Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Leukemia ; 37(10): 2006-2016, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37634013

RESUMEN

Patients harboring CRLF2-rearranged B-lineage acute lymphocytic leukemia (B-ALL) face a 5-year survival rate as low as 20%. While significant gains have been made to position targeted therapies for B-ALL treatment, continued efforts are needed to develop therapeutic options with improved duration of response. Here, first we have demonstrated that patients with CRLF2-rearranged Ph-like ALL harbor elevated thymic stromal lymphopoietin receptor (TSLPR) expression, which is comparable with CD19. Then we present and evaluate the anti-tumor characteristics of 1B7/CD3, a novel CD3-redirecting bispecific antibody (BsAb) that co-targets TSLPR. In vitro, 1B7/CD3 exhibits optimal binding to both human and cynomolgus CD3 and TSLPR. Further, 1B7/CD3 was shown to induce potent T cell activation and tumor lytic activity in both cell lines and primary B-ALL patient samples. Using humanized cell- or patient-derived xenograft models, 1B7/CD3 treatment was shown to trigger dose-dependent tumor remission or growth inhibition across donors as well as induce T cell activation and expansion. Pharmacokinetic studies in murine models revealed 1B7/CD3 to exhibit a prolonged half-life. Finally, toxicology studies using cynomolgus monkeys found that the maximum tolerated dose of 1B7/CD3 was ≤1 mg/kg. Overall, our preclinical data provide the framework for the clinical evaluation of 1B7/CD3 in patients with CRLF2-rearranged B-ALL.


Asunto(s)
Anticuerpos Biespecíficos , Linfoma de Células B , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Humanos , Animales , Ratones , Complejo CD3 , Anticuerpos Biespecíficos/farmacología , Anticuerpos Biespecíficos/uso terapéutico , Linfoma de Células B/tratamiento farmacológico , Antígenos CD19 , Línea Celular , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Receptores de Citocinas
3.
J Immunother Cancer ; 9(7)2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34326171

RESUMEN

BACKGROUND: Acute myeloid leukemia (AML) stem cells (LSCs) are capable of surviving current standard chemotherapy and are the likely source of deadly, relapsed disease. While stem cell transplant serves as proof-of-principle that AML LSCs can be eliminated by the immune system, the translation of existing immunotherapies to AML has been met with limited success. Consequently, understanding and exploiting the unique immune-evasive mechanisms of AML LSCs is critical. METHODS: Analysis of stem cell datasets and primary patient samples revealed CD200 as a putative stem cell-specific immune checkpoint overexpressed in AML LSCs. Isogenic cell line models of CD200 expression were employed to characterize the interaction of CD200+ AML with various immune cell subsets both in vitro and in peripheral blood mononuclear cell (PBMC)-humanized mouse models. CyTOF and RNA-sequencing were performed on humanized mice to identify novel mechanisms of CD200-mediated immunosuppression. To clinically translate these findings, we developed a fully humanized CD200 antibody (IgG1) that removed the immunosuppressive signal by blocking interaction with the CD200 receptor while also inducing a potent Fc-mediated response. Therapeutic efficacy of the CD200 antibody was evaluated using both humanized mice and patient-derived xenograft models. RESULTS: Our results demonstrate that CD200 is selectively overexpressed in AML LSCs and is broadly immunosuppressive by impairing cytokine secretion in both innate and adaptive immune cell subsets. In a PBMC-humanized mouse model, CD200+ leukemia progressed rapidly, escaping elimination by T cells, compared with CD200- AML. T cells from mice with CD200+ AML were characterized by an abundance of metabolically quiescent CD8+ central and effector memory cells. Mechanistically, CD200 expression on AML cells significantly impaired OXPHOS metabolic activity in T cells from healthy donors. Importantly, CD200 antibody therapy could eliminate disease in the presence of graft-versus-leukemia in immune competent mice and could significantly improve the efficacy of low-intensity azacitidine/venetoclax chemotherapy in immunodeficient hosts. CONCLUSIONS: Overexpression of CD200 is a stem cell-specific marker that contributes to immunosuppression in AML by impairing effector cell metabolism and function. CD200 antibody therapy is capable of simultaneously reducing CD200-mediated suppression while also engaging macrophage activity. This study lays the groundwork for CD200-targeted therapeutic strategies to eliminate LSCs and prevent AML relapse.


Asunto(s)
Antígenos CD/metabolismo , Evasión Inmune/genética , Leucemia Mieloide Aguda/genética , Animales , Humanos , Ratones , Ratones Endogámicos NOD
4.
Front Immunol ; 9: 3153, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30713535

RESUMEN

Despite substantial advances in the treatment of acute myeloid leukemia (AML), only 30% of patients survive more than 5 years. Therefore, new therapeutics are much needed. Here, we present a novel therapeutic strategy targeting PR1, an HLA-A2 restricted myeloid leukemia antigen. Previously, we have developed and characterized a novel T-cell receptor-like monoclonal antibody (8F4) that targets PR1/HLA-A2 and eliminates AML xenografts by antibody-dependent cellular cytotoxicity (ADCC). To improve the potency of 8F4, we adopted a strategy to link T-cell cytotoxicity with a bi-specific T-cell-engaging antibody that binds PR1/HLA-A2 on leukemia and CD3 on neighboring T-cells. The 8F4 bi-specific antibody maintained high affinity and specific binding to PR1/HLA-A2 comparable to parent 8F4 antibody, shown by flow cytometry and Bio-Layer Interferometry. In addition, 8F4 bi-specific antibody activated donor T-cells in the presence of HLA-A2+ primary AML blasts and cell lines in a dose dependent manner. Importantly, activated T-cells lysed HLA-A2+ primary AML blasts and cell lines after addition of 8F4 bi-specific antibody. In conclusion, our studies demonstrate the therapeutic potential of a novel bi-specific antibody targeting the PR1/HLA-A2 leukemia-associated antigen, justifying further clinical development of this strategy.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Antígenos de Neoplasias/inmunología , Antígeno HLA-A2/inmunología , Leucemia Mieloide Aguda/inmunología , Linfocitos T/inmunología , Animales , Anticuerpos Biespecíficos/farmacología , Especificidad de Anticuerpos/inmunología , Antígenos de Neoplasias/metabolismo , Células CHO , Línea Celular , Cricetulus , Citotoxicidad Inmunológica , Antígeno HLA-A2/metabolismo , Humanos , Inmunoterapia Adoptiva , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/terapia , Activación de Linfocitos , Unión Proteica , Linfocitos T/metabolismo
5.
J Biol Chem ; 292(24): 10295-10305, 2017 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-28468826

RESUMEN

Neutrophil elastase (NE) can be rapidly taken up by tumor cells that lack endogenous NE expression, including breast cancer, which results in cross-presentation of PR1, an NE-derived HLA-A2-restricted peptide that is an immunotherapy target in hematological and solid tumor malignancies. The mechanism of NE uptake, however, remains unknown. Using the mass spectrometry-based approach, we identify neuropilin-1 (NRP1) as a NE receptor that mediates uptake and PR1 cross-presentation in breast cancer cells. We demonstrated that soluble NE is a specific, high-affinity ligand for NRP1 with a calculated Kd of 38.7 nm Furthermore, we showed that NRP1 binds to the RRXR motif in NE. Notably, NRP1 knockdown with interfering RNA or CRISPR-cas9 system and blocking using anti-NRP1 antibody decreased NE uptake and, subsequently, susceptibility to lysis by PR1-specific cytotoxic T cells. Expression of NRP1 in NRP1-deficient cells was sufficient to induce NE uptake. Altogether, because NRP1 is broadly expressed in tumors, our findings suggest a role for this receptor in immunotherapy strategies that target cross-presented antigens.


Asunto(s)
Absorción Fisiológica , Neoplasias de la Mama/metabolismo , Reactividad Cruzada , Elastasa de Leucocito/metabolismo , Proteínas de Neoplasias/metabolismo , Neuropilina-1/metabolismo , Secuencias de Aminoácidos , Anticuerpos Bloqueadores/metabolismo , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Sistemas CRISPR-Cas , Línea Celular Tumoral , Femenino , Humanos , Cinética , Elastasa de Leucocito/química , Elastasa de Leucocito/inmunología , Ligandos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Neuropilina-1/antagonistas & inhibidores , Neuropilina-1/química , Neuropilina-1/genética , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Dominios y Motivos de Interacción de Proteínas , Interferencia de ARN , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Solubilidad , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo
6.
Methods Mol Biol ; 1321: 73-90, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26082216

RESUMEN

While yeast are lower eukaryotic organisms, they share many common features and biological processes with higher eukaryotes. As such, yeasts have been used as model organisms to facilitate our understanding of such features and processes. To this end, a large number of powerful genetic tools have been developed to investigate and manipulate these organisms. Going hand-in-hand with these genetic tools is the ability to efficiently scale up the fermentation of these organisms, thus making them attractive hosts for the production of recombinant proteins. A key feature of producing recombinant proteins in yeast is that these proteins can be readily secreted into the culture supernatant, simplifying any downstream processing. A consequence of this secretion is that the proteins typically pass through the secretory pathway, during which they may be exposed to various posttranslational modifications. The addition of glycans is one such modification. Unfortunately, while certain aspects of glycosylation are shared between lower and higher eukaryotes, significant differences exist. Over the last two decades much research has focused on engineering the glycosylation pathways of yeast to more closely resemble those of higher eukaryotes, particularly those of humans for the production of therapeutic proteins. In the current review we shall highlight some of the key achievements in yeast glyco-engineering which have led to humanization of both the N- and O-linked glycosylation pathways.


Asunto(s)
Proteínas Fúngicas/genética , Proteínas Recombinantes/genética , Levaduras/genética , Ingeniería Genética/métodos , Glicosilación , Humanos , Polisacáridos/genética , Procesamiento Proteico-Postraduccional/genética
7.
J Biotechnol ; 208: 13-21, 2015 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-26015261

RESUMEN

Monoclonal antibody (mAb) therapy has been successfully used for the treatment of B-cell lymphomas and is currently extended for the treatment of multiple myeloma (MM). New developments in MM therapeutics have achieved significant survival gains in patients but the disease still remains incurable. Elotuzumab (HuLuc63), an anti-CS1 monoclonal IgG1 antibody, is believed to induce anti-tumor activity and MM cytotoxicity through antibody dependent cellular cytotoxicity (ADCC) and inhibition of MM cell adhesion to bone marrow stromal cells (BMSCs). Modulations of the Fc glycan composition at the N297 site by selective mutations or afucosylation have been explored as strategies to develop bio-better therapeutics with enhanced ADCC activity. Afucosylated therapeutic antibodies with enhanced ADCC activity have been reported to possess greater efficacy in tumor growth inhibition at lower doses when compared to fucosylated therapeutic antibodies. The N-linked glycosylation pathway in Pichia pastoris has been engineered to produce human-like N-linked glycosylation with uniform afucosylated complex type glycans. The purpose of this study was to compare afucosylated anti-CS1 mAb expressed in glycoengineered Pichia pastoris with fucosylated anti-CS1 mAb expressed in mammalian HEK293 cells through in vitro ADCC and in vivo tumor inhibition models. Our results indicate that Fc glycosylation is critical for in vivo efficacy and afucosylated anti-CS1 mAb expressed in glycoengineered Pichia pastoris shows a better in vivo efficacy in tumor regression when compared to fucosylated anti-CS1 mAb expressed in HEK293 cells. Glycoengineered Pichia pastoris could provide an alternative platform for generating homogeneous afucosylated recombinant antibodies where Fc mediated immune effector function is important for efficacy.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Antineoplásicos , Ingeniería Celular , Mieloma Múltiple/tratamiento farmacológico , Neoplasias Experimentales/tratamiento farmacológico , Pichia , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/farmacología , Anticuerpos Antineoplásicos/biosíntesis , Anticuerpos Antineoplásicos/química , Anticuerpos Antineoplásicos/genética , Línea Celular Tumoral , Glicosilación , Células HEK293 , Humanos , Ratones SCID , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Pichia/genética , Pichia/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Ensayos Antitumor por Modelo de Xenoinjerto
8.
PLoS One ; 8(7): e70190, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23875020

RESUMEN

State-of-the-art monoclonal antibody (mAb) discovery methods that utilize surface display techniques in prokaryotic and eukaryotic cells require multiple steps of reformatting and switching of hosts to transition from display to expression. This results in a separation between antibody affinity maturation and full-length mAb production platforms. Here, we report for the first time, a method in Glyco-engineered Pichiapastoris that enables simultaneous surface display and secretion of full-length mAb molecules with human-like N-glycans using the same yeast cell. This paradigm takes advantage of homo-dimerization of the Fc portion of an IgG molecule to a surface-anchored "bait" Fc, which results in targeting functional "half" IgGs to the cell wall of Pichiapastoris without interfering with the secretion of full length mAb. We show the utility of this method in isolating high affinity, well-expressed anti-PCSK9 leads from a designed library that was created by mating yeasts containing either light chain or heavy chain IgG libraries. Coupled with Glyco-engineered Pichiapastoris, this method provides a powerful tool for the discovery and production of therapeutic human mAbs in the same host thus improving drug developability and potentially shortening the discovery time cycle.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Formación de Anticuerpos , Pichia , Ingeniería de Proteínas/métodos , Anticuerpos Monoclonales/genética , Afinidad de Anticuerpos/genética , Afinidad de Anticuerpos/inmunología , Formación de Anticuerpos/genética , Separación Celular/métodos , Glicosilación , Humanos , Organismos Modificados Genéticamente , Biblioteca de Péptidos , Pichia/genética , Pichia/metabolismo , Multimerización de Proteína , Procesamiento Proteico-Postraduccional
9.
PLoS One ; 8(7): e68325, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23840891

RESUMEN

The methylotrophic yeast, Pichiapastoris, is an important organism used for the production of therapeutic proteins. However, the presence of fungal-like glycans, either N-linked or O-linked, can elicit an immune response or enable the expressed protein to bind to mannose receptors, thus reducing their efficacy. Previously we have reported the elimination of ß-linked glycans in this organism. In the current report we have focused on reducing the O-linked mannose content of proteins produced in P. pastoris, thereby reducing the potential to bind to mannose receptors. The initial step in the synthesis of O-linked glycans in P. pastoris is the transfer of mannose from dolichol-phosphomannose to a target protein in the yeast secretory pathway by members of the protein-O-mannosyltransferase (PMT) family. In this report we identify and characterize the members of the P. pastoris PMT family. Like Candida albicans, P. pastoris has five PMT genes. Based on sequence homology, these PMTs can be grouped into three sub-families, with both PMT1 and PMT2 sub-families possessing two members each (PMT1 and PMT5, and PMT2 and PMT6, respectively). The remaining sub-family, PMT4, has only one member (PMT4). Through gene knockouts we show that PMT1 and PMT2 each play a significant role in O-glycosylation. Both, by gene knockouts and the use of Pmt inhibitors we were able to significantly reduce not only the degree of O-mannosylation, but also the chain-length of these glycans. Taken together, this reduction of O-glycosylation represents an important step forward in developing the P. pastoris platform as a suitable system for the production of therapeutic glycoproteins.


Asunto(s)
Manosiltransferasas/genética , Pichia/enzimología , Pichia/genética , Candida albicans/enzimología , Candida albicans/genética , Técnicas de Inactivación de Genes , Genes Fúngicos , Glicosilación , Manosiltransferasas/metabolismo , Filogenia , Pichia/metabolismo , Polisacáridos/metabolismo
10.
Methods Mol Biol ; 988: 31-43, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23475712

RESUMEN

Currently, mammalian cells are the most commonly used hosts for the production of therapeutic monoclonal antibodies (mAbs). These hosts not only secrete mAbs with properly assembled two heavy and two light chains but also deliver mAbs with a glycosylation profile that is compatible with administration into humans. GlycoFi, a wholly owned subsidiary of Merck & Co., Inc., humanized the Pichia glycosylation pathway which allows it to express glycoproteins with a human-like glycan profile. This offers an alternative mAb production platform similar to mammalian hosts and in some cases it even provides more homogenous product and better efficacy, such as enhanced effector function. This chapter describes a protocol for using glycoengineered Pichia to produce full-length mAbs. It covers a broad spectrum of mAb expression technologies in yeast including expression vector construction, yeast transformation, high-throughput strain selection to fermentation, and antibody purification.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Pichia/metabolismo , Ingeniería de Proteínas/métodos , Secuencia de Aminoácidos , Anticuerpos Monoclonales/aislamiento & purificación , Clonación Molecular , Medios de Cultivo Condicionados/química , Fermentación , Vectores Genéticos , Glicosilación , Humanos , Datos de Secuencia Molecular , Pichia/crecimiento & desarrollo , Plásmidos/genética , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Coloración y Etiquetado , Transformación Genética
11.
J Immunol Methods ; 386(1-2): 34-42, 2012 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-22982058

RESUMEN

Previous studies have shown that glycoproteins expressed in wild-type Pichia pastoris bind to Dendritic cell-SIGN (DC-Specific Intercellular adhesion molecule-3 Grabbing Nonintegrin), a mannose-binding receptor found on dendritic cells in peripheral tissues which is involved in antigen presentation and the initiation of an immune response. However, the binding of DC-SIGN to glycoproteins purified from P. pastoris strains engineered to express humanized N- and O-linked glycans has not been tested to date. In this study, the binding of glycoproteins with specific high-mannose or human N- and O-linked glycan structures to DC-SIGN was tested. Proteins with humanized N-glycans including Man5 structures and O-glycans (up to as many as 24) with single mannose chain length showed DC-SIGN binding that was comparable to that measured for a CHO-produced IgG1 which lacks O-linked mannose. Glycoproteins with wild-type N-glycans and mannotriose and higher O-glycans bound to DC-SIGN in a manner that was strongly inhibited by either the use of enzymatic N-deglycosylation or sodium meta-periodate oxidation. Mannan purified from humanized P. pastoris also showed lower ability to inhibit DC-SIGN binding to glycoproteins with wild type fungal glycosylation than mannan purified from wild type strains. This study shows that humanized P. pastoris can produce glycoproteins that do not bind to DC-SIGN.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Glicoproteínas/metabolismo , Inmunoglobulina G/metabolismo , Lectinas Tipo C/metabolismo , Pichia/genética , Polisacáridos/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Células CHO , Cricetinae , Glicoproteínas/genética , Glicosilación , Humanos , Inmunoglobulina G/genética , Manosa/metabolismo , Unión Proteica/genética , Ingeniería de Proteínas
12.
J Immunol Methods ; 375(1-2): 159-65, 2012 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-22019510

RESUMEN

A fragment of antigen binding (Fab) surface display system was developed using a glycoengineered Pichia pastoris host strain genetically modified to secrete glycoproteins with mammalian mannose-type Man(5)GlcNAc(2) N-linked glycans. The surface display method described here takes advantage of a pair of coiled-coil peptides as the linker while using the Saccharomyces cerevisiae Sed1p GPI-anchored cell surface protein as an anchoring domain. Several Fabs were successfully displayed on the cell surface using this system and the expression level of the displayed Fabs was correlated to that of secreted Fabs from the same glycoengineered host in the absence of the cell wall anchor. Strains displaying different model Fabs were mixed and, through cell sorting, the strain displaying more expressed Fab molecule or the strain displaying the Fab with higher affinity for an antigen was effectively enriched by FACS. This novel yeast surface display system provides a general platform for the display of Fab libraries for affinity and/or expression maturation using glycoengineered Pichia.


Asunto(s)
Glicoproteínas/genética , Glicoproteínas/metabolismo , Fragmentos Fab de Inmunoglobulinas/genética , Fragmentos Fab de Inmunoglobulinas/metabolismo , Pichia/genética , Pichia/metabolismo , Antígenos de Superficie/genética , Antígenos de Superficie/metabolismo , Pared Celular/genética , Pared Celular/metabolismo , Vectores Genéticos/genética , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Péptidos/genética , Péptidos/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
13.
Glycobiology ; 21(12): 1606-15, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21798867

RESUMEN

The N-glycosylation pathway in Pichia pastoris has been humanized by the deletion of genes responsible for fungal-type glycosylation (high mannose) as well as the introduction of heterologous genes capable of forming human-like N-glycosylation. This results in a yeast host that is capable of expressing therapeutic glycoproteins. A thorough investigation was performed to examine whether glycoproteins expressed in glycoengineered P. pastoris strains may contain residual fungal-type high-mannose structures. In a pool of N-linked glycans enzymatically released by protein N-glycosidase from a reporter glycoprotein expressed in a developmental glycoengineered P. pastoris strain, an oligosaccharide with a mass consistent with a Hexose(9)GlcNAc(2) oligosaccharide was identified. When this structure was analyzed by a normal-phase high-performance liquid chromatography (HPLC), its retention time was identical to a Man(9)GlcNAc(2) standard. However, this Hexose(9)GlcNAc(2) oligosaccharide was found to be resistant to α-1,2-mannosidase as well as endomannosidase, which preferentially catabolizes endoplasmic reticulum oligosaccharides containing terminal α-linked glucose. To further characterize this oligosaccharide, we purified the Hexose(9)GlcNAc(2) oligosaccharide by HPLC and analyzed the structure by high-field one-dimensional (1D) and two-dimensional (2D) (1)H NMR (nuclear magnetic resonance) spectroscopy followed by structural elucidation by homonuclear and heteronuclear 1D and 2D (1)H and (13)C NMR spectroscopy. The results of these experiments lead to the identification of an oligosaccharide α-Man-(1 → 2)-ß-Man-(1 → 2)-ß-Man-(1 → 2)-α-Man-(1 → 2) moiety as part of a tri-antennary structure. The difference in enzymatic reactivity can be attributed to multiple ß-linkages on the α-1,3 arm of the Man(9)GlcNAc(2) oligosaccharide.


Asunto(s)
Manosidasas/metabolismo , Proteínas de la Membrana/metabolismo , Oligosacáridos/biosíntesis , Oligosacáridos/química , Pichia/metabolismo , Humanos , Manosidasas/genética , Proteínas de la Membrana/genética , Pichia/genética , Conformación Proteica , Relación Estructura-Actividad
14.
Biologicals ; 39(4): 205-10, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21723741

RESUMEN

A glycoengineered Pichia pastoris host was used to produce an IgG1 with either afucosylated N-glycosylation (afucosylated biantennary complex) or without N-glycosylation (N297A) while a wild type P. pastoris host was used to produce an IgG1 containing fungal-type N- and O-linked glycosylation. The PK properties of these antibodies were compared to a commercial IgG1 produced in CHO cells following intravenous administration in wild type C57B6, FcγR-/- or hFcRn transgenic mice. MAbs produced in glycoengineered yeast exhibited similar PK properties in wild type mice or FcγR-/- mice with respect to clearance (CL), volume of distribution at steady-state (Vss) and half-life (t(1/2)) to that produced in mammalian (CHO) cells, while the mAb produced in wild type yeast exhibited ∼2-3-fold faster CL, which might be due to the high mannose content interacting with mannose receptors. Furthermore, in vitro binding affinity to human FcRn or mouse FcRn was similar between the reference mAb and mAbs produced in humanized yeast, and the glycovariants produced in humanized yeast exhibited similar PK patterns in human FcRn transgenic mice and in wild type mice. These results suggest the potential application of P. pastoris as a production platform for clinically viable mAbs.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Pichia/genética , Animales , Anticuerpos Monoclonales/genética , Células CHO , Cricetinae , Cricetulus , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
15.
MAbs ; 3(3): 289-98, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21487242

RESUMEN

Mammalian cell culture systems are used predominantly for the production of therapeutic monoclonal antibody (mAb) products. A number of alternative platforms, such as Pichia engineered with a humanized N-linked glycosylation pathway, have recently been developed for the production of mAbs. The glycosylation profiles of mAbs produced in glycoengineered Pichia are similar to those of mAbs produced in mammalian systems. This report presents for the first time the comprehensive characterization of an anti-human epidermal growth factor receptor 2 (HER2) mAb produced in a glycoengineered Pichia, and a study comparing the anti-HER2 from Pichia, which had an amino acid sequence identical to trastuzumab, with trastuzumab. The comparative study covered a full spectrum of preclinical evaluation, including bioanalytical characterization, in vitro biological functions, in vivo anti-tumor efficacy and pharmacokinetics in both mice and non-human primates. Cell signaling and proliferation assays showed that anti-HER2 from Pichia had antagonist activities comparable to trastuzumab. However, Pichia-produced material showed a 5-fold increase in binding affinity to FcγIIIA and significantly enhanced antibody dependant cell-mediated cytotoxicity (ADCC) activity, presumably due to the lack of fucose on N-glycans. In a breast cancer xenograft mouse model, anti-HER2 was comparable to trastuzumab in tumor growth inhibition. Furthermore, comparable pharmacokinetic profiles were observed for anti-HER2 and trastuzumab in both mice and cynomolgus monkeys. We conclude that glycoengineered Pichia provides an alternative production platform for therapeutic mAbs and may be of particular interest for production of antibodies for which ADCC is part of the clinical mechanism of action.


Asunto(s)
Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales/inmunología , Pichia/genética , Receptor ErbB-2/inmunología , Proteínas Recombinantes/inmunología , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Afinidad de Anticuerpos/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Área Bajo la Curva , Unión Competitiva/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Evaluación Preclínica de Medicamentos , Fucosa/metabolismo , Ingeniería Genética , Humanos , Macaca fascicularis , Ratones , Ratones Endogámicos C57BL , Pichia/metabolismo , Polisacáridos/metabolismo , Unión Proteica/inmunología , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacología , Trastuzumab , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Protein Expr Purif ; 76(1): 7-14, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21074617

RESUMEN

A robust and scalable purification process was developed to quickly generate antibody of high purity and sufficient quantity from glycoengineered Pichia pastoris fermentation. Protein A affinity chromatography was used to capture the antibody from fermentation supernatant. A pH gradient elution was applied to the Protein A column to prevent antibody precipitation at low pH. Antibody from Protein A chromatography contained some product related impurities, which were the misassembling of cleaved heavy chain, heavy chain and light chain. It also had some process related impurities, including Protein A residues, endotoxin, host cell DNA and proteins. Cation exchange chromatography with optimal NaCl gradient at pH 4.5-6.0 efficiently removed these product and process related impurities. The antibody from glycoengineered P. pastoris was comparable to its commercial counterpart in heterotetramer folding, physical stability and binding affinity.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Pichia/metabolismo , Proteínas Recombinantes/biosíntesis , Anticuerpos Monoclonales/aislamiento & purificación , Organismos Modificados Genéticamente , Pichia/genética , Proteínas Recombinantes/aislamiento & purificación
17.
J Ind Microbiol Biotechnol ; 37(9): 961-71, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20711797

RESUMEN

The methylotrophic yeast Pichia pastoris has recently been engineered to express therapeutic glycoproteins with uniform human N-glycans at high titers. In contrast to the current art where producing therapeutic proteins in mammalian cell lines yields a final product with heterogeneous N-glycans, proteins expressed in glycoengineered P. pastoris can be designed to carry a specific, preselected glycoform. However, significant variability exists in fermentation performance between genotypically similar clones with respect to cell fitness, secreted protein titer, and glycan homogeneity. Here, we describe a novel, multidimensional screening process that combines high and medium throughput tools to identify cell lines producing monoclonal antibodies (mAbs). These cell lines must satisfy multiple selection criteria (high titer, uniform N-glycans and cell robustness) and be compatible with our large-scale production platform process. Using this selection process, we were able to isolate a mAb-expressing strain yielding a titer (after protein A purification) in excess of 1 g/l in 0.5-l bioreactors.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Ingeniería Genética , Glicoproteínas/biosíntesis , Pichia/aislamiento & purificación , Proteínas Recombinantes/biosíntesis , Anticuerpos Monoclonales/genética , Reactores Biológicos , Técnicas de Cultivo de Célula , Línea Celular , ADN de Hongos/genética , Fermentación , Expresión Génica , Glicoproteínas/genética , Glicosilación , Humanos , Técnicas Microbiológicas , Pichia/genética , Pichia/metabolismo , Proteínas Recombinantes/genética , Selección Genética , Transformación Genética
18.
J Immunol Methods ; 358(1-2): 66-74, 2010 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-20338179

RESUMEN

A simple cell labeling method for sorting yeast Pichia pastoris antibody expressing strains is described. A small portion of secreted recombinant antibody retained on the cell surface was labeled with fluorescence detection antibody. The signal intensity of the labeled cell was correlated with the cell's antibody productivity. Using this labeling technique to sort a mixture model induced in the same fermenter where the cells of high producing strain were spiked into a population of a low producing strain at the frequency of 1:100,000, one round of sorting achieved a approximately 5000-fold enrichment of the high producing strain. A variety of P.pastoris strains expressing antibody sorted based on the signal intensity on the cell surface yielded titer improvements by 30% to 300%. Our data demonstrate that Pichia cell surface labeling is a simple, effective and reliable method for sorting Pichia antibody expressing strains for productivity improvement.


Asunto(s)
Inmunoglobulina G/biosíntesis , Proteínas de la Membrana/análisis , Proteínas de la Membrana/inmunología , Pichia/aislamiento & purificación , Pichia/metabolismo , Proteínas Recombinantes/biosíntesis , Coloración y Etiquetado/métodos , Animales , Anticuerpos/inmunología , Reactores Biológicos , Citometría de Flujo , Cabras , Humanos , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Microscopía Confocal , Pichia/clasificación , Pichia/citología , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes/análisis , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología
19.
J Biotechnol ; 139(4): 318-25, 2009 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-19162096

RESUMEN

The growing antibody market and the pressure to improve productivity as well as reduce cost of production have fueled the development of alternative expression systems. The therapeutic function of many antibodies is influenced by N-linked glycosylation, which is affected by a combination of the expression host and culture conditions. This paper reports the generation of a glycoengineered Pichia pastoris strain capable of producing more than 1 g l(-1) of a functional monoclonal antibody in a robust, scalable and portable cultivation process with uniform N-linked glycans of the type Man(5)GlcNAc(2). N-linked glycan uniformity and volumetric productivity have been maintained across a range of cultivation process conditions including pH (5.5-7.5), temperature (16-24 degrees C), dissolved oxygen concentration (0.85-3.40 mg l(-1)) and specific methanol feed rate (9-19 mg g(-1) h(-1)) as well as across different cultivation scales (0.5, 3.0, 15 and 40 l). Compared to a marketed CHO-produced therapeutic antibody, the glycoengineered yeast-produced antibody has similar motilities on SDS-PAGE, comparable size exclusion chromatograms (SEC) and antigen binding affinities. This paper provides proof of concept that glycoengineered yeast can be used to produce functional full-length monoclonal antibodies at commercially viable productivities.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Inmunoglobulina G/biosíntesis , Pichia/genética , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/aislamiento & purificación , Afinidad de Anticuerpos , Sitios de Unión de Anticuerpos , Reactores Biológicos , Células Cultivadas , Ingeniería Genética , Inestabilidad Genómica , Glicosilación , Humanos , Concentración de Iones de Hidrógeno , Inmunoglobulina G/genética , Inmunoglobulina G/aislamiento & purificación , Metanol/química , Oxígeno/química , Pichia/metabolismo , Temperatura
20.
Nat Biotechnol ; 24(2): 210-5, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16429149

RESUMEN

As the fastest growing class of therapeutic proteins, monoclonal antibodies (mAbs) represent a major potential drug class. Human antibodies are glycosylated in their native state and all clinically approved mAbs are produced by mammalian cell lines, which secrete mAbs with glycosylation structures that are similar, but not identical, to their human counterparts. Glycosylation of mAbs influences their interaction with immune effector cells that kill antibody-targeted cells. Here we demonstrate that human antibodies with specific human N-glycan structures can be produced in glycoengineered lines of the yeast Pichia pastoris and that antibody-mediated effector functions can be optimized by generating specific glycoforms. Glycoengineered P. pastoris provides a general platform for producing recombinant antibodies with human N-glycosylation.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Mejoramiento Genético/métodos , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/genética , Pichia/genética , Pichia/metabolismo , Ingeniería de Proteínas/métodos , Anticuerpos Monoclonales/genética , Glicosilación , Humanos , Proteínas Recombinantes/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA