Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nat Rev Drug Discov ; 22(5): 410-427, 2023 05.
Article in English | MEDLINE | ID: mdl-36810917

ABSTRACT

Over the past 3 years, the first bivalent protein degraders intentionally designed for targeted protein degradation (TPD) have advanced to clinical trials, with an initial focus on established targets. Most of these clinical candidates are designed for oral administration, and many discovery efforts appear to be similarly focused. As we look towards the future, we propose that an oral-centric discovery paradigm will overly constrain the chemical designs that are considered and limit the potential to drug novel targets. In this Perspective, we summarize the current state of the bivalent degrader modality and propose three categories of degrader designs, based on their likely route of administration and requirement for drug delivery technologies. We then describe a vision for how parenteral drug delivery, implemented early in research and supported by pharmacokinetic-pharmacodynamic modelling, can enable exploration of a broader drug design space, expand the scope of accessible targets and deliver on the promise of protein degraders as a therapeutic modality.


Subject(s)
Drug Design , Proteins , Humans , Proteins/therapeutic use , Proteins/metabolism , Proteolysis , Drug Delivery Systems
2.
Chem Soc Rev ; 51(9): 3477-3486, 2022 May 10.
Article in English | MEDLINE | ID: mdl-35438107

ABSTRACT

Targeted protein degradation has emerged from the chemical biology toolbox as one of the most exciting areas for novel therapeutic development across the pharmaceutical industry. The ability to induce the degradation, and not just inhibition, of target proteins of interest (POIs) with high potency and selectivity is a particularly attractive property for a protein degrader therapeutic. However, the physicochemical properties and mechanism of action for protein degraders can lead to unique pharmacokinetic (PK) and pharmacodynamic (PD) properties relative to traditional small molecule drugs, requiring a shift in perspective for translational pharmacology. In this review, we provide practical insights for building the PK-PD understanding of protein degraders in the context of translational drug development through the use of quantitative mathematical frameworks and standard experimental assays. Published datasets describing protein degrader pharmacology are used to illustrate the applicability of these insights. The learnings are consolidated into a translational PK-PD roadmap for targeted protein degradation that can enable a systematic, rational design workflow for protein degrader therapeutics.


Subject(s)
Models, Biological , Proteolysis
3.
Front Psychol ; 12: 624254, 2021.
Article in English | MEDLINE | ID: mdl-33658964

ABSTRACT

INTRODUCTION: Approaches to improve heart rate variability and reduce stress such as breathing retraining are more frequently being integrated into psychotherapy but little research on their effectiveness has been done to date. Specifically, no studies to date have directly compared using a breathing pacer at 6 breaths per minute with compassion focused soothing rhythm breathing. CURRENT STUDY: In this randomized controlled experiment, 6 breaths per minute breathing using a pacer was compared with compassion focused soothing rhythm breathing, with a nature video being used as a control group condition. METHODS: Heart rate variability (HRV) measures were assessed via electrocardiogram (ECG) and respiration belt, and an automated blood pressure machine was used to measure systolic diastolic blood pressure, and heart rate (HR). A total of 96 participants were randomized into the three conditions. Following a 5-min baseline, participants engaged in either 6 breath per minute breathing, soothing rhythm breathing, or watched a nature video for 10 min. To induce a stressful state, participants then wrote for 5 min about a time they felt intensely self-critical. Participants then wrote for 5 min about a time they felt self-compassionate, and the experiment ended with a 10-min recovery period. RESULTS: Conditions did not significantly differ at baseline. Overall, HRV, as measured by standard deviation of NN intervals (SDNN), low frequency HRV (LF HRV), and LF/HF ratio, increased during the intervention period, decreased during self-critical writing, and then returned to baseline levels during the recovery period. High frequency HRV (HF HRV) was not impacted by any of the interventions. The participants in the 6 breath per minute pacer condition were unable to consistently breathe at that rate and averaged about 12 breaths per minute. Time by Condition analyses revealed that both the 6 breaths per minute pacer and soothing breathing rhythm conditions lead to significantly higher SDNN than the nature video condition during breathing practice but there were no significant differences between conditions in response to the self-critical and self-compassionate writing or recovery periods. The 6 breath per minute pacer condition demonstrated a higher LF HRV and LF/HF ratio than the soothing rhythm breathing condition, and both intervention conditions had a higher LF HRV and LF/HF ratio than the nature video. CONCLUSIONS: Although the 6 breath per minute pacer condition participants were not able to breath consistently at the low pace, both the participants attempting to breathe at 6 breaths per minute as well as those in the soothing rhythm breathing condition effectively increased HR variability as measured by SDNN, and attempting to breathe at 6 breaths per minute led to the highest LF HRV and LF/HF ratio. Both breathing approaches impacted HRV more than watching a relaxing nature video and can potentially be used as key adjuncts in psychotherapy to aid in regulating physiological functioning, although it appears that consistent breathing practice would be needed.

4.
J Pharmacokinet Pharmacodyn ; 48(1): 149-163, 2021 02.
Article in English | MEDLINE | ID: mdl-33090299

ABSTRACT

Bispecific protein degraders (BPDs) engage the ubiquitin-proteasome system (UPS) to catalytically degrade intracellular proteins through the formation of ternary complexes with the target protein and E3 ubiquitin ligases. Here, we describe the development of a mechanistic modeling framework for BPDs that includes the reaction network governing ternary complex formation and degradation via the UPS. A critical element of the model framework is a multi-step process that results in a time delay between ternary complex formation and protein degradation, thereby balancing ternary complex stability against UPS degradation rates akin to the kinetic proofreading concept that has been proposed to explain the accuracy and specificity of biological processes including protein translation and T cell receptor signal transduction. Kinetic proofreading likely plays a central role in the cell's ability to regulate substrate recognition and degradation by the UPS, and the model presented here applies this concept in the context of a quantitative pharmacokinetic (PK)-pharmacodynamic (PD) framework to inform the design of potent and selective BPDs.


Subject(s)
Drug Design , Proteasome Endopeptidase Complex/drug effects , Proteolysis/drug effects , Ubiquitin/agonists , Computer Simulation , Humans , Models, Biological , Proteasome Endopeptidase Complex/metabolism , Protein Binding/drug effects , Signal Transduction/drug effects , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
5.
AAPS J ; 22(2): 53, 2020 03 02.
Article in English | MEDLINE | ID: mdl-32124093

ABSTRACT

Static in vitro cell culture studies cannot capture the dynamic concentration profiles of drugs, nutrients, and other factors that cells experience in physiological systems. This limits the confidence in the translational relevance of in vitro experiments and increases the reliance on empirical testing of exposure-response relationships and dose optimization in animal models during preclinical drug development, introducing additional challenges owing to species-specific differences in drug pharmacokinetics (PK) and pharmacodynamics (PD). Here, we describe the development of a microfluidic cell culture device that enables perfusion of cells under 2D or 3D culture conditions with temporally programmable concentration profiles. Proof-of-concept studies using doxorubicin and gemcitabine demonstrated the ability of the microfluidic PK-PD device to examine dose- and time-dependent effects of doxorubicin as well as schedule-dependent effects of doxorubicin and gemcitabine combination therapy on cell viability using both step-wise drug concentration profiles and species-specific (i.e., mouse, human) drug PK profiles. The results demonstrate the importance of including physiologically relevant dynamic drug exposure profiles during in vitro drug testing to more accurately mimic in vivo drug effects, thereby improving translatability across nonclinical studies and reducing the reliance on animal models during drug development.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Breast Neoplasms/drug therapy , Deoxycytidine/analogs & derivatives , Doxorubicin/pharmacology , Doxorubicin/pharmacokinetics , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques , Models, Biological , Antineoplastic Combined Chemotherapy Protocols/chemistry , Breast Neoplasms/pathology , Cell Survival/drug effects , Deoxycytidine/chemistry , Deoxycytidine/pharmacokinetics , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Doxorubicin/chemistry , Humans , MCF-7 Cells , Perfusion , Proof of Concept Study , Tissue Culture Techniques , Gemcitabine
6.
PLoS One ; 14(5): e0217276, 2019.
Article in English | MEDLINE | ID: mdl-31112568

ABSTRACT

Cancer cells harness immune checkpoints such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1) and indoleamine 2,3-dioxygenase 1 (IDO1) to evade immune control. Checkpoint inhibitors have demonstrated durable anti-tumor efficacy in human and preclinical models. Liver toxicity is one of the common immune-related adverse events associated with checkpoint inhibitors (CPIs) and its frequency and severity often increase significantly during CPI combination therapies. We aim to develop a mouse model to elucidate the immune mechanisms of CPI-associated liver toxicity. Co-administration of CTLA-4 blocking antibody, 9D9, and/or an IDO1 inhibitor, epacadostat in wild-type and PD-1-/- mice (to simulate the effect of PD1 blockade) synergistically induced liver injury and immune cell infiltration. Infiltrated cells were primarily composed of CD8+ T cells and positively associated with hepatocyte necrosis. Strikingly, sites of hepatocyte necrosis were frequently surrounded by clusters of mononuclear immune cells. CPI treatments resulted in increased expression of genes associated with hepatocyte cell death, leukocyte migration and T cell activation in the liver. In conclusion, blockade of immune checkpoints PD-1, CTLA-4, and IDO1 act synergistically to enhance T cell infiltration and activity in the liver, leading to hepatocyte death.


Subject(s)
CTLA-4 Antigen/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Liver/immunology , Liver/injuries , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Disease Models, Animal , Female , Hepatocytes/pathology , Humans , Ipilimumab/administration & dosage , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nivolumab/administration & dosage , Oximes/administration & dosage , Programmed Cell Death 1 Receptor/deficiency , Programmed Cell Death 1 Receptor/genetics , Sulfonamides/administration & dosage
7.
J Pharm Sci ; 108(9): 3124-3129, 2019 09.
Article in English | MEDLINE | ID: mdl-31136767

ABSTRACT

Microdialysis is a technique that utilizes a semipermeable membrane to sample analytes present within tissue interstitial fluid. Analyte-specific calibration is required for quantitative microdialysis, but these calibration methods are tedious, require significant technical skill, and often cannot be performed jointly with the experimental measurements. Here, we describe a method using retrodialysis with stable-isotope-labeled analytes that enables simultaneous calibration and quantification for in vivo tumor microdialysis. Isotope-labeled amino acids relevant to immuno-metabolism in the tumor microenvironment (tryptophan, kynurenine, glutamine, and glutamate) were added to the microdialysis perfusate, and microdialysis probes were inserted in subcutaneous CT26 and MC38 tumors in mice. The levels of both the endogenous and isotope-labeled amino acids in the perfusate outlet were quantified using LC-MS/MS. Plasma and tumor tissue samples were also collected from the same mice and amino acid levels quantified using LC-MS/MS. Amino acids which showed statistically significant differences between the CT26-bearing and MC38-bearing mice in tumor lysate (tryptophan, kynurenine, and glutamine) and plasma (glutamate) were not the same as those identified as significantly different in tumor interstitial fluid (kynurenine and glutamate), underscoring how microdialysis can provide unique and complementary insights into tumor and immune metabolism within the tumor microenvironment.


Subject(s)
Amino Acids/analysis , Antineoplastic Agents/pharmacology , Drug Monitoring/methods , Microdialysis/standards , Tumor Microenvironment/drug effects , Amino Acids/metabolism , Animals , Antineoplastic Agents/therapeutic use , Calibration , Cell Line, Tumor/transplantation , Chromatography, High Pressure Liquid/methods , Dialysis Solutions/analysis , Dialysis Solutions/chemistry , Disease Models, Animal , Female , Humans , Isotope Labeling , Metabolic Networks and Pathways/drug effects , Mice , Microdialysis/methods , Neoplasms/drug therapy , Neoplasms/metabolism , Tandem Mass Spectrometry/methods
8.
Mol Cancer Ther ; 17(12): 2530-2542, 2018 12.
Article in English | MEDLINE | ID: mdl-30232146

ABSTRACT

Tumors use indoleamine 2,3-dioxygenase-1 (IDO1) as a major mechanism to induce an immunosuppressive microenvironment. IDO1 expression is upregulated in many cancers and considered to be a resistance mechanism to immune checkpoint therapies. IDO1 is induced in response to inflammatory stimuli such as IFNγ and promotes immune tolerance by depleting tryptophan and producing tryptophan catabolites, including kynurenine, in the tumor microenvironment. This leads to effector T-cell anergy and enhanced Treg function through upregulation of FoxP3. As a nexus for the induction of key immunosuppressive mechanisms, IDO1 represents an important immunotherapeutic target in oncology. Here, we report the identification and characterization of the novel selective, orally bioavailable IDO1 inhibitor EOS200271/PF-06840003. It reversed IDO1-induced T-cell anergy in vitro In mice carrying syngeneic tumor grafts, PF-06840003 reduced intratumoral kynurenine levels by over 80% and inhibited tumor growth both in monotherapy and, with an increased efficacy, in combination with antibodies blocking the immune checkpoint ligand PD-L1. We demonstrate that anti-PD-L1 therapy results in increased IDO1 metabolic activity thereby providing additional mechanistic rationale for combining PD-(L)1 blockade with IDO1 inhibition in cancer immunotherapies. Supported by these preclinical data and favorable predicted human pharmacokinetic properties of PF-06840003, a phase I open-label, multicenter clinical study (NCT02764151) has been initiated.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Biocatalysis , Enzyme Inhibitors/pharmacology , Immunotherapy , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoles/pharmacology , Succinimides/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , B7-H1 Antigen/metabolism , CTLA-4 Antigen/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon-gamma/metabolism , Kynurenine/blood , Lymphocytes, Tumor-Infiltrating/drug effects , Mice, Inbred BALB C , Mice, Inbred C57BL , Stereoisomerism , Substrate Specificity/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/drug effects
9.
Gene Regul Syst Bio ; 11: 1177625017710941, 2017.
Article in English | MEDLINE | ID: mdl-28804243

ABSTRACT

Reduction in low-density lipoprotein cholesterol (LDL-C) is associated with decreased risk for cardiovascular disease. Alirocumab, an antibody to proprotein convertase subtilisin/kexin type 9 (PCSK9), significantly reduces LDL-C. Here, we report development of a quantitative systems pharmacology (QSP) model integrating peripheral and liver cholesterol metabolism, as well as PCSK9 function, to examine the mechanisms of action of alirocumab and other lipid-lowering therapies, including statins. The model predicts changes in LDL-C and other lipids that are consistent with effects observed in clinical trials of single or combined treatments of alirocumab and other treatments. An exploratory model to examine the effects of lipid levels on plaque dynamics was also developed. The QSP platform, on further development and qualification, may support dose optimization and clinical trial design for PCSK9 inhibitors and lipid-modulating drugs. It may also improve our understanding of factors affecting therapeutic responses in different phenotypes of dyslipidemia and cardiovascular disease.

10.
Front Pharmacol ; 5: 232, 2014.
Article in English | MEDLINE | ID: mdl-25374542

ABSTRACT

Multiple software programs are available for designing and running large scale system-level pharmacology models used in the drug development process. Depending on the problem, scientists may be forced to use several modeling tools that could increase model development time, IT costs and so on. Therefore, it is desirable to have a single platform that allows setting up and running large-scale simulations for the models that have been developed with different modeling tools. We developed a workflow and a software platform in which a model file is compiled into a self-contained executable that is no longer dependent on the software that was used to create the model. At the same time the full model specifics is preserved by presenting all model parameters as input parameters for the executable. This platform was implemented as a model agnostic, therapeutic area agnostic and web-based application with a database back-end that can be used to configure, manage and execute large-scale simulations for multiple models by multiple users. The user interface is designed to be easily configurable to reflect the specifics of the model and the user's particular needs and the back-end database has been implemented to store and manage all aspects of the systems, such as Models, Virtual Patients, User Interface Settings, and Results. The platform can be adapted and deployed on an existing cluster or cloud computing environment. Its use was demonstrated with a metabolic disease systems pharmacology model that simulates the effects of two antidiabetic drugs, metformin and fasiglifam, in type 2 diabetes mellitus patients.

11.
Mol Pharm ; 11(11): 3965-73, 2014 Nov 03.
Article in English | MEDLINE | ID: mdl-24779727

ABSTRACT

Engineered antibody fragments offer faster delivery with retained tumor specificity and rapid clearance from nontumor tissues. Here, we demonstrate that positron emission tomography (PET) based detection of prostate specific membrane antigen (PSMA) in prostatic tumor models using engineered bivalent antibodies built on single chain fragments (scFv) derived from the intact antibody, huJ591, offers similar tumor delineating properties but with the advantage of rapid targeting and imaging. (89)Zr-radiolabeled huJ591 scFv (dimeric scFv-CH3; (89)Zr-Mb) and cysteine diabodies (dimeric scFv; (89)Zr-Cys-Db) demonstrated internalization and similar Kds (∼2 nM) compared to (89)Zr-huJ591 in PSMA(+) cells. Tissue distribution assays established the specificities of both (89)Zr-Mb and (89)Zr-Cys-Db for PSMA(+) xenografts (6.2 ± 2.5% ID/g and 10.2 ± 3.4% ID/g at 12 h p.i. respectively), while minimal accumulation in PSMA(-) tumors was observed. From the PET images, (89)Zr-Mb and (89)Zr-Cys-Db exhibited faster blood clearance than the parent huJ591 while tumor-to-muscle ratios for all probes show comparable values across all time points. Ex vivo autoradiography and histology assessed the distribution of the probes within the tumor. Imaging PSMA-expressing prostate tumors with smaller antibody fragments offers rapid tumor accumulation and accelerated clearance; hence, shortened wait periods between tracer administration and high-contrast tumor imaging and lower dose-related toxicity are potentially realized.


Subject(s)
Antibodies, Monoclonal , Antigens, Surface/immunology , Glutamate Carboxypeptidase II/immunology , Molecular Imaging/methods , Prostatic Neoplasms/diagnostic imaging , Radiopharmaceuticals , Single-Chain Antibodies , Zirconium , Animals , Antibodies, Monoclonal/pharmacokinetics , Humans , Immunoglobulin Fragments , Iodine Radioisotopes/pharmacokinetics , Male , Mice , Positron-Emission Tomography/methods , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Radiopharmaceuticals/pharmacokinetics , Single-Chain Antibodies/pharmacokinetics , Tissue Distribution , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Zirconium/pharmacokinetics
12.
J Biomed Opt ; 18(10): 101304, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23752742

ABSTRACT

Antibody fragments including diabodies have more desirable pharmacokinetic characteristics than whole antibodies. An activatable optical imaging probe based on a cys-diabody targeting prostate-specific membrane antigen conjugated with the near-infrared fluorophore, indocyanine green (ICG), was designed such that it can only be activated when bound to the tumor, leading to high signal-to-background ratios. We employed short polyethylene glycol (PEG) linkers between the ICG and the reactive functional group (Sulfo-OSu group), resulting in covalent conjugation of ICG to the cys-diabody, which led to lower dissociation of ICG from cys-diabody early after injection, reducing hepatic uptake. However, unexpectedly, high and long-term fluorescence was observed in the kidneys, liver, and blood pool more than 1 h after injection of the cys-diabody PEG-ICG conjugate. A biodistribution study using I125-labeled cys-diabody-ICG showed immediate uptake in the kidneys followed by a rapid decrease, while gastric activity increased due to released radioiodine during rapid cys-diabody-ICG catabolism in the kidneys. To avoid this catabolic pathway, it would be preferable to use antibody fragments large enough not to be filtered through glomerulus or to conjugate the fragments with fluorescent dyes that are readily excreted into urine when cleaved from the cys-diabody to achieve high tumor-specific detection.


Subject(s)
Fluorescent Dyes/chemistry , Indocyanine Green/chemistry , Microscopy, Fluorescence/methods , Molecular Imaging/methods , Animals , Antibodies/chemistry , Antibodies/metabolism , Antigens, Surface/immunology , Antigens, Surface/metabolism , Female , Fluorescent Dyes/pharmacokinetics , Glutamate Carboxypeptidase II/immunology , Glutamate Carboxypeptidase II/metabolism , Indocyanine Green/pharmacokinetics , Kinetics , Mice , Mice, Nude , Polyethylene Glycols , Tissue Distribution
13.
Bioconjug Chem ; 19(9): 1927-37, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18720981

ABSTRACT

Increasing interest in the use of radiolabeled antibodies for cancer imaging and therapy drives the need for more efficient production of the antibody conjugates. Here, we illustrate a method for rapid and efficient production of radiolabeled antibody conjugates using vacuum diafiltration guided by mathematical modeling. We apply this technique to the production of 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA)-conjugated antibodies at the milligram and gram production scale and achieve radiolabeling efficiencies >95% using In-111. Using vacuum diafiltration, antibody-chelate conjugation and purification can be accomplished within the same vessel, and the entire process can be completed in <24 h. Vacuum diafiltration also offers safer and gentler processing conditions by eliminating the need to keep the retentate vessel under positive pressure through applied gas pressure or shear-inducing restriction points in the retentate flow path. Experimental data and mathematical model calculations suggest there exists a weak binding affinity (approximately 10(4)M(-1)) between the charged chelate molecules (e.g., DOTA) and the antibodies that slows the removal of excess chelate during purification. By analyzing the radiolabeling efficiency as a function of the number of diavolumes, we demonstrate the importance of balancing the removal of free chelate with the introduction of metal contaminants from the diafiltration buffer and also illustrate how to optimize radiolabeling of antibody conjugates under a variety of operating conditions. This methodology is applicable to the production of antibody conjugates in general.


Subject(s)
Antibodies, Monoclonal , Filtration/methods , Heterocyclic Compounds, 1-Ring/chemistry , Immunoconjugates/chemistry , Isotope Labeling/methods , Radioimmunotherapy/methods , Radiopharmaceuticals/chemistry , Animals , Chelating Agents/chemistry , Humans , Models, Theoretical , Vacuum
14.
Biotechnol Bioeng ; 99(4): 975-85, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-17929316

ABSTRACT

This study addresses issues of relevance for siRNA nanoparticle delivery by investigating the functional impact of tumor-specific targeting and dosing schedule. The investigations are performed using an experimental system involving a syngeneic mouse cancer model and a theoretical system based on our previously described mathematical model of siRNA delivery and function. A/J mice bearing subcutaneous Neuro2A tumors approximately 100 mm(3) in size were treated by intravenous injection with siRNA-containing nanoparticles formed with cyclodextrin-containing polycations (CDP). Three consecutive daily doses of transferrin (Tf)-targeted nanoparticles carrying 2.5 mg/kg of two different siRNA sequences targeting ribonucleotide reductase subunit M2 (RRM2) slowed tumor growth, whereas non-targeted nanoparticles were significantly less effective when given at the same dose. Furthermore, administration of the three doses on consecutive days or every 3 days did not lead to statistically significant differences in tumor growth delay. Mathematical model calculations of siRNA-mediated target protein knockdown and tumor growth inhibition are used to elucidate possible mechanisms to explain the observed effects and to provide guidelines for designing more effective siRNA-based treatment regimens regardless of delivery methodology and tumor type.


Subject(s)
Gene Targeting/methods , Genetic Therapy/methods , Nanoparticles/administration & dosage , Neuroblastoma/genetics , Neuroblastoma/therapy , RNA, Small Interfering/administration & dosage , Transferrin/genetics , Animals , Cell Proliferation , Dose-Response Relationship, Drug , Female , Injections, Intravenous , Mice , Neuroblastoma/pathology
15.
Proc Natl Acad Sci U S A ; 104(39): 15549-54, 2007 Sep 25.
Article in English | MEDLINE | ID: mdl-17875985

ABSTRACT

Targeted delivery represents a promising approach for the development of safer and more effective therapeutics for oncology applications. Although macromolecules accumulate nonspecifically in tumors through the enhanced permeability and retention (EPR) effect, previous studies using nanoparticles to deliver chemotherapeutics or siRNA demonstrated that attachment of cell-specific targeting ligands to the surface of nanoparticles leads to enhanced potency relative to nontargeted formulations. Here, we use positron emission tomography (PET) and bioluminescent imaging to quantify the in vivo biodistribution and function of nanoparticles formed with cyclodextrin-containing polycations and siRNA. Conjugation of 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid to the 5' end of the siRNA molecules allows labeling with (64)Cu for PET imaging. Bioluminescent imaging of mice bearing luciferase-expressing Neuro2A s.c. tumors before and after PET imaging enables correlation of functional efficacy with biodistribution data. Although both nontargeted and transferrin-targeted siRNA nanoparticles exhibit similar biodistribution and tumor localization by PET, transferrin-targeted siRNA nanoparticles reduce tumor luciferase activity by approximately 50% relative to nontargeted siRNA nanoparticles 1 d after injection. Compartmental modeling is used to show that the primary advantage of targeted nanoparticles is associated with processes involved in cellular uptake in tumor cells rather than overall tumor localization. Optimization of internalization may therefore be key for the development of effective nanoparticle-based targeted therapeutics.


Subject(s)
Cyclodextrins/administration & dosage , Drug Delivery Systems , Nanoparticles/chemistry , Neoplasms/therapy , RNA, Small Interfering/metabolism , Animals , Chemistry, Pharmaceutical/methods , Cyclodextrins/chemistry , Drug Carriers , Heterocyclic Compounds, 1-Ring/chemistry , Humans , Mice , Mice, SCID , Neoplasm Transplantation , Positron-Emission Tomography/methods , Technology, Pharmaceutical/methods
16.
Clin Cancer Res ; 13(7): 2207-15, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17404105

ABSTRACT

PURPOSE: Ribonucleotide reductase (RR) is a therapeutic target for DNA replication-dependent diseases such as cancer. Here, a potent small interfering RNA (siRNA) duplex against the M2 subunit of RR (RRM2) is developed and shown to reduce the growth potential of cancer cells both in vitro and in vivo. EXPERIMENTAL DESIGN: Three anti-RRM2 siRNAs were identified via computational methods, and the potency of these and additional "tiling" duplexes was analyzed in cultured cells via cotransfections using a RRM2-luciferase fusion construct. Knockdown of RRM2 by the best duplex candidates was confirmed directly by Western blotting. The effect of potent duplexes on cell growth was investigated by a real-time cell electronic sensing assay. Finally, duplex performance was tested in vivo in luciferase-expressing cells via whole animal bioluminescence imaging. RESULTS: Moderate anti-RRM2 effects are observed from the three duplexes identified by computational methods. However, the tiling experiments yielded an extremely potent duplex (siR2B+5). This duplex achieves significant knockdown of RRM2 protein in cultured cells and has pronounced antiproliferative activity. S.c. tumors of cells that had been transfected with siR2B+5 preinjection grew slower than those of control cells. CONCLUSIONS: An anti-RRM2 siRNA duplex is identified that exhibits significant antiproliferative activity in cancer cells of varying human type and species (mouse, rat, monkey); these findings suggest that this duplex is a promising candidate for therapeutic development.


Subject(s)
Genetic Therapy/methods , RNA, Small Interfering/chemical synthesis , RNA, Small Interfering/genetics , Ribonucleoside Diphosphate Reductase/antagonists & inhibitors , Ribonucleoside Diphosphate Reductase/genetics , Animals , Base Sequence , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Humans , In Vitro Techniques , Molecular Sequence Data , Sequence Homology, Nucleic Acid , Transfection
17.
Bioconjug Chem ; 18(2): 456-68, 2007.
Article in English | MEDLINE | ID: mdl-17326672

ABSTRACT

Nucleic acid-based therapeutics have the potential to provide potent and highly specific treatments for a variety of human ailments. However, systemic delivery continues to be a significant hurdle to success. Multifunctional nanoparticles are being investigated as systemic, nonviral delivery systems, and here, we describe the physicochemical and biological characterization of cyclodextrin-containing polycations (CDP) and their nanoparticles formed with nucleic acids including plasmid DNA (pDNA) and small interfering RNA (siRNA). These polycation/nucleic acid complexes can be tuned by formulation conditions to yield particles with sizes ranging from 60 to 150 nm, zeta potentials from 10 to 30 mV, and molecular weights from approximately 7 x 107 to 1 x 109 g mol-1 as determined by light scattering techniques. Inclusion complexes formed between adamantane (AD)-containing molecules and the beta-cyclodextrin molecules enable the modular attachment of poly(ethylene glycol) (AD-PEG) conjugates for steric stabilization and targeting ligands (AD-PEG-transferrin) for cell-specific targeting. A 70 nm particle can contain approximately 10 000 CDP polymer chains, approximately 2000 siRNA molecules, approximately 4000 AD-PEG5000 molecules, and approximately 100 AD-PEG5000-Tf molecules; this represents a significant payload of siRNA and a large ratio of siRNA to targeting ligand (20:1). The particles protect the nucleic acid payload from nuclease degradation, do not aggregate at physiological salt concentrations, and cause minimal erythrocyte aggregation and complement fixation at the concentrations typically used for in vivo application. Uptake of the nucleic acid-containing particles by HeLa cells is measured by flow cytometry and visualized by confocal microscopy. Competitive uptake experiments show that the transferrin-targeted particles display enhanced affinity for the transferrin receptor through avidity effects (multiligand binding). Functional efficacy of the delivered pDNA and siRNA is demonstrated through luciferase reporter protein expression and knockdown, respectively. The analysis of the CDP delivery vehicle provides insights that can be applied to the design of targeted nucleic acid delivery vehicles in general.


Subject(s)
Cyclodextrins/chemistry , Drug Carriers/chemistry , Nanoparticles , Nucleic Acids/chemistry , Polyamines/chemistry , Transferrin/chemistry , Adamantane/metabolism , Animals , Cattle , Cyclodextrins/metabolism , Drug Carriers/metabolism , Drug Delivery Systems , Electrophoretic Mobility Shift Assay , Erythrocyte Aggregation , Gene Transfer Techniques , HeLa Cells , Humans , Nucleic Acids/metabolism , Plasmids , Polyamines/metabolism , Polyelectrolytes , Polyethylene Glycols/chemistry , RNA, Small Interfering , Receptors, Transferrin/metabolism , Transferrin/metabolism
18.
Biotechnol Bioeng ; 97(4): 909-21, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17154307

ABSTRACT

Small interfering RNA (siRNA) molecules achieve sequence-specific gene silencing through the RNA interference (RNAi) mechanism. Here, live-cell and live-animal bioluminescent imaging (BLI) is used to directly compare luciferase knockdown by unmodified and nuclease-stabilized siRNAs in rapidly (HeLa) and slowly (CCD-1074Sk) dividing cells to reveal the impact of cell division and siRNA nuclease stability on the kinetics of siRNA-mediated gene silencing. Luciferase knockdown using unmodified siRNAs lasts approximately 1 week in HeLa cells and up to 1 month in CCD-1074Sk cells. There is a slight increase in the duration of luciferase knockdown by nuclease-stabilized siRNAs relative to unmodified siRNAs after cationic lipid transfection, but this difference is not observed after electroporation. In BALB/cJ mice, a fourfold increase in maximum luciferase knockdown is observed after hydrodynamic injection (HDI) of nuclease-stabilized siRNAs relative to unmodified siRNAs, yet the overall kinetics of the recovery after knockdown are nearly identical. By using a mathematical model of siRNA-mediated gene silencing, the trends observed in the experimental data can be duplicated by changing model parameters that affect the stability of the siRNAs before they reach the cytosolic compartment. Based on these findings, we hypothesize that the stabilization advantages of nuclease-stabilized siRNAs originate primarily from effects prior to and during internalization before the siRNAs can interact with the intracellular RNAi machinery.


Subject(s)
Endonucleases/metabolism , Gene Silencing , RNA Interference , RNA, Small Interfering/administration & dosage , Animals , Electroporation , Enzyme Stability , HeLa Cells , Humans , In Vitro Techniques , Kinetics , Luciferases/genetics , Luminescent Agents , Luminescent Measurements , Mice , Mice, Inbred BALB C , Plasmids , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transfection
19.
Clin Cancer Res ; 12(5): 1606-14, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16533788

ABSTRACT

Preclinical efficacy of i.v. IT-101, a nanoparticulate conjugate of 20(S)-camptothecin and a cyclodextrin-based polymer, was investigated in several mouse xenografts. The effects of different multiple dosing schedules on tumor growth of LS174T colon carcinoma xenografts are elucidated. All multiple dosing schedules administered over 15 to 19 days resulted in enhanced efficacy compared with untreated or single-dose groups. Further improvements in antitumor efficacy were not observed when the dosing frequency was increased from three weekly doses to five doses at 4-day intervals or 5 days of daily dosing followed by 2 days without dosing repeated in three cycles using similar cumulative doses. This observation was attributed to the extended release characteristics of camptothecin from the polymer. Antitumor efficacy was further evaluated in mice bearing six different s.c. xenografts (LS174T and HT29 colorectal cancer, H1299 non-small-cell lung cancer, H69 small-cell lung cancer, Panc-1 pancreatic cancer, and MDA-MB-231 breast cancer) and one disseminated xenograft (TC71-luc Ewing's sarcoma). In all cases, a single treatment cycle of three weekly doses of IT-101 resulted in a significant antitumor effect. Complete tumor regression was observed in all animals bearing H1299 tumors and in the majority of animals with disseminated Ewing's sarcoma tumors. Importantly, IT-101 is effective in a number of tumors that are resistant to treatment with irinotecan (MDA-MB-231, Panc-1, and HT29), consistent with the hypothesis that polymeric drug conjugates may be able to overcome certain kinds of multidrug resistance. Taken together, these results indicate that IT-101 has good tolerability and antitumor activity against a wide range of tumors.


Subject(s)
Camptothecin/therapeutic use , Cyclodextrins/chemistry , Disease Models, Animal , Polymers/chemistry , Xenograft Model Antitumor Assays , Animals , Breast Neoplasms/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Small Cell/drug therapy , Colorectal Neoplasms/drug therapy , Female , Humans , Lung Neoplasms/drug therapy , Mice , Mice, Nude , Nanotechnology , Pancreatic Neoplasms/drug therapy , Sarcoma, Ewing/drug therapy , Tumor Cells, Cultured
20.
Nucleic Acids Res ; 34(1): 322-33, 2006.
Article in English | MEDLINE | ID: mdl-16410612

ABSTRACT

Small interfering RNA (siRNA) molecules are potent effectors of post-transcriptional gene silencing. Using noninvasive bioluminescent imaging and a mathematical model of siRNA delivery and function, the effects of target-specific and treatment-specific parameters on siRNA-mediated gene silencing are monitored in cells stably expressing the firefly luciferase protein. In vitro, luciferase protein levels recover to pre-treatment values within <1 week in rapidly dividing cell lines, but take longer than 3 weeks to return to steady-state levels in nondividing fibroblasts. Similar results are observed in vivo, with knockdown lasting approximately 10 days in subcutaneous tumors in A/J mice and 3-4 weeks in the nondividing hepatocytes of BALB/c mice. These data indicate that dilution due to cell division, and not intracellular siRNA half-life, governs the duration of gene silencing under these conditions. To demonstrate the practical use of the model in treatment design, model calculations are used to predict the dosing schedule required to maintain persistent silencing of target proteins with different half-lives in rapidly dividing or nondividing cells. The approach of bioluminescent imaging combined with mathematical modeling provides useful insights into siRNA function and may help expedite the translation of siRNA into clinically relevant therapeutics for disease treatment and management.


Subject(s)
RNA Interference , RNA, Small Interfering/administration & dosage , Animals , Cell Line , Cell Proliferation , Hepatocytes/metabolism , Kinetics , Luciferases, Firefly/analysis , Luciferases, Firefly/genetics , Luminescent Agents , Mice , Mice, Inbred BALB C , Models, Biological , Neoplasms/genetics , Neoplasms/metabolism , RNA, Small Interfering/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...