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










Publication year range
1.
Nature ; 547(7662): 222-226, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28678784

ABSTRACT

T cells directed against mutant neo-epitopes drive cancer immunity. However, spontaneous immune recognition of mutations is inefficient. We recently introduced the concept of individualized mutanome vaccines and implemented an RNA-based poly-neo-epitope approach to mobilize immunity against a spectrum of cancer mutations. Here we report the first-in-human application of this concept in melanoma. We set up a process comprising comprehensive identification of individual mutations, computational prediction of neo-epitopes, and design and manufacturing of a vaccine unique for each patient. All patients developed T cell responses against multiple vaccine neo-epitopes at up to high single-digit percentages. Vaccine-induced T cell infiltration and neo-epitope-specific killing of autologous tumour cells were shown in post-vaccination resected metastases from two patients. The cumulative rate of metastatic events was highly significantly reduced after the start of vaccination, resulting in a sustained progression-free survival. Two of the five patients with metastatic disease experienced vaccine-related objective responses. One of these patients had a late relapse owing to outgrowth of ß2-microglobulin-deficient melanoma cells as an acquired resistance mechanism. A third patient developed a complete response to vaccination in combination with PD-1 blockade therapy. Our study demonstrates that individual mutations can be exploited, thereby opening a path to personalized immunotherapy for patients with cancer.


Subject(s)
Cancer Vaccines/genetics , Cancer Vaccines/immunology , Melanoma/immunology , Melanoma/therapy , Mutation/genetics , Precision Medicine/methods , RNA/genetics , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , B7-H1 Antigen/immunology , CD8 Antigens/immunology , Cancer Vaccines/therapeutic use , Epitopes/genetics , Epitopes/immunology , Humans , Immunotherapy/methods , Melanoma/genetics , Neoplasm Metastasis , Neoplasm Recurrence, Local/prevention & control , Nivolumab , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/immunology , Vaccination , beta 2-Microglobulin/deficiency
2.
Histochem Cell Biol ; 148(3): 313-329, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28397143

ABSTRACT

Lentiviruses are suitable to transfer potential therapeutic genes into non-replicating cells such as neurons, but systematic in vivo studies on transduction of neural cells within the complete brain are missing. We analysed the distribution of transduced cells with respect to brain structure, virus tropism, numbers of transduced neurons per brain, and influence of the Vpx or Vpr accessory proteins after injection of vectors based on SIVsmmPBj, HIV-2, and HIV-1 lentiviruses into the right striatum of the mouse brain. Transduced cells were found ipsilaterally around the injection canal, in corpus striatum and along corpus callosum, irrespective of the vector type. All vectors except HIV-2SEW transduced also single cells in the olfactory bulb, hippocampus, and cerebellum. Vector HIV-2SEW was the most neuron specific. However, vectors PBjSEW and HIV-1SEW transduced more neurons per brain (means 41,299 and 32,309) than HIV-2SEW (16,102). In the presence of Vpx/Vpr proteins, HIV-2SEW(Vpx) and HIV-1SEW(Vpr) showed higher overall transduction efficiencies (30,696 and 27,947 neurons per brain) than PBjSEW(Vpx) (6636). The distances of transduced cells from the injection canal did not differ among the viruses but correlated positively with the numbers of transduced neurons. The presence of Vpx/Vpr did not increase the numbers of transduced neurons. Parental virus type and the vector equipment seem to influence cellular tropism and transduction efficiency. Thus, precision of injection and choice of virus pseudotype are not sufficient when targeted lentiviral vector transduction of a defined brain cell population is required.


Subject(s)
Brain/virology , Genetic Vectors/pharmacokinetics , HIV-1/metabolism , HIV-2/metabolism , Lentivirus/genetics , Simian Immunodeficiency Virus/metabolism , Transduction, Genetic/methods , Viral Tropism , Animals , Brain/metabolism , Cells, Cultured , Female , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , HIV-1/genetics , HIV-2/genetics , Lentivirus/metabolism , Mice , Mice, Inbred C57BL , Pregnancy , Qualitative Research , Simian Immunodeficiency Virus/genetics
3.
Retrovirology ; 13(1): 46, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27368163

ABSTRACT

BACKGROUND: Feline immunodeficiency virus (FIV) is a global pathogen of Felidae species and a model system for Human immunodeficiency virus (HIV)-induced AIDS. In felids such as the domestic cat (Felis catus), APOBEC3 (A3) genes encode for single-domain A3Z2s, A3Z3 and double-domain A3Z2Z3 anti-viral cytidine deaminases. The feline A3Z2Z3 is expressed following read-through transcription and alternative splicing, introducing a previously untranslated exon in frame, encoding a domain insertion called linker. Only A3Z3 and A3Z2Z3 inhibit Vif-deficient FIV. Feline A3s also are restriction factors for HIV and Simian immunodeficiency viruses (SIV). Surprisingly, HIV-2/SIV Vifs can counteract feline A3Z2Z3. RESULTS: To identify residues in feline A3s that Vifs need for interaction and degradation, chimeric human-feline A3s were tested. Here we describe the molecular direct interaction of feline A3s with Vif proteins from cat FIV and present the first structural A3 model locating these interaction regions. In the Z3 domain we have identified residues involved in binding of FIV Vif, and their mutation blocked Vif-induced A3Z3 degradation. We further identified additional essential residues for FIV Vif interaction in the A3Z2 domain, allowing the generation of FIV Vif resistant A3Z2Z3. Mutated feline A3s also showed resistance to the Vif of a lion-specific FIV, indicating an evolutionary conserved Vif-A3 binding. Comparative modelling of feline A3Z2Z3 suggests that the residues interacting with FIV Vif have, unlike Vif-interacting residues in human A3s, a unique location at the domain interface of Z2 and Z3 and that the linker forms a homeobox-like domain protruding of the Z2Z3 core. HIV-2/SIV Vifs efficiently degrade feline A3Z2Z3 by possible targeting the linker stretch connecting both Z-domains. CONCLUSIONS: Here we identified in feline A3s residues important for binding of FIV Vif and a unique protein domain insertion (linker). To understand Vif evolution, a structural model of the feline A3 was developed. Our results show that HIV Vif binds human A3s differently than FIV Vif feline A3s. The linker insertion is suggested to form a homeo-box domain, which is unique to A3s of cats and related species, and not found in human and mouse A3s. Together, these findings indicate a specific and different A3 evolution in cats and human.


Subject(s)
Cytidine Deaminase/chemistry , Cytidine Deaminase/metabolism , Gene Products, vif/metabolism , HIV-1/metabolism , Immunodeficiency Virus, Feline/metabolism , Animals , Cats , Cell Line , Cytidine Deaminase/genetics , Evolution, Molecular , Gene Products, vif/genetics , Genes, Homeobox , HIV-1/genetics , Humans , Immunodeficiency Virus, Feline/genetics , Models, Molecular , Recombinant Fusion Proteins/metabolism
8.
Cancer Immunol Immunother ; 61(9): 1473-84, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22331080

ABSTRACT

Targeted delivery of tumor-associated antigens to professional antigen-presenting cells (APC) is being explored as a strategy to enhance the antitumoral activity of cancer vaccines. Here, we generated a cell-based system for continuous in vivo production of a CTLA-4-ErbB2 fusion protein as a therapeutic vaccine. The chimeric CTLA-4-ErbB2 molecule contains the extracellular domain of CTLA-4 for specific targeting to costimulatory B7 molecules on the surface of APC, genetically fused to residues 1-222 of human ErbB2 (HER2) as an antigenic determinant. In wild-type BALB/c mice, inoculation of syngeneic epithelial cells continuously secreting the CTLA-4-ErbB2 fusion vaccine in the vicinity of subcutaneously growing ErbB2-expressing renal cell carcinomas resulted in the rejection of established tumors, accompanied by the induction of ErbB2-specific antibodies and cytotoxic T cells. In contrast, treatment with CTLA-4-ErbB2 vaccine-secreting producer cells alone was insufficient to induce tumor rejection in ErbB2-transgenic WAP-Her-2 F1 mice, which are characterized by pronounced immunological tolerance to the human self-antigen. When CTLA-4-ErbB2 producer cells were modified to additionally secrete interleukin (IL)-15, antigen-specific antitumoral activity of the vaccine in WAP-Her-2 F1 mice was restored, documented by an increase in survival, and marked inhibition of the growth of established ErbB2-expressing, but not antigen-negative tumors. Our results demonstrate that continuous in vivo expression of an APC-targeted ErbB2 fusion protein results in antigen-specific immune responses and antitumoral activity in tumor-bearing hosts, which is augmented by the pleiotropic cytokine IL-15. This provides a rationale for further development of this approach for specific cancer immunotherapy.


Subject(s)
Antigen-Presenting Cells/immunology , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Interleukin-15/immunology , Receptor, ErbB-2/immunology , Animals , Antigen-Presenting Cells/metabolism , CTLA-4 Antigen/biosynthesis , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Cancer Vaccines/genetics , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/therapy , Female , Humans , Immunotherapy, Active , Kidney Neoplasms/immunology , Kidney Neoplasms/therapy , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology
9.
Mol Biotechnol ; 47(3): 262-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20938757

ABSTRACT

Lentiviral gene transfer vectors are suitable for genetically modifying non-cycling primary human cells. In this study, we analyzed transduced human dendritic cells (DC) generated by the use of three different GFP-encoding lentiviral vectors, HIV-2 ROD A Δenv-GFP (ROD A), SIVsmm PBj ΔE EGFP (PBj), and SIVmac ΔE EGFP (SIVmac). CD14+ monocytes were isolated from buffy coat, transduced, and differentiated to immature and mature DC. Cytofluometric analysis of DC revealed high transduction efficiencies at MOI 1 for simian immunodeficiency virus (SIV)-derived vectors PBj and SIVmac ranging between 80-90 and 70-90%, respectively. In contrast, transduction with ROD A resulted only in approximately 30%-positive DC at the same MOI. Of note, none of the analyzed vectors affected expression of maturation and/or activation markers. Moreover, transduction with PBj or SIVmac did not induce significant cytokine responses whereas ROD A transduction stimulated weak interferon-alpha responses. SIVmac transduced DC showed normal phagocytosis of antigen and normal allo T cell stimulatory capacity when compared with untreated DC. Thus, the SIVmac lentiviral transduction vector is suitable for efficient genetic modification of human DC without affecting phenotype or function and thus qualifies this vector as a versatile tool for use in basic research.


Subject(s)
Dendritic Cells/metabolism , Genetic Vectors/genetics , Lentivirus/genetics , Cells, Cultured , Flow Cytometry , Humans , Phagocytosis/genetics , Phagocytosis/physiology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Transduction, Genetic
10.
J Gene Med ; 12(5): 446-52, 2010 May.
Article in English | MEDLINE | ID: mdl-20440755

ABSTRACT

BACKGROUND: Lentiviral vectors allow stable gene transfer into nonreplicating cells and are increasingly used in clinical gene therapy approaches. Vectors derived from different origins can show distinct target cell transduction properties. Therefore, the construction of modern vector systems of different viral origin remains desirable. The generation of safe and efficient lentivirus-derived transfer vectors by gradual enhancing cloning steps is a time-consuming process that depends on the presence of suitable restriction sites. Multiple-step cloning protocols also enhance the risk of acquisition of mutations or other genetic instabilities. METHODS: We constructed novel HIV-2 and SIVsmmPBj-derived transfer vectors by amplification of three essential segments of the viral genome [5'-long terminal repeat (LTR), rev responsive element, DeltaU3-3'-LTR] on the template of the lentiviral full-length genome by a highly flexible three-step fusion polymerase chain reaction approach. Further necessary vector elements, as well as a multiple cloning site, were included into the resulting vector by extension of the primer sequences. The respective vesicular stomatitis virus G pseudotyped lentiviral vector particles were generated and analysed. RESULTS: Two novel transfer vectors of different lentiviral origin were successfully generated. Titers for the corresponding SIVsmmPBj- and HIV-2-derived vectors reached up to 9.9 x 10(7) transforming units (TU)/ml and 1.2 x 10(8) TU/ml, respectively. The specific capacity to transduce primary human monocytes was maintained in both newly-generated vector systems. CONCLUSIONS: We anticipate that this novel and fast way of generating any lentiviral transfer vector will improve the generation of such vectors. The HIV-2- and SIVsmmPBj-derived vectors described will prove valuable for future gene therapy strategies.


Subject(s)
Genetic Vectors/genetics , HIV-2/genetics , Polymerase Chain Reaction/methods , Simian Immunodeficiency Virus/genetics , Cell Line , Cloning, Molecular , HIV-2/physiology , Humans , Monocytes/metabolism , Plasmids/genetics , Simian Immunodeficiency Virus/physiology , Transduction, Genetic , Virus Replication/physiology
11.
PLoS One ; 4(9): e7098, 2009 Sep 21.
Article in English | MEDLINE | ID: mdl-19768115

ABSTRACT

BACKGROUND: Human primary monocytes are refractory to infection with the human immunodeficiency virus 1 (HIV-1) or transduction with HIV-1-derived vectors. In contrast, efficient single round transduction of monocytes is mediated by vectors derived from simian immunodeficiency virus of sooty mangabeys (SIVsmmPBj), depending on the presence of the viral accessory protein Vpx. METHODS AND FINDINGS: Here we analyzed whether Vpx of SIVsmmPBj is sufficient for transduction of primary monocytes by HIV-1-derived vectors. To enable incorporation of PBj Vpx into HIV-1 vector particles, a HA-Vpr/Vpx fusion protein was generated. Supplementation of HIV-1 vector particles with this fusion protein was not sufficient to facilitate transduction of human monocytes. However, monocyte transduction with HIV-1-derived vectors was significantly enhanced after delivery of Vpx proteins by virus-like particles (VLPs) derived from SIVsmmPBj. Moreover, pre-incubation with Vpx-containing VLPs restored replication capacity of infectious HIV-1 in human monocytes. In monocytes of non-human primates, single-round transduction with HIV-1 vectors was enabled. CONCLUSION: Vpx enhances transduction of primary human and even non-human monocytes with HIV-1-derived vectors, only if delivered in the background of SIVsmmPBj-derived virus-like particles. Thus, for accurate Vpx function the presence of SIVsmmPBj capsid proteins might be required. Vpx is essential to overcome a block of early infection steps in primary monocytes.


Subject(s)
HIV-1/genetics , HIV-1/metabolism , Monocytes/virology , Viral Regulatory and Accessory Proteins/metabolism , Virus Replication , Animals , Blotting, Western , Cercocebus , Genetic Vectors , HeLa Cells , Humans , Models, Genetic , Monocytes/metabolism , Simian Immunodeficiency Virus/metabolism , vpr Gene Products, Human Immunodeficiency Virus/genetics , vpr Gene Products, Human Immunodeficiency Virus/metabolism
12.
J Biol Chem ; 284(9): 5819-26, 2009 Feb 27.
Article in English | MEDLINE | ID: mdl-19074429

ABSTRACT

The APOBEC3 cytidine deaminases are part of the intrinsic defense of cells against retroviruses. Lentiviruses and spumaviruses have evolved essential accessory proteins, Vif and Bet, respectively, which counteract the APOBEC3 proteins. We show here that Bet of the Prototype foamy virus inhibits the antiviral APOBEC3C activity by a mechanism distinct to Vif: Bet forms a complex with APOBEC3C without inducing its degradation. Bet abolished APOBEC3C dimerization as shown by coimmunoprecipitation and cross-linking experiments. These findings implicate a physical interaction between Bet and the APOBEC3C. Subsequently, we identified the Bet interaction domain in human APOBEC3C in the predicted APOBEC3C dimerization site. Taken together, these data support the hypothesis that Bet inhibits incorporation of APOBEC3Cs into retroviral particles. Bet likely achieves this by trapping APOBEC3C protein in complexes rendering them unavailable for newly generated viruses due to direct immobilization.


Subject(s)
Cytidine Deaminase/antagonists & inhibitors , Cytidine Deaminase/metabolism , Proviruses/genetics , Retroviridae Proteins/physiology , APOBEC-3G Deaminase , Animals , Cells, Cultured , Chlorocebus aethiops , Cricetinae , Cross-Linking Reagents , Dimerization , Gene Products, vif/physiology , Humans , Immunoblotting , Immunoprecipitation , Kidney/cytology , Kidney/metabolism , Lentivirus/genetics , Lentivirus/metabolism , Macaca mulatta , Macaca nemestrina , Pan troglodytes , Simian foamy virus , Transfection , Virus Assembly , Virus Replication/drug effects , Virus Replication/genetics
13.
Virology ; 364(2): 330-41, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17418360

ABSTRACT

Although lentiviruses like HIV-1 are able to infect non-dividing cells, particular resting cells such as non-stimulated primary peripheral blood mononuclear cells (PBMC) are resistant to infection. In contrast to other lentiviruses, SIVsmmPBj can replicate in non-stimulated PBMC. Moreover, SIVsmmPBj-derived, but not HIV-1-derived, replication-incompetent vectors enable gene transfer into G(0)-arrested human cell lines and primary human monocytes. Here, we demonstrate that transduction of G(0)-arrested cell lines by SIVsmmPBj-derived vectors is independent of the viral accessory proteins Vif, Vpx, Vpr, or Nef. In contrast, for the transduction of primary human monocytes, the Vpx protein proved to be essential. However, trans-complementation of HIV-1 vectors with SIVsmmPBj Vpx did not provide the property of gene transfer into monocytes. Taken together, these data indicate that Vpx is essential for the infection of primary monocytes by SIVsmmPBj. Additionally, further genome functions besides the accessory proteins are required for the particular capacity of SIVsmmPBj in transduction or infection events.


Subject(s)
Retroviridae Proteins/physiology , Simian Immunodeficiency Virus/physiology , Simian Immunodeficiency Virus/pathogenicity , Viral Regulatory and Accessory Proteins/physiology , Base Sequence , Cell Line , DNA, Viral/genetics , Gene Deletion , Genes, Viral , Genetic Complementation Test , Genetic Vectors , HIV/genetics , Humans , Monocytes/virology , Resting Phase, Cell Cycle , Retroviridae Proteins/genetics , Simian Immunodeficiency Virus/genetics , Transduction, Genetic , Viral Regulatory and Accessory Proteins/genetics , Virus Replication
14.
J Virol ; 81(13): 7048-60, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17459941

ABSTRACT

The productive replication of human immunodeficiency virus type 1 (HIV-1) occurs exclusively in defined cells of human or chimpanzee origin, explaining why heterologous animal models for HIV replication, pathogenesis, vaccination, and therapy are not available. This lack of an animal model for HIV-1 studies prompted us to examine the susceptibility of feline cells in order to evaluate the cat (Felis catus) as an animal model for studying HIV-1. Here, we report that feline cell lines harbor multiple restrictions with respect to HIV-1 replication. The feline CD4 receptor does not permit virus infection. Feline T-cell lines MYA-1 and FeT-1C showed postentry restrictions resulting in low HIV-1 luciferase reporter activity and low expression of viral Gag-Pol proteins when pseudotyped vectors were used. Feline fibroblastic CrFK and KE-R cells, expressing human CD4 and CCR5, were very permissive for viral entry and HIV-long terminal repeat-driven expression but failed to support spreading infection. KE-R cells displayed a profound block with respect to release of HIV-1 particles. In contrast, CrFK cells allowed very efficient particle production; however, the CrFK cell-derived HIV-1 particles had low specific infectivity. We subsequently identified feline apolipoprotein B-editing catalytic polypeptide 3 (feAPOBEC3) proteins as active inhibitors of HIV-1 particle infectivity. CrFK cells express at least three different APOBEC3s: APOBEC3C, APOBEC3H, and APOBEC3CH. While the feAPOBEC3C did not significantly inhibit HIV-1, the feAPOBEC3H and feAPOBEC3CH induced G to A hypermutations of the viral cDNA and reduced the infectivity approximately 10- to approximately 40-fold.


Subject(s)
Disease Models, Animal , HIV Infections/metabolism , HIV-1/metabolism , Virus Replication , Animals , Base Sequence , CD4 Antigens/metabolism , Cats , Cell Line , Cytosine Deaminase/metabolism , Evaluation Studies as Topic , Fusion Proteins, gag-pol/biosynthesis , Humans , Molecular Sequence Data , Pan troglodytes , Receptors, CCR5/metabolism , Species Specificity , Terminal Repeat Sequences
SELECTION OF CITATIONS
SEARCH DETAIL
...