Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Adv Mater ; 36(9): e2309039, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37903320

RESUMO

Activation of the innate immune system counteracts tumor-induced immunosuppression. Hence, small molecule-based toll-like receptor 7/8 agonists (TLR7/8a), which can modulate immunosuppression in the tumor microenvironment along with the activation of innate immunity, are emerging as essential components of cancer immunotherapy. However, the clinical application of synthetic TLR7/8a therapies is limited by systemic immune-associated toxicity and immune tolerance induced by uncontrolled stimulatory activities and repeated treatments. To address these limitations, a dynamic immunomodulation strategy incorporating masking and temporal recovery of the activity of TLR7/8a through prodrug-like TLR7/8a (pro-TLR7/8a) at the molecular level and a sustained and controlled release of active TLR7/8a from nanoliposome (pro-TLR7/8a) (NL(pro-TLR7/8)) in a macroscale depot are designed. Immunization with cationic NL(pro-TLR7/8) and anionic antigens triggers robust activation of innate immune cells as well as antigen-specific T cell responses, eliciting reprogramming of immunosuppressive cells into tumor-suppressive cells, with decreased systemic adverse effects and immune tolerance. Combination treatment with NL(pro-TLR7/8a) and immune checkpoint inhibitors (anti-CTLA-4 plus anti-PD-L1) or nanoliposomes (Doxorubicin) has synergistic effects on antitumor immunity in various tumor models. The concept of pro-TLR7/8a suggested herein may facilitate the advancement of small-molecule-based immunomodulators for clinical translation and safe and effective cancer immunotherapy.


Assuntos
Neoplasias , Receptor 7 Toll-Like , Humanos , Fatores Imunológicos , Adjuvantes Imunológicos/farmacologia , Tolerância Imunológica , Neoplasias/tratamento farmacológico , Microambiente Tumoral
2.
Small ; 20(13): e2307694, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37967333

RESUMO

Although adoptive cell-based therapy is illuminated as one of the promising approaches in cancer immunotherapy, it shows low antitumor efficacy because transferred cells adapt and alter toward a pro-tumoral phenotype in response to the tumor's immunosuppressive milieu. Herein, nanoengineered macrophages anchored with functional liposome armed with cholesterol-conjugated Toll-like receptor 7/8 agonist (masked TLR7/8a, m7/8a) are generated to overcome the shortcomings of current macrophage-based therapies and enhance the remodeling of the immunosuppressive tumor microenvironment (TME). The liposome-anchored macrophages (LAMΦ-m7/8a), are fabricated by anchoring dibenzocyclooctyne-modified liposome(m7/8a) onto azido-expressing macrophages via a bio-orthogonal click reaction, are continuously invigorated due to the slow internalization of liposome(m7/8a) and sustained activation. LAMΦ-m7/8a secreted ≈3 and 33-fold more IL-6 and TNF-α than conventional M1-MΦ, maintained the M1 phenotype, and phagocytosed tumor cells for up to 48 h in vitro. Both intratumoral and intravenous injections of LAMΦ-m7/8a induced effective antitumor efficacy when treated in combination with doxorubicin-loaded liposomes in 4T1-tumor bearing mice. It not only increases the infiltration of antigen-specific CD8+ T cells secreting granzyme B, IFN-γ, and TNF-α within the TME, but also reduces myeloid-derived suppressor cells. These results suggest that LAMΦ-m7/8a may provide a suitable alternative to next-generation cell-based therapy platform.


Assuntos
Neoplasias , Receptor 7 Toll-Like , Camundongos , Animais , Linfócitos T CD8-Positivos , Fator de Necrose Tumoral alfa , Lipossomos , Microambiente Tumoral , Macrófagos , Neoplasias/terapia , Imunoterapia/métodos , Adjuvantes Imunológicos , Linhagem Celular Tumoral
4.
Nat Nanotechnol ; 18(4): 390-402, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36635335

RESUMO

Although conventional innate immune stimuli contribute to immune activation, they induce exhausted immune cells, resulting in suboptimal cancer immunotherapy. Here we suggest a kinetically activating nanoadjuvant (K-nanoadjuvant) that can dynamically integrate two waves of innate immune stimuli, resulting in effective antitumour immunity without immune cell exhaustion. The combinatorial code of K-nanoadjuvant is optimized in terms of the order, duration and time window between spatiotemporally activating Toll-like receptor 7/8 agonist and other Toll-like receptor agonists. K-nanoadjuvant induces effector/non-exhausted dendritic cells that programme the magnitude and persistence of interleukin-12 secretion, generate effector/non-exhausted CD8+ T cells, and activate natural killer cells. Treatment with K-nanoadjuvant as a monotherapy or in combination therapy with anti-PD-L1 or liposomes (doxorubicin) results in strong antitumour immunity in murine models, with minimal systemic toxicity, providing a strategy for synchronous and dynamic tailoring of innate immunity for enhanced cancer immunotherapy.


Assuntos
Linfócitos T CD8-Positivos , Neoplasias , Animais , Camundongos , Imunoterapia/métodos , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/uso terapêutico , Imunidade Inata , Neoplasias/terapia
5.
J Control Release ; 343: 564-583, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35124126

RESUMO

Although cancer immunotherapy has emerged as a novel cancer treatment modality, it still suffers from low therapeutic efficacy in clinics due to the presence of a low number of activated immune cells and immunosuppressive factors in the tumor microenvironment (TME). Immunomodulatory ribonucleic acids (RNAs) have been developed to improve the therapeutic efficacy of cancer immunotherapy through either regulating target cell functions [i.e., messenger RNA (mRNA) or small interfering RNA (siRNA)] or stimulating immune cells [i.e., toll-like receptors (TLRs) or cytosolic retinoic acid-inducible gene I (RIG-I) agonist]. However, RNA-based therapeutics face many biological barriers, including ineffective delivery to target cells, degradation by ribonucleases (RNases), and difficulties in passing through the cellular membranes. In this review, we discuss nanoparticle-based delivery strategies that can overcome these hurdles to enhance RNA-based immunomodulation in cancer immunotherapy. Various nanoparticle-based delivery has been reported to increase the delivery efficacy of RNAs, by improving cellular uptake, RNA stability, and accumulation at the desired sites (target cells and intracellular compartments). The nanoparticle-based delivery of multifaceted immunomodulatory RNAs could enhance cancer immunotherapy through the regulating functions of immune cells, tumor cells, and immunosuppressive factors as well as stimulating the immune cells by recognition of endosomal TLRs and cytosolic RIG-I. Nanotechnology-assisted RNA-based therapeutics are expected to offer tremendous potential and advances for treating cancer, viral infections, and other diseases.


Assuntos
Nanopartículas , RNA Mensageiro , RNA Interferente Pequeno , Sistemas de Liberação de Medicamentos , Humanos , Imunoterapia , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , RNA Mensageiro/uso terapêutico , Receptores Toll-Like , Microambiente Tumoral
6.
Adv Sci (Weinh) ; 8(19): e2102043, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34363349

RESUMO

The deficiency of antigen-specific T cells and the induction of various treatment-induced immunosuppressions still limits the clinical benefit of cancer immunotherapy. Although the chemo-immunotherapy adjuvanted with Toll-like receptor 7/8 agonist (TLR 7/8a) induces immunogenic cell death (ICD) and in situ vaccination effect, indoleamine 2,3-dioxygenase (IDO) is also significantly increased in the tumor microenvironment (TME) and tumor-draining lymph node (TDLN), which offsets the activated antitumor immunity. To address the treatment-induced immunosuppression, an assemblable immune modulating suspension (AIMS) containing ICD inducer (paclitaxel) and supra-adjuvant (immune booster; R848 as a TLR 7/8a, immunosuppression reliever; epacadostat as an IDO inhibitor) is suggested and shows that it increases cytotoxic T lymphocytes and relieves the IDO-related immunosuppression (TGF-ß, IL-10, myeloid-derived suppressor cells, and regulatory T cells) in both TME and TDLN, by the formation of in situ depot in tumor bed as well as by the efficient migration into TDLN. Local administration of AIMS increases T cell infiltration in both local and distant tumors and significantly inhibits the metastasis of tumors to the lung. Reverting treatment-induced secondary immunosuppression and reshaping "cold tumor" into "hot tumor" by AIMS also increases the response rate of immune checkpoint blockade therapy, which promises a new nanotheranostic strategy in cancer immunotherapy.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/imunologia , Terapia de Imunossupressão/métodos , Imunoterapia/métodos , Nanomedicina/métodos , Animais , Modelos Animais de Doenças , Imunoterapia/efeitos adversos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...