Page 40«..1020..39404142..5060..»

What might next 100 years hold for cardiovascular disease prevention and care? – Japan Today

Posted: July 11, 2024 at 2:43 am

The past century brought profound advances in the understanding and treatment of cardiovascular disease, leading to dramatic reductions in deaths linked to heart disease and stroke. But what will the next 100 years bring?

While no one can say for certain, experts point to three areas they expect may play a crucial role in the decades ahead: a push to halt the rise of cardiovascular risk factors using the knowledge gained in previous decades; the continued development of innovative technologies; and a heightened focus on the root causes of health disparities to prevent them from widening as the U.S. population grows more diverse.

Prevention: Putting knowledge gains to use

Over the past century, an enormous body of research emerged surrounding the factors and behaviors that contribute to cardiovascular disease. The American Heart Association compiled this knowledge into eight key measures that, if properly managed, could help to substantially lower the risk for heart disease, stroke and other major health problems. These include physical activity, diet, tobacco use, body weight, sleep duration and blood pressure, glucose and cholesterol levels.

"The challenge is how to translate that knowledge into health actions," said Dr. Adrian Hernandez, executive director of the Duke Clinical Research Institute and vice dean of Duke University School of Medicine in Durham, North Carolina. "That's where we have to do a lot more."

Research shows the rates of many cardiovascular risk factors have been rising in the U.S. and are projected to keep doing so. High blood pressure, diabetes and obesity rates are expected to continue to climb over the next 30 years, along with coronary artery disease, heart failure, stroke and atrial fibrillation. Similar increases are projected among children.

A recent AHA presidential advisory estimates that by 2050, high blood pressure and obesity will affect more than half the population overall and more than 80% of some subgroups, such as Black and older adults. This is expected to have a direct impact on heart disease and stroke rates.

Most strokes could be prevented if these risk factors were reduced, said Dr. Hugo Aparicio, an associate professor of neurology at the Chobanian and Avedisian School of Medicine at Boston University.

"There are some risk factors we've been better at reducing," like smoking, he said. "But there are other risk factors, such as high blood pressure and obesity, with a stubborn increase, especially among younger adults."

Better education about how to manage those risks delivered early in life will be critical, said Dr. Alex Crystal, chief of cardiology at Mackenzie Health and director of Lawrence Park Cardiology, both in Toronto.

"There appears to be a gap in current public health education efforts regarding preventative health care measures for young people," he said. "This is particularly concerning for chronic diseases like obesity, where early intervention can significantly improve long-term health outcomes."

Teens and young adults don't understand those long-term health risks, such as the development of diabetes or cardiovascular disease early in life, he said. "Prioritizing educational initiatives targeting adolescents and young adults to emphasize the importance of healthy lifestyle choices, including regular exercise, proper nutrition and preventive screenings, and investing in targeted public health campaigns that effectively communicate these long-term health risks could be beneficial."

Innovative technologies and therapies

One way to help people understand those consequences could be to show them their personalized health trajectories using innovative, genetics-related technologies and wearable devices that collect personal health data, Hernandez said.

"If we can create a map of human health and show people where they are on that map, it might help them understand why they need to prevent the conditions that lead to cardiovascular disease," he said. "We could show people how those risks add up."

Genetics will likely play a growing role in identifying and lowering a person's risk for disease, Aparicio said. Polygenetic risk scores, calculated by the presence or absence of gene variants, tell people their chances of developing medical conditions. Gene editing, a field that has emerged over the past decade, allows scientists to change the DNA of an organism, making it theoretically possible to reduce disease risk. While still experimental, it has been successfully used to treat childhood leukemia and has led to the development of new therapies for hemophilia and cystic fibrosis.

Hernandez and Aparicio see an expanding role for gene editing in the years ahead.

For example, someone with a high genetic risk for stroke might be able to lower that risk through gene editing, Hernandez said.

Even if the genes can't be altered, between testing and passive data collected with wearable technology, such as smartwatches that monitor heart rate, heart rhythms and physical activity levels, "we should be able to get good information to understand what that data means for each person's cardiovascular risk profile," Aparicio said.

In the future, wearable technology could evolve beyond smartwatches to include devices implanted in clothing or eyeglasses that gather data not just from individuals but from the environment they're in, Aparicio said. He envisions it being able to alert people to other contributing health factors, such as poor air quality, to help them reduce their personal risks.

"Decades ahead, we should have much more information about our individual health and health trajectories than ever before," Hernandez said. "The combination of where someone stands with cardiovascular risk factors, their genome sequencing and assessments of daily health through passive, digital technology should allow us to better understand their total health and what it might look like over the coming years. It closes the loop for precision health."

Faster treatment may reduce brain damage

Even if stroke rates rise, Aparicio sees a future in which they could do less harm.

He noted a growing area of research into medications that could be given immediately following a stroke, even in an ambulance, and possibly improve stroke outcomes. "They could protect the brain immediately," Aparicio said. "A lot of this is being developed right now."

Other innovative technologies such as mobile CT scanners that can fit in an ambulance also would allow faster diagnosis and treatment of people who have strokes. They are already being used in some areas and will likely become more commonplace, he said. Aparicio envisions a time when other non-invasive technologies, such as MRIs, might also be deployed in this way.

There are also exciting new technologies on the horizon to help resuscitate people who experience cardiac arrest, when the heart suddenly stops working, said Dr. Sarah Perman, an associate professor of emergency medicine at Yale University School of Medicine in New Haven, Connecticut.

ECPR, or extracorporeal cardiopulmonary resuscitation, allows a machine to pump blood for the heart. This allows doctors to preserve organ function as they try to determine what caused the cardiac arrest and treat it. ECPR for in-hospital cardiac arrests has been shown to reduce mortality and is becoming more widespread, but efforts to use it to treat out-of-hospital cardiac arrest are fairly new, gaining traction in Europe and in a few trials in the U.S., Perman said.

According to AHA's advanced life support guidelines update co-authored by Perman, ECPR is reasonable to use on people who have a cardiac arrest but their heart remains unresponsive after extended CPR attempts. But it would require a well-trained and equipped team.

"It's pretty rare right now, still in the early stages," Perman said. "But there are some pretty exciting opportunities here to save lives."

While new technologies can be expensive during the early stages, and available only to a select few, that tends to change over time, Aparicio said.

"Eventually, all of these technologies will likely become cheaper and more accessible and widespread," he said.

Tackling the underlying causes of health disparities

Addressing racial, ethnic, gender and socioeconomic health disparities already a significant problem will become more critical in the coming years, experts say.

The U.S. Census Bureau projects that by 2030, immigration will become the main driver of population growth. By 2060, the Hispanic, Asian and multiracial populations are expected to increase significantly, which is likely to exacerbate underlying social determinants of health such as access to health care and healthy foods and potentially widen disparities.

Black adults already have disproportionately higher rates of high blood pressure and obesity, and they continue to have higher death rates from cardiovascular disease than their white peers. American Indians and Alaska Natives are 50% more likely than white people to be diagnosed with heart disease. Because of shifting demographics, researchers project that within 30 years, Hispanic adults will see the largest total increase in cardiovascular disease and stroke, and Hispanic children will see the largest rises in high blood pressure, diabetes and obesity rates, while Black children are expected to have the highest overall rates of high blood pressure and diabetes.

Identifying and treating risk factors earlier in life critical for all populations will be even more important within these higher-risk groups, Aparicio said.

"Certainly, as the population becomes more diverse, more attention needs to be paid to ensure that all people have the same chance to age with a healthy heart and a healthy brain," he said. "If the root causes of health disparities are not addressed, these populations are going to be more at risk."

Much research has already been done to identify those root causes, which include structural racism and its consequences related to income, employment and housing inequities, less access to health care, healthy foods and safe environments.

Some disparities exist because entire groups of people are left out of the research, Perman said. That's why it's so important when developing new technologies or treatments to ensure equal representation.

"As devices and technologies are being developed, we want to make sure we're not creating more disparities for people with lower socioeconomic status, underrepresented race or sex," she said. "We need to make sure we're being purposeful in how we design studies and who we are including in those studies. And, if we don't have data for women, we need to make sure we are careful if we generalize what we know about men to apply it to women."

Reversing deeply entrenched disparities won't be easy, experts say.

Research shows health care outcomes improve when people are treated by a more diverse medical workforce. Health care professionals who represent the patient community they serve have greater cultural sensitivity to patient needs, fewer language barriers and foster greater trust, which can be critical in getting people to comply with medical advice.

"In communities with the greatest health disparities, we need to take action to improve trust in the health system," Hernandez said.

"And we need to make sure we improve access to health care everywhere."

See original here:
What might next 100 years hold for cardiovascular disease prevention and care? - Japan Today

Posted in Preventative Medicine | Comments Off on What might next 100 years hold for cardiovascular disease prevention and care? – Japan Today

Genomic variants associated with age at diagnosis of childhood-onset type 1 diabetes – Nature.com

Posted: July 11, 2024 at 2:43 am

Patterson CC, Karuranga S, Salpea P, Saeedi P, Dahlquist G, Soltesz G et al. Worldwide estimates of incidence, prevalence and mortality of type 1 diabetes in children and adolescents: Results from the International Diabetes Federation Diabetes Atlas. Diabetes Res Clin Pract. 2019;157:107842.

Article PubMed Google Scholar

Green A, Hede SM, Patterson CC, Wild SH, Imperatore G, Roglic G, et al. Type 1 diabetes in 2017: global estimates of incident and prevalent cases in children and adults. Diabetologia. 2021;64:274150.

Article PubMed PubMed Central Google Scholar

Leete P, Mallone R, Richardson SJ, Sosenko JM, Redondo MJ, Evans-Molina C. The effect of age on the progression and severity of type 1 diabetes: potential effects on disease mechanisms. Curr diabetes Rep. 2018;18:18.

Article CAS Google Scholar

Rakyan VK, Beyan H, Down TA, Hawa MI, Maslau S, Aden D, et al. Identification of Type 1 Diabetes Associated DNA Methylation Variable Positions That Precede Disease Diagnosis. PLOS Genet. 2011;7:e1002300.

Article CAS PubMed PubMed Central Google Scholar

Kumar D, Gemayel NS, Deapen D, Kapadia D, Yamashita PH, Lee M, et al. North- American twins with IDDM. Genetic, etiological, and clinical significance of disease concordance according to age, zygosity, and the interval after diagnosis in first twin. Diabetes. 1993;42:135163.

Article CAS PubMed Google Scholar

Olmos P, AHern R, Heaton DA, Millward BA, Risley D, Pyke DA et al. The significance of the concordance rate for type 1 diabetes in identical twins. Diabetologia. 1988;31:74750.

Kyvik KO, Green A, Beck-Nielsen H. Concordance rates of insulin dependent diabetes mellitus: a population based study of young Danish twins. BMJ. 1995;311:9137.

Article CAS PubMed PubMed Central Google Scholar

Hyttinen V, Kaprio J, Kinnunen L, Koskenvuo M, Tuomilehto J. Genetic liability of type 1 diabetes and the onset age among 22,650 young Finnish twin pairs: a nationwide follow-up study. Diabetes. 2003;52:10525.

Article CAS PubMed Google Scholar

Nerup J, Platz P, Andersen OO, Christy M, Lyngsoe J, Poulsen JE, et al. HL-A antigens and diabetes mellitus. Lancet Lond Engl. 1974;2:8646.

Article CAS Google Scholar

Bell GI, Horita S, Karam JH. A polymorphic locus near the human insulin gene is associated with insulin- dependent diabetes mellitus. Diabetes. 1984;33:17683.

Article CAS PubMed Google Scholar

Barrett JC, Clayton DG, Concannon P, Akolkar B, Cooper JD, Erlich HA, et al. Genome-wide association study and meta- analysis find that over 40 loci affect risk of type 1 diabetes. Nate Genet. 2009;41:7037.

Article CAS Google Scholar

Bradfield JP, Qu HQ, Wang K, Zhang H, Sleiman PM, Kim CE, et al. A genome-wide meta-analysis of six type 1 diabetes cohorts identifies multiple associated loci. PLoS Genet. 2011;7:e1002293.

Article CAS PubMed PubMed Central Google Scholar

Onengut-Gumuscu S, Chen WM, Burren O, Cooper NJ, Quinlan AR, Mychaleckyj JC, et al. Fine mapping of type 1 diabetes susceptibility loci and evidence for colocalization of causal variants with lymphoid gene enhancers. Nat Genet. 2015;47:3816.

Article CAS PubMed PubMed Central Google Scholar

Virgin HW, Todd JA. Metagenomics and personalized medicine. Cell. 2011;147:4456.

Article CAS PubMed PubMed Central Google Scholar

Inshaw JRJ, Cutler AJ, Burren OS, Stefana MI, Todd JA. Approaches and advances in the genetic causes of autoimmune disease and their implications. Nat Immunol. 2018;19:67484.

Article CAS PubMed Google Scholar

Caillat-Zucman S, Garchon HJ, Timsit J, Assan R, Boitard C, Djilali-Saiah I et al. Age- dependent HLA genetic heterogeneity of type 1 insulin-dependent diabetes mellitus. J Clin Invest. 1992;90:224250.

Article CAS PubMed PubMed Central Google Scholar

Howson JMM, Cooper JD, Smyth DJ, Walker NM, Stevens H, She JX, et al. Evidence of Gene-Gene Interaction and Age-at-Diagnosis Effects in Type 1 Diabetes. Diabetes. 2012;61:30127.

Article CAS PubMed PubMed Central Google Scholar

Inshaw JRJ, Cutler AJ, Crouch DJM, Wicker LS, Todd JA. Genetic Variants Predisposing Most Strongly to Type 1 Diabetes Diagnosed Under Age 7 Years Lie Near Candidate Genes That Function in the Immune System and in Pancreatic -Cells. Diabetes Care. 2020;43:16977.

Article CAS PubMed Google Scholar

Syreeni A, Sandholm N, Sidore C, Cucca F, Haukka J, Harjutsalo V, et al. Genome-wide search for genes affecting the age at diagnosis of type 1 diabetes. J Intern Med. 2021;289:66274.

Article CAS PubMed Google Scholar

Moffatt MF, Gut IG, Demenais F, Strachan DP, Bouzigon E, Heath S, et al. A large-scale, consortium-based genomewide association study of asthma. N Engl J Med. 2010;363:121121.

Article CAS PubMed PubMed Central Google Scholar

Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun. 2019;10:1523.

Article PubMed PubMed Central Google Scholar

Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;11:2498504.

Article Google Scholar

Piero J, Bravo , Queralt-Rosinach N, Gutirrez-Sacristn A, Deu-Pons J, Centeno E, et al. DisGeNET: a comprehensive platform integrating information on human disease-associated genes and variants. Nucleic acids Res. 2017;45:D833D839.

Article PubMed Google Scholar

Benjamini Y, Hochberg Y. Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing. J R Stat Soc Ser B Methodol. 1995;57:289300.

Article Google Scholar

Kielevinen V, Turtinen M, Luopajrvi K, Hrknen T, Ilonen J, Knip M, et al. Increased HLA class II risk is associated with a more aggressive presentation of clinical type 1 diabetes. Acta Paediatr. 2023;112:5228.

Article PubMed Google Scholar

Pllnen PM, Lempainen J, Laine AP, Toppari J, Veijola R, Vhsalo P, et al. Characterisation of rapid progressors to type 1 diabetes among children with HLA-conferred disease susceptibility. Diabetologia. 2017;60:128493.

Article PubMed Google Scholar

Bougnres P, Valleron AJ. Causes of early-onset type 1 diabetes: toward data-driven environmental approaches. J Exp Med. 2008;205:29537.

Article PubMed PubMed Central Google Scholar

Bougnres P, LeFur S, Valleron AJ. Early varicella infection is associated with a delayed onset of childhood type 1 diabetes. Diabetes Metabol. 2022;48:101394.

Verlaan DJ, Berlivet S, Hunninghake GM, Madore AM, Larivire M, Moussette S, et al. Allele-Specific Chromatin Remodeling in the ZPBP2/GSDMB/ORMDL3 Locus Associated with the Risk of Asthma and Autoimmune Disease. Am J Hum Genet. 2009;85:37793.

Article CAS PubMed PubMed Central Google Scholar

Kochi Y, Yamada R, Suzuki A, Harley JB, Shirasawa S, Sawada T, et al. A functional variant in FCRL3, encoding Fc receptor-like 3, is associated with rheumatoid arthritis and several autoimmunities. Nat Genet. 2005;37:47885.

Article CAS PubMed PubMed Central Google Scholar

Eskdale J, McNicholl J, Wordsworth P, Jonas B, Huizinga T, Field M, et al. Interleukin-10 microsatellite polymorphisms and IL-10 locus alleles in rheumatoid arthritis susceptibility. Lancet. 1998;352:12823.

Article CAS PubMed Google Scholar

Lin YJ, Wan L, Sheu JJC, Huang CM, Lin CW, Lan YC, et al. G/T polymorphism in the interleukin-2 exon 1 region among Han Chinese systemic lupus erythematosus patients in Taiwan. Clin Immunol. 2008;129:369.

Article CAS PubMed Google Scholar

Sun SC. Deubiquitylation and regulation of the immune response. Nat Rev Immunol. 2008;8:50111.

Article CAS PubMed PubMed Central Google Scholar

Jostins L, Ripke S, Weersma RK, Duerr RH, McGovern DP, Hui KY, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012;491:11924.

Article CAS PubMed PubMed Central Google Scholar

Pothlichet J, Quintana-Murci L. The genetics of innate immunity sensors and human disease. Int Rev Immunol. 2013;32:157208.

Article CAS PubMed Google Scholar

Kahlmann D, Davalos-Misslitz ACM, Ohl L, Stanke F, Witte T, Frster R. Genetic variants of chemokine receptor CCR7 in patients with systemic lupus erythematosus, Sjogrens syndrome and systemic sclerosis. BMC Genet. 2007;8:33.

Article PubMed PubMed Central Google Scholar

Sigurdsson S, Nordmark G, Gring HHH, Lindroos K, Wiman AC, Sturfelt G, et al. Polymorphisms in the Tyrosine Kinase 2 and Interferon Regulatory Factor 5 Genes Are Associated with Systemic Lupus Erythematosus. Am J Hum Genet. 2005;76:52837.

Article CAS PubMed PubMed Central Google Scholar

Read the rest here:
Genomic variants associated with age at diagnosis of childhood-onset type 1 diabetes - Nature.com

Posted in Human Genetics | Comments Off on Genomic variants associated with age at diagnosis of childhood-onset type 1 diabetes – Nature.com

The untold story of the Human Genome Project: How one mans DNA became a pillar of genetics – STAT

Posted: July 11, 2024 at 2:43 am

STAT is co-publishing this investigation by Undark.

They numbered 20 in all 10 men and 10 women who came to a sprawling medical campus in downtown Buffalo, N.Y., to volunteer for what a news report had billed as the worlds biggest science project.

It was the spring of 1997, and the Human Genome Project, an ambitious attempt to read and map a human genetic code in its entirety, was building momentum. The projects scientists had refined techniques to read out the chemical sequences the series of As, Cs, Ts, and Gs that encode the building blocks of life. Now, the researchers just needed suitable human DNA to work with. More exactly, they needed DNA from ordinary people willing to have their genetic information published for the world to see. The volunteers who showed up at Buffalos Roswell Park Cancer Institute had come to answer the call.

To take part in the study was to assume risks that were hard to calculate or predict. If the volunteers were publicly outed, project scientists told them, they might be contacted by the media or by critics of genetic research of whom there were many. If the published sequences revealed a worrisome genetic condition that could be tied back to the volunteers, they might face discrimination from potential employers or insurers. And it was impossible to know how future scientists might use or abuse genetic information. No ones genome had ever been sequenced before.

But the volunteers were also informed that measures had been put in place to protect them: They would remain anonymous, and to minimize the chances that any one of them could be identified based on their unique genetic sequence, the published genome would be a patchwork, derived not from one person but stitched together from the DNA of a large number of volunteers. If we use the blood you donate to prepare DNA samples, the consent form read, we expect that no more than 10% of the eventual DNA sequence will have been obtained from your DNA.

Soon, however, those assurances began to wither. When a much-celebrated working draft of the human genome was published in 2001, the vast majority of it nearly 75 percent came from just one Roswell Park volunteer, an anonymous male donor known as RP11.

To this day, the story of how and why RP11 came to be the centerpiece of one of biologys crowning achievements has largely escaped public scrutiny. Even the scientists who helped orchestrate it disagree about the particulars.

To piece the story together, Undark reviewed more than 100 emails, letters, and other digital documents housed within the History of Genomics Archive at the National Human Genome Research Institute. The documents, provided to Undark through an institutional research collaboration agreement, reveal that the projects sourcing of human genetic material was more ethically fraught than official publications portrayed it to be, and included DNA harvested from a cadaver, and from one of the projects own scientists. The records, along with interviews with many of the projects central figures and with experts in law and bioethics, paint a picture in which high-ranking project officials constrained by their own experimental protocols and accelerated timelines veered from their guiding principles and pushed the boundaries of informed consent.

We were panicking, recalled Aristides Patrinos, who led the Department of Energys efforts in the Human Genome Project and, along with National Human Genome Research Institute director Francis Collins, helped steer the project to completion. So a lot of these issues were not front and center. Thats no excuse, but it was a reason. We were under a lot of pressure to make sure we finished by the time we finished.

The revelations potentially cast a stain on a project that had been extolled for its high ethical standards. Its a big deal when researchers act deceptively, which is to say they do things that they said they werent going to do, or dont do things that they said they were,said Paul Appelbaum, a Columbia University professor who specializes in legal and ethical issues in medicine, psychiatry, and genetics.It has the potential to negatively impact the research enterprise in general, and the benefits that can potentially come from it.

To the extent that an injustice was done, it has propagated far and wide. The genetic sequence that emerged from the Human Genome Projectcontinues to serve as a cornerstone resource of modern biology as a so-called reference genome, used ubiquitously by clinicians and researchers to identify genetic variants, sequence new genomes, and aid tests that determine patients genetic risks. Although the reference genome has undergone several refinements and incorporated new genetic material over the years, RP11 remains at the center of it all, with his DNA still constituting more than 70 percent of the most recent versions.

RP11 is likely unaware that his DNA played, and continues to play, such a pivotal role in the march of genetic science. Project leaders, hamstrung, they say, by a decades-old ethics panel decision, have never attempted to inform him.

Well, I think at this point, it probably would be a good idea to come out in the open and tell everybody what happened, said Patrinos. And give as many specifics as possible.

The Human Genome Project is often compared to the achievement of putting humans on the moon. Launched in 1990 by the Department of Energy and the National Institutes of Health, the project took 13 years and, at the time, around $3 billion to complete. By 2000, scientists had sequenced around 85 percent of the genome, and the milestone was marked with a White House ceremony. President Bill Clinton described it as more than just an epic-making triumph of science and reason. U.K. Prime Minister Tony Blair, who joined by satellite, called it the kind of breakthrough that takes humankind across a frontier and into a new era.

But in 1996, the project was at a crossroads. Francis Collins, then the director of NIHs National Center for Human Genome Research later renamed the National Human Genome Research Institute, or NHGRI was leading the international consortium of laboratories tasked with completing the sequence. Still in his mid-40s, the physicians star was rising. He had succeeded Nobel laureate James Watson years earlier as the centers director, and Barack Obama would later appoint him to the helm of NIH, the worlds largest public funder of biomedical and behavioral research. People who worked with him described him as a brilliant mind and a great communicator a passionate leader with legendary powers of persuasion.

Collins needed all of those qualities to manage the first sequencing of a human genome. It was a staggeringly complex operation. First, the entirety of a persons DNA a molecular sequence of more than 3 billion pairs of nucleotide bases, typically represented as As, Cs, Ts, and Gs had to be broken into fragments roughly 100,000 to 200,000 base pairs long. The fragments were then isolated and cloned, typically by specially preparing each one and inserting it into a bacterium, which copied the fragment as it reproduced. In this way, the teams scientists could make a physical copy of a persons full, albeit fragmented, genome known as a clone library.

Identical clone libraries could then be shipped to different laboratories around the world, allowing many research groups to read the fragments, and piece the sequences back together, in parallel. In a way, it was like distributing sets of the same, extraordinarily difficult jigsaw puzzle to a lineup of the worlds best puzzle solvers: They could work on different sections of the puzzle simultaneously and, if need be, check each others work.

By 1996, clone libraries were already being distributed to a variety of labs. But that spring, project members learned that several of the libraries had been constructed without any informed consent process and with no oversight from institutional review boards, or IRBs bodies that, according to federal policy, should have ethical purview over research with human subjects. Rumors swirled that some of the DNA had come from scientists involved with the project, a scenario that project members speculated could raise ethical questions about consent and invite charges of elitism. Internal project correspondence and tissue bank donation records reviewed by Undark suggest that another DNA source was the cadaver of a 19-year-old who had died by suicide; the family had donated the body to science but had not specifically consented to its use in the Human Genome Project.

It bothered Collins that at least one donors identity was known to project scientists, and that the donor was aware his DNA was being used to create a library. It sounds as if the donor knows who he is, he wrote in an email that March, after being briefed on a clone library that had been constructed at the California Institute of Technology. Thats not the way it should have been done.

In the wake of the revelation, Collins and Patrinos consulted an array of advisers and came up with a new plan, outlined in a joint guidance. They would find new donors and make new clone libraries, under new protocols. Unlike the old libraries, the new ones would be obtained through a double-blind procedure: Scientists involved with the project would not know the identities of the donors, and donors wouldnt know for certain whether their DNA was being used in the project. According to internal correspondence and interviews, project leadership was concerned not only about the genetic privacy of the donors, but also about the possibility that a donor might trumpet their role to the media and create a spectacle.

It seemed like it would create a major distraction from what we wanted to generate, recalled Robert Waterston, who headed one of the five centers that did the majority of the sequencing for the project.

We wanted the human genome, he added meaning a reference that everyone could relate to. Its not Joe Blows genome. Its your genome. Its my genome. Its representative of everybodys genome.

To further protect the two-way confidentiality, the completed representation of the human genome would be a mosaic, assembled from the DNA of not one but multiple donors. The thinking, among the projects inner circle, was that a mosaic would not only complicate attempts to identify donors based on the genetic sequence but also reduce the incentive for wanting to know the donors identities to begin with. If a donors identity did come to light, limiting their contributions might minimize their exposure to potential harms and deter them from attempting to claim property or ownership rights over the published sequence.

In a June 1996 email that appears to have been written by Melvin Simon, who led a cloning operation at Caltech, the scientist told Human Genome Project leadership, including Patrinos, that, as he understood it, no matter what waiver a volunteer is willing to sign, he or she would not lose ownership or property rights. Thus only by a true patchwork or anonymizing approach can it be made extremely difficult to claim such rights, the email read. (Simon confirmed the sentiment behind the email in an interview with Undark.)

Simons Caltech team and a laboratory at the Roswell Park Cancer Institute were each commissioned to create new clone libraries under the new protocols. Soon, however, the plans for a mosaic genome would veer off course, and the Human Genome Project would find itself in a consent conundrum with one person, RP11, caught in the middle.

Pieter de Jong, who led the cloning project at the Roswell Park Cancer Institute, had been behind some of the problematic libraries that had sparked Collins consternation in the spring of 1996. But he had a long history with the project, and he was a foremost expert at DNA cloning. So when the Human Genome Project enacted its new plan, they commissioned him to build at least five new libraries, de Jong recalled to Undark.

This time, de Jong used a lottery-like process to select donors. On March 23, 1997, he ran an advertisement in the Buffalo News seeking 20 volunteers. The edition also featured a front-page story about the project, which de Jong says he helped arrange. In the weeks that followed, the volunteers each came in, met with a genetic counselor, signed a consent form, and donated a few tablespoons of blood. The genetic counselor labeled each blood sample with a number, but created no records linking the samples to their donors.

The 20 samples were then transferred to de Jong, who chose two at random one male and one female to use for clone libraries. The only personal information the facility retained were the names and signatures on the consent forms, which were sealed in envelopes and stored in a locked file cabinet. As a result, it would be virtually impossible for anyone at Roswell Park to determine who the two donors were.

A postdoctoral researcher, Kazutoyo Osoegawa, did most of the work building the first library. Osoegawa was skillful, de Jong recalled, with a knack for coaxing large fragments of DNA from a sample for cloning: The larger the fragments, the more easily scientists could map them for sequencing, and the fewer fragments overall they would have to sequence to finish the job.

By August of 1997, de Jong, Osoegawa and their colleagues had begun distributing the first of the new Roswell Park clone libraries, RP11, and it was a good one with enough fragments for scientists to be fairly certain that they spanned essentially the entire genome, with few missing gaps. A second library was in the works, with more to follow. But, before those libraries could materialize, the Human Genome Projects plans took a turn.

On the evening of Sept. 20, 1998, Francis Collins emailed NHGRI brass, including Jane Peterson, a program director involved with the sequencing effort, and Mark Guyer, the institutes assistant director for scientific coordination, about an unhappy circumstance. I have been feeling uneasy about the RPC11 library ever since Jane uncovered the language that Pieter de Jong used for the consent form, he wrote. (The RP11 library was often referred to as RPC11 or RPCI-11 in correspondence.)

The specific language that unsettled Collins was the passage conveying that no more than 10 percent of the genetic sequence was expected to come from their DNA. And it was resurfacing at an inopportune moment.

The Human Genome Project was in the midst of what Maynard Olson, who led one of the projects sequencing labs, described in an email that September as a de facto drift away from the concept of a genome sequence that is a mosaic of contributions from many individuals. When de Jong crafted the consent language, he was under the impression that 10 new clone libraries would be built and integrated into the completed genome. But now project leaders were lurching toward a strategy that would draw most of the final sequence between 60 and 90 percent from a single clone library. And RP11 was their library of choice.

In his email to his NHGRI colleagues, Collins wrote that the document of general principles he and Patrinos had shared suggested an intent to include several donors but wasnt specific about it, nor does it put a ceiling on the amount of sequence that could come from a single person.

The 10 percent language in the consent form worried him, however. Attempting to reconsent RP11 under new terms would be complicated: RP11 could have been any of the 10 male donors, and all the researchers had to go on were the names on the consent forms. The only way he could think to do it, he wrote, would require asking every volunteer if they objected to the raising of the 10 percent restriction and then holding our breath that none of them do.

Technically, the word expect didnt forbid using RP11 for more than 10 percent of the sequence, Collins wrote, but how far can we push this?

The next month, Collins joined a conference call with de Jong, Roswell Park IRB chair Harold Douglass, and other Roswell Park and NHGRI staff. According to handwritten notes, Collins told them that limiting use of the clone library to 10 percent would devastate the momentum of the project and that there were concerns about recontacting all 10 male donors. The notes indicate that Douglass mentioned the IRB would ask about the benefit of fast-tracking the project, and Collins said there was a medical reason: to find as many genes ASAP to understand disease. (Speaking to Undark, Collins confirmed his participation in the call. He said the notes, taken by a different participant, used phrasing he wouldnt have used, but seemed correct.)

Days later, the Roswell Park IRB met and according to a written summary that was shared with Guyer voted unanimously against any attempts to try to find and reconsent the ten donors. Among the IRBs stated justifications were that the expectation expressed to the donors was not a guarantee, and that attempting to reconsent the 10 male volunteers would be difficult and could jeopardize RP11s anonymity. To delay the project by not expanding the use of RP11s library, the panel added, would itself be unethical, given the number of people who stood to derive health benefits from the timely completion of the human genome. (Douglass declined to comment for this story.)

Recently, Collins spoke to Undark about RP11 and the Human Genome Projects donor sourcing strategies. He was joined by Eric Green, who was also involved with the project and currently leads the National Human Genome Research Institute.

According to Collins and Green, project leaders did initially aim to construct 10 new clone libraries for use in the completed genome. But they soon realized it would be inefficient and chaotic to work with 10 libraries at once. There would be lots of complexities that would come out by having too much blending going on, Green said.

Collins explained that structural differences between individual genomes such as large-scale insertions or deletions of genes can make it difficult to stitch together an accurate sequence from two different human sources. If you go from one person to 10, he said, and then you try to fit the whole thing together, its going to be potentially much more error-prone.

It was primarily those technical challenges, Collins and Green said recently, that prompted the decision to derive most of the genome from a single donor. And RP11 with its well-sized fragments and comprehensive coverage of the genome stood out from the other libraries as the ideal one to work with, they said. Also, Green added, RP11 at the time was further along than any of the other new libraries in the process of being characterized and prepared for sequencing.

But Collins and Greens recollections diverge in key ways from those of other scientists involved in the Human Genome Project. Robert Waterston, for instance, who was among the small circle of researchers who guided project strategy, recalls that the complexities of blending clone libraries were only a minor consideration. Yes, structural differences in DNA could complicate the task of meshing one persons genetic sequence with anothers, he said, but only in certain regions of the genome, such as those marked by repeat sequences that differ in number and complexity from one person to the next.

The bigger factor, said Waterston, was time. And the Human Genome Project was pressed for time, he said, thanks to a man named J. Craig Venter.

In May 1998, the scientist Venter whose nonprofit Institute for Genomic Research had done pilot work for the Human Genome Project launched a venture built to rival the publicly funded initiative. That June, Venter and his colleagues pledged in a Science article that they would sequence a human genome by 2001 years ahead of the Human Genome Projects 2005 target deadline and at a fraction of the cost. The enterprise, known as Celera Genomics Group, set up shop in Rockville, Maryland, just miles from NHGRIs Bethesda headquarters.

Correspondence from that time suggests the news lit a fire under the Human Genome Project. Obviously there would be significant political advantages to getting something out a year earlier than Venter is proposing, provided we can defend its utility, wrote Phil Green, an investigator at the University of Washingtons sequencing center, in an email that was shared with Collins shortly after word of Venters plans began to spread.

Project members worried about the implications of a commercial enterprise owning, and possibly monetizing, the first human genome. For some of them, competition itself and the specter of a stinging defeat seemed to be motivation enough. In an email that September, NHGRIs Peterson described Eric Lander who led the Whitehead/MIT Center for Genome Research, one of the five large centers that sequenced the majority of the genome as having called her in a very depressed mood. Lander believed Venter would have a draft of the human genome done before next summer and will take continual pot shots at us, Peterson wrote. (Lee McGuire, chief communication officer at the Broad Institute, where Eric Lander is a member and founding director, told Undark that Lander was unavailable to be interviewed for this story.)

In a move that was widely reported in the media as being prompted by the Celera announcement, Collins announced that September that the Human Genome Project would aim to finish its genome two years earlier than planned, by 2003, and release a working draft by 2001.

We came into this crush with Celera, and everything just had to get done as quickly as possible, recalled Waterston. The complement of libraries theyd envisioned wasnt ready yet, and it wouldve taken time to make and distribute them, he said. They had to work with what they had, and what they had was RP11.

There just wasnt an alternative, Waterston recalled. We didnt have a second library to go to.

Marco Marra and John McPherson who along with Waterston did much of the preliminary characterization of clone libraries at Washington University similarly remember that it was the dearth of available libraries, more than the challenge of blending them together, that led the project to focus on a single donor.

That aligns with de Jongs recollection. RP11 was a good library, he told Undark, but so were subsequent libraries he built. The problem was that there was no time to wait. (De Jong shared records with Undark indicating that his lab had not yet completed the second of its planned new libraries by September 1998, when the issues around RP11s consent language arose; it is unclear whether the Caltech laboratory had completed and distributed the first of its planned new libraries to sequencing centers by that time, but Waterston recalls they hadnt.)

Although de Jong said he was not heavily involved in discussions of sequencing strategy, he thinks it began to dawn on the scientists how much additional work, and money, would be required to prepare and sequence 10 libraries, rather than one or two. They couldnt potentially keep up the same speed as Venter with his commercial effort if they would have stayed with the original plan, said de Jong. So I think it was mostly because they didnt want to lose the race.

Other members of the Human Genome Project who spoke with Undark expressed similar sentiments, including one of its highest-ranking figures. We got pretty panicky that we were going to lose this, Patrinos said of the competition with Celera. So at that time, we had to follow paths that would get us to the conclusion as fast as possible.

Asked if he felt Celera contributed to a sense of urgency at that time, Collins told Undark he didnt recall that being a factor that the rush, instead, was to get the job done to provide benefits for understanding health and disease. In a follow-up call, Collins clarified: I think Celeras intentions to produce a for-profit human genome sequence was an issue that everybody was fully aware of, so that was in the air, if you will. But he said it was not the driving factor at all in the decision to move as quickly as possible to obtain a complete public sequence.

In any case, on Oct. 27, 1998 five months after Venter launched his rival to the Human Genome Project, a month and a half after the project gave itself a new, ambitious deadline, weeks after Collins concerned email about RP11s consent language, and days after Collins conference call with the chair of the Roswell Park IRB the ethics panel gave Collins and his team carte blanche to dramatically expand the use of RP11s DNA, without telling any of the Roswell Park donors about the change.

That same month Simon and collaborator Hiroaki Shizuya having finished their first Caltech library under the new donor protection protocols told the DOEs Marvin Frazier that although the group had genetic material in hand to begin a second library, they had been informed that there was no longer a great deal of interest in new libraries, and they were instead moving on to new research pursuits.

Archival correspondence suggests the turn of events didnt sit well with all of the lead scientists involved in the project. I was deeply distressed to have the director of a major genome center already start building the case that the informed-consent form for DNA used to build RPC-11 did not really mean what it said, wrote Olson in a November 1998 email to Collins and his University of Washington colleague Phil Green. The ethical, legal, and social issues related to the library sourcing will not go away, he predicted.

Speaking to Undark, Olson said he does not recall which consent language, or which director, he was referring to in his email. But he remembers there being tension between the ethicists and technical experts involved with the project. Some of the ethicists resented the idea that technical considerations should factor into discussions, he said, and a lot of the more technically well-informed participants in the project just actually werent terribly interested in the ethics issues.

Undark invited several biomedical ethicists and legal experts to review the Roswell Park consent form and the IRBs ruling on RP11. Their responses called into question many of the justifications the ethics panel gave for its decision.

The big deal is that the 10% is not just a minor aspect of the consent form, wrote Hank Greely, a Stanford University Professor who works on ethical, legal, and social issues in the biosciences, in an email to Undark. Rather, he noted, it is a substantial part of the argument about confidentiality. Greely said that he didnt find any of the panels justifications convincing. He doesnt think the IRB acted nefariously, but he said that he would not have so hastily dismissed the possibility of attempting to reconsent the volunteers, and that doing so wouldnt necessarily have heightened the risks to the donor. Weve got these 10 names. Lets see if theyre in the phone book, he said, later adding, lets see how locatable they are.

Jonathan Moreno, a professor of medical ethics and health policy at the University of Pennsylvania who declined the offer to review documents but was briefed by Undark on the IRB decision, agreed that the volunteers should have been reconsented.

Appelbaum, the Columbia University legal and ethics specialist, was one of several experts who took issue with the panels interpretation of the 10 percent expectation. I think a reasonable person would take away from that that the intent of the research team was to use no more than 10 percent of his or her genome in the project, he said. And so playing with words in that way, I think, is really not appropriate in this context.

Appelbaum also thought it was odd for Collins, representing a sponsoring agency, to meet directly with an IRB chair on an ethical issue related to work the agency was sponsoring. There is a risk, he said, of exerting undue influence on the oversight process. Bruce Gordon, the assistant vice chancellor for regulatory affairs at the University of Nebraska Medical Center, told Undark that, generally speaking, the best practice would be that funders shouldnt be interacting with the IRB under any circumstance, though he described it as an unspoken rule, and not a strict standard.

Collins said he agreed the conference call was an unusual step, but that the significance of the situation justified it. I counted on the IRB to do what they always do, he said, which is to step back and take up a purely objective view of an ethical question and render their best opinion. I do not believe I put pressure on them at all.

Although ethicists and legal experts who spoke to Undark raised questions about the rationale of the IRBs ruling, many said it was unlikely that RP11 had suffered concrete harms as a result a point also expressed by Collins and other key figures from the Human Genome Project. Protections enacted in the U.S. since the completion of the Human Genome Project make it illegal for employers or health insurers to discriminate based on a persons genetic information. And experts say that without a matching DNA sample, it remains difficult to identify a person based solely on a genetic sequence. With a matching sample, however, it would be straightforward to identify the donor, whether their contribution was 70% or 7%.

I think its fair to say RP11 was probably misled about what was going to happen, said R. Alta Charo, a professor emerita of law and bioethics at the University of Wisconsin Madison. (Like Moreno, Charo declined the offer to review documents, but was briefed by Undark on the IRB decision.) The real question, however, said Charo, is whether the decision made him more identifiable, whether it exposed him to more risk. I dont know how to answer that question.

Appelbaum said it may be truethat RP11s risks werent substantially heightened by the decision to expand the use of his genetic sequence. But it seems to me that thats different from saying that the action wasnt consequential, he said, in the sense that it can be highly consequential, I think, for the research enterprise in this country to make promises to people in signed consent forms, and then violate those promises.

Appelbaum described the episode as illustrative of a long history of deceptions that have contributed to a lack of trust in the research enterprise, especially in minoritized communities. One of the big issues in human subjects research, which has assumed even greater salience in genomic research, has been the issue of trust, he said. If I agree to be in your project, are you leveling with me about whats going to happen to me? And if I agree to donate blood, or some other tissue sample, are you telling me the truth about how its going to be used?

The June 2000 White House ceremony that marked the Human Genome Projects sequencing milestone was a joint ceremony: At the presidential lectern that day, President Clinton was flanked on one side by Francis Collins and on the other by Craig Venter, whose Celera team was also nearing the finish line.

The following winter, the two teams each published landmark genome papers, with the Human Genome Projects report on its draft genome sequence officially appearing in the Feb. 15 issue of the prestigious journal Nature, and Celeras sequencing results appearing in the rival journal Science one day later.

Celera reported that its genome had been assembled from five unnamed donors, one of whom the majority donor Venter later revealed was himself.

Meanwhile, the Human Genome Project was circumspect about the donors behind its published sequence. A table in the Nature paper listed eight clone libraries that were described as having contributed the bulk of the sequence. Among them was RP11, which the table noted accounted for just over 74 percent of the draft genome. The other seven each contributed between 1.6 and 4.3 percent of the total. Additional libraries, neither named nor tallied in the paper, collectively accounted for the remaining 8.4 percent of the sequence.

The paper described the libraries as originating from anonymous DNA donors, according to a lottery-like process like the one used at Roswell Park. What was left unsaid but what consent documents, internal memos, and other records reviewed by Undark reveal is that six of the eight named libraries were the same ones that had raised ethics concerns early in the project: the library sourced from the 19-year-old cadaver; the libraries suspected to have been built with the DNA of project scientists; the libraries whose donors were known to project researchers. Collins and Patrinos had agreed in 1996 to let scientists use those libraries, provided the donors were properly consented, protocols were cleared by IRBs, and the libraries contributed minimally to the final sequence. (Caltechs Simon told Undark that it was a lab technicians husband and not a postdoc, as had been rumored who produced the sperm from which one of his early libraries was built.)

Also left unsaid was that four of the eight libraries had all been derived from the same donor.

Collins and NHGRI director Green could not confirm to Undark how many, if any, of the libraries outside of the top eight had been approved by IRBs. Collins also said he did not know if the family of the 19-year-old tissue donor had been reconsented in accordance with the 1996 guidelines.

Asked if he feels the project should have been more forthright in the 2001 paper about the sourcing of DNA donors, Collins said its always good in hindsight to be transparent and forthright in every way. To be honest though, I dont think in my view, that this was such a major substantial issue that it would have required a deep debate about exactly how to put that forward. He added, I dont believe that individuals were significantly put at risk by the way in which this was laid out. And I hope that doesnt get lost.

To Appelbaum, however, the idea that the Human Genome Projects landmark paper may have misrepresented donor procedures is gravely concerning the kind of transgression that can erode public trust in science more broadly. Perhaps an argument could be made to defend the projects DNA sourcing, Appelbaum said, but Im not sure theres any argument on the other side about covering up what you did when you publish your results. I think youve got to be open about that.

If you made certain decisions along the way, he said, you describe the decisions you made and the justification for them.

The culmination of the Human Genome Project was, in a way, the beginning of a long scientific afterlife for RP11s genetic sequence. A 2010 study, published in the journal Science, analyzed the reference genome and concluded that RP11 was of mixed African and European genetic ancestry, and likely identified as Black or African American.

Perhaps most consequential, however, is that the sequence that emerged from the human genome project has evolved into a foundational resource of modern genetics. It has been revised and improved through the years, each new edition, or reference assembly, augmented with new annotations and fixes.

Deanna Church, who led an international collaboration that managed the reference assemblies in the years following the Human Genome Projects completion, likens them to maps that give scientists a shared coordinate system for describing, comparing, and understanding genetic sequences. Researchers use them to interpret and identify fragments of DNA; clinicians and genetic testing companies use them as benchmarks to determine which genetic variants a person carries. The reference assembly that emerged from the Human Genome Project has become the foundation for all genomic data and databases, wrote the authors of a 2019 opinion piece in the journal Genome Biology.

And to this day, the most widely used reference assemblies continue to derive more than 70 percent of their sequence from a person who did not clearly consent to that level of use.

In recent years, Church and other experts have argued that it is time for a new reference model: The assemblies from the Human Genome Project do not adequately reflect the breadth of human genetic variation, they say. And although those reference assemblies are of exceptional quality by genome standards, a newer sequence, sourced from new DNA and known as the telomere-to-telomere assembly, is both more accurate and more comprehensive.

But a reference assemblys usefulness stems in large part from the information, annotations, and standards that are built on top of it, and it will take time for scientists to duplicate that infrastructure for a new reference genome.

Leslie Biesecker, chief of the Center for Precision Health Research at the NHGRI, estimates it will be three to five years before the community transitions to a new reference. There are so many pieces of machinery that need to be moved forward at the same time in order for that whole system to work.

Stanfords Greely, a lawyer by training, said its conceivable that were RP11 to learn of the outsized role his DNA played in genetic science, he might seek financial compensation. Without wanting to get into the merits of the claims, it could play out kind of the way the Henrietta Lacks story has, said Greely, referring to a Black woman who died of cervical cancer in 1951, and whose cells were harvested for science without her consent. (Lacks family members were recently awarded an undisclosed settlement from Thermo Fisher Scientific, over allegations the company unjustly profited from her cells.) If I were NIH, I would worry hey, if this guy knows, he might sue us or make trouble for us, Greely said.

Documents suggest the architects of the Human Genome Project worried about just such a scenario: a clause in the original consent form used at Roswell Park asserted that, by signing, a donor waived their rights to claim any part of conceivable profits resulting from research performed on the blood and products derived from the blood you donated. But emails sent to NHGRI leadership in July 1997 indicate that when Department of Health and Human Services officials learned of the clause, they argued it ran afoul of a federal regulation that bars consent language that could be construed as a waiver of legal rights. Although RP11 had likely already signed the original version, the waiver was removed from the consent form by that August.

These days, the trim beard Pieter de Jong wore during the days of the Human Genome Project has turned to gray. He now lives near Seattle, where he still runs a small clone library supply operation. This year, to free up space, he finally destroyed three of the five clone libraries he built for the Human Genome Project two of which he says the project never used, and a third that was incorporated into the reference sequence only in the genomes later revisions.

De Jong no longer knows the whereabouts of the 20 consent forms that were collected from the Roswell Park volunteers the only known records that identify the participants by name. Although study protocols stipulated that Roswell Park staff would maintain a chain of custody for the forms, Annie Deck-Miller, director of public relations at the center, now known as the Roswell Park Comprehensive Cancer Center, told Undark in an email that the facility no longer possesses any forms related to de Jongs study. In a subsequent emailed statement, representatives of Roswell Park indicated that documents related to the Human Genome Project were stored onsite for a number of years, as required by federal regulations. They declined to comment further, however, citing a lack of capacity to engage in a review of decisions purported to have taken place in a confidential meeting conducted 26 years ago. Collins and Green say they have never attempted to notify Roswell Park donors about the change to the sequencing plan, and that the IRB decision does not permit them to.

There is, however, one Human Genome Project donor whose whereabouts de Jong knows precisely: the person behind the four clone libraries that accounted for more than 9 percent of the draft sequence.

De Jong recalls that he and a visiting collaborator created those libraries in the summer of 1993. They did it quickly he was in a hurry to apply for grants and get something going and he said there were few ethical guardrails to guide them. De Jong felt it would be inappropriate to solicit DNA from one of his lab workers, so my collaborator my visitor and me, we exchanged, we both tossed up and we gave blood samples for the project.

One of those samples yielded clone libraries that helped spark the 1996 panic over donors: libraries whose origins project leaders worried might leak to the press, de Jong said, but that nonetheless found their way into the worlds first human genome sequence.

It ended up being me, de Jong said, matter-of-factly. The reference genome is maybe 80 percent or 75 percent RP11, and maybe 10 percent me.

If you or someone you know donated to or otherwise participated in the Human Genome Project and you would like to share your story, Undark and STAT would like to hear from you. Contact us at[emailprotected].

Undark is a nonprofit, editorially independent digital magazine exploring the intersection of science and society.

See original here:
The untold story of the Human Genome Project: How one mans DNA became a pillar of genetics - STAT

Posted in Human Genetics | Comments Off on The untold story of the Human Genome Project: How one mans DNA became a pillar of genetics – STAT

Metabolic gene function discovery platform GeneMAP identifies SLC25A48 as necessary for mitochondrial choline import – Nature.com

Posted: July 11, 2024 at 2:43 am

Prosser, G. A., Larrouy-Maumus, G. & de Carvalho, L. P. S. Metabolomic strategies for the identification of new enzyme functions and metabolic pathways. EMBO Rep. 15, 657669 (2014).

Article PubMed PubMed Central Google Scholar

Pizzagalli, M. D., Bensimon, A. & Superti-Furga, G. A guide to plasma membrane solute carrier proteins. FEBS J. 288, 27842835 (2021).

Article PubMed Google Scholar

Wiedmer, T., Ingles-Prieto, A., Goldmann, U., Steppan, C. M. & Superti-Furga, G. Accelerating SLC transporter research: streamlining knowledge and validated tools. Clin. Pharmacol. Ther. 112, 439442 (2022).

Article PubMed PubMed Central Google Scholar

Csar-Razquin, A. et al. A call for systematic research on solute carriers. Cell 162, 478487 (2015).

Article PubMed Google Scholar

Shi, X. et al. Combinatorial GxGxE CRISPR screen identifies SLC25A39 in mitochondrial glutathione transport linking iron homeostasis to OXPHOS. Nat. Commun. 13, 2483 (2022).

Article CAS PubMed PubMed Central Google Scholar

Kenny, T. C. et al. Integrative genetic analysis identifies FLVCR1 as a plasma-membrane choline transporter in mammals. Cell Metab. 35, 10571071.e12 (2023).

Article CAS PubMed PubMed Central Google Scholar

Wang, Y. et al. SLC25A39 is necessary for mitochondrial glutathione import in mammalian cells. Nature 599, 136140 (2021).

Article CAS PubMed PubMed Central Google Scholar

Unlu, G. et al. Metabolic-scale gene activation screens identify SLCO2B1 as a heme transporter that enhances cellular iron availability. Mol. Cell 82, 28322843.e7 (2022).

Article CAS PubMed PubMed Central Google Scholar

Dvorak, V. et al. An overview of cell-based assay platforms for the solute carrier family of transporters. Front. Pharmacol. 12, 722889 (2021).

Article CAS PubMed PubMed Central Google Scholar

Barroso, I. & McCarthy, M. I. The genetic basis of metabolic disease. Cell 177, 146161 (2019).

Article PubMed PubMed Central Google Scholar

Rios, S. et al. Plasma metabolite profiles associated with the World Cancer Research Fund/American Institute for Cancer Research lifestyle score and future risk of cardiovascular disease and type 2 diabetes. Cardiovasc. Diabetol. 22, 252 (2023).

Article CAS PubMed PubMed Central Google Scholar

Wang, F. et al. Plasma metabolomic profiles associated with mortality and longevity in a prospective analysis of 13,512 individuals. Nat. Commun. 14, 5744 (2023).

Article CAS PubMed PubMed Central Google Scholar

Schlosser, P. et al. Genetic studies of paired metabolomes reveal enzymatic and transport processes at the interface of plasma and urine. Nat. Genet. 55, 9951008 (2023).

Article CAS PubMed PubMed Central Google Scholar

Yin, X. et al. Genome-wide association studies of metabolites in Finnish men identify disease-relevant loci. Nat. Commun. 13, 1644 (2022).

Article CAS PubMed PubMed Central Google Scholar

Chen, Y. et al. Genomic atlas of the plasma metabolome prioritizes metabolites implicated in human diseases. Nat. Genet. 55, 4453 (2023).

Article PubMed PubMed Central Google Scholar

Surendran, P. et al. Rare and common genetic determinants of metabolic individuality and their effects on human health. Nat. Med. 28, 23212332 (2022).

Article CAS PubMed PubMed Central Google Scholar

Yin, X. et al. Integrating transcriptomics, metabolomics, and GWAS helps reveal molecular mechanisms for metabolite levels and disease risk. Am. J. Hum. Genet. 109, 17271741 (2022).

Lotta, L. A. et al. A cross-platform approach identifies genetic regulators of human metabolism and health. Nat. Genet. 53, 5464 (2021).

Article CAS PubMed PubMed Central Google Scholar

Gamazon, E. R. et al. A gene-based association method for mapping traits using reference transcriptome data. Nat. Genet. 47, 10911098 (2015).

Article PubMed PubMed Central Google Scholar

Gusev, A. et al. Integrative approaches for large-scale transcriptome-wide association studies. Nat. Genet. 48, 245252 (2016).

Article CAS PubMed PubMed Central Google Scholar

Porcu, E. et al. Mendelian randomization integrating GWAS and eQTL data reveals genetic determinants of complex and clinical traits. Nat. Commun. 10, 3300 (2019).

Article PubMed PubMed Central Google Scholar

Barbeira, A. N. et al. Exploring the phenotypic consequences of tissue specific gene expression variation inferred from GWAS summary statistics. Nat. Commun. 9, 1825 (2018).

Article PubMed PubMed Central Google Scholar

Zhou, D. et al. A unified framework for joint-tissue transcriptome-wide association and Mendelian randomization analysis. Nat. Genet. 52, 12391246 (2020).

Article CAS PubMed PubMed Central Google Scholar

Mogil, L. S. et al. Genetic architecture of gene expression traits across diverse populations. PLoS Genet. 14, e1007586 (2018).

Article PubMed PubMed Central Google Scholar

Storey, J. D. & Tibshirani, R. Statistical significance for genomewide studies. Proc. Natl Acad. Sci. USA 100, 94409445 (2003).

Article PubMed PubMed Central Google Scholar

Jumper, J. et al. Highly accurate protein structure prediction with AlphaFold. Nature 596, 583589 (2021).

Article CAS PubMed PubMed Central Google Scholar

Cheng, J. et al. Accurate proteome-wide missense variant effect prediction with AlphaMissense. Science 381, eadg7492 (2023).

Article CAS PubMed Google Scholar

Rentzsch, P., Schubach, M., Shendure, J. & Kircher, M. CADD-Spliceimproving genome-wide variant effect prediction using deep learning-derived splice scores. Genome Med. 13, 31 (2021).

Article CAS PubMed PubMed Central Google Scholar

Jeong, H., Tombor, B., Albert, R., Oltval, Z. N. & Barabsl, A. L. The large-scale organization of metabolic networks. Nature 407, 651654 (2000).

Article CAS PubMed Google Scholar

Stacey, D. et al. ProGeM: a framework for the prioritization of candidate causal genes at molecular quantitative trait loci. Nucleic Acids Res. 47, e3 (2019).

Article CAS PubMed Google Scholar

Li, F., Chen, Y., Anton, M. & Nielsen, J. GotEnzymes: an extensive database of enzyme parameter predictions. Nucleic Acids Res. 51, D583D586 (2023).

Article CAS PubMed Google Scholar

Aguet, F. et al. The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science 369, 13181330 (2020).

Article CAS Google Scholar

Zeisel, S. H. & Da Costa, K. A. Choline: an essential nutrient for public health. Nutr. Rev. 67, 615623 (2009).

Article PubMed Google Scholar

Combs, G. F. Jr. & McClung, J. P. (eds) in The Vitamins 523589 (Academic Press, 2022).

Kennedy, E. P. & Weiss, S. B. The function of cytidine coenzymes in the biosynthesis of phospholipides. J. Biol. Chem. 222, 193214 (1956).

Article CAS PubMed Google Scholar

Ducker, G. S. & Rabinowitz, J. D. One-carbon metabolism in health and disease. Cell Metab. 25, 2742 (2017).

Article PubMed Google Scholar

Dragolovich, J. Dealing with salt stress in animal cells: the role and regulation of glycine betaine concentrations. J. Exp. Zool. 268, 139144 (1994).

Article CAS Google Scholar

Ueland, P. M. Choline and betaine in health and disease. J. Inherit. Metab. Dis. 34, 315 (2011).

Article CAS PubMed Google Scholar

Chen, W. W., Freinkman, E., Wang, T., Birsoy, K. & Sabatini, D. M. Absolute quantification of matrix metabolites reveals the dynamics of mitochondrial metabolism. Cell 166, 13241337.e11 (2016).

Article CAS PubMed PubMed Central Google Scholar

Palmieri, F. The mitochondrial transporter family SLC25: identification, properties and physiopathology. Mol. Aspects Med. 34, 465484 (2013).

Article CAS PubMed Google Scholar

Chen, S. et al. A genome-wide mutational constraint map quantified from variation in 76,156 human genomes. Preprint at bioRxiv https://doi.org/10.1101/2022.03.20.485034 (2022).

Son, Y., Kenny, T. C., Khan, A., Birsoy, K. & Hite, R. K. Structural basis of lipid head group entry to the Kennedy pathway by FLVCR1. Nature 629, 710716 (2024).

Article CAS PubMed Google Scholar

Ri, K. et al. Molecular mechanism of choline and ethanolamine transport in humans. Nature 630, 501508 (2024).

Verkerke, A. R. P., Shi, X., Abe, I., Gerszten, R. E. & Kajimura, S. Mitochondrial choline import regulates purine nucleotide pools via SLC25A48. Preprint at bioRxiv https://doi.org/10.1101/2023.12.31.573776 (2024).

Patil, S. et al. SLC25A48 is a human mitochondrial choline transporter. Preprint at medRxiv https://doi.org/10.1101/2023.12.04.23299390 (2023).

Ardlie, K. G. et al. The GenotypeTissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science 348, 648660 (2015).

Article Google Scholar

Giambartolomei, C. et al. Bayesian test for colocalisation between pairs of genetic association studies using summary statistics. PLoS Genet. 10, e1004383 (2014).

Article PubMed PubMed Central Google Scholar

Lonsdale, J. et al. The GenotypeTissue Expression (GTEx) project. Nat. Genet. 45, 580585 (2013).

Article CAS Google Scholar

Yavorska, O. O. & Burgess, S. MendelianRandomization: an R package for performing Mendelian randomization analyses using summarized data. Int. J. Epidemiol. 46, 17341739 (2017).

Article PubMed PubMed Central Google Scholar

Burgess, S., Thompson, S. G. & CRP CHD Genetics Collaboration. Avoiding bias from weak instruments in Mendelian randomization studies. Int. J. Epidemiol. 40, 755764 (2011).

Foley, C. N. et al. A fast and efficient colocalization algorithm for identifying shared genetic risk factors across multiple traits. Nat. Commun. 12, 764 (2021).

Article CAS PubMed PubMed Central Google Scholar

Wu, P. et al. Mapping ICD-10 and ICD-10-CM codes to phecodes: workflow development and initial evaluation. JMIR Med. Inform. 7, e14325 (2019).

Article PubMed PubMed Central Google Scholar

Wu, M. C. et al. Rare-variant association testing for sequencing data with the sequence kernel association test. Am. J. Hum. Genet. 89, 8293 (2011).

Article PubMed PubMed Central Google Scholar

Choi, S. W. & OReilly, P. F. PRSice-2: polygenic risk score software for biobank-scale data. Gigascience 8, giz082 (2019).

Read more from the original source:
Metabolic gene function discovery platform GeneMAP identifies SLC25A48 as necessary for mitochondrial choline import - Nature.com

Posted in Human Genetics | Comments Off on Metabolic gene function discovery platform GeneMAP identifies SLC25A48 as necessary for mitochondrial choline import – Nature.com

Transhumanist author predicts artificial super-intelligence, immortality, and the Singularity by 2045 – TechSpot

Posted: July 11, 2024 at 2:42 am

Dystopian Kurzweil: As Big Tech continues frantically pushing AI development and funding, many users have become concerned about the outcome and dangers of the latest AI advancements. However, one man is more than sold on AI's ability to bring humanity to its next evolutionary level.

Raymond Kurzweil is a well-known computer scientist, author, and artificial intelligence enthusiast. Over the years, he has promoted radical concepts such as transhumanism and technological singularity, where humanity and advanced technology merge to create an evolved hybrid species. Kurzweil's latest predictions on AI and the future of tech essentially double down on twenty-year-old predictions.

In a recent interview with the Guardian, Kurzweil introduced his latest book, "The Singularity Is Nearer," a sequel to his bestselling 2005 book, "The Singularity Is Near: When Humans Transcend Biology." Kurzweil predicted that AI would reach human-level intelligence by 2029, with the merging between computers and humans (the singularity) happening in 2045. Now that AI has become the most talked-about topic, he believes his predictions still hold.

Kurzweil believes that in five years, machine learning will possess the same abilities as the most skilled humans in almost every field. A few "top humans" capable of writing Oscar-level screenplays or conceptualizing deep new philosophical insights will still be able to beat AI, but everything will change when artificial general intelligence (AGI) finally surpasses humans at everything.

Bringing large language models (LLM) to the next level simply requires more computing power. Kurzweil noted that the computing paradigm we have today is "basically perfect," and it will just get better and better over time. The author doesn't believe that quantum computing will turn the world upside down. He says there are too many ways to continue improving modern chips, such as 3D and vertically stacked designs.

Kurzweil predicts that machine-learning engineers will eventually solve the issues caused by hallucinations, uncanny AI-generated images, and other AI anomalies with more advanced algorithms trained on more data. The singularity is still happening and will arrive once people start merging their brains with the cloud. Advancements in brain-computer interfaces (BCIs) are already occurring. These BCIs, eventually comprised of nanobots "noninvasively" entering the brain through capillaries, will enable humans to possess a combination of natural and cybernetic intelligence.

Kurzweil's imaginative nature as a book author and enthusiastic transhumanist is plain to see. Science still hasn't discovered an effective way to deliver drugs directly into the brain because human physiology doesn't work the way the futurist thinks. However, he remains confident that nanobots will make humans "a millionfold" more intelligent within the next twenty years.

Kurzweil concedes that AI will radically change society and create a global automated economy. People will lose jobs but will also adapt to new employment roles and opportunities advanced tech brings. A universal basic income will also ease the pain. He expects the first tangible transformative plans will emerge in the 2030s. The inevitable Singularity will enable humans to live forever or extend our living prospects indefinitely. Technology could even resurrect the dead through AI avatars and virtual reality.

Kurzweil says people are misdirecting their worries regarding AI.

"It is not going to be us versus AI: AI is going inside ourselves," he said. "It will allow us to create new things that weren't feasible before. It'll be a pretty fantastic future."

View post:
Transhumanist author predicts artificial super-intelligence, immortality, and the Singularity by 2045 - TechSpot

Posted in Transhumanism | Comments Off on Transhumanist author predicts artificial super-intelligence, immortality, and the Singularity by 2045 – TechSpot

Stem Cell-Derived Therapy Shows Promise Against Treatment-Resistant Liver Cancer – University of California San Diego

Posted: July 11, 2024 at 2:42 am

Researchers at University of California San Diego have found that the most common form of liver cancer one with a high mortality rate can be better targeted and treated using an innovative new stem cell-derived therapy, according to a recently published study in Cell Stem Cell.

The treatment, not yet studied in patients, involves the lab engineering of natural killer (NK) cells white blood cells that destroy tumor cells to more effectively battle hepatocellular carcinoma (HCC), one of the most treatment-resistant types of solid tumor.

Genetically modified NK-cell therapy doesn't require personalization like chimeric antigen receptor (CAR)-expressing T-cell therapy a relatively new, personalized form of immunotherapy. That means an NK-cell therapy could be mass produced and shelf-ready for patients, who could begin therapy without delay, their new research shows.

To some extent all tumor cells perhaps hepatocellular carcinoma more so inhibit immune cells that try to kill them, said UC San Diego School of Medicine Professor Dan Kaufman, M.D., Ph.D., lead author on the study, director of the Sanford Advanced Therapy Center at the universitys Sanford Stem Cell Institute and Moores Cancer Center member.

This is one key reason why some immunotherapies like CAR T cells have been less successful for solid tumors than for blood cancers the immunosuppressive tumor microenvironment.

Kaufman and his team produced stem cell-derived NK cells in which the receptor for transforming growth factor beta (TGF-) a protein that impairs immune function was disabled. HCC tumors and the liver in general contain copious amounts of the substance, which both inhibits the immune cell activity and allows cancer to proliferate.

They found that typical NK cells without the disabled receptor, like CAR T cells, were not very effective in battling the cancer. These are pretty resistant tumors when we put them in mice, they grow and kill the mice, he said. The five-year survival rate for HCC in humans is less than 20 percent.

When researchers tested the modified NK cells against the cancer, however, we got very good anti-tumor activity and significantly prolonged survival, he noted.

These studies demonstrate that it is crucial to block transforming growth factor beta at least for NK cells, but I also think its true for CAR T cells, Kaufman said. If you unleash NK cells by blocking this inhibitory pathway, they should kill cancer quite nicely.

Kaufman anticipates that his teams discovery will manifest itself in the clinical trials of many research groups and companies whether theyre working on CAR T-cell or NK-cell therapies, battling hepatocellular carcinoma or other challenging types of solid tumors.

Anyone developing such therapies for solid tumors should be working to inhibit transforming growth factor beta activity to improve cancer-killing and attain effective anti-tumor activity, he said.

Co-authors of this study include Jaya Lakshmi Thangaraj; Michael Coffey; and Edith Lopez, all of the Division of Regenerative Medicine at UC San Diegos School of Medicine.

This work was made possible by the NIH/NCI grants U01CA217885, P30CA023100 (administrative supplement), and the Sanford Stem Cell Institute at the University of California San Diego.

Disclosures: Kaufman is a co-founder and advisor to Shoreline Biosciences and has an equity interest in the company. He also consults VisiCELL Medical and RedC Bio, for which he receives income and/or equity. Studies in this work are not related to work of those companies. The terms of these arrangements have been reviewed and approved by the University of California San Diego in accordance with its conflict-of-interest policies. The remaining authors declare no competing interest.

The UC San Diego Sanford Stem Cell Institute (SSCI) is a global leader in regenerative medicine and a hub for stem cell science and innovation in space. SSCI aims to catalyze critical basic research discoveries, translational advances and clinical progress terrestrially and in space to develop and deliver novel therapeutics to patients.

Visit link:
Stem Cell-Derived Therapy Shows Promise Against Treatment-Resistant Liver Cancer - University of California San Diego

Posted in Cell Medicine | Comments Off on Stem Cell-Derived Therapy Shows Promise Against Treatment-Resistant Liver Cancer – University of California San Diego

A new way to miniaturize cell production for cancer treatment – MIT News

Posted: July 11, 2024 at 2:42 am

Researchers from the Singapore-MIT Alliance for Research and Technology (SMART), MITs research enterprise in Singapore, have developed a novel way to produce clinical doses of viable autologous chimeric antigen receptor (CAR) T-cells in a ultra-small automated closed-system microfluidic chip, roughly the size of a pack of cards.

This is the first time that a microbioreactor is used to produce autologous cell therapy products. Specifically, the new method was successfully used to manufacture and expand CAR-T cells that are as effective as cells produced using existing systems in a smaller footprint and less space, and using fewer seeding cell numbers and cell manufacturing reagents. This could lead to more efficient and affordable methods of scaling-out autologous cell therapy manufacturing, and could even potentially enable point-of-care manufacturing of CAR T-cells outside of a laboratory setting such as in hospitals and wards.

CAR T-cell therapy manufacturing requires the isolation, activation, genetic modification, and expansion of a patients own T-cells to kill tumor cells upon reinfusion into the patient. Despite how cell therapies have revolutionized cancer immunotherapy, with some of the first patients who received autologous cell therapies in remission for more than 10 years, the manufacturing process for CAR-T cells has remained inconsistent, costly, and time-consuming. It can be prone to contamination, subject to human error, and requires seeding cell numbers that are impractical for smaller-scale CAR T-cell production. These challenges create bottlenecks that restrict both the availability and affordability of these therapies despite their effectiveness.

In a paper titled A high-density microbioreactor process designed for automated point-of-care manufacturing of CAR T cells published in the journal Nature Biomedical Engineering, SMART researchers detailed their breakthrough: Human primary T-cells can be activated, transduced, and expanded to high densities in a 2-mililiter automated closed-system microfluidic chip to produce over 60 million CAR T-cells from donors with lymphoma, and over 200 million CAR T-cells from healthy donors. The CAR T-cells produced using the microbioreactor are as effective as those produced using conventional methods, but in a smaller footprint and less space, and with fewer resources. This translates to lower cost of goods manufactured (COGM), and potentially to lower costs for patients.

The groundbreaking research was led by members of the Critical Analytics for Manufacturing Personalized-Medicine (CAMP) interdisciplinary research group at SMART. Collaborators include researchers from the Duke-NUS Medical School; the Institute of Molecular and Cell Biology at the Agency for Science, Technology and Research; KK Womens and Childrens Hospital; and Singapore General Hospital.

This advancement in cell therapy manufacturing could ultimately offer a point-of-care platform that could substantially increase the number of CAR T-cell production slots, reducing the wait times and cost of goods of these living medicines making cell therapy more accessible to the masses. The use of scaled-down bioreactors could also aid process optimization studies, including for different cell therapy products, says Michael Birnbaum, co-lead principal investigator at SMART CAMP, associate professor of biological engineering at MIT, and a co-senior author of the paper.

With high T-cell expansion rates, similar total T-cell numbers could be attained with a shorter culture period in the microbioreactor (seven to eight days) compared to gas-permeable culture plates (12 days), potentially shortening production times by 30-40 percent. The CAR T-cells from both the microfluidic bioreactor and gas-permeable culture plates only showed subtle differences in cell quality. The cells were equally functional in killing leukemia cells when tested in mice.

This new method suggests that a dramatic miniaturization of current-generation autologous cell therapy production is feasible, with the potential of significantly alleviating manufacturing limitations of CAR T-cell therapy. Such a miniaturization would lay the foundation for point-of-care manufacturing of CAR T-cells and decrease the good manufacturing practice (GMP) footprint required for producing cell therapies which is one of the primary drivers of COGM, says Wei-Xiang Sin, research scientist at SMART CAMP and first author of the paper.

Notably, the microbioreactor used in the research is a perfusion-based, automated, closed system with the smallest footprint per dose, smallest culture volume and seeding cell number, as well as the highest cell density and level of process control attainable. These microbioreactors previously only used for microbial and mammalian cell cultures were originally developed at MIT and have been advanced to commercial production by Millipore Sigma.

The small starting cell numbers required, compared to existing larger automated manufacturing platforms, means that smaller amounts of isolation beads, activation reagents, and lentiviral vectors are required per production run. In addition, smaller volumes of medium are required (at least tenfold lower than larger automated culture systems) owing to the extremely small culture volume (2 milliliters; approximately 100-fold lower than larger automated culture systems) which contributes to significant reductions in reagent cost. This could benefit patients, especially pediatric patients who have low or insufficient T-cell numbers to produce therapeutic doses of CAR T-cells.

Moving forward, SMART CAMP is working on further engineering sampling and/or analytical systems around the microbioreactor so that CAR-T production can be performed with reduced labor and out of a laboratory setting, potentially facilitating the decentralized bedside manufacturing of CAR T-cells. SMART CAMP is also looking to further optimize the process parameters and culture conditions to improve cell yield and quality for future clinical use.

The research was conducted by SMART and supported by the National Research Foundation Singapore under its Campus for Research Excellence and Technological Enterprise (CREATE) program.

Follow this link:
A new way to miniaturize cell production for cancer treatment - MIT News

Posted in Cell Medicine | Comments Off on A new way to miniaturize cell production for cancer treatment – MIT News

Longeveron scores RMAT designation for Alzheimers cell therapy – Pharmaceutical Technology

Posted: July 11, 2024 at 2:42 am

The US Food and Drug Administration (FDA) has awarded a Regenerative Medicine Advanced Therapy (RMAT) designation to Longeverons Lomecel-B for treating mild Alzheimers Disease.

An RMAT designation provides the benefits of both fast track and breakthrough designations, thereby, allowing for accelerated approval based on surrogate or intermediate endpoints. It also allows for multiple meetings with the FDA to expedite drug development.

Following the news, Longeveron stock was up by over 20% in pre-market trading today, compared to the market close on 9 July.

Lomecel-B is made up of human mesenchymal stem cells, derived from the bone marrow of healthy human donors. It is being evaluated as a treatment for hypoplastic left heart syndrome, and ageing-related frailty, in addition to Alzheimers disease.

Lomecel-B met the primary safety endpoint in a Phase II CLERMIND trial (NCT05233774), which enrolled 50 patients with mild Alzheimers. There were three reported serious events within 30 days of Lomecel-B administration, one in each of three different treatment dose groups, compared to none in the placebo group.

There were no incidences of hypersensitivity, notable changes in laboratory evaluations and electrocardiogram (EKG), or cases of amyloid-related imaging abnormalities (ARIA). Additionally, no clinically asymptomatic microhaemorrhages were noted on MRI. The therapy also improved cognitive functions and reduced neuroinflammation assessed by diffusion tensor imaging, an MRI imaging technique.

Access the most comprehensive Company Profiles on the market, powered by GlobalData. Save hours of research. Gain competitive edge.

Your download email will arrive shortly

We are confident about the unique quality of our Company Profiles. However, we want you to make the most beneficial decision for your business, so we offer a free sample that you can download by submitting the below form

Country * UK USA Afghanistan land Islands Albania Algeria American Samoa Andorra Angola Anguilla Antarctica Antigua and Barbuda Argentina Armenia Aruba Australia Austria Azerbaijan Bahamas Bahrain Bangladesh Barbados Belarus Belgium Belize Benin Bermuda Bhutan Bolivia Bonaire, Sint Eustatius and Saba Bosnia and Herzegovina Botswana Bouvet Island Brazil British Indian Ocean Territory Brunei Darussalam Bulgaria Burkina Faso Burundi Cambodia Cameroon Canada Cape Verde Cayman Islands Central African Republic Chad Chile China Christmas Island Cocos Islands Colombia Comoros Congo Democratic Republic of the Congo Cook Islands Costa Rica Cte d"Ivoire Croatia Cuba Curaao Cyprus Czech Republic Denmark Djibouti Dominica Dominican Republic Ecuador Egypt El Salvador Equatorial Guinea Eritrea Estonia Ethiopia Falkland Islands Faroe Islands Fiji Finland France French Guiana French Polynesia French Southern Territories Gabon Gambia Georgia Germany Ghana Gibraltar Greece Greenland Grenada Guadeloupe Guam Guatemala Guernsey Guinea Guinea-Bissau Guyana Haiti Heard Island and McDonald Islands Holy See Honduras Hong Kong Hungary Iceland India Indonesia Iran Iraq Ireland Isle of Man Israel Italy Jamaica Japan Jersey Jordan Kazakhstan Kenya Kiribati North Korea South Korea Kuwait Kyrgyzstan Lao Latvia Lebanon Lesotho Liberia Libyan Arab Jamahiriya Liechtenstein Lithuania Luxembourg Macao Macedonia, The Former Yugoslav Republic of Madagascar Malawi Malaysia Maldives Mali Malta Marshall Islands Martinique Mauritania Mauritius Mayotte Mexico Micronesia Moldova Monaco Mongolia Montenegro Montserrat Morocco Mozambique Myanmar Namibia Nauru Nepal Netherlands New Caledonia New Zealand Nicaragua Niger Nigeria Niue Norfolk Island Northern Mariana Islands Norway Oman Pakistan Palau Palestinian Territory Panama Papua New Guinea Paraguay Peru Philippines Pitcairn Poland Portugal Puerto Rico Qatar Runion Romania Russian Federation Rwanda Saint Helena, Ascension and Tristan da Cunha Saint Kitts and Nevis Saint Lucia Saint Pierre and Miquelon Saint Vincent and The Grenadines Samoa San Marino Sao Tome and Principe Saudi Arabia Senegal Serbia Seychelles Sierra Leone Singapore Slovakia Slovenia Solomon Islands Somalia South Africa South Georgia and The South Sandwich Islands Spain Sri Lanka Sudan Suriname Svalbard and Jan Mayen Swaziland Sweden Switzerland Syrian Arab Republic Taiwan Tajikistan Tanzania Thailand Timor-Leste Togo Tokelau Tonga Trinidad and Tobago Tunisia Turkey Turkmenistan Turks and Caicos Islands Tuvalu Uganda Ukraine United Arab Emirates US Minor Outlying Islands Uruguay Uzbekistan Vanuatu Venezuela Vietnam British Virgin Islands US Virgin Islands Wallis and Futuna Western Sahara Yemen Zambia Zimbabwe Kosovo

Industry * Academia & Education Aerospace, Defense & Security Agriculture Asset Management Automotive Banking & Payments Chemicals Construction Consumer Foodservice Government, trade bodies and NGOs Health & Fitness Hospitals & Healthcare HR, Staffing & Recruitment Insurance Investment Banking Legal Services Management Consulting Marketing & Advertising Media & Publishing Medical Devices Mining Oil & Gas Packaging Pharmaceuticals Power & Utilities Private Equity Real Estate Retail Sport Technology Telecom Transportation & Logistics Travel, Tourism & Hospitality Venture Capital

Tick here to opt out of curated industry news, reports, and event updates from Pharmaceutical Technology.

Submit and download

The global market for Alzheimers treatments is expected to be worth over $15.9bn in 2030, as per a GlobalData report. The field is dominated by amyloid-targeting therapies, with Eli Lillys Kisunla (donanemab) being the latest FDA-approved therapy in this class.

GlobalData is the parent company of Pharmaceutical Technology.

The news comes as a welcome reprieve for the company after it discontinued a Phase II trial of Lomecel-B in ageing-related frailty in Japan, in February. The decision was a strategic move by the Longeveron to focus on developing Lomecel-B in hypoplastic left heart syndrome, a rare congenital heart defect.

The company is evaluating Lomecel-B in a Phase IIb ELPIS II trial (NCT04925024) in hypoplastic left heart syndrome patients under 12 years of age. The trial plans to recruit 38 participants, with enrolment expected to be completed by the end of the year.

Stem cell treatments are a relatively new area in the Alzheimers treatment space. Regeneration Biomedical is evaluating its autologous stem cell treatment, RB-ADSC, in mild-to-moderate Alzheimers in a Phase I trial (NCT05667649).

Cell & Gene Therapy coverage on Pharmaceutical Technology is supported by Cytiva.

Editorial content is independently produced and follows thehighest standardsof journalistic integrity. Topic sponsors are not involved in the creation of editorial content.

Give your business an edge with our leading industry insights.

See more here:
Longeveron scores RMAT designation for Alzheimers cell therapy - Pharmaceutical Technology

Posted in Cell Medicine | Comments Off on Longeveron scores RMAT designation for Alzheimers cell therapy – Pharmaceutical Technology

City of Hope and Mount Sinai scientists first – EurekAlert

Posted: July 11, 2024 at 2:42 am

video:

3-D image of a mouse kidney that has received human islet grafts. Beta cells are visible in green; red reveals the entire mouse kidney.

Credit: City of Hope/Mount Sinai Health System

LOS ANGELES and NEW YORK In preclinical studies, a team of researchers from City of Hope in Los Angeles and Mount Sinai Health System in New York reports new findings on a therapeutic combination that regenerated human insulin-producing beta cells,providing a possible new treatment for diabetes. The findings were published today in Science Translational Medicine.

This work, led by Andrew F. Stewart, M.D., Irene and Dr. Arthur M. Fishberg Professor of Medicine and Director of the Mount Sinai Diabetes, Obesity and Metabolism Institute, began at the Icahn School of Medicine at Mount Sinai in 2015. The studies were a team effort. Adolfo Garcia-Ocaa, Ph.D., formerly a professor at Mount Sinai who is now at City of Hope, a leading research center for diabetes and one of the largest cancer research and treatment organizations in the United States, and is the Ruth B. & Robert K. Lanman Chair in Gene Regulation and Drug Discovery Research and chair of theDepartment of Molecular & Cellular Endocrinology, and his research team designed the studies and performed the novel, extensive and detailed animal transplant and drug treatment models using beta cells from donors. Final studies took place at City of Hope in 2023.

For the study, the natural product harmine, which is found in some plants, was combined with a widely used class of type 2 diabetes therapy called GLP1 receptor agonists. Researchers transplanted a small number of human beta cells into mice that had no immune system and that also served as a standard model of type 1 and type 2 diabetes; these mice were treated with the combination therapy and their diabetes was rapidly reversed. Strikingly, human beta cell numbers increased by 700% over three months with this drug combination.

This is the first time scientists have developed a drug treatment that is proven to increase adult human beta cell numbers in vivo. This research brings hope for the use of future regenerative therapies to potentially treat the hundreds of millions of people with diabetes, said Dr. Garcia-Ocaa, the papers corresponding author.

It has been remarkable to watch this story unfold over the past 15 years, said Dr. Stewart, who, along with Peng Wang, Ph.D., professor of Medicine (Endocrinology, Diabetes and Bone Disease) at Icahn Mount Sinai, conceived of and performed the initial high-throughput drug screen that led to the discovery of harmine described in Nature Medicine in 2015. The steady progression from the most basic human beta cell biology, through robotic drug screening and now moving to human studies, illustrates the essential role for physician-scientists in academia and pharma.

Growing new beta cells More than 10% of the worlds adult population has diabetes, a disease defined by high blood sugar levels. In both type 1 and type 2 diabetes, a reduction in both the quantity and quality of insulin-producing beta cells causes high blood sugar. Unfortunately, none of the many commonly used diabetes therapies are able to increase human beta cell numbers, and therefore cannot completely reverse diabetes.

Fortunately, most people with diabetes have some residual beta cells, which is what inspired the research team to search for ways to restore their numbers. The team had previously shown that several different inhibitors of an enzyme in beta cells called DYRK1A can induce the proliferation of adult human beta cells in a tissue culture dish for a few days. But prior to this study, no one had shown the ability to expand human beta cells numbers in vivo in human islet grafts used in an animal model over many months.

To accurately measure the mass of human beta cells in the islet grafts, the team turned to Sarah A. Stanley, M.B.B.Ch., Ph.D., associate professor of Medicine (Endocrinology, Diabetes and Bone Disease), and Neuroscience, at Icahn Mount Sinai. Using an advanced laser microscopy tool called iDISCO+ that effectively makes biological tissue transparent, Dr. Stanley saw that beta cell mass was dramatically increased through mechanisms that included enhanced proliferation, function and survival of the human beta cells. The technology allowed for accurate and rigorous quantitative assessment of engrafted human beta cells for the first time.

Translating results to the clinic The Mount Sinai team recently completed a phase 1 clinical trial of harmine in healthy volunteers to test its safety and tolerability. At the same time, Robert J. DeVita, Ph.D., professor of Pharmacological Sciences and director of the Marie-Jose and Henry R. Kravis Drug Discovery Institute at Mount Sinai, has developed next-generation DYRK1A inhibitors. Mount Sinai is conducting studies to test these in humans for potential toxicity risks and estimate dosing for clinical trials, and is planning to initiate first-in-human trials with independent research teams next year. Mount Sinai owns an extensive patent portfolio covering these technologies.

Researchers also want to address the fact that in patients with type 1 diabetes, the immune system will continue to kill new beta cells. At City of Hope, Dr. Garcia-Ocaa and colleague Alberto Pugliese, M.D., Samuel Rahbar Chair in Diabetes & Drug Discovery, chair of the Department of Diabetes Immunology and director of The Wanek Family Project for Type 1 Diabetes within the Arthur Riggs Diabetes & Metabolism Research Institute, plan to test inducers of beta cell regeneration together with immunomodulators that regulate the immune system. Their goal is for the combination to allow new beta cells to thrive and improve insulin levels.

Our studies pave the way for moving DYRK1A inhibitors into human clinical trials, and it's very exciting to be close to seeing this novel treatment used in patients, Dr. Garcia-Ocaa said. There is nothing like this available to patients right now.

The work outlined in the Science Translational Medicine paper was funded by grants from the National Institutes of Health (NIH), the National Institute of Diabetes and Digestive and Kidney Disease, and BreakthroughT1D (formerly JDRF), as well as from philanthropic donations to Mount Sinai, support from The Wanek Family Project for Type 1 Diabetesat City of Hope and additional generous philanthropic gifts.

Other critical members of the team include Mount Sinais Carolina Rosselot, Ph.D., Yansui Li, Ph.D., and Alexandra Alvarsson, Ph.D. Additional City of Hope authors on the paper are Geming Lu, M.D., assistant research professor, and Randy Kang, senior research associate, who are both members of Dr. Garcia-Ocaas lab.

Drs. Stewart and DeVita are named co-inventors on patent applications for DYRK1A inhibitors, such as harmine, for the treatment of diabetes. These patent applications are filed through the Icahn School of Medicine at Mount Sinai and are currently unlicensed.

# # #

About City of Hope City of Hope's mission is to make hope a reality for all touched by cancer and diabetes.Founded in 1913,City of Hopehas grown into one of the largest cancer research and treatment organizations in the U.S. and one of the leading research centers for diabetes and other life-threatening illnesses. City of Hope research has been the basis fornumerous breakthrough cancer medicines, as well as human synthetic insulin and monoclonal antibodies. With an independent, National Cancer Institute-designated comprehensive cancer center at its core, City of Hope brings a uniquely integrated model to patients spanning cancer care, research and development, academics and training, and innovation initiatives. City of Hopes growing national system includes its Los Angeles campus, a network of clinical care locations across Southern California, a new cancer center in Orange County, California, and cancer treatment centers and outpatient facilities in the Atlanta, Chicago and Phoenix areas. City of Hopes affiliated group of organizations includesTranslational Genomics Research InstituteandAccessHopeTM. For more information about City of Hope, follow us onFacebook,X,YouTube,InstagramandLinkedIn.

About the Mount Sinai Health System Mount Sinai Health System is one of the largest academic medical systems in the New York metro area, with 48,000 employees working across eight hospitals, more than 400 outpatient practices, more than 600 research and clinical labs, a school of nursing, and a leading school of medicine and graduate education. Mount Sinai advances health for all people, everywhere, by taking on the most complex health care challenges of our timediscovering and applying new scientific learning and knowledge; developing safer, more effective treatments; educating the next generation of medical leaders and innovators; and supporting local communities by delivering high-quality care to all who need it.

Through the integration of its hospitals, labs, and schools, Mount Sinai offers comprehensive health care solutions from birth through geriatrics, leveraging innovative approaches such as artificial intelligence and informatics while keeping patients medical and emotional needs at the center of all treatment. The Health System includes approximately 9,000 primary and specialty care physicians and 11 free-standing joint-venture centers throughout the five boroughs of New York City, Westchester, Long Island, and Florida. Hospitals within the System are consistently ranked by Newsweeks The Worlds Best Smart Hospitals, Best in State Hospitals, World Best Hospitals and Best Specialty Hospitals and by U.S. News & World Report's Best Hospitals and Best Childrens Hospitals. The Mount Sinai Hospital is on the U.S. News & World Report Best Hospitals Honor Roll for 2023-2024.

For more information, visithttps://www.mountsinai.org or find Mount Sinai onFacebook,Twitter andYouTube.

Science Translational Medicine

Excerpt from:
City of Hope and Mount Sinai scientists first - EurekAlert

Posted in Cell Medicine | Comments Off on City of Hope and Mount Sinai scientists first – EurekAlert

PHI aims for US listing – BioStock – Connecting Innovation and Capital

Posted: July 11, 2024 at 2:42 am

11 juli, 2024

Swedish Medtech Phase Holographic Imaging is making significant strides in the United States, establishing itself at the forefront of the rapidly growing regenerative medicine field. PHI is now taking an important step by applying for listing its shares in the US OTCQB market to enhance its presence and opportunities in the US. BioStock talked to CEO Patrik Eschricht to learn more about what this means for the company.

The core technology of the Phase Holographic Imaging (PHI) commercial HoloMonitor system is so-called Quantitative Phase Imaging (QPI), capable of assessing cell culture quality and health without influencing or damaging cells like traditional measurement methods. PHIs overarching goal with its technology is to contribute to making cell-based therapies affordable, accessible, and patient-safe.

Since 2022, the company has had a laser focus on regenerative medicine. This growing field involves the repair, replacement, or regeneration of dysfunctional cells, tissues, and organs to amplify our natural healing process in the places where it is needed most. Allowing for precise monitoring and verification of cell quality, the QPI technology is considered a valuable asset in this cutting-edge field. According to PHI, QPI measurements would fit seamlessly into an automated manufacturing workflow, setting it apart from other cell quality control methods that require removing precious cells from the process.

With the US spearheading the field of regenerative medicine, PHI has put a lot of emphasis on establishing itself in this major market. PHI got its first US customer already back in 2014 and has always had a strong academic foothold, collaborating with leading US research institutes, such as Northeastern University, Huntsman Cancer Institute, UCSF and the National Institute of Aging.

In 2019, as part of the ongoing expansion of activities in the US, PHI established its first fully owned subsidiary, PHI Inc., in Boston. Two years ago, the company joined the RegenMed Development Organization (ReMDO) located in Winston-Salem. PHI also joined an alliance for advancing cell-based biomanufacturing with other companies in ReMDO and the pioneering Wake Forest Institute for Regenerative Medicine (WFIRM). Together, they aim to automate the manufacturing process of cell-based therapies using recent technological developments in cell culturing, analytical instrumentation, and artificial intelligence.

In 2022, as a next step in its investment in regenerative medicine, PHI opened a new development office in the Innovation Quarter in Winston-Salem to become present at the epicentre of this rapidly growing field.

Now, the company has decided to push even further into the major US market, applying to list its shares on the US-based OTCQB Venture Market. This dual listing would not only offer PHIs current and prospective shareholders an additional trading platform but also enhance PHIs credibility among US investors and partners. BioStock has talked to PHI CEO Patrik Eschricht to learn more about what the dual listing means for the company and its plans moving forward.

We have thought about it before, but now that everyone is talking about regenerative medicine and AI, the time is right. We are very happy about the possibility of dual-listing on the OTCQB, which would be another significant step in PHIs growth journey as a global company. This dual listing would make it more accessible for international investors to invest in PHI, positively increasing liquidity, boosting shareholder value, and supporting our ongoing growth.

We have the worlds eye on Winston-Salem and the leading institute, WFIRM, for pioneering regenerative medicine and bringing scientific discoveries to the clinic and patients. The time is now, as we see the momentum within regenerative medicine and the, therefore, increasing relevance of PHIs unique technology. We have made a strategic decision to push further into the major US market and react to the growing interest in PHI from the US and regenerative medicine partners.

This dual listing would allow international investors to trade in PHI shares, expanding our investor community and enhancing the liquidity of our shares. I see this as a significant benefit to our current shareholders and PHIs ongoing growth.

Certainly to recap, were spearheading an important Smart Manufacturing project with our technology development alliance with WFIRM, ReMDO, analytics leader SAS, BioSpherix, and QIAGEN. This project focuses on creating new standards for cell therapy quality control. A cornerstone of this project is the Cell Report Card, which integrates QPI cell quality data with omics information.

This card tracks a cell sample throughout the lifetime of the cells and the patient, providing data crucial for ensuring quality and safety in cell therapies. We have finalized another round of experiments and continue to evaluate the HoloMonitor data together with our partners.

We are happy to see the substantial growth of the RegeneratOR test bed, particularly with our partner QIAGENs recent commitment to expanding its contributions. The test bed initiative is at the forefront of regenerative medicine, offering access to cutting-edge technologies and expert resources.

In my eyes, its a must-have to drive novel prototyping in the regenerative medicine field and the right place for PHI to present our unique technology for the development of cell-based therapies. At our latest meeting, it was obvious to everyone that not one company can solve all the problems; we need to work together. And at the RegeneratOR test bed, we have a very strong Starting Eleven, a starting lineup with over 60 companies.

We are still thrilled about the USD 160 million NSF grant, which underscores Winston-Salems role as a global hub for regenerative medicine! With the complexity and number of partners involved in this grant, we patiently await the presentation of details and how this grant will directly impact PHIso we, too, can share the news. So far, the increased focus on the region certainly has boosted our visibility and may create new business opportunities.

Our collaboration with WFIRM places us at the forefront of innovation in regenerative medicine. Being at the epicenter of such significant advancements reinforces our strategic positioning and commitment to establishing our QPI technology as the gold standard in cell quality control.

* anger obligatoriska flt

Follow this link:
PHI aims for US listing - BioStock - Connecting Innovation and Capital

Posted in Regenerative Medicine | Comments Off on PHI aims for US listing – BioStock – Connecting Innovation and Capital

Page 40«..1020..39404142..5060..»