Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Cancer Res ; 29(5): 888-898, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36342102

RESUMO

PURPOSE: This phase 1 study (NCT03440437) evaluated the safety, tolerability, pharmacokinetics (PK), and activity of FS118, a bispecific antibody-targeting LAG-3 and PD-L1, in patients with advanced cancer resistant to anti-PD-(L)1 therapy. PATIENTS AND METHODS: Patients with solid tumors, refractory to anti-PD-(L)1-based therapy, received intravenous FS118 weekly with an accelerated dose titration design (800 µg to 0.3 mg/kg) followed by 3+3 ascending dose expansion (1 to 20 mg/kg). Primary objectives were safety, tolerability, and PK. Additional endpoints included antitumor activity, immunogenicity, and pharmacodynamics. RESULTS: Forty-three patients with a median of three prior regimens in the locally advanced/metastatic setting, including at least one anti-PD-(L)1 regimen, received FS118 monotherapy. FS118 was well tolerated, with no serious adverse events relating to FS118 reported. No dose-limiting toxicities (DLT) were observed, and an MTD was not reached. The recommended phase 2 dose of FS118 was established as 10 mg/kg weekly. The terminal half-life was 3.9 days. Immunogenicity was transient. Pharmacodynamic activity was prolonged throughout dosing as demonstrated by sustained elevation of soluble LAG-3 and increased peripheral effector cells. The overall disease control rate (DCR) was 46.5%; this disease control was observed as stable disease, except for one late partial response. Disease control of 54.8% was observed in patients receiving 1 mg/kg or greater who had acquired resistance to PD-(L)1-targeted therapy. CONCLUSIONS: FS118 was well tolerated with no DLTs observed up to and including 20 mg/kg QW. Further studies are warranted to determine clinical benefit in patients who have become refractory to anti-PD-(L)1 therapy. See related commentary by Karapetyan and Luke, p. 835.


Assuntos
Anticorpos Biespecíficos , Antineoplásicos , Neoplasias , Humanos , Interferons , Antígeno B7-H1 , Neoplasias/patologia , Antineoplásicos/efeitos adversos , Anticorpos Biespecíficos/efeitos adversos , Imunoterapia , Biologia
2.
Clin Cancer Res ; 26(15): 4154-4167, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32345647

RESUMO

PURPOSE: With the increased prevalence in checkpoint therapy resistance, there remains a significant unmet need for additional therapies for patients with relapsing or refractory cancer. We have developed FS222, a bispecific tetravalent antibody targeting CD137 and PD-L1, to induce T-cell activation to eradicate tumors without the current toxicity and efficacy limitations seen in the clinic. EXPERIMENTAL DESIGN: A bispecific antibody (FS222) was developed by engineering CD137 antigen-binding sites into the Fc region of a PD-L1 IgG1 mAb. T-cell activation by FS222 was investigated using multiple in vitro assays. The antitumor efficacy, survival benefit, pharmacodynamics, and liver pharmacology of a murine surrogate molecule were assessed in syngeneic mouse tumor models. Toxicology and the pharmacokinetic/pharmacodynamic profile of FS222 were investigated in a non-human primate dose-range finding study. RESULTS: We demonstrated simultaneous binding of CD137 and PD-L1 and showed potent T-cell activation across CD8+ T-cell activation assays in a PD-L1-dependent manner with a CD137/PD-L1 bispecific antibody, FS222. FS222 also activated T cells in a human primary mixed lymphocyte reaction assay, with greater potency than the monospecific mAb combination. FS222 showed no signs of liver toxicity up to 30 mg/kg in a non-human primate dose-range finding study. A surrogate molecule caused significant tumor growth inhibition and survival benefit, concomitant with CD8+ T-cell activation, in CT26 and MC38 syngeneic mouse tumor models. CONCLUSIONS: By targeting CD137 agonism to areas of PD-L1 expression, predominantly found in the tumor microenvironment, FS222 has the potential to leverage a focused, potent, and safe immune response augmenting the PD-(L)1 axis blockade.


Assuntos
Anticorpos Biespecíficos/fisiologia , Antineoplásicos Imunológicos/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Neoplasias Colorretais/tratamento farmacológico , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/antagonistas & inibidores , Animais , Anticorpos Biespecíficos/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Linhagem Celular Tumoral/transplante , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Modelos Animais de Doenças , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Células HEK293 , Humanos , Leucócitos Mononucleares , Macaca fascicularis , Camundongos , Cultura Primária de Células , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
3.
Clin Cancer Res ; 26(13): 3333-3344, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32299814

RESUMO

PURPOSE: Although programmed death-ligand 1 (PD-L1) antibody-based therapy has improved the outcome of patients with cancer, acquired resistance to these treatments limits their clinical efficacy. FS118 is a novel bispecific, tetravalent antibody (mAb2) against human lymphocyte activation gene-3 (LAG-3) and PD-L1 with the potential to reinvigorate exhausted immune cells and overcome resistance mechanisms to PD-L1 blockade. Here, using FS118 and a murine surrogate, we characterized the activity and report a novel mechanism of action of this bispecific antibody. EXPERIMENTAL DESIGN: This study characterizes the binding activity and immune function of FS118 in cell lines and human peripheral blood mononuclear cells and further investigates its antitumor activity and mechanism of action using a surrogate murine bispecific antibody (mLAG-3/PD-L1 mAb2). RESULTS: FS118 demonstrated simultaneous binding to LAG-3 and PD-L1 with high affinity and comparable or better activity than the combination of the single component parts of the mAb2 in blocking LAG-3- and PD-L1-mediated immune suppression and enhancing T-cell activity. In syngeneic tumor mouse models, mLAG-3/PD-L1 mAb2 significantly suppressed tumor growth. Mechanistic studies revealed decreased LAG-3 expression on T cells following treatment with the mouse surrogate mLAG-3/PD-L1 mAb2, whereas LAG-3 expression increased upon treatment with the combination of mAbs targeting LAG-3 and PD-L1. Moreover, following binding of mLAG-3/PD-L1 mAb2 to target-expressing cells, mouse LAG-3 is rapidly shed into the blood. CONCLUSIONS: This study demonstrates a novel benefit of the bispecific approach over a combination of mAbs and supports the further development of FS118 for the treatment of patients with cancer.


Assuntos
Anticorpos Biespecíficos/farmacologia , Antígenos CD/metabolismo , Antineoplásicos Imunológicos/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Ativação Linfocitária/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Animais , Afinidade de Anticorpos , Biomarcadores Tumorais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Imunofenotipagem , Ativação Linfocitária/imunologia , Camundongos , Ligação Proteica , Linfócitos T/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína do Gene 3 de Ativação de Linfócitos
4.
J Immunol ; 199(10): 3395-3405, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-29046347

RESUMO

Nasal allergen challenge (NAC) is a human model of allergic rhinitis (AR) that delivers standardized allergens locally to the nasal mucosa allowing clinical symptoms and biospecimens such as peripheral blood to be collected. Although many studies have focused on local inflammatory sites, peripheral blood, an important mediator and a component of the systemic immune response, has not been well studied in the setting of AR. We sought to investigate immune gene signatures in peripheral blood collected after NAC under the setting of AR. Clinical symptoms and peripheral blood samples from AR subjects were collected during NAC. Fuzzy c-means clustering method was used to identify immune gene expression patterns in blood over time points (before NAC and 1, 2, and 6 h after NAC). We identified and validated seven clusters of differentially expressed immune genes after NAC onset. Clusters 2, 3, and 4 were associated with neutrophil and lymphocyte frequencies and neutrophil/lymphocyte ratio after the allergen challenge. The patterns of the clusters and immune cell frequencies were associated with the clinical symptoms of the AR subjects and were significantly different from healthy nonallergic subjects who had also undergone NAC. Our approach identified dynamic signatures of immune gene expression in blood as a systemic immune response associated with clinical symptoms after NAC. The immune gene signatures may allow cross-sectional investigation of the pathophysiology of AR and may also be useful as a potential objective measurement for diagnosis and treatment of AR combined with the NAC model.


Assuntos
Células Sanguíneas/imunologia , Mucosa Nasal/imunologia , Rinite Alérgica/imunologia , Adulto , Alérgenos/imunologia , Estudos Transversais , Feminino , Humanos , Imunidade , Masculino , Pessoa de Meia-Idade , Família Multigênica/genética , Testes de Provocação Nasal , Pólen/imunologia , Rinite Alérgica/diagnóstico , Rinite Alérgica/genética , Transcriptoma
5.
Cytokine ; 85: 101-8, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27309676

RESUMO

In October 2013, the International Life Sciences Institute - Health and Environmental Sciences Institute Immunotoxicology Technical Committee (ILSI-HESI ITC) held a one-day workshop entitled, "Workshop on Cytokine Release: State-of-the-Science, Current Challenges and Future Directions". The workshop brought together scientists from pharmaceutical, academic, health authority, and contract research organizations to discuss novel approaches and current challenges for the use of in vitro cytokine release assays (CRAs) for the identification of cytokine release syndrome (CRS) potential of novel monoclonal antibody (mAb) therapeutics. Topics presented encompassed a regulatory perspective on cytokine release and assessment, case studies regarding the translatability of preclinical cytokine data to the clinic, and the latest state of the science of CRAs, including comparisons between mAb therapeutics within one platform and across several assay platforms, a novel physiological assay platform, and assay optimization approaches such as determination of FcR expression profiles and use of statistical tests. The data and approaches presented confirmed that multiple CRA platforms are in use for identification of CRS potential and that the choice of a particular CRA platform is highly dependent on the availability of resources for individual laboratories (e.g. positive and negative controls, number of human blood donors), the assay through-put required, and the mechanism-of-action of the therapeutic candidate to be tested. Workshop participants agreed that more data on the predictive performance of CRA platforms is needed, and current efforts to compare in vitro assay results with clinical cytokine assessments were discussed. In summary, many laboratories continue to focus research efforts on the improvement of the translatability of current CRA platforms as well explore novel approaches which may lead to more accurate, and potentially patient-specific, CRS prediction in the future.


Assuntos
Citocinas/sangue , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Bioensaio/métodos , Humanos , Doenças do Sistema Imunitário/sangue , Doenças do Sistema Imunitário/tratamento farmacológico
6.
Bioanalysis ; 3(23): 2657-63, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22136053

RESUMO

The induction of cytokine release is a common consequence of the administration of therapeutic antibodies and in most cases is either tolerated by the patient or can be managed clinically by the administration of corticosteroids. However, in 2006, the administration of TGN1412 to six patients in a Phase I trial resulted in a unprecedentedly high level of cytokine release, systemic organ failure and the hospitalization of the subjects. Whilst the path to failure in this incident was multifactorial, at least one contributing factor was the lack of a robust in vitro model that would allow the prediction of the in vivo activity of a therapeutic antibody. In this article we review the current 'state of the art' of in vitro cytokine release assays and explore potential future developments.


Assuntos
Anticorpos Monoclonais/efeitos adversos , Citocinas/análise , Citocinas/metabolismo , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/uso terapêutico , Humanos
7.
Immunology ; 111(3): 262-72, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15009426

RESUMO

Previous studies have identified a 210 000-molecular weight molecule expressed at a high level on the surface of dendritic cells (DCs) in afferent lymph of cattle and evident on cells with the morphology of DCs in lymphoid tissues. Expression is either absent from other immune cells or is present at a lower level. The molecular weight and cellular distribution suggested that the molecule, called bovine WC6 antigen (workshop cluster), might be an orthologue of human DEC-205 (CD205). To establish whether this was the case, the open reading frame of bovine DEC-205 was amplified, by polymerase chain reaction, from thymic cDNA (accession no. AY264845). The cDNA sequence of bovine DEC-205 had 86% and 78% nucleic acid identity with human and mouse molecules, respectively. COS-7 cells transfected with a plasmid containing the cattle DEC-205 coding region expressed a molecule that stained with WC6-specific monoclonal antibody, showing that ruminant WC6 is an orthologue of DEC-205. Two-colour flow cytometry of mononuclear cells from afferent lymph draining cattle skin, and from blood, confirmed the high level of expression on large cells in lymph that were uniformly DC-LAMP positive and major histocompatibility complex class II positive. Within this DEC-205+ DC-LAMP+ population were subpopulations of cells that expressed the mannose receptor or SIRPalpha. The observations imply that DCs in afferent lymph are all DEC-205high, but not a uniform population of homogeneous mature DCs.


Assuntos
Antígenos CD/análise , Células Dendríticas/imunologia , Lectinas Tipo C/análise , Linfa/imunologia , Receptores de Superfície Celular/análise , Sequência de Aminoácidos , Animais , Antígenos CD/imunologia , Antígenos de Superfície/imunologia , Sequência de Bases , Células COS , Bovinos , Movimento Celular/imunologia , Chlorocebus aethiops , DNA Circular/imunologia , Citometria de Fluxo/métodos , Humanos , Imuno-Histoquímica/métodos , Lectinas Tipo C/imunologia , Leucócitos Mononucleares/imunologia , Proteínas de Membrana Lisossomal , Receptor de Manose , Lectinas de Ligação a Manose/análise , Camundongos , Antígenos de Histocompatibilidade Menor , Reação em Cadeia da Polimerase/métodos , Receptores de Superfície Celular/imunologia , Pele/imunologia , Transfecção
8.
Eur J Immunol ; 32(5): 1472-81, 2002 05.
Artigo em Inglês | MEDLINE | ID: mdl-11981836

RESUMO

Two major sub-populations of dendritic cells (DC) are present in afferent lymph draining the skin of cattle distinguished by expression of signal regulator protein alpha (SIRPalpha). The SIRPalpha(-) population expresses the uncharacterized bovine WC10 antigen (Ag). Initial N-terminal sequencing of the WC10 protein purified by affinity chromatography showed significant homology with human CD26. A cDNA encoding bovine CD26 was cloned and the recombinant molecule expressed in COS-7 cells. Transfectants abrogated the ability of macrophage-derived chemokine (MDC) to cause a calcium flux in bovine PBMC indicating enzymatic activity characteristic of CD26. They also stained with WC10 monoclonal antibody confirming that the Ag is CD26. This is the first description of CD26 expression by DC in vivo or in vitro. It is expressed on a sub-population of ex vivo DC in afferent lymph draining the skin and on sub-populations of DC isolated from prescapular and mesenteric lymph nodes draining the skin or intestine, respectively. CD26 is an exopeptidase with specificity for motifs within the receptor-binding domain of several chemokines including MDC. CD26 mediated truncation of MDC affects the Th cell response effected by the chemokine and may produce a Th1 bias. Transcripts for MDC were present in both CD26(+) and CD26(-) DC, thus CD26 mediated modification of MDC may bias the immune response induced in naive T cells by DC.


Assuntos
Células Dendríticas/imunologia , Dipeptidil Peptidase 4/genética , Dipeptidil Peptidase 4/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Células COS , Bovinos , Quimiocinas/metabolismo , Clonagem Molecular , DNA Complementar/genética , Células Dendríticas/classificação , Células Dendríticas/enzimologia , Expressão Gênica , Humanos , Linfonodos/citologia , Linfonodos/imunologia , Macrófagos/imunologia , Dados de Sequência Molecular , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Homologia de Sequência de Aminoácidos , Pele/citologia , Pele/enzimologia , Pele/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...