CRISPR Ethics: Moral
Considerations for Applications of a
Powerful Tool
Carolyn Brokowski
1
and Mazhar Adli
2
1 - Department of Emergency Medicine, Yale School of Medicine, 464 Congress Avenue, New Haven, CT 06519-1362, USA
2 - Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, 1340 Jefferson Park Avenue,
Charlottesville, VA 22908, USA
Correspondence to Mazhar Adli: [email protected]
https://doi.org/10.1016/j.jmb.2018.05.044
Edited by Prashant Mali
Abstract
With the emergence of CRISPR techno logy, targeted editing of a wide variety of genomes is no longer an
abstract hypothetical, but occurs reg ularly. As application areas of CRISPR are exceeding beyond research
and biomedical therapies, new and existing ethical concerns abound throughout the global community about
the appropriate scope of the systems' use. Here we review fundamental ethical issues including the following:
1) the extent to which CRISPR use should be permitted; 2) access to CRISPR applications; 3) wheth er a
regulatory framework(s) for clinical research involving human subjects might accommodate all types of human
genome editing, including editing of the germline; and 4) whether international regulations governing
inappropriate CRISPR utilization should be crafted and publicized. We conclude that moral decision making
should evolve as the science of genomic engineering advances and hold that it would be reasonable for
national and supranational legislatures to consider evidence-based regulation of certain CRISPR applications
for the betterment of human health and progress.
© 2018 Elsevier Ltd. All rights reserved.
Introduction
The CRISPR (Clustered Regularly Interspaced
Short Palindromic Repeats)-Cas9 (CRISPR-associ-
ated protein 9) system (CRISPR or the system)is
the most versatile genomic engineering tool created
in the history of molecular biology to date. This
system's ability to edit divers e genome types with
unprecedented ease has caused considerable ex-
citement and alarm throughout the international
biomedical community.
CRISPR appears to offer considerable promise in
a wide variety of disease contexts. For example,
around the world at least 15 clinical trialsfocused
on multiple myeloma; esophageal, lung, prostate,
and bladder cancer; solid tumors; melanoma;
leukemia; human papilloma virus; HIV-1; gastroin-
testinal infection; β-thalassemia; sickle-cell anemia;
and other diseasesinvolving CRISPR appli cations
have been developed [13]. Moreover, as of May,
2018, in China at least 86 individuals have had their
genes altered as part of clinical trials [4].
While significant public support exists for thera-
peutic applications [5], ethical (moral) and safety
concerns about certain areas of CRISPR applica-
tions, such as ger mline editing, are apparent around
the world [6]. Notably, such discussions commenced
during the Napa Valley meeting of 2015 when a
leading group of CRISPRCas9 developers, scien-
tists, and ethicist s met to examine the biomedical,
legal, and ethical aspects of CRISPR systems [7].
From this meeting, more extensive deliberations
were solicited, and the United States (US) National
Academies of Sciences, Engineering, and Medicine
(NASEM or The Committee) invited the Chinese
Academy of Sciences and the United Kingdom's
(UK) Royal Society to participate in the International
Summit on Human Gene Editing [8]. The goal of this
meeting was to examine when, where, and how the
technology might be applied in humans. This
discussion continued in February of 2017 when a
multidisciplinary committee of the NASEM published
a comprehensive report examining numerous as-
pects of human genome editing [9].
KDC YJMBI-65762; No. of pages: 14; 4C:
0022-2836/© 2018 Elsevier Ltd. All rights reserved. J Mol Biol (2018) xx, xxxxxx
Review
Please cite this article as: C. Brokowski, M. Adli, CRISPR Ethics: Moral Considerations for Applications of a Powerful Tool, J. Mol. Biol.
(2018), https://doi.org/10.1016/j.jmb.2018.05.044
To date, the NASEM report provides perh aps the
most influential, extensive analysis examining wide-
ranging concerns about human genome editing [10].
Importantly, the Committee favored somatic genome
editing, but did not permit genomic modification for
any kind of enhancement [9, 11]. Also, though
impermissible at present, the Committee concluded
cautiously that human heritable genome editing, the
modification of the germline with the goal of creating
a new person who could potentially transfer the
genomic edit to future generations, would be
permissible under certain conditions: In light of the
technical and social concerns involved heritable
genome-editing research trials might be permitted,
but only following much more research aimed at
meeting existing risk/benefit standards for authoriz-
ing clinical trials and even then, only for compelling
reasons and under strict oversight. [9] Although by
law, US federal funding cannot be used to support
research involving human em bryos [1214], the
NASEM report suggests that when technical and
safety risks are better understood then clinical trials
involving germline editing might begin [9].
In this review, we aim to summarize fundamental
ethical concerns about CRISPR use in general, but
the list is not exhaustive. First, we briefly review
CRISPR systems and their applications in editing
genomes and epigenomes. Second, we describe
how complexities of CRISPR science affect those of
CRISPR ethics and vice versa. Third, we assess
several key ethical considerations. Notably, while
some of these concerns are specific to CRISPR
technology, many, such as research on human
embryos, have been debated long before the
CRISPR revolution [15]. Moreover, since CRISPR
is still a maturing technology, novel applications in
the future may raise new ethical quandaries meriting
further attention and dissection. Fourth, it is impor-
tant to point out that, though morality and law often
overlap, significant differences exist. Although law
may affect ethics and vice versa, we focus mostly on
ethics. Finally, while discussing these issues, we
assume no position on any topic; our account is
merely descriptive. Therefore, we mak e no attempt
to settle any of the controversies presented herein.
CRISPR systems and their uses
Different CRISPR systems in genome editing
CRISPR is a natural bacterial defense system
against invading viruses and nucleic acids. Over
billions of years, multiple CRISPR-type immune
systems have evolved. Naturally occurring CRISPR
systems are typically classified by their repertoires of
CRISPR-associated (cas) genes, which are often
found adjacent to the CRISP R arrays [16, 17].
Although the characterization is yet to be finalized,
two major classes of CRISPRCas adaptive immune
systems have been identified in prokaryotes [1820].
This division is based on the organization of effector
modules. Class 1 CRISPRCas systems employ
multi-protein effector complexes and encompass
three types (I, III, and IV). By contrast, Class 2
systems utilize single protein effectors and encom-
pass three other types (II, V, and VI). Although various
natural CRISPRCas systems have been repurposed
for genome editing, due to its robust gene-editing
efficiency and broader genome-targeting scope owing
to its simple NGG PAM sequence requirement, the
Cas9 from Streptococcus pyogenes (spCas9) is
currently the most commonly used CRISPRCas9
protein. It is worth noting that multiple efforts are
underway to discover novel Cas9 variants or re-
engineer the existing Cas9 proteins, which will have
less dependence on the stringent PAM-sequence
requirement [21, 22].
CRISPR goes beyond genome editing
The DNA-editing capacity of CRISPRCas9 is due
to the ability of the WT Cas9 protein to cause double-
stranded breaks at the target site that is determined
by the custom-designed short guiding RNA [23]. The
repair of DNA breaks frequen tly results in indels, due
to the non-homologous end joining (NHEJ) repair
mechanism. However, when a complementary tem-
plate is available, homology-directed repair (HDR)
machinery can use it and thereby achieve more
precise gene editing. Notably, a single-point mutation
in either of the two catalytic domains of Cas9 results
in a nickase Cas9 (nCas9), whereas mutations in
both domains (D10A and H840A for spCas9)
diminish Cas9's catalytic activity, resulting in dead
Cas9 (dCas9) [24]. Interestingly, the application
areas of modified Cas9 proteins are exceeding that
of WT Cas9 [25]. Such uses are largely possible
because the nCas9 or dCas9 can still be guided to
the target sequence [26]. Researchers employed
these Cas9 variants for unique purposes. For
example, tandem targeting of nCas9 has been
utilized to improve targeting specificity [27, 28].
More recently, this enzyme has been used as the
base platform for second generation genome-editing
tools called base editors [29, 30] Base-editing
technology employs cytidine or adenine deaminase
enzymes to achieve the programmable conversion of
one base into another (C to T or A to G). Most
importantly, the targeted base transition happens
without DNA double-stranded breaks [29, 31].We
recently utilized this technology to edit the universal
genetic code and introduced a stop codon in the
genes [32].
In addition to nCas9, researchers uti lized the
guidable capacity of dCas9 as a platform to recruit
various effector proteins to a specific locus in living
cells. Generally, these activities ca n be classified as
2 Review: CRISPR Ethics
Please cite this article as: C. Brokowski, M. Adli, CRISPR Ethics: Moral Considerations for Applications of a Powerful Tool, J. Mol. Biol.
(2018), https://doi.org/10.1016/j.jmb.2018.05.044
epigenetic editing (to alter locus-specific epigenetic
information), gene regulation (to turn the activities of
single or multiple genes on or off), chromatin imaging
(to label and monitor chromatin dynamics in living
cells), and manipulation of chromatin topology (to
alter 3D chromatin structure in the nuclear space)
[33].
CRISPR research is progressing at a rapid pace.
Recently, scientists have a lso uncovered new
CRISPRCas systems (Cas13) that can target RNA
instead of DNA [34, 35]. By enabling targeted RNA
recognition and editing, these newer RNA-targeting
CRISPR tools have their unique applications ranging
from biomedical and biotechnological to the detection
of nucleic acids [36, 37]. Although many ethical
concerns are related to the catalytic activities of WT
Cas9partly because it permanently alters the
genetic informationsome of these activities of
catalytically inactive dCas9, nickase-Cas9-based
platforms, such as base editors and recently discov-
ered RNA-targeting Cas proteins, may raise compa-
rable moral issues depending on the duration of the
exerted effect and the purpose of the experiments.
Detailed discussion of such issues, however, is
beyond the scope of this review (Table 1).
CRISPR ethics and science:
Uncomfortable bedfellows
Moral decisions, especially in biomedicine, are
empirically informed and involve assessing potential
risk-benefit ratiosattempting to maximize the latter
while minimizing the former. To navigate ethical
decision making, it is critical to consider the range of
possible outcomes, the probabilities of each instantiat-
ing, and the possible justifications driving the results of
any one. The ethical concerns about CRISPR genome
engineering technology are largely due to at least three
important reasons.
First, there are concerns about the power and
technical limitations of CRISPR technology. These
include the possibilities of limited on-target editin g
efficiency [38, 39], incomplete editing (mosaicism)
[40, 41], and inaccurate on- or off-target editing [42,
43]. These limitations have been reported in CRISPR
experiments involving animals and human cell lines.
However, the technology is evolving at an unprece-
dented pace. As m ore efficient and sensitiv e
CRISPR tools are developed, many of these con-
cerns may become obsolete. Yet for the sake of this
review, we mention these limitations as one of the
principal worries about widespread CRISPR utiliza-
tion. Second, it is unclear whether modified organ-
isms will be affected indefinitely and whether the
edited genes will be transferred to future generations,
potentially affecting them in unexpected ways.
Combined with technical limitations and the com-
plexities of biological systems, making precise
predictions about the future of an edited organism
and gauging potential risks and benefits might be
difficult, if not impossible. Thus, uncertainty resulting
from these factors hinders accurate risk-benefit
analysis, complicating moral decision making.
Finally, the skeptical view is that even if the genome
is edited as expected and the desired functional output
is achieved at the given time, the complex relationship
between genetic information and biolo gical phenotyp es
is not fully understood. Therefore, the biological
consequence of editing a gene in germline and/or
somatic cells may be unclear and unpredictable
depending on the context. Many biological traits are
determined by the complex regulatory actions of
numerous genes. Hence, is it difficult, if not impossible,
to design a biological phenotype at the whole-
organism level. Across biological outcomes, whether
in normal or in disease development, it is uncommon
that a single gene is the only factor shaping a complex
biological trait. Other genetic regulatory factors such as
additional genes or distal regulatory elements (e.g.,
enhancer or repressor elements), as well as environ-
mental and epigenetic factors, contribute to the
emergence of a biological phenotype. To argue that
modifying a gene changes a desired phenotype (under
certain conditions) implies at least a reasonable
understanding of other independent variables contrib-
uting to the phenotype's instantiation. But this under-
standing is still far from complete in many normal and
disease processes [44, 45]. Given the uncertainty
regarding how gene expression and modification
influence complex biological outcomes, it is difficult to
appraise potential risk and benefit. This ambiguity
creates a challenge on its own and is one of the sources
obscuring efficient ethical deliberation and decision
making.
Nevertheless, regulations governing ce llular- and
gene-therapy research may facilitate the safe devel-
opment and oversight of some clinical trials involving
CRISPR-based-editing applications. In the United
States, for instance, cellular- and gene-therapy
products, including many CRISPR applications, at
this time are defined generally by the Food and Drug
Administration as biological products and are regu-
lated by the Food and Drug Administration's Center
for Biologics Evaluation and Research/Office of
Cellular, Tissue , and Gene Therapies [4648].
Although the r isks and benefits of many such
therapies increasingly are better understood [49],
questions regarding safety and effi cacy remain.
Thus, future advancements likely will con tinue to
improve the benefits of this revolutionary technology,
while minimizing the potential risk s. Regardless of
the uncertainty posed by novel CRISPR technologies
and applications, in several locations around the
world significant regulatory frameworks exist by
which risks may be monitored and contained.
However, wherever such infrastructure and oversight
are lacking, safety and privacy risks might increase.
3Review: CRISPR Ethics
Please cite this article as: C. Brokowski, M. Adli, CRISPR Ethics: Moral Considerations for Applications of a Powerful Tool, J. Mol. Biol.
(2018), https://doi.org/10.1016/j.jmb.2018.05.044
Ethical concerns
To what extent should CRISPR experimentation
be permitted in basic and pre-clinical biomedical
research?
Although it is less than a decade old, CRISPR
Cas9 has demonstrated unprecedented potential to
revolutionize innovation in basic science. From viruses
and bacteria [50, 51], to simple model organisms, such
as Drosophila melanogaster (fruit fly) [52], Anopheles
gambiae (mosquito) [53], Saccharomyces cerevisiae
(budding yeast) [54], Hydra magnipapillata (hydra)
[55], Caenorhabditis elegans (round worm) [56],
Danio rerio (zebra fish) [57], and Arabidopsis thaliana
(rockcress) [58], to larger animals such as pigs [59],
cattle [60], and monkeys [61], and even human
Table 1. Risk-benefit considerations in CRISPR technology
Benefit(s) Risk(s)/Harm(s)
Basic and pre-clinical research New model organisms and cell lines
Increased gene-editing efficiency
High-throughput screens
Novel drug targets
Access to totipotent cells
Identification of novel signaling,
regulatory, and developmental pathways
Development of novel gene-editing
approaches (base editing and RNA targeting)
Knowledge advancement
Experimentation involving human embryos is
controversial and illegal in some countries
Potential for privacy and confidentiality breaches
Translational and
clinical medicine
Immunotherapy
Organoids
Novel drug targets
Artificial intelligence
Modification of pathological genes
Novel therapeutics and fertility
applications
Procreative liberty
Ability to fix single base changes
Knowledge advancement
Potential for equitable access
Serious injury, disability, and/or death to research
participant(s) and/or offspring
Blurry distinction between therapeutic and
enhancement applications, leading to potential
subtle or obvious exacerbation of inequalities
Misapplications
Eugenics
Potential for inequitable access and exacerbation
of inequalities
Non-therapeutic
applications
Enhancement to augment select faulty
or normal human characteristics
Fortification of crops and livestock
Successful control of pests, invasive
species, and reservoirs (gene drives)
Disease/infection control (e.g., malaria,
dengue fever, Lyme and Chagas
disease, schistosomiasis)
Ecosystem alteration to protect
endangered species (gene drives)
Safety
Crop cultivation
Knowledge advancement
Eugenics
Exacerbation of racism and inequality
Theoretical risk for damage to ecosystems
Theoretical risk of misuse
Access to CRISPR
technology
Inexpensive (technology itself)
Widely available
Profit, economic growth
Innovation
Price gouging
Prohibitively expensive applications
Regulations for
clinical research involving
human subjects
Established framework in some
countries to manage research risks
Legal mechanisms for redress
already exist, depending on location
Lack of appropriate supervisory infrastructure,
oversight, and/or regulatory framework
in many nations
Unclear how to supervise the research even in
some countries with regulatory oversight
Over-regulation might hinder progress
National and international
regulations, law, and policy
Prevention against misuses of technology
Safeguard against risky, potentially
harmful conditions
Potential to encroach on individual, scientific,
and societal autonomy
Limit discovery and progress
Difficult enforcement
Lack of uniformity may create inconsistencies
in applications of laws/regulations
4 Review: CRISPR Ethics
Please cite this article as: C. Brokowski, M. Adli, CRISPR Ethics: Moral Considerations for Applications of a Powerful Tool, J. Mol. Biol.
(2018), https://doi.org/10.1016/j.jmb.2018.05.044
zygotes [62, 63], CRISPR experimentation has led to
novel, important findings. Such benefits include at least
the following: increased overall efficiency in gene
editing compared with previous genomic engineering
techniques like transcription activator-like effector
nucleases (TALENs) and zinc finger nucleases
(ZNFs) [64]; significant insights into the evolutionary
transformation of fish fins into tetrapod limbs [65];
investigation into new organisms [66]; genetic and
epigenetic screens [67, 68]; the creation of novel cell
lines [69]; high-throughput screens and libraries [70];
the elucidation of novel genomic and epigenomic
regulatory pathways [71, 72]; insights into the devel-
opment of butterfly coloring and patterning [73];the
functional characterization of key genes and molecular
signaling pathways [74, 75]; and drug-targeting
screens [76, 77]. Data from such experimenta tion
provide essential clues and understanding that pro-
mote biomedical discovery, advancement, and the
basis for potential medical benefits.
One of the major controversies about CRISPR
technology emerges from its possible application in
hum an embryos. This controversy is not about
CRISPR itself, but instead is largely due to the lack of
clarity about the status of the human embryo. Although
some in the scie ntific community main tain that it is
ethically impermissible to exper iment on human
embryos a fter 14 days [78, 79], it is impossible for
any one partywhether it is a government, laboratory,
funding agency, panel of experts, court, religious
organization, or other groupto decide the status of
a human embryo [80, 81] and whether and precisely
when it has personhoo d [82]: Is the entity merely a ball
of cells whose status is like that of human skin, which
sheds regularly without further ado? Or does the entity
hold complete personhood statuswith irreducible,
inalienable moral rights and to whom we owe important
directed duties? Or is the embryo's status characterized
optimally as something in between? And if so, which
moral rights might this entity hold, and which duties
might we owe to it? Despite this perplexing complexity,
decisions one way or the other must be executed,
because proceeding with research or failing to do so
has important consequences: Banning or significantly
limiting research on human embryos stymies progress
at least by making unavailable or curtailing an option to
investigate the therapeutic potentials of stem cells and
the biology of totipotent cells, which currently are not
known to be present in any other viable human tissue
sources. Totipotent cells can divide indefinitely and
have the capacity to develop into all tissue types.
Depending on how the status of the embryo is
appraised, however, the ban also could save it from
potentially unjust, lethal research-related harms. Even
if the research is justified because of its potential benefit
to the embryo itself and/or to others, the embryo as
such cannot give informed consent at the time of the
research, since the entity is not sufficiently developed.
But from the research, it could experience potentially
life-altering consequencesgood or badthat may
extend throughout the lifespan and future generations.
By contrast, promoting such investigation may
facilitate the development of novel in vitro fertilization
techniques and advances for conditions such as
spinal cord injury [83], Parkinson's disease [84],
burns [85], cardiomyopathy [86], and other ailments
that might be ameliorated by approaches involving
regeneration. Taken together, countries must con-
tinue to decide as the science progresses whether
and how to legalize experimentation on human
embryos. Current positions across the globe vary
widelyfrom outright banning of the research to
illegalizing its federal funding only (while still allowing
private funding for research and the research itself)
to authorizing federal monies for experimentation
[4, 6, 87, 88].
To what extent should CRISPR use be permitted
in translational and clinical medicine?
CRISPR is significantly benefitting, and is likely to
improve, immunotherapy [89], organoid engineering
and development [90], in vivo drug target identifica-
tion [91], machine learning and artificial intelligence
[92], and disease-gene modification in viable human
embryos [62]. The system offers nearly boundless
potential to promote progress in combating HIV [93],
hemophilia [94], cancer [95], Duchenne muscular
dystrophy [96], amyotrophic lateral sclerosis [97],
sickle-cell anemia [98], cystic fibrosis [99], infertility
[100], and any number of novel diseases. The
potential both for gaining knowledge and for devel-
oping treatments in humans seems nearly endless.
However, such knowledge and treatment acquisition
are not without potential risk. With experimentation
involving somatic cells, risk assessment seems at least
comparable to that which arises in regularly practiced
biomedical testing. The chief objective of phase-2
oncology trials, for example, is to evaluate the efficacy
of a new drug or device [101]. Study participants may
assume significant harms, including possible irrevers-
ible side effects and death [102, 103].Inmany
countries, respect for autonomy permits assuming
such risk with the requirement that informed consent
occurs before enrollment in the research, regardless of
whether individuals are enrolling themselves or their
dependents. If this risk is considered morally (and
legally) permissible, then it would seem unjustified and
unreasonable to not allow risk posed by investigations
involving CRISPR-based genome engineering. At the
time of this writing, there is no empirical support
suggesting that CRISPR experimentation would nec-
essarily pose greater risk: the overall risk profile of
CRISPR experimentation in human subjects remains
unknown.
It could be argued, however, that heritable germ-
line editing might present additional risk, because it
involves not only the research participant but also
5Review: CRISPR Ethics
Please cite this article as: C. Brokowski, M. Adli, CRISPR Ethics: Moral Considerations for Applications of a Powerful Tool, J. Mol. Biol.
(2018), https://doi.org/10.1016/j.jmb.2018.05.044
potentially his or her descendants. Of course, whether
germline engineering technologies introduce risk be-
yond that which might be present in more common
testing scenarios is an empirical matter. For instance, it
is well established that routinely used chemotherapies
have mutagenic properties: alkylating agents, including
cisplatin and cyclophosphamides, cause DNA adducts
and crosslinks; antimetabolites, such as hydroxyurea,
gemcitabine, and 5-fluorouracil, are nucleoside ana-
logs and inhibit thymidine synthase; topoisomerases,
such as etoposide, cause topoisomerase II inhibition,
leading to double-stranded breaks in DNA; and
anthracyclines, like doxorubicin, cause DNA intercala-
tion [104]. Therefore, significant exp osure to any of
these agents increases the probability of both incurring
genetic mutations and passing on these unintended
genomic alterations to future generations. Whether the
risk level presented by such exposure is greater than,
equal to, or less than that presented by CRISPR
experimentation must be quantitatively determined by
empirical evidence. It is also an empirical matter
whether CRISPR introduces risk that is statistically
significant beyond that which is incurred in the daily
experience of a healthy individual with little-to-no
exposure to mutagenic agents. Thus, to determine
with confidence whether it is exceptionally risky to
involve humans in CRISPR translational and clinical
research, possible research-related risks must be
compared with those in other potentially dangerous
experimental and every-day contexts. This is difficult,
however, given that CRISPR technology is new and
that robust, reliable data about CRISPR risk in human
subjects are unavailable. Nevertheless, decision mak-
ing about assuming risk in studies and therapeutics
should be considered according to legal infrastructure,
national and possibly international regulatory agencies,
and ultimately navigated by research participants and/
or their legally authorized representative(s).
Important questions also arise about whether
experiments involving heritable germline editing
yield reliable, interpretable data. One objection is
that such experiments are unlikely to be controlled
and/or predicted [105] because it could be impossi-
ble to analyze or understand the results from such
experimentation until considerable time (decades or
even generations) passes [106]. As previously
noted, the central concern here is the uncertainty
in the causal connection between gene expression/
modification and the potential involvement of other
factors shaping biological outcomes in the future.
Another risk, shared globally, is posed by the
greater society. It is possible, for instance, that
allowing CRISPR germline editing, even if only for
medical purposes, might in some respect(s) lead to
the return of eugenics, whose proponents believed
that the human population can be improved by
controlled breeding to increase the occurrence of
desirable, heritable characteristics [107].Unfortu-
nately, historically, this selective weeding of people
with bad genes and breeding of those with good
ones resulted in many atrocities, including the forced
sterilization of individuals and the propagation of
racially discriminatory policiesboth of which were
backed by state authorities and even educated elites
in different societies. In the notorious case Buck v. Bell
[108], for example, the United States Supreme Court
(the Court) upheld a Virginia statute permitting the
compulsory sterilization of individuals, such as Carrie
Buck, who were considered mentally unfit. Buck was
an economically disadvantaged woman who was
labeled as feebleminded like her other family
members of past generations. She was committed
to the Virginia Colony for Epileptics and Feeble-
Minded and was forcibly sterilized [109]. Unfortunate-
ly, however, the evidence of the case strongly
indicates that Buck, like the others in her family, was
normal and that the Court erred gravely [110].Its
decision, authored by eugenics proponent Associate
Justice Oliver Wendell Holmes, led to the sterilization
of 50,000 Americans, set a precedent for the Nazi
racial hygiene program, and is yet to be overturned
[111]. Hence, history reveals that egregious, system-
atic mistakes are always possible.
To what extent should CRISPR use be permitted
for non-therapeutic purposes?
Impor tant et hical questions also arise in non-
therapeutic contexts including enhancement of
crops, livestock, gene drives, and human features
[112].
Certain areas of CRISPR applications, such as the
enhancement of crops and livestock, are likely to
significantly impact society and humanity at large. In
2016, the United Nati ons Food and Agriculture
Organization estimated that 795 million people in the
world were undernourished [113]. And according to
the World Health Organization, 2 billion people are
unable to obtain key nutrients like iron and vitamin A
[114]. A bundant evidence demonstrates that
CRISPRCas systems could be used to improve
nutrient content in foods [106, 115123]. In principle,
CRISPR has the potential to fortify foods efficiently for
individuals who are suffering from a lack of basic
nutrients. Why not decrease malnutrition by maximiz-
ing access to foods of higher quality? Promoting
benefit in this way carries moral weight at least
comparable to any other ethical concern raised
herein, especially given the very large number of
those who are nutrient-deprived. Nevertheless, with
this benefit arise worries about accessibility to these
product(s); this issue is discussed further below.
Gene drive technology is another CRISPR appli-
cation with unprecedented potential to directly benefit
andsavemillionsoflives[124]. In using gene drives,
researchers employ CRISPR to speed up genetic
recombination such that a gene of interest is rapidly
distributed to the entire population much faster than in
6 Review: CRISPR Ethics
Please cite this article as: C. Brokowski, M. Adli, CRISPR Ethics: Moral Considerations for Applications of a Powerful Tool, J. Mol. Biol.
(2018), https://doi.org/10.1016/j.jmb.2018.05.044
a typical Mendelian inheritance rate. Therefore, this
application has the potential to edit the genome of an
entire population or even an entire species. Using
CRISPR-mediated gene drives, investigators have
demonstrated that a gene allele providing a parasite-
resistance phenotype in mosquitos could quickly
spread through the population in a non-Mendelian
fashion [111, 125]. This highly cost-effective technol-
ogy has many potential benefits and applications for
public health, species conservation, agriculture, and
basic research [126]: Gene drives may provide a
fundamental tool to fight against deadly diseases such
as malaria [127129], dengue fever [130],Chagas
and Lyme disease [131], and schistosomiasis [132].
This application also may control and/or alter a wide
variety of animals (e.g., rodents and bats), invasive
plant pests, and reservoirs [133, 134]. Thus, the
technology has unprecedented power that may save
millions of lives each year. However, it is also
important to consider the expected and unexpected
risks. Once applied, gene drives will eventually affect
every individual of the entire species. Knowing this,
researchers are developing and incorporating key
safety off-switch features such as novel ways to (i)
control, (ii) inhibit, and (iii) reverse/eliminate gene
drive systems from the population in case of an
unexpected or emergency event [134137].
Furthermore, were it possible, would it be it morally
permissible to employ CRISPR techniques to en-
hance human features such as height, muscle mass,
vision, or cognitive factors like learning aptitude and
memory? Answering this question is problematic
largely because of the difficulty with deciding about
what counts as pathology vs. what is merely a minor
or even moderate deviation from the norm in a given
context. Moreover, accurately characterizing norms in
the first place is often very difficult. Hence, medical
necessity often becomes ambiguous, and the bound-
ary between therapy and enhancement can be
murky. For example, a gene-editing approach may
allow for a reduction in bad cholesterol, thus leading to
a healthier life style. Whether this hypoth etical
scenario, which may benefit both the individual and
society in the long run, should be classified as
enhancement or a medical need is unclear.
Aside from concerns about well characterizing
medical necessity, positive moral liberties are granted
and backed by legal rights in many countries, especially
in the West. Should medical enhancement by CRISPR
technologies be considered a form of free speech and/
or expression? [138] If so, how, if at all, might these
rights be limited, and why? Who has and/or should
have the authority to decide?
Who should have access to CRISPR technology
and/or its products?
Benefits from CRISPR innovation raise concerns
and controversies about fairness and distributive
justice across all layers of society. These matters are
not specific to CRISPR technology, but may apply to
all other technologies arising from academic re-
search. Like many novel biomedical advancements,
new CRISPR applications are expected to be
profitable for patent holders. At least the initial prices
of CRISPR-bas ed products, such as gene therapy,
are likely to be costly [139]. To this end, an ethical
question is whether the high price-tag will make the
CRISPR product available to only the world's elites.
Since much of the funding for CRISPR characteri-
zation and development was provided by grants
from government funds and thus taxpayers' money
[140151], it is morally problematic to deny poten-
tially lifesaving benefits of the technology to the very
individuals who funded much of its development in
the first place. Moreover, even if it were affordable for
some, there may be economic harms associated
with high-price purchases. For instance, those
needing CRISPR-based applications to maintain a
reasonable quality of life, or even life itself, might be
forced to ma ke painful economic choices about
whether to spend funds on therapuetics, food, or
other essential living necessities. While this problem is
not unique to genomic engineering advancements,
allowing price gouging to continue unaddressed is
unhelpful and potentially allows physical, psycholog-
ical, and economic harms to continue. Encouraging
the establishment of anti-price-gouging laws, where
possible, could ameliorate some of these concerns
[152].
As discussed in the previous section, CRISPR
may be used to fortify foods. Those residing in some
of the most impoverished areas of the world are
positioned to benefit the greatest from the se
products. How might such individuals gain access
to CRISPR-modified foods, especially in places with
armed combat and rogue governments? How, if at
all, could companies benefit such that reaching out
to these populations mig ht be desirable and/or
lucrative?
Limiting human genome editing? Somatic
versus germline editing
As noted above, obvious applications of CRISPR
technology are cell and gene therapies. To date, gene
therapy mostly involves the use of genome-
engineering technologies to edit somatic cells to
treat genetic diseases. Clinical t rials involving
CRISPR-based gene therapy are already under
way. Although clinical gene and cell therapies have
had major road blocks in the past, due to unanticipat-
ed injuries and death [153, 154], significant safety
improvements have been implemented over the last
decade [155]. With the advances of CRISPR technol-
ogy and safer delivery approaches, t herapeutic
applications of gene therapy are on the rise [156].In
the United States and elsewhere, federal regulations
7Review: CRISPR Ethics
Please cite this article as: C. Brokowski, M. Adli, CRISPR Ethics: Moral Considerations for Applications of a Powerful Tool, J. Mol. Biol.
(2018), https://doi.org/10.1016/j.jmb.2018.05.044
provide the needed legal and ethical frameworks,
through the institutional review board system, to
potentially minimize and manage po tenti al risks
[156160].
At present, there is considerable excitement
about such experimentation in the United States. In
January of 2018, for example, the US National
Institutes of He alth launched the Somatic Cell
Genome Editing program, seeking to improve the
delivery mechanisms for target ing gene editing tools
in patients, develop new and improved genome
editors, develop assays for testing the safety and
efficacy of the genome editing tools in animal and
human cells, and assemble a genome editing toolkit
containing the resulting knowledge, methods, and
tools to be shared with the scientific community
[161].
Heritable genome editing, by contrast, is perhaps
the CRISPR systems' greatest discussed controver-
sy. Recently, professional scientific and medical
societies, industry organizations, and CRISPR pio-
neers together have released g reater than 60
statements and reports about whether such editing
in humans is morally permissible [6]. Most state-
ments hold that heritable germline experimentation
should be prohibited currently, although reports from
the Netherlands [162], the United Kingdom [163],
Spain [164], and the United States [9] suggest that
editing could be permissible if certain requir ements
were satisfied. The NASEM Committee's report on
germline editing, for example, specified that the
following provisions must be met for human her itable
germline research to commence: the absence of
reasonable alternatives; restriction to preventing a
serious disease or condition; restriction to editin g
genes that have been convincingly demonstrated to
cause or to strongly predispose to that disease or
condition; restriction to converting such genes to
versions that are prevalent in the population and are
known to be associated with ordinary health with little
or no evidence of adverse effects; the availability of
credible preclinical and/or clinical data on risks and
potential health benefits of the procedures; ongoing,
rigorous oversight during clinical trials of the effect s
of the procedure on the health and safety of the
research participants; comprehensive plans for long-
term, multigenerational follow up that still respect
personal autonomy; maximum transparency consis-
tent with patient privacy; continued reassessment of
both health and societal benefits and risks, with
broad ongoing participat ion and input by the public;
and reliable oversight mechanisms t o prevent
extension to uses other than preventing a serious
disease or condit ion [9]. Although fears about
misuse in this context abound, it is important to
point out that there are reasonable arguments
supporting heritable germline editing in research,
such as the protection of defective embryos [165],
the elimination of certain diseases that might be
obliterated optimally early in embryonic develop-
ment, and the exerc ise of free speech and/or
expression [138].
Should international regulations governing
CRISPR use be crafted and promulgated?
Although ethics statements are important, by
themselves, they provide little force. Typically, if
ethics guidelines are infringed, the consequences
suffered by the violator(s) are fairly minimal com-
pared to those arising when in violation of certain
laws. Violations of ethics statements may lead to
loss of funding, the retraction of a publication(s), job
loss, and mistrust among the biomedical community.
By contrast, punishments by law may lead to heavy
fines and potentially incarceration. Given the signif-
icant potential promise, the dark history of eugenics,
the potentially serious transgenerational risks, and
the theoretical potential for misuse, it is reasonable
for the global community to consider instantiating
national and supranational regulations, if not revising
older agreements such as the Geneva [166] and the
United Nations Conventions on Biological and Toxin
Weapons, [167] to reflect changes in genomic
engineering technologies. While doing so likely will
not eliminate all risks, it is arguably one of the few
options available to reasonably control and/or
minimize them.
Conclusions and future directions
CRISPR technology continues to mature, and
existing systems are being engineered to contain
innovative capabilities; excitingly new CRISPR sys-
tems with novel functions are still being discovered.
The potential benefits of such revolutionary tools are
endless. However, like any powerful tool, there are also
associated risks raising moral concerns. To make truly
informed decisions about areas of ethical controversy,
well-controlled, reproducible experimentation and clin-
ical trials are warranted. Currently, this is difficult
because many international laws discourage or
ban such research and/or inhibit its funding for
certain types of investigation. Thus, widespread
data about benefits and risks are unavailable. It is
critical, however, for countries to examine their
reasoning behind these prohibitions to ensure that
they are not simply arising out of fear and without
reasonable justification.
Going forward, many support establishing an
organization that will decide how best to address the
aforem entioned ethical complexities. Recently, a
group of European scientists founded the Association
for Responsible Research and Innovation in Genome
Editing (ARRIGE) to examine and provide guidance
about the ethical use of genome editing [168, 169].
Furthermore, Jasanoff and Hurlbut [170] recently
8 Review: CRISPR Ethics
Please cite this article as: C. Brokowski, M. Adli, CRISPR Ethics: Moral Considerations for Applications of a Powerful Tool, J. Mol. Biol.
(2018), https://doi.org/10.1016/j.jmb.2018.05.044
advocated for the development of an international,
interdisciplinary global observatory for gene editing.
Briefly, they argued that deliberations about moral
issues in gene editing should not be dominated by the
scientific community, but instead should include a
network of scholars and organizations similar to
those established for human rights and climate
change. The network would be dedicated to gathering
information from dispersed sources, bringing to the
fore perspectives that are often overlooked, and
promoting exchange across disciplinary and cultural
divides [170].
As the technology evolves, so will discussions
about ethical and legal frameworks circumscribing
its uses. The above-mentioned platforms present
interesting ideas for furthering debates and potential
resolutions. The research and ethical guidelines
from national and international organizations, where
diverse disciplines of societies contribute, will be
critical for federal funding agencies and institutional
review boards to enforce and regulate, to minimize
the potentials risks and maximize the potential
benefits of CRISPR technology. However, it is likely
that the enforcement of research laws and ethical
guidelines ultimately will be assumed by govern-
ments and their legal systems, principal investiga-
tors, and institutional review boards.
Acknowledgment
We are thankful to all the Adli lab members. The
research in Dr. Adli's lab is supported through local
funds from University of Virginia School of Medicine
and federal grants from National Institutes of Health/
National Cancer Institute 1R01 CA211648-01.
Author Contributions: M.A. and C.B. conceptu-
alized the study and wrote the manuscript.
Received 18 March 2018;
Received in revised form 30 May 2018;
Accepted 30 May 2018
Available online xxxx
Keywords:
CRISPRCas9;
research ethics;
genome editing;
genetic engineering;
bioengineering
Abbreviations used:
CRISPR, Clustered Regularly Interspaced Short
Palindromic Repeats; Cas9, CRISPR-associated protein 9;
NASEM, US National Academies of Sciences, Engineering,
and Medicine.
References
[1] D. Cyranoski, CRISPR gene-editing tested in a person for
the first time, Nature 539 (2016) 479.
[2] CRISPR to debut in clinical trials, D. Kwon, Israel Molecular
Medicine, [cited 2018 May 15] Available from: http://www.
molecular-medicine-israel.co.il/crispr-debut-clinical-trials/
2017.
[3] ClinicalTrials.gov, U.S. National Library of medicine, [cited
2018 May 20]. Available from: https://clinicaltrials.gov/ct2/
results?cond=&t erm=CRISPR&cntry=&state=&city=&dist=
2018.
[4] L. The, Editing the human genome: balancing safety and
regulation, Lancet 391 (2018) 402.
[5] D.A. Scheufele, M.A. Xenos, E.L. Howell, K.M. Rose, D.
Brossard, B.W. Hardy, U.S. attitudes on human genome
editing, Science 357 (2017) 553554.
[6] C. Brokowski, Do CRISPR germline ethics statements cut
it? CRISPR J. 1 (2) (2018) 115125.
[7] D. Baltimore, P. Berg, M. Botchan, D. Carroll, R.A. Charo,
G. Church, et al., Biotechnology. A prudent path forward for
genomic engineering and germline gene modification,
Science 348 (2015) 3638.
[8] International Summit on Human Gene Editing: A Global
Discussion [Internet]. National Academy of Sciences
Engineering and Medicine, U.S., 2015, [cited 2018 Feb
16]. Available from: http://nationalacademies.org/cs/groups/
pgasite/documents/webpage/pga_170455.pdf.
[9] National Academies of Sciences Engineering and Medicine
(U.S.), Human Genome Editing: Science, Ethics, and
Governance, The National Academies Press, Washington,
DC, 2017.
[10] Many countries and international decision-making bodies,
including the Nuffield Council on Bioethics, the European
Society of Human Genetics, and the European Society of
Reproduction have issued reports and statements about
ethical issues in genomic engineering, focusing specifically
on the moral permissibility of germline editing. However, to
date, the U.S. NASEM report (2017) is the most robust.
[11] With stringent oversight, heritable germline editing clinical trials
could one day be permitted for serious conditions; non-
heritable clinical trials should be limited to treating or preventing
disease or disability at this time, National Academies of
Sciences Engineering and Medicine (News). [2018 May 20].
Available from: http://www8.nationalacademies.org/onpinews/
newsitem.aspx?RecordID=24623 2017.
[12] Code of Federal Regulations: Research Involving Pregnant
Women or Fetuses, 45 C.F.R. Sect 462017.
[13] United States Code: Institutional Review Boards; Ethics
Guidance Program, 42 U.S. Code Sect 2892017.
[14] Consolidated Appropriations Act of 2016 [statute on the
internet] c2017, [cited 2017 Nov]. Available from: https://
www.congress.gov/114/bills/hr2029/BILLS-114hr2029enr.
pdf.
[15] J.A. Robertson, Human embryonic stem cell research:
ethical and legal issues, Nat. Rev. Genet. 2 (2001) 7478.
[16] D.H. Haft, J. Selengut, E.F. Mongodin, K.E. Nelson, A guild
of 45 CRISPR-associated (Cas) protein families and
multiple CRISPR/Cas subtypes exist in prokaryotic ge-
nomes, PLoS Comput. Biol. 1 (2005), e60.
[17] K.S. Makarova, D.H. Haft, R. Barrangou, S.J. Brouns, E.
Charpentier, P. Horvath, et al., Evolution and classification
of the CRISPRCas systems, Nat. Rev. Microbiol. 9 (2011)
467477.
9Review: CRISPR Ethics
Please cite this article as: C. Brokowski, M. Adli, CRISPR Ethics: Moral Considerations for Applications of a Powerful Tool, J. Mol. Biol.
(2018), https://doi.org/10.1016/j.jmb.2018.05.044
[18] K.S. Makarova, Y.I. Wolf, O.S. Alkhnbashi, F. Costa, S.A.
Shah, S.J. Saunders, et al., An updated evolutionary
classification of CRISPRCas systems, Nat. Rev. Microbiol.
13 (11) (2015) 722736.
[19] E.V. Koonin, K.S. Makarova, F. Zhang, Diversity, classifi-
cation and evolution of CRISPRCas systems, Curr. Opin.
Microbiol. 37 (2017) 6778.
[20] S. Shmakov, A. Smargon, D. Scott, D. Cox, N. Pyzocha, W.
Yan, et al., Diversity and evolution of class 2 CRISPRCas
systems, Nat. Rev. Microbiol. 15 (3) (2017) 169182.
[21] B.P. Kleinstiver, M.S. Prew, S.Q. Tsai, V.V. Topkar, N.T.
Nguyen, Z. Zheng, et al., Engineered CRISPRCas9
nucleases with altered PAM specificities, Nature 523
(2015) 481485.
[22] J.H. Hu, S.M. Miller, M.H. Geurts, W. Tang, L. Chen, N. Sun,
et al., Evolved Cas9 variants with broad PAM compatibility
and high DNA specificity, Nature 556 (2018) 5763.
[23] H. Nishimasu, F.A. Ran, P.D. Hsu, S. Konermann, S.I.
Shehata, N. Dohmae, et al., Crystal structure of Cas9 in
complex with guide RNA and target DNA, Cell 156 (2014)
935949.
[24] L.S. Qi, M.H. Larson, L.A. Gilbert, J.A. Doudna, J.S.
Weissman, A.P. Arkin, et al., Repurposing CRISPR as an
RNA-guided platform for sequence-specific control of gene
expression, Cell 152 (2013) 11731183.
[25] M. Adli, The CRISPR tool kit for genome editing and
beyond, Nat. Commun. 2018 (9) (1911).
[26] M. Jinek, K. Chylinski, I. Fonfara, M. Hauer, J.A. Doudna, E.
Charpentier, A programmable dual-RNA-guided DNA en-
donuclease in adaptive bacterial immunity, Science 337
(2012) 816821.
[27] P. Mali, J. Aach, P.B. Stranges, K.M. Esvelt, M. Moosburner,
S. Kosuri, et al., CAS9 transcriptional activators for target
specificity screening and paired nickases for cooperative
genome engineering, Nat. Biotechnol. 31 (2013) 833838.
[28] F.A. Ran, P.D. Hsu, J. Wright, V. Agarwala, D.A. Scott, F.
Zhang, Genome engineering using the CRISPRCas9
system, Nat. Protoc. 8 (2013) 22812308.
[29] N.M. Gaudelli, A.C. Komor, H.A. Rees, M.S. Packer, A.H.
Badran, D.I. Bryson, et al., Programmable base editing of
A*T to G*C in genomic DNA without DNA cleavage, Nature
551 (2017) 464471.
[30] A.C. Komor, Y.B. Kim, M.S. Packer, J.A. Zuris, D.R. Liu,
Programmable editing of a target base in genomic DNA
without double-stranded DNA cleavage, Nature 533 (2016)
420424.
[31] G.T. Hess, J. Tycko, D. Yao, M.C. Bassik, Methods and
applications of CRISPR-mediated base editing in eukaryotic
genomes, Mol. Cell 68 (2017) 2643.
[32] C. Kuscu, M. Parlak, T. Tufan, J. Yang, K. Szlachta, X. Wei,
et al., CRISPR-STOP: gene silencing through base-editing-
induced nonsense mutations, Nat. Methods 14 (2017)
710712.
[33] A.C. Komor, A.H. Badran, D.R. Liu, CRISPR-based
technologies for the manipulation of eukaryotic genomes,
Cell 169 (2017) 559.
[34] O.O. Abudayyeh, J.S. Gootenberg, P. Essletzbichler, S.
Han, J. Joung, J.J. Belanto, et al., RNA targeting with
CRISPRCas13, Nature 550 (2017) 280284.
[35] S.C. Strutt, R.M. Torrez, E. Kaya, O.A. Negrete, J.A.
Doudna, RNA-dependent RNA targeting by CRISPR
Cas9, elife 7 (2018).
[36] CRISPRCas12a target binding unleashes indiscriminate
single-stranded DNase activity, Science 360 (2018) 436439.
[37] J.S. Gootenberg, O.O. Abudayyeh, M.J. Kellner, J. Joung,
J.J. Collins, F. Zhang, Multiplexed and portable nucleic acid
detection platform with Cas13, Cas12a, and Csm6, Science
360 (2018) 439444.
[38] R. Peng, G. Lin, J. Li, Potential pitfalls of CRISPR/Cas9-
mediated genome editing, FEBS J. 283 (2016) 12181231.
[39] D. Bohaciakova, T. Renzova, V. Fedorova, M. Barak, M.
Kunova Bosakova, A. Hampl, et al., An efficient method for
generation of knockout human embryonic stem cells using
CRISPR/Cas9 System, Stem Cells Dev. 26 (2017) 15211527.
[40] X. Guo, X.J. Li, Targeted genome editing in primate
embryos, Cell Res. 25 (2015) 767768.
[41] Z. Tu, W. Yang, S. Yan, A. Yin, J. Gao, X. Liu, et al.,
Promoting Cas9 degradation reduces mosaic mutations in
non-human primate embryos, Sci. Rep. 7 (2017), 42081.
[42] K.A. Schaefer, W.H. Wu, D.F. Colgan, S.H. Tsang, A.G.
Bassuk, V.B. Mahajan, Unexpected mutations after
CRISPRCas9 editing in vivo, Nat. Methods 14 (2017)
547548.
[43] J. Zischewski, R. Fischer, L. Bortesi, Detection of on-target
and off-target mutations generated by CRISPR/Cas9 and
other sequence-specific nucleases, Biotechnol. Adv. 35
(2017) 95104.
[44] A. Jelenkovic, R. Sund, Y.M. Hur, Y. Yokoyama, J.V.
Hjelmborg, S. Moller, et al., Genetic and environmental
influences on height from infancy to early adulthood: An
individual-based pooled analysis of 45 twin cohorts, Sci.
Rep. 6 (2016), 28496.
[45] S. Wu, S. Powers, W. Zhu, Y.A. Hannun, Substantial
contribution of extrinsic risk factors to cancer development,
Nature 529 (2016) 4347.
[46] Code of Federal Regulations: Food and Drugs, 21 C.F.R.
(scattered), 2017.
[47] United States Code: Regulation of Biological Products, 42
U.S. Code Sect 2622017.
[48] Such guidance is inapplicable to cells, tissues, and cellular-
and tissue-based products regulated under United States
Code: Regulations to Control Communicable Diseases, 42
U.S. Code Sect 2642017.
[49] J. Kimmelman, Ethics of cancer gene transfer clinical
research, Methods Mol. Biol. 1317 (2015) 263285.
[50] Y.D. Tang, J.T. Liu, T.Y. Wang, M.X. Sun, Z.J. Tian, X.H.C.
R.I.S.P.R. Cai, Cas9-mediated multiple single guide RNAs
potently abrogate pseudorabies virus replication, Arch.
Virol. 162 (2017) 38813886.
[51] S.L. Shipman, J. Nivala, J.D. Macklis, G.M. Church,
CRISPRCas encoding of a digital movie into the genomes
of a population of living bacteria, Natu re 547 (2017)
345
349.
[52] S.J. Gratz, A.M. Cummings, J.N. Nguyen, D.C. Hamm, L.K.
Donohue, M.M. Harrison, et al., Genome engineering of
Drosophila with the CRISPR RNA-guided Cas9 nuclease,
Genetics 194 (2013) 10291035.
[53] S. Dong, J. Lin, N.L. Held, R.J. Clem, A.L. Passarelli, A.W.
Franz, Heritable CRISPR/Cas9-mediated genome editing in
the yellow fever mosquito, Aedes aegypti, PLoS One 10
(2015), e0122353.
[54] J.E. DiCarlo, J.E. Norville, P. Mali, X. Rios, J. Aach, G.M.
Church, Genome engineering in Saccharomyces cerevisiae
using CRISPRCas systems, Nucleic Acids Res. 41 (2013)
43364343.
[55] Y. Kraus, A. Aman, U. Technau, G. Genikhovich, Pre-
bilaterian origin of the blastoporal axial organizer, Nat.
Commun. 7 (2016), 11694.
10 Review: CRISPR Ethics
Please cite this article as: C. Brokowski, M. Adli, CRISPR Ethics: Moral Considerations for Applications of a Powerful Tool, J. Mol. Biol.
(2018), https://doi.org/10.1016/j.jmb.2018.05.044
[56] A.E. Friedland, Y.B. Tzur, K.M. Esvelt, M.P. Colaiacovo, G.M.
Church, J.A. Calarco, Heritable genome editing in C. elegans
via a CRISPRCas9 system, Nat. Methods 10 (2013)
741743.
[57] U. Irion, J. Krauss, C. Nusslein-Volhard, Precise and
efficient genome editing in zebrafish using the CRISPR/
Cas9 system, Development 141 (2014) 48274830.
[58] Z. Feng, B. Zhang, W. Ding, X. Liu, D.L. Yang, P. Wei, et al.,
Efficient genome editing in plants using a CRISPR/Cas
system, Cell Res. 23 (2013) 12291232.
[59] T. Hai, F. Teng, R. Guo, W. Li, Q. Zhou, One- step
generation of knockout pigs by zygote injection of
CRISPR/Cas system, Cell Res. 24 (2014) 372375.
[60] M. Ikeda, S. Matsuyama, S. Akagi, K. Ohkoshi, S.
Nakamura, S. Minabe, et al., Correction of a disease
mutation using CRISPR/Cas9-assisted genome editing in
Japanese black cattle, Sci. Rep. 7 (2017), 17827.
[61] E. Zuo, Y.J. Cai, K. Li, Y. Wei, B.A. Wang, Y. Sun, et al.,
One-step generation of complete gene knockout mice and
monkeys by CRISPR/Cas9-mediated gene editing with
multiple sgRNAs, Cell Res. 27 (2017) 933945.
[62] H. Ma, N. Marti-Gutierrez, S.W. Park, J. Wu, Y. Lee, K.
Suzuki, et al., Correction of a pathogenic gene mutation in
human embryos, Nature 548 (2017) 413419.
[63] X. Kang, W. He, Y. Huang, Q. Yu, Y. Chen, X. Gao, et al.,
Introducing precise genetic modifications into human 3PN
embryos by CRISPR/Cas-mediated genome editing, J. Assist.
Reprod. Genet. 33 (2016) 581588.
[64] T. Gaj, C.A. Gersbach, C.F. Barbas III, ZFN, TALEN, and
CRISPR/Cas-based methods for genome engineering,
Trends Biotechnol. 31 (2013) 397405.
[65] T. Nakamura, A.R. Gehrke, J. Lemberg, J. Szymaszek, N.H.
Shubin, Digits and fin rays share common developmental
histories, Nature 537 (2016) 225228.
[66] J.J. Russell, J.A. Theriot, P. Sood, W.F. Marshall, L.F.
Landweber, L. Fritz-Laylin, et al., Non-model model
organisms, BMC Biol. 15 (2017) 55.
[67] W. Shang, F. Wang, G. Fan, H. Wang, Key elements for
designing and performing a CRISPR/Cas9-based genetic
screen, J. Genet. Genomics 44 (2017) 439449.
[68] H.K. Liao, F. Hatanaka, T. Araoka, P. Reddy, M.Z. Wu, Y.
Sui, et al., In vivo target gene activation via CRISPR/Cas9-
Mmdiated trans-epigenetic modulation, Cell 171 (2017)
(1495-507.e15).
[69] M. Yang, L. Zhang, J. Stevens, G. Gibson, CRISPR/Cas9
mediated generation of stable chondrocyte cell lines with
targeted gene knockouts; analysis of an aggrecan knockout
cell line, Bone 69 (2014) 118125.
[70] X.J. Lu, Y.Y. Xiang, X.J. Li, CRISPR screen: a high-
throughput approach for cancer genetic research, Clin.
Genet. 88 (2015) 3233.
[71] E. Charpentier, H. Richter, J. van der Oost, M.F. White,
Biogenesis pathways of RNA guides in archaeal and
bacterial CRISPRCas adaptive immunity, FEMS Microbiol.
Rev. 39 (2015) 428441.
[72] T.S. Klann, J.B. Black, M. Chellappan, A. Safi, L. Song, I.B.
Hilton, et al., CRISPRCas9 epigenome editing enables
high-throughput screening for functional regulatory ele-
ments in the human genome, Nat. Biotechnol. 35 (2017)
561
568.
[73] A. Mazo-Vargas, C. Concha, L. Livraghi, D. Massardo, R.W.
R. Wallbank, L. Zhang, et al., Macroevolutionary shifts of
WntA function potentiate butterfly wing-pattern diversity,
Proc. Natl. Acad. Sci. U. S. A. 114 (2017) 1070110706.
[74] P. Atanes, I. Ruz-Maldonado, R. Hawkes, B. Liu, S.J.
Persaud, S. Amisten, Identifying signall ing pathways
regulated by GPRC5B in beta-cells by CRISPRCas9-
mediated genome editing, Cell. Physiol. Biochem. 45 (2018)
656666.
[75] S. Chi, A. Weiss, H.A. Wang, CRISPR-based toolbox for
studying T cell signal transduction, Biomed. Res. Int. 2016
(2016), 5052369.
[76] C. Kasap, O. Elemento, T.M. Kapoor, DrugTargetSeqR: a
genomics- and CRISPRCas9-based method to analyze
drug targets, Nat. Chem. Biol. 10 (2014) 626628.
[77] B.E. Housden, A.J. Valvezan, C. Kelley, R. Sopko, Y. Hu, C.
Roesel, et al., Identification of potential drug targets for
tuberous sclerosis complex by synthetic screens combining
CRISPR-based knockouts with RNAi, Sci. Signal. 8 (2015) rs9.
[78] I. Hyun, A. Wilkerson, J. Johnston, Embryology policy:
revisit the 14-day rule, Nature 533 (7602) (2016) 169171.
[79] Human embryo culture: discussions concerning the statu-
tory time limit for maintaining human embryos in culture in
light of some recent scientific developments. Nuffield
Council on Bioethics, 2017 [cited 2018 May 29]. Available
from: http://nuffi eldbioethics.org/wp-content/uploads/
Human-Embryo-Culture-web-FINAL.pdf.
[80] R.A. Charo, The hunting of the snark: the moral status of
embryos, right-to-lifers, and Third World women, Stanf. Law
Pol. Rev. 6 (1995) 1137.
[81] B. Wennergr en, Hum an ri ghts of an em bryo , J. Int .
Bioethique 2 (1991) 4649.
[82] J.J. Miklavcic, P. Flaman, Personhood status of the human
zygote, embryo, fetus, Linacre Q. 84 (2017) 130144.
[83] J.W. McDonald, X.Z. Liu, Y. Qu, S. Liu, S.K. Mickey, D.
Turetsky, et al., Transplanted embryonic stem cells survive,
differentiate and promote recovery in injured rat spinal cord,
Nat. Med. 5 (1999) 14101412.
[84] L.M. Bjorklund, R. Sanchez-Pernaute, S. Chung, T.
Andersson, I.Y. Chen, K.S. McNaught, et al., Embryonic
stem cells develop into functional dopaminergic neurons
after transplantation in a Parkinson rat model, Proc. Natl.
Acad. Sci. U. S. A. 99 (2002) 23442349.
[85] V.F. Hamrahi, J. Goverman, W. Jung, J.C. Wu, A.J.
Fischman, R.G. Tompkins, et al., In vivo molecular imaging
of murine embryonic stem cells delivered to a burn wound
surface via Integra(R) scaffolding, J. Burn Care Res. 33
(2012) e49e54.
[86] D.K. Singla, A. Ahmed, R. Singla, B. Yan, Embryonic stem
cells improve cardiac function in doxorubicin-induced
cardiomyopathy mediated through multiple mechanisms,
Cell Transplant. 21 (2012) 19191930.
[87] F.P. Busardo, M. Gulino, S. Napoletano, S. Zaami, P. Frati,
The evolution of legislation in the field of medically assisted
reproduction and embryo stem cell research in European
union members, Biomed. Res. Int. 2014 (2014), 307160.
[88] R. Dajani, Jordan's stem-cell law can guide the Middle East,
Nature 510 (2014) 189.
[89] R.T. Manguso, H.W. Pope, M.D. Zimmer, F.D. Brown, K.B.
Yates, B.C. Miller, et al., In vivo CRISPR screening
identifies Ptpn2 as a cancer immunotherapy target, Nature
547 (2017) 413418.
[90] J. Drost, R. van Boxtel, F. Blokzijl, T. Mizutani, N. Sasaki, V.
Sasselli, et al., Use of CRISPR-modified human stem cell
organoids to study the origin of mutational signatures in
cancer, Science 358 (2017) 234
238.
[91] J. Chu, G. Galicia-Vazquez, R. Cencic, J.R. Mills, A.
Katigbak, J.A. Porco Jr., et al., CRISPR-mediated drug-
11Review: CRISPR Ethics
Please cite this article as: C. Brokowski, M. Adli, CRISPR Ethics: Moral Considerations for Applications of a Powerful Tool, J. Mol. Biol.
(2018), https://doi.org/10.1016/j.jmb.2018.05.044
target validation reveals selective pharmacological inhibi-
tion of the RNA helicase, eIF4A, Cell Rep. 15 (2016)
23402347.
[92] S.H. Hough, A. Ajetunmobi, L. Brody, N. Humphryes-Kirilov, E.
Perello, Desktop Genetics, Perinat. Med. 13 (2016) 517521.
[93] Z. Huang, A. Tomitaka, A. Raymond, M. Nair, Current
application of CRISPR/Cas9 gen e-editing technique to
eradication of HIV/AIDS, Gene Ther. 24 (2017) 377384.
[94] L.A. George, S.K. Sullivan, A. Giermasz, J.E.J. Rasko, B.J.
Samelson-Jones, J. Ducore, et al., Hemophilia B gene therapy
with a high-specific-activity factor IX variant, N. Engl. J. Med.
377 (2017) 22152227.
[95] C. Wei, F. Wang, W. Liu, W. Zhao, Y. Yang, K. Li, et al.,
CRISPR/Cas9 targeting of the androgen receptor sup-
presses the growth of LNCaP human prostate cancer cells,
Mol. Med. Rep. 17 (2018) 29012906.
[96] T.W.Y. Wong, R.D. Cohn, Therapeutic applications of
CRISPR/Cas for Duchenne muscular dystrophy, Curr.
Gene Ther. 17 (2017) 301308.
[97] T. Gaj, D.S. Ojala, F.K. Ekman, L.C. Byrne, P. Limsirichai,
D.V. Schaffer, In vivo genome editing improves motor
function and extends survival in a mouse model of ALS, Sci.
Adv. 3 (2017) eaar3952.
[98] J. Wen, W. Tao, S. Hao, Y. Zu, Cellular function reinstitution of
offspring red blood cells cloned from the sickle cell disease
patient blood post CRISPR genome editing, J. Hematol.
Oncol. 10 (2017) 119.
[99] G. Schwank, B.K. Koo, V. Sasselli, J.F. Dekkers, I. Heo, T.
Demircan, et al., Functional repair of CFTR by CRISPR/
Cas9 in intestinal stem cell organoids of cystic fibrosis
patients, Cell Stem Cell 13 (2013) 653658.
[100] Y. Wu, H. Zhou, X. Fan, Y. Zhang, M. Zhang, Y. Wang,
et al., Correction of a genetic disease by CRISPRCas9-
mediated gene editing in mouse spermatogonial stem cells,
Cell Res. 25 (2015) 6779.
[101] Code of Federal Regulations: Investigational New Drug, 21
C.F.R. Sec 3122017.
[102] G. Suntharalingam, M.R. Perry, S. Ward, S.J. Brett, A.
Castello-Cortes, M.D. Brunner, et al., Cytokine storm in a
phase 1 trial of the anti-CD28 monoclonal antibody
TGN1412, N. Engl. J. Med. 355 (2006) 10181028.
[103] R. Kaur, P. Sidhu, S. Singh, What failed BIA 10-2474 Phase
I clinical trial? Global speculations and recommendations
for future Phase I trials, J. Pharmacol. Pharmacother. 7
(2016) 120126.
[104] B.Szikriszt,A.Poti,O.Pipek,M.Krzystanek,N.Kanu,J.
Molnar, et al., A comprehensive survey of the mutagenic impact
of common cancer cytotoxics, Genome Biol. 17 (2016) 99.
[105] T. Friedmann, E.C. Jonlin, King NMP, B.E. Torbett, N.A.
Wivel, Y. Kaneda, et al., ASGCT and JSGT Joint position
statement on human genomic editing, Mol. Ther. 23 (2015)
1282.
[106] X. Wang, Y. Niu, J. Zhou, H. Yu, Q. Kou, A. Lei, et al.,
Multiplex gene editing via CRISPR/Cas9 exhibits desirable
muscle hypertrophy without detectable off-target effects in
sheep, Sci. Rep. 6 (2016), 32271.
[107] C. Brokowski, M. Pollack, R. Pollack, Cutting eugenics out
of CRISPRCas9, EBEM 6 (2015) 263.
[108] Buck v. Bell, 274 U.S. 2001927.
[109] J.D. Smith, K.R. Nelson, The Sterilization of Carrie Buck,
New Horizon Press, Far Hills, N.J., 1989
[110] P.A. Lombardo, Three Generations, No Imbeciles: Eugenics,
the Supreme Court, and Buck v. Bell, Johns Hopkins
University Press, Baltimore, 2008.
[111] V.M. Gantz, N. Jasinskiene, O. Tatarenkova, A. Fazekas, V.M.
Macias, E. Bier, et al., Highly efficient Cas9-mediated gene
drive for population modification of the malaria vector mosquito
Anopheles stephensi,Proc.Natl.Acad.Sci.U.S.A.112
(2015) E6736E6743.
[112] J. Savulescu, N. Bostrom, Human Enhancement, Oxford
University Press, New York, 2009.
[113] United Nations, Food, [cited 2017 Dec 23]. Available from:
http://www.un.org/sustainabledevelopment/hunger/.
[114] World Health Organization, Nutrition, [cited 2017 Dec 23].
Available from: http://apps.who.int/iris/bitstream/10665/
43412/1/9241594012_eng.pdf?ua=1.
[115] E. Stout, T. Klaenhammer, R. Barrangou, CRISPRCas
technologies and applications in food bacteria, Annu. Rev.
Food Sci. Technol. 8 (2017) 413437.
[116] Y. Demirci, B. Zhang, T. Unver, CRISPR/Cas9: an RNA-
guided highly precise synthetic tool for plant genome
editing, J. Cell. Physiol. 233 (2018) 18441859.
[117] J. Zhang, H. Zhang, J.R. Botella, J.K. Zhu, Generation of
new glutinous rice by CRISPR/Cas9-targeted mutagenesis
of the Waxy gene in elite rice varieties, J. Integr. Plant Biol.
60 (2018) 369375.
[118] S.R. Rajendran, Y.Y. Yau, D. Pandey, A. Kumar, CRISPR
Cas9 based genome engineering: opportunities in agri-
food-nutrition and healthcare, OMICS 19 (2015) 261275.
[119] H. Du, X. Zeng, M. Zhao, X. Cui, Q. Wang, H. Yang, et al.,
Efficient targeted mutagenesis in soybean by TALENs and
CRISPR/Cas9, J. Biotechnol. 217 (2016) 9097.
[120] S. Svitashev, J.K. Young, C. Schwartz, H. Gao, S.C. Falco,
Cigan A.M. Targeted Mutagenesis, Precise gene editing,
and site-specific gene insertion in maize using Cas9 and
guide RNA, Plant Physiol. 169 (2015) 931945.
[121] K. Brinegar, K.Y. A, S. Choi, E. Vallillo, G.U. Ruiz-Esparza,
A.M. Prabhakar, et al., The commercialization of genome-
editing technologies, Crit. Rev. Biotechnol. 37 (2017)
924932.
[122] F. Georges, H. Ray, Genome editing of crops: a renewed
opportunity for food security, GM Crops Food. 8 (2017)
112.
[123] S. Gonen, J. Jenko, G. Gorjanc, A .J. Mileham, C.B.
Whitelaw, J.M. Hickey, Potential of gene drives with
genome editing to increase gene tic gain in l ivestock
breeding programs, Genet. Sel. Evol. 49 (2017) 3.
[124] V.M. Gantz, E. Bier, Genome editing. The mutagenic chain
reaction: a method for converting heterozygous to homo-
zygous mutations, Science 348 (2015) 442444.
[125] A. Hammond, R. Galizi, K. Kyrou, A. Simoni, C. Siniscalchi, D.
Katsanos, et al., A CRISPRCas9 gene drive system
targeting female reproduction in the malaria mosquito vector
Anopheles gambiae, Nat. Biotechnol. 34 (2016) 7883.
[126] National Academies of Sciences Engineering and Medicine
(U.S.), Committee on Gene Drive Research in Non-Human
Organisms: Recommendations for Responsible Conduct.
Gene Drives on the Horizon: Advancing Science, Navigating
Uncertainty, and Aligning Research With Public VALUes, The
National Academies Press, Washington, D.C., 2016
[127] N. Windbichler, M. Menichelli, P.A. Papathanos, S.B.
Thyme, H. Li, U.Y. Ulge, et al., A synthetic homing
endonuclease-based gene drive system in the human
malaria mosquito, Nature 473 (2011) 212215.
[128] P.A. Eckhoff, E.A. Wenger, H.C. Godfray, A. Burt, Impact of
mosquito gene drive on malaria elimination in a computa-
tional model with explicit spatial and temporal dynamics,
Proc. Natl. Acad. Sci. U. S. A. 114 (2017) (E255-E64).
12 Review: CRISPR Ethics
Please cite this article as: C. Brokowski, M. Adli, CRISPR Ethics: Moral Considerations for Applications of a Powerful Tool, J. Mol. Biol.
(2018), https://doi.org/10.1016/j.jmb.2018.05.044
[129] S. James, F.H. Collins, P.A. Welkhoff, C. Emerson, H.C.J.
Godfray, M. Gottlieb, et al., Pathway to deployment of gene
drive mosquitoes as a potential biocontrol tool for elimina-
tion of malaria in sub-Saharan Africa: recommendations of a
scientific working group, Am. J. Trop. Med. Hyg. 98 (2018)
149.
[130] N.L. Achee, F. Gould, T.A. Perkins, R.C. Reiner Jr., A.C.
Morrison, S.A. Ritchie, et al., A critical assessment of vector
control for dengue prevention, PLoS Negl. Trop. Dis. 9
(2015), e0003655.
[131] K.M. Esvelt, A.L. Smidler, F. Catteruccia, G.M. Church,
Concerning RNA-guided gene drives for the alteration of
wild populations, elife 3 (2014).
[132] S.H. Sokolow, C.L. Wood, I.J. Jones, K.D. Lafferty, A.M.
Kuris, M.H. Hsieh, et al., To reduce the global burden of
human schistosomiasis, use old fashioned snail control,
Trends Parasitol. 34 (2018) 2340.
[133] D. Moro, M. Byrne, M. Kennedy, S. Campbell, M. Tizard,
Identifying knowledge gaps for gene drive research to
control invasive animal species: the next CRISPR step,
Glob. Ecol. Conserv. 13 (2018), e00363.
[134] J.E. DiCarlo, A. Chavez, S.L. Dietz, K.M. Esvelt, G.M.
Church, Safeguarding CRISPRCas9 gene drives in yeast,
Nat. Biotechnol. 33 (2015) 12501255.
[135] E. Roggenkamp, R.M. Giersch, M.N. Schrock, E. Turnquist,
M. Halloran, G.C. Finnigan, Tuning CRISPRCas9 gene
drives in Saccharomyces cerevisiae, G3 (Bethesda) 8
(2018) 9991018.
[136] E.M. Basgall, S.C. Goetting, M.E. Goeckel, R.M. Giersch, E.
Roggenkamp, M.N. Schrock, et al., Gene drive inhibition by the
anti-CRISPR proteins AcrIIA2 and AcrIIA4 in Saccharomyces
cerevisiae, Microbiology 164 (2018) 464474.
[137] M.R. Vella, C.E. Gunning, A.L. Lloyd, F. Gould, Evaluating
strategies for reversing CRISPRCas9 gene drives, Sci.
Rep. 7 (2017), 11038.
[138] U.S. CONST. amend. 1791, 1.
[139] E.K. White, Killing U.S. slowly: curing the epidemic rise
of cancer drug prices, Food Drug Law J. 72 (2017)
189224.
[140] L. Cong, F.A. Ran, D. Cox, S. Lin, R. Barretto, N. Habib,
et al., Multiplex genome engineering using CRISPR/Cas
systems, Science 339 (2013) 819823.
[141] S. Chen, N.E. Sanjana, K. Zheng, O. Shalem, K. Lee, X.
Shi, e t al ., Ge nome-wi de CRIS PR scr een in a mouse
model of tumor growth and metastasis, Cell 160 (2015)
12461260.
[142] H. Wang, H. Yang, C.S. Shivalila, M.M. Dawlaty, A.W.
Cheng, F. Zhang, et al., One-step generation of mice
carrying mutations in multiple genes by CRISPR/Cas-
mediated genome engineering, Cell 153 (2013) 910918.
[143] F.A. Ran, L. Cong, W.X. Yan, D.A. Scott, J.S. Gootenberg,
A.J. Kriz, et al., In vivo genome editing using Staphylococ-
cus aureus Cas9, Nature 520 (2015) 186191.
[144] I.M. Slaymaker, L. Gao, B. Zetsche, D.A. Scott, W.X. Yan,
F. Zhang, Rationally engineered Cas9 nucle ases with
improved specificity, Science 351 (2016) 8488.
[145] R.D. Chow, C.D. Guzman, G. Wang, F. Schmidt, M.W.
Youngblood, L. Ye, et al., AAV-mediated direct in vivo
CRISPR screen identifies functional suppressors in glio-
blastoma, Nat. Neurosci. 20 (2017) 13291341.
[146] J. Doudna, A. Mason, Federal Funding for Basic Research
Led to the Gene-Editing Revolution. Don't Cut It, Vox, 2017.
[147] M.A. Moreno-Mateos, J.P. Fernandez, R. Rouet, C.E.
Vejnar, M.A. Lane, E. Mis, et al., CRISPRCpf1 mediates
efficient homology-directed repair and temperature-
controlled genome editing, Nat. Commun. 8 (2017) 2024.
[148] D. Burstein, L.B. Harrington, S.C. Strutt, A.J. Probst, K.
Anantharaman, B.C. Thomas, et al., New CRISPRCas
systems from uncultivated microbes, Nature 542 (2017)
237241.
[149] S.H. Sternberg, B. LaFrance, M. Kaplan, J.A. Doudna,
Conformational control of DNA target cleavage by
CRISPRCas9, Nature 527 (2015) 110113.
[150] K. Schumann, S. Lin, E. Boyer, D.R. Simeonov, M.
Subramani am, R.E. Gate , et al., G enerat ion of kn ock-
in primary human T cells using Cas9 ribonucleopro-
teins,Proc.Natl.Acad.Sci.U.S.A.112(2015)
1043710442.
[151] M.R. O'Connell, B.L. Oakes, S.H. Sternberg, A. East-
Seletsky, M. Kaplan, J.A. Doudna, Programmable RNA
recognition and cleavage by CRISPR/Cas9, Nature 516
(2014) 263266.
[152] J.A. Greene, W.V. Padula, Targeting unconscionab le
prescription-drug pricesMaryland's Anti-Price-Gouging
Law, N. Engl. J. Med. 377 (2017) 101103.
[153] E. Marshall, Gene therapy death prompts review of
adenovirus vector, Science 286 (1999) 22442245.
[154] K.M. Frank, D.K. Hogarth, J.L. Miller, S. Mandal, P.J.
Mease, R.J. Samulski, et al., Investigation of the cause of
death in a gene-therapy trial, N. Engl. J. Med. 361 (2009)
161169.
[155] C.E. Dunbar, K.A. High, J.K. Joung, D.B. Kohn, K. Ozawa,
M. Sadelain, Gene therapy comes of age, Science 359
(2018).
[156] Code of Federal Regulations: Food and Drugs, Institutional
Review Boards, 21 C.F.R. 562017.
[157] Code of Federal Regulations: Protection of Human Subjects,
45 C.F.R. Sect 462017.
[158] Code of Federal Regulations: Food and Drugs, Protection of
Human Subjects, 21 C.F.R. 502017.
[159] Code of Federal Regulations: Food and Drugs, Investiga-
tional New Drug Application, 21 C.F.R. 3122017.
[160] Code of Federal Regulations: Food and Drugs, Investiga-
tional Device, 21 C.F.R. 8122017.
[161] NIH to launch genome editing research program, https://www.
nih.gov/news-events/news-releases/nih-launch-genome-
editing-research-program.
[162] Commission on Genet ic Modifi cat ion and the Healt h
Council of the Netherlands, Editing human DNA: moral
and social implications of germline genetic modification,
2017, Accessed 26 May 2018.
[163] The Academy of Medical Sciences' response to the Nuffield
Council on Bioethics Genome editing and human reproduc-
tion', Acad. Med. Sci. (2017) [[press release] [cited 2018 May
26]. Available from: https://acmedsci.ac.uk/file-download/
89659337].
[164] Document on bioethics and gene editing in humans, [cited
2018 May 26]. Available from: Bioethics and Law Observa-
tory of the University of Barcelona, http://www.publicacions.
ub.edu/refs/observatoriBioEticaDret/documents/0854 3.pdf
2016.
[165] G. Church , Compelling reasons for repairing human
germlines, N. Engl. J. Med. 377 (2017) 19091911.
[166] Geneva Convention, Protocol for the prohibition of the use
in war of asphyxiating, poisonous or other gases, and of
bacteriological methods of warfare, http://avalon.law.yale.
edu/20th_century/geneva01.asp 1925, Accessed date: 20
December 2017.
13Review: CRISPR Ethics
Please cite this article as: C. Brokowski, M. Adli, CRISPR Ethics: Moral Considerations for Applications of a Powerful Tool, J. Mol. Biol.
(2018), https://doi.org/10.1016/j.jmb.2018.05.044
[167] United Nations, Convention on the prohibition of the
development, production and stockpiling of bacteriological
(biological) and toxin weapons and on their destruction,
http://avalon.law.yale.edu/20th_century/bact.asp 1975,
Accessed date: 20 December 2017.
[168] M. Enserink, Interested in responsible genome editing?
Join the new club, Science (2018), https://doi.org/10.1126/
science.aat7183.
[169] M. Lluis, M. Jennifer, H. François, A. Marion, J. Pierre, B.
Bernard, S.M. Cyril, C. Hervé, ARRIGE arrives: toward the
responsible use of genome editing, CRISPR J. 1 (2) (2018)
128129.
[170] S. Jasanoff, J.B. Hurlbut, A global observatory for gene
editing, Nature 555 (2018) 435437.
14 Review: CRISPR Ethics
Please cite this article as: C. Brokowski, M. Adli, CRISPR Ethics: Moral Considerations for Applications of a Powerful Tool, J. Mol. Biol.
(2018), https://doi.org/10.1016/j.jmb.2018.05.044