Abstract
With the emergence of CRISPR technology, targeted editing of a wide variety of genomes is no longer an abstract hypothetical, but occurs regularly. As application areas of CRISPR are exceeding beyond research and biomedical therapies, new and existing ethical concerns abound throughout the global community about the appropriate scope of the systems’ use. Here we review fundamental ethical issues including the following: 1) the extent to which CRISPR use should be permitted; 2) access to CRISPR applications; 3) whether a regulatory framework(s) for clinical research involving human subjects might accommodate all types of human genome editing, including editing of the germline; and 4) whether international regulations governing inappropriate CRISPR use should be crafted and publicized. We conclude that moral decision making should evolve as CRISPR science advances and hold that it would be reasonable for national and supranational legislatures to consider evidence-based regulation of certain CRISPR applications for the betterment of human health and progress.
Keywords: CRISPR-Cas9, research ethics, genome editing, genetic engineering, research involving human subjects, bioengineering
I. Introduction
The CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats)-Cas9 (CRISPR-associated protein 9) system (“CRISPR” or “the system”) is the most versatile genomic engineering tool created in the history of molecular biology to date. This system’s ability to edit diverse genome types with unprecedented ease has caused considerable excitement and alarm throughout the international biomedical community.
CRISPR appears to offer considerable promise in a wide variety of disease contexts. For example, around the world at least 15 clinical trials— focused on multiple myeloma, esophageal, lung, prostate, and bladder cancer, solid tumors, melanoma, leukemia, human papilloma virus, HIV-1, gastrointestinal infection, β-thalassemia, sickle cell disease, and other diseases— involving CRISPR applications have been developed1–3. Moreover, as of May, 2018, in China at least 86 individuals have had their genes altered as part of clinical trials4.
While significant public support exists for therapeutic applications5, ethical (moral) and safety concerns about certain areas of CRISPR applications, such as germline editing, are apparent around the world6. Notably, such discussions commenced during the Napa Valley meeting of 2015 when a leading group of CRISPR-Cas9 developers, scientists, and ethicists met to examine the biomedical, legal, and ethical aspects of CRISPR systems7. From this meeting, more extensive deliberations were solicited, and the United States (U.S.) National Academies of Sciences, Engineering, and Medicine (NASEM or “The Committee”) invited the Chinese Academy of Sciences and the United Kingdom’s (U.K) Royal Society to participate in the International Summit on Human Gene Editing8. The goal of this meeting was to examine when, where, and how the technology might be applied in humans. This discussion continued in February of 2017 when a multidisciplinary committee of the NASEM published a comprehensive report examining numerous aspects of human genome editing9.
To date, the NASEM report provides perhaps the most influential, extensive analysis examining wide-ranging concerns about human genome editing10. Importantly, the Committee favored somatic genome editing, but did not permit genomic modification for any kind of enhancement9,11. Also, though impermissible at present, the Committee concluded that human heritable genome editing, the modification of the germline with the goal of creating a new person who could potentially transfer the genomic edit to future generations, would be permissible under certain conditions: “In light of the technical and social concerns involved… heritable genome-editing trials might be permitted, but only following much more research aimed at meeting existing risk/benefit standards for authorizing clinical trials and even then, only for compelling reasons and under strict oversight…”9 Although by law U.S. federal funding cannot be used to support research involving human embryos12–14, the NASEM report suggests that when technical and safety risks are better understood, then clinical trials involving germline editing in human subjects might begin9.
In this review, we aim to summarize fundamental ethical concerns about CRISPR use in general, but the list is not exhaustive. First, we briefly review CRISPR systems and their applications in editing genomes and epigenomes. Second, we describe how complexities of CRISPR science affect those of CRISPR ethics and vice versa. Third, we assess several key ethical considerations. Notably, although some of the ethical concerns are specific to CRISPR technology, many, such as research on human embryos, have been debated long before CRISPR15. Moreover, since CRISPR is still a maturing technology, novel applications in the future may raise new ethical quandaries meriting further attention and dissection. Fourth, it is important to point out that, though morality and law often overlap, significant differences exist. Even though law may affect ethics and vice versa, this discussion centers mostly on ethics. Finally, while discussing these issues, we assume no position on any topic; our account is merely descriptive. Therefore, we make no attempt to settle the aforementioned issues.
II. CRISPR systems and their uses
Different CRISPR systems in genome editing
CRISPR is a natural bacterial defense system against invading viruses and nucleic acids. Over billions of years, multiple CRISPR-type immune systems have evolved. Naturally occurring CRISPR systems are typically classified by their repertoires of CRISPR-associated (cas) genes, which are often found adjacent to the CRISPR arrays16,17. Although the characterization is yet to be finalized, two major classes of CRISPR-Cas adaptive immune systems have been identified in prokaryotes18–20. This division is based on the organization of effector modules. Class 1 CRISPR-Cas systems employ multi-protein effector complexes and encompass 3 types (I, III, and IV). By contrast, Class 2 systems utilize single protein effectors and encompass 3 types (II, V, VI). Although various natural CRISPR-Cas systems have been repurposed for genome editing, due to its robust gene-editing efficiency and broader genome-targeting scope owing to its simple NGG PAM sequence requirement, the Cas9 from S. pyogenes (spCas9) is currently the most commonly used CRISPR-Cas9 protein. It is worth noting that multiple efforts are underway to discover novel Cas9 variants or re-engineer the existing Cas9 proteins, which will have less dependence on the stringent PAM-sequence requirement21,22.
CRISPR goes beyond genome editing
The DNA-editing capacity of CRSPR-Cas9 is due to the ability of the WT Cas9 protein to cause double-stranded breaks at the target site that is determined by the custom-designed short guiding RNA (sgRNA)23. The repair of DNA breaks frequently results in indels, due to the non-homologous end joining (NHEJ) repair mechanism. However, when a complementary template is available, homology-directed repair (HDR) machinery can use it and thereby achieve more precise gene editing. Notably, a single point mutation in either of the two catalytic domains of Cas9 results in a nickase Cas9 (nCas9), whereas mutations in both domains (D10A and H840A for spCas9) diminish Cas9’s catalytic activity, resulting in dead Cas9 (dCas9)24. Interestingly, the application areas of modified Cas9 proteins are exceeding that of WT Cas925. Such uses are largely possible because the nCas9 or dCas9 can still be guided to the target sequence26. Researchers employed these Cas9 variants for unique purposes. For example, tandem targeting of nCas9 has been utilized to improve targeting specificity27,28. More recently, this enzyme has been used as the base platform for second generation genome-editing tools called “base editors”29,30. Base-editing technology employs cytidine or adenine deaminase enzymes to achieve the programmable conversion of one base into another (C to T or A to G). Most importantly, the targeted base transition happens without DNA double-stranded breaks29,31. We recently utilized this technology to edit the universal genetic code and introduced a “stop” codon in the genes32.
In addition to nCas9, researchers utilized the guidable capacity of dCas9 as a platform to recruit various effector proteins to a specific locus in living cells. Generally, these activities can be classified as epigenetic editing (to alter locus-specific epigenetic information), gene regulation (to turn the activities of single or multiple genes on or off), chromatin imaging (to label and monitor chromatin dynamics in living cells), and manipulation of chromatin topology (to alter 3D chromatin structure in the nuclear space)33.
CRISPR research is progressing at a rapid pace. Recently, scientists have also uncovered new CRISPR-Cas systems (Cas13) that can target RNA instead of DNA34,35. By enabling targeted RNA recognition and editing, these newer RNA-targeting CRISPR tools have their unique applications ranging from biomedical and biotechnological to the detection of nucleic acids36,37. Although many ethical concerns are related to the catalytic activities of WT Cas9—partly because it permanently alters the genetic information—some of these activities of catalytically inactive dCas9, nickase-Cas9-based platforms, such as base editors and recently discovered RNA-targeting Cas proteins, may raise comparable moral issues depending on the duration of the exerted effect and the purpose of the experiments. Detailed discussion of such issues, however, is beyond the scope of this review.
III. CRISPR ethics and science: Uncomfortable bedfellows
Moral decisions, especially in biomedicine, are empirically informed and involve assessing potential risk-benefit ratios—attempting to maximize the latter while minimizing the former. To navigate ethical decision making, it is critical to consider the range of possible outcomes, the probabilities of each instantiating, and the possible justifications driving the results of any one. The ethical concerns about CRISPR genome engineering technology are largely due to at least three important reasons.
Firstly, there are concerns about the power and technical limitations of CRISPR technology. These include the possibilities of limited on-target editing efficiency38,39, incomplete editing (mosaicism)40,41, and inaccurate on- or off-target editing42,43. These limitations have been reported in CRISPR experiments involving animals and human cell lines. However, the technology is evolving at an unprecedented pace. As more efficient and sensitive CRISPR tools are developed, many of these concerns may become obsolete. Yet for the sake of this review, we mention these limitations as one of the principal worries about CRISPR. The second concern is about the future of the modified organisms: whether they will be affected indefinitely and whether the edited genes will be transferred to future generations, potentially affecting them in unexpected ways. Combined with previously mentioned technical limitations and the complexities of biological systems, making precise predictions about the future of an edited organism and gauging potential risks and benefits might be difficult, if not impossible. Thus, uncertainty resulting from these factors hinders accurate risk/benefit analysis, complicating moral decision making.
Finally, the skeptical view is that even if the genome is edited as expected and the desired functional output is achieved at the time, the complex relationship between genetic information and biological phenotypes is not fully understood. Therefore, the biological consequence of editing a gene in germline and/or somatic cells may be unclear depending on the context. Many biological traits are determined by the complex regulatory actions of numerous genes. Hence, is it difficult, if not impossible, to “design” a biological phenotype at the whole-organism level. Across biological outcomes, whether in normal or in disease development, it is uncommon that a single gene is the only factor shaping a complex biological trait. Other genetic regulatory factors such as additional genes or distal regulatory elements (e.g., enhancer or repressor elements), as well as environmental and epigenetic factors, contribute to the emergence of a biological phenotype. To argue that modifying a gene changes a desired phenotype (under certain conditions) implies at least a reasonable understanding of other independent variables contributing to the phenotype’s instantiation. But this understanding is still far from complete in many normal and disease processes44,45. Given the uncertainty regarding how gene expression and modification influence complex biological outcomes, it is difficult to appraise potential risk and benefit. This ambiguity creates a challenge on its own and is one of the sources obscuring efficient ethical deliberation and decision making.
Nevertheless, regulations governing cellular- and gene-therapy research may facilitate the safe development and oversight of some clinical trials involving CRISPR-based-editing applications. In the United States, for instance, cellular- and gene-therapy products, including many CRISPR applications, at this time are defined generally by the Food and Drug Administration (FDA) as biological products and are regulated by the FDA’s Center for Biologics Evaluation and Research (CBER)/Office of Cellular, Tissue, and Gene Therapies (OCTGT)46–48. Even though the risks and benefits of many such therapies increasingly are better understood49, questions regarding safety and efficacy remain. Thus, future advancements will continue to improve the benefits of this revolutionary technology, while minimizing the potential risks. Regardless of the uncertainty posed by novel CRISPR technologies and applications, in several locations around the world significant regulatory frameworks exist by which risks may be monitored and contained. However, wherever such infrastructure and oversight are lacking, safety and privacy risks—due to lack of regulatory oversight, monitoring, and the possibility of data breaches— might increase.
IV. Ethical concerns
To what extent should CRISPR experimentation be permitted in basic and pre-clinical biomedical research?
Though it is less than a decade old, CRISPR-Cas9 has demonstrated unprecedented potential to revolutionize innovation in basic science. From viruses and bacteria50,51, to simple model organisms, such as Drosophila melanogaster (fruit fly)52, Anopheles gambiae (mosquito)53, Saccharomyces cerevisiae (budding yeast)54, Hydra magnipapillata (hydra)55, Caenorhabditis elegans (round worm)56, Danio rerio (zebra fish)57, and Arabidopsis thaliana (rockcress)58, to larger animals such as pigs59, cattle60, and monkeys61, and even human zygotes62,63, CRISPR experimentation has led to novel, important findings. Such benefits include at least the following: increased overall efficiency in gene editing compared with previous genomic engineering techniques like transcription activator-like effector nucleases (TALENs) and zinc finger nucleases (ZNFs)64; significant insights into the evolutionary transformation of fish fins into tetrapod limbs65; investigation into new organisms66; genetic and epigenetic screens67,68; the creation of novel cell lines69; high-throughput screens and libraries70; the elucidation of novel genomic and epigenomic regulatory pathways71,72; insights into the development of butterfly coloring and patterning73; the functional characterization of key genes and molecular signaling pathways74,75; and drug-targeting screens76,77. Data from such experimentation provide essential clues and understanding that promote biomedical discovery, advancement, and the basis for potential medical benefits.
One of the major controversies about CRISPR technology emerges from its potential application in human embryos. This controversy is not about CRISPR itself, but instead is largely due to the lack of clarity about the status of the human embryo. Although some in the scientific community maintain that it is ethically impermissible to experiment on human embryos after 14 days78,79, it is impossible for any one party—whether it is a government, laboratory, funding agency, panel of experts, court, religious organization, or other group—to decide the status of a human embryo80,81 and whether and precisely when it has “personhood”82: Is the entity merely a ball of cells whose status is like that of human skin, which sheds regularly without further ado? Or does the entity hold complete personhood status— with irreducible, inalienable moral rights and to whom we owe important directed duties? Or is the embryo’s status characterized optimally as something in between? And if so, which moral rights might this entity hold, and which duties might we owe to it? Despite this perplexing complexity, decisions one way or the other must be executed, because proceeding with research or failing to do so has consequences: Banning or significantly limiting research on human embryos stymies progress at least by making unavailable or curtailing an option to investigate the therapeutic potentials of stem cells and the biology of totipotent cells, which currently are not known to be present in any other viable human tissue sources. Totipotent cells can divide indefinitely and have the capacity to develop into all tissue types. Depending on how the status of the embryo is appraised, however, the ban also could save it from potentially unjust, lethal research-related harms. Even if the research is justified because of its potential benefit to the embryo itself and/or to others, the embryo as such cannot give informed consent at the time of the research, since the entity is not sufficiently developed. But from the research it could experience potentially lifealtering consequences—good or bad— that may extend throughout the lifespan and future generations.
By contrast, promoting such investigation may facilitate the development of novel in vitro fertilization (IVF) techniques and advances for conditions such as spinal cord injury83, Parkinson’s disease84, burns85, cardiomyopathy86, and other ailments that might be ameliorated by approaches involving regeneration. Taken together, countries must continue to decide as the science progresses whether and how to legalize experimentation on human embryos. Current positions across the globe vary widely—from outright banning of the research to illegalizing its federal funding only (while still allowing private funding for research and the research itself) to authorizing federal monies for experimentation4,6,87,88.
To what extent should CRISPR use be permitted in translational and clinical medicine?
CRISPR is significantly benefitting and improving immunotherapy89, organoid engineering and development90, in vivo drug target identification91, machine learning and artificial intelligence92, and disease-gene modification in viable human embryos32. The system offers nearly boundless potential to promote progress in combating HIV93, hemophilia94, cancer95, Duchenne muscular dystrophy96, amyotrophic lateral sclerosis97, sickle-cell anemia98, cystic fibrosis99, infertility100, and any number of novel diseases. The potential both for gaining knowledge and for developing treatments in humans seems nearly endless.
Yet such knowledge and treatment acquisition are not without potential risk. With experimentation involving somatic cells, risk assessment seems at least comparable to that which arises in regularly practiced biomedical testing. The chief objective of phase-2 oncology trials, for example, is to evaluate the efficacy of a new drug or device101. Study participants may assume significant harms, including possibly irreversible side effects and death102,103. In many countries, respect for autonomy permits assuming such risk with the requirement that informed consent occurs before enrollment in the research, regardless of whether participants are enrolling themselves or their dependents. If this risk is considered morally (and legally) permissible, then it would seem unjustified and unreasonable to not allow risk posed by CRISPR investigation. At the time of this writing, there is no empirical support to suggest that CRISPR experimentation would necessarily pose greater risk: the overall risk profile of CRISPR experimentation in human subjects remains unknown.
It could be argued, however, that heritable germline editing may present additional risk, because it involves not only the research participant, but also potentially his or her descendants. Of course, whether germline engineering technologies introduce risk beyond that which might be present in more common testing scenarios is an empirical matter. For instance, it is well-established that routinely used chemotherapies have mutagenic properties: alkylating agents, including cisplatin and cyclophosphamides, cause DNA adducts and crosslinks; antimetabolites, such as hydroxyurea, gemcitabine, and 5-fluorouracil, are nucleoside analogues and inhibit thymidine synthase; topoisomerases, such as etoposide, cause topoisomerase II inhibition, leading to double-stranded breaks in DNA; and anthracyclines, like doxorubicin, cause DNA intercalation104. Therefore, significant exposure to any of these agents increases the probability of both incurring genetic mutations and passing on these unintended genomic alterations to future generations. Whether the risk level presented by such exposure is greater than, equal to, or less than that presented by CRISPR experimentation must be quantitatively determined by empirical evidence. It is also an empirical matter whether CRISPR introduces risk that is statistically significant beyond that which is incurred in the daily experience of a healthy individual with little-to-no exposure to mutagenic agents. Thus, to determine with confidence whether it is exceptionally risky to involve humans in CRISPR translational and clinical research, possible research-related risks must be compared with those in other potentially dangerous medical and every-day contexts. This is difficult, however, given that CRISPR technology is new and that robust, reliable data about CRISPR risk in human subjects are unavailable. Nevertheless, decision making about assuming risk in CRISPR studies and therapeutics should be considered according to legal infrastructure, national and possibly international regulatory agencies, and ultimately navigated by research participants and/or their legally authorized representative(s).
Important questions also arise about whether experiments involving heritable germline editing yield reliable, interpretable data. One objection is that such experiments are unlikely to be controlled and or predicted105 because it could be impossible to analyze or understand the results from such experimentation until considerable time (decades or even generations) passes122. As previously noted, the central concern here is the uncertainty in the causal connection between gene expression/modification and the potential involvement of other factors shaping biological outcomes in the future.
Another risk, shared globally, is posed by the greater society. It is possible, for instance, that allowing CRISPR germline editing, even if only for medical purposes, might in some respect(s) lead to the return of eugenics, whose proponents believed that the human population can be improved by controlled breeding to increase the occurrence of desirable, heritable characteristics106. Unfortunately, historically, this selective weeding of people with “bad” genes and breeding of those with “good” ones resulted in many atrocities, including the forced sterilization of individuals and the propagation of racially discriminatory policies—both of which were backed by state authorities and even educated elites in different societies. In the notorious case Buck v. Bell107, for example, the United States Supreme Court (“the Court”) upheld a Virginia statute permitting the compulsory sterilization of individuals, such as Carrie Buck, who were considered “mentally unfit.” Buck was an economically disadvantaged woman who was labeled as “feebleminded” like her other family members of past generations. She was committed to the Virginia Colony for Epileptics and Feeble-Minded and was forcibly sterilized108. Unfortunately, however, the evidence of the case strongly indicates that Buck, like the others in her family, was normal and that the high Court erred gravely109. Its decision, authored by eugenics proponent Associate Justice Oliver Wendell Holmes, led to the sterilization of 50,000 Americans, set a precedent for the Nazi racial hygiene program, and is yet to be overturned124. Hence, history reveals that egregious, systematic mistakes are always possible.
To what extent should CRISPR use be permitted for non-therapeutic purposes?
Important ethical questions also arise in non-therapeutic contexts such as enhancement of crops, livestock, gene drives, and human features110.
Certain areas of CRISPR applications, such as enhancement of crops and livestock, may have greater impact on society and humanity at large. In 2016 the United Nations Food and Agriculture Organization estimated that 795 million people in the world were undernourished111. And according to the World Health Organization, 2 billion people are unable to obtain key nutrients like iron and vitamin A112. Abundant evidence demonstrates that CRISPR-Cas systems could be used to improve nutrient content in foods113–122. Why not decrease malnutrition by maximizing access to foods of higher quality? In principle, CRISPR has the potential to fortify foods efficiently for individuals who are suffering from a lack of basic nutrients. Promoting benefit in this way carries moral weight at least comparable to any other ethical concern raised herein, especially given the very large number of those who are nutrient-deprived. Nevertheless, with this benefit arise worries about “accessibility” to the product(s): Who will benefit from it? Will such food products be available to all who need it, and at what price?
Another CRISPR application that has unprecedented potential to directly benefit and save millions of lives is “gene drive technology”123. In using gene drives, researchers employ CRISPR to speed up genetic recombination such that a “gene of interest” is rapidly distributed to the entire population much faster than in a typical Mendelian inheritance rate. Therefore, this application has the potential to edit the genome of an entire population or even the entire species. Using CRISPR-mediated gene drives, investigators have demonstrated that a gene allele providing a parasite-resistance phenotype in mosquitos could quickly spread through the population in a non-Mendelian fashion124,125. This highly cost-effective technology has many potential benefits and applications for public health, species conservation, agriculture, and basic research126: Gene drives may provide a fundamental tool to fight against deadly diseases such as malaria127–129, dengue fever130, Chagas and Lyme disease131, and schistosomiasis132. This application also may control and/or alter a wide variety of animals (e.g., rodents and bats), invasive plant pests, and reservoirs135–134. Thus, the technology has unprecedented power that may save millions of lives each year. However, it is also important to consider the expected and unexpected risks of this technology: Once applied, it will eventually affect every individual of the entire species. Knowing this, researchers are developing and incorporating key safety “offswitch” features such as novel ways to (i) control, (ii) inhibit, and (iii) reverse/eliminate gene drive systems from the population in case of an unexpected or emergency event135–138.
Furthermore, is it morally permissible to use CRISPR to enhance human features such as height, muscle mass, vision, or cognitive factors like learning aptitude and memory? It becomes particularly difficult to answer this question because deviation from a “norm” may become hallmark of human diseases for many of these features. The real challenge, then, is to define what is a “norm” and when a biological phenotype is a disease deserving treatment. “Medical necessity” often becomes ambiguous. Therefore, the boundary between “therapy” and “enhancement” can be murky3. For example, a gene-editing approach may allow a reduction in bad cholesterol, thus leading to a healthier life style. Whether this hypothetical scenario, which may benefit both the individual and society in the long run, can be classified as enhancement or a medical need is unclear.
Also, in certain regions of the world, such as the United States, positive liberties are granted morally and are constitutional rights. Will medical enhancement be considered as a form of free speech?139 Alternatively, what authority should may stop individuals from exercising their right in this context?
Who should have access to CRISPR technology and/or its products?
Benefits from CRISPR innovation raise concerns and controversies about fairness and distributive justice across all layers of society. These matters are not specific to CRISPR technology, but may apply to all other technologies arising from academic research. Like many novel biomedical advancements, new CRISPR applications are expected to be profitable for patent holders. At least the initial prices of CRISPR-based products, such as gene therapy, are likely to be costly140. To this end, an ethical question is whether the high price-tag will make the CRISPR-product available to only a special class of people across societies. Since much of the funding for CRISPR characterization and development was provided by grants from government funds and thus taxpayers’ money141–152, it is ethically problematic to deny potentially life-saving benefits of the technology to the very individuals who funded much of the development in the first place. Moreover, even if it were affordable for some, there may be economic harms associated with high-price purchases. For instance, those needing the technology and its applications to maintain a reasonable quality of life, or even life itself, might be forced to make painful economic choices about whether to spend funds on treatment, food, or other essential living necessities. While this problem is not unique to genomic engineering advancements, allowing price gouging to continue unaddressed is unhelpful and potentially allows physical, psychological, and economic harms to continue. Encouraging the establishment of anti-pricegouging laws, where possible, could ameliorate some of these concerns153.
As noted earlier, CRISPR may be used to fortify foods. Those residing in some of the most impoverished areas of the world are positioned to benefit the greatest from these products. How might such individuals gain access to these foods? How could companies benefit such that reaching out to these populations might be desirable and/or lucrative? Will such foods be able to safely and successfully surpass rogue governments?
Limiting human genome editing? Somatic vs. germline editing
As noted previously, an obvious application of CRISPR technology is cell and gene therapy. To date, gene therapy mostly involves the use of genome-engineering technologies to edit somatic cells to treat genetic diseases. As mentioned above, clinical trials involving CRISPR-based gene therapy are already under way. Although clinical gene and cell therapies have had major road blocks in the past, due to unanticipated injuries and death154,155, significant safety improvements have been implemented over the last decade156. With the advances of CRISPR technology and safer delivery approaches, therapeutic applications of gene therapy are on the rise159. In the United States and elsewhere, federal regulations provide the needed legal and ethical frameworks, through the institutional review board system, to potentially minimize and manage potential risks157–161
At present there is considerable excitement about such experimentation in the United States. In January of 2018, for example, the U.S. National Institutes of Health (NIH) launched the Somatic Cell Genome Editing program, seeking “to improve the delivery mechanisms for targeting gene editing tools in patients, develop new and improved genome editors, develop assays for testing the safety and efficacy of the genome editing tools in animal and human cells, and assemble a genome editing toolkit containing the resulting knowledge, methods, and tools to be shared with the scientific community”162. Heritable genome editing, by contrast, is perhaps the systems’ greatest discussed controversy. Recently, professional scientific and medical societies, industry organizations, and CRISPR pioneers together have released greater than 60 statements and reports about whether such editing in humans is morally permissible6. Most statements hold that heritable germline experimentation should be prohibited currently, although statements from the Netherlands163, the United Kingdom164, Spain165, and the United States9 suggest that editing could be permissible if certain requirements were satisfied. The NASEM Committee’s report on germline editing, for example, specified that the following provisions must be met for human heritable germline research to commence: “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; ongoing, rigorous oversight during clinical trials 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; and reliable oversight mechanisms to prevent extension to uses other than preventing a serious disease or condition”9. Though fears about misuse in this context abound, it is also important to point out that there are reasonable arguments supporting heritable germline editing in research, such as the protection of defective embryos,166 the elimination of certain disease that might be obliterated optimally early in embryonic development, and the exercise of free speech.139
Should international regulations governing CRISPR use be crafted and promulgated?
Though ethics statements are important, by themselves, they provide little force. Typically, if ethics guidelines are infringed, the consequences suffered by the violator(s) are fairly minimal, compared to those arising when in violation of certain laws. Violations of ethics statements may lead to loss of funding, the retraction of publication, job loss, and mistrust among the scientific community. By contrast, punishments by law may lead to heavy fines and potentially incarceration. Given the significant potential promise, the dark history of eugenics, the potentially serious transgenerational risks, and the theoretical potential for misuse, it is reasonable for the global community to consider instantiating national and supranational regulations, if not revising older agreements such as the Geneva167 and United Nations Conventions on Biological and Toxin Weapons168 to reflect changes in genomic engineering technologies. While doing so likely will not eliminate all risks, it is arguably one of the few options available to reasonably control and/or minimize them.
V. Conclusions and future directions
CRISPR technology continues to mature, and existing systems are being engineered to contain innovative capabilities; excitingly new CRISPR systems with novel functions are still being discovered. The potential benefits of such revolutionary tools are endless. However like any powerful tool, there are also potential associated risks raising moral concerns. To make truly informed decisions about areas of ethical controversy, well-controlled, reproducible experimentation and clinical trials research are warranted. Currently, this is difficult because many international laws discourage or ban such research and/or inhibit its funding for certain types of investigation. Thus, widespread data about benefits and risks are unavailable. It is critical, however, for countries to examine their reasoning behind these prohibitions to ensure that they are not simply arising out of fear and without reasonable justification.
Going forward, many support establishing an organization that will decide how best to address the aforementioned ethical complexities. Recently, a group of European scientists founded the Association for Responsible Research and Innovation in Genome Editing (ARRIGE) to examine, and provide guidance about, the ethical use of genome editing169,170. Furthermore, Janasoff and Hurlbut recently advocated for the development of an international, interdisciplinary “global observatory for gene editing”171. Briefly, they argued that deliberations about moral issues in gene editing should not be dominated by the scientific community, but instead should include a “network of scholars and organizations similar to those established for human rights and climate change. The network would be dedicated to gathering information from dispersed sources, bringing to the fore perspectives that are often overlooked, and promoting exchange across disciplinary and cultural divides”198.
As the technology evolves, so will discussions about ethical and legal frameworks circumscribing its uses. The above-mentioned platforms present interesting ideas for furthering debates and potential resolutions. The research and ethical guidelines from national and international organizations, where diverse disciplines of society contribute, will be critical for federal funding agencies and IRBs to enforce and regulate, to minimize the potentials risks and maximize the potential benefits of CRISPR technology. However, it is likely that the enforcement of ethical research laws and guidelines ultimately will be assumed by legal systems, principal investigators, and institutional review boards.
Table 1.
Benefit(s) | Risk(s)/Harm(s) | |
---|---|---|
Basic and pre-clinical research |
|
|
Translational and clinical medicine |
|
|
Non-therapeuticapplications |
|
|
Access to CRISPRtechnology |
|
|
Regulations for clinicalresearch involvinghuman subjects |
|
|
National andinternationalregulations, law, andpolicy |
|
|
Acknowledgments
We are thankful to all the Adli lab members. The research in Dr. Adli’s lab is supported through local funds from University of Virginia School of Medicine and federal grants from NIH/NCI 1R01 CA211648-01.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Author Contributions
MA and CB conceptualized the study and wrote the manuscript.
References
- 1.Cyranoski D. CRISPR gene-editing tested in a person for the first time. Nature. 2016;539:479. doi: 10.1038/nature.2016.20988. [DOI] [PubMed] [Google Scholar]
- 2.CRISPR to debut in clinical trials. [cited 2018 May 15];Kwon D. Israel Molecular Medicine. 2017 Available from: http://www.molecular-medicine-israel.co.il/crispr-debut-clinical-trials/
- 3.ClinicalTrials.gov. [cited 2018 May 20];U.S. National Library of medicine. 2018 Available from: https://clinicaltrials.gov/ct2/results?cond=&term=CRISPR&cntry=&state=&city=&dist=
- 4.The L. Editing the human genome: balancing safety and regulation. Lancet. 2018;391:402. doi: 10.1016/S0140-6736(18)30153-3. [DOI] [PubMed] [Google Scholar]
- 5.Scheufele DA, Xenos MA, Howell EL, Rose KM, Brossard D, Hardy BW. U.S. attitudes on human genome editing. Science. 2017;357:553–4. doi: 10.1126/science.aan3708. [DOI] [PubMed] [Google Scholar]
- 6.Brokowski C. Do CRISPR germline ethics statements cut it? The CRISPR Journal. 2018;1(2):115–125. doi: 10.1089/crispr.2017.0024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Baltimore D, Berg P, Botchan M, Carroll D, Charo RA, Church G, et al. Biotechnology. A prudent path forward for genomic engineering and germline gene modification. Science. 2015;348:36–8. doi: 10.1126/science.aab1028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.International summit on human gene editing: a global discussion [Internet] [cited 2018 Feb 16];National Academy of Sciences Engineering and Medicine (U.S.) 2015 Available from: http://nationalacademies.org/cs/groups/pgasite/documents/webpage/pga_170455.pdf.
- 9.National Academies of Sciences Engineering and Medicine (U.S.). Human genome editing: science, ethics, and governance. Washington, DC: The National Academies Press; 2017. [PubMed] [Google Scholar]
- 10.Many countries and international decision-making bodies, including the Nuffield Council on Bioethics, the European Society of Human Genetics, and the European Society of Reproduction have issued reports and statements about ethical issues in genomic engineering, focusing specifically on the moral permissibility of germline editing. However, to date, the U.S. NASEM report is the most robust.
- 11.With stringent oversight, heritable germline editing clinical trials could one day be permitted for serious conditions; non-heritable clinical trials should be limited to treating or preventing disease or disability at this time. [2018 May 20];National Academies of Sciences Engineering and Medicine. News. 2017 Available from: http://www8.nationalacademies.org/onpinews/newsitem.aspx?RecordID=24623.
- 12.Code of Federal Regulations: Research Involving Pregnant Women or Fetuses, 45 C.F.R. Sect 46. 2017 [Google Scholar]
- 13.United States Code: Institutional Review Boards; Ethics Guidance Program, 42 U.S. Code Sect 289. 2017 [Google Scholar]
- 14. [cited 2017 Nov];Consolidated Appropriations Act of 2016 [statute on the internet] c2017 Available from: https://www.congress.gov/114/bills/hr2029/BILLS-114hr2029enr.pdf.
- 15.Robertson JA. Human embryonic stem cell research: ethical and legal issues. Nat Rev Genet. 2001;2:74–8. doi: 10.1038/35047594. [DOI] [PubMed] [Google Scholar]
- 16.Haft DH, Selengut J, Mongodin EF, Nelson KE. A guild of 45 CRISPR-associated (Cas) protein families and multiple CRISPR/Cas subtypes exist in prokaryotic genomes. PLoS Comput Biol. 2005;1:e60. doi: 10.1371/journal.pcbi.0010060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Makarova KS, Haft DH, Barrangou R, Brouns SJ, Charpentier E, Horvath P, et al. Evolution and classification of the CRISPR-Cas systems. Nat Rev Microbiol. 2011;9:467–77. doi: 10.1038/nrmicro2577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Makarova KS, Wolf YI, Alkhnbashi OS, Costa F, Shah SA, Saunders SJ, et al. An updated evolutionary classification of CRISPR-Cas systems. Nat Rev Microbiol. 2015;13(11):722–36. doi: 10.1038/nrmicro3569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Koonin EV, Makarova KS, Zhang F. Diversity, classification and evolution of CRISPR-Cas systems. Curr Opin Microbiol. 2017;37:67–78. doi: 10.1016/j.mib.2017.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Shmakov S, Smargon A, Scott D, Cox D, Pyzocha N, Yan W, et al. Diversity and evolution of class 2 CRISPR-Cas systems. Nat Rev Microbiol. 2017;15(3):169–82. doi: 10.1038/nrmicro.2016.184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Kleinstiver BP, Prew MS, Tsai SQ, Topkar VV, Nguyen NT, Zheng Z, et al. Engineered CRISPR-Cas9 nucleases with altered PAM specificities. Nature. 2015;523:481–5. doi: 10.1038/nature14592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Nishimasu H, Ran FA, Hsu PD, Konermann S, Shehata SI, Dohmae N, et al. Crystal structure of Cas9 in complex with guide RNA and target DNA. Cell. 2014;156:935–49. doi: 10.1016/j.cell.2014.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Qi LS, Larson MH, Gilbert LA, Doudna JA, Weissman JS, Arkin AP, et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell. 2013;152:1173–83. doi: 10.1016/j.cell.2013.02.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Adli M. The CRISPR tool kit for genome editing and beyond. Nat Commun. 2018;9:1911. doi: 10.1038/s41467-018-04252-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Mali P, Aach J, Stranges PB, Esvelt KM, Moosburner M, Kosuri S, et al. CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering. Nat Biotechnol. 2013;31:833–8. doi: 10.1038/nbt.2675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Ran FA, Hsu PD, Wright J, Agarwala V, Scott DA, Zhang F. Genome engineering using the CRISPR-Cas9 system. Nat Protoc. 2013;8:2281–308. doi: 10.1038/nprot.2013.143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Gaudelli NM, Komor AC, Rees HA, Packer MS, Badran AH, Bryson DI, et al. Programmable base editing of A*T to G*C in genomic DNA without DNA cleavage. Nature. 2017;551:464–71. doi: 10.1038/nature24644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Komor AC, Kim YB, Packer MS, Zuris JA, Liu DR. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature. 2016;533:420–4. doi: 10.1038/nature17946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Hess GT, Tycko J, Yao D, Bassik MC. Methods and applications of CRISPR-mediated base editing in eukaryotic genomes. Mol Cell. 2017;68:26–43. doi: 10.1016/j.molcel.2017.09.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Kuscu C, Parlak M, Tufan T, Yang J, Szlachta K, Wei X, et al. CRISPR-STOP: gene silencing through base-editing-induced nonsense mutations. Nat Methods. 2017;14:710–2. doi: 10.1038/nmeth.4327. [DOI] [PubMed] [Google Scholar]
- 33.Komor AC, Badran AH, Liu DR. CRISPR-based technologies for the manipulation of eukaryotic genomes. Cell. 2017;169:559. doi: 10.1016/j.cell.2017.04.005. [DOI] [PubMed] [Google Scholar]
- 34.Abudayyeh OO, Gootenberg JS, Essletzbichler P, Han S, Joung J, Belanto JJ, et al. RNA targeting with CRISPR-Cas13. Nature. 2017;550:280–4. doi: 10.1038/nature24049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Strutt SC, Torrez RM, Kaya E, Negrete OA, Doudna JA. RNA-dependent RNA targeting by CRISPR-Cas9. Elife. 2018;7 doi: 10.7554/eLife.32724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.CRISPR-Cas12a target binding unleashes indiscriminate single-stranded DNase activity. Science. 2018;360:436–9. doi: 10.1126/science.aar6245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Gootenberg JS, Abudayyeh OO, Kellner MJ, Joung J, Collins JJ, Zhang F. Multiplexed and portable nucleic acid detection platform with Cas13, Cas12a, and Csm6. Science. 2018;360:439–44. doi: 10.1126/science.aaq0179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Peng R, Lin G, Li J. Potential pitfalls of CRISPR/Cas9-mediated genome editing. FEBS J. 2016;283:1218–31. doi: 10.1111/febs.13586. [DOI] [PubMed] [Google Scholar]
- 39.Bohaciakova D, Renzova T, Fedorova V, Barak M, Kunova Bosakova M, Hampl A, et al. An efficient method for generation of knockout human embryonic stem cells using CRISPR/Cas9 System. Stem Cells Dev. 2017;26:1521–7. doi: 10.1089/scd.2017.0058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Guo X, Li XJ. Targeted genome editing in primate embryos. Cell Res. 2015;25:767–8. doi: 10.1038/cr.2015.64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Tu Z, Yang W, Yan S, Yin A, Gao J, Liu X, et al. Promoting Cas9 degradation reduces mosaic mutations in non-human primate embryos. Sci Rep. 2017;7:42081. doi: 10.1038/srep42081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Schaefer KA, Wu WH, Colgan DF, Tsang SH, Bassuk AG, Mahajan VB. Unexpected mutations after CRISPR-Cas9 editing in vivo. Nat Methods. 2017;14:547–8. doi: 10.1038/nmeth.4293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Zischewski J, Fischer R, Bortesi L. Detection of on-target and off-target mutations generated by CRISPR/Cas9 and other sequence-specific nucleases. Biotechnol Adv. 2017;35:95–104. doi: 10.1016/j.biotechadv.2016.12.003. [DOI] [PubMed] [Google Scholar]
- 44.Jelenkovic A, Sund R, Hur YM, Yokoyama Y, Hjelmborg JV, Moller S, et al. Genetic and environmental influences on height from infancy to early adulthood: An individual-based pooled analysis of 45 twin cohorts. Sci Rep. 2016;6:28496. doi: 10.1038/srep28496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Wu S, Powers S, Zhu W, Hannun YA. Substantial contribution of extrinsic risk factors to cancer development. Nature. 2016;529:43–7. doi: 10.1038/nature16166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Code of Federal Regulations: Food and Drugs, 21 C.F.R. (scattered) [Google Scholar]
- 47. United States Code: Regulation of Biological Products, 42 U.S. Code Sect 262. 2017 [Google Scholar]
- 48. Such guidance is inapplicable to cells, tissues, and cellular- and tissue-based products regulated under United States Code: Regulations to Control Communicable Diseases, 42 U.S. Code Sect 264. 2017 [Google Scholar]
- 49.Kimmelman J. Ethics of cancer gene transfer clinical research. Methods Mol Biol. 2015;1317:263–85. doi: 10.1007/978-1-4939-2727-2_15. [DOI] [PubMed] [Google Scholar]
- 50.Tang YD, Liu JT, Wang TY, Sun MX, Tian ZJ, Cai XH. CRISPR/Cas9-mediated multiple single guide RNAs potently abrogate pseudorabies virus replication. Arch Virol. 2017;162:3881–6. doi: 10.1007/s00705-017-3553-4. [DOI] [PubMed] [Google Scholar]
- 51.Shipman SL, Nivala J, Macklis JD, Church GM. CRISPR-Cas encoding of a digital movie into the genomes of a population of living bacteria. Nature. 2017;547:345–9. doi: 10.1038/nature23017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Gratz SJ, Cummings AM, Nguyen JN, Hamm DC, Donohue LK, Harrison MM, et al. Genome engineering of Drosophila with the CRISPR RNA-guided Cas9 nuclease. Genetics. 2013;194:1029–35. doi: 10.1534/genetics.113.152710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Dong S, Lin J, Held NL, Clem RJ, Passarelli AL, Franz AW. Heritable CRISPR/Cas9-mediated genome editing in the yellow fever mosquito, Aedes aegypti. PLoS One. 2015;10:e0122353. doi: 10.1371/journal.pone.0122353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.DiCarlo JE, Norville JE, Mali P, Rios X, Aach J, Church GM. Genome engineering in Saccharomyces cerevisiae using CRISPR-Cas systems. Nucleic Acids Res. 2013;41:4336–43. doi: 10.1093/nar/gkt135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Kraus Y, Aman A, Technau U, Genikhovich G. Pre-bilaterian origin of the blastoporal axial organizer. Nat Commun. 2016;7:11694. doi: 10.1038/ncomms11694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Friedland AE, Tzur YB, Esvelt KM, Colaiacovo MP, Church GM, Calarco JA. Heritable genome editing in C. elegans via a CRISPR-Cas9 system. Nat Methods. 2013;10:741–3. doi: 10.1038/nmeth.2532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Irion U, Krauss J, Nusslein-Volhard C. Precise and efficient genome editing in zebrafish using the CRISPR/Cas9 system. Development. 2014;141:4827–30. doi: 10.1242/dev.115584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Feng Z, Zhang B, Ding W, Liu X, Yang DL, Wei P, et al. Efficient genome editing in plants using a CRISPR/Cas system. Cell Res. 2013;23:1229–32. doi: 10.1038/cr.2013.114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Hai T, Teng F, Guo R, Li W, Zhou Q. One-step generation of knockout pigs by zygote injection of CRISPR/Cas system. Cell Res. 2014;24:372–5. doi: 10.1038/cr.2014.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Ikeda M, Matsuyama S, Akagi S, Ohkoshi K, Nakamura S, Minabe S, et al. Correction of a Disease Mutation using CRISPR/Cas9-assisted Genome Editing in Japanese Black Cattle. Sci Rep. 2017;7:17827. doi: 10.1038/s41598-017-17968-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Zuo E, Cai YJ, Li K, Wei Y, Wang BA, Sun Y, et al. One-step generation of complete gene knockout mice and monkeys by CRISPR/Cas9-mediated gene editing with multiple sgRNAs. Cell Res. 2017;27:933–45. doi: 10.1038/cr.2017.81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.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. 2017;548:413–9. doi: 10.1038/nature23305. [DOI] [PubMed] [Google Scholar]
- 63.Kang X, He W, Huang Y, Yu Q, Chen Y, Gao X, et al. Introducing precise genetic modifications into human 3PN embryos by CRISPR/Cas-mediated genome editing. J Assist Reprod Genet. 2016;33:581–8. doi: 10.1007/s10815-016-0710-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Gaj T, Gersbach CA, Barbas CF., 3rd ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol. 2013;31:397–405. doi: 10.1016/j.tibtech.2013.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Nakamura T, Gehrke AR, Lemberg J, Szymaszek J, Shubin NH. Digits and fin rays share common developmental histories. Nature. 2016;537:225–8. doi: 10.1038/nature19322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Russell JJ, Theriot JA, Sood P, Marshall WF, Landweber LF, Fritz-Laylin L, et al. Non-model model organisms. BMC Biol. 2017;15:55. doi: 10.1186/s12915-017-0391-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Shang W, Wang F, Fan G, Wang H. Key elements for designing and performing a CRISPR/Cas9-based genetic screen. J Genet Genomics. 2017;44:439–49. doi: 10.1016/j.jgg.2017.09.005. [DOI] [PubMed] [Google Scholar]
- 68.Liao HK, Hatanaka F, Araoka T, Reddy P, Wu MZ, Sui Y, et al. In vivo target gene activation via CRISPR/Cas9-Mmdiated trans-epigenetic modulation. Cell. 2017;171:1495–507 e15. doi: 10.1016/j.cell.2017.10.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Yang M, Zhang L, Stevens J, Gibson G. CRISPR/Cas9 mediated generation of stable chondrocyte cell lines with targeted gene knockouts; analysis of an aggrecan knockout cell line. Bone. 2014;69:118–25. doi: 10.1016/j.bone.2014.09.005. [DOI] [PubMed] [Google Scholar]
- 70.Lu XJ, Xiang YY, Li XJ. CRISPR screen: a high-throughput approach for cancer genetic research. Clin Genet. 2015;88:32–3. doi: 10.1111/cge.12606. [DOI] [PubMed] [Google Scholar]
- 71.Charpentier E, Richter H, van der Oost J, White MF. Biogenesis pathways of RNA guides in archaeal and bacterial CRISPR-Cas adaptive immunity. FEMS Microbiol Rev. 2015;39:428–41. doi: 10.1093/femsre/fuv023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Klann TS, Black JB, Chellappan M, Safi A, Song L, Hilton IB, et al. CRISPR-Cas9 epigenome editing enables high-throughput screening for functional regulatory elements in the human genome. Nat Biotechnol. 2017;35:561–8. doi: 10.1038/nbt.3853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Mazo-Vargas A, Concha C, Livraghi L, Massardo D, Wallbank RWR, Zhang L, et al. Macroevolutionary shifts of WntA function potentiate butterfly wing-pattern diversity. Proc Natl Acad Sci U S A. 2017;114:10701–6. doi: 10.1073/pnas.1708149114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Atanes P, Ruz-Maldonado I, Hawkes R, Liu B, Persaud SJ, Amisten S. Identifying Signalling Pathways Regulated by GPRC5B in beta-Cells by CRISPR-Cas9-Mediated Genome Editing. Cell Physiol Biochem. 2018;45:656–66. doi: 10.1159/000487159. [DOI] [PubMed] [Google Scholar]
- 75.Chi S, Weiss A, Wang H. A CRISPR-Based Toolbox for Studying T Cell Signal Transduction. Biomed Res Int. 2016;2016:5052369. doi: 10.1155/2016/5052369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Kasap C, Elemento O, Kapoor TM. DrugTargetSeqR: a genomics- and CRISPR-Cas9-based method to analyze drug targets. Nat Chem Biol. 2014;10:626–8. doi: 10.1038/nchembio.1551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Housden BE, Valvezan AJ, Kelley C, Sopko R, Hu Y, Roesel C, et al. Identification of potential drug targets for tuberous sclerosis complex by synthetic screens combining CRISPR-based knockouts with RNAi. Sci Signal. 2015;8:rs9. doi: 10.1126/scisignal.aab3729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Hyun I, Wilkerson A, Johnston J. Embryology policy: Revisit the 14-day rule. Nature. 2016;533(7602):169–71. doi: 10.1038/533169a. [DOI] [PubMed] [Google Scholar]
- 79.Human embryo culture: Discussions concerning the statutory time limit for maintaining human embryos in culture in light of some recent scientific developments. [cited 2018 May 29];Nuffield Council on bioethics. 2017 Available from: http://nuffieldbioethics.org/wp-content/uploads/Human-Embryo-Culture-web-FINAL.pdf.
- 80.Charo RA. The hunting of the snark: the moral status of embryos, right-to-lifers, and Third World women. Stanford Law Pol Rev. 1995;6:11–37. [PubMed] [Google Scholar]
- 81.Wennergren B. Human rights of an embryo. J Int Bioethique. 1991;2:46–9. [PubMed] [Google Scholar]
- 82.Miklavcic JJ, Flaman P. Personhood status of the human zygote, embryo, fetus. Linacre Q. 2017;84:130–44. doi: 10.1080/00243639.2017.1299896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.McDonald JW, Liu XZ, Qu Y, Liu S, Mickey SK, Turetsky D, et al. Transplanted embryonic stem cells survive, differentiate and promote recovery in injured rat spinal cord. Nat Med. 1999;5:1410–2. doi: 10.1038/70986. [DOI] [PubMed] [Google Scholar]
- 84.Bjorklund LM, Sanchez-Pernaute R, Chung S, Andersson T, Chen IY, McNaught KS, et al. Embryonic stem cells develop into functional dopaminergic neurons after transplantation in a Parkinson rat model. Proc Natl Acad Sci U S A. 2002;99:2344–9. doi: 10.1073/pnas.022438099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Hamrahi VF, Goverman J, Jung W, Wu JC, Fischman AJ, Tompkins RG, et al. In vivo molecular imaging of murine embryonic stem cells delivered to a burn wound surface via Integra(R) scaffolding. J Burn Care Res. 2012;33:e49–54. doi: 10.1097/BCR.0b013e3182331d1c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Singla DK, Ahmed A, Singla R, Yan B. Embryonic stem cells improve cardiac function in Doxorubicin-induced cardiomyopathy mediated through multiple mechanisms. Cell Transplant. 2012;21:1919–30. doi: 10.3727/096368911X627552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Busardo FP, Gulino M, Napoletano S, Zaami S, Frati P. The evolution of legislation in the field of Medically Assisted Reproduction and embryo stem cell research in European union members. Biomed Res Int. 2014;2014:307160. doi: 10.1155/2014/307160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Dajani R. Jordan's stem-cell law can guide the Middle East. Nature. 2014;510:189. doi: 10.1038/510189a. [DOI] [PubMed] [Google Scholar]
- 89.Manguso RT, Pope HW, Zimmer MD, Brown FD, Yates KB, Miller BC, et al. In vivo CRISPR screening identifies Ptpn2 as a cancer immunotherapy target. Nature. 2017;547:413–8. doi: 10.1038/nature23270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Drost J, van Boxtel R, Blokzijl F, Mizutani T, Sasaki N, Sasselli V, et al. Use of CRISPR-modified human stem cell organoids to study the origin of mutational signatures in cancer. Science. 2017;358:234–8. doi: 10.1126/science.aao3130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Chu J, Galicia-Vazquez G, Cencic R, Mills JR, Katigbak A, Porco JA, Jr, et al. CRISPR-Mediated Drug-Target Validation Reveals Selective Pharmacological Inhibition of the RNA Helicase, eIF4A. Cell Rep. 2016;15:2340–7. doi: 10.1016/j.celrep.2016.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Hough SH, Ajetunmobi A, Brody L, Humphryes-Kirilov N, Perello E. Desktop Genetics. Per Med. 2016;13:517–21. doi: 10.2217/pme-2016-0068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Huang Z, Tomitaka A, Raymond A, Nair M. Current application of CRISPR/Cas9 gene-editing technique to eradication of HIV/AIDS. Gene Ther. 2017;24:377–84. doi: 10.1038/gt.2017.35. [DOI] [PubMed] [Google Scholar]
- 94.George LA, Sullivan SK, Giermasz A, Rasko JEJ, Samelson-Jones BJ, Ducore J, et al. Hemophilia B Gene Therapy with a High-Specific-Activity Factor IX Variant. N Engl J Med. 2017;377:2215–27. doi: 10.1056/NEJMoa1708538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Wei C, Wang F, Liu W, Zhao W, Yang Y, Li K, et al. CRISPR/Cas9 targeting of the androgen receptor suppresses the growth of LNCaP human prostate cancer cells. Mol Med Rep. 2018;17:2901–6. doi: 10.3892/mmr.2017.8257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Wong TWY, Cohn RD. Therapeutic Applications of CRISPR/Cas for Duchenne Muscular Dystrophy. Curr Gene Ther. 2017;17:301–8. doi: 10.2174/1566523217666171121165046. [DOI] [PubMed] [Google Scholar]
- 97.Gaj T, Ojala DS, Ekman FK, Byrne LC, Limsirichai P, Schaffer DV. In vivo genome editing improves motor function and extends survival in a mouse model of ALS. Sci Adv. 2017;3:eaar3952. doi: 10.1126/sciadv.aar3952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Wen J, Tao W, Hao S, Zu Y. Cellular function reinstitution of offspring red blood cells cloned from the sickle cell disease patient blood post CRISPR genome editing. J Hematol Oncol. 2017;10:119. doi: 10.1186/s13045-017-0489-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Schwank G, Koo BK, Sasselli V, Dekkers JF, Heo I, Demircan T, et al. Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients. Cell Stem Cell. 2013;13:653–8. doi: 10.1016/j.stem.2013.11.002. [DOI] [PubMed] [Google Scholar]
- 100.Wu Y, Zhou H, Fan X, Zhang Y, Zhang M, Wang Y, et al. Correction of a genetic disease by CRISPR-Cas9-mediated gene editing in mouse spermatogonial stem cells. Cell Res. 2015;25:67–79. doi: 10.1038/cr.2014.160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Code of Federal Regulations: Investigational New Drug, 21 C.F.R. Sec 312. 2017 [Google Scholar]
- 102.Suntharalingam G, Perry MR, Ward S, Brett SJ, Castello-Cortes A, Brunner MD, et al. Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N Engl J Med. 2006;355:1018–28. doi: 10.1056/NEJMoa063842. [DOI] [PubMed] [Google Scholar]
- 103.Kaur R, Sidhu P, Singh S. What failed BIA 10-2474 Phase I clinical trial? Global speculations and recommendations for future Phase I trials. J Pharmacol Pharmacother. 2016;7:120–6. doi: 10.4103/0976-500X.189661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Szikriszt B, Poti A, Pipek O, Krzystanek M, Kanu N, Molnar J, et al. A comprehensive survey of the mutagenic impact of common cancer cytotoxics. Genome Biol. 2016;17:99. doi: 10.1186/s13059-016-0963-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.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;23:1282. doi: 10.1038/mt.2015.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Brokowski C, Pollack M, Pollack R. Cutting eugenics out of CRISPR-Cas9. EBEM. 2015;6:263. [Google Scholar]
- 107.Buck v. Bell, 274 U.S. 200. 1927 [Google Scholar]
- 108.Smith JD, Nelson KR. The sterilization of Carrie Buck. Far Hills, N.J: New Horizon Press; 1989. [Google Scholar]
- 109.Lombardo PA. Bell. Baltimore: Johns Hopkins University Press; 2008. Three generations, no imbeciles: eugenics, the Supreme Court, and Buck v. [Google Scholar]
- 110.Savulescu J, Bostrom N. Human enhancement. Oxford ; New York: Oxford University Press; 2009. [Google Scholar]
- 111.United Nations. [cited 2017 Dec 23];Food. Available from: http://www.un.org/sustainabledevelopment/hunger/
- 112.World Health Organization. [cited 2017 Dec 23];Nutrition. Available from: http://apps.who.int/iris/bitstream/10665/43412/1/9241594012_eng.pdf?ua=1.
- 113.Stout E, Klaenhammer T, Barrangou R. CRISPR-Cas technologies and applications in food bacteria. Annu Rev Food Sci Technol. 2017;8:413–37. doi: 10.1146/annurev-food-072816-024723. [DOI] [PubMed] [Google Scholar]
- 114.Demirci Y, Zhang B, Unver T. CRISPR/Cas9: An RNA-guided highly precise synthetic tool for plant genome editing. J Cell Physiol. 2018;233:1844–59. doi: 10.1002/jcp.25970. [DOI] [PubMed] [Google Scholar]
- 115.Zhang J, Zhang H, Botella JR, Zhu JK. Generation of new glutinous rice by CRISPR/Cas9-targeted mutagenesis of the Waxy gene in elite rice varieties. J Integr Plant Biol. 2017 doi: 10.1111/jipb.12620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Rajendran SR, Yau YY, Pandey D, Kumar A. CRISPR-Cas9 Based Genome Engineering: Opportunities in Agri-Food-Nutrition and Healthcare. OMICS. 2015;19:261–75. doi: 10.1089/omi.2015.0023. [DOI] [PubMed] [Google Scholar]
- 117.Du H, Zeng X, Zhao M, Cui X, Wang Q, Yang H, et al. Efficient targeted mutagenesis in soybean by TALENs and CRISPR/Cas9. J Biotechnol. 2016;217:90–7. doi: 10.1016/j.jbiotec.2015.11.005. [DOI] [PubMed] [Google Scholar]
- 118.Svitashev S, Young JK, Schwartz C, Gao H, Falco SC, Cigan AM. Targeted Mutagenesis, Precise Gene Editing, and Site-Specific Gene Insertion in Maize Using Cas9 and Guide RNA. Plant Physiol. 2015;169:931–45. doi: 10.1104/pp.15.00793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Brinegar K, A KY, Choi S, Vallillo E, Ruiz-Esparza GU, Prabhakar AM, et al. The commercialization of genome-editing technologies. Crit Rev Biotechnol. 2017;37:924–32. doi: 10.1080/07388551.2016.1271768. [DOI] [PubMed] [Google Scholar]
- 120.Georges F, Ray H. Genome editing of crops: A renewed opportunity for food security. GM Crops Food. 2017;8:1–12. doi: 10.1080/21645698.2016.1270489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Gonen S, Jenko J, Gorjanc G, Mileham AJ, Whitelaw CB, Hickey JM. Potential of gene drives with genome editing to increase genetic gain in livestock breeding programs. Genet Sel Evol. 2017;49:3. doi: 10.1186/s12711-016-0280-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Wang X, Niu Y, Zhou J, Yu H, Kou Q, Lei A, et al. Multiplex gene editing via CRISPR/Cas9 exhibits desirable muscle hypertrophy without detectable off-target effects in sheep. Sci Rep. 2016;6:32271. doi: 10.1038/srep32271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Gantz VM, Bier E. Genome editing. The mutagenic chain reaction: a method for converting heterozygous to homozygous mutations. Science. 2015;348:442–4. doi: 10.1126/science.aaa5945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Gantz VM, Jasinskiene N, Tatarenkova O, Fazekas A, Macias VM, Bier E, et al. Highly efficient Cas9-mediated gene drive for population modification of the malaria vector mosquito Anopheles stephensi. Proc Natl Acad Sci U S A. 2015;112:E6736–43. doi: 10.1073/pnas.1521077112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Hammond A, Galizi R, Kyrou K, Simoni A, Siniscalchi C, Katsanos D, et al. A CRISPR-Cas9 gene drive system targeting female reproduction in the malaria mosquito vector Anopheles gambiae. Nat Biotechnol. 2016;34:78–83. doi: 10.1038/nbt.3439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Gene drives on the horizon : advancing science, navigating uncertainty, and aligning research with public values. Washington, D.C: The National Academies Press; 2016. National Academies of Sciences Engineering and Medicine (U.S.). Committee on Gene Drive Research in Non-Human Organisms: Recommendations for Responsible Conduct. [PubMed] [Google Scholar]
- 127.Windbichler N, Menichelli M, Papathanos PA, Thyme SB, Li H, Ulge UY, et al. A synthetic homing endonuclease-based gene drive system in the human malaria mosquito. Nature. 2011;473:212–5. doi: 10.1038/nature09937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Eckhoff PA, Wenger EA, Godfray HC, Burt A. Impact of mosquito gene drive on malaria elimination in a computational model with explicit spatial and temporal dynamics. Proc Natl Acad Sci U S A. 2017;114:E255–E64. doi: 10.1073/pnas.1611064114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Eckhoff PA, Wenger EA, Godfray HC, Burt A. Impact of mosquito gene drive on malaria elimination in a computational model with explicit spatial and temporal dynamics. Proc Natl Acad Sci U S A. 2017;114:E255–E64. doi: 10.1073/pnas.1611064114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Achee NL, Gould F, Perkins TA, Reiner RC, Jr, Morrison AC, Ritchie SA, et al. A critical assessment of vector control for dengue prevention. PLoS Negl Trop Dis. 2015;9:e0003655. doi: 10.1371/journal.pntd.0003655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Esvelt KM, Smidler AL, Catteruccia F, Church GM. Concerning RNA-guided gene drives for the alteration of wild populations. Elife. 2014;3 doi: 10.7554/eLife.03401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Sokolow SH, Wood CL, Jones IJ, Lafferty KD, Kuris AM, Hsieh MH, et al. To reduce the global burden of human schistosomiasis, use 'old fashioned' snail control. Trends Parasitol. 2018;34:23–40. doi: 10.1016/j.pt.2017.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Courtier-Orgogozo V, Morizot B, Boete C. Using CRISPR-based gene drive for agriculture pest control. EMBO Rep. 2017;18:1481. doi: 10.15252/embr.201744822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.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. Global Ecology and Conservation. 2018;13:e00363. [Google Scholar]
- 135.DiCarlo JE, Chavez A, Dietz SL, Esvelt KM, Church GM. Safeguarding CRISPR-Cas9 gene drives in yeast. Nat Biotechnol. 2015;33:1250–5. doi: 10.1038/nbt.3412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Roggenkamp E, Giersch RM, Schrock MN, Turnquist E, Halloran M, Finnigan GC. Tuning CRISPR-Cas9 Gene Drives in Saccharomyces cerevisiae. G3 (Bethesda) 2018;8:999–1018. doi: 10.1534/g3.117.300557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Basgall EM, Goetting SC, Goeckel ME, Giersch RM, Roggenkamp E, Schrock MN, et al. Gene drive inhibition by the anti-CRISPR proteins AcrIIA2 and AcrIIA4 in Saccharomyces cerevisiae. Microbiology. 2018;164:464–74. doi: 10.1099/mic.0.000635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Vella MR, Gunning CE, Lloyd AL, Gould F. Evaluating strategies for reversing CRISPR-Cas9 gene drives. Sci Rep. 2017;7:11038. doi: 10.1038/s41598-017-10633-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.U.S. CONST. amend. 1 [Google Scholar]
- 140.White EK. Killing U.S. Slowly: Curing the Epidemic Rise of Cancer Drug Prices. Food Drug Law J. 2017;72:189–224. [PubMed] [Google Scholar]
- 141.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Chen S, Sanjana NE, Zheng K, Shalem O, Lee K, Shi X, et al. Genome-wide CRISPR screen in a mouse model of tumor growth and metastasis. Cell. 2015;160:1246–60. doi: 10.1016/j.cell.2015.02.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Wang H, Yang H, Shivalila CS, Dawlaty MM, Cheng AW, Zhang F, et al. One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell. 2013;153:910–8. doi: 10.1016/j.cell.2013.04.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Ran FA, Cong L, Yan WX, Scott DA, Gootenberg JS, Kriz AJ, et al. In vivo genome editing using Staphylococcus aureus Cas9. Nature. 2015;520:186–91. doi: 10.1038/nature14299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Chow RD, Guzman CD, Wang G, Schmidt F, Youngblood MW, Ye L, et al. AAV-mediated direct in vivo CRISPR screen identifies functional suppressors in glioblastoma. Nat Neurosci. 2017;20:1329–41. doi: 10.1038/nn.4620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Doudna J, Mason A. Federal funding for basic research led to the gene-editing revolution. Don’t cut it. Vox. 2017 [Google Scholar]
- 148.Moreno-Mateos MA, Fernandez JP, Rouet R, Vejnar CE, Lane MA, Mis E, et al. CRISPR-Cpf1 mediates efficient homology-directed repair and temperature-controlled genome editing. Nat Commun. 2017;8:2024. doi: 10.1038/s41467-017-01836-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Burstein D, Harrington LB, Strutt SC, Probst AJ, Anantharaman K, Thomas BC, et al. New CRISPR-Cas systems from uncultivated microbes. Nature. 2017;542:237–41. doi: 10.1038/nature21059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Sternberg SH, LaFrance B, Kaplan M, Doudna JA. Conformational control of DNA target cleavage by CRISPR-Cas9. Nature. 2015;527:110–3. doi: 10.1038/nature15544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Schumann K, Lin S, Boyer E, Simeonov DR, Subramaniam M, Gate RE, et al. Generation of knock-in primary human T cells using Cas9 ribonucleoproteins. Proc Natl Acad Sci U S A. 2015;112:10437–42. doi: 10.1073/pnas.1512503112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Greene JA, Padula WV. Targeting Unconscionable Prescription-Drug Prices - Maryland's Anti-Price-Gouging Law. N Engl J Med. 2017;377:101–3. doi: 10.1056/NEJMp1704907. [DOI] [PubMed] [Google Scholar]
- 154.Marshall E. Gene therapy death prompts review of adenovirus vector. Science. 1999;286:2244–5. doi: 10.1126/science.286.5448.2244. [DOI] [PubMed] [Google Scholar]
- 155.Frank KM, Hogarth DK, Miller JL, Mandal S, Mease PJ, Samulski RJ, et al. Investigation of the cause of death in a gene-therapy trial. N Engl J Med. 2009;361:161–9. doi: 10.1056/NEJMoa0801066. [DOI] [PubMed] [Google Scholar]
- 156.Dunbar CE, High KA, Joung JK, Kohn DB, Ozawa K, Sadelain M. Gene therapy comes of age. Science. 2018;359 doi: 10.1126/science.aan4672. [DOI] [PubMed] [Google Scholar]
- 157. Code of Federal Regulations: Protection of Human Subjects, 45 C.F.R. Sect 46. 2017 [Google Scholar]
- 158.Code of Federal Regulations: Food and Drugs, Protection of Human Subjects, 21 C.F.R. 50. 2017 [Google Scholar]
- 159.Code of Federal Regulations: Food and Drugs, Institutional Review Boards, 21 C.F.R. 56. 2017 [Google Scholar]
- 160.Code of Federal Regulations: Food and Drugs, Investigational New Drug Application, 21 C.F.R. 312. 2017 [Google Scholar]
- 161.Code of Federal Regulations: Food and Drugs, Investigational Device, 21 C.F.R. 812. 2017 [Google Scholar]
- 162.NIH to launch genome editing research program. https://www.nih.gov/news-events/news-releases/nihlaunch-genome-editing-research-program.
- 163.Commission on Genetic Modification and the Health Council of the Netherlands. [Accessed 26 May 2018];Editing human DNA: Moral and social implications of germline genetic modification. [Google Scholar]
- 164.The Academy of Medical Sciences’ response to the Nuffield Council on Bioethics. Genome editing and human reproduction. [cited 2018 May 26];The Academy of Medical Sciences. 2017 [press release]. Available from: https://acmedsci.ac.uk/file-download/89659337.
- 165.Document on bioethics and gene editing in humans. Bioethics and Law Observatory of the University of Barcelona. [cited 2018 May 26];Bioethics and Law Observatory of the University of Barcelona. 2016 Available from: http://www.publicacions.ub.edu/refs/observatoriBioEticaDret/documents/08543.pdf.
- 166.Church G. Compelling Reasons for Repairing Human Germlines. N Engl J Med. 2017;377:1909–11. doi: 10.1056/NEJMp1710370. [DOI] [PubMed] [Google Scholar]
- 167.Geneva Convention. [accessed 20 December 2017];Protocol for the Prohibition of the Use in war of Asphyxiating, Poisonous or Other Gases, and of bacteriological methods of warfare. 1925 http://avalon.law.yale.edu/20th_century/geneva01.asp.
- 168.United Nations. [accessed 20 December 2017];Convention on the Prohibition of the Development, Production and Stockpiling of Bacteriological (Biological) and Toxin Weapons and on Their Destruction. 1975 http://avalon.law.yale.edu/20th_century/bact.asp.
- 169.Enserink M. Interested in responsible genome editing? Join the new club. Science. 2018 doi: 10.1126/science.aat7183. [DOI] [Google Scholar]
- 170.Lluis M, Jennifer M, François H, Marion A, Pierre J, Bernard B, Cyril SM, Hervé C. ARRIGE arrives: toward the responsible use of genome editing. The CRISPR Journal. 2018;1(2):128–129. doi: 10.1089/crispr.2018.29012.mon. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Jasanoff S, Hurlbut JB. A global observatory for gene editing. Nature. 2018;555:435–7. doi: 10.1038/d41586-018-03270-w. [DOI] [PubMed] [Google Scholar]