Genetic Modification of Human Beings

Genetic Modification of Human Beings

Read 461-502 in Barnet and Bedau
After reading the essays presenting the pros and cons on each one of the five issues in Part 4, choose one of the five topics presented and write your own reasoned view. Look at the thought-provoking questions at the end of the essay. Where do you stand on this issue? Why?
In your essay you should quote, paraphrase, and summarize the argument from the essay to which you are responding. Then you must go to EBSCO and find two articles that help you see your topic differently or more clearly.
Paper must use the MLA documentation method.
Length: 5-7 pages
Genetic Modification of Human Beings: Is It Acceptable?
Opinions Essay Topic from the book Current Issues by Sylvan Barnet and Hugo Bedau page495-498
Building Baby From the Genes Up

By Ronald M. Green
Sunday, April 13, 2008
The two British couples no doubt thought that their appeal for medical help in conceiving a child was entirely reasonable. Over several generations, many female members of their families had died of breast cancer. One or both spouses in each couple had probably inherited the genetic mutations for the disease, and they wanted to use in-vitro fertilization and preimplantation genetic diagnosis (PGD) to select only the healthy embryos for implantation. Their goal was to eradicate breast cancer from their family lines once and for all.
In the United States, this combination of reproductive and genetic medicine — what one scientist has dubbed “reprogenetics” — remains largely unregulated, but Britain has a formal agency, the Human Fertilization and Embryology Authority (HFEA), that must approve all requests for PGD. In July 2007, after considerable deliberation, the HFEA approved the procedure for both families. The concern was not about the use of PGD to avoid genetic disease, since embryo screening for serious disorders is commonplace now on both sides of the Atlantic. What troubled the HFEA was the fact that an embryo carrying the cancer mutation could go on to live for 40 or 50 years before ever developing cancer, and there was a chance it might never develop. Did this warrant selecting and discarding embryos? To its critics, the HFEA, in approving this request, crossed a bright line separating legitimate medical genetics from the quest for “the perfect baby.”

Like it or not, that decision is a sign of things to come — and not necessarily a bad sign. Since the completion of the Human Genome Project in 2003, our understanding of the genetic bases of human disease and non-disease traits has been growing almost exponentially. The National Institutes of Health has initiated a quest for the “$1,000 genome,” a 10-year program to develop machines that could identify all the genetic letters in anyone’s genome at low cost (it took more than $3 billion to sequence the first human genome). With this technology, which some believe may be just four or five years away, we could not only scan an individual’s — or embryo’s — genome, we could also rapidly compare thousands of people and pinpoint those DNA sequences or combinations that underlie the variations that contribute to our biological differences.
With knowledge comes power. If we understand the genetic causes of obesity, for example, we can intervene by means of embryo selection to produce a child with a reduced genetic likelihood of getting fat. Eventually, without discarding embryos at all, we could use gene-targeting techniques to tweak fetal DNA sequences. No child would have to face a lifetime of dieting or experience the health and cosmetic problems associated with obesity. The same is true for cognitive problems such as dyslexia. Geneticists have already identified some of the mutations that contribute to this disorder. Why should a child struggle with reading difficulties when we could alter the genes responsible for the problem?
Many people are horrified at the thought of such uses of genetics, seeing echoes of the 1997 science-fiction film “Gattaca,” which depicted a world where parents choose their children’s traits. Human weakness has been eliminated through genetic engineering, and the few parents who opt for a “natural” conception run the risk of producing offspring — “invalids” or “degenerates” — who become members of a despised underclass. Gattaca’s world is clean and efficient, but its eugenic obsessions have all but extinguished human love and compassion.
These fears aren’t limited to fiction. Over the past few years, many bioethicists have spoken out against genetic manipulations. The critics tend to voice at least four major concerns. First, they worry about the effect of genetic selection on parenting. Will our ability to choose our children’s biological inheritance lead parents to replace unconditional love with a consumerist mentality that seeks perfection?
Second, they ask whether gene manipulations will diminish our freedom by making us creatures of our genes or our parents’ whims. In his book “Enough,” the techno-critic Bill McKibben asks: If I am a world-class runner, but my parents inserted the “Sweatworks2010 GenePack” in my genome, can I really feel pride in my accomplishments? Worse, if I refuse to use my costly genetic endowments, will I face relentless pressure to live up to my parents’ expectations?
Third, many critics fear that reproductive genetics will widen our social divisions as the affluent “buy” more competitive abilities for their offspring. Will we eventually see “speciation,” the emergence of two or more human populations so different that they no longer even breed with one another? Will we re-create the horrors of eugenics that led, in Europe, Asia and the United States, to the sterilization of tens of thousands of people declared to be “unfit” and that in Nazi Germany paved the way for the Holocaust?
Finally, some worry about the religious implications of this technology. Does it amount to a forbidden and prideful “playing God”?
To many, the answers to these questions are clear. Not long ago, when I asked a large class at Dartmouth Medical School whether they thought that we should move in the direction of human genetic engineering, more than 80 percent said no. This squares with public opinion polls that show a similar degree of opposition. Nevertheless, “babies by design” are probably in our future — but I think that the critics’ concerns may be less troublesome than they first appear.
Will critical scrutiny replace parental love? Not likely. Even today, parents who hope for a healthy child but have one born with disabilities tend to love that child ferociously. The very intensity of parental love is the best protection against its erosion by genetic technologies. Will a child somehow feel less free because parents have helped select his or her traits? The fact is that a child is already remarkably influenced by the genes she inherits. The difference is that we haven’t taken control of the process. Yet.
Knowing more about our genes may actually increase our freedom by helping us understand the biological obstacles — and opportunities — we have to work with. Take the case of Tiger Woods. His father, Earl, is said to have handed him a golf club when he was still in the playpen. Earl probably also gave Tiger the genes for some of the traits that help make him a champion golfer. Genes and upbringing worked together to inspire excellence. Does Tiger feel less free because of his inherited abilities? Did he feel pressured by his parents? I doubt it. Of course, his story could have gone the other way, with overbearing parents forcing a child into their mold. But the problem in that case wouldn’t be genetics, but bad parenting.
Granted, the social effects of reproductive genetics are worrisome. The risks of producing a “genobility,” genetic overlords ruling a vast genetic underclass, are real. But genetics could also become a tool for reducing the class divide. Will we see the day when perhaps all youngsters are genetically vaccinated against dyslexia? And how might this contribute to everyone’s social betterment?
As for the question of intruding on God’s domain, the answer is less clear than the critics believe. The use of genetic medicine to cure or prevent disease is widely accepted by religious traditions, even those that oppose discarding embryos. Speaking in 1982 at the Pontifical Academy of Sciences, Pope John Paul II observed that modern biological research “can ameliorate the condition of those who are affected by chromosomic diseases,” and he lauded this as helping to cure “the smallest and weakest of human beings . . . during their intrauterine life or in the period immediately after birth.” For Catholicism and some other traditions, it is one thing to cure disease, but another to create children who are faster runners, longer-lived or smarter.
But why should we think that the human genome is a once-and-for-all-finished, untamperable product? All of the biblically derived faiths permit human beings to improve on nature using technology, from agriculture to aviation. Why not improve our genome? I have no doubt that most people considering these questions for the first time are certain that human genetic improvement is a bad idea, but I’d like to shake up that certainty.
Genomic science is racing toward a future in which foreseeable improvements include reduced susceptibility to a host of diseases, increased life span, better cognitive functioning and maybe even cosmetic enhancements such as whiter, straighter teeth. Yes, genetic orthodontics may be in our future. The challenge is to see that we don’t also unleash the demons of discrimination and oppression. Although I acknowledge the risks, I believe that we can and will incorporate gene technology into the ongoing human adventure.
Ronald M. Green is a professor of ethics at Dartmouth College. His most recent book is “Babies by Design: The Ethics of Genetic Choice.”
Citation:
Barnet, Sylvan, and Hugo Adam Bedau. “Part Four Current Issues: Occasions for Debate.” Current Issues and Enduring Questions a Guide to Critical Thinking and Argument, with Readings. Boston: Bedford/St. Martins, 2014. 495-98. Print.

Personal point of view:
Human modification should not be acceptable because as humans we make mistakes; making a mistake with human life could be irreversible and dangerous. If is possible to create new humans, it may be also possible to find cures to all the diseases that gene modification claims will “prevent”. Before we make a mistake that could cost a highly price to humanity, scientist should find a cure to poverty, instead of pleasing rich people with a perfect baby.

The following are the articles:

Engineering the Perfect Baby. (cover story)
Images

Authors:
Regalado, Antonio
Source:
MIT Technology Review. May/Jun2015, Vol. 118 Issue 3, p26-33. 8p. 1 Color Photograph, 1 Graph.
Document Type:
Article
Subjects:
DNA — Modification & restriction
HUMAN genetic engineering
CRISPRS (Genetics)
LUHAN Yang
STEM cells
SPERMATOZOA
Abstract:
The article reports if scientists should stop to develop ways to edit the DNA of future children. Topics include the role played by scientist Luhan Yang in developing CRISPR-Cas9 which is a technology for editing DNA, the emergence of CRISPR technology as kind of tool for biologists to alter DNA, and the easiness of editing a human embryo using CRISPR. Also mentioned is the possibility of using stem cells to produce eggs and sperm in the laboratory.
Lexile:
1220
Full Text Word Count:
5163
ISSN:
1099-274X
Accession Number:
102929707
Database:
MasterFILE Complete

Engineering the Perfect Baby
Scientists are developing ways to edit the DNA of tomorrow’s children. Should they stop before it’s too late?
If anyone had devised away to create a genetically engineered baby, I figured George Church would know about it.
At his labyrinthine laboratory on the Harvard Medical School campus, you can find researchers giving E. Coli a novel genetic code never seen in nature. Around another bend, others are carrying out a plan to use DNA engineering to resurrect the woolly mammoth. His lab, Church likes to say, is the center of a new technological genesis–one in which man rebuilds creation to suit himself.
When I visited the lab last June, Church proposed that I speak to a young postdoctoral scientist named Luhan Yang. A Harvard recruit from Beijing, she’d been a key player in developing a powerful new technology for editing DNA, called CRISPR-Cas9. With Church, Yang had founded a small biotechnology company to engineer the genomes of pigs and cattle, sliding in beneficial genes and editing away bad ones.
As I listened to Yang, I waited for a chance to ask my real questions: Can any of this be done to human beings? Can we improve the human gene pool? The position of much of mainstream science has been that such meddling would be unsafe, irresponsible, and even impossible. But Yang didn’t hesitate. Yes, of course, she said. In fact, the Harvard laboratory had a project under way to determine how it could be achieved. She flipped open her laptop to a PowerPoint slide titled “Germline Editing Meeting.”
Here it was: a technical proposal to alter human heredity. “Germ line” is biologists’ jargon for the egg and sperm, which combine to form an embryo. By editing the DNA of these cells or the embryo itself, it could be possible to correct disease genes and pass those genetic fixes on to future generations. Such a technology could be used to rid families of scourges like cystic fibrosis. It might also be possible to install genes that offer lifelong protection against infection, Alzheimer’s, and, Yang told me, maybe the effects of aging. Such history-making medical advances could be as important to this century as vaccines were to the last.
That’s the promise. The fear is that germ-line engineering is a path toward a dystopia of superpeople and designer babies for those who can afford it. Want a child with blue eyes and blond hair? Why not design a highly intelligent group of people who could be tomorrow’s leaders and scientists?
Just three years after its initial development, CRISPR technology is already widely used by biologists as a kind of search-and-replace tool to alter DNA, even down to the level of a single letter. It’s so precise that it’s expected to turn into a promising new approach for gene therapy in people with devastating illnesses. The idea is that physicians could directly correct a faulty gene, say, in the blood cells of a patient with sickle-cell anemia (see “Genome Surgery,” March/April 2014). But that kind of gene therapy wouldn’t affect germ cells, and the changes in the DNA wouldn’t get passed to future generations.
In contrast, the genetic changes created by germ-line engineering would be passed on, and that’s what has made the idea seem so objectionable. So far, caution and ethical concerns have had the upper hand. A dozen countries, not including the United States, have banned germ-line engineering, and scientific societies have unanimously concluded that it would be too risky to do. The European Union’s convention on human rights and biomedicine says tampering with the gene pool would be a crime against “human dignity” and human rights.
But all these declarations were made before it was actually feasible to precisely engineer the germ line. Now, with CRISPR, it is possible.
The experiment Yang described, though not simple, would go like this: The researchers hoped to obtain, from a hospital in New York, the ovaries of a woman undergoing surgery for ovarian cancer caused by a mutation in a gene called BRCA1. Working with another Harvard laboratory, that of antiaging specialist David Sinclair, they would extract immature egg cells that could be coaxed to grow and divide in the laboratory. Yang would use CRISPR in these cells to correct the DNA of the BRCA1 gene. They would try to create a viable egg without the genetic error that caused the woman’s cancer.
Yang would later tell me that she dropped out of the project not long after we spoke. Yet it remained difficult to know if the experiment she described was occurring, canceled, or awaiting publication. Sinclair said that a collaboration between the two labs was ongoing, but then, like several other scientists whom I’d asked about germ-line engineering, he stopped replying to my e-mails.
Regardless of the fate of that particular experiment, human germ-line engineering has become a burgeoning research concept. At least three other centers in the United States are working on it, as are scientists in China, in the U.K., and at a biotechnology company called OvaScience, based in Cam bridge, Massachusetts, that boasts some of the world’s leading fertility doctors on its advisory board.
The objective of these groups is to demonstrate that it’s possible to produce children free of specific genes involved in inherited disease. If it’s possible to correct the DNA in a woman’s egg, or a man’s sperm, those cells could be used in an in vitro fertilization (IVF) clinic to produce an embryo and then a child. It might also be possible to directly edit the DNA of an early-stage IVF embryo using CRISPR. Several people interviewed by MIT Technology Review said that such experiments had already been carried out in China and that results describing edited embryos were pending publication. These people, including two high-ranking specialists, didn’t wish to comment publicly because the papers are under review.
All this means that germ-line engineering is much further along than anyone imagined. “What you are talking about is a major issue for all humanity,” says Merle Berger, one of the founders of Boston IVF, a network of fertility clinics that is among the largest in the world and helps more than a thousand women get pregnant each year. “It would be the biggest thing that ever happened in our field.” Berger predicts that repairing genes involved in serious inherited diseases will win wide public acceptance but says the idea of using the technology beyond that would cause a public uproar because “everyone would want the perfect child”: people might pick and choose eye color and eventually intelligence. “These are things we talk about all the time,” he says. “But we have never had the opportunity to do it.”
Editing embryos
How easy would it be to edit a human embryo using CRISPR? Very easy, experts say. “Any scientist with molecular biology skills and knowledge of how to work with [embryos] is going to be able to do this,” says Jennifer Doudna, a biologist at the University of California, Berkeley, who in 2012 co-discovered how to use CRISPR to edit genes.
To find out how it could be done, I visited the lab of Guoping Feng, a biologist at MIT’s McGovern Institute for Brain Research, where a colony of marmoset monkeys is being established with the aim of using CRISPR to create accurate models of human brain diseases. To create the models, Feng will edit the DNA of embryos and then transfer them into female marmosets to produce live monkeys. One gene Feng hopes to alter in the animals is SHANK3. The gene is involved in how neurons communicate; when it’s damaged in children, it is known to cause autism.
Feng said that before CRISPR, it was not possible to introduce precise changes into a primate’s DNA. With CRISPR, the technique should be relatively straightforward. The CRISPR system includes a gene-snipping enzyme and a guide molecule that can be programmed to target unique combinations of the DNA letters, A, G, C, and T; get these ingredients into a cell and they will cut and modify the genome at the targeted sites.
But CRISPR is not perfect–and it would be a very haphazard way to edit human embryos, as Feng’s efforts to create gene-edited marmosets show. To employ the CRISPR system in the monkeys, his students simply inject the chemicals into a fertilized egg, which is known as a zygote–the stage just before it starts dividing.
Feng said the efficiency with which CRISPR can delete or disable a gene in a zygote is about 40 percent, whereas making specific edits, or swapping DNA letters, works less frequently–more like 20 percent of the time. Like a person, a monkey has two copies of most genes, one from each parent. Sometimes both copies get edited, but sometimes just one does, or neither. Only about half the embryos will lead to live births, and of those that do, many could contain a mixture of cells with edited DNA and without. If you add up the odds, you find you’d need to edit 20 embryos to get a live monkey with the version you want.
That’s not an insurmountable problem for Feng, since the MIT breeding colony will give him access to many monkey eggs and he’ll be able to generate many embryos. However, it would present obvious problems in humans. Putting the ingredients of CRISPR into a human embryo would be scientifically trivial. But it wouldn’t be practical for much just yet. This is one reason that many scientists view such an experiment (whether or not it has really occurred in China) with scorn, seeing it more as a provocative bid to grab attention than as real science. Rudolf Jaenisch, an MIT biologist who works across the street from Feng and who in the 1970s created the first gene-modified mice, calls attempts to edit human embryos “totally premature.” He says he hopes these papers will be rejected and not published. “It’s just a sensational thing that will stir things up,” says Jaenisch. “We know it’s possible, but is it of practical use? I kind of doubt it.”
For his part, Feng told me he approves of the idea of germ-line engineering. Isn’t the goal of medicine to reduce suffering? Considering the state of the technology, however, he thinks actual gene-edited humans are “10 to 20 years away.” Among other problems, CRISPR can introduce off-target effects or change bits of the genome far from where scientists had intended. Any human embryo altered with CRISPR today would carry the risk that its genome had been changed in unexpected ways. But, Feng said, such problems may eventually be ironed out, and edited people will be born. “To me, it’s possible in the long run to dramatically improve health, lower costs. It’s a kind of prevention,” he said. “It’s hard to predict the future, but correcting disease risks is definitely a possibility and should be supported. I think it will be a reality.”
Editing eggs
Elsewhere in the Boston area, scientists are exploring a different approach to engineering the germ line, one that is technically more demanding but probably more powerful. This strategy combines CRISPR with unfolding discoveries related to stem cells. Scientists at several centers, including Church’s, think they will soon be able to use stem cells to produce eggs and sperm in the laboratory. Unlike embryos, stem cells can be grown and multiplied. Thus they could offer a vastly improved way to create edited offspring with CRISPR. The recipe goes like this: First, edit the genes of the stem cells. Second, turn them into an egg or sperm. Third, produce an offspring.
Some investors got an early view of the technique on December 17, at the Benjamin Hotel in Manhattan, during commercial presentations by OvaScience. The company, which was founded four years ago, aims to commercialize the scientific work of David Sinclair, who is based at Harvard, and Jonathan Tilly, an expert on egg stem cells and the chairman of the biology department at Northeastern University (see “10 Emerging Technologies: Egg Stem Cells,” May/June 2012). It made the presentations as part of a successful effort to raise $132 million in new capital during January.
During the meeting, Sinclair, a velvet-voiced Australian whom Time last year named one of the “100 Most Influential People in the World,” took the podium and provided Wall Street with a peek at what he called “truly world-changing” developments. People would look back at this moment in time and recognize it as a new chapter in “how humans control their bodies,” he said, because it would let parents determine “when and how they have children and how healthy those children are actually going to be.”
The company has not perfected its stem-cell technology–it has not reported that the eggs it grows in the lab are viable-but Sinclair predicted that functional eggs were “a when, and not an if.” Once the technology works, he said, infertile women will be able to produce hundreds of eggs, and maybe hundreds of embryos. Using DNA sequencing to analyze their genes, they could pick among them for the healthiest ones.
Genetically improved children may also be possible. Sinclair told the investors that he was trying to alter the DNA of these egg stem cells using gene editing, work he later told me he was doing with Church’s lab. “We think the new technologies with genome editing will allow it to be used on individuals who aren’t just interested in using IVF to have children but have healthier children as well, if there is a genetic disease in their family,” Sinclair told the investors. He gave the example of Huntington’s disease, caused by a gene that will trigger a fatal brain condition even in someone who inherits only one copy. Sinclair said gene editing could be used to remove the lethal gene defect from an egg cell. His goal, and that of OvaScience, is to “correct those mutations before we generate your child,” he said. “It’s still experimental, but there is no reason to expect it won’t be possible in coming years.”
Sinclair spoke to me briefly on the phone while he was navigating in a cab across a snowed-in Boston, but later he referred my questions to OvaScience. When I contacted OvaScience, Cara Mayfield, a spokeswoman, said its executives could not comment because of their travel schedules but confirmed that the company was working on treating inherited disorders with gene editing. What was surprising to me was that OvaScience’s research in “crossing the germ line,” as critics of human engineering sometimes put it, has generated scarcely any notice. In December of 2013, OvaScience even announced it was putting $1.5 million into a joint venture with a synthetic-biology company called Intrexon, whose R&D objectives include gene-editing eggs to “prevent the propagation” of human disease “in future generations.”
When I reached Tilly at Northeastern, he laughed when I told him what I was calling about. “It’s going to be a hot-button issue,” he said. Tilly also said his lab was trying to edit egg stem cells with CRISPR “right now” to rid them of an inherited genetic disease that he didn’t want to name. Tilly emphasized that there are “two pieces of the puzzle”–one being stem cells and the other gene editing. The ability to create large numbers of egg stem cells is critical, because only with sizable quantities can genetic changes be stably introduced using CRISPR, characterized using DNA sequencing, and carefully studied to check for mistakes before producing an egg.
Tilly predicted that the whole end-to-end technology–cells to stem cells, stem cells to sperm or egg and then to offspring–would end up being worked out first in animals, such as cattle, either by his lab or by companies such as eGenesis, the spinoff from the Church lab working on livestock. But he isn’t sure what the next step should be with edited human eggs. You wouldn’t want to fertilize one “willy nilly,” he said. You’d be making a potential human being. And doing that would raise questions he’s not sure he can answer. He told me, “‘Can you do it?’ is one thing. If you can, then the most important questions come up. ‘Would you do it? Why would you want to do it? What is the purpose?’ As scientists we want to know if it’s feasible, but then we get into the bigger questions, and it’s not a science question–it’s a society question.”
Improving humans
If germ-line engineering becomes part of medical practice, it could lead to transformative changes in human well-being, with consequences to people’s life span, identity, and economic output. But it would create ethical dilemmas and social challenges. What if these improvements were available only to the richest societies, or the richest people? An in vitro fertility procedure costs about $20,000 in the United States. Add genetic testing and egg donation or a surrogate mother, and the price soars toward $100,000.
Others believe the idea is dubious because it’s not medically necessary. Hank Greely, a lawyer and ethicist at Stanford University, says proponents “can’t really say what it is good for.” The problem, says Greely, is that it’s already possible to test the DNA of IVF embryos and pick healthy ones, a process that adds about $4,000 to the cost of a fertility procedure. A man with Huntington’s, for instance, could have his sperm used to fertilize a dozen of his partner’s eggs. Half those embryos would not have the Huntington’s gene, and those could be used to begin a pregnancy.
Indeed, some people are adamant that germ-line engineering is being pushed ahead with “false arguments.” That is the view of Edward Lanphier, CEO of Sangamo Biosciences, a California biotechnology company that is using another gene-editing technique, called zinc fingers nucleases, to try to treat HIV in adults by altering their blood cells. “We’ve looked at [germ-line engineering] for a disease rationale, and there is none,” he says. “You can do it. But there really isn’t a medical reason. People say, well, we don’t want children born with this, or born with that–but it’s a completely false argument and a slippery slope toward much more unacceptable uses.”
Critics cite a host of fears. Children would be the subject of experiments. Parents would be influenced by genetic advertising from IVF clinics. Germ-line engineering would encourage the spread of allegedly superior traits. And it would affect people not yet born, without their being able to agree to it. The American Medical Association, for instance, holds that germ-line engineering shouldn’t be done “at this time” because it “affects the welfare of future generations” and could cause “unpredictable and irreversible results.” But like a lot of official statements that forbid changing the genome, the AMA’s, which was last updated in 1996, predates today’s technology. “A lot of people just agreed to these statements,” says Greely. “It wasn’t hard to renounce something that you couldn’t do.”
Others predict that hard-to-oppose medical uses will be identified. A couple with several genetic diseases at once might not be able to find a suitable embryo. Treating infertility is another possibility. Some men don’t produce any sperm, a condition called azoospermia. One cause is a genetic defect in which a region of about one million to six million DNA letters is missing from the Y chromosome. It might be possible to take a skin cell from such a man, turn it into a stem cell, repair the DNA, and then make sperm, says Werner Neuhausser, a young Austrian doctor who splits his time between the Boston IVF fertility-clinic network and Harvard’s Stem Cell Institute. “That will change medicine forever, right? You could cure infertility, that is for sure,” he says.
I spoke with Church several times by telephone over the last few months, and he told me what’s driving everything is the “incredible specificity” of CRISPR. Although not all the details have been worked out, he thinks the technology could replace DNA letters essentially without side effects. He says this is what makes it “tempting to use.” Church says his laboratory is focused mostly on experiments in engineering animals. He added that his lab would not make or edit human embryos, calling such a step “not our style.”
What is Church’s style is human enhancement. And he’s been making a broad case that CRISPR can do more than eliminate disease genes. It can lead to augmentation. At meetings, some involving groups of “transhumanists” interested in next steps for human evolution, Church likes to show a slide on which he lists naturally occurring variants of around 10 genes that, when people are born with them, confer extraordinary qualities or resistance to disease. One makes your bones so hard they’ll break a surgical drill. Another drastically cuts the risk of heart attacks. And a variant of the gene for the amyloid precursor protein, or APP, was found by Icelandic researchers to protect against Alzheimer’s. People with it never get dementia and remain sharp into old age.
Church thinks CRISPR could be used to provide people with favorable versions of genes, making DNA edits that would act as vaccines against some of the most common diseases we face today. Although he told me anything “edgy” should be done only to adults who can consent, it’s obvious to him that the earlier such interventions occur, the better.
Church tends to dodge questions about genetically modified babies. The idea of improving the human species has always had “enormously bad press,” he wrote in the introduction to Regenesis, his 2012 book on synthetic biology, whose cover was a painting by Eustache Le Sueur of a bearded God creating the world. But that’s ultimately what he’s suggesting: enhancements in the form of protective genes. “An argument will be made that the ultimate prevention is that the earlier you go, the better the prevention,” he told an audience at MIT’s Media Lab last spring. “I do think it’s the ultimate preventive, if we get to the point where it’s very inexpensive, extremely safe, and very predictable.” Church, who has a less cautious side, proceeded to tell the audience that he thought changing genes “is going to get to the point where it’s like you are doing the equivalent of cosmetic surgery.”
Some thinkers have concluded that we should not pass up the chance to make improvements to our species. “The human genome is not perfect,” says John Harris, a bioethicist at Manchester University, in the U.K. “It’s ethically imperative to positively support this technology.” By some measures, U.S. public opinion is not particularly negative toward the idea. A Pew Research survey carried out last August found that 46 percent of adults approved of genetic modification of babies to reduce the risk of serious diseases.
The same survey found that 83 percent said genetic modification to make a baby smarter would be “taking medical advances too far.” But other observers say higher IQ is exactly what we should be considering. Nick Bostrom, an Oxford philosopher best known for his 2014 book Superintelligence, which raised alarms about the risks of artificial intelligence in computers, has also looked at whether humans could use reproductive technology to improve human intellect. Although the ways in which genes affect intelligence aren’t well understood and there are far too many relevant genes to permit easy engineering, such realities don’t dim speculation on the possibility of high-tech eugenics.
What if everyone could be a little bit smarter? Or a few people could be a lot smarter? Even a small number of “super-enhanced” individuals, Bostrom wrote in a 2013 paper, could change the world through their creativity and discoveries, and through innovations that everyone else would use. In his view, genetic enhancement is an important long-range issue like climate change or financial planning by nations, “since human problem-solving ability is a factor in every challenge we face.”
To some scientists, the explosive advance of genetics and biotech means germ-line engineering is inevitable. Of course, safety questions would be paramount. Before there’s a genetically edited baby saying “Mama,” there would have to be tests in rats, rabbits, and probably monkeys, to make sure they are normal. But ultimately, if the benefits seem to outweigh the risks, medicine would take the chance. “It was the same with IVF when it first happened,” says Neuhausser. “We never really knew if that baby was going to be healthy at 40 or 50 years. But someone had to take the plunge.”
Wine country
In January, on Saturday the 24th, around 20 scientists, ethicists, and legal experts traveled to Napa Valley, California, for a retreat among the vineyards at the Carneros Inn. They had been convened by Doudna, the Berkeley scientist who co-discovered the CRISPR system a little over two years ago. She had become aware that scientists might be thinking of crossing the germ line, and she was concerned. Now she wanted to know: could they be stopped?
“We as scientists have come to appreciate that CRISPR is incredibly powerful. But that swings both ways. We need to make sure that it’s applied carefully,” Doudna told me. “The issue is especially human germ-line editing and the appreciation that this is now a capability in everyone’s hands.”
At the meeting, along with ethicists like Greely, was Paul Berg, a Stanford biochemist and Nobel Prize winner known for having organized the Asilomar Conference, a historic 1975 forum at which biologists reached an agreement on how to safely proceed with recombinant DNA, the newly discovered method of splicing DNA into bacteria.
Should there be an Asilomar for germ-line engineering? Doudna thinks so, but the prospects for consensus seem dim. Biotechnology research is now global, involving hundreds of thousands of people. There’s no single authority that speaks for science, and no easy way to put the genie back in the bottle. Doudna told me she hoped that if American scientists agreed to a moratorium on human germ-line engineering, it might influence researchers elsewhere in the world to cease their work.
Doudna said she felt that a self-imposed pause should apply not only to making gene-edited babies but also to using CRISPR to alter human embryos, eggs, or sperm–as researchers at Harvard, Northeastern, and OvaScience are doing. “I don’t feel that those experiments are appropriate to do right now in human cells that could turn into a person,” she told me. “I feel that the research that needs to be done right now is to understand safety, efficacy, and delivery. And I think those experiments can be done in nonhuman systems. I would like to see a lot more work done before it’s done for germ-line editing. I would favor a very cautious approach.”
Not everyone agrees that germ-line engineering is such a big worry, or that experiments should be padlocked. Greely notes that in the United States, there are piles of regulations to keep lab science from morphing into a genetically modified baby anytime soon. “I would not want to use safety as an excuse for a non-safety-based ban,” says Greely, who says he pushed back against talk of a moratorium. But he also says he agreed to sign Doudna’s letter, which now reflects the consensus of the group. “Although I don’t view this as a crisis moment, I think it’s probably about time for us to have this discussion,” he says.
(After this article was published online in March, Doudna’s editorial appeared in Science. Along with Greely, Berg, and 15 others, she called for a global moratorium on any effort to use CRISPR to generate gene-edited children until researchers could determine “what clinical applications, if any, might in the future be deemed permissible.” The group, however, endorsed basic research, including applying CRISPR to embryos. The final list of signatories included Church, although he did not attend the Napa meeting.)
As news has spread of germ-line experiments, some biotechnology companies now working on CRISPR have realized that they will have to take a stand. Nessan Bermingham is CEO of Intellia Therapeutics, a Boston startup that raised $15 million last year to develop CRISPR into gene therapy treatments for adults or children. He says germ-line engineering “is not on our commercial radar,” and he suggests that his company could use its patents to prevent anyone from commercializing it.
“The technology is in its infancy,” he says. “It is not appropriate for people to even be contemplating germ-line applications.”
Bermingham told me he never imagined he’d have to be taking a position on genetically modified babies so soon. Modifying human heredity has always been a theoretical possibility. Suddenly it’s a real one. But wasn’t the point always to understand and control our own biology–to become masters over the processes that created us?
Doudna says she is also thinking about these issues. “It cuts to the core of who we are as people, and it makes you ask if humans should be exercising that kind of power,” she told me. “There are moral and ethical issues, but one of the profound questions is just the appreciation that if germ-line editing is conducted in humans, that is changing human evolution.” One reason she feels the research should slow down is to give scientists a chance to spend more time explaining what their next steps could be.
“Most of the public,” she says, “does not appreciate what is coming.”
CRISPR in the Human Gene Pool
Key players in the development of human germ-line editing.
Jennifer Doudna University of California, Berkeley
Key achievement: In 2012, co-developed CRISPR editing system, using bacteria.
Current work: Has expressed concern about CRISPR technology and its potential germ-line applications.
George Church Harvard Medical School
Key achievement: In 2013, demonstrated that CRISPR can work in human cells.
Current work: Project to engineer genomes of animals, including pigs whose organs can be transplanted to human patients.
OvaScience Cambridge, Massachusetts
Key achievement: Raised nearly $300 million to commercialize egg stem-cell technology for IVF clinics.
Current work: Exploring the possibility of editing eggs to remove disease genes from future generations.
Jinsong Li Shanghai Institute for Biological Sciences
Key achievement: In 2015, corrected a genetic disease in mice by editing the DNA of sperm cells. Current work: Hopes to edit human sperm and demonstrate medical applications for IVF procedures.
Xingxu Huang ShanghaiTech University
Key achievement: In 2014, was part of team that bred the first CRISPR-edited monkeys in China.
Current work: Seeking permission to genetically modify discarded IVF embryos.
Azim Surani University of Cambridge, U.K.
Key achievement: In late 2014, showed that human skin cells can be turned into primitive eggs and sperm.
Current work: Using CRISPR in stem cells to study basic questions in developmental biology.
Genetic Modification of Babies
Percentage of U.S. adults saying that changing a baby’s genetic characteristics for each purpose is …
Appropriate Taking medical advances too far
To make the baby more intelligent 15% 83%
To reduce the risk of serious diseases 46% 50%
~~~~~~~~
By Antonio Regalado
Antonio Regalado is MIT Technology Review’s biomedicine editor.
________________________________________
Copyright of MIT Technology Review is the property of MIT Technology Review and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder’s express written permission. However, users may print, download, or email articles for individual use.
AMA(American Medical Assoc.)
Reference List
Regalado A. Engineering the Perfect Baby. (cover story). MIT Technology Review [serial online]. May 2015;118(3):26. Available from: MasterFILE Complete, Ipswich, MA. Accessed June 26, 2016.
http://eds.b.ebscohost.com.libraryaccess.sdmesa.edu/eds/detail/detail?vid=2&sid=b581606e-f68d-4282-8339-e019ee5ee743%40sessionmgr107&hid=108&bdata=JnNpdGU9ZWRzLWxpdmU%3d#AN=102929707&db=f6h
WHAT HUMAN GENETIC MODIFICATION MEANS FOR WOMEN.
Authors:
Levine, Judith
Source:
World Watch. Jul/Aug2002, Vol. 15 Issue 4, p26. 4p.
Document Type:
Article
Subjects:
BIOTECHNOLOGY
FEMINISM
GENE therapy
Abstract:
Focuses on the view of the feminists on human genetic engineering. Threat of genetic engineering on women; Existence of fertility therapy; Effectiveness of genetic therapy. INSET: THE ROLE OF NORTH AMERICAN WOMEN..
Lexile:
1140
Full Text Word Count:
2711
ISSN:
0896-0615
Accession Number:
7198350
Database:
MasterFILE Complete

WHAT HUMAN GENETIC MODIFICATION MEANS FOR WOMEN
Supporters of the new eugenics want it framed as an issue of “choice.” But feminists know we can support abortion rights and still oppose eugenics.
Seduced by the medical promises of genetic science or fearful of losing reproductive autonomy, many feminists have been slow to oppose human genetic engineering. But GE is a threat to women, and in the broadest sense a feminist issue. Here’s why.
If anyone should be wary of medical techniques to “improve” ordinary reproduction—as GE purports to do—it’s women. History is full of such “progress,” and its grave results. When limbless babies were born to mothers who took thalidomide, the drug was recalled. But the deadly results of another “pregnancy-enhancing” drug, DES, showed up only years later, as cancer in the daughters of DES mothers. The high-estrogen Pill was tested first on uninformed Puerto Rican mothers, some of whom may have died from it.
Today’s fertility industry takes in $4 billion a year, even though in-vitro fertilization (IVF) succeeds in only 3 of 10 cases. Virtually unregulated and highly competitive, these fertility doctors often undertake experimental treatments. Recently, the Institute for Reproductive Medicine and Science at New Jersey’s St. Barnabas Medical Center announced the success of a new fertility “therapy” called cytoplasmic transfer, in which some of the cellular material outside the nucleus of one woman’s egg is transferred into the egg of another woman who is having difficulty sustaining embryo survival. The transferred cytoplasm contains mitochondria (organelles that produce energy for the cell), which have a small number of their own genes. So the embryo produced with cytoplasmic transfer can end up with two genetic mothers. This mixing, called “mitochondrial heteroplasmy,” can cause life-threatening symptoms that don’t show up until later in life. When the Public Broadcasting Service’s Nova enthusiastically reported on the procedure, complete with footage of its cute outcome, Katy, it mentioned no risks.
Didn’t these patients give informed consent? Yes and no. Most read warnings and signed their names. But with genetic therapies there’s no such thing as “informed,” says Judy Norsigian of the Boston Women’s Health Collective, “because the risks can’t be known.” Adds biologist Ruth Hubbard, the deadliness of DES was discovered “only because it showed itself in an otherwise very rare condition. If the effects [of human genetic engineering] are delayed, and if they are not associated with a particularly unusual pathology, it could take quite a long time to find out.” Or indeed, “we might never know.”
“PERFECTING” HUMAN GENETIC MODIFICATION WOULD REQUIRE EXPERIMENTATION ON WOMEN AND CHILDREN.
Scottish biologist Ian Wilmut, the “father” of the famously first-cloned sheep Dolly, provided these statistics in 2001: Of the 31,007 sheep, mice, pig, and other mammal eggs that had undergone somatic cell nuclear transfer (cloning), 9,391 viable embryos resulted. From those embryos came 267 live-born offspring. In these animals, The New Tork Times reported, “random errors” were ubiquitous—including fatal heart and lung defects, malfunctioning immune systems, and grotesque obesity. In all, “fewer than 3 percent of all cloning efforts succeed.” Dolly may be a victim of accelerated aging, another problem in cloned animals. In January, it was reported that she has arthritis, at the unusually early age of five and a half. Mothers of clones are endangered too, since their bodies have trouble supporting the abnormally large fetuses that cloning often produces.
It’s likely that scientists will get better at cloning animals, and at the more complex procedures required to produce inheritable genetic alterations. Then, as health activists quip, if it works on a mouse, they will try it on a woman. The problem, warns Stuart Newman, a cell biologist at New York Medical College in Valhalla, is that if it works on a mouse, it is likely not to work on a woman: “Every species presents a new set of problems.” How might the process be perfected in humans? In clinical trials?
“The degree of risk to be taken should never exceed that determined by the humanitarian importance of the problem to be solved by the experiment,” reads the Nuremburg Code, drawn up after World War II to forbid future torturous experiments of the sort Nazi “scientists” inflicted on concentration-camp inmates. What is the humanitarian importance of creating a faster 100-meter sprinter? Or even curing a disease with genetic engineering when other options are still untried? The science to find “safe” means of human GE, says Newman, would constitute “an entirely experimental entcrprise with little justification.” In other words, “We can’t get there from here.”
WE ARE NOT OUR GENES.
When the Human Genome Project finished its map of our DNA, its press releases called it the “blueprint” of humanity, the very Book of Life. The newspapers had already been filling up with reports of the discovery of a “gene for” breast cancer, and a “gene for” gayness. Many people had begun to believe our genes determine who we become.
This line of thinking should sound familiar to women. Not long ago, we were told that hormones, not sexism, explained why there has never been a U.S. female president (she might start a nuclear war in a fit of PMS). A decade after that came the notion that gender is “hard-wired” into the brain. Not incidentally, these claims were made just when social movements were proving Simone de Beauvoir’s adage that women are not born but made. Now the old determinism is raising its ugly head once again, with genetics. As “non-traditional” families finally bring legitimacy to social parenting, proponents of inheritable genetic modification tell us not only that we can pre-determine the natures of our children, but that cloning is the only means by which gays and lesbians can become real parents. “Real” parental ties, they imply, are biological, genetic.
“Genetic determinism” is not biologically accurate. “It is very unlikely that a simple and directly causal link between genes and most common diseases will ever be found,” writes Richard Horton, editor of the British medical journal The Lancet. If this is true of disease, it is even more true of musicality, optimism, or sexual orientation. The more complex a trait, the less useful genetics are to explain it. Hubbard writes, “The lens of genetics really is one of the narrowest foci to define our biology, not to mention what our social being is about.”
GENETIC MODIFICATION IS NOT A REPRODUCTIVE “CHOICE.”
For feminists, one of the most galling aspects of the debate about human genetic manipulation is the way its proponents have hijacked the language of “choice” to sell its products. IVF clinics and biotech research shouldn’t be regulated, say the companies that run them, because that would impinge on “choice” (for the paying customers, if not for their unsuspecting offspring). The Book of Life is becoming a “catalogue” of “consumer eugenics,” says sociologist Barbara Katz Rothman.
Some ethicists, too, have posited a reproductive “right” to prenatal baby design. People decide whether or not to reproduce based on an expected “package of experiences,” wrote John Robertson, an influential bioethicist, in 1998. “Since the makeup of the packet will determine whether or not they reproduce…some right to choose characteristics, either by negative exclusion or positive selection, should follow as well.” Already, selective abortion is widely accepted after prenatal genetic screening uncovers an “anomaly.” Although some (notably disability rights activists) critique such “negative eugenics,” many people accept this practice for serious medical conditions. In any case, selecting from among a small number of embryos is a far cry from rearranging the DNA of a future child to achieve some preferred traits.
What feminists mean by “choice”—the ability to control fertility with safe and legal birth control and abortion—is far more concrete. It confers existential equality on the female half of the human race, which is why women worldwide have sought it for centuries. But genetic engineering designs in inequality: it will artificially confer heritable advantages only on those who can afford to buy them. Performed prenatally, moreover, it affects the new person without that person’s prior consent and possibly to her physical or emotional detriment. “Ending an unwanted pregnancy is apples, and mucking around with genes is oranges,” says Marcy Darnovsky of the Center for Genetics and Society. “We support abortion rights because we support a right to not have a child—or to have one. But we don’t support a woman’s right to do anything to that child once it’s alive, like abuse it or kill it.” Ironically, as Lisa Handwerker of the National Women’s Health Network has pointed out, anti-choice, anti-GE forces share with GE’s proponents an obsessive focus on the embryo as an independent entity, while they both virtually ignore the pregnant woman and the child she may bear.
BANS ON DANGEROUS GENETIC TECHNOLOGIES DO NOT GIVE FETUSES “RIGHTS.”
Some choice advocates fear that any perceived concern about embryos will cede territory to anti-abortionists, who want full legal protection of embryos and fetuses. U.S. Congressman Henry Waxman reflected this confusion when he said at a Congressional hearing, “I do not believe that the Congress should prohibit potentially life-saving research on genetic cell replication because it accords a cell—a special cell, but only a cell—the same rights and protections as a person.”
But pro-choice opponents of cloning do not propose to give cells rights. Rather, we worry that cloned embryos might be implanted by unscrupulous fertility entrepreneurs into desperate women, where they’ll grow into cloned humans. And from cloning, it is not a big step to designing children.
For legal, political, and philosophical reasons, University of Chicago medical ethicist Mary Mahowald proposes clarifying the pro-choice position. “It does feminist support for abortion no good to confuse life with personhood,” she told me. “We can admit that the embryo is life and therefore afford it respect—the respect, for instance, of not exchanging its genes with those of another cell. But respecting life is not the same as granting rights. Rights are reserved for living persons.”
INDIVIDUAL FREEDOM MUST BE BALANCED WITH SOCIAL JUSTICE.
“We’re against bans,” said a member of a coalition of mainstream reproductive-rights groups, explaining why the coalition was reluctant to join a campaign against human cloning. This reaction is not surprising in the United States, where defense of personal freedom can often trump the public interest.
Women’s liberation means more than personal freedom, though. Rooted in the Left, feminism is a critique of all kinds of domination and therefore a vision of an egalitarian world—racially and economically, as well as sexually.
In the case of species-altering procedures, social justice must prevail over individual “choice.” Arguing for an international ban on reproductive cloning and regulation of related research, Patricia Baird, chair of Canada’s Royal Commission on New Reproductive Technologies, put it this way: “The framework of individual autonomy and reproductive choice is dangerously incomplete, because it leaves out the effects on others and on social systems, and the effects on the child and future generations.” The good news is that good public policy protects individuals too. Baird offered the example of overfishing, which might benefit the fisherman in the short run but deplete the fishery for everyone, including that fisherman, in the long run. Regulation sustains his and his children’s livelihoods. “We all have a stake in the kind of community we live in,” Baird said.
FEMINISTS CAN WORK ALONGSIDE ANTI-ABORTION CONSERVATIVES AGAINST SPECIES-ALTERING PROCEDURES.
“We are repelled by the prospect of cloning human beings…because we intuit and we feel, immediately and without argument, the violation of things that we rightfully hold dear,” wrote Leon Kass, conservative social critic and chair of President Bush’s committee to investigate stem-cell research.
Not every feminist holds dear what Kass holds dear: the “sanctity” of the family based in God-given, “natural” forms of reproduction. Still, Kass sat beside Judy Norsigian and Stuart Newman to testify before the U.S. Congress against cloning.
The genetic engineering debate has made strange bedfellows. But it has also rearranged the political definitions that made those bed-fellows strangers. “Social conservatives believe [genetic engineering] is playing God and therefore unethical, while anti-biotech activists [of the Left] see it as the first step into a brave new world divided by biological castes,” writes social critic Jeremy Rifkin. “Both oppose the emergence of a commercial eugenics civilization.” Others suggest that the new political landscape divides differently, between libertarians and communitarians. Whether of the Left or the Right, the former would support an individual right to choose just about any intervention on one’s own body or one’s offspring, whereas the latter esteem public health and social equality and would reject those interventions, including GE, that endanger them.
Choice activists may at first be Surprised when they find that their anti-cloning and anti-eugenics sentiments are shared by opponents of reproductive rights. But passionate arguments for the same position from historically sworn enemies can only make a legislator, or any citizen, listen up. Feminists need sacrifice no part of the defense of women’s reproductive autonomy when we champion health and social justice for the future human community.
PHOTO (COLOR): Thousands of cloning experiments on mammals have yielded these results so far: for every 3.3 cloned eggs, 1 viable embryo, and for every 35 viable embryos, 1 live-born offspring. Ratio of eggs to live offspring: about 116 to 1. Most of the offspring suffered from grave defects. To get better at cloning will require much more experimentation. To get good at cloning humans, or performing other genetic operations on them, will require experimenting on women, men, and children—and accepting the inevitable failures.
~~~~~~~~
By Judith Levine
Judith Levine has written on women’s issues for Ms., My Generation, New York Woman, Oxygen, and Salon, among others. She is the author of My Enemy, My Love: Women, Men, and the Dilemma of Gender (Anchor Doubleday, 1993), and Harmful to Minors: the Perils of Protecting Children From Sex (University of Minnesota Press, 2002).
THE ROLE OF NORTH AMERICAN WOMEN
Mainstream American women’s and reproductive rights organizations have been slow to understand species-altering technologies as their issues. This hasn’t been true in Europe and the global South, or among indigenous women in North America. In 1992, for instance, European Green Party women discovered a patent application from a U.S. biotech company for a process to synthesize nonhuman “biological active agents” in human mammary glands, from which they’d be secreted in milk and transmitted to nursing infants. To dramatize the commodification of women that lurked in this idea, the women’s propaganda featured the image of a pregnant belly with a bar code emblazoned across it. It was one of the first feminist campaigns against patenting a life form, and it was successful. But if such success is to have any chance of being parlayed into a comprehensive global ban, given the aggressive rush of U.S. industry toward this lucrative new trade, more active intervention will be needed from Americans—and especially from American women.
When proposals to ban human cloning were introduced in the U.S. House of Representatives a year ago, progressive opponents of genetic engineering were only partly pleased. The problem was, the legislation did not come from other progressives, or their friends. Rather, the bills were all sponsored by hard-right Republicans like Florida Congressman David Weldon and Pennsylvania’s James Greenwood, and the bills’ loudest supporters were anti-abortion fundamentalists.
This demanded fast and tricky politicking. The sponsors’ sympathies, showing more tenderness toward blastocysts than toward living women and children, made pro-choice representatives want to run in the other direction. “The problem with the Weldon bill was Dave Weldon,” said Judy Norsigian, executive director of the Boston Women’s Health Collective, after lobbying the House on behalf of that bill. The press fanned moderates’ misgivings by characterizing the debate as one of science versus religion, or of medical progress versus Luddite alarmism.
Last summer, U.S. feminists began to catch up. More than 100 groups and individuals—from the National Women’s Health Network to the National Latina Health Organization, and from disability rights feminist Adrienne Asch to anti-globalization activist Naomi Klein—signed the Boston Women’s Health Collective petition supporting a ban on reproductive cloning and a moratorium on embryo cloning. The leadership of the Health Collective’s executive director was emblematic as well as real: as the prospect of human genetic engineering looms, the title of the feminist classic her group wrote—Our Bodies, Ourselves—assumes more urgent meaning.
________________________________________
.AMA
(American Medical Assoc.)
Reference List
Levine J. WHAT HUMAN GENETIC MODIFICATION MEANS FOR WOMEN. World Watch [serial online]. July 2002;15(4):26. Available from: MasterFILE Complete, Ipswich, MA. Accessed June 26, 2016.
http://web.a.ebscohost.com.libraryaccess.sdmesa.edu/ehost/detail/detail?vid=6&sid=472f2e20-7e02-4da2-8b5f-1ff5826a58db%40sessionmgr4005&hid=4204&bdata=JnNpdGU9ZWhvc3QtbGl2ZQ%3d%3d#AN=7198350&db=f6h

Order from us and get better grades. We are the service you have been looking for.