Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cancer Immunol Immunother ; 72(2): 409-425, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35925286

RESUMO

Recently, chimeric antigen receptor T cell (CAR-T) therapy has received increasing attention as an adoptive cellular immunotherapy that targets tumors. However, numerous challenges remain for the effective use of CAR-T to treat solid tumors, including ovarian cancer, which is an aggressive and metastatic cancer with a poor therapeutic response. We screened for an effective anti-MSLN single-chain Fv antibody with comparable binding activity and non-off-target properties using human phage display library. A second-generation of anti-MSLN CAR was designed and generated. We demonstrated the efficacy of our anti-MSLN CAR-T cells for ovarian cancer treatment in an in vitro experiment to kill ovarian tumor cell lines. The anti-MSLN CAR-T cells impeded MSLN-positive tumor growth concomitant with a significant increase in cytokine levels compared with the control. Then, we demonstrated the efficacy of anti-MSLN CAR-T cells in an in vivo experiment against ovarian cancer cell-derived xenografts. Furthermore, we herein report three cases with ovarian cancer who were treated with autologous anti-MSLN CAR-T cells and evaluate the safety and effectiveness of adoptive cell therapy. In this investigator-initiated clinical trials, no patients experienced cytokine release syndrome or neurological symptoms over 2 grads. Disease stabilized in two patients, with progression-free survival times of 5.8 and 4.6 months. Transient CAR expression was detected in patient blood after infusion each time. The tumor partially subsided, and the patient's condition was relieved. In conclusion, this work proves the efficacy of the anti-MSLN CAR-T treatment strategy in ovarian cancer and provides preliminary data for the development of further clinical trials.


Assuntos
Imunoterapia Adotiva , Neoplasias Ovarianas , Receptores de Antígenos Quiméricos , Feminino , Humanos , Linhagem Celular Tumoral , Proteínas Ligadas por GPI , Imunoterapia , Neoplasias Ovarianas/terapia , Animais
2.
Gene Ther ; 29(7-8): 407-417, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-33414522

RESUMO

Camptothecin has been used in tumor therapy for a long time but its antitumor effect is rather limited due to the side effect and the drug resistance. FEN1, a major component of DNA repair systems, plays important roles in maintaining genomic stability via DNA replication and repair. Here we found that FEN1 inhibitor greatly sensitizes cancer cells to low-dose camptothecin. The combinative treatment of FEN1 inhibitor and 1 nM camptothecin induced a synthetic lethal effect, which synergistically suppressed cancer cell proliferation and significantly mediated apoptosis both in vitro and in vivo. Our study suggested that targeting FEN1 could be a potent strategy for tumor-targeting cancer therapy.


Assuntos
Camptotecina , Endonucleases Flap , Neoplasias , Apoptose , Camptotecina/farmacologia , Dano ao DNA , Endonucleases Flap/antagonistas & inibidores , Humanos , Mitocôndrias/metabolismo
3.
Photodermatol Photoimmunol Photomed ; 38(6): 522-530, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35175655

RESUMO

BACKGROUND: Ultraviolet (UV) radiation-induced oxidative stress is the main cause of photodamage to the skin. Glutathione (GSH) serves important physiological functions, including scavenging oxygen-free radicals and maintaining intracellular redox balance. γ-glutamylcysteine (γ-GC), as an immediate precursor of GSH and harboring antioxidant and anti-inflammatory properties, represents an unexplored option for skin photodamage treatment. PURPOSE: The purpose of this study was to investigate whether γ-GC can reduce UVB-induced NIH-3T3 cell damage. METHODS: The experimental groups were as follows: control, UVB radiation, UVB radiation after pretreatment with γ-GC. Cell counting kit-8 (CCK-8) assays were used to measure cell proliferation, flow cytometry, and immunoblotting to detect the apoptosis rate and apoptosis-associated proteins. The levels of Reactive Oxygen Species (ROS), Superoxide Dismutase (SOD), and GSH/GSSG (oxidized GSH) were measured to assess oxidative stress. Immunoblotting and immunofluorescence were used to detect DNA damage. The members of the MAPK signaling pathways were detected by immunoblotting. RESULTS: UVB irradiation significantly reduced cell viability and destroyed the oxidative defense system. Pretreatment with γ-GC reduced UVB-induced cytotoxicity, restored the oxidation defense system, and inhibited activation of the MAPK pathway. It also reduced the apoptosis rate, downregulated the levels of cleaved caspase 3 and cleaved PARP. Furthermore, pretreatment with γ-GC reduced the accumulation of γH2AX after UVB radiation exposure, indicating that γ-GC could protect cells from DNA damage. CONCLUSION: γ-GC protected NIH-3T3 from damage caused by UVB irradiation. The photoprotective effect of γ-GC is mediated via strengthening the endogenous antioxidant defense system, which prevents DNA damage and inhibits the activation of the MAPK pathway.


Assuntos
Estresse Oxidativo , Raios Ultravioleta , Humanos , Camundongos , Animais , Células NIH 3T3 , Raios Ultravioleta/efeitos adversos , Dipeptídeos/metabolismo , Dipeptídeos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Antioxidantes/farmacologia , Glutationa/metabolismo , Apoptose
4.
J Transl Med ; 18(1): 295, 2020 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-32746924

RESUMO

BACKGROUND: Treatment of hepatocellular carcinoma (HCC) using antibody-based targeted therapies, such as antibody conjugates and chimeric antigen receptor T (CAR-T) cell therapy, shows potent antitumor efficacy. Glypican-3 (GPC3) is an emerging HCC therapeutic target; therefore, antibodies against GPC3 would be useful tools for developing immunotherapies for HCC. METHODS: We isolated a novel human monoclonal antibody, 32A9, by phage display technology. We determined specificity, affinity, epitope and anti-tumor activity of 32A9, and developed 32A9-based immunotherapy technologies for evaluating the potency of HCC treatment in vitro or in vivo. RESULTS: 32A9 recognized human GPC3 with potent affinity and specificity. The epitope of 32A9 was located in the region of the GPC3 protein core close to the modification sites of the HS chain and outside of the Wnt-binding site of GPC3. The 32A9 antibody significantly inhibited HCC xenograft tumor growth in vivo. We then pursued two 32A9-based immunotherapeutic strategies by constructing an immunotoxin and CAR-T cells. The 32A9 immunotoxin exhibited specific cytotoxicity to GPC3-positive cancer cells, while 32A9 CAR-T cells efficiently eliminated GPC3-positive HCC cells in vitro and caused HCC xenograft tumor regressions in vivo. CONCLUSIONS: Our study provides a rationale for 32A9 as a promising GPC3-specific antibody candidate for HCC immunotherapy.


Assuntos
Carcinoma Hepatocelular , Imunotoxinas , Neoplasias Hepáticas , Carcinoma Hepatocelular/terapia , Glipicanas , Humanos , Neoplasias Hepáticas/terapia , Linfócitos T , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Clin Immunol ; 205: 116-124, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31228581

RESUMO

Autoimmune diseases are caused by self-immune responses to autoantigens, which damage body tissues and severely affect the patient's quality of life. Therapeutic drugs are associated with adverse side effects and their beneficial effects are limited to specific populations. Evidence indicates that exosomes which are small vesicles secreted by most cell types and body fluids, and may play roles in both immune stimulation and tolerance since they are involved in many processes such as immune signaling, inflammation and angiogenesis. Exosomes have also emerged as promising tools for therapeutic delivery, given their intrinsic features such as stability, biocompatibility and a capacity for stealth. In this review, we summarize existing literature regarding the production, efficacy, action mechanism, and potential therapeutic uses of exosomes in the contexts of autoimmune diseases such as type 1 diabetes mellitus, multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, and Sjogren's syndrome.


Assuntos
Doenças Autoimunes/terapia , Exossomos/transplante , Artrite Reumatoide/terapia , Doenças Autoimunes/imunologia , Diabetes Mellitus Tipo 1/terapia , Sistemas de Liberação de Medicamentos , Exossomos/química , Exossomos/imunologia , Humanos , Lúpus Eritematoso Sistêmico/terapia , Esclerose Múltipla/terapia , Síndrome de Sjogren/terapia
6.
Exp Lung Res ; 45(5-6): 123-134, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31210057

RESUMO

Pulmonary fibrosis (PF) is an end-stage change in lung disease characterized by fibroblast proliferation, massive extracellular matrix (ECM) aggregation with inflammatory damage, and severe structural deterioration. PD29 is a 29-amino acid peptide which has the potential to alleviate PF pathogenesis via three mechanisms: anti-angiogenesis, inhibition of matrix metalloproteinase activities, and inhibition of integrins. In this study, fibrotic lung injuries were induced in SD rats by a single intratracheal instillation of 5 mg/kg bleomycin (BLM). Then, these rats were administered 7.5, 5, or 2.5 mg/kg PD29 daily for 30 days. BLM induced-syndromes including structure distortion, excessive deposition of ECM, excessive inflammatory infiltration, and pro-inflammatory cytokine release were used to evaluate the protective effect of PD-29. Oxidative stress damage in lung tissues was attenuated by PD29 in a dose-dependent manner. The expression of TGF-ß1 and the phosphorylation of Smad-2/-3-its downstream targets-were enhanced by BLM and weakened by PD29. In vitro, PD29 inhibited TGF-ß1-induced epithelial-mesenchymal transition (EMT) and transformation in A549 cells and mouse primary fibroblasts into myofibroblasts. In summary, PD29 reversed EMT and transformation of fibroblasts into myofibroblasts in vitro and prevented PF in vivo possibly by suppressing the TGF-ß1/Smad pathway.


Assuntos
Pulmão/efeitos dos fármacos , Fibrose Pulmonar/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Células A549 , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Bleomicina , Avaliação Pré-Clínica de Medicamentos , Humanos , Pulmão/metabolismo , Metaloproteinases da Matriz/metabolismo , Camundongos , Cultura Primária de Células , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , Ratos Sprague-Dawley , Proteínas Smad/metabolismo , Inibidores Teciduais de Metaloproteinases/metabolismo , Fator de Crescimento Transformador beta/metabolismo
8.
Transl Res ; 259: 1-12, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-36977441

RESUMO

Chimeric antigen receptor T cell (CAR-T) therapy has limited efficacy for treating glioma because of the infiltrative nature of the blood-brain barrier (BBB) and T cell exhaustion. Conjugation with rabies virus glycoprotein (RVG) 29 enhances the brain-related efficacy of various agents. Here we assess whether RVG enhances the ability of CAR-T cells to cross the BBB and improves their immunotherapy. We generated 70R CAR-T cells (anti-CD70 CAR-T modified with RVG29) and validated their tumor-killing efficacy in vitro and in vivo. We validated their effects on tumor regression in a human glioma mouse orthotopic xenograft model as well as in patient-derived orthotopic xenograft (PDOX) models. The signaling pathways activated in 70R CAR-T cells were revealed by RNA sequencing. The 70R CAR-T cells we generated showed effective antitumor function against CD70+ glioma cells both in vitro and in vivo. 70R CAR-T cells were better able to cross the BBB into the brain than CD70 CAR-T cells under the same treatment conditions. Moreover, 70R CAR-T cells significantly promote the regression of glioma xenografts and improve the physical characteristics of mice without causing overt adverse effects. RVG modification enables CAR-T cells to cross the BBB, and stimulation with glioma cells induces 70R CAR-T cells to expand in a resting state. The modification of RVG29 has a positive impact on CAR-T therapy for brain tumors and may have potential in CAR-T therapy for glioma.


Assuntos
Glioma , Vírus da Raiva , Receptores de Antígenos Quiméricos , Humanos , Animais , Camundongos , Glioma/terapia , Glioma/metabolismo , Glicoproteínas , Imunoterapia Adotiva , Ensaios Antitumorais Modelo de Xenoenxerto , Linhagem Celular Tumoral
9.
J Chemother ; 34(6): 391-400, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34870566

RESUMO

The therapeutic efficacy of TMZ, a common used drug for chemotherapy, is limited by the resistance from colorectal cancer cells. Base excision repair (BER) pathway has been identified as one of the reasons for drug resistance. By blocking Polß-dependent BER (Base Excision Repair) pathway, the efficacy of TMZ treatment can be improved greatly. Several Polß inhibitors that have been identified could not become approved drugs due to lack of potency or specificity. To find therapeutic candidates with exquisite specificity and high affinity to Polß, phage display technology was used in the current research. We screened out a candidate Polß inhibitor, 10 D, that can inhibit the activity of Polßand SP-BER (Short-Patch Base excision Repair) pathway. Co-treatment with 10 D enhanced the sensitivity of colorectal cancer (CRC) cells to TMZ both in vitro and in vivo. Our data suggested that the novel Polß inhibitor we identified can improve TMZ efficacy and optimize CRC chemotherapy.


Assuntos
Bacteriófagos , Neoplasias Colorretais , DNA Polimerase beta , Bacteriófagos/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , DNA Polimerase beta/genética , DNA Polimerase beta/metabolismo , Reparo do DNA , Humanos , Biblioteca de Peptídeos , Temozolomida/farmacologia , Temozolomida/uso terapêutico
10.
Transl Cancer Res ; 11(11): 4185-4188, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36523319

RESUMO

Background: Trastuzumab, a monoclonal antibody which binds to the extracellular domain of human epidermal growth factor receptor 2 (HER2), is the first biological drug approved for the treatment of HER2-positive breast cancer. However, trastuzumab exhibits a series of clinical adverse effects, including cardiac toxicity, nerve damage, abnormal liver function, thrombocytopenia, etc. Case Description: We report a case of a 46-year-old female patient with invasive breast ductal carcinoma (classified as Stage 2B) who developed a rare severe edema in neck, face, chest, abdomen and both upper limbs after a single dose trastuzumab treatment. The patient (~60 kg weight) was administered with trastuzumab (420 mg) with an infusion time of 2 hours. The most severe edema symptom was observed in patient's hands, in which the epidermis was markedly transparent and tight. One month after trastuzumab administration, the patient was administered with methylprednisone (80 mg per day) for 5 days. The edema in patient's neck, face and both upper limbs was mildly reduced though the CT image showed no significant reduction of edema. Conclusions: Trastuzumab treatment for breast cancer patients, particularly those who have an allergic constitution, may have an adverse effect to develop severe edema, and treatment with methylprednisone at early stage can reduce such adverse reaction.

11.
J Biomed Res ; 36(3): 155-166, 2022 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-35545451

RESUMO

High-affinity antibodies are widely used in diagnostics and for the treatment of human diseases. However, most antibodies are isolated from semi-synthetic libraries by phage display and do not possess in vivo affinity maturation, which is triggered by antigen immunization. It is therefore necessary to engineer the affinity of these antibodies by way of in vitro assaying. In this study, we optimized the affinity of two human monoclonal antibodies which were isolated by phage display in a previous related study. For the 42A1 antibody, which targets the liver cancer antigen glypican-3, the variant T57H in the second complementarity-determining region of the heavy chain (CDR-H2) exhibited a 2.6-fold improvement in affinity, as well as enhanced cell-binding activity. For the I4A3 antibody to severe acute respiratory syndrome coronavirus 2, beneficial single mutations in CDR-H2 and CDR-H3 were randomly combined to select the best synergistic mutations. Among these, the mutation S53P-S98T improved binding affinity (about 3.7 fold) and the neutralizing activity (about 12 fold) compared to the parent antibody. Taken together, single mutations of key residues in antibody CDRs were enough to increase binding affinity with improved antibody functions. The mutagenic combination of key residues in different CDRs creates additive enhancements. Therefore, this study provides a safe and effective in vitro strategy for optimizing antibody affinity.

12.
J Immunother Cancer ; 9(4)2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33833049

RESUMO

BACKGROUND: Glypican-3 (GPC3), a cell surface glycoprotein that is pathologically highly expressed in hepatocellular carcinoma (HCC), is an attractive target for immunotherapies, including chimeric antigen receptor (CAR) T cells. The serum GPC3 is frequently elevated in HCC patients due to the shedding effect of cell surface GPC3. The shed GPC3 (sGPC3) is reported to block the function of cell-surface GPC3 as a negative regulator. Therefore, it would be worth investigating the potential influence of antigen shedding in anti-GPC3 CAR-T therapy for HCC. METHODS: In this study, we constructed two types of CAR-T cells targeting distinct epitopes of GPC3 to examine how sGPC3 influences the activation and cytotoxicity of CAR-T cells in vitro and in vivo by introducing sGPC3 positive patient serum or recombinant sGPC3 proteins into HCC cells or by using sGPC3-overexpressing HCC cell lines. RESULTS: Both humanized YP7 CAR-T cells and 32A9 CAR-T cells showed GPC3-specific antitumor functions in vitro and in vivo. The existence of sGPC3 significantly inhibited the release of cytokines and the cytotoxicity of anti-GPC3 CAR-T cells in vitro. In animal models, mice carrying Hep3B xenograft tumors expressing sGPC3 exhibited a worse response to the treatment with CAR-T cells under both a low and high tumor burden. sGPC3 bound to CAR-T cells but failed to induce the effective activation of CAR-T cells. Therefore, sGPC3 acted as dominant negative regulators when competed with cell surface GPC3 to bind anti-GPC3 CAR-T cells, leading to an inhibitory effect on CAR-T cells in HCC. CONCLUSIONS: We provide a proof-of-concept study demonstrating that GPC3 shedding might cause worse response to CAR-T cell treatment by competing with cell surface GPC3 for CAR-T cell binding, which revealed a new mechanism of tumor immune escape in HCC, providing a novel biomarker for patient enrolment in future clinical trials and/or treatments with GPC3-targeted CAR-T cells.


Assuntos
Biomarcadores Tumorais/antagonistas & inibidores , Carcinoma Hepatocelular/terapia , Glipicanas/antagonistas & inibidores , Imunoterapia Adotiva , Neoplasias Hepáticas/terapia , Receptores de Antígenos Quiméricos/genética , Linfócitos T/transplante , Animais , Ligação Competitiva , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/imunologia , Carcinoma Hepatocelular/sangue , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Citocinas/metabolismo , Citotoxicidade Imunológica , Feminino , Glipicanas/sangue , Glipicanas/imunologia , Neoplasias Hepáticas/sangue , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Ativação Linfocitária , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos Nus , Estudo de Prova de Conceito , Ligação Proteica , Receptores de Antígenos Quiméricos/imunologia , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Carga Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
13.
J Hematol Oncol ; 14(1): 152, 2021 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-34556152

RESUMO

Chimeric antigen receptor T-cell (CAR-T) therapy has shown tremendous success in eradicating hematologic malignancies. However, this success has not yet been extrapolated to solid tumors due to the limited infiltration and persistence of CAR-T cells in the tumor microenvironment (TME). In this study, we screened a novel anti-CD70 scFv and generated CD70 CAR-T cells that showed effective antitumor functions against CD70+ renal carcinoma cells (RCCs) both in vitro and in vivo. We further evaluated the effect and explored the molecular mechanism of a PARP inhibitor (PARPi) in CAR-T cell immunotherapy by administering the PARPi to mouse xenografts model derived from human RCC cells. Treatment with the PARPi promoted CAR-T cell infiltration by stimulating a chemokine milieu that promoted CAR-T cell recruitment and the modulation of immunosuppression in the TME. Moreover, our data demonstrate that PARPi modulates the TME by activating the cGAS-STING pathway, thereby altering the balance of immunostimulatory signaling and enabling low-dose CAR-T cell treatment to induce effective tumor regression. These data demonstrate the application of CD70 CAR-T cell therapeutic strategies for RCC and the cross-talk between targeting DNA damage responses and antitumor CAR-T cell therapy. These findings provide insight into the mechanisms of PARPis in CAR-T cell therapy for RCC and suggest a promising adjuvant therapeutic strategy for CAR-T cell therapy in solid tumors.


Assuntos
Ligante CD27/antagonistas & inibidores , Carcinoma de Células Renais/terapia , Imunoterapia Adotiva/métodos , Neoplasias Renais/terapia , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Anticorpos de Cadeia Única/uso terapêutico , Animais , Ligante CD27/imunologia , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/imunologia , Linhagem Celular Tumoral , Dano ao DNA/efeitos dos fármacos , Humanos , Neoplasias Renais/genética , Neoplasias Renais/imunologia , Proteínas de Membrana/imunologia , Camundongos , Nucleotidiltransferases/imunologia , Transdução de Sinais
14.
DNA Repair (Amst) ; 95: 102953, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32861926

RESUMO

The up-regulation of PRMT1 is critical to the cell growth and cancer progression of lung cancer cells. In our research, we found that PRMT1 is important to the DNA repair ability and drug resistance of lung cancer cells. To demonstrate the functions of PRMT1, we identified Flap endonuclease 1 (FEN1) as a post-translationally modified downstream target protein of PRMT1. As a major component of Base Excision Repair pathway, FEN1 plays an important role in DNA replication and DNA damage repair. However, the detailed mechanism of FEN1 up-regulation in lung cancer cells remains unclear. In our study, we identified PRMT1 as a key factor that maintains the high expression levels of FEN1, which is critical to the DNA repair ability and the chemotherapeutic drug resistance of lung cancer cells.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Endonucleases Flap/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/patologia , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Repressoras/metabolismo , Células A549 , Apoptose/genética , Proliferação de Células/genética , Reparo do DNA , Epigênese Genética , Técnicas de Silenciamento de Genes , Humanos , Proteína-Arginina N-Metiltransferases/deficiência , Proteína-Arginina N-Metiltransferases/genética , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Regulação para Cima
15.
Sci Rep ; 8(1): 14517, 2018 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-30266938

RESUMO

Shift metabolism profile from mitochondrial oxidative phosphorylation to aerobic glycolysis (Warburg effect) is a key for tumor cell growth and metastasis. Therefore, suppressing the tumor aerobic glycolysis shows a great promise in anti-tumor therapy. In the present study, we study the role of shikonin, a naphthoquinone isolated from the traditional Chinese medicine Lithospermum, in inhibiting tumor aerobic glycolysis and thus tumor growth. We found that shikonin dose-dependently inhibited glucose uptake and lactate production in Lewis lung carcinoma (LLC) and B16 melanoma cells, confirming the inhibitory effect of shikonin on tumor aerobic glycolysis. Treatment of shikonin also decreased tumor cell ATP production. Furthermore, pyruvate kinase M2 (PKM2) inhibitor or activator respectively altered the effect of shikonin on tumor cell aerobic glycolysis, suggesting that suppression of cell aerobic glycolysis by shikonin is through decreasing PKM2 activity. Western blot analysis confirmed that shikonin treatment reduced tumor cell PKM2 phosphorylation though did not reduce total cellular PKM2 level. In vitro assay also showed that shikonin treatment significantly promoted tumor cell apoptosis compared to untreated control cells. Finally, when mice implanted with B16 cells were administered with shikonin or control vehicle, only shikonin treatment significantly decreased B16 tumor cell growth. In conclusion, this study demonstrates that shikonin inhibits tumor growth in mice by suppressing PKM2-mediated aerobic glycolysis.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Medicamentos de Ervas Chinesas/uso terapêutico , Inibidores Enzimáticos/farmacologia , Glicólise/efeitos dos fármacos , Melanoma Experimental/tratamento farmacológico , Naftoquinonas/uso terapêutico , Proteínas de Neoplasias/antagonistas & inibidores , Piruvato Quinase/antagonistas & inibidores , Trifosfato de Adenosina/metabolismo , Aerobiose/efeitos dos fármacos , Animais , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Medicamentos de Ervas Chinesas/farmacologia , Masculino , Camundongos , Camundongos Nus , Camundongos SCID , Terapia de Alvo Molecular , Naftoquinonas/farmacologia , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Neoplasias Gástricas/patologia
16.
Int J Clin Exp Med ; 7(12): 5857-61, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25664120

RESUMO

OBJECTIVE: To investigate the correlation of peripheral DPYD gene polymorphism with 5-fluorouracil (5-FU) susceptibility and side effect in patients with colon cancer. METHODS: The total DNA of peripheral mononuclear cells was extracted in 100 cases of colon cancer patients. Quantitative PCR was conducted to measure DPYD gene 14G1A, A1627G, T85C 3 loci polymorphism, and analyze the correlation of 5-FU susceptibility, side effects with DPYD gene polymorphism. RESULTS: Mutations were detected at position 14G1A (mutation rate 14%), A1627G (mutation rate 11%), and T85C (mutation rate 17%). The effective rate of 3 loci of wild type was significantly higher than that of mutant type (P < 0.05). With respect to side effects such as myelosuppression, hand-foot syndrome, diarrhea, and gastrointestinal reactions, the incidence in mutation type was significantly higher than that in the wild type (P < 0.05). CONCLUSION: DPYD gene polymorphism plays a guiding role in predicting efficacy and toxicity of 5-FU, which can be used as an important reference index of 5-FU individualized administration scheme.

17.
Int J Clin Exp Med ; 7(11): 4272-81, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25550942

RESUMO

Malignant ascites (MA) is a pathological condition due to a variety of primary abdominal and extra-abdominal neoplasms. It is a primary cause of morbidity and presents many difficulties in evaluation and treatment. In this study we used dendritic cell vaccines combined with cytokine-induced killer (CIK) cells intraperitoneal injected in patients with MA, and evaluated the safety and efficacy of this treatment. The results showed that the percentage of CD3(+) CD56(+) CIK cells after treatment increased significantly while the percentage of CD4(+) CD25(+) Treg cells decreased (P < 0.05). The clinical response rate (RR) was 40.9% and disease control rate (DCR) was 77.3%. We then studied and identified the mechanisms of the anti-tumor effects of the vaccines by analyzing a series of cytokines that are commonly involved in tumor progression and ascitic development including granulocyte macrophage colony stimulating factor (GM-CSF), interleukin-10 (IL-10), interferon-γ (IFN-γ), tumor necrosis factor-α (TGF-α), tumor necrosis factor-ß (TGF-ß), Vascular endothelial growth factor (VEGF) and monocyte chemotactic protein-1 (MCP-1). These data demonstrated that intraperitoneal injection with DC vaccines combined with CIK cells in patients with malignant peritoneal effusion is safe and feasible. This therapy modality can achieve a certain clinical benefit even in patients resistant to conventional treatments.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA