Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Hepatology ; 68(4): 1391-1411, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29405333

RESUMO

Early detection and clear delineation of microscopic lesions during surgery are critical to the prognosis and survival of patients with hepatocellular carcinoma (HCC), a devastating malignancy without effective treatments except for resection. Tools to specifically identify and differentiate micronodules from normal tissue in HCC patients can have a positive impact on survival. Here, we discovered a peptide that preferentially binds to HCC cells through phage display. Significant accumulation of the fluorescence-labeled peptide in tumor from ectopic and orthotopic HCC mice was observed within 2 hours of systemic injection. Contrast between tumor and surrounding liver is up to 6.5-fold, and useful contrast lasts for 30 hours. Micronodules (0.03 cm in diameter) in liver and lung can clearly be distinguished from normal tissue with this fluorescence-labeled peptide in orthotopic HCC mice and HCC patients. Compared to indocyanine green, a Food and Drug Administration-approved imaging contrast agent, an up to 8.7-fold higher differentiation ratio of tumor to fibrosis is achieved with this fluorescence-labeled peptide. Importantly, this peptide enables up to 10-fold differentiation between HCC and peritumoral tissue in human tissues and the complete removal of tumor in HCC mice with surgical navigation. No abnormalities in behavior or activity are observed after systemic treatment, indicating the absence of overt toxicity. The peptide is metabolized with a half-life of approximately 4 hours in serum. CONCLUSION: Our findings demonstrate that micronodules can be specifically differentiated with high sensitivity from surrounding tissue with this molecule, opening clinical possibilities for early detection and precise surgery of HCC. (Hepatology 2018).


Assuntos
Carcinoma Hepatocelular/diagnóstico por imagem , Carcinoma Hepatocelular/cirurgia , Verde de Indocianina , Neoplasias Hepáticas/diagnóstico por imagem , Neoplasias Hepáticas/cirurgia , Animais , Biópsia por Agulha , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Feminino , Fluorescência , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Modelos Animais , Peptídeos , Valor Preditivo dos Testes , Sensibilidade e Especificidade
2.
Chem Res Toxicol ; 30(1): 73-80, 2017 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-28092939

RESUMO

Superparamagnetic iron oxide nanoparticles (SPIONs) have recently been used as an effective magnetic resonance imaging (MRI) contrast agent for the noninvasive diagnosis of chronic liver diseases including nonalcohol fatty liver diseases, nonalcohol steatohepatitis, and cirrhosis as well as liver tumors. However, the potential risk of the iron overload by SPIONs has been highly underestimated in chronic liver diseases. While most of SPIONs have been shown safe in the healthy group, significant toxicity potential by the iron overload has been revealed through immunotoxicity, lipid peroxidation, and fatty acid and cholesterol metabolism in cirrhosis as a high risk factor. As a result, the systems toxicology assessments of SPIONs are crucial in both healthy ones and chronic liver disease models to determine the margin of safety. In addition, the challenge of the iron overload by SPIONs requires better designed SPIONs as MRI contrast agents for chronic liver diseases such as the biodegradable nanocluster assembly with urine clearance.


Assuntos
Meios de Contraste/efeitos adversos , Compostos Férricos/efeitos adversos , Sobrecarga de Ferro/induzido quimicamente , Hepatopatias/diagnóstico por imagem , Nanopartículas/efeitos adversos , Animais , Meios de Contraste/uso terapêutico , Compostos Férricos/uso terapêutico , Humanos , Sobrecarga de Ferro/prevenção & controle , Imageamento por Ressonância Magnética , Nanopartículas/uso terapêutico
3.
BMC Cancer ; 15: 159, 2015 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-25881156

RESUMO

BACKGROUND: High levels of thymidine kinase 1 (TK1) and thymidine phosphorylase (TYMP) are key molecular targets by thymidine therapeutics in cancer treatment. The dual roles of TYMP as a tumor growth factor and a key activation enzyme of anticancer metabolites resulted in a mixed outcome in cancer patients. In this study, we investigated the roles of TK1 and TYMP on a thymidine quinoxaline conjugate to evaluate an alternative to circumvent the contradictive role of TYMP. METHODS: TK1 and TYMP levels in multiple liver cell lines were assessed along with the cytotoxicity of the thymidine conjugate. Cellular accumulation of the thymidine conjugate was determined with organelle-specific dyes. The impacts of TK1 and TYMP were evaluated with siRNA/shRNA suppression and pseudoviral overexpression. Immunohistochemical analysis was performed on both normal and tumor tissues. In vivo study was carried out with a subcutaneous liver tumor model. RESULTS: We found that the thymidine conjugate had varied activities in liver cancer cells with different levels of TK1 and TYMP. The conjugate mainly accumulated at endothelial reticulum and was consistent with cytosolic pathways. TK1 was responsible for the cytotoxicity yet high levels of TYMP counteracted such activities. Levels of TYMP and TK1 in the liver tumor tissues were significantly higher than those of normal liver tissues. Induced TK1 overexpression decreased the selectivity of dT-QX due to the concurring cytotoxicity in normal cells. In contrast, shRNA suppression of TYMP significantly enhanced the selective of the conjugate in vitro and reduced the tumor growth in vivo. CONCLUSIONS: TK1 was responsible for anticancer activity of dT-QX while levels of TYMP counteracted such an activity. The counteraction by TYMP could be overcome with RNA silencing to significantly enhance the dT-QX selectivity in cancer cells.


Assuntos
Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Redes e Vias Metabólicas , Quinoxalinas/metabolismo , Quinoxalinas/farmacologia , Timidina/metabolismo , Animais , Antineoplásicos/toxicidade , Linhagem Celular Tumoral , Citosol/metabolismo , Modelos Animais de Doenças , Expressão Gênica , Inativação Gênica , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Neoplasias/metabolismo , Quinoxalinas/toxicidade , Timidina Quinase/genética , Timidina Quinase/metabolismo , Timidina Fosforilase/genética , Timidina Fosforilase/metabolismo , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
4.
J Biol Chem ; 288(6): 4076-84, 2013 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-23275339

RESUMO

Peroxisome proliferator-activated receptor γ (PPARγ), a subgroup of ligand-activated nuclear receptors, plays critical roles in cell cycle regulation, differentiation, apoptosis, and invasion. PPARγ is involved in tumorigenesis and is a potent target for cancer therapy. PPARγ transactivation of KLF4 has been demonstrated in various studies; however, how PPARγ regulates KLF4 expression is not clear. In this study, we reveal that PPARγ regulates the expression of KLF4 by binding directly to the PPAR response element (PPRE) within the KLF4 promoter. The PPRE resides at -1657 to -1669 bp upstream of the KLF4 ATG codon, which is essential for the transactivation of troglitazone-induced KLF4 expression. Furthermore, we found that stable silencing of KLF4 obviously suppressed the G(1)/S arrest and anti-proliferation effects induced by PPARγ ligands. Taken together, our data indicate that up-regulation of KLF4 upon PPARγ activation is mediated through the PPRE in the KLF4 promoter, thus providing further insights into the PPARγ signal transduction pathway as well as a novel cancer therapeutic strategy.


Assuntos
Pontos de Checagem da Fase G1 do Ciclo Celular , Regulação Neoplásica da Expressão Gênica , Fatores de Transcrição Kruppel-Like/biossíntese , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , PPAR gama/agonistas , PPAR gama/metabolismo , Elementos de Resposta , Pontos de Checagem da Fase S do Ciclo Celular , Animais , Linhagem Celular Tumoral , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Camundongos Nus , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patologia , Neoplasias/terapia , PPAR gama/genética , Transdução de Sinais , Ativação Transcricional/genética
5.
Bioorg Med Chem Lett ; 24(24): 5716-5720, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25453807

RESUMO

Allosteric inhibition of coagulation enzymes offers the advantage of controlled inhibition. In this study, a small library of mono sulfated indole and benzothiazole based molecules was synthesized and screened against the panel of coagulation proteases. The results reveal that selected molecules inhibit the thrombin, factor Xa and factor XIa with moderate potency. Compound 6a was found to have an allosteric mode of inhibition against thrombin. Plasma clotting assays suggest that selected inhibitors 14b, 14c and 14d prolong both prothrombin and activated partial thromboplastin time. Overall, this work presents the newer class of allosteric inhibitors of thrombin and factor XIa with improved aqueous solubility profile.


Assuntos
Anticoagulantes/síntese química , Benzotiazóis/química , Coagulação Sanguínea/efeitos dos fármacos , Fator XIa/antagonistas & inibidores , Fator Xa/química , Trombina/antagonistas & inibidores , Regulação Alostérica , Anticoagulantes/farmacologia , Desenho de Fármacos , Humanos , Interações Hidrofóbicas e Hidrofílicas , Concentração Inibidora 50 , Cinética , Modelos Moleculares , Estrutura Molecular , Tempo de Tromboplastina Parcial , Tempo de Protrombina , Relação Estrutura-Atividade
6.
Bioorg Med Chem Lett ; 23(1): 355-9, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23164711

RESUMO

The design of sulfated, small, nonsaccharide molecules as modulators of proteins is still in its infancy as standard drug discovery tools such as library of diverse sulfated molecules and in silico docking and scoring protocol have not been firmly established. Databases, such as ZINC, contain too few sulfate-containing nonsaccharide molecules, which severely limits the identification of new hits. Lack of a generally applicable protocol for scaffold hopping limits the development of sulfated small molecules as synthetic mimetics of the highly sulfated glycosaminoglycans. We explored a sequential ligand-based (LBVS) and structure-based virtual screening (SBVS) approach starting from our initial discovery of monosulfated benzofurans to discover alternative scaffolds as allosteric modulators of thrombin, a key coagulation enzyme. Screening the ZINC database containing nearly 1 million nonsulfated small molecules using a pharmacophore developed from the parent sulfated benzofurans followed by a genetic algorithm-based dual-filter docking and scoring screening identified a group of 10 promising hits, of which three top-scoring hits were synthesized. Each was found to selectively inhibit human alpha-thrombin suggesting the possibility of this approach for scaffold hopping. Michaelis-Menten kinetics showed allosteric inhibition mechanism for the best molecule and human plasma studies confirmed good anticoagulation potential as expected. Our simple sequential LBVS and SBVS approach is likely to be useful as a general strategy for identification of sulfated small molecules hits as modulators of glycosaminoglycan-protein interactions.


Assuntos
Materiais Biomiméticos/química , Glicosaminoglicanos/química , Bibliotecas de Moléculas Pequenas/química , Sulfatos/química , Algoritmos , Regulação Alostérica/efeitos dos fármacos , Materiais Biomiméticos/síntese química , Materiais Biomiméticos/farmacologia , Avaliação Pré-Clínica de Medicamentos , Humanos , Cinética , Bibliotecas de Moléculas Pequenas/síntese química , Bibliotecas de Moléculas Pequenas/farmacologia , Trombina/antagonistas & inibidores , Trombina/metabolismo
7.
J Adv Res ; 35: 259-266, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35003803

RESUMO

Introduction: Cancer cells induced into immunogenic cell death (ICD) in vitro can be directly used as a whole cell vaccine for tumor immunotherapy with many advantages, especially enacting immediate and intense 'eat me' signals to engage immune system. Unfortunately, there have been few successes with in vitro ICD cancer cells as a treatment vaccine. Objective: To demonstrate that cancer cells treated in vitro with a new class of potent ICD inducer, naphthylquinoxaline thymidine conjugate (NAP) followed by UVA irradiation would be able to act as an effective tumor immunotherapy directly. Methods: The therapeutic potentials of treated cancer cell plus different vaccine adjuvants were assessed by in vivo liver tumor model and in vitro mixed lymphocyte reaction studies. The elicited activated T cells were determined with immunohistochemistry and T cell induced cytotoxicity studies. Results: Treatment of established H22 tumor with in vitro NAP and UVA treated cancer cell vaccine led to significantly improved survival. Further mixed lymphocyte reaction study implied that adjuvants alum and CpG would improve the therapeutic potential whereas poly IC would not be as effective. Subsequent in vivo validation of alum and CpG adjuvants indicated that only CpG in NAP and UVA treated cell vaccine resulted in markedly enhanced survival (median at 71 days and 50% tumor-free) as compared with PBS group (14.5 days, 0%) and CpG alone (36 days, 0%). It was revealed that the enhanced efficacy by CpG was specific to NAP and UVA treated cells. Moreover, the effective tumor immunotherapy was achieved through the infiltration of active CD4 and CD8 T cells in tumors and acquisition of cancer cell-specific cytotoxic CD8 T cells. Conclusion: In vitro NAP and UVA treated cancer cells plus CpG adjuvant are effective tumor therapeutic vaccines per se.


Assuntos
Adjuvantes de Vacinas , Vacinas Anticâncer , Neoplasias , Oligodesoxirribonucleotídeos , Animais , Humanos , Neoplasias/terapia , Timidina
8.
J Hematol Oncol ; 15(1): 46, 2022 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-35488312

RESUMO

BACKGROUND: Personalized immunotherapy utilizing cancer vaccines tailored to the tumors of individual patients holds promise for tumors with high genetic heterogeneity, potentially enabling eradication of the tumor in its entirety. METHODS: Here, we demonstrate a general strategy for biological nanovaccines that trigger tailored tumor-specific immune responses for hepatocellular carcinoma (HCC). Dendritic cell (DC)-derived exosomes (DEX) are painted with a HCC-targeting peptide (P47-P), an α-fetoprotein epitope (AFP212-A2) and a functional domain of high mobility group nucleosome-binding protein 1 (N1ND-N), an immunoadjuvant for DC recruitment and activation, via an exosomal anchor peptide to form a "trigger" DEX vaccine (DEXP&A2&N). RESULTS: DEXP&A2&N specifically promoted recruitment, accumulation and activation of DCs in mice with orthotopic HCC tumor, resulting in enhanced cross-presentation of tumor neoantigens and de novo T cell response. DEXP&A2&N elicited significant tumor retardation and tumor-specific immune responses in HCC mice with large tumor burdens. Importantly, tumor eradication was achieved in orthotopic HCC mice when antigenic AFP peptide was replaced with the full-length AFP (A) to form DEXP&A&N. Supplementation of Fms-related tyrosine kinase 3 ligand greatly augmented the antitumor immunity of DEXP&A&N by increasing immunological memory against tumor re-challenge in orthotopic HCC mice. Depletion of T cells, cross-presenting DCs and other innate immune cells abrogated the functionality of DEXP&A&N. CONCLUSIONS: These findings demonstrate the capacity of universal DEX vaccines to induce tumor-specific immune responses by triggering an immune response tailored to the tumors of each individual, thus presenting a generalizable approach for personalized immunotherapy of HCC, by extension of other tumors, without the need to identify tumor antigens.


Assuntos
Vacinas Anticâncer , Carcinoma Hepatocelular , Exossomos , Neoplasias Hepáticas , Animais , Vacinas Anticâncer/uso terapêutico , Humanos , Imunidade Inata , Imunoterapia/métodos , Camundongos , Peptídeos , alfa-Fetoproteínas
9.
Chem Res Toxicol ; 24(3): 402-11, 2011 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-21306116

RESUMO

Quinone methides (QMs) are involved in the metabolism of many drugs and carcinogens as reactive intermediates to form covalent nucleobase adducts in DNA that associate with high mutagenicity. Recently, a plethora of synthetic QM DNA alkylating agents have been developed to form various nucleobase adducts as potential antitumor agents. However, the mutagenic potential of these synthetic QM alkylating agents has not been fully investigated. In this report, N-methylquinolinium QM was developed as a synthetic model to study biological consequences of the formation of nucleobase adducts in a DNA target. N-Methylquinolinium QM was generated in situ via an elimination process from a bis-quaternary ammonium precursor that was synthesized from a quinoline derivative. Alkylation with N-methylquinolinium QM on a DNA target produced mostly a stable N(2)-dG adduct as revealed by gel electrophoresis and DNA digestion assays and confirmed by mass and NMR analyses. The formation of N(2)-dG adducts of a DNA target was found to cause extensive stops in the primer extension with high fidelity DNA polymerase T7 and even low fidelity error prone Dpo4. The direct biological impact of a prealkylated green fluorescence protein plasmid with N-methylquinolinium QM was demonstrated as significant suppression of protein expression in A549 cells. Overall, our results suggested that nucleobase-QM adducts could potentially block nucleobase mismatch/translesion in the error-prone process to reduce the mutagenic potential if designed carefully.


Assuntos
Adutos de DNA/química , DNA Polimerase Dirigida por DNA/metabolismo , DNA/química , Desoxiguanosina/química , Proteínas de Fluorescência Verde/metabolismo , Compostos de Quinolínio/química , Alquilação , Linhagem Celular Tumoral , DNA/metabolismo , DNA Polimerase beta/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas de Fluorescência Verde/genética , Humanos , Espectroscopia de Ressonância Magnética , Plasmídeos/genética , Plasmídeos/metabolismo , Reação em Cadeia da Polimerase , Compostos de Quinolínio/toxicidade
10.
Nat Commun ; 11(1): 1790, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32286296

RESUMO

Treating large established tumors is challenging for dendritic cell (DC)-based immunotherapy. DC activation with tumor cell-derived exosomes (TEXs) carrying multiple tumor-associated antigen can enhance tumor recognition. Adding a potent adjuvant, high mobility group nucleosome-binding protein 1 (HMGN1), boosts DCs' ability to activate T cells and improves vaccine efficiency. Here, we demonstrate that TEXs painted with the functional domain of HMGN1 (TEX-N1ND) via an exosomal anchor peptide potentiates DC immunogenicity. TEX-N1ND pulsed DCs (DCTEX-N1ND) elicit long-lasting antitumor immunity and tumor suppression in different syngeneic mouse models with large tumor burdens, most notably large, poorly immunogenic orthotopic hepatocellular carcinoma (HCC). DCTEX-N1ND show increased homing to lymphoid tissues and contribute to augmented memory T cells. Importantly, N1ND-painted serum exosomes from cancer patients also promote DC activation. Our study demonstrates the potency of TEX-N1ND to strengthen DC immunogenicity and to suppress large established tumors, and thus provides an avenue to improve DC-based immunotherapy.


Assuntos
Alarminas/metabolismo , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/metabolismo , Exossomos/metabolismo , Proteína HMGN1/metabolismo , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/metabolismo , Animais , Carcinoma Hepatocelular/terapia , Linhagem Celular , Proteína HMGN1/genética , Células HeLa , Humanos , Imuno-Histoquímica , Imunoterapia , Neoplasias Hepáticas/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Linfócitos T/metabolismo
11.
Malar J ; 8: 139, 2009 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-19555505

RESUMO

BACKGROUND: Malaria is the third most prevalent cause of infectious disease in the world. Resistance of the parasite to classical drugs makes the discovery of new and effective drugs more urgent. The oxidized derivative of hydroxy-cis terpenone (OHCT) is a synthetic molecule that is not toxic to cultured human liver cells at concentrations as high as 60 microM and inhibits activity of cytochrome P450s that metabolize many drugs. METHODS: OHCT activity against chloroquine-sensitive and -resistant strains of Plasmodium falciparum, and a P. falciparum clone that is partially resistant to artemisinin was assayed in vitro. RESULTS: OHCT at nanomolar concentrations was effective against all intraerythrocytic stages of P. falciparum and exhibited activity in vitro against both chloroquine-sensitive and -resistant strains of P. falciparum as well as a P. falciparum clone that is partially resistant to artemisinin. Moreover, OHCT exhibited potent activity against gametocytes, the form that is transmitted by mosquitoes and essential for the spread of malaria. CONCLUSION: OHCT displays strong growth inhibitory activity against all stages of P. falciparum and no evidence of toxicity to human cells in culture. It is easily synthesized and has the potential for inhibiting metabolism of drugs used in combination therapies.


Assuntos
Antimaláricos/farmacologia , Resistência a Medicamentos , Malária Falciparum/tratamento farmacológico , Plasmodium falciparum/efeitos dos fármacos , Terpenos/farmacologia , Animais , Antimaláricos/uso terapêutico , Células Cultivadas/efeitos dos fármacos , Eritrócitos/efeitos dos fármacos , Eritrócitos/parasitologia , Humanos , Concentração Inibidora 50 , Testes de Sensibilidade Parasitária , Terpenos/síntese química
12.
Bioorg Med Chem Lett ; 19(15): 4126-9, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19540113

RESUMO

Designing non-saccharide functional mimics of heparin is a major challenge. In this work, a library of small, aromatic molecules based on the sulfated DHP scaffold was synthesized and screened against thrombin and factor Xa. The results reveal that (i) selected monomeric benzofuran derivatives inhibit the two enzymes, albeit weakly; (ii) the two enzymes recognize different structural features in the benzofurans studied suggesting significant selectivity of recognition; and (iii) the mechanism of inhibition is allosteric. The molecules represent the first allosteric small molecule inhibitors of the two enzymes.


Assuntos
Fator Xa/química , Trombina/antagonistas & inibidores , Sítio Alostérico , Anticoagulantes/síntese química , Anticoagulantes/farmacologia , Benzofuranos/química , Sítios de Ligação , Química Farmacêutica/métodos , Desenho de Fármacos , Heparina/química , Humanos , Cinética , Modelos Químicos , Polímeros/química , Polímeros/farmacologia
13.
Eur J Med Chem ; 171: 255-264, 2019 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-30925340

RESUMO

Anticancer anthracyclines are cytotoxic drugs that can induce antitumor immune response as a secondary effect through immunogenic cell death (ICD) mechanism. However, the immunogenic potency is quite limited, possibly due to that these chemotherapeutic agents are not specifically developed as ICD inducers. Thus, new drug entities through studies focusing on enhanced ICD induction would significantly promote antitumor immune response in the vaccination application. We report here a naphthyl quinoxaline thymidine conjugate as a new class of cytotoxic compounds that effectively induced in vivo antitumor activity through the vaccination application. Synthesized naphthyl quinoxaline conjugates were weak fluorescent thymidine analog yet exhibited a pronounced anticancer activity in the low nanomolar range post UVA activation. The potent activity of naphthyl conjugate was able to induce the marked detection of ICD markers including ATP and HMGB1 extracellular and calreticulin intracellularly at 2 h post UVA activation. Most importantly, mice vaccinated with cells treated with naphthyl conjugate plus UVA exhibited complete tumor growth inhibition in the tumor challenge study, and the induced immunogenic inhibition was much more effective than that of mitoxantrone anthracycline drug. All these results demonstrate the high potential of naphthyl quinoxaline conjugate for the cancer cell vaccine against tumor.


Assuntos
Antineoplásicos/farmacologia , Quinoxalinas/farmacologia , Timidina/farmacologia , Raios Ultravioleta , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Molecular , Quinoxalinas/química , Relação Estrutura-Atividade , Timidina/química , Vacinação
14.
Eur J Med Chem ; 167: 499-509, 2019 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-30784882

RESUMO

Conventional chemotherapeutic and photodynamic therapy have recently been shown to also elicit immune response against cancer through the immunogenic cell death mechanism, which can be potentially translated into effective cancer vaccines. However, there are few studies on the potential of nanodelivery system to promote the immunogenic cell death as a cancer vaccine. We reported here that cRGD target liposome delivery system was capable to promote the immunogenic cell death through enhanced potency of a thymidine conjugate post UVA activation as a cancer vaccine. Liposomes and cRGD target liposomes were found to significantly increase the cellular accumulation of the thymidine conjugate and subsequently translated into enhanced cytotoxic potency after UVA activation. More importantly, cRGD target liposomes of the thymidine conjugate markedly promoted the early detection of immunogenic cell death markers including ATP, HMGB1 and calreticulin. Subsequent in vivo vaccination-challenge study confirmed effective tumor growth inhibition by the cRGD liposomal thymidine conjugate and UVA treated cancer cells as the cancer vaccine. In addition, liposomes and cRGD target liposomes alone did not shown any induction of the immunogenic cell death markers, revealing the adjuvant nature of the nanodelivery system.


Assuntos
Vacinas Anticâncer/química , Morte Celular/imunologia , Sistemas de Liberação de Medicamentos/métodos , Oligopeptídeos/antagonistas & inibidores , Timidina/uso terapêutico , Raios Ultravioleta , Adjuvantes Imunológicos , Animais , Antineoplásicos/administração & dosagem , Vacinas Anticâncer/administração & dosagem , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Humanos , Lipossomos/uso terapêutico
15.
RSC Adv ; 9(25): 14051-14059, 2019 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-35519327

RESUMO

Superparamagnetic iron oxide nanoclusters (SPIONs) modified with pH (low) insertion peptide (pHLIP) could be advantageous for magnetic resonance imaging (MRI) diagnosis of liver tumors at the early stage due to their unique responsiveness to the tumor acidic microenvironment when tumor markers are unknown. However, many critical aspects including the effectiveness of selective MRI in liver tumors, types of delivery and the potential safety profile in cirrhosis need to be fully evaluated. In this study, we report the evaluation of non-targeting, C- or N-pHLIP modified SPIONs as the contrast agent for selective MRI of liver tumors and their potential toxicity profile in cirrhosis. It was found that N-pHLIP modified SPIONs did not result in the loss of liver tumor in the T2-weight MRI but provided additional dynamic details of tumor structures that would enhance the diagnosis of liver tumors at a small size below 8 mm. In addition, an enhanced safety profile was found for N-pHLIP modified SPIONs with almost fully recoverable impact in cirrhosis. In contrast, the poly-d-lysine assembled SPIONs and C-terminus linked pHLIP SPIONs had non-tumor specific MRI contrast enhancement and potential safety risks in cirrhosis due to the iron overload post injection. All these results implied the promising potential of N-terminus linked pHLIP SPIONs as an MRI contrast agent for the diagnosis of liver tumors.

16.
Sci Transl Med ; 10(444)2018 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-29875202

RESUMO

Exosomes are circulating nanovesicular carriers of macromolecules, increasingly used for diagnostics and therapeutics. The ability to load and target patient-derived exosomes without altering exosomal surfaces is key to unlocking their therapeutic potential. We demonstrate that a peptide (CP05) identified by phage display enables targeting, cargo loading, and capture of exosomes from diverse origins, including patient-derived exosomes, through binding to CD63-an exosomal surface protein. Systemic administration of exosomes loaded with CP05-modified, dystrophin splice-correcting phosphorodiamidate morpholino oligomer (EXOPMO) increased dystrophin protein 18-fold in quadriceps of dystrophin-deficient mdx mice compared to CP05-PMO. Loading CP05-muscle-targeting peptide on EXOPMO further increased dystrophin expression in muscle with functional improvement without any detectable toxicity. Our study demonstrates that an exosomal anchor peptide enables direct, effective functionalization and capture of exosomes, thus providing a tool for exosome engineering, probing gene function in vivo, and targeted therapeutic drug delivery.


Assuntos
Exossomos/metabolismo , Peptídeos/metabolismo , Animais , Linhagem Celular , Exossomos/efeitos dos fármacos , Exossomos/ultraestrutura , Inflamação/patologia , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Morfolinos/farmacologia , Músculos/efeitos dos fármacos , Músculos/metabolismo , Soro/metabolismo , Tetraspanina 30/metabolismo
17.
Acta Biomater ; 55: 194-203, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28363789

RESUMO

Superparamagnetic iron oxide nanoparticles (SPION) are contrast agents used for noninvasive tumor magnetic resonance imaging (MRI). SPION with active targeting by tumor-specific ligands can effectively enhance the MRI sensitivity and specificity of tumors. However, the challenge remains when the tumor specific markers are yet to be determined, especially in the case of early tumor detection. In this study, the effectiveness of pH-responsive SPION via a pH low insertion peptide (pHLIP) to target tumor acidic microenvironments was investigated. Polylysine polymers were first successfully modified with pHLIP to have the pH-responsive capability. SPION pHLIP nanoclusters of 64, 82, 103, and 121nm size were then assembled by the pH-responsive polymers in a size-controlled manner. The pH-responsive SPION nanoclusters of the 64nm size exhibited the most effective pH-responsive retention in cells and tumor selective imaging in MRI. More importantly, the unique contrast enhancement of tumor inner core by the pH-responsive SPION in three different tumor models demonstrated the clinical potential to target tumor acidic microenvironment through pHLIP for tumor early detection and diagnosis by MRI. STATEMENT OF SIGNIFICANCE: Detection and diagnosis of tumors at early stage are critical for the improvement of the survival rate of cancer patients. However, the challenge remains when the tumor specific markers are yet to be determined, especially in early tumor detection. pH low insertion peptide (pHLIP) has been used as a specific ligand to target the tumor acidic microenvironment for tumors at early and metastatic stages. Superparamagnetic iron nanoparticles (SPION) are contrast enhancing agents used in the noninvasive magnetic resonance imaging for tumors. This research has demonstrated that pH-responsive pHLIP nanoclusters of SPION were able to target different tumors and facilitate the noninvasive diagnosis of tumors by MRI.


Assuntos
Meios de Contraste , Sistemas de Liberação de Medicamentos , Imageamento por Ressonância Magnética , Nanopartículas de Magnetita , Proteínas de Membrana , Neoplasias Experimentais/diagnóstico por imagem , Animais , Linhagem Celular Tumoral , Meios de Contraste/química , Meios de Contraste/farmacologia , Humanos , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/uso terapêutico , Masculino , Proteínas de Membrana/química , Proteínas de Membrana/farmacologia , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia
18.
Emerg Microbes Infect ; 6(12): e108, 2017 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-29209052

RESUMO

The M gene segment of influenza A virus has been shown to be a contributing factor to the high growth phenotype. However, it remains largely unknown why matrix protein 1 (M1), the major structural protein encoded by M gene, exhibits pH-dependent conformational changes during virus replication. Understanding the mechanisms underlying efficient virus replication can help to develop strategies not only to combat influenza infections but also to improve vaccine supplies. M(NLS-88R) and M(NLS-88E) are two M1 mutants differing by only a single amino acid: G88R vs G88E. G88R but not G88E was the compensatory mutation naturally selected by the virus after its nuclear localization signal was disrupted. Our study shows that, compared with M(NLS-88E) M1, M(NLS-88R) M1 dissociated quickly from viral ribonucleoproteins (vRNPs) at higher pH and took less time to dissemble in vitro, despite forming thicker matrix layer and having stronger association with vRNP in assembled virions. Correspondingly, M(NLS-88R) replicated more efficiently and was genetically more stable than M(NLS-88E). Crystallographic analysis indicated that M(NLS-88R) M1, like wild-type M1, is able to switch from a face-to-back-oriented conformation to a face-to-face-oriented conformation when pH drops from neutral to acidic, whereas G88E mutation causes M(NLS-88E) M1 to be trapped in a face-to-face-arranged conformation regardless of environmental pH. Our results suggest that maintaining M1 pH-dependent conformational flexibility is critical for efficient virus replication, and position 88 is a key residue controlling M1 pH-dependent conformational changes. Our findings provide insights into developing M1-based antiviral agents.


Assuntos
Vírus da Influenza A/fisiologia , Influenza Humana/virologia , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/metabolismo , Replicação Viral , Animais , Cães , Humanos , Concentração de Íons de Hidrogênio , Vírus da Influenza A/química , Vírus da Influenza A/genética , Células Madin Darby de Rim Canino , Modelos Moleculares , Conformação Proteica , Proteínas da Matriz Viral/genética
19.
Eur J Med Chem ; 107: 180-91, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26584085

RESUMO

Thymidine quinoxaline conjugate (dT-QX) is a thymidine analog with selective cytotoxicity against different cancer cells. In this study, the structure activity relationship study of dT-QX analogs was carried out under the low radiance of black fluorescent (UVA-1) light. Significantly enhanced cytotoxicity was observed under UVA-1 activation among analogs containing both thymidine and quinoxaline moieties with different length of the linker, stereochemical configuration and halogenated substituents. Among these analogs, the thymidine dichloroquinoxaline conjugate exhibited potent activity under UVA-1 activation as the best candidate with EC50 at 0.67 µM and 1.3 µM against liver and pancreatic cancer cells, respectively. In contrast, the replacement of thymidine moiety with a galactosyl residue or the replacement of quinoxaline moiety with a fluorescent pyrenyl residue or a simplified diketone structure resulted in the full loss of activity. Furthermore, it was revealed that the low radiance of UVA-1 at 3 mW/cm(2) for 20 min was sufficient enough to induce the full cytotoxicity of thymidine dichloroquinoxaline conjugate and that the cytotoxic mechanism was achieved through a rapid and steady production of reactive oxygen species.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Fotoquimioterapia/métodos , Relação Estrutura-Atividade , Antineoplásicos/síntese química , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos da radiação , Técnicas de Química Sintética , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Humanos , Quinoxalinas/química , Espécies Reativas de Oxigênio/metabolismo , Timidina/análogos & derivados , Timidina/química , Raios Ultravioleta
20.
Sci Rep ; 6: 29110, 2016 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-27357559

RESUMO

Superparamagnetic iron oxide nanoparticles (SPIONs) as a contrast agent have been widely used in magnetic resonance imaging for tumor diagnosis and theranostics. However, there has been safety concern of SPIONs with cirrhosis related to excess iron-induced oxidative stress. In this study, the impact of iron overload by SPIONs was assessed on a mouse cirrhosis model. A single dose of SPION injection at 0.5 or 5 mg Fe/kg in the cirrhosis group induced a septic shock response at 24 h with elevated serum levels of liver and kidney function markers and extended impacts over 14 days including high levels of serum cholesterols and persistent low serum iron level. In contrast, full restoration of liver functions was found in the normal group with the same dosages over time. Analysis with PCR array of the toxicity pathways revealed the high dose of SPIONs induced significant expression changes of a distinct subset of genes in the cirrhosis liver. All these results suggested that excess iron of the high dose of SPIONs might be a risk factor for cirrhosis because of the marked impacts of elevated lipid metabolism, disruption of iron homeostasis and possibly, aggravated loss of liver functions.


Assuntos
Cirrose Hepática/fisiopatologia , Fígado/efeitos dos fármacos , Nanopartículas de Magnetita/efeitos adversos , Estresse Oxidativo/efeitos dos fármacos , Animais , Meios de Contraste/efeitos adversos , Meios de Contraste/uso terapêutico , Modelos Animais de Doenças , Humanos , Sobrecarga de Ferro/induzido quimicamente , Sobrecarga de Ferro/fisiopatologia , Fígado/fisiopatologia , Cirrose Hepática/induzido quimicamente , Imageamento por Ressonância Magnética , Nanopartículas de Magnetita/uso terapêutico , Camundongos , Nanomedicina Teranóstica , Distribuição Tecidual/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa