BMB Reports 2023; 56(1): 43-48  https://doi.org/10.5483/BMBRep.2022-0155
Development of bioinformatics and multi-omics analyses in organoids
Doyeon Ha1 , JungHo Kong1 , Donghyo Kim1 , Kwanghwan Lee1 , Juhun Lee1 , Minhyuk Park1 , Hyunsoo Ahn2 , Youngchul Oh & Sanguk Kim1,2,*
1Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, 2Graduate School of Artificial Intelligence, Pohang University of Science and Technology, Pohang 37673, Korea
Correspondence to: Tel: +82-54-279-2348; Fax: +82-54-279-2199; E-mail: sukim@postech.ac.kr
Received: May 16, 2022; Revised: October 25, 2022; Accepted: October 25, 2022; Published online: October 25, 2022.
© Korean Society for Biochemistry and Molecular Biology. All rights reserved.

cc This is an open-access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
ABSTRACT
Pre-clinical models are critical in gaining mechanistic and biological insights into disease progression. Recently, patient-derived organoid models have been developed to facilitate our understanding of disease development and to improve the discovery of therapeutic options by faithfully recapitulating in vivo tissues or organs. As technological developments of organoid models are rapidly growing, computational methods are gaining attention in organoid researchers to improve the ability to systematically analyze experimental results. In this review, we summarize the recent advances in organoid models to recapitulate human diseases and computational advancements to analyze experimental results from organoids.
Keywords: Computational biology, Multi-omics analysis, Organoids, Systems biology
INTRODUCTION

The use of cell lines and animal models has greatly improved our mechanistic understanding of human developmental processes and disease initiation. Identifying disease mechanisms using cell lines and animal models has guided the development of potential drugs, however, there exist inherent differences between classical pre-clinical models and humans, which limits the clinical translation of research findings. The development of 3D culture organoid models, which are self-organizing cells that mimic in vivo human tissue, can help reduce these limitations.

In this review, we describe examples of organoid models and computational methods and their application in detecting human disease signatures using genetic mutations, expression patterns, multi-omics analyses, and image data. It includes examples of sequencing techniques used in organoid research, as well as computational methods developed to improve the detection of true signals from the sequencing results.

GENOMIC ANALYSES OF ORGANOIDS

Organoids are powerful tools for identifying and validating disease mechanisms since (i) they provide an accurate representation of human tissue and (ii) they can be genetically modified. For instance, Li and colleagues observed that genetic modification of tumor suppressors or oncogenes can recapitulate colon tumorigenesis in organoid models (1). In another example, Artegiani and colleagues discovered that loss of BRCA 1 Associated Protein 1 (BAP1), a deubiquitinating enzyme, in liver cancer organoid models resulted in a loss of multiple epithelial characteristics and acquisition of tumorigenic features, supporting that BAP1 can act as a tumor suppressor in the liver (2).

Genetic manipulation in organoid models revealed key oncogenic variants in cancer. Specifically, studies have found context-specific oncogenic signaling pathways and tumor suppressor genes (TSGs) through genome-wide Clustered Regularly Interspaced Short Palindromic Repeats-Cas9 (CRISPR-Cas9) screening of organoids. For example, several TSGs were identified as negative regulators of the Transforming Growth Factor (TGF)-beta pathway in the context of mutant Adenomatous Polyposis Coli (APC), including subunits of the Switch/Sucrose Non-Fermentable (SWI/SNF) chromatin remodeling complex (3). Transforming Growth Factor Beta Receptor 2 (TGFBR2) was known to be the most prevalent TSG in the context of mutant APC and Kirsten rat sarcoma virus (KRAS) for colorectal cancer (4). For genetically defined benign tumor-derived organoids carrying APC and KRAS mutations, tumorigenic ability of the mutation in Activin A Receptor Type 1B (ACVR1B), Activin A Receptor Type 2A (TACVR2A), and AT-Rich Interaction Domain 2 (ARID2) was evaluated by disrupting TSG function and the role of Transformation-related Protein 53 (TRP53) was uncovered in tumor metastasis (5).

Genetic manipulation in organoid models has also been conducted to observe developmental processes or to identify various human disease variants. Ungricht and colleagues found that the combination of inducible genome-editing and longitudinal sampling of human kidney organoids not only provided the knowledge of early differentiation into the kidney lineage, but also helped to identify childhood/chronic kidney disease-related genes, such as ciliopathies (6). Beumer and colleagues discovered that CRISPR-Cas9 engineered organoid biobank can be used to identify essential host factors for coronaviruses (7). Furthermore, CRISPR-Cas9 gene editing can be applied to human intestinal organoids to correct disease-causing mutations. Correction of Cystic Fibrosis Transmembrane Conductance Regulator (CFTF) locus by homologous recombination using CRISPR-Cas9 gene editing system in cultured intestinal stem cells of cystic fibrosis patients restored the level of swelling comparable to that of wild-type organoids (8).

Testing all genetic variants for their functional role will require a vast amount of resources. Therefore, evaluating the impact of the genetic variants prior to functional validation in organoid models can effectively reduce experimental time and cost. Many computational methods have been developed to predict the impact of genetic variants using evolutionary selections on noncoding regions (9) or coding regions (10, 11), protein 3D structures, (12) and splicing patterns (13). These computational methods first identify disease variants, which can then be validated in organoids. For instance, to identify and validate cancer driver genes, Arnold and colleagues used a computational tool, PolyPhen-2, to identify potential pathogenic mutations in pancreatic ductal adenocarcinoma patients (14). They identified D48Y and R609H missense mutations in RAD50 Interactor 1 (RINT1), where the variants were suggested to hamper RINT1 function by impairing the interaction with zw10 kinetochore protein (ZW10) and RAD50 Double Strand Break Repair Protein (RAD50). They proved the disease-association of the mutations based on the genetic alteration of RINT1 on patient-derived organoids. These results suggest that computational methods may facilitate the discovery of novel disease variants when combined with functional validation in organoid models.

GENE EXPRESSION PROFILING OF ORGANOIDS

A cell’s functional role is determined by the expressed genes and therefore, gene expression is considered as an intermediate phenotype (15, 16) in understanding various human diseases. Accordingly, gene expression profiling has been conducted on organoid models to measure molecular similarity between organoids and human, especially by comparing the similarities in developmental trajectories or cellular compositions. For example, Cowan and colleagues developed human retinal organoids with functional synapses, and sequenced the organoids at seven developmental time points that span 46 weeks (17). The results showed that the organoids developed into matured state at 30 to 38 weeks, with transcriptomes matching that of a developing human retina in vivo.

In addition to retinal organoids, human cerebral organoids have been developed to understand the developmental processes or disease-related signatures of the human brain. For instance, Kanton and colleagues developed human, chimpanzee, and macaque cerebral organoids, and conducted single-cell RNA sequencing (scRNA-seq) to detect human-specific expression patterns, which revealed that human neuronal development was much slower than two other primates (18). Another group analyzed cerebral organoids generated from eight bipolar disorder patients and eight healthy control individuals using a reprogramming human induced pluripotent stem cells, and found downregulation of gene expression in the biological processes including cell adhesion and neurodevelopment (19).

Gene expression profiling also revealed key similarities between cancer organoids and human primary tumors. Liu and colleagues evaluated transcriptomic similarity of organoids and cancer cell lines and showed that organoids more closely resembled metastatic breast cancer samples compared to cancer cell lines (20). Another recent study found that a pancreatic ductal adenocarcinoma cancer (PDAC) organoid has multiple subtypes, suggesting that organoid models are useful in recapitulating intra-tumor heterogeneity (21). In detail, scRNA-seq revealed that the two molecular subtypes (classical and basal) of PDAC coexist in the same organoid sample. Given the transcriptome similarity between organoids and in vivo tissue samples, gene expression profiling in organoid models are useful in identifying mechanisms of action of therapeutic drugs and drug biomarkers. For instance, Norkin and colleagues developed a method for systematically conducting drug screening and gene expression profiling in colorectal cancer organoids (22). They applied the method in wild-type and cancer organoids to screen cell differentiation inducing anti-cancer drugs.

Despite the usefulness of gene expression analysis, results derived from expression profiles often do not generate reproducible results (23). One of the approaches that can improve the reproducibility of gene expression analyses is the use of a protein-protein interaction network. Recent studies suggest that genes associated with diseases would likely form clusters in protein interactome (24, 25). Based on these observations, a method has been developed to identify anti-cancer drug biomarkers from drug screening data in organoids (26). Kong and colleagues showed that drug response biomarkers can be derived using a protein-protein interaction network with drug screening and transcriptomics data from patient-derived organoid models, which are predictive of drug response in 5-Fluorouracil-treated colorectal cancer and cisplatin-treated bladder cancer patients.

MULTI-OMICS ANALYSES OF ORGANOIDS

Multi-omics analysis has played an essential role in identifying physiological similarities between in vitro organoids and in vivo organs. For the brain, Amiri and colleagues provided evidence that human-induced, pluripotent stem cell (hiPSC)-derived organoids recapitulate human cortical development from the embryonic to the prenatal stage (27). They compared chromatin immunoprecipitation sequencing (ChIP-seq) data and RNA-seq data obtained from hiPSC-derived organoids with those from isogenic human postmortem fetal samples. They identified enhancer-gene links that are closely related to early cortical development. In addition, they showed that genes related to cortical development during early fetal stages are also associated with neurodevelopmental disorders such as autism spectrum disorder (ASD) (27). Additionally, brain organoids have been developed to model the postnatal stage of brain development. Gordon and colleagues demonstrated that the developmental process observed in 3D organoids derived from human stem cells displayed high transcriptomic and epigenetic similarities with neurodevelopmental processes observed in-vivo (28). These findings were further confirmed by an observation that organoids underwent chromatin remodeling, which resembled developmental processes observed in the in-vivo human forebrain (29).

In addition to brain organoids, multi-omics analyses demonstrated that 3D organoids derived from primary tumors recapitulated the features of in-vivo human tumors. For example, Broutier and colleagues showed that organoid cultures from eight liver cancer patients successfully recapitulated genomic, transcriptomic, and histological features of the primary tumor even after the organoids had been cultured for long-term periods (30). Gao and colleagues leveraged genomic and transcriptomic data to confirm the resemblance of organoids derived from prostate cancer patients (31), and Boj and colleagues utilized transcriptomic and proteomic analyses to comprehensively understand pancreatic organoids (32). Integrated analyses of genomic, transcriptomic, and phosphor-proteomic data derived from colorectal cancer patient-derived organoids also showed that patient-derived organoids could recapitulate patients’ tumors at the molecular level (33).

Since various omics data showed that patients-derived organoid models accurately mimicked original tissue development and disease pathology, many precision medicine methods were developed through drug screening in organoid models. Specifically, cancer organoid models have been developed and applied to test the efficacy of various anticancer agents. For example, using a high-throughput drug screening platform for patient-derived colorectal organoids, Toshimitsu and colleagues discovered that bromodomain and extra-terminal (BET) bromodomain protein inhibitor as a cancer-selective growth suppressor that targets genes were aberrantly activated in colorectal cancer (34). Using genome and transcriptome data together, they discovered an association between checkpoint with fork-head and ring finger domains (CHFR) silencing and paclitaxel sensitivity. Similarly, Broutier and colleagues identified novel drugs for liver cancer treatment (30). They developed an organoid model for the three most common primary liver cancer (PLC) subtypes: hepatocellular carcinoma (HCC), cholangiocarcinoma (CC), and combined HCC/CC (CHC) tumors. Multi-omics analysis using genomics and transcriptomics data of PLC-derived organoids discovered the ERK inhibitor SCH772984 as a potential therapeutic agent for primary liver cancer. Using tumor organoids from mouse endometrial cancer, Chen and colleagues performed drug screening with a library of small molecules that targets epigenetic factors. Their RNA-seq and CHIP-seq results showed that the hypoxia-inducible factor (HIF) pathway was the most significantly downregulated pathway by MI-136 treatment (35).

Moreover, the accumulation of organoid models has led to the establishment of organoid biobanks and enabled a large-scale drug screening which successfully revealed several unexpected drugs to be tested and approved in clinical trials. Yan et al. established a primary gastric cancer organoid (GCO) biobank that comprised normal, dysplastic, cancer, and lymph node metastases (n = 63) from 34 patients with detailed whole-exome and transcriptome analysis (36). They identified gastric cancer therapeutic agents in the clinical phase of development, including Napabucasin, Abemaciclib, and the ATR inhibitor VE-822. The OncoTrack consortium built a large biobank of 106 colorectal cancer patients (stages I-IV) and developed a pre-clinical platform composed of 35 organoids and 59 xenografts, with extensive omics data comprised of a compendium of drug sensitivity data totaling >4,000 assays testing 16 clinical drugs (37). Linking molecular profiles with drug sensitivity patterns identifies novel biomarkers, including a signature that outperformed RAS/RAF mutations in predicting sensitivity to the EGFR inhibitor cetuximab.

The proteomics data from organoids is also applied in drug screening. Schumacher and colleagues analyzed a cohort of well-characterized colorectal cancer organoids and found heterogeneous activation of mitogen-activated protein kinase (MAPK) signaling pathway in the targeted proteomics data. They showed that the heterogeneity resulted in variable responses to epidermal growth factor receptor (EGFR) inhibition (38). Drug screening was also conducted in diverse disease models including cancer models. Mills and colleagues performed functional screening of 105 small molecules with pro-regenerative potential in cardiac organoids (39).

The key objective in the multi-omics analysis is to capture a robust biological signature. Common approaches for identifying robust signatures include (i) leveraging regulatory or protein-protein interaction networks (40, 41) and (ii) using machine-learning (ML) techniques, such as deep neural networks (42). As important signatures may be undiscovered when using single-omics data, the application of multi-omics methods in organoid models will become increasingly important as more sequencing data from organoid models become available.

IMAGE-BASED ANALYSES OF ORGANOIDS

Unlike omics-based sequencing methods, image-based data provide information on cell composition, morphology, and their positional location during cell development, which are closely related to cellular function and phenotypic outcome of an organism (36). Conventional image-based data includes the use of brightfield microscopy, H&E imaging, and immunostaining techniques (36). Interestingly, recent advances in high-resolution microscopy and 3D confocal staining have made it possible to generate 3D organoid images, which in turn have led to the development of high-content screening (HCS) (43, 44). Lukonin and colleagues used HCS to characterize intestinal organoids and to accurately identify their developmental trajectory (45). In brain research, where research using organoids are currently being actively conducted, high-throughput screening results were verified by checking the shape, size, total gene expression, cell composition, and structure through image data (46). Recently, there have been efforts to develop a drug screening platform based on high-resolution 3D imaging technology (34). All of these efforts resulted in an increased number of image data for various organoids.

As high-throughput imaging techniques become more available, it is likely that ML techniques will be actively applied to help automate analyses. Specifically, organoid images form optically opaque aggregates due to dense, diverse, and tightly interacting cells, posing difficulties for human experts to conduct cell segmentation and phenotypic annotation of organoid image data (47). To solve such limitations, recent studies have focused on applying ML techniques to accurately interpret image data from organoids. For instance, Gritti and colleagues utilized a relatively simple ML model (i.e. Logistic Regression and Multi-layer perceptron) for efficient segmentation, quantification, and visualization of large-scale image data of organoids (48). Also, there were efforts to handle organoid images with more sophisticated ML techniques including convolutional neural networks (CNNs). Kassis and colleagues utilized a CNN for automated recognition and quantification of human intestinal organoids with brightfield images (49). Kok and colleagues tracked the development of organoids by recognizing histone H2B-mCherry stained organoid images in single-cell resolution relying on a CNN (50). An interesting research opportunity in organoid image-based tasks would be to apply high-performance image recognition ML models that are being developed in the field of computer vision.

CONCLUSION

Organoids show close similarities in molecular profiles, morphological characteristics, and drug responses with human tissues with respect to it. However, because organoid technology is still in its infancy, the ability to accurately mimic mature organs observed in human requires further investigation. One of the approaches for improving organoid models is to incorporate a tissue microenvironment. Kim and colleagues created a bladder organoid that includes tissue stem cells with stromal components termed assembloids (51). It was able to recapitulate regenerative responses to bladder injury that matches in vivo regeneration patterns. Moreover, the bladder assembloid revealed a master pioneer factor, Forkhead Box A1 (FOXA1), that determines tumor phenotype in bladder cancer. Neal and colleagues used an air-liquid interface (ALI) to incorporate tumor organoids with immune cells (52). The organoid model using ALI method was treated with immunotherapies (anti-programmed cell death protein 1 or anti-programmed cell death ligand 1) and successfully observed tumor cell death. Altogether, further investigations to improve organoid models will be the key to the development of experimental techniques that will adequately incorporate stromal and immune cell components that match in vivo tissue environments.

A better understanding of the similarities between human and organoid models for improved organoid technology is likely to be an active research subject. In this respect, computational methods developed to quantify similarities between pre-clinical models and humans can be applied in organoid research. Han and colleagues have previously developed computational methods to understand similarities between human and mouse models (53, 54), which can be applied in organoid research as the number of sequencing data and phenotypic profiling in organoid models increases.

CONFLICTS OF INTEREST

The authors have no conflicting interests.

REFERENCES
  1. Li X, Nadauld L, Ootani A et al (2014) Oncogenic transformation of diverse gastrointestinal tissues in primary organoid culture. Nat Med 20, 769-777
    Pubmed KoreaMed CrossRef
  2. Artegiani B, van Voorthuijsen L, Lindeboom RGH et al (2019) Probing the tumor suppressor function of BAP1 in CRISPR-engineered human liver organoids. Cell Stem Cell 24, 927-943
    Pubmed CrossRef
  3. Ringel T, Frey N, Ringnalda F et al (2020) Genome-scale CRISPR screening in human intestinal organoids identifies drivers of TGF-β resistance. Cell Stem Cell 26, 431-440
    Pubmed CrossRef
  4. Michels BE, Mosa MH, Streibl BI et al (2020) Pooled in vitro and in vivo CRISPR-Cas9 screening identifies tumor suppressors in human colon organoids. Cell Stem Cell 26, 782-792
    Pubmed CrossRef
  5. Takeda H, Kataoka S, Nakayama M et al (2019) CRISPR-Cas9-mediated gene knockout in intestinal tumor organoids provides functional validation for colorectal cancer driver genes. Proc Natl Acad Sci U S A 116, 15635-15644
    Pubmed KoreaMed CrossRef
  6. Ungricht R, Guibbal L, Lasbennes MC et al (2022) Genome-wide screening in human kidney organoids identifies developmental and disease-related aspects of nephrogenesis. Cell Stem Cell 29, 160-175
    Pubmed CrossRef
  7. Beumer J, Geurts MH, Lamers MM et al (2021) A CRISPR/Cas9 genetically engineered organoid biobank reveals essential host factors for coronaviruses. Nat Commun 12, 5498
    Pubmed KoreaMed CrossRef
  8. Schwank G, Koo BK, Sasselli V et al (2013) Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients. Cell Stem Cell 13, 653-658
    Pubmed CrossRef
  9. Khurana E, Fu Y, Colonna V et al (2013) Integrative annotation of variants from 1092 humans: application to cancer genomics. Science 342, 1235587
    Pubmed KoreaMed CrossRef
  10. Ng PC and Henikoff S (2001) Predicting deleterious amino acid substitutions. Genome Res 11, 863-874
    Pubmed KoreaMed CrossRef
  11. Kim D, Han SK, Lee K, Kim I, Kong JH and Kim S (2019) Evolutionary coupling analysis identifies the impact of disease-associated variants at less-conserved sites. Nucleic Acids Res 47, e94
    Pubmed KoreaMed CrossRef
  12. Adzhubei IA, Schmidt S, Peshkin L et al (2010) A method and server for predicting damaging missense mutations. Nat Methods 7, 248-249
    Pubmed KoreaMed CrossRef
  13. Desmet FO, Hamroun D, Lalande M, Collod-Bëroud G, Claustres M and Béroud C (2009) Human splicing finder: an online bioinformatics tool to predict splicing signals. Nucleic Acids Res 37, e67
    Pubmed KoreaMed CrossRef
  14. Arnold F, Gout J, Wiese H et al (2021) RINT1 regulates SUMOylation and the DNA damage response to preserve cellular homeostasis in pancreatic cancer. Cancer Res 81, 1758-1774
    Pubmed CrossRef
  15. Schadt EE, Lamb J, Yang X et al (2005) An integrative genomics approach to infer causal associations between gene expression and disease. Nat Genet 37, 710-717
    Pubmed KoreaMed CrossRef
  16. Chen Y, Zhu J, Lum PY et al (2008) Variations in DNA elucidate molecular networks that cause disease. Nature 452, 429-435
    Pubmed KoreaMed CrossRef
  17. Cowan CS, Renner M, de Gennaro M et al (2020) Cell types of the human retina and its organoids at single-cell resolution. Cell 182, 1623-1640
    Pubmed KoreaMed CrossRef
  18. Kanton S, Boyle MJ, He Z et al (2019) Organoid single-cell genomic atlas uncovers human-specific features of brain development. Nature 574, 418-422
    Pubmed CrossRef
  19. Kathuria A, Lopez-Lengowski K, Vater M, McPhie D, Cohen BM and Karmacharya R (2020) Transcriptome analysis and functional characterization of cerebral organoids in bipolar disorder. Genome Med 12, 34
    Pubmed KoreaMed CrossRef
  20. Liu K, Newbury PA, Glicksberg BS et al (2019) Evaluating cell lines as models for metastatic breast cancer through integrative analysis of genomic data. Nat Commun 10, 2138
    Pubmed KoreaMed CrossRef
  21. Krieger TG, le Blanc S, Jabs J et al (2021) Single-cell analysis of patient-derived PDAC organoids reveals cell state heterogeneity and a conserved developmental hierarchy. Nat Commun 12, 5826
    Pubmed KoreaMed CrossRef
  22. Norkin M, Ordóñez-Morán P and Huelsken J (2021) High-content, targeted RNA-seq screening in organoids for drug discovery in colorectal cancer. Cell Rep 35, 109026
    Pubmed CrossRef
  23. Raspe E, Decraene C and Berx G (2012) Gene expression profiling to dissect the complexity of cancer biology: Pitfalls and promise. Semin Cancer Biol 22, 250-260
    Pubmed CrossRef
  24. Menche J, Sharma A, Kitsak M et al (2015) Uncovering disease-disease relationships through the incomplete interactome. Science 347, 1257601
    Pubmed KoreaMed CrossRef
  25. Hofree M, Shen JP, Carter H, Gross A and Ideker T (2013) Network-based stratification of tumor mutations. Nat Methods 10, 1108-1115
    Pubmed KoreaMed CrossRef
  26. Kong JH, Lee H, Kim D et al (2020) Network-based machine learning in colorectal and bladder organoid models predicts anti-cancer drug efficacy in patients. Nat Commun 11, 5485
    Pubmed KoreaMed CrossRef
  27. Amiri A, Coppola G, Scuderi S et al (2018) Transcriptome and epigenome landscape of human cortical development modeled in organoids. Science 362, eaat6720
    Pubmed KoreaMed CrossRef
  28. Gordon A, Yoon SJ, Tran SS et al (2021) Long-term maturation of human cortical organoids matches key early postnatal transitions. Nat Neurosci 24, 331-342
    Pubmed KoreaMed CrossRef
  29. Trevino AE, Sinnott-Armstrong N, Andersen J et al (2020) Chromatin accessibility dynamics in a model of human forebrain development. Science 367, eaay1645
    Pubmed KoreaMed CrossRef
  30. Broutier L, Mastrogiovanni G, Verstegen MMA et al (2017) Human primary liver cancer-derived organoid cultures for disease modeling and drug screening. Nat Med 23, 1424-1435
    Pubmed KoreaMed CrossRef
  31. Gao D, Vela I, Sboner A et al (2014) Organoid cultures derived from patients with advanced prostate cancer. Cell 159, 176-187
    Pubmed KoreaMed CrossRef
  32. Boj SF, Hwang CI, Baker LA et al (2015) Organoid models of human and mouse ductal pancreatic cancer. Cell 160, 324-338
    Pubmed KoreaMed CrossRef
  33. Codrich M, Dalla E, Mio C et al (2021) Integrated multi-omics analyses on patient-derived CRC organoids highlight altered molecular pathways in colorectal cancer progression involving PTEN. J Exp Clin Cancer Res 40, 198
    Pubmed KoreaMed CrossRef
  34. Toshimitsu K, Takano A, Fujii M et al (2022) Organoid screening reveals epigenetic vulnerabilities in human colorectal cancer. Nat Chem Biol 18, 605-614
    Pubmed CrossRef
  35. Chen J, Zhao L, Peng H et al (2021) An organoid-based drug screening identified a menin-MLL inhibitor for endometrial cancer through regulating the HIF pathway. Cancer Gene Ther 28, 112-125
    Pubmed CrossRef
  36. Yan HHN, Siu HC, Law S et al (2018) A comprehensive human gastric cancer organoid biobank captures tumor subtype heterogeneity and enables therapeutic screening. Cell Stem Cell 23, 882-897
    Pubmed CrossRef
  37. Schütte M, Risch T, Abdavi-Azar N et al (2017) Molecular dissection of colorectal cancer in pre-clinical models identifies biomarkers predicting sensitivity to EGFR inhibitors. Nat Commun 8, 14262
    Pubmed KoreaMed CrossRef
  38. Schumacher D, Andrieux G, Boehnke K et al (2019) Heterogeneous pathway activation and drug response modelled in colorectal-tumor-derived 3D cultures. PLoS Genet 15, e1008076
    Pubmed KoreaMed CrossRef
  39. Mills RJ, Parker BL, Quaife-Ryan GA et al (2019) Drug screening in human PSC-cardiac organoids identifies pro-proliferative compounds acting via the mevalonate pathway. Cell Stem Cell 24, 895-907
    Pubmed CrossRef
  40. Webber JT, Kaushik S and Bandyopadhyay S (2018) Integration of tumor genomic data with cell lines using multi-dimensional network modules improves cancer pharmacogenomics. Cell Syst 7, 526-536
    Pubmed KoreaMed CrossRef
  41. Han SK, Kong J, Kim S, Lee JH and Han DH (2019) Exomic and transcriptomic alterations of hereditary gingival fibromatosis. Oral Dis 25, 1374-1383
    Pubmed CrossRef
  42. Sharifi-Noghabi H, Zolotareva O, Collins CC and Ester M (2019) MOLI: multi-omics late integration with deep neural networks for drug response prediction. Bioinformatics 35, i501-i509
    Pubmed KoreaMed CrossRef
  43. Boutros M, Heigwer F and Laufer C (2015) Microscopy-based high-content screening. Cell 163, 1314-1325
    Pubmed KoreaMed CrossRef
  44. Dekkers JF, Alieva M, Wellens LM et al (2019) High-resolution 3D imaging of fixed and cleared organoids. Nat Protoc 14, 1756-1771
    Pubmed CrossRef
  45. Lukonin I, Serra D, Challet Meylan L et al (2020) Phenotypic landscape of intestinal organoid regeneration. Nature 586, 275-280
    Pubmed KoreaMed CrossRef
  46. Renner H, Grabos M, Becker KJ et al (2020) A fully automated high-throughput workflow for 3d-based chemical screening in human midbrain organoids. Elife 9, e52904
    Pubmed KoreaMed CrossRef
  47. Renner H, Schöler HR and Bruder JM (2021) Combining automated organoid workflows with artificial intelligence-based analyses: opportunities to build a new generation of interdisciplinary high-throughput screens for Parkinson's disease and beyond. Mov Disord 36, 2745-2762
    Pubmed CrossRef
  48. Gritti N, le Lim J, Anlaş K et al (2021) Morgana: accessible quantitative analysis of organoids with machine learning. Development 148, dev199611
    Pubmed KoreaMed CrossRef
  49. Kassis T, Hernandez-Gordillo V, Langer R and Griffith LG (2019) OrgaQuant: human intestinal organoid localization and quantification using deep convolutional neural networks. Sci Rep 9, 12479
    Pubmed KoreaMed CrossRef
  50. Kok RNU, Hebert L, Huelsz-Prince G et al (2020) OrganoidTracker: efficient cell tracking using machine learning and manual error correction. PLoS One 15, e0240802
    Pubmed KoreaMed CrossRef
  51. Kim E, Choi S, Kang B et al (2020) Creation of bladder assembloids mimicking tissue regeneration and cancer. Nature 588, 664-669
    Pubmed CrossRef
  52. Neal JT, Li X, Zhu J et al (2018) Organoid modeling of the tumor immune microenvironment. Cell 175, 1972-1988
    Pubmed KoreaMed CrossRef
  53. Han SK, Kim D, Lee H, Kim I and Kim S (2018) Divergence of noncoding regulatory elements explains gene-phenotype differences between human and mouse orthologous genes. Mol Biol Evol 35, 1653-1667
    Pubmed CrossRef
  54. Ha D, Kim D, Kim I et al (2022) Evolutionary rewiring of regulatory networks contributes to phenotypic differences between human and mouse orthologous genes. Nucleic Acids Res 50, 1849-1863
    Pubmed KoreaMed CrossRef


This Article

e-submission

Archives