Annotated bibliography (20 hours)

We are confident that we have the best essaywriters in the market. We have a team of experienced writers who are familiar with all types of essays, and we are always willing to help you with any questions or problems you might face. Plus, our writers are always available online so you can always get the help you need no matter where you are in the world.


Order a Similar Paper Order a Different Paper

 

APA format

Save your time - order a paper!

Get your paper written from scratch within the tight deadline. Our service is a reliable solution to all your troubles. Place an order on any task and we will take care of it. You won’t have to worry about the quality and deadlines

Order Paper Now

1) Minimum 4 pages  (No word count per page)- Follow the 3 x 3 rule: minimum of three paragraphs per page

You must strictly comply with the number of paragraphs requested per page

.

2)¨******APA norms

         All paragraphs must be narrative and cited in the text- each paragraph

         The writing must be coherent, using connectors or conjunctive to extend, add information, or contrast information. 

         Bulleted responses are not accepted

         Don’t write in the first person 

         Don’t copy and paste the questions.

         Answer the question objectively, do not make introductions to your answers, answer it when you start the paragraph

3)****************************** It will be verified by Turnitin (Identify the percentage of exact match of writing with any other resource on the internet and academic sources, including universities and data banks) 

********************************It will be verified by SafeAssign (Identify the percentage of similarity of writing with any other resource on the internet and academic sources, including universities and data banks)

4) References (APA format)  should only refer to the  8 attached articles

5) Identify your answer with the numbers, according to the question. Start your answer on the same line, not the next

Example:

Q 1. Nursing is XXXXX

Q 2. Health is XXXX

__________________________________________________________________________________

Question: Should gene-editing be abolished?

Position: Agree

Topic: Gene-editing  should be abolished

Annotated bibliography (APA format)

Use only the eight (8) attached documents to make an annotated bibliography

Keep in mind that articles 2, 3, 4, 5, 6, 7 and 8 are against human gene editing; therefore, when reviewing the articles, you should focus on the disadvantages, risks or topic problems.

Only article 1 should reflect the perspective in favor of human gene editing

1. Half page for each article (Two articles per page)

2. Answer the following questions on a line at the end of the analysis of each article

to. What is the source about?

b. How and where do you plan to use the information in the research pap3r?

c. How do you know if the source is credible?

i. Who is the author, publisher, or what database is it from?

Centre for Biomedicine, Self and Society; Usher Institute; University of Edinburgh, Old Medical
School, Teviot Place, Edinburgh, EH8 9AG, United Kingdom.

Email: [email protected]

Perspectives in Biology and Medicine, volume 63, number 1 (winter 2020): 111–125.
© 2020 by Johns Hopkins University Press

111

Playing it Safe?

precaution, risk, and responsibility
in human genome editing

Sarah Chan

ABSTRACT Human germline genetic modification has long been a controversial
topic. Until recently it remained largely a hypothetical debate: whether one accepted
or opposed the idea in principle, it was not only too risky but impractical to execute
in reality. With the advent of genome editing technologies, however, heritable modi-
fications to the human genome became a much more concrete possibility; nonetheless,
the consensus has to date remained that human heritable genome editing is not yet safe
enough for clinical application. The announcement of the birth of two genome-ed-
ited babies in late 2018, therefore, was condemned almost universally as premature,
irresponsible, and dangerous. But what does responsibility require, and from whom?
How should risk and precaution be balanced in assessing heritable genome editing, and
against what alternatives? This paper reexamines commonly held assumptions about
risk and responsibility with respect to human genome editing and argues that the pre-
cautionary approach that has so far been favored is not well justified, that the risks of
heritable versus somatic genome editing should be reassessed, and that a fuller account
of responsibility—scientific, social, and global—is required for the ethical governance
of genome editing.

On november 26, 2018, the world awoke to the news that genome edit-ing had for the first time been used to create genetically modified human

Sarah Chan

112 Perspectives in Biology and Medicine

beings. He Jiankui, a scientist then employed by Southern University of Science
and Technology of China, Shenzhen, announced via social media and the pop-
ular press that he had performed genome editing on embryos with the aim of
disrupting the CCR5 gene in order to induce immunity to HIV, implanted the
embryos, and that twin girls had been born.

In the wake of this announcement, arguments, claims, and accusations flew.
Most commentators declared He’s actions “irresponsible,” pointing to various
statements by scientific organizations agreeing that human embryo genome edit-
ing was not yet at an appropriate stage for clinical use. Another common criticism
was the lack of transparency surrounding the work, and that He failed to follow
appropriate governance procedures with respect to consent and ethical review.
Many also condemned the experiment on the grounds that the babies had been
exposed to unjustified risk relative to the possible benefits.

This case, then, was almost universally regarded as an unfortunate occurrence.
Nonetheless, in exposing some potential fault lines in our thinking about human
genome editing with respect to risk, harm, and responsibility, it provides a timely
opportunity to reexamine these concepts. He’s actions may have been a failure
of responsibility—but responsibility to whom and for what? What does it mean
to “be responsible,” “act responsibly,” or “take responsibility” with respect to
human genome editing? In addressing these questions, I will argue that in dealing
with human genome editing, the following elements are required: an approach
to risk and responsibility that goes beyond precaution; a more robust conceptu-
alization of scientific responsibility and what it requires; a reconsideration of the
nature and distribution of harms and benefits of genome editing; and attention
to collective social responsibility and its global dimensions with respect to the
development and governance of science.

Precaution and Responsibility in Genome Editing

The generally accepted approach to human heritable genome editing (HGE)
so far has been cautious, if not outright precautionary. A recent review of over
60 statements on the subject found that most “were expressly against heritable
genome editing at the current time” (Brokowski 2018), with both the risk of
known possible harms and uncertainty and the possibility of unforeseen adverse
effects being prominent concerns. Even those that were willing to entertain the
possibility of HGE in principle, notable among them the US National Academies
(2017) and the UK’s Nuffield Council on Bioethics (2018), agreed that it should
not be permitted in practice until issues of risk and safety had been adequately
addressed, and that this was not yet the case. In light of these concerns, it seems
justified to label He’s actions as premature and irresponsible. But what level of
caution in relation to genome editing is appropriate, and is a precautionary ap-
proach warranted?

Playing it Safe?

113winter 2020 • volume 63, number 1

Early formulations of the precautionary principle emerged in relation to envi-
ronmental policy. As originally articulated, it was intended to justify taking action
to forestall harm: “Where there are threats of serious or irreversible damage, lack
of scientific certainty shall not be used as a reason for postponing cost-effective
measures to prevent environmental degradation” (UN Conference on Environ-
ment and Development 1992). Subsequent iterations shifted the focus of the
principle to a presumption against doing, by placing the burden of proof on the
would-be doer to demonstrate safety (SEHN1998). This form of the principle, in
which precaution is the opposite of pro-action, is the most common characteriza-
tion in bioethical debate (Glannon 2002; Harris and Holm 2002). In this context,
it also tends to incorporate assumptions about the relative merits of the status quo
or natural, versus a human-engineered state of affairs, “a sense of nature’s ulti-
macy that precedes or supersedes human ingenuity” (Fuller and Lipinska 2014).

The Nuffield Council’s report on human heritable genome editing, in an in-
teresting reversal, entertains a more pro-actionary version of the principle itself.
Considering possible harms that might be averted by genome editing in a situ-
ation where looming environmental catastrophe threatens human survival, they
write that “at minimum [the precautionary principle] permits taking action in
the present even in the absence of clear evidence of the likelihood of a harm that
might occur in the future” (Nuffield Council 2018, 90). Nevertheless, the main
approach to managing risks and uncertainties of heritable human genome editing
can best be described as either that we ought not to proceed until we can be fairly
sure that nothing bad will happen; or that, because we cannot be sure of this, we
ought not to proceed at all, notwithstanding the Nuffield Council’s exception for
a threat to human survival—where, in a sense, all bets are off.

Adopting this sort of precautionary attitude to human genome editing com-
bines (at least) two intertwined claims about responsibility. One is quasi-empiri-
cal, about where the balance of risk lies and what would therefore be a responsible
course of action to take; the other is a moral claim about what our responsibility
would be for the consequences of our choices in various circumstances.

To explain further, the empirical balance-of-risk claim concerns how likely
it is that a change deliberately engineered by humans will produce unintended
deleterious side effects. Existing genetic variation at least has history on its side,
having survived thus far; what therefore are the chances that any change we
make is on-balance more likely to disrupt this and produce negative side-effects?
Advocates of a precautionary approach favor the idea that we ought to give
nature the benefit of the doubt (President’s Council 2003), a claim sometimes
expressed in terms of the “wisdom of nature” (Bostrom and Sandberg 2009).
Powell and Buchanan (2011), in critiquing this claim, call it the Master Engineer
Analogy: the idea that “from an evolutionary perspective, the human organism
is like the product of an engineering genius—a delicately balanced, completed,
well-functioning masterwork” and that therefore attempts to interfere with this,

Sarah Chan

114 Perspectives in Biology and Medicine

even if well-intentioned and scientifically-grounded, are likely to be “disastrously
counterproductive” (Powell and Buchanan 2011, 7). This analogy, they argue,
relies on misconceived interpretations of evolutionary theory: we should resist
the presumption that intentional genetic modification is likely to be more dan-
gerous than “unintentional genetic modification”—that is, the process of muta-
tion, inheritance, and selection that occurs whether we like it or not. It may be
that random genetic changes are more likely to be deleterious than beneficial, but
this is not necessarily true of targeted modifications designed with some under-
standing of their likely effects.

Moreover, judgments about risk are not absolute but comparative: whether a
likely consequence is better than or worse than the alternative. Precaution as a
presumption against action assumes that the consequences of doing are probably
going to be worse than not-doing. In the case of genome editing for serious dis-
ease, it is far from evident that this is so.

To Do or Not to Do?

The moral responsibility claim concerns blameworthiness for consequences: spe-
cifically, whether more blame should attach to those consequences we cause via
our action, than those we allow via inaction. Here the precautionary approach
is often expressed in terms of “letting nature take its course.” By analogy with
the “wisdom of nature,” we might call this a “responsibility of nature” claim:
that if something happens as a result of nature, we are not responsible for having
allowed it.

Evaluating risk requires us to consider both how severe a harm would be and
how likely it is to occur. A persistent issue with heritable genome editing is that
we cannot necessarily foresee all possible harms in order to assess their severity
nor their likelihood. This is not, however, by any means a problem unique to
genome editing. We also cannot predict all the possible consequences of mobile
phone use or non-use; of fracking or not-fracking; or of failing to stop climate
change. There will be consequences either way, of a decision to use or not to
use technology; why then should we focus in the case of genome editing on the
possible consequences of doing, more than of not-doing?

The precautionary approach to human genome editing seems to presume that
it would be worse for a harm to occur as the result of our deliberate action,
than for a similar-magnitude harm to occur as the consequences of a deliberate
omission. This, however, seems to conflate causal and moral responsibility. Are
we failing to do what would be most responsible, in order to avoid being held
responsible—that is, held to blame—in the event that something goes wrong?

In moral philosophy, the distinction between acts and omissions has been most
addressed in relation to the ethics of killing versus letting die. Rachels’s (1975)
thought experiment of the evil uncles (one of whom kills his nephew in the

Playing it Safe?

115winter 2020 • volume 63, number 1

bath, while the other “merely” but deliberately allows him to drown) attempts to
demonstrate that the same harm brought about by action and by deliberate omis-
sion is morally equivalent. Genome editing, however, is not quite analogous: this
is a case where in acting to attempt to prevent one foreseeable harm, we incur a
risk of causing a different harm. How then should we weigh our responsibility for
allowing versus causing harm in each instance?

Perhaps a more appropriate analogy is a Good Samaritan case, in which one is
presented with an opportunity to render aid to the needy. In such cases, a per-
son who is “not responsible” for the situation, such as a passer-by, is not usually
legally liable for failing to help. A person who “takes responsibility” by choosing
to attempt to help, however, might subsequently be “held responsible” if their
efforts to help somehow go wrong, providing they are sufficiently careless in the
attempt.

The law in general attaches more responsibility to actions than omissions,
a distinction that has led to some convoluted legal reasoning particularly with
respect to end-of-life decisions (Airedale NHS Trust v Bland (1993) 1 All ER
821). Imposing a precautionary, rather than a pro-actionary, approach to human
genome editing, however, would seem to broaden this rather narrow legal ap-
proach to acts versus omissions much more widely in order to draw the same
conclusion about moral responsibility—namely that we should be considered
more blameworthy for harms we cause (even unintentionally) than for those we
deliberately allow.

The acts/omissions distinction certainly has legal relevance, but should it have
moral relevance? Moral philosophy is quicker than law to recognize obligations
of beneficence, such as a “duty of rescue,” where accomplishing the rescue
would not cause undue or disproportionate burden to the rescuer. Moreover,
even within a legalistic frame, consider that in the Good Samaritan example, a
genuine attempt to assist by acting in good faith, with the reasonable belief that
one’s actions would help rather than harm, is unlikely to result in either legal or
moral blame if the attempt miscarries. Our evaluation of what responsible parents
and scientists should do with respect to genome editing, and what they ought to
be held responsible (morally blameworthy) for, ought also to take into account
similar criteria of reasonable belief and intention.

Furthermore, parents are not bystanders: they do have special moral responsi-
bility for their children’s welfare. Given this, a presumption against action seems
unwarranted. As the Nuffield Council (2018) report puts it, “we have to take
responsibility for both acts and conscious omissions: deciding not to use an avail-
able technology and whether to discover knowledge about the genome or to
intervene in it may still count as choices that engage moral responsibility” (73).

Sarah Chan

116 Perspectives in Biology and Medicine

Who Is Responsible?

To summarize so far, responsibility with respect to human genome editing en-
compasses both what it would be responsible to do, and for what consequences
we should be held responsible. These two senses are entwined, in that (1) if we
fail to behave responsibly, it is reasonable to think that (2) we ought to be held
responsible for that failure. But there is one further element needed to connect
these points: namely, (3) our having responsibility (or taking it, or being respon-
sible) for doing the right thing in the first place.

For example, imagine a teenager charged with looking after his younger sib-
lings for the evening; instead of watching them, he goes upstairs to read a book,
falls asleep, and they set the house on fire. Clearly, he has been irresponsible and
should be held to account for any harms that result. (Note also that this is respon-
sibility for an omission, failing to act.) Alternatively, now imagine that his mother
comes home earlier than expected and tells him, “Go upstairs and read; I’ll watch
the kids.” If in this situation he falls asleep reading and, his mother failing to pay
attention, the children burn the house down, it seems that he should not be held
responsible.

One might argue that, despite having been “let off the hook,” it would none-
theless “be responsible” for the teenager to take additional responsibility for en-
suring all is well with his family and to check on them. All this shows, however,
is that transferring responsibility is not necessarily simply a matter of two parties
agreeing between themselves that one will be responsible and the other not; ex-
traneous factors may still lead us to conclude that the would-be transferor retains
some responsibility. Unilateral assumptions and assignations of responsibility also
require cautious evaluation: simply declaring myself to be responsible does not
necessarily make me so, nor can I render someone else responsible just by saying
so.

In the case of human genome editing, then, who is responsible, who takes
responsibility, and who should be held accountable? It is now generally accepted
that scientists bear some degree of responsibility for their work, collectively as
well as individually: that is, being a scientist in itself imposes certain responsibility
(Ehni 2008; Jonas 1984; Verhoog 1981). We should also, though, recognize the
significance of responsibility-taking as a form of asserting authority. Calling He’s
work a “failure of self-regulation” (Cyranoski 2018) not only takes responsibility
on behalf of scientists, it also stakes an implicit claim to governance. This claim,
and the associated questions of how scientific responsibility is distributed and
how it is to be discharged, demand more critical scrutiny.

What Does Scientific Responsibility Require?

Scientific responsibility in human genome editing is not just a matter of indi-
vidual case-by-case risk management: calling something “irresponsible” is not

Playing it Safe?

117winter 2020 • volume 63, number 1

identical with saying it is “too dangerous.” The statement issued after the first
International Summit on Human Gene Editing in 2015 declared that:

It would be irresponsible to proceed with any clinical use of germline editing
unless and until (i) the relevant safety and efficacy issues have been resolved,
based on appropriate understanding and balancing of risks, potential benefits,
and alternatives, and (ii) there is broad societal consensus about the appropriate-
ness of the proposed application. (NAS 2017)

The implication is that conducting heritable human genome editing at the
present time would be irresponsible because it is currently too dangerous, but
that with proper investigation it might become less so. Some have suggested that
responsibility may even require us to undertake such investigation: Lovell-Badge
(2019), for example, comments that “it would actually be irresponsible for us
not to explore this opportunity further.” This is partly because of the medical
need that genome editing might help to address, an argument that parallels bio-
ethical claims about moral obligations to pursue gene therapy and enhancement.
Additionally, however, scientific responsibility requires complying with norms
of scientific knowledge production and communication, including “high-quali-
ty experimental design,” “appropriate review,” and “transparency” (NAS 2017,
24). This includes appropriate standards and procedures at a community-wide
level, for which a thorough understanding of the relevant science is necessary.

Assigning and judging scientists’ responsibility to avoid bad behavior is rel-
atively simple. It is clear that in carrying out heritable human genome editing,
against almost all scientific opinion and without any chance for public discussion,
clandestinely, and without attention to appropriate standards of clinical research
design or review, He breached scientific responsibility on multiple counts. Less
easy, however, is determining what positive duties scientific responsibility might
impose, and on whom. Who else might bear some responsibility in this case?
Although He’s name is most prominently associated with the work, he did not
act alone: fertility specialists must have been involved in the procedures required.
Further, from reports that have since emerged, it seems other academics in the
US may have been directly involved, while an additional number knew or sus-
pected what was afoot. This appears to be more than just an individual failure on
He’s part.

The more difficult problem, however, is what these other scientists could or
should have done in response. If peer review and ethical oversight are the ex-
pected routes via which scientific work is subjected to examination, what should
scientists do about work that avoids or evades these pathways? Are scientists ex-
pected to be aware of regulatory requirements across all national contexts and
to act as whistleblowers to promote enforcement? How far does the positive
obligation to discourage or prevent the malfeasance of other scientists extend:
might there be a collective responsibility to develop community standards and

Sarah Chan

118 Perspectives in Biology and Medicine

mechanisms that would have enabled those in the know more effectively to act?
More work is required to turn the claims about irresponsibility that have attached
to He’s case into a robust framework for scientific responsibility in human ge-
nome editing. Does knowing that a scientist is abrogating agreed upon standards
of research and behavior make those scientists in the know culpable?

Relative Risks and Sociocultural Harms

A focal criticism of He’s experiment was that the procedure was largely unnec-
essary and of marginal benefit, given that HIV infection can easily be avoided in
other ways and can be well managed with current treatments, and that therefore
exposing the babies to the risks of heritable genome editing was unjustified.

He’s argument was that with an HIV-positive parent, the children were at
greater than normal risk of infection; that within the Chinese health system, ac-
cess to top-line antiretrovirals is not necessarily assured; and that furthermore, in
China, those who are HIV-positive face high levels of stigma and social discrimi-
nation. In a context where HIV is severely stigmatized and has drastic social, not
only health, implications, might the culturally relative risk be higher and there-
fore change the risk-benefit ratio of the treatment?

Risks can be differently materially relative—that is, the same event can have
different consequences in light of different material circumstances. For example,
suffering a simple cut to one’s finger at home is fairly insignificant and unlikely to
cause much of a problem; the same wound suffered in the jungle, without access
to antibiotics and several days’ journey from medical assistance, might result in
infection, loss of a finger or limb, or even death. One would be justified in taking
proportionately greater precautions to avoid the latter than the former. Insofar as
cultural context makes up part of the material circumstances in which our deci-
sions are taken, it may well be the case that a particular characteristic represents
more of a harm in one social context than in another. In such circumstances it
seems sensible that the level of risk that can justifiably be incurred in trying to
avoid it would likewise vary.

One response might be that the appropriate solution to the social disadvantage
of being HIV-positive is to seek to reduce discrimination, rather than to geneti-
cally engineer immunity. Disability advocates and scholars have long argued for
this “social model” with respect to people with disabilities or genetic conditions
such as Down syndrome (Shakespeare 2013): that the “harm” of such conditions
is at least in part socially constructed, and that it is society that ought to change
rather than imposing a technological solution. Nonetheless, it is commonly seen
as justifiable to expose embryos in vitro or fetuses in utero to procedures such as
pre-implantation genetic testing (PGT) and prenatal screening in order to avoid
such conditions.

Playing it Safe?

119winter 2020 • volume 63, number 1

Comparing responses to He’s work with the practices and attitudes surround-
ing the use of reproductive technologies we already commonly accept, if we are
to apply the “social model” to judge one, we should apply it in equal measure
to the other. In both cases, we ought to give more consideration to the ways in
which welfare is, as the Nuffield Council report recognizes, “to an extent, so-
cially and historically determined.” Importantly, both for this case and as genome
editing develops globally, we should also acknowledge the socioculturally relative
ways in which our attitudes towards different uses of technology are conditioned,
and how this might differentially affect assessments of justifiable risk in each case.

Harm and Benefit to Whom?

A wrinkle in assessing the harms and benefits of HGE is that, at least as we
presently envisage its application, it would need to be performed in advance of
any genetic testing of the embryo, at the single-cell stage or even at the point
of fertilization. If using it to increase the chances of obtaining a suitable embryo
for transfer where some embryos might already have the desired genotype, as has
been one proposed application, this would mean some embryos would have been
exposed unnecessarily to the procedure—but we would not know which.

This is in some ways comparable to the existing distribution of risk in PGT:
the embryos that are eventually transferred are subjected to blastomere biopsy
only to prove that they are healthy. Insofar as this procedure might entail some
risk of harm, had they been implanted without testing they might have been
better off. For potential children produced from both PGT and genome editing,
however, the application of the technique should be regarded as a precondi-
tion of their existence—assuming that parents would otherwise not reproduce or
(more likely) would choose to reproduce in another way, meaning the creation
of different embryos resulting in different future persons.

In the case of the genome-edited twins, their parents might still have chosen
assisted reproduction in order to enable the sperm-washing procedure to prevent
HIV-transmission, but differences in the timing and circumstances would surely
have resulted in a different combination of egg and sperm, and thus a differ-
ent child or children being born. And, although He’s decision (on his account,
in accordance with the parents’ wishes) to transfer one of the embryos despite
knowing that the genome editing had not worked as intended has been criticized
for incurring the risk of HGE with no justificatory benefit, the alternative for that
child would have been nonexistence.

The task of assigning harm or benefit to children who might result from
potential genome editing procedures thus encounters the knotty philosophical
problem of identity, or rather nonidentity (Parfit 1984): can we say a child has
been harmed by being subjected to genome editing when the alternative, had
their parents not chosen to use this process, is that a different child altogether
would have been born?

Sarah Chan

120 Perspectives in Biology and Medicine

Identity issues raise various difficulties for reproductive law. The regulation of
PGT and its acceptable uses has been criticized for inconsistency in this regard
(Sheldon and Wilkinson 2004); indeed, the entire category of “wrongful life”
cases is problematic in comparing the harms of existence to nonexistence. Such
abstract philosophical considerations are also rather far removed from parents
making decisions about their future families. For example, in the debate over
mitochondrial replacement therapy (MRT), philosophers were quick to pounce
on and dissect the question of whether MRT affects numerical identity and how
this might influence assessments of harm (see, for example, Cavaliere and Pala-
cios-Gonzalez 2018; Liao 2017; Palacios-Gonzalez 2017; Rulli 2017; Wrigley,
Wilkinson, and Appleby 2015). Prospective parents and would-be users’ views,
however, centered much more on concern for the health of future children and
how the parent-child relationship might be negotiated via genetic and social fac-
tors (Nuffield Council 2012).

What this means for genome editing is that solving the nonidentity problem
is not our biggest concern. We should not be so caught up in metaphysical co-
nundrums regarding future persons that we fail to cash out harms and benefits
to current persons. In fact, HGE is also for the benefit of prospective parents, in
enabling them to fulfil significant procreative interests (Nuffield Council 2018).
Likewise, although clearly it is to be hoped that the genome-edited twins are
healthy and unharmed, we should consider who else might be harmed by He’s
actions, or by others who might be inclined to attempt a similar exercise. Nota-
bly, if premature, reckless or “irresponsible” (in any sense of the word) application
of genome editing leads to a delay in development of valuable therapies, those
patients and their families who miss out on beneficial treatment will be harmed.
We should therefore consider the possible dangers of an over-hasty approach, not
only for eager patients who may be willing to try experimental genome editing
procedures no matter how risky, but also for those who await benefit from ge-
nome editing developed in a responsible and acceptable way.

Intergenerational Risk and the
Somatic/Germline Distinction

It is usually taken for granted that somatic genome editing is “less risky,” and
that this justifies pursuing it in preference to germline editing. This assumption,
however, warrants further questioning. The risks and harms involved in germline
editing may be of a different nature and differently distributed, but it is not neces-
sarily straightforwardly the case that assessment of risks and possible harms should
lead us to favor somatic genome editing.

For example, somatic gene therapy for inherited anemias involves bone mar-
row harvesting (a nontrivial procedure), in vitro manipulation, and re-transplan-
tation (Dever and Porteus 2017). Each stage has its own attendant risks; there is

Playing it Safe?

121winter 2020 • volume 63, number 1

uncertainty regarding whether it will succeed overall or have some unintended
adverse consequence; and during the whole procedure the patient suffers the ef-
fects of the disease. The latter can be significant: a recent case of somatic therapy,
for instance, involved a young boy with junctional epidermolysis bullosa, a severe
skin adhesion disorder (Hirsch et al. 2017). The treatment was a success for the
patient and as an example of cell and gene therapy, but had it been possible to
cure the disease at an embryonic stage instead, this would have saved the patient
from enduring several years of pain and suffering, including losing 80% of the
skin from his body before being treated.

Additionally, if the recipients of somatic genome editing wish to have genet-
ically related children and avoid passing the disease on, they must resort to IVF
and PGT, procedures which carry their own risks and burdens and are not always
successful. These procedures are also currently not available for carrier status, at
least in the UK; thus, a patient with a recessive genetic condition who has been
cured by somatic genome editing would, unless their reproductive partner also
has the condition or is a carrier, not necessarily have access to PGT to remove the
mutation from their line of descent.

In other words, to continually prefer somatic to germline intervention is to
say that each manifestation of disease in subsequent generations should be treated
with somatic therapy, which in itself entails a not-inconsequential degree of risk
and suffering. So even if a single instance of germline editing carries greater risk
in comparison to a single instance of somatic editing, a decision over which to
pursue must take into account the cumulative risks and harms of the multiple
instances of somatic editing that are being traded off.

A further consideration is that the risk of germline editing falls primarily
on one individual, while the risk of repeated somatic procedures is distributed
amongst many, including persons who do not yet exist. We might of course say
that in the case of germline editing, not only the “editee” but their potential fu-
ture descendants are exposed to risk. This depends, however, on the reproductive
choices made by the editee, choices that are likely to be strongly influenced by
whether the genome editing procedure has worked as intended, or whether some
unintended adverse consequence has occurred.

Nevertheless, it seems that there may be a relevant difference between a single
risk-taking event followed by decisions that can be made knowing what the con-
sequences in fact were, versus multiple sequential risk-taking events each involv-
ing possible negative consequences and influencing subsequent decisions. What
this demonstrates is that we need a more thorough ethical, as well as scientific,
analysis of risk in both somatic and germline genome editing, in order properly
to be able to make decisions about when to pursue each possibility.

Sarah Chan

122 Perspectives in Biology and Medicine

Responsibilities to Global Society

Thus far we have focused mainly on the responsibilities that scientists, scientific
communities, and parents have with respect to genome editing at the individual
level. What might collective social responsibility with respect to genome editing
require?

In a wider social context, we might also have obligations regarding how we
use (or refrain from using), in the broadest sense of the word, human genome
editing. The Nuffield Council suggests that heritable genome editing should be
“permitted only in circumstances in which it cannot reasonably be expected to
produce or exacerbate social division or the unmitigated marginalization or dis-
advantage of groups within society.” We might take this a step further: if we can
envisage a way in which this technology might be used that would give us a rea-
sonable expectation of reducing social division, marginalization, or disadvantage,
these circumstances might provide a reason not only to permit HGE, but a posi-
tive reason to use it. Additionally, though, we ought not to assume that it is only
the direct application of genome editing itself that will lead to marginalization, or
that only genome-edited humans will either be its victims or agents.

What is usually envisaged when equity concerns over genetic modification are
raised is the division of society into GeneRich and GenePoor: that those who
are already sufficiently advantaged to afford genetic technologies will become
even better off as a result, widening existing inequities. Extreme versions of this
concern envision this resulting in the fragmentation of humanity into different
species, who will then inevitably turn on each other (Annas, Andrews, and Isasi
2002). This argument ignores, however, that as history unfortunately shows, hu-
mans are quite capable of creating social divisions liable to end in genocide even
without the help of genetic modification. Avoiding human genome editing only
solves part of this problem, and not the most immediate part: the “fix” here must
be social, not anti-technological. Indeed, if the spectre of genome-edited spe-
ciation prompts us to develop better moral and political frameworks in order to
avoid future posthuman genocide, this might contribute to reducing conflict and
promoting more equitable treatment of all human beings today—which would
certainly be a good thing!

In any case, however, there are at least two other, probably more immediate
ways in which genome editing might lead indirectly to social division and mar-
ginalization. The first is via the geneticization of discourse: even before genome
editing comes into widespread use, the way in which we discuss its potential and
our moral obligations with respect to it might promote “geneticized” thinking,
which in turn can lead to discrimination. Overbroad versions of the “moral im-
perative” argument, which posit genome editing as the remedy for all manner of
complex biosocial problems such as health-care resource allocation and inequali-
ties in socioeconomic status and educational outcomes (Gyngell, Bowman-Smart,
and Savulescu 2019), may pose a particular danger here: it is not a very great leap

Playing it Safe?

123winter 2020 • volume 63, number 1

from urging that genes are the solution we have a moral obligation to pursue, to
concluding that genes are the cause of the problem. Given how far we are from
the promissory future in which genome editing does solve such problems (if in-
deed it ever can), this is just as likely to result in pseudo-genetic discrimination.

Second, genes aside and on a global scale, we are already ScienceRich and
SciencePoor. Vast disparities exist between countries in terms of scientific capi-
tal: those countries with more advanced scientific capacity also tend to dominate
ethical and regulatory discourse and wield greater influence in determining the
global norms of science. Looking at the recent history of human genome edit-
ing as well as other emerging technologies, there is a very real possibility that
the trajectory through which human genome editing is realized, including the
discourses surrounding and shaping its realization, will increase inequity between
countries and worsen divisions amongst global scientific communities (Chan,
Palacios-Gonzalez, and De Jesus Medina Arellano 2017). Even before He’s an-
nouncement, China’s human genome editing endeavors were already disadvan-
taged by stereotypical perceptions of a “Wild East” deficient in ethics, regulation,
and scientific norms (Ho 2016; Sipp and Pei 2016). Regardless of whether “ob-
jectively” justified or not, such perceptions are also a product of existing scientific
capital; reiterating them, whether as justification or critique, further reinforces
inequalities. We must attend to the effects of this in considering how we move
forward with, and shape discourse around, human genome editing and its gov-
ernance at a global level. Discussions of genome editing must therefore move
us to new modes of global ethical discourse and scientific governance, not only
because of the technology’s potential to cross borders, but because of its role as a
paradigmatic example of emerging, ethically contested science.

References

Annas, G. J., L. B. Andrews, and R. M. Isasi. 2002. “Protecting the Endangered Human:
Toward an International Treaty Prohibiting Cloning and Inheritable Alterations.” Am
J Law Med 28 (2–3): 151–78.

Bostrom, N., and A. Sandberg. 2009. “The Wisdom of Nature: An Evolutionary Heu-
ristic for Human Enhancement.” In Human Enhancement, ed. J. Savulescu and N.
Bostrom, 375–416. Oxford: Oxford University Press.

Brokowski, C. 2018. “Do CRISPR Germline Ethics Statements Cut It?” CRISPR J 1
(2). DOI: 10.1089/crispr.2017.0024.

Cavaliere, G., and C. Palacios-Gonzalez. 2018. “Lesbian Motherhood and Mitochondrial
Replacement Techniques: Reproductive Freedom and Genetic Kinship.” J Med Ethics
44 (12): 835–42. DOI: 10.1136/medethics-2017-104450.

Chan, S., C. Palacios-Gonzalez, and M. De Jesus Medina Arellano. 2017. “Mitochondrial
Replacement Techniques, Scientific Tourism, and the Global Politics of Science.”
Hastings Cent Rep 47 (5): 7–9. DOI: 10.1002/hast.763.

Cyranoski, D. 2018. “CRISPR-Baby Scientist Fails to Satisfy Critics.” Nature 564: 13–14.

Sarah Chan

124 Perspectives in Biology and Medicine

Dever, D. P., and M. H. Porteus. 2017. “The Changing Landscape of Gene Editing in
Hematopoietic Stem Cells: A Step Towards Cas9 Clinical Translation.” Curr Opin
Hematol 24 (6): 481–88. DOI: 10.1097/MOH.0000000000000385.

Ehni, H. J. 2008. “Dual Use and the Ethical Responsibility of Scientists.” Arch Immunol
Ther Exp (Warsz) 56 (3): 147–52. DOI: 10.1007/s00005-008-0020-7.

Fuller, S., and V.Lipinska. 2014. The Proactionary Imperative: A Foundation for Transhuman-
ism. London: Palgrave Macmillan.

Glannon, W. 2002. “Extending the Human Life Span.” J Med Philos 27 (3): 339–54.
DOI: 10.1076/jmep.27.3.339.2978.

Gyngell, C., H. Bowman-Smart, and J. Savulescu. 2019. “Moral Reasons to Edit the Hu-
man Genome: Picking Up from the Nuffield Report.” J Med Ethics. DOI: 10.1136/
medethics-2018-105084.

Harris, J., and S. Holm. 2002. “Extending Human Lifespan and the Precautionary Para-
dox.” J Med Philos 27 (3): 355–68.

Hirsch, T., et al. 2017. “Regeneration of the Entire Human Epidermis Using Transgenic
Stem Cells.” Nature 551 (7680): 327–32. DOI: 10.1038/nature24487.

Ho, C. W.-L. 2016. “CRISPR Gene-Editing Controversy Shows Old Ideas About East
And West Still Prevail.” The Conversation. http://theconversation.com/crispr-gene-
editing-controversy-shows-old-ideas-about-east-and-west-still-prevail-66918.

Jonas, H. 1984. The Imperative of Responsibility. Chicago: University of Chicago Press.
Liao, S. M. 2017. “Do Mitochondrial Replacement Techniques Affect Qualitative or

Numerical Identity?” Bioethics 31 (1): 20–26. DOI: 10.1111/bioe.12308.
Lovell-Badge, R. 2019. “CRISPR Babies: A View from the Centre of the Storm.” De-

velopment 146 (3). DOI: 10.1242/dev.175778.
National Academies of Sciences, Engineering and Medicine (NAS). 2015. On Human

Gene Editing: International Summit Statement. Washington, DC: National Academy of
Sciences, Engineering and Medicine. http://www8.nationalacademies.org/onpin-
ews/newsitem.aspx?RecordID=12032015a.

National Academies of Sciences, Engineering and Medicine (NAS). 2017. Human Ge-
nome Editing: Science, Ethics and Governance. Washington, DC: National Academies.

Nuffield Council on Bioethics. 2012. Novel Techniques for the Prevention of Mitochondrial
DNA Disorders: An Ethical Review. London: Nuffield Council.

Nuffield Council on Bioethics. 2018. Genome Editing and Human Reproduction. London:
Nuffield Council.

Palacios-Gonzalez, C. 2017. “Does Egg Donation For Mitochondrial Replacement
Techniques Generate Parental Responsibilities?” J Med Ethics. DOI: 10.1136/mede-
thics-2017-104400.

Parfit, D. 1984. Reasons and Persons. New York: Oxford University Press.
Powell, R., and A. Buchanan. 2011. “Breaking Evolution’s Chains: The Prospect of De-

liberate Genetic Modification in Humans.” J Med Philos 36 (1): 6–27. DOI: 10.1093/
jmp/jhq057.

President’s Council on Bioethics. 2003. Beyond Therapy: Biotechnology and the Pursuit of
Happiness. Washington, DC: President’s Council on Bioethics. https://bioethicsar-
chive.georgetown.edu/pcbe/reports/beyondtherapy/fulldoc.html.

Rachels, J. 1975. “Active and Passive Euthanasia.” N Engl J Med 292 (2): 78–80. DOI:
10.1056/NEJM197501092920206.

Playing it Safe?

125winter 2020 • volume 63, number 1

Rulli, T. 2017. “The Mitochondrial Replacement ‘Therapy’ Myth.” Bioethics 31 (5):
368–74. DOI: 10.1111/bioe.12332.

Science and Environmental Health Network (SEHN). 1998. The Wingspread Statement
on the Precautionary Principle. https://www.sehn.org/precautionary-principle-un-
derstanding-science-in-regulation.

Shakespeare, T. 2013. “The Social Model of Disability.” In The Disability Studies Reader,
ed. L. Davis, 214–21. New York: Routledge.

Sheldon, S., and S. Wilkinson. 2004. “Should Selecting Saviour Siblings Be Banned?” J
Med Ethics 30 (6): 533–37. DOI: 10.1136/jme.2003.004150.

Sipp, D., and D. Pei. 2016. “Bioethics in China: No Wild East.” Nature 534 (7608):
465–68. DOI: DOI:10.1038/534465a.

United Nations (UN) Conference on Environment and Development. 1992. Agenda 21,
Rio Declaration, Forest Principles. New York: United Nations.

Verhoog, H. 1981. “The Responsibilities of Scientists.” Minerva 19 (4): 582–604.
Wrigley, A., S. Wilkinson, and J. B. Appleby. 2015. “Mitochondrial Replacement: Ethics

and Identity.” Bioethics 29 (9): 631–38. DOI: 10.1111/bioe.12187.

Reproduced with permission of copyright owner. Further
reproduction prohibited without permission.

514 Gyngell C, et al. J Med Ethics 2019;45:514–523. doi:10.1136/medethics-2018-105084

Moral reasons to edit the human genome: picking up
from the Nuffield report
Christopher Gyngell,1,2 Hilary Bowman-Smart,  2 Julian Savulescu2,3

Feature article

To cite: Gyngell C,
Bowman-Smart H,
Savulescu J. J Med Ethics
2019;45:514–523.

1Department of Paediatrics,
University of Melbourne,
Melbourne, Victoria, Australia
2Murdoch Children’s Research
Institute, Melbourne, Victoria,
Australia
3Faculty of Philosophy, Oxford
Uehiro Centre for Practical
Ethics, Oxford, UK

Correspondence to
Professor Julian Savulescu,
Faculty of Philosophy, The
Oxford Uehiro Centre for
Practical Ethics, Oxford OX1
1PT, UK;
julian. [email protected] philosophy.
ox. ac. uk

Received 7 September 2018
Revised 2 December 2018
Accepted 11 December 2018
Published Online First
24 January 2019

► http:// dx. doi. org/ 10. 1136/
medethics- 2018- 105316

► http:// dx. doi. org/ 10. 1136/
medethics- 2019- 105390

► http:// dx. doi. org/ 10. 1136/
medethics- 2019- 105395

► http:// dx. doi. org/ 10. 1136/
medethics- 2019- 105713

© Author(s) (or their
employer(s)) 2019. Re-use
permitted under CC BY.
Published by BMJ.

AbsTrACT
In July 2018, the Nuffield Council of Bioethics released
its long-awaited report on heritable genome editing
(HGE). The Nuffield report was notable for finding that
HGE could be morally permissible, even in cases of
human enhancement. In this paper, we summarise the
findings of the Nuffield Council report, critically examine
the guiding principles they endorse and suggest ways
in which the guiding principles could be strengthened.
While we support the approach taken by the Nuffield
Council, we argue that detailed consideration of the
moral implications of genome editing yields much
stronger conclusions than they draw. Rather than being
merely ’morally permissible’, many instances of genome
editing will be moral imperatives.

InTroduCTIon
Genome editing technologies have developed
rapidly in the last few years, and point to a future
where we can precisely edit the human germline.
The most powerful gene editing technology is the
CRISPR (Clustered Regularly Interspaced Short
Palindromic Repeats)-Cas9 system. CRISPR-Cas9 is
found naturally in bacteria, where it functions as a
defence against viruses by cutting viral DNA into
small, non-functional fragments. In 2012, a team at
UC Berkeley showed that CRISPR-Cas9 could be
modified in the lab, so that it could target virtu-
ally any DNA sequence.1 This allows researchers
to cut effectively any part of the genome. Further-
more, once a DNA strand is broken, the cell’s own
repair mechanisms could be recruited to delete,
add or modify the sequence. In April 2015, it was
announced that CRISPR had been used to make
edits in human embryos for the first time.2 In
August 2017, researchers in the USA used CRISPR
to correct a mutation in human embryos that
leads to a fatal heart condition—with virtually no
off-target mutations.3 In November 2018, Dr He
Jiankui announced that he had used the CRIS-
PR-Cas9 system to edit the genomes of twins Lulu
and Nana, in an attempt to make them resistant to
HIV.4 Although this has not been independently
confirmed, if true, it would be the first use of
genome editing for human reproduction. This
attempt has largely been met with condemnation,
including by one of the authors of this paper,5 due
to its experimental nature and lack of consideration
for the welfare of the children. Such research has
generated wide debate about the ethics of genome
modification.

Genome editing of germ cells (embryos, sperm
and egg cells) was initially very controversial and
caused some to call for an outright ban on this
application.6 Despite this, there has been a broad

consensus among expert bodies that genome editing
in research is morally permissible (see table 1 for
summary). However, genome editing for reproduc-
tion, a practice called heritable gene editing (HGE),
has been much more contentious.

In July 2018, the Nuffield Council on Bioethics
released its report ‘Genome editing and human
reproduction: social and ethical issues’.7 The
report is significant for advocating an approach
to the assessment of HGE based on ethical princi-
ples rather than applications. Any particular use of
HGE could be morally permissible, provided it was
consistent with promoting individual welfare and
social solidarity.

In this paper, we critically analyse the approach
taken by Nuffield report and its conclusions.

In the first section, The Nuffield Council’s report
on genome editing and human reproduction, we
provide a detailed summary of the Nuffield Coun-
cil’s approach. While we applaud the Nuffield
Council report as a significant step forward in the
debate, we argue there is room to build on and
strengthen their approach. In the second section,
Social harms and collective action problems, we
suggest ways their guiding principles could be
improved by removing an implicit asymmetry
between the two principles. In the third section,
Categorical limits and moral imperatives in HGE,
we argue that the conclusions stated in the report
do not go far enough. Some uses of HGE are not
merely morally permissible but are moral impera-
tives, even beyond the treatment of disease. Finally,
in the section Governance and public attitudes,
we examine the implications of recent attitudinal
research by the Pew Centre which shows that the
general public may support HGE for treatment but
not enhancement.

The nuFFIeld CounCIl’s reporT on genome
edITIng And humAn reproduCTIon
In 2016, following its report which looked at the
ethical issues associated with genome editing more
broadly (e.g., including food), the Nuffield Council
of Bioethics formed a working group to ‘examine
ethical questions relating to the attempted influ-
ence of inherited characteristics in humans, in the
light of the likely impact of genome editing tech-
nologies’. After 2 years of work, the working party
released its report titled ‘Genome editing and
human reproduction’.

The first two sections of the report contextualise
HGE within its immediate potential role as a repro-
ductive technology used by individuals, its possible
future applications and the social context in which
those applications might evolve.

515Gyngell C, et al. J Med Ethics 2019;45:514–523. doi:10.1136/medethics-2018-105084

Feature article

Ta
b

le
1

Re

ce
nt

o
rg

an
is

at
io

na
l o

r
ex

pe
rt

b
od

y
st

at
em

en
ts

o
n

th
e

et
hi

cs
o

f h
er

ita
bl

e
ge

ne
e

di
tin

g
(H

G
E)

*
Ye

ar
b

o
d

y
Ti

tl
e

K
ey

p
o

in
ts

20
18

N
uf

fie
ld

C
ou

nc
il

on
B

io
et

hi
cs

G
en

om
e

ed
iti

ng
a

nd
h

um
an

r
ep

ro
du

ct
io

n
A

pp
lic

at
io

ns
o

f H
G

E
m

ay
b

e
m

or
al

ly
p

er
m

is
si

bl
e

if
th

ey
a

re
c

on
si

st
en

t
w

ith
t

he
w

el
fa

re
o

f t
he

fu
tu

re
p

er
so

n
an

d
so

ci
al

ju
st

ic
e.

T
hi

s
m

ay
a

llo
w

fo
r

ap
pl

ic
at

io
ns

b
ey

on
d

pr
ev

en
tin

g
di

se
as

e.
F

ur
th

er
r

es
ea

rc
h

m
us

t
be

d
on

e
in

to
e

ffi
ca

cy
a

nd
s

af
et

y
an

d
a

br
oa

de
r

so
ci

et
al

d
eb

at
e

m
us

t
be

fa
ci

lit
at

ed
, a

s
w

el
l a

s
en

su
rin

g
ad

eq
ua

te
g

ov
er

na
nc

e.

20
18

G
én

om
e

Q

be
c

&
C

en
tr

e
of

G
en

om
ic

s
an

d
Po

lic
y

H
um

an
g

en
om

e
ed

iti
ng

: e
th

ic
al

a
nd

p
ol

ic
y

co
ns

id
er

at
io

ns
Th

e
m

od
ifi

ca
tio

n
of

h
um

an
g

er
m

c
el

ls
a

nd
e

m
br

yo
s

sh
ou

ld
b

e
pe

rm
itt

ed
in

a
b

as
ic

o
r

pr
ec

lin
ic

al
r

es
ea

rc
h

co
nt

ex
t,

an
y

fu
tu

re
c

lin
ic

al
g

er
m

lin
e

m
od

ifi
ca

tio
ns

s
ho

ul
d

be
fo

r
se

rio
us

di

se
as

es
o

nl
y

(a
nd

c
rit

er
ia

n
ee

d
to

b
e

ou
tli

ne
d)

a
nd

a
n

in
cl

us
iv

e
ap

pr
oa

ch
t

o
po

lic
ym

ak
in

g
sh

ou
ld

b
e

en
co

ur
ag

ed
.

20
17

N
at

io
na

l A
ca

de
m

y
of

S
ci

en
ce

s,
E

ng
in

ee
rin

g
an

d
M

ed
ic

in
e

H
um

an
g

en
om

e
ed

iti
ng

: s
ci

en
ce

, e
th

ic
s

an
d

go
ve

rn
an

ce
If

te
ch

ni
ca

l c
ha

lle
ng

es
r

el
at

in
g

to
s

af
et

y
an

d
ef

fic
ac

y
w

er
e

ov
er

co
m

e,
c

lin
ic

al
t

ria
ls

fo
r

H
G

E
sh

ou
ld

o
nl

y
be

p
er

m
itt

ed
if

t
he

re
is

a
n

ab
se

nc
e

of
r

ea
so

na
bl

e
al

te
rn

at
iv

es
, b

e
re

st
ric

te
d

to

ge
ne

s
th

at
c

le
ar

ly
c

au
se

o
r

pr
ed

is
po

se
t

o
a

se
ve

re
d

is
ab

ili
ty

o
r

di
se

as
e,

w
ith

c
on

ve
rs

io
n

on
ly

t
o

ge
ne

s
th

at
c

ur
re

nt
ly

e
xi

st
in

t
he

p
op

ul
at

io
n.

R
eg

ul
at

or
y

ov
er

si
gh

t
an

d
pu

bl
ic

d
eb

at
e

is

es
se

nt
ia

l.
N

o
tr

ia
ls

fo
r

pu
rp

os
es

o
th

er
t

ha
n

th
e

pr
ev

en
tio

n
of

d
is

ea
se

s
ho

ul
d

be
p

er
m

itt
ed

a
t

th
is

t
im

e,
b

ut
p

ub
lic

d
eb

at
e

on
t

he
m

at
te

r
sh

ou
ld

b
e

fa
ci

lit
at

ed
.

20
17

A
m

er
ic

an
S

oc
ie

ty
o

f H
um

an
G

en
et

ic
s

(A
SH

G
)†

H
um

an
g

er
m

lin
e

ge
no

m
e

ed
iti

ng
: A

SH
G

p
os

iti
on

s
ta

te
m

en
t

1.

A
t

th
is

t
im

e,
it

is
n

ot
a

pp
ro

pr
ia

te
t

o
co

nd
uc

t
H

G
E

th
at

r
es

ul
ts

in
h

um
an

p
re

gn
an

cy
.

2.

A
s

lo
ng

a
s

th
er

e
is

a
pp

ro
pr

ia
te

o
ve

rs
ig

ht
, t

he
re

is
n

o
re

as
on

t
o

pr
oh

ib
it

or
n

ot
u

se
p

ub
lic

fu
nd

s
fo

r
in

v
itr

o
ge

rm
lin

e
ge

ne
e

di
tin

g
fo

r
re

se
ar

ch
in

to
c

lin
ic

al
a

pp
lic

at
io

ns
.

3.

Fu
tu

re
c

lin
ic

al
a

pp
lic

at
io

ns
s

ho
ul

d
on

ly
p

ro
ce

ed
if

t
he

re
is

: (
i)

a
m

ed
ic

al
r

at
io

na
le

, (
ii)

s
tr

on
g

ev
id

en
ce

b
as

e,
(i

ii)
a

n
et

hi
ca

l j
us

tifi
ca

tio
n

an
d

(iv
) a

t
ra

ns
pa

re
nt

p
ub

lic
p

ro
ce

ss
.

20
17

O
pi

ni
on

G
ro

up
o

f t
he

B
io

et
hi

cs
a

nd
L

aw
O

bs
er

va
to

ry
, U

ni
ve

rs
ity

of

B
ar

ce
lo

na
D

ec
la

ra
tio

n
on

b
io

et
hi

cs
a

nd
g

en
e

ed
iti

ng
in

h
um

an
s

A
m

or
at

or
iu

m
is

u
se

le
ss

in
a

n
in

te
rn

at
io

na
l a

nd
c

om
pe

tit
iv

e
sc

ie
nt

ifi
c

w
or

ld
. R

eg
ul

at
or

y
fr

am
ew

or
ks

a
re

n
ee

de
d

to
g

ov
er

n
an

y
cl

in
ic

al
a

pp
lic

at
io

n
of

g
er

m
lin

e
ge

no
m

e
ed

iti
ng

, a
nd

it

sh
ou

ld
n

ot
b

e
us

ed
fo

r
en

ha
nc

em
en

t.

20
17

C
ou

nc
il

of
E

ur
op

e
Re

co
m

m
en

da
tio

n
21

15
: t

he
u

se
o

f n
ew

g
en

et
ic

t
ec

hn
ol

og
ie

s
in

hu

m
an

b
ei

ng
s

D
el

ib
er

at
e

ge
rm

lin
e

ed
iti

ng
in

h
um

an
b

ei
ng

s
w

ou
ld

c
ro

ss
a

li
ne

v
ie

w
ed

a
s

et
hi

ca
lly

in
vi

ol
ab

le
. A

r
eg

ul
at

or
y

fr
am

ew
or

k
ne

ed
s

to
b

e
de

ve
lo

pe
d

an
d

a
br

oa
d

an
d

in
fo

rm
ed

p
ub

lic
d

eb
at

e
sh

ou
ld

b
e

fo
st

er
ed

.

20
17

Fe
de

ra
tio

n
of

E
ur

op
ea

n
A

ca
de

m
ie

s
of

M
ed

ic
in

e
(F

EA
M

)
Th

e
ap

pl
ic

at
io

n
of

g
en

om
e

ed
iti

ng
in

h
um

an
s

Th
er

e
ar

e
m

aj
or

e
th

ic
al

, s
af

et
y

an
d

ef
fic

ac
y

is
su

es
t

ha
t

m
us

t
be

r
es

ol
ve

d
be

fo
re

c
lin

ic
al

a
pp

lic
at

io
ns

o
f H

G
E

ca
n

be
c

on
si

de
re

d.
T

he
re

is
n

o
br

oa
d

so
ci

et
al

c
on

se
ns

us
in

E
ur

op
e.

G

en
er

al
ly

, F
EA

M
d

oe
s

no
t

su
pp

or
t

ge
no

m
e

ed
iti

ng
fo

r
no

n-
m

ed
ic

al
in

te
rv

en
tio

ns
(i

nc
lu

di
ng

s
om

at
ic

g
en

om
e

ed
iti

ng
),

bu
t

an
tic

ip
at

es
a

w
id

e-
ra

ng
in

g
pu

bl
ic

d
is

cu
ss

io
n

on
t

hi
s

m
at

te
r.

20
17

Eu
ro

pe
an

A
ca

de
m

ie
s

Sc
ie

nc
e

A
dv

is
or

y
C

ou
nc

il
G

en
om

e
ed

iti
ng

: s
ci

en
tifi

c
op

po
rt

un
iti

es
, p

ub
lic

in
te

re
st

s
an

d
po

lic
y

op
tio

ns
in

t
he

E
ur

op
ea

n
U

ni
on

It
w

ou
ld

b
e

irr
es

po
ns

ib
le

t
o

pr
oc

ee
d

w
ith

g
er

m
lin

e
ed

iti
ng

u
nt

il
th

e
nu

m
er

ou
s

sa
fe

ty
a

nd
e

ffi
ca

cy
c

on
ce

rn
s

ha
ve

b
ee

n
re

so
lv

ed
. T

he
re

is
a

n
ee

d
fo

r
an

o
ng

oi
ng

fo
ru

m
.

20
17

A
m

er
ic

an
C

ol
le

ge
o

f M
ed

ic
al

G
en

et
ic

s
an

d
G

en
om

ic
s

G
en

om
e

ed
iti

ng
in

c
lin

ic
al

g
en

et
ic

s:
p

oi
nt

s
to

c
on

si
de

r
Th

er
e

ar
e

te
ch

ni
ca

l c
on

ce
rn

s
(e

g,
t

he
r

is
k

of
o

ff
-t

ar
ge

t
ef

fe
ct

s
or

e
pi

ge
ne

tic
e

ff
ec

ts
t

ha
t

m
ay

a
ff

ec
t

m
an

y
fu

tu
re

g
en

er
at

io
ns

) a
s

w
el

l a
s

et
hi

ca
l c

on
ce

rn
s

(w
hi

ch
v

ar
ia

nt
s

th
at

a
re

t
o

be

ed
ite

d
ne

ed
s

to
b

e
di

sc
us

se
d

at
a

s
oc

ie
ta

l l
ev

el
).

20
15

N
at

io
na

l A
ca

de
m

y
of

S
ci

en
ce

s,
E

ng
in

ee
rin

g
an

d
M

ed
ic

in
e

In
te

rn
at

io
na

l s
um

m
it

on
h

um
an

g
en

e
ed

iti
ng

It
w

ou
ld

b
e

irr
es

po
ns

ib
le

t
o

pr
oc

ee
d

w
ith

a
ny

c
lin

ic
al

u
se

o
f g

er
m

lin
e

ed
iti

ng
u

nl
es

s
an

d
un

til
(i

) t
he

r
el

ev
an

t
sa

fe
ty

a
nd

e
ffi

ca
cy

is
su

es
h

av
e

be
en

r
es

ol
ve

d,
b

as
ed

o
n

ap
pr

op
ria

te

un
de

rs
ta

nd
in

g
an

d
ba

la
nc

in
g

of
r

is
ks

, p
ot

en
tia

l b
en

efi
ts

a
nd

a
lte

rn
at

iv
es

, a
nd

(i
i)

th
er

e
is

b
ro

ad
s

oc
ie

ta
l c

on
se

ns
us

a
bo

ut
t

he
a

pp
ro

pr
ia

te
ne

ss
o

f t
he

p
ro

po
se

d
ap

pl
ic

at
io

n.

20
15

A
ca

de
m

y
of

M
ed

ic
al

S
ci

en
ce

s
(U

K)

G
en

om
e

ed
iti

ng
in

h
um

an
c

el
ls


in

iti
al

jo
in

t
st

at
em

en
t

Et
hi

ca
l a

nd
r

eg
ul

at
or

y
qu

es
tio

ns
m

us
t

be
d

eb
at

ed
b

ef
or

e
cl

in
ic

al
a

pp
lic

at
io

ns
o

f e
di

tin
g

ge
rm

c
el

ls
c

an
b

e
co

ns
id

er
ed

.

20
15

H
in

xt
on

G
ro

up
§

St
at

em
en

t
on

g
en

om
e

ed
iti

ng
t

ec
hn

ol
og

ie
s

an
d

hu
m

an
g

er
m

lin
e

ge
ne

tic
m

od
ifi

ca
tio

n
W

he
n

al
l s

af
et

y,
e

ffi
ca

cy
a

nd
g

ov
er

na
nc

e
ne

ed
s

ar
e

m
et

, t
he

re
m

ay
b

e
m

or
al

ly
a

cc
ep

ta
bl

e
us

es
fo

r
H

G
E

fo
r

re
pr

od
uc

tiv
e

pu
rp

os
es

. F
ur

th
er

d
eb

at
e

as
t

o
w

ha
t

sp
ec

ifi
c

us
es

a
re

m
or

al
ly

ac

ce
pt

ab
le

is
r

eq
ui

re
d.

20
15

A
m

er
ic

an
S

oc
ie

ty
fo

r
G

en
e

an
d

C
el

l T
he

ra
py

(A
SG

C
T)

a
nd

J
ap

an

So
ci

et
y

of
G

en
e

Th
er

ap
y

(J
SG

T)
A

SG
C

T
an

d
JS

G
T

jo
in

t
po

si
tio

n
st

at
em

en
t

on
h

um
an

g
en

om
ic

e
di

tin
g

A
s

av
ai

la
bl

e
ge

no
m

e
ed

iti
ng

t
ec

hn
ol

og
ie

s
ar

e
in

ad
eq

ua
te

ly
u

nd
er

st
oo

d,
a

nd
b

ec
au

se
t

he
r

es
ul

ts
o

f s
uc

h
m

an
ip

ul
at

io
n

co
ul

d
no

t
be

u
nd

er
st

oo
d

fo
r

de
ca

de
s

or
g

en
er

at
io

ns
, t

he
se

s
af

et
y

an
d

ef
fic

ac
y

co
nc

er
ns

a
re

s
uf

fic
ie

nt
ly

s
er

io
us

t
o

su
pp

or
t

a
st

ro
ng

s
ta

nc
e

ag
ai

ns
t

H
G

E.

20
15

A
lli

an
ce

fo
r

Re
ge

ne
ra

tiv
e

M
ed

ic
in


D

o
no

t
ed

it
th

e
hu

m
an

g
er

m
lin

e
Th

e
sc

ie
nt

ifi
c

co
m

m
un

ity
is

u
rg

ed
t

o
en

ga
ge

in
d

ia
lo

gu
e

an
d

pu
t

in
p

la
ce

a
v

ol
un

ta
ry

m
or

at
or

iu
m

t
o

di
sc

ou
ra

ge
h

um
an

g
er

m
lin

e
m

od
ifi

ca
tio

n.
T

hi
s

is
d

ue
t

o
sc

ie
nt

ifi
c

an
d

et
hi

ca
l r

is
ks

su

ch
a

s
th

at
p

er
m

itt
in

g
ev

en
u

na
m

bi
gu

ou
sl

y
th

er
ap

eu
tic

in
te

rv
en

tio
ns

c
ou

ld
s

ta
rt

d
ow

n
a

pa
th

t
ow

ar
ds

n
on

-t
he

ra
pe

ut
ic

g
en

et
ic

e
nh

an
ce

m
en

t.

20
15

IG
I F

or
um

o
n

Bi
oe

th
ic

s,
N

ap
a,

C
al

ifo
rn

ia
**

A
p

ru
de

nt
p

at
h

fo
rw

ar
d

fo
r

ge
no

m
ic

e
ng

in
ee

rin
g

an
d

ge
rm

lin
e

ge
ne

m

od
ifi

ca
tio

n
1.

A

ny
a

tt
em

pt
s

at
c

lin
ic

al
a

pp
lic

at
io

ns
o

f g
er

m
lin

e
ge

ne
e

di
tin

g
ar

e
st

ro
ng

ly
d

is
co

ur
ag

ed
.

2.

Ex
pe

rt
fo

ru
m

s
of

s
ci

en
tis

ts
a

nd
b

io
et

hi
ci

st
s

m
us

t
be

h
el

d
to

fu
rt

he
r

di
sc

us
s

th
es

e
is

su
es

.
3.

Tr

an
sp

ar
en

t
re

se
ar

ch
s

ho
ul

d
be

e
nc

ou
ra

ge
d

an
d

su
pp

or
te

d.
4.

A

g
lo

ba
lly

r
ep

re
se

nt
at

iv
e

gr
ou

p
of

a
w

id
e

ra
ng

e
of

s
ta

ke
ho

ld
er

s
(in

cl
ud

in
g

th
e

ge
ne

ra
l p

ub
lic

a
nd

g
ov

er
nm

en
t

bo
di

es
) s

ho
ul

d
be

a
ss

em
bl

ed
t

o
fu

rt
he

r
di

sc
us

s
th

es
e

is
su

es
a

nd

m
ak

e
po

lic
y

re
co

m
m

en
da

tio
ns

.

20
15

In
te

rn
at

io
na

l S
oc

ie
ty

fo
r

St
em

C
el

l R
es

ea
rc

h
(IS

SC
R)

††
Th

e
IS

SC
R

st
at

em
en

t
on

h
um

an
g

er
m

lin
e

ge
no

m
e

m
od

ifi
ca

tio
n

A
ny

c
on

si
de

ra
tio

n
of

a
pp

ly
in

g
nu

cl
ea

r
ge

no
m

e
ed

iti
ng

t
o

th
e

hu
m

an
g

er
m

lin
e

in
c

lin
ic

al
p

ra
ct

ic
e

ra
is

es
s

ig
ni

fic
an

t
et

hi
ca

l,
so

ci
et

al
a

nd
s

af
et

y
co

ns
id

er
at

io
ns

. T
he

re
fo

re
, t

he
IS

SC
R

ca
lls

fo

r
a

m
or

at
or

iu
m

o
n

ge
rm

lin
e

ge
no

m
e

ed
iti

ng
in

c
lin

ic
al

p
ra

ct
ic

e
(e

xc
ep

tin
g

m
ito

ch
on

dr
ia

l r
ep

la
ce

m
en

t
th

er
ap

y)
, a

lth
ou

gh
in

v
itr

o
re

se
ar

ch
is

s
up

po
rt

ed
.

20
15

N
at

io
na

l I
ns

tit
ut

es
o

f H
ea

lth
(N

IH
)

St
at

em
en

t
on

N
IH

fu
nd

in
g

of
r

es
ea

rc
h

us
in

g
ge

ne
-e

di
tin

g
te

ch
no

lo
gi

es
in

h
um

an
e

m
br

yo
s

Sa
fe

ty
, e

th
ic

al
a

nd
r

eg
ul

at
or

y
is

su
es

m
ea

ns
t

ha
t

al
te

rin
g

th
e

hu
m

an
g

er
m

lin
e

in
e

m
br

yo
s

is
‘a

li
ne

t
ha

t
sh

ou
ld

n
ot

b
e

cr
os

se
d’

. N
IH

w
ill

n
ot

fu
nd

a
ny

u
se

o
f g

en
e

ed
iti

ng
in

e
m

br
yo

s.
(N

.B
.:

as
o

f 2
01

8,
N

IH
is

fu
nd

in
g

re
se

ar
ch

in
to

s
om

at
ic

c
el

l g
en

om
e

ed
iti

ng
).

*O
rg

an
is

at
io

na
l r

ec
om

m
en

da
tio

ns
a

re
in

cl
ud

ed
h

er
e.

A
s

ea
rc

h
w

as
c

on
du

ct
ed

u
si

ng
a

b
ra

nc
he

d
st

ra
te

gy
, b

eg
in

ni
ng

w
ith

t
he

u
se

o
f s

ea
rc

h
te

rm
s

in
d

at
ab

as
es

s
uc

h
as

P
ub

M
ed

a
nd

G
oo

gl
e

Sc
ho

la
r

us
in

g
se

ar
ch

t
er

m
s

su
ch

a
s

‘h
um

an
h

er
ita

bl
e

ge
no

m
e

ed
iti

ng
’,

‘g
er

m
lin

e
ge

ne
e

di
tin

g’
, ‘

ge
no

m
e

ed
iti

ng
e

m
br

yo
s’

, ‘
ge

rm
lin

e
en

gi
ne

er
in

g’
, ‘

hu
m

an
g

er
m

lin
e

ed
iti

ng
’+

‘e
th

ic
s’

, ‘
po

lic
y’

a
nd

/o
r

‘r
ec

om
m

en
da

tio
ns

’.
Fu

rt
he

rm
or

e,
c

ita
tio

ns
a

nd
r

ef
er

en
ce

s
w

er
e

ex
am

in
ed

t
o

ge
ne

ra
te

fu
rt

he
r

re
su

lts
. A

la
rg

e
nu

m
be

r
of

p
ap

er
s

w
er

e
id

en
tifi

ed
d

is
cu

ss
in

g
th

es
e

is
su

es
, i

nc
lu

di
ng

a
n

um
be

r
of

r
ec

om
m

en
da

tio
ns

. R
es

ul
ts

w
er

e
lim

ite
d

to
t

ho
se

fr
om

2
00

8
on

w
ar

ds
(i

e,
t

he
p

as
t

de
ca

de
a

t
tim

e
of

w
rit

in
g)

; a
ll

pa
pe

rs
b

y
in

di
vi

du
al

s
or

g
ro

up
s

of
in

di
vi

du
al

s
no

t
re

pr
es

en
tin

g
an

o
rg

an
is

at
io

n,
b

od
y

or

co
nf

er
en

ce
w

er
e

di
sc

ar
de

d,
a

s
w

el
l a

s
em

pi
ric

al
r

es
ea

rc
h

in
to

o
pi

ni
on

s
on

g
er

m
lin

e
ed

iti
ng

b
y

sp
ec

ifi
c

se
ct

or
s

of
s

oc
ie

ty
(e

g,
g

en
er

al
p

ub
lic

, h
ea

lth
ca

re
p

ro
fe

ss
io

na
ls

).
Th

e
fin

al
g

ro
up

o
f r

es
ul

ts
w

as
li

m
ite

d
to

o
rg

an
is

at
io

ns
w

ho
se

r
em

it
in

cl
ud

es
h

um
an

g
en

et
ic

s,
g

en
et

ic
s

po
lic

y,
b

io
et

hi
cs

, r
ep

ro
du

ct
iv

e
te

ch
no

lo
gi

es
a

nd
/o

r
em

er
gi

ng
t

ec
hn

ol
og

ie
s.

O
rg

an
is

at
io

na
l r

es
po

ns
es

t
o

st
at

em
en

ts
a

nd

jo
ur

na
l e

di
to

ria
ls

w
er

e
no

t
in

cl
ud

ed
. T

hi
s

lis
t

is
n

ot
e

xh
au

st
iv

e.
†S

ta
te

m
en

t
en

do
rs

ed
b

y
U

K
A

ss
oc

ia
tio

n
of

G
en

et
ic

N
ur

se
s

an
d

C
ou

ns
el

lo
rs

, C
an

ad
ia

n
A

ss
oc

ia
tio

n
of

G
en

et
ic

C
ou

ns
el

lo
rs

, I
nt

er
na

tio
na

l G
en

et
ic

E
pi

de
m

io
lo

gy
S

oc
ie

ty
, U

S
N

at
io

na
l S

oc
ie

ty
o

f G
en

et
ic

C
ou

ns
el

or
s,

A
m

er
ic

an
S

oc
ie

ty
fo

r
Re

pr
od

uc
tiv

e
M

ed
ic

in
e,

A
si

a
Pa

ci
fic

S
oc

ie
ty

o
f H

um
an

G
en

et
ic

s,
B

rit
is

h
So

ci
et

y
fo

r
G

en
et

ic
M

ed
ic

in
e,

H
um

an
G

en
et

ic
s

So
ci

et
y

of
A

us
tr

al
as

ia
, P

ro
fe

ss
io

na
l

So
ci

et
y

of
G

en
et

ic
C

ou
ns

el
or

s
in

A
si

a,
a

nd
S

ou
th

er
n

A
fr

ic
an

S
oc

ie
ty

fo
r

H
um

an
G

en
et

ic
s.

T
he

A
lli

an
ce

fo
r

Re
ge

ne
ra

tiv
e

M
ed

ic
in

e
(a

n
in

te
rn

at
io

na
l m

ul
tis

ta
ke

ho
ld

er
g

ro
up

, i
nc

lu
di

ng
c

om
m

er
ci

al
s

ta
ke

ho
ld

er
s)

, r
el

ea
se

d
a

st
at

em
en

t
su

pp
or

tin
g

th
is

p
os

iti
on

.
‡J

oi
nt

s
ta

te
m

en
t

w
ith

t
he

A
ss

oc
ia

tio
n

of
M

ed
ic

al
R

es
ea

rc
h

C
ha

rit
ie

s,
t

he
M

ed
ic

al
R

es
ea

rc
h

C
ou

nc
il

an
d

th
e

W
el

lc
om

e
Tr

us
t.

§A
g

ro
up

c
on

si
st

in
g

of
a

n
in

te
rn

at
io

na
l c

ol
le

ct
io

n
of

a
ca

de
m

ic
s

fr
om

d
is

ci
pl

in
es

s
uc

h
as

t
he

li
fe

s
ci

en
ce

s,
p

hi
lo

so
ph

y,
m

ed
ic

in
e

an
d

ec
on

om
ic

s,
fr

om
u

ni
ve

rs
iti

es
in

cl
ud

in
g

Jo
hn

H
op

ki
ns

, O
xf

or
d

an
d

Lo
nd

on
S

ch
oo

l o
f E

co
no

m
ic

s.

A
g

ro
up

c
on

si
st

in
g

of
c

om
m

er
ci

al
c

om
pa

ni
es

, r
es

ea
rc

h
in

st
itu

tio
ns

, n
on

-p
ro

fit
o

rg
an

is
at

io
ns

a
nd

p
at

ie
nt

a
dv

oc
ac

y
gr

ou
ps

. C
or

po
ra

tio
ns

c
om

pr
is

e
73

%
o

f s
ta

ke
ho

ld
er

s
an

d
th

us
t

hi
s

or
ga

ni
sa

tio
n

sh
ou

ld
b

e
pr

im
ar

ily
c

on
si

de
re

d
to

r
ep

re
se

nt
t

he
v

ie
w

s
of

c
or

po
ra

tio
ns

s
uc

h
BD

B
io

sc
ie

nc
es

, G
E

H
ea

lth
ca

re
, G

SK
, M

ill
ip

or
eS

ig
m

a,
N

ov
ar

tis
a

nd
P

fiz
er

, a
m

on
g

m
an

y
ot

he
rs

. T
he

y
la

te
r

re
le

as
ed

a

st
at

em
en

t
su

pp
or

tin
g

th
e

A
SH

G
p

os
iti

on
s

ta
te

m
en

t
(s

ee
fo

ot
no

te
1

).
**

A
la

rg
e

nu
m

be
r

of
e

xp
er

t
st

ak
eh

ol
de

rs
a

tt
en

de
d

an
d

m
ad

e
th

is
p

os
iti

on
s

ta
te

m
en

t,
in

cl
ud

in
g

th
e

in
ve

nt
or

s
of

C
RI

SP
R-

C
as

9.
††

A
n

on
-p

ro
fit

o
rg

an
is

at
io

n
en

co
ur

ag
in

g
re

se
ar

ch
in

to
t

he
u

se
o

f s
te

m
c

el
ls

.

516 Gyngell C, et al. J Med Ethics 2019;45:514–523. doi:10.1136/medethics-2018-105084

Feature article

The report states that genome editing in the context of repro-
duction must be situated against a background of increased
genetic knowledge informing reproductive options. Increased
knowledge about genetic differences has created an ‘epistemic
shift’ revealing previous dichotomies between states of health
and disease, and thus therapeutic and non-therapeutic applica-
tions, to be inadequate (p. 26). Even if one views HGE as only
permissible within the context of ‘therapeutic’ applications, its
position as a reproductive technology means that it cannot be
viewed as straightforwardly therapeutic in the same way as other
medical technologies. HGE is not therapeutic in the sense that
nobody who is in a current state of disease is being treated, but
nor is it straightforwardly preventative (because the risk could be
addressed by not having children) (pp. 22–23). Genomic inter-
vention in reproduction is distinct from other human applica-
tions because it deals with possible persons rather than existing
persons; it must be viewed as a means of fulfilling reproductive
desires rather than a means of preventing disease (p. 47, foot-
note 143).

The report goes on to say that genetic information (that can
be acquired by a number of technologies) places new responsi-
bilities on whether to act or not act on this information when
considering reproduction. Prospective parents who wish to
use this knowledge to avoid or ensure certain genetic variants
in their offspring are seeking specific outcomes. They desire a
specific kind of child, that is, one that is genetically related to
them and does/does not have a specific trait, condition or char-
acteristic (p. 23). Existing assisted reproductive technologies,
such as pre-implantation genetic diagnosis, provide one way for
parents to pursue such goals, but rely on a sufficient abundance
of embryos. If this is not possible, HGE may be more accept-
able than other means to achieve parenthood such as donated
gametes, due to the strong preference many people have for
genetically related offspring (p. 25).

The report next situates the development of genome editing
within its technical possibilities and a social and political
context. Here, the report highlights that genome editing should
not be viewed in isolation as an ‘innovation’, but instead encour-
ages us to consider what a society in which it is widely available
might look like. It outlines various strategies by which HGE
might be deployed, including at the zygote stage on embryos
created through in vitro fertilisation (IVF), and the possibility of
creating modified gametes from induced pluripotent stem (iPS)
cells instead of editing embryos directly (pp. 37–39).

There are a number of situations identified in the report where
HGE may be the only option to create a genetically related child.
These include the cases of Y-chromosome defects, or dominant
conditions where one parent is homozygous (p. 45). More
likely scenarios where HGE may be necessary are those where
the chance of an unaffected embryo would be low to very low.
For example, one or both parents may be heterozygous for a
dominant condition, or cases where there are multiple undesired
independently sorting variants (p. 45). Although not mentioned
in the report, another circumstance where HGE might be the
only option to create healthy genetically related children is when
dominant de novo mutations occur in the germ cell line, such as
within spermatogonial stem cells.

The report even raises the possibility of using HGE to avoid
complex conditions that are common in populations and are
difficult to avoid through selective approaches, because of the
large numbers of genes involved (p. 46). Looking forward to a
future in which HGE is widely available, the report envisages
a wide range of possible applications. These include increased
immunity and resistance to disease, tolerance for adverse

environmental conditions (such as that of space), superabilities
or other various factors such as the ability to make vitamins
rather than having to consume them (p. 47).

The report notes that the social and political drivers for
the development of this technology may initially be the use of
embryos for ‘basic’ research. However, researchers cannot be
morally insulated from the ethical implications of further uses
that might develop from applied research (pp. 48–49). The
report supports responsible research and innovation, encour-
aging reflection that may counteract technological momentum
(p. 49). The use of genome editing in human reproduction has
the potential to be socially transformative, and policy and regu-
lation will play a key role in how this transformation may play
out. The report identifies three kinds of concerns about the inte-
gration of new technologies into the landscape: first, that we
may simply ‘sleepwalk’ into a new world, due to uncontrolled
technological momentum; second, that the technology may
be subject to function creep, where its use expands in possibly
morally troubling ways; and third, that we may be headed down
a slippery slope with no reliable ethical or legal means to distin-
guish morally unacceptable applications from morally acceptable
ones (p. 55). Identifying these concerns can help to recognise
key points in the process where better governance and ethical
reflection may play an even more important role.

The report’s approach to the ethical issues surrounding HGE
is largely framed around the concept of human rights and inter-
ests, with a view to the production of general principles. It exam-
ines the ethical issues through the lens of three different kinds
of interests: (i) individuals directly affected by the technology
(the parents and the future person), (ii) society, particularly those
who may collaterally affected in less immediate and direct ways
(such as people with genetic conditions) and (iii) the interests of
humanity in general and future generations (p. 59). This exam-
ination concludes that ‘none of the considerations raised yields
an ethical principle that would constitute a categorical reason to
prohibit heritable genome editing interventions’ (p. xviii).

The report observes that, in terms of the interests of individ-
uals, the use of HGE must be balanced between the prospective
parents’ interests and the welfare of the future person that may
result. Prospective parents often desire a child that is geneti-
cally related to them, for a variety of reasons, most of which
are ‘felt as well as reasoned’ (p. 59). Nonetheless, it does not
necessarily follow that these desires constitute any sort of moral
claim to a genetically related child. However, the report looks at
two reasons why individuals should be supported in their repro-
ductive projects. The first is from the view that procreation is a
good, with a naturalist perspective seeing it as an essential part
of human function and flourishing or the satisfaction of a natural
human desire. More moderately, a view of procreation as a good
can come from a recognition that social arrangements favour
procreation. However, as the report concludes, it is distinctly
difficult to position procreation as a good in itself (pp. 62–63).
The second reason people should be supported in their repro-
ductive projects stems from a respect for procreative interests.
These interests lead to both a negative right and a positive right,
although the extent to which the latter is applicable is likely to
be dependent on social context (pp. 63–64).

The reproductive interests of the parents must, however, be
constrained by another set of interests: that of the future person.
This, of course, raises issues such as the non-identity problem,
which the report addresses in the following fashion. There are
a number of possible children that exist in the form of mental
images that the parents may have; as more and more decisions
are made during the reproduction process, the diversity of these

517Gyngell C, et al. J Med Ethics 2019;45:514–523. doi:10.1136/medethics-2018-105084

Feature article

possible children narrows to become closer to the nature of the
actual future child. These decisions can be about various proper-
ties of the child, and such properties can include those relating
to the child’s levels of welfare. The parents bear responsibility
for the state of affairs that result from their decisions, which
carry moral weight (pp. 66–67). The report examines the extent
and limits of this responsibility. It discusses arguments that have
been put forward that (i) no genome editing is permissible (such
as those autonomy-based arguments from Habermas), (ii) some
genome editing is permissible (generally drawing a distinction
between ‘therapeutic’ and ‘enhancement’ applications) and (iii)
some genome editing is morally required (similar to the principle
of procreative beneficence).

The report addresses a number of issues with all of these
approaches. The first set of arguments raise the issue of genetic
determinism, and the question of how genome editing can be
sufficiently demarcated from other parenting strategies in a
way that is morally coherent (p. 68). The second falls prey to
the difficulty of distinguishing therapeutic applications from
enhancement, including but not limited to disability rights and
feminist critiques of the normativity implicit in any such argu-
ment (pp. 69–72). About the third, the report raises concerns
about the application of this principle in practice, such as the
ability to sufficiently and reliably identify genomic variants asso-
ciated with welfare, and the risk of the burden of expectation
(p. 72). However, all these approaches inform the first principle
that the report formulates: the use of genome editing technolo-
gies must secure and be consistent with the welfare of any future
person that may be born as a result of those technologies (p. 75).
This principle is necessary but not sufficient for any application
of HGE to be morally permissible.

principle 1: the welfare of the future person
Gametes or embryos that have been subject to genome editing
procedures (or that are derived from cells that have been subject
to such procedures) should be used only where the procedure is
carried out in a manner and for a purpose that is intended to secure
the welfare of and is consistent with the welfare of a person who
may be born as a consequence of treatment using those cells.

It is notable that in the formulation of this principle, the Nuffield
Council has specifically referenced a general approach focused
on the welfare of the future person rather than any particular
distinction between different applications of HGE, such as ‘ther-
apeutic’ versus ‘enhancement’. As the report itself notes, it is
very difficult to draw a clear distinction between therapeutic
and non-therapeutic applications and this approach sidesteps
this difficulty. The report explicitly states that there is no a priori
reason that applications beyond the prevention of disease would
not also be consistent with the welfare of the future person (p.
76). This is a significant step in opening the door for a variety of
traits and characteristics to be considered for HGE. For example,
this raises the possibility of actively including certain welfare-im-
proving characteristics, rather than limiting HGE to the removal
of welfare-diminishing characteristics.

The report goes on to address the interests of society, noting
that reproduction takes place in a social context. Reproductive
behaviours already can change the composition of a society
without deliberate coordination, but reproductive technologies
allow for this with greater certainty (p. 78). Other people in
society may be collaterally and indirectly affected by the use of
HGE. The report highlights questions surrounding diversity,
shifting norms, disability critiques, social virtues and equity and
justice.

The report notes that it is possible that genome editing could
lead to changes in the level of diversity within the population—
whether more or less would depend strongly on prevailing social
factors (pp. 79–80). If the use of HGE becomes standard, this
could lead to a shift in norms and the expectation of its use,
which could decrease freedom. This can be reflected in current
concerns around prenatal screening—what people typically do
becomes what people should do, and thus what people feel pres-
sured to do (pp. 80–82). This follows on to disability critiques,
such as the expressivist objection, which states that attempts to
deselect or prevent disability expresses or presupposes a negative
view of people with a disability. However, the Nuffield Council
explicitly rejects this objection when it comes to genetic condi-
tions that significantly affect both quality and length of life (p.
82). Another set of critiques is that HGE represents attempts to
overcome fragility and weakness, which are in themselves a valu-
able part of the human condition and should not be removed.
However, the difficulty with these approaches is that a person
would have to value such fragility enough that they would be
willing to impart it on their own children (pp. 82–85). Finally,
the report considers the importance of equity and justice and
concludes that HGE should be restricted so as not to result in
unfair advantages for certain groups of people (pp. 85–86). This
leads on to the second principle formulated in the report (p. 87).

principle 2: social justice and solidarity
The use of gametes or embryos that have been subject to genome
editing procedures (or that are derived from cells that have
been subject to such procedures) should be permitted only in
circumstances in which it cannot reasonably be expected to produce
or exacerbate social division or the unmitigated marginalisation or
disadvantage of groups within society.

The third set of interests that the report addresses is that of
humanity and future generations. Potential adverse effects of
HGE may only manifest themselves after several generations,
and here the notion that we have responsibilities or moral
obligations to future generations is key (p. 89). One perspec-
tive is that HGE could offer a means to remedy harms that
have already been set in motion for future generations, such as
runaway climate change (pp. 89–90). However, invoking the
‘precautionary principle’ would suggest that the uncertainty
and possible negative consequences of HGE mean that it should
not be applied. However, as the report notes, this constitutes
no reason not to continue research and the development of the
technology as a means of hedging bets against future events (pp.
90–91).

The report also addresses the relationship between HGE
and transhumanism. Transhumanism is a concept that is closely
linked to the emergence of technologies such as HGE. HGE
could lead to the ‘self-overcoming’ of the human species to a
grander, more capable species, and the report questions whether
this constitutes a moral reason not to apply it. This may stem
from a notion of a fundamental human dignity. HGE might be
troubling because it threatens the integrity of human genetic
inheritance (interference with the line of transmission that links
the human family together) and the integrity of human genomic
identity (distinguishing the human family from non-human
beings) (p. 91). These concerns could be mitigated by replacing
variants only with wild-type or typical variants, but of course
the question of what is a wild-type or mutant variant is often
value-laden in itself. The human genome is enormously varied,
and many new variants are possible and likely to emerge. The
report responds to this by stating that concerns about moving

518 Gyngell C, et al. J Med Ethics 2019;45:514–523. doi:10.1136/medethics-2018-105084

Feature article

the human genome away from wild-type variants are pruden-
tial, rather than a categorical moral reason not to do so (p. 92).
However, questions remain surrounding justice and equity, and
the possibility of schisms between the ‘gene-rich’ and the ‘gene-
poor’ (pp. 92–95).

The report concludes by stating that there are no categorical
limits on the use of genome editing technologies, as long as (p.
97):

► They are not biologically reckless;
► They are consistent with the welfare of future people;
► They are not socially divisive;
► They are not initiated without prior societal debate.
We applaud the Nuffield Council’s approach to HGE and

think it is an important step forward in the debate. A strength
of the approach is that it outlines quite specific moral principles
rather than merely appealing to broad concepts—such as earlier
reports.

For example, the National Academiy of Sciences (NAS) report
on genome editing endorses the principle of:

Promoting well-being: the principle of promoting well-being
supports providing benefit and preventing harm to those affected,
often referred to in the bioethics literature as the principles of
beneficence and nonmaleficence.

It is left unspecified whose well-being we should be promoting
through genome editing, what the components of well-being/harm
are and exactly how that principle should apply to genome editing.
Nearly all people would freely endorse this principle (and many of
the other principles relied on in the National Academy of sciences
report), but might draw radically different conclusions regarding
genome editing. In contrast, the Nuffield Council’s first principle
is much more specific and the scope and force is clear.

Nonetheless, there is room to improve and build from the
Nuffield Council’s approach. In the next section, we show how
an asymmetry in the structure between the two guiding princi-
ples leads to counterintuitive implications for an important set
of possible uses for HGE, and suggest ways their second prin-
ciple could be improved.

soCIAl hArms And ColleCTIve ACTIon problems
The first guiding principle adopted by the Nuffield Council
concerns its effect on individuals. The report draws on the
term ‘welfare’, which is explained as ‘a broader concept than
well-being (“being well”, ie, “healthy”). In this sense, psychoso-
cial welfare, and not just good health, is an important consid-
eration’ (p. 76). Genome editing is only morally permissible if
it is ‘carried out in a manner and for a purpose that is intended
to secure the welfare of and is consistent with the welfare of a
person who may be born’ (p. 75).

A relatively broad range of ways in which individuals could be
harmed or wronged is discussed in the report. This includes the
effects of genome editing on one’s physical health, and people’s
psychological well-being, including any state ‘that might give the
future person reasonable grounds to reprove their parents’ (p. 96).

In contrast, principle 2—which looks at the social effects of
genome editing—is quite specific. Genome editing would be
impermissible if it were ‘to produce or exacerbate social division
or the unmitigated marginalisation or disadvantage of groups’
(p. 87). But this principle seems to overlook the fact that there
are many ways in which society could be made worse that do
not involve the creation of social division or marginalisation.
If everyone in society were to be made significantly worse off,

but in a way that did not increase inequality (or even possibly
decreased it), we would still most likely view this as an undesir-
able state of affairs.

One strand of arguments in the literature on genetic enhance-
ment centre on so-called ‘collective action problems’.8–10 In
a collective action problem, one option is optimific from the
perspective of an individual but results in collective harms if
everyone pursues it. Imagine if it becomes possible to use HGE
select for or against the predisposition to particular personality
traits, such as extroversion. Extroversion is highly heritable11
and associated with increased levels of subjective well-being.12
Furthermore, recent studies have shown that mothers value
extroversion in their children above other traits such as intelli-
gence and conscientiousness.13 It is therefore plausible that were
parents able to access HGE to increase their chances of having
an extroverted child, they would use it. This would be consis-
tent with both of the Nuffield Council’s guiding principles. As
extroversion is associated with higher levels of subjective well-
being, such a change would be consistent with the welfare of the
child. While decreasing the frequency of introverts in society
might lead to increased division and marginalisation as they
become ‘the odd one out’, this is not necessarily so (they might
become more highly prized as they become rarer). At any rate,
the wrongness of such selection should not depend entirely on
the contingent response of individuals to the decreasing features
of a trait such as introversion.

If HGE were to dramatically increase the rate of extroverts in
society, there is a sense that this would make society imperson-
ally worse. Introverts contribute important forms of cognitive
diversity which can benefit group problem solving.14 15 Having
introverts in society can thus benefit many areas of life including
achievement in the science and the arts.16

The ability of parents to target other characteristics through
HGE, including height and innate immunity, could also lead to
collective action problems.8 Such edits would be consistent with
welfare of the child, yet if many people made those changes to
their children, it could make society worse off in ways that do
not necessarily involve increasing social division or marginalisa-
tion. If the average height of the population increased, this could
increase the amount of resources that we use, and thus damage
the environment. If many people selected similar immune genes
for their children, this could leave us more susceptible to novel
pathogens in the future.

This is not a criticism of the Nuffield Council’s report, as
the type of HGE applications which could lead to collective
problems are a long way off—and their consideration is not
a pressing concern for the regulation of HGE now. However,
reflection on them suggests ways in which the Nuffield report’s
guiding principles could be improved. Namely, the second prin-
ciple should evoke a broader concept of social harm, analogous
to the concept of welfare in the first principle.

modified principle 2: social harms
The use of gametes or embryos that have been subject to genome
editing procedures (or that are derived from cells that have
been subject to such procedures) should be permitted only in
circumstances in which it cannot reasonably be expected to produce
or exacerbate social harms, including increased social division or
the unmitigated marginalisation or disadvantage of groups within
society.

While this principle loses some of the advantageous of spec-
ificity, it has the resources to respond to concerns relating to
collective action problems.

519Gyngell C, et al. J Med Ethics 2019;45:514–523. doi:10.1136/medethics-2018-105084

Feature article

CATegorICAl lImITs And morAl ImperATIves In hge
As stated above, the central conclusion of the Nuffield report on
HGE was that there are no categorical limits on its use, provided
applications are consistent with its guiding principles and
preceded with broad public debate. We believe much stronger
conclusions regarding the ethics of HGE can be drawn.

Technologies like HGE cannot be good or bad absolutely. We
can speak of whether a particular application of a technology
is good or bad, or whether their availability has good or bad
effects on society—but technologies themselves are not the type
of object to which the property of ‘good’ or ‘bad’ attaches.

The most basic ethical questions regarding HGE is therefore
whether particular applications of it are good, bad, permissible,
desirable, etc. In this section, we will examine some possible
applications of HGE and show that rather than being merely
morally permissible, some applications will be moral imperatives.

single gene disorders
A mark of success of medical genetics has been the diagnosis
of the disease phenylketonuria (PKU) at birth. This is an inher-
ited metabolic disorder in which levels of the enzyme phenylal-
anine hydroxylase are lowered. This means individuals cannot
metabolise the amino acid phenylalanine. In 1962, a test was
devised that allowed PKU to be diagnosed through a blood
test.17 The ‘heel prick test’ is now routinely given to infants as
part of newborn screening. Those children who are identified
as suffering from PKU are put on a low phenylalanine diet or
else they will develop severe intellectual disability. This diet
means no bread, pasta, soybeans, egg whites, meat, legumes,
nuts, watercress and fish. Such an environmental intervention is
demanding. There is always a risk that foods containing phenyl-
alanine will be consumed by mistake. The ubiquitous sweetener,
aspartame, can cause a crisis.

Imagine an artificial enzyme was developed to replace phenyl-
alanine. If this was administered regularly it would allow
sufferers of PKU to consume a normal diet. Such a cure would be
hailed as a breakthrough. There would be a moral imperative to
provide this cure, just as there is an imperative to provide blood
transfusion for severe bleeding, and antibiotics for infection.

Now imagine that instead of getting a pharmaceutical
company to manufacture the enzyme, we could get the body to
manufacture it. By altering the DNA of someone with PKU, we
could get a patient’s own cells to produce the missing enzyme,
phenylalanine hydroxylase. There are many advantages to not
relying on pharmaceutical companies. Production inside the
body allows for a more targeted response and more accurate
dosages. Furthermore, it removes all chance that a patient would
be unable to access the treatment, such as when the company has
supply chain problems.i

Just as there would be a moral imperative to provide a replace-
ment enzyme therapy for PKU, there would be an imperative
to make safe genome edits which prevent PKU. If it becomes
possible for carriers of the PKU mutation to prevent PKU in their

i It should be noted that the parents of the affected child will need
to access HGE again if they want to ensure any future children
they have are also unaffected. Given the cost associated with
HGE, this may be seen as creating a division between parents
who can and cannot afford it. However, such issues of access
already exist for parents of children with PKU, which can cost
around US$10 000 per year for the medical food and formula.
HGE will certainly be much cheaper than the cost of treating
PKU over a lifetime—it could thus help reduce such inequities.
For a more complete discussions, see Gyngell et al.43 We thank
an anonymous reviewer for making us confront this issue.

future children through HGE, they will have an obligation to use
this technology, in the same way they would have an obligation
to use an enzyme replacement therapy.

Preimplantation genetic testing and HGE
The Nuffield report notes that in all but ‘extremely rare’ (p.44)
cases, monogenic diseases like PKU can already be prevented
through IVF in combination with preimplantation genetic testing
(PGT), with the proviso that ‘it might not be reasonable to
expect sufficient viable embryos with the characteristics sought
to be available’ (p. 46). Let us try to put some numbers around
the cases in which HGE would provide benefits over PGT in
preventing single gene disorders due to a lack of viable embryos.
In 2013 (the last year for which data are available), 18% of IVF
cycles conducted in the UK produced only one viable embryo.18
So, for every 100 couples who go through IVF with the intention
of using PGT to avoid disease, approximately 18 will produce a
single viable embryo. In 2016 (the last year for which there is
data), there were roughly 700 cycles of PGT for genetic disease
in the UK.19 So, every year in the UK, around 126 IVF cycles
are conducted for PGT and only produce one viable embryo.
In these cases, it will not be possible to use genetic selection to
avoid diseases. As people choose to attempt to conceive children
later and later in life, in part for educational and career reasons,
there will be a greater and greater scarcity of embryos.

The most common scenario in which couples use PGT is when
they are both are carriers for recessive conditions. In these cases,
there is a 25% chance that an embryo will carry both copies
of the disease-predisposing mutation. This would imply that
there are 31 cases in the UK per year in which HGE could avoid
genetic disease in an embryo which PGT cannot. However, this is
likely a conservative estimate. When parents are homozygous for
dominant conditions like Huntington’s disease, or cases where
there are multiple undesirable independently sorting variants,
the number of affected embryos will be closer to 50%. One IVF
company is on record as estimate that 48% of embryos which
undergo PGT are affected by a genetic condition,20 although this
will vary clinic to clinic.

Extrapolating from the above numbers would imply that,
worldwide, there are several hundred cases a year where HGE
would be the only option to produce unaffected offspring.

While several hundred cases a year can be considered rare,
it is not negligible. If a public health measure could reduce the
incidence of serious disease by several hundred a year, then we
would have strong reasons to implement it. It would not merely
be ‘morally permissible’ to take such a measure, but something
that we actively ought to do. Of course, in situations of limited
resources we have reasons to prefer interventions that maximise
benefit, but this does not negate the moral reasons we have to
benefit the few.

In sum, the application of HGE to prevent of single gene
disorders is a good application of technology, and something we
have moral reasons to pursue. If it were possible to use HGE to
prevent single gene disorders, there would be a moral imperative
to use it for this purpose. Of course, given this application alone
may not benefit a large number of people, it may not justify
using limited health resources developing HGE which could
be spent on more effective health measures. But HGE also has
potential to prevent far more common causes of disease, as we
will explain in the next section.

polygenic diseases
Most diseases are not the result of just a few genetic changes. They
are the result of many, sometimes hundreds, of genes combining

520 Gyngell C, et al. J Med Ethics 2019;45:514–523. doi:10.1136/medethics-2018-105084

Feature article

together with environmental effects. Such polygenic diseases
are among the world’s biggest killers. Cardiovascular disease is
emerging as the biggest cause of death in the low-income and
middle-income world. Together deaths from chronic diseases in
those under 70 years are responsible for approximately 30% of
all deaths worldwide.21 In addition to causing pain and death
to individuals, chronic diseases place a huge burden on national
health systems, consuming resources that could be used else-
where. One study found that the healthcare cost associated with
treating cardiovascular disease totalled €104 billion annually, for
countries within the European Union.22

We know that there are genetic contributions to chronic
diseases. Genome-wide association studies have identified at
least 44 genes involved in diabetes23; 35 genes involved in coro-
nary artery disease24 and over 300 genes involved in common
cancers.25

It is possible to differentiate between individuals based on
their genetic risk of developing chronic diseases. Using next-gen-
eration sequencing technologies (like whole genome or whole
exome sequencing), polymorphisms occurring across many
genes can be tallied and weighted giving an individual a ‘poly-
genic risk score’ that reflects their genetic predispositions to
develop particular diseases and traits. Individuals can then be
stratified into different risk categories (such as high risk, medium
risk and low risk) based on their polygenic risk score.26

As genome editing technologies can target many genes at one
time, it may become possible to use them to alter an individual’s
polygenic risk score at the embryonic stage,ii and shift individ-
uals from a high-risk category to a low-risk category.iiiiv

Alternatively, it will be possible for individuals who know they
have a high polygenic risk to particular diseases to use HGE to
alter their gametes to ensure they do not pass this high risk on
to their children.

For example, by editing around 27 mutations associated with
coronary heart disease, it would be possible to reduce an individ-
ual’s lifetime risk by 42%27; by editing 12 genetic variants one’s
lifetime risk of bladder cancer could be reduced by almost 75%.28

This application cannot be achieved through current methods
of genetic selection. Say a couple want to use PGT to select for
15 different genes in an embryo, to reduce their likelihood of
cardiovascular disease. Then they would need to create thou-
sands of embryos to make it sufficiently likely that one will have
the right combination at all 15 loci. The chance of the couple
having such an embryo would be <1% with traditional IVF and
PGD.29

Given the massive disease burden caused by chronic diseases,
we have strong moral reasons to develop technologies that
reduce their incidence—whether these operate through genetic
or environmental mechanisms. Imagine scientists develop a new
technology which potentially could be incorporated into exhaust
filters, and would drastically reduce the amount of air pollution
cars emit. In cities where cars are fitted with the exhaust filter,
the incidence of respiratory disease would be decreased by 40%.

ii This first require an embryo to be biopsied at very early stage
(eg, two-cell or four-cell stage), to reduce risks of mosaicism.
iii This possibility was first brought to our attention by Roman
Teo Oliynyk’s unpublished manuscript ‘Could future gene
therapy prevent aging diseases?’44
iv In each of these case, there is a possibility that people in the
low-risk group are overall worse off than those in the high-risk
group, as the genes associated with high risk are beneficial in
some other way. There will be a need for greater research into
the overall effects of particular mutation before this application
of HGE was undertaken.

There are clearly strong moral reasons to develop this tech-
nology and pursue its applications. Developing the exhaust filter
is not merely something it would be permissible to do, but some-
thing that there is an imperative to do. The very same reasons
apply to the development of HGE.

One might respond that the clear difference between this case
and HGE, is that HGE makes heritable changes and will thus
affect future generations. However, air pollution is a known
epigenetic modifier,30 that is, it makes changes to gene expres-
sion which can be inherited by future generations.31 Hence,
reducing air pollution could also affect future generations. Of
course, we need to consider what the long-term effects of any
changes will be. But if the likely effect of a genetic change in one
generation is to reduce risk of disease in future generations, this
seems only to strengthen the case in favour of those changes.

If HGE could make genetic changes which reduce risks of
polygenic disease in current and future generations, there would
be an imperative to use it. Obviously, this application is a long
way away from being plausible, possibly decades. One major
difficulty is that we do not understand polygenic scores well
enough to accurately predict the effects of large-scale changes.
Still, we have moral reasons to develop HGE with the inten-
tion of using them for this purpose. First, it will reduce rates of
premature death and disability due to chronic disease. Second,
the use of HGE to make the highest risk individuals the same
as the lowest risk individuals will be equality-promoting. Third,
using HGE to lower the incidence of chronic disease will also
promote justice. As stated above, health systems spend billions
in resources to treat and prevent chronic disease. Using HGE
in germline cells will probably be a relatively cheap way (in the
proximity of US$20 000) of reducing someone’s susceptibility to
chronic diseases. In a world of limited resources, taking a more
expensive therapy has the opportunity cost of preventing the
treatment of someone else’s disease. Justice requires we choose
the most cost-effective option, other things being equal. If we do
not invest in the most cost-effective option, we harm others who
could use these resources.

enhancement
Just as polygenic scores could in theory be used to reduce rates of
complex disease, they can target complex traits like intelligence.

General intelligence—the ability to learn, reason and solve
problems—is the best known predictor of education and occu-
pational outcomes.32

For decades, it has been known that around 50% of the observed
variation in intelligence is due to genetic factors. A number of recent
large studies have identified many polymorphisms, which help
explain 20% of the heritable variation in intelligence.32

As with complex disease, using the polygenic scores it is
possible to stratify the population into three board groups ‘high
predisposition to high intelligence’; ‘medium predisposition to
high intelligence’ and ‘low predisposition to high intelligence’.
It will become theoretically possible to use HGE to shift indi-
viduals from the low or medium predisposition groups, into the
high predisposition group.

Enhancing based on intelligence using polygenic scores would,
in the words of the Nuffield report, be a form of enhancement
that uses only ‘wild-type’ variants (variants that already exist in
the species) rather than a form of enhancing that goes beyond
what currently exists in the species. In other words, it is a form
of ‘normal range human enhancement’.33 While it may be
possible in the future to enhance intelligence beyond levels that
are currently observed in the species—such forms of enhance-
ment are much less feasible at present.

521Gyngell C, et al. J Med Ethics 2019;45:514–523. doi:10.1136/medethics-2018-105084

Feature article

Imagine a prenatal nutritional programme was developed,
which was predicted to increase intelligence in children born
with low innate predisposition to high intelligence. This would
be seen as a breakthrough. We may soon be able to achieve the
same with HGE.

One of the most intuitive concerns about technologies like
germline engineering is the effect on equality. It is feared that
germline engineering would only be available to the rich, and
that it could widen the gap between rich and poor, adding
biological advantages to already existing social ones. This is an
important and complex issue, faced not just by genome editing
but other goods like education. Ethically, we must take steps to
ensure that the benefits and costs of HGE are evenly or fairly
shared. As recognised by the Nuffield Council, this is not a
reason to ban the technology, or fail to develop it, but a reason
to ensure it is developed responsibly.

However, it is also possible to use HGE to directly improve
equality, as the intelligence example shows. Nature is a biolog-
ical lottery which has no mind to fairness. Some are born gifted
and talented, others with short painful lives or severe disabili-
ties. Currently, diet, education, special services and other social
interventions are used to correct natural inequality. It may be
that targeting combinations of genes is an effective means of
promoting equality in education. For example, there are natural
variations in people’s innate ability to learn how to read. This
often matters little for people in higher socioeconomic groups,
who can afford to spend extra time with their children teaching
them how to read, or employ tutors, etc. However, for those
in lower socioeconomic groups, this predisposition can leave
them illiterate for life. While other measures could in theory no
doubt remedy this inequity of outcomes, evening out the genetic
starting point could prove the most effective way. This method
would have the additional benefit of being passed to future
generations. Genome editing could be used as a part of public
healthcare for egalitarian reasons.

Boosting intelligence and other cognitive traits through HGE
will be an ‘enhancement’, rather than disease prevention. As
noted by the Nuffield report, this does not by itself reduce the
moral reasons we have to pursue it. We have a moral imperative
to use all reasonable means to produce equality in education.

Future generations and intergenerational justice
One of the key interests considered by the Nuffield Council
report is that of future generations. It is crucial that the very long-
term consequences of developing or failing to develop HGE be
considered. Humans often exhibit a cognitive bias towards the
near future and neglect then how our actions may affect the very
far future. This can distort our appraisal of technologies.

The obligations we have to future generations are often
described in terms of intergenerational justice. We owe future
generations the same considerations that we owe our contempo-
raries. We should not unnecessarily deplete the ozone layer, for
example, if this will greatly harm future persons at an only small
benefit to ourselves.

Some worry that by engaging in HGE we risk harming future
generations by negatively altering our genome. There is no doubt
that some application of HGE could harm future generations
(e.g., see discussion of collective action problems in Section 3);
however, such applications are not the inevitable consequences
of the development of HGE, and can be mitigated or avoided.

Moreover, a deep engagement with the interests of future
generations will show why there is strong moral imperative to
develop HGE as a matter of intergenerational justice.34

Modern medicine is removing selection pressures that humans
have historically been subjected to. This is increasing the rate of
random mutations accumulating in the genome and poses a risk
to future generations, as made clear by Michael Lynch in a 2016
article in the journal Genetics:

What is exceptional about humans is the recent detachment
from the challenges of the natural environment and the ability to
modify phenotypic traits in ways that mitigate the fitness effects
of mutations, for example, precision and personalized medicine.
This results in a relaxation of selection against mildly deleterious
mutations, including those magnifying the mutation rate itself.
The long-term consequence of such effects is an expected genetic
deterioration in the baseline human condition, potentially
measurable on the timescale of a few generations in westernized
societies.35

As we develop effective and accessible treatments for disease,
we all but guarantee that the incidence of those diseases will
increase in future generations. This is because mutations which
arise that contribute to those diseases are no longer selected
against.

For example, short sightedness (myopia) has been historically
very rare because it was selected against in hunter-gatherer soci-
eties.36 Modern technologies such as glasses, contact lenses and
Lasik eye surgery help correct such vision problems. In modern
societies, those with naturally poor eyesight have the same
fitness as those who have naturally good eyesight. This allows
deleterious mutations to occur in the genes which influence
vision and not be selected against. Rates of myopia are now over
50% in many countries, making populations increasingly reliant
on technology for this basic biological function. It is likely that
reduced selection against poor vision has caused some of this
increase. While it is easy to correct for myopia, the same process
will allow mutations to accumulate in genes which influence
other biological functions.

The percentage of people who require blood pressure medi-
cation,37 assisted reproductive technologies38 and have genetic
predispositions to deafness, are all increasing. While social
changes play a major role in these changes (eg, poor diet and
sedentary lifestyle, delayed childbearing), biological factors also
play an important part.39 40 In future generations, nearly all
people may be reliant on technologies for these basic functions,
as well as many others.

This will be bad for individuals, who become increasingly
dependent on technologies for basic functions, and need to spend
much of their time and money acquiring a range of therapeutic
goods. Similarly, society will become burdened with spiralling
healthcare costs. Furthermore, the consequence of natural disas-
ters will become much more severe if people are reliant on a
variety of complex technologies whose supply can be disrupted.

Fortunately, there is a way for our descendants to avoid such
a medicalised future. Using HGE, we could edit out disease-
causing mutations as they arise in our genome. This will allow
our descendants to enjoy the same level of genetic health as we
enjoy today.

Of course, many diseases have a lifestyle element—we have
mentioned cardiovascular disease and infertility. Many resist
using biological interventions to treat lifestyle problems. For
example, it seems absurd to genetically modify human beings
to be able to tolerate a diet consisting solely of foods with low
nutritional value.

However, as we have argued, there are biological components
to many contemporary diseases that are worthy of modification.

522 Gyngell C, et al. J Med Ethics 2019;45:514–523. doi:10.1136/medethics-2018-105084

Feature article

Moreover, even if such diseases were entirely lifestyle or social
in origin, which intervention we ought to choose—modifying
the biological, psychological, social or natural environment—
depends on the costs and benefits of the particular interven-
tion, and relevant moral values. For example, it may be possible
to prevent skin cancer by avoiding exposure to the sun, or
by increasing the production of melanin, or by increasing the
capacity of our immune cells to attack skin cancers. Which we
should choose depends on the context.

A common assumption in environmental ethics is that we have
obligations to members of future generations. According to one
principle of intergenerational justice, ‘existing generations ought
not act so as to worsen the position of future generations by
depleting non-renewable resources with no compensatory action
or recompense’.41

It is clear that the use of modern medicine is worsening the
position of members of future generation, by allowing random
mutations to occur to our genome. Fortunately, there is a straight-
forward compensatory action—developing HGE. This is not
something that is merely permissible—but a moral imperative.

governAnCe And publIC ATTITudes
Ultimately, it is up to the public to make decisions about the
ways genome editing can be applied. This is a guiding principle
of liberal democracies. As noted by the Nuffield Council (p.
162), before any changes are made to the laws governing HGE,
broad and inclusive public debate is necessary.

We endorse this view, but wish to add that public debates
surrounding HGE need to be supplemented by public educa-
tion initiatives. Making truly informed decisions about complex
scientific matters requires people to understand science. A recent
study by the Pew Study showed that 86% of Americans with high
scientific knowledge approved of the use of HGE to prevent
diseases that would be apparent at birth. This drops to 56% of
people with low knowledge of science.42 Such research shows
how familiarity with a subject matter shapes one’s view of it.

The Pew Research also shows a great divide between people
who think HGE is permissible to prevent disease (72% for
disease present at birth; 60% for later onset diseases) and those
that think it is permissible for enhancement (18%). This is inter-
esting because as noted by the Nuffield Council, there seems
not to be essential reasons as to why the use of HGE to prevent
disease is different than its use for human enhancement. What
is important is that any use be consistent with promoting indi-
vidual welfare, and does not negatively impact society.

Just as is the case with science, for people to make truly
informed decision on ethical matters, ethical education is
required. People should learn about concepts such as justice,
freedom and well-being from an earlier age, and learn how to
think critically about such topics. Only then can we truly make
informed decisions about technologies like HGE.

ConClusIon
Genome editing technologies are developing rapidly, and so too
is our understanding of their moral implications. The consensus
of various expert bodies on the ethical implications of genome
editing has shifted in response to greater engagement with the
underlying philosophical issues. This has been exemplified by
the recent Nuffield Council report, ‘genome editing and human
reproduction’. Rather than drawing arbitrary lines between
different possible uses of HGE, the Nuffield Council report
engages with the fundamental ethical principles that should

guide our appraisal of genome editing—concerns for individ-
uals, for society as a whole and for future generations.

Nonetheless, we think deep engagement with underlying
ethical issues of HGE yields much stronger conclusions than
those drawn by the Nuffield Council. It will be ‘morally permis-
sible’ to engage in HGE and will be morally ‘required’ in some
instances.

The human genome was created by a blind process of muta-
tion and selection occurring over thousands of generations. This
process had no foresight for the creatures it would produce.
This has resulted in vast natural inequality. The most extreme
examples are single gene disorders, where some people become
destined to a short life with much pain due to random quirks in
their DNA. Others are born with high risks of chronic disease
like heart disease and cancer. We ought to use powerful technol-
ogies like HGE to correct these inequalities and promote human
flourishing. Such actions are moral imperatives.

Contributors Dr CG conducted the initial research, helped drafted the initial
manuscript, made revisions and prepared the manuscript for submission. HBS
researched and summarised the Nuffield Council report and other institutional
statements, and edited the manuscript. Professor JS conceptualised the project,
helped draft the initial manuscript, provided feedback on drafts and made revisions
to the manuscript. All authors approved the final manuscript as submitted and agree
to be accountable for all aspects of the work.

Funding CG, HBS and JS, through their involvement with the Murdoch, received
funding from the Victorian State Government through the Operational Infrastructure
Support (OIS) Programme.

Competing interests None declared.

patient consent Not required.

provenance and peer review Commissioned; externally peer reviewed.

data sharing statement Not applicable.

open access This is an open access article distributed in accordance with the
Creative Commons Attribution 4.0 Unported (CC BY 4.0) license, which permits
others to copy, redistribute, remix, transform and build upon this work for any
purpose, provided the original work is properly cited, a link to the licence is given,
and indication of whether changes were made. See: https:// creativecommons. org/
licenses/ by/ 4. 0/.

reFerenCes
1 Jinek M, Chylinski K, Fonfara I, et al. A programmable dual-RNA-guided DNA

endonuclease in adaptive bacterial immunity. Science 2012;337:816–21.
2 Liang P, Xu Y, Zhang X, et al. CRISPR/Cas9-mediated gene editing in human

tripronuclear zygotes. Protein Cell 2015;6:363–72.
3 Ma H, Marti-Gutierrez N, Park SW, et al. Correction of a pathogenic gene mutation in

human embryos. Nature 2017;548:413–9.
4 Practical Ethics. The fundamental ethical flaw in Jiankui he’s alleged gene editing

experiment. 2018 https://www. apnews. com/ 4997 bb7a a36c 4544 9b48 8e19 ac83e86d
5 News AP. Chinese researcher claims first gene-edited babies. 2018 http:// blog.

practicalethics. ox. ac. uk/ 2018/ 11/ the- fundamental- ethical- flaw- in- jiankui- hes- alleged-
gene- editing-experiment/

6 Lanphier E, Urnov F, Haecker SE, et al. Don’t edit the human germ line. Nature
2015;519:410–1.

7 The Nuffield Council on Bioethics. Genome editing and human reproduction: social
and ethical issues. 2018 http:// nuffieldbioethics. org/ project/ genome- editing- human-
reproduction (Accessed 19 Jul 2018).

8 Gyngell C, Douglas T. Stocking the genetic supermarket: reproductive genetic
technologies and collective action problems. Bioethics 2015;29:241–50.

9 In: Buchanan AE, Brock DW, eds. rom chance to choice: genetics and justice. 8th
printing. Cambridge: Cambridge Univ. Press, 2007.

10 Singer P. The human genome and the genetic supermarket. Free Inq 2001;21.
11 Power RA, Pluess M. Heritability estimates of the big five personality traits based on

common genetic variants. Transl Psychiatry 2015;5:e604.
12 Diener E, Sandvik E, Pavot W, et al. Extraversion and subjective well-being in a U.S.

national probability sample. J Res Pers 1992;26:205–15.
13 Latham RM, von Stumm S. Mothers want extraversion over conscientiousness or

intelligence for their children. Pers Individ Dif 2017;119:262–5.
14 Jung JH, Lee Y, Karsten R. The moderating effect of extraversion–introversion

differences on group idea generation performance. Small Group Res 2012;43:30–49.
15 Gyngell C, Douglas T. Selecting against disability: the liberal eugenic challenge and

the argument from cognitive diversity. J Appl Philos 2016.

523Gyngell C, et al. J Med Ethics 2019;45:514–523. doi:10.1136/medethics-2018-105084

Feature article

16 Cain S. Quiet: the power of introverts in a world that can’t stop talking. 1st pbk. ed.
New York: Broadway Paperbacks, 2013.

17 Bhattacharya K, Wotton T, Wiley V. The evolution of blood-spot newborn screening.
Transl Pediatr 2014;3:63–70.

18 Human Fertilisation and Embryology Authorit. Fertility Treatment in 2013.
19 Human Fertilisation and Embryology Authority. Fertility treatment 2014–2016: trends

and figures. 2018 https://www. hfea. gov. uk/ media/ 2563/ hfea- fertility- trends- and-
figures- 2017- v2. pdf.

20 Medical Service Advisory Committee. Assessment of preimplantation genetic
diagnosis. 2015.

21 World Health Organisation. Preventing Chronic Diseases a vital investment. http://
www. who. int/ chp/ chronic_ disease_ report/ contents/ part2. pdf (Accessed 31 Aug
2018).

22 Leal J, Luengo-Fernández R, Gray A, et al. Economic burden of cardiovascular diseases
in the enlarged European Union. Eur Heart J 2006;27:1610–9.

23 Wheeler E, Barroso I. Genome-wide association studies and type 2 diabetes. Brief
Funct Genomics 2011;10:52–60.

24 Peden JF, Farrall M. Thirty-five common variants for coronary artery disease: the fruits
of much collaborative labour. Hum Mol Genet 2011;20:R198–205.

25 Chang CQ, Yesupriya A, Rowell JL, et al. A systematic review of cancer GWAS and
candidate gene meta-analyses reveals limited overlap but similar effect sizes. Eur J
Hum Genet 2014;22:402–8.

26 Chatterjee N, Shi J, García-Closas M. Developing and evaluating polygenic risk
prediction models for stratified disease prevention. Nat Rev Genet 2016;17:392–406.

27 Mega JL, Stitziel NO, Smith JG, et al. Genetic risk, coronary heart disease events, and
the clinical benefit of statin therapy: an analysis of primary and secondary prevention
trials. Lancet 2015;385:2264–71.

28 Garcia-Closas M, Rothman N, Figueroa JD, et al. Common genetic polymorphisms
modify the effect of smoking on absolute risk of bladder cancer. Cancer Res
2013;73:2211–20.

29 Bourne H, Douglas T, Savulescu J. Procreative beneficence and in vitro gametogenesis.
Monash Bioeth Rev 2012;30:29–48.

30 Maghbooli Z, Hossein-Nezhad A, Ramezani M, et al. Epigenetic alterations and
exposure to air pollutants: protocol for a birth cohort study to evaluate the

association between adverse birth outcomes and global DNA methylation. JMIR Res
Protoc 2017;6:e29.

31 Bohacek J, Mansuy IM. Epigenetic inheritance of disease and disease risk.
Neuropsychopharmacology 2013;38:220–36.

32 Plomin R, von Stumm S, von SS. The new genetics of intelligence. Nat Rev Genet
2018;19:148–59.

33 Kahane G, Savulescu J. Normal human variation: refocussing the enhancement
debate. Bioethics 2015;29:133–43.

34 Powell R. In genes we trust: germline engineering, eugenics, and the future of the
human genome. J Med Philos 2015;40:jhv025.

35 Lynch M. Mutation and human exceptionalism: our future genetic load. Genetics
2016;202:869–75.

36 Cordain L, Eaton SB, Brand Miller J, et al. An evolutionary analysis of the aetiology
and pathogenesis of juvenile-onset myopia. Acta Ophthalmol Scand 2002;80:125–35.

37 Gu Q, Burt VL, Dillon CF, et al. Trends in antihypertensive medication use and
blood pressure control among United States adults with hypertension: the
National Health And Nutrition Examination Survey, 2001 to 2010. Circulation
2012;126:2105–14.

38 Adamson GD, Tabangin M, Macaluso M, et al. The number of babies born globally
after treatment with the assisted reproductive technologies (ART). Fertil Steril
2013;100:S42.

39 Hanevik HI, Hessen DO, Sunde A, et al. Can IVF influence human evolution? Hum
Reprod 2016;31:1397–402.

40 Nance WE, Kearsey MJ. Relevance of connexin deafness (DFNB1) to human evolution.
Am J Hum Genet 2004;74:1081–7.

41 Page E. Intergenerational Justice and Climate Change. Polit Stud 1999;47:53–66.
42 Funk C, Hefferon M. Public Views of Gene Editing for Babies Depend on How It Would

Be Used. Pew Res Cent Internet Sci Tech 2018 http://www. pewinternet. org/ 2018/
07/ 26/ public- views- of- gene- editing- for- babies- depend- on- how- it- would- be- used/
(Accessed 30 Aug 2018).

43 Gyngell C, Douglas T, Savulescu J. The ethics of germline gene editing. J Appl Philos
2017;34.

44 Oliynyk RT. Quantifying the potential for future gene therapy to lower lifetime risk of
polygenic late-onset diseases. bioRxiv 2018.

  • Moral reasons to edit the human genome: picking up from the Nuffield report
    • Abstract
    • Introduction
    • The Nuffield Council’s report on genome editing and human reproduction
      • Principle 1: the welfare of the future person
      • Principle 2: social justice and solidarity
    • Social harms and collective action problems
      • Modified principle 2: social harms
    • Categorical limits and moral imperatives in HGE
      • Single gene disorders
        • Preimplantation genetic testing and HGE
      • Polygenic diseases
      • Enhancement
      • Future generations and intergenerational justice
    • Governance and public attitudes
    • Conclusion
    • References

Computational and Structural Biotechnology Journal 18 (2020) 887–896

journal homepage: www.elsevier.com/locate/csbj

Review

Ethical issues related to research on genome editing in human embryos

https://doi.org/10.1016/j.csbj.2020.03.014
2001-0370/� 2020 The Authors. Published by Elsevier B.V. on behalf of Research Network of Computational and Structural Biotechnology.
This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

⇑ Corresponding author.
E-mail addresses: [email protected] (E. Niemiec), Heidi.ho[email protected]

(H.C. Howard).

Emilia Niemiec ⇑, Heidi Carmen Howard
Centre for Research Ethics and Bioethics, Uppsala University, Box 564, 751 22 Uppsala, Sweden

a r t i c l e i n f o a b s t r a c t

Article history:
Received 15 November 2019
Received in revised form 13 March 2020
Accepted 14 March 2020
Available online 21 March 2020

Keywords:
Genome editing
CRISPR-Cas9
Oocyte donation
Egg donation
Whole genome sequencing
Research ethics

Although the potential advantages of clinical germline genome editing (GGE) over currently available
methods are limited, the implementation of GGE in the clinic has been proposed and discussed. Ethical
issues related to such an application have been extensively debated, meanwhile, seemingly less attention
has been paid to ethical implications of studies which would have to be conducted in order to evaluate
potential clinical uses of GGE.
In this article, we first provide an overview of the debate on potential clinical uses of GGE. Then, we

discuss questions and ethical issues related to the studies relevant to evaluation of potential clinical uses
of GGE. In particular, we describe the problems related to the acceptable safety threshold, current tech-
nical hurdles in human GGE, the destruction of human embryos used in the experiments, involvement of
egg donors, and genomic sequencing performed on the samples of the research participants.
The technical and ethical problems related to studies on GGE should be acknowledged and carefully

considered in the process of deciding to apply technology in such a way that will provide benefits and
minimize harms.

� 2020 The Authors. Published by Elsevier B.V. on behalf of Research Network of Computational and
Structural Biotechnology. This is an open access article under the CC BY license (http://creativecommons.

org/licenses/by/4.0/).

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 887
2. Germline genome editing in the clinic: potential benefits, risks and ethical issues . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 888
3. Current guidelines on the potential use of GGE in the clinic: how do these impact research? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 889
4. Research context of GGE and ethical implications. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 889

4.1. Challenges related to the evaluation of safety and efficacy of GGE . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 889
4.2. Safety issues in germline genome editing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 890
4.3. Use of embryos . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 892
4.4. Oocyte procurement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 892
4.5. Genomic sequencing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 893
4.6. Other issues related to research on GGE . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 894

5. Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 894
CRediT authorship contribution statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 894
Declaration of Competing Interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 894
Acknowledgments. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 894
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 895

1. Introduction

The CRISPR-Cas9 genome editing (GE) system allows for precise,
efficient, relatively cheap, and fast modification of DNA in various
organisms and types of cells. GE has been found to have applica-
tions in many research areas, including, among others, gene func-

888 E. Niemiec, H.C. Howard / Computational and Structural Biotechnology Journal 18 (2020) 887–896

tion studies, gene therapy studies, drug development, and produc-
tion of modified crops in agriculture [1]. In 2015, the first experi-
ment with CRISPR-Cas9 on human embryos was reported; the
authors demonstrated that the GE system performed targeted
cleavage in the b-globin gene, which when mutated causes b-
thalassemia [2]. Following this publication, discussion on prospec-
tive clinical applications of germline genome editing (GGE) in
humans and related ethical issues was ignited. While the term
GGE refers to applications of GE on progenitor cells of gametes,
gametes, or embryos, in this article, we focus our reflection on
GGE conducted on human embryos.

In spite of legal prohibitions on germline genome modification
to establish a pregnancy (i.e. what we consider a clinical use)
enacted in many countries [3], clinical uses of GGE have been dis-
cussed by prominent scientists [5,64]. Many meetings and groups
have been convened to address the ethical questions surrounding
the technique and a voluminous academic literature on this topic
has quickly grown. The majority of the scientific community cur-
rently considers the clinical applications of GGE premature and
many groups have called for a moratorium for such uses [5,6]. Nev-
ertheless, future implementation of GGE in the clinic has not been
excluded as a possibility. Many policy documents, professional rec-
ommendations, and groups of authors either state or seem to imply
that GGE in the clinic could be acceptable if certain conditions are
met. Requirements specified most often include adequate safety
and efficacy of the method, societal debate and/or societal consen-
sus, and appropriate governance [7–11].

Benefits, risks, and ethical issues of potential clinical uses of GGE
have been discussed at length, meanwhile, seemingly less attention
has been given to the questions raised by research on human GGE
which would have to be conducted in order to address safety and
efficacy of the technique. In this article we offer an analysis of eth-
ical issues related to the research context of GGE, including interre-
lated conceptual and technical aspects. To place this analysis in a
broader context, we first present the discussion on potential clinical
uses of GGE in humans focusing on claimed benefits, risks and
selected ethical issues. We then tackle the questions related specif-
ically to research on human GGE concerning: challenges in the eval-
uation of safety and efficacy; technical shortcomings and the safety
risks they pose; problems related to use of embryos in research;
oocyte procurement; and genomic sequencing.

2. Germline genome editing in the clinic: potential benefits,
risks and ethical issues

Although claims of the potential benefits of GGE often relate to
the possibilities of curing or preventing disease (see for example
Gyngell, 2017 [12]), the actual advantage of GGE over available
methods is uncertain and currently appears limited. Firstly, GGE
does not have curative aims per se since there is no patient involved
in the procedure who could be cured1 [13]. Instead, GGE in the
clinic would be coupled with the in vitro fertilization (IVF) technique
in order to create a genetically-related child (for a given couple) who
would then possess a desired trait (e.g. would not be affected by a
given disease).

This approach would be offered in the first instance to couples
who have a combination of genotypes that will result (at least in
theory) in some of their children being affected by a genetic
disease and who are aware of this and would like to avoid passing

1 In one of the approaches of GGE, the embryo can be considered as the subject
which could be cured, that is, when GGE is applied on an existing embryo. This
approach, however, is known to cause mosaicism in embryos, and as such, is unlikely
to be seriously considered for clinical uses. The method of adding the components of a
GE system at the moment of fertilization seems more advantageous in terms of safety
of potential clinical uses.

the disease to their offspring. Importantly, such couples currently
have a number of options available to achieve the desired goal:
1) undergo IVF coupled with preimplantation genetic diagnosis
(PGD) to select ‘‘unaffected” embryos and use them to establish
pregnancy; PGD can detect post-IVF embryos which carry
disease-causing alleles and allows for the selection of embryos
without these (combinations of) alleles; 2) use donor gametes in
IVF; 3) adopt a child; 4) decide to not have a child together.

In the majority of cases, IVF coupled with PGD can be applied2

and GGE does not seem to have a clear advantage over it, as we
explain in the paragraph below. Yet, at least in theory, there are rare
cases where all children of a couple would be affected by a disease
(for example, when one parent is homozygous for a dominant dis-
ease, or both parents are homozygous for a recessive mutation) and
there is no option for PGD and embryo selection. In such situations,
GGE might potentially be the only alternative to have a genetically
related child not affected by a disease, and, as such, it could bring
an advantage over currently available methods [14]. Importantly, it
is not clear whether such couples exist and if they do exist, whether
they would be willing to undergo GGE. Indeed, the theoretical esti-
mation based on available data on prevalence of genetic disorders
in the USA suggests that the clinical demand for GGE would be very
small [15]. For example, the analysis indicates that in the USA in a
given time there is only one couple at reproductive age in which both
persons are homozygous for variants causing cystic fibrosis [15].

Some authors have suggested another benefit of GGE, namely
the possibility to rescue embryos already created in IVF, where
disease-causing variants are detected by PGD and which would
normally be discarded [16,17]. As argued by these authors, such
embryos could undergo GGE and subsequently be transferred in
utero to establish a pregnancy; this, as the authors claim, would
rescue embryos and increase efficiency of the procedure of IVF.
This view of ‘‘rescuing” embryos is, however, challenged by techni-
cal hurdles, for example, the fact that GGE seems to be best applied
at the moment of fertilization to prevent mosaicism, which pre-
cludes the possibility of testing and identifying embryos with a
disease-causing gene variant [14,15].

Another suggested group of applications of GGE has, as a goal,
the general enhancement of (often complex) traits and an
increased resistance to diseases (the latter often regarded as a sub-
set of enhancement, namely disease-prevention enhancement).
The proposals to enhance traits such as intelligence and other com-
plex traits are currently premature due to scientific and technical
limitations. These are primarily related to an incomplete knowl-
edge about the (often limited) genetic contributions to complex
traits, as well as difficulties related to the more complicated
approach that would have to be employed to edit multiple gene
variants. Regarding the suggestion to increase resistance to a dis-
ease, controversially, the first reported case of clinical GGE repre-
sents such an attempt. In November 2018, He Jiankui, a scientist
at the Southern University of Science and Technology in Shenzhen
(China) showed results of his clinical study in which he edited gen-
omes of human embryos that were subsequently used to establish
a pregnancy [18]. He claims that two baby girls with edited gen-
omes were born as a result of this experiment. The goal of this
GGE clinical study was to modify the gene CCR5 with an aim to
increase resistance to HIV in children whose biological father
was affected by AIDS. The study of He Jiankui has been widely crit-
icized not only due to a lack of medical need justifying the research
and the presence of alternative measures to avoid contraction of

2 The risks and impact of PGD on the human organism are more understood than
those of GGE, since PGD has been used in the clinic for nearly 30 years. Yet, PGD raises
ethical issues as well, many of which are common with GGE, for example, the
questions related to the potential impacts on family relationships and more broadly
on the whole society (see the discussion below).

E. Niemiec, H.C. Howard / Computational and Structural Biotechnology Journal 18 (2020) 887–896 889

HIV in that situation, but also due to violation of a number of eth-
ical requirements including not seeking and gaining ethical
approval of the study and not ensuring that adequate informed
consent was obtained [18].

Potential benefits of GGE may be discussed, as argued by some
authors, from a broader perspective which includes long-term ben-
efits on a population level such as a decrease in prevalence of
genetic diseases [19]. Importantly, such a prognosis assumes not
only that the technique will be efficient and safe, but also that its
uptake will be high, neither of which can be taken for granted. Even
if the prospect of reducing the frequency of diseases in a popula-
tion was realistic, such an advantage and related economic gains
should be weighed against harm which may be caused to individ-
uals, on which we elaborate below.

Importantly, in both types of GGE applications (to obtain a child
without a disease-causing gene and for enhancement), the
approach would be used to satisfy a desire to have a genetically-
related child with a chosen feature. It is important to acknowledge
that as such, GGE does not fulfill a therapeutic purpose per se, since
the technique is being used to create the child (with a specific trait)
and not to treat an already existing child. As such, the rationale of
fulfilling the desire for a biologically related child with trait ‘‘X”
funded by (public) healthcare services can be challenged, as has
been done with other reproductive technologies [20].

Furthermore, GGE in the clinic poses many additional ethical
problems and risks. First, proceeding with first-in-human uses of
GGE, unlike in the case of other ‘‘standard” clinical trials, will
involve creating a research participant, who would be involuntarily
involved in a research project. Importantly, the main expected ben-
efit of such a trial would be, as already mentioned, satisfying the
parents’ desire to have a genetically-related child not affected by
a given disease. To achieve this goal, a child would be involved in
an experiment with relatively high uncertainties, risks, and poten-
tially irreversible consequences. Furthermore, the experimentation
and consequent need for monitoring may result in such a person (as
well as their children and grandchildren etc.) taking part in a life-
long experiment or clinical trial essentially without having given
consent. Such a situation raises fundamental questions about the
autonomy and best interest of a child. Smolenski (2015) suggests
that a decision to place a child in such a situation is not similar to
other decisions parents routinely make for their children (e.g.
because it is irreversible) and may extend the limits of parental lib-
erty and decision making over a child to unacceptable levels [21].

Additionally, there are questions about the impact of such an
intervention on child-parent relationships. In particular, the risk
of reducing these relationships ‘‘to an overt commercial transaction”
has been discussed [22], as well as the risk of making ‘‘parents less
tolerant of perceived imperfections or differences within their fami-
lies”, and of eroding ‘‘parental instincts for unconditional acceptance.”
[9] Moreover, the efforts to obtain a child with desired traits, raise
concerns about the rise of consumer eugenics ‘‘in which affluent
parents seek to choose socially preferred qualities for their children”
[23]. An increased focus on avoiding certain traits and increasing
others may also lead to a decrease of compassion and general
acceptance of disabled persons in society.

While not specific to GGE, ethical concerns about the creation
and destruction of supernumerary embryos created during IVF but
not used to establish a pregnancy may be also debated. We discuss
ethical positions on the moral status of embryos in the section
below.

3. Current guidelines on the potential use of GGE in the clinic:
how do these impact research?

The theoretical or potential benefits of GGE in the clinic
currently appear limited and uncertain (especially given the

availability of alternative approaches), while the potential risks
including the ethical problems it raises are numerous. There are
authors who have been explicit about their view that GGE should
not be pursued [24,25]. There seems to be, however, many recom-
mendations issued by professional organizations, policy-making
groups, and other groups of authors (not representing any one
group) that state that GGE is not currently permissible, but that
such applications might be permissible provided certain conditions
are met [7–11].

What conditions should be met to pursue the (currently theo-
retical) limited benefits of GGE in the clinic according to the docu-
ments mentioned above? Most common recommendations refer to
the need to address the current uncertainties surrounding the
science, especially safety and efficacy concerns, and consequently
to continue research on GGE [8–11]. The other, often expressed
conditions are of societal debate and/or societal consensus on the
issues of clinical GGE and appropriate oversight [7–11].

In the remainder of this article we focus on the research context
of GGE, as it is being supported, especially as stated above to
address further uncertainties surrounding the science of GGE and
especially its safety and efficacy (which we consider herein as
including the basic science of how well genome editing tools are
working on target edits).

4. Research context of GGE and ethical implications

4.1. Challenges related to the evaluation of safety and efficacy of GGE

As mentioned above, many policy documents and recommen-
dations issued by professional groups as well as by individual
authors state that safety and efficacy of GGE must be further stud-
ied and evaluated in order to consider its potential implementation
in the clinic. However, less attention has been given to trying to
answer the question: what is an acceptable threshold of safety
and efficacy to proceed with clinical trials? And, exactly what kind
of studies should be conducted in order to provide satisfactory evi-
dence? While we do not offer the answers to these questions, we
show in this subsection how these aspects are particularly prob-
lematic for GGE research and how this is recognised by different
professional and policy groups.

Importantly, the above-mentioned questions arise in a broader
context of a benefit-risk evaluation, which should precede any
decision on initiation of a clinical trial. The classic and widely
accepted medical ethics document, the Declaration of Helsinki
states:

‘‘Medical research involving human subjects may only be con-
ducted if the importance of the objective outweighs the risks and
burdens to the research subjects. (. . .) All medical research involv-
ing human subjects must be preceded by careful assessment of pre-
dictable risks and burdens to the individuals and groups involved in
the research in comparison with foreseeable benefits to them and
to other individuals or groups affected by the condition under
investigation.’’ [26]

In this specific context of GGE, a question may be posed: could
the importance of the objective of the trial, that is to create a
genetically related child not affected by a given condition ever out-
weigh and justify exposing a child to the risks and uncertainties
involved in GGE? Or, in other words, should the desire to have a
genetically-related child be satisfied in spite of the harm the pro-
cess can cause?

Of note, a positive answer to this question has already been
given in the context of other reproductive technologies such as
IVF, PGD, and more recently, nuclear genome transfer (also known
as a mitochondrial replacement), which are now used in the clinic.

890 E. Niemiec, H.C. Howard / Computational and Structural Biotechnology Journal 18 (2020) 887–896

The acceptance and clinical uses of nuclear genome transfer, which
is considered by some as the first germline genome modification in
the clinic,3 may, to some extent, pave the way to the potential
implementation of GGE in the clinic. In this context, it is important
to recognize that these approaches raised safety concerns and ethical
issues, which were deemed tolerable as clinical uses were allowed.
Flaws in the process of policy-making on these technologies have
been discussed by commentators. For example, with regard to
nuclear genome transfer, Drabiak explains:

“The HFEA Review acknowledged the potential for complications
pertaining to safety and efficacy, but unilaterally disregarded what
the scientific community has described as numerous substantial
barriers.” [27]

These concerns should prompt us to increase vigilance over the
processes leading to decisions on the uses of GGE.

The challenges around safety assessments pertain not only to
the fact that GGE in the clinic would not be, strictly speaking, ther-
apeutic and involve the creation of a child, but also to the problem
of long-term impacts from the irreversible changes introduced,
both on the developing organism of a ‘‘genome-modified” person
as well as on her descendants. The American Society for Gene
and Cell Therapy and the Japan Society of Gene Therapy address
this issue with the following:

‘‘The requirement that the results of an experiment be susceptible
to analysis and characterization before further applications are
undertaken cannot be met with human germ-line modification
with current methods, because the results of any such manipula-
tion could not be analyzed or understood for decades or genera-
tions—a situation incompatible with ethical imperatives and with
the scientific method.’’ [28]

In spite of the open question of whether a thorough assessment
of the technique is possible at all, efforts have been taken to pro-
vide some guidance regarding what evidence is needed. The Amer-
ican Society of Human Genetics (2017) specified that the

‘‘(. . .) minimum necessary developments should include the
following:
-Definitions of broadly acceptable methodologies and minimum
standards for measuring off-target mutagenesis.
-Consensus regarding the likely impact of, and maximum accept-
able thresholds for, off-target mutations.
-Consensus regarding the types of acceptable genome edits with
regard to their potential for unintended consequences.”[9]

In the summer of 2019, the International Commission on the
Clinical Use of Human Germline Genome Editing (convened by
the UK’s Royal Society and the US National Academies of Sciences
and Medicine) issued a call for evidence, that is, a request directed
to experts in the field of genome editing to answer questions such
as

‘‘If there were to be an appropriate use case for human germline
genome editing, what evidence would be needed to proceed to
first in human use?”

‘‘What is the status of the technology for validating that a correct edit
(on target characterization) has been made and that unintended
edits (e.g., off target effects, mosaicism, etc.) have not occurred in a

3 While technically, nuclear genome transfer does imply a change in the
genome since there is no longer the same combination of nuclear DNA and
mitochondrial DNA – lumping both genome editing of specific nucleotides with the
much coarser exchange of mitochondria (and cytoplasm) is somewhat misleading.
Indeed, these two proceedures do not work at all on the same scale or entail the same
type of changes at the DNA level and consequently the same types of risks and
benefits. It has been suggested that by grouping them in the same category, the
acceptance of one will open the door to the acceptance of the other.

range of cell and tissue types? If possible, please provide evidence
drawn from work on early stage human embryos.” [29]

The Commission has, as its main goal, the development of ‘‘a
framework for considering technical, scientific, medical, regulatory,
and ethical requirements for germline genome editing, should society
conclude such applications are acceptable.” [58]. In addition, they list
specific requirements (or tasks) to identify appropriate pre-clinical
approaches to assess on- and off- target effects, mosaicism, and
long-term side effects, among other thing [58].

In the next section, we aim to provide a general overview of the
selected technical aspects of the studies which raise important eth-
ical implications. Importantly, studies addressing safety and effi-
cacy of human GGE may be performed on animals or human
embryos. Although we do not dismiss the importance of address-
ing ethics of the former, due to the limited space herein we focus
on the problems raised by research on human embryos.

4.2. Safety issues in germline genome editing

In Table 1 we list studies published in English that have used
GGE in human embryos. Although the goals of the experiments
vary, all these studies may, to varying degrees, provide information
on the functioning of genome editing in embryos. We may distin-
guish research which alludes to GGE in the clinic more directly,
that is, studies in which disease-causing variants have been cor-
rected with GE [30–32] or a study in which an allele of the gene
CCR5 associated with a resistance or slower progression of HIV
infections has been introduced [33]. There are also studies which
aimed to show feasibility of a given approach, but unlike the pre-
vious group of experiments, they did not focus on correcting
disease-causing genes or variants relevant to disease resistance
[34,35]. Furthermore, research examining the role of a gene in
embryogenesis using GE has been published [36].

The studies presented in Table 1 reveal, among other aspects,
various technical hurdles, which render potential clinical applica-
tions of GGE unsafe. The main technical problems with safety
implications for potential clinical GGE in human embryos revealed
therein include:

1) mosaicism, a situation where not all cells of an embryo/or-
ganism have the same DNA – in this case the desired DNA
modification;

2) off-target effects, where unintended changes in the genome
outside of the targeted sequence occur;

3) on-target undesired modifications introduced within or next
to the targeted locus [16].

All of these phenomena may have negative and difficult to pre-
dict effects on an organism. Some strategies exist to address these
problems, albeit to varying degrees. For example, a study by Ma
et al. (2017) has shown that injection of CRISPR-Cas9 system at
the moment of fertilization reduces mosaicism [30]. Ma and col-
leagues also explicitly admitted technical shortcomings of their
approach, in particular occurrence of on-target effects:

‘‘Despite remarkable targeting efficiency and high HDR [homology-
directed repair] frequency, some CRISPR–Cas9-treated human
embryos demonstrated NHEJ [non-homologous end joining]
induced indels and thus would not be suitable for transfer. There-
fore, genome editing approaches must be further optimized before
clinical application of germline correction can be considered.” [30]

An alternative GE approach has been developed and tested in
human embryos, whereby, unlike in CRISPR-Cas9 system, sequence
is not cut, but a base pair is directly modified [32,34,36]. As such,
this base editing approach does not involve a repair mechanism

Table 1
Studies conducted using germline genome editing on human embryos. This table is based on Table 1 included in the article by Niemiec and Howard (2020) published under
Creative Common Attribution Noncommercial License (https://creativecommons.org/licenses/by-nc/4.0/).

Year Authors Title Type of modification introduced Type of embryos used

Clinic 2018 He Jiankui (unpublished,
presented at the
International Summit on
Human Gene Editing, Hong
Kong, 2018)

Developing a CCR5-targeted
gene editing strategy for
embryos using CRISPR/Cas9

Modification of CCR5 gene to
increase resistance to HIV
infections

Embryos created with sperm of a man who
contracted AIDS. Two embryos were
implanted to establish a pregnancy which
resulted in the live birth of twin girls

Research 2019 Li et al. Efficient generation of
pathogenic A-to-G mutations in
human tripronuclear embryos
via ABE-mediated base editing

Single nucleotide substitutions
in a few genes (base editing)

Tripronuclear embryos created in clinical IVF
procedures

2019 Zhang et al. Human cleaving embryos
enable robust homozygotic
nucleotide substitutions by base
editors

Single nucleotide substitutions
in a few genes (base editing)

Embryos created using immature oocytes
from patients undergoing clinical IVF
procedures and sperm from donors
Tripronuclear embryos obtained in clinical
IVF procedures

2018 Zeng et al. Correction of the Marfan
syndrome pathogenic FBN1
mutation by base editing in
human cells and heterozygous
embryos

Correction of a mutation in FBN1
gene causing Marfan syndrome
by base editing

Embryos created for the purpose of research
using immature oocytes from women
undergoing IVF procedures

2017 Zhou et al. Highly efficient base editing in
human tripronuclear zygotes

Single nucleotide substitutions
in a few genes (base editing)

Tripronuclear embryos

2017 Li et al. Highly efficient and precise base
editing in discarded human
tripronuclear embryos

Single nucleotide substitutions
in a few genes (base editing)

Tripronuclear embryos created in clinical IVF
procedures

2017 Ma et al. Correction of a pathogenic gene
mutation in human embryos

Correction of a mutation that
causes hypertrophic
cardiomyopathy

Embryos created for the purpose of research
(over 100 embryos were created) using
oocytes and sperm procured specifically for
research

2017 Tang et al. CRISPR/Cas9-mediated gene
editing in human zygotes using
Cas9 protein

Correction of a mutation in HBB
gene causing b-thalassemia and
a mutation in G6PD gene related
to an enzyme deficiency

Embryos created for the purpose of research
using immature oocytes and sperm from
patients undergoing clinical IVF procedures
Tripronuclear embryos created in clinical IVF
procedures

2017 Liang et al. Correction of b-thalassemia
mutant by base editor in human
embryos

Correction of a mutation in the
HBB gene which causes b-
thalassemia (base editing)

Embryos obtained by somatic cell nuclear
transfer; immature oocytes were donated by
women undergoing IVF procedures

2017 Fogarty et al. Genome editing reveals a role
for OCT4 in human
embryogenesis

Study of the function of the
pluripotency transcription factor
OCT4 during embryogenesis

Surplus embryos created in clinical IVF
procedures

2016 Kang et al. Introducing Precise Genetic
Modifications into Human 3PN
Embryos by CRISPR/Cas-
Mediated Genome Editing.

Introduction of an allele of the
gene CCR5 associated with a
resistance or slower progression
of HIV infections

Tripronuclear embryos created in clinical IVF
procedures

2015 Liang et al. CRISPR/Cas9-Mediated Gene
Editing in Human Tripronuclear
Zygotes

Modification of HBB gene, which
when mutated causes b-
thalassemia

Tripronuclear embryos created in clinical IVF
procedures

E. Niemiec, H.C. Howard / Computational and Structural Biotechnology Journal 18 (2020) 887–896 891

which could introduce undesired on-target modifications. Yet, this
technique is not only relatively new and not thoroughly tested, but
also was shown to introduce off-target modifications [59,60].

Not only does the elimination of these undesired events in the
genome pose challenges, but so do the very techniques used to
attempt to detect these changes. In order to control the effects of
genome editing, an embryo has to be biopsied, DNA isolated and
sequenced. Since the amount of isolated DNA in such context is rel-
atively low, DNA has to be pre-amplified which introduces risks of
errors [16]. Furthermore, there are problems with an adequate ref-
erence genome sequence to which the sequence of an edited
embryo could be compared. The DNA sequence of an edited

embryo can be compared to the parents’ sequences and reference
genome (assembled, representative for humans whole genome
sequence); in this case, however, potential off-target modifications
have to be distinguished from other variations among genomes
[16].

Besides the technical shortcomings revealed by the studies,
there are also broader problems related to cell physiology and
genomic interactions, which may be relevant to safety of potential
clinical uses of GGE. For example, epigenetic effects may occur,
which, as the American College of Medical Genetics and Genomics
explains, ‘‘may alter normal patterns of gene expression in some tis-
sues” [37]. Additionally, we may inquire about what would be

4 For an overview of arguments related to moral status/value of human embryos,
see for example, the report of the Nuffiled Council on Bioethics on the related topic
[41].

5 It may be argued that 14-days rule protects more developed embryos from use
and destruction in research, however, it does so by requesting destruction of less
developed embryos.

892 E. Niemiec, H.C. Howard / Computational and Structural Biotechnology Journal 18 (2020) 887–896

the impact of a given DNA modification more broadly on the func-
tioning of a given organism which may vary depending on, for
instance, environmental factors. There are also questions about
multiple (sometimes not yet discovered) functions of edited genes.
Given the complexity of the human genome and the variability
among individuals as well as epigenetic mechanisms (which are
not yet completely understood), it is difficult or even impossible
to predict all the consequences of GGE on an organism (and on
future generations). Furthermore, the effects of each modification
may be different, depending on what gene is edited, its function
and location on the chromosome, as well as the type of editing
used. Additionally, the impact of factors such as medium used to
culture the embryos and the way of administrating the GE system
may be considered.

Above we presented a general and non-exhaustive description
of technical hurdles in GGE. Indeed, there are many nuances of
methodological aspects, which may have impact on the technical
outcome of GGE and its potential safety in humans, which we do
not address here. To thoroughly evaluate the current state-of-art
of GGE with regard to technical issues, safety, and efficacy, a sys-
tematic literature review should be conducted (of research con-
ducted both in human cells and animals). Importantly, such a
review can only be meaningful if so called ‘‘unsuccessful” experi-
ments are also published and accessible [38]. In addition to techni-
cal shortcomings, there are questions about methodology and
reproducibility of the studies.

Furthermore, the process of answering whether some technical
deficiencies and uncertainties are acceptable in the pursuit of the
potential limited (added) benefits of clinical GGE will involve dif-
ferent value judgments by stakeholders with a priori different val-
ues, priorities, and opinions. This brings us back to the more
fundamental questions of whether safety issues can be fully
addressed and what level of uncertainty we are able or willing to
accept.

Notwithstanding the limitations of this overview, it is clear that
there remain a number of important technical problems (and
hence with safety) with GGE and currently there are no straightfor-
ward solutions to surmount them. We may expect that many stud-
ies would have to be conducted to address these issues, both on
human embryos and animals. Below we attend to the ethical prob-
lems raised by the former group of experiments.

4.3. Use of embryos

The use of embryos in research, including the studies listed in
Table 1, raises a number of ethical aspects. One commonly dis-
cussed ethical issue is that related to the destruction of human
embryos. We can distinguish the following types of embryos used
in GGE research based on their source:

1) so called supernumerary or surplus embryos, which are left
over after clinical IVF procedures,

2) embryos created specifically for the purpose of research
using gametes left over (surplus) from IVF,

3) embryos created specifically for the purpose of research
using gametes procured specifically for research.

Furthermore, we may distinguish viable and non-viable
embryos. The former term means that embryos are considered to
be able to develop “normally” and result in a live birth when
implanted into a woman’s uterus; non-viable embryos (e.g.
tripronuclear embryos) are considered unable to develop “nor-
mally” and to result in a live birth if implanted into a woman’s
uterus. Of note, from the scientific point of view, viable embryos
are, in general, more advantageous than non-viable embryos, as
the latter type possess abnormalities impacting their functioning.

Since human embryos are humans in the earliest developmen-
tal stage, their destruction raises ethical questions. While the full
discussion behind different considerations of the human embryo
is beyond the remit of this article, we can distinguish broadly,
three main positions in discussions on the moral status/value4 of
the human embryo:

a) human embryos have the same moral status as any other
born human;

b) human embryos have some moral status/value, but not the
same as a born human; there are variations within this view,
for example, some say that moral status or value of embryos
increases during their development;

c) human embryos have no moral status or their moral status/-
value is the same as of any other type of human cells.

If we assume the position that human embryos have the same
moral status as persons, GGE experiments are considered unac-
ceptable. Research on embryos is prohibited in some countries
(e.g. Austria, Germany, Italy, Poland), however, the formulations
of these legislations differ (https://hpscreg.eu/map). Furthermore,
two important public agencies that fund research, the European
Commission and National Institutes of Health in the USA, do not
fund research projects involving the destruction of embryos
[39,40].

The second view on the moral status of embryos has been
entrenched in the legislation of more countries whereby research
on embryos is allowed with some restrictions (that is, some protec-
tion is granted to embryos) (see https://hpscreg.eu/map). An
example, although debatable,5 of such protection is the so called
14-day rule stating that research can be conducted only until 14 days
after fertilization; after this time, embryos have to be destroyed [41].
Furthermore, if assumed that human embryos have some moral sta-
tus/value, the type of research they are used in and their number
may be discussed, and conditioned, for example, upon the expected
benefit of research. Moreover, some laws make a distinction
between using embryos left over from IVF procedures and those cre-
ated specifically for research. The Oviedo Convention, for instance,
states that the creation of embryos specifically for research is not
permissible [42].

In the studies on GGE, both viable and non-viable embryos have
been used; in one study, by Ma et al. (2017) embryos were created
specifically for the purpose of research. Notably, the usual number
of embryos used in such procedures can be high. For instance, in
the study conducted by Ma et al. (2017), over 100 human embryos
were created and destroyed. The continuation of such experiments
would certainly multiply these numbers. We believe that this eth-
ical issue related to the implementation of GGE in the clinic, should
be acknowledged and discussed, and society informed about it
when public discussions are conducted.

4.4. Oocyte procurement

The study of Ma et al. (2017) showed that the strategy of
administering CRISPR-Cas9 system at the point of fertilization is
advantageous as it reduces the mosaicism in embryos [30]. To fol-
low such a strategy, embryos have to be created specifically for
research. Furthermore, since the authors aimed to ‘‘correct” a
specific gene mutation, it was essential to obtain, and, in this case,
create embryos with exactly this genotype. Creating embryos for

E. Niemiec, H.C. Howard / Computational and Structural Biotechnology Journal 18 (2020) 887–896 893

GGE research raises questions about the source of gametes used,
particularly human eggs. While supernumerary oocytes and sperm
from IVF procedures can be used, there may be limited availability
of gametes with desired genotypes. If a scientist would be inter-
ested to study embryos heterozygous for a given (disease-
causing) gene, an alternative approach could consist of deriving
sperm from an affected man and using wild types oocytes donated
as surplus after IVF, as was done in the study of Zhang et al. (2019)
[35]. Oocytes can also be procured from women specifically for
research, which raises more profound ethical issues.

Within the studies on human GGE listed in Table 1, five exper-
iments involved egg donation: in one study, oocytes were procured
specifically for research [30], in the remaining studies, immature
oocytes obtained in clinical IVF procedures were used
[31,32,35,61]. Although immature oocytes retrieved in IVF proce-
dures are usually discarded, it has been shown that some of such
oocytes can undergo in vitro maturation, be fertilized and develop
into embryos and live births [62]. Consequently, it may be ques-
tioned whether all immature oocytes can be considered useless
in the context of clinical IVF and whether women should be invited
to donate such eggs. When donation of mature and “healthy”
oocytes obtained during IVF process is considered, such questions
seem even more pertinent. Ballantyne and de Lacey explain that:

‘‘Women having IVF have the option of fertilizing all the eggs
retrieved and freezing spare embryos for future use. When women
do not wish, for personal reasons, to freeze embryos, freezing spare
eggs for fertilization and transfer in future attempts is a viable
option. If the woman donates some of her eggs for research and
her initial embryos fail to implant, she must undergo additional
cycles of egg retrieval that may otherwise have been unnecessary.
Although the personal costs of egg donation will differ for each indi-
vidual woman, depending on her specific fertility problem, there
are currently no cost-free eggs. Due to the uncertainties surround-
ing egg fertilization, embryo implantation, successful pregnancy,
and the desire for future children; infertile women always are being
asked to donate a potentially valuable resource. Therefore, it is not
the case that women are being asked to donate ‘‘spare” or ‘‘surplus”
eggs. Rather, they are being asked to donate eggs that are a poten-
tially valuable personal resource that has typically required signif-
icant investment of time, money, discomfort, and anxiety to
produce.” [43]

Egg donation specifically for the purpose of research raises
additional concerns. Oocyte procurement is a physically invasive
procedure, which involves ovarian suppression, followed by ovar-
ian stimulation and a surgical procedure of oocyte retrieval. The
whole process involves not only many inconveniences, but also
risks to the physical health or even the life of the woman involved.
Discomforts include frequent visits to doctor’s office, injections of
medicines, and undergoing a surgical procedure under sedation.
Most frequent side effects include nausea, irritability and head-
aches, among others [63]. Ovarian hyperstimulation syndrome is
a rarer, yet more serious complication, which may, in the worst-
case scenario, result in death [44]. Notably, in the consent forms
used in the study by Ma et al. (2017) death was mentioned three
times in the context of different procedures6. Additionally, Schnei-
der et al. have drawn attention to long-term potential risks for
oocyte donors, such as breast cancer, which has not yet been ade-
quately studied [45].

The question of whether experiments involving the procedure
of oocyte retrieval are ethically acceptable is not new and has been
discussed in the context of stem cell research. Magnus and Cho

6 The forms were provided at our request by one of the co-authors of the study of
Ma et al. (2017). To our knowledge they are not available online.

highlight the disproportion between the risks involved and poten-
tial benefits in this context:

‘‘These women are not pursuing the procedure for any reproductive
or medical benefit to themselves; rather, they are exposing them-
selves to risk entirely for the benefit of others. If we were to think
of them as simply clinical patients, their physician’s fiduciary obli-
gations would seem to require counsel against undergoing such a
procedure for no benefit.” [46]

Importantly, in the case of GGE, which is strictly speaking not a
therapeutic procedure, as explained above, the ratio of benefits to
risks is even more difficult to accept.

Despite serious arguments against studies involving oocyte
retrieval, such research on stem cells has been conducted and
accepted by professional societies as permissible [47,48] raising
other sets of questions on how women should be engaged in such
studies. In particular, a key issue is on ensuring that consent to
research is informed and women are adequately compensated for
the risks and inconveniences to which they are exposed. Sums of
a few thousand dollars in compensation (in the study of Ma et al.
(2017) of 5000 US dollars) do not seem inflated when we consider
the serious risks involved in egg procurement. On the other hand,
such amounts of money may constitute undue inducement to
some women who are suffering various degrees of financial hard-
ship or socio-economic disadvantage. Indeed, it may be difficult to
avoid undue inducement and at the same time offer fair
compensation.

As explained above, studies involving the creation of human
embryos seem to be currently advantageous over other approaches
from scientific point of view. Until now (to our knowledge) only
one study involving embryo creation and egg donation for purpose
of research has been performed to study GE, yet, if the goal of
addressing safety and efficacy of GGE is to be pursued, we may
expect more of such studies. This will entail exposing many
women to risks. We believe that these aspects should be recog-
nized and acknowledged in the discussions on GGE.

4.5. Genomic sequencing

As mentioned above, in order to verify whether an embryo has
been edited in the desired way and to assess for off-target events,
genome sequencing of embryonic cells is conducted. The entire
genome of gamete donors is also sequenced (e.g. from blood) in
order to act as a reference sequence. In these ways, researchers also
obtain a lot of genomic sequencing information from gamete
donors. One may ask if gamete donors are aware that all (or a large
part of) their DNA will be sequenced and know the implications of
this fact. Indeed, our recent study of informed consent forms used
in the study of Ma et al. (2017) shows that genomic sequencing has
not been explicitly mentioned in the forms, which raises questions
about whether research participants were adequately informed
about this important aspect of the research [49].

Importantly, the use of whole genome sequencing (WGS) and
whole exome sequencing (WES) of research subjects in the “regu-
lar” (i.e. non GGE) genomic research context already raises impor-
tant ethical, legal and social issues (ELSI). These ELSI commonly
revolve around issues of privacy and confidentiality of the genomic
data; how to obtain fully informed consent from research subjects;
the possibility of subjects to withdraw from research; as well as
issues regarding the return of research results, including the right
not to know. While going into depth into each of these issues is
beyond the scope of this article (see Pinxten and Howard 2014
for a review) [50], we provide an example of the complexity of
some ELSI by explaining the challenges of obtaining informed con-
sent below. We also highlight that in general, the ELSI surrounding

894 E. Niemiec, H.C. Howard / Computational and Structural Biotechnology Journal 18 (2020) 887–896

the use of WGS and WES in research is still being debated and the
logistics and policies needed to be implemented to offer responsi-
ble genomic sequencing are still being developed. Hence the use of
an already ethically challenging approach such as genomic
sequencing within the highly ethically contentious context of
GGE only serves to exacerbate the ELSI, especially where gamete
donors are concerned.

The difficulties related to obtaining truly informed consent for
genomic sequencing (even outside of the context of GGE) have
been discussed [51]. A requirement for informed consent is com-
municating relevant possible risks and benefits to research sub-
jects. With genome sequencing, the amount of data generated,
and the different results that could be obtained above and beyond
the results for the initial reason to sequence, as well as the uncer-
tainties still surrounding sequencing [52] are so great that it is not
obvious that all this information can be transmitted explicitly and
meaningfully during the informed consent process. For example,
data about family relations, or about DNA variants that may cause
susceptibility to other diseases (other than the one that triggered
the initial need for sequencing) may be revealed by the sequence
information. Questions then arise about which types of informa-
tion should be returned to research participants, how it would be
done and by whom? Ideally, the consent process should include
all these possibilities, but ironically this is likely to result in very
long consent forms, the length and volume of information alone
sometimes contributing to confusion and lack of understanding.
Consent procedures should also address the security of data stor-
age and for how long data will be kept and if it will be used for sec-
ondary purposes or shared with any third parties nationally or
internationally. Clearly this is a lot of information and providing
it in a way that truly supports decision making (instead of simply
being an exchange and signature of forms) is a challenge.

4.6. Other issues related to research on GGE

In addition to the issues described above, there are other con-
cerns related to the research which would have to be conducted
to introduce GGE to the clinic. As mentioned earlier, in addition
to the studies on human embryos, research on animals would be
necessary to assess the impact of embryonic DNA modifications
on the development and functioning of an adult organism and
future generations. Such research, similarly to the studies con-
ducted in humans would likely involve oocyte retrieval, in vitro
fertilization, and implantation of the fertilized eggs to establish
pregnancy; these procedures may cause pain and distress to ani-
mals. Although the problem of harm caused to animals in research
is neither new nor unique for the studies on GGE, the question of
whether the objective of the study can justify the harm caused to
animals seems to be particular in the context of GGE given the
problems its potential clinical use entails.

Research on animals is widely (yet not universally) accepted if
the experiments are scientifically justified and follow the frame-
work of three ‘‘Rs” relating to the reduction of the number of ani-
mals in the experiments, refinement of the protocols (so that
animals’ suffering is minimized) and replacement with other
approaches not involving animals where possible (see, for exam-
ple, the Directive of the European Union [53]). Each study involving
GGE in animals will have to follow the local legislation and guide-
lines for this kind of research.

Another ethical aspect related to GGE research is the opportu-
nity costs of funding this type of research instead of other types,
as Baylis puts it in her recent book:

‘‘What other valuable research is not being done as a result of this
investment? (. . .) what other medical needs are being
underfunded?” [54]

We could also ask, more concretely, whether research efforts,
talents, and funds should not be directed to the studies investigat-
ing somatic GE, which does not involve so many contentious ethi-
cal aspects as GGE, yet, is a promising approach to treat or even
cure a number of genetic diseases [55].

5. Conclusions

Despite the limited and uncertain medical need for clinical GGE,
numerous ethical issues and risks related to such an application,
and current prohibitions of germline genome modification in many
countries, uses of GGE have been proposed and discussed. Further-
more, in some circles, it seems that the focus of the debate has
recently shifted from the question of if GGE should be introduced
to the clinic to how it should be done [56,57]. Indeed, influential
actors such as the US National Academy of Sciences have under-
taken efforts to establish a framework for a potential translational
pathway for clinical GGE [58].

Not only does the potential clinical use of GGE raise ethical
issues, but so does the research context of this approach, including
all the studies that would be required to evaluate GGE before its
potential introduction into the clinic. Firstly, it can be questioned
whether safety of the procedure could ever be sufficiently evalu-
ated in the non-clinical studies since the effects of GGE on a devel-
oping human organism cannot be fully predicted. Related to this,
there are questions about the degree to which numerous technical
and scientific hurdles would have to be addressed and the type of
evidence needed from the studies both on human embryos and
animals. GGE research using embryos raises questions about the
moral status of the human embryo, the involvement of egg dona-
tion, which entails serious risks to women, as well as the ethical
issues related to whole genome sequencing. To evaluate the effects
of germline DNA modification on a developing and adult organism
as well as on future generations, research on animals would have
to be involved, which raises ethical concerns as well. Last but not
least, we may question whether continuation of such research is
the best allocation of the resources, both in terms of funds and
personnel.

We argue that these additional ‘‘costs” of bringing GGE to the
clinic, related to the research context, should be acknowledged
and carefully considered along with the potential benefits when
evaluating further research and the potential clinical applications
of GGE.

CRediT authorship contribution statement

Emilia Niemiec: Conceptualization, Investigation, Writing –
original draft. Heidi Carmen Howard: Conceptualization, Investi-
gation, Writing – original draft, Supervision.

Declaration of Competing Interest

The authors declare that they have no known competing finan-
cial interests or personal relationships that could have appeared
to influence the work reported in this paper.

Acknowledgments

We thank Dr. Oliver Feeney for his help with language editing
and insightful comments on this manuscript.

This work is supported by a grant from the Swedish Research
Council ‘‘Ethical, legal, and social issues of gene editing” (2017–
01710). Some sections of this article are also partly based on
reports 2.1 and 2.4 from the SIENNA project (Stakeholder-
informed ethics for new technologies with high socio-economic

E. Niemiec, H.C. Howard / Computational and Structural Biotechnology Journal 18 (2020) 887–896 895

and human rights impact) – which has received funding under the
European Union’s H2020 research and innovation programme
under grant agreement No 741716.

This article and its contents reflect only the views of the authors
and does not intend to reflect those of the European Commission.
The European Commission is not responsible for any use that
may be made of the information it contains.

References

[1] Knott GJ, Doudna JA. CRISPR-Cas guides the future of genetic engineering.
Science 2018;361:866–9. https://doi.org/10.1126/science.aat5011.

[2] Liang P, Xu Y, Zhang X, Ding C, Huang R, Zhang Z, et al. CRISPR/Cas9-mediated
gene editing in human tripronuclear zygotes. Protein Cell 2015;6(5):363–72.
https://doi.org/10.1007/s13238-015-0153-5.

[3] Araki M, Ishii T. International regulatory landscape and integration of
corrective genome editing into in vitro fertilization. Reprod Biol Endocrinol
2014;12(8):1–12. https://doi.org/10.1186/1477-7827-12-108.

[4] Daley GQ, Lovell-Badge R, Steffann J. After the storm – A responsible path for
genome editing. N Engl J Med 2019;380:897–9. https://doi.org/10.1056/
NEJMp1900504.

[5] Brokowski C. Do CRISPR germline ethics statements cut it? Cris J 2018. https://
doi.org/10.1089/crispr.2017.0024.

[6] Lander ES, Baylis F, Zhang F, Charpentier E, Berg P, Bourgain C, et al. Adopt a
moratorium on heritable genome editing. Nature 2019;567:165–8. https://doi.
org/10.1038/d41586-019-00726-5.

[7] The Hinxton Group. Statement on Genome Editing Technologies and Human
Germline Genetic Modification 2015.

[8] De Wert G, Pennings G, Clarke A, Eichenlaub-Ritter U, van El CG, et al. Human
germline gene editing: Recommendations of ESHG and ESHRE. Eur J Hum
Genet 2018;26:445–9. https://doi.org/10.1093/hropen/hox024.

[9] Ormond KE, Mortlock DP, Scholes DT, Bombard Y, Brody LC, Faucett WA, et al.
Human germline genome editing. Am J Hum Genet 2017;101:167–76. https://
doi.org/10.1016/j.ajhg.2017.06.012.

[10] Nuffield Council on Bioethics. Genome editing and human reproduction. 2018.
[11] National Academies of Sciences Engineering and Medicine. Human Genome

Editing: Science, Ethics and Governance. Washington, D.C.: 2017.
doi:10.17226/24623.

[12] Gyngell C, Douglas T, Savulescu J. The ethics of germline gene editing. J Appl
Philos 2017;34:498–513. https://doi.org/10.1111/japp.12249.

[13] Rulli T. Reproductive CRISPR does not cure disease. Bioethics 2019;33
(9):1072–82. https://doi.org/10.1111/bioe.12663.

[14] Ranisch R. Germline genome editing versus preimplantation genetic diagnosis:
is there a case in favour of germline interventions? Bioethics 2019;34(1):60–9.
https://doi.org/10.1111/bioe.12635.

[15] Viotti M, Victor AR, Griffin DK, Groob JS, Brake AJ, Zouves CG, et al. Estimating
demand for germline genome editing: an in vitro fertilization clinic
perspective. Cris J 2019;2(5):304–15. https://doi.org/10.1089/
crispr.2019.0044.

[16] Wolf DP, Mitalipov PA, Mitalipov SM. Principles of and strategies for germline
gene therapy. Nat Med 2019;25:890–7. https://doi.org/10.1038/s41591-019-
0473-8.

[17] Steffann J, Jouannet P, Bonnefont JP, Chneiweiss H, Frydman N. Could failure in
preimplantation genetic diagnosis justify editing the human embryo genome?.
Cell Stem Cell 2018;22:481–2. https://doi.org/10.1016/j.stem.2018.01.004.

[18] Krimsky S. Breaking the germline barrier in a moral vacuum. Account Res
2019;26(6):351–68. https://doi.org/10.1080/08989621.2019.1644171.

[19] Savulescu J, Pugh J, Douglas T, Gyngell C. The moral imperative to continue
gene editing research on human embryos. Protein Cell 2015;6:476–9. https://
doi.org/10.1007/s13238-015-0184-y.

[20] Denier Y. Need or desire? A conceptual and moral phenomenology of the child
wish. Int J Appl Philos 2006;20:81–95.

[21] Smolenski J. CRISPR/Cas9 and germline modification: new difficulties in
obtaining informed consent. Am J Bioeth 2015;15:35–7. https://doi.org/
10.1080/15265161.2015.1103816.

[22] Cussins J, Lowthorp L. Germline modification and policymaking: the
relationship between mitochondrial replacement and gene editing. New
Bioeth 2018;24:74–94. https://doi.org/10.1080/20502877.2018.1443409.

[23] Center for Genetics and Society. Open Letter on Reproductive Human Germline
Modification Organized by Center for Genetics and Society 2015. https://
www.geneticsandsociety.org/internal-content/open-letter-calls-prohibition-
reproductive-human-germline-modification.

[24] Lanphier E, Urnov F, Haecker SE, Werner M, Smolenski J. Don’t edit the human
germ line. Nature 2015;519:410–1. https://doi.org/10.1038/519410a.

[25] Darnovsky M. Genetically modifying future children isn’t just wrong. It would
harm all of us. The Guardian; 2018.

[26] World Medical Association. WMA Declaration of Helsinki – Ethical Principles
for Medical Research Involving Human Subjects 2013. https://www.wma.net/
policies-post/wma-declaration-of-helsinki-ethical-principles-for-medical-
research-involving-human-subjects/ (accessed March 31, 2020).

[27] Drabiak K. Emerging governance of mitochondrial replacement therapy:
assessing coherence between scientific evidence and policy outcomes.
DePaul J Health Care Law 2018;20.

[28] Friedmann T, Jonlin EC, King NMP, Torbett BE, Wivel NA, Kaneda Y, et al.
ASGCT and JSGT joint position statement on human genomic editing. Mol Ther
2015;23(8):1282. https://doi.org/10.1038/mt.2015.118.

[29] International Commission on the Clinical Use of Human Germline Genome
Editing. Call for evidence 2019. https://www.nationalacademies.org/_cache_
58c4/content/4885770000148509.pdf (accessed March 31, 2020).

[30] Ma H, Marti-Gutierrez N, Park S-W, Wu J, Lee Y, Suzuki K, et al. Correction of a
pathogenic gene mutation in human embryos. Nature 2017;548:413–9.
https://doi.org/10.1038/nature23305.

[31] Tang L, Zeng Y, Du H, Gong M, Peng J, Zhang B, et al. CRISPR/Cas9-mediated
gene editing in human zygotes using Cas9 protein. Mol Genet Genomics
2017;292:525–33. https://doi.org/10.1007/s00438-017-1299-z.

[32] Zeng Y, Li J, Li G, Huang S, Yu W, Zhang Y, et al. Correction of the Marfan
syndrome pathogenic FBN1 mutation by base editing in human cells and
heterozygous embryos. Mol Ther 2018;26:2631–7. https://doi.org/10.1016/j.
ymthe.2018.08.007.

[33] Kang X, He W, Huang Y, Yu Q, Chen Y. Introducing precise genetic
modifications into human 3PN embryos by CRISPR / Cas-mediated genome
editing. J Assist Reprod Genet 2016:581–8. https://doi.org/10.1007/s10815-
016-0710-8.

[34] Li G, Liu X, Huang S, Zeng Y, Yang G, Lu Z, et al. Efficient generation of
pathogenic A-to-G mutations in human tripronuclear embryos via ABE-
mediated base editing. Mol Ther – Nucleic Acids 2019;17:289–96. https://
doi.org/10.1016/j.omtn.2019.05.021.

[35] Zhang M, Zhou C, Wei Y, Xu C, Pan H, Ying W, et al. Human cleaving embryos
enable robust homozygotic nucleotide substitutions by base editors. Genome
Biol 2019;20:1–7. https://doi.org/10.1186/s13059-019-1703-6.

[36] Fogarty NME, McCarthy A, Snijders KE, Powell BE, Kubikova N, Blakeley P, et al.
Genome editing reveals a role for OCT4 in human embryogenesis. Nature
2017;550:67–73. https://doi.org/10.1038/nature24033.

[37] ACMG Board of Directors. Genome editing in clinical genetics: Points to
consider-A statement of the American College of Medical Genetics and
Genomics. Genet Med 2017;19:723–4. https://doi.org/10.1038/gim.2016.195.

[38] Howard HC, van El CG, Forzano F, Radojkovic D, Rial-Sebbag E, de Wert G, et al.
One small edit for humans, one giant edit for humankind? Points and
questions to consider for a responsible way forward for gene editing in
humans. Eur J Hum Genet 2017:1–11. https://doi.org/10.1038/s41431-017-
0024-z.

[39] National Institutes of Health. Human Embryo Research and Cloning
Prohibitions 2019. https://oir.nih.gov/sourcebook/ethical-conduct/special-
research-considerations/human-stem-cell-use/human-embryo-research-
cloning-prohibitions.

[40] European Commission Directorate General for Research & Innovation. Horizon
2020 Programme: Guidance how to complete your ethics self-assessment
2019. https://ec.europa.eu/research/participants/data/ref/h2020/grants_manual/
hi/ethics/h2020_hi_ethics-self-assess_en.pdf.

[41] Nuffield Council on Bioethics. Human embryo culture: discussions concerning
the statutory time limit for maintaining human embryos in culture in the light
of some recent scientific developments; 2017.

[42] Council of Europe. Convention for the Protection of Human Rights and Dignity
of the Human Being with regard to the Application of Biology and Medicine:
Convention on Human Rights and Biomedicine; 1997.

[43] Ballantyne A, de Lacey S. Wanted — egg donors for research: a research ethics
approach to donor recruitment and compensation. J Fem Approaches Bioeth
2008;1:145–64.

[44] Jayaprakasan K, Herbert M, Moody E, Stewart JA, Murdoch AP. Estimating the
risks of ovarian hyperstimulation syndrome (OHSS): Implications for egg
donation for research. Hum Fertil 2007;10:183–7. https://doi.org/10.1080/
14647270601021743.

[45] Schneider J, Lahl J, Kramer W. Long-term breast cancer risk following ovarian
stimulation in young egg donors: a call for follow-up, research and informed
consent. Reprod Biomed Online 2017;34:480–5. https://doi.org/10.1016/j.
rbmo.2017.02.003.

[46] Magnus D, Cho MK. Issues in oocyte donation for stem cell research. Science
2005;308:1747–8. https://doi.org/10.1126/science.1114454.

[47] International Society for Stem Cells Research. Guidlines for stem cell research
and clinical translation. 2016.

[48] National Research Council. Guidelines for Human Embryonic Stem Cell
Research. Washington, D.C.: The National Academies Press; 2005.
doi:10.17226/11278.

[49] Niemiec E, Howard HC. Germline genome editing research: what are gamete
donors (not) informed about in consent forms ? 2020;3:52–63. doi:10.1089/
crispr.2019.0043.

[50] Pinxten W, Howard HC. Ethical issues raised by whole genome sequencing.
Best Pract Res Clin Gastroenterol 2014;28:269–79. https://doi.org/10.1016/j.
bpg.2014.02.004.

[51] Niemiec E and Howard HC (2019) Chapter 3: Consenting patients to genome
sequencing. In: Tibben A and Biesecker BB (eds.) Clinical genome sequencing:
Psychological considerations, Elsevier.

[52] Howard HC, Iwarsson E. Mapping uncertainty in genomics. J Risk Res
2018;21:117–28. https://doi.org/10.1080/13669877.2016.1215344.

[53] European Parliament and Council of the European Union. Directive 2010/63/
EU of the European Parliament and of the Council of 22 September 2010 on the
protection of animals used for scientific purposes.

[54] Baylis F. Altered inheritance: CRISPR and the Ethics of Human Genome
Editing. Cambridge MA: Harvard University Press; 2019.

896 E. Niemiec, H.C. Howard / Computational and Structural Biotechnology Journal 18 (2020) 887–896

[55] Maeder ML, Gersbach CA. Genome-editing technologies for gene and cell
therapy. Mol Ther 2016;24:430–46. https://doi.org/10.1038/mt.2016.10.

[56] Hurlbut JB. Human genome editing: ask whether, not how. Nature
2019;565:135. https://doi.org/10.1038/d41586-018-07881-1.

[57] Baylis F. Questioning the proposed translational pathway for germline genome
editing. Nat Hum Behav 2019;3:200. https://doi.org/10.1038/s41562-019-
0544-3.

[58] The National Academies of Sciences, Engineering, Medicine. International
Commission on the Clinical Use of Human Germline Genome Editing 2019.
https://www.nationalacademies.org/our-work/international-commission-on-
the-clinical-use-of-human-germline-genome-editing (accessed March 31,
2020).

[59] Zuo E, Sun Y, Wei W, Yuan T, Ying W, Sun H, et al. Cytosine base editor
generates substantial off-target single-nucleotide variants in mouse embryos.
Science 2019;364(6437):289–92. https://doi.org/10.1126/science.aav9973.

[60] Jin S, Zong Y, Gao Q, Zhu Z, Wang Y, Qin P, et al. Cytosine, but not adenine, base
editors induce genome-wide off-target mutations in rice. Science 2019;364
(6437):292–5. https://doi.org/10.1126/science.aaw7166.

[61] Liang P, Ding C, Sun H, Xie X, Xu Y, Zhang X, et al. Correction of b-thalassemia
mutant by base editor in human embryos. Protein & Cell 2017;8:811–22.
https://doi.org/10.1007/s13238-017-0475-6.

[62] Martin-Palomino Olid N, García D, Rodríguez A, Vassena R, et al. Could fertility
clinics offer a sizable improvement of live birth rates by maturing post-GVBD
oocytes in vitro? Journal of Assisted Reproduction and Genetics
2019;36:1927–34. https://doi.org/10.1007/s10815-019-01540-8.

[63] OHSU Center for Women’s Health. OHSU Fertility Egg Donation Process; 2020.
[Accessed 30 March 2020].

[64] Church G. Compelling Reasons for Repairing Human Germlines. The New
England Journal of Medicine 2017;377(20):1909–11. https://doi.org/10.1056/
NEJMp1710370. In this issue.

  • Ethical issues related to research on genome editing in human embryos
    • 1 Introduction
    • 2 Germline genome editing in the clinic: potential benefits, risks and ethical issues
    • 3 Current guidelines on the potential use of GGE in the clinic: how do these impact research?
    • 4 Research context of GGE and ethical implications
      • 4.1 Challenges related to the evaluation of safety and efficacy of GGE
      • 4.2 Safety issues in germline genome editing
      • 4.3 Use of embryos
      • 4.4 Oocyte procurement
      • 4.5 Genomic sequencing
      • 4.6 Other issues related to research on GGE
    • 5 Conclusions
    • CRediT authorship contribution statement
    • Declaration of Competing Interest
    • ack15
    • Acknowledgments
    • References

ARTICLE

Civil liability for damages related to germline
and embryo editing against the legal admissibility
of gene editing
Dorota Krekora-Zając 1✉

ABSTRACT The creators of CRISPR-Cas9 method have turned to the world community,

including lawyers, to undertake a public discussion on the implications that it can create. One

of the most important problems to be resolved in the future, will be the issue of establishing

very clear legal principles of compensatory liability for damages resulting from the editing of

genes in human embryos and reproductive cells. It is necessary to show possible legal

problems that may arise and—what is more—the fact that they will certainly appear in future

legislative work in the world. Questions must be asked to which world legal experts will seek

answers. And this is the goal of this paper was set—showing possible legal problems and

asking questions related to liability for damages resulting from the editing of genes in human

embryos and reproductive cells that will be answered in the future. The most important

research questions are therefore: what is the genetic nature of the genes edition—is it a

treatment whose aim is to treat infertility of parents or the future child? How to determine

the scope of responsibility in the situation when it comes to the “cure” of one mutation, but

there is a tendency to develop a disease in the future? What then is the scope of the doctor’s

duty to inform? How to qualify the editing of a gene that is not intended to cure the existing

disease, but to obtain a certain specific immunity? What legal obligations will weigh on

parents who decide to edit the genes of the embryo or in the preconception phase? Finally,

the question arises about the time limits of this gene-editing responsibility. If we make

genetic modification of hereditary nature, then will the children or grandchildren subjected to

gene editing be able to make claims? In this paper, the provisions of international European

law, common law and continental law on the example of Polish law have been analysed. The

key findings of this paper are to show that legal problems in gene editing are not limited to

answering the question whether it should be admissible or not. For this reason, the role of

legal discourse, and in particular of private law, should focus on the reinterpretation of

traditional compensation structures, so that they also protect the rights of people whose

genome has been modified.

The newest developments in genome editing will demand that we think again about how to balance hope and fear
(Caroll and Charo, 2015).

https://doi.org/10.1057/s41599-020-0399-2 OPEN

1 Department of Civil Law, Faculty of Law and Administration, Warsaw’s University, Warsaw, Poland. ✉email: [email protected]

PALGRAVE COMMUNICATIONS | (2020)6:30 | https://doi.org/10.1057/s41599-020-0399-2 | www.nature.com/palcomms 1

12
3
4
5
6
7
8
9
0
()
:,;

Introduction

T
he development of gene therapy, in particular gene editing
using the CRISPR-Cas9 method, has prompted a lively
discussion around the world about how deeply you can

interfere with the human genome. The creators of this method
have turned to the world community, including lawyers, to
undertake a public discussion of the implications that it can create
(The National Academies of Sciences Engineering Medicine,
2015). The most important problem to be resolved in the future,
in my opinion, will be the issue of establishing very clear legal
principles of liability for damages resulting from the editing of
genes in human embryos and reproductive cells. However, before
this happens, it is necessary to show the possible legal problems
that may arise and that will certainly appear in future legislative
work in the world. Questions must be asked to which world legal
experts will need to seek answers. The goal of this paper is to
show the possible legal problems and ask questions related to the
liability for damages resulting from the editing of genes in human
embryos and reproductive cells that will be answered in the
future.

Private law considerations will be based on Polish law,
although it should be pointed out that the conclusions derived
from them appear to be of universal nature for different legal
systems. Despite the fact that legal considerations will refer to the
regulation of Polish law, the subject of the analysis will also be the
differences in the legal qualification of reproductive cells and
embryos in other European legislations. It seems that nowhere in
the world are there special regulations regarding the liability for
damage related to the genetic editing of reproductive cells or
embryos. Therefore, there is a need to present new challenges for
classic private law institutions, such as legal abilities, torts, or
liability for damages. Due to the lack of uniform European reg-
ulations and different conflicts of rights the subject of analysis will
not be wrongful life and wrongful birth actions, but only claims of
prenatal damage to a child.

The first major legal problem facing the international com-
munity is, of course, the question of the legal acceptability of the
editing of genes of human reproductive cells and embryos (van
Dijke et al., 2018). In this regard, it should be pointed out that
despite the initial demand to ban such editing, over time,
increasingly more scientists have pointed to the fact that it is not
possible to maintain such a moratorium (Doudna and Sternberg,
2017). Jiankui’s presentation at the Second International Summit
on Human Genome Editing on November 27, 2018, showed that
the introduction of a moratorium on genetic modifications of
embryos in Europe, the condemnation of such research by a
group of 120 of the greatest geneticists, even the Chinese reg-
ulations (Zhang and Lie, 2018) will not limit its conduct (Cyra-
noski and Ledford, 2018). Globalization of the medical market
means that if any procedures are allowed on other continents,
they will also become available to Europeans (Lunshof, 2016). It
should be noted that even without proper legal approval some
scientists in various countries intend to genetically modify
embryos (Cohen, 2019).

This issue is even more important because for years, we have
been observing the expansion of protection of human health in
earlier stages of development both in Poland and other Eur-
opean countries. (Kmieciak, 2015). Across Europe, we are
struggling with falling birth rates and a significant number of
miscarriages caused by various foetal abnormalities, including
genetic mutations (Araki and Ishii, 2014). It should therefore
be assumed that as soon as geneticists and biotechnologists
allow the elimination of these mutations, giving parents the
chance to give birth to healthy children, lawmakers will also be
forced to start working on the legalization of such therapy
(Simonstein, 2017).

The controversial CRISPR-Cas9 method and new conflicts of
law between protected rights
The possibilities offered by gene editing are undoubtedly very
promising for the future. Gene editing can involve modifying
somatic or reproductive cells. In the first case, the modification is
not hereditary. The second type of modification affects not only the
performance of a particular unit but also the possible transmission
of this modification to subsequent generations (Sykora, 2018).

Visions regarding genetic modifications and gene therapies have
been the subject of deliberations and medical activities for several
years, but the discovery of the CRIPSP-Cas9 method has given rise
to the greatest controversy (De Miguel Beriaina and Marcos del
Cano, 2018). Undoubtedly, this is because this method allows the
precise removal of a damaged gene and its replacement with the
correct one. This method is much cheaper, easier, safer and faster
than the ones used before. The method may be used to genetically
modify plants, animals and humans. For the first time, therefore, we
have a method by which we can control the randomness of the
evolutionary processes in all of nature (Ishii, 2017).

The first publication about using gene editing on a person
infected with HIV appeared in 2014 (Tebas et al., 2014). Then the
next use of gene editing took place in the UK in 2015, where this
treatment was given to a one-year old girl with leukaemia
(Reardon, 2015). The treatment was carried out using immune
cells derived from a donor. The application of this method to
living people (on somatic cells) does not raise major controversies
and is considered acceptable in the world of experimental medical
treatments (van Dijke et al., 2018). However, there are more
doubts about the use of this method on human reproductive cells
and embryos due to the possible inheritability of the modifica-
tions made. These fears are related not so much to the possibility
of a future child’s health being threatened as to the transmission
of such changes to the next generations and—connected to this—
taking of control over the evolution of humanity, the possibility of
deepening social differences, etc. (Doudna and Sternberg, 2017).
However, there are opposite views indicating the moral obligation
to continue research on gene editing (Savulescu et al., 2015; De
Miguel et al., 2018).

The discussion on methods of genetic editing of embryos and
germ cells shows the competition between two legally protected
rights: the right to undertake therapy, i.e., enabling the birth of a
healthy child, and the right to protect the biodiversity of future
generations. This is a conflict between relatively new values and is
significantly different from the “traditional” considerations
regarding the prenatal stage. Until now, there was only a possi-
bility of conflict in internal relations, i.e., between parents and a
child, and not in external relations, i.e., between the child’s right
to health and life and the rights of future generations, as has been
indicated. Traditionally, therefore, there have been indications of
possible conflicts between women’s rights and children’s rights
(this conflict has been mainly analysed in relation to the termi-
nation of pregnancy and medically assisted procreation proce-
dures) (Davey, 1989) and the rights of a man (father) and the
rights of a child/woman regarding the right not to consent to the
implanting of the embryo (Cohen, 2010).

In the field of gene editing of embryos and the human germ-
line, this conflict directly translates into the classic division
between the good of humanity and the good of the individual
(Agar, 2019).

International legal regulations and international community’s
opinions on gene therapy in embryos and reproductive cells
It is widely recognized that both the majority of the European
legislation and acts of international law prohibit genetic

ARTICLE PALGRAVE COMMUNICATIONS | https://doi.org/10.1057/s41599-020-0399-2

2 PALGRAVE COMMUNICATIONS | (2020)6:30 | https://doi.org/10.1057/s41599-020-0399-2 | www.nature.com/palcomms

modification of human embryos (Ishii, 2017; Charo, 2016). Fur-
ther analysis will show that this prohibition is not absolute. The
only definitive ban can be based on art. 90 al. 2 of the new
Regulation no. 536/2014 of the European Parliament and of the
Council of 16 April 2014 on clinical trials on medicinal products
for human use, repealing Directive 2001/20/EC that was not yet
transposed to the member states of the European Union (Howard
et al., 2018). However, this article does not seem to ban all types
of gene editing but only those that alter “the subject’s germ line
genetic identity”. This prohibition is also present in the art 9 of
Directive 2001/20/EC of The European Parliament and of The
Council of 4 April 2001 on the approximation of the laws, reg-
ulations and administrative provisions of the Member States
relating to the implementation of good clinical practice in the
conduct of clinical trials on medicinal products for human use.

The first act of international law in this area is the Universal
Declaration on the Human Genome and Human Rights of United
Nations Educational, Scientific and Cultural Organization
(UNESCO). The declaration has no subjective limitations and due
to its nature, it refers not only to the protection of living human
beings but also to the protection of humanity as such, including
future generations.

The declaration permits modifications of the human genome,
specifying in art. 12 that everyone should have access to
achievements in the field of genetics and medicine related to the
human genome. The content of the declaration shows that
modifications of the human genome can be carried out for health
and scientific purposes. It seems that in both cases, according to
art. 12 b, we should aim to relieve suffering and improve the
health of individuals and humanity as such.

In addition, it should be pointed out that the UNESCO
declaration does not prohibit genetic modification of human
embryos but restricts the possibility of such modifications in
reproductive cells. According to article 24, interventions regard-
ing the germ cell line may (but not need) be considered as
practices that “could be contrary to human dignity”. It should be
emphasized that the regulation of article 24 therefore differs from
Article 11 of the declaration, which explicitly prohibits the use of
reproductive cloning as a practice contrary to human dignity.
Therefore, it cannot be categorically indicated that according to
the declaration, any form of genetic interference in reproductive
cells is unacceptable; on the contrary, it must be recognized that
such interference is possible and does not have to be a threat to
human dignity (humanity). In addition, it should be noted that
pursuant to article 24 of the declaration, the UNESCO Interna-
tional Bioethics Committee should present recommendations and
guidelines on the application of the declaration, presumably also
with regard to genetic interference in the germline.

The lack of a complete ban on gene editing makes it possible to
recognize the right of a person, as expressed in article 8 of the
declaration, to be compensated for losses caused by direct and
decisive interference in his or her genome.

Another legal act relating to the recognition of the genome as a
common good is the Bioethical Convention (The Convention for
the Protection of Human Rights and Dignity of the Human Being
with regard to the Application of Biology and Medicine: Con-
vention on Human Rights and Biomedicine (ETS No. 164, so-
called Oviedo Convention). According to its article 13, an inter-
vention aimed at making changes in the human genome can only
be carried out for prophylactic, therapeutic or diagnostic pur-
poses when its aim is not to cause hereditary genetic changes in
the offspring. As pointed out by Sykora and Caplan (2017), one
can argue that the ban on germline gene therapy in article 13 is in
contradiction with one of the motivations of the Oviedo Con-
vention: that progress in biomedicine would be used solely for the
benefit of future generations”. Nevertheless in 2015, the

International Bioethics Committee called on the member states to
adopt a joint temporary moratorium on germline editing.
According to point 46 of the report “Updating its Reflection on
the Human Genome and Human Rights”, the reasons for a ban
on gene editing are the “uncertainties on the effect of germline
modification on the future generations”.

With this formulation of the Oviedo Convention, it seems
necessary to interpret the very concept of purpose; if the purpose
of the intervention is to “repair” the genome of the embryo or
reproductive cells, causing the possible transmission of these
modifications to subsequent generations, can it be concluded that
the inheritance of these changes is the goal of the intervention or
only a side effect that we know of? This is important in the
context of article 1 of the convention, which shows that the main
purpose of the convention is the protection of individual rights. I
do not find any justification for the view presented in the lit-
erature that this prohibition also applies to the “modification of
the genome of spermatozoids and oocytes intended for fertiliza-
tion”. In my opinion, it is impossible to explicitly exclude such an
interpretation of article 13 of the convention, according to which
interventions aimed at a significant therapeutic or prophylactic
goal for the individual should be treated differently from those
whose main purpose is to cause changes in the offspring. In the
first case, it should be pointed out that because the primary
purpose is to protect the health and life of the future child
embryo, such interventions cannot be considered contrary to the
convention (Sykora Peter and Caplan Arthur, 2017). According
to de Miguel Beriain et al. (2019) article 13 of Oviedo Convection
prohibits only clinical applications of germline gene editing. They
forecast that there will be new redaction of Article 13 that allows
clinical use of germline gene editing.

There is also no general prohibition on gene editing in the
Charter of Fundamental Rights of the European Union. There is
no doubt that the charter indicates a total ban on all eugenic
practices, but in a situation in which gene editing is to be used for
therapeutic purposes, it is difficult to clearly interpret such a
prohibition.

According to point 40 of the preamble of the directive on the
legal protection of biotechnological interventions of the European
Parliament and the Council of the European Union, there are
prohibitions on the patentability of processes modifying the
germline genetic identity of human beings, and germline gene
editing is treated as an offence against public order and morality.

According to the recommendations of the German Ethics
Council from the 9th of May 2019, a temporary ban on gene editing
of the human germline is also needed. The temporality of this
prohibition is justified, among others, by the fact that genome
editing is a very young field of research and that only two cases are
known in which intervention in the germline to prevent polygenic
and multifactorial disease would have any chance of success.
According to point 99 of the executive summary of the recom-
mendations, it will be possible to use germline gene editing only if
the technology is sufficiently safe, effective and tolerable. In the
executive summary of the recommendations, it is pointed out that
“Germline interventions will presumably change the network of
relationships between the members of a society”. However, the
“ethical concepts of the protection of life, of freedom and of ben-
eficence suggest some a duty to permit such interventions”. Based
on point 4 of recommendations, the moratorium should undergo a
regular transparent review. International groups of ethicists and
researchers also call for a global but not permanent moratorium,
including some of those who originally developed CRISPR-Cas9 as
a gene-editing tool (Lander et al., 2019).

The Nuffield Council on Bioethics recommends that heritable
genome editing interventions should only be permitted provided
that arrangements are in place to monitor the effects on those

PALGRAVE COMMUNICATIONS | https://doi.org/10.1057/s41599-020-0399-2 ARTICLE

PALGRAVE COMMUNICATIONS | (2020)6:30 | https://doi.org/10.1057/s41599-020-0399-2 | www.nature.com/palcomms 3

whose interests can be collaterally affected and on society more
generally and provided that effective mechanisms are in place to
redress any such effects (The Nuffield Council on Bioethics,
2018).

The report of an international committee convened by the U.S.
National Academy of Sciences (NAS) and the National Academy
of Medicine in Washington, DC, concludes that human embryo
editing “might be permitted, but only following much more
research” on risks and benefits and “only for compelling reasons
and under strict oversight”. The U.S. government prohibits the
use of federal funding for research involving human embryos.
However, gene editing of human embryos can be performed
using private funding. The Food and Drug Administration is
barred from considering any studies that would involve using
genetically modified human embryos to create a pregnancy.
However, laws that govern the creation of genetically modified
babies vary widely internationally.

The analysis clearly shows that there are still no definitive
prohibitions on gene editing of embryos or the germline. Existing
prohibitions have a temporary character that protects the per-
missiveness of such operations before they are better known and
safe (Nordberg et al., 2019). S. Holm indicates that the concept
“only when it is safe” can be deceptive and hides “the full
implications of the arguments made about ethics of gene editing
and their underlying philosophical justifications” (Holm, 2018).
Transferring these doubts to private law, one should ask ques-
tions about the legal nature of such treatment.

The legal nature of germline and embryo gene editing
Legal regulations regarding gene editing of embryos and repro-
ductive cells can be found in legal acts regarding clinical trials,
biomedical research (EU Regulation), medically assisted pro-
creation procedures or codes of medical ethics. The first method
of regulation has already been discussed above, and it refers to the
public law regulation answering the question of whether such a
procedure should be prohibited. The second and third refer to the
personal rights of the patient and thus to private law.

Such legal solutions will be discussed on the basis of Polish law.
However, it should be noted that in this respect Polish law is
modelled on the Oviedo Convention (Haberko, 2016) and is
therefore not significantly different from the regulations adopted
by other European countries.

In Polish law, two acts can be pointed out that directly refer to
genetic modifications. The first is the act on the treatment of
infertility, and the second is the code of medical ethics.

According to article 25 para. 2 of the act on the treatment of
infertility (Ustawa o leczeniu niepłodności Ustawa z dnia 26
czerwca 2015 r. o leczeniu niepłodności), “(n) creation with the
use of techniques for the medically assisted procreation of chi-
meras and hybrids and intervention aimed at making inheritable
changes in the human genome that can be passed on to next
generations is not allowed”. A fundamental question arises here
about the interpretation of this norm and its ratio legis that guides
the legislator. Assuming the rationality of this legislator, it should
be recognized that the lawmaker did not forbid the editing of
genes with the preceding technique for assisted procreation but
only forbids the type of intervention which can be passed on to
future generations.

Analysing in depth the content of article 25 para. 2, it should be
pointed out that two models of interpretation are possible. The
first would lead to the conclusion that the act on the treatment of
infertility is more restrictive than the Bioethical Convention and
that it prohibits any genetic modification that may (or not, as the
convention indicates “are intended”) change the offspring. An
open question arises in this respect: what does the term “may”

mean? Is it possible, according to current knowledge, that mod-
ifications will appear in the offspring or that on the contrary,
modifications are forbidden because theoretically we cannot
exclude their hereditary nature, although we lack evidence for
their heredity?

The second model of linguistic interpretation would hold that
because gene editing is not a technique of assisted procreation,
the prohibition does not apply to it. Such an interpretation would
be consistent with article 4 of the act, assuming that “infertility
treatment is carried out (…) with particular emphasis on the legal
protection of life, health, good and the rights of the child”.
Therefore, it is difficult to argue that a situation where genetic
modification would allow the birth of a healthy child could be
covered by the prohibition in article 25.

Again, an open question arises—in a situation where the main
purpose of the modification is to remove from a particular
embryo a particular mutation and the secondary goal is to
transfer the modified gene to the next generations, is prohibiting
such practices justified?

Referring to the Code of Medical Ethics (Kodeks Etyki
Lekarskiej), it should be pointed out that pursuant to article 51
par. 4, “(l) the doctor may not participate in activities aimed at
inducing hereditary genetic changes in a human being”. Assum-
ing that in most cases genetic modification will be done by a
geneticist, not a doctor, the regulation of the code will not apply
to the medical practitioner. In such cases, gene editing will be
qualified as a medical experiment and will require, among others,
the opinion of the bioethics commission.

Summarizing this part of the considerations, it should be
emphasized that it is extremely difficult to unequivocally indicate
that Polish law prohibits gene editing of embryos and germline
cells. In Poland, no such modifications have been carried out.
There is no doubt, however, that in the coming years, we will also
face the dilemma as to whether to destroy embryos that due to
their genetic mutations cannot be implanted in a woman’s body
or whether to treat them with the help of gene editing. The
question then arises as to whether a child or his or her parents
will have claims to compensation and if so to whom, if gene
therapy causes harm to the child or if it is not performed despite
its availability.

Legal classification of genetic modifications of human
embryos and reproductive cells
At the beginning of the second part of the study, the following
questions should be asked: who would be the patient in a situa-
tion where genetic modifications were made to reproductive cells
and would such modifications be considered a medical
procedure?

To answer these questions, the legal status of human germ cells
and embryos should be determined. In many European legisla-
tions the term embryo is defined by law of medically assisted
procreation. However, nasciturus is the term of traditional civil
law based on Roman’s origin law. In some countries there is a
debate as to whether it is possible to extend the concept of nas-
citurus to the embryo (Cacace, 2013). In Polish law, the term
embryo has been defined as a group of cells formed as a result of
the extracorporeal connection of the female and male reproduc-
tive cells, from the end of the process of fusion of the germ cells
(karyogamy) to implantation in the endometrium witch is a stage
without legal personality. After the implementation in the
endometrium the embryo is considered to be a nasciturus and a
potential human who has conditional legal capacity. The doctrine
indicates that despite the lack of explicit statutory regulation in
the field of compensation for damages, the embryo should be
considered a nasciturus. In reference to cells of human origin, in

ARTICLE PALGRAVE COMMUNICATIONS | https://doi.org/10.1057/s41599-020-0399-2

4 PALGRAVE COMMUNICATIONS | (2020)6:30 | https://doi.org/10.1057/s41599-020-0399-2 | www.nature.com/palcomms

the absence of explicit statutory regulation, it is postulated to
consider them as the subject of law (res with a very limited
turnover).

Similarly, it should be pointed out that the issue of the status of
human body parts in various European countries raises a lot of
controversy. In Italian law, it can be indicated that the recognition
of whether a given element can be protected by legal claims or by
the protection of personal rights depends on the nature of the
body parts and the purpose for which they were disconnected
from the human body. As De Cupis (1985) points out, parts
detached from the human body, the disconnection of which is not
associated with irreversible damage to the body, become movable
goods that can be disposed of with certain restrictions arising
from article 5 of codice civile (Civil Code Regio Decreto 16 marzo
1942, n. 262 (G. U. n. 79 del 4-4-1942)). It should be noted that
things (beni) within the meaning of article 810 c.c. are things that
can be subject to rights. According to this theory, parts of the
human body become things when they become detached from
the body.

As in Polish law, the Italian Civil Code uses the concept of
nasciturus, guaranteeing, for example, rights related to inheri-
tance, and the concept of embryo was specified in the Act on
Medically Assisted Procreation Legge 19 febbraio 2004, n 40,
Norme in materia di procreazione medicalmente assistita (l.40/
2004).

The German jurist Wolf (Simić, 2018) stated: “Research into
the civil law confirmed that nasciturus does have legal person-
ality”, concluding that nasciturus, as a legal subject can suffer
harm to his/her health. According to the German Civil Code, in
order to provide future right to support, it is possible to obtain a
temporary order in favour of nasciturus and in the order to
preserve his/her future rights, it is possible to appoint his/her a
guardian”.

Initially, in Great Britain there was no doubt that the human
body or even a human being could be the object of property, the
best proof of which was slavery and feudal relations. This view
was maintained until the beginning of the 19th century, i.e. until
the abolition of slavery, although in jurisprudence it had aroused
much controversy (Nwabueze, 2007). It should be noted that
English positive law does not de facto determine the legal status of
the human body, leaving the regulation to case-law. The general
principle arising from common law is therefore the recognition
that it has no property on the human body, but the development
of medical science and commercialization of scientific research
results have led the jurisprudence to create many exceptions to
this principle. Referring to the subject of reproductive cells, the
courts often referred to exceptions to the rule of no property on
human body. The admissibility of qualifying male semen as an
object of property rights was indicated (R v. Welsh (1974)
RTR478).

Under Polish law, all relations between the patient and
healthcare service providers are regulated by civil law obligations
(Borysiak, 2019).

There is no doubt that as long as the cells are in the human
body, they share its legal status (Krekora-Zając The legal …
2015a, 2015b). This means, therefore, that in a situation where
reproductive cells are genetically modified, the patient is the person
from whom they originate. Regarding this treatment, it should be
pointed out that the therapeutic goal is reproductive cell therapy
aimed at the treatment of infertility. Despite its title, the act does
not apply to every infertility treatment. Gene modifications of
reproductive cells would only be acceptable if the mutations held
by these cells could not lead to the conception and birth of a child
before the modification was applied. I am referring to the most
serious diseases that cause infertility or a high risk of foetal death
(or spontaneous abortion). There is no doubt, that the concept of

serious illnesses has not been defined, and as Kleiderman et al.
(2019) show it can be interpreted differently and based on objective
criteria (morbidity) or flexible and evolving elements (disability as
impacted by social arrangements or treatability as impacted by
effectiveness and cost of the treatment).

The situation would look differently if the occurrence of a
genetic disease did not cause a foetus to die (or its non-existence)
or cause the birth of a sick child. It is difficult to stipulate that
such a treatment that affects the genotype is therapeutic for the
patient who is a cell donor. Is it possible to indicate that in this
regard the doctor performs a medical treatment for a future legal
person, that is, the person who the child will become? The answer
to this question is not easy, although some proponents of the
doctrine indicate that legal subjectivity is not a feature that
determines the status of a patient. Traditionally, however, the
subject of consideration was the legal nature of nasciturus, the
embryo, and not the reproductive cells from which it would arise.
Karkowska (2010) explicitly indicates that an unborn child has
the right to the protection of his or her life and health, although
due to the lack of legal capacity, he or she is not subject to all
patients’ rights. In turn, according to Kmiciak (2018), “(n) I doubt
that a child conceived de facto is automatically put in the
“position” of the patient.” In the opinion of Gałązka (2018), the
very fact of conception with the help of medically assisted pro-
creation is a sui generis medical procedure whereby one of the
patients is a future child.

Since most of the doctrine’s proponents detach the state of
being a patient from being a legal entity, pointing to the sepa-
rateness of the rights of the future child in relation to the mother,
it must be assumed that in the field of genetic modification of
human cells and embryos, we can also determine such rights of
the future child (Karkowska, 2010; Kmiecika, 2018, Gałązka,
2018). At the same time, it should be emphasized that such a view
seems more justified/unmistakable in the cases discussed here
than in the case of “traditional” procedures on the nasciturus
because treatments on the embryo and reproductive cells are
performed on a separate being without interfering with a
woman’s body. Of course the procedure precedes the collection of
cells from the body but genetic modification is performed on a
cell taken from the woman’s body.

Prenatal damage and preconception injures
Prenatal damage was directly defined in article 4461 of the Polish
civil code (c.c.). Similar regulations exist in many European coun-
tries (Simić, 2018). This regulation has made it possible for a child
to claim damages for the damage that he or she has suffered before
birth. Using the language interpretation, it should be noted that art.
4461 c.c. does not indicate the child’s right to claim for damages
caused only during pregnancy. In my opinion, there are no grounds
for limiting the scope of article 4461 c.c. to only the damage that has
occurred since the conception of the child. Moreover, it should be
pointed out that in situations where the legislator wants to link
permission with the “status” of the nasciturus, this directly defines
the status in the regulation. For example, it can be pointed out that
such a referral can be found in article 927 § 2 c.c. (a child already
conceived). I therefore accept that article 4461 c.c. is the basis for a
child’s claims related to events that arose during foetal life as well as
during preconception.

In her article, Wojtaszek (1990) aptly pointed out that legal
subjectivity matters only when the claim is made and that com-
pensation for preconception damage is subject to damage con-
sisting in a health disorder. The author, referring to the views of
Rezler, indicates that unlawfulness is a feature of a cause and may
be a future violation of the legally protected interests of an entity
that is to be created in the future.

PALGRAVE COMMUNICATIONS | https://doi.org/10.1057/s41599-020-0399-2 ARTICLE

PALGRAVE COMMUNICATIONS | (2020)6:30 | https://doi.org/10.1057/s41599-020-0399-2 | www.nature.com/palcomms 5

Preconception damages have traditionally been combined with
damages that are de facto suffered by the mother of the child and
that are only secondary in terms of the child, in contrast to
prenatal damage, in regard to which Kaliński (2018) aptly points
out that “an act directed against the mother is also an act directly
aimed at the child”.

The procedures of assisted procreation and genetic modifica-
tion of human reproductive cells have shown, however, that it is
possible to cause damage that will only become apparent after the
birth of the child and that will not be damage done to the mother.
This issue is not only related to genetic modification but also
occurs in relation to mistakes in the in vitro procedure, e.g.,
in situations where the implantation of a non-maternal embryo is
done (Krekora-Zając W świetle… 2015a, 2015b).

Another problem that can arise with regard to legal subjectivity is
the argument of identity (Soniewicka, 2018). It is difficult to indicate
the possibility of pursuing claims showing the guilt of the doctor
(Article 415 of the Polish Civil Code). It will also be difficult to
indicate the causal relationship between the damage and the event.

An example of such a situation may be the correction of both
mutated copies of the gene encoding beta-globin in people with
sickle cell of anaemia, which will relieve them of the disease but
deprive them of the mutation’s protection against malaria. When
such a child develops malaria, it will be difficult to prove that the
immediate cause was the modification, not the mosquito infec-
tion. Such a relationship can be demonstrated in two cases: first,
in a situation where the child has a high probability of contact
with malaria, e.g., living in an area where this disease occurs, and
second, in a situation where the doctor/legal guardian did not
fulfil the information obligation towards the parents/child.

Even greater doubts may arise when the gene is not edited to cure
the existing disease but to obtain a certain specific immunity.
Therefore, this case is about making a modification that has no
therapeutic purpose and that is preventive. It is well known that a
modification in the CCR5 gene (a similar modification has just been
carried out by He Jiankui) causes resistance to HIV but increases
the probability of becoming sick (susceptibility to a virus) with the
West Nile fever. Undoubtedly, for the legality of such a medical
experiment, a comparison of benefits and risks will be required.
However, the question remains open whether a child who is ill with
West Nile fever will have a claim against the entity that made these
modifications. As in the previous case, will we be able to indicate an
adequate causal relationship between the event and the damage?

Another problem is identifying the entity responsible for the
damage. Fundamental in this respect is the problem of the
responsibility of the parents who would agree to such a mod-
ification. It seems unresolved whether pursuant to article 441 c.c.
they will become entities jointly and severally liable and whether
in general they are entitled to give such consent. These issues are
directly related to the rights and obligations of future parents
towards future children and the legal status of the human embryo
and reproductive cells. There is no doubt that it can be proble-
matic to justify blaming the parents (the classic association of
guilt in supervision). According to R. Scott (2000) there are very
restrictive limitations to a duty of care toward an unborn child.
First, it is doubtful whether there would be a legal basis for the
consent of future parents. Indeed, it cannot be said that by
agreeing the parents would be making decisions that fall under
parental authority, which would arise only if the child was born.
Therefore, it would be possible to consider their responsibility
only when the parental authority was also extended to the pre-
natal period. Second, according to the current legal doctrine,
there are no cases in which consent to a child’s legal medical
procedure could be the basis of parents’ responsibility.

Another important threat associated with gene editing is the
right to privacy and autonomy. Many scientists indicate that

children who were conceived from genetically modified cells or
from embryos subjected to gene editing should be monitored for
a long time (Ishii and de Miguel Berian, 2019).

Finally, a question arises about the time limits on this
responsibility. According to art. 4421 c.c. the limitation of claims
by a minor for remedying a person’s injury cannot end before the
expiration of 2 years from the date when he or she reaches the age
of majority. If we make a genetic modification of a hereditary
nature, will the children or grandchildren of the subject whose
genome had been subjected to gene editing be able to make
claims? It seems that responsibility will always be limited by the
sine qua non premise and an adequate causal relationship. At the
current level of knowledge, we can indicate that the genome of
each new generation will result not only from a single mod-
ification but also from subsequent modifications associated with
environmental and associated changes that will naturally arise as
a result of conception. However, it cannot be ruled out that in the
future, as genetics develop, this reasoning should be rejected.

Conclusions
The belief that legal regulation will prohibit the editing of human
genes seems illusory. As pointed out by Łętowska (2018), the faith
that the law will secure all possibilities is an illusion. It can help a
little, but it will not change the world. For this reason, the legal
discourse, and in particular private law, should be focused on the
reinterpretation of traditional compensation structures so that
they can also protect the rights of people whose genome has been
modified. Today, it is too early to formulate such solutions. It
seems that the aim of the discussion, including the legal one,
should be to indicate problems in the area of liability for damages,
which is relevant not only for us as lawyers but also for us as
people (Savulescu et al., 2015). This discussion should take place
even if today we believe that the clinical use of gene editing is
premature and irresponsible (Team of experts from the Polish
Episcopate for bioethical matters 2016) and we have a lot of
doubts related to using gene editing to human enhancement
(Soniewicka and Lewandowski, 2019).

The ability to edit genes creates some new legal problems but
also forces legal reconsiderations of previously existing issues,
ultimately unresolved by law and lawyers, such as the legal
status of the embryo and sex cells and the rights and duties
of future parents, especially in the period before conception
(Sykora, 2018).

As I pointed out in the “Introduction”, this paper did not aim
to solve these problems—which is a long-term process—but
aimed only to show the problems to start the discussion.

Data availability
Data sharing not applicable to this article as no datasets were
generated or analysed during the current study.

Received: 24 July 2019; Accepted: 17 January 2020;
Published online: 25 February 2020

References
Agar N (2019) Why we should defend gene editing as eugenics. Camb Q Healthc

Ethics 28:9–19. https://www.cambridge.org/core/journals/cambridge-quarterly-
of-healthcare-ethics/article/why-we-should-defend-gene-editing-as-eugenics/
00B15AEB625379F8543C43E286160B87

Araki M, Ishii T (2014) International regulatory landscape and integration of
corrective genome editing into in vitro fertilization. Reprod Biol Endocrinol
12:108. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4251934

Borysiak W (2019) Freedom of contract in the medical legal relationship. Stud Prawa
Prywatnego 52:71–82. https://czasopisma.beck.pl/studia-prawa-prywatnego/
artykul/freedom-of-contract-in-the-medical-legal-relationship/

Cacace S (2013) Identità e Statuto Dell’embrione Umano: Soggetto di Diritto/Oggetto
Di Tutela? Riv Ital Med Leg (e del Diritto in campo sanitario) 4:1736–1751.

ARTICLE PALGRAVE COMMUNICATIONS | https://doi.org/10.1057/s41599-020-0399-2

6 PALGRAVE COMMUNICATIONS | (2020)6:30 | https://doi.org/10.1057/s41599-020-0399-2 | www.nature.com/palcomms

https://www.iusexplorer.it/Rivista/Rivista_Italiana_di_Medicina_Legale/
Identità_e_statuto_dell’embrione_u?IdDatabanks=144&IdDocMaster=4087732

Caroll D, Charo RA (2015) The societal opportunities and challenges of genome
editing. Genome Biol 16:242

Charo RA (2016) The legal and regulatory context for human gene editing. Issues
Sci Technol 32. https://issues.org/the-legal-and-regulatory-context-for-
human-gene-editing/. Accessed 22 July 2019

Charter of Fundamental Rights of the European Union (OJ C 364/1). https://www.
europarl.europa.eu/charter/pdf/text_en.pdf

Codice Civile Regio Decreto 16 marzo 1942, n. 262 (G. U. n. 79 del 4-4-1942).
https://www.gazzettaufficiale.it/anteprima/codici/codiceCivile

Cohen G (2010) The right not to be a genetic parent? South Calif Law Rev
81:1115–1196. https://papers.ssrn.com/sol3/papers.cfm?abstract_id=1116269

Cohen J (2019) Russian geneticist answers challenges to his plan to make gene-
edited babies. Science (advance online publication 12 June). https://doi.org/
10.1126/science.aay3879

The Convention for the Protection of Human Rights and Dignity of the Human Being
with regard to the Application of Biology and Medicine: Convention on Human
Rights and Biomedicine (ETS No. 164). https://rm.coe.int/16808ff399

De Cupis A (1985) I diritti della personalità. In: Cicu A, Messineo F (ed) Trattato di
diritto civile e comerciale. Milano, p. 159

Cyranoski D, Ledford H (2018) Genome-edited baby claim provokes international
outcry. Nature (advance online publication November). https://doi.org/
10.1038/d41586-018-07545-0

Davey DA (1989) The right to be born. Med Law 7(5):475–482
De Miguel Beriain I, Marcos del Cano AM (2018) Gene editing in human embryos. A

comment on the Ethical Issues Involved. In: Soniewicka M (ed) The ethics of
reproductive, philosophy and medicine, vol. 128. Springer, Cham, pp. 173–187

Van Dijke I, Bosch L, Bredenoord AL, Cornel M, Repping S, Hendriks S (2018) The
ethics of clinical applications of germline genome modification: a systematic
review of reasons. Hum Reprod 9(33):1777–1796. https://www.ncbi.nlm.nih.
gov/pmc/articles/PMC6454467/

Directive 2001/20/EC of The European Parliament and of The Council of 4 April
2001on the approximation of the laws, regulations and administrative pro-
visions of the Member States relating to the implementation of good clinical
practice in the conduct of clinical trials on medicinal products for human use
(OJ L 121, 1.5.2001, p. 34). https://ec.europa.eu/health/sites/health/files/files/
eudralex/vol-1/dir_2001_20/dir_2001_20_en.pdf

Doudna JA, Sternberg SH (2017) A crack in creation: gene editing and the
unthinkable power to control evolution. Houghton Mifflin Harcourt,
Houghton

Gałązka M (2018) Świadczenia zdrowotne wspomagające prokreację. In: Bosek L,
Wnukiewicz-Kozłowska A (ed) System Prawa Medycznego. Szczególne
śwadczenia zdrowotne, vol. 2. C.H. Beck, Warsaw, p. 138

German Civil Code BGB 2002 (FLG I p. 738). https://www.gesetze-im-internet.de/
englisch_bgb/englisch_bgb.pdf

German Ethics Council (2019) Intervening in the human germline opinion
Executive summary & recommendations. https://www.ethikrat.org/en/press-
releases/2019/ethics-council-germline-interventions-currently-too-risky-but-
not-ethically-out-of-the-question/. Accessed 22nd July 2019

Haberko J (2016) Ustawa o leczeniu niepłodności. Komentarz. Wolters Kluwer
Polska, Warsaw

Holm S (2018) Let us assume that gene editing is safe-the role of safety arguments
in the gene editing debate. Camb Q Healthc Eth 28:110–111. https://www.
cambridge.org/core/journals/cambridge-quarterly-of-healthcare-ethics/
article/let-us-assume-that-gene-editing-is-safethe-role-of-safety-arguments-
in-the-gene-editing-debate/E760EB3B2463C32DF5F08DEE2F39AF0E

Howard HC, van El CG, Forzano F, Radojkovic D, Rial-Sebbag E, de wert G, Borry
P, Cornel MC (2018) One small edit for humans, one giant edit for
humankind? Points and questions to consider for a responsible way forward
for gene editing humans. Eur J Hum Genet 26:1–11. https://www.ncbi.nlm.
nih.gov/pmc/articles/PMC5839051/

International Bioethics Committee UNESCO (2015) Report of the IBC on updating
its reflection on the Human Genome and Human Rights. https://unesdoc.
unesco.org/ark:/48223/pf0000233258. Accessed 22 Jul 2019

Ishii T (2017) Germ line genome editing in clinics: the approaches, objective and
global society. Brief Funct Genomics 16(1):46–56. https://www.ncbi.nlm.nih.
gov/pmc/articles/PMC5291189/

Ishii T, de Miguel Beriain I (2019) Safety of germline genome editing for genetically
related “future” children as perceived by parents. CRISPR J (published online
19 Nov 2019). https://doi.org/10.1089/crispr.2019.0010

Kaliński M (2018) Odpowiedzialność odszkodowawcza. In: Olejniczak A (ed.)
System Prawa Prywatnego. Prawo zobowiązań-część ogólna. C. H. Beck,
Warsaw, pp. 48–49

Karkowska D (2010) Ustawa o prawach pacjenta i Rzeczniku Praw Pacjenta.
Komentarz, Wolters Kluwer Polska, Warsaw

Kmieciak B (2015) W stronę chirurgii zarodka. Socjoprawne aspekty innowa-
cyjnych technik wspomaganego rozrodu. Ethics Prog 6(1):141–153

Kmiciak B (2018) Pacjent i jego status prawny. In: Kubiak R, Kubicki L (eds)
System Prawa Medycznego. Pojęcie, źródła i zakres prawa medycznego. Tom
I. Wolters Kluwer Polska, Warsaw, p. 156

Kleiderman E, Ravitsky V, Knoppers BM (2019) The ‘serious’ factor in germline
modification J Med Eth 45:508–513. https://jme.bmj.com/content/45/8/508

Kodeks Etyki Lekarskiej. https://www.nil.org.pl/__data/assets/pdf_file/0003/4764/
Kodeks-Etyki-Lekarskiej.pdf

Krekora-Zając (2015a) The legal status of human biological samples used in
research purposes. Stud Prawnicze 203:89–134. http://cejsh.icm.edu.pl/
cejsh/element/bwmeta1.element.desklight-04af6bb2-cfd7-489d-8607-
e81cd9dcd31f

Krekora-Zając (2015b) W świetle prawa matka jest pewna, ale …, Gazeta Prawna
27.02.2015. https://prawo.gazetaprawna.pl/artykuly/855751,w-swietle-prawa-
matka-jest-pewna-ale-sa-pewne-wyjatki.html

Lander E, Baylis F, Zhang F, Charpentier E, Berg P, Baurgain C, Friedrich B, Joung
JK Li. J, Liu D, Naldino L, Nie J-B, Qui R, Schoene-seifer B, Shao F, Terry S,
Wei W, Wunnacker E-L (2019) Adopt a moratorium on heritable genome
editing. Nature (advance online publication 13 March). https://doi.org/
10.1038/d41586-019-00726-5

Legge 19 febbraio 2004, n 40, Norme in materia di procreazione medicalmente
assistita (G. U. n 45 del 24-2.2004). http://www.parlamento.it/parlam/leggi/
04040l.htm

Lunshof JE (2016) Human germ line editing-roles and responsibilities. Protein Cell
7(1):7–10. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4707153/

Łętowska E, Woś R (2018) Kiedy prawo ma wąsy. Tygodnik Powszechny 49.
https://www.tygodnikpowszechny.pl/kiedy-prawo-ma-wasy-156563

de Miguel Beriain I, Armaza Armaza E, Sánchez AD (2019) Human germline
editing is not prohibited by the Oviedo Convention: an argument. Med Law
Int (first published July). https://doi.org/10.1177/0968533219862590

National Academies of Sciences Engineering Medicine, Baltimore D, Baylis F, Berg
P, Daley GQ,Doudna JA, Lander ES, Lovell-Badge R, Ossorio P, Pei D,
Thrasher A, Winnacker E-L, Zhou Q (2015) On Human Gene Editing:
International Summit Statement. http://www8.nationalacademies.org/
onpinews/newsitem.aspx?RecordID=12032015a. Accessed 22 Jul 2019

Nordberg A, Minssen T, Feeney O, de Miguel Beriain I, Galvagni L, Wartiovaara K
(2019) Regulating germline editing in assisted reproductive technology: an
EU cross-disciplinary perspective. Bioethics (special issue: Human germline
editing) 12:16–32. https://doi.org/10.1111/bioe.12705

Nuffield Council on Bioethics (2018) Genome editing and human reproduction:
social and ethical issues. http://nuffieldbioethics.org/project/genome-editing-
human-reproduction. Accessed 22 Jul 2019

Nwabueze RN (2007) Biotechnology and the challenge of property. Property rights in
dead body, body parts, and genetic information. Routledge, Hampshire, p. 44

Reardon S (2015) Leukaemia success herald wave of gene-editing therapies. Nature
527:146–147. https://www.nature.com/news/leukaemia-success-heralds-
wave-of-gene-editing-therapies-1.18737

Regulation no. 536/2014 of the European Parliament and of the Council of 16 April
2014 on clinical trials on medicinal products for human use, repealing Directive
2001/20/EC (OJ L 158, 27.5.2014, pp. 1–76). https://ec.europa.eu/health/sites/
health/files/files/eudralex/vol-1/reg_2014_536/reg_2014_536_en.pdf

Savulescu J, Pungh J, Doiglas T, Gyngell Ch (2015) The moral imperative to
continue gene editing research on human embryos. Protein Cell 6
(7):476–479. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4491050/

Scott R (2000) Maternal duties toward the Unborn? Sounding from the Law of Tort
Med Law Rev 8(1):1–68. https://academic.oup.com/medlaw/article-abstract/
8/1/1/938923?redirectedFrom=fulltext

Simić J (2018) The Protection of Nasciturus within the civil law. Pravno Zapisi
God. IX. http://pravnifakultet.rs/wp-content/uploads/2018/12/Pravni-zapisi-
2018-02-03-Simic.pdf

Simonstein F (2017) Gene editing, enhancing and women’s role. Sci Eng Eth
(advance online publication 2 February). https://doi.org/10.1007/s11948-017-
9875-5

Soniewicka M (2018) Selekcja Genetyczna w Prokreacji Medycznie Wspomaganej.
Wolters Kluwer, Warsaw

Soniewicka M, Lewandowski W (2019) Human genetic selection and enhancement.
Parental perspectives and law. Peter Lang GmbH.

Sykora P, Caplan A (2017) Germline gene therapy in compatible with human
dignity. EMBO Rep. 18(12):2086. https://www.ncbi.nlm.nih.gov/pmc/articles/
PMC5709747/

Syrkora P (2018) Germline gene therapy in the era of precise genome editing: how
far should we go? In: Soniewicka M (ed.) The ethics of reproductive, philo-
sophy and medicine, vol. 128. Springer, Cham, pp. 157–171

Tebas P, Stein D, Tang WW, Frank I, Wang SQ, Lee G, Spratt SK, Surosky RT, Giedlin
MA, Nichol G, Holmes MC, Gregory PD, Ando DG, Kalos M, Collman RG,

PALGRAVE COMMUNICATIONS | https://doi.org/10.1057/s41599-020-0399-2 ARTICLE

PALGRAVE COMMUNICATIONS | (2020)6:30 | https://doi.org/10.1057/s41599-020-0399-2 | www.nature.com/palcomms 7

Binder-Scholl G, Plesa G, Hwang W-T, Levine BL, June CH (2014) Gene editing
of CCR% in autologous CD4T cells of persons infected with HIV. N Engl J Med
370:901–910. https://www.nejm.org/doi/10.1056/NEJMoa1300662

Universal Declaration on the Human Genome and Human Rights of United
Nations Educational, Scientific and Cultural Organisation (UNESCO)
adopted on 11 November 1997. http://www.unesco.org/new/en/social-and-
human-sciences/themes/bioethics/human-genome-and-human-rights/

Ustawa z dnia 26 czerwca 2015 r. o leczeniu niepłodności (Dz.U. 2015, poz. 1087).
http://prawo.sejm.gov.pl/isap.nsf/DocDetails.xsp?id=WDU20150001087

Wojtaszek E (1990) Odpowiedzialność cywilna z tytułu szkody wyrządzonej
dziecku przed jego poczęciem. Nowe Prawo 10–12:96

Zhang D, Lie RK (2018) Ethical issues in human germline gene editing; a per-
spective from China. Monash Bioeth Rev 36:23–35. https://link.springer.com/
article/10.1007/s40592-018-0091-0

Acknowledgements
The work was supported by National Science Center, Poland Grant No. 2016/23/D/HS5/
00411 and inspired by IS1203—Citizen’s Health through public–private Initiatives:
Public health, Market and Ethical perspective (CHIP ME).

Competing interests
The author declares no competing interests.

Additional information
Correspondence and requests for materials should be addressed to D.K.-Z.

Reprints and permission information is available at http://www.nature.com/reprints

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in
published maps and institutional affiliations.

Open Access This article is licensed under a Creative Commons
Attribution 4.0 International License, which permits use, sharing,

adaptation, distribution and reproduction in any medium or format, as long as you give
appropriate credit to the original author(s) and the source, provide a link to the Creative
Commons license, and indicate if changes were made. The images or other third party
material in this article are included in the article’s Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the
article’s Creative Commons license and your intended use is not permitted by statutory
regulation or exceeds the permitted use, you will need to obtain permission directly from
the copyright holder. To view a copy of this license, visit http://creativecommons.org/
licenses/by/4.0/.

© The Author(s) 2020

ARTICLE PALGRAVE COMMUNICATIONS | https://doi.org/10.1057/s41599-020-0399-2

8 PALGRAVE COMMUNICATIONS | (2020)6:30 | https://doi.org/10.1057/s41599-020-0399-2 | www.nature.com/palcomms

  • Civil liability for damages related to germline and�embryo editing against the legal admissibility of gene editing
    • Introduction
    • The controversial CRISPR-Cas9 method and new conflicts of law between protected rights
    • International legal regulations and international community&#x02019;s opinions on gene therapy in embryos and reproductive cells
    • The legal nature of germline and embryo gene editing
    • Legal classification of genetic modifications of human embryos and reproductive cells
    • Prenatal damage and preconception injures
    • Conclusions
    • Data availability
    • References
    • Acknowledgements
    • Competing interests
    • Additional information

cells

Review

CRISPR-Cas and Its Wide-Ranging Applications: From Human
Genome Editing to Environmental Implications, Technical
Limitations, Hazards and Bioethical Issues

Roberto Piergentili 1 , Alessandro Del Rio 2,*, Fabrizio Signore 3, Federica Umani Ronchi 2, Enrico Marinelli 2

and Simona Zaami 2

����������
�������

Citation: Piergentili, R.; Del Rio, A.;

Signore, F.; Umani Ronchi, F.;

Marinelli, E.; Zaami, S. CRISPR-Cas

and Its Wide-Ranging Applications:

From Human Genome Editing to

Environmental Implications,

Technical Limitations, Hazards and

Bioethical Issues. Cells 2021, 10, 969.

https://doi.org/10.3390/cells10050969

Academic Editor: Tetsushi Sakuma

Received: 31 March 2021

Accepted: 19 April 2021

Published: 21 April 2021

Publisher’s Note: MDPI stays neutral

with regard to jurisdictional claims in

published maps and institutional affil-

iations.

Copyright: © 2021 by the authors.

Licensee MDPI, Basel, Switzerland.

This article is an open access article

distributed under the terms and

conditions of the Creative Commons

Attribution (CC BY) license (https://

creativecommons.org/licenses/by/

4.0/).

1 Institute of Molecular Biology and Pathology, Italian National Research Council (CNR-IBPM),
00185 Rome, Italy; [email protected]

2 Department of Anatomical, Histological, Forensic, and Orthopedic Sciences, Sapienza University of Rome,
00161 Rome, Italy; [email protected] (F.U.R.); [email protected] (E.M.);
[email protected] (S.Z.)

3 Obstetrics and Gynecology Department, USL Roma2, Sant’Eugenio Hospital, 00144 Rome, Italy;
[email protected]

* Correspondence: [email protected] or [email protected]

Abstract: The CRISPR-Cas system is a powerful tool for in vivo editing the genome of most organ-
isms, including man. During the years this technique has been applied in several fields, such as
agriculture for crop upgrade and breeding including the creation of allergy-free foods, for eradicating
pests, for the improvement of animal breeds, in the industry of bio-fuels and it can even be used as a
basis for a cell-based recording apparatus. Possible applications in human health include the making
of new medicines through the creation of genetically modified organisms, the treatment of viral
infections, the control of pathogens, applications in clinical diagnostics and the cure of human genetic
diseases, either caused by somatic (e.g., cancer) or inherited (mendelian disorders) mutations. One of
the most divisive, possible uses of this system is the modification of human embryos, for the purpose
of preventing or curing a human being before birth. However, the technology in this field is evolving
faster than regulations and several concerns are raised by its enormous yet controversial potential. In
this scenario, appropriate laws need to be issued and ethical guidelines must be developed, in order
to properly assess advantages as well as risks of this approach. In this review, we summarize the
potential of these genome editing techniques and their applications in human embryo treatment. We
will analyze CRISPR-Cas limitations and the possible genome damage caused in the treated embryo.
Finally, we will discuss how all this impacts the law, ethics and common sense.

Keywords: CRISPR-Cas; germline genome editing; human embryo; bioethics; biosecurity

1. Introduction

Although its first, serendipitous discovery dates back to 1987 [1], the potential of the
CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) system for genome
modification exploded less than 10 years ago [2], and earned Jennifer Doudna and Em-
manuelle Charpentier the Nobel Prize in Chemistry in 2020 [3]. The CRISPR system is the
way Bacteria and Archaea defend themselves from viral infections (Figure 1). It has been
shown [4] that natural occurring, defective variants of bacteriophages that are still able to
inject their genome inside the host but are unable to complete their life cycle, or are too
slow in doing that, or even wild type phages that are in some way inactivated during their
infection through other host defenses, are likely the principal route for bacteria to acquire
virus resistance through the CRISPR system. In brief, if the host survives a first viral attack
(Figure 1A), part of the phage genome may be incorporated inside a specific locus called
CRISPR, composed of short DNA repeats (length: 23–55 base pairs, bp) [5], thus becoming
a CRISPR DNA spacer (length: 21–72 bp) (Figure 1B). CRISPR loci are located at multiple

Cells 2021, 10, 969. https://doi.org/10.3390/cells10050969 https://www.mdpi.com/journal/cells

Cells 2021, 10, 969 2 of 24

sites inside the bacterial genome, (up to 23 in Methanocaldococcus sp.) and each locus may
contain several spacers (up to ca. 600 spacers in Haliangium ochraceum) [6]. This part of the
process is known as “adaptation”. If the bacterium is infected again by the same type of
virus (Figure 1C), the CRISPR locus, which contains usually ca. 50 spacers per CRISPR ar-
ray [5], is transcribed into a long RNA (CRISPR RNA, or crRNA) which is then cleaved into
short interfering crRNA (Figure 1D) (process: “expression”). These pieces, together with
the Cas protein and tracrRNA (see below), will bind to the newly injected viral genome,
promoting its degradation through a nucleolytic cut (Figures 1D and 2), a process known
as “interference”. The protein(s) responsible for the pairing and cut is/are a member of the
Cas (CRISPR-associated protein) family of proteins. They differ in their nucleic acid target
(DNA or RNA; single or double stranded; linear or circular DNA; other structural features),
the type of cut (blunt ends or overhangs) and the way of action; they are sorted in a total of
two classes, and divided in six types and 33 subtypes [7]. The systems that use more than
one Cas protein for DNA degradation belong to class I, while those belonging to class II use
only one, larger Cas protein. Cas9 endonuclease, one of the most used for genome editing,
belongs to class II. The Cas9 complex is formed by the Cas9 protein, which contains two
magnesium-dependent endonuclease domains responsible for the DNA cutting (namely,
HNH and RuvC) and two RNA, a crRNA and a trans-activating CRISPR RNA (tracrRNA),
the latter being necessary for crRNA maturation and cleavage through the formation of
a RNA duplex. Once processed by RNAse III, the complex crRNA/tracrRNA/Cas9 is
“guided” to their target, and thus the name “guide RNA” (gRNA). A crucial role in the
Cas9-mediated target degradation is played by the PAM (Protospacer Adjacent Motif)
sequence. PAM is a short DNA string (usually 3–8 bp long, length and sequence depending
on the bacterial species Cas9 comes from) adjacent to the cleavage site on the nontarget
strand (Figure 2B). Usually, it is located 2–6 nucleotides at the 3′ end of the DNA sequence
targeted by the guide RNA and the Cas nuclease cuts three nucleotides upstream of it.
Notably, PAM is not present in the crRNA sequence (i.e., it is part of the viral, but not the
bacterial, genome) and its absence is sufficient to impair the cleavage activity of the entire
complex. This allows the complex to recognize self vs. non-self DNA, thus avoiding that it
erroneously cuts bacterial DNA at the CRISPR locus. As such, the role of PAM is equally
pivotal during genome engineering, for precise DNA targeting.

The work of Doudna and Charpentier was aimed at modifying this system to tar-
get specific genomic sequences in virtually any organisms and promote their cleavage;
the modified system allows both gene knock out or knock in, depending on the repair
mechanism involved. In particular, Doudna and Charpentier re-engineered Cas9 complex
into a more controllable, two-component system by fusing the two RNA molecules into a
synthetic, single guide RNA (sgRNA) that is sufficient to find and cut the target DNA of
choice (Figure 2A, bottom), a solution explored and validated also by other groups in the
same year [8]. In this way, it is possible to create in vitro a custom-made sgRNA that drives
the Cas9 endonuclease to a specific target inside the genome. Once the sgRNA recognizes
the homologue sequence, Cas9 cuts the DNA. This triggers the DNA repair machinery of
the host cell, involving the error-prone NHEJ (non-homologous end joining) mechanism,
which induces some errors in the joining ends, thus inactivating gene function (knock out).
However, if a suitable exogenous template is provided (donor DNA), this can be used for a
homology-driven DNA repair, thus substituting the original sequence with another one of
choice, either (i) introducing a specific mutation in a wild type sequence (knock out), or (ii)
restoring the wild type copy of a mutated gene (gene modification), or (iii) inserting an
entire new gene or even multiple genes (knock in). Consequently, the donor DNA may
potentially be of any size, ranging from a few base pairs targeting a gene point mutation,
to larger elements containing one or more genes with specific promoters and additional
regulatory elements. The relatively ease of use and the efficient and precise targeting of
DNA sequences allowed to spread the use of this technique to modify genomic DNA in
virtually all living organisms, including plants, animals and even humans [9].

Cells 2021, 10, 969 3 of 24

Cells 2021, 10, x FOR PEER REVIEW 3 of 24

and additional regulatory elements. The relatively ease of use and the efficient and precise
targeting of DNA sequences allowed to spread the use of this technique to modify genomic
DNA in virtually all living organisms, including plants, animals and even humans [9].

Figure 1. The CRISPR antiviral defense system of prokaryotes. A population of viruses may contain both wild type (black)
and defective phages (gray; see text for further explanation). Upon infection of a defective bacteriophage as in the shown
example (A), part of the viral genome is inserted inside one of the CRISPR loci of the bacterial genome (adaptation, B). In
case of a second infection, even in case of a wild type phage (C) the CRISPR locus is transcribed (expression) and promotes
viral genome degradation by site-specific, Cas-mediated cleavage (interference, D).

Figure 1. The CRISPR antiviral defense system of prokaryotes. A population of viruses may contain both wild type (black)
and defective phages (gray; see text for further explanation). Upon infection of a defective bacteriophage as in the shown
example (A), part of the viral genome is inserted inside one of the CRISPR loci of the bacterial genome (adaptation, B). In
case of a second infection, even in case of a wild type phage (C) the CRISPR locus is transcribed (expression) and promotes
viral genome degradation by site-specific, Cas-mediated cleavage (interference, D).

Cells 2021, 10, 969 4 of 24Cells 2021, 10, x FOR PEER REVIEW 4 of 24

Figure 2. Mechanism of action of the Cas complex. (A): difference between natural (top) and engineered (bottom) guide
RNA (gRNA). The natural system is composed of two parts, crRNA (dark blue) and tracrRNA (light blue), which are
paired and drive Cas9 to target the invading viral DNA. The portion of the crRNA recognizing the target is indicated as a
dotted line. In the engineered form, the synthetic, single guide RNA (sgRNA) is one molecule that mimics the shape of its
natural counterpart, including the target recognition site (dotted line). (B): for genome editing, the sgRNA (orange) is
incorporated inside the Cas9 protein (green) and recognizes the double-stranded target DNA (black-grey) promoting the
pairing. In case of homology (pairing sequence length: 21–72 bp, indicated in gray) and in the presence of a PAM sequence
(red) on the target DNA, the Cas protein cuts the DNA 3 bp upstream of PAM, causing a double strand break, thus inac-
tivating target gene function. The repair of the target chromosome damage in eukaryotic cells usually employs the error
prone NHEJ (non-homologous end joining) mechanism; however, in presence of an exogenous DNA template (injected
with the Cas complex and sgRNA), the cell may fix the break through homologous-driven repair, thus introducing the
sequence of interest inside the genome.

1.1. Overview of the Possible Applications of CRISPR for the Improvement of the Human Qual-
ity of Life

The flexibility of the system opened up the possibility to modify host genome in very
diverse ways, to achieve important results in both eukaryotic and prokaryotic organisms.
Plant genomes can be easily modified with CRISPR [10]. In general, this technique allows
to have crop improvement either for yield increase [11], better nutritional content [12,13],
fruit ripening control [14,15], to create plants resistant to parasites, fungi and other pests
[16–19] or resistant to environmental stresses [20,21]. In addition, also more “aesthetic”
aims are pursued, such as more intense color, larger size or a more regular shape of fruits
and vegetables, improved flowering of ornamental plants [22–24] or even the creation of
vegetables with new flavors [25]. Some modifications also have a direct impact on human
health; beyond the nutritional content mentioned above, there are groups working to
lower the allergenic content of food, such as depleting allergens content of soybean [26]
or to create low/free gluten wheat for coeliac people [27,28].

CRISPR had been used to manipulate animal genomes to create strains that are re-
sistant to disease. This had been achieved for example for pigs resistant to Porcine Respir-
atory and Reproductive Syndrome virus (PRRSV) or the African swine fever or even to
produce coronavirus resistant pigs, by manipulating the pig proteins the virus recognizes
to invade cells. Pigs had been also manipulated to improve xenogeneic transplantation
and tolerance [29,30]. Similar gene editing approaches had been used also for improving

Figure 2. Mechanism of action of the Cas complex. (A): difference between natural (top) and engineered (bottom) guide
RNA (gRNA). The natural system is composed of two parts, crRNA (dark blue) and tracrRNA (light blue), which are paired
and drive Cas9 to target the invading viral DNA. The portion of the crRNA recognizing the target is indicated as a dotted
line. In the engineered form, the synthetic, single guide RNA (sgRNA) is one molecule that mimics the shape of its natural
counterpart, including the target recognition site (dotted line). (B): for genome editing, the sgRNA (orange) is incorporated
inside the Cas9 protein (green) and recognizes the double-stranded target DNA (black-grey) promoting the pairing. In
case of homology (pairing sequence length: 21–72 bp, indicated in gray) and in the presence of a PAM sequence (red) on
the target DNA, the Cas protein cuts the DNA 3 bp upstream of PAM, causing a double strand break, thus inactivating
target gene function. The repair of the target chromosome damage in eukaryotic cells usually employs the error prone
NHEJ (non-homologous end joining) mechanism; however, in presence of an exogenous DNA template (injected with the
Cas complex and sgRNA), the cell may fix the break through homologous-driven repair, thus introducing the sequence of
interest inside the genome.

1.1. Overview of the Possible Applications of CRISPR for the Improvement of the Human Quality
of Life

The flexibility of the system opened up the possibility to modify host genome in very
diverse ways, to achieve important results in both eukaryotic and prokaryotic organisms.
Plant genomes can be easily modified with CRISPR [10]. In general, this technique allows to
have crop improvement either for yield increase [11], better nutritional content [12,13], fruit
ripening control [14,15], to create plants resistant to parasites, fungi and other pests [16–19]
or resistant to environmental stresses [20,21]. In addition, also more “aesthetic” aims
are pursued, such as more intense color, larger size or a more regular shape of fruits
and vegetables, improved flowering of ornamental plants [22–24] or even the creation of
vegetables with new flavors [25]. Some modifications also have a direct impact on human
health; beyond the nutritional content mentioned above, there are groups working to lower
the allergenic content of food, such as depleting allergens content of soybean [26] or to
create low/free gluten wheat for coeliac people [27,28].

CRISPR had been used to manipulate animal genomes to create strains that are
resistant to disease. This had been achieved for example for pigs resistant to Porcine
Respiratory and Reproductive Syndrome virus (PRRSV) or the African swine fever or even
to produce coronavirus resistant pigs, by manipulating the pig proteins the virus recognizes
to invade cells. Pigs had been also manipulated to improve xenogeneic transplantation and
tolerance [29,30]. Similar gene editing approaches had been used also for improving animal
welfare after diffuse practices such as horn removal, male castration, or mulesing (reviewed

Cells 2021, 10, 969 5 of 24

in [31]). The poultry industry is deeply involved as well, for the creation of chicken and
quail lines that are resistant to specific disease-causing microorganism such as avian
influenza virus or avian leukosis virus; similarly, great efforts are in place for enhancing
muscle growth, thus increasing the weight and quality of human food (reviewed in [32]).
These animals are also used to create chickens that are efficient bioreactor systems for
producing valuable proteins in poultry species (see [32] and references therein). Similarly
to plants, also in animals CRISPR had been used for aesthetic purposes, such as the creation
of ‘micropigs’ to be used as pets [33]; the same company also works on koi carps with
custom size, color and patterns. Finally, also for animals, studies are running for allergen
depletion in food; examples include goat milk [34] and chicken eggs [35].

As for direct research in human health, and beyond, its use in embryo manipula-
tion that will be discussed in the next sections, we recall here briefly a few examples
of CRISPR/Cas9 used as a tool to treat human diseases. This system is very useful to
create disease models or to discover new etiological agents, allowing researchers to better
understand their biology. Examples include cancer, neurological diseases, cardiovascular
diseases, immunodeficiency, infectious diseases, sickle cell disease, hemophilia, metabolic
diseases, cystic fibrosis, retinitis pigmentosa, and several others (reviewed in [36–42]). In
particular, the treatment and characterization of cancer is very promising, and hundreds
of publications are available on this topic (for recent reviews, see for example [43–51]).
Pavani and collaborators showed its use in the treatment of beta-thalassemia [52] and
there are ongoing projects for the treatment of AIDS [53]. Several works exist describing a
possible CRISPR-based approach in the treatment of liver diseases such as viral hepatitis,
hepatocellular carcinoma and hereditary tyrosinemia type I (reviewed in [54]). This system
has also been used to specifically remove entire chromosomes from the genome. Zuo and
collaborators [55] demonstrated that it is possible to specifically remove sex chromosomes
from mouse cultured cells, embryos, and tissues in vivo, and also eliminate from mouse
cells human chromosomes that are otherwise stably transmitted through generations, such
as chromosomes 14 and 21; this is also possible in human cultured cells, such as aneuploid
cancer cells or cells from Down’s syndrome patients. This work [55] suggests the potential
use of CRISPR/Cas9 as a therapeutic strategy for human aneuploidy diseases caused by
supernumerary chromosomes.

We conclude this section by citing also work aimed at eradicating human, pest-derived
diseases such as malaria, by trying to control their vectors, i.e., mosquitoes [56]. This is a
typical case of artificial ‘gene drive’, i.e., the possibility to spread a particular gene (or a set
of genes) in a population by altering its probability of transmission through the generations.
The potential of this technique has been greatly enhanced by the use of CRISPR, thanks to
the possibility, in heterozygotes, to target (cut) the wild type gene and then repair it using
the mutated copy as a template; as a consequence, the probability to transmit the mutated
gene to the offspring is nearly 100%, instead of 50% as expected according to Mendel’s
laws. This approach overcomes the problems of inserting a particular mutation inside a
genome and waiting for it to naturally expand (if natural selection does not swipe it away),
and allows to genetically modify specific populations or even entire species. However, this
raises a plethora of ethical issues about the possibility to permanently modify the genome
of the target species worldwide, influence its ecology and that of the related species, the
impossibility to foresee all possible side effects of the manipulation of the targeted gene,
and possibly causing species extinction [57,58].

1.2. Technical Risks of Human Embryo Modifications

First of all, the fundamental difference needs to be stressed between modifying a
human embryo, for research purposes, and then discarding it after data collection, and
allowing its implantation. Both are highly controversial topics, but there is a general
consensus in opposition to the latter possibility. Laws currently in force, although still
insufficiently implemented, ban this practice in most countries.

Cells 2021, 10, 969 6 of 24

The first report of CRISPR-Cas driven editing of human embryos is only five years
old [59], indicating that this approach for gene therapy is just in its infancy, as someone
already noted [60,61]. Despite this, CRISPR potential and ease of use suggest that human
gene editing could become a possible way to treat—at least some of the—human genetic
diseases, either inherited (Mendelian diseases) or caused by newly appeared mutations
(as in idiopathic cancer) at the individual level, towards the implementation of precision
medicine [62]. This can be achieved in two ways: by deleting a wild type (wt), target
gene that predisposes to a disease or infection, or by substituting a mutated gene with
its wt counterpart. The efficiency of both approaches heavily depend on the number of
somatic cells that can be edited, thus the idea of manipulating the zygote or the embryo
at a very early stage, being much lower the number of cells to be targeted, or to act
on gametes to significantly reduce mosaicism [63]. An example of the first approach,
used in embryos, is the very debated experiment by the Chinese biophysicist He Jiankui
and his staff, aimed at preventing HIV infections by inducing a random deletion in an
otherwise wt protein coding gene (namely, CCR5)—we already described how this story
ended [64]; moreover, the several theoretical, technical and methodological pitfalls of those
experiments had been extensively analyzed in the past [65]. Obviously, most efforts are
based on the second method, i.e., restoring the wt sequence. The ethical and legal issues
of using these technologies in human embryo modifications will be extensively discussed
in the following sections and the possibility to modify the method to avoid this change
to pass on the next generation (“one-generation germline therapy”) has been discussed
as well [66]. In particular, the one-generation germline therapy is promising because
it would allow to treat an inherited condition in the somatic line leaving the germ line
almost untouched. In brief, it consists of introducing not just one gene template, but a
more complex transgene cassette hosting the following modules: (i) the gene of interest
under the control of a selected promoter (for example, the wild type copy of a mutated
gene under its physiological promoter); (ii) a DNA recombinase (such as Cre) under the
control of a germ line specific promoter; (iii) specific flanking sequences, recognized by
the recombinase; (iv) an insulator (a cis-acting DNA sequence) that avoids the unwanted
activation of the recombinase promoter in the soma, under the influence of the upstream
promoter. In the soma, the gene of interest is activated under the stimuli specific for the
promoter of choice, while the recombinase gene remains silent and the cassette is stably
inserted in the genome and passed through cell divisions. Instead, in the germ line, the
promoter of the recombinase is activated, and the protein promotes the cleavage of the
entire cassette at the flanking sequences (recognition sites), stimulating the DNA repair
machinery that would restore chromosome integrity but without the cassette and with
only a small DNA footprint, i.e., a few additional DNA nucleotides at the site of repaired
cleavage. The possibility to insert the cassette in a specific location where the footprint
would (likely) cause no health problem is, obviously, critical.

Beyond possible, subtle mutations that can be a byproduct of off-target Cas9 action,
and the possibility to induce chromosome alterations in model embryo systems [67,68]
and in human somatic cells [69,70], potential risks for whole genome integrity had been
highlighted in at least three reports published in 2020, indicating that large and unexpected
karyotype modifications can occur, causing potentially severe health problems, that might
arise during CRISPR use in human embryo treatment. The first report we discuss is the one
by Alanis-Lobato and collaborators [71]. On the basis of previous work, suggesting that
gene editing is occurring through interhomologue homologous recombination (IH-HR)
driven by the maternal allele [63], the authors tried to replicate the experiments using
as a target the pluripotency factor OCT4, encoded by the POU5F1 gene on the p-arm
of chromosome 6. Despite the overall good results (i.e., restoration of the wt allele),
the authors did not obtain the PCR amplification of the supposedly edited gene. An
in-depth analysis revealed that almost one third of the targeted samples (8/25) showed
abnormalities on chromosome 6. In particular, the authors were able to identify diverse
rearrangements, including segmental loss or gain of chromosome fragments next to the

Cells 2021, 10, 969 7 of 24

POU5F1 locus (p-arm), whole gain of chromosome 6, and segmental gain on the q-arm,
totaling approximately 16% of samples and spanning 4 kb to at least 20 kb in length. In
addition, also the search for loss of heterozygosity (LOH) was positive. In the second
paper, Zuccaro and collaborators [72] reported the results of substituting the EYS locus
(mapping at 6q12, associated with retinitis pigmentosa and causing blindness, [73]) in
embryos. The starting point was the use of spermatozoa from a man homozygous for
an intragenic deletion causing the formation of a premature stop codon at exon 34 of the
gene, exploiting the fact that DSB (double strand break) mediated recombination between
homologous chromosomes in eggs is apparently quite efficient and preferentially uses the
maternal allele as a template [63]. Indeed, Zuccaro and collaborators found that 17 out
of 20 analyzed samples had the restoration of the wt genotype, suggesting either cell
type differences in DSB repair and/or cell survival after a chromosome break. To better
understand what happened, the authors performed several additional analyses, finding
that in many cases the wt homozygous genotype for EYS was achieved with a contemporary
LOH of the surrounding regions of chromosome 6. This LOH was caused by different
gross chromosome 6 segmental rearrangements, including (i) distal 6q arm loss; (ii) 6q arm
gain (with the consequent increase of paternal genes copy number) and a contemporary
movement of the EYS gene far from flanking sequences or with breakpoints inside the EYS
locus, resulting in the impossibility to amplify and detect it in the samples; (iii) monosomy
of the maternal chromosome 6, i.e., complete loss of the paternal homologue; (iv) gain of
one or more paternal chromosome 6. This occurred in embryos either injected with Cas9
RNP at fertilization or at the 2-cell stage, in 19 out of 20 samples screened. In conclusion,
the authors wrote that in their system—i.e., very early stage of development—the loss of
paternal alleles is a ‘common outcome’ and that this occurs through aneuploidy, not efficient
interhomolog repair. Interestingly, additional chromosome aberrations and aneuploidies
involving chromosome 16 were scored, mostly in the form of mosaic; at least some of
them are due to off-target CRISPR-mediated cleavage. All together, these results suggest
that in human preimplantation embryos DNA repair pathways might be—or behave—
different(ly) from other cell types (in [72] there is evidence of the involvement of the
MMEJ—microhomology-mediated end joining—pathway [74]), which could explain the
high incidence of endogenous genome instability. The third paper we discuss is by Liang
and collaborators [75] in which no karyotype alteration is presented, yet extensive gene
conversion is reported as a consequence of CRISPR-mediated DNA damage. Additionally,
in this case the authors performed experiments on preimplantation heterozygous embryos
and using a heterozygous mutation in exon 22 of the MYH7 gene located on chromosome
14, implicated in familial hypertrophic cardiomyopathy (HCM). The authors noted that
DNA damage in this system is mainly repaired through gene conversion, a form of HDR
(homology directed repair) that uses an exogenous homologous sequence as a template for
repairing. Interestingly, HDR is usually far less used in cells than NHEJ, also because it can
occur only after DNA replication while the other may act throughout the cell cycle. The
main difference between classical HDR and gene conversion is that, after strand annealing,
the repair machinery may extend the copy of the template beyond the microhomology
region used for the first strand invasion, thus making the template and the invaded
strand identical (noncrossover recombination), and consequently causing a LOH that
might extend for several kilobases and, in some instances, entire chromosome arms. In
addition, the search for chromosome deletions in those cells was negative, confirming
the gene conversion mechanism involved in this experiment and indicating that ‘a large
percentage of DSBs (41.7%, 50/120), are resolved by gene conversion’ [75]. Similar results
were obtained targeting the MYBPC3 and LDLRAP1 loci [75].

Despite the different results obtained by Liang and collaborators [75] compared to the
previous two [69,70], i.e., LOH as main output but no chromosome deletion vs. karyotype
alterations including deletions, duplications and chromosome loss, all three works point to
the same conclusions: (i) embryos respond differently to DNA damage when compared
to somatic cells upon CRISPR-Cas9 manipulation, and the reasons of this difference are

Cells 2021, 10, 969 8 of 24

still far from clarified; (ii) the output of this manipulation is still largely unpredictable and
amply variable, including several different types of genomic damage; (iii) in all cases, the
percentage of damaged cells is extremely high—around half of them show gross alterations.
For all such reasons, we believe that using in embryos the same protocols used for somatic
cell editing is presently inadvisable. Taken together, these results are against the use of
CRISPR-Cas9 for manipulating embryo genomes because of the formation of DSBs that
might either remain unrepaired or being erroneously repaired, through mechanisms that
are still poorly understood, and the extensive LOH that might reveal additional recessive
mutations inside the egg genome. It would be advisable to opt for alternative gene editing
methods that do not cause DNA damage [76]; nonetheless, even that approach needs
further testing and verification.

2. Beyond Therapeutic Safety and Efficacy, Genome Editing Entails Polarizing Ethical
and Legal Quandaries

Ethical concerns about CRISPR-based genome engineering techniques arise from vari-
ous lines of reasoning. Firstly, the scope and limitations of CRISPR technology, including
the risks stemming from limited on-target editing efficiency, mosaicism [77], and inaccurate
on- or off-target editing, are still largely unknown [78]. Such flaws have been documented
in CRISPR experiments with animals as well as human cell lines. Still, as the technology
is gradually honed and perfected, such concerns may no longer be warranted over time.
Nonetheless, ethically tenable decision making in biomedicine needs to be informed on
an empirical basis, by means of a thorough appraisal of risk–benefit ratios. To that end,
ethical decisions have to be grounded in a thorough analysis of possible outcomes, the
likelihood of each manifesting itself, and the purposes and possible justifications that
determine the end results. When it comes to CRISPR genome engineering technology,
however, assessing potential risks and benefits with an acceptable degree of accuracy may
be extremely hard, given the difficulties of making reliable predictions about the future of
a genetically edited organism. It is unclear whether modified organisms will be affected
indefinitely and whether and to what extent the edited genes will be transferred to future
generations, potentially affecting them in unexpected ways. An accurate risk–benefit
analysis is therefore significantly complicated, not only by technical limitations, but also
on account of the complexities inherent to biological systems.

2.1. Current Lack of Understanding Stands in the Way of thorough Risk–Benefit Analysis

Critics have in fact pointed out that even if genome-editing procedures are successfully
carried out, and the expected functional effect is achieved in a timely fashion, genetic
information and biological phenotypes are related in ways that are not yet fully understood.
Genetic pleiotropic effects in fact constitute a primary source of phenotypic variability in
humans. It is therefore still undetermined in what way CRISPR-Cas systems could bring
about pleiotropic effects, even though we were able to successfully establish that genes act
as prominent causal factors in disease development. Some disease phenotypes, it is worth
stressing, can potentially be altered or even obliterated by pleiotropic effects [79]. Hence,
editing a gene in germline and/or somatic cells may bring about unpredictable biological
consequences. It is in fact the complex regulatory actions of numerous genes which
determine a wide array of biological traits. That makes it extremely difficult to “engineer”
a biological phenotype at the level of a whole organism [80]. In other words, a single
gene is very unlikely to be the only factor that molds and develops a complex biological
trait. The emergence of a biological phenotype is in fact determined by environmental and
epigenetic factors and several other genetic regulatory factors, e.g., additional genes or
distal regulatory elements (such as enhancer or repressor elements). By virtue of that, a
thorough understanding of other independent variables contributing to the phenotype’s
instantiation is a key element, in addition to genetic modification. Such an understanding
is nevertheless still far from being complete enough in many normal and disease processes.
Since uncertainty still lingers as to how gene expression and modification affect and drive
complex biological outcomes, a thorough risk–benefit analysis is difficult to produce [81].

Cells 2021, 10, 969 9 of 24

In addition, as previously noted, a seemingly unsolvable ethical and legal debate has
been ignited by the potential application of CRISPR technology on human embryos [82].
Such a controversy is further complicated by the lack of consensus among ethicists and
legal scholars as to the status of the human embryo itself [83,84]. Even though many in
the scientific community contend that experimentation on human embryos after 14 days
is ethically intolerable, it is all but impossible to find common ground for determining
the status of a human embryo and when it acquires “personhood” [85,86]. That is the
fundamental reason why many nations regulate medically assisted procreation via in vitro
fertilization with varying degrees of restrictions [87].

2.2. The Unsolved Quandary of Embryonic Status

If embryos are to be ascribed personhood status, then they are entitled to have their
inalienable human rights upheld. If, on the other hand, they are deemed as something
in between, i.e., less than human beings but more than mere pools of cells, what moral
rights should they have acknowledged, if any? Certainly, some point out that the first
experiments using CRISPR to edit human embryos occurred in 2015, and since then, only
few teams around the world have focused on the process and its potential [85], but recent
studies have highlighted an underappreciated risk of CRISPR–Cas9 editing: if embryos are
deemed to have the right to at least some degree of legal safeguards, such safety concerns
are likely to significantly inform the ongoing debate on the matter. In light of such major
unsolved controversies, some have called for an international moratorium on all embryo
editing [88,89], and some countries, including Canada, already have policies that ban
human-embryo gene editing, irrespective of whether or not the edited embryo would
be meant for implantation [90]. In the United States and Britain, on the other hand, an
intermediate regulatory approach has been chosen. The US Food and Drug Administration
views any use of CRISPR/Cas9 gene editing in humans as gene therapy, regulated by
the FDA’s Center for Biologics Evaluation and Research (CBER). Clinical studies of gene
therapy in human beings therefore require the submission of an investigational new drug
application (IND) before they can be legally initiated in the United States. In addition,
marketing gene therapy products calls for the submission and approval of a biologics
license application (BLA). As a result of such requirements and restrictions, operating a
private lab, with private funds, and conducting nonclinical, human gene therapy research
is not illegal. Nonetheless, marketing such therapeutic options in the US would require
FDA approval in terms of clinical studies and marketing. As far as it could be determined,
no instances exist of a germline gene therapy product in the US; only somatic cell gene
therapy products have been granted approval; currently, federal law prevents the FDA
from reviewing or approving any application involving manipulated human embryos [91].
Again, it is necessary to draw a clear distinction between embryo editing for research
purposes and the implantation of such edited embryos, which is ethically far more con-
tentious. In the United Kingdom, for instance, the use of genome editing in embryos for
the purpose of implantation is banned, albeit gene editing on discarded IVF embryos is
lawful, provided that such embryos are destroyed immediately afterwards. In vitro culture
of human embryos beyond 14 days after onset of embryo creation, i.e., after the appearance
of the primitive streak, is prohibited: such a ban is enshrined in the Human Fertilisation
and Embryology (HFE) Acts of 1990 and 2008 [92]. In Italy, specific provisions in law
40/2004 recognize the embryo as having rights from the moment of fertilization [93]. The
law prohibits the use of embryos for any research unless it is specifically aimed towards
improving the therapeutic and medical condition of the embryo itself [94,95]. Critics have
pointed out the apparent paradox behind such a restriction, considering that in vivo em-
bryos can be terminated up to 24 weeks through voluntary termination of pregnancy [96].
Some may in fact find it confusing that embryo research is required to stop so much ear-
lier, particularly in light of the fact that it is arguably more ethically sustainable to use
abandoned supernumerary embryos for research purposes that could benefit humanity,
than to just dispose of them. While abortion ethics is beyond the scope of this review, it

Cells 2021, 10, 969 10 of 24

is worth pointing out that in these two scenarios, different fundamental goals are in play:
legal termination of pregnancy stems from the need to uphold the right of women to have
a choice and be in control of their body, whereas in vitro embryo research does not entail
that issue. As for embryo experimentation, such intermediate regulatory approaches bear
witness to the current uncertainty as to how strictly such techniques ought to be regulated,
for the purpose of striking a balance between upholding bioethics precepts and fostering
scientific progress for the common good. Nonetheless, nations with more lax, ambiguous
or nonspecific regulatory frameworks governing new biomedical technologies may result
in a worrisome “maverick” scientific environment in which untested techniques are made
available. That has been found to be the case with mitochondrial replacement therapy
(MRT), a form of nuclear transfer used as a germline therapy and believed to prevent the
transmission of mitochondrial diseases and increase the likelihood of success in pregnan-
cies [97]. Although MRT is banned in many countries due to its still dubious safety, clinics
in Spain, Albania, Russia, Ukraine, and Israel have been found to offer the procedure [98].
That said, and irrespective of how individual countries decide to govern such techniques,
the issue of whether scientists should seek to edit human embryos to prevent genetic
diseases is controversial in itself, because the genomic change which it creates is permanent
and may be passed down for generations. Even if embryo experimentation should be
deemed justified, by virtue of its potential benefit to the embryo itself and others, embryos
obviously cannot grant informed consent, but are still liable to experience life-altering
consequences which can extend throughout their lifetimes and affect future generations as
well. Besides, as mentioned earlier on, the enforcement and practice of both ethical precepts
and legal provisions are inextricably linked to a set of notions that are hardly carved in
stone and universally acknowledged. Hence, the time at which a human life (whether
embryo or fetus) is deemed a fully-fledged human being has far-reaching ramifications that
encompass the crucial realms of health care, law- and policy making and the inalienable
right of individual autonomy of all humans [99]. There are no easy answers in our ever
more culturally and ethically diverse societies: one-size-fits-all approaches seem doomed
to fail, yet finding common ground is vital. If human embryos are to be deemed human
beings with full personhood status, major implications ensue. That is the perspective
espoused by Catholic doctrine, best exemplified by the late Pope John Paul II, who in 1995
famously stated that “the mere probability that a human person is involved would suffice
to justify an absolutely clear prohibition of any intervention aimed at killing a human
embryo” [100]. That approach does not differentiate between embryos edited for research
purposes and edited embryos to be implanted. Conversely, prominent philosophers such
as Kant, Locke and Fletcher have laid out criteria for identifying personhood closely tied
to self-awareness, the capability to relate to others, self-control, rationality, and the use of
memory, among others [101–104]. On the other hand, all such complexities and apparently
irreconcilable views notwithstanding, there is no denying that banning or constraining
research on human embryos could put a damper on scientific progress and stymie the
development of therapies that could defeat currently untreatable diseases. Would that not
be a moral and ethical imperative outweighing previously reported concerns? Again, no
easy answers.

2.3. Broadening Fields of Application

Early research seeking treatment for neurodegenerative diseases has shown that
CRISPR can even be used in combination with other elements, e.g., nonpathogenic viruses,
to help improve target specificity on any genomic sequences [105,106]. CRISPR-Cas9
may also be pooled with multiple guide RNAs, allowing the editing of multiple genes in
one step. This pool of guide RNAs allows the Cas enzyme, which cuts the DNA, to be
guided to many different parts of the genome. It could prove valuable to target multiple
genes at once through pooled guide RNAs in order to shed light on systemic effects, e.g.,
response to therapy or how metabolism is affected [107,108]. Another ethically sensitive
research field that holds great promise involves the investigation and development of the

Cells 2021, 10, 969 11 of 24

therapeutic potentials of stem cells and the biology of totipotent cells, which can divide
indefinitely and give rise to any of the 220 cell types found in an embryo as well as extra-
embryonic cells (i.e., the placenta) and pluripotent cells (which can give rise to all cell
types of the body except for the placenta) [109,110]. Totipotent and pluripotent cells are in
fact not found in any viable human tissue sources other than embryos. In fact, embryonic
cells within the first couple of cell divisions after fertilization are the only cells that are
totipotent, derived from the early cells of a fertilized egg, while pluripotent cells are found
in the inner cell mass of blastocysts [111,112]. In the current pandemic scenario, studies
have found that CRISPR/Cas9 has potential applications to human-induced pluripotent
stem cells (hiPSCs), ranging from gene therapy to the induction of the immunological
response to specific virus infection, such as HIV and SARS-Cov-2 itself [113]. The potential
applications of CRISPR/Cas9 and hiPSCs in antiviral response, including SARS-Cov-
2 research, are centered around a testing platform meant to replicate the human lung,
differentiating wild type (WT)-hiPSCs into pneumocytes type II [114], and treating them
with pseudoviruses capable of replicating SARS-Cov-2 infection [115]. Although future
prospects for therapeutic applications are still far from conclusive, especially as far as
mutation-prone viruses such as SARS-Cov-2 are concerned, that may certainly be one
way in which gene editing could be harnessed for the repression or the upregulation of
genes that play a role in viral activity, in addition to the introduction of polymorphisms
that could protect against or predispose to the viral infection [116]. Moreover, cells that
have undergone editing can be used to test the capacity of a number of compounds to
fight the infections. A rather versatile and apparently effective approach has recently been
devised for the purpose of targeting viral RNA through CRISPR/Cas9; researchers are
looking into the possibility of specifically using it on SARS-CoV-2 RNA genome, in order to
constrain its ability to reproduce. That prospect could pave the way for a great opportunity
to effectively deal with fast-evolving viruses that have the capability to develop resistance
rapidly and give rise to tragic consequences such as overwhelmed health care systems
and ethical quandaries in the delivery of care [117,118]. In light of the above instances
of therapeutic applications, it is not hard to figure out why it is so essential to strike
a balance between bioethics precepts and the needs and fast-developing dynamics of
scientific research, for the sake of public health. On the other hand, several roadblocks
still lie on the path of techniques such as the Prophylactic Antiviral CRISPR in huMAN
cells (PAC-MAN). Research has shown that such methods could inhibit RNA viruses in
human cells through CRISPR-Cas13d for the purpose of viral gene expression inhibition
through targeted RNA degradation. Difficulties in that approach include the lack of a
reliably effective delivery mechanism, since the component parts of CRISPR are just too
large to enter the target cells. Progress is in fact being made in terms of developing lipitoids,
i.e., synthetic molecules to be used as CRISPR delivery system [119]. Still, clinically viable
CRISPR-based potential therapeutic options such as PAC-MAN [120] may still be far from
mainstream therapeutic use, in terms of accuracy, applicability and costs, hardly feasible
solutions for the current scenario.

3. When the Line between “Therapy” and “Enhancement” Is Blurred

Provided that genome editing techniques undoubtedly have therapeutic value and
hold great promise in terms of providing new forms of treatment for incurable diseases,
prominent bioethicists have advocated for it by pointing out that they may even be ethically
desirable [121,122]. In fact, gene-editing technologies could even result in fewer embryos
being destroyed in assisted reproduction procedures. At the time being, if a carrier of a
genetic disease seeks to have a child who will not have the same condition, the carrier often
resorts to IVF and preimplantation genetic diagnosis (PGD) for the purpose of selecting
an embryo unaffected by that condition. Still, it behooves us to bear in mind that IVF
often entails producing a considerable number of supernumerary unwanted embryos,
which will eventually be destroyed even if viable. This ethically objectionable practice
would be obsolete if safe and effective gene-editing technologies were available to all;

Cells 2021, 10, 969 12 of 24

carriers of genetic diseases in fact would no longer have to produce large numbers of
surplus embryos in order to make it possible to have children not affected by the genetic
condition of which their parents are carriers, e.g., autosomal recessive mutations [123],
responsible for autosomal recessive monogenic diseases such as cystic fibrosis and sickle
cells anemia [124,125]. The feasibility of such prospective applications has, however, been
called into question, given the potential for harm, off-target effects or mis-edits that could
make any such theoretical advantages ultimately unattainable [126]. Plus, it is unlikely
for gene editing to replace PGD, since embryo testing will always be necessary, as will
embryo selection. Testing will in fact still have to be performed following the editing
intervention, to minimize the risks arising from the adverse effects of germline genome
editing. Hence, as long as editing techniques are still quite far from 100% accurate, embryos
might again be discarded even after the intervention [127]. As the United States National
Academy of Science (NAS) stated in a report issued in 2020, in fact, neither the currently
available editing technologies, nor sequencing embryonic DNA, aimed at keeping in check
possible off-target effects, are as yet reliable enough, hence strict criteria ought to be met
for couples to avail themselves of such techniques in order to have biological children that
will not inherit the mutation. Key factors include the nature or severity of the disease,
awareness of correlations between genotype–phenotype, and the availability of already
mentioned alternative options, such as prenatal genetic testing (PGT) and noninvasive
genetic diagnosis (NIPT) [128]. Not all possible applications of heritable human germline
editing can rely on responsible translation pathways, hence a thorough risk–benefit analysis
rests on multiple complex variables from which an assessment has to be worked out:
disease severity, the genetic situation of the couple, the mode of inheritance of the disease,
the nature of the proposed sequence change, and the availability of alternatives [129].
Outside of its therapeutic potential, however, an exhaustive discussion as to the ethics of
gene editing cannot leave out the all too relevant aspect of possible applications aimed at
biomedical enhancement. Currently, the primary means for the enhancement of cognitive
functions and/or physical performances is pharmacological in nature. Pharmacological
cognitive enhancement refers to the off-label use of drugs or supplements by healthy
individuals in order to augment performance. Such drugs are referred to as nootropics or
“smart drugs”. Nootropic use is universally and increasingly popular, both by professionals
and students, in addition to recreational users [130–132]. Among the most widespread
substances are methylphenidate [133], fluoxetine, and sildenafil [134,135]. Pharmacological
enhancers already pose a major and immediate challenge for regulators and policy makers,
which would only be compounded if the use of genome editing techniques were used to
enhance the capabilities of human beings yet to be born. Hence, through such interventions,
individual traits and characteristics could be selected during its gestation according to the
wishes and decisions of third parties. That would amount to a sort of “germline genetic
enhancement”, i.e., overcoming the limitations of the human body and mind, going well
beyond the therapeutic use meant to restore or sustain health [136]. Even though the
dream (or according to opponents, the nightmare) of being in full control of human biology
might still be far off, gene editing research has already made giant strides [137,138]. Critics
have expressed concerns that this approach would generate the need for a reflection on
the ethical, social and legal implications of these techniques and their implementation in
society. Fears that scientists may one day start “playing God” are not new after all: decades
ago, in vitro fertilization (IVF) has been harshly criticized by many as “unnatural” or taking
on prerogatives that do not belong to men [139–141]. IVF techniques are still morally and
ethically controversial and often restricted, as are fertility preservation procedures capable
of prolonging the time frame in which parenthood is achievable [142–145].

3.1. Is Enhancement Headed down a Dangerous Path?

Despite the potentially enormous benefits such techniques may yield, detractors have
gone so far as to liken gene editing-based enhancement to eugenics: enhancement is after
all an intervention aimed at improving capabilities, functioning or appearance that are

Cells 2021, 10, 969 13 of 24

already within the normal range. Eugenics was steeped in utilitarian philosophy precepts
based on Darwinian natural selection. By virtue of that, eugenicists encouraged those
deemed “fit”, typically belonging to middle and upper classes to have large families,
whereas the destitutes, deemed “unfit”, were to breed less. Over the 20th century, it became
clear that adjudging moral worth on the basis of mental or physical fitness would lead to
atrocities of horrific magnitude, such as forced sterilization, euthanasia and genocide [146].
Although from the late 19th century eugenics was advocated for in Western countries, from
the United States (suffice it to say that the Rockefeller, Carnegie, and Ford Foundations
actively funded eugenics research) to the United Kingdom, Germany and Sweden, such
ethically and scientifically indefensible thinking was ultimately exposed as folly, and
rightly rejected. Nowadays, however, prominent supporters of human enhancement
include academics and philosophers who argue that the aspiration to augment human
capabilities, even at the genetic level, is grounded on solid science, individual consent and
determination to improve oneself, thus devoid of any element of coercion [147], and that
even a moral obligation exists to produce the best possible children [148]. Those are the
underpinnings of the so-called Principle of Procreative Beneficence (PPB), a philosophically
complex and contentious framework [149], which should in our view be rejected, since it
would entail ascribing degrees of human value on the basis of capabilities or “desirable”
attributes and quite possibly, placing lower moral value on the disabled or on those less
endowed [150–152]. The alleged “obligation” to pursue such objectives is an integral
part of the transhumanist doctrine, whose followers have espoused nontherapeutic gene
editing and other new and emerging technologies for enhancement purposes [153]. In fact,
transhumanists have embraced the desirability and inevitability of germline and enhancing
gene therapies, while also calling for public financing of research and a regulatory process to
ensure their safety [154]. Aside from extremes such as the notion of a “morally obligatory”
enhancement of those yet to be born, three aspects should in our view be defined and
specified when weighing the ethical feasibility of genome editing applied to reproductive
technologies: the already mentioned moral status of embryos that should be enhanced,
the legal status of the individual poised to be enhanced, and the responsibility of the
agents carrying out the enhancement interventions. It is in fact undeniable that genetic
enhancement can impact inalienable human rights such as identity, dignity, and good
lifetime of individuals particularly vulnerable and without autonomy, such as embryos or
a newborn [155]. Ultimately, as remarked in the Second International Summit on Human
Genome Editing [156], the risks and benefits of germline genome editing are not fully
understood and clarified yet, at least not enough to allow germline genome editing to
proceed. That being said, expanding scientific understanding and recent research seem to
point out that it may be time to start outlining what a well-balanced pathway for future
clinical use should entail [157]. Mapping out the way forward would call for greater
awareness, further discussion and consensus building on multiple highly complex issues;
yet, those issues need to be thoroughly dealt with, since over time, the definition of a
tenable and consistent clinical pathway for germline genome editing could become both
morally essential and necessary to prevent irresponsible practices that would violate both
clinical ethics and the core values on which our civilization is built.

3.2. Gene Drives: A Potential Threat to the Environment and Ecosystems?

As previously mentioned, in addition to its human therapeutic applications, CRISPR
has already been used to modify animals [158], insects [159], plants [160], and microor-
ganisms [161]. Successful trials [162] have also involved bacterial pathogens, such as the
rather recent characterization of a new type 2 CRISPR system from Francisella novicida
(FnCas9), an unofficial fourth subspecies of the Francisella tularensis, the causative agent of
tularemia. Specifically, FnCas9 has been found to direct homology-directed repair (HDR)
as well as nonhomologous end-joining–mediated DNA repair, while giving rise to a higher
rate of HDR and extremely low off-targeting. A substantially high degree of specificity
has been shown in terms of binding to its intended targets, which demonstrates that the

Cells 2021, 10, 969 14 of 24

horizon of genome editing technologies is poised to broaden considerably further [163].
Although such uses may not appear to create new ethical problems in such contexts, a real
risk does exist that CRISPR could run counter to regulations governing the creation and
release of genetically modified organisms (GMOs). Generating modified organisms could
be valuable in terms of possibly eradicating infectious diseases through the elimination
of their vectors and invasive species [164]. In 2014, Esvelt and coworkers suggested that
CRISPR/Cas9 could be harnessed to build endonuclease gene drives [165], and studies doc-
umenting the successful engineering of CRISPR-based gene drives in Saccharomyces [166],
Drosophila [167], and mosquitoes [168] were published soon after. Inheritance distortion
has been found by all four studies, efficient over future generations. More recent exper-
iments demonstrate that a CRISPR/Cas9-based gene drive can spread a targeted gene
throughout nearly all of laboratory populations of yeast, fruit flies, or mosquitoes [169].
Among the noteworthy examples of such research have involved the Aedes aegypti mosquito,
which spreads dengue fever, and even some Anopheles subspecies [170,171], which carry
the Plasmodium parasite [172,173]. Biotech researchers have been resorting to “gene drives”,
aimed at stemming disease transmission through the editing of mosquitos for the ulti-
mate purpose of making them incapable of carrying the disease, inducing sterility in male
mosquitos, or shortening the lifespan of their offspring [174]. Although such methods
could prove extremely valuable in eradicating deadly diseases (malaria has struck 228
million people in 2018, causing upwards of 400,000 deaths [175]), environmental concerns
have been expressed that they could cause the extinction of entire species, with harmful
environmental consequences. Gene drive is a powerful tool that can make the edited trait
heritable. Hence, genetically modified organisms which are released into the environment
find wild-type mates, and their offspring is estimated to have a 50% chance of inheriting
the modified genes [176]. Nonetheless, such fears (and hopes) appear to be overblown,
being largely based on anecdotal, speculative theories, rather than solid empirical research
and analysis. The progress in such techniques notwithstanding, there is still no proving
how dangerous or promising they may turn out. While the introduction of small numbers
of edited mosquitos or other pests is unlikely to cause major effects, gene drive is capable
of copying mutations made on one chromosome by CRISPR to its partner chromosome,
thus passing the edited genome on to future generations [177]. Although those dynamics
could be instrumental in greatly limiting transmission rates of various infectious diseases,
gene drives do entail considerable risks to the environment and ecosystems: they have in
fact the potential to decimate entire species [178,179], disrupting food chains, or lead to the
uncontrolled proliferation and spread of invasive species, without sufficient containment
mechanisms [180–182]. It is therefore of utmost importance to identify the knowledge gaps
that could thwart or complicate attempts to control CRISPR-modified species, as there
may be unintended effects such as altering gene flow within a population [183]. Even
though attempts have been made to model gene drives within populations [184], it is quite
hard for such assessments to encompass ecosystems rife with complexities and extremely
dynamic [185,186]. In order to face such risks and potential threats to ecosystems and
biodiversity, research has been looking into ways to make gene drives reversible, through
self-exhausting forms of CRISPR-based gene drive that have been dubbed “daisy-chain
drive” [187,188], in order to achieve reversible alterations, meant to be limited in space and
scope [189].

3.3. Should CRISPR Be Viewed as a Potential Biosecurity Hazard?

In 2017, the US Defense Advanced Research Projects Agency (DARPA)’s Safe Genes
programme announced the allocation of USD 65 million to fund research into how to control,
counter and reverse gene drives [190,191]. The program has been devised to pursue three
fundamental technical goals: developing technologies for spatial, temporal, and reversible
control of genome editors applied to living organisms; molecular countermeasures aimed
at creating viable prophylactic and treatment solutions to stave off or constrain genome
editing in organisms and preserve genomic backgrounds in populations; the capability

Cells 2021, 10, 969 15 of 24

to root out undesired edited genes from ecosystems, thus restoring them to their genetic
baseline state. It is stressed that the development of these powerful techniques has to
unfold and progress within a strict ethical framework which prioritizes safety and progress
achieved in a responsible fashion [192]. DARPA program manager for the Safe Genes
program has likened the measures and technologies to control, inhibit, or reverse genome
editing to “brakes in cars”, which are not meant to make people drive slowly, but rather to
enable them to drive fast and stop when they need to. The manager went on to remark that
“the steep drop in the costs of genomic sequencing and gene editing toolkits, along with the
increasing accessibility of this technology”, has engendered even greater opportunity to
experiment with genetic modifications. In addition, this convergence of low cost and high
availability means that applications for gene editing, both positive and negative, could
arise from individuals or state entities that operate outside the scope and supervision of the
traditional scientific community [193]. The ethical implications of such remarks are quite
startling, in light of the potential such techniques indisputably possess. In fact, CRISPR can
be accurately viewed as a dual-use technology: although it has a broad range of potential
benefits to medicine, science, and public health, it can be used with malicious intent. As
pointed out in a 2018 report [194] by the United States National Academies of Sciences,
Engineering, and Medicine, requested by the U.S. Department of Defense, it is critical
that biotechnology continue to be researched and developed, but at the same time and
just as importantly, its innovative applications in the scientific and public realms ought
to be thoroughly evaluated via a specifically targeted assessment framework. Possible
risks were also identified in the report such as the re-creation of known pathogens (e.g.,
smallpox) as one of the gravest concerns, whereas the creation of a new pathogen was
deemed a lower risk. As a matter of fact, CRISPR could make it possible to edit a given
pathogen so that it will acquire the degree of virulence of another pathogen, or it might
enable rogue scientists to replicate a known pathogen whose genome has been published.
In light of those areas of concern, the misuse of CRISPR deserves to be acknowledged as
having the potential to pose a real threat to biosecurity. How effective such oversight efforts
at the national level will prove remains to be seen. Certainly, there are no silver bullet
solutions that can be put in place in order to lower the risks of misuse to near zero [195].
Awareness of that fact makes it even clearer just how urgent the need is for targeted, globally
shared regulations meant to define standards for the testing and environmental release of
GMOs. Such innovative applications are in fact inadequately governed by current national
and international regulations, particularly in terms of guidance and oversight. Those
shortcomings are likely to induce public mistrust in the safety and viability of gene editing
and GMOs, as well as in the regulatory bodies and agencies charged with overseeing such
techniques. One of the main concerns in that respect is that public misunderstanding and
mistrust of GMOs will hinder scientific progress and the highly beneficial applications of
CRISPR. Thoroughly weighing, planning—and above all, getting right—the regulations
and research ethics for these applications of CRISPR could go a long way towards creating
an ethical framework for human germ line editing that can be shared at least by nations
with common core values.

3.4. Towards a Shared Regulatory Framework?

A common set of standards to be applied to gene editing is therefore more essential
than ever and needs to be based on the awareness that research ethics needs to be based on
a painstaking process of gaining informed consent from those who will be individually
impacted by a given procedure. Still, that plain and indisputable standard is not easily
applied to gene editing and gene drive research, which are designed to modify entities yet
to be born or even environments and ecosystems. The cornerstone of ethics-based research
and innovation in those realms must therefore be based on fundamental values applicable
to categories of research in the broad sense, in keeping with the fundamental precepts
enshrined in human rights treaties and conventions. The very essence of human rights
undoubtedly comprises both the duty of nations to uphold the people’s rights, in terms

Cells 2021, 10, 969 16 of 24

of physical well-being and autonomy, and to enable them to rely on a wholesome living
environment. It is after all the very notion of human dignity on which the United Nations
Universal Declaration of Human Rights is based [196]. Guidelines and recommendations
by national and international scientific institutions must be drawn upon as valuable frame
of reference, for the purpose of updating the moral and ethical compass through which
genome editing is regulated. To that end, the already mentioned recent analysis by the NAS
has outlined valuable guidance for the gradual and supervised development of the most
controversial form of gene editing: heritable human genome editing [129]. Specifically,
dramatic and highly consequential breakthroughs need to be regulated by prioritizing
safety. That of course calls for an extremely cautious selection of relatively few initial cases,
based on a thoroughly implemented risk–benefit analysis, and initial outcomes must be
reviewed and evaluated before further procedures are given the nod. Moreover, when a
high degree of uncertainty exists as to the procedure’s efficacy, interventions need to be
focused on patients suffering from clinical conditions with high mortality and/or morbidity
and for which no alternatives are available. Abiding by such standards would be in keeping
with the most accurate assessment of potential harms versus benefits, and could ensure
that human applications of such techniques are offered after an ethically tenable informed
consent process. The NAS also calls attention to two ethical issues that further buttress those
three standards: as mentioned earlier, heritable human genome editing affects the future
existence of individuals who would be created as a result of the technology, but obviously
cannot provide consent, which can be granted by their parents only. Secondly, germline
editing gives rise to genetic alterations which can be inherited by future generations. Those
points make the need for shared solutions and criteria all the more urgent, however hard
it may be to achieve it. That perspective had already been highlighted by three reports
issued by the World Health Organization Expert Advisory Committee on Developing
Global Standards for Governance and Oversight of Human Genome Editing, a global
multidisciplinary expert panel that set out to delve into the scientific, ethical, social and
legal challenges linked to human genome editing, whether somatic or germ cell [197]. The
WHO Committee, which held its first meeting from 18–19 March 2019, pointed out that
relatively few countries have as yet outlined and codified an appropriate translational
pathway for somatic human genome editing interventions, relying on well-balanced and
enforceable regulation and oversight mechanisms aimed at patient safety and public
confidence. Hence, looking forward, the priority set by the WHO’s expert working group
was to gain an understanding and awareness of how to best foster transparency and
reliable practices, and to make sure that any decision and authorization will be based on
appropriate risk/benefit assessments, in apparent agreement with the NAS considerations.

4. Conclusions: When Technology Outpaces Regulatory Governance, Harmonization
May Be Key

Like all potentially revolutionary technologies that seem poised to call into question
core values that are deeply held in our societies, genome editing is bound to entail daunting
quandaries. Ethical complexities and apparently irreconcilable elements need to be dealt
with through balanced legal and regulatory approaches aimed at upholding the rights of
all parties involved and no less importantly, fostering public health in the face of extraordi-
narily challenging circumstances. That prospect has to rely on a broad-ranging risk–benefit
analysis. This is currently difficult to achieve, considering that several international laws
severely restrict or ban such research or prevent it from being adequately funded. Reliable
data concerning benefits and risks are largely unavailable. It is of utmost importance that
governments reconsider their reasoning for putting in place such restrictions, so as to
ensure that they are really warranted and not merely rooted in fear. At the same time,
CRISPR applications to non-human organisms cannot be overlooked in their potential
to pose biothreats. The extraordinary significance of germline alterations for individuals
and societies has not yet been publicly debated. Yet, a multidisciplinary discourse could
reliably and efficiently enable policymakers, funders, research institutions, and users to
draw distinctions between appropriate applications of a technology from those and those

Cells 2021, 10, 969 17 of 24

that are inappropriate, intolerable, or even dangerous. Going forward, many support es-
tablishing an organization that will decide how best to address the aforementioned ethical
complexities. Initiatives such as the International Summit on Human Genome Editing are
a step toward the ultimate goal of finding middle ground and shared solutions at least
among nations founded on a common framework of core values.

Author Contributions: Conceptualization, R.P., S.Z. and A.D.R.; supervision, F.S., F.U.R. and E.M.;
writing—original draft preparation R.P., A.D.R., E.M. and S.Z.; writing—review and editing, F.U.R.,
R.P. and S.Z. All authors have read and agreed to the published version of the manuscript.

Funding: This research received no external funding.

Institutional Review Board Statement: Not applicable.

Informed Consent Statement: Not applicable.

Data Availability Statement: The data presented in this study are available on request from the
corresponding author.

Conflicts of Interest: The authors declare no conflict of interest.

References
1. Ishino, Y.; Shinagawa, H.; Makino, K.; Amemura, M.; Nakatura, A. Nucleotide sequence of the iap gene, responsible for alkaline

phosphatase isoenzyme conversion in Escherichia coli, and identification of the gene product. J. Bacteriol. 1987, 169, 5429–5433.
[CrossRef]

2. Jinek, M.; Chylinski, K.; Fonfara, I.; Hauer, M.; Doudna, J.A.; Charpentier, E. A programmable dual-RNA-guided DNA
endonuclease in adaptive bacterial immunity. Science 2012, 337, 816–821. [CrossRef]

3. Press Release: The Nobel Prize in Chemistry 2020. Available online: https://www.nobelprize.org/prizes/chemistry/2020/press-
release/ (accessed on 16 March 2021).

4. Hynes, A.P.; Villion, M.; Moineau, S. Adaptation in bacterial CRISPR-Cas immunity can be driven by defective phages. Nat.
Commun. 2014, 5, 1–6.

5. Barrangou, R.; Marraffini, L.A. CRISPR-cas systems: Prokaryotes upgrade to adaptive immunity. Mol. Cell 2014, 54, 234–244.
[CrossRef] [PubMed]

6. Westra, E.R.; Dowling, A.J.; Broniewski, J.M.; Van Houte, S. Evolution and Ecology of CRISPR. Annu. Rev. Ecol. Evol. Syst. 2016,
47, 307–331. [CrossRef]

7. Makarova, K.S.; Wolf, Y.I.; Iranzo, J.; Shmakov, S.A.; Alkhnbashi, O.S.; Brouns, S.J.J.; Charpentier, E.; Cheng, D.; Haft, D.H.;
Horvath, P.; et al. Evolutionary classification of CRISPR–Cas systems: A burst of class 2 and derived variants. Nat. Rev. Microbiol.
2020, 18, 67–83. [CrossRef] [PubMed]

8. Cong, L.; Ran, F.A.; Cox, D.; Lin, S.; Barretto, R.; Habib, N.; Hsu, P.D.; Wu, X.; Jiang, W.; Marraffini, L.A.; et al. Multiplex Genome
Engineering Using CRISPR/Cas Systems. Science 2013, 339, 819–823. [CrossRef] [PubMed]

9. Burgess, D.J. Technology: A CRISPR Genome-Editing Tool. Nat. Rev. Genet. 2013, 14, 80. [CrossRef]
10. Li, Q.; Sapkota, M.; van der Knaap, E. Perspectives of CRISPR/Cas-mediated cis-engineering in horticulture: Unlocking the

neglected potential for crop improvement. Hortic. Res. 2020, 7, 36.
11. Huang, X.; Hilscher, J.; Stoger, E.; Christou, P.; Zhu, C. Modification of cereal plant architecture by genome editing to improve

yields. Plant Cell Rep. 2021, 1–26. [CrossRef]
12. Ku, H.-K.; Ha, S.-H. Improving Nutritional and Functional Quality by Genome Editing of Crops: Status and Perspectives. Front.

Plant Sci. 2020, 11, 23. [CrossRef] [PubMed]
13. Arora, L.; Narula, A. Gene editing and crop improvement using CRISPR-cas9 system. Front. Plant Sci. 2017, 8, 1932. [CrossRef]

[PubMed]
14. Martín-Pizarro, C.; Posé, D. Genome editing as a tool for fruit ripening manipulation. Front. Plant Sci. 2018, 9, 1415. [CrossRef]
15. Shipman, E.N.; Yu, J.; Zhou, J.; Albornoz, K.; Beckles, D.M. Can gene editing reduce postharvest waste and loss of fruit, vegetables,

and ornamentals? Hortic. Res. 2021, 8, 1. [CrossRef]
16. Schenke, D.; Cai, D. Applications of CRISPR/Cas to Improve Crop Disease Resistance: Beyond Inactivation of Susceptibility

Factors. iScience 2020, 23, 101478. [CrossRef]
17. Ahmad, S.; Wei, X.; Sheng, Z.; Hu, P.; Tang, S. CRISPR/Cas9 for development of disease resistance in plants: Recent progress,

limitations and future prospects. Brief. Funct. Genom. 2020, 19, 26–39. [CrossRef]
18. Borrelli, V.M.G.; Brambilla, V.; Rogowsky, P.; Marocco, A.; Lanubile, A. The enhancement of plant disease resistance using

crispr/cas9 technology. Front. Plant Sci. 2018, 9, 1245. [CrossRef]
19. Jaganathan, D.; Ramasamy, K.; Sellamuthu, G.; Jayabalan, S.; Venkataraman, G. CRISPR for crop improvement: An update review.

Front. Plant Sci. 2018, 9, 985. [CrossRef]

Cells 2021, 10, 969 18 of 24

20. Martignago, D.; Rico-Medina, A.; Blasco-Escámez, D.; Fontanet-Manzaneque, J.B.; Caño-Delgado, A.I. Drought Resistance by
Engineering Plant Tissue-Specific Responses. Front. Plant Sci. 2020, 10, 1676. [CrossRef] [PubMed]

21. Zafar, S.A.; Zaidi, S.S.E.A.; Gaba, Y.; Singla-Pareek, S.L.; Dhankher, O.P.; Li, X.; Mansoor, S.; Pareek, A. Engineering abiotic stress
tolerance via CRISPR/Cas-mediated genome editing. J. Exp. Bot. 2020, 71, 470–479. [CrossRef]

22. Wang, T.; Zhang, H.; Zhu, H. CRISPR technology is revolutionizing the improvement of tomato and other fruit crops. Hortic. Res.
2019, 6, 77. [CrossRef]

23. Corte, L.E.D.; Mahmoud, L.M.; Moraes, T.S.; Mou, Z.; Grosser, J.W.; Dutt, M. Development of improved fruit, vegetable, and
ornamental crops using the CRISPR/cas9 genome editing technique. Plants 2019, 8, 601. [CrossRef]

24. Gao, Q.; Luo, H.; Li, Y.; Liu, Z.; Kang, C. Genetic modulation of RAP alters fruit coloration in both wild and cultivated strawberry.
Plant Biotechnol. J. 2020, 18, 1550–1561. [CrossRef]

25. Naves, E.R.; de Ávila Silva, L.; Sulpice, R.; Araújo, W.L.; Nunes-Nesi, A.; Peres, L.E.P.; Zsögön, A. Capsaicinoids: Pungency
beyond Capsicum. Trends Plant Sci. 2019, 24, 109–120. [CrossRef]

26. Sugano, S.; Hirose, A.; Kanazashi, Y.; Adachi, K.; Hibara, M.; Itoh, T.; Mikami, M.; Endo, M.; Hirose, S.; Maruyama, N.; et al.
Simultaneous induction of mutant alleles of two allergenic genes in soybean by using site-directed mutagenesis. BMC Plant Biol.
2020, 20. [CrossRef] [PubMed]

27. Jouanin, A.; Gilissen, L.J.W.J.; Schaart, J.G.; Leigh, F.J.; Cockram, J.; Wallington, E.J.; Boyd, L.A.; van den Broeck, H.C.; van der Meer,
I.M.; America, A.H.P.; et al. CRISPR/Cas9 Gene Editing of Gluten in Wheat to Reduce Gluten Content and Exposure—Reviewing
Methods to Screen for Coeliac Safety. Front. Nutr. 2020, 7, 51. [CrossRef] [PubMed]

28. Sánchez-León, S.; Gil-Humanes, J.; Ozuna, C.V.; Giménez, M.J.; Sousa, C.; Voytas, D.F.; Barro, F. Low-gluten, nontransgenic wheat
engineered with CRISPR/Cas9. Plant Biotechnol. J. 2018, 16, 902–910. [CrossRef]

29. Cowan, P.J.; Hawthorne, W.J.; Nottle, M.B. Xenogeneic transplantation and tolerance in the era of CRISPR-Cas9. Curr. Opin.
Organ Transplant. 2019, 24, 5–11. [CrossRef]

30. Kararoudi, M.N.; Hejazi, S.S.; Elmas, E.; Hellström, M.; Kararoudi, M.N.; Padma, A.M.; Lee, D.; Dolatshad, H. Clustered regularly
interspaced short palindromic repeats/Cas9 gene editing technique in xenotransplantation. Front. Immunol. 2018, 9, 1711.
[CrossRef]

31. Menchaca, A.; dos Santos-Neto, P.C.; Mulet, A.P.; Crispo, M. CRISPR in livestock: From editing to printing. Theriogenology 2020,
150, 247–254. [CrossRef]

32. Khwatenge, C.N.; Nahashon, S.N. Recent Advances in the Application of CRISPR/Cas9 Gene Editing System in Poultry Species.
Front. Genet. 2021, 12, 627714. [CrossRef]

33. Cyranoski, D. Gene-edited pigs to be sold as pets. Nature 2015, 526, 18. [CrossRef]
34. Zhou, W.; Wan, Y.; Guo, R.; Deng, M.; Deng, K.; Wang, Z.; Zhang, Y.; Wang, F. Generation of beta-lactoglobulin knock-out goats

using CRISPR/Cas9. PLoS ONE 2017, 12, e0186056. [CrossRef]
35. Mukae, T.; Yoshii, K.; Watanobe, T.; Tagami, T.; Oishi, I. Production and characterization of eggs from hens with ovomucoid gene

mutation. Poult. Sci. 2021, 100, 452–460. [CrossRef]
36. Torres-Ruiz, R.; Rodriguez-Perales, S. CRISPR-Cas9 technology: Applications and human disease modelling. Brief. Funct. Genom.

2017, 16, 4–12. [CrossRef]
37. Alagoz, M.; Kherad, N. Advance genome editing technologies in the treatment of human diseases: CRISPR therapy (Review). Int.

J. Mol. Med. 2020, 46, 521–534. [CrossRef]
38. Van Giau, V.; Lee, H.; Shim, K.H.; Bagyinszky, E.; An, S.S.A. Genome-editing applications of CRISPR–Cas9 to promote in vitro

studies of Alzheimer’s disease. Clin. Interv. Aging 2018, 13, 221–233. [CrossRef] [PubMed]
39. Safari, F.; Hatam, G.; Behbahani, A.B.; Rezaei, V.; Barekati-Mowahed, M.; Petramfar, P.; Khademi, F. CRISPR System: A

High-throughput Toolbox for Research and Treatment of Parkinson’s Disease. Cell. Mol. Neurobiol. 2020, 40, 477–493. [CrossRef]
40. Vermersch, E.; Jouve, C.; Hulot, J.S. CRISPR/Cas9 gene-editing strategies in cardiovascular cells. Cardiovasc. Res. 2020, 116,

894–907. [CrossRef] [PubMed]
41. Sharma, G.; Sharma, A.R.; Bhattacharya, M.; Lee, S.S.; Chakraborty, C. CRISPR-Cas9: A Preclinical and Clinical Perspective for

the Treatment of Human Diseases. Mol. Ther. 2021, 29, 571–586. [CrossRef] [PubMed]
42. Karimian, A.; Gorjizadeh, N.; Alemi, F.; Asemi, Z.; Azizian, K.; Soleimanpour, J.; Malakouti, F.; Targhazeh, N.; Majidinia, M.;

Yousefi, B. CRISPR/Cas9 novel therapeutic road for the treatment of neurodegenerative diseases. Life Sci. 2020, 259. [CrossRef]
43. Chen, M.; Mao, A.; Xu, M.; Weng, Q.; Mao, J.; Ji, J. CRISPR-Cas9 for cancer therapy: Opportunities and challenges. Cancer Lett.

2019, 447, 48–55. [CrossRef]
44. Xing, H.; Meng, L. CRISPR-cas9: A powerful tool towards precision medicine in cancer treatment. Acta Pharmacol. Sin. 2020, 41,

583–587. [CrossRef]
45. Jiang, C.; Meng, L.; Yang, B.; Luo, X. Application of CRISPR/Cas9 gene editing technique in the study of cancer treatment. Clin.

Genet. 2020, 97, 73–88. [CrossRef] [PubMed]
46. Mirza, Z.; Karim, S. Advancements in CRISPR/Cas9 technology—Focusing on cancer therapeutics and beyond. Semin. Cell Dev.

Biol. 2019, 96, 13–21. [CrossRef]
47. Saber, A.; Liu, B.; Ebrahimi, P.; Haisma, H.J. CRISPR/Cas9 for overcoming drug resistance in solid tumors. DARU J. Pharm. Sci.

2020, 28, 295–304. [CrossRef] [PubMed]

Cells 2021, 10, 969 19 of 24

48. Liu, B.; Saber, A.; Haisma, H.J. CRISPR/Cas9: A powerful tool for identification of new targets for cancer treatment. Drug Discov.
Today 2019, 24, 955–970. [CrossRef]

49. Vallone, C.; Rigon, G.; Gulia, C.; Baffa, A.; Votino, R.; Morosetti, G.; Zaami, S.; Briganti, V.; Catania, F.; Gaffi, M.; et al. Non-Coding
RNAs and Endometrial Cancer. Genes 2018, 9, 187. [CrossRef]

50. Esposito, R.; Bosch, N.; Lanzós, A.; Polidori, T.; Pulido-Quetglas, C.; Johnson, R. Hacking the Cancer Genome: Profiling
Therapeutically Actionable Long Non-coding RNAs Using CRISPR-Cas9 Screening. Cancer Cell 2019, 35, 545–557. [CrossRef]
[PubMed]

51. Piergentili, R.; Zaami, S.; Cavaliere, A.F.; Signore, F.; Scambia, G.; Mattei, A.; Marinelli, E.; Gulia, C.; Perelli, F. Non-Coding RNAs
as Prognostic Markers for Endometrial Cancer. Int. J. Mol. Sci. 2021, 22, 3151. [CrossRef]

52. Pavani, G.; Fabiano, A.; Laurent, M.; Amor, F.; Cantelli, E.; Chalumeau, A.; Maule, G.; Tachtsidi, A.; Concordet, J.-P.;
Cereseto, A.; et al. Correction of β-thalassemia by CRISPR/Cas9 editing of the α-globin locus in human hematopoietic stem cells.
Blood Adv. 2021, 5, 1137–1153. [CrossRef]

53. Xiao, Q.; Guo, D.; Chen, S. Application of CRISPR/Cas9-based gene editing in HIV-1/AIDS therapy. Front. Cell. Infect. Microbiol.
2019, 9, 69. [CrossRef]

54. Xu, T.; Li, L.; Liu, Y.C.; Cao, W.; Chen, J.S.; Hu, S.; Liu, Y.; Li, L.Y.; Zhou, H.; Meng, X.M.; et al. Crispr/cas9-related technologies in
liver diseases: From feasibility to future diversity. Int. J. Biol. Sci. 2020, 16, 2283–2295. [CrossRef]

55. Zuo, E.; Huo, X.; Yao, X.; Hu, X.; Sun, Y.; Yin, J.; He, B.; Wang, X.; Shi, L.; Ping, J.; et al. CRISPR/Cas9-mediated targeted
chromosome elimination. Genome Biol. 2017, 18, 224. [CrossRef]

56. North, A.R.; Burt, A.; Godfray, H.C.J. Modelling the Suppression of a Malaria Vector Using a CRISPR-Cas9 Gene Drive to Reduce
Female Fertility. BMC Biol. 2020, 18, 98. [CrossRef]

57. Annas, G.J.; Beisel, C.L.; Clement, K.; Crisanti, A.; Francis, S.; Galardini, M.; Galizi, R.; Grünewald, J.; Immobile, G.;
Khalil, A.S.; et al. A Code of Ethics for Gene Drive Research. Cris. J. 2021, 4, 19–24. [CrossRef]

58. Callies, D.E. The ethical landscape of gene drive research. Bioethics 2019, 33, 1091–1097. [CrossRef] [PubMed]
59. Kang, X.; He, W.; Huang, Y.; Yu, Q.; Chen, Y.; Gao, X.; Sun, X.; Fan, Y. Introducing precise genetic modifications into human 3PN

embryos by CRISPR/Cas-mediated genome editing. J. Assist. Reprod. Genet. 2016, 33, 581–588. [CrossRef]
60. Yang, Y.; Huang, Y. The CRIPSR /Cas gene-editing system—An immature but useful toolkit for experimental and clinical

medicine. Anim. Model. Exp. Med. 2019, 2, 5–8. [CrossRef]
61. Ma, Y.; Zhang, L.; Qin, C. The first genetically gene-edited babies: It’s “irresponsible and too early”. Anim. Model. Exp. Med. 2019,

2, 1–4. [CrossRef]
62. Razzouk, S. CRISPR-Cas9: A cornerstone for the evolution of precision medicine. Ann. Hum. Genet. 2018, 82, 331–357. [CrossRef]

[PubMed]
63. Ma, H.; Marti-Gutierrez, N.; Park, S.W.; Wu, J.; Lee, Y.; Suzuki, K.; Koski, A.; Ji, D.; Hayama, T.; Ahmed, R.; et al. Correction of a

pathogenic gene mutation in human embryos. Nature 2017, 548, 413–419. [CrossRef] [PubMed]
64. Zaami, S.; Piergentili, R.; Marinelli, E.; Montanari Vergallo, G. Commentary—CRISPR-based techniques: Cas9, Cas13 and their

applications in the era of COVID-19. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 1752–1761.
65. Wang, H.; Yang, H. Gene-edited babies: What went wrong and what could go wrong. PLoS Biol. 2019, 17, e3000224. [CrossRef]
66. Krishan, K.; Kanchan, T.; Singh, B. Human Genome Editing and Ethical Considerations. Sci. Eng. Ethics 2016, 22, 597–599.

[CrossRef] [PubMed]
67. Korablev, A.; Lukyanchikova, V.; Serova, I.; Battulin, N. On-target CRISPR/CAS9 activity can cause undesigned large deletion in

mouse zygotes. Int. J. Mol. Sci. 2020, 21, 3604. [CrossRef]
68. Kosicki, M.; Tomberg, K.; Bradley, A. Repair of double-strand breaks induced by CRISPR–Cas9 leads to large deletions and

complex rearrangements. Nat. Biotechnol. 2018, 36, 765. [CrossRef]
69. Przewrocka, J.; Rowan, A.; Rosenthal, R.; Kanu, N.; Swanton, C. Unintended on-target chromosomal instability following

CRISPR/Cas9 single gene targeting. Ann. Oncol. 2020, 31, 1270–1273. [CrossRef]
70. Cullot, G.; Boutin, J.; Toutain, J.; Prat, F.; Pennamen, P.; Rooryck, C.; Teichmann, M.; Rousseau, E.; Lamrissi-Garcia, I.; Guyonnet-

Duperat, V.; et al. CRISPR-Cas9 genome editing induces megabase-scale chromosomal truncations. Nat. Commun. 2019, 10, 1–14.
[CrossRef]

71. Alanis-Lobato, G.; Zohren, J.; McCarthy, A.; Fogarty, N.M.E.; Kubikova, N.; Hardman, E.; Greco, M.; Wells, D.; Turner, J.M.A.;
Niakan, K.K. Frequent loss-of-heterozygosity in CRISPR-Cas9-edited early human embryos. Proc. Natl. Acad. Sci. USA 2021.
[CrossRef]

72. Zuccaro, M.V.; Xu, J.; Mitchell, C.; Marin, D.; Zimmerman, R.; Rana, B.; Weinstein, E.; King, R.T.; Palmerola, K.L.; Smith, M.E.; et al.
Allele-Specific Chromosome Removal after Cas9 Cleavage in Human Embryos. Cell 2020, 183, 1650–1664. [CrossRef]

73. Abd El-Aziz, M.M.; Barragan, I.; O’Driscoll, C.A.; Goodstadt, L.; Prigmore, E.; Borrego, S.; Mena, M.; Pieras, J.I.; El-Ashry, M.F.;
Safieh, L.A.; et al. EYS, encoding an ortholog of Drosophila spacemaker, is mutated in autosomal recessive retinitis pigmentosa.
Nat. Genet. 2008, 40, 1285–1287. [CrossRef] [PubMed]

74. Sfeir, A.; Symington, L.S. Microhomology-Mediated End Joining: A Back-up Survival Mechanism or Dedicated Pathway? Trends
Biochem. Sci. 2015, 40, 701–714. [CrossRef]

75. Liang, D.; Gutierrez, N.M.; Chen, T.; Lee, Y.; Park, S.W.; Ma, H.; Koski, A.; Ahmed, R.; Darby, H.; Li, Y.; et al. Frequent gene
conversion in human embryos induced by double strand breaks. bioRxiv 2020. [CrossRef]

Cells 2021, 10, 969 20 of 24

76. Anzalone, A.V.; Randolph, P.B.; Davis, J.R.; Sousa, A.A.; Koblan, L.W.; Levy, J.M.; Chen, P.J.; Wilson, C.; Newby, G.A.;
Raguram, A.; et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 2019, 576, 149–157.
[CrossRef]

77. Tu, Z.; Yang, W.; Yan, S.; Yin, A.; Gao, J.; Liu, X.; Zheng, Y.; Zheng, J.; Li, Z.; Yang, S.; et al. Promoting Cas9 degradation reduces
mosaic mutations in non-human primate embryos. Sci. Rep. 2017, 7, 42081. [CrossRef]

78. Peng, R.; Lin, G.; Li, J. Potential pitfalls of CRISPR/Cas9-mediated genome editing. FEBS J. 2016, 283, 1218–1231. [CrossRef]
[PubMed]

79. Brokowski, C.; Pollack, M.; Pollack, R. Cutting Eugenics Out of CRISPR-Cas9. Ethics Biol. Eng. Med. 2015, 6, 263–279. [CrossRef]
80. Brokowski, C.; Adli, M. CRISPR Ethics: Moral Considerations for Applications of a Powerful Tool. J. Mol. Biol. 2019, 431, 88–101.

[CrossRef]
81. Wu, S.; Powers, S.; Zhu, W.; Hannun, Y.A. Substantial contribution of extrinsic risk factors to cancer development. Nature 2016,

529, 43–47. [CrossRef]
82. Napoletano, S.; Piersanti, V.; Rallo, G. CRISPR -Cas9: A groundbreaking new technique which ushers in new prospects and just

as many doubts. Clin. Ter. 2021, 171, e52–e54. [CrossRef]
83. Marinelli, S.; Del Rio, A. Beginning of Life Ethics at the Dawn of a New Era of Genome Editing: Are Bioethical Precepts and

Fast-Evolving Biotechnologies Irreconcilable? Clin. Ter. 2020, 171, e407–e411.
84. Gilbert, S.F. When “Personhood” Begins in the Embryo: Avoiding a Syllabus of Errors. Birth Defects Res. C Embryo Today 2008, 84,

164–173. [CrossRef]
85. Charo, R.A. The hunting of the snark: The moral status of embryos, right-to-lifers, and Third World women. Stanf. Law Pol. Rev.

1995, 6, 11–37. [PubMed]
86. Ledford, H. CRISPR gene editing in human embryos wreaks chromosomal mayhem. Nature 2020, 583, 17–18. [CrossRef]
87. Ricci, G.; Campanozzi, L.L.; Marinelli, S.; Midolo, E.; Ruggeri, L. The Human Embryo, Subjectivity and Legal Capacity. Notes in

the Light of Art. 1 of the Italian Law on “Medically Assisted Procreation”. Clin. Ter. 2019, 170, e102–e107. [CrossRef]
88. Botkin, J.R. The Case for Banning Heritable Genome Editing. Genet. Med. 2020, 22, 487–489. [CrossRef] [PubMed]
89. Musunuru, K. Why Human Embryo Editing Should Be Banned. CRISPR J. 2019, 2, 356–358. [CrossRef] [PubMed]
90. Grant, E.V. FDA Regulation of Clinical Applications of CRISPR-CAS Gene-Editing Technology. Food Drug L. J. 2016, 71, 608–633.
91. Public Law 114–113—18 December 2015. Consolidated Appropriations Act. 2016. Available online: https://www.congress.gov/

114/plaws/publ113/PLAW-114publ113.pdf (accessed on 15 February 2021).
92. Williams, K.; Johnson, M.H. Adapting the 14-Day Rule for Embryo Research to Encompass Evolving Technologies. Reprod. Biomed.

Soc. Online 2020, 10, 1–9. [CrossRef]
93. Law 40, 24 February 2004, Regulation of Medically Assisted Human Reproduction, Legge 24 Febbraio 2004, n. 40, Norme in

Materia di Procreazione Medicalmente Assistita, G. U. N. 45 24-2-2004. Available online: https://www.camera.it/parlam/leggi/
04040l.htm (accessed on 15 February 2021).

94. Montanari, V.G.; Zaami, S.; Bruti, V.; Signore, F. How the legislation in medically assisted procreation has evolved in Italy. Med.
Law 2017, 36, 5.

95. Negro, F.; Marinelli, S. Is There Anything Left of the Italian Law Governing Medically-Assisted Procreation? Clin. Ter. 2021, 171,
e57–e59. [CrossRef]

96. Castelyn, G. Embryo Experimentation: Is There a Case for Moving beyond the “14-Day Rule”. Monash Bioeth. Rev. 2020, 38,
181–196. [CrossRef]

97. Dobler, R.; Dowling, D.K.; Morrow, E.H.; Reinhardt, K. A Systematic Review and Meta-Analysis Reveals Pervasive Effects of
Germline Mitochondrial Replacement on Components of Health. Hum. Reprod. Update 2018, 24, 519–534. [CrossRef]

98. Cyranoski, D. The CRISPR-Baby Scandal: What’s next for Human Gene-Editing. Nature 2019, 566, 440–442. [CrossRef]
99. Miklavcic, J.J.; Flaman, P. Personhood Status of the Human Zygote, Embryo, Fetus. Linacre Q. 2017, 84, 130–144. [CrossRef]
100. Paul, P.J., II. You Shall Not Kill in Idem, The Gospel of Life (Evangelium Vitae); Times Books; Random House: New York, NY, USA,

1995; pp. 92–140.
101. Nimbalkar, N. John Locke on Personal Identity. Mens. Sana Monogr. 2011, 9, 268–275. [CrossRef]
102. Manninen, B.A. Are Human Embryos Kantian Persons? Kantian Considerations in Favor of Embryonic Stem Cell Research.

Philos. Ethics Humanit. Med. 2008, 3, 4. [CrossRef]
103. Donovan, P. When Does Personhood Begin? Fam. Plan. Perspect. 1983, 15, 40–44. [CrossRef]
104. Wagner, N.-F.; Northoff, G. Habits: Bridging the Gap between Personhood and Personal Identity. Front. Hum. Neurosci. 2014, 8,

330. [CrossRef]
105. Fan, H.-C.; Chi, C.-S.; Lee, Y.-J.; Tsai, J.-D.; Lin, S.-Z.; Harn, H.-J. The Role of Gene Editing in Neurodegenerative Diseases. Cell

Transplant. 2018, 27, 364–378. [CrossRef]
106. Gallego, C.; Gonçalves, M.A.F.V.; Wijnholds, J. Novel Therapeutic Approaches for the Treatment of Retinal Degenerative Diseases:

Focus on CRISPR/Cas-Based Gene Editing. Front. Neurosci. 2020, 14, 838. [CrossRef]
107. Gaj, T.; Perez-Pinera, P. The Continuously Evolving CRISPR Barcoding Toolbox. Genome Biol. 2018, 19, 143. [CrossRef]
108. Wang, H.; La Russa, M.; Qi, L.S. CRISPR/Cas9 in Genome Editing and Beyond. Annu. Rev. Biochem. 2016, 85, 227–264. [CrossRef]
109. Zhang, J.-H.; Adikaram, P.; Pandey, M.; Genis, A.; Simonds, W.F. Optimization of Genome Editing through CRISPR-Cas9

Engineering. Bioengineered 2016, 7, 166–174. [CrossRef]

Cells 2021, 10, 969 21 of 24

110. Ben Jehuda, R.; Shemer, Y.; Binah, O. Genome Editing in Induced Pluripotent Stem Cells Using CRISPR/Cas9. Stem Cell Rev. Rep.
2018, 14, 323–336. [CrossRef]

111. Geng, B.; Choi, K.-H.; Wang, S.; Chen, P.; Pan, X.; Dong, N.; Ko, J.-K.; Zhu, H. A Simple, Quick, and Efficient CRISPR/Cas9
Genome Editing Method for Human Induced Pluripotent Stem Cells. Acta Pharmacol. Sin. 2020, 41, 1427–1432. [CrossRef]
[PubMed]

112. Lu, F.; Zhang, Y. Cell Totipotency: Molecular Features, Induction, and Maintenance. Natl. Sci. Rev. 2015, 2, 217–225. [CrossRef]
113. Le, R.; Huang, Y.; Zhao, A.; Gao, S. Lessons from Expanded Potential of Embryonic Stem Cells: Moving toward Totipotency. J.

Genet. Genom. 2020, 47, 123–130. [CrossRef]
114. De Masi, C.; Spitalieri, P.; Murdocca, M.; Novelli, G.; Sangiuolo, F. Application of CRISPR/Cas9 to Human-Induced Pluripotent

Stem Cells: From Gene Editing to Drug Discovery. Hum. Genom. 2020, 14, 25. [CrossRef]
115. Spitalieri, P.; Quitadamo, M.C.; Orlandi, A.; Guerra, L.; Giardina, E.; Casavola, V.; Novelli, G.; Saltini, C.; Sangiuolo, F. Rescue of

Murine Silica-Induced Lung Injury and Fibrosis by Human Embryonic Stem Cells. Eur. Respir. J. 2012, 39, 446–457. [CrossRef]
[PubMed]

116. Nie, J.; Li, Q.; Wu, J.; Zhao, C.; Hao, H.; Liu, H.; Zhang, L.; Nie, L.; Qin, H.; Wang, M.; et al. Establishment and Validation of a
Pseudovirus Neutralization Assay for SARS-CoV-2. Emerg. Microbes Infect. 2020, 9, 680–686. [CrossRef] [PubMed]

117. Uddin, F.; Rudin, C.M.; Sen, T. CRISPR Gene Therapy: Applications, Limitations, and Implications for the Future. Front. Oncol.
2020, 10, 1387. [CrossRef] [PubMed]

118. Nguyen, T.M.; Zhang, Y.; Pandolfi, P.P. Virus against Virus: A Potential Treatment for 2019-NCov (SARS-CoV-2) and Other RNA
Viruses. Cell. Res. 2020, 30, 189–190. [CrossRef] [PubMed]

119. Straiton, J. CRISPR vs. COVID-19: How Can Gene Editing Help Beat a Virus? BioTechniques 2020, 69, 327–329. [CrossRef]
[PubMed]

120. Abbott, T.R.; Dhamdhere, G.; Liu, Y.; Lin, X.; Goudy, L.; Zeng, L.; Chemparathy, A.; Chmura, S.; Heaton, N.S.; Debs, R.; et al.
Development of CRISPR as an Antiviral Strategy to Combat SARS-CoV-2 and Influenza. Cell 2020, 181, 865–876. [CrossRef]
[PubMed]

121. Marinelli, E.; Busardò, F.P.; Zaami, S. Intensive and Pharmacological Care in Times of COVID-19: A “Special Ethics” for
Emergency? BMC Med. Ethics 2020, 21, 117. [CrossRef]

122. Savulescu, J.; Pugh, J.; Douglas, T.; Gyngell, C. The Moral Imperative to Continue Gene Editing Research on Human Embryos.
Protein Cell 2015, 6, 476–479. [CrossRef]

123. Cavaliere, G. Genome Editing and Assisted Reproduction: Curing Embryos, Society or Prospective Parents? Med. Health Care
Philos 2018, 21, 215–225. [CrossRef]

124. Schleidgen, S.; Dederer, H.-G.; Sgodda, S.; Cravcisin, S.; Lüneburg, L.; Cantz, T.; Heinemann, T. Human Germline Editing in the
Era of CRISPR-Cas: Risk and Uncertainty, Inter-Generational Responsibility, Therapeutic Legitimacy. BMC Med. Ethics 2020, 21,
87. [CrossRef]

125. Gyngell, C.; Bowman-Smart, H.; Savulescu, J. Moral Reasons to Edit the Human Genome: Picking up from the Nuffield Report. J.
Med. Ethics 2019, 45, 514–523. [CrossRef] [PubMed]

126. Ledford, H. CRISPR Babies: When Will the World Be Ready? Nature 2019, 570, 293–296. [CrossRef] [PubMed]
127. Ranisch, R. Germline Genome Editing versus Preimplantation Genetic Diagnosis: Is There a Case in Favour of Germline

Interventions? Bioethics 2020, 34, 60–69. [CrossRef] [PubMed]
128. Zaami, S.; Orrico, A.; Signore, F.; Cavaliere, A.F.; Mazzi, M.; Marinelli, E. Ethical, Legal and Social Issues (ELSI) Associated with

Non-Invasive Prenatal Testing: Reflections on the Evolution of Prenatal Diagnosis and Procreative Choices. Genes 2021, 12, 204.
[CrossRef] [PubMed]

129. International Commission on the Clinical Use of Human Germline Genome Editing; National Academy of Medicine; National
Academy of Sciences; The Royal Society. Heritable Human Genome Editing; National Academies Press: Washington, DC, USA, 2020;
ISBN 9780309671132.

130. Bourne, H.; Douglas, T.; Savulescu, J. Procreative Beneficence and in Vitro Gametogenesis. Monash Bioeth. Rev. 2012, 30, 29–48.
[CrossRef]

131. Zaami, S.; Tagliabracci, A.; Berretta, P.; Busardò, F.P.; Marinelli, E. Use of Methylphenidate Analogues as Cognitive Enhancers:
The Prelude to Cosmetic Neurology and an Ethical Issue. Front. Psychiatry 2019, 10, 1006. [CrossRef]

132. Wise, J. Cognitive Enhancers Are Set to Change Workplaces. BMJ 2012, 345, e7538. [CrossRef]
133. Carlier, J.; Giorgetti, R.; Varì, M.R.; Pirani, F.; Ricci, G.; Busardò, F.P. Use of Cognitive Enhancers: Methylphenidate and Analogs.

Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 3–15. [CrossRef]
134. Urban, K.R.; Gao, W.-J. Performance Enhancement at the Cost of Potential Brain Plasticity: Neural Ramifications of Nootropic

Drugs in the Healthy Developing Brain. Front. Syst. Neurosci. 2014, 8, 38. [CrossRef]
135. Ragan, C.I.; Bard, I.; Singh, I. Independent Scientific Committee on Drugs What Should We Do about Student Use of Cognitive

Enhancers? An Analysis of Current Evidence. Neuropharmacology 2013, 64, 588–595. [CrossRef]
136. d’Angelo, L.-S.C.; Savulich, G.; Sahakian, B.J. Lifestyle Use of Drugs by Healthy People for Enhancing Cognition, Creativity,

Motivation and Pleasure. Br. J. Pharmacol. 2017, 174, 3257–3267. [CrossRef]
137. Wiegel, C.; Sattler, S.; Göritz, A.S.; Diewald, M. Work-Related Stress and Cognitive Enhancement among University Teachers.

Anxiety Stress Coping 2016, 29, 100–117. [CrossRef]

Cells 2021, 10, 969 22 of 24

138. Johnston, J. Shaping the CRISPR Gene-Editing Debate: Questions About Enhancement and Germline Modification. Perspect. Biol.
Med. 2020, 63, 141–154. [CrossRef]

139. Cwik, B. Moving Beyond “Therapy” and “Enhancement” in the Ethics of Gene Editing. Camb. Q. Healthc. Ethics 2019, 28, 695–707.
[CrossRef] [PubMed]

140. Gumer, J.M. The Wisdom of Germline Editing: An Ethical Analysis of the Use of CRISPR-Cas9 to Edit Human Embryos. New
Bioeth. 2019, 25, 137–152. [CrossRef] [PubMed]

141. Locke, L.G. The Promise of CRISPR for Human Germline Editing and the Perils of “Playing God”. CRISPR J. 2020, 3, 27–31.
[CrossRef] [PubMed]

142. Zaami, S. Assisted Heterologous Fertilization and the Right of Donorconceived Children to Know Their Biological Origins. Clin.
Ter. 2018, 169, e39–e43. [CrossRef] [PubMed]

143. Borovecki, A.; Tozzo, P.; Cerri, N.; Caenazzo, L. Social Egg Freezing under Public Health Perspective: Just a Medical Reality or a
Women’s Right? An Ethical Case Analysis. J. Public Health Res. 2018, 7, 1484. [CrossRef]

144. Zaami, S.; Busardò, F.P. Elective Egg Freezing: Can You Really Turn Back the Clock? Eur. Rev. Med. Pharmacol. Sci. 2015, 19,
3537–3538.

145. Harwood, K. On the Ethics of Social Egg Freezing and Fertility Preservation for Nonmedical Reasons. Medicolegal Bioeth. 2015, 59.
[CrossRef]

146. Vizcarrondo, F.E. Human Enhancement: The New Eugenics. Linacre Q. 2014, 81, 239–243. [CrossRef] [PubMed]
147. Sparrow, R. A Not-so-New Eugenics. Harris and Savulescu on Human Enhancement. Hastings Cent. Rep. 2011, 41, 32–42.

[CrossRef]
148. Savulescu, J.; Kahane, G. The Moral Obligation to Create Children with the Best Chance of the Best Life. Bioethics 2009, 23,

274–290. [CrossRef] [PubMed]
149. Bennett, R. When Intuition Is Not Enough. Why the Principle of Procreative Beneficence Must Work Much Harder to Justify Its

Eugenic Vision: When Intuition Is Not Enough. Bioethics 2014, 28, 447–455. [CrossRef]
150. Bennett, R. The Fallacy of the Principle of Procreative Beneficence. Bioethics 2009, 23, 265–273. [CrossRef] [PubMed]
151. Holm, S.; Bennett, R. The Proper Scope of the Principle of Procreative Beneficence Revisited. Monash Bioeth. Rev. 2014, 32, 22–32.

[CrossRef]
152. Parker, M. The Best Possible Child. J. Med. Ethics 2007, 33, 279–283. [CrossRef]
153. Porter, A. Bioethics and Transhumanism. J. Med. Philos. 2017, 42, 237–260. [CrossRef]
154. Bostrom, N. Human Genetic Enhancements: A Transhumanist Perspective. J. Value Inq. 2003, 37, 493–506. [CrossRef]
155. Liao, S.M. Designing Humans: A Human Rights Approach. Bioethics 2019, 33, 98–104. [CrossRef]
156. Policy and Global Affairs; National Academies of Sciences, Engineering, and Medicine. Second International Summit on Human

Genome Editing: Continuing the Global Discussion: Proceedings of a Workshop in Brief ; Olson, S., Ed.; National Academies Press:
Washington, DC, USA, 2019; ISBN 9780309487900.

157. National Academies of Sciences, Engineering, and Medicine, Human Genome Editing: Science, Ethics, and Governance (The
National Academies Press: Washington, DC, USA, 2017) and Nuffield Council on Bioethics, Genome Editing and Human
Reproduction: Social and Ethical Issues (Nuffield Council on Bioethics: London, UK, 2018). Available online: https://www.
nuffieldbioethics.org/wp-cont (accessed on 27 February 2021).

158. Zheng, Q.; Lin, J.; Huang, J.; Zhang, H.; Zhang, R.; Zhang, X.; Cao, C.; Hambly, C.; Qin, G.; Yao, J.; et al. Reconstitution of UCP1
Using CRISPR/Cas9 in the White Adipose Tissue of Pigs Decreases Fat Deposition and Improves Thermogenic Capacity. Proc.
Natl. Acad. Sci. USA 2017, 114, E9474–E9482. [CrossRef] [PubMed]

159. Taning, C.N.T.; Van Eynde, B.; Yu, N.; Ma, S.; Smagghe, G. CRISPR/Cas9 in Insects: Applications, Best Practices and Biosafety
Concerns. J. Insect Physiol. 2017, 98, 245–257. [CrossRef] [PubMed]

160. Zhang, Y.; Ma, X.; Xie, X.; Liu, Y.-G. CRISPR/Cas9-Based Genome Editing in Plants. Prog. Mol. Biol. Transl. Sci. 2017, 149, 133–150.
[CrossRef] [PubMed]

161. Javed, M.R.; Sadaf, M.; Ahmed, T.; Jamil, A.; Nawaz, M.; Abbas, H.; Ijaz, A. CRISPR-Cas System: History and Prospects as a
Genome Editing Tool in Microorganisms. Curr. Microbiol. 2018, 75, 1675–1683. [CrossRef]

162. Acharya, S.; Mishra, A.; Paul, D.; Ansari, A.H.; Azhar, M.; Kumar, M.; Rauthan, R.; Sharma, N.; Aich, M.; Sinha, D.; et al.
Francisella Novicida Cas9 Interrogates Genomic DNA with Very High Specificity and Can Be Used for Mammalian Genome
Editing. Proc. Natl. Acad. Sci. USA 2019, 116, 20959–20968. [CrossRef] [PubMed]

163. Zetsche, B.; Gootenberg, J.S.; Abudayyeh, O.O.; Slaymaker, I.M.; Makarova, K.S.; Essletzbichler, P.; Volz, S.E.; Joung, J.; van der
Oost, J.; Regev, A.; et al. Cpf1 Is a Single RNA-Guided Endonuclease of a Class 2 CRISPR-Cas System. Cell 2015, 163, 759–771.
[CrossRef]

164. Windbichler, N.; Menichelli, M.; Papathanos, P.A.; Thyme, S.B.; Li, H.; Ulge, U.Y.; Hovde, B.T.; Baker, D.; Monnat, R.J.;
Burt, A.; et al. A Synthetic Homing Endonuclease-Based Gene Drive System in the Human Malaria Mosquito. Nature 2011, 473,
212–215. [CrossRef]

165. Esvelt, K.M.; Smidler, A.L.; Catteruccia, F.; Church, G.M. Concerning RNA-Guided Gene Drives for the Alteration of Wild
Populations. Elife 2014, 3, e03401. [CrossRef]

166. DiCarlo, J.E.; Chavez, A.; Dietz, S.L.; Esvelt, K.M.; Church, G.M. Safeguarding CRISPR-Cas9 Gene Drives in Yeast. Nat. Biotechnol.
2015, 33, 1250–1255. [CrossRef]

Cells 2021, 10, 969 23 of 24

167. Gantz, V.M.; Bier, E. Genome Editing. The Mutagenic Chain Reaction: A Method for Converting Heterozygous to Homozygous
Mutations. Science 2015, 348, 442–444. [CrossRef]

168. Hammond, A.; Galizi, R.; Kyrou, K.; Simoni, A.; Siniscalchi, C.; Katsanos, D.; Gribble, M.; Baker, D.; Marois, E.; Russell, S.; et al.
A CRISPR-Cas9 Gene Drive System Targeting Female Reproduction in the Malaria Mosquito Vector Anopheles Gambiae. Nat.
Biotechnol. 2016, 34, 78–83. [CrossRef]

169. Collins, J.P. Gene Drives in Our Future: Challenges of and Opportunities for Using a Self-Sustaining Technology in Pest and
Vector Management. BMC Proc. 2018, 12, 9. [CrossRef]

170. Kyrou, K.; Hammond, A.M.; Galizi, R.; Kranjc, N.; Burt, A.; Beaghton, A.K.; Nolan, T.; Crisanti, A. A CRISPR–Cas9 Gene Drive
Targeting Doublesex Causes Complete Population Suppression in Caged Anopheles Gambiae Mosquitoes. Nat. Biotechnol. 2018,
36, 1062–1066. [CrossRef]

171. Gantz, V.M.; Jasinskiene, N.; Tatarenkova, O.; Fazekas, A.; Macias, V.M.; Bier, E.; James, A.A. Highly Efficient Cas9-Mediated
Gene Drive for Population Modification of the Malaria Vector Mosquito Anopheles Stephensi. Proc. Natl. Acad. Sci. USA 2015, 112,
E6736–E6743. [CrossRef]

172. Isaacs, A.T.; Li, F.; Jasinskiene, N.; Chen, X.; Nirmala, X.; Marinotti, O.; Vinetz, J.M.; James, A.A. Engineered Resistance to
Plasmodium Falciparum Development in Transgenic Anopheles Stephensi. PLoS Pathog. 2011, 7, e1002017. [CrossRef] [PubMed]

173. Kranjc, N.; Crisanti, A.; Nolan, T.; Bernardini, F. Anopheles Gambiae Genome Conservation as a Resource for Rational Gene
Drive Target Site Selection. Insects 2021, 12, 97. [CrossRef] [PubMed]

174. Scudellari, M. Self-Destructing Mosquitoes and Sterilized Rodents: The Promise of Gene Drives. Nature 2019, 571, 160–162.
[CrossRef] [PubMed]

175. World Health Organization. World Malaria Report 2019. Issued on 4 December 2019. Available online: https://apps.who.int/
iris/rest/bitstreams/1262394/retrieve (accessed on 27 February 2021).

176. Esvelt, K.M.; Gemmell, N.J. Conservation Demands Safe Gene Drive. PLoS Biol. 2017, 15, e2003850. [CrossRef] [PubMed]
177. Zentner, G.E.; Wade, M.J. The Promise and Peril of CRISPR Gene Drives: Genetic Variation and Inbreeding May Impede the

Propagation of Gene Drives Based on the CRISPR Genome Editing Technology. Bioessays 2017, 39, 1700159. [CrossRef] [PubMed]
178. Frieß, J.L.; von Gleich, A.; Giese, B. Gene Drives as a New Quality in GMO Releases-a Comparative Technology Characterization.

PeerJ. 2019, 7, e6793. [CrossRef]
179. Wedell, N.; Price, T.A.R.; Lindholm, A.K. Gene Drive: Progress and Prospects. Proc. Biol. Sci. 2019, 286, 20192709. [CrossRef]
180. López Del Amo, V.; Bishop, A.L.; Sánchez, C.; Bennett, J.B.; Feng, X.; Marshall, J.M.; Bier, E.; Gantz, V.M. A Transcomplementing

Gene Drive Provides a Flexible Platform for Laboratory Investigation and Potential Field Deployment. Nat. Commun. 2020, 11,
352. [CrossRef] [PubMed]

181. Caplan, A.L.; Parent, B.; Shen, M.; Plunkett, C. No Time to Waste—The Ethical Challenges Created by CRISPR: CRISPR/Cas,
Being an Efficient, Simple, and Cheap Technology to Edit the Genome of Any Organism, Raises Many Ethical and Regulatory
Issues beyond the Use to Manipulate Human Germ Line Cells. EMBO Rep. 2015, 16, 1421–1426. [CrossRef] [PubMed]

182. Hayirli, T.C.; Martelli, P.F. Gene Drives as a Response to Infection and Resistance. Infect. Drug Resist. 2019, 12, 229–234. [CrossRef]
[PubMed]

183. Rode, N.O.; Estoup, A.; Bourguet, D.; Courtier-Orgogozo, V.; Débarre, F. Population Management Using Gene Drive: Molecular
Design, Models of Spread Dynamics and Assessment of Ecological Risks. Conserv. Genet. 2019, 20, 671–690. [CrossRef]

184. Sánchez, C.; Bennett, J.B.; Wu, S.L.; Rašić, G.; Akbari, O.S.; Marshall, J.M. Modeling Confinement and Reversibility of Threshold-
Dependent Gene Drive Systems in Spatially-Explicit Aedes Aegypti Populations. BMC Biol. 2020, 18, 50. [CrossRef]

185. Moro, D.; Byrne, M.; Kennedy, M.; Campbell, S.; Tizard, M. Identifying Knowledge Gaps for Gene Drive Research to Control
Invasive Animal Species: The next CRISPR Step. Glob. Ecol. Conserv. 2018, 13, e00363. [CrossRef]

186. Hayes, K.R.; Hosack, G.R.; Dana, G.V.; Foster, S.D.; Ford, J.H.; Thresher, R.; Ickowicz, A.; Peel, D.; Tizard, M.; De Barro, P.; et al.
Identifying and Detecting Potentially Adverse Ecological Outcomes Associated with the Release of Gene-Drive Modified
Organisms. J. Responsible Innov. 2018, 5, S139–S158. [CrossRef]

187. Noble, C.; Min, J.; Olejarz, J.; Buchthal, J.; Chavez, A.; Smidler, A.L.; DeBenedictis, E.A.; Church, G.M.; Nowak, M.A.; Esvelt, K.M.
Daisy-Chain Gene Drives for the Alteration of Local Populations. Proc. Natl. Acad. Sci. USA 2019, 116, 8275–8282. [CrossRef]

188. Brossard, D.; Belluck, P.; Gould, F.; Wirz, C.D. Promises and Perils of Gene Drives: Navigating the Communication of Complex,
Post-Normal Science. Proc. Natl. Acad. Sci. USA 2019, 116, 7692–7697. [CrossRef]

189. Schairer, C.E.; Triplett, C.; Buchman, A.; Akbari, O.S.; Bloss, C.S. Interdisciplinary Development of a Standardized Introduction to
Gene Drives for Lay Audiences. BMC Med. Res. Methodol. 2020, 20, 273. [CrossRef]

190. Faunce, T.; Ray, A.; Gardiner, C.; Preiss, T.; Burgio, G. Regulating RNA Research and CRISPR Gene Drives to Combat Biosecurity
Threats. J. Law Med. 2018, 26, 208–213. [PubMed]

191. Wegrzyn, R.D.; Lee, A.H.; Jenkins, A.L.; Stoddard, C.D.; Cheever, A.E. Genome Editing: Insights from Chemical Biology to
Support Safe and Transformative Therapeutic Applications. ACS Chem. Biol. 2018, 13, 333–342. [CrossRef] [PubMed]

192. Defense Advanced Research Project Agency (DARPA). Building the Safe Genes Toolkit; DARPA: Arlington, VA, USA, 2017;
Available online: https://www.darpa.mil/news-events/2017-07-19 (accessed on 27 February 2021).

193. Janik, E.; Niemcewicz, M.; Ceremuga, M.; Krzowski, L.; Saluk-Bijak, J.; Bijak, M. Various Aspects of a Gene Editing System-
CRISPR-Cas9. Int. J. Mol. Sci. 2020, 21, 6904. [CrossRef] [PubMed]

Cells 2021, 10, 969 24 of 24

194. National Academies of Sciences, Engineering, and Medicine (U.S.) (Ed.) Biodefense in the Age of Synthetic Biology; National
Academies Press: Washington, DC, USA, 2018; ISBN 9780309465182.

195. West, R.M.; Gronvall, G.K. CRISPR Cautions: Biosecurity Implications of Gene Editing. Perspect. Biol. Med. 2020, 63, 73–92.
[CrossRef] [PubMed]

196. Morsink, J. (Ed.) Universal Declaration of Human Rights. In The Universal Declaration of Human Rights: Origins, Drafting and
Intent; Pennsylvania Studies in Human, Rights; Ratified by the General Assembly as Resolution 217 during Its Third Session on
10 December 1948 in Paris, France; University of Pennsylvania Press: Philadelphian, PA, USA, 2000; ISBN 9780812217476.

197. Expert Advisory Committee on Developing Global Standards for Governance and Oversight of Human Genome Editing.
Background Paper Governance 1. Human Genome Editing. March 2019. Available online: https://www.who.int/ethics/topics/
human-genome-editing/WHO-Commissioned-Governance-1-paper-March-19.pdf (accessed on 2 March 2021).

  • Introduction
    • Overview of the Possible Applications of CRISPR for the Improvement of the Human Quality of Life
    • Technical Risks of Human Embryo Modifications
  • Beyond Therapeutic Safety and Efficacy, Genome Editing Entails Polarizing Ethical and Legal Quandaries
    • Current Lack of Understanding Stands in the Way of thorough Risk–Benefit Analysis
    • The Unsolved Quandary of Embryonic Status
    • Broadening Fields of Application
  • When the Line between “Therapy” and “Enhancement” Is Blurred
    • Is Enhancement Headed down a Dangerous Path?
    • Gene Drives: A Potential Threat to the Environment and Ecosystems?
    • Should CRISPR Be Viewed as a Potential Biosecurity Hazard?
    • Towards a Shared Regulatory Framework?
  • Conclusions: When Technology Outpaces Regulatory Governance, Harmonization May Be Key
  • References

REVIEW
published: 07 August 2020

doi: 10.3389/fonc.2020.01387

Frontiers in Oncology | www.frontiersin.org 1 August 2020 | Volume 10 | Article 1387

Edited by:

Israel Gomy,

Dana–Farber Cancer Institute,

United States

Reviewed by:

Nan Wu,

Peking Union Medical College

Hospital (CAMS), China

Tanja Kunej,

University of Ljubljana, Slovenia

Martin Roffe,

A.C. Camargo Cancer Center, Brazil

*Correspondence:

Charles M. Rudin

[email protected]

Triparna Sen

[email protected]

†Lead Contact

Specialty section:

This article was submitted to

Cancer Genetics,

a section of the journal

Frontiers in Oncology

Received: 09 March 2020

Accepted: 30 June 2020

Published: 07 August 2020

Citation:

Uddin F, Rudin CM and Sen T (2020)

CRISPR Gene Therapy: Applications,

Limitations, and Implications for the

Future. Front. Oncol. 10:1387.

doi: 10.3389/fonc.2020.01387

CRISPR Gene Therapy: Applications,
Limitations, and Implications for the
Future
Fathema Uddin1, Charles M. Rudin1,2* and Triparna Sen1,2*†

1 Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY,

United States, 2 Weill Cornell Medicine, Cornell University, New York, NY, United States

A series of recent discoveries harnessing the adaptive immune system of prokaryotes

to perform targeted genome editing is having a transformative influence across the

biological sciences. The discovery of Clustered Regularly Interspaced Short Palindromic

Repeats (CRISPR) and CRISPR-associated (Cas) proteins has expanded the applications

of genetic research in thousands of laboratories across the globe and is redefining our

approach to gene therapy. Traditional gene therapy has raised some concerns, as its

reliance on viral vector delivery of therapeutic transgenes can cause both insertional

oncogenesis and immunogenic toxicity. While viral vectors remain a key delivery vehicle,

CRISPR technology provides a relatively simple and efficient alternative for site-specific

gene editing, obliviating some concerns raised by traditional gene therapy. Although it

has apparent advantages, CRISPR/Cas9 brings its own set of limitations which must be

addressed for safe and efficient clinical translation. This review focuses on the evolution

of gene therapy and the role of CRISPR in shifting the gene therapy paradigm. We review

the emerging data of recent gene therapy trials and consider the best strategy to move

forward with this powerful but still relatively new technology.

Keywords: gene therapy, CRISPR/Cas9, homology-directed repair (HDR), non-homologous end joining (NHEJ),

clinical trial, ethics

INTRODUCTION

Gene therapy as a strategy to provide therapeutic benefit includes modifying genes via disruption,
correction, or replacement (1). Gene therapy has witnessed both early successes and tragic
failures in a clinical setting. The discovery and development of the CRISPR/Cas9 system has
provided a second opportunity for gene therapy to recover from its stigma and prove to be
valuable therapeutic strategy. The recent advent of CRISPR technology in clinical trials has
paved way for the new era of CRISPR gene therapy to emerge. However, there are several
technical and ethical considerations that need addressing when considering its use for patient
care. This review aims to (1) provide a brief history of gene therapy prior to CRISPR and
discuss its ethical dilemmas, (2) describe the mechanisms by which CRISPR/Cas9 induces gene
edits, (3) discuss the current limitations and advancements made for CRISPR technology for
therapeutic translation, and (4) highlight a few recent clinical trials utilizing CRISPR gene therapy
while opening a discussion for the ethical barriers that these and future trials may hinge upon.

Uddin et al. CRISPR Gene Therapy Current Applications

GENE THERAPY PRIOR TO
CRISPR—HISTORY, HURDLES, AND ITS
FUTURE

Origins of Gene Therapy
The introduction of gene therapy into the clinic provided hope
for thousands of patients with genetic diseases and limited
treatment options. Initially, gene therapy utilized viral vector
delivery of therapeutic transgenes for cancer treatment (2) or
monogenic disease (3). One of these pioneering clinical trials
involved ex vivo retroviral delivery of a selective neomycin-
resistance marker to tumor infiltrating leukocytes (TILs)
extracted from advanced melanoma patients (4). Although the
neomycin tagging of TILs did not have a direct therapeutic
intent and was used for tracking purposes, this study was the
first to provide evidence for both the feasibility and safety of
viral-mediated gene therapy. Soon after, the first clinical trial
that used gene therapy for therapeutic intent was approved in
1990 for the monogenic disease adenosine deaminase-severe
combined immunodeficiency (ADA-SCID). Two young girls
with ADA-SCID were treated with retroviruses for ex vivo
delivery of a wildtype adenosine deaminase gene to autologous
T-lymphocytes, which were then infused back into the patients
(5, 6). While one patient showed moderate improvement, the
other did not (5, 6) Although initial results were suboptimal, the
early evidence of feasibility prompted multiple subsequent gene
therapy trials using viral-mediated gene edition. However, this
was followed by some major setbacks.

Tragic Setbacks for Gene Therapy
Jesse Gelsinger, an 18-year-old with a mild form of the
genetic disease ornithine transcarbamylase (OTC) deficiency,
participated in a clinical trial which delivered a non-mutated
OTC gene to the liver through a hepatic artery injection of the
recombinant adenoviral vector housing the therapeutic gene.
Unfortunately, Jesse passed away 4 days after treatment (7). The
adenovirus vector triggered a much stronger immune response
in Jesse than it had in other patients, causing a chain of multiple
organ failures that ultimately led to his death (8). At the time
of the trial, adenoviral vectors were considered reasonably safe.
In preclinical development, however, two of the rhesus monkeys
treated with the therapy developed a similar pattern of fatal
hepatocellular necrosis (9). Shortly after, another gene therapy
trial led to the development of leukemia in several young
children induced by insertional oncogenesis from the therapy
(10). These trials opened for two forms of SCID (SCID-X1 or
common È chain deficiency) and adenosine deaminase deficiency
(ADA). The therapy used È-retroviral vectors for ex vivo delivery
of therapeutic transgenes to autologous CD34+ hematopoietic
stem cells, which were reintroduced to the patients (10). Five
patients developed secondary therapy-related leukemia, one of
whom died from the disease (11). Further investigation revealed
integration of the therapeutic gene into the LMO2 proto-
oncogene locus, presumably resulting in the development of
leukemia (12). Subsequent analyses have suggested a higher
frequency of insertional mutagenesis events with È-retroviral
vectors relative to other vectors (13). Together, these tragic events

prompted substantial post-hoc concerns regarding the nature
of appropriate informed consent and the stringency of safety
and eligibility parameters for gene therapy experimentation in
humans (14).

Shifting the Gene Therapy Paradigm
Almost two decades after these cases, gene therapy returned
in clinical trials with reengineered viruses designed with safety
in mind. Current clinical approaches are being scrutinized for
evidence of insertional mutagenesis and adverse immunogenic
reactions (15–18). Non-viral vectors have been used as an
alternative method for gene delivery, which have reduced
immunogenicity compared to their viral counterparts and
therefore greater tolerance for repeated administration. A
concern is whether these methods can be optimized to provide
equivalent efficiency of gene delivery to that provided by
viruses (19).

While viral vectors continue to be essential for current gene
therapy, the concerns and limitations of viral-mediated gene
edition has broadened the diversity of gene-editing approaches
being considered. Rather than introducing the therapeutic
gene into a novel (and potentially problematic) locus, a more
attractive strategy would be to directly correct the existing
genetic aberrations in situ. This alternative would allow the
pathological mutation to be repaired while averting the risk
of insertional oncogenesis. The discovery and repurposing of
nucleases for programmable gene editing made this possible,
beginning with the development of zinc finger nucleases
(ZFN) (20, 21), followed by transcription activator-like effector
nucleases (TALENs), meganucleases, and most recently, the
CRISPR/Cas system (22). While the other gene-editing tools
can induce genome editing at targeted sites under controlled
conditions, the CRISPR/Cas system has largely supplanted these
earlier advances due to its relatively low price, ease of use, and
efficient and precise performance. However, this technology is
often delivered with adeno-associated virus (AAV) vectors, and
thus does not completely avert risks associated with viruses.
Other delivery options are available to circumvent this issue,
each with their own advantages and challenges (see Delivery of
CRISPR Gene Therapy section). Of the CRISPR/Cas systems,
CRISPR/Cas9 is the most developed and widely used tool for
current genome editing.

CRISPR/Cas9 MEDIATED GENE EDITING

Pioneering Discoveries in CRISPR/Cas9
Technology
The bacterial CRISPR locus was first described by Francisco
Mojica (23) and later identified as a key element in the
adaptive immune system in prokaryotes (24). The locus
consists of snippets of viral or plasmid DNA that previously
infected the microbe (later termed “spacers”), which were found
between an array of short palindromic repeat sequences. Later,
Alexander Bolotin discovered the Cas9 protein in Streptococcus
thermophilus, which unlike other known Cas genes, Cas9 was
a large gene that encoded for a single-effector protein with
nuclease activity (25). They further noted a common sequence

Frontiers in Oncology | www.frontiersin.org 2 August 2020 | Volume 10 | Article 1387

Uddin et al. CRISPR Gene Therapy Current Applications

FIGURE 1 | Hallmarks of CRISPR Gene Therapy. Timeline highlighting major events of traditional gene therapy, CRISPR development, and CRISPR gene therapy. The

text in red denotes gene therapy events which have raised significant ethical concerns.

in the target DNA adjacent to the spacer, later known as
the protospacer adjacent motif (PAM)—the sequence needed
for Cas9 to recognize and bind its target DNA (25). Later
studies reported that spacers were transcribed to CRISPR RNAs
(crRNAs) that guide the Cas proteins to the target site of
DNA (26). Following studies discovered the trans-activating
CRISPR RNA (tracrRNA), which forms a duplex with crRNA
that together guide Cas9 to its target DNA (27). The potential
use of this system was simplified by introducing a synthetic
combined crRNA and tracrRNA construct called a single-guide
RNA (sgRNA) (28). This was followed by studies demonstrating
successful genome editing by CRISPR/Cas9 in mammalian cells,
thereby opening the possibility of implementing CRISPR/Cas9 in
gene therapy (29) (Figure 1).

Mechanistic Overview of
CRISPR/Cas9-Mediated Genome Editing
CRISPR/Cas9 is a simple two-component system used for
effective targeted gene editing. The first component is the
single-effector Cas9 protein, which contains the endonuclease
domains RuvC and HNH. RuvC cleaves the DNA strand
non-complementary to the spacer sequence and HNH cleaves
the complementary strand. Together, these domains generate
double-stranded breaks (DSBs) in the target DNA. The second
component of effective targeted gene editing is a single guide
RNA (sgRNA) carrying a scaffold sequence which enables
its anchoring to Cas9 and a 20 base pair spacer sequence
complementary to the target gene and adjacent to the PAM
sequence. This sgRNA guides the CRISPR/Cas9 complex to
its intended genomic location. The editing system then relies
on either of two endogenous DNA repair pathways: non-
homologous end-joining (NHEJ) or homology-directed repair
(HDR) (Figure 2). NHEJ occurs much more frequently in
most cell types and involves random insertion and deletion

of base pairs, or indels, at the cut site. This error-prone
mechanism usually results in frameshift mutations, often creating
a premature stop codon and/or a non-functional polypeptide.
This pathway has been particularly useful in genetic knock-out
experiments and functional genomic CRISPR screens, but it can
also be useful in the clinic in the context where gene disruption
provides a therapeutic opportunity. The other pathway, which is
especially appealing to exploit for clinical purposes, is the error-
free HDR pathway. This pathway involves using the homologous
region of the unedited DNA strand as a template to correct the
damaged DNA, resulting in error-free repair. Experimentally,
this pathway can be exploited by providing an exogenous donor
template with the CRISPR/Cas9 machinery to facilitate the
desired edit into the genome (30).

LIMITATIONS AND ADVANCEMENTS OF
CRISPR/Cas9

Off-Target Effects
A major concern for implementing CRISPR/Cas9 for gene
therapy is the relatively high frequency of off-target effects
(OTEs), which have been observed at a frequency of ≥50% (31).
Current attempts at addressing this concern include engineered
Cas9 variants that exhibit reduced OTE and optimizing guide
designs. One strategy that minimizes OTEs utilizes Cas9 nickase
(Cas9n), a variant that induces single-stranded breaks (SSBs),
in combination with an sgRNA pair targeting both strands
of the DNA at the intended location to produce the DSB
(32). Researchers have also developed Cas9 variants that are
specifically engineered to reduce OTEs while maintaining editing
efficacy (Table 1). SpCas9-HF1 is one of these high-fidelity
variants that exploits the “excess-energy” model which proposes
that there is an excess affinity between Cas9 and target DNA
which may be enabling OTEs. By introducing mutations to 4

Frontiers in Oncology | www.frontiersin.org 3 August 2020 | Volume 10 | Article 1387

Uddin et al. CRISPR Gene Therapy Current Applications

FIGURE 2 | CRISPR/Cas9 mediated gene editing. Cas9 in complex with the sgRNA targets the respective gene and creates DSBs near the PAM region. DNA

damage repair proceeds either through the NHEJ pathway or HDR. In the NHEJ pathway, random insertions and deletions (indels) are introduced at the cut side and

ligated resulting in error-prone repair. In the HDR pathway, the homologous chromosomal DNA serves as a template for the damaged DNA during repair, resulting in

error-free repair.

residues involved in direct hydrogen bonding between Cas9
and the phosphate backbone of the target DNA, SpCas9-HF1
has been shown to possess no detectable off-target activity in
comparison to wildtype SpCas9 (35). Other Cas9 variants that

have been developed include evoCas9 and HiFiCas9, both of
which contain altered amino acid residues in the Rec3 domain
which is involved in nucleotide recognition. Desensitizing
the Rec3 domain increases the dependence on specificity

Frontiers in Oncology | www.frontiersin.org 4 August 2020 | Volume 10 | Article 1387

Uddin et al. CRISPR Gene Therapy Current Applications

TABLE 1 | Cas9 variants.

Bacterial origin Cas9 variant Advantage Variant mutation PAM References

Streptococcus pyogenes Cas9-D1135E Improved

PAM

recognition

D1135E NGG (33)

Cas9-VQR Altered

PAM

D1135V/R1335Q/T1337R NGAN or

NGNG

Cas9-EQR Altered

PAM

D1135E/R1335Q/T1337R NGAG

Cas9-VRER Altered

PAM

D1135V/G1218R/R1335E/T1337R NGCG

Cas9-VRQR Altered

PAM

M495V/Y515N/K526E/R661Q NGA

Cas9-QQR1 Altered

PAM

G1218R/N1286Q/I1331F/D1332K/R1333Q/R1335Q/T1337R NAAG (34)

SpCas9-HF1 Reduced

OTE

N497A/R661A/Q695A/Q926A NGG (35)

eSpCas9 Reduced

OTE

K846A/K1003A/R1060A NGG (36)

HeFSpCas9 Reduced

OTE

N497A/R661A/Q695A/K846A/Q926A/K1003A/R1060A NGG (37)

evoCas9 Reduced

OTE

M495V/Y515N/K526E/R661Q NGG (38)

HiFiCas9 Reduced

OTE

R691A NGG (39)

Cas9n/Cas9D10A SSB

instead of DSB,

Reduced OTE

D10A NGG (40, 41)

Dimeric

dCas9-FokI

Reduced

OTE

dCas9 fused to FokI endonuclease domain NGG (42)

xCas9-3.7 Broad

PAM specificity

A262T/R324L/S409I/E480K/E543D/M694I/E1219V NG, GAA

or GAT

(43)

SpCas9-NG Minimal

PAM

R1335V/L1111R/D1135V/G1218R/E1219F/A1322R/T1337R NGN (44)

HypaCas9 Reduced

OTE

N692A/M694A/Q695A/H698A NGG (45)

Sniper-Cas9 Reduced

OTE

F539S/M763I/K890N NGG (46)

SpG Cas9 Minimal

PAM

D1135L/S1136W/G1218K/E1219Q/R1335Q/T1337R NGN (47)

SpRY Cas9 Minimal

PAM

D1135L/S1136W/G1218K/E1219Q/R1335Q/T1337R/L1111R/

A1322R/A61R/N1317R/R1333P

NRN>NYN

SpCas9-HF1 plus Reduced

OTE

N497A/Q695A/Q926A; amino acids 1005-1013 replaced

with two glycine

NGG (48)

eSpCas9 plus Reduced

OTE

K848A/R1060A; amino acids 1005-1013 replaced with two

glycine

NGG

Cas9_R63A/Q768A Reduced

OTE

R63A/Q768A NGG (49)

Staphylococcus aureus KKH SaCas9 Relaxed

PAM

E782K/N968K/R1015H NNNRRT (33)

SaCas9-HF Reduced

OTE

R245A/N413A/N419A/R654A NNGRRT (50)

SaCas9-NR Relaxed

PAM

N986R NNGRR (51)

SaCas9-RL Relaxed

PAM

N986R/R991L NNGRR

Streptococcus canis ScCas9 Minimal

PAM

N/A (wildtype) NNG (52)

Frontiers in Oncology | www.frontiersin.org 5 August 2020 | Volume 10 | Article 1387

Uddin et al. CRISPR Gene Therapy Current Applications

for the DNA:RNA heteroduplex to induce DSBs, thereby
reducing OTEs while maintaining editing efficacy (38, 39).
One of the more recent developments is the Cas9_R63A/Q768A
variant, in which the R63A mutation destabilizes R-loop
formation in the presence of mismatches and Q768A mutation
increases sensitivity to PAM-distal mismatches (49). Despite
the different strategies, the rational for generating many Cas9
variants with reduced OTEs has been to ultimately reduce general
Cas9 and DNA interactions and give a stronger role for the
DNA:RNA heteroduplex in facilitating the edits.

Optimizing guide designs can also reduce the frequency of
OTEs (31). Many features in an sgRNA determine specificity
including the seed sequence (a 10–12 bp region proximal to
PAM on 3′ of spacer sequence) (29, 53), GC content (54, 55),
and modifications such as 5′ truncation of the sgRNA (56).
Several platforms have also been designed to provide optimized
guide sequences against target genes, including E-Crisp (31, 57),
CRISPR-design, CasOFFinder, and others (31). However, many
of these tools are designed based on computational algorithms
with varying parameters or rely on phenotypic screens that may
be specific to cell types and genomes, generating appreciable
noise and lack of generalizability across different experimental
setups (58, 59). Recently, an additional guide design tool named
sgDesigner was developed that addressed these limitations by
employing a novel plasmid library in silico that contained
both the sgRNA and the target site within the same construct.
This allowed collecting Cas9 editing efficiency data in an
intrinsic manner and establish a new training dataset that avoids
the biases introduced through other models. Furthermore, a
comparative performance evaluation to predict sgRNA efficiency
of sgDesigner with 3 other commonly used tools (Doench Rule
Set 2, Sequence Scan for CRISPR and DeepCRISPR) revealed that
sgDesigner outperformed all 3 designer tools in 6 independent
datasets, suggesting that sgDesigner may be a more robust and
generalizable platform (60).

Protospacer Adjacent Motif Requirement
An additional limitation of the technology is the requirement for
a PAM near the target site. Cas9 from the bacteria Streptococcus
pyogenes (SpCas9) is one of the most extensively used Cas9s
with a relatively short canonical PAM recognition site: 5′NGG3′,
where N is any nucleotide. However, SpCas9 is relatively large
and difficult to package into AAV vectors (61, 62), the most
common delivery vehicle for gene therapy. Staphylococcus aureus
Cas9 (SaCas9) is a smaller ortholog that can be packaged
more easily in AAV vectors but has a longer PAM sequence:
5′NNGRRT3′ or 5′NNGRR(N)3′, where R is any purine, which
further narrows the window of therapeutic targeting sites.
Engineered SaCas9 variants have been made, such as KKH
SaCas9, which recognizes a 5′NNNRRT3′ PAM, broadening
the human targeting sites by 2- to 4-fold. OTEs, however, are
observed with frequencies similar to wildtype SaCas9 and need
to be considered in designing any therapeutic application (33).
Several other variants of SpCas9 have also been engineered
for broadening the gene target window including SpCas9-NG,
which recognizes a minimal NG PAM (44) and xCas9, which
recognizes a broad range of PAM including NG, GAA, and GAT

(43). A side by side comparison of both variants revealed that
while SpCas9-NG had a broader PAM recognition, xCas9 had
the lowest OTE in human cells (63). Another Cas9 ortholog
from the bacteria Streptococcus canis, ScCas9, has been recently
characterized with a minimal PAM specificity of 5′NNG3′ and
an 89.2% sequence homology to SpCas9 and comparable editing
efficiency to SpCas9 in both bacterial and human cells (52). The
most recent development is a variant of SpCas9 named SpRY that
has been engineered to be nearly PAMless, recognizing minimal
NRN > NYN PAMs. This new variant can potentially edit any
gene independent of a PAM requirement, and hence can be used
therapeutically against several genetic diseases (47).

Alternatively, RNA-targeting Cas9 variants have been
developed which also broaden the gene targeting spectrum by
mitigating PAM requirement restrictions. S. pyogenese Cas9
(SpyCas9) can be manipulated to target RNA by providing
a short oligonucleotide with a PAM sequence, known as a
PAMmer (64, 65), and thus eliminates the need for a PAM site
within the target region. Other subsets of Cas enzymes have
also been discovered that naturally target RNA independent of a
PAM, such as Cas13d. Upon further engineering of this effector,
CasRx was developed for efficient RNA-guided RNA targeting
in human cells (66, 67). Although RNA-targeting CRISPR
advances provide a therapeutic opportunity without the risk of
DNA-damage toxicity, they exclude the potential for editing a
permanent correction into the genome.

DNA-Damage Toxicity
CRISPR-induced DSBs often trigger apoptosis rather than the
intended gene edit (68). Further safety concerns were revealed
when using this tool in human pluripotent stem cells (hPSCs)
which demonstrated that p53 activation in response to the toxic
DSBs introduced by CRISPR often triggers subsequent apoptosis
(69). Thus, successful CRISPR edits are more likely to occur
in p53 suppressed cells, resulting in a bias toward selection
for oncogenic cell survival (70). In addition, large deletions
spanning kilobases and complex rearrangements as unintended
consequences of on-target activity have been reported in several
instances (71, 72), highlighting a major safety issue for clinical
applications of DSB-inducing CRISPR therapy. Other variations
of Cas9, such as catalytically inactive endonuclease dead Cas9
(dCas9) in which the nuclease domains are deactivated, may
provide therapeutic utility while mitigating the risks of DSBs (73).
dCas9 can transiently manipulate expression of specific genes
without introducing DSBs through fusion of transcriptional
activating or repressing domains or proteins to the DNA-binding
effector (74). Other variants such as Cas9n can also be considered,
which induces SSBs rather than DSBs. Further modifications
of these Cas9 variants has led to the development of base
editors and prime editors, a key innovation for safe therapeutic
application of CRISPR technology (see Precision Gene Editing
With CRISPR section).

Immunotoxicity
In addition to technical limitations, CRISPR/Cas9, like
traditional gene therapy, still raises concerns for immunogenic
toxicity. Charlesworth et al. showed that more than half of the

Frontiers in Oncology | www.frontiersin.org 6 August 2020 | Volume 10 | Article 1387

Uddin et al. CRISPR Gene Therapy Current Applications

human subjects in their study possessed preexisting anti-Cas9
antibodies against the most commonly used bacterial orthologs,
SaCas9 and SpCas9 (75). Furthermore, AAV vectors are also
widely used to deliver CRISPR components for gene therapy. To
this end, several Cas9 orthologs and AAV serotypes were tested
based on sequence similarities and predicted binding strength
to MHC class I and class II to screen for immune orthologs that
can be used for safe repeated administration of AAV-CRISPR
gene therapy. Although no two AAV serotypes were found to
completely circumvent immune recognition, the study verified
3 Cas9 orthologs [SpCas9, SaCas9, and Campylobacter jejuni
Cas9 (CjCas9)] which showed robust editing efficiency and
tolerated repeated administration due to reduced immunogenic
toxicity in mice immunized against AAV and Cas9 (76). A
major caveat is pre-existing immunity in humans against 2 of
these orthologs—SpCas9 and SaCas9, leaving CjCas9 as the
only current option for this cohort of patients. However, this
ortholog has not been well-studied in comparison to the other 2
orthologs and will need further investigation to provide evidence
for its safety and efficacy for clinical use. Future studies may
also identify other Cas9 immune-orthogonal orthologs for safe
repeated gene therapy.

Precision Gene Editing With CRISPR
Precise-genome editing is essential for prospects of CRISPR
gene therapy. Although HDR pathways can facilitate a desired
edit, its low efficiency renders its utility for precise gene editing
for clinical intervention highly limiting, with NHEJ as the
default pathway human cells take for repair. Enhancement of
HDR efficiency has been achieved via suppression of the NHEJ
pathway through chemical inhibition of key NHEJ modulating
enzymes such as Ku (77), DNA ligase IV (78), and DNA-
dependent protein kinases (DNA-PKcs) (79). Other strategies
that improve HDR efficiency include using single-stranded
oligodeoxynucleotide (ssODN) template, which contains the
homology arms to facilitate recombination and the desired
edit sequence, instead of double-stranded DNA (dsDNA).
Rationally designed ssODN templates with optimized length
complementarity have been shown to increase HDR rates up
to 60% in human cells for single nucleotide substitution (80).
Furthermore, cell cycle stage plays a key role in determining
the DNA-damage repair pathway a cell may take. HDR events
are generally restricted to late S and G2 phases of the cell
cycle, given the availability of the sister chromatid to serve as a
template at these stages, whereas NHEJ predominates the G1, S,
and G2 phases (81). Pharmacological arrest at the S phase with
aphidicolin increased HDR frequency in HEK293T with Cas9-
guide ribonucleoprotein (RNP) delivery. Interestingly, cell arrest
in the M phase using nocodazole with low concentrations of the
Cas9-guide RNP complex yielded higher frequencies of HDR
events in these cells, reaching a maximum frequency of up to
31% (82). Although HDR is considered to be restricted to mitotic
cells, a recent study revealed that the CRISPR/Cas9 editing
can achieve HDR in mature postmitotic neurons. Nishiyama
et al. successfully edited the CaMKIIα locus through HDR in
postmitotic hippocampal neurons of adult mice in vitro using
an AAV delivered Cas9, guide RNA, and donor template in

the CaMKIIα locus, which achieved successful HDR-mediated
edits in ∼30% of infected cells. Although HDR efficiency was
dose-dependent on AAV delivered HDR machinery and off-
target activity was not monitored, this study demonstrated
CRISPR’s potential utility for translational neuroscience after
further developments (83). To further exploit cell-cycle stage
control as a means to favor templated repair, Cas9 conjugation
to a part of Geminin, a substrate for G1 proteosome degradation,
can limit Cas9 expression to S, G2, and M stages. This strategy
was shown to facilitate HDR events while mitigating undesired
NHEJ edits in human immortalized and stem cells (84, 85). A
more recent strategy combined a chemically modified Cas9 to
the ssODN donor or a DNA adaptor that recruits the donor
template, either of which improved HDR efficiency by localizing
the donor template near the cleavage site (86). Despite these
advancements, HDR is still achieved at a relatively low efficiency
in eukaryotic cells and use of relatively harmful agents in
cells such as NHEJ chemical inhibitors may not be ideal in a
clinical setting.

A recent advancement that allows precision gene editing
independent of exploiting DNA damage response mechanisms
is the CRISPR base editing (BE) system. In this system,
a catalytically inactive dead Cas9 (dCas9) is conjugated to
deaminase, which can catalyze the conversion of nucleotides
via deamination. For increased editing efficiency, Cas9 nickase
(Cas9n) fused with deaminase is recently being more utilized
over dCas9 for base editing, as the nicks created in a single
strand of DNA induce higher editing efficiency. Currently, the
two types of CRISPR base editors are cytidine base editors
(CBEs) and adenosine base editors (ABEs). CBEs catalyze the
conversion of cytidine to uridine, which becomes thymine after
DNA replication. ABEs catalyze the conversion of adenosine
to inosine which becomes guanine after DNA replication (87).
Base editors provide a means to edit single nucleotides without
running the risk of causing DSB-induced toxicity. However,
base editors are limited to “A to T” and “C to G” conversions,
narrowing its scope for single-base gene edition to only these
bases. In addition, base editors still face some of the same
challenges as the previously described CRISPR systems, including
OTEs, more so with CBEs than ABEs (88, 89) and packaging
constraints, namely in AAV vectors due to the large size of base
editors (90). Furthermore, the editing window for base editors
are limited to a narrow range of a few bases upstream of the
PAM (90). More recently, prime editing has been developed as
a strategy to edit the genome to insert a desired stretch of edits
without inducing DSBs (91). This technology combines fusion of
Cas9n with a reverse transcriptase and a prime editing guide RNA
(pegRNA), which contains sgRNA sequence, primer binding site
(PBS), and an RNA template encoding the desired edit on the
3′ end. Prime editors use Cas9n to nick one strand of the DNA
and insert the desired edit via reverse transcription of the RNA
template. The synthesized edit is incorporated into the genome
and the unedited strand is cleaved and repaired to match the
inserted edit. With an optimized delivery system in place, base
editors and primer editors can open the door for precision gene
editing to correct and potentially cure a multitude of genetic
diseases (Figure 3).

Frontiers in Oncology | www.frontiersin.org 7 August 2020 | Volume 10 | Article 1387

Uddin et al. CRISPR Gene Therapy Current Applications

FIGURE 3 | Precise Gene Editing. (A) CRISPR/Cas9-HDR. Cas9 induces a DSB. The exogenous ssODN carrying the sequence for the desired edit and homology

arms is used as a template for HDR-mediated gene modification. (B) Base Editor. dCas9 or Cas9n is tethered to the catalytic portion of a deaminase. Cytosine

deaminase catalyzes the formation of uridine from cytosine. DNA mismatch repair mechanisms or DNA replication yield an C:G to T:A single nucleotide base edit.

Adenosine deaminase catalyzes the formation of inosine from adenosine. DNA mismatch repair mechanisms or DNA replication yield an A:T to G:C single nucleotide

base edit. (C) Prime Editor. Cas9n is tethered to the catalytic portion of reverse transcriptase. The prime editor system uses pegRNA, which contains the guide spacer

sequence, reverse transcriptase primer, which includes the sequence for the desired edit and a primer binding site (PBS). PBS hybridizes with the complementary

region of the DNA and reverse transcriptase transcribes new DNA carrying the desired edit. After cleavage of the resultant 5′ flap and ligation, DNA repair mechanisms

correct the unedited strand to match the edited strand. HDR, homology directed repair. DSB, double stranded break; SSB, single-stranded break; ssODN,

single-stranded oligodeoxynucleotide.

Frontiers in Oncology | www.frontiersin.org 8 August 2020 | Volume 10 | Article 1387

Uddin et al. CRISPR Gene Therapy Current Applications

Delivery of CRISPR Gene Therapy
The delivery modality of CRISPR tools greatly influences
its safety and therapeutic efficacy. While traditional gene
therapy utilizing viruses have been scrutinized for the risk
of immunotoxicity and insertional oncogenesis, AAV vectors
remain a key delivery vehicle for CRISPR gene therapy and
continues to be extensively used for its high efficiency of delivery
(92). The CRISPR toolkit can be packaged as plasmid DNA
encoding its components, including Cas9 and gRNA, or can be
delivered as mRNA of Cas9 and gRNA. Nucleic acids of CRISPR
can be packaged in AAV vectors for delivery or introduced to
target cells via electroporation/nucleofection or microinjection,
with the latter methods averting virus-associated risks. However,
microinjection can be technically challenging and is only suited
for ex vivo delivery. Electroporation is also largely used for
ex vivo but can be used in vivo for certain target tissues
(93). However, high-voltage shock needed to permeabilize cell
membranes via electroporation can be toxic and can lead to
permanent permeabilization of treated cells (94). In addition
to viral toxicity, AAV delivery of CRISPR components yields
longevity of expression, leading to greater incidence of OTEs.
Alternatively, delivery of the Cas9 protein and gRNA as RNP
complexes has reduced OTEs while maintained editing efficacy,
owing to its transient expression and rapid clearance in the
cell (95).

Once the delivery modality is selected, CRISPR/Cas9 edits can
be facilitated either ex vivo where cells are genetically modified
outside of the patient and reintroduced back, or in vivo with
delivery of the CRISPR components directly into the patient
where cells are edited (Figure 4). Both systems pose their own
set of advantages and challenges. Advantages for ex vivo delivery
include greater safety since patients are not exposed to the gene
altering tool, technical feasibility, and tighter quality control of
the edited cells. However, challenges to this method include
survival and retention of in vivo function of cells outside the
patient after genetic manipulation and extensive culture in vitro.
Also, an adequate supply of cells is needed for efficient re-
engraftment. These conditions limit this method to certain cell
types that can survive and be expanded in culture, such as
hematopoietic stem and progenitor cells (HSPCs) (96) and T
cells (97).

While ex vivo gene therapy has provided therapeutic benefit
for hematological disorders and cancer immunotherapy, many
tissue types are not suited for this method, severely limiting
its therapeutic utility for other genetic diseases. in vivo
manipulation is thus needed to expand CRISPR’s utility to treat
a broader range of genetic diseases, such as Duchenne muscular
dystrophy (DMD) (98) and hereditary tyrosinemia (99). CRISPR
components can be delivered in vivo systemically through
intravenous injections or can be locally injected to specific tissues
(Figure 4). With systemic delivery, the CRISPR components
and its vehicle are introduced into the circulatory system
where expression of the gene editing toolkit can be controlled
to target specific organs via tissue-specific promoters (100).
However, challenges of in vivo delivery include degradation by
circulating proteases or nucleases, opsonization by opsonins,
or clearance by the mononuclear phagocyte system (MPS).

Furthermore, the cargo must reach the target tissue and bypass
the vascular endothelium, which are often tightly connected
by cell-cell junctions (101), preventing accessibility to larger
delivery vehicles (>1 nm diameter). Additionally, once the
cargo has reached the target cells, they must be internalized,
which is generally facilitated through endocytosis where they
can be transported and degraded by lysosomal enzymes (102).
In addition, localization of the editing machinery near the
point of injection can result in uneven distribution of the
edited cell repertoire within the tissue, which may result in
suboptimal therapeutic outcomes (102). While advancements
are continuing to refine delivery techniques, the current
systems have allowed CRISPR gene therapy to be used in
the clinic.

BIOLOGICAL INTERVENTION OF
CRISPR/Cas9 IN CLINICAL TRIALS

Cancer Immunotherapy
The first CRISPR Phase 1 clinical trial in the US opened in
2018 with the intent to use CRISPR/Cas9 to edit autologous
T cells for cancer immunotherapy against several cancers with
relapsed tumors and no further curative treatment options. These
include multiple myeloma, melanoma, synovial sarcoma and
myxoid/round cell liposarcoma. This trial was approved by the
United States Food and Drug Administration (FDA) after careful
consideration of the risk to benefit ratios of this first application
of CRISPR gene therapy into the clinic. During this trial, T
lymphocytes were collected from the patients’ blood and ex vivo
engineered with CRISPR/Cas9 to knockout the α and β chains of
the endogenous T cell receptor (TCR), which recognizes a specific
antigen to mediate an immune response, and the programmed
cell death-1 (PD-1) protein, which attenuates immune response.
The cells were then transduced with lentivirus to deliver a gene
encoding a TCR specific for a NY-ESO-1 antigen, which has
been shown to be highly upregulated in the relapsed tumors
and thus can serve as a therapeutic target. Since then, many
trials have opened for CRISPR-mediated cancer immunotherapy
and is currently the most employed strategy for CRISPR gene
therapy (Table 2). A trial implementing this strategy using other
tools had already been conducted in both pre-clinical and clinical
settings, but this was the first time CRISPR/Cas9 was used to
generate the genetically modified T cells (97). The moderate
transition of switching only the tool used for an already approved
therapeutic strategy may have been key to paving the road for
using CRISPR’s novel abilities for gene manipulation, such as
targeted gene disruption.

Gene Disruption
The first clinical trial in the US using CRISPR to catalyze
gene disruption for therapeutic benefit were for patients
with sickle-cell anemia (SCD) and later β-thalassemia, by
Vertex Pharmaceuticals and CRISPR Therapeutics. This
therapy, named CTX001, increases fetal hemoglobin (HbF)
levels, which can occupy one or two of four hemoglobin
binding pockets on erythrocytes and thereby provides clinical
benefit for major β-hemoglobin diseases such as SCD and

Frontiers in Oncology | www.frontiersin.org 9 August 2020 | Volume 10 | Article 1387

Uddin et al. CRISPR Gene Therapy Current Applications

FIGURE 4 | Delivery of CRISPR Therapy. Nucleic acids encoding CRISPR/Cas9 or its RNP complex can be packaged into delivery vehicles. Once packaged, edits

can be facilitated either ex vivo or in vivo. Ex vivo editing involves extraction of target cells from the patient, cell culture, and expansion in vitro, delivery of the CRISPR

components to yield the desired edits, selection, and expansion of edited cells, and finally reintroduction of therapeutic edited cells into the patient. In vivo editing can

be systemically delivered via intravenous infusions to the patient, where the CRISPR cargo travels through the bloodstream via arteries leading to the target tissue, or

locally delivered with injections directly to target tissue. Once delivered, the edits are facilitated in vivo to provide therapeutic benefit.

β-thalassemia (103). The trial involved collecting autologous
hematopoietic stem and progenitor cells from peripheral blood
and using CRISPR/Cas9 to disrupt the intronic erythroid-specific
enhancer for the BCL11A gene (NCT03745287) as disruption

of this gene increases HbF expression (104–106). Genetically
modified hematopoietic stem cells with BCL11A disruption are
delivered by IV infusion after myeloablative conditioning
with busulfan to destroy unedited hematopoietic stem

Frontiers in Oncology | www.frontiersin.org 10 August 2020 | Volume 10 | Article 1387

Uddin et al. CRISPR Gene Therapy Current Applications

TABLE 2 | Biological intervention of CRISPR gene therapy in clinical trials.

Sponsor/affiliation Disease Gene target Clinial Trial ID CRISPR-Cas9 mediated intervention

University of Pennsylvania/Parker

Institute for Cancer

Immunotherapy/Tmunity

Multiple Myeloma, Melanoma,

Synovial Sarcoma,

Myxoid/Round Cell Liposarcoma

TCRα, TCRβ,

PDCD1

NCT03399448 NY-ESO-1 redirected autologous T cells with

CRISPR edited endogenous TCR and PD-1

Affiliated Hospital to Academy of

Military Medical Sciences/Peking

University/Capital Medical

University

HIV-1 CCR5 NCT03164135 CD34+ hematopoietic stem/progenitor cells

from donor are treated with CRISPR/Cas9

targeting CCR5 gene

CRISPR Therapeutics AG Multiple Myeloma TCRα, TCRβ, B2M NCT04244656 CTX120 B-cell maturation antigen

(BCMA)-directed T-cell immunotherapy

comprised of allogeneic T cells genetically

modified ex vivo using CRISPR-Cas9 gene

editing components

Crispr Therapeutics/Vertex Beta-Thalassemia, Thalassemia,

Genetic Diseases Inborn,

Hematologic Diseases,

Hemoglobinopathies

BCL11A NCT03655678 CTX001 (autologous CD34+ hHSPCs modified

with CRISPR-Cas9 at the erythroid

lineage-specific enhancer of the BCL11A gene)

Crispr Therapeutics B-cell MalignancyNon-Hodgkin

LymphomaB-cell Lymphoma

TCRα, TCRβ NCT04035434 CTX110 (CD19-directed T-cell immunotherapy

comprised of allogeneic T cells genetically

modified ex vivo using CRISPR-Cas9 gene

editing components)

Editas Medicine, Inc./Allergan Leber Congenital Amaurosis 10 CEP290 NCT03872479 Single escalating doses of AGN-151587

(EDIT-101) administered via subretinal injection

Vertex Pharmaceuticals

Incorporated/CRISPR

Therapeutics

Sickle Cell Disease,

Hematological Diseases,

Hemoglobinopathies

BCL11A NCT03745287 CTX001 (autologous CD34+ hHSPCs modified

with CRISPR-Cas9 at the erythroid

lineage-specific enhancer of the BCL11A gene)

Allife Medical Science and

Technology Co., Ltd.

Thalassemia HBB NCT03728322 Investigate the safety and efficacy of the gene

correction of HBB in patient-specific iHSCs

using CRISPR/Cas9

Yang Yang, The Affiliated Nanjing

Drum Tower Hospital of Nanjing

University Medical School

Stage IV Gastric Carcinoma,

Stage IV Nasopharyngeal

Carcinoma, T-Cell Lymphoma

Stage IV, Stage IV Adult Hodgkin

Lymphoma, Stage IV Diffuse

Large B-Cell Lymphoma

PDCD1 NCT03044743 CRISPR-Cas9 mediated PD-1 knockout-T cells

from autologous origin

First Affiliated Hospital, Sun

Yat-Sen University/Jingchu

University of Technology

Human Papillomavirus-Related

Malignant Neoplasm

HPV16 and HPV18

E6/E7 DNA

NCT03057912 Evaluate the safety and efficacy of TALEN-HPV

E6/E7 and CRISPR/Cas9-HPV E6/E7 in

treating HPV Persistency and HPV-related

Cervical Intraepithelial NeoplasiaI

Sichuan University/Chengdu

MedGenCell, Co., Ltd.

Metastatic Non-small Cell Lung

Cancer

PDCD1 NCT02793856 CRISPR-Cas9 mediated PD-1 knockout-T cells

from autologous origin

Peking University Metastatic Renal Cell Carcinoma PDCD1 NCT02867332 CRISPR-Cas9 mediated PD-1 knockout-T cells

from autologous origin

Peking University Hormone Refractory Prostate

Cancer

PDCD1 NCT02867345 CRISPR-Cas9 mediated PD-1 knockout-T cells

from autologous origin

Peking University Invasive Bladder Cancer Stage IV PDCD1 NCT02863913 CRISPR-Cas9 mediated PD-1 knockout-T cells

from autologous origin

Hangzhou Cancer

Hospital/Anhui Kedgene

Biotechnology Co., Ltd

Esophageal Cancer PDCD1 NCT03081715 CRISPR-Cas9 mediated PD-1 knockout-T cells

from autologous origin

Chinese PLA General Hospital Solid Tumor, Adult TCRα, TCRβ,

PDCD1

NCT03545815 Evaluate the feasibility and safety of

CRISPR-Cas9 mediated PD-1 and TCR

gene-knocked out chimeric antigen receptor

(CAR) T cells in patients with mesothelin

positive multiple solid tumors

Baylor College of Medicine/The

Methodist Hospital System

T-cell Acute Lymphoblastic

Leukemia, T-cell Acute

Lymphoblastic Lymphoma,

T-non-Hodgkin Lymphoma

CD7 NCT03690011 CRISPR-Cas9 mediated CD7 knockout-T cells

from autologous origin

(Continued)

Frontiers in Oncology | www.frontiersin.org 11 August 2020 | Volume 10 | Article 1387

Uddin et al. CRISPR Gene Therapy Current Applications

TABLE 2 | Continued

Sponsor/affiliation Disease Gene target Clinial Trial ID CRISPR-Cas9 mediated intervention

Chinese PLA General Hospital B Cell Leukemia, B Cell

Lymphoma

PDCD1 NCT03398967 Determine the safety of the allogenic

CRISPR-Cas9 gene-edited dual specificity

CD19 and CD20 or CD22 CAR-T cells

Chinese PLA General Hospital B Cell Leukemia, B Cell

Lymphoma

TCRα, TCRβ, B2M NCT03166878 CRISPR-Cas9 mediated TCR and B2M

knockout-T cells from allogenic origin for CD19

CAR-T

Chinese PLA General Hospital Solid Tumor, Adult PDCD1 NCT03747965 CRISPR-Cas9 mediated PD-1 knockout-T cells

from autologous origin

Xijing Hospital/Xi’An Yufan

Biotechnology Co., Ltd

Leukemia, Lymphoma HPK1 NCT04037566 CRISPR Gene Edited to Eliminate Endogenous

HPK1 (XYF19 CAR-T Cells)

cells in the bone marrow. Preliminary findings from
two patients receiving this treatment seem promising.
One SCD patient was reported to have 46.6% HbF and
94.7% erythrocytes expressing HbF after 4 months of CTX001
transfusions and one β-thalassemia patient is expressing 10.1
g/dL HbF out of 11.9 g/dL total hemoglobin, and 99.8%
erythrocytes expressing HbF after 9 months of the therapy.
Results from the clinical trial that has opened for this therapy
(NCT04208529) to assess the long-term risks and benefits of
CTX001 will dictate whether this approach can provide a novel
therapeutic opportunity for a disease that otherwise has limited
treatment options.

In vivo CRISPR Gene Therapy
While the aforementioned trials rely on ex vivo editing and
subsequent therapy with modified cells, in vivo approaches
have been less extensively employed. An exciting step forward
with CRISPR gene therapy has been recently launched with
a clinical trial using in vivo delivery of CRISPR/Cas9 for
the first time in patients. While in vivo editing has been
largely limited by inadequate accessibility to the target tissue,
a few organs, such as the eye, are accessible. Leber congenital
amaurosis (LCA) is a debilitating monogenic disease that results
in childhood blindness caused by a bi-allelic loss-of-function
mutation in the CEP290 gene, with no treatment options.
This therapy, named EDIT-101, delivers CRISPR/Cas9 directly
into the retina of LCA patients specifically with the intronic
IVS26 mutation, which drives aberrant splicing resulting in
a non-functional protein. The therapy uses an AAV5 vector
to deliver nucleic acid instructions for Staphylococcus aureus
Cas9 and two guides targeting the ends of the CEP290
locus containing the IVS26 mutation. The DSB induced
by Cas9 and both guides result in either a deletion or
inversion of the IVS26 intronic region, thus preventing the
aberrant splicing caused by the genetic mutation and enabling
subsequent translation of the functional protein (107). Potential
immunotoxicity or OTEs arising from nucleic acid viral
delivery will have to be closely monitored. Nonetheless, a
possibly curative medicine for genetic blindness using an in
vivo approach marks an important advancement for CRISPR
gene therapy.

CRISPR Editing in Human Embryos and
Ethical Considerations
While somatic editing for CRISPR therapy has been permitted
after careful consideration, human germline editing for
therapeutic intent remains highly controversial. With somatic
edition, any potential risk would be contained within the
individual after informed consent to partake in the therapy.
Embryonic editing not only removes autonomy in the decision-
making process of the later born individuals, but also allows
unforeseen and permanent side effects to pass down through
generations. This very power warrants proceeding with caution
to prevent major setbacks as witnessed by traditional gene
therapy. However, a controversial CRISPR trial in human
embryos led by Jiankui He may have already breached the
ethical standards set in place for such trials. This pilot study
involved genetic engineering of the C-C chemokine receptor
type 5 (CCR5) gene in human embryos, with the intention of
conferring HIV-resistance, as seen by a naturally occurring
CCR5132 mutation in a few individuals (108). However, based
on the limited evidence, CRISPR/Cas9 was likely used to target
this gene, but rather than replicate the naturally observed and
beneficial 32-base deletion, the edits merely induced DSBs at one
end of the deletion, allowing NHEJ to repair the damaged DNA
while introducing random, uncharacterized mutations. Thus, it
is unknown whether the resultant protein will function similarly
to the naturally occurring CCR5132 protein and confer HIV
resistance. In addition, only one of the two embryos, termed
with the pseudonym Nana, had successful edits in both copies
of the CCR5 gene, whereas the other embryo, with pseudonym
Lulu, had successful editing in only one copy. Despite these
findings, both embryos were implanted back into their mother,
knowing that the HIV-resistance will be questionable in Nana
and non-existent in Lulu (109, 110).

Furthermore, recent studies have shown that the mechanism
for infection of some variants of the highly mutable HIV
virus may heavily rely on the C-X-C chemokine receptor
type 4 (CXCR4) co-receptor (108, 111). With no attempts
at editing CXCR4, this adds yet another layer of skepticism
toward achieving HIV resistance by this strategy. In addition,
OTEs, particularly over the lifetime of an individual, remain
a major concern for applying this technology in humans.

Frontiers in Oncology | www.frontiersin.org 12 August 2020 | Volume 10 | Article 1387

Uddin et al. CRISPR Gene Therapy Current Applications

The recent advances in the editing tool to limit OTEs, such
as using high fidelity Cas9 variants, has not been exploited.
Furthermore, the rationale for selecting HIV prevention for the
first use of CRISPR in implanted human embryos contributes
to the poor risk to benefit ratio of this study, considering HIV
patients can live long, healthy lives on a drug regimen. A
more appropriate first attempt would have been to employ this
technology for a more severe disease. For example, correction
of the MYBPC3 gene is arguably a better target for embryonic
gene editing, as mutations in MYBPC3 can cause hypertrophic
cardiomyopathy (HCM), a heart condition responsible for most
sudden cardiac deaths in people under the age of 30. Gene
correction for this pathological mutation was achieved recently
for the first time in the US in viable human embryos using the
HDR-mediated CRISPR/Cas9 system. However, these embryos
were edited for basic research purposes and not intended for
implantation. In this study, sperm carrying the pathogenic
MYBPC3 mutation and the CRISPR/Cas9 machinery as an
RNP complex were microinjected into healthy donor oocytes
arrested at MII, achieving 72.4% homozygous wildtype embryos
as opposed to 47.4% in untreated embryos. The HDR-mediated
gene correction was observed at considerably high frequencies
with no detectable OTEs in selected blastomeres, likely owing
to the direct microinjection delivery of the RNP complex in the
early zygote. Interestingly, the maternal wildtype DNA was used
preferentially for templated repair over the provided exogenous
ssODN template (112). While evidence for gene correction was
promising, NHEJ mediated DNA repair was still observed in
many embryos, highlighting the need to improve HDR efficiency
before clinical application can be considered. Although strategies
have been developed to improve HDR, such as chemical
inhibitors of NHEJ (77–79), such techniques may have varying
outcomes in embryonic cells and side effects that may arise
from treatment needs to be investigated. Germline gene editing
will remain to be ethically unfavorable at its current state and
its discussions may not be considered until sufficient long-term
studies of the ongoing somatic CRISPR therapy clinical trials
are evaluated.

POTENTIAL FOR CRISPR THERAPEUTICS
DURING COVID-19 PANDEMIC

The rapidly advancing CRISPR technology may provide aid
during our rapidly evolving times. The recent outbreak of a novel
severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)
has led to a global pandemic (113). These pressing times call for
an urgent response to develop quick and efficient testing tools
and treatment options for coronavirus disease 2019 (COVID-
19) patients. Currently available methods for testing are relatively
time consuming with suboptimal accuracy and sensitivity (114).
The two predominant testing methods are molecular testing
or serological testing. The US Centers for Disease Control and
Prevention (CDC) has developed a real-time RT-PCR assay for
molecular testing for the presence of viral RNA to detect COVID-
19 (115). However, this assay has a roughly ∼30% false negative
rate (116, 117) with the turnaround time of several hours to
>24 h. Serological testing methods are much more rapid but lack

the ability to detect acute respiratory infection since antibodies
used to detect infection can take several days or weeks to develop.

Recently, a CRISPR Cas12-based assay named SARS-CoV-2
DETECTR has been developed for detection of COVID-19 with
a short turnaround time of about 40 min and a 95% reported
accuracy. The assay involves RNA extraction followed by reverse
transcription and simultaneous isothermal amplification using
the RT-LAMP method. Cas12 and a guide RNA against regions
of the N (nucleoprotein) gene and E (envelope) gene of SARS-
CoV-2 are then targeted, which can be visualized by cleavage of a
fluorescent reporter molecule. The assay also includes a laminar
flow strip for a visual readout, where a single band close to where
the sample was applied indicates a negative test and 2 higher
bands or a single higher band would indicate cleavage of the
fluorescent probe and hence positive for SARS-CoV-2 (118).

In addition to CRISPR’s diagnostic utility, CRISPR may
provide therapeutic options for COVID-19 patients. The recently
discovered Cas13 is an RNA-guided RNA-targeting endonuclease
may serve as a potential therapeutic tool against COVID-19.
PAC-MAN (Prophylactic Antiviral CRISPR in huMAN cells)
has been developed, which utilizes the Ruminococcus flavefaciens
derived VI-D CRISPR-Cas13d variant, selected for its small size
facilitating easier packaging in viral vehicles, high specificity,
and strong catalytic activity in human cells. This technique
was developed to simultaneously target multiple regions for
RNA degradation, opening the door for a much-needed pan-
coronavirus targeting strategy, given the evidence suggesting
relatively high mutation and recombination rates of SARS-CoV-
2 (119). With these advances, the CRISPR/Cas machinery may
again be implemented to serve its original purpose as a virus-
battling system to provide aid during this pandemic.

DISCUSSION

The birth of gene therapy as a therapeutic avenue began with
the repurposing of viruses for transgene delivery to patients
with genetic diseases. Gene therapy enjoyed an initial phase
of excitement, until the recognition of immediate and delayed
adverse effects resulted in death and caused a major setback.
More recently, the discovery and development of CRISPR/Cas9
has re-opened a door for gene therapy and changed the way
scientists can approach a genetic aberration—by fixing a non-
functional gene rather than replacing it entirely, or by disrupting
an aberrant pathogenic gene. CRISPR/Cas9 provides extensive
opportunities for programmable gene editing and can become a
powerful asset for modern medicine. However, lessons learned
from traditional gene therapy should prompt greater caution
in moving forward with CRISPR systems to avoid adverse
events and setbacks to the development of what may be a
unique clinically beneficial technology. A failure to take these
lessons into account may provoke further backlash against
CRISPR/Cas9 development and slow down progression toward
attaining potentially curative gene editing technologies.

Although CRISPR editing in humans remains a highly
debated and controversial topic, a few Regulatory Affairs
Certification (RAC)-reviewed and FDA-approved CRISPR gene
therapy trials have opened after thorough consideration of the
risk to benefit ratios. These first few approved trials, currently

Frontiers in Oncology | www.frontiersin.org 13 August 2020 | Volume 10 | Article 1387

Uddin et al. CRISPR Gene Therapy Current Applications

in Phase I/II, are only for patients with severe diseases, such
as cancers or debilitating monogenic diseases. The outcomes
of these trials will dictate how rapidly we consider using this
system to treat less severe diseases, as the risks of the technology
are better understood. A concern remains whether normalizing
CRISPR/Cas9 editing for less debilitating diseases may act as a
gateway for human genome editing for non-medical purposes,
such as altering genes in embryos to create offspring with certain
aesthetic traits. This fear of unnatural selection for unethical
reasons has likely become more tangible in the public’s view with
the strong media attention of the edited “CRISPR babies.” The
lasting effects of that trial and outcomes of the approved clinical
trials will greatly influence CRISPR’s future in gene therapy
and begin to answer the key questions we must consider as
we further explore this technology. These key questions include
how to avoid the mistakes of the past, who should decide
CRISPR’s therapeutic future, and how the ethical boundaries of
its applications should best be drawn.

AUTHOR CONTRIBUTIONS

FU researched and drafted the article. TS and CR supervised the
content. All authors wrote, reviewed, and edited the manuscript
before submission.

FUNDING

This work was supported by grants from the US National
Institutes of Health, including U24CA213274 and R01CA197936
(CR); Parker Institute of Cancer Immunotherapy grant (TS).

ACKNOWLEDGMENTS

The authors would like to thank Ms. Emily Costa, Dr. Alvaro
Quintanal Villalonga, and Dr. Rebecca Caesar for their excellent
assistance with editing the review.

REFERENCES

1. Humbert O, Davis L, Maizels N. Targeted gene therapies: tools,
applications, optimization. Crit Rev Biochem Mol Biol. (2012) 47:264–81.
doi: 10.3109/10409238.2012.658112

2. Amer MH. Gene therapy for cancer: present status and future perspective.
Mol Cell Ther. (2014) 2:27. doi: 10.1186/2052-8426-2-27

3. Prakash V, Moore M, Yanez-Munoz RJ. Current progress in therapeutic
gene editing for monogenic diseases. Mol Ther. (2016) 24:465–74.
doi: 10.1038/mt.2016.5

4. Rosenberg SA, Aebersold P, Cornetta K, Kasid A, Morgan RA,
et al. Gene transfer into humans–immunotherapy of patients with
advanced melanoma, using tumor-infiltrating lymphocytes modified
by retroviral gene transduction. N Engl J Med. (1990) 323:570–8.
doi: 10.1056/NEJM199008303230904

5. Blaese RM, Culver KW, Miller AD, Carter CS, Fleisher T, Clerici M, et al. T
lymphocyte-directed gene therapy for ADASCID: Initial trial results after 4
years. Science. (1995) 270:475–80. doi: 10.1126/science.270.5235.475

6. Bordignon C, Notarangelo LD, Nobili N, Ferrari G, Casorati G, Panina
P, et al. Gene therapy in peripheral blood lymphocytes and bone
marrow for ADA- immunodeficient patients. Science. (1995) 270:470–5.
doi: 10.1126/science.270.5235.470

7. Sibbald B. Death but one unintended consequence of gene-therapy trial.
CMAJ. (2001) 164:1612.

8. Raper SE, Chirmule N, Lee FS, Wivel NA, Bagg A, Gao GP, et al. Fatal
systemic inflammatory response syndrome in a ornithine transcarbamylase
deficient patient following adenoviral gene transfer. Mol Genet Metab. (2003)
80:148–58. doi: 10.1016/j.ymgme.2003.08.016

9. Nunes FA, Furth EE, Wilson JM, Raper SE. Gene transfer into the
liver of nonhuman primates with E1-deleted recombinant adenoviral
vectors: safety of readministration. Hum Gene Ther. (1999) 10:2515–26.
doi: 10.1089/10430349950016852

10. Cavazzana-Calvo M, Hacein-Bey S, De Saint Basile G, Gross F, Yvon
E, Nusbaum P, et al. Gene therapy of human severe combined
immunodeficiency (SCID)-X1 disease. Science. (2000) 288:669–72.
doi: 10.1126/science.288.5466.669

11. Herzog RW. Gene therapy for SCID-X1: round 2. Mol Ther. (2010) 18:1891.
doi: 10.1038/mt.2010.228

12. Hacein-Bey-Abina S, Von Kalle C, Schmidt M, Mccormack MP, Wulffraat
N, Leboulch P, et al. LMO2-associated clonal T cell proliferation in
two patients after gene therapy for SCID-X1. Science. (2003) 302:415–9.
doi: 10.1126/science.1088547

13. Maetzig T, Galla M, Baum C, Schambach A. Gammaretroviral vectors:
biology, technology and application. Viruses. (2011) 3:677–713.
doi: 10.3390/v3060677

14. Hammer MJ, Eckardt P, Barton-Burke M. Informed consent: a
clinical trials perspective. Oncol Nurs Forum. (2016) 43:694–6.
doi: 10.1188/16.ONF.694-696

15. Lee CS, Bishop ES, Zhang R, Yu X, Farina EM, Yan S, et al. Adenovirus-
mediated gene delivery: potential applications for gene and cell-based
therapies in the new era of personalized medicine. Genes Dis. (2017) 4:43–63.
doi: 10.1016/j.gendis.2017.04.001

16. Goswami R, Subramanian G, Silayeva L, Newkirk I, Doctor D, Chawla
K, et al. Gene therapy leaves a vicious cycle. Front Oncol. (2019) 9:297.
doi: 10.3389/fonc.2019.00297

17. Sadelain M. Insertional oncogenesis in gene therapy: how much of a risk?
Gene Ther. (2004) 11:569–73. doi: 10.1038/sj.gt.3302243

18. Nayak S, Herzog RW. Progress and prospects: immune responses
to viral vectors. Gene Ther. (2010) 17:295–304. doi: 10.1038/gt.
2009.148

19. Ramamoorth M, Narvekar A. Non viral vectors in gene therapy- an overview.
J Clin Diagn Res. (2015) 9:Ge01–06. doi: 10.7860/JCDR/2015/10443.5394

20. Bibikova M, Golic M, Golic KG, Carroll D. Targeted chromosomal cleavage
and mutagenesis in Drosophila using zinc-finger nucleases. Genetics. (2002)
161:1169–75. Available online at: https://www.genetics.org/content/161/3/
1169

21. Bibikova M, Beumer K, Trautman JK, Carroll D. Enhancing gene
targeting with designed zinc finger nucleases. Science. (2003) 300:764.
doi: 10.1126/science.1079512

22. Chandrasegaran S, Carroll D. Origins of programmable nucleases
for genome engineering. J Mol Biol. (2016) 428:963–89.
doi: 10.1016/j.jmb.2015.10.014

23. Mojica FJ, Juez G, Rodriguez-Valera F. Transcription at different salinities
of Haloferax mediterranei sequences adjacent to partially modified PstI
sites. Mol Microbiol. (1993) 9:613–21. doi: 10.1111/j.1365-2958.1993.
tb01721.x

24. Mojica FJ, Diez-Villasenor C, Soria E, Juez G. Biological significance
of a family of regularly spaced repeats in the genomes of archaea,
bacteria and mitochondria. Mol Microbiol. (2000) 36:244–6.
doi: 10.1046/j.1365-2958.2000.01838.x

25. Bolotin A, Quinquis B, Sorokin A, Ehrlich SD. Clustered regularly
interspaced short palindrome repeats (CRISPRs) have spacers
of extrachromosomal origin. Microbiology. (2005) 151:2551–61.
doi: 10.1099/mic.0.28048-0

Frontiers in Oncology | www.frontiersin.org 14 August 2020 | Volume 10 | Article 1387

Uddin et al. CRISPR Gene Therapy Current Applications

26. Brouns SJ, Jore MM, Lundgren M, Westra ER, Slijkhuis RJ, Snijders AP, et al.
Small CRISPR RNAs guide antiviral defense in prokaryotes. Science. (2008)
321:960–4. doi: 10.1126/science.1159689

27. Deltcheva E, Chylinski K, Sharma CM, Gonzales K, Chao Y, Pirzada ZA,
et al. CRISPR RNA maturation by trans-encoded small RNA and host factor
RNase III. Nature. (2011) 471:602–7. doi: 10.1038/nature09886

28. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier
E. A programmable dual-RNA-guided DNA endonuclease in adaptive
bacterial immunity. Science. (2012) 337:816–21. doi: 10.1126/science.
1225829

29. Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, et al. Multiplex
genome engineering using CRISPR/Cas systems. Science. (2013) 339:819–23.
doi: 10.1126/science.1231143

30. Cong L, Zhang F. Genome engineering using CRISPR-Cas9 system. Methods
Mol Biol. (2015) 1239:197–217. doi: 10.1007/978-1-4939-1862-1_10

31. Zhang XH, Tee LY, Wang XG, Huang QS, Yang SH. Off-target effects in
CRISPR/Cas9-mediated genome engineering. Mol Ther Nucleic Acids. (2015)
4:e264. doi: 10.1038/mtna.2015.37

32. Ran FA, Hsu PD, Lin CY, Gootenberg JS, Konermann S, Trevino AE, et al.
Double nicking by RNA-guided CRISPR Cas9 for enhanced genome editing
specificity. Cell. (2013) 154:1380–9. doi: 10.1016/j.cell.2013.08.021

33. Kleinstiver BP, Prew MS, Tsai SQ, Nguyen NT, Topkar VV, Zheng Z,
et al. Broadening the targeting range of Staphylococcus aureus CRISPR-
Cas9 by modifying PAM recognition. Nat Biotechnol. (2015) 33:1293–8.
doi: 10.1038/nbt.3404

34. Anders C, Bargsten K, Jinek M. Structural plasticity of PAM recognition by
engineered variants of the RNA-guided endonuclease Cas9. Mol Cell. (2016)
61:895–902. doi: 10.1016/j.molcel.2016.02.020

35. Kleinstiver BP, Pattanayak V, Prew MS, Tsai SQ, Nguyen NT, Zheng Z,
et al. High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide
off-target effects. Nature. (2016) 529:490–5. doi: 10.1038/nature16526

36. Slaymaker IM, Gao L, Zetsche B, Scott DA, Yan WX, Zhang F. Rationally
engineered Cas9 nucleases with improved specificity. Science. (2016) 351:84–
8. doi: 10.1126/science.aad5227

37. Kulcsár PI, Tálas A, Huszár K, Ligeti Z, Tóth E, Weinhardt N, et al. Crossing
enhanced and high fidelity SpCas9 nucleases to optimize specificity and
cleavage. Genome Biol. (2017) 18:190. doi: 10.1186/s13059-017-1318-8

38. Casini A, Olivieri M, Petris G, Montagna C, Reginato G, Maule G, et al. A
highly specific SpCas9 variant is identified by in vivo screening in yeast. Nat
Biotechnol. (2018) 36:265–71. doi: 10.1038/nbt.4066

39. Vakulskas CA, Dever DP, Rettig GR, Turk R, Jacobi AM, Collingwood
MA, et al. A high-fidelity Cas9 mutant delivered as a ribonucleoprotein
complex enables efficient gene editing in human hematopoietic stem and
progenitor cells. Nat Med. (2018) 24:1216–24. doi: 10.1038/s41591-018-
0137-0

40. Hsu PD, Scott DA, Weinstein JA, Ran FA, Konermann S, Agarwala V, et al.
DNA targeting specificity of RNA-guided Cas9 nucleases. Nat Biotechnol.
(2013) 31:827–32. doi: 10.1038/nbt.2647

41. Shen B, Zhang W, Zhang J, Zhou J, Wang J, Chen L, et al. Efficient genome
modification by CRISPR-Cas9 nickase with minimal off-target effects. Nat
Methods. (2014) 11:399–402. doi: 10.1038/nmeth.2857

42. Wyvekens N, Topkar VV, Khayter C, Joung JK, Tsai SQ. Dimeric
CRISPR RNA-guided FokI-dCas9 nucleases directed by truncated gRNAs
for highly specific genome editing. Hum Gene Ther. (2015) 26:425–31.
doi: 10.1089/hum.2015.084

43. Hu JH, Miller SM, Geurts MH, Tang W, Chen L, Sun N, et al. Evolved Cas9
variants with broad PAM compatibility and high DNA specificity. Nature.
(2018) 556:57–63. doi: 10.1038/nature26155

44. Nishimasu H, Shi X, Ishiguro S, Gao L, Hirano S, Okazaki S, et al.
Engineered CRISPR-Cas9 nuclease with expanded targeting space. Science.
(2018) 361:1259–62. doi: 10.1126/science.aas9129

45. Chen JS, Dagdas YS, Kleinstiver BP, Welch MM, Sousa AA,
Harrington LB, et al. Enhanced proofreading governs CRISPR-Cas9
targeting accuracy. Nature. (2017) 550:407–10. doi: 10.1038/nature
24268

46. Lee JK, Jeong E, Lee J, Jung M, Shin E, Kim YH, et al. Directed evolution
of CRISPR-Cas9 to increase its specificity. Nat Commun. (2018) 9:3048.
doi: 10.1038/s41467-018-05477-x

47. Walton RT, Christie KA, Whittaker MN, Kleinstiver BP. Unconstrained
genome targeting with near-PAMless engineered CRISPR-Cas9 variants.
Science. (2020) 368:290–6. doi: 10.1126/science.aba8853

48. Kulcsár PI, Tálas A, Tóth E, Nyeste A, Ligeti Z, Welker Z, et al. Blackjack
mutations improve the on-target activities of increased fidelity variants
of SpCas9 with 5′G-extended sgRNAs. Nat Commun. (2020) 11:1223.
doi: 10.1038/s41467-020-15021-5

49. Bratovič M, Fonfara I, Chylinski K, Gálvez EJC, Sullivan TJ, Boerno S, et al.
Bridge helix arginines play a critical role in Cas9 sensitivity to mismatches.
Nat Chem Biol. (2020) 16:587–95. doi: 10.1038/s41589-020-0490-4

50. Tan Y, Chu A, Bao S, Hoang DA, Kebede FT, Xiong W, et al.
Rationally engineered Staphylococcus aureus Cas9 nucleases with high
genome-wide specificity. Proc Natl Acad Sci USA. (2019) 116:20969–76.
doi: 10.1073/pnas.1906843116

51. Luan B, Xu G, Feng M, Cong L, Zhou R. Combined computational–
experimental approach to explore the molecular mechanism of SaCas9 with
a broadened DNA targeting range. J Am Chem Soc. (2019) 141:6545–52.
doi: 10.1021/jacs.8b13144

52. Chatterjee P, Jakimo N, Jacobson JM. Minimal PAM specificity
of a highly similar SpCas9 ortholog. Sci Adv. (2018) 4:eaau0766.
doi: 10.1126/sciadv.aau0766

53. Semenova E, Jore MM, Datsenko KA, Semenova A, Westra ER, Wanner B,
et al. Interference by clustered regularly interspaced short palindromic repeat
(CRISPR) RNA is governed by a seed sequence. Proc Natl Acad Sci USA.
(2011) 108:10098–103. doi: 10.1073/pnas.1104144108

54. Liu X, Homma A, Sayadi J, Yang S, Ohashi J, Takumi T. Sequence features
associated with the cleavage efficiency of CRISPR/Cas9 system. Sci Rep.
(2016) 6:19675. doi: 10.1038/srep19675

55. Bruegmann T, Deecke K, Fladung M. Evaluating the efficiency of gRNAs
in CRISPR/Cas9 mediated genome editing in poplars. Int J Mol Sci. (2019)
20:3623. doi: 10.3390/ijms20153623

56. Fu Y, Sander JD, Reyon D, Cascio VM, Joung JK. Improving CRISPR-
Cas nuclease specificity using truncated guide RNAs. Nat Biotechnol. (2014)
32:279–84. doi: 10.1038/nbt.2808

57. Heigwer F, Kerr G, Boutros M. E-CRISP: fast CRISPR target site
identification. Nat Methods. (2014) 11:122–3. doi: 10.1038/nmeth.2812

58. Yan J, Chuai G, Zhou C, Zhu C, Yang J, Zhang C, et al. Benchmarking
CRISPR on-target sgRNA and design. Brief Bioinform. (2017) 15:1–4.
doi: 10.1093/bib/bbx001

59. Cui Y, Xu J, Cheng M, Liao X, Peng S. Review of CRISPR/Cas9
sgRNA design tools. Interdiscipl Sci Comput Life Sci. (2018) 10:455–65.
doi: 10.1007/s12539-018-0298-z

60. Hiranniramol K, Chen Y, Liu W, Wang X. Generalizable sgRNA design for
improved CRISPR/Cas9 editing efficiency. Bioinformatics. (2020) 36:2684–
89. doi: 10.1093/bioinformatics/btaa041

61. Lino CA, Harper JC, Carney JP, Timlin JA. Delivering CRISPR: a
review of the challenges and approaches. Drug Deliv. (2018) 25:1234–57.
doi: 10.1080/10717544.2018.1474964

62. Lau CH, Suh Y. In vivo genome editing in animals using AAV-CRISPR
system: applications to translational research of human disease. F1000Res.
(2017) 6:2153. doi: 10.12688/f1000research.11243.1

63. Kim HK, Lee S, Kim Y, Park J, Min S, Choi JW, et al. High-throughput
analysis of the activities of xCas9, SpCas9-NG and SpCas9 at matched
and mismatched target sequences in human cells. Nat Biomed Eng. (2020)
4:111–24. doi: 10.1038/s41551-019-0505-1

64. O’ Connell MR, Oakes BL, Sternberg SH, East-Seletsky A, Kaplan M, Doudna
JA. Programmable RNA recognition and cleavage by CRISPR/Cas9. Nature.
(2014) 516:263–6. doi: 10.1038/nature13769

65. Strutt SC, Torrez RM, Kaya E, Negrete OA, Doudna JA. RNA-
dependent RNA targeting by CRISPR-Cas9. Elife. (2018) 7:e32724.
doi: 10.7554/eLife.32724

66. Konermann S, Lotfy P, Brideau NJ, Oki J, Shokhirev MN, Hsu PD.
Transcriptome engineering with RNA-targeting type VI-D CRISPR effectors.
Cell. (2018) 173:665–76.e14. doi: 10.1016/j.cell.2018.02.033

67. Larochelle S. CRISPR–Cas goes RNA. Nat Methods. (2018) 15:312.
doi: 10.1038/nmeth.4681

68. Hu Z, Yu L, Zhu D, Ding W, Wang X, Zhang C, et al. Disruption of
HPV16-E7 by CRISPR/Cas system induces apoptosis and growth inhibition

Frontiers in Oncology | www.frontiersin.org 15 August 2020 | Volume 10 | Article 1387

Uddin et al. CRISPR Gene Therapy Current Applications

in HPV16 positive human cervical cancer cells. BioMed Res Int. (2014)
2014:612823. doi: 10.1155/2014/612823

69. Ihry RJ, Worringer KA, Salick MR, Frias E, Ho D, Theriault K, et al. p53
inhibits CRISPR–Cas9 engineering in human pluripotent stem cells. Nat
Med. (2018) 24:939–46. doi: 10.1038/s41591-018-0050-6

70. Haapaniemi E, Botla S, Persson J, Schmierer B, Taipale J. CRISPR-Cas9
genome editing induces a p53-mediated DNA damage response. Nat Med.
(2018) 24:927–30. doi: 10.1038/s41591-018-0049-z

71. Kosicki M, Tomberg K, Bradley A. Repair of double-strand breaks induced
by CRISPR–Cas9 leads to large deletions and complex rearrangements. Nat
Biotechnol. (2018) 36:765–71. doi: 10.1038/nbt.4192

72. Cullot G, Boutin J, Toutain J, Prat F, Pennamen P, Rooryck C, et al. CRISPR-
Cas9 genome editing induces megabase-scale chromosomal truncations. Nat
Commun. (2019) 10:1136. doi: 10.1038/s41467-019-09006-2

73. Moses C, Nugent F, Waryah CB, Garcia-Bloj B, Harvey AR, Blancafort
P. Activating PTEN tumor suppressor expression with the CRISPR/dCas9
system. Nucleic Acids. (2019) 14:287–300. doi: 10.1016/j.omtn.2018.12.003

74. Xu X, Qi LS. A CRISPR-dCas toolbox for genetic engineering and synthetic
biology. J Mol Biol. (2019) 431:34–47. doi: 10.1016/j.jmb.2018.06.037

75. Charlesworth CT, Deshpande PS, Dever DP, Camarena J, Lemgart
VT, Cromer MK, et al. Identification of preexisting adaptive
immunity to Cas9 proteins in humans. Nat Med. (2019) 25:249–54.
doi: 10.1038/s41591-018-0326-x

76. Moreno AM, Palmer N, Alemán F, Chen G, Pla A, Jiang N, et al. Immune-
orthogonal orthologues of AAV capsids and of Cas9 circumvent the immune
response to the administration of gene therapy. Nat Biomed Eng. (2019)
3:806–16. doi: 10.1038/s41551-019-0431-2

77. Li G, Liu D, Zhang X, Quan R, Zhong C, Mo J, et al. Suppressing
Ku70/Ku80 expression elevates homology-directed repair efficiency
in primary fibroblasts. Int J Biochem Cell Biol. (2018) 99:154–60.
doi: 10.1016/j.biocel.2018.04.011

78. Maruyama T, Dougan SK, Truttmann MC, Bilate AM, Ingram JR, Ploegh
HL. Increasing the efficiency of precise genome editing with CRISPR-Cas9 by
inhibition of nonhomologous end joining. Nat Biotechnol. (2015) 33:538–42.
doi: 10.1038/nbt.3190

79. Robert F, Barbeau M, Éthier S, Dostie J, Pelletier J. Pharmacological
inhibition of DNA-PK stimulates Cas9-mediated genome editing. Genome
Med. (2015) 7:93. doi: 10.1186/s13073-015-0215-6

80. Richardson CD, Ray GJ, DeWitt MA, Curie GL, Corn JE. Enhancing
homology-directed genome editing by catalytically active and inactive
CRISPR-Cas9 using asymmetric donor DNA. Nat Biotechnol. (2016) 34:339–
44. doi: 10.1038/nbt.3481

81. Heyer WD, Ehmsen KT, Liu J. Regulation of homologous
recombination in eukaryotes. Annu Rev Genet. (2010) 44:113–39.
doi: 10.1146/annurev-genet-051710-150955

82. Lin S, Staahl BT, Alla RK, Doudna JA. Enhanced homology-directed human
genome engineering by controlled timing of CRISPR/Cas9 delivery. eLife.
(2014) 3:e04766. doi: 10.7554/eLife.04766.010

83. Nishiyama J, Mikuni T, Yasuda R. Virus-mediated genome editing
via homology-directed repair in mitotic and postmitotic cells in
mammalian brain. Neuron. (2017) 96:755–68.e5. doi: 10.1016/j.neuron.2017.
10.004

84. Gutschner T, Haemmerle M, Genovese G, Draetta GF, Chin L. Post-
translational regulation of Cas9 during G1 enhances homology-directed
repair. Cell Rep. (2016) 14:1555–66. doi: 10.1016/j.celrep.2016.01.019

85. Lomova A, Clark DN, Campo-Fernandez B, Flores-Bjurström C, Kaufman
ML, Fitz-Gibbon S, et al. Improving gene editing outcomes in human
hematopoietic stem and progenitor cells by temporal control of DNA repair.
Stem Cells. (2019) 37:284–94. doi: 10.1002/stem.2935

86. Ling X, Xie B, Gao X, Chang L, Zheng W, Chen H, et al. Improving the
efficiency of precise genome editing with site-specific Cas9-oligonucleotide
conjugates. Sci Adv. (2020) 6:eaaz0051. doi: 10.1126/sciadv.aaz0051

87. Eid A, Alshareef S, Mahfouz MM. CRISPR base editors: genome
editing without double-stranded breaks. Biochem J. (2018) 475:1955–64.
doi: 10.1042/BCJ20170793

88. Xin H, Wan T, Ping Y. Off-targeting of base editors: BE3 but not ABE induces
substantial off-target single nucleotide variants. Sig Transduct Target Ther.
(2019) 4:9. doi: 10.1038/s41392-019-0044-y

89. Lee HK, Smith HE, Liu C, Willi M, Hennighausen L. Cytosine base editor
4 but not adenine base editor generates off-target mutations in mouse
embryos. Commun Biol. (2020) 3:19. doi: 10.1038/s42003-019-0745-3

90. Rees HA, Liu DR. Base editing: precision chemistry on the genome
and transcriptome of living cells. Nat Rev Genet. (2018) 19:770–88.
doi: 10.1038/s41576-018-0059-1

91. Anzalone AV, Randolph PB, Davis JR, Sousa AA, Koblan LW, Levy JM, et al.
Search-and-replace genome editing without double-strand breaks or donor
DNA. Nature. (2019) 576:149–57. doi: 10.1038/s41586-019-1711-4

92. Xu X, Wan T, Xin H, Li D, Pan H, Wu J, et al. Delivery of
CRISPR/Cas9 for therapeutic genome editing. J Gene Med. (2019) 21:e3107.
doi: 10.1002/jgm.3107

93. Wilbie D, Walther J, Mastrobattista E. Delivery aspects of CRISPR/Cas
for in vivo genome editing. Accounts Chem Res. (2019) 52:1555–64.
doi: 10.1021/acs.accounts.9b00106

94. Hui S-W. Overview of drug delivery and alternative methods to
electroporation bt – electroporation protocols: preclinical and clinical gene
medicine. In: Li S, editor. Electroporation Protocols Methods in Molecular
Biology. Totowa, NJ: Humana Press (2008). p. 91–107.

95. Kim S, Kim D, Cho SW, Kim J, Kim JS. Highly efficient RNA-guided genome
editing in human cells via delivery of purified Cas9 ribonucleoproteins.
Genome Res. (2014) 24:1012–9. doi: 10.1101/gr.171322.113

96. Yu KR, Natanson H, Dunbar CE. Gene editing of human hematopoietic
stem and progenitor cells: promise and potential hurdles. Human Gene Ther.
(2016) 27:729–40. doi: 10.1089/hum.2016.107

97. Baylis F, Mcleod M. First-in-human phase 1 CRISPR gene editing
cancer trials: are we ready? Curr Gene Ther. (2017) 17:309–19.
doi: 10.2174/1566523217666171121165935

98. Zhang Y, Li H, Min YL, Sanchez-Ortiz E, Huang J, Mireault AA, et al.
Enhanced CRISPR-Cas9 correction of Duchenne muscular dystrophy in
mice by a self-complementary AAV delivery system. Sci Adv. (2020)
6:eaay6812. doi: 10.1126/sciadv.aay6812

99. Pankowicz FP, Barzi M, Legras X, Hubert L, Mi T, Tomolonis JA, et al.
Reprogramming metabolic pathways in vivo with CRISPR/Cas9 genome
editing to treat hereditary tyrosinaemia. Nat Commun. (2016) 7:12642.
doi: 10.1038/ncomms12642

100. Ablain J, Zon LI. Tissue-specific gene targeting using CRISPR/Cas9. Methods
Cell Biol. (2016) 135:189–202. doi: 10.1016/bs.mcb.2016.03.004

101. Komarova Y, Malik AB. Regulation of endothelial permeability via
paracellular and transcellular transport pathways. Annu Rev Physiol. (2010)
72:463–93. doi: 10.1146/annurev-physiol-021909-135833

102. Tong S, Moyo B, Lee CM, Leong K, Bao G. Engineered materials for in
vivo delivery of genome-editing machinery. Nat Rev Mater. (2019) 4:726–37.
doi: 10.1038/s41578-019-0145-9

103. Akinsheye I, Alsultan A, Solovieff N, Ngo D, Baldwin CT, Sebastiani P,
et al. Fetal hemoglobin in sickle cell anemia. Blood. (2011) 118:19–27.
doi: 10.1182/blood-2011-03-325258

104. Basak A, Sankaran VG. Regulation of the fetal hemoglobin silencing factor
BCL11A. Ann NY Acad Sci. (2016) 1368:25–30. doi: 10.1111/nyas.13024

105. Basak A, Hancarova M, Ulirsch JC, Balci TB, Trkova M, Pelisek
M, et al. BCL11A deletions result in fetal hemoglobin persistence
and neurodevelopmental alterations. J Clin Invest. (2015) 125:2363–8.
doi: 10.1172/JCI81163

106. A Safety and Efficacy Study Evaluating CTX001 in Subjects With Severe
Sickle Cell Disease. Vertex Pharmaceuticals Incorporated. Available online
at: https://clinicaltrials.gov/ct2/show/NCT03745287

107. Maeder ML, Stefanidakis M, Wilson CJ, Baral R, Barrera LA, Bounoutas
GS, et al. Development of a gene-editing approach to restore vision
loss in Leber congenital amaurosis type 10. Nat Med. (2019) 25:229–33.
doi: 10.1038/s41591-018-0327-9

108. Ye L, Wang J, Beyer AI, Teque F, Cradick TJ, Qi Z, et al. Seamless
modification of wild-type induced pluripotent stem cells to the natural
CCR5Delta32 mutation confers resistance to HIV infection. Proc Natl Acad
Sci USA. (2014) 111:9591–6. doi: 10.1073/pnas.1407473111

109. Cohen J. Did CRISPR help—or harm—the first-ever gene-edited babies?
Science. (2019). doi: 10.1126/science.aay9569

110. Greely HT. CRISPR’d babies: human germline genome editing in the ’He
Jiankui affair’. J Law Biosci. (2019) 6:111–83. doi: 10.1093/jlb/lsz010

Frontiers in Oncology | www.frontiersin.org 16 August 2020 | Volume 10 | Article 1387

Uddin et al. CRISPR Gene Therapy Current Applications

111. Alkhatib G. The biology of CCR5 and CXCR4. Curr Opin HIV AIDS. (2009)
4:96–103. doi: 10.1097/COH.0b013e328324bbec

112. Ma H, Marti-Gutierrez N, Park SW, Wu J, Lee Y, Suzuki K, et al. Correction
of a pathogenic gene mutation in human embryos. Nature. (2014) 548:413–9.
doi: 10.1038/nature23305

113. Sironi M, Hasnain SE, Rosenthal B, Phan T, Luciani F, Shaw MA,
et al. SARS-CoV-2 and COVID-19: a genetic, epidemiological, and
evolutionary perspective. Infect Genet Evolut. (2020) 84:104384.
doi: 10.1016/j.meegid.2020.104384

114. Zhai P, Ding Y, Wu X, Long J, Zhong Y, Li Y. The epidemiology, diagnosis
and treatment of COVID-19. Int J Antimicrobial Agents. (2020) 55:105955.
doi: 10.1016/j.ijantimicag.2020.105955

115. US Centers for Disease Control and Prevention. Real-time RT–PCR Panel for
Detection 2019-nCoV. Centers for Disease Control and Prevention (2020).
Available online at: https://www.cdc.gov/coronavirus/2019-ncov/lab/rt-pcr-
detection-instructions.html

116. Fang Y, Zhang H, Xie J, Lin M, Ying L, Pang P. Sensitivity of chest
CT for COVID-19: comparison to RT-PCR. Radiology. (2020) 200432.
doi: 10.1148/radiol.2020200432

117. Wikramaratna P, Paton RS, Ghafari M, Lourenco J. Estimating false-
negative detection rate of SARS-CoV-2 by RT-PCR. BMJ. (2020).
doi: 10.1101/2020.04.05.20053355

118. Broughton JP, Deng X, Yu G, Fasching CL, Servellita V, Singh J, et al.
CRISPR–Cas12-based detection of SARS-CoV-2. Nat Biotechnol. (2020)
38:870–4. doi: 10.1038/s41587-020-0513-4

119. Abbott TR, Dhamdhere G, Liu Y, Lin X, Goudy L, Zeng L, et al. Development
of CRISPR as an antiviral strategy to combat SARS-CoV-2 and influenza.
Cell. (2020) 181:865–76.e12. doi: 10.1016/j.cell.2020.04.020

Conflict of Interest: CR has consulted regarding oncology drug development
with AbbVie, Amgen, Ascentage, Astra Zeneca, Celgene, Daiichi Sankyo,
Genentech/Roche, Ipsen, Loxo, and Pharmar, and is on the scientific advisory
boards of Harpoon Therapeutics and Bridge Medicines.

The remaining authors declare that the research was conducted in the absence of
any commercial or financial relationships that could be construed as a potential
conflict of interest.

Copyright © 2020 Uddin, Rudin and Sen. This is an open-access article distributed

under the terms of the Creative Commons Attribution License (CC BY). The use,

distribution or reproduction in other forums is permitted, provided the original

author(s) and the copyright owner(s) are credited and that the original publication

in this journal is cited, in accordance with accepted academic practice. No use,

distribution or reproduction is permitted which does not comply with these terms.

Frontiers in Oncology | www.frontiersin.org 17 August 2020 | Volume 10 | Article 1387

  • CRISPR Gene Therapy: Applications, Limitations, and Implications for the Future
    • Introduction
    • Gene Therapy Prior to Crispr—History, Hurdles, and its Future
      • Origins of Gene Therapy
      • Tragic Setbacks for Gene Therapy
      • Shifting the Gene Therapy Paradigm
    • CRISPR/Cas9 Mediated Gene Editing
      • Pioneering Discoveries in CRISPR/Cas9 Technology
      • Mechanistic Overview of CRISPR/Cas9-Mediated Genome Editing
    • Limitations and Advancements of CRISPR/Cas9
      • Off-Target Effects
      • Protospacer Adjacent Motif Requirement
      • DNA-Damage Toxicity
      • Immunotoxicity
      • Precision Gene Editing With CRISPR
      • Delivery of CRISPR Gene Therapy
    • Biological Intervention of CRISPR/Cas9 in Clinical Trials
      • Cancer Immunotherapy
      • Gene Disruption
      • In vivo CRISPR Gene Therapy
      • CRISPR Editing in Human Embryos and Ethical Considerations
    • Potential for CRISPR Therapeutics During COVID-19 Pandemic
    • Discussion
    • Author Contributions
    • Funding
    • Acknowledgments
    • References

Leading Edge

Review

Heritable human genome editing:
Research progress, ethical considerations,
and hurdles to clinical practice
Jenna Turocy,1 Eli Y. Adashi,2 and Dieter Egli1,3,4,*
1Department of Obstetrics and Gynecology, Columbia University, New York, NY 10032, USA
2Professor of Medical Science, Brown University, Providence, RI, USA
3Department of Pediatrics and Naomi Berrie Diabetes Center, Columbia University, New York, NY 10032, USA
4Columbia University Stem Cell Initiative, New York, NY 10032, USA
*Correspondence: [email protected]

https://doi.org/10.1016/j.cell.2021.02.036

ll

SUMMARY

Our genome at conception determines much of our health as an adult. Most human diseases have a heritable
component and thus may be preventable through heritable genome editing. Preventing disease from the
beginning of life before irreversible damage has occurred is an admirable goal, but the path to fruition remains
unclear. Here, we review the significant scientific contributions to the field of human heritable genome edit-
ing, the unique ethical challenges that cannot be overlooked, and the hurdles that must be overcome prior to
translating these technologies into clinical practice.

INTRODUCTION

The introduction of genome editing using clustered regularly

interspaced short palindromic repeats (CRISPR)-based tech-

nologies generated tremendous enthusiasm as well as con-

troversy within the medical and public communities. Herita-

ble Human Genome Editing (HHGE) has the potential to

treat or even eradicate genetic diseases. By addressing ge-

netic disease before the defect is amplified through cell pro-

liferation during development, HHGE may prove to be more

effective than any other treatment being developed today,

including somatic gene or drug therapy (Figure 1). Somatic

gene therapies are limited in their ability to reverse damage

that has already occurred and to reach the billions of cells

needed to adequately treat the disease. The brief in vitro

culture of a human embryo as routinely practiced in IVF

clinics provides a readily accessible window for potential

prevention of numerous conditions that later in life are diffi-

cult to manage, let alone cure. This hope provides the ratio-

nale for research, but not yet for therapy, as criteria of effi-

cacy and safety have yet to be met. HHGE also raises

difficult ethical and regulatory questions. Manipulations of

the early embryo are highly consequential, both with regard

to potential benefits but also with regard to risks. Other re-

views have also discussed heritable genome editing, empha-

sizing its potential for understanding gene function in the

early human embryo (Lea and Niakan, 2019; Rossant,

2018; Plaza Reyes and Lanner, 2017). Here, we summarize

the research on human heritable genome editing including

its potential therapeutic value, ethical implications, and

new risks that were identified in recent studies.

DNA DOUBLE-STRAND BREAKS ALLOW GENOME
EDITING IN HUMAN EMBRYOS

Studies since the early 1980s have demonstrated the power of

using DNA double-stand breaks (DSBs) for targeted genetic

change, first in yeast and then in mammalian cells (reviewed by

Jasin and Rothstein, 2013). DNA DSBs may occur during the

DNA replication process, after exposure to ionizing radiation or

chemotherapy, or after experimental manipulation and are re-

paired mainly by one of two repair pathways: nonhomologous

end joining (NHEJ) or homology directed repair (HDR). Based

on the pioneering studies in model organisms, it became evident

that genome editing through the targeted induction of a DSB may

be an ideal approach for the correction of disease-causing mu-

tations.

A critical breakthrough was the discovery of DNA sequence

specific nucleases amenable to design. Engineered nucleases

can target a genetic sequence and create a DSB at a unique po-

sition. During the repair process, the original DNA sequence in

the vicinity of the cut is altered, creating a new sequence. Several

biological systems and types of nucleases such as transcription

activator-like effector nucleases (TALENs) and zinc finger nucle-

ases (ZFNs) provide these capabilities for editing double-

stranded DNA. Both TALENs and ZFNs consist of a combination

of repetitive protein segments, each with DNA binding specificity

of 1 or 3 nucleotides, to generate a protein with a DNA binding

domain that is specific to a single site in the genome. However,

it was the discovery of RNA-guided nucleases that made

changes in the genome readily accessible and scalable for a

wide range of applications. Shortly after the function of the

RNA-guided endonuclease CRISPR-Cas9 was first described

Cell 184, March 18, 2021 ª 2021 Elsevier Inc. 1561

Figure 1. Pathways to parenthood without disease
Review of potential pathways to parenthood for couples with known heritable genetic disease who wish to avoid disease inheritance. Currently, couples may
choose adoption, prenatal genetic diagnosis followed by selective pregnancy termination if affected, use of donor gametes, or preimplantation genetic testing for
IVF-generated embryos. Mitochondrial replacement therapy and heritable human genome editing may provide a future pathway for couples wishing to avoid
passing on a known genetic disease. Research in genome editing has targeted somatic cells, in utero fetal cells, embryos, gametes, as well as in vitro-derived
gametes and haploid cells. Potential disease targets include single-gene, polygenic disease, disease risk alleles, aneuploidy, and mitochondrial disease.

ll

1562 Cell 184, March 18, 2021

Review

ll
Review

(Gasiunas et al., 2012; Jinek et al., 2012), it was adapted to modi-

fying the genome of cultured mammalian cells (Cong et al., 2013;

Mali et al., 2013) followed by human embryos (Liang et al., 2015;

Kang et al., 2016; Tang et al., 2017) and reviewed in Lea and Nia-

kan (2019).

Initial CRISPR-Cas9-mediated studies in human embryos

focused on rates of mutation correction, introduction of off-

target edits, and mosaicism—multiple different genetic out-

comes within the same embryo (Liang et al., 2015; Tang et al.,

2017; Kang et al., 2016) (Figure 2). The first such study made

use of non-viable tripronuclear (3PN) embryos. While this exper-

imental approach took advantage of embryos that would be clin-

ically discarded, developmental outcomes and karyotypes are

difficult to interpret and were hence not evaluated. After injecting

3PN zygotes with CRISPR-Cas9, Liang and colleagues found

only 4 of 71 (5.6%) embryos contained the desired genetic

change in hemoglobin b gene (HBB) (Liang et al., 2015). The

correctly edited embryos were mosaic and off-target mutations

were common as the CRISPR-Cas9 complex was acting at

two other sites of the genome. Kang and colleagues injected

126 3PN zygotes with Cas9 mRNA, guide RNA (gRNA), and

correction template to modify the immune cell gene CCR5

(Kang et al., 2016). Genetic analysis showed that 2 embryos

were successfully modified with the intended 32 bp deletion.

The use of two gRNAs to delete the same DNA segment without

the need for HDR was successful in 4/26 embryos. Embryos with

the intended genetic change were mosaic. Also using 3PN em-

bryos, Tang et al. found integration of a homologous template

in 2/30 (6.6%) zygotes at the G6PD locus, and 14% (2/14)

showed homologous editing at the HBB locus (Tang et al.,

2017). Using diploid 2PN human embryos fertilized with mutant

sperm, Tang et al. (2017) showed that mutations in HBB and

G6PD could be corrected. All told, however, the study was

limited by the low number of embryos all of which proved to be

mosaic.

Another study targeting MYBPC3, a mutation of which causes

hypertrophic cardiomyopathy, also reported a low efficiency of

template integration through HDR in 2PN embryos; only a single

embryo out of 58 showed template integration in some of the

cells (Ma et al., 2017). As the sperm donor used was heterozy-

gous, an estimated 29 embryos were generated with mutant

sperm, resulting in an HDR efficiency using the template of �3%.
For HDR to be therapeutically relevant for the precise repair of

disease-causing mutations in the human embryos, the efficiency

of editing will need to be increased. Low efficiency of HDR may

be due to the lack of control over when DSBs occur and in which

cell-cycle phase. HDR is more active in the S and G2 phases than

in G1, though this assertion has not been directly tested in hu-

man embryos. Furthermore, the timing of the cell-cycle phases

and of the kinetics of Cas9 cleavage in the human embryo

have not yet been determined. Timing of microinjection at the

G2 phase of the cell cycle has proved useful in promoting HDR

in the mouse embryo (Gu et al., 2018). According to Gu et al.

(2018), the efficiency of targeted integration could be further

increased to 95% by tethering a biotinylated DNA repair tem-

plate to Cas9, which was modified by fusion to monomeric

avidin. This approach has not yet been tested in human embryos

but appears promising. In embryonic stem cells as well as

somatic cells, the efficiency of homologous recombination can

be increased �2-fold through interference with the function of
53BP1 (Nambiar et al., 2019; Canny et al., 2018). 53BP1 inhibits

DSB repair by HDR and promotes NHEJ (Bunting et al., 2010),

and thus, through its deletion, more DSBs are repaired by

HDR, though the effect is modest. An alternative approach is

to increase the expression of factors involved in HDR. Injection

of mouse zygotes with Rad51, a protein involved in the search

for a homologous repair template, appears to increase HDR ef-

ficiency in mouse embryos (Wilde et al., 2018). However, reliance

on Rad51 could also result in detrimental genetic changes

including translocations due to increased recombination be-

tween homologous sequences in the human genome (Richard-

son et al., 2004). Neither approach has thus far been tested in hu-

man embryos.

Most groups have focused on using HDR to introduce pro-

grammed edits into the human germline genome (Liang et al.,

2015; Kang et al., 2016; Tang et al., 2017; Ma et al.2017). How-

ever, NHEJ repair may also be used to restore or disrupt gene

function. The outcomes are generally novel alleles that have no

precedent in the human population. For instance, recurrent

end-joining events restore the reading frame of a mutation at

the EYS locus but results in alleles with an altered amino acid

sequence relative to the wild type (Zuccaro et al., 2020). The gen-

eration of novel alleles may be very useful in a research context,

such as to study the function of a gene product in early embry-

onic development. This experimental approach has been used

to identify a requirement of POU5F1 for normal blastocyst devel-

opment (Fogarty et al., 2017). However, in the context of repro-

duction, functional testing of a novel allele in a human being is

not possible, or acceptable, and therefore the effect on the

health of a person cannot be known. A recent report by the Royal

Society and the National Academies of Sciences, Engineering,

and Medicine specifically called for only the introduction of com-

mon variants in the relevant population to correct a mutation

(NAS, National Academy of Medicine, National Academy of Sci-

ences and the Royal Society, 2020). Thus, HDR is preferable to

NHEJ as a path to germline gene correction. However, low effi-

ciency of HDR was found in all studies; in aggregate, less than

10% of cells showed the intended modification. The formation

of indels predominates about 10-fold (Figure 2).

ADVERSE CONSEQUENCES OF DNA BREAKS IN HUMAN
EMBRYOS

In addition to small indels and precise repair by homologous

recombination, Cas9-induced DSBs can also result in more

extensive genetic changes. In mouse embryos, Cas9 gave rise

to deletions of several hundred base pairs in almost half of the

embryos (Adikusuma et al., 2018). Such deletions are also com-

mon in mouse embryonic stem cells (Kosicki et al., 2018). Kosicki

and colleagues found that more than 20% of the targeted alleles

in mouse embryonic stem cells contained large deletions

>250 bp extending up to 6 kb away from the CRISPR cut site.

Mutations also included complex genomic rearrangements at

the targeted sites. In contrast, Ma and colleagues noted a lack

of large deletions by performing long-range PCR and SNP anal-

ysis in human embryos (Ma et al., 2018). Large deletions were

Cell 184, March 18, 2021 1563

Figure 2. Efficiencies of genome-editing out-

comes in human embryos
Shown are human embryo genome editing outcomes
with the stage of Cas9 injection specified. Genes that
are not represented in all categories were either not
studied for this aspect, or the number of embryos
examined was deemed too low (2 or less). The
endogenous repair template for HBB is the HBD locus,
and the homologous chromosome is the repair tem-
plate for EYS. *Number of embryos analyzed with 8 or
more cells, from Figure 3a of Fogarty et al., 2017.
2-cell injections display the number of blastomeres for
HDR, indels, and off-target activity instead of the
number of embryos. Reference for HBB in 3PN
embryos (Liang et al., 2015), CCR5 (Kang et al., 2016),
G6PD and HBB in 2PN embryos (Tang et al., 2017),
MYBPC3 (Ma et al., 2017), POU5F1 (Alanis-Lobato et
al., 2020; Fogarty et al., 2017), and EYS (Zuccaro et al.,
2020). In Ma et al., the numbers of mutant embryos are
inferred based on the frequency of mutant sperm
from a heterozygous donor. This study also reported
efficient HDR repair with the maternal chromosome
as a template based on the absence of a disease-
causing mutation at MYBPC3 (33% in zygotes [9/27]
and 45% in MII oocytes [13/29]). The genetic nature
of these embryos is not fully understood, as
several outcomes, including chromosome loss,
mitotic recombination, or more complex genetic
change, could result in the absence of a detectable
mutation. HDR, homology directed repair; 2PN, two
pronuclear; 3PN, tripronuclear.

ll

1564 Cell 184, March 18, 2021

Review

ll
Review

similarly missing in another study targeting the EYS locus (Zuc-

caro et al., 2020). Fogarty and colleagues found deletions of

up to �28 bp (Fogarty et al., 2017). Though this observation
does not exclude the possibility of large deletions, human em-

bryos do not appear to incur deletions of several hundred base

pairs at the frequency seen in mice, thereby pointing to spe-

cies-specific differences in DSB repair.

Surprisingly, Cas9 resulted in frequent chromosomal changes

in human embryos. Zuccaro and colleagues targeted a blind-

ness-causing mutation in the EYS gene with CRISPR-Cas9

(Zuccaro et al., 2020). DNA breaks in approximately half of the

embryos injected remained unrepaired, resulting in the loss of

a chromosome arm or a whole chromosome. Similarly, Fogarty

et al. (2017) used CRISPR-Cas9 to create mutations in the

POU5F1 gene for embryo developmental studies and also found

chromosomal changes (Alanis-Lobato et al., 2020). The multiple

possible outcomes associated with the generation of a DSB pre-

sent daunting challenges. Chromosomal changes as a conse-

quence of Cas9 cleavage can also occur in human differentiated

cells, though at about 10-fold lower frequency than in human

embryos (Leibowitz et al., 2020), pointing to differences in DNA

repair in embryos and somatic cells. A large body of work

will be needed to better understand the endogenous embryo

repair machinery and increase the frequency of desired repair

outcomes.

A primary limitation of all gene-editing approaches is the ability

to prevent mosaicism. By restricting the activity of gene editors

to the first cell cycle, and potentially in advance of the replication

of the targeted locus, mosaicism could potentially be avoided.

The discovery of anti-CRISPR proteins (Pawluk et al., 2018)

may well enable such temporal control through timed injection

of the anti-CRISPR protein relative to Cas9. Interestingly, Cas9

activity at off-target sites appears to occur with a delay relative

to the on-target site in human embryos. While the majority of

on-target sites were modified within the first cell cycle of embryo

development, most off-target genetic change occurred in later

cell cycles and were mosaic (Zuccaro et al., 2020). In somatic

cells, delayed addition of the anti-Crispr protein AcrIIA4 inhibits

off-target cleavage while still allowing on target activity (Shin

et al., 2017). Anti-Crispr proteins have promise to reduce on-

target and off-target mosaicism but have not as yet been tested

in human embryos.

BASE AND PRIME EDITING IN HUMAN EMBRYOS

Recently developed editing systems such as base and prime ed-

iting do not use DSBs and thus may avoid some of the aforemen-

tioned undesirable repair outcomes. In base editing, a catalyti-

cally inactive Cas9 serves to guide a deaminase to a specific

site in the genome to mediate site-specific nucleotide to nucleo-

tide conversions (Komor et al., 2016). Base editing has been

tested in human embryos (Zhou et al., 2017; Zhang et al.,

2019; Li et al., 2017;, Zeng et al., 2018) and was found to be high-

ly efficient: editing efficiency was higher than 50% in most cases.

Indels occurred at a lower frequency than after Cas9 cleavage

and may be due to a response of the endogenous repair mech-

anisms to the deaminated base. Studies also reported mosai-

cism and base changes at sites flanking the targeted base.

Base editors with a more limited editing window and less

bystander effects have been developed (Kim et al., 2017) but

have not as of yet been tested in human embryos.

While more efficient than HDR, base editing can only correct

four out of 12 nucleotide substitutions and cannot repair genetic

changes such as indels. About 60% of disease-causing mutations

may be corrected using base editing (Rees and Liu, 2018). In 2019,

Anzalone and colleagues described the prime editing method to

overcome these limitations. Prime editing uses a Cas9 nickase

for the recruitment of a reverse transcriptase to the target site

and the introduction of a break in just one of the two DNA strands.

The attendant gRNA is modified to include an RNA template for

the repair of the mutation by copying it through reverse transcrip-

tion into the single-stranded nick (Anzalone et al., 2019). Prime ed-

iting also allows for the repair of a much wider spectrum of

mutations without the need for a donor DNA template or the gen-

eration of a DSB. Using prime editing to correct the genes respon-

sible for sickle cell disease and Tay Sachs, Liu’s group reported a

higher or similar efficiency and lower off-target editing compared

to CRISPR-triggered HDR (Anzalone et al., 2019). While the key

steps of gene editing are performed by exogenously engineered

enzymes, the endogenous DNA repair machinery is still required

to seal the nick induced in the DNA. Thus, the outcomes of prime

editing may be cell type and species dependent. It is therefore

important to study the consequences of this approach directly

in the relevant cell type. In mouse embryos, intended edits could

be introduced in 10%–50% of the embryos at different loci (Aida

et al., 2020; Liu et al., 2020). However, in addition to the intended

genetic change, indels were also observed in up to 60% of the

embryos, which proved mosaic for both intended and unintended

genetic change. Prime editing has yet to be tested in human em-

bryos, and there is insufficient understanding of endogenous DNA

repair pathways in the human embryo to anticipate its associated

outcomes.

MITOCHONDRIAL REPLACEMENT THERAPY

In addition to nuclear DNA mutations, heritable genetic disease

can also be caused by mutations in mitochondrial DNA. Mito-

chondrial disorders are among the most common inherited

metabolic diseases and can be debilitating or fatal at an early

age. Given the lack of effective pharmacologic agents for the

treatment of mitochondrial DNA disorders, current treatment is

largely supportive. Forms of prevention have focused on preim-

plantation genetic diagnosis or mitochondrial replacement.

Mitochondrial replacement entails the replacement of the

mutated mitochondrial DNA of the oocyte with a healthy mito-

chondrial genome from a donated oocyte (Herbert and Turnbull,

2018). Nuclear content of a maternal oocyte is transferred to a

normal ‘‘enucleated’ oocyte from a donor female prior to implan-

tation. The offspring will have all the nuclear genetic components

of the parents but mitochondrial DNA from a female donor. Mito-

chondrial replacement therapy has been approved for use in the

United Kingdom strictly to prevent genetic disease, and a clinical

trial is ongoing. In 2016, mitochondrial replacement therapy was

used to avoid transmitting the hereditary disease, Leigh syn-

drome, resulting in a child with predominantly normal mitochon-

drial DNA (Zhang et al., 2017).

Cell 184, March 18, 2021 1565

ll
Review

Within an oocyte, thousands of mitochondrial DNA molecules

exist. Mutant and wild-type mitochondrial DNA may co-exist and

only result in disease if the mutant mitochondrial DNA exceeds a

certain threshold. The specific cleavage of mutant mitochondria

through TALENs to reduce the percentage of mutant mitochon-

drial DNA may be an alternative approach to treating mitochon-

drial disease (Reddy et al., 2015). Only recently has it become

possible to edit the mitochondrial genome using TALEN nucle-

ases fused to a deaminase that can act on double-stranded

DNA (Mok et al., 2020). This latest base editor adds yet another

possible tool to the prevention of mitochondrial DNA disease in

the context of human reproduction. Neither approach has been

tested in human embryos.

An important distinction between mitochondrial replacement

and the use of TALENs for cleavage or editing is that mitochon-

drial replacement does not involve any direct change to the

genome itself. Rather, it is a manipulation to alter the pattern of

mitochondrial DNA inheritance and provides no path to intro-

ducing genetic variants that do not already exist in the human

population. Thus, some of the concerns relating to heritable

genome editing do not apply to this technology.

EDITING OF IN VITRO-DERIVED GAMETES AND
EMBRYOS

Spermatogonial stem cells
Cultured cells provide several practical advantages for gene ed-

iting over embryos. They allow for a larger number of modifica-

tions, a comprehensive genetic analysis prior to generating an

embryo, and the ability to avoid mosaicism. Several different

cell types have reproductive potential. In men, germline cells

continue to proliferate throughout adulthood as spermatogonial

stem cells and may provide a path to generating edited mature

sperm. Wu et al. (2015) reported the editing of mouse spermato-

gonial stem cells followed by testicular transplantation, resulting

in the repair of a cataract-causing mutation. Fertilization using

spermatids derived from these edited spermatogonial stem cells

gave rise to offspring with the corrected phenotype at 100% ef-

ficiency. Gene editing of spermatogonial stem cells, perhaps

even in vivo, might provide a path to efficient gene editing of

the paternal genome while also avoiding mosaicism. This

approach could, for instance, be used to restore spermatogen-

esis due to mutations in genes required for sperm maturation

and prevent offspring from inheriting these same mutations.

Induced pluripotent stem cells
In vitro-derived gametes induced from pluripotent cells are also

an active area of research. Pluripotent stem cells are frequently

used as a target for genome editing in the context of human dis-

ease modeling. For example, Schwank et al. (2013) used

CRISPR-Cas9 to successfully correct cystic fibrosis mutations

in induced pluripotent stem cells derived from cystic fibrosis pa-

tients. Pluripotent cells can give rise to all cell types of the body

including germ cells. When induced from somatic cells, they are

diploid and need to undergo meiosis for use in reproduction.

Hayashi et al. (2011) reported mouse primordial germ cell-like

cells derived from male pluripotent stem cells that have been

transplanted into the seminiferous tubules of germ cell-ablated

1566 Cell 184, March 18, 2021

mice and yielded functional sperm. Hayashi also showed mouse

primordial germ cell-like cells derived from female embryonic

stem cells and induced pluripotent stem cells developed into

fully grown oocytes that contributed to healthy offspring (Haya-

shi et al., 2012). More recently, Hayashi’s laboratory identified

transcription factors that allow conversion of mouse pluripotent

stem cells to oocyte-like cells that proved fertilization compe-

tent, although they failed to undergo normal meiosis and give

rise to embryos (Hamazaki et al., 2021). Though primordial

germ cell-like cells have been made from human pluripotent

stem cells (Yamashiro et al., 2018), no mature gametes have

been reported to date.

Haploid human stem cells
Further progress toward reproduction has been made in haploid

human stem cells (Figure 3). Haploid human pluripotent stem

cells are derived from either parthenogenetically activated

oocytes that develop without fertilization (Sagi et al., 2016) or

from sperm injected into enucleated oocytes (Zhang et al.,

2020). Haploid pluripotent stem cells have the chromosomal

equivalent of a gamete, containing only 23 chromosomes.

Remarkably, haploid stem cells derived from sperm can act

like sperm fertilize a human oocyte and allow development to

the blastocyst stage (Zhang et al., 2020). Imprinting patterns of

these embryos are indistinguishable from IVF controls, and

gene-expression patterns were very similar, though a small num-

ber of genes of unknown significance were differentially ex-

pressed. Developmental efficiency to the blastocyst stage was

lower than in ICSI control embryos; of 130 oocytes injected

with paternal haploid cells, only 5 formed euploid blastocysts.

Pregnancy was not attempted with these 5 euploid blastocysts.

Haploid cells can be clonally expanded thereby allowing

extensive genetic modification, selection, and detailed genetic

and epigenetic analysis. Editing in haploid stem cells is efficient

(Safier et al., 2020), and their genomes have been modified at

scale in the context of genetic screens (Yilmaz et al., 2020).

Sequential editing or multiplexing may allow one to eliminate

damaging variants from the human genome. The estimated

number of mutations disrupting protein coding genes in the

human genome is �100 (MacArthur et al., 2012). However, there
is not yet sufficient understanding of the health impact of these

mutations and whether or not they might contribute to normal

phenotypic variation. Such information may ultimately be gained

through a combination of genome sequencing, and molecular

and functional studies in cellular models of human disease as

well as in animal models. These efforts are ongoing worldwide

to understand mechanisms of human disease for the develop-

ment of adult therapies. Though the motivation is different and

independent of germline genome therapy, these efforts will

also inform which and how many variants might be meaningful

to target in the germline.

While the correction of numerous disease-causing variants in

cultured cells may be an attractive concept, routine culture will

also introduce novel mutations as well as epigenetic changes,

which could affect the health of the resulting embryo. By some

measurements, mutation rates in human pluripotent stem cells

are about 3–4 base substitutions per genome and population

doubling, or within about a day of culture (Kuijk et al., 2020).

Figure 3. Editing haploid human pluripotent stem cells
Shown is a proposed pathway for reproduction with genome-edited paternal haploid stem cells: (1) The maternal genome is removed from the oocyte followed by
sperm injection. The resultant haploid embryo is cultured to the blastocyst stage where the inner cell mass is used to generate a haploid pluripotent embryonic
stem cell (ESC) line. Only X sperm give rise to ESCs as monosomy Y is lethal. (2) One or multiple edits can be performed with CRISPR/Cas9 or prime editing in
ESCs followed by clonal expansion. Whole genome sequencing can identify genetic mutations including novel, culture-induced genetic, and/or epigenetic
changes. (3) Haploid cells spontaneously convert to diploid cells at a rate of 1–5% per day, requiring selection of haploid cells through sorting for low (haploid)
DNA content (Sagi et al., 2016). (4) Instead of fertilization by sperm, a single haploid ESC with an edited paternal genome is injected into a nucleated oocyte.
Because ESCs contain mitochondrial DNA from the oocyte donor used for derivation, a match of mitochondria DNA genotypes might be needed to ensure
stability of the mitochondria DNA genotype. (5) The resulting diploid embryo is cultured and biopsied for genome sequencing and epigenetic characterization.

(legend continued on next page)

ll

Cell 184, March 18, 2021 1567

Review

ll
Review

Culture-induced genetic or epigenetic change will be clonally in-

herited and remain of unknown functional significance until a hu-

man being is made. Compared to the use of sperm and oocytes,

reproduction from cultured pluripotent stem cells adds a

sequence of cellular manipulations with high risks. This is partic-

ularly true for induced pluripotent stem cells derived from so-

matic cells, which contain mutations of somatic origin as well

as from in vitro reprogramming (Gore et al., 2011; Bhutani

et al., 2016; Rouhani et al., 2016; Yoshihara et al., 2017). Thus,

this approach, in most instances, will be inconsistent with the

above-stated goal of avoiding the introduction of novel alleles.

Further considerations under which circumstances such risk

might be justified will need to be made.

ETHICAL CONSIDERATIONS OF HERITABLE HUMAN
GENOME EDITING

With the introduction of efficient genome editing tools such as

CRISPR-Cas9, the plausibility of safely editing the genome of

the human germline is currently the subject of many academic,

industry, and policy discussions. In contrast to gene editing in

somatic cells, gene editing in human gametes or embryos to

permanently modify the germline raises significant ethical con-

cerns. Shortly before the 2018 International Human Genome Ed-

iting Summit, reports emerged that two girls were born after

germline genome editing to prevent the expression of the HIV re-

ceptor CCR5 (Cyranoski and Ledford, 2018). This announce-

ment was met with strong statements condemning the practice

in general, the lack of a sound medical basis, the lack of safety

assessments, the inadequacy of the informed-consent docu-

ments signed by the prospective parents, and the lack of public

discussion and input regarding the personal and societal conse-

quences of HHGE (Savulescu and Singer, 2019).

Many called for a moratorium on clinical HHGE (Lander et al.,

2019) and the ethical debate on HHGE continues to intensify.

Because it is associated with human reproduction, HHGE often

evokes spiritual, religious, or deeply personal issues for many.

The following ethical discussion is not meant to be exhaustive

but instead focuses on key issues surrounding HHGE as it relates

to the four core bioethical principles: beneficence, non-malefi-

cence, autonomy, and justice (Figure 4).

Beneficence: Therapeutic benefits
With over 10,000 known monogenic diseases, heritable diseases

collectively affect roughly 5%–7% of the human population (Ko-

fler and Kraschel, 2018). Millions more are affected by genetic

variants that increase disease risk to common disorders such

as diabetes, obesity, or cardiovascular disease. The correction

of mutations in the germline would allow patients to create em-

bryos free of disease-causing mutations. The disease gene

would no longer be passed on to subsequent generations and

over multiple generations. This alone could reduce disease prev-

Maternal and paternal genomes in the embryo result in a diploid, 46 XX karyotyp
attempted. For use of a maternal haploid ESC, its edited genome would replace th
and male offspring (not illustrated). Below: tools for genome editing. Cas9 induces
or through end joining. Cleavage between regions of microhomology (red nucleo
base editing (not illustrated), involve a single strand DNA cut.

1568 Cell 184, March 18, 2021

alence and even eliminate selected heritable diseases. Some

argue that the medical need for HHGE is so compelling that pro-

ceeding with this use of genome editing is a moral imperative

(Gyngell et al., 2019) and that doing so would help to ‘‘lighten

the burden of human existence’’ (Harris, 2016).

Some may argue that IVF with preimplantation genetic testing

for monogenic disease (PGT-M) already allows couples to have

genetically related children without the risk of inheriting a known

familial disorder (NAS, National Academy of Medicine, National

Academy of Sciences and the Royal Society, 2020). Patients

with a genetic disorder can use PGT-M to screen IVF embryos

for the disease-causing gene of interest and transfer only dis-

ease-free embryos. Few scenarios exist in which couples would

not be eligible for PGT-M and would necessitate the use of

HHGE. For instance, if a parent is homozygous for an autosomal

dominant disorder, every embryo will inherit at least one copy of

the causative allele and will thus be affected. If both parents are

homozygous for a recessive disorder such as cystic fibrosis, all

embryos will be homozygous. Given the rarity of these scenarios,

analysis of prevalence data of common genetic disorders sug-

gests that the clinical need for HHGE for cases that are not

amenable to PGT-M is exceedingly small (Viotti et al., 2019).

An analysis by Viotti et al. (2019) estimated that if all of the pa-

tients who are ineligible for PGT-M opted for HHGE, HHGE

would benefit at most 100 births per year in the United States.

Preimplantation genetic testing, however, is not equally effec-

tive for all couples. Even without PGT-M, a limited number of oo-

cytes retrieved during IVF will fertilize and result in an embryo for

transfer. IVF success rates decline significantly with age with

only an estimated 12% of oocytes retrieved result in live birth

(De Rycke et al., 2017). The exclusion of embryos due to a muta-

tion can result in no embryos available for transfer. One study re-

ported 39.8% of cycles performed with PGT-M and aneuploidy

resulted in no transferable embryos (Minasi et al., 2017). HHGE

could improve the efficiency of PGT-M by increasing the number

of transferrable embryos, thus decreasing the need for multiple

IVF cycles with its associated physical risks and costs. Based

on the percentage of cycles with no transferable embryos due

to a genetic mutation, we estimate that �3,000 IVF cycles annu-
ally would benefit from this approach in the US alone (Viotti et al.,

2019; Minasi et al., 2017).

Another common argument against HHGE is that somatic gene

therapyordrugsprovideasaferpathtotherapywithoutriskstothe

germline (Lanphier et al., 2015). While somatic gene therapy trials

are encouraging, limitations to effective therapy include an im-

munebarrierand treatmentafterirreversible damagefromdisease

onset has occurred. In utero gene therapy may provide a window

of opportunity for effective treatment due to small fetal size, tolero-

genic fetal immune system, accessible stem and/or progenitor

cells, permeable blood-brain barrier, and potential to treat before

disease onset, critical for diseases with high prenatal or perinatal

morbidity and mortality (Palanki et al., 2021). The clinical utility of

e. The last step, to establish a pregnancy for the birth of a child, has not been
e genome of the oocyte, and would be fertilized by sperm, allowing both female
a double-stranded break, which can be repaired with a homologous template

tides) can result in predictable outcomes. In contrast, prime editing, as well as

Figure 4. Ethical considerations of heritable

human genome editing
The four principles of biomedical ethics (beneficence,
non-maleficence, autonomy, and justice) are used as
framework to describe the ethical concerns sur-
rounding human heritable genome editing. (A) Benef-
icence: Genome editing has the potential to treat
or even eradicate heritable disease. (B) Autonomy:
Parents have right to make their own reproductive
decisions which will affect offspring and subsequent
generations. (C) Non-maleficence: Unintended out-
comes of CRISPR/Cas9 can have dire consequences.
Some worry germline editing is a slippery slope that
can lead to ‘‘designer babies’’ and affect genetic and
phenotypic diversity in human reproduction. (D) Jus-
tice: Access to assisted reproductive technologies is
limited to individuals and countries who can afford it
(figured adapted from (DeWeerdt, 2020).

ll

Cell 184, March 18, 2021 1569

Review

ll
Review

in utero gene therapy has so far been limited by poor efficacy and

safety concerns, including that a therapy may rescue a pregnancy

but still result in a child with disease. Effective somatic or in utero

gene therapiesmayinfactincrease the needforgermlineinterven-

tionascoupleswithhomozygousmutationswillbemorecommon.

For instance, patients with cystic fibrosis now live long enough to

raise a family and may wish toavoid passing on the disease totheir

children. Hence, effective somatic gene therapy does not obviate

gene therapy of the germline. A patient who is a carrier for a dis-

ease-causing mutation may also wish to avoid passing on the mu-

tation to help prevent the risk of disease in subsequent genera-

tions. This could expand the utility of HHGE to millions of couples.

Embryos with chromosomal aneuploidy are more commonly

encountered than embryos with homozygous disease-causing

mutations. The number of IVF-generated aneuploid embryos in-

creases exponentially with increasing maternal age, reaching

85% of all embryos by the age of 43 (Franasiak et al., 2014).

Intriguing new data demonstrated the possibility of targeting and

deleting an entire chromosome in both mouse and human em-

bryos using Cas9 (Adikusuma et al., 2017; Zuo et al., 2017; Zuc-

caro et al., 2020). This potential use of Cas9 will still require exten-

sive basic research to investigate the mechanisms of

chromosome loss and the risk of adverse outcomes, including

retention of chromosome segments that could cause develop-

mental issues. Viotti et al. (2019) estimated that 20% of cycles pro-

duce only aneuploid embryos, which in the United States alone

amounts to �17,600 cycles. Approximately 5% of human oocytes
contain single-chromosome gains (McCoy et al., 2015); these oo-

cytes may be the most amenable to correction of chromosomal

content.

Patients with a Robertsonian chromosome translocation could

also benefit from this technology. Robertsonian translocations

are the most common form of chromosomal translocations in hu-

mans, identified in approximately 1 in 1,000 individuals (Hamer-

ton et al., 1975). It occurs when the long q arms of two acrocen-

tric chromosomes merge by translocation, and their short p arms

are lost. Because the lost short p arms do not contain unique ge-

netic sequences, these individuals are typically phenotypically

normal but are at increased risk for recurrent pregnancy loss

due to embryonic aneuploidy. Individuals with a Robertsonian

21q;21q translocation, in particular, may benefit from removing

a chromosome since essentially all resulting offspring have tri-

somy 21, which carries an 85% risk of pregnancy loss (Ercis

and Balci, 1999). Approximately 2% of individuals with trisomy

21 are due to a Robertsonian 21q;21q translocation (Mutton

et al., 1996). While years of extensive preclinical testing is still

needed to determine feasibility and safety, both gene editing

and correction of aneuploidies have the potential to benefit the

health of the embryo and the resulting child.

Non-maleficence: Risks and safety
Rigorous affirmation of the scientific proof of concept and the

copious preclinical evidence to establish the knowledge of risks

and benefits is essential prior to clinical translation. Genome ed-

iting carries risks beyond those incurred by natural reproduction

or IVF alone. Mosaicism and off-target effects are major con-

cerns. Genetic engineering of an embryo or gamete without a

mutation would mean to expose it to risk of off-target effects

1570 Cell 184, March 18, 2021

without any benefits to the resulting child. Accordingly, the Royal

Society and the National Academies of Sciences, Engineering,

and Medicine limit the use of genome editing to only affected

embryos with known pathogenic variants (NAS, National Acad-

emy of Medicine, National Academy of Sciences and the Royal

Society, 2020). Identifying which embryos to target for genetic

modification is a challenge, in particular at the earliest stages

when genome editing can be performed on one copy to avoid

mosaicism. In studies involving the fertilization of oocytes with

sperm from a donor with a heterozygous mutation, all embryos,

including wild-type embryos, were injected with Cas9 (Ma et al.,

2017; Tang et al., 2017) or with a base editor (Zeng et al., 2018;

Liang et al., 2017). For oocytes, though not for sperm, it is

possible to infer the genetic content through analysis of the polar

bodies. Thus, HHGE in the embryo may be most applicable to

the female germline and to homozygous mutations in the male

germline. While stem cell-derived gametes would allow for

extensive genetic analysis prior to embryo generation, in vitro-

derived gametes also add enormous new risks. In vitro gameto-

genesis, in particular, from induced pluripotent stem cells com-

bines an extensive sequence of cellular manipulations. The

associated risks are numerous from incomplete reprogramming

to genetic and epigenetic changes.

Non-maleficence: Exploitation for non-therapeutic
modification
Some are concerned that HHGE may lead to a ‘‘slippery slope’’

going beyond disease prevention to select for desirable traits.

This has been referred to as enhancement or ‘‘designer babies.’’

While some argue that this can be controlled through policy and

regulation, others worry that perceived enhancement technolo-

gies will be used in other countries without sufficient regulations

or oversight (Ethics Committee of the American Society for

Reproductive Medicine, 2020). Beyond the interests of parents

and the health of a child, genetic diversity is the principal asset

of human reproduction. It is the basis for phenotypic diversity

and thereby contributes to the formation of a highly complex so-

ciety. This diversity could be compromised if pressures experi-

enced by parents are projected to the genotypes of their chil-

dren. It is also difficult to know which, among the innumerable

normal variants, prepares their children best for the future.

Autonomy: Reproductive rights and lack of ability to
consent before birth
Given alternative paths to parenthood including adoption or

gamete donation, the desire for genetic relatedness will be

weighed against the risks of HHGE. Risks and benefits of exist-

ing and novel reproductive treatments impact both parents and

child with an important distinction; the prospective parents can

evaluate risks and benefits and consent to them, while the hu-

man being to be created cannot. Still others assert parents

make countless decisions that shape their children’s future

and that the parental desire to enhance the health and happiness

of their children is an existing and indeed admirable aspect of

parenthood, which often begins prior to conception and con-

tinues throughout the child’s life (Ethics Committee of the Amer-

ican Society for Reproductive Medicine, 2020).

ll
Review

Justice: Equitable access and allocation of resources
Additional concerns within the debate on the ethics of HHGE

relate to inequality. As with many reproductive technologies,

HHGE may only be accessible to individuals and countries

who can afford it. This reality may increase health disparities

among socioeconomic classes (Nuffield Council on Bioethics,

2018). Others argue that HHGE could instead balance the in-

equalities brought on by genetic diseases and variants (Gyngell

et al., 2019; De Wert et al., 2018). Data on human genetic diver-

sity and the role of gene variants under different genomic and

external environments are fundamental to gene editing. This

knowledge is dependent on genetic data from diverse ethnic

backgrounds and environments. Some argue that the current

genetic repositories are not representative of the global popula-

tion (Cavaliere, 2019). Costs and allocation of resources is also a

consideration. For reproductive treatments, the expenses are

generated at the beginning; the costs of managing chronic med-

ical conditions, however, may be significantly greater in compar-

ison due to repeated hospitalization, testing, and treatment.

Embryo research and destruction
Preclinical evidence of efficacy and safety of HHGE requires a

large body of research. Preclinical research on genome editing

in human embryos is incompatible with their use in reproduction,

resulting in their destruction. For some, such outcome is ethically

unacceptable on the grounds that embryos should be granted

the full rights of a living person. For some patients, IVF and the

discarding of embryos also pose moral and ethical dilemmas.

These concerns can be further augmented after preimplantation

testing reveals one or more of the embryos in question contain a

disease-causing mutation. Theoretically, HHGE could repair and

salvage the diseased embryos that otherwise would be dis-

carded. However, while beneficial for some embryos, HHGE is

unlikely to result in the transfer of every embryo created in an

IVF cycle. Technologies such as HHGE and PGT-M could also

play a role in avoiding pregnancy termination based on the dis-

covery of a genetic anomaly through prenatal testing (Ethics

Committee of the American Society for Reproductive Medicine,

2018). For some, the discarding of embryos is more acceptable

than prenatal diagnosis followed by abortion (Cameron and Wil-

liamson, 2003).

REGULATION

HHGE is largely forbidden globally by laws and regulations. A

2020 policy survey found that the majority of countries (96 of

106) surveyed have policy documents—legislation, regulations,

guidelines, codes, and international treaties—relevant to the

use of HHGE (Baylis et al., 2020). No country explicitly permits

the use of genetically modified embryos for reproductive intent.

Five countries (Colombia, Panama, Belgium, Italy, and United

Arab Emirates) would allow potential exceptions for HHGE and

therapeutic purposes. For instance, Colombia allows for HHGE

if ‘‘aimed at relieving suffering or improving the health of the per-

son and humanity’’ (Baylis et al., 2020).

National policies surrounding germline editing for research

purposes without reproductive intent are much more mixed.

Most surveyed countries do not have specific regulation, either

permissive or prohibitive. Seventeen countries, including Can-

ada, Germany, Brazil, and Switzerland, do not permit human

germline genome research. Alternatively, 12 countries, including

the United States, the United Kingdom, Japan, China, Sweden,

Ireland, Norway, Thailand, Iran, Congo, Burundi, and India,

permit research on human germline genome editing for research

without reproductive intent (Baylis et al., 2020).

In the United States, scientific research into the editing of the

genome of the human embryo without transfer for reproductive

purposes remains permissible although ineligible for public fund-

ing. The Food and Drug Administration (FDA) is also prohibited

from considering applications for clinical trials ‘‘in which a human

embryo is intentionally created or modified to include heritable

genetic modification’’ (Congress.gov, 2015). Of note, no legis-

lator has stated whether the language of the moratorium applies

to editing of oocytes, sperm, or gamete precursors (Cohen et al.,

2020). These regulations may all have similar intent: to prevent

inappropriate or premature use of a technology with transforma-

tive potential. While all nations aim to improve human health,

complete prohibitions limit the potential for translational

research and demonstrate the lack of public confidence in regu-

lation’s ability to distinguish appropriate from inappropriate use,

both of which have yet to be defined. Future policy discussions

should continue to engage the scientific, medical, and public

communities to reflect shared national interests and define

acceptable use.

CONCLUSIONS

Science and imagination possess the freedom to reach beyond

what can be translated into clinical practice for the purpose of

better understanding human biology, reproduction, and ge-

netics. While science continues to rapidly advance, in order to

arrive at decisions with far-reaching implications, the public,

medical, and scientific community should be engaged in mean-

ingful discussions regarding the pursuit and potential of these

powerful reproductive tools.

The United Kingdom’s handling of mitochondrial replacement

therapy may serve as a model in this context (Claiborne et al.,

2016). Years of preclinical basic research preceded its consider-

ation for use in human reproduction. At the same time, the United

Kingdom actively sought the public’s opinion to help inform pol-

icymakers, industry, and the research community. This led to

state-sanctioned clinical trials and could be used as a model

for the ethically defensible and publicly acceptable pursuit of

HHGE. In stark contrast to the birth of the two girls with edited

CCR5 genes in 2018, decisions to move forward with mitochon-

drial replacement were not made by individual scientists or doc-

tors, but by independent regulators and the public. This path

takes more time but will likely have a longer-lasting benefit to pa-

tients. For instance, while a mitochondrial replacement proced-

ure was successfully performed in Mexico in 2017 (Zhang et al.,

2017), apparently based on institutional rather than state-level

oversight, no additional cases have since been reported. Alter-

natively, state-sanctioned mitochondrial donation studies are

ongoing in the United Kingdom. While HHGE has the potential

to transform the field of reproductive medicine, it is the

consensus of many (Plaza Reyes and Lanner, 2017; Rossant,

Cell 184, March 18, 2021 1571

ll
Review

2018; Adashi and Cohen, 2020; Lea and Niakan, 2019) that great

caution must be exercised with an eye on the broader societal

and ethical issues that surround its application.

ACKNOWLEDGMENTS

This work was supported by the NYSTEM award #C32564GG to D.E. J.T. is

supported by a clinical fellowship in reproductive endocrinology and infertility.

DECLARATION OF INTERESTS

D.E. is a member of the Cell Editorial Board. E.Y.A. serves as Co-Chair of the

Safety Advisory Board of Ohana Biosciences. J.T. declares no conflicts of

interest.

REFERENCES

Adashi, E.Y., and Cohen, I.G. (2020). Therapeutic Germline Editing: Sense and

Sensibility. Trends Genet. 36, 315–317.

Adikusuma, F., Williams, N., Grutzner, F., Hughes, J., and Thomas, P. (2017).

Targeted Deletion of an Entire Chromosome Using CRISPR/Cas9. Mol. Ther.

25, 1736–1738.

Adikusuma, F., Piltz, S., Corbett, M.A., Turvey, M., McColl, S.R., Helbig, K.J.,

Beard, M.R., Hughes, J., Pomerantz, R.T., and Thomas, P.Q. (2018). Large de-

letions induced by Cas9 cleavage. Nature 560, E8–E9.

Aida, T., Wilde, J.J., Yang, L., Hou, Y., Li, M., Xu, D., Lin, J., Qi, P., Lu, Z., and

Feng, G. (2020). Prime editing primarily induces undesired outcomes in mice.

bioRxiv, 2020.08.06.239723.

Alanis-Lobato, G., Zohren, J., Mccarthy, A., Fogarty, N.M.E., Kubikova, N.,

Hardman, E., Greco, M., Wells, D., Turner, J.M.A., and Niakan, K.K. (2020).

Frequent loss-of-heterozygosity in CRISPR-Cas9-edited early human em-

bryos. bioRxiv, 2020.06.05.135913.

Anzalone, A.V., Randolph, P.B., Davis, J.R., Sousa, A.A., Koblan, L.W., Levy,

J.M., Chen, P.J., Wilson, C., Newby, G.A., Raguram, A., and Liu, D.R.

(2019). Search-and-replace genome editing without double-strand breaks or

donor DNA. Nature 576, 149–157.

Baylis, F., Darnovsky, M., Hasson, K., and Krahn, T.M. (2020). Human Germ

Line and Heritable Genome Editing: The Global Policy Landscape. CRISPR

J. 3, 365–377.

Bhutani, K., Nazor, K.L., Williams, R., Tran, H., Dai, H., D�zakula, �Z., Cho, E.H.,

Pang, A.W.C., Rao, M., Cao, H., et al. (2016). Whole-genome mutational

burden analysis of three pluripotency induction methods. Nat. Commun.

7, 10536.

Bunting, S.F., Callén, E., Wong, N., Chen, H.T., Polato, F., Gunn, A., Bothmer,

A., Feldhahn, N., Fernandez-Capetillo, O., Cao, L., et al. (2010). 53BP1 inhibits

homologous recombination in Brca1-deficient cells by blocking resection of

DNA breaks. Cell 141, 243–254.

Cameron, C., and Williamson, R. (2003). Is there an ethical difference between

preimplantation genetic diagnosis and abortion? J. Med. Ethics 29, 90–92.

Canny, M.D., Moatti, N., Wan, L.C.K., Fradet-Turcotte, A., Krasner, D., Ma-

teos-Gomez, P.A., Zimmermann, M., Orthwein, A., Juang, Y.C., Zhang, W.,

et al. (2018). Inhibition of 53BP1 favors homology-dependent DNA repair

and increases CRISPR-Cas9 genome-editing efficiency. Nat. Biotechnol. 36,

95–102.

Cavaliere, G. (2019). The Ethics of Human Genome Editing. WHO Expert Advi-

sory Committee on Developing Global Standards for Governance and Oversight

of Human Genome Editing, Background Paper. https://www.who.int/ethics/

topics/human-genome-editing/WHO-Commissioned-Ethics-paper-March19.

pdf. (Accessed 25 January 2021).

Claiborne, A.B., English, R.A., and Kahn, J.P. (2016). ETHICS OF NEW TECH-

NOLOGIES. Finding an ethical path forward for mitochondrial replacement.

Science 351, 668–670.

1572 Cell 184, March 18, 2021

Cohen, I.G., Sherkow, J.S., and Adashi, E.Y. (2020). Gene Editing Sperm and

Eggs (not Embryos): Does it Make a Legal or Ethical Difference? J. Law Med.

Ethics 48, 619–621.

Cong, L., Ran, F.A., Cox, D., Lin, S., Barretto, R., Habib, N., Hsu, P.D., Wu, X.,

Jiang, W., Marraffini, L.A., and Zhang, F. (2013). Multiplex genome engineering

using CRISPR/Cas systems. Science 339, 819–823.

Congress.Gov (2015). Public Law 114-92. Consolidated Appropriations Act,

2016. Sec 749. December 18, 2015. https://www.congress.gov/bill/

114th-congress/house-bull/2029/text. (Accessed 25 January 2021).

Cyranoski, D., and Ledford, H. (2018). Genome-edited baby claim provokes in-

ternational outcry. Nature 563, 607–608.

De Rycke, M., Goossens, V., Kokkali, G., Meijer-Hoogeveen, M., Coonen, E.,

and Moutou, C. (2017). ESHRE PGD Consortium data collection XIV-XV: cy-

cles from January 2011 to December 2012 with pregnancy follow-up to

October 2013. Hum. Reprod. 32, 1974–1994.

De Wert, G., Heindryckx, B., Pennings, G., Clarke, A., Eichenlaub-Ritter, U.,

van El, C.G., Forzano, F., Goddijn, M., Howard, H.C., Radojkovic, D., et al.; Eu-

ropean Society of Human Genetics and the European Society of Human

Reproduction and Embryology (2018). Responsible innovation in human germ-

line gene editing: Background document to the recommendations of ESHG

and ESHRE. Eur. J. Hum. Genet. 26, 450–470.

DeWeerdt, S. (2020). How much should having a baby cost? Nature 588,

S174–S176.

Ercis, M., and Balci, S. (1999). Can a parent with balanced Robertsonian trans-

location t(21q;21q) have a non-Down’s offspring? Lancet 353, 751.

Ethics Committee of the American Society for Reproductive Medicine. (2018).

Use of preimplantation genetic testing for monogenic defects (PGT-M) for

adult-onset conditions: an Ethics Committee opinion. Fertil. Steril. 109,

989–992.

Ethics Committee of the American Society for Reproductive Medicine (2020).

Ethics in embryo research: a position statement by the ASRM Ethics in Embryo

Research Task Force and the ASRM Ethics Committee. Fertil. Steril. 113,

270–294.

Fogarty, N.M.E., McCarthy, A., Snijders, K.E., Powell, B.E., Kubikova, N., Bla-

keley, P., Lea, R., Elder, K., Wamaitha, S.E., Kim, D., et al. (2017). Genome ed-

iting reveals a role for OCT4 in human embryogenesis. Nature 550, 67–73.

Franasiak, J.M., Forman, E.J., Hong, K.H., Werner, M.D., Upham, K.M., Treff,

N.R., and Scott, R.T. (2014). Aneuploidy across individual chromosomes at the

embryonic level in trophectoderm biopsies: changes with patient age and

chromosome structure. J. Assist. Reprod. Genet. 31, 1501–1509.

Gasiunas, G., Barrangou, R., Horvath, P., and Siksnys, V. (2012). Cas9-crRNA

ribonucleoprotein complex mediates specific DNA cleavage for adaptive im-

munity in bacteria. Proc. Natl. Acad. Sci. USA 109, E2579–E2586.

Gore, A., Li, Z., Fung, H.L., Young, J.E., Agarwal, S., Antosiewicz-Bourget, J.,

Canto, I., Giorgetti, A., Israel, M.A., Kiskinis, E., et al. (2011). Somatic coding

mutations in human induced pluripotent stem cells. Nature 471, 63–67.

Gu, B., Posfai, E., and Rossant, J. (2018). Efficient generation of targeted large

insertions by microinjection into two-cell-stage mouse embryos. Nat. Bio-

technol. 36, 632–637.

Gyngell, C., Bowman-Smart, H., and Savulescu, J. (2019). Moral reasons to

edit the human genome: picking up from the Nuffield report. J. Med. Ethics

45, 514–523.

Hamazaki, N., Kyogoku, H., Araki, H., Miura, F., Horikawa, C., Hamada, N.,

Shimamoto, S., Hikabe, O., Nakashima, K., Kitajima, T.S., et al. (2021). Recon-

stitution of the oocyte transcriptional network with transcription factors. Nature

589, 264–269.

Hamerton, J.L., Canning, N., Ray, M., and Smith, S. (1975). A cytogenetic sur-

vey of 14,069 newborn infants. I. Incidence of chromosome abnormalities.

Clin. Genet. 8, 223–243.

Harris, J. (2016). Germline Modification and the Burden of Human Existence.

Camb. Q. Healthc. Ethics 25, 6–18.

ll
Review

Hayashi, K., Ohta, H., Kurimoto, K., Aramaki, S., and Saitou, M. (2011). Recon-

stitution of the mouse germ cell specification pathway in culture by pluripotent

stem cells. Cell 146, 519–532.

Hayashi, K., Ogushi, S., Kurimoto, K., Shimamoto, S., Ohta, H., and Saitou, M.

(2012). Offspring from oocytes derived from in vitro primordial germ cell-like

cells in mice. Science 338, 971–975.

Herbert, M., and Turnbull, D. (2018). Progress in mitochondrial replacement

therapies. Nat. Rev. Mol. Cell Biol. 19, 71–72.

Jasin, M., and Rothstein, R. (2013). Repair of strand breaks by homologous

recombination. Cold Spring Harb. Perspect. Biol. 5, a012740.

Jinek, M., Chylinski, K., Fonfara, I., Hauer, M., Doudna, J.A., and Charpentier,

E. (2012). A programmable dual-RNA-guided DNA endonuclease in adaptive

bacterial immunity. Science 337, 816–821.

Kang, X., He, W., Huang, Y., Yu, Q., Chen, Y., Gao, X., Sun, X., and Fan, Y.

(2016). Introducing precise genetic modifications into human 3PN embryos

by CRISPR/Cas-mediated genome editing. J. Assist. Reprod. Genet. 33,

581–588.

Kim, Y.B., Komor, A.C., Levy, J.M., Packer, M.S., Zhao, K.T., and Liu, D.R.

(2017). Increasing the genome-targeting scope and precision of base editing

with engineered Cas9-cytidine deaminase fusions. Nat. Biotechnol. 35,

371–376.

Kofler, N., and Kraschel, K.L. (2018). Treatment of heritable diseases using

CRISPR: Hopes, fears, and reality. Semin. Perinatol. 42, 515–521.

Komor, A.C., Kim, Y.B., Packer, M.S., Zuris, J.A., and Liu, D.R. (2016). Pro-

grammable editing of a target base in genomic DNA without double-stranded

DNA cleavage. Nature 533, 420–424.

Kosicki, M., Tomberg, K., and Bradley, A. (2018). Repair of double-strand

breaks induced by CRISPR-Cas9 leads to large deletions and complex rear-

rangements. Nat. Biotechnol. 36, 765–771.

Kuijk, E., Jager, M., van der Roest, B., Locati, M.D., Van Hoeck, A., Korzelius,

J., Janssen, R., Besselink, N., Boymans, S., van Boxtel, R., and Cuppen, E.

(2020). The mutational impact of culturing human pluripotent and adult stem

cells. Nat. Commun. 11, 2493.

Lander, E.S., Baylis, F., Zhang, F., Charpentier, E., Berg, P., Bourgain, C., Frie-

drich, B., Joung, J.K., Li, J., Liu, D., et al. (2019). Adopt a moratorium on her-

itable genome editing. Nature 567, 165–168.

Lanphier, E., Urnov, F., Haecker, S.E., Werner, M., and Smolenski, J. (2015).

Don’t edit the human germ line. Nature 519, 410–411.

Lea, R.A., and Niakan, K. (2019). Human germline genome editing. Nat. Cell

Biol. 21, 1479–1489.

Leibowitz, M.L., Papathanasiou, S., Doerfler, P.A., Blaine, L.J., Yao, Y., Zhang,

C.-Z., Weiss, M.J., and Pellman, D. (2020). Chromothripsis as an on-target

consequence of CRISPR-Cas9 genome editing. bioRxiv, 2020.07.13.200998.

Li, G., Liu, Y., Zeng, Y., Li, J., Wang, L., Yang, G., Chen, D., Shang, X., Chen, J.,

Huang, X., and Liu, J. (2017). Highly efficient and precise base editing in dis-

carded human tripronuclear embryos. Protein Cell 8, 776–779.

Liang, P., Xu, Y., Zhang, X., Ding, C., Huang, R., Zhang, Z., Lv, J., Xie, X., Chen,

Y., Li, Y., et al. (2015). CRISPR/Cas9-mediated gene editing in human tripronu-

clear zygotes. Protein Cell 6, 363–372.

Liang, P., Ding, C., Sun, H., Xie, X., Xu, Y., Zhang, X., Sun, Y., Xiong, Y., Ma, W.,

Liu, Y., et al. (2017). Correction of b-thalassemia mutant by base editor in hu-

man embryos. Protein Cell 8, 811–822.

Liu, Y., Li, X., He, S., Huang, S., Li, C., Chen, Y., Liu, Z., Huang, X., and Wang,

X. (2020). Efficient generation of mouse models with the prime editing system.

Cell Discov. 6, 27.

Ma, H., Marti-Gutierrez, N., Park, S.W., Wu, J., Lee, Y., Suzuki, K., Koski, A., Ji,

D., Hayama, T., Ahmed, R., et al. (2017). Correction of a pathogenic gene mu-

tation in human embryos. Nature 548, 413–419.

Ma, H., Marti-Gutierrez, N., Park, S.W., Wu, J., Hayama, T., Darby, H., Van

Dyken, C., Li, Y., Koski, A., Liang, D., et al. (2018). Ma et al. reply. Nature

560, E10–E23.

MacArthur, D.G., Balasubramanian, S., Frankish, A., Huang, N., Morris, J.,

Walter, K., Jostins, L., Habegger, L., Pickrell, J.K., Montgomery, S.B., et al.;

1000 Genomes Project Consortium (2012). A systematic survey of loss-of-

function variants in human protein-coding genes. Science 335, 823–828.

Mali, P., Yang, L., Esvelt, K.M., Aach, J., Guell, M., DiCarlo, J.E., Norville, J.E.,

and Church, G.M. (2013). RNA-guided human genome engineering via Cas9.

Science 339, 823–826.

McCoy, R.C., Demko, Z.P., Ryan, A., Banjevic, M., Hill, M., Sigurjonsson, S.,

Rabinowitz, M., and Petrov, D.A. (2015). Evidence of Selection against Com-

plex Mitotic-Origin Aneuploidy during Preimplantation Development. PLoS

Genet. 11, e1005601.

Minasi, M.G., Fiorentino, F., Ruberti, A., Biricik, A., Cursio, E., Cotroneo, E.,

Varricchio, M.T., Surdo, M., Spinella, F., and Greco, E. (2017). Genetic dis-

eases and aneuploidies can be detected with a single blastocyst biopsy: a suc-

cessful clinical approach. Hum. Reprod. 32, 1770–1777.

Mok, B.Y., de Moraes, M.H., Zeng, J., Bosch, D.E., Kotrys, A.V., Raguram, A.,

Hsu, F., Radey, M.C., Peterson, S.B., Mootha, V.K., et al. (2020). A bacterial

cytidine deaminase toxin enables CRISPR-free mitochondrial base editing.

Nature 583, 631–637.

Mutton, D., Alberman, E., and Hook, E.B.; National Down Syndrome Cytoge-

netic Register and the Association of Clinical Cytogeneticists (1996). Cytoge-

netic and epidemiological findings in Down syndrome, England and Wales

1989 to 1993. J. Med. Genet. 33, 387–394.

Nambiar, T.S., Billon, P., Diedenhofen, G., Hayward, S.B., Taglialatela, A., Cai,

K., Huang, J.W., Leuzzi, G., Cuella-Martin, R., Palacios, A., et al. (2019). Stim-

ulation of CRISPR-mediated homology-directed repair by an engineered

RAD18 variant. Nat. Commun. 10, 3395.

NAS, National Academy of Medicine, National Academy of Sciences and the

Royal Society (2020). Heritable Human Genome Editing (The National Acade-

mies Press).

Nuffield Council on Bioethics (2018). Genome Editing and Human Reproduc-

tion: Social and Ethical Issues (Nuffield Council on Bioethics).

Palanki, R., Peranteau, W.H., and Mitchell, M.J. (2021). Delivery technologies

for in utero gene therapy. Adv. Drug Deliv. Rev. 169, 51–62.

Pawluk, A., Davidson, A.R., and Maxwell, K.L. (2018). Anti-CRISPR: discovery,

mechanism and function. Nat. Rev. Microbiol. 16, 12–17.

Plaza Reyes, A., and Lanner, F. (2017). Towards a CRISPR view of early human

development: applications, limitations and ethical concerns of genome editing

in human embryos. Development 144, 3–7.

Reddy, P., Ocampo, A., Suzuki, K., Luo, J., Bacman, S.R., Williams, S.L., Su-

gawara, A., Okamura, D., Tsunekawa, Y., Wu, J., et al. (2015). Selective elim-

ination of mitochondrial mutations in the germline by genome editing. Cell 161,

459–469.

Rees, H.A., and Liu, D.R. (2018). Base editing: precision chemistry on the

genome and transcriptome of living cells. Nat. Rev. Genet. 19, 770–788.

Richardson, C., Stark, J.M., Ommundsen, M., and Jasin, M. (2004). Rad51

overexpression promotes alternative double-strand break repair pathways

and genome instability. Oncogene 23, 546–553.

Rossant, J. (2018). Gene editing in human development: ethical concerns and

practical applications. Development 145, dev150888.

Rouhani, F.J., Nik-Zainal, S., Wuster, A., Li, Y., Conte, N., Koike-Yusa, H., Ku-

masaka, N., Vallier, L., Yusa, K., and Bradley, A. (2016). Mutational History of a

Human Cell Lineage from Somatic to Induced Pluripotent Stem Cells. PLoS

Genet. 12, e1005932.

Safier, L.Z., Zuccaro, M.V., and Egli, D. (2020). Efficient SNP editing in haploid

human pluripotent stem cells. J. Assist. Reprod. Genet. 37, 735–745.

Sagi, I., Chia, G., Golan-Lev, T., Peretz, M., Weissbein, U., Sui, L., Sauer, M.V.,

Yanuka, O., Egli, D., and Benvenisty, N. (2016). Derivation and differentiation of

haploid human embryonic stem cells. Nature 532, 107–111.

Savulescu, J., and Singer, P. (2019). An ethical pathway for gene editing.

Bioethics 33, 221–222.

Cell 184, March 18, 2021 1573

ll
Review

Schwank, G., Koo, B.K., Sasselli, V., Dekkers, J.F., Heo, I., Demircan, T., Sa-

saki, N., Boymans, S., Cuppen, E., van der Ent, C.K., et al. (2013). Functional

repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic

fibrosis patients. Cell Stem Cell 13, 653–658.

Shin, J., Jiang, F., Liu, J.-J., Bray, N.L., Rauch, B.J., Baik, S.H., Nogales, E.,

Bondy-Denomy, J., Corn, J.E., and Doudna, J.A. (2017). Disabling Cas9 by

an anti-CRISPR DNA mimic. Sci. Adv. 3, e1701620.

Tang, L., Zeng, Y., Du, H., Gong, M., Peng, J., Zhang, B., Lei, M., Zhao, F.,

Wang, W., Li, X., and Liu, J. (2017). CRISPR/Cas9-mediated gene editing in hu-

man zygotes using Cas9 protein. Mol. Genet. Genomics 292, 525–533.

Viotti, M., Victor, A.R., Griffin, D.K., Groob, J.S., Brake, A.J., Zouves, C.G., and

Barnes, F.L. (2019). Estimating Demand for Germline Genome Editing: An

In Vitro Fertilization Clinic Perspective. CRISPR J. 2, 304–315.

Wilde, J.J., Aida, T., Wienisch, M., Zhang, Q., Qi, P., and Feng, G. (2018). Effi-

cient Zygotic Genome Editing via RAD51-Enhanced Interhomolog Repair. bio-

Rxiv, 263699.

Wu, Y., Zhou, H., Fan, X., Zhang, Y., Zhang, M., Wang, Y., Xie, Z., Bai, M., Yin,

Q., Liang, D., et al. (2015). Correction of a genetic disease by CRISPR-Cas9-

mediated gene editing in mouse spermatogonial stem cells. Cell Res. 25,

67–79.

Yamashiro, C., Sasaki, K., Yabuta, Y., Kojima, Y., Nakamura, T., Okamoto, I.,

Yokobayashi, S., Murase, Y., Ishikura, Y., Shirane, K., et al. (2018). Generation

of human oogonia from induced pluripotent stem cells in vitro. Science 362,

356–360.

Yilmaz, A., Braverman-Gross, C., Bialer-Tsypin, A., Peretz, M., and Benve-

nisty, N. (2020). Mapping Gene Circuits Essential for Germ Layer Differentia-

tion via Loss-of-Function Screens in Haploid Human Embryonic Stem Cells.

Cell Stem Cell 27, 679–691.

1574 Cell 184, March 18, 2021

Yoshihara, M., Araki, R., Kasama, Y., Sunayama, M., Abe, M., Nishida, K., Ka-

waji, H., Hayashizaki, Y., and Murakawa, Y. (2017). Hotspots of De Novo Point

Mutations in Induced Pluripotent Stem Cells. Cell Rep. 21, 308–315.

Zeng, Y., Li, J., Li, G., Huang, S., Yu, W., Zhang, Y., Chen, D., Chen, J., Liu, J.,

and Huang, X. (2018). Correction of the Marfan Syndrome Pathogenic FBN1

Mutation by Base Editing in Human Cells and Heterozygous Embryos. Mol.

Ther. 26, 2631–2637.

Zhang, J., Liu, H., Luo, S., Lu, Z., Chávez-Badiola, A., Liu, Z., Yang, M., Merhi,

Z., Silber, S.J., Munné, S., et al. (2017). Live birth derived from oocyte spindle

transfer to prevent mitochondrial disease. Reprod. Biomed. Online 34,

361–368.

Zhang, M., Zhou, C., Wei, Y., Xu, C., Pan, H., Ying, W., Sun, Y., Sun, Y., Xiao,

Q., Yao, N., et al. (2019). Human cleaving embryos enable robust homozygotic

nucleotide substitutions by base. Genome Biol. 20, 101.

Zhang, X.M., Wu, K., Zheng, Y., Zhao, H., Gao, J., Hou, Z., Zhang, M., Liao, J.,

Zhang, J., Gao, Y., et al. (2020). In vitro expansion of human sperm through nu-

clear transfer. Cell Res. 30, 356–359.

Zhou, C., Zhang, M., Wei, Y., Sun, Y., Sun, Y., Pan, H., Yao, N., Zhong, W., Li,

Y., Li, W., et al. (2017). Highly efficient base editing in human tripronuclear zy-

gotes. Protein Cell 8, 772–775.

Zuccaro, M.V., Xu, J., Mitchell, C., Marin, D., Zimmerman, R., Rana, B., Wein-

stein, E., King, R.T., Palmerola, K.L., Smith, M.E., et al. (2020). Allele-Specific

Chromosome Removal after Cas9 Cleavage in Human Embryos. Cell 183,

1650–1664.

Zuo, E., Huo, X., Yao, X., Hu, X., Sun, Y., Yin, J., He, B., Wang, X., Shi, L., Ping,

J., et al. (2017). CRISPR/Cas9-mediated targeted chromosome elimination.

Genome Biol. 18, 224.

  • Heritable human genome editing: Research progress, ethical considerations, and hurdles to clinical practice
    • DNA double-strand breaks allow genome editing in human embryos
    • Adverse consequences of DNA breaks in human embryos
    • Mitochondrial replacement therapy
    • Editing of in vitro-derived gametes and embryos
    • Spermatogonial stem cells
    • Induced pluripotent stem cells
    • Haploid human stem cells
    • Ethical considerations of heritable human genome editing
    • Beneficence: Therapeutic benefits
    • Non-maleficence: Risks and safety
    • Non-maleficence: Exploitation for non-therapeutic modification
    • Autonomy: Reproductive rights and lack of ability to consent before birth
    • Justice: Equitable access and allocation of resources
    • Embryo research and destruction
    • Conclusions
    • Acknowledgments
    • Declaration of interests
    • References

Commentary

Gene editing and the health of future generations

Christopher Gyngell
Oxford Uehiro Centre for Practical Ethics, Oxford University, Oxford OX1 3PA, UK

Corresponding author: Christopher Gyngell. Email: [email protected]

The CRISPR-cas9 gene-editing system (CRISPR) is a
revolutionary technology that promises unparalleled
abilities. It is the first technology that allows for the
precise, efficient modification of DNA sequences. Less
than five years since it was first developed, it has been
used to alter a diverse range of organisms, including
plants, livestock, insects and primates. There is little
doubt that it will soon be technically possible to use
the CRISPR system to rewrite the human genome. It is
crucial that we consider the impact such technologies
will have on future generations. The ability to alter our
biological makeup will create immense opportunities
but also pose novel threats. It is crucial that we make
sensible decisions about the development and use of
gene-editing technologies.

In this commentary, I discuss the effect that germ-
line gene editing will have on the health of future gen-
erations (Note: By ‘germline’ gene editing, I mean the
editing of DNA in cells which could potentially be her-
itable, e.g. germ cells or embryonic cells.). I argue that
provided germline gene editing is well regulated, it
could greatly improve the health of our descendants.
The use of germline gene editing in research will greatly
increase our knowledge of development and could lead
to novel treatments for disease. Germline gene editing
also has enormous potential as a clinical tool. It could
soon be used to prevent simple genetic diseases, and
eventually to reduce the incidents of polygenic dis-
eases. While the use of germline gene editing to prevent
disease raises contentious philosophical issues, concep-
tual uncertainty should not prevent the development of
germline gene editing as a research tool and a treatment
for fatal genetic conditions.

The research applications of germline gene
editing

The most significant question currently facing coun-
ties in regards to germline gene editing is whether to
use germline gene editing for research purposes. In
many countries around the world, such as Canada,
Australia and most of Europe, any form of research

using germline gene editing is banned. Many of these
bans were legislated in eras of far cruder genetic
engineering technologies. Gene-editing techniques
like CRISPR-cas9 are much more precise and effi-
cient than previous methods, and are the first tech-
nologies with serious potential to be used to modify
the human germline.

The research case in favour of pursuing germline
gene editing is very strong.

1
Editing human embry-

onic stem cells could be a breakthrough for the study
of early human development. Many theories regard-
ing how many events happen in early development
are based on mice models, which are proving to be
unreliable.

2
Early human development remains lar-

gely a mystery. Using germline gene editing to inves-
tigate the activity of specific groups of genes allows
researchers to better understand the processes that
drive development.

Improving our knowledge of development will
help provide better cures of infertility. Less than a
third of fertilised embryos survive pregnancy.

3
We

have a poor understanding of why this is. Using
germline gene editing to study early development
could lead to a great understanding of the causes of
infertility and to better treatment options.

Germline gene editing can also improve our under-
standing of genetic diseases. Gene editing allows
researchers to generate embryonic system cell lines
with different specific disease alleles on the same gen-
etic background.

4
Such cell lines can be used for the

study of genetic disease. For example, the CRISPR
system could be used to alter embryonic system cells
to contain mutations associated with Parkinson’s dis-
ease. These cells could then be induced to grow into
nerve cells (which malfunction in Parkinson’s disease).
These nerve cells could be used for the detailed study of
the mechanisms involved in Parkinson’s disease, and
serve as a platform to test potential treatments.
Germline gene editing could thus expedite the develop-
mentof pharmacological therapies for genetic diseases.

While such research can be performed using
induced pluripotent stem cells, embryonic system

Journal of the Royal Society of Medicine; 2017, Vol. 110(7) 276–279

DOI: 10.1177/0141076817705616

! The Royal Society of Medicine 2017

Reprints and permissions: sagepub.co.uk/journalsPermissions.nav

cells may have technical advantages.
5
Induced pluri-

potent stem cell models are created from somatic
cells, which may have undergone epigenetic changes.
As a result, induced pluripotent stem cells may be
more diverse and behave less predictably than
embryonic system cells in certain applications.

The use of germline gene editing in research, there-
fore, could improve the health of future generations.
By providing a new way to study human develop-
ment, germline gene editing may lead to better treat-
ments for infertility. Furthermore, germline gene
editing could be used to create cellular models and
further our understanding of genetic disease. Such
knowledge may be valuable in its own right, in add-
ition to leading to treatments for serious disease.

Single gene disorders

Beyond research, it may soon be feasible to use germ-
line gene editing in human reproduction. The most
obvious clinical use of germline gene editing will be to
correct the mutations associated with fatal single gene
disorders such as Tay Sachs disease, Duchenne mus-
cular dystrophy, cystic fibrosis and spinal muscular
atrophy. These conditions are caused by well-under-
stood genetic mechanisms and can reduce life expect-
ancy by decades. We currently use genetic selection
techniques like preimplantation genetic diagnosis
(preimplantation genetic diagnosis) to reduce the
incidence of these conditions, but preimplantation
genetic diagnosis is not always effective. When IVF
only produces a small number of viable embryos,
selection is not possible. Furthermore, preimplanta-
tion genetic diagnosis is useless to those who are
homozygotes for dominant conditions like
Huntington’s disease. In these cases, using germline
gene editing will be the only way that individuals can
avoid serious disease in their children.

Many object that such cases are rare, and that
preimplantation genetic diagnosis is effective in the
vast majority of cases. But even when selection can
be used to avoid disease, germline gene editing may
provide the more desirable option. Preimplantation
genetic diagnosis involves creating a number of
embryos, testing each and then only implanting
those most likely to be healthy. Preimplantation
genetic diagnosis nearly always results in embryos
being discarded. For some, this is an undesirable fea-
ture of preimplantation genetic diagnosis, which
germline gene editing can avoid. (Note: Such benefits
largely depend on germline gene editing developing to
the point where it is efficient enough to be used on a
single embryo.) Furthermore, the way in which germ-
line gene editing avoids disease may be preferable to
preimplantation genetic diagnosis. Selection prevents

disease by changing who comes into existence,
whereas gene editing ensures those who come into
existence have the best shot of living a full life.
Using germline gene editing to avoid disease thus
seems more analogous to curing a disease than pre-
implantation genetic diagnosis.

Furthermore, germline gene editing may be more
preferable than selection in the treatment of single
gene disorders because of its potential to reduce
rates of genetic diseases in the next generation.
Preimplantation genetic diagnosis is often not used
to select against carriers of a condition, partly
because this is difficult to achieve with the number
of embryos couples typically produce through IVF.
In the case of autosomal recessive disorders, children
who are born as the result of preimplantation genetic
diagnosis are likely to be carriers of condition their
parents selected against. Germline gene editing will
provide a way to remove all disease-causing genes
from an embryo, and so the germ cells in that
embryo will not carry the mutation. Using germline
gene editing to prevent single gene disorders will thus
provide a more effective way to reduce the incidence
of these diseases in future generations than preim-
plantation genetic diagnosis.

Chronic diseases

In the far future – perhaps in a few decades – we may
be in a position to use embryonic gene editing to
target other causes of death. Roughly 30% of all
deaths worldwide are due to chronic diseases (such
as heart disease, cancer and diabetes) in those under
70.

6
Many billions of dollars are spent each year

trying to develop new treatments to these disorders
and reduce their impact on mortality. We know that
chronic disease is affected by our genetic make-up.
For example, genome-wide association studies have
identified at least 44 genes involved in diabetes

7
;

35 genes involved in coronary artery disease
8
and

over 300 genes involved in common cancers.
9
As we

understand more about genetics, and more about the
aetiology of these disorders, it will be possible to
reduce our susceptibility to these diseases. The ability
of germline gene editing to target multiple genes sim-
ultaneously means it could potentially be used to
reduce the incidence of these disorders.

Disability, diversity and risks to human
health

As the above section argues, it is clear that germline
gene editing could be used to reduce genetic disease.
But this raises the question – how far to do we go? Do
we use germline gene editing to target all diseases – all

Gyngell 277

undesirable traits? Such questions are complex and
controversial. A common theme of the disability
pride movement is that our common sense views of
disability are mistaken. Many of the conditions that
we view as diseases and disability are not, in fact
something bad, but rather something to take pride
in. This presents a worry. If germline gene editing is
used to eradicate conditions that are in fact not nega-
tive, this will not improve the health of future gener-
ations at all.

Worse, if we use germline gene editing overzea-
lously, it may harm future generations, by removing
valuable forms of human diversity.

10
Human groups

benefit from certain types of diversity, including
immuno-diversity (diversity in the genes that influ-
ence innate immunity) and cognitive diversity (diver-
sity in the genes that affect our cognitive traits). It is
plausible that some conditions we think of as diseases
may contribute to valuable forms of diversity. For
example, it is plausible that Asperger’s syndrome
and dyslexia are sources of valuable forms of cogni-
tive diversity.

11
Similarly, conditions like deafness

which cause people to experience the world in
unique ways, may also contribute to valuable forms
of diversity.

These questions are difficult and complex. It is the
subject of intense debate in philosposphy how to dis-
tinguish healthy forms of human diversity from dis-
ease and disability. However, we should not let this
conceptual uncertainty be barrier to the development
of germline gene editing.

As noted above, germline gene editing is valuable
as a research tool; independent of whether it is ever
used in a clinical setting. Furthermore, even if some
diseases and disabilities may be valuable forms of
diversity, many are clearly not. No one plausibly
holds that Tay Sachs syndrome (a degenerative dis-
ease of the nervous system that commonly causes
death before four years of age) is a valuable form
of human diversity rather than a horrible disease.
Similarly, there are other diseases which have
simple genetic mechanisms, and which take decades
of life from people (including cystic fibrosis and
spinal muscular atrophy). Such diseases seem likely
to be negative rather than neutral forms of genetic
diversity.

One option, then, is to limit the use of germline
gene editing, to the prevention of severe fatal condi-
tions. Similar principles already govern access to
other reproductive technologies like preimplantation
genetic diagnosis. In the UK, regulations limit pre-
implantation genetic diagnosis to being used to select
against ‘serious’ inherited conditions. However, what
is regarded as ‘serious’ is considered on a case-by-case
basis. Each proposed use of preimplantation genetic

diagnosis is examined individually. Those that are
deemed to be risky can be rejected.

There is no reason why such a system could not
work for germline gene editing. A case-by-case
system could work both to reduce rates of fatal genetic
disease and avoid risking traits that may represent
valuable types of diversity. If regulated in such a
way, germline gene editing could greatly improve the
health of future generations

Declarations

Competing Interests: None declared

Funding: Christopher Gyngell would like to thank the Marie
Curie Actions of the European Union’s 2014 Horizon2020 work

programme (grant agreement n� 659700) for its funding.

Ethical approval: Not applicable

Guarantor: CG.

Contributorship: Sole authorship.

Acknowledgements: This article builds from the arguments I
presented in a debate at the Royal Society for Medicine event

‘Gene editing in medicine: breakthrough or thin edge of the

wedge?’.

Provenance: Commissioned; editorial review.

References

1. Gyngell C, Douglas T and Savulescu J. The ethics of
germline gene editing. J Appl Philos. Epub ahead of

print November 2016. DOI: 10.1111/japp.12249.
2. Irie N, Weinberger L, Tang WW, Kobayashi T, Viukov

S, Manor YS, et al. SOX17 is a critical specifier of

human primordial germ cell fate. Cell 2015; 160:
253–268.

3. Macklon NS, Geraedts JPM and Fauser BCJM.
Conception to ongoing pregnancy: the ‘black box’ of

early pregnancy loss. Hum Reprod Update 2002; 8:

333–343.
4. Committee on Human Gene Editing: Scientific,

Medical, and Ethical Considerations, National
Academy of Sciences, National Academy of Medicine,

et al. Human Genome Editing: Science, Ethics, and

Governance. Washington, D.C.: National Academies
Press. See www.nap.edu/catalog/24623 (last checked 20

March 2017).
5. Bilic J and Izpisua Belmonte JC. Concise review:

Induced pluripotent stem cells versus embryonic stem
cells: close enough or yet too far apart? Stem Cells

2012; 30: 33–41.
6. Chronic diseases and health promotion, World Health

Organisation website. See www.who.int/chp/en/ (last

checked 20 March 2017).
7. Berndt SI, Gustafsson S, Mägi R, Ganna A, Wheeler E,

Feitosa MF, et al. Genome-wide meta-analysis identifies
11 new loci for anthropometric traits and provides

insights into genetic architecture. Nat Genet 2013; 45:

501–512.

278 Journal of the Royal Society of Medicine 110(7)

8. Peden JF and Farrall M. Thirty-five common variants
for coronary artery disease: the fruits of much collab-
orative labour. Hum Mol Genet 2011; 20: R198–205.

9. Chang CQ, Yesupriya A, Rowell JL, Pimentel CB,
Clyne M, Gwinn M, et al. A systematic review of
cancer GWAS and candidate gene meta-analyses reveals
limited overlap but similar effect sizes. Eur J Hum Genet

2014; 22: 402–408.

10. Gyngell C. Enhancing the species: genetic engineering
technologies and human persistence. Philos Technol
2012; 25: 495–512.

11. Gyngell C and Douglas T. Selecting against disability:
the liberal eugenic challenge and the argument from
cognitive diversity. Journal of Applied Philosophy.
DOI: 10.1111/japp.12199.

Gyngell 279

Writerbay.net

Do you have a lot of essay writing to do? Do you feel like you’re struggling to find the right way to go about it? If so, then you might want to consider getting help from a professional essay writer. Click one of the buttons below.


Order a Similar Paper Order a Different Paper