Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.225
Filtrar
1.
Cancer Med ; 12(12): 13573-13585, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37199371

RESUMO

OBJECTIVE: In order to ensure the stable transcription of target genes, we constructed a eukaryotic high expression vector carrying an immune-check inhibitor PD-1v and a variety of cytokines, and studied their effects on activating immune response to inhibit tumor growth. METHODS: A novel eukaryotic expression plasmid vector named pT7AMPCE containing T7RNA polymerase, T7 promoter, internal ribosome entry site (IRES), and poly A tailing signal was constructed by T4 DNA ligase, on which homologous recombination was used to clone and construct the vector carrying PD-1v, IL-2/15, IL-12, GM-CSF, and GFP. In vitro transfection of CT26 cells was performed, and the protein expression of PD-1v, IL-12 and GM-CSF was detected by Western blot and ELISA after 48 h. Mice were subcutaneously inoculated with CT26-IRFP tumor cells in the rib abdomen, and the tumor tissues were injected with PD-1v, IL-2/15, IL-12, and GM-CSF recombinant plasmids for treatment during the experimental period. The efficacy of the treatment was evaluated by assay tumor size and survival time of tumor-bearing mice during the experiment. Expression levels of IFN-γ, TNF, IL-4, IL-2, and IL-5 in mouse blood were measured using the CBA method. Tumor tissues were extracted and immune cell infiltration in tumor tissues was detected by HE staining and the IHC method. RESULTS: The recombinant plasmids carrying PD-1v, IL-2/15, IL-12, and GM-CSF were successfully constructed, and the Western blot and ELISA results showed that PD-1v, IL-12, and GM-CSF were expressed in the supernatant of CT26 cells 48 h after in vitro cell transfection. The combined application of PD-1v, IL-2/15, IL-12, and GM-CSF recombinant plasmids significantly inhibited tumor growth in mice, and the tumor growth rate was significantly lower than that in the blank control group and GFP plasmid control group (p < 0.05). Cytometric bead array data suggested that the combination of PD-1v and various cytokines can effectively activate immune cells. HE and IHC analysis revealed plenty of immune cell infiltrates in the tumor tissue, and a large proportion of tumor cells showed the necrotic phenotype in the combination treatment group. CONCLUSION: The combination of immune check blockade and multiple cytokine therapy can significantly activate the body's immune response and inhibit tumor growth.


Assuntos
Marcação de Genes , Vetores Genéticos , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Imunidade , Interleucina-12 , Neoplasias , Receptor de Morte Celular Programada 1 , Receptor de Morte Celular Programada 1/genética , Animais , Camundongos , Regiões Promotoras Genéticas , RNA Polimerases Dirigidas por DNA/genética , Proteínas Virais/genética , Interleucina-12/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Feminino , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Neoplasias/imunologia , Neoplasias/terapia , Transfecção , Imunidade/genética , Marcação de Genes/métodos , Proteínas de Fluorescência Verde
2.
Nat Immunol ; 24(5): 869-883, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37081150

RESUMO

To date, no immunotherapy approaches have managed to fully overcome T-cell exhaustion, which remains a mandatory fate for chronically activated effector cells and a major therapeutic challenge. Understanding how to reprogram CD8+ tumor-infiltrating lymphocytes away from exhausted effector states remains an elusive goal. Our work provides evidence that orthogonal gene engineering of T cells to secrete an interleukin (IL)-2 variant binding the IL-2Rßγ receptor and the alarmin IL-33 reprogrammed adoptively transferred T cells to acquire a novel, synthetic effector state, which deviated from canonical exhaustion and displayed superior effector functions. These cells successfully overcame homeostatic barriers in the host and led-in the absence of lymphodepletion or exogenous cytokine support-to high levels of engraftment and tumor regression. Our work unlocks a new opportunity of rationally engineering synthetic CD8+ T-cell states endowed with the ability to avoid exhaustion and control advanced solid tumors.


Assuntos
Linfócitos T CD8-Positivos , Imunoterapia Adotiva , Interleucina-2 , Neoplasias Experimentais , Linfócitos T CD8-Positivos/imunologia , Exaustão das Células T , Linfócitos do Interstício Tumoral/imunologia , Interleucina-2/farmacologia , Interleucina-33 , Engenharia de Proteínas , Feminino , Animais , Camundongos , Camundongos Endogâmicos C57BL , Linhagem Celular Tumoral , Neoplasias Experimentais/terapia , Receptor de Morte Celular Programada 1/metabolismo
3.
ACS Appl Mater Interfaces ; 14(45): 50592-50600, 2022 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-36334044

RESUMO

CpG, an agonist of toll-like receptor 9 (TLR9), has become a novel adjuvant that substantially potentiates cellular immunity. However, this agonist may increase systemic toxicity by diffusing into blood after administration and is difficult to be internalized by immune cells to reach TLR9 located in endosomes as a result of the characteristics of negative charge of CpG. Here, we applied a scalable and controllable flash nanocomplexation technology to prepare nanoparticulate CpG adjuvant (npCpG), CpG encapsulated in a physical cross-linking network of protamine and TPP. The nanoadjuvant could redirect CpG into draining lymph nodes to reduce systemic diffusion to improve safety. Further, a combination of npCpG and influenza H1N1 hemagglutinin antigen showed excellent humoral and cellular immunity, evoking high levels of antibodies and cytokines and inducing a great expansion of splenocytes in immunized mice. Also, the nanoadjuvant combined with ovalbumin antigen led to a potent cytotoxic T-cell response, substantially inhibited tumor growth, and improved the survival rate of mice in a melanoma model. This study showed the universal performances of npCpG in infectious disease prevention and tumor immunotherapy to demonstrate the translational potential.


Assuntos
Adjuvantes Imunológicos , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Vírus da Influenza A Subtipo H1N1 , Neoplasias Experimentais , Animais , Camundongos , Adjuvantes Imunológicos/farmacologia , Camundongos Endogâmicos C57BL , Oligodesoxirribonucleotídeos , Receptor Toll-Like 9/agonistas , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Ilhas de CpG , Nanopartículas , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia
4.
Biochem Biophys Res Commun ; 634: 114-121, 2022 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-36242917

RESUMO

Clostridium ghonii (C. ghonii) is a non-pathogenic Clostridium species and a strictly anaerobic, spore-forming bacterium. However, its bacterial oncolytic capabilities and applications have not yet been reported. This study aimed to determining the bacterial oncolytic capability of C. ghonii for the treatment of experimental solid tumors. C. ghonii secreted collagenase IV and phospholipase c and significantly promoted apoptosis and necrosis in cultured A549 cells. C. ghonii spores specially germinated and were distributed in the tumors, and elicited the immune responses after intratumoral injection in tumor-bearing mice. C. ghonii spores decreased tumor volumes and increased tumor necrosis and inhibition rates in tumor-bearing mice. Furthermore, the combination of radiation and C. ghonii exerted additive anti-tumor effects. Taken together, our data indicate that C. ghonii is a bacteriolytic therapeutic agent against solid tumors. Given the proven natural safety of C. ghonii, it is attractive as a potential novel bacteriolytic therapy for solid tumors.


Assuntos
Neoplasias Experimentais , Neoplasias , Camundongos , Animais , Esporos Bacterianos , Clostridium , Neoplasias/patologia , Neoplasias Experimentais/terapia , Necrose
5.
Exp Oncol ; 44(2): 155-158, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35964647

RESUMO

AIM: To study the antitumor and antimetastatic effects of B. subtilis IMV B-7724 lectin used in neoadjuvant and adjuvant settings in vivo. MATERIALS AND METHODS: Studies were performed on C57Bl/6J mice; Lewis lung carcinoma (LLC) was used as an experimental tumor. В. subtilis ІМV В-7724 lectin was administered to tumor-bearing mice or to mice which underwent surgical resection of the primary tumor. The lectin was injected subcutaneously, 10 times, at a single dose of 5 or 1 mg/kg of body weight. The standard indicators of tumor growth and metastasis were evaluated. RESULTS: Independently of the application settings, the lectin at a dose of 1 mg/kg of b.w. caused more pronounced effect than at a dose of 5 mg/kg of b.w. The administration of B. subtilis IMV B-7724 lectin to the mice with LLC in neoadjuvant setting did not cause notable antitumor effect but led to a significant decrease in the number and volume of lung metastases. The lectin administration in adjuvant setting significantly inhibited metastasis: the metastasis inhibition index reached 63.0% and 100% in the mice treated with the lectin at a dose of 5 mg/kg and 1 mg/kg respectively. The mean survival time of the treated animals significantly increased. CONCLUSION: A pronounced antimetastatic effect of B. subtilis IMV B-7724 lectin administered in an adjuvant setting was demonstrated.


Assuntos
Carcinoma Pulmonar de Lewis , Neoplasias Pulmonares , Neoplasias Experimentais , Animais , Bacillus subtilis , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Carcinoma Pulmonar de Lewis/patologia , Lectinas , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Experimentais/terapia
6.
Proc Natl Acad Sci U S A ; 119(24): e2202719119, 2022 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-35675425

RESUMO

Schwannomas are slow-growing benign neoplasms that develop throughout the body causing pain, sensory/motor dysfunction, and death. Because bacterial immunotherapy has been used in the treatment of some malignant neoplasms, we evaluated attenuated Salmonella typhimurium strains as immunotherapies for benign murine schwannomas. Several bacterial strains were tested, including VNP20009, a highly attenuated strain that was previously shown to be safe in human subjects with advanced malignant neoplasms, and a VNP20009 mutant that was altered in motility and other properties that included adherence and invasion of cultured mammalian cells. VNP20009 controlled tumor growth in two murine schwannoma models and induced changes in cytokine and immune effector cell profiles that were consistent with induction of enhanced innate and adaptive host immune responses compared with controls. Intratumoral (i.t.) injection of S. typhimurium led to tumor cell apoptosis, decreased tumor angiogenesis, and lower growth of the injected schwannoma tumors. Invasive VNP20009 was significantly more efficacious than was a noninvasive derivative in controlling the growth of injected tumors. Bacterial treatment apparently induced systemic antitumor immunity in that the growth of rechallenge schwannomas implanted following primary bacterial treatment was also reduced. Checkpoint programmed death-1 (PD-1) blockade induced by systemic administration of anti-PD-1 antibodies controlled tumor growth to the same degree as i.t. injection of S. typhimurium, and together, these two therapies had an additive effect on suppressing schwannoma growth. These experiments represent validation of a bacterial therapy for a benign neoplasm and support development of S. typhimurium VNP20009, potentially in combination with PD-1 inhibition, as a schwannoma immunotherapy.


Assuntos
Imunoterapia , Neurilemoma , Salmonella typhimurium , Animais , Apoptose , Humanos , Imunoterapia/métodos , Injeções Intralesionais , Camundongos , Neoplasias Experimentais/terapia , Neurilemoma/terapia , Receptor de Morte Celular Programada 1 , Salmonella typhimurium/genética
7.
Proc Natl Acad Sci U S A ; 119(7)2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35131941

RESUMO

Lysine-specific demethylase 6A (KDM6A), also named UTX, is frequently mutated in bladder cancer (BCa). Although known as a tumor suppressor, KDM6A's therapeutic potential in the metastasis of BCa remains elusive. It also remains difficult to fulfill the effective up-regulation of KDM6A levels in bladder tumor tissues in situ to verify its potential in treating BCa metastasis. Here, we report a mucoadhesive messenger RNA (mRNA) nanoparticle (NP) strategy for the intravesical delivery of KDM6A-mRNA in mice bearing orthotopic Kdm6a-null BCa and show evidence of KDM6A's therapeutic potential in inhibiting the metastasis of BCa. Through this mucoadhesive mRNA NP strategy, the exposure of KDM6A-mRNA to the in situ BCa tumors can be greatly prolonged for effective expression, and the penetration can be also enhanced by adhering to the bladder for sustained delivery. This mRNA NP strategy is also demonstrated to be effective for combination cancer therapy with other clinically approved drugs (e.g., elemene), which could further enhance therapeutic outcomes. Our findings not only report intravesical delivery of mRNA via a mucoadhesive mRNA NP strategy but also provide the proof-of-concept for the usefulness of these mRNA NPs as tools in both mechanistic understanding and translational study of bladder-related diseases.


Assuntos
Histona Desmetilases/farmacologia , Nanopartículas/química , Metástase Neoplásica/prevenção & controle , RNA Mensageiro/farmacologia , Adesividade , Administração Intravesical , Animais , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Terapia Genética , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Humanos , Camundongos , Camundongos Nus , Mucosa , Neoplasias Experimentais/terapia , RNA Mensageiro/administração & dosagem , RNA Mensageiro/metabolismo , Neoplasias da Bexiga Urinária
8.
Chem Commun (Camb) ; 58(19): 3202-3205, 2022 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-35174839

RESUMO

Low levels of immune infiltrates in the tumor milieu hinder the effectiveness of immunotherapy against immune-cold tumors. In the current work, a tumor-targeting drug delivery system composed of Endo-loaded platelets (Endo@PLT) was developed to relieve immunosuppression by achieving tumor vascular normalization. Endo@PLT reprogrammed the immunostimulatory phenotype, achieving excellent PD-1 immunotherapy in vivo.


Assuntos
Inibidores da Angiogênese/farmacologia , Neoplasias/imunologia , Neovascularização Patológica/imunologia , Microambiente Tumoral/imunologia , Inibidores da Angiogênese/química , Animais , Humanos , Terapia de Imunossupressão , Imunoterapia , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/terapia , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Neovascularização Patológica/terapia , Microambiente Tumoral/efeitos dos fármacos
9.
J Clin Invest ; 132(3)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35104810

RESUMO

PD-1 signaling on T cells is the major pathway that limits T cell immunity, but the efficacy of anti-PD-1 therapy has been limited to a small proportion of patients with advanced cancers. We fortuitously observed that anti-PD-1 therapy depends on IL-2 signaling, which raises the possibility that a lack of IL-2 limits anti-PD-1-induced effector T cell expansion. To selectively deliver IL-2 to PD-1+CD8+ tumor-infiltrating lymphocytes (TILs), we engineered a low-affinity IL-2 paired with anti-PD-1 (PD-1-laIL-2), which reduced affinity to peripheral Treg cells but enhanced avidity to PD-1+CD8+ TILs. PD-1-laIL-2 exerted better tumor control and lower toxicity than single or mixed treatments. Mechanistically, PD-1-laIL-2 could effectively expand dysfunctional and tumor-specific CD8+ T cells. Furthermore, we discovered that presumably dysfunctional PD-1+TIM3+ TILs are the dominant tumor-specific T cells responding to PD-1-laIL-2. Collectively, these results highlight that PD-1-laIL-2 can target and reactivate tumor-specific TILs for tumor regression as a unique strategy with stronger efficacy and lower toxicity.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Imunidade Celular/efeitos dos fármacos , Interleucina-2/farmacologia , Linfócitos do Interstício Tumoral/imunologia , Neoplasias Experimentais/imunologia , Receptor de Morte Celular Programada 1/imunologia , Animais , Imunidade Celular/genética , Interleucina-2/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Neoplasias Experimentais/genética , Neoplasias Experimentais/terapia , Receptor de Morte Celular Programada 1/genética
10.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35101971

RESUMO

Metastatic osteosarcoma has a poor prognosis with a 2-y, event-free survival rate of ∼15 to 20%, highlighting the need for the advancement of efficacious therapeutics. Chimeric antigen receptor (CAR) T-cell therapy is a potent strategy for eliminating tumors by harnessing the immune system. However, clinical trials with CAR T cells in solid tumors have encountered significant challenges and have not yet demonstrated convincing evidence of efficacy for a large number of patients. A major bottleneck for the success of CAR T-cell therapy is our inability to monitor the accumulation of the CAR T cells in the tumor with clinical-imaging techniques. To address this, we developed a clinically translatable approach for labeling CAR T cells with iron oxide nanoparticles, which enabled the noninvasive detection of the iron-labeled T cells with magnetic resonance imaging (MRI), photoacoustic imaging (PAT), and magnetic particle imaging (MPI). Using a custom-made microfluidics device for T-cell labeling by mechanoporation, we achieved significant nanoparticle uptake in the CAR T cells, while preserving T-cell proliferation, viability, and function. Multimodal MRI, PAT, and MPI demonstrated homing of the T cells to osteosarcomas and off-target sites in animals administered with T cells labeled with the iron oxide nanoparticles, while T cells were not visualized in animals infused with unlabeled cells. This study details the successful labeling of CAR T cells with ferumoxytol, thereby paving the way for monitoring CAR T cells in solid tumors.


Assuntos
Neoplasias Ósseas , Óxido Ferroso-Férrico/farmacologia , Imunoterapia Adotiva , Imageamento por Ressonância Magnética , Nanopartículas/uso terapêutico , Neoplasias Experimentais , Osteossarcoma , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Animais , Neoplasias Ósseas/diagnóstico por imagem , Neoplasias Ósseas/imunologia , Neoplasias Ósseas/terapia , Camundongos , Neoplasias Experimentais/diagnóstico por imagem , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Osteossarcoma/diagnóstico por imagem , Osteossarcoma/imunologia , Osteossarcoma/terapia
11.
Cancer Sci ; 113(3): 875-890, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34978120

RESUMO

T cells survival, proliferation, and anti-tumor response are closely linked to their mitochondrial health. Complement C1q binding protein (C1QBP) promotes mitochondrial fitness through regulation of mitochondrial metabolism and morphology. However, whether C1QBP regulates T cell survival, proliferation, and anti-tumor immune function remains unclear. Our data demonstrated that C1QBP knockdown induced the accumulation of reactive oxygen species (ROS) and the loss of mitochondrial membrane potential to impair T cell mitochondrial fitness. At the same time, C1QBP insufficiency reduced the recruitment of the anti-apoptotic proteins, including Bcl-2 and Bcl-XL, and repressed caspase-3 activation and poly (ADP-ribose) polymerase cleavage, which consequently accelerated the T cell apoptotic process. In contrast, C1QBP knockdown rendered T cells with relatively weaker proliferation due to the inhibition of AKT/mTOR signaling pathway. To investigate the exact role of C1QBP in anti-tumor response, C1QBP+/- and C1QBP+/+ mice were given a subcutaneous injection of murine MC38 cells. We found that C1QBP deficiency attenuated T cell tumor infiltration and aggravated tumor-infiltrating T lymphocytes (TIL) exhaustion. Moreover, we further clarified the potential function of C1QBP in chimeric antigen receptor (CAR) T cell immunotherapy. Our data showed that C1QBP+/- CAR T cells exhibited relatively weaker anti-tumor response than the corresponding C1QBP+/+ CAR T cells. Given that C1QBP knockdown impairs T cells' anti-apoptotic capacity, proliferation as well as anti-tumor immune function, development of the strategy for potentiation of T cells' mitochondrial fitness through C1QBP could potentially optimize the efficacy of the related immunotherapy.


Assuntos
Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Linfócitos T/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Humanos , Imunoterapia Adotiva , Linfócitos do Interstício Tumoral/citologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Camundongos , Proteínas Mitocondriais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/terapia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores de Antígenos Quiméricos , Transdução de Sinais , Linfócitos T/citologia , Linfócitos T/imunologia
12.
Molecules ; 27(2)2022 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-35056740

RESUMO

Theranostic agents for concurrent cancer therapy and diagnosis have begun attracting attention as a promising modality. However, accurate imaging and identification remains a great challenge for theranostic agents. Here, we designed and synthesized a novel theranostic agent H6M based on the "double-locked" strategy by introducing an electron-withdrawing nitro group into 1-position of a pH-responsive 3-amino-ß-carboline and further covalently linking the hydroxamic acid group, a zinc-binding group (ZBG), to the 3-position of ß-carboline to obtain histone deacetylase (HDAC) inhibitory effect for combined HDAC-targeted therapy. We found that H6M can be specifically reduced under overexpressed nitroreductase (NTR) to produce H6AQ, which emits bright fluorescence at low pH. Notably, H6M demonstrated a selective fluorescence imaging via successive reactions with NTR (first "key") and pH (second "key"), and precisely identified tumor margins with a high S/N ratio to guide tumor resection. Finally, H6M exerted robust HDAC1/cancer cell inhibitory activities compared with a known HDAC inhibitor SAHA. Therefore, the NTR/pH-activated theranostic agent provided a novel tool for precise diagnosis and efficient tumor therapy.


Assuntos
Antineoplásicos/farmacologia , Inibidores de Histona Desacetilases/química , Inibidores de Histona Desacetilases/farmacologia , Medicina de Precisão/métodos , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Feminino , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/diagnóstico por imagem , Neoplasias Experimentais/cirurgia , Neoplasias Experimentais/terapia , Nitrorredutases/metabolismo , Ratos , Espectrometria de Fluorescência , Cirurgia Assistida por Computador , Ensaios Antitumorais Modelo de Xenoenxerto
13.
J Mater Sci Mater Med ; 33(1): 10, 2022 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-35022850

RESUMO

Herein we report synthesis of hematite (α-Fe2O3) nanorods by calcinating hydrothermally synthesized goethite nanorods at 5000C. The structural, optical and MRI imaging guided cancer therapeutic properties of fabricated nanorods have been discussed in this manscript. FESEM and TEM imaging techniques were used to confirm the nanorod like morphology of as prepared materials. As we know that Fe2O3 nanorods with size in the range of 25-30 nm exhibit super magnetism. After coating with the PEG, the as prepared nanorods can be used as T2 MR imaging contrast agents. An excellent T2 MRI contrast of 38.763 mM-1s-1 achieved which is highest reported so far for α-Fe2O3. Besides the as prepared nanorods display an excellent photothermal conversion efficiency of 39.5% thus acts as an excellent photothermal therapeutic agent. Thus, we envision the idea of testing our nanorods for photothermal therapy and MR imaging application both in vitro and in vivo, achieving an excellent T2 MRI contrast and photothermal therapy effect with as prepared PEGylated nanorods.


Assuntos
Compostos Férricos/química , Nanotubos/química , Animais , Materiais Biocompatíveis/química , Linhagem Celular , Sobrevivência Celular , Feminino , Compostos Férricos/toxicidade , Células HeLa , Humanos , Técnicas In Vitro , Imageamento por Ressonância Magnética , Teste de Materiais , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Microscopia Eletrônica de Varredura , Nanotubos/toxicidade , Nanotubos/ultraestrutura , Neoplasias Experimentais/patologia , Neoplasias Experimentais/terapia , Fototerapia/métodos , Polietilenoglicóis/química , Análise Espectral Raman , Difração de Raios X
14.
ACS Appl Mater Interfaces ; 14(2): 2464-2477, 2022 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-35045602

RESUMO

Nanomedicine, constructed from therapeutics, presents an advantage in drug delivery for cancer therapies. However, nanocarrier-based treatment systems have problems such as interbatch variability, multicomponent complexity, poor drug delivery, and carrier-related toxicity. To solve these issues, the natural molecule honokiol (HK), an anticancer agent in a phase I clinical trial (CTR20170822), was used to form a self-assembly nanoparticle (SA) through hydrogen bonding and hydrophobicity. The preparation of SA needs no molecular precursors or excipients in aqueous solution, and 100% drug-loaded SA exhibited superior tumor-targeting ability due to the enhanced permeability and retention (EPR) effect. Moreover, SA significantly enhanced the antitumor immunity relative to free HK, and the mechanism has notable selectivity to the p53 pathway. Furthermore, SA exhibited excellent physiological stability and inappreciable toxicity. Taken together, this supramolecular self-assembly strategy provides a safe and "molecular economy" model for rational design of clinical therapies and is expected to promote targeted therapy of HK, especially in colorectal cancer patients with obvious p53 status.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Materiais Biocompatíveis/farmacologia , Compostos de Bifenilo/farmacologia , Neoplasias Colorretais/terapia , Imunoterapia , Lignanas/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Proteína Supressora de Tumor p53/antagonistas & inibidores , Animais , Antineoplásicos Fitogênicos/síntese química , Antineoplásicos Fitogênicos/química , Materiais Biocompatíveis/síntese química , Materiais Biocompatíveis/química , Compostos de Bifenilo/síntese química , Compostos de Bifenilo/química , Neoplasias Colorretais/imunologia , Feminino , Humanos , Lignanas/síntese química , Lignanas/química , Substâncias Macromoleculares/síntese química , Substâncias Macromoleculares/química , Teste de Materiais , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Estrutura Molecular , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Bibliotecas de Moléculas Pequenas/síntese química , Bibliotecas de Moléculas Pequenas/química , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/imunologia
15.
ACS Appl Mater Interfaces ; 14(1): 452-463, 2022 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-34961307

RESUMO

Trastuzumab combined with chemotherapy is the first-line treatment for advanced HER2-positive gastric cancer, but it still suffers from limited therapeutic efficiency and serious side effects, which are usually due to the poor delivery efficiency and the drug resistance of tumor cells to the chemotherapeutic drugs. Herein, a type of ultrasound microbubble for simultaneous delivery of sonosensitizers and therapeutic antibodies to achieve targeting combination of sonodynamic therapy and antibody therapy of HER2-positive gastric cancer was constructed from pyropheophorbide-lipid followed by trastuzumab conjugation (TP MBs). In vitro and in vivo studies showed that TP MBs had good biological safety, and their in vivo delivery can be monitored by ultrasound/fluorescence bimodal imaging. With ultrasound (US) located at the tumor area, TP MBs can be converted into nanoparticles (TP NPs) in situ by US-targeted microbubble destruction; plus the enhanced permeability and retention effects and the targeting effects of trastuzumab, the enrichment of sonosensitizers and antibodies in the tumor tissue can be greatly enhanced (∼2.1 times). When combined with ultrasound, TP MBs can not only increase the uptake of sonosensitizers in HER2-positive gastric cancer NCI-N87 cells but also efficiently generate singlet oxygen to greatly increase the killing effect on cells, obviously inhibiting the tumor growth in HER2-positive gastric cancer NCI-N87 cell models with a tumor inhibition rate up to 79.3%. Overall, TP MBs combined with US provided an efficient way for co-delivery of sonosensitizers and antibodies, greatly enhancing the synergistic therapeutic effect on HER2-positive gastric cancer while effectively reducing the side effects.


Assuntos
Anticorpos/farmacologia , Antineoplásicos Imunológicos/farmacologia , Materiais Biocompatíveis/farmacologia , Receptor ErbB-2/antagonistas & inibidores , Neoplasias Gástricas/terapia , Trastuzumab/farmacologia , Terapia por Ultrassom , Animais , Anticorpos/química , Antineoplásicos Imunológicos/química , Materiais Biocompatíveis/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Teste de Materiais , Camundongos , Camundongos Nus , Microbolhas , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/terapia , Receptor ErbB-2/metabolismo , Neoplasias Gástricas/metabolismo , Trastuzumab/química , Ondas Ultrassônicas
16.
Proc Natl Acad Sci U S A ; 119(1)2022 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-34969858

RESUMO

Brain metastases are a leading cause of death in patients with breast cancer. The lack of clinical trials and the presence of the blood-brain barrier limit therapeutic options. Furthermore, overexpression of the human epidermal growth factor receptor 2 (HER2) increases the incidence of breast cancer brain metastases (BCBM). HER2-targeting agents, such as the monoclonal antibodies trastuzumab and pertuzumab, improved outcomes in patients with breast cancer and extracranial metastases. However, continued BCBM progression in breast cancer patients highlighted the need for novel and effective targeted therapies against intracranial metastases. In this study, we engineered the highly migratory and brain tumor tropic human neural stem cells (NSCs) LM008 to continuously secrete high amounts of functional, stable, full-length antibodies against HER2 (anti-HER2Ab) without compromising the stemness of LM008 cells. The secreted anti-HER2Ab impaired tumor cell proliferation in vitro in HER2+ BCBM cells by inhibiting the PI3K-Akt signaling pathway and resulted in a significant benefit when injected in intracranial xenograft models. In addition, dual HER2 blockade using anti-HER2Ab LM008 NSCs and the tyrosine kinase inhibitor tucatinib significantly improved the survival of mice in a clinically relevant model of multiple HER2+ BCBM. These findings provide compelling evidence for the use of HER2Ab-secreting LM008 NSCs in combination with tucatinib as a promising therapeutic regimen for patients with HER2+ BCBM.


Assuntos
Antineoplásicos Imunológicos/metabolismo , Neoplasias Encefálicas , Neoplasias Experimentais , Células-Tronco Neurais , Oxazóis/farmacologia , Piridinas/farmacologia , Quinazolinas/farmacologia , Receptor ErbB-2 , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Nus , Metástase Neoplásica , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Neoplasias Experimentais/terapia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Células-Tronco Neurais/transplante , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Cancer Sci ; 113(3): 864-874, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34971473

RESUMO

NY-ESO-1 is a cancer/testis antigen expressed in various cancer types. However, the induction of NY-ESO-1-specific CTLs through vaccines is somewhat difficult. Thus, we developed a new type of artificial adjuvant vector cell (aAVC-NY-ESO-1) expressing a CD1d-NKT cell ligand complex and a tumor-associated antigen, NY-ESO-1. First, we determined the activation of invariant natural killer T (iNKT) and natural killer (NK) cell responses by aAVC-NY-ESO-1. We then showed that the NY-ESO-1-specific CTL response was successfully elicited through aAVC-NY-ESO-1 therapy. After injection of aAVC-NY-ESO-1, we found that dendritic cells (DCs) in situ expressed high levels of costimulatory molecules and produced interleukn-12 (IL-12), indicating that DCs undergo maturation in vivo. Furthermore, the NY-ESO-1 antigen from aAVC-NY-ESO-1 was delivered to the DCs in vivo, and it was presented on MHC class I molecules. The cross-presentation of the NY-ESO-1 antigen was absent in conventional DC-deficient mice, suggesting a host DC-mediated CTL response. Thus, this strategy helps generate sufficient CD8+ NY-ESO-1-specific CTLs along with iNKT and NK cell activation, resulting in a strong antitumor effect. Furthermore, we established a human DC-transferred NOD/Shi-scid/IL-2γcnull immunodeficient mouse model and showed that the NY-ESO-1 antigen from aAVC-NY-ESO-1 was cross-presented to antigen-specific CTLs through human DCs. Taken together, these data suggest that aAVC-NY-ESO-1 has potential for harnessing innate and adaptive immunity against NY-ESO-1-expressing malignancies.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Antígenos de Neoplasias/administração & dosagem , Vacinas Anticâncer/administração & dosagem , Células Dendríticas/imunologia , Imunoterapia/métodos , Proteínas de Membrana/administração & dosagem , Adjuvantes Imunológicos/metabolismo , Animais , Antígenos CD1d/imunologia , Antígenos CD1d/metabolismo , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/metabolismo , Apresentação Cruzada , Células HEK293 , Humanos , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Células NIH 3T3 , Células T Matadoras Naturais/imunologia , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Linfócitos T Citotóxicos/imunologia
18.
Bioorg Chem ; 119: 105549, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34929517

RESUMO

Ecto-nucleotide pyrophosphatase/phosphodiesterases 1 (ENPP1 or NPP1), is an attractive therapeutic target for various diseases, primarily cancer and mineralization disorders. The ecto-enzyme is located on the cell surface and has been implicated in the control of extracellular levels of nucleotide, nucleoside and (di) phosphate. Recently, it has emerged as a critical phosphodiesterase that hydrolyzes cyclic 2'3'- cGAMP, the endogenous ligand for STING (STimulator of INterferon Genes). STING plays an important role in innate immunity by activating type I interferon in response to cytosolic 2'3'-cGAMP. ENPP1 negatively regulates the STING pathway and hence its inhibition makes it an attractive therapeutic target for cancer immunotherapy. Herein, we describe the design, optimization and biological evaluation studies of a series of novel non-nucleotidic thioguanine based small molecule inhibitors of ENPP1. The lead compound 43 has shown good in vitro potency, stability in SGF/SIF/PBS, selectivity, ADME properties and pharmacokinetic profile and finally potent anti-tumor response in vivo. These compounds are a good starting point for the development of potentially effective cancer immunotherapy agents.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Imunoterapia , Neoplasias Pulmonares/terapia , Pirofosfatases/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Tioguanina/farmacologia , Células A549 , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Relação Dose-Resposta a Droga , Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Feminino , Humanos , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Molecular , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/terapia , Diester Fosfórico Hidrolases/metabolismo , Pirofosfatases/metabolismo , Bibliotecas de Moléculas Pequenas/síntese química , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade , Tioguanina/síntese química , Tioguanina/química
19.
Nat Commun ; 12(1): 7264, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34907171

RESUMO

Antibodies targeting costimulatory receptors of T cells have been developed for the activation of T cell immunity in cancer immunotherapy. However, costimulatory molecule expression is often lacking in tumor-infiltrating immune cells, which can impede antibody-mediated immunotherapy. Here, we hypothesize that delivery of costimulatory receptor mRNA to tumor-infiltrating T cells will enhance the antitumor effects of antibodies. We first design a library of biomimetic nanoparticles and find that phospholipid nanoparticles (PL1) effectively deliver costimulatory receptor mRNA (CD137 or OX40) to T cells. Then, we demonstrate that the combination of PL1-OX40 mRNA and anti-OX40 antibody exhibits significantly improved antitumor activity compared to anti-OX40 antibody alone in multiple tumor models. This treatment regimen results in a 60% complete response rate in the A20 tumor model, with these mice being resistant to rechallenge by A20 tumor cells. Additionally, the combination of PL1-OX40 mRNA and anti-OX40 antibody significantly boosts the antitumor immune response to anti-PD-1 + anti-CTLA-4 antibodies in the B16F10 tumor model. This study supports the concept of delivering mRNA encoding costimulatory receptors in combination with the corresponding agonistic antibody as a strategy to enhance cancer immunotherapy.


Assuntos
Materiais Biomiméticos/administração & dosagem , Imunoterapia/métodos , Linfócitos do Interstício Tumoral/imunologia , Nanopartículas/administração & dosagem , RNA Mensageiro/administração & dosagem , Linfócitos T/imunologia , Animais , Materiais Biomiméticos/química , Sistemas de Liberação de Medicamentos , Glicolipídeos/administração & dosagem , Glicolipídeos/química , Linfócitos do Interstício Tumoral/metabolismo , Camundongos , Nanopartículas/química , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Fosfolipídeos/administração & dosagem , Fosfolipídeos/química , RNA Mensageiro/química , Receptores OX40/antagonistas & inibidores , Receptores OX40/genética , Receptores OX40/imunologia , Receptores OX40/metabolismo , Linfócitos T/metabolismo , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/antagonistas & inibidores , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo
20.
Biomater Sci ; 10(1): 258-269, 2021 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-34850790

RESUMO

Combination therapy has been widely studied due to its promising applications in tumor therapy. However, a sophisticated nanoplatform and sequential irradiation with different laser sources for phototherapy complicate the treatment process. Unlike the integration of therapeutic agents, we report a FeS2@SRF@BSA nanoplatform for the combination of chemo-combined photothermal therapy (PTT) enhanced photodynamic therapy (PDT) and chemodynamic therapy (CDT) to achieve an "all-in-one" therapeutic agent. Ultrasmall FeS2 nanoparticles (NPs) with a size of 7 nm exhibited higher Fenton reaction rates due to their large specific surface areas. A photodynamic reaction could be triggered and could generate 1O2 to achieve PDT under 808 nm irradiation. FeS2 NPs also exhibited the desired photothermal properties under the same wavelength of the laser. The Fenton reaction and photodynamic reaction were both significantly improved to accumulate more reactive oxygen species (ROS) with an increase of temperature under laser irradiation. Besides, loading of the chemotherapeutic drug sorafenib (SRF) further improved the efficacy of tumor treatment. To realize long blood circulation, bovine serum albumin (BSA) was used as a carrier to encapsulate FeS2 NPs and SRF, remarkably improving the biocompatibility and tumor enrichment ability of the nanomaterials. Additionally, the tumors on mice treated with FeS2@SRF@BSA almost disappeared under 808 nm irradiation. To sum up, FeS2@SRF@BSA NPs possess good biocompatibility, stability, and sufficient therapeutic efficacy in combination therapy for cancer treatment. Our study pointed out a smart design of the nanoplatform as a multifunctional therapeutic agent for combination cancer therapy in the near future.


Assuntos
Nanopartículas , Neoplasias Experimentais/terapia , Fotoquimioterapia , Terapia Fototérmica , Animais , Linhagem Celular Tumoral , Camundongos , Soroalbumina Bovina , Sorafenibe
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...