Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 52019 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-31039141

RESUMO

Chimeric antigen receptor (CAR) T cell therapies have achieved promising outcomes in several cancers, however more challenging oncology indications may necessitate advanced antigen receptor designs and functions. Here we describe a bipartite receptor system comprised of separate antigen targeting and signal transduction polypeptides, each containing an extracellular dimerization domain. We demonstrate that T cell activation remains antigen dependent but can only be achieved in the presence of a dimerizing drug, rapamycin. Studies performed in vitro and in xenograft mouse models illustrate equivalent to superior anti-tumor potency compared to currently used CAR designs, and at rapamycin concentrations well below immunosuppressive levels. We further show that the extracellular positioning of the dimerization domains enables the administration of recombinant re-targeting modules, potentially extending antigen targeting. Overall, this novel regulatable CAR design has exquisite drug sensitivity, provides robust anti-tumor responses, and is uniquely flexible for multiplex antigen targeting or retargeting, which may further assist the development of safe, potent and durable T cell therapeutics.


Assuntos
Antígenos CD19/imunologia , Imunoterapia Adotiva/métodos , Neoplasias/terapia , Receptores de Antígenos Quiméricos/genética , Proteínas Recombinantes de Fusão/genética , Animais , Antígenos CD19/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Ativação Linfocitária , Camundongos , Neoplasias/imunologia , Neoplasias/patologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Domínios Proteicos/genética , Multimerização Proteica/efeitos dos fármacos , Multimerização Proteica/genética , Receptores de Antígenos Quiméricos/imunologia , Receptores de Antígenos Quiméricos/metabolismo , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/metabolismo , Sirolimo/administração & dosagem , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/transplante , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Hum Gene Ther ; 29(5): 585-601, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29641319

RESUMO

B-cell maturation antigen (BCMA) expression has been proposed as a marker for the identification of malignant plasma cells in patients with multiple myeloma (MM). Nearly all MM tumor cells express BCMA, while normal tissue expression is restricted to plasma cells and a subset of mature B cells. Consistent BCMA expression was confirmed on MM biopsies (29/29 BCMA+), and it was further demonstrated that BCMA is expressed in a substantial number of lymphoma samples, as well as primary chronic lymphocytic leukemia B cells. To target BCMA using redirected autologous T cells, lentiviral vectors (LVV) encoding chimeric antigen receptors (CARs) were constructed with four unique anti-BCMA single-chain variable fragments, fused to the CD137 (4-1BB) co-stimulatory and CD3ζ signaling domains. One LVV, BB2121, was studied in detail, and BB2121 CAR-transduced T cells (bb2121) exhibited a high frequency of CAR + T cells and robust in vitro activity against MM cell lines, lymphoma cell lines, and primary chronic lymphocytic leukemia peripheral blood. Based on receptor quantification, bb2121 recognized tumor cells expressing as little as 222 BCMA molecules per cell. The in vivo pharmacology of anti-BCMA CAR T cells was studied in NSG mouse models of human MM, Burkitt lymphoma, and mantle cell lymphoma, where mice received a single intravenous administration of vehicle, control vector-transduced T cells, or anti-BCMA CAR-transduced T cells. In all models, the vehicle and control CAR T cells failed to inhibit tumor growth. In contrast, treatment with bb2121 resulted in rapid and sustained elimination of the tumors and 100% survival in all treatment models. Together, these data support the further development of anti-BCMA CAR T cells as a potential treatment for not only MM but also some lymphomas.


Assuntos
Antígeno de Maturação de Linfócitos B/antagonistas & inibidores , Neoplasias Hematológicas/terapia , Mieloma Múltiplo/terapia , Receptores de Antígenos Quiméricos/imunologia , Animais , Antígeno de Maturação de Linfócitos B/imunologia , Complexo CD3/genética , Complexo CD3/imunologia , Linhagem Celular Tumoral , Citotoxicidade Imunológica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/patologia , Humanos , Imunoterapia Adotiva , Lentivirus/genética , Camundongos , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Receptores de Antígenos Quiméricos/uso terapêutico , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
3.
MAbs ; 5(3): 384-96, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23549103

RESUMO

The CTLA4-Ig fusion proteins abatacept and belatacept are clinically proven immunosuppressants used for rheumatoid arthritis and renal transplant, respectively. Given that both biologics are typically administered chronically by infusion, a need exists for a next-generation CTLA4-Ig with more convenient dosing. We used structure-based protein engineering to optimize the affinity of existing CTLA4-Ig therapeutics for the ligands CD80 and CD86, and for the neonatal Fc receptor, FcRn. From a rationally designed library, we identified four substitutions that enhanced binding to human CD80 and CD86. Coupled with two IgG1 Fc substitutions that enhanced binding to human FcRn, these changes comprise the novel CTLA4-Ig fusion protein, XPro9523. Compared with abatacept, XPro9523 demonstrated 5.9-fold, 23-fold, and 12-fold increased binding to CD80, CD86, and FcRn, respectively; compared with belatacept, CD80, CD86, and FcRn binding increased 1.5-fold, 7.7-fold, and 11-fold, respectively. XPro9523 and belatacept suppressed human T cell proliferation and IL-2 production more potently than abatacept. XPro9523 also suppressed inflammation in the mouse collagen-induced arthritis model. In cynomolgus monkeys, XPro9523 saturated CD80 and CD86 more effectively than abatacept and belatacept, potently inhibited IgM and IgG immunization responses, and demonstrated longer half-life. Pharmacokinetic modeling of its increased potency and persistence suggests that, in humans, XPro9523 may demonstrate superior efficacy and dosing convenience compared with abatacept and belatacept.


Assuntos
Artrite Experimental/terapia , Artrite Reumatoide/terapia , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Rejeição de Enxerto/terapia , Antígenos de Histocompatibilidade Classe I/metabolismo , Imunoconjugados/metabolismo , Ligação Proteica/efeitos dos fármacos , Receptores Fc/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Abatacepte , Animais , Afinidade de Anticorpos , Formação de Anticorpos/efeitos dos fármacos , Antígeno B7-1/imunologia , Antígeno B7-2/imunologia , Células Cultivadas , Feminino , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Imunoconjugados/genética , Imunoconjugados/farmacologia , Terapia de Imunossupressão , Transplante de Rim , Ativação Linfocitária/efeitos dos fármacos , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos DBA , Mutação/genética , Ligação Proteica/imunologia , Engenharia de Proteínas , Receptores Fc/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , Relação Estrutura-Atividade
4.
J Allergy Clin Immunol ; 129(4): 1102-15, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22257644

RESUMO

BACKGROUND: Sequestration of IgE to prevent its binding to high-affinity IgE receptor FcεRI on basophils and mast cells is an effective therapy for allergic asthma. IgE production requires differentiation of activated IgE(+) B cells into plasma cells upon allergen sensitization. B-cell receptor signaling is suppressed by the inhibitory IgG Fc receptor FcγRIIb; therefore, we reasoned that a therapeutic antibody that coengages FcγRIIb and IgE B-cell receptor would not only sequester IgE but also suppress its production by blocking IgE(+) B-cell activation and differentiation to IgE-secreting plasma cells. OBJECTIVE: To explore the effects of IgE sequestration versus IgE suppression by comparing omalizumab to FcγRIIb-optimized anti-IgE antibodies in humanized mouse models of immunoglobulin production. METHODS: By using a murine anti-IgE antibody as a template, we humanized, increased IgE binding, and modified its Fc domain to increase affinity for FcγRIIb. We next compared effects of this antibody (XmAb7195) versus omalizumab on the secretion of IgE and other isotypes in human PBMC cultures and in PBMC-engrafted severe combined immunodeficiency mice. RESULTS: Relative to omalizumab, XmAb7195 has a 5-fold higher affinity for human IgE and more than 400-fold higher affinity for FcγRIIb. In addition to sequestering soluble IgE, XmAb7195 inhibited plasma cell differentiation and consequent human IgE production through coengagement of IgE B-cell receptor with FcγRIIb. In PBMC-engrafted mice, XmAb7195 reduced total human IgE (but not IgG or IgM) levels by up to 40-fold relative to omalizumab. CONCLUSION: XmAb7195 acts by IgE sequestration coupled with an FcγRIIb-mediated inhibitory mechanism to suppress the formation of IgE-secreting plasma cells and reduce both free and total IgE levels.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Imunoglobulina E/biossíntese , Receptores de Antígenos de Linfócitos B/antagonistas & inibidores , Receptores de IgE/antagonistas & inibidores , Receptores de IgG/antagonistas & inibidores , Animais , Antialérgicos/farmacologia , Anticorpos Anti-Idiotípicos/sangue , Anticorpos Anti-Idiotípicos/imunologia , Anticorpos Anti-Idiotípicos/farmacologia , Anticorpos Monoclonais Humanizados/sangue , Anticorpos Monoclonais Humanizados/genética , Afinidade de Anticorpos/imunologia , Humanos , Imunoglobulina E/metabolismo , Fragmentos Fc das Imunoglobulinas/genética , Imunoglobulina G/biossíntese , Imunoglobulina G/sangue , Imunoglobulina M/biossíntese , Imunoglobulina M/sangue , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/transplante , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Omalizumab , Ligação Proteica/imunologia , Receptores de Antígenos de Linfócitos B/metabolismo , Receptores de IgE/metabolismo , Receptores de IgG/genética , Receptores de IgG/metabolismo
5.
Blood ; 119(9): 2074-82, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22246035

RESUMO

HM1.24, an immunologic target for multiple myeloma (MM) cells, has not been effectively targeted with therapeutic monoclonal antibodies (mAbs). In this study, we investigated in vitro and in vivo anti-MM activities of XmAb5592, a humanized anti-HM1.24 mAb with Fc-domain engineered to significantly enhance FcγR binding and associated immune effector functions. XmAb5592 increased antibody-dependent cellular cytotoxicity (ADCC) several fold relative to the anti-HM1.24 IgG1 analog against both MM cell lines and primary patient myeloma cells. XmAb5592 also augmented antibody dependent cellular phagocytosis (ADCP) by macrophages. Natural killer (NK) cells became more activated by XmAb5592 than the IgG1 analog, evidenced by increased cell surface expression of granzyme B-dependent CD107a and MM cell lysis, even in the presence of bone marrow stromal cells. XmAb5592 potently inhibited tumor growth in mice bearing human MM xenografts via FcγR-dependent mechanisms, and was significantly more effective than the IgG1 analog. Lenalidomide synergistically enhanced in vitro ADCC against MM cells and in vivo tumor inhibition induced by XmAb5592. A single dose of 20 mg/kg XmAb5592 effectively depleted both blood and bone marrow plasma cells in cynomolgus monkeys. These results support clinical development of XmAb5592, both as a monotherapy and in combination with lenalidomide, to improve patient outcome of MM.


Assuntos
Anticorpos Monoclonais Humanizados/imunologia , Antígenos CD/imunologia , Fragmentos Fc das Imunoglobulinas/imunologia , Mieloma Múltiplo/terapia , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Degranulação Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Técnicas de Cocultura , Sinergismo Farmacológico , Feminino , Proteínas Ligadas por GPI/imunologia , Humanos , Células Matadoras Naturais/imunologia , Lenalidomida , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Depleção Linfocítica , Macaca fascicularis , Camundongos , Camundongos SCID , Fagocitose/efeitos dos fármacos , Fagocitose/imunologia , Plasmócitos/efeitos dos fármacos , Plasmócitos/imunologia , Talidomida/administração & dosagem , Talidomida/análogos & derivados , Talidomida/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Immunol ; 186(7): 4223-33, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21357255

RESUMO

Engagement of the low-affinity Ab receptor FcγRIIb downregulates B cell activation, and its dysfunction is associated with autoimmunity in mice and humans. We engineered the Fc domain of an anti-human CD19 Ab to bind FcγRIIb with high affinity, promoting the coengagement of FcγRIIb with the BCR complex. This Ab (XmAb5871) stimulated phosphorylation of the ITIM of FcγRIIb and suppressed BCR-induced calcium mobilization, proliferation, and costimulatory molecule expression of human B cells from healthy volunteers and systemic lupus erythematosus (SLE) patients, as well as B cell proliferation induced by LPS, IL-4, or BAFF. XmAb5871 suppressed humoral immunity against tetanus toxoid and reduced serum IgM, IgG, and IgE levels in SCID mice engrafted with SLE or healthy human PBMC. XmAb5871 treatment also increased survival of mice engrafted with PBMC from a unique SLE patient. Unlike anti-CD20 Ab, coengagement of FcγRIIb and BCR complex did not promote B cell depletion in human PBMC cultures or in mice. Thus, amplification of the FcγRIIb inhibitory pathway in activated B cells may represent a novel B cell-targeted immunosuppressive therapeutic approach for SLE and other autoimmune diseases that should avoid the complications associated with B cell depletion.


Assuntos
Sítios de Ligação de Anticorpos , Comunicação Celular/imunologia , Imunidade Humoral , Lúpus Eritematoso Sistêmico/imunologia , Receptores de Antígenos de Linfócitos B/metabolismo , Receptores de IgG/metabolismo , Animais , Antígenos CD19/imunologia , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Subpopulações de Linfócitos B/patologia , Comunicação Celular/genética , Modelos Animais de Doenças , Feminino , Amplificação de Genes/imunologia , Células HEK293 , Humanos , Imunidade Humoral/genética , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/transplante , Lúpus Eritematoso Sistêmico/patologia , Lúpus Eritematoso Sistêmico/prevenção & controle , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos SCID , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos B/genética , Receptores de Antígenos de Linfócitos B/fisiologia , Receptores de IgG/deficiência , Receptores de IgG/fisiologia
7.
Blood ; 116(16): 3004-12, 2010 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-20616215

RESUMO

CD40 is highly expressed on various B-lineage malignancies and represents an attractive immunotherapy target for neoplastic disease. Previous work showed that engineering the Fc domain of an antibody for increased binding to Fcγ receptors (FcγRs) significantly enhanced Fc-mediated immune effector function and antitumor activity in vitro and in vivo. We developed a humanized anti-CD40 antibody similarly Fc-engineered for increased FcγR binding (XmAbCD40) and compared its efficacy with that of an anti-CD40 native IgG1 analog and the anti-CD20 antibody rituximab. XmAbCD40 increased antibody-dependent cell-mediated cytotoxicity (ADCC) up to 150-fold relative to anti-CD40 IgG1 against B-lymphoma, leukemia, and multiple myeloma cell lines, and significantly enhanced ADCC against primary tumors. XmAbCD40 was also superior to rituximab in enhancing ADCC (both in cell lines and primary tumors) and in augmenting antibody-dependent cellular phagocytosis. XmAbCD40 significantly inhibited lymphoma growth in disseminated and established mouse xenografts and was more effective than the IgG1 analog or rituximab. An anti-CD40 antibody constructed to abrogate FcγR binding showed no reduction of tumor growth, indicating that the in vivo antitumor activity of XmAbCD40 is primarily mediated via FcγR-dependent mechanisms. These data demonstrate that XmAbCD40 displays potent antitumor efficacy and merits further evaluation for the treatment of CD40(+) malignancies.


Assuntos
Anticorpos/imunologia , Anticorpos/uso terapêutico , Citotoxicidade Celular Dependente de Anticorpos , Antígenos CD40/imunologia , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/terapia , Receptores de IgG/imunologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Imunoterapia , Leucemia/imunologia , Leucemia/terapia , Leucemia Linfocítica Crônica de Células B/imunologia , Leucemia Linfocítica Crônica de Células B/terapia , Leucemia Plasmocitária/imunologia , Leucemia Plasmocitária/terapia , Linfoma/imunologia , Linfoma/terapia , Camundongos , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/terapia , Células Tumorais Cultivadas
8.
Nat Biotechnol ; 28(2): 157-9, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20081867

RESUMO

Improved affinity for the neonatal Fc receptor (FcRn) is known to extend antibody half-life in vivo. However, this has never been linked with enhanced therapeutic efficacy. We tested whether antibodies with half-lives extended up to fivefold in human (h)FcRn transgenic mice and threefold in cynomolgus monkeys retain efficacy at longer dosing intervals. We observed that prolonged exposure due to FcRn-mediated enhancement of half-life improved antitumor activity of Fc-engineered antibodies in an hFcRn/Rag1(-/-) mouse model. This bridges the demand for dosing convenience with the clinical necessity of maintaining efficacy.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Animais , Meia-Vida , Macaca fascicularis , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
9.
Cancer Res ; 68(19): 8049-57, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18829563

RESUMO

CD19 is a pan B-cell surface receptor expressed from pro-B-cell development until its down-regulation during terminal differentiation into plasma cells. CD19 represents an attractive immunotherapy target for cancers of lymphoid origin due to its high expression levels on the vast majority of non-Hodgkin's lymphomas and some leukemias. A humanized anti-CD19 antibody with an engineered Fc domain (XmAb5574) was generated to increase binding to Fcgamma receptors on immune cells and thus increase Fc-mediated effector functions. In vitro, XmAb5574 enhanced antibody-dependent cell-mediated cytotoxicity 100-fold to 1,000-fold relative to an anti-CD19 IgG1 analogue against a broad range of B-lymphoma and leukemia cell lines. Furthermore, XmAb5574 conferred antibody-dependent cell-mediated cytotoxicity against patient-derived acute lymphoblastic leukemia and mantle cell lymphoma cells, whereas the IgG1 analogue was inactive. XmAb5574 also increased antibody-dependent cellular phagocytosis and apoptosis. In vivo, XmAb5574 significantly inhibited lymphoma growth in prophylactic and established mouse xenograft models, and showed more potent antitumor activity than its IgG1 analogue. Comparisons with a variant incapable of Fcgamma receptor binding showed that engagement of these receptors is critical for optimal antitumor efficacy. These results suggest that XmAb5574 exhibits potent tumor cytotoxicity via direct and indirect effector functions and thus warrants clinical evaluation as an immunotherapeutic for CD19(+) hematologic malignancies.


Assuntos
Anticorpos Monoclonais/genética , Anticorpos Monoclonais/uso terapêutico , Antígenos CD19/imunologia , Fragmentos Fc das Imunoglobulinas/genética , Leucemia/terapia , Linfoma/terapia , Animais , Anticorpos Monoclonais/biossíntese , Antineoplásicos/uso terapêutico , Feminino , Humanos , Fragmentos Fc das Imunoglobulinas/biossíntese , Fragmentos Fc das Imunoglobulinas/química , Imunoterapia , Leucemia/imunologia , Linfoma/imunologia , Camundongos , Camundongos Knockout , Camundongos SCID , Ligação Proteica , Engenharia de Proteínas/métodos , Receptores de IgG/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Methods Mol Biol ; 423: 361-72, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18370214

RESUMO

Electroporation (EP)-assisted intralesional delivery of Interleukin-2 (IL-2) plasmid (pDNA) has the potential to increase the local concentration of the expressed cytokine for an extended time in the injected tumors while minimizing its systemic concentration, in comparison with systemic delivery of the recombinant cytokine. Nonclinical Investigational New Drug application-enabling studies were performed in mice to evaluate the effect of intratumoral administration of murine IL-2 pDNA on local expression and systemic distribution of IL-2 transgene as well as the inhibition of established tumor growth. The safety of repeated administrations of a human IL-2 pDNA product candidate with EP was evaluated in rats. Following the nonclinical safety and efficacy studies, a human IL-2 pDNA product candidate intralesionally administered with EP to metastatic melanoma patients is currently being investigated in a phase I clinical trial.


Assuntos
Eletroquimioterapia/métodos , Terapia Genética/métodos , Interleucina-2/genética , Plasmídeos/administração & dosagem , Plasmídeos/genética , Animais , Linhagem Celular Tumoral , DNA Recombinante/administração & dosagem , DNA Recombinante/genética , Terapia Genética/efeitos adversos , Humanos , Interleucina-2/administração & dosagem , Melanoma/secundário , Melanoma/terapia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos DBA , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Segurança
11.
Biologicals ; 35(2): 123-9, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17071101

RESUMO

The contribution of Bacillus anthracis lethal factor (LF)-specific immune responses to protection against anthrax disease in humans remains incompletely defined due, in part, to a paucity of qualified reagents and a lack of standardized serological assays. Toward this end, we have identified and characterized suitable positive quality control and standard reference sera and developed, optimized, and qualified an enzyme-linked immunosorbent assay (ELISA) to measure LF-binding IgG. Herein we describe the performance characteristics of this ELISA and propose criteria for its use in the detection and quantification of anti-LF IgG in human serum.


Assuntos
Antígenos de Bactérias/imunologia , Antitoxinas/análise , Toxinas Bacterianas/imunologia , Ensaio de Imunoadsorção Enzimática/normas , Imunoglobulina G/análise , Antraz/prevenção & controle , Antitoxinas/uso terapêutico , Humanos , Padrões de Referência , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...