Tag Archives: Dietram Scheufele

CRISPR and editing the germline in the US (part 3 of 3): public discussions and pop culture

After giving a basic explanation of the technology and some of the controversies in part 1 and offering more detail about the technology and about the possibility of designer babies in part 2; this part covers public discussion, a call for one and the suggestion that one is taking place in popular culture.

But a discussion does need to happen

In a move that is either an exquisite coincidence or has been carefully orchestrated (I vote for the latter), researchers from the University of Wisconsin-Madison have released a study about attitudes in the US to human genome editing. From an Aug. 11, 2017 University of Wisconsin-Madison news release (also on EurekAllert),

In early August 2017, an international team of scientists announced they had successfully edited the DNA of human embryos. As people process the political, moral and regulatory issues of the technology — which nudges us closer to nonfiction than science fiction — researchers at the University of Wisconsin-Madison and Temple University show the time is now to involve the American public in discussions about human genome editing.

In a study published Aug. 11 in the journal Science, the researchers assessed what people in the United States think about the uses of human genome editing and how their attitudes may drive public discussion. They found a public divided on its uses but united in the importance of moving conversations forward.

“There are several pathways we can go down with gene editing,” says UW-Madison’s Dietram Scheufele, lead author of the study and member of a National Academy of Sciences committee that compiled a report focused on human gene editing earlier this year. “Our study takes an exhaustive look at all of those possible pathways forward and asks where the public stands on each one of them.”

Compared to previous studies on public attitudes about the technology, the new study takes a more nuanced approach, examining public opinion about the use of gene editing for disease therapy versus for human enhancement, and about editing that becomes hereditary versus editing that does not.

The research team, which included Scheufele and Dominique Brossard — both professors of life sciences communication — along with Michael Xenos, professor of communication arts, first surveyed study participants about the use of editing to treat disease (therapy) versus for enhancement (creating so-called “designer babies”). While about two-thirds of respondents expressed at least some support for therapeutic editing, only one-third expressed support for using the technology for enhancement.

Diving even deeper, researchers looked into public attitudes about gene editing on specific cell types — somatic or germline — either for therapy or enhancement. Somatic cells are non-reproductive, so edits made in those cells do not affect future generations. Germline cells, however, are heritable, and changes made in these cells would be passed on to children.

Public support of therapeutic editing was high both in cells that would be inherited and those that would not, with 65 percent of respondents supporting therapy in germline cells and 64 percent supporting therapy in somatic cells. When considering enhancement editing, however, support depended more upon whether the changes would affect future generations. Only 26 percent of people surveyed supported enhancement editing in heritable germline cells and 39 percent supported enhancement of somatic cells that would not be passed on to children.

“A majority of people are saying that germline enhancement is where the technology crosses that invisible line and becomes unacceptable,” says Scheufele. “When it comes to therapy, the public is more open, and that may partly be reflective of how severe some of those genetically inherited diseases are. The potential treatments for those diseases are something the public at least is willing to consider.”

Beyond questions of support, researchers also wanted to understand what was driving public opinions. They found that two factors were related to respondents’ attitudes toward gene editing as well as their attitudes toward the public’s role in its emergence: the level of religious guidance in their lives, and factual knowledge about the technology.

Those with a high level of religious guidance in their daily lives had lower support for human genome editing than those with low religious guidance. Additionally, those with high knowledge of the technology were more supportive of it than those with less knowledge.

While respondents with high religious guidance and those with high knowledge differed on their support for the technology, both groups highly supported public engagement in its development and use. These results suggest broad agreement that the public should be involved in questions of political, regulatory and moral aspects of human genome editing.

“The public may be split along lines of religiosity or knowledge with regard to what they think about the technology and scientific community, but they are united in the idea that this is an issue that requires public involvement,” says Scheufele. “Our findings show very nicely that the public is ready for these discussions and that the time to have the discussions is now, before the science is fully ready and while we have time to carefully think through different options regarding how we want to move forward.”

Here’s a  link to and a citation for the paper,

U.S. attitudes on human genome editing by Dietram A. Scheufele, Michael A. Xenos, Emily L. Howell, Kathleen M. Rose, Dominique Brossard1, and Bruce W. Hardy. Science 11 Aug 2017: Vol. 357, Issue 6351, pp. 553-554 DOI: 10.1126/science.aan3708

This paper is behind a paywall.

A couple of final comments

Briefly, I notice that there’s no mention of the ethics of patenting this technology in the news release about the study.

Moving on, it seems surprising that the first team to engage in germline editing in the US is in Oregon; I would have expected the work to come from Massachusetts, California, or Illinois where a lot of bleeding edge medical research is performed. However, given the dearth of financial support from federal funding institutions, it seems likely that only an outsider would dare to engage i the research. Given the timing, Mitalipov’s work was already well underway before the recent about-face from the US National Academy of Sciences (Note: Kaiser’s Feb. 14, 2017 article does note that for some the recent recommendations do not represent any change).

As for discussion on issues such as editing of the germline, I’ve often noted here that popular culture (including advertising with the science fiction and other dramas laid in various media) often provides an informal forum for discussion. Joelle Renstrom in an Aug. 13, 2017 article for slate.com writes that Orphan Black (a BBC America series featuring clones) opened up a series of questions about science and ethics in the guise of a thriller about clones. She offers a précis of the first four seasons (Note: A link has been removed),

If you stopped watching a few seasons back, here’s a brief synopsis of how the mysteries wrap up. Neolution, an organization that seeks to control human evolution through genetic modification, began Project Leda, the cloning program, for two primary reasons: to see whether they could and to experiment with mutations that might allow people (i.e., themselves) to live longer. Neolution partnered with biotech companies such as Dyad, using its big pharma reach and deep pockets to harvest people’s genetic information and to conduct individual and germline (that is, genetic alterations passed down through generations) experiments, including infertility treatments that result in horrifying birth defects and body modification, such as tail-growing.

She then provides the article’s thesis (Note: Links have been removed),

Orphan Black demonstrates Carl Sagan’s warning of a time when “awesome technological powers are in the hands of a very few.” Neolutionists do whatever they want, pausing only to consider whether they’re missing an opportunity to exploit. Their hubris is straight out of Victor Frankenstein’s playbook. Frankenstein wonders whether he ought to first reanimate something “of simpler organisation” than a human, but starting small means waiting for glory. Orphan Black’s evil scientists embody this belief: if they’re going to play God, then they’ll control not just their own destinies, but the clones’ and, ultimately, all of humanity’s. Any sacrifices along the way are for the greater good—reasoning that culminates in Westmoreland’s eugenics fantasy to genetically sterilize 99 percent of the population he doesn’t enhance.

Orphan Black uses sci-fi tropes to explore real-world plausibility. Neolution shares similarities with transhumanism, the belief that humans should use science and technology to take control of their own evolution. While some transhumanists dabble in body modifications, such as microchip implants or night-vision eye drops, others seek to end suffering by curing human illness and aging. But even these goals can be seen as selfish, as access to disease-eradicating or life-extending technologies would be limited to the wealthy. Westmoreland’s goal to “sell Neolution to the 1 percent” seems frighteningly plausible—transhumanists, who statistically tend to be white, well-educated, and male, and their associated organizations raise and spend massive sums of money to help fulfill their goals. …

On Orphan Black, denial of choice is tantamount to imprisonment. That the clones have to earn autonomy underscores the need for ethics in science, especially when it comes to genetics. The show’s message here is timely given the rise of gene-editing techniques such as CRISPR. Recently, the National Academy of Sciences gave germline gene editing the green light, just one year after academy scientists from around the world argued it would be “irresponsible to proceed” without further exploring the implications. Scientists in the United Kingdom and China have already begun human genetic engineering and American scientists recently genetically engineered a human embryo for the first time. The possibility of Project Leda isn’t farfetched. Orphan Black warns us that money, power, and fear of death can corrupt both people and science. Once that happens, loss of humanity—of both the scientists and the subjects—is inevitable.

In Carl Sagan’s dark vision of the future, “people have lost the ability to set their own agendas or knowledgeably question those in authority.” This describes the plight of the clones at the outset of Orphan Black, but as the series continues, they challenge this paradigm by approaching science and scientists with skepticism, ingenuity, and grit. …

I hope there are discussions such as those Scheufele and Brossard are advocating but it might be worth considering that there is already some discussion underway, as informal as it is.

-30-

Part 1: CRISPR and editing the germline in the US (part 1 of 3): In the beginning

Part 2: CRISPR and editing the germline in the US (part 2 of 3): ‘designer babies’?

CRISPR and editing the germline in the US (part 2 of 3): ‘designer babies’?

Having included an explanation of CRISPR-CAS9 technology along with the news about the first US team to edit the germline and bits and pieces about ethics and a patent fight (part 1), this part hones in on the details of the work and worries about ‘designer babies’.

The interest flurry

I found three articles addressing the research and all three concur that despite some of the early reporting, this is not the beginning of a ‘designer baby’ generation.

First up was Nick Thieme in a July 28, 2017 article for Slate,

MIT Technology Review reported Thursday that a team of researchers from Portland, Oregon were the first team of U.S.-based scientists to successfully create a genetically modified human embryo. The researchers, led by Shoukhrat Mitalipov of Oregon Health and Science University, changed the DNA of—in MIT Technology Review’s words—“many tens” of genetically-diseased embryos by injecting the host egg with CRISPR, a DNA-based gene editing tool first discovered in bacteria, at the time of fertilization. CRISPR-Cas9, as the full editing system is called, allows scientists to change genes accurately and efficiently. As has happened with research elsewhere, the CRISPR-edited embryos weren’t implanted—they were kept sustained for only a couple of days.

In addition to being the first American team to complete this feat, the researchers also improved upon the work of the three Chinese research teams that beat them to editing embryos with CRISPR: Mitalipov’s team increased the proportion of embryonic cells that received the intended genetic changes, addressing an issue called “mosaicism,” which is when an embryo is comprised of cells with different genetic makeups. Increasing that proportion is essential to CRISPR work in eliminating inherited diseases, to ensure that the CRISPR therapy has the intended result. The Oregon team also reduced the number of genetic errors introduced by CRISPR, reducing the likelihood that a patient would develop cancer elsewhere in the body.

Separate from the scientific advancements, it’s a big deal that this work happened in a country with such intense politicization of embryo research. …

But there are a great number of obstacles between the current research and the future of genetically editing all children to be 12-foot-tall Einsteins.

Ed Yong in an Aug. 2, 2017 article for The Atlantic offered a comprehensive overview of the research and its implications (unusually for Yong, there seems to be mildly condescending note but it’s worth ignoring for the wealth of information in the article; Note: Links have been removed),

… the full details of the experiment, which are released today, show that the study is scientifically important but much less of a social inflection point than has been suggested. “This has been widely reported as the dawn of the era of the designer baby, making it probably the fifth or sixth time people have reported that dawn,” says Alta Charo, an expert on law and bioethics at the University of Wisconsin-Madison. “And it’s not.”

Given the persistent confusion around CRISPR and its implications, I’ve laid out exactly what the team did, and what it means.

Who did the experiments?

Shoukhrat Mitalipov is a Kazakhstani-born cell biologist with a history of breakthroughs—and controversy—in the stem cell field. He was the scientist to clone monkeys. He was the first to create human embryos by cloning adult cells—a move that could provide patients with an easy supply of personalized stem cells. He also pioneered a technique for creating embryos with genetic material from three biological parents, as a way of preventing a group of debilitating inherited diseases.

Although MIT Tech Review name-checked Mitalipov alone, the paper splits credit for the research between five collaborating teams—four based in the United States, and one in South Korea.

What did they actually do?

The project effectively began with an elevator conversation between Mitalipov and his colleague Sanjiv Kaul. Mitalipov explained that he wanted to use CRISPR to correct a disease-causing gene in human embryos, and was trying to figure out which disease to focus on. Kaul, a cardiologist, told him about hypertrophic cardiomyopathy (HCM)—an inherited heart disease that’s commonly caused by mutations in a gene called MYBPC3. HCM is surprisingly common, affecting 1 in 500 adults. Many of them lead normal lives, but in some, the walls of their hearts can thicken and suddenly fail. For that reason, HCM is the commonest cause of sudden death in athletes. “There really is no treatment,” says Kaul. “A number of drugs are being evaluated but they are all experimental,” and they merely treat the symptoms. The team wanted to prevent HCM entirely by removing the underlying mutation.

They collected sperm from a man with HCM and used CRISPR to change his mutant gene into its normal healthy version, while simultaneously using the sperm to fertilize eggs that had been donated by female volunteers. In this way, they created embryos that were completely free of the mutation. The procedure was effective, and avoided some of the critical problems that have plagued past attempts to use CRISPR in human embryos.

Wait, other human embryos have been edited before?

There have been three attempts in China. The first two—in 2015 and 2016—used non-viable embryos that could never have resulted in a live birth. The third—announced this March—was the first to use viable embryos that could theoretically have been implanted in a womb. All of these studies showed that CRISPR gene-editing, for all its hype, is still in its infancy.

The editing was imprecise. CRISPR is heralded for its precision, allowing scientists to edit particular genes of choice. But in practice, some of the Chinese researchers found worrying levels of off-target mutations, where CRISPR mistakenly cut other parts of the genome.

The editing was inefficient. The first Chinese team only managed to successfully edit a disease gene in 4 out of 86 embryos, and the second team fared even worse.

The editing was incomplete. Even in the successful cases, each embryo had a mix of modified and unmodified cells. This pattern, known as mosaicism, poses serious safety problems if gene-editing were ever to be used in practice. Doctors could end up implanting women with embryos that they thought were free of a disease-causing mutation, but were only partially free. The resulting person would still have many tissues and organs that carry those mutations, and might go on to develop symptoms.

What did the American team do differently?

The Chinese teams all used CRISPR to edit embryos at early stages of their development. By contrast, the Oregon researchers delivered the CRISPR components at the earliest possible point—minutes before fertilization. That neatly avoids the problem of mosaicism by ensuring that an embryo is edited from the very moment it is created. The team did this with 54 embryos and successfully edited the mutant MYBPC3 gene in 72 percent of them. In the other 28 percent, the editing didn’t work—a high failure rate, but far lower than in previous attempts. Better still, the team found no evidence of off-target mutations.

This is a big deal. Many scientists assumed that they’d have to do something more convoluted to avoid mosaicism. They’d have to collect a patient’s cells, which they’d revert into stem cells, which they’d use to make sperm or eggs, which they’d edit using CRISPR. “That’s a lot of extra steps, with more risks,” says Alta Charo. “If it’s possible to edit the embryo itself, that’s a real advance.” Perhaps for that reason, this is the first study to edit human embryos that was published in a top-tier scientific journal—Nature, which rejected some of the earlier Chinese papers.

Is this kind of research even legal?

Yes. In Western Europe, 15 countries out of 22 ban any attempts to change the human germ line—a term referring to sperm, eggs, and other cells that can transmit genetic information to future generations. No such stance exists in the United States but Congress has banned the Food and Drug Administration from considering research applications that make such modifications. Separately, federal agencies like the National Institutes of Health are banned from funding research that ultimately destroys human embryos. But the Oregon team used non-federal money from their institutions, and donations from several small non-profits. No taxpayer money went into their work. [emphasis mine]

Why would you want to edit embryos at all?

Partly to learn more about ourselves. By using CRISPR to manipulate the genes of embryos, scientists can learn more about the earliest stages of human development, and about problems like infertility and miscarriages. That’s why biologist Kathy Niakan from the Crick Institute in London recently secured a license from a British regulator to use CRISPR on human embryos.

Isn’t this a slippery slope toward making designer babies?

In terms of avoiding genetic diseases, it’s not conceptually different from PGD, which is already widely used. The bigger worry is that gene-editing could be used to make people stronger, smarter, or taller, paving the way for a new eugenics, and widening the already substantial gaps between the wealthy and poor. But many geneticists believe that such a future is fundamentally unlikely because complex traits like height and intelligence are the work of hundreds or thousands of genes, each of which have a tiny effect. The prospect of editing them all is implausible. And since genes are so thoroughly interconnected, it may be impossible to edit one particular trait without also affecting many others.

“There’s the worry that this could be used for enhancement, so society has to draw a line,” says Mitalipov. “But this is pretty complex technology and it wouldn’t be hard to regulate it.”

Does this discovery have any social importance at all?

“It’s not so much about designer babies as it is about geographical location,” says Charo. “It’s happening in the United States, and everything here around embryo research has high sensitivity.” She and others worry that the early report about the study, before the actual details were available for scrutiny, could lead to unnecessary panic. “Panic reactions often lead to panic-driven policy … which is usually bad policy,” wrote Greely [bioethicist Hank Greely].

As I understand it, despite the change in stance, there is no federal funding available for the research performed by Mitalipov and his team.

Finally, University College London (UCL) scientists Joyce Harper and Helen O’Neill wrote about CRISPR, the Oregon team’s work, and the possibilities in an Aug. 3, 2017 essay for The Conversation (Note: Links have been removed),

The genome editing tool used, CRISPR-Cas9, has transformed the field of biology in the short time since its discovery in that it not only promises, but delivers. CRISPR has surpassed all previous efforts to engineer cells and alter genomes at a fraction of the time and cost.

The technology, which works like molecular scissors to cut and paste DNA, is a natural defence system that bacteria use to fend off harmful infections. This system has the ability to recognise invading virus DNA, cut it and integrate this cut sequence into its own genome – allowing the bacterium to render itself immune to future infections of viruses with similar DNA. It is this ability to recognise and cut DNA that has allowed scientists to use it to target and edit specific DNA regions.

When this technology is applied to “germ cells” – the sperm and eggs – or embryos, it changes the germline. That means that any alterations made would be permanent and passed down to future generations. This makes it more ethically complex, but there are strict regulations around human germline genome editing, which is predominantly illegal. The UK received a licence in 2016 to carry out CRISPR on human embryos for research into early development. But edited embryos are not allowed to be inserted into the uterus and develop into a fetus in any country.

Germline genome editing came into the global spotlight when Chinese scientists announced in 2015 that they had used CRISPR to edit non-viable human embryos – cells that could never result in a live birth. They did this to modify the gene responsible for the blood disorder β-thalassaemia. While it was met with some success, it received a lot of criticism because of the premature use of this technology in human embryos. The results showed a high number of potentially dangerous, off-target mutations created in the procedure.

Impressive results

The new study, published in Nature, is different because it deals with viable human embryos and shows that the genome editing can be carried out safely – without creating harmful mutations. The team used CRISPR to correct a mutation in the gene MYBPC3, which accounts for approximately 40% of the myocardial disease hypertrophic cardiomyopathy. This is a dominant disease, so an affected individual only needs one abnormal copy of the gene to be affected.

The researchers used sperm from a patient carrying one copy of the MYBPC3 mutation to create 54 embryos. They edited them using CRISPR-Cas9 to correct the mutation. Without genome editing, approximately 50% of the embryos would carry the patients’ normal gene and 50% would carry his abnormal gene.

After genome editing, the aim would be for 100% of embryos to be normal. In the first round of the experiments, they found that 66.7% of embryos – 36 out of 54 – were normal after being injected with CRIPSR. Of the remaining 18 embryos, five had remained unchanged, suggesting editing had not worked. In 13 embryos, only a portion of cells had been edited.

The level of efficiency is affected by the type of CRISPR machinery used and, critically, the timing in which it is put into the embryo. The researchers therefore also tried injecting the sperm and the CRISPR-Cas9 complex into the egg at the same time, which resulted in more promising results. This was done for 75 mature donated human eggs using a common IVF technique called intracytoplasmic sperm injection. This time, impressively, 72.4% of embryos were normal as a result. The approach also lowered the number of embryos containing a mixture of edited and unedited cells (these embryos are called mosaics).

Finally, the team injected a further 22 embryos which were grown into blastocyst – a later stage of embryo development. These were sequenced and the researchers found that the editing had indeed worked. Importantly, they could show that the level of off-target mutations was low.

A brave new world?

So does this mean we finally have a cure for debilitating, heritable diseases? It’s important to remember that the study did not achieve a 100% success rate. Even the researchers themselves stress that further research is needed in order to fully understand the potential and limitations of the technique.

In our view, it is unlikely that genome editing would be used to treat the majority of inherited conditions anytime soon. We still can’t be sure how a child with a genetically altered genome will develop over a lifetime, so it seems unlikely that couples carrying a genetic disease would embark on gene editing rather than undergoing already available tests – such as preimplantation genetic diagnosis or prenatal diagnosis – where the embryos or fetus are tested for genetic faults.

-30-

As might be expected there is now a call for public discussion about the ethics about this kind of work. See Part 3.

For anyone who started in the middle of this series, here’s Part 1 featuring an introduction to the technology and some of the issues.

CRISPR and editing the germline in the US (part 1 of 3): In the beginning

There’s been a minor flurry of interest in CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats; also known as CRISPR-CAS9), a gene-editing technique, since a team in Oregon announced a paper describing their work editing the germline. Since I’ve been following the CRISPR-CAS9 story for a while this seems like a good juncture for a more in-depth look at the topic. In this first part I’m including an introduction to CRISPR, some information about the latest US work, and some previous writing about ethics issues raised when Chinese scientists first announced their work editing germlines in 2015 and during the patent dispute between the University of California at Berkeley and Harvard University’s Broad Institute.

Introduction to CRISPR

I’ve been searching for a good description of CRISPR and this helped to clear up some questions for me (Thank you to MIT Review),

For anyone who’s been reading about science for a while, this upbeat approach to explaining how a particular technology will solve all sorts of problems will seem quite familiar. It’s not the most hyperbolic piece I’ve seen but it barely mentions any problems associated with research (for some of the problems see: ‘The interest flurry’ later in part 2).

Oregon team

Steve Connor’s July 26, 2017 article for the MIT (Massachusetts Institute of Technology) Technology Review breaks the news (Note: Links have been removed),

The first known attempt at creating genetically modified human embryos in the United States has been carried out by a team of researchers in Portland, Oregon, MIT Technology Review has learned.

The effort, led by Shoukhrat Mitalipov of Oregon Health and Science University, involved changing the DNA of a large number of one-cell embryos with the gene-editing technique CRISPR, according to people familiar with the scientific results.

Until now, American scientists have watched with a combination of awe, envy, and some alarm as scientists elsewhere were first to explore the controversial practice. To date, three previous reports of editing human embryos were all published by scientists in China.

Now Mitalipov is believed to have broken new ground both in the number of embryos experimented upon and by demonstrating that it is possible to safely and efficiently correct defective genes that cause inherited diseases.

Although none of the embryos were allowed to develop for more than a few days—and there was never any intention of implanting them into a womb—the experiments are a milestone on what may prove to be an inevitable journey toward the birth of the first genetically modified humans.

In altering the DNA code of human embryos, the objective of scientists is to show that they can eradicate or correct genes that cause inherited disease, like the blood condition beta-thalassemia. The process is termed “germline engineering” because any genetically modified child would then pass the changes on to subsequent generations via their own germ cells—the egg and sperm.

Some critics say germline experiments could open the floodgates to a brave new world of “designer babies” engineered with genetic enhancements—a prospect bitterly opposed by a range of religious organizations, civil society groups, and biotech companies.

The U.S. intelligence community last year called CRISPR a potential “weapon of mass destruction.”

Here’s a link to a citation for the groundbreaking paper,

Correction of a pathogenic gene mutation in human embryos by Hong Ma, Nuria Marti-Gutierrez, Sang-Wook Park, Jun Wu, Yeonmi Lee, Keiichiro Suzuki, Amy Koski, Dongmei Ji, Tomonari Hayama, Riffat Ahmed, Hayley Darby, Crystal Van Dyken, Ying Li, Eunju Kang, A.-Reum Park, Daesik Kim, Sang-Tae Kim, Jianhui Gong, Ying Gu, Xun Xu, David Battaglia, Sacha A. Krieg, David M. Lee, Diana H. Wu, Don P. Wolf, Stephen B. Heitner, Juan Carlos Izpisua Belmonte, Paula Amato, Jin-Soo Kim, Sanjiv Kaul, & Shoukhrat Mitalipov. Nature (2017) doi:10.1038/nature23305 Published online 02 August 2017

This paper appears to be open access.

CRISPR Issues: ethics and patents

In my May 14, 2015 posting I mentioned a ‘moratorium’ on germline research, the Chinese research paper, and the stance taken by the US National Institutes of Health (NIH),

The CRISPR technology has reignited a discussion about ethical and moral issues of human genetic engineering some of which is reviewed in an April 7, 2015 posting about a moratorium by Sheila Jasanoff, J. Benjamin Hurlbut and Krishanu Saha for the Guardian science blogs (Note: A link has been removed),

On April 3, 2015, a group of prominent biologists and ethicists writing in Science called for a moratorium on germline gene engineering; modifications to the human genome that will be passed on to future generations. The moratorium would apply to a technology called CRISPR/Cas9, which enables the removal of undesirable genes, insertion of desirable ones, and the broad recoding of nearly any DNA sequence.

Such modifications could affect every cell in an adult human being, including germ cells, and therefore be passed down through the generations. Many organisms across the range of biological complexity have already been edited in this way to generate designer bacteria, plants and primates. There is little reason to believe the same could not be done with human eggs, sperm and embryos. Now that the technology to engineer human germlines is here, the advocates for a moratorium declared, it is time to chart a prudent path forward. They recommend four actions: a hold on clinical applications; creation of expert forums; transparent research; and a globally representative group to recommend policy approaches.

The authors go on to review precedents and reasons for the moratorium while suggesting we need better ways for citizens to engage with and debate these issues,

An effective moratorium must be grounded in the principle that the power to modify the human genome demands serious engagement not only from scientists and ethicists but from all citizens. We need a more complex architecture for public deliberation, built on the recognition that we, as citizens, have a duty to participate in shaping our biotechnological futures, just as governments have a duty to empower us to participate in that process. Decisions such as whether or not to edit human genes should not be left to elite and invisible experts, whether in universities, ad hoc commissions, or parliamentary advisory committees. Nor should public deliberation be temporally limited by the span of a moratorium or narrowed to topics that experts deem reasonable to debate.

I recommend reading the post in its entirety as there are nuances that are best appreciated in the entirety of the piece.

Shortly after this essay was published, Chinese scientists announced they had genetically modified (nonviable) human embryos. From an April 22, 2015 article by David Cyranoski and Sara Reardon in Nature where the research and some of the ethical issues discussed,

In a world first, Chinese scientists have reported editing the genomes of human embryos. The results are published1 in the online journal Protein & Cell and confirm widespread rumours that such experiments had been conducted — rumours that sparked a high-profile debate last month2, 3 about the ethical implications of such work.

In the paper, researchers led by Junjiu Huang, a gene-function researcher at Sun Yat-sen University in Guangzhou, tried to head off such concerns by using ‘non-viable’ embryos, which cannot result in a live birth, that were obtained from local fertility clinics. The team attempted to modify the gene responsible for β-thalassaemia, a potentially fatal blood disorder, using a gene-editing technique known as CRISPR/Cas9. The researchers say that their results reveal serious obstacles to using the method in medical applications.

“I believe this is the first report of CRISPR/Cas9 applied to human pre-implantation embryos and as such the study is a landmark, as well as a cautionary tale,” says George Daley, a stem-cell biologist at Harvard Medical School in Boston, Massachusetts. “Their study should be a stern warning to any practitioner who thinks the technology is ready for testing to eradicate disease genes.”

….

Huang says that the paper was rejected by Nature and Science, in part because of ethical objections; both journals declined to comment on the claim. (Nature’s news team is editorially independent of its research editorial team.)

He adds that critics of the paper have noted that the low efficiencies and high number of off-target mutations could be specific to the abnormal embryos used in the study. Huang acknowledges the critique, but because there are no examples of gene editing in normal embryos he says that there is no way to know if the technique operates differently in them.

Still, he maintains that the embryos allow for a more meaningful model — and one closer to a normal human embryo — than an animal model or one using adult human cells. “We wanted to show our data to the world so people know what really happened with this model, rather than just talking about what would happen without data,” he says.

This, too, is a good and thoughtful read.

There was an official response in the US to the publication of this research, from an April 29, 2015 post by David Bruggeman on his Pasco Phronesis blog (Note: Links have been removed),

In light of Chinese researchers reporting their efforts to edit the genes of ‘non-viable’ human embryos, the National Institutes of Health (NIH) Director Francis Collins issued a statement (H/T Carl Zimmer).

“NIH will not fund any use of gene-editing technologies in human embryos. The concept of altering the human germline in embryos for clinical purposes has been debated over many years from many different perspectives, and has been viewed almost universally as a line that should not be crossed. Advances in technology have given us an elegant new way of carrying out genome editing, but the strong arguments against engaging in this activity remain. These include the serious and unquantifiable safety issues, ethical issues presented by altering the germline in a way that affects the next generation without their consent, and a current lack of compelling medical applications justifying the use of CRISPR/Cas9 in embryos.” …

The US has modified its stance according to a February 14, 2017 article by Jocelyn Kaiser for Science Magazine (Note: Links have been removed),

Editing the DNA of a human embryo to prevent a disease in a baby could be ethically allowable one day—but only in rare circumstances and with safeguards in place, says a widely anticipated report released today.

The report from an international committee convened by the U.S. National Academy of Sciences (NAS) and the National Academy of Medicine in Washington, D.C., concludes that such a clinical trial “might be permitted, but only following much more research” on risks and benefits, and “only for compelling reasons and under strict oversight.” Those situations could be limited to couples who both have a serious genetic disease and for whom embryo editing is “really the last reasonable option” if they want to have a healthy biological child, says committee co-chair Alta Charo, a bioethicist at the University of Wisconsin in Madison.

Some researchers are pleased with the report, saying it is consistent with previous conclusions that safely altering the DNA of human eggs, sperm, or early embryos—known as germline editing—to create a baby could be possible eventually. “They have closed the door to the vast majority of germline applications and left it open for a very small, well-defined subset. That’s not unreasonable in my opinion,” says genome researcher Eric Lander of the Broad Institute in Cambridge, Massachusetts. Lander was among the organizers of an international summit at NAS in December 2015 who called for more discussion before proceeding with embryo editing.

But others see the report as lowering the bar for such experiments because it does not explicitly say they should be prohibited for now. “It changes the tone to an affirmative position in the absence of the broad public debate this report calls for,” says Edward Lanphier, chairman of the DNA editing company Sangamo Therapeutics in Richmond, California. Two years ago, he co-authored a Nature commentary calling for a moratorium on clinical embryo editing.

One advocacy group opposed to embryo editing goes further. “We’re very disappointed with the report. It’s really a pretty dramatic shift from the existing and widespread agreement globally that human germline editing should be prohibited,” says Marcy Darnovsky, executive director of the Center for Genetics and Society in Berkeley, California.

Interestingly, this change of stance occurred just prior to a CRISPR patent decision (from my March 15, 2017 posting),

I have written about the CRISPR patent tussle (Harvard & MIT’s [Massachusetts Institute of Technology] Broad Institute vs the University of California at Berkeley) previously in a Jan. 6, 2015 posting and in a more detailed May 14, 2015 posting. I also mentioned (in a Jan. 17, 2017 posting) CRISPR and its patent issues in the context of a posting about a Slate.com series on Frankenstein and the novel’s applicability to our own time. This patent fight is being bitterly fought as fortunes are at stake.

It seems a decision has been made regarding the CRISPR patent claims. From a Feb. 17, 2017 article by Charmaine Distor for The Science Times,

After an intense court battle, the US Patent and Trademark Office (USPTO) released its ruling on February 15 [2017]. The rights for the CRISPR-Cas9 gene editing technology was handed over to the Broad Institute of Harvard University and the Massachusetts Institute of Technology (MIT).

According to an article in Nature, the said court battle was between the Broad Institute and the University of California. The two institutions are fighting over the intellectual property right for the CRISPR patent. The case between the two started when the patent was first awarded to the Broad Institute despite having the University of California apply first for the CRISPR patent.

Heidi Ledford’s Feb. 17, 2017 article for Nature provides more insight into the situation (Note: Links have been removed),

It [USPTO] ruled that the Broad Institute of Harvard and MIT in Cambridge could keep its patents on using CRISPR–Cas9 in eukaryotic cells. That was a blow to the University of California in Berkeley, which had filed its own patents and had hoped to have the Broad’s thrown out.

The fight goes back to 2012, when Jennifer Doudna at Berkeley, Emmanuelle Charpentier, then at the University of Vienna, and their colleagues outlined how CRISPR–Cas9 could be used to precisely cut isolated DNA1. In 2013, Feng Zhang at the Broad and his colleagues — and other teams — showed2 how it could be adapted to edit DNA in eukaryotic cells such as plants, livestock and humans.

Berkeley filed for a patent earlier, but the USPTO granted the Broad’s patents first — and this week upheld them. There are high stakes involved in the ruling. The holder of key patents could make millions of dollars from CRISPR–Cas9’s applications in industry: already, the technique has sped up genetic research, and scientists are using it to develop disease-resistant livestock and treatments for human diseases.

….

I also noted this eyebrow-lifting statistic,  “As for Ledford’s 3rd point, there are an estimated 763 patent families (groups of related patents) claiming CAS9 leading to the distinct possibility that the Broad Institute will be fighting many patent claims in the future.)

-30-

Part 2 covers three critical responses to the reporting and between them describe the technology in more detail and the possibility of ‘designer babies’.  CRISPR and editing the germline in the US (part 2 of 3): ‘designer babies’?

Part 3 is all about public discussion or, rather, the lack of and need for according to a couple of social scientists. Informally, there is some discussion via pop culture and Joelle Renstrom notes although she is focused on the larger issues touched on by the television series, Orphan Black and as I touch on in my final comments. CRISPR and editing the germline in the US (part 3 of 3): public discussions and pop culture

BRAIN and ethics in the US with some Canucks (not the hockey team) participating (part two of five)

The Brain research, ethics, and nanotechnology (part one of five) May 19, 2014 post kicked off a series titled ‘Brains, prostheses, nanotechnology, and human enhancement’ which brings together a number of developments in the worlds of neuroscience*, prosthetics, and, incidentally, nanotechnology in the field of interest called human enhancement. Parts one through four are an attempt to draw together a number of new developments, mostly in the US and in Europe. Due to my language skills which extend to English and, more tenuously, French, I can’t provide a more ‘global perspective’. Part five features a summary.

Before further discussing the US Presidential Commission for the Study of Bioethical Issues ‘brain’ meetings mentioned in part one, I have some background information.

The US launched its self-explanatory BRAIN (Brain Research through Advancing Innovative Neurotechnologies) initiative (originally called BAM; Brain Activity Map) in 2013. (You can find more about the history and details in this Wikipedia entry.)

From the beginning there has been discussion about how nanotechnology will be of fundamental use in the US BRAIN initiative and the European Union’s 10 year Human Brain Project (there’s more about that in my Jan. 28, 2013 posting). There’s also a 2013 book (Nanotechnology, the Brain, and the Future) from Springer, which, according to the table of contents, presents an exciting (to me) range of ideas about nanotechnology and brain research,

I. Introduction and key resources

1. Nanotechnology, the brain, and the future: Anticipatory governance via end-to-end real-time technology assessment by Jason Scott Robert, Ira Bennett, and Clark A. Miller
2. The complex cognitive systems manifesto by Richard P. W. Loosemore
3. Analysis of bibliometric data for research at the intersection of nanotechnology and neuroscience by Christina Nulle, Clark A. Miller, Harmeet Singh, and Alan Porter
4. Public attitudes toward nanotechnology-enabled human enhancement in the United States by Sean Hays, Michael Cobb, and Clark A. Miller
5. U.S. news coverage of neuroscience nanotechnology: How U.S. newspapers have covered neuroscience nanotechnology during the last decade by Doo-Hun Choi, Anthony Dudo, and Dietram Scheufele
6. Nanoethics and the brain by Valerye Milleson
7. Nanotechnology and religion: A dialogue by Tobie Milford

II. Brain repair

8. The age of neuroelectronics by Adam Keiper
9. Cochlear implants and Deaf culture by Derrick Anderson
10. Healing the blind: Attitudes of blind people toward technologies to cure blindness by Arielle Silverman
11. Ethical, legal and social aspects of brain-implants using nano-scale materials and techniques by Francois Berger et al.
12. Nanotechnology, the brain, and personal identity by Stephanie Naufel

III. Brain enhancement

13. Narratives of intelligence: the sociotechnical context of cognitive enhancement by Sean Hays
14. Towards responsible use of cognitive-enhancing drugs by the healthy by Henry T. Greeley et al.
15. The opposite of human enhancement: Nanotechnology and the blind chicken debate by Paul B. Thompson
16. Anticipatory governance of human enhancement: The National Citizens’ Technology Forum by Patrick Hamlett, Michael Cobb, and David Guston
a. Arizona site report
b. California site report
c. Colorado site reportd. Georgia site report
e. New Hampshire site report
f. Wisconsin site report

IV. Brain damage

17. A review of nanoparticle functionality and toxicity on the central nervous system by Yang et al.
18. Recommendations for a municipal health and safety policy for nanomaterials: A Report to the City of Cambridge City Manager by Sam Lipson
19. Museum of Science Nanotechnology Forum lets participants be the judge by Mark Griffin
20. Nanotechnology policy and citizen engagement in Cambridge, Massachusetts: Local reflexive governance by Shannon Conley

Thanks to David Bruggeman’s May 13, 2014 posting on his Pasco Phronesis blog, I stumbled across both a future meeting notice and documentation of the  Feb. 2014 meeting of the Presidential Commission for the Study of Bioethical Issues (Note: Links have been removed),

Continuing from its last meeting (in February 2014), the Presidential Commission for the Study of Bioethical Issues will continue working on the BRAIN (Brain Research through Advancing Innovative Neurotechnologies) Initiative in its June 9-10 meeting in Atlanta, Georgia.  An agenda is still forthcoming, …

In other developments, Commission staff are apparently going to examine some efforts to engage bioethical issues through plays.  I’d be very excited to see some of this happen during a Commission meeting, but any little bit is interesting.  The authors of these plays, Karen H. Rothenburg and Lynn W. Bush, have published excerpts in their book The Drama of DNA: Narrative Genomics.  …

The Commission also has a YouTube channel …

Integrating a theatrical experience into the reams of public engagement exercises that technologies such as stem cell, GMO (genetically modified organisms), nanotechnology, etc. tend to spawn seems a delightful idea.

Interestingly, the meeting in June 2014 will coincide with the book’s release date. I dug further and found these snippets of information. The book is being published by Oxford University Press and is available in both paperback and e-book formats. The authors are not playwrights, as one might assume. From the Author Information page,

Lynn Bush, PhD, MS, MA is on the faculty of Pediatric Clinical Genetics at Columbia University Medical Center, a faculty associate at their Center for Bioethics, and serves as an ethicist on pediatric and genomic advisory committees for numerous academic medical centers and professional organizations. Dr. Bush has an interdisciplinary graduate background in clinical and developmental psychology, bioethics, genomics, public health, and neuroscience that informs her research, writing, and teaching on the ethical, psychological, and policy challenges of genomic medicine and clinical research with children, and prenatal-newborn screening and sequencing.

Karen H. Rothenberg, JD, MPA serves as Senior Advisor on Genomics and Society to the Director, National Human Genome Research Institute and Visiting Scholar, Department of Bioethics, Clinical Center, National Institutes of Health. She is the Marjorie Cook Professor of Law, Founding Director, Law & Health Care Program and former Dean at the University of Maryland Francis King Carey School of Law and Visiting Professor, Johns Hopkins Berman Institute of Bioethics. Professor Rothenberg has served as Chair of the Maryland Stem Cell Research Commission, President of the American Society of Law, Medicine and Ethics, and has been on many NIH expert committees, including the NIH Recombinant DNA Advisory Committee.

It is possible to get a table of contents for the book but I notice not a single playwright is mentioned in any of the promotional material for the book. While I like the idea in principle, it seems a bit odd and suggests that these are purpose-written plays. I have not had good experiences with purpose-written plays which tend to be didactic and dull, especially when they’re not devised by a professional storyteller.

You can find out more about the upcoming ‘bioethics’ June 9 – 10, 2014 meeting here.  As for the Feb. 10 – 11, 2014 meeting, the Brain research, ethics, and nanotechnology (part one of five) May 19, 2014 post featured Barbara Herr Harthorn’s (director of the Center for Nanotechnology in Society at the University of California at Santa Barbara) participation only.

It turns out, there are some Canadian tidbits. From the Meeting Sixteen: Feb. 10-11, 2014 webcasts page, (each presenter is featured in their own webcast of approximately 11 mins.)

Timothy Caulfield, LL.M., F.R.S.C., F.C.A.H.S.

Canada Research Chair in Health Law and Policy
Professor in the Faculty of Law
and the School of Public Health
University of Alberta

Eric Racine, Ph.D.

Director, Neuroethics Research Unit
Associate Research Professor
Institut de Recherches Cliniques de Montréal
Associate Research Professor,
Department of Medicine
Université de Montréal
Adjunct Professor, Department of Medicine and Department of Neurology and Neurosurgery,
McGill University

It was a surprise to see a couple of Canucks listed as presenters and I’m grateful that the Presidential Commission for the Study of Bioethical Issues is so generous with information. in addition to the webcasts, there is the Federal Register Notice of the meeting, an agenda, transcripts, and presentation materials. By the way, Caulfield discussed hype and Racine discussed public understanding of science with regard to neuroscience both fitting into the overall theme of communication. I’ll have to look more thoroughly but it seems to me there’s no mention of pop culture as a means of communicating about science and technology.

Links to other posts in the Brains, prostheses, nanotechnology, and human enhancement five-part series:

Part one: Brain research, ethics, and nanotechnology (May 19, 2014 post)

Part three: Gray Matters: Integrative Approaches for Neuroscience, Ethics, and Society issued May 2014 by US Presidential Bioethics Commission (May 20, 2014)

Part four: Brazil, the 2014 World Cup kickoff, and a mind-controlled exoskeleton (May 20, 2014)

Part five: Brains, prostheses, nanotechnology, and human enhancement: summary (May 20, 2014)

* ‘neursocience’ corrected to ‘neuroscience’ on May 20, 2014.

Unintended consequences of reading science news online

University of Wisconsin-Madison researchers Dominique Brossard and  Dietram Scheufele have written a cautionary piece for the AAAS’s (American Association for the Advancement of Science) magazine, Science, according to a Jan. 3, 2013 news item on ScienceDaily,

A science-inclined audience and wide array of communications tools make the Internet an excellent opportunity for scientists hoping to share their research with the world. But that opportunity is fraught with unintended consequences, according to a pair of University of Wisconsin-Madison life sciences communication professors.

Dominique Brossard and Dietram Scheufele, writing in a Perspectives piece for the journal Science, encourage scientists to join an effort to make sure the public receives full, accurate and unbiased information on science and technology.

“This is an opportunity to promote interest in science — especially basic research, fundamental science — but, on the other hand, we could be missing the boat,” Brossard says. “Even our most well-intended effort could backfire, because we don’t understand the ways these same tools can work against us.”

The Jan. 3, 2012 University of Wisconsin-Madison news release by Chris Barncard (which originated the news item) notes,

Recent research by Brossard and Scheufele has described the way the Internet may be narrowing public discourse, and new work shows that a staple of online news presentation — the comments section — and other ubiquitous means to provide endorsement or feedback can color the opinions of readers of even the most neutral science stories.

Online news sources pare down discussion or limit visibility of some information in several ways, according to Brossard and Scheufele.

Many news sites use the popularity of stories or subjects (measured by the numbers of clicks they receive, or the rate at which users share that content with others, or other metrics) to guide the presentation of material.

The search engine Google offers users suggested search terms as they make requests, offering up “nanotechnology in medicine,” for example, to those who begin typing “nanotechnology” in a search box. Users often avail themselves of the list of suggestions, making certain searches more popular, which in turn makes those search terms even more likely to appear as suggestions.

Brossard and Scheufele have published an earlier study about the ‘narrowing’ effects of search engines such as Google, using the example of the topic ‘nanotechnology’, as per my May 19, 2010 posting. The researchers appear to be building on this earlier work,

The consequences become more daunting for the researchers as Brossard and Scheufele uncover more surprising effects of Web 2.0.

In their newest study, they show that independent of the content of an article about a new technological development, the tone of comments posted by other readers can make a significant difference in the way new readers feel about the article’s subject. The less civil the accompanying comments, the more risk readers attributed to the research described in the news story.

“The day of reading a story and then turning the page to read another is over,” Scheufele says. “Now each story is surrounded by numbers of Facebook likes and tweets and comments that color the way readers interpret even truly unbiased information. This will produce more and more unintended effects on readers, and unless we understand what those are and even capitalize on them, they will just cause more and more problems.”

If even some of the for-profit media world and advocacy organizations are approaching the digital landscape from a marketing perspective, Brossard and Scheufele argue, scientists need to turn to more empirical communications research and engage in active discussions across disciplines of how to most effectively reach large audiences.

“It’s not because there is not decent science writing out there. We know all kinds of excellent writers and sources,” Brossard says. “But can people be certain that those are the sites they will find when they search for information? That is not clear.”

It’s not about preparing for the future. It’s about catching up to the present. And the present, Scheufele says, includes scientific subjects — think fracking, or synthetic biology — that need debate and input from the public.

Here’s a citation and link for the Science article,

Science, New Media, and the Public by Dominique Brossard and Dietram A. Scheufele in Science 4 January 2013: Vol. 339 no. 6115 pp. 40-41 DOI: 10.1126/science.1232329

This article is behind a paywall.

Science communication at the US National Academy of Sciences

I guess it’s going to be a science communication kind of day on this blog. Dr. Andrew Maynard on his 2020 Science blog posted a May 22, 2012 piece about a recent two-day science communication event at the US National Academy of Sciences in Washington, DC.

Titled The Science of Science Communication and held May 21 – 22, 2012, I was a little concerned about the content since it suggests a dedication to metrics (which are useful but I find often misused) and the possibility of a predetermined result for science communication. After watching a webcast of the first session (Introduction and Overviews offered by Baruch Fischhof [Carnegie Mellon University] and Dietram Scheufele [University of Wisconsin at Madison], 55:35 mins.), I’m relieved to say that the first two presenters mostly avoided those pitfalls.

You can go here to watch any of the sessions held during that two days, although I will warn you that these are not TED talks. The shortest are roughly 27 mins. with most running over 1 hour, while a couple  of them run over two hours.

Getting back to Andrew and his take on the proceedings, excerpted from his May 22, 2012 posting,

It’s important that the National Academies of Science are taking the study of science communication (and its practice) seriously.  Inviting a bunch of social scientists into the National Academies – and into a high profile colloquium like this – was a big deal.  And irrespective of the meeting’s content, it flags a commitment to work closely with researchers studying science communication and decision analysis to better ensure informed and effective communication strategies and practice.  Given the substantial interest in the colloquium – on the web as well as at the meeting itself – I hope that the National Academies build on this and continue to engage fully in this area.

Moving forward, there needs to be more engagement between science communication researchers and practitioners.  Practitioners of science communication – and the practical insight they bring – were notable by their absence (in the main) from the colloquium program.  If the conversation around empirical research is to connect with effective practice, there must be better integration of these two communities.

It’s interesting to read about the colloquia (the science communication event was one of a series events known as the Arthur M. Sackler Colloquia) from the perspective of a someone who was present in real time.

Preview of new book on nanotechnology and public attitudes

For anyone who has been following the discussion about public perceptions, opinion polling, and nanotechnology, there’s a new book on the horizon: Nanotechnology: Public Perception and Risk Communication by Susanna Priest. The proposed publication date is Aug. 15, 2011. Dietram Scheufele (nanopublic blog) will be contributing a chapter of which a draft copy is currently being featured on his blog. Here’s an excerpt from Dietram’s draft,

Patterns of news coverage on nanotechnology are developing in ways that mirror issue cycles for previous technologies, including agricultural biotechnology. In particular, early coverage of nanotechnology was dominated by a general optimism about the scientific potential and economic impacts of this new technology (Dudo, Dunwoody, & Scheufele, forthcoming; Friedman & Egolf, 2005; Friedman & Egolf, 2007). This is in part related to the fact that a sizeable proportion of nanotechnology news coverage – at least in newspapers – continues to be provided by a handful of science journalists and business writers (Dudo, Choi, & Scheufele, 2011; Dudo et al., forthcoming).

Attitudes without Knowledge?

The overall positive framing of nanotechnology in news outlets is also linked to support for more research and funding among the general public (Cobb & Macoubrie, 2004; Scheufele & Lewenstein, 2005). This connection between media coverage and support for nanotechnology, however, does not follow traditional knowledge deficit models (for an overview, see Brossard, Lewenstein, & Bonney, 2005). Instead, most research on public attitudes toward nanotechnology does not show an impact of media coverage on lay audiences’ understanding of the technology, which – according to knowledge deficit models – would produce more positive attitudes toward the technology. Instead, most recent research has found that the driving factor behind public attitudes are various forms of heuristics or cognitive shortcuts that audiences use to make sense of the technology, even in the absence of information (Scheufele, 2006).

S.NET 2011 annual meeting

S.NET is the Society for the Study of  Nanoscience and Emerging Technologies and members will be holding their 3rd annual meeting in Tempe, Arizona from the 7th to the 10th of November 2011 according to Dietram Scheufele’s Jan. 11, 2011 posting on his nanopublic blog (I can’t link directly to the posting but you can find it by scrolling down). From Dietram’s posting,

Invitation. S.NET invites contributions to the Third Annual Meeting of the The Society for the Study of Nanoscience and Emerging Technologies (S.NET) to be held in Tempe (Phoenix), Arizona. The workshop will engage diverse scholars, practitioners, and policy makers in the development and implications of emerging technologies.

About S.NET. S.NET is an international association that promotes intellectual exchange and critical inquiry about the advancement of nanoscience and emerging technologies in society. The aim of the association is to advance critical reflection on developments in a broad range of new and emerging fields of science and technology, including, but not limited to, nanoscale science and engineering, biotechnology, synthetic biology, cognitive science, and geoengineering.

Eligibility. S.NET includes diverse communities, viewpoints, and methodologies from across the social sciences and humanities, and welcomes contributions from scientists, engineers, and other practitioners.

To Apply. The program committee (see below) invites submissions from the full breadth of disciplines, methodologies, and epistemologies, as well as from applied, participatory, and practical approaches to studying these emerging fields and from different regional or comparative perspectives. Committed to diverse styles of communication, S.NET welcomes proposals for individual papers, posters, traditional panels, roundtable discussions, and other more innovative formats. In particular, the program committee encourages proposals for topics and formats that will encourage greater dialogue and interaction. Details of the submission process are available online at cns.ucsb.edu/snet2011. All proposals should be submitted online between 1 Feb and 1 March 2011.

Stipends. Travel stipends may be available for US graduate students, and post-doctoral scholars, and non-US participants from the Global South.

I mentioned the 2010 S.Net annual meeting in my Sept. 14, 2010 posting and briefly in my Nov. 8, 2010 posting. In both cases, you will have to scroll down to find the information as the meeting was not the main subject.

Flaws in opinion polls about science

If you’ve ever had the experience of trying to answer an opinion poll and wanting to scream in frustration because the questions are vague or cover too much ground, this is the study for you: Measuring risk/benefit perceptions of emerging technologies and their potential impact on communication of public opinion toward science published online in the journal Public Understanding of Science, Jan. 6, 2011. From the Jan. 13, 2011 news item on physorg.com,

A new study from North Carolina State University highlights a major flaw in attempting to use a single survey question to assess public opinion on science issues. Researchers found that people who say that risks posed by new science fields outweigh benefits often actually perceive more benefits than risks when asked more detailed questions.

We set out to determine whether we can accurately assess public opinion on complex science issues with one question, or if we need to break the issue down into questions on each of the issue’s constituent parts,” says Dr. Andrew Binder, an assistant professor of communication at NC State and lead author of the study. “We found that, to varying degrees, accuracy really depends on breaking it down into multiple questions for people.” [emphasis mine]

To assess the problematic nature of a single-question surveys, the researchers developed two surveys; one focused on nanotechnology and the other on biofuels. In each survey, respondents were asked an overarching question: do the risks associated with nanotechnology/biofuels outweigh the benefits; do the benefits outweigh the risks; or are the risks and benefits approximately the same? The researchers then asked survey participants a series of questions about specific risks and benefits associated with nanotechnology or biofuels.

Precisely! Your answer to questions like these tends to be informed by the situation. In other words, you might see a benefit outweighing a risk where self-cleaning windows are concerned but not where transgenic goats (e.g. goats with spider genes) are concerned. Both of these are nanotechnology oriented, the windows being an application and the spidery goats supplying milk that can be spun for nanotechnology applications.

The article is behind a paywall but you can find out more about the study at nanopublic, Dietram Scheufele’s blog (from his Jan. 14 2011 posting,  [note: I cannot link to directly to the post so you may need to scroll down or search for it by date],

Studies tapping public perceptions of the risks and benefits surrounding new technologies have long relied on a single-item measure asking respondents a variant of the following question: “Do the risks associated with technology x outweigh the benefits; do the benefits outweigh the risks; or are the risks and benefits approximately the same?” More recently, we raised concerns about this single item measure and suggested that — especially for nanotechnology — a more application-specific look at risk perceptions might be useful.

Dietram is one of the paper’s authors.