Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
EBioMedicine ; 69: 103478, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34256345

RESUMO

BACKGROUND: Approximately 40% of human pregnancies are unintended, indicating a need for more acceptable effective contraception methods. New antibody production systems make it possible to manufacture reagent-grade human monoclonal antibodies (mAbs) for clinical use. We used the Nicotiana platform to produce a human antisperm mAb and tested its efficacy for on-demand topical contraception. METHODS: Heavy and light chain variable region DNA sequences of a human IgM antisperm antibody derived from an infertile woman were inserted with human IgG1 constant region sequences into an agrobacterium and transfected into Nicotiana benthamiana. The product, an IgG1 mAb ["Human Contraception Antibody" (HCA)], was purified on Protein A columns, and QC was performed using the LabChip GXII Touch protein characterization system and SEC-HPLC. HCA was tested for antigen specificity by immunofluorescence and western blot assays, antisperm activity by sperm agglutination and complement dependent sperm immobilization assays, and safety in a human vaginal tissue (EpiVaginal™) model. FINDINGS: HCA was obtained at concentrations ranging from 0.4 to 4 mg/ml and consisted of > 90% IgG monomers. The mAb specifically reacted with a glycan epitope on CD52g, a glycoprotein produced in the male reproductive tract and found in abundance on sperm. HCA potently agglutinated sperm under a variety of relevant physiological conditions at concentrations ≥ 6.25 µg/ml, and mediated complement-dependent sperm immobilization at concentrations ≥ 1 µg/ml. HCA and its immune complexes did not induce inflammation in EpiVaginal™ tissue. INTERPRETATION: HCA, an IgG1 mAb with potent sperm agglutination and immobilization activity and a good safety profile, is a promising candidate for female contraception. FUNDING: This research was supported by grants R01 HD095630 and P50HD096957 from the National Institutes of Health.


Assuntos
Anticorpos Monoclonais/imunologia , Antígeno CD52/imunologia , Anticoncepção Imunológica/métodos , Espermatozoides/imunologia , Vacinas Anticoncepcionais/imunologia , Especificidade de Anticorpos , Feminino , Humanos , Masculino
2.
Front Immunol ; 12: 792465, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34975892

RESUMO

Multiple sclerosis (MS) is a chronic autoimmune disease driven by T and B lymphocytes. The remyelination failure and neurodegeneration results in permanent clinical disability in MS patients. A desirable therapy should not only modulate the immune system, but also promote neuroprotection and remyelination. To investigate the neuroprotective effect of CD52 antibody in MS, both C57BL/6J and SJL mice with experimental autoimmune encephalomyelitis (EAE) were treated with CD52 antibody at the peak of disease. Treatment with CD52 antibody depleted T but not B lymphocytes in the blood, reduced the infiltration of T lymphocytes and microglia/macrophages in the spinal cord. Anti-CD52 therapy attenuated EAE scores during the recovery phase. It protected neurons immediately after treatment (within 4 days) as shown by reducing the accumulation of amyloid precursor proteins. It potentially promoted remyelination as it increased the number of olig2/CC-1-positive mature oligodendrocytes and prevented myelin loss in the following days (e.g., 14 days post treatment). In further experiments, EAE mice with a conditional knockout of BDNF in neurons were administered with CD52 antibodies. Neuronal deficiency of BDNF attenuated the effect of anti-CD52 treatment on reducing EAE scores and inflammatory infiltration but did not affect anti-CD52 treatment-induced improvement of myelin coverage in the spinal cord. In summary, anti-CD52 therapy depletes CD4-positive T lymphocytes, prevents myelin loss and protects neurons in EAE mice. Neuronal BDNF regulates neuroprotective and anti-inflammatory effect of CD52 antibody in EAE mice.


Assuntos
Anti-Inflamatórios/farmacologia , Anticorpos/farmacologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Antígeno CD52/antagonistas & inibidores , Encefalomielite Autoimune Experimental/tratamento farmacológico , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Medula Espinal/efeitos dos fármacos , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Antígeno CD52/imunologia , Antígeno CD52/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Mediadores da Inflamação/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/imunologia , Neurônios/metabolismo , Neurônios/patologia , Remielinização/efeitos dos fármacos , Medula Espinal/imunologia , Medula Espinal/metabolismo , Medula Espinal/patologia
3.
Eur J Immunol ; 50(9): 1362-1373, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32388861

RESUMO

Lymphocyte depletion using anti-CD52 antibody effectively reduces relapses of multiple sclerosis (MS). To begin to understand what mechanisms might control this outcome, we examined the effect of a murine-CD52-specific mAb on the depletion and repopulation of immune cells in mice with experimental autoimmune encephalomyelitis (EAE), a model of MS. We tested whether the tolerance-promoting receptor programmed cell death protein-1 (PD-1) is required for disease remission post anti-CD52, and found that PD-1-deficient mice with a more severe EAE were nevertheless effectively treated with anti-CD52. Anti-CD52 increased the proportions of newly generated T cells and double-negative (DN) T cells while reducing newly generated B cells; the latter effect being associated with a higher expression of CD52 by these cells. In the longer term, anti-CD52 caused substantial increases in the proportion of newly generated lymphocytes and DN T cells in mice with EAE. Thus, the rapid repopulation of lymphocytes from central lymphoid organs post anti-CD52 may limit further disease. Furthermore, these data identify DN T cells, a subset with immunoregulatory potential, as a significant hyperrepopulating subset following CD52-mediated depletion.


Assuntos
Linfócitos B/imunologia , Antígeno CD52/antagonistas & inibidores , Encefalomielite Autoimune Experimental/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T/imunologia , Alemtuzumab/farmacologia , Animais , Antígeno CD52/imunologia , Camundongos , Receptor de Morte Celular Programada 1
4.
Nat Commun ; 11(1): 1776, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32296059

RESUMO

Asthma is a chronic and genetically complex respiratory disease that affects over 300 million people worldwide. Here, we report a genome-wide analysis for asthma using data from the UK Biobank and the Trans-National Asthma Genetic Consortium. We identify 66 previously unknown asthma loci and demonstrate that the susceptibility alleles in these regions are, either individually or as a function of cumulative genetic burden, associated with risk to a greater extent in men than women. Bioinformatics analyses prioritize candidate causal genes at 52 loci, including CD52, and demonstrate that asthma-associated variants are enriched in regions of open chromatin in immune cells. Lastly, we show that a murine anti-CD52 antibody mimics the immune cell-depleting effects of a clinically used human anti-CD52 antibody and reduces allergen-induced airway hyperreactivity in mice. These results further elucidate the genetic architecture of asthma and provide important insight into the immunological and sex-specific relevance of asthma-associated risk variants.


Assuntos
Asma/genética , Antígeno CD52 , Predisposição Genética para Doença/epidemiologia , Fatores Sexuais , Adulto , Idoso , Animais , Asma/imunologia , Antígeno CD52/genética , Antígeno CD52/imunologia , Estudos de Casos e Controles , Feminino , Estudo de Associação Genômica Ampla , Humanos , Sistema Imunitário/citologia , Sistema Imunitário/patologia , Linfócitos/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas , Fatores de Risco , Reino Unido/epidemiologia
5.
Front Immunol ; 11: 124, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32117274

RESUMO

Alemtuzumab was designed to reduce the immunogenicity of the parent CD52-specific rat immunoglobulin. Although originally marketed for use in cancer (Mabcampath®), alemtuzumab is currently licensed and formulated for the treatment of relapsing multiple sclerosis (Lemtrada®). Perhaps due to its history as the first humanized antibody, the potential of immunogenicity of the molecule has been considered inconsequential, and anti-drug antibodies (ADA) responses were similarly reported as being clinically insignificant. Nonetheless, despite humanization and depletion of peripheral T and B cells, alemtuzumab probably generates the highest frequency of binding and neutralizing ADA of all humanized antibodies currently in clinical use, and they occur rapidly in a large majority of people with MS (pwMS) on alemtuzumab treatment. These ADA appear to be an inherent issue of the biology of the molecule-and more importantly, the target-such that avoidance of immunogenicity-related effects has been facilitated by the dosing schedule used in clinical practice. At the population level this enables the drug to work in most pwMS, but in some individuals, as we show here, antibody neutralization appears to be sufficiently severe to reduce efficacy and allow disease breakthrough. It is therefore imperative that efficacy of lymphocyte depletion and the anti-drug response is monitored in people requiring additional cycles of treatment, notably following disease breakthrough. This may help inform whether to re-treat or to switch to another disease-modifying treatment.


Assuntos
Alemtuzumab/imunologia , Anticorpos Monoclonais Humanizados/imunologia , Antígeno CD52/imunologia , Esclerose Múltipla Recidivante-Remitente/imunologia , Animais , Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Neutralizantes/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Humanos , Depleção Linfocítica/métodos , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Ratos
6.
Biotechniques ; 68(4): 185-190, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32096651

RESUMO

Aim: The humanized anti-CD52 monoclonal antibody alemtuzumab depletes lymphocytes and is currently used to treat relapsing multiple sclerosis. During treatment, anti-alemtuzumab antibodies may develop and reduce effective lymphocyte depletion in future treatment cycles. Results: Alemtuzumab-Alexa Fluor 488 conjugate binding to the CHO-CD52 cell surface was inhibited by anti-alemtuzumab antibodies. Conclusion: In this proof-of-concept study, a CHO-CD52 cell line has been developed and used to detect the presence of anti-alemtuzumab neutralizing antibodies. This platform provides the basis of an assay for routine screening of serum for neutralizing antibodies from patients treated with alemtuzumab.


Assuntos
Alemtuzumab/imunologia , Anticorpos Neutralizantes , Técnicas Citológicas/métodos , Imunoensaio/métodos , Alemtuzumab/uso terapêutico , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Ligação Competitiva/imunologia , Antígeno CD52/imunologia , Antígeno CD52/metabolismo , Células CHO/química , Células CHO/metabolismo , Cricetulus , Fluoresceínas , Humanos , Depleção Linfocítica/métodos , Esclerose Múltipla/tratamento farmacológico , Ácidos Sulfônicos
7.
Hum Immunol ; 81(2-3): 101-104, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32046870

RESUMO

Most sialic acid-binding immunoglobulin-like lectins (Siglecs) suppress immune cell function but are expressed at low levels on human T cells. We found that soluble CD52 inhibited T cell signalling by ligating Siglec-10, but the presence of Siglec-10 on human T cells has been questioned. To address this concern, we examined the expression of Siglec-10 at the RNA and protein level in human CD4+ T cells. Analysis by RNAseq, qPCR and flow cytometry demonstrated that, in contrast to other Siglecs, after activation of CD4+ T cells Siglec-10 was selectively upregulated in a subset of cells also high for CD52 expression. This observation is consistent with a homeostatic role for Siglec-10 in human CD4+ T cells.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Lectinas/metabolismo , Receptores de Superfície Celular/metabolismo , Linfócitos T CD4-Positivos/imunologia , Antígeno CD52/imunologia , Antígeno CD52/metabolismo , Humanos , Lectinas/imunologia , Ativação Linfocitária/imunologia , Receptores de Superfície Celular/imunologia , Regulação para Cima
8.
Eur J Pharmacol ; 871: 172923, 2020 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-31962100

RESUMO

The humanized anti-CD52 antibody alemtuzumab is successfully used in the treatment of multiple sclerosis (MS) and is thought to exert most of its therapeutic action by depletion and repopulation of mainly B and T lymphocytes. Although neuroprotective effects of alemtuzumab have been suggested, direct effects of anti-CD52 treatment on glial cells and neurons within the CNS itself have not been investigated so far. Here, we show CD52 expression in murine neurons, astrocytes and microglia, both in vitro and in vivo. As expected, anti CD52-treatment caused profound lymphopenia and improved disease symptoms in mice subjected to experimental autoimmune encephalomyelitis (EAE). CD52 blockade also had a significant effect on microglial morphology in organotypic hippocampal slice cultures but did not affect microglial functions. Furthermore, anti-CD52 neither changed baseline neuronal calcium, nor did it act neuroprotective in excitotoxicity models. Altogether, our findings argue against a functionally significant role of CD52 blockade on CNS neurons and microglia. The beneficial effects of alemtuzumab in MS may be exclusively mediated by peripheral immune mechanisms.


Assuntos
Alemtuzumab/imunologia , Antígeno CD52/imunologia , Microglia/patologia , Neurônios/patologia , Animais , Antígeno CD52/metabolismo , Cálcio/metabolismo , Encefalomielite Autoimune Experimental/patologia , Regulação da Expressão Gênica/imunologia , Camundongos
9.
Front Immunol ; 11: 626820, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33658999

RESUMO

Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by B cell dysregulation and breaks in tolerance that lead to the production of pathogenic autoantibodies. We performed single-cell RNA sequencing of B cells from healthy donors and individuals with SLE which revealed upregulated CD52 expression in SLE patients. We further demonstrate that SLE patients exhibit significantly increased levels of B cell surface CD52 expression and plasma soluble CD52, and levels of soluble CD52 positively correlate with measures of lupus disease activity. Using CD52-deficient JeKo-1 cells, we show that cells lacking surface CD52 expression are hyperresponsive to B cell receptor (BCR) signaling, suggesting an inhibitory role for the surface-bound protein. In healthy donor B cells, antigen-specific BCR-activation initiated CD52 cleavage in a phospholipase C dependent manner, significantly reducing cell surface levels. Experiments with recombinant CD52-Fc showed that soluble CD52 inhibits BCR signaling in a manner partially-dependent on Siglec-10. Moreover, incubation of unstimulated B cells with CD52-Fc resulted in the reduction of surface immunoglobulin and CXCR5. Prolonged incubation of B cells with CD52 resulted in the expansion of IgD+IgMlo anergic B cells. In summary, our findings suggest that CD52 functions as a homeostatic protein on B cells, by inhibiting responses to BCR signaling. Further, our data demonstrate that CD52 is cleaved from the B cell surface upon antigen engagement, and can suppress B cell function in an autocrine and paracrine manner. We propose that increased expression of CD52 by B cells in SLE represents a homeostatic mechanism to suppress B cell hyperactivity.


Assuntos
Autoanticorpos/sangue , Linfócitos B/imunologia , Antígeno CD52/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Receptores de Antígenos de Linfócitos B/metabolismo , Linfócitos B/metabolismo , Antígeno CD52/sangue , Antígeno CD52/metabolismo , Quimiocina CXCL13/metabolismo , Regulação da Expressão Gênica/imunologia , Genes MHC da Classe II/imunologia , Humanos , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Lúpus Eritematoso Sistêmico/metabolismo , Lúpus Eritematoso Sistêmico/fisiopatologia , RNA-Seq , Receptores CXCR5/metabolismo , Índice de Gravidade de Doença , Transdução de Sinais/imunologia , Análise de Célula Única , Fosfolipases Tipo C/metabolismo
10.
Front Immunol ; 10: 1967, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31507595

RESUMO

Human CD52 is a small glycopeptide (12 amino acid residues) with one N-linked glycosylation site at asparagine 3 (Asn3) and several potential O-glycosylation serine/threonine sites. Soluble CD52 is released from the surface of activated T cells and mediates immune suppression via its glycan moiety. In suppressing activated T cells, it first sequesters the pro-inflammatory high mobility group Box 1 (HMGB1) protein, which facilitates its binding to the inhibitory sialic acid-binding immunoglobulin-like lectin-10 (Siglec-10) receptor. We aimed to identify the features of CD52 glycan that underlie its bioactivity. Analysis of native CD52 purified from human spleen revealed extensive heterogeneity in N-glycosylation and multi-antennary sialylated N-glycans with abundant polyLacNAc extensions, together with mainly di-sialylated O-glycosylation type structures. Glycomic (porous graphitized carbon-ESI-MS/MS) and glycopeptide (C8-LC-ESI-MS) analysis of recombinant soluble human CD52-immunoglobulin Fc fusion proteins revealed that CD52 bioactivity was correlated with a high abundance of tetra-antennary α-2,3/6 sialylated N-glycans. Removal of α-2,3 sialylation abolished bioactivity, which was restored by re-sialylation with α-2,3 sialyltransferases. When glycoforms of CD52-Fc were fractionated by anion exchange MonoQ-GL chromatography, bioactive fractions displayed mainly tetra-antennary, α-2,3 sialylated N-glycan structures and a lower relative abundance of bisecting GlcNAc structures compared to non-bioactive fractions. In addition, O-glycan core type-2 di-sialylated structures at Ser12 were more abundant in bioactive CD52 fractions. Understanding the structural features of CD52 glycan required for its bioactivity will aid its development as an immunotherapeutic agent.


Assuntos
Antígeno CD52/imunologia , Antígeno CD52/metabolismo , Imunomodulação , Antígeno CD52/sangue , Antígeno CD52/isolamento & purificação , Cromatografia por Troca Iônica , Ensaio de Imunoadsorção Enzimática , Glicosilação , Humanos , Polissacarídeos/metabolismo , Proteínas Recombinantes , Baço/imunologia , Baço/metabolismo
11.
Transpl Immunol ; 56: 101226, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31344441

RESUMO

OBJECTIVES: Alemtuzumab (Ale) is a recombinant monoclonal antibody which binds to CD52 causing profound lymphodepletion, thus allowing its use in renal transplantation induction therapy. However, patients may be at increased risk for opportunistic infections, such as Cytomegalovirus (CMV). We analyzed CMV infection in renal allograft recipients administered low-dose valganciclovir (VGCV) prophylaxis with alemtuzumab induction and steroid minimization. MATERIALS AND METHODS: In this retrospective analysis, 678 kidney transplant recipients were evaluated, with 606 included for analysis. Patients were excluded for receiving induction therapy other than Ale, or for lack of follow-up within 1 year. VGCV prophylaxis was stratified by recipient CMV risk status and low-dose (450 mg) VGCV was given 3 times a week to low and moderate risk patients and daily to high risk individuals. Subject records were examined for recipient demographics, donor and recipient CMV serostatus, CMV viremia, and invasive infection. RESULTS: Of the 606 recipients, 154 were defined as low risk for CMV infection (donor and recipient both negative, or D-/R-), 236 as moderate risk without mismatch (D+/R+), 122 as moderate risk with mismatch (D-/R+), and 94 as high risk (D+/R-). Twenty-nine (29) individuals (4.8%) tested positive by PCR for CMV viremia and 10 (1.7%) patients developed invasive CMV disease, including colitis (n = 4), esophagitis (n = 1), enteritis (n = 1), nephritis (n = 1), and pneumonia (n = 3). High risk recipients (D+/R-) accounted for the majority of invasive CMV disease (n = 5), followed by moderate risk (n = 4). CMV viremia was also more common in high risk and moderate risk (D+/R+) individuals. Overall rejection rate for our study population was 27%. CONCLUSION: In this institution's experience, CMV incidence was reduced compared to historically reported data by using low-dose (450 mg) VGCV prophylaxis in combination with Ale induction and steroid minimization. However, overall rejection rate was significantly higher in our population, possibly influenced by the degree of steroid minimization.


Assuntos
Alemtuzumab/uso terapêutico , Antivirais/uso terapêutico , Infecções por Citomegalovirus/prevenção & controle , Citomegalovirus/fisiologia , Rejeição de Enxerto/tratamento farmacológico , Transplante de Rim , Valganciclovir/uso terapêutico , Adulto , Idoso , Alemtuzumab/efeitos adversos , Antígeno CD52/imunologia , Infecções por Citomegalovirus/etiologia , Cálculos da Dosagem de Medicamento , Humanos , Depleção Linfocítica , Pessoa de Meia-Idade , Período Pós-Operatório , Estudos Retrospectivos , Risco , Esteroides/uso terapêutico , Transplante Homólogo , Resultado do Tratamento
12.
MAbs ; 11(7): 1266-1275, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31199181

RESUMO

Deamidation evaluation and mitigation is an important aspect of therapeutic antibody developability assessment. We investigated the structure and function of the Asn-Gly deamidation in a human anti-CD52 IgG1 antibody light chain complementarity-determining region 1, and risk mitigation through protein engineering. Antigen binding affinity was found to decrease about 400-fold when Asn33 was replaced with an Asp residue to mimic the deamidation product, suggesting significant impacts on antibody function. Other variants made at Asn33 (N33H, N33Q, N33H, N33R) were also found to result in significant loss of antigen binding affinity. The co-crystal structure of the antigen-binding fragment bound to a CD52 peptide mimetic was solved at 2.2Å (PDB code 6OBD), which revealed that Asn33 directly interacts with the CD52 phosphate group via a hydrogen bond. Gly34, but sits away from the binding interface, rendering it more amendable to mutagenesis without affecting affinity. Saturation mutants at Gly34 were prepared and subjected to forced deamidation by incubation at elevated pH and temperature. Three mutants (G34R, G34K and G34Q) showed increased resistance to deamidation by LC-MS peptide mapping, while maintaining high binding affinity to CD52 antigen measured by Biacore. A complement -dependent cytotoxicity assay indicated that these mutants function by triggering antibody effector function. This study illustrates the importance of structure-based design and extensive mutagenesis to mitigate antibody developability issues.


Assuntos
Anticorpos Monoclonais/química , Antígeno CD52/química , Regiões Determinantes de Complementaridade/química , Imunoglobulina G/química , Cadeias Leves de Imunoglobulina/química , Amidas/química , Anticorpos Monoclonais/genética , Citotoxicidade Celular Dependente de Anticorpos , Asparagina/genética , Bioengenharia , Antígeno CD52/genética , Antígeno CD52/imunologia , Regiões Determinantes de Complementaridade/genética , Cristalografia por Raios X , Humanos , Imunoglobulina G/genética , Cadeias Leves de Imunoglobulina/genética , Mutagênese Sítio-Dirigida , Mapeamento de Peptídeos , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade
13.
Methods ; 154: 70-76, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30145356

RESUMO

Bispecific antibodies (biAb) targeting two different antigens or two distinct epitopes on the same antigen have demonstrated broad therapeutic utility. CD52 and CD20 are co-expressed on the cell surface of malignant B cells in B-cell non-Hodgkin lymphoma (B-NHL) and chronic lymphocytic leukemia (CLL) and increased expression of both antigens is detected on dividing or recently divided cells ("proliferative fraction") in CLL. The CD52-targeting monoclonal antibody (mAb) alemtuzumab (atz) not only depletes malignant B cells but also healthy CD52+ B and T lymphocytes and monocytes, causing severe immunosuppression. Loss of CD20 can occur in CLL after treatment with rituximab (rtx) and other CD20-targeting mAbs. To broaden the benefit of atz and rtx, we engineered an IgG1-like biAb, atz × rtx scFv-Fc. The Fc fragment of the biAb facilitates purification by Protein A affinity chromatography and supports a longer circulatory half-life. While atz × rtx scFv-Fc retained both antigen binding specificities, it showed superior binding to CD52+CD20+ B cells compared to CD52+CD20- T cells. Moreover, atz × rtx scFv-Fc mediated potent complement-dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC) in vitro and exhibited B-cell depleting but T-cell sparing activities in vivo in a CLL patient-derived xenograft model. B-cell depletion was more pronounced for cells of the proliferative fraction.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Antígenos CD20/imunologia , Antígeno CD52/antagonistas & inibidores , Imunoterapia , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Alemtuzumab , Animais , Anticorpos Biespecíficos/farmacologia , Antígenos de Neoplasias/imunologia , Antineoplásicos Imunológicos/farmacologia , Antineoplásicos Imunológicos/uso terapêutico , Antígeno CD52/imunologia , Humanos , Fragmentos Fc das Imunoglobulinas , Imunoglobulina G/farmacologia , Imunoglobulina G/uso terapêutico , Leucemia Linfocítica Crônica de Células B/imunologia , Leucemia Linfocítica Crônica de Células B/metabolismo , Leucemia Linfocítica Crônica de Células B/terapia , Camundongos , Rituximab , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Appl Microbiol Biotechnol ; 103(3): 1217-1229, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30554388

RESUMO

Hyperosmolality has been commonly investigated due to its effects on the production and quality characteristics of monoclonal antibodies (mAbs) produced in CHO cell fed-batch cultures. However, the application of hyperosmolality at different times and its effect on biopotency have seldom been researched, especially in perfusion culture. In our study, different degrees of hyperosmolality induced by sodium chloride were investigated in anti-IgE rCHO cell fed-batch cultures and anti-CD52 rCHO cell perfusion cultures during the initial and stable phases. The results showed that the initial hyperosmolality group (IHG) in fed-batch and early phase of perfusion cultures exhibited significant suppression of the viable cell density yet an enhancement in specific productivity, whereas the stable hyperosmolality group (SHG) achieved higher mAb production in both fed-batch and perfusion cultures. Additionally, the SHG produced less aggregates and acidic charge variants than IHG in fed-batch culture, which differed from perfusion cultures. However, the contents of non-glycosylation heavy chain (NGHC) and man5 were higher in SHG than in IHG in fed-batch cultures at plus 60 and 120 mOsm/kg, which was similar to perfusion cultures. Furthermore, the biopotency in the IHG was higher than in the SHG at plus 60 and 120 mOsm/kg in fed-batch cultures, which is similar to complement-dependent cytotoxicity (CDC) efficacy in perfusion cultures. The biopotency of all group was acceptable, except FI3. Thus, the study shows that hyperosmolality at a certain level could be beneficial for both mAb production, quality and biopotency, which could play an important role in process development for commercial production.


Assuntos
Anticorpos Monoclonais/biossíntese , Técnicas de Cultura Celular por Lotes/métodos , Antígeno CD52/imunologia , Imunoglobulina E/biossíntese , Concentração Osmolar , Cloreto de Sódio/análise , Animais , Reatores Biológicos/microbiologia , Células CHO , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Cricetinae , Cricetulus
15.
J Neuroinflammation ; 15(1): 225, 2018 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-30098594

RESUMO

BACKGROUND: Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) for which several new treatment options were recently introduced. Among them is the monoclonal anti-CD52 antibody alemtuzumab that depletes mainly B cells and T cells in the immune periphery. Considering the ongoing controversy about the involvement of B cells and in particular the formation of B cell aggregates in the brains of progressive MS patients, an in-depth understanding of the effects of anti-CD52 antibody treatment on the B cell compartment in the CNS itself is desirable. METHODS: We used myelin basic protein (MBP)-proteolipid protein (PLP)-induced experimental autoimmune encephalomyelitis (EAE) in C57BL/6 (B6) mice as B cell-dependent model of MS. Mice were treated intraperitoneally either at the peak of EAE or at 60 days after onset with 200 µg murine anti-CD52 vs. IgG2a isotype control antibody for five consecutive days. Disease was subsequently monitored for 10 days. The antigen-specific B cell/antibody response was measured by ELISPOT and ELISA. Effects on CNS infiltration and B cell aggregation were determined by immunohistochemistry. Neurodegeneration was evaluated by Luxol Fast Blue, SMI-32, and Olig2/APC staining as well as by electron microscopy and phosphorylated heavy neurofilament serum ELISA. RESULTS: Treatment with anti-CD52 antibody attenuated EAE only when administered at the peak of disease. While there was no effect on the production of MP4-specific IgG, the treatment almost completely depleted CNS infiltrates and B cell aggregates even when given as late as 60 days after onset. On the ultrastructural level, we observed significantly less axonal damage in the spinal cord and cerebellum in chronic EAE after anti-CD52 treatment. CONCLUSION: Anti-CD52 treatment abrogated B cell infiltration and disrupted existing B cell aggregates in the CNS.


Assuntos
Anticorpos/uso terapêutico , Linfócitos B/patologia , Antígeno CD52/imunologia , Sistema Nervoso Central/patologia , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/patologia , Animais , Células Apresentadoras de Antígenos/efeitos dos fármacos , Antígenos CD/metabolismo , Linfócitos B/efeitos dos fármacos , Linfócitos B/fisiologia , Doenças Desmielinizantes/tratamento farmacológico , Doenças Desmielinizantes/etiologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/complicações , Feminino , Citometria de Fluxo , Adjuvante de Freund/toxicidade , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteína Básica da Mielina/toxicidade , Proteína Proteolipídica de Mielina/toxicidade , Proteínas de Neurofilamentos/metabolismo , Fator de Transcrição 2 de Oligodendrócitos/metabolismo , Fatores de Tempo
16.
Proc Natl Acad Sci U S A ; 115(30): 7783-7788, 2018 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-29997173

RESUMO

CD52, a glycophosphatidylinositol (GPI)-anchored glycoprotein, is released in a soluble form following T cell activation and binds to the Siglec (sialic acid-binding Ig-like lectin)-10 receptor on T cells to suppress their function. We show that binding of CD52-Fc to Siglec-10 and T cell suppression requires the damage-associated molecular pattern (DAMP) protein, high-mobility group box 1 (HMGB1). CD52-Fc bound specifically to the proinflammatory Box B domain of HMGB1, and this in turn promoted binding of the CD52 N-linked glycan, in α-2,3 sialic acid linkage with galactose, to Siglec-10. Suppression of T cell function was blocked by anti-HMGB1 antibody or the antiinflammatory Box A domain of HMGB1. CD52-Fc induced tyrosine phosphorylation of Siglec-10 and was recovered from T cells complexed with HMGB1 and Siglec-10 in association with SHP1 phosphatase and the T cell receptor (TCR). Thus, soluble CD52 exerts a concerted immunosuppressive effect by first sequestering HMGB1 to nullify its proinflammatory Box B, followed by binding to the inhibitory Siglec-10 receptor, triggering recruitment of SHP1 to the intracellular immunoreceptor tyrosine-based inhibitory motif of Siglec-10 and its interaction with the TCR. This mechanism may contribute to immune-inflammatory homeostasis in pathophysiologic states and underscores the potential of soluble CD52 as a therapeutic agent.


Assuntos
Antígeno CD52/imunologia , Proteína HMGB1/imunologia , Lectinas/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Motivos de Aminoácidos , Anticorpos/farmacologia , Feminino , Proteína HMGB1/antagonistas & inibidores , Humanos , Masculino , Domínios Proteicos , Proteína Tirosina Fosfatase não Receptora Tipo 6/imunologia
19.
Expert Opin Biol Ther ; 18(3): 323-334, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29309202

RESUMO

INTRODUCTION: Alemtuzumab is a monoclonal antibody that targets for the destruction CD52+ cells, particularly B and T cells. Alemtuzumab is approved in more than 50 countries around the world for the treatment of adult patients with relapsing remitting multiple sclerosis (MS). Areas covered: In this review, the authors summarize biological, clinical and safety data related to the use of alemtuzumab in patients with MS. The authors then provide their expert opinion on alemtuzumab and the field as of whole before providing their perspectives for the future. Expert opinion: Alemtuzumab is highly efficacious; more so than first line treatments but comparable to natalizumab. Treatment schedule makes alemtuzumab administration easy and attractive to patients. However, its safety profile makes it a choice for a very limited number of patients, in a specific disease window. As of now, a cure for MS remains elusive and there is an unmet need for a safe and highly potent agent at the level of and beyond the blood brain barrier.


Assuntos
Alemtuzumab/uso terapêutico , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Alemtuzumab/efeitos adversos , Alemtuzumab/farmacocinética , Antígeno CD52/imunologia , Ensaios Clínicos como Assunto , Meia-Vida , Humanos , Imageamento por Ressonância Magnética , Esclerose Múltipla Recidivante-Remitente/diagnóstico por imagem , Esclerose Múltipla Recidivante-Remitente/patologia , Natalizumab/farmacocinética , Natalizumab/uso terapêutico , Vigilância de Produtos Comercializados , Resultado do Tratamento
20.
Clin Immunol ; 187: 50-57, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29031579

RESUMO

The cell-surface glycoprotein CD52 is widely expressed in lymphocytes. CD4+CD52hi T cells are functioning suppressor CD4+T cells. We investigated the role of the immune regulation of CD4+CD52 T cells in systemic lupus erythematosus (SLE). CD4+CD52lo T cells were increased in SLE patients, in positive correlation with SLEDAI, anti-ds-DNA antibody, and IgG concentration. Circulating follicular helper-like T cells (Tfh-like cells) were also increased in SLE, in positive correlation with CD4+CD52lo T cells. Chemokine receptor 8 (CCR8) expression in CD4+CD52lo T cells was increased. In vitro experiments using CD4 T cells of SLE patients showed that thymus and activation-regulated chemokine (TARC), a ligand of CCR8, contributed to the development of CD4+CD52hi T cells into CD4+CD52lo T cells. Our findings suggest that CD4+CD52lo T-cell upregulation is involved in the production of pathogens by autoantibodies, and TARC may contribute to the development of SLE through an aberrant induction of CD4+CD52lo T cells.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Antígeno CD52/imunologia , Quimiocina CCL17/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Adolescente , Adulto , Anticorpos Antinucleares/imunologia , Autoanticorpos/imunologia , Estudos de Casos e Controles , Feminino , Humanos , Imunoglobulina G/imunologia , Técnicas In Vitro , Masculino , Pessoa de Meia-Idade , Receptores CCR8/imunologia , Índice de Gravidade de Doença , Regulação para Cima , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA