Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Immunol Cell Biol ; 97(6): 526-537, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30680791

RESUMEN

Human IgG1 and IgG3 antibodies (Abs) can mediate Ab-dependent cellular cytotoxicity (ADCC), and engineering of the Ab Fc (point mutation; defucosylation) has been shown to affect ADCC by modulating affinity for FcRγIIIa. In the absence of a CH 1 domain, many camelid heavy-chain Abs (HCAbs) naturally bear very long and flexible hinge regions connecting their VH H and CH 2 domains. To better understand the influence of hinge length and structure on HCAb ADCC, we produced a series of hinge-engineered epidermal growth factor receptor (EGFR)-specific chimeric camelid VH H-human Fc Abs and characterized their affinities for recombinant EGFR and FcRγIIIa, their binding to EGFR-positive tumor cells, and their ability to elicit ADCC. In the case of one chimeric HCAb (EG2-hFc), we found that variants bearing longer hinges (IgG3 or camelid hinge regions) showed dramatically improved ADCC in comparison with a variant bearing the human IgG1 hinge, in similar fashion to a variant with reduced CH 2 fucosylation. Conversely, an EG2-hFc variant bearing a truncated human IgG1 upper hinge region failed to elicit ADCC. However, there was no consistent association between hinge length and ADCC for four similarly engineered chimeric HCAbs directed against distinct EGFR epitopes. These findings demonstrate that the ADCC of some HCAbs can be modulated simply by varying the length of the Ab hinge. Although this effect appears to be heavily epitope-dependent, this strategy may be useful to consider during the design of VH H-based therapeutic Abs for cancer.


Asunto(s)
Adenocarcinoma/terapia , Anticuerpos Monoclonales/metabolismo , Neoplasias de la Mama/terapia , Inmunoterapia/métodos , Proteínas Recombinantes de Fusión/genética , Adenocarcinoma/inmunología , Animales , Anticuerpos Monoclonales/genética , Afinidad de Anticuerpos , Citotoxicidad Celular Dependiente de Anticuerpos , Neoplasias de la Mama/inmunología , Camelidae , Línea Celular Tumoral , Receptores ErbB/inmunología , Receptores ErbB/metabolismo , Femenino , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoglobulina G/genética , Cadenas Pesadas de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/genética , Mutación/genética , Unión Proteica , Ingeniería de Proteínas
2.
Mol Ther Oncol ; 32(1): 200775, 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38596311

RESUMEN

Chimeric antigen receptor (CAR) T cell therapies targeting B cell-restricted antigens CD19, CD20, or CD22 can produce potent clinical responses for some B cell malignancies, but relapse remains common. Camelid single-domain antibodies (sdAbs or nanobodies) are smaller, simpler, and easier to recombine than single-chain variable fragments (scFvs) used in most CARs, but fewer sdAb-CARs have been reported. Thus, we sought to identify a therapeutically active sdAb-CAR targeting human CD22. Immunization of an adult Llama glama with CD22 protein, sdAb-cDNA library construction, and phage panning yielded >20 sdAbs with diverse epitope and binding properties. Expressing CD22-sdAb-CAR in Jurkat cells drove varying CD22-specific reactivity not correlated with antibody affinity. Changing CD28- to CD8-transmembrane design increased CAR persistence and expression in vitro. CD22-sdAb-CAR candidates showed similar CD22-dependent CAR-T expansion in vitro, although only membrane-proximal epitope targeting CD22-sdAb-CARs activated direct cytolytic killing and extended survival in a lymphoma xenograft model. Based on enhanced survival in blinded xenograft studies, a lead CD22sdCAR-T was selected, achieving comparable complete responses to a benchmark short linker m971-scFv CAR-T in high-dose experiments. Finally, immunohistochemistry and flow cytometry confirm tissue and cellular-level specificity of the lead CD22-sdAb. This presents a complete report on preclinical development of a novel CD22sdCAR therapeutic.

3.
PLoS One ; 18(6): e0273884, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37347762

RESUMEN

Bi-specific T-cell engager antibodies (BiTEs) are synthetic fusion molecules that combine multiple antibody-binding domains to induce active contact between T-cells and antigen expressing cells in the body. Blinatumomab, a CD19-CD3 BiTE is now a widely used therapy for relapsed B-cell malignancies, and similar BiTE therapeutics have shown promise for treating various other forms of cancer. The current process for new BiTE development is time consuming and costly, requiring characterization of the individual antigen binding domains, followed by bi-specific design, protein production, purification, and eventually functional screening. Here, we sought to establish a more cost-efficient approach for generating novel BiTE sequences and assessing bioactivity through a function first approach without purification. We generate a plasmid with a bi-modular structure to allow high-throughput exchange of either binding arm, enabling rapid screening of novel tumour-targeting single chain variable (scFv) domains in combination with the well-characterized OKT3 scFv CD3-targeting domain. We also demonstrate two systems for high throughput functional screening of BiTE proteins based on Jurkat T cells (referred to as BiTE-J). Using BiTE-J we evaluate four EGFRvIII-scFv sequenced in BiTE format, identifying two constructs with superior activity for redirecting T-cells against the EGFRvIII-tumour specific antigen. We also confirm activity in primary T cells, where novel EGFRvIII-BiTEs induced T cell activation and antigen selective tumor killing. We finally demonstrate similar exchange the CD3-interacting element of our bi-modular plasmid. By testing several novel CD3-targeting scFv elements for activity in EGFRvIII-targeted BiTEs, we were able to identify highly active BiTE molecules with desirable functional activity for downstream development. In summary, BiTE-J presents a low cost, high-throughput method for the rapid assessment of novel BiTE molecules without the need for purification and quantification.


Asunto(s)
Anticuerpos Biespecíficos , Recurrencia Local de Neoplasia , Humanos , Complejo CD3/metabolismo , Anticuerpos Biespecíficos/farmacología , Células Jurkat , Antígenos de Neoplasias , Linfocitos B/metabolismo
4.
Front Immunol ; 13: 864868, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35935988

RESUMEN

Epidermal growth factor family receptor (EGFR) is commonly overexpressed in many solid tumors and an attractive target for chimeric antigen receptor (CAR)-T therapy, but as EGFR is also expressed at lower levels in healthy tissues a therapeutic strategy must balance antigenic responsiveness against the risk of on-target off-tumor toxicity. Herein, we identify several camelid single-domain antibodies (also known as nanobodies) that are effective EGFR targeting moieties for CARs (EGFR-sdCARs) with very strong reactivity to EGFR-high and EGFR-low target cells. As a strategy to attenuate their potent antigenic sensitivity, we performed progressive truncation of the human CD8 hinge commonly used as a spacer domain in many CAR constructs. Single amino acid hinge-domain truncation progressively decreased both EGFR-sdCAR-Jurkat cell binding to EGFR-expressing targets and expression of the CD69 activation marker. Attenuated signaling in hinge-truncated EGFR-sdCAR constructs increased selectivity for antigen-dense EGFR-overexpressing cells over an EGFR-low tumor cell line or healthy donor derived EGFR-positive fibroblasts. We also provide evidence that epitope location is critical for determining hinge-domain requirement for CARs, as hinge truncation similarly decreased antigenic sensitivity of a membrane-proximal epitope targeting HER2-CAR but not a membrane-distal EGFRvIII-specific CAR. Hinge-modified EGFR-sdCAR cells showed clear functional attenuation in Jurkat-CAR-T cells and primary human CAR-T cells from multiple donors in vitro and in vivo. Overall, these results indicate that hinge length tuning provides a programmable strategy for throttling antigenic sensitivity in CARs targeting membrane-proximal epitopes, and could be employed for CAR-optimization and improved tumor selectivity.


Asunto(s)
Receptores Quiméricos de Antígenos , Anticuerpos de Dominio Único , Epítopos , Receptores ErbB , Humanos , Inmunoterapia Adoptiva/métodos , Receptor ErbB-2/genética , Receptores Quiméricos de Antígenos/genética , Linfocitos T
5.
Mol Ther Methods Clin Dev ; 16: 238-254, 2020 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-32083149

RESUMEN

Chimeric antigen receptor (CAR) development involves extensive empirical characterization of antigen-binding domain (ABD)/CAR constructs for clinical suitability. Here, we present a cost-efficient and rapid method for evaluating CARs in human Jurkat T cells. Using a modular CAR plasmid, a highly efficient ABD cloning strategy, plasmid electroporation, short-term co-culture, and flow-cytometric detection of CD69, this assay (referred to as CAR-J) evaluates sensitivity and specificity for ABDs. Assessing 16 novel anti-CD22 single-chain variable fragments derived from mouse monoclonal antibodies, CAR-J stratified constructs by response magnitude to CD22-expressing target cells. We also characterized 5 novel anti-EGFRvIII CARs for preclinical development, identifying candidates with varying tonic and target-specific activation characteristics. When evaluated in primary human T cells, tonic/auto-activating (without target cells) EGFRvIII-CARs induced target-independent proliferation, differentiation toward an effector phenotype, elevated activity against EGFRvIII-negative cells, and progressive loss of target-specific response upon in vitro re-challenge. These EGFRvIII CAR-T cells also showed anti-tumor activity in xenografted mice. In summary, CAR-J represents a straightforward method for high-throughput assessment of CAR constructs as genuine cell-associated antigen receptors that is particularly useful for generating large specificity datasets as well as potential downstream CAR optimization.

7.
J Neuroimmunol ; 139(1-2): 119-32, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12799029

RESUMEN

Human gammadelta T cells are an integral part of the innate immune system and have been difficult to study owing primarily to their relatively low abundance and their fastidious culture properties associated with short in vitro lifespan. Their increased presence within multiple sclerosis (MS) white matter plaques compared to peripheral blood (PB) suggests a specific interaction with central nervous system (CNS) tissues. This fact, together with their innate ability to lyse human oligodendrocytes in culture implicate them possibly in the pathogenesis of MS. To further investigate their potential role in MS, we studied whether gammadelta T cells could be effectively immortalized using Herpesvirus saimiri (HVS), so that they could be studied in longer-term cultures. Effective culture conditions were established resulting in efficient HVS growth transformation of multiple PB and CSF gammadelta T cell lines and clones that could exist in IL-2-dependent culture for periods in excess of 2 years. Phenotypic and functional comparison studies with parental nontransformed gammadelta T cells were performed to characterize the changes that possibly induced by viral transformation. Using panels of transformed gammadelta T cell clones representing discrete gammadelta TcR subtypes, there was no apparent correlation between intracytoplasmic cytokine expression or tumor cell cytotoxicity with a specific TcR. All transformed gammadelta T cells analyzed, regardless of their compartment of origin, strongly expressed intracytoplasmic IFN-gamma and TNF-alpha, but little IL-2 or anti-inflammatory IL-4 or IL-10. These results indicate that HVS transformation of gammadelta T cells can be used to generate lines and clones from both the CSF and PB compartments for further study and elucidation of their potential role in MS pathogenesis.


Asunto(s)
Línea Celular Transformada/virología , Vectores Genéticos , Herpesvirus Saimiriino 2/inmunología , Esclerosis Múltiple/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/virología , Técnicas de Cultivo de Célula , Línea Celular Transformada/citología , Línea Celular Transformada/inmunología , Células Clonales/efectos de los fármacos , Células Clonales/inmunología , Citotoxinas/farmacología , Vectores Genéticos/inmunología , Humanos , Interferón gamma/metabolismo , Interleucina-2/farmacología , Esclerosis Múltiple/sangre , Esclerosis Múltiple/líquido cefalorraquídeo , Fenotipo , Linfocitos T/citología , Linfocitos T/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
8.
Glycobiology ; 17(3): 249-60, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17172262

RESUMEN

The inherent promiscuity of the polysialic acid (PSA) biosynthetic pathway has been exploited by the use of exogenous unnatural sialic acid precursor molecules to introduce unnatural modifications into cellular PSA, and has found applications in nervous system development and tumor vaccine studies. The sialic acid precursor molecules N-propionyl- and N-butanoyl-mannosamine (ManPr, ManBu) have been variably reported to affect PSA biosynthesis ranging from complete inhibition to de novo production of modified PSA, thus illustrating the need for further investigation into their effects. In this study, we have used a monoclonal antibody (mAb) 13D9, specific to both N-propionyl-PSA and N-butanoyl-PSA (NPrPSA and NBuPSA), together with flow cytometry, to study precursor-treated tumor cells and NT2 neurons at different stages of their maturation. We report that both ManPr and ManBu sialic acid precursors are metabolized and the resultant unnatural sialic acids are incorporated into de novo surface sialylglycoconjugates in murine and human tumor cells and, for the first time, in human NT2 neurons. Furthermore, neither precursor treatment deleteriously affected endogenous PSA expression; however, with NT2 cells, PSA levels were naturally downregulated as a function of their maturation into polarized neurons independent of sialic acid precursor treatment.


Asunto(s)
Neuronas/metabolismo , Polisacáridos Bacterianos/biosíntesis , Polisacáridos/biosíntesis , Ácidos Siálicos/biosíntesis , Animales , Anticuerpos Monoclonales/inmunología , Ingeniería Biomédica/métodos , Línea Celular Tumoral , Membrana Celular/química , Regulación hacia Abajo , Citometría de Flujo , Hexosaminas/metabolismo , Hexosaminas/farmacología , Humanos , Ratones , Ácido N-Acetilneuramínico/biosíntesis , Neuronas/efectos de los fármacos , Neuronas/ultraestructura , Polisacáridos/análisis , Polisacáridos/inmunología , Polisacáridos Bacterianos/análisis , Polisacáridos Bacterianos/inmunología
9.
J Biol Chem ; 279(24): 25390-9, 2004 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-15047693

RESUMEN

N-Propionyl, N-butyryl (N-Bu), and N-benzoyl mannosamine, as precursors of sialic acid biosynthesis, were incubated with human melanoma SK-MEL-28 cells and resulted in the replacement of N-acetyl groups on the cell surface sialic acid residues, including those associated with GD3. Meanwhile, vaccines containing GD3 and modified GD3 tetrasaccharide-keyhole limpet hemocyanin conjugates were synthesized, and BALB/c mice were immunized with them together with monophosphoryl lipid A adjuvant. The GD3Bu-keyhole limpet hemocyanin conjugate raised the highest IgG titers without any cross-reactivity to unmodified GD3. Expression of GD3Bu epitopes on the surface of SK-MEL-28 cells was confirmed in vitro and in vivo by the binding of a polyclonal antiserum and monoclonal antibody (mAb) 2A, both of which specifically recognize GD3Bu, and by mass spectroscopic analysis of glycolipids extracted from cells. Following expression of GD3Bu on the surface of SK-MEL-28 cells, the cells could be lysed by mAb 2A and GD3Bu antiserum in the presence of complement. Although less effective in the control of existing large size tumors ( approximately 10 mm inner diameter) on BALB/c nu/nu mice, mAb 2A in combination with ManNBu effectively protected mice from SK-MEL-28 tumor grafting. This approach may provide a method to augment the immunogenicity of sialylated human antigens and to avoid generating an autoimmune response to them at same time.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Gangliósidos/inmunología , Melanoma/terapia , Animales , Anticuerpos Monoclonales/inmunología , Ingeniería Biomédica , Línea Celular Tumoral , Femenino , Glicoconjugados/inmunología , Hemocianinas/inmunología , Humanos , Inmunización , Melanoma/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Vacunas Conjugadas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA