Monday, November 26, 2018

Gene edited babies


A Chinese researcher claims that he helped make the world’s first genetically edited babies — twin girls born this month whose DNA he said he altered with a powerful new tool capable of rewriting the very blueprint of life.

If true, it would be a profound leap of science and ethics.

A U.S. scientist said he took part in the work in China, but this kind of gene editing is banned in the United States because the DNA changes can pass to future generations and it risks harming other genes.

Many mainstream scientists think it’s too unsafe to try, and some denounced the Chinese report as human experimentation. 

The researcher, He Jiankui of Shenzhen, said he altered embryos for seven couples during fertility treatments, with one pregnancy resulting thus far. He said his goal was not to cure or prevent an inherited disease, but to try to bestow a trait that few people naturally have — an ability to resist possible future infection with HIV, the AIDS virus.

He said the parents involved declined to be identified or interviewed, and he would not say where they live or where the work was done.

There is no independent confirmation of He’s claim, and it has not been published in a journal, where it would be vetted by other experts. He revealed it Monday in Hong Kong to one of the organizers of an international conference on gene editing that is set to begin Tuesday, and earlier in exclusive interviews with The Associated Press.

“I feel a strong responsibility that it’s not just to make a first, but also make it an example,” He told the AP. “Society will decide what to do next” in terms of allowing or forbidding such science.

Some scientists were astounded to hear of the claim and strongly condemned it.

It’s “unconscionable ... an experiment on human beings that is not morally or ethically defensible,” said Dr. Kiran Musunuru, a University of Pennsylvania gene editing expert and editor of a genetics journal.

“This is far too premature,” said Dr. Eric Topol, who heads the Scripps Research Translational Institute in California. “We’re dealing with the operating instructions of a human being. It’s a big deal.”

However, one famed geneticist, Harvard University’s George Church, defended attempting gene editing for HIV, which he called “a major and growing public health threat.”

“I think this is justifiable,” Church said of that goal.

In recent years scientists have discovered a relatively easy way to edit genes, the strands of DNA that govern the body. The tool, called CRISPR-cas9, makes it possible to operate on DNA to supply a needed gene or disable one that’s causing problems.

It’s only recently been tried in adults to treat deadly diseases, and the changes are confined to that person. Editing sperm, eggs or embryos is different — the changes can be inherited. In the U.S., it’s not allowed except for lab research. China outlaws human cloning but not specifically gene editing.

He Jiankui (HEH JEE’-an-qway), who goes by “JK,” studied at Rice and Stanford universities in the U.S. before returning to his homeland to open a lab at Southern University of Science and Technology of China in Shenzhen, where he also has two genetics companies. The university said He’s work “seriously violated academic ethics and standards” and planned to investigate. A spokesman for He confirmed that he has been on leave from teaching since early this year, but he remains on the faculty and has a lab at the school.

The U.S. scientist who worked with him on this project after He returned to China was physics and bioengineering professor Michael Deem, who was his adviser at Rice in Houston. Deem also holds what he called “a small stake” in — and is on the scientific advisory boards of — He’s two companies.

The Chinese researcher said he practiced editing mice, monkey and human embryos in the lab for several years and has applied for patents on his methods.

He said he chose embryo gene editing for HIV because these infections are a big problem in China. He sought to disable a gene called CCR5 that forms a protein doorway that allows HIV, the virus that causes AIDS, to enter a cell.

All of the men in the project had HIV and all of the women did not, but the gene editing was not aimed at preventing the small risk of transmission, He said. The fathers had their infections deeply suppressed by standard HIV medicines and there are simple ways to keep them from infecting offspring that do not involve altering genes.

Instead, the appeal was to offer couples affected by HIV a chance to have a child that might be protected from a similar fate.

He recruited couples through a Beijing-based AIDS advocacy group called Baihualin. Its leader, known by the pseudonym “Bai Hua,” told the AP that it’s not uncommon for people with HIV to lose jobs or have trouble getting medical care if their infections are revealed.

Here is how He described the work:

The gene editing occurred during IVF, or lab dish fertilization. First, sperm was “washed” to separate it from semen, the fluid where HIV can lurk. A single sperm was placed into a single egg to create an embryo. Then the gene editing tool was added.

When the embryos were 3 to 5 days old, a few cells were removed and checked for editing. Couples could choose whether to use edited or unedited embryos for pregnancy attempts. In all, 16 of 22 embryos were edited, and 11 embryos were used in six implant attempts before the twin pregnancy was achieved, He said.

Tests suggest that one twin had both copies of the intended gene altered and the other twin had just one altered, with no evidence of harm to other genes, He said. People with one copy of the gene can still get HIV, although some very limited research suggests their health might decline more slowly once they do.

The Rice scientist, Deem, said he was present in China when potential participants gave their consent and that he “absolutely” thinks they were able to understand the risks.

Deem said he worked with He on vaccine research at Rice and considers the gene editing similar to a vaccine.

“That might be a layman’s way of describing it,” he said.

Both men are physics experts with no experience running human clinical trials.

The Chinese scientist, He, said he personally made the goals clear and told participants that embryo gene editing has never been tried before and carries risks. He said he also would provide insurance coverage for any children conceived through the project and plans medical follow-up until the children are 18 and longer if they agree once they’re adults.

Further pregnancy attempts are on hold until the safety of this one is analyzed and experts in the field weigh in, but participants were not told in advance that they might not have a chance to try what they signed up for once a “first” was achieved, He acknowledged. Free fertility treatment was part of the deal they were offered.

He sought and received approval for his project from Shenzhen Harmonicare Women’s and Children’s Hospital, which is not one of the four hospitals that He said provided embryos for his research or the pregnancy attempts.

Some staff at some of the other hospitals were kept in the dark about the nature of the research, which He and Deem said was done to keep some participants’ HIV infection from being disclosed.

“We think this is ethical,” said Lin Zhitong, a Harmonicare administrator who heads the ethics panel.

Any medical staff who handled samples that might contain HIV were aware, He said. An embryologist in He’s lab, Qin Jinzhou, confirmed to the AP that he did sperm washing and injected the gene editing tool in some of the pregnancy attempts.

The study participants are not ethicists, He said, but “are as much authorities on what is correct and what is wrong because it’s their life on the line.”

“I believe this is going to help the families and their children,” He said. If it causes unwanted side effects or harm, “I would feel the same pain as they do and it’s going to be my own responsibility.”

https://www.apnews.com/4997bb7aa36c45449b488e19ac83e86d

13 comments:

  1. Sooner or later it was bound to happen: A rogue scientist in China claims to have edited a gene in two human embryos and implanted them in the mother’s womb, resulting in the birth of genetically altered twin girls. We’re no longer in the realm of science fiction. If true, this hacking of their biological operating instructions, which they will pass on to their children and generations to come, is a dangerous breach of medical ethics and responsible research and must be condemned.

    This is not to say that medicine won’t someday employ gene-editing technologies in similar ways. But that time has not arrived. There are still too many risks, too many unknowns, about tinkering with our heritable genetic blueprints.

    In recent years genome editing has been appropriately heralded as the most important advance of biotechnology of our generation, and most likely the past century. Known as Crispr, this technique, and related DNA editing tools, enable their users to cut and paste discrete letters of the genome. This ability has markedly advanced science, shedding new light on the complex human genome with its billions of A, C, T and G letters that are the architecture of who we are.

    Already, many clinical trials using this technology are underway involving patients with rare diseases like hemophilia, thalassemia and sickle cell anemia. The difference between these efforts and what reportedly happened in China is that these genome editing trials involve cells from the patient’s body. The manipulations are not transmissible to the next generation.(continued)

    ReplyDelete
  2. These trials are in their early stages, and we don’t yet have results to show whether this type of editing is safe or provides effective treatment. But whatever happens, the consequences are confined to a patient who has consented to the experimental treatment.

    While there have been reports of human embryo editing experiments in laboratory petri dishes, until now, so far as we know, none of these embryos have been implanted in humans. In such cases, the hazards become markedly amplified because the editing intervenes in so-called germ line cells that are transmitted from one generation to another. Conceivably every cell in the body, some 37 trillion, could be programmed with the edits.

    In the Chinese study, led by He Jiankui, a physicist, not a medical doctor, on the faculty of Southern University of Science and Technology in Shenzhen, the targeted gene was CCR5, which helps enable H.I.V. to enter cells. While preventing this intrusion might sound like an advance in the fight against AIDS, it is completely unnecessary and may even carry the hazard of increasing the subject’s susceptibility to other types of infection, such as influenza and West Nile fever. Previous genome editing studies have shown it is possible to disable the CCR5 gene in adults without working at the embryo level.

    So the experiment was needless. It had no scientific basis and must be considered unethical when balanced against the known and unknown risks.

    The predominant risks are the potential impacts of the editing on other letters of the genome, which could induce diseases. We don’t have the assurance yet that Crispr provides laserlike precision in editing — for example, certain important genes for suppressing cancer are particularly susceptible to unintended editing. The way we assess this risk is to sequence the genome before and after editing, to see whether changes were made in genes other than the target gene.
    But our ability to discern these changes is still rudimentary, and it is entirely likely that we will miss something. The fact that we may not have seen unintended mutations brought about by editing is by no means proof of their absence.

    With six billion letters in the genome that could be affected, the risk of unintended editing is considerable and requires extensive scrutiny to understand and mitigate. That’s partly why the implantation of edited human embryos has been widely banned…

    We don’t know whether the intended human genome editing was achieved in the twins and have no idea whether it will prove to be safe if it was accomplished.

    But we can conclude that this was a misguided, reckless use of powerful gene-altering tools to create edited human beings. We should not proceed down this road until we know far more about the consequences of what we are doing.

    https://www.nytimes.com/2018/11/27/opinion/genetically-edited-babies-china.html

    ReplyDelete
  3. The news has broken about one of the most significant experiments ever undertaken in the history of human genetics: an attempt to alter genes in a human embryo. A Chinese scientist reported that he had done so in at least two girls who have now been born. And he says that more children may be on the way. He used the new technique CRISPR, which basically involves snipping genes out or inserting genes. It's kind of like a scissors that can be used to alter the genetics in a body cell or, in the case of human embryos, in the embryonic cell itself. The reason I say that this experiment is hugely significant is because it's the first known attempt to try to alter genetic material that will be passed on to future generations. It's not the same as gene therapy, where you try to change genes in the cells of the body...

    I think it's one of the most amazing experiments ever seen and stupendously immoral.

    This work was done without the consensus of the scientific community that CRISPR is safe enough to alter genes in embryos. There have been a few experiments in human embryos in dishes and some experiments in animal embryos. But we are far from knowing whether we can accurately target a particular set of genes and change, delete, or alter them without other changes taking place in other parts of the genome. Were that to happen, you could get birth defects, premature aging, and all manner of sickness and illness occurring in the children. When it comes to something like germline genetic engineering, safety [should come] first. That has been violated and ignored here.

    Second, the experiment is grandstanding. All of the announcements from China have come via YouTube and press conferences. That is no way to do momentous scientific research. Nothing can be validated. No one knows what methods were used. No one can assess the data. There is no way to track what really was done, and that is completely morally irresponsible.

    Worse, the researcher says that he spent 2 hours with the women who were going to have their embryos altered, getting their informed consent. This researcher has applied for patents and he is running a fertility clinic where people are drawn to spend a lot of money to perhaps alter their children's genes. In this case, the goal of the alteration was to try to get more resistance to HIV. Apparently, the women's husbands were infected with HIV, so they were looking for some way to prevent transmission to the children. But the goal, in a sense, does not matter. Explaining this in 2 hours by a person who has conflicts of interest, who is making money trying to become first in line [in this area] and promoting a patented technique, is not informed consent. It's bamboozlement…

    I'm not against germline genetic engineering. I think repairing diseases like sickle cell, Huntington's disease, or hemophilia is a great application of the technique—but only once it's proven safe and reliable, and in the hands of people known to be competent at handling engineering in something like an embryo. That is years away. It's going to come. I hope it comes quickly.

    Renegade science like this offends both basic principles of human experimentation and scientific consensus about safety. It endangers the field; this experiment could set back germline engineering rather than advance it as people react with prohibitions or withdraw funds.

    Some people see Professor He, who did this experiment in China, and his team, as heroes. I see them as villains. I think they have done something hugely immoral and threatening to the future of important science, and they set a horrible example for anyone interested in trying to undertake pioneering research in the realm of genetics.

    https://www.medscape.com/viewarticle/905598

    ReplyDelete
  4. In 2010, a Rice University graduate student named He Jiankui published a paper describing the nitty-gritty details of a then-arcane bacterial immune system called CRISPR. It was well before scientists unlocked the knowledge that CRISPR could be used to manipulate DNA with a precision and ease that other genome editors lacked.

    Now, just eight years later, and after returning to his native China, He has burst onto the global stage in the most spectacular way — an entrance he seems to have meticulously choreographed even as he ignored guidelines established by global scientific panels. Using YouTube rather than an academic journal, He claimed that with the aid of CRISPR, he had helped create the world’s first babies — twin girls born a few weeks ago — whose genomes had been edited as embryos. The announcement dropped like a surprise Beyonce album.

    The claim, which has not been verified by outside researchers, was heralded by a few as a scientific milestone, an unprecedented step toward preventing all sorts of diseases. But others viewed it as quite the opposite: the reckless breaking of a scientific taboo for personal gain. After all, many far more experienced and respected researchers had the technical know-how to attempt what He did, but they honored the widely accepted ethical barriers. Such research is also illegal in the United States.

    He Jiankui (pronounced HEH JEE’-an-qway), who was previously not known to many Western CRISPR experts, immediately found himself at the center of a firestorm of criticism over the lack of transparency in his research, the choice of gene that was edited, and his pursuit of this research at all.(continued)

    ReplyDelete
  5. (continued)
    “I am trying to understand what may have motivated the work he describes,” said a scientist who helped organize a major Hong Kong summit on human genome editing that starts Tuesday and who asked not to be named. “As far as I can tell, it was a combination of hubris, naivete, and perhaps a genuine desire to help people in need. He does not seem to have anticipated the profound public backlash against his work and the way it was conducted and publicized.”…

    Even He’s own university, from which he has been on leave since February, distanced itself from the research, saying that it knew nothing about the work and that it was “conducted outside of the campus” and “seriously violated academic ethics and codes of conduct.”…

    Beyond the condemnation, leading genome editing experts were simply left puzzled by the researcher behind the announcement. He doesn’t have an extensive history of publishing papers about genome editing or embryology or the gene he disabled in the embryos, CCR5, to try to confer resistance to HIV infection. Much of the social media scuttlebutt came back to this question: What was this guy doing editing embryos?...

    He also co-authored a paper published this week in the CRISPR Journal proposing five “ethical principles” to guide the use of genome editing in the clinic. The second author on the paper: Ryan Ferrell, the American public relations professional who worked with He on the rollout of the CRISPR’d embryos project.

    Among the elements He called for: open dialogue…(continued)

    ReplyDelete
  6. (continued)In April 2016, He wrote to CRISPR pioneer Feng Zhang of the Broad Institute, identifying himself as CEO of his DNA sequencing company Direct Genomics and requesting a tour of Zhang’s lab in Cambridge, Mass. He did not visit then, but he did briefly drop by to visit Zhang’s lab this past August, Zhang told STAT on the sidelines of the genome editing summit.

    What did He want to talk about? Methods to reduce off-target effects — cuts CRISPR can make in unintended places — in editing of mouse and human embryos, Zhang recalled.

    “It was clear to me that he was having the same challenges as other researchers around lack of efficiency and lack of precision,” Zhang said. “I told him that the technology is neither efficient nor precise enough for real-world application in embryos, including in human IVF applications.”

    For Zhang, that conversation has taken on a new tone in light of this week’s news. “Of course, now we know that by August, he must have been quite far along in his work in humans, at least according to his most recent claims,” Zhang said. “He certainly never mentioned this work when I saw him.”

    He’s career trajectory highlights a few salient facts: His scientific background is wide-ranging, but lacking in deep expertise in CRISPR and embryology. Public records indicate that he is just 34 years old, an age when many top researchers are just opening up their first labs.

    He, who appears to have been born and raised in China, earned his undergraduate degree in physics from the University of Science and Technology of China in 2006. He then moved to Texas to earn his Ph.D., in biophysics, at Rice University in Houston; his adviser there, the bioengineering professor Michael Deem, would later collaborate with him on the CRISPR’d embryos project.

    After graduating in 2010, He spent about a year doing his postdoc in the lab of Stanford bioengineer Stephen Quake. It was there that He learned about single-molecule sequencing ― a then-revolutionary method of sequencing single DNA molecules without first having to copy them using polymerase chain reaction, according to a 2015 story in the publication Bio-IT World…

    In his 2017 talk, He raised the 1999 death of Jesse Gelsinger in an early gene therapy trial, which set that field back by more than a decade.

    Researchers who might be tempted to use a CRISPR’d embryo to start a pregnancy should remember that case, He suggested.

    “I want to remind everyone that we should do this kind of slow and with a bit of caution,” He said, “because a single case of failure may kill the entire field.”

    https://www.statnews.com/2018/11/26/he-jiankui-gene-edited-babies-china/

    ReplyDelete
  7. He Jiankui, Ryan Ferrell, Chen Yuanlin, Qin Jinzhou, and Chen Yangran. Draft Ethical Principles for Therapeutic Assisted Reproductive Technologies. The CRISPR Journal. In press. Published Online:28 Nov 2018https://doi.org/10.1089/crispr.2018.0051

    Abstract
    The germline gene-editing field has several detailed preclinical research guidelines but lacks concise and plain-language ethics statements on ultimate clinical uses. The general public deserves a clear vision of the future to gauge the field's ultimate intentions and have meaningful input and discussion about its progress. We propose the identification of a core set of fundamental human values to frame, guide, and restrict clinical applications that communities around the world can share and localize based on religious beliefs, culture, and public-health challenges.

    ReplyDelete
  8. The whereabouts of He Jiankui, a Chinese scientist who claims to have created the world’s first gene-edited babies, remain unknown after his former workplace refuted media reports that he was detained over the weekend.

    A spokeswoman for China’s Southern University of Science and Technology said that media reports of He’s detention were inaccurate but added that the university would be unable to answer any further questions regarding the matter, Hong Kong’s South China Morning Post reported.

    The scientist stunned the medical community last week in a YouTube video, in which he claimed to have altered embryos for a couple during fertility treatments.

    At a conference in Hong Kong on Wednesday, he gave his first public remarks on the DNA manipulation, saying that he altered the genetic material of two human embryos with the aim of making them resistant to HIV infections. The former Stanford University postdoctoral fellow then claimed that the gene-edited twin girls were born “normal and healthy” earlier this month.

    His controversial revelation earned him the nickname of ‘Chinese Frankenstein’ and was met with a huge backlash from the wider scientific community. Similar forms of gene-editing are banned in the U.S., amid concerns about the consequences that such alterations have on future generations.

    The Chinese scientist is now facing an investigation from China’s National Health Commission on the experiment, with a top official at the Ministry of Science and Technology telling state media that He’s work was “extremely abominable.”

    http://time.com/5469111/he-jiankui-scientist-missing-gene-edited-babies/

    ReplyDelete
  9. Doudna, who co-led a 2012 study showing that a weird bacterial immune system called CRISPR could edit DNA as niftily as Word edits documents, and hundreds of other experts were in Hong Kong for the International Human Genome Editing Summit. He Jiankui, who was scheduled to speak at the summit on Wednesday, had asked to meet privately with Doudna, one of the summit’s organizers. In his presentation, He had planned to talk about the ethics of embryo editing and his experiments on mouse, monkey, and human embryos, with nary a hint that two of those embryos were now living, breathing, baby girls whom He, in an astonishing YouTube birth announcement, called Nana and Lulu. Was that okay?, he asked Doudna as they sat in the lobby.

    Um, Doudna replied, you’ve dropped this shocking news on the world, right before our summit, and you’re not planning to mention it? He seemed surprised that she expected him to but agreed to have dinner with her and other members of the summit organizing committee that evening to talk it out.

    “His demeanor was an odd combination of hubris and naivete,” she recalled in an interview. “He was very confident in his work, and totally not understanding what an explosion he had caused” — one that, some scientists feared, could derail hopes for using CRISPR to prevent some of the most devastating diseases lurking in the double helix….

    He confided in at least two U.S. scientists about his plan, but ignored their arguments that he was making a potentially disastrous mistake. He studied recommended ethical guidelines for embryo editing — but flouted them. He claimed he had been transparent about working toward pregnancies with CRISPR’d embryos — yet never breathed a word about those plans in his talks at science meetings and stalled for months before listing his experiment on an official Chinese registry of clinical trials.

    For a driven and fame-seeking scientist who had set his star on changing the world, heeding doubters and sticklers wasn’t part of the plan.

    He believed he would be hailed for his scientific first, especially in his homeland, as someone who did for China what the Sputnik engineers did for the old Soviet Union. In conversations with scientists and others, he brought up Dr. Robert Edwards, part of the team who created the world’s first test tube baby, won the 2010 Nobel prize for it, and brought joy to millions of otherwise infertile couples.

    No wonder He seemed stunned that Monday, as worldwide condemnation of his work grew and even the stars of the CRISPR firmament weren’t applauding him. Over the hastily arranged Cantonese buffet dinner at Le Méridien, Doudna and three other summit organizers peppered him with technical questions (How many embryos did you try to edit with CRISPR? How many succeeded? How did you decide which embryos to implant? What tests did you run to see if the editing worked as planned?) and challenged the ethics of the experiment (Why did you pick the gene CCR5, which is involved in HIV infection, to edit? Did the parents understand the risks to their potential child? How do you know?).

    After just over an hour, He had enough, participants told STAT. He pulled some cash out of his pocket, threw it on the table, and stormed off. Fearful of his safety, he left the Méridien and checked into another hotel. His dinner companions were left wondering if he would even show up for his scheduled talk at the summit on Wednesday…

    He had written no important CRISPR papers before his shocking announcement. (He still hasn’t: The CRISPR babies experiment remains unpublished, and a study editing mouse, monkey, and human embryos without starting pregnancies has been rejected.) He was on no one’s radar screen…(continued)

    ReplyDelete
  10. (continued) Maybe we should invite him, Doudna proposed to Hurlbut. They did. He came. On the workshop’s second day, in a session called “Evolution and Human Development,” He presented work on using CRISPR to edit mouse, monkey, and human embryos (without pregnancies). His talk did not leave much of an impression, “and I don’t think it was received very well,” Doudna said.

    That was partly because He was, in a sense, two years late. In 2015, scientists at Sun Yat-sen University in Guangzhou used CRISPR to edit the gene whose abnormality causes the often-fatal blood disease beta thalassemia. Their experiment, which also sent shock waves around the world, used nonviable IVF embryos. He, too, was using nonviable embryos. It didn’t seem like he was moving the science forward.

    Worse, another attendee recalled, scientists said He’s “science was sloppy and the application unnecessary.” One biologist challenged He on technical details of his work, especially how he analyzed the edited genomes for the unintended edits called off-target effects, a critical safety concern.

    Other scholars who attended were struck by what Harvard’s Sheila Jasanoff called He’s “great smoothness.” Although He did not explicitly discuss his ethical views, Jasanoff said, he “clearly did not have deep misgivings about plugging ahead with gene editing, and I sensed no exposure to the sorts of ethical debates our guys are routinely involved in.”…

    But now something new entered the discussions. He told DeWitt he was planning to start pregnancies with CRISPR’d human embryos. DeWitt was aghast, he told STAT, and argued that there was no justification for such an experiment. The technology simply wasn’t ready to use on babies-to-be….

    “I knew where he was heading,” Hurlbut said. “I tried to give him a sense of the practical and moral implications,” including ethical objections to research on human embryos. He pushed back; wasn’t it only a fringe group in the U.S. that adamantly opposes that?, he asked; if CRISPR can be used to prevent a dreaded genetic disorder in a baby who would otherwise inherit it, why should we hold a one-cell embryo in the same ethical regard as a suffering child?

    “My overall feeling,” Hurlbut said, “was that he’s a well-meaning person who wants his efforts to count for good.”…

    In July 2017 He gave an updated version of his Berkeley talk, at a meeting on “Genome Engineering: The CRISPR-Cas Revolution” at Cold Spring Harbor Laboratory on New York’s Long Island. The data from his experiments in mouse, monkey, and human embryos included ways to improve CRISPR’s efficiency and measurements of its accuracy. He had injected CRISPR into the first human embryo, he said, on Nov. 10, 2016, doing two or three each month (though four that December). He reminded his audience of the many ways embryo editing could fail, including off-target edits and mosaicism (when only some of an organism’s cells are edited, creating a genetic patchwork with unknown implications for health).

    Viewing He as less than a heavy hitter in the genome-editing world, many skipped his talk. Computational biologist Max Haeussler of UC Santa Cruz, who shared a double room with He and did attend, is struck in retrospect at He’s discussing how dangerous editing human embryos is. “I found this remark already strange back then,” he said. “Everyone in the room knew that it’s out of the question to edit human embryos. Why mention that it’s dangerous?”(continued)

    ReplyDelete
  11. (continued) In the room they shared, He and Haeussler talked shop, including how to detect off-target edits and how deeply He probed to find any. One red flag was that He was doing what’s called “short read sequencing,” meaning he sequenced short segments of DNA. That can miss big rearrangements along a chromosome. It’s similar to proofreading a document a sentence or two at a time….

    After dashing across town for a 3:30 press conference, where they mostly begged off on commenting about He’s unverified claim, several of the organizers crowded into a van for the return trip — and, for some of them, dinner with He in the hotel’s Nam Fong restaurant.

    “That,” said bioethicist and organizing committee member Alta Charo of the University of Wisconsin, “was an interesting dinner.”

    Over the Cantonese buffet, Charo fired questions at He, focusing on the bioethics of his experiment: how he recruited families, what he told them, why he chose CCR5. He opened his laptop and showed a spreadsheet, but it was in Chinese, leaving the scientists hardly better informed about key details. Asked why he had kept his experiment a secret, He said he had presented his work at science meetings. But he had never said he was going to establish pregnancies, and seemed not to understand why that was such a great leap beyond the embryo experiments he did talk about.

    https://www.statnews.com/2018/12/17/crispr-shocker-genome-editing-scientist-he-jiankui/

    ReplyDelete
  12. The journal Nature has named its Top 10 people who mattered in science in 2018, and there’s one controversial name on the list.

    Chinese scientist He Jiankui captured global attention last month when he announced he had edited the genomes of twin baby girls, though neither proof nor a peer-reviewed journal paper was offered. This drew widespread condemnation over concerns that he had ignored ethical considerations and exposed the infants – if they exist – to potential risks.

    But then, as Nature’s chief features editor Rick Monastersky says, some memorable scientific events “force us to confront difficult questions about who we are, where we have come from, and where we are going”.

    https://cosmosmagazine.com/society/nature-journal-nominates-its-top-10-people-for-2018

    ReplyDelete
  13. Chinese scientist He Jiankui shook the world last month when he claimed that he had created the world’s first gene-edited babies.

    On Thursday, He was named by the science journal Nature as a member of a select group of 10 people “who mattered” in 2018.


    But unlike the nine other nominees, mostly scientists, He stands out on the list as a figure whose work’s impact on humanity is questionable at best.

    Nature called the Chinese scientist the “CRISPR rogue,” referring to the shorthand for the technique he claimed to have used to edit the genes of embryos that have resulted in the birth of twin girls, Lulu and Nana.

    The other nine people on the list include scientists who advanced the understanding of nature and ourselves, as well as advocates for environmental protection and diversity in science.

    But the journal said that when the geneticist altered the genomes of two human embryos, as he has claimed, He knew he was “crossing a new bioethical boundary.”

    He initially defended himself, telling a packed auditorium at a conference last month that he was proud of his research and believed it was a viable method for curing inherited diseases.

    He’s claims haven’t been independently verified or published in a peer-reviewed journal. But the procedure He claims to have carried out has been criticized by prominent scientists worldwide as unnecessary, unethical and risky.

    Since then, China’s science authorities have moved to cut ties with and condemned He.

    The Chinese Ministry of Science and Technology has ordered research institutes to suspend He’s projects.

    He has not made further public statements or appearances since the November 28 conference.

    https://www.inkstonenews.com/science/chinese-scientist-he-jiankui-chosen-nature-10-people-who-mattered-2018/article/2178912

    ReplyDelete