Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cancer Res ; 82(7): 1409-1422, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35039320

RESUMO

The standard treatment of nonmuscle invasive bladder cancer (NMIBC) is transurethral resection of the tumors, followed by intravesical therapy (IT), which comprises a direct instillation of a solution of Bacillus Calmette-Guérin vaccine or chemotherapy into the bladder. However, the recurrence rate in this disease remains unacceptably high. IT is a local treatment that fails to reach tumors developed in the upper urinary tract (ureter and renal pelvis). The catheterization procedure required for IT is invasive, painful, and poses an increased infection risk, resulting in poor patient quality of life and compliance. There is an unmet need for a potent, comprehensive, and noninvasive option. Without chemical modifications, peptides are rapidly removed by renal clearance. This "shortcoming" can be advantageous when used as a drug carrier for directing therapy to NMIBC. Here we develop a urinary drug-disposing (UDD) approach to improve NMIBC treatment. A 12-amino acid bio-inert peptide (Bdd) that can be exclusively eliminated via renal filtration was generated for delivering the microtubule inhibitor DM1 to NMIBC with minimal nonspecific accumulation in other organs. The UDD approach prolonged survival of mice bearing human bladder tumors. Unlike IT, the treatment was given noninvasively (intravenously). Furthermore, it was more effective at suppressing tumor growth than clinically used IT (mitomycin) and safer than free DM1. The application of this UDD approach to treat kidney tumors and deliver other drugs such as doxorubicin was also demonstrated. Overall, the rapid renal clearance of peptides can be exploited to direct cancer therapies to the urinary system. SIGNIFICANCE: A noninvasive drug delivery approach that targets the urinary system overcomes the current barriers facing effective treatment of bladder cancer.


Assuntos
Neoplasias da Bexiga Urinária , Adjuvantes Imunológicos , Animais , Vacina BCG/uso terapêutico , Humanos , Camundongos , Invasividade Neoplásica/patologia , Recidiva Local de Neoplasia/patologia , Preparações Farmacêuticas , Qualidade de Vida , Bexiga Urinária/patologia , Neoplasias da Bexiga Urinária/patologia
2.
BMC Cancer ; 20(1): 661, 2020 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-32678032

RESUMO

BACKGROUND: Tumors are heterogeneous in nature, composed of different cell populations with various mutations and/or phenotypes. Using a single drug to encounter cancer progression is generally ineffective. To improve the treatment outcome, multiple drugs of distinctive mechanisms but complementary anticancer activities (combination therapy) are often used to enhance antitumor efficacy and minimize the risk of acquiring drug resistance. We report here the synergistic effects of salinomycin (a polyether antibiotic) and dasatinib (a Src kinase inhibitor). METHODS: Functionally, both drugs induce cell cycle arrest, intracellular reactive oxygen species (iROS) production, and apoptosis. We rationalized that an overlapping of the drug activities should offer an enhanced anticancer effect, either through vertical inhibition of the Src-STAT3 axis or horizontal suppression of multiple pathways. We determined the toxicity induced by the drug combination and studied the kinetics of iROS production by fluorescence imaging and flow cytometry. Using genomic and proteomic techniques, including RNA-sequencing (RNA-seq), reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and Western Blot, we subsequently identified the responsible pathways that contributed to the synergistic effects of the drug combination. RESULTS: Compared to either drug alone, the drug combination showed enhanced potency against MDA-MB-468, MDA-MB-231, and MCF-7 human breast cancer (BC) cell lines and tumor spheroids. The drug combination induces both iROS generation and apoptosis in a time-dependent manner, following a 2-step kinetic profile. RNA-seq data revealed that the drug combination exhibited synergism through horizontal suppression of multiple pathways, possibly through a promotion of cell cycle arrest at the G1/S phase via the estrogen-mediated S-phase entry pathway, and partially via the BRCA1 and DNA damage response pathway. CONCLUSION: Transcriptomic analyses revealed for the first time, that the estrogen-mediated S-phase entry pathway partially contributed to the synergistic effect of the drug combination. More importantly, our studies led to the discoveries of new potential therapeutic targets, such as E2F2, as well as a novel drug-induced targeting of estrogen receptor ß (ESR2) approach for triple-negative breast cancer treatment, currently lacking of targeted therapies.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/tratamento farmacológico , Dasatinibe/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Piranos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Dasatinibe/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Fator de Transcrição E2F2/genética , Fator de Transcrição E2F2/metabolismo , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Células MCF-7 , Piranos/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Transcriptoma/efeitos dos fármacos
3.
Neuro Oncol ; 22(10): 1495-1504, 2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-32301996

RESUMO

BACKGROUND: Diffuse intrinsic pontine glioma (DIPG) is a highly lethal malignancy that occurs predominantly in children. DIPG is inoperable and post-diagnosis survival is less than 1 year, as conventional chemotherapy is ineffective. The intact blood-brain barrier (BBB) blocks drugs from entering the brain. Convection-enhanced delivery (CED) is a direct infusion technique delivering drugs to the brain, but it suffers from rapid drug clearance. Our goal is to overcome the delivery barrier via CED and maintain a therapeutic concentration at the glioma site with a payload-adjustable peptide nanofiber precursor (NFP) that displays a prolonged retention property as a drug carrier. METHODS: The post-CED retention of 89Zr-NFP was determined in real time using PET/CT imaging. Emtansine (DM1), a microtubule inhibitor, was conjugated to NFP. The cytotoxicity of the resulting DM1-NFP was tested against patient-derived DIPG cell lines. The therapeutic efficacy was evaluated in animals bearing orthotopic DIPG, according to glioma growth (measured using bioluminescence imaging) and the long-term survival. RESULTS: DM1-NFP demonstrated potency against multiple glioma cell lines. The half-maximal inhibitory concentration values were in the nanomolar range. NFP remained at the infusion site (pons) for weeks, with a clearance half-life of 60 days. DM1-NFP inhibited glioma progression in animals, and offered a survival benefit (median survival of 62 days) compared with the untreated controls (28 days) and DM1-treated animal group (26 days). CONCLUSIONS: CED, in combination with DM1-NFP, complementarily functions to bypass the BBB, prolong drug retention at the fusion site, and maintain an effective therapeutic effect against DIPG to improve treatment outcome.


Assuntos
Neoplasias do Tronco Encefálico , Glioma Pontino Intrínseco Difuso , Nanofibras , Animais , Neoplasias do Tronco Encefálico/tratamento farmacológico , Criança , Convecção , Humanos , Peptídeos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Radioisótopos , Zircônio
4.
Methods Mol Biol ; 2115: 79-90, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32006395

RESUMO

The silencing of an oncogene with a small interfering RNA (siRNA) is a promising way for cancer therapy. Its efficacy can be further enhanced by integrating with other therapeutics; however, transporting siRNA and other active ingredients to the same location at the same time is challenging. Here, we report a novel multifunctional nanodelivery platform by sequentially layering several functional ingredients, such as siRNAs, microRNAs, peptides, and targeting ligands, onto a core through charge-charge interaction. The prepared nanovectors effectively and programmably delivered multiple active components to maximize therapeutic combination with minimal off-targeting effects.


Assuntos
Portadores de Fármacos/química , Nanopartículas/química , Neoplasias/terapia , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/uso terapêutico , Animais , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Feminino , Ouro/química , Humanos , Ácido Hialurônico/química , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/genética , Peptídeos/química , RNA Interferente Pequeno/farmacocinética , Terapêutica com RNAi/métodos , Eletricidade Estática
5.
Res Vet Sci ; 128: 86-89, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31760317

RESUMO

Chemotherapeutic drugs are given parenterally to treat various canine tumors. A limitation of parenteral administration is low drug penetration into the tumor, which reduces tumoricidal activity. Various drug carriers have been used to enhance tumor delivery, including albumin, liposomes and nanoparticles. A novel peptide-based nanofiber precursor (NFP) has been developed that is designed to take advantage of the leaky tumor neovasculature to promote drug delivery after parenteral administration. In this study, we loaded aldoxorubicin, an albumin-bound prodrug version of doxorubicin, onto NFP and tested the in vitro cytotoxicity in canine mammary carcinoma (CMT12, CMT25) and osteosarcoma (HMPOS, D-17, Abrams) cell lines. The half maximal inhibitory concentration (IC50) was determined with a luminescence-based cell viability assay. The IC50 for aldoxorubicin-loaded NFP was lower than free aldoxorubicin or doxorubicin in all cell lines, whereas non-drug loaded NFP had no cytotoxic effects. There were differences in IC50 between the osteosarcoma lines, with lower and higher IC50 for HMPOS and D-17 cells, respectively, with all drugs (aldoxorubicin-loaded NFP, free aldoxorubicin or free doxorubicin). Our results indicate that drug-loaded NFPs are cytotoxic for various canine mammary carcinoma and osteosarcoma cell lines in vitro and hold promise as a mechanism for enhancing delivery of chemotherapeutic agents to canine tumors.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Doenças do Cão/tratamento farmacológico , Doxorrubicina/análogos & derivados , Hidrazonas/farmacologia , Neoplasias Mamárias Animais/tratamento farmacológico , Nanofibras , Osteossarcoma/veterinária , Animais , Linhagem Celular Tumoral , Cães , Doxorrubicina/farmacologia , Osteossarcoma/tratamento farmacológico
6.
ACS Chem Neurosci ; 10(5): 2287-2298, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-30838861

RESUMO

The blood-brain barrier (BBB) represents a major obstacle in delivering therapeutics to brain lesions. Convection-enhanced delivery (CED), a method that bypasses the BBB through direct, cannula-mediated drug delivery, is one solution to maintaining increased, effective drug concentration at these lesions. CED was recently proven safe in a phase I clinical trial against diffuse intrinsic pontine glioma (DIPG), a childhood cancer. Unfortunately, the exact relationship between drug size, charge, and pharmacokinetic behavior in the brain parenchyma are difficult to observe in vivo. PET imaging of CED-delivered agents allows us to determine these relationships. In this study, we label different modifications of the PDGFRA inhibitor dasatinib with fluorine-18 or via a nanofiber-zirconium-89 system so that the effect of drug structure on post-CED behavior can accurately be tracked in vivo, via PET. Relatively unchanged bioactivity is confirmed in patient- and animal-model-derived cell lines of DIPG. In naïve mice, significant individual variability in CED drug clearance is observed, highlighting a need to accurately understand drug behavior during clinical translation. Generally, the half-life for a drug to clear from a CED site is short for low molecular weight dasatinib analogs that bare different charge; 1-3 (1, 32.2 min (95% CI: 27.7-37.8), 2, 44.8 min (27.3-80.8), and 3, 71.7 min (48.6-127.6) minutes) and is much longer for a dasatinib-nanofiber conjugate, 5, (42.8-57.0 days). Positron emission tomography allows us to accurately measure the effect of drug size and charge in monitoring real-time drug behavior in the brain parenchyma of live specimens.


Assuntos
Antineoplásicos/farmacocinética , Barreira Hematoencefálica/efeitos dos fármacos , Corpo Estriado/efeitos dos fármacos , Dasatinibe/farmacocinética , Animais , Antineoplásicos/uso terapêutico , Barreira Hematoencefálica/metabolismo , Neoplasias do Tronco Encefálico/tratamento farmacológico , Neoplasias do Tronco Encefálico/metabolismo , Neoplasias do Tronco Encefálico/patologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Corpo Estriado/metabolismo , Dasatinibe/uso terapêutico , Glioma Pontino Intrínseco Difuso/tratamento farmacológico , Glioma Pontino Intrínseco Difuso/metabolismo , Glioma Pontino Intrínseco Difuso/patologia , Sistemas de Liberação de Medicamentos , Humanos , Camundongos , Estrutura Molecular , Distribuição Tecidual
7.
Adv Funct Mater ; 28(44)2018 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-30505260

RESUMO

An effective tumoral delivery system should show minimal removal by the reticuloendothelial system (RES), promote tumor uptake and penetration, and minimize on-site clearance. This study reports the design and synthesis of advanced self-assembling peptide nanofiber precursor (NFP) analogues. The peptidic nature of NFP offers the design flexibility for on-demand customization with imaging agents and surface charges while maintaining a set size, allowing for real-time monitoring of kinetic and dynamic tumoral delivery by multimodal fluorescence/positron emission tomography/computed tomography (fluo/PET/CT) imaging, for formulation optimization. The optimized glutathione (GSH)-NFP displays a reduced capture by the RES as well as excellent tumor targeting and tissue invasion properties compared to naive NFP. Inside a tumor, GSH-NFP can structurally transform into ten times larger interfibril networks, serving as in situ depot that promotes weeks-long local retention. This nanofiber, which can further be designed to release the active pharmacophores within a tumor microenvironment, displays a superior therapeutic efficacy for inhibiting disease progression and improving the survival of animals bearing triple-negative breast cancer tumors compared to free drug and liposome formulation of the drug, in addition to a favorable toxicity profile.

8.
ACS Med Chem Lett ; 9(2): 114-119, 2018 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-29456798

RESUMO

Histone deacetylase (HDAC) inhibition is becoming an increasingly popular approach to treat cancer, as HDAC overexpression is common in many malignancies. The blood-brain barrier (BBB) prevents systemically delivered drugs from reaching brain at effective concentration, making small-molecule-HDAC inhibition in brain tumors particularly challenging. To circumvent the BBB, novel routes for administering therapeutics are being considered in the clinic, and a need exists for drugs whose deliveries can be directly imaged, so that effective delivery across the BBB can be monitored. We report chemistry for radiolabeling the HDAC inhibitor, panobinostat, with fluoride-18 (compound-1). Like panobinostat, compound 1 retains nanomolar efficacy in diffuse intrinsic pontine glioma (DIPG IV and XIII) cells (IC50 = 122 and 108 nM, respectively), with lesser activity against U87 glioma. With a favorable therapeutic ratio, 1 is highly selective to glioma and demonstrates considerably less toxicity toward healthy astrocyte controls (IC50 = 5265 nM). Compound 1 is stable in aqueous solution at physiological pH (>7 days, fetal bovine serum), and its delivery can be imaged by positron emission tomography (PET). Compound 1 is synthesized in two steps, and employs rapid, late-stage aqueous isotopic exchange 18F-radiochemistry. PET is used to image the in vivo delivery of [18F]-1 to the murine central nervous system via convection enhanced delivery.

9.
J Neurosurg ; 129(1): 10-18, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-28885119

RESUMO

OBJECTIVE Drug clearance may be a limiting factor in the clinical application of convection-enhanced delivery (CED). Peptide-based nanofibers (NFPs) have a high aspect ratio, and NFPs loaded with drugs could potentially maintain effective drug concentrations for an extended period sufficient for cancer therapy. The objective of this study was to assess the volume of distribution (Vd) and clearance of variable lengths of NFPs when administered using CED. METHODS NFPs composed of multiple methoxypolyethylene glycol (mPEG)-conjugated constructs (mPEG2000-KLDLKLDLKLDL-K( FITC)-CONH2, for which FITC is fluorescein isothiocyanate) were assembled in an aqueous buffer. The NFPs were approximately 5 nm in width and were formulated into different lengths: 100 nm (NFP-100), 400 nm (NFP-400), and 1000 nm (NFP-1000). The NFP surface was covalently conjugated with multiple Cy5.5 fluorophores as the optical reporters to track the post-CED distribution. Forty-two 6- to 8-week-old Ntv-a;p53fl/fl mice underwent CED to the striatum. Animals were killed immediately, 24 hours or 72 hours after CED. The brains were extracted and sectioned for assessing NFP Vd to volume of infusion (Vi) ratio, and clearance using fluorescence microscopy. RESULTS CED of NFPs was well tolerated by all the animals. The average Vd/Vi ratios for NFP-100, NFP-400, NFP-1000, and unconjugated positive control (free Cy5.5) were 1.87, 2.47, 1.07, and 3.0, respectively, which were statistically different (p = 0.003). The percentages remaining of the original infusion volume at 24 hours for NFP-100, -400, and -1000 were 40%, 90%, and 74%, respectively. The percentages remaining at 72 hours for NFP-100, -400, and -1000 were 15%, 30%, and 46%, respectively. Unconjugated Cy5.5 was not detected at 24 or 72 hours after CED. CONCLUSIONS CED of NFPs is feasible with Vd/Vi ratios and clearance rates comparable to other nanocarriers. Of the 3 NFPs, NFP-400 appears to provide the best distribution and slowest clearance after 24 hours. NFP provides a dynamic theranostic platform, with the potential to deliver clinically efficacious drug payload to brain tumor after CED.


Assuntos
Encéfalo/metabolismo , Nanofibras , Peptídeos/metabolismo , Peptídeos/farmacocinética , Animais , Portadores de Fármacos , Camundongos , Distribuição Tecidual
10.
Mol Cancer Ther ; 16(12): 2902-2912, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28978723

RESUMO

The blood brain barrier can limit the efficacy of systemically delivered drugs in treating neurological malignancies; therefore, alternate routes of drug administration must be considered. The Abl-kinase inhibitor, dasatinib, is modified to give compound 1 ([18F]-1) so that 18F-positron emission tomography (PET) and fluorescent imaging can both be used to observe drug delivery to murine orthotopic glioma. In vitro Western blotting, binding studies (IC50 = 22 ± 5 nmol/L), and cell viability assays (IC50 = 46 ± 30 nmol/L) confirm nanomolar, in vitro effectiveness of [18F]-1, a dasatinib derivative that is visible by 18F-PET and fluorescence. [18F]-1 is used to image dynamic direct drug delivery via two different drug delivery techniques to orthotopic murine brainstem glioma (mBSG) bearing mice. Convection enhanced delivery (CED) delivers higher concentrations of drug to glioma-containing volumes versus systemic, tail-vein delivery. Accurate delivery and clearance data pertaining to dasatinib are observed, providing personalized information that is important in dosimetry and redosing. Cases of missed drug delivery are immediately recognized by PET/CT, allowing for prompt intervention in the case of missed delivery. Mol Cancer Ther; 16(12); 2902-12. ©2017 AACR.


Assuntos
Antineoplásicos/farmacologia , Neoplasias do Tronco Encefálico/diagnóstico por imagem , Neoplasias do Tronco Encefálico/tratamento farmacológico , Dasatinibe/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Glioma/diagnóstico por imagem , Glioma/tratamento farmacológico , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Animais , Neoplasias do Tronco Encefálico/patologia , Sistemas Computacionais , Modelos Animais de Doenças , Glioma/patologia , Camundongos
11.
PLoS One ; 12(2): e0171044, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28196146

RESUMO

Ovarian cancer (OVC) patients who receive chemotherapy often acquire drug resistance within one year. This can lead to tumor reoccurrence and metastasis, the major causes of mortality. We report a transient increase of a small distinctive CXCR4High/CD24Low cancer stem cell population (CXCR4High) in A2780 and SKOV-3 OVC cell lines in response to cisplatin, doxorubicin, and paclitaxel, treatments. The withdrawal of the drug challenges reversed this cell-state transition. CXCR4High exhibits dormancy in drug resistance and mesenchymal-like invasion, migration, colonization, and tumor formation properties. The removal of this cell population from a doxorubicin-resistant A2780 lineage (A2780/ADR) recovered the sensitivity to drug treatments. A cytotoxic peptide (CXCR4-KLA) that can selectively target cell-surface CXCR4 receptor was further synthesized to investigate the therapeutic merits of targeting CXCR4High. This peptide was more potent than the conventional CXCR4 antagonists (AMD3100 and CTCE-9908) in eradicating the cancer stem cells. When used together with cytotoxic agents such as doxorubicin and cisplatin, the combined drug-peptide regimens exhibited a synergistic cell-killing effect on A2780, A2780/ADR, and SKOV-3. Our data suggested that chemotherapy could establish drug-resistant and tumor-initiating properties of OVC via reversible CXCR4 cell state transition. Therapeutic strategies designed to eradicate rather than antagonize CXCR4High might offer a far-reaching potential as supportive chemotherapy.


Assuntos
Citotoxinas/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Proteínas de Neoplasias/antagonistas & inibidores , Células-Tronco Neoplásicas/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Peptídeos/farmacologia , Receptores CXCR4/antagonistas & inibidores , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Feminino , Humanos , Invasividade Neoplásica , Metástase Neoplásica , Proteínas de Neoplasias/metabolismo , Células-Tronco Neoplásicas/patologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Receptores CXCR4/metabolismo
12.
Macromol Biosci ; 17(2)2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27654639

RESUMO

The unsatisfactory outcomes of typical multiple cytotoxic chemotherapeutic combination therapies used to treat patients have fostered a need for new unconventional combinations of therapeutic agents. Among the candidates, siRNA has been widely discussed and tested. However, the right time right place codelivery of siRNA with other types of active ingredients is challenging because of the possible differences among their physiochemical and pharmacodynamics properties. To accomplish a synergistic cytotoxic effect, a nanoassembly is thus designed to codeliver siRNA with other therapeutic agents. A siRNA, targeting prosurvival gene for the p75 neurotrophin receptor, and an organelle-fusing peptide, targeting mitochondria, are layered onto a nanotemplate by charge-charge interaction, followed by a layer of CD44 targeting ligand. The formulated triple-functional nanomedicine is efficiently internalized by the CD44 expressing triple-negative breast cancer cells. The encapsulated siRNA and the pro-apoptotic peptide are released inside cells, silencing the intended prosurvival gene, and inducing apoptosis by fusing the mitochondrial membrane, respectively. A synergistic effect is achieved by this three-agent combination. The design of the developed multifunctional nanomedicine can be generalized to deliver other siRNA and drugs for a maximum therapeutic combination with minimal off-targeting effects.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Nanopartículas/química , RNA Interferente Pequeno/administração & dosagem , Morte Celular , Linhagem Celular Tumoral , Terapia Combinada , Feminino , Inativação Gênica , Humanos , Receptores de Hialuronatos/metabolismo , Nanomedicina , Peptídeos/química , Neoplasias de Mama Triplo Negativas/patologia
13.
JCI Insight ; 1(19): e90064, 2016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-27882353

RESUMO

Clinical monitoring of adoptive T cell transfer (ACT) utilizes serial blood analyses to discern T cell activity. While useful, these data are 1-dimensional and lack spatiotemporal information related to treatment efficacy or toxicity. We utilized a human genetic reporter, somatostatin receptor 2 (SSTR2), and PET, to quantitatively and longitudinally visualize whole-body T cell distribution and antitumor dynamics using a clinically approved radiotracer. Initial evaluations determined that SSTR2-expressing T cells were detectable at low densities with high sensitivity and specificity. SSTR2-based PET was applied to ACT of chimeric antigen receptor (CAR) T cells targeting intercellular adhesion molecule-1, which is overexpressed in anaplastic thyroid tumors. Timely CAR T cell infusions resulted in survival of tumor-bearing mice, while later infusions led to uniform death. Real-time PET imaging revealed biphasic T cell expansion and contraction at tumor sites among survivors, with peak tumor burden preceding peak T cell burden by several days. In contrast, nonsurvivors displayed unrelenting increases in tumor and T cell burden, indicating that tumor growth was outpacing T cell killing. Thus, longitudinal PET imaging of SSTR2-positive ACT dynamics enables prognostic, spatiotemporal monitoring with unprecedented clarity and detail to facilitate comprehensive therapy evaluation with potential for clinical translation.


Assuntos
Imunoterapia Adotiva , Neoplasias Experimentais/terapia , Tomografia por Emissão de Pósitrons , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/citologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Genes Reporter , Humanos , Células Jurkat , Camundongos , Transdução Genética , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Biomacromolecules ; 17(6): 2040-9, 2016 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-27180972

RESUMO

We previously reported a high aspect ratio peptide nanofiber that could be effectively delivered to tumors with minimal nonspecific uptake by other organs. The peptidic nature offers the design flexibility of smart formulation with unique responsiveness. Two new formulations that behave congruously as nanotransformers (NTFs) are reported herein. NTF1 and NTF2 could biomechanically remodel upon enzyme activation to generate a degradable and an aggregable effect, respectively, within the lysosomal compartment. These NTFs were further evaluated as carriers of mertansine (DM1), a microtubule inhibitor. DM1-loaded NTF1 could be degraded by cathepsin B (CathB) to release the same active metabolite, as previously described in the lysosomal degradation of antibody-DM1 conjugate. In contrast, CathB only partially digested DM1-loaded NTF2 and induced aggregate formation to become a storage reservoir with slow payload release property. The DM1-loaded NTF1 exhibited a comparable cytotoxicity to the free drug and was more effective than the NTF2 formulation in eradicating triple negative breast cancer. Our data suggested that biological transformers with distinct enzyme-induced structural changes and payload release profiles could be designed for the intracellular delivery of cytotoxic and imaging agents.


Assuntos
Catepsina B/metabolismo , Portadores de Fármacos/química , Maitansina/análogos & derivados , Nanofibras/química , Oligopeptídeos/química , Moduladores de Tubulina/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/enzimologia , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Liberação Controlada de Fármacos , Feminino , Humanos , Maitansina/administração & dosagem , Maitansina/farmacologia , Maitansina/uso terapêutico , Microscopia de Fluorescência , Moduladores de Tubulina/farmacologia , Moduladores de Tubulina/uso terapêutico
15.
Int J Biochem Cell Biol ; 57: 54-62, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25310905

RESUMO

Recent studies have suggested that the receptor for advanced glycation end products (RAGE) participates in melanoma progression by promoting tumor growth. However, the mechanisms of RAGE activation in melanoma tumors are not clearly understood. To get deeper insights into these mechanisms, we transfected a melanoma cell line, which was established from a human melanoma primary tumor, with RAGE, and studied the effect of RAGE overexpression on cell proliferation and migration in vitro. We observed that overexpression of RAGE in these cells not only resulted in significantly increased migration rates compared to control cells, but also in decreased proliferation rates (Meghnani et al., 2014). In the present study, we compared the growth of xenograft tumors established from RAGE overexpressing WM115 cells, to that of control cells. We observed that when implanted in mice, RAGE overexpressing cells generated tumors faster than control cells. Analysis of protein tumor extracts showed increased levels of the RAGE ligands S100B, S100A2, S100A4, S100A6 and S100A10 in RAGE overexpressing tumors compared to control tumors. We show that the tumor growth was significantly reduced when the mice were treated with anti-RAGE antibodies, suggesting that RAGE, and probably several S100 proteins, were involved in tumor growth. We further demonstrate that the anti-RAGE antibody treatment significantly enhanced the efficacy of the alkylating drug dacarbazine in reducing the growth rate of RAGE overexpressing tumors.


Assuntos
Melanoma/metabolismo , Receptores Imunológicos/metabolismo , Proteínas S100/biossíntese , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Dacarbazina/farmacologia , Feminino , Humanos , Ligantes , Melanoma/tratamento farmacológico , Camundongos , Camundongos SCID , Distribuição Aleatória , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/antagonistas & inibidores , Receptores Imunológicos/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Small ; 9(12): 2129-39, 2013 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-23359548

RESUMO

The ability to map multiple biomarkers at the same time has far-reaching biomedical and diagnostic applications. Here, a series of biocompatible poly(D,L-lactic-co-glycolic acid) and polyethylene glycol particles for multicolor and multiplexed imaging are reported. More than 30 particle formulations that exhibit distinct emission signatures (ranging from the visible to NIR wavelength region) are designed and synthesized. These particles are encapsulated with combinations of carbocyanine-based fluorophores DiO, Dil, DiD, and DiR, and are characterized as <100 nm in size and brighter than commercial quantum dots. A particle formulation is identified that simultaneously emits fluorescence at three different wavelengths upon a single excitation at 485 nm via sequential and multiple FRET cascade events for multicolor imaging. Three other particles that display maximum fluorescence intensities at 570, 672, or 777 nm for multiplexed imaging are also identified. These particles are individually conjugated with specific (Herceptin or IgG2A11 antibody) or nonspecific (heptaarginine) ligands for targeting and, thus, could be applied to differentiate different cancer cells from a cell mixture according to the expressions of cell-surface human epidermal growth factor receptor 2 and the receptor for advanced glycation endproducts. Using an animal model subcutaneously implanted with the particles, it is further demonstrated that the developed platform could be useful for in vivo multiplexed imaging.


Assuntos
Diagnóstico por Imagem/métodos , Transferência Ressonante de Energia de Fluorescência/métodos , Nanopartículas/química , Polímeros/química , Ácido Láctico/química , Polietilenoglicóis/química , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico
17.
J Control Release ; 167(1): 1-10, 2013 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-23352910

RESUMO

Targeted nano-particulate systems hold extraordinary potential for delivery of therapeutics across blood brain barrier (BBB). In this work, we investigated the potential of novel bi-ligand (transferrin-poly-l-arginine) liposomal vector for delivery of desired gene to brain, in vivo. The in vivo evaluation of the delivery vectors is essential for clinical translation. We followed an innovative approach of combining transferrin receptor targeting with enhanced cell penetration to design liposomal vectors for improving the transport of molecules into brain. The biodistribution profile of 1, 1'-dioctadecyl-3,3,3',3'-tetramethyl-indocarbocyanine iodide(DiR)-labeled liposomes was evaluated in adult rats after single intravenous injection at dose of 15.2µmoles of phospholipids/kg body weight. We demonstrated that bi-ligand liposomes accumulated in rat brain at significantly (p<0.05) higher concentrations as compared to the single-ligand (transferrin) or plain liposomes. In addition, the bi-ligand liposomes resulted in increased expression of ß-galactosidase(ß-gal) plasmid in rat brain tissue in comparison to the single-ligand liposomes. Histological examination of the transfected tissues did not show any signs of tissue necrosis or inflammation. Hemolysis assay further authenticated the biocompatibility of bi-ligand liposomes in blood up to 600 nmoles of phospholipids/1.4×10(7) erythrocytes. The findings of this study provide important and detailed information regarding the distribution of bi-ligand liposomes in vivo and accentuate their ability to demonstrate improved brain penetration and transfection potential over single-ligand liposomes.


Assuntos
Encéfalo/metabolismo , Peptídeos Penetradores de Células/farmacocinética , Peptídeos/farmacocinética , Transferrina/farmacocinética , Animais , Carbocianinas/química , Carbocianinas/farmacocinética , Peptídeos Penetradores de Células/química , Eritrócitos/citologia , Corantes Fluorescentes/química , Corantes Fluorescentes/farmacocinética , Hemólise , Ligantes , Lipossomos , Peptídeos/química , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual , Transfecção/métodos , Transferrina/química , beta-Galactosidase/metabolismo
18.
Free Radic Biol Med ; 57: 49-60, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23261941

RESUMO

Through free radical-mediated peroxidation, cyclooxygenase (COX) can metabolize dihomo-γ-linolenic acid (DGLA) and arachidonic acid (AA) to form well-known bioactive metabolites, namely, the 1-series of prostaglandins (PGs1) and the 2-series of prostaglandins (PGs2), respectively. Unlike PGs2, which are generally viewed as proinflammatory and procarcinogenic PGs, PGs1 may possess anti-inflammatory and anti-cancer activity. Previous studies using ovine COX along with spin trapping and the LC/ESR/MS technique have shown that certain exclusive free radicals are generated from different free radical reactions in DGLA and AA peroxidation. However, it has been unclear whether the differences were associated with the contrasting bioactivity of DGLA vs AA. The aim of this study was to refine the LC/MS and spin trapping technique to make it possible for the association between free radicals and cancer cell growth to be directly tested. Using a colon cancer cell line, HCA-7 colony 29, and LC/MS along with a solid-phase extraction, we were able to characterize the reduced forms of radical adducts (hydroxylamines) as the free radicals generated from cellular COX-catalyzed peroxidation. For the first time, free radicals formed in the COX-catalyzed peroxidation of AA vs DGLA and their association with cancer cell growth were assessed (cell proliferation via MTS and cell cycle distribution via propidium iodide staining) in the same experimental setting. The exclusive free radicals formed from the COX-catalyzed peroxidation of AA and DGLA were shown to be correlated with the cell growth response. Our results indicate that free radicals generated from the distinct radical reactions in COX-catalyzed peroxidation may represent the novel metabolites of AA and DGLA that correspond to their contrasting bioactivity.


Assuntos
Ácido 8,11,14-Eicosatrienoico/metabolismo , Ácido Araquidônico/metabolismo , Radicais Livres/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Cromatografia Líquida , Neoplasias do Colo , Espectroscopia de Ressonância de Spin Eletrônica , Humanos , Hidroxilaminas/metabolismo , Espectrometria de Massas , Prostaglandinas/metabolismo , Detecção de Spin
19.
Nanomedicine ; 9(4): 449-57, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23178287

RESUMO

The cellular interactions and in vivo distribution of the nanomaterials are known to be strongly influenced by their physiochemical properties. Here, we investigated and compared the biocompatibility, pharmacokinetics, and biodistribution of previously reported peptide-based nanofiber (NFP), with commercially available nanomaterials. The NFP was a 2-dimensional (2D) structure with an extremely narrow width (4 nm) and a controllable length (50 to 400 nm). NFP was found to be non-toxic, hemocompatible, and with a minimum uptake by macrophages. In vivo studies further demonstrated that NFP could be delivered to the tumor site more effectively, and within a very shorter period of time, than spherical nanoparticles. Importantly, the undelivered NFP was rapidly eliminated by renal clearance and, thus, avoiding its accumulation in the spleen or liver. Overall, our data suggested a new paradigm in drug delivery via using a short circulating NFP, rather than a long circulating 3D nanoparticle, as a delivery cargo. FROM THE CLINICAL EDITOR: In this study, the role of small peptide-based nanocarriers is investigated for tumor-specific delivery, reporting excellent targeting properties and a favorable toxicity profile.


Assuntos
Antineoplásicos/administração & dosagem , Nanofibras , Neoplasias/tratamento farmacológico , Peptídeos/administração & dosagem , Animais , Antineoplásicos/farmacocinética , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos SCID , Distribuição Tecidual
20.
Analyst ; 137(23): 5487-90, 2012 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-23061092

RESUMO

Herein, we report the application of synthesized fluorescent, water soluble polymers for post-translational subtyping and differentiation of breast cancer cells in vitro. The fluorescence emission spectra from these polymers were modulated differently in the presence of conditioned cell culture media from various breast cancer cells. These polymers differentiate at a post-translation level possibly due to their ability to interact with extracellular enzymes that are over-expressed in cancerous conditions.


Assuntos
Neoplasias da Mama/classificação , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Meios de Cultivo Condicionados , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Feminino , Corantes Fluorescentes , Células HEK293 , Células HeLa , Humanos , Metaloproteínas/análise , Metaloproteínas/antagonistas & inibidores , Polímeros , Espectrometria de Fluorescência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA