Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Planta Med ; 90(4): 305-315, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38373705

RESUMEN

Checkpoint blockade immunotherapy has revolutionized cancer treatment, with monoclonal antibodies targeting immune checkpoints, yielding promising clinical benefits. However, with the advent of resistance to immune checkpoint inhibitor treatment in clinical trials, developing next-generation antibodies with potentially increased efficacy is critical. Here, we aimed to generate a recombinant bispecific monoclonal antibody for dual inhibition of programmed cell death protein 1/programmed cell death ligand 1 and cytotoxic T-lymphocyte-associated protein 4 axes. The plant system was used as an alternative platform for bispecific monoclonal antibody production. Dual variable domain immunoglobulin atezolizumab × 2C8 is a plant-derived bispecific monoclonal antibody that combines both programmed cell death ligand 1 and cytotoxic T-lymphocyte-associated protein 4 blockade into a single molecule. Dual variable domain immunoglobulin atezolizumab × 2C8 was transiently expressed in Nicotiana benthamiana and the expression level was determined to be the highest after 4 days of infiltration. The size and assembly of the purified bispecific monoclonal antibody were determined, and its function was investigated in vitro and in vivo. The molecular structures of plant-produced dual variable domain immunoglobulin atezolizumab × 2C8 are as expected, and it was mostly present as a monomer. The plant-produced dual variable domain immunoglobulin atezolizumab × 2C8 showed in vitro binding to programmed cell death ligand 1 and cytotoxic T-lymphocyte-associated protein 4 proteins. The antitumor activity of plant-produced bispecific monoclonal antibody was tested in vivo by treating humanized Balb/c mice bearing a CT26 colorectal tumor. Plant-produced dual variable domain immunoglobulin atezolizumab × 2C8 significantly inhibited tumor growth by reducing tumor volume and weight. Body weight changes indicated that the plant-produced bispecific monoclonal antibody was safe and tolerable. Overall, this proof of concept study demonstrated the viability of plants to produce functional plant-based bispecific immunotherapy.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias Colorrectales , Neoplasias , Ratones , Animales , Antígeno CTLA-4/uso terapéutico , Antígeno B7-H1/uso terapéutico , Ligandos , Neoplasias/tratamiento farmacológico , Anticuerpos Monoclonales/farmacología , Anticuerpos Biespecíficos/farmacología , Anticuerpos Biespecíficos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico
2.
J Cancer Res Clin Oncol ; 148(1): 177-190, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34741682

RESUMEN

PURPOSE: CD19 is a cell surface protein that is found on both healthy and malignant B cells. Accordingly, it has become an important target for novel treatments for non-Hodgkin lymphomas and B-cell leukaemia. Three anti-CD19 monoclonal antibodies with distinct mechanisms of action have been developed for the treatment of B-cell malignancies. METHODS: We reviewed the preclinical and clinical data on the development of the newly approved anti-CD19 monoclonal antibodies blinatumomab, tafasitamab and loncastuximab tesirine, and consider their place in the treatment of relapsed or refractory B-cell malignancies. RESULTS: Blinatumomab is a bispecific T-cell engager that binds to both CD19 on B cells and CD3 on T cells, facilitating antibody-dependent cytotoxicity. Blinatumomab significantly prolongs overall survival in patients with relapsed or refractory B-cell acute lymphoblastic leukaemia, although cytokine release syndrome and severe neurotoxicity may necessitate discontinuation. Tafasitamab, which has modified anti-CD19 Fab and Fc regions, has significantly enhanced affinity for both CD19 and effector cell receptors compared with unmodified anti-CD19. In L-MIND, tafasitamab plus lenalidomide provided an overall response rate (ORR) of 57.5% in patients with relapsed or refractory diffuse large B-cell lymphoma (DLBCL) in patients non-transplant eligible. Loncastuximab tesirine is an antibody-drug conjugate that has been studied as monotherapy and in combination with ibrutinib in 3L + relapsed or refractory DLBCL. The ORR was 48.3% in a phase II trial of loncastuximab tesirine. The optimal place of anti-CD19 monoclonal antibodies in therapy has yet to be determined, but the prospect of improved outcomes for at least some patients with treatment-resistant B-cell malignancies appears likely, particularly in those with limited therapeutic options and poor prognosis.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antígenos CD19/inmunología , Antineoplásicos/uso terapéutico , Benzodiazepinas/uso terapéutico , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Adenina/análogos & derivados , Adenina/uso terapéutico , Linfocitos B/inmunología , Linfocitos B/metabolismo , Evaluación Preclínica de Medicamentos , Humanos , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Piperidinas/uso terapéutico
3.
Ann Hematol ; 100(11): 2727-2732, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34331563

RESUMEN

Treatment options for relapsed or refractory B-lymphoblastic leukaemia (r/r B-ALL) are limited and the prognosis of these patients remains dismal, but novel immunotherapeutic options such as the anti-CD22 antibody-drug-conjugate Inotuzumab-Ozogamicin (InO) have improved outcomes in these patients. Flow cytometry is essential to assess antigen-expression prior to treatment initiation of antigen-directed immunotherapies. Here, we present flow cytometric and clinical data of three adult patients with r/r B-ALL who failed treatment with InO associated with reduced or lost antigen-expression. In addition, we present comparative data on two different diagnostic CD22-specific antibody clones that exhibit significant differences in staining intensities.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Linfocitos B/química , Inotuzumab Ozogamicina/uso terapéutico , Subgrupos Linfocitarios/química , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Lectina 2 Similar a Ig de Unión al Ácido Siálico/análisis , Adulto , Anciano de 80 o más Años , Aloinjertos , Anticuerpos Biespecíficos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Linfocitos B/patología , Células Clonales , Femenino , Citometría de Flujo , Trasplante de Células Madre Hematopoyéticas , Humanos , Mesilato de Imatinib/administración & dosificación , Inmunofenotipificación , Subgrupos Linfocitarios/patología , Masculino , Persona de Mediana Edad , Leucemia-Linfoma Linfoblástico de Células Precursoras B/sangre , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Recurrencia , Terapia Recuperativa , Sorafenib/uso terapéutico , Insuficiencia del Tratamiento , Adulto Joven
4.
Value Health ; 24(6): 839-845, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34119082

RESUMEN

OBJECTIVES: To evaluate alternative methods to calculate and/or attribute economic surplus in the cost-effectiveness analysis of single or short-term therapies. METHODS: We performed a systematic literature review of articles describing alternative methods for cost-effectiveness analysis of potentially curative therapies whose assessment using traditional methods may suggest unaffordable valuations owing to the magnitude of estimated long-term quality-adjusted life-year (QALY) gains or cost offsets. Through internal deliberation and discussion with staff at the Health Technology Assessment bodies in England and Canada, we developed the following 3 alternative methods for further evaluation: (1) capping annual costs in the comparator arm at $150 000 per year; (2) "sharing" the economic surplus with the health sector by apportioning only 50% of cost offsets or 50% of cost offsets and QALY gains to the value of the therapy; and (3) crediting the therapy with only 12 years of the average annual cost offsets or cost offsets and QALY gains over the lifetime horizon. The impact of each alternative method was evaluated by applying it in an economic model of 3 hypothetical condition-treatment scenarios meant to reflect a diversity of chronicity and background healthcare costs. RESULTS: The alternative with greatest impact on threshold price for the fatal pediatric condition spinal muscular atrophy type 1 was the 12-year cutoff scenario. For a hypothetical one-time treatment for hemophilia A, capping cost offsets at $150 000 per year had the greatest impact. For chimeric antigen receptor T-cell treatment of non-Hodgkin's lymphoma, capping cost offsets or using 12-year threshold had little impact, whereas 50% sharing of surplus including QALY gains and cost offsets greatly reduced threshold pricing. CONCLUSIONS: Health Technology Assessment bodies and policy makers will wrestle with how to evaluate single or short-term potentially curative therapies and establish pricing and payment mechanisms to ensure sustainability. Scenario analyses using alternative methods for calculating and apportioning economic surplus can provide starkly different assessment results. These methods may stimulate important societal dialogue on fair pricing for these novel treatments.


Asunto(s)
Quimioterapia/economía , Terapia Genética/economía , Costos de la Atención en Salud , Inmunoterapia Adoptiva/economía , Evaluación de la Tecnología Biomédica/economía , Anticuerpos Biespecíficos/economía , Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales Humanizados/economía , Anticuerpos Monoclonales Humanizados/uso terapéutico , Productos Biológicos/economía , Productos Biológicos/uso terapéutico , Ahorro de Costo , Análisis Costo-Beneficio , Costos de los Medicamentos , Terapia Genética/efectos adversos , Hemofilia A/tratamiento farmacológico , Hemofilia A/economía , Humanos , Inmunoterapia Adoptiva/efectos adversos , Linfoma no Hodgkin/economía , Linfoma no Hodgkin/terapia , Modelos Económicos , Años de Vida Ajustados por Calidad de Vida , Proteínas Recombinantes de Fusión/economía , Proteínas Recombinantes de Fusión/uso terapéutico , Inducción de Remisión , Atrofias Musculares Espinales de la Infancia/economía , Atrofias Musculares Espinales de la Infancia/genética , Atrofias Musculares Espinales de la Infancia/terapia , Factores de Tiempo , Resultado del Tratamiento
5.
Lancet ; 397(10274): 630-640, 2021 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-33460559

RESUMEN

Congenital haemophilia A (factor VIII deficiency) and B (factor IX deficiency) are X-linked bleeding disorders. Replacement therapy has been the cornerstone of the management of haemophilia, aiming to reduce the mortality and morbidity of chronic crippling arthropathy. Frequent intravenous injections are burdensome and costly for patients, consequently with poor adherence and restricted access to therapy for many patients worldwide. Bioengineered clotting factors with enhanced pharmacokinetic profiles can reduce the burden of treatment. However, replacement therapy is associated with a risk for inhibitor development that adversely affects bleeding prevention and outcomes. Novel molecules that are subcutaneously delivered provide effective prophylaxis in the presence or absence of inhibitors, either substituting for the procoagulant function of clotting factors (eg, emicizumab) or targeting the natural inhibitors of coagulation (ie, antithrombin, tissue factor pathway inhibitor, or activated protein C). The ultimate goal of haemophilia treatment would be a phenotypical cure achievable with gene therapy, currently under late phase clinical investigation.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Factor VIII/uso terapéutico , Terapia Genética , Hemofilia A/terapia , Hemofilia B/terapia , Anticuerpos Neutralizantes , Factor IX/uso terapéutico , Factor VIIa/uso terapéutico , Semivida , Hemorragia/tratamiento farmacológico , Hemorragia/prevención & control , Humanos , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Inyecciones Subcutáneas , Polietilenglicoles/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Proteínas Recombinantes/uso terapéutico , Albúmina Sérica/uso terapéutico , Factor de von Willebrand/metabolismo
6.
Immunotherapy ; 13(2): 125-141, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33172323

RESUMEN

Aim: We report results of a first-in-human study of pasotuxizumab, a PSMA bispecific T-cell engager (BiTE®) immune therapy mediating T-cell killing of tumor cells in patients with advanced castration-resistant prostate cancer. Patients & methods: We assessed once-daily subcutaneous (SC) pasotuxizumab. All SC patients developed antidrug antibodies; therefore, continuous intravenous (cIV) infusion was assessed. Results: A total of 47 patients received pasotuxizumab (SC: n = 31, 0.5-172 µg/d; cIV: n = 16, 5-80 µg/d). The SC maximum tolerated dose was 172.0 µg/d. A sponsor change stopped the cIV cohort early; maximum tolerated dose was not determined. PSA responders occurred (>50% PSA decline: SC, n = 9; cIV, n = 3), including two long-term responders. Conclusion: Data support pasotuxizumab safety in advanced castration-resistant prostate cancer and represent evidence of BiTE monotherapy efficacy in solid tumors. Clinical trial registration: NCT01723475 (ClinicalTrials.gov).


Asunto(s)
Anticuerpos Biespecíficos , Antineoplásicos Inmunológicos , Neoplasias de la Próstata Resistentes a la Castración , Anciano , Anciano de 80 o más Años , Humanos , Masculino , Persona de Mediana Edad , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/farmacocinética , Anticuerpos Biespecíficos/uso terapéutico , Antígenos de Superficie/inmunología , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/farmacocinética , Antineoplásicos Inmunológicos/uso terapéutico , Biomarcadores de Tumor/sangre , Complejo CD3/inmunología , Glutamato Carboxipeptidasa II/inmunología , Inmunoterapia , Infusiones Intravenosas , Inyecciones Subcutáneas , Dosis Máxima Tolerada , Neoplasias de la Próstata Resistentes a la Castración/sangre , Neoplasias de la Próstata Resistentes a la Castración/inmunología , Neoplasias de la Próstata Resistentes a la Castración/patología , Neoplasias de la Próstata Resistentes a la Castración/terapia , Resultado del Tratamiento
7.
Theranostics ; 10(23): 10498-10512, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32929362

RESUMEN

Rationale: Current traditional treatment options are frequently ineffective to fight against ovarian cancer due to late diagnosis and high recurrence. Therefore, there is a vital need for the development of novel therapeutic agents. B7H3, an immune checkpoint protein, is highly expressed in various cancers, representing it a promising target for cancer immunotherapy. Although targeting B7H3 by bispecific T cell-engaging antibodies (BiTE) has achieved successes in hematological malignancies during recent years, attempts to use them for the treatment of solid cancers are less favorable, in part due to the heterogeneity of tumors. Sorafenib is an unselective inhibitor of multiple kinases currently being tested in clinical trials for several tumors, including ovarian cancer which showed limited activity and inevitable side effect for ovarian cancer treatment. However, it is able to enhance antitumor immune response, which indicates sorafenib may improve the efficiency of immunotherapy. Methods: We evaluated the expression of B7H3 in ovarian cancer using online database and validated its expression of tumor tissues by immunohistochemistry staining. Then, B7H3 expression and the effects of sorafenib on ovarian cancer cell lines were determined by flow cytometry. In addition, 2D and 3D ovarian cancer models were established to test the combined therapeutic effect in vitro. Finally, the efficiency of B7H3×CD3 BiTE alone and its combination with sorafenib were evaluated both in vitro and in vivo. Results: Our data showed that B7H3 was highly expressed in ovarian cancer compared with normal samples. Treatment with sorafenib inhibited ovarian cancer cell proliferation and induced a noticeable upregulation of B7H3 expression level. Further study suggested that B7H3×CD3 BiTE was effective in mediating T cell killing to cancer cells. Combined treatment of sorafenib and B7H3×CD3 BiTE had synergistic anti-tumor effects in ovarian cancer models. Conclusions: Overall, our study indicates that combination therapy with sorafenib and B7H3×CD3 BiTE may be a new therapeutic option for the further study of preclinical treatment of OC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Antígenos B7/antagonistas & inhibidores , Carcinoma Epitelial de Ovario/terapia , Neoplasias Ováricas/terapia , Sorafenib/farmacología , Animales , Anticuerpos Biespecíficos/farmacología , Anticuerpos Biespecíficos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Antígenos B7/análisis , Antígenos B7/metabolismo , Complejo CD3/antagonistas & inhibidores , Carcinoma Epitelial de Ovario/inmunología , Carcinoma Epitelial de Ovario/mortalidad , Carcinoma Epitelial de Ovario/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Conjuntos de Datos como Asunto , Sinergismo Farmacológico , Femenino , Células HEK293 , Humanos , Estimación de Kaplan-Meier , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Recurrencia Local de Neoplasia , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/mortalidad , Neoplasias Ováricas/patología , Ovario/patología , Sorafenib/uso terapéutico , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Expert Rev Clin Immunol ; 16(7): 689-709, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32536227

RESUMEN

INTRODUCTION: Treatment of colorectal cancer as one of the most commonly diagnosed and a frequent cause of cancer-related deaths is of great challenges in health-related issues. AREAS COVERED: Immunotherapy is the fourth pillar of cancer treatment which provides more novel therapeutic options with expanding investigational potentials. One of the modalities in immunotherapy is the use of bispecific antibodies. Despite demonstrating many promising roles, it still needs more advanced studies to identify the actual pros and cons. In this review, the application of bispecific antibody in the treatment of colorectal cancer has been explained, based on preclinical and clinical studies. The literature search was conducted mainly through PubMed in June and September 2019. EXPERT OPINION: Bispecific antibody is in its early stages in colorectal cancer treatment, requiring modern technologies in manufacturing, better biomarkers and more specific target antigens, more studies on individual genetic variations, and conducting later phase clinical trials and systematic reviews to achieve better survival benefits.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Vacunas contra el Cáncer/inmunología , Neoplasias Colorrectales/terapia , Inmunoterapia/tendencias , Linfocitos T/inmunología , Animales , Ensayos Clínicos como Asunto , Neoplasias Colorrectales/inmunología , Evaluación Preclínica de Medicamentos , Humanos
9.
Thromb Haemost ; 120(6): 968-976, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32384547

RESUMEN

BACKGROUND: Emicizumab prophylaxis is a promising treatment that reduces bleeding events in severely affected patients with hemophilia A (PwHA). It is anticipated that emicizumab could be similarly effective in mild/moderate PwHA (PwMHA) although this effect has not been investigated. AIM: We evaluated ex vivo coagulant effects of emicizumabin PwMHA. METHODS: Clot waveform analysis (CWA) triggered by prothrombin time/activated partial prothrombin time-mixed reagents was utilized to examine coagulant effects of emicizumabin factor (F)VIII-deficient plasma mixed with recombinant (r)FVIIIand in native plasmas from 16 PwMHA. The CWA parameter, adjusted-|min1| (Ad|min1|), was used. Increases in Ad|min1| (ΔAd|min1|) mediated by emicizumab were calculated from the slopes of regression lines in the presence of rFVIII. RESULTS: Ad|min1| in FVIII-deficient plasma with various concentrations of rFVIII negatively correlated with ΔAd|min1|by adding emicizumab, and these data were defined as standard reference values. Ad|min1| (4.57 ± 0.50) in 16 PwMHA increased to 5.05 ± 0.54 and 5.37 ± 0.60 by adding emicizumab at 50 and 100 µg/mL, respectively, but remained lower than the normal range (7.22 ± 0.21). ΔAd|min1| levels were 1.5 to 2-fold higher in five cases and 0.4 to 0.6-fold lower in four cases, compared with reference values determined by rFVIII. In some cases, genetic analyses suggested that specific point mutations could have contributed to these findings. Further studies using rFVIII mutants indicated, however, that the differences in ΔAd|min1| were not related to individual FVIII gene defects. CONCLUSION: Emicizumab enhances coagulation potential in PwMHA. Assessment of ex vivo coagulant activity of emicizumab could be helpful for predicting coagulant potentials prior to treatment in these patients.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Hemofilia A/sangre , Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Factor VIII/genética , Factor VIII/farmacología , Hemofilia A/tratamiento farmacológico , Hemofilia A/genética , Hemorragia/prevención & control , Humanos , Técnicas In Vitro , Mutación Missense , Tiempo de Tromboplastina Parcial , Plasma , Tiempo de Protrombina , Proteínas Recombinantes/farmacología
10.
Front Immunol ; 11: 476, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32265927

RESUMEN

Replacement therapy with coagulation factor VIII (FVIII) represents the current clinical treatment for patients affected by hemophilia A (HA). This treatment while effective is, however, hampered by the formation of antibodies which inhibit the activity of infused FVIII in up to 30% of treated patients. Immune tolerance induction (ITI) protocols, which envisage frequent infusions of high doses of FVIII to confront this side effect, dramatically increase the already high costs associated to a patient's therapy and are not always effective in all treated patients. Therefore, there are clear unmet needs that must be addressed in order to improve the outcome of these treatments for HA patients. Taking advantage of preclinical mouse models of hemophilia, several strategies have been proposed in recent years to prevent inhibitor formation and eradicate the pre-existing immunity to FVIII inhibitor positive patients. Herein, we will review some of the most promising strategies developed to avoid and eradicate inhibitors, including the use of immunomodulatory drugs or molecules, oral or transplacental delivery as well as cell and gene therapy approaches. The goal is to improve and potentiate the current ITI protocols and eventually make them obsolete.


Asunto(s)
Factor VIII/inmunología , Hemofilia A/tratamiento farmacológico , Tolerancia Inmunológica/efectos de los fármacos , Factores Inmunológicos/uso terapéutico , Isoanticuerpos/inmunología , Traslado Adoptivo , Animales , Anticuerpos Biespecíficos/farmacología , Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Portadores de Fármacos , Composición de Medicamentos , Evaluación Preclínica de Medicamentos , Factor IXa/inmunología , Factor VIII/administración & dosificación , Factor VIII/uso terapéutico , Factor X/inmunología , Femenino , Terapias Fetales , Terapia Genética , Hemofilia A/inmunología , Hemofilia A/terapia , Humanos , Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Inmunoterapia Adoptiva , Isoanticuerpos/biosíntesis , Tejido Linfoide/inmunología , Ratones , Modelos Animales , Células Vegetales , Embarazo , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/uso terapéutico , Linfocitos T Reguladores/trasplante
11.
BioDrugs ; 34(2): 111-119, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31916225

RESUMEN

In autoimmune diseases, a highly complex network comprising diverse cytokines and their receptors on immune cells drives the inflammatory response. A number of therapeutic antibodies targeting these disease-related molecules have been approved for the treatment of autoimmune diseases. Bispecific antibodies (bsAbs), with binding specificity for two different target molecules, have recently been developed for a range of autoimmune diseases, such as rheumatoid arthritis, systemic lupus erythematosus, and psoriasis, and tested in clinical trials. This review briefly describes the three main categories of bsAb structures developed for autoimmune diseases, including immunoglobulin G (IgG)-like, natural IgG, and tandem antibody fragment formats. The bsAbs developed and evaluated to date mainly target the depletion of T or B cells, the inhibition of T cell differentiation or activation, or the neutralization of proinflammatory cytokines. The clinical evaluation of bsAbs in autoimmune diseases is ongoing, with both successes (phase II trials of obexelimab in systemic lupus erythematosus) and failures (phase II trials of lutikizumab in osteoarthritis and romilkimab in idiopathic pulmonary fibrosis), and this review aims to provide a comprehensive, up-to-date summary of the clinical progress of bsAbs in this therapeutic area. Although many challenges remain, bsAbs offer new therapeutic options in the future direction of autoimmune disease treatments.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Enfermedades Autoinmunes/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Animales , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/inmunología , Ensayos Clínicos como Asunto , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Humanos , Terapia Molecular Dirigida , Resultado del Tratamiento
12.
Nat Rev Drug Discov ; 18(8): 585-608, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31175342

RESUMEN

The term bispecific antibody (bsAb) is used to describe a large family of molecules designed to recognize two different epitopes or antigens. BsAbs come in many formats, ranging from relatively small proteins, merely consisting of two linked antigen-binding fragments, to large immunoglobulin G (IgG)-like molecules with additional domains attached. An attractive bsAb feature is their potential for novel functionalities - that is, activities that do not exist in mixtures of the parental or reference antibodies. In these so-called obligate bsAbs, the physical linkage of the two binding specificities creates a dependency that can be temporal, with binding events occurring sequentially, or spatial, with binding events occurring simultaneously, such as in linking an effector to a target cell. To date, more than 20 different commercialized technology platforms are available for bsAb creation and development, 2 bsAbs are marketed and over 85 are in clinical development. Here, we review the current bsAb landscape from a mechanistic perspective, including a comprehensive overview of the pipeline.


Asunto(s)
Anticuerpos Biespecíficos , Diseño de Fármacos , Neoplasias , Animales , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/uso terapéutico , Sitios de Unión de Anticuerpos , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Humanos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología
13.
Rinsho Ketsueki ; 60(5): 475-479, 2019.
Artículo en Japonés | MEDLINE | ID: mdl-31168016

RESUMEN

Regular prophylaxis using factor (F) VIII products to prevent bleeding in patients with hemophilia A (PWHAs) results in markedly suppressed onset of arthropathy and greatly contributes to improved quality of life. However, some issues remain with the use of clotting factor replacement therapy. The need for multiple intravenous infusions is associated with substantial mental and physical burden, and the inhibitor development results in difficulty of hemostatic management. To overcome these unmet needs, a recombinant humanized anti-FIXa/FX bispecific antibody mimicking FVIIIa function (emicizumab) was created. In phase 1/2 clinical studies, Japanese patients with PWHAs were treated with once-weekly subcutaneous administration of emicizumab. The annual bleeding rates were markedly reduced, irrespective of inhibitor use. A phase 3 global study for PWHA with inhibitor demonstrated a statistically significant efficacy, whereas thrombotic microangiopathy and thromboembolism were reported in 5 patients treated with emicizumab concomitantly with multiple infusions of activated prothrombin complex concentrates. In Japan, emicizumab (HEMLIBRA®) was approved for treatment of PWHAs with inhibitor on March 2018. In conclusion, emicizumab has promising features. Its subcutaneous bioavailability and long half-life (T1/2; approximately 30 days) enable effective bleeding prophylactic treatment at inhibitor status. Additional studies are warranted to establish clinical data on its safety and to define suitable assays for hemostatic monitoring.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Hemofilia A/tratamiento farmacológico , Hemostáticos/uso terapéutico , Ensayos Clínicos como Asunto , Factor VIII , Humanos , Japón , Calidad de Vida
14.
Expert Rev Hematol ; 12(7): 515-524, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31130012

RESUMEN

Introduction: Hemophilia A (HA) is an inherited bleeding disorder that, if not properly treated, is associated with debilitating joint damage due to recurrent hemarthroses as well as life-threatening bleeds including intracranial hemorrhage. For decades, the only method to prevent bleeding events was to infuse factor (F) VIII concentrates intravenously two to three times weekly. Although successful in reducing bleeding frequency, preventing a high proportion of joint disease, and extending life expectancy, standard continuous prophylaxis with FVIII is burdensome and insufficiently effective at preventing long-term joint dysfunction in some patients. In October 2018, the Federal Drug Administration approved a novel agent, emicizumab-kxwh, for the treatment of patients with HA without inhibitors. Areas covered: In this article, the preclinical and clinical development of emicizumab-kxwh will be reviewed. Data from licensure phase 3 clinical trials will be reviewed in detail discussing issues of both safety and efficacy. Expert opinion: As emicizumab-kxwh leads the way of a shift in treatment paradigm for patients with HA without inhibitors, a number of questions remain, including the impact of emicizumab-kxwh on joint and bone health, inhibitor development, and thrombotic risk. Ultimately, time and clinical investigation will be able to elucidate the influence of emicizumab-kxwh in these areas.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Hemofilia A/tratamiento farmacológico , Animales , Anticuerpos Biespecíficos/administración & dosificación , Anticuerpos Biespecíficos/efectos adversos , Anticuerpos Biespecíficos/farmacocinética , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/farmacocinética , Antineoplásicos Inmunológicos/administración & dosificación , Antineoplásicos Inmunológicos/efectos adversos , Antineoplásicos Inmunológicos/farmacocinética , Inhibidores de Factor de Coagulación Sanguínea/inmunología , Ensayos Clínicos como Asunto , Terapia Combinada , Evaluación Preclínica de Medicamentos , Factor VIII/inmunología , Hemartrosis/etiología , Hemartrosis/prevención & control , Hemartrosis/terapia , Hemofilia A/complicaciones , Hemofilia A/inmunología , Hemorragia/etiología , Hemorragia/prevención & control , Hemorragia/terapia , Humanos , Isoanticuerpos/sangre , Isoanticuerpos/inmunología , Resultado del Tratamiento
15.
Methods Mol Biol ; 1884: 335-347, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30465214

RESUMEN

Development of antibody-based immunotherapeutics has progressed from direct tumor-targeting, with antibodies such as rituximab, to blocking of immune checkpoints to reactivate antitumor immunity. In addition, bispecific antibodies/antibody fragments are also of great interest in cancer therapy, as these constructs have the ability to redirect immune effector cells to cancer targets and, thereby, enhance therapeutic efficacy. A number of bispecific antibody formats have been reported, with the first FDA-approved bispecific antibody being blinatumomab, a so-called bispecific T cell engager (BiTE), which redirects and potently activates T cell immune responses. Recently, we described an additional novel bispecific antibody derivative, termed RTX-CD47, which was designed to inhibit the innate immune checkpoint CD47-SIRPα only on -positive cancer cells. RTX-CD47 contains two antibody fragments in tandem and has monovalent binding specificity for CD47 and . Only upon dual binding to and CD47 RTX-CD47 blocks CD47 "Don't eat me" signaling. Here, we provide a detailed protocol for the construction and functional evaluation of such a bispecific antibody derivative.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Antineoplásicos/farmacología , Bioensayo/métodos , Neoplasias/tratamiento farmacológico , Proteínas Recombinantes de Fusión/inmunología , Animales , Anticuerpos Biespecíficos/uso terapéutico , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/inmunología , Antígenos de Diferenciación/metabolismo , Antineoplásicos/uso terapéutico , Bioensayo/instrumentación , Antígeno CD47/genética , Antígeno CD47/inmunología , Antígeno CD47/metabolismo , Células CHO , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Separación Celular/instrumentación , Separación Celular/métodos , Cromatografía de Afinidad/instrumentación , Cromatografía de Afinidad/métodos , Cricetulus , Evaluación Preclínica de Medicamentos/instrumentación , Evaluación Preclínica de Medicamentos/métodos , Células HEK293 , Humanos , Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/patología , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
16.
J Allergy Clin Immunol ; 142(4): 1185-1193.e4, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29890236

RESUMEN

BACKGROUND: Asthma is a chronic inflammatory airway disease in which innate and adaptive immune cells act together to cause eosinophilic inflammation, goblet cell metaplasia (GCM), and bronchial hyperreactivity (BHR). In clinical trials using biologicals against IL-4 receptor (IL-4R) α or IL-5, only a subset of patients with moderate-to-severe asthma responded favorably, suggesting that distinct pathophysiologic mechanisms are at play in subgroups of patients called endotypes. However, the effect of multiple cytokine blockade using bispecific antibodies has not been tested. OBJECTIVE: We sought to target simultaneously the IL-4, IL-13, and IL-5 signaling pathways with a novel IL-4Rα/IL-5-bispecific antibody in a murine house dust mite (HDM) model of asthma. METHODS: Two mAbs neutralizing IL-4Rα and IL-5 were generated by using a llama-based antibody platform. Their heavy and light chains were then cotransfected in mammalian cells, resulting in a heterogeneous antibody mixture from which the bispecific antibody was isolated by using a dual anti-idiotypic purification process. C57BL/6J mice were finally sensitized and challenged to HDM extracts and treated during challenge with the antibodies. RESULTS: We successfully generated and characterized the monospecific and bispecific antibodies targeting IL-4Rα and IL-5. The monospecific antibodies could suppress eosinophilia, IgE synthesis, or both, whereas only the IL-4Rα/IL-5-bispecific antibody and the combination of monospecific antibodies additionally inhibited GCM and BHR. CONCLUSION: Type 2 cytokines act synergistically to cause GCM and BHR in HDM-exposed mice. These preclinical results show the feasibility of generating bispecific antibodies that target multiple cytokine signaling pathways as superior inhibitors of asthma features, including the difficult-to-treat GCM.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico , Asma/tratamiento farmacológico , Citocinas/antagonistas & inhibidores , Eosinofilia/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/uso terapéutico , Asma/inmunología , Asma/patología , Asma/fisiopatología , Camélidos del Nuevo Mundo , Línea Celular , Citocinas/inmunología , Eosinofilia/inmunología , Eosinofilia/patología , Eosinofilia/fisiopatología , Escherichia coli , Femenino , Células Caliciformes/efectos de los fármacos , Células Caliciformes/patología , Humanos , Ratones Endogámicos C57BL , Pyroglyphidae/inmunología
17.
Expert Rev Anticancer Ther ; 16(10): 1019-27, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27530056

RESUMEN

INTRODUCTION: Peritoneal carcinomatosis (PC) is a cancer disease with an urgent need for effective treatment. Conventional chemotherapy failed to show acceptable results. Cytoreductive surgery and hyperthermic chemoperfusion (HIPEC) are only beneficial in few patients with resectable peritoneal metastasis. Immunotherapy could be attractive against PC, as all requirements for immunotherapy are available in the peritoneal cavity. AREAS COVERED: This review analyzes the present literature for immunotherapy of PC. Advances from immune stimulators, radionucleotide-conjugated- and bispecific antibodies to future developments like adoptive engineered T-cells with chimeric receptors are discussed. The clinical development of catumaxomab, which was the first intraperitoneal immunotherapy to be approved for clinical treatment, is discussed. The requirements for future developments are illustrated. Expert commentary: Immunotherapy of peritoneal carcinomatosis is manageable, showing striking cancer cell killing. Improved profiles of adverse events by therapy-induced cytokine release, enhanced specific killing and optimal treatment schedules within multimodal treatment will be key factors.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Inmunoterapia/métodos , Neoplasias Peritoneales/terapia , Animales , Anticuerpos Biespecíficos/administración & dosificación , Anticuerpos Biespecíficos/efectos adversos , Terapia Combinada , Citocinas/inmunología , Procedimientos Quirúrgicos de Citorreducción/métodos , Humanos , Hipertermia Inducida/métodos , Inmunoterapia/efectos adversos , Neoplasias Peritoneales/inmunología
18.
Haemophilia ; 22 Suppl 5: 36-41, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27405674

RESUMEN

The development of inhibitors to factor VIII (FVIII) or factor IX (FIX) remains a major treatment complication encountered in the treatment of haemophilia. Not all patients with even the same severity and genotype develop inhibitors suggesting an underlying mechanism of tolerance against FVIII- or FIX-related immunity. One mechanism may be central tolerance observed in patients in whom the FVIII mutation enables some production of the protein. The other is a peripheral tolerance mechanism which may be evident in patients with null mutation. Recently, recombinant porcine FVIII (rpFVIII, Obixur, OBI-1, BAX801) has been developed for the haemostatic treatment of both congenital haemophilia with inhibitor (CHAWI) and acquired haemophilia A (AHA). In 28 subjects with AHA with life-/limb-threatening bleeding, rpFVIII reduced or stopped bleeding in all patients within 24 h. The cross-reactivity of anti-human FVIII antibodies to rpFVIII remains around 30-50%. Recently, new therapeutics based on the quite novel concepts have been developed and clinical studies are ongoing. These are humanized asymmetric antibody mimicking FVIIIa function by maintaining a suitable interaction between FIXa and FX (Emicizumab, ACE910), and small interfering RNAs (siRNA, ALN-AT3) suppress liver production of AT through post-transcriptional gene silencing and a humanized anti-TFPI monoclonal antibody (Concizumab). Their main advantages are longer half-life, subcutaneous applicability and efficacy irrespective of the presence of inhibitors which will make it easier to initiate more effective treatment especially early childhood.


Asunto(s)
Factor VIII/inmunología , Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales Humanizados/biosíntesis , Anticuerpos Monoclonales Humanizados/genética , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Neutralizantes/sangre , Factor VIII/uso terapéutico , Factor X/inmunología , Factor X/metabolismo , Factor Xa/inmunología , Factor Xa/metabolismo , Hemofilia A/tratamiento farmacológico , Hemofilia A/inmunología , Humanos , Tolerancia Inmunológica , Interferencia de ARN
19.
Rinsho Ketsueki ; 57(6): 709-14, 2016 06.
Artículo en Japonés | MEDLINE | ID: mdl-27384849

RESUMEN

Serious issues in current hemostatic treatment of hemophilia A are the requirement for frequent intravenous administrations of factor (F) VIII, FVIII inhibitor development, and hemostatic treatment for patients with this inhibitor. For the purpose of overcoming these challenges, the FVIIIa-substituting bispecific antibody against FIXa/FX (ACE910, INN emicizumab) was produced. Emicizumab demonstrated marked hemostatic effects on both ongoing and spontaneous joint bleeding in the acquired hemophilia A primate model. The clinical phase 1 study designed to assess the pharmacokinetics, pharmacodynamics and safety of emicizumab has been initiated. Severe emicizumab-related adverse events were minimal. The t1/2 was approximately 30 days, and bleeding events were significantly decreased by weekly subcutaneous administration in severe hemophilia A patients, independently of the presence of the inhibitor. Currently, the phase 1/2 extension study is ongoing. We anticipate that emicizumab will show the benefits of prophylactic efficacy with subcutaneous administration at a much lower frequency.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Materiales Biomiméticos/uso terapéutico , Factor VIII/inmunología , Hemofilia A/tratamiento farmacológico , Animales , Ensayos Clínicos como Asunto , Terapias Complementarias , Diseño de Fármacos , Hemofilia A/inmunología , Humanos
20.
Leuk Lymphoma ; 57(5): 1021-32, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27050240

RESUMEN

Blinatumomab is a member of a novel class of T cell-engaging bispecific antibodies, so-called Bispecific T cell Engager (BiTEs). It is directed against the B cell differentiation antigen CD19 and intended for treatment of B cell malignancies. In clinical phase I/II trials, blinatumomab showed remarkable single-agent activity in patients with relapsed and/or refractory (R/R) non-Hodgkin lymphoma and R/R B cell precursor acute lymphoblastic leukemia (B-precursor ALL). Cytokine release syndrome and neurological side effects were dose-limiting. Adverse effects were well manageable and transient in nature. Based on results of an international phase II trial, blinatumomab received FDA approval for the treatment of R/R B-precursor ALL in December 2014. Ongoing and future trials will contribute to further optimization of blinatumomab-based T cell therapy and have to show that integration of blinatumomab in current and innovative treatment protocols improves overall survival and quality of life of patients with B cell malignancies.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Animales , Anticuerpos Biespecíficos/economía , Anticuerpos Biespecíficos/farmacología , Antígenos CD19/metabolismo , Antineoplásicos/economía , Antineoplásicos/farmacología , Complejo CD3/metabolismo , Ensayos Clínicos como Asunto , Análisis Costo-Beneficio , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos , Humanos , Linfoma no Hodgkin/tratamiento farmacológico , Linfoma no Hodgkin/inmunología , Linfoma no Hodgkin/metabolismo , Linfoma no Hodgkin/patología , Terapia Molecular Dirigida , Neoplasias/inmunología , Neoplasias/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Recurrencia , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA