David Torcivia:

[0:00] I'm David Torcivia.

Daniel Forkner:

I’m Daniel Forkner.

David Torcivia:

And this is Ashes Ashes, a podcast about systemic issues, cracks in civilization, collapse of the environment, and if we're unlucky, the end of the world.

Daniel Forkner:

[0:13] But if we learn from all this maybe we can stop that. The world might be broken but it doesn't have to be.

David Torcivia:

[0:18] Now this week we're going to discuss a very new and rapidly evolving technology in the fields of molecular biology and genetics.

And this is all going to have enormous effects on our health, society at large, and life as a whole. And there's a lot of science here and if you're familiar with gene therapy, CRISPR, and all this related technology, then this is not going to be anything new to you, at least at first you can skip ahead a little bit.

For the rest we’re going to do a quick basic overview of this tech and see how it works and then really dig into what it means for all of us. Daniel, you want to get us started?

Brief History Of Gene Therapy

Daniel Forkner:

[0:48] Sure let's look at the historical overview of gene therapy which is not a new field right? Gene therapy which is the addition of new genes into the DNA found in cells, is something that scientists have experimented with since the 60s.

A researcher studying a virus that infected rabbits – this Shope Papilloma virus - noticed that in addition to causing harmful warts, rabbits that got infected with this virus had a lower level of arginine in their blood - this is an amino acid. And he theorized that when the virus inserted its DNA into rabbit cells, part of that DNA included a gene that just happened to encode for an enzyme that reduced Arginine levels. [1:31] At around this time a genetic disorder was discovered in humans that caused the body to overproduce Arginine in the blood which led to spasms, epilepsy, and mental retardation. So in an attempt to cure this disorder and test his hypothesis, this researcher - Stanfield Rogers - injected patients that had this disorder with large quantities of this Shope virus. And as you might expect….

[1:57] It didn't work at all; it didn't do anything to these patient and a lot of scientist severely criticized Rogers for even attempting this experiment calling it reckless.

And he never tried anything like this again. Later in fact we even looked at the genetic material of this virus and figured out that it doesn't even have this gene that he thought it did so it was just a failed experiment.

But this concept of using a virus to deliver genetic payload you want into a patient - that idea revolutionized genetics and really got scientists thinking.

[2:31] And in the 70s and 80s researchers had figured out a crude method for copy and pasting segments of DNA into their own synthesized DNA called recombinant DNA. And combining this with viruses - so taking out the bad DNA in a virus and replacing it with this DNA created in the lab - really opened the door for a new type medicine to treat genetic disorders. Although many of these medicines, what we call viral vectors, have had several complications along the way they are still the most popular method of delivering genetic payloads.

In addition to this new technology that will talk about soon.

David Torcivia:

[3:09] Yeah that's right so I mean this technology is really amazing and the ability to actually physically edit the DNA inside of us was a huge jump in medical technology, but the problem with it at the time - and still now using the same tech that they were at the time - is that it's not very accurate. So it's sort of like trying to make a very detailed and fine sculpture only using a shotgun.

Yeah you can do it if you're really careful, and spend a long time on it but odds are you’re going to mess up somewhere along the way, and this was for a long time really the process of this gene therapy technology.

Daniel Forkner:

[3:43] Yeah David it's this shotgun approach that is really the biggest drawback to using viral vectors with recombinant DNA.

Viruses are extremely good at delivering their DNA to a cell - that's what they evolved to do - but once that DNA is in the cell it kind of just randomly and haphazardly fits its way into the cell's DNA. So it's fine if you're just trying to introduce a new gene - which is useful in correcting some genetic disorders - but if you want to target and correct a defective gene that's already in the cell, or remove that gene, or any other kind of specific manipulation, you can't do it using this method.

David Torcivia:

[4:22] That technology developed, and as more research was done in that area scientists got better at being more specific about adding smaller and smaller bits of DNA and doing more stuff with that DNA. So a big breakthrough here was figuring out how to insert this DNA into fertilized eggs, and from there as this embryo developed this modified DNA would be in the entire organism once it mature.

And that was a huge breakthrough saying like “oh it's not just about adjusting some certain sections of the cells in the body. We can take this DNA, and insert it into an organism, and our introduced DNA becomes that organism’s actual DNA.”

For a long time this was sort of the evolution of this technology: we would have a viral vector that introduces large amounts of DNA into the subject, and as time goes on we were getting better and better at doing smaller and smaller bits of DNA, may be putting them in the right place, figuring out how to break DNA and insert things into certain areas… but it was all still a very rough process. But then all that changed a few years ago.

What Is CRISPR?

Daniel Forkner:

[5:20] That's right and it's that targeting a specific location that had eluded scientists for a long time. It’s like “okay we can introduce this DNA but how do we make it go to a certain location?” The Breakthrough came with CRISPR and so maybe you've heard of this technology called CRISPR so what is it exactly?

There're a thousand videos you can go online and you can see exactly how it works in animated form and can be very useful, but let's just break it down really simply. CRISPR explains a part of bacterial genomes the function to fight off and remember encounters with viruses.

It's essentially a genetic immune system, and within this section of the DNA of bacteria, there are regularly spaced “palindromic repeats.” There are short segments of DNA that act as markers for these different Cas proteins that always accompany the CRISPR genes.

And in between these short segments are unique 20 letter strings of DNA called spacers which are exact copies of viral DNA.

The CRISPR section acts as a sort of library for all the viral DNA this bacteria has encountered in the past, so when a virus attacks a bacterial cell, it injects its DNA and this CRISPR system responds. If it's a first encounter, proteins in the cell will splice off a section of the viral DNA and store it and its own genome as another spacer, and if this cell and counters the same virus again all of these spacers get transcribed into RNA molecules which then bind with a Cas protein, and if any of these RNA molecules match with the viral DNA, these Cas proteins go to work destroying the invading DNA.

David Torcivia:

[6:56] That’s a lot of words, a lot of acronyms – DNA, RNA, CRISPR, Cass - but very simply think of it like this:

So what CRISPR does is it records a big library all these infections that happen to a cell. When a cell gets infected it says “oh we got infected by this thing, here’s a recording of what this thing’s DNA is; if this DNA pops up again destroy it.”

And so every time new DNA is inserted into the cell it checks it against this database, this library, and when it finds a match it says “this is a bad thing send out these proteins, these Cass proteins to go out and rip up this DNA.”

The proteins go out and it saves the cell so it can live another day.

[7:37] You’ve got this like very simple library system that also enables the cell to defend itself from these Invaders.

Daniel Forkner:

[7:45] Yeah and when researchers figured that out they got really excited because you know this promised to answer that question that had been eluding them is: how do you target a very specific location in a genome?

Because if you can do that you can do all kinds of things; you can introduce new genes into specific areas; you can correct genetic defects; you can remove bad genes; you can do new kinds of targeted research into understanding cancer cells and all types of diseases and we'll get into these benefits and applications in a little bit.

David Torcivia:

[8:13] The real breakthrough in this field occurred in 2012 when researchers at the University of Berkeley discovered that one of these CRISPR proteins - one of those little things that runs out and checks DNA and comes back and it cuts DNA up and stuff – they discovered one of these called Cas9, and these Cas proteins, there's a lot of them they all do different things, but Cas9 was really special. And this discovery, it hasn't won a Nobel Prize yet but it's guaranteed that it’s going to within the next couple years, because what it enables, what Cas9 enables researchers, these geneticists, these medical doctors to do is very accurately, very precisely, target specific sections of DNA and do basically whatever you want with it.

Whether that’s cut it out, insert things in, move stuff around; it makes it fast and makes it accurate and it's going to totally transform our world.

Daniel Forkner:

[9:05] In many ways it already has, because they were able to quickly commercialize this Cas system to be used in any lab worldwide for very cheap and to apply to almost any cell you wanted to.

David Torcivia:

[9:16] We can't understand how important this discovery is going to be. I mean this is the same thing as discovering nuclear power, this is the same thing as discovering germs, this is the same as discovering the double helix in DNA; this is a world-shattering groundbreaking discovery that’s going to totally transform all of our lives over the next few years and decades.

So let's take a look at some of those possibility.

Daniel Forkner:

[9:39] There's a lot of benefits that come with CRISPR, for instance you can correct genetic defects in animals and humans including making changes to stem cells, and one example of that is you can correct the genetic defect that causes sickle-cell anemia.

Benefits Of CRISPR

By taking these stem cells out of an individual’s bone marrow, making these CRISPR changes to it, correcting that genetic defect, reintroducing the stem cells into their bone marrow and all of a sudden now their blood will be free of this genetic defect.

David Torcivia:

[10:06] Yeah I mean at this point really any sort of genetic disease - things that are caused by a single gene or a few genes - could be something that could be fixed by this technology. There's lots of blood diseases, skin diseases, different types of disabilities that just a quick fix with CRISPR could totally eliminate this defect from somebody's DNA and cure them of that disease; something that would have been previously and a completely incurable illness.

It's huge.

Daniel Forkner:

[10:32] CRISPR opens a wide range of opportunities for research and a big one is cancer. One of the reasons why it's so difficult to understand cancer at the genetic level is because cancerous cells mutate so quickly, but why CRISPR allows you to do is take a group of cancer cells and kind of isolate different genes within different groups of cancerous cells, and see which genes are absolutely critical to the development of these cancerous cells, and once we know that it's much easier to target these genes away.

David Torcivia:

[11:01] In the same vein with cancer, imagine being able to take a pill that has targeted attack vectors that are only going to seek out the cancerous cells in your body because they’ve biopsied these cells and they realize what part of the DNA flipped and went wrong, and so these killer cells are going into you, detecting only that cancerous DNA, killing all those cells and then, you know you take this pill and a week later you wake up, you feel a little sick but you have no more cancer.

That's the type of stuff CRISPR might enable: these super hyper-personalized DNA-level genomic medicine which is something that we've never seen before, and is going to totally transform how we do medicine, how we think about medicine and it's going to be worth trillions and trillions of dollars.

Daniel Forkner:

[11:45] To take that even one step further you could even introduce genetic changes into someone's genome so that the body is able to identify these cancerous cells and fight them off itself. There have already been many real-world applications in experiments with this CRISPR; researchers have taken goats for example and used CRISPR to engineer them to produce more wool and to be more meaty so that they're more productive economically.

We've used it in agriculture to create disease-resistant rice, or you can take a potato and genetically alter it so that it doesn't produce that neurotoxin that's bad for you in large quantities.

David Torcivia:

[12:19] Yeah or in the more dramatic realm researchers are even talking about we could engineer mosquitoes that aren't able to carry malaria, or stepping up even one more than that, being able to directly target and wipeout the malaria parasite using just this specific gene technology, and we'll get more to that later.

Daniel Forkner:

[12:37] There's so many possibilities every single genetic disease is potentially treatable, and when you combine this at the germline level, so you at the embryo level, you're talking about changes that will affect every cell in an individual's body and eventually be passed down to its offspring, and it's become a big question: what are the ethical limits of this technology?

Some scientists will say “look we need to slow down and really think about what we're doing because if we make some mistakes at a genetic level they might not be reversible.”

David Torcivia:

[13:09] You know what I'm really looking forward to its technology?

Daniel Forkner:

What?

David Torcivia:

Permanent kittens and puppies, teeny tiny dogs and cats, that stay that way forever. You laugh now but this is totally possible with this technology. I think there's billions of dollars being wasted that we should be researching these things right now.

Daniel Forkner:

[13:26] They actually did that in China with pigs. They engineered these, what they're called micro pigs, they're very expensive, but you know these pigs that normally grow to be like 300 pounds they made them into little Chihuahua sized pets for you, you know that just run around your house and don't cause any problems.

David Torcivia:

[13:42] See that's what I'm saying we need that but with like permanent kittens. I want a cat that acts like an old cat but looks like a kitten. Scientists if you're listening to this get on this. Smaller animals are good for the environment too. Less footprint right?

Daniel Forkner:

[13:55] Haha, yea..

Part of the ethical consideration is this technology gives us so many potential opportunities for medicine.

Like right now it's very expensive to produce some medicines where you have to synthesize a lot of proteins; you have to do this in the lab and you need incubators and all these processes that can be complicated. But with CRISPR say we know “we need this protein to go to this medicine,” we can just engineer a cow to produce that for us and it will get secreted out with the cow's milk and we just separate it out and now we have two products from a cow's milk instead of one, and it's very cheap.

David Torcivia:

[14:31] Everyone remembers the Ebola outbreak a few years ago, and there was a dramatic thing, they couldn't get enough - well first there were no vaccines and they rushed through a lot of stuff. They started developing ebola vaccines for the first time but they couldn't get enough of them made because it was very hard to synthesize and create.

And then some lab came up on the idea of “well let’s synthesize this Ebola vaccine into a plant using CRISPR” I think it might have been a tobacco plant, you might have to double-check me on that.

But they grew Ebola vaccines in a greenhouse using this genetically modified CRISPR plant. So we're growing vaccines with plants, using this technology, saving lives right now.

It's incredible and this is just a couple of years after this technology was first discovered. That's how rapidly this field is advancing.

Daniel Forkner:

[15:17] This technology is developing so quickly, I mean scientist have stopped referring it to Gene editing and now call this whole field “genome engineering” because of the possibilities and because of how fast is thing has evolved.

David Torcivia:

[15:32] There are over 7,000 human genetic diseases that are caused by just a single known gene mutation. All those diseases using this CRISPR technology could be gone in the next couple of years and certainly in the next few decades.

[15:46] And if that isn't crazy enough I mean scientists are literally talking about bringing extinct species back to life, like straight out of Jurassic Park with this CRISPR technology.

Researchers are looking at bringing the woolly mammoth back by using Asian elephants and inserting this this woolly mammoth DNA, which we have from all the preserved woolly mammoth carcasses that they found frozen in the permafrost as it quickly melts from all this global warming…

But they take this, they extract the DNA and we have a mostly complete woolly mammoth genome at this point and they just insert that into Asian elephant embryo and next thing you know we've got woolly mammoths again.

Daniel Forkner:

[16:25] Bringing back the woolly mammoth is actually one of the strategies that some people are saying is a way to curb climate change, you know we’ll just release all these woolly mammoths back into their natural habitat in the Arctic tundra, they’ll eat all the grass and clear all the shrubs, exposing that permafrost to more colder air and preventing it from thawing out and releasing all that methane that we talked about in earlier episode.

Seems a bit far-fetched to me but I'd love to see it

David Torcivia:

[16:50] Yeah I mean that’s a technofix if I've ever heard but I definitely want to see woolly mammoths so I’m going to let this one slide.

So as you can see I mean all this technology has the potential to do amazing wonderful almost science fiction like things.

What’s incredible about this is that this all happened really in just the past few years.

So if you had gone 7, 8 years ago people would have said that this stuff is impossible, but six years ago after the discovery of this CRISPR-Cas9 technology, and there are new technologies using CRISPR like CPF1 which might prove to be even easier and cheaper, and they're researching more constantly all the time so by the time you hear this this might already be out of date - that's how quick of this field is changing.

But it's really incredible what's happened in just a few years. You've maybe head of Moore's law in the Computing world where things get faster and faster all the time at a certain rate, and this makes that look like nothing; this is exponential growth at its largest and purist sense.

Daniel Forkner:

[17:45] It’s the cost of genetic sequencing that's one component of this rapid transformation. This Precision medicine, this CRISPR technology, is made possible through two realities. One is the rapidly declining cost in sequencing genomes, so understanding the DNA makeup of any individual or cell, but also this technology of targeting very specific places in the genome and delivering target DNA to that location.

And both of these components are really making just astonishing progress, and it's hard to keep up with.

David Torcivia:

[18:17] Yeah I mean so the Human Genome Project right, that sort of Manhattan Project a few decades ago to map the entire Human Genome which really sort of kicked off all this stuff.

What did that cost? It was like billions of dollars at the time, and now I can go and spend a tube for less than $100 and it's not a full genomic sequence but I can have most of my DNA sequence spit out and then they tell me I'm how ever much Neanderthal and that I'm actually from whatever country. That's amazing, don't do that by the way, we'll talk about why you shouldn't later, but the technology is incredible and the price of it is incredible.

Daniel Forkner:

[18:51] This technology has been progressing so fast and becoming so accessible and so easy to manipulate that the Director of National Intelligence of the United States in 2016 came out with a report that listed genetic engineering as a weapon of mass destruction and a potential threat to our security.

Maybe we should talk a little bit about why this is concerning to us and why we might be worried about it from a security standpoint and as individuals.

Biohacking

David Torcivia:

[19:18] This is why I really wanted to do this show because for me this is why this technology is so interesting and quite frankly so terrifying.

We've already established that this is an amazing tech, in terms of the potential it has in changing our lives for the better, but at the same time this is just as easy to really completely destroy the world in a way that we've never seen before, and I think surpasses even the power of nuclear weapons. First of all, this technology is cheap.

Already you know we're barely five years passed the discovery of this; I can go online, I can order a CRISPR kit to do my own CRISPR testing for $1,500, and then I can start developing CRISPR genes in my garage. That’s nothing right? If you wanted to build a nuclear bomb, like that is prohibitively expensive and difficult to source all those things. I'm not saying people haven't done that, maybe you've heard the story of the nuclear Boy Scout, who using free samples and a lot of earnest letters was able to build an almost functioning nuclear reactor in his garage before the Nuclear regulatory committee and FBI showed up and busted him. The knowledge is out there, it's easy to do but finding all those materials is difficult.

With CRISPR that's not the case. The knowledge is out there and the materials to do this are simple and accessible.

And that’s something we've never seen before as a threat.

Daniel Forkner:

[20:34] I like how you liken it to nuclear power because it's an example of how technology can have dual use right, it can be used for good and it can obviously be used for very destructive means.

And this is just one of those technologies, just like all the others, that has very good potential uses but also very malicious uses that are possible, and the fact that it's so easy for someone to get their hands on this technology is one of the concerning aspects of it.

Like you mentioned it's very cheap to start experimenting with it, and as the research goes forward it will become easier and easier to utilize this technology.

And as we know like that the stuff that happens in Labs can have you know a real worldwide impact right, and we have a pretty tough time already dealing with very natural pandemics and very natural biological agents; imagine now that not only do we have to deal with natural pathogens, but now people are in their basement engineering pandemics and engineering pathogens with this CRISPR and related technologies.

David Torcivia:

[21:35] You mentioned escaping the lab, so yeah that's always every scientists like worst fear, that working on one of these highly infectious pathogens, it gets out of lab security and causes a pandemic all around the world.

There's lots of safeguards and stuff that they take, there’s different levels of labs and different safety precautions, and only a couple labs have some highly infectious things like smallpox - only in one or two labs - but there is precedence of these pathogens escaping these laboratory conditions.

Maybe you’ve heard of the 1918 influenza - the Spanish Flu.

The Spanish influenza was one of the worst outbreaks in modern history.

Daniel Forkner:

[22:10] Killed 1% of the human population.

David Torcivia:

[22:13] Yeah 1%, which doesn't sound like a lot but I mean that's millions and millions of people.

Yeah there were other diseases in the past that killed more; Bubonic plague in Europe killed 25% of all Europeans, but in terms of raw numbers nothing has killed as much as this 1918 flu virus.

And it was a huge problem, lots of people died all around the world, there are only one or two places in the entire Earth that this virus didn't reach and millions and millions of people die. Eventually they were able to vaccinate against the flu and it went away and that particular strain of flu, H1, disappeared from the earth it was only found in laboratories. Following that there were more flu varieanrt that appread. H2, and then another one H3. H2 has disappeared we don't see that anymore, and H3 is one of the strains we have today.

But in 1977 all of a sudden people started getting sick with H1 again, and it's not unusual for these flue viruses to come back and reappear. What is unusual is this H1 virus that reappeared was identical genetically to the 1918 strain H1N1 that killed all those people 60 years before this.

And the only way that something like that could happen was if somebody had preserved this virus, which they had in all sorts of labs around the world, and then that virus was able to get out of the lab and back into the population. And what was really interesting about this outbreak was that it wasn't killing the sick and elderly like the flu virus normally does, it was killing the young, and that's because the elderly had been exposed to this virus when they were young and had survived and had those antibodies built up in them.

[23:45] But the young, the healthy, college kids, people in their 20s and 30s, they have never been exposed to this variant of the virus and it wreaked havoc on them. Lots of people got sick, many people died - not as nearly as much as in 1918 because we had a lot more preparation and our medicine was better at the time – but it was a serious outbreak, and today we're still dealing with this. We have both the H1 and H3 variants around us and we have to be immunized for both come flu season.

That's the kind of potential danger that we’re facing with these lab outbreaks.

Daniel Forkner:

[24:14] And some of these can have extremely long lasting effects like you mentioned the H1 virus that went away is still with us today.

The UK experimented with Anthrax on an island which came to be known as Anthrax Island during World War II and it was uninhabitable for 50 years after that.

David Torcivia:

[24:31] So then the question becomes if these are just like regular naturally occurring diseases like the flu, and they can cause all that damage when they escape from labs, what happens when a potentially maliciously engineered virus or bacteria or whatever it is escapes?

Daniel Forkner:

[24:49] Or even not escapes but is deliberately deployed upon different people.

David Torcivia:

[24:54] I mean that's an interesting point right, so historically biological warfare hasn't been particularly useful because yes it's devastating to your enemy but the chances that it crosses from your enemy and starts impacting yourself is very dramatically High.

And so then the trade-off becomes “is just worth it? I’m probably going to hurt myself it's probably not” and so it really relegated these biological weapons to “we’re screwed and we're going to take everyone out with us.”

But with CRISPR, and with genetically targeted pathogens that might not be the case anymore.

Daniel Forkner:

[25:27] CRISPR allows you to engineer something that is so precise you can literally write almost like an if-then statement into this pathogen:

IF female and IF Asian, THEN deploy this mechanism that causes infertility.

If someone was really anti-Asian and wanted to wipe the Asian populations out, you know maybe you could design something they could actually do that. It’s was very precise what you can do with this.

David Torcivia:

[25:53] Let me let me jump in there, before you start saying “but but but! There's no genes that encode for Asian” or whatever. Yeah you're right but there are lots of genetic quirks that are present in certain different types of race, or ethnicity, or families, or all sorts of different specific things you can break down.

So for example Asians carry the gene called EDAR and almost 87% of Asians have this, so it’s a very easy way to target of racial group. These genetic quirks that have emerged in different geographical areas, can be very specifically engineered using this CRISPR technology to identify people and then deploy whatever it is you want to deploy to them. Not just that but it’s sex, it’s ethnic groups, so like Ashkenazi Jews you could target them, they have a lot of very specific things that are unique to that ethnic group.

Daniel Forkner:

[26:42] And obviously if you share your individual genome like through one of these services, ancestry or something like that, that data might not be protected and anyone who gets their hands on that could design something specifically for you.

David Torcivia:

[26:55] Right and not just you but also your family. So maybe you didn't do that but you have a sister or father or somebody that did, and now they can target your specific family members and your whole line and deploy something that targets you and your family.

Daniel Forkner:

[27:08] Okay but David, I mean why would someone want to do this? I mean, sure I guess you could go through all the money and expense and really the difficulty and inconvenience of trying to design some kind of genetic pathogen; some CRISPR related vector that goes into your body and affects you.

But why would someone go to that trouble?

David Torcivia:

[27:27] Well this is my favorite part of this game, it's the “what if game.” What if we wanted to be the bad guys for a minute?

And they're all sorts of things, and remember lots of these people wouldn't call themselves bad guys, but they realize that this technology enables them to get what they want in a way that's never been possible before.

A lot of this information we pulled from a DEF CON talk from the head of medical technology at Intel and he gave us a couple of great examples that gets us started.

So say I'm a militant vegan, or a militant vegetarian.

Daniel Forkner:

[27:57] I've ever met anyone like that.

David Torcivia:

[28:00] And maybe you don’t want anyone to eat meat.

Well there are specific genes that you can target that if you flip this or you break this switch, then all of a sudden you become intolerant to meat; you can't eat it anymore and you are forced to have a vegetarian lifestyle.

It's theoretically possible and in fact getting easier all the time that somebody can engineer a virus or disease or something, that when it infects you it flips this thing in your genes and you can no longer eat meat, and so the whole world is suddenly vegetarian within a couple generations, especially if you’re using gene drive technology which is something we'll discuss in a little bit.

Or say if I have some Vendetta against sex and I want the world to be sexually chased, I can engineer something that, I don't know makes gonorrhea hyper-infective so if you sleep with somebody outside of whatever monogamous relationship you have, then the odds of you getting this hyper gonorrhea is suddenly very high.

Or if I don't want anybody having sex at all like I can engineer erectile dysfunction for everybody on Earth; maybe if I sell Viagra I would want to do that anyway.

I can make people intolerant to alcohol; I can make people intolerant to the Sun so they have to veil themselves all the time.

Just about any genetic disease that can be flipped with one or two or just a few genes, you can give to people, and a lot of those you can do for ideological reasons.

Daniel Forkner:

[29:19] A lot of those that you mentioned David sound like they stem from ideological incentives and the types of things we might expect different terrorist groups or individuals to carry out, but what's most alarming to me about all this is that there's actually a lot of incentives for institutional abuse of this type of thing.

For example I could be a pharmaceutical company, and “hey instead of producing a better medicine why don't I just you encourage the disease that I can cure and that will cause more demand for this medicine that I can provide” so kind of create the disease and then supply the cure.

Or even at a national level say two countries are really competing with each other economically. One nation could potentially impact the citizens of the other with something like narcolepsy if it wanted to interrupt its worker's productivity. Or maybe it would want its competitor’s National Healthcare cost to rise so it introduces early onset Alzheimer's disease, or cancer, or Parkinson's, or something like that that will really cripple its citizens.

David Torcivia:

[30:20] Right the potential for abuse from both individuals, from terrorist groups, from nation state actors, and from corporations is humongous. And the thing is a lot of this stuff is hard to tie back to an individual.

So say if I'm the bad actor at McDonald's and I want to tweak people's biology so that they process saturated fats better, so they’re more likely to eat my food. I could do that and then I can release that and nobody would know that it was me, and suddenly this change in the population happened. I benefit from it, there's nothing that comes back and bites me for it because it's hard to trace back to the source, and they're very hard to detect and by the time we catch it it's often too late to do anything about it.

It's a very terrifying time here where this offensive technology is hugely powerful, and it's evolving, and getting cheaper, and more accessible literally every single day but the defensive technology - to stop this, to protect these things, really doesn't exist at all yet.

Daniel Forkner:

[31:16] There's also a host of embarrassing and inconvenient things that you could do to someone's genome. You could cause total baldness for someone who may be recognized for their hair right, maybe this could be a celebrity or someone in politics.

You could give someone a fishy body odor which is really intolerable; no one wants to be around this person anymore and it would be quite embarrassing for them in a public sphere.

David Torcivia:

[31:40] You could make them fat, you give them diarrhea, give them Tourette’s where they're constantly cursing and you're not going to be a politician or CEO or public speaker if you have these problems.

The potential for abuse for this is huge.

And it's not limited just to people either; economic targets also are big thing. One great example that I heard was: I'm a California wine grower, and I want to target the wines of France because they’re cutting into my profits. I can genetically engineer, using my Silicon Valley buds, this CRISPR pathogen, we release it on the wine fields of France, and wipe out their vineyards, so the world’s wine markets have to turn to California for an alternative. No one would stop me, no one could do anything about that, and I would get away with very little risk.

Daniel Forkner:

[32:23] I mentioned how you could design sort of like an IF-THEN statement, you know IF this person THEN deploy this genetic alteration. You can also manipulate the time though, you could introduce a change that only takes place after a certain time delay.

David Torcivia:

[32:38] And not just time but also environment.

So this is when this technology starts getting really crazy; you can integrate the epigenetic - that is the affect the environment has on the expression of these genes - to regulate when these CRISPR sections of the DNA are activated.

So like environmental variables, at certain temperatures outside, or certain humidity levels, when people are menstrating you could activate it, how much sleep you have. If somebody isn't getting enough sleep then all of a sudden this DNA kicks in and causes some worse problem.

Even silly things like it only kicks on when you drink alcohol, or when you're drinking water, or eating certain foods, you get not only this hyper-targeted on a DNA level selection, but you can use this influence our environment has on the expression of our genes to really specify not just who you're targeting, but when and how as well.

Combining Tracking With Genetic Data

Daniel Forkner:

[33:28] And if you've listened to our Permanent Record episode and you're familiar with how corporations right now track us in so many ways and compile so many variables about our life: when we do this, when we do that, what kind of products we buy, also the medicines we take…

They're already acquiring that type of data on us, it's not a far leap to start using that data to influence us, you know, if they have this genetic tool at their disposal.

David Torcivia:

[33:52] Okay I'm just spit balling here, but the potential for this, and the abuse of this is just literally insane. So imagine a future where these CRISPR technologies can be very quickly and easily engineering, because that future is coming and it's going to be mostly automatically synthesized on computers in these machines that make it and then ‘poof’ you’ve got a little thing and you deploy to whoever you’re going to using a variety of techniques.

And so let's imagine some horrible dystopian future, which I think we have a more than decent chance of someday seeing if it doesn't all fall apart first. But you’re on vacation; you're going to go out to the beach and somebody makes you more susceptible to sun damage so they can sell you more sunscreen. Or oh you’re an active person but they want you to stay in and watch Netflix and eat junk food so they hit you with a gene that makes you tired and physical exertion wrecks you so you have to stay in and sleep, and now they're selling all these online services to you and junk food instead.

Daniel Forkner:

[34:48] Or every time you take a sip of alcohol you suddenly have erectile dysfunction, and then an ad pops up on your phone that says “oh do you have ED? You can One Touch button to get our delivery of this pill that will cure your erectile dysfunction.”

David Torcivia:

[35:01] Yeah that's a good one especially because how much alcohol and places where you don't need erectile dysfunction go together.

So basically any dystopic eventuality that you can come up with where somebody knows literally everything about you. Both how you live your life - from all the information that is collected that we discuss in Permanent Record - to who you are on an individual, biological, genetic level. Combining these two breeds a perfect dystopia of being able to influence you - literally change who you are physically on a genetic level - in order to influence your behavior in a way that suits – whether these are companies; whether these are individuals who are doing this technology maybe to hurt you; nation states; enemies; terrorist groups.

Literally anybody who has access to this technology which again is getting cheaper, and easier, every single day.

Daniel Forkner:

[35:55] Okay so there are a lot of possibilities for malicious intent right.

David Torcivia:

[35:59] You're right, you're right, okay so this is a lot of bad stuff but at the same time I guess we could have people biohacking and trying to release these things for good right?

Gene Drives

Daniel Forkner:

[36:08] One of the uses of CRISPR that's being researched right now that has a lot of potential, and there's a lot of hype around it, and it has a lot of potential for good; these are gene drives.

Under normal gene editing you insert a new strand of DNA or whatever into a cell, and it's a one time thing. I mean yes you have the chance to pass it down potentially to your offspring depending on where this DNA goes in your body, but a gene drive is something a little bit different.

In the context of CRISPR it’s when you insert a strand of DNA with a section that encodes for CRISPR itself to put this gene into every part of the genome of this individual, and the result is that it really propagates itself throughout a population - a species population - much much faster than if you just had a natural process of heredity and passing this down to your offspring.

[37:02] And one of the biggest opportunities for this technology would be to say eradicate the malaria-carrying strand of mosquito, because if you can introduce this gene into one mosquito, within just a few generations it now exists in almost every single individual in this population.

So it propagate to self very quickly.

David Torcivia:

[37:23] Yea so the potential here is just how quickly these genes can propagate, because you’re guaranteeing that it’s going to be passed on from generation to generation, which is very rare for everybody for remembers there Punnett Squares in middle school biology.

And that means the potential for this to spread - so if one of these genetically modified using gene drive technology organisms, mosquitoes, fruit flies, whatever, escaped from one of these labs just like the flu virus did back in 1977… if what one of these escaped, accidentally or on purpose maybe, the potential for that to wreak havoc on ecosystems - the environment and these species in the actual world - is profound, and would be very rapid, especially in these short life span creatures like mosquitoes, like fruit flies, and very quickly you would have these genetically modified organisms being the predominant, if not only version of this organism in the world.

If we make a mistake, that could have profound effects, and if these gene drives mutate - because there's a possibility that the gene that has this gene drive technology on it breaks or mutates - then all of a sudden this mutation is the thing being passed on by this reproductive technology.

That could have very serious ramifications if the mutation is negative; if it's something that we don't want to see happen. And because of this there are a lot of scientists, a lot of researchers, who are coming out and saying “we need to be very, very careful with this technology; we need to not be releasing this into the world until we better understand the possible effects this might have on the environment; on the ecological systems that we want to be influenced using gene drives.”

Daniel Forkner:

[39:03] If you're talking about wiping out an entire species like a malaria-carrying mosquito.

David Torcivia:

[39:08] Or Malaria itself.

Daniel Forkner:

[39:09] Or Malaria itself. I don't think it's really possible to understand the ecological system enough to really calculate how that subtraction of species is going to impact the overall system right?

And you mentioned that scientist are really concerned about how to go forward with this, and that's important, but the way they talk about going about safeguarding these gene drives is:

One, when you do research with these species to make sure you can contain them at all costs. For the very first experiment that was done using a gene drive on a fruit fly, it has been estimated that if one individual fruit fly had escaped that experiment it would have affected you know between 20 and 50% of all fruit flies worldwide. So scientists are coming out saying “look if you do these experiments you need to make sure that these species are contained.”

Two, there are possibilities to reverse some of these gene drives; they’re called reverse drives, and the ideas is if you were to introduce a gene drive and then you say “oh you know what, can we redo that?” then you can introduce another gene drive into the population which basically cuts that original gene drive out. The problem with that though is that you have to design that in the context of the initial experiment. You have to have it in mind that we're going to create this, you know vaccine for lack of better words, to solve a problem if it arises. Which of course is expensive and some labs may opt not to do that.

But the other problem means that one of our options for curing a potential problem of gene drive is this very reactive response like “oh no we need to fix this so let’s introduce another gene drive to fix it” and if something went wrong with the first one there's no guarantee that the reverse drive is going to be any more successful.

David Torcivia:

[40:51] So then you just keep releasing more gene drives hoping we'll fix it eventually.

You know this reminds me of?

Daniel Forkner:

[40:57] What does it remind you of?

David Torcivia:

[40:58] That song “There Was an Old Lady Who Swallowed a Fly,” and hopefully we don't end up with a situation like that; continuously swallowing larger and larger things to fix our fuck ups until of course we die.

Daniel Forkner:

[41:10] Well obviously screwing up in the context of a legitimate experiment, or a legitimate clinical trial of one of these gene dries is a concern.

But if you're talking about like designing better experiments to contain these gene drives, that does nothing to address someone who might have a malicious intent and doesn't really care about containing their experiment because their intent is to introduce a harmful gene drive into a population.

You know to bring it back to the malicious intent that we talked about in the biohacking, it would be perfectly possible to introduce a gene drive say into mosquitoes that causes mosquitoes to produce a toxin that's only harmful to humans, and it would be very hard to detect that until it was too late and then very difficult to fix it before it has wrecked damage upon the human population.

David Torcivia:

[42:01] I briefly brought up the concept of defense for some of these gene technologies and gene drives.

But we didn't really explore what that might be, and what they might entail, because for one the research isn't done there and scientists and biohackers and whatever aren't entirely sure what they will be, but I mean it comes down to a multi-component thing.

There’s the regulatory framework right; we need to ban research that could be dangerous but that doesn't stop people acting outside the law, and it doesn't stop people in countries that don't respect the law, and there's a lot of black and gray areas beyond these regulations, so counting on them is a fool's errand.

What does that mean beyond that? Well one way would be to constantly test people's genetic code and make sure we're not getting infected by these gene attacks. And not just people but also plants, animals, whatever it is that's important to you, important to the ecosystems that you care about. Fortunately that's becoming cheaper and more possible as these gene sequencing technologies reduce in price. But the scope and scale of being able to constantly monitor all this is crazy.

I mean there might be a day in the not-too-distant future where you know you wake up, you weigh yourself on your scale, you swab yourself to make sure your DNA is still intact and what it's supposed to be, and then you know you take your shower, brush your teeth, and go on with your day.

In the same way, being able to have backups of your DNA of this pure stem cell level of you to fix things when they might come up, when you might have unwanted genetic alteration, might be something that is just done in standard. So you're born, they already will take your umbilical cord, take that cord blood, freeze it somewhere in case you ever need the stem cells that are in there, but also have like physical digital backups of your DNA in case you need to re-splice stuff back in and rework all those details.

This might be a weird just accepted part of our life in the future. I certainly hope that's not the case but right now that seems to be as far as we can figure out the best line of defense for these types of technology, in addition to quick government response when these large scale infections are discovered, and sort of quickly scaling up vaccines and deploying them to thousands, hundreds of thousands, millions, and hopefully not but possibly even billions of people within the course of weeks.

Okay wait wait, I want to jump topics totally and dramatically, sorry for this not so smooth transition but

Designer Babies

[44:18] When I hear is gene therapy, when I hear gene improvements, what's the first thing that comes in mind? Of course it's designer babies. It's everyone's favorite ethical question: if you could engineer certain things in your children would you do it? Would you make them smarter, tall, or strong or whatever it is? Well now the technology’s catching up to that and some of those things might be possible. Some are possible right now and some might be in the coming decades. What does that mean, what can we do at the moment, and what implications will that have for all of us?

You want to start us on this Daniel?

Daniel Forkner:

[44:50] Yeah and to start I think we should just clarify right, there's a lot of talk that goes on with designer babies and it always seems to reduce to this:

“oh you know 15 years from now my next door neighbor has a kid and they choose to have a designer baby so their kid comes out super intelligent, super athletic, super beautiful and tall, but because I didn't elect to do that, my child's just kind of normal and therefore not as good as their child.

David Torcivia:

[45:15] The GATTACA scenario if you will.

Daniel Forkner:

[45:17] Yeah and it we should just clarify that when it comes to like general intelligence, and general athletic ability, and stuff like that, we're nowhere near a place where we can even understand what general intelligence is just psychologically right? And when you're talking about something that has no doubtless a genetic component, but also environmental and some of these epigenetic factors that you talked about, maybe the way your raised plays a role in this… The idea that we can somehow tease out this complex relationship of genes and codes for some mythical general intelligences is very unlikely if not impossible, at least not in the time frame that we should be worried about any of this.

But of course when it comes to designer babies, or making genetic changes to embryos, obviously the priority first goes to curing or preventing any genetic defects that might encode for disorders or diseases.

[46:09] And then yeah I guess there are some genes out there that we can identify and point to and say “hey if we change this gene maybe you'll have stronger bones, or you know the hemoglobin in your blood will be more efficient so when it comes to athletic activities you have more endurance.”

There's even a gene that we feel fairly confident that if we can alter that, you as an individual might need less sleep. So there's very specific things that we as a society could think of as improvements or enhancements, but they're very specific and they're not so general as just like an intelligent super baby.

David Torcivia:

So instead of looking at generic, across the board improvements like intelligence, whatever that means, where there's a million genes playing into it as well as all these other factors, it's just a matter of “oh we found this gene when it flips gives you extra muscles. And that’s the future you’re talking about Daniel where people are super-strong or super-endurant or don't need much sleep and that technology more less exists right now yeah?

Daniel Forkner:

[47:12] It's possible yeah.

But we should take a step back for a second because where the science is right now when it comes to embryonic gene editing, there are a lot of concerns with unintended side effects, and changes in the biodiversity of us as a species globally. And these are concerns that might not go away regardless of how well the science progresses, and how good the technology gets.

And this is going on right now; researchers are using embryos as we speak, testing this CRISPR technology to correct genetic defects. The first experiment took place in China in 2015, and the United States just did its first embryonic experiment in 2017.

So we still have a lot of questions right about what the ethical lines are in what will become clinical trials of embryonic gene editing, but the research is going full speed ahead, and so we have a limited time frame to kind of address and answer some of these questions and concerns that we have.

Unintended Consequences

David Torcivia:

[48:09] The technology to make these edits and to make these fixes and improvements on embryos exists right now; we can do this at this moment.

But the problem is we don't have a complete understanding of our genome and the way that all these individual genes interact with each other.

Though we've sequenced everything, and we can point to all the different ATCG bits of the DNA, understanding how one set of genes can affect every single other part, is still something that we're working on developing all the time. And we realize that things that sound like they make improvements can often times have unintended consequences and hurt us another ways.

One example, so there's a gene called CCR5, and when you edit that people who have this gene are more resistant to HIV, but at the same time they're more susceptible to West Nile virus.

You can do other things like you can fix the beta globin genes so people who have sickle cell disease, this would carry them of that but then all of a sudden they’re more susceptible to malaria, which in certain areas of the world that would be something you don't want.

There's another one that causes Cystic Fibrosis; if you fix that you’re suddenly more susceptible to tuberculosis.

[49:16] So there's a lot of give-and-take with these. Sometimes it's an individual gene and sometimes its how genes interact with each other, and we're just beginning to seek out and learn the way that all these individual components, how that dance, that interaction between it all is playing out.

Going in there and poking around and making these edits on something that's going to actually live and have to live with these consequences as a human, is ethically very grey and a lot of people say that we should not do this, we should not meddle in these things until we can guarantee that the changes that we make are only going to benefit this person, make their life better, even that is a question that is up for a lot of interpretation.

Lack Of Biodiversity

Daniel Forkner:

[49:51] Yeah I think that's a great example those three that you gave, and that's a great point that we really don't understand all these interactions. The ones that you pointed to, yeah we understand that the sickle cell trait makes you more resistant to malaria.

[50:05] But as an individual, if you go into the doctor and the doctor gives you the option to remove the sickle cell trait from your child and thus prevent the child from potentially passing on this defect to their children, and causing the full sickle cell anemia, there's really no reason not to do that right? It's very clearly a good thing.

[50:24] But for us as a species - going back to the ecologically consequences - what does that do for us when we eradicate this thing from our entire population and now we have increased susceptibility as a whole to malaria?

These are the types of biodiversity problems that it's impossible to predict but would no doubt have a profound impact.

David Torcivia:

[50:44] Historically in the actual real world, organisms that are very biologically similar or even biological clones like for example bananas, they might flourish for a brief bout of time and then a pathogen comes that takes advantage of their homogeneity, can wipe out a species completely and that's what happened to the old banana until we replaced it with our current form a banana that you see that's much less sweet and so much bigger, it’s a different color, and is an entirely different banana.

In fact our current banana is at risk of a fungus wiping it out once again because these are clones of the same exact banana everywhere. It’s susceptible to this disease and maybe somebody can come in with CRISPR and prevent this happening, but at the current rate it looks like the bananas we know it might not live for a couple more decades, and then we'll have to find a new banana to clone and do this all over again with.

So those are the kind of dangers that this biological similarity, this lack of diversity in a species, can bring about.

Social Conformity, Inequality, And Stratification In Society And Sports

Daniel Forkner:

[51:40] Beyond the biodiversity, and this problem of unintended side effects, why don't we talk a little bit about some of the implications for society that everyone has kind of come to associate with designer babies, which is equality and discrimination.

I mean we have a world that's right now very stratified, and if you think it's unequal now just imagine how much more unequal it will become when some of these societal inequalities get reinforced by genetic changes.

[52:10] It's a big concern among disability advocacy groups. When we look at certain differences in our population as problems that need to be fixed, then it encourages more discrimination against people who have these differences like blindness, or deafness, or maybe someone's a little bit on the autism spectrum. And now we point to them and say “look this is a problem that needs to be fixed” and that mentality has the potential to widen these stratifications in our society.

David Torcivia:

[52:39] So we have genetic bills right now that prevent discrimination at least in terms of healthcare based on your genes; there's a bill called GINA that does this, but they’re already writing loopholes around it they’re trying to get around it as genetic sequencing becomes more affordable and we have a better understanding what all these genomes do. Imagine a future where your child is born, and they didn't get some gene fixed on them and they're more susceptible to heart disease, and they're denied medical insurance because “sorry you’re a risk, you’re too expensive to insure because we know you have a higher risk for heart disease so sorry you're out of luck, you gotta buy that yourself.”

There's lots of different forms that this discrimination can occur that we don't consider, but a lot of this has been discussed before and it always ends up degrading into that sort of Time Machine world where there's a race of super genetically perfect people, and then like the rest of us are Neanderthal Morlocks wandering around, and I think lots has been said about that and a lot of it is interesting.

I don't know how much more we can add to it but one thing I want to discuss, in this same vein is like what does this mean for sports right? And what does CRISPR mean for sports, so not just designer babies but what if I can come and CRISPR myself, and give myself more muscles, more endurance, more hemoglobin production, and this is my genetic code that's producing this; that's not doping that's not doing drugs or steroids – artificially changing myself - this is my genetic code making my body do these things.

How they going to get around that? How can you possibly have a fair playing field when that's the case, when people are basically engineered at that point on a genetic level to be hyper athletes?

Daniel Forkner:

[54:16] You know I think it would be impossible to stop but perhaps, you know how like NASCAR right now has weight limits on cars right? And in fighting; in wrestling and all this you have different weight classes.

Maybe we'll see a world where you as as an athlete step up to the plate and they say “hold on let's take your blood and let’s sequence your genome and if you have this gene activated you need to go back into the lab and get it corrected because that gives you an unfair advantage,” but probably more realistically it's like you said we'll just see even more of these huge gaps between elite athletes who can take advantage of very specialized modifications that improve their endurance or whatever.

David Torcivia:

[54:54] Well even more immediately, when are we going to see the first genetically engineered Olympic team?

This technology exists and I don't want to point fingers at China all the time but they do get a lot of flack with the way they train their Olympic team, especially with some of the people, and China is right now probably the forefront of this CRISPR, of this gene editing technology - they were the first to try embryonic experiments - and they’ve gotten a lot of flak from some of the western researchers saying that they're doing too much too quickly and they’re being reckless with it, but beyond that it's not a big jump to say well maybe China will field the first CRISPR modified hyper genetic team at the Olympics and how is that going to play out?

Daniel Forkner:

[55:32] That'll be a record year for viewership at the Olympics.

David Torcivia:

[55:35] What if, to get back into our malicious biohacking, let's say I'm a bookie and I am betting against a team or whatever and the World Cup is in Qatar and it's hot. So we infect these soccer players, these football players, with diseases - these genetic vectors that only will activate at certain temperatures, or certain endurance levels of their body - and then you can take out opponents; make them sick, make them underperform, and make millions of dollars off your bets.

[56:05] That'll be a fun application of this technology in a way that has direct economic benefits. I think it’s very possible to happen at some point.

Daniel Forkner:

[56:14] There's certainly a lot of money being pumped into the technology right now so it's not infeasible; there's a lot of people betting that this technology is going to be able to reap billions and billions of dollars.

David Torcivia:

[56:25] Trillions. Let’s be honest this is trillions of dollars here.

Daniel Forkner:

[56:28] We're talking about enhancements to athletes but what about other cohorts of our population like employees or workers of factories?

David Torcivia:

[56:36] So genetically engineering people to be more productive workers, to decrease ADHD, or to tweak genes that make people better at concentrating, or need less sleep as we mentioned before.

I mean that's a really interesting topic where we can almost create a class of people who are designed to be workers, and it becomes a necessity if you want to survive and live in the world you have to have these performance enhancements in order to be able to make do.

But what do we lose when we start doing that? Like “yeah okay you’ll live a little better life economically maybe, but what are we sacrificing in order to create these things?

Adapting To Underlying Societal Flaws

Daniel Forkner:

[57:10] For me this is the biggest part of all this.

Once we start talking about enhancements or improvements to our genetic code, we have to realize that when we say the word “Improvement” or “enhancement” it's within the context of our underlying societal structures and values, and if we are not comfortable or even aware of these underlying societal values and structures, then we may become disappointed when these enhancements really only deepen these unfavorable values.

Like you're talking about workers. If a very high priority and a very big societal value that we have is productivity, then that means that our enhancements may fall along the same line. If we can suddenly engineer ourselves to have less sleep, why would that be an improvement? Could it be because now instead of having an 8-hour workday employers can feel justified by demanding an 11-hour work day?

And would that really feel like an improvement on our lives?

David Torcivia:

[58:09] Well for me also it's the traits that we decide to select for. So these societal pressures of our economic systems of capitalism, of our cultural pressures, will create a specific type of individual. But how much is created in advance by people who live outside that box? Who are the weirdos and strangers, who are our artists, and our philosophers?

People that aren't content to live in this certain way and want to explore something else because they are creative; because they have mental problems; because they are prone to depression; or whatever it is that gives them an alternate viewpoint of the world. Something we might lose when we genetically edit people and embryos in order to better maintain a specific way of seeing the world, of living with the world, and optimizing themselves for that particular mold.

We I think as a society, as a race, are going to lose out on a lot of things and a lot of potential when we start doing that, and unfortunately I see the likelihood of this occurring as fairly great.

Daniel Forkner:

[59:03] That's a great point. We pointed out that a reduction of biodiversity being an unintended side-effect of some of these changes, but you point “hey we could be losing societal diversity” right? The types of people that we have in our society may become more and more conformed to a certain lifestyle as we reinforce those lifestyles with genetic alterations.

David Torcivia:

[59:24] Right and then those systems that become entrenched that we optimize people for, if it turns out we have better systems… alternatives, alternatives to capitalism whatever it is, then it becomes harder to switch to those because we’ve optimize ourselves for a way of living and experiencing the world and putting ourselves and organizing all this together in what might not be the best way.

At that point the question becomes how do we change, how do we fix this? And it's that much harder than it already is.

Daniel Forkner:

[59:50] If we can take one more example, I like how you mentioned earlier you know some malicious executive at McDonald's could convince someone to deploy a genetic change that makes us better able to process or digest their food.

Well obesity is a big problem on the rise in our society, and let's say we look at obesity and we say “okay this is a problem” obviously the source of this problem is some kind of underlying lifestyle, some variable that's been introduced into society that -

David Torcivia:

[1:00:20] Maybe carbon dioxide.

Daniel Forkner:

[1:00:22] Maybe carbon dioxide that's a good point.

But if we just look to the easy solution of genetic alterations and say “well let's just change a few genes here and there and we'll change our ability to digest certain food and we'll just get rid of obesity and it won't be a problem anymore.”

The result will be that we haven't really changed the underlined lifestyle or variables that were causing obesity in the first place; all we've done is adapt our self to those harmful lifestyles. So now like you said it becomes even harder, and even more difficult to get away from those habits and variables that have been introduced right? So now we're more depending on a certain type of food or we're more adapted to this sedentary lifestyle that reduces the variability in our lives.

David Torcivia:

[1:01:06] Well and also I mean how many times has the government gotten like our nutritional scale wrong?

They keep rebuilding that food pyramid into different things and we might find that we optimize ourselves for things that are just factually, physically, scientifically wrong.

And at that point we start changing reality around us to better suit whatever it is that we think is best but might not be the case. I mean that has really interesting philosophical questions talking about redefining everything around us to better match the way that we want to live, including ourselves like our genetic code.

That seems crazy to me but is well within the realm of possibility with this technology.

Daniel Forkner:

[1:01:46] Many dietitians will tell us that eating and consuming dairy products every day is not so healthy for us, and whether that's true or not, I'm sure that with the money involved with that product if the dairy industry got its way then we would all be genetically altered so that it is factually true that it's healthy for us to consume dairy and we would not have a say in it anymore.

David Torcivia:

[1:02:09] This is I mean really the Wild West of what's possible. We really need to totally rethink our relationship with everything at this point, because our ability to change ourselves in order to modify ourselves, is going to change how we interact with the world and with each other.

[1:02:25] And if we aren't asking ourselves these questions as we go along, as we look at this technology… not just saying “well how can this be used wrong,” but “why should we use this in the first place; why should we make these changes beyond curing very basic diseases?”

If we aren’t asking these tough questions, evaluating not just the surface level but also systemic questions, then we have a very great risk of just fixing symptoms and not curing anything at all, and making all of our lives and the world a worse place because of that, especially to those who don't have access to these Technologies.

What Could Have Been

Daniel Forkner:

[1:02:58] You know a thought just occurred to me as we were talking about this; I think it's one of the sad things about our world and the way we look at history, is that it's impossible to see what could have been.

So even if some dystopian genetic altered reality of conformity comes about in the future, people that exist in that future will be able to point to something in their reality that disproves everything we’re talking about right now; all of our concerns.

They’ll be able to say “look how healthy and financially secure we are. Those people that worried about this germ line editing and the consequences for society, they didn't know what they were talking about we have a good life.”

But they will only be looking at the reality in front of them, and not the reality that could have been. In the same way, how much are we missing out on right now as a society because of things like our ambient levels of CO2 or that lead exposure that we talked about last week. Or our underlying capitalistic structures that thrives on exploitation and marginalizing people.

For all the good that we can identify, and all the bad, it is impossible to know how dumb or disadvantaged, or behind we are compared to a world that could have been.

[1:04:10] So we should spend more time thinking about how the world could be, in hopes that we can actually get there.

What Can We Do?

[1:04:17] The question at the end of these episodes always comes back to what can we do? And there are some ideas being posed by researchers and experts in these fields related to our regulatory environment.

So there has to be a balance right between the regulations that are imposed on these technologies. If you're too strict and say you know you can't research this, or you know this is too dangerous so we don't want researchers involved with it, then you're really just going to push scientists to behind closed doors where they're not going to be sharing their research and that's bad, or you'll just encourage other countries with maybe less ethical standards to pursue these types of experiments and research.

So we definitely want our scientists and our researchers to be involved in a healthy regulatory environment, but at the same time, if we're to lax then we could be looking at more risk right? With some of these gene drives that could potentially wreck ecological havoc; we want to be able to weigh the benefits with the risks, and so there has to be a balance, and that's going to take a lot of discussion, it's going to take a lot of voices involved not just scientist, but experts in law, and bioethics, and us as well.

David Torcivia:

[1:05:31] And that “and us” part is really important.

Because it's so easy, because this technology is difficult to understand and strange, to just leave this for scientists and politicians to figure out and sort out in a regulatory matter, and just say “whatever it's too complicated, we’re going to let them deal with it.”

But something that’s going to have such huge effects on each of our individual lives and our society and culture at large, is something that we all need to be involved in. We need to be part of this conversation, we need to be talking to each other, and we need to be talking to our politicians, and we need to be talking with researchers about what we feel we want this technology to be, how we want it to be used, what direction we want it to be pushed in, and what regulatory framework we want on the way. Whether we want to see this weaponized research, whether we think the military like DARPA - who is one of the major funders of these gene drive - should be putting money in it.

Whether these billionaires should be able to patent all these genes as they go along. These are all questions that we say “does this technology belong to the researchers, to the politicians who are making these laws?” Or is this because it's a part of us, a physical genetic part of us discovered in nature, reused by scientists in order to modify each and every one of us…

This is our technology, this is us, this is life, this is the genetic code that flows through all of us. We need to be a part of this, we need to be a part of this conversation, and we need to be able to direct where this goes and how it's used because ultimately we're the ones are going to have to deal with the consequences.

Daniel Forkner:

[1:07:00] There are a lot of changes going on in our society, and a lot of these changes are going on behind closed doors, and we as people of this society need to take a bigger role in these conversations, whether that's CRISPR - yes the science can be complicated but these ethical considerations and the implications of this technology, are something that we can all grasp and that we can all talk about.

In a big F U to America, Ajit Pai and the FCC board came out with their vote a couple months ago to get rid of net neutrality against the broader wishes of the American people. There were over 22 million comments related to the net neutrality vote that the American people had put forward, and at this vote in the comments that the FCC board made, they did not mention at all these comments that we as American people had expressed to them; they didn't even address it.

We as a broader people are being left out of some of the most important decisions being made that will shape our future, and if we don't assert ourselves and we don't get involved in the conversation, it’s been demonstrated pretty clearly that no one else is going to take our concerns into consideration.

That's something that we just absolutely have to change.

David Torcivia:

So that's a lot to think about; that’s a lot to take in; hopefully you’ll bring this to your friends, your family, discuss these topics because they are important. Played them this show, tell them that they need to be concerned about this stuff and we really hope that all this kicks you into action.

That wraps it up for this week. If you want to learn more about this we have a bunch of research papers and sources on our website at ashesashes.org.

You can also find news, links, and even a couple memes on our social media @ashesashescast on your favorite network.

Daniel Forkner:

A lot of time and research goes into making these episodes possible. We will never use ads to support this podcast, so if you enjoy it and would like us to keep going you can support us by giving us a review and recommending us to a friend.

David Torcivia:

Next week we got a really great show continuing our surveillance series. We hope you'll tune in for that, but until then this is Ashes Ashes.

Bye bye.

Daniel Forkner:

Bye.