Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(11)2023 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-37298069

RESUMO

Chimeric antigen receptor T cell (CAR-T) therapy in hematologic malignancies has made great progress, but there are still some problems. First, T cells from tumor patients show an exhaustion phenotype; thus, the persistence and function of the CAR-Ts are poor, and achieving a satisfactory curative effect is difficult. Second, some patients initially respond well but quickly develop antigen-negative tumor recurrence. Thirdly, CAR-T treatment is not effective in some patients and is accompanied by severe side effects, such as cytokine release syndrome (CRS) and neurotoxicity. The solution to these problems is to reduce the toxicity and enhance the efficacy of CAR-T therapy. In this paper, we describe various strategies for reducing the toxicity and enhancing the efficacy of CAR-T therapy in hematological malignancies. In the first section, strategies for modifying CAR-Ts using gene-editing technologies or combining them with other anti-tumor drugs to enhance the efficacy of CAR-T therapy are introduced. The second section describes some methods in which the design and construction of CAR-Ts differ from the conventional process. The aim of these methods is to enhance the anti-tumor activity of CAR-Ts and prevent tumor recurrence. The third section describes modifying the CAR structure or installing safety switches to radically reduce CAR-T toxicity or regulating inflammatory cytokines to control the symptoms of CAR-T-associated toxicity. Together, the knowledge summarized herein will aid in designing better-suited and safer CAR-T treatment strategies.


Assuntos
Neoplasias Hematológicas , Receptores de Antígenos Quiméricos , Humanos , Receptores de Antígenos Quiméricos/genética , Recidiva Local de Neoplasia/etiologia , Imunoterapia Adotiva/efeitos adversos , Imunoterapia Adotiva/métodos , Neoplasias Hematológicas/patologia , Terapia Baseada em Transplante de Células e Tecidos , Receptores de Antígenos de Linfócitos T/genética
2.
Front Immunol ; 14: 1145441, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37180104

RESUMO

Background: CD44v6 chimeric antigen receptor T (CD44v6 CAR-T) cells demonstrate strong anti-tumor ability and safety in acute myeloid leukemia (AML). However, the expression of CD44v6 on T cells leads to transient fratricide and exhaustion of CD44v6 CAR-T cells, which affect the application of CD44v6 CAR-T. The exhaustion and function of T cells and CD44v6 expression of AML cells are associated with DNA methylation. Hypomethylating agents (HAMs) decitabine (Dec) and azacitidine (Aza) have been widely used to treat AML. Therefore, there may be synergy between CD44v6 CAR-T cells and HAMs in the treatment of AML. Methods: CD44v6 CAR-T cells pretreated with Dec or Aza were co-cultured with CD44v6+ AML cells. Dec or aza pretreated AML cells were co-cultured with CD44v6 CAR-T cells. The cytotoxicity, exhaustion, differentiation and transduction efficiency of CAR-T cells, and CD44v6 expression and apoptosis in AML cells were detected by flow cytometry. The subcutaneous tumor models were used to evaluate the anti-tumor effect of CD44v6 CAR-T cells combined with Dec in vivo. The effects of Dec or Aza on gene expression profile of CD44v6 CAR-T cells were analyzed by RNA-seq. Results: Our results revealed that Dec and Aza improved the function of CD44v6 CAR-T cells through increasing the absolute output of CAR+ cells and persistence, promoting activation and memory phenotype of CD44v6 CAR-T cells, and Dec had a more pronounced effect. Dec and Aza promoted the apoptosis of AML cells, particularly with DNA methyltransferase 3A (DNMT3A) mutation. Dec and Aza also enhanced the CD44v6 CAR-T response to AML by upregulating CD44v6 expression of AML cells regardless of FMS-like tyrosine kinase 3 (FLT3) or DNMT3A mutations. The combination of Dec or Aza pretreated CD44v6 CAR-T with pretreated AML cells demonstrated the most potent anti-tumor ability against AML. Conclusion: Dec or Aza in combination with CD44v6 CAR-T cells is a promising combination therapy for AML patients.


Assuntos
Leucemia Mieloide Aguda , Receptores de Antígenos Quiméricos , Humanos , Receptores de Antígenos Quiméricos/genética , Linhagem Celular Tumoral , Leucemia Mieloide Aguda/genética , Azacitidina/farmacologia , Azacitidina/uso terapêutico , Metilases de Modificação do DNA , Linfócitos T
3.
Small ; 19(36): e2302194, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37118855

RESUMO

The α-phase formamidinium lead tri-iodide (α-FAPbI3 ) has become the most promising photovoltaic absorber for perovskite solar cells (PSCs) due to its outstanding semiconductor properties and astonishing high efficiency. However, the incomplete crystallization and phase transition of α-FAPbI3 substantially undermine the performance and stability of PSCs. In this work, a series of the protic amine carboxylic acid ion liquids are introduced as the precursor additives to efficiently regulate the crystal growth and phase transition processes of α-FAPbI3 . The MA2 Pb3 I8 ·2DMSO phase is inhibited in annealing process, which remarkably optimizes the phase transition process of α-FAPbI3 . It is noted that the functional groups of carboxyl and ammonium passivate the undercoordinated lead ions, halide vacancies, and organic vacancies, eliminating the deleterious nonradiative recombination. Consequently, the small-area devices incorporated with 2% methylammonium butyrate (MAB) and 1.5% n-butylammonium formate (BAFa) in perovskite show champion efficiencies of 25.10% and 24.52%, respectively. Furthermore, the large-area modules (5 cm × 5 cm) achieve PCEs of 21.26% and 19.27% for MAB and BAFa additives, indicating the great potential for commercializing large-area PSCs.

4.
Front Immunol ; 14: 1063454, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36761742

RESUMO

Chimeric antigen receptor T-cell (CAR-T-cell) therapy has been well researched to date because of its ability to target malignant tumor cells. The most common CAR-T cells are CD19 CAR-T cells, which play a large role in B-cell leukemia treatment. However, most CAR-T cells are associated with relapse after clinical treatment, so the quality and persistence of CAR-T cells need to be improved. With continuous optimization, there have been four generations of CARs and each generation of CARs has better quality and durability than the previous generation. In addition, it is important to increase the proportion of memory cells in CAR-T cells. Studies have shown that an immunosuppressive tumor microenvironment (TME) can lead to dysfunction of CAR-T cells, resulting in decreased cell proliferation and poor persistence. Thus, overcoming the challenges of immunosuppressive molecules and targeting cytokines in the TME can also improve CAR-T cell persistence. In this paper, we explored how to improve the durability of CAR-T cell therapy by improving the structure of CARs, increasing the proportion of memory CAR-T cells and improving the TME.


Assuntos
Receptores de Antígenos Quiméricos , Receptores de Antígenos Quiméricos/genética , Imunoterapia Adotiva/métodos , Antígenos CD19 , Microambiente Tumoral , Terapia Baseada em Transplante de Células e Tecidos
5.
Polymers (Basel) ; 14(24)2022 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-36559914

RESUMO

Traditional hydrogels have drawbacks such as surgical implantation, large wound surfaces, and uncontrollable drug release during tumor treatment. In this paper, targeted nanomedicine has been combined with injectable hydrogel for photothermal-chemotherapy combination therapy. First, targeted nanomedicine (ICG-MTX) was fabricated by combining near-infrared (NIR) photothermal reagents (ICG) and chemotherapy drugs (MTX). The ICG-MTX was then mixed with the hydrogel precursor and radical initiator to obtain an injectable hydrogel precursor solution. Under the irradiation of NIR light, the precursor solution could release alkyl radicals, which promote the transition of the precursor solution from a liquid to a colloidal state. As a result, the nanomedicine could effectively remain at the site of the tumor and continue to be released from the hydrogel. Due to the targeted nature of MTX, the released ICG-MTX could target tumor cells and improve the accuracy of photothermal-chemo combination therapy. The results indicated that the injectable nanomedicine-hydrogel system has a favorable therapeutic effect on tumors.

6.
Clin Transl Med ; 12(9): e1043, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36163632

RESUMO

BACKGROUND: Chimeric antigen receptor T-cell (CAR-T) therapy for acute myeloid leukaemia (AML) has thus far been elusive, in part due to target restriction and phenotypic heterogeneity of AML cells. Mutations of the FMS-like tyrosine kinase 3 (FLT3) and DNA methyltransferase 3A (DNMT3A) genes are common driver mutations that present with a poor prognosis in AML patients. We found that AML patients with FLT3 or DNMT3A mutations had higher expression of CD44 isoform 6 (CD44v6) compared to normal specimens. Therefore, we intended to demonstrate CD44v6 could be a specific option for AML with FLT3 or DNMT3A mutations. METHODS: Internal tandem duplication (ITD) mutations of FLT3 (FLT3/ITD) knock-in clone and DNMT3A-R882H mutant clones of SKM-1 cells were generated using CRISPR/Cas9 and lentiviral transfection, respectively. CD44v6 CAR-T cells were constructed by transfecting T cells with lentivirus containing CD44v6 CAR. CD44v6 expression in AML cell lines, AML patients and healthy donors was evaluated by flow cytometry. DNA methylation assays were used to analyse the mechanisms of FLT3 and DNMT3A mutations affecting CD44v6 expression. RESULTS: Aberrant overexpression of CD44v6 was observed in AML cell lines with FLT3 or DNMT3A mutations compared to the wild-type SKM-1 or K562 cells. AML patients with FLT3 or DNMT3A mutations had higher expression of CD44v6 compared to normal specimens. Then we constructed CD44v6 CAR-T cells and found that CD44v6 CAR-T specifically lysed CD44v6+ cells, accompanied by cytokines release. No significant killing effect was observed from CD44v6- AML cells and normal cells after co-culture with CD44v6 CAR-T. These results were also observed in vivo. Furthermore, we found that FLT3 or DNMT3A mutations induced CD44v6 overexpression by downregulating the CpG methylation of CD44 promoter. CONCLUSIONS: Collectively, CD44v6 is a promising target of CAR-T for AML patients with FLT3 or DNMT3A mutations.


Assuntos
Leucemia Mieloide Aguda , Receptores de Antígenos Quiméricos , Citocinas/genética , DNA (Citosina-5-)-Metiltransferases/genética , DNA Metiltransferase 3A , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Mutação/genética , Receptores de Antígenos Quiméricos/genética , Especificidade do Receptor de Antígeno de Linfócitos T , Tirosina Quinase 3 Semelhante a fms/genética
7.
J Phys Chem Lett ; 13(14): 3188-3196, 2022 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-35377654

RESUMO

The inclusion of potassium in perovskite solar cells (PSCs) has been widely demonstrated to enhance the power conversion efficiency and eliminate the hysteresis effect. However, the effects of the locations K+ cations on the charge-carrier dynamics remain unknown with respect to achieving a more delicate passivation design for perovskite interfaces and bulk films. Herein, we employ the combined electrical and ultrafast dynamics analysis for the perovskite film to distinguish the effects of bulk doping and interfacial passivation of the potassium cation. Transient absorption spectroscopy indicates an enhancement of charge-carrier diffusion for K+-doped PSCs (from 808 to 605 ps), and charge-carrier transfer is significantly promoted by K+ interface passivation (from 12.34 to 1.23 ps) compared with that of the pristine sample. Importantly, K+ doping can suppress the formation of wide bandgap perovskite phases (e.g., FAPbI0.6Br2.4 and FAPbI1.05Br1.95) that generate an energy barrier on the charge-carrier transport channel.

8.
Small ; 17(42): e2102527, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34528387

RESUMO

The success of phototheranostics is hampered by some intrinsic defects, such as limited light penetration depth, heat resistance of tumor cells to photothermal therapy (PTT) induced by heat shock protein (HSP) and stress resistance against photodynamic therapy (PDT) caused by hypoxia microenvironment of tumor. Herein, a second near infrared (NIR-II) light excitation phototheranostic nanomedicine has been fabricated by integrating the semiconducting polymer, azo compound, and HSP inhibitor into a thermosensitive liposome, followed by modification with targeting aptamer, forming Lip(PTQ/GA/AIPH) for multimodal phototheranostics of triple-negative breast cancer (TNBC). The phototheranostic nanomedicine provides tumor targeting NIR-II fluorescence and photoacoustic dual-modal imaging, as well as NIR-II PTT. The released HSP inhibitor can effectively inhibit the activity of HSP for enhanced NIR-II PTT. Moreover, azo compound can be decomposed by the NIR-II photothermal activation, generating cytotoxic free radicals and realizing oxygen-irrelevant photonic thermodynamic therapy (PTDT) effects. Under the NIR-II laser irradiation, NIR-II fluorescence/photoacoustic dual-modal imaging guided enhanced NIR-II PTT and PTDT by Lip(PTQ/GA/AIPH), can achieve precise diagnosis and effective suppression of deep-seated TNBC with negligible side effects. This work develops a promising NIR-II excitation phototheranostic nanomedicine for spatiotemporally specific diagnosis and combination therapy of TNBC.


Assuntos
Nanopartículas , Neoplasias , Técnicas Fotoacústicas , Fotoquimioterapia , Linhagem Celular Tumoral , Fluorescência , Humanos , Nanomedicina , Neoplasias/tratamento farmacológico , Fototerapia , Nanomedicina Teranóstica , Termodinâmica , Microambiente Tumoral
9.
ACS Appl Bio Mater ; 4(10): 7595-7604, 2021 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-35006703

RESUMO

An injectable hydrogel sustained drug release system could be a promising technique for in situ treatment. Herein, an injectable hydrogel was prepared for photothermal-chemo therapy of cancer based on the thermosensitive liposomal hydrogel (Lip-Gel). The Lip-Gel system was fabricated by encapsulation of the NIR-II photothermal agent (DPP-BTz) and chemotherapy drugs (GEM) in thermosensitive liposomes and then combined with hydrogel precursor solution. The hydrogel precursor was used as an injectable flowing solution at room temperature and transferred into a cross-linked gel structure at physiological temperature. After being injected into the tumor, DPP-BTz in the Lip-Gel system can generate heat under irradiation of 1064 nm laser, breaking the thermosensitive liposomes and releasing GEM to kill tumor cells. From the treatment results, the Lip-Gel system showed a significant antitumor effect through chemo-/photothermal therapy combination therapy triggered by the NIR-II laser. This work provides a useful scheme for the development of drug delivery and drug treatment directions for local cancer therapy.


Assuntos
Hidrogéis , Neoplasias Pancreáticas , Humanos , Lipossomos , Neoplasias Pancreáticas/tratamento farmacológico , Fototerapia/métodos , Terapia Fototérmica , Neoplasias Pancreáticas
10.
ACS Appl Mater Interfaces ; 12(48): 53634-53645, 2020 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-33205657

RESUMO

The development of effective and safe tumor nanotheranostics remains a research imperative. Herein, tumor microenvironment (TME)-responsive Fe(III)-porphyrin (TCPP) coordination nanoparticles (FT@HA NPs) were prepared using a simple one-pot method followed by modification with hyaluronic acid (HA). FT@HA NPs specifically accumulated in CD44 receptor-overexpressed tumor tissues through the targeting property of HA and upon endocytosis by tumor cells. After cell internalization, intracellular acidic microenvironments and high levels of glutathione (GSH) triggered the rapid decomposition of FT@HA NPs to release free TCPP molecules and Fe(III) ions. The released Fe(III) ions could trigger GSH depletion and Fenton reaction, activating chemodynamic therapy (CDT). Meanwhile, the fluorescence and photodynamic effects of the TCPP could be also activated, achieving controlled reactive oxygen species (ROS) generation and avoiding side effects on normal tissues. Moreover, the rapid consumption of GSH further enhanced the efficacy of CDT and photodynamic therapy (PDT). The in vivo experiments further demonstrated that the antitumor effect of these nanotheranostics was significantly enhanced and that their toxicity and side effects against normal tissues were effectively suppressed. The FT@HA NPs can be applied for activated tumor combination therapy under the guidance of dual-mode imaging including fluorescence imaging and magnetic resonance imaging, providing an effective strategy for the design and preparation of TME-responsive multifunctional nanotheranostics for precise tumor imaging and combination therapy.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...