Vapers Show Chemical Changes in Their Genome Linked to Cancer – Global Health News Wire

This is the latest study from a Keck School of Medicine of USC research team to show e-cigarette users develop cancer-related molecular changes similar to smokers. Image Credit: CASEY CHIN

Biologically important changes in DNA seen in smokers are also being found in people who vape, according to a new study published in the journalEpigenetics.

A team of scientists at the Keck School of Medicine of USC have found people who vape exhibit similar chemical modifications in their overall genome and in parts of their DNA as people who smoke cigarettes.

These specific chemical alterations, also known as epigenetic changes, can cause genes to malfunction and are commonly found in nearly all types of human cancer as well as other serious diseases.

The findings add to a growing list of health concerns associated with vaping, which is perceived by many as a safer alternative to smoking. E-cigarette use has soared among youth, with more than 25 percent of high school students now using the products, according to the CDC.

The new study, led by Ahmad Besaratinia, PhD, associate professor at the Keck School of Medicine of USC, examined a group of people matched for age, gender and race, divided equally into three categories: vapers only, smokers only and a control group of people who neither vaped nor smoked.

Blood was drawn from each of the participants and tested for changes in levels of two specific chemical tags attached to DNA that are known to impact gene activity and/or function. These chemical tags include: (1) methyl groups in a specific DNA sequence, named Long Interspersed Nucleotide Element 1 (LINE-1); and (2) hydroxymethyl groups in the genome overall. Changes in the levels of these chemical tags, which are important for genomic stability and regulation of gene expression, occur in various stages of development, as well as in diseases such as cancer.

Of the 45 study participants, vapers and smokers both showed significant reduction in the levels of both chemical tags compared to the control group. This is the first study to show that vapers, like smokers, have these biologically important changes detectable in their blood cells.

That doesnt mean that these people are going to develop cancer, said Besaratinia. But what we are seeing is that the same changes in chemical tags detectable in tumors from cancer patients are also found in people who vape or smoke, presumably due to exposure to cancer-causing chemicals present in cigarette smoke and, generally at much lower levels, in electronic cigarettes vapor.

This is the newest study Besaratinias team has done on vapers and smokers. Their earlier study published last year (IJMS, 2019) examined changes in gene expression in epithelial cells taken from the mouths of vapers and smokers compared to a control group. In that study, both vapers and smokers showed abnormal gene expression in a large number of genes linked to cancer.

Our new study adds an important piece to that puzzle by demonstrating that epigenetic mechanisms, specifically changes in chemical tags attached to the DNA, may contribute to the abnormal expression of genes in vapers and smokers alike, said Besaratinia.

He and his team plan to continue their research. The next step is to look at the whole genome and identify all the genes targeted by these two chemical changes in vapers versus smokers.

Considering the established role many genes play in human diseases, this investigation should provide invaluable information, which may have immediate public health and policy implications, said Besaratinia. The epidemic of teen vaping and the recent outbreak of vaping-related severe lung injury and deaths in the U.S. underscore the importance of generating scientific evidence on which future regulations for electronic cigarette manufacturing, marketing, and distribution can be based.

Originally posted here:
Vapers Show Chemical Changes in Their Genome Linked to Cancer - Global Health News Wire

How in utero Zika virus infection can lead to microcephaly in newborns: Baylor research – Outbreak News Today

A new study led by researchers at Baylor College of Medicine revealed how in utero Zika virus infection can lead to microcephaly in newborns. The team discovered that the Zika virus protein NS4A disrupts brain growth by hijacking a pathway that regulates the generation of new neurons. The findings point at the possibility of developing therapeutic strategies to prevent microcephaly linked to Zika virus infection. The study appeared Thursday in the journal Developmental Cell.

Patients with rare genetic mutations shed light on how Zika virus causes microcephaly

The current study was initiated when a patient presented with a small brain size at birth and severe abnormalities in brain structures at the Baylor Hopkins Center for Mendelian Genomics (CMG), a center directed by Dr. Jim Lupski, professor of pediatrics, molecular and human genetics at Baylor College of Medicine and attending physician at Texas Childrens Hospital, said Dr. Hugo J. Bellen, professor at Baylor, investigator at the Howard Hughes Medical Institute and Jan and Dan Duncan Neurological Research Institute at Texas Childrens Hospital.

This patient and others in a cohort at CMG had not been infected by Zika virus in utero. They had a genetic defect that caused microcephaly. CMG scientists determined that the ANKLE2 gene was associated with the condition. Interestingly, a few years back the Bellen lab had discovered in the fruit fly model that ANKLE2 gene was associated with neurodevelopmental disorders. Knowing that Zika virus infection in utero can cause microcephaly in newborns, the team explored the possibility that Zika virus was mediating its effects in the brain via ANKLE2.

In a subsequent fruit fly study, the researchers demonstrated that overexpression of Zika protein NS4A causes microcephaly in the flies by inhibiting the function of ANKLE2, a cell cycle regulator that acts by suppressing the activity of VRK1 protein.

Since very little is known about the role of ANKLE2 or VRK1 in brain development, Bellen and his colleagues applied a multidisciplinary approach to tease apart the exact mechanism underlying ANKLE2-associated microcephaly.

The fruit fly helps clarify the mystery

The team found that fruit fly larvae with mutations in ANKLE2 gene had small brains with dramatically fewer neuroblasts brain cell precursors and could not survive into adulthood. Experimental expression of the normal human version of ANKLE2 gene in mutant larvae restored all the defects, establishing the loss of Ankle2 function as the underlying cause.

To understand why ANKLE2 mutants have fewer neuroblasts and significantly smaller brains, we probed deeper into asymmetric cell divisions, a fundamental process that produces and maintains neuroblasts, also called neural stem cells, in the developing brains of flies and humans, said first author Dr. Nichole Link, postdoctoral associate in the Bellen lab.

Asymmetric cell division is an exquisitely regulated process by which neuroblasts produce two different cell types. One is a copy of the neuroblast and the other is a cell programmed to become a different type of cell, such as a neuron or glia.

Proper asymmetric distribution and division of these cells is crucial to normal brain development, as they need to generate a correct number of neurons, produce diverse neuronal lineages and replenish the pool of neuroblasts for further rounds of division.

When flies had reduced levels of Ankle2, key proteins, such as Par complex proteins and Miranda, were misplaced in the neuroblasts of Ankle2 larvae. Moreover, live imaging analysis of these neuroblasts showed many obvious signs of defective or incomplete cell divisions. These observations indicated that Ankle2 is a critical regulator of asymmetric cell divisions, said Link.

Further analyses revealed more details about how Ankle2 regulates asymmetric neuroblast division. They found that Ankle2 protein interacts with VRK1 kinases, and that Ankle2 mutants alter this interaction in ways that disrupt asymmetric cell division.

The Zika connection

Linking our findings to Zika virus-associated microcephaly, we found that expressing Zika virus protein NS4A in flies caused microcephaly by hijacking the Ankle2/VRK1 regulation of asymmetric neuroblast divisions. This offers an explanation to why the severe microcephaly observed in patients with defects in the ANKLE2 and VRK1 genes is strikingly similar to that of infants with in utero Zika virus infection, Link said.

Subscribe to Outbreak News TV

For decades, researchers have been unsuccessful in finding experimental evidence between defects in asymmetric cell divisions and microcephaly in vertebrate models. The current work makes a giant leap in that direction and provides strong evidence that links a single evolutionarily conserved Ankle2/VRK1 pathway as a regulator of asymmetric division of neuroblasts and microcephaly, Bellen said.

Moreover, it shows that irrespective of the nature of the initial triggering event, whether it is a Zika virus infection or congenital mutations, the microcephaly converges on the disruption of Ankle2 and VRK1, making them promising drug targets.

Another important takeaway from this work is that studying a rare disorder (which refers to those resulting from rare disease-causing variations in ANKLE2 or VRK1 genes) originally observed in a single patient can lead to valuable mechanistic insights and open up exciting therapeutic possibilities to solve common human genetic disorders and viral infections.

Originally posted here:
How in utero Zika virus infection can lead to microcephaly in newborns: Baylor research - Outbreak News Today

DNA Nudge app review: can this wristband tell you the best diet for your genes? – The Times

After a super-fast DNA test developed by scientists at Imperial College, Helen Rumbelow trialled their new gadget it lets people choose food to suit their genes

The Times,November 12 2019, 12:01am

Will it be my grandmothers cancer, or the family weakness for Alzheimers that will get me in the end? Our genes contain instructions for our death as well as our life, but they have always played dumb. Until now.

Now I can wear a wristband with my genetic vulnerability for fatal diseases coded into it. Which is by turns futuristic and kind of terrifying. For me, its like shaking hands with my heart attack scheduled for 2050: Nice to get to know you at last!

Weird, but I soon get used to it when I take the wristband shopping. Its the opposite experience to taking a toddler, endlessly pestering for sweeties, to the supermarket. When I aim the tiny scanner of the DNA Nudge wristband

Want to read more?

Subscribe now and get unlimited digital access on web and our smartphone and tablet apps, free for your first month.

Read the original post:
DNA Nudge app review: can this wristband tell you the best diet for your genes? - The Times

She was destined to get early Alzheimer’s, but didn’t. Did a rare mutation protect her – STAT

A woman with a genetic mutation thought to inevitably cause Alzheimers disease in peoples 50s escaped that fate, living into her 70s before she developed mild dementia and researchers think they know why.

In addition to the Alzheimers mutation, they reported on Monday, she has a rare form of a gene best known for producing molecules that help carry cholesterol through the bloodstream. Somehow, the second gene prevented the devastating consequences of the first, a finding that might one day open up new approaches to treating or preventing Alzheimers.

This is an excellent and thought-provoking study, said Dr. Michael Greicius of Stanford University School of Medicine, an expert in Alzheimers genetics who was not involved in the research. He emphasized, however, that because the patients combination of genes is exceedingly uncommon and possibly unique, the study published in Nature Medicine is hypothesis-generating but far from definitive.

advertisement

He and five other Alzheimers researchers cautioned that this is a single case report, not a large study, and the rarity of the womans genetics may well make it impossible to prove that the supposedly protective gene really did keep her from developing early-onset Alzheimers.

The woman (unnamed, to protect her privacy) belongs to a large extended Colombian family. Descended from a Basque couple who migrated to Colombia 300 years ago, roughly 1,200 of its 6,000 living members carry a mutation in a gene called PSEN1, discovered in 1987. The mutation (known as E280A) causes the brain to overproduce the protein fragment beta-amyloid, which forms sticky plaques between neurons and is a diagnostic hallmark (though not necessarily the cause) of Alzheimers.

Because the Colombian family is the largest single group with mutations that cause early-onset Alzheimers half of those with the gene develop mild cognitive impairment by age 44 and dementia by 49 they have been a key part of studies of the disease.

Through one such study, the woman came to the attention of neuropsychologist Yakeel Quiroz of Massachusetts General Hospital. The womans memory and thinking had been basically fine well into her 50s and 60s, her family said. Although brain imaging revealed extremely high levels of amyloid as is expected with PSEN1 only in her 70s did she develop mild cognitive impairment, three decades after relatives who also have the amyloid-superproducing PSEN1 mutation.

In fact, she has more amyloid plaques than relatives whose cognition began crashing in their 40s. She also has relatively low brain levels of tau, also a protein fragment but one that accumulates inside (and kills) neurons. She also has little neurodegeneration.

To figure out how the woman avoided early-onset Alzheimers, Quiroz and her colleagues sequenced her genome. One of her genes, APOE3, was extremely unusual: Both copies (one from her mother and one from her father) carried the rare Christchurch mutation, named for the New Zealand city where it was discovered in 1987. Despite that history, it is found almost exclusively in Latinos; Stanfords Grecius estimates that only about 1 in 100 million people have two copies.

Like every other human, the woman has thousands of other unusual genetic variants. But Quiroz zeroed in on the Christchurch mutation based on an algorithm that ranks variants for their role in particular diseases. Different forms of APOE have long been associated with Alzheimers: APOE4 raises the risk of developing the disease, APOE2 lowers the risk, and APOE3 is neutral (there is no APOE1). We felt confident the Christchurch variant of APOE3 was of interest, Quiroz said.

To test that hunch, she and her team studied how the Christchurch form of the APOE3 molecule interacts with other molecules that play a role in Alzheimers. In lab dishes, the Christchurch form didnt bind as well as ordinary APOE3 to sugar molecules (called heparan sulphate proteoglycans). Those sugars, previous studies showed, are critical enablers of tau, the neuron-killing Alzheimers-related molecule: Bound to APOE, the sugars allow tau to spread from one neuron to another, jumping around the brain in a dance as lethal as glowing embers in a wildfire.

The Christchurch mutation, Quiroz and her team concluded, dampens tau formation and neuronal death even when the brain is awash in amyloid.

If theyre right, it might be possible to prevent or treat Alzheimers through a route very different from removing brain amyloid, as most experimental drugs have (in virtually every case, without success). Instead, antibodies or other molecules that keep APOE from binding to the tau-spreading sugars could reproduce [the] potentially protective effect of the Christchurch mutation, including in people with ordinary genes, Quiroz and her colleagues wrote. That could have a profound impact on the treatment and prevention of Alzheimers disease.

Other scientists werent so sure. The main doubt: This patient, like everyone, has tens of thousands of other rare variants, any one of which might be why she did not develop early-onset Alzheimers as her PSEN1 mutation should have caused.

There are thousands of variants in our genome, said Nikolaos Robakis of the Icahn School of Medicine at Mount Sinai, who discovered one of the first mutations for early-onset Alzheimers. So, from the get-go, its unlikely that this is the one that let the woman escape what would have otherwise been her genetic fate.

One reason for doubt: Having one copy of the Christchurch variant (as seven of 117 members of the patients extended family do) rather than two (as she does) has apparently no benefit, Stanfords Greicius pointed out. All of the seven developed early-onset Alzheimers.

It would have been most convincing to show that while two copies of the Christchurch variant move the age of onset from early 40s to early 70s, one copy had a middling effect, moving the age to the early 50s, he said. But there was no dose effect. That, agreed Robakis, is evidence against the claim that this rare form of APOE3 acts as an anti-Alzheimers talisman.

But even skeptics agreed on one thing: The role of APOE in Alzheimers is vastly understudied. Remedying that could be the Colombian womans most valuable contribution to Alzheimers research.

Go here to see the original:
She was destined to get early Alzheimer's, but didn't. Did a rare mutation protect her - STAT

New therapy to tackle cystic fibrosis highly effective – RTE.ie

A new triple-drug therapy that tackles the genetic causes of cystic fibrosis has been shown to be highly effective in treating the rare life-threatening disorder, scientists reportedfollowing landmark clinical trials.

The finding, 30 years after the gene that causes the disease was identified, was called a "cause for celebration" by Francis Collins, the director of the National Institutes of Health who was part of the team that made the original genetic discovery.

Trikafta is a combination of three drugs that target the CFTR gene responsible for the disease, which affects over 1,000 people in Ireland.

The gene results in the formation of thick mucus build-up in the lungs, digestive tract and other parts of the body.

That in turn results in respiratory and digestive problems, and exposes patients to complications such as infections and diabetes.

Average life expectancy is in the 30s and 40s and was historically even lower before advances in drugs that alleviate symptoms.

The new therapy targets the most common mutation of the CFTR gene, the Phe508del mutation, which represents around 90%of cases.

"The results of a pair of phase 3 clinical trials in the Journal and in a simultaneous publication in the Lancet document impressive benefits," Mr Collins wrote in an editorial for the New England Journal of Medicine.

The two trials examined how much air patients could expel in a forced breath, an established marker of the disease's progression.

In the first trial, mean levels increased by 13.8%compared to a placebo, while in the second trial, mean levels rose 10% from baseline compared to an earlier drug combination.

"This should be a cause for major celebration," said Mr Collins, but he added that more work was needed on patients with other forms of mutations who would not respond to the new therapy.

Beyond that, he added, the best outcome would come when the more than 70,000 people with the disease worldwide do not need drugs because of a permanent cure achieved through gene editing.

The rest is here:
New therapy to tackle cystic fibrosis highly effective - RTE.ie

The Journal of Gene Medicine | RG Impact & Description …

Background/objectives: Selenoprotein P (SeP) is involved in transporting selenium from the liver to target tissues. As SeP confers protection against disease by reducing chronic oxidative stress, this study aims to assess the level of SeP in the serum of patients with metabolic syndrome (MetS) with a history of cardiovascular disease (CVD).Subjects/methods: A cross-sectional study was conducted on 63 and 71 subjects with and without MetS in the presence of documented CVD. All demographic, anthropometric and with cardiometabolic variables (lipids, blood glucose, blood pressure) were assessed. Lifestyle related factors and personal history and familial CVD risk factors were recorded. The expression of SELP in mRNA and protein levels in the serum was measured, and MetS was determined using ATPIII criteria. Binary logistic regression analysis demonstrated MetS and SeP as dependent and independent variables, respectively.Results: Mean of systolic and diastolic blood pressure, triglyceride, HDL-C, fasting blood sugar (FBS), BMI, and waist circumference were higher among subjects with MetS (P = 0.05). Mean of selenium is higher among subjects with MetS whereas the mean of SeP was lower among subjects with MetS (P < 0.001). In the unadjusted model, the SeP had decreased odds for MetS (OR, 0.995; 95% CI, 0.989 to 1.00) (P < 0.04). Furthermore, the association between MetS and SeP levels remained marginally significant even after adjusting for potential confounders such as age, gender, family history, smoking status, and nutrition. SeP and WC has a significant relationship (OR, 0.995; 95% CI, 0.990 to 1.00), (P < 0.033).Conclusions: In conclusion, we demonstrated a significant decrease in circulating SeP levels according to MetS status in patients with documented cardiovascular disease.

View original post here:
The Journal of Gene Medicine | RG Impact & Description ...

Hopkins joins gene cloning project to advance medicine development – Baltimore Sun

Scientists at Johns Hopkins, Rutgers and Harvard universities, as well as the University of Trento in Italy, have created a new technique that allows thousands of genes in a DNA sequence to be cloned at once.

Researchers hope the advance in gene cloning will allow them to more quickly identify markers for diseases and discover new medicines.

Until now genes had to be cloned individually in a time-consuming process. The new molecular method allows thousands of the long DNA strands that make up genes to be isolated and cloned at the same time.

The discovery was published June 26 in Nature Biomedical Engineering.

"Our goal is to make it cheap and easy for any researcher in any field to clone and express the entire set of proteins from any organism," said Ben Larman, an assistant professor of pathology in the Johns Hopkins School of Medicine and the study's co-senior author, in a statement. "Until now, such a prospect was only realistic for high-powered research consortia studying model organisms like fruit flies or mice."

The scientists call their technique for capturing DNA strands that make up genes the LASSO method, for long adapter single-stranded oligonucleotid. They also liken it to capturing cattle with a rope.

The new process speeds up the genes' creation of proteins, which manage cell activity, compared to the old process of cloning individual genes.

To test the method, the scientists sought to capture more than 3,000 DNA strands from the E. coli bacterial genome, commonly used as a model organism, and were successful with most of the targets. They also were able to use the strands to analyze what the gene's proteins do.

"We're very excited about all the potential applications for LASSO cloning," Larman said. "Our hope is that by greatly expanding the number of proteins that can be expressed and screened in parallel, the road to interesting biology and new therapeutic biomolecules will be dramatically shortened for many researchers."

The next step, already underway, is improving the cloning process and building libraries of proteins from DNA samples for use in research, said Biju Parekkadan, an associate professor in the Department of Biomedical Engineering at Rutgers University-New Brunswick.

Funding for the research came from the Shriners Hospitals for Children, the Prostate Cancer Foundation and the National Institutes of Health.

Larman, Parekkadan and a Harvard scientist on the project have sought a patent for the method, which is pending.

meredith.cohn@baltsun.com

See the original post:
Hopkins joins gene cloning project to advance medicine development - Baltimore Sun

What can genetic testing really tell you? – Popular Science

Once difficult and expensive even for the most technologically advanced labs, genetic testing is fast becoming a cheap and easy consumer product. With a little spit and 200 dollars, you can find out your risk for everything from cystic fibrosis to lactose intolerance.

But its important to remember that not all genetic tests are created equal. And even the best clinical genetic test, carried out in a medical lab under a doctor's supervision, isn't perfectgenes are important, but they don't seal your fate.

Genetic tests are diagnostic, so anyone who is curious about their health can get one done. But they're more informative if you think you might be at risk for a genetic disorder.

Heavy-duty genetic tests have been used as a clinical tool for almost half a centurylong before 23andMe and Ancestry.com began offering direct-to-consumer tests. Lets say that many women in your family have had breast cancer. You can get a genetic test to see if you may have inherited an abnormal version of the BRCA gene, known to increase your risk for breast cancer.

Heidi Rehm, associate professor of pathology at Harvard Medical School, is the director of the Laboratory for Molecular Medicine, where patients get tested for diseases that can be traced to specific genetic roots. She says it is most common for people to get tested when they either suspect or know that they have a genetic disease; it may have affected multiple people in their family or they could show symptoms of something widely known to be genetic, like sickle cell anemia. For these people, genetic tests can provide a much-needed explanation for an illness and help doctors determine the best course of treatment. Babies are often tested for genetic diseases, either while they are still fetuses or shortly after birth.

Others get genetic tests if they and their partner both have family histories of an inherited diseaseeven if they dont have the disease themselves. For example, cystic fibrosis is linked to one particular gene, but you have to inherit the abnormal version of the gene from both your parents to get the disease. If you only inherit one copy, you may never knowyou wont display any of the symptoms. But if you and your partner both carry one copy of the faulty gene, your child could still inherit two copies. Genetic tests can forewarn you of that possibility.

But Rehm says there has been a recent trend of healthy people getting tested to predict whether theyll get certain diseases. I do think there are settings where predictive genetic testing is incredibly important and useful, Rehm says; for example, knowing that youre at risk for breast cancer gives you the opportunity for early intervention (remember when Angelina Jolie got a double mastectomy upon finding out she had a mutated BRCA gene?)

But Rehm also points out that genetic tests may not be as straightforward as they seem. For example, some genes are thought to increase risk of getting a certain disease, but it might only happen if you have specific family history, or you might be able to reduce your risk with lifestyle changes. So remember that a genetic test isnt the final verdictthere are other factors at play too.

Not entirelyits scope is limited. For starters, not all diseases are caused by genes. Plenty of conditions stem from environmental and lifestyle factors; they may interact with your genes, but the external factors are the real trigger.

But even if a disease is caused solely by faulty instructions written in your genes, you wont necessarily be able to test for it. Thats because genetic tests are mainly used for diseases that are penetrant, a term that scientists use to describe a strong connection between having a certain gene (or multiple genes) and getting a disease.

Genetic tests are surprisingly simple on the surface. All thats required of you is a small sample of cells, like a blood sample or saliva (which doesnt have DNA itself, but picks up cheek cells during its journey out of your mouth). It get sent to a lab where sequencing machines match up small pieces of synthetic DNA with your DNA to figure out the overall sequence.

Once they have your sequence, geneticists can compare it with "normal" or disease-causing sequences. In the end, they might give you a yes or no answer, or sometimes youll get a probabilitya measure of how much your genes increase your risk of developing the disease. Then, its up to your doctor to figure out what these genes (in combination with your lifestyle, family history and other risk factors) mean for your health.

With penetrant diseases, theres a very, very high ability to explain the disease, Rehm says. For example, the breast cancer-related gene BRCA1 can give you a 60 percent chance of getting breast cancer (in Jolies case, with her family history, the risk was 87 percent.)

This makes genetic tests better at detecting so-called rare diseases, says Steven Schrodi, associate research scientist at the Marshfield Clinic Research Institutes Center for Human Genetics, but theyre less useful when it comes to more common diseases, like heart disease or diabetes. Genetics can increase your likelihood of getting these disease, but scientists still dont know quite how much. Part of the problem is that there may be dozens or hundreds of genes responsible for these diseases, Schrodi says.

We have an incomplete understanding of why people get diseases, Schrodi says. A large part of it hinges on how we define diseases. Perhaps physicians have inadvertently combined multiple diseases together into a single entity.

Consumer genetic teststhe ones where you send in samples from homesometimes claim to test for these more complex traits, but be careful: Their results might not be very medically relevant, Rehm says. If they tell you that your genes make you twice as likely to develop diabetes, for example, that's a marginal increase that doesn't significantly affect your risk, especially when you take into account lifestyle factors.

Genes do seem to play a role in determining lifespan. After all, some family reunions stretch from great-great-grandparents all the way down to infants. Scientists have studied centenarianspeople who lived to be 100 years oldand found that people with certain versions of genes involved in repairing DNA tend to live longer.

This makes sense because aging leaves its mark on your DNA. Environmental factors can damage DNA, and even the routine chore of replicating cells can introduce errors as the three billion units of your DNA are copied over and over. Long-lived individuals have different sequences that seem to make their cells better at keeping DNA in mint condition.

But figuring out your expiration date is more complex than just testing for a few genes, says Jan Vijg, professor of genetics at Albert Einstein College of Medicine. In theory, you could design a test that looks at specific genes that might measure your risk for developing Alzheimers Disease or other age-related diseases, or your risk for aging quickly. To some extent, yes: Biomarkers will tell you something about your chances of living a long life, Vijg says. Still, that will only work if you live a careful life. And that means no accidents, infections, or cancers.

Aging also affects the exposed ends of your DNA, called "telomeres." DNA is stored as chromosomes, those X-like structures that you may have seen in biology textbooks. The most vulnerable parts of the chromosome are the chromosomes tips, which get shorter as you age because they arent properly replicated. But while telomere length might let you compare your DNA now with your DNA from a decade ago, you cant compare your own telomeres with other peoples telomeres. Theres a lot of variation between individuals, Vijg says. Some of us are just old souls (on the genomic level, that is.)

The methylation test, which looks at how the presence of small chemical groups attached to your DNA changes as you age, might be a better bet. A study at UCLA showed that changes were slower in longer-lived people. But Vijg is hesitant: I would not put my hopes on that as a marker to predict when exactly youre going to die.

For now, just enjoy your life, because you cant predict death. And if you decide to unlock the secrets of your DNA with an at-home test, don't take those results for more than their worth.

The rest is here:
What can genetic testing really tell you? - Popular Science

Kashmiri doctor helps gene editing of human embryos in US – Hindustan Times

For the first time, genetically modified human embryos have been developed in the US and Kashmir-born doctor Sanjeev Kaul has played a lead role in this breakthrough.

Scientists have now demonstrated an effective way of using a gene-editing tool to correct a disease-causing gene mutation in human embryos and stop it from passing to future generations.

Though this is not a full-fledged start of a revolution of having designer babies, the first steps, however, have been laid. China attempted this earlier.

A team of scientists has altered human embryos using a new technique called CRISPR CAS9 that edits genes and in this case it helped remove a fatal mutation that leads to heart attacks.

This now opens up an ethical Pandoras Box if germline repairs and enhancements may become a thing in vogue.

As of now, the human embryos were not implanted in humans. But this now opens up exciting prospects of the world having designer babies soon.

The research published in British journal Nature shows the first genetically modified human embryos made in America.

A team of South Korean, Chinese and American scientists has identified how they could edit out a faulty gene that causes heart attacks in later life due to the thickening of heart walls.

One of the team members is Dr Kaul, who was born in Kashmir, studied in New Delhi and later immigrated to America.

Although the rare heart mutation affects men and women of all ages, it is a common cause of sudden cardiac arrest in young people, and it could be eliminated in one generation in a particular family, said co-author Kaul, a professor of medicine (cardiovascular medicine) in the OHSU School of Medicine and director of the OHSU Knight Cardiovascular Institute.

Thanks to advances in stem cell technologies and gene editing, we are finally starting to address disease-causing mutations that impact potentially millions of people, says Juan Carlos Izpisua Belmonte, a professor in California-based Salk Institutes Gene Expression Laboratory and a corresponding author of the paper.

Gene editing is still in its infancy so even though this preliminary effort was found to be safe and effective, it is crucial that we continue to proceed with the utmost caution, paying the highest attention to ethical considerations.

CRISPR CAS9 or Clustered Regularly Interspaced Short Palindromic Repeats is a kind of a precise molecular scissor the scientists use to edit faulty genes.

Only selected healthy embryos were allowed to grow further that too only for a few days. The embryos were not implanted in humans.

The big step forward is that a higher percentage embryos were found to have been repaired in this American experiment than earlier attempts.

CRISPR holds promise for correcting mutations in the human genome to prevent genetic disease. Using an enzyme called Cas9, it is possible to snip a specific target sequence on a mutant gene.

The new study found that human embryos effectively repair these breaks in the mutant gene using the normal copy of this gene from a second parent as a template.

The resulting embryos contain now repaired, mutation-free copies of this gene.

The technique already has been used in animals for generating mutant models; however, the new study is the first to demonstrate that technique can be used in human embryos to convert mutant genes back to normal.

The study also demonstrated a way for overcoming a crucial problem in genome editing in embryos known as mosaicism.

Mosaicism refers to an outcome when not all cells in a multicellular embryo get repaired and some cells still carry a mutation.

Every generation on would carry this repair because we have removed the disease-causing gene variant from that familys lineage, said senior author Shoukhrat Mitalipov, PhD, who directs the Center for Embryonic Cell and Gene Therapy at Oregon Health and Science University (OHSU), in Portland, Oregon, USA.

By using this technique, it is possible to reduce the burden of this heritable disease on the family and eventually the human population.

The study provides new insight into a technique that could apply to thousands of inherited genetic disorders affecting millions of people worldwide.

The gene-editing technique described in this study, done in concert with in vitro fertilisation, could provide a new avenue for people with known heritable disease-causing genetic mutations to eliminate the risk of passing the disease to their children.

If proven safe, this technique could potentially decrease the number of cycles needed for people trying to have children free of genetic disease, said co-author Paula Amato, associate professor of obstetrics and gynaecology in the OHSU School of Medicine.

Designer babies could be in the offing.

Our results demonstrate the great potential of embryonic gene editing, but we must continue to realistically assess the risks as well as the benefits, adds Belmonte.

In this landmark study, the researchers worked with healthy donated human oocytes and sperm carrying the genetic mutation that causes cardiomyopathy or the thickening of heart walls.

Embryos created in this study were used to answer pre- clinical questions about safety and effectiveness. The study noted that genome editing approaches must be further optimised before moving to clinical trials.

This research significantly advances scientific understanding of the procedures that would be necessary to ensure the safety and efficacy of germline gene correction, said Daniel Dorsa, senior vice president for research at OHSU.

The ethical considerations of moving this technology to clinical trials are complex and deserve significant public engagement before we can answer the broader question of whether its in humanitys interest to alter human genes for future generations.

Existing ethical guidelines did not permit the team to implant the genetically modified human embryos into women.

See the rest here:
Kashmiri doctor helps gene editing of human embryos in US - Hindustan Times

UK’s chief medical officer calls for gene testing revolution in cancer treatment – Daily Nation

Saturday July 8 2017

Kenyans mark World Cancer Day on February 4, 2016 in Eldoret town. Tiny errors in DNA code can lead to cancer and other illnesses. PHOTO | JARED NYATAYA | NATION MEDIA GROUP

A revolution in the search for cancer treatments has been proposed by Englands chief medical officer.

Prof Sally Davies wants gene-testing to be introduced on a routine basis.

I want the National Health Service to be offering genomic medicine, that means diagnosis of our genes, to patients where they can possibly benefit, she said.

GENETIC TESTS Testing, she said, should be standard across cancer care as well as some other areas of medicine, including rare diseases and infections.

Doctors are already using genetic tests to identify and better treat different strains of the infectious disease, tuberculosis.

Humans have about 20,000 genes, bits of DNA code or instructions that control how our bodies work.

Tiny errors in this code can lead to cancer and other illnesses.

Gene-screening can reveal these errors by comparing tumour and normal DNA samples from the patient.

Professor Davies says in about two-thirds of cases, this information can improve their diagnosis and care.

Doctors can tailor treatments to the individual, picking the drugs most likely to be effective.

Currently, genetic testing in England is done at 25 regional laboratories, as well as some other small centres.

Professor Davies wants to centralise the service and set up a national network to ensure equal access to the testing across the country.

She said one hurdle could be doctors themselves, who dont like change.

Patients should persuade them to move from a local to a national service. *** Joe Furness was in Newcastle upon Tyne when he was invited to a party in London.

A three-hour, one-way train trip would cost him 78.50 (Sh10,517) and a plane flight 106, but Joe, aged 21, is a poor student and didnt have much money.

What he did have however was time. So Joe decided to take a detour via Spain.

CAR HIRE Flying from Newcastle to the Spanish island of Menorca cost him 16.00.

There he hired a car for 7.50 and spent the night in it, while sipping a 4.50 cocktail.

Next morning he flew to London for 11.00, joined the party, then grabbed a lift home with a pal afterwards.

Total cost of 39 was a saving of 39.50 on a train journey from Newcastle and 67 on a flight.

Distance travelled was 2,350 miles, against 290 miles from the North to London. *** Bradley Lowery is a six-year-old boy who won the hearts of the nation by campaigning for his beloved Sunderland Football Club and for its top scorer, Jermaine Defoe.

TV film of Defoe holding a smiling Bradley in his arms before a recent game appeared on nationwide television.

What everyone knows, of course, is that Bradley is dying from the childhood cancer neuroblastoma.

A fund-raising campaign raised money for him and will be used for other sick children when Bradley dies.

Now it seems fraudsters have been setting up pages on the internet claiming to be collecting for the boys cause.

His family have warned against them. Please be vigilant, they said in a message on Facebook.

You have to wonder, how low can some people stoop?

*** Some 400 plastic bottles are sold per second in this country and millions end up, along with other garbage, in the worlds oceans.

In fact, scientists calculate that by 2050, the oceans will contain more plastic by weight than fish.

The opposition Labour party is pressing for the introduction of a money-back return scheme, which has been introduced in many other countries and has proved successful in reducing the scale of littering.

You pay a bit extra for your drink but you get it back if you return the bottle, which the drinks company then recycles.

Coca-Cola, among others, is backing the idea. *** Famous one-liners:

Doctors recommend eight glasses of water per day. Why does this seem impossible when eight glasses of beer is so easy? Anonymous.

If you want to know what God thinks of money, look at the people he gave it to. American writer Dorothy Parker.

The two most beautiful words in the English language are Cheque enclosed. Dorothy Parker.

PLAGIARISM I asked God for a bike but I know He doesnt work that way, so I stole a bike and asked for forgiveness. Internet.

I wouldnt say I was the best football manager in the business, but I was in the top one. Brian Clough, British football manager.

To steal ideas from one person is plagiarism; to steal from many is research. Anonymous

England and America are two countries separated by a common language. Irish writer George Bernard Shaw.

If I agreed with you, then we would both be wrong. Internet.

Interior Cabinet Secretary collapsed in his house and was rushed to hospital by family and

He is becoming the Presidents go-to-guy when things need to be fixed.

See original here:
UK's chief medical officer calls for gene testing revolution in cancer treatment - Daily Nation

In northern humans, evolution favored shorter bones but with a painful trade-off – Stanford Medical Center Report

Many people think of osteoarthritis as a kind of wear-and-tear disease, but theres clearly a genetic component at work here as well.

The researchers were studying a gene called GDF5 that Kingsleys laboratory first linked to skeletal growth in the early 1990s. GDF5 is involved in bone growth and joint formation, and mutations in the coding portion of the gene have been shown to cause malformations in leg-bone structure in mice. In humans, GDF5 mutations are associated with shorter stature and joint problems; in particular, two nucleotide changes immediately upstream of the gene have been strongly associated with a 1.2- to 1.8-fold increase in the risk of osteoarthritis.

In the new study, the researchers were interested in learning more about how the DNA sequences surrounding GDF5 might affect the genes expression. Often, these noncoding sequences contain key regulatory regions known as promoters and enhancers. Capellini, Chen and Cao were able to identify a previously unknown enhancer region they termed GROW1, which is several thousand nucleotides downstream of GDF5.

When the researchers analyzed the sequence of GROW1 in the 1,000 Genomes Project database, which collects and compares sequences from many human populations around the globe, they identified a single nucleotide change that is highly prevalent in Europeans and Asians but that rarely occurs in Africans. When they introduced this nucleotide change into laboratory mice, they found that it decreased the activity of GDF5 in the growth plates of the long bones of fetal mice.

Further research showed that this nucleotide change has been repeatedly favored during human evolution. Modern humans migrated from Africa between 50,000 and 100,000 years ago. But they werent the first to leave the continent. Neanderthals and Denisovans moved north into Europe and Asia about 600,000 years ago. Interestingly, the researchers found that the same GROW1 variant was found in the DNA of both ancient and modern humans in Europe and Asia.

However, theres a dark side to this stocky, hardy body type: The GDF5 variant that reduces bone length comes hand-in-hand with the two upstream nucleotide changes known to confer an increased risk for osteoarthritis.

Its clear that the genetic machinery around a gene can have a dramatic impact on how it works, said Capellini. The variant that decreases height is lowering the activity ofGDF5in the growth plates of the bone. Interestingly, the region that harbors this variant is closely linked to other mutations that affect GDF5 activity in the joints, increasing the risk of osteoarthritis in the knee and hip.

The potential medical impact of the finding is very interesting because so many people are affected, said Kingsley. This is an incredibly prevalent, and ancient, variant. Many people think of osteoarthritis as a kind of wear-and-tear disease, but theres clearly a genetic component at work here as well. Now weve shown that positive evolutionary selection has given rise to one of the most common height variants and arthritis risk factors known in human populations.

A researcher from the University of Waterloo in Ontario, Canada, also contributed to the study.

The research was supported by the National Sciences and Engineering Research Council of Canada, the Arthritis Foundation, the National Institutes of Health (grant AR42236), the Howard Hughes Medical Institute, the Milton Fund of Harvard, the China Scholarship Council and the Jason S. Bailey Fund of Harvard.

Stanfords Department of Developmental Biology also supported the work.

Originally posted here:
In northern humans, evolution favored shorter bones but with a painful trade-off - Stanford Medical Center Report

Debate Rages Over Safety Of CRISPR Gene-Editing Technique … – IFLScience

In the last few years, CRISPR the precise and easily available gene-editing technique has gone from strength to strength. Right now, its more potential than practical, but its uses are almost countless. From removing genetic diseases from babies before they are born to causing cancerous cells to self-destruct, its more likely than not it will rapidly become a key tenet of modern medicine.

The technique is being used in a massive number of trials right now, and a few of them have highlighted that its far from fully understood and far from being perfected. One particularly controversial study released back in May claimed that CRISPR was in fact dangerous, but now another study, currently sitting on the bioRxiv pre-print server, has dismissed this research as flawed.

The original study looked at the effect of using CRISPR in mice, and found that it introduced a large number of previously unknown mutations into their genome. The mice were blind, and the gene-editing technique was used to effectively cure them of this affliction.

While it was successful, it also appeared to inadvertently lead to mutations elsewhere at least 1,500 of them, in fact. Of these, just over 100 mutations were more serious, involving not just DNA base pair changes, but full deletions or insertions of new segments of DNA.

What is CRISPR? Wired via YouTube

Although there were no physiological side effects, unwanted mutations are always a cause for concern. As a result, the authors concluded that CRISPR has the potential to be quite hazardous to the normal operation of biological systems.

When this study was released, it caused quite a stir. Similar studies had only shown a handful of changes, which turned out to be harmless. People were quick to point out that the study was very small, involving just three mice, and that the changes made elsewhere on their genomes could easily be attributed to normal genetic mutations that take place all the time.

Now, a Harvard University-led team have suggested that these unwanted mutations have a far more innocuous explanation.

Apart from the fact that there is no direct line of evidence demonstrating that CRISPR caused the mutations, the team note that the mice used in the study were very closely related. This means that whatever mutations one had, the other would have had too.

Combined with the fact that the mutations were found nowhere near the actual gene-editing site, the authors conclude that, once again, CRISPR is probably safe after all. Plenty more trials are needed to verify this, of course but the point here is that this study raised a massive red flag when it wasnt justified in doing so.

[H/T: New Scientist]

Read more:
Debate Rages Over Safety Of CRISPR Gene-Editing Technique ... - IFLScience

Heres How Coronavirus Tests Workand Who Offers Them – Scientific American

As the new coronavirus explodes in cities across the U.S., public health agencies and hospitals are making testingwhich was initially plagued by significant shortagesincreasingly available. And biotech companies are ramping up production of test kitsbut states are still struggling to meet demand.

COVID-19 testing is currently available in every U.S. state, as well as Washington, D.C., Guam and Puerto Rico, according to the U.S. Centers for Disease Control and Prevention. As of Tuesday, 92 U.S. public health laboratories had completed the CDCs verification process and were offering tests. The CDCs own labs have tested 4,654 specimens, and public health laboratories have tested 98,576. A New York Timesdatabase reports that at least 75,178 cases had been confirmed as positive by lab tests as of Thursday afternoon.

Testing availability varies by state. A sampling of state public health agencies in New York, California and Texas all referred Scientific Americans questions about such availability, as well as about where patients should go to get tested, to their respective coronavirus information pages. But the basic process is the same nationwide.

The first step in any coronavirus test is collecting a sample. Doing so involves placing a sterile swab at the back of a patients nasal passage, where it connects to the throat via the nasopharynx, for several seconds to absorb secretions. Scott Wesley Long, a clinical microbiologist who directs Houston Methodist Hospitals diagnostic microbiology lab, says the swab is thinless than three millimeters in diameter at its tip. Once you place it in the back of the throat, its uncomfortable, but you can still breathe and talk, he says. Its not as bad as it looks. After a sample is collected, the swab goes into a liquid-filled tube for transport.

Shortages of swabs and reagents for collection kits were among the several roadblocks that stymied public health agencies ability to perform widespread testing in recent weeks, according to David Harris, who directs the biorepository at the University of Arizona. Harris says the university produced 1,600 generic collection kits last weekend to distribute to local health providers free of charge to help fill the gap, adding that all such kits are pretty much the same. The university plans to produce thousands more until availability begins to meet demand.

Recently, two health start-ups began marketing at-home collection kits that they claimed would allow patients to take their own samples and send them to labs for testing. The Food and Drug Administration quickly issued a warning that no such tests have been authorized, and the companies halted sales of the kits, which had been priced at about $170 to $180 apiece.

To determine whether a nasopharyngeal sample is positive for the coronavirus, biotechnicians use a technique known as reverse transcriptase polymerase chain reaction, or RT-PCR. The World Health Organizations and CDCs test kits both use this method, as do all of the kits the latter has approved to date.

Theres a lot of hands-on work involved in performing RT-PCR tests, Long says. First, a technician extracts viral genetic material called RNAif it is presentfrom the sample and uses it to produce a complimentary strand of DNA that the RT-PCR technique amplifies, or makes thousands of copies of, to get a measurable result. The primary difference from one kit to another is which coronavirus genes each test targets. CDC-approved kits target regions on a gene that codes for the protein that makes the viruss nucleocapsid, an envelope that houses its RNA. The biotechnology companies Roche Diagnostics, LabCorp and Thermo Fisher Scientific are among the top suppliers of commercial coronavirus RT-PCR kits.

Stephanie Caccomo, a spokesperson for the FDA, says the positive predictive value, or likelihood a positive test result correctly reflects that a patient has COVID-19, depends on how widespread the disease isand that situation is changing quickly. Based on what is known about the pathophysiology of COVID-19, the data provided and our previous experience with respiratory pathogen tests, the false-positive rate for authorized tests is likely to be very low, and the true-positive rate is likely to be high, Caccomo says. However, performance characteristics are specific to each test. And there is a risk of a false-negative result if the sample is not taken correctly, says Vincent Racaniello, a professor of microbiology and immunology at Columbia University. This possibility could explain why people recovering from the disease sometimes test negative initially and then positive later, he says.

Most RT-PCR tests take anywhere from a few hours to a few days to process, but the FDA recently began granting emergency use authorization (EUA) to rapid diagnostic PCR tests that manufacturers say can deliver results in less than an hour. The authorization allows medical devices that have not yet been approved by the agency to be used during public health emergencies. Caccomo says the FDA conducts rolling reviews of diagnostic-test-validation data provided by laboratories. This has enabled authorization of most EUA requests within a few days, she adds.

On Saturday Cepheid, a Silicon Valleybased molecular diagnostics company, said the FDA had granted it authorization for a COVID-19 test that can deliver results in about 45 minutes. And on Tuesday Mesa Biotech in San Diego announced it had received the go-ahead for a handheld test kit that Hong Cai, the companys CEO, says can deliver results at bedside in about half an hour. Cai says the tests will begin shipping this week to several hospitals and that her company has tens of thousands of units ready to go, adding that Mesa is planning to triple its production capacity.

Another approach relies on identifying antibodies to the coronavirus (SARS-CoV-2) in a patients bloodstream to determine whether that person previously had COVID-19. Florian Krammer, a microbiologist at the Icahn School of Medicine at Mount Sinai, recently developed one of these tests, which is described in a preprint study posted last week on medRxiv. This is not a test for [ongoing] infections, he says. It basically looks for antibodies after the fact, after you had an infection. Like other serological, or antibody-based, diagnostic assays, it uses an enzyme-linked immunosorbent assay (ELISA), which employs a portion of the target virus to find antibodies. Although serological tests are not useful for quickly identifying whether a patient currently has COVID-19, Krammer says they can help researchers understand how humans produce antibodies to the virus.

Patients with severe cases of COVID-19 may be able to be treated with blood plasma from people who have developed antibodies and are immune to the disease in what is effectively an antibody-transfer operation. But to do that, clinicians first have to screen donors to determine who has a strong immune response to the virus, Krammer says. His lab is currently running a serological assay to find potential plasma donors. On Tuesday the FDA approved the emergency use of plasma for critically ill coronavirus patients.

Additionally, serological tests can also help determine if a person has been infected whether or not the individual had symptomssomething an RNA test kit cannot do after the fact, because it only looks for the virus itself. That means serological tests could be used to survey a population to determine how widespread infection rates were. It also could allow public health agencies to figure out who is already immune to COVID-19. So if you would roll this out on a very wide scale, you could potentially identify everybody who is immune and then ask them to go back to their regular life and go back to work, Krammer says. This approach could be especially useful for health care providers who are working with COVID-19 patients. They might feel much more comfortable working with those patients, [knowing] that they can't get sick anymore, knowing that they cant pass on the virus to others, he says.

Read more about the coronavirus outbreakhere.

Original post:
Heres How Coronavirus Tests Workand Who Offers Them - Scientific American

Gene Cloning Just Became A Thousand Times Easier – Wall Street Pit

Presently, the process of DNA sequencing to discover a genes function is a bit time-consuming because it has to be done one gene at a time. But with the invention of a team of researchers from Rutgers University-New Brunswick, Johns Hopkins University and Harvard Medical School, that process can now be considerably shortened as they have found a way to simultaneously clone thousands of genes.

The new technique is called LASSO cloning LASSO is an acronym for long-adapter single-strand oligonucleotide and through it, isolating thousands of DNA sequences can now be done at the same time. As a result, the new technology can supposedly expedite the creation of proteins the end product of genes which can in turn lead to faster discovery of new medicines and biomarkers for scores of diseases.

The technique is considered as quite an improvement over an existing method known as molecular inversion probes (MIPs), which can only capture about 200 bases of DNA. Raising this number to a thousand is especially significant because the typical size of a genes protein-coding sequence can reach up to thousands of DNA base pairs.

The technology works through a tool the team refers to as a LASSO probe. Using a number of these probes enables grabbing of target DNA sequences, similar with how a lasso (a rope) is used to round up cattle. What makes this special is the fact that with a single effort, the technique can grab thousands of DNA sequences at the same time.

In their proof-of-concept study, the team used their LASSO probes to simultaneously capture over 3,000 E. coli bacteria DNA fragments. They were successful in capturing around 95% of their gene targets. More importantly, the sequences were captured in such a way that allowed the researchers to observe and evaluate what the expressed proteins do.

As an added bonus, the team also helped solved a perennial problem in the genome sequencing field. As it is, genetic sequencing currently involves sequencing small fragments of DNA one at a time, then overlaying them in order to map out the entire genome. This doesnt just take time; its also harder to interpret and kind of prone to errors. With the LASSO probes, sequencing long fragments has now been made possible, which also translates to a faster and more accurate gene sequencing process.

As one of the researchers, Biju Parekkadan of the Rutgers University-New Brunswick, said: We think that the rapid, affordable, and high-throughput cloning of proteins and other genetic elements will greatly accelerate biological research to discover functions of molecules encoded by genomes and match the pace at which new genome sequencing data is coming out.

Going forward, the team is now working on improving the cloning process, building libraries and discovering the therapeutic proteins found in our genes.

The research was recently published in the journal Nature Biomedical Engineering.

Read more here:
Gene Cloning Just Became A Thousand Times Easier - Wall Street Pit

AZ adds gene silencing tech to pipeline with Silence Therapeutics deal – – pharmaphorum

AstraZeneca has announced a collaboration with UK biotech Silence Therapeutics to develop gene silencing drugs for cardiovascular, renal, metabolic and respiratory diseases.

The deal could see AZ adding small interfering RNA (siRNA) technology to its pipeline in one of its main areas of research.

Silence Therapeutics says its technology can selectively inhibit any gene in the genome, specifically silencing the production of disease-causing proteins.

The UK biotech says it has achieved an additional level of accuracy by delivering its therapeutic RNA molecules exclusively to target cells.

siRNA is a technology that showed much promise after biologists Andrew Fire and Craig Mello received the Nobel Prize in Physiology or Medicine for discovering the technology in 2006.

But in 2010 it became apparent that it was harder to convert into a working therapy because of the challenge of delivering therapeutic RNA molecules to target tissues and big pharma quickly lost interest.

But companies like Silence have managed to overcome this hurdle and its rival Alnylam made history in 2018 when its siRNA drug Onpattro was approved by the FDA to treat hereditary transthyretin (hATTR) amyloidosis, which causes the build-up of amyloid protein in nerves and organs.

AZ will pay $60 million up front and invest $20 million in Silence, and will pay up to $400 million in milestone payments plus tiered royalties.

The companies expect to work on five targets within the first three years of the collaboration, and AstraZeneca has the option to extend it to a further five targets.

Silence has technology that can inhibit liver-expressed gene targets and the companies will collaborate to find siRNA molecules to other tissues including the heart, kidney and lung.

The UK biotech will design SiRNA molecules against gene targets selected by AZ, and will manufacture material to support toxicology and phase 1 clinical studies.

AZ and Silence will collaborate during the discovery phase, while AZ will lead clinical trials and marketing.

Silence will have the option to negotiate for co-development of two drugs of their choice starting from phase 2.

AZ will pay an option fee of $10 million for each selected target when a drug candidate is nominated, and Silence could receive up to $140 million in development milestones, and up to $250 million in market milestones.

Link:
AZ adds gene silencing tech to pipeline with Silence Therapeutics deal - - pharmaphorum

Alnylam Pharmaceuticals and Gen Sign Distribution Agreement in Turkey for ONPATTRO (patisiran), the First-in-Class Gene-Silencing RNAi Therapeutic -…

Alnylam Pharmaceuticals, Inc. (Nasdaq: ALNY), the leading RNAi therapeutics company, and Gen, a GMP-certified pharmaceutical company specializing in rare diseases, today announced an exclusive Distribution Agreement for ONPATTRO, a first-in-class RNAi therapeutic for the treatment of hATTR amyloidosis in adults with Stage 1 or Stage 2 polyneuropathy.

"Our partnership with Gen enables us to extend access to ONPATTRO to patients suffering from hereditary ATTR (hATTR) amyloidosis with polyneuropathy in Turkey where we currently dont have a presence," said Brendan Martin, Vice President and Acting Head of Canada, Europe, Middle East and Africa, Alnylam Pharmaceuticals. "There are a significant number of patients in Turkey who urgently need new treatment options and we are delighted to partner with Gen to bring ONPATTRO to those in need."

Abidin Glms, CEO of Gen stated: "We are proud of our reputation as one of Turkey's leading specialty pharmaceutical companies and are excited to have partnered with Alnylam. Through collaborations with leading international companies, we aim to bring innovative medicines to patients in Turkey in the fastest and most reliable way possible."

Patients in Turkey were among those who participated in the randomized, double-blind, placebo-controlled, global Phase 3 APOLLO study, the largest-ever study in hATTR amyloidosis patients with polyneuropathy, which led to the approval of ONPATTRO in the U.S. and EU in 2018.

About ONPATTRO (patisiran)

ONPATTRO is an RNAi therapeutic that was approved in the United States and Canada for the treatment of the polyneuropathy of hATTR amyloidosis in adults. ONPATTRO is also approved in the European Union, Switzerland and Brazil for the treatment of hATTR amyloidosis in adults with Stage 1 or Stage 2 polyneuropathy, and in Japan for the treatment of hATTR amyloidosis with polyneuropathy. Based on Nobel Prize-winning science, ONPATTRO is an intravenously administered RNAi therapeutic targeting transthyretin (TTR). It is designed to target and silence TTR messenger RNA, thereby blocking the production of TTR protein before it is made. ONPATTRO blocks the production of TTR in the liver, reducing its accumulation in the bodys tissues in order to halt or slow down the progression of the polyneuropathy associated with the disease. For more information about ONPATTRO, visit ONPATTRO.com.

Important Safety Information (ISI) for ONPATTRO

Infusion-Related Reactions

Infusion-related reactions (IRRs) have been observed in patients treated with patisiran. In a controlled clinical study, 19% of patisiran-treated patients experienced IRRs, compared to 9% of placebo-treated patients. The most common symptoms of IRRs with patisiran were flushing, back pain, nausea, abdominal pain, dyspnoea, and headache. Hypotension, which may include syncope, has also been reported as a symptom of IRRs.

To reduce the risk of IRRs, patients should receive premedication with a corticosteroid, paracetamol, and antihistamines (H1 and H2 blockers) at least 60 minutes prior to patisiran infusion. Monitor patients during the infusion for signs and symptoms of IRRs. If an IRR occurs, consider slowing or interrupting the infusion and instituting medical management as clinically indicated. If the infusion is interrupted, consider resuming at a slower infusion rate only if symptoms have resolved. In the case of a serious or life-threatening IRR, the infusion should be discontinued and not resumed.

Reduced Serum Vitamin A Levels and Recommended Supplementation

Patisiran treatment leads to a decrease in serum vitamin A levels. Patients receiving patisiran should take oral supplementation of approximately 2500 IU vitamin A per day to reduce the potential risk of ocular toxicity due to vitamin A deficiency. Doses higher than 2500 IU vitamin A per day should not be given to try to achieve normal serum vitamin A levels during treatment with patisiran, as serum levels do not reflect the total vitamin A in the body. Patients should be referred to an ophthalmologist if they develop ocular symptoms suggestive of vitamin A deficiency (e.g. including reduced night vision or night blindness, persistent dry eyes, eye inflammation, corneal inflammation or ulceration, corneal thickening or corneal perforation).

Story continues

Adverse Reactions

The most common adverse reactions that occurred in patients treated with patisiran were peripheral oedema (30%) and infusion-related reactions (19%).

About RNAi

RNAi (RNA interference) is a natural cellular process of gene silencing that represents one of the most promising and rapidly advancing frontiers in biology and drug development today. Its discovery has been heralded as "a major scientific breakthrough that happens once every decade or so," and was recognized with the award of the 2006 Nobel Prize for Physiology or Medicine. By harnessing the natural biological process of RNAi occurring in our cells, a new class of medicines, known as RNAi therapeutics, is now a reality. Small interfering RNA (siRNA), the molecules that mediate RNAi and comprise Alnylams RNAi therapeutic platform, function upstream of todays medicines by potently silencing messenger RNA (mRNA) the genetic precursors that encode for disease-causing proteins, thus preventing them from being made. This is a revolutionary approach with the potential to transform the care of patients with genetic and other diseases.

About Alnylam

Alnylam (Nasdaq: ALNY) is leading the translation of RNA interference (RNAi) into a whole new class of innovative medicines with the potential to transform the lives of people afflicted with rare genetic, cardio-metabolic, hepatic infectious, and central nervous system (CNS)/ocular diseases. Based on Nobel Prize-winning science, RNAi therapeutics represent a powerful, clinically validated approach for the treatment of a wide range of severe and debilitating diseases. Founded in 2002, Alnylam is delivering on a bold vision to turn scientific possibility into reality, with a robust RNAi therapeutics platform. Alnylams commercial RNAi therapeutic products are ONPATTRO (patisiran), approved in the U.S., EU, Canada, Japan, Brazil and Switzerland, and GIVLAARI (givosiran), approved in the U.S and the EU. Alnylam has a deep pipeline of investigational medicines, including five product candidates that are in late-stage development. Alnylam is executing on its "Alnylam 2020" strategy of building a multi-product, commercial-stage biopharmaceutical company with a sustainable pipeline of RNAi-based medicines to address the needs of patients who have limited or inadequate treatment options. Alnylam is headquartered in Cambridge, MA.

About Gen

Gen is the fastest growing pharmaceutical company in Turkey. Teamed up with its leading international partners and compliant with ethical and scientific principles, Gen supplies products used in treatment of rare diseases and disorders in different branches and aims to bring these products to patients in the easiest, fastest and most reliable way possible while striving to find and bring new treatments to patients with unmet medical needs. With its GMP certificated production facility based in Ankara, Gen exports its products to different countries and has offices in Ankara (HQ), stanbul, zmir, Trabzon, Azerbaijan, Kazakhstan and Russia with 400+ employees. For more information please visit the Gen website.

Alnylam Forward Looking Statements

Various statements in this release concerning future expectations, plans and prospects, including, without limitation, Alnylam's views and plans with respect to the ability to extend patient access to ONPATTRO in Turkey through the announced Distribution Agreement with Gen, and the number of patients in Turkey within the approved indication for ONPATTRO who are in need of new treatment options, Gens views and plans with respect to the speed and reliability with which it is able to bring innovative medicines to patients in Turkey, and Alnylams expectations regarding the continued execution on its "Alnylam 2020" guidance for the advancement and commercialization of RNAi therapeutics, constitute forward-looking statements for the purposes of the safe harbor provisions under The Private Securities Litigation Reform Act of 1995. Actual results and future plans may differ materially from those indicated by these forward-looking statements as a result of various important risks, uncertainties and other factors, including, without limitation: Alnylam's ability to discover and develop novel drug candidates; the pre-clinical and clinical results for its product candidates, which may not be replicated or continue to occur in other subjects or in additional studies or otherwise support further development of product candidates for a specified indication or at all; actions or advice of regulatory agencies, which may affect the design, initiation, timing, continuation and/or progress of clinical trials or result in the need for additional pre-clinical and/or clinical testing; delays, interruptions or failures in the manufacture and supply of its product candidates or its marketed products, including ONPATTRO in Turkey; obtaining, maintaining and protecting intellectual property; intellectual property matters including potential patent litigation relating to its platform, products or product candidates; obtaining regulatory approval for its product candidates, including lumasiran and product candidates developed in collaboration with others, including inclisiran, and maintaining regulatory approval and obtaining pricing, reimbursement and access for its products, including ONPATTRO and GIVLAARI; progress in continuing to establish a commercial and ex-United States infrastructure, including in Europe; successfully launching, marketing and selling its approved products globally, including ONPATTRO and GIVLAARI, and achieve net product revenues for ONPATTRO within its expected range during 2020; potential risks to Alnylams business, activities and prospects as a result of the COVID-19 pandemic, or delays or interruptions resulting therefrom, including without limitation, any risks affecting access to ONPATTRO in Turkey, Alnylams ability to successfully expand the indication for ONPATTRO in the future; competition from others using technology similar to Alnylam's and others developing products for similar uses; Alnylam's ability to manage its growth and operating expenses within the ranges of its expected guidance and achieve a self-sustainable financial profile in the future, obtain additional funding to support its business activities, and establish and maintain strategic business alliances and new business initiatives; Alnylam's dependence on third parties, including Regeneron, for development, manufacture and distribution of certain products, including eye and CNS products, and Ironwood, for assistance with the education about and promotion of GIVLAARI in the U.S.; the outcome of litigation; the risk of government investigations; and unexpected expenditures, as well as those risks more fully discussed in the "Risk Factors" filed with Alnylam's most recent Annual Report on Form 10-K filed with the Securities and Exchange Commission (SEC) and in other filings that Alnylam makes with the SEC. In addition, any forward-looking statements represent Alnylam's views only as of today and should not be relied upon as representing its views as of any subsequent date. Alnylam explicitly disclaims any obligation, except to the extent required by law, to update any forward-looking statements.

View source version on businesswire.com: https://www.businesswire.com/news/home/20200325005133/en/

Contacts

Alnylam Pharmaceuticals, Inc. Christine Regan Lindenboom(Investors and Media)+1-617-682-4340

Fiona McMillan(Media, Europe)+44 1628 244960

Gen Ayhan Yener, MD(Medical Director)+90 554 566 57 40

See the article here:
Alnylam Pharmaceuticals and Gen Sign Distribution Agreement in Turkey for ONPATTRO (patisiran), the First-in-Class Gene-Silencing RNAi Therapeutic -...

‘Headed down the wrong road’: The quest for precision medicine distracts us from what already works – Genetic Literacy Project

Human health and biomedical science have been transformed in tandem over the past two centuries. From around 1850 to 1920, better nutrition and prevention of epidemic infections were the main contributors to massive improvements in life expectancy and reductions in infant and maternal mortality. These challenges have now receded for much of the world. Chronic diseases have become the principal threats to a healthy lifespan, and in the past half century we have seen a second wave of improved health, above all from enormous gains in control of cardiovascular diseases and cancer. Whereas the increase in life expectancy in the industrialized world from about 50 years in 1900 to nearly 70 years in 1950 is well-known, the addition of nearly a decade in life expectancy since 1970 is far less appreciated. But a rich body of theoretical and practical experience on what drove this more recent wave of success yields a clear set of principles that are securely established as foundational concepts in biomedical and related sciences.

What does transformational progress in reducing the burden of chronic disease look like and how did it happen? What can it tell us about the most promising pathways for future population-wide advances in health?

About two-thirds of all deaths in the United States and most industrialized countries are caused by cardiovascular disease (CVD) or cancer. Although the burden and character of both these disease categories have evolved dramatically in recent decades, for CVD the magnitude of the reduction in incidence (occurrence of new cases, both fatal or nonfatal), prevalence (proportion of the population living with a chronic condition), and death toll in the past 60 years is not widely appreciated. Mortality rates from coronary heart disease (CHD), the most common form of CVD, in the United States peaked in 1968, and have declined by 2%-3% every year since, until the past two years. The total reduction, after adjusting for changes in the age structure of the population, is now over 75%. Though CVD of course remains common, there are at least six hundred thousand fewer CVD deaths per year in the United States as result of this decline, and the total number of deaths averted since 1968 is on the order of twenty million. This unprecedented success in reducing the burden of the leading cause of death in industrialized countries was achieved not by any single intervention, but through advances along multiple pathways: the development of a clear understanding of the etiologic process; determined efforts to create public awareness, especially about the role of diet and physical activity; successful policy interventions to promote such things as low-fat dairy products and removal of trans fats from food items; introduction of safe, cheap, and effective medications to treat causal risk factors such as high cholesterol and blood pressure; and improved treatment of acute cases and advanced disease of the coronary arteries. Rapid declines in mortality from stroke, the other component of CVD, have been even larger than the declines from CHD, and are continuing as well.

Although the magnitude is considerably smaller, progress has now been made in lowering cancer death rates. Age-adjusted total cancer mortalityhas declined over 30%in the past 25 years in the United States. Reduction in tobacco use is the most important factor, and for men accounts for 40%-50% of the overall decline in all cancer deaths. Male smoking prevalence rates have dropped from 65% to 20% in the past 70 years. Lung cancer mortality has declined 50% in men since 1991; colon cancer deaths fell by 50% for both sexes; breast cancer deaths in women are now 40% lower. Cervical cancer death rates have declined by 60% since 1975, and prostate cancer mortality has declined by more than 50% since 1994.

Cancer, of course, is a composite of a wide range of tumors, each with distinct causes, natural histories, and challenges to prevention, detection, and treatment. Nonetheless, the main drivers can be identified. The great decline in smoking is responsible for reductions in lung cancer in both sexes and for fewer deaths from laryngeal and likely bladder cancer. Widespread adoption of technologies that permit early diagnosis (for breast and prostate cancer) or detection of premalignant states (cervix and colon) appear to have been the largest contributors to the reductions in mortality of these cancers, but improved surgical techniques and treatment options have probably played a role as well.

Thus modern biomedicine, especially the component focused on prevention, has brought transformational change not only to infectious diseases but to chronic diseases that less than five decades ago posed hopeless challenges. In the course of this transformation, we have accumulated a rich knowledge base of what research, what tools, and what implementation strategies work in reducing the burden of disease and death.

Throughout most of this remarkable period of improved public health, the field of genetics functioned within well-defined subdisciplines in such diverse areas as selective breeding of animals and improvement of crop yields, statistical modeling of heredity, and experimental work to understand monogenic disorders. One of the most significant accomplishments early on in genetics was the clear demonstration that most common or complex traits were highly polygenicthat is, they resulted from the combined small effects of many genes. Characterization of the double helix structure of DNA in 1953 and parsing how genes are expressed in terms of molecular function ushered in a new era of intense focus on the molecular pathways that shape the growth and maturation of the organism. In the 1980s the advent of faster, more efficient gene sequencing technology ignited an explosion of new research opportunities, and eventually the transformation of genetics, a reasonably discrete scientific discipline, into genomics, a vastly more open-ended project with its sights set on establishing a precise, mechanistic description of the fundamental rules of biologynot only of intergenerational transfer of information via germline DNA, the traditional focus of genetic research, but of the causal sequence underlying virtually all disease processes. The launching pad for this new era was the Human Genome Project, led in its first phase by James Watson, codiscoverer of the double helix. At a projected cost of $3 billion, only the federal government, primarily the National Institutes of Health, could afford such a large and focused endeavor.

This massive project encouraged people in the genomics community to see themselves as transformational actors in all forms of medical research, and to promise equally transformative benefits for health. As NIH proclaims in its mission statement the goal of NIH research is to acquire new knowledge to help prevent, detect, diagnose, and treat disease and disability. Genomics became key to that mission. In June 2000, at the ceremony hosted by President Bill Clinton announcing the completion of the Human Genome Project, the world was offered the hope that genome science will revolutionize the diagnosis, prevention and treatment of most, if not all, human diseases. Twenty years on, the scale of investment, the dynamism within the field, and the far-reaching claims for transformational impact on health and medicine have only accelerated.

Genomics has further matured since 2000, and broadened into an open-ended pursuit spanning domains from how cells regulate metabolic activity to the impact of all environmental exposures that individuals encounter over their lifetime. As a consequence, an enormous growth in resources devoted to research, training, technology development, and implementation, including a substantial de facto reallocation of resources that had been used in traditional biomedical disciplines, has been directed to genomics-oriented biomedicine. NIH, with its $39 billion annual budget, has aggressively promoted this agenda, and currently invests roughly half its resources in genomics-related research. In parallel, NIH spends less and less on research into prevention and public health. In pursuit of the goal to make genomics the basis for a new era of precision medicine, NIH Director Francis Collins has launched the All of Us research program, with a goal of recruiting one million Americans to have their whole genome sequenced, at a total cost of about $1 billion. NIH continues to advance an undiminished message of promise for this science, as Collins declared in May 2018: We would expect to see more effective prevention of many diseases, fewer diagnoses of serious illness, and an extension in health span. Genomics science is now being offered as the foundation for a population-based medicine of the future.

We thus appear to be in the early stages of a decisive transition between the multifaceted approach that has yielded such progress over the past two centuries, and the emerging new model driven by genomic sciences and captured by the term precision medicine. What, then, should we expect from genomics and personalized medicine in meeting the enormous chronic health challenges that remain, such as diabetes, dementia, arthritis, renal failureand, of course, CVD and cancer? And what about the huge challenges presented by violence and suicide? Based on well-established principles, drawn from the history of biomedicine and public health, are the promissory notes issued by genomics likely to be cashed for value? Does the evidence of success from the first two decades of the genomics era justify the original and continued predictions of transformational progress in population health?

The current undeniable crisis within the health care delivery system notwithstanding, we now stand on a massive edifice of technology and basic biology. We can muster a vast array of effective pharmacologic agents, transplant many solid organs, and perform extraordinarily complex surgical procedures; we can detect and treat many conditions in their earliest stages; we have developed many ways of preventing disease before it starts, especially with the weapons of social policy. At the core of these advances lies a set of principles about what causes disease and how large-scale population-level improvements are achieved.

In the middle of the nineteenth century, the great German pathologist and public health advocate Rudolf Virchow declared mass disease means society is out of joint. A more contemporary framing of that principle would emphasize that like the rest of the animal and plant kingdom, humans long evolutionary lineage has left us well adapted to life on this planet. As a corollary, the vast majority of health risk, certainly more than 90%, is derived from deleterious environmental exposure over our life course, not information encoded in our DNA. Human genetic adaptability is no match for newly emerging threats to health, as the history of epidemics shows. When a new agent appears on the scene, whether cholera, a new flu virus variant, cigarette smoking, or a huge increase in animal fat consumption, the human genome is unable to prevent the premature deaths of millions of humans. In other words, the human genome does not express itself as a dominant or primary cause of mass disease.

Occasionally, evolutionary forces have conferred new protective adaptations to region-specific epidemicsthe role of sickle-cell disease and thalassemia in reducing risk of severe malaria among some African populations is the paradigmatic example here. But the genetic repertoire of our species leaves us susceptible to many noxious exposures that vary with time and place too rapidly for evolutionary forces to react to. Mass disease with a global scopeillnesses that occur in at least 5%-10% of the worlds populationalmost always occur because of widespread insults that arise external to the organism, whether it be the sanitary conditions in medieval cities that gave rise to plague, or the conditions of trench warfare that contributed to the 1918 flu pandemic. Mass diseases are products of the societies in which we live.

In earlier periods, both insufficient total calories and inadequate sources of specific nutrients were the primary drivers of risk. In the modern era, all too often excessive exposure to substances that are inhaled, ingested, or absorbed through the skin are the fundamental causes of common disease. The transformational events in our progress against chronic disease have been driven by mitigation of those exposures through a familiar litany of interventions such as vaccination, improved diet, and altered behavior. What we have learned about disease, medicine, and population health over the past two centuries tells us that the primary domain of interest for disease prevention consists of external factors, which are products of sick societies, rather than heritable factors that modify individual-level risk. From this perspective, the prediction that precision genomic medicine will lead to population-wide health demands a sharp break from the source of such benefits in the past.

The introduction of genomics into the mainstream of biomedical research violates another historical precedent. Though technology has clearly played a key role in helping build the current medical armamentariumfrom imaging to clinical chemistry to drug developmentwith rare exception those technological advances emerged from focused research on a disease challenge (vaccines), had self-evident utility at the moment of discovery (the Roentgenogram), or were imported from outside the medical enterprise (lasers, fiber optics). Scientific advances that have led to improved health have nearly always been the result of research that matched technologies to specific human health problems and their clinical solution.

The grand theory of human molecular genetics is that the gene is a code that needs only to be deciphered in order to solve the problem of human disease. This theory arose in the 1950s simultaneously with, and reinforced by, the development of modern computer science, with coding of programs at its root. A few decades later, as we have noted, genomics developed as a laboratory-based technology, erupting into widespread use with the development of rapid and accurate sequencing methods. Only then was genomics injected into mainstream biomedicine, and retrofitted to address problems beyond its inherent scope. As applied to clinical medicine, DNA sequencing technology is first and foremost a tool to study germline errors in the code, preeminently Mendelian (single-gene) traits. The ethos of genetic determinism, and the irresistible allure of technological solutions, have opened the door for the adoption of genomics for the study of conditions where it has no or minimal relevancenamely most, if not all, human diseases. Sequencing (and the rapid acceleration of its throughput) was quickly promoted as a tool for gene mapping, and thereby attracted much broader research interest. The current wave of large-scale gene-mapping projects has been justified through an appeal to genetic determinismmore causal knowledge self-evidently must lead to solutions. This new causal knowledge would come, in ways we could not foresee, from as-yet-unexplored domains.

When tested, however, these hypotheses have not, to date, been adequately verified. Likewise, the effect of individual genetic variants being sought have been grossly overestimated, in clear contradiction of the established theory of the supposed polygenic inheritance of complex traits. The outcome was almost preordainedgenomic theory emerged without feedback from the clinical or any other empirical setting, and it advanced and captured increasing swaths of biomedical science without evidence of improved population-wide medical advance. Indeed, the more we learn about the genome, the more distant it seems to be from a role as a causative agent in most widespread diseases. We have every reason to believe that the same will be true for those diseases where the etiology remains obscure.

Beyond an appeal to principles, we now have the accumulated experience of some 25 years of genomic research, with a few projects reaching back as far as the 1970s. Anything approaching a broad summary of this rapidly evolving science is clearly beyond our scope here. However, an empirical basis for our concerns is required, and several conclusions have now achieved general consensus in the academic community. First, however, it is necessary to reaffirm what no one disputesthat adequate support for all branches of science is an essential investment in the infrastructure of modern society. Nor can those investments be limited to science that promises near-term benefits. For genomics at the present, this trade-off was articulated by former National Cancer Institute director Harold Varmuss sentiment that genomics is a way to do science, not medicine. Second, the advent of genomic technology has already generated a huge array of new tools beyond DNA sequencing that have transformed many lab sciences, and advanced public health, for example contributing crucially to our understanding of the spread of viruses in epidemics and the evolution of drug resistance in microorganisms, to new diagnostic assays, and to immunotherapy for cancer.

Major changes in population health, and extension of healthy years of life, however, belong to a dimension far removed from these incremental, niche advances, as beneficial as they are for many patients.

Enthusiasm for genomics and precision medicine builds on expectations for major scientific and medical progress in at least five major areas.

1. Enabling disease prediction.Although thousands of familial, genetic syndromes had been catalogued in the pre-genomic era, it is now possible to define the DNA sequence variations in great detail, and early success with cystic fibrosis, the so-called BRCA complex related to breast cancer, and Huntingtons disease offered the promise of much wider translational success for genomics. As noted, however, for most diseases the impacts for specific genetic factors are small, and studies of unprecedented size were required. Many of these have now been completedat enormous cost, needless to sayand a robust literature exists for common disorders such as CHD, diabetes, hypertension, obesity, and other metabolic traits.

Focusing on two pressing public health concerns of the moment, CHD and diabetes, we have conclusive evidence regarding risk prediction from DNA markers. Collectively representing cohorts of almost half a million patients, four major studies have now published virtually identical results. As is well known, the odds of dying from CHD is driven by four major risk factors: elevated cholesterol, cigarette smoking, high blood pressure, and diabetes. After accounting for these easily measured traits, DNA markers offer trivial additional information, perhaps identifying 2%-3% of individuals who might be reclassified as low or high risk. The sole response to this information would be adjustment of the dose of a cholesterol-reducing statin at a younger age. Roughly 80%-90% of the risk of the common adult form of diabetes, type 2 diabetes, can be determined from body mass index (BMI; a simple ratio of height to weight), and randomized trials have shown that in almost half of patients type 2 diabetes can be prevented, and indeed normal glucose control restored, with weight loss. Regular fitness activity and cessation of smoking also modify risk. The very large studies already completed demonstrate that virtually no additive predictive information can be derived from more genome-wide searches for additional risk variants. Similar knowledge has emerged for hypertension, stroke, dementia, and numerous other conditions.

Genetic prediction of cancers similarly struggles with predictors that are too weak in most cases to be useful in clinical practice. For example, in a very large European database in which the average lifetime risk of breast cancer is 5.1%, the risk for women in the top 5% of gene scores is 12%, and in the bottom 25% it was 2.4%. These results have no impact on clinical practice: preemptive invasive procedures cannot be justified for a group of women whose likelihood of not getting breast cancer is 88%, and 2.4% risk is still too high to abandon screening.

2.Providing critical new insights into molecular pathways.The rise of genomics has encouraged the view that once the DNA mutations underlying a trait have been identified, no matter how small, downstream metabolic consequences would be revealed and, along with them, targets for clinical intervention. Efforts to define cell-based pathways using molecular technology have in fact met with some success. We now know, for example, much more about immune function, control of fetal hemoglobin, and lipid regulatory mechanisms, in large part through application of genetic and molecular technology. However, most metabolic networks are so intricate, redundant, and multidimensional that following Ariadnes thread is mere childs play compared with an attempt to move from identifying a mutation to tracing that mutation to a specific physiological outcome. Complexity involved in inference from genotype to organism has been evident for years. In sickle cell anemia, for example, an apparently simple genetic changethe single nucleotide substitution of adenine for thymine in the hemoglobin geneproduces strokes, pulmonary hemorrhage, painful bony crises, and enhanced susceptibility to the pneumococcus bacteria. The linkage of genetic change to clinical manifestation is sufficiently complex that six decades after the underlying molecular basis of the disorder was discovered, we still have no specific therapy for the condition.

A technique that will knock out altogether the action of a gene almost invariably does not lead to the expected observable consequence in the organism, and there is as yet little evidence that genome-wide association studies that statistically link multiple genetic sites to risk markers for diseases are leading to significant improvement in understanding pathophysiologic processes. The rare successes that have been achieved (for example, identification of an allelic variant in the genetic locus PCSK9 that influences cholesterol metabolism) are still being derived from study populations where the link between risk and genetics (for example, in high-risk families) was already long-established. Living organisms are simply too complex to yield up a set of fundamental laws, and instead reveal more and more intricate processes and networks that wriggle and squirm across time and space, refusing to cast a fixed image.

3. Isolating genetic mutations that predispose patients to severe adverse drug reactions.Pharmaceutical agents are essentially foreign bodies, as far as our species is concerned, that evolution has never been called on to protect us from. It should be unsurprising, then, that many drugs have side effects, as well as some variation in absorption, metabolism, or effect depending on the individual. Genetic predisposition therefore can play a role in modulating person-level response. Some important successes have been achieved, especially in the identification of people at risk for severe adverse reactions. Early in the experience of so-called pharmacogenomic testing, variation in the efficacy of drugs used to prevent blood clots was identified. The added value of characterizing the relevant genes has now been studied in clinical trials. The most important examples are warfarin and clopidogrel, drugs that inhibit clotting by modifying platelet function. Both have significant side effects. Clopidogrel requires further metabolic conversion in the liver to make the active compound, and person-level variation in enzyme function produces the genetic effect. The original molecule was reformulated to avoid this variability in response; the most recent agent to become available, ticagrelor, avoids the between-person variability seen with clopidogrel, and thereby obviates the need for gene testing. Clearly drug companies have great incentive to market their drugs to the widest possible sales base to maximize profits, and want to avoid the step of gene testing if possible. Additional new drugsthe so-called non-vitamin K oral anticoagulantsare now showing promise for patients experiencing serious side effects from warfarin, further limiting the role of gene testing for that drug.

Current use of the new anticoagulants, however, requires conventional assays of platelet function. This return to traditional practiceomitting gene prediction and measuring the physiologic variables that are the direct target of the treatment (e.g., serum lipids, blood pressure, blood sugar)reaffirms our assertion that decision-making for individual patients will continue to be based on biochemical or other basic parameters. The complexities along the pathway from gene to physiologic outcomes are almost always influenced by too many other factors for us to be able to make clinically useful decisions from genetic information.

For other classes of common drugs, even these modest successes have rarely been seen. For example, a very extensive, long-running NIH-funded project on medications used for high blood pressure resulted in genetic scores that at best predicted 1-2 mmHg difference in response between individuals after testing; as above, they merely confirmed that direct measurement of blood pressure after you prescribe the drug will remain the basis of clinical practice. A vast array of other minor findings has been reported, but over time the scenarios we outline here have been repeated: either new agents replaced drugs that required gene testing (including a drug for hepatitis C), or the genetic effect was trivial. Pharmacogenomics overall has therefore not lived up to early expectations. Additional efforts face a stiff challenge to success, for reasons that should now be familiar: links between genomic makeup and patient response to drugs are too complex to have much clinical value, and actual measurement of physiological end points are almost always more informative.

4. Identifying targets for new drugs.At the very earliest stages of the genomic revolution, the pharmaceutical industry and innumerable start-up companies invested heavily in the search for novel targets that could be identified through DNA association studies. Though some new agents discovered from genetic research are in clinical trials (for example, a protein inhibitor for elevated triglycerides and an RNAi blocker for fatty liver disease), these efforts have yielded surprisingly little. In fact, a crisis has emerged with a drastic reduction in new drugs coming to market in the past two decades. An important exception relates to drugs influencing immune response, including autoimmune diseases. And there may well be drugs in the pharmaceutical pipeline based on genomic research that could yet translate into useful products.

5. Unlocking at long last the secrets of cancer, which after all, according to the current dominant theory, is a genetic disorder.Despite years of intense, well-funded research, progress toward effective treatment, let alone cure, of most cancers remains an elusive goal. To oversimplify the general proposition, harmful DNA mutations at least at some stage may drive the growth of tumor cells, and ultimately the metastases that prove fatal. Identifying driver mutations and blocking their effects could thus possibly offer cures. Unfortunately, the results across all these hope-filled propositions have, in sum, been dismal. From a historical perspective genomics is a young science, and the unexpected will occur with time. However, for some hypotheses, accumulating research is asymptotically approaching a null result.

The dominant theory in cancer biology remains gene-centric: either somatic mutations, occurring in the absence of known external cause, allow a clone of cells to escape from normal control of cell replication and death, or pathologic mutations in some less-defined way act at the earliest stages to drive growth and metastasis of tumor cells. Whereas it is incontrovertible that carcinogenic agents of diverse types, including viruses, ionizing radiation, and aromatic hydrocarbons, do cause pathologic mutations, a vigorous debate continues within oncology as to whether this is actually the process that triggers and sustains cancer development. For example, recent work demonstrates that normal tissue adjoining tumors harbors the same mutations as the tumors themselves; conversely, tumors transplanted from one model organism to another usually do not survive. In other words, the mutations themselves are clearly not the sole actors, or perhaps not even the causal driver, of tumor growth. Thus, a complementary field theory has been proposed that emphasizes tissue-level factors, particularly cell-to-cell communication. Recent experimental evidence now conclusively shows that at least some of these abnormal functional states, which cannot be explained in terms of mutations, must exist for tumors to propagate locally and, more importantly, to metastasize. Though this brief summary hardly does justice to a complex, rapidly evolving field, we hope it begins to communicate why large-scale sequencing projects of tumors have not delineated clear causal pathways, and more importantly why agents developed to block driver mutations have usually not met expectations, or, if they succeed, seem to act through entirely unexpected and independent mechanisms.

Despite these unresolved questions, substantial success has been achieved with several classes of new antitumor drugs. The drug imatinib mesylate, marketed as Gleevec, has been widely celebrated in this regard. Although it is the sole example in current use of target-selective therapy, it has led to lasting remission for some two thousand patients in the United States per year with chronic leukemia, without the debilitating side effects of chemotherapy. But Gleevecs success may be (and so far has been) hard to repeat: it works on a specific causal chromosomal abnormality of the sort that is uncommon in most cancers. Moreover, life-long therapy is required at an approximate average cost of $1 million per patient.

Biomarker-driven molecular research, leading to development of antibodies on the cell surface of individual tumors, has also met with substantial success. For example, a panel of genetic tests for individual tumors that allows better matches for drug therapy has entered the clinical arena. Immunotherapy, an important example of the transition from molecular research to translationespecially the so-called check point inhibitorscan specifically target solid tumors in about 15% of patients, although these are not genetic targets. An increasing, but small, proportion of patients with melanomas have attained durable long-term remission with a combination of new genetic/immunotherapies.

But the central theory of cancer as a genetic disorder, with its corollary that the ability to identify unique driver mutations will lead to therapies that can block their action, has not been verified. Instead, it has become a piece of a much more complex puzzle. Whereas any research toward safe, effective antitumor drugs is of enormous value, when entered onto the balance sheet of factors that account for the 30% decline in cancer rates achieved in the past several decades, the contribution of new curative agents developed through molecular techniques to improvements in health on a scale measurable in population-level statistics remains, at best, somewhere in the range of 2% or less. New knowledge will increase this contribution, yet predictions of a truly transformative role for treatment of advanced cancer lack empirical justification. Most invasive solid tumors have remained stubbornly resistant to curative or durable palliative therapy. At the same time, two new immunizing agents against viruses established to cause cancer in the past few decadesthe human papilloma virus that causes cervical cancer, and the hepatitis B virus that underlies most cases of hepatocellular carcinomapromise, if widely used, to virtually eradicate these two cancers without regard to genomic variability, potentially saving more than a million lives a year worldwide.

Many observers will no doubt find this account overly pessimistic. Numerous success stories have been omitted. The advances enabled by the advent of genomic technology are far-reaching and of great scientific importance. Whole subdisciplines, from human evolutionary history to epidemic surveillance and vaccine preparation for conditions such as Ebola and influenza, have indeed been transformed. But a key distinction is that these advances are due to the power of genomics when applied toagentsof human disease, not to the diseasehost. Many infectious organisms must take advantage of molecular targets on the surface of cells or be targets for killer white cells. Thus an understanding of species-specific susceptibility to bacteria or virusesfor example, pneumococcus or Ebolacan be very informative in vaccine preparation. Distinguishing molecular signatures of pathogens within a species has made outbreak investigation much more precise. Buthumangenomics and precision medicine have not transformed human health. Nor, in our view, is there a basis from which to argue that they will do socertainly not in any foreseeable future.

Meanwhile, the opportunity costs are enormous. To help bring them into focus, we offer a somber, indeed heart-breaking story that has played out in one of the oldestand thereby most matureexperiments to employ genomics as a tool to improve human health. The Pima Indians of the Sonoran Desert in central Arizona were deprived of irrigation water from the Gila River around 1900 when it was diverted upstream by commercial farmers. Isolated, and confronted with famine, they became dependent on food subsidies from the US Department of Agriculture, and adopted a diet low in nutritional value but high in calories. In the following decades an epidemic of obesity of unprecedented magnitude swept the reservation. The prevalence of type 2 diabetes rose to 50%, and even adolescents with the disease have now required dialysis for renal failure. In the 1970s, NIH established a research institute in nearby Phoenix to search for the unique genetic factors that predisposed the Pima to this crippling disease and use this knowledge to cure or prevent the disease. Despite five decades of research no important genetic mutations were identified, and the sum of available evidence showed that those susceptibility loci that could be isolated were no different, and no more common, in the Pima than in the majority US population.

The depths of the intellectual poverty of this long-running experiment can be summarized by the following quotation, posted as a research advance by the Phoenix group on the website of the director of the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) in 2016: In a prospective study conducted between 1965 and 2007, NIDDK investigators followed children from Arizonas Gila River Indian Community for development of type 2 diabetes. They found that BMI and impaired glucose tolerance were strong predictors of type 2 diabetes, but other components of the metabolic syndrome were not. It should be noted here that the scientific observations highlighted in this advance was known to the Ayurvedic medical tradition in India in the fifth century BCE, and was certainly common knowledge in the modern era by the seventeenth century. Yet even today this genomic research continues along the line explored for so many years among Pima populations. The rationale for an NIDDK project started in 2015 states: When it comes to kidney disease years of exposure to diabetes may change the way the body reads its DNA, increasing the risk of kidney disease being exposed to high blood sugars or high blood pressure may cause people who have a genetic susceptibility to have kidney disease in the future.

About 10 years ago the Gila community broke off collaboration with NIDDK and mounted its own preventive campaigns based on weight control and increasing physical activity. The risk of hyperbole notwithstanding, this unhappy saga ranks with the Tuskegee Experiment as another egregious project where the natural history of a fatal illness in a vulnerable population was allowed to run its course, under the careful observation of government-funded scientists, in pursuit of a narrow, unjustified hypothesis, built on notions of genetic determinism and race-based susceptibility, while available preventive or curative alternative interventions were ignored or actively shunned.

We wonder if the story of the Gila River Indian Community, writ large, will be the outcome of the headlong rush toward precision medicine across the entire spectrum of biomedicine. In the cancer realm, for example, consider that fatty liver from obesity is now becoming a major cause of liver cancer in some countriesyet another widespread example of sick societies. Or pancreatic cancer, whose etiology has been an enigma, and for which early detection and effective treatment remain elusive goalsyet very recent evidence suggests fungal infection from the gut could play a major role. A similar causal pathway emerged between the Helicobacter pylori bacterium and stomach cancer, and treatment of this type of bacteria has had an important impact on peptic ulcer disease, and may yet influence the risk of stomach cancer. Preventive interventions for more cancers are almost certainly possible. The search for environmental causes of cancer proceeds apace, but the effort remains modest compared with the work expended on searching for genomic correlates. We have learned the importance of radiation, microorganisms, toxic chemicals, and lifestyle factors such as obesity, and (for breast cancer) age at first birth, but we still have much to learn about the nongenetic causes of cancer. Implementation of modalities to prevent and detect premalignant lesionsas with colonoscopycould achieve much less costly and more rapid downturns in cancer mortality than are likely to result from the long road to curative therapies derived from molecular research, which are often applied near the end-stage of disease.

Progress is also being observed in two other significant chronic disease challenges. There is a growing awareness that vascular disease has been underappreciated as a cause of dementia, and improving heart health is now accompanied by a welcome decline in incidence of dementia in the elderly. Type 2 diabetes is a major global threat to health, moving rapidly to nonindustrialized countries. As noted, whereas obesity accounts for 70%-80% of risk, and reversing the obesity epidemic has thus far proven difficult, type 2 diabetes incidence rates appear to have leveled off or declined in many countries. Randomized trials have demonstrated that weight loss of as little as 15 pounds can lead to a 40% reduction in onset of type 2 diabetes in high-risk patients, and return 40% of patients with recent onset of the disease to nondiabetic status. Policy to influence food production and sales, as well as eating patterns, is in its infancy and can boast only modest success, but that is clearly the only solution to the challenge of obesity and type 2 diabetes. It goes without saying that the opioid epidemic in the United States, which has cost 770,000 lives since 1999 and reduced overall life expectancy, is a poster child of a social disease whose amelioration will not be driven by precision genomic medicine.

We do not believe genomics and precision medicine will transform biomedicine and population health. Though the history of science will have the final word on this era, we believe that large segments of the biomedical community, supported by tens of billions of public dollars, are in effect headed down the wrong road, if not into a cul-de-sac. To understand this assertion, it is essential to recognize the distinction between transformational change and widespread niche advances. The concern that we have addressed here lies singularly with population health, with benefits accruing to millions. Scientific understanding of both the reasons for enormous gains in population-wide health, and the origins of disease, are being largely displaced by a reductionist, technology- and theory- (and career- and profit-) driven approach to health and medicine that remains largely unproven (and wildly expensive). Of course we want to explore and pursue many new research avenues, but the powerful legacy of genetic determinism and the devotion to technological solutions have narrowed the scope of research aimed at improving population health, and thus narrowed and reduced the benefits that biomedical science could and should be providing, right now.

Ironically, two decades into the genomics revolution life expectancy in the United States has declined for three consecutive years, the reduction in cardiovascular disease rates has leveled off, and a surge of opioid deaths has devastated many communities. These adverse events have no direct relationship to genomics or precision medicine, but just as clearly we have not observed the promised bonus of more effective prevention of many diseases, fewer diagnoses of serious illness, and an extension in health span. We could, of course, be accused of making a grossly premature judgment. Two decades is a reasonable interval, however, in which at a minimum to demonstrate proof-of-concept, and we see no evidence of that modest milestone having been reached. More to the point, we argue that genes as a cause and precision medicine as the cure violate basic precepts of health and medicine. Biomedical science should be reoriented and reprioritized to expand its scope in accord with what we actually know about health and disease, and to expand the benefits of science for all.

Read the original:
'Headed down the wrong road': The quest for precision medicine distracts us from what already works - Genetic Literacy Project

This Startup is on a Mission to Decentralize Cell and Gene Therapy Clinical Trials – BioBuzz

For the cell and gene therapy revolution to be fullyrealized, physicians, research scientists, biomanufacturing experts, advocacygroups, regulatory bodies like the Food & Drug Administration (FDA) andother key stakeholders have to Think Different, as Apple famously encouraged.

The vein-to-vein, one batch to one patientformula of personalized medicine is radically and rapidly forcing changes onbiomanufacturing where cell and gene therapy best practices are not yetcalcified and are changing as we speak. Personalized medicine supply chainchallenges are emerging and still being worked through and the high cost ofthese therapies remains a daunting challenge for life science companies andpatients.

Cell and gene therapy challenges across R&D, manufacturing, commercialization, and supply chain as well as bioethical challenges yet to be fully confronted or resolved have been well documented and discussed by experts across the BioHealth Capital Region (BHCR).

One company Jeeva InformaticsSolutions, Inc. (Jeeva) in Herndon, Virginia is developing BigData solutions for a less publicized but equally urgent cell and gene therapychallenge: How to manage a radically new form of clinical trial that could spandecades. Founder and CEO of Jeeva, Harsha K. Rajasimha, is building a companythat leverages Big Data, AI and mobile tech to decentralize clinical trials.Jeevas goal is to make it easier for biotech companies to collect, aggregate,analyze and report required clinical trial data while helping patients stayenrolled and compliant over longer periods of time with little travel from theconvenience of their homes.

The FDA recently passed newindustry guidelines requiring long-term follow up (LTFU) periods ofup to 15 years for gene and cell therapies, representing a sea change for howclinical trials will operate in the future.

The cell and gene therapy space provides significant hope for cures that can reverse genetic mutations. This space is growing. There are more than 900 clinical trials ongoing. Hundreds if not thousands of genetic diseases are likely to have therapies for the first time in our lifetime, stated Rajasimha.

At Jeeva, we are trying to solve a number ofissues but have focused on one particular problem that needs to be addressed ifthese cell and gene therapies are going to be delivered to patients: No oneknows the long term implications of these therapies because they are a one anddone type treatment. If a patient receives therapy today, they are done, and sothe FDA has mandated that recipients need to be monitored for up to 15 years.We are looking to address this issue with digital health technologies and AI,he added.

Jeeva believes that these long-term monitoringchallenges can be managed by making clinical trial participation easier viamobile applications, video conferencing consultations and centralizedscheduling, to name just a few of Jeevas product features. Leveraging digitalhealth tech can reduce the need for travel and eliminate inconveniences thatmight cause a patient to become non-compliant or, worse, drop out of a trialaltogether.

Decentralized clinical trials eliminate the heavy burden of patient travel and makes the process simpler and more efficient. Utilizing a Bring-Your-Own-Device (BYOD) approach and an eVisit consultation model to create decentralized trials can reduce brick and mortar visits by 20% to 80%, according to Jeeva.

Thenew FDA guidelines is the latest attempt to grapple with unchartedsafety protocols for cell and gene therapies. Clinical trial challenges are notnew to the biotechnology industry, however; rather, the approach to clinicaltrials has been inefficient and static for decades, leading to industry-wideproblems with clinical trial enrollment and recruitment that has a dominoeffect that lengthens the commercialization process and increases drugdevelopment costs.

We want to be a catalyst for accelerating thedrug development and delivery process. Patient recruitment is a huge barrierand has made the biopharmaceutical industry unsustainable. The average cost ofbringing a drug to market is $2.5B and takes 10-15 years to get to market. Wefeel that by educating and informing the global community about clinical trialsand enrollment opportunities using AI and digital health tech, we can help allstakeholders in getting people earlier access to treatments and getting thetreatments to markets faster, Rajasimha said.

Jeeva not only can help small to midsize biotechs improve trial recruitment and longer-term safety monitoring, but the company uses AI to improve clinical trial operations. By using AI and high tech tools, biotech companies can leverage historical trial data to shape new trials while empowering real-time adjustments to trials based on real-time monitoring to improve overall success rates.

Rajasimha continued, We have been building AItools to solve our customers specific needs, not just for AIs sake. Wesurveyed our customers last year and they told us Every single clinical trialseems like the first trial ever conducted by mankind. Even companies likePfizer and Novartis, which have been conducting hundreds of trials for decades,feel that when they launch a new trialit is no more efficient than theprevious trial.

So, we have been building an AI assistant that learns from past clinical trials data to make the next trial more efficient, he added. Rajasimha quickly reinforced patient centricity by saying it is not something you fix with an all technology solution, unless the robustly tested technology solution is combined with the human elements and focus on patients perspectives. Having been a global patient advocate in the U.S. over the past six years has given me a unique perspective on how to integrate technology in the lives of patients and caregivers. Moreover, a growing number of trials are recruiting patients from multiple countries and reducing international travel burden on patients over extended durations will be critical to achieve enrollment.

While the concept of virtual clinical trialsmight seem futuristic, Rajasimha and the Jeeva team believe the market is readyfor change.

A number of pilot projects or proof of concept clinical trials, about 20 of them, have been published where patients didnt go to the clinic at all. The feasibility of conducting such remote patients studies has been validated multiple times by the industry now. The tipping point has arrived. One of the key barriers for widespread adoption of decentralized clinical trials was a lack of FDA guidelines. Now the FDA has clarified its expectations about how the industry and stakeholders can share the responsibility to reduce the burden on patients. Enough validation and regulatory guidelines have put us in a position to give our customers what they need, stated Rajasimha.

Rajasimha sees partnering with smaller to midsize biotechs early on in the drug development process meaning well before the start of Phase II or III trials as an inflection point where it can deliver the greatest impact. In addition, we are seeing some initial interest from the Medical Cannabis industry, opioid crisis intervention for chronic pain management, and patient advocacy groups, where patients often live in remote, rural areas, can also benefit from decentralized, hybrid virtual clinical trials. Finally, real-world evidence studies, or longitudinal cohort studies, is also a growing market because companies need to collect and manage patients across longer time horizons, which is Jeevas sweet spot.

Rajasimha and Jeeva are starting to see this growing market interest manifest itself in new funding partnerships. Jeeva recently announced that CIT GAP Funds had invested in the company. Jeeva is currently in an early-stage investment round and the company is in active product development with a validated prototype. Jeeva is seeking new customer pilot projects to add to its ongoing pilots, which include chronic pain, medical cannabis, oncology and cell, and gene therapy products. Later this year, the company plans to complete multiple pilot projects and have validation in Good Clinical Practices (GCP) settings.

Rajasimha and his Jeeva team are certainlyembracing a think different approach to the future of clinical trials. Jeevaand its AI-driven, virtual clinical trial model is poised to help biotechcompanies thrive and meet the unmet medical needs of more patients across theglobe.

You can listen to Rajasimhas interviewwith podcast host Daniel Levine earlier this month on iheart radio here.

Team Jeeva is seeking customer pilot projectsand strategic partners to join the journey and will be exhibiting at the NationalInstitutes of Health Rare Disease Day event on Feb 28, 2020.Rajasimha will also be delivering a keynote speech on AI in rare diseases atthe BIO-IT World West Conference at San Franciscoon March 3rd, 2020.

Steve has over 20 years experience in copywriting, developing brand messaging and creating marketing strategies across a wide range of industries, including the biopharmaceutical, senior living, commercial real estate, IT and renewable energy sectors, among others. He is currently the Principal/Owner of StoryCore, a Frederick, Maryland-based content creation and execution consultancy focused on telling the unique stories of Maryland organizations.

See the rest here:
This Startup is on a Mission to Decentralize Cell and Gene Therapy Clinical Trials - BioBuzz

NIH Director: Were on an Exponential Curve – The Atlantic

Collins later met a Methodist pastor, Sam MacMillan, who was a very willing partner for me, tolerating my blasphemous questions and assuring me that if God was real there would be answers. It was MacMillan who introduced Collins to the work of C.S. Lewis, starting with Mere Christianity.

I realized in the very first two or three pages of that book that most of my objections against faith were utterly simplistic. They were arguments from a schoolboy. Here was an Oxford intellectual giant who had traveled the same path from atheism to faith, and had a way of describing why that made sense that was utterly disarming. It was also very upsetting. It was not the answer I was looking for. But it was, for Collins, the answer he eventually found, and at age 27, he became a Christian.

The embrace of that faith transformed not only his relationship with God, but also how he viewed other people, and himself. They are all, as Lewis said, angels around you. And the notion therefore that it is okay to put yourself in the driver's seat in every way regardless of what effect that has on others, it's simply indefensible. I think it did take what had been for me a pretty strong ambitious driving approach and moderate it, not to say that I didn't retain a fair amount of that, but maybe in a somewhat more loving, forgiving approach.

When I asked him how he sees faith now, in his late 60s, compared to how he saw things in his late 20s, he told me, I think I've also arrived at a place where my faith has become a really strong support for dealing with life's struggles. It took me awhile, I thinkthat sense that God is sufficient and that I don't have to be strong in every circumstance.

I found that striking, particularly in this moment. One of my great puzzles when I first became a Christian is that verse, My grace is sufficient for you, because My strength is made perfect in your weakness, he told me. That was so completely upside down for me. Weakness? And now I embrace that with the fullness of everything around me when I'm realizing that my strength is inadequate, whether it's coronavirus or some family crisis, God's strength is always sufficient. That is a such a great comfort, but it took me a long time to get to the point of really owning that one.

Collins was the founder and creative force behind BioLogos, an organization that invites the church and the world to see the harmony between science and biblical faith. (BioLogos was launched at the time Collins was asked by President Obama to become the director of NIH, in April 2009, which required him having no other affiliations with any other organizations.)

I asked Collins what he hopes more Christians would understand about science and what he hopes more scientists would understand about faith.

To Christians I would say, think of science as a gift from the creator. The curiosity that we have been instilled with to understand how the universe works can inspire even greater awe of the creator. This gift could hardly be a threat to God, the author of it all. Celebrate what science can teach us. Think of science as a form of worship.

Read more:
NIH Director: Were on an Exponential Curve - The Atlantic

Genes Previously Linked to Heart Condition Disputed – Clinical OMICs News

A new analysis of available data has convinced a panel genomic experts that nine genes previously believed to be associated with a rare, genetic heart conditionlong QT syndromewere an erroneously linked to the condition, as revealed in a new study funded by the National Human Genome Research Institute (NHGRI), a division of the National Institutes of Health (NIH).

Geneticists and heart specialists around the world had previously reported 17 genes to cause long QT syndrome. However, the Clinical Genome Resources (ClinGen) expert panel has critically reevaluated the scientific evidence for all 17 reported genes, and has concluded at least nine of the genes cannot be linked to the disease, and only three of the genes can be definitively associated with the most common form of the disease.

Long QT syndrome is caused by mutations in genes that regulate the hearts electrical activity. These mutations can cause the heart to have sudden, irregular heart rhythms, or arrhythmias. People with long QT syndrome can have arrythmias that are both unprovoked or as a result of stress and exercise. These arrythmias can be fatal.

Many people with long QT syndrome may be unaware they have the condition, unless they get an unrelated electrocardiogram, know their family history, and have undergone genetic testing.

Ever since the syndrome was described in 1957, researchers have engaged in a genetic race to identify the genes associated with it, which currently includes the 17 genes. By using such a standardized, evidence-based framework, the international ClinGen panel experts on long QT syndrome were able to classify the 17 genes into specific groups.

Three genes, KCNQ1, KCNH2 and SCN5A, had sufficient evidence to be implicated as definitive genetic causes for typical long QT syndrome. Four other genes had strong or definitive evidence supporting their role in causing atypical forms of long QT syndrome, particularly if they presented in the newborn period with associated heart block, seizures or delays in development.

The remaining ten genes were deemed to not have sufficient evidence to support a causal role in the syndrome. In fact, nine of these 10 remaining genes were placed in the limited or disputed category. The study authors suggest that these genes not be routinely tested in clinical settings when evaluating patients and families with long QT syndrome, because they lack sufficient scientific evidence as a cause for the condition.

This removal of genes from the testing list impacts genetic testing providers, who use research papers to determine which genes to include in their testing panels for diagnostic reporting to physicians. Published papers reporting gene-disease associations vary widely in their study design and strength of evidence to support their conclusions. Until recently, standard guidelines that can differentiate between genes found with strong and valid scientific approaches versus those with insufficient evidence did not exist. Clearly, this is a problematic approach, and led to several studies drawing early conclusions.

ClinGens expert panels include researchers, clinicians, and genetic counselors who apply an evidence-based framework in evaluating the available data from research papers to place gene-disease relationships into definitive, strong, moderate, limited, disputed, or refuted categories.

ClinGen is an impressive community effort. With over 1,000 researchers and clinicians from 30 countries volunteering their time and expertise, ClinGen is providing much needed clarity for the clinical genomics community regarding which gene-disease pairs have sufficient evidence to be used clinically, said Erin Ramos, Ph.D., project scientist for ClinGen and program director in the Division of Genomic Medicine at NHGRI.

Our study highlights the need to take a step back and to critically evaluate the level of evidence for all reported gene-disease associations, especially when applying genetic testing for diagnostic purposes in our patients. Testing genes with insufficient evidence to support disease causation only creates a risk of inappropriately interpreting the genetic information and leading to patient harm, says Michael Gollob, M.D., senior author of the paper and researcher at the Toronto General Hospital Research Institute.

Moreover, testing for genes not definitively associated with long QT syndrome can result in inappropriate and costly medical interventions such as implanting of a cardioverter-defibrillator.

This is not the first time a team at ClinGen has clarified published research for clinicians. The same team of researchers published a similar study in 2018, covering another heart condition called Brugada syndrome. In 2019, the American Society of Human Genetics considered the paper as one of the top 10 advances in genomic medicine.

ClinGen is an NHGRI-funded resource created to define the clinical relevance and validity of genes associated with various genetic disorders. It comprises more than 20 expert panels working on a variety of genetically influenced diseases, ensuring the reliability of gene-disease linkage. This work is also instrumental in determining which specific genes should be targeted for further study in precision medicine and research.

More here:
Genes Previously Linked to Heart Condition Disputed - Clinical OMICs News