Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Blood Adv ; 7(6): 900-908, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36044391

ABSTRACT

The American Trial Using Tranexamic Acid (TXA) in Thrombocytopenia (A-TREAT, NCT02578901) demonstrated no superiority of TXA over placebo in preventing World Health Organization (WHO) grade 2 or higher bleeding in patients with severe thrombocytopenia requiring supportive platelet transfusion following myeloablative therapy for hematologic disorders. In this ancillary study, we sought to determine whether this clinical outcome could be explained on the basis of correlative assays of fibrinolysis. Plasma was collected from A-TREAT participants (n = 115) before the initiation of study drug (baseline) and when TXA was at steady-state trough concentration (follow-up). Global fibrinolysis was measured by 3 assays: euglobulin clot lysis time (ECLT), plasmin generation (PG), and tissue-type plasminogen activator (tPA)-challenged clot lysis time (tPA-CLT). TXA was quantified in follow-up samples by tandem mass spectrometry. Baseline samples did not demonstrate fibrinolytic activation by ECLT or tPA-CLT. Furthermore, neither ECLT nor levels of plasminogen activator inhibitor-1, tPA, plasminogen, alpha2-antiplasmin, or plasmin-antiplasmin complexes were associated with a greater risk of WHO grade 2+ bleeding. TXA trough concentrations were highly variable (range, 0.7-10 µg/mL) and did not correlate with bleeding severity, despite the fact that plasma TXA levels correlated strongly with pharmacodynamic assessments by PG (Spearman r, -0.78) and tPA-CLT (r, 0.74). We conclude that (1) no evidence of fibrinolytic activation was observed in these patients with thrombocytopenia, (2) trough TXA concentrations varied significantly between patients receiving the same dosing schedule, and (3) tPA-CLT and PG correlated well with TXA drug levels.


Subject(s)
Antifibrinolytic Agents , Blood Coagulation Disorders , Thrombocytopenia , Tranexamic Acid , Humans , Tranexamic Acid/therapeutic use , Tranexamic Acid/pharmacology , Antifibrinolytic Agents/therapeutic use , Antifibrinolytic Agents/pharmacology , Fibrinolysin/pharmacology , Fibrinolysis/physiology , Hemorrhage/etiology , Thrombocytopenia/drug therapy , Thrombocytopenia/etiology
2.
Antibodies (Basel) ; 10(3)2021 Jul 26.
Article in English | MEDLINE | ID: mdl-34449544

ABSTRACT

Antibody-drug conjugates (ADCs) appear to be in a developmental boom, with five FDA approvals in the last two years and a projected market value of over $4 billion by 2024. Major advancements in the engineering of these novel cytotoxic drug carriers have provided a few early success stories. Although the use of these immunoconjugate agents are still in their infancy, valuable lessons in the engineering of these agents have been learned from both preclinical and clinical failures. It is essential to appreciate how the various mechanisms used to engineer changes in ADCs can alter the complex pharmacology of these agents and allow the ADCs to navigate the modern-day therapeutic challenges within oncology. This review provides a global overview of ADC characteristics which can be engineered to alter the interaction with the immune system, pharmacokinetic and pharmacodynamic profiles, and therapeutic index of ADCs. In addition, this review will highlight some of the engineering approaches being explored in the creation of the next generation of ADCs.

3.
Pharmaceutics ; 13(1)2021 Jan 17.
Article in English | MEDLINE | ID: mdl-33477395

ABSTRACT

Major developments in nanomedicines, such as nanoparticles (NPs), nanosomes, and conjugates, have revolutionized drug delivery capabilities over the past four decades. Although nanocarrier agents provide numerous advantages (e.g., greater solubility and duration of systemic exposure) compared to their small-molecule counterparts, there is considerable inter-patient variability seen in the systemic disposition, tumor delivery and overall pharmacological effects (i.e., anti-tumor efficacy and unwanted toxicity) of NP agents. This review aims to provide a summary of fundamental factors that affect the disposition of NPs in the treatment of cancer and why they should be evaluated during preclinical and clinical development. Furthermore, this chapter will highlight some of the translational challenges associated with elements of NPs and how these issues can only be addressed by detailed and novel pharmacology studies.

4.
Antibodies (Basel) ; 8(1)2019 Jan 01.
Article in English | MEDLINE | ID: mdl-31544809

ABSTRACT

The rapid advancement in the development of therapeutic proteins, including monoclonal antibodies (mAbs) and antibody-drug conjugates (ADCs), has created a novel mechanism to selectively deliver highly potent cytotoxic agents in the treatment of cancer. These agents provide numerous benefits compared to traditional small molecule drugs, though their clinical use still requires optimization. The pharmacology of mAbs/ADCs is complex and because ADCs are comprised of multiple components, individual agent characteristics and patient variables can affect their disposition. To further improve the clinical use and rational development of these agents, it is imperative to comprehend the complex mechanisms employed by antibody-based agents in traversing numerous biological barriers and how agent/patient factors affect tumor delivery, toxicities, efficacy, and ultimately, biodistribution. This review provides an updated summary of factors known to affect the disposition of mAbs/ADCs in development and in clinical use, as well as how these factors should be considered in the selection and design of preclinical studies of ADC agents in development.

5.
Nanomedicine ; 21: 102076, 2019 10.
Article in English | MEDLINE | ID: mdl-31394261

ABSTRACT

Pretargeting is an increasingly explored strategy to improve nanoparticle targeting, in which pretargeting molecules that bind both selected epitopes on target cells and nanocarriers are first administered, followed by the drug-loaded nanocarriers. Bispecific antibodies (bsAb) represent a promising class of pretargeting molecules, but how different bsAb formats may impact the efficiency of pretargeting remains poorly understood, in particular Fab valency and Fc receptor (FcR)-binding of bsAb. We found the tetravalent bsAb markedly enhanced PEGylated nanoparticle binding to target HER2+ cells relative to the bivalent bsAb in vitro. Pretargeting with tetravalent bsAb with abrogated FcR binding increased tumor accumulation of PEGylated liposomal doxorubicin (PLD) 3-fold compared to passively targeted PLD alone, and 5-fold vs pretargeting with tetravalent bsAb with normal FcR binding in vivo. Our work demonstrates that multivalency and elimination of FcRn recycling are both important features of pretargeting molecules, and further supports pretargeting as a promising nanoparticle delivery strategy.


Subject(s)
Antibodies, Bispecific , Antineoplastic Agents, Immunological , Drug Carriers , Neoplasms, Experimental , Polyethylene Glycols , Receptor, ErbB-2/antagonists & inhibitors , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/pharmacology , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/pharmacology , Cell Line, Tumor , Drug Carriers/chemistry , Drug Carriers/pharmacology , Female , Humans , Mice, Nude , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Xenograft Model Antitumor Assays , omega-Chloroacetophenone
6.
Cancer Chemother Pharmacol ; 83(1): 61-70, 2019 01.
Article in English | MEDLINE | ID: mdl-30327876

ABSTRACT

PURPOSE: Obesity may alter mononuclear phagocyte system (MPS) function and the pharmacology and efficacy of nanoparticles therapies, such as PEGylated liposomal doxorubicin (PLD). We aimed to evaluate the relationships between hormone and chemokine mediators of MPS function and the pharmacokinetic (PK) exposure of PLD in obese and normal weight patients with ovarian and endometrial cancer. METHODS: Hormone and chemokine mediators in obese and normal weight ovarian and endometrial cancer patients were measured. A separate pharmacology study was performed that evaluated the relationship between serum hormone concentrations, MPS function, and PK disposition of PLD in refractory ovarian cancer patients. RESULTS: Univariate analysis revealed a significant relationship between serum estradiol and body mass index (OR 8.64, 95% CI 2.67-28.0, p < 0.001). Estrone and testosterone concentrations were positively correlated with MPS function (ρ = 0.57 and 0.53, p = 0.14 and 0.18, respectively) and inversely correlated with PLD PK exposure (ρ = - 0.75 and - 0.76, respectively, p = 0.02 for both). CONCLUSIONS: Higher MPS function resulting in reduced PLD exposure is a potential mechanism for reduced efficacy of PLD and other nanoparticles observed in obese patients with cancer. PK simulations suggest higher doses of PLD are required in obese patients to achieve similar exposures as standard dosing in normal weight patients.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Doxorubicin/analogs & derivatives , Endometrial Neoplasms/pathology , Mononuclear Phagocyte System/physiopathology , Nanoparticles/administration & dosage , Obesity/physiopathology , Ovarian Neoplasms/pathology , Antibiotics, Antineoplastic/chemistry , Case-Control Studies , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Endometrial Neoplasms/drug therapy , Female , Follow-Up Studies , Humans , Middle Aged , Nanoparticles/chemistry , Ovarian Neoplasms/drug therapy , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Prognosis
7.
Adv Drug Deliv Rev ; 136-137: 82-96, 2018.
Article in English | MEDLINE | ID: mdl-30273617

ABSTRACT

Nanotechnology provides many solutions to improve conventional drug delivery and has a unique niche in the areas related to the specific targeting of the immune system, such as immunotherapies and vaccines. Preclinical studies in this field rely heavily on the combination of in vitro and in vivo methods to assess the safety and efficacy of nanotechnology platforms, nanoparticle-formulated drugs, and vaccines. While certain types of toxicities can be evaluated in vitro and good in vitro-in vivo correlation has been demonstrated for such tests, animal studies are still needed to address complex biological questions and, therefore, provide a unique contribution to establishing nanoparticle safety and efficacy profiles. The genetic, metabolic, mechanistic, and phenotypic diversity of currently available animal models often complicates both the animal choice and the interpretation of the results. This review summarizes current knowledge about differences in the immune system function and immunological responses of animals commonly used in preclinical studies of nanomaterials. We discuss challenges, highlight current gaps, and propose recommendations for animal model selection to streamline preclinical analysis of nanotechnology formulations.


Subject(s)
Immune System/innervation , Models, Animal , Nanostructures/chemistry , Nanotechnology , Animals , Immune System/immunology
8.
Methods Mol Biol ; 1831: 201-228, 2018.
Article in English | MEDLINE | ID: mdl-30051434

ABSTRACT

Major advances in carrier-mediated agents (CMAs), which include nanoparticles, nanosomes, and conjugates, have revolutionized drug delivery capabilities over the past decade. While providing numerous advantages, such as greater solubility, duration of exposure, and delivery to the site of action over their small molecule counterparts, there is substantial variability in systemic clearance and distribution, tumor delivery, and pharmacologic effects (efficacy and toxicity) of these agents. In this chapter, we focus on the analytical and phenotypic methods required to design a study that characterizes the pharmacokinetics (PK) and pharmacodynamics (PD) of all forms of these nanoparticle-based drug agents. These methods include separation of encapsulated and released drugs, ultrafiltration for measurement of non-protein bound active drug, microdialysis to measure intra-tumor drug concentrations, immunomagnetic separation and flow cytometry for sorting cell types, and evaluation of spatial distribution of drug forms relative to tissue architecture by mass spectrometry imaging and immunohistochemistry.


Subject(s)
Drug Carriers/chemistry , Drug Delivery Systems/methods , Pharmacokinetics , Animals , Humans , Nanoparticles/chemistry , Phenotype
9.
Antibodies (Basel) ; 7(1)2018 Feb 07.
Article in English | MEDLINE | ID: mdl-31544862

ABSTRACT

Major advances in therapeutic proteins, including antibody-drug conjugates (ADCs), have created revolutionary drug delivery systems in cancer over the past decade. While these immunoconjugate agents provide several advantages compared to their small-molecule counterparts, their clinical use is still in its infancy. The considerations in their development and clinical use are complex, and consist of multiple components and variables that can affect the pharmacologic characteristics. It is critical to understand the mechanisms employed by ADCs in navigating biological barriers and how these factors affect their biodistribution, delivery to tumors, efficacy, and toxicity. Thus, future studies are warranted to better understand the complex pharmacology and interaction between ADC carriers and biological systems, such as the mononuclear phagocyte system (MPS) and tumor microenvironment. This review provides an overview of factors that affect the pharmacologic profiles of ADC therapies that are currently in clinical use and development.

10.
Nanomedicine (Lond) ; 12(16): 2021-2042, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28745129

ABSTRACT

Major advances in carrier-mediated agents, including nanoparticle, conjugates and antibody-drug conjugates, have created revolutionary drug delivery systems in cancer over the past two decades. While these agents provide several advantages, such as greater duration of exposure and solubility, compared with their small-molecule counterparts, there is substantial variability in delivery of these agents to tissues and especially tumors. This review provides an overview of tumor microenvironment factors that affect the pharmacokinetics and pharmacodynamics of carrier-mediated agents observed in preclinical models and patients.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems/methods , Neoplasms/drug therapy , Tumor Microenvironment/physiology , Animals , Antineoplastic Agents/pharmacokinetics , Drug Liberation , Humans , Molecular Targeted Therapy/methods , Nanoparticles/chemistry , Neoplasms/metabolism , Permeability
11.
Int J Pharm ; 526(1-2): 443-454, 2017 Jun 30.
Article in English | MEDLINE | ID: mdl-28473237

ABSTRACT

Studies have shown that nanoparticles (NPs) are cleared through the mononuclear phagocyte system (MPS). Pharmacokinetic studies of Doxil, DaunoXome, micellar doxorubicin (SP1049C) and small molecule (SM) doxorubicin were performed in SCID mice, Sprague-Dawley rats, and beagle dogs. An ex vivo MPS profiling platform was used to evaluate the interaction between the same agents, as well as colloid-forming and non-colloid forming SM drugs. In all species, the systemic clearance was highest for SP1049C and lowest for Doxil. With the exception of dog blood, the MPS screening results of mouse and rat blood showed that the greatest reduction in phagocytosis occurred after the ex vivo addition of SM-doxorubicin>SP1049C>DaunoXome>Doxil. The MPS profiling platform in rats, but not dogs, could differentiate between colloid forming and non-colloid forming drugs. The results of the MPS profiling platform were generally consistent with in vivo clearance rates of NP and SM anticancer drugs in mice and rats. This study suggests the MPS profiling platform is an effective method to screen and differentiate the important characteristics of NPs and colloid-forming drugs that affect their in vivo clearance. Implications of these findings on preclinical prediction of human clearance are discussed.


Subject(s)
Colloids/pharmacology , Mononuclear Phagocyte System/drug effects , Nanoparticles/chemistry , Animals , Dogs , Humans , Mice , Mice, SCID , Rats , Rats, Sprague-Dawley
12.
Nanomedicine ; 13(2): 471-482, 2017 02.
Article in English | MEDLINE | ID: mdl-27720926

ABSTRACT

The mononuclear phagocyte system (MPS) has previously been shown to significantly affect the clearance, tumor delivery, and efficacy of nanoparticles (NPs). This study profiled MPS cell infiltration in murine preclinical tumor models and evaluated how these differences may affect tumor disposition of PEGylated liposomal doxorubicin (PLD) in models sensitive and resistant to PLD. Significant differences in MPS presence existed between tumor types (e.g. ovarian versus endometrial), cell lines within the same tumor type, and location of tumor implantation (i.e. flank versus orthotopic xenografts). Further, the differences in MPS presence of SKOV-3 ovarian and HEC1A endometrial orthotopic cancer models may account for the 2.6-fold greater PLD tumor exposure in SKOV-3, despite similar plasma, liver and spleen exposures. These findings suggest that profiling the presence of MPS cells within and between tumor types is important in tumor model selection and in tumor types and patients likely to respond to NP treatment.


Subject(s)
Antibiotics, Antineoplastic/pharmacokinetics , Doxorubicin/pharmacology , Macrophages/drug effects , Polyethylene Glycols , Animals , Antibiotics, Antineoplastic/administration & dosage , Doxorubicin/administration & dosage , Female , Humans , Mice , Models, Biological , Nanoparticles , Neoplasms/drug therapy , Ovarian Neoplasms/drug therapy , Phospholipids/therapeutic use
13.
Nanomedicine ; 12(7): 2007-2017, 2016 10.
Article in English | MEDLINE | ID: mdl-27288666

ABSTRACT

Nanoparticles (NP) including liposomes are cleared by phagocytes of the mononuclear phagocyte system. High inter-patient variability in pharmacokinetics of PEGylated liposomal doxorubicin (PLD) has been reported. We hypothesized that genetic factors may be associated with the variable disposition of PLD. We evaluated plasma and tissue disposition of doxorubicin after administration of PLD at 6mg/kg IV ×1 via tail vein in 23 different male inbred mouse strains. An approximately 13-fold difference in plasma clearance of PLD was observed among inbred strains. We identified a correlation between strain-specific differences in PLD clearance and genetic variation within a genomic region encoding GULP1 (PTB domain containing engulfment adapter 1) protein using haplotype associated mapping and the efficient mixed-model association algorithms. Our results also show that Gulp1 expression in adipose tissue was associated with PLD disposition in plasma. Our findings suggest that genetic variants may be associated with inter-individual pharmacokinetic differences in NP clearance.


Subject(s)
Doxorubicin , Nanoparticles , Adaptor Proteins, Signal Transducing , Animals , Humans , Kinetics , Liposomes , Male , Mice , Mice, Inbred Strains , Pharmacogenomic Variants , Polyethylene Glycols
14.
Article in English | MEDLINE | ID: mdl-26846457

ABSTRACT

Major advances in carrier-mediated agents (CMAs), which include nanoparticles and conjugates, have revolutionized drug delivery capabilities over the past decade. While providing numerous advantages over their small-molecule counterparts, there is substantial variability in how individual CMA formulations and patient characteristics affect the pharmacology, pharmacokinetics (PK), and pharmacodynamics (PD) (efficacy and toxicity) of these agents. Development or selection of animal models is used to predict the effects within a particular human disease. A breadth of studies have begun to emphasize the importance of preclinical animal models in understanding and evaluating the interaction between CMAs and the immune system and tumor matrix, which ultimately influences CMA PK (clearance and distribution) and PD (efficacy and toxicity). It is fundamental to study representative preclinical tumor models that recapitulate patients with diseases (e.g., cancer) and evaluate the interplay between CMAs and the immune system, including the mononuclear phagocyte system (MPS), chemokines, hormones, and other immune modulators. Furthermore, standard allometric scaling using body weight does not accurately predict drug clearance in humans. Future studies are warranted to better understand the complex pharmacology and interaction of CMA carriers within individual preclinical models and their biological systems, such as the MPS and tumor microenvironment, and their application to allometric scaling across species. WIREs Nanomed Nanobiotechnol 2016, 8:642-653. doi: 10.1002/wnan.1394 For further resources related to this article, please visit the WIREs website.


Subject(s)
Antineoplastic Agents , Chemistry, Pharmaceutical , Drug Carriers , Nanomedicine , Nanoparticles , Animals , Drug Evaluation , Drug Evaluation, Preclinical , Humans , Mice
15.
J Pharm Biomed Anal ; 119: 122-9, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26678179

ABSTRACT

Doxorubicin, a widely used anticancer agent, exhibits antitumor activity against a wide variety of malignancies. The drug exerts its cytotoxic effects by binding to and intercalating within the DNA of tumor and tissue cells. However, current assays are unable to accurately determine the concentration of the intracellular active form of doxorubicin. Thus, the development of a sample processing method and a high-performance liquid chromatography (HPLC) methodology was performed in order to quantify doxorubicin that is associated with DNA in tumors and tissues, which provided an intracellular cytotoxic measure of doxorubicin exposure after administration of small molecule and nanoparticle formulations of doxorubicin. The assay uses daunorubicin as an internal standard; liquid-liquid phase extraction to isolate drug associated with DNA; a Shimadzu HPLC with fluorescence detection equipped with a Phenomenex Luna C18 (2µm, 2.0×100mm) analytical column and a gradient mobile phase of 0.1% formic acid in water or acetonitrile for separation and quantification. The assay has a lower limit of detection (LLOQ) of 10ng/mL and is shown to be linear up to 3000ng/mL. The intra- and inter-day precision of the assay expressed as a coefficient of variation (CV%) ranged from 4.01 to 8.81%. Furthermore, the suitability of this assay for measuring doxorubicin associated with DNA in vivo was demonstrated by using it to quantify the doxorubicin concentration within tumor samples from SKOV3 and HEC1A mice obtained 72h after administration of PEGylated liposomal doxorubicin (Doxil(®); PLD) at 6mg/kg IV x 1. This HPLC assay allows for sensitive intracellular quantification of doxorubicin and will be an important tool for future studies evaluating intracellular pharmacokinetics of doxorubicin and various nanoparticle formulations of doxorubicin.


Subject(s)
Antibiotics, Antineoplastic/analysis , Chromatography, High Pressure Liquid/methods , DNA Adducts/analysis , DNA/chemistry , Doxorubicin/analysis , Liver/metabolism , Ovarian Neoplasms/metabolism , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Cell Line, Tumor , DNA Adducts/pharmacokinetics , Doxorubicin/pharmacokinetics , Drug Stability , Female , Humans , Limit of Detection , Mice, SCID , Reproducibility of Results , Tissue Distribution
16.
Hosp Pharm ; 51(3): 237-245, 2016 Mar.
Article in English | MEDLINE | ID: mdl-38745569

ABSTRACT

Background: In 2013, the American Society of Health-System Pharmacists (ASHP) endorsed a policy recommending the development of nationally standardized drug concentrations and dosing units. Although many hospitals have started standardizing their intravenous (IV) solutions, few have taken the practice beyond their institutions or local geographical areas. Objective: This project evaluates the current IV standardization practices for adult patients across hospitals in North Carolina and compares them with each other. In addition, this project proposed standards and evaluated them for their impact on reducing observed variability. Methods: In the fall of 2013, an e-mail request was sent to select hospital pharmacy leaders in North Carolina asking them to voluntarily submit a copy of their adult IV standard concentrations and dosing guidelines. From these lists, the data were summarized and compiled to evaluate trends and compare the various policies. Results: A total of 18 different hospitals and health systems responded. Survey results showed more than 255 concentrations for 84 unique drugs. Of these, 37 were high-risk medications, with 135 unique drug concentrations. From this list, a single proposed concentration was developed for each medication. If utilized, this standardization would result in a greater than 65% reduction in potential drug concentrations in use. A single dosing unit was also proposed for all medications reviewed, resulting in a greater than 21% reduction in variation. Conclusions: Standardization of IV drug concentrations and dosing units statewide could reduce the variability in IV therapies and promote safer and more consistent practices in medication administration.

17.
Expert Opin Drug Metab Toxicol ; 11(9): 1419-33, 2015.
Article in English | MEDLINE | ID: mdl-26173794

ABSTRACT

INTRODUCTION: Major advances in carrier-mediated agents (CMAs), which include nanoparticles and conjugates, have revolutionized drug delivery capabilities over the past decade. While providing numerous advantages such as increased exposure duration, greater solubility and delivery to tumor sites over their small molecule counterparts, there is substantial variability in how individual CMA formulations affect the pharmacology, pharmacokinetics and pharmacodynamics (efficacy and toxicity) of these agents. AREAS COVERED: CMA formulations are complex in nature compared to their small molecule counterparts and consist of multiple components and variables that can affect the pharmacological profile. This review provides an overview of factors that affect the pharmacologic profiles observed in CMA-formulated chemotherapy, primarily in liposomal formulations, that are currently in preclinical or early clinical development. EXPERT OPINION: Despite the numerous advantages that CMA formulations provide, their clinical use is still in its infancy. It is critical that we understand the mechanisms and effects of CMAs in navigating biological barriers and how these factors affect their biodistribution and delivery to tumors. Future studies are warranted to better understand the complex pharmacology and interaction between CMA carriers and biological systems, such as the mononuclear phagocyte system and tumor microenvironment.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems , Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Drug Carriers/chemistry , Drug Design , Humans , Liposomes , Nanoparticles , Neoplasms/pathology , Solubility , Tissue Distribution , Tumor Microenvironment
18.
PLoS One ; 9(11): e112823, 2014.
Article in English | MEDLINE | ID: mdl-25396429

ABSTRACT

The translationally-controlled tumor protein (TCTP) is a highly conserved, ubiquitously expressed, abundant protein that is broadly distributed among eukaryotes. Its biological function spans numerous cellular processes ranging from regulation of the cell cycle and microtubule stabilization to cell growth, transformation, and death processes. In this work, we propose a new function for TCTP as a "buffer protein" controlling cellular homeostasis. We demonstrate that binding of hemin to TCTP is mediated by a conserved His-containing motif (His76His77) followed by dimerization, an event that involves ligand-mediated conformational changes and that is necessary to trigger TCTP's cytokine-like activity. Mutation in both His residues to Ala prevents hemin from binding and abrogates oligomerization, suggesting that the ligand site localizes at the interface of the oligomer. Unlike heme, binding of Ca2+ ligand to TCTP does not alter its monomeric state; although, Ca2+ is able to destabilize an existing TCTP dimer created by hemin addition. In agreement with TCTP's proposed buffer function, ligand binding occurs at high concentration, allowing the "buffer" condition to be dissociated from TCTP's role as a component of signal transduction mechanisms.


Subject(s)
Biomarkers, Tumor/chemistry , Biomarkers, Tumor/metabolism , Hemin/metabolism , Homeostasis/physiology , Amino Acid Motifs/genetics , Amino Acid Sequence , Calcium/metabolism , Chromatography, High Pressure Liquid , Circular Dichroism , Computational Biology , Cross-Linking Reagents , Dimerization , Electrophoresis, Polyacrylamide Gel , Fluorescence , Homeostasis/genetics , Humans , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Biosynthesis/physiology , Protein Conformation , Sequence Alignment , Tumor Protein, Translationally-Controlled 1 , Ultracentrifugation
SELECTION OF CITATIONS
SEARCH DETAIL
...