Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 644: 1-7, 2023 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-36621147

RESUMEN

Pan-T cell targeting by CD3-based T cell engagers has brought program-shift treatment and management of blood tumors. However, these modalities have been shown to provoke all types of T cells leading to cytokine storm syndrome, and activate Treg cells. Thus, modulating and potentiating the antitumor responses of a specific T cell subset was encouraged. We initially found that high purity of mucosa-associated invariant T (MAIT) cells could be expanded by the combination of plate-immobilized Vα7.2 mAb (Clone 3C10) and IL2 plus IL15. Then, we generated a novel anti-Vα7.2 TCR bsAb, Vα7.2 x PD-L1, to invoke the anti-tumor potency of these expanded MAIT cells. Furthermore, our data have demonstrated that Vα7.2 x PD-L1 could mediate the cell-to-cell conjunction between MAIT cell and tumor cell line, selectively elicit the activation, cytokine production, degranulation, and cytotoxicity of the expanded MAIT cells in the presence of target cell only. Collectively, this proof-of-concept study provides a new tool to explore the clinical potential of MAIT cells in fighting against PD-L1 positive solid tumors and suggests additional encouragement in designing novel T cell engagers targeting TCR alpha chain specific innate-like T cells subsets, other than pan CD3+ T cells.


Asunto(s)
Células T Invariantes Asociadas a Mucosa , Antígeno B7-H1/metabolismo , Subgrupos de Linfocitos T , Receptores de Antígenos de Linfocitos T/metabolismo , Membrana Mucosa
2.
J Immunother Cancer ; 10(12)2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36460337

RESUMEN

BACKGROUND: Agents blocking programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) have been approved for triple-negative breast cancer (TNBC). However, the response rate of anti-PD-1/PD-L1 is still unsatisfactory, partly due to immunosuppressive factors such as transforming growth factor-beta (TGF-ß). In our previous pilot study, the bispecific antibody targeting TGF-ß and murine PD-L1 (termed YM101) showed potent antitumor effect. In this work, we constructed a bispecific antibody targeting TGF-ß and human PD-L1 (termed BiTP) and explored the antitumor effect of BiTP in TNBC. METHODS: BiTP was developed using Check-BODYTM bispecific platform. The binding affinity of BiTP was measured by surface plasmon resonance, ELISA, and flow cytometry. The bioactivity was assessed by Smad and NFAT luciferase reporter assays, immunofluorescence, western blotting, and superantigen stimulation assays. The antitumor activity of BiTP was explored in humanized epithelial-mesenchymal transition-6-hPDL1 and 4T1-hPDL1 murine TNBC models. Immunohistochemical staining, flow cytometry, and bulk RNA-seq were used to investigate the effect of BiTP on immune cell infiltration. RESULTS: BiTP exhibited high binding affinity to dual targets. In vitro experiments verified that BiTP effectively counteracted TGF-ß-Smad and PD-L1-PD-1-NFAT signaling. In vivo animal experiments demonstrated that BiTP had superior antitumor activity relative to anti-PD-L1 and anti-TGF-ß monotherapy. Mechanistically, BiTP decreased collagen deposition, enhanced CD8+ T cell penetration, and increased tumor-infiltrating lymphocytes. This improved tumor microenvironment contributed to the potent antitumor activity of BiTP. CONCLUSION: BiTP retains parent antibodies' binding affinity and bioactivity, with superior antitumor activity to parent antibodies in TNBC. Our data suggest that BiTP might be a promising agent for TNBC treatment.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias de la Mama Triple Negativas , Humanos , Ratones , Animales , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Factor de Crecimiento Transformador beta , Proyectos Piloto , Anticuerpos Biespecíficos/farmacología , Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Antineoplásicos , Microambiente Tumoral
3.
J Hematol Oncol ; 15(1): 142, 2022 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-36209176

RESUMEN

BACKGROUND: Non-inflamed tumors, including immune-excluded and immune-desert tumors, are commonly resistant to anti-PD-1/PD-L1 (α-PD-1/PD-L1) therapy. Our previous study reported the potent antitumor activity of anti-TGF-ß/PD-L1 bispecific antibody YM101 in immune-excluded tumors. However, YM101 had limited antitumor activity in immune-desert models. MSA-2 is a novel oral stimulator of interferon genes (STING) agonist, which activates the innate immune system and may synergize with YM101 in overcoming immunotherapy resistance. METHODS: The dose-dependent effect of MSA-2 on STING signaling was determined by interferon-ß level. The maturation and function of dendritic cell (DC) were measured by flow cytometry, RNA-seq, one-way mixed lymphocyte reaction (MLR), OVA peptide pulse, and cytokine/chemokine detection. The synergistic effect between MSA-2 and YM101 was assessed by one-way MLR. The macrophage activation was measured by flow cytometry and cytokine/chemokine detection. The in vivo antitumor activity of MSA-2 combined with YM101 was explored in syngeneic murine tumor models. After treatments, the alterations in the tumor microenvironment (TME) were detected by flow cytometry, immunohistochemistry staining, immunofluorescence staining, RNA-seq, and single-cell RNA-seq (scRNA-seq). RESULTS: MSA-2 could promote the maturation and antigen presentation capability of murine DC. In the one-way MLR assay, MSA-2 synergized with YM101 in enhancing naive T cell activation. Moreover, MSA-2 stimulated the classical activation of macrophage, without significant influence on alternative activation. Further in vivo explorations showed that MSA-2 increased multiple proinflammatory cytokines and chemokines in the TME. MSA-2 combined with YM101 remarkedly retarded tumor growth in immune-excluded and immune-desert models, with superior antitumor activity to monotherapies. Flow cytometry, bulk RNA-seq, and scRNA-seq assays indicated that the combination therapy simultaneously boosted the innate and adaptive immunity, promoted antigen presentation, improved T cell migration and chemotaxis, and upregulated the numbers and activities of tumor-infiltrating lymphocytes. CONCLUSION: Our results demonstrate that MSA-2 synergizes with YM101 in boosting antitumor immunity. This immune cocktail therapy effectively overcomes immunotherapy resistance in immune-excluded and immune-desert models.


Asunto(s)
Anticuerpos Biespecíficos , Proteínas de la Membrana/agonistas , Neoplasias , Animales , Anticuerpos Biespecíficos/farmacología , Anticuerpos Biespecíficos/uso terapéutico , Antígeno B7-H1 , Línea Celular Tumoral , Citocinas , Humanos , Inmunoterapia , Interferón beta/farmacología , Interferones/farmacología , Ratones , Factor de Crecimiento Transformador beta , Microambiente Tumoral
4.
Front Immunol ; 13: 923969, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35784353

RESUMEN

The potent cytotoxic property of Vγ2Vδ2 T cells makes them attractive for adoptive T cell transfer therapy. The transfusing of the expanded Vγ2Vδ2 T cells into cancer patients shows well-tolerated, but the clinical response rates are required to be improved, implying that there is still an unmet efficacy with low toxicity for this novel anti-tumor therapy. In this study, we test the anti-tumor efficacy of a Y-body-based bispecific antibody (bsAb) Vγ2 x PD-L1 that preferentially redirects Vγ2Vδ2 T cells to combat PD-L1 positive tumor cells. With nanomolar affinity levels to Vγ2Vδ2 T cells and PD-L1+ tumor cells, Vγ2 x PD-L1 bridges a Vγ2Vδ2 T cell with a SKOV3 tumor cell to form a cell-to-cell conjugation. In a PD-L1-dependent manner, the bsAb elicits effective activation (CD25+CD69+), IFNγ releasing, degranulation (CD107a+), and cytokine production (IFNγ+ and TNFα+) of expanded Vγ2Vδ2 T cells. The activations of the Vγ2Vδ2 T cells eliminate PD-L1-expressing human cancer cell lines, including H1975, SKOV3, A375, H1299, and H2228 cells, but not PD-L1 negative cells including HEK-293 (293) cells and healthy PBMCs. Finally, we show that combining Vγ2 x PD-L1 with adoptively transferring Vγ2Vδ2 T cells inhibits the growth of existing tumor xenografts and increases the number of Vγ2Vδ2 T cells into the tumor bed. Vγ2 x PD-L1 represents a promising reagent for increasing the efficacy of adoptively transferred Vγ2Vδ2 T cells in the treatment of PD-L1 positive malignant tumors.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias , Anticuerpos Biespecíficos/farmacología , Antígeno B7-H1/inmunología , Células HEK293 , Humanos , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T , Linfocitos T
5.
Cell Commun Signal ; 20(1): 101, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35778766

RESUMEN

BACKGROUND: Coinfection with HIV and Plasmodium parasites is fairly common, but the sequence of infection with these two pathogens and their impact on disease progression are poorly understood. METHODS: A Chinese rhesus macaque HIV and Plasmodium coinfection model was established to compare the impact of pre-existing and subsequent malaria on the progression of SIV infection. RESULTS: We found that a pre-existing malaria caused animals to produce a greater number of CD4+CCR5+ T cells for SIV replication, resulting in higher viral loads. Conversely, subsequent malaria induced a substantially larger proportion of CD4+CD28highCD95high central memory T cells and a stronger SIV-specific T cell response, maintained the repertoire diversity of SIV-specific T cell receptors, and generated new SIV-specific T cell clonotypes to trace SIV antigenic variation, resulting in improved survival of SIV-infected animals. CONCLUSION: The complex outcomes of this study may have important implications for research on human HIV and malaria coinfection. The infection order of the two pathogens (HIV and malaria parasites) should be emphasized. Video abstract.


Asunto(s)
Coinfección , Infecciones por VIH , Malaria , Síndrome de Inmunodeficiencia Adquirida del Simio , Virus de la Inmunodeficiencia de los Simios , Animales , Macaca mulatta , Virus de la Inmunodeficiencia de los Simios/fisiología
6.
J Healthc Eng ; 2021: 5906855, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34733454

RESUMEN

Objective: To study the effect of a care bundle combined with continuous positive airway pressure (CPAP) in the postanesthesia care unit (PACU) on rapid recovery after pulmonary tumor resection. Methods: A total of 135 patients requiring anesthesia resuscitation after pulmonary tumor resection in our hospital from June 2020 to February 2021 were selected. They were randomly divided into three groups: the PACU experimental group, PACU control group, and operating room resuscitation (OR) group. Subsequently, their intraoperative clinical symptoms, parameters in monitoring postoperative respiratory status, and follow-up results were compared among the three groups. Results: The PACU experimental group had the highest number of right lesions, while the OR group had the highest intraoperative blood transfusion volume, urine volume, intraoperative colloid volume, intrapulmonary shunt, and intraoperative physician handover rate (P < 0.05). Before surgery, serum potassium (K) in the PACU experimental group was significantly higher than that in the OR group but lower than that in the PACU control group (P < 0.01). During the time in the PACU, blood partial pressure of oxygen (PO2) and oxygen index (OI) levels in the PACU experimental group were significantly higher than those in the other groups (P < 0.01). After surgery, total PACU stay time, time from PACU to extubation, and stay after extubation were markedly reduced in the PACU experimental group (P < 0.05). The highest number of patients with drainage was found in the PACU experimental group, while the highest number of patients without drainage was found in the PACU control group. Conclusion: A care bundle combined with CPAP in the PACU can improve the monitoring time of respiratory status and improve blood gas parameters, thus accelerating the postoperative rehabilitation process of patients undergoing pulmonary tumor resection.


Asunto(s)
Neoplasias Pulmonares , Paquetes de Atención al Paciente , Presión de las Vías Aéreas Positiva Contínua , Humanos , Pulmón/cirugía , Neoplasias Pulmonares/cirugía , Oxígeno
7.
Cell Discov ; 7(1): 82, 2021 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-34493710

RESUMEN

The pandemic of COVID-19 caused by SARS-CoV-2 has raised a new challenges to the scientific and industrious fields after over 1-year spread across different countries. The ultimate approach to end the pandemic is the timely application of vaccines to achieve herd immunity. Here, a novel SARS-CoV-2 receptor-binding domain (RBD) homodimer was developed as a SARS-CoV-2 vaccine candidate. Formulated with aluminum adjuvant, RBD dimer elicited strong immune response in both rodents and non-human primates, and protected mice from SARS-CoV-2 challenge with significantly reducing viral load and alleviating pathological injury in the lung. In the non-human primates, the vaccine could prevent majority of the animals from SARS-CoV-2 infection in the respiratory tract and reduce lung damage. In addition, antibodies elicited by this vaccine candidate showed cross-neutralization activities to SARS-CoV-2 variants. Furthermore, with our expression system, we provided a high-yield RBD homodimer vaccine without additional biosafety or special transport device supports. Thus, it may serve as a safe, effective, and low-cost SARS-CoV-2 vaccine candidate.

8.
J Hematol Oncol ; 14(1): 146, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34526097

RESUMEN

BACKGROUND: Our previous work showed that the anti-TGF-ß/PD-L1 bispecific antibody YM101 effectively overcame anti-PD-L1 resistance in immune-excluded tumor models. However, in immune-desert models, the efficacy of YM101 was limited. Bivalent manganese (Mn2+) is identified as a natural stimulator of interferon genes (STING) agonist, which might enhance cancer antigen presentation and improve the therapeutic effect of YM101. METHODS: The effect of Mn2+ on STING pathway was validated by western blotting and enzyme-linked immunosorbent assay. Dendritic cell (DC) maturation was measured by flow cytometry. The synergistic effect between Mn2+ and YM101 in vitro was determined by one-way mixed lymphocyte reaction, CFSE dilution assay, and cytokine detection. The in vivo antitumor effect of Mn2+ plus YM101 therapy was assessed in CT26, EMT-6, H22, and B16 tumor models. Flow cytometry, RNA-seq, and immunofluorescent staining were adopted to investigate the alterations in the tumor microenvironment. RESULTS: Mn2+ could activate STING pathway and promote the maturation of human and murine DC. The results of one-way mixed lymphocyte reaction showed that Mn2+ synergized YM101 in T cell activation. Moreover, in multiple syngeneic murine tumor models, Mn2+ plus YM101 therapy exhibited a durable antitumor effect and prolonged the survival of tumor-bearing mice. Relative to YM101 monotherapy and Mn2+ plus anti-PD-L1 therapy, Mn2+ plus YM101 treatment had a more powerful antitumor effect and a broader antitumor spectrum. Mechanistically, Mn2+ plus YM101 strategy simultaneously regulated multiple components in the antitumor immunity and drove the shift from immune-excluded or immune-desert to immune-inflamed tumors. The investigation in the TME indicated Mn2+ plus YM101 strategy activated innate and adaptive immunity, enhanced cancer antigen presentation, and upregulated the density and function of tumor-infiltrating lymphocytes. This normalized TME and reinvigorated antitumor immunity contributed to the superior antitumor effect of the combination therapy. CONCLUSION: Combining Mn2+ with YM101 has a synergistic antitumor effect, effectively controlling tumor growth and prolonging the survival of tumor-bearing mice. This novel cocktail strategy has the potential to be a universal regimen for inflamed and non-inflamed tumors.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Manganeso/uso terapéutico , Neoplasias/terapia , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Sinergismo Farmacológico , Humanos , Inmunoterapia/métodos , Ratones Endogámicos BALB C , Neoplasias/inmunología , Factor de Crecimiento Transformador beta/inmunología
9.
Front Immunol ; 12: 654080, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34040604

RESUMEN

Vγ2Vδ2 T cell-based immunotherapy has benefited some patients in clinical trials, but the overall efficacy is low for solid tumor patients. In this study, a bispecific antibody against both PD-L1 and CD3 (PD-L1 x CD3), Y111, could efficiently bridge T cells and PD-L1 expressing tumor cells. The Y111 prompted fresh CD8+ T cell-mediated lysis of H358 cells, but spared this effect on the fresh Vδ2+ T cells enriched from the same donors, which suggested that Y111 could bypass the anti-tumor capacity of the fresh Vγ2Vδ2 T cells. As the adoptive transfer of the expanded Vγ2Vδ2 T cells was approved to be safe and well-tolerated in clinical trials, we hypothesized that the combination of the expanded Vγ2Vδ2 T cells with the Y111 would provide an alternative approach of immunotherapy. Y111 induced the activation of the expanded Vγ2Vδ2 T cells in a dose-dependent fashion in the presence of PD-L1 positive tumor cells. Moreover, Y111 increased the cytotoxicity of the expanded Vγ2Vδ2 T cells against various NSCLC-derived tumor cell lines with the releases of granzyme B, IFNγ, and TNFα in vitro. Meanwhile, the adoptive transferred Vγ2Vδ2 T cells together with the Y111 inhibited the growth of the established xenografts in NPG mice. Taken together, our data suggested a clinical potential for the adoptive transferring the Vγ2Vδ2 T cells with the Y111 to treat PD-L1 positive solid tumors.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Antineoplásicos Inmunológicos/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Complejo CD3/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Traslado Adoptivo , Animales , Anticuerpos Biespecíficos/aislamiento & purificación , Citocinas , Citotoxicidad Inmunológica , Femenino , Humanos , Inmunoterapia Adoptiva , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Activación de Linfocitos , Ratones , Unión Proteica , Proteínas Recombinantes de Fusión , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Hematol Oncol ; 14(1): 27, 2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33593403

RESUMEN

BACKGROUND: Therapeutic antibodies targeting programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) axis induce potent and durable anti-tumor responses in multiple types of cancers. However, only a subset of patients benefits from anti-PD-1/PD-L1 therapies. As a negative regulator of anti-tumor immunity, TGF-ß impairs the efficacy of anti-PD-1/PD-L1 and induces drug resistance. Developing a novel treatment strategy to simultaneously block PD-1/PD-L1 and TGF-ß would be valuable to enhance the effect of anti-PD-1/PD-L1 and relieve drug resistance. METHODS: Based on the Check-BODY™ technology platform, we developed an anti-TGF-ß/PD-L1 bispecific antibody YM101. The bioactivity of the anti-TGF-ß moiety was determined by Smad-luciferase reporter assay, transwell assay, western blotting, CCK-8, and flow cytometry. The bioactivity of the anti-PD-L1 moiety was measured by T cell activation assays. EMT-6, CT26, and 3LL tumor models were used to investigate the anti-tumor activity of YM101 in vivo. RNA-seq, immunohistochemical staining, and flow cytometry were utilized to analyze the effect of YM101 on the tumor microenvironment. RESULTS: YM101 could bind to TGF-ß and PD-L1 specifically. In vitro experiments showed that YM101 effectively counteracted the biological effects of TGF-ß and PD-1/PD-L1 pathway, including activating Smad signaling, inducing epithelial-mesenchymal transition, and immunosuppression. Besides, in vivo experiments indicated the anti-tumor activity of YM101 was superior to anti-TGF-ß and anti-PD-L1 monotherapies. Mechanistically, YM101 promoted the formation of 'hot tumor': increasing the numbers of tumor infiltrating lymphocytes and dendritic cells, elevating the ratio of M1/M2, and enhancing cytokine production in T cells. This normalized tumor immune microenvironment and enhanced anti-tumor immune response might contribute to the robust anti-tumor effect of YM101. CONCLUSION: Our results demonstrated that YM101 could simultaneously block TGF-ß and PD-L1 pathways and had a superior anti-tumor effect compared to the monotherapies.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias/tratamiento farmacológico , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Animales , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/farmacología , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/farmacología , Línea Celular Tumoral , Humanos , Inhibidores de Puntos de Control Inmunológico/química , Inhibidores de Puntos de Control Inmunológico/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias/patología
11.
Expert Opin Investig Drugs ; 29(11): 1295-1308, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32822558

RESUMEN

INTRODUCTION: CD38 is expressed by some cells of hematological malignancies and tumor-related immunosuppressive cells, including regulatory T cells, regulatory B cells, and myeloid-derived suppressor cells. CD38 is an effective target in some hematological malignancies such as multiple myeloma (MM). Daratumumab (Dara), a CD38-targeting antibody, can eliminate CD38high immune suppressor cells and is regarded as a standard therapy for MM because of its outstanding clinical efficacy. Other CD38 monospecific antibodies, such as isatuximab, MOR202, and TAK079, showed promising effects in clinical trials. AREA COVERED: This review examines the expression, function, and targeting of CD38 in MM and its potential to deplete immunosuppressive cells in solid cancers. We summarize the distribution and biological function of CD38 and discuss the application of anti-CD38 drugs in hematological malignancies. We also analyz the role of CD38+ immune cells in the tumor microenvironment to encourage additional investigations that target CD38 in solid cancers. PubMed and ClinicalTrials were searched to identify relevant literature from the database inception to 30 April 2020. EXPERT OPINION: There is convincing evidence that CD38-targeted immunotherapeutics reduce CD38+ immune suppressor cells. This result suggests that CD38 can be exploited to treat solid tumors by regulating the immunosuppressive microenvironment.


Asunto(s)
Antineoplásicos/farmacología , Mieloma Múltiple/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , ADP-Ribosil Ciclasa 1/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Humanos , Inmunoterapia/métodos , Glicoproteínas de Membrana/inmunología , Terapia Molecular Dirigida , Mieloma Múltiple/inmunología , Neoplasias/inmunología , Microambiente Tumoral/inmunología
12.
BMC Infect Dis ; 19(1): 965, 2019 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-31718574

RESUMEN

BACKGROUND: The co-occurrence of human immunodeficiency virus (HIV) infection and malaria in humans in endemic areas raises the question of whether one of these infections affects the course of the other. Although epidemiological studies have shown the impact of HIV infection on malaria, the mechanism(s) are not yet fully understood. Using a Chinese rhesus macaque coinfection model with simian immunodeficiency virus (SIV) and Plasmodium cynomolgi (Pc) malaria, we investigated the effect of concurrent SIV infection on the course of malaria and the underlying immunological mechanism(s). METHODS: We randomly assigned ten Chinese rhesus monkeys to two groups based on body weight and age. The SIV-Pc coinfection animals (S + P group) were infected intravenously with SIVmac251 eight weeks prior to malaria infection, and the control animals (P group) were infected intravenously with only Pc-infected red blood cells. After malaria was cured with chloroquine phosphate, we also initiated a secondary malaria infection that lasted 4 weeks. We monitored body weight, body temperature and parasitemia, measured SIV viral loads, hemoglobin and neopterin, and tracked the CD4+, CD8+, and CD4+ memory subpopulations, Ki67 and apoptosis by flow cytometry. Then, we compared these parameters between the two groups. RESULTS: The animals infected with SIV prior to Pc infection exhibited more severe malaria symptoms characterized by longer episodes, higher parasitemia, more severe anemia, greater body weight loss and higher body temperature than the animals infected with Pc alone. Concurrent SIV infection also impaired immune protection against the secondary Pc challenge infection. The coinfected animals showed a reduced B cell response to Pc malaria and produced lower levels of Pc-specific antibodies. In addition, compared to the animals subjected to Pc infection alone, the animals coinfected with SIV and Pc had suppressed total CD4+ T cells, CD4+CD28highCD95high central memory T cells, and CD4+CD28lowCD95- naïve T cells, which may result from the imbalanced immune activation and faster CD4+ T cell turnover in coinfected animals. CONCLUSIONS: SIV infection aggravates malaria physiologically and immunologically in Chinese rhesus monkeys. This nonhuman primate SIV and Pc malaria coinfection model might be a useful tool for investigating human HIV and malaria coinfection and developing effective therapeutics.


Asunto(s)
Malaria/patología , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , China , Coinfección/complicaciones , Modelos Animales de Enfermedad , Humanos , Inmunidad Humoral , Macaca mulatta , Malaria/complicaciones , Malaria/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/complicaciones , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/fisiología , Carga Viral
13.
J Exp Clin Cancer Res ; 38(1): 355, 2019 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-31412896

RESUMEN

BACKGROUND: Human epidermal growth factor receptor 2 (HER2) is overexpressed in multiple cancers, which is associated with poor prognosis. Herceptin and other agents targeting HER2 have potent antitumor efficacy in patients with HER2-positive cancers. However, the development of drug resistance adversely impacts the efficacy of these treatments. It is therefore urgent to develop new HER2-targeted therapies. Bispecific antibodies (BsAbs) could guide immune cells toward tumor cells, and produced remarkable effects in some cancers. METHODS: A BsAb named M802 that targets HER2 and CD3 was produced by introducing a salt bridge and knobs-into-holes (KIHs) packing into the structure. Flow cytometry was performed to determine its binding activity and cytotoxicity. CCK-8, Annexin V/PI staining, western blotting, and ELISA were utilized to study its effect on cell proliferation, apoptosis, the signaling pathways of tumor cells, and the secretion of cytokines by immune cells. Subcutaneous tumor mouse models were used to analyze the in vivo antitumor effects of M802. RESULTS: We generated a new format of BsAb, M802, consisting of a monovalent unit against HER2 and a single chain unit against CD3. Our in vitro and in vivo experiments indicated that M802 recruited CD3-positive immune cells and was more cytotoxic than Herceptin in cells with high expression of HER2, low expression of HER2, and Herceptin resistance. Although M802 showed weaker effects than Herceptin on the PI3K/AKT and MAPK pathways, it was more cytotoxic due to its specific recognition of HER2 and its ability to recruit effector cells via its anti-CD3 moiety. CONCLUSIONS: Our results indicated that M802 exhibited potent antitumor efficacy in vitro and in vivo. M802 retained the function of Herceptin in antitumor signaling pathways, and also recruited CD3-positive immune cells to eliminate HER2-positive tumor cells. Therefore, M802 might be a promising HER2 targeted agent.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Antineoplásicos Inmunológicos/farmacología , Complejo CD3/antagonistas & inhibidores , Receptor ErbB-2/antagonistas & inhibidores , Animales , Anticuerpos Biespecíficos/química , Antineoplásicos Inmunológicos/química , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Citotoxicidad Inmunológica/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Melanoma Experimental , Ratones , Peso Molecular , Transducción de Señal/efectos de los fármacos , Análisis Espectral , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Hum Immunol ; 70(2): 79-84, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19100802

RESUMEN

Generation and adoptive transfusion of alloreactive cytotoxic T lymphocytes (CTLs) specific for tumor are expected to circumvent tumor tolerance. Here we describe a novel protocol to raise allo-restricted, peptide-specific CTLs by co-culture of murine splenocytes and autologous macrophage bearing an allogeneic H-2K molecule associated with its restricted peptide (peptide/allo-MHC). The extracellular domains of H-2K(d) were fused with constant domains of murine IgG2a heavy chain to generate a fusion protein (peptide/H-2K(d)/IgG2aFc, the dimer) consisting of divalent TCR-ligands and an IgG Fc receptor type I (FcgammaRI)-reactive moiety. The dimer is able to bind to macrophage (Mvarphi) of H-2K(k) via the interaction of the Fc part with FcgammaRI, and cause the H-2K(k) Mvarphi to be coated with the peptide/H-2K(d) complex. The results show that proliferation of CD8+ cells is enhanced and that the specific-tetramer stained CD8+ cells appear more frequently by co-culture of H-2K(k) splenocytes with the autologous Mvarphi loaded with the dimer. Furthermore, the CD8+ T cells from the co-cultural bulk exhibit an elevated cytotoxicity against a specific target (H-2K(d)-restricted, peptide-specific cytotoxicity), compared with that against the irrelevant targets. This study provides a strategy for preparation of allo-restricted, peptide-specific CTLs, which may add to our arsenal for adoptive immunotherapy to eliminate chronic virally infected or tumor cells.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/inmunología , Macrófagos/citología , Macrófagos/inmunología , Bazo/citología , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Animales , Linfocitos T CD8-positivos/citología , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Citotoxicidad Inmunológica , Femenino , Antígenos H-2/genética , Antígenos H-2/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/inmunología , Ratones , Ratones Endogámicos BALB C/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo
15.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 33(4): 359-64, 2008 Apr.
Artículo en Chino | MEDLINE | ID: mdl-18460782

RESUMEN

OBJECTIVE: To determine the cellular compatibility of combined deproteinized bone(DPB) coated with hepatocyte growth factor (HGF), and to observe the adherent effect of osteoblasts in response to HGF. METHODS: Osteoblasts were isolated from fetal rabbits. Osteoblasts were cultured with DPB coated with HGF and deproteinized bone as experimental group and contral group, respectively. The proliferation and alkalinephosphatase activity were tested. Their growth was examined by inverted phase contrast microscope and scanning electronmicroscope. RESULTS: The osteoblasts were attached to the outside and inside surfaces and grew well. HGF/DPB could stimulate the alkalinephosphatase activity of the osteoblasts and improve the proliferation of the osteoblasts. CONCLUSION: HGF/DPB has good biocompatibility and bone induction. HGF could improve the adherent effect of DPB on osteoblasts, and it could be used as scaffold material for the bone tissue engineering.


Asunto(s)
Materiales Biocompatibles/farmacología , Factor de Crecimiento de Hepatocito/farmacología , Osteoblastos/citología , Ingeniería de Tejidos/métodos , Andamios del Tejido , Animales , Sustitutos de Huesos/metabolismo , Huesos/citología , Proliferación Celular , Células Cultivadas , Femenino , Feto , Osteogénesis , Embarazo , Conejos
16.
Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi ; 22(12): 1441-4, 2008 Dec.
Artículo en Chino | MEDLINE | ID: mdl-19137885

RESUMEN

OBJECTIVE: To investigate the clinical characteristics and methods of diagnosis and treatment multiple level thoracolumbar spinal fractures. METHODS: From March 2002 to March 2006, 17 patients with 35 thoracolumbar spinal fractures were treated, 13 males and 4 females, aged 21-52 years old (36.4 on average), among whom there were 10 cases of traffic accident injury and 7 of high falling injury. One fracture was located at T2, 1 at T3, 1 at T10, 4 at T11, 6 at T12, 5 at L1, at L2, 7 at L3, 5 at L4, and 2 at L15, with a total of 35 segments including 26 segments with unstable fractures and 9 segments with stable compression fractures. According to the Frankel grade, there was 1 case of grade A, 1 of grade B, 2 of grade C, 5 of grade and 8 of grade E. The preoperative height of the anterior border of the vertebral body was (20.8 +/- 3.8) mm and the preoperative kyphosis angle was (16.2 +/- 3.4) degrees. All the unstable fractures were performed operation. Sixteen injured vertebras were treated with long-segment pedicle screw internal fixation; 8 were treated with short-segment pedicle screw internal fixation, and 2 were treated with anterior fusion and fixation. Five injured vertebras with stable compression fractures were not treated and 4 were treated with pedicle screw implantation. RESULTS: The operation time was 1.8-4.2 hours and the amount of blood loss was 300-900 mL. The incisions obtained healing by first intention after the operation. All 17 patients were followed up for 13-41 months (18 months on average), and radiological evaluation showed no failure of the internal fixation. After the operation, Frankel scale assessment showed that 1 patient of grade A improved to grade B, 1 of grade B improved to grade C, 1 of grade improved to grade D, 1 of grade C improved to grade E, 5 of grade D improved to grade E, and 8 of grade E had no improvement. At the final postoperative follow-up, the height of the anterior border of the vertebral body was (31.9 -/+ 3.2) mm and kyphosis angle was (6.8 +/- 3.7) degrees, which were significantly different from those of preoperation (P < 0.01). CONCLUSION: The treatment of multiple level thoracolumbar spinal fractures should be individualized according to the patients' actual conditions in order to obtain decompression and stability of spines.


Asunto(s)
Vértebras Lumbares/lesiones , Fracturas de la Columna Vertebral/cirugía , Vértebras Torácicas/lesiones , Adulto , Tornillos Óseos , Femenino , Fijación Interna de Fracturas , Humanos , Masculino , Persona de Mediana Edad , Radiografía , Fracturas de la Columna Vertebral/clasificación , Fracturas de la Columna Vertebral/diagnóstico por imagen
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...