ArticlePDF Available

Abstract

Recent studies using gene editing tools such as CRISPR/Cas9 have shown an enormously broad spectrum of possible future applications. This genetic engineering technique allows researchers to easily disable, insert, or replace DNA fragments, while raising risks such as incomplete or inaccurate editing, off-target mutations, and other unintended consequences. Whereas there is consensus that possible future uses in somatic cells hold enormous promise in clinical contexts, human germline interventions, i.e., the genetic modification of human gametes or embryos, are highly controversial (Baltimore et al., 2015; Lander, 2015; Lanphier et al., 2015; Miller, 2015; Pollack, 2015; Savulescu et al., 2015).
OPINION
published: 09 May 2016
doi: 10.3389/fgene.2016.00081
Frontiers in Genetics | www.frontiersin.org 1May 2016 | Volume 7 | Article 81
Edited by:
Dov Greenbaum,
Yale University, USA
Reviewed by:
Amy L. Fletcher,
University of Canterbury, New Zealand
Nanibaa’ A. Garrison,
Seattle Children’s Research Institute,
USA
Bettina Bock Von Wülfingen,
Humboldt-Universität zu Berlin,
Germany
*Correspondence:
Elisabeth Hildt
ehildt@iit.edu
Specialty section:
This article was submitted to
ELSI in Science and Genetics,
a section of the journal
Frontiers in Genetics
Received: 24 February 2016
Accepted: 20 April 2016
Published: 09 May 2016
Citation:
Hildt E (2016) Human Germline
Interventions–Think First.
Front. Genet. 7:81.
doi: 10.3389/fgene.2016.00081
Human Germline Interventions–Think
First
Elisabeth Hildt *
Center for the Study of Ethics in the Professions, Illinois Institute of Technology, Chicago, IL, USA
Keywords: germline, gene editing, CRISPR, gene therapy, human embryo, ethics, societal implications, ELSI
Recent studies using gene editing tools such as CRISPR/Cas9 have shown an enormously broad
spectrum of possible future applications. This genetic engineering technique allows researchers to
easily disable, insert, or replace DNA fragments, while raising risks such as incomplete or inaccurate
editing, off-target mutations, and other unintended consequences. Whereas there is consensus that
possible future uses in somatic cells hold enormous promise in clinical contexts, human germline
interventions, i.e., the genetic modification of human gametes or embryos, are highly controversial
(Baltimore et al., 2015; Lander, 2015; Lanphier et al., 2015; Miller, 2015; Pollack, 2015; Savulescu
et al., 2015).
Up to now, at least in Western countries, there has been a broad consensus to
ban interventions that aim to modify the human germline. For example, the Council of
Europe’s Convention on Human Rights and Biomedicine from 1997 (http://conventions.
coe.int/Treaty/en/Treaties/Html/164.htm) reads in Article 13:
“An intervention seeking to modify the human genome may only be undertaken for preventive,
diagnostic, or therapeutic purposes and only if its aim is not to introduce any modification in the
genome of any descendants.”
In December 2015, the Council of Europe’s Committee on Bioethics adopted a Statement
on Genome Editing Technologies (https://rm.coe.int/CoERMPublicCommonSearchServices/
DisplayDCTMContent?documentId=090000168049034a) that stresses the relevance of the
Convention on Human Rights and Biomedicine with regard to gene editing techniques.
Article 1 of UNESCO’s Universal Declaration on the Human Genome and Human Rights
from 1997 states (http://portal.unesco.org/en/ev.php-URL_ID=13177&URL_DO=DO_TOPIC&
URL_SECTION=201.html):
“The human genome underlies the fundamental unity of all members of the human family, as well as the
recognition of their inherent dignity and diversity. In a symbolic sense, it is the heritage of humanity.”
This can be seen in the context of UNESCO’s Universal Declaration on Bioethics and Human
Rights (2005) (http://portal.unesco.org/en/ev.php-URL_ID=31058&URL_DO=DO_TOPIC&
URL_SECTION=201.html) which says in Article 16: “The impact of life sciences on future
generations, including on their genetic constitution, should be given due regard.”
In October 2015, a UNESCO panel of experts called for a temporary ban on
editing the human germline (http://www.unesco.org/new/en/social-and-human-sciences/
themes/bioethics/sv0/news/unesco_panel_of_experts_calls_for_ban_on_editing_of_human_dna_
to_avoid_unethical_tampering_with_hereditary_traits/#.VwgJUvkrJaQ).
Many countries ban germline genetic modification either by law or by guidelines, while other
countries are ambiguous about the legal status of germline modifications (cf. Araki and Ishii, 2014).
These regulations and agreements are to be seen against the background of a long
interdisciplinary debate on ethical and societal issues of germline interventions that began in the
Hildt Human Germline Interventions–Think First
1980s. In this debate, a broad spectrum of topics has been
discussed, including medical issues concerning safety and
efficacy, possible health benefits, risks and unintended
consequences for future generations, and ethical issues such as
individual and collective responsibility, reproductive autonomy,
human dignity, the moral status of human embryos, human
nature, tampering with human evolution, playing God, eugenics,
discrimination, human enhancement, and transhumanist
visions.
Up to a few months ago, in the absence of a technique to
modify the human germline by disabling, inserting, or replacing
DNA fragments, the debate on human germline interventions
seemed to be rather academic and theoretical, far away from
any reality. It is the advent of this new technique of gene
editing and the observation that it can in principle be used to
modify the DNA of the resulting organism and its offspring
that revitalizes the debate on germline interventions and gives
it a new direction. In this, the mere availability of a technique
that allows us to modify the human germline seems to provide
a strong push toward human germline interventions. Whereas
the plausibility or implausibility of the various arguments
in the debate around human germline interventions has not
changed, the situation around this debate is quickly evolving:
the worldwide scientific community watches an exciting scientific
breakthrough, scientists are engaging in a new field of research,
and manuscripts are appearing that put pressure on scientific
journals to publish them (Sharma and Scott, 2015). All of this
seems to develop a strong momentum of its own, resulting
in a rush toward using this new technique for germline
intervention.
Recently, a group of Chinese researchers published the
results of human germline modification experiments involving
tripronuclear zygotes, i.e., zygotes that are not capable of
developing normally (Liang et al., 2015). In December 2015, the
members of the Organizing Committee for the International
Summit on Human Gene Editing, in which leading researchers
in the field participated, published a summit statement
(http://www8.nationalacademies.org/onpinews/newsitem.aspx?
RecordID=12032015a). In this, they do not exclude in-vitro
research involving human germline interventions as long as
the modified cells are not used to establish a pregnancy, but
for the time being argue against any clinical use of germline
editing. During the past few months, also others have spoken in
favor of running in-vitro human germline genetic interventions
(Baltimore et al., 2015; Miller, 2015). Some scientists have argued
toward a general ban of human germline modifications using
the CRISPR/Cas9 technique and similar techniques, however,
(Lanphier et al., 2015; Pollack, 2015).
There is an urgent need to step back and think about
what is going on. The recent move toward running in-vitro
human germline interventions is a remarkable development.
This is particularly true as currently, in view of the high
risks and unresolved practical, societal and ethical issues
involved, nobody would ever argue toward attempting
human germline modifications that involve initiating a
pregnancy. However, the move toward in-vitro human
germline interventions attempts to set the stage for future
uses involving human reproduction. Even if in-vitro studies
involving only early stages of human embryonic development
are far less problematic from a practical and ethical point of
view than research involving the initiation of a pregnancy or
the birth of a child, they are still highly problematic. In-vitro
research with early human embryos clearly is not done in
a vacuum but has to be seen in the broader context of the
attempt to find possible future applications involving human
reproduction.
THERE ARE NO NEW POWERFUL
ARGUMENTS IN FAVOR OF HUMAN
GERMLINE INTERVENTIONS
Are there any new powerful arguments in favor of human
germline interventions? I do not see any.
The main argument put forward in favor of clinical germline
interventions is that they may serve to prevent diseases and avoid
possible suffering in future generations. It has been proposed
that germline interventions might be an appropriate tool for
the prevention of monogenic diseases in situations in which
there is a 100% probability that the disease will be passed on to
the progeny. This is the case when one parent is homozygous
for a dominant disease or both parents are homozygous for
an autosomal recessive disease. Sickle cell anemia or recessive
deafness would be examples of the latter (Wivel and Walters,
1993; Lander, 2015; Miller, 2015). However, situations like these
are very rare. It is questionable whether there would be broader
justifiable medical uses for germline interventions, especially in
view of the availability of genetic testing and pre-implantation
genetic diagnosis.
Furthermore, speculations on possible future additional
applications seem to play a considerable role in the rush toward
human germline interventions. These include the possibility
of risk reduction in widespread multigenic and multifactorial
disorders such as cardiovascular disease or cancer. However,
interactions between the numerous genetic and environmental
factors are most often very complicated so that it is questionable
whether the risks involved would ever outweigh the potential
benefits (Wivel and Walters, 1993; Lander, 2015).
In addition, germline interventions may be considered a
future option for parents who attempt to specify more than
one trait of their future offspring. It might be easier to
genetically modify the traits in question than to attempt to
select embryos with the desired traits from a limited number of
available embryos by using pre-implantation genetic diagnosis
(cf. Savulescu et al., 2015).
Beyond that, according to further speculations, germline
gene editing may render it possible to influence a broad
spectrum of traits or enhance human capabilities. With germline
genetic interventions, the choices available for future parents
would increase considerably. This opens up the debate for
speculations on improving mankind, genetic enhancements,
eugenics and designer babies, all of these clearly underlining the
manifold ethical and societal issues involved in human germline
interventions. Interestingly, with regard to human germline
Frontiers in Genetics | www.frontiersin.org 2May 2016 | Volume 7 | Article 81
Hildt Human Germline Interventions–Think First
interventions, several authors consider a development toward
genetic enhancements to be inevitable (Wivel and Walters, 1993;
Baylis and Robert, 2004).
AVOID PREMATURE EXPERIMENTS
In his article “Human gene therapy: scientific and ethical
considerations” published in 1985, W. French Anderson named
three conditions that should be met prior to any attempt to
undergo germline gene therapy in humans: Substantial previous
experience with somatic cell gene therapy in humans proving
safety and efficacy of the approach; adequate animal research
that shows the reproducibility, reliability, and safety of germline
therapeutic interventions; and the informed public approval of
the procedure (Anderson, 1985).
In spite of the fact that neither comprehensive knowledge with
gene editing of somatic cells nor with germline applications in
animal studies is available at the moment, in-vitro experiments
involving human germline modifications have already been
initiated (Liang et al., 2015)1. Whereas in-vitro studies are far
less problematic than studies using gene editing in reproductive
contexts, it should not be forgotten that in-vitro studies involving
human embryos bear considerable practical and ethical issues
when it comes to the creation and destruction of human embryos
for research purposes or to the use of surplus in-vitro fertilization
(IVF) embryos. For the time being, considering the moral status
of human embryos and the lack of adequate experience with gene
editing tools, in-vitro human germline interventions involving
human embryos can hardly be justified.
Furthermore, before overriding general standards that have
been agreed upon widely in Western societies, it will be absolutely
1Albeit in this particular study, the tripronuclear zygotes involved were not capable
to develop normally.
necessary to thoroughly reflect on the issues involved. For this, a
broad societal debate on the practical, societal, and ethical issues
in human germline interventions involving the various groups
within society is urgently needed. Beyond an interdisciplinary
academic debate involving disciplines such as medicine, natural
sciences, humanities, social sciences and the law, it will be
particularly important for scientists to inform the public of the
ongoing development and to engage in a serious dialog with
critical stakeholders.
Only widely held and well informed public approval can
legitimize researchers to go on in a sensitive field like this which
has the power to affect society as a whole. For the time being,
however, not much is known on the opinion of the public. Up
to now, the controversy on gene editing and human germline
interventions is dominated by scientists involved in the field. The
general public, it seems, has difficulties keeping abreast with the
quick development and understanding the issues at stake.
For sure, a balanced debate that involves a broad spectrum of
the various societal groups will have an open outcome. It may be
the case that societal values change over time, that the balancing
of pros and cons is done differently now than it was done in the
past, or that new and important arguments in favor of or against
germline interventions will come up. But all of this needs careful
and thorough reflection.
In view of the unresolved practical, ethical and societal issues,
for the time being, it is more than advisable to refrain from any
experiments involving human germline interventions. Instead:
Think first. Don’t hastily override existing standards broadly
agreed upon.
AUTHOR CONTRIBUTIONS
The author confirms being the sole contributor of this work and
approved it for publication.
REFERENCES
Anderson, W. F. (1985). Human gene therapy: scientific and ethical considerations.
J. Med. Philos. 10, 275–291. doi: 10.1093/jmp/10.3.275
Araki, M., and Ishii, T. (2014). International regulatory landscape and integration
of corrective genome editing into in vitro fertilization. Reprod. Biol. Endocrinol.
12:108. doi: 10.1186/1477-7827-12-108
Baltimore, D., Berg, P., Botchan, M., Carroll, D., Charo, R. A., Church, G., et al.
(2015). A prudent path forward for genomic engineering and germline gene
modification. Science 348, 36–38. doi: 10.1126/science.aab1028
Baylis, F., and Robert, J. S. (2004). The inevitability of genetic enhancement
technologies. Bioethics 18, 1–26. doi: 10.1111/j.1467-8519.2004.00376.x
Lander, E. S. (2015). Brave new genome. N. Engl. J. Med. 373, 5–8. doi:
10.1056/NEJMp1506446
Lanphier, E., Urnov, F., Haecker, S. E., Werner, M., and Smolenski, J. (2015). Don’t
edit the human germ line. Nature 519, 410–411. doi: 10.1038/519410a
Liang, P., Xu, Y., Zhang, X., Ding, C., Huang, R., Zhang, Z., et al. (2015).
CRISPR/Cas9-mediated gene editing in human tripronuclear zygotes. Protein
Cell 6, 363–372. doi: 10.1007/s13238-015-0153-5
Miller, H. I. (2015). Germline gene therapy: we’re ready. Science 348:1325. doi:
10.1126/science.348.6241.1325-a
Pollack, R. (2015). Eugenics lurk in the shadow of CRISPR. Science 348:871. doi:
10.1126/science.348.6237.871-a
Savulescu, J., Pugh, J., Douglas, T., and Gyngell, C. (2015). The moral imperative
to continue gene editing research on human embryos. Protein Cell 6, 476–479.
doi: 10.1007/s13238-015-0184-y
Sharma, A., and Scott, C. T. (2015). The ethics of publishing human germline
research. Nat. Biotechnol. 33, 590–592. doi: 10.1038/nbt.3252
Wivel, N. A., and Walters, L. (1993). Germ-line gene modification and disease
prevention: some medical and ethical perspectives. Science 262, 533–538. doi:
10.1126/science.8211180
Conflict of Interest Statement: The author declares 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 © 2016 Hildt. 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) or licensor
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 Genetics | www.frontiersin.org 3May 2016 | Volume 7 | Article 81
... The possibility of analysing the genetically modified embryo is limited, since this analysis would have to be carried out during the subsequent in vitro culture period; this would impose limitations in terms of both technique and time. In addition, reducing the usable embryos to those obtained from in vitro fertilization processes, as opposed to being able to specifically create embryos for research, would reduce the quantity and quality of the embryos available 4 [23]. ...
... Hildt [23] argued that any attempt to genetically modify the germline should meet three conditions that do not tend to be met at present: 1. Previous solid and safe experience in gene therapy with somatic cells 2. Tests on animals that would guarantee safety and reproducibility and suggest that any subsequent human interventions would be successful 3. Public approval (social, political…) of the technique ...
... Hence, in July 2017, Chneiweiss, Hirsch, Montoliu and other researchers (representing over 20 institutions) proposed creating a European Steering Committee to analyse and evaluate the advantages and dangers of the new technique[23]. ...
Article
Full-text available
The field of genetics has seen major advances in recent decades, particularly in research, prevention and diagnosis. One of the most recent developments, the genomic editing technique Clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9, has opened the possibility for genetic therapies through genome modification. The technique marks an improvement on previous procedures but poses some serious ethical conflicts. Bioethics is the discipline geared at finding answers to ethical challenges posed by progress in medicine and biology and examining their repercussions for society. It can also offer a conceptualization of these ethical dilemmas. The aim of this paper is to offer a map of the ethical dilemmas associated with this technique by way of a critical analysis of current literature. The main issues can be grouped in four areas: efficacy and security; the types of cells which can be targeted by the technique (somatic, embryonic and gametes); the goal of the therapy; and accessibility and justice.
... However, this technology is not available clinically due to an international moratorium based on ethical and safety concerns. These concerns relate to the moral status of the embryos and the potential for permanent modification on the germline to affect future generations, as well as off-target implications whereby germline editing results in unplanned, harmful changes (Greely, 2019;Hildt, 2016;Ormond et al., 2017). ...
... Germline genome editing remains technically and ethically challenging, and most countries ban germline genetic modification either by law or by guidelines. [43] Somatic gene therapy, on the other hand, led to significant breakthroughs in many different clinical applications in PGDs. This review will focus on somatic gene therapy, with a discussion of gene therapy strategies and the associated risks, recent advances in gene therapy for paediatric genetic disorders and challenges of gene therapy in precision medicine [ Table 2]. ...
Article
Full-text available
There are more than 7,000 paediatric genetic diseases (PGDs) but less than 5% have treatment options. Treatment strategies targeting different levels of the biological process of the disease have led to optimal health outcomes in a subset of patients with PGDs, where treatment is available. In the past 3 decades, there has been rapid advancement in the development of novel therapies, including gene therapy, for many PGDs. The therapeutic success of treatment relies heavily on knowledge of the genetic basis and the disease mechanism. Specifically, gene therapy has been shown to be effective in various clinical trials, and indeed, these trials have led to regulatory approvals, paving the way for gene therapies for other types of PGDs. In this review, we provide an overview of the treatment strategies and focus on some of the recent advancements in therapeutics for PGDs.
... However, the application of the CRISPR/Cas9 technology in editing the human embryo has revived the debate about germline modification that began decades ago. Previously, the debate was based on speculative assumptions, but the ethical issues raised are nonetheless similar (Baumann 2016;Hildt 2016). Human germline gene editing raises more complex issues than somatic gene editing because it would result in heritable changes to the human genome. ...
Article
This paper describes maslahah, a fundamental concept in Islam and its application in deliberating permissibility of human germline gene editing from an Islamic perspective. This paper refers to He Jiankui’s research that led to the birth of the world’s first gene edited babies, who were edited to be protected from HIV. The objective, procedure, and output of the research were assessed against the conditions of maslahah. It can be concluded that the experiment did not meet the conditions; it is inconsistent with the objectives of shariah (maqasid al-shariah) and some fundamental Islamic teachings that it did not preserve greater benefit, hence it could be considered impermissible.
... The Ethics of Gene Editing from an Islamic Perspective… heritage of humanity and in 2005 UNESCO stated in its Article 16 that "The impact of life sciences on future generations, including on their genetic constitution, should be given due regard" and in 2015 again confirmed through its advisory panel for a temporary ban on any germline genetic modifications (Hildt 2016; Universal Declaration on Bioethics and Human Rights 2005). ...
Article
Full-text available
In light of the development of “CRISPR” technology, new promising advances in therapeutic and preventive approaches have become a reality. However, with it came many ethical challenges. The most recent worldwide condemnation of the first use of CRISPR to genetically modify a human embryo is the latest example of ethically questionable use of this new and emerging field. Monotheistic religions are very conservative about such changes to the human genome and can be considered an interference with God’s creation. Moreover, these changes could cause perpetual changes to future generations. The Muslim scholars establish their decisions by addressing five foundations of Islamic law i.e. “maqāṣid al sharı̄`a”; the purposes of the law. These are dın̄ (religion), nafs (life), nasl (progeny), `aql (intellect) and māl (wealth). To achieve this, the five principles should all be met before approval of an experiment like the Chinese embryo modifications; Qaṣd (intention) which is achieved in this case as it aims to protect the embryo from HIV. Yaqın̄ (certainty) and Ḍarar (injury) were not satisfied as they require strong scientific certainty of the procedures, and evidence of safety. Ḍarūra (necessity) by which the alternatives being compared; in this case more established and proven safe alternatives to protect the HIV transmission from the father are available, so this principle is not met. The final principle is `Urf (custom), by which the social context of using any contemporary technology should be taken in consideration, and clearly this was not achieved. Collectively, germline changes are rejected from an Islamic perspective until the five principles are fulfilled. In the Chinese Twins gene editing case, there was clearly no justification or support for it according to the Muslim Jurisprudence laws. These laws and approaches can serve as an ethical checklist for such controversial research, especially in early stages of the research.
Article
Full-text available
Abstract: The concept of gene therapy has been around for over a hundred years. However, the research and development necessary to prove the efficacy and safety of gene therapy are tremendously time-consuming and require huge investments. Moreover, the application of gene therapy depends on the management techniques of genetic materials, which have advanced rapidly just recently. The history of clinical trials on gene therapy from the past to the present reveals both successes and failures. Correspondingly, a clinical trial on gene therapy has to be approved by a scientific review committee and an ethics committee to ensure that the benefits outweigh the risks. At present, there are several approved gene therapy products for the treatment of some diseases, while many products are currently undergoing late-phase clinical trials or waiting for approval. The development of gene therapy is adapted accordingly to innovations such as CRISPR/Cas9 gene editing that enable the utilization and modernization of gene therapy. In this review, we describe the concepts and principles of gene therapy, namely, the definition, key events in the history of gene therapy, therapeutic indications, type of gene therapy, strategy of gene therapy, ex vivo and in vivo gene therapy, delivery systems, immune responses, and adverse drug reactions, and lastly, we address some concerns about ethical issues.
Article
Full-text available
Produkty lecznicze terapii zaawansowanych (ATMP) dają nadzieję na uzyskanie korzyści zdrowotnych w tych wszystkich sytuacjach, w których tradycyjne metody terapii zawodzą lub, z różnych przyczyn, nie mogą być stosowane. Zasadniczym celem niniejszego artykułu jest analiza pojęcia innowacyjności używanego w odniesieniu do biotechnologii wykorzystywanych w produkcji ATMP. Analiza tego pojęcia pozwoli wyodrębnić pięć głównych kontekstów znaczeniowych, które konstytuują jego sposób rozumienia: zmiana sposobu myślenia o dostępnym spektrum procedur medycznych, krótki czas rozpoznania technologii w modelach eksperymentalnych i teoretycznych, brak danych klinicznych dotyczących efektów stosowanych technologii w terapiach pacjentów, zwiększenie złożoności technologi oraz zwiększenie różnorodności stosowanych w tych technologiach substratów. Przeprowadzona analiza wskazuje na konieczność zachowania daleko idącej ostrożności w ocenie bezpieczeństwa terapii ATMP, w których wykorzystuje się innowacyjne technologie.
Chapter
Contemporary techniques in reproductive medicine, such as in vitro fertilization (IVF) and preimplantation genetic testing (PGT), have enabled fertilization, and the selection and transfer of embryos, without temporal and spatial restrictions. These powerful technologies have been used for both medical and non-medical reasons by couples hoping to build a family, and in the process, they have engendered tremendous controversy worldwide. The major points of contention are potential transgressions of religion, health risks to the resultant children, questions about the balance between the reproductive autonomy of parents and the welfare of children, and reluctance to accept genetic unrelatedness in the family system. For countries, it has often been difficult to discuss these points, because individuals in such countries tend to have widely varying views regarding the moral status of human prenatal lives, and they often lack sufficient knowledge of reproductive medicine. However, lessons learned from past debates illuminate an important path for regulating the new reproductive techniques with higher uncertainty. Before enacting legislation around these powerful methods, the people must weigh the likely benefits and inevitable risks to the future children, the prospective parents, and the society at large. Of course, it is challenging for most countries to reach a social consensus on reproductive medicine. However, each country must steadily tread the path to as broad a consensus as possible in order to make a socially responsible decision and empower prospective parents. This, in turn, will lead to effective enforcement of the legislation governing these powerful but nascent reproductive technologies.
Chapter
Recent progressAuthenticity in biomedical research in the fields of stem cell technology and genome editing has entailed a revival of ethical concerns regarding the autonomyAutonomy and authenticityAuthenticity of future persons who might be created by prospective novel means of assisted reproductive technologies (ART), e.g., on the basis of in vitro gametogenesis or germline interventionsGermline intervention. In this regard, critical authors refer to the HabermaseanHabermas, Jürgen concern that persons “made” by technological means instead of coming into existence in a “natural” way could be deprived of forming a self-concept of being autonomous and the authentic “authors of their life histories”. Since the recent birth of two genetically modified girls, these concerns have gained topicality. The notion of being the “author” of one’s life history appears to relate to the idea that we can—somehow and within certain limits—actively influence who we are. It is hard to see why this idea could not include the possibility of integrating the circumstances of one’s coming into existence—be they “artificial” or “natural”—in anAuthenticity authenticity-preserving way. Given that (future) persons are (will be) aware of these circumstances, they should be able to adapt their life histories accordingly. Assuming a relational structure of autonomyAutonomy and authenticityAuthenticity in general, it appears plausible that no person’s life history and self-concept are constituted and maintained in an isolated and individualistic manner. Quite the contrary, (self-)ascriptions of autonomyAutonomy and authenticityAuthenticity require adequate social relationships, in the first place. Based on this assumption, it can be supposed that deprivation of a confident self-concept and feeling of “objectification” by their “creators” in (future) persons conceived by means of novel ART would be due to flawed communicative relationships or prejudice rather than an actual lack of essentialist authenticityAuthenticity. The HabermaseanHabermas, Jürgen concern can thus be regarded as revealing a structural social problem rather than one raised by biotechnology per se, and it can be smoothed out by Habermas’Habermas, Jürgen own conceptual conditions for the development of personal identityPersonal identity, authenticityAuthenticity and autonomyAutonomy.
Article
Full-text available
The publication of the first study to use gene editing techniques in human embryos (Liang et al., 2015) has drawn outrage from many in the scientific community. The prestigious scientific journals Nature and Science have published commentaries which call for this research to be strongly discouraged or halted all together (Lanphier et al., 2015; Baltimore et al., 2015). We believe this should be questioned. There is a moral imperative to continue this research.
Article
Full-text available
A recent paper describes the use of CRISPR-Cas9 to modify the endogenous beta-globin gene (HBB) in single-celled, non-viable, triploid human zygotes—byproducts of in vitro fertilization (IVF) created due to fertilization of single eggs with two human sperm1. The research, published in the journal Protein & Cell, provoked a firestorm of controversy, reigniting old fears about gene therapy and raising new ethical concerns about human germline modification and designer babies. With clearly stated therapeutic goals, the scientists targeted endogenous HBB, the mutated form of which causes beta-thalassemia. The research also probed questions about safety and efficacy. The Chinese research team reported three major findings, all which had been previously reported in animal studies: first, low overall editing efficiency, with only 4/28 embryos repaired using the guide template; second, genetic mosaicism, resulting from embryonic cell division before the corrections are complete; and importantly, the detection of a number of off-target mutations. Precision gene editing has been hailed as a major improvement on first-generation gene transfer technologies using viral vectors. Early gene transfer studies were inefficient, failed to persist in host cells, had short-lived transgene expression and suffered from off-target editing. The HBB/CRISPR-Cas9 result suggests that dramatic improvements in gene targeting fidelity and specificity are needed before the system can be used safely for therapies.
Article
Full-text available
![Figure][1] Eugenics on the horizon?PHOTO: LAWRENCE LAWRY / SCIENCE SOURCE IN CALLING THEIR Perspective “A prudent path forward for genomic engineering and germline gene modification” (3 April, p. [36][1]; published online 19 March), D. Baltimore et al. show at once the size of the
Article
Full-text available
Genome editing tools such as the clustered regularly interspaced short palindromic repeat (CRISPR)-associated system (Cas) have been widely used to modify genes in model systems including animal zygotes and human cells, and hold tremendous promise for both basic research and clinical applications. To date, a serious knowledge gap remains in our understanding of DNA repair mechanisms in human early embryos, and in the efficiency and potential off-target effects of using technologies such as CRISPR/Cas9 in human pre-implantation embryos. In this report, we used tripronuclear (3PN) zygotes to further investigate CRISPR/Cas9-mediated gene editing in human cells. We found that CRISPR/Cas9 could effectively cleave the endogenous β-globin gene (HBB). However, the efficiency of homologous recombination directed repair (HDR) of HBB was low and the edited embryos were mosaic. Off-target cleavage was also apparent in these 3PN zygotes as revealed by the T7E1 assay and whole-exome sequencing. Furthermore, the endogenous delta-globin gene (HBD), which is homologous to HBB, competed with exogenous donor oligos to act as the repair template, leading to untoward mutations. Our data also indicated that repair of the HBB locus in these embryos occurred preferentially through the non-crossover HDR pathway. Taken together, our work highlights the pressing need to further improve the fidelity and specificity of the CRISPR/Cas9 platform, a prerequisite for any clinical applications of CRSIPR/Cas9-mediated editing. Electronic supplementary material The online version of this article (doi:10.1007/s13238-015-0153-5) contains supplementary material, which is available to authorized users.
Article
Full-text available
Heritable human genetic modifications pose serious risks, and the therapeutic benefits are tenuous, warn Edward Lanphier, Fyodor Urnov and colleagues.
Article
Full-text available
Genome editing technology, including zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and clustered regularly interspaced short palindromic repeat (CRISPR)/Cas, has enabled far more efficient genetic engineering even in non-human primates. This biotechnology is more likely to develop into medicine for preventing a genetic disease if corrective genome editing is integrated into assisted reproductive technology, represented by in vitro fertilization. Although rapid advances in genome editing are expected to make germline gene correction feasible in a clinical setting, there are many issues that still need to be addressed before this could occur. We herein examine current status of genome editing in mammalian embryonic stem cells and zygotes and discuss potential issues in the international regulatory landscape regarding human germline gene modification. Moreover, we address some ethical and social issues that would be raised when each country considers whether genome editing-mediated germline gene correction for preventive medicine should be permitted. Electronic supplementary material The online version of this article (doi:10.1186/1477-7827-12-108) contains supplementary material, which is available to authorized users.
Article
Recently, Chinese researchers attempted to modify embryos using “germline gene therapy.” If successful, this technique would create a heritable change and affect future generations, were those embryos to be used ([ 1 ][1]). A firestorm in the scientific community followed, with some researchers
Article
Fifty years ago, microbiologists sparked the recombinant-DNA revolution with the discovery that bacteria have innate immune systems based on restriction enzymes. These enzymes bind and cut invading viral genomes at specific short sequences, and scientists rapidly repurposed them to cut and paste DNA in vitro - transforming biologic science and giving rise to the biotechnology industry. Ten years ago, microbiologists discovered that bacteria also harbor adaptive immune systems, and subsequent progress has been breathtakingly rapid.(1) Between 2005 and 2009, microbial genetic studies conducted by the laboratories of Mojica, Jansen, Koonin, Horvath, van der Oost, Sontheimer, Marraffini, and others revealed that . . .
Article
A framework for open discourse on the use of CRISPR-Cas9 technology to manipulate the human genome is urgently needed. Copyright © 2015, American Association for the Advancement of Science.
Article
The term ‘gene therapy’ encompasses at least four types of application of genetic engineering for the insertion of genes into humans. The scientific requirements and the ethical issues associated with each type are discussed. Somatic cell gene therapy is technically the simplest and ethically the least controversial. The first clinical trials will probably be undertaken within the next year. Germ line gene therapy will require major advances in our present knowledge and it raises ethical issues that are now being debated. In order to provide guidelines for determining when germ line gene therapy would be ethical, the author presents three criteria which should be satisfied prior to the time that a clinical protocol is attempted in humans. Enhancement genetic engineering presents significant, and troubling, ethical concerns. Except where this type of therapy can be justified on the grounds of preventive medicine, enhancement engineering should not be performed. The fourth type, eugenic genetic engineering, is impossible at present and will probably remain so for the foreseeable future, despite the widespread media attention it has received.