Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 523(1): 135-139, 2020 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-31839218

RESUMO

Cancer cells are methionine (MET) and methylation addicted and are highly sensitive to MET restriction. The present study determined the efficacy of oral-recombinant methioninase (o-rMETase) and the DNA methylation inhibitor, decitabine (DAC) on restricting MET in an undifferentiated-soft tissue sarcoma (USTS) patient-derived orthotopic xenograft (PDOX) nude-mouse model. The USTS PDOX models were randomized into five treatment groups of six mice: Control; doxorubicin (DOX) alone; DAC alone; o-rMETase alone; and o-rMETase-DAC combination. Tumor size and body weight were measured during the 14 days of treatment. Tumor growth was arrested only in the o-rMETase-DAC condition. Tumors treated with the o-rMETase-DAC combination exhibited tumor necrosis with degenerative changes. This study demonstrates that the o-rMETase-DAC combination could arrest the USTS PDOX tumor suggesting clinical promise.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Liases de Carbono-Enxofre/metabolismo , Decitabina/farmacologia , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias Musculares/tratamento farmacológico , Sarcoma/tratamento farmacológico , Administração Oral , Animais , Antimetabólitos Antineoplásicos/administração & dosagem , Liases de Carbono-Enxofre/administração & dosagem , Terapia Combinada , Decitabina/administração & dosagem , Feminino , Humanos , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Neoplasias Musculares/patologia , Neoplasias Musculares/cirurgia , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/patologia , Neoplasias Experimentais/cirurgia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo , Sarcoma/patologia , Sarcoma/cirurgia
2.
Biochem Biophys Res Commun ; 518(2): 306-310, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31421825

RESUMO

The aim of this study was to determine the efficacy of oral recombinant methioninase (o-rMETase) on a colon cancer primary tumor using a patient-derived orthotopic xenograft (PDOX) nude mouse model. Forty colon cancer primary tumor PDOX mouse models were divided into 4 groups of 10 mice each (total 40 mice) by measuring the tumor size. The groups were as follows: untreated control; 5-fluorouracil (5-FU) (50 mg/kg, once a week for two weeks, N = 10 mice) and oxaliplatinum (OXA) (6 mg/kg, once a week for two weeks, N = 10 mice); o-rMETase (100 units/day, oral 14 consecutive days, N = 10 mice); combination of 5-FU + OXA and o-rMETase (N = 10 mice). All treatments inhibited tumor growth compared to the untreated control. The combination of 5-FU + OXA and o-rMETase was significantly more efficacious than other treatments. The present study demonstrates the efficacy of o-rMETase combination therapy on a PDOX colon cancer primary tumor, suggesting potential clinical development of o-rMETase in recalcitrant cancer.


Assuntos
Antineoplásicos/uso terapêutico , Liases de Carbono-Enxofre/uso terapêutico , Neoplasias do Colo/tratamento farmacológico , Fluoruracila/uso terapêutico , Administração Oral , Animais , Antineoplásicos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Liases de Carbono-Enxofre/administração & dosagem , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Fluoruracila/administração & dosagem , Humanos , Camundongos , Camundongos Nus , Camundongos Transgênicos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico , Resultado do Tratamento , Células Tumorais Cultivadas
3.
Invest New Drugs ; 37(2): 201-209, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-29948359

RESUMO

The anti-cancer efficacy of methionine γ-lyase (MGL) from Clostridium sporogenes (C. sporogenes) is described. MGL was active against cancer models in vitro and in vivo. The calculated EC50 values for MGL were 4.4 U/ml for A549, 7.5 U/ml for SK-BR3, 2.4 U/ml for SKOV3, and 0.4 U/ml for MCF7 cells. The combination of doxorubicin (DOX) and MGL was more effective for A549 human lung cancer growth inhibition than either agent alone in vitro and in vivo. MGL reduced the EC50 of doxorubicin from 35.9 µg/mL to 0.01-0.265 µg/mL. The growth inhibitory effect of DOX + MGL on A549 xenografts in vivo was reflective of the results obtained in vitro. The inhibition rate of tumor growth in the combined arm was 57%, significantly higher than that in the doxorubicin (p = 0.033)-alone arm.


Assuntos
Liases de Carbono-Enxofre/administração & dosagem , Cisplatino/farmacologia , Clostridium/enzimologia , Doxorrubicina/farmacologia , Sinergismo Farmacológico , Neoplasias/tratamento farmacológico , Células A549 , Animais , Antibióticos Antineoplásicos/farmacologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/enzimologia , Neoplasias/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Br J Cancer ; 118(9): 1189-1199, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29576621

RESUMO

BACKGROUND: To investigate the mechanism of lentiviral vector carrying methioninase enhances the sensitivity of drug-resistant gastric cancer cells to Cisplatin. METHODS: Death receptors, anti-apoptotic protein, NF-κB, and TRAIL pathway-related factors were detected. The influence of LV-METase transfection on cell viability and pathway-related proteins were assessed by MTT method and western blot, respectively. Different treatments (NF-κB or caspase-3 inhibitor induction, TRAIL supplement, etc.) were performed in gastric cancer cells and the above parameters were analysed. Moreover, the connection between miR-21 and NF-κB or caspase-8 was determined by Chip and luciferase assay, respectively. LV-METase transfection drug-resistant gastric cancer cells were injected subcutaneously into mice. RESULTS: The expression of free MET, miR-21-5p, MDR1, P-gp, and DR5 was significantly increased in drug-resistant gastric cancer cell lines. When cells were transfected with LV-METase, intracellular TRAIL signalling was activated while NF-κB pathway was inhibited. Besides, enhanced TRAIL signalling or repressed NF-κB pathway can promote the sensitivity of drug-resistant strains to Cisplatin, and the combination shows more sensitive to sensitisation. LV-METase promoted TRAIL expression by reducing NF-κB, thereby contributing to the downregulation of P-gp and enhancing the susceptibility of drug-resistant gastric cancer cells to Cisplatin. Furthermore, miR-21 regulated by NF-κB mediated the expression of P-gp protein via inhibiting caspase-8, thus regulating Cisplatin-induced cell death. CONCLUSIONS: Our results suggest that LV-METase has potential as a therapeutic agent for gastric cancer treatment.


Assuntos
Liases de Carbono-Enxofre/administração & dosagem , Liases de Carbono-Enxofre/genética , Cisplatino/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Terapia Genética/métodos , Lentivirus/genética , Neoplasias Gástricas/tratamento farmacológico , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular Tumoral , Terapia Combinada , Sinergismo Farmacológico , Feminino , Vetores Genéticos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Biochem Biophys Res Commun ; 503(4): 3086-3092, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30166061

RESUMO

Melanoma is a recalcitrant cancer. To improve and individualize treatment for this disease, we previously developed a patient-derived orthotopic xenograft (PDOX) model for melanoma. We previously reported the individual efficacy of tumor-targeting Salmonella typhimurium A1-R (S. typhimurium A1-R) and recombinant methioninase (rMETase) for melanoma in the PDOX models of this disease. In the present study, we evaluated the efficacy of the combination of S. typhimurium A1-R with orally-administered rMETase (o-rMETase) for BRAF-V600E-negative melanoma in a PDOX model. Three weeks after implantation, 60 PDOX mouse models were randomized into six groups of 10 mice each: untreated control, temozolomide (TEM); o-rMETase; S. typhimurium A1-R; TEM + rMETase, S. typhimurium A1-R + rMETase. All treatments inhibited tumor growth compared to untreated control (TEM: p < 0.0001, rMETase: p < 0.0001, S. typhimurium A1-R: p < 0.0001, TEM + rMETase: p < 0.0001, S. typhimurium A1-R + rMETase: p < 0.0001). The most effective was the combination of S. typhimurium A1-R + o-rMETase which regressed this melanoma PDOX, thereby indicating a new paradigm for treatment of metastatic melanoma.


Assuntos
Antineoplásicos/uso terapêutico , Liases de Carbono-Enxofre/uso terapêutico , Melanoma/terapia , Pseudomonas putida/enzimologia , Salmonella typhimurium , Temozolomida/uso terapêutico , Administração Oral , Animais , Antimetabólitos Antineoplásicos/uso terapêutico , Antineoplásicos/administração & dosagem , Liases de Carbono-Enxofre/administração & dosagem , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Humanos , Masculino , Melanoma/genética , Melanoma/microbiologia , Melanoma/patologia , Camundongos Nus , Mutação Puntual , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico , Salmonella typhimurium/fisiologia , Temozolomida/administração & dosagem
6.
Anticancer Res ; 44(9): 3885-3889, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39197890

RESUMO

BACKGROUND/AIM: Pancreatic cancer has a very poor prognosis with a 5-year survival rate of less than 5% among patients with distant metastasis, a figure that has not improved over many decades. Only 10 to 20% patients are candidates for curative surgery at presentation due to the aggressive nature and asymptomatic progression of pancreatic cancer. Although first-line chemotherapy, such as FOLFIRINOX and gemcitabine + nab paclitaxel, improved the median survival from 8.5 to 11.1 months, more effective treatments are immediately needed. The aim of the present study was to evaluate the efficacy of methionine restriction with oral rMETase (o-rMETase) and a low-methionine diet combined with first-line chemotherapy on a patient with stage IV metastatic pancreatic cancer. CASE REPORT: A 63-year-old female was diagnosed with metastatic pancreatic cancer in October 2023. The patient started FOLFIRINOX as first-line chemotherapy in combination with methionine restriction, which comprised o-rMETase 250 units twice a day and a low-methionine diet. The patient was monitored using computed tomography and CA19-9 blood tests. After five months from the start of combination therapy, the size of the primary tumor decreased by 40% along with liver-metastasis regression. The CA19-9 blood marker decreased by 86%. The patient sustains a high performance status and continues the combination therapy without severe side effects. CONCLUSION: Methionine restriction consisting of o-rMETase and a low-methionine diet, in combination with first-line chemotherapy, was highly effective in a patient with inoperable stage IV pancreatic cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica , Liases de Carbono-Enxofre , Metionina , Neoplasias Pancreáticas , Humanos , Feminino , Liases de Carbono-Enxofre/administração & dosagem , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/sangue , Pessoa de Meia-Idade , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Metionina/administração & dosagem , Estadiamento de Neoplasias , Biomarcadores Tumorais/sangue , Fluoruracila/administração & dosagem , Antígeno CA-19-9/sangue , Leucovorina/administração & dosagem , Leucovorina/uso terapêutico , Irinotecano/administração & dosagem , Irinotecano/uso terapêutico , Oxaliplatina/administração & dosagem , Oxaliplatina/uso terapêutico , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico , Administração Oral
7.
Anticancer Res ; 44(9): 3785-3791, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39197928

RESUMO

BACKGROUND/AIM: Drug resistance has been a recalcitrant problem for sarcoma patients for many decades. Trabectedin is a second-line chemotherapy for soft-tissue sarcoma that often leads to resistance and death of the patients. The objective of the present study was to address the issue of trabectedin-chemoresistance in HT1080 fibrosarcoma cells by combining recombinant methioninase (rMETase) with trabectedin and examining their efficacy on trabectedin-resistant fibrosarcoma cells in vitro. MATERIALS AND METHODS: Trabectedin-resistant HT1080 (TR-HT1080) cells were generated by subjecting HT1080 human fibrosarcoma cells to increasing trabectedin concentrations (3.3-8 nM). IC50 values for trabectedin and rMETase were compared for HT1080 and TR-HT1080 cells. TR-HT 1080 cells were placed into four groups to determine synergy of rMETase and trabectedin on TR-HT1080 cells: a control group with no treatment; a group treated with trabectedin (3.3 nM); a group treated with rMETase (0.75 U/ml); and a group treated with both trabectedin (3.3 nM) and rMETase (0.75 U/ml). RESULTS: The IC50 value of trabectedin- on TR-HT1080 cells was 42.9 nM, whereas the IC50 value of trabectedin on the parental HT1080 cells was 3.3 nM, indicating a 13-fold increase. The combination of rMETase (0.75 U/ml) and trabectedin (3.3 nM) was synergistic on TR-HT1080 cells resulting in an inhibition of 64.2% compared to trabectedin alone (5.7%) or rMETase alone (50.5%) (p<0.05). rMETase increased the efficacy of trabectedin 11-fold on trabectedin-resistant fibrosarcoma cells. CONCLUSION: The combined administration of trabectedin and rMETase was synergistic on the viability of TR-HT1080 cells in vitro. The combination of rMETase and trabectedin has promising clinical potential for overcoming chemo-resistance of soft-tissue sarcoma.


Assuntos
Antineoplásicos Alquilantes , Liases de Carbono-Enxofre , Dioxóis , Resistencia a Medicamentos Antineoplásicos , Proteínas Recombinantes , Tetra-Hidroisoquinolinas , Trabectedina , Humanos , Trabectedina/farmacologia , Liases de Carbono-Enxofre/administração & dosagem , Liases de Carbono-Enxofre/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Tetra-Hidroisoquinolinas/farmacologia , Tetra-Hidroisoquinolinas/administração & dosagem , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Alquilantes/uso terapêutico , Dioxóis/farmacologia , Dioxóis/uso terapêutico , Dioxóis/administração & dosagem , Proteínas Recombinantes/farmacologia , Linhagem Celular Tumoral , Sarcoma/tratamento farmacológico , Sarcoma/patologia , Fibrossarcoma/tratamento farmacológico , Fibrossarcoma/patologia , Sinergismo Farmacológico
8.
Anticancer Res ; 44(6): 2359-2367, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38821601

RESUMO

BACKGROUND/AIM: The alkylating agent trabectedin, which binds the minor groove of DNA, is second-line therapy for soft-tissue sarcoma but has only moderate efficacy. The aim of the present study was to determine the synergistic efficacy of recombinant methioninase (rMETase) and trabectedin on fibrosarcoma cells in vitro, compared with normal fibroblasts. MATERIALS AND METHODS: HT1080 human fibrosarcoma cells expressing green fluorescent protein (GFP) in the nucleus and red fluorescent protein (RFP) in the cytoplasm and Hs27 normal human fibroblasts, were used. Each cell line was cultured in vitro and divided into four groups: no-treatment control; trabectedin treated; rMETase treated; and trabectedin plus rMETase treated. The dual-color HT1080 cells were used to quantitate nuclear fragmentation in each treatment group. RESULTS: The combination of rMETase and trabectedin was highly synergistic to decrease HT1080 cell viability. In contrast, there was no synergy on Hs27 cells. Moreover, nuclear fragmentation occurred synergistically with the combination of trabectedin and rMETase on dual-color HT1080 cells. CONCLUSION: The combination treatment of trabectedin plus rMETase was highly synergistic on fibrosarcoma cells in vitro suggesting that the combination can improve the outcome of trabectedin alone in future clinical studies. The lack of synergy of rMETase and trabectedin on normal fibroblasts suggests the combination is not toxic to normal cells. Synergy of the two drugs may be due to the high rate of nuclear fragmentation on treated HT1080 cells, and the late-S/G2 cell-cycle block of cancer cells by rMETase, which is a target for trabectedin. The results of the present study suggest the future clinical potential of the combination of rMETase and trabectedin for soft-tissue sarcoma.


Assuntos
Liases de Carbono-Enxofre , Sobrevivência Celular , Dioxóis , Sinergismo Farmacológico , Fibroblastos , Fibrossarcoma , Tetra-Hidroisoquinolinas , Trabectedina , Humanos , Fibrossarcoma/tratamento farmacológico , Fibrossarcoma/patologia , Fibrossarcoma/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Trabectedina/farmacologia , Liases de Carbono-Enxofre/farmacologia , Liases de Carbono-Enxofre/administração & dosagem , Tetra-Hidroisoquinolinas/farmacologia , Dioxóis/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Linhagem Celular Tumoral , Antineoplásicos Alquilantes/farmacologia , Núcleo Celular/metabolismo , Núcleo Celular/efeitos dos fármacos
9.
J Nanosci Nanotechnol ; 13(1): 178-83, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23646714

RESUMO

Stealth cationic liposomes (SCLs) modified with tumor-targeting single-chain fragment variable (scFV) antibody for systemic delivery of recombinant methioninase (rMETase) for gastric cancer were prepared successfully. These functional SCL nanoparticles are composed of cationic lipids, dioleoylphosphatidylethanolamine, and distearoylphosphatidylethanolamine-polyethylene glycol, which have lower gene transfection efficiencies compared with Lipofectamine 2000, and can also be used as effective gene delivery vectors. Increased therapeutic efficiency of rMETase-loaded scFV-SCLs were tested in SGC-7901 cells and compared with free rMETase in solution and rMETase-loaded SCLs. In addition, scFV-SCLs (effective diameter: 185.7 nm; polydispersity index: 0.236) can significantly boost rhodamine 123 cellular accumulation in the cytoplasm. The scFV-targeted SCLs can be used as a potentially effective drug delivery system.


Assuntos
Liases de Carbono-Enxofre/administração & dosagem , Lipossomos/química , Nanocápsulas/administração & dosagem , Serina Endopeptidases/farmacocinética , Anticorpos de Cadeia Única/farmacocinética , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/metabolismo , Liases de Carbono-Enxofre/química , Cátions , Linhagem Celular Tumoral , Difusão , Humanos , Nanocápsulas/química , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/química , Serina Endopeptidases/uso terapêutico , Anticorpos de Cadeia Única/uso terapêutico , Neoplasias Gástricas/patologia , Resultado do Tratamento
10.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 28(4): 839-42, 2011 Aug.
Artigo em Zh | MEDLINE | ID: mdl-21936392

RESUMO

Methionine-dependent increase in tumor cells is a specific metabolic defect. This metabolic defect is also a target for selective treatment of cancer. Studies found that the methionine gamma-lyase (methioninase, L-methionine gamma-lyase) can specificly split the methionine of extracellular and intracellular, so it can strongly inhibit the growth of tumor cells and induce apoptosis of tumor cells. However, no effect on normal cells has been found, so the methionine gamma-lyase may play the anti-tumor role. We also explored in the present study another effect of methionine gamma-lyase as a single agent on DNA methylation levels and DNA synthesis, which may change as a result of deprivation of methionine, and thus may enhance anti-tumor effects. Animal studies and clinical trials showed that a variety of methionine dependent methionine gamma-lyase can eliminate the tumor cells. Therefore, methionine restriction is an effective anticancer strategy. Methionine gamma-lyase has shown great prospects as a new type of gene therapy. This article made a review of it.


Assuntos
Liases de Carbono-Enxofre/administração & dosagem , Terapia Genética/métodos , Neoplasias/tratamento farmacológico , Animais , Humanos , Neoplasias/terapia , Proteínas Recombinantes/administração & dosagem , Transfecção
11.
Anticancer Res ; 41(4): 1745-1751, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33813378

RESUMO

BACKGROUND/AIM: Osteosarcoma is the most frequent malignant bone tumor. Failure of first-line therapy results in poor prognosis of osteosarcoma. In the present report, we examined the efficacy of the combination of oral recombinant methioninase (o-rMETase) and docetaxel (DOC) on an osteosarcoma patient-derived orthotopic xenograft (PDOX) mouse model. MATERIALS AND METHODS: Osteosarcoma-PDOX models were established by tumor insertion within the tibia of nude mice. The osteosarcoma PDOX models were randomized into four groups (4-5 mice per group): control; o-rMETae alone; DOC alone; o- rMETase combined with DOC. The treatment period was 3 weeks. RESULTS: The combination of o-rMETase and DOC showed significant efficacy compared to the control group (p=0.03). In contrast, there was no significant efficacy of o-rMETase alone or DOC alone (p=0.65, 0.60, respectively). CONCLUSION: o-rMETase converted an osteosarcoma PDOX from DOC-resistant to -sensitive. This combination therapy may be effective against recalcitrant osteosarcoma and other recalcitrant cancers.


Assuntos
Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias Ósseas/tratamento farmacológico , Liases de Carbono-Enxofre/administração & dosagem , Docetaxel/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Osteossarcoma/tratamento farmacológico , Tíbia/efeitos dos fármacos , Administração Oral , Adolescente , Animais , Neoplasias Ósseas/patologia , Humanos , Masculino , Camundongos Nus , Osteossarcoma/patologia , Proteínas Recombinantes/administração & dosagem , Tíbia/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Anticancer Res ; 41(4): 1921-1926, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33813397

RESUMO

BACKGROUND/AIM: Methionine addiction is a general and fundamental hallmark of cancer due to the excess use of methionine for transmethylation reactions, termed the "Hoffman Effect". Methionine addiction has been shown to be a highly-effective target for cancer therapy by methionine restriction with oral recombinant methioninase (o-rMETase) in preclinical studies, including patient- derived orthotopic xenograft (PDOX) mouse models of cancer. A clinical study of o-rMETase as a supplement showed a 70% reduction of PSA levels in a patient with bone-metastatic prostate cancer. MATERIALS AND METHODS: In the present study, two advanced prostate-cancer patients took o-rMETase as a supplement for approximately one month. RESULTS: One of the patients taking o-rMETase showed a 38% reduction of PSA levels and the second patient showed a 20% PSA reduction. CONCLUSION: o-rMETase shows promise for treating patients with advanced prostate cancer.


Assuntos
Liases de Carbono-Enxofre/administração & dosagem , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/tratamento farmacológico , Administração Oral , Idoso de 80 Anos ou mais , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/secundário , Progressão da Doença , Regulação para Baixo/efeitos dos fármacos , Terapia de Reposição de Enzimas , Humanos , Masculino , Metionina/sangue , Metionina/efeitos dos fármacos , Pessoa de Meia-Idade , Projetos Piloto , Antígeno Prostático Específico/efeitos dos fármacos , Neoplasias da Próstata/sangue , Neoplasias da Próstata/patologia , Proteínas Recombinantes/administração & dosagem
13.
In Vivo ; 34(2): 489-494, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32111745

RESUMO

BACKGROUND/AIM: obesity is a world-wide recalcitrant problem leading to many diseases. Dietary methionine restriction (MR) has been shown to prevent obesity, but it is an onerous regimen. The present study aimed to determine the effects of oral recombinant methionase (o-rMETase), on preventing obesity in mice on a high-fat diet. MATERIALS AND METHODS: Male C57BL/6J mice in the control group were fed a control diet (CD) (+6.5% fat) for 25 days, and others were fed a high-fat (HF) diet (+34.3% fat) for 25 days. Then, the mice were divided into three dietary groups: 1) HF + phosphate buffered saline (PBS) group; 2) HF + o-rMETase group; and 3) untreated non-HF group. RESULTS: The mice on the CD increased in body weight by 14% during experimental period of 25 days; in contrast the mice in the HF+PBS group increased by 33%; however, the mice on the HF+o-rMETase group increased only by 14% (p=0.02, HF+PBS vs HF+o-rMETase). CONCLUSION: The HF+ o-rMETase group had the same weight increase as untreated mice on a normal fat diet, demonstrating the potential for o-rMETase to eliminate the need for dieting to maintain normal body weight.


Assuntos
Liases de Carbono-Enxofre/administração & dosagem , Dieta Hiperlipídica , Obesidade/etiologia , Obesidade/prevenção & controle , Proteínas Recombinantes/administração & dosagem , Animais , Peso Corporal , Modelos Animais de Doenças , Humanos , Masculino , Camundongos
14.
In Vivo ; 34(3): 979-984, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32354883

RESUMO

BACKGROUND/AIM: We have recently shown that oral recombinant methionase (o-rMETase) prevents obesity and diabetes onset in mice on a high-fat (HF) diet. The present study aimed to determine if o-rMETase can inhibit the onset of nonalcoholic fatty liver disease (NAFLD) onset in mice on a high-fat diet. MATERIALS AND METHODS: Male C57BL/6J mice in the control group were fed a normal-fat diet (NFD) (+6.5% fat), and other mice were fed a high-fat (HF) diet (+34.3% fat). Then, the mice on the HF diet were divided into two dietary groups: i) HF+phosphate buffered saline (PBS) group, and ii) HF+o-rMETase group. RESULT: The fatty change score in the livers of mice treated with HF+PBS increased to an average of 2.6 during the experimental period of 8 weeks. In contrast, the fatty change in the livers of mice on the HF+o-rMETase group had an average score of 0.92 (p=0.04, HF+PBS vs HF+o-rMETase). CONCLUSION: o-rMETase inhibited the onset of NAFLD as well as prevented obesity and the onset of diabetes on a high-fat diet, offering a possibility of a new paradigm to prevent liver cirrhosis or liver cancer via NAFLD.


Assuntos
Liases de Carbono-Enxofre/administração & dosagem , Dieta Hiperlipídica/efeitos adversos , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Proteínas Recombinantes/administração & dosagem , Administração Oral , Animais , Modelos Animais de Doenças , Fígado/metabolismo , Fígado/patologia , Masculino , Metionina/metabolismo , Camundongos , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia
15.
In Vivo ; 34(3): 973-978, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32354882

RESUMO

BACKGROUND/AIM: We have recently shown that oral recombinant methionase (o-rMETase) prevents obesity in mice on a high-fat (HF) diet. The present study aimed to determine if o-rMETase can inhibit the onset of diabetes in mice on a HF diet. MATERIALS AND METHODS: The mice on a HF diet were divided into two groups: 1) HF+phosphate buffered saline (PBS) group; 2) HF+o-rMETase group. RESULTS: The blood glucose level in the HF+PBS group increased to average of 201 mg/dl during the experimental period of 8 weeks. In contrast, the blood glucose level in the HF+o-rMETase group maintained an average of 126 mg/dl (p<0.01, HF+PBS vs. HF+o-rMETase). The glucose tolerance test showed a significant increase in tolerance in the HF+o-rMETase group at 120 min after glucose injection compared to the HF+PBS group (p=0.04). Visceral adipose tissue was significantly less in the HF+o-rMETase group than the HF+PBS group (p=0.05). There was no difference in insulin levels, cholesterol or triglycerides between the HF+PBS and HF+o-rMETase groups. CONCLUSION: o-rMETase inhibited the onset of diabetes as well as prevented obesity on a high-fat diet, offering a possibility of a new and easy-to-use alternative to severe dieting or insulin injections.


Assuntos
Liases de Carbono-Enxofre/administração & dosagem , Diabetes Mellitus Experimental/metabolismo , Hipoglicemiantes/administração & dosagem , Proteínas Recombinantes/administração & dosagem , Animais , Biomarcadores , Glicemia , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/etiologia , Diabetes Mellitus Experimental/prevenção & controle , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Glucose/metabolismo , Lipídeos/sangue , Camundongos
16.
Anticancer Res ; 40(9): 4869-4874, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32878774

RESUMO

BACKGROUND/AIM: In the present study, we evaluated the efficacy of adjuvant administration of oral recombinant methioninase (o-rMETase) against recurrence and metastasis in a 4T1 murine breast-cancer syngeneic model. MATERIALS AND METHODS: 4T1 cells were orthotopically implanted into the 2nd mammary fat pad of BALB/c mice. The 4T1 orthotopic syngeneic models were randomized into 2 groups after primary tumor resection: untreated control and o-rMETase (100 units, oral, daily, 2 weeks). RESULTS: The frequency and extent of local recurrence were reduced by o-rMETase. The number of individual cancer cells and metastatic nodules on the lung surface was significantly lower in the o-rMETase-treated mice than the untreated control mice. CONCLUSION: Adjuvant o-rMETase inhibited local recurrence and lung metastasis after primary tumor resection.


Assuntos
Antimetabólitos Antineoplásicos/administração & dosagem , Liases de Carbono-Enxofre/administração & dosagem , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/tratamento farmacológico , Administração Oral , Animais , Linhagem Celular Tumoral , Quimioterapia Adjuvante , Esquema de Medicação , Feminino , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/cirurgia , Camundongos , Camundongos Endogâmicos BALB C , Recidiva Local de Neoplasia/prevenção & controle , Proteínas Recombinantes/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Anticancer Res ; 40(11): 6083-6091, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33109546

RESUMO

BACKGROUND/AIM: The aim of the study was to determine if oral recombinant methioninase (o-rMETase) can sensitize an orthotopic bladder tumor in nude mice to low-dose cisplatinum (CDDP). MATERIALS AND METHODS: The green fluorescent protein (GFP)-expressing UM-UC-3-GFP bladder cancer was surgically orthotopically implanted (SOI) to the bladder in nude mice. The treatment was initiated when the primary tumor volume reached 100 mm3 Mice were assigned to 3 groups: G1: Saline vehicle (0.1 ml per mouse, oral, twice per day); G2: low-dose CDDP (0.5 mg/kg, intraperitoneal twice per week); G3: o-rMETase + low-dose CDDP (100 units per mouse, oral, twice per day + 0.5 mg/kg, intraperitoneal twice per week, respectively). Tumor volume and body weight were measured twice per week. The expression of Ki-67 was detected by immunohistochemistry to evaluate cell proliferation. RESULTS: The combination of o-rMETase and low-dose CDDP increased inhibition efficacy compared to low-dose CDDP monotherapy, on primary-tumor growth (p=0.032) and metastasis (p=0.002). CONCLUSION: The combination of o-rMETase with low-dose CDDP has future clinical potential for bladder cancer.


Assuntos
Liases de Carbono-Enxofre/uso terapêutico , Cisplatino/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Neoplasias da Bexiga Urinária/tratamento farmacológico , Neoplasias da Bexiga Urinária/patologia , Ensaios Antitumorais Modelo de Xenoenxerto , Administração Oral , Animais , Liases de Carbono-Enxofre/administração & dosagem , Liases de Carbono-Enxofre/farmacologia , Proliferação de Células/efeitos dos fármacos , Cisplatino/administração & dosagem , Cisplatino/farmacologia , Relação Dose-Resposta a Droga , Feminino , Humanos , Camundongos Nus , Metástase Neoplásica , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Carga Tumoral/efeitos dos fármacos
18.
Cancer Lett ; 492: 174-184, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32739322

RESUMO

Methionine addiction is a fundamental and general hallmark of cancer. Gene expression analysis showed that methionine restriction (MR) of methionine-addicted cancer cells increases TNF-related apoptosis-induced ligand receptor-2 (TRAIL-R2) expression. Here, we determined the effects of MR on TRAIL-R2 targeted therapy in pancreatic cancer by the TRAIL-R2 agonist tigatuzumab. Human pancreatic cancer cell lines were cultured in control or methionine-free medium. The effects of MR on TRAIL-R2 expression and sensitivity to tigatuzumab were evaluated in vitro. An orthotopic pancreatic cancer mouse model was established to evaluate the efficacy of MR using oral recombinant methioninase (o-rMETase), and the efficacy of tigatuzumab and their combination. MR enabled tigatuzumab-induced apoptosis, by increasing TRAIL-R2 expression in pancreatic cancer cells in vitro. The protein expression level of the melanoma-associated antigen MAGED2, which reduces TRAIL-R2 expression, was decreased by MR. In the orthotopic pancreatic cancer mouse model, o-rMETase increased TRAIL-R2 expression level in the tumors and enabled the antitumor efficacy of tigatuzumab. MR, effected by o-rMETase, enabled the efficacy of the TRAIL-R2 agonist tigatuzumab by increasing TRAIL-R2 expression in pancreatic cancer. Our results suggest that o-rMETase has clinical potential for treating pancreatic cancer.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Liases de Carbono-Enxofre/administração & dosagem , Neoplasias Pancreáticas/tratamento farmacológico , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/análise , Proteínas Adaptadoras de Transdução de Sinal/análise , Administração Oral , Animais , Antígenos de Neoplasias/análise , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Metionina/administração & dosagem , Metionina/sangue , Camundongos , Neoplasias Pancreáticas/química , Neoplasias Pancreáticas/patologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/agonistas , Proteínas Recombinantes/administração & dosagem
19.
Clin Pharmacol Drug Dev ; 9(8): 1003-1014, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32627420

RESUMO

ABP 798 is a proposed biosimilar to rituximab reference product (RP), an anti-CD20 monoclonal antibody. Pharmacokinetics (PK), pharmacodynamics (PD), and safety results from the comparative clinical study that evaluated the PK, PD, safety, efficacy, and immunogenicity of ABP 798 versus rituximab RP are presented here. Subjects with moderate to severe rheumatoid arthritis (RA) received 2 doses of ABP 798, United States-sourced RP (rituximab US) or European Union-sourced RP (rituximab EU), each consisting of two 1000-mg infusions 2 weeks apart. For the second dose (week 24), ABP 798- and rituximab EU-treated subjects received the same treatment; rituximab US-treated subjects transitioned to ABP 798. End points included area under the serum concentration-time curve from time 0 extrapolated to infinity and maximum observed serum concentration following the second infusion of the first dose (PK) and percentage of subjects with complete CD19+ cell depletion days 1-33 (PD). Primary analysis established PK similarity between ABP 798 and rituximab RP based on 90% confidence intervals of the adjusted geometric mean ratios being within a prespecified equivalence margin of 0.8 and 1.25. Complete CD19+ B-cell depletion on day 3 among groups confirmed PD similarity. These findings demonstrated PK/PD similarity between ABP 798 and rituximab RP in subjects with moderate to severe RA.


Assuntos
Antirreumáticos/farmacocinética , Artrite Reumatoide/tratamento farmacológico , Medicamentos Biossimilares/farmacocinética , Liases de Carbono-Enxofre/farmacocinética , Rituximab/farmacocinética , Adulto , Idoso , Antineoplásicos Imunológicos/administração & dosagem , Antineoplásicos Imunológicos/sangue , Antineoplásicos Imunológicos/farmacocinética , Antineoplásicos Imunológicos/farmacologia , Antirreumáticos/administração & dosagem , Antirreumáticos/sangue , Antirreumáticos/farmacologia , Área Sob a Curva , Medicamentos Biossimilares/administração & dosagem , Medicamentos Biossimilares/sangue , Medicamentos Biossimilares/farmacologia , Liases de Carbono-Enxofre/administração & dosagem , Liases de Carbono-Enxofre/sangue , Liases de Carbono-Enxofre/farmacologia , Método Duplo-Cego , Feminino , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Rituximab/administração & dosagem , Rituximab/sangue , Rituximab/farmacologia , Segurança , Índice de Gravidade de Doença , Equivalência Terapêutica , Resultado do Tratamento
20.
In Vivo ; 34(3 Suppl): 1593-1596, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32503816

RESUMO

The Covid-19 pandemic is a world-wide crisis without an effective therapy. While most approaches to therapy are using repurposed drugs that were developed for other diseases, it is thought that targeting the biology of the SARS-CoV-2 virus, which causes Covid-19, can result in an effective therapeutic treatment. The coronavirus RNA cap structure is methylated by two viral methyltransferases that transfer methyl groups from S-adenosylmethionine (SAM). The proper methylation of the virus depends on the level of methionine in the host to form SAM. Herein, we propose to restrict methionine availability by treating the patient with oral recombinant methioninase, aiming to treat Covid-19. By restricting methionine we not only interdict viral replication, which depends on the viral RNA cap methyaltion, but also inhibit the proliferation of the infected cells, which have an increased requirement for methionine. Most importantly, the virally-induced T-cell- and macrophage-mediated cytokine storm, which seems to be a significant cause for Covid-19 deaths, can also be inhibited by restricting methionine, since T-cell and macrophrage activation greatly increases the methionine requirement for these cells. The evidence reviewed here suggests that oral recombinant methioninase could be a promising treatment for coronavirus patients.


Assuntos
Antivirais/uso terapêutico , Betacoronavirus/efeitos dos fármacos , Liases de Carbono-Enxofre/uso terapêutico , Infecções por Coronavirus/tratamento farmacológico , Metionina/metabolismo , Pneumonia Viral/tratamento farmacológico , Capuzes de RNA/efeitos dos fármacos , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , RNA Viral/efeitos dos fármacos , Administração Oral , Antivirais/administração & dosagem , Proteínas de Bactérias/administração & dosagem , Proteínas de Bactérias/uso terapêutico , Betacoronavirus/fisiologia , COVID-19 , Liases de Carbono-Enxofre/administração & dosagem , Ensaios Clínicos como Assunto , Infecções por Coronavirus/complicações , Infecções por Coronavirus/imunologia , Síndrome da Liberação de Citocina/prevenção & controle , Humanos , Ativação Linfocitária/efeitos dos fármacos , Ativação de Macrófagos/efeitos dos fármacos , Metanálise como Assunto , Metilação/efeitos dos fármacos , Pandemias , Pneumonia Viral/complicações , Pneumonia Viral/imunologia , Pseudomonas putida/enzimologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico , S-Adenosilmetionina/metabolismo , SARS-CoV-2 , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA