Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
J Control Release ; 365: 89-100, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37981052

ABSTRACT

A recent development in cancer chemotherapy is to use cytotoxics to induce tumor-specific immune response through immunogenic cell death (ICD). In ICD, calreticulin is translocated from endoplasmic reticulum to cell membrane (ecto-CRT) which serves as the 'eat-me-signal' to antigen-presenting cells. Ecto-CRT measurements, e.g., by ecto-CRT immunostaining plus flow cytometry, can be used to study the pharmacodynamics of ICD in single cells, whereas ICD studies in intact 3-dimensional tissues such as human tumors require different approaches. The present study described a method that used (a) immunostaining with fluorescent antibodies followed by confocal microscopy to obtain the spatial locations of two molecules-of-interest (CRT and a marker protein WGA), and (b) machine-learning (trainable WEKA segmentation) and additional image processing tools to locate the target molecules, remove the interfering signals in the nucleus, cytosol and extracellular space, enable the distinction of the inner and outer edges of the cell membrane and thereby identify the cells with ecto-CRT. This method, when applied to 3-dimensional human bladder cancer cell spheroids, yielded drug-induced ecto-CRT measurements that were qualitatively comparable to the flow cytometry results obtained with single cells disaggregated from spheroids. This new method was applied to study drug-induced ICD in short-term cultures of surgical specimens of human patient bladder tumors.


Subject(s)
Antineoplastic Agents , Urinary Bladder Neoplasms , Humans , Immunogenic Cell Death , Antineoplastic Agents/therapeutic use , Cell Membrane/metabolism , Urinary Bladder Neoplasms/drug therapy , Protein Transport , Cell Line, Tumor
2.
AoB Plants ; 15(5): plad069, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37937046

ABSTRACT

Chlorophyll fluorescence measured at the leaf scale through pulse amplitude modulation (PAM) has provided valuable insight into photosynthesis. At the canopy- and satellite-scale, solar-induced fluorescence (SIF) provides a method to estimate the photosynthetic activity of plants across spatiotemporal scales. However, retrieving SIF signal remotely requires instruments with high spectral resolution, making it difficult and often expensive to measure canopy-level steady-state chlorophyll fluorescence under natural sunlight. Considering this, we built a novel low-cost photodiode system that retrieves far-red chlorophyll fluorescence emission induced by a blue light emitting diode (LED) light source, for 2 h at night, above the canopy. Our objective was to determine if an active remote sensing-based night-time photodiode method could track changes in canopy-scale LED-induced chlorophyll fluorescence (LEDIF) during an imposed drought on a broadleaf evergreen shrub, Polygala myrtifolia. Far-red LEDIF (720-740 nm) was retrieved using low-cost photodiodes (LEDIFphotodiode) and validated against measurements from a hyperspectral spectroradiometer (LEDIFhyperspectral). To link the LEDIF signal with physiological drought response, we tracked stomatal conductance (gsw) using a porometer, two leaf-level vegetation indices-photochemical reflectance index and normalized difference vegetation index-to represent xanthophyll and chlorophyll pigment dynamics, respectively, and a PAM fluorimeter to measure photochemical and non-photochemical dynamics. Our results demonstrate a similar performance between the photodiode and hyperspectral retrievals of LEDIF (R2 = 0.77). Furthermore, LEDIFphotodiode closely tracked drought responses associated with a decrease in photochemical quenching (R2 = 0.69), Fv/Fm (R2 = 0.59) and leaf-level photochemical reflectance index (R2 = 0.59). Therefore, the low-cost LEDIFphotodiode approach has the potential to be a meaningful indicator of photosynthetic activity at spatial scales greater than an individual leaf and over time.

3.
J Control Release ; 361: 717-726, 2023 09.
Article in English | MEDLINE | ID: mdl-37574051

ABSTRACT

Cytoreductive surgery (CRS) has emerged as a survival-extending treatment of peritoneal metastasis (PM); recent advances include using intraperitoneal chemotherapy (IPC) at normothermic or hyperthermic temperatures, or under pressure (CRS + IPC). Clinical CRS + IPC research has established its highly variable efficacy and suggested tumor size, tumor locations and presence of ascites as potential determinants. On the other hand, there is limited knowledge on the effects of pharmaceutical properties on treatment outcomes. The present study investigated the inter-subject variability of paclitaxel binding to proteins in patient ascites because some PM patients show accumulation of ascites and because activity and transport of highly protein-bound drugs such as paclitaxel are affected by protein binding. Ascites samples were collected from 26 patients and investigated for their protein contents using LC/MS/MS proteomics analysis and for the concentrations of total proteins and two major paclitaxel-binding proteins (human serum albumin or HSA and α-1-acid glycoprotein or AAG). The association constants of paclitaxel to HSA and AAG and the extent of protein binding of paclitaxel in patient ascites were studied using equilibrium dialysis. Proteomic analysis of four randomly selected samples revealed 288 proteins, >90% of which are also present in human plasma. Between 72% - 94% of paclitaxel was bound to proteins in patient ascites. The concentrations of HSA and AAG in ascites showed substantial inter-subject variations, ranging from 14.7 - 46.3 mg/mL and 0.13-2.56 mg/mL, respectively. The respective paclitaxel association constants to commercially available HSA and AAG were âˆ¼ 3.5 and âˆ¼ 120 mM. Calculation using these constants and the HSA and AAG concentrations in individual patient ascites indicated that these two proteins accounted for >85% of the total protein-binding of paclitaxel in ascites. The extensive drug binding to ascites proteins, by reducing the pharmacologically active free fraction, may lead to the diminished CRS efficacy in PM patients with ascites. Clinical advances in CRS + IPC have outpaced current knowledge of pharmaceutical properties in this setting. IPC, as a locally acting therapy, is subjected to processes different from those governing systemic treatments. This study, to our knowledge, is the first to illustrate the implications of drug properties in the CRS + IPC efficacy against PM. While drugs are now an integral part of PM patient management, there is limited pharmaceutical research in this treatment setting (e.g., effects of hyperthermia or pressure on drug transport or release from delivery systems, pharmacokinetics, pharmacodynamics). Hence, CRS + IPC of PM represents an area where additional pharmaceutical research can assist further development and optimization.


Subject(s)
Colorectal Neoplasms , Hyperthermia, Induced , Peritoneal Neoplasms , Pharmaceutical Research , Humans , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/secondary , Ascites/drug therapy , Proteomics , Tandem Mass Spectrometry , Paclitaxel/therapeutic use , Pharmaceutical Preparations , Combined Modality Therapy , Antineoplastic Combined Chemotherapy Protocols , Colorectal Neoplasms/drug therapy
4.
AAPS J ; 23(5): 102, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34435271

ABSTRACT

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.


Subject(s)
Drug Development/methods , Drug Interactions , Liver-Specific Organic Anion Transporter 1/metabolism , Models, Biological , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Area Under Curve , Computer Simulation , Hepatocytes/metabolism , Homeostasis , Humans
5.
Pharmaceutics ; 13(7)2021 Jun 30.
Article in English | MEDLINE | ID: mdl-34209265

ABSTRACT

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.

6.
BMC Infect Dis ; 21(1): 75, 2021 Jan 14.
Article in English | MEDLINE | ID: mdl-33446137

ABSTRACT

BACKGROUND: The elderly population in Hong Kong is rapidly growing, and the need for residential care homes (RCHs) is increasing. The risk of being infected with micro-organisms increases among the frail and the vulnerable elderly population as their immunity system begins to deteriorate. Furthermore, the residents in RCHs are at high risk of healthcare-associated infections (HAIs) due to the confined living environments and individual co-morbidities. In relation to this, infection control practice (ICP) is considered a crucial and effective approach in preventing HAIs. This study aimed to observe the daily ICP of healthcare workers in RCH settings. METHODS: An observational study was conducted to observe daily ICP among healthcare workers in private and subsidized RCHs. Each RCH was separated into different units based on the location (common area and bedroom area) and nature of residents for successive days. The ICP episodes were observed until 200 opportunities in each unit. The ICP episodes were recorded by an electronic tool called "eRub," which is an ICP checklist based on international guidelines. RESULTS: The most frequent observed ICP episodes were hand hygiene (n = 1053), the use of gloves (n = 1053) and respiratory protection (n = 1053). The overall compliance of hand hygiene was poor, with only 15% of participants performing this during the "five moments for hand hygiene." Furthermore, the observations showed that 77.9% improperly performed the use of gloves, and 31.8% failed to wear a mask during the care provision for the elderly. However, the results showed that most healthcare workers can wear the mask in a proper way when they should. Generally, the personal care workers were the worst in terms of hand hygiene and use of gloves compared with the other types of healthcare workers. CONCLUSIONS: Despite the fact that the practice of hand hygiene, the use of gloves, and respiratory protection were the important elements of ICP, overall compliance to these elements was still poor. Personal care workers had the most frequent contact with the residents, but they had the worst compliance rate. Hence, continued monitoring and training among healthcare workers is needed, particularly personal care workers, in this healthcare service setting.


Subject(s)
Cross Infection/prevention & control , Guideline Adherence/statistics & numerical data , Health Facilities , Health Personnel , Homes for the Aged , Infection Control/methods , Nursing Homes , Cross Infection/epidemiology , Gloves, Protective , Hand Disinfection/methods , Hong Kong/epidemiology , Humans , Masks , Practice Guidelines as Topic , Respiratory Protective Devices
7.
Am J Infect Control ; 49(6): 746-752, 2021 06.
Article in English | MEDLINE | ID: mdl-33127437

ABSTRACT

BACKGROUND: The risk of hand recontamination is often neglected after using hand washing facilities, which can increase the spread of pathogens. The study aimed to evaluate the hygienic condition of posthandwashing facilities in public washrooms at different timeslots, particularly those near food courts and restaurants located in shopping malls. METHODS: This observational study was conducted in 12 public washrooms that ranged from low-end, middle-end, to high-end category on 3 different timeslots including baseline, T1 (immediate postcleaning) and T2 (1-hour postcleaning, with counting the footfall). Hand-touch surfaces with a high risk of recontamination after handwashing, which included paper tower dispensers, air drying outlets, and exit door handles, were evaluated by the surface adenosine triphosphate (ATP) bioluminescence method (ATP-value). ATP-values <500 relative light units (RLUs) were considered a good hygiene. Cleaning schedules and footfalls of each sampled washroom were obtained by direct observations. RESULTS: The overall mean ATP value of washroom was 785 RLU (N = 108); the ATP values of female and male washrooms at T2 were 203 and 3,718 RLUs, respectively. The highest ATP value was found on the exit door handles of male washrooms (range = 13-26,695 RLUs, mean = 3,229 RLU). Regarding passed/failed hygiene conditions, there were significant differences in the proportion of exit door handles between genders (P = .018) and timeslots (P = .007) as well as that of paper towel button/screw between timeslots (P= .025). CONCLUSION: Attention should be paid at the exit door handles of male washrooms, where are high risks of cross and re-contamination.


Subject(s)
Hand Disinfection , Hygiene , Adenosine Triphosphate , Colony Count, Microbial , Female , Humans , Luminescent Measurements , Male
8.
Pharmacol Rev ; 71(2): 157-169, 2019 04.
Article in English | MEDLINE | ID: mdl-30846487

ABSTRACT

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.


Subject(s)
Drug Delivery Systems , Drug Development/methods , Nanostructures/administration & dosage , Animals , Humans , Models, Theoretical , Nanomedicine/methods , Neoplasms/drug therapy , Pharmacology , Systems Biology/methods , Time Factors , Tumor Microenvironment
9.
AAPS J ; 21(2): 24, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30710324

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Approval , Drug Carriers/pharmacokinetics , Drugs, Generic/pharmacokinetics , United States Food and Drug Administration/standards , Antineoplastic Agents/pharmacokinetics , Humans , Nanoparticles , Neoplasms/drug therapy , Particle Size , Therapeutic Equivalency , Tissue Distribution , United States , United States Food and Drug Administration/legislation & jurisprudence
10.
Clin Pharmacol Ther ; 104(1): 27-30, 2018 07.
Article in English | MEDLINE | ID: mdl-29603191

ABSTRACT

On October 2nd and 3rd , 2017, the US Food and Drug Administration (FDA) hosted a public workshop titled "Leveraging Quantitative Methods and Modeling to Modernize Generic Drug Development and Review."1 This report summarizes Session 2 of the public workshop: "Model Informed Drug Development and Review for Generic Products." The session focused on the application of quantitative methods and modeling in modernizing the generic drug development and review.


Subject(s)
Drug Approval , Drug Development , Drugs, Generic , United States Food and Drug Administration , Education , Humans , Therapeutic Equivalency , United States
11.
J Control Release ; 270: 101-113, 2018 01 28.
Article in English | MEDLINE | ID: mdl-29203416

ABSTRACT

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.


Subject(s)
Heparin/administration & dosage , RNA, Small Interfering/administration & dosage , Suramin/administration & dosage , Survivin/genetics , Biological Availability , Gene Silencing , HT29 Cells , Humans , Liposomes , Transfection
12.
J Control Release ; 268: 147-158, 2017 Dec 28.
Article in English | MEDLINE | ID: mdl-29054369

ABSTRACT

PURPOSE: Exosomes are small membrane vesicles (30-100nm in diameter) secreted by cells into extracellular space. The present study evaluated the effect of chemotherapeutic agents on exosome production and/or release, and quantified the contribution of exosomes to intercellular drug transfer and pharmacodynamics. METHODS: Human cancer cells (breast MCF7, breast-to-lung metastatic LM2, ovarian A2780 and OVCAR4) were treated with paclitaxel (PTX, 2-1000nM) or doxorubicin (DOX, 20-1000nM) for 24-48h. Exosomes were isolated from the culture medium of drug-treated donor cells (Donor cells) using ultra-centrifugation, and analyzed for acetylcholinesterase activity, total proteins, drug concentrations, and biological effects (cytotoxicity and anti-migration) on drug-naïve recipient cells (Recipient cells). These results were used to develop computational predictive quantitative pharmacology models. RESULTS: Cells in exponential growth phase released ~220 exosomes/cell in culture medium. PTX and DOX significantly promoted exosome production and/or release in a dose- and time-dependent manner, with greater effects in ovarian cancer cells than in breast cancer cells. Exosomes isolated from Donor cells contained appreciable drug levels (2-7pmole/106 cells after 24h treatment with 100-1000nM PTX), and caused cytotoxicity and inhibited migration of Recipient cells. Quantitative pharmacology models that integrated cellular PTX pharmacokinetics with PTX pharmacodynamics successfully predicted effects of exosomes on intercellular drug transfer, cytotoxicity of PTX on Donor cells and cytotoxicity of PTX-containing exosomes on Recipient cells. Additional model simulations indicate that within clinically achievable PTX concentrations, the contribution of exosomes to active drug efflux increased with drug concentration and exceeded the p-glycoprotein efflux when the latter was saturated. CONCLUSIONS: Our results indicate (a) chemotherapeutic agents stimulate exosome production or release, and (b) exosome is a mechanism of intercellular drug transfer that contributes to pharmacodynamics of neighboring cells.


Subject(s)
Antineoplastic Agents/pharmacology , Exosomes/metabolism , Models, Biological , Biological Transport , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/pharmacology , Humans , Paclitaxel/pharmacology , Pharmacology
13.
Curr Cancer Drug Targets ; 17(8): 735-755, 2017.
Article in English | MEDLINE | ID: mdl-28359247

ABSTRACT

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.


Subject(s)
Drug Interactions , Pharmaceutical Preparations/administration & dosage , Cell Line, Tumor , Computer Simulation , Dose-Response Relationship, Drug , Humans
14.
Adv Drug Deliv Rev ; 97: 280-301, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26686425

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/administration & dosage , Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Drug Delivery Systems , Extracellular Matrix/metabolism , Humans , Nanoparticles/administration & dosage , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Neoplasms/immunology , Tumor Microenvironment
15.
J Control Release ; 216: 103-10, 2015 Oct 28.
Article in English | MEDLINE | ID: mdl-26272765

ABSTRACT

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.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacology , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Pancreatic Neoplasms/therapy , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacology , Animals , Cell Line, Tumor , Drug Delivery Systems , Drug Resistance, Neoplasm , Female , Gene Silencing/drug effects , Genetic Therapy/methods , Humans , Inhibitor of Apoptosis Proteins/drug effects , Injections, Intravenous , Lipids/chemistry , Mice , Mice, Nude , Pancreatic Neoplasms/drug therapy , Peritoneal Neoplasms/drug therapy , RNA, Neoplasm/metabolism , Survivin , Transfection , Xenograft Model Antitumor Assays
16.
AAPS J ; 17(6): 1475-82, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26286676

ABSTRACT

RNAi therapeutics provide an opportunity to correct faulty genes, and several RNAi have entered clinical evaluation. The existing quantification methods typically use radioactivity- or fluorescence-labeled RNAi, require large blood volumes, and/or have a limited dynamic detection range. We established a quantitative reverse transcriptase real-time polymerase chain reaction (RT-qPCR) assay to measure RNAi; the model analyte was survivin siRNA (siSurvivin). A second siRNA was used as the internal standard. The three major steps were (a) extraction of the two siRNAs from blood or water, (b) synthesis of their cDNA by poly-A extension, and (c) qPCR of cDNA. Standard curves were established. Utility of the assay was demonstrated in a pharmacokinetic study where all 12 samples for the blood concentration-time profile were obtained from a single mouse given an intravenous dose of 1 nmole siSurvivin (prepared as lipoplex with pegylated cationic liposomes). The RT-qPCR assay was sensitive (lower detection limit of 100 fM) and had a 5 × 107-fold dynamic range and low sample volume requirement (10 µL). The 16-point standard curves constructed using whole blood samples were linear (R (2) > 0.98). The intraday and interday variations for the slopes were ≤6%, although the variations for accuracy and precision at individual concentrations were substantially higher (58-145%). Standard curves prepared with water in place of blood showed similar results (<6% difference), indicating water may be used when blood is not available. The current RT-qPCR assay enabled the measurement of nonlabeled siRNA in small volume of blood samples.


Subject(s)
Blood Volume/physiology , Inhibitor of Apoptosis Proteins/blood , Inhibitor of Apoptosis Proteins/pharmacokinetics , RNA, Small Interfering/blood , RNA, Small Interfering/pharmacokinetics , Real-Time Polymerase Chain Reaction/standards , Repressor Proteins/blood , Repressor Proteins/pharmacokinetics , Animals , Anticoagulants/blood , Anticoagulants/pharmacokinetics , Dose-Response Relationship, Drug , Female , Limit of Detection , Mice , Mice, Nude , Real-Time Polymerase Chain Reaction/methods , Survivin
17.
AAPS J ; 17(5): 1065-79, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26089090

ABSTRACT

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.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Paclitaxel/administration & dosage , Peritoneal Neoplasms/drug therapy , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/therapeutic use , Drug Carriers/chemistry , Drug Design , Humans , Injections, Intraperitoneal , Microspheres , Paclitaxel/pharmacokinetics , Paclitaxel/therapeutic use , Peritoneal Neoplasms/pathology , Polyethylene Glycols/chemistry , Polymers/chemistry
18.
J Chem Ecol ; 41(6): 513-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25994224

ABSTRACT

Identifying specific plant secondary metabolites that influence feeding behavior can be challenging, but a solid understanding of animal preferences can guide efforts. Common brushtail possums (Trichosurus vulpecula) predominantly eat Eucalyptus species belonging to the subgenus Symphyomyrtus, and avoid eating those belonging to the Monocalyptus subgenus (also called subgenus Eucalyptus). Using an unbiased (1)H NMR metabolomics approach, a previous study identified unsubstituted B ring flavanones in most species of monocalypts examined, whereas these compounds were absent from symphyomyrtles. We hypothesised that unsubstituted B ring flavanones act as feeding deterrents for common brushtail possums. In the current study, we tested this hypothesis by comparing how much possums ate of a basal diet, with diets containing one of four structurally related compounds; pinocembrin, flavanone (unsubstituted B ring flavanones), chrysin (the flavone analogue of pinocembrin), and naringenin (a flavanone with B ring substitution). We found that pinocembrin and flavanone deterred feeding relative to the basal diet, but that chrysin and naringenin did not at equivalent concentrations. Thus, unsubstituted B-ring flavanones may explain why brushtail possums avoid eating monocalypt species. Furthermore, small differences in the structure of secondary compounds can have a large impact on antifeedant properties. These results demonstrate that metabolomics can be a valuable tool for ecologists seeking to understand herbivore feeding preferences.


Subject(s)
Eucalyptus/chemistry , Flavanones/chemistry , Herbivory , Metabolome , Plant Leaves/chemistry , Trichosurus/physiology , Animals , Diet , Male , Metabolomics
19.
J Urol ; 194(1): 230-7, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25681288

ABSTRACT

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.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Carcinoma, Transitional Cell/drug therapy , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Mitomycin/therapeutic use , RNA Interference , RNA, Small Interfering/pharmacology , RNA, Small Interfering/therapeutic use , Animals , Carcinoma, Transitional Cell/genetics , Female , Heterografts , Humans , Injections, Intravenous , Mice , Mice, Nude , Survivin , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
20.
AAPS J ; 17(1): 268-76, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25425294

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Suramin/pharmacology , Telomerase/antagonists & inhibitors , Telomere Shortening/drug effects , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/drug therapy , Neoplasms/pathology , Suramin/administration & dosage , Telomere/drug effects , Telomere/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...