Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Molecules ; 27(23)2022 Dec 02.
Article in English | MEDLINE | ID: mdl-36500549

ABSTRACT

The majority of clear cell renal cell carcinomas (ccRCCs) are characterized by mutations in the Von Hippel−Lindau (VHL) tumor suppressor gene, which leads to the stabilization and accumulation of the HIF2α transcription factor that upregulates key oncogenic pathways that promote glucose metabolism, cell cycle progression, angiogenesis, and cell migration. Although FDA-approved HIF2α inhibitors for treating VHL disease-related ccRCC are available, these therapies are associated with significant toxicities such as anemia and hypoxia. To improve ccRCC-specific drug delivery, peptide amphiphile micelles (PAMs) were synthesized incorporating peptides targeted to the CD70 marker expressed by ccRCs and anti-HIF2α siRNA, and the ability of HIF2α-CD27 PAMs to modulate HIF2α and its downstream targets was evaluated in human ccRCC patient-derived cells. Cell cultures were derived from eight human ccRCC tumors and the baseline mRNA expression of HIF2A and CD70, as well as the HIF2α target genes SLC2A1, CCND1, VEGFA, CXCR4, and CXCL12 were first determined. As expected, each gene was overexpressed by at least 63% of all samples compared to normal kidney proximal tubule cells. Upon incubation with HIF2α-CD27 PAMs, a 50% increase in ccRCC-binding was observed upon incorporation of a CD70-targeting peptide into the PAMs, and gel shift assays demonstrated the rapid release of siRNA (>80% in 1 h) under intracellular glutathione concentrations, which contributed to ~70% gene knockdown of HIF2α and its downstream genes. Further studies demonstrated that knockdown of the HIF2α target genes SLC2A1, CCND1, VEGFA, CXCR4, and CXCL12 led to inhibition of their oncogenic functions of glucose transport, cell proliferation, angiogenic factor release, and cell migration by 50−80%. Herein, the development of a nanotherapeutic strategy for ccRCC-specific siRNA delivery and its potential to interfere with key oncogenic pathways is presented.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , RNA, Small Interfering/genetics , Micelles , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , CD27 Ligand/genetics , CD27 Ligand/metabolism
2.
Oncotarget ; 13: 1054-1067, 2022.
Article in English | MEDLINE | ID: mdl-36128328

ABSTRACT

Loss-of-function mutations in genes encoding the Krebs cycle enzymes Fumarate Hydratase (FH) and Succinate Dehydrogenase (SDH) induce accumulation of fumarate and succinate, respectively and predispose patients to hereditary cancer syndromes including the development of aggressive renal cell carcinoma (RCC). Fumarate and succinate competitively inhibit αKG-dependent dioxygenases, including Lysine-specific demethylase 4A/B (KDM4A/B), leading to suppression of the homologous recombination (HR) DNA repair pathway. In this study, we have developed new syngeneic Fh1- and Sdhb-deficient murine models of RCC, which demonstrate the expected accumulation of fumarate and succinate, alterations in the transcriptomic and methylation profile, and an increase in unresolved DNA double-strand breaks (DSBs). The efficacy of poly ADP-ribose polymerase inhibitors (PARPis) and temozolomide (TMZ), alone and in combination, was evaluated both in vitro and in vivo. Combination treatment with PARPi and TMZ results in marked in vitro cytotoxicity in Fh1- and Sdhb-deficient cells. In vivo, treatment with standard dosing of the PARP inhibitor BGB-290 and low-dose TMZ significantly inhibits tumor growth without a significant increase in toxicity. These findings provide the basis for a novel therapeutic strategy exploiting HR deficiency in FH and SDH-deficient RCC with combined PARP inhibition and low-dose alkylating chemotherapy.


Subject(s)
Carcinoma, Renal Cell , Dioxygenases , Kidney Neoplasms , Adenosine Diphosphate Ribose , Animals , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Citric Acid Cycle , DNA , Fumarate Hydratase/genetics , Fumarates , Humans , Jumonji Domain-Containing Histone Demethylases , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Lysine , Mice , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Succinate Dehydrogenase/genetics , Succinates , Temozolomide/pharmacology
3.
Urology ; 99: 270-277, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27720772

ABSTRACT

OBJECTIVE: To evaluate the safety and feasibility of single and serial instillations of MitoGel into the upper urinary tract using a preclinical swine animal model. MitoGel is a novel sustained release formulation of mitomycin C (MMC) based on RTGel, a proprietary thermosensitive hydrogel technology. MitoGel is liquid at cold temperatures and solidifies to gel state at body temperature. It is intended as a treatment for upper tract urothelial carcinoma, given its ability to provide sustained release of MMC in the upper urinary tract. MATERIALS AND METHODS: We utilized 23 pigs in a 3-phase design. All animals underwent bilateral nephrostomy tube placement. During phase 1, 3 animals underwent antegrade RTGel instillation, imaging, and euthanasia within 12 hours. In phase 2, 10 pigs underwent single antegrade instillation, unilateral nephrectomy 3 days following instillation, and contralateral nephrectomy and euthanasia 30 days following instillation. During phase 3, 10 animals underwent 6 instillations over 3 weeks, followed by bilateral nephrectomy and necropsy 30 days postinstillation. MitoGel (2 mg/mL and 4 mg/mL), aqueous MMC (2 mg/mL and 4 mg/mL), and RTGel alone were evaluated. RESULTS: MitoGel remained visible within the pelvicalyceal system on fluoroscopic and computed tomography imaging for 4-6 hours. MMC plasma levels were well within acceptable safety thresholds. There was no evidence of urinary obstruction, acute kidney injury, sepsis, or myelosuppression. Histologic changes in the urinary system were mild and transient. CONCLUSION: Antegrade MitoGel delivery to the pelvicalyceal system of Yorkshire swine is feasible and safe. Further evaluation of MitoGel in human clinical trials is warranted.


Subject(s)
Carcinoma, Transitional Cell/drug therapy , Mitomycin/administration & dosage , Neoplasms, Experimental , Polymers/administration & dosage , Urinary Bladder Neoplasms/drug therapy , Administration, Intravesical , Animals , Cross-Linking Reagents/administration & dosage , Delayed-Action Preparations , Drug Evaluation, Preclinical , Feasibility Studies , Female , Instillation, Drug , Swine
4.
J Urol ; 182(2): 728-34, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19539328

ABSTRACT

PURPOSE: We assessed CA9 single nucleotide polymorphisms and mutations, and their association with CAIX protein expression, overall survival and response to interleukin-2 in white patients with metastatic clear cell renal cell carcinoma. MATERIALS METHODS: Genomic DNA was extracted from frozen tumor samples of 54 metastatic clear cell renal cell carcinomas. The CA9 gene exons and flanking regions were amplified by polymerase chain reaction and sequenced. The monoclonal antibody M75 was used to evaluate CAIX protein expression in the primary tumor by immunohistochemistry. RESULTS: CA9 reference single nucleotide polymorphisms rs2071676, rs12553173, rs3829078 and rs1048638 were found in 59%, 15%, 11% and 33% of patients, respectively. The deletion c.376del393 was observed in 2 patients. CAIX expression was greater than 85% in 65% of patients. No single nucleotide polymorphisms were significantly associated with CAIX expression. Patients with the C allele variant of rs12553173 had improved median survival (27.3 vs 13.6 months, p = 0.0431) and a greater likelihood of an interleukin-2 response (57% vs 22%, p = 0.081) Likewise high CAIX expression was associated with longer median survival (25.5 vs 8.5 months, p <0.0001) and a greater interleukin-2 response rate (37% vs 8%, p = 0.070). In a multivariate Cox model the C allele variant of CA9 single nucleotide polymorphism rs12553173 and CAIX expression were retained as independent prognostic factors. CONCLUSIONS: CA9 single nucleotide polymorphisms are common in patients with metastatic clear cell renal cell carcinoma. The synonymous C allele variant of rs12553173 may be associated with improved overall survival and a greater likelihood of a response to interleukin-2. CA9 rs12553173 and CAIX are independent prognostic factors of overall survival and complementary for predicting the prognosis of metastatic clear cell renal cell carcinoma.


Subject(s)
Antigens, Neoplasm/genetics , Carbonic Anhydrases/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/secondary , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Polymorphism, Single Nucleotide , Adult , Aged , Antineoplastic Agents/therapeutic use , Carbonic Anhydrase IX , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/mortality , Female , Humans , Interleukin-2/therapeutic use , Kidney Neoplasms/drug therapy , Kidney Neoplasms/mortality , Male , Middle Aged , Prognosis , Survival Rate
5.
Urology ; 68(5): 1132-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17095063

ABSTRACT

OBJECTIVES: Carbonic anhydrase-IX (CA-IX) is a cell surface tumor-associated antigen expressed by most clear cell renal cell carcinomas (RCCs). The specificity and the prognostic value of CA-IX provide impetus to create a mouse model of CA-IX-expressing RCC for testing CA-IX-targeted therapies against RCC. METHODS: A retrovirus encoding the human CA-IX gene was used to transduce the murine RCC line, RENCA. In vivo growth kinetics and CA-IX expression were compared between RENCA and RENCA/CA-IX using heterotopic, metastatic, and orthotopic models. RESULTS: Transduction of RENCA created the RENCA/CA-IX line with nearly 100% CA-IX surface expression. In the heterotopic model, subcutaneous injection of 500,000 and 50,000 cells led to tumor formation at 2 to 2.5 weeks after injection, with similar growth kinetics between the two cell lines at either cell number. In the pulmonary metastatic model, a similar number of metastases was noted after inoculation of RENCA and RENCA/CA-IX. In the orthotopic model, autopsy revealed a CA-IX-expressing renal tumor, as well as CA-IX-expressing metastases to the lungs, liver, contralateral kidney, intestines, and lymph nodes. In all the above models, the RENCA/CA-IX tumors retained expression of CA-IX, as demonstrated by immunohistochemistry staining. CONCLUSIONS: RENCA/CA-IX is the first tumor model that manifests in immunocompetent Balb/c mice and stably expresses a defined kidney cancer-associated antigen. It maintains antigen expression, forms metastases, and produces reliable tumor growth kinetics equivalent to that of its parental cell line.


Subject(s)
Carbonic Anhydrases/biosynthesis , Carcinoma, Renal Cell/enzymology , Disease Models, Animal , Kidney Neoplasms/enzymology , Animals , Carbonic Anhydrase IX , Carbonic Anhydrases/analysis , Carcinoma, Renal Cell/chemistry , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/chemistry , Kidney Neoplasms/pathology , Mice , Mice, Inbred BALB C
6.
Clin Cancer Res ; 10(4): 1421-9, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14977845

ABSTRACT

PURPOSE: Carbonic anhydrase 9 (CA9) is the most promising molecular marker described for renal cell carcinoma (RCC) to date. We investigated whether transduction of monocytes from peripheral blood with adenovirus encoding the CA9 gene (AdV-CA9) could stimulate a T-cell mediated immune response against cancer cells expressing CA9. The ability to consistently generate a T-cell response is an important step toward the development of a CA9-specific RCC vaccine. EXPERIMENTAL DESIGN: AdV-CA9 was generated using the AdEasy system. AdV-CA9-transduced peripheral blood mononuclear cell (PBMC)-derived monocytes were used to raise CTLs from autologous peripheral blood lymphocytes (PBLs). The ability of CTLs to lyse targets expressing CA9 was assessed by (51)Cr-release. RESULTS: Monocytes were efficiently transduced with AdV-CA9. In five of six experiments, AdV-CA9-transduced monocytes were able to induce a population of CTLs from bulk PBLs. CTLs were capable of lysing autologous, but not allogeneic monocytes expressing CA9. Furthermore, CTLs were able to lyse autologous RCC tumor cells expressing CA9. The ability of CTLs to lyse relevant targets was blocked by anti-CD3, anti-CD8, and anti-MHC class I antibodies demonstrating a MHC class I restricted response. CONCLUSIONS: These results suggest that PBMC-derived monocytes transduced with AdV-CA9 can generate RCC-specific MHC class I restricted CTLs capable of lysing CA9-expressing cancer cells. Transduction of PBMC-derived monocytes with adenovirus provides a simple and effective alternative to the use of dendritic cells for the induction of antigen-specific CTL.


Subject(s)
Adenoviridae/genetics , Cancer Vaccines , Carbonic Anhydrases/genetics , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/therapy , Kidney Neoplasms/immunology , Kidney Neoplasms/therapy , Monocytes/immunology , Blotting, Western , Cell Adhesion , Cell Line, Tumor , Cloning, Molecular , Cytokines/metabolism , Flow Cytometry , Genetic Vectors , Humans , Immunotherapy , Leukocytes, Mononuclear/metabolism , Microscopy, Phase-Contrast , Monocytes/metabolism , Phenotype , T-Lymphocytes/metabolism
7.
Cancer Res ; 63(16): 4952-9, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12941820

ABSTRACT

A metastatic renal cell carcinoma (RCC) tumor model xenograft that expresses the targetable, membrane-bound tumor-associated antigen carbonic anhydrase type 9 (CA IX) is described. The xenograft, established from a high-grade type-2 chromophil RCC (cRCC), has been serially transplanted in immune compromised mice, in which it grows orthotopically under the renal capsule, doubling its size every 9 weeks and sending metastases to the lung and liver at approximately 20 weeks. Tumors were capable of being imaged using a micro-PET (micro-positron emission tomograph) with an 18-fluorodeoxyglucose (18-FDG) tracer. Subsequent xenograft generations have conserved immunohistochemical and ultrastructural properties typical for malignant renal epithelium-derived neoplasia (vimentin+, CK-19+, CA IX+ with hypoxia-inducible factor (HIF)-1 alpha constitutive expression) and have demonstrated extensive proliferation, lack of apoptosis, severe genetic alterations, and molecular expression alterations; transforming growth factor beta 1 (TGF-beta 1), hepatocyte growth factor (HGF), proto-oncogene (c-met), matrix metalloproteinase (MMP)-1, and vascular endothelial growth factor (VEGF) C and D were overexpressed, whereas human epidermal growth factor receptor (HER)-2, MMP-2 and MMP-9, VEGF-R3, p53, and p27 were severely down-regulated, suggesting a proangiogenic environment, local invasiveness, and facilitated lymphatic metastasis. Altogether, LABAZ1 provides a relevant and flexible model to study the biology of cRCC, the role of CA IX in RCC tumorigenesis, progression, and metastasis, and a platform for testing new targeted therapeutic strategies.


Subject(s)
Antigens, Neoplasm/analysis , Carbonic Anhydrases/analysis , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/secondary , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Neoplasm Proteins/analysis , Aged , Animals , Carbonic Anhydrase IX , Carcinoma, Renal Cell/genetics , Disease Models, Animal , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Immunohistochemistry , Kidney Neoplasms/genetics , Male , Mice , Mice, SCID , Microscopy, Electron , Proto-Oncogene Mas , Transcription Factors/analysis , Translocation, Genetic
8.
Clin Cancer Res ; 9(5): 1906-16, 2003 May.
Article in English | MEDLINE | ID: mdl-12738749

ABSTRACT

PURPOSE: We investigated the ability of the fusion protein granulocyte-macrophage colony-stimulating factor and carbonic anhydrase IX (GMCA-9)(1) to induce an immune response in vitro and in vivo for the development of a GMCA-9-based kidney cancer vaccine. EXPERIMENTAL DESIGN: Human dendritic cells (DCs) were transduced with a recombinant adenovirus containing the GMCA-9 gene and tested for their capacity to induce CA9-specific cytotoxic T lymphocytes in vitro. Tumor growth was studied in severe compromised immunodeficiency disease (SCID) mice s.c. injected with R11-GMCA-9, a human renal cell carcinoma cell line stably transfected with the GMCA-9 gene. Involvement of natural killer (NK) cells in the antitumor activity of GMCA-9 was determined in SCID mice treated with the NK-blocking agent anti-asialoGM-1. RESULTS: DC and R11 cells transduced with GMCA-9 produced a GMCA-9 protein that is targeted to the cell membrane and partially processed to granulocyte macrophage colony-stimulating factor- and CA9-like products. Furthermore, GMCA-9 was capable of inducing DC maturation, as well as CA9-specific cytotoxic lymphocytes in vitro. Tumor growth of R11 cells in SCID mice was significantly inhibited after transfection with the GMCA-9 fusion gene (P < 0.01). In mice treated with anti-asialoGM-1, R11-GMCA-9 tumors grew significantly faster than those of control mice (P < 0.05), suggesting an involvement of NK cells. CONCLUSIONS: Our results suggest that the fusion protein GMCA-9 is capable of generating an immune response both in vitro and in vivo. Additional studies will confirm the utility of ex vivo GMCA-9-transduced DCs as a kidney cancer vaccine.


Subject(s)
Antigens, Neoplasm/genetics , Carbonic Anhydrases/genetics , Carcinoma, Renal Cell/therapy , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Immunotherapy/methods , Kidney Neoplasms/therapy , Neoplasm Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Adenoviridae/genetics , Animals , Carbonic Anhydrase IX , Carcinoma, Renal Cell/immunology , Dendritic Cells/drug effects , Dendritic Cells/physiology , Genetic Therapy/methods , Genetic Vectors/genetics , Genetic Vectors/pharmacology , Humans , Isoenzymes , Kidney Neoplasms/immunology , Killer Cells, Natural/immunology , Male , Mice , Mice, SCID , Mice, Transgenic , Recombinant Fusion Proteins/genetics , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/physiology , Transduction, Genetic
9.
Int J Oncol ; 22(2): 431-7, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12527945

ABSTRACT

Most anti-cancer therapies induce apoptotic cell death, but a major barrier to long-term cancer treatments is the generation of apoptosis-resistant tumor cells. Tumor cells that become resistant to one therapy are usually cross-resistant to subsequent therapies, including those with different cellular/molecular targets, suggesting that resistant tumor cells acquire modifications of the general apoptotic pathway. Most solid tumors are characterized by infiltration of lymphocytes (tumor infiltrating lymphocytes, TIL), which may serve as a basis for new strategies to generate tumor specific lymphocytes. However, TIL frequently are unable to kill autologous tumor cells suggesting that they are anergic/tolerant. It is possible that the TIL are functional but the tumor cells are resistant to TIL-mediated apoptotic pathways. Previous findings revealed that resistant tumor cells can be sensitized with cytokines or subtoxic concentrations of chemotherapeutic drugs and restore killing by cytotoxic lymphocytes. In this study, we examined whether TIL can kill autologous and allogeneic tumor cells following sensitization with chemotherapeutic drugs. Renal and prostate cancer-derived TIL were cytotoxic to chemosensitized resistant tumor cells. Killing by TIL was found to be perforin-dependent and perforin-independent. These findings demonstrate that combination drug and immunotherapy may be able to overcome tumor cell resistance to killing by TIL. Further, in vivo sensitization of drug-resistant tumor cells by subtoxic doses of sensitizing chemotherapeutic drugs may result in tumor regression by the host immune system.


Subject(s)
Adenocarcinoma/immunology , Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/immunology , Carcinoma/immunology , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Interleukin-2/pharmacology , Kidney Neoplasms/immunology , Killer Cells, Lymphokine-Activated/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Ovarian Neoplasms/immunology , Prostatic Neoplasms/immunology , Adenocarcinoma/pathology , Apoptosis/drug effects , Apoptosis/physiology , Carcinoma/pathology , Carcinoma, Renal Cell/pathology , Combined Modality Therapy , Cytotoxicity, Immunologic/drug effects , Egtazic Acid/pharmacology , Fas Ligand Protein , Female , Humans , Ionomycin/pharmacology , Kidney Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/drug effects , Magnesium Chloride/pharmacology , Male , Membrane Glycoproteins/physiology , Ovarian Neoplasms/pathology , Perforin , Pore Forming Cytotoxic Proteins , Prostatic Neoplasms/pathology , Recombinant Proteins/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/immunology , fas Receptor/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...