Ethical Issues in Genetic Modification and Why Application Matters

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Available online at www.sciencedirect.

com

ScienceDirect

Ethical issues in genetic modification and why


application matters
Megan Munsie1 and Christopher Gyngell2

Advances in genome editing techniques have generated engineering, raising the spectre of ‘editing’ the genomes
renewed interest in the ethical implications of genetic our own cells or even those of our descendants at will.
modification. In this article, we review the recent literature and
discuss in detail ethical issues pertaining to the application of The most notable of these advanced ‘genome editing’
this technology to five areas; human embryo research, techniques is the clustered regularly interspaced short
organoid research, the prospect of genetically modified babies, palindromic repeats (CRISPR-Cas9) system. This
mitochondrial replacement therapy and the creation of approach, developed by a team at UC Berkeley in
chimeric organisms. We point to a central issue which cuts 2012, showed that CRISPR-Cas9 could be modified so
through these different areas: the need to clearly frame how that it could target virtually any DNA sequence [5],
using the technology provides benefit that cannot be met by giving researchers the ability to delete, add, or modify
other means. Failure to provide reasonable justification, and DNA sequences with greatly increased precision. The
address how risks — if any — will be mitigated, is likely to CRISPR-Cas9 approach has since been used to modify
erode public trust and undermine progress in medical research the genome of mice [6], dogs [7], pigs [8] and primates [9].
and its clinical translation.
These developments have brought to focus a number of
important ethical questions. In this review, we discuss the
Addresses ethical issues raised by genome modification technologies
1
Centre for Stem Cell Systems, School of Biomedical Sciences, and why application remains a paramount consideration
University of Melbourne, Parkville, Australia for the future.
2
Murdoch Children’s Research Institute and University of Melbourne,
Parkville, Australia
Recent controversy — editing the human germline
Corresponding author: Munsie, Megan (megan.munsie@unimelb.edu.au) In April 2015, a team in China became the first to use
genome editing technologies on human embryos [10].
The study, which attempted to correct the gene respon-
Current Opinion in Genetics & Development 2018, 52:7–12
sible for b-thalassaemia using ‘non-viable’ IVF embryos,
This review comes from a themed issue on Cell reprogramming,
sparked a worldwide debate about how research involving
regeneration and repair
germline genome editing (a practice we will call GGE3)
Edited by Knut Woltjen and Alex Bortvin should be regulated. Some scientists and public interest
For a complete overview see the Issue and the Editorial groups, including the United Nations Educational Scien-
Available online 23rd May 2018 tific and Cultural Organization have called for an inter-
https://doi.org/10.1016/j.gde.2018.05.002
national ban on any gene editing research in human
embryos [11]. The U.S. based National Institutes of
0959-437/ã 2018 Elsevier Ltd. All rights reserved.
Health, maintained that performing such research passed
‘a line that should not be crossed’ [12]. Opinion articles in
the leading journals Nature and Science called for a mora-
torium on any GGE research [13,14].

Introduction In response to this development, some called for collec-


For over 40 years, scientists have been able to genetically tive efforts to carefully analyse the ethical, legal, and
modify the genome of mammalian cells. The first social implications for altering the germline, and called for
‘transgenic’ mouse was created by Rudolf Jaenisch in broad public discussion on the issue [15]. In December
1974 [1]. Since this time various techniques have been 2015, the Hinxton Group, an international consortium of
used to insert, delete or modify DNA, in order to create scientists, ethicists and policy experts, convened a meet-
animals with altered physical [2], cognitive [3] and social ing to analyse the ethics of gene editing technologies.
[4] characteristics. While invaluable to advance basic The Hinxton Report resisted calls for moratorium on all
research, the potential clinical applications have been GGE research. While it makes clear that any reproductive
hampered by the challenge of reliably modifying the
desired genome sequence without any off-target effects. 3
By GGE, we mean the editing of any cells, including gametes and
However, advances over the past six years have led to cells in early cleavage stage embryos, in which the changes made to the
more precise and efficient methods for genetic genome could be inherited by decedents of that cell.

www.sciencedirect.com Current Opinion in Genetics & Development 2018, 52:7–12


8 Cell reprogramming, regeneration and repair

use of GGE is premature, it highlights the important roles directly . . . ’ [21]. However, it is also unclear how infor-
that GGE could achieve in basic research, such as helping mative experiments using genetically abnormal embryos
us understand the mechanisms that underlie early human are in advancing the technology given that information
development [16]. about off-target mutations, mosaicism and about human
development, cannot be easily extrapolated [16].
In 2016, International Society for Stem Cell Research
released its ‘Guidelines for Stem Cell Science and Clini- These limitations raise the question of how ‘normally’
cal Translation’. Like the Hinxton Report, this publica- fertilised human embryos could be ethically sourced. Most
tion highlighted the importance of basic research which human embryos used in research are ‘left-over’ from IVF
could be accomplished with GGE. However, it takes a treatment. These embryos, normally fertilised and at the
harder line on the use of these technologies for reproduc- early stages of pre-implantation embryonic development,
tive purposes, starting that that such uses ought to be are no longer required by the couple and maybe available
prohibited; and that there needs to be ‘a deeper and more to donate to research. Using CRISPR-Cas9 on these
rigorous deliberation on the ethical, legal, and societal embryos (often 4–8 cell or blastocyst stage) is likely to
implications of modifying the human germ line is essen- give variable results with genetic modification being
tial if clinical application is ever to be sanctioned’ [17]. achieved in some cells and not others, resulting in a
mosaic of edited and non-edited expression across the
In 2017, a comprehensive joint report from the U.S. embryo. Normally fertilised zygotes are rarely available
National Academy of Sciences and U.S. National Acad- for research. Therefore, from a research perspective, it has
emy of Medicine, was released which looked at the been argued that it may be better to specifically create
ethical and regulatory implications for human genome zygotes using donated eggs and sperm for the purpose of
editing. This report builds on fundamental ethical prin- research — an approach adopted in two recent gene edit-
ciples [18] to describe the overarching considerations for ing studies [22,23]. While such a strategy may be accept-
the conduct and oversight of genome editing technologies able to some, and has received ethics approval, such an
for basic research and clinical application [19]. Notably, approach opens these studies up to objections that they
the report states that clinical trials using GGE could be are unethical by creating an embryo whose sole purpose is
considered if 10 steps are met, importantly whether the to be used as a means for research [24]. How, where and
clinical objectives are unable to be reached through from whom to source appropriate material to progress
reasonable alternatives (see Table 1). GGE using human gametes and embryos, and how to
regulate such research, is likely to be an ongoing chal-
The recent public attention on modifying human lenge for the field.
embryos has generated much discussion on the ethics
of such research; but also the ethical issues raised by Organoids
genetically modifying human cells more generally. We Research involving organoids — clusters of cells derived
review the major issues below, distinguishing between from tissue or pluripotent stem cells that self-organize in
issues raised by genetic modification in basic research and ways which mimic tissue and organ function — allows
issues raised through areas of future clinical application of scientists to create models which can further our under-
genetic modification. standing of biological processes underpinning develop-
ment and the progression of disease [25]. It has also been
proposed that in the future organoids may be a source of
Ethical issues raised in research
functional tissues and organs for transplantation, a quest
Genetically modifying embryos
that still has many challenges in terms of meeting neces-
To date, all of the research involving the germline modi-
sary standards of scale and maturity. However, the recent
fication has taken place during early embryo development;
report where gene editing techniques were used to
at, or immediately after, fertilisation (see Table 2). For
restore normal function in intestinal organoids derived
those who think that early embryos have the same moral
from cystic fibrosis patients provide an indication of
status as adult humans, this is morally problematic [20].
future therapeutic applications [26].
Initial GGE studies using human embryos tried to miti-
gate these concerns by using ‘non-viable’ IVF embryos4
Organoid research raises a number of important ethical
that were identified as being abnormally fertilised and
issues [27]. Specifically, organoid research raises ques-
therefore not suitable for infertility treatment. As Savu-
tions around moral status of these structures, about
lescu et al., commented ‘as trialling the CRISPR system in
whether their creation constitutes ‘life’ or if they hold
these zygotes had no chance of resulting in a live birth, it is
independent interest and rights. This is particularly rele-
unclear how the study could harm or wrong anyone
vant for research involving gastruloids, structures that are
4
Researchers used one-cell embryos, also referred to as zygotes, that
made in the lab from pluripotent stem cells and that
displayed more than two pro-nuclei following fertilisation; indicating mimic post-implantation embryonic development [28].
abnormal fertilisation. While most jurisdictions limit the development of human

Current Opinion in Genetics & Development 2018, 52:7–12 www.sciencedirect.com


Ethics and genetic modification: why application matters Munsie and Gyngell 9

Table 1

Key considerations that NAS recommends must be met before undertaking clinical research using heritable human germline genome
editing [19]

Consideration
The absence of reasonable alternatives
Restriction to preventing a serious disease or condition
Restriction to editing genes that have been convincingly demonstrated to cause or to strongly predispose to that disease or condition
Restriction to converting such genes to versions that are prevalent in the population and are known to be associated with ordinary health with little or
no evidence of adverse effects
The availability of credible preclinical and/or clinical data on risks and potential health benefits of the procedures
During the trial, ongoing, rigorous oversight of the effects of the procedure on the health and safety of the research participants
Comprehensive plans for long-term, multigenerational follow-up that still respect personal autonomy
Maximum transparency consistent with patient privacy
Continued reassessment of both health and societal benefits and risks, with broad ongoing participation and input by the public
Reliable oversight mechanisms to prevent extension to uses other than preventing a serious disease or condition

Table 2

Summary of stated rationale for GGM in recent human embryos experiments

Publication Developmental stage used Rationale for GGM Alternative approach


Liang et al., 2015 [10] Abnormally fertilised (3PN) zygotes — Correct common b-thalassemia mutation PGD on IVF embryos
donated by IVF patients
Kang et al., 2016 [52] Abnormally fertilised (3PN) zygotes — Introduce mutation to disrupt receptor for Strategies to address
donated by IVF patients HIV-1 thereby reducing risk of infection HIV infection
Tang et al., 2017 [23] Abnormally fertilised (3PN) zygotes and Correct common b-thalassemia mutation PGD on IVF embryos
normally fertilised embryos created from and X-linked mutation causing glucose-6-
immature eggs — donated by IVF patients phosphate dehydrogenase deficiency
Ma et al., 2017 [22] Zygotes (normally fertilised) and co- Correction of mutation of which causes a PGD on IVF embryos
injection with sperm at fertilisation — type of heart disease (hypertrophic
embryos created for research cardiomyopathy) to prevent germline
transmission and ‘rescue mutant embryos,
increase the number of embryos available
for transfer and ultimately improve
pregnancy rates.’
Fogarty et al., 2017 [53] Normally fertilised zygotes — donated by Explore the molecular mechanism of early None
IVF patients development by inducing mutation in OCT4
gene

embryos in a lab to 14 days post fertilisation, gastruloids Such questions are vital to how we assess the moral
make it possible study development beyond this point implications of this research. It is crucial that such ques-
and thereby provide insight into early human develop- tions are considerations as research continues [27].
ment during first trimester pregnancy and miscarriage
[28]. But is such research ethical? Do gastruloids have An additional and fundamental consideration in organoid
the same moral standing as early human embryos derived research relates to informed consent [30]. What type of
from fertilisation, and should the same ethical principles consent procedures are appropriate? Does this need to be
apply to them? [28]. modified where the research involves creation of gastru-
loids or cerebral organoids? What processes are in place to
Growing cerebral organoids in a dish — where they mimic ensure that the donor’s intentions are honoured? Further-
certain structural and functional aspects of the brain — is more, as organoids can be stored in biobanks for future
also an area that has attracted significant concern. While research, much like a 3D cell line, questions about the
structural complexity is currently limited due to technical ownership, distribution and commercialisation may need
challenges of maintaining these structures for sufficient to be considered after consent has been given [31].
time to allow them to mature, in time these models are
likely to become more sophisticated and better able to Ethical issues raised by future clinical applications
imitate functions of a mature brain. Questions must be While there remain considerable technical and ethical
asked about the moral standing of these structures [29]. challenges in delivering gene therapy through genetic
When do they begin to have experiences, or even gain the modification of somatic cells [32], we have elected in this
capacity to feel pain? How can we ascertain if they do? review to focus on three future applications that have

www.sciencedirect.com Current Opinion in Genetics & Development 2018, 52:7–12


10 Cell reprogramming, regeneration and repair

attracted substantial community interest, generate novel the organ transplant shortage, one U.S. group is seeking to
ethical challenges, and merit careful consideration around create genetically modified pigs, where the pancreas is
justification and public engagement. composed of human cells [46]. The resulting genetically
modified pigs could help save lives with some arguing we
Genome edited babies have strong moral imperative to develop these chimeras
By far, the prospect of genetically modified children is the for use in organ transplantation [47]. Others point to
ethical issue that has generated the most discussion in worries that growing human cells in an animal could
relation genome editing technologies. Proponents of this inadvertently affect the developing animal in ways that
application state that such technologies can help fight go well beyond the localized formation of human organs.
disease and reduce suffering more broadly — and that For example, it is possible that human cells could migrate
there is therefore a moral imperative to develop them to the nervous system and effect brain development,
[33]. In contrast, many critics of gene editing claim it will resulting in animals with ambiguous moral status [48].
be wrong at one or more levels [34]. They will harm the
child as they will lead to the commodification of children The need to justify application
[35]; they will harm parents by hurting their relationships A theme cutting through all of the above ethical issues is
with their children [36]; harm society by exacerbating the need for a feasible justification to underpin the
social inequalities [37], or harm the species by altering our application. While deepening understanding and advanc-
evolutionary trajectory [38]. Proponents argue that either ing progress towards a therapeutic outcome may be
these concerns are either unfounded, or that they also sufficient to justify GGE in basic research, applications
apply to existing reproductive technologies, like Pre- aiming at reproductive uses of GGE should only be
implantation Genetic Diagnosis (PGD) — where considered in a limited set of circumstances (see Table 1).
embryos are selected based on their pattern of gene Despite this, four out the five initial studies performing
expression — and the risks could be captured under GGE have justified their work through speculative ther-
existing regulatory frameworks [39].therapy apeutic goals that could arguably be met using existing
While the use of GGE for reproduction remains in the reproductive technologies (see Table 2). Only one study
future, another type of genetic modification has been has used CRISPR to investigate early development.
legalised in the United Kingdom [40]. Mitochondrial Given the controversial nature of genetically modifying
replacement therapy (MRT) is a broad term referring children; and the fact that more advanced methods of
to techniques which involve the creation of human gene editing is likely to be developed in the near future,
embryos using mitochondria from third party [41]. the justification for such research is precarious. What is
Employed to prevent transmission of mitochondrial dis- the long-term value of knowing how effectively CRISPR-
ease and allow a woman to conceive a child that is Cas9 can alter HIV resistance genes, when we do not even
biologically hers, a child created using this technology know how significant of a health problem HIV will be in
will inherit their genomic (nuclear) DNA from their 50 years? Why correct for a mutation that causes thalas-
‘biological’ parents but their mitochondria (and the semia or heart disease when PGD has already been shown
DNA contained within these organelles) from an egg to be able to select embryos that do not carry these, and
donor. similar genetic conditions, without the risk of off-site
genetic modifications to the child?
The key ethical issues that MRT raises include questions
about the long-term safety of the technology [42]; The risks are not just to scientific progress. While surveys
whether mitochondrial changes can alter the identity of of public opinions are generally supportive of gene edit-
a child [43,44]; and whether it should be considered as a ing technologies [49,50], this could quickly erode. We
type of germline therapy [41]. In response to some of echo concerns expressed elsewhere that pursuit of gene
these concerns, the National Academy of Sciences has editing research aimed at reproductive applications needs
recommended limiting MRT, to male children [45]; who to consider assumptions related to reproductive options
do not pass their mitochondria on to future generations. and disease ‘seriousness’ [51]. Researchers have a duty of
care to be involved in the ongoing debate.
Chimeric organisms
Chimeras are organisms composed of cells from two or Conflict of interest
more ‘individuals’. While chimeric mice — usually cre- Neither MM or CG work for, consult, own shares in or
ated by aggregating cells from different embryos or by receive funding from any company or organisation that
introducing an embryonic stem cell into a developing would benefit from this article.
blastocyst — have been an essential aspect of biological
research for decades, more recently researchers have been
using these techniques to create animals where the chi- Acknowledgements
Christopher Gyngell, through his involvement with the Murdoch Children’s
meric contribution from human cells is predominantly Research Institute, received funding from the Victorian State Government
restricted one organ. For example, in an effort to address through the Operational Infrastructure Support (OIS) Program.

Current Opinion in Genetics & Development 2018, 52:7–12 www.sciencedirect.com


Ethics and genetic modification: why application matters Munsie and Gyngell 11

References and recommended reading 18. For example, those outlined by the National Commission for the
Protection of Human Subjects of Biomedical and Behavioral
Papers of particular interest, published within the period of review, Research, Department of Health, Education and Welfare (DHEW).
have been highlighted as: The Belmont Report (PDF). Washington, DC: United States
Government Printing Office; 1978.
 of special interest
 of outstanding interest 19. National Academy of Sciences: Human Genome Editing: Science,
 Ethics and Governance. Washington, DC, USA: National
1. Jaenisch R, Mintz B: Simian Virus 40 DNA Sequences in DNA of Academies Press; 2017.
Healthy Adult Mice Derived from Preimplantation Blastocysts This comprehensive report followed a summit, jointly organised by the U.
Injected with Viral DNA. Proceedings of the National Academy of S. National Academy Sciences, U.S. National Academy of Medicine, the
Sciences of the United States of America 1974, 71:1250-1254. UK Royal Society and the Chinese Academy of Sciences. It summarises
the latest research being conducted with the techniques, which broadly
2. Hakimi P, Yang J, Casadesus G, Massillon D, Tolentino-Silva F, fall into three categories ‘basic research’, ‘clinical use to treat disease’,
Nye CK, Cabrera ME, Hagen DR, Utter CB, Baghdy Y et al.: and ‘germline editing. It outlines seven ethical principles which should
Overexpression of the cytosolic form of phosphoenolpyruvate guide the governance of human genome editing: promoting well-being;
carboxykinase (GTP) in skeletal muscle repatterns energy transparency; due care; respect for persons; fairness and transnational
metabolism in the mouse. J Biol Chem 2007, 282:32844-32855. cooperation. The Report is notable for not calling for an outright ban on
heritable germline changes, but instead describing 10 conditions that
3. Tang YP, Shimizu E, Dube GR, Rampon C, Kerchner GA, Zhuo M, must be met before clinical trials should be considered. It also stresses
Liu G, Tsien JZ: Genetic enhancement of learning and memory the important role in which public engagement should play in shaping the
in mice. Nature 1999, 401:63-69. development of these genome editing technologies.
4. Lim MM, Wang Z, Olazábal DE, Ren X, Terwilliger EF, Young LJ: 20. Oderberg DS: The metaphysical status of the embryo: some
Enhanced partner preference in a promiscuous species by arguments revisited. J Appl Philos 2008, 25:263-276.
manipulating the expression of a single gene. Nature 2004,
429:754-757. 21. Savulescu J, Pugh J, Douglas T, Gyngell C: The moral imperative
to continue gene editing research on human embryos. Protein
5. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Cell 2015, 6:476-479.
Charpentier E: A Programmable dual-RNA-guided DNA
endonuclease in adaptive bacterial immunity. Science 2012, 22. Ma H, Marti-Gutierrez N, Park S-W, Wu J, Lee Y, Suzuki K, Koski A,
337:816-821. Ji D, Hayama T, Ahmed R et al.: Correction of a pathogenic gene
mutation in human embryos. Nature 2017, 548:413-419.
6. Iyer V, Shen B, Zhang W, Hodgkins A, Keane T, Huang X,
Skarnes WC: Off-target mutations are rare in Cas9-modified 23. Tang L, Zeng Y, Du H, Gong M, Peng J, Zhang B, Lei M, Zhao F,
mice. Nat Methods 2015, 12:479. Wang W, Li X, Liu J: CRISPR/Cas9-mediated gene editing in
human zygotes using Cas9 protein. Mol Genet Genomics 2017,
7. Zou Q, Wang X, Liu Y, Ouyang Z, Long H, Wei S, Xin J, Zhao B,
292:525-533.
Lai S, Shen J et al.: Generation of gene-target dogs using
CRISPR/Cas9 system. J Mol Cell Biol 2015 http://dx.doi.org/ 24. See, for example FitzPatrick W: Surplus embryos,
10.1093/jmcb/mjv061. nonreproductive cloning, and the intend/foresee distinction.
8. Yang L, Güell M, Niu D, George H, Lesha E, Grishin D, Aach J, Hastings Cent Rep 2003, 33:29-36.
Shrock E, Xu W, Poci J et al.: Genome-wide inactivation of 25. Willyard C: The boom in mini stomachs, brains, breasts,
porcine endogenous retroviruses (PERVs). Science 2015 http:// kidneys and more. Nature 2015, 523:520-522.
dx.doi.org/10.1126/science.aad1191.
26. Schwank G, Koo B-K, Sasselli V, Dekkers JF, Heo I, Demircan T,
9. Niu Y, Shen B, Cui Y, Chen Y, Wang J, Wang L, Kang Y, Zhao X, Sasaki N, Boymans S, Cuppen E, van der Ent CK et al.: Functional
Si W, Li W et al.: Generation of gene-modified cynomolgus repair of CFTR by CRISPR/Cas9 in intestinal stem cell
monkey via Cas9/RNA-mediated gene targeting in one-cell organoids of cystic fibrosis patients. Cell Stem Cell 2013,
embryos. Cell 2014, 156. 13:653-658.
10. Liang P, Xu Y, Zhang X, Ding C, Huang R, Zhang Z, Lv J, Xie X,
27. Munsie M, Hyun I, Sugarman J: Ethical issues in human
Chen Y, Li Y et al.: CRISPR/Cas9-mediated gene editing in
organoid and gastruloid research. Development 2017, 144:942-
human tripronuclear zygotes. Protein Cell 2015, 6:363-372.
945.
11. International Bioethics Committee of the United Nations
Educational Scientific and Cultural Organization: Report of the IBC 28. Denker HW: Self-organization of stem cell colonies and of early
on Updating Its Reflection on the Human Genome and Human mammalian embryos: recent experiments shed new light on
Rights. 2015. http://unesdoc.unesco.org/images/0023/002332/ the role of autonomy vs. external instructions in basic body
233258E.pdf. plan development. Cells 2016, 5:E39.

12. Collins F: Statement on NIH Funding of Research Using Gene- 29. Hyun I: Engineering ethics and self-organizing models of
Editing Technologies in Human Embryos. 2015. https://www.nih. human development: opportunities and challenges. Cell Stem
gov/about-nih/who-we-are/nih-director/statements/statement- Cell 2017, 21:718-720.
nih-funding-research-using-gene-editing-technologies-human-
30. Bredenoord AL, Clevers H, Knoblich JA: Human tissues in a dish:
embryos.
the research and ethical implications of organoid technology.
13. Baltimore D, Berg P, Botchan M, Carroll D, Charo RA, Church G, Science 2017, 355:eaaf9414.
Corn JE, Daley GQ, Doudna JA, Fenner M et al.: Biotechnology. A
prudent path forward for genomic engineering and germline 31. Boers SN, van Delden JJ, Clevers H, Bredenoord AL: Organoid
gene modification. Science 2015, 348:36-38. biobanking: identifying the ethics: organoids revive old and
raise new ethical challenges for basic research and
14. Lanphier E, Urnov F, Haecker SE, Werner M, Smolenski J: Don’t therapeutic use. EMBO Rep 2016, 17:938-941.
edit the human germ line. Nature 2015, 519:410-411.
32. Naldini L: Gene therapy returns to centre stage. Nature 2015,
15. Sugarman J: Ethics and germline gene editing. EMBO Rep 2015, 526:351-360.
16:879-880.
33. Harris J: Germline manipulation and our future worlds. Am J
16. The Hinxton Group, an International Consortium on Stem Cells, Bioethics 2015, 15:30-34.
Ethics and Law: Statement on Genome Editing Technologies and
Human Germline Genetic Modification. 2015. (PDF). 34. For a good summary of some of the historical criticisms of gene
editing for reproduction; see American Association for the
17. International Society for Stem Cell Research: Guidelines for Stem Advancement of Science (2000) Human Inheritable Genetic
Cell Science and Clinical Translation. 2016. http://www.isscr.org/ Modifications — Assessing Scientific, Ethical, Religious, and
professional-resources/policy/2016-guidelines. Policy Issues (PDF).

www.sciencedirect.com Current Opinion in Genetics & Development 2018, 52:7–12


12 Cell reprogramming, regeneration and repair

35. Ishii T: The ethics of creating genetically modified children Mitochondrial_2_2_2016&utm_medium=email&utm_term=0_


using genome editing. Curr Opin Endocrinol Diabetes Obes 2017, 211686812e-fb857bd5b7-180391841.
24:418-423.
46. Wu J, Vilarino M, Suzuki K, Okamura D, Bogliotti YS, Park I,
36. Ishii T: Germline genome-editing research and its socioethical Rowe J, McNabb B, Ross PJ, Belmonte JCI: CRISPR-Cas9
implications. Trends Mol Med 2015, 21:473-481. mediated one-step disabling of pancreatogenesis in pigs. Sci
Rep 2017, 7.
37. Baylis F: Human germline genome editing and broad societal
consensus. Nat Hum Behav 2017, 1:0103. 47. Savulescu J: Human–animal transgenesis and chimeras might
be an expression of our humanity. Am J Bioethics 2003, 3:22-25.
38. Lander ES: Brave new genome. N Engl J Med 2015, 373:5-8.
39. Gyngell C, Douglas T, Savulescu J: The ethics of germline gene 48. Hyun I: What’s wrong with human/nonhuman chimera
 editing. J Appl Philos 2017, 34:498-513.  research? PLOS Biol 2016, 14:e1002535.
This article discusses the ethical arguments for and against germline gene Frames a discussion about human/nonhuman chimera research by not-
editing (GGE). If first outlines the case in favour of GGE. The authors ing that the National Institutes of Health (NIH) is poised to lift its funding
distinguish between the medical case for GGE and the research case for moratorium on research. Notes how the creation of chimeric organisms
GGE. The research case relates to the potential for GGE to be used in has been possible for decades, however recent technologies — such as
basic research, for example, to investigate the process by which embryo- blastocyst complementation — allow for acute levels of human/nonhu-
nic cells differentiate into specialised cell types. The medical case relates man mixing in stem-cell–based chimeras, which may create novel ethical
to the long-term potential of GGE to prevent disease. The article then issues. Hyun, next notes the benefits of such research, including that it
reviews the arguments that have been offered against GGE. Including that could allow human organs to be grown in others animals for transplanta-
it will used for enhancement purposes, it will have negative evolutionary tion. Hyun then discusses a risk of such research, namely that embryo
consequences, and that we lack the consent of future people who will be complementation could inadvertently affect the developing animal in
impacted by the technology. It concludes that none these arguments ways that cause it to have an ambiguous moral status, for example,
justify very strong prohibitions against GGE, which limits its use in through effecting the development of the organisations brain. This worry
research. Hyun refers to as ‘moral humanising’ of the research animal. It concludes
that ‘chimera research should prioritize animal welfare principles while at
40. Callaway E: UK moves closer to allowing “three-parent” the same time enabling scientific progress in areas of humanitarian
babies. Nature 2016 http://dx.doi.org/10.1038/ importance, albeit in a manner consistent with these principles.’.
nature.2016.21067.
49. Scheufele DA, Xenos MA, Howell EL, Rose KM, Brossard D,
41. Newson AJ, Wilkinson S, Wrigley A: Ethical and legal issues in Hardy BW: U.S. attitudes on human genome editing. Science
 mitochondrial transfer. EMBO Mol Med 2016, 8:589-591. 2017, 357:553-554.
This article follows the U.S. National Academy of Science report on
mitochondrial transfer which the article describes as ‘broadly supportive 50. Gaskell G, Bard I, Allansdottir A, da Cunha RV, Eduard P,
of MRT’. The article first describes two major techniques of MTR: Hampel J, Hildt E, Hofmaier C, Kronberger N, Laursen S,
maternal spindle transfer (MST) and pronuclear transfer (PNT). It provides Meijknecht A et al.: Public views on gene editing and its uses.
a summary of the current legal and regulatory status of MTR, noting Nat Biotechnol 2017, 35:1021-1023.
recent changes in UK law which make the practice legal. It then discusses
some of the ethical issues that MTR raises, such as whether it should be 51. Hyun I, Osbourne C: Query the merits of embryo editing for
considered a form of germline therapy, whether MTR should be limited to  reproductive research now. Nat Biotechnol 2017, 35:1023-1025.
male children, and whether mitochondria donors should be able to Letter to the editor in relation to ‘questionable value assumptions’ raised
maintain anonymity. It then argues that it is unlikely that recipients of in first report to correct gene mutation in viable human embryos. Places
MTR who suffered suboptimal health as a result of the practice, could this in context of recent NAS guidelines highlighting gaps/ambiguity in
take legal action against their providers of the technology. interpretation of three of these suggested requirements: Firstly, determin-
ing if there are reasonable alternatives; secondly, how to determine
42. Callaway E: Reproductive medicine: the power of three. Nature seriousness of disease or condition and finally, how to establish health
2014, 509:414-417. and societal benefits AND risks via broad and ongoing public engage-
ment. Noting ‘waiting until after the preclinical stages of research are well
43. Liao SM: Do mitochondrial replacement techniques affect under way bypasses discussion about the philosophical and ethical
qualitative or numerical identity? Do mitochondrial wisdom of even pursuing clinical embryo editing in the first place’.
replacement techniques affect qualitative or numerical
identity?. Bioethics 2017, 31:20-26. 52. Kang X, He W, Huang Y, Yu Q, Chen Y, Gao X, Sun X, Fan Y:
Introducing precise genetic modifications into human 3PN
44. Wrigley A, Wilkinson S, Appleby JB: Mitochondrial replacement: embryos by CRISPR/Cas-mediated genome editing. J Assist
ethics and identity. Bioethics 2015, 29:631-638. Reprod Genet 2016, 33:581-588.
45. National Academy of Sciences: Mitochondrial Replacement
53. Fogarty NME, McCarthy A, Snijders KE, Powell BE, Kubikova N,
Techniques: Ethical, Social, and Policy Considerations. 2016.
Blakeley P, Lea R, Elder K, Wamaitha SE, Kim D et al.: Genome
http://www.nationalacademies.org/hmd/reports/2016/
editing reveals a role for OCT4 in human embryogenesis.
Mitochondrial-Replacement-Techniques?utm_source=IOM
Nature 2017, 550:67-73.
+Email+List&utm_campaign=fb857bd5b7-02_03_16_

Current Opinion in Genetics & Development 2018, 52:7–12 www.sciencedirect.com

You might also like