P035 iPSC-derived macrophages from cystic fibrosis patients as a cell source to establish in vitro infection models and a drug screening platform

2021 ◽  
Vol 20 ◽  
pp. S49
Author(s):  
C. Rodríguez-González ◽  
A. Rafiei Hashtchin ◽  
S. Merkert ◽  
A. Munder ◽  
N. Lachmann
2019 ◽  
Vol 14 (16) ◽  
pp. 1437-1450 ◽  
Author(s):  
Thomas J O'Brien ◽  
Martin Welch

The airways of persons with cystic fibrosis are prone to infection by a diverse and dynamic polymicrobial consortium. Currently, no models exist that permit recapitulation of this consortium within the laboratory. Such microbial ecosystems likely have a network of interspecies interactions, serving to modulate metabolic pathways and impact upon disease severity. The contribution of less abundant/fastidious microbial species on this cross-talk has often been neglected due to lack of experimental tractability. Here, we critically assess the existing models for studying polymicrobial infections. Particular attention is paid to 3Rs-compliant in vitro and in silico infection models, offering significant advantages over mammalian infection models. We outline why these models will likely become the ‘go to’ approaches when recapitulating polymicrobial cystic fibrosis infection.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4677-4677 ◽  
Author(s):  
Deepak Balaji balaji Thimiri Govinda Raj ◽  
Mariaserena Giliberto ◽  
Andrea Cremaschi ◽  
Sigrid Strand Skånland ◽  
Alexandra Gade ◽  
...  

Abstract Introduction Multiple Myeloma (MM) is considered incurable and MM patients eventually relapse despite use of many promising approved drugs in standard-of-care treatment. It has been challenging to design precision medicine protocols to tailor personalized treatment for MM patients that relapse despite availability of novel drugs. In-vitro drug screening has been hampered by lack of in-vitro culture protocols that mimic tumor microenvironment and that accommodates for low cell number. Here, we report our novel MM proliferation protocol along with an in-vitro functional screening platform, that allow us to assess drug sensitivity on MM patient samples with a customized panel of 30 myeloma drugs. Using our novel drug sensitivity screening platform, we aim to identify efficient drugs for individual patients with progressive disease and select the best treatment option. Methods Previously, we have established culture settings that mimic the tumor microenvironment for MM (Wang D. et al Leukemia 2017). Here, we implemented a novel protocol that allowed primary MM cells to proliferate in a 384 well-format. Stimulated CD138+ MM cells were tested against a customized library of 30 clinically approved drugs including proteasome inhibitors (PI) and drugs that are in clinical trials. CD138+ MM cells were cultured in 384-well format in the presence of individual drugs in a concentration range over 6 logs for 72 hours (3 days). To define drugs that inhibit malignant plasma cell growth, we used the cell-based assays CellTiter-Glo® luminescent cell viability assay and CellTox™ green cytotoxicity assay as readouts by assessing drug sensitivity at day 3. We performed MM drug screening on 18 patient samples and 6 healthy B-cell (BC) control samples. We performed drug screening on myeloma cells SK-MM2 (patient derived cell line) for 527 drugs at 5 concentrations. We are currently performing drug screening on 11 MM cell lines which represents diverse cancer stage. For each patient sample, a Drug Sensitivity Score (DSS) was calculated for every drug using the IC50 value, slope and the area under the curve (AUC). Next, DSS values for the full MM patient cohort were compared to those of healthy controls to generate a selective DSS (sDSS) for each drug (sDSS = DSSpatient - average DSShealthy). Drugs which had sDSS >5 were considered clearly more effective for patient samples in the in vitro test. MM patient samples were assessed for sDSS score using our screening data and we ranked all the drugs by their sDSS score. We have generated sDSS score for both CTG (cell viability) and CTxG (cell toxicity) datasets. Results and conclusion To date we have performed MM drug sensitivity screening on 18 MM patient samples and 6 healthy B cells donors. We adopted a quantitative scoring approach using sDSS to rank drugs that are selective and effective in inhibiting myeloma cells. Based on our drug sensitivity analysis, proteasome inhibitors such as bortezomib and carfilzomib were more effective in inhibiting myeloma cell proliferation compared to other drugs in all 18 patient samples as well as in the 6 healthy donors. Surprising, doxorubicin showed the highest average sDSS score in 10 patients with score 12.96 followed by prednisolone with average sDSS score 6.73 (Figure 1), while proteasome inhibitor bortezomib showed average sDSS score of 4.14 and carfilzomib showed average sDSS score of 1. In addition, we observed that samples from dexamethasone-treated patients showed lower sDSS score for dexamethasone in the in vitro drug screening compared to samples from untreated patients (MM0905 and MM0706). Based on the screening data and clustering analysis, we concluded that the observed diversity in drug effectiveness between patient samples supports the hypothesis of tumor heterogeneity and creates a basis for exploring the possibility to individualize treatment choices. Figure 1: Selective Drug Sensitivity Screening (sDSS) score for 30 drugs for 13 MM patient samples. HB, Healthy donor B cells (Euclidean distance, Ward linkage method) Disclosures Schjesvold: Novartis: Honoraria; Oncopeptides: Consultancy; Janssen: Consultancy, Honoraria, Research Funding; Adaptive: Consultancy; Bayer: Consultancy; Bristol Myers Squibb: Consultancy; Takeda: Consultancy, Honoraria; Celgene: Consultancy, Honoraria; Amgen: Consultancy, Honoraria, Research Funding; Abbvie: Honoraria.


2016 ◽  
Vol 4 (43) ◽  
pp. 7000-7008 ◽  
Author(s):  
Ching-Yun Chen ◽  
Tsai-Shin Chiang ◽  
Ling-Ling Chiou ◽  
Hsuan-Shu Lee ◽  
Feng-Huei Lin

Since clinical drugs need to be approved for their liver metabolism efficiency before commercialization, a powerful in vitro drug-screening platform is imperative and indispensable for the clinical medicine and pharmaceutical industries.


Pneumologie ◽  
2014 ◽  
Vol 68 (06) ◽  
Author(s):  
R Haller ◽  
S Ulrich ◽  
S Weinreich ◽  
S Merkert ◽  
C Bednarski ◽  
...  

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3624-3624
Author(s):  
Stephen Sze Yuen Lam ◽  
Chae Yin Cher ◽  
Nelson Ka Lam Ng ◽  
Eric SK Ho ◽  
Cheuk Him Man ◽  
...  

Abstract Acute myeloid leukaemia (AML) is a heterogeneous group of diseases with distinct clinicopathologic, cytogenetic and genetic characteristics. The heterogeneity has made unified and regimental approach unsuccessful for most patients, and outcome with standard chemotherapy and allogeneic haematopoietic stem cell transplantation (HSCT) was unsatisfactory with an overall cure rate of 30-40%. We hypothesised that an optimised in vitro drug screening platform for primary AML samples might help to identify the best personalised therapeutic agents for AML patients from whom the samples were obtained at real time. Primary mononuclear cells isolated from peripheral blood (PB) or bone marrow (BM) of AML patients at different stages of disease were seeded onto 96-well plates and treated with a panel of 25 selected drugs at 1000-fold concentration range for 3 days. The drugs included 17 tyrosine kinase inhibitors: axitinib, crizotinib, pazopanib, erlotinib, gefitinib, lapatinib, vandetanib, vemurafenib, sorafenib, quizartinib, ponatinib, lestaurtinib, nilotinib, dasatinib, ruxolitinib, TG101209, tofacitinib; 2 differentiation agents: arsenic trioxide, all-trans retinoic acid; a protein translation inhibitor: homoharringtonine; a proteasome inhibitor: bortezomib; a chemotherapy: cytarabine; a histone deacetylase inhibitor: vorinostat; a DNA methyltransferase inhibitor: azacitidine; and an mTOR inhibitor: rapamycin. The blast percentage in each sample was above 50% as confirmed by film review of the cytospin preparation. The inhibitory effect of each drug was evaluated by a high throughput PrestoBlue® fluorometric assay that was a measure of viable cell number. The results were expressed with reference to the vehicle control (0.1% DMSO) for each sample. There was significant cell death when primary AML cells were cultured in IMDM medium supplemented with 10% FBS (IMDM10) for 3 days (Annexin V+ cells: 53.8% ± 4.5% , n=33). To identify the optimal culture condition, multiple culture conditions were tested for each sample (n=53 samples). The 3-day post-culture survival of primary AML cells improved significantly in a 1:1 mix of IMDM10 and medium conditioned by a mixture of mouse fibroblast lines engineered to produce human G-CSF, SCF, IL-3 and Flt3L (50%CM) (47.1% ± 6.5% improvement in Annexin V/7AAD -/- cell count normalised to day 0 input compared to IMDM10, p<0.0001). Assay readout was highly reproducible between replicates of the same samples (r=0.9681). There was significant correlation in drug response between PB and BM myeloblasts (r=0.9356, p<0.0001, 16 pairs), supporting the feasibility of drug screen based on PB samples. The readout might be predictive of clinical drug response in patients, as exemplified by the superior in vitro response to sorafenib in FLT3-ITD+ than FLT3 wild type AML samples (p<0.01 at 1 and 10 µM Sorafenib) that corroborated with the clinical observation for these patients treated with sorafenib. Furthermore, sorafenib-naïve FLT3-ITD+ AML samples were more sensitive to sorafenib in vitro than the resistant samples collected from the same patients during subsequent leukaemia progression. To identify personalised agents effective for individual patients, a total of 85 samples from 60 AML patients were screened in 50%CM. Axitinib and vemurafenib had minimal cytotoxic effect on all samples whereas bortezomib, ponatinib, lestaurtinib, homoharringtonine and vorinostat exhibited significant anti-leukaemia effect (>75%) to over 90% samples at concentrations around or below the peak plasma concentrations in respective pharmacokinetic studies. The remaining 18 drugs exhibited variable effects on different samples. Further experiments and analyses are underway to identify distinct combinations and correlate with in vivo efficacy in xenotransplantation mouse model. This optimised and clinically relevant in vitro drug screening platform might provide the laboratory basis for the selection of personalised treatment regime for AML patients with a short turnover time. Disclosures No relevant conflicts of interest to declare.


Author(s):  
Liang Ma ◽  
Jeremy Barker ◽  
Changchun Zhou ◽  
Biaoyang Lin ◽  
Wei Li

A cell culture microfluidic device has been developed to test the cytotoxicity of anticancer drugs while reproducing multi-organ interactions in vitro. Cells were cultured in separate chambers representing the liver and tumor. The two chambers were connected through a channel to mimick the blood flow. Glioblastoma (GBM) cancer cells (M059K) and hepatoma cells (HepG2) were cultured in the tumor and the liver chambers, respectively. The cytotoxic effect of cancer treatment drug Temolozomide (TMZ) was tested using this two chamber system. The experimental results showed that with the liver cells, the cancer cells showed much higher viability than those without the liver cells. This indicates that the liver metabolism has strong effect on the toxicity of the anticancer drug. The results demonstrated that the perfused two chamber cell culture system has the potential to be used as a platform for drug screening in a more physiologically realistic environment.


2018 ◽  
Vol 23 (6) ◽  
pp. 599-613 ◽  
Author(s):  
Sandra Laternser ◽  
Hansjoerg Keller ◽  
Olivier Leupin ◽  
Martin Rausch ◽  
Ursula Graf-Hausner ◽  
...  

Two-dimensional (2D) cell cultures do not reflect the in vivo situation, and thus it is important to develop predictive three-dimensional (3D) in vitro models with enhanced reliability and robustness for drug screening applications. Treatments against muscle-related diseases are becoming more prominent due to the growth of the aging population worldwide. In this study, we describe a novel drug screening platform with automated production of 3D musculoskeletal-tendon-like tissues. With 3D bioprinting, alternating layers of photo-polymerized gelatin-methacryloyl-based bioink and cell suspension tissue models were produced in a dumbbell shape onto novel postholder cell culture inserts in 24-well plates. Monocultures of human primary skeletal muscle cells and rat tenocytes were printed around and between the posts. The cells showed high viability in culture and good tissue differentiation, based on marker gene and protein expressions. Different printing patterns of bioink and cells were explored and calcium signaling with Fluo4-loaded cells while electrically stimulated was shown. Finally, controlled co-printing of tenocytes and myoblasts around and between the posts, respectively, was demonstrated followed by co-culture and co-differentiation. This screening platform combining 3D bioprinting with a novel microplate represents a promising tool to address musculoskeletal diseases.


2020 ◽  
Author(s):  
Marvin G. Gunawan ◽  
Sarabjit S. Sangha ◽  
Sanam Shafaattalab ◽  
Eric Lin ◽  
Danielle A. Heims-Waldron ◽  
...  

AbstractCurrent drug development efforts for the treatment of atrial fibrillation (AF) are hampered by the fact that many preclinical models have been unsuccessful in reproducing human cardiac atrial physiology and its response to medications. In this study, we demonstrated an approach using human induced pluripotent stem cell-derived atrial and ventricular cardiomyocytes (hiPSC-aCMs and hiPSC-vCMs, respectively) coupled with a sophisticated optical mapping system for drug screening of atrial-selective compounds in vitro.We optimized differentiation of hiPSC-aCMs by modulating the WNT and retinoid signalling pathways. Characterization of the transcriptome and proteome revealed that retinoic acid pushes the differentiation process into the atrial lineage and generated hiPSC-aCMs. Functional characterization using optical mapping showed that hiPSC-aCMs have shorter action potential durations and faster Ca2+ handling dynamics compared to hiPSC-vCMs. Furthermore, pharmacological investigation of hiPSC-aCMs captured atrial-selective effects by displaying greater sensitivity to atrial-selective compounds 4-aminopyridine, AVE0118, UCL1684, and vernakalant when compared to hiPSC-vCMs.These results established that a model system incorporating hiPSC-aCMs combined with optical mapping is well-suited for pre-clinical drug screening of novel and targeted atrial selective compounds.


Sign in / Sign up

Export Citation Format

Share Document