Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Appl Microbiol Biotechnol ; 101(7): 2747-2766, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28013405

RESUMEN

D2C7-(scdsFv)-PE38KDEL (D2C7-IT) is a novel recombinant Pseudomonas exotoxin A-based immunotoxin (IT), targeting both wild-type epidermal growth factor receptor (EGFRwt) and mutant EGFR variant III (EGFRvIII) proteins overexpressed in glioblastomas. Initial pre-clinical testing demonstrated the anti-tumor efficacy of D2C7-IT against orthotopic glioblastoma xenograft models expressing EGFRwt, EGFRvIII, or both EGFRwt and EGFRvIII. A good laboratory practice (GLP) manufacturing process was developed to produce sufficient material for a phase I/II clinical trial. D2C7-IT was expressed under the control of the T7 promoter in Escherichia coli BLR (λ DE3). D2C7-IT was produced by a 10-L batch fermentation process and was then purified from inclusion bodies using anion exchange, size exclusion, and an endotoxin removal process that achieved a yield of over 300 mg of purified protein. The final vialed batch of D2C7-IT for clinical testing was at a concentration of 0.12 ± 0.1 mg/mL, the pH was at 7.4 ± 0.4, and endotoxin levels were below the detection limit of 10 EU/mL (1.26 EU/mL). The stability of the vialed D2C7-IT has been monitored over a period of 42 months through protein concentration, sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE), isoelectric focusing, size exclusion chromatography, cytotoxicity, sterility, and pH measurements. The vialed D2C7-IT is currently being tested in a phase I/II clinical trial by intratumoral convection-enhanced delivery for 72 h in patients with recurrent glioblastoma (NCT02303678, D2C7 for Adult Patients with Recurrent Malignant Glioma; clinicaltrials.gov ).


Asunto(s)
Inmunotoxinas/metabolismo , ADP Ribosa Transferasas/genética , Adulto , Toxinas Bacterianas/genética , Línea Celular Tumoral , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Estabilidad de Medicamentos , Electroforesis en Gel de Poliacrilamida , Receptores ErbB/genética , Receptores ErbB/metabolismo , Escherichia coli/genética , Exotoxinas/genética , Fermentación , Glioblastoma/tratamiento farmacológico , Humanos , Inmunotoxinas/química , Inmunotoxinas/genética , Inmunotoxinas/uso terapéutico , Control de Calidad , Factores de Virulencia/genética , Exotoxina A de Pseudomonas aeruginosa
2.
Invest New Drugs ; 34(2): 149-58, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26728879

RESUMEN

D2C7-(scdsFv)-PE38KDEL (D2C7-IT) is a novel immunotoxin that reacts with wild-type epidermal growth factor receptor (EGFRwt) and mutant EGFRvIII proteins overexpressed in glioblastomas. This study assessed the toxicity of intracerebral administration of D2C7-IT to support an initial Food and Drug Administration Investigational New Drug application. After the optimization of the formulation and administration, two cohorts (an acute and chronic cohort necropsied on study days 5 and 34) of Sprague-Dawley (SD) rats (four groups of 5 males and 5 females) were infused with the D2C7-IT formulation at total doses of 0, 0.05, 0.1, 0.4 µg (the acute cohort) and 0, 0.05, 0.1, 0.35 µg (the chronic cohort) for approximately 72 h by intracerebral convection-enhanced delivery using osmotic pumps. Mortality was observed in the 0.40 µg (5/10 rats) and 0.35 µg (4/10 rats) high-dose groups of each cohort. Body weight loss and abnormal behavior were only revealed in the rats treated with high doses of D2C7-IT. No dose-related effects were observed in clinical laboratory tests in either cohort. A gross pathologic examination of systemic tissues from the high-dose and control groups in both cohorts exhibited no dose-related or drug-related pathologic findings. Brain histopathology revealed the frequent occurrence of dose-related encephalomalacia, edema, and demyelination in the high-dose groups of both cohorts. In this study, the maximum tolerated dose of D2C7-IT was determined to be between 0.10 and 0.35 µg, and the no-observed-adverse-effect-level was 0.05 µg in SD rats. Both parameters were utilized to design the Phase I/II D2C7-IT clinical trial.


Asunto(s)
Convección , Sistemas de Liberación de Medicamentos , Evaluación Preclínica de Medicamentos , Inmunoconjugados/administración & dosificación , Inmunoconjugados/toxicidad , Inmunotoxinas/administración & dosificación , Inmunotoxinas/toxicidad , Anticuerpos de Cadena Única/administración & dosificación , Anticuerpos de Cadena Única/toxicidad , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Femenino , Concentración 50 Inhibidora , Inyecciones Intraventriculares , Masculino , Ratas Sprague-Dawley
3.
J Immunol ; 193(1): 48-55, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24890727

RESUMEN

Immunogenicity remains the "Achilles' heel" of protein-based therapeutics. Anti-drug Abs produced in response to protein therapeutics can severely limit both the safety and efficacy of this expanding class of agent. In this article, we report that monotherapy of mice with tofacitinib (the JAK inhibitor) quells Ab responses to an immunotoxin derived from the bacterial protein Pseudomonas exotoxin A, as well as to the model Ag keyhole limpet hemocyanin. Thousand-fold reductions in IgG1 titers to both Ags were observed 21 d post immunization. In fact, suppression was evident for all IgG isotypes and IgM. A reduction in IgG3 production was also noted with a thymus-independent type II Ag. Mechanistic investigations revealed that tofacitinib treatment led to reduced numbers of CD127+ pro-B cells. Furthermore, we observed fewer germinal center B cells and the impaired formation of germinal centers of mice treated with tofacitinib. Because normal Ig levels were still present during tofacitinib treatment, this agent specifically reduced anti-drug Abs, thus preserving the potential efficacy of biological therapeutics, including those used as cancer therapeutics.


Asunto(s)
ADP Ribosa Transferasas/farmacología , Formación de Anticuerpos/efectos de los fármacos , Toxinas Bacterianas/farmacología , Exotoxinas/farmacología , Hemocianinas/farmacología , Inmunotoxinas/farmacología , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Pirroles/farmacología , Factores de Virulencia/farmacología , Animales , Linfocitos B/inmunología , Femenino , Centro Germinal/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/inmunología , Ratones , Ratones Endogámicos BALB C , Exotoxina A de Pseudomonas aeruginosa
4.
Blood ; 121(7): 1165-74, 2013 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-23243285

RESUMEN

Immune targeting of B-cell malignancies using chimeric antigen receptors (CARs) is a promising new approach, but critical factors impacting CAR efficacy remain unclear. To test the suitability of targeting CD22 on precursor B-cell acute lymphoblastic leukemia (BCP-ALL), lymphoblasts from 111 patients with BCP-ALL were assayed for CD22 expression and all were found to be CD22-positive, with median CD22 expression levels of 3500 sites/cell. Three distinct binding domains targeting CD22 were fused to various TCR signaling domains ± an IgG heavy chain constant domain (CH2CH3) to create a series of vector constructs suitable to delineate optimal CAR configuration. CARs derived from the m971 anti-CD22 mAb, which targets a proximal CD22 epitope demonstrated superior antileukemic activity compared with those incorporating other binding domains, and addition of a 4-1BB signaling domain to CD28.CD3 constructs diminished potency, whereas increasing affinity of the anti-CD22 binding motif, and extending the CD22 binding domain away from the membrane via CH2CH3 had no effect. We conclude that second-generation m971 mAb-derived anti-CD22 CARs are promising novel therapeutics that should be tested in BCP-ALL.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Receptores de Antígenos de Linfocitos T/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T/inmunología , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores , Lectina 2 Similar a Ig de Unión al Ácido Siálico/inmunología , Animales , Línea Celular Tumoral , Citocinas/metabolismo , Citotoxicidad Inmunológica , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Recombinantes de Fusión/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Int J Cancer ; 132(10): 2339-48, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23115013

RESUMEN

Our study demonstrates the glioma tumor antigen podoplanin to be present at very high levels (>90%) in both glioblastoma (D2159MG, D08-0308MG and D08-0493MG) and medulloblastoma (D283MED, D425MED and DAOY) xenografts and cell line. We constructed a novel recombinant single-chain antibody variable region fragment (scFv), NZ-1, specific for podoplanin from the NZ-1 hybridoma. NZ-1-scFv was then fused to Pseudomonas exotoxin A, carrying a C-terminal KDEL peptide (NZ-1-PE38KDEL). The immunotoxin (IT) was further stabilized by a disulfide (ds) bond between the heavy-chain and light-chain variable regions as the construct NZ-1-(scdsFv)-PE38KDEL. NZ-1-(scdsFv)-PE38KDEL exhibited significant reactivity to glioblastoma and medulloblastoma cells. The affinity of NZ-1-(scdsFv), NZ-1-(scdsFv)-PE38KDEL and NZ-1 antibody for podoplanin peptide was 2.1 × 10(-8) M, 8.0 × 10(-8) M and 3.9 × 10(-10) M, respectively. In a protein stability assay, NZ-1-(scdsFv)-PE38KDEL retained 33-98% of its activity, whereas that of NZ-1-PE38KDEL declined to 13% of its initial levels after incubation at 37°C for 3 days. In vitro cytotoxicity of the NZ-1-(scdsFv)-PE38KDEL was measured in cells isolated from glioblastoma xenografts, D2159MG, D08-0308MG and D08-0493MG, and in the medulloblastoma D283MED, D425MED and DOAY xenografts and cell line. The NZ-1-(scdsFv)-PE38KDEL IT was highly cytotoxic, with an 50% inhibitory concentration in the range of 1.6-29 ng/ml. Significantly, NZ-1-(scdsFv)-PE38KDEL demonstrated tumor growth delay, averaging 24 days (p < 0.001) and 21 days (p < 0.001) in D2159MG and D283MED in vivo tumor models, respectively. Crucially, in the D425MED intracranial tumor model, NZ-1-(scdsFv)-PE38KDEL caused a 41% increase in survival (p ≤ 0.001). In preclinical studies, NZ-1-(scdsFv)-PE38KDEL exhibited significant potential as a targeting agent for malignant brain tumors.


Asunto(s)
ADP Ribosa Transferasas/inmunología , Toxinas Bacterianas/inmunología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/inmunología , Exotoxinas/inmunología , Glioblastoma/tratamiento farmacológico , Glioblastoma/inmunología , Meduloblastoma/tratamiento farmacológico , Meduloblastoma/inmunología , Glicoproteínas de Membrana/inmunología , Anticuerpos de Cadena Única/inmunología , Factores de Virulencia/inmunología , ADP Ribosa Transferasas/uso terapéutico , Toxinas Bacterianas/uso terapéutico , Línea Celular Tumoral , Exotoxinas/uso terapéutico , Femenino , Humanos , Masculino , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/uso terapéutico , Factores de Virulencia/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Exotoxina A de Pseudomonas aeruginosa
6.
Sci Transl Med ; 15(682): eabn5649, 2023 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-36753564

RESUMEN

D2C7-immunotoxin (IT), a dual-specific IT targeting wild-type epidermal growth factor receptor (EGFR) and mutant EGFR variant III (EGFRvIII) proteins, demonstrates encouraging survival outcomes in a subset of patients with glioblastoma. We hypothesized that immunosuppression in glioblastoma limits D2C7-IT efficacy. To improve the response rate and reverse immunosuppression, we combined D2C7-IT tumor cell killing with αCD40 costimulation of antigen-presenting cells. In murine glioma models, a single intratumoral injection of D2C7-IT+αCD40 treatment activated a proinflammatory phenotype in microglia and macrophages, promoted long-term tumor-specific CD8+ T cell immunity, and generated cures. D2C7-IT+αCD40 treatment increased intratumoral Slamf6+CD8+ T cells with a progenitor phenotype and decreased terminally exhausted CD8+ T cells. D2C7-IT+αCD40 treatment stimulated intratumoral CD8+ T cell proliferation and generated cures in glioma-bearing mice despite FTY720-induced peripheral T cell sequestration. Tumor transcriptome profiling established CD40 up-regulation, pattern recognition receptor, cell senescence, and immune response pathway activation as the drivers of D2C7-IT+αCD40 antitumor responses. To determine potential translation, immunohistochemistry staining confirmed CD40 expression in human GBM tissue sections. These promising preclinical data allowed us to initiate a phase 1 study with D2C7-IT+αhCD40 in patients with malignant glioma (NCT04547777) to further evaluate this treatment in humans.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Inmunotoxinas , Humanos , Animales , Ratones , Glioblastoma/patología , Inmunotoxinas/genética , Linfocitos T CD8-positivos , Inmunidad Adaptativa , Receptores ErbB/metabolismo , Línea Celular Tumoral , Neoplasias Encefálicas/terapia
7.
Int J Mol Med ; 24(1): 51-5, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19513534

RESUMEN

The anoctamin (ANO) family of proteins, consisting of 10 members in mammals, are transmembrane proteins that have Ca2+-activated Cl- channel activity. The transmembrane channel-like (TMC) family of proteins, consisting of 8 members in mammals, are also transmembrane proteins of which mutations are implicated in various human conditions, such as hearing loss and epidermodysplasia verruciformis. Here we show that ANO and TMC proteins share high sequence similarity and probably the same membrane topology, indicating that these proteins are evolutionarily related. We found many conserved amino acid residues between the two families of proteins, especially in regions spanning the transmembrane domains TM1, TM4-TM5, and TM6-TM7. These findings imply that these proteins form one large family, which we term ANO/TMC superfamily and that TMC proteins also function as channels for Cl- or other ions. The ANO/TMC superfamily proteins are present in almost all diverse groups of eukaryotic organisms, suggesting that the proteins function in important biological processes, such as ion homeostasis, in eukaryotic cells.


Asunto(s)
Calcio/fisiología , Canales de Cloruro/química , Evolución Molecular , Proteínas de la Membrana/química , Homología de Secuencia de Aminoácido , Secuencia de Aminoácidos , Canales de Cloruro/genética , Humanos , Proteínas de la Membrana/genética , Datos de Secuencia Molecular
8.
PLoS One ; 14(1): e0210608, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30625226

RESUMEN

Standard treatment, unfortunately, yields a poor prognosis for patients with primary or metastatic cancers in the central nervous system, indicating a necessity for novel therapeutic agents. Immunotoxins (ITs) are a class of promising therapeutic candidates produced by fusing antibody fragments with toxin moieties. In this study, we investigated if inherent resistance to IT cytotoxicity can be overcome by rational combination with pro-apoptotic enhancers. Therefore, we combined ITs (9.2.27-PE38KDEL or Mel-14-PE38KDEL) targeting chondroitin sulfate proteoglycan 4 (CSPG4) with a panel of Bcl-2 family inhibitors (ABT-737, ABT-263, ABT-199 [Venetoclax], A-1155463, and S63845) against patient-derived glioblastoma, melanoma, and breast cancer cells/cell lines. In vitro cytotoxicity assays demonstrated that the addition of the ABT compounds, specifically ABT-737, sensitized the different tumors to IT treatment, and improved the IC50 values of 9.2.27-PE38KDEL up to >1,000-fold. Mechanistic studies using 9.2.27-PE38KDEL and ABT-737 revealed that increased levels of intracellular IT, processed (active) exotoxin, and PARP cleavage correlated with the enhanced sensitivity to the combination treatment. Furthermore, we confirmed the synergistic effect of 9.2.27-PE38KDEL and ABT-737 combination therapy in orthotopic GBM xenograft and cerebral melanoma metastasis models in nude mice. Our study defines strategies for overcoming IT resistance and enhancing specific antitumor cytotoxicity in primary and metastatic brain tumors.


Asunto(s)
Antineoplásicos/uso terapéutico , Compuestos de Bifenilo/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/secundario , Inmunotoxinas/uso terapéutico , Nitrofenoles/uso terapéutico , Sulfonamidas/uso terapéutico , Animales , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Benzotiazoles/farmacología , Benzotiazoles/uso terapéutico , Compuestos de Bifenilo/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Sinergismo Farmacológico , Endocitosis/efectos de los fármacos , Exotoxinas/farmacología , Furina/farmacología , Humanos , Inmunotoxinas/farmacología , Isoquinolinas/farmacología , Isoquinolinas/uso terapéutico , Proteínas de la Membrana/metabolismo , Ratones Desnudos , Modelos Biológicos , Nitrofenoles/farmacología , Piperazinas/farmacología , Piperazinas/uso terapéutico , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología , Análisis de Supervivencia , Tiofenos/farmacología , Tiofenos/uso terapéutico , Factores de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Immunother Cancer ; 7(1): 142, 2019 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-31142380

RESUMEN

BACKGROUND: D2C7-IT is a novel immunotoxin (IT) targeting wild-type epidermal growth factor receptor (EGFRwt) and mutant EGFR variant III (EGFRvIII) proteins in glioblastoma. In addition to inherent tumoricidal activity, immunotoxins induce secondary immune responses through the activation of T cells. However, glioblastoma-induced immune suppression is a major obstacle to an effective and durable immunotoxin-mediated antitumor response. We hypothesized that D2C7-IT-induced immune response could be effectively augmented in combination with αCTLA-4/αPD-1/αPD-L1 therapies in murine models of glioma. METHODS: To study this, we overexpressed the D2C7-IT antigen, murine EGFRvIII (dmEGFRvIII), in established glioma lines, CT-2A and SMA560. The reactivity and therapeutic efficacy of D2C7-IT against CT-2A-dmEGFRvIII and SMA560-dmEGFRvIII cells was determined by flow cytometry and in vitro cytotoxicity assays, respectively. Antitumor efficacy of D2C7-IT was examined in immunocompetent, intracranial murine glioma models and the role of T cells was assessed by CD4+ and CD8+ T cell depletion. In vivo efficacy of D2C7-IT/αCTLA-4/αPD-1 monotherapy or D2C7-IT+αCTLA-4/αPD-1 combination therapy was evaluated in subcutaneous unilateral and bilateral CT-2A-dmEGFRvIII glioma-bearing immunocompetent mice. Further, antitumor efficacy of D2C7-IT+αCTLA-4/αPD-1/αPD-L1/αTim-3/αLag-3/αCD73 combination therapy was evaluated in intracranial CT-2A-dmEGFRvIII and SMA560-dmEGFRvIII glioma-bearing mice. Pairwise differences in survival curves were assessed using the generalized Wilcoxon test. RESULTS: D2C7-IT effectively killed CT-2A-dmEGFRvIII (IC50 = 0.47 ng/mL) and SMA560-dmEGFRvIII (IC50 = 1.05 ng/mL) cells in vitro. Treatment of intracranial CT-2A-dmEGFRvIII and SMA560-dmEGFRvIII tumors with D2C7-IT prolonged survival (P = 0.0188 and P = 0.0057, respectively), which was significantly reduced by the depletion of CD4+ and CD8+ T cells. To augment antitumor immune responses, we combined D2C7-IT with αCTLA-4/αPD-1 in an in vivo subcutaneous CT-2A-dmEGFRvIII model. Tumor-bearing mice exhibited complete tumor regressions (4/10 in D2C7-IT+αCTLA-4 and 5/10 in D2C7-IT+αPD-1 treatment groups), and combination therapy-induced systemic antitumor response was effective against both dmEGFRvIII-positive and dmEGFRvIII-negative CT-2A tumors. In a subcutaneous bilateral CT-2A-dmEGFRvIII model, D2C7-IT+αCTLA-4/αPD-1 combination therapies showed dramatic regression of the treated tumors and measurable regression of untreated tumors. Notably, in CT-2A-dmEGFRvIII and SMA560-dmEGFRvIII intracranial glioma models, D2C7-IT+αPD-1/αPD-L1 combinations improved survival, and in selected cases generated cures and protection against tumor re-challenge. CONCLUSIONS: These data support the development of D2C7-IT and immune checkpoint blockade combinations for patients with malignant glioma.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Receptores ErbB/uso terapéutico , Inmunotoxinas/efectos de los fármacos , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Receptores ErbB/farmacología , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL
10.
Gene ; 366(2): 238-45, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16364570

RESUMEN

The POTE family genes encode a highly homologous group of primate-specific proteins that contain ankyrin repeats and coiled coil domains. At least 13 paralogous POTE family genes are found on 8 human chromosomes (2, 8, 13, 14, 15, 18, 21 and 22), which can be sorted into 3 groups based on sequence similarity. We identified by a database search a group of additional human ankyrin repeat domain proteins, of which ANKRD26 and ANKRD30A are the best characterized; these are more distant homologs of POTE family proteins. A comprehensive comparison of the genomic organization indicates that ANKRD26 has the genomic structure of the possible ancestor of ANKRD30A and all POTE family genes. Extensive remodeling involving segmental loss and internal duplication appears to have reshaped the ANKRD30A and POTE family genes after the primal duplication of the ancestor gene. We also identified a mouse homolog of human ANKRD26, but failed to find a mouse homolog that bears the structural characteristics of any of the POTE family of proteins. The mouse Ankrd26 may serve as a useful model for the study of the function of human ANKRD26, ANKRD30A and POTE family proteins.


Asunto(s)
Ancirinas/genética , Cromosomas Humanos/genética , Evolución Molecular , Duplicación de Gen , Animales , Humanos , Ratones , Estructura Terciaria de Proteína/genética , Homología de Secuencia de Aminoácido
11.
Clin Cancer Res ; 19(17): 4717-27, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23857604

RESUMEN

PURPOSE: The EGF receptor gene (EGFR) is most frequently amplified and overexpressed, along with its deletion mutant, EGFRvIII, in glioblastoma. We tested the preclinical efficacy of the recombinant immunotoxin, D2C7-(scdsFv)-PE38KDEL, which is reactive with a 55-amino acid (AA) region present in the extracellular domain of both EGFRwt (583-637 AAs) and EGFRvIII (292-346 AAs) proteins. EXPERIMENTAL DESIGN: The binding affinity and specificity of D2C7-(scdsFv)-PE38KDEL for EGFRwt and EGFRvIII were measured by surface-plasmon resonance and flow cytometry. In vitro cytotoxicity of D2C7-(scdsFv)-PE38KDEL was measured by inhibition of protein synthesis in human EGFRwt-transfected NR6 (NR6W), human EGFRvIII-transfected NR6 (NR6M), EGFRwt-overexpressing A431-epidermoid-carcinoma, and glioblastoma xenograft cells (43, D08-0493MG, D2159MG, and D270MG). In vivo antitumor efficacy of D2C7-(scdsFv)-PE38KDEL was evaluated using 43, NR6M, and D270MG orthotopic tumor models. RESULTS: The KD of D2C7-(scdsFv)-PE38KDEL for EGFRwt and EGFRvIII was 1.6×10(-9) mol/L and 1.3×10(-9) mol/L, respectively. Flow cytometry with NR6W and NR6M cells confirmed the specificity of D2C7-(scdsFv)-PE38KDEL for EGFRwt and EGFRvIII. The D2C7-(scdsFv)-PE38KDEL IC50 was 0.18 to 2.5 ng/mL on cells expressing EGFRwt (NR6W, A431, 43, and D08-0493MG). The D2C7-(scdsFv)-PE38KDEL IC50 was approximately 0.25 ng/mL on EGFRvIII-expressing cells (NR6M) and on EGFRwt- and EGFRvIII-expressing glioblastoma xenograft cells (D2159MG and D270MG). Significantly, in intracranial tumor models of 43, NR6M, and D270MG, treatment with D2C7-(scdsFv)-PE38KDEL by convection-enhanced delivery prolonged survival by 310% (P=0.006), 28% (P=0.002), and 166% (P=0.001), respectively. CONCLUSIONS: In preclinical studies, the D2C7-(scdsFv)-PE38KDEL immunotoxin exhibited significant potential for treating brain tumors expressing EGFRwt, EGFRvIII, or both.


Asunto(s)
Neoplasias Encefálicas/inmunología , Receptores ErbB/inmunología , Glioblastoma/inmunología , Inmunotoxinas/administración & dosificación , Antígenos de Neoplasias/inmunología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , Epítopos/aislamiento & purificación , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Citometría de Flujo , Glioblastoma/patología , Glioblastoma/terapia , Humanos , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Inmunotoxinas/genética , Resonancia por Plasmón de Superficie , Ensayos Antitumor por Modelo de Xenoinjerto
12.
MAbs ; 5(5): 748-62, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23924792

RESUMEN

About 60 percent of glioblastomas highly express the gangliosides 3'-isoLM1 and 3',6'-isoLD1 on the cell surface, providing ideal targets for brain tumor immunotherapy. A novel recombinant immunotoxin, DmAb14m-(scFv)-PE38KDEL (DmAb14m-IT), specific for the gangliosides 3'-isoLM1 and 3',6'-isoLD1, was constructed with improved affinity and increased cytotoxicity for immunotherapeutic targeting of glioblastoma. We isolated an scFv parental clone from a previously established murine hybridoma, DmAb14, that is specific to both 3'-isoLM1 and 3',6'-isoLD1. We then performed in vitro affinity maturation by CDR hotspot random mutagenesis. The binding affinity and specificity of affinity-matured DmAb14m-IT were measured by surface-plasmon resonance, flow cytometry, and immunohistochemical analysis. In vitro cytotoxicity of DmAb14m-IT was measured by protein synthesis inhibition and cell death assays in human cell lines expressing gangliosides 3'-isoLM1 and 3',6'-isoLD1 (D54MG and D336MG) and xenograft-derived cells (D2224MG). As a result, the KD of DmAb14m-IT for gangliosides 3'-isoLM1 and 3',6'-isoLD1 was 2.6 × 10(-9)M. Also, DmAb14m-IT showed a significantly higher internalization rate in cells expressing 3'-isoLM1 and 3',6'-isoLD1. The DmAb14m-IT IC 50 was 80 ng/mL (1194 pM) on the D54MG cell line, 5 ng/ml (75 pM) on the D336MG cell line, and 0.5 ng/ml (7.5 pM) on the D2224MG xenograft-derived cells. There was no cytotoxicity on ganglioside-negative HEK293 cells. Immunohistochemical analysis confirmed the specific apparent affinity of DmAb14m-IT with 3'-isoLM1 and 3',6'-isoLD1. In conclusion, DmAb14m-IT showed specific binding affinity, a significantly high internalization rate, and selective cytotoxicity on glioma cell lines and xenograft-derived cells expressing 3'-isoLM1 and 3',6'-isoLD1, thereby displaying robust therapeutic potential for testing the antitumor efficacy of DmAb14m-IT at the preclinical level and eventually in the clinical setting.


Asunto(s)
Neoplasias Encefálicas/inmunología , Gangliósidos/inmunología , Glioma/inmunología , Inmunotoxinas/inmunología , Secuencia de Aminoácidos , Animales , Afinidad de Anticuerpos/inmunología , Especificidad de Anticuerpos/inmunología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , Supervivencia Celular/inmunología , Regiones Determinantes de Complementariedad/genética , Regiones Determinantes de Complementariedad/inmunología , Citometría de Flujo , Glioma/patología , Glioma/terapia , Células HEK293 , Xenoinjertos , Humanos , Inmunohistoquímica , Inmunoterapia/métodos , Inmunotoxinas/genética , Inmunotoxinas/uso terapéutico , Ratones , Datos de Secuencia Molecular , Mutación , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/uso terapéutico , Homología de Secuencia de Aminoácido , Resonancia por Plasmón de Superficie
14.
Cancer Immunol Immunother ; 57(1): 115-21, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17634939

RESUMEN

PURPOSE: Immunotoxins as anti-cancer therapeutics have several potential advantages over conventional agents including a high specificity, extraordinary potency, and a lack of an identified mechanism for resistance. It has been clearly demonstrated that Pseudomonas-based immunotoxins have a direct cytotoxic effect. However, delayed and often dramatic antitumor responses seen in human studies with targeted toxins led us to hypothesize that immunologic responses may be a secondary mechanism that enhances the therapeutic efficacy of these novel drugs. EXPERIMENTAL DESIGN: This hypothesis was tested in a murine system using an immunotoxin, MR1-1 [MR1-1(dsFv)-PE38KDEL], that targets a syngeneic murine homologue of the tumor-specific human epidermal growth factor mutation, EGFRvIII, expressed on a murine cell line. RESULTS: Intratumoral treatment with MR1-1 eliminated EGFRvIII-expressing tumors (P < 0.0001). The antitumor activity of MR1-1 was dependent on the expression of EGFRvIII on some, but not all tumors cells, and was significantly inhibited in the absence of CD4+ (P = 0.0193) and CD8+ (P = 0.0193) T cells. MR1-1 induced EGFRvIII-specific immunity (P < 0.0005) and produced long lasting immunity against tumors expressing EGFRvIII as well as EGFRvIII-negative tumors. CONCLUSIONS: These data suggest that immunotoxins may not be strictly dependent on direct cytotoxicity for their efficacy, but may also be potent inducers of antitumor immunity active even against cells that do not express the targeted antigen.


Asunto(s)
Antineoplásicos/uso terapéutico , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Receptores ErbB/antagonistas & inhibidores , Inmunotoxinas/uso terapéutico , Neoplasias Experimentales/tratamiento farmacológico , Animales , Línea Celular Tumoral , Femenino , Humanos , Depleción Linfocítica , Ratones
15.
Proc Natl Acad Sci U S A ; 102(22): 7946-51, 2005 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-15905329

RESUMEN

Because CD30 is highly expressed on Hodgkin's lymphoma and anaplastic large cell lymphoma, it is a promising target for immunotherapy. Soluble CD30, the extracellular domain of CD30 that is shed from the cells, can reduce the effects of CD30-targeting agents by competitive binding. In this study, we identified two epitopes on membrane-associated CD30 that are missing on soluble CD30 probably because of a conformational change upon shedding. These epitopes are potentially superior targets for immunotherapy because targeting them should be free from the competitive effects of soluble CD30. We studied 27 anti-native CD30 mAbs that were assigned to 8 different topographical epitopes. Soluble CD30 was prepared from culture supernatants of L540 cells or Karpas 299 cells. In an ELISA, the mAbs to two epitopes, Ep2 (amino acids 107-153) and Ep7 (amino acids 282-338), showed less than a 2% average cross-reactivity to soluble CD30 compared with a CD30-Fc fusion protein. In addition, these mAbs bound to CD30 on cells in the presence of an excess of soluble CD30. These epitopes (Ep2 and Ep7) are, therefore, more efficiently presented on cell-associated CD30 than on soluble CD30 (membrane-specific epitopes). Also, soluble CD30 in the sera of mice bearing L540 tumors did not form immune complexes with the membrane-specific mAbs analyzed by size-exclusion chromatography. In contrast, mAbs to the other epitopes reacted with both soluble CD30 and membrane CD30. Our results suggest that it may be possible to find membrane-specific epitopes on other immunotherapy target molecules.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Epítopos/metabolismo , Inmunoterapia/métodos , Antígeno Ki-1/inmunología , Antígeno Ki-1/metabolismo , Linfoma/terapia , Animales , Western Blotting , Línea Celular Tumoral , Cromatografía en Gel , Reacciones Cruzadas/inmunología , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunoglobulina G/metabolismo , Ratones
16.
Biochem Biophys Res Commun ; 303(3): 842-7, 2003 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-12670487

RESUMEN

Copines are ubiquitously expressed, phospholipid-binding proteins that have been conserved through evolution. In this paper, we report the cloning and molecular characterization of a new member of the Copine family, Copine 8. This gene has been isolated and characterized using a combination of bioinformatic and experimental approaches. Using an algorithm to cluster ESTs (expressed sequence tags) that are available through the public "GoldenPath" database, Copine 8 was initially identified as a gene predominantly expressed in prostate and testis. Cloning and molecular analysis revealed that this gene is expressed in low-levels in most tissues examined. Two different isoforms of this gene have been isolated. Strongest expression of Copine 8 mRNA is seen in the prostate, heart, and brain. Taken together, this data suggest that Copine 8 may have an important role to play in prostate regulation and development.


Asunto(s)
Proteínas Portadoras/genética , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Clonación Molecular , ADN Complementario/genética , Etiquetas de Secuencia Expresada , Femenino , Expresión Génica , Humanos , Técnicas In Vitro , Masculino , Datos de Secuencia Molecular , Familia de Multigenes , Embarazo , Próstata/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Conejos , Homología de Secuencia de Aminoácido , Testículo/metabolismo , Distribución Tisular
17.
Biochem Biophys Res Commun ; 312(4): 1209-15, 2003 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-14652002

RESUMEN

We have combined computer-based screening and experimental expression analysis to identify genes that are expressed in normal prostate and/or prostate cancer but not in essential human tissues. Using this approach we identified a new gene that is specifically expressed in testis, prostate, and placenta. The gene has one major transcript of 1.0kb in size and encodes for a protein of 30.7kDa molecular weight. We named this gene TEPP (expressed in testis, prostate, and placenta). The amino acid sequence analysis of TEPP using SignalP program shows that it has a signal peptide with a predicted cleavage site between amino acids 19 and 20, indicating that it might be a secreted protein. Analysis of the predicted TEPP orthologs from different species shows that these proteins are highly conserved in chordates. In addition we have identified a splice variant of TEPP, which encodes a 37kDa protein. In conclusion, a combination of bioinformatic and molecular approaches is useful in the identification of genes expressed in specific tissues. Selective expression of TEPP in testis, prostate, and in placenta and its high conservation among different species indicate that TEPP might have a role in reproductive biology.


Asunto(s)
Secuencia Conservada/genética , Regulación de la Expresión Génica/genética , Placenta/química , Próstata/química , Proteínas/química , Proteínas/genética , Análisis de Secuencia de Proteína/métodos , Testículo/química , Secuencia de Aminoácidos , Animales , Cordados , Ciona intestinalis , Clonación Molecular , Etiquetas de Secuencia Expresada , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Peso Molecular , Especificidad de Órganos , Placenta/metabolismo , Proteínas Gestacionales/química , Proteínas Gestacionales/clasificación , Proteínas Gestacionales/genética , Próstata/metabolismo , Proteínas/clasificación , Ratas , Alineación de Secuencia/métodos , Homología de Secuencia de Aminoácido , Especificidad de la Especie , Porcinos , Testículo/metabolismo , Xenopus laevis
18.
Proc Natl Acad Sci U S A ; 101(36): 13257-61, 2004 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-15326299

RESUMEN

Chromosomal rearrangements resulting in gene fusions are frequently involved in carcinogenesis. Here, we describe a semiautomatic procedure for identifying fusion gene transcripts by using publicly available mRNA and EST databases. With this procedure, we have identified 96 transcript sequences that are derived from 60 known fusion genes. Also, 47 or more additional sequences appear to be derived from 20 or more previously unknown putative fusion genes. We have experimentally verified the presence of a previously unknown IRA1/RGS17 fusion in the breast cancer cell line MCF7. The fusion gene encodes the full-length RGS17 protein, a regulator of G protein-coupled signaling, under the control of the IRA1 gene promoter. This study demonstrates that databases of ESTs can be used to discover fusion genes resulting from structural rearrangement of chromosomes.


Asunto(s)
Aberraciones Cromosómicas , Etiquetas de Secuencia Expresada , Proteínas de Fusión Oncogénica/genética , Línea Celular Tumoral , Bases de Datos como Asunto , Femenino , Humanos , Hibridación Fluorescente in Situ , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA