Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
J Nanobiotechnology ; 20(1): 190, 2022 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-35418151

RESUMO

BACKGROUND: Neoantigens are considered ideal targets for immunotherapy, especially tumor vaccine, because of their strong specificity and immunogenicity. Here, we developed a neoantigen nanovaccine used liposomes with lymph-node targeting characteristic. METHODS: Our nanovaccine was composed of neoantigens, an amphiphilic liposome and an adjuvant Montanide™ ISA 51. Small animal imaging system and immunofluorescence staining were used to identify the distribution of nanovaccines. A subcutaneous-tumor-resection mouse model of melanoma was established to evaluate the anti-tumor efficacy. Flow cytometry was performed to assay the immune responses initiated by nanovaccines. RESULTS: Nanovaccines could traffic to lymph nodes, be uptaken by CD11c+ DCs and promote DCs maturity. After the treatment of our neoantigen nanovaccines, the average recurrence time was extended from 11 to 16 days and the median survival time was even prolonged 7.5 days relative to the control group (NS group). Nanovaccines increased neoantigen-specific T cells to 10-fold of free vaccines, and upregulated Th1 cytokines, such as IFN-γ and TNF-α. The anti-tumor activity of spleen lymphocytes in the nanovaccine group was significantly stronger than that of other groups. However, some immune-inhibitory cells or molecules in tumor microenvironment have been detected upregulated under the immune pressure of neoantigen nanovaccines, such as Tregs and PD-L1. The efficacy of the neoantigen nanovaccine combined with anti-PD1 antibody or Treg inhibiting peptide P60 was better than that of the single treatment. CONCLUSIONS: We developed a general vaccine strategy, triggering specific T cell responses, and provided feasible combination strategies for better anti-tumor efficacy.


Assuntos
Vacinas Anticâncer , Melanoma , Animais , Antígenos de Neoplasias , Imunidade , Imunoterapia/métodos , Linfonodos , Melanoma/terapia , Camundongos , Microambiente Tumoral
2.
Cell Immunol ; 365: 104376, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33984534

RESUMO

Adoptive T cell transfer is one of the most promising ways to combat solid tumors. However, the weak infiltration of T cells into tumor sites has restricted their antitumor efficacy. To overcome this obstacle, we used the lipophilic protein painting strategy to improve tumor targeting and penetrating capacity of lymphocytes for the first time. We synthesized the lipid anchor consisting of a bispecific recombinant protein iRGD-antiEGFR and DSPE-PEG derivates, then successfully inserted it into the membranes of T cells. This surface modification was non-invasive and could efficiently improve the infiltration ability of T cells into multicellular spheroids and tumor masses. The surface modified T cells also displayed superior antitumor activities in EGFR-positive tumor xenografts via systematic infusion. Moreover, the permeability and antitumor efficacy of these surface painted T cells could be remarkably enhanced when used in combination with local low-dose irradiation.


Assuntos
Membrana Celular/metabolismo , Imunoterapia Adotiva/métodos , Linfócitos do Interstício Tumoral/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Anticorpos de Domínio Único/metabolismo , Neoplasias Gástricas/terapia , Linfócitos T/metabolismo , Animais , Linhagem Celular Tumoral , Receptores ErbB/imunologia , Receptores ErbB/metabolismo , Engenharia Genética , Humanos , Lipídeos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosfatidiletanolaminas , Polietilenoglicóis , Receptores de Antígenos de Linfócitos T/genética , Anticorpos de Domínio Único/genética , Neoplasias Gástricas/imunologia , Linfócitos T/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Acta Pharmacol Sin ; 39(4): 649-658, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29219946

RESUMO

4-(4-Pyridinyl methylene) curcumin (C1206) is a new derivative of curcumin that is more active than curcumin in inhibition of heat shock protein 90 (Hsp90) and antitumor action. In this study we investigated the relationship between C1206-induced inhibition of Hsp90 and its anti-leukemic effects. The fluorescence quenching experiments showed that C1206 seemed to bind the middle dimerization domain of Hsp90. The interaction between C1206 and Hsp90 was driven mainly by electrostatic interaction. In in vitro enzyme activity assay, C1206 dose-dependently inhibited Hsp90 ATPase activity with an IC50 value of 4.17 µmol/L. In both imatinib-sensitive K562 chronic myeloid leukemia cells and imatinib-resistant K562/G01 chronic myeloid leukemia cells, C1206 (0.4-3.2 µmol/L) dose-dependently caused the degradation of Hsp90 client proteins and downstream proteins (AKT, MEK, ERK, C-RAF, P-AKT, P-MEK and P-ERK). Furthermore, C1206 (0.4-3.2 µmol/L) dose-dependently induced apoptosis of K562 and K562/G01 cells through triggering mitochondrial pathway. Consistent with this result, C1206 inhibited the proliferation of K562 and K562/G01 cells with IC50 values of 1.10 and 0.60 µmol/L, respectively. These results suggest that C1206 is a novel Hsp90 inhibitor and a promising therapeutic agent for chronic myeloid leukemia.


Assuntos
Adenosina Trifosfatases/antagonistas & inibidores , Antineoplásicos/farmacologia , Curcumina/análogos & derivados , Curcumina/farmacologia , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Adenosina Trifosfatases/química , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Curcumina/química , Ensaios Enzimáticos , Proteínas de Choque Térmico HSP90/química , Humanos , Células K562 , Mitocôndrias/metabolismo , Pontos de Checagem da Fase S do Ciclo Celular/efeitos dos fármacos
4.
J Immunol ; 194(9): 4489-97, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25810397

RESUMO

Interstitial leukocyte migration plays a critical role in inflammation and offers a therapeutic target for treating inflammation-associated diseases such as multiple sclerosis. Identifying small molecules to inhibit undesired leukocyte migration provides promise for the treatment of these disorders. In this study, we identified vibsanin B, a novel macrocyclic diterpenoid isolated from Viburnum odoratissimum Ker-Gawl, that inhibited zebrafish interstitial leukocyte migration using a transgenic zebrafish line (TG:zlyz-enhanced GFP). We found that vibsanin B preferentially binds to heat shock protein (HSP)90ß. At the molecular level, inactivation of HSP90 can mimic vibsanin B's effect of inhibiting interstitial leukocyte migration. Furthermore, we demonstrated that vibsanin B ameliorates experimental autoimmune encephalomyelitis in mice with pathological manifestation of decreased leukocyte infiltration into their CNS. In summary, vibsanin B is a novel lead compound that preferentially targets HSP90ß and inhibits interstitial leukocyte migration, offering a promising drug lead for treating inflammation-associated diseases.


Assuntos
Movimento Celular/efeitos dos fármacos , Diterpenos/farmacologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Leucócitos/efeitos dos fármacos , Leucócitos/imunologia , Animais , Linhagem Celular , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Diterpenos/administração & dosagem , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/patologia , Feminino , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Humanos , Leucócitos/metabolismo , Camundongos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Medula Espinal/imunologia , Medula Espinal/metabolismo , Medula Espinal/patologia , Peixe-Zebra
5.
Mol Pharm ; 13(11): 3756-3763, 2016 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-27715057

RESUMO

Marine natural products are served as attractive source of anticancer therapeutics, with the great success of "first-in-class" drugs, such as Yondelis, Halaven, and Brentuximab vendotin. Lagunamides A-C from marine cyanobacterium, Lyngbya majuscula, exhibit exquisite growth inhibitory activities against cancer cells. In this study, we have systematically investigated the structure-activity relationships (SARs) of a concise collection of lagunamide A and its analogues constructed by total chemical synthesis against a broad panel of cancer cells derived from various tissues or organs, including A549, HeLa, U2OS, HepG2, BEL-7404, BGC-823, HCT116, MCF-7, HL-60, and A375. The R configuration of lagunamide A at C-39 position was found to be the structure determinant for anticancer activity. Further molecular mechanism study in A549 cells revealed that lagunamide A induced caspase-mediated mitochondrial apoptosis. Accompanied with the dissipation of mitochondrial membrane potential (Δφm) and overproduction of reactive oxygen species (ROS), lagunamide A led to mitochondrial dysfunction and finally caused cell death. Moreover, both anti- and pro-apoptotic B-cell lymphoma 2 (Bcl-2) family proteins participated in lagunamide A-induced mitochondrial apoptosis, especially myeloid cell leukemia-1 (Mcl-1). Overexpression of Mcl-1 partly rescued A549 cells from lagunamide A-induced apoptosis. This study suggests that lagunamide A may exert anticancer property through mitochondrial apoptosis. Together, our findings would provide insightful information for the design of new anticancer drugs derived from lagunamides.


Assuntos
Antineoplásicos/farmacologia , Depsipeptídeos/farmacologia , Células A549 , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Células HCT116 , Células HL-60 , Células HeLa , Células Hep G2 , Humanos , Células MCF-7 , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Microscopia Eletrônica de Transmissão , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/ultraestrutura , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Relação Estrutura-Atividade
6.
Anticancer Drugs ; 26(1): 25-34, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25014191

RESUMO

Heat shock protein 90 (Hsp90) functions within multiple signaling pathways on the basis of its ability to serve as a chaperone for more than 100 client proteins. Thus, inhibition of Hsp90 alone can trigger numerous pathways. Mycoepoxydiene (MED) can inhibit Hsp90 function and induce apoptosis in cervical cancer cells. However, the antitumor efficacy of MED in vivo is still not clear. We examined the efficacy of MED in a mouse xenograft model to further elucidate HeLa cell fate and also assessed the mechanism of altered protein signaling in response to this compound in vitro. Our data showed that Hsp90 inhibition simultaneously triggers signaling that regulates both cell death and cell proliferation, and that HeLa cell death may be a result of the disequilibrium of these signals. MED induces cell death as a result of the destabilization of Akt and IKK, which may promote cell death through a reduction in the activation of Bad and nuclear factor-κB. However, MED also induces the MEK/ERK pathway, which is classically considered to promote cell survival. MEK/ERK activation leads to an increase in p21, a cyclin-dependent kinase inhibitor, and is independent of Raf, but is shown to be mediated by p53. MED also leads to a decrease in several additional G2/M regulatory proteins independent of the MEK/ERK pathway. These results indicate an interesting mechanism of cross-talk between the inhibition of Akt phosphorylation and the activation of the MEK pathway by MED and provide in-vivo evidence for the potential of inhibiting Hsp90 as a candidate anticancer treatment.


Assuntos
Antineoplásicos/farmacologia , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Pironas/farmacologia , Neoplasias do Colo do Útero/patologia , Animais , Antineoplásicos/metabolismo , Antineoplásicos/uso terapêutico , Hidrocarbonetos Aromáticos com Pontes/metabolismo , Hidrocarbonetos Aromáticos com Pontes/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Proteínas de Choque Térmico HSP90/metabolismo , Xenoenxertos , Humanos , MAP Quinase Quinase Quinases/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pironas/metabolismo , Pironas/uso terapêutico , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Neoplasias do Colo do Útero/tratamento farmacológico
7.
Acta Biochim Biophys Sin (Shanghai) ; 47(6): 451-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25948110

RESUMO

Heat shock protein 90 (Hsp90) is a fascinating target for cancer therapy due to its significant role in the crossroad of multiple signaling pathways associated with cell proliferation and regulation. Hsp90 inhibitors have the potential to be developed into anti-cancer drugs. Here, we identified nicotinic-mycoepoxydiene (NMD), a structurally novel compound as Hsp90 inhibitor to perform the anti-tumor activity. The compound selectively bound to the Hsp90 N-terminal domain, and degraded the Hsp90 client protein Akt. The degradation of Akt detained Bad in non-phosphorylation form. NMD-associated apoptosis was characterized by the formation of fragmented nuclei, poly(ADP-ribose) polymerase cleavage, cytochrome c release, caspase-3 activation, and the increased proportion of sub-G1 phase cells. Interestingly, the apoptosis was accompanied with autophagy, by exhibiting the increased expression of LC-3 and the decrease of lysosome pH value. Our findings provide a novel cellular mechanism by which Hsp90 inhibitor adjusts cell apoptosis and autophagy in vitro, suggesting that NMD not only has a potential to be developed into a novel anti-tumor pharmaceutical, but also exhibits a new mechanism in regulating cancer cell apoptosis and autophagy via Hsp90 inhibition.


Assuntos
Apoptose/fisiologia , Autofagia/fisiologia , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Proteínas de Choque Térmico HSP90/fisiologia , Nicotina/química , Pironas/farmacologia , Hidrocarbonetos Aromáticos com Pontes/química , Cloroquina/farmacologia , Citocromos c/metabolismo , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Células HeLa , Humanos , Pironas/química , Transdução de Sinais
8.
Mol Cancer ; 13: 150, 2014 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-24927996

RESUMO

BACKGROUND: Heat shock protein 90 (Hsp90) is a promising therapeutic target and inhibition of Hsp90 will presumably result in suppression of multiple signaling pathways. FW-04-806, a bis-oxazolyl macrolide compound extracted from China-native Streptomyces FIM-04-806, was reported to be identical in structure to the polyketide Conglobatin. METHODS: We adopted the methods of chemproteomics, computational docking, immunoprecipitation, siRNA gene knock down, Quantitative Real-time PCR and xenograft models on the research of FW-04-806 antitumor mechanism, through the HER2-overexpressing breast cancer SKBR3 and HER2-underexpressing breast cancer MCF-7 cell line. RESULTS: We have verified the direct binding of FW-04-806 to the N-terminal domain of Hsp90 and found that FW-04-806 inhibits Hsp90/cell division cycle protein 37 (Cdc37) chaperone/co-chaperone interactions, but does not affect ATP-binding capability of Hsp90, thereby leading to the degradation of multiple Hsp90 client proteins via the proteasome pathway. In breast cancer cell lines, FW-04-806 inhibits cell proliferation, caused G2/M cell cycle arrest, induced apoptosis, and downregulated Hsp90 client proteins HER2, Akt, Raf-1 and their phosphorylated forms (p-HER2, p-Akt) in a dose and time-dependent manner. Importantly, FW-04-806 displays a better anti-tumor effect in HER2-overexpressed SKBR3 tumor xenograft model than in HER2-underexpressed MCF-7 model. The result is consistent with cell proliferation assay and in vitro apoptosis assay applied for SKBR-3 and MCF-7. Furthermore, FW-04-806 has a favorable toxicity profile. CONCLUSIONS: As a novel Hsp90 inhibitor, FW-04-806 binds to the N-terminal of Hsp90 and inhibits Hsp90/Cdc37 interaction, resulting in the disassociation of Hsp90/Cdc37/client complexes and the degradation of Hsp90 client proteins. FW-04-806 displays promising antitumor activity against breast cancer cells both in vitro and in vivo, especially for HER2-overexpressed breast cancer cells.


Assuntos
Neoplasias da Mama/genética , Proteínas de Ciclo Celular/genética , Chaperoninas/genética , Proteínas de Choque Térmico HSP90/genética , Macrolídeos/administração & dosagem , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Proteínas de Ciclo Celular/antagonistas & inibidores , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Chaperoninas/antagonistas & inibidores , Feminino , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Humanos , Células MCF-7 , Complexos Multiproteicos/antagonistas & inibidores , Complexos Multiproteicos/efeitos dos fármacos , Oxazóis/administração & dosagem , Ligação Proteica/efeitos dos fármacos , Receptor ErbB-2/genética
9.
Nat Chem Biol ; 8(11): 897-904, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22983157

RESUMO

Liver kinase B1 (LKB1) has important roles in governing energy homeostasis by regulating the activity of the energy sensor kinase AMP-activated protein kinase (AMPK). The regulation of LKB1 function, however, is still poorly understood. Here we demonstrate that the orphan nuclear receptor Nur77 binds and sequesters LKB1 in the nucleus, thereby attenuating AMPK activation. This Nur77 function is antagonized by the chemical compound ethyl 2-[2,3,4-trimethoxy-6-(1-octanoyl)phenyl]acetate (TMPA), which interacts with Nur77 with high affinity and at specific sites. TMPA binding of Nur77 results in the release and shuttling of LKB1 to the cytoplasm to phosphorylate AMPKα. Moreover, TMPA effectively reduces blood glucose and alleviates insulin resistance in type II db/db and high-fat diet- and streptozotocin-induced diabetic mice but not in diabetic littermates with the Nur77 gene knocked out. This study attains a mechanistic understanding of the regulation of LKB1-AMPK axis and implicates Nur77 as a new and amenable target for the design and development of therapeutics to treat metabolic diseases.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Fenilacetatos/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Animais , Glicemia/efeitos dos fármacos , Células Cultivadas , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Ativação Enzimática/efeitos dos fármacos , Células HEK293 , Humanos , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Modelos Moleculares , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/antagonistas & inibidores , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Fenilacetatos/química , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Transporte Proteico/efeitos dos fármacos , Estreptozocina , Relação Estrutura-Atividade
10.
Cancer Manag Res ; 16: 691-701, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38948681

RESUMO

Purpose: As one of the most important breakthroughs in cancer therapy, immune checkpoint inhibitors have greatly prolonged survival of patients with breast cancer. However, their application and efficacy are limited, especially for advanced HER2-negative breast cancer. It has been reported that epigenetic modulation of the histone deacetylase (HDAC) inhibitor chidamide, as well as immune microenvironment modulation of radiotherapy are potentially synergistic with immunotherapy. Thus, the combination of chidamide, radiotherapy and immunotherapy is expected to improve prognosis of patients with advanced HER2-negative breast cancer. Patients and Methods: This is a single-arm, open, prospective clinical trial investigating the efficacy and safety of the combination of HDAC inhibitor chidamide, anti-PD-1 antibody sintilimab, and the novel immuno-radiotherapy, which aims to enhance efficacy of immunotherapy, in subsequent lines of therapy of HER2-negative breast cancer. Our study will include 35 patients with advanced breast cancer that has failed endocrine therapy and first-line chemotherapy. Participants will receive 30 mg of chidamide twice a week, 200 mg of sintilimab once every 3 weeks, combined with immuno-radiotherapy. Radiotherapy will be centrally 8 Gy for at least one lesion, and at least 1 Gy for the other lesions. We will complete three fractions of radiotherapy in one cycle. The primary endpoint is progression-free survival, and secondary endpoints are objective response rate, disease control rate and safety. Moreover, biomarkers including cytokines and lymphocyte subgroups will be explored. Conclusion: As a single-arm clinical trial, the analysis of the influence of each single treatment is limited. Besides, our study is an open study, which involves neither randomization nor blinding. In spite of the abovementioned limitations, this prospective clinical trial will give an insight into subsequent lines of therapy of HER2-negative advanced breast cancer, prolong the survival or achieve long remission for these participants, and identify potential responders.

11.
Inflamm Res ; 62(2): 165-72, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23064679

RESUMO

OBJECTIVE: Gambogic acid (GBA) targeted Heat shock protein 90 (Hsp90) and prohibited TNF-α/NF-κB signaling pathway. It can be inferred that the anti-inflammatory activity of GBA results from inhibiting the cytokine production via NF-κB signaling pathway. We used the RAW264.7 cell line and the endotoxin shock mouse model to confirm the hypothesis that GBA protects mice from endotoxin shock by suppressing cytokine synthesis. METHOD: RAW264.7 cells were cultured and the endotoxin shocked mice model was constructed. ELISA was employed to evaluate the change of cytokine secretion levels. The effects of GBA on the activation of NF-κB signaling pathway were also determined by western blot and immune-fluorescent analysis. Cell viability was determined by MTT assay, and the cell migration was tested by wound healing assay. RESULT: Our results demonstrated that GBA significantly inhibited the LPS-induced release of pro-inflammatory factors both in cell lines and mice serum, thereby protecting mice from endotoxin shock. Furthermore, we observed that the reduction of inflammatory cytokines interleukin 1-beta, interleukin 6 and TNF-α resulted from the Hsp90's client protein IKK degradation and the suppression of NF-κB pathway. Moreover, GBA suppressed the migration of LPS-induced RAW264.7 cells. CONCLUSION: Our results indicate that GBA has a potential both as an antitumor and anti-inflammatory therapeutic agent.


Assuntos
Anti-Inflamatórios/uso terapêutico , Choque Séptico/tratamento farmacológico , Xantonas/uso terapêutico , Animais , Anti-Inflamatórios/farmacologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Citocinas/imunologia , Quinase I-kappa B/imunologia , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/imunologia , Choque Séptico/imunologia , Cicatrização , Xantonas/farmacologia
12.
J Cancer Res Clin Oncol ; 149(16): 15003-15011, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37610673

RESUMO

OBJECTIVE: Immunotherapy is an attractive treatment for breast cancer. Cancer-testis antigens (CTAs) are potential targets for immunotherapy for their restricted expression. Here, we investigate the expression of CTAs in breast cancer and their value for prognosis. So as to hunt for a potential panel of CTAs for universal immunotherapeutic targets. MATERIAL AND METHODS: A total of 137 breast cancer tissue specimens including 51 triple-negative breast cancer (TNBC) were assessed for MAGE-A4, MAGEA1, NY-ESO-1, KK-LC-1 and PRAME expression by immunohistochemistry. The expression of PD-L1 and TILs was also calculated and correlated with the five CTAs. Clinical data were collected to evaluate the CTA's value for prognosis. Data from the K-M plotter were used as a validation cohort. RESULTS: The expression of MAGE-A4, NY-ESO-1 and KK-LC-1 in TNBC was significantly higher than in non-TNBC (P = 0.012, P = 0.005, P < 0.001 respectively). 76.47% of TNBC expressed at least one of the five CTAs. Patients with positive expression of either MAGE-A4 or PRAME had a significantly extended disease-free survival (DFS). Data from the Kaplan-Meier plotter confirm our findings. CONCLUSIONS: MAGE-A4, NY-ESO-1, PRAME and KK-LC-1 are overexpressed in breast cancer, especially in TNBC. Positive expression of MAGE-A4 or PARME may be associated with prolonged DFS. A panel of CTAs is attractive universal targets for immunotherapy.


Assuntos
Neoplasias de Mama Triplo Negativas , Masculino , Humanos , Neoplasias de Mama Triplo Negativas/terapia , Neoplasias de Mama Triplo Negativas/metabolismo , Biomarcadores Tumorais/metabolismo , Testículo , Proteínas de Membrana/metabolismo , Antígenos de Neoplasias/metabolismo , Imunoterapia
13.
Biochem Biophys Res Commun ; 424(1): 100-4, 2012 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-22732400

RESUMO

AMP-activated protein kinase (AMPK) plays a key role in maintaining intracellular and whole-body energy homeostasis. Activation of AMPK has been shown to ameliorate the symptoms of metabolic diseases, such as type 2 diabetes and obesity. Here we show that gambogic acid (GB), a known antitumor agent, activates AMPK by increasing the phosphorylation of AMPKα and its downstream substrate ACC in various cell lines. Further study revealed that GB stimulated AMPK activity independent of upstream kinases. Moreover, the AMPK inhibitor, compound C, has no effects on the GB-induced AMPK activation. We also found that GB promptly increased intracellular ROS level, and antioxidants attenuated the ROS production. Interestingly, only the thiol antioxidants significantly abolished GB-enhanced AMPK activation. In addition, analysis of binding and dissociation kinetics indicated that GB bound to the AMPKα subunit. Collectively, these results suggest that GB may be a novel direct activator of AMPK.


Assuntos
Proteínas Quinases Ativadas por AMP/biossíntese , Antineoplásicos/farmacologia , Xantonas/farmacologia , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Animais , Linhagem Celular , Camundongos , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Espécies Reativas de Oxigênio/metabolismo
14.
Front Oncol ; 12: 905832, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35734599

RESUMO

Breast cancer remains the most frequently diagnosed malignancy worldwide. Advanced breast cancer is still an incurable disease mainly because of its heterogeneity and limited immunogenicity. The great success of cancer immunotherapy is paving the way for a new era in cancer treatment, and therapeutic cancer vaccination is an area of interest. Vaccine targets include tumor-associated antigens and tumor-specific antigens. Immune responses differ in different vaccine delivery platforms. Next-generation sequencing technologies and computational analysis have recently made personalized vaccination possible. However, only a few cases benefiting from neoantigen-based treatment have been reported in breast cancer, and more attention has been given to overexpressed antigen-based treatment, especially human epidermal growth factor 2-derived peptide vaccines. Here, we discuss recent advancements in therapeutic vaccines for breast cancer and highlight near-term opportunities for moving forward.

15.
Front Oncol ; 12: 900832, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36505863

RESUMO

Breast cancer is considered a malignant tumor with the highest incidence among women and is prone to develop distant metastasis. Small intestinal metastasis of breast cancer, however, is relatively rare. This case report describes a 49-year-old Chinese female patient who presented with small intestinal obstruction and was diagnosed with lobular breast cancer with small intestinal and contralateral breast metastasis. Clinical manifestations, clinicopathological features and potential mechanisms of metastasis, along with diagnosis and treatment, are discussed with a review of the relevant literature. Although small intestinal metastasis is rare in breast cancer, we should keep high alert on the possibility of gastrointestinal metastasis when treating lobular breast cancer patients.

16.
Biochem Biophys Res Commun ; 403(3-4): 282-7, 2010 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-21074517

RESUMO

Gambogic acid (GB) is an important anti-cancer drug candidate, but the target protein by which it exerts its anti-cancer effects has not been identified. This study is the first to show that GB inhibits heat shock protein 90 (Hsp90) and down-regulates TNF-α/NF-κB in HeLa cells. The effects of GB on Hsp90 were studied by characterizing its physical interactions with Hsp90 upon binding, the noncompetitive inhibition of Hsp90 ATPase activity, and the degradation of Hsp90 client proteins (i.e., Akt, IKK) in HeLa cells. GB seems to bind to the N-terminal ATP-binding domain of Hsp90. Additionally, GB suppresses the activation of TNF-α/NF-κB and decreases XIAP expression levels and the ratio of Bcl-2/Bax, which in turn induces HeLa cell apoptosis. Thus, GB represents a promising therapeutic agent for cancer; it may also be useful as a probe to increase understanding of the biological functions of Hsp90.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Xantonas/farmacologia , Adenosina Trifosfatases/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proteínas de Choque Térmico HSP90/química , Proteínas de Choque Térmico HSP90/metabolismo , Células HeLa , Humanos , Conformação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo , Xantonas/metabolismo
17.
Nat Chem Biol ; 4(9): 548-56, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18690216

RESUMO

Nuclear orphan receptor Nur77 has important roles in many biological processes. However, a physiological ligand for Nur77 has not been identified. Here, we report that the octaketide cytosporone B (Csn-B) is a naturally occurring agonist for Nur77. Csn-B specifically binds to the ligand-binding domain of Nur77 and stimulates Nur77-dependent transactivational activity towards target genes including Nr4a1 (Nur77) itself, which contains multiple consensus response elements allowing positive autoregulation in a Csn-B-dependent manner. Csn-B also elevates blood glucose levels in fasting C57 mice, an effect that is accompanied by induction of multiple genes involved in gluconeogenesis. These biological effects were not observed in Nur77-null (Nr4a1-/-) mice, which indicates that Csn-B regulates gluconeogenesis through Nur77. Moreover, Csn-B induced apoptosis and retarded xenograft tumor growth by inducing Nur77 expression, translocating Nur77 to mitochondria to cause cytochrome c release. Thus, Csn-B may represent a promising therapeutic drug for cancers and hypoglycemia, and it may also be useful as a reagent to increase understanding of Nur77 biological function.


Assuntos
Antineoplásicos , Proteínas de Ligação a DNA/agonistas , Gluconeogênese/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Fenilacetatos , Receptores de Esteroides/agonistas , Animais , Antineoplásicos/isolamento & purificação , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Ascomicetos/química , Glicemia/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Gluconeogênese/genética , Humanos , Ligantes , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Modelos Moleculares , Transplante de Neoplasias , Neoplasias/metabolismo , Neoplasias/patologia , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares , Fenilacetatos/isolamento & purificação , Fenilacetatos/farmacologia , Fenilacetatos/uso terapêutico , Ligação Proteica , Transporte Proteico , Receptores de Esteroides/biossíntese , Receptores de Esteroides/genética , Ativação Transcricional , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Cancer Manag Res ; 12: 863-870, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32104068

RESUMO

OBJECTIVE: The use of hepatic artery infusion (HAI) as a regional therapy against liver metastasis has rarely been reported in gastric cancer. This study aimed to evaluate the efficacy and safety of HAI oxaliplatin plus oral S-1 chemotherapy in first-line palliative therapy for gastric cancer with multiple liver metastases (GCLM). METHODS: We reviewed the records of five patients with GCLM who received HAI oxaliplatin (70-80 mg/m2 2 hrs d1,15) administered via a port-catheter system and S-1 with oral (35-40 mg/m2 twice daily for d1-14, 28 days for one cycle). Follow-up examination and efficacy evaluation were executed periodically. RESULTS: Until the 4th cycle response evaluation, the local effective rate and control rate were 40% and 80%, respectively; only one patient developed progression. HAI chemotherapy had a better local control against liver metastases (median progression-free survival: hepatic, 8.8 months vs. extrahepatic, 6.2 months), accompanied by less systemic toxicity, decreased tumour markers and symptomatic relief. CONCLUSION: HAI oxaliplatin plus oral S-1 chemotherapy can be considered as a new choice of first-line treatment for GCLM, which is also a good approach for controlling extrahepatic lesions with less adverse events.

19.
Cancer Manag Res ; 11: 8937-8945, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31802936

RESUMO

PURPOSE: Inhibition of heat shock protein 90 (Hsp90) can lead to degradation of multiple client proteins, which are involved in tumor progression. Elevated Hsp90 expression has been linked to poor prognosis in patients with non-small cell lung cancer (NSCLC). Discovery of effective drug is a promising strategy to improve patient survival. This study aims to investigate the synergistic antitumor mechanism of C086 combined with gefitinib in NSCLC cells in vitro. METHODS: The binding of C086, gefitinib, and the combinations to Hsp90 was characterized by fluorescence quenching experiments. The inhibition of A549 or NCI-H1975 cell proliferation and apoptosis by C086 and gefitinib as a single agent or in combinations were performed using CFSE staining assays, AnnexinV-APC/PI and Western blot. RESULTS: C086 alone or with gefitinib reduces proliferation and increases proapoptotic caspase activation of both wild-type and mutation NSCLC, with NCI-H1975 cells showing much greater sensitivity to C086 and the combinations than A549 cells. The combination of C086 and gefitinib showed synergistic reduction of EGFR expression and the downstream PI3K/Akt and Ras-Raf-Erk pathways enhanced suppression of Erk signaling. CONCLUSION: C086 combined gefitinib has a good synergistic antitumor effect in vitro. Therefore, the combination of C086 and gefitinib may provide a new theoretical basis and ideas for the treatment of NSCLC patients.

20.
Nat Commun ; 10(1): 1336, 2019 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-30902997

RESUMO

Poor infiltration of activated lymphocytes into tumors represents a fundamental factor limiting the therapeutic effect of adoptive cell immunotherapy. A tumor-penetrating peptide, iRGD, has been widely used to deliver drugs into tumor tissues. In this study, we demonstrate for the first time that iRGD could also facilitate the infiltration of lymphocytes in both 3D tumor spheroids and several xenograft mouse models. In addition, combining iRGD modification with PD-1 knockout lymphocytes reveals a superior anti-tumor efficiency. Mechanistic studies demonstrate that the binding of iRGD to neuropilin-1 results in tyrosine phosphorylation of the endothelial barrier regulator VE-cadherin, which plays a role in the opening of endothelial cell contacts and the promotion of transendothelial lymphocyte migration. In summary, these results demonstrate that iRGD modification could promote tumor-specific lymphocyte infiltration, and thereby overcome the bottleneck associated with adoptive immune cell therapy in solid tumors.


Assuntos
Técnicas de Inativação de Genes , Imunoterapia , Linfócitos/imunologia , Oligopeptídeos/uso terapêutico , Receptor de Morte Celular Programada 1/genética , Neoplasias Gástricas/imunologia , Neoplasias Gástricas/terapia , Animais , Antígenos CD/metabolismo , Caderinas/metabolismo , Linhagem Celular Tumoral , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Peritoneais/metabolismo , Neoplasias Peritoneais/patologia , Fosfatidiletanolaminas/química , Fosforilação , Fosfotirosina/metabolismo , Polietilenoglicóis/química , Receptor de Morte Celular Programada 1/metabolismo , Esferoides Celulares/metabolismo , Neoplasias Gástricas/patologia , Tela Subcutânea/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA