You are on page 1of 15

PROGRESS REPORT

www.advancedscience.com

Is it Time for Reviewer 3 to Request Human Organ Chip


Experiments Instead of Animal Validation Studies?
Donald E. Ingber

their premise is that the results they gen-


For the past century, experimental data obtained from animal studies have erate in their cell cultures will translate to
been required by reviewers of scientific articles and grant applications to humans. Because in vitro studies generally
validate the physiological relevance of in vitro results. At the same time, lack the natural three dimensional (3D) con-
pharmaceutical researchers and regulatory agencies recognize that results text, vascular flow, and physico-chemical
microenvironment of living tissues and or-
from preclinical animal models frequently fail to predict drug responses in
gans, as well as the multi-organ physiol-
humans. This Progress Report reviews recent advances in human ogy of whole organisms, many question the
organ-on-a-chip (Organ Chip) microfluidic culture technology, both with clinical relevance of findings obtained with
single Organ Chips and fluidically coupled human “Body-on-Chips” these simplified models. For this reason,
platforms, which demonstrate their ability to recapitulate human physiology most researchers who submit a grant ap-
plication or publication based on in vitro
and disease states, as well as human patient responses to clinically relevant
findings commonly expect to find at least
drug pharmacokinetic exposures, with higher fidelity than other in vitro one reviewer (the classic exasperating “Re-
models or animal studies. These findings raise the question of whether viewer 3”) who demands that additional an-
continuing to require results of animal testing for publication or grant funding imal experiments be carried out to validate
still makes scientific or ethical sense, and if more physiologically relevant their findings before the work could be ac-
human Organ Chip models might better serve this purpose. This issue is ceptable for publication or funding. This ar-
ticle seeks to provoke a conversation in the
addressed in this article in context of the history of the field, and advantages
scientific community by asking two simple
and disadvantages of Organ Chip approaches versus animal models are questions: does this make sense, and if not,
discussed that should be considered by the wider research community. is there a better alternative? I address these
questions by reviewing recent progress that
has been made using organoids and engi-
neered microphysiological systems (MPS)
1. Introduction with a focus on microfluidic organ-on-a-
chip (Organ Chip) culture technologies.
Biomedical research grant applications and manuscripts com-
monly begin with an explanation of the potential clinical rel-
evance of the work for human health. Most basic researchers 2. Why Do We Require Animal Studies to Validate
carry out experiments using in vitro models, and thus, key to In Vitro Findings?
Animal experiments have been the mainstay of scientific re-
D. E. Ingber search since the time of Aristotle, and there are good reasons
Wyss Institute for Biologically Inspired Engineering at Harvard University for this. Many molecular mechanisms, physiological processes,
Boston, MA 02115, USA
and disease states involve complex tissue- and organ-level struc-
E-mail: don.ingber@wyss.harvard.edu
tures and functions, as well as interplay among multiple organ
D. E. Ingber
Vascular Biology Program, Department of Surgery systems. The effects that drugs produce in our bodies not only
Boston Children’s Hospital and Harvard Medical School depend on their effects within individual cells; they are also gov-
Boston, MA 02115, USA erned by their pharmacokinetics (PK), which defines how their
D. E. Ingber levels change over time in flowing blood and in different tis-
Harvard John A. Paulson School of Engineering and Applied Sciences sues depending on the dynamics of their absorption, distribu-
Cambridge, MA 02138, USA
tion, metabolism, and excretion (ADME) behaviors. It is not pos-
The ORCID identification number(s) for the author(s) of this article sible to model these complex responses, disease phenotypes, and
can be found under https://doi.org/10.1002/advs.202002030 PK behaviors in vitro using conventional culture technology, and
© 2020 The Authors. Published by Wiley-VCH GmbH. This is an open hence, it has been difficult to study the relation between molec-
access article under the terms of the Creative Commons Attribution ular mechanism, pathogenesis, and effects of drugs or toxins in
License, which permits use, distribution and reproduction in any a meaningful way without carrying out animal experiments. An-
medium, provided the original work is properly cited. other excellent reason for doing animal studies is when experi-
DOI: 10.1002/advs.202002030 ments in humans would be unethical, for example, when testing

Adv. Sci. 2020, 2002030 2002030 (1 of 15) © 2020 The Authors. Published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

the effects of exposure to lethal toxins, high energy radiation, or tionalities, and organ-level responses not possible in the past.
potentially deadly pathogens, such as infection by Ebola or the Organoids, for example, are clusters of cells that contain a sub-
SARS-CoV-2 virus. set of self-renewing stem cells, which grow and self-organize into
Reviewers of grants and publications appreciate the impor- small closed spheres that undergo cytodifferentiation and recre-
tance of animal models for all of the above reasons, but per- ate 3D tissue-like structures and functions seen in living organs
haps more central to their argument is the ability of animals, and when grown embedded within solid extracellular matrix (ECM)
particularly mice, to be genetically engineered to mimic human gels in vitro (Figure 1).[7] Methods for generating organoids have
disease states. This has been one of the central tenets of basic been published for many organs, including intestine, lung, liver,
biomedical research for the past 40 years, particularly in studies kidney, and brain, among others, and because they contain stem
focused on discovery of new therapeutics to alleviate human suf- cells, they can be used to continuously expand organ-specific hu-
fering. But are animal models good predictors of human patient man cell populations for additional in vitro studies.
outcomes? The answer to this question might be surprising. Importantly, organoids generated with human patient-derived
Studies by the pharmaceutical industry, as well as regula- cells or human-induced pluripotent stem (iPS) cells have proven
tory and government agencies around the world, including the to be extremely valuable for studying mechanisms relating to dif-
United States Food and Drug Administration (FDA) and National ferentiation, morphogenesis, pathogenesis, and drug action, as
Institutes of Health (NIH), have consistently found that results well as discovery of disease biomarkers that act at the cell and tis-
from drug testing in preclinical animal models are poor predic- sue levels.[7] Thus, they have a key role to play in the preclinical
tors of human responses.[1–3] Estimates of failure rates range as drug development pipeline, and they may be used to replace an-
high 90%, and this relates to both lack of efficacy and safety prob- imal studies that are focused on cell and molecular mechanisms
lems when the drugs are evaluated in human clinical trials. of disease.
Many animal models, including genetically engineered mice, However, because organoids are closed structures that lack
can mimic human disease phenotypes; however, the underlying tissue–tissue interfaces, vascular flow, circulating immune cells,
molecular, cellular, and physiological mechanisms are often dis- and physiologically relevant mechanical cues, they cannot fully
tinct. For example, while mice can generate phenotypes reminis- recapitulate organ-level responses or be used to study drug ef-
cent of multi-system inflammatory responses in humans, such as fects under pharmacologically relevant conditions that depend
sepsis and acute respiratory distress (ARDS), the genomic path- upon dynamic drug PK exposure profiles. Integration of self-
ways that underlie these responses differ significantly.[4] Simi- organized capillary networks within 3D organoid cultures[8] and
larly, while mice can be engineered to develop neurofibrillary addition of immune cells to the surrounding ECM gel[9] may help
tangles similar to those seen in the brains of humans with to overcome some of these limitations. But there are still signif-
Alzheimer’s Disease, the molecular mechanisms uncovered in icant limitations, such as an inability control vascular architec-
mouse models and drugs developed based on these insights have ture and flow dynamics, and hence how chemicals, inflamma-
not translated well when the drugs were moved into the clinic.[5] tory molecules, drugs, and immune cells are transported to and
Perhaps this is why hundreds of drugs that were found to be ef- from these tissues as they are within living vascularized organs in
fective when tested on animals for diseases ranging from stroke our bodies. Because organoids are closed structures surrounded
to inflammation were not found to be active in humans, and why by a thick dense ECM, it is also difficult to measure of transport
many vaccines have similarly failed in clinical trials even after and absorption of nutrients, chemicals, or drugs across the ep-
working well in non-human primates.[6] Furthermore, recent ad- ithelial tissue, experimentally sample contents of the internal lu-
vances in the biotechnology industry have led to biologic thera- men, sustain co-cultures with living microbiome, and integrate
pies, such as therapeutic monoclonal antibodies, that are so spe- sensors for on-line functional measurement using these models.
cific for their human targets that they do not even cross react with
related molecules in non-human primates, and so there is no
other option than to test them in a human model. Finally, a major 3.2. Static Microphysiological Systems
issue that has always concerned the field of drug development is
that there may be many therapies out there that were killed be- Over the past decade, there also has been an explosion in the de-
cause they failed to produce positive results in animals, yet they velopment of advanced MPS, such as engineered 3D tissues and
could have potentially saved human lives. So why is Reviewer 3 microfluidic Organ Chips with dynamic flow. Some static MPS
still demanding that animal studies be carried out before scien- models that lack fluid flow can produce high levels of function-
tific findings can be shared openly with the wider community or ality and mimicry of tissue functions, and they have been used
funding is provided to advance a potentially important research in combination with primary human cells or iPS-derived cells
program? It is likely because they feel that there is no alternative; from patients to gain insight into human physiology and patho-
but this is not the case. genesis that would be difficult to accomplish using animal mod-
els. For example, contractile heart tissues engineered using iPS
3. Are there Viable In Vitro Alternatives to Animal cell-derived cardiomyocytes from patients with Barth syndrome
Models? were able to mimic key features of the cardiomyopathy these pa-
tients experience in vitro, and this work led to new mechanis-
3.1. Human Organoids tic insights into the pathogenesis of this disease.[10] Tissue engi-
neering with iPS-derived cardiomyocytes from human patients
Over the past decade, there have been many advances in culture along with physiological electrical conditioning also enabled de-
technology that provide levels of cytodifferentiation, tissue func- velopment of electrophysiologically distinct atrial and ventricular

Adv. Sci. 2020, 2002030 2002030 (2 of 15) © 2020 The Authors. Published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

Figure 1. Potential in vitro replacements for animal testing. Schematics of mice, organoids that grow as small closed spheres which undergo organ-
specific cyto- and histo-differentiation within 3D ECM gels, and microfluidic Organ Chips that may be lined with cells from organoids (dashed line),
iPS cell-derived cells, or primary cells. Organ Chips reconstitute tissue–tissue interfaces between organ-specific epithelium and endothelium, vascular
perfusion, interstitial flow or air–liquid interfaces, and mechanical cues (e.g., by applying cyclic strain via application of suction to hollow side chambers)
to mimic breathing or peristalsis motions. Circulating or resident immune cells, connective tissue cells, nerve cells, and other cell types can be integrated
into the Organ Chips as needed to recapitulate increasing levels of complexity.

tissues that mimicked chamber-specific drug responses and gene many types of animal testing (Figure 1). These devices may be
expression, which permitted modeling of polygenic left ventricu- lined with either primary cells, iPS cells, or cells isolated from
lar hypertrophy in vitro.[11] There are also numerous other exam- patient-derived organoids. Importantly, while organoids may be
ples of how static MPS models can mimic human cell and tissue maintained in perfusion cultures, the advantage of microfluidic
pathophysiology, as well as cellular responses to drug therapies, Organ Chips is that dynamic fluid flow is delivered through an
and so they too represent viable replacements for some animal endothelium-lined channel as it is in living organs, and hence nu-
studies.[12] However, these MPS models do not enable dynamic trients, wastes, and drugs are transferred back and forth across
flow required to mimic drug PK or support multi-organ coupling, the endothelial interface with surrounding tissues in the chip as
and hence, they only satisfy a subset of requirements for replac- occurs in vivo.
ing animal studies, much like organoids. Many Organ Chip designs have been explored,[13] however, a
common embodiment is a small microfluidic device the size of a
computer memory stick composed of an optically clear flexible
3.3. Microfluidic Organ Chips polymer, such as poly-dimethylsiloxane (PDMS), that contains
two linear hollow channels each less than 1 mm wide that are sep-
Microfluidic Organ Chips are a form of dynamic MPS that reca- arated by a thin porous membrane made of the same clear flexible
pitulate vascular perfusion, tissue–tissue interfaces, and organ- material (Figure 1).[14] By coating the membrane with ECM, it is
relevant mechanical motions, while also allowing integration of possible to culture and differentiate organ-specific epithelial cells
circulating immune cells, connective tissues cells, and complex on one side of the membrane while growing endothelium from
microbiome, which could serve as meaningful alternatives to the same organ on the other. Culture medium or whole blood

Adv. Sci. 2020, 2002030 2002030 (3 of 15) © 2020 The Authors. Published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

can be flowed through the endothelium-lined microchannel to cytokine production, and suppressed ciliary function of asthmat-
mimic vascular perfusion, and medium can be flowed through ics when stimulated with interleukin-13.[18] When these same
the epithelial channel as a form of interstitial fluid (e.g., in Liver Airway Chips were lined with epithelial cells from patients with
Chips) or the channel may be filled with air to create an air–liquid chronic obstructive pulmonary disease (COPD), they recapitu-
interface (ALI) (e.g., in Lung Chips). Immune cells, either cir- lated the selective cytokine hypersecretion and enhanced recruit-
culating (Figure 1) or resident, as well as connective tissue cells ment of circulating human neutrophils that are flowed through
(e.g., fibroblasts, astrocytes), may be incorporated into these de- the endothelium-lined vascular channel, as well as clinical exac-
vices as well. In addition, by applying cyclic suction to full height erbation by exposure to viral and bacterial toxins, which are seen
side chambers on the either side of the central microchannels clinically. This inflammatory disease model was also used to de-
in some devices, it is possible to rhythmically stretch and relax tect synergistic effects of lung endothelium and epithelium on
the side walls and attached porous membrane with the adher- cytokine production, identify potential biomarkers of COPD exac-
ent tissue–tissue interface (Figure 1), thereby mimicking organ- erbation, and measure responses to anti-inflammatory drugs that
relevant motions, such as breathing in the lung or peristalsis in suppress cytokine-induced recruitment of circulating immune
the intestine. cells. Because the model incorporates dynamic fluid flow, it was
Importantly, many different Organ Chips have been shown to possible to mimic recruitment of circulating immune cells and
recapitulate human physiology, disease states, host–microbiome their inhibition by anti-inflammatory drugs more effectively than
interactions, and responses to clinically relevant drug and radi- static Transwell cultures that contained the same tissue–tissue
ation exposures with a level of fidelity that is as good or better interface.
than animal models. For example, a Lung Alveolus Chip lined Furthermore, by connecting the Lung Airway Chips lined by
by human A549 lung alveolar epithelial cells and pulmonary vas- primary human epithelial cells to a cigarette smoking robot that
cular endothelium that experienced cyclic breathing movements breathes whole cigarette smoke in and out of the air channel of
reproduced many features of human lung physiology including the chips, it was possible to carry out matched comparative anal-
gas exchange and responses to bacteria, inflammatory cytokines, ysis of the same patients before and after smoke exposure. This
and airborne nanoparticulates introduced into the alveolar space, led to the identification of ciliary abnormalities, as well as COPD-
as well as revealing previously unknown effects of physiological specific cytokine and transcriptomic signatures (Figure 2), that
breathing on these responses.[14] The same chip mimicked drug closely matched those detected in prior clinical studies in smok-
toxicity-induced pulmonary edema observed in human patients ers who were otherwise healthy.[19] Given the anatomic and phys-
treated with the cancer drug interleukin-2 (IL-2) at a clinically iological differences between species, these studies could not be
relevant dose and over the same time frame, in addition to re- done as effectively in animal models, and particularly not in mice
vealing that physiological breathing motions were required for given their small size.
this response while immune cells were not.[15] Importantly, this Most recently, an influenza virus infection model was devel-
work led to the identification of a potential therapeutic (TRPV4 oped using the Lung Airway Chip, which replicates the virulence
ion channel inhibitor) and helped to enable its entry into human of different viral strains (e.g., H1N1 vs H3N2 and H5N1) and en-
clinical trials. ables quantification of host cytokine and immune cell responses,
A later version of the Lung Alveolus Chip lined with pri- as well as mimicry of clinical responses to antiviral therapies and
mary human lung alveolar cells and pulmonary microvascu- spontaneous viral evolution in response to drug exposure that
lar endothelium that supported flowing whole human blood is enabled by human-to-human transmission.[20,21] Microfluidic
through the device faithfully recapitulated organ-level contri- Airway Chips are now being used to model human lung infection
butions to platelet–endothelial dynamics and inflammation- by SARS-CoV-2 virus in vitro, and to repurpose existing FDA-
induced thrombosis.[16] Using this chip, it was discovered that approved drugs as potential COVID-19 therapeutics.[21,22] Impor-
bacterial lipopolysaccharide (LPS) endotoxin stimulates intravas- tantly, it is extremely difficult to develop animal models of viral
cular thrombosis via activation of the alveolar epithelium, rather infection, and none replicate human host responses as well as
than acting directly on endothelium. In addition, this device was these Organ Chip models that incorporate highly differentiated
leveraged to deliver a potential adenoviral vector-based gene ther- human lung cells grown under an ALI while exposed to dynamic
apy encoding a TRPV4 inhibiting peptide directly to the lung fluid flow at their base.
alveolar cells (i.e., through the airspace of the epithelial channel) The same chip design has been used to model the responses
while experience breathing motions, and this was able to sup- of human intestine to various stimuli in ways that are com-
press pulmonary vascular leakage.[17] Thus, in addition to mim- parable to, or more accurate than, animal models. Intestine
icking human responses to pathogens, toxins, drugs, and gene Chips that were initially lined by human Caco-2 intestinal ep-
therapies, these Organ Chip studies led to new insights into the ithelial cells,[23–25] and later by primary cells isolated from hu-
cellular and molecular basis of pathophysiology due to their abil- man patient-derived duodenal or colon organoids along with pri-
ity to enable independent control of cell composition, chemical mary human intestinal microvascular endothelium,[26,27] have
factors, fluid flow, and mechanical forces, which is not possi- been shown to exhibit histological appearance similar to that ob-
ble using either conventional cell culture, organoids, or animal served in vivo (e.g., villi-like structures in duodenum) (Figure 3A)
models. and high levels of goblet cell differentiation (Figure 3B), as well
Human Lung Airway Chips lined by primary human bron- as improved intestinal barrier function, enhanced drug metabo-
chiolar epithelium grown under an ALI in one microchan- lizing activities, and more mucus production than static cultures.
nel with an underlying microvascular endothelium in another Interestingly, when transcriptomic analysis was carried put, the
also can reconstitute the goblet cell hyperplasia, increased Small Intestine Chips more closely resembled living duodenum

Adv. Sci. 2020, 2002030 2002030 (4 of 15) © 2020 The Authors. Published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

Figure 2. Human Lung Airway Chip recreates airway epithelial architecture and replicates patient responses to cigarette smoke in vitro. A) Still image
captured from a video recording from the side of cilia beating on the apical surface of the differentiated airway epithelium on-chip. B) Scanning electron
micrograph of the apical surface of the airway epithelium formed on-chip showing ciliated cells (blue) and non-ciliated cells (brown). C) Graph showing
changes in interleukin 8 (IL-8) secretion in Lung Airway Chips lined with epithelial cells isolated from normal or COPD patients, with or without exposure
to whole cigarette smoke for 75 min (smoking) (**p < 0.01). D) Heatmap comparing expression of 29 genes associated with cellular oxidation–reduction
in lung airway epithelial cells obtained from small airways of different normal human smokers compared with samples obtained from three Lung Airway
Chips that were exposed to whole cigarette smoke on-chip. Note that the patterns of induced and suppressed genes in the Airway Chip mimic those
seen in human patients. (B) Reproduced with permission.[18] Copyright 2016, Springer Nature. (C,D) Reproduced with permission.[19] Copyright 2016,
Cell Press.

in vivo than the organoids from which they were derived (Fig- flammatory cytokines (IL-8, IL-6, IL-1𝛽, TNF-𝛼) elicited by LPS
ure 3C).[26] Moreover, the Colon Chips lined by organoid-derived endotoxin and human immune cells that are necessary and suffi-
cells spontaneously accumulate a mucus layer with the same im- cient to compromise intestinal barrier function.[25] Interestingly,
penetrable and penetrable layers, and the same thickness, as is use of mechanically active Organ Chips also revealed that ces-
observed in human colon, whereas this is not observed in the sation of mechanical peristalsis-like motions stimulates over-
organoids cultured under the same conditions without flow.[27] growth of bacteria,[26–28] as seen in patients with ileus and in-
Mucus accumulation in the Colon Chips also can be studied non- flammatory bowel disease (IBD). Moreover, similar results were
invasively over time on-chip using live imaging, which is not pos- obtained using a human Colon Chip, which showed that
sible in other cell cultures or animal models. peristalsis-like mechanical deformations influence infection by
Small Intestine Chips lined by cells isolated from patient- the human pathogen Shigella.[31] Finally, additional complexity
derived organoids have been used to study induction of CYP450 has been integrated into Intestine Chip models by establishing
metabolizing enzymes that can complicate the development of a hypoxia gradient across the tissue–tissue interface, which en-
new drugs, and provide insight into the molecular and cellular ba- ables extended co-culture of human intestinal epithelium in di-
sis of drug–drug interactions that can cause changes in drug PK, rect contact (via its natural overlying mucus layer) with complex
safety, and efficacy.[28] Relative to animal models, Small Intestine living human gut microbiome containing over 200 different types
Chips better mimic infection by enteric viruses[29] and intesti- of microbial strains over multiple days in vitro.[32] This is yet an-
nal injuries induced by acute exposure to 𝛾-radiation (including other advantage of human Organ Chips over static MPS cultures,
dosage sensitivities and responses to countermeasure therapies) organoids, and animal models, as they do not permit long-term
exhibited by humans as well.[30] human microbiome co-cultures.
Additional Small Intestine Chip studies showed that probi- Yet another example where Organ Chip experiments provide
otic and antibiotic therapies can prevent villus injury induced by advantages over animal studies relates to modeling the blood–
pathogenic bacteria, and this led to identification of four proin- brain barrier, which has distinct permeability properties and

Adv. Sci. 2020, 2002030 2002030 (5 of 15) © 2020 The Authors. Published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

lar cells isolated from patients also were able to recreate disease-
specific transporter deficiencies and barrier disruption.[34] More-
over, when the vascular lumen was perfused with whole blood,
the engineered blood–brain barrier protected neural cells from
plasma-induced toxicity and accurately predicted brain perme-
ability of drugs. These observations raise the possibility that hu-
man Blood–Brain Barrier Chips may provide a more physiologi-
cally relevant way to develop shuttle molecules for delivery of ther-
apeutic antibodies and other brain-targeting therapeutics than
mouse or non-human primate models, which are used today.
Different useful functionalities and biological mimicry has
been demonstrated using a human Bone Marrow Chip that main-
tains differentiation and maturation of multiple blood cell lin-
eages over 1 month by co-culturing CD34+ cells and marrow-
derived stromal cells in a fibrin gel in one channel interfaced with
human microvascular endothelium in the other (Figure 4A).[36]
This chip reproduces marrow toxicities induced by exposures to
clinically relevant doses of anti-cancer drugs (Figure 4B) or ioniz-
ing radiation, as well as bone marrow recovery after drug-induced
myelosuppression, whereas other preclinical models do not.
Importantly, using this microfluidic chip, it was possible to
recapitulate both drug exposure profiles that were previously
measured in PK studies in human patients (Figure 4C) and as-
sociated regimen-specific myeloerythroid toxicity responses pre-
viously measured in those same patients (Figure 4D). Equally
important, characteristic hematopoietic defects of a rare genetic
disorder (Shwachman–Diamond syndrome) were recapitulated
on-chip by incorporating cells into the Bone Marrow Chips that
Figure 3. Human Intestine Chips lined by cells isolated from patient- were isolated from patients, and this work led to new insight in
derived organoids exhibit differentiated structures and functions that the underlying pathogenesis of these disease. The ability of this
closely resemble those displayed by living intestine in vivo. A) Differen- Bone Marrow Chip to replicate drug PK-dependent effects, clin-
tial interference microscopic image (top) and immunofluorescence mi-
croscopic image of F-actin staining in green (bottom) of vertical sec-
ically relevant radiation sensitivities, and a rare human disease
tions through an Intestine Chip showing villus protrusions formed by phenotype using patient-specific cells under these dynamic mod-
primary intestinal (duodenal) epithelium cultured on-chip for 12 days. eling conditions, is a clear example of why human Organ Chips
B) Immunofluorescence microscopic image of a vertical section through should be considered as alternatives to animal models.
a human Colon Chip showing a high polarized epithelium with basolat- Human Organ Chips also have been used to create “ortho-
eral adherens junctions labeled with E-cadherin (green) and brush border topic” Cancer Chip models by growing tumor cells in organ-
stained for F-actin (white) restricted to the apical regions, Hoechst stained
relevant contexts, rather than relying on injecting human tu-
nuclei (blue) localized at the cell base, and goblet cells stained for Muc2
(magenta). C) A curated heatmap showing gene expression profiles in the mor cells into their organs of origin in mice. For example, when
mechanically active Intestine Chip (with fluid flow and cyclic stretching to human non-small-cell lung cancer (NSCLC) cells that arise at
mimic peristalsis-like motions) lined by cells from patient-derived duode- the bronchiolar–alveolar interface but grow preferentially within
nal organoids, the duodenal organoids from which the cells were derived, alveoli in patients, were cultured in human Lung Alveolus and
and duodenum in vivo. Note that the expression of pattern of the Intestine Airway Chips, they recapitulated these organ microenvironment-
Chip more closely resembles native intestine compared to the organoids.
specific behaviors by proliferating rapidly in the former and ex-
(A,C) Reproduced with permission.[26] Copyright 2018, Springer Nature.
(B) Reproduced with permission.[27] Copyright 2020, Elsevier Inc. hibiting tumor dormancy in the latter.[37] In addition, these mod-
els replicated differences in responses to tyrosine kinase inhibitor
(TKI) therapies observed in human patients, and revealed that
transport functions in humans. Human Blood–Brain Barrier breathing motions can influence lung cancer cell growth and in-
Chips have been developed that are lined by iPS cell-derived vasion, as well as therapeutic responses. Again, these insights
human brain microvascular endothelium interfaced with either would not be possible using animal models.
primary human brain pericytes and astrocytes[33] or human iPS- In another cancer study, a microfluidic Organ Chip model of
derived astrocytes and neurons,[37] which reconstitute the high the human bone perivascular niche containing vascular cell net-
permeability restrictions of the human barrier. When differen- works and marrow-derived mesenchymal stem cells within a na-
tiated appropriately, the iPS cell-derived endothelium expresses tive bone matrix with controlled flow hemodynamics and oxy-
high levels of human-specific efflux pumps and tight junction gen gradients revealed that human breast tumor cells exposed
proteins, which enables recapitulation of differential transport to interstitial flow persist in a dormant state on-chip that is as-
and selective transcytosis of antibodies and peptides, as well as ex- sociated with increased drug resistance.[38] A perfused model of
tracellular vesicles, that have been observed in vivo.[33–35] Blood– metastatic tumor seeding in liver that incorporates human hepa-
Brain Barrier Chips containing iPS-derived human neurovascu- tocytes, nonparenchymal liver cells, and breast cancer cells, and

Adv. Sci. 2020, 2002030 2002030 (6 of 15) © 2020 The Authors. Published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

Adv. Sci. 2020, 2002030 2002030 (7 of 15) © 2020 The Authors. Published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

which was instrumented with oxygen sensors and micropumps cherichia coli (EHEC) compared to mice.[45] Because fluids are
that enabled diurnal variations in nutrient and hormone profiles, continuously flowed through these chips, it was possible to carry
was similarly used to recreate the human-specific metastatic mi- out metabolomic analysis in this study using a Colon Chip, which
croenvironment and probe the paracrine effects between liver led to the identification of specific chemical metabolites that are
cells and metastatic tumor cells.[39] More recently, a microfluidic responsible for this different susceptibility to EHEC infection.
model containing a 3D collagen gel populated by human liver Rat, dog, and human Liver Chips containing primary hepato-
cancer cells in one channel was challenged with human T cells cytes, liver sinusoidal endothelium, Kupffer cells and stellate cells
engineered to express tumor-specific T cell receptors in an ad- also have been created that replicate species-specific hepatotoxi-
jacent channel while modulating oxygen levels and inflamma- cities induced by multiple drugs (Figure 5).[46] This is an impor-
tory cues to assess the preclinical efficacies of adoptive T cell tant finding because preclinical rat and dog liver toxicity studies
immunotherapies in an immunosuppressive environment.[40] are key requirements for regulatory approval, and they can of-
These types of studies can provide insight into human-specific ten produce conflicting results, leading to confusion or removal
cancer and immune cell responses that would not be possible of good drugs from the pipeline. These results suggest that hu-
with animal models. man Organ Chips offer a potential way to cross-validate these re-
Organ Chips with different fluidic designs have produced ad- sults between animals and humans early in the drug develop-
ditional evidence showing that this technological approach can ment process. These animal Organ Chips also might serve as re-
be used with primary or patient-derived cells to gain insight into placements for animal validation studies, for example, in veteri-
human-relevant cellular and molecular mechanisms of patho- nary applications or be used to benchmark in vitro results versus
genesis, as well as drug actions, more effectively than animal findings obtained in past animal experiments.
models. For instance, a human Organ Chip model of the kid- More sophisticated microenvironmental control can be inte-
ney proximal tubule was combined with quantitative systems grated into Organ Chips because of their microfluidic design. For
pharmacology-based computational models to enable in vitro- example, continuous oxygen zonation can be generated in Liver
to-in vivo translation (IVIVT) of experimental results for the Chips by regulating flow rate in the vascular and hepatic chan-
biomarker kidney injury molecule-1 (KIM-1), which predicted nels, which can provide insight into its contribution to human
human drug-induced renal toxicities, and helped to identify fa- liver physiology, toxicology, and disease progression, as well as
vorable dosing regimens.[41] The same approach was leveraged recruitment of circulating immune cells.[47] By populating per-
to uncover a unique mechanism by which an FDA-approved an- fused Liver Chips with freshly cryopreserved hepatocytes from
tibiotic drug (polymyxin) induces kidney injury, and to identify five different human donors, it also has been possible to quan-
related chemical analogues that do not induce this toxicity.[42] An- tify metabolic clearance rates for multiple drugs, and this led to
other Kidney Chip was used to model viral infection of the kidney the finding that albumin, urea, lactate dehydrogenase, and cy-
distal tubule and how it alters sodium reabsorption, which can tochrome P450 mRNA levels can serve as predictors of these
lead to serum electrolyte abnormalities.[43] Finally, a microfluidic pharmacologically relevant responses.[48] In addition, by overlay-
model of the kidney glomerulus lined by human iPS cell-derived ing a computational, population-level, physiologically based PK
kidney podocytes interfaced with kidney microvascular endothe- model, in vitro data from this study were related to their ob-
lium and exposed to cyclic mechanical deformations (mimicking served PK behavior in vivo, and model simulations predicted ob-
changes in blood flow with each beat of the heart) reconstituted served clinical concentration-time profiles and associated popu-
differential clearance of albumin and inulin via glomerular filtra- lation variability in drug metabolism. The same perfused Liver
tion, as well as albuminuria and podocyte injury induced by the Chip also was able to recapitulate the clinical impact of an anti-
cancer drug adriamycin.[44] IL-6R monoclonal antibody used to treat rheumatoid arthritis on
While most work in this field has focused on creating Organ small molecule drug PK, and it was shown to be mediated by
Chips lined by human cells, species-specific Organ Chips also modulation of cytochrome P450 enzyme activities.[49]
have been made that, for example, can replicate the much higher Another example where Organ Chips have been shown to
susceptibility of humans to infection by enterohemorrhagic Es- be a valuable alternative to animals is in ophthalmologic drug

Figure 4. Prediction of clinically observed hematotoxicities at patient-relevant drug exposures using a human Bone Marrow Chip. A) Left, schematic of
bone and insert showing normal human marrow histology. Left middle, schematic of the human Bone Marrow Chip at the time of seeding, showing
dispersed CD34+ progenitor cells and bone marrow stromal cells in an ECM gel filling the top channel, and a vascular endothelium beginning to line
the adjacent channel. Right middle, within 2 weeks of culture, the endothelium covers all four sides of the lower channel, creating a vascular lumen, and
the CD34+ cells undergo expansion and multilineage differentiation. Right, immunofluorescence view of a cross-section through the ECM gel grown
on-chip for 14 days (yellow, megakaryocyte lineage; magenta, erythroid lineage; blue, neutrophil and other hematopoietic lineages; EC, endothelial cell).
B) Effects of treating the Bone Marrow Chips, suspension cultures, and static ECM gel co-cultures for 48 h with various doses of the cancer drug, 5-
fluorouracil (5-FU) on total cell number (left) versus only neutrophil lineage cells (right)(***p < 0.001). The range of patient plasma 5-FU concentrations
for a 2 day infusion that are known to cause myelosuppression is indicated in orange. Note that the Bone Marrow Chip replicates this sensitivity to a
clinically relevant dose exposure whereas the other in vitro models do not. C) Graphs showing that dynamic changes in concentrations of the cancer drug
AZD2811 measured by mass spectrometry in chip outlet (circles), which were used to fit PK models of Bone Marrow Chip drug exposure (black line) for
2 and 48 h infusions. Note that these drug exposure profiles that were generated in the microfluidic Bone Marrow Chip closely resembled plasma levels
of AZD2811 in vivo (grey), which were simulated for an average patient at a range of clinical doses based on the known PK characteristics of AZD2811.
D) Graphs showing the effects of infusing the Bone Marrow Chips with varying doses of AZD2811 for 2 versus 48 h; total neutrophil (blue) and erythroid
(red) cell numbers on day 12 are shown (***p < 0.001). Note that the clinical observation of unusual regimen-specific neutropenia and anemia with 2
h dosing, but only neutropenia with 48 h dosing, was replicated on-chip. Reproduced with permission.[36] Copyright 2020, Springer Nature.

Adv. Sci. 2020, 2002030 2002030 (8 of 15) © 2020 The Authors. Published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

Figure 5. A human Liver Chip recapitulates species-specific drug toxicities in rat, dog, and human. A) Schematic of the Liver Chip that contains an upper
parenchymal channel lined by primary rat, dog, or human hepatocytes grown in ECM sandwich, while species-specific liver sinusoidal endothelial cells
(LSECs), Kupffer cells, and stellate cells are cultured on the opposite side of the same membrane in the lower vascular channel. B) Previously observed
species-specific effects of the drug bosentan on albumin secretion were recreated in microfluidic human, dog, and rat Liver Chips (Chip, open circles)
whereas this was not replicated in static hepatocyte sandwich monocultures (Plate, closed triangles). Reproduced with permission.[46] Copyright 2019,
American Association for the Advancement of Science.

development where results from animal experiments often do PK studies. Two microfluidic human Blood Brain Barrier Chips
not translate to humans. Recently, a Retina Chip was created (lined by primary brain microvascular endothelial cells interfaced
that is lined by retinal organoids derived from human iPS cells with human astrocytes and pericytes) were fluidically coupled via
that form complex stratified retinal tissues containing more than their interstitial (cerebral spinal fluid) channels before and af-
seven different retinal cell types, and that permits physiological ter a chip containing primary human brain neuronal networks
interactions between mature photoreceptor segments with reti- (Figure 6A–C) to model drug influx across the human blood–
nal pigmented epithelium.[50] This chip reproduced the retino- brain barrier, permeation into the brain parenchymal compart-
pathic side effects of the anti-malaria drug chloroquine and the ment, and efflux across the barrier.[54] This model of the hu-
antibiotic gentamicin in vitro, and now offers a preclinical hu- man neurovascular unit effectively mimicked the barrier open-
man model to study drug efficacy and toxicities in a way that is ing properties of the psychoactive drug methamphetamine. It
not possible in animal models. also permitted independent metabolomic analysis of each of the
Perhaps the clearest argument against using animal models different fluidic compartments of the neurovascular unit, which
comes from studies that show human Organ Chips can repli- revealed previously unknown metabolic coupling between the
cate pathophysiological responses observed in humans that were brain microvascular endothelium and neurons that would be im-
never detected in preclinical animal studies. One example is a possible to do in animal studies.
human Kidney Chip model of the proximal tubule that can repli- In a separate study, a microfluidic multi-organ model con-
cate toxicities induced by cisplatin through a human-specific Pgp taining gut, liver, and circulating Treg and Th17 cell compo-
efflux transporter that is not expressed in animals.[51] Another is nents was used to model the contributions of microbiome and
the demonstration that a human Blood Vessel Chip reproduces liver metabolism to IBD.[55] This work revealed that microbiome-
thrombotic toxicities in response to treatment with a therapeu- derived short-chain fatty acids can influence IBD severity based
tic monoclonal antibody that caused deaths in human clinical on the involvement of effector CD4 T cells. Multi-Organ Chip
trials even though this toxicity was not observed in preclinical systems also have been used to model preclinical drug toxicities
animal studies.[52] Finally, one challenge that is difficult for an- and metabolic conversion,[56] as well as sequential multi-organ
imal models is to faithfully mimic complex function of the hu- metabolism of drugs and xenobiotics that can impact therapeu-
man immune system, such as vaccination responses. However, tic action and side effects.[57] In the latter study, Organ Chips
recent studies suggest that this also may be possible using a hu- representing the jejunum, liver, and kidney (the major absorp-
man Organ Chip that supports self-assembly of human blood- tion, metabolism, and clearance organs) were evaluated, as well
derived B and T lymphocytes into germinal center-like lymphoid as skeletal muscle and neurovascular models, and organ-specific
follicles.[53] Also, the human Bone Marrow Chip supports con- ADME processing of various drugs was shown to be consistent
tinued production of multiple blood cell lineages in vitro, as well with human clinical data.
as spontaneous intravasation of more differentiated blood cells It has even been possible to study organism-level, hormonal
into the endothelium-lined vascular channel of these microflu- cross-talk, such as between insulin production in pancreas and
idic chips,[36] raising the possibility of creating fluidically linked glucose regulation in liver by fluidically coupling two microflu-
single or multiple Organ Chip systems that incorporate all cells idic chips lined by human pancreatic islet microtissues and liver
and tissues derived from the same patient, including circulating spheroids.[58] A functional hormonal feedback loop between the
immune cells. human liver and insulin-secreting islet tissues was demonstrated
in this study, which suggests that it might be useful for study-
3.4. Multi-Organ Chip Models ing diabetes in humans. Even more impressively, a functioning
female reproductive system that can be used to study the hu-
One of the greatest advantages of microfluidic Organ Chips man 28-day menstrual cycle and pregnancy-associated hormonal
over static engineered MPS and organoids is that multiple chips loops has been successfully recreated in vitro by fluidically cou-
can be linked fluidically to model multi-organ physiology and pling organ modules for the ovary, fallopian tube, uterus, cervix,
even create human “Body-on-Chips” models that can be used for and liver.[59]

Adv. Sci. 2020, 2002030 2002030 (9 of 15) © 2020 The Authors. Published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

Figure 6. Multi-Organ Chip model of the human neurovascular unit. A) Schematic of the influx Blood–Brain Barrier Chip containing brain microvascular
endothelium (magenta) in its vascular channel separated by a porous membrane from brain astrocytes (blue) and pericytes (yellow) in the parenchymal
channel through which medium mimicking cerebral spinal fluid (CSF) flows. The CSF fluid is transferred to a similar channel within a Brain Chip that is
separated by a porous membrane from a channel containing cultured human brain neuronal cells (green) and astrocytes (blue), and from there to the
parenchymal channel of a second efflux Blood–Brain Barrier Chip. Medium mimicking blood is flowed separately through the lower vascular channel. B)
3D confocal microscopic reconstruction of the Blood–Brain Barrier chip viewed from the side showing a continuous endothelium stained for VE-cadherin
(purple) forming a lumen in the lower vascular channel, as well as pericytes (F-actin, yellow) and astrocytes (GFAP, blue) on the top surface of the porous
membrane in the upper channel. C) Confocal fluorescence microscopic view of networks of human brain neurons (𝛽-III-tubulin, green) and astrocytes
(glial fibrillary astrocytic protein, GFAP, blue) in the lower compartment of the Brain Chip. Reproduced with permission.[54] Copyright 2018, Springer
Nature.

An outstanding example of a capability that Organ Chip tech- pharmacodynamics obtained in a fluidically linked Bone Marrow
nology offers that animal models do not relate to the modeling of Chip also matched previously reported patient data.[70] Together,
human drug PK properties using human Body-on-Chips mod- these studies show how human Body-on-Chips approaches can
els that can maintain up to ten different human Organ Chips enable quantitative IVIVT of human PK and pharmacodynamic
when fluidically coupled for 1 month in vitro.[60,61] PK studies parameters, as well as predict drug ADME properties and toxici-
have been carried out in many multi-Organ Chip models by ties, which is not currently possible with animal models.
transfering fluids between them, and analyzing drug levels and
metabolism using mass spectrometry.[59,62–65] In some studies, 4. Advantages and Disadvantages of Organ
qualitative predictions of drug toxicity responses have been made Chip Models
using PK and pharmacodynamic models with coupled Organ
Chips.[60,66–69] However, in these studies, the medium contain- From this analysis, it appears that human Organ Chips can
ing drug flowed directly from one parenchymal tissue type to an- provide much of the organ-level information that Reviewer 3 is
other without passing through the endothelium, which is crucial seeking when he or she requests additional animal validation
for defining drug PK behavior in vivo, and thus, the physiologi- studies, and even provide useful insight into multi-organ physiol-
cal accuracy of these results is limited. However, more recently, ogy. The presence of dynamic fluid flow, tissue–tissue interfaces,
it has been possible to quantitatively predict drug PK parame- and physiological mechanical cues enhances the fidelity of cell
ters (e.g., Cmax , T1/2 ) measured in human patients by applying differentiation and increases expression of tissue-specific func-
physiologically based PK modeling to experimental data obtained tionalities on-chip relative to conventional cultures, static MPS,
with a human Body-on-Chips platform that coupled fluid flows and even organoids.[14,18,23,26,27,33,36,44,46,71] Tissues in Organ Chips
between endothelium-lined vascular channels of Liver, Kidney, also can be probed using virtually any type of analytic technique
and Intestine Chips to model first-pass drug ADME behaviors that is used in other in vitro models or animal studies, including
(Figure 7A).[61,70] This multi-Organ Chip platform also incorpo- methods leveraging high resolution microscopy, flow cytometry,
rates an arterio-venous fluid mixing reservoir so that drug con- transcriptomics, proteomics, metabolomics, and histological
centrations within the vascular channels of all chips can be mea- analysis, and as recently demonstrated, automated high content
sured by sampling this shared compartment, much like taking confocal imaging with fluorescent reporters.[72] In addition, be-
venous blood samples in a patient. Quantitative PK predictions cause Organ Chips can be instrumented with in-line electrical,
were demonstrated for two different drugs (cisplatin and nico- chemical, mechanical, and optical sensors (both alone and in
tine) that were administered via two different routes (intravenous combination), they can incorporate real-time readouts and en-
and oral, respectively) (Figure 7B), and predictions of cisplatin able probes of critical cell and tissue functions, including tissue

Adv. Sci. 2020, 2002030 2002030 (10 of 15) © 2020 The Authors. Published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

Figure 7. A first-pass human Body-on-Chips model that predicts human PK parameters. A) Top, photographs of the Gut, Liver, and Kidney Chips that
were fluidically coupled to each other and to an arterio-venous mixer reservoir to create the first-pass PK model in vitro. Bottom, diagrams of the different
2-channel microfluidic chips with arrows indicating the manner in which the chips and reservoir are fluidically linked to each other and to the AV reservoir
(red arrows indicate flow direction). B) Predictions of how nicotine blood concentrations will change over time for three different oral doses (different
colored dashed lines) made by a physiological PK model using experimental data obtained from the human Body-on-Chips platform compared with
previously published blood nicotine levels measured in patients who received orally administered nicotine in the form of nicotine gum (blue), pouched
chewing tobacco known as “snus” (black), or loose snus (green) at three different doses (4, 9, and 13–16 mg). Note that the experimental platform was
able to quantitatively predict these human PK behaviors. C) Graph showing changes in cisplatin concentrations in blood over time predicted by the same
PK model using data from a Body-on-Chips configuration containing linked Liver, Kidney, and Bone Marrow Chips along with an arterio-venous reservoir
for infusion periods (dotted lines) of either 1 h (black) or 3 h (blue). Note that these predictions closely match previously published blood cisplatin
levels measured in patients who received cisplatin injected intravenously over these same time periods (solid lines). Reproduced with permission.[70]
Copyright 2020, Springer Nature.

barrier integrity, neuronal cell electrical activity, oxygen uti- clearly needs greater exploration before animal studies can be re-
lization, pH, molecular transport, and multiple other relevant placed. Quantitative analysis of the effects of large-scale forces
measures of differentiation state and viability.[10,11,32,47,73–79] The and deformations on large organs (e.g., long bones, spinal disks,
ability to control the contents of different parallel flow paths in- whole joints), or of the contributions of macroscale 3D architec-
dividually enables regulation of oxygen and chemical gradients ture to host responses (e.g., infections of the lung apex vs base),
across tissue–tissue interfaces as well, which permits stable co- also cannot be studied using Organ Chip technology, although
culture of living human tissues in direct contact with living com- some simplified models can be developed to study some of the
plex microbiome as in the human intestine,[32] as well as recre- underlying cellular mechanisms (e.g., local effects of compres-
ation of oxygen zonation patterns in organs, such as the liver.[47] sion on bone or cartilage tissue). While mimicry of some hor-
All of these measurements can be carried out under dynamic monal axes has been accomplished using microfluidic Organ
flow conditions as occurs in vivo, and many would be difficult, if Chips, such as the reproduction of the menstrual cycle in vitro,[58]
not impossible, to carry out in animal studies or even in human it is difficult to model temporal variations in many different hu-
trials. man hormones simultaneously as can occur in vivo; moreover,
Organ Chips offer a potentially useful alternative to animal many bloodborne regulatory factors that could be important for
testing, but they cannot replicate many complex functions and biological regulation are still unknown. But because of flow con-
responses that are still better studied in animals. The most ob- trol, it is possible to replicate temporal changes in hormone levels
vious examples relate to studies focused on cognitive functions in Organ Chips for hormones that have been well characterized
and complex organismal behaviors, which remain beyond the ca- in vivo.
pabilities of models that rely on cultured cells. Similarly, while The lack of fat tissues that can absorb many drugs and
brain neurons have been integrated into some microfluidic Or- lipophilic compounds in vivo also could produce abnormal
gan Chips,[34,54] peripheral neurons have not; this is an area that results in experiments studying how drugs distribute in

Adv. Sci. 2020, 2002030 2002030 (11 of 15) © 2020 The Authors. Published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

multi-Organ Chip models. Some materials that are used to con- model,[70] which could potentially accelerate clinical trials. This
struct Organ Chip devices, such as PDMS, can absorb lipophilic inability of a single Organ Chip to model all facets of that organ’s
drugs and hence, this could similarly interfere with drug stud- function, and the need to modify both single and multiple Organ
ies; interestingly, however, by quantifying the level of absorption Chip systems based on the questions being posed, are challenges
into these materials (e.g., using mass spectrometry), this fea- that animal models do not have. Furthermore, regardless of the
ture can be used to mimic fat absorption in human Body-on- device design, all chips must be experimentally validated in terms
Chips modeling.[61,70] Another advantage human Organ Chips of their ability to recapitulate the key physiological or pathophysi-
could potentially provide relative to animal models is the abil- ological properties of the particular organ, disease state, or multi-
ity to replicate human-specific immune and inflammatory re- organ coupling that is the focus of the study, and to do it in a
sponses that are not seen in other species. Circulating immune consistent and robust manner.
cells have been incorporated into multiple human Organ Chip
models[9,14,18,21,25] and complex vaccination responses have been 5. Conclusion
replicated on-chip.[53] But to meet this goal of fully recapitulat-
ing human immune responses in vitro, it will be necessary to This article began with a simple question: does it make sense
incorporate tissue-resident immune cells and obtain parenchy- for reviewers to demand animal validation studies before being
mal, connective tissue, endothelial, and immune cells all from willing to fund grants or share articles with the larger scientific
the same donor. community, and if not, is there a better alternative? As described
Another challenge that the Organ Chip field faces relates to above, the problem mainly lies in the fact that many animal mod-
cell sourcing. While established cell lines generally fail to repli- els are physiologically irrelevant when considering human dis-
cate tissue-specific functions with high fidelity, primary human ease, and thus, demanding use of a poor animal model for the
cells and organoids can display significant donor-to-donor vari- sole sake of “satisfying Reviewers” should be discouraged. Given
ability. The quality of commercially available primary cells (e.g., recent advances in human Organ Chip technologies, I suggest
doubling time, passage number, differentiation potential) also that these new forms of human experimentation in vitro provide
can vary greatly between different suppliers and even between more physiologically and clinically relevant preclinical models for
different lots from the same company. However, these issues can studying both pathophysiology and pharmacological responses
be managed by establishing protocols for “qualifying” each cell than many animal studies, and they should be considered for this
source in terms of the consistency and quality of their growth and purpose in the future. Organoids and other MPS models that of-
differentiation potential before initiating critical experiments. ten can be carried out at a higher throughput than microfluidic
Moreover, inter-donor variability, while frustrating relative to Organ Chips also have a very important role to play, particularly
working with established cell lines or inbred animals, is much in studies focused on cellular and molecular mechanisms of tis-
more clinically relevant as it mimics the diversity of responses sue regulation and drug action. Organ Chips can be used for this
seen in clinical trials and broader human populations. This chal- purpose as well; however, their greatest novelty comes from their
lenge can be overcome by repeating studies with cells from mul- ability to reconstitute dynamic vascular perfusion, tissue–tissue
tiple donors, and exploring whether male and female cell sources interfaces, and organ-specific mechanical cues that are impor-
produce different results can lead to helpful insights as well. In- tant for in vivo-like functions, and to provide an ability to se-
terestingly, however, studies with Lung Airway Chips[18,19] and lectively add complexity to these models in the form of soluble
Bone Marrow Chips[36] suggest that results can be quite repro- factors and different cell types (e.g., epithelial, endothelial, con-
ducible between different donors. nective tissue, immune, nerve, muscle, commensal microbes).
Finally, it is important to note that Organ Chips do not mimic Thus, Organ Chips are a form of synthetic biology that allows
the function of a whole organ; rather, they are effectively living 3D one to build artificial models with defined composition at the cell,
cross-sections of a major functional subunit of an organ, such an tissue, and organ level, and thereby, to identify key cellular and
alveolus, airway, glomerulus, or proximal tubule. So before any molecular contributors to human physiology and pathophysiol-
animal model can be replaced, the Organ Chip must be designed ogy. For example, it would be extremely difficult, if not impossi-
so that it contains the key features of the relevant organ unit that ble, to identify the individual contributions of microbiome, me-
is the key site of this activity, while minimizing system complex- chanical cues, and specific cytokines to pathophysiology at the or-
ity. For example, while a Lung Alveolus Chip containing only alve- gan and multi-organ level using animal models, whereas this has
olar epithelium, endothelium, and immune cells might be used been easily done using Organ Chips. At the very least, manuscript
to replace an animal model of pulmonary edema that is focused and grant reviewers should first consider non-animal alterna-
on preventing pulmonary vascular leakage, it would need to be tives, such as human Organ Chips, and second, whether or not a
modified by adding additional connective tissue cells to replace fully validated animal model is available.
an in vivo pulmonary fibrosis model. Thus, the goal is not to use Importantly, this article is not an argument to stop all animal
Organ Chips to replace animal models in a generic way; rather, it testing, and to carry out all validation studies using human Or-
is to replace one specific type of animal model at a time. Similarly, gan Chips instead. Clearly, we will need to continue some forms
human Body-on-Chips platforms need to be designed to model of animal testing for many years to come in order to cross-validate
the organism-level properties of interest, and recent studies sug- Organ Chip results with the current state-of-the-art, and to pro-
gest that meaningful results can be obtained using this approach gressively convince animal researchers and regulatory scientists
that may be even more useful than those obtained from animal of their value. And it is likely that some animal studies will be very
studies, as demonstrated by the ability to quantitative predict hu- hard to replace (e.g., those focused on cognition and behavior).
man drug PK parameters using a first pass intestine–liver–kidney Rather, my goal here is to initiate a conversation among members

Adv. Sci. 2020, 2002030 2002030 (12 of 15) © 2020 The Authors. Published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

of our community, and to ask scientists who serve as reviewers to [10] G. Wang, M. L. McCain, L. Yang, A. He, F. S. Pasqualini, A. Agarwal,
consider the realities as they stand today. As our collective inter- H. Yuan, D. Jiang, D. Zhang, L. Zangi, J. Geva, A. E. Roberst, Q. Ma,
est is in human health, our goal should not be to validate results J. Dian, J. Chen, D.-Z. Wang, K. Li, J. Wang, R. J. A. Wanders, W. Kulik,
against animals, but rather against humans, because in the end F. M. Vaz, M. A. Laflamme, C. E. Murry, K. R. Chien, R. I. Kelley, G. M.
Church, K. K. Parker, W. T. Pu, Nat. Med. 2014, 20, 616.
we all know that mice are not men. Perhaps it is time for review-
[11] Y. Zhao, N. Rafatian, N. T. Feric, B. J. Cox, R. Aschar-Sobbi, E. Y.
ers to accept this reality.
Wang, P. Aggarwal, B. Zhang, G. Conant, K. Ronaldson-Bouchard, A.
Pahnke, S. Protze, J. H. Lee, L. Davenport Huyer, D. Jekic, A. Wickeler,
Acknowledgements H. E. Naguib, G. M. Keller, G. Vunjak-Novakovic, U. Broekcel, P. H.
Backx, M. Radisic, Cell 2019, 176, 913.
The author thanks all of the research team members, past trainees, [12] U. Marx, T. Akabane, T. B. Andersson, E. Baker, M. Beilmann, S.
collaborators, and other members of the scientific community (includ- Beken, S. Brendler-Schwaab, M. Cirit, R. David, E.-M. Dehne, I.
ing those he might have inadvertently failed to cite) for helping to es- Durieux, L. Ewart, S. C. Fitzpatrick, O. Frey, F. Fuchs, L. G. Griffith,
tablish and build the Organ Chip field. Work from the laboratory re- G. A. Hamilton, T. Hartung, J. Hoeng, H. Hogberg, D. J. Hughes, D.
viewed here was funded by DARPA (W911NF1920023, W911NF1920027, E. Ingber, A. Iskander, T. Kanamori, H. Kojima, J. Kuehnl, M. Leist, B.
W911NF-12-2-0036, HR0011-20-2-0040), FDA (75F40119C10098), NIH
Li, P. Loskill, D. L. Mendrick, et al., Altex 2020, 37, 364.
(UH3HL141797), Cancer Research United Kingdom (C25640/A29057),
[13] S. N. Bhatia, D. E. Ingber, Nat. Biotechnol. 2014, 32, 760.
and Gates Foundation (OPP1173198, INV-002164).
[14] D. Huh, B. D. Matthews, A. Mammoto, M. Montoya-Zavala, H. Y.
Hsin, D. E. Ingber, Science 2010, 328, 1662.
Conflict of Interest [15] D. Huh, D. C. Leslie, B. D. Matthews, J. P. Fraser, S. Jurek, G. A. Hamil-
ton, K. S. Thorneloe, M. A. McAlexander, D. E. Ingber, Science Trans.
The author has the following potential conflicts: Emulate Inc., equity, con- Med. 2012, 4, 159ra147.
sulting, chair of SAB; BOA Biomedical Inc., equity, consulting, chair of SAB, [16] A. Jain, R. Barrile, A. van der Meer, A. Mammoto, T. Mammoto, K.
board member; Free Flow Medical Device, equity; SynDevRx, equity; Con- DeCeunyck, O. Aisku, M. A. Otieno, C. S. Louden, G. A. Hamilton, R.
sortia Rx, equity, board member; Roche, consulting; Astrazeneca, spon- Flaumenhaft, D. E. Ingber, Clin. Pharmacol. Therap. 2018, 103, 332.
sored research; Fulcrum Therapeutics, sponsored research; Kraft Heinz,
[17] J. Li, A. Wen, R. Potla, E. Benshirim, A. Seebarran, M. A. Benz, O.
sponsored research; and inventor of multiple patent applications.
Henry, R. Prantil-Baun, S. Gilpin, O. Levy, D. E. Ingber, APL Bioeng.
2019, 3, 046103.
Keywords [18] K. H. Benam, R. Villenave, C. Lucchesi, A. Varone, C. Hubeau, H.-H.
Lee, S. E. Alves, M. Salmon, T. C. Ferrante, J. Weaver, G. A. Hamilton,
microfluidics, microphysiological systems, organoids, organ-on-a-chip, A. Bahinski, D. E. Ingber, Nat. Methods 2016,13, 151.
preclinical studies [19] K. Benam, R. Novak, J. Nawroth, M. Hirano-Kobayashi, T. Ferrante,
Y. Choe, R. Prantil-Baun, A. Bahinski, K. K. Parker, D. E. Ingber, Cell
Received: May 29, 2020 Syst. 2016, 3, 456.
Revised: July 22, 2020 [20] L. Si, R. Prantil-Baun, K. Benam, H. Bai, M. Rodas, M. Burt, D. E.
Published online: Ingber, bioRxiv:685552, 2019, https://doi.org/10.1101/685552.
[21] L. Si, H. Bai, M. Rodas, W. Cao, C. Y. Oh, A. Jiang, R. Moller, D.
Hoagland, K. Oishi, S. Horiuchi, S. Uhl, D. Blanco-Melo, R. A. Al-
brecht, W.-C. Liu, T. Jordan, B. E. Nilsson-Payant, J. Logue, R. Haupt,
[1] A. Akthar, Camb. Q. Healthc. Ethics 2015, 24, 407. M. McGrath, S. Weston, A. Nurani, S. M. Kim, D. Y. Zhu, K. H. Benam,
[2] L. Meigs, L. Smirnova, C. Rovida, M. Leist, T. Hartung, Altex 2018, 35, G. Goyal, S. E. Gilpin, R. Prantil-Baun, R. K. Powers, K. Carlson, M.
275. Frieman, et al., bioRxiv:04.13.039917, 2020, https://doi.org/10.1101/
[3] P. Pound, M. Ritskes-Hoitinga, J.Transl. Med. 2018, 16, 304. 2020.04.13.039917.
[4] J. Seok, H. S. Warren, A. G. Cuenca, M. N. Mindrinos, H. V. Baker, W. [22] A. L. Gard, R. Maloney, B. P. Cain, C. R. Miller, R. J. Luu, J. R. Coppeta,
Xu, D. R. Richards, G. P. McDonald-Smith, H. Gao, L. Hennessy, C. C. P. Liu, J. P. Wang, H. Azizgolshani, R. F. Fezzie, J. L. Balestrini, B. C.
Finnerty, C. M. López, S. Honari, E. E. Moore, J. P. Minei, J. Cuschieri, Isenberg, R. W. Finberg, J. T. Borenstein, bioRxiv:05.23.112797, 2020,
P. E. Bankey, J. L. Johnson, J. Sperry, A. B. Nathens, T. R. Billiar, M. https://doi.org/10.1101/2020.05.23.112797.
A. West, M. G. Jeschke, M. B. Klein, R. L. Gamelli, N. S. Gibran, B. [23] H. J. Kim, D. Huh, G. A. Hamilton, D. E. Ingber, Lab Chip 2012, 12,
H. Brownstein, C. Miller-Graziano, S. E. Calvano, P. H. Mason, et al., 2165.
Proc. Natl. Acad. Sci. U. S. A. 2013, 110, 3507. [24] H. J. Kim, D. E. Ingber, Integr. Biol. 2013, 5, 1130.
[5] R. Franco, A. Cedazo-Minguez, Front. Pharmacol 2014, 5, 146. [25] H. J. Kim, H. Li, J. J. Collins, D. E. Ingber, Proc. Natl. Acad. Sci. U. S.
[6] R. S. Herati, E. J. Wherry, Cold Spring Harb. Perspect. Biol. 2018, 10, A. 2016, 113, E7.
a031583. [26] M. Kasendra, A. Tovaglieri, A. Sontheimer-Phelps, S. Jalili-
[7] H. Clevers, Cell 2016, 165, 1586. Firoozinezhad, A. Bein, A. Chalkiadaki, W. Scholl, C. Zhang, H.
[8] K. A. Homan, N. Gupta, K. T. Kroll, D. Kolesky, M. Skylar-Scott, T. Rickner, C. A. Richmond, H. Li, D. T. Breault, D. E. Ingber, Sci. Rep.
Miyoshi, D. Mau, M. Todd Valerius, T. Ferrante, J. V. Bonventre, J. A. 2018, 8, 2871.
Lewis, R. Morizane, Nat. Methods 2019, 16, 255. [27] A. Sontheimer-Phelps, D. B. Chou, A. Tovaglieri, T. C. Ferrante,
[9] J. T. Neal, X. Li, J. Zhu, V. Giangarra, C. L. Grzeskowiak, J. Ju, I. H. Liu, T. Duckworth, C. Fadel, V. Frismantas, S. Jalili-Firoozinezhad, M.
S. Chiou, A. A. Salahudeen, A. R. Smith, B. C. Deutsch, L. Liao, A. J. Ze- Kasendra, E. Stas, J. C. Weaver, C. A. Richmond, O. Levy, R. Prantil-
mek, F. Zhao, K. Karlsson, L. M. Schultz, T. J. Metzner, L. D. Nadauld, Baun, D. T. Breault, D. E. Ingber, Cell. Mol. Gastroenterol. Hepatol.
Y. Tseng, S. Alkahairy, C. Oh, P. Keskula, D. Mendoza-Villanueva, F. 2020, 9, 507.
M. De La Vega, P. L. Kunz, J. C. Liao, J. T. Leppert, J. B. Sunwoo, C. [28] M. Kasendra, R. Luc, J. Yin, D. V. Manatakis, G. Kulkarni, C. Lucch-
Sabatti, J. S. Boehm, W. C. Hahn, G. X. Y. Zheng, M. M. Davis, C. J. esi, J. Sliz, A. Apostolou, L. Sunawar, J. Obrigewitch, K.-J. Kang, G. A.
Kuo, Cell 2018, 175, 1972. Hamilton, M. Donowitz, K. Karalis, Elife 2020, 9, e50135.

Adv. Sci. 2020, 2002030 2002030 (13 of 15) © 2020 The Authors. Published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

[29] R. Villenave, S. Q. Wales, T. Hamkins-Indik, E. Papafragkou, J. C. S. Haney, D. E. Ingber, G. A. Hamilton, Sci. Trans. Med. 2019, 11,
Weaver, T. C. Ferrante, A. Bahinski, C. Elkins, M. Kulka, D. E. Ingber, eaax5516.
PLoS One 2017, 12, e0169412. [47] X. Li, S. M. George, L. Vernetti, A. H. Gough, D. L. Taylor, Lab Chip
[30] S. Jalili-Firoozinezhad, R. Prantil-Baun, A. Jiang, T. Mammoto, J. C. 2018, 18, 2614.
Weaver, T. C. Ferrante, H. J. Kim, J. M. S. Cabral, O. Levy, D. E. Ingber, [48] N. Tsamandouras, T. Kostrzewski, C. L. Stokes, L. G. Griffith, D. J.
Cell Death Dis. 2018, 9, 223. Hughes, M. Cirit, J. Pharmacol. Exp. Ther. 2017, 360, 95.
[31] A. Grassart, V. Malardé, S. Gobaa, A. Sartori-Rupp, J. Kerns, K. Karalis, [49] T. J. Long, P. A. Cosgrove, R. T. Dunn, Drug Metab. Dispos. 2016, 44,
B. Marteyn, P. Sansonetti, N. Sauvonnet, Cell Host Microbe 2019, 26, 1940.
435. [50] K. Achberger, C. Probst, J. Haderspeck, S. Bolz, J. Rogal, J. Chuchuy,
[32] S. Jalili-Firoozinezhad, F. S. Gazzaniga, E. L. Calamari, D. M. Cama- M. Nikolova, V. Cora, L. Antkowiak, W. Haq, N. Shen, K. Schenke-
cho, C. W. Fadel, B. Nestor, M. J. Cronce, A. Tovaglieri, O. Levy, K. E. Layland, M. Ueffing, S. Liebau, P. Loskill, Elife 2019, 8, e46188.
Gregory, D. T. Breault, J. M. S. Cabral, D. L. Kasper, R. Novak, D. E. [51] K. J. Jang, A. P. Mehr, G. A. Hamilton, L. A. McPartlin, K.-Y. Suh, D. E.
Ingber, Nat. Biomed. Eng. 2019, 3, 520. Ingber, Integr. Biol. 2013, 5, 1119.
[33] T.-E. Park, N. Mustafaoglu, A. Herland, R. Hasselkus, R. Mannix, E. [52] R. Barrile, A. D. van der Meer, H. Park, J. Fraser, F. Teng, D. Simic, D.
A. FitzGerald, R. Prantil Baun, A. Watters, O. Henry, M. Benz, H. Conegliano, J. Nguyen, T. Almy, D. Weinstock, Y. Wang, C. Louden, K.
Sanchez, H. J. McCrea, L. C. Coumnerova, H. K. Wilson, S. P. Pale- Karalis, D. E. Ingber, G. A. Hamilton, M. Otieno, Clin. Pharm. Therap.
cek, E. Shusta, D. E. Ingber, Nat. Commun. 2019, 10, 2621. 2018, 104, 1240.
[34] G. D. Vatine, R. Barrile, M. J. Workman, S. Sances, B. Barriga, M. Rah- [53] G. Goyal, B. Bausk, P. Prabhala, L. Xie, D. Curran, J. Long, L. Cohen, O.
nama, S. Barthakur, M. Kasendra, C. Lucchesi, J. Kerns, N. Wen, W. Levy, R. Prantil-Baun, D. R. Walt, D. E. Ingber, bioRxiv:806505, 2019,
R. Spivia, Z. Chen, J. Van Eyk, C. Svendsen, Cell Stem Cell 2019, 24, https://doi.org/10.1101/806505.
995. [54] B. M. Maoz, A. Herland, E. A. Fitzgerald, T. Grevesse, C. Vidoudez, A.
[35] G. Morad, C. Carman, E. Hagedorn, J. Perlin, L. Zon, N. Mustafaoglu, R. Pacheco, S. P. Sheehy, T.-E. Park, S. Dauth, R. Mannix, N. Budnik,
T.-E. Park, D. E. Ingber, C. Daisy, M. Moses, ACS Nano 2019, 13, K. Shores, A. Cho, J. C. Nawroth, D. Segre, B. Budnik, D. E. Ingber, K.
13853. K. Parker, Nat. Biotechnol. 2018, 36, 865.
[36] D. B. Chou, V. Frismantas, Y. Milton, R. David, P. Pop-Damkov, D. Fer- [55] M. Trapecar, C. Communal, J. Velazquez, C. A. Mauss, Y.-J. Huang, K.
guson, A. MacDonald, O. Vargel-Bolukbasi, C. E. Joyce, L. S. Moreira- Schneider, C. W. Wright, V. Butty, G. Eng, O. Yilmaz, D. Trumper, L.
Teixeira, A. Rech, A. Jiang, E. Calamari, S. Jalili Firoozinezhad, B. A. G. Griffith, Cell Syst. 2020, 10, 223.
Furlong, L. R. O’Sullivan, C. F. Ng, Y. Choe, S. Marquez, K. C. Myers, [56] C. W. McAleer, C. J. Long, D. Elbrecht, T. Sasserath, L. R. Bridges,
O. K. Weinberg, R. P. Hasserjian, R. Novak, O. Levy, R. Prantil-Baun, J. W. Rumsey, C. Martin, M. Schnepper, Y. Wang, F. Schuler, A. B.
C. D. Novina, A. Shimamura, L. Ewart, D. E. Ingber, Nat. Biomed. Eng. Roth, C. Funk, M. L. Schuler, J. J. Hickman, Sci. Transl. Med. 2019, 11,
2020, 4, 394. eaav1386.
[37] B. Hassell, G. Goyal, E. Lee, O. Levy, C. S. Chen, D. E. Ingber, Cell [57] L. Vernetti, A. Gough, N. Baetz, S. Blutt, J. R. Broughman, J. A. Brown,
Rep. 2017, 21, 508. J. Foulke-Abel, N. Hasan, J. In, E. Kelly, O. Kovbasnjuk, J. Repper, N.
[38] A. Marturano-Kruik, M. M. Nava, K. Yeager, A. Chramiec, L. Hao, S. Senutovitch, J. Stabb, C. Yeung, N. C. Zachos, M. Donowitz, M. Estes,
Robinson, E. Guo, M. T. Raimondi, G. Vunjak-Novakovic, Proc. Natl. J. Himmelfarb, G. Truskey, J. P. Wikswo, D. L. Taylor, Sci. Rep. 2017,
Acad. Sci. U. S. A. 2018, 115, 1256. 7, 42296.
[39] S. E. Wheeler, J. T. Borenstein, A. M. Clark, M. R. Ebrahimkhani, I. J. [58] S. Bauer, C. Wennberg Huldt, K. P. Kanebratt, I. Dureiex, D. Gunne,
Fox, L. Griffith, W. Inman, D. Lauffenburger, T. Nguyen, V. C. Pillai, R. S. Andersson, L. Ewart, W. G. Haynes, I. Maschmeyer, A. Winter, C.
Prantil-Baun, D. B. Stolz, D. Taylor, T. Ulrich, R. Venkataramanan, A. Ammala, U. Marx, T. B. Andersson, Sci. Rep. 2017, 7, 14620 [published
Wells, C. Young, Stem Cell Res. Ther. 2013, 4, S11. correction appears in Sci. Rep. 2018, 8, 1672].
[40] A. Pavesi, A. T. Tan, S. Koh, A. Chia, M. Colombo, E. Antonecchia, C. [59] S. Xiao, J. R. Coppeta, H. B. Rogers, B. C. Isenberg, J. Zhu, S. A.
Miccolis, E. Ceccarello, G. Adriani, M. T. Raimondi, R. D. Kamm, A. Olalekan, K. E. McKinnon, D. Dokic, A. S. Rashedi, D. J. Haisen-
Bertoletti, J. CIin. Insight 2017, 2, 89762. leder, S. S. Malpani, C. A. Arnold-Murray, K. Chen, M. Jiang, L. Bai,
[41] C. Maass, N. B. Sorensen, J. Himmelfarb, E. J. Kelly, C. L. Stokes, M. C. T. Nguyen, J. Zhang, M. M. Laronda, T. J. Hope, K. P. Maniar,
Cirit, CPT: Pharmacometrics Syst. Pharmacol. 2019, 8, 316. M. E. Pavone, M. J. Avra, E. C. Sefton, S. Getsios, J. E. Burdette,
[42] E. J. Weber, K. A. Lidberg, L. Wang, T. K. Bammler, J. W. MacDonald, J. J. Kim, J. T. Borenstein, T. K. Woodruff, Nat. Commun. 2017, 8,
M. J. Li, M. Redhair, W. M. Atkins, C. Tran, K. M. Hines, J. Herron, L. 14584.
Xu, M. B. Monteiro, S. Ramm, V. Vaidya, M. Vaara, T. Vaara, J. Him- [60] C. D. Edington, W. L. K. Chen, E. Geishecker, T. Kassis, L. R. Soenksen,
melfarb, E. J. Kelly, JCI Insight 2018, 3, e123673. B. M. Bhushan, D. Freake, J. Kirschner, C. Maass, N. Tsamandouras,
[43] J. Wang, C. Wang, N. Xu, Z. F. Liu, D. W. Pang, Z. L. Zhang, Biomate- J. Valdez, C. D. Cook, T. Parent, S. Snyder, J. Yu, E. Suter, M. Schockley,
rials 2019, 219, 119367. J. Velazquez, J. J. Velazquez, L. Stockdale, J. P. Papps, I. Lee, N. Vann,
[44] S. Musah, T. Mammoto, A. Mammoto, T. C. Ferrante, M. Hirano- M. Gamboa, M. E. LaBarge, Z. Zhong, X. Wang, L. A. Boyer, D. A.
Kobayashi, K. Roberts, S. Chung, R. Novak, M. Ingram, T. Fatanat- Lauffenburger, R. L. Carrier, C. Communal, S. R. Tannenbaum, C. L.
Didar, J. C. Weaver, G. M. Church, D. E. Ingber, Nat. Biomed. Eng. Stokes, D. J. Hughes, G. Rohatgi, D. L. Trumper, M. Cirti, L. G. Griffith,
2017, 1, 69. Sci. Rep. 2018, 8, 4530.
[45] A. Tovaglieri, A. Sontheimer-Phelps, A. Geirnaert, R. Prantil-Baun, [61] R. Novak, M. Ingram, S. Marquez, D. Das, A. Delahanty, A. Herland,
D. M. Camacho, D. B. Chou, S. Jalili-Firoozinezhad, T. Wouters, M. B. M. Maoz, S. S. F. Jeanty, M. R. Somayaji, M. Burt, E. Calamari,
Kasendra, M. Super, M. Cartwright, C. A. Richmond, D. T. Breault, C. A. Chalkiadaki, A. Cho, Y. Choe, D. B. Chou, M. Cronce, S. Dauth, T.
Lacroix, D. E. Ingber, Microbiome 2019, 7, 43. Divic, J. Fernandez-Alcon, T. Ferrante, J. Ferrier, E. A. FitzGerald, R.
[46] K.-J. Jang, M. A. Otieno, J. Ronxhi, K. Kodella, J. Rubins, D. Petropolis, Fleming, S. Jalili-Firoozinezhad, T. Grevesse, J. A. Goss, T. Hamkins-
G. Kulkarni, D. Simic, H.-K. Lim, W. Lam, M. Singer, E. Barale, B. Indik, O. Henry, C. Hinojosa, T. Huffstater, et al., Nat. Biomed. Eng.
Singh, M. Sonee, A. J. Streeter, C. Manthey, L. Ewart, B. Jones, A. 2020, 4, 407.
Srivastava, H. Park, J. Nawroth, D. Conegliano, J. Sliz, A. Herland, [62] H. E. Abaci, M. Shuler, Integr. Biol. 2015, 7, 383.

Adv. Sci. 2020, 2002030 2002030 (14 of 15) © 2020 The Authors. Published by Wiley-VCH GmbH
www.advancedsciencenews.com www.advancedscience.com

[63] M. B. Esch, H. Ueno, D. R. Applegate, M. L. Shuler, Lab Chip 2016, Sareen, G. A. Hamilton, C. N. Svendsen, Stem Cell Rep. 2018, 10,
16, 2719. 1222.
[64] J. R. Coppeta, M. J. Mescher, B. C. Isenberg, A. J. Spencer, E. S. Kim, A. [72] S. Peel, A. M. Corrigan, B. Ehrhardt, K.-J. Jang, P. Caetano-Pino, M.
R. Lever, T. J. Mulhern, R. Prantil-Baun, J. C. Comolli, J. T. Borenstein, Boeckeler, J. E. Rubins, K. Kodella, D. B. Petropolis, J. Ronxhi, G.
Lab Chip 2016, 17, 134. Kulkarni, A. J. Foster, D. Williams, G. A. Hamilton, L. Ewart, Lab Chip
[65] I. Wagner, E.-M. Materne, S. Brincker, U. Süssbier, C. Frädrich, M. 2019, 19, 410.
Busek, F. Sonntag, D. A. Sakharov, E. V. Trushkin, A. G. Tonevitsky, R. [73] S. B. Kim, K.-I. Koo, H. Bae, M. R. Dokmeci, G. A. Hamilton, A. Bahin-
Lauster, U. Marx, Lab Chip 2013, 13, 3538. ski, S. M. Kim, D. E. Ingber, A. Khademhosseini, Lab Chip 2012, 12,
[66] C. L. Stokes, M. Cirit, D. Lauffenburger, CPT: Pharmacometrics Syst. 3976.
Pharmacol. 2015, 4, 559. [74] M. Odijk, A. D. van der Meer, D. Levner, H. J. Kim, M. W. van der
[67] D. Bovard, A. Sandoz, K. Luettich, S. Frentzel, A. Iskandar, D. Helm, L. I. Segerink, J. P. Frimat, G. A. Hamilton, D. E. Ingber, A. van
Marescotti, K. Trivedi, E. Guedj, Q. Dutertre, M. C. Peitsch, J. Hoeng, den Berg, Lab Chip 2015, 15, 745.
Lab Chip 2018, 18, 3814. [75] S. A. Mousavi, F. De Ferrari, Y. S. Zhang, M. Nabavinia, N. B. Mo-
[68] C. Oleaga, B. Bernabini, A. S. Smith, B. Srinivasan, M. Jackson, W. hammad, J. Ryan, A. Pourmand, E. Laukaitis, R. B. Sadeghian, A.
McLamb, V. Platt, R. Bridges, Y. Cai, N. Santhanam, B. Berry, S. Najjar, Nadhman, S. R. Shin, A. S. Nezhad, A. Khademhosseini, M. R. Dok-
N. Akanda, X. Guo, C. Martin, G. Ekman, M. B. Esch, J. Langer, G. meci, Biomicrofluidics 2016, 10, 044111.
Ouedraogo, J. Cotovio, L. Breton, M. L. Shuler, J. J. Hickman, Sci. [76] O. Y. F. Henry, R. Villenave, R. D. E. Ingber, Lab Chip 2017, 17, 2264.
Rep. 2016, 6, 20030. [77] B. M. Moaz, A. Herland, O. Y. F. Henry, W. Leineweber, M. Yadid, R.
[69] I. Maschmeyer, A. K. Lorenz, K. Schimek, T. Hasenberg, A. P. Ramme, Mannix, V. J. Kujala, E. A. Fitzgerald, K. K. Parker, D. E. Ingber, Lab
J. Hübner, M. Lindner, C. Drewell, S. Bauer, A. Thomas, N. S. Sambo, Chip 2017, 17, 2294.
F. Sonntag, R. Lauster, U. Marx, Lab Chip 2015, 15, 2688. [78] D. Sheyn, D. Cohn-Yakubovich, S. Ben-David, S. De Mel, V. Chan,
[70] A. Herland, B. M. Maoz, D. Debarun, M. R. Somayaji, R. Prantil- C. Hinojosa, N. Wen, G. A. Hamilton, D. Gazit, Z. Gazit, Microfluid.
Baun, R. Novak, M. Cronce, T. Huffstater, S. S. F. Jeanty, M. In- Nanofluid. 2019, 23, 99.
gram, A. Chalkiadaki, D. B. Chou, S. Marquez, A. Delahanty, S. Jalili- [79] Y. S. Zhang, J. Aleman, S. R. Shin, T. Kilic, D. Kim, S. A. M. Shaegh,
Firoozinezhad, Y. Milton, A. Sontheimer-Phelps, B. Swenor, O. Levy, S. Massa, R. Riahi, S. Chae, N. Hu, H. Avci, W. Zhang, A. Silvestri,
K. K. Parker, A. Przekwas, D. E. Ingber, Nat. Biomed. Eng. 2020, 4, A. S. Nezhad, A. Manbohi, F. D. Ferrari, A. Polini, G. Calzone, N.
421. Shaikh, P. Alerasool, E. Budina, J. Kang, N. Bhise, J. Ribas, A. Pour-
[71] S. Sances, R. Ho, G. Vatine, D. West, A. Laperle, A. Meyer, M. mand, A. Skardal, T. Shupe, C. E. Bishop, M. R. Dokmeci, A. Atala, A.
Godoy, P. S. Kay, B. Mandefro, S. Hatata, C. Hinojosa, N. Wen, D. Khademhosseini, Proc. Natl. Acad. Sci. U. S. A. 2017, 114, E2293.

Donald Ingber is the founding director of the Wyss Institute for Biologically Inspired Engineering at
Harvard University, Judah Folkman Professor of Vascular Biology at Harvard Medical School and
Boston Children’s Hospital, and professor of bioengineering at the Harvard John A. Paulson School
of Engineering and Applied Sciences. He received his B.A., M.A., M.Phil., M.D., and Ph.D. from Yale
University. He has made significant contributions to many disciplines and helped to pioneer the devel-
opment of microfluidic Organ Chips.

Adv. Sci. 2020, 2002030 2002030 (15 of 15) © 2020 The Authors. Published by Wiley-VCH GmbH

You might also like