Bio


I'm Joy, currently an Instructor in the lab of Prof. Hiro Nakauchi. My academic journey began with an undergraduate degree in Genetic Engineering from India, leading me to further studies with an M.S. and a Ph.D. in Sweden. I then went to Prof Nakauchi's lab at the University of Tokyo, Japan to familiarize myself with stem cell biology before joining his Stanford lab.

Under Prof. Jonas Nilsson's mentorship during my Ph.D., I explored the molecular intricacies of cancer. This helped me identify strategies that could potentially prevent pluripotent stem cells from developing malignant characteristics, ensuring their safety in clinical applications.

My ongoing research revolves around three core objectives:

1. Defining Pluripotency in Mammals: Our work has led to the identification of specific conditions that sustain pluripotency across species without requiring small molecules or cytokines.

2. Overcoming Xeno-barriers in Organ Development: By employing genome-wide perturbation techniques, we're working to identify and bypass barriers that hinder the effective integration of xenogeneic donor cells. This is crucial for generating complete organs in research animals.

3. Evolving Organ Preservation Methods: Drawing inspiration from natural processes, we are on the path to formulating innovative ex vivo organ preservation techniques and thereby, enhancing the success rates of transplantations.

Honors & Awards


  • International Postdoc Grant (2017-0034, Spring 2017), Swedish Research Council (June 2017)
  • PhD thesis of the year 2016 for Institute of Clinical Sciences, University of Gothenburg, The Sahlgrenska Academy, University of Gothenburg , Sweden (May 2017)
  • Assar Gabrielssons Best PhD Thesis Award 2017 in Experimental Research, Stiftelsen Assar Gabrielssons Fond, Sweden (May 2017)

Current Research and Scholarly Interests


My goal is to generate universally transplantable human organs in research animals.

All Publications


  • Age-related changes in the hematopoietic stem cell pool revealed via quantifying the balance of symmetric and asymmetric divisions. PloS one Kawahigashi, T., Iwanami, S., Takahashi, M., Bhadury, J., Iwami, S., Yamazaki, S. 2024; 19 (1): e0292575

    Abstract

    Hematopoietic stem cells (HSCs) are somatic stem cells that continuously generate lifelong supply of blood cells through a balance of symmetric and asymmetric divisions. It is well established that the HSC pool increases with age. However, not much is known about the underlying cause for these observed changes. Here, using a novel method combining single-cell ex vivo HSC expansion with mathematical modeling, we quantify HSC division types (stem cell-stem cell (S-S) division, stem cell-progenitor cell (S-P) division, and progenitor cell-progenitor cell (P-P) division) as a function of the aging process. Our time-series experiments reveal how changes in these three modes of division can explain the increase in HSC numbers with age. Contrary to the popular notion that HSCs divide predominantly through S-P divisions, we show that S-S divisions are predominant throughout the lifespan of the animal, thereby expanding the HSC pool. We, therefore, provide a novel mathematical model-based experimental validation for reflecting HSC dynamics in vivo.

    View details for DOI 10.1371/journal.pone.0292575

    View details for PubMedID 38285676

  • Secreted Particle Information Transfer (SPIT) - A Cellular Platform forIn VivoGenetic Engineering. bioRxiv : the preprint server for biology Charlesworth, C. T., Homma, S., Suchy, F., Wang, S., Bhadhury, J., Amaya, A. K., Camarena, J., Zhang, J., Tan, T. K., Igarishi, K., Nakauchi, H. 2024

    Abstract

    A multitude of tools now exist that allow us to precisely manipulate the human genome in a myriad of different ways. However, successful delivery of these tools to the cells of human patients remains a major barrier to their clinical implementation. Here we introduce a new cellular approach for in vivo genetic engineering, Secreted Particle Information Transfer (SPIT) that utilizes human cells as delivery vectors for in vivo genetic engineering. We demonstrate the application of SPIT for cell-cell delivery of Cre recombinase and CRISPR-Cas9 enzymes, we show that genetic logic can be incorporated into SPIT and present the first demonstration of human cells as a delivery platform for in vivo genetic engineering in immunocompetent mice. We successfully applied SPIT to genetically modify multiple organs and tissue stem cells in vivo including the liver, spleen, intestines, peripheral blood, and bone marrow. We anticipate that by harnessing the large packaging capacity of a human cell's nucleus, the ability of human cells to engraft into patients' long term and the capacity of human cells for complex genetic programming, that SPIT will become a paradigm shifting approach for in vivo genetic engineering.

    View details for DOI 10.1101/2024.01.11.575257

    View details for PubMedID 38260654

  • Physioxia improves the selectivity of hematopoietic stem cell expansion cultures. Blood advances Igarashi, K. J., Kucinski, I., Chan, Y. Y., Tan, T., Khoo, H. M., Kealy, D., Bhadury, J., Hsu, I., Ho, P. Y., Niizuma, K., Hickey, J. W., Nolan, G., Bridge, K. S., Czechowicz, A., Gottgens, B., Nakauchi, H., Wilkinson, A. C. 2023

    Abstract

    Hematopoietic stem cells (HSCs) are a rare hematopoietic cell type that can entirely reconstitute the blood and immune systems following transplantation. Allogeneic HSC transplantation (HSCT) is used clinically as a curative therapy for a range of hematolymphoid diseases, but remains a high-risk therapy due to potential side effects including poor graft function and graft-vs-host disease (GvHD). Ex vivo HSC expansion has been suggested as an approach to improve hematopoietic reconstitution from low-cell dose grafts. Here, we demonstrate that we can improve the selectivity of polyvinyl alcohol (PVA)-based mouse HSC cultures through the use of physioxic culture conditions. Single-cell transcriptomic analysis confirmed inhibition of lineage-committed progenitor cells in physioxic cultures. Long-term physioxic expansion also afforded culture-based ex vivo HSC selection from whole bone marrow, spleen, and embryonic tissues. Furthermore, we provide evidence that HSC-selective ex vivo cultures deplete GvHD-causing T cells and that this approach can be combined with genotoxic-free antibody-based conditioning HSCT approaches. Our results offer a simple approach to improve PVA-based HSC cultures and the underlying molecular phenotype, as well as highlight the potential translational implications of selective HSC expansion systems for allogeneic HSCT.

    View details for DOI 10.1182/bloodadvances.2023009668

    View details for PubMedID 36809781

  • LARGE-SCALE IN VIVO CRISPR SCREENS IDENTIFY SAGA COMPLEX MEMBERS AS KEY REGULATORS OF HAEMATOPOIESIS Wilkinson, A., Haney, M., Shankar, A., Hsu, I., Miyauchi, M., Palovics, R., Olender, L., Khoo, H., Igarashi, K., Bhadury, J., Munson, C., Mack, P., Tan, T., Nakauchi, H., Wyss-Coray, T. ELSEVIER SCIENCE INC. 2023: S43
  • Chimpanzee and pig-tailed macaque iPSCs: Improved culture and generation of primate cross-species embryos. Cell reports Roodgar, M., Suchy, F. P., Nguyen, L. H., Bajpai, V. K., Sinha, R., Vilches-Moure, J. G., Van Bortle, K., Bhadury, J., Metwally, A., Jiang, L., Jian, R., Chiang, R., Oikonomopoulos, A., Wu, J. C., Weissman, I. L., Mankowski, J. L., Holmes, S., Loh, K. M., Nakauchi, H., VandeVoort, C. A., Snyder, M. P. 2022; 40 (9): 111264

    Abstract

    As our closest living relatives, non-human primates uniquely enable explorations of human health, disease, development, and evolution. Considerable effort has thus been devoted to generating induced pluripotent stem cells (iPSCs) from multiple non-human primate species. Here, we establish improved culture methods for chimpanzee (Pan troglodytes) and pig-tailed macaque (Macaca nemestrina) iPSCs. Such iPSCs spontaneously differentiate in conventional culture conditions, but can be readily propagated by inhibiting endogenous WNT signaling. As a unique functional test of these iPSCs, we injected them into the pre-implantation embryos of another non-human species, rhesus macaques (Macaca mulatta). Ectopic expression of gene BCL2 enhances the survival and proliferation of chimpanzee and pig-tailed macaque iPSCs within the pre-implantation embryo, although the identity and long-term contribution of the transplanted cells warrants further investigation. In summary, we disclose transcriptomic and proteomic data, cell lines, and cell culture resources that may be broadly enabling for non-human primate iPSCs research.

    View details for DOI 10.1016/j.celrep.2022.111264

    View details for PubMedID 36044843

  • Streamlined and quantitative detection of chimerism using digital PCR. Scientific reports Suchy, F. P., Nishimura, T., Seki, S., Wilkinson, A. C., Higuchi, M., Hsu, I., Zhang, J., Bhadury, J., Nakauchi, H. 2022; 12 (1): 10223

    Abstract

    Animal chimeras are widely used for biomedical discoveries, from developmental biology to cancer research. However, the accurate quantitation of mixed cell types in chimeric and mosaic tissues is complicated by sample preparation bias, transgenic silencing, phenotypic similarity, and low-throughput analytical pipelines. Here, we have developed and characterized a droplet digital PCR single-nucleotide discrimination assay to detect chimerism among common albino and non-albino mouse strains. In addition, we validated that this assay is compatible with crude lysate from all solid organs, drastically streamlining sample preparation. This chimerism detection assay has many additional advantages over existing methods including its robust nature, minimal technical bias, and ability to report the total number of cells in a prepared sample. Moreover, the concepts discussed here are readily adapted to other genomic loci to accurately measure mixed cell populations in any tissue.

    View details for DOI 10.1038/s41598-022-14467-5

    View details for PubMedID 35715477

  • Generating human artery and vein cells from pluripotent stem cells highlights the arterial tropism of Nipah and Hendra viruses. Cell Ang, L. T., Nguyen, A. T., Liu, K. J., Chen, A., Xiong, X., Curtis, M., Martin, R. M., Raftry, B. C., Ng, C. Y., Vogel, U., Lander, A., Lesch, B. J., Fowler, J. L., Holman, A. R., Chai, T., Vijayakumar, S., Suchy, F. P., Nishimura, T., Bhadury, J., Porteus, M. H., Nakauchi, H., Cheung, C., George, S. C., Red-Horse, K., Prescott, J. B., Loh, K. M. 2022

    Abstract

    Stem cell research endeavors to generate specific subtypes of classically defined "cell types." Here, we generate >90% pure human artery or vein endothelial cells from pluripotent stem cells within 3-4 days. We specified artery cells by inhibiting vein-specifying signals and vice versa. These cells modeled viral infection of human vasculature by Nipah and Hendra viruses, which are extraordinarily deadly (∼57%-59% fatality rate) and require biosafety-level-4 containment. Generating pure populations of artery and vein cells highlighted that Nipah and Hendra viruses preferentially infected arteries; arteries expressed higher levels of their viral-entry receptor. Virally infected artery cells fused into syncytia containing up to 23 nuclei, which rapidly died. Despite infecting arteries and occupying ∼6%-17% of their transcriptome, Nipah and Hendra largely eluded innate immune detection, minimally eliciting interferon signaling. We thus efficiently generate artery and vein cells, introduce stem-cell-based toolkits for biosafety-level-4 virology, and explore the arterial tropism and cellular effects of Nipah and Hendra viruses.

    View details for DOI 10.1016/j.cell.2022.05.024

    View details for PubMedID 35738284

  • Generation of Functional Organs Using a Cell-Competitive Niche in Intra- and Inter-species Rodent Chimeras. Cell stem cell Nishimura, T., Suchy, F. P., Bhadury, J., Igarashi, K. J., Charlesworth, C. T., Nakauchi, H. 2020

    Abstract

    Interspecies organ generation via blastocyst complementation has succeeded in rodents, but not yet in evolutionally more distant species. Early developmental arrest hinders the formation of highly chimeric fetuses. We demonstrate that the deletion of insulin-like growth factor 1 receptor (Igf1r) in mouse embryos creates a permissive "cell-competitive niche" in several organs, significantly augmenting both mouse intraspecies and mouse/rat interspecies donor chimerism that continuously increases from embryonic day 11 onward, sometimes even taking over entire organs within intraspecies chimeras. Since Igf1r deletion allows the evasion of early developmental arrest, interspecies fetuses with high levels of organ chimerism can be generated via blastocyst complementation. This observation should facilitate donor cell contribution to host tissues, resulting in whole-organ generation via blastocyst complementation across wide evolutionary distances.

    View details for DOI 10.1016/j.stem.2020.11.019

    View details for PubMedID 33373620

  • Activated HoxB4-induced Hematopoietic Stem Cells from Murine Pluripotent Stem Cells via Long-Term Programming. Experimental hematology Izawa, K., Yamazaki, S., Becker, H. J., Bhadury, J., Kakegawa, T., Sakaguchi, M., Tojo, A. 2020

    Abstract

    Hematopoietic stem cells (HSCs) are multipotent cells that form the entire blood system and have the potential to cure several pathogenic conditions directly or indirectly arising from defects within the HSC compartment. Pluripotent stem cells (PSCs) or induced pluripotent stem cells (iPSCs) can give rise to all embryonic cell types, however, efficient in vitro differentiation of HSCs from PSCs remains challenging. HoxB4 is a key regulator orchestrating the differentiation of PSCs into all cells type across the mesodermal lineage, including HSCs. Moreover, the ectopic expression of HoxB4 enhances the in vitro generation and expansion of HSCs. However, several aspects of HoxB4 biology including its regulatory functions are not fully understood. Here, we describe the role of HoxB4 in indirectly inhibiting the emergence of mature CD45+ HSCs from iPSCs in vitro. Forced activation of HoxB4 permitted long-term maintenance of functional hematopoietic stem and progenitor cells (HSPCs), which efficiently reconstituted hematopoiesis upon transplantation. Our method enables an easy and scalable in vitro platform for the generation of HSCs from iPSCs, which will ultimately lead to a better understanding of HSC biology and facilitate in preparing the roadmap for producing an unrestricted supply of HSCs for several curative therapies.

    View details for DOI 10.1016/j.exphem.2020.08.003

    View details for PubMedID 32795499

  • BET bromodomain inhibitors synergize with ATR inhibitors in melanoma in melanoma. Cell death & disease Muralidharan, S. V., Einarsdottir, B. O., Bhadury, J., Lindberg, M. F., Wu, J., Campeau, E., Bagge, R. O., Stierner, U., Ny, L., Nilsson, L. M., Nilsson, J. A. 2017; 8 (8): e2982

    Abstract

    Metastatic malignant melanoma continues to be a challenging disease despite clinical translation of the comprehensive understanding of driver mutations and how melanoma cells evade immune attack. In Myc-driven lymphoma, efficacy of epigenetic inhibitors of the bromodomain and extra-terminal domain (BET) family of bromodomain proteins can be enhanced by combination therapy with inhibitors of the DNA damage response kinase ATR. Whether this combination is active in solid malignancies like melanoma, and how it relates to immune therapy, has not previously investigated. To test efficacy and molecular consequences of combination therapies cultured melanoma cells were used. To assess tumor responses to therapies in vivo we use patient-derived xenografts and B6 mice transplanted with B16F10 melanoma cells. Concomitant inhibition of BET proteins and ATR of cultured melanoma cells resulted in similar effects as recently shown in lymphoma, such as induction of apoptosis and p62, implicated in autophagy, senescence-associated secretory pathway and ER stress. In vivo, apoptosis and suppression of subcutaneous growth of patient-derived melanoma and B16F10 cells were observed. Our data suggest that ATRI/BETI combination therapies are effective in melanoma.

    View details for DOI 10.1038/cddis.2017.383

    View details for PubMedID 28796244

    View details for PubMedCentralID PMC5596569

  • Global analysis of somatic structural genomic alterations and their impact on gene expression in diverse human cancers PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Alaei-Mahabadi, B., Bhadury, J., Karlsson, J. W., Nilsson, J. A., Larsson, E. 2016; 113 (48): 13768-13773

    Abstract

    Tumor genomes are mosaics of somatic structural variants (SVs) that may contribute to the activation of oncogenes or inactivation of tumor suppressors, for example, by altering gene copy number amplitude. However, there are multiple other ways in which SVs can modulate transcription, but the general impact of such events on tumor transcriptional output has not been systematically determined. Here we use whole-genome sequencing data to map SVs across 600 tumors and 18 cancers, and investigate the relationship between SVs, copy number alterations (CNAs), and mRNA expression. We find that 34% of CNA breakpoints can be clarified structurally and that most amplifications are due to tandem duplications. We observe frequent swapping of strong and weak promoters in the context of gene fusions, and find that this has a measurable global impact on mRNA levels. Interestingly, several long noncoding RNAs were strongly activated by this mechanism. Additionally, SVs were confirmed in telomere reverse transcriptase (TERT) upstream regions in several cancers, associated with elevated TERT mRNA levels. We also highlight high-confidence gene fusions supported by both genomic and transcriptomic evidence, including a previously undescribed paired box 8 (PAX8)-nuclear factor, erythroid 2 like 2 (NFE2L2) fusion in thyroid carcinoma. In summary, we combine SV, CNA, and expression data to provide insights into the structural basis of CNAs as well as the impact of SVs on gene expression in tumors.

    View details for DOI 10.1073/pnas.1606220113

    View details for Web of Science ID 000388835700070

    View details for PubMedID 27856756

    View details for PubMedCentralID PMC5137778

  • BET bromodomain inhibitors synergize with ATR inhibitors to induce DNA damage, apoptosis, senescence-associated secretory pathway and ER stress in Myc-induced lymphoma cells ONCOGENE Muralidharan, S. V., Bhadury, J., Nilsson, L. M., Green, L. C., McLure, K. G., Nilsson, J. A. 2016; 35 (36): 4689-4697

    Abstract

    Inhibiting the bromodomain and extra-terminal (BET) domain family of epigenetic reader proteins has been shown to have potent anti-tumoral activity, which is commonly attributed to suppression of transcription. In this study, we show that two structurally distinct BET inhibitors (BETi) interfere with replication and cell cycle progression of murine Myc-induced lymphoma cells at sub-lethal concentrations when the transcriptome remains largely unaltered. This inhibition of replication coincides with a DNA-damage response and enhanced sensitivity to inhibitors of the upstream replication stress sensor ATR in vitro and in mouse models of B-cell lymphoma. Mechanistically, ATR and BETi combination therapy cause robust transcriptional changes of genes involved in cell death, senescence-associated secretory pathway, NFkB signaling and ER stress. Our data reveal that BETi can potentiate the cell stress and death caused by ATR inhibitors. This suggests that ATRi can be used in combination therapies of lymphomas without the use of genotoxic drugs.

    View details for DOI 10.1038/onc.2015.521

    View details for Web of Science ID 000383324700002

    View details for PubMedID 26804177

  • Hypoxia-regulated gene expression explains differences between melanoma cell line-derived xenografts and patient-derived xenografts ONCOTARGET Bhadury, J., Einarsdottir, B. O., Podraza, A., Bagge, R. O., Stierner, U., Ny, L., Lopez, M. D., Nilsson, J. A. 2016; 7 (17): 23801-23811

    Abstract

    Cell line-derived xenografts (CDXs) are an integral part of drug efficacy testing during development of new pharmaceuticals against cancer but their accuracy in predicting clinical responses in patients have been debated. Patient-derived xenografts (PDXs) are thought to be more useful for predictive biomarker identification for targeted therapies, including in metastatic melanoma, due to their similarities to human disease. Here, tumor biopsies from fifteen patients and ten widely-used melanoma cell lines were transplanted into immunocompromised mice to generate PDXs and CDXs, respectively. Gene expression profiles generated from the tumors of these PDXs and CDXs clustered into distinct groups, despite similar mutational signatures. Hypoxia-induced gene signatures and overexpression of the hypoxia-regulated miRNA hsa-miR-210 characterized CDXs. Inhibition of hsa-miR-210 with decoys had little phenotypic effect in vitro but reduced sensitivity to MEK1/2 inhibition in vivo, suggesting down-regulation of this miRNA could result in development of resistance to MEK inhibitors.

    View details for DOI 10.18632/oncotarget.8181

    View details for Web of Science ID 000377706200062

    View details for PubMedID 27009863

    View details for PubMedCentralID PMC5029664

  • Cancer Differentiating Agent Hexamethylene Bisacetamide Inhibits BET Bromodomain Proteins CANCER RESEARCH Nilsson, L. M., Green, L. C., Muralidharan, S. V., Demir, D., Welin, M., Bhadury, J., Logan, D. T., Walse, B., Nilsson, J. A. 2016; 76 (8): 2376-2383

    Abstract

    Agents that trigger cell differentiation are highly efficacious in treating certain cancers, but such approaches are not generally effective in most malignancies. Compounds such as DMSO and hexamethylene bisacetamide (HMBA) have been used to induce differentiation in experimental systems, but their mechanisms of action and potential range of uses on that basis have not been developed. Here, we show that HMBA, a compound first tested in the oncology clinic over 25 years ago, acts as a selective bromodomain inhibitor. Biochemical and structural studies revealed an affinity of HMBA for the second bromodomain of BET proteins. Accordingly, both HMBA and the prototype BET inhibitor JQ1 induced differentiation of mouse erythroleukemia cells. As expected of a BET inhibitor, HMBA displaced BET proteins from chromatin, caused massive transcriptional changes, and triggered cell-cycle arrest and apoptosis in Myc-induced B-cell lymphoma cells. Furthermore, HMBA exerted anticancer effects in vivo in mouse models of Myc-driven B-cell lymphoma. This study illuminates the function of an early anticancer agent and suggests an intersection with ongoing clinical trials of BET inhibitor, with several implications for predicting patient selection and response rates to this therapy and starting points for generating BD2-selective BET inhibitors. Cancer Res; 76(8); 2376-83. ©2016 AACR.

    View details for DOI 10.1158/0008-5472.CAN-15-2721

    View details for Web of Science ID 000374170700032

    View details for PubMedID 26941288

  • Small RNA deep sequencing discriminates subsets of extracellular vesicles released by melanoma cells - Evidence of unique microRNA cargos RNA BIOLOGY Lunavat, T. R., Cheng, L., Kim, D., Bhadury, J., Jang, S. C., Lasser, C., Sharples, R. A., Lopez, M. D., Nilsson, J., Gho, Y. S., Hill, A. F., Lotvall, J. 2015; 12 (8): 810-823

    Abstract

    Melanoma cells release different types of extracellular vesicles (EVs) into the extracellular milieu that are involved with communication and signaling in the tumor microenvironment. Subsets of EVs include exosomes, microvesicles, and apoptotic bodies that carry protein and genetic (RNA) cargos. To define the contribution of the RNA cargo of melanoma cell derived EVs we performed small RNA sequencing to identify different small RNAs in the EV subsets. Using validated centrifugation protocols, we separated these EV subsets released by the melanoma cell line MML-1, and performed RNA sequencing with the Ion Torrent platform. Various, but different, non-coding RNAs were detected in the EV subsets, including microRNA, mitochondrial associated tRNA, small nucleolar RNA, small nuclear RNA, Ro associated Y-RNA, vault RNA and Y-RNA. We identified in total 1041 miRNAs in cells and EV subsets. Hierarchical clustering showed enrichment of specific miRNAs in exosomes, including hsa-miR-214-3p, hsa-miR-199a-3p and hsa-miR-155-5p, all being associated with melanoma progression. Comparison of exosomal miRNAs with miRNAs in clinical melanoma samples indicate that multiple miRNAs in exosomes also are expressed specifically in melanoma tissues, but not in benign naevi. This study shows for the first time the presence of distinct small RNAs in subsets of EVs released by melanoma cells, with significant similarities to clinical melanoma tissue, and provides unique insights into the contribution of EV associated extracellular RNA in cancer.

    View details for DOI 10.1080/15476286.2015.1056975

    View details for Web of Science ID 000359659700006

    View details for PubMedID 26176991

    View details for PubMedCentralID PMC4615768

  • Melanoma patient-derived xenografts accurately model the disease and develop fast enough to guide treatment decisions ONCOTARGET Einarsdottir, B. O., Bagge, R. O., Bhadury, J., Jespersen, H., Mattsson, J., Nilsson, L. M., Truve, K., Lopez, M. D., Naredi, P., Nilsson, O., Stierner, U., Ny, L., Nilsson, J. A. 2014; 5 (20): 9609-9618

    Abstract

    The development of novel therapies against melanoma would benefit from individualized tumor models to ensure the rapid and accurate identification of biomarkers of therapy response. Previous studies have suggested that patient-derived xenografts (PDXes) could be useful. However, the utility of PDXes in guiding real-time treatment decisions has only been reported in anecdotal forms. Here tumor biopsies from patients with stage III and IV metastatic malignant melanoma were transplanted into immunocompromised mice to generate PDXes. 23/26 melanoma biopsies generated serially transplantable PDX models, and their histology, mutation status and expression profile resembled their corresponding patient biopsy. The potential treatment for one patient was revealed by an in vitro drug screen and treating PDXes with the MEK inhibitor trametinib. In another patient, the BRAF mutation predicted the response of both the patient and its corresponding PDXes to MAPK-targeted therapy. Importantly, in this unselected group of patients, the time from biopsy for generation of PDXes until death was significantly longer than the time required to reach the treatment phase of the PDXes. Thus, it could be clinically meaningful to use this type of platform for melanoma patients as a pre-selection tool in clinical trials.

    View details for Web of Science ID 000348036500007

    View details for PubMedID 25228592

    View details for PubMedCentralID PMC4259423

  • Identification of tumorigenic and therapeutically actionable mutations in transplantable mouse tumor cells by exome sequencing. Oncogenesis Bhadury, J., López, M. D., Muralidharan, S. V., Nilsson, L. M., Nilsson, J. A. 2013; 2

    Abstract

    Cancer development occurs in response to the successive accumulation of mutations that eventually targets key regulators of cell proliferation. As most mutations likely occur randomly, cancer driver mutations can only be found if they are recurrent. Here we use exome sequencing of the mouse cell lines Panc02, L1210 and Colon 26 to identify genetic alterations (single-nucleotide polymorphisms and small insertion and deletions) that occurred in three different strains of mice and that resulted in tumorigenesis. We identify known mutations in genes like Kras, Cdkn2a/b, Smad4 and Trp53 and a large list of genes whose causal link to cancer is unknown. Interestingly, by screening a compound library we find that the identified oncogenic Kras mutation in Colon 26 cells correlates with its sensitivity to MEK inhibitors in vitro and in vivo. Our analysis of these mouse tumor exomes show that their manageable number of mutations could facilitate the identification of novel mutations or pathways driving tumor development. Furthermore, their use as tools is now enhanced as they can be used to create syngenic transplant models for utilization in drug discovery and validation. Finally, by showing that Kras mutant Colon 26 cells are sensitive to MEK inhibitors, we provide one proof-of-principle experiment that a platform containing targeted resequencing and drug screens could be a valuable addition in the clinic to devise anti-cancer drug schemes.

    View details for DOI 10.1038/oncsis.2013.8

    View details for PubMedID 23588493

    View details for PubMedCentralID PMC3641362