6,818 research outputs found

    High-throughput screening for drug discovery targeting the cancer cell-microenvironment interactions in hematological cancers

    Get PDF
    Introduction The interactions between leukemic blasts and cells within the bone marrow environment affect oncogenesis, cancer stem cell survival, as well as drug resistance in hematological cancers. The importance of this interaction is increasingly being recognized as a potentially important target for future drug discoveries and developments. Recent innovations in the high throughput drug screening-related technologies, novel ex-vivo disease-models, and freely available machine-learning algorithms are advancing the drug discovery process by targeting earlier undruggable proteins, complex pathways, as well as physical interactions (e.g. leukemic cell-bone microenvironment interaction). Area covered In this review, the authors discuss the recent methodological advancements and existing challenges to target specialized hematopoietic niches within the bone marrow during leukemia and suggest how such methods can be used to identify drugs targeting leukemic cell-bone microenvironment interactions. Expert opinion The recent development in cell-cell communication scoring technology and culture conditions can speed up the drug discovery by targeting the cell-microenvironment interaction. However, to accelerate this process, collecting clinical-relevant patient tissues, developing culture model systems, and implementing computational algorithms, especially trained to predict drugs and their combination targeting the cancer cell-bone microenvironment interaction are needed.Peer reviewe

    Pre-Training on In Vitro and Fine-Tuning on Patient-Derived Data Improves Deep Neural Networks for Anti-Cancer Drug-Sensitivity Prediction

    Get PDF
    Large-scale databases that report the inhibitory capacities of many combinations of candidate drug compounds and cultivated cancer cell lines have driven the development of preclinical drug-sensitivity models based on machine learning. However, cultivated cell lines have devolved from human cancer cells over years or even decades under selective pressure in culture conditions. Moreover, models that have been trained on in vitro data cannot account for interactions with other types of cells. Drug-response data that are based on patient-derived cell cultures, xenografts, and organoids, on the other hand, are not available in the quantities that are needed to train high-capacity machine-learning models. We found that pre-training deep neural network models of drug sensitivity on in vitro drug-sensitivity databases before fine-tuning the model parameters on patient-derived data improves the models’ accuracy and improves the biological plausibility of the features, compared to training only on patient-derived data. From our experiments, we can conclude that pre-trained models outperform models that have been trained on the target domains in the vast majority of cases

    Transcriptomic data integration for precision medicine in leukemia

    Get PDF
    This thesis is comprised of three studies demonstrating the application of different statistical and bioinformatic approaches to address distinct challenges of implementing precision medicine strategies for hematological malignancies. The approaches focus on the analysis of next-generation sequencing data, including both genomic and transcriptomics, to deconvolute disease biology and underlying mechanisms of drug sensitivities and resistance. The outcomes of the studies have clinical implications for advancing current diagnosis and treatment paradigms in patients with hematological diseases. Study I, RNA sequencing has not been widely adopted in a clinical diagnostic setting due to continuous development and lack of standardization. Here, the aim was to evaluate the efficiency of two different RNA-seq library preparation protocols applied to cells collected from acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) patients. The poly-A-tailed mRNA selection (PA) and ribo- depletion (RD) based RNA-seq library preparation protocols were compared and evaluated for detection of gene fusions, variant calling and gene expression profiling. Overall, both protocols produced broadly consistent results and similar outcomes. However, the PA protocol was more efficient in quantifying expression of leukemia marker genes and drug targets. It also provided higher sensitivity and specificity for expression-based classification of leukemia. In contrast, the RD protocol was more suitable for gene fusion detection and captured a greater number of transcripts. Importantly, high technical variations were observed in samples from two leukemia patient cases suggesting further development of strategies for transcriptomic quantification and data analysis. Study II, the BCL-2 inhibitor venetoclax is an approved and effective agent in combination with hypomethylating agents or low dose cytarabine for AML patients, unfit for intensive induction chemotherapy. However, a limited number of patients responding to venetoclax and development of resistance to the treatment presents a challenge for using the drug to benefit the majority of the AML patients. The aim was to investigate genomic and transcriptomic biomarkers for venetoclax sensitivity and enable identification of the patients who are most responsive to venetoclax treatment. We found that venetoclax sensitive samples are enriched with WT1 and IDH1/IDH2 mutations. Intriguingly, HOX family genes, including HOXB9, HOXA5, HOXB3, HOXB4, were found to be significantly overexpressed in venetoclax sensitive patients. Thus, these HOX-cluster genes expression biomarkers can be explored in a clinical trial setting to stratify AML patients responding to venetoclax based therapies. Study III, venetoclax treatment does not benefit all AML patients that demands identifying biomarkers to exclude the patients from venetoclax based therapies. The aim was to investigate transcriptomic biomarkers for ex vivo venetoclax resistance in AML patients. The correlation of ex vivo venetoclax response with gene expression profiles using a machine learning approach revealed significant overexpression of S100 family genes, S100A8 and S100A9. Moreover, high expression ofS100A9was found to be associated with birabresib (BET inhibitor) sensitivity. The overexpression of S100A8 and S100A9 could potentially be used to detect and monitor venetoclax resistance. The combination of BCL-2 and BET inhibitors may sensitize AML cells to venetoclax upon BET inhibition and block leukemic cell survival.In this thesis, the aim was to utilize gene expression information for advanced precision medicine outcomes in patients with hematological malignancies. In the study, I, the contemporary mainstream library preparation protocols, Ribo-depletion and PolyA enrichment used for RNA sequencing, were compared in order to select the protocol that suffices the goal of the experiment, especially in patients with acute leukemias. In study II, we applied bioinformatics approaches to identify IDH1/2 mutation and HOX family gene expression correlated with ex vivo sensitivity to BCL-2 inhibitor venetoclax in acute myeloid leukemia (AML) patients. In study III, statistical and machine learning methods were implemented to identify S100A8/A9 gene expression biomarkers for ex vivo resistance to venetoclax in AML patients. In summary, this thesis addresses the challenges of utilizing gene expression information to stratify patients based on biomarkers to promote precision medicine practice in hematological malignancies

    A Patient-Derived Cell Atlas Informs Precision Targeting of Glioblastoma

    Get PDF
    Glioblastoma (GBM) is a malignant brain tumor with few therapeutic options. The disease presents with a complex spectrum of genomic aberrations, but the pharmacological consequences of these aberrations are partly unknown. Here, we report an integrated pharmacogenomic analysis of 100 patient-derived GBM cell cultures from the human glioma cell culture (HGCC) cohort. Exploring 1,544 drugs, we find that GBM has two main pharmacological subgroups, marked by differential response to proteasome inhibitors and mutually exclusive aberrations in TP53 and CDKN2A/B. We confirm this trend in cell and in xenotransplantation models, and identify both Bcl-2 family inhibitors and p53 activators as potentiators of proteasome inhibitors in GBM cells, We can further predict the responses of individual cell cultures to several existing drug classes, presenting opportunities for drug repurposing and design of stratified trials. Our functionally profiled biobank provides a valuable resource for the discovery of new treatments for GBM.Patrik Johansson, Cecilia Krona and Soumi Kundu share first authorship</p

    Tumor heterogeneity in glioblastoma:a real-life brain teaser

    Get PDF

    INTEGRATIVE ANALYSIS OF OMICS DATA IN ADULT GLIOMA AND OTHER TCGA CANCERS TO GUIDE PRECISION MEDICINE

    Get PDF
    Transcriptomic profiling and gene expression signatures have been widely applied as effective approaches for enhancing the molecular classification, diagnosis, prognosis or prediction of therapeutic response towards personalized therapy for cancer patients. Thanks to modern genome-wide profiling technology, scientists are able to build engines leveraging massive genomic variations and integrating with clinical data to identify “at risk” individuals for the sake of prevention, diagnosis and therapeutic interventions. In my graduate work for my Ph.D. thesis, I have investigated genomic sequencing data mining to comprehensively characterise molecular classifications and aberrant genomic events associated with clinical prognosis and treatment response, through applying high-dimensional omics genomic data to promote the understanding of gene signatures and somatic molecular alterations contributing to cancer progression and clinical outcomes. Following this motivation, my dissertation has been focused on the following three topics in translational genomics. 1) Characterization of transcriptomic plasticity and its association with the tumor microenvironment in glioblastoma (GBM). I have integrated transcriptomic, genomic, protein and clinical data to increase the accuracy of GBM classification, and identify the association between the GBM mesenchymal subtype and reduced tumorpurity, accompanied with increased presence of tumor-associated microglia. Then I have tackled the sole source of microglial as intrinsic tumor bulk but not their corresponding neurosphere cells through both transcriptional and protein level analysis using a panel of sphere-forming glioma cultures and their parent GBM samples.FurthermoreI have demonstrated my hypothesis through longitudinal analysis of paired primary and recurrent GBM samples that the phenotypic alterations of GBM subtypes are not due to intrinsic proneural-to-mesenchymal transition in tumor cells, rather it is intertwined with increased level of microglia upon disease recurrence. Collectively I have elucidated the critical role of tumor microenvironment (Microglia and macrophages from central nervous system) contributing to the intra-tumor heterogeneity and accurate classification of GBM patients based on transcriptomic profiling, which will not only significantly impact on clinical perspective but also pave the way for preclinical cancer research. 2) Identification of prognostic gene signatures that stratify adult diffuse glioma patientsharboring1p/19q co-deletions. I have compared multiple statistical methods and derived a gene signature significantly associated with survival by applying a machine learning algorithm. Then I have identified inflammatory response and acetylation activity that associated with malignant progression of 1p/19q co-deleted glioma. In addition, I showed this signature translates to other types of adult diffuse glioma, suggesting its universality in the pathobiology of other subset gliomas. My efforts on integrative data analysis of this highly curated data set usingoptimizedstatistical models will reflect the pending update to WHO classification system oftumorsin the central nervous system (CNS). 3) Comprehensive characterization of somatic fusion transcripts in Pan-Cancers. I have identified a panel of novel fusion transcripts across all of TCGA cancer types through transcriptomic profiling. Then I have predicted fusion proteins with kinase activity and hub function of pathway network based on the annotation of genetically mobile domains and functional domain architectures. I have evaluated a panel of in -frame gene fusions as potential driver mutations based on network fusion centrality hypothesis. I have also characterised the emerging complexity of genetic architecture in fusion transcripts through integrating genomic structure and somatic variants and delineating the distinct genomic patterns of fusion events across different cancer types. Overall my exploration of the pathogenetic impact and clinical relevance of candidate gene fusions have provided fundamental insights into the management of a subset of cancer patients by predicting the oncogenic signalling and specific drug targets encoded by these fusion genes. Taken together, the translational genomic research I have conducted during my Ph.D. study will shed new light on precision medicine and contribute to the cancer research community. The novel classification concept, gene signature and fusion transcripts I have identified will address several hotly debated issues in translational genomics, such as complex interactions between tumor bulks and their adjacent microenvironments, prognostic markers for clinical diagnostics and personalized therapy, distinct patterns of genomic structure alterations and oncogenic events in different cancer types, therefore facilitating our understanding of genomic alterations and moving us towards the development of precision medicine

    Cancer Drug Screening Scale-up: Combining Biomimetic Microfluidic Platforms and Deep Learning Image Analysis

    Full text link
    The development of cancer drugs is usually costly and time-consuming, mainly due to growing complexity in screening large number of candidate compounds and high failure rates in translation from preclinical trials to clinical approval. Despite the great efforts, the preclinical screening platforms combing good clinical relevance and high throughput for large-scale drug testing is still lacking. In addition, accumulating evidence suggests that cancer drug response can be altered by tumor microenvironment (TME), which includes not only cancer cells but also physical, and biochemical cues in niches. To improve the current cancer drug screening assays, it is important to mimic local TME to achieve better physiological relevance. In the first part of this dissertation, three TME-mimicking microfluidic platforms were introduced for three different in-vitro TME-mimicking tumor sphere models: spheres in matrix, self-aggregated spheres, and single-cell clonal spheres. First, a 3D gel-island chip investigated the heterogeneity of single-cell drug responses in biomimetic extracellular matrix (ECM). With 1,500 isolated single cell chambers containing ECM, it was demonstrated that ECM support was favorable for some population of cancer cells to maintain stemness and develop drug resistance. This result suggested the importance of drug screening at single-cell resolution in TME-mimicking platforms. Secondly, a drug combination screening chip enabling high-throughput and scalable combinatorial drug screening was demonstrated for the aggregated sphere model. Instead of screening a single drug on each of the tumors, this chip allows the screening of all pairwise drug combinations from eight different cancer drugs, in total 172 different treatment conditions, and 1,032 tested samples in a single microfluidic chip. The presented design approach was easily scalable to incorporate arbitrary number of drugs for large-scale drug screening. Finally, single-cell Hi-Sphere chip enabled high-throughput clonal sphere culture and selective retrieval. Combining fluorescent dye on-situ staining techniques, we identified rare cancer stem-like cell population and confirms its location at the leading edge of spheres. Advance in experimental throughput generates massive data, which demands the corresponding automatic analysis and intelligent interpretation capabilities. The second part of this dissertation focuses on the applications of computer vision and machine learning algorithms to automated biomedical data processing. Image analysis with convolutional neural network was applied for drug efficacy evaluation in a fast and label-free manner. The estimated drug efficacy is highly correlated with the experimental ground truth (R-value > 0.93), while the predicted half-maximal inhibitory concentration is within 8% error range. In addition, metastatic fast-moving cells could be identified after extracting morphological features from the microscope images and applying deep learning algorithm for image analysis, achieving over 99% accuracy for cell movement direction prediction and 91% for speed prediction. In summary, this dissertation presents high-throughput TME-mimicking microfluidics and deep learning image analysis for large-scale drug screening solutions.PHDElectrical EngineeringUniversity of Michigan, Horace H. Rackham School of Graduate Studieshttp://deepblue.lib.umich.edu/bitstream/2027.42/163039/1/zhangzx_1.pd

    The landscape of combination therapies against glioblastoma:From promises to challenges

    Get PDF
    We demonstrate in this thesis how new targets can be identified and highlight the challenges that lie in front of us when trying to translate these steps toward the clinic. We conclude that the blood-brain barrier, PD/PK of drugs, and therapy resistance are still major challenges and explain the limited improvement in treatment options for patients with GBM. First, GBM is a diffuse glioma located in the brain where the blood-brain barrier prevents the crossing of drugs and thereby limits the efficacy of treatment. Second, inter- and intratumoral heterogeneity have been observed in GBM leading to different cellular subpopulations with distinctive genetic profiles. Hence, treating these subpopulations with targeted drugs allows until now still survival of certain subpopulations that are not sensitive to this treatment. Lastly, therapy resistance is often seen in GBM patients and is probably related to intratumoral heterogeneity, but the intrinsic molecular mechanism is still not fully understood. Together they lead to the inevitable recurrence of the tumor

    Glioblastoma and the search for non-hypothesis driven combination therapeutics in academia

    Get PDF
    Glioblastoma (GBM) remains a cancer of high unmet clinical need. Current standard of care for GBM, consisting of maximal surgical resection, followed by ionisation radiation (IR) plus concomitant and adjuvant temozolomide (TMZ), provides less than 15-month survival benefit. Efforts by conventional drug discovery to improve overall survival have failed to overcome challenges presented by inherent tumor heterogeneity, therapeutic resistance attributed to GBM stem cells, and tumor niches supporting self-renewal. In this review we describe the steps academic researchers are taking to address these limitations in high throughput screening programs to identify novel GBM combinatorial targets. We detail how they are implementing more physiologically relevant phenotypic assays which better recapitulate key areas of disease biology coupled with more focussed libraries of small compounds, such as drug repurposing, target discovery, pharmacologically active and novel, more comprehensive anti-cancer target-annotated compound libraries. Herein, we discuss the rationale for current GBM combination trials and the need for more systematic and transparent strategies for identification, validation and prioritisation of combinations that lead to clinical trials. Finally, we make specific recommendations to the preclinical, small compound screening paradigm that could increase the likelihood of identifying tractable, combinatorial, small molecule inhibitors and better drug targets specific to GBM.Peer reviewe
    • 

    corecore