Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 356
Filtrar
1.
Med ; 3(3): 157-158, 2022 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-35291730

RESUMEN

The therapeutic and prophylactic uses of monoclonal antibodies (mABs) against SARS-CoV-2 are limited by their short half-life and need for intravenous delivery. In this issue, Cobb et al.1 engineer a neutralizing mAB cocktail with extended half-life that can be delivered intramuscularly to provide prophylactic protection against infection in rhesus macaques.


Asunto(s)
Antineoplásicos Inmunológicos , COVID-19 , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico , COVID-19/prevención & control , Humanos , Macaca fascicularis/inmunología , Macaca mulatta/inmunología , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/inmunología
2.
Nature ; 601(7893): 410-414, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34794169

RESUMEN

The CVnCoV (CureVac) mRNA vaccine for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) was recently evaluated in a phase 2b/3 efficacy trial in humans1. CV2CoV is a second-generation mRNA vaccine containing non-modified nucleosides but with optimized non-coding regions and enhanced antigen expression. Here we report the results of a head-to-head comparison of the immunogenicity and protective efficacy of CVnCoV and CV2CoV in non-human primates. We immunized 18 cynomolgus macaques with two doses of 12 µg lipid nanoparticle-formulated CVnCoV or CV2CoV or with sham (n = 6 per group). Compared with CVnCoV, CV2CoV induced substantially higher titres of binding and neutralizing antibodies, memory B cell responses and T cell responses as well as more potent neutralizing antibody responses against SARS-CoV-2 variants, including the Delta variant. Moreover, CV2CoV was found to be comparably immunogenic to the BNT162b2 (Pfizer) vaccine in macaques. Although CVnCoV provided partial protection against SARS-CoV-2 challenge, CV2CoV afforded more robust protection with markedly lower viral loads in the upper and lower respiratory tracts. Binding and neutralizing antibody titres were correlated with protective efficacy. These data demonstrate that optimization of non-coding regions can greatly improve the immunogenicity and protective efficacy of a non-modified mRNA SARS-CoV-2 vaccine in non-human primates.


Asunto(s)
Vacunas contra la COVID-19/genética , Vacunas contra la COVID-19/inmunología , COVID-19/prevención & control , Inmunogenicidad Vacunal , Nucleósidos/química , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas de ARNm/genética , Vacunas de ARNm/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Vacuna BNT162/inmunología , COVID-19/inmunología , COVID-19/virología , Vacunas contra la COVID-19/normas , Femenino , Macaca fascicularis/inmunología , Masculino , Células B de Memoria/inmunología , Nucleósidos/genética , Sistema Respiratorio/inmunología , Sistema Respiratorio/virología , SARS-CoV-2/inmunología , Linfocitos T/inmunología , Vacunas Sintéticas/normas , Carga Viral , Vacunas de ARNm/normas
3.
PLoS One ; 16(12): e0260010, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34855793

RESUMEN

Little is known about how specific individual viral lineages replicating systemically during acute Human Immunodeficiency Virus or Simian Immunodeficiency Virus (HIV/SIV) infection persist into chronic infection. In this study, we use molecularly barcoded SIV (SIVmac239M) to track distinct viral lineages for 12 weeks after intravenous (IV) or intrarectal (IR) challenge in macaques. Two Mafa-A1*063+ cynomolgus macaques (Macaca fascicularis, CM) were challenged IV, and two Mamu-A1*001+ rhesus macaques (Macaca mulatta, RM) were challenged IR with 200,000 Infectious Units (IU) of SIVmac239M. We sequenced the molecular barcode of SIVmac239M from all animals over the 12 weeks of the study to characterize the diversity and persistence of virus lineages. During the first three weeks post-infection, we found ~70-560 times more unique viral lineages circulating in the animals challenged IV compared to those challenged IR, which is consistent with the hypothesis that the challenge route is the primary driver restricting the transmission of individual viral lineages. We also characterized the sequences of T cell epitopes targeted during acute SIV infection, and found that the emergence of escape variants in acutely targeted epitopes can occur on multiple virus templates simultaneously, but that elimination of some of these templates is likely a consequence of additional host factors. These data imply that virus lineages present during acute infection can still be eliminated from the systemic virus population even after initial selection.


Asunto(s)
Membrana Mucosa/virología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/crecimiento & desarrollo , Animales , Epítopos/inmunología , Femenino , Productos del Gen tat/genética , Inyecciones Intravenosas , Macaca fascicularis/inmunología , Macaca fascicularis/virología , Macaca mulatta/inmunología , Macaca mulatta/virología , Membrana Mucosa/inmunología , Mutación , ARN Viral/sangre , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Linfocitos T/inmunología , Linfocitos T/virología , Carga Viral , Viremia/inmunología , Viremia/virología
4.
Proc Natl Acad Sci U S A ; 118(43)2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34625475

RESUMEN

The pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a global threat to human health and life. A useful pathological animal model accurately reflecting human pathology is needed to overcome the COVID-19 crisis. In the present study, COVID-19 cynomolgus monkey models including monkeys with underlying diseases causing severe pathogenicity such as metabolic disease and elderly monkeys were examined. Cynomolgus macaques with various clinical conditions were intranasally and/or intratracheally inoculated with SARS-CoV-2. Infection with SARS-CoV-2 was found in mucosal swab samples, and a higher level and longer period of viral RNA was detected in elderly monkeys than in young monkeys. Pneumonia was confirmed in all of the monkeys by computed tomography images. When monkeys were readministrated SARS-CoV-2 at 56 d or later after initial infection all of the animals showed inflammatory responses without virus detection in swab samples. Surprisingly, in elderly monkeys reinfection showed transient severe pneumonia with increased levels of various serum cytokines and chemokines compared with those in primary infection. The results of this study indicated that the COVID-19 cynomolgus monkey model reflects the pathophysiology of humans and would be useful for elucidating the pathophysiology and developing therapeutic agents and vaccines.


Asunto(s)
COVID-19/inmunología , Modelos Animales de Enfermedad , Macaca fascicularis/inmunología , Enfermedades de los Primates/inmunología , SARS-CoV-2/inmunología , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , COVID-19/virología , Femenino , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Pulmón/diagnóstico por imagen , Pulmón/inmunología , Pulmón/virología , Macaca fascicularis/virología , Masculino , Enfermedades de los Primates/virología , SARS-CoV-2/fisiología , Tomografía Computarizada por Rayos X/métodos , Esparcimiento de Virus/inmunología , Esparcimiento de Virus/fisiología
5.
PLoS Pathog ; 17(7): e1009668, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34280241

RESUMEN

SARS-CoV-2 infection presents clinical manifestations ranging from asymptomatic to fatal respiratory failure. Despite the induction of functional SARS-CoV-2-specific CD8+ T-cell responses in convalescent individuals, the role of virus-specific CD8+ T-cell responses in the control of SARS-CoV-2 replication remains unknown. In the present study, we show that subacute SARS-CoV-2 replication can be controlled in the absence of CD8+ T cells in cynomolgus macaques. Eight macaques were intranasally inoculated with 105 or 106 TCID50 of SARS-CoV-2, and three of the eight macaques were treated with a monoclonal anti-CD8 antibody on days 5 and 7 post-infection. In these three macaques, CD8+ T cells were undetectable on day 7 and thereafter, while virus-specific CD8+ T-cell responses were induced in the remaining five untreated animals. Viral RNA was detected in nasopharyngeal swabs for 10-17 days post-infection in all macaques, and the kinetics of viral RNA levels in pharyngeal swabs and plasma neutralizing antibody titers were comparable between the anti-CD8 antibody treated and untreated animals. SARS-CoV-2 RNA was detected in the pharyngeal mucosa and/or retropharyngeal lymph node obtained at necropsy on day 21 in two of the untreated group but undetectable in all macaques treated with anti-CD8 antibody. CD8+ T-cell responses may contribute to viral control in SARS-CoV-2 infection, but our results indicate possible containment of subacute viral replication in the absence of CD8+ T cells, implying that CD8+ T-cell dysfunction may not solely lead to viral control failure.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , COVID-19/veterinaria , Macaca fascicularis/inmunología , Macaca fascicularis/virología , Enfermedades de los Monos/inmunología , Enfermedades de los Monos/virología , SARS-CoV-2/inmunología , SARS-CoV-2/fisiología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , COVID-19/inmunología , COVID-19/virología , Modelos Animales de Enfermedad , Femenino , Humanos , Cinética , Depleción Linfocítica/veterinaria , Masculino , ARN Viral/genética , ARN Viral/metabolismo , SARS-CoV-2/genética , Replicación Viral/inmunología
6.
Emerg Microbes Infect ; 10(1): 1457-1470, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34120576

RESUMEN

Zika virus (ZIKV) is an emerging arbovirus with recent global expansion. Historically, ZIKV infections with Asian lineages have been associated with mild disease such as rash and fever. However, recent Asian sub-lineages have caused outbreaks in the South Pacific and Latin America with increased prevalence of neurological disorders in infants and adults. Asian sub-lineage differences may partially explain the range of disease severity observed. However, the effect of Asian sub-lineage differences on pathogenesis remains poorly characterized. Current study conducts a head-to-head comparison of three Asian sub-lineages that are representative of the circulating ancestral mild Asian strain (ZIKV-SG), the 2007 epidemic French Polynesian strain (ZIKV-FP), and the 2013 epidemic Brazil strain (ZIKV-Brazil) in adult Cynomolgus macaques. Animals infected intervenously or subcutaneously with either of the three clinical isolates showed sub-lineage-specific differences in viral pathogenesis, early innate immune responses and systemic inflammation. Despite the lack of neurological symptoms in infected animals, the epidemiologically neurotropic ZIKV sub-lineages (ZIKV-Brazil and/or ZIKV-FP) were associated with more sustained viral replication, higher systemic inflammation (i.e. higher levels of TNFα, MCP-1, IL15 and G-CSF) and greater percentage of CD14+ monocytes and dendritic cells in blood. Multidimensional analysis showed clustering of ZIKV-SG away from ZIKV-Brazil and ZIKV-FP, further confirming sub-lineage differences in the measured parameters. These findings highlight greater systemic inflammation and monocyte recruitment as possible risk factors of adult ZIKV disease observed during the 2007 FP and 2013 Brazil epidemics. Future studies should explore the use of anti-inflammatory therapeutics as early treatment to prevent ZIKV-associated disease in adults.


Asunto(s)
Inmunidad Innata , Infección por el Virus Zika/inmunología , Virus Zika/clasificación , Virus Zika/inmunología , Virus Zika/patogenicidad , Adulto , Animales , Asia , Brasil , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Humanos , Interleucina-15/genética , Interleucina-15/inmunología , Macaca fascicularis/inmunología , Macaca fascicularis/virología , Monocitos/inmunología , Especificidad de la Especie , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Virulencia , Replicación Viral , Virus Zika/genética , Infección por el Virus Zika/virología
7.
Sci Rep ; 11(1): 8617, 2021 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-33883656

RESUMEN

Many groups are working to improve the results of clinical allogeneic islet transplantation in a primate model. However, few studies have focused on the optimal islet dose for achieving normal glycemia without exogenous insulin after transplantation in primate models or on the relationship between rejection and islet amyloid polypeptide (IAPP) expression. We evaluated the dose (10,000, 20,000, and > 25,000 islet equivalents (IEQ)/kg) needed to achieve normal glycemia without exogenous insulin after transplantation using eleven cynomolgus monkeys, and we analyzed the characteristics exhibited in the islets after transplantation. 10,000 IEQ/kg (N = 2) failed to control blood glucose level, despite injection with the highest dose of exogenous insulin, and 20,000 IEQ/kg group (N = 5) achieved unstable control, with a high insulin requirement. However, 25,000 IEQ/kg (N = 4) achieved normal glycemia without exogenous insulin and maintained it for more than 60 days. Immunohistochemistry results from staining islets found in liver biopsies indicated that as the number of transplanted islets decreased, the amount of IAPP accumulation within the islets increased, which accelerated CD3+ T cell infiltration. In conclusion, the optimal transplantation dose for achieving a normal glycemia without exogenous insulin in our cynomolgus monkey model was > 25,000 IEQ/kg, and the accumulation of IAPP early after transplantation, which depends on the transplanted islet dose, can be considered one factor in rejection.


Asunto(s)
Diabetes Mellitus Experimental/inmunología , Insulina/inmunología , Islotes Pancreáticos/inmunología , Macaca fascicularis/inmunología , Animales , Complejo CD3/inmunología , Prueba de Tolerancia a la Glucosa/métodos , Inmunohistoquímica/métodos , Polipéptido Amiloide de los Islotes Pancreáticos/inmunología , Trasplante de Islotes Pancreáticos/métodos , Trasplante Heterólogo/métodos
8.
Sci Rep ; 11(1): 8810, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33893359

RESUMEN

Rhesus (Macaca mulatta) and cynomolgus (Macaca fasicularis) macaques of distinct genetic origin are understood to vary in susceptibility to Mycobacterium tuberculosis, and therefore differences in their immune systems may account for the differences in disease control. Monocyte:lymphocyte (M:L) ratio has been identified as a risk factor for M. tuberculosis infection and is known to vary between macaque species. We aimed to characterise the constituent monocyte and lymphocyte populations between macaque species, and profile other major immune cell subsets including: CD4+ and CD8+ T-cells, NK-cells, B-cells, monocyte subsets and myeloid dendritic cells. We found immune cell subsets to vary significantly between macaque species. Frequencies of CD4+ and CD8+ T-cells and the CD4:CD8 ratio showed significant separation between species, while myeloid dendritic cells best associated macaque populations by M. tuberculosis susceptibility. A more comprehensive understanding of the immune parameters between macaque species may contribute to the identification of new biomarkers and correlates of protection.


Asunto(s)
Susceptibilidad a Enfermedades/inmunología , Macaca fascicularis/inmunología , Macaca mulatta/inmunología , Tuberculosis/inmunología , Animales , Inmunofenotipificación , Subgrupos Linfocitarios , Estudios Retrospectivos , Especificidad de la Especie
9.
MAbs ; 13(1): 1850395, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33459147

RESUMEN

We report here the discovery and optimization of a novel T cell retargeting anti-GUCY2C x anti-CD3ε bispecific antibody for the treatment of solid tumors. Using a combination of hybridoma, phage display and rational design protein engineering, we have developed a fully humanized and manufacturable CD3 bispecific antibody that demonstrates favorable pharmacokinetic properties and potent in vivo efficacy. Anti-GUCY2C and anti-CD3ε antibodies derived from mouse hybridomas were first humanized into well-behaved human variable region frameworks with full retention of binding and T-cell mediated cytotoxic activity. To address potential manufacturability concerns, multiple approaches were taken in parallel to optimize and de-risk the two antibody variable regions. These approaches included structure-guided rational mutagenesis and phage display-based optimization, focusing on improving stability, reducing polyreactivity and self-association potential, removing chemical liabilities and proteolytic cleavage sites, and de-risking immunogenicity. Employing rapid library construction methods as well as automated phage display and high-throughput protein production workflows enabled efficient generation of an optimized bispecific antibody with desirable manufacturability properties, high stability, and low nonspecific binding. Proteolytic cleavage and deamidation in complementarity-determining regions were also successfully addressed. Collectively, these improvements translated to a molecule with potent single-agent in vivo efficacy in a tumor cell line adoptive transfer model and a cynomolgus monkey pharmacokinetic profile (half-life>4.5 days) suitable for clinical development. Clinical evaluation of PF-07062119 is ongoing.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Complejo CD3/inmunología , Inmunoterapia Adoptiva/métodos , Neoplasias/terapia , Receptores de Enterotoxina/inmunología , Animales , Anticuerpos Biespecíficos/farmacocinética , Anticuerpos Biespecíficos/uso terapéutico , Línea Celular Tumoral , Femenino , Humanos , Hibridomas , Macaca fascicularis/inmunología , Macaca fascicularis/metabolismo , Ratones Endogámicos BALB C , Neoplasias/inmunología , Neoplasias/metabolismo , Ingeniería de Proteínas/métodos , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/farmacocinética , Anticuerpos de Cadena Única/uso terapéutico , Linfocitos T/inmunología , Linfocitos T/metabolismo
10.
Front Immunol ; 12: 798207, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35069580

RESUMEN

Tuberculosis (TB) is a major global health problem and the only currently-licensed vaccine, BCG, is inadequate. Many TB vaccine candidates are designed to be given as a boost to BCG; an understanding of the BCG-induced immune response is therefore critical, and the opportunity to relate this to circumstances where BCG does confer protection may direct the design of more efficacious vaccines. While the T cell response to BCG vaccination has been well-characterized, there is a paucity of literature on the humoral response. We demonstrate BCG vaccine-mediated induction of specific antibodies in different human populations and macaque species which represent important preclinical models for TB vaccine development. We observe a strong correlation between antibody titers in serum versus plasma with modestly higher titers in serum. We also report for the first time the rapid and transient induction of antibody-secreting plasmablasts following BCG vaccination, together with a robust and durable memory B cell response in humans. Finally, we demonstrate a functional role for BCG vaccine-induced specific antibodies in opsonizing mycobacteria and enhancing macrophage phagocytosis in vitro, which may contribute to the BCG vaccine-mediated control of mycobacterial growth observed. Taken together, our findings indicate that the humoral immune response in the context of BCG vaccination merits further attention to determine whether TB vaccine candidates could benefit from the induction of humoral as well as cellular immunity.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Vacuna BCG/inmunología , Inmunoglobulina G/inmunología , Células B de Memoria/inmunología , Células Plasmáticas/inmunología , Adulto , Animales , Anticuerpos Antibacterianos/sangre , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/metabolismo , Vacuna BCG/administración & dosificación , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/inmunología , Inmunidad Humoral/efectos de los fármacos , Inmunidad Humoral/inmunología , Inmunoglobulina A/sangre , Inmunoglobulina A/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/metabolismo , Inmunoglobulina M/sangre , Inmunoglobulina M/inmunología , Macaca fascicularis/inmunología , Macaca mulatta/inmunología , Masculino , Células B de Memoria/metabolismo , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/fisiología , Células Plasmáticas/metabolismo , Tuberculosis/inmunología , Tuberculosis/microbiología , Tuberculosis/prevención & control , Vacunación/métodos
11.
Front Immunol ; 12: 815680, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35087534

RESUMEN

Macaques are frequently used to evaluate candidate vaccines and to study infection-induced antibody responses, requiring an improved understanding of their naïve immunoglobulin (IG) repertoires. Baseline gene usage frequencies contextualize studies of antigen-specific immune responses, providing information about how easily one may stimulate a response with a particular VDJ recombination. Studies of human IgM repertoires have shown that IG VDJ gene frequencies vary several orders of magnitude between the most and least utilized genes in a manner that is consistent across many individuals but to date similar analyses are lacking for macaque IgM repertoires. Here, we quantified VDJ gene usage levels in unmutated IgM repertoires of 45 macaques, belonging to two species and four commonly used subgroups: Indian and Chinese origin rhesus macaques and Indonesian and Mauritian origin cynomolgus macaques. We show that VDJ gene frequencies differed greatly between the most and least used genes, with similar overall patterns observed in macaque subgroups and individuals. However, there were also clear differences affecting the use of specific V, D and J genes. Furthermore, in contrast to humans, macaques of both species utilized IGHV4 family genes to a much higher extent and showed evidence of evolutionary expansion of genes of this family. Finally, we used the results to inform the analysis of a broadly neutralizing HIV-1 antibody elicited in SHIV-infected rhesus macaques, RHA1.V2.01, which binds the apex of the Env trimer in a manner that mimics the binding mode of PGT145. We discuss the likelihood that similar antibodies could be elicited in different macaque subgroups.


Asunto(s)
Inmunoglobulina M/genética , Macaca fascicularis/genética , Macaca fascicularis/inmunología , Macaca mulatta/genética , Macaca mulatta/inmunología , Recombinación V(D)J , Animales , Formación de Anticuerpos/genética , Formación de Anticuerpos/inmunología , Biología Computacional/métodos , VIH-1/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunogenética/métodos , Cadenas Pesadas de Inmunoglobulina/genética , Virus de la Inmunodeficiencia de los Simios/inmunología , Especificidad de la Especie
12.
Nature ; 590(7845): 320-325, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33260195

RESUMEN

The expanding pandemic of coronavirus disease 2019 (COVID-19) requires the development of safe, efficacious and fast-acting vaccines. Several vaccine platforms are being leveraged for a rapid emergency response1. Here we describe the development of a candidate vaccine (YF-S0) for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that uses live-attenuated yellow fever 17D (YF17D) vaccine as a vector to express a noncleavable prefusion form of the SARS-CoV-2 spike antigen. We assess vaccine safety, immunogenicity and efficacy in several animal models. YF-S0 has an excellent safety profile and induces high levels of SARS-CoV-2 neutralizing antibodies in hamsters (Mesocricetus auratus), mice (Mus musculus) and cynomolgus macaques (Macaca fascicularis), and-concomitantly-protective immunity against yellow fever virus. Humoral immunity is complemented by a cellular immune response with favourable T helper 1 polarization, as profiled in mice. In a hamster model2 and in macaques, YF-S0 prevents infection with SARS-CoV-2. Moreover, a single dose conferred protection from lung disease in most of the vaccinated hamsters within as little as 10 days. Taken together, the quality of the immune responses triggered and the rapid kinetics by which protective immunity can be attained after a single dose warrant further development of this potent SARS-CoV-2 vaccine candidate.


Asunto(s)
Vacunas contra la COVID-19/inmunología , COVID-19/inmunología , COVID-19/prevención & control , Vectores Genéticos/genética , SARS-CoV-2/inmunología , Vacunas Atenuadas/inmunología , Vacuna contra la Fiebre Amarilla/genética , Animales , Vacunas contra la COVID-19/administración & dosificación , Vacunas contra la COVID-19/efectos adversos , Vacunas contra la COVID-19/genética , Cricetinae , Modelos Animales de Enfermedad , Femenino , Glicosilación , Macaca fascicularis/genética , Macaca fascicularis/inmunología , Macaca fascicularis/virología , Masculino , Mesocricetus/genética , Mesocricetus/inmunología , Mesocricetus/virología , Ratones , Seguridad , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/genética
13.
Front Immunol ; 11: 586251, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193411

RESUMEN

Allogeneic hematopoietic stem cell transplants can lead to dramatic reductions in human immunodeficiency virus (HIV) reservoirs. This effect is partially mediated by donor T cells recognizing lymphocyte-expressed minor histocompatibility antigens (mHAgs). The potential to mark malignant and latently infected cells for destruction makes mHAgs attractive targets for cellular immunotherapies. However, testing such HIV reservoir reduction strategies will likely require preclinical studies in non-human primates (NHPs). In this study, we used a combination of alloimmunization, whole exome sequencing, and bioinformatics to identify an mHAg in Mauritian cynomolgus macaques (MCMs). We mapped the minimal optimal epitope to a 10-mer peptide (SW10) in apolipoprotein B mRNA editing enzyme catalytic polypeptide-like 3C (APOBEC3C) and determined the major histocompatibility complex class I restriction element as Mafa-A1∗063, which is expressed in almost 90% of MCMs. APOBEC3C SW10-specific CD8+ T cells recognized immortalized B cells but not fibroblasts from an mHAg-positive MCM. These results provide a framework for identifying mHAgs in a non-transplant setting and suggest that APOBEC3C SW10 could be used as a model antigen to test mHAg-targeted therapies in NHPs.


Asunto(s)
Citidina Desaminasa/inmunología , Macaca fascicularis/inmunología , Antígenos de Histocompatibilidad Menor/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Epítopos de Linfocito T/inmunología
14.
Monoclon Antib Immunodiagn Immunother ; 39(5): 175-183, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32996830

RESUMEN

During preclinical studies, there is a great need to develop monoclonal antibodies (mAbs) that are specific to human immunoglobulin (IgG), without binding to monkey IgG, to detect therapeutic human mAb in non-human primates. We took advantage of the latest rabbit B cell cloning technology to develop six unique rabbit anti-human IgG mAb clones for this purpose. These clones are capable of binding to both human IgG and Fab with high affinity without nonspecific binding to cynomolgus monkey IgG. These clones have been evaluated as a generic capture reagent for the detection of human IgG and Fab, in the presence of cynomolgus monkey serum, by Gyrolab™ immunoassay. They may be used in singlet or as pairs for the detection of human IgG, in any host animal, to meet the need for therapeutic mAb development in preclinical studies.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Inmunoensayo , Fragmentos Fab de Inmunoglobulinas/inmunología , Inmunoglobulina G/inmunología , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/farmacología , Humanos , Fragmentos Fab de Inmunoglobulinas/biosíntesis , Fragmentos Fab de Inmunoglobulinas/farmacología , Macaca fascicularis/inmunología , Conejos
15.
Life Sci ; 259: 118244, 2020 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-32791145

RESUMEN

AIM: Protein therapeutics have potential to elicit immune responses resulting in undesirable anti-drug antibodies (ADA) that might affect product efficacy and patient safety, and should be assessed in animals before applying the treatment to humans. In this paper, we aim to assess the immunogenicity and toxicokinetics of the mono-PEGylated recombinant human interleukin-11 (rhIL-11), a novel protein therapeutic for the treatment of chemotherapy-induced thrombocytopenia, in repeated administration to cynomolgus monkeys. MAIN METHODS: Enzyme-linked immunosorbent assay (ELISA) methods were developed to measure ADA responses and plasma PEGylated IL-11 (PEG-IL11) concentration in monkeys. Assay parameters of immunogenicity and toxicokinetics methods were evaluated during validation in accordance with regulatory guidelines. We also employed cell-based assays to test the neutralizing activity of ADA provoked in monkeys. KEY FINDINGS: The results showed that weak immunogenicity occurred in some monkeys after receiving repeated dose of 0.1-0.3 mg/kg by subcutaneous administration and disappeared after the recovery period. More pronounced immunogenicity occurred at high dose of 0.9 mg/kg, with a higher positive rate and titer, and some ADAs had neutralizing activity, but it can be greatly reduced after recovery. Such ADAs generated in monkeys may be accounted for the plasma toxicokinetics changes of PEG-IL11 and a minor reduction in systemic exposure. SIGNIFICANCE: These methods have been successfully applied to immunogenicity and toxicokinetic studies of PEG-IL11 in repeated dose toxicity following subcutaneous administration to monkeys, and could be successfully used in clinical trials after some modifications.


Asunto(s)
Interleucina-11/inmunología , Interleucina-11/uso terapéutico , Animales , Anticuerpos Monoclonales/inmunología , Ensayo de Inmunoadsorción Enzimática/métodos , Humanos , Fenómenos Inmunogenéticos/genética , Fenómenos Inmunogenéticos/fisiología , Interleucina-11/metabolismo , Macaca fascicularis/inmunología , Preparaciones Farmacéuticas , Polietilenglicoles/farmacología , Proteínas Recombinantes/uso terapéutico , Toxicocinética
16.
Xenotransplantation ; 27(6): e12632, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32781479

RESUMEN

BACKGROUND: Allogeneic skin recovered from human deceased donors (HDD) has been a mainstay interim treatment for severe burns, but unfortunately risk of infectious disease and availability limitations exist. Genetically engineered ɑ-1,3 galactosyltransferase knockout (GalT-KO) porcine source animals for viable skin xenotransplants may provide a promising clinical alternative. METHODS: Four cynomolgus macaque recipients received full-thickness surgical wounds to model the defects arising from excision of full-thickness burn injury and were treated with biologically active skin xenotransplants derived from GalT-KO, Designated Pathogen Free (DPF) miniature swine. Evaluations were conducted for safety, tolerability, and recipient immunological response. RESULTS: All skin xenotransplants demonstrated prolonged survival, vascularity, and persistent dermal adhesion until the study endpoint at post-operative day 30. No adverse outcomes were observed during the study. Varying levels of epidermolysis coincided with histologic detection of CD4+ and CD8+ T cells, and other cellular infiltrates in the epidermis. Recipient sera IgM and IgG demonstrated significant antibody immune response to non-α-1,3-galactose porcine xenoantigens. Separately, specific wound healing mediators were quantified. Neither porcine cell migration nor PERV were detected in circulation or any visceral organs. CONCLUSIONS: These results provide a detailed analysis of vital skin xenotransplants utilizing a non-human primate model to predict the anticipated immunological response of human patients. The lack of adverse rejection even in the presence of elevated Ig indicates this is a prospective therapeutic option. The findings reported here directly supported regulatory clearance for a first-in-man, Phase I xenotransplantation clinical trial.


Asunto(s)
Macaca fascicularis/inmunología , Trasplante de Piel , Trasplante Heterólogo , Animales , Animales Modificados Genéticamente , Galactosiltransferasas , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Modelos Animales , Estudios Prospectivos , Porcinos , Porcinos Enanos , Linfocitos T/inmunología
17.
Biologicals ; 67: 21-28, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32828642

RESUMEN

Antibodies have strong affinity to their target molecules, a characteristic that is utilized in antibody drugs. For antibody drugs, target molecule specificity and long duration pharmacokinetics, along with strong affinity to the target molecule are important characteristics. Plasminogen activator inhibitor-1 (PAI-1) is one of the key regulators of the fibrinolysis system, and the benefits of PAI-1 activity inhibition have been widely reported for multiple thrombosis and fibrosis-related diseases. Here, we generated a novel antibody, AS3288802, with high selectivity for active PAI-1. AS3288802 exhibited prolonged and strong inhibition of PAI-1 activity in cynomolgus monkey blood in vivo. Given that AS3288802 showed prolonged antigen inhibition activity due to its high target molecule selectivity, we propose that increasing target molecule selectivity may be a key strategy for lengthening the efficacy duration of antibody drugs. AS3288802 may be a promising anti-PAI-1 antibody drug with multiple clinical applications including thrombosis and fibrosis-related diseases.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos/inmunología , Macaca fascicularis/inmunología , Inhibidor 1 de Activador Plasminogénico/inmunología , Animales , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/farmacología , Humanos , Macaca fascicularis/metabolismo , Masculino , Ratones , Inhibidor 1 de Activador Plasminogénico/metabolismo , Trombosis/inmunología , Trombosis/prevención & control
18.
Signal Transduct Target Ther ; 5(1): 157, 2020 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-32814760

RESUMEN

Identification of a suitable nonhuman primate (NHP) model of COVID-19 remains challenging. Here, we characterized severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in three NHP species: Old World monkeys Macaca mulatta (M. mulatta) and Macaca fascicularis (M. fascicularis) and New World monkey Callithrix jacchus (C. jacchus). Infected M. mulatta and M. fascicularis showed abnormal chest radiographs, an increased body temperature and a decreased body weight. Viral genomes were detected in swab and blood samples from all animals. Viral load was detected in the pulmonary tissues of M. mulatta and M. fascicularis but not C. jacchus. Furthermore, among the three animal species, M. mulatta showed the strongest response to SARS-CoV-2, including increased inflammatory cytokine expression and pathological changes in the pulmonary tissues. Collectively, these data revealed the different susceptibilities of Old World and New World monkeys to SARS-CoV-2 and identified M. mulatta as the most suitable for modeling COVID-19.


Asunto(s)
Betacoronavirus/patogenicidad , Callithrix/virología , Infecciones por Coronavirus/epidemiología , Modelos Animales de Enfermedad , Macaca fascicularis/virología , Macaca mulatta/virología , Pandemias , Neumonía Viral/epidemiología , Animales , Anticuerpos Antivirales/biosíntesis , Betacoronavirus/inmunología , Temperatura Corporal , Peso Corporal , COVID-19 , Callithrix/inmunología , Infecciones por Coronavirus/diagnóstico por imagen , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/patología , Citocinas/biosíntesis , Citocinas/clasificación , Citocinas/inmunología , Susceptibilidad a Enfermedades , Femenino , Humanos , Pulmón/diagnóstico por imagen , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Macaca fascicularis/inmunología , Macaca mulatta/inmunología , Masculino , Neumonía Viral/diagnóstico por imagen , Neumonía Viral/inmunología , Neumonía Viral/patología , SARS-CoV-2 , Especificidad de la Especie , Tomografía Computarizada por Rayos X , Carga Viral , Replicación Viral
19.
Am J Pathol ; 190(10): 2095-2110, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32598882

RESUMEN

Inhalational anthrax, a disease caused by inhaling Bacillus anthracis spores, leads to respiratory distress, vascular leakage, high-level bacteremia, and often death within days. Anthrax lethal toxin and edema toxin, which are composed of protective antigen (PA) plus either lethal factor (LF) or edema factor (EF), respectively, play an important yet incompletely defined role in the pulmonary pathophysiology. To better understand their contribution, we examined the structural integrity of the alveolar-capillary barrier in archival formalin-fixed lungs of cynomolgus monkeys challenged with the fully virulent B. anthracis Ames wild-type strain or the isogenic toxin-deficient mutants ΔEF, ΔLF, and ΔPA. Pulmonary spore challenge with the wild-type strain caused high mortality, intra-alveolar hemorrhages, extensive alveolar septal sequestration of bacteria and neutrophils, diffuse destabilization of epithelial and endothelial junctions, increased markers of coagulation and complement activation (including tissue factor and C5a), and multifocal intra-alveolar fibrin deposition. ΔEF challenge was lethal and showed similar alveolar-capillary alterations; however, intra-alveolar hemorrhages, bacterial deposition, and markers of coagulation or complement were absent or markedly lower. In contrast, ΔLF or ΔPA challenges were nonlethal and showed no signs of alveolar bacterial deposition or alveolar-capillary changes. These findings provide evidence that lethal toxin plays a determinative role in bacterial dissemination and alveolar-capillary barrier dysfunction, and edema toxin may significantly exacerbate pulmonary pathologies in a systemic infection.


Asunto(s)
Carbunco/patología , Bacillus anthracis/patogenicidad , Bacteriemia/patología , Pulmón/patología , Infecciones del Sistema Respiratorio/patología , Animales , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/farmacología , Toxinas Bacterianas/farmacología , Pulmón/efectos de los fármacos , Macaca fascicularis/inmunología , Neutrófilos/inmunología , Esporas Bacterianas/inmunología , Esporas Bacterianas/patogenicidad , Virulencia/inmunología
20.
Birth Defects Res ; 112(1): 105-117, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31746560

RESUMEN

Antibody-like biopharmaceuticals cross the placenta by utilizing existing transport pathways (e.g., FcRn receptor). There are limited data evaluating this transfer during organogenesis in any species. Understanding placental transfer of antibody-like biopharmaceuticals can help to predict risk of developmental toxicity across species, including humans. To complement previously published placental transfer data in the rat with humanized IgGΔ2 (hIgG2), the timing and magnitude of transfer in the cynomolgus monkey and embryo/fetal biodistribution of maternally administered 125 I-radiolabeled hIgG2 was quantified on gestation days (GD) 35, 40, 50, 70, and 140 using gamma counting and whole body autoradiography 24 hr following intravenous injection. Chorioallantoic placental tissues were collected at all time points for Western Blot analysis with anti-FcRn antibody. Maternally administered 125 I-hIgG2 was found in embryo/fetal tissues at all time points, including organogenesis. Embryo/fetal plasma 125 I-hIgG2 concentration increased during gestation, but only slightly up to GD 70 in embryo/fetal tissues, with hIgG2 tissue concentrations generally similar between GD70 and 140. The embryo/fetal:maternal 125 I-hIgG2 plasma concentration ratio was approximately 2.3 fold higher on GD 140, in comparison to ratios on GD 40. Importantly, placental FcRn protein expression was confirmed at all timepoints. These data demonstrate placental transfer of hIgG2 in a nonhuman primate model, and at levels comparable to the rat model, raising the potential for adverse developmental outcomes by direct antibody binding to biological targets.


Asunto(s)
Inmunoglobulina G/inmunología , Intercambio Materno-Fetal/inmunología , Placenta/inmunología , Animales , Anticuerpos Monoclonales/metabolismo , Embrión de Mamíferos/metabolismo , Femenino , Feto/metabolismo , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/metabolismo , Radioisótopos de Yodo/metabolismo , Macaca fascicularis/inmunología , Macaca fascicularis/metabolismo , Intercambio Materno-Fetal/fisiología , Organogénesis/inmunología , Organogénesis/fisiología , Embarazo , Distribución Tisular/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...