Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 37
Filtrer
1.
J Infect Dis ; 228(5): 555-563, 2023 08 31.
Article de Anglais | MEDLINE | ID: mdl-37062677

RÉSUMÉ

Emerging variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) possess mutations that prevent antibody therapeutics from maintaining antiviral binding and neutralizing efficacy. Monoclonal antibodies (mAbs) shown to neutralize Wuhan-Hu-1 SARS-CoV-2 (ancestral) strain have reduced potency against newer variants. Plasma-derived polyclonal hyperimmune drugs have improved neutralization breadth compared with mAbs, but lower titers against SARS-CoV-2 require higher dosages for treatment. We previously developed a highly diverse, recombinant polyclonal antibody therapeutic anti-SARS-CoV-2 immunoglobulin hyperimmune (rCIG). rCIG was compared with plasma-derived or mAb standards and showed improved neutralization of SARS-CoV-2 across World Health Organization variants; however, its potency was reduced against some variants relative to ancestral, particularly omicron. Omicron-specific antibody sequences were enriched from yeast expressing rCIG-scFv and exhibited increased binding and neutralization to omicron BA.2 while maintaining ancestral strain binding and neutralization. Polyclonal antibody libraries such as rCIG can be utilized to develop antibody therapeutics against present and future SARS-CoV-2 threats.


Sujet(s)
COVID-19 , Humains , SARS-CoV-2/génétique , Anticorps monoclonaux/usage thérapeutique , Antiviraux , Saccharomyces cerevisiae , Anticorps neutralisants/usage thérapeutique , Glycoprotéine de spicule des coronavirus/génétique , Anticorps antiviraux/usage thérapeutique
2.
J Clin Immunol ; 43(5): 940-950, 2023 07.
Article de Anglais | MEDLINE | ID: mdl-36826743

RÉSUMÉ

PURPOSE: Most individuals with antibody deficiency (hypogammaglobulinemia) need immunoglobulin replacement therapy (IgG-RT) from healthy plasma donors to stay clear of infections. However, a small subset of hypogammaglobulinemic patients do not require this substitution therapy. We set out to investigate this clinical conundrum by asking whether the peripheral B cell receptor repertoires differ between antibody-deficient patients who do and do not need IgG-RT. METHODS: We sequenced and analyzed IgG and IgM heavy chain B cell receptor repertoires from peripheral blood mononuclear cells (PBMCs) isolated from patients with low serum IgG concentrations who did or did not require IgG-RT. RESULTS: Compared to the patients who did not need IgG-RT, those who needed IgG-RT had higher numbers of IgG antibody clones, higher IgM diversity, and less oligoclonal IgG and IgM repertoires. The patient cohorts had different heavy chain variable gene usage, and the patients who needed IgG-RT had elevated frequencies of IgG clones with higher germline identity (i.e., fewer somatic hypermutations). CONCLUSION: Antibody-deficient patients with infection susceptibility who needed IgG-RT had more diverse peripheral antibody repertoires that were less diverged from germline and thus may not be as optimal for targeting pathogens, possibly contributing to infection susceptibility.


Sujet(s)
Immunoglobuline G , Agranulocytes , Humains , Immunoglobuline M , Séquence nucléotidique , Récepteurs pour l'antigène des lymphocytes B/génétique
3.
Pathogens ; 11(7)2022 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-35890050

RÉSUMÉ

Conventionally, hyperimmune globulin drugs manufactured from pooled immunoglobulins from vaccinated or convalescent donors have been used in treating infections where no treatment is available. This is especially important where multi-epitope neutralization is required to prevent the development of immune-evading viral mutants that can emerge upon treatment with monoclonal antibodies. Using microfluidics, flow sorting, and a targeted integration cell line, a first-in-class recombinant hyperimmune globulin therapeutic against SARS-CoV-2 (GIGA-2050) was generated. Using processes similar to conventional monoclonal antibody manufacturing, GIGA-2050, comprising 12,500 antibodies, was scaled-up for clinical manufacturing and multiple development/tox lots were assessed for consistency. Antibody sequence diversity, cell growth, productivity, and product quality were assessed across different manufacturing sites and production scales. GIGA-2050 was purified and tested for good laboratory procedures (GLP) toxicology, pharmacokinetics, and in vivo efficacy against natural SARS-CoV-2 infection in mice. The GIGA-2050 master cell bank was highly stable, producing material at consistent yield and product quality up to >70 generations. Good manufacturing practices (GMP) and development batches of GIGA-2050 showed consistent product quality, impurity clearance, potency, and protection in an in vivo efficacy model. Nonhuman primate toxicology and pharmacokinetics studies suggest that GIGA-2050 is safe and has a half-life similar to other recombinant human IgG1 antibodies. These results supported a successful investigational new drug application for GIGA-2050. This study demonstrates that a new class of drugs, recombinant hyperimmune globulins, can be manufactured consistently at the clinical scale and presents a new approach to treating infectious diseases that targets multiple epitopes of a virus.

4.
MAbs ; 14(1): 2069075, 2022.
Article de Anglais | MEDLINE | ID: mdl-35482911

RÉSUMÉ

The antibody drug field has continually sought improvements to methods for candidate discovery and engineering. Historically, most such methods have been laboratory-based, but informatics methods have recently started to make an impact. Deep learning, a subfield of machine learning, is rapidly gaining prominence in the biomedical research. Recent advances in microfluidics technologies and next-generation sequencing have not only revolutionized therapeutic antibody discovery, but also contributed to a vast amount of antibody repertoire sequencing data, providing opportunities for deep learning-based applications. Previously, we used microfluidics, yeast display, and deep sequencing to generate a panel of binder and non-binder antibody sequences to the cancer immunotherapy targets PD-1 and CTLA-4. Here we encoded the antibody light and heavy chain complementarity-determining regions (CDR3s) into antibody images, then built and trained convolutional neural network models to classify binders and non-binders. To improve model interpretability, we performed in silico mutagenesis to identify CDR3 residues that were important for binder classification. We further built generative deep learning models using generative adversarial network models to produce synthetic antibodies against PD-1 and CTLA-4. Our models generated variable length CDR3 sequences that resemble real sequences. Overall, our study demonstrates that deep learning methods can be leveraged to mine and learn patterns in antibody sequences, offering insights into antibody engineering, optimization, and discovery.


Sujet(s)
Apprentissage profond , Anticorps , Antigène CTLA-4 , Régions déterminant la complémentarité/composition chimique , Récepteur-1 de mort cellulaire programmée
5.
BMC Biol ; 19(1): 107, 2021 05 25.
Article de Anglais | MEDLINE | ID: mdl-34030676

RÉSUMÉ

BACKGROUND: The anti-tumor activity of anti-PD-1/PD-L1 therapies correlates with T cell infiltration in tumors. Thus, a major goal in oncology is to find strategies that enhance T cell infiltration and efficacy of anti-PD-1/PD-L1 therapy. TGF-ß has been shown to contribute to T cell exclusion, and anti-TGF-ß improves anti-PD-L1 efficacy in vivo. However, TGF-ß inhibition has frequently been shown to induce toxicity in the clinic, and the clinical efficacy of combination PD-L1 and TGF-ß blockade has not yet been proven. To identify strategies to overcome resistance to PD-L1 blockade, the transcriptional programs associated with PD-L1 and/or TGF-ß blockade in the tumor microenvironment should be further elucidated. RESULTS: We used single-cell RNA sequencing in a mouse model to characterize the transcriptomic effects of PD-L1 and/or TGF-ß blockade on nearly 30,000 single cells in the tumor and surrounding microenvironment. Combination treatment led to upregulation of immune response genes, including multiple chemokine genes such as CCL5, in macrophages, and downregulation of extracellular matrix genes in fibroblasts. Analysis of publicly available tumor transcriptome profiles showed that the chemokine CCL5 was strongly associated with immune cell infiltration in various human cancers. Further investigation with in vivo models showed that intratumorally administered CCL5 enhanced cytotoxic lymphocytes and the anti-tumor activity of anti-PD-L1. CONCLUSIONS: Taken together, our data could be leveraged translationally to complement or find alternatives to anti-PD-L1 plus anti-TGF-ß combination therapy, for example through companion biomarkers, and/or to identify novel targets that could be modulated to overcome resistance.


Sujet(s)
Tumeurs , Animaux , Antigène CD274/génétique , Souris , Transcriptome , Facteur de croissance transformant bêta , Microenvironnement tumoral
6.
Nat Biotechnol ; 39(8): 989-999, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-33859400

RÉSUMÉ

Plasma-derived polyclonal antibody therapeutics, such as intravenous immunoglobulin, have multiple drawbacks, including low potency, impurities, insufficient supply and batch-to-batch variation. Here we describe a microfluidics and molecular genomics strategy for capturing diverse mammalian antibody repertoires to create recombinant multivalent hyperimmune globulins. Our method generates of diverse mixtures of thousands of recombinant antibodies, enriched for specificity and activity against therapeutic targets. Each hyperimmune globulin product comprised thousands to tens of thousands of antibodies derived from convalescent or vaccinated human donors or from immunized mice. Using this approach, we generated hyperimmune globulins with potent neutralizing activity against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) in under 3 months, Fc-engineered hyperimmune globulins specific for Zika virus that lacked antibody-dependent enhancement of disease, and hyperimmune globulins specific for lung pathogens present in patients with primary immune deficiency. To address the limitations of rabbit-derived anti-thymocyte globulin, we generated a recombinant human version and demonstrated its efficacy in mice against graft-versus-host disease.


Sujet(s)
Lymphocytes B/immunologie , COVID-19/thérapie , Globulines/biosynthèse , SARS-CoV-2/immunologie , Animaux , Anticorps antiviraux/immunologie , Cellules CHO , Cricetulus , Test ELISA , Globulines/immunologie , Humains , Immunisation passive , Souris , Protéines recombinantes/biosynthèse , Protéines recombinantes/immunologie , Virus Zika/immunologie , Sérothérapie COVID-19
7.
MAbs ; 12(1): 1803646, 2020.
Article de Anglais | MEDLINE | ID: mdl-32744131

RÉSUMÉ

IN VITRO: affinity maturation of therapeutic monoclonal antibodies is commonly applied to achieve desired properties, such as improved binding kinetics and affinity. Currently there are no universally accepted protocols for generation of variegated antibody libraries or selection thereof. Here, we performed affinity maturation using a yeast-based single-chain variable fragment (scFv) expression system to compare two mutagenesis methods: random mutagenesis across the entire V(D)J region by error-prone PCR, and a novel combinatorial mutagenesis process limited to the complementarity-determining regions (CDRs). We applied both methods of mutagenesis to four human antibodies against well-known immuno-oncology target proteins. Detailed sequence analysis showed an even mutational distribution across the entire length of the scFv for the error-prone PCR method and an almost exclusive targeting of the CDRs for the combinatorial method. Though there were distinct mutagenesis profiles for each target antibody and mutagenesis method, we found that both methods improved scFv affinity with similar efficiency. When a subset of the affinity-matured antibodies was expressed as full-length immunoglobulin, the measured affinity constants were mostly comparable to those of the respective scFv, but the full-length antibodies were inferior to their scFv counterparts for one of the targets. Furthermore, we found that improved affinity for the full-length antibody did not always translate into enhanced binding to cell-surface expressed antigen or improved immune checkpoint blocking ability, suggesting that screening with full-length antibody or antigen-binding fragment formats might be advantageous and the subject of a future study.


Sujet(s)
Affinité des anticorps/génétique , Mutagenèse , Anticorps à chaîne unique , Régions déterminant la complémentarité/composition chimique , Régions déterminant la complémentarité/génétique , Humains , Réaction de polymérisation en chaîne , Anticorps à chaîne unique/composition chimique , Anticorps à chaîne unique/génétique
8.
Nat Biotechnol ; 38(5): 609-619, 2020 05.
Article de Anglais | MEDLINE | ID: mdl-32393905

RÉSUMÉ

T cells engineered to express antigen-specific T cell receptors (TCRs) are potent therapies for viral infections and cancer. However, efficient identification of clinical candidate TCRs is complicated by the size and complexity of T cell repertoires and the challenges of working with primary T cells. Here we present a high-throughput method to identify TCRs with high functional avidity from diverse human T cell repertoires. The approach used massively parallel microfluidics to generate libraries of natively paired, full-length TCRαß clones, from millions of primary T cells, which were then expressed in Jurkat cells. The TCRαß-Jurkat libraries enabled repeated screening and panning for antigen-reactive TCRs using peptide major histocompatibility complex binding and cellular activation. We captured more than 2.9 million natively paired TCRαß clonotypes from six healthy human donors and identified rare (<0.001% frequency) viral-antigen-reactive TCRs. We also mined a tumor-infiltrating lymphocyte sample from a patient with melanoma and identified several tumor-specific TCRs, which, after expression in primary T cells, led to tumor cell killing.


Sujet(s)
Antigènes/analyse , Récepteur lymphocytaire T antigène, alpha-bêta/immunologie , Lymphocytes T/cytologie , Ingénierie cellulaire , Banque de gènes , Humains , Cellules Jurkat , Lymphocytes TIL/immunologie , Mélanome/immunologie , Lymphocytes T/immunologie , Virus/immunologie
9.
Antibodies (Basel) ; 8(1)2019 Feb 19.
Article de Anglais | MEDLINE | ID: mdl-31544823

RÉSUMÉ

To discover therapeutically relevant antibody candidates, many groups use mouse immunization followed by hybridoma generation or B cell screening. One modern approach is to screen B cells by generating natively paired single chain variable fragment (scFv) display libraries in yeast. Such methods typically rely on soluble antigens for scFv library screening. However, many therapeutically relevant cell-surface targets are difficult to express in a soluble protein format, complicating discovery. In this study, we developed methods to screen humanized mouse-derived yeast scFv libraries using recombinant OX40 protein in cell lysate. We used deep sequencing to compare screening with cell lysate to screening with soluble OX40 protein, in the context of mouse immunizations using either soluble OX40 or OX40-expressing cells and OX40-encoding DNA vector. We found that all tested methods produce a unique diversity of scFv binders. However, when we reformatted forty-one of these scFv as full-length monoclonal antibodies (mAbs), we observed that mAbs identified using soluble antigen immunization with cell lysate sorting always bound cell surface OX40, whereas other methods had significant false positive rates. Antibodies identified using soluble antigen immunization and cell lysate sorting were also significantly more likely to activate OX40 in a cellular assay. Our data suggest that sorting with OX40 protein in cell lysate is more likely than other methods to retain the epitopes required for antibody-mediated OX40 agonism.

10.
MAbs ; 11(5): 870-883, 2019 07.
Article de Anglais | MEDLINE | ID: mdl-30898066

RÉSUMÉ

Immunization of mice followed by hybridoma or B-cell screening is one of the most common antibody discovery methods used to generate therapeutic monoclonal antibody (mAb) candidates. There are a multitude of different immunization protocols that can generate an immune response in animals. However, an extensive analysis of the antibody repertoires that these alternative immunization protocols can generate has not been performed. In this study, we immunized mice that transgenically express human antibodies with either programmed cell death 1 protein or cytotoxic T-lymphocyte associated protein 4 using four different immunization protocols, and then utilized a single cell microfluidic platform to generate tissue-specific, natively paired immunoglobulin (Ig) repertoires from each method and enriched for target-specific binders using yeast single-chain variable fragment (scFv) display. We deep sequenced the scFv repertoires from both the pre-sort and post-sort libraries. All methods and both targets yielded similar oligoclonality, variable (V) and joining (J) gene usage, and divergence from germline of enriched libraries. However, there were differences between targets and/or immunization protocols for overall clonal counts, complementarity-determining region 3 (CDR3) length, and antibody/CDR3 sequence diversity. Our data suggest that, although different immunization protocols may generate a response to an antigen, performing multiple immunization protocols in parallel can yield greater Ig diversity. We conclude that modern microfluidic methods, followed by an extensive molecular genomic analysis of antibody repertoires, can be used to quickly analyze new immunization protocols or mouse platforms.


Sujet(s)
Anticorps monoclonaux humanisés/génétique , Diversité des anticorps , Immunisation/méthodes , Microfluidique/méthodes , Animaux , Anticorps monoclonaux humanisés/immunologie , Lymphocytes B/immunologie , Antigène CTLA-4/immunologie , Régions déterminant la complémentarité/génétique , Génomique/méthodes , Séquençage nucléotidique à haut débit , Humains , Hybridomes , Souris , Souris transgéniques , Banque de peptides , Récepteur-1 de mort cellulaire programmée/immunologie , Anticorps à chaîne unique/génétique , Anticorps à chaîne unique/immunologie
11.
MAbs ; 10(3): 431-443, 2018 04.
Article de Anglais | MEDLINE | ID: mdl-29376776

RÉSUMÉ

Deep sequencing and single-chain variable fragment (scFv) yeast display methods are becoming more popular for discovery of therapeutic antibody candidates in mouse B cell repertoires. In this study, we compare a deep sequencing and scFv display method that retains native heavy and light chain pairing with a related method that randomly pairs heavy and light chain. We performed the studies in a humanized mouse, using interleukin 21 receptor (IL-21R) as a test immunogen. We identified 44 high-affinity binder scFv with the native pairing method and 100 high-affinity binder scFv with the random pairing method. 30% of the natively paired scFv binders were also discovered with the randomly paired method, and 13% of the randomly paired binders were also discovered with the natively paired method. Additionally, 33% of the scFv binders discovered only in the randomly paired library were initially present in the natively paired pre-sort library. Thus, a significant proportion of "randomly paired" scFv were actually natively paired. We synthesized and produced 46 of the candidates as full-length antibodies and subjected them to a panel of binding assays to characterize their therapeutic potential. 87% of the antibodies were verified as binding IL-21R by at least one assay. We found that antibodies with native light chains were more likely to bind IL-21R than antibodies with non-native light chains, suggesting a higher false positive rate for antibodies from the randomly paired library. Additionally, the randomly paired method failed to identify nearly half of the true natively paired binders, suggesting a higher false negative rate. We conclude that natively paired libraries have critical advantages in sensitivity and specificity for antibody discovery programs.


Sujet(s)
Lymphocytes B/immunologie , Banque de gènes , Chaines légères des immunoglobulines , Sous-unité alpha du récepteur à l'interleukine 21 , Anticorps à chaîne unique , Animaux , Humains , Chaines légères des immunoglobulines/biosynthèse , Chaines légères des immunoglobulines/génétique , Chaines légères des immunoglobulines/immunologie , Sous-unité alpha du récepteur à l'interleukine 21/antagonistes et inhibiteurs , Sous-unité alpha du récepteur à l'interleukine 21/immunologie , Souris , Anticorps à chaîne unique/génétique , Anticorps à chaîne unique/immunologie
12.
MAbs ; 9(8): 1282-1296, 2017.
Article de Anglais | MEDLINE | ID: mdl-28846502

RÉSUMÉ

Affinity-matured, functional anti-pathogen antibodies are present at low frequencies in natural human repertoires. These antibodies are often excellent candidates for therapeutic monoclonal antibodies. However, mining natural human antibody repertoires is a challenge. In this study, we demonstrate a new method that uses microfluidics, yeast display, and deep sequencing to identify 247 natively paired anti-pathogen single-chain variable fragments (scFvs), which were initially as rare as 1 in 100,000 in the human repertoires. Influenza A vaccination increased the frequency of influenza A antigen-binding scFv within the peripheral B cell repertoire from <0.1% in non-vaccinated donors to 0.3-0.4% in vaccinated donors, whereas pneumococcus vaccination did not increase the frequency of antigen-binding scFv. However, the pneumococcus scFv binders from the vaccinated library had higher heavy and light chain Replacement/Silent mutation (R/S) ratios, a measure of affinity maturation, than the pneumococcus binders from the corresponding non-vaccinated library. Thus, pneumococcus vaccination may increase the frequency of affinity-matured antibodies in human repertoires. We synthesized 10 anti-influenza A and nine anti-pneumococcus full-length antibodies that were highly abundant among antigen-binding scFv. All 10 anti-influenza A antibodies bound the appropriate antigen at KD<10 nM and neutralized virus in cellular assays. All nine anti-pneumococcus full-length antibodies bound at least one polysaccharide serotype, and 71% of the anti-pneumococcus antibodies that we tested were functional in cell killing assays. Our approach has future application in a variety of fields, including the development of therapeutic antibodies for emerging viral diseases, autoimmune disorders, and cancer.


Sujet(s)
Anti-infectieux/immunologie , Anticorps monoclonaux/immunologie , Affinité des anticorps/immunologie , Génomique/méthodes , Microfluidique/méthodes , Séquence d'acides aminés , Anti-infectieux/administration et posologie , Anti-infectieux/métabolisme , Anticorps monoclonaux/administration et posologie , Anticorps monoclonaux/métabolisme , Séquençage nucléotidique à haut débit , Humains , Virus de la grippe A/effets des médicaments et des substances chimiques , Virus de la grippe A/immunologie , Banque de peptides , Anticorps à chaîne unique/génétique , Anticorps à chaîne unique/immunologie , Anticorps à chaîne unique/métabolisme , Streptococcus pneumoniae/effets des médicaments et des substances chimiques , Streptococcus pneumoniae/immunologie
13.
MAbs ; 9(8): 1270-1281, 2017.
Article de Anglais | MEDLINE | ID: mdl-28846506

RÉSUMÉ

Conventionally, mouse hybridomas or well-plate screening are used to identify therapeutic monoclonal antibody candidates. In this study, we present an alternative to hybridoma-based discovery that combines microfluidics, yeast single-chain variable fragment (scFv) display, and deep sequencing to rapidly interrogate and screen mouse antibody repertoires. We used our approach on six wild-type mice to identify 269 molecules that bind to programmed cell death protein 1 (PD-1), which were present at an average of 1 in 2,000 in the pre-sort scFv libraries. Two rounds of fluorescence-activated cell sorting (FACS) produced populations of PD-1-binding scFv with a mean enrichment of 800-fold, whereas most scFv present in the pre-sort mouse repertoires were de-enriched. Therefore, our work suggests that most of the antibodies present in the repertoires of immunized mice are not strong binders to PD-1. We observed clusters of related antibody sequences in each mouse following FACS, suggesting evolution of clonal lineages. In the pre-sort repertoires, these putative clonal lineages varied in both the complementary-determining region (CDR)3K and CDR3H, while the FACS-selected PD-1-binding subsets varied primarily in the CDR3H. PD-1 binders were generally not highly diverged from germline, showing 98% identity on average with germline V-genes. Some CDR3 sequences were discovered in more than one animal, even across different mouse strains, suggesting convergent evolution. We synthesized 17 of the anti-PD-1 binders as full-length monoclonal antibodies. All 17 full-length antibodies bound recombinant PD-1 with KD < 500 nM (average = 62 nM). Fifteen of the 17 full-length antibodies specifically bound surface-expressed PD-1 in a FACS assay, and nine of the antibodies functioned as checkpoint inhibitors in a cellular assay. We conclude that our method is a viable alternative to hybridomas, with key advantages in comprehensiveness and turnaround time.


Sujet(s)
Anticorps monoclonaux/immunologie , Affinité des anticorps/immunologie , Génomique/méthodes , Microfluidique/méthodes , Récepteur-1 de mort cellulaire programmée/immunologie , Animaux , Anticorps monoclonaux/métabolisme , Anticorps monoclonaux/pharmacologie , Régions déterminant la complémentarité/génétique , Régions déterminant la complémentarité/immunologie , Régions déterminant la complémentarité/métabolisme , Cytométrie en flux , Séquençage nucléotidique à haut débit , Humains , Hybridomes , Souris , Banque de peptides , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Liaison aux protéines/immunologie , Anticorps à chaîne unique/génétique , Anticorps à chaîne unique/immunologie , Anticorps à chaîne unique/métabolisme
14.
Genes Dev ; 28(10): 1068-84, 2014 May 15.
Article de Anglais | MEDLINE | ID: mdl-24788092

RÉSUMÉ

The spliceosome machinery is composed of multimeric protein complexes that generate a diverse repertoire of mRNA through coordinated splicing of heteronuclear RNAs. While somatic mutations in spliceosome components have been discovered in several cancer types, the molecular bases and consequences of spliceosome aberrations in cancer are poorly understood. Here we report for the first time that PRPF6, a member of the tri-snRNP (small ribonucleoprotein) spliceosome complex, drives cancer proliferation by preferential splicing of genes associated with growth regulation. Inhibition of PRPF6 and other tri-snRNP complex proteins, but not other snRNP spliceosome complexes, selectively abrogated growth in cancer cells with high tri-snRNP levels. High-resolution transcriptome analyses revealed that reduced PRPF6 alters the constitutive and alternative splicing of a discrete number of genes, including an oncogenic isoform of the ZAK kinase. These findings implicate an essential role for PRPF6 in cancer via splicing of distinct growth-related gene products.


Sujet(s)
Tumeurs du côlon/génétique , Tumeurs du côlon/anatomopathologie , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Épissage alternatif , Lignée cellulaire , Lignée cellulaire tumorale , Prolifération cellulaire , Humains , Isoformes de protéines , Facteurs d'épissage des ARN , Splicéosomes
15.
BMC Biotechnol ; 13: 55, 2013 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-23826749

RÉSUMÉ

BACKGROUND: Monoclonal antibody therapeutics are rapidly gaining in popularity for the treatment of a myriad of diseases, ranging from cancer to autoimmune diseases and neurological diseases. Multiple forms of antibody therapeutics are in use today that differ in the amount of human sequence present in both the constant and variable regions, where antibodies that are more human-like usually have reduced immunogenicity in clinical trials. RESULTS: Here we present a method to quantify the humanness of the variable region of monoclonal antibodies and show that this method is able to clearly distinguish human and non-human antibodies with excellent specificity. After creating and analyzing a database of human antibody sequences, we conducted an in-depth analysis of the humanness of therapeutic antibodies, and found that increased humanness score is correlated with decreased immunogenicity of antibodies. We further discovered a surprisingly similarity in the immunogenicity of fully human antibodies and humanized antibodies that are more human-like based on their humanness score. CONCLUSIONS: Our results reveal that in most cases humanizing an antibody and confirming the humanness of the final form may be sufficient to eliminate immunogenicity issues to the same extent as using fully human antibodies. We created a public website to calculate the humanness score of any input antibody sequence based on our human antibody database. This tool will be of great value during the preclinical drug development process for new monoclonal antibody therapeutics.


Sujet(s)
Anticorps monoclonaux humanisés/composition chimique , Anticorps monoclonaux humanisés/immunologie , Spécificité des anticorps/génétique , Animaux , Anticorps monoclonaux humanisés/usage thérapeutique , Bases de données factuelles , Humains , Phénomènes immunogénétiques/génétique , Région variable d'immunoglobuline/composition chimique , Région variable d'immunoglobuline/génétique , Souris , Rats
16.
Clin Cancer Res ; 19(4): 773-84, 2013 Feb 15.
Article de Anglais | MEDLINE | ID: mdl-23224736

RÉSUMÉ

PURPOSE: This study is aimed to identify genes within the KRAS genomic amplicon that are both coupregulated and essential for cell proliferation when KRAS is amplified in lung cancer. EXPERIMENTAL DESIGN: We used an integrated genomic approach to identify genes that are coamplified with KRAS in lung adenocarcinomas and subsequently preformed an RNA interference (RNAi) screen to uncover functionally relevant genes. The role of lactate dehydrogenase B (LDHB) was subsequently investigated both in vitro and in vivo by siRNA and short hairpin RNA (shRNA)-mediated knockdown in a panel of lung adenocarcinoma cells lines. LDHB expression was also investigated in patient tumors using microarray and immunohistochemistry analyses. RESULTS: RNAi-mediated depletion of LDHB abrogated cell proliferation both in vitro and in xenografted tumors in vivo. We find that LDHB expression correlates to both KRAS genomic copy number gain and KRAS mutation in lung cancer cell lines and adenocarcinomas. This correlation between LDHB expression and KRAS status is specific for lung cancers and not other tumor types that harbor KRAS mutations. Consistent with a role for LDHB in glycolysis and tumor metabolism, KRAS-mutant lung tumors exhibit elevated expression of a glycolysis gene signature and are more dependent on glycolysis for proliferation compared with KRAS wild-type lung tumors. Finally, high LDHB expression was a significant predictor of shorter survival in patients with lung adenocarcinomas. CONCLUSION: This study identifies LDHB as a regulator of cell proliferation in a subset of lung adenocarcinoma and may provide a novel therapeutic approach for treating lung cancer.


Sujet(s)
Adénocarcinome/génétique , L-Lactate dehydrogenase/génétique , Tumeurs du poumon/génétique , Protéines proto-oncogènes/métabolisme , Protéines G ras/métabolisme , Adénocarcinome/métabolisme , Adénocarcinome/anatomopathologie , Adénocarcinome pulmonaire , Animaux , Lignée cellulaire tumorale , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Techniques de knock-down de gènes , Humains , Isoenzymes/génétique , Isoenzymes/métabolisme , Estimation de Kaplan-Meier , L-Lactate dehydrogenase/métabolisme , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Souris , Pronostic , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes p21(ras) , Petit ARN interférent , Transplantation hétérologue , Protéines G ras/génétique
17.
Cancer Res ; 72(22): 5812-23, 2012 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-23139210

RÉSUMÉ

Breast cancer has been redefined into three clinically relevant subclasses: (i) estrogen/progesterone receptor positive (ER+/PR+), (ii) HER2/ERRB2 positive, and (iii) those lacking expression of all three markers (triple negative or basal-like). While targeted therapies for ER+/PR+ and HER2+ tumors have revolutionized patient treatment and increased lifespan, an urgent need exists for identifying novel targets for triple-negative breast cancers. Here, we used integrative genomic analysis to identify candidate oncogenes in triple-negative breast tumors and assess their function through loss of function screening. Using this approach, we identify lactate dehydrogenase B (LDHB), a component of glycolytic metabolism, as an essential gene in triple-negative breast cancer. Loss of LDHB abrogated cell proliferation in vitro and arrested tumor growth in fully formed tumors in vivo. We find that LDHB and other related glycolysis genes are specifically upregulated in basal-like/triple-negative breast cancers as compared with other subtypes, suggesting that these tumors are distinctly glycolytic. Consistent with this, triple-negative breast cancer cell lines were more dependent on glycolysis for growth than luminal cell lines. Finally, we find that patients with breast cancer and high LDHB expression in their tumors had a poor clinical outcome. While previous studies have focused on the ubiquitous role of LDHA in tumor metabolism and growth, our data reveal that LDHB is upregulated and required only in certain cancer genotypes. These findings suggest that targeting LDHB or other components of lactate metabolism would be of clinical benefit in triple-negative breast cancer.


Sujet(s)
Tumeurs du sein/génétique , Lactate dehydrogenases/génétique , Animaux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Processus de croissance cellulaire/génétique , Lignée cellulaire tumorale , Femelle , Techniques de knock-down de gènes , Humains , Lactate dehydrogenases/biosynthèse , Cellules MCF-7 , Souris , Souris nude , Pronostic , Récepteur ErbB-2/métabolisme , Récepteurs des oestrogènes/métabolisme , Récepteurs à la progestérone/métabolisme , Transplantation hétérologue
18.
Cancer Discov ; 2(7): 638-51, 2012 Jul.
Article de Anglais | MEDLINE | ID: mdl-22728437

RÉSUMÉ

UNLABELLED: The transcription factor ZNF217 is a candidate oncogene in the amplicon on chromosome 20q13 that occurs in 20% to 30% of primary human breast cancers and that correlates with poor prognosis. We show that Znf217 overexpression drives aberrant differentiation and signaling events, promotes increased self-renewal capacity, mesenchymal marker expression, motility, and metastasis, and represses an adult tissue stem cell gene signature downregulated in cancers. By in silico screening, we identified candidate therapeutics that at low concentrations inhibit growth of cancer cells expressing high ZNF217. We show that the nucleoside analogue triciribine inhibits ZNF217-induced tumor growth and chemotherapy resistance and inhibits signaling events [e.g., phospho-AKT, phospho-mitogen-activated protein kinase (MAPK)] in vivo. Our data suggest that ZNF217 is a biomarker of poor prognosis and a therapeutic target in patients with breast cancer and that triciribine may be part of a personalized treatment strategy in patients overexpressing ZNF217. Because ZNF217 is amplified in numerous cancers, these results have implications for other cancers. SIGNIFICANCE: This study finds that ZNF217 is a poor prognostic indicator and therapeutic target in patients with breast cancer and may be a strong biomarker of triciribine treatment efficacy in patients. Because previous clinical trials for triciribine did not include biomarkers of treatment efficacy, this study provides a rationale for revisiting triciribine in the clinical setting as a therapy for patients with breast cancer who overexpress ZNF217.


Sujet(s)
Marqueurs biologiques tumoraux/génétique , Tumeurs du sein/génétique , Régulation de l'expression des gènes tumoraux , Transactivateurs/génétique , Animaux , Antibiotiques antinéoplasiques/pharmacologie , Marqueurs biologiques tumoraux/métabolisme , Technique de Western , Tumeurs du sein/anatomopathologie , Tumeurs du sein/prévention et contrôle , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Évolution de la maladie , Doxorubicine/pharmacologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Femelle , Analyse de profil d'expression de gènes , Humains , Cellules MCF-7 , Souris , Cellules NIH 3T3 , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Séquençage par oligonucléotides en batterie , Pronostic , RT-PCR , Ribonucléosides/pharmacologie , Analyse de survie , Transactivateurs/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
19.
Cancer Res ; 72(8): 2129-39, 2012 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-22345154

RÉSUMÉ

CDK8 is a cyclin-dependent kinase that mediates transcriptional control of pathways linked to both cancer and stem cells. In this study, we show that CDK8 is required for both tumor growth and maintenance of tumor dedifferentiation in vivo and uncover a common role for CDK8 in controlling cancer and stem cell function. Acute CDK8 loss in vivo strongly inhibited tumor growth and promoted differentiation. Transcriptional profiling identified a set of embryonic stem cell-related genes that are activated by CDK8 in cancer. Consistent with this, we found that CDK8 expression correlated to the embryonic stem cell pluripotency state and loss of CDK8 caused embryonic stem cells to differentiate. This effect was, at least partially, mediated by the ability of CDK8 to regulate MYC protein and downstream MYC target gene expression. Similar regulation of MYC target genes by CDK8 was observed in colon tumor cells, and increased expression of a CDK8-regulated, embryonic stem cell MYC target gene signature was associated with loss of differentiation and poor outcome in primary human colon cancers. Together, these observations reveal that CDK8 acts, at least in part, through MYC to maintain both tumors and embryonic stem cells in an undifferentiated state. This raises the intriguing possibility that targeting CDK8 therapeutically may specifically inhibit the stem-like properties of cancer cells.


Sujet(s)
Dédifférenciation cellulaire/physiologie , Cyclin-Dependent Kinase 8/métabolisme , Cellules souches embryonnaires/enzymologie , Tumeurs expérimentales/enzymologie , Tumeurs expérimentales/anatomopathologie , Cellules souches pluripotentes/enzymologie , Animaux , Technique de Western , Lignée cellulaire tumorale , Séparation cellulaire , Cellules souches embryonnaires/cytologie , Cytométrie en flux , Régulation de l'expression des gènes tumoraux/physiologie , Gènes myc , Humains , Immunohistochimie , Souris , Séquençage par oligonucléotides en batterie , Cellules souches pluripotentes/cytologie , Transfection
20.
J Cell Biol ; 191(4): 809-25, 2010 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-21059851

RÉSUMÉ

The ability of progenitor cells to exit the cell cycle is essential for proper embryonic development and homeostasis, but the mechanisms governing cell cycle exit are still not fully understood. Here, we tested the requirement for the retinoblastoma (Rb) protein and its family members p107 and p130 in G0/G1 arrest and differentiation in mammalian cells. We found that Rb family triple knockout (TKO) mouse embryos survive until days 9-11 of gestation. Strikingly, some TKO cells, including in epithelial and neural lineages, are able to exit the cell cycle in G0/G1 and differentiate in teratomas and in culture. This ability of TKO cells to arrest in G0/G1 is associated with the repression of key E2F target genes. Thus, G1 arrest is not always dependent on Rb family members, which illustrates the robustness of cell cycle regulatory networks during differentiation and allows for the identification of candidate pathways to inhibit the expansion of cancer cells with mutations in the Rb pathway.


Sujet(s)
Différenciation cellulaire/physiologie , Phase G1/physiologie , Protéine du rétinoblastome/métabolisme , Animaux , Plan d'organisation du corps/physiologie , Cycle cellulaire/physiologie , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/métabolisme , Cellules cultivées , Embryon de mammifère/anatomie et histologie , Embryon de mammifère/physiologie , Cellules souches embryonnaires/cytologie , Cellules souches embryonnaires/physiologie , Femelle , Analyse de profil d'expression de gènes , Humains , Mâle , Souris , Souris knockout , Neurones/cytologie , Neurones/physiologie , Protéine du rétinoblastome/génétique , Protéine p107 de type rétinoblastome/génétique , Protéine p107 de type rétinoblastome/métabolisme , Protéine p130 de type rétinoblastome/génétique , Protéine p130 de type rétinoblastome/métabolisme , Tératome/métabolisme , Tératome/anatomopathologie , Facteurs de transcription/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE