1,128 research outputs found

    The immune microenvironment in mantle cell lymphoma : Targeted liquid and spatial proteomic analyses

    Get PDF
    The complex interplay of the tumour and immune cells affects tumour growth, progression, and response to treatment. Restorationof effective immune response forms the basis of onco-immunology, which further enabled the development of immunotherapy. Inthe era of precision medicine, pin-pointing patient biological heterogeneity especially in relation to patient-specific immunemicroenvironment is a necessity for the discovery of novel biomarkers and for development of patient stratification tools for targetedtherapeutics. Mantle cell lymphoma (MCL) is a rare and aggressive subtype of B-cell lymphoma with poor survival and high relapserates. Previous investigations of MCL have largely focused on the tumour itself and explorations of the immune microenvironmenthave been limited. This thesis and the included five papers, investigates multiple aspects of the immune microenvironment withrespect to proteomic analysis performed on tissue and liquid biopsies of diagnostic and relapsed/refractory (R/R) MCL cohorts.Analyses based on liquid biopsies (serum) in particular are relevant for aggressive cases such as in relapse, where invasiveprocedures for extracting tissues is not recommended. Thus, paper I-II probes the possibility of using serum for treatment andoutcome-associated biomarker discovery in R/R MCL, using a targeted affinity-based protein microarray platform quantifyingimmune-regulatory and tumor-secretory proteins in sera. Analysis performed in paper I using pre-treatment samples, identifies 11-plex biomarker signature (RIS – relapsed immune signature) associated with overall survival. Further integration of RIS with mantlecell lymphoma international prognostic index (MIPI) led to the development of MIPIris index for the stratification of R/R MCL intothree risk groups. Moreover, longitudinal analysis can be important in understanding how patient respond to treatment and thiscan further guide therapeutic interventions. Thus, paper II is a follow-up study wherein longitudinal analyses was performed onpaired samples collected at pre-treatment (baseline) and after three months of chemo-immunotherapy (on-treatment). We showhow genetic aberrations can influence systemic profiles and thus integrating genetic information can be crucial for treatmentselection. Furthermore, we observe that the inter-patient heterogeneity associated with absolute values can be circumvented byusing velocity of change to capture general changes over time in groups of patients. Thus, using velocity of change in serumproteins between pre- and on-treatment samples identified response biomarkers associated with minimal residual disease andprogression. While exploratory analysis using high dimensional omics-based data can be important for accelerating discovery,translating such information for clinical utility is a necessity. Thus, in paper III, we show how serum quantification can be usedcomplementary tissue-identified prognostic biomarkers and this can enable faster clinical implementation. Presence of CD163+M2-like macrophages has shown to be associated with poor outcome in MCL tissues. We show that higher expression of sCD163levels in sera quantified using ELISA, is also associated with poor outcome in diagnostic and relapsed MCL. Furthermore, wesuggest a cut-off for sCD163 levels that can be used for clinical utility. Further exploration of the dynamic interplay of tumourimmunemicroenvironment is now possible using spatial resolved omics for tissue-based analysis. Thus, in paper IV and V, weanalyse cell-type specific proteomic data collected from tumour and immune cells using GeoMx™ digital spatial profiler. In paperIV, we show that presence as well as spatial localization of CD163+ macrophage with respect to tumour regions impactsmacrophage phenotypic profiles. Further modulation in the profile of surrounding tumour and T-cells is observed whenmacrophages are present in the vicinity. Based on this analysis, we suggest MAPK pathway as a potential therapeutic target intumours with CD163+ macrophages. Immune composition can be defined not just by the type of cells, but also with respect tofrequency and spatial localization and this is explored in paper V with respect to T-cell subtypes. Thus, in paper V, we optimizeda workflow of multiplexed immunofluorescence image segmentation that allowed us to extract cell metrics for four subtypes ofCD3+ T-cells. Using this data, we show that higher infiltration of T-cells is associated with a positive outcome in MCL. Moreover,by combining image derived metrics to cell specific spatial omics data, we were able to identify immunosuppressivemicroenvironment associated with highly infiltrated tumours and suggests new potential targets of immunotherapy with respect toIDO1, GITR and STING. In conclusion, this thesis explores systemic and tumor-associated immune microenvironment in MCL, fordefining patient heterogeneity, developing methods of patient stratification and for identifying novel and actionable biomarkers

    Developing computational tools for studying cancer metabolism and genomics

    Get PDF
    The interplay between different genomic and epigenomic alterations lead to different prognoses in cancer patients. Advances in high-throughput technologies, like gene expression profiling, next-generation sequencing, proteomics, and fluxomics, have enabled detailed molecular characterization of various tumors, yet studying this interplay is a complex computational problem.Here we set to develop computational approaches to identify and study emerging challenges in cancer metabolism and genomics. We focus on three research questions, addressed by different computational approaches: (1) What is the set of metabolic interactions in cancer metabolism? To this end we generated a computational framework that quantitatively predicts synthetic dosage lethal (SDL) interactions in human metabolism, by developing a new algorithmic-modeling approach. SDLs offer a promising way to selectively kill cancer cells by targeting the SDL partners of activated oncogenes in tumors, which are often difficult to target directly. (2) What is the landscape of metabolic regulation in breast cancer? To this end we established a new framework that utilizes different data types to perform multi-omics data integration and flux prediction, by incorporating machine learn- ing techniques with Genome Scale Metabolic Modeling (GSMM). This enabled us to study the regulation of breast cancer cell line under different growth conditions, from multiple omics data. (3) What is the power of somatic mutations derived from RNA in estimating the tumor mutational burden? Here we develop a new tool to detect somatic mutations from RNA sequencing data without a matched- normal sample. To this end we developed a machine learning pipeline that takes as input a list of single nucleotide variants and classifies them as either somatic or germline, based on read-level features as well as position-specific variant statistics and common germline databases. We showed that detecting somatic mutations directly from RNA enables the identification of expressed mutations, and therefore represent a more relevant metric in estimating the tumor mutational burden, which is significantly associated with patient survival. In sum, my work has been focused around developing computational methods to tackle different research questions in cancer metabolism and genomics, utilizing various types of omics data and a variety of computational approaches. These methods provide new solutions to some important computational challenges, and their applications help to generate promising leads for cancer research, and can be utilized in many future applications, analyzing novel and existing datasets

    The role of network science in glioblastoma

    Get PDF
    Network science has long been recognized as a well-established discipline across many biological domains. In the particular case of cancer genomics, network discovery is challenged by the multitude of available high-dimensional heterogeneous views of data. Glioblastoma (GBM) is an example of such a complex and heterogeneous disease that can be tackled by network science. Identifying the architecture of molecular GBM networks is essential to understanding the information flow and better informing drug development and pre-clinical studies. Here, we review network-based strategies that have been used in the study of GBM, along with the available software implementations for reproducibility and further testing on newly coming datasets. Promising results have been obtained from both bulk and single-cell GBM data, placing network discovery at the forefront of developing a molecularly-informed-based personalized medicine.This work was partially supported by national funds through Fundação para a Ciência e a Tecnologia (FCT) with references CEECINST/00102/2018, CEECIND/00072/2018 and PD/BDE/143154/2019, UIDB/04516/2020, UIDB/00297/2020, UIDB/50021/2020, UIDB/50022/2020, UIDB/50026/2020, UIDP/50026/2020, NORTE-01-0145-FEDER-000013, and NORTE-01-0145-FEDER000023 and projects PTDC/CCI-BIO/4180/2020 and DSAIPA/DS/0026/2019. This project has received funding from the European Union’s Horizon 2020 research and innovation program under Grant Agreement No. 951970 (OLISSIPO project)

    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

    Incorporating Prior Knowledge in Deep Learning Models via Pathway Activity Autoencoders

    Full text link
    Motivation: Despite advances in the computational analysis of high-throughput molecular profiling assays (e.g. transcriptomics), a dichotomy exists between methods that are simple and interpretable, and ones that are complex but with lower degree of interpretability. Furthermore, very few methods deal with trying to translate interpretability in biologically relevant terms, such as known pathway cascades. Biological pathways reflecting signalling events or metabolic conversions are Small improvements or modifications of existing algorithms will generally not be suitable, unless novel biological results have been predicted and verified. Determining which pathways are implicated in disease and incorporating such pathway data as prior knowledge may enhance predictive modelling and personalised strategies for diagnosis, treatment and prevention of disease. Results: We propose a novel prior-knowledge-based deep auto-encoding framework, PAAE, together with its accompanying generative variant, PAVAE, for RNA-seq data in cancer. Through comprehensive comparisons among various learning models, we show that, despite having access to a smaller set of features, our PAAE and PAVAE models achieve better out-of-set reconstruction results compared to common methodologies. Furthermore, we compare our model with equivalent baselines on a classification task and show that they achieve better results than models which have access to the full input gene set. Another result is that using vanilla variational frameworks might negatively impact both reconstruction outputs as well as classification performance. Finally, our work directly contributes by providing comprehensive interpretability analyses on our models on top of improving prognostication for translational medicine

    The Translational Status of Cancer Liquid Biopsies

    Get PDF
    Precision oncology aims to tailor clinical decisions specifically to patients with the objective of improving treatment outcomes. This can be achieved by leveraging omics information for accurate molecular characterization of tumors. Tumor tissue biopsies are currently the main source of information for molecular profiling. However, biopsies are invasive and limited in resolving spatiotemporal heterogeneity in tumor tissues. Alternative non-invasive liquid biopsies can exploit patient’s body fluids to access multiple layers of tumor-specific biological information (genomes, epigenomes, transcriptomes, proteomes, metabolomes, circulating tumor cells, and exosomes). Analysis and integration of these large and diverse datasets using statistical and machine learning approaches can yield important insights into tumor biology and lead to discovery of new diagnostic, predictive, and prognostic biomarkers. Translation of these new diagnostic tools into standard clinical practice could transform oncology, as demonstrated by a number of liquid biopsy assays already entering clinical use. In this review, we highlight successes and challenges facing the rapidly evolving field of cancer biomarker research. Lay Summary: Precision oncology aims to tailor clinical decisions specifically to patients with the objective of improving treatment outcomes. The discovery of biomarkers for precision oncology has been accelerated by high-throughput experimental and computational methods, which can inform fine-grained characterization of tumors for clinical decision-making. Moreover, advances in the liquid biopsy field allow non-invasive sampling of patient’s body fluids with the aim of analyzing circulating biomarkers, obviating the need for invasive tumor tissue biopsies. In this review, we highlight successes and challenges facing the rapidly evolving field of liquid biopsy cancer biomarker research

    Knowledge Management Approaches for predicting Biomarker and Assessing its Impact on Clinical Trials

    Get PDF
    The recent success of companion diagnostics along with the increasing regulatory pressure for better identification of the target population has created an unprecedented incentive for the drug discovery companies to invest into novel strategies for stratified biomarker discovery. Catching with this trend, trials with stratified biomarker in drug development have quadrupled in the last decade but represent a small part of all Interventional trials reflecting multiple co-developmental challenges of therapeutic compounds and companion diagnostics. To overcome the challenge, varied knowledge management and system biology approaches are adopted in the clinics to analyze/interpret an ever increasing collection of OMICS data. By semi-automatic screening of more than 150,000 trials, we filtered trials with stratified biomarker to analyse their therapeutic focus, major drivers and elucidated the impact of stratified biomarker programs on trial duration and completion. The analysis clearly shows that cancer is the major focus for trials with stratified biomarker. But targeted therapies in cancer require more accurate stratification of patient population. This can be augmented by a fresh approach of selecting a new class of biomolecules i.e. miRNA as candidate stratification biomarker. miRNA plays an important role in tumorgenesis in regulating expression of oncogenes and tumor suppressors; thus affecting cell proliferation, differentiation, apoptosis, invasion, angiogenesis. miRNAs are potential biomarkers in different cancer. However, the relationship between response of cancer patients towards targeted therapy and resulting modifications of the miRNA transcriptome in pathway regulation is poorly understood. With ever-increasing pathways and miRNA-mRNA interaction databases, freely available mRNA and miRNA expression data in multiple cancer therapy have created an unprecedented opportunity to decipher the role of miRNAs in early prediction of therapeutic efficacy in diseases. We present a novel SMARTmiR algorithm to predict the role of miRNA as therapeutic biomarker for an anti-EGFR monoclonal antibody i.e. cetuximab treatment in colorectal cancer. The application of an optimised and fully automated version of the algorithm has the potential to be used as clinical decision support tool. Moreover this research will also provide a comprehensive and valuable knowledge map demonstrating functional bimolecular interactions in colorectal cancer to scientific community. This research also detected seven miRNA i.e. hsa-miR-145, has-miR-27a, has- miR-155, hsa-miR-182, hsa-miR-15a, hsa-miR-96 and hsa-miR-106a as top stratified biomarker candidate for cetuximab therapy in CRC which were not reported previously. Finally a prospective plan on future scenario of biomarker research in cancer drug development has been drawn focusing to reduce the risk of most expensive phase III drug failures
    • …
    corecore