Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.064
Filtrar
1.
Dokl Biochem Biophys ; 516(1): 53-57, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38700816

RESUMEN

Study of CD4+ T cell response and T cell receptor (TCR) specificity is crucial for understanding etiology of immune-mediated diseases and developing targeted therapies. However, solubility, accessibility, and stability of synthetic antigenic peptides used in T cell assays may be a critical point in such studies. Here we present a T cell activation reporter system using recombinant proteins containing antigenic epitopes fused with bacterial thioredoxin (trx-peptides) and obtained by bacterial expression. We report that co-incubation of CD4+ HA1.7 TCR+ reporter Jurkat 76 TRP cells with CD80+ HLA-DRB1*01:01+ HeLa cells or CD4+ Ob.1A12 TCR+ Jurkat 76 TRP with CD80+ HLA-DRB1*15:01+ HeLa cells resulted in activation of reporter Jurkat 76 TPR after addition of recombinant trx-peptide fusion proteins, containing TCR-specific epitopes. Trx-peptides were comparable with corresponding synthetic peptides in their capacity to activate Jurkat 76 TPR. These data demonstrate that thioredoxin as a carrier protein (trx) for antigenic peptides exhibits minimal interference with recognition of MHC-specific peptides by TCRs and consequent T cell activation. Our findings highlight potential feasibility of trx-peptides as a reagent for assessing the immunogenicity of antigenic fragments.


Asunto(s)
Linfocitos T CD4-Positivos , Péptidos , Receptores de Antígenos de Linfocitos T , Proteínas Recombinantes de Fusión , Tiorredoxinas , Humanos , Tiorredoxinas/inmunología , Tiorredoxinas/genética , Células Jurkat , Linfocitos T CD4-Positivos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Péptidos/farmacología , Péptidos/inmunología , Péptidos/química , Activación de Linfocitos/efectos de los fármacos , Células HeLa
2.
Vaccine ; 42(15): 3474-3485, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38641492

RESUMEN

Respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) vaccines have been long overdue. Structure-based vaccine design created a new momentum in the last decade, and the first RSV vaccines have finally been approved in older adults and pregnant individuals. These vaccines are based on recombinant stabilized pre-fusion F glycoproteins administered as soluble proteins. Multimeric antigenic display could markedly improve immunogenicity and should be evaluated in the next generations of vaccines. Here we tested a new virus like particles-based vaccine platform which utilizes the direct fusion of an immunogen of interest to the structural human immunodeficient virus (HIV) protein Gag to increase its surface density and immunogenicity. We compared, in mice, the immunogenicity of RSV-F or hMPV-F based immunogens delivered either as soluble proteins or displayed on the surface of our VLPs. VLP associated F-proteins showed better immunogenicity and induced superior neutralizing responses. Moreover, when combining both VLP associated and soluble immunogens in a heterologous regimen, VLP-associated immunogens provided added benefits when administered as the prime immunization.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Metapneumovirus , Ratones Endogámicos BALB C , Vacunas de Partículas Similares a Virus , Proteínas Virales de Fusión , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/sangre , Ratones , Metapneumovirus/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Vacunas de Partículas Similares a Virus/administración & dosificación , Femenino , Proteínas Virales de Fusión/inmunología , Proteínas Virales de Fusión/genética , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Virus Sincitial Respiratorio Humano/inmunología , Inmunogenicidad Vacunal , Humanos , Vacunas contra Virus Sincitial Respiratorio/inmunología , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/genética , Infecciones por Virus Sincitial Respiratorio/prevención & control , Infecciones por Virus Sincitial Respiratorio/inmunología , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación
3.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167159, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38583815

RESUMEN

Chimeric antigen receptor T (CAR-T) cell therapy is regarded as a potent immunotherapy and has made significant success in hematologic malignancies by eliciting antigen-specific immune responses. However, response rates of CAR-T cell therapy against solid tumors with immunosuppressive microenvironments remain limited. Co-engineering strategies are advancing methods to overcome immunosuppressive barriers and enhance antitumor responses. Here, we engineered an IL-2 mutein co-engineered CAR-T for the improvement of CAR-T cells against solid tumors and the efficient inhibition of solid tumors. We equipped the CAR-T cells with co-expressing both tumor antigen-targeted CAR and a mutated human interleukin-2 (IL-2m), conferring enhanced CAR-T cells fitness in vitro, reshaped immune-excluded TME, enhanced CAR-T infiltration in solid tumors, and improved tumor control without significant systemic toxicity. Overall, this subject demonstrates the universal CAR-T cells armed strategy for the development and optimization of CAR-T cells against solid tumors.


Asunto(s)
Inmunoterapia Adoptiva , Interleucina-2 , Neoplasias , Receptores Quiméricos de Antígenos , Linfocitos T , Humanos , Interleucina-2/genética , Interleucina-2/inmunología , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/genética , Inmunoterapia Adoptiva/métodos , Animales , Neoplasias/terapia , Neoplasias/inmunología , Neoplasias/genética , Ratones , Linfocitos T/inmunología , Linfocitos T/metabolismo , Microambiente Tumoral/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/genética , Línea Celular Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Cancer Res ; 84(10): 1550-1559, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38381555

RESUMEN

Lipid nanoparticle (LNP)-encapsulated mRNA has been used for in vivo production of several secreted protein classes, such as IgG, and has enabled the development of personalized vaccines in oncology. Establishing the feasibility of delivering complex multispecific modalities that require higher-order structures important for their function could help expand the use of mRNA/LNP biologic formulations. Here, we evaluated whether in vivo administration of mRNA/LNP formulations of SIRPα-Fc-CD40L and TIGIT-Fc-LIGHT could achieve oligomerization and extend exposure, on-target activity, and antitumor responses comparable with that of the corresponding recombinant fusion proteins. Intravenous infusion of the formulated LNP-encapsulated mRNAs led to rapid and sustained production of functional hexameric proteins in vivo, which increased the overall exposure relative to the recombinant protein controls by ∼28 to 140 fold over 96 hours. High concentrations of the mRNA-encoded proteins were also observed in secondary lymphoid organs and within implanted tumors, with protein concentrations in tumors up to 134-fold greater than with the recombinant protein controls 24 hours after treatment. In addition, SIRPα-Fc-CD40L and TIGIT-Fc-LIGHT mRNAs induced a greater increase in antigen-specific CD8+ T cells in the tumors. These mRNA/LNP formulations were well tolerated and led to a rapid increase in serum and intratumoral IL2, delayed tumor growth, extended survival, and outperformed the activities of benchmark mAb controls. Furthermore, the mRNA/LNPs demonstrated improved efficacy in combination with anti-PD-L1 relative to the recombinant fusion proteins. These data support the delivery of complex oligomeric biologics as mRNA/LNP formulations, where high therapeutic expression and exposure could translate into improved patient outcomes. SIGNIFICANCE: Lipid nanoparticle-encapsulated mRNA can efficiently encode complex fusion proteins encompassing immune checkpoint blockers and costimulators that functionally oligomerize in vivo with extended pharmacokinetics and durable exposure to induce potent antitumor immunity.


Asunto(s)
Nanopartículas , ARN Mensajero , Proteínas Recombinantes de Fusión , Animales , Ratones , ARN Mensajero/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/inmunología , Nanopartículas/química , Humanos , Femenino , Ratones Endogámicos C57BL , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/genética , Lípidos/química , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Línea Celular Tumoral
5.
Clin Cancer Res ; 30(10): 2025-2038, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38190116

RESUMEN

Cytokines are key mediators of immune responses that can modulate the antitumor activity of immune cells. Cytokines have been explored as a promising cancer immunotherapy. However, there are several challenges to cytokine therapy, especially a lack of tumor targeting, resulting in high toxicity and limited efficacy. To overcome these limitations, novel approaches have been developed to engineer cytokines with improved properties, such as chimeric cytokines. Chimeric cytokines are fusion proteins that combine different cytokine domains or link cytokines to antibodies (immunocytokines) or other molecules that can target specific receptors or cells. Chimeric cytokines can enhance the selectivity and stability of cytokines, leading to reduced toxicity and improved efficacy. In this review, we focus on two promising cytokines, IL2 and IL15, and summarize the current advances and challenges of chimeric cytokine design and application for cancer immunotherapy. Most of the current approaches focus on increasing the potency of cytokines, but another important goal is to reduce toxicity. Cytokine engineering is promising for cancer immunotherapy as it can enhance tumor targeting while minimizing adverse effects.


Asunto(s)
Citocinas , Inmunoterapia , Neoplasias , Proteínas Recombinantes de Fusión , Humanos , Neoplasias/terapia , Neoplasias/inmunología , Neoplasias/tratamiento farmacológico , Inmunoterapia/métodos , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico , Citocinas/metabolismo , Animales , Interleucina-2/uso terapéutico , Interleucina-2/inmunología , Interleucina-2/efectos adversos
6.
J Interferon Cytokine Res ; 42(7): 316-328, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35834651

RESUMEN

Cytokines are powerful mediators of immune responses and some, such as interleukin-2 (IL-2), have achieved dramatic responses as cancer immunotherapies. Unfortunately, systemic administration often results in deleterious side effects, prompting exploration of strategies to localize cytokine activity to the tumor microenvironment (TME). To this end, we constructed an IL-2/IL2Ra fusion protein (IL-2FP) with an MMP2/9-specific cleavage site, designed to exploit the dysregulated protease activity in the TME to selectively activate IL-2 in the tumor. To determine if TME protease activity is sufficient to cleave the FP and if FP activity is due to specific cleavage, we created Colon 38 tumor cell lines expressing similar levels of IL-2FPs with either a functional cleavage site [H11(cs-1FP)] or a scrambled, noncleavable sequence [H2(scramFP)]. H11(cs-1FP) tumors demonstrated reduced tumor growth, characterized by regressions not observed in H2(scramFP) tumors. Analysis through qRT-PCR, flow cytometry, and immunohistochemistry indicate robust CD8 responses in the H11(cs-1FP) tumors. Interferon gamma (IFNg) knockout mice revealed that the immune effects of the cleavable FP are mediated through both IFNg-dependent and IFNg-independent mechanisms. Collectively, these data suggest that matrix metalloproteinases (MMPs) in the TME can cleave the IL-2FP specifically, thus enhancing an antitumor response, and provide a rationale for further developing this approach.


Asunto(s)
Línea Celular Tumoral , Inmunidad , Interferón gamma , Interleucina-2 , Proteínas Recombinantes de Fusión , Microambiente Tumoral , Animales , Línea Celular Tumoral/inmunología , Inmunidad/inmunología , Interferón gamma/inmunología , Interleucina-2/inmunología , Interleucina-2/farmacología , Ratones , Péptido Hidrolasas/inmunología , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacología , Microambiente Tumoral/inmunología
7.
Nature ; 607(7918): 360-365, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35676488

RESUMEN

Synthetic receptor signalling has the potential to endow adoptively transferred T cells with new functions that overcome major barriers in the treatment of solid tumours, including the need for conditioning chemotherapy1,2. Here we designed chimeric receptors that have an orthogonal IL-2 receptor extracellular domain (ECD) fused with the intracellular domain (ICD) of receptors for common γ-chain (γc) cytokines IL-4, IL-7, IL-9 and IL-21 such that the orthogonal IL-2 cytokine elicits the corresponding γc cytokine signal. Of these, T cells that signal through the chimeric orthogonal IL-2Rß-ECD-IL-9R-ICD (o9R) are distinguished by the concomitant activation of STAT1, STAT3 and STAT5 and assume characteristics of stem cell memory and effector T cells. Compared to o2R T cells, o9R T cells have superior anti-tumour efficacy in two recalcitrant syngeneic mouse solid tumour models of melanoma and pancreatic cancer and are effective even in the absence of conditioning lymphodepletion. Therefore, by repurposing IL-9R signalling using a chimeric orthogonal cytokine receptor, T cells gain new functions, and this results in improved anti-tumour activity for hard-to-treat solid tumours.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Inmunoterapia Adoptiva , Subunidad gamma Común de Receptores de Interleucina , Neoplasias , Receptores de Interleucina-9 , Proteínas Recombinantes de Fusión , Linfocitos T , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Inmunoterapia Adoptiva/métodos , Subunidad gamma Común de Receptores de Interleucina/genética , Subunidad gamma Común de Receptores de Interleucina/inmunología , Interleucinas/genética , Interleucinas/inmunología , Melanoma/inmunología , Ratones , Neoplasias/genética , Neoplasias/inmunología , Neoplasias Pancreáticas/inmunología , Receptores de Interleucina-9/genética , Receptores de Interleucina-9/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Factores de Transcripción STAT/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
8.
Front Immunol ; 13: 767530, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35154097

RESUMEN

Natural killer (NK) cells have been studied extensively in humans and mice for their vital role in the vertebrate innate immune system. They are known to rapidly eliminate tumors or virus infected cells in an immune response utilizing their lytic properties. The natural cytotoxicity receptors (NCRs) NKp30 (NCR3), NKp44 (NCR2), and NKp46 (NCR1) are important mediators of NK-cell cytotoxicity. NKp44 expression was reported for NK cells in humans as well as in some non-human primates and found exclusively on activated NK cells. Previously, no information was available on NKp44 protein expression and its role in porcine lymphocytes due to the lack of species-specific monoclonal antibodies (mAbs). For this study, porcine-specific anti-NKp44 mAbs were generated and their reactivity was tested on blood and tissue derived NK cells in pigs of different age classes. Interestingly, NKp44 expression was detected ex vivo already on resting NK cells; moreover, the frequency of NKp44+ NK cells was higher than that of NKp46+ NK cells in most animals analyzed. Upon in vitro stimulation with IL-2 or IL-15, the frequency of NKp44+ NK cells, as well as the intensity of NKp44 expression at the single cell level, were increased. Since little is known about swine NK cells, the generation of a mAb (clone 54-1) against NKp44 will greatly aid in elucidating the mechanisms underlying the differentiation, functionality, and activation of porcine NK cells.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Citotoxicidad Inmunológica , Células Asesinas Naturales/inmunología , Receptor 2 Gatillante de la Citotoxidad Natural/inmunología , Receptor 2 Gatillante de la Citotoxidad Natural/metabolismo , Adolescente , Adulto , Animales , Anticuerpos Monoclonales/sangre , Donantes de Sangre , Células Cultivadas , Femenino , Humanos , Inmunización/métodos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Interleucina-4/administración & dosificación , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/inmunología , Porcinos , Adulto Joven
9.
Comput Math Methods Med ; 2022: 6244175, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35222687

RESUMEN

BACKGROUND: Researches have confirmed that the abnormal signals of OX40 and PD-1 lead to the changes of T cell biological behavior, thus participating the immunopathological process of RA. However, the pathogenesis of RA immunopathological process has not been clarified yet. METHODS: 30 DBA/1 mice were randomly divided into 5 groups (6 mice per group): control group, collagen-induced arthritis (CIA) group, PD-1-Fc/CIA group, OX40-Fc/CIA group, and PD-1-Fc + OX40-Fc/CIA group. The pathological changes in mice joints were observed by H&E staining. The proportion of CD4+ T, CD8+ T, CD28+, and CD19+ cells in peripheral blood mononuclear cells (PBMCs) was detected by flow cytometry. Serum inflammatory factors (CRP, IL-2, IL-4, IL-1ß, INF-γ) and bone metabolism-related genes (CTX-I, TRACP-5b, BALP) were detected by ELISA assay. Western blotting was applied to measure the NF-κB signaling pathway-related protein (p-IKKß, p-IκBα, p50) expression in synovial tissue of mice joint. RESULTS: Compared with the control group, CIA mice showed significant increases in arthritis score and pathological score. In the CIA group, a marked decrease was identified in the proportion of CD8+ T, CD19+, and CD68+ cells. Additionally, the CIA group was associated with upregulation of secretion of inflammatory factors in serum and expression of bone metabolism-related genes and NF-κB pathway-related proteins. Compared with the CIA group, the same indexes above showed a further aggravation in the PD-1-Fc group while all indexes improved in the OX40-Fc group. Besides, OX40-Fc fusion protein slowed down significantly the further deterioration of CIA mouse pathological process caused by PD-1-Fc fusion protein. CONCLUSION: OX40-Fc fusion protein alleviates PD-1-Fc-aggravated RA by inhibiting inflammatory response. This research provides biological markers with clinical significance for diagnosis and prognosis of RA, as well as offers theoretical and experimental foundation to the new targets for immune intervention.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptores OX40/uso terapéutico , Animales , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/patología , Artritis Experimental/fisiopatología , Artritis Reumatoide/patología , Artritis Reumatoide/fisiopatología , Biomarcadores/metabolismo , Huesos/efectos de los fármacos , Huesos/metabolismo , Colágeno Tipo I/metabolismo , Biología Computacional , Humanos , Fragmentos Fc de Inmunoglobulinas/inmunología , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Inflamación/tratamiento farmacológico , Inflamación/patología , Inflamación/fisiopatología , Masculino , Ratones , Ratones Endogámicos DBA , FN-kappa B/metabolismo , Péptidos/metabolismo , Receptor de Muerte Celular Programada 1/inmunología , Receptores OX40/inmunología , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico , Transducción de Señal/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Fosfatasa Ácida Tartratorresistente/metabolismo
10.
MAbs ; 14(1): 2004982, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34978527

RESUMEN

Antibodies facilitate targeted cell killing by engaging with immune cells such as natural killer cells through weak binding interactions with Fcγ receptors on the cell surface. Here, we evaluate the binding affinity of the receptor FcγRIIIa V158 (CD16a) for several therapeutic antibody classes, isoforms, and Fc-fusion proteins using an immobilized receptor affinity liquid chromatography (LC) approach coupled with online mass spectrometry (MS) detection. Aglycosylated FcγRIIIa was used in the affinity chromatography and compared with published affinities using glycosylated receptors. Affinity LC-MS differentiated the IgG1 antibodies primarily according to their Fc glycosylation patterns, with highly galactosylated species having greater affinity for the immobilized receptors and thus eluting later from the column (M5< G0F < G0 afucosylated ≅ G1F < G2F). Sialylated species bound weaker to their asialylated counterparts as reported previously. High mannose glycoforms bound weaker than G0F, contrary to previously published studies using glycosylated receptors. Also, increased receptor binding affinity associated with afucosylated antibodies was not observed with the aglycosylated FcγRIIIa. This apparent difference from previous findings highlighted the importance of the glycans on the receptors for mediating stronger binding interactions. Characterization of temperature-stressed samples by LC-MS peptide mapping revealed over 200 chemical and post-translational modifications, but only the Fc glycans, deamidation of EU N325, and an unknown modification to either proline or cysteine residues of the hinge region were found to have a statistically significant impact on binding.Abbreviations: Antibody-dependent cell-mediated cytotoxicity (ADCC), chimeric antigen receptor (CAR), Chinese hamster ovary (CHO), dithiothreitol (DTT), electrospray ionization (ESI), hydrogen-deuterium exchange (HDX), filter aided-sample preparation (FASP), Fcγ receptor (FcγR), fragment crystallizable (Fc), high-pressure liquid chromatography (HPLC), immunoglobulin G (IgG), liquid chromatography (LC), monoclonal antibody (mAb), mass spectrometry (MS), natural killer (NK), N-glycolylneuraminic acid (NGNA), N-acetylneuraminic acid (NANA), principal component analysis (PCA), surface plasmon resonance (SPR), trifluoroacetic acid (TFA), and extracted mass chromatogram (XMC).


Asunto(s)
Cromatografía de Afinidad , Fragmentos Fc de Inmunoglobulinas/química , Espectrometría de Masas , Receptores de IgG/química , Proteínas Recombinantes de Fusión/química , Animales , Células CHO , Cricetulus , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/inmunología , Receptores de IgG/genética , Receptores de IgG/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología
11.
Nat Commun ; 13(1): 109, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013154

RESUMEN

Direct injection of therapies into tumors has emerged as an administration route capable of achieving high local drug exposure and strong anti-tumor response. A diverse array of immune agonists ranging in size and target are under development as local immunotherapies. However, due to the relatively recent adoption of intratumoral administration, the pharmacokinetics of locally-injected biologics remains poorly defined, limiting rational design of tumor-localized immunotherapies. Here we define a pharmacokinetic framework for biologics injected intratumorally that can predict tumor exposure and effectiveness. We find empirically and computationally that extending the tumor exposure of locally-injected interleukin-2 by increasing molecular size and/or improving matrix-targeting affinity improves therapeutic efficacy in mice. By tracking the distribution of intratumorally-injected proteins using positron emission tomography, we observe size-dependent enhancement in tumor exposure occurs by slowing the rate of diffusive escape from the tumor and by increasing partitioning to an apparent viscous region of the tumor. In elucidating how molecular weight and matrix binding interplay to determine tumor exposure, our model can aid in the design of intratumoral therapies to exert maximal therapeutic effect.


Asunto(s)
Colágeno/genética , Inmunoterapia/métodos , Interleucina-2/farmacología , Melanoma Experimental/terapia , Receptores Inmunológicos/genética , Neoplasias Cutáneas/terapia , Aloinjertos , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Línea Celular Tumoral , Colágeno/inmunología , Femenino , Biblioteca de Genes , Inyecciones Intralesiones , Interleucina-2/genética , Interleucina-2/inmunología , Interleucina-2/farmacocinética , Melanoma Experimental/diagnóstico por imagen , Melanoma Experimental/genética , Melanoma Experimental/mortalidad , Ratones , Ratones Endogámicos C57BL , Péptidos/genética , Péptidos/inmunología , Tomografía de Emisión de Positrones , Unión Proteica , Ingeniería de Proteínas/métodos , Receptores Inmunológicos/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Albúmina Sérica/genética , Albúmina Sérica/inmunología , Neoplasias Cutáneas/diagnóstico por imagen , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/mortalidad , Análisis de Supervivencia , Carga Tumoral/efectos de los fármacos
12.
Protein Expr Purif ; 189: 105973, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34560256

RESUMEN

The novel anti-PD-L1/TGFBR2-ECD fusion protein (BR102) comprises an anti-PD-L1 antibody (HS636) which is fused at the C terminus of the heavy chain to a TGF-ß1 receptor Ⅱ ectodomain (TGFBR2-ECD), and which can sequester the PD-1/PD-L1 pathway and TGF-ß bioactivity in the immunosuppressive tumor microenvironment. In the expression of TGFBR2-ECD wild-type fused protein (BR102-WT), a 50 kDa clipped species was confirmed to be induced by proteolytic cleavage at a "QKS" site located in the N-terminus of the ectodomain, which resulted in the formation of IgG-like clipping. The matrix metalloproteinase-9 was determined to be associated with BR102-WT digestion. In addition, it was observed that the N-glycosylation modifications of the fusion protein were tightly involved in regulating proteolytic activity and the levels of cleavage could be significantly suppressed by MMP-inhibitors. To avoid proteolytic degradation, eliminating protease-sensitive amino acid motifs and introducing potential glycosylation were performed. Three sensitive motifs were mutated, and the levels of clipping were strongly restrained. The mutant candidates exhibited similar binding affinities to hPD-L1 and hTGF-ß1 as well as highly purified BR102-WT2. Furthermore, the mutants displayed more significant proteolytic resistance than that of BR102-WT2 in the lysate incubation reaction and the plasma stability test. Moreover, the bifunctional candidate Mu3 showed an additive antitumor effect in MC38/hPD-L1 bearing models as compared to that of with anti-PD-L1 antibody alone. In conclusion, in this study, the protease-sensitive features of BR102-WT were well characterized and efficient optimization was performed. The candidate BR102-Mutants exhibited advanced druggability in drug stability and displayed desirable antitumor activity.


Asunto(s)
Anticuerpos Antineoplásicos/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Neoplasias del Colon/terapia , Procesamiento Proteico-Postraduccional , Receptor Tipo II de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/genética , Animales , Anticuerpos Antineoplásicos/genética , Anticuerpos Antineoplásicos/inmunología , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Células CHO , Neoplasias del Colon/genética , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Cricetulus , Femenino , Glicosilación , Humanos , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/inmunología , Ratones , Ratones Endogámicos C57BL , Mutación , Dominios Proteicos , Proteolisis , Receptor Tipo II de Factor de Crecimiento Transformador beta/genética , Receptor Tipo II de Factor de Crecimiento Transformador beta/inmunología , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacología , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/inmunología , Microambiente Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Life Sci ; 289: 120222, 2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-34902436

RESUMEN

In order to overcome limitations of conventional cancer therapy methods, immunotoxins with the capability of target-specific action have been designed and evaluated pre-clinically, and some of them are in clinical studies. Targeting cancer cells via antibodies specific for tumour-associated surface proteins is a new biomedical approach that could provide the selectivity that is lacking in conventional cancer therapy methods such as radiotherapy and chemotherapy. A successful example of an approved immunotoxin is represented by immunoRNases. ImmunoRNases are fusion proteins in which the toxin has been replaced by a ribonuclease. Conjugation of RNase molecule to monoclonal antibody or antibody fragment was shown to enhance specific cell-killing by several orders of magnitude, both in vitro and in animal models. There are several RNases obtained from different mammalian cells that are expected to be less immunogenic and systemically toxic. In fact, RNases are pro-toxins which become toxic only upon their internalization in target cells mediated by the antibody moiety. The structure and large size of the antibody molecules assembled with the immunoRNases have always been a challenge in the application of immunoRNases as an antitoxin. To overcome this obstacle, we have offered a new strategy for the application of immunoRNases as a promising approach for upgrading immunoRNAses with maximum affinity and high stability in the cell, which can ultimately act as an effective large-scale cancer treatment. In this review, we introduce the optimized antibody-like molecules with small size, approximately 10 kD, which are presumed to significantly enhance RNase activity and be a suitable agent with the potential for anti-cancer functionality. In addition, we also discuss new molecular entities such as monobody, anticalin, nonobody and affilin as refined versions in the development of immunoRNases. These small molecules express their functionality with the suitable small size as well as with low immunogenicity in the cell, as a part of immunoRNases.


Asunto(s)
Antineoplásicos Inmunológicos , Antineoplásicos , Inmunotoxinas , Neoplasias , Proteínas Recombinantes de Fusión , Ribonucleasas , Animales , Antineoplásicos/inmunología , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/farmacocinética , Antineoplásicos Inmunológicos/farmacología , Humanos , Inmunotoxinas/genética , Inmunotoxinas/inmunología , Inmunotoxinas/farmacocinética , Inmunotoxinas/farmacología , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/farmacología , Ribonucleasas/genética , Ribonucleasas/inmunología , Ribonucleasas/farmacocinética , Ribonucleasas/farmacología
14.
Infect Genet Evol ; 97: 105188, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34920098

RESUMEN

The best and most effective way to combat pandemics is to use effective vaccines and live attenuated vaccines are among the most effective vaccines. However, one of the major problems is the length of time it takes to get the attenuated vaccines. Today, the CRISPR toolkit (Clustered Regularly Inerspaced Short Palindromic Repeats) has made it possible to make changes with high efficiency and speed. Using this toolkit to make point mutations on the RNA virus's genome in a coculture of permissive and nonpermissive cells and under controlled conditions can accelerate changes in the genome and accelerate natural selection to obtain live attenuated vaccines.


Asunto(s)
Vacunas contra la COVID-19/genética , COVID-19/prevención & control , Sistemas CRISPR-Cas , Edición Génica/métodos , Tasa de Mutación , SARS-CoV-2/genética , Proteínas Virales/genética , Desaminasas APOBEC/genética , Desaminasas APOBEC/inmunología , Adenosina Desaminasa/genética , Adenosina Desaminasa/inmunología , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , COVID-19/inmunología , Vacunas contra la COVID-19/biosíntesis , Endonucleasas/genética , Endonucleasas/inmunología , Expresión Génica , Genoma Viral , Humanos , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , SARS-CoV-2/inmunología , Selección Genética , Vacunas Atenuadas , Proteínas Virales/inmunología
15.
Protein Expr Purif ; 189: 105966, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34627999

RESUMEN

Nerve growth factor (NGF) is produced and released in injured tissues or chronic pain tissues caused by other diseases. Studies have shown that monoclonal antibodies targeting NGF have a good efficacy in the treatment of osteoarthritis (OA), low back pain and chronic pain, which may be a promising therapy. In this study, DNA sequences of NGF-his and NGF-hFc were synthesized using eukaryotic expression system and subcloned into pTT5 expression vector. After that, NGF proteins were expressed by transient expression in HEK293E cells. We immunized mice with NGF-hFc protein and fused mouse spleen cells to prepare hybridomas. NGF-His protein was used to screen out the hybridoma supernatant that could directly bind to NGF. Antibodies were purified from hybridioma supernatant. Futhermore, via surface plasmon resonance (SPR) screening, six anti-NGF mAbs were screened to block the binding of NGF and TrkA receptor in the treatment of chronic pain. Among them, 58F10G10H showed high affinity (KD = 1.03 × 10-9 M) and even better than that of positive control antibody Tanezumab (KD = 1.53 × 10-9 M). Moreover, the specific reactivity of 58F10G10H was demonstrated by TF-1 cell proliferation activity experiments, competitive binding Enzyme-linked immunosorbent assay (ELISA) and the arthritis animal models in mice, respectively. In conclusion, in this study, a method for the preparation of high-yield NGF-HFC and NGF-His proteins was designed, and a high-affinity monoclonal antibody against NGF with potential for basic research and clinical application was prepared.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Artritis/tratamiento farmacológico , Factor de Crecimiento Nervioso/antagonistas & inhibidores , Dolor/prevención & control , Receptor trkA/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Monoclonales Humanizados/farmacología , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Artritis/genética , Artritis/inmunología , Artritis/patología , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Células HEK293 , Humanos , Hibridomas/química , Hibridomas/inmunología , Inmunización , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/inmunología , Linfocitos/química , Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Factor de Crecimiento Nervioso/genética , Factor de Crecimiento Nervioso/inmunología , Dolor/genética , Dolor/inmunología , Dolor/patología , Receptor trkA/genética , Receptor trkA/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología
16.
Int Immunopharmacol ; 101(Pt B): 108368, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34857479

RESUMEN

Due to the high affinity for binding to target molecules and also other unique attributes, affibodies have a great potential to be used in immunotherapeutic and diagnostic approaches. However, the possibility of undesirable immune response is still a great concern. In the current study, we investigated the possible antigenicity, allergenicity and cytotoxicity of the HER2-targeting affibody ZHER2. The binding affinity of potential epitopes of the affibody to murine major histocompatibility complex (MHC) molecules was investigated by immunoinformatics tools and docking approaches. The possible interaction of ZHER2 with human leukocyte antigens HLA-DP, HLA-DM, HLA-DQ and HLA-DR was also studied by protein-protein docking. Additionally, the synthesized affibody gene was expressed and the protein was purified for boosted immunization of Balb/c mice. Induced secretion of IFN-γ, IL-2, IL-4 and IL-10, and total serum IgG were assessed in the immunized mice. Furthermore, MTT cell viability test was performed to evaluate the cytotoxicity of ZHER2 in splenocytes of the treated mice. In silico analyses showed the possible induction of the immune response by ZHER2. While the affibody could elicit the secretion of cellular immune cytokines, it could not induce a significant humoral response in the treated mice and did not show any cytotoxic effects on the exposed splenocytes. These findings explain the practicability of ZHER2 for therapeutic and in vivo diagnostic usages, though its ubiquitous application may need more studies.


Asunto(s)
Anticuerpos , Imitación Molecular , Receptor ErbB-2/inmunología , Proteínas Recombinantes de Fusión/inmunología , Alérgenos , Animales , Supervivencia Celular , Clonación Molecular , Simulación por Computador , Citocinas/genética , Citocinas/metabolismo , Epítopos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Conformación Proteica , Dominios Proteicos
17.
Viruses ; 13(12)2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34960713

RESUMEN

Congenital tremor (CT) type A-II in piglets is caused by an emerging atypical porcine pestivirus (APPV), which is prevalent in swine herds and a serious threat to the pig production industry. This study aimed to construct APPV E2 subunit vaccines fused with Fc fragments and evaluate their immunogenicity in piglets. Here, APPV E2Fc and E2ΔFc fusion proteins expressed in Drosophila Schneider 2 (S2) cells were demonstrated to form stable dimers in SDS-PAGE and western blotting assays. Functional analysis revealed that aE2Fc and aE2ΔFc fusion proteins could bind to FcγRI on antigen-presenting cells (APCs), with the affinity of aE2Fc to FcγRI being higher than that of aE2ΔFc. Moreover, subunit vaccines based on aE2, aE2Fc, and aE2ΔFc fusion proteins were prepared, and their immunogenicity was evaluated in piglets. The results showed that the Fc fusion proteins emulsified with the ISA 201VG adjuvant elicited stronger humoral and cellular immune responses than the IMS 1313VG adjuvant. These findings suggest that APPV E2 subunit vaccines fused with Fc fragments may be a promising vaccine candidate against APPV.


Asunto(s)
Inmunidad Celular , Inmunidad Humoral , Pestivirus/inmunología , Porcinos/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Línea Celular , Inmunogenicidad Vacunal , Fragmentos Fc de Inmunoglobulinas/inmunología , Fragmentos Fc de Inmunoglobulinas/metabolismo , Activación de Linfocitos , Infecciones por Pestivirus/inmunología , Infecciones por Pestivirus/veterinaria , Multimerización de Proteína , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/virología , Células Th2/inmunología , Vacunas de Subunidad/inmunología , Proteínas Estructurales Virales/química , Proteínas Estructurales Virales/inmunología , Proteínas Estructurales Virales/metabolismo
18.
Front Immunol ; 12: 753371, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34721427

RESUMEN

Many pathogens enter the host via the gut, causing disease in animals and humans. A robust intestinal immune response is necessary to protect the host from these gut pathogens. Despite being best suited for eliciting intestinal immunity, oral vaccination remains a challenge due to the gastrointestinal environment, a poor uptake of vaccine antigens by the intestinal epithelium and the tolerogenic environment pervading the gut. To improve uptake, efforts have focused on targeting antigens towards the gut mucosa. An interesting target is aminopeptidase N (APN), a conserved membrane protein present on small intestinal epithelial cells shown to mediate epithelial transcytosis. Here, we aimed to further optimize this oral vaccination strategy in a large animal model. Porcine APN-specific monoclonal antibodies were generated and the most promising candidate in terms of epithelial transcytosis was selected to generate antibody fusion constructs, comprising a murine IgG1 or porcine IgA backbone and a low immunogenic antigen: the F18-fimbriated E. coli tip adhesin FedF. Upon oral delivery of these recombinant antibodies in piglets, both mucosal and systemic immune responses were elicited. The presence of the FedF antigen however appeared to reduce these immune responses. Further analysis showed that F18 fimbriae were able to disrupt the antigen presenting capacity of intestinal antigen presenting cells, implying potential tolerogenic effects of FedF. Altogether, these findings show that targeted delivery of molecules to epithelial aminopeptidase N results in their transcytosis and delivery to the gut immune systems. The results provide a solid foundation for the development of oral subunit vaccines to protect against gut pathogens.


Asunto(s)
Adhesinas Bacterianas/inmunología , Anticuerpos Monoclonales/inmunología , Antígenos Bacterianos/inmunología , Antígenos CD13/inmunología , Proteínas de Escherichia coli/inmunología , Inmunoconjugados/inmunología , Inmunoglobulina A/biosíntesis , Mucosa Intestinal/inmunología , Intestino Delgado/inmunología , Porcinos/inmunología , Transcitosis , Vacunas Sintéticas/inmunología , Adhesinas Bacterianas/administración & dosificación , Administración Oral , Animales , Anticuerpos Antibacterianos/biosíntesis , Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales/administración & dosificación , Afinidad de Anticuerpos , Células Presentadoras de Antígenos/inmunología , Antígenos Bacterianos/administración & dosificación , Antígenos CD13/fisiología , Escherichia coli Enterotoxigénica/inmunología , Células Epiteliales/metabolismo , Proteínas de Escherichia coli/administración & dosificación , Femenino , Fimbrias Bacterianas/inmunología , Inmunoconjugados/administración & dosificación , Inmunoglobulina A/administración & dosificación , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Intestino Delgado/enzimología , Ratones , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/inmunología , Transcitosis/fisiología , Vacunación/veterinaria
19.
Sci Rep ; 11(1): 21856, 2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34750487

RESUMEN

Hepatitis C is a major threat to public health for which an effective treatment is available, but a prophylactic vaccine is still needed to control this disease. We designed a vaccine based on chimeric HBV-HCV envelope proteins forming subviral particles (SVPs) that induce neutralizing antibodies against HCV in vitro. Here, we aimed to increase the neutralizing potential of those antibodies, by using HBV-HCV SVPs bearing apolipoprotein E (apoE). These particles were produced by cultured stable mammalian cell clones, purified and characterized. We found that apoE was able to interact with both chimeric HBV-HCV (E1-S and E2-S) proteins, and with the wild-type HBV S protein. ApoE was also detected on the surface of purified SVPs and improved the folding of HCV envelope proteins, but its presence lowered the incorporation of E2-S protein. Immunization of New Zealand rabbits resulted in similar anti-S responses for all rabbits, whereas anti-E1/-E2 antibody titers varied according to the presence or absence of apoE. Regarding the neutralizing potential of these anti-E1/-E2 antibodies, it was higher in rabbits immunized with apoE-bearing particles. In conclusion, the association of apoE with HCV envelope proteins may be a good strategy for improving HCV vaccines based on viral envelope proteins.


Asunto(s)
Apolipoproteínas E/administración & dosificación , Apolipoproteínas E/inmunología , Hepacivirus/inmunología , Virus de la Hepatitis B/inmunología , Vacunas contra Hepatitis Viral/administración & dosificación , Vacunas contra Hepatitis Viral/inmunología , Animales , Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Neutralizantes/sangre , Presentación de Antígeno/inmunología , Línea Celular , Femenino , Hepatitis C/inmunología , Hepatitis C/prevención & control , Anticuerpos contra la Hepatitis C/biosíntesis , Anticuerpos contra la Hepatitis C/sangre , Humanos , Evasión Inmune , Conejos , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/inmunología , Proteínas del Envoltorio Viral/administración & dosificación , Proteínas del Envoltorio Viral/inmunología
20.
J Immunol Res ; 2021: 7855808, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34790830

RESUMEN

Interleukin 2 (IL-2) plays a fundamental role in both immune activation and tolerance and has revolutionized the field of cancer immunotherapy since its discovery. The ability of IL-2 to mediate tumor regression in preclinical and clinical settings led to FDA approval for its use in the treatment of metastatic renal cell carcinoma and metastatic melanoma in the 1990s. Although modest success is observed in the clinic, cancer patients receiving IL-2 therapy experience a wide array of side effects ranging from flu-like symptoms to life-threatening conditions such as vascular leak syndrome. Over the past three decades, efforts have focused on circumventing IL-2-related toxicities by engineering methods to localize IL-2 to the tumor or secondary lymphoid tissue, preferentially activate CD8+ T cells and NK cells, and alter pharmacokinetic properties to increase bioavailability. This review summarizes the various IL-2-based strategies that have emerged, with a focus on chimeric fusion methods.


Asunto(s)
Interleucina-2/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Proteínas Recombinantes de Fusión/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Humanos , Células Asesinas Naturales/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA