Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 130(6): 2920-2927, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32045385

ABSTRACT

BACKGROUNDInterventions that interrupt Plasmodium vivax transmission or eliminate dormant P. vivax liver-stage parasites will be essential for malaria elimination. Development of these interventions has been hindered by the lack of P. vivax in vitro culture and could be accelerated by a safe and reproducible clinical model in malaria-naive individuals.METHODSHealthy, malaria-naive adults were enrolled in 2 studies to assess the safety, infectivity, and transmissibility of a new P. vivax isolate. Participants (Study 1, n = 2; Study 2, n = 24) were inoculated with P. vivax-infected red blood cells to initiate infection, and were treated with artemether-lumefantrine (Study 1) or chloroquine (Study 2). Primary endpoints were safety and infectivity of the new isolate. In Study 2, transmission to mosquitoes was also evaluated using mosquito feeding assays, and sporozoite viability was assessed using in vitro cultured hepatocytes.RESULTSParasitemia and gametocytemia developed in all participants and was cleared by antimalarial treatment. Adverse events were mostly mild or moderate and none were serious. Sixty-nine percent of participants (11/16) were infectious to Anopheles mosquitoes at peak gametocytemia. Mosquito infection rates reached 97% following membrane feeding with gametocyte-enriched blood, and sporozoites developed into liver-stage schizonts in culture.CONCLUSIONWe have demonstrated the safe, reproducible, and efficient transmission of P. vivax gametocytes from humans to mosquitoes, and have established an experimental model that will accelerate the development of interventions targeting multiple stages of the P. vivax life cycle.TRIAL REGISTRATIONACTRN12614000930684 and ACTRN12616000174482.FUNDING(Australian) National Health and Medical Research Council Program Grant 1132975 (Study 1). Bill and Melinda Gates Foundation (OPP1111147) (Study 2).


Subject(s)
Artemether, Lumefantrine Drug Combination/administration & dosage , Chloroquine/administration & dosage , Malaria, Vivax , Plasmodium vivax/metabolism , Adolescent , Adult , Animals , Anopheles , Female , Humans , Malaria, Vivax/drug therapy , Malaria, Vivax/metabolism , Malaria, Vivax/transmission , Male , Middle Aged , Models, Biological , Pilot Projects
2.
ACS Appl Mater Interfaces ; 11(10): 9814-9823, 2019 Mar 13.
Article in English | MEDLINE | ID: mdl-30724549

ABSTRACT

Three-dimensional (3D) multicellular structures allow cells to behave and interact with each other in a manner that mimics the in vivo environment. In recent years, many 3D cell culture methods have been developed with the goal of producing the most in vivo-like structures possible. Whilst strongly preferable to  conventional cell culture, these approaches are often poorly reproducible, time-consuming, expensive, and labor-intensive and require specialized equipment. Here, we describe a novel 3D culture platform, which we have termed the naked liquid marble (NLM). Cells are cultured in a liquid drop (the NLM) in superhydrophobic-coated plates, which causes the cells to naturally form 3D structures. Inside the NLMs, cells are free to interact with each other, forming multiple 3D spheroids that are uniform in size and shape in less than 24 h. We showed that this system is highly reproducible, suitable for cell coculture, compound screening, and also compatible with laboratory automation systems. The low cost of production, small volume of each NLM, and production via automated liquid handling make this 3D cell-culturing system particularly suitable for high-throughput screening assays such as drug testing as well as numerous other cell-based research applications.


Subject(s)
Cell Culture Techniques/methods , Drug Screening Assays, Antitumor , High-Throughput Screening Assays , Spheroids, Cellular/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Humans , Spheroids, Cellular/pathology
3.
BMC Cancer ; 18(1): 41, 2018 01 06.
Article in English | MEDLINE | ID: mdl-29304770

ABSTRACT

BACKGROUND: Cancer cell resistance to therapeutics can result from acquired or de novo-mediated factors. Here, we have utilised advanced breast cancer cell culture models to elucidate de novo doxorubicin resistance mechanisms. METHODS: The response of breast cancer cell lines (MCF-7 and MDA-MB-231) to doxorubicin was examined in an in vitro three-dimensional (3D) cell culture model. Cells were cultured with Matrigel™ enabling cellular arrangements into a 3D architecture in conjunction with cell-to-extracellular matrix (ECM) contact. RESULTS: Breast cancer cells cultured in a 3D ECM-based model demonstrated altered sensitivity to doxorubicin, when compared to those grown in corresponding two-dimensional (2D) monolayer culture conditions. Investigations into the factors triggering the observed doxorubicin resistance revealed that cell-to-ECM interactions played a pivotal role. This finding correlated with the up-regulation of pro-survival proteins in 3D ECM-containing cell culture conditions following exposure to doxorubicin. Inhibition of integrin signalling in combination with doxorubicin significantly reduced breast cancer cell viability. Furthermore, breast cancer cells grown in a 3D ECM-based model demonstrated a significantly reduced proliferation rate in comparison to cells cultured in 2D conditions. CONCLUSION: Collectively, these novel findings reveal resistance mechanisms which may contribute to reduced doxorubicin sensitivity.


Subject(s)
Breast Neoplasms/drug therapy , Doxorubicin/administration & dosage , Extracellular Matrix Proteins/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Culture Techniques , Cell Proliferation/drug effects , Cell Survival/drug effects , Collagen/chemistry , Drug Combinations , Drug Resistance, Neoplasm/genetics , Female , Humans , Laminin/chemistry , MCF-7 Cells , Proteoglycans/chemistry , Signal Transduction/drug effects
4.
Molecules ; 22(10)2017 Oct 12.
Article in English | MEDLINE | ID: mdl-29023425

ABSTRACT

Kinetoplastid parasites cause vector-borne parasitic diseases including leishmaniasis, human African trypanosomiasis (HAT) and Chagas disease. These Neglected Tropical Diseases (NTDs) impact on some of the world's lowest socioeconomic communities. Current treatments for these diseases cause severe toxicity and have limited efficacy, highlighting the need to identify new treatments. In this study, the Davis open access natural product-based library was screened against kinetoplastids (Leishmania donovani DD8, Trypanosoma brucei brucei and Trypanosoma cruzi) using phenotypic assays. The aim of this study was to identify hit compounds, with a focus on improved efficacy, selectivity and potential to target several kinetoplastid parasites. The IC50 values of the natural products were obtained for L. donovani DD8, T. b. brucei and T. cruzi in addition to cytotoxicity against the mammalian cell lines, HEK-293, 3T3 and THP-1 cell lines were determined to ascertain parasite selectivity. Thirty-one compounds were identified with IC50 values of ≤ 10 µM against the kinetoplastid parasites tested. Lissoclinotoxin E (1) was the only compound identified with activity across all three investigated parasites, exhibiting IC50 values < 5 µM. In this study, natural products with the potential to be new chemical starting points for drug discovery efforts for kinetoplastid diseases were identified.


Subject(s)
Antiprotozoal Agents/pharmacology , Biological Products/pharmacology , Drug Evaluation, Preclinical , Kinetoplastida/drug effects , Small Molecule Libraries , Animals , Biological Products/chemistry , Cell Line , Drug Discovery , Humans , Inhibitory Concentration 50 , Mice , Parasitic Sensitivity Tests , Trypanosoma brucei gambiense/drug effects , Trypanosoma cruzi/drug effects , Trypanosomiasis, African/drug therapy
5.
Article in English | MEDLINE | ID: mdl-28674055

ABSTRACT

Open-access drug discovery provides a substantial resource for diseases primarily affecting the poor and disadvantaged. The open-access Pathogen Box collection is comprised of compounds with demonstrated biological activity against specific pathogenic organisms. The supply of this resource by the Medicines for Malaria Venture has the potential to provide new chemical starting points for a number of tropical and neglected diseases, through repurposing of these compounds for use in drug discovery campaigns for these additional pathogens. We tested the Pathogen Box against kinetoplastid parasites and malaria life cycle stages in vitro Consequently, chemical starting points for malaria, human African trypanosomiasis, Chagas disease, and leishmaniasis drug discovery efforts have been identified. Inclusive of this in vitro biological evaluation, outcomes from extensive literature reviews and database searches are provided. This information encompasses commercial availability, literature reference citations, other aliases and ChEMBL number with associated biological activity, where available. The release of this new data for the Pathogen Box collection into the public domain will aid the open-source model of drug discovery. Importantly, this will provide novel chemical starting points for drug discovery and target identification in tropical disease research.


Subject(s)
Antimalarials/pharmacology , Malaria/drug therapy , Cell Line , Chagas Disease/drug therapy , Drug Discovery/methods , HEK293 Cells , Humans , Leishmaniasis/drug therapy , Neglected Diseases/drug therapy , Trypanosomiasis, African/drug therapy
6.
Drug Discov Today ; 22(10): 1516-1531, 2017 10.
Article in English | MEDLINE | ID: mdl-28647378

ABSTRACT

Leishmaniasis, caused by the trypanosomatid protozoan Leishmania, is endemic in 98 countries worldwide, with morbidity and mortality increasing daily. Despite available drugs, leishmaniasis faces the challenge of emerging resistance and toxicity concerns for current drug regimes. Identification of anti-leishmanial compounds representing new chemistry and novel mechanisms of action is essential to populate the drug discovery pipeline. The in vitro assays currently available have shown poor translational outcomes, with high compound attrition rates. It is therefore imperative that more physiologically relevant assays are developed to identify anti-leishmanial compounds. This review provides an overview of the disease, current treatment options and compares the various technologies and assay formats currently available for leishmanial drug discovery.


Subject(s)
Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/therapeutic use , Leishmaniasis/drug therapy , Animals , Drug Discovery/methods , Humans , Leishmania/drug effects
7.
Assay Drug Dev Technol ; 14(7): 367-80, 2016 09.
Article in English | MEDLINE | ID: mdl-27552143

ABSTRACT

Pancreatic cancer continues to have one of the poorest prognoses among all cancers. The drug discovery efforts for this disease have largely failed, with no significant improvement in survival outcomes for advanced pancreatic cancer patients over the past 20 years. Traditional in vitro cell culture techniques have been used extensively in both basic and early drug discovery; however, these systems offer poor models to assess emerging therapeutics. More predictive cell-based models, which better capture the cellular heterogeneity and complexities of solid pancreatic tumors, are urgently needed not only to improve drug discovery success but also to provide insight into the tumor biology. Pancreatic tumors are characterized by a unique micro-environment that is surrounded by a dense stroma. A complex network of interactions between extracellular matrix (ECM) components and the effects of cell-to-cell contacts may enhance survival pathways within in vivo tumors. This biological and physical complexity is lost in traditional cell monolayer models. To explore the predictive potential of a more complex cellular system, a three-dimensional (3D) micro-tumor assay was evaluated. Efficacy of six current chemotherapeutics was determined against a panel of primary and metastatic pancreatic tumor cell lines in a miniaturized ECM-based 3D cell culture system. Suitability for potential use in high-throughput screening applications was assessed, including ascertaining the effects that miniaturization and automation had on assay robustness. Cellular health was determined by utilizing an indirect population-based metabolic activity assay and a direct imaging-based cell viability assay.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Culture Techniques/methods , Miniaturization/methods , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Antineoplastic Agents/therapeutic use , Dose-Response Relationship, Drug , Humans , Pancreatic Neoplasms/drug therapy , Treatment Outcome
8.
Expert Opin Drug Discov ; 11(9): 885-94, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27454169

ABSTRACT

INTRODUCTION: Cell culture models have been at the heart of anti-cancer drug discovery programs for over half a century. Advancements in cell culture techniques have seen the rapid evolution of more complex in vitro cell culture models investigated for use in drug discovery. Three-dimensional (3D) cell culture research has become a strong focal point, as this technique permits the recapitulation of the tumor microenvironment. Biologically relevant 3D cellular models have demonstrated significant promise in advancing cancer drug discovery, and will continue to play an increasing role in the future. AREAS COVERED: In this review, recent advances in 3D cell culture techniques and their application in tumor modeling and anti-cancer drug discovery programs are discussed. The topics include selection of cancer cells, 3D cell culture assays (associated endpoint measurements and analysis), 3D microfluidic systems and 3D bio-printing. EXPERT OPINION: Although advanced cancer cell culture models and techniques are becoming commonplace in many research groups, the use of these approaches has yet to be fully embraced in anti-cancer drug applications. Furthermore, limitations associated with analyzing information-rich biological data remain unaddressed.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery/methods , Neoplasms/drug therapy , Animals , Bioprinting , Cell Culture Techniques , Drug Design , Humans , Microfluidic Analytical Techniques/methods , Models, Biological , Neoplasms/pathology , Printing, Three-Dimensional
9.
J Cancer Res Clin Oncol ; 141(5): 951-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25773123

ABSTRACT

PURPOSE: Utilization of miniaturized three-dimensional (3D) cell culture-based assays enables investigation into the anticancer activity of drug candidates and further elucidation of the anticancer profile of standard-of-care chemotherapeutic agents against tumor cells. Drug discovery assays established using 3D cell culture, which better recapitulate the tumor microenvironment, may more accurately reflect the antitumor activity of compounds. METHODS: Several standard-of-care anticancer drugs, epirubicin, paclitaxel and vinorelbine, were evaluated against a panel of breast cancer cell lines grown in a 3D cell culture microenvironment in the presence of extracellular matrix. A comparison of this antitumor activity in 3D conditions was made with that observed in traditional two-dimensional (2D) monolayer conditions. RESULTS: Examination of the above mentioned drugs against breast tumor cells cultured in 3D conditions demonstrated significantly altered potency and efficacy in comparison with cells propagated in a 2D monolayer system. The differences observed were cell line-dependent and drug-specific; the triple-negative cell line MDA-MB-231 and the endocrine receptor-positive cell line MCF-7 consistently displayed resistance to therapeutics with distinct modes of action (i.e., topoisomerase II and microtubules) in 3D cell culture in comparison with ErbB2 receptor-positive BT-474 cells. CONCLUSION: The data presented herein demonstrates the cellular viability and physical changes observed within the 3D spheroid following exposure to drug, which is not always reflected in 2D cell culture models.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Area Under Curve , Cell Culture Techniques , Cell Line, Tumor , Cell Proliferation , Epirubicin/administration & dosage , Female , Humans , Paclitaxel/administration & dosage , Treatment Outcome , Vinblastine/administration & dosage , Vinblastine/analogs & derivatives , Vinorelbine
10.
Biology (Basel) ; 3(2): 345-67, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24887773

ABSTRACT

Human cancer cell lines are an integral part of drug discovery practices. However, modeling the complexity of cancer utilizing these cell lines on standard plastic substrata, does not accurately represent the tumor microenvironment. Research into developing advanced tumor cell culture models in a three-dimensional (3D) architecture that more prescisely characterizes the disease state have been undertaken by a number of laboratories around the world. These 3D cell culture models are particularly beneficial for investigating mechanistic processes and drug resistance in tumor cells. In addition, a range of molecular mechanisms deconstructed by studying cancer cells in 3D models suggest that tumor cells cultured in two-dimensional monolayer conditions do not respond to cancer therapeutics/compounds in a similar manner. Recent studies have demonstrated the potential of utilizing 3D cell culture models in drug discovery programs; however, it is evident that further research is required for the development of more complex models that incorporate the majority of the cellular and physical properties of a tumor.

11.
Assay Drug Dev Technol ; 11(7): 435-48, 2013 Sep.
Article in English | MEDLINE | ID: mdl-25310845

ABSTRACT

A more relevant in vitro cell culture model that closely mimics tumor biology and provides better predictive information on anticancer therapies has been the focus of much attention in recent years. We have developed a three-dimensional (3D) human tumor cell culture model that attempts to recreate the in vivo microenvironment and tumor biology in a miniaturized 384-well plate format. This model aims to exploit the potential of 3D cell culture as a screening tool for novel therapeutics for discovery programs. Here we have evaluated a Matrigel™ based induction of 3D tumor formation using standard labware and plate reading equipment. We have demonstrated that with an optimized protocol, reproducible proliferation, and cell viability data can be obtained across a range of cell lines and reagent batches. A panel of reference drugs was used to validate the suitability of the assays for a high throughput drug discovery program. Indicators of assay reproducibility, such as Z'-factor and coefficient of variation, as well as dose response curves confirmed the robustness of the assays. Several methods of drug activity determination were examined, including metabolic and imaging based assays. These data demonstrate this model as a robust tool for drug discovery bridging the gap between monolayer cell culture and animal models, providing insights into drug efficacy at an earlier time point, ultimately reducing costs and high attrition rates.


Subject(s)
Antineoplastic Agents/administration & dosage , Batch Cell Culture Techniques/instrumentation , Biological Assay/instrumentation , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Drug Evaluation, Preclinical/instrumentation , Models, Biological , Apoptosis/drug effects , Breast Neoplasms/physiopathology , Cell Line, Tumor , Cell Survival/drug effects , Equipment Design , Equipment Failure Analysis , Female , Humans , Miniaturization , Tumor Microenvironment/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...