Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
N Engl J Med ; 389(18): 1672-1684, 2023 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-37870974

RESUMO

BACKGROUND: Neoadjuvant or adjuvant immunotherapy can improve outcomes in patients with resectable non-small-cell lung cancer (NSCLC). Perioperative regimens may combine benefits of both to improve long-term outcomes. METHODS: We randomly assigned patients with resectable NSCLC (stage II to IIIB [N2 node stage] according to the eighth edition of the AJCC Cancer Staging Manual) to receive platinum-based chemotherapy plus durvalumab or placebo administered intravenously every 3 weeks for 4 cycles before surgery, followed by adjuvant durvalumab or placebo intravenously every 4 weeks for 12 cycles. Randomization was stratified according to disease stage (II or III) and programmed death ligand 1 (PD-L1) expression (≥1% or <1%). Primary end points were event-free survival (defined as the time to the earliest occurrence of progressive disease that precluded surgery or prevented completion of surgery, disease recurrence [assessed in a blinded fashion by independent central review], or death from any cause) and pathological complete response (evaluated centrally). RESULTS: A total of 802 patients were randomly assigned to receive durvalumab (400 patients) or placebo (402 patients). The duration of event-free survival was significantly longer with durvalumab than with placebo; the stratified hazard ratio for disease progression, recurrence, or death was 0.68 (95% confidence interval [CI], 0.53 to 0.88; P = 0.004) at the first interim analysis. At the 12-month landmark analysis, event-free survival was observed in 73.4% of the patients who received durvalumab (95% CI, 67.9 to 78.1), as compared with 64.5% of the patients who received placebo (95% CI, 58.8 to 69.6). The incidence of pathological complete response was significantly greater with durvalumab than with placebo (17.2% vs. 4.3% at the final analysis; difference, 13.0 percentage points; 95% CI, 8.7 to 17.6; P<0.001 at interim analysis of data from 402 patients). Event-free survival and pathological complete response benefit were observed regardless of stage and PD-L1 expression. Adverse events of maximum grade 3 or 4 occurred in 42.4% of patients with durvalumab and in 43.2% with placebo. Data from 62 patients with documented EGFR or ALK alterations were excluded from the efficacy analyses in the modified intention-to-treat population. CONCLUSIONS: In patients with resectable NSCLC, perioperative durvalumab plus neoadjuvant chemotherapy was associated with significantly greater event-free survival and pathological complete response than neoadjuvant chemotherapy alone, with a safety profile that was consistent with the individual agents. (Funded by AstraZeneca; AEGEAN ClinicalTrials.gov number, NCT03800134.).


Assuntos
Antineoplásicos Imunológicos , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Adjuvantes Imunológicos/uso terapêutico , Administração Intravenosa , Antineoplásicos Imunológicos/administração & dosagem , Antineoplásicos Imunológicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Antígeno B7-H1/administração & dosagem , Antígeno B7-H1/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/cirurgia , Terapia Combinada , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/cirurgia , Recidiva Local de Neoplasia/tratamento farmacológico
2.
Acta Biomater ; 136: 473-484, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34571271

RESUMO

The continuous activation and expansion of tumor-specific T cells by various means are the main goal of cancer immunotherapy. Tumor cells overexpress fibrinogen-like protein 1 (FGL1) and programmmed death-ligand 1 (PD-L1), which respectively bind to lymphocyte-activation gene 3 (LAG-3) and programmmed death-1(PD-1) on T cells, forming important signaling pathways (FGL1/LAG-3 and PD-1/PD-L1) that negatively regulate immune responses. In order to interfere with the inhibitory function of FGL1 and PD-L1 proteins, we designed a new type of reactive oxygen species (ROS)-sensitive nanoparticles to load FGL1 siRNA (siFGL1) and PD-L1 siRNA (siPD-L1), which was formed from a stimuli-responsive polymer with a poly-l-lysine-thioketal and modified cis-aconitate to facilitate endosomal escape. Moreover, tumor-penetrating peptide iRGD and ROS-responsive nanoparticles were co-administered to further enhance the delivery efficiency of siFGL1 and siPD-L1, thereby significantly reducing the protein levels of FGL1 and PD-L1 in tumor cells. Our findings indicated that the dual delivery of FGL1/PD-L1 siRNA was a new and powerful treatment method, which was characterized by increasing the infiltration of effector CD4+ and CD8+ T cells, effectively alleviating the tumor immunosuppressive microenvironment. These findings also supported the superiority and feasibility of nanoparticle-mediated tumor immunotherapy, and may provide a different perspective for cancer treatment. STATEMENT OF SIGNIFICANCE: In addition to the idea that cancer vaccines can promote T cell immune responses, nanoparticle delivery modulators (such as small interfering RNA (siRNA) targeting immunosuppressive pathways) may provide more information for the research of nanoparticle-mediated cancer immunotherapy. In this study, we designed a new intelligent nano-delivery system for co-delivery of siFGL1 and siPD-L1, and demonstrated the ability to down-regulate the expression levels of FGL1 and PD-L1 proteins in tumor cells in vitro and in vivo. The constructed nanoparticle had a good tumor microenvironment responsiveness, and the delivery efficiency was enhanced by co-injection with tumor penetrating peptide iRGD. This project proposed a new strategy for tumor immunotherapy based on smart nano-delivery systems, and explored more possibilities for tumor therapy.


Assuntos
Antígeno B7-H1 , Fibrinogênio/administração & dosagem , Nanopartículas , Oligopeptídeos/uso terapêutico , Animais , Antígeno B7-H1/administração & dosagem , Linhagem Celular Tumoral , Imunoterapia , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio , Microambiente Tumoral
3.
Biomed Res Int ; 2021: 5574282, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34497850

RESUMO

Programmed cell death 1 ligand (PD-L1) and its receptor (PD-1) are key molecules for immunoregulation and immunotherapy. PD-L1 binding PD-1 is an effective way to regulate T or B cell immunity in autoimmune diseases such as rheumatoid arthritis (RA). In our study, we overexpressed PD-L1 by constructing a recombinant of PD-L1-lentiviral vector, which was subsequently used to transfect mouse bone marrow mesenchymal stem cells (MBMMSCs) and significantly suppressed the development of collagen-induced arthritis (CIA) in DBA/1j mice. In addition, PD-L1-transfected MBMMSCs (PD-L1-MBMMSCs) ameliorated joint damage, reduced proinflammatory cytokine expression, and inhibited T and B cell activation. Furthermore, PD-L1-MBMMSCs decreased the number of dendritic cells and increased the numbers of regulatory T cells and regulatory B cells in joints of CIA mice. In conclusion, our results provided a potential therapeutic strategy for RA treatment with PD-L1-MBMMSC-targeted therapy.


Assuntos
Artrite Experimental/terapia , Artrite Reumatoide/terapia , Antígeno B7-H1/administração & dosagem , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Linfócitos T Reguladores/imunologia , Animais , Artrite Experimental/imunologia , Artrite Experimental/patologia , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Células Cultivadas , Modelos Animais de Doenças , Ativação Linfocitária , Masculino , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos DBA
4.
Carbohydr Polym ; 265: 118065, 2021 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-33966829

RESUMO

Wound bacterial infections and tumor recurrence are the main reasons for the poor prognosis after primary tumor resection. Here, we fabricated a novel therapeutic nanocomposite using chitosan (CS) hydrogel combined with black phosphate nanosheets (BPNSs) and in situ grown copper nanoparticles (CuNPs). The obtained hydrogel (CS@BPNSs@CuNPs), possessing a remarkable temperature-sensitive spongy-like state, offered 24.98 % blood clotting index. The released BPNSs@CuNPs could produce reactive oxygen species (ROS) to kill infected invasive bacteria (98.1 %) and inhibit local residual tumor cell regeneration (11.3 %). Moreover, by coupling the photothermal properties of BPNSs, the BPNSs@CuNPs showed 19.6 % penetration rate to cross the blood tumor barrier (BTB) for treating brain tumors. The hydrogel platform was further combined with aPD-L1-based immunotherapy to employ its synergetic therapeutic effect in the prevention of tumors. The in vivo studies showed that biodegradable hydrogel could hold a great potential as a novel strategy for improving postoperative therapy and multi-tumor treatments.


Assuntos
Quitosana/administração & dosagem , Cobre/química , Hidrogéis/administração & dosagem , Nanocompostos/química , Neoplasias/tratamento farmacológico , Infecção da Ferida Cirúrgica/tratamento farmacológico , Animais , Antibacterianos/administração & dosagem , Antibacterianos/química , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Antígeno B7-H1/administração & dosagem , Antígeno B7-H1/química , Quitosana/química , Hemostáticos/administração & dosagem , Hemostáticos/química , Humanos , Hidrogéis/química , Injeções/métodos , Camundongos , Nanocompostos/administração & dosagem , Nanopartículas/química , Recidiva Local de Neoplasia/tratamento farmacológico , Fósforo/química , Espécies Reativas de Oxigênio/metabolismo , Temperatura
5.
Zhongguo Fei Ai Za Zhi ; 24(4): 217-235, 2021 Apr 20.
Artigo em Chinês | MEDLINE | ID: mdl-33896153

RESUMO

Non-small cell lung cancer (NSCLC) is the most common pathological type of lung cancer. The systemic antitumor therapy of advanced NSCLC has undergone renovations of chemotherapy, targeted therapy and immunotherapy, which results in greatly improved survival for patients with advanced NSCLC. Immune checkpoint inhibitors (ICIs), especially targeting programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1), has changed the treatment paradigm of NSCLC. ICIs have become the standard treatment for advanced NSCLC without epidermal growth factor receptor(EGFR) mutation or anaplastic lymphomakinase(ALK) translocation in the first- or second-line setting, and for locally advanced NSCLC following concurrent radiotherapy and chemotherapy. ICIs are also promising in adjuvant/neoadjuvant therapy. More and more ICIs have been approved domestically for the treatment of NSCLC. Led by the NSCLC expert committee of Chinese Society of Clinical Oncology (CSCO), this consensus was developed and updated based on thoroughly reviewing domestic and foreign literatures, clinical trial data, systematic reviews, experts' discussion and the consensus(2019 version). This consensus will aid domestic clinicians in the treatment of NSCLC with ICIs.
.


Assuntos
Antineoplásicos Imunológicos/administração & dosagem , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Inibidores de Checkpoint Imunológico/administração & dosagem , Neoplasias Pulmonares/tratamento farmacológico , Antígeno B7-H1/administração & dosagem , China , Consenso , Humanos , Imunoterapia , Receptor de Morte Celular Programada 1/administração & dosagem
6.
Int J Radiat Oncol Biol Phys ; 110(2): 510-520, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33383126

RESUMO

PURPOSE: The establishment of a preclinical model of the abscopal effect on hepatocellular carcinoma (HCC) and evaluation of whether the hypofractionated radiation therapy (RT) multitumor Hepa1-6 mouse HCC model could be used to suppress nonradiated tumor mass was performed in this study. METHODS AND MATERIALS: Hepa1-6 mouse liver cancer cell lines were used to form tumors. Immunogenicity was analyzed using ELISpot and immune cell labeled antibody. Interferon (IFN) ß expression was confirmed through polymerase chain reaction. RESULTS: After investigation, the intratumoral transcription of type Ⅰ IFN increased by 2-fold. The antitumor immune response to Hepa 1-6 cells induced by radiation was increased. Moreover, the influx of activated CD8+ T cells was increased in nonirradiated tumors. The number of dendritic cells and activation status were evaluated by flow cytometry on the second day after irradiation. Flow cytometry revealed a significantly increased dendritic cell population expressing the CD11c molecule in tumor-draining lymph nodes. Furthermore, because irradiation leads to adaptation of immune resistance of tumor cells against RT, we sought to elucidate a potent tool to overcome the resistance and confirm the ability of PD-L1 antibody to survive late RT resistance. CONCLUSIONS: The immunologic mechanism of the abscopal effect was revealed and the application of PD-L1 inhibitor successfully performed as a breakthrough in late RT resistance in the Hepa1-6 tumor model.


Assuntos
Carcinoma Hepatocelular/terapia , Inibidores de Checkpoint Imunológico/uso terapêutico , Neoplasias Hepáticas/terapia , Tolerância a Radiação/imunologia , Radiocirurgia/métodos , Animais , Antineoplásicos , Antígeno B7-H1/administração & dosagem , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/metabolismo , Antígenos CD11/metabolismo , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/efeitos da radiação , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Terapia Combinada/métodos , Células Dendríticas/citologia , Células Dendríticas/imunologia , Células Dendríticas/efeitos da radiação , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Interferon Tipo I/metabolismo , Interferon beta/metabolismo , Interferon gama/imunologia , Interferon gama/metabolismo , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Linfonodos/metabolismo , Linfócitos do Interstício Tumoral/citologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Hipofracionamento da Dose de Radiação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Carga Tumoral/efeitos da radiação
7.
Int J Radiat Oncol Biol Phys ; 110(2): 492-506, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32768562

RESUMO

PURPOSE: Mounting evidence demonstrates that combining radiation therapy (RT) with immunotherapy can reduce tumor burden in a subset of patients. However, conventional systemic delivery of immunotherapeutics is often associated with significant adverse effects, which force treatment cessation. The aim of this study was to investigate a minimally invasive therapeutics delivery approach to improve clinical response while attenuating toxicity. METHODS AND MATERIALS: We used a nanofluidic drug-eluting seed (NDES) for sustained intratumoral delivery of combinational antibodies CD40 and PDL1. To enhance immune and tumor response, we combined the NDES intratumoral platform with RT to treat the 4T1 murine model of advanced triple negative breast cancer. We compared the efficacy of NDES against intraperitoneal administration, which mimics conventional systemic treatment. Tumor growth was recorded, and local and systemic immune responses were assessed via imaging mass cytometry and flow cytometry. Livers and lungs were histologically analyzed for evaluation of toxicity and metastasis, respectively. RESULTS: The combination of RT and sustained intratumoral immunotherapy delivery of CD40 and PDL1 via NDES (NDES CD40/PDL1) showed an increase in both local and systemic immune response. In combination with RT, NDES CD40/PDL1 achieved significant tumor burden reduction and liver inflammation mitigation compared with systemic treatment. Importantly, our treatment strategy boosted the abscopal effect toward attenuating lung metastatic burden. CONCLUSIONS: Overall, our study demonstrated superior efficacy of combination treatment with RT and sustained intratumoral immunotherapy via NDES, offering promise for improving therapeutic index and clinical response.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Antineoplásicos/administração & dosagem , Antígenos CD40/imunologia , Imunoterapia/métodos , Nanomedicina Teranóstica/métodos , Neoplasias de Mama Triplo Negativas/terapia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Humanizados/efeitos adversos , Antineoplásicos/efeitos adversos , Antígeno B7-H1/administração & dosagem , Antígeno B7-H1/imunologia , Antígenos CD40/administração & dosagem , Linfócitos T CD8-Positivos , Linhagem Celular Tumoral , Terapia Combinada/métodos , Implantes de Medicamento , Feminino , Liofilização , Imunoterapia/efeitos adversos , Injeções Intralesionais/métodos , Injeções Intraperitoneais , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos BALB C , Intervalo Livre de Progressão , Hipofracionamento da Dose de Radiação , Distribuição Aleatória , Critérios de Avaliação de Resposta em Tumores Sólidos , Resultado do Tratamento , Neoplasias de Mama Triplo Negativas/imunologia , Neoplasias de Mama Triplo Negativas/patologia , Carga Tumoral
8.
Biotechnol Lett ; 42(5): 727-736, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32006351

RESUMO

Programmed death ligand 1 (PDL1, CD274, B7-H1) has been identified as the ligand for the immune inhibitory receptor programmed death 1 protein (PD1/PDCD1). PDL1 is a member of B7 family of immune molecules and this protein together with PDL2, are two ligands for PD1 expressed on activated lymphoid cells. By binding to PD1 on activated T cells, PDL1 may inhibit T cell responses by inducing apoptosis. Accordingly, it leads to the immune evasion of cancers and contribute to tumor growth, thus PDL1 is regarded as therapeutic target for malignant cancers. We selected PDL1 specific nanobodies from a high quality dromedary camel immune library by phage display technology, three anti-PDL1-VHHs were developed.


Assuntos
Antígeno B7-H1/administração & dosagem , Neoplasias/imunologia , Anticorpos de Domínio Único/metabolismo , Animais , Antígeno B7-H1/imunologia , Camelus/metabolismo , Técnicas de Visualização da Superfície Celular , Células HEK293 , Humanos , Imunização , Neoplasias/tratamento farmacológico , Receptor de Morte Celular Programada 1/metabolismo , Anticorpos de Domínio Único/farmacologia , Evasão Tumoral/efeitos dos fármacos
9.
Nat Med ; 24(12): 1852-1858, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30478423

RESUMO

Programmed cell death protein-1/ligand-1 (PD-1/PD-L1) blockade is effective in a subset of patients with several tumor types, but predicting patient benefit using approved diagnostics is inexact, as some patients with PD-L1-negative tumors also show clinical benefit1,2. Moreover, all biopsy-based tests are subject to the errors and limitations of invasive tissue collection3-11. Preclinical studies of positron-emission tomography (PET) imaging with antibodies to PD-L1 suggested that this imaging method might be an approach to selecting patients12,13. Such a technique, however, requires substantial clinical development and validation. Here we present the initial results from a first-in-human study to assess the feasibility of imaging with zirconium-89-labeled atezolizumab (anti-PD-L1), including biodistribution, and secondly test its potential to predict response to PD-L1 blockade (ClinicalTrials.gov identifiers NCT02453984 and NCT02478099). We imaged 22 patients across three tumor types before the start of atezolizumab therapy. The PET signal, a function of tracer exposure and target expression, was high in lymphoid tissues and at sites of inflammation. In tumors, uptake was generally high but heterogeneous, varying within and among lesions, patients, and tumor types. Intriguingly, clinical responses in our patients were better correlated with pretreatment PET signal than with immunohistochemistry- or RNA-sequencing-based predictive biomarkers, encouraging further development of molecular PET imaging for assessment of PD-L1 status and clinical response prediction.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Antígeno B7-H1/administração & dosagem , Tomografia por Emissão de Pósitrons , Radioisótopos/administração & dosagem , Zircônio/administração & dosagem , Adulto , Idoso , Anticorpos Monoclonais/química , Anticorpos Monoclonais Humanizados , Antígeno B7-H1/química , Biópsia , Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Radioisótopos/química , Compostos Radiofarmacêuticos/administração & dosagem , Compostos Radiofarmacêuticos/química , Distribuição Tecidual/efeitos dos fármacos , Neoplasias de Mama Triplo Negativas/diagnóstico por imagem , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias da Bexiga Urinária/diagnóstico por imagem , Neoplasias da Bexiga Urinária/patologia , Zircônio/química
10.
BMJ ; 362: k3529, 2018 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-30201790

RESUMO

OBJECTIVE: To evaluate the relative efficacy of programmed cell death 1 (PD-1) or programmed cell death ligand 1 (PD-L1) inhibitors versus conventional drugs in patients with cancer that were PD-L1 positive and PD-L1 negative. DESIGN: Meta-analysis of randomised controlled trials. DATA SOURCES: PubMed, Embase, Cochrane database, and conference abstracts presented at the American Society of Clinical Oncology and European Society of Medical Oncology up to March 2018. REVIEW METHODS: Studies of PD-1 or PD-L1 inhibitors (avelumab, atezolizumab, durvalumab, nivolumab, and pembrolizumab) that had available hazard ratios for death based on PD-L1 positivity or negativity were included. The threshold for PD-L1 positivity or negativity was that PD-L1 stained cell accounted for 1% of tumour cells, or tumour and immune cells, assayed by immunohistochemistry staining methods. RESULTS: 4174 patients with advanced or metastatic cancers from eight randomised controlled trials were included in this study. Compared with conventional agents, PD-1 or PD-L1 inhibitors were associated with significantly prolonged overall survival in both patients that were PD-L1 positive (n=2254, hazard ratio 0.66, 95% confidence interval 0.59 to 0.74) and PD-L1 negative (1920, 0.80, 0.71 to 0.90). However, the efficacies of PD-1 or PD-L1 blockade treatment in patients that were PD-L1 positive and PD-L1 negative were significantly different (P=0.02 for interaction). Additionally, in both patients that were PD-L1 positive and PD-L1 negative, the long term clinical benefits from PD-1 or PD-L1 blockade were observed consistently across interventional agent, cancer histotype, method of randomisation stratification, type of immunohistochemical scoring system, drug target, type of control group, and median follow-up time. CONCLUSIONS: PD-1 or PD-L1 blockade therapy is a preferable treatment option over conventional therapy for both patients that are PD-L1 positive and PD-L1 negative. This finding suggests that PD-L1 expression status alone is insufficient in determining which patients should be offered PD-1 or PD-L1 blockade therapy.


Assuntos
Antígeno B7-H1/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Imunoterapia/métodos , Neoplasias/tratamento farmacológico , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Antineoplásicos Imunológicos/administração & dosagem , Antineoplásicos Imunológicos/uso terapêutico , Antígeno B7-H1/administração & dosagem , Carcinoma Pulmonar de Células não Pequenas/secundário , Humanos , Metástase Neoplásica/tratamento farmacológico , Nivolumabe/administração & dosagem , Nivolumabe/uso terapêutico , Receptor de Morte Celular Programada 1/efeitos dos fármacos , Ensaios Clínicos Controlados Aleatórios como Assunto , Análise de Sobrevida , Resultado do Tratamento
11.
PLoS One ; 13(6): e0199497, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29944697

RESUMO

BACKGROUND: Multidrug resistant (MDR) bacterial pathogens are a serious problem of increasing importance facing the medical community. MDR bacteria typically infect the most immunologically vulnerable: patients in intensive care units, patients with extensive comorbidities, oncology patients, hemodialysis patients, and other immune suppressed individuals are likely to fall victim to these pathogens. One promising novel approach to treatment of MDR bacteria is immuno-adjuvant therapy to boost patient immunity. Success with this strategy would have the major benefit of providing protection against a number of MDR pathogens. OBJECTIVES: This study had two main objectives. First, immunophenotyping of peripheral blood mononuclear cells from patients with sepsis associated with MDR bacteria was performed to examine for findings indicative of immunosuppression. Second, the ability of three immuno-adjuvants with distinct mechanisms of action to reverse CD4 and CD8 T cell dysfunction, a pathophysiological hallmark of sepsis, was evaluated. RESULTS: Septic patients with MDR bacteria had increased expression of the inhibitory receptor PD-1 and its ligand PD-L1 and decreased monocyte HLA-DR expression compared to non-septic patients. All three immuno-adjuvants, IL-7, anti-PD-L1, and OX-40L, increased T cell production of IFN-γ in a subset of septic patients with MDR bacteria: IL-7 was most efficacious. There was a strong trend toward increased mortality in patients whose T cells failed to increase IFN-γ production in response to the three treatments. CONCLUSION: Immuno-adjuvant therapy reversed T cell dysfunction, a key pathophysiological mechanism in septic patients with MDR bacteria.


Assuntos
Antígeno B7-H1/administração & dosagem , Farmacorresistência Bacteriana Múltipla/imunologia , Interleucina-7/administração & dosagem , Receptores OX40/administração & dosagem , Sepse/imunologia , Sepse/microbiologia , Linfócitos T/imunologia , Adjuvantes Imunológicos/farmacologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Apresentação de Antígeno/efeitos dos fármacos , Apresentação de Antígeno/imunologia , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Feminino , Humanos , Interferon gama/metabolismo , Leucócitos/efeitos dos fármacos , Leucócitos/imunologia , Masculino , Pessoa de Meia-Idade , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Ligante OX40/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Receptores de Interleucina-7/metabolismo , Sepse/sangue , Linfócitos T/efeitos dos fármacos
12.
Sci Rep ; 8(1): 1605, 2018 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-29371688

RESUMO

Programmed cell death 1 ligand 1 (PD-L1) is a ligand of programmed cell death 1 (PD-1) that functions as an immune checkpoint by down-regulating immune responses. To determine whether PD-L1 is a therapy target in vitiligo treatment, Pmel-1 vitiligo mice were treated with a PD-L1 fusion protein. Treatment with this fusion protein significantly reversed/suppressed depigmentation development in adult Pmel-1 mice. Mechanistically, enrichment of regulatory T cells (Treg) in the skin was detected after PD-L1 fusion protein treatment in Pmel-1 mice. Furthermore, Tregs abundance was also increased in both the spleen and circulation of Pmel-1 mice treated with PD-L1. These data indicate that PD-L1 protein therapy inhibits the immune response and reverses depigmentation development in Pmel-1 vitiligo mice.


Assuntos
Antígeno B7-H1/administração & dosagem , Pele/patologia , Linfócitos T Reguladores/imunologia , Vitiligo/patologia , Vitiligo/terapia , Animais , Camundongos , Proteínas Recombinantes de Fusão/administração & dosagem , Resultado do Tratamento
13.
Lancet Oncol ; 18(12): e731-e741, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29208439

RESUMO

Use of immune checkpoint inhibitors targeting the programmed cell death protein-1/programmed cell death-ligand 1 and cytotoxic T lymphocyte-associated protein-4 axes has yielded impressive results in some clinical trials. However, only a subset of patients initially respond to these inhibitors, and increasing clinical evidence indicates that a substantial proportion of initial responders ultimately relapse with lethal, drug-resistant disease months or years later. Studies that have used massively parallel sequencing have shed light on the rich functional landscape of mutations that endow tumour cells with the ability to evade T-cell-mediated immunosurveillance. Cancer genomes bear signatures of clonal evolution and selection, particularly implicating acquired defects in interferon receptor signalling and antigen presentation. In this Review, we discuss the biological processes that operate in the formation of so-called immunoresistant niches, and describe the latest progress in the development of combination strategies to reinstate immunosurveillance in immune-refractory tumours.


Assuntos
Antígeno B7-H1/antagonistas & inibidores , Pontos de Checagem do Ciclo Celular/imunologia , Imunoterapia/métodos , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Anticorpos Monoclonais/uso terapêutico , Antígeno B7-H1/administração & dosagem , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Feminino , Humanos , Masculino , Terapia de Alvo Molecular , Neoplasias/genética , Prognóstico , Receptor de Morte Celular Programada 1/administração & dosagem , Medição de Risco , Resultado do Tratamento
14.
Med Sci Monit ; 23: 3360-3366, 2017 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-28697172

RESUMO

BACKGROUND Renal cell carcinoma (RCC) is among the most common malignant cancers of males worldwide. For advanced RCC patients, there still is no effective therapy. Immune checkpoint blockade therapies have shown benefits for many cancers, but previous clinical trials of immune checkpoint blockade therapies in RCC patients achieved only modest results. MATERIAL AND METHODS We explored the effects of combining chemotherapy with immune checkpoint blockade therapy in RCC xenograft mouse models. We also studied the potential mechanisms by which chemotherapy might enhance the efficacy of immune checkpoint blockade therapy, both in vitro and in vivo. RESULTS Our results showed that many commonly used chemotherapy agents can induce immunogenic marker release in RCC cell lines. Importantly, the RCC xenograft mouse model mice who received the combination treatment of 5-fluorouracil (5-FU) and anti-programmed cell death-ligand 1 (PD-L1) antibodies (Abs) had longer survival times compared to those who received 5-FU or anti-PD-L1 Abs alone. Also, increased key cytokines that promote tumor immunity, such as IL-2, IFN-γ, and TNF-α, as well as tumor-infiltrating cytotoxic T cells, were also increased after the combination treatment. CONCLUSIONS We conclude that 5-FU can sensitize RCC to anti-PD-L1 treatment by releasing the immune suppression in the tumor microenvironment.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/imunologia , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/imunologia , Animais , Antígeno B7-H1/administração & dosagem , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Terapia Combinada , Citocinas/imunologia , Modelos Animais de Doenças , Fluoruracila/administração & dosagem , Humanos , Interleucina-2/imunologia , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Camundongos , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Microambiente Tumoral , Fator de Necrose Tumoral alfa/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Stroke ; 48(8): 2255-2262, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28706113

RESUMO

BACKGROUND AND PURPOSE: Intracerebral hemorrhage (ICH) is a neurologically destructive stroke, for which no valid treatment is available. This preclinical study examined the therapeutic effect of PD-L1 (programmed death ligand 1), a B7 family member and a ligand for both PD-1 (programmed death 1) and B7-1 (CD80), in a murine ICH model. METHODS: ICH was induced by injecting autologous blood into 252 male C57BL/6 and Rag1-/- mice. One hour later, ICH mice were randomly assigned to receive an intraperitoneal injection of vehicle, PD-L1, or anti-PD-L1 antibody. Neurological function was assessed along with brain edema, brain infiltration of immune cells, blood-brain barrier integrity, neuron death, and mTOR (mammalian target of rapamycin) pathway products. RESULTS: PD-L1 significantly attenuated neurological deficits, reduced brain edema, and decreased hemorrhage volume in ICH mice. PD-L1 specifically downsized the number of brain-infiltrating CD4+ T cells and the percentages of Th1 and Th17 cells but increased the percentages of Th2 and regulatory T cells. In the PD-L1-treated group, we observed an amelioration of the inflammatory milieu, decreased cell death, and enhanced blood-brain barrier integrity. PD-L1 also inhibited the mTOR pathway. The administration of anti-PD-L1 antibody produced the opposite effects to those of PD-L1 in ICH mice. CONCLUSIONS: PD-L1 provided protection from the damaging consequences of ICH.


Assuntos
Antígeno B7-H1/administração & dosagem , Lesões Encefálicas/prevenção & controle , Hemorragia Cerebral/prevenção & controle , Modelos Animais de Doenças , Fármacos Neuroprotetores/administração & dosagem , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/patologia , Lesões Encefálicas/imunologia , Lesões Encefálicas/patologia , Hemorragia Cerebral/imunologia , Hemorragia Cerebral/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia
16.
Nat Commun ; 7: 10501, 2016 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-26883990

RESUMO

Despite compelling antitumour activity of antibodies targeting the programmed death 1 (PD-1): programmed death ligand 1 (PD-L1) immune checkpoint in lung cancer, resistance to these therapies has increasingly been observed. In this study, to elucidate mechanisms of adaptive resistance, we analyse the tumour immune microenvironment in the context of anti-PD-1 therapy in two fully immunocompetent mouse models of lung adenocarcinoma. In tumours progressing following response to anti-PD-1 therapy, we observe upregulation of alternative immune checkpoints, notably T-cell immunoglobulin mucin-3 (TIM-3), in PD-1 antibody bound T cells and demonstrate a survival advantage with addition of a TIM-3 blocking antibody following failure of PD-1 blockade. Two patients who developed adaptive resistance to anti-PD-1 treatment also show a similar TIM-3 upregulation in blocking antibody-bound T cells at treatment failure. These data suggest that upregulation of TIM-3 and other immune checkpoints may be targetable biomarkers associated with adaptive resistance to PD-1 blockade.


Assuntos
Imunidade Adaptativa , Neoplasias Pulmonares/imunologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Idoso , Animais , Antígeno B7-H1/administração & dosagem , Antígeno B7-H1/imunologia , Receptor Celular 2 do Vírus da Hepatite A , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Masculino , Camundongos , Pessoa de Meia-Idade , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/imunologia , Receptores Virais/genética , Receptores Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA