Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
AAPS J ; 23(5): 102, 2021 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-34435271

RESUMO

Mathematical modeling has been an important tool in pharmaceutical research for 50 + years and there is increased emphasis over the last decade on using modeling to improve the efficiency and effectiveness of drug development. In an earlier commentary, we applied a multiscale model linking 6 scales (whole body, tumor, vasculature, cell, spatial location, time), together with literature data on nanoparticle and tumor properties, to demonstrate the effects of nanoparticle particles on systemic disposition. The current commentary used a 4-scale model (cell membrane, intracellular organelles, spatial location, time) together with literature data on the intracellular processing of membrane receptors and transporters to demonstrate disruption of transporter homeostasis can lead to drug-drug interaction (DDI) between victim drug (VD) and perpetrator drug (PD), including changes in the area-under-concentration-time-curve of VD in cells that are considered significant by the US Food and Drug Administration (FDA). The model comprised 3 computational components: (a) intracellular transporter homeostasis, (b) pharmacokinetics of extracellular and intracellular VD/PD concentrations, and (c) pharmacodynamics of PD-induced stimulation or inhibition of an intracellular kinetic process. Model-based simulations showed that (a) among the five major endocytic processes, perturbation of transporter internalization or recycling led to the highest incidence and most extensive DDI, with minor DDI for perturbing transporter synthesis and early-to-late endosome and no DDI for perturbing transporter degradation and (b) three experimental conditions (spatial transporter distribution in cells, VD/PD co-incubation time, extracellular PD concentrations) were determinants of DDI detection. We propose modeling is a useful tool for hypothesis generation and for designing experiments to identify potential DDI; its application further aligns with the model-informed drug development paradigm advocated by FDA.


Assuntos
Desenvolvimento de Medicamentos/métodos , Interações Medicamentosas , Transportador 1 de Ânion Orgânico Específico do Fígado/metabolismo , Modelos Biológicos , Membro 1B3 da Família de Transportadores de Ânion Orgânico Carreador de Soluto/metabolismo , Área Sob a Curva , Simulação por Computador , Hepatócitos/metabolismo , Homeostase , Humanos
2.
Pharmaceutics ; 13(7)2021 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-34209265

RESUMO

Exosomes, naturally occurring vesicles secreted by cells, are undergoing development as drug carriers. We used experimental and computational studies to investigate the kinetics of intracellular exosome processing and exosome-mediated drug efflux and the effects of exosome inhibition. The experiments used four human-breast or ovarian cancer cells, a cytotoxic drug paclitaxel (PTX), two exosome inhibitors (omeprazole (OME), which inhibits exosome release, and GW4869 (GW), which inhibits synthesis of sphingolipid ceramide required for exosome formation), LC-MS/MS analysis of PTX levels in exosomes, and confocal microscopic study of endocytic transport (monitored using fluorescent nanoparticles and endocytic organelle markers). In all four cells, exosome production was enhanced by PTX but diminished by OME or GW (p < 0.05); the PTX enhancement was completely reversed by OME or GW. Co-treatment with OME or GW simultaneously reduced PTX amount in exosomes and increased PTX amount and cytotoxicity in exosome-donor cells (corresponding to >2-fold synergy as indicated by curve shift and uncertainty envelope analyses). This synergy is consistent with the previous reports that OME co-administration significantly enhances the taxane activity in tumor-bearing mice and in patients with triple negative metastatic breast cancer. The experimental results were used to develop a quantitative pharmacology model; model simulations revealed the different effects of the two exosome inhibitors on intracellular PTX processing and subcellular distribution.

3.
Pharmacol Rev ; 71(2): 157-169, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30846487

RESUMO

Quantitative systems pharmacology (QSP), an emerging field that entails using modeling and computation to interpret, interrogate, and integrate drug effects spanning from the molecule to the whole organism to forecast treatment outcomes, is expected to enhance the efficiency of drug development. Since late 2017, the U.S. Food and Drug Administration has advocated the use of an analogous approach of model-informed drug development. This review focuses on issues pertaining to nanosized medicines (NP) and the potential utility of QSP to determine NP delivery and residence at extracellular or intracellular targets in vivo. The kinetic processes governing NP disposition and transport, interactions with biologic matrix components, binding and internalization in cells, and intracellular trafficking are determined, sometimes jointly, by NP properties (e.g., dimension, materials, surface charge and modifications, shape, and geometry) and target tissue properties (e.g., perfusion status, vessel pore size and wall thickness, vessel and cell density, composition of extracellular matrix, and void volume fraction). These various determinants, together with the heterogeneous tissue structures and microenvironment factors in solid tumors, lead to environment-, spatial-, and time-dependent changes in NP concentrations that are difficult to predict. Adding to the complexity is the recent discovery that NP surface-coating protein corona, whose composition depends on NP properties and which undergoes continuous evolution with time and local protein environments, is yet another unpredictable variable. Examples are provided to demonstrate the potential utility of QSP-based multiscale modeling to capture the physicochemical and biologic processes in equations to enable computational studies of the key kinetic processes in cancer treatments.


Assuntos
Sistemas de Liberação de Medicamentos , Desenvolvimento de Medicamentos/métodos , Nanoestruturas/administração & dosagem , Animais , Humanos , Modelos Teóricos , Nanomedicina/métodos , Neoplasias/tratamento farmacológico , Farmacologia , Biologia de Sistemas/métodos , Fatores de Tempo , Microambiente Tumoral
4.
AAPS J ; 21(2): 24, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30710324

RESUMO

Approval of generic drugs by the US Food and Drug Administration (FDA) requires the product to be pharmaceutically equivalent to the reference listed drug (RLD) and demonstrate bioequivalence (BE) in effectiveness when administered to patients under the conditions in the RLD product labeling. Effectiveness is determined by drug exposure at the target sites. However, since such measurement is usually unavailable, systemic exposure is assumed to equal target site exposure and systemic BE to equal target site BE. This assumption, while it often applies to small molecule drug products that are readily dissolved in biological fluids and systemically absorbed, is unlikely to apply to nanotechnology products (NP) that exist as heterogeneous systems and are subjected to dimension- and material-dependent changes. This commentary provides an overview of the intersecting and spatial-dependent processes and variables governing the delivery and residence of oncologic NP in solid tumors. In order to provide a quantitative perspective of the collective effects of these processes, we used quantitative systems pharmacology (QSP) multi-scale modeling to capture the physicochemical and biological events on several scales (whole-body, organ/suborgan, cell/subcellular, spatial locations, time). QSP is an emerging field that entails using modeling and computation to facilitate drug development; an analogous approach (i.e., model-informed drug development) is advocated by to FDA. The QSP model-based simulations illustrated that small changes in NP attributes (e.g., size variations during manufacturing, interactions with proteins in biological milieu) could lead to disproportionately large differences in target site exposure, rending systemic BE unlikely to equal target site BE.


Assuntos
Antineoplásicos/administração & dosagem , Aprovação de Drogas , Portadores de Fármacos/farmacocinética , Medicamentos Genéricos/farmacocinética , United States Food and Drug Administration/normas , Antineoplásicos/farmacocinética , Humanos , Nanopartículas , Neoplasias/tratamento farmacológico , Tamanho da Partícula , Equivalência Terapêutica , Distribuição Tecidual , Estados Unidos , United States Food and Drug Administration/legislação & jurisprudência
5.
J Control Release ; 270: 101-113, 2018 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-29203416

RESUMO

RNA Interference (RNAi) is a potentially useful tool to correct the detrimental effects of faulty genes; several RNAi are undergoing clinical evaluation in various diseases. The present study identified the relative contributions of three mechanisms by which polyanion drugs reduced the gene silencing activity of Lipoplex, a complex of small interfering RNA (siRNA) and cationic liposomes. The study used a siRNA against the chemoresistance gene survivin and two model polyanion drugs (suramin, heparin). Products of Lipoplex destabilization were separated, identified, and/or quantified using ultrafiltration, gel electrophoresis, and RT-qPCR (quantitative reverse transcription polymerase chain reaction). Cell binding and endocytosis of fluorescence-labeled Lipoplex and the amount of siRNA at its site of action RISC (RNA-induced silencing complex) were evaluated using endocytosis markers, confocal microscopy, quantitative image analysis, immunoprecipitation, and RT-qPCR. The results show suramin and heparin exerted multiple concentration-dependent effects. First, these agents altered several Lipoplex properties (i.e., reduced particle size, changed surface charge, modified composition of protein biocorona). Second, both caused Lipoplex destabilization to release double- and single-strand siRNA and/or smaller siRNA-lipid complexes with reduced siRNA cargo. Third, both prevented the cell surface binding and internalization of Lipoplex, diminished the siRNA concentration in RISC, and retarded the mRNA knockdown. Suramin and heparin yielded qualitatively and quantitatively different results. Analysis of the experimental results of suramin using quantitative pharmacology (QP) modeling indicated the major cause of gene silencing activity loss depended on drug concentration, changing from inhibition of endocytosis at lower concentration (accounting for 60% loss at ~9µM) to inhibition of cell surface binding and loss of siRNA cargo at higher concentrations (accounting for 64% and 27%, respectively, at 70µM). In summary, the present study demonstrates the complex and dynamic interactions between polyanions and Lipoplex, and the use of QP modeling to delineate the contributions of three mechanisms to the eventual loss of gene silencing activity.


Assuntos
Heparina/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , Suramina/administração & dosagem , Survivina/genética , Disponibilidade Biológica , Inativação Gênica , Células HT29 , Humanos , Lipossomos , Transfecção
6.
Curr Cancer Drug Targets ; 17(8): 735-755, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28359247

RESUMO

BACKGROUND: Commonly used methods for analyzing interactivity between drugs (e.g. synergy, antagonism) such as isobologram, combination index, and curve shift are based on the Loewe Additivity principle of dose equivalence and the inherent assumption of similar concentration- effect (C-E) including parallel curves and equal maximum effects (Emax), and therefore are not suitable for drugs with dissimilar C-E. This study describes a new method that is without this limitation and has the additional advantage of enabling statistical analysis. METHODS AND RESULTS: The method comprises two steps. First, based on the dose equivalence principle, the experimentally obtained C-E of one drug was used to calculate the equally effective C-E of the other drug at no interactivity; the resulting two zero-interactivity C-E formed the upper and lower boundaries of Additivity Envelope. Next, 95% confidence intervals calculated from experimental data were added to Additivity Envelope to obtain Uncertainty Envelope (UE). Experimentally observed effects of drug combinations (C-Ecomb,observed) located within UE indicate additivity whereas C-Ecomb,observed located above or below UE indicate statistically significant (p<0.05) synergy or antagonism, respectively. Additional in silico studies demonstrated the shape and size of Additivity Envelope, which determines the ability to detect drug interactivity, depended on the Drug A-to-B concentration ratios and the ratios of their C-E curve shape parameter. Analyses of experimental results of combinations of drugs with nonparallel C-E and/or unequal Emax indicated UE as more versatile and provided more information, compared to earlier methods. CONCLUSION: UE is a broadly applicable method for analysis, including statistical significance assessment, of drug interactivity.


Assuntos
Interações Medicamentosas , Preparações Farmacêuticas/administração & dosagem , Linhagem Celular Tumoral , Simulação por Computador , Relação Dose-Resposta a Droga , Humanos
7.
Adv Drug Deliv Rev ; 97: 280-301, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26686425

RESUMO

Advances in molecular medicine have led to identification of worthy cellular and molecular targets located in extracellular and intracellular compartments. Effectiveness of cancer therapeutics is limited in part by inadequate delivery and transport in tumor interstitium. Parts I and II of this report give an overview on the kinetic processes in delivering therapeutics to their intended targets, the transport barriers in tumor microenvironment and extracellular matrix (TME/ECM), and the experimental approaches to overcome such barriers. Part III discusses new concepts and findings concerning nanoparticle-biocorona complex, including the effects of TME/ECM. Part IV outlines the challenges in animal-to-human translation of cancer nanotherapeutics. Part V provides an overview of the background, current status, and the roles of TME/ECM in immune checkpoint inhibition therapy, the newest cancer treatment modality. Part VI outlines the development and use of multiscale computational modeling to capture the unavoidable tumor heterogeneities, the multiple nonlinear kinetic processes including interstitial and transvascular transport and interactions between cancer therapeutics and TME/ECM, in order to predict the in vivo tumor spatiokinetics of a therapeutic based on experimental in vitro biointerfacial interaction data. Part VII provides perspectives on translational research using quantitative systems pharmacology approaches.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias/metabolismo , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Sistemas de Liberação de Medicamentos , Matriz Extracelular/metabolismo , Humanos , Nanopartículas/administração & dosagem , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Microambiente Tumoral
8.
J Control Release ; 216: 103-10, 2015 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-26272765

RESUMO

The major barrier for using small interfering RNA (siRNA) as cancer therapeutics is the inadequate delivery and transfection in solid tumors. We have previously shown that paclitaxel tumor priming, by inducing apoptosis, expands the tumor interstitial space, improves the penetration and dispersion of nanoparticles and siRNA-lipoplexes in 3-dimensional tumor histocultures, and promotes the delivery and transfection efficiency of siRNA-lipoplexes under the locoregional setting in vivo (i.e., intraperitoneal treatment of intraperitoneal tumors). The current study evaluated whether tumor priming is functional for systemically delivered siRNA via intravenous injection, which would subject siRNA to several additional delivery barriers and elimination processes. We used the same pegylated cationic (PCat)-siRNA lipoplexes as in the intraperitoneal study to treat mice bearing subcutaneous human pancreatic Hs766T xenograft tumors. The target gene was survivin, an inducible chemoresistance gene. The results show single agent paclitaxel delayed tumor growth but also significantly induced the survivin protein level in residual tumors, whereas addition of PCat-siSurvivin completely reversed the paclitaxel-induced survivin and enhanced the paclitaxel activity (p<0.05). In comparison, PCat-siSurvivin alone did not yield survivin knockdown or antitumor activity, indicating the in vivo effectiveness of intravenous siRNA-mediated gene silencing requires paclitaxel cotreatment. Additional in vitro studies showed that paclitaxel promoted the cytoplasmic release of siGLO, a 22 nucleotide double-stranded RNA that has no mRNA targets, from its PCat lipoplex and/or endosomes/lysosomes. Taken together, our earlier and current data show paclitaxel tumor priming, by promoting the interstitial transport and cytoplasmic release, is critical to promote the delivery and transfection of siRNA in vivo. In addition, because paclitaxel has broad spectrum activity and is used to treat multiple types of solid tumors including the hard-to-treat pancreatic cancer, the synergistic paclitaxel+siSurvivin combination represents a potentially useful chemo-gene therapy.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/farmacologia , Paclitaxel/administração & dosagem , Paclitaxel/farmacologia , Neoplasias Pancreáticas/terapia , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/farmacologia , Animais , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Resistencia a Medicamentos Antineoplásicos , Feminino , Inativação Gênica/efeitos dos fármacos , Terapia Genética/métodos , Humanos , Proteínas Inibidoras de Apoptose/efeitos dos fármacos , Injeções Intravenosas , Lipídeos/química , Camundongos , Camundongos Nus , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Peritoneais/tratamento farmacológico , RNA Neoplásico/metabolismo , Survivina , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
9.
AAPS J ; 17(5): 1065-79, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26089090

RESUMO

Intraperitoneal (IP) chemotherapy confers significant survival benefits in cancer patients. However, several problems, including local toxicity and ineffectiveness against bulky tumors, have prohibited it from becoming a standard-of-care. We have developed drug-loaded, tumor-penetrating microparticles (TPM) to address these problems. TPM comprises two components and uses the versatile PLGA or poly(lacticco-glycolic acid) copolymer to provide tumor-selective adherence and pharmacodynamically optimized fractionated dosing to achieve the desired tumor priming (which promotes particle penetration into tumors) plus immediate and sustained antitumor activity. Preclinical studies show that TPM is less toxic and more effective against several IP metastatic tumors with different characteristics (fast vs. slow growing, porous vs. densely packed structures, wide-spread vs. solitary tumors, early vs. late stage, with or without peritoneal carcinomatosis or ascites), compared to the intravenous paclitaxel/Cremophor micellar solution that has been used off-label in previous IP studies. TPM further requires less frequent dosing. These encouraging preclinical results have motivated the follow-up clinical development of TPM. We are working with National Institutes of Health on the IND-enabling studies.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Paclitaxel/administração & dosagem , Neoplasias Peritoneais/tratamento farmacológico , Animais , Antineoplásicos Fitogênicos/farmacocinética , Antineoplásicos Fitogênicos/uso terapêutico , Portadores de Fármacos/química , Desenho de Fármacos , Humanos , Injeções Intraperitoneais , Microesferas , Paclitaxel/farmacocinética , Paclitaxel/uso terapêutico , Neoplasias Peritoneais/patologia , Polietilenoglicóis/química , Polímeros/química
10.
J Urol ; 194(1): 230-7, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25681288

RESUMO

PURPOSE: Survivin inhibits apoptosis and enables tumor cells to escape from therapy induced senescence. High survivin expression is associated with bladder cancer aggressiveness and recurrence. We evaluated whether survivin expression is reduced by siRNA and whether survivin silencing would enhance mitomycin C activity in human RT4 bladder transitional cell tumors in vitro and in vivo. MATERIALS AND METHODS: We assessed the effectiveness of siRNA therapy using 2 newly developed pegylated cationic liposome carriers, PCat and PPCat. Each has a fusogenic lipid to destabilize the endosomal membrane. PPCat further contains paclitaxel to enhance in vivo delivery and transfection of survivin siRNA. In vitro antitumor activity was evaluated by short-term MTT and long-term clonogenicity cytotoxicity assays. In vivo intravenous therapy was assessed in mice bearing subcutaneous tumors. RESULTS: Nontarget siRNA showed no antitumor activity in vitro or in vivo. Treatment of cultured cells with mitomycin C at a 50% cytotoxic concentration enhanced survivin mRNA and protein levels. Adding PPCat or PCat containing survivin siRNA reversed survivin induction and enhanced mitomycin C activity (p <0.05). In tumor bearing mice single agent mitomycin C delayed tumor growth and almost tripled the survivin protein level in residual tumors. Adding PPCat-survivin siRNA, which alone resulted in a minor survivin decrease of less than 10%, completely reversed mitomycin C induced survivin and enhanced mitomycin C activity (p <0.05). CONCLUSIONS: Results indicate that there is effective in vivo survivin silencing and synergism between mitomycin C and PPCat-survivin siRNA. This combination represents a potentially useful chemo-gene therapy for bladder cancer.


Assuntos
Antibióticos Antineoplásicos/uso terapêutico , Carcinoma de Células de Transição/tratamento farmacológico , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Mitomicina/uso terapêutico , Interferência de RNA , RNA Interferente Pequeno/farmacologia , RNA Interferente Pequeno/uso terapêutico , Animais , Carcinoma de Células de Transição/genética , Feminino , Xenoenxertos , Humanos , Injeções Intravenosas , Camundongos , Camundongos Nus , Survivina , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
11.
AAPS J ; 17(1): 268-76, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25425294

RESUMO

We reported that suramin is an effective chemosensitizer at noncytotoxic concentrations (<50 µM); this effect was observed in multiple types of human xenograft tumors in vitro and in vivo. Clinical evaluation of noncytotoxic suramin is ongoing. Because (a) suramin inhibits reverse transcriptase, (b) telomerase is a reverse transcriptase, and (c) inhibition of telomerase enhances tumor chemosensitivity, we studied the pharmacodynamics of noncytotoxic suramin on telomerase activity and telomere length in cultured cells and tumors grown in animals. In three human cancer cells that depend on telomerase for telomere maintenance (pharynx FaDu, prostate PC3, breast MCF7), suramin inhibited telomerase activity in cell extracts and intact cells at concentrations that exhibited no cytotoxicity (IC50 of telomerase was between 1 and 3 µM vs. >60 µM for cytotoxicity), and continuous treatment at 10-25 µM for 6 weeks resulted in gradual telomere shortening (maximum of 30%) and cell senescence (measured by ß-galactosidase activity and elevation of mRNA levels of two senescence markers p16 and p21). In contrast, noncytotoxic suramin did not shorten the telomere in telomerase-independent human osteosarcoma Saos-2 cells. In mice bearing FaDu tumors, treatment with noncytotoxic suramin for 6 weeks resulted in telomere erosion in >95% of the tumor cells with an average telomere shortening of >40%. These results indicate noncytotoxic suramin inhibits telomerase, shortens telomere and induces cell senescence, and suggest telomerase inhibition as a potential mechanism of its chemosensitization.


Assuntos
Antineoplásicos/farmacologia , Suramina/farmacologia , Telomerase/antagonistas & inibidores , Encurtamento do Telômero/efeitos dos fármacos , Animais , Antineoplásicos/administração & dosagem , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Humanos , Concentração Inibidora 50 , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Suramina/administração & dosagem , Telômero/efeitos dos fármacos , Telômero/metabolismo
12.
J Control Release ; 192: 10-8, 2014 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-24995948

RESUMO

Nanotechnology is widely used in cancer research. Models that predict nanoparticle transport and delivery in tumors (including subcellular compartments) would be useful tools. This study tested the hypothesis that diffusive transport of cationic liposomes in 3-dimensional (3D) systems can be predicted based on liposome-cell biointerface parameters (binding, uptake, retention) and liposome diffusivity. Liposomes comprising different amounts of cationic and fusogenic lipids (10-30mol% DOTAP or 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine, 1-20mol% DOPE or 1,2-dioleoyl-3-trimethylammonium-propane, +25 to +44mV zeta potential) were studied. We (a) measured liposome-cell biointerface parameters in monolayer cultures, and (b) calculated effective diffusivity based on liposome size and spheroid composition. The resulting parameters were used to simulate the liposome concentration-depth profiles in 3D spheroids. The simulated results agreed with the experimental results for liposomes comprising 10-30mol% DOTAP and ≤10mol% DOPE, but not for liposomes with higher DOPE content. For the latter, model modifications to account for time-dependent extracellular concentration decrease and liposome size increase did not improve the predictions. The difference among low- and high-DOPE liposomes suggests concentration-dependent DOPE properties in 3D systems that were not captured in monolayers. Taken together, our earlier and present studies indicate the diffusive transport of neutral, anionic and cationic nanoparticles (polystyrene beads and liposomes, 20-135nm diameter, -49 to +44mV) in 3D spheroids, with the exception of liposomes comprising >10mol% DOPE, can be predicted based on the nanoparticle-cell biointerface and nanoparticle diffusivity. Applying the model to low-DOPE liposomes showed that changes in surface charge affected the liposome localization in intratumoral subcompartments within spheroids.


Assuntos
Lipossomos/metabolismo , Neoplasias/metabolismo , Transporte Biológico , Simulação por Computador , Difusão , Ácidos Graxos Monoinsaturados/química , Ácidos Graxos Monoinsaturados/metabolismo , Humanos , Lipossomos/química , Fosfatidiletanolaminas/química , Fosfatidiletanolaminas/metabolismo , Compostos de Amônio Quaternário/química , Compostos de Amônio Quaternário/metabolismo , Esferoides Celulares , Células Tumorais Cultivadas
13.
AAPS J ; 16(3): 424-39, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24570339

RESUMO

This study established a multiscale computational model for intraperitoneal (IP) chemotherapy, to depict the time-dependent and spatial-dependent drug concentrations in peritoneal tumors as functions of drug properties (size, binding, diffusivity, permeability), transport mechanisms (diffusion, convection), spatial-dependent tumor heterogeneities (vessel density, cell density, pressure gradient), and physiological properties (peritoneal pressure, peritoneal fluid volume). Equations linked drug transport and clearance on three scales (tumor, IP cavity, whole organism). Paclitaxel was the test compound. The required model parameters (tumor diffusivity, tumor hydraulic conductivity, vessel permeability and surface area, microvascular hydrostatic pressure, drug association with cells) were obtained from literature reports, calculation, and/or experimental measurements. Drug concentration-time profiles in peritoneal fluid and plasma were the boundary conditions for tumor domain and blood vessels, respectively. The finite element method was used to numerically solve the nonlinear partial differential equations for fluid and solute transport. The resulting multiscale model accounted for intratumoral spatial heterogeneity, depicted diffusive and convective drug transport in tumor interstitium and across blood vessels, and provided drug flux and concentration as a function of time and spatial position in the tumor. Comparison of model-predicted tumor spatiokinetics with experimental results (autoradiographic data of 3H-paclitaxel in IP ovarian tumors in mice, 6 h posttreatment) showed good agreement (1% deviation for area under curve and 23% deviations for individual data points, which were several-fold lower compared to the experimental intertumor variations). The computational multiscale model provides a tool to quantify the effects of drug-, tumor-, and host-dependent variables on the concentrations and residence time of IP therapeutics in tumors.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Injeções Intraperitoneais/métodos , Algoritmos , Animais , Líquido Ascítico/metabolismo , Simulação por Computador , Difusão , Absorção Intestinal , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Distribuição Tecidual
14.
J Control Release ; 178: 79-85, 2014 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-24462901

RESUMO

Cancers originating from the digestive system account for 290,000 or ~20% of all new cancer cases annually in the US. We previously developed paclitaxel-loaded tumor-penetrating microparticles (TPM) for intraperitoneal (IP) treatment of peritoneal tumors (Lu et al., 2008; Tsai et al., 2007; Tsai et al., 2013). TPM is undergoing NIH-supported IND-enabling studies for clinical evaluation. The present study evaluated the hypothesis that TPM, via inducing apoptosis and expanding the interstitial space, promotes the delivery and transfection of lipid vectors containing siRNA. The in vivo model was the metastatic human Hs766T pancreatic tumor that, upon IP injection, produced widely distributed solid tumors and ascites in the peritoneal cavity in 100% of animals. The target gene was survivin, an anti-apoptotic protein induced by chemotherapy and associated with metastases and poor prognosis of patients with gastric and colorectal cancers. The siRNA carrier was pegylated liposomes comprising cationic and neutral lipids plus a fusogenic lipid (PCat). PCat-loaded with survivin siRNA (PCat-siSurvivin) was active in cultured cells (decreased survivin mRNA and protein levels, reduced cell clonogenicity, enhanced paclitaxel activity), but lost its activity in vivo; this difference is consistent with the well-known problem of inadequate delivery and transfection of siRNA in vivo. In comparison, single agent TPM prolonged animal survival and, as expected, induced survivin expression in tumors. Addition of PCat-siSurvivin reversed the TPM-induced survivin expression and enhanced the antitumor activity of TPM. The finding that in vivo survivin knockdown by PCat-siSurvivin was successful only when it was given in combination with TPM provides the proof-of-concept that tumor priming promotes the delivery and transfection of liposomal siRNA. The data further suggest the TPM/PCat-siSurvivin combination as a potentially useful chemo-gene therapy for peritoneal cancer.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Paclitaxel/administração & dosagem , Neoplasias Peritoneais/terapia , RNA Interferente Pequeno/administração & dosagem , Animais , Antineoplásicos Fitogênicos/farmacocinética , Linhagem Celular Tumoral , Terapia Combinada , Feminino , Humanos , Proteínas Inibidoras de Apoptose/genética , Proteínas Inibidoras de Apoptose/metabolismo , Lipossomos , Camundongos , Camundongos Nus , Paclitaxel/farmacocinética , Neoplasias Peritoneais/metabolismo , RNA Interferente Pequeno/farmacocinética , Survivina , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Curr Cancer Drug Targets ; 14(1): 70-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24200079

RESUMO

Intraperitoneal (IP) chemotherapy confers significant survival benefits in cancer patients. However, several problems, including local toxicity and ineffectiveness against bulky tumors, have prohibited it from becoming a standard of care. We have developed drug-loaded, polymeric tumor-penetrating microparticles (TPM) to address these problems. Initial studies showed that TPM provides tumor-selective delivery and is effective against ovarian SKOV3 tumors of relatively small size (<50 mg). The present study evaluated whether the TPM activity extends to other tumor types that are more bulky and have different morphologies and disease presentation. We evaluated TPM in mice bearing two IP human pancreatic tumors with different growth characteristics and morphologies (rapidly growing, large and porous Hs766T vs. slowly growing, smaller and densely packed MiaPaCa2), and at different disease stage (early stage with smaller tumors vs. late stage with larger tumors plus peritoneal carcinomatosis). Comparison of treatments with TPM or paclitaxel in Cremophor micelles, at equi-toxic doses, shows, in all tumor types: (a) higher paclitaxel levels in tumors (up to 55-fold) for TPM, (b) greater efficacy for TPM, including significantly longer survival and higher cure rate, and (c) a single dose of TPM was equally efficacious as multiple doses of paclitaxel/Cremophor. The results indicate tumor targeting property and superior antitumor activity of paclitaxel-loaded TPM are generalizable to small and large peritoneal tumors, with or without accompanying carcinomatosis.


Assuntos
Antineoplásicos/administração & dosagem , Portadores de Fármacos/farmacologia , Paclitaxel/administração & dosagem , Neoplasias Pancreáticas/tratamento farmacológico , Polietilenoglicóis/administração & dosagem , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Portadores de Fármacos/farmacocinética , Sistemas de Liberação de Medicamentos/métodos , Feminino , Humanos , Injeções Intraperitoneais , Camundongos , Camundongos Nus , Paclitaxel/farmacocinética , Paclitaxel/farmacologia , Neoplasias Pancreáticas/patologia , Tamanho da Partícula , Polietilenoglicóis/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
16.
J Am Anim Hosp Assoc ; 50(1): 12-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24216494

RESUMO

Although conventional treatment of dogs with osteosarcoma (OSA) by amputation and chemotherapy results in reported survival times (STs) of 262-413 days, no major improvements in STs have occurred in the past 2 decades. Suramin is a polysulfonated napthylurea, which at noncytotoxic concentrations in vitro, increases tumor sensitivity to chemotherapy, including doxorubicin. The study authors evaluated the combination of noncytotoxic suramin and doxorubicin after amputation in dogs with OSA. The hypothesis was that treatment of dogs with appendicular OSA with amputation, adjuvant doxorubicin, and noncytotoxic suramin would be well tolerated and result in STs at least comparable to those of doxorubicin alone. Forty-seven dogs received 6.75 mg/kg of suramin IV followed by 30 mg/m(2) of doxorubicin IV 4 hr later. Treatment was repeated q 2 wk for five doses. The median disease free time (DFI) was 203 days (range, 42-1,580+ days) and the median ST for all dogs was 369 days (range, 92-1,616+ days). There was no statistical difference in ST and DFI between greyhounds and nonngreyhounds. Adjuvant doxorubicin and noncytotoxic suramin was well tolerated in dogs with OSA following amputation. Additional studies are needed to determine if this combination treatment protocol provides additional clinical benefit compared with doxorubicin alone.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Neoplasias Ósseas/veterinária , Doenças do Cão/tratamento farmacológico , Osteossarcoma/veterinária , Amputação Cirúrgica/veterinária , Animais , Neoplasias Ósseas/tratamento farmacológico , Quimioterapia Adjuvante/veterinária , Doenças do Cão/cirurgia , Cães , Doxorrubicina/administração & dosagem , Osteossarcoma/tratamento farmacológico , Suramina/administração & dosagem , Análise de Sobrevida , Resultado do Tratamento
17.
J Control Release ; 172(3): 737-44, 2013 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-24056144

RESUMO

Intraperitoneal therapy (IP) has demonstrated survival advantages in patients with peritoneal cancers, but has not become a widely practiced standard-of-care in part due to local toxicity and sub-optimal drug delivery. Paclitaxel-loaded, polymeric microparticles were developed to overcome these limitations. The present study evaluated the effects of microparticle properties on paclitaxel release (extent and rate) and in vivo pharmacodynamics. In vitro paclitaxel release from microparticles with varying physical characteristics (i.e., particle size, copolymer viscosity and composition) was evaluated. A method was developed to simulate the dosing rate and cumulative dose released in the peritoneal cavity based on the in vitro release data. The relationship between the simulated drug delivery and treatment outcomes of seven microparticle compositions was studied in mice bearing IP human pancreatic tumors, and compared to that of the intravenous Cremophor micellar paclitaxel solution used off-label in previous IP studies. Paclitaxel release from polymeric microparticles in vitro was multi-phasic; release was greater and more rapid from microparticles with lower polymer viscosities and smaller diameters (e.g., viscosity of 0.17 vs. 0.67 dl/g and diameter of 5-6 vs. 50-60 µm). The simulated drug release in the peritoneal cavity linearly correlated with treatment efficacy in mice (r(2)>0.8, p<0.001). The smaller microparticles, which distribute more evenly in the peritoneal cavity compared to the large microparticles, showed greater dose efficiency. For single treatment, the microparticles demonstrated up to 2-times longer survival extension and 4-times higher dose efficiency, relative to the paclitaxel/Cremophor micellar solution. Upon repeated dosing, the paclitaxel/Cremophor micellar solution showed cumulative toxicity whereas the microparticle that yielded 2-times longer survival did not display cumulative toxicity. The efficacy of IP therapy depended on both temporal and spatial factors that were determined by the characteristics of the drug delivery system. A combination of fast- and slow-releasing microparticles with 5-6 µm diameter provided favorable spatial distribution and optimal drug release for IP therapy.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Preparações de Ação Retardada/química , Ácido Láctico/química , Paclitaxel/administração & dosagem , Neoplasias Pancreáticas/tratamento farmacológico , Ácido Poliglicólico/química , Animais , Antineoplásicos Fitogênicos/farmacocinética , Antineoplásicos Fitogênicos/uso terapêutico , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Nus , Paclitaxel/farmacocinética , Paclitaxel/uso terapêutico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico
18.
AAPS J ; 15(3): 816-31, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23605950

RESUMO

The rapidly evolving nanotechnology field highlights the need of better understanding the relationship between nanoparticle (NP) properties and NP transport in solid tumors. The present study tested the hypothesis that the diffusive transport and spatial distribution of NP can be predicted based on the following parameters: interstitial NP diffusivity, NP-cell interaction parameters (cell surface binding capacity, rate constants of association, dissociation, and internalization). We (a) established the models and equations; (b) experimentally measured, in monolayer pharynx FaDu cells, the model parameters for three NP formulations (negatively charged polystyrene beads, near-neutral liposomes, and positively charged liposomes, with respective diameter of 20, 110, and 130 nm); and (c) used the models and parameters to simulate NP diffusion in 3-dimensional (3D) systems. We next measured the NP concentration-depth profiles in tumor cell spheroids, an avascular 3D system, and found good agreement between model-simulated and experimental data in spheroids for the negative and neutral NP (>90% predicted data points at three NP concentrations and three treatment times were within the 95% confidence intervals of experimental data). Model performance was inferior for positive liposomes containing a fusogenic lipid. The present study demonstrated the possibility of using in vitro NP-cell biointerface data in monolayer cultures with in silico studies to predict the NP diffusive transport and concentration-time-depth profiles in 3D systems, as functions of NP concentrations and treatment times. Extending this approach to include convective transport may yield a cost-effective means to predict the NP delivery and residence in solid tumors.


Assuntos
Imageamento Tridimensional/métodos , Modelos Moleculares , Nanopartículas , Neoplasias/metabolismo , Esferoides Celulares/metabolismo , Transporte Biológico/fisiologia , Difusão , Previsões , Humanos , Neoplasias/patologia , Esferoides Celulares/patologia , Células Tumorais Cultivadas
19.
Cancer Chemother Pharmacol ; 70(1): 49-56, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22729159

RESUMO

PURPOSE: Suramin, a polysulfonated naphthylurea, inhibits the actions of polypeptide growth factors including acidic and basic fibroblast growth factors (aFGF and bFGF), which confer broad spectrum chemotherapy resistance. We hypothesized that suramin at non-cytotoxic doses in combination with weekly paclitaxel would be well tolerated and demonstrate anti-tumor activity. METHODS: Women with metastatic breast cancer who had been previously treated with a taxane in the adjuvant or metastatic setting were eligible. The primary objective of the phase I was to determine the dose of intravenous (IV) weekly suramin that resulted in plasma concentrations between 10 and 50 umol/l over 8-48 h (or the target range) in combination with IV 80 mg/m(2) of weekly paclitaxel. The primary objective of the phase II trial was to determine the anti-tumor activity of the dosing regimen defined in phase I. Therapy was continued until disease progression or development of unacceptable toxicity. RESULTS: Thirty-one patients were enrolled (9: phase I; 22: phase II). In phase I, no dose-limiting toxicities were observed. Pharmacokinetics during the first cycle showed suramin concentrations within the target range for 21 of 24 weekly treatments (88 %). In phase II, the objective response rate (ORR) was 23 % (95 % CI 8-45 %), the median progression-free survival was 3.4 months (95 % CI 2.1-4.9 months), and the median overall survival was 11.2 months (95 % CI 6.6-16.0 months). CONCLUSIONS: Non-cytotoxic doses of suramin in combination with weekly paclitaxel were well tolerated. The efficacy was below the pre-specified criteria required to justify further investigation.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Neoplasias da Mama/sangue , Neoplasias da Mama/patologia , Intervalo Livre de Doença , Relação Dose-Resposta a Droga , Esquema de Medicação , Fadiga/induzido quimicamente , Feminino , Fator 2 de Crescimento de Fibroblastos/sangue , Humanos , Leucopenia/induzido quimicamente , Pessoa de Meia-Idade , Náusea/induzido quimicamente , Metástase Neoplásica , Paclitaxel/administração & dosagem , Paclitaxel/efeitos adversos , Suramina/administração & dosagem , Suramina/efeitos adversos , Suramina/farmacocinética , Taxoides/administração & dosagem , Resultado do Tratamento
20.
Adv Drug Deliv Rev ; 64(1): 29-39, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21569804

RESUMO

Advances in molecular medicines have led to identification of promising targets on cellular and molecular levels. These targets are located in extracellular and intracellular compartments. The latter include cytosol, nucleus, mitochondrion, Golgi apparatus and endoplasmic reticulum. This report gives an overview on the barriers to delivering nanomedicines to various target sites within a solid tumor, the experimental approaches to overcome such barriers, and the potential utility of nanotechnology.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Nanomedicina/métodos , Nanotecnologia/métodos , Neoplasias/tratamento farmacológico , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...