Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Genome Biol ; 25(1): 25, 2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38243310

ABSTRACT

CRISPR genome editing approaches theoretically enable researchers to define the function of each human gene in specific cell types, but challenges remain to efficiently perform genetic perturbations in relevant models. In this work, we develop a library cloning protocol that increases sgRNA uniformity and greatly reduces bias in existing genome-wide libraries. We demonstrate that our libraries can achieve equivalent or better statistical power compared to previously reported screens using an order of magnitude fewer cells. This improved cloning protocol enables genome-scale CRISPR screens in technically challenging cell models and screen formats.


Subject(s)
CRISPR-Cas Systems , RNA, Guide, CRISPR-Cas Systems , Humans , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Library , Gene Editing , Cloning, Molecular
2.
Genome Biol ; 24(1): 281, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-38062486

ABSTRACT

GCLiPP is a global RNA interactome capture method that detects RNA-binding protein (RBP) occupancy transcriptome-wide. GCLiPP maps RBP-occupied sites at a higher resolution than phase separation-based techniques. GCLiPP sequence tags correspond with known RBP binding sites and are enriched for sites detected by RBP-specific crosslinking immunoprecipitation (CLIP) for abundant cytosolic RBPs. Comparison of human Jurkat T cells and mouse primary T cells uncovers shared peaks of GCLiPP signal across homologous regions of human and mouse 3' UTRs, including a conserved mRNA-destabilizing cis-regulatory element. GCLiPP signal overlapping with immune-related SNPs uncovers stabilizing cis-regulatory regions in CD5, STAT6, and IKZF1.


Subject(s)
RNA-Binding Proteins , Transcriptome , Animals , Humans , Mice , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Binding Sites/genetics , RNA/metabolism , Protein Binding , Immunoprecipitation
3.
Nature ; 615(7953): 687-696, 2023 03.
Article in English | MEDLINE | ID: mdl-36356599

ABSTRACT

T cell receptors (TCRs) enable T cells to specifically recognize mutations in cancer cells1-3. Here we developed a clinical-grade approach based on CRISPR-Cas9 non-viral precision genome-editing to simultaneously knockout the two endogenous TCR genes TRAC (which encodes TCRα) and TRBC (which encodes TCRß). We also inserted into the TRAC locus two chains of a neoantigen-specific TCR (neoTCR) isolated from circulating T cells of patients. The neoTCRs were isolated using a personalized library of soluble predicted neoantigen-HLA capture reagents. Sixteen patients with different refractory solid cancers received up to three distinct neoTCR transgenic cell products. Each product expressed a patient-specific neoTCR and was administered in a cell-dose-escalation, first-in-human phase I clinical trial ( NCT03970382 ). One patient had grade 1 cytokine release syndrome and one patient had grade 3 encephalitis. All participants had the expected side effects from the lymphodepleting chemotherapy. Five patients had stable disease and the other eleven had disease progression as the best response on the therapy. neoTCR transgenic T cells were detected in tumour biopsy samples after infusion at frequencies higher than the native TCRs before infusion. This study demonstrates the feasibility of isolating and cloning multiple TCRs that recognize mutational neoantigens. Moreover, simultaneous knockout of the endogenous TCR and knock-in of neoTCRs using single-step, non-viral precision genome-editing are achieved. The manufacture of neoTCR engineered T cells at clinical grade, the safety of infusing up to three gene-edited neoTCR T cell products and the ability of the transgenic T cells to traffic to the tumours of patients are also demonstrated.


Subject(s)
Cell- and Tissue-Based Therapy , Gene Editing , Neoplasms , Precision Medicine , Receptors, Antigen, T-Cell , T-Lymphocytes , Transgenes , Humans , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Biopsy , Cell- and Tissue-Based Therapy/adverse effects , Cell- and Tissue-Based Therapy/methods , Cytokine Release Syndrome/complications , Disease Progression , Encephalitis/complications , Gene Knock-In Techniques , Gene Knockout Techniques , Genes, T-Cell Receptor alpha , Genes, T-Cell Receptor beta , Mutation , Neoplasms/complications , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/therapy , Patient Safety , Precision Medicine/adverse effects , Precision Medicine/methods , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transgenes/genetics , HLA Antigens/immunology , CRISPR-Cas Systems
4.
J Exp Med ; 218(11)2021 11 01.
Article in English | MEDLINE | ID: mdl-34586363

ABSTRACT

MicroRNAs (miRNAs, miRs) regulate cell fate decisions by post-transcriptionally tuning networks of mRNA targets. We used miRNA-directed pathway discovery to reveal a regulatory circuit that influences Ig class switch recombination (CSR). We developed a system to deplete mature, activated B cells of miRNAs, and performed a rescue screen that identified the miR-221/222 family as a positive regulator of CSR. Endogenous miR-221/222 regulated B cell CSR to IgE and IgG1 in vitro, and miR-221/222-deficient mice exhibited defective IgE production in allergic airway challenge and polyclonal B cell activation models in vivo. We combined comparative Ago2-HITS-CLIP and gene expression analyses to identify mRNAs bound and regulated by miR-221/222 in primary B cells. Interrogation of these putative direct targets uncovered functionally relevant downstream genes. Genetic depletion or pharmacological inhibition of Foxp1 and Arid1a confirmed their roles as key modulators of CSR to IgE and IgG1.


Subject(s)
Immunoglobulin Class Switching/genetics , MicroRNAs/genetics , Recombination, Genetic/genetics , Animals , B-Lymphocytes/immunology , Female , Gene Expression/genetics , Gene Expression/immunology , Gene Regulatory Networks/genetics , Hypersensitivity/genetics , Hypersensitivity/immunology , Immunoglobulin Class Switching/immunology , Immunoglobulin E/genetics , Immunoglobulin G/genetics , Male , Mice , MicroRNAs/immunology , Recombination, Genetic/immunology
5.
Cell Rep ; 28(11): 2795-2806.e3, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31509743

ABSTRACT

The 3' UTR (UTR) of human mRNAs plays a critical role in controlling protein expression and function. Importantly, 3' UTRs of human messages are not invariant for each gene but rather are shaped by alternative polyadenylation (APA) in a cell state-dependent manner, including in response to T cell activation. However, the proteins and mechanisms driving APA regulation remain poorly understood. Here we show that the RNA-binding protein CELF2 controls APA of its own message in a signal-dependent manner by competing with core enhancers of the polyadenylation machinery for binding to RNA. We further show that CELF2 binding overlaps with APA enhancers transcriptome-wide, and almost half of 3' UTRs that undergo T cell signaling-induced APA are regulated in a CELF2-dependent manner. These studies thus reveal CELF2 to be a critical regulator of 3' UTR identity in T cells and demonstrate an additional mechanism for CELF2 in regulating polyadenylation site choice.


Subject(s)
CELF Proteins/metabolism , Gene Expression Regulation/genetics , Nerve Tissue Proteins/metabolism , Polyadenylation/genetics , RNA-Binding Proteins/metabolism , 3' Untranslated Regions , CELF Proteins/genetics , Cell Line, Tumor , Cleavage And Polyadenylation Specificity Factor/genetics , Cleavage And Polyadenylation Specificity Factor/metabolism , Enhancer Elements, Genetic , Humans , Introns/genetics , Nerve Tissue Proteins/genetics , Protein Binding , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , RNA-Seq , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction/genetics , Splicing Factor U2AF/genetics , Splicing Factor U2AF/metabolism , Transcriptome
6.
Cell Rep ; 28(8): 2169-2181.e4, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31433990

ABSTRACT

Coordinate control of T cell proliferation, survival, and differentiation are essential for host protection from pathogens and cancer. Long-lived memory cells, whose precursors are formed during the initial immunological insult, provide protection from future encounters, and their generation is the goal of many vaccination strategies. microRNAs (miRNAs) are key nodes in regulatory networks that shape effective T cell responses through the fine-tuning of thousands of genes. Here, using compound conditional mutant mice to eliminate miR-15/16 family miRNAs in T cells, we show that miR-15/16 restrict T cell cycle, survival, and memory T cell differentiation. High throughput sequencing of RNA isolated by cross-linking immunoprecipitation of AGO2 combined with gene expression analysis in miR-15/16-deficient T cells indicates that these effects are mediated through the direct inhibition of an extensive network of target genes within pathways critical to cell cycle, survival, and memory.


Subject(s)
Cell Cycle , Cell Differentiation , Immunologic Memory , MicroRNAs/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , Antigens/metabolism , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Survival/genetics , Gene Expression Regulation , Gene Regulatory Networks , Genetic Loci , Lymphocytic choriomeningitis virus/physiology , Mice, Transgenic , MicroRNAs/genetics
7.
Genome Res ; 29(6): 896-906, 2019 06.
Article in English | MEDLINE | ID: mdl-31152051

ABSTRACT

Compared to coding sequences, untranslated regions of the transcriptome are not well conserved, and functional annotation of these sequences is challenging. Global relationships between nucleotide composition of 3' UTR sequences and their sequence conservation have been appreciated since mammalian genomes were first sequenced, but the functional relevance of these patterns remain unknown. We systematically measured the effect on gene expression of the sequences of more than 25,000 RNA-binding protein (RBP) binding sites in primary mouse T cells using a massively parallel reporter assay. GC-rich sequences were destabilizing of reporter mRNAs and come from more rapidly evolving regions of the genome. These sequences were more likely to be folded in vivo and contain a number of structural motifs that reduced accumulation of a heterologous reporter protein. Comparison of full-length 3' UTR sequences across vertebrate phylogeny revealed that strictly conserved 3' UTRs were GC-poor and enriched in genes associated with organismal development. In contrast, rapidly evolving 3' UTRs tended to be GC-rich and derived from genes involved in metabolism and immune responses. Cell-essential genes had lower GC content in their 3' UTRs, suggesting a connection between unstructured mRNA noncoding sequences and optimal protein production. By reducing gene expression, GC-rich RBP-occupied sequences act as a rapidly evolving substrate for gene regulatory interactions.


Subject(s)
3' Untranslated Regions , Base Composition , Conserved Sequence , Gene Expression Regulation , Gene Expression , Genes, Reporter , RNA, Messenger/genetics , Animals , Base Sequence , Evolution, Molecular , GC Rich Sequence , Humans , Mice , Nucleic Acid Conformation , RNA Stability , RNA, Messenger/chemistry
8.
PLoS One ; 10(5): e0125565, 2015.
Article in English | MEDLINE | ID: mdl-25933216

ABSTRACT

Glioblastoma (GBM) is among the most invasive and lethal of cancers, frequently infiltrating surrounding healthy tissue and giving rise to rapid recurrence. It is therefore critical to establish experimental model systems and develop therapeutic approaches that enhance anti-tumor immunity. In the current study, we have employed a newly developed murine glioma model to assess the efficacy of a novel picornavirus vaccination approach for the treatment of established tumors. The GL261-Quad system is a variation of the GL261 syngeneic glioma that has been engineered to expresses model T cell epitopes including OVA257-264. MRI revealed that both GL261 and GL261-Quad tumors display characteristic features of human gliomas such as heterogeneous gadolinium leakage and larger T2 weighted volumes. Analysis of brain-infiltrating immune cells demonstrated that GL261-Quad gliomas generate detectable CD8+ T cell responses toward the tumor-specific Kb:OVA257-264 antigen. Enhancing this response via a single intracranial or peripheral vaccination with picornavirus expressing the OVA257-264 antigen increased anti-tumor CD8+ T cells infiltrating the brain, attenuated progression of established tumors, and extended survival of treated mice. Importantly, the efficacy of the picornavirus vaccination is dependent on functional cytotoxic activity of CD8+ T cells, as the beneficial response was completely abrogated in mice lacking perforin expression. Therefore, we have developed a novel system for evaluating mechanisms of anti-tumor immunity in vivo, incorporating the GL261-Quad model, 3D volumetric MRI, and picornavirus vaccination to enhance tumor-specific cytotoxic CD8+ T cell responses and track their effectiveness at eradicating established gliomas in vivo.


Subject(s)
Antigens, Neoplasm/immunology , Brain Neoplasms/therapy , Cancer Vaccines/administration & dosage , Glioma/therapy , Picornaviridae/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, Neoplasm/genetics , Biomarkers/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Brain Neoplasms/mortality , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Line, Tumor , Cell Movement/immunology , Disease Models, Animal , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Gene Expression , Glioma/genetics , Glioma/immunology , Glioma/mortality , Humans , Injections, Intraventricular , Mice , Mice, Inbred C57BL , Ovalbumin/genetics , Ovalbumin/immunology , Peptide Fragments/genetics , Peptide Fragments/immunology , Picornaviridae/genetics , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/immunology , Survival Analysis , Treatment Outcome , Vaccination
9.
Cancer Immunol Res ; 2(9): 839-45, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24852944

ABSTRACT

Cytotoxic T cells typically are expanded ex vivo in culture with IL2 for adoptive immunotherapy. This culture period leads to a differentiated phenotype and acquisition of effector function, as well as a loss of in vivo proliferative capability and antitumor efficacy. Here, we report antigen-specific and polyclonal expansion of cytotoxic T cells in a cocktail of cytokines and small molecules that leads to a memory-like phenotype in mouse and human cells even during extended culture, leading to enhanced in vivo expansion and tumor control in mice.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Immunologic Memory , Immunotherapy, Adoptive , Lymphocytes, Tumor-Infiltrating/immunology , Animals , Cell Proliferation , Cell Survival , Cells, Cultured , Cytokines/immunology , Epitopes , Humans , Mice , Mice, Inbred C57BL , Neoplasms/therapy
10.
Oncoimmunology ; 2(10): e26294, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24251080

ABSTRACT

Alkylating chemotherapy exerts both antineoplastic and immunostimulatory effects. However, in addition to depleting regulatory T cells (Treg), alkylating agents also mediate a long lasting antiproliferative effect on responder lymphocytes. Our recent findings indicate that this antiproliferative effect profoundly impairs vaccination-induced immune responses, especially in the case of vaccines that target specific tumor-associated neo-antigens that do not require Treg depletion.

11.
Mol Ther ; 21(8): 1526-35, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23689601

ABSTRACT

Clinical trials reveal that plasmid DNA (pDNA)-based gene delivery must be improved to realize its potential to treat human disease. Current pDNA platforms suffer from brief transgene expression, primarily due to the spread of transcriptionally repressive chromatin initially deposited on plasmid bacterial backbone sequences. Minicircle (MC) DNA lacks plasmid backbone sequences and correspondingly confers higher levels of sustained transgene expression upon delivery, accounting for its success in preclinical gene therapy models. In this study, we show for the first time that MC DNA also functions as a vaccine platform. We used a luciferase reporter transgene to demonstrate that intradermal delivery of MC DNA, relative to pDNA, resulted in significantly higher and persistent levels of luciferase expression in mouse skin. Next, we immunized mice intradermally with DNA encoding a peptide that, when presented by the appropriate major histocompatibility complex class I molecule, was recognized by endogenous CD8(+) T cells. Finally, immunization with peptide-encoding MC DNA, but not the corresponding full-length (FL) pDNA, conferred significant protection in mice challenged with Listeria monocytogenes expressing the model peptide. Together, our results suggest intradermal delivery of MC DNA may prove more efficacious for prophylaxis than traditional pDNA vaccines.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , DNA, Circular/immunology , Epitopes, T-Lymphocyte/immunology , Plasmids/immunology , Adoptive Transfer , Animals , Antigen Presentation/immunology , Cell Line , DNA, Circular/genetics , Epitopes, T-Lymphocyte/genetics , Female , Gene Expression , Gene Order , Genes, Reporter , Genetic Vectors/genetics , Humans , Listeriosis/immunology , Listeriosis/prevention & control , Mice , Plasmids/genetics , Skin/metabolism , Transgenes/immunology , Vaccines, DNA/genetics , Vaccines, DNA/immunology
12.
J Immunol ; 190(12): 6259-68, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23686484

ABSTRACT

Overall, cancer vaccines have had a record of failure as an adjuvant therapy for malignancies that are treated with alkylating chemotherapy, and the contribution of standard treatment to that failure remains unclear. Vaccines aim to harness the proliferative potential of the immune system by expanding a small number of tumor-specific lymphocytes into a large number of antitumor effectors. Clinical trials are often conducted after treatment with alkylating chemotherapy, given either as standard therapy or for immunomodulatory effect. There is mounting evidence for synergy between chemotherapy and adoptive immunotherapy or vaccination against self-Ags; however, the impact of chemotherapy on lymphocytes primed against tumor neoantigens remains poorly defined. We report that clinically relevant dosages of standard alkylating chemotherapies, such as temozolomide and cyclophosphamide, significantly inhibit the proliferative abilities of lymphocytes in mice. This proliferative impairment was long-lasting and led to quantitative and qualitative defects in B and T cell responses to neoantigen vaccines. High-affinity responder lymphocytes receiving the strongest proliferative signals from vaccines experienced the greatest DNA damage responses, skewing the response toward lower-affinity responders with inferior functional characteristics. Together, these defects lead to inferior efficacy and overall survival in murine tumor models treated by neoantigen vaccines. These results suggest that clinical protocols for cancer vaccines should be designed to avoid exposing responder lymphocytes to alkylating chemotherapy.


Subject(s)
Adaptive Immunity/drug effects , Antineoplastic Agents, Alkylating/toxicity , Cancer Vaccines/administration & dosage , Neoplasms, Experimental/therapy , Adaptive Immunity/immunology , Animals , Antineoplastic Agents, Alkylating/administration & dosage , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Combined Modality Therapy , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Experimental/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
13.
J Immunol ; 190(2): 613-20, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23248259

ABSTRACT

Malignant gliomas are lethal brain tumors for which novel therapies are urgently needed. In animal models, vaccination with tumor-associated Ags efficiently primes T cells to clear gliomas. In clinical trials, cancer vaccines have been less effective at priming T cells and extending survival. Generalized immune suppression in the tumor draining lymph nodes has been documented in multiple cancers. However, a systematic analysis of how vaccination at various distances from the tumor (closest to farthest) has not been reported. We investigated how the injection site chosen for vaccination dictates CD8 T cell priming and survival in an OVA-transfected murine glioma model. Glioma-bearing mice were vaccinated with Poly:ICLC plus OVA protein in the neck, hind leg, or foreleg for drainage into the cervical, inguinal, or axillary lymph nodes, respectively. OVA-specific CD8 T cell number, TCR affinity, effector function, and infiltration into the brain decreased as the vaccination site approached the tumor. These effects were dependent on the presence of the tumor, because injection site did not appreciably affect CD8 T cell priming in tumor-free mice. Our data suggest the site of vaccination can greatly impact the effectiveness of cancer vaccines. Considering that previous and ongoing clinical trials have used a variety of injection sites, vaccination site is potentially a critical aspect of study design that is being overlooked.


Subject(s)
Brain Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Glioma/immunology , Animals , Brain Neoplasms/mortality , Brain Neoplasms/therapy , Disease Models, Animal , Female , Glioma/mortality , Glioma/therapy , Mice , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism
14.
PLoS One ; 7(1): e29336, 2012.
Article in English | MEDLINE | ID: mdl-22235286

ABSTRACT

The rationale for using small molecule inhibitors of oncogenic proteins as cancer therapies depends, at least in part, on the assumption that metastatic tumors are primarily clonal with respect to mutant oncogene. With the emergence of BRAF(V600E) as a therapeutic target, we investigated intra- and inter-tumor heterogeneity in melanoma using detection of the BRAF(V600E) mutation as a marker of clonality. BRAF mutant-specific PCR (MS-PCR) and conventional sequencing were performed on 112 tumors from 73 patients, including patients with matched primary and metastatic specimens (n = 18). Nineteen patients had tissues available from multiple metastatic sites. Mutations were detected in 36/112 (32%) melanomas using conventional sequencing, and 85/112 (76%) using MS-PCR. The better sensitivity of the MS-PCR to detect the mutant BRAF(V600E) allele was not due to the presence of contaminating normal tissue, suggesting that the tumor was comprised of subclones of differing BRAF genotypes. To determine if tumor subclones were present in individual primary melanomas, we performed laser microdissection and mutation detection via sequencing and BRAF(V600E)-specific SNaPshot analysis in 9 cases. Six of these cases demonstrated differing proportions of BRAF(V600E)and BRAF(wild-type) cells in distinct microdissected regions within individual tumors. Additional analyses of multiple metastatic samples from individual patients using the highly sensitive MS-PCR without microdissection revealed that 5/19 (26%) patients had metastases that were discordant for the BRAF(V600E) mutation. In conclusion, we used highly sensitive BRAF mutation detection methods and observed substantial evidence for heterogeneity of the BRAF(V600E) mutation within individual melanoma tumor specimens, and among multiple specimens from individual patients. Given the varied clinical responses of patients to BRAF inhibitor therapy, these data suggest that additional studies to determine possible associations between clinical outcomes and intra- and inter-tumor heterogeneity could prove fruitful.


Subject(s)
Melanoma/genetics , Melanoma/secondary , Mutation , Proto-Oncogene Proteins B-raf/genetics , Base Sequence , DNA Mutational Analysis , Humans , Melanoma/pathology , Microdissection , Neoplasm Metastasis , Polymerase Chain Reaction
15.
Cancer Res ; 71(21): 6583-9, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21908554

ABSTRACT

With recent approval of the first dendritic cell (DC) vaccine for patient use, many other DC vaccine approaches are now being tested in clinical trials. Many of these DC vaccines employ tumor cell lysates (TL) generated from cells cultured in atmospheric oxygen (∼20% O2) that greatly exceeds levels found in tumors in situ. In this study, we tested the hypothesis that TLs generated from tumor cells cultured under physiologic oxygen (∼5% O2) would be more effective as a source for DC antigens. Gene expression patterns in primary glioma cultures established at 5% O2 more closely paralleled patient tumors in situ and known immunogenic antigens were more highly expressed. DCs treated with TLs generated from primary tumor cells maintained in 5% O2 took up and presented antigens to CD8 T cells more efficiently. Moreover, CD8 T cells primed in this manner exhibited superior tumoricidal activity against target cells cultured in either atmospheric 20% O2 or physiologic 5% O2. Together, these results establish a simple method to greatly improve the effectiveness of DC vaccines in stimulating the production of tumoricidal T cells, with broad implications for many of the DC-based cancer vaccines being developed for clinical application.


Subject(s)
Antigens, Neoplasm/immunology , Brain Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/pathology , Oxygen/physiology , T-Lymphocytes, Cytotoxic/immunology , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Cancer Vaccines , Cell Extracts/immunology , Cell Hypoxia/immunology , Culture Media/chemistry , Culture Media/pharmacology , Culture Media, Serum-Free/pharmacology , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Glioblastoma/genetics , Glioblastoma/immunology , Glioblastoma/metabolism , HLA-A2 Antigen/immunology , Humans , In Vitro Techniques , Oxygen/pharmacology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...