Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Pharmacol Res ; 196: 106912, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37696483

RESUMEN

Microglia are first responders to acute brain insults and initiate neuroinflammation to drive secondary tissue injury. Yet the key molecular switches in control of the inflammatory activity of microglia remain poorly understood. Intracerebral hemorrhage (ICH) is a devastating stroke subtype whereby a hematoma is formed within the brain parenchyma and associated with high mortality. Using a mouse model of ICH, we found upregulation of CD22 that predominantly occurred in microglia. Antibody blockade of CD22 led to a reduction in neurological deficits, brain lesion and hematoma volume. This was accompanied by reduced inflammatory activity, increased expression of alternative activation markers (CD206 and IL-10) and enhanced phagocytosis activity in microglia after ICH. CD22 blockade also led to an increase of phosphorylated SYK and AKT after ICH. Notably, the benefits of CD22 blockade were ablated in ICH mice subjected to microglial depletion with a colony-stimulating factor 1 receptor inhibitor PLX5622. Additionally, the protective effects of CD22 blockade was diminished in ICH mice receiving a SYK inhibitor R406. Together, our findings highlight CD22 as a key molecular switch to control the detrimental effects of microglia after acute brain injury, and provide a novel strategy to improve the outcome of ICH injury.


Asunto(s)
Lesiones Encefálicas , Microglía , Lectina 2 Similar a Ig de Unión al Ácido Siálico , Encéfalo/metabolismo , Hemorragia Cerebral/tratamiento farmacológico , Hemorragia Cerebral/metabolismo , Hematoma/complicaciones , Hematoma/metabolismo , Hematoma/patología , Enfermedades Neuroinflamatorias , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo , Animales , Ratones
2.
Front Immunol ; 12: 684430, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34140954

RESUMEN

Microglia, the innate immune cells of the brain, are essential for maintaining homeostasis by their ramified, highly motile processes and for orchestrating the immune response to pathological stimuli. They are implicated in several neurodegenerative diseases like Alzheimer's and Parkinson's disease. One commonality of these diseases is their strong correlation with aging as the highest risk factor and studying age-related alterations in microglia physiology and associated signaling mechanism is indispensable for a better understanding of age-related pathomechanisms. CD22 has been identified as a modifier of microglia phagocytosis in a recent study, but not much is known about the function of CD22 in microglia. Here we show that CD22 surface levels are upregulated in aged versus adult microglia. Furthermore, in the amyloid mouse model PS2APP, Aß-containing microglia also exhibit increased CD22 signal. To assess the impact of CD22 blockage on microglia morphology and dynamics, we have established a protocol to image microglia process motility in acutely prepared brain slices from CX3CR1-GFP reporter mice. We observed a significant reduction of microglial ramification and surveillance capacity in brain slices from aged versus adult mice. The age-related decrease in surveillance can be restored by antibody-mediated CD22 blockage in aged mice, whereas surveillance in adult mice is not affected by CD22 inhibition. Moreover to complement the results obtained in mice, we show that human iPSC-derived macrophages exhibit an increased phagocytic capacity upon CD22 blockage. Downstream analysis of antibody-mediated CD22 inhibition revealed an influence on BMP and TGFß associated gene networks. Our results demonstrate CD22 as a broad age-associated modulator of microglia functionality with potential implications for neurodegenerative disorders.


Asunto(s)
Envejecimiento/fisiología , Encéfalo/citología , Microglía/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores , Envejecimiento/efectos de los fármacos , Envejecimiento/genética , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Recuento de Células , Modelos Animales de Enfermedad , Humanos , Macrófagos/metabolismo , Masculino , Ratones , Microglía/citología , Fagocitosis/genética , Lectina 2 Similar a Ig de Unión al Ácido Siálico/genética , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo , Transducción de Señal
4.
Cancer Biother Radiopharm ; 36(8): 672-681, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33887152

RESUMEN

Background: BAY 1862864 is an α-particle emitting 227Th-labeled CD22-targeting antibody. This first-in-human dose-escalation phase I study evaluated BAY 1862864 in patients with CD22-positive relapsed/refractory B cell non-Hodgkin lymphoma (R/R-NHL). Materials and Methods: BAY 1862864 intravenous injections were administered at the starting 227Th radioactivity dose of 1.5 MBq (2 or 10 mg antibody), and the radioactivity dose escalated in ∼1.5 MBq increments (10 mg antibody) until the maximum tolerated dose (MTD) was reported. The primary objective was to determine the safety, tolerability, and MTD. Results: Twenty-one patients received BAY 1862864. Two dose-limiting toxicities (grade 3 febrile neutropenia and grade 4 thrombocytopenia) were reported in one patient in the 4.6 MBq (10 mg antibody) cohort. The MTD was not reached. Ten (48%) patients reported grade ≥3 treatment-emergent adverse events, with the most common being neutropenia, thrombocytopenia, and leukopenia, each occurring in 3 (14%) patients. Pharmacokinetics demonstrated the dose proportionality and stability of BAY 1862864 in the blood. The objective response rate (ORR) was 25% (5/21 patients) according to the LUGANO 2014 criteria, including 1 complete and 4 partial responses. The ORR was 11% (1/9) and 30% (3/10) in patients with relapsed high- and low-grade lymphomas, respectively. Conclusions: BAY 1862864 was safe and tolerated in patients with R/R-NHL. Clinical Trial Registration numbers: NCT02581878 and EudraCT 2014-004140-36.


Asunto(s)
Leucopenia , Linfoma no Hodgkin , Neutropenia , Lectina 2 Similar a Ig de Unión al Ácido Siálico , Torio/farmacología , Trombocitopenia , Anciano , Anticuerpos Monoclonales Humanizados/farmacología , Relación Dosis-Respuesta a Droga , Monitoreo de Drogas/métodos , Femenino , Humanos , Inyecciones Intravenosas , Leucopenia/inducido químicamente , Leucopenia/diagnóstico , Linfoma no Hodgkin/patología , Linfoma no Hodgkin/radioterapia , Masculino , Dosis Máxima Tolerada , Clasificación del Tumor , Estadificación de Neoplasias , Neutropenia/inducido químicamente , Neutropenia/diagnóstico , Radioterapia/métodos , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores , Lectina 2 Similar a Ig de Unión al Ácido Siálico/inmunología , Trombocitopenia/inducido químicamente , Trombocitopenia/diagnóstico , Resultado del Tratamiento
6.
Minerva Med ; 111(5): 478-490, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32955830

RESUMEN

Patients with acute lymphoblastic leukemia (ALL) are characterized by an unfavorable outcome in the majority of adult cases. Several clinical trials have confirmed the usefulness of a pediatric-type therapy applied to adult patients. Adults present with higher risk features at diagnosis that predispose them to chemotherapy resistance and disease relapse after an initial achievement of complete remission. The recent introduction of novel immune-targeted therapies, including monoclonal antibodies (MoAbs) targeting B cell-associated antigens such as CD19 (blinatumumab) and CD22 (inotuzumab), tyrosine kinase inhibitors targeting BCR-ABL1 tyrosine kinase, bispecific antibodies and chimeric antigen receptor T- cell therapy (CAR-T), circumvent B-ALL cell chemo-refractoriness through novel mechanisms of action, potentially eradicating minimal residual disease (MRD) and enabling more patients to receive allogeneic hematopoietic stem cell transplantation and to achieve a better clinical outcome.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Inhibidores de Proteínas Quinasas/uso terapéutico , Enfermedad Aguda , Adulto , Anticuerpos Biespecíficos/uso terapéutico , Ensayos Clínicos como Asunto , Resistencia a Antineoplásicos , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Trasplante de Células Madre Hematopoyéticas , Humanos , Imidazoles/uso terapéutico , Inmunoterapia Adoptiva/métodos , Inotuzumab Ozogamicina/uso terapéutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Piridazinas/uso terapéutico , Recurrencia , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores
10.
Drug Metab Dispos ; 47(10): 1146-1155, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31358513

RESUMEN

Antibody-drug conjugates (ADCs) contain a disease-receptor antibody and a payload drug connected via a linker. The payload delivery depends on both tumor properties and ADC characteristics. In this study, we used different linkers, attachment sites, and doses to modulate payload delivery of several ADCs bearing maytansinoids (e.g., DM1), auristatins (e.g., MMAE), and DNA alkylating agents [e.g., pyrrolo[2,1-c][1,4]benzodiazepine-dimer (PBD)] as payloads in HER2- or CD22-expressing xenograft models. The tumor growth inhibition and ADC stability and exposure data were collected and analyzed from these dosed animals. The trend analysis suggests that intratumoral payload exposures that directly related the combination of conjugate linker and dose correlate with the corresponding efficacies of three payload types in two antigen-expressing xenograft models. These preliminary correlations also suggest that a minimal threshold concentration of intratumoral payload is required to support sustained efficacy. In addition, an ADC can deliver an excessive level of payload to tumors that does not enhance efficacy ("Plateau" effect). In contrast to tumor payload concentrations, the assessments of systemic exposures of total antibody (Tab) as well as the linker, dose, site of attachment, plasma stability, and drug-to-antibody ratio changes of these ADCs did not consistently rationalize the observed ADC efficacies. The requirement of a threshold payload concentration for efficacy is further supported by dose fractionation studies with DM1-, MMAE-, and PBD-containing ADCs, which demonstrated that single-dose regimens showed better efficacies than fractionated dosing. Overall, this study demonstrates that 1) the linker and dose together determine the tissue payload concentration that correlates with the antitumor efficacy of ADCs and 2) an ADC can deliver an unnecessary level of payload to tumors in xenograft models.


Asunto(s)
Antineoplásicos Inmunológicos/farmacocinética , Inmunoconjugados/farmacocinética , Receptor ErbB-2/antagonistas & inhibidores , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores , Ado-Trastuzumab Emtansina/administración & dosificación , Ado-Trastuzumab Emtansina/farmacocinética , Animales , Antineoplásicos Inmunológicos/administración & dosificación , Antineoplásicos Inmunológicos/química , Benzodiazepinas/química , Brentuximab Vedotina/administración & dosificación , Brentuximab Vedotina/farmacocinética , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Inmunoconjugados/administración & dosificación , Ratones , Ratones Transgénicos , Pirroles/química , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Drug Metab Dispos ; 47(8): 884-889, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31072822

RESUMEN

Carfilzomib (CFZ) is a proteasome inhibitor used for oncology indications including treating multiple myeloma. CFZ is a potent cytotoxic agent with an IC50 value in the nanomolar range in various cancer cell lines and was considered as a potential payload for antibody drug conjugates (ADCs); however, the conjugated CFZ to anti-CD22 or anti-HER2 antibody totally abolishes the in vitro potency. This was a surprise since with other payloads such as monomethyl auristatin E (MMAE), where potent antiproliferation efficacy was retained as MMAE alone or as a payload in an ADC. Further investigations were conducted using CFZ alone, CFZ with a linker, and CFZ-ADC with tissue matrices including lysosomal enzymes. With CFZ linked to the ADC, cathepsin B (a lysosomal enzyme) was efficient in liberating CFZ from the ADC by cleavage of the valine-citrulline linker. At the same time, the liberated CFZ in the lysosome was inactivated due to further metabolism by lysosomal enzymes. The products from epoxide and amide hydrolysis were identified from these incubations. These results suggested that the CFZ-ADC upon uptake and internalization specifically delivers CFZ payload to the lysosomes, where CFZ was inactivated. On the other hand, CFZ by itself is not as vulnerable and could reach its target. Therefore, lysosomal stability is an important criterion in the selection of a payload for making the next generation of potent ADC therapeutics.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Antineoplásicos/farmacocinética , Inmunoconjugados/farmacocinética , Lisosomas/enzimología , Oligopéptidos/farmacocinética , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/química , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Catepsina B/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Estudios de Factibilidad , Humanos , Inmunoconjugados/administración & dosificación , Inmunoconjugados/química , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Oligopéptidos/administración & dosificación , Oligopéptidos/química , Receptor ErbB-2/antagonistas & inhibidores , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores
12.
Leukemia ; 33(12): 2854-2866, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31110217

RESUMEN

Despite worldwide promising clinical outcome of CD19 CAR-T therapy, relapse after this therapy is associated with poor prognosis and has become an urgent problem to be solved. We conducted a CD22 CAR T-cell therapy in 34 relapsed or refractory (r/r) B-ALL pediatric and adult patients who failed from previous CD19 CAR T-cell therapy. Complete remission (CR) or CR with incomplete count recovery (CRi) was achieved in 24 of 30 patients (80%) that could be evaluated on day 30 after infusion, which accounted for 70.5% of all 34 enrolled patients. Most patients only experienced mild cytokine-release syndrome and neurotoxicity. Seven CR patients received no further treatment, and 3 of them remained in remission at 6, 6.6, and 14 months after infusion. Eleven CR patients were promptly bridged to transplantation, and 8 of them remained in remission at 4.6 to 13.3 months after transplantation, resulted in 1-year leukemia-free survival rate of 71.6% (95% CI, 44.2-99.0). CD22 antigen loss or mutation was not observed to be associated with relapsed patients. Our study demonstrated that our CD22 CAR T-cells was highly effective in inducing remission in r/r B-ALL patients, and also provided a precious window for subsequent transplantation to achieve durable remission.


Asunto(s)
Inmunoterapia Adoptiva , Leucemia-Linfoma Linfoblástico de Células Precursoras B/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Quiméricos de Antígenos/metabolismo , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores , Linfocitos T/inmunología , Linfocitos T/metabolismo , Adolescente , Adulto , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Biopsia , Niño , Preescolar , Terapia Combinada/efectos adversos , Terapia Combinada/métodos , Citocinas/metabolismo , Citotoxicidad Inmunológica , Manejo de la Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Lactante , Masculino , Persona de Mediana Edad , Leucemia-Linfoma Linfoblástico de Células Precursoras B/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/etiología , Receptores de Antígenos de Linfocitos T/genética , Receptores Quiméricos de Antígenos/genética , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo , Adulto Joven
13.
Nature ; 568(7751): 187-192, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30944478

RESUMEN

Microglia maintain homeostasis in the central nervous system through phagocytic clearance of protein aggregates and cellular debris. This function deteriorates during ageing and neurodegenerative disease, concomitant with cognitive decline. However, the mechanisms of impaired microglial homeostatic function and the cognitive effects of restoring this function remain unknown. We combined CRISPR-Cas9 knockout screens with RNA sequencing analysis to discover age-related genetic modifiers of microglial phagocytosis. These screens identified CD22, a canonical B cell receptor, as a negative regulator of phagocytosis that is upregulated on aged microglia. CD22 mediates the anti-phagocytic effect of α2,6-linked sialic acid, and inhibition of CD22 promotes the clearance of myelin debris, amyloid-ß oligomers and α-synuclein fibrils in vivo. Long-term central nervous system delivery of an antibody that blocks CD22 function reprograms microglia towards a homeostatic transcriptional state and improves cognitive function in aged mice. These findings elucidate a mechanism of age-related microglial impairment and a strategy to restore homeostasis in the ageing brain.


Asunto(s)
Envejecimiento/fisiología , Encéfalo/citología , Homeostasis/efectos de los fármacos , Microglía/efectos de los fármacos , Ácido N-Acetilneuramínico/farmacología , Fagocitosis/efectos de los fármacos , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores , Envejecimiento/efectos de los fármacos , Envejecimiento/genética , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Proteína 9 Asociada a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Cognición/efectos de los fármacos , Cognición/fisiología , Femenino , Homeostasis/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Ácido N-Acetilneuramínico/química , Fagocitosis/genética , Análisis de Secuencia de ARN , Lectina 2 Similar a Ig de Unión al Ácido Siálico/genética , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo
14.
Blood Adv ; 3(1): 96-104, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30622147

RESUMEN

Inotuzumab ozogamicin (InO) is a recently US Food and Drug Administration-approved antibody-drug conjugate for the treatment of relapsed/refractory B-cell acute lymphoblastic leukemia (ALL). InO consists of a CD22-targeting immunoglobulin G4 humanized monoclonal antibody conjugated to calicheamicin. Although initially developed for the treatment of non-Hodgkin lymphoma (NHL) because of activity in preclinical models and high response rates in indolent lymphomas, a phase 3 trial was negative and further development focused on CD22+ ALL. Although results in NHL were disappointing, parallel testing in early-phase trials of CD22+ ALL demonstrated feasibility and efficacy. Subsequently, the randomized phase 3 Study Of Inotuzumab Ozogamicin Versus Investigator's Choice Of Chemotherapy In Patients With Relapsed Or Refractory Acute Lymphoblastic Leukemia trial showed that InO was superior to standard of care regimens with a significantly improved complete remission (CR) rate in patients with relapsed/refractory disease (80.7% vs 29.4%, P < .001). Patients achieving CR with InO also had a significantly higher rate of undetectable minimal residual disease compared with chemotherapy (78.4% vs 28.1%, P < .001). InO-specific side effects, including veno-occlusive disease, have been an ongoing area of concern, and consensus guidelines for minimizing toxicities are now available. Ongoing trials are investigating the combination of InO with other agents in the relapse setting and the addition of InO to frontline therapy. This review details the preclinical and clinical development of InO, focusing on how best to use it and future directions for further development.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Inotuzumab Ozogamicina/uso terapéutico , Terapia Molecular Dirigida , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Animales , Antineoplásicos Inmunológicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Estudios Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos , Humanos , Inotuzumab Ozogamicina/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/mortalidad , Pronóstico , Recurrencia , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores , Resultado del Tratamiento
15.
BMC Cancer ; 18(1): 1117, 2018 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-30442119

RESUMEN

BACKGROUND: Extramedullary involvement of B-cell Acute Lymphoblastic Leukemia (EM-ALL) is a rare occurrence, characterized by dismal outcome and the absence of a defined and shared therapeutic approach. In the landscape of innovative compounds, inotuzumab ozogamicin (IO) is a promising drug, whose mechanism of action relies on the killing of CD22 positive leukemic cells, through the delivery, after cell binding, of a molecule of calicheamicin. CASE PRESENTATION: We report two cases of CD22 positive relapsed EM-ALL treated with IO, obtained as compassionate use. Case 1, a 66 years old woman, affected by Philadelphia (Ph) negative B-ALL, relapsed with extramedullary involvement after 6 standard chemotherapy courses, who reached a complete metabolic response with IO treatment. Case 2, a 67 years old man with Ph positive B-ALL, initially treated with ponatinib, a third generation tyrosine-kinase inhibitor (TKI), obtaining a prolonged deep molecular remission. Nevertheless, for skin relapse during TKI treatment, the patient received local radiotherapy and, shortly after, standard chemotherapy, as multiple abdominal sites of relapse were detected too, with no response. The patient then received IO, obtained as compassionate use, with a good metabolic response. CONCLUSIONS: These two cases suggest a possible key role of IO in the setting of advanced CD22 positive ALL, and underline its potential activity also in patients with EM involvement, relapsed after or refractory to conventional chemotherapy. Despite the well known hepatotoxic effect of the compound (Sinusoid Occlusive Syndrome), neither of them had such adverse event, moreover the second patient safely underwent allogeneic bone marrow transplantation.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/uso terapéutico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores , Anciano , Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Femenino , Humanos , Imidazoles/farmacología , Imidazoles/uso terapéutico , Inotuzumab Ozogamicina , Masculino , Recurrencia Local de Neoplasia/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Piridazinas/farmacología , Piridazinas/uso terapéutico , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo , Resultado del Tratamiento
16.
Drugs ; 78(16): 1763-1767, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30357593

RESUMEN

Moxetumomab pasudotox-tdfk (LUMOXITI™), an anti CD22 recombinant immunotoxin, has been developed by MedImmune and its parent company AstraZeneca for the treatment of hairy cell leukaemia. The product, discovered at the National Cancer Institute, is an optimised version of immunotoxin CAT-3888. Moxetumomab pasudotox is composed of the Fv fragment of an anti-CD22 monoclonal antibody fused to a 38 kDa fragment of Pseudomonas exotoxin A, PE38. The Fv portion of moxetumomab pasudotox binds to CD22, a cell surface receptor expressed on a variety of malignant B-cells, thereby delivering the toxin moiety PE38 directly to tumour cells. Once internalised, PE38 catalyses the ADP ribosylation of the diphthamide residue in elongation factor-2 (EF-2), resulting in the rapid fall in levels of the anti-apoptotic protein myeloid cell leukaemia 1 (Mcl-1), leading to apoptotic cell death. This article summarizes the milestones in the development of moxetumomab pasudotox leading to this first approval for the treatment of adults with relapsed or refractory hairy cell leukaemia who received at least two prior systemic therapies, including treatment with a purine nucleoside analogue. Development of moxetumomab pasudotox for non-Hodgkin's lymphoma, chronic lymphocytic leukaemia and precursor cell lymphoblastic leukaemia/lymphoma was discontinued.


Asunto(s)
Antineoplásicos , Toxinas Bacterianas , Exotoxinas , Leucemia Linfoide/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Antineoplásicos Inmunológicos , Toxinas Bacterianas/administración & dosificación , Toxinas Bacterianas/efectos adversos , Toxinas Bacterianas/uso terapéutico , Exotoxinas/administración & dosificación , Exotoxinas/efectos adversos , Exotoxinas/uso terapéutico , Humanos , Región Variable de Inmunoglobulina , Inmunotoxinas/administración & dosificación , Inmunotoxinas/efectos adversos , Inmunotoxinas/química , Inmunotoxinas/uso terapéutico , Persona de Mediana Edad , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores
17.
JAMA Oncol ; 4(10): 1413-1420, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29931220

RESUMEN

Importance: Remarkable progress has occurred in understanding the pathophysiology and in developing improved personalized therapies in adult acute lymphoblastic leukemia (ALL). Observations: We searched MEDLINE (1990-2018), the American Society of Clinical Oncology, and American Society of Hematology websites (2010-2018). We used the search terms "acute lymphoblastic or lymphocytic leukemia" or "ALL." We largely selected publications in the past 5 years but did not exclude commonly referenced and highly regarded older publications. Target therapies toward specific transcripts (eg, BCR-ABL1 tyrosine kinase oncoprotein by tyrosine kinase inhibitors) and specific leukemic cell surface antigens (eg, CD20, CD22, and CD19 monoclonal antibodies) are major breakthroughs. Current treatments produce long-term survival in 50% of patients with precursor B-cell ALL including 50% to 70% with Philadelphia chromosome (Ph)-positive ALL, 50% to 60% with T-cell ALL, and 80% with mature B-cell ALL. Next-generation sequencing and genomic profiling in ALL have identified new prognostic markers, targets, and ALL subtypes (eg, Ph-like ALL). Monoclonal antibodies, bispecific antibody constructs, and chimeric antigen receptor T cellular therapies developed in the past 5 to 7 years have revolutionized the treatment of ALL and resulted in US Food and Drug Administration approvals of blinatumomab in 2014, as well as inotuzumab and tisagenlecleucel in 2017 as ALL salvage strategies. Their use in combined modalities as salvage and frontline therapies is currently under investigation. Conclusions and Relevance: Therapies targeting specific transcripts or leukemic cell surface antigens are major breakthroughs in the treatment of adults with ALL. The incorporation of new monoclonal antibodies and other targeted approaches into frontline regimens is showing promising results. If confirmed, such strategies may increase the cure rates in adults to levels achieved in pediatric ALL and reduce the need for intensive and prolonged chemotherapy.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Terapia Molecular Dirigida/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores , Adulto , Anticuerpos Biespecíficos/uso terapéutico , Antígenos CD19/inmunología , Antígenos CD19/metabolismo , Antígenos CD20/inmunología , Antígenos CD20/metabolismo , Antineoplásicos/uso terapéutico , Proteínas de Fusión bcr-abl/metabolismo , Humanos , Inmunoterapia/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Terapia Recuperativa/métodos , Lectina 2 Similar a Ig de Unión al Ácido Siálico/inmunología , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo , Análisis de Supervivencia
18.
Mol Cancer Ther ; 17(7): 1486-1493, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29695631

RESUMEN

Tac (CD25) is expressed on multiple hematologic malignancies and is a target for cancer therapies. LMB-2 is an extremely active anti-Tac recombinant immunotoxin composed of an Fv that binds to Tac and a 38-kDa fragment of Pseudomonas exotoxin A (PE38). Although LMB-2 has shown high cytotoxicity toward Tac-expressing cancer cells in clinical trials, its efficacy was hampered by the formation of anti-drug antibodies against the immunogenic bacterial toxin and by dose-limiting off-target toxicity. To reduce toxin immunogenicity and nonspecific toxicity, we introduced six point mutations into domain III that were previously shown to reduce T-cell immunogenicity and deleted domain II from the toxin, leaving only the 11aa furin cleavage site, which is required for cytotoxic activity. Although this strategy has been successfully implemented for mesothelin and CD22-targeting immunotoxins, we found that removal of domain II significantly lowered the cytotoxic activity of anti-Tac immunotoxins. To restore cytotoxic activity in the absence of PE domain II, we implemented a combined rational design and screening approach to isolate highly active domain II-deleted toxin variants. The domain II-deleted variant with the highest activity contained an engineered disulfide-bridged furin cleavage site designed to mimic its native conformation within domain II. We found that this approach restored 5-fold of the cytotoxic activity and dramatically improved the MTD. Both of these improvements led to significantly increased antitumor efficacy in vivo We conclude that the next-generation anti-Tac immunotoxin is an improved candidate for targeting Tac-expressing malignancies. Mol Cancer Ther; 17(7); 1486-93. ©2018 AACR.


Asunto(s)
ADP Ribosa Transferasas/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Toxinas Bacterianas/administración & dosificación , Exotoxinas/administración & dosificación , Neoplasias Hematológicas/tratamiento farmacológico , Ingeniería de Proteínas , Factores de Virulencia/administración & dosificación , ADP Ribosa Transferasas/genética , ADP Ribosa Transferasas/inmunología , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Toxinas Bacterianas/genética , Toxinas Bacterianas/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citotoxicidad Inmunológica/genética , Exotoxinas/genética , Exotoxinas/inmunología , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/inmunología , Humanos , Subunidad alfa del Receptor de Interleucina-2/antagonistas & inhibidores , Subunidad alfa del Receptor de Interleucina-2/inmunología , Mesotelina , Ratones , Mutación Puntual/genética , Dominios Proteicos/genética , Dominios Proteicos/inmunología , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores , Lectina 2 Similar a Ig de Unión al Ácido Siálico/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Factores de Virulencia/genética , Factores de Virulencia/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto , Exotoxina A de Pseudomonas aeruginosa
19.
Expert Rev Clin Immunol ; 14(4): 245-258, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29542345

RESUMEN

INTRODUCTION: Systemic lupus erythematosus (SLE) is a multi-system autoimmune disease. There are three drugs licensed for the treatment of lupus: corticosteroids, hydroxychloroquine and belimumab. Immunosuppressants such as azathioprine, methotrexate and mycophenolate are also used. Despite these treatments there is still considerable morbidity. New treatments are needed for the management of active lupus. Epratuzumab a humanized IgG1 monoclonal antibody that targets CD22 resulting in selective B cell modulation that has been considered a potential treatment for SLE. Areas covered: Summary of the relevant pathogenesis and disease activity measurements used in SLE patients, current treatments and unmet needs in SLE, pharmacokinetics and pharmacodynamics of epratuzumab therapy, and a summary of the 7 clinical trials that have investigated the efficacy and safety of epratuzumab in SLE. Expert commentary: It is not clear why trials have failed to demonstrate efficacy but high placebo response rates from optimisation of standard of care and a sub-optimal dosing regimen may have played a role. Post-hoc analysis suggested that there may be subgroups that did respond, such as anti-SSA positive patients with features of Sjogren's syndrome. Further research is needed to explore this and other potential sub-groups that might respond.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Linfocitos B/inmunología , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/inmunología , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores , Anticuerpos Monoclonales Humanizados/farmacocinética , Linfocitos B/patología , Humanos , Lupus Eritematoso Sistémico/patología , Ensayos Clínicos Controlados Aleatorios como Asunto , Lectina 2 Similar a Ig de Unión al Ácido Siálico/inmunología
20.
Cancer ; 124(10): 2151-2160, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29508899

RESUMEN

BACKGROUND: Inotuzumab ozogamicin (InO), an anti-CD22 antibody-calicheamicin conjugate, demonstrated superior clinical activity versus standard-of-care (SOC) chemotherapies for relapsed/refractory B-cell acute lymphoblastic leukemia in the phase 3 randomized controlled INO-VATE trial. The authors assessed patient-reported outcomes (PROs) from that study. METHODS: Patients were randomized to receive either InO (1.8 mg/m2 per cycle for ≤6 cycles) or SOC (fludarabine/cytarabine [ara-C]/granulocyte colony-stimulating factor, or ara-C plus mitoxantrone, or high-dose ara-C for ≤4 cycles) and completed the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire and the EuroQoL 5 Dimensions Questionnaires at baseline, on day 1 of each cycle, and at the end of treatment. Treatment differences in PROs were assessed using longitudinal mixed-effects models with random intercepts and slopes. RESULTS: Questionnaire completion rates in the InO (n = 164) and SOC (n = 162) arms were 85% and 65%, respectively. Baseline scores were similar between arms. Patients who received InO reported better quality of life (QoL), functioning, and symptom scores (except for constipation and emotional functioning). Least-squares mean (95% confidence interval [CI]) differences in physical, role, and social functioning and in appetite loss were significant (6.9 [95% CI, 1.4-12.3], 11.4 [95% CI, 3.2-19.5], 8.4 [95% CI, 0.7-16.1], and -8.7 [95% CI, -16.0 to -1.4], respectively; all P < .05) and had exceeded the minimally important difference of 5. Mean treatment differences in favor of InO on the EuroQoL visual analog scale and the global health status/QoL, dyspnea, and fatigue scales reached or approached the minimally important difference of 5, although without statistical significance. No dimensions were significantly worse with InO versus SOC. CONCLUSIONS: The current PRO data support the favorable benefit/risk ratio of InO for the treatment of relapsed/refractory acute lymphoblastic leukemia, with superior clinical efficacy and better QoL. Cancer 2018;124:2151-60. © 2018 American Cancer Society.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Medición de Resultados Informados por el Paciente , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Resistencia a Antineoplásicos , Disnea/inducido químicamente , Disnea/epidemiología , Fatiga/inducido químicamente , Fatiga/epidemiología , Femenino , Estudios de Seguimiento , Estado de Salud , Humanos , Inotuzumab Ozogamicina , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/mortalidad , Leucemia-Linfoma Linfoblástico de Células Precursoras/microbiología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Calidad de Vida , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...