Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.461
Filtrar
1.
J Clin Invest ; 130(9): 4921-4934, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32544087

RESUMO

Cachexia, a devastating wasting syndrome characterized by severe weight loss with specific losses of muscle and adipose tissue, is driven by reduced food intake, increased energy expenditure, excess catabolism, and inflammation. Cachexia is associated with poor prognosis and high mortality and frequently occurs in patients with cancer, chronic kidney disease, infection, and many other illnesses. There is no effective treatment for this condition. Hypothalamic melanocortins have a potent and long-lasting inhibitory effect on feeding and anabolism, and pathophysiological processes increase melanocortin signaling tone, leading to anorexia, metabolic changes, and eventual cachexia. We used 3 rat models of anorexia and cachexia (LPS, methylcholanthrene sarcoma, and 5/6 subtotal nephrectomy) to evaluate efficacy of TCMCB07, a synthetic antagonist of the melanocortin-4 receptor. Our data show that peripheral treatment using TCMCB07 with intraperitoneal, subcutaneous, and oral administration increased food intake and body weight and preserved fat mass and lean mass during cachexia and LPS-induced anorexia. Furthermore, administration of TCMCB07 diminished hypothalamic inflammatory gene expression in cancer cachexia. These results suggest that peripheral TCMCB07 treatment effectively inhibits central melanocortin signaling and therefore stimulates appetite and enhances anabolism, indicating that TCMCB07 is a promising drug candidate for treating cachexia.


Assuntos
Caquexia/tratamento farmacológico , Receptor Tipo 4 de Melanocortina/antagonistas & inibidores , Insuficiência Renal Crônica/tratamento farmacológico , Sarcoma Experimental/tratamento farmacológico , Animais , Apetite/efeitos dos fármacos , Caquexia/etiologia , Caquexia/metabolismo , Caquexia/patologia , Masculino , Ratos , Ratos Sprague-Dawley , Receptor Tipo 4 de Melanocortina/metabolismo , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia , Sarcoma Experimental/complicações , Sarcoma Experimental/metabolismo , Sarcoma Experimental/patologia
2.
Acta Biomater ; 106: 256-266, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32058082

RESUMO

Here, we describe innovative synthesis of well-defined biocompatible N-(2-hydroxypropyl) methacrylamide (HPMA)-based polymer carriers and their drug conjugates with pirarubicin intended for controlled drug delivery and pH-triggered drug activation in tumor tissue. Polymer carrier synthesis was optimized to obtain well-defined linear HPMA-based polymer precursor with dispersity close to 1 and molar mass close to renal threshold with minimal synthesis steps. The developed synthesis enables preparation of tailored polymer nanomedicines with highly enhanced biological behavior in vivo, especially the biodistribution, urine elimination, tumor accumulation and anticancer activity. STATEMENT OF SIGNIFICANCE: The manuscript reports on novel synthesis and detailed physicochemical characterization and in vivo evaluation of well-defined biocompatible hydrophilic copolymers based on N-(2-hydroxypropyl)methacrylamide (HPMA) and their drug conjugates with pirarubicin enabling controlled drug delivery and pH-triggered drug activation in tumor tissue. Polymer carrier synthesis was optimized to obtain well-defined linear HPMA-based polymer precursor with minimal synthesis steps using controlled polymerization. Compared to previously published HPMA-based polymer drug conjugates whose polymer carriers were prepared by classical route via free radical polymerization, the newly prepared polymer drug conjugates exhibited enhanced biological behavior in vivo, especially the prolonged blood circulation, urine elimination, tumor accumulation and excellent anticancer activity. We believe that the newly prepared well-defined polymer conjugates could significantly enhance tumor therapy in humans.


Assuntos
Acrilamidas/uso terapêutico , Antineoplásicos/uso terapêutico , Doxorrubicina/análogos & derivados , Sarcoma Experimental/tratamento farmacológico , Acrilamidas/síntese química , Acrilamidas/farmacocinética , Animais , Antineoplásicos/síntese química , Antineoplásicos/farmacocinética , Caproatos/síntese química , Caproatos/farmacocinética , Caproatos/uso terapêutico , Linhagem Celular Tumoral , Doxorrubicina/síntese química , Doxorrubicina/farmacocinética , Doxorrubicina/uso terapêutico , Sistemas de Liberação de Medicamentos , Camundongos , Nanomedicina/métodos , Polimerização
3.
J Control Release ; 304: 242-258, 2019 06 28.
Artigo em Francês | MEDLINE | ID: mdl-31071376

RESUMO

Cancer-Immunotherapy was the most exciting topic. However, either insensitivity due to singleness of therapeutic target or immune evasion leads to the failure of the treatment. Ingenol-3-mebutate (I3A) can inhibit cancer through synergy between immunotherapy and chemotherapy, however, the speculation and accurate mechanism haven't been confirmed in vivo limited by its hydrophobicity and pH-instability, which also hindered its clinical translation. Herein we developed a polymeric micelle with 'acidic core' provided by single alcoholic hydroxyl (-CH(CH3)-OH) encapsulating I3A (I3A-PM), which successfully overcome the aforementioned problems and reduce the toxicity in vivo. To test the synergy, S180 tumor-bearing mice were subjected to I3A-PM through intravenous and intratumoral administration, we found I3A-PM presented significant antitumor effect, and promoted Th1 polarization by upregulating the level of Th1 cytokines (IL-12, IL-2, IFN-γ and TNF-α), and accelerated the expansion of CD4+ and CD8+ T cells, meanwhile, I3A-PM depleted regulatory T cells, Th2 cytokine IL-6 through inhibiting TGF-ß signaling pathway. Furthermore, we appealed to virtual screening of tumor target, and found a new pathway of I3A as a TGF-ß receptor type I inhibitor to improve immunostimulatory effects. These results demonstrated I3A-PM as a promising nanoagent for cancer immunotherapy strategy. The synergistic therapeutic effects are encouraged to further evaluate in different cancer model compared with commercial products to facilitate research finding (I3A-PM) entering the clinic.


Assuntos
Diterpenos/administração & dosagem , Imunoterapia/métodos , Nanopartículas , Sarcoma Experimental/tratamento farmacológico , Animais , Citocinas/metabolismo , Diterpenos/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Micelas , Nanotecnologia , Polímeros/química , Sarcoma Experimental/imunologia , Transdução de Sinais/efeitos dos fármacos
4.
Invest New Drugs ; 37(5): 1044-1051, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30680583

RESUMO

Obtustatin, isolated from the Levantine Viper snake venom (Macrovipera lebetina obtusa -MLO), is the shortest known monomeric disintegrin shown to specifically inhibit the binding of the α1ß1 integrin to collagen IV. Its oncostatic effect is due to the inhibition of angiogenesis, likely through α1ß1 integrin inhibition in endothelial cells. To explore the therapeutic potential of obtustatin, we studied its effect in S-180 sarcoma-bearing mice model in vivo as well as in human dermal microvascular endothelial cells (HMVEC-D) in vitro, and tested anti-angiogenic activity in vivo using the chick embryo chorioallantoic membrane assay (CAM assay). Our in vivo results show that obtustatin inhibits tumour growth by 33%. The expression of vascular endothelial growth factor (VEGF) increased after treatment with obtustatin, but the level of expression of caspase 8 did not change. In addition, our results demonstrate that obtustatin inhibits FGF2-induced angiogenesis in the CAM assay. Our in vitro results show that obtustatin does not exhibit cytotoxic activity in HMVEC-D cells in comparison to in vivo results. Thus, our findings disclose that obtustatin might be a potential candidate for the treatment of sarcoma in vivo with low toxicity.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos/farmacologia , Neovascularização Patológica/tratamento farmacológico , Sarcoma Experimental/tratamento farmacológico , Venenos de Víboras/farmacologia , Animais , Apoptose , Proliferação de Células , Embrião de Galinha , Membrana Corioalantoide , Integrina alfa1beta1/antagonistas & inibidores , Camundongos , Neovascularização Patológica/patologia , Sarcoma Experimental/irrigação sanguínea , Sarcoma Experimental/patologia , Células Tumorais Cultivadas
5.
Cancer Chemother Pharmacol ; 83(2): 361-374, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30506269

RESUMO

Macrophage-mediated drug delivery system has emerged and gained wide interest as a novel strategy for cancer treatment. Among them, RAW264.7 cell was commonly used as the macrophage model for antitumor drug loading and delivery. However, this cell line was a macrophage-like cancerous cell with both immunogenicity and pro-tumorigenic properties, which may interfere with the positive response of the host immune system to developed tumor. Thus, the safety and efficacy of the RAW264.7 cell line as a drug carrier for cancer therapy remain questionable. Here, we constructed doxorubicin-loaded RAW264.7 cells and examined its antitumor efficacy in S180 tumor-bearing mice. The bio-distribution of RAW264.7 cells was determined by in vivo imaging technique, showing a high accumulation level of RAW264.7 cells in mice livers, spleens, and thymuses. A phenomenon of accelerated tumor growth was observed in mice treated with doxorubicin-loaded RAW264.7 cells. Thereafter, the effect of frequency, dose, and viability of injected RAW264.7 cells on S180 tumor growth was further investigated. The underlying mechanism was confirmed, attributing to the immune tolerance induced by excessive RAW264.7 cells. Our findings emphasized the latent limitation of RAW264.7 cells as drug carrier in current researches, and provided an experimental basis for the clinical safety of cell-mediated drug delivery system.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Materiais Biomiméticos/administração & dosagem , Doxorrubicina/administração & dosagem , Sistemas de Liberação de Medicamentos , Macrófagos/transplante , Sarcoma Experimental/tratamento farmacológico , Animais , Antibióticos Antineoplásicos/farmacocinética , Apoptose , Proliferação de Células , Doxorrubicina/farmacocinética , Liberação Controlada de Fármacos , Humanos , Macrófagos/citologia , Masculino , Camundongos , Células RAW 264.7 , Sarcoma Experimental/patologia , Células Tumorais Cultivadas
6.
Anticancer Agents Med Chem ; 19(3): 365-374, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30523769

RESUMO

OBJECTIVE: The aim of this study is to investigate the inhibitory effect of camptothecin derivative 3j on Non-Small Cell Lung Cancer (NSCLCs) cells and the potential anti-tumor mechanisms. BACKGROUND: Camptothecin compounds are considered as the third largest natural drugs which are widely investigated in the world and they suffered restriction because of serious toxicity, such as hemorrhagic cystitis and bone marrow suppression. METHODS: Using cell proliferation assay and S180 tumor mice model, a series of 20(S)-O-substituted benzoyl 7- ethylcamptothecin compounds were screened and evaluated the antitumor activities in vitro and in vivo. Camptothecin derivative 3j was selected for further study using flow cytometry in NSCLCs cells. Cell cycle related protein cyclin A2, CDK2, cyclin D and cyclin E were detected by Western Blot. Then, computer molecular docking was used to confirm the interaction between 3j and Topo I. Also, DNA relaxation assay and alkaline comet assay were used to investigate the mechanism of 3j on DNA damage. RESULTS: Our results demonstrated that camptothecin derivative 3j showed a greater antitumor effect in eleven 20(S)-O-substituted benzoyl 7-ethylcamptothecin compounds in vitro and in vivo. The IC50 of 3j was 1.54± 0.41 µM lower than irinotecan with an IC50 of 13.86±0.80 µM in NCI-H460 cell, which was reduced by 8 fold. In NCI-H1975 cell, the IC50 of 3j was 1.87±0.23 µM lower than irinotecan (IC50±SD, 5.35±0.38 µM), dropped by 1.8 fold. Flow cytometry analysis revealed that 3j induced significant accumulation in a dose-dependent manner. After 24h of 3j (10 µM) treatment, the percentage of NCI-H460 cell in S-phase significantly increased (to 93.54 ± 4.4%) compared with control cells (31.67 ± 3.4%). Similarly, the percentage of NCI-H1975 cell in Sphase significantly increased (to 83.99 ± 2.4%) compared with control cells (34.45 ± 3.9%) after treatment with 10µM of 3j. Moreover, increased levels of cyclin A2, CDK2, and decreased levels of cyclin D, cyclin E further confirmed that cell cycle arrest was induced by 3j. Furthermore, molecular docking studies suggested that 3j interacted with Topo I-DNA and DNA-relaxation assay simultaneously confirmed that 3j suppressed the activity of Topo I. Research on the mechanism showed that 3j exhibited anti-tumour activity via activating the DNA damage response pathway and suppressing the repair pathway in NSCLC cells. CONCLUSION: Novel camptothecin derivative 3j has been demonstrated as a promising antitumor agent and remains to be assessed in further studies.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Camptotecina/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Dano ao DNA , DNA Topoisomerases Tipo I/metabolismo , Inibidores da Topoisomerase I/farmacologia , Animais , Antineoplásicos Fitogênicos/síntese química , Antineoplásicos Fitogênicos/química , Camptotecina/síntese química , Camptotecina/química , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Camundongos , Estrutura Molecular , Sarcoma Experimental/tratamento farmacológico , Sarcoma Experimental/metabolismo , Sarcoma Experimental/patologia , Relação Estrutura-Atividade , Inibidores da Topoisomerase I/síntese química , Inibidores da Topoisomerase I/química , Células Tumorais Cultivadas
7.
PLoS One ; 13(8): e0201220, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30080874

RESUMO

In recent years, immunogenic cell death (ICD) has emerged as a revolutionary concept in the development of novel anticancer therapies. This particular form of cell death is able, through the spatiotemporally defined emission of danger signals by the dying cell, to induce an effective antitumor immune response, allowing the immune system to recognize and eradicate malignant cells. To date, only a restricted number of chemotherapeutics can trigger ICD of cancer cells. We previously reported that a peptide, called RT53, spanning the heptad leucine repeat region of the survival protein AAC-11 fused to a penetrating sequence, selectively induces cancer cell death in vitro and in vivo. Interestingly, B16F10 melanoma cells treated by RT53 were able to mediate anticancer effects in a tumor vaccination model. Stimulated by this observation, we investigated whether RT53 might mediate ICD of cancer cells. Here, we report that RT53 treatment induces all the hallmarks of immunogenic cell death, as defined by the plasma membrane exposure of calreticulin, release of ATP and the exodus of high-mobility group box 1 protein (HMGB1) from dying cancer cells, through a non-regulated, membranolytic mode of action. In a prophylactic mouse model, vaccination with RT53-treated fibrosarcomas prevented tumor growth at the challenge site. Finally, local intratumoral injection of RT53 into established cancers led to tumor regression together with T-cell infiltration and the mounting of an inflammatory response in the treated animals. Collectively, our results strongly suggest that RT53 can induce bona fide ICD of cancer cells and illustrate its potential use as a novel antitumor and immunotherapeutic strategy.


Assuntos
Antineoplásicos/farmacologia , Proteínas Reguladoras de Apoptose/farmacologia , Morte Celular/efeitos dos fármacos , Morte Celular/imunologia , Peptídeos Penetradores de Células/farmacologia , Proteínas Nucleares/farmacologia , Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Antineoplásicos/química , Proteínas Reguladoras de Apoptose/química , Linhagem Celular Tumoral , Peptídeos Penetradores de Células/química , Células HL-60 , Humanos , Células Jurkat , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/química , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacologia , Peptídeos/química , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/farmacologia , Sarcoma Experimental/tratamento farmacológico , Sarcoma Experimental/imunologia , Sarcoma Experimental/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Curr Med Sci ; 38(4): 697-703, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30128881

RESUMO

This study examined the effect of saponins from Tupistra chinensis Bak (STCB) on the growth of sarcoma S-180 cells in vitro and in mouse xenografts as well as the underlying mechanisms. Cell proliferation was assessed by MTT assay. Cell cycle distribution was determined by flow cytometry. Sarcoma S-180 tumor-bearing mice were treated with different doses of STCB with 10 µg/mL 5-fluorouracil (5-Fu) as a positive control. The activity of nuclear factor (NF)-κB was detected by gel mobility shift assay. The mRNA level of NF-κB was determined by real-time quantitative RT-PCR. The results showed that in vitro STCB inhibited the growth of S-180 cells in a concentration-dependent manner, which was accompanied by cell cycle arrest at S-phase. In vivo STCB significantly inhibited the growth of S-180 tumor mouse xenografts in a dose-dependent manner with apparent induction of cell apoptosis. Moreover, STCB inhibited the activity of NF-κB p65 and reduced the expression of NF-κB p65 mRNA in mouse xenografts. It was concluded that STCB inhibits the proliferation and cell cycle progression of S-180 cells by suppressing NF-κB signaling in mouse xenografts. Our findings suggest STCB is a promising agent for the treatment of sarcoma.


Assuntos
Antineoplásicos/uso terapêutico , Saponinas/uso terapêutico , Sarcoma Experimental/tratamento farmacológico , Fator de Transcrição RelA/metabolismo , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Asparagaceae/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Saponinas/farmacologia , Sarcoma Experimental/metabolismo , Transdução de Sinais , Fator de Transcrição RelA/genética
9.
Theranostics ; 8(7): 1782-1797, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29556356

RESUMO

Gold nanoparticles (AuNPs) are emerging as promising agents for both cancer therapy and computed tomography (CT) imaging. AuNPs absorb x-rays and subsequently release low-energy, short-range photoelectrons during external beam radiation therapy (RT), increasing the local radiation dose. When AuNPs are near tumor vasculature, the additional radiation dose can lead to increased vascular permeability. This work focuses on understanding how tumor vascular permeability is influenced by AuNP-augmented RT, and how this effect can be used to improve the delivery of nanoparticle chemotherapeutics. Methods: Dual-energy CT was used to quantify the accumulation of both liposomal iodine and AuNPs in tumors following AuNP-augmented RT in a mouse model of primary soft tissue sarcoma. Mice were injected with non-targeted AuNPs, RGD-functionalized AuNPs (vascular targeting), or no AuNPs, after which they were treated with varying doses of RT. The mice were injected with either liposomal iodine (for the imaging study) or liposomal doxorubicin (for the treatment study) 24 hours after RT. Increased tumor liposome accumulation was assessed by dual-energy CT (iodine) or by tracking tumor treatment response (doxorubicin). Results: A significant increase in vascular permeability was observed for all groups after 20 Gy RT, for the targeted and non-targeted AuNP groups after 10 Gy RT, and for the vascular-targeted AuNP group after 5 Gy RT. Combining targeted AuNPs with 5 Gy RT and liposomal doxorubicin led to a significant tumor growth delay (tumor doubling time ~ 8 days) compared to AuNP-augmented RT or chemotherapy alone (tumor doubling time ~3-4 days). Conclusions: The addition of vascular-targeted AuNPs significantly improved the treatment effect of liposomal doxorubicin after RT, consistent with the increased liposome accumulation observed in tumors in the imaging study. Using this approach with a liposomal drug delivery system can increase specific tumor delivery of chemotherapeutics, which has the potential to significantly improve tumor response and reduce the side effects of both RT and chemotherapy.


Assuntos
Antineoplásicos/administração & dosagem , Vasos Sanguíneos/efeitos da radiação , Doxorrubicina/administração & dosagem , Ouro/metabolismo , Permeabilidade/efeitos da radiação , Sarcoma/tratamento farmacológico , Sarcoma/radioterapia , Animais , Ouro/administração & dosagem , Iodo/administração & dosagem , Lipossomos/administração & dosagem , Nanopartículas Metálicas/administração & dosagem , Camundongos , Oligopeptídeos/administração & dosagem , Oligopeptídeos/metabolismo , Sarcoma/diagnóstico por imagem , Sarcoma Experimental/diagnóstico por imagem , Sarcoma Experimental/tratamento farmacológico , Sarcoma Experimental/radioterapia , Tomografia Computadorizada por Raios X , Resultado do Tratamento
10.
Sci Rep ; 8(1): 2194, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29391414

RESUMO

A newly designed organoselenium compound, methyl substituted umbelliferone selenocyanate (MUS), was synthesized as a primary hit against the myelotoxic activity of carboplatin. MUS was administered at 6 mg/kg b.wt, p.o. in concomitant and pretreatment schedules with carboplatin (12 mg/kg b.wt, i.p. for 10 days) in female Swiss albino mouse. MUS treatment reduced (P < 0.001) the percentage of chromosomal aberrations, micronuclei formation, DNA damage and apoptosis in murine bone marrow cells and also enhanced (P < 0.001) the bone marrow cell proliferation of the carboplatin-treated mice. These activities cumulatively restored the viable bone marrow cell count towards normalcy. Myeloprotection by MUS was achieved, in part, due to a significant reduction in the ROS/RNS formation and restoration of glutathione redox pool. Additionally, MUS synergistically enhanced the cytotoxicity of carboplatin against two human cancer cell lines (MCF-7 and Colo-205). Furthermore, MUS can effectively potentiate the antitumour activity of carboplatin against two murine cancers (Dalton's Lymphoma and Sarcoma-180) in vivo. These preclinical findings clearly indicate that MUS can improve the therapeutic index of carboplatin and ensures more effective therapeutic strategy against cancer for clinical development.


Assuntos
Antineoplásicos/farmacologia , Cumarínicos/química , Desenho de Fármacos , Linfoma/tratamento farmacológico , Mielopoese/efeitos dos fármacos , Compostos Organosselênicos/farmacologia , Sarcoma Experimental/tratamento farmacológico , Animais , Antineoplásicos/química , Antioxidantes/química , Antioxidantes/farmacologia , Apoptose , Proliferação de Células , Quimioterapia Adjuvante , Aberrações Cromossômicas , Dano ao DNA/efeitos dos fármacos , Feminino , Peroxidação de Lipídeos , Linfoma/patologia , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Sarcoma Experimental/patologia , Células Tumorais Cultivadas
11.
J BUON ; 23(6): 1904-1911, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30610820

RESUMO

PURPOSE: To examine the antitumor effects of Mebendazole (MZ) in a model of experimental fibrosarcoma induced by inoculation of BHK-21/C13 cells in Syrian golden hamster. METHODS: Hamsters were inoculated with a suspension of BHK cells by subcutaneous injection and randomly divided into 5 experimental and 2 control groups. Treatment started on the 10th day after inoculation, when the tumor grew to a diameter of 5mm. The experimental design was based on distributing the total amount of drug MZ(z) in different protocols and approaches (oral/intraperitoneal) to the 5 experimental groups. The positive control group received doxorubicin intraperitoneally. Negative control group received olive oil orally. The total amount of MZ(z) was chosen to be the highest for the animal to survive during the experiment. For antitumor effect evaluation, the main parameters were tumor size, number of mitoses, cytochrome-C immunopositivity and tumor tissue morphology incuding cytoarchitecture and percentage of preserved tumor tissue in stereologically reconstructed tumor mass. RESULTS: The results of this study showed absence of objective MZ antitumor effect on experimental fibrosarcoma. MZ does not exhibit activity similar to DNA-damaging agents on the fibrosarcoma model. CONCLUSIONS: It might be postulated that soft tissue tumors on animal models could show high level of resistance to MZ effect.


Assuntos
Proliferação de Células/efeitos dos fármacos , Reposicionamento de Medicamentos/métodos , Fibrossarcoma/patologia , Mebendazol/uso terapêutico , Sarcoma Experimental/patologia , Moduladores de Tubulina/uso terapêutico , Animais , Fibrossarcoma/tratamento farmacológico , Mesocricetus , Sarcoma Experimental/tratamento farmacológico
12.
Cancer Lett ; 416: 1-10, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29104145

RESUMO

Inhibition of the VEGF/VEGF receptor (VEGFR) and angiopoietin-2 (Ang-2)/TEK receptor tyrosine kinase (Tie-2) pathway is a potential target for tumor angiogenesis. We previously showed that a peptide AS16 which dually inhibits VEGFR/Ang-2 could reduce the tumor growth and decrease the number of microvessels in tumor. However, its short circulating half-life in the serum limits its clinical applications. In this study, as an effort to prolong the short in vivo half-life of AS16, we designed a fusion protein containing peptide AS16 and an IgG Fc fragment. Pharmacokinetic study also revealed that AS16-Fc has a prolonged circulating half-life of about 231 min in rats. We examined the effects of treatment on the tumor vasculature and immune cell populations, tumor growth, in both the MCA-205 and S180 tumor models. We found that AS16-Fc dramatically reduced tumor volume, vascular density and tumor-associated macrophages. Macrophages were identified as potential novel targets following anti-angiogenic therapy, our findings imply a novel role for anti-angiogenic peptide AS16-Fc. These findings indicate that AS16-Fc could be more effective on inhibiting tumor growth angiogenesis and tumor immune microenvironment than that of peptide AS16.


Assuntos
Imunoconjugados/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Neovascularização Patológica/prevenção & controle , Sarcoma Experimental/tratamento farmacológico , Inibidores da Angiogênese/química , Inibidores da Angiogênese/farmacocinética , Inibidores da Angiogênese/farmacologia , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Imunoconjugados/química , Imunoconjugados/farmacocinética , Fragmentos Fc das Imunoglobulinas/química , Imunoglobulina G/química , Ativação de Macrófagos/imunologia , Macrófagos/classificação , Macrófagos/imunologia , Camundongos , Peptídeos/química , Ratos Sprague-Dawley , Sarcoma Experimental/irrigação sanguínea , Sarcoma Experimental/imunologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
13.
J Med Chem ; 60(24): 10220-10230, 2017 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-29202233

RESUMO

Novel hybrid molecule containing 2-mercaptoethylamine was synthesized starting from O-propyloxime-N-propoxy bacteriopurpurinimide (dipropoxy-BPI), which was readily oxidized in oxygen atmosphere yielding the corresponding disulfide analogue (disulfide-BPI). Spectral, photophysical, photodynamic, and biological properties of compound were properly evaluated. Compounds bearing disulfide moiety can directly interact with glutathione (GSH), thereby reducing its intracellular concentration. Indeed, mice sarcoma S37 cell line was treated in vitro with disulfide-BPI, yielding a CC50 value of 0.05 ± 0.005 µM. A relatively high level of singlet oxygen was detected. It was demonstrated (by fluorescence) that the PS was rapidly accumulated in a cancer nest (S37) at a relatively high level after 2 h upon intravenous administration. After 24 h, no traces of the molecule were detected in the tumor mass. Moreover, high photodynamic efficiency was demonstrated at doses of 150-300 J/cm2 against two different in vivo tumor models, achieving 100% regression of cancer growth.


Assuntos
Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Administração Intravenosa , Animais , Linhagem Celular Tumoral , Técnicas de Química Sintética , Dissulfetos/química , Feminino , Glutationa/metabolismo , Camundongos , Fármacos Fotossensibilizantes/síntese química , Fármacos Fotossensibilizantes/farmacocinética , Porfirinas/química , Ratos , Sarcoma Experimental/tratamento farmacológico , Oxigênio Singlete/química , Distribuição Tecidual
14.
PLoS One ; 12(8): e0183783, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28854214

RESUMO

Hemocyanin is a multifunctional glycoprotein, which also plays multiple roles in immune defense. While it has been demonstrated that hemocyanin from some mollusks can induce potent immune response and is therefore undergoing clinical trials to be used in anti-tumor immunotherapy, little is currently known about how hemocyanin from arthropods affect tumors. In this study we investigated the anti-tumor activity of hemocyanin from Litopenaeus vannamei on Sarcoma-180 (S180) tumor-bearing mice model. Eight days treatment with 4mg/kg bodyweight of hemocyanin significantly inhibited the growth of S180 up to 49% as compared to untreated. Similarly, histopathology analysis showed a significant decrease in tumor cell number and density in the tissues of treated mice. Moreover, there was a significant increase in immune organs index, lymphocyte proliferation, NK cell cytotoxic activity and serum TNF-α level, suggesting that hemocyanin could improve the immunity of the S180 tumor-bearing mice. Additionally, there was a significant increase in superoxide dismutase (SOD) activity and a decrease in the level of malondialdehyde (MDA) in serum and liver, which further suggest that hemocyanin improved the anti-oxidant ability of the S180 tumor-bearing mice. Collectively, our data demonstrated that L. vannamei hemocyanin had a significant antitumor activity in mice.


Assuntos
Proteínas de Artrópodes/farmacologia , Hemocianinas/farmacologia , Penaeidae/metabolismo , Sarcoma Experimental/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Células HeLa , Humanos , Immunoblotting , Fígado/efeitos dos fármacos , Fígado/metabolismo , Linfócitos/citologia , Linfócitos/efeitos dos fármacos , Masculino , Malondialdeído/sangue , Malondialdeído/metabolismo , Camundongos , Sarcoma Experimental/metabolismo , Superóxido Dismutase/sangue , Superóxido Dismutase/metabolismo , Carga Tumoral/efeitos dos fármacos , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/metabolismo
15.
Drug Deliv Transl Res ; 7(5): 642-653, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28695431

RESUMO

Tocopheryl polyethylene glycol 1000 succinate (TPGS) is considered a promising surfactant, but its high critical micelle concentration (CMC) limits its application. Cholesterol is hydrophobic, can act as a tumor-targeting ligand, and has strong binding ability with taxoids. Based on this information, we coupled cholesterol with TPGS to synthesize cholesterol-coupled TPGS (TPGS-CHMC), which had a lower CMC than pure TPGS. The TPGS-CHMC was used to prepare micelles loading with docetaxel (DTX) by a self-assembly method. DTX-loaded TPGS-CHMC micelles were globule-shaped, 13.3 ± 2.0 nm in size, and had a zeta potential of -4.66 ± 0.41 mv. In vitro release studies demonstrated the delayed release property of the micelles, which also had a relatively high encapsulation efficiency and drug loading content of 99.2 and 3.20%, respectively. Furthermore, the micelles were stable in vitro at a dilution of 100-fold. In vivo antitumor studies showed that the DTX-loaded TPGS-CHMC micelles significantly enhanced the antitumor activity of DTX in S180 tumor-bearing mice. Interestingly, the blank TPGS-CHMC micelles also showed antitumor activity. Our results demonstrate that TPGS-CHMC is a promising system for DTX delivery that may be suitable for other hydrophobic antitumor drugs.


Assuntos
Colesterol/química , Sarcoma Experimental/tratamento farmacológico , Taxoides/administração & dosagem , Vitamina E/química , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Química Farmacêutica , Docetaxel , Camundongos , Micelas , Tamanho da Partícula , Taxoides/química , Taxoides/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Integr Biol (Camb) ; 9(6): 555-565, 2017 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-28513646

RESUMO

Magnetic liposome-mediated combined chemotherapy and hyperthermia is gaining importance as an effective therapeutic modality for cancer. However, control and maintenance of optimum hyperthermia are major challenges in clinical settings due to the overheating of tissues. To overcome this problem, we developed a novel magnetic liposomes formulation co-entrapping a dextran coated biphasic suspension of La0.75Sr0.25MnO3 (LSMO) and iron oxide (Fe3O4) nanoparticles for self-controlled hyperthermia and chemotherapy. However, the general apprehension about biocompatibility and safety of the newly developed formulation needs to be addressed. In this work, in vitro and in vivo biocompatibility and therapeutic evaluation studies of the novel magnetic liposomes are reported. Biocompatibility study of the magnetic liposomes formulation was carried out to evaluate the signs of preliminary systemic toxicity, if any, following intravenous administration of the magnetic liposomes in Swiss mice. Therapeutic efficacy of the magnetic liposomes formulation was evaluated in the fibrosarcoma tumour bearing mouse model. Fibrosarcoma tumour-bearing mice were subjected to hyperthermia following intratumoral injection of single or double doses of the magnetic liposomes with or without chemotherapeutic drug paclitaxel. Hyperthermia (three spurts, each at 3 days interval) with drug loaded magnetic liposomes following single dose administration reduced the growth of tumours by 2.5 fold (mean tumour volume 2356 ± 550 mm3) whereas the double dose treatment reduced the tumour growth by 3.6 fold (mean tumour volume 1045 ± 440 mm3) compared to their corresponding control (mean tumour volume 3782 ± 515 mm3). At the end of the tumour efficacy studies, the presence of MNPs was studied in the remnant tumour tissues and vital organs of the mice. No significant leaching or drainage of the magnetic liposomes during the study was observed from the tumour site to the other vital organs of the body, suggesting again the potential of the novel magnetic liposomes formulation for possibility of developing as an effective modality for treatment of drug resistant or physiologically vulnerable cancer.


Assuntos
Hipertermia Induzida/métodos , Lipossomos/uso terapêutico , Magnetismo , Neoplasias/terapia , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Materiais Biocompatíveis/uso terapêutico , Linhagem Celular Tumoral , Terapia Combinada , Feminino , Humanos , Lipossomos/administração & dosagem , Lipossomos/toxicidade , Nanopartículas de Magnetita/administração & dosagem , Nanopartículas de Magnetita/uso terapêutico , Nanopartículas de Magnetita/toxicidade , Teste de Materiais , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Paclitaxel/administração & dosagem , Sarcoma Experimental/tratamento farmacológico , Sarcoma Experimental/metabolismo , Sarcoma Experimental/terapia , Distribuição Tecidual
17.
Oncotarget ; 8(25): 40713-40723, 2017 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-28489574

RESUMO

Angiogenesis is essential for tumor growth and metastasis, controlling angiogenesis is a promising strategy in cancer treatment. However, thus farther severe side effects of anti-angiogenic drugs have been rather demonstrated, stimulating interest in seeking novel targets of anti-angiogenesis. Neurokinin receptors, also known as tachykinin receptors, are usually considered as drug targets due to diverse physiological functions and their tractability. Although Neurokinin B, the selective natural agonist of neurokinin-3 receptor, have been shown to exhibit anti-angiogenesis activity, the effect and mechanism of neurokinin-3 receptor-mediated angiogenesis still remains unclear. In the present study, we demonstrated that [Mephe7]NKB, an analogue of NKB, possess significant anti-angiogenic effect on CAM. Furthermore, by introducing the tumor angiogenesis homing sequence (NGR), we designed and synthesized two novel agonist analogues of NK3R, NK3R-A1 and NK3R-A2. Both of the two analogues exhibit more efficient anti-migration effect on HUVECs by activating NK3R in vitro, and showed potent antitumor activities with no significant side effects in vivo. Taken together, our results illuminated that NK3R might be a potential novel target for the anti-angiogenesis therapy. Notably, NK3R-A1 might be used as a template for the development of the anti-tumor drugs on the basis of the anti-angiogenesis strategy.


Assuntos
Inibidores da Angiogênese/farmacologia , Neurocinina B/farmacologia , Receptores da Neurocinina-3/agonistas , Sarcoma Experimental/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Embrião de Galinha , Membrana Corioalantoide/irrigação sanguínea , Membrana Corioalantoide/efeitos dos fármacos , Membrana Corioalantoide/metabolismo , Feminino , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos Endogâmicos BALB C , Neurocinina B/análogos & derivados , Receptores da Neurocinina-3/metabolismo , Sarcoma Experimental/irrigação sanguínea , Sarcoma Experimental/metabolismo , Carga Tumoral/efeitos dos fármacos
18.
Cancer Res ; 77(6): 1296-1309, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28082401

RESUMO

Adoptive immunotherapy utilizing chimeric antigen receptor (CAR) T cells has demonstrated high success rates in hematologic cancers, but results against solid malignancies have been limited to date, due in part to the immunosuppressive tumor microenvironment. Activation of the 4-1BB (CD137) pathway using an agonistic α-4-1BB antibody is known to provide strong costimulatory signals for augmenting and diversifying T-cell responses. We therefore hypothesized that a combination of α-4-1BB and CAR T-cell therapy would result in improved antitumor responses. Using a human-Her2 self-antigen mouse model, we report here that α-4-1BB significantly enhanced CAR T-cell efficacy directed against the Her2 antigen in two different established solid tumor settings. Treatment also increased the expression of IFNγ and the proliferation marker Ki67 in tumor-infiltrating CAR T cells when combined with α-4-1BB. Strikingly, α-4-1BB significantly reduced host immunosuppressive cells at the tumor site, including regulatory T cells and myeloid-derived suppressor cells, correlating with an increased therapeutic response. We conclude that α-4-1BB has a multifunctional role for enhancing CAR T-cell responses and that this combination therapy has high translational potential, given current phase I/II clinical trials with α-4-1BB against various types of cancer. Cancer Res; 77(6); 1296-309. ©2017 AACR.


Assuntos
Anticorpos Monoclonais/farmacologia , Neoplasias do Colo/terapia , Imunoterapia Adotiva , Neoplasias Mamárias Experimentais/tratamento farmacológico , Receptores de Antígenos de Linfócitos T/imunologia , Sarcoma Experimental/tratamento farmacológico , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Citotoxicidade Imunológica/imunologia , Feminino , Humanos , Ativação Linfocitária , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptor ErbB-2/imunologia , Receptor ErbB-2/metabolismo , Sarcoma Experimental/imunologia , Sarcoma Experimental/patologia , Células Tumorais Cultivadas , Microambiente Tumoral/imunologia
19.
Vet Comp Oncol ; 15(3): 754-763, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26991424

RESUMO

Spontaneously occurring soft tissue sarcoma (STS) is relatively common in canine cancer patients. Because of the similarities to human disease, canine STSs are a valuable and readily available resource for the study of new therapeutics. In this study, a canine patient-derived xenograft (PDX) model, CDX-STS2, was established. The CDX-STS2 model was engrafted and expanded for systemic administration studies with chemotherapeutic agents commonly used to treat STS, including doxorubicin, docetaxel and gemcitabine. Immunohistochemistry for drug-specific biomarkers and tumour growth measurement revealed tumour sensitivity to doxorubicin and docetaxel, whereas gemcitabine had no effect on tumour growth. Although many human PDX tumour models have been established, relatively few canine PDX models have been reported to date. CDX-STS2 represents a new STS PDX research model of canine origin that will be useful in bridging preclinical research with clinical studies of STS in pet dogs.


Assuntos
Transplante de Neoplasias/veterinária , Sarcoma Experimental/tratamento farmacológico , Sarcoma/veterinária , Animais , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais , Modelos Animais de Doenças , Cães , Xenoenxertos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias/métodos , Sarcoma/tratamento farmacológico , Sarcoma/patologia , Sarcoma Experimental/patologia
20.
Mol Pharm ; 13(12): 4106-4115, 2016 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-27934482

RESUMO

Many conjugates of water-soluble polymers with biologically active molecules were developed during the last two decades. Although, therapeutic effects of these conjugates are affected by the properties of carriers, the properties of the attached drugs appear more important than the same carrier polymer in this case. Pirarubicin (THP), a tetrahydropyranyl derivative of doxorubicin (DOX), demonstrated more rapid cellular internalization and potent cytotoxicity than DOX. Here, we conjugated the THP or DOX to N-(2-hydroxypropyl)methacrylamide copolymer via a hydrazone bond. The polymeric prodrug conjugates, P-THP and P-DOX, respectively, had comparable hydrodynamic sizes and drug loading. Compared with P-DOX, P-THP showed approximately 10 times greater cellular uptake during a 240 min incubation and a cytotoxicity that was more than 10 times higher during a 72-h incubation. A marginal difference was seen in P-THP and P-DOX accumulation in the liver and kidney at 6 h after drug administration, but no significant difference occurred in the tumor drug concentration during 6-24 h after drug administration. Antitumor activity against xenograft human pancreatic tumor (SUIT2) in mice was greater for P-THP than for P-DOX. To sum up, the present study compared the biological behavior of two different drugs, each attached to an N-(2-hydroxypropyl)methacrylamide copolymer carrier, with regard to their uptake by tumor cells, body distribution, accumulation in tumors, cytotoxicity, and antitumor activity in vitro and in vivo. No differences in the tumor cell uptake of the polymer-drug conjugates, P-THP and P-DOX, were observed. In contrast, the intracellular uptake of free THP liberated from the P-THP was 25-30 times higher than that of DOX liberated from P-DOX. This finding indicates that proper selection of the carrier, and especially conjugated active pharmaceutical ingredient (API) are most critical for anticancer activity of the polymer-drug conjugates. THP, in this respect, was found to be a more preferable API for polymer conjugation than DOX. Hence the treatment based on enhanced permeability and retention (EPR) effect that targets more selectively to solid tumors can be best achieved with THP, although both polymer conjugates of DOX and THP exhibited the EPR effects and drug release profiles in acidic pH similarly.


Assuntos
Acrilamidas/química , Antibióticos Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Doxorrubicina/análogos & derivados , Doxorrubicina/farmacologia , Portadores de Fármacos/química , Polímeros/química , Animais , Antibióticos Antineoplásicos/química , Antineoplásicos/química , Antineoplásicos/farmacologia , Doxorrubicina/química , Portadores de Fármacos/administração & dosagem , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Polímeros/administração & dosagem , Sarcoma Experimental/tratamento farmacológico , Sarcoma Experimental/metabolismo , Sarcoma Experimental/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...