Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.982
Filter
1.
Biomater Adv ; 160: 213839, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38579521

ABSTRACT

Pulmonary delivery of protein therapeutics poses significant challenges that have not been well addressed in the research literature or practice. In fact, there is currently only one commercial protein therapeutic that is delivered through aerosolization and inhalation. In this study, we propose a drug delivery strategy that enables a high-concentration dosage for the pulmonary delivery of antibodies as an aerosolizable solid powder with desired stability. We utilized zwitterionic polymers for their promising properties as drug delivery vehicles and synthesized swellable, biodegradable poly(sulfo-betaine) (pSB) microparticles. The microparticles were loaded with Immunoglobulin G (IgG) as a model antibody. We quantified the microparticle size and morphology, and the particles were found to have an average diameter of 1.6 µm, falling within the optimal range (~1-5 µm) for pulmonary drug delivery. In addition, we quantified the impact of the crosslinker to monomer ratio on particle morphology and drug loading capacity. The results showed that there is a trade-off between desired morphology and drug loading capacity as the crosslinker density increases. In addition, the particles were aerosolized, and our data indicated that the particles remained intact and retained their initial morphology and size after aerosolization. The combination of morphology, particle size, antibody loading capacity, low cytotoxicity, and ease of aerosolization support the potential use of these particles for pulmonary delivery of protein therapeutics.


Subject(s)
Aerosols , Betaine , Betaine/analogs & derivatives , Particle Size , Betaine/chemistry , Humans , Administration, Inhalation , Immunoglobulin G/chemistry , Immunoglobulin G/administration & dosage , Drug Delivery Systems/methods , Polymers/chemistry , Drug Carriers/chemistry , Animals , Antibodies/chemistry , Microspheres
2.
s.l; CONETEC; 7 nov. 2023.
Non-conventional in Spanish | BRISA/RedTESA | ID: biblio-1519668

ABSTRACT

INTRODUCCIÓN: La hipercolesterolemia familiar (FH) es un trastorno hereditario que exhibe herencia autosómica dominante y que se expresa como una eliminación retardada de LDL plasmático, causada por mutaciones de los genes implicados en la vía mediada por el receptor de LDL. Se estima que la FH heterocigótica (HeFH) ocurre en aproximadamente 1 de cada 200 a 300 personas. Por el contrario, la FH homocigótica (HoFH) es una enfermedad que se encuentra en el listado de enfermedades poco frecuentes del Ministerio de Salud de la Nación (ORPHACODE 391665), con una prevalencia mundial estimada de aproximadamente 1:300.000 a 1:400.000. El diagnóstico de la HoFH debe ser mediante pruebas genéticas en personas con sospecha clínica: LDL-C no tratado >500 mg/dL (>13 mmol/L) o LDL-C tratado ≥300 mg/dL (>8 mmol/L), junto con xantoma cutáneo o tendinoso antes de los 10 años de edad, y/o niveles elevados de LDL-C compatibles con HeFH en ambos padres. Es importante señalar que se pueden observar niveles de LDL-C no tratados. Los personas con FH heterocigoto (HeFH) portan el gen mutado en un solo alelo y tienen niveles plasmáticos de LDL-C el doble de lo normal o más, y pueden experimentar el primer evento cardiovascular tan pronto como a los treinta años. Con mutaciones en ambos alelos, la HoFH exhibe niveles de LDL-C que duplican los de la HeFH, o incluso más, y los personas desarrollan complicaciones cardiovasculares incluso en la primera década de sus vidas.1,2 Los niños con HoFH deben someterse a una evaluación cardiovascular integral continuo con electrocardiogramas, ecocardiografías, pruebas funcionales de esfuerzo e imágenes de la arteria coronaria. El manejo de la enfermedad comprende la adopción de hábitos saludables para el corazón junto con tratamiento farmacológico. En este documento se plantea evaluar la eficacia y seguridad del uso de evinacumab (Evkeeza®) en niños con HoFH. TECNOLOGÍA: Evinacumab-dgnb es un anticuerpo monoclonal de isotopo IgG4 humano recombinante que se une e inhibe la proteína similar a la angiopoyetina 3 (ANGPTL3), una proteína reguladora que desempeña un papel en el metabolismo de los lípidos mediante la inhibición de la lipoproteína lipasa (LPL) y la lipasa endotelial (EL). 6 La inhibición de ANGPTL3 conduce a reducciones en LDLC, HDL-C y triglicéridos (TG). Evinacumab-dgnb reduce el LDL-C, independientemente de la presencia del receptor de LDL (con actividad del receptor de LDL prácticamente ausente o alterada), al promover el procesamiento y la eliminación de las lipoproteínas de muy baja densidad y la formación de LDL. OBJETIVO: El objetivo del presente informe es evaluar rápidamente los parámetros de eficacia, seguridad, costos y recomendaciones disponibles acerca del empleo del uso de evinacumab (Evkeeza®) en niños con HoFH. MÉTODOS: Se realizó una búsqueda bibliográfica en las principales bases de datos tales como PUBMED, LILACS, BRISA, COCHRANE, SCIELO, EMBASE, TRIPDATABASE como así también en sociedades científicas, agencias reguladoras, financiadores de salud y agencias de evaluación de tecnologías sanitarias. Se priorizó la inclusión de revisiones sistemáticas, ensayos clínicos controlados aleatorizados, evaluación de tecnología sanitaria y guías de práctica clínica de alta calidad metodológica. RECOMENDACIONES: No se hallaron guías de práctica clínica actualizadas en Argentina y en el Mundo que recomienden la tecnología en la indicación evaluada, antes o después de la autorización de comercialización por parte de la FDA.5,13,14 La Sociedad Europea de Aterosclerosis publicó en 2023 una actualización sobre su consenso en personas con HoFH.5 El panel hace una mención para niños/adolecentes y adultos por igual, donde menciona a la aféresis, el evinacumab o la lomitapida (no disponible en Argentina) como alternativas en personas con HoFH que no responden a la combinación de estatinas a dosis máximas tolerables, ezetimibe e inhibidores de PCSK9. También menciona que en muchos países de Europa el precio del fármaco será una limitante para su uso y serán alternativas viables comenzar con la aféresis. El Instituto Nacional para la Excelencia en Salud y Atención (NICE, sus siglas del inglés National Institute for Health and Care Excellence) de Reino Undo y la Agencia Canadiense de Medicamentos y Tecnologías en Salud (CADTH, su sigla del inglés Canadian Agency for Drugs and Technologies in Health) recientemente han comenzado a evaluar la tecnología en la indicación evaluada. CONCLUSIONES: La evidencia que sustenta la aprobación de comercialización por parte de los Estados Unidos de evinacumab-dgnb (Evkeeza®) en niños con hipercolesterolemia familiar homocigota se basa en un único ensayo clínico abierto y sin comparador de fase III. Cabe señalar que actualmente la Europa lo autorizó solo en mayores de 12 años. No se hallaron estudios que hayan evaluado el evinacumab-dgnb sobre desenlaces clínicos relevantes para la enfermedad o la reducción de la aféresis, como tampoco que lo compare frente a otros tratamientos farmacológicos disponibles para la población objetivo. El estudio identificado demostró que en 14 niños con hipercolesterolemia familiar homocigota resistentes al tratamiento farmacológico, la administración de evinacumab-dgnb podría mejorar al muy corto plazo parámetros de laboratorios importantes para el seguimiento de la enfermedad. No se hallaron guías de práctica clínica actualizadas en Argentina y en el Mundo que recomienden la tecnología en la indicación evaluada, antes o después de la autorización de comercialización por parte de los Estados Unidos. Una guía europea la menciona en niños pero por fuera de la actual autorización de comercialización del continente. Según los precios de adquisición relevados en ese país, para un niño de 25 kg, el costo mensual del fármaco sería de aproximadamente USD 12.288,53 (ARS 4.491.458 noviembre/23).


Subject(s)
Immunoglobulin G/administration & dosage , Homozygous Familial Hypercholesterolemia/drug therapy , Argentina , Efficacy , Cost-Benefit Analysis
4.
Proc Natl Acad Sci U S A ; 119(10): e2123002119, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35235456

ABSTRACT

Therapeutic human IgG antibodies are routinely tested in mouse models of oncologic, infectious, and autoimmune diseases. However, assessing the efficacy and safety of long-term administration of these agents has been limited by endogenous anti-human IgG immune responses that act to clear human IgG from serum and relevant tissues, thereby reducing their efficacy and contributing to immune complex­mediated pathologies, confounding evaluation of potential toxicity. For this reason, human antibody treatment in mice is generally limited in duration and dosing, thus failing to recapitulate the potential clinical applications of these therapeutics. Here, we report the development of a mouse model that is tolerant of chronic human antibody administration. This model combines both a human IgG1 heavy chain knock-in and a full recapitulation of human Fc receptor (FcγR) expression, providing a unique platform for in vivo testing of human monoclonal antibodies with relevant receptors beyond the short term. Compared to controls, hIgG1 knock-in mice mount minimal anti-human IgG responses, allowing for the persistence of therapeutically active circulating human IgG even in the late stages of treatment in chronic models of immune thrombocytopenic purpura and metastatic melanoma.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Immunoglobulin G/immunology , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/toxicity , Antibody Formation/genetics , Chronic Disease , Humans , Immune Tolerance , Immunoglobulin G/administration & dosage , Immunoglobulin G/genetics , Immunoglobulin Heavy Chains/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Transgenic , Models, Animal , Purpura, Thrombocytopenic, Idiopathic/immunology , Purpura, Thrombocytopenic, Idiopathic/therapy
5.
Pharm Res ; 39(2): 239-250, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35118567

ABSTRACT

PURPOSE: We have hypothesized that a high concentration of circulating monocytes and macrophages may contribute to the fast weight-based clearance of monoclonal antibodies (mAbs) in young children. Exploring this hypothesis, this work uses modeling to clarify the role of monocytes and macrophages in the elimination of mAbs. METHODS: Leveraging pre-clinical data from mice, a minimal physiologically-based pharmacokinetic model was developed to characterize mAb uptake and FcRn-mediated recycling in circulating monocytes, macrophages, and endothelial cells. The model characterized IgG disposition in complex scenarios of site-specific FcRn deletion and variable endogenous IgG levels. Evaluation was performed for predicting IgG disposition with co-administration of high dose IVIG. A one-at-a-time sensitivity analysis quantified the role of relevant cellular parameters on IgG elimination in various scenarios. RESULTS: The plasma AUC of mAbs was highly sensitive to endothelial cell parameters, but had near-nil sensitivity to monocyte and macrophage parameters, even in scenarios with 90% loss of FcRn expression/activity. In mice with normal FcRn expression, simulations suggest that less than 2% of an IV dose is eliminated in macrophages, while endothelial cells are predicted to dominate mAb elimination. CONCLUSIONS: The model suggests that the role of monocytes and macrophages in IgG homeostasis includes extensive uptake and highly efficient FcRn-mediated protection, but not appreciable degradation when FcRn is present. Therefore, it is very unlikely that a high concentration of circulating monocytes can contribute to explaining the fast weight-based clearance of mAbs in very young children, even if FcRn expression/activity was 90% lower in children than in adults.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Histocompatibility Antigens Class I/metabolism , Immunoglobulin G/metabolism , Macrophages/metabolism , Models, Biological , Monocytes/metabolism , Receptors, Fc/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Drug Elimination Routes , Endothelial Cells/metabolism , Histocompatibility Antigens Class I/genetics , Immunoglobulin G/administration & dosage , Immunoglobulins, Intravenous/administration & dosage , Injections, Intravenous , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Fc/genetics
6.
Int Immunopharmacol ; 104: 108472, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35008008

ABSTRACT

A population pharmacokinetic (PK) model for comparing the PK of subcutaneously administered immunoglobulin G (IgG) replacement therapy (SCIG) with Gamunex-C 10% or SCIG 20% formulations in patients with primary immunodeficiency diseases was developed using data from 3 clinical trials (N = 95, 69.5% adults, 30.5% <18 years) of intravenous IG (IVIG) 10% and SCIG 10% or SCIG 20%. Serum IgG exposure following switches from IVIG 10% every 3 or 4 weeks to biweekly SCIG 20% (dose adjustment factor 1.0 or 1.37) and from weekly SCIG 20% to biweekly SCIG 20% or SCIG 20% 2-7 times/week was simulated. The PK of IVIG 10% and SCIG 20% were adequately described by a 2-compartment model with first-order absorption rate constant of exogenous IgG from an SC depot compartment into the central compartment and first-order elimination from the central compartment. Switching from IVIG 10% every 4 weeks to biweekly SCIG 20% produced similar serum IgG exposure, with lower peak and higher trough serum IgG concentrations. Switching from IVIG 10% every 3 or 4 weeks to weekly and biweekly SCIG 20% yielded comparable IgG exposure and clinically effective trough IgG concentrations.


Subject(s)
Immunoglobulin G/administration & dosage , Models, Biological , Primary Immunodeficiency Diseases/metabolism , Administration, Intravenous , Adolescent , Adult , Aged , Child , Child, Preschool , Computer Simulation , Cross-Over Studies , Female , Humans , Immunoglobulin G/blood , Injections, Subcutaneous , Male , Middle Aged , Primary Immunodeficiency Diseases/blood , Young Adult
7.
Immunohorizons ; 6(1): 90-103, 2022 01 24.
Article in English | MEDLINE | ID: mdl-35074850

ABSTRACT

Maternal alloantibodies toward paternally inherited Ags on fetal platelets can cause thrombocytopenia and bleeding complications in the fetus or neonate, referred to as fetal and neonatal alloimmune thrombocytopenia (FNAIT). This is most commonly caused by Abs against the human platelet Ag (HPA)-1a in Caucasians, and a prophylactic regimen to reduce the risk for alloimmunization to women at risk would be beneficial. We therefore aimed to examine the prophylactic potential of a fully human anti-HPA-1a IgG1 (mAb 26.4) with modified Fc region or altered N-glycan structures. The mAb 26.4 wild-type (WT) variants all showed efficient platelet clearance capacity and ability to mediate phagocytosis independent of their N-glycan structure, compared with an effector silent variant (26.4.AAAG), although the modified N-glycan variants showed differential binding to FcγRs measured in vitro. In an in vivo model, female mice were transfused with platelets from transgenic mice harboring an engineered integrin ß3 containing the HPA-1a epitope. When these preimmunized mice were bred with transgenic males, Abs against the introduced epitope induced thrombocytopenia in the offspring, mimicking FNAIT. Prophylactic administration of the mAb 26.4.WT, and to some extent the mAb 26.4.AAAG, prior to platelet transfusion resulted in reduced alloimmunization in challenged mice and normal platelet counts in neonates. The notion that the effector silent variant hampered alloimmunization demonstrates that rapid platelet clearance, as seen with mAb 26.4.WT, is not the sole mechanism in action. Our data thus successfully demonstrate efficient Ab-mediated immunosuppression and prevention of FNAIT by anti-HPA-1a monoclonal variants, providing support for potential use in humans.


Subject(s)
Antigens, Human Platelet/immunology , Integrin beta3/immunology , Isoantibodies/blood , Thrombocytopenia, Neonatal Alloimmune/immunology , Thrombocytopenia, Neonatal Alloimmune/prevention & control , Animals , Antibodies, Monoclonal/administration & dosage , Female , Humans , Immunoglobulin G/administration & dosage , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Protein Isoforms , THP-1 Cells
8.
Sci Rep ; 11(1): 24067, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34911992

ABSTRACT

Asunercept (company code APG101 [Apogenix AG]; company code CAN008 [CANbridge Pharmaceuticals]) is a novel glycosylated fusion protein that has shown promising effectiveness in glioblastoma. This Phase I study was initiated to evaluate the tolerability and safety of asunercept in combination with standard radiotherapy and temozolomide (RT/TMZ) in Asian patients with newly diagnosed glioblastoma. This was the Phase I portion of a Phase I/II open label, multicenter trial of asunercept plus standard RT/TMZ. Adults with newly-diagnosed glioblastoma received surgical resection followed by standard RT/TMZ plus asunercept 200 mg/week (Cohort 1) or 400 mg/week (Cohort 2) by 30-min IV infusion. The primary endpoint was the safety and tolerability of asunercept during concurrent asunercept and RT/TMZ; dose-limiting toxicities were observed for each dose. Secondary endpoints included pharmacokinetics (PK) and 6-month progression-free survival (PFS6). All patients (Cohort 1, n = 3; Cohort 2, n = 7) completed ≥ 7 weeks of asunercept treatment. No DLTs were experienced. Only one possibly treatment-related treatment emergent adverse event (TEAE), Grade 1 gingival swelling, was observed. No Grade > 3 TEAEs were reported and no TEAE led to treatment discontinuation. Systemic asunercept exposure increased proportionally with dose and showed low inter-patient variability. The PFS6 rate was 33.3% and 57.1% for patients in Cohort 1 and 2, respectively. Patients in Cohort 2 maintained a PFS rate of 57.1% at Month 12. Adding asunercept to standard RT/TMZ was safe and well tolerated in patients with newly-diagnosed glioblastoma and 400 mg/week resulted in encouraging efficacy.Trial registration NCT02853565, August 3, 2016.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/therapy , Glioblastoma/therapy , Radiotherapy , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Asian People , Biomarkers , Brain Neoplasms/diagnosis , Brain Neoplasms/etiology , Brain Neoplasms/mortality , Combined Modality Therapy , Disease Management , Drug Monitoring , Glioblastoma/diagnosis , Glioblastoma/etiology , Glioblastoma/mortality , Humans , Immunoglobulin G/administration & dosage , Kaplan-Meier Estimate , Prognosis , Recombinant Fusion Proteins/administration & dosage , Temozolomide/administration & dosage , Treatment Outcome , fas Receptor/administration & dosage
9.
Front Immunol ; 12: 749891, 2021.
Article in English | MEDLINE | ID: mdl-34867979

ABSTRACT

Broadly neutralising antibodies (bNAbs) may play an important role in future strategies for HIV control. The development of anti-drug antibody (ADA) responses can reduce the efficacy of passively transferred bNAbs but the impact of ADA is imperfectly understood. We previously showed that therapeutic administration of the anti-HIV bNAb PGT121 (either WT or LALA version) controlled viraemia in pigtailed macaques with ongoing SHIV infection. We now report on 23 macaques that had multiple treatments with PGT121. We found that an increasing number of intravenous doses of PGT121 or human IgG1 isotype control antibodies (2-4 doses) results in anti-PGT121 ADA induction and low plasma concentrations of PGT121. ADA was associated with poor or absent suppression of SHIV viremia. Notably, ADA within macaque plasma recognised another human bNAb 10E8 but did not bind to the variable domains of PGT121, suggesting that ADA were primarily directed against the constant regions of the human antibodies. These findings have implications for the development of preclinical studies examining multiple infusions of human bNAbs.


Subject(s)
Broadly Neutralizing Antibodies/administration & dosage , HIV Antibodies/administration & dosage , Immunoglobulin G/administration & dosage , Simian Acquired Immunodeficiency Syndrome/prevention & control , Viremia/prevention & control , Animals , Broadly Neutralizing Antibodies/blood , Broadly Neutralizing Antibodies/immunology , HIV Antibodies/blood , HIV Antibodies/immunology , Macaca nemestrina/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Viremia/immunology
10.
Lancet Haematol ; 8(11): e808-e817, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34627593

ABSTRACT

BACKGROUND: Intravenous TTI-621 (SIRPα-IgG1 Fc) was previously shown to have activity in relapsed or refractory haematological malignancies. This phase 1 study evaluated the safety and activity of TTI-621 in patients with percutaneously accessible relapsed or refractory mycosis fungoides, Sézary syndrome, or solid tumours. Here we report the clinical and translational results among patients with mycosis fungoides or Sézary syndrome. METHODS: This multicentre, open-label, phase 1 study was conducted at five academic health-care and research centres in the USA. Eligible patients were aged 18 years or older; had injectable, histologically or cytologically confirmed relapsed or refractory cutaneous T-cell lymphoma (CTCL) or solid tumours; Eastern Cooperative Oncology Group performance status of 2 or less; and adequate haematological, renal, hepatic, and cardiac function. TTI-621 was injected intralesionally in a sequential dose escalation (cohorts 1-5; single 1 mg, 3 mg, or 10 mg injection or three 10 mg injections weekly for 1 or 2 weeks) and in expansion cohorts (cohorts 6-9; 2 week induction at the maximum tolerated dose; weekly continuation was allowed). In cohort 6, patients were injected with TTI-621 in a single lesion and in cohort 7, they were injected in multiple lesions. In cohort 8, TTI-621 was combined with pembrolizumab 200 mg injections per product labels. In cohort 9, TTI-621 was combined with the standard labelled dose of subcutaneous pegylated interferon alpha-2a 90 µg. The primary endpoint was the incidence and severity of adverse events. The study is registered with ClinicalTrials.gov, NCT02890368, and was closed by the sponsor to focus on intravenous studies with TTI-621. FINDINGS: Between Jan 30, 2017, and March 31, 2020, 66 patients with mycosis fungoides, Sézary syndrome, other CTCL, or solid tumours were screened, 35 of whom with mycosis fungoides or Sézary syndrome were enrolled and received intralesional TTI-621 (escalation, n=13; expansion, n=22). No dose-limiting toxicities occurred; the maximum tolerated dose was not established. In the dose expansion cohorts, the maximally assessed regimen (10 mg thrice weekly for 2 weeks) was used. 25 (71%) patients had treatment-related adverse events; the most common (occurring in ≥10% of patients) were chills (in ten [29%] patients), injection site pain (nine [26%]), and fatigue (eight [23%]). No treatment-related adverse events were grade 3 or more or serious. There were no treatment-related deaths. Rapid responses (median 45 days, IQR 17-66) occurred independently of disease stage or injection frequency. 26 (90%) of 29 evaluable patients had decreased Composite Assessment of Index Lesion Severity (CAILS) scores; ten (34%) had a decrease in CAILS score of 50% or more (CAILS response). CAILS score reductions occurred in adjacent non-injected lesions in eight (80%) of ten patients with paired assessments and in distal non-injected lesions in one additional patient. INTERPRETATION: Intralesional TTI-621 was well tolerated and had activity in adjacent or distal non-injected lesions in patients with relapsed or refractory mycosis fungoides or Sézary syndrome, suggesting it has systemic and locoregional abscopal effects and potential as an immunotherapy for these conditions. FUNDING: Trillium Therapeutics.


Subject(s)
CD47 Antigen/antagonists & inhibitors , Immune Checkpoint Inhibitors/therapeutic use , Immunoglobulin G/therapeutic use , Mycosis Fungoides/drug therapy , Sezary Syndrome/drug therapy , Skin Neoplasms/drug therapy , Aged , CD47 Antigen/immunology , Female , Humans , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/adverse effects , Immunoglobulin G/administration & dosage , Immunoglobulin G/adverse effects , Male , Maximum Tolerated Dose , Middle Aged , Mycosis Fungoides/immunology , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/immunology , Sezary Syndrome/immunology , Skin Neoplasms/immunology
11.
Front Immunol ; 12: 750160, 2021.
Article in English | MEDLINE | ID: mdl-34712239

ABSTRACT

Epidermolysis bullosa acquisita (EBA) is an autoimmune blistering disorder characterized and caused by autoantibodies against type VII collagen (COL7). Although it has been noticed that EBA in both patients and mice is associated with an increased scratching, it is not clear whether and how the scratching contributes to disease manifestation. Hence, we here aimed to validate this clinical observation and also to investigate the potential contribution of increased scratching in EBA pathogenesis in mice. Longitudinal assessment of scratching behavior revealed an increased frequency of scratching as early as 12 hours after injection of anti-COL7 IgG into the skin of mice. Subsequently, scratching events became even more frequent in mice. In contrast, mice injected with a control antibody showed an unaltered scratching behavior throughout the observation period. Based on these observations, we hypothesized that mechanical irritation may promote the induction of inflammation in experimental EBA. To challenge this assumption, the local anesthetic dyclonine hydrochloride was topically applied before injection of anti-COL7 IgG. Dyclonine hydrochloride reduced the scratching events and impaired clinical disease manifestation. In therapeutic experimental settings, i.e. administration of the local anesthetic 24 hours after injection of anti-COL7 IgG, dyclonine hydrochloride only inhibited the scratching behavior, but had no significant effect on clinical disease development. In addition, eosinophils were detected in the skin before the injection of anti-COL7 IgG and significantly increased 48 hours after the antibody injection. Collectively, our results suggest that scratching behavior contributes to the initiation phase of disease manifestation in experimental EBA.


Subject(s)
Anesthetics, Local/administration & dosage , Epidermolysis Bullosa Acquisita/drug therapy , Propiophenones/administration & dosage , Administration, Topical , Animals , Collagen Type VII/immunology , Disease Models, Animal , Female , Immunoglobulin G/administration & dosage , Mice, Inbred BALB C
12.
PLoS One ; 16(10): e0258831, 2021.
Article in English | MEDLINE | ID: mdl-34665826

ABSTRACT

Cancer causes mitochondrial alterations in skeletal muscle, which may progress to muscle wasting and, ultimately, to cancer cachexia. Understanding how exercise adaptations are altered by cancer and cancer treatment is important for the effective design of exercise interventions aimed at improving cancer outcomes. We conducted an exploratory study to investigate how tumor burden and cancer immunotherapy treatment (anti-PD-1) modify the skeletal muscle mitochondrial response to exercise training in mice with transplantable tumors (B16-F10 melanoma and EO771 breast cancer). Mice remained sedentary or were provided with running wheels for ~19 days immediately following tumor implant while receiving no treatment (Untreated), isotype control antibody (IgG2a) or anti-PD-1. Exercise and anti-PD-1 did not alter the growth rate of either tumor type, either alone or in combination therapy. Untreated mice with B16-F10 tumors showed increases in most measured markers of skeletal muscle mitochondrial content following exercise training, as did anti-PD-1-treated mice, suggesting increased mitochondrial content following exercise training in these groups. However, mice with B16-F10 tumors receiving the isotype control antibody did not exhibit increased skeletal muscle mitochondrial content following exercise. In untreated mice with EO771 tumors, only citrate synthase activity and complex IV activity were increased following exercise. In contrast, IgG2a and anti-PD-1-treated groups both showed robust increases in most measured markers following exercise. These results indicate that in mice with B16-F10 tumors, IgG2a administration prevents exercise adaptation of skeletal muscle mitochondria, but adaptation remains intact in mice receiving anti-PD-1. In mice with EO771 tumors, both IgG2a and anti-PD-1-treated mice show robust skeletal muscle mitochondrial exercise responses, while untreated mice do not. Taken together, we postulate that immune modulation may enhance skeletal muscle mitochondrial response to exercise in tumor-bearing mice, and suggest this as an exciting new avenue for future research in exercise oncology.


Subject(s)
Immune Checkpoint Inhibitors/administration & dosage , Immunoglobulin G/administration & dosage , Mammary Neoplasms, Experimental/therapy , Melanoma, Experimental/therapy , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Physical Conditioning, Animal/methods , Animals , Cell Line, Tumor , Citrate (si)-Synthase/metabolism , Electron Transport Complex IV/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Immune Checkpoint Inhibitors/pharmacology , Immunoglobulin G/pharmacology , Immunotherapy , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice , Muscle, Skeletal/drug effects , Random Allocation , Treatment Outcome
13.
Int Immunopharmacol ; 100: 108044, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34601202

ABSTRACT

BACKGROUND: The pharmacokinetics of Ig20Gly, a 20% subcutaneous immunoglobulin (IG) therapy, is well characterized in IG-experienced patients with primary immunodeficiency diseases (PID). Data from IG-naïve patients are limited. OBJECTIVE: Simulate serum total immunoglobulin G (IgG) pharmacokinetic profiles in IG-naïve patients with PID for different Ig20Gly initiation and maintenance dosing regimens. METHODS: A population pharmacokinetic model developed with data from pivotal phase 2/3 trials of weekly Ig20Gly in PID (NCT01412385, NCT01218438) was used to simulate pharmacokinetic profiles of IgG in various scenarios with 400- or 800-mg/kg total loading doses (administered as split doses over 1-2 weeks) and corresponding 100- or 200-mg/kg weekly maintenance doses, respectively. Endogenous baseline IgG levels (1.5, 2.0, 4.0, 6.0 g/L) were evaluated for each scenario; time to putative therapeutic target IgG trough level (7 g/L) was determined. RESULTS: Serum IgG levels reached steady-state by approximately Week 12 for all scenarios and baseline endogenous IgG levels. Time to target trough level generally occurred sooner with 1-week versus 2-week loading schemes. Endogenous baseline IgG levels <4 g/L required a 1-week 800-mg/kg total loading dose to achieve target levels within 2 weeks. Both maintenance regimens sustained serum IgG above target level. CONCLUSIONS: Simulations indicated IG-naïve patients with PID can achieve protective serum IgG levels within 1-3 weeks using appropriate Ig20Gly loading regimens. Patients with low endogenous IgG may benefit most from an 800-mg/kg/month loading dose. 400- or 800-mg/kg/month Ig20Gly maintenance regimens appeared adequate to maintain stable IgG levels. Serum IgG monitoring and clinical status can guide dosing parameters.


Subject(s)
Immunoglobulin G/administration & dosage , Immunologic Factors/pharmacokinetics , Models, Biological , Primary Immunodeficiency Diseases/drug therapy , Adolescent , Adult , Biological Variation, Population , Child , Child, Preschool , Computer Simulation , Drug Administration Schedule , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunologic Factors/administration & dosage , Immunologic Factors/blood , Injections, Subcutaneous , Male , Primary Immunodeficiency Diseases/immunology , Young Adult
14.
Clin Exp Immunol ; 206(3): 422-438, 2021 12.
Article in English | MEDLINE | ID: mdl-34487545

ABSTRACT

Graft versus host disease (GvHD) is a major clinical problem with a significant unmet medical need. We examined the role of cytotoxic T lymphocyte antigen-4 (CTLA-4) in a xenogenic GvHD (xeno-GvHD) model induced by injection of human peripheral mononuclear cells (hPBMC) into irradiated non-obese diabetic (NOD) SCID gamma (NSG) mice. Targeting the CTLA-4 pathway by treatment with CTLA-4 immunoglobulin (Ig) prevented xeno-GvHD, while anti-CTLA-4 antibody treatment exacerbated the lethality and morbidity associated with GvHD. Xeno-GvHD is associated with infiltration of hPBMCs into the lungs, spleen, stomach, liver and colon and an increase in human proinflammatory cytokines, including interferon (IFN)-γ, tumor necrosis factor (TNF)-α and interleukin (IL)-5. Infiltration of donor cells and increases in cytokines were attenuated by treatment with CTLA-4 Ig, but remained either unaffected or enhanced by anti-CTLA-4 antibody. Further, splenic human T cell phenotyping showed that CTLA-4 Ig treatment prevented the engraftment of human CD45+ cells, while anti-CTLA-4 antibody enhanced donor T cell expansion, particularly CD4+ (CD45RO+ ) subsets, including T box transcription factor TBX21 (Tbet)+ CXCR3+ and CD25+ forkhead box protein 3 (FoxP3) cells. Comprehensive analysis of transcriptional profiling of human cells isolated from mouse spleen identified a set of 417 differentially expressed genes (DEGs) by CTLA-4 Ig treatment and 13 DEGs by anti-CTLA-4 antibody treatment. The CTLA-4 Ig regulated DEGs mapped to down-regulated apoptosis, inflammasome, T helper type 17 (Th17) and regulatory T cell (Treg ) pathways and enhanced Toll-like receptor (TLR) receptor signaling, TNF family signaling, complement system and epigenetic and transcriptional regulation, whereas anti-CTLA-4 antibody produced minimal to no impact on these gene pathways. Our results show an important role of co-inhibitory CTLA-4 signaling in xeno-GvHD and suggest the therapeutic utility of other immune checkpoint co-inhibitory pathways in the treatment of immune-mediated diseases driven by hyperactive T cells.


Subject(s)
CTLA-4 Antigen/immunology , Cytokines/blood , Graft vs Host Disease/immunology , Heterografts/immunology , Leukocytes, Mononuclear/immunology , Alanine Transaminase/blood , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Aspartate Aminotransferases/blood , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/immunology , Ipilimumab/pharmacology , Lymphocyte Activation/immunology , Mice , Mice, Inbred NOD , Mice, SCID , T-Lymphocytes, Cytotoxic/immunology
15.
mSphere ; 6(5): e0055621, 2021 10 27.
Article in English | MEDLINE | ID: mdl-34494884

ABSTRACT

Discovered in 1931, Rift Valley fever virus (RVFV) is an arbovirus that causes disease in humans and livestock. In humans, disease ranges from a self-limiting febrile illness to a more severe hepatitis or encephalitis. There are currently no licensed human therapeutics for RVFV disease. Given the recent advances in the use of monoclonal antibodies (MAbs) for treating infectious disease, a panel of anti-RVFV Gn glycoprotein MAbs was developed and characterized. RVFV MAbs spanned a range of neutralizing abilities and mapped to distinct epitopes along Gn. The contribution of Fc effector functions in providing MAb-mediated protection from RVFV was assessed. IgG2a version MAbs had increased capacity to induce effector functions and conferred better protection from RVFV challenge in a lethal mouse model than IgG1 version MAbs. Overall, this study shows that Fc-mediated functions are a critical component of humoral protection from RVFV. IMPORTANCE Rift Valley fever virus (RVFV) is a mosquito-borne virus found throughout Africa and into the Middle East. It has a substantial disease burden; in areas of endemicity, up to 60% of adults are seropositive. With a case fatality rate of up to 3% and the ability to cause hemorrhagic fever and encephalitis, RVFV poses a serious threat to human health. Despite the known human disease burden and the fact that it is a NIAID category A priority pathogen and a WHO priority disease for research and development, there are no vaccines or therapeutics available for RVF. In this study, we developed and characterized a panel of monoclonal antibodies against the RVFV surface glycoprotein, Gn. We then demonstrated therapeutic efficacy in the prevention of RVF in vivo in an otherwise lethal mouse model. Finally, we revealed a role for Fc-mediated function in augmenting the protection provided by these antibodies.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Viral/administration & dosage , Glycoproteins/immunology , Immunoglobulin G/administration & dosage , Rift Valley Fever/prevention & control , Animals , Disease Models, Animal , Epitopes/immunology , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Rift Valley Fever/immunology , Rift Valley fever virus/immunology , Survival Analysis , Treatment Outcome
16.
Article in English | MEDLINE | ID: mdl-34580181

ABSTRACT

BACKGROUND AND OBJECTIVES: To determine in a mouse model whether neonatal Fc receptor (FcRn) blockade prevents the placental transfer of class G immunoglobulin (IgG) derived from patients with anti-NMDA receptor (NMDAR) encephalitis and their pathogenic effects on the fetuses and offspring. METHODS: Pregnant C57BL/6J mice were administered via tail vein FcRn antibody (FcRn-ab) or saline solution 6 hours before administration of patients' or controls' IgG on days 14, 15, and 16 of gestation. Three experimental groups were established: mice receiving controls' IgG, patients' IgG, or patients' IgG along with pretreatment with FcRn-ab. Immunohistochemical staining, confocal microscopy, hippocampal long-term potentiation, and standardized developmental and behavioral tasks were used to assess the efficacy of treatment with FcRn-ab. RESULTS: In pregnant mice that received patients' IgG, treatment with FcRn-ab prevented the IgG from reaching the fetal brain, abrogating the decrease of NMDAR clusters and the reduction of cortical plate thickness that were observed in fetuses from untreated pregnant mice. Moreover, among the offspring of mothers that received patients' IgG, those whose mothers were treated with FcRn-ab did not develop the alterations that occurred in offspring of untreated mothers, including impairment in hippocampal plasticity, delay in innate reflexes, and visuospatial memory deficits. DISCUSSION: FcRn blockade prevents placental transfer of IgG from patients with anti-NMDAR encephalitis and abrogates the synaptic and neurodevelopmental alterations caused by patients' antibodies. This model has potential therapeutic implications for other antibody-mediated diseases of the CNS during pregnancy.


Subject(s)
Anti-N-Methyl-D-Aspartate Receptor Encephalitis/immunology , Antibodies, Blocking/administration & dosage , Autoantibodies/administration & dosage , Histocompatibility Antigens Class I/immunology , Immunoglobulin G/administration & dosage , Maternal-Fetal Exchange/drug effects , Placental Circulation/drug effects , Receptors, Fc/immunology , Animals , Animals, Newborn , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred C57BL , Pregnancy
17.
Sci Rep ; 11(1): 17440, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34465819

ABSTRACT

The use of antibody-based therapies for the treatment of high consequence viral pathogens has gained interest over the last fifteen years. Here, we sought to evaluate the use of unique camelid-based IgG antibodies to prevent lethal hantavirus pulmonary syndrome (HPS) in Syrian hamsters. Using purified, polyclonal IgG antibodies generated in DNA-immunized alpacas, we demonstrate that post-exposure treatments reduced viral burdens and organ-specific pathology associated with lethal HPS. Antibody treated animals did not exhibit signs of disease and were completely protected. The unique structures and properties, particularly the reduced size, distinct paratope formation and increased solubility of camelid antibodies, in combination with this study support further pre-clinical evaluation of heavy-chain only antibodies for treatment of severe respiratory diseases, including HPS.


Subject(s)
Antibodies, Viral/administration & dosage , Disease Models, Animal , Glycoproteins/immunology , Hantavirus Infections/prevention & control , Hantavirus Pulmonary Syndrome/prevention & control , Immunoglobulin G/administration & dosage , Orthohantavirus/immunology , Animals , Antibodies, Viral/immunology , Camelids, New World , Female , Hantavirus Infections/immunology , Hantavirus Infections/virology , Hantavirus Pulmonary Syndrome/immunology , Hantavirus Pulmonary Syndrome/virology , Immunoglobulin G/immunology , Male , Mesocricetus
18.
Nat Commun ; 12(1): 5151, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34446712

ABSTRACT

Poorly inflamed carcinomas do not respond well to immune checkpoint blockade. Converting the tumour microenvironment into a functionally inflamed immune hub would extend the clinical benefit of immune therapy to a larger proportion of cancer patients. Here we show, by using comprehensive single-cell transcriptome, proteome, and immune cell analysis, that Entinostat, a class I histone deacetylase inhibitor, facilitates accumulation of the necrosis-targeted recombinant murine immune-cytokine, NHS-rmIL12, in experimental mouse colon carcinomas and poorly immunogenic breast tumours. This combination therapy reprograms the tumour innate and adaptive immune milieu to an inflamed landscape, where the concerted action of highly functional CD8+ T cells and activated neutrophils drive macrophage M1-like polarization, leading to complete tumour eradication in 41.7%-100% of cases. Biomarker signature of favourable overall survival in multiple human tumor types shows close resemblance to the immune pattern generated by Entinostat/NHS-rmIL12 combination therapy. Collectively, these findings provide a rationale for combining NHS-IL12 with Entinostat in the clinical setting.


Subject(s)
Benzamides/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Immunoglobulin G/administration & dosage , Interleukin-12/administration & dosage , Pyridines/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Adaptive Immunity/drug effects , Animals , Breast Neoplasms/mortality , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Colonic Neoplasms/mortality , Drug Therapy, Combination , Female , Humans , Immunity, Innate/drug effects , Macrophages/immunology , Mice , Mice, Inbred BALB C , Tumor Microenvironment/drug effects
20.
J Clin Invest ; 131(13)2021 07 01.
Article in English | MEDLINE | ID: mdl-34196305

ABSTRACT

Fibromyalgia syndrome (FMS) is characterized by widespread pain and tenderness, and patients typically experience fatigue and emotional distress. The etiology and pathophysiology of fibromyalgia are not fully explained and there are no effective drug treatments. Here we show that IgG from FMS patients produced sensory hypersensitivity by sensitizing nociceptive neurons. Mice treated with IgG from FMS patients displayed increased sensitivity to noxious mechanical and cold stimulation, and nociceptive fibers in skin-nerve preparations from mice treated with FMS IgG displayed an increased responsiveness to cold and mechanical stimulation. These mice also displayed reduced locomotor activity, reduced paw grip strength, and a loss of intraepidermal innervation. In contrast, transfer of IgG-depleted serum from FMS patients or IgG from healthy control subjects had no effect. Patient IgG did not activate naive sensory neurons directly. IgG from FMS patients labeled satellite glial cells and neurons in vivo and in vitro, as well as myelinated fiber tracts and a small number of macrophages and endothelial cells in mouse dorsal root ganglia (DRG), but no cells in the spinal cord. Furthermore, FMS IgG bound to human DRG. Our results demonstrate that IgG from FMS patients produces painful sensory hypersensitivities by sensitizing peripheral nociceptive afferents and suggest that therapies reducing patient IgG titers may be effective for fibromyalgia.


Subject(s)
Fibromyalgia/immunology , Fibromyalgia/physiopathology , Animals , Case-Control Studies , Disease Models, Animal , Female , Fibromyalgia/etiology , Ganglia, Spinal/physiopathology , Humans , Immunization, Passive , Immunoglobulin G/administration & dosage , Immunoglobulin G/blood , Male , Mice , Mice, Inbred C57BL , Nociceptors/immunology , Nociceptors/physiology , Pain/physiopathology , Pain Threshold/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...