RESUMO
Amyloidosis is a malignant pathology associated with the formation of proteinaceous amyloid fibrils that deposit in organs and tissues, leading to dysfunction and severe morbidity. More than 25 proteins have been identified as components of amyloid, but the most common form of systemic amyloidosis is associated with the deposition of amyloid composed of Ig light chains (AL). Clinical management of amyloidosis focuses on reducing synthesis of the amyloid precursor protein. However, recently, passive immunotherapy using amyloid fibril-reactive antibodies, such as 11-1F4, to remove amyloid from organs has been shown to be effective at restoring organ function in patients with AL amyloidosis. However, 11-1F4 does not bind amyloid in all AL patients, as evidenced by PET/CT imaging, nor does it efficiently bind the many other forms of amyloid. To enhance the reactivity and expand the utility of the 11-1F4 mAb as an amyloid immunotherapeutic, we have developed a pretargeting "peptope" comprising a multiamyloid-reactive peptide, p5+14, fused to a high-affinity peptide epitope recognized by 11-1F4. The peptope, known as p66, bound the 11-1F4 mAb in vitro with subnanomolar efficiency, exhibited multiamyloid reactivity in vitro and, using tissue biodistribution and SPECT imaging, colocalized with amyloid deposits in a mouse model of systemic serum amyloid A amyloidosis. Pretreatment with the peptope induced 11-1F4 mAb accumulation in serum amyloid A deposits in vivo and enhanced 11-1F4-mediated dissolution of a human AL amyloid extract implanted in mice.
Assuntos
Amiloidose/metabolismo , Amiloidose/terapia , Anticorpos Monoclonais/fisiologia , Amiloide/metabolismo , Proteínas Amiloidogênicas/metabolismo , Animais , Anticorpos Biespecíficos/imunologia , Anticorpos Monoclonais/imunologia , Cadáver , Epitopos/metabolismo , Humanos , Cadeias Leves de Imunoglobulina/imunologia , Camundongos , Peptídeos/metabolismo , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Ligação Proteica , Proteína Amiloide A Sérica/metabolismo , Distribuição Tecidual , Resultado do TratamentoRESUMO
Recent years have seen the use of recombinant proteins in the treatment of different diseases. Among them, monoclonal antibodies (mAbs) are currently the fastest growing class of bio-therapeutic recombinant proteins. Chinese hamster ovary (CHO) cells are the most commonly used host cells for production of these recombinant mAbs. Expression vectors determine the expression level and quality of recombinant mAbs. Currently, few construction strategies for recombinant mAbs expression vectors in CHO cells have been developed, including monocistronic vector, multiple-promoter expression vector, and tricistronic vector mediated by internal ribosome entry site (IRES) or Furin-2A element. Among them, Furin-2A-mediated vector is an effective approach due to advantages of high "self-cleavage" efficiency, and equal expression of light and heavy chains from a single open reading frame. Here, we have reviewed the progress in development of different strategies for constructing recombinant mAb expression vectors in CHO cells and its potential advantages and disadvantages.
Assuntos
Anticorpos Monoclonais/genética , Clonagem Molecular/métodos , Engenharia de Proteínas/métodos , Animais , Anticorpos Monoclonais/fisiologia , Formação de Anticorpos/genética , Células CHO , Cricetulus , Vetores Genéticos/síntese química , Vetores Genéticos/genética , Humanos , Regiões Promotoras Genéticas , Proteínas Recombinantes/genética , Transfecção/métodosRESUMO
PURPOSE OF REVIEW: Standard structural imaging procedures such as endoscopy, ultrasonography, or MRI are an integral part of the rational management of patients with inflammatory bowel diseases (IBDs). There is nevertheless the need for further refined imaging approaches that are able to overcome the limitations of currently used formats. The advent of molecular imaging modalities that allow real-time visualization of cellular processes not only in the preclinical setting but also in clinical trials has demonstrated its ability to improve current therapeutic strategies. The purpose of this review is to present and discuss advancements in the field of molecular imaging approaches in the IBD field. RECENT FINDINGS: Recent preclinical and clinical studies have addressed the applicability of molecular imaging for improving the differentiation between benign and malignant mucosal alterations, increasing the detection of dysplastic intestinal lesions, and predicting individual responses to biological therapies. SUMMARY: Molecular imaging in IBD represents an exciting and evolving field that has the potential to impact on current diagnostic and therapeutic algorithms in the treatment of IBD patients by analyzing and visualizing the molecular mechanisms that drive mucosal inflammation. It might enable us to base individualized therapeutic decisions on preceded molecular level analysis by suitable imaging modalities.
Assuntos
Anticorpos Monoclonais/fisiologia , Endoscopia Gastrointestinal/métodos , Doenças Inflamatórias Intestinais/diagnóstico por imagem , Imagem Molecular , Sistemas de Apoio a Decisões Clínicas , Humanos , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/patologia , Imagem Molecular/instrumentação , Imagem Molecular/tendências , Valor Preditivo dos Testes , Sensibilidade e EspecificidadeRESUMO
With the majority of HIV infections resulting from mucosal transmission, induction of an effective mucosal immune response is thought to be pivotal in preventing transmission. HIV-specific IgA, but not IgG, has been detected in the genital tract, seminal fluid, urethral swabs, urine, and vaginal wash samples of HIV-negative sex workers and HIV-status discordant couples. Purified mucosal and plasma IgA from some individuals with highly exposed, persistently seronegative status can neutralize infection and present cross-clade neutralization activity, though present at low levels. We generated a CD4-induced human mAb, F425A1g8, and characterized the impact of its isotype variants on HIV neutralizing activity. The result showed that, in contrast to little neutralization by the F425A1g8 IgG1 in the absence of sCD4, the IgA1 variant of the Ab displayed significant independent neutralization activity against a range of HIV clade B isolates in the absence of sCD4. Studies of the neutralizing function of IgA isotypes, and the functional relationship between different antigenic epitopes and IgA Abs, may also suggest strategies for the intervention of virus transmission and spread within the mucosa of the host, as well as serve to inform the design of vaccine strategies that may be more effective at preventing mucosal transmission. This research clearly suggests that IgA isotype, because of its unique molecular structure, may play an important role in HIV neutralization.
Assuntos
Anticorpos Monoclonais/química , Anticorpos Monoclonais/fisiologia , Anticorpos Anti-HIV/química , Anticorpos Anti-HIV/fisiologia , HIV-1/imunologia , Imunoglobulina A/fisiologia , Regiões Constantes de Imunoglobulina/química , Regiões Constantes de Imunoglobulina/fisiologia , Anticorpos Monoclonais/metabolismo , Sítios de Ligação de Anticorpos , Anticorpos Anti-HIV/metabolismo , HIV-1/química , HIV-1/metabolismo , Humanos , Regiões Constantes de Imunoglobulina/metabolismo , Isotipos de Imunoglobulinas/química , Isotipos de Imunoglobulinas/metabolismo , Isotipos de Imunoglobulinas/fisiologia , Testes de NeutralizaçãoRESUMO
The unexpected outcome of the clinical trial of the superagonistic CD28 mAb TGN1412 (IgG4κ) continues to stimulate interest. We show that TGN1412 binds similarly to human and cynomolgus macaque FcγR, eliminating the possibility that differences in Fc-mediated interactions with FcγR contributed to the failure of preclinical testing in macaques to predict toxicity in humans. The influence of the Fc domain and C region structure on the in vitro functional activity of TGN1412 was investigated using F(ab')(2) and Fab fragments derived from TGN1412 recovered from the trial and recombinant TGN1412 subclass variants and mutants. Superagonistic activity, as measured by cytokine release and proliferation, was assessed by exposing PBMCs to immobilized mAbs/fragments or to aqueous mAbs/fragments in the presence of HUVEC monolayers. Removing the Fc generally curtailed or abolished PBMC activation. However, eliminating detectable FcγR-binding of the IgG4 by mutation (L235E) did not abrogate activity. Stabilizing the "wild-type" IgG4 hinge (S228P) enhanced activity without increasing FcγR binding, which could only partially be explained by inhibition of Fab arm-exchange. Subclass switching the IgG4 mAb to IgG1 decreased activity, whereas switching to IgG2 markedly increased activity. We conclude that the C region strongly influences in vitro CD28-mediated superagonistic signaling. Superagonism requires an intact Fc, as shown by the absence of activity of TGN1412 Fab and F(ab')(2) fragments, but, notably, appears to be relatively independent of FcγR-binding properties. We propose that the Fc, potentially through restricting flexibility, maintains a favorable V region conformation to allow superagonistic activity. These findings have important implications for Ab design strategies.
Assuntos
Anticorpos Monoclonais Humanizados/metabolismo , Anticorpos Monoclonais/fisiologia , Regiões Constantes de Imunoglobulina/fisiologia , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais Humanizados/fisiologia , Sítios de Ligação de Anticorpos/imunologia , Proliferação de Células , Células Cultivadas , Ensaios Clínicos Fase I como Assunto , Técnicas de Cocultura , Citocinas/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Regiões Constantes de Imunoglobulina/química , Regiões Constantes de Imunoglobulina/metabolismo , Fragmentos Fab das Imunoglobulinas/fisiologia , Fragmentos Fc das Imunoglobulinas/fisiologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Macaca fascicularis , Ligação Proteica/imunologia , Receptores de IgG/metabolismo , Receptores de IgG/fisiologia , Proteínas Recombinantes/metabolismoRESUMO
Allergen-specific IgE plays an essential role in the pathogenesis of allergic asthma. Although there has been increasing evidence suggesting the involvement of IL-17 in the disease, the relationship between IL-17 and IgE-mediated asthmatic responses has not yet been defined. In this study, we attempted to elucidate the contribution of IL-17 to an IgE-mediated late-phase asthmatic response and airway hyperresponsiveness (AHR). BALB/c mice passively sensitized with an OVA-specific IgE mAb were challenged with OVA intratracheally four times. The fourth challenge caused a late-phase increase in airway resistance associated with elevated levels of IL-17(+)CD4(+) cells in the lungs. Multiple treatments with a C3a receptor antagonist or anti-C3a mAb during the challenges inhibited the increase in IL-17(+)CD4(+) cells. Meanwhile, a single treatment with the antagonist or the mAb at the fourth challenge suppressed the late-phase increase in airway resistance, AHR, and infiltration by neutrophils in bronchoalveolar lavage fluid. Because IL-17 production in the lungs was significantly repressed by both treatments, the effect of an anti-IL-17 mAb was examined. The late-phase increase in airway resistance, AHR, and infiltration by neutrophils in bronchoalveolar lavage fluid was inhibited. Furthermore, an anti-Gr-1 mAb had a similar effect. Collectively, we found that IgE mediated the increase of IL-17(+)CD4(+) cells in the lungs caused by repeated Ag challenges via C3a. The mechanisms leading to the IgE-mediated late-phase asthmatic response and AHR are closely associated with neutrophilic inflammation through the production of IL-17 induced by C3a.
Assuntos
Asma/imunologia , Asma/patologia , Hiper-Reatividade Brônquica/imunologia , Complemento C3a/fisiologia , Imunoglobulina E/fisiologia , Interleucina-17/fisiologia , Neutrófilos/imunologia , Neutrófilos/patologia , Animais , Anticorpos Monoclonais/fisiologia , Asma/metabolismo , Hiper-Reatividade Brônquica/patologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Complemento C3a/antagonistas & inibidores , Complemento C3a/imunologia , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/fisiologia , Interleucina-17/biossíntese , Interleucina-17/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Receptores de Complemento/antagonistas & inibidores , Fatores de TempoRESUMO
Chikungunya virus (CHIKV) is an alphavirus responsible for numerous epidemics in Africa and Asia. Infection by CHIKV is often characterized by long-lasting, incapacitating arthritis, and some fatal cases have been described among elderly and newborns. Currently, there is no available vaccine or specific treatment against CHIKV. Blood B cells from a donor with history of CHIKV infection were activated, immortalized, amplified, and cloned. Two human mAbs against CHIKV, 5F10 and 8B10, were identified, sequenced, and expressed in recombinant form for characterization. In a plaque reduction neutralization test, 5F10 and 8B10 show mean IC(50) of 72 and 46 ng/ml, respectively. Moreover, both mAbs lead to a strong decrease in extracellular spreading of infectious viral particles from infected to uninfected cells. Importantly, the mAbs neutralize different CHIKV isolates from Singapore, Africa, and Indonesia, as well as O'nyong-nyong virus, but do not recognize other alphaviruses tested. Both mAbs are specific for the CHIKV envelope: 5F10 binds to the E2 glycoprotein ectodomain and 8B10 to E1 and/or E2. In conclusion, these two unique human mAbs strongly, broadly, and specifically neutralize CHIKV infection in vitro and might become possible therapeutic tools against CHIKV infection, especially in individuals at risk for severe disease. Importantly, these mAbs will also represent precious tools for future studies on host-pathogen interactions and the rational design of vaccines against CHIKV.
Assuntos
Infecções por Alphavirus/imunologia , Infecções por Alphavirus/virologia , Anticorpos Monoclonais/fisiologia , Anticorpos Neutralizantes/fisiologia , Vírus Chikungunya/imunologia , Testes de Neutralização/métodos , Proteínas do Envelope Viral/imunologia , Infecções por Alphavirus/prevenção & controle , Animais , Anticorpos Monoclonais/metabolismo , Anticorpos Neutralizantes/metabolismo , Sítios de Ligação de Anticorpos , Linhagem Celular Transformada , Vírus Chikungunya/patogenicidade , Chlorocebus aethiops , Células HEK293 , Humanos , Imunização Passiva/métodos , Imunoglobulina G/metabolismo , Proteínas Recombinantes/metabolismo , Células VeroRESUMO
Despite promising results in the use of anti-epidermal growth factor receptor (EGFR) Abs for cancer therapy, several issues remain to be addressed. An increasing emphasis is being placed on immune effector mechanisms. It has become clear for other Abs directed to tumor targets that their effects involve the adaptive immunity, mainly by the contribution of Fc region-mediated mechanisms. Given the relevance of EGFR signaling for tumor biology, we wonder whether the oncogene inhibition could contribute to Ab-induced vaccine effect. In a mouse model in which 7A7 (an anti-murine EGFR Ab) and AG1478 (an EGFR-tyrosine kinase inhibitor) displayed potent antimetastatic activities, depletion experiments revealed that only in the case of the Ab, the effect was dependent on CD4(+) and CD8(+) T cells. Correspondingly, 7A7 administration elicited a remarkable tumor-specific CTL response in hosts. Importantly, experiments using 7A7 F(ab')(2) suggested that in vivo Ab-mediated EGFR blockade may play an important role in the linkage with adaptive immunity. Addressing the possible mechanism involved in this effect, we found quantitative and qualitative differences between 7A7 and AG1478-induced apoptosis. EGFR blocking by 7A7 not only prompted a higher proapoptotic effect on tumor metastases compared with AG1478, but also was able to induce apoptosis with immunogenic potential in an Fc-independent manner. As expected, 7A7 but not AG1478 stimulated exposure of danger signals on tumor cells. Subcutaneous injection of 7A7-treated tumor cells induced an antitumor immune response. This is the first report, to our knowledge, of a tumor-specific CTL response generated by Ab-mediated EGFR inhibition, suggesting an important contribution of immunogenic apoptosis to this effect.
Assuntos
Anticorpos Monoclonais/fisiologia , Especificidade de Anticorpos/fisiologia , Apoptose/imunologia , Carcinoma Pulmonar de Lewis/imunologia , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/imunologia , Animais , Anticorpos Bloqueadores/fisiologia , Carcinoma Pulmonar de Lewis/patologia , Carcinoma Pulmonar de Lewis/secundário , Linhagem Celular Tumoral , Células Cultivadas , Receptores ErbB/metabolismo , Feminino , Fragmentos Fc das Imunoglobulinas/fisiologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Melanoma Experimental/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Citotóxicos/patologiaRESUMO
Circulating alloantibodies in transplant recipients are often associated with increased Ab-mediated as well as cellular rejection. We tested the hypothesis that alloantibodies facilitate cellular rejection by functioning as opsonins to enhance T cell activation using a BALB/c to C57BL/6 heart or skin transplant model. Long-term heart and skin survival induced with anti-CD154 alone or in combination with donor-specific transfusion (DST), respectively, was abrogated by the presence of anti-K(d) mAbs, and alloreactive T cell activation as well as acute rejection was observed. The prevention of graft acceptance in the skin model was dependent on anti-K(d) binding to and converting DST from tolerigenic to immunogenic. Adoptive transfer of CFSE-labeled TCR-transgenic T cells into B6 recipients treated with anti-CD154/DST revealed the ability of anti-K(d) to enhance the proliferation of anti-K(d)-specific T cells via the indirect pathway as well as of non-K(d)-reactive, recipient MHC-restricted CD4(+) and CD8(+) T cells. Thus, alloantibodies with restricted specificity are able to facilitate the indirect presentation as well as the cross-presentation of a larger repertoire of "linked" donor-derived Ags. These observations highlight the ability of alloantibodies to function not only in classical humoral rejection but also as opsonins that facilitate the CD40-CD154-independent activation of alloreactive T cells.
Assuntos
Regulação para Baixo/imunologia , Isoanticorpos/fisiologia , Ativação Linfocitária/imunologia , Subpopulações de Linfócitos T/imunologia , Tolerância ao Transplante/imunologia , Regulação para Cima/imunologia , Animais , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/fisiologia , Ligante de CD40/imunologia , Regulação para Baixo/genética , Feminino , Rejeição de Enxerto/genética , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/patologia , Sobrevivência de Enxerto/genética , Sobrevivência de Enxerto/imunologia , Antígenos H-2/imunologia , Isoanticorpos/metabolismo , Transfusão de Leucócitos/métodos , Ativação Linfocitária/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Transplante de Pele/imunologia , Transplante de Pele/métodos , Transplante de Pele/patologia , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/transplante , Tolerância ao Transplante/genética , Regulação para Cima/genéticaRESUMO
Neuromyelitis optica (NMO) is thought to be caused by immunoglobulin G autoantibodies (NMO-IgG) against astrocyte water channel aquaporin-4 (AQP4). A recent study (Hinson et al. (2012) Proc Natl Acad Sci USA 109:1245-1250) reported that NMO-IgG inhibits AQP4 water permeability directly and causes rapid cellular internalization of the M1 but not M23 isoform of AQP4, resulting in AQP4 clustering, enhanced complement-dependent cytotoxicity, and tissue swelling. Here, we report evidence challenging this proposed mechanism of NMO-IgG-mediated pathology. We measured osmotic water permeability by stopped-flow light scattering on plasma membrane vesicles isolated from AQP4-expressing CHO cells, an approach that can detect changes in water permeability as small as 5% and is not confounded by internalization effects. We found similar single-molecule water permeability for M1-AQP4 tetramers and M23-AQP4 clusters (orthogonal arrays of particles, OAPs). Exposure of AQP4 to high concentrations of NMO-IgG from six seropositive NMO patients, and to high-affinity recombinant monoclonal NMO antibodies, did not reduce AQP4 water permeability. Also, NMO-IgG did not reduce water permeability in AQP4-reconstituted proteoliposomes. In transfected cells expressing M1- or M23-AQP4 individually, NMO-IgG caused more rapid internalization of M23- than M1-AQP4. In cells coexpressing both isoforms, M1- and M23-AQP4 comingled in OAPs that were internalized together in response to NMO-IgG. Super-resolution imaging and native gel electrophoresis showed that the size of AQP4 OAPs was not altered by NMO sera or recombinant NMO antibodies. We conclude that NMO-IgG does not: (i) inhibit AQP4 water permeability, (ii) cause preferential internalization of M1-AQP4, or (iii) cause intramembrane AQP4 clustering.
Assuntos
Aquaporina 4/metabolismo , Autoanticorpos/fisiologia , Permeabilidade da Membrana Celular/fisiologia , Membrana Celular/metabolismo , Imunoglobulina G/fisiologia , Neuromielite Óptica/imunologia , Animais , Anticorpos Monoclonais/fisiologia , Aquaporina 4/antagonistas & inibidores , Aquaporina 4/química , Células CHO , Linhagem Celular Tumoral , Membrana Celular/patologia , Cricetinae , Cricetulus , Humanos , Complexos Multiproteicos/antagonistas & inibidores , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Neuromielite Óptica/metabolismo , Neuromielite Óptica/patologia , Ligação Proteica/fisiologia , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Água/metabolismoRESUMO
Intestinal remodeling and stricture formation is a complication of inflammatory bowel disease (IBD) that often requires surgical intervention. Although eosinophils are associated with mucosal remodeling in other organs and are increased in IBD tissues, their role in IBD-associated remodeling is unclear. Histological and molecular features of ileitis and remodeling were assessed using immunohistochemical, histomorphometric, flow cytometric, and molecular analysis (real-time RT-PCR) techniques in a murine model of chronic eosinophilic ileitis. Collagen protein was assessed by Sircol assay. Using a spontaneous eosinophilic Crohn's-like mouse model SAMP1/SkuSlc, we demonstrate an association between ileitis progression and remodeling over the course of 40 weeks. Mucosal and submucosal eosinophilia increased over the time course and correlated with increased histological inflammatory indices. Ileitis and remodeling increased over the 40 weeks, as did expression of fibronectin. CCR3-specific antibody-mediated reduction of eosinophils resulted in significant decrease in goblet cell hyperplasia, muscularis propria hypertrophy, villus blunting, and expression of inflammatory and remodeling genes, including fibronectin. Cellularity of local mesenteric lymph nodes, including T- and B-lymphocytes, was also significantly reduced. Thus, eosinophils participate in intestinal remodeling, supporting eosinophils as a novel therapeutic target.
Assuntos
Eosinófilos/fisiologia , Ileíte/fisiopatologia , Receptores CCR3/antagonistas & inibidores , Animais , Anti-Inflamatórios/farmacologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/fisiologia , Quimiocina CCL11/metabolismo , Quimiocina CCL24/metabolismo , Doença Crônica , Citocinas/metabolismo , Dexametasona/farmacologia , Feminino , Fibrose , Ileíte/tratamento farmacológico , Ileíte/patologia , Imunoglobulina G/farmacologia , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos , Mucosa/patologia , Permeabilidade , Receptores CCR3/imunologia , Receptores CCR3/metabolismoRESUMO
Accumulating evidence has designated B cells as central players in the pathogenesis of immune diseases. In the late 1990s, anti-CD20 monoclonal antibodies were developed for the treatment of B-cell non-Hodgkin lymphomas, offering the opportunity to efficiently deplete the B-cell compartment for therapeutic immunointerventions. Several studies have since established the beneficial effect of this drug on the course of a wide range of immune diseases. However, paradoxically, it has also been reported that rituximab sometimes worsens the symptoms of the very same conditions. The explanation that reconciles such apparently conflicting results has recently emerged from basic studies, which demonstrate that (1) B cells are also endowed with immune-regulatory properties and (2) the opposing contributions of B cells may overlap during the course of the disease. Caution should therefore be exercised when considering B-cell depletion because the therapeutic effect will depend on the relative contributions of the opposing B-cell activities at the time of the drug administration.
Assuntos
Anticorpos Monoclonais/fisiologia , Linfócitos B/fisiologia , Doenças do Sistema Imunitário/etiologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Murinos , Antígenos CD20/imunologia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linfócitos B/imunologia , Humanos , Doenças do Sistema Imunitário/imunologia , Imunoterapia/métodos , Linfoma de Células B/complicações , Linfoma de Células B/etiologia , Linfoma de Células B/imunologia , Linfoma de Células B/terapia , Modelos Biológicos , Rituximab , Resultado do TratamentoRESUMO
The blockade of immune suppression against antitumor responses is a particularly attractive strategy when combined with agents that promote tumor-specific CTLs. In this study, we have attempted to further improve the CTL induction and potent antitumor efficacy of a combination mAb-based therapy (termed "trimAb therapy") that comprises tumor cell death-inducing anti-death receptor 5 mAb and immune activating anti-CD40 and anti-CD137 mAbs. Among trimAb-treated tumors, the infiltration of CD4(+) Foxp3(+) cells was greater in progressing tumors compared with stable tumors. Blockade of CTLA-4 (CD152)-mediated signals by an antagonistic mAb substantially increased the tumor rejection rate of trimAb therapy, although the immune responses of draining lymph node cells were not augmented. Interestingly, by comparison, additional treatment with agonistic anti-glucocorticoid-induced TNF receptor mAb, antagonistic anti-programmed death-1 (CD279) mAb, or agonistic anti-OX40 (CD134) mAb significantly augmented immune responses of draining lymph node cells, but did not augment the therapeutic effect of trimAb. CD4 T cell depletion reduced the antitumor effect of anti-CTLA-4 mAb treatment alone, but did not reduce the tumor rejection rate of trimAb in conjunction with anti-CTLA-4 mAb. Thus, the blockade of the CTLA-4-mediated inhibitory signal in tumor infiltrating CTL may be the most effective strategy to augment the effect of immune therapies that generate tumor-specific CTL.
Assuntos
Anticorpos Bloqueadores/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/terapia , Animais , Anticorpos Bloqueadores/fisiologia , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/fisiologia , Antígenos CD/imunologia , Antígenos CD/metabolismo , Antígenos de Superfície/imunologia , Proteínas Reguladoras de Apoptose/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Antígeno CTLA-4 , Citotoxicidade Imunológica , Proteína Relacionada a TNFR Induzida por Glucocorticoide , Linfonodos/imunologia , Linfonodos/metabolismo , Linfonodos/patologia , Ativação Linfocitária/imunologia , Depleção Linfocítica , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Receptor de Morte Celular Programada 1 , Transporte Proteico/imunologia , Receptores de Fator de Crescimento Neural/imunologia , Receptores OX40/imunologia , Receptores do Fator de Necrose Tumoral/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/patologiaRESUMO
Our laboratory has developed a process for generating mAbs with selectivity to unique peptides in the context of MHC molecules. Recently, we reported that RL4B, an mAb that we have called a TCR mimic (TCRm) because it recognizes peptide in the context of MHC, has cytotoxic activity in vitro and prevented growth of tumor cells in a prophylactic setting. When presented in the context of HLA-A2, RL4B TCRm recognizes the peptide GVLPALPQV derived from human chorionic gonadotropin (hCG)-beta. In this study, we show that RL4B TCRm has strong binding affinity for the GVLPALPQV peptide/HLA-A2 epitope and fine binding specificity for cells that express endogenous hCGbeta Ag and HLA-A2. In addition, suppression of tumor growth with RL4B TCRm was observed in orthotopic models for breast cancer. Using two aggressive human tumor cell lines, MDA-MB-231 and MCF-7, we provide evidence that RL4B TCRm significantly retards tumor growth, supporting a possible role for TCRm agents in therapeutic settings. Moreover, tumors in mice responded to RL4B TCRm therapy in a dose-dependent manner, eliminating tumors at the highest dose. RL4B TCRm strongly detects the hCGbeta peptide/HLA-A2 epitope in human primary breast tumor tissue, but does not react or reacts weakly with normal breast tissue from the same patient. These results further illustrate the selective nature of TCRm Abs and the clinical relevance of the GVLPALPQV peptide/HLA-A2 epitope expression in tumor cells, because they provide the first evidence that Abs that mimic the TCR can be used to markedly reduce and suppress tumor growth.
Assuntos
Anticorpos Monoclonais/metabolismo , Afinidade de Anticorpos , Especificidade de Anticorpos , Neoplasias da Mama/terapia , Gonadotropina Coriônica Humana Subunidade beta/imunologia , Inibidores do Crescimento/fisiologia , Mimetismo Molecular/imunologia , Fragmentos de Peptídeos/metabolismo , Animais , Anticorpos Monoclonais/fisiologia , Anticorpos Monoclonais/uso terapêutico , Apresentação de Antígeno/genética , Apresentação de Antígeno/imunologia , Antineoplásicos/metabolismo , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/imunologia , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Linhagem Celular Transformada , Linhagem Celular Tumoral , Gonadotropina Coriônica Humana Subunidade beta/metabolismo , Testes Imunológicos de Citotoxicidade , Epitopos de Linfócito T/biossíntese , Epitopos de Linfócito T/genética , Marcação de Genes , Inibidores do Crescimento/uso terapêutico , Antígenos HLA-A/imunologia , Antígenos HLA-A/metabolismo , Antígeno HLA-A2 , Humanos , Camundongos , Camundongos Nus , Fragmentos de Peptídeos/imunologiaRESUMO
Mucosal (nasal or oral) administration of anti-CD3 mAb is effective in ameliorating animal models of autoimmunity (experimental autoimmune encephalomyelitis, diabetes, and lupus) by inducing LAP(+) regulatory T cells. We tested this approach in an arthritis model using type II collagen. We found that nasal anti-CD3 was more effective than oral anti-CD3 in attenuating the development of arthritis. Nasal anti-CD3 induced a LAP(+) regulatory T cell that secreted high levels of IL-10 and suppressed collagen-specific T cell proliferation and anti-collagen Ab production. However, neither nasal nor oral anti-CD3 attenuated disease when given to animals with ongoing arthritis, and this was associated with a lack of induction of LAP(+) regulatory T cells. We found, however, that coadministration of a novel emulsome adjuvant, which enhances Th2 responses, resulted in the induction of LAP(+) regulatory T cells and suppression of ongoing arthritis by both nasal and oral anti-CD3. Suppression of arthritis by mucosal anti-CD3 was associated with less joint damage, a decrease of TNF-alpha and IFN-gamma mRNA expression in joints, and a reduction in anti-collagen Abs. These results demonstrate that mucosal anti-CD3 therapy may serve as a therapeutic approach in arthritis and that the biologic effect is enhanced by an emulsome-based adjuvant.
Assuntos
Adjuvantes Imunológicos/administração & dosagem , Anticorpos Monoclonais/administração & dosagem , Artrite Experimental/prevenção & controle , Complexo CD3/imunologia , Colágeno Tipo II/toxicidade , Precursores de Proteínas/biossíntese , Linfócitos T Reguladores/imunologia , Células Th2/patologia , Fator de Crescimento Transformador beta/biossíntese , Regulação para Cima/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Anticorpos Monoclonais/fisiologia , Artrite Experimental/imunologia , Artrite Experimental/patologia , Diferenciação Celular/imunologia , Células Cultivadas , Emulsões , Masculino , Camundongos , Camundongos Endogâmicos DBA , Mucosa Bucal/imunologia , Mucosa Bucal/metabolismo , Mucosa Nasal/imunologia , Mucosa Nasal/metabolismo , Peptídeos/fisiologia , Precursores de Proteínas/fisiologia , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/patologia , Células Th2/imunologia , Fator de Crescimento Transformador beta/fisiologiaRESUMO
TGF-beta is an ubiquitous cytokine that plays a pivotal role in the maintenance of self-tolerance and prevention of immunopathologies. Under steady-state conditions, TGF-beta keeps naive T cells in a resting state and inhibits Th1 and Th2 cell differentiation. Because rapid generation of Th1 and Th2 effector cells is needed in response to pathogen invasion, how do naive T cells escape from the quiescent state maintained by TGF-beta? We hypothesized that stimulation by strong TCR agonists might interfere with TGF-beta signaling. Using both primary mouse CD4(+) T cells and human Jurkat cells, we observed that strong TCR agonists swiftly suppress TGF-beta signaling. TCR engagement leads to a rapid increase in SMAD7 levels and decreased SMAD3 phosphorylation. We present evidence that TCR signaling hinders SMAD3 activation by inducing recruitment of TGF-betaRs in lipid rafts together with inhibitory SMAD7. This effect is dependent on protein kinase C, a downstream TCR signaling intermediary, as revealed by both pharmacological inhibition and expression of dominant-negative and constitutively active protein kinase C mutants. This work broadens our understanding of the cross-talk occurring between the TCR and TGF-beta signaling pathways and reveals that strong TCR agonists can release CD4 T cells from constitutive TGF-beta signaling. We propose that this process may be of vital importance upon confrontation with microbial pathogens.
Assuntos
Linfócitos T CD4-Positivos/imunologia , Isoenzimas/fisiologia , Ativação Linfocitária/imunologia , Proteína Quinase C/fisiologia , Transdução de Sinais/imunologia , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Fator de Crescimento Transformador beta1/fisiologia , Animais , Anticorpos Monoclonais/fisiologia , Antígenos CD28/imunologia , Complexo CD3/imunologia , Linfócitos T CD4-Positivos/enzimologia , Linfócitos T CD4-Positivos/metabolismo , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Humanos , Isoenzimas/genética , Células Jurkat , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/imunologia , Microdomínios da Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosforilação/imunologia , Proteína Quinase C/genética , Proteína Quinase C-theta , Receptores de Antígenos de Linfócitos T/agonistas , Receptores de Antígenos de Linfócitos T/antagonistas & inibidores , Receptores de Antígenos de Linfócitos T/fisiologia , Fase de Repouso do Ciclo Celular/imunologia , Transdução de Sinais/efeitos dos fármacos , Proteína Smad3/antagonistas & inibidores , Proteína Smad3/metabolismoRESUMO
Dulanermin (rhApo2L/TRAIL) and conatumumab bind to transmembrane death receptors and trigger the extrinsic cellular apoptotic pathway through a caspase-signaling cascade resulting in cell death. Tumor size time series data from rodent tumor xenograft (COLO205) studies following administration of either of these two pro-apoptotic receptor agonists (PARAs) were combined to develop a intracellular-signaling tumor-regression model that includes two levels of signaling: upstream signals unique to each compound (representing initiator caspases), and a common downstream apoptosis signal (representing executioner caspases) shared by the two agents. Pharmacokinetic (PK) models for each drug were developed based on plasma concentration data following intravenous and/or intraperitoneal administration of the compounds and were used in the subsequent intracellular-signaling tumor-regression modeling. A model relating the PK of the two PARAs to their respective and common downstream signals, and to the resulting tumor burden was developed using mouse xenograft tumor size measurements from 448 experiments that included a wide range of dose sizes and dosing schedules. Incorporation of a pro-survival signal--consistent with the hypothesis that PARAs may also result in the upregulation of pro-survival factors that can lead to a reduction in effectiveness of PARAs with treatment--resulted in improved predictions of tumor volume data, especially for data from the long-term dosing experiments.
Assuntos
Anticorpos Monoclonais/fisiologia , Proteínas Reguladoras de Apoptose/agonistas , Proteínas Reguladoras de Apoptose/fisiologia , Líquido Intracelular/fisiologia , Modelos Biológicos , Transdução de Sinais/fisiologia , Ligante Indutor de Apoptose Relacionado a TNF/fisiologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Linhagem Celular Tumoral , Feminino , Humanos , Líquido Intracelular/efeitos dos fármacos , Camundongos , Camundongos Nus , Distribuição Aleatória , Ratos , Indução de Remissão/métodos , Transdução de Sinais/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodosRESUMO
CD40 is a member of the TNF family of receptors that has been shown to play a crucial role in enhancing dendritic cell activity and fostering anti-tumor immune responses. In this study, we demonstrate the in vitro properties and in vivo efficacious activity of the CD40 agonist antibody, CP-870,893. CP-870,893 is a fully human, IgG2 antibody that selectively interacts with CD40 at a site distinct from its ligand-binding region with a KD of 0.4 nM. It enhances the expression of MHC class II, CD54, CD86, and CD23 on human B cells in vitro. CP-870,893 also enhances dendritic cell activity as evidenced by cytokine secretion (IL-12, IL-23, IL-8), the upregulation of CD86 and CD83, and the ability to prime T cells to secrete IFNγ. In SCID-beige mice, a single parenteral injection of CP-870,893 was therapeutically effective against several CD40(pos) human tumors (B-cell lymphoma, breast, colon, and prostate) indicating direct effects on tumor cell survival and/or growth. When mice were co-implanted with human T cells and dendritic cells, the activity of CP-870,893 against CD40(pos) tumors increased, and efficacy was also observed against CD40(neg) and CD40(low) tumors demonstrating the ability of CP-870,893 to enhance anti-tumor immune function in vivo. These studies suggest that CP-870,893 has the potential to be efficacious against a wide range of tumor types through both direct and immune-mediated effects.
Assuntos
Anticorpos Monoclonais/fisiologia , Linfócitos B/imunologia , Antígenos CD40/imunologia , Células Dendríticas/imunologia , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/prevenção & controle , Animais , Anticorpos Monoclonais Humanizados , Linfócitos B/citologia , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/citologia , Citometria de Fluxo , Humanos , Ativação Linfocitária , Camundongos , Camundongos SCIDRESUMO
CD8αß heterodimers are mainly expressed on cytotoxic T lymphocytes. This study demonstrated the detection of CD8αß heterodimers on human monocytes by whole blood erythrocyte lysis method in flow cytometry. Results revealed that CD8αß heterodimers were not produced by monocytes themselves, but were transferred from T cells to monocytes when these cells were coincubated in plasma and with anti-CD8 monoclonal antibody (mAb). For completion of CD8 translocation from T cells to monocytes, cell-to-cell contact between T cells and monocytes, as well as binding of the Fc portion of the anti-CD8 mAb and Fcγ receptor II (FcγRII) on monocytes were required. Furthermore, the dynamism of cell membrane and cytoskeleton were involved in the mechanism of CD8 translocation. Interestingly, CD3 and αßT cell receptor (TCR) were also transferred from T cells to monocytes accompanied by CD8. These phenomena are consistent with Ab-dependent and FcγR-mediated trogocytosis, which is recently recognized as one of the intercellular communication processes of the immune system. Trogocytosis means exchange of plasma membrane including cell surface molecules in conjugates formed between immune cells. Results of this study could provide another model of trogocytosis and clearly indicated that putative plasma factors were critically implicated in the mechanism of Ab-dependent and FcγR-mediated trogocytosis.
Assuntos
Anticorpos Monoclonais/fisiologia , Antígenos CD8/sangue , Monócitos/metabolismo , Receptores de IgG/fisiologia , Linfócitos T/metabolismo , Anticorpos Monoclonais/farmacologia , Complexo CD3/metabolismo , Comunicação Celular , Membrana Celular/metabolismo , Técnicas de Cocultura , Citoesqueleto/metabolismo , Humanos , Plasma , Transporte Proteico , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismoRESUMO
We previously reported that some human antiphospholipid Abs (aPL) in patients with the antiphospholipid syndrome (APS) bind to the homologous enzymatic domains of thrombin and the activated coagulation factor X (FXa). Moreover, some of the reactive Abs are prothrombotic and interfere with inactivation of thrombin and FXa by antithrombin (AT). Considering the enzymatic domain of activated coagulation factor IX (FIXa) is homologous to those of thrombin and FXa, we hypothesized that some aPLs in APS bind to FIXa and hinder AT inactivation of FIXa. To test this hypothesis, we searched for IgG anti-FIXa Abs in APS patients. Once the concerned Abs were found, we studied the effects of the Ab on FIXa inactivation by AT. We found that 10 of 12 patient-derived monoclonal IgG aPLs bound to FIXa and that IgG anti-FIXa Abs in APS patients were significantly higher than those in normal controls (p < 0.0001). Using the mean + 3 SD of 30 normal controls as the cutoff, the IgG anti-FIXa Abs were present in 11 of 38 (28.9%) APS patients. Importantly, 4 of 10 FIXa-reactive monoclonal aPLs (including the B2 mAb generated against beta(2)-glycoprotein I significantly hindered AT inactivation of FIXa. More importantly, IgG from two positive plasma samples were found to interfere with AT inactivation of FIXa. In conclusion, IgG anti-FIXa Ab occurred in approximately 30% of APS patients and could interfere with AT inactivation of FIXa. Because FIXa is an upstream procoagulant factor, impaired AT regulation of FIXa might contribute more toward thrombosis than the dysregulation of the downstream FXa and thrombin.