36
ISSN 0740-9737

GeneWatch Vol. 26 No. 1

Embed Size (px)

DESCRIPTION

Genetics in Agriculture

Citation preview

Page 1: GeneWatch Vol. 26 No. 1

ISSN 0740-9737

Page 2: GeneWatch Vol. 26 No. 1

January-March 2013 2 GeneWatch

GeneWatch is published by the Council for Responsible Genetics (CRG), a national, nonprofit, tax-exempt organization. Founded in 1983, CRG’s mission

is to foster public debate on the social, ethical, and environmental implications of new genetic technologies. The views expressed herein do not necessarily represent

the views of the staff or the CRG Board of Directors.

address 5 Upland Road, Suite 3 Cambridge, MA 02140 Phone 617.868.0870 Fax 617.491.5344

www.councilforresponsiblegenetics.org

board of directors

sheldon KrimsKy, Phd, board chair Tufts University

evan balaban, PhdMcGill University

Paul billings, md, PhdLife Technologies Corporation

sujatha byravan, Phd

Centre for Development Finance, India

robert desalle, Phd

American Museum of Natural History

robert green, md, mPhHarvard University

jeremy gruber, jdCouncil for Responsible Genetics

rayna raPP, PhdNew York University

Patricia Williams, jdColumbia University

staff

Jeremy Gruber, President and Executive DirectorSheila Sinclair, Manager of Operations

Samuel Anderson, Editor of GeneWatchAndrew Thibedeau, Senior Fellow

Vani Kilakkathi, Fellow

editorial & creative consultant

Grace Twesigye

GeneWatchJanuary-March 2013VoluMe 26 nuMber 1

editor and designer: Samuel W. Andersoneditorial committee: Jeremy Gruber, Sheldon Krimsky,

Ruth Hubbard

Unless otherwise noted, all material in this publication is protected by copyright by the Council for Responsible Genetics. All rights reserved. GeneWatch 26,1

0740-973

This issue brought up a few “worlds colliding” moments for me, but in a good way. When I’m not editing GeneWatch, I work at a nonprofit called New Entry Sustainable Farming Project, where I’m the livestock and poultry guy. I also grew up on a sheep farm, and across the twenty-plus years my dad has been keeping detailed re-cords on each ewe and lamb, I have seen firsthand the subtle but remarkable effects of selective breeding.

A couple of years ago, through my work at New Entry, I had the privilege of attending the International Poultry Exposition in Atlan-ta. It’s the largest poultry industry event in the world, and I do mean “industry”—these are the guys in business suits, not Carhartts. I al-ready knew that most conventional poultry farmers—particularly the ones who raise meat chickens, or “broilers”—don’t end up mak-ing much money for their increasingly limited role in the poultry supply chain; but the gaudiness of the industry booths (three-story chicken-shaped displays, open bars galore, that sort of thing) said very clearly that someone was making money. A typical broiler spends all but two or three days of its life under the care of a farmer, who has signed a contract with Tyson or Perdue or Pilgrim’s Pride to shelter and feed the birds from when they arrive as day-old chicks until the company sends a crew to take them to the processing plant at five or six weeks. But all the real action happens before and after that. I could go on for pages about the “after” (seriously, send me an email if you really want to hear about it), but what concerns us in this issue of GeneWatch, since we are examining the role of genetic technologies and research in agriculture, is the “before.”

The vast majority of broilers in the U.S. come from just four poultry genetics companies, all of which were attempting to make a splash at the International Poultry Expo. Each of their extrava-gant displays featured information about the company’s genetic offerings, different broiler pedigrees which are supposed to be tai-lored to meet subtle differences in demand by the hatcheries and the vertically-integrated poultry companies that bring the birds to market. Aviagen is the largest of the broiler companies, yet it offers just a handful of pedigree lines: Indian River, Arbor Acres Plus, Ross 308, Ross 708 and Ross PM3. Aviagen had made glossy one-pagers for each brand, each with a photograph of that variety’s clean white spokeschicken and a list of its “specs”—what size it will be at dif-ferent ages, how much feed it will need to get there, and figures denoting flock uniformity. Uniformity struck me as an especially

Editor’s Note Samuel W. anderSon

comments and submissionsGeneWatch welcomes article submissions, comments and letters to the editor. Please email [email protected] if you would

like to submit a letter or any other comments or queries, including proposals for article submissions.

founding members of the council for responsible geneticsRuth Hubbard • Jonathan King • Sheldon Krimsky • Philip Bereano

Stuart Newman • Claire Nader • Liebe Cavalieri • Barbara Rosenberg Anthony Mazzocchi • Susan Wright • Colin Gracey • Martha Herbert

Continued on page15

Page 3: GeneWatch Vol. 26 No. 1

GeneWatch Vol. 26 No. 14 The Living Legacy The President of Seed Savers Exchange

talks with GeneWatch about a grand collective effort to preserve crop diversity, from seed banks to garden plots all over North America. Interview with John Torgrimson

7 The Safe Seed Initiative

8 Patented Seeds vs. Free Inquiry Restrictions on independent research reduce our knowledge of genetically engineered crops. By Martha L. Crouch

10 Creatures Great and Small You’ve heard about seed banks for preserving crop diversity, but what about livestock? SVF Foundation is on it. Interview with Sarah Bowley

13 Food, Made From Scratch Are synthetically modified organisms the next step in agricultural biotechnology? By Eric Hoffman

16 Bill’s Excellent African Adventure: A Tale of Technocratic Agroindustrial Philanthrocapitalism Bill Gates’ approach to philanthropy promotes the wrong option for African agriculture. By Phil Bereano

18 Ag Biotech Policy: 2012 in Review From GMO labeling to new problems with genetically engineered salmon, it was an eventful year. By Colin O’Neil

20 In the Bullpen: Livestock Cloning With the backing of a few deep-pocketed investors, commercial livestock cloning is no longer a thing of the distant future. By Jaydee Hanson

21 GMO Labeling: What’s Next?

22 Agricultural Technologies for a Warming World GMOs have been presented as a silver bullet to shield agriculture from the effects of climate change, but ecoagricultural systems hold far more promise and far less risk. By Lim Li Ching

24 The State of the Science Most of the evidence for the safety of genetically modified food crops comes from studies that only look at short-term effects … and from information provided by the same companies who sell those seeds. By Stuart A. Newman

26 Glypho-gate Last fall, a paper finding health problems in rats fed Roundup herbicide and genetically modified Roundup-resistant corn brought out strong reactions from proponents and opponents of GM crops. Now the authors are presenting new findings on the most widely used herbicide in the world. With Gilles-Eric Séralini, Robin Mesnage, and Benoît Bernay

28 Life, the Remix They’re both “biotechnology,” but genetic engineering is fundamentally different from traditional breeding. By Martin Dagoberto

30 Book Review: Race in a Bottle Jonathan Kahn’s book tells the story of the rise, fall, and fallout of BiDil, the notorious race-based drug. By Lundy Braun

32 Notes: CRG Founder Debates Human Genetic Engineering on NPR, PBS

32 Topic Update: Gene Patents Monsanto Takes a Farmer to Supreme Court

33 Endnotes

Image: Seed Savers Exchange

Page 4: GeneWatch Vol. 26 No. 1

January-March 2013 4 GeneWatch

Seed Savers Exchange is a non-profit organization dedicated to saving and sharing heirloom seeds, based in Decorah, Iowa. Seed Savers Exchange was one of the original signers of the Safe Seed Pledge in 1999. Learn more at www.seedsavers.org.

John Torgrimson is Executive Director of Seed Savers Exchange.

Today Seed Savers is both a grass-roots organization and a giant in preserving North American crop genetics. How has the organiza-tion evolved since it was founded in 1975?

Seed Savers was started in 1975 when Diane Ott Whealy and her husband Kent received two varieties from Di-ane’s grandfather. One was a morning glory, now known as “Grandpa Ott’s” morning glory, and one was the Ger-man Pink tomato, and he told them the seeds had come from Bavaria with Diane’s great-grandfather. Both Kent and Diane were quite taken with the responsibility that they now had, and

they thought: “Are there other people in the United States who have simi-lar responsibility?” They put a letter to the editor in Mother Earth News and Organic Gardening and all of the sudden people started corresponding with them. But I don’t think they had any vision that Seed Savers would grow to where it is today.

Those are our humble beginnings, but pretty much the same thought process is in place today: the need to protect heirloom varieties and open-pollinated varieties, but also to ensure that they are being distrib-uted and shared, and that people are growing them.

We approach our preservation work in two ways. One is that we’re very much about participatory pres-ervation; we have 13,000 members worldwide, and we have an inter-nal exchange called our “yearbook,” where members grow and list variet-ies that they want to share with other members. Right now our 2013 year-book has about 700 members offer-ing about 19,000 different varieties. This kind of participatory preserva-tion is how we started.

Secondary to that is the ex situ seed bank that we maintain. We have a seed bank on site where we maintain more than 20,000 varieties of seed. We back it up at the USDA seed bank in Fort Collins, and we are now also distributing some of our seeds to Svalbard Global Seed Vault in Norway.

It seems that today, gardeners and certainly farmers are much less likely to collect and save their seeds. How did that come about?

Prior to World War II, most farmers would take their best seed from the harvest and carry it over for the next spring. That changed dramatically with the onset of hybrids after World War II, in the 1950s and beyond. Farmers started looking for better yields and increased sugar content, for example, in corn. That really changed how farming was done in America, and continues today with genetically engineered seeds being the predominant crops grown in the United States.

A hundred years ago, almost every

The Living Legacy

The President of Seed Savers Exchange talks with GeneWatch about a grand collective effort to preserve

crop diversity, from seed banks to garden plots all

over North America. IntervIeW WIth

John torgrImSon

Image: Seed Savers Exchange

Page 5: GeneWatch Vol. 26 No. 1

GeneWatch 5VoluMe 26 nuMber 1

community of reasonable size had their own seed company. In Decorah, Iowa, which was about 800 people, there was Adams Seed Company. Like many seed companies, they fo-cused their attention on varieties that were being grown regionally. Over time, there was a consolidation of these seed companies, and post-1950, the demand for these regional products started to decline, and we started to lose those seed companies.

Part of our mission has been to not just get seeds that have been passed on generationally in families, but also to find seeds that were grown com-mercially and are no longer in the marketplace. So part of our collec-tion is family heirlooms, part of it is former commercial open-pollinated varieties, and we also have some varieties from expeditions to other continents.

Say a gardener goes to buy seeds off the rack at the nearest hardware store. How many of those are likely to be open-pollinated today?

There’s actually a tremendous de-mand for heirloom varieties now. Even Burpee’s, which has traditional-ly been a seed company that special-izes in hybrids, is now, I’ve been told, selling over 400 varieties of heirloom seeds. You’ve got several seed com-panies that have built their business plans solely around heirloom seeds. Baker Creek, out of Mansfield, Mis-souri, is a good example.

That market continues to grow as gardeners start to look at the kind of varieties they want to grow in their garden. And quite frankly, I don’t have anything against hybrids—I’ve grown hybrids in my own garden. It really depends what the gardener is looking for. I grow about six types of tomatoes, for example. They’re all heirloom now, but if I have a problem with a pest or disease, I might get a

A Seed Savers Exchange field crew member bags corn tassels in order to collect pollen for hand pollination. Photo: Seed Savers Exchange

Page 6: GeneWatch Vol. 26 No. 1

January-March 2013 6 GeneWatch

grown, or the amount of genetic di-versity within a crop type. Take last year’s drought for example—some plants perform better given the con-ditions. Last season we noticed late-pollinating corn fared better because it benefited from late and much needed rains. But the point is you will never be able to predict all the traits that will benefit your crops—that’s why diversity is so important.

What’s the process of collecting a new variety at Seed Savers—do people just send you seeds and ask if you could include it in the collection?

Any varieties sent to SSE must meet our accession policy. Varieties are documented as they come in to en-sure a thorough history and horticul-tural understanding. Someone might donate seed and say something like “my great-aunt Molly used to grow this back in Indiana in the 1800s,” and usually there’s some documen-tation that accompanies the seeds; but we also know that there are many varieties that are synonyms. In other words, you may call it “Aunt Molly’s tomato,” but its roots go back to a to-mato in a seed catalog being distrib-uted in the 1850s. Right now we’re actually going through a review of our collection. We have a seed histo-rian on staff whose job is to look at the providence of the varieties in our collection and trace the documenta-tion, so we can better classify them for what we do—so we can clearly classify what it is, where it originated, and what its traits are.

Going forward a little bit, we know that there will be modern heirlooms that will be developed. The “Green Zebra” tomato, which was created by Tom Wagner, is a good example. The Green Zebra is probably about 37 years old, and its genetics come from four different tomato plants. If

you mention when somebody asks: “Why is this work important?”

In the Irish potato famine, the “lump-er” potato was the only variety of po-tato that was grown in Ireland at the time. That speaks strongly in itself for the need for diversity. The adaptabil-ity of seeds to regional conditions is extremely important. Certain types of heirloom or open-pollinated va-rieties, if grown for many years in a particular area and seeds are saved, will become adapted to the climate conditions, the soil type, even the area’s pests.

But there are many reasons diver-sity is important, whether we’re talk-ing about the diversity of crops being

hybrid that deals with that particular problem.

The point is that gardeners, for the most part, are growing varieties in their garden for things like taste—there’s nothing better than a vine-ripened heirloom tomato, compared to what you might buy in a grocery store. And there’s the proliferation of farmer’s markets and CSAs—they put heirlooms on a pedestal, and heirlooms can command a better price because of their taste.

When people talk about the impor-tance of crop diversity, the Irish po-tato famine, where late blight wiped out the country’s potatoes, comes up a lot. Is there anything else that

Imag

e: S

eed

Save

rs E

xcha

nge

Page 7: GeneWatch Vol. 26 No. 1

GeneWatch 7VoluMe 26 nuMber 1

you grow a Green Zebra today and keep the seed for next year, you’re going to get another Green Zebra. So we would classify that as a “modern heirloom,” something that is post-1950 and is part of the American gar-dening heritage.

Is there a crop equivalent to the passenger pigeon or the dodo—something that could have been saved but disappeared?

I can’t think of something specifi-cally, but we certainly know that it has happened. I’ll give you a differ-ent example though, of seeds that we rescued. Back in the 1970s, a Swiss chard called Five Color Silver-beet disappeared from the American market. We located this variety being grown in Australia, by a group called Digger’s Club, and we worked with them to reintroduce it to the Ameri-can market. You can go to any heir-loom seed catalog today and you will find Five Color Silverbeet.

Here’s another specific example. We have a historic apple orchard with 550 varieties of pre-1900 apples. Our orchard manager, Dan Bussey—he’s working on a book—he has identified more than 20,000 named varieties of apples that existed in the United States from the 1620s to the year 2000. As of the year 2000, he has identified about 4,000 varieties that continue to be grown. That would mean we’ve lost about 16,000 varieties of apples—they could have been applesauce apples, cider apples, winter storage apples … and all of that came to America, where our conditions make us just about a perfect nursery for growing apples. Now, of those 4,000 apple varieties that remain, most of them are not commercially available. nnn

In 1999, nine seed companies and the Council for Responsible Genetics created the Safe Seed Pledge to connect seed sellers with gardeners and small farmers looking for non-GM seeds. Businesses that sign the Pledge commit to not knowingly buying or selling genetically engi-neered seeds.

CRG formally recognizes vendors through the Safe Seed Sourcebook, available online (see link below). Sellers are en-couraged to advertise the Pledge to consumers through seed catalogs and package labels. So far, over 100 seed sellers have joined this grow-ing movement for agricultural sustainability. If you are a seed vendor and would like to sign the Safe Seed Pledge, just copy and paste the form below into an email, fill it out, and send to [email protected]. If you prefer, you can print the form and fax it to 617.491.5344, or mail to:

Council for Responsible Genetics 5 Upland Road, Suite 3 Cambridge, MA 02140 USA

Safe Seed Pledge Signup 2013

COMPANY NAME:SIGNER’S NAME & TITLE:

ADDRESS:

The Safe Seed PledgeAgriculture and seeds provide the basis upon which our lives depend. We must protect this foundation as a safe and genetically stable source for future generations. For the ben-efit of all farmers, gardeners and consumers who want an al-ternative, we pledge that we do not knowingly buy or sell genetically engineered seeds or plants. The mechanical transfer of genetic material outside of natural reproductive methods and between genera, families or kingdoms, poses great biological risks as well as economic, political and cul-tural threats. We feel that genetically engineered varieties have been insufficiently tested prior to public release. More research and testing is necessary to further assess the poten-tial risks of genetically engineered seeds.

Thank you for your participation!

For the full list of participating companies, visit: www.councilforresponsiblegenetics.org/viewpage.aspx?pageid=261

The Safe Seed Initiative

Page 8: GeneWatch Vol. 26 No. 1

January-March 2013 8 GeneWatch

Legally, the only way to study Roundup Ready soybeans or any oth-er genetically engineered crop, com-mercialized or not, is to go through the company that owns the patents. If the company is willing, it will of-fer researchers—or more likely to-day, their institutions – confidential agreements with the terms under which research can be conducted.

In other words, the companies that stand to profit or lose from the re-sults are ultimately in control of who gets to do research and who doesn’t. Some scientists who have labored under such contracts think that this restricted access gives the seed in-dustry too much say in the kinds of research scientists do and the way their data are reported to the public, stating that “no truly independent

research can be legally conducted on many critical questions” involving these crops.4,5,6

What, then, are tangible conse-quences of biotech companies hold-ing the keys to research on the genet-ically engineered crops that surround us? From looking at the literature, it is inherently difficult to say that a specific study is missing or skewed because of such a policy. However, since learning about the “no re-search” clause, I’ve had opportunities to ask agricultural scientists about how this particular dependence on agribusiness has affected them.7

Some admitted to being put off of particular projects even before ask-ing the company for a contract. They weren’t affiliated with an institution that normally conducts agricultural research and thus weren’t covered by a blanket agreement, or they be-longed to an institution that could not accept the terms offered. One scientist didn’t want to disclose her methods or theories for fear that her ideas would be pilfered at an early stage by company scientists who had more resources and experience with the system she wanted to study.

In fact, it seems that the superior resources of corporations compared with many academic labs have dis-couraged some graduate students from doing projects with genetically engineered crops. Students asking for research materials reported being told by company scientists that “we know everything there is to know

Several years ago I was approached by a colleague who wanted to do preliminary experiments on trans-location of the herbicide glyphosate into flowers of Roundup Ready soy-beans. He needed a few handfuls of a named variety of seed. Since I live in soybean country he figured I might know a farmer willing to part with such a small quantity. “Sure”, I said, “No problem.”

It was, I discovered, a big problem. As part of the technology agreement that farmers sign when they purchase Roundup Ready soybeans, they “…may NOT plant and may not trans-fer to others for planting any Seed for crop breeding, research, or genera-tion of herbicide registration data.”1

This was a surprise to me. I was quite familiar with prohibitions on farmers replanting seeds of geneti-cally engineered crops, a controver-sial innovation of Monsanto’s2 that they vigorously enforce and which has been adopted by other companies as well.3 But as I scientist, I bridled at being prohibited from simply walk-ing into a store or farmer’s shed and leaving with material to study one of the most common plants in my en-vironment. After all, we are swim-ming in these Roundup Ready soy-beans here in Indiana. Every fourth acre of the state is planted in them. Seeds fall out of trucks, volunteer in subsequent crops, and are piled high in grain elevators along our county roads. Soybeans, soybeans every-where, and not a seed to study?

Patented Seeds vs. Free InquiryRestrictions on independent research reduce our knowledge of genetically engineered crops. By martha l. CrouCh

The companies that stand to profit or lose

from the results are ultimately in control of

who gets to do research and who

doesn’t.

Page 9: GeneWatch Vol. 26 No. 1

GeneWatch 9VoluMe 26 nuMber 1

for information about the levels of glyphosate in pollen and nectar of Roundup Ready crops in light of the crises honeybees and other pollina-tors are experiencing. I didn’t find any relevant studies in the public domain.

Maybe if I had been able to send my colleague some Roundup Ready soybeans when he asked, we would know about glyphosate levels in pol-

len and nectar by now, and thus be better equipped to assess environmental impacts. And I wonder how many other studies are missing at least partly because of the impedi-ments to free inquiry from patent-driven research restrictions.

I believe it is in the pub-lic’s interest to bring seeds of genetically engineered crops back into the com-mon sphere where they can be used freely in research. Although doing so won’t remove all corporate influ-ence from scientific stud-ies, it is a concrete step in the right direction towards transparent, reliable infor-mation about these new and impactful technologies. nnn

Martha L. Crouch, PhD, was a graduate student at Yale Universi-ty studying the development of seeds and flowers when genes were first cloned. By the time she headed her own plant mo-lecular biology lab at Indiana University, plant genes were being patented. Now she consults from her home in Bloomington, Indiana, on issues of biotechnology and agriculture for non-profit groups and law firms.

more say, including having seats on university boards.8 I have argued that even public money for basic research is steered towards projects that will support agribusiness.9

It is a wonder that any truly in-dependent studies of genetically en-gineered crops get done against the backdrop of all of these corporate in-fluences. Some scientists rise to the challenge, but my sense is that many

more find it too much of a hassle and decide to work on other issues.

Certainly, when I comb the sci-entific literature for impacts of par-ticular genetically engineered crop systems I am dismayed at how few independent studies I find. This is especially true for impacts on non-target organisms, meaning all of us except weeds and pest insects. For example, recently I was searching

about [whatever],” leaving them with the impression that it would be dif-ficult to carve out a niche in compe-tition with the “big guys.” I’ve also heard of students being strongly en-couraged to change direction when it appeared that their research was heading for a conclusion that was not in the company’s interest. In one case, a student said he was offered a grant if he dropped his current proj-ect in favor of another one after his preliminary re-sults pointed to an issue with crop performance.

Researchers who did persevere and in the end reported results that might damage the com-pany’s bottom line were sometimes refused further access to seeds or other materials. They also faced coordinated attacks on their published work, well beyond what most aca-demics experience dur-ing the normal give and take of scientific critique. Often their work was dis-counted for deficiencies in methodology, such as lack of the most appropriate control plants or reagents, at the same time that they were denied access to these materials.

Other factors weigh against independent re-search in agriculture, of course. Public research in agriculture is in-fluenced by private money and guid-ance at every level. For years, as pub-lic money has dwindled, grants and contracts from corporations have in-creased. So have industry-sponsored endowed chairs, graduate student fellowships, undergraduate teach-ing grants, internships, and other partnerships that give corporations

Image: S. W

. Anderson

Page 10: GeneWatch Vol. 26 No. 1

January-March 2013 10 GeneWatch

SVF Foundation preserves germplasm (semen and embryos) from rare and endangered breeds of food and fiber livestock. In collaboration with Tufts University’s Cummings School of Veterinary Medicine, SVF maintains a library of cryopreserved genetic material from rare cattle, sheep and goats. Learn more at svffoundation.org.

Sarah Bowley is Program Manager of SVF Foundation.

First things first: What goes on at Swiss Village Farm?

SVF is a cryopreservation facil-ity focusing on rare and endangered breeds of livestock. We have a labo-ratory facility, a cryo room, and a sur-gical suite on the farm, and we col-lect and preserve samples—semen, embryos, cells and blood—from en-dangered livestock breeds, focusing on cattle, sheep and goats. We bring the animals here to the farm—col-laborating with Tufts University, who provides all the veterinary care—and after we do the germplasm collec-tion, which takes about a year for each animal, we send the animals to working farms around the country that are trying to keep these breeds going, as we say, “on the hoof.”

Cryopreservation seems pretty in-volved and expensive—why bother with it?

Seed banks have been well es-tablished and are an expected part of agriculture now; that’s how we maintain our agricultural history and genetic diversity for plants. The no-tion of doing genetic preservation for livestock didn’t really take off until the early 2000s. It was thanks to a lot of foresight, especially from Dr. (George) Saperstein (at the Tufts Cummings School of Veterinary Medicine), to say: Look, we are los-ing breeds at an extremely alarming rate. The Food and Agriculture Or-ganization estimates that globally, we are losing one breed of livestock per month. It has taken eight to twelve thousand years to domesticate all of these species and create the diver-sity within these species that has al-lowed humans to acclimate ourselves around the world to all these differ-ent regions, bringing livestock with us and finely tuning them through selective breeding. Now, with in-dustrial agriculture and all the ways we manipulate the environment, the genetic base of livestock that we use has really narrowed. All of these dif-ferent breeds that took us thousands of years to develop are being lost very rapidly; but if we can cryopreserve those genetics, they will be available to us even if the breed goes extinct.

Here’s a question you must hear a lot: If these breeds are rare, isn’t there a reason for it? Doesn’t that mean that they aren’t productive

enough for farmers to want them?

Absolutely, but the key is that they aren’t productive enough in today’s agriculture. Since the 1950s we’ve seen a rapid shift in the way we pro-duce food. It involves a lot of fossil fuels, and for livestock it involves a lot of intensive management. This means feeding a lot of grain, using a lot of antibiotics, using deworm-ers, most animals need help giving birth and then don’t necessarily take care of their own babies … because in order to mass produce food for a cheap price, we have to step in and manage the animals in each aspect of their production. What we’ve lost are a lot of those genetic traits that allow the animal to produce on its own. So if you put one of these finely-tuned Holstein cows out on pasture and expect it to get bred by a bull, raise

Creatures Great and SmallYou’ve heard about seed banks for preserving crop diversity, but what about livestock? SVF Foundation is on it. IntervIeW WIth Sarah BoWley

Page 11: GeneWatch Vol. 26 No. 1

GeneWatch 11VoluMe 26 nuMber 1

or their milk or their meat has differ-ent nutritional qualities—but very few of those have been scientifically studied to date. So the Gulf Coast Native sheep are exciting because it’s one breed that was looked at, some money was invested into figuring out why they don’t seem to be affected by parasites, and they ended up proving the genetic link.

Another trait is the Santa Cruz sheep’s tail. This was a feral island population that became its own breed because it was geographically isolated for so long. They were left on an island off the coast of California, and for three or four hundred years there was no human intervention and no outbreeding with any other type of sheep. When they were taken off of the island in the 1970s, people noticed they had developed a trait where they shed their wool—since there was nobody to shear them on the island—and they had developed something called “rat tail,” a very short, woolless tail. Tail docking is a problem in the sheep industry; people feel that it’s inhumane, but if you leave the tail on then when it gets dirty, especially when it’s time to lamb and in the summer, the sheep can have serious problems with flies. So it’s inhumane whether you leave the tail on or take it off; but with sheep that have these small, woolless tails, there’s no need to dock.

Traits like that can develop in these island breeds. Unfortunately, the Santa Cruz breed is not only crit-ically endangered, it’s also really hard to get anybody to work with them because they’re not economically vi-able—they’re very small, they don’t produce a lot of wool, and they don’t grow a very heavy lamb. It’s hard to convince farmers to invest in this kind of sheep just because it could have useful traits for farmers in the future, but it’s one that we represent in our collection here. If we isolate

we are especially excited to represent in our bank. Gulf Coast Native sheep are one breed that is critically en-dangered, but they are scientifically proven to be genetically resistant to internal parasites. There is a differ-ence in the lining of their digestive tract which prevents parasites from latching on like they do in most sheep breeds. When a sheep is infected, the worms suck blood from the sheep’s gastrointestinal tract, the sheep be-comes anemic, and can eventually die with a heavy enough parasite load. Gulf Coast Native sheep are much less susceptible to this, and studies

at Louisiana State University and a couple others proved the genetic link and how it actually works.

There’s a lot of anecdotal evidence that accompanies most of these breeds—they’re really good mothers,

a calf, eat only grass, and produce a lot of milk, you’re going to fail. That’s because we have provided so much for her over so many generations that she has lost that ability to reproduce and take care of her offspring on her own.

What we’re really interested in representing with these rare breeds are those genetic outliers that con-tribute to things like mothering abil-ity, disease resistance, parasite re-sistance, feed efficiency—and there are a lot of behaviors and nutritional aspects that we aren’t even aware of yet. And we’re losing breeds before we know what traits we’re losing with them.

Why focus on cattle, sheep and goats?

When this project started in 1998, the reproductive technology for cryopreserving samples from rumi-nants—cattle, sheep and goats—was already there. Collecting and freez-ing semen and embryos has been done for cattle for a long time, and it’s where a lot of the technology came from that human IVF therapies are now based on. So it wasn’t a guessing game: we knew we could collect em-bryos from cattle, sheep and goats, freeze them, and have them be viable afterwards. The technology isn’t quite there yet to allow us to successfully freeze and then utilize swine germ-plasm. A lot of people now freeze se-men from boars, but mostly they are commercial lines, and there’s a wide variation; whereas with ruminants, if you can freeze one goat’s genetic material, you can freeze pretty much any goat’s.

Among endangered breeds, are there any traits you especially tar-get for preservation?

There are some cases we know of that

Sarah Bowley of SVF Foundation with Drs. George Saperstein and David Matsas of the Tufts Cummings School of Veterinary Medicine. Photo: SVF Foundation

Page 12: GeneWatch Vol. 26 No. 1

January-March 2013 12 GeneWatch

Unfortunately, that’s the situation we are replicating with the Holstein today. We have come to rely on one very inbred type of cow to provide over 95% of all our dairy products. If we get some superbug that comes along and wipes out the Holsteins, we’re going to be in a lot of trouble, because we haven’t supported those other populations to fall back on.

The Irish potato famine was caused by late blight; for cattle, do you have any idea what that superbug might be?

Not at this point, but if you talk to any dairy farmer, they are very aware that the Holsteins are a susceptible population. There’s an estimated ef-fective population of 34 individuals among the whole Holstein-Friesian population. So it’s a pretty fragile, inbred population that we’re relying on. If you look at other breeds, there may only be 400 American Milking Devon cattle left, but their genetic diversity is actually much more than the Holsteins, even though there are so many more Holsteins out there.

Instead of a superbug, could it be something like skyrocketing grain prices, where it would become a problem to rely on breeds designed for grain instead of grass?

Absolutely. And we’re seeing a lot more interest in these breeds as the sustainable farming movement grows. It’s no longer just a fad to have a rare breed. These smaller produc-tion breeds are really fitting into grass-based farming, going back to these types of farming that are easier on the environment. They’re not high production or high output breeds, but they usually do very well fitting into a small farm with multiple spe-cies on it or that has both crops and livestock. nnn

disease resistance if they were going to survive to carry on their genetic traits.

When we at SVF are trying to explain our mission to the public, two things that we like to bring up are dog breeds and the Irish potato famine. When people are confused about why there are different types of cows or different types of sheep, we talk about how there are different types of dogs. Dogs are one species, but we have developed many differ-ent breeds to suit our purposes; you would never ask a German Shepherd to do what a collie does, or vice ver-sa. We’ve done the same thing with cows: they are adapted for different parts of the world and different jobs we need them to do.

And the Irish potato famine is a good example of monoculture. Irish peasant farmers relied on one vari-ety of potato, and when blight came through and wiped out that potato crop, millions of people starved or were displaced because they only had one variety of crop available to them.

that genetic trait for short tails, we may be able to introduce it into com-mercial sheep breeds, which would be a major change in the industry.

It seems like there’s a theme here—a lot of the breeds SVF works with developed survival traits as a feral population.

There are two main categories of breeds that we’re looking at. One is the heritage breeds, which have a kind of cool, cultural story that ac-company them. These are the ones we think of as what our forefathers brought over, the cattle that gave you milk and pulled the plow and then you ate it at the end of its productive life.

The other type are the ones that were feral, the island or landrace breeds. These have some of the more interesting genetic characteristics because for generations it was sur-vival of the fittest. They had to raise their own babies, find their own food, and develop some sort of parasite or

Page 13: GeneWatch Vol. 26 No. 1

GeneWatch 13VoluMe 26 nuMber 1

have been engineered and patented by chemical companies, including Monsanto and Bayer, to either with-stand increasingly heavy doses of herbicides or to produce their own systemic pesticide.

Synthetic Biology - Extreme Genetic Engineering

In the second decade of the 21st century, we are likely to see even more radical changes on the horizon,

this time via a rapidly growing field known as synthetic biology. Syn-thetic biology is a broad term used to describe a collection of new bio-technologies that push the limits of what was previously possible with “conventional” genetic engineer-ing. Rather than moving one or two genes between different organisms, synthetic biology enables the writing and re-writing of genetic code on a computer, working with hundreds and thousands of DNA sequences at a time and even trying to reengineer entire biological systems. Synthetic biology’s techniques, scale, and its use of novel and synthetic genetic sequences make it, in essence, an ex-treme form of genetic engineering.

Synthetic biology is a nascent but rapidly growing field, worth over

$1.6 billion in annual sales today and expected to grow to 10.8 bil-lion by 2016.3 Many of the largest

energy, chemical, forestry, phar-maceutical, food and agribusiness

corporations are investing in syn-thetic biology research and develop-ment or establishing joint ventures,

and a handful of products have al-ready reached the cosmetic, food, and medical sectors with many others not far behind. Much of

this focus is being placed on agriculture applications to become the next wave of GMOs – let’s call them

synthetically modified or-ganisms (SMOs).

Synthetically Modified Organisms

Monsanto, the biotech and

“Agriculture as we know it needs to disappear. …We can design better and healthier proteins than we get from nature.” – J. Craig Venter1

A (very) short history of agriculture

For ten thousand years humans have been manipulating plants for food production. This began at a very basic level, saving slips or seeds from the fastest growing, highest yield-ing, best tasting and most nutritious plants for the following season. This form of conventional breeding even-tually led to the development of hy-brid crops which involved cross-breeding two genetically different lines in the same genus and usually the same species. These changes in the plants were limited to the genes already present within the plants.

This all changed dramatically with the advent of genetic engi-neering in the 1970s and 1980s. Genetic engineering allowed the transfer of genes between species, even between species of different kingdoms, as when bacteria DNA were inserted into plants—and court decisions allowed, for the first time, patents on life. Since then, genetically engineered organisms, often called genetically modified organisms (GMOs), have become a ubiquitous fea-ture of industrial agriculture in the U.S., comprising roughly 88% of the corn, 94% of the soy-beans, 90% of the canola, 90% of the cotton, and 95% of the sugar beets grown in the country.2 These crops

Food, Made From ScratchAre synthetically modified organisms the next step in agricultural biotechnology? By erIC hoffman

Page 14: GeneWatch Vol. 26 No. 1

January-March 2013 14 GeneWatch

of how these genes, and the biologi-cal systems they are being inserted into, actually work. It is already dif-ficult to assess the safety of a single genetically engineered organism, and synthetic biology raises this level of complexity enormously. To date, there has been no scientific effort to thoroughly assess the environmental or health risk of any synthetic organ-ism, which can have tens or hundreds of entirely novel genetic sequences.

Biotechnology is already regulated poorly in the U.S., and SMOs will only push the boundaries of this anti-quated regulatory system. For exam-ple, the U.S. Department of Agricul-ture regulates GMOs through plant pest laws, since most have been en-gineered through a plant virus. Syn-thetic biology opens up the possibil-ity for SMOs to be created without plant viruses, meaning those crops may be completely unregulated by the USDA—or any agency.

Our risk assessment models for biotechnology are quickly becom-ing outdated as well. Safety of GMOs is typically determined if it is “sub-stantially equivalent” to its natural counterpart. This idea of “substantial equivalence” quickly breaks down when looking at the risk of an SMO which has genes that have never ex-isted before in nature and whose “parent is a computer.”9

An End to Industrial Agriculture As We Know It

Synthetic biology may hold some promises, but is a dangerous path to follow if we don’t know better where it leads. The past few decades of ag-riculture biotechnology have pro-duced a multitude of problems, many of which will be exacerbated by syn-thetic biology, including genetic con-tamination, super-weeds, an increas-ing dependence on ever more toxic industrial chemicals, larger areas of

coconut oil, animal feed additives, and even genetically engineered ani-mals with synthesized genes. Food flavorings may sound benign, but ac-tually pose another set of risks: eco-nomic risks to farmers. These natural botanical markets are worth an esti-mated $65 billion annually and cur-rently provide livelihoods for small farmers, particularly in the global South.7 Replacing the natural pro-duction of these products by farmers with synthetic biology in biotech vats in the U.S. and Europe will have ma-jor socio-economic impacts and may drive smallholder farmers further into poverty.

The Perils of Synthetic Biology

While some of these develop-ments sound promising, synthetic bi-ology also has a dark side. If an SMO were to be released into the environ-ment, either intentionally (say, as an agricultural crop) or unintention-ally from a lab, they could have seri-ous and irreversible impacts on the ecosystem. Synthetic organisms may become our next invasive organisms, finding an ecological niche, displac-ing wild populations and disrupting entire ecosystems.8 SMOs will lead to genetic pollution—as happens commonly with GMOs—and create synthetic genetic pollution which will be impossible to clean up or re-call. Using genes synthesized on a computer instead of those originally found in nature also raises questions about human safety and the possibil-ity that SMOs could become a new source of food allergens or toxins.

What’s different and possibly more hazardous about synthetic bi-ology is that the DNA sequences and genes being used are increasingly different than those found in nature. Our ability to synthesize new genes has far outpaced our understanding

chemical giant, recently announced a joint venture with Sapphire Energy, a synthetic biology algae company. Monsanto is interested in algae be-cause most types of algae reproduce daily, compared to traditional agri-culture crops which only reproduce once or twice a year. Monsanto hopes to isolate traits in algae at a much faster rate than can be done in plants, which could then be engineered and inserted into crops.4 Such technolo-gies will potentially allow increas-ing numbers of (and more extreme) genetically engineered crops on our fields.

J. Craig Venter, a leading synthetic biologist who built the world’s first synthetic genome by copying a rath-er simple goat pathogen’s genome, created a new company, Agradis, to focus on applying synthetic biology to agriculture. Agradis aims to cre-ate “superior” crops and improved methods for crop growth and crop protection. The company plans to create higher-yielding castor and sweet sorghum for biofuels through undisclosed “genomic technologies.”5

There are even plans to “improve” photosynthesis in plants through synthetic biology. Researchers at the Department of Energy’s National Re-newable Energy Laboratory in Colo-rado believe that “the efficiency of photosynthesis could be improved by re-engineering the structure of plants through modern synthetic bi-ology and genetic manipulation. Us-ing synthetic biology, these engineers hope plants can be built from scratch, starting with amino acid building blocks, allowing the formation of op-timum biological band gaps,”6 mean-ing plants could turn a broader spec-trum of light into energy than done naturally through photosynthesis.

Other food and agriculture ap-plications of synthetic biology in the works include food flavorings, stevia,

Page 15: GeneWatch Vol. 26 No. 1

GeneWatch 15VoluMe 26 nuMber 1

unsustainable monocultures, fights over intellectual property and the su-ing of farmers, and the further con-centration of corporate control over our food supply.

Far from making “agriculture as we know it disappear,” as Craig Venter hopes to do, we should work to make industrial agriculture as we know it (and its dependence on biotechnol-ogy and toxic chemicals) disappear, refocusing our energies on agricul-tural systems we know to work, such as agro-ecology and organic farming. For example, a recent USDA study found that simple sustainable chang-es in farming, such as crop rotation, produced better yields, significantly reduced the need for nitrogen fertil-izer and herbicides, and reduced the amounts of toxins in groundwater, all without having any impact on farm profit.10 Such systems have shown to be equally if not more productive than industrial agriculture systems, and are also better for the planet and our climate11 and produce food that is healthier and more nutritious with-out a dependence on hazardous, ex-pensive and unproven technologies.

A moratorium on the environ-mental release and commercial use of synthetic biology is necessary to ensure that our ability to assess its risks and regulate it to protect hu-man health and the environment keep pace with the technology’s rap-id developments, and to provide time to explore and support alternatives.12 Instead of continuing down the road of GMOs to SMOs, let’s look to so-lutions that already exist to create a vibrant, healthy, sustainable and just food system. nnn

Eric Hoffman is a food and technology campaigner for Friends of the Earth.

Editor’s Note (continued from page 2)

ironic figure. I know why it’s there—uniformity is a virtue in the broiler industry largely because it suits the extraordinary amount of mechani-zation, from incubation to process-ing—but it’s a funny thing to say one brand is especially uniform when the photographs on all five flyers looked very much like pictures of one chick-en from five slightly different angles. The differences in their stats—or, rather, the minuteness of those differ-ences—was even more striking. At 35 days (around slaughter time), the Ross 308 is 4.96 lbs, having eaten 7.77 lbs of feed, while the Ross 708 is 4.71 lbs, eating 7.26 lbs of feed.[1] So the Ross 708 converts feed more efficiently— a whopping 1.6% more efficiently, to be exact.

The incredible uniformity of broil-ers derives from their pyramid-shaped family tree. At the bottom of the pyra-mid is a hybrid bird (billions of them, actually) which, thanks to hybrid vig-or, is an even better meat bird than ei-ther of its parent lines; however, if you were to cross two of the hybrid meat birds with each other, that hybrid vig-or would be lost in the next genera-tion. These are end-of-the-line chick-ens, created only for meat and not for passing on their genetics—what in other types of livestock breeding you would call a “terminal cross.”

In order to create this hybrid vigor, you need distinct male and female parent lines. Males are selected for growth rate, edible meat yield, and feed conversion ratio, all traits that make them efficient for meat produc-tion; females are selected for those traits too, and also for egg production, which makes them a more attractive buy for hatcheries. Some of the par-ent stock may also be hybrids, care-fully selected to combine traits that fit (or “nick,” in industry language) with each other and with the opposite par-ent’s line. Hatcheries buy parent stock from genetic companies, which guard the grandparent and great-grand-parent genetics. The top of the pyra-mid—the great-great-grandparents of the birds that will end up as chicken

nuggets—are the “pureline” pedigrees. In 1998, the genetics of the world’s 400 billion Cornish Cross broilers could be traced back to 400,000 indi-vidual birds from just 35 to 40 pure-lines. In other words, for every one million meat birds, there is one com-mon great-great-grandparent.

The following articles tackle the broad topic of “genetics in agricul-ture” from different angles, from ag-ricultural biodiversity to transgenic technologies, from research priori-ties to consumer activism. There will be plenty of discussion about geneti-cally modified crops, but even if that is where your passions lie—in fact, especially if that is where your pas-sions lie—I hope you will read further. There is plenty to say about GMOs, but the role of genetics in shaping the plants and animals that feed us goes much, much deeper. nnn

Cover image: Adapted (but not much) from the cover of the 1906 Johnson & Stokes seed catalog, of Philadelphia. Amazingly, although it has changed hands several times since being founded in 1881, Stokes Seed still exists today, now based in Buffalo, New York and Thorold, Ontario.

Page 16: GeneWatch Vol. 26 No. 1

January-March 2013 16 GeneWatch

From 2009 to 2011, Bill Gates’ foundation spent $478,302,627 to in-fluence African agricultural develop-ment. Adding in the value of agricul-tural grants going to multiple regions and those for 2012, the Founda-tion’s outlay to influence Afri-can agriculture is around $1 bil-lion. Of course, Gates is not an African, not a scholar of Africa, not a farmer, and not a devel-opment expert. But he is a very rich man, and he knows how he wants to remake the world.

Gates’ support for ag devel-opment strategies favors indus-trial, high-tech, capitalist mar-ket approaches. In particular, his support for genetically en-gineered crops as a solution for world hunger is of concern to those of us—in Africa and the U.S.—involved in promoting sustainable, equitable agricul-tural policies.

First, his technocratic ide-ology runs counter to the best informed science. The World Bank and the UN funded 400 scientists, over three years, to compile the International Assessment of Agricultur-al Knowledge, Science and Technology for Development (IAASTD). Its conclusions in 2009 were diametrically opposed, at both philosophical and practical levels, to those espoused by Gates. It recom-mended research that “would focus on local priorities identified through

participatory and transparent pro-cesses, and favor multifunctional so-lutions to local problems,” and it con-cluded that biotechnology alone will not solve the food needs of Africa.

The IAASTD suggests that rather than pursuing industrial farming models, “agro-ecological” methods provide the most viable, proven, and reliable means to enhance global food security, especially in light of climate

change. These include implementing practical scientific research based on traditional ecological approaches, so farmers avoid disrupting the natural carbon, nitrogen and water cycles,

as conventional agriculture has done.

Olivier De Shutter, the UN’s Special Rapporteur on the Right to Food, reinforces the IAASTD research. He too concludes that agro-ecological farming has far greater potential for fighting hunger, particularly during eco-nomic and climatically uncer-tain times.

Agroecological practices have consistently proven ca-pable of sustainably increasing productivity. Conversely, the present GM crops, based on industrial agriculture, generally have not increased yields over the long run, despite their in-creased input costs and depen-dence. The Union of Concerned Scientists details GM crops’ un-derperformance in their 2009 report, “Failure to Yield.”1

Second, Gates funds African front groups whose work with Monsanto and other multina-tional agricultural corporations directly undermines existing

grassroots efforts at improving Af-rican agricultural production. Gates has become a stalking horse for corporate proponents promoting industrial agricultural paradigms, which view African hunger simply

Bill’s Excellent African Adventure: A Tale of Technocratic Agroindustrial PhilanthrocapitalismBill Gates’ approach to philanthropy promotes the wrong option for African agriculture. By PhIl Bereano

Imag

e: S

. W. A

nder

son

Page 17: GeneWatch Vol. 26 No. 1

GeneWatch 17VoluMe 26 nuMber 1

with agro-ecological techniques and traditional knowledge.

Mariam Mayet of the African Centre for Biosafety said of the Gates Foundation grant, “(Genetically modified) nitrogen-fixing crops are not the answer to improving the fer-tility of Africa’s soils. African farmers are the last people to be asked about such projects. This often results in the wrong technologies being devel-oped, which many farmers simply cannot afford.”

She said farmers need ways to build up resilient soils that are both fertile and adaptable to extreme weather. “We also want our knowledge and skills to be respected and not to have inappropriate solutions imposed on us by distant institutions, charitable bodies or governments,” Mayet said.

While successful in his chosen field, Gates has no expertise in the farm field. This is not to say that he and his fellow philanthropists cannot contribute—they certainly can. How-ever, some circumspection and hu-mility would go a long way to heal the rifts they have opened. African farm-ers never asked to be beaten with the big stick of high-input proprietary technology; doing so continues neo-imperialism and the perpetuation of foreign-imposed African “’failure.” Africans urge Bill Gates to engage with them in a more broadly con-sultative, agroecological approach. nnn

Phil Bereano, JD, is a Professor Emeritus at the University of Washington and a co-founder of the Council for Responsible Ge-netics. This essay is based on work he and other researchers have done for AGRA Watch, a project of Seattle’s Community Alliance for Global Justice (http://www.seattleglobaljustice.org)

ground in African agriculture. The Foundation’s press release describes it as:

(Providing) tools to ensure that ag-ricultural development does not de-grade natural systems and the ser-vices they provide, especially for smallholder farmers. It will also fill a critical unmet need for integrat-ing measurements of agriculture, ecosystem services and human well-being by pooling near real-time and multi-scale data into an open-access online dashboard that policy makers will be able to freely use and custom-ize to inform smart decision making. The raw data will be fully accessible and synthesized into six simple holis-tic indicators that communicate di-agnostic information about complex agro-ecosystems, such as: availability of clean water, the resilience of crop production to climate variability or the resilience of ecosystem services and livelihoods to changes in the ag-ricultural system.3

This is really a top-down tech-nocratic program, hardly qualify-ing as agroecological. In fact—while it might be a beneficial activity—it could be used as a perfect illustration of trying to use an appealing label to whitewash its opposite. A Gates of-ficial claims that it will be “for deci-sion-makers,” but these users appear to be hierarchical elites, not small-holders—who are unlikely to have “an open-access online dashboard” in their fields.

Genetically modified crops are also supported by the Gates Foun-dation, although they threaten con-ventional and organic production as well as the autonomy of African pro-ducers and nations. In 2002, Emmy Simmons, then-assistant administra-tor of the U.S. Agency for Interna-tional Development, stated that “in four years, enough (genetically engi-neered) crops will have been planted in South Africa that the pollen will have contaminated the entire conti-nent.” Biotechnology cannot coexist

as a business opportunity. His foun-dation has referred to the world’s poor as presenting “a fast growing consumer market.” Referring to the world’s poor as “BOP” (the bottom of the pyramid), he insists they must be subsumed into a global capitalist sys-tem, one which has done so well to enrich him. His philanthropy is really “philanthrocapitalism.”

By and large, Gates’ grants do not support locally defined priorities, they do not fit within the holistic ap-proach urged by many development experts, and they do not investigate the long-term effectiveness and risks of genetic modification. The choice of a high-risk, high-tech project over more modest but effective agricul-tural techniques is problematic, of-fering no practical solutions for the present and near-future concerns of the people who run small farms.

For example, the Gates Founda-tion touted a $10 million grant to Conservation International in 2012 as “agroecological,” an important concept emerging as a touchstone criterion for assessing development assistance. Using the guidelines that Miguel Altieri has laid down, it con-sists of “broad performance criteria which includes properties of eco-logical sustainability, food security, economic viability, resource conser-vation and social equity, as well as increased production. . . . To attain this understanding agriculture must be conceived of as an ecological sys-tem as well as a human dominated socio-economic system.”2 This goes far beyond the definition used, for example, by the OECD as “the study of the relation of agricultural crops and environment.” In other words, in addition to embodying the idea of sustainability, agroecology includes principles of democracy.

However, the Conservation Inter-national grant is merely a program of monitoring what is happening on the

Page 18: GeneWatch Vol. 26 No. 1

January-March 2013 18 GeneWatch

as a result of sustained opposition by farmers, environmental groups and consumers.

GE Labeling Movement Marches Forward

In the fall of 2011 the Center for Food Safety filed a groundbreak-ing legal petition with the U.S. Food and Drug Administration (FDA) de-manding that the agency utilize its existing authorities to require the labeling of all food produced using genetic engineering. On March 12, 2012, fifty-five Members of Congress joined a bicameral letter led by Sena-tor Barbara Boxer (D-CA) and Repre-sentative Peter DeFazio (D-OR) that was sent to the FDA Commissioner in support of the labeling petition. By March 27, over one million public comments had been submitted to the FDA in support of the petition—the largest public response the FDA has ever received.

The labeling movement did not stop at the FDA. Over a dozen la-beling bills were introduced at the state level in 2012 that would have required the labeling of GE fish, GE wholefoods or all GE foods. Ulti-mately, the bills failed under indus-try pressure; but already in 2013, 34 more bills have been introduced in 21 states including Hawaii, Washing-ton, Indiana, Missouri and Vermont, with many more expected by year’s end.

Biotech Riders Emerge in House Bills

Despite increased calls for label-ing and better oversight of GE crops

and foods, Republican members in the U.S. House of Representatives at-tempted multiple times to roll back the clock on GE crop regulations.

On the heels of federal court deci-sions that found approvals of several GE crops to be unlawful, a dangerous policy rider (Sec. 733) was inserted into the FY 2013 House Agriculture Appropriations bill. The rider was intended to strip federal courts of their authority to halt the sale and planting of GE crops and compel USDA to allow continued planting of those crops upon request by indus-try. The rider drew sharp criticism from groups like the American Civil Liberties Union, Earthjustice and the National Family Farm Coalition who viewed it as an assault on the fun-damental safeguards of our judicial system and one that would negative-ly impact the environment, public health and farmers across America.

Following the appropriations rid-er, a suite of policy riders (Sec. 10011, 10013, and 10014) were buried in the House Agriculture Committee’s draft 2012 Farm Bill. These riders sought to: dramatically weaken the oversight and regulation of GE crops and spe-cifically eliminate the critical roles of our most important environmental laws; dramatically shrink the time USDA has to analyze biotech crops, while withholding funds for USDA to conduct environmental reviews; lim-it the regulatory authority of the EPA and other agencies; establish mul-tiple backdoor approval mechanisms for GE crop applications; and force USDA to adopt a national policy of

In a year filled with attention-grabbing headlines about reelec-tion campaigns and congressional roadblocks, the narrowly defeated California Proposition 37 may be the story about genetically engineered (GE) foods that people most recall. However, for those directly affected by biotech policy, 2012 was shaped by more than just Prop. 37. Its issues were driven by chemical companies, farmers and consumers.

Farmer and Environmental Opposition Slows the Chemical Arms Race

The year began like most years in biotech policy, with a major an-nouncement by the Administration during the holidays. In this case, 2012 was ushered in with a statement by the U.S. Department of Agriculture (USDA) revealing that it was moving forward with Dow AgroSciences’ ap-plication for its controversial Enlist GE corn that is engineered to with-stand exposure to the herbicide 2,4-D, a component in the Vietnam era defoliant Agent Orange that has been linked to a number of human health and environmental harms.1 This an-nouncement brought sharp criticism from farmers, environmental groups and consumers, and in January 2013, after a year of strong opposition, Dow announced that it would be delaying the release of its 2,4-D corn until at least the 2014 planting season.2 Mon-santo’s dicamba-tolerant soybean also inched further toward approval in 2012, yet by year’s end no decision had been made by the agency likely

Ag Biotech Policy: 2012 in Review From GMO labeling to new problems with genetically engineered salmon, it was an eventful year. By ColIn o’neIl

Page 19: GeneWatch Vol. 26 No. 1

GeneWatch 19VoluMe 26 nuMber 1

working together will we be able to halt the march toward the further industrialization of agriculture.

nnn

Colin O’Neil is the Director of Gov-ernment Affairs at the Center for Food Safety.

surfaced during a Canadian investi-gation which found that AquaBoun-ty’s Prince Edward Island facility was contaminated in 2009 with a new strain of Infectious Salmon Ane-mia (ISA),4 the deadly fish flu that is devastating fish stocks around the world. This information was hidden from the public and potentially other Federal agencies and the FDA’s own Veterinary Medicine Advisory Com-mittee (VMAC).

Looming Battles in 2013

Much remains to be seen about the impact that Prop. 37 will have on other state labeling initiates and where continued opposition to the next generation of GE crops can be maintained. However it is clear that Members of Congress are now ready to intervene on the chemical industry’s behalf and only with the strong will of farmers, advocacy groups and

members of industry

allowable levels of GE contamination in crops and foods. The riders were widely opposed by industry includ-ing the Grocery Manufacturers’ As-sociation, the National Grain and Feed Association, the Snack Food Association and the Corn Refiners Association, as well as environmen-tal, consumer and farm groups.

Neither the appropriations rider nor the Farm Bill riders were includ-ed in any final legislation.

A Fish with a Drug Problem

In keeping with holiday tradition, on December 21, 2012, FDA officials released their Draft Environmental Assessment (EA) and opened a pub-lic comment period concerning the AquAdvantage Salmon produced by AquaBounty Technologies. The GE Atlantic salmon being considered for approval under FDA’s new animal drug law was developed by artificially combining growth hormone genes from an unrelated Pacific salmon with DNA from the anti-freeze genes of an arctic eelpout. This modifica-tion causes production of growth-hormone year-round, creating a fish the company claims grows at twice the rate of conventional farmed salmon, allowing factory fish farms to further confine fish and still get high production rates.

Since the FDA first announced its approval process for GE salmon in 2010, numerous environmental, health, economic and animal safety concerns have been raised by advo-cacy groups and the scientific com-munity. A 2011 study published by Canadian scientists concluded that if GE Atlantic salmon males, like those used in the company’s facility, were to escape from captivity they could succeed in breeding and passing their genes into the wild.3 More re-cently, previously hidden documents

Page 20: GeneWatch Vol. 26 No. 1

January-March 2013 20 GeneWatch

On February 22, 1997, a team of Scottish scientists announced that the world’s first cloned mammal, a sheep cloned from an adult cell, had been born the previous July.1 The scientists cloned a ewe by inserting DNA from a single sheep cell into an egg and implanted it in a surrogate mother. Actually, cloned embryos were placed in 277 surrogate ewes and the sheep, called Dolly, was the only success. Dolly was cloned as a way to help copy sheep that had been genetically engineered to pro-duce human proteins in their milk or blood. In effect, cloning was intended to help make pharmaceutical drugs.

By 2006, it was clear that a num-ber of cattle, pig, and goat breeders were planning to introduce cloned animals and their offspring into the food market. The U.S. Food and Drug Administration drafted a risk assess-ment on the use of clones for food in 2006 and eventually approved their use for food in 2008. However, the FDA risk assessment relied on minimal research for their approval of the meat and milk of clones for food.2 The FDA assessment found no peer-reviewed studies on meat from cloned cows or on milk or meat from their offspring. No peer-reviewed studies were found on meat and milk from cloned goats or their offspring either, nor for pigs. The three peer re-viewed studies on milk from cloned cows showed marked differences in milk from clones that should have prompted further research. None-theless, based on submissions of

data from two cloning companies, the FDA approved food and milk from cloned cattle, pigs, and goats as safe for human consumption. Ironi-cally, while the first mammal cloned was a sheep, the FDA recommended against approving meat or milk from sheep due to lack of data.

After the approval of meat and milk from cloned cattle, goats, and pigs, the U.S. Department of Agri-culture asked cloners to voluntarily keep such products off the market. The U.S. Congress requested a study of the economic effects of cloning on livestock producers, but USDA has yet to release the report. Our re-search at the Center for Food Safety has found that at least six U.S. sell-ers of bull semen for the artificial in-semination of cattle are advertising that they have cloned bull semen for sale. While there is no tracking of the number of cloned bull semen straws that have been sold, we assume that a small portion of the cattle sold in the U.S. are the offspring of clones. While cloning is sold as a way to im-prove cattle and pig genetics, most of the U.S. pedigrees now require the cloning status of an animal to be list-ed in its pedigree. The three breed as-sociations with the most cloned ani-mals—Angus, Hereford, and Texas Longhorn—all require cloning status to be listed in the pedigree. In Europe and Canada, most breeding associa-tions simply prohibit the listing of a cloned animal in the pedigree of the breed.3

In the Bullpen: Livestock Cloning With the backing of a few deep-pocketed investors, commercial livestock cloning is no longer a thing of the distant future. By Jaydee hanSon

Page 21: GeneWatch Vol. 26 No. 1

GeneWatch 21VoluMe 26 nuMber 1

is working on improving the efficien-cy of the cloning process.6

Regulating animal cloning for food

Only the United States has explic-itly approved animal clones for food. The European Union has had a sig-nificant discussion about whether food from clones and their offspring should be approved. The Agriculture Ministers of most EU countries have called for approving meat and milk from clones and their offspring.7 The European Parliament, on the other hand, has called for an outreach ban of clones and their offspring in food and forced a stop to efforts by the European Commission to approve clones for food. The Health and Consumers agency of the European Commission conducted a public comment period last fall and is ex-pected to issue a report soon.8 That report will likely engender another international discussion of animal cloning for food.

Who is doing the cloning?

Most of the cloned animals pro-duced in or for the U.S. market are cloned by two companies: Viagen, a Texas company owned by billionaire John Sperling,4 and Cyagra, former-ly a U.S. company but now owned by Argentina’s richest man, Grego-rio Perez Companc.5 Viagen claims that it has produced a total of 1,000 clones, mostly cows but also racing horses and pigs. Japanese company Kirin Pharmaceuticals now owns an-other U.S. cloning and animal genetic engineering company, Hematech. Its scientific officer, James Robl, told me that while the company has an appli-cation before the FDA for approval of its cloned and genetically engineered “mad cow” resistant cow, it plans to use that animal only to produce pharmaceutical drugs, not for food. The next generation of clones could come from wealthy Chinese corpora-tions. The Beijing Genome Institute, a powerhouse in DNA sequencing, has set up a pig cloning company that

GMO Labeling: What’s Next?

Last fall, California’s GMO Labeling ballot initiative (Prop 37) fell short by a 6% margin, having been ahead in the polls until biotech companies began pouring millions of dollars into stopping the initiative. The is-sue is still very much alive, though: Upwards of 70%1 of processed foods are estimated to contain ingredients from genetically modified crops, and groups across the country are gearing up for new efforts to require foods containing genetically modified ingredients to be la-beled as such.

The grassroots campaigns in 37 states plus Can-ada have formed The Coalition of States for GMO Labeling in an effort to coordinate state

Conclusion

Comparatively few cloned animals or their offspring have entered the U.S. or European Union markets. It is likely that imports of meat from Ar-gentina or China will be the source of cloned meat products in the future. The U.S. should take steps to be able to track clones and their offspring. At the very least, the National Organic Program should implement the 2007 recommendation of the National Or-ganic Standard Board that clones and their offspring should be excluded from USDA certified organic prod-ucts, so that consumers who want to avoid clones and their offspring in meat and milk products would have a viable option. nnn

Jaydee Hanson is Senior Policy Analyst at the Center for Food Safety.

efforts to pass mandatory GMO labeling laws. For more information on the coalition, please contact: [email protected].

To see what’s happening in your state, vis-it: http://gefoodlabels.org/gmo-labeling/state-and-local-labeling-initiatives.

If you want to let people know your state’s grassroots efforts to enact GMO labeling—or even your struggles, as it may be—email [email protected] (men-tion GeneWatch in the subject line). Compiled with contributions from Tara Cook-Littman.1. http://truefoodnow.org/campaigns/

genetically-engineered-foods/

Page 22: GeneWatch Vol. 26 No. 1

January-March 2013 22 GeneWatch

South Asia, one of the most affected regions in terms of crop production, could decline by 14.3 to 14.5 percent by 2050, maize production by 8.8-18.5 percent and wheat production by 43.7 to 48.8 percent, relative to 2000 levels.2 As such, unchecked cli-mate change will have major negative effects on agricultural productivity, with yield declines and price increas-es for the world’s staples.

The number of people at risk of hunger will therefore increase. More-over, the impacts of climate change will fall disproportionately on devel-oping countries, although they con-tributed least to the causes. The ma-jority of the world’s rural poor who live in areas that are resource-poor, highly heterogeneous and risk-prone will be hardest hit. Smallholder and subsistence farmers, pastoralists and artisanal fisherfolk will suffer com-plex, localized impacts of climate change. For these vulnerable groups, even minor changes in climate can have disastrous impacts on their livelihoods.

No wonder then that the world is desperately seeking solutions. Genet-ically modified organisms (GMOs) are one of the proposed options, for example through the development of drought-tolerant GM crops. There has been rather a lot of hype about these new GM crops, but closer

examination reveals constraints. From the limited data supplied by Monsanto to the U.S. Department of Agriculture, its drought-tolerant corn (recently deregulated in the U.S.) only provides approximately six percent reduction in yield loss in times of moderate drought.3

Drought is a complex challenge, varying in severity and timing, and other factors such as soil quality af-fect the ability of crops to withstand drought. These complications make it unlikely that any single approach or gene used to make a GM crop will be useful in all—or even most—types of drought. Furthermore, genetic en-gineering’s applicability for drought tolerance is limited insofar as it can only manipulate a few genes at a time, while many genes control drought tolerance in plants, raising questions as to whether the technology is fit for this purpose.

In contrast, conventional breed-ing has increased drought tolerance in U.S. corn by an estimated one per-cent per year over the past several decades. According to the Union of Concerned Scientists,

… that means traditional meth-ods of improving drought tolerance may have been two to three times as effective as genetic engineering, considering the 10 to 15 years typi-cally required to produce a geneti-cally engineered crop. If traditional

Climate change endangers the livelihoods and food security of the planet’s poor and vulnerable, largely because it threatens to disturb agri-cultural production in many parts of the world. The Intergovernmen-tal Panel on Climate Change (IPCC) projects that crop productivity would actually increase slightly at mid- to high latitudes for local mean temper-ature increases of up to 1-3o Celsius, depending on the crop. However, at lower latitudes, especially in season-ally dry and tropical regions, crop productivity is projected to decrease for even small local temperature in-creases (1-2oC). In some African countries, yields from rain-fed agri-culture, which is important for the poorest farmers, could be reduced by up to 50 percent by 2020. Further warming above 3oC would have in-creasingly negative impacts in all regions.

Recent studies suggest the IPCC may have significantly understated the potential impacts of climate change on agriculture. New research suggests that production losses across Africa in 2050 (consistent with global warming of around 1.5oC) are likely to be in the range of 18 to 22 per-cent for maize, sorghum, millet and groundnut, with worst-case losses of 27 to 32 percent.1 Other research suggests that rice production in

Agricultural Technologies for a Warming World GMOs have been presented as a silver bullet to shield agriculture from the effects of climate change, but ecoagricultural systems hold far more promise and far less risk. By lIm lI ChIng

Imag

e: N

ew E

ntry

Sus

tain

able

Far

min

g Pr

ojec

t

Page 23: GeneWatch Vol. 26 No. 1

GeneWatch 23VoluMe 26 nuMber 1

climate change, pest and diseases and encourages the use of traditional and locally-adapted drought and heat-tolerant varieties and species.

There is also increasing evidence that ecological agriculture can in-crease yields where they matter most—in small farmers’ fields—with low-cost, readily adoptable and ac-cessible technologies that build on farmers’ knowledge. A review of 286 ecological agriculture projects in 57 countries showed a 116 percent in-crease in yields for African projects and a 128 percent increase for East Africa.6 During times of drought, scientific side-by-side comparisons at the Rodale Institute, USA, have demonstrated that organic yields are higher than both conventional and GM agriculture.7

While there is great potential in ecological agriculture, there has been little attention to it in terms of re-search, investment, training and poli-cy focus. The challenge is to re-orient agriculture policies and significantly increase funding to support climate-resilient ecological agricultural tech-nologies. Research and development efforts should be refocused towards ecological agriculture in the context of climate change, while at the same time strengthening existing farmer knowledge and innovation.

In conclusion, a comparison of genetic engineering with other tech-nologies, such as conventional breed-ing and ecological agriculture, shows that the latter are more effective than the former at meeting the climate challenge, and at lower cost. An ex-cessive focus on genetic engineering at the expense of other approaches is a risky strategy. nnn

Lim Li Ching, M.Phil., works in the bio-safety and sustainable agriculture pro-grams at Third World Network.

world needs to move away from con-ventional, energy- and input-intensive agriculture, which has been the domi-nant model to date. Thus, the call has been for a serious transition towards sustainable/ecological agriculture. The International Assessment on Ag-ricultural Knowledge, Science and Technology for Development (IAAS-TD),5 stressed this in an extraordi-narily comprehensive assessment of the global state of agriculture, involv-ing more than 400 scientists.

The ecological model of agricul-tural production, which is based on principles that create healthy soils and cultivate biological diversity and which prioritizes farmers and tradi-tional knowledge, is climate-resilient as well as productive. Ecological ag-riculture practices and technologies are the bases for the adaptation ef-forts so urgently needed by develop-ing-country farmers, who will suffer disproportionately from the effects of climate change. Many answers al-ready exist in farmers’ knowledge of their region and their own land—for example, how to create healthy soils that store more water under drought conditions or how to grow a diversity of crops to create the resilience need-ed to face increased unpredictability in weather patterns.

Ecological agriculture practices improve and sustain soil quality and fertility, enhance agricultural biodi-versity and emphasize water man-agement and harvesting techniques. Practices such as using compost, green manures, cover crops, mulch-ing and crop rotation increase soil fertility and organic matter, which reduce negative effects of drought, enhance soil water-holding capac-ity and increase water infiltration capacity, providing resilience under unpredictable conditions. Moreover, cultivating a high degree of diversity allows farmers to respond better to

approaches have improved corn’s drought tolerance by just 0.3 percent to 0.4 percent per year, they have pro-vided as much extra drought protec-tion as Monsanto’s GE corn over the period required to develop it.4

While water availability during times of drought is also an impor-tant issue, there is little evidence that genetic engineering can help crops use water more efficiently, i.e. to use less water to achieve normal yields. Drought-tolerant crops typically do not require less water to produce a normal amount of food or fiber, and Monsanto has not supplied any data measuring water use by its drought-tolerant corn to suggest that it has also improved water use efficiency.

There are, moreover, biosafety is-sues with GMOs, and they have po-tential environmental, health and so-cio-economic risks. That is why there is an international law regulating GMOs: the Cartagena Protocol on Biosafety, ratified by 164 countries. Parties to the Cartagena Protocol have obligations to ensure that the risks are robustly assessed. There are also obligations in terms of risk man-agement, monitoring, addressing il-legal and unintentional transbound-ary movements and public awareness and participation. Therefore, any de-cision to approve or release a GMO has to be weighed seriously and deci-sion-makers should consider the full range of options available.

With regard to climate change and its implications for poor farmers, a key question to ask then is whether the proposed option can meet the needs of small farmers with the least cost/most benefit, and lowest risks. What option can best contribute to resilience to deal with unpredictable climatic options? And given that the climate change challenge is so urgent, what can deliver results quickly?

The emerging consensus is that the

Page 24: GeneWatch Vol. 26 No. 1

January-March 2013 24 GeneWatch

applications proposed. Given con-temporaneous findings in molecu-lar genetics, such as the recognition that a mutation in a single gene could promote a cell’s transformation to cancerous state,1 it was unsurprising

that concerns were raised about the capability of the transgenic methods to dramatically change the biochem-istry or ecological stability of plants. Some critics suggested that the qual-ity and safety of fruits and vegetables could be impaired, making them allergenic or toxic to humans and nonhumans who consume them, or that “superweeds” might be created which could disrupt wild or farmed ecosystems.

By 2005, however, when more than 90 percent of the annual soy-bean crop and 50 percent of the corn crop in the United States had come to be genetically engineered – a trans-formation in agricultural production that took less than a decade2 – efforts at testing and regulation of geneti-cally modified (GM) foods were in-creasingly portrayed as irrational. A perusal of the summaries of recent policy articles on the PubMed data-base turns up dozens in which reser-vations about the massive introduc-tion of GM food into the food chain are represented as scientifically igno-rant, economically suicidal, and cruel to the world’s hungry. One abstract in the journal Nature reads: “Unjus-tified and impractical legal require-ments are stopping genetically en-gineered crops from saving millions from starvation and malnutrition.”3

These papers—many by Euro-pean commentators decrying the successful efforts to keep GM foods out of the markets there, and some by U.S. commentators bemoaning

When scientists first learned in the late 1970s how to sequence DNA and transfer it from one kind of or-ganism to another, improving foods and other crop plants by introduc-ing foreign genes was among the first

The State of the Science Most of the evidence for the safety of genetically modified food crops comes from studies that only look at short-term effects … and from information provided by the same companies who sell those seeds. By Stuart a. neWman

Illustration by Boris Artzybasheff for Fortune magazine, June 1943.

Page 25: GeneWatch Vol. 26 No. 1

GeneWatch 25VoluMe 26 nuMber 1

a skeptical public, the biotech food industry has depended on compliant regulators,10 on its proponents’ ridicule of biotech industry critics’ supposed scientific ignorance,11,12 and on expensive campaigns against labeling of prepared foods that would draw undue attention to the presence of GM components, which they claim to be natural and ordinary.13 (These are the same components that when presented to the Patent Office and potential investors are portrayed as novel and unique.) A food crop that actually benefited the people who eat it rather than only those who sell it would likely open the floodgates of greatly weakened regulation. Golden Rice, designed to provide Vitamin A to malnourished children, has failed to overcome the hurdles for approval for dietary use since it was first described in 2000. Though very limited in its ability to alleviate malnutrition, it has some merit in the prevention of blindness, and seems poised for approval in the next year or so.14 If so, it will almost certainly help agribusiness tighten its grip on the world food supply and increase its capacity to foist products that are much more questionable on their captive clientele—that is, everyone.

nnn

Stuart Newman, Ph.D., is Professor of Cell Biology and Anatomy at New York Medical College and a founding member of the Council for Responsible Genetics.

have not indicated any risk to human health. In spite of this clear state-ment, it is quite amazing to note that the review articles published in inter-national scientific journals during the current decade did not find, or the number was particularly small, refer-ences concerning human and animal toxicological/health risks studies on GM foods.7

The same group revisited the lit-erature four years later, reporting that whereas the number of citations found in databases had dramatically increased in the intervening period, new information on products such as potatoes, cucumber, peas or toma-toes, among others was not available. Regarding corn, rice, and soybeans, there was a balance in the number of studies suggesting that GM corn and soybeans are as safe and nutritious as the respective conventional non-GM plant, and those raising still serious concerns. They also note that “most of these studies have been conducted by biotechnology companies respon-sible [for] commercializing these GM plants.”8

Given the uncertainties of the long-term health impact of GM foods, it is significant that so far, vir-tually all genetic modification of food and fiber crops has focused on the economic aspects of production (i.e., making crops resistant to herbicides and insect damage, increasing trans-portability and shelf-life) rather than the more elusive goals of improving nutrition or flavor. Introducing bio-logical qualities that enhance pro-duction, transportability and shelf life can compromise palatability, as seen with the Flavr Savr tomato, the first GM crop to be approved by the FDA for human consumption, two decades ago.9

To protect its investment against

the necessity to test these products at all—mainly support their cases by referencing short-term feeding stud-ies of animals. But this type of study is not adequate to allay valid concerns. One group, reviewing the relevant areas, has written, “It appears that there are no adverse effects of GM crops on many species of animals in acute and short-term feeding studies, but serious debates of effects of long-term and multigenerational feeding studies remain.”4

According to another group that has looked into these issues:

The most detailed regulatory tests on the GMOs are three-month long feeding trials of laboratory rats, which are biochemically assessed…The test data and the corresponding results are kept in secret by the com-panies. Our previous analyses…of three GM maize [varieties] led us to conclude that [liver and kidney] tox-icities were possible, and that longer testing was necessary.5

Another team actually performed such long-term studies, with the find-ings that mice that were fed for five consecutive generations with trans-genic grain resistant to a herbicide showed enlarged lymph nodes and increased white blood cells, a signifi-cant decrease in the percentage of T lymphocytes in the spleen and lymph nodes and of B lymphocytes in lymph nodes and blood in comparison to control fed for the same number of generations with conventional grain.6

A central issue for crop foods, of course, is their effects on humans. The most comprehensive review of this subject as of 2007 stated:

…the genetically modified (GM) products that are currently on the international market have all passed risk assessments conducted by na-tional authorities. These assessments

Page 26: GeneWatch Vol. 26 No. 1

January-March 2013 26 GeneWatch

found to be 1.3 to 2.3 times greater with large palpable tumors occurring 4 times more than controls.

The paper was met with a firestorm of reaction. Some scientists and regu-latory bodies found the study incon-clusive, citing methodological flaws or limitations in the study design or statistical analysis, and recommend-ed that the study be repeated. Oth-ers dismissed the study outright as biased and requested that the journal withdraw it. However, over a hundred scientists from universities and insti-tutes throughout the world signed on to an open letter supporting Séralini et al. against what they viewed as cor-porate influence over the science of GM crops.

Many media outlets dismissed the study without even waiting for the paper to be fully aired in the scien-tific community. Faced with an un-precedented reaction to their journal publication, the editors of Food and Chemical Toxicology wrote: “The edi-tors and publishers wish to make clear that the normal thorough peer review process was applied to the Séralini

et al. paper. The paper was published after being objectively and anony-mously peer reviewed with a series of revisions made by the authors and the corrected paper then accepted by the editor.”

Séralini et al. issued an 8-page re-sponse to critics where they provided a table of criticisms and answers.2

Professor Séralini sent GeneWatch an announcement in which his re-search group identifies the most toxic chemical in Ready Roundup—the most widely used herbicide in the world--which is not the active ingredi-ent glyphosate but a substance called POE-15. This substance is an adju-vant added to glyphosate, which the authors state is toxic to human cells. Adjuvant chemicals often escape the rigorous testing of active ingredients in pesticides. Another example of an adjuvant in synthetic pyrethroids is piperonyl butoxide, which is a poten-tial carcinogen.

Sheldon KrimskyChair, Board of DirectorsCouncil for Responsible Genetics

Background: Séralini et. al

Gilles Eric Séralini is a professor of molecular biology at Caen Univer-sity, located in the town of Caen in Normandy, France. Professor Séralini was the lead author among a team of 8 scientists who submitted a paper to the peer-reviewed journal Food and Chemical Toxicology on the long term toxicity of Roundup herbicide and Roundup-tolerant genetically modi-fied maize. The paper was received by the journal on April 11, 2012, sent out for review, accepted for publication on August 2, 2012, available online September 19, 2012 and appeared in print in the Elsevier journal Novem-ber 2012.1

Séralini et al. exposed rats to GM maize and Glyphosate and studied them for 2 years. They found that female rats died at a rate 2-3 times greater than controls. Female rats de-veloped large mammary tumors more often and earlier in life than the con-trol groups. In treated male rats liver congestion and necrosis were ob-served 2.5 to 5.5 more frequently than controls; severe kidney disease was

Glypho-gate Last fall, a paper finding health problems in rats fed Roundup herbicide and genetically modified Roundup-resistant corn brought out strong reactions from proponents and opponents of GM crops. Now the authors are presenting new findings on the most widely used herbicide in the world.

WIth gIlleS-erIC SéralInI, roBIn meSnage, and Benoît Bernay

January-March 2013 26 GeneWatch

Page 27: GeneWatch Vol. 26 No. 1

GeneWatch 27VoluMe 26 nuMber 1

cannot be carried out. These assess-ments are therefore neither neutral nor independent. They should, as a first step, make public on the Internet all the data that underpin the com-mercial release and positive opinions on the use of Roundup and similar products. The industry toxicological data must be legally made public.

Adjuvants of the POE-15 family (polyethoxylated tallowamine) have now been revealed as actively toxic to human cells, and must be regulated as such. The complete formulations must be tested in long-term toxicity studies and the results taken into ac-count in regulatory assessments. The regulatory authorization process for pesticides released into the environ-ment and sold in stores must be re-vised. Moreover, since the toxic con-fidential adjuvants are in general use in pesticide formulations, we fear ac-cording to these discoveries that the toxicity of all pesticides has been very significantly underestimated.

The full study: Mesnage R., Bernay B., Séralini

G-E. (2013, in press). Ethoxylated adjuvants of glyphosate-based herbi-cides are active principles of human cell toxicity. Toxicology. http://dx.doi.org/10.1016/j.tox.2012.09.006

This study was conducted in the Uni-versity of Caen with the structural support of CRIIGEN in the European Network of Scientists for Social and Environmental Responsibility. www.criigen.org | www.ensser.org

The health and environmental agen-cies and pesticide companies assess the long-term effects on mammals of glyphosate alone, and not the full for-mulation. The details of this regula-tory assessment are kept confidential by companies like Monsanto and by health and environmental agencies.

This study demonstrates that all the glyphosate-based herbicides test-ed are more toxic than glyphosate alone, and explains why. Thus their regulatory assessments and the maxi-mum residue levels authorized in the environment, food, and feed, are er-roneous. A drink (such as tap water contaminated by Roundup residues) or a food made with a Roundup toler-ant GMO (like a transgenic soybean or corn) were already demonstrated as toxic in the recent rat feeding study1 from Prof. Séralini’s team. The researchers have also published re-sponses to critics of the study.2 This new research explains and confirms the scientific results of the rat feeding study.

Overall, it is a great matter of con-cern for public health. First, all au-thorizations of Roundup-type herbi-cides have to be questioned urgently. Second, the regulatory assessment rules have to be fully revised. They should be analyzed in a transparent and contradictory manner by the sci-entific community. Agencies that give opinions to government authorities, in common with the pesticide com-panies, generally conclude safety. The agencies’ opinions are wrong because they are made on the basis of lax as-sessments and much of the industry data is kept confidential, meaning that a full and transparent assessment

In a new study published in the scientific journal Toxicology, Robin Mesnage, Benoît Bernay and Profes-sor Gilles-Eric Séralini, from the Uni-versity of Caen, France, have proven from a study of nine Roundup-like herbicides that their most toxic com-pound is not glyphosate—the sub-stance the most assessed by regula-tory authorities—but a compound that is not always listed on the label, called POE-15. Modern methods were applied at the cellular level (on three human cell lines), and mass spectrometry (studies on the nature of molecules). This allowed the re-searchers to identify and analyze the effects of these compounds.

Glyphosate is supposed to be the “active ingredient” of Roundup, the most widely used herbicide in the world, and it is present in a large group of Roundup-like herbicides. It has been safety tested on mammals for the purposes of regulatory risk as-sessment. But the commercial formu-lations of these pesticides as they are sold and used contain added ingre-dients, or adjuvants. These are often classified confidential and described as “inerts.” However, they help to sta-bilize the chemical compound glypho-sate and help it to penetrate plants, in the manner of corrosive detergents. The formulated herbicides (including Roundup) can affect all living cells, especially human cells. This danger is overlooked because glyphosate and Roundup are treated as the same by industry and regulators on long-term studies. The supposed non-toxicity of glyphosate serves as a basis for the commercial release of Roundup.

Study: Glyhposate Not the Most Toxic Chemical in Roundup

GeneWatch 27VoluMe 26 nuMber 1

Page 28: GeneWatch Vol. 26 No. 1

January-March 2013 28 GeneWatch

Biotechnology is nothing new; hu-mans have been manipulating other species’ genetics to serve our needs and desires for thousands of years. Tomatoes, potatoes, corn, beer, poodles—none would exist as they do today without the deliberate, me-thodical, and at times questionable (poodles?), intervention of humans.

However, there are several impor-tant distinctions between the more recent techniques commonly re-ferred to as “genetic engineering” or “genetic modification” and the tech-niques we employed in the several millennia before it was possible to combine the DNA of a flounder and a tomato plant. To collapse the dis-tinction between these two branches of biotechnology is to muddy the de-bate over the possible ramifications of genetic engineering.

It’s important to note that the term “genetic modification” is used in in-ternational laws to refer to the use of recombinant DNA techniques to transfer genetic material between organisms in a way that would not take place naturally. The term is also sometimes used incorrectly to re-fer to marker-assisted selection, in which genes are simply identified (tagged) and then manipulated via more traditional breeding methods.

The most immediate difference between traditional breeding and ge-netic engineering is that with tradi-tional breeding, we are working with sex cells. We are deliberately mixing the gametes of one individual with those of another, in hopes of bringing

out desired results of this sexual re-combination. With genetic engineer-ing, we are inserting the manually-remixed genetic material from one or more organisms into the genome of another, well outside the context of sexual reproduction as it is com-monly understood.

With traditional breeding, we are able to select for aspects of genetic potential which already existed with-in a given species, or at most, be-tween closely related species or gen-era. We are limited to working within existing contexts of genetic material. When we bring out a desired set of traits, resulting even in something as novel as a poodle, that possibility—however “unnatural” seeming—had always existed within the genetic po-tential of the species.

There are several reasons, de-scribed below, why genetic engineer-ing may cause unpredictable inter-ruptions of normal gene function. This unpredictability is demonstrat-ed most readily by the vast difference in the production of viable offspring between the two processes. In tradi-tional breeding, most of the offspring produced are viable, functional or-ganisms, although they very well may not express the desired traits. Though advances in genetic engi-neering have decreased the relative number of failures, the process still requires a large number of attempts (and failures) before viable organ-isms are produced.

In brief, genetic engineering in-volves the deliberate insertion of a

Life, the Remix They’re both “biotechnology,” but genetic engineering is fundamentally different from traditional breeding. By martIn dagoBerto

28 GeneWatch

Page 29: GeneWatch Vol. 26 No. 1

GeneWatch 29VoluMe 26 nuMber 1

genetic sequences in a relatively con-trolled fashion, as compared to the more unpredictable gene insertion carried out with genetic engineering.

One final major difference worth noting in this brief review is the prevalent use in genetic engineering of marker genes which code for anti-biotic resistance, employed to screen cells for expression of the transgene. This technique introduces risks asso-ciated with the creation of antibiotic resistant bacteria which do not exist with traditional breeding.

With genetic engineering, we are now capable of re-mixing genes from every kingdom of life and expressing them in ways that could never have possibly existed before. Nothing re-motely similar to this could ever oc-cur with the use of traditional breed-ing. As the commercial prevalence of genetic engineering increases and for the sake of meaningful public dis-course about the possible health and environmental impacts, it is critical to maintain the distinction between these two branches of biotechnology.

nnn

Martin Dagoberto is Co-Founder and Network Facilitator of Massachusetts Right to Know GMOs, a state-wide net-work of safe food advocates supporting mandatory labeling of Genetically Modi-fied foods.

isolated units. They are connected to an extraordinarily complex whole. Unfortunately, we do not yet under-stand how all the pieces can fit and move together. A single change to the DNA can cause widespread changes in the organism which we cannot predict. Nor can we adequately pre-dict how any given “GM” organism or sequence will interact, mutate or persist within a complex living eco-system, once it is released from the laboratory environment.

The processes currently used to create commercial GM crops are not able to target where exactly the gene is inserted, nor can they ensure stable expression. GM sequences (or random fragments of them) can be randomly inserted into the middle of an existing gene or otherwise dis-rupt or alter normal gene regulation. The process of tissue culture itself can also be highly mutagenic. Un-intended modifications due to such mutations (for example, the produc-tion of a new allergen or change in nutritional value) may not be detect-ed or selected against by the genetic engineer before the GMO is put into commercial production and the pub-lic food supply.

With traditional breeding, the ex-change of genetic material occurs be-tween two organisms with a relative-ly similar recent evolutionary history. In this case, compatible mechanisms have evolved which are able to shuffle

foreign sequence of DNA into the genetic makeup and expression of an existing organism. One way to force a novel DNA sequence into a target genome is to coat it onto gold pellets and shoot it at a culture of host cells with a “gene gun.” Another way is to perforate the cell membranes with chemicals or electricity, or to use bac-teria to carry the sequence and infect the cells. In order to move, integrate and express the desired gene, the inserted DNA sequence must often include genetic segments modified from viruses and bacteria. The cells which survive the insertion process are screened for the expression of the desired sequences and grown out in an artificial culture using hormones. Those tissues/organisms which ap-pear to function properly are then tested for commercial application. As compared to traditional methods of cross-pollination or animal pair-ing, genetic engineering is clearly a distinct process.

Further, common processes of gene insertion are still relatively crude, in that they inherently cause unpredictable mutations in the DNA of the host cells. This collateral dam-age can alter the functioning of the natural genes of the organism in ran-dom and potentially harmful ways. These processes were developed us-ing a dated perception of the genetic landscape. It is now becoming clear that genes cannot be considered

Page 30: GeneWatch Vol. 26 No. 1

January-March 2013 30 GeneWatch

for Warfarin metabolism, even when specific genetic mutations that influ-ence its metabolism are known.

Critical to Kahn’s argument re-garding evidence is the fact that the clinical trials on which the company based its patent application for Bi-Dil were never designed to compare racial difference in response to the drug. Using “care of the data” as an organizing theme, Kahn highlights one of the many troubling aspects of this controversy: the extraordinarily

failure, NitroMed filed a claim with the U.S. Patent and Trademark Of-fice. As a necessary condition for FDA approval, they conducted a clinical trial that enrolled only Afri-can American subjects. Not surpris-ingly, the drug was effective in Afri-can Americans, just as it had been on other groups in earlier trials. On June 23, 2005 the FDA approved BiDil for the treatment of heart failure only in African Americans. Physicians were free to prescribe the drug off label for other patients but the drug was lauded for its unique effects in Afri-can Americans. Its exorbitant price compared to the inexpensive gener-ics, however, doomed BiDil, at least for the time being.

Several overarching questions frame Kahn’s analysis: How and why did race become an object of phar-macogenomics, such that it could reasonably be used in a patent appli-cation? How specifically did race play out in the medical, legal, and regula-tory arenas? What are the broader social and political implications of racialized biomedicine? Focusing on BiDil, Kahn argues convincingly that innovations in genomics, the vagaries of U.S. patent law, powerful commercial interests, federal man-dates for inclusion, and uncritical advocacy around racial disparities in heart disease converged to create the conditions for NitroMed’s successful patent application. He then extends his analysis to biotechnology more broadly, examining how race has been invoked in diagnostic testing

In Race in a Bottle, Jonathan Kahn tracks the contentious history of Bi-Dil, the first drug targeted specifical-ly to African Americans. Ironically, race-based drug treatment emerged in the wake of the sequencing of the human genome, a project that theo-retically promised both to scientifi-cally refute the notion of genetically distinct racial groups and to usher in an era of personalized medicine. Though hyped by researchers, the FDA, and the press as an important first step toward personalized medi-cine, BiDil is a drug administered to patients based on their membership in a group.

This is a complex case that Kahn makes accessible through careful examination of how market forces shaped what counted as “evidence” in BiDil’s convoluted path to the mar-ket. As Kahn recounts, BiDil is not a “new” drug. Rather it is a combination of two generic vasodilators, long con-sidered effective in the management of heart failure—for all people, re-gardless of race. Because the new for-mulation as a single pill did not work any better than the generic drugs, it was unable to win FDA approval. NitroMed, the company that gained the intellectual rights to the combi-nation drug, got creative, drawing on race in their attempt to extend pat-ent protection. With the barest hint of evidence for a differential effect in African Americans (based on only 49 African American subjects) from one of the large but poorly designed clinical trials of treatment for heart

Book Review: Race in a Bottle Jonathan Kahn’s book tells the story of the rise, fall, and fallout of BiDil, the notorious race-based drug. By lundy Braun

Race in a Bottle: The Story of BiDil and Racialized Medicine in a Post-Genomic AgeBy Jonathan KahnColumbia University Press

Page 31: GeneWatch Vol. 26 No. 1

GeneWatch 31VoluMe 26 nuMber 1

shaping of the very questions scien-tists ask, this book will be of inter-est to scientists, clinicians on the frontline of race-based medicine, and patients. For advocates seriously invested in the social justice project of eliminating racial disparities in disease, Kahn’s book should be re-quired reading. At a time when the ties between scientific researchers and the pharmaceutical industry are becoming ever more entangled, Race in a Bottle provides valuable insights into the consequences of these con-nections for health and health care – and, importantly, for what passes as knowledge. nnn

Lundy Braun, PhD, is a Professor of Af-ricana Studies and Pathology and Labo-ratory Medicine and a member of the Science and Technology Studies Program at Brown University.

loose, if not sloppy, construction of what passed as evidence in the pat-ent application and FDA hearings. From the use of misleading statis-tics on mortality from heart failure in African Americans, to the fail-ure to define the central variable of race, to the design of a clinical trial (A-HeFT) that included only African Americans (and therefore could not determine differential efficacy) to the lack of any mechanistic understand-ing for a differential effect, Kahn shows that attention to the data was consistently problematic when it came to matters of race. The chapter on the FDA hearings is particularly illuminating.

Kahn is a widely acknowledged ex-pert in the history of BiDil. As a his-torian of science and a lawyer with expertise in patent law, Professor Kahn is ideally positioned to analyze the complex interactions between biomedicine, the law, the market, the

state, and an uncritical media in ex-ploiting race in the context of drug development. As he notes, Kahn has been both a participant in these de-bates and an analyst. Importantly, he places the case studies of BiDil and Warfarin in the larger frame of con-temporary debates over race, medi-cine, and genetics, thus providing the reader with tools for analyzing new cases as they arise. The specif-ics of each case will differ, as he il-lustrates in his comparison of BiDil to Warfarin, but the principles for understanding the problems with race-based medicine, and particular-ly the subtle forms of discrimination race-based medicine engenders, are generalizable.

Race in a Bottle is an important book for anyone interested in sort-ing through the morass of hype and biomedical evidence in the age of genomics. In probing the complex-ity of commercial interests in the

CRG is excited to announce that GeneWatch magazine has launched its new Youtube video channel: GeneWatch TV.

Each new issue of GeneWatch magazine will have a video component highlighting the key people and hot topics in its pages.

www.youtube.com/thecrgchannel2

GeneWatch Multimedia

Page 32: GeneWatch Vol. 26 No. 1

January-March 2013 32 GeneWatch

On Feb. 13, CRG Board Chair and Co-founder Professor Sheldon Krim-sky participated in a debate held by the program Intelligence Squared, which airs on NPR and PBS. The top-ic: Should we prohibit genetically en-gineered babies? Professor Krimsky argued in favor of a ban, joined by Robert Winston, professor at Impe-rial College London. Nita Farahany, professor at Duke University and a member of the Presidential Com-mission for the Study of Bioethical Issues, and Lee Silver, professor at Princeton University, argued against the ban.

As Prof. Krimsky pointed out, there are two main reasons that par-ents might consider prenatal genetic modification of reproductive cells (sperm, eggs, or zygote): for curing or preventing genetic disease, or for “enhancement of a person.”

“For genetic diseases, in the great majority of cases there are simpler, less risky, less costly, less ethically controversial, and more dependable methods” of detecting and avoiding severe genetic abnormalities through prenatal embryo diagnosis, Prof. Krimsky said. This means that oth-er than a couple of rare exceptions, such as mitochondrial disease, “the only sensible rationale for engaging in genetic modification of the fertil-ized egg is for the enhancement of a child”—selecting for (or attempting to select for) traits such as height, physical appearance, or intelligence.

“Engaging in genetic modification of human gametes, the human re-productive cells, for enhancement is where I find the greatest moral failure and the greatest scientific folly,” Prof.

Krimsky said. He and Prof. Winston outlined some of the many things that could go wrong in the course of attempting to carry out genetic mod-ification of human reproductive cells, including—since this can’t be fully tested in rats, or even chimpanzees—the prospect of human clinical trials.

Furthermore, Prof. Krimsky said, in regard to parents genetically modifying their future child, “from a biological and developmental stand-point, the so-called traits under con-sideration cannot remotely be en-hanced by the modification of a gene or two.” Traits from muscle tone to nearly anything related to human behavior are shaped by a complex array of factors, both genetic and en-vironmental. You can’t think of the human genome as “a Lego set,” Prof. Krimsky said, “where pieces of DNA can be plugged in or out without in-terfering with the other parts of the system. Actually, the human genome is more like an ecosystem where all the parts interrelate and are in mu-tual balance.”

“I am all for human enhancement, but it must start after an egg is fertil-ized,” Prof. Krimsky said, “beginning in utero by protecting the fetus from toxic chemicals and continuing post-natally through environmental, nu-tritional and cognitive enhancement and moral education. Enhancement through genetic engineering of hu-man germ plasm is a fool’s paradise and will lead to no good.”

The full debate is available online at www.intelligencesquaredus.org. nnn

Bowman v. Monsanto, in which the largest agribusiness company in the world is seeking damages from Indiana farmer Vernon Hugh Bowman for violating the corpora-tion’s patent on genetically modified Roundup Ready seeds, is not going well for Bowman.

Farmers who buy Monsanto’s Roundup Ready seeds are required to sign a contract which forbids them from saving seeds from that crop and replanting them the following year

CRG Founder Debates Human Genetic Engineering on NPR, PBS

notes from the field

TOPIC UPDATE: Gene Patents

Monsanto Takes a Farmer to Supreme Court; Guess Who’s Winning?

Page 33: GeneWatch Vol. 26 No. 1

GeneWatch 33VoluMe 26 nuMber 1

(instead of having to go back to Mon-santo for their seeds each year).

Vernon Bowman had legally pur-chased Roundup Ready seeds for several years before he hatched a plan to circumvent both having to buy the herbicide-resistant seeds from Monsanto each year, and hav-ing to sign the contract forbidding him from replanting those seeds. He bought soybeans from a local grain silo, knowing that it would include a mix of Roundup Ready and non-Roundup Ready soybeans - and that, although it was meant for livestock feed, many of the beans would still be viable as seed. Bowman planted the soybeans and sprayed the field with

image: thefoodlabelmovement.org

Thomson Reuters, 8 Mar. 2011.5. “Biofuel Crops Breeding and

Improvement.” Agradis, Web.6. National Renewable Energy Laboratory.

NREL’s Multi-Junction Solar Cells Teach Scientists How to Turn Plants into Powerhouses. N.p., 12 May 2011.

7. Synthetic Biology: 10 Key Points for Delegates. ETC Group, Oct. 2012. <http://www.etcgroup.org/sites/www.etcgroup.org/files/synbio_ETC4COP11_4web_0.pdf>.

8. Rodemeyer, Michael. New Life, Old Bottles: Regulating the First-Generation Products of Synthetic Biology. Woodrow Wilson International Center for Scholars, Synthetic Biology Project, 2009.

9. Wade, Nicholas. “Researchers Say They Created a ‘Synthetic Cell.’” The New York Times. 20 May 2010.

10. Bittman, Mark. “A Simple Fix for Farming.” New York Times, 19 Oct. 2012.

11. “Agroecology and Sustainable Development.” Pesticide Action Network North America, Apr. 2009.

12. “Principles for the Oversight of Synthetic Biology.” Friends of the Earth, Mar. 2012.

Phil Bereano, p. 16

1. http://www.ucsusa.org/food_and_ag-riculture/our-failing-food-system/genetic-engineering/failure-to-yield.html

2. http://nature.berkeley.edu/~miguel-alt/what_is_agroecology.html

3. http://www.conservation.org/newsroom/

www.landesbioscience.com/journals/gmcrops/02SappingtonGMC1-2.pdf, accessed 18 Feb 2013

6. Stutz B (2010) Companies Put Restrictions on Research into GM Crops. Yale Environmnet 360. http://e360.yale.edu/content/print.msp?id=2273, accessed 18 Feb 2013

7. Understandably, the people who told me their experiences did so in confidence.

8. Food & Water Watch (2012) Public Research, Private Gain: Corporate Influence Over University Agricultural Research. http://documents.foodandwater-watch.org/doc/PublicResearchPrivateGain.pdf, accessed 18 Feb 2013

9. Crouch ML (1991) The very structure of scientific research mitigates against devel-oping products to help the environment, the poor, and the hungry. J. Agricultural and Environmental Ethics 4:151-158

Eric Hoffman, p. 13

1. Hylton, Wils S. “Craig Venter’s Bugs Might Save the World.” New York Times 30 May 2012

2. “What Is GMO?” The Non-GMO Project, <http://www.nongmoproject.org/learn-more/what-is-gmo/>.

3. “Global Market for Synthetic Biology to Grow to $10.8 Billion by 2016.” BCC Research, Nov. 2011.

4. Fehrenbacher, Katie. “Monsanto Backs Algae Startup Sapphire Energy.”

Editor’s Note, p. 2

1. http://www.poultryhub.org/wp-content/uploads/2012/06/Ross308BroilerPerfObj2012R1.pdf; http://en.aviagen.com/assets/Tech_Center/Ross_Broiler/Ross708BroilerPerfObj2012R1.pdf

Martha Crouch, p. 8

1. 2009 Monsanto Technology/Stewardship Agreement (Limited Use License)

2. Gorman ME, Simmonds J, Ofiesh C, Smith R, and Werhane PH (2001) Monsanto and Intellectual Property, In “Teaching Ethics, Fall 2001”, University of Virginia Darden School Foundation, Charlottesville, VA. http://www.uvu.edu/ethics/seac/Monsanto%20and%20Intellectual%20Property.pdf; accessed 18 Feb 2013.

3. Center for Food Safety & Save Our Seeds (2013) Gene Giants vs. U.S. Farmers. http://www.centerforfoodsafety.org/wp-content/uploads/2013/02/Seed-Giants_final.pdf; accessed 18 Feb 2013

4. Waltz E (2009) Under wraps. Nature Biotechnology 27 (10): 880 – 882. http://www.emilywaltz.com/Biotech_crop_research_restrictions_Oct_2009.pdf, accessed 18 Feb 2013

5. Sappington TW, Ostlie KR, DiFonzo C, Hibbard BE, Kurpke CH, Porter P, Pueppke S, Shields EJ and Tollefson JJ (2010) Conducting public-sector re-search on commercialized transgenic seed. GM Crops 1 (2): 55 – 58. http://

Endnotes

Roundup; the plants that survived were the Roundup Ready ones, and he saved and replanted those seeds for several years.

But in 2007, Monsanto found out and sued Bowman for patent infringement. The Indiana Federal Court and the United States Court of Appeals for the Federal Circuit both ruled that Bowman owes Mon-santo over $84,000. Now the case has reached the Supreme Court, but after the Court heard arguments from both sides (but especially from Monsanto’s lawyer, who had signifi-cantly more uninterrupted speaking time), the verdict doesn’t look likely to change.

Bowman’s lawyer, Mark Walters, challenged not just Monsanto’s case against Bowman but the patents themselves. The Supreme Court - even the more liberal justices, such as Justice Sonia Sotamayor - did not appear convinced.

“He can plant and harvest and eat or sell,” Justice Antonin Scalia said, echoing the sentiments of Chief Justice John Roberts. “He just can’t plant, harvest, and then replant.”

“We disagree that the activ-ity of basic farming could be consid-ered making the invention,” Walter responded.

The Court is expected to hand down its ruling by June. nnn

Page 34: GeneWatch Vol. 26 No. 1

January-March 2013 34 GeneWatch

4. Zhang, W. & F. Shi. 2010. Do geneti-cally modified crops affect animal re-production? A review of the ongoing debate. Animal. 5: 1048-1059.

5. de Vendomois, J. S., et al. 2010. Debate on GMOs health risks after statistical findings in regulatory tests. Int J Biol Sci. 6: 590-598.

6. Krzyzowska, M., et al. 2010. The effect of multigenerational diet containing geneti-cally modified triticale on immune system in mice. Pol J Vet Sci. 13: 423-430.

7. Domingo, J. L. 2007. Toxicity stud-ies of genetically modified plants: a review of the published literature. Crit Rev Food Sci Nutr. 47: 721-733.

8. Domingo, J. L. & J. Gine Bordonaba. 2011. A literature review on the safety assessment of genetically modified plants. Environ Int. 37: 734-742.

9. Redenbaugh, K. 1992. Safety assessment of genetically engineered fruits and veg-etables: a case study of the FLAVR SAVR tomato. CRC Press. Boca Raton, Fla.

10. Newman, S. A. 2009. Genetically modified foods and the attack on nature. Capitalism Nature Socialism. 20: 22-31.

11. Silver, L. M. 2006. Why GM Is good for us: genetically modified foods may be greener than organic ones. In Newsweek International, March 20: 57-58. http://128.112.44.57/CNmedia/articles/06newsweekpig1s1.pdf

12. Shermer, M. 2013. The liberals’ war on science. ScientficAmerican.com, January 21. http://www.scientificamerican.com/article.cfm?id=the-liberals-war-on-science

13. Vaughan, A. 2012. Prop 37: Californian voters reject GM food labelling. Guardian.co.uk, November 7. http://www.guard-ian.co.uk/environment/2012/nov/07/prop-37-californian-gm-labelling

14. Haskell, M. J. 2012. The challenge to reach nutritional adequacy for vita-min A: beta-carotene bioavailability and conversion--evidence in humans. Am J Clin Nutr. 96: 1193S-1203S.

Seralini et. al, p. 26

1. Gilles-Eric Seralini, Emile Clair, Robin Mesnage, Steeve Gress, Nicolas Defarge, Manuela Malatesta, Didier Hennequin, Joёl Spiroux de Vendômors. Longterm toxicity of a Roundup her-bicide and a Roundup-tolerant geneti-cally modified maize. Food and Chemical Toxicology 50(2012):4221-4231.

2. Gilles-Eric Seralini, et. al. Answers to critics: Why there is a long term toxicity due to a Roundup-tolerant genetically modified maize and to a Roundup herbicide. Food and Chemical Toxicology 53(2013):476-483.

html#ixzz2LqRAcC3B6. See BGI Ark Biotechnology Co. LTD

Shenzen (BAB) http://www.bab-genomics.com/list.aspx?catid=168 and Christine Larson, Inside China’s Genome Factory, MIT Technology Review, Feb. 11, 2013 available at http://www.technolo-gyreview.com/featuredstory/511051/inside-chinas-genome-factory/

7. See http://www.dw.de/eu-ministers-approve-sale-of-food-from-cloned-animals-offspring/a-4414990

8. See Director General, SANCO, “Measures on animal cloning for food production in the EU” available at http://ec.europa.eu/dgs/health_consumer/dgs_consultations/animal_cloning_consultation_en.htm

Lim Li Ching, p. 22

1. Schlenker, W. and D.B. Lobell (2010). Robust negative impacts of cli-mate change on African agriculture. Environmental Research Letters, 5, doi:10.1088/1748-9326/5/1/014010.

2. Nelson, G.C., M.W. Rosegrant, J. Koo, R. Robertson, T. Sulser, T. Zhu, C. Ringler, S. Msangi, A. Palazzo, M. Batka, M. Magalhaes, R. Valmonte-Santos, M. Ewing and D. Lee (2009). Climate Change: Impact on Agriculture and Costs of Adaptation. IFPRI, Washington, DC.

3. Gurian-Sherman, D. (2012). High and dry: Why genetic engineering is not solving agriculture’s drought problem in a thirsty world. Union of Concerned Scientists, Cambridge, MA. Available at: http://www.ucsusa.org/assets/documents/food_and_agriculture/high-and-dry-report.pdf

4. Ibid, p.3.5. IAASTD (2009). Agriculture at a

Crossroads. International Assessment of Agricultural Knowledge, Science and Technology for Development. Island Press, Washington, DC. http://www.agassessment.org

6. UNEP-UNCTAD Capacity-building Task Force on Trade, Environment and Development (2008). Organic Agriculture and Food Security in Africa. United Nations, New York and Geneva.

7. http://www.rodaleinstitute.org/fst30years/yields

Stuart Newman, p. 24

1. Weinstein, I. B., et al. 1984. Cellular targets and host genes in multistage carcinogenesis. Fed Proc. 43: 2287-2294.

2. Hsieh-Li, H. M., et al. 1995. Hoxa 11 structure, extensive antisense transcrip-tion, and function in male and female fertility. Development. 121: 1373-1385.

3. Potrykus, I. 2010. Regulation must be revolutionized. Nature. 466: 561.

pressreleases/Pages/Global_Tool_to_Gauge_Earths_and_Humanitys_Vital_Signs_Launches_in_Africa.aspx

Colin O’Neil, p. 18

1. For more information, see the Center for Food Safety’s Food Safety Review “Going Backwards: Dow’s 2,4-D-Resistant Crops and a More Toxic Future.” Winter 2012. http://www.centerforfoodsafety.org/wp-content/uploads/2012/02/FSR_24-D.pdf

2. Gillam, Carey. “Dow’s controversial new GMO corn delayed amid protests.” Reuters. January 18, 2012. Available online at http://www.reuters.com/article/2013/01/18/dow-biotech-idUSL1E9CIBN320130118

3. Darek T. R. Moreau, Corinne Conway, Ian A. Fleming. (2011) “Reproductive per-formance of alternative male phenotypes of growth hormone transgenic Atlantic salmon (Salmo salar).” Evolutionary Applications, Blackwell Publishing, Ltd.

4. Living Oceans Society, Media Release. “ISA virus confirmed in AquaBounty’s genetically-engineered salmon.” Reposted on December 20, 2012. Available at: http://www.livingoceans.org/media/releases/salmon-farming/isa-virus-confirmed-aquabounty’s-genetically-engin

Jaydee Hanson, p. 20

1. John Gurdon, won the Nobel Prize for Medicine for his work developing the techniques now used for cloning for his work in frogs in 1962. The Scottish team’s achievement was figuring out how to use this technique, previously successful only in amphibians and fish, in mammals.

2. See Center For Food Safety, “Not Ready for Prime Time: FDA’s Flawed Approach to Assessing the Safety of Food from Animal Clones,” March 2007, available at: http://www.centerforfoodsafety.org/pubs/FINAL_FORMATTEDprime%20time.pdf

3. See Jaydee Hanson, Comments to the US Department of Agriculture, National Organic Program on tracking animal clones using pedigrees, September 20, 2011, pgs. 253-259. Available at: http://www.ams.usda.gov/AMSv1.0/getfile?dDocName=STELPRDC5095829

4. See Melissa Del Bosque, Clone on the Range, Texas Observer, September 14, 2011 available at: http://www.texasob-server.org/clone-on-the-range-2/

5. See Daniel Boffey, “El Cardinal, the Opus Dei devotee behind cloning firm”, The Daily Mail, UK, August 20, 2010 avail-able at: http://www.dailymail.co.uk/news/article-1301215/Wisconsins-king-copy-cattle-Farmer-sold-cloned-cow-embryos-Britain-claims-fell-sales-patter-promising-prize-animal-live-ever.

Page 35: GeneWatch Vol. 26 No. 1

GeneWatch 35VoluMe 26 nuMber 1

Can genes determine which fifty-year-old will succumb to Alzheimer’s, which citizen will turn out on voting day, and which child will be marked for a life of crime? Yes, according to the Internet, a few scientific studies, and some in the biotechnology industry who should know better. Sheldon Krimsky and Jeremy Gruber gather a team of genetic experts to argue that treating genes as the holy grail of our physical being is a patently unscientific endeavor. Genetic Explanations urges us to replace our faith in genetic determinism with scientific knowledge about how DNA actually contributes to human development.

The concept of the gene has been steadily revised since Watson and Crick discovered the structure of the DNA molecule in 1953. No longer viewed by scientists as the cell’s fixed set of master molecules, genes and DNA are seen as a dynamic script that is ad-libbed at each stage of development. Rather than an autonomous predictor of disease, the DNA we inherit interacts continuously with the environment and functions differently as we age. What our parents hand down to us is just the beginning. Emphasizing relatively new understandings of genetic plasticity and epigenetic inheritance, the authors put into a broad developmental context the role genes are known to play in disease, behavior, evolution, and cognition.

Rather than dismissing genetic reductionism out of hand, Krimsky and Gruber ask why it persists despite opposing scientific evidence, how it influences attitudes about human behavior, and how it figures in the politics of research funding.

Sheldon Krimsky is Professor of Urban & Environmental Policy & Planning in the School of Arts and Sciences and Adjunct Professor of Public Health and Community Medicine in the School of Medicine at Tufts University. Jeremy Gruber is President and Executive Director of the Council for Responsible Genetics.

HARVARD UNIVERSITY PRESSWWW.HUP.HARVARD.EDU • HARVARDPRESS.TYPEPAD.COM • 800.405.1619

GEnEtic ExplanationSSense and Nonsense

Edited by ShEldon KrimSKy and JErEmy GrubEr

new in cloth / $45.00384 pages • 6⅛ x 9¼ inches • 2 graphs, 4 tables

Read more about this book:www.hup.harvard.edu/catalog.php?isbn=9780674064461

For email sign-up or to receive HUP catalogs, visit: www.hup.harvard.edu/news/email

Page 36: GeneWatch Vol. 26 No. 1

council For resPonsible Genetics

5 Upland Road, Suite 3Cambridge, MA 02140

GENEWATCH IS FREEON THE WEB - BUT YOU CAN STILL SUBSCRIBE TO RECEIVE PRINT ISSUES.

ANNUAL PRINT SUBSCRIPTION RATES

INDIVIDUAL: $35NON - PROFIT: $50 LIBRARY: $70 CORPORATION: $100

All figures in US dollars. Future issues of GeneWatch will be exclusively electronic and free at www.councilforresponsiblegenetics.org. If you wish to obtain a special print copy of a single issue, please send a request to the address below or call 617.868.0870.

PAYMENT INFORMATION

Please send a subscription request with your full name, address and email address, along with a check or credit card (MasterCard or Visa) information, to:

Council for Responsible Genetics5 Upland Road, Suite 3 Cambridge, MA 02140

Orders are also accepted online(www.councilforresponsiblegenetics.org)and by phone (617.868.0870). Additional donations are welcome. Thank you!

IMPORTANT TAX INFORMATION: THE FAIR MARKET VALUE OF A ONEYEAR SUBSCRIPTION TO GENEWATCH IS $35.00

(PROOF OF NON-PROFIT STATUS MAY BE REQUIRED)

Support from people like you makes CRG’s work possible.

Much of our income comes from individuals. Your support helps keep our programs free of the restrictions that come with funding from pharmaceutical and health care companies or government sources. We are the watchdogs for accurate and unbiased information about biotechnology, even when the truth doesn’t suit current political or commercial agendas. And we depend on you to be able to do what we do.

There are many ways you can help CRG. You can become a donor: an annual gift in quarterly installments of $25, $50 or $100 gives us a wonderful and predictable support with a minimal shock to your budget. You may also be able to designate CRG through your workplace giving program, including the Combined Federal Campaign. Many companies will actually match or even double-match your donation. Check with your employer about its matching gift program. You might also consider making an investment in a future where biotechnology is properly used by remembering CRG in your will with a bequest or charitable trust gift.

To learn more about helping CRG, please call us at 617.868.0870. You may also write the Council for Responsible Genetics at 5 Upland Road, Suite 3, Cambridge MA 02140, or visit www.councilforresponsiblegenetics.org.

ISSN 0740-9737