Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
2.
ESMO Open ; 6(3): 100101, 2021 06.
Article in English | MEDLINE | ID: mdl-33901870

ABSTRACT

BACKGROUND: Among patients with advanced renal cell carcinoma (RCC), those with sarcomatoid histology (sRCC) have the poorest prognosis. This analysis assessed the efficacy of avelumab plus axitinib versus sunitinib in patients with treatment-naive advanced sRCC. METHODS: The randomized, open-label, multicenter, phase III JAVELIN Renal 101 trial (NCT02684006) enrolled patients with treatment-naive advanced RCC. Patients were randomized 1 : 1 to receive either avelumab plus axitinib or sunitinib following standard doses and schedules. Assessments in this post hoc analysis of patients with sRCC included efficacy (including progression-free survival) and biomarker analyses. RESULTS: A total of 108 patients had sarcomatoid histology and were included in this post hoc analysis; 47 patients in the avelumab plus axitinib arm and 61 in the sunitinib arm. Patients in the avelumab plus axitinib arm had improved progression-free survival [stratified hazard ratio, 0.57 (95% confidence interval, 0.325-1.003)] and a higher objective response rate (46.8% versus 21.3%; complete response in 4.3% versus 0%) versus those in the sunitinib arm. Correlative gene expression analyses of patients with sRCC showed enrichment of gene pathway scores for cancer-associated fibroblasts and regulatory T cells, CD274 and CD8A expression, and tumors with The Cancer Genome Atlas m3 classification. CONCLUSIONS: In this subgroup analysis of JAVELIN Renal 101, patients with sRCC in the avelumab plus axitinib arm had improved efficacy outcomes versus those in the sunitinib arm. Correlative analyses provide insight into this subtype of RCC and suggest that avelumab plus axitinib may increase the chance of overcoming the aggressive features of sRCC.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Axitinib , Carcinoma, Renal Cell , Kidney Neoplasms , Sunitinib , Antineoplastic Combined Chemotherapy Protocols , Axitinib/therapeutic use , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Sunitinib/therapeutic use
3.
Biotechniques ; 33(1): 190-2, 194, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12139245

ABSTRACT

The development of peptide-based therapeutics has suffered from challenges associated with delivery to intact tissue. In skin, an array of protein targets resides only tens of micrometers below the surface; however, because of difficulties in traversing the cutaneous barrier, the potentialfor peptide-based therapeutics remains unrealized. We have developed a general approach for topical peptide delivery into skin using releasable protein transduction sequences to enable peptide transport across tissue boundaries. Upon entry into the cell, the disulfide linkage between the peptide transduction sequences and peptide cargo is cleaved, permitting the dissociation of the highly charged peptide transduction sequences from the active peptide. A protype cargo peptide, the hemagglutinin (HA) epitope, was conjugated to a hepta-arginine protein transduction sequence via a releasable disulfide linkage. This construct penetrated the skin to deep dermis within 1 h after topical application. Consistent with the dissociation of the protein transduction and cargo sequences, absorbed protein transduction sequences and HA peptides displayed differential intracellular localization. Reversible protein transduction sequence linkage thus represents a noninvasive platform for tissue delivery of intact peptides with no requirement for viral vectors or parenteral injection and may be of broad utility in molecular therapy.


Subject(s)
Drug Delivery Systems/methods , Keratinocytes/metabolism , Protein Transport/genetics , Proteins/genetics , Proteins/pharmacokinetics , Signal Transduction/genetics , Administration, Cutaneous , Animals , Humans , Mice , Mice, Inbred BALB C , Mice, Nude
4.
Hum Gene Ther ; 12(12): 1551-8, 2001 Aug 10.
Article in English | MEDLINE | ID: mdl-11506697

ABSTRACT

The skin offers a tissue site accessible for delivery of gene-based therapeutics. To develop the capability for sustained systemic polypeptide delivery via cutaneous gene transfer, we generated and injected pseudotyped HIV-1 lentiviral vectors intradermally at a range of doses into human skin grafted on immune-deficient mice. Unlike Moloney murine leukemia virus (MLV)-based retrovectors, which failed to achieve detectable cutaneous gene transfer by this approach, lentivectors effectively targeted all major cell types within human skin tissue, including fibroblasts, endothelial cells, keratinocytes, and macrophages. After a single injection, lentivectors encoding human erythropoietin (EPO) produced dose-dependent increases in serum human EPO levels and hematocrit that increased rapidly within one month and remained stable subsequently. Delivered gene expression was confined locally at the injection site. Excision of engineered skin led to rapid and complete loss of human EPO in the bloodstream, confirming that systemic EPO delivery was entirely due to lentiviral targeting of cells within skin rather than via spread of the injected vector to visceral tissues. These findings indicate that the skin can sustain dosed systemic delivery of therapeutic polypeptides via direct lentivector injection and thus provide an accessible and reversible approach for gene-based delivery to the bloodstream.


Subject(s)
Gene Transfer Techniques , Lentivirus/genetics , Skin/metabolism , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium/metabolism , Erythropoietin/blood , Erythropoietin/genetics , Fibroblasts/metabolism , Genetic Vectors , Hematocrit , Humans , Keratinocytes/metabolism , Macrophages/metabolism , Mice , Mice, SCID , Peptides/genetics , Time Factors
5.
Hum Gene Ther ; 12(11): 1443-8, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11485635

ABSTRACT

Molecular therapy studies to date have examined only a limited number of corrective parameters. To assess more global impacts on cellular gene expression for two major molecular therapeutic approaches, we compared gene versus protein delivery in the human genetic disease junctional epidermolysis bullosa (JEB). Both gene and protein replacement of the laminin 5 beta3 (beta3) adhesion molecule restored normal growth and adhesion to poorly viable JEB cells. Gene expression profiling was then performed using cDNA microarrays. The expression of more genes was normalized after beta3 gene transfer than after protein transfer. As anticipated for beta3 delivery, many of the genes whose expression was restored to the normal range were those encoding adhesion molecules and hemidesmosome components. Although gene transfer normalized the expression of a higher percentage of genes than did protein transfer, neither approach fully normalized expression of all genes examined. In addition, both approaches disrupted the expression of some genes, but protein transfer altered expression of a larger proportion of the genes studied. Our findings suggest that therapeutic gene and protein delivery may exert different effects on gene expression and thus may have implications for the development and analysis of molecular therapies for the treatment of genetic disorders.


Subject(s)
Cell Adhesion Molecules/genetics , Epidermolysis Bullosa, Junctional/therapy , Antigens, CD/immunology , Base Sequence , Biopsy , Cell Adhesion Molecules/immunology , Cells, Cultured , Epidermolysis Bullosa, Junctional/genetics , Epidermolysis Bullosa, Junctional/pathology , Gene Expression Profiling , Gene Expression Regulation , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors/genetics , Green Fluorescent Proteins , Humans , Integrin beta3 , Integrins/immunology , Keratinocytes/metabolism , Keratinocytes/pathology , Kinetics , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Platelet Membrane Glycoproteins/immunology , Retroviridae/genetics , Transduction, Genetic , Kalinin
6.
Proc Natl Acad Sci U S A ; 98(9): 5193-8, 2001 Apr 24.
Article in English | MEDLINE | ID: mdl-11296269

ABSTRACT

The blistering disorder, lethal junctional epidermolysis bullosa (JEB), can result from mutations in the LAMB3 gene, which encodes laminin 5 beta3 (beta3). Appropriate expression of LAMbeta3 in JEB skin tissue could potentially ameliorate the symptoms of the underlying disease. To explore the utility of this therapeutic approach, primary keratinocytes from six unrelated JEB patients were transduced with a retroviral vector encoding beta3 and used to regenerate human skin on severe combined immunodeficient (SCID) mice. Tissue regenerated from beta3-transduced JEB keratinocytes produced phenotypically normal skin characterized by sustained beta3 expression and the formation of hemidesmosomes. Additionally, beta3 gene transfer corrected the distribution of a number of important basement membrane zone proteins including BPAG2, integrins beta4/beta1, and laminins alpha3/gamma2. Skin produced from beta3-negative (beta3[-]) JEB cells mimicked the hallmarks of the disease state and did not exhibit any of the aforementioned traits. Therefore, by effecting therapeutic gene transfer to beta3-deficient primary keratinocytes, it is possible to produce healthy, normal skin tissue in vivo. These data support the utility of gene therapy for JEB and highlight the potential for gene delivery in the treatment of human genetic skin disease.


Subject(s)
Cell Adhesion Molecules/metabolism , Epidermolysis Bullosa, Junctional/metabolism , Epidermolysis Bullosa, Junctional/pathology , Genetic Therapy , Basement Membrane/cytology , Basement Membrane/metabolism , Basement Membrane/pathology , Cell Adhesion Molecules/genetics , Cell Division , Cell Polarity , Cell Size , Cells, Cultured , Epidermolysis Bullosa, Junctional/genetics , Gene Transfer Techniques , Genetic Vectors/genetics , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Keratinocytes/pathology , Kinetics , Regeneration , Skin/cytology , Skin/metabolism , Skin/pathology , Stem Cells/cytology , Stem Cells/metabolism , Stem Cells/pathology , Kalinin
7.
Blood ; 94(10): 3349-57, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10552944

ABSTRACT

Retroviral vectors based on the Moloney murine leukemia virus (MoMuLV) are currently the most commonly used vehicles for stable gene transfer into mammalian hematopoietic cells. But, even with reasonable transduction efficiency, expression only occurs in a low percentage of transduced cells and decreases to undetectable levels over time. We have previously reported the modified MND LTR (myeloproliferative sarcoma virus enhancer, negative control region deleted, dl587rev primer-binding site substituted) to show increased expression frequency and decreased methylation in transduced murine embryonic stem cells and hematopoietic stem cells. We have now compared expression of the enhanced green fluorescent protein (eGFP) from a vector using the MoMuLV LTR (LeGFPSN) with that from the modified vector (MNDeGFPSN) in mature hematopoietic and lymphoid cells in the mouse bone marrow transplant (BMT) model. In primary BMT recipients, we observed a higher frequency of expression from the MND LTR (20% to 80%) in hematopoietic cells of all lineages in spleen, bone marrow, thymus, and blood compared with expression from the MoMuLV LTR (5% to 10%). Expression from the MND LTR reached 88% in thymic T lymphocytes and 54% in splenic B lymphocytes for up to 8 months after BMT. The mean fluorescence intensity of the individual cells, indicating the amount of protein synthesized, was 6- to 10-fold higher in cells expressing MNDeGFPSN compared with cells expressing LeGFPSN. Transduction efficiencies determined by DNA polymerase chain reaction of vector copy number were comparable for the 2 vectors. Therefore, the MND vector offers an improved vehicle for reliable gene expression in hematopoietic cells.


Subject(s)
Bone Marrow Transplantation , Gene Transfer Techniques , Genetic Vectors , Hematopoietic Stem Cells/metabolism , Lymphocytes/metabolism , 3T3 Cells , Animals , Female , Gene Dosage , Gene Expression , Genetic Therapy/methods , Genetic Vectors/genetics , Green Fluorescent Proteins , Leukemia Virus, Murine/genetics , Luminescent Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Retroviridae/genetics , Time Factors , Transduction, Genetic
8.
Hum Gene Ther ; 9(16): 2321-30, 1998 Nov 01.
Article in English | MEDLINE | ID: mdl-9829531

ABSTRACT

Retroviral vectors based on the Moloney murine leukemia virus (Mo-MuLV) are among the most commonly used vectors for stable gene transfer into mammalian cells. However, expression from the transcription unit of the Mo-MuLV long terminal repeat (LTR) has often been unsatisfactory. Transcriptional suppression of retroviral vectors in vitro in embryonal carcinoma (EC) cells and in vivo in hematopoietic stem cells (HSCs) has been associated with increased levels of cytosine methylation in the vector 5' LTR. To obtain a comprehensive picture of the methylation pattern in the 5' LTR of retroviral vectors, we employed the bisulfite genomic sequencing technique, which allows detection of the methylation pattern of every CpG dinucleotide in a target sequence. We studied the 5' LTR within the Mo-MuLV-based vector, LN, and a series of multiply modified vectors, which show improved expression in vitro and in vivo. Methylation patterns of the vectors were compared in PA317 (3T3-derived) fibroblasts, which are permissive for expression from all of the vectors, and in F9 embryonal carcinoma (EC) cells, which are restrictive for expression from the parental Mo-MuLV LTR but show improved expression from the modified vectors. These analyses revealed that the levels of methylation of CpG dinucleotides were globally consistent throughout the entire LTR, including the region of transcriptional factor binding. All vectors showed no measurable methylation of CpG dinucleotides throughout the 5' LTR in the PA317 fibroblasts. The CpG dinucleotides of the standard Mo-MuLV-based vector (LN) were highly methylated in F9 EC cells (49.1%). The doubly modified vector, MD-neo, which did not show improved expression, exhibited a relatively high level of methylation (45%), similar to that found in the LN vector. In contrast, the CpG dinucleotides of the triply modified vectors, which showed improved expression in EC cells (MND-neo and MTD-neo), were much less methylated (26.2 and 23.4%, respectively). The results extend our previous findings of an inverse correlation between gene expression and methylation of cytosine residues of the LTR of retroviral vectors.


Subject(s)
Cytosine/analysis , Cytosine/metabolism , Retroviridae/genetics , Sequence Analysis, DNA/methods , Terminal Repeat Sequences , Animals , Blotting, Northern , Carcinoma, Embryonal/virology , CpG Islands , DNA Methylation , DNA Primers , Gene Expression Regulation, Viral , Genetic Vectors , Mice , Moloney murine leukemia virus/genetics , Sulfites , Thy-1 Antigens/genetics , Tumor Cells, Cultured
9.
Proc Natl Acad Sci U S A ; 95(17): 10182-7, 1998 Aug 18.
Article in English | MEDLINE | ID: mdl-9707621

ABSTRACT

Retroviral vectors based on the Moloney murine leukemia virus (MoMuLV) have shown inconsistent levels and duration of expression as well as a propensity for the acquisition of de novo methylation in vivo. MoMuLV-based vectors are known to contain sequences that are capable of suppressing or preventing expression from the long terminal repeat. Previously, we constructed a series of modified retroviral vectors and showed that they function significantly better than MoMuLV-based vectors in vitro. To test the efficacy of the modified vectors in hematopoietic stem cells in vivo, we examined gene expression and proviral methylation in differentiated hematopoietic colonies formed in the spleens of mice after serial transplantation with transduced bone marrow (2 degreesCFU-S). We found a significant increase in the frequency of expression with our modified vectors (>90% expression in vector DNA containing 2 degreesCFU-S) over the frequency observed with the standard MoMuLV-based vector (28% expression in vector containing 2 degreesCFU-S). Expression from the modified vectors was highly consistent, with expression in >50% of the vector-containing 2 degreesCFU-S from all 20 transplant recipients analyzed, whereas expression from the standard MoMuLV-based vector was inconsistent, with expression in 0-10% of the vector containing 2 degreesCFU-S from 8 recipients and expression in >50% of the vector-containing 2 degreesCFU-S from 4 other recipients. In addition, we established that the modified vectors had a lower level of DNA methylation than the control vector. These findings represent significant advances in the development and evaluation of effective retroviral vectors for application in vivo.


Subject(s)
Genetic Vectors , Hematopoietic Stem Cells/metabolism , Retroviridae/genetics , Animals , Base Sequence , Bone Marrow Transplantation , Colony-Forming Units Assay , DNA Methylation , DNA Primers/genetics , DNA, Recombinant/genetics , DNA, Recombinant/metabolism , Female , Founder Effect , Gene Expression , Gene Transfer Techniques , Hematopoietic Stem Cells/cytology , Male , Mice , Mice, Inbred C57BL , Moloney murine leukemia virus/genetics , Repetitive Sequences, Nucleic Acid , Transduction, Genetic , Transplantation, Isogeneic , Virus Integration/genetics
10.
J Virol ; 71(12): 9466-74, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9371608

ABSTRACT

Gene expression from the Moloney murine leukemia retrovirus (Mo-MuLV) is highly restricted in embryonic carcinoma (EC) and embryonic stem (ES) cells. We compared levels of expression in PA317 fibroblasts, F9 (EC) cells, and CCE (ES) cells by Mo-MuLV-based vectors and vectors based on our previously reported MND backbone, which has alterations to address three viral elements implicated as repressors of expression by Mo-MuLV: the enhancer, the primer binding site, and the negative-control region. Expression was evaluated with three reporter genes, the chloramphenicol acetyltransferase (CAT) gene, whose expression was measured by enzymatic assay and by Northern blotting; a truncated nerve growth factor receptor (tNGFR), whose expression was measured by fluorescence-activated cell sorting (FACS) as a cell surface protein; and the enhanced green fluorescent protein (EGFP), whose expression was measured intracellularly by flow cytometry. We found significantly higher levels of CAT activity (5- to 300-fold) and greater quantities of vector-specific transcripts in ES and EC cells transduced with the modified MND-CAT-SN vector than in those transduced with L-CAT-SN. Northern blot analysis indicated that long terminal repeat transcripts from MND-CAT-SN are >80 times more abundant than the L-CAT-SN transcripts. FACS analysis of tNGFR expression from a pair of vectors, L-tNGFR-SN and MND-tNGFR-SN, indicated that only 1.04% of the CCE cells containing the L-tNGFR-SN vector expressed the cell surface reporter, while the MND-tNGFR-SN vector drove expression in 99.54% of the CCE cells. Of the F9 cells containing the L-tNGFR-SN vector, 13.32% expressed tNGFR, while 99.89% of the F9 cells transduced with MND-tNGFR-SN showed expression. Essentially identical results were produced with an analogous pair of vectors encoding EGFP. In unselected pools of F9 cells 48 h posttransduction, the L-EGFP-SN vector drove expression in only 5% of the population while the MND-EGFP-SN vector drove expression in 88% of the cells. After more than 3 weeks in culture without selection, the proportion of cells showing expression from L-EGFP-SN decreased slightly to 3% while expression from the MND-EGFP-SN vector persisted in 80% of the cells. Interestingly, in the few ES and EC cells which did show expression from the L-tNGFR-SN or L-EGFP-SN vectors, the magnitude of reporter expression was similar to that from the MND-tNGFR-SN or MND-EGFP-SN vector in nearly all cells, suggesting that the MND vectors are far less susceptible to position-dependent variegation of expression than are the Mo-MuLV-based vectors. Therefore, the modified retroviral vector, MND, achieves higher net levels of expression due to a greater frequency of expression, which may be useful for the expression of exogenous genes in EC and ES cells.


Subject(s)
Gene Expression , Genetic Vectors , Moloney murine leukemia virus , 3T3 Cells , Animals , Blotting, Northern , Blotting, Southern , Carcinoma, Embryonal , Cell Line , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , DNA Methylation , Genes, Reporter , Green Fluorescent Proteins , Luminescent Proteins/genetics , Mice , Proviruses , Receptors, Nerve Growth Factor/genetics , Retroviridae , Stem Cells , Tumor Cells, Cultured , Virus Integration
11.
J Immunol ; 147(9): 2892-901, 1991 Nov 01.
Article in English | MEDLINE | ID: mdl-1918997

ABSTRACT

Results and conclusions concerning the ability of HIV glycoprotein (gp) 120 to stimulate monokine secretion have been equivocal, based on observations using natural gp120 derived from infected human cells and a Chinese hamster ovary (CHO) cell-derived recombinant fusion protein. Current studies were designed to determine whether differences in recombinant gp120 proteins could result in failure to trigger monokine production. We found that natural gp120 could stimulate monocytes to release TNF-alpha, IL-1 beta, IL-6, and granulocyte-macrophage-CSF, and this effect could be blocked with soluble CD4. Full-length rgp120 either expressed from an adenovirus vector and purified from infected human cells, or derived from CHO cells, could function similarly. In contrast, full-length recombinant envelope protein expressed in a baculovirus system and a CHO cell-derived recombinant fusion protein tested previously, consistently failed to stimulate monokine production. The stimulatory capacity of both natural and full-length CHO cell-derived gp120 was eliminated by heating at 100 degrees C, and could be blocked with excess CHO cell-derived gp120 fusion protein. Inasmuch as the baculovirus-expressed gp120 and the CHO cell-derived recombinant fusion protein can bind to CD4, these results suggest that HIV gp120 binding to CD4 on the monocyte surface may of itself be insufficient for stimulation of monokine secretion. Therefore, primary protein structure, as well as posttranslational protein modifications, may determine this activity.


Subject(s)
HIV Envelope Protein gp120/immunology , HIV-1/immunology , Macrophages/physiology , Monocytes/physiology , Monokines/metabolism , Animals , CHO Cells , Cricetinae , Endotoxins/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , HIV Core Protein p24/immunology , HIV Envelope Protein gp120/chemistry , Humans , In Vitro Techniques , Interleukin-1/metabolism , Interleukin-6/metabolism , Lipopolysaccharides , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/metabolism
12.
J Immunol ; 143(2): 470-5, 1989 Jul 15.
Article in English | MEDLINE | ID: mdl-2544645

ABSTRACT

We have previously described model systems for cytokine-induced regulation of chronically HIV-infected promonocyte and T cell clones. Using these systems, we have shown that monokines contained in supernatants from LPS-stimulated human monocyte/macrophages (MO) up-regulate HIV expression, reflected by an increase in reverse transcriptase activity, viral RNA levels, and expressed viral proteins. Current studies were designed to determine whether viral Ag can interact with MO and secondarily affect HIV1 expression by stimulating monokine production. We found that certain herpes-group viruses, including CMV and EBV, augment HIV1 expression by inducing monokine production, whereas others, such as HSV1, HSV2, varicella-zoster virus, and human herpes virus 6 were unable to function in this capacity. The HSV1 and HSV2 Ag which failed to stimulate monokine production did not interfere with MO stimulation by CMV Ag, suggesting that failure to induce HIV expression was not attributable to MO suppression. When nonherpes group viruses were tested, we found that human adenovirus, hepatitis B virus, and vaccinia virus all failed to stimulate the production of monokines capable of activating HIV in the chronically infected cell lines. In contrast, HIV1 can augment its own expression by inducing the secretion of monokines which up-regulate HIV expression in the infected cells. The viral Ag-induced MO supernatants capable of up-regulating HIV expression did so in a dose-dependent manner, whereas viral Ag alone produced no significant change. Monokine production mediated by viral Ag was not attributable to contaminating endotoxin. These studies provide a model to determine whether other opportunistic infections may induce the expression of HIV by indirect mechanisms, such as the stimulation of cytokine production.


Subject(s)
Antigens, Viral/immunology , Biological Factors/biosynthesis , HIV-1/physiology , Monocytes/metabolism , Virus Activation , Acquired Immunodeficiency Syndrome/immunology , Adenoviruses, Human/immunology , Biological Factors/physiology , Cell Line , Endotoxins/physiology , HIV Antigens/immunology , HIV-1/immunology , Hepatitis B virus/immunology , Humans , Monocytes/immunology , Monokines , Simplexvirus/immunology , Vaccinia virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...