Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Sci Immunol ; 7(73): eabm3723, 2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35857577

RESUMO

Antibodies protect from infection, underpin successful vaccines and elicit therapeutic responses in otherwise untreatable cancers and autoimmune conditions. The human IgG2 isotype displays a unique capacity to undergo disulfide shuffling in the hinge region, leading to modulation of its ability to drive target receptor signaling (agonism) in a variety of important immune receptors, through hitherto unexplained molecular mechanisms. To address the underlying process and reveal how hinge disulfide orientation affects agonistic activity, we generated a series of cysteine to serine exchange variants in the hinge region of the clinically relevant monoclonal antibody ChiLob7/4, directed against the key immune receptor CD40. We report how agonistic activity varies with disulfide pattern and is afforded by the presence of a disulfide crossover between F(ab) arms in the agonistic forms, independently of epitope, as observed in the determined crystallographic structures. This structural "switch" affects directly on antibody conformation and flexibility. Small-angle x-ray scattering and ensemble modeling demonstrated that the least flexible variants adopt the fewest conformations and evoke the highest levels of receptor agonism. This covalent change may be amenable for broad implementation to modulate receptor signaling in an epitope-independent manner in future therapeutics.


Assuntos
Dissulfetos , Imunoglobulina G , Anticorpos Monoclonais , Dissulfetos/química , Epitopos , Humanos , Conformação Proteica
2.
J Biol Chem ; 290(9): 5424-37, 2015 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-25568316

RESUMO

Type I anti-CD20 mAb such as rituximab and ofatumumab engage with the inhibitory FcγR, FcγRIIb on the surface of B cells, resulting in immunoreceptor tyrosine-based inhibitory motif (ITIM) phosphorylation. Internalization of the CD20·mAb·FcγRIIb complex follows, the rate of which correlates with FcγRIIb expression. In contrast, although type II anti-CD20 mAb such as tositumomab and obinutuzumab also interact with and activate FcγRIIb, this interaction fails to augment the rate of CD20·mAb internalization, raising the question of whether ITIM phosphorylation plays any role in this process. We have assessed the molecular requirements for the internalization process and demonstrate that in contrast to internalization of IgG immune complexes, FcγRIIb-augmented internalization of rituximab-ligated CD20 occurs independently of the FcγRIIb ITIM, indicating that signaling downstream of FcγRIIb is not required. In transfected cells, activatory FcγRI, FcγRIIa, and FcγRIIIa augmented internalization of rituximab-ligated CD20 in a similar manner. However, FcγRIIa mediated a slower rate of internalization than cells expressing equivalent levels of the highly homologous FcγRIIb. The difference was maintained in cells expressing FcγRIIa and FcγRIIb lacking cytoplasmic domains and in which the transmembrane domains had been exchanged. This difference may be due to increased degradation of FcγRIIa, which traffics to lysosomes independently of rituximab. We conclude that the cytoplasmic domain of FcγR is not required for promoting internalization of rituximab-ligated CD20. Instead, we propose that FcγR provides a structural role in augmenting endocytosis that differs from that employed during the endocytosis of immune complexes.


Assuntos
Anticorpos Monoclonais Murinos/metabolismo , Antígenos CD20/metabolismo , Endocitose , Receptores de IgG/metabolismo , Biotinilação , Western Blotting , Linhagem Celular Tumoral , Humanos , Microscopia Confocal , Mutação , Receptores de IgG/genética , Rituximab , Transdução de Sinais
3.
Clin Cancer Res ; 21(6): 1321-8, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25589626

RESUMO

PURPOSE: This phase I study aimed to establish the biologic effects and MTD of the agonistic IgG1 chimeric anti-CD40 antibody ChiLob7/4 in patients (pts) with a range of CD40-expressing solid tumors and diffuse large B-cell lymphoma, resistant to conventional therapy. Potential mechanisms of action for agonistic anti-CD40 include direct cytotoxic effects on tumor cells and conditioning of antigen-presenting cells. EXPERIMENTAL DESIGN: ChiLob7/4 was given by IV infusion weekly for 4 doses at a range from 0.5 to 240 mg/dose. Validated ELISAs were used to quantify ChiLob7/4 in serum and test for anti-chimeric MAb (HACA) responses. Pharmacodynamic assessments included quantitation of T-cell, natural killer-cell, and B-cell numbers and activation in blood by flow cytometry and a panel of cytokines in plasma by Luminex technology. Planned dose escalation was in cohorts of 3 patients until MTD or biologic effect, defined as reduction of peripheral blood CD19(+) B cells to 10% or less of baseline. RESULTS: Twenty-nine courses of treatment were given to 28 subjects. The MTD was 200 mg × 4, with dose-limiting toxicity of liver transaminase elevations at 240 mg. At 200 mg (range between 2.1 mg/kg and 3.3 mg/kg based on patient body weight), the trough level pretreatment was above 25 µg/mL. Grade 1-2 infusion reactions were seen above the dose of 16 mg, but could be prevented with single-dose corticosteroid premedication. HACA responses were seen after doses between 1.6 mg and 50 mg, but not above this. There were dose-dependent falls in blood B-cell numbers accompanied by reduced expression of CD21, and transient reductions in NK cell numbers with increased CD54 expression from 50 mg upward. MIP-1ß and IL12 plasma concentrations rose after doses above 16 mg. Fifteen of 29 treatments were accompanied by disease stabilization for a median 6 months, the longest for 37 months. CONCLUSIONS: ChiLob7/4 can activate B and NK cells at doses that can be administered safely, and should be tested in combination with other antibodies and chemotherapy agents.


Assuntos
Linfócitos B/imunologia , Antígenos CD40/antagonistas & inibidores , Imunoglobulina G/uso terapêutico , Células Matadoras Naturais/imunologia , Ativação Linfocitária/efeitos dos fármacos , Linfócitos T/imunologia , Adulto , Idoso , Anticorpos Monoclonais/uso terapêutico , Células Apresentadoras de Antígenos/imunologia , Antineoplásicos/uso terapêutico , Antígenos CD40/biossíntese , Antígenos CD40/imunologia , Quimiocina CCL4/sangue , Feminino , Humanos , Imunoglobulina G/efeitos adversos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Molécula 1 de Adesão Intercelular/biossíntese , Subunidade p35 da Interleucina-12/sangue , Ativação Linfocitária/imunologia , Contagem de Linfócitos , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Linfoma Difuso de Grandes Células B/patologia , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Receptores de Complemento 3d/biossíntese , Transaminases/metabolismo
4.
Blood ; 123(5): 669-77, 2014 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-24227819

RESUMO

A major feature that distinguishes type I from type II anti-CD20 monoclonal antibodies (mAbs) and reduces their therapeutic efficacy is the tendency to internalize from the cell surface. We have shown previously that the extent of internalization correlates with the capacity of type I mAb to simultaneously engage both CD20 and the inhibitory Fcγ receptor, FcγRIIb, in a bipolar configuration. Here, we investigated whether mAbs directed at other B-cell surface receptors also engaged FcγRIIb and whether this interaction promoted internalization. Most mAbs engaged and activated FcγRIIb, with the strength of activation related to the level of mAb bound to the cell surface. However, engagement did not affect internalization of most mAb-ligated receptors, either in cell lines or primary chronic lymphocytic leukemia cells with the exception of CD19 and CD38. Furthermore, at high cell concentrations/density both cis and trans interactions between cell-surface bound mAb and FcγRIIb were evident, but trans interactions did not inhibit type I anti-CD20 mAb-mediated internalization. These data identify that FcγRIIb is engaged by many mAbs in both cis and trans configurations, triggering its activation, but that internalization via FcγRIIb occurs for only a select subset. These findings have implications when designing new antibody-based therapeutics.


Assuntos
Anticorpos Monoclonais/imunologia , Receptores de IgG/imunologia , Anticorpos Monoclonais/metabolismo , Especificidade de Anticorpos , Antígenos CD20/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Humanos , Isoformas de Proteínas/imunologia , Transporte Proteico
5.
J Immunol ; 191(8): 4130-40, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-24026082

RESUMO

Genetic deficiency of the inhibitory Fc receptor, FcγRIIB (CD32b), has been shown to augment the activity of activatory FcγR and promote mAb immunotherapy. To investigate whether mAbs capable of blocking FcγRIIB have similar capacity, we recently generated a panel of specific anti-mouse FcγRIIB mAbs that do not cross-react with other FcRs, allowing us to study the potential of FcγRIIB as a therapeutic target. Previous work revealed a number of these mAbs capable of eliciting programmed cell death of targets, and in the present study we demonstrated their ability to promote target cell phagocytosis. However, in a variety of murine tumor models, anti-FcγRIIB mAbs demonstrated limited therapeutic activity despite optimized treatment regimens. Unexpectedly, we observed that the anti-FcγRIIB mAbs are rapidly and extensively consumed in vivo, both by the tumor and host cells, including B cells, leading to a precipitous loss from the circulation. Closer analysis revealed that the anti-FcγRIIB mAbs become extensively internalized from the cell surface within 24 h in vivo, likely explaining their suboptimal efficacy. Subsequent studies revealed that anti-FcγRIIB mAb immunotherapy was effective when used against FcγRIIB(+) tumors in FcγRIIB(-/-) recipients, indicating that consumption of the mAb by nontumor cells is the primary limitation of these reagents. Importantly, similar rates of internalization were not seen on human target cells, at least in vitro. These studies further highlight the need to determine the propensity of mAb therapeutics to internalize target receptors and also identify potential key differences between human and mouse cells in this respect.


Assuntos
Anticorpos Monoclonais/imunologia , Linfoma de Células B/imunologia , Macrófagos/imunologia , Mieloma Múltiplo/imunologia , Receptores de IgG/imunologia , Animais , Anticorpos Monoclonais/metabolismo , Apoptose/imunologia , Células Cultivadas , Feminino , Humanos , Imunoterapia , Linfoma de Células B/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout , Mieloma Múltiplo/terapia , Receptores de IgG/genética
6.
Blood ; 117(17): 4519-29, 2011 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-21378274

RESUMO

The anti-CD20 mAb rituximab has substantially improved the clinical outcome of patients with a wide range of B-cell malignancies. However, many patients relapse or fail to respond to rituximab, and thus there is intense investigation into the development of novel anti-CD20 mAbs with improved therapeutic efficacy. Although Fc-FcγR interactions appear to underlie much of the therapeutic success with rituximab, certain type II anti-CD20 mAbs efficiently induce programmed cell death (PCD), whereas rituximab-like type I anti-CD20 mAbs do not. Here, we show that the humanized, glycoengineered anti-CD20 mAb GA101 and derivatives harboring non-glycoengineered Fc regions are type II mAb that trigger nonapoptotic PCD in a range of B-lymphoma cell lines and primary B-cell malignancies. We demonstrate that GA101-induced cell death is dependent on actin reorganization, can be abrogated by inhibitors of actin polymerization, and is independent of BCL-2 overexpression and caspase activation. GA101-induced PCD is executed by lysosomes which disperse their contents into the cytoplasm and surrounding environment. Taken together, these findings reveal that GA101 is able to potently elicit actin-dependent, lysosomal cell death, which may potentially lead to improved clearance of B-cell malignancies in vivo.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Morte Celular/efeitos dos fármacos , Morte Celular/imunologia , Linfoma de Células B , Actinas/efeitos dos fármacos , Actinas/imunologia , Anticorpos Monoclonais Humanizados , Anticorpos Monoclonais Murinos/farmacologia , Catepsinas/farmacologia , Adesão Celular/imunologia , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular/imunologia , Resistencia a Medicamentos Antineoplásicos/imunologia , Humanos , Linfoma de Células B/tratamento farmacológico , Linfoma de Células B/imunologia , Linfoma de Células B/patologia , Lisossomos/efeitos dos fármacos , Lisossomos/imunologia , Rituximab
7.
Mol Immunol ; 48(5): 760-8, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21196049

RESUMO

This work aims to enhance killing of antibody-coated human tumor cells by altering the antibody's affinity for two effector-cell Fcγ-receptors (FcγR). Affinity for the activating FcγRIII is raised, affinity for the inhibitory FcγRIIB is reduced, with the ratio between the two association constants increasing >1000-fold. We use as a standard tool the Fab'γ from a monoclonal antibody specific for human FcγRIII. This Fab'γ module is bonded to an IgG antibody by a tandem thioether link running between cysteine residues in the hinge vicinity of each protein, thus forming a bispecific FabIgG construct. Simultaneously, effector function of the IgG module is adjusted by leaving its hinge open and adding negative charges. FabIgG constructs derived from the chimeric IgG1 antibody rituximab show the following properties. (1) The titer for antibody-dependent cellular cytotoxicity is enhanced by 12-100-fold, reflecting the affinity of the Fab'γ module for effector-cell FcγRIII. (2) Two functions of the construct's Fcγ, activation of complement and prolonged metabolic survival, are moderately reduced. (3) In contrast, affinities of the Fcγ for all FcγR are severely reduced, with two anticipated consequences. First, attacks by macrophages on antibody-coated cells are favored by reduced engagement of the inhibitory FcγRIIB. Second, reduced engagement of activating FcγR by the Fcγ lowers the probability of untoward crosslinkings of FcγR, which have been shown to provoke toxicity. If the Fab'γ module possesses human constant regions, the linkage strategy requires prior genetic deletion of at least one cysteine residue. With both Fab'γ and IgG modules available, FabIgG can be prepared in 35 h.


Assuntos
Anticorpos Biespecíficos/imunologia , Imunoglobulina G/imunologia , Receptores de IgG/imunologia , Animais , Anticorpos Biespecíficos/metabolismo , Linhagem Celular , Humanos , Imunoglobulina G/metabolismo , Camundongos
8.
Semin Hematol ; 47(2): 107-14, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20350657

RESUMO

The last decade has seen the monoclonal antibody (mAb), rituximab, transform clinical management of many non-Hodgkin lymphomas and more recently provide new opportunities for controlling autoimmune conditions, such as rheumatoid arthritis. Although not yet fully determined, the explanation for this success appears to lie with the inherent properties of its target, CD20, which allow rituximab to recruit potent cytotoxic effectors with unusual efficiency. In this review we detail the properties of CD20 that make it such an effective therapeutic target and describe how different mAbs change the membrane distribution and internalization of CD20 and have distinct modes of cytotoxic activity.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos CD20/fisiologia , Animais , Anticorpos Monoclonais/classificação , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Murinos , Especificidade de Anticorpos , Citotoxicidade Celular Dependente de Anticorpos , Reações Antígeno-Anticorpo , Antígenos CD20/química , Antígenos CD20/imunologia , Antígenos CD20/metabolismo , Antígenos de Superfície/imunologia , Antígenos de Superfície/metabolismo , Antineoplásicos/uso terapêutico , Linfócitos B/imunologia , Linfócitos B/metabolismo , Sinalização do Cálcio/fisiologia , Morte Celular , Sistemas de Liberação de Medicamentos , Humanos , Linfoma de Células B/tratamento farmacológico , Camundongos , Camundongos Transgênicos , Conformação Proteica , Transporte Proteico , Rituximab
9.
J Clin Invest ; 119(8): 2143-59, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19620786

RESUMO

mAbs are becoming increasingly utilized in the treatment of lymphoid disorders. Although Fc-FcgammaR interactions are thought to account for much of their therapeutic effect, this does not explain why certain mAb specificities are more potent than others. An additional effector mechanism underlying the action of some mAbs is the direct induction of cell death. Previously, we demonstrated that certain CD20-specific mAbs (which we termed type II mAbs) evoke a nonapoptotic mode of cell death that appears to be linked with the induction of homotypic adhesion. Here, we reveal that peripheral relocalization of actin is critical for the adhesion and cell death induced by both the type II CD20-specific mAb tositumomab and an HLA-DR-specific mAb in both human lymphoma cell lines and primary chronic lymphocytic leukemia cells. The cell death elicited was rapid, nonapoptotic, nonautophagic, and dependent on the integrity of plasma membrane cholesterol and activation of the V-type ATPase. This cytoplasmic cell death involved lysosomes, which swelled and then dispersed their contents, including cathepsin B, into the cytoplasm and surrounding environment. The resulting loss of plasma membrane integrity occurred independently of caspases and was not controlled by Bcl-2. These experiments provide what we believe to be new insights into the mechanisms by which 2 clinically relevant mAbs elicit cell death and show that this homotypic adhesion-related cell death occurs through a lysosome-dependent pathway.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígenos CD20/metabolismo , Apoptose/efeitos dos fármacos , Antígenos HLA-DR/metabolismo , Leucemia/tratamento farmacológico , Linfoma/tratamento farmacológico , Lisossomos/fisiologia , Actinas/fisiologia , Anticorpos Monoclonais/uso terapêutico , Antígenos CD20/imunologia , Autofagia , Adesão Celular/efeitos dos fármacos , Comunicação Celular , Linhagem Celular , Antígenos HLA-DR/imunologia , Humanos , Leucemia/patologia , Linfoma/patologia , Lisossomos/efeitos dos fármacos , Microdomínios da Membrana/fisiologia , Microvilosidades/fisiologia
10.
Blood ; 112(10): 4170-7, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18583569

RESUMO

Anti-CD20 monoclonal antibodies (mAbs) are classified into type I (rituximab-like) or type II (tositumomab-like) based on their ability to redistribute CD20 molecules in the plasma membrane and activate various effector functions. To compare type I and II mAbs directly in vivo and maximize Fc effector function, we selected and engineered mAbs with the same mouse IgG(2)a isotype and assessed their B-cell depleting activity in human CD20 transgenic mice. Despite being the same isotype, having similar affinity, opsonizing activity for phagocytosis, and in vivo half-life, the type II mAb tositumomab (B1) provided substantially longer depletion of B cells from the peripheral blood compared with the type I mAb rituximab (Rit m2a), and 1F5. This difference was also evident within the secondary lymphoid organs, in particular, the spleen. Failure to engage complement did not explain the efficacy of the type II reagents because type I mAbs mutated in the Fc domain (K322A) to prevent C1q binding still did not display equivalent efficacy. These results give support for the use of type II CD20 mAbs in human B-cell diseases.


Assuntos
Anticorpos Monoclonais/farmacologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Antígenos CD20/imunologia , Antineoplásicos/farmacologia , Ativação do Complemento/efeitos dos fármacos , Depleção Linfocítica/métodos , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais Murinos , Citotoxicidade Celular Dependente de Anticorpos/genética , Antígenos CD20/metabolismo , Antineoplásicos/imunologia , Antineoplásicos/metabolismo , Ativação do Complemento/genética , Ativação do Complemento/imunologia , Complemento C1q/imunologia , Complemento C1q/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Regiões Constantes de Imunoglobulina/genética , Regiões Constantes de Imunoglobulina/imunologia , Mutação de Sentido Incorreto , Ligação Proteica/genética , Ligação Proteica/imunologia , Receptores de IgG/genética , Receptores de IgG/imunologia , Rituximab
11.
J Biol Chem ; 283(25): 16971-84, 2008 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-18426802

RESUMO

The anti-CD20 monoclonal antibody (mAb) rituximab is now routinely used for the treatment of non-Hodgkins lymphoma and is being examined in a wide range of other B-cell disorders, such as rheumatoid arthritis. Despite intensive study, the mechanism of action still remains uncertain. In the current study, anti-CD20 mAb-induced calcium signaling was investigated. Previously, we grouped anti-CD20 mAbs into Type I (rituximab-like) and Type II (B1-like) based upon various characteristics such as their ability to induce complement activation and redistribute CD20 into detergent-insoluble membrane domains. Here we show that only Type I mAbs are capable of inducing a calcium flux in B cells and that this is tightly correlated with the expression of the B-cell antigen receptor (BCR). Inhibitor analysis revealed that the signaling cascade employed by CD20 was strikingly similar to that utilized by the BCR, with inhibitors of Syk, Src, and PI3K, but not EGTA, p38, or ERK1/2, completely ablating calcium flux. Furthermore, binding of Type I but not Type II mAbs caused direct association of CD20 with the BCR as measured by FRET and resulted in the phosphorylation of BCR-specific adaptor proteins BLNK and SLP-76. Crucially, variant Ramos cells lacking BCR expression but with unchanged CD20 expression were completely unable to induce calcium flux following ligation of CD20. Collectively, these data indicate that CD20 induces cytosolic calcium flux through its ability to associate with and "hijack" the signaling potential of the BCR.


Assuntos
Antígenos CD20/metabolismo , Linfócitos B/metabolismo , Citosol/metabolismo , Receptores de Antígenos de Linfócitos B/metabolismo , Anticorpos Monoclonais/química , Cálcio/química , Cálcio/metabolismo , Sinalização do Cálcio , Comunicação Celular , Separação Celular , Quelantes/farmacologia , Humanos , Modelos Biológicos , Fosforilação , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
Blood ; 104(6): 1793-800, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15172969

RESUMO

Despite the rapid and widespread integration of chimeric CD20 monoclonal antibody (mAb), rituximab, into the management of non-Hodgkin lymphoma, its efficacy remains variable and often modest when used as a single agent. To develop more potent reagents, human immunoglobulin transgenic mice were used to generate a panel of immunoglobulin G1kappa (IgG1kappa) CD20 mAbs. All reagents bound strongly to CD20(+) cells and recruited mononuclear cells for the lysis of malignant B cells. However, 2 mAbs, 2F2 and 7D8, were exceptionally active in complement-dependent cytotoxicity (CDC), being able to lyse a range of rituximab-resistant targets, such as CD20-low chronic lymphocytic leukemia (CLL), in the presence of human plasma or unfractionated blood. Further analysis showed that 2F2 and 7D8, like rituximab, redistributed CD20 into Triton X-100-insoluble regions of the plasma membrane, but that they had markedly slower off-rates. To determine whether off-rate influenced CDC, a non-complement activating F(ab')(2) antihuman kappa reagent was used. This reagent markedly slowed the off-rate of rituximab and increased its CDC activity to that of 2F2 and 7D8. Thus, with increasing evidence that mAb therapeutic activity in vivo depends on complement activation, these new CD20 reagents with their slow off-rates and increased potency in CDC hold considerable promise for improved clinical activity.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos CD20/imunologia , Citotoxicidade Imunológica , Linfoma não Hodgkin/imunologia , Linfoma não Hodgkin/patologia , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Sítios de Ligação de Anticorpos/imunologia , Linhagem Celular Tumoral , Testes de Fixação de Complemento , Proteínas do Sistema Complemento/imunologia , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Imunoglobulina G/imunologia , Cinética , Leucemia Linfocítica Crônica de Células B/imunologia , Leucemia Linfocítica Crônica de Células B/patologia , Camundongos , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA