Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Bhagwan Yadav is active.

Publication


Featured researches published by Bhagwan Yadav.


Scientific Reports | 2015

Quantitative scoring of differential drug sensitivity for individually optimized anticancer therapies

Bhagwan Yadav; Tea Pemovska; Agnieszka Szwajda; Evgeny Kulesskiy; Mika Kontro; Riikka Karjalainen; Muntasir Mamun Majumder; Disha Malani; Astrid Murumägi; Jonathan Knowles; Kimmo Porkka; Caroline Heckman; Olli Kallioniemi; Krister Wennerberg; Tero Aittokallio

We developed a systematic algorithmic solution for quantitative drug sensitivity scoring (DSS), based on continuous modeling and integration of multiple dose-response relationships in high-throughput compound testing studies. Mathematical model estimation and continuous interpolation makes the scoring approach robust against sources of technical variability and widely applicable to various experimental settings, both in cancer cell line models and primary patient-derived cells. Here, we demonstrate its improved performance over other response parameters especially in a leukemia patient case study, where differential DSS between patient and control cells enabled identification of both cancer-selective drugs and drug-sensitive patient sub-groups, as well as dynamic monitoring of the response patterns and oncogenic driver signals during cancer progression and relapse in individual patient cells ex vivo. An open-source and easily extendable implementation of the DSS calculation is made freely available to support its tailored application to translating drug sensitivity testing results into clinically actionable treatment options.


PLOS Computational Biology | 2013

Target Inhibition Networks: Predicting Selective Combinations of Druggable Targets to Block Cancer Survival Pathways

Jing Tang; Leena Karhinen; Tao Xu; Agnieszka Szwajda; Bhagwan Yadav; Krister Wennerberg; Tero Aittokallio

A recent trend in drug development is to identify drug combinations or multi-target agents that effectively modify multiple nodes of disease-associated networks. Such polypharmacological effects may reduce the risk of emerging drug resistance by means of attacking the disease networks through synergistic and synthetic lethal interactions. However, due to the exponentially increasing number of potential drug and target combinations, systematic approaches are needed for prioritizing the most potent multi-target alternatives on a global network level. We took a functional systems pharmacology approach toward the identification of selective target combinations for specific cancer cells by combining large-scale screening data on drug treatment efficacies and drug-target binding affinities. Our model-based prediction approach, named TIMMA, takes advantage of the polypharmacological effects of drugs and infers combinatorial drug efficacies through system-level target inhibition networks. Case studies in MCF-7 and MDA-MB-231 breast cancer and BxPC-3 pancreatic cancer cells demonstrated how the target inhibition modeling allows systematic exploration of functional interactions between drugs and their targets to maximally inhibit multiple survival pathways in a given cancer type. The TIMMA prediction results were experimentally validated by means of systematic siRNA-mediated silencing of the selected targets and their pairwise combinations, showing increased ability to identify not only such druggable kinase targets that are essential for cancer survival either individually or in combination, but also synergistic interactions indicative of non-additive drug efficacies. These system-level analyses were enabled by a novel model construction method utilizing maximization and minimization rules, as well as a model selection algorithm based on sequential forward floating search. Compared with an existing computational solution, TIMMA showed both enhanced prediction accuracies in cross validation as well as significant reduction in computation times. Such cost-effective computational-experimental design strategies have the potential to greatly speed-up the drug testing efforts by prioritizing those interventions and interactions warranting further study in individual cancer cases.


Journal of Biological Chemistry | 2012

Obatoclax, saliphenylhalamide and gemcitabine inhibit influenza A virus infection

Oxana V. Denisova; Laura Kakkola; Lin Feng; Jakob Stenman; A. Nagaraj; Johanna Lampe; Bhagwan Yadav; Tero Aittokallio; Pasi Kaukinen; Tero Ahola; Olli Vapalahti; Anu Kantele; Janne Tynell; Ilkka Julkunen; Hannimari Kallio-Kokko; Henrik Paavilainen; Veijo Hukkanen; Richard M. Elliott; Jef K. De Brabander; Xavier Saelens; Denis E. Kainov

Background: Novel options should be developed for treatment of IAV infections. Results: Obatoclax, saliphenylhalamide, and gemcitabine target host factors and inhibit IAV and several other viruses infections. Conclusion: These compounds represent potent antiviral agents. Significance: These compounds could be exploited in treatment of severe viral infections. Influenza A viruses (IAVs) infect humans and cause significant morbidity and mortality. Different treatment options have been developed; however, these were insufficient during recent IAV outbreaks. Here, we conducted a targeted chemical screen in human nonmalignant cells to validate known and search for novel host-directed antivirals. The screen validated saliphenylhalamide (SaliPhe) and identified two novel anti-IAV agents, obatoclax and gemcitabine. Further experiments demonstrated that Mcl-1 (target of obatoclax) provides a novel host target for IAV treatment. Moreover, we showed that obatoclax and SaliPhe inhibited IAV uptake and gemcitabine suppressed viral RNA transcription and replication. These compounds possess broad spectrum antiviral activity, although their antiviral efficacies were virus-, cell type-, and species-specific. Altogether, our results suggest that phase II obatoclax, investigational SaliPhe, and FDA/EMEA-approved gemcitabine represent potent antiviral agents.


Computational and structural biotechnology journal | 2015

Searching for Drug Synergy in Complex Dose-Response Landscapes Using an Interaction Potency Model.

Bhagwan Yadav; Krister Wennerberg; Tero Aittokallio; Jing Tang

Rational design of multi-targeted drug combinations is a promising strategy to tackle the drug resistance problem for many complex disorders. A drug combination is usually classified as synergistic or antagonistic, depending on the deviation of the observed combination response from the expected effect calculated based on a reference model of non-interaction. The existing reference models were proposed originally for low-throughput drug combination experiments, which make the model assumptions often incompatible with the complex drug interaction patterns across various dose pairs that are typically observed in large-scale dose–response matrix experiments. To address these limitations, we proposed a novel reference model, named zero interaction potency (ZIP), which captures the drug interaction relationships by comparing the change in the potency of the dose–response curves between individual drugs and their combinations. We utilized a delta score to quantify the deviation from the expectation of zero interaction, and proved that a delta score value of zero implies both probabilistic independence and dose additivity. Using data from a large-scale anticancer drug combination experiment, we demonstrated empirically how the ZIP scoring approach captures the experimentally confirmed drug synergy while keeping the false positive rate at a low level. Further, rather than relying on a single parameter to assess drug interaction, we proposed the use of an interaction landscape over the full dose–response matrix to identify and quantify synergistic and antagonistic dose regions. The interaction landscape offers an increased power to differentiate between various classes of drug combinations, and may therefore provide an improved means for understanding their mechanisms of action toward clinical translation.


Leukemia | 2017

HOX gene expression predicts response to BCL-2 inhibition in acute myeloid leukemia

Mika Kontro; Ashwini Kumar; Muntasir Mamun Majumder; Samuli Eldfors; Alun Parsons; Tea Pemovska; Jani Saarela; Bhagwan Yadav; Disha Malani; Y Fløisand; Martin Höglund; Kari Remes; Bjørn Tore Gjertsen; Olli Kallioniemi; Krister Wennerberg; Caroline Heckman; K Porkka

Inhibitors of B-cell lymphoma-2 (BCL-2) such as venetoclax (ABT-199) and navitoclax (ABT-263) are clinically explored in several cancer types, including acute myeloid leukemia (AML), to selectively induce apoptosis in cancer cells. To identify robust biomarkers for BCL-2 inhibitor sensitivity, we evaluated the ex vivo sensitivity of fresh leukemic cells from 73 diagnosed and relapsed/refractory AML patients, and then comprehensively assessed whether the responses correlated to specific mutations or gene expression signatures. Compared with samples from healthy donor controls (nonsensitive) and chronic lymphocytic leukemia (CLL) patients (highly sensitive), AML samples exhibited variable responses to BCL-2 inhibition. Strongest CLL-like responses were observed in 15% of the AML patient samples, whereas 32% were resistant, and the remaining exhibited intermediate responses to venetoclax. BCL-2 inhibitor sensitivity was associated with genetic aberrations in chromatin modifiers, WT1 and IDH1/IDH2. A striking selective overexpression of specific HOXA and HOXB gene transcripts were detected in highly BCL-2 inhibitor sensitive samples. Ex vivo responses to venetoclax showed significant inverse correlation to β2-microglobulin expression and to a lesser degree to BCL-XL and BAX expression. As new therapy options for AML are urgently needed, the specific HOX gene expression pattern can potentially be used as a biomarker to identify venetoclax-sensitive AML patients for clinical trials.


Cell Death and Disease | 2013

Anticancer compound ABT-263 accelerates apoptosis in virus-infected cells and imbalances cytokine production and lowers survival rates of infected mice

Laura Kakkola; Oxana V. Denisova; Janne Tynell; Johanna Viiliäinen; Tine Ysenbaert; R. C. Matos; A. Nagaraj; Tiina Öhman; Henrik Paavilainen; Lin Feng; Bhagwan Yadav; Ilkka Julkunen; Olli Vapalahti; Veijo Hukkanen; Jakob Stenman; Tero Aittokallio; Emmy W. Verschuren; Päivi M. Ojala; Tuula A. Nyman; Xavier Saelens; K. Dzeyk; Denis E. Kainov

ABT-263 and its structural analogues ABT-199 and ABT-737 inhibit B-cell lymphoma 2 (Bcl-2), BCL2L1 long isoform (Bcl-xL) and BCL2L2 (Bcl-w) proteins and promote cancer cell death. Here, we show that at non-cytotoxic concentrations, these small molecules accelerate the deaths of non-cancerous cells infected with influenza A virus (IAV) or other viruses. In particular, we demonstrate that ABT-263 altered Bcl-xL interactions with Bcl-2 antagonist of cell death (Bad), Bcl-2-associated X protein (Bax), uveal autoantigen with coiled-coil domains and ankyrin repeats protein (UACA). ABT-263 thereby activated the caspase-9-mediated mitochondria-initiated apoptosis pathway, which, together with the IAV-initiated caspase-8-mediated apoptosis pathway, triggered the deaths of IAV-infected cells. Our results also indicate that Bcl-xL, Bcl-2 and Bcl-w interact with pattern recognition receptors (PRRs) that sense virus constituents to regulate cellular apoptosis. Importantly, premature killing of IAV-infected cells by ABT-263 attenuated the production of key pro-inflammatory and antiviral cytokines. The imbalance in cytokine production was also observed in ABT-263-treated IAV-infected mice, which resulted in an inability of the immune system to clear the virus and eventually lowered the survival rates of infected animals. Thus, the results suggest that the chemical inhibition of Bcl-xL, Bcl-2 and Bcl-w could potentially be hazardous for cancer patients with viral infections.


Molecular Cancer | 2016

Identification of selective cytotoxic and synthetic lethal drug responses in triple negative breast cancer cells

Prson Gautam; Leena Karhinen; Agnieszka Szwajda; Sawan Kumar Jha; Bhagwan Yadav; Tero Aittokallio; Krister Wennerberg

BackgroundTriple negative breast cancer (TNBC) is a highly heterogeneous and aggressive type of cancer that lacks effective targeted therapy. Despite detailed molecular profiling, no targeted therapy has been established. Hence, with the aim of gaining deeper understanding of the functional differences of TNBC subtypes and how that may relate to potential novel therapeutic strategies, we studied comprehensive anticancer-agent responses among a panel of TNBC cell lines.MethodThe responses of 301 approved and investigational oncology compounds were measured in 16 TNBC cell lines applying a functional profiling approach. To go beyond the standard drug viability effect profiling, which has been used in most chemosensitivity studies, we utilized a multiplexed readout for both cell viability and cytotoxicity, allowing us to differentiate between cytostatic and cytotoxic responses.ResultsOur approach revealed that most single-agent anti-cancer compounds that showed activity for the viability readout had no or little cytotoxic effects. Major compound classes that exhibited this type of response included anti-mitotics, mTOR, CDK, and metabolic inhibitors, as well as many agents selectively inhibiting oncogene-activated pathways. However, within the broad viability-acting classes of compounds, there were often subsets of cell lines that responded by cell death, suggesting that these cells are particularly vulnerable to the tested substance. In those cases we could identify differential levels of protein markers associated with cytotoxic responses. For example, PAI-1, MAPK phosphatase and Notch-3 levels associated with cytotoxic responses to mitotic and proteasome inhibitors, suggesting that these might serve as markers of response also in clinical settings. Furthermore, the cytotoxicity readout highlighted selective synergistic and synthetic lethal drug combinations that were missed by the cell viability readouts. For instance, the MEK inhibitor trametinib synergized with PARP inhibitors. Similarly, combination of two non-cytotoxic compounds, the rapamycin analog everolimus and an ATP-competitive mTOR inhibitor dactolisib, showed synthetic lethality in several mTOR-addicted cell lines.ConclusionsTaken together, by studying the combination of cytotoxic and cytostatic drug responses, we identified a deeper spectrum of cellular responses both to single agents and combinations that may be highly relevant for identifying precision medicine approaches in TNBC as well as in other types of cancers.


Blood | 2017

JAK1/2 and BCL2 inhibitors synergize to counteract bone marrow stromal cell-induced protection of AML

Riikka Karjalainen; Tea Pemovska; Mihaela Popa; Minxia Liu; Komal K. Javarappa; Muntasir Mamun Majumder; Bhagwan Yadav; David Tamborero; Jing Tang; Dmitrii Bychkov; Mika Kontro; Alun Parsons; Minna Suvela; Mireia Mayoral Safont; Kimmo Porkka; Tero Aittokallio; Olli Kallioniemi; Emmet McCormack; Bjørn Tore Gjertsen; Krister Wennerberg; Jonathan Knowles; Caroline Heckman

The bone marrow (BM) provides a protective microenvironment to support the survival of leukemic cells and influence their response to therapeutic agents. In acute myeloid leukemia (AML), the high rate of relapse may in part be a result of the inability of current treatment to effectively overcome the protective influence of the BM niche. To better understand the effect of the BM microenvironment on drug responses in AML, we conducted a comprehensive evaluation of 304 inhibitors, including approved and investigational agents, comparing ex vivo responses of primary AML cells in BM stroma-derived and standard culture conditions. In the stroma-based conditions, the AML patient cells exhibited significantly reduced sensitivity to 12% of the tested compounds, including topoisomerase II, B-cell chronic lymphocytic leukemia/lymphoma 2 (BCL2), and many tyrosine kinase inhibitors (TKIs). The loss of TKI sensitivity was most pronounced in patient samples harboring FLT3 or PDGFRB alterations. In contrast, the stroma-derived conditions enhanced sensitivity to Janus kinase (JAK) inhibitors. Increased cell viability and resistance to specific drug classes in the BM stroma-derived conditions was a result of activation of alternative signaling pathways mediated by factors secreted by BM stromal cells and involved a switch from BCL2 to BCLXL-dependent cell survival. Moreover, the JAK1/2 inhibitor ruxolitinib restored sensitivity to the BCL2 inhibitor venetoclax in AML patient cells ex vivo in different model systems and in vivo in an AML xenograft mouse model. These findings highlight the potential of JAK inhibitors to counteract stroma-induced resistance to BCL2 inhibitors in AML.


Nature | 2016

Consistency in drug response profiling

John Patrick Mpindi; Bhagwan Yadav; Päivi Östling; Prson Gautam; Disha Malani; Astrid Murumägi; Akira Hirasawa; Sara Kangaspeska; Krister Wennerberg; Olli Kallioniemi; Tero Aittokallio

The comparative analysis by Haibe-Kains et al.1 concluded that data from two large-scale studies of cancer cell lines2,3 showed highly discordant results for drug sensitivity measurements, whereas gene expression data were reasonably concordant. Here, we crosscompared the two original datasets2,3 against our own data of drug response profiles in overlapping cancer cell line panels. Our results indicate that it is possible to achieve concordance between different laboratories for drug response measurements by paying attention to the harmonization of assays and experimental procedures. There is a Reply to this Comment by Safikhani, Z. et al. Nature 540, http://dx.doi.org/10.1038/ nature20172 (2016). Haibe-Kains et al.1 reported on a comparative evaluation of two drug sensitivity and molecular profiling datasets, one from the Cancer Genome Project (CGP)2 and the other from the Cancer Cell Line Encyclopedia (CCLE)3. In their analyses, gene expression profiles between hundreds of common cancer cell lines across all genes showed high consistency between the two studies (median rank correlation (MRC) = 0.85), whereas the drug response data for 15 common compounds were highly discordant (MRC = 0.28 for halfmaximum inhibitory concentration (IC50) values). This report1 and the accompanying commentary4 suggested that differences in laboratory protocols, compounds and their tested concentration ranges, and computational methods may account for the differences, but these reports did not elaborate which of these factors are important and whether they can be controlled for. Here, we reanalysed the dose–response data from both CGP and CCLE using a standardized area under the curve (AUC) response metric, which we call the drug sensitivity score (DSS)5. We then compared the CGP and CCLE data with a new dataset of drug responses profiled using the Institute for Molecular Medicine Finland (FIMM) compound testing assay6, covering 308 drugs across 106 cancer cell lines. The FIMM data included 45 compounds in common with CGP and 14 with the CCLE in 50 cell lines (Supplementary Data 1). In the AUC calculation, we unified the drug concentration ranges across the CGP, CCLE and FIMM assays. We observed a significantly higher level of consistency (P = 4.2 × 10−5), especially between the CCLE and FIMM drug response data (MRC = 0.74), as compared to the consistency between FIMM and CGP data (MRC = 0.54) (Fig. 1a). Similar experimental protocols were applied at FIMM and CCLE, including the same readout (CellTiter-Glo, Promega), similar controls (vehicle as negative control and positive controls of toxic compounds 100 μ M benzethonium chloride or 1 μ M MG132). However, there were also differences, such as the plate format used (1,536 versus 384 wells). Importantly, there was no effort made to standardize cell numbers used or any other parameters between the three laboratories, such as the source, passage number and media used for cells, nor the origin and handling of drugs. Therefore, this observed level of drug response agreement could be substantially improved by further standardization of the laboratory protocols. The CGP experimental protocol differed from the two others in terms of the readout (fluorescent nucleic acid stain Syto 60, Life Technologies), in the use of controls (drug-free cells as negative and no cells as positive controls), and the plate format used (96or 384-well plates). We compared the drug response profiles between the same cell lines from different laboratories, in line with the approach of Haibe-Kains et al.1, in which they showed consistency in gene expression profiles from CGP and CCLE (MRC = 0.85)1. The Haibe-Kains et al.1 approach, in which the correlation is calculated for each drug separately across the cell lines, showed more variability (Fig. 1b), owing to the fact that some drugs show minimal efficacy in all the tested cell lines. Analogously, gene expression correlations vary more widely when analysed at the level of genes across cell lines (MRC = 0.58 between CGP and CCLE), as certain genes are not expressed above technical noise. Although both ways to compare the data are relevant to the overall goal of personalized


Leukemia | 2017

Enhanced sensitivity to glucocorticoids in cytarabine-resistant AML.

Disha Malani; Astrid Murumägi; Bhagwan Yadav; Mika Kontro; Samuli Eldfors; Ashwini Kumar; Riikka Karjalainen; Muntasir Mamun Majumder; P Ojamies; Tea Pemovska; Krister Wennerberg; Caroline Heckman; K Porkka; Maija Wolf; Tero Aittokallio; Olli Kallioniemi

We sought to identify drugs that could counteract cytarabine resistance in acute myeloid leukemia (AML) by generating eight resistant variants from MOLM-13 and SHI-1 AML cell lines by long-term drug treatment. These cells were compared with 66 ex vivo chemorefractory samples from cytarabine-treated AML patients. The models and patient cells were subjected to genomic and transcriptomic profiling and high-throughput testing with 250 emerging and clinical oncology compounds. Genomic profiling uncovered deletion of the deoxycytidine kinase (DCK) gene in both MOLM-13- and SHI-1-derived cytarabine-resistant variants and in an AML patient sample. Cytarabine-resistant SHI-1 variants and a subset of chemorefractory AML patient samples showed increased sensitivity to glucocorticoids that are often used in treatment of lymphoid leukemia but not AML. Paired samples taken from AML patients before treatment and at relapse also showed acquisition of glucocorticoid sensitivity. Enhanced glucocorticoid sensitivity was only seen in AML patient samples that were negative for the FLT3 mutation (P=0.0006). Our study shows that development of cytarabine resistance is associated with increased sensitivity to glucocorticoids in a subset of AML, suggesting a new therapeutic strategy that should be explored in a clinical trial of chemorefractory AML patients carrying wild-type FLT3.

Collaboration


Dive into the Bhagwan Yadav's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mika Kontro

University of Helsinki

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge