Abstract
As genome editors move into clinical trials, there is a need to establish ex vivo multicellular systems to rapidly assess and predict toxic effects of genome editors in physiologically relevant human models. Advancements in organoid and organs-on-chip technologies offer the possibility to create multicellular systems that replicate the cellular composition and metabolic function of native tissues. Some multicellular systems have been validated in multiple applications for drug discovery and could be easily adapted to test genome editors; other models, especially those of the adaptive immune system, will require validation before being used as benchmarks for testing genome editors. Likewise, protocols to assess immunogenicity, to detect off-target effects, and to predict ex vivo to in vivo translation will need to be established and validated. This review will discuss key aspects to consider when designing, building, and/or adopting in vitro human multicellular systems for testing genome editors.
Keywords: CRISPR-Cas, Off-target effects, Organoids, Organ-on-chips, Adaptive immunity
Graphical Abstract

1. Introduction
The Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR) system has revolutionized genome editing due to ease of engineering and programmability [1]. For safe human translation, CRISPR-based gene editing must have no off-target effects in the genome [2]. CRISPR itself or its delivery vehicle may have unintended adverse effects within cells or systemically, which warrants detailed elucidation of the toxic effects in appropriate model systems before human administration [3,4]. Significant progress has been made to modify CRISPR-Associated (Cas) nucleases or the guide RNAs (gRNAs) to tackle specificity [5,6]. Likewise, novel viral and non-viral strategies with reduced immunogenicity and increased delivery efficacy for genome editors are underway [7] (Table 1). Precise genome-wide techniques for detection of off-target insertions/deletions (indels) have also been created [8,9]. In contrast, little progress has been made to examine the gene editors in platforms that can yield human relevant readouts to enable us to rapidly predict and assess toxic effects of CRISPR. Most studies have used human cell lines or animal models, which fall short in accurately predicting the response of primary human cells to genome editors. Through integration of stem cells or multiple cell types, physical and biochemical cues in three dimensions (3D), organs-on-chip and organoid technologies seek to establish ex vivo human models that can capture the complexity of their in vivo tissue counterparts [10–12]. These multicellular systems provide a platform to fill the gap between animal studies and human trials, enabling prolonged maintenance and manipulation of primary human cells ex vivo. Therefore, human-based multicellular platforms can facilitate studies of the efficacy, toxicity, and immunogenicity of CRISPR and its delivery vehicles.
Table 1:
Challenges of CRISPR-based genome editors for human translation
| Challenge | Possible solutions |
|---|---|
| Genome Editor Efficacy | Development of Cas proteins with improved activity. Identification of other Cas proteins (e.g, Cas13) or smaller Cas protein for easy virus packaging. Directed evolution of Cas orthologs. |
| Genome Editor Efficiency | Improved delivery system, lentivirus, adeno-associated virus (AAV), nanoparticles with cell-specific receptors for target cell delivery. |
| Genome Editor Immunogenic Activity | Improved delivery system with reduced immunogenicity (e.gs., Cas9 epitope modification, humanized Cas9 protein orthologous to build all-human Cas enzyme), immune orthogonal Cas orthologs |
| Genome Editor in vitro to in vivo Translation | Development of near-physiological multicellular tissue constructs with controllable flow, oxygen, and long-term culture. Possibility of connecting multiple tissue constructs |
2. Multicellular Systems for Modeling Genome Editing in Humans
2.1. Organoids
Organoids are self-organizing 3D cellular structures that develop from adult stem cells or from induced pluripotent stem cells (iPSCs) that resemble organ functionality and tissue patterning similar to in vivo tissues [13,14]. The self-organizing capacity of stem cells enables initiation and execution of intricate cellular interactions and developmental processes that eventually result in functional tissue structures that enable decoding design principles of tissue morphogenesis, physiology, and disease states [15,16].
A pioneering study by Schwank, et al., in 2013 combined organoid and CRISPR-Cas9 technologies for the successful therapeutic repair of the cystic fibrosis transmembrane conductor receptor (CFTR) gene in intestinal organoids derived from cystic fibrosis patients [17]. This approach has been recently extended to repair the CFTR gene in patient-derived upper-airway organoids using CRISPR-Cas9 [18] and to repair a CFTR point mutation in multiple patient-derived intestinal organoid lines using CRISPR-based adenine base editors [19]. Further, tissue-derived or iPSC-derived organoids can be used to create in vitro models of genetic mutations found in cancer patients using CRISPR-based genome editing [20,21].
Organoid biobanks can provide an easy to use, medium-throughput platform to capture disease heterogeneity, sex-specific disease differences, donor-to-donor (epi)genetic variability, and patient-to-patient variable responses to genome editors [19,22]. As most gRNAs are designed based on the human reference genome, these gRNAs run the risk of having off-target effects due to population genome variability [23]. Therefore, it might be possible to use patient-derived organoid biobanks and in silico screening, to validate the best gRNA for individual patients. Organoids contain a proliferative stem cell population that can be propagated continuously, thus, they might be useful models to follow changes in the genome (on- and off-target) over multiple generations after genome editing. One challenge is that some organoid models undergo necrosis due to the accumulation of apoptotic cells and lack of oxygenation and need to be passaged every 7-10 days; therefore, long-term effects of genome editors are limited without disrupting the original organoid population [12].
While organoids provide an emerging platform for assessment of genome editors using human stem cell populations, the lack of immune cells in some organoids [24] can reduce their utility for investigating immune response that could be harmful in vivo [25]. Additionally, in the context of iPSC-derived organoids, limited maturity can impede their capacity to capture adultlike human phenotypes. However, recent advances in iPSC-derived organoids have improved organoid maturation, vascularization, and tissue function which collectively provide a promising outlook for using organoids to test genome editors [11,26–31].
2.2. Organs-on-chips
Unlike organoids in which the 3D architecture spontaneously develops, organs-on-chips offer the possibility of designing topographical features of the native tissue. Organs-on-chips are complex ex vivo models that are built using microprinting, soft lithography, and polymer molds [32,33]. The integration of cell culture flow for perfusion, mechanical stimulation, and oxygen gradient regulation in organs-on-chips mimic features of native tissues in ways unachievable with organoids [10,12,34]. Various organs-on-chip models have been created for brain, lung, liver, gut, heart, bone marrow, thymus, and kidney tissues [33].
An attractive feature of organs-on-chips is engineering physiochemical properties of tissue such as the flow of culture media to allow controlled nutrient replenishment and continuous measurement of secreted cytokines or metabolites that might be affected by genome editing, which combined with multi-omics (e.gs., proteomics, metabolomics) and deep sequencing (e.g., RNAseq), offers a comprehensive analysis of the effects of genome editors in multicellular tissue constructs [34]. However, organs-on-chips models have been heavily developed using cell lines (e.g., cancer cells) that are easy to manipulate and more resistant to stress [10]. Other organs-on-chip models combine cell types from different sources (e.gs., immortalized cell lines, iPSCs, human and mouse primary cells) which results in tissue-models of mixed cell phenotype population, or tissue-models that represent early states in human development, especially when using iPSC-derived cells that usually do not mature beyond a fetal state [33,35]. The organs-on-chips field has started to use primary cells isolated from patient tissues to faithfully mimic physiological processes [32,33,35]. While the use of primary cells allows for the creation of tissue models for personalized medicine and the study of donor-to-donor variability, the use of primary cells raises additional technical challenges including: a) reproducibility in methods to promote tissue formation due to donor’s age and pathological state, b) cell loss during tissue processing and cell isolation, c) a common media formulation to maintain different cell types, d) a suitable scaffold to support multiple cell types and tissue geometries, and e) the lack of feasibility to mark and enrich primary cells that undergo a genome editing event.
New organ-on-chip applications combine patient-derived iPSCs with microfluidic flow to create organoids or iPSC-derived organs-on-chips that can also improve cell maturation and tissue organization [12,32]. Altogether, organs-on-chip models offer possibilities as platforms to test genome editors at different stages of technical development; however, it will be critical to engineer organs-on-chip models that enable the viability and assessment of multiple cell types, particularly from cells derived from the same donor. Recent ongoing efforts have also focused on studying human physiology by combining up to ten organ systems modularly [10,36–38]. These efforts together with organoids will establish a human-based multicellular system that enables assessment and improvement of genome editors in a physiologically relevant model and can fill the gap between mouse studies and human trials.
3. Delivery Options for Genome Editors in Multicellular systems
Wild-type CRISPR-associated protein 9 from Streptococcus pyogenes (WT-SpCas9) or some other CRISPR orthologs such as Staphylococcus aureus (WT-SaCas9) induces double stranded breaks (DSBs) in the genome [39–41]. DSBs are repaired either through error-prone non-homologous end joining (NHEJ) or through homology directed repair (HDR) [42]. The improper repair of on-target DSBs can lead to small indels, large deletions, or inversions [43]. Engineered Cas9 variants or other CRISPR orthologs have been introduced to increase the specificity of gene editing, single base pair replacement, targeted epigenome engineering, modification of RNA molecules, and decreased immunogenicity, to name a few [1,5,42,44–46]. CRISPR-Cas genome editors can be delivered using physical methods (e.gs. electroporation or microinjection), viral transduction (e.gs. lentivirus (LV) or adeno-associated virus (AAV)), or nanoparticles (e.g. lipid nanoparticles) [47]. Collectively, all of these methods have demonstrated utility to edit cells grown as monolayers, dispersed single cells in suspension, and in animal models [47,48].
Although the utility of organoids, organs-on-chips, and CRISPR-based genome editing is well documented, there are limited reports using organoids or organs-on-chips as models for CRISPR-based genome editing. The most extensive use of CRISPR-based genome editors with organoids has been to correct a specific mutated gene or to generate organoid cell lines with mutations found in cancer patients, using a parental organoid cell line. For most in vitro editing of tissue-derived organoids, electroporation is the preferred approach for CRISPR-Cas delivery, followed by positive antibiotic selection and expansion of transduced stem cell population [17–19]. Once the edited population is expanded, they can be enriched to detect on- and off-targets genomic effects. This approach works well for organoids that contain a stem cell population but might not be applicable for terminally differentiated cells present in multicellular ex vivo models that cannot be expanded after editing. Further, as 3D multicellular systems recapitulate some aspect of tissue architecture found in vivo, the delivery of genome editors in these ex vivo models encounters more challenges compared to dispersed single cells in suspension. For instance, cells located at the periphery of a multilayer 3D cellular system will be modified whereas cells in the center might not. Further, unless there is a fluorescence reporter or a selection marker (i.e., antibiotic resistance gene) added into the delivery vector, the identification and isolation of edited cells will be challenging, which will impact the quantitative analysis of on- and off-targets effects.
An additional challenge for efficient delivery of genome editors in multicellular systems is the selective editing of a cell population within the ex vivo model. This can be accomplished by engineering viral vectors with cellular tropism or by engineering nanoparticles with cell-specific epitopes for receptor-mediated delivery. Although both approaches work, in the case of engineered nanoparticles, there might be additional optimization for endosomal escape and effective genome editing [49]. Ultimately, the identification of an efficient delivery method of genome editors to multicellular systems will require the empirical test of multiple delivery methods in parallel, followed by quantitative analyses of delivery efficiency and off-target effects.
4. Capturing CRISPR-based DNA Cleavage and its Off-target Toxicity in Multicellular Systems
CRISPR-Cas uses a gRNA to direct the Cas nuclease to the target site via Watson-Crick base pairing (on-target) to create DSBs; however, the Cas nuclease can often create DSBs elsewhere in the genome (off-target) too [1]. The host DNA repair mechanism in response to induced DSBs can sometimes result in unintended genome alterations such as indels, chromosome rearrangements, and translocations that could affect cellular function, morphology, or the proliferative potential of the edited cells negatively (toxic effect, leading to apoptosis) or positively (turning on oncogenes, leading to over proliferation) [39,40,50].
One of the first approaches to predict and reduce off-target effects is to use in silico screening (extensively review elsewhere in ref [51]). Multiple tools (e.gs., E-CRISP, Cas-OFFinder, COSMID, Breaking-Cas, CRISTA) exist that computationally predict off-target mutations based on the gRNA and the gene editor used [52]. However, computational models often fail to predict rare off-target events and large chromosomal rearrangements and should be paired with additional in vitro, ex vivo, and in vivo testing.
Quantitative in vitro approaches for on- and off-target detection using genomic DNA isolated from edited cells include mismatch cleavage assays, such as T7E1, TIDE, and IDAA, Sanger Sequencing, targeted sequencing, and exome sequencing [50,53]. Circularization for in vitro reporting of cleavage effects by sequencing (CIRCLE-seq) has been proposed as a superior in vitro technique, compared to mismatch cleavage assays, to detect genome-wide off target mutations by CRISPR [54,55]. CIRCLE-seq has two main advantages over previous assays; it is highly sensitive despite possible cell-type-specific single-nucleotide polymorphisms (SNPs), and is able to detect off-target effects without using a reference genome [54]. Furthermore, there are methods analogous to in vitro mismatch cleavage assays, such as genome-wide unbiased identification of DSB enabled by sequencing (GUIDE-seq) and iGUIDE which employ double strand oligodeoxynucleotide tags integrated in sites of DNA breaks in the genome to detect on- and off-target cleaved sites [8,55]. Most recently, DISCOVER-Seq, a method for in situ discovery of DSBs in a single step that preserves the integrity of chromatin state and modifications, could allow better detection of translocations and large deletions [9].
Together, the techniques described above are enhancing our ability to detect CRISPR off-target events with greater precision in purified single-cell type populations, but these techniques require further refinement in sensitivity and detection capabilities to be able to detect large chromosomal rearrangements, especially when using primary human cells in which genetic variability could influence genome editing in unknown ways. There is also the need to perform a direct comparison of these methods using model cell lines to establish best practices across research groups to detect off-target effects. Adapting these techniques to detect off-target events in multicellular systems might require iteration of experimental conditions that mimic in vivo conditions. A first challenge is to pair a highly sensitive method to detect rare mutation events with the appropriate time scale for tissue collection after genome editing. Further, in multicellular systems with primary human cells, it is unlikely that edited cells will have a selection marker (i.e. antibiotic selection), thus their unsorted harvest can result in the dilution of off-target genome changes, especially for rare off-target events. Waiting too long for cell harvesting could lead to the unintended selection of cells that gained a growth advantage after gene editing, thus masking early off-target toxic events that lead to cell death. The time-dependency of testing for off-target toxicity is exemplified in an experiment that delivered Cas9/Pcsk9-gP in mice followed by on- and off-target effects detection for four days. Off-target mutations were highest after 24 hours and steadily declined for the next few days [9]. Hence, interpreting and extrapolating off-target toxic effects after genome editing using ex vivo multicellular systems will require careful experimental design. Likewise, linking a specific mutation to a cellular biological response in complex multicellular systems would require the establishment of novel experimental approaches to analyze genomic, transcriptomic, proteomic, or metabolomic implications after unintended genome editing events (Table 2).
Table 2:
Proposed metrics to evaluate the effect of genome editors on multicellular systems
| Possible readouts | Considerations | |
|---|---|---|
| Cell Metabolism | Cell Growth Cell Death | Continuous analysis, easy to set up, colorimetric, fluorometric, quantitative, non-invasive, high throughput, non-cell specific |
| Cell-secreted metabolites | Continuous analysis, easy to set up, quantitative, non-invasive, non-cell specific, high throughput (e.g., Luminex) | |
| Enzymatic Metabolic Activity | Continuous analysis, easy to set up, quantitative, non-invasive, high throughput, cell-type specific | |
| Cell Identity | Cell surface markers | End point analysis, requires optimization, quantitative, high throughput, cell-type specific, (e.gs., Flow cytometry, CyTOF) |
| Imaging analysis | Bright Field and epifluorescence microscopy | Continuous analysis, easy to set up, can be quantitative by downstream image analysis, medium throughput, non-invasive, fluorescence based on reporter (e.gs., GFP, Tdtomato, etc.). Low resolution |
| Second harmonic generation microscopy | Cumbersome to set up, can be quantitative, limited to Collagen I fibers, microtubules, and myosin. Low throughput, high resolution | |
| Confocal microscopy | Requires some optimization, can be quantitative, limited to commercially available antibodies. Low throughout, high resolution | |
| Light sheet microscopy | Requires extensive optimization, can be quantitative, limited to commercially available antibodies. Low throughout, high resolution of large tissue structures | |
| Omics | Genomics Epigenomics Transcriptomics Proteomics Glycomics Metabolomics Lipidomics | End point analysis, requires some optimization, generates terabytes of data for downstream analysis, very low throughput, quantitative. |
5. Challenges to Investigate Immunogenicity of CRISPR in Humans
While there have been significant advances in engineered genome editors for improved specificity, other areas, such as the activation of the human immunological responses to these novel genome editors, are less developed [56]. The delivery method of choice, Cas protein used, or the secondary structure of the gRNA, all independently can be recognized by the immune system and trigger a cascade of immune reactions with the release of cytokines or chemokines that activate antigen presenting cells to recruit the adaptive immune system [4]. Further, as humans are exposed to the same bacteria from which Cas9 is often derived (S. pyogenes and S. aureus), it is possible to develop a pre-existing immunity against Cas9 [7,57,58]. This preexisting immunity could inactivate Cas9 and negate the therapeutic effects of genome editors in vivo [7,57]. To reduce the immunogenic potential of bacterial-derived WT-Cas9 proteins, it is possible to synthetically alter Cas9 by mutating the precise epitopes that are recognized by T-cells while maintaining its nuclease activity [59]. An alternative approach is to use orthogonal Cas9 proteins sequentially [7] or create a Cas-like protein using human homologous parts, as demonstrated by engineering an epitranscriptomic regulator with comparable results to current CRISPR RNA-targeting systems [60].
The SpCas9 protein can induce a humoral immune reaction in animal models [61], in some cases leading to enlarged lymph nodes and activation of CD4+ T cells [62]. However, animal models do not accurately recapitulate safety profiles in humans due to differences in the immune system of human versus mouse, particularly in the major histocompatibility complex antigens [63]. The differences between animals and humans reinforce the need to engineer human microphysiological immune tissue platforms that can be used to investigate the safety of CRISPR-based editors.
An idealized human immunocompetent model for testing genome editors should have multicellular architecture and should be reproducible with controllable experimental parameters to accurately represent the innate and adaptive immune system. Primary human immune organs or tissues, such as, lymph node, lymphatic vessels, spleen, thymus, tonsils, or leukocyte-producing bone marrow platforms could facilitate genome editor testing in a physiologically relevant way [64]. However, ex vivo models of the human adaptive immune system are challenging to create because of their complexity in immune cell maturation and the need of specialized niches (recently reviewed in refs [64,65]). For instance, lymphoid T cells, involved in pathogen detection via antigen-presentation dependency, mature in the thymus, whereas B cells, involved in antibody production for long-term immunity, first mature in the bone marrow then differentiate in secondary lymphoid nodes [64]. As part of the adaptive immune system, both B and T cells are derived from hematopoietic stem cells that reside in the bone marrow. Multicellular systems that recapitulate some aspects of the innate immune system (e.gs., dendritic cells, macrophages) have been a major focus in the engineering of tissue chips. For instance, gut and liver systems have been developed, as these two organs contribute considerably to the overall immunological state of the body and are important in drug discovery [66–69].
Although current multicellular immune competent models were not designed to test CRISPR-Cas genome editors, some of them can be useful to study innate and potentially adaptive immune activation. Liver chip models can mimic the innate immune system as they consist of hepatocytes, endothelial cells, Kupffer cells (resident liver macrophages) and stellate cells, all combined in a microfluidic device to provide oxygen to maintain functional hepatocytes [70,71]. The addition of an inflammatory cue (e.g., lipopolysaccharides (LPS)) causes the release of inflammatory cytokines IL-6 and TNFa [72]. Likewise, advanced models of gut-liver interaction with innate and adaptive immune cells can be achieved by combining dendritic cells, macrophages, Treg, and Th7 cells, in a gut chip model and hepatocytes and Kupffer cells, in the liver chip. These chips allow the study of inflammatory gut-liver crosstalk in healthy and ulcerative colitis gut models [73]. Collectively, these gut and liver immune competent models respond to exogenously added cues (e.g., LPS) and therefore can be adapted to study genome editor effects in tissues during homeostasis and inflammatory processes.
In summary, some multicellular innate immune competent models (e.gs. gut and liver) are well characterized and their reproducibility is well documented, thus they can be easily adapted to model genome editor toxicity. Multicellular models of adaptive immunity are in their infancy and will need to be validated before being combined with genome editor testing. Ideally, interconnected tissue chips of the adaptive and innate immune system, especially from cells derived from the same donor, could provide a powerful platform to test genome editors in a physiologically relevant way.
6. Other Limitations of Current Multicellular Systems
Although current multicellular systems can be adapted to test genome editors, there are still some technical challenges that need to be addressed. For instance, as the number and diversity of cells that constitute a given multicellular system increases, it will be necessary to identify appropriate media formulations that support a diverse population of cells. This is critical to allow autocrine and paracrine signaling loops that occur in vivo. Likewise, as the cellular complexity of the ex vivo model increases, there will be a need to design the multicellular systems with the appropriate ratio of cells that can capture in vivo physiology (scaling), especially when the magnitude of cell-cell crosstalk is important to reach metabolic concentrations of cell-secreted factors for correct paracrine signaling or in vivo-like tissue injury response [32,33].
One of the most pressing challenges will be to build multicellular systems using cells from a single donor because using cells from different donors could result in unexpected cell crosstalk or unintended tissue function that does not accurately represent human physiology. This is vital in multicellular systems that will include cells from the adaptive immune system to mimic local or systemic inflammation states. A possible solution is to derive all the cell types from a single iPSC donor, either by the addition of tissue morphogens or via synthetic biology approaches [11,74]. These cells, however, often only mature to a fetal-like state [33,35], thus do not represent adult tissue, providing an important area of future studies in which synthetic biology and genome engineering approaches could turn iPSC-derived immature cells into adultlike tissue.
In addition to the technical and biological challenges described above, the increase in cellular diversity in multicellular systems decreases the ability to correlate an observed response to a given cell type, secreted metabolite, or autocrine and paracrine signaling, so there will be a need to establish metrics for validation and identification of biological outcomes (Table 2). An approach to gain insights into the biology of these multicellular systems is to perform multi-omics (e.gs. RNAseq, metabolomics) analyses. Although possible with a single multicellular system from a single donor, comparing several donors and experimental conditions often results in enormous amounts of data that needs to be interpreted using systems biology approaches and will need further validation to be used as predictor for in-vitro-to-in-vivo-extrapolation [75]. Nevertheless, the establishment of the Somatic Cell Genome Editing working group by the NIH to bring together experts in different fields to share reagents, protocols, unpublished data, and established guidelines for genome editing are moving the field in the right direction [76].
Figure 1. Overall strategy to evaluate genome editors using ex vivo multicellular systems.

Top panel, CRISPR-Cas genome editors and delivery vector engineering (1), can be validated using in silico approaches (2), that further move into the pipeline to be tested in ex vivo multicellular system that recapitulate some aspect of human physiology (3) to predict off-target effects in vivo and establish models for clinical translation (4-5). Bottom panel describes the opportunities and challenges when developing genome editors at each step in the pipeline. Note to editor. Image should be in color.
Acknowledgments:
This paper was supported partly by funds from the Pittsburgh liver research center, PLRC (NIH/NIDDK P30DK120531), the National Institutes of Health, NIH 8-U01-EB029372-02. The National Institute of Biomedical Imaging and Bioengineering, R01 (EB028532) and from the National Heart, Lung, and Blood Institute, R01 (HL141805).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Disclosure: Samira Kiani is the co-founder of SafeGen Therapeutics Inc.
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
References
* Special Interest
** Outstanding interest
- [1].Doudna JA, The promise and challenge of therapeutic genome editing, Nature. 578 (2020) 229–236. doi: 10.1038/s41586-020-1978-5. [DOI] [PMC free article] [PubMed] [Google Scholar]; ** Describes the field of genome engineering, differentiating between current in vivo and ex vivo approaches, as well as the many Cas orthologs that are available for genome editing. Doudna gives full descriptions of delivery barriers and progress (tissue specific; viral/non-viral); accuracy, precision, safety; and diseases that genome engineering could be applied to.
- [2].Wang D, Zhang F, Gao G, CRISPR-Based Therapeutic Genome Editing: Strategies and In Vivo Delivery by AAV Vectors, Cell. 181 (2020) 136–150.doi: 10.1016/j.cell.2020.03.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Mehta A, Merkel OM, Immunogenicity of Cas9 Protein, J. Pharm. Sci 109 (2020) 62–67. doi: 10.1016/j.xphs.2019.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Kim S, Koo T, Jee HG, Cho HY, Lee G, Lim DG, Shin HS, Kim JS, CRISPR RNAs trigger innate immune responses in human cells, Genome Res. 28 (2018) 367–373. doi: 10.1101/gr.231936.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Kleinstiver BP, Pattanayak V, Prew MS, Tsai SQ, Nguyen NT, Zheng Z, Joung JK, High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off-target effects, Nature. 529 (2016) 490–495. doi: 10.1038/nature16526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Kleinstiver BP, Sousa AA, Walton RT, Tak YE, Hsu JY, Clement K, Welch MM, Horng JE, Malagon-Lopez J, Scarfò I, Maus MV, Pinello L, Aryee MJ, Joung JK, Engineered CRISPR-Cas12a variants with increased activities and improved targeting ranges for gene, epigenetic and base editing, Nat. Biotechnol 37 (2019) 276–282. doi: 10.1038/s41587-018-0011-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Moreno AM, Palmer N, Alemán F, Chen G, Pla A, Jiang N, Leong Chew W, Law M, Mali P, Immune-orthogonal orthologues of AAV capsids and of Cas9 circumvent the immune response to the administration of gene therapy, Nat. Biomed. Eng 3 (2019) 806–816. doi: 10.1038/s41551-019-0431-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Nobles CL, Reddy S, Salas-Mckee J, Liu X, June CH, Melenhorst JJ, Davis MM, Zhao Y, Bushman FD, IGUIDE: An improved pipeline for analyzing CRISPR cleavage specificity Jin-Soo Kim, Genome Biol. 20 (2019) 14. doi: 10.1186/s13059-019-1625-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Wienert B, Wyman SK, Richardson CD, Yeh CD, Akcakaya P, Porritt MJ, Morlock M, Vu JT, Kazane KR, Watry HL, Judge LM, Conklin BR, Maresca M, Corn JE, Unbiased detection of CRISPR off-targets in vivo using DISCOVER-Seq, Science (80-. ). 364 (2019) 286–289. doi: 10.1126/science.aav9023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Ronaldson-Bouchard K, Vunjak-Novakovic G, Organs-on-a-Chip: A Fast Track for Engineered Human Tissues in Drug Development, Cell Stem Cell. 22 (2018) 310–324. doi: 10.1016/j.stem.2018.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Sharma A, Sances S, Workman MJ, Svendsen CN, Multi-lineage Human iPSC-Derived Platforms for Disease Modeling and Drug Discovery, Cell Stem Cell. 26 (2020) 309–329. doi: 10.1016/j.stem.2020.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Takebe T, Zhang B, Radisic M, Synergistic Engineering: Organoids Meet Organs-on-a-Chip, Cell Stem Cell. 21 (2017) 297–300. doi: 10.1016/j.stem.2017.08.016. [DOI] [PubMed] [Google Scholar]
- [13].Lancaster MA, Knoblich JA, Organogenesisin a dish: Modeling development and disease using organoid technologies, Science (80-. ). 345 (2014). doi: 10.1126/science.1247125. [DOI] [PubMed] [Google Scholar]
- [14].Takebe T, Wells JM, Organoids by design, Science (80-. ). 364 (2019) 956–959. doi: 10.1126/science.aaw7567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Kretzschmar K, Clevers H, Organoids: Modeling Development and the Stem Cell Niche in a Dish, Dev. Cell 38 (2016) 590–600. doi: 10.1016/j.devcel.2016.08.014. [DOI] [PubMed] [Google Scholar]
- [16].McCauley HA, Wells JM, Pluripotent stem cell-derived organoids: using principles of developmental biology to grow human tissues in a dish, Development. 144 (2017) 958–962. doi: 10.1242/dev.140-731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Schwank G, Koo BK, Sasselli V, Dekkers JF, Heo I, Demircan T, Sasaki N, Boymans S, Cuppen E, Van Der Ent CK, Nieuwenhuis EES, Beekman JM, Clevers H, Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients, Cell Stem Cell. 13 (2013) 653–658. doi: 10.1016/j.stem.2013.11.002. [DOI] [PubMed] [Google Scholar]; * Landmark paper demonstrating the utility of CRISPR-Cas editing to repair the CFTR genes in patient-derived intestinal organoid. This paper set the stage for the potential application of CRISPR-Cas editing in vivo on monogenetic diseases.
- [18].Vaidyanathan S, Salahudeen AA, Sellers ZM, Bravo DT, Choi SS, Batish A, Le W, Baik R, de la O S, Kaushik MP, Galper N, Lee CM, Teran CA, Yoo JH, Bao G, Chang EH, Patel ZM, Hwang PH, Wine JJ, Milla CE, Desai TJ, Nayak JV, Kuo CJ, Porteus MH, High-Efficiency, Selection-free Gene Repair in Airway Stem Cells from Cystic Fibrosis Patients Rescues CFTR Function in Differentiated Epithelia, Cell Stem Cell. 26 (2020) 161–171.e4. doi: 10.1016/j.stem.2019.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Geurts MH, de Poel E, Amatngalim GD, Oka R, Meijers FM, Kruisselbrink E, van Mourik P, Berkers G, de Winter-de Groot KM, Michel S, Muilwijk D, Aalbers BL, Mullenders J, Boj SF, Suen SWF, Brunsveld JE, Janssens HM, Mall MA, Graeber SY, van Boxtel R, van der Ent CK, Beekman JM, Clevers H, CRISPR-Based Adenine Editors Correct Nonsense Mutations in a Cystic Fibrosis Organoid Biobank, Cell Stem Cell. 26 (2020) 503–510.e7. doi: 10.1016/j.stem.2020.01.019. [DOI] [PubMed] [Google Scholar]; ** The authors demonstrate the repair of CFTR mutations via adenine base editors in an organoid biobank derived from 664 patients. Further, there were no detectable off-target effects found in whole genome sequencing, implying possible safety of adenine base editors in the clinic.
- [20].Roper J, Yilmaz ÖH, Breakthrough Moments: Genome Editing and Organoids, Cell Stem Cell. 24 (2019) 841–842. doi: 10.1016/j.stem.2019.05.008. [DOI] [PubMed] [Google Scholar]
- [21].Fujii M, Clevers H, Sato T, Modeling Human Digestive Diseases With CRISPR-Cas9–Modified Organoids, Gastroenterology. 156 (2019) 562–576. doi: 10.1053/j.gastro.2018.11.048. [DOI] [PubMed] [Google Scholar]
- [22].Sachs N, de Ligt J, Kopper O, Gogola E, Bounova G, Weeber F, Balgobind AV, Wind K, Gracanin A, Begthel H, Korving J, van Boxtel R, Duarte AA, Lelieveld D, van Hoeck A, Ernst RF, Blokzijl F, Nijman IJ, Hoogstraat M, van de Ven M, Egan DA, Zinzalla V, Moll J, Boj SF, Voest EE, Wessels L, van Diest PJ, Rottenberg S, Vries RGJ, Cuppen E, Clevers H, A Living Biobank of Breast Cancer Organoids Captures Disease Heterogeneity, Cell. 172 (2018) 373–386.e10. doi: 10.1016/j.cell.2017.11.010. [DOI] [PubMed] [Google Scholar]
- [23].Scott DA, Zhang F, Implications of human genetic variation in CRISPR-based therapeutic genome editing, Nat. Med 23 (2017) 1095–1101. doi: 10.1038/nm.4377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].de Souza N, Organoids, Nat. Methods 15 (2018) 23. doi: 10.1038/nmeth.4576. [DOI] [Google Scholar]
- [25].Eisenstein M, Organoids: the body builders, Nat. Methods 15 (2018) 19–22. doi: 10.1038/nmeth.4538. [DOI] [Google Scholar]
- [26].Takebe T, Enomura M, Yoshizawa E, Kimura M, Koike H, Ueno Y, Matsuzaki T, Yamazaki T, Toyohara T, Osafune K, Nakauchi H, Yoshikawa HY, Taniguchi H, Vascularized and Complex Organ Buds from Diverse Tissues via Mesenchymal Cell-Driven Condensation, Cell Stem Cell. 16 (2015) 556–565. doi: 10.1016/j.stem.2015.03.004. [DOI] [PubMed] [Google Scholar]
- [27].Grebenyuk S, Ranga A, Engineering Organoid Vascularization, Front. Bioeng. Biotechnol 7 (2019) 39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Velazquez JJ, LeGraw R, Moghadam F, Tan Y, Kilbourne J, Hislop J, Liu S, Cats D, de S. Lopes SMC, Plaisier C, Cahan P, Kiani S, Ebrahimkhani MR, Synthetic Maturation of Multilineage Human Liver Organoids via Genetically Guided Engineering, BioRxiv. (2020) 2020.05.10.087445. doi: 10.1101/2020.05.10.087445. [DOI] [Google Scholar]
- [29].Ronaldson-Bouchard K, Ma SP, Yeager K, Chen T, Song LJ, Sirabella D, Morikawa K, Teles D, Yazawa M, Vunjak-Novakovic G, Advanced maturation of human cardiac tissue grown from pluripotent stem cells, Nature. 556 (2018) 239–243. doi: 10.1038/s41586-018-0016-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Garreta E, Prado P, Tarantino C, Oria R, Fanlo L, Martí E, Zalvidea D, Trepat X, Roca-Cusachs P, Gavaldà-Navarro A, Cozzuto L, Campistol JM, Izpisúa Belmonte JC, Hurtado del Pozo C, Montserrat N, Fine tuning the extracellular environment accelerates the derivation of kidney organoids from human pluripotent stem cells, Nat. Mater 18 (2019) 397–405. doi: 10.1038/s41563-019-0287-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Homan KA, Gupta N, Kroll KT, Kolesky DB, Skylar-Scott M, Miyoshi T, Mau D, Valerius MT, Ferrante T, Bonventre JV, Lewis JA, Morizane R, Flow-enhanced vascularization and maturation of kidney organoids in vitro, Nat. Methods 16 (2019) 255–262. doi: 10.1038/s41592-019-0325-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Ronaldson-Bouchard K, Vunjak-Novakovic G, Organs-on-a-Chip: A Fast Track for Engineered Human Tissues in Drug Development, Cell Stem Cell. 22 (2018) 310–324. doi: 10.1016/j.stem.2018.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Zhang B, Korolj A, Lai BFL, Radisic M, Advances in organ-on-a-chip engineering, Nat. Rev. Mater 3 (2018) 257–278. doi: 10.1038/s41578-018-0034-7. [DOI] [Google Scholar]; * This review details the use of organs-on-chips for modeling human physiology, with emphasis on tissue barrier properties, parenchymal tissue function, and multi-organ interactions. The authors describe cell sourcing, functional hallmarks that are tested within each organ-on-chip, and the disease models and drugs tested within each model.
- [34].Park SE, Georgescu A, Huh D, Organoids-on-a-chip, Science (80-. ) 364 (2019) 960–965. doi: 10.1126/science.aaw7894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Ware BR, Khetani SR, Engineered Liver Platforms for Different Phases of Drug Development, Trends Biotechnol. 35 (2017) 172–183. doi: 10.1016/j.tibtech.2016.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Novak R, Ingram M, Marquez S, Das D, Delahanty A, Herland A, Maoz BM, Jeanty SSF, Somayaji MR, Burt M, Calamari E, Chalkiadaki A, Cho A, Choe Y, Chou DB, Cronce M, Dauth S, Divic T, Fernandez-Alcon J, Ferrante T, Ferrier J, FitzGerald EA, Fleming R, Jalili-Firoozinezhad S, Grevesse T, Goss JA, Hamkins-Indik T, Henry O, Hinojosa C, Huffstater T, Jang KJ, Kujala V, Leng L, Mannix R, Milton Y, Nawroth J, Nestor BA, Ng CF, O’Connor B, Park TE, Sanchez H, Sliz J, Sontheimer-Phelps A, Swenor B, Thompson G, Touloumes GJ, Tranchemontagne Z, Wen N, Yadid M, Bahinski A, Hamilton GA, Levner D, Levy O, Przekwas A, Prantil-Baun R, Parker KK, Ingber DE, Robotic fluidic coupling and interrogation of multiple vascularized organ chips, Nat. Biomed. Eng 4 (2020) 407–420. doi: 10.1038/s41551-019-0497-x. [DOI] [PMC free article] [PubMed] [Google Scholar]; **Functional coupling of up to ten organ chips was enabled by robotic liquid-handling to allow for physiologically relevant vascular perfusion. The integration of in situ microscopy imaging and in silico modeling allowed for the prediction and measurement of pharmacodynamics in a “body-on-a-chip”.
- [37].Herland A, Maoz BM, Das D, Somayaji MR, Prantil-Baun R, Novak R, Cronce M, Huffstater T, Jeanty SSF, Ingram M, Chalkiadaki A, Benson Chou D, Marquez S, Delahanty A, Jalili-Firoozinezhad S, Milton Y, Sontheimer-Phelps A, Swenor B, Levy O, Parker KK, Przekwas A, Ingber DE, Quantitative prediction of human pharmacokinetic responses to drugs via fluidically coupled vascularized organ chips, Nat. Biomed. Eng 4 (2020) 421–436. doi: 10.1038/s41551-019-0498-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Edington CD, Chen WLK, Geishecker E, Kassis T, Soenksen LR, Bhushan BM, Freake D, Kirschner J, Maass C, Tsamandouras N, Valdez J, Cook CD, Parent T, Snyder S, Yu J, Suter E, Shockley M, Velazquez J, Velazquez JJ, Stockdale L, Papps JP, Lee I, Vann N, Gamboa M, Labarge ME, Zhong Z, Wang X, Boyer LA, Lauffenburger DA, Carrier RL, Communal C, Tannenbaum SR, Stokes CL, Hughes DJ, Rohatgi G, Trumper DL, Cirit M, Griffith LG, Interconnected Microphysiological Systems for Quantitative Biology and Pharmacology Studies, Sci. Rep 8 (2018) 1–18. doi: 10.1038/s41598-018-22749-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E, A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity, Science (80-. ). 337 (2012) 816–821. doi: 10.1126/science.1225829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Wang D, Zhang F, Gao G, CRISPR-Based Therapeutic Genome Editing: Strategies and In Vivo Delivery by AAV Vectors, Cell. 181 (2020) 136–150. doi: 10.1016/j.cell.2020.03.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Ran FA, Cong L, Yan WX, Scott DA, Gootenberg JS, Kriz AJ, Zetsche B, Shalem O, Wu X, Makarova KS, Koonin EV, Sharp PA, Zhang F, In vivo genome editing using Staphylococcus aureus Cas9, Nature. 520 (2015) 186–191. doi: 10.1038/nature14299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Jiang F, Doudna JA, CRISPR–Cas9 Structures and Mechanisms, Annu. Rev. Biophys 46 (2017) 505–529. doi: 10.1146/annurev-biophys-062215-010822. [DOI] [PubMed] [Google Scholar]
- [43].Kosicki M, Tomberg K, Bradley A, Repair of double-strand breaks induced by CRISPR–Cas9 leads to large deletions and complex rearrangements, Nat. Biotechnol 36 (2018) 765–771. doi: 10.1038/nbt.4192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Komor AC, Badran AH, Liu DR, CRISPR-Based Technologies for the Manipulation of Eukaryotic Genomes, Cell. 168 (2017) 20–36. doi: 10.1016/j.cell.2016.10.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Anzalone AV, Randolph PB, Davis JR, Sousa AA, Koblan LW, Levy JM, Chen PJ, Wilson C, Newby GA, Raguram A, Liu DR, Search-and-replace genome editing without double-strand breaks or donor DNA, Nature. 576 (2019) 149–157. doi: 10.1038/s41586-019-1711-4. [DOI] [PMC free article] [PubMed] [Google Scholar]; ** The authors describe a new paradigm of genome editing, called Prime Editing, in which dCas9 is fused to an engineered reverse transcriptase and combined with new gRNAs. This application allows for targeted indels, as well as all 12 types of point mutations, all without inducing a double stranded break.
- [46].Cox DBT, Gootenberg JS, Abudayyeh OO, Franklin B, Kellner MJ, Joung J, Zhang F, RNA editing with CRISPR-Cas13, Science (80-. ). 358 (2017) 1019–1027. doi: 10.1126/science.aaq0180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Yin H, Kauffman KJ, Anderson DG, Delivery technologies for genome editing, Nat. Rev. Drug Discov 16 (2017) 387–399. doi: 10.1038/nrd.2016.280. [DOI] [PubMed] [Google Scholar]
- [48].Glass Z, Lee M, Li Y, Xu Q, Engineering the Delivery System for CRISPR-Based Genome Editing, Trends Biotechnol. 36 (2018) 173–185. doi: 10.1016/j.tibtech.2017.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Rouet R, Thuma BA, Roy MD, Lintner NG, Rubitski DM, Finley JE, Wisniewska HM, Mendonsa R, Hirsh A, de On L, Compte Barro J, McLellan TJ, Bellenger J, Feng X, Varghese A, Chrunyk BA, Borzilleri K, Hesp KD, Zhou K, Ma N, Tu M, Dullea R, McClure KF, Wilson RC, Liras S, Mascitti V, Doudna JA, Receptor-Mediated Delivery of CRISPR-Cas9 Endonuclease for Cell-Type-Specific Gene Editing, (2018). doi: 10.1021/jacs.8b01551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Hendel A, Fine EJ, Porteus MH, Quantifying on-and off-target genome editing, Trends Biotechnol. 33 (2015) 132–140. doi: 10.1016/j.tibtech. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Lee CM, Cradick TJ, Fine EJ, Bao G, Nuclease target site selection for maximizing on-target activity and minimizing off-target effects in genome editing, Mol. Ther 24 (2016) 475–487. doi: 10.1038/mt.2016.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Gkazi SA, Quantifying CRISPR off-target effects, Emerg. Top. Life Sci 3 (2019) 327–334. doi: 10.1042/etls20180146. [DOI] [PubMed] [Google Scholar]
- [53].Sentmanat MF, Peters ST, Florian CP, Connelly JP, Pruett-Miller SM, A Survey of Validation Strategies for CRISPR-Cas9 Editing, Sci. Rep 8 (2018) 1–8. doi: 10.1038/s41598-018-19441-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Tsai SQ, Nguyen NT, Malagon-Lopez J, Topkar VV, Aryee MJ, Joung JK, CIRCLE-seq: A highly sensitive in vitro screen for genome-wide CRISPR-Cas9 nuclease off-targets, Nat. Methods 14 (2017) 607–614. doi: 10.1038/nmeth.4278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Tsai SQ, Joung JK, Defining and improving the genome-wide specificities of CRISPR-Cas9 nucleases, Nat. Rev. Genet 17 (2016) 300–312. doi: 10.1038/nrg.2016.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Dunbar CE, High KA, Joung JK, Kohn DB, Ozawa K, Sadelain M, Gene therapy comes of age, Science (80-. ). 359 (2018). doi: 10.1126/science.aan4672. [DOI] [PubMed] [Google Scholar]; ** After a brief overview of the history of gene therapy, the authors describe the use of CRISPR and other genome engineering tools to treat diseases in the clinic. In addition to specific clinical trials and diseases that are being targeted, the authors show the promise of pursuing both in vivo and ex vivo approaches to gene therapy.
- [57].Charlesworth CT, Deshpande PS, Dever DP, Camarena J, Lemgart VT, Cromer MK, Vakulskas CA, Collingwood MA, Zhang L, Bode NM, Behlke MA, Dejene B, Cieniewicz B, Romano R, Lesch BJ, Gomez-Ospina N, Mantri S, Pavel-Dinu M, Weinberg KI, Porteus MH, Identification of preexisting adaptive immunity to Cas9 proteins in humans, Nat. Med 25 (2019) 249–254. doi: 10.1038/s41591-018-0326-x. [DOI] [PMC free article] [PubMed] [Google Scholar]; ** The authors demonstrated pre-existing immunity to Staphylococcus aureus and Streptococcus pyogenes in 78% and 58% of donors, respectively, by using an immunoabsorbant assay for anti-Cas9 antibodies. This was the first study demonstrating pre-existing immunity to common Cas9 orthologs.
- [58].Simhadri VL, McGill J, McMahon S, Wang J, Jiang H, Sauna ZE, Prevalence of Preexisting Antibodies to CRISPR-Associated Nuclease Cas9 in the USA Population, Mol. Ther. - Methods Clin. Dev 10 (2018) 105–112. doi: 10.1016/j.omtm.2018.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Ferdosi SR, Ewaisha R, Moghadam F, Krishna S, Park JG, Ebrahimkhani MR, Kiani S, Anderson KS, Multifunctional CRISPR-Cas9 with engineered immunosilenced human T cell epitopes, Nat. Commun 10 (2019) 1–10. doi: 10.1038/s41467-019-09693-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Rauch S, He E, Srienc M, Zhou H, Zhang Z, Dickinson BC, Programmable RNA-Guided RNA Effector Proteins Built from Human Parts, Cell. 178 (2019) 122–134.e12. doi: 10.1016/j.cell.2019.05.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Wang D, Mou H, Li S, Li Y, Hough S, Tran K, Li J, Yin H, Anderson DG, Sontheimer EJ, Weng Z, Gao G, Xue W, Adenovirus-Mediated Somatic Genome Editing of Pten by CRISPR/Cas9 in Mouse Liver in Spite of Cas9-Specific Immune Responses, Hum. Gene Ther 26 (2015) 432–442. doi: 10.1089/hum.2015.087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Chew WL, Tabebordbar M, Cheng JKW, Mali P, Wu EY, Ng AHM, Zhu K, Wagers AJ, Church GM, A multifunctional AAV-CRISPR-Cas9 and its host response, Nat. Methods 13 (2016) 868–874. doi: 10.1038/nmeth.3993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Kenney LL, Shultz LD, Greiner DL, Brehm MA, Humanized Mouse Models for Transplant Immunology, Am. J. Transplant 16 (2016) 389–397. doi: 10.1111/ajt.13520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Kim S, Shah SB, Graney PL, Singh A, Multiscale engineering of immune cells and lymphoid organs, Nat. Rev. Mater 4 (2019) 355–378. doi: 10.1038/s41578-019-0100-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Polini A, del Mercato LL, Barra A, Zhang YS, Calabi F, Gigli G, Towards the development of human immune-system-on-a-chip platforms, Drug Discov. Today 24 (2019) 517–525. doi: 10.1016/j.drudis.2018.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Chassaing B, Kumar M, Baker MT, Singh V, Vijay-Kumar M, Mammalian gut immunity, Biomed. J 37 (2014) 246–258. doi: 10.4103/2319-4170.130922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Blériot C, Ginhoux F, Understanding the Heterogeneity of Resident Liver Macrophages, Front. Immunol. 10 (2019). doi: 10.3389/fimmu.2019.02694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Kasendra M, Luc R, Yin J, Manatakis DV, Kulkarni G, Lucchesi C, Sliz J, Apostolou A, Sunuwar L, Obrigewitch J, Jang KJ, Hamilton GA, Donowitz M, Karalis K, Duodenum intestine-chip for preclinical drug assessment in a human relevant model, Elife. 9 (2020). doi: 10.7554/eLife.50135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Shin W, Kim HJ, Intestinal barrier dysfunction orchestrates the onset of inflammatory host-microbiome cross-talk in a human gut inflammation-on-a-chip, Proc. Natl. Acad. Sci. U. S. A 115 (2018) E10539–E10547. doi: 10.1073/pnas.1810819115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].Jang KJ, Otieno MA, Ronxhi J, Lim HK, Ewart L, Kodella KR, Petropolis DB, Kulkarni G, Rubins JE, Conegliano D, Nawroth J, Simic D, Lam W, Singer M, Barale E, Singh B, Sonee M, Streeter AJ, Manthey C, Jones B, Srivastava A, Andersson LC, Williams D, Park H, Barrile R, Sliz J, Herland A, Haney S, Karalis K, Ingber DE, Hamilton GA, Reproducing human and cross-species drug toxicities using a Liver-Chip, Sci. Transl. Med 11 (2019). doi: 10.1126/scitranslmed.aax5516. [DOI] [PubMed] [Google Scholar]; * Using a microfluidic Liver-Chip with primary hepatocytes, endothelial cells, Kupffer cells, and stellate cells from rats, dogs, and humans, the authors convincingly described the mechanisms behind the differential species response to drug-induced toxicity. In doing so, this research bridged the divide between animal models and many current in vitro models for drug testing
- [71].Beckwitt CH, Clark AM, Wheeler S, Taylor DL, Stolz DB, Griffith L, Wells A, Liver ‘organ on a chip,’ Exp. Cell Res 363 (2018) 15–25. doi: 10.1016/j.yexcr.2017.12.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [72].Sarkar U, Rivera-Burgos D, Large EM, Hughes DJ, Ravindra KC, Dyer RL, Ebrahimkhani MR, Wishnok JS, Griffith LG, Tannenbaum SR, Metabolite Profiling and Pharmacokinetic Evaluation of Hydrocortisone in a Perfused Three-Dimensional Human Liver Bioreactor, Drug Metab. Dispos 43 (2015) 1091–1099. doi: 10.1124/dmd.115.063495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Trapecar M, Communal C, Velazquez J, Maass CA, Huang YJ, Schneider K, Wright CW, Butty V, Eng G, Yilmaz O, Trumper D, Griffith LG, Gut-Liver Physiomimetics Reveal Paradoxical Modulation of IBD-Related Inflammation by Short-Chain Fatty Acids, Cell Syst. 10 (2020) 223–239.e9. doi: 10.1016/j.cels.2020.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [74].Guye P, Ebrahimkhani MR, Kipniss N, Velazquez JJ, Schoenfeld E, Kiani S, Griffith LG, Weiss R, Genetically engineering self-organization of human pluripotent stem cells into a liver bud-like tissue using Gata6, Nat. Commun 7 (2016) 1–12. doi: 10.1038/ncomms10243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Fabian E, Gomes C, Birk B, Williford T, Hernandez TR, Haase C, Zbranek R, van Ravenzwaay B, Landsiedel R, In vitro-to-in vivo extrapolation (IVIVE) by PBTK modeling for animal-free risk assessment approaches of potential endocrine-disrupting compounds, Arch. Toxicol 93 (2019) 401–416. doi: 10.1007/s00204-018-2372-z. [DOI] [PubMed] [Google Scholar]
- [76].Perry ME, Valdes KM, Wilder E, Austin CP, Brooks PJ, Genome editing to ‘rewrite’ wrongs, Nat. Rev. Drug Discov 17 (2018) 689–690. doi: 10.1038/nrd.2018.91. [DOI] [PubMed] [Google Scholar]
