/therapy , Dendritic Cells/immunology , Electroporation , Kidney Neoplasms/therapy , Animals , Cancer Vaccines/immunology , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/therapy , Carcinoma, Renal Cell/immunology , Cell Line, Tumor , Dextrans/metabolism , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/metabolism , Immunotherapy , Kidney Neoplasms/immunology , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Transplantation , Spleen/cytology , Spleen/immunology , T-Lymphocytes/immunology
13.
Cancer Gene Ther ; 11(5): 346-53, 2004 May.
Article in English | MEDLINE | ID: mdl-15031722

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.


Subject(s)
Carcinoma, Renal Cell/metabolism , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/metabolism , Melanoma/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Angiostatins/biosynthesis , Angiostatins/genetics , Animals , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/genetics , 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 , Genetic Therapy , Genetic Vectors , Interleukin-12/biosynthesis , Interleukin-12/genetics , Kidney Neoplasms/blood supply , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Lung Neoplasms/secondary , Male , Melanoma/blood supply , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Transfection , Viruses/genetics
14.
Technol Cancer Res Treat ; 1(5): 341-50, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12625759

ABSTRACT

Electroporation is widely used to transfect and load cells with various molecules. Traditional electroporation using a static mode is typically restricted to volumes less than 1 mL, which limits its use in clinical and industrial bioprocessing applications. Here we report efficient, large volume transfection results by using a scalable-volume electroporation system. Suspended (Jurkat) and adherent cells (10T1/2 and Huh-7) were tested. A large macromolecule, FITC-conjugated dextran (MW=500 kD) was used to measure cell uptake, while a plasmid carrying the gene coding for enhanced green fluorescence protein (eGFP) was used to quantitate the flow electrotransfection efficiency as determined by flow cytometry. The flow electroloading efficiency of FITC-dextran was >90%, while the cell viability was highly maintained (>90%). High flow electrotransfection efficiency (up to 75%) and cell viability (up to 90%) were obtained with processing volumes ranging from 1.5 to 50 mL. No significant difference of electrotransfection efficiency was observed between flow and static electrotransfection. When 50 mL of cell volume was processed and samples collected at different time points during electroporation, the transgene expression and cell viability results were identical. We also demonstrated that DNA plasmid containing EBNA1-OriP elements from Epstein-Barr virus were more efficient in transgene expression than standard plasmid without the elements (at least 500 too 1000-fold increase in expression level). Finally, to examine the feasibility of utilizing flow electrotransfected cells as a gene delivery vehicle, 10T1/2 cells were transfected with a DNA plasmid containing the gene coding for mIL12. mIL12 transfected cells were injected subcutaneously into mice, and produced functional mIL12, as demonstrated by anti-angiogenic activity. This is the first demonstration of efficient, large volume, flow electroporation and the in vivo efficacy of flow electrotransfected cells. This technology may be useful for clinical gene therapy and large-scale bioprocesses.


Subject(s)
Electroporation/methods , Animals , Cell Adhesion , Cell Line , Cell Survival , Coloring Agents/pharmacology , Flow Cytometry , Herpesvirus 4, Human/genetics , Humans , Jurkat Cells , Mice , Neovascularization, Pathologic , Plasmids/metabolism , Time Factors , Transfection , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...