The Government Should Stop Mandating the Use of "Race" in Medical and Scientific Studies – Reason

I was asked to contribute to a symposium on race, racism, and administrative law at Notice and Comment, the blog of the Yale Journal of Regulation. Given my current research on the American law of race, some of which I have blogged here, I had a lot to choose from; the most egregious forms of racial classification in the U.S. are largely the product of administrative decisions, rather than legislation. I ultimately decided to write about how the FDA and NIH require the use of ridiculously unscientific "racial" categories, adopted by the OMB for entirely different purposes in 1977, in biomedical research, and why this use of "race" should be abolished. Here's a taste, but you can read the whole thing at this link:

As of this writing, the federal government is considering using race and ethnicity to allocate access to a new Coronavirus vaccine to combat Covid-19 when one becomes available. More specifically, the government is considering giving preference to African Americans and Latinos because they have been disproportionately affected by the pandemic.

There are obvious dangers to allocating medical resources by race especially in a politically sensitive an area such as vaccines, where the public is already all-too-prone to accept various conspiracy theories and quackery that leads them to oppose vaccination. Instead of expanding the use of race in this way, science and medicine should be moving away from considering race and ethnicity at all.

Unfortunately, the FDA and NIH have mandated the use of race and ethnicity since the late 1990s. As a result of this mandate, the use of race has become so common in the scientific and medical communities that most people in the field fail to consider whether there is any justification for doing so. As one scientist reports, "we don't tend to think a lot about that [race] variable, what it means, how it's defined, how it's being used. We just sort of use it blindly."

This is very unfortunate, because, in addition to other problems discussed below, the FDA and NIH mandated that the "race variable' be based on the arbitrary (but now standard in American life) racial and ethnic classifications established by the Office of Management and Budget in 1977 for civil rights enforcement purposes. At the time, the OMB warned that the "classifications should not be interpreted as being scientific or anthropological in nature." This did not stop the FDA and NIH from institutionalizing them into medical and scientific research.

Any discussion of race in science and medicine must start with the recognition that variations in DNA that may have scientific or medical implications are not specific to race, as such, but to geographical distance between different populations. Additionally, there is no known example of polymorphism that is found exclusively in any particular "racial" group.

Even if at one time race may have been useful as a crude proxy for genetic heterogeneity, as DNA testing has become more available and much less expensive, race is a poor substitute for looking at actual discernible genetic differences between people. "Pooling people in race silos," an editorial in Nature Biotechnology declared, "is akin to zoologists grouping racoons, tigers, and okapis on the basis that they are all stripey."

The OMB category of Asian, meanwhile, is absurdly non-specific and unscientific. It includes people with origins everywhere from the Philippines to the Indian subcontinent. There are vast differences among the various ethnic groups that comprise the two billion or so people who live within the Indian subcontinent, much less between South Asians and East Asians.

Hispanic/Latino is an even more problematic category. Latinos' origins can be any combination of African, Asian, European, and Indigenous. Nor are they culturally homogenous. There is no reason to believe that data about Dominican residents of New York City is applicable to indigenous Mexican farm workers in California.

[S]upport for the idea that we should allow the government to use research based on arbitrary, scientifically ridiculous OMB racial categories to allocate medical resources to people based on those categories seems both fantastical and an unjustified triumph of unscientific racialist thinking. Unfortunately, this is what NIH's and FDA's imposition of the OMB categories into scientific research has wrought.

Original post:

The Government Should Stop Mandating the Use of "Race" in Medical and Scientific Studies - Reason

It’s time to to start a public conversation about genomics – World Economic Forum

Genomics. Genomic medicine. Personalised medicine. These are very much the buzzwords of our time when it comes to healthcare. But on hearing them for the first time, what do these terms actually mean to people? One research participant who had never heard of the term genomics before, broke the word down phonetically and suggested: "Is genomics something to do with genes and economics?"

It's not an unreasonable question, and it's helpful to be reminded periodically that as a biotechnology community we need to bring everyone up to speed with what we are actually talking about.

Genomics is emerging as one of the main sources of data across many disciplines of medicine and healthcare, so there has never been a more important time to spark public interest in what it can do for people and how they might benefit (and contribute). But how do we start these conversations when often the science, as well as the surrounding language, can seem alien and impenetrable?

It is likely that some of us have already had form of genomic test already, and even if we havent personally, someone we are biologically related to may have done. This may be as part of a routine or in-depth health check, for research purposes or even as part of an online ancestry test.

The first time we come in contact with genomic technology may be an overwhelming or even confusing experience, revealing much more information than we may have anticipated. For others, the situation may be less intense or even recreational, meaning less care will be given about the science that exists behind those tests or consideration of the implications.

Whatever the experience for the individual, it is crucial we reach people with the right information to both dispel the myths around genomics, and make it meaningful. For many, results from genetic tests will end up being a wider conversation, where individuals will be expected to consider what the test means, and its future implications for other relatives.

This means work is needed to find a language that is both memorable and resonates with everyone. But how is this achieved? How can we make genomics and genomic testing seem much less daunting to talk about? What sort of framings narratives, metaphors, mantras and memes can we use to socialise an otherwise dense topic that even healthcare professionals find difficult to navigate?

The only way to achieve a model that works for both professionals and members of the public is to open up dialogue to all groups of people to create a new shared understanding, and an accompanying language that provides clear guidelines around the tools and narratives to be used to achieve best practice. There needs to be a balance of investment where increasing genomic literacy through education (valuable though it is) is only one part of the package. Often what is missed is investment in how to engage and reach currently disconnected members of the public who have never given any thought to the subject. Why is this important? Because as genomic medicine goes mainstream, they may be a patient in waiting or related to someone who is. This means that the relevance of genomics has moved beyond the individual to the family, and then on to broader society.

Healthcare professionals need a clear understanding about how to talk to patients. Recognising that patients do not need to understand the technical concepts to make sense of the testing process, or to make informed choices for themselves and their family, is a first step in creating a mutual respect between these stakeholders. Patients need to feel more confident and familiar with what genomics means for them, and more at ease about discussing it, just as we would for common illnesses.

Its time for us collectively to turn conversations about the impact of genomics into something sociable, easy to navigate and familiar. Just as we have started to embrace discussions on the impact of vaccines and climate change, so too is this necessary for genomics as without it, the impact of genetics will be misinterpreted. Cross-disciplinary expertise is needed for this drawing together input from both the arts and sciences but also from storytellers from the advertising and film industries, and utilising evidence-based engagement and narrative techniques. This evidence must not be one-sided, however, and should incorporate different experiential and social understandings held by different members of society.

The application of precision medicine to save and improve lives relies on good-quality, easily-accessible data on everything from our DNA to lifestyle and environmental factors. The opposite to a one-size-fits-all healthcare system, it has vast, untapped potential to transform the treatment and prediction of rare diseasesand disease in general.

But there is no global governance framework for such data and no common data portal. This is a problem that contributes to the premature deaths of hundreds of millions of rare-disease patients worldwide.

The World Economic Forums Breaking Barriers to Health Data Governance initiative is focused on creating, testing and growing a framework to support effective and responsible access across borders to sensitive health data for the treatment and diagnosis of rare diseases.

The data will be shared via a federated data system: a decentralized approach that allows different institutions to access each others data without that data ever leaving the organization it originated from. This is done via an application programming interface and strikes a balance between simply pooling data (posing security concerns) and limiting access completely.

The project is a collaboration between entities in the UK (Genomics England), Australia (Australian Genomics Health Alliance), Canada (Genomics4RD), and the US (Intermountain Healthcare).

Constructing a global PR campaign takes commitment to messaging, resources and creativity; it also needs collaboration across academia, industry and medicine, with no discipline taking dominance. It also needs to tap into the seldom-heard public voices from people who have had no interaction with this technology yet. It needs to seek out the views of indigenous peoples and representative members of the public, but also those who are fearful and afraid of the technology. Without this collaboration the conversations about genomics will remain disconnected, patchy and without a consistent beneficiary.

This is about all of us not for profit, but for humankind.

License and Republishing

World Economic Forum articles may be republished in accordance with the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International Public License, and in accordance with our Terms of Use.

Written by

Anna Middleton, Professor and Head, Society and Ethics Research Group, Wellcome Genome Campus (Sanger Institute), University of Cambridge

Mavis Machirori, Research Associate, Policy, Ethics and Life Sciences, Newcastle University

Jenniffer Mabuka-Maroa, Consultant, The African Academy of Sciences

Tiffany Boughtwood, Manager, Australian Genomics Health Alliance

The views expressed in this article are those of the author alone and not the World Economic Forum.

Read the original here:

It's time to to start a public conversation about genomics - World Economic Forum

Merus announces FDA Orphan Drug Designation of Zenocutuzumab for the Treatment of Pancreatic Cancer – BioSpace

UTRECHT, The Netherlands and CAMBRIDGE, Mass., July 27, 2020 (GLOBE NEWSWIRE) -- Merus N.V. (Nasdaq: MRUS), a clinical-stage oncology company developing innovative, full-length multispecific antibodies (Biclonics and Triclonics) for cancer, today announced that the U.S. Food and Drug Administration (FDA) has granted Orphan Drug Designation to Zenocutuzumab (Zeno) for the treatment of patients with pancreatic cancer.

Zeno is a first in class bispecific antibody that potently binds to the HER2 and HER3 receptors, to potently block the interaction of HER3 with its ligand, neuregulin 1 (NRG1). Zeno has demonstrated promising early clinical responses in patients with previously treated pancreatic cancer harboring NRG1 gene fusions, as presented at the AACR/NCI/EORTC International Conference on Molecular Targets and Cancer Therapeutics in October 2019. The NRG1 gene fusion is a rare, powerful driver of cancer cell growth found in pancreatic, lung and other types of solid tumors. Zeno is now being evaluated in a global phase 1/2 clinical trial called the eNRGy trial.

Receiving Orphan Drug Designation for Zeno is another important milestone for our lead program, and it validates the significant unmet need in patients with pancreatic cancer, said Bill Lundberg, M.D., President, Chief Executive Officer and Principal Financial Officer of Merus. We are pleased with the progress we are making in our ongoing global clinical trial, and believe that Zeno has the potential to play a significant role in shifting the treatment paradigm for NRG1 fusion cancers from conventional chemotherapy to a personalized medicine approach.

The FDA grants Orphan Drug Designation to drugs that are intended to treat rare diseases that affect fewer than 200,000 people in the U.S. Orphan Drug Designation may provide Merus certain benefits, such as grant funding towards clinical trial costs, tax advantages and eligibility for seven-year market exclusivity.

Pancreatic cancer is estimated to occur in approximately 57,000 patients annually in the United States, according to the NCI SEER database. Pancreatic ductal adenocarcinoma (PDAC), the most common subtype of pancreatic cancer, is one of the most aggressive solid tumor cancers and the fourth leading cause of cancer related deaths.

About the eNRGy Clinical Trial Merus is currently enrolling patients on the Phase 1/2 eNRGy trial to assess the safety and anti-tumor activity of Zeno monotherapy in NRG1+ cancers. The eNRGy trial consists of three cohorts: NRG1+ pancreatic cancer; NRG1+ non-small cell lung cancer; and NRG1+ other solid tumors. Further details, including current trial sites, can be found at http://www.ClinicalTrials.gov and Merus trial website at http://www.nrg1.com or by calling 1-833-NRG-1234.

About NRG1 FusionsThe NRG1 gene encodes for neuregulin 1 (also known as heregulin), the ligand for HER3.Fusions between NRG1 and partner genes are rare genetic events occurring in patients with certain lung, pancreatic and other solid tumors, associated with activation of HER2/HER3 signaling and growth of cancer cells. NRG1 fusions are estimated to occur at a rate of approximately 0.5% - 1.5% in PDAC, based on the limited available published data.

About ZenoZeno is an antibody-dependent cell-mediated cytotoxicity (ADCC)-enhanced Biclonics that utilizes the Merus Dock & Block mechanism to inhibit the neuregulin/HER3 tumor-signaling pathway in solid tumors. Through its unique mechanism of binding to HER2 and potently blocking the interaction of HER3 with its ligand NRG1 or NRG1-fusion proteins, Zeno has the potential to be particularly effective against NRG1+ cancers. In preclinical studies, Zeno also potently inhibits HER2/HER3 heterodimer formation and tumor growth in models harboring NRG1 fusions.

Learn more aboutZeno Dock& Block athttps://merus.nl/technology/.

About MerusMerus is a clinical-stage oncology company developing innovative full-length human bispecific and trispecific antibody therapeutics, referred to as Multiclonics. Multiclonics are manufactured using industry standard processes and have been observed in preclinical and clinical studies to have several of the same features of conventional human monoclonal antibodies, such as long half-life and low immunogenicity. For additional information, please visit Merus website,www.merus.nlandhttps://twitter.com/MerusNV.

Forward Looking StatementsThis press release contains forward-looking statements within the meaning of the Private Securities Litigation Reform Act of 1995. All statements contained in this press release that do not relate to matters of historical fact should be considered forward-looking statements, including without limitation, the potential of Zeno to play a significant role in shifting the treatment paradigm for NRG1 fusion cancers from conventional chemotherapy to a personalized medicine approach; the progress of the eNRGy trial; and the potential benefits of the orphan drug designation of Zeno , such as grant funding towards clinical trial costs, tax advantages and eligibility for seven-year market exclusivity. These forward-looking statements are based on managements current expectations. These statements are neither promises nor guarantees, but involve known and unknown risks, uncertainties and other important factors that may cause our actual results, performance or achievements to be materially different from any future results, performance or achievements expressed or implied by the forward-looking statements, including, but not limited to, the following: our need for additional funding, which may not be available and which may require us to restrict our operations or require us to relinquish rights to our technologies or Biclonics Triclonics and multispecific antibody candidates; potential delays in regulatory approval, which would impact our ability to commercialize our product candidates and affect our ability to generate revenue; the lengthy and expensive process of clinical drug development, which has an uncertain outcome; the unpredictable nature of our early stage development efforts for marketable drugs; potential delays in enrollment of patients, which could affect the receipt of necessary regulatory approvals; our reliance on third parties to conduct our clinical trials and the potential for those third parties to not perform satisfactorily; impacts of the COVID-19 pandemic; we may not identify suitable Biclonics or bispecific antibody candidates under our collaborations or our collaborators may fail to perform adequately under our collaborations; our reliance on third parties to manufacture our product candidates, which may delay, prevent or impair our development and commercialization efforts; protection of our proprietary technology; our patents may be found invalid, unenforceable, circumvented by competitors and our patent applications may be found not to comply with the rules and regulations of patentability; we may fail to prevail in potential lawsuits for infringement of third-party intellectual property; and our registered or unregistered trademarks or trade names may be challenged, infringed, circumvented or declared generic or determined to be infringing on other marks.

These and other important factors discussed under the caption Risk Factors in our Quarterly Report on Form 10-Q for the quarterly period ended March 31, 2020 filed with the Securities and Exchange Commission, or SEC, on May 11, 2020, and our other reports filed with the SEC, could cause actual results to differ materially from those indicated by the forward-looking statements made in this press release. Any such forward-looking statements represent managements estimates as of the date of this press release. While we may elect to update such forward-looking statements at some point in the future, we disclaim any obligation to do so, even if subsequent events cause our views to change, except as required under applicable law. These forward-looking statements should not be relied upon as representing our views as of any date subsequent to the date of this press release.

Follow this link:

Merus announces FDA Orphan Drug Designation of Zenocutuzumab for the Treatment of Pancreatic Cancer - BioSpace

DNA changes that cause neuropsychiatric disorders should be included in genomic screening programs, researchers say – BioSpace

DANVILLE, Pa., July 23, 2020 /PRNewswire/ --Geisinger researchers have concluded there is sufficient evidence to consider including DNA changes that cause neurodevelopmental and psychiatric disorders in genomic screening programs.

Geisinger is a leader in population-based genomic screening, and its MyCode Community Health Initiative is the only such research program that routinely returns clinically relevant results to patient-participants. The Geisinger team discovered that close to 1 percent of the more than 250,000 individuals enrolled in MyCode have a DNA change that is known to cause learning disorders, autism spectrum disorder, epilepsy, or other psychiatric illnesses. By analyzing electronic health record data, the research team determined that up to 70 percent of these individuals had a related clinical symptom documented, but most were unaware of their underlying genetic diagnosis.

The team published their findings in the Journal of the American Medical Association, Psychiatry on July 22.

"Our results show that DNA changes that cause certain brain conditions are at least as common as those that cause some cancers and cardiac diseases that are already being screened for in similar population-based DNA screening programs," said Christa Lese Martin, Ph.D., associate chief scientific officer for Geisinger and professor and director of the Autism & Developmental Medicine Institute. "When we talked with participants about their medical history and found that such a significant proportion had symptoms related to their genetic diagnosis, they shared that the genetic results 'medicalized' what they had been dealing with their whole lives."

When presented with their screening results, a subset of more than 140 patients responded positively and found the information to be valuable. Participants frequently noted that the DNA results helped them understand their own medical and personal history related to the conditions being studied, and many intended to share their results with family members since these DNA changes can be inherited.

"These DNA results are likely to have had a large impact on health and wellbeing throughout life for these individuals," said Karen E. Wain, MS, assistant professor for Geisinger's Autism & Developmental Medicine Institute. "It is important to know how people feel about these results, for themselves and their family, so we can ensure they have access to their genetic information with appropriate support."

Advances in genetic testing have made it possible to identify a genetic cause of neurodevelopmental and psychiatric disorders in more than 40 percent of individuals tested. However, most testing is ordered for children with developmental concerns and is rarely offered to adult patients with intellectual disabilities or psychiatric conditions. The Geisinger team noted that only about 6 percent of individuals in their study had received a genetic diagnosis through clinical testing. This indicates that many people who could benefit from genomic information have not had access to this information, and that their health care providers have not been informed of the additional health risks that the genomic result confers.

"There is an important care gap and knowledge gap when it comes to genetic testing in adults with neuropsychiatric conditions," said David Ledbetter, Ph.D., executive vice president and chief scientific officer for Geisinger. "We hope the positive clinical and personal utility seen in our MyCode population will help to encourage broader use of genetic testing in adults with these conditions."

About GeisingerGeisinger is committed to making better health easier for the more than 1.5 million consumers it serves. Founded more than 100 years ago by Abigail Geisinger, the system now includes 13 hospital campuses, a 600,000-member health plan, two research centers and the Geisinger Commonwealth School of Medicine. With 32,000 employees and 1,800 employed physicians, Geisinger boosts its hometown economies in Pennsylvania and New Jersey by billions of dollars annually. Learn more atgeisinger.org or connect with us on Facebook, Instagram, LinkedIn and Twitter.

CONTACT: Ashley Andyshak Hayes717-972-4043arandyshakhayes@geisinger.edu

View original content:http://www.prnewswire.com/news-releases/dna-changes-that-cause-neuropsychiatric-disorders-should-be-included-in-genomic-screening-programs-researchers-say-301099119.html

SOURCE Geisinger

More:

DNA changes that cause neuropsychiatric disorders should be included in genomic screening programs, researchers say - BioSpace

2 Stocks to Hold for the Next 20 Years – Motley Fool

As 2020 has demonstrated, the stock market can move in any direction in the short term. But as historical stock market data suggest, the stock market wields rock-solid wealth-building potential in the long run. That's why individual investors are best served by adopting a long-term mindset with a buy-and-hold strategy.

There are a number of ways to go about it. Most portfolios should own a collection of blue chip stocks, even if that simply relies on indexing, although buy-and-hold investing doesn't have to be boring. After all, investors that held onto today's blue-chip businesses before they were trendy have enjoyed awesome returns thanks to the power of compound interest.

With that in mind, investors might want to take a closer look at Dicerna Pharmaceuticals (NASDAQ:DRNA) and Fate Therapeutics (NASDAQ:FATE). The two development-stage biopharmas have much to prove, but these pharma stocks could appreciate considerably in the coming decades.

Image source: Getty Images.

First-generation cell therapies were based on chimeric antigen receptor (CAR) T cells, or CAR-T. While the approach has plenty of room for improvement, CAR-T cells have several notable drawbacks. For instance, the cell type isn't ideal for treating solid tumor cancers. Additionally, harvesting immune cells from each individual patient is a cost- and labor-intensive process that delays treatment. This all suggests it's worth exploring other immune cells as the starting point for next-generation cell therapies and new production methods, at the very least to augment the capabilities of CAR-T.

Fate Therapeutics is all-in on that line of thinking. Most of the company's 13 unique pipeline programs are based on natural killer (NK) cells, which have inherent advantages compared to other types of cell therapies. NK cells can target solid tumor cancer cells, rally the rest of the immune system to reduce tumor burden, and be dosed multiple times. The latter feature creates many new treatment opportunities, such as driving longer durations of response or being used in combination with other cell therapies.

The development-stage business has also taken the "next-generation" label seriously. Rather than harvest immune cells from patients -- a long, complicated, expensive, and risky procedure -- the company is pursuing an off-the-shelf strategy. In other words, most of the pipeline candidates are grown from master cell lines, which allows each cell therapy to be genetically engineered with reproducible edits and to be produced in batches. Fate Therapeutics estimates its approach would save weeks during crucial treatment windows and drop the cost of treatment from $425,000 per dose to just $2,500 per dose. Better yet, the approach is cell-type agnostic, meaning it can be applied to NK cells and CAR-T cells.

Although the early stage pipeline lacks concrete data for investors to digest, there are a handful of significant partnerships that boost the company's credentials. Fate Therapeutics has a collaboration with Johnson & Johnson subsidiary Janssen that could be worth up to $3 billion in milestone payments, a collaboration with ONO Pharmaceutical that could be worth over $1.3 billion in milestone payments, and a partnership with Inscripta providing access to next-generation gene-editing tools.

Fate Therapeutics ended March with $204 million in cash, which is sufficient to get the business through several data readouts from early clinical trials. If the results suggest the cell therapy approach has merit, then the biopharma stock should earn a higher valuation. Given the ambitious volume of assets -- 13 unique pipeline programs is very large for a development-stage company -- no single failure should have a disastrous effect on the stock price. Investors with a long-term mindset should give this company a closer look.

Image source: Getty Images.

Gene editing and gene therapies gobble up most of the attention when it comes to genetic medicines, but investors shouldn't forget about RNA interference (RNAi). The gene-silencing technique encountered some stumbles in the past two decades, but a few simple tweaks to how the therapeutic payload is delivered into cells appears to have resurrected the technology's intriguing potential.

Dicerna Pharmaceuticals is one of the leading investment opportunities in the space. On the one hand, the company hasn't commercialized a single pipeline asset and is relatively far behind RNAi peers Alnylam Pharmaceuticals and Arrowhead Pharmaceuticals. On the other hand, the company ended March with half its market cap in cash and partnerships with six of the world's leading pharmaceutical companies.

That includes an unusual collaboration with Alnylam Pharmaceuticals. The duo reached an agreement to co-develop competing drug candidates for alpha-1 liver disease, in which Dicerna Pharmaceuticals has the right to develop Alnylam's ALN-AAT02 and its own DCR-A1AT. The RNAi competitor-collaborators also agreed to share intellectual property for their pulmonary hypertension (PH) programs. Hefty royalties -- in both directions -- are the bounty for success.

That's not the only intriguing partnership in the pipeline. Dicerna Pharmaceuticals and Roche are developing an experimental treatment aimed at chronic hepatitis B (CHB) infections. The asset is likely being developed as a functional cure for the disease, which would follow in the footsteps of an RNAi combination therapy from peer Arrowhead Pharmaceuticals. A safe and effective functional cure for CHB could generate tens of billions of dollars in lifetime sales.

Despite the early stage nature of the pipeline, investors cannot overlook the potential of the RNAi medicines being developed by Dicerna Pharmaceuticals. The gene-silencing approach could carve out and maintain dominant market positions in various indications. Market shares might change when curative gene editing tools and gene therapies arrive, although that could take much longer than many investors expect due to several technical obstacles facing the hyped-up approaches. Investors looking for a contrarian pick in genetic medicine might want to give serious consideration to this biopharma stock.

See the rest here:

2 Stocks to Hold for the Next 20 Years - Motley Fool

Explained: How a Black woman saved lives without her consent and without due acknowledgement – The Indian Express

Written by Pooja Pillai | New Delhi | Updated: July 27, 2020 12:22:45 am Henrietta Lacks historical marker in Clover, Virginia. (Photo: Wikimedia Commons)

In the coming week (on August 1) is the birth centenary of Henrietta Lacks, an African American woman who made one of the most significant contributions to modern medical science without her knowledge or consent.

The story of Lacks and the HeLa cell line that was harvested from her and which still forms the basis of a lot of medical research isimportant for an understanding of the ethical issues in medical research on human subjects. This is especially so right now, given the urgency to develop an effective COVID-19 vaccine, which requires that it be tested on human cells.

Who was Henrietta Lacks?

Henrietta Lacks was an African American woman, who, according toThe Immortal Life of Henrietta Lacks(2010, Crown) by Rebecca Skloot, grew up on a tobacco farm in rural Virginia. She was married to David Lacks and had five children.

Also Read | In Covid year, why unsung heroine of DNA Rosalind Franklin needs to be remembered

On January 29, 1951, she visited the John Hopkins Hospital in Baltimore, Maryland, for the diagnosis and treatment of a lump in her abdomen. It turned out to be an aggressive form of cervical cancer. Lacks died at the age of 31 on October 4, 1951.

What is HeLa and whats so special about it?

When Lacks was at John Hopkins, her tumour was biopsied and tissues from this were used for research by Dr George Otto Gey, the head of the Tissue Culture Laboratory at the hospital. The cells were found to be growing at a remarkable rate, doubling in count in 24 hours. Their astonishing growth rate made them ideal for mass replication for use in medical research.

Prior to this, researchers had attempted to immortalise human cells in vitro, but the cells always eventually died. The HeLa cells named after the donor were the first ones to be successfully immortalised.

How have HeLa cells advanced medical science?

The HeLa cell line is one of the most important cell lines in the history of medical science and has been the foundation for some of the most significant advances in this field.

HeLa cells were the first human cells to be successfully cloned and were used by Jonas Salk to test the polio vaccine. Significantly, they helped in identifying the human papilloma virus (HPV) as being the main cause of many forms of cervical cancer including the one that killed Lacks and were instrumental in the development of the HPV vaccine, which won its creator, Harald zur Hausen, the Nobel Prize for Medicine in 2008.

Theyve been used widely in cancer research and were used to establish that human cells contain 23 pairs of chromosomes, not 24, as previously thought.

When was Lacks recognised as the donor of the HeLa cells?

Lacks was an unwitting donor; neither she, nor her family were aware that her cells had been extracted and were to be used for medical research. Lacks was a poor, uneducated Black woman and her consent was not considered necessary by the medical establishment at the time.

Also Read | Meet Dr Sarah Gilbert, one of the scientists leading the race to find a coronavirus vaccine

While thousands of studies and developments worth many billions of dollars happened due to the HeLa cells, Lacks herself was only acknowledged as their source in the 1970s when researchers sought blood samples from her family. Moreover, her descendants had no control over the cell line until 2013, when the National Institutes of Health arrived at an agreement with them, granting them a degree of control over how Lacks genetic material was to be used.

Race and non-ethical medical research

In 1947, during the Nuremberg Trials, the Allied forces developed what came to be known as the Nuremberg Code, a set of 10 ethical principles for human experimentation. The code was created in response to the German experiments on human subjects during World War II and the first principle it enshrined was that voluntary consent was essential in human experimentation.

By the time Lacks cells were harvested and used without her consent, the code had been in existence for four years. Unfortunately, the violation of Lacks consent was only the latest chapter in a long history of medical research which has scorned ethics as far as non-white bodies are concerned.

Take the case of J Marion Sims, the 19th century physician who is often called the father of modern gynecology. He pioneered the surgical treatment of the vesicovaginal fistula, a common complication of childbirth in which a tear develops between the bladder and vaginal wall, causing pain, infection and urine leakage. Sims performed his surgical experiments on Alabama slaves, without their consent and without the benefit of anaesthesia.

Or consider the infamous Tuskegee Syphilis Study, conducted by the US Public Health Service, from 1932 to 72, which examined how untreated syphilis progressed through African American men and how different it was from the way it affected white men.

Alabamas Tuskegee Institute (now Tuskegee University) was recruited for the study and the subjects 399 were infected patients and 201 uninfected control patients were all poor sharecroppers. While treatment with arsenic, bismuth and mercury was initially part of the study, the subjects were later given no treatment at all. Even after penicillin began to be widely available for use in treatment of syphilis in the 1940s, it was withheld from the subjects of the Tuskegee study. More than 100 are believed to have died; the study finally ended only after public exposure in the Washington Star.

Express Explainedis now onTelegram. Clickhere to join our channel (@ieexplained)and stay updated with the latest

Unethical, non-consensual experiments on human subjects took place elsewhere too; in 2013, food historian Ian Mosby revealed highly unethical nutritional experiments conducted by the Canadian government on Aboriginal children in six residential schools between 1942 and 52.

As part of the study, malnourished children were denied adequate nutrition; parents were neither informed, nor was their consent sought.

In 2004, a senate inquiry into the experiences of Australian Aboriginal children forced into state care similarly revealed their use in medical experiments and trials, from the 1920s till as late as 1970.

The Indian Express is now on Telegram. Click here to join our channel (@indianexpress) and stay updated with the latest headlines

For all the latest Explained News, download Indian Express App.

IE Online Media Services Pvt Ltd

See the original post:

Explained: How a Black woman saved lives without her consent and without due acknowledgement - The Indian Express

COVID-19 tests: There’s an insurmountable backlog of virus tests. A rapid test could help. – NBC News

As the U.S. deals with massive delays in COVID-19 testing, doctors and scientists say another type of diagnostic test could alleviate the stress on labs. Rapid, or point-of-care, tests deliver results in just minutes, while lab-based tools can take days.

"Every day they wait is another day they need to quarantine, or if they're not, it's another day they could be infecting other people," Dr. Keith Jerome, who directs the molecular virology lab at the University of Washington medical school, said in an interview. "If you're getting results within 20 minutes, you can start taking the appropriate actions right away."

The National Institutes of Health announced Wednesday what it called an "unprecedented effort" to ramp up testing technology. Funded by $1.5 billion in federal stimulus money, the program will focus on creating rapid tests and distributing them more widely.

It's also being called for by lawmakers and top federal health officials. President Donald Trump promised more rapid testing during his briefing Tuesday. And Dr. Brett Giroir, who is overseeing the nation's COVID-19 testing, said this month that he expected 5 million additional "point-of-care" tests in July, with a goal of 20 million or more by September.

The tests produce such quick results because samples aren't sent off to labs. Instead, they're inserted directly into a machine housed at a doctor's office or a hospital. The machine does the entire analysis, so instead of hours, it takes just minutes to get results similar to rapid flu or strep tests used by most doctors.

Six point-of-care tests are authorized by the Food and Drug Administration, including two antigen tests, which look for certain proteins in the virus rather than genetic material.

As promising as the rapid tests seem to be, a significant problem prevents more doctors and clinicians from using them. Most aren't as accurate as lab-based tests, and, in some cases, they can have shockingly high rates of false negatives.

Full coverage of the coronavirus outbreak

Let our news meet your inbox. The news and stories that matters, delivered weekday mornings.

Joseph Petrosino, director of molecular virology and microbiology at Baylor College of Medicine, compares it to eating at a fast-food joint.

"If you go to a gourmet restaurant, you don't expect your meal to be ready in five minutes," he said. "But if you're on the go and you eat fast food, the quality of food is usually a sacrifice, compared to the gourmet restaurant. That's the same thing in the testing world."

One of the more popular rapid tests, Abbott Labs' ID NOW point-of-care test, which promises results in as little as five minutes and was once touted by the White House, came under fire in recent weeks after a small study found that it returned false negatives for nearly 50 percent of certain samples compared to a rival test. While other studies found more accurate results, it was enough for the FDA to issue an alert in May. The agency has received 147 adverse event reports about the test.

"The reality is that trying to do this really fast the combination of fast and sensitive turns out to be really a challenge," said Dr. Christopher Polage, director of the clinical microbiology laboratory at Duke University Health System.

Polage said COVID-19 testing is a lengthy process. In the lab, scientists use special reagents that amplify or copy a sample's genetic material to test for the virus. The process takes several hours. When you try to short-cut it for a rapid test, you can end up trading off the test's sensitivity.

"No patient is ever going to wait at a clinic for eight hours," Polage said. "So it's really difficult and, in some cases, impossible to get an equivalent result in a fraction of the time."

Jerome said, "You have to really keep in mind that there is a trade-off that you've made for that speed, and the trade-off is they're not as sensitive, which means they're going to miss some people who actually have COVID and tell them COVID isn't there."

A point-of-care test made by Cepheid Inc. of Sunnyvale, California, which gives results in about an hour, has been shown to be nearly as accurate as lab-based tests. But scientists say that the machine is expensive and that, as with some lab-based tests, some of the reagents it uses are in short supply.

"If you can wait an hour, you can get really good results," Jerome said. "The issue with those has been just shortages of reagents, and the machine itself is just not available enough that everybody can have one." He said UW Medicine, the health care system affiliated with the University of Washington, can use it only in the emergency room, where it needs to quickly test trauma patients so doctors and nurses know what kind of protective gear they need to wear.

But some doctors say that instead of focusing on rapid tests, which are notoriously difficult to perfect both rapid strep and flu tests also have issues with accuracy the U.S. should focus on fixing capacity issues with lab-based tests, which are being slowed in part because of supply shortages.

"We have the equipment, but we can't get the reagents," Jerome said. "We did a little over 7,000 tests yesterday in my laboratory. But we could have done 7,000 more if we had full allotments of reagents."

Download the NBC News app for full coverage and alerts about the coronavirus outbreak

The delays in testing have a real impact for people like Frank Borsa, 55, who anxiously waited for days to find out whether he was infected after he returned to Brooklyn, New York, from Miami.

"It's really frustrating," Borsa said while he was still waiting for the results. "I've called repeatedly. What you get is 'it's taking a little bit longer.' It's very difficult, because you don't know how to go forward."

He finally got his results 12 days after he took the test at a New York City urgent care center. He was negative. But he said he now understands what the hubbub around testing is all about.

"Even though there might be hundreds of thousands of tests performed per day, if people are not getting their results, this is never going to end," he said. "The communication is there's tons of tests. But if there's no results, what good is it?"

Originally posted here:

COVID-19 tests: There's an insurmountable backlog of virus tests. A rapid test could help. - NBC News

Why Black and Hispanic residents are more likely to become infected by the coronavirus – Poynter

Black and Hispanic residents are more likely than white residents to become infected by the coronavirus and Black residents are more likely to die from it. Dr. Sherita Hill Golden, vice president and chief diversity officer for Johns Hopkins Medicine, talked with me this week about why that is the case.

She discussed the impact of systemic racism, dispelled some myths and highlighted ways health care institutions and government can respond to make a difference.

Here are the highlights of our conversation, which has been edited and condensed for clarity.

Tim Nickens: Statistics show Black and Hispanic residents are more likely to become infected by the COVID-19 virus and more likely to die from it. Why is that?

Dr. Sherita Hill Golden, vice president and chief diversity officer for Johns Hopkins Medicine (Courtesy: Johns Hopkins Medicine)

Dr. Sherita Hill Golden: There are several contributing factors. I think of them in three buckets, and two of them are historical. One is that there are historical practices that were embedded in our medical and health care environments. During slavery, African American slaves were often experimented upon without their consent and without anesthesia. Even in modern-day medicine, there are some erroneous beliefs that somehow Blacks are more tolerant of pain and need less pain medication. Thats an example of a bias that still exists in the health care system that results in inadequate pain control during hospitalization.

There were also situations like the Guatemala syphilis experiment (in the 1940s) and the Tuskegee syphilis experiment (from the 1930s to 1972) where Hispanics and African Americans were withheld treatment so scientists could learn the natural history of syphilis. The Tuskegee experiment wasnt uncovered until 1972. Thats fairly recent in our countrys history. All of those things led to a distrust among minority populations of our medical system.

At the turn of the last century, there were also racial and ethnic groups that were considered biologically inferior to others African Americans, Latinos, and recent immigrants to the U.S. We know today that is absolutely not true. There is no scientific foundation that there are any groups that are genetically inferior to others.

All of those things have contributed to minority patients experiencing bias in the health care system resulting in less likelihood of seeking care and poor experiences in the health care setting. So we now have African American and Latino patients who may be getting sick but not coming into the health care system because they may have had poor experiences before, or coming to the health care system and not being believed when they are presenting symptoms of COVID-19.

Second, there is a social context, policies that have been in place in our country that started after the Civil War era that have contributed to structural and institutional racism in things like housing, jobs and education. African Americans were coming from the South to settle in cities in the north, and a lot of those neighborhoods would become redlined and African Americans were often subjected to predatory loans. City governments would stop investing in public works in those neighborhoods, stop investing in the school system, and stop investing in economic development. So today we have neighborhoods that still have a lot of housing instability, food insecurity where there isnt access to healthy foods, and a lack of access to parks for physical activity and recreation. We know those factors increase the risk of chronic diseases that have been associated with COVID-19.

The third bucket is that African American and Latino residents are more likely to be working in jobs in the service sector that are considered essential during the pandemic the food service industry, environmental services, security, public transportation. They have had to continue to go to work, often without proper personal protective equipment, especially at the beginning of the pandemic, so they were more likely to be exposed. Many of them are also living in crowded, multigenerational housing.

All of those things contribute to increased COVID exposure. Then if the population is also more likely to have a risk of diabetes, heart disease and lung disease because of these historical issues, and on top of that they are more likely to be exposed to and infected by COVID, that is going to result in worse outcomes. Its not so much that these diseases make you more susceptible to infection; its that they contribute to a poorer outcome once you get infected.

Nickens: Does it frustrate you that some believe Black and Hispanic residents are more likely to be infected by COVID-19 because of genetics?

Golden: Theres nothing genetic about being housing insecure, food insecure, or living in an environment where you have exposure to chemicals that increase your risk of chronic diseases. Those are social and institutional contributors to health that have nothing to do with a persons genetics.

Nickens: Are the lifestyle changes needed to guard against the virus harder to make in poorer neighborhoods?

Golden: They can be. Fortunately, now there are all kinds of masks available. Washing your hands frequently is critical. But if you are living in crowded housing, it can make social distancing difficult to impossible.

One thing important to recognize in the African American community is that everybody who is dying from COVID is not low-income and living in these types of circumstances. There are well-off African Americans who also have diabetes, obesity or cardiovascular disease who are dying from COVID. Even when you are in a situation where you can implement those public health practices, this population is still very much at risk.

Nickens: A lot has been written about historical stress contributing to this situation.

Golden: I think it is a significant contributor. African Americans are more likely to contract COVID and more likely to die. The Hispanic population is more likely to get COVID, but the death rate is not as high and is closer to that of white people. Part of the reason is that Hispanics who are getting infected are younger. But I also think the difference is that African Americans have been exposed to generational stress that results from dealing with discrimination in every aspect of life. Our Latino immigrant community has come to the U.S. more recently, so there hasnt been the same amount of time for that chronic stress to perhaps have as significant impact in terms of mortality.

We really should be thinking about how we eliminate that discriminatory stress for all of our vulnerable communities.

Nickens: What have you seen from the government and the medical community that has been effective in helping people of color and low-income communities deal with the virus?

Golden: Meeting people where they are in the community is key. Those who are undocumented immigrants dont have access to all of the usual benefits that citizens have. In Baltimore, we have established partnerships with community organizations and corporations to get meals delivered to them. We are also using our Johns Hopkins excess testing capacity to provide mobile testing in the community where there are hot spots.

If you are partnering with trusted community partners, they can also help you with contact tracing. People are often uncomfortable about wanting to say who they have been in contact with, but we have to know who they have been in contact with if they are infected so that we can make quarantine and isolation recommendations to stop the spread of the virus.

Nickens: Do you have any hope for positive structural change to come out of this pandemic as these disparities are highlighted?

Golden: Ive been a doctor for 26 years. When youre in medical school, youre told you are going to use this medicine to treat this disease and the patient is going to get better. Then you start practicing, and you realize there are all of these extraneous factors that contribute to the ability of the patient to get the medicine and to take the medicine. We have to really think about how we use our policies and legislation to address these structural, social determinants of health.

One fire hydrant is required for so many houses in a neighborhood. It seems like for so many houses, there should be a store where you can get affordable fresh fruits and vegetables and healthy food. How do we use our power of legislation to address these issues? As we think about good health, much of this needs to happen in collaboration with the health care system but also outside of it. Thats a very different way of thinking than when I was in medical school in the early 90s.

Nickens: From the news coverage you have seen on racial disparities regarding the virus, are there any particular points where the coverage is off-base or where journalists could be more thoughtful about how they approach the issue?

Golden: It is important for journalists to report on and raise awareness about the contribution of structural racism to the social determinants of health that are foundational to the disparities in COVID-19 and the chronic medical conditions that worsen outcomes from COVID-19. This will prevent reporting suggesting that it is just the chronic diseases and that it is the fault of the vulnerable populations for making poor health choices. I recall seeing such reports early during the pandemic, and they were very upsetting because they assume that everyone lives in an environment where they can make healthy choices; unfortunately, that is not the case.

It is also important to emphasize that it is not only poor African Americans who are dying from COVID-19 but also those who are adequately resourced, further shedding light on the generational impact of racism and the resulting stress on health.

Nickens: What have we missed in this conversation?

Golden: Im an African American physician. I have been shocked by how many people have died from COVID-19. Im flabbergasted that we could have this many deaths and a quarter of them are in my own community. My husband and I both know people who have had COVID-19 or have died from it. Its horrible, but if it can actually wake us up to think about what we really need to do to deliver adequate care to people and advocate for environmental justice, that would be a great outcome.

Dr. Sherita Hill Golden is vice president and chief diversity officer for Johns Hopkins Medicine. Her expertises include cardiovascular diseases, diabetes, diabetes mellitus, endocrinology and lipid disorders.

Tim Nickens recently retired as editor of editorials for the Tampa Bay Times. He and a colleague won the 2013 Pulitzer Prize for editorial writing that successfully persuaded Pinellas County to resume adding fluoride to drinking water. This is part of a series funded by a grant from the Rita Allen Foundation to report and present stories about the disproportionate impact of the virus on people of color, Americans living in poverty and other vulnerable groups.

View post:

Why Black and Hispanic residents are more likely to become infected by the coronavirus - Poynter

The Vital Role Genetic Testing Plays in Bladder Cancer Treatments – Cancer Therapy Advisor

Sponsored Content by the Janssen Pharmaceutical Companies of Johnson & Johnson

Recent research suggests cancer patients who have undergone genetic testing for inherited or acquired mutations were more likely to have prolonged overall survival than patients who did not, as genetic tests can direct healthcare providers to targeted treatment paradigms featuring therapies specific to an individuals genetic makeup.1,2

Various genetic alterations impact how specific tumors can arise or develop, including BRCA1/2, p53, HER2 and the RAS family of genes, among others.3 As researchers continue to study how genetic changes impact cancer development, findings have led to improvements in cancer care including the increased use of targeted therapies and early detection strategies.3

Treating with an inhibitor or a targeted therapy may slow or halt uncontrolled cell growth that may be driven, in part, by a genetically-altered cancer and can provide clinical benefits to patients, said Tracy McGowan, MD, Strategic Area Lead for Medical Affairs at Janssen Biotech, Inc. Analyzing a patients DNA or RNA for actionable genetic alterations can help direct a cancer patients treatment plan, and is intended to provide patients and providers with treatment options that are specific to the unique characteristics of an individuals specific tumor type.

An example of the above is the fibroblast growth factor receptor (FGFR) genetic alteration, which occurs in approximately one in five patients with metastatic urothelial carcinoma (mUC).4,5 FGFR genes, a family of receptor tyrosine kinases, impact tumor cell proliferation, migration and survival in both non-muscle-invasive bladder cancer (NMIBC) and muscle-invasive bladder cancer (MIBC) including mUC.6 This is also true in a variety of other tumor types as well.4

Dr. McGowan is hopeful that, over time, more healthcare providers will recommend genetic testing earlier in the course of bladder cancer to ensure that appropriate treatment options are available for consideration. In the evolving era of precision medicine, it is important to inform patients and their families about the role of genetic testing to help them navigate treatment opportunities. For healthcare providers, talking to patients about the role of genetic testing in their care plan is the beginning of important conversations especially when specific targeted therapies are now available.

References

See the original post here:

The Vital Role Genetic Testing Plays in Bladder Cancer Treatments - Cancer Therapy Advisor

CRADLE GENOMICS: Expands Executive Team, Accelerating Efforts to Transform Non-Invasive Prenatal Testing and Patient Care – Patient Daily

Cradle Genomics issued the following announcement on July 24.

Cradle Genomics today announced the expansion of its executive team with the additions ofTanya Moreno, PhD, as Vice President of Development andSue Gross, MD, as Chief Medical Officer.

"Our mission at Cradle is to deliver genetic knowledge for life, with a vision of better outcomes for every pregnancy," saidTristan Orpin, CEO of Cradle Genomics. "The outstanding additions of Dr. Moreno to lead clinical development and Dr. Gross as CMO will enable Cradle to achieve our vision of transforming prenatal testing and patient care."

Dr. Moreno has over 13 years of experience in diagnostics development and the commercialization of genomic tests. As the head of clinical sciences, in multiple clinical laboratories, she has led development programs across a broad range of advanced genomic tools to empower patients and physicians with precision medicine. Dr. Moreno earned her Ph.D. in molecular genetics and developmental biology at the California Institute of Technology and received her postdoctoral training at the Salk Institute for Biological Studies. She is widely published in peer-reviewed journals and has both led and participated in a number of research studies in consumer and clinical genomics. She is an inventor of several novel genomic applications with multiple patents pending and issued.

Dr. Gross has had a distinguished clinical career with roles at Montefiore Einstein where she was Professor of Obstetrics and Gynecology, Pediatrics and Genetics; Natera where she was Chief Medical Officer; and most recently at Mount Sinai Hospital and Sema4 where she was the Medical Director for the Reproductive Lab and Clinical Analysis Division. She is double board certified in Obstetrics and Gynecology as well as Medical Genetics.Dr. Gross has served on national and international guideline committees and has lectured and published extensively on prenatal screening and genetic testing, with a focus on new technologies and public health policy.

About Cradle GenomicsCradle Genomics is headquartered inSan Diego, California.Cradle is developing novel fetal genetic analysis and pregnancy health solutions at the earliest stages of pregnancy.For more information, visit http://www.cradlegenomics.com

Original source can be found here.

Go here to read the rest:

CRADLE GENOMICS: Expands Executive Team, Accelerating Efforts to Transform Non-Invasive Prenatal Testing and Patient Care - Patient Daily

Elevation Oncology Emerges from Stealth with $32.5M Series A to Develop Precision Medicines for Tumors Harboring Rare Genetic Driver Alterations -…

NEW YORK, July 21, 2020 /PRNewswire/ --Elevation Oncology, a clinical stage biopharmaceutical company focused on the development of precision medicines for patients with genomically defined cancers, announced today the launch of the Company with a $32.5M Series A financing, initiation of the Phase 2 CRESTONE study, and new partnerships with Next Generation Sequencing diagnostic providers including Ashion Analytics, Strata Oncology, and Tempus to explore innovative models for real-time identification, patient referral, and enrollment of patients with tumors driven by rare genomic alterations. The Series A financing was led by Aisling Capital and a syndicate of investors including Vertex Ventures HC, Qiming Venture Partners USA, Driehaus Capital Management, and BVF Partners.

"At Elevation Oncology, we envision a future in which each unique genomic testing result can be matched with a purpose-built precision medicine and bring clarity to the patient treatment journey. Focused drug development paired with open collaboration will be instrumental for our industry to fully realize the potential of precision medicine for all patients with cancer," said Shawn Leland, PharmD, RPh, Founder and Chief Business Officer of Elevation Oncology. "With our lead development program, seribantumab, and the partnerships announced today, we are taking our first steps toward this future."

Seribantumab and the Tumor-agnostic CRESTONE Study

Seribantumab is a fully human IgG2 monoclonal antibody that binds to human epidermal growth factor receptor 3 (ERBB3 or HER3). HER3 is traditionally activated through binding of its primary ligand, neuregulin-1 (NRG1). The NRG1 gene fusion is a rare genomic alteration that combines NRG1 with another partner gene to create chimeric NRG1 "fusion proteins."

Seribantumab was acquired in 2019 by Elevation Oncology, and the development program builds on prior clinical experience from over 800 patients demonstrating consistent safety and tolerability. Previous clinical trials with seribantumab did not select for tumors with an NRG1 fusion. The CRESTONE study leverages seribantumab's rational design with recent discoveries on the significance of the NRG1 gene fusion and improvements in diagnostic sensitivity. Novel preclinical data generated by Elevation Oncology demonstrating the ability of seribantumab to prevent the activation of HER3 signaling in NRG1 fusion models are supportive of CRESTONE and are expected to be released in publications and at scientific conferences later this year.

Although rare, NRG1 gene fusions are oncogenic drivers that can be found in a variety of solid tumors, including lung, pancreatic, gallbladder, breast, ovarian, colorectal, and neuroendocrine cancers, and sarcomas. Importantly, NRG1 gene fusions are mutually exclusive with other known driver mutations and are considered a unique oncogenic driver event essential for tumor cell survival. Following recent regulatory approvals of tumor-agnostic treatments associated with oncogenic drivers, CRESTONE is designed as a registration-enabling Phase 2 "basket trial" to evaluate the efficacy and safety of seribantumab in patients with any solid tumor that harbor an NRG1 fusion.

"Genomic testing and matched precision therapeutics are creating a revolution in oncology development and regulatory approval paths," said Lori Kunkel, MD, Chair of the Elevation Oncology Scientific Advisory Board and former Chief Medical Officer LOXO Oncology. "The FDA has recently approved several oncology therapeutics for tumor-agnostic indications. I am encouraged to see the evolution in our understanding of how to achieve better clinical outcomes to address the unmet clinical need among patients with a genomically defined cancer, regardless of its tissue of origin. The CRESTONE study potentially expands the actionability of genomic tests to tumors with an NRG1 fusion and is a promising approach for furthering this genomically-driven tumor-agnostic development pathway."

Innovative Models for Patient Enrollment

Sarah Cannon Research Institute(Sarah Cannon) has been selected as the first strategic site for CRESTONE and is open and enrolling patients today. Sarah Cannon's world class clinical research leadership and insights as well as additional prospectively selected clinical sites are foundational to ensuring rigorous study conduct.

"Identifying potential driver alterations, such as NRG1 gene fusions, enables us to approach cancer treatment in a more targeted way," said David Spigel, MD, Chief Scientific Officer, Sarah Cannon Research Institute at Tennessee Oncology and one of the investigators of the CRESTONE study. "Today, all cancer patients facing a treatment decision without clear standard of care should consider comprehensive genomic testing for their tumor. Collaborations across the healthcare ecosystem help to ensure that the value of each genomic test is maximized and to expand access to critical treatment opportunities for patients."

Diagnostic partnerships will enhance traditional patient enrollment in the CRESTONE study through real-time, nationwide identification of NRG1 fusion positive patients withintheAshion, Strata Oncology, Tempus, and other partner networks. Through various partnership models, patients may also be enrolled in CRESTONE through active referral to current strategic sites or "just-in-time" site initiation.

These innovative models address specific challenges encountered by genomically-driven, tumor-agnostic trials such as the rarity of genomic driver alterations and the impracticality of comprehensive clinical site coverage by both geography and organ-system of study. In addition, these models may reduce the burden on patients by minimizing the number of diagnostic tests they may need and maximizing the treatment opportunities available to them, regardless of where they may live. Bringing clinical trials to patients using the "just-in-time" site initiation model can further help to minimize travel and keep patients safe in the face of ongoing travel restrictions due to COVID-19.

"With the strong backing of a dynamic and experienced investor syndicate, Elevation Oncology is well positioned to execute on our mission of delivering precision medicines to physicians and their patients with cancer," said Steve Elms, Chairman and Interim Chief Executive Officer of Elevation Oncology and Managing Partner of Aisling Capital. "Our development approach to seribantumab sets the stage for our broader vision: the elevation of precision medicine to the forefront of every patient journey through building a collaborative industry-wide ecosystem. Together with diagnostic developers, clinical researchers, patient advocates, and the Elevation Oncology team, we are looking to build a pipeline of precision oncology medicines that can amplify each other's efforts towards our shared goal of improving patient outcomes."

About Elevation Oncology

Elevation Oncology is founded on the belief that every patient with cancer deserves to know what is driving the growth of their disease and have access to therapeutics that can stop it. We make genomic tests actionable by selectively developing drugs to inhibit the specific alterations that have been identified as drivers of disease. Together with our peers we work towards a future in which each unique test result can be matched with a purpose-built precision medicine to enable an individualized treatment plan for each patient. Our lead candidate, seribantumab, inhibits tumor growth driven by NRG1 fusions and is currently being clinically tested in the Phase 2 CRESTONE study for patients with tumors of any origin that have an NRG1 fusion. Details on CRESTONE are available at http://www.NRG1fusion.com. For more information visit http://www.ElevationOncology.com.

About Ashion Analytics

Ashion Analytics, LLC, is a CLIA-certified and CAP-accredited clinical laboratory that uses advanced genomic technologies to offer a wide range of testing capabilities to assist physicians, health systems, research and commercial partners to provide precision cancer treatments. Ashion was developed and launched by the Translational Genomics Research Institute (TGen), an affiliate of City of Hope. TGen is a pioneer in the use of genomics to identify treatment options for cancer patients.

About Strata Oncology

Strata Oncology, Inc. is a precision medicine company dedicated to transforming cancer care by building a platform to systematize precision oncology across a network of health systems and biopharma companies. Strata Oncology empowers health systems to deliver a comprehensive, system-wide precision oncology program that integrates cutting-edge tumor molecular profiling and a portfolio of biomarker-guided with routine care, so that all patients with advanced cancer have the opportunity to benefit. This large network of trial-ready health systems provides a mechanism to rapidly and predictably enroll precision therapy trials. For more information visitwww.strataoncology.com.

About Tempus

Tempus is a technology company advancing precision medicine through the practical application of artificial intelligence in healthcare. With one of the world's largest libraries of clinical and molecular data, and an operating system to make that data accessible and useful, Tempus enables physicians to make real-time, data-driven decisions to deliver personalized patient care and in parallel facilitates discovery, development and delivery of optimal therapeutics. Additionally, the TIME TrialNetwork leverages a unique and comprehensive infrastructure to bring the right clinical trials to the right patients in under two weeks.The goal is for each patient to benefit from the treatment of others who came before by providing physicians with tools that learn as the company gathers more data. For more information, visit tempus.com.

Media Contact:Elevation OncologyDavid Rosen, Argot PartnersPhone: +1 (716) 371-1125Email: [emailprotected]

SOURCE Elevation Oncology

Home

View post:

Elevation Oncology Emerges from Stealth with $32.5M Series A to Develop Precision Medicines for Tumors Harboring Rare Genetic Driver Alterations -...

Cradle Genomics Expands Executive Team, Accelerating Efforts to Transform Non-Invasive Prenatal Testing and Patient Care – PRNewswire

SAN DIEGO, July 24, 2020 /PRNewswire/ --Cradle Genomics today announced the expansion of its executive team with the additions of Tanya Moreno, PhD, as Vice President of Development and Sue Gross, MD, as Chief Medical Officer.

"Our mission at Cradle is to deliver genetic knowledge for life, with a vision of better outcomes for every pregnancy," saidTristan Orpin, CEO of Cradle Genomics. "The outstanding additions of Dr. Moreno to lead clinical development and Dr. Gross as CMO will enable Cradle to achieve our vision of transforming prenatal testing and patient care."

Dr. Moreno has over 13 years of experience in diagnostics development and the commercialization of genomic tests. As the head of clinical sciences, in multiple clinical laboratories, she has led development programs across a broad range of advanced genomic tools to empower patients and physicians with precision medicine. Dr. Moreno earned her Ph.D. in molecular genetics and developmental biology at the California Institute of Technology and received her postdoctoral training at the Salk Institute for Biological Studies. She is widely published in peer-reviewed journals and has both led and participated in a number of research studies in consumer and clinical genomics. She is an inventor of several novel genomic applications with multiple patents pending and issued.

Dr. Gross has had a distinguished clinical career with roles at Montefiore Einstein where she was Professor of Obstetrics and Gynecology, Pediatrics and Genetics; Natera where she was Chief Medical Officer; and most recently at Mount Sinai Hospital and Sema4 where she was the Medical Director for the Reproductive Lab and Clinical Analysis Division. She is double board certified in Obstetrics and Gynecology as well as Medical Genetics.Dr. Gross has served on national and international guideline committees and has lectured and published extensively on prenatal screening and genetic testing, with a focus on new technologies and public health policy.

About Cradle Genomics Cradle Genomics is headquartered inSan Diego, California.Cradle is developing novel fetal genetic analysis and pregnancy health solutions at the earliest stages of pregnancy.For more information, visitwww.cradlegenomics.com

SOURCE Cradle Genomics

Read more:

Cradle Genomics Expands Executive Team, Accelerating Efforts to Transform Non-Invasive Prenatal Testing and Patient Care - PRNewswire

How six months of pandemic have changed Illinois forever – Crain’s Chicago Business

Since Illinois reported its first case of the novel coronavirus on Jan. 24, COVID-19 has cut through every aspect of lifefrom the most basic social interactions to education, health care and travel.

In a span of six months, 168,000 Illinoisans have tested positive for the virus and more than 7,300 have died. Roughly 1.5 million people have applied for unemployment benefits in Illinois since March, employers' costs have skyrocketed as they implement new safety measures and governments statewide face steep budget gaps and rising calls for relief. More than 4,000 Chicago-area businesses shut down as stay-home restrictions and fear of infection kept customers out of restaurants and stores. Meanwhile, as case rates rise in other states, leaders are bracing for a new surge.

But in the midst of the gravest public health crisis in generations, people are adapting to a new way of life, full of e-learning, mask wearing and socially distant celebrations. Whether the pandemic will lead to permanent work flexibility, bigger investments in public health and unemployment, or meaningful moves toward equity is still unknown.

But how did we get here? Crain's asked Chicago-area decision-makers, business leaders and workers how the first six months of the pandemic in Illinoisfrom the controversial calls to the most pivotal momentshave affected them and what life is going to look like in the future as a result. Here's what they said. (Interviews have been edited for length and clarity.)

Continued here:

How six months of pandemic have changed Illinois forever - Crain's Chicago Business

The COVID-19 Host Genome Structural Variant Consortium Formed by Dr. Ravindra Kolhe at Augusta University Creates a Massive Expansion in the Scope of…

SAN DIEGO, July 24, 2020 (GLOBE NEWSWIRE) -- Bionano Genomics, Inc. (Nasdaq: BNGO) announced today that a recently formed international consortium of clinical and research sites is using its Saphyr genome imaging system to identify genomic variants that influence resistance or sensitivity to the SARS-CoV2 virus, or COVID-19 disease progression and drug response. The consortium is comparing the genome structures of those patients who show no or mild symptoms and those who show severe illness, while controlling for the known risk factors of age and chronic illness such as asthma, heart disease, diabetes, or other immune-compromising disease. The team plans to analyze at least 1,000 patient genomes with Saphyr.

The consortium was founded by Dr. Ravindra Kolhe, the Vice-Chair of Pathology and Section Chief of Molecular and Genetic Pathology at Augusta University and Dr. Alka Chaubey, Scientific Director for the Georgia Esoteric & Molecular Lab at Augusta University. Additionally, it consists of co-investigators from Baylor College of Medicine, Bostons Children's Hospital of Harvard University, Childrens National Medical Center, Columbia University, George Mason University, MD Anderson Cancer Center, the National Cancer Institute, Oregon Health and Science University, Rockefeller University, San Francisco State University, Sanford Burnham Prebys, UC San Diego, UC Santa Cruz, and Virginia Commonwealth University, with many more in the process of joining.

A number of companies have committed to supporting this effort as part of a global Tech Against Covid initiative. Rescale, the High-Performance Computing cloud platform fully integrated with Saphyr, and Amazon, a leading provider of on-demand cloud computing, are donating compute time for the Bionano data analysis. Genoox, the platform for annotation and classification of genomic variants, is donating its compute resources to analyze available sequencing data combined with Bionanos structural variation calls for an integrated analysis of small and large genomic variants.

Initial unpublished findings from the first 30 patients that have been analyzed show that Saphyr detects large amounts of structural variation in many putatively relevant genes, demonstrating that point mutations alone are unlikely to explain disease differences between patients. Bionanos Saphyr system is expected to provide the crucial structural variation data needed for a full understanding of genome structure in patients.

Dr. Ravindra Kolhe, founder of the consortium commented: We strongly believe that Bionanos Saphyr platform is uniquely capable of identifying variants that play an important role in regulating COVID-19 in patients, and may be able to explain some of the extreme variation in disease severity and progression that we see in patients. Other studies of the host genome are based on short-read sequencing or SNP-microarrays, and those technologies are unable to detect and account for the large amounts of structural variation thats present in clinically important regions of the genome. We are hopeful that our initial results will translate into discoveries that truly advance our understanding of this devastating disease, and will improve our ability to treat the sickest patients.

Erik Holmlin, PhD, CEO of Bionano Genomics commented: The COVID-19 Host Genome Structural Variation Consortium is an important expansion of Bionanos efforts to help the scientific and medical community in the development of novel, targeted, antiviral therapies or vaccines. We believe that Bionanos genome imaging technology is the only technology capable of detecting the structural variants that could protect against or predispose patients to the viral infection and influence the severity of the disease. We are thrilled that our technology is being used in a global effort to help bring this pandemic to a halt.

About Bionano GenomicsBionano is a genome analysis company providing tools and services based on its Saphyr system to scientists and clinicians conducting genetic research and patient testing. Bionanos Saphyr system is a platform for ultra-sensitive and ultra-specific structural variation detection that enables researchers and clinicians to accelerate the search for new diagnostics and therapeutic targets and to streamline the study of changes in chromosomes, which is known as cytogenetics. The Saphyr system is comprised of an instrument, chip consumables, reagents and a suite of data analysis tools, and genome analysis services to provide access to data generated by the Saphyr system for researchers who prefer not to adopt the Saphyr system in their labs. For more information, visitwww.bionanogenomics.com.

Forward-Looking StatementsThis press release contains forward-looking statements within the meaning of the Private Securities Litigation Reform Act of 1995. Words such as may, will, expect, plan, anticipate, estimate, intend and similar expressions (as well as other words or expressions referencing future events, conditions or circumstances) convey uncertainty of future events or outcomes and are intended to identify these forward-looking statements. Forward-looking statements include statements regarding our intentions, beliefs, projections, outlook, analyses or current expectations concerning, among other things: planned scope of the consortiums research; preliminary findings regarding COVID-19 through the use of Saphyr; and Saphyrs ability to contribute to research and treatment of COVID-19, including discoveries that can advance an understanding of COVID-19 and improve the ability to treat patients. Each of these forward-looking statements involves risks and uncertainties. Actual results or developments may differ materially from those projected or implied in these forward-looking statements. Factors that may cause such a difference include the risks and uncertainties associated with: the impact of the COVID-19 pandemic on our business and the global economy; general market conditions; changes in the competitive landscape and the introduction of competitive products; changes in our strategic and commercial plans; our ability to obtain sufficient financing to fund our strategic plans and commercialization efforts; the loss of key members of management and our commercial team; and the risks and uncertainties associated withour business and financial condition in general, including the risks and uncertainties described in our filings with the Securities and Exchange Commission, including, without limitation, our Annual Report on Form 10-K for the year ended December 31, 2019 and in other filings subsequently made by us with the Securities and Exchange Commission. All forward-looking statements contained in this press release speak only as of the date on which they were made and are based on management's assumptions and estimates as of such date. We do not undertake any obligation to publicly update any forward-looking statements, whether as a result of the receipt of new information, the occurrence of future events or otherwise.

CONTACTSCompany Contact:Erik Holmlin, CEOBionano Genomics, Inc.+1 (858) 888-7610eholmlin@bionanogenomics.com

Investor Relations Contact:Ashley R. RobinsonLifeSci Advisors, LLC+1 (617) 430-7577arr@lifesciadvisors.com

Media Contact:Kirsten ThomasThe Ruth Group+1 (508) 280-6592kthomas@theruthgroup.com

Go here to see the original:

The COVID-19 Host Genome Structural Variant Consortium Formed by Dr. Ravindra Kolhe at Augusta University Creates a Massive Expansion in the Scope of...

UBC scientists sequencing the genomes of Canadians with COVID-19 – UBC Faculty of Medicine

For years, genome sequencing has helped scientists better understand the factors that predispose humans to disease. Now, the tool is being used in the fight against COVID-19.

Through the Canadian COVID Genomics Network (CanCOGeN)a new initiative launched by Genome Canada and backed by $40 million in federal fundingscientists across Canada are collaborating on genomics-based research to find solutions to COVID-19.

Dr. Terry Snutch

Among those involved are UBCs Dr. Terry Snutch and Dr. Steven Jones, both professors in the faculty of medicine. Dr. Snutch, professor in UBCs Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health, is leading the $20 million COVID-19 genome sequencing component of the initiative, called CanCOGeN-VirusSeq. Meanwhile, Dr. Jones, who is co-director and head of bioinformatics at Canadas BC Michael Smith Genome Sciences Centre (GSC) at BC Cancer, is leading the B.C. branch of the CanCOGeN-HostSeq team to sequence the genomes of 10,000 Canadians who have tested positive for COVID-19an initiative supported by another $20 million investment from the federal government.

In this Q&A, Drs. Snutch and Jones discuss how their findings could lead to better assessment and treatment of the most vulnerable COVID-19 patients.

Dr. Steven Jones

TS: By determining SARS-CoV-2 genome sequences, we can easily identify small mutations, or variants, that arise as the virus is generating billions of copies of itself. By following the trail of these variants as they appear in patients across different regions, we can determine where that variant originated and, combined with that persons travel history, when it was introduced into the region. This is crucial to distinguishing between infections arising from travel versus community spread and provides health authorities with critical information towards recommendations regarding outbreak control and where to direct resources.

SJ: We know that COVID-19 affects people differently. Seniors and those with other diseases tend to have worse symptoms and outcomes compared with young and healthy people. But some young and otherwise relatively healthy people are also seriously affected. By assessing people who have tested positive for COVID-19, and comparing the genomes of those that have responded poorly to infection to those who responded relatively well, we can tease out differences in genes that may indicate why people respond to the virus differently.

A flow cell, the site where DNA sequencing takes place. (Photo credit: GSC)

SJ: Although we did not sequence this particular virus (laboratories in China did shortly after its initial discovery in Wuhan), UBC researchers were the first in the world to sequence a related virus, SARS-COV-1, that was responsible for the SARS epidemic in 2003. We know from that work that understanding the sequence of the virus genome can accelerate the process of finding a vaccine.

TS: The virus genome codes for about two dozen proteins. By chance, some mutations will arise spontaneously in proteins that are currently targeted for vaccine development. It is crucial that we identify and understand how viral mutations appear in vaccine-targeted proteins. If the virus mutates at a critical targeted site, a vaccine that took months or years to develop might only be partially effective by the time it is available to the public.

TS: On the genome data side, I hope that the effort will evolve into the collective Canada-wide ability to track the virus in near real time (e.g., 24-48 hours) thus becoming an invaluable tool towards COVID-19 surveillance as new outbreaks arise and the predicted second wave appears. I also hope that the viral genome data will allow us to predict and track the development of viral resistance to therapeutics treatments.

SJ: The genes associated with outcomes for COVID-19 are part of a complex puzzle. We think there is a genetic connection associated with disease trajectory and outcome, but we wont know the extent of that until we complete this study analyzing the genomes of people affected by the virus. The studies that come from this work will tell us a lot more about the genetics of viral infections. While we dont know the immediate impact the current COVID-19 pandemic, we intend to be better armed with knowledge of the behaviour of this virus, which will help us to be better prepared for future pandemics.

Project collaborators include Dr. Richard Harrigan, professor in the UBC department of medicine, Dr. Marco Marra, head of the UBC department of medical genetics and director of Canadas Michael Smith Genome Science Centre at the BC Cancer Research Institute, and Dr. Naveed Aziz, chief administrative and chief scientific officer at CGEn.

See the article here:

UBC scientists sequencing the genomes of Canadians with COVID-19 - UBC Faculty of Medicine

Rare genetic mutation leaves people at higher risk for multiple cancers – Penn: Office of University Communications

Rare inherited mutations in the bodys master regulator of the DNA repair systemthe TP53 genecan leave people at a higher risk of developing multiple types of cancer over the course of their lives. Now, for the first time, a team led by researchers in theBasser Center for BRCAat theAbramson Cancer Center details the potential implications of a lower risk TP53 mutation, including an association with a specific type of Li-Fraumeni syndrome (LFS), an inherited predisposition to a wide range of cancers. The findings raise questions about how to appropriately screen patients for this mutation and whether the standard process of full-body scans for LFS patients should be modified for this group, since their risk profile is different than those with classic LFS. The researchers published their findingsinCancer Research, a journal of the American Association for Cancer Research.

Mutations in the TP53 gene are the most commonly acquired mutations in cancer. The p53 protein, made by the TP53 gene, normally acts as the supervisor in the cell as the body tries to repair damaged DNA. Different mutations can determine how well or how poorly that supervisor is able to direct the response. The more defective the mutation, the greater the risk. When TP53 mutations are inherited, they cause LFS, a disease that leaves people with a 90 percent chance of developing cancer in their lifetime. There are currently no therapies that target the p53 pathway.

Researchers determined that there is an inherited set of genetic material shared among people who have this mutation, suggesting its whats called a founder mutationa mutation that tracks within one ethnicity. In this case, that ethnicity is the Ashkenazi Jewish population.

Due to the wide variety of disease types associated with inherited TP53 mutations and the early age of cancer diagnoses, cancer screening is exceptionally aggressive. However, we do not yet know if all mutations require the same high level of screening, says the studys senior authorKara N. Maxwell, an assistant professor of hematology-oncology and Genetics in thePerelman School of Medicine and a member of the Abramson Cancer Center and the Basser Center for BRCA. It is therefore critical to study the specifics of individual TP53 mutations so we can understand how best to screen people who carry lower risk mutations.

Read more at Penn Medicine News.

Continue reading here:

Rare genetic mutation leaves people at higher risk for multiple cancers - Penn: Office of University Communications

Cleveland Clinic Researchers Identify Genetic Factors that May Influence COVID-19 Susceptibility – Health Essentials from Cleveland Clinic

A new Cleveland Clinic study has identified genetic factors that may influence susceptibility to COVID-19. Published today in BMC Medicine, the study findings could guide personalized treatment for COVID-19.

While the majority of confirmed COVID-19 cases result in mild symptoms, the virus does pose a serious threat to certain individuals. Morbidity and mortality rates rise dramatically with age and co-existing health conditions, such as cancer and cardiovascular disease. However, even young and otherwise healthy individuals have unpredictably experienced severe illness and death. These clinical observations suggest that genetic factors may influence COVID-19 disease susceptibility, but these factors remain largely unknown.

In this new study, a team of researchers led by Feixiong Cheng, Ph.D., of Cleveland Clinics Genomic Medicine Institute, investigated genetic susceptibility to COVID-19 by examining DNA polymorphisms (variations in DNA sequences) in the ACE2 and TMPRSS2 genes. These genes produce enzymes (ACE2 and TMPRSS2, respectively) that enable the virus to enter and infect human cells.

Because we currently have no approved drugs for COVID-19, repurposing already approved drugs could be an efficient and cost-effective approach to developing prevention and treatment strategies, Cheng said. The more we know about the genetic factors influencing COVID-19 susceptibility, the better we will be able to determine the clinical efficacy of potential treatments.

Looking at 81,000 human genomes from three genomic databases, they found 437 genetic variants in the protein-coding regions of ACE2 and TMPRSS2. They identified multiple polymorphisms in both genes that offer potential explanations for different genetic susceptibility to COVID-19 as well as for risk factors.

These findings demonstrate a possible association between ACE2 and TMPRSS2 polymorphisms and COVID-19 susceptibility, indicating that identification of the functional polymorphisms of these variants among different populations could pave the way for precision medicine and personalized treatment strategies for COVID-19.

However, all investigations in this study were performed in general populations, not with COVID-19 patient genetic data. Therefore, Cheng calls for a human genome initiative to validate the teams findings and to identify new clinically actionable variants to accelerate precision medicine for COVID-19.

This study was supported by the National Heart, Lung, and Blood Institute and the National Institute of Aging (both part of the National Institutes of Health) as well as Cleveland Clinics VeloSano Pilot Program. Serpil Erzurum, M.D., chair of Cleveland Clinics Lerner Research Institute, and Charis Eng, M.D., Ph.D., chair of the Genomic Medicine Institute, are co-authors of the study.

More here:

Cleveland Clinic Researchers Identify Genetic Factors that May Influence COVID-19 Susceptibility - Health Essentials from Cleveland Clinic

Rhythm Pharmaceuticals Announces Appointment of David Meeker, MD, as Chief Executive Officer – BioSpace

BOSTON, July 20, 2020 (GLOBE NEWSWIRE) -- Rhythm Pharmaceuticals, Inc. (Nasdaq:RYTM), a late-stage biopharmaceutical company aimed at developing and commercializing therapies for the treatment of rare genetic disorders of obesity, today announced that David Meeker, M.D., the Chairman of Rhythms Board of Directors, has been appointed as the President and Chief Executive Officer (CEO) of the company, effective immediately. Dr. Meeker succeeds Hunter Smith, the Companys Interim President and CEO and Chief Financial Officer (CFO), who will continue in his role as CFO.

I am delighted to announce Davids appointment as Rhythms new CEO, said Hunter Smith, CFO of Rhythm. Since he joined our Board in 2015, David has played a key role in shaping the clinical and commercial strategy for setmelanotide and in fostering our collaborative and patient-focused culture. As we continue to work toward the first potential approval of setmelanotide in pro-opiomelanocortin (POMC) and leptin receptor (LEPR) deficiency obesities later this year, Davids extensive experience leading commercial organizations and managing the launches of new medicines for rare genetic diseases, coupled with his proven ability to build strong relationships with patient and clinician communities, will be invaluable. The Rhythm team is energized by the opportunity to work more closely with David in an effort to deliver setmelanotide and potentially transform the care of people living with rare genetic disorders of obesity.

Rhythm is an exciting company that I have long admired, both for its scientifically-rigorous approach to drug development and its commitment to patients with rare genetic disorders of obesity, said David Meeker, M.D. With setmelanotide, we have the opportunity to bring one of the first meaningful therapeutic candidates to a segment of that community in dire need. Moreover, we hope our efforts will create visibility for rare genetic disorders of obesity, enabling better care for the people affected and catalyzing ongoing research efforts globally. The current management team has done a great job leading the organization through the transition and I am honored to take the CEO role.

Dr. Meeker has served as Chairman of Rhythm Pharmaceuticals since April 2017 and as a member of the Board since November 2015. Most recently, he served as President and CEO of KSQ Therapeutics. Previously, Dr. Meeker was the Executive Vice President and Head of Sanofi Genzyme, the specialty-care global business unit of Sanofi that focuses on rare diseases, multiple sclerosis, oncology and immunology. Dr. Meeker joined Genzyme in 1994 as Medical Director and, over the course of his tenure, served the company as Vice President of Medical Affairs, Chief Operating Officer, and Chief Executive Officer. He led Genzyme's commercial organization and global market access functions and managed the launch of several treatments for rare genetic diseases, including Aldurazyme, Fabrazyme and Myozyme. Prior to his tenure with Genzyme, Dr. Meeker was Director of the Pulmonary Critical Care Fellowship at the Cleveland Clinic and an Assistant Professor of Medicine at Ohio State University. Dr. Meeker earned his M.D. from the University of Vermont Medical School and completed the advanced management program at Harvard Business School.

About Rhythm PharmaceuticalsRhythm is a late-stage biopharmaceutical company focused on the development and commercialization of therapies for the treatment of rare genetic disorders of obesity. The U.S. Food and Drug Administration (FDA) has accepted for filing Rhythms New Drug Application (NDA) for setmelanotide for the treatment of POMC deficiency obesity and LEPR deficiency obesity with Priority Review of the NDA and assigned a Prescription Drug User Fee Act (PDUFA) goal date of November 27, 2020. Rhythm also submitted a Marketing Authorization Application (MAA) for setmelanotide to treat individuals living with POMC deficiency obesity or LEPR deficiency obesity to the European Medicines Agency (EMA) in June 2020. Rhythm is also evaluating setmelanotide for reduction in hunger and body weight in a pivotal Phase 3 trial in people living with Bardet-Biedl and Alstrm syndromes, with topline data from this trial expected in the fourth quarter of 2020 or early in the first quarter of 2021. Rhythm is leveraging the Rhythm Engine -- comprised of its Phase 2 basket study, TEMPO Registry, GO-ID genotyping study and Uncovering Rare Obesity program -- to improve the understanding, diagnosis and potentially the treatment of rare genetic disorders of obesity. For healthcare professionals, visitwww.UNcommonObesity.comfor more information. For patients and caregivers, visitwww.LEADforRareObesity.comfor more information. The company is based inBoston, MA.

Forward-Looking Statements This press release contains forward-looking statements within the meaning of the Private Securities Litigation Reform Act of 1995. All statements contained in this press release that do not relate to matters of historical fact should be considered forward-looking statements, including without limitation statements regarding the potential, safety, efficacy, and regulatory and clinical progress of setmelanotide, including anticipated timing of data readouts and our expectations surrounding potential regulatory approvals and timing thereof. Statements using words such as expect, anticipate, believe, may, will and similar terms are also forward-looking statements. These statements are neither promises nor guarantees, but involve known and unknown risks, uncertainties and other important factors that may cause our actual results, performance or achievements to be materially different from any future results, performance or achievements expressed or implied by the forward-looking statements, including, but not limited to, the impact of our management transition, our ability to enroll patients in clinical trials, the design and outcome of clinical trials, the impact of competition, the ability to achieve or obtain necessary regulatory approvals, risks associated with data analysis and reporting, our liquidity and expenses, the impact of the COVID-19 pandemic on our business and operations, including our preclinical studies, clinical trials and commercialization prospects, and general economic conditions, and other important factors discussed under the caption Risk Factors in our Quarterly Report on Form 10-Q for the quarterly period ended March 31, 2020 and our other filings with the Securities and Exchange Commission. Except as required by law, we undertake no obligations to make any revisions to the forward-looking statements contained in this release or to update them to reflect events or circumstances occurring after the date of this release, whether as a result of new information, future developments or otherwise.

Corporate Contact:David ConnollyHead of Investor Relations and Corporate CommunicationsRhythm Pharmaceuticals, Inc.857-264-4280dconnolly@rhythmtx.com

Investor Contact:Hannah DeresiewiczStern Investor Relations, Inc.212-362-1200hannah.deresiewicz@sternir.com

Media Contact:Adam DaleyBerry & Company Public Relations212-253-8881adaley@berrypr.com

A photo accompanying this announcement is available at https://www.globenewswire.com/NewsRoom/AttachmentNg/66fae683-bf9b-4498-9dff-20f74fa14502

David Meeker, M.D., President and Chief Executive Officer

Rhythm Pharmaceuticals Announces Appointment of David Meeker, M.D., as President and Chief Executive Officer

See original here:

Rhythm Pharmaceuticals Announces Appointment of David Meeker, MD, as Chief Executive Officer - BioSpace

MyoKardia and Fulcrum Therapeutics Announce Multi-Target Collaboration to Discover Novel Targeted Therapies for Genetic Cardiomyopathies -…

BRISBANE, Calif. and CAMBRIDGE, Mass., July 21, 2020 (GLOBE NEWSWIRE) -- MyoKardia, Inc. (Nasdaq: MYOK), a clinical-stage biopharmaceutical company discovering and developing targeted therapies for the treatment of serious cardiovascular diseases and Fulcrum Therapeutics, Inc. (Nasdaq: FULC), a clinical-stage biopharmaceutical company focused on improving the lives of patients with genetically defined rare diseases, announced today that they have entered into a strategic collaboration and license agreement to discover, develop and commercialize novel targeted therapies for the treatment of genetic cardiomyopathies.

Under the agreement, MyoKardia will access Fulcrums unique, proprietary target discovery engine to identify therapeutics that control the expression of genes that are known to be underlying drivers of genetic cardiomyopathies. The collaboration focuses joint discovery efforts on certain undisclosed genetic targets. MyoKardia will be responsible for all development and commercialization activities for, and will have global rights to, any potential therapeutics identified through this collaboration.

Fulcrum will receive a payment of $12.5 million at the close of the transaction and may be eligible to receive research, development and commercial milestone payments and additional research reimbursement of up to $302.5 million for a first product to progress through development and commercialization. If MyoKardia chooses to develop and commercialize products directed to additional targets under the collaboration, Fulcrum may be eligible for up to $150.0 million in milestone payments. Fulcrum may also be eligible to receive tiered royalty payments in the mid-single-digit to low double-digit range on net sales for any products under the collaboration that are commercialized.

MyoKardia intends to select targets for further exploration under this collaboration informed by its integrated research and development engine, which includes capabilities in translational research, proprietary and novel disease models, clinical development, and patient engagement and identification. Potential diseases associated with such undisclosed targets are expected to share common characteristics with indications currently being pursued by MyoKardia: strong and genetically-validated mechanistic rationale, high unmet patient need, potentially efficient pathway to approval, and synergy with the commercial organization that MyoKardia is building.

This partnership is a natural extension of MyoKardias investments over the last eight years in building a world-leading cardiovascular research, translational, clinical and commercial organization. We believe this collaboration will enable us to leverage our unique strengths to expand thoughtfully in identifying new therapeutic candidates for the potential treatment of heritable cardiomyopathies, said Robert S. McDowell, Ph.D., MyoKardias Chief Scientific Officer. We have been impressed by Fulcrums ability to discover new biology around genetic muscle disorders. By working together, we hope to further our mission to treat patients suffering from serious cardiovascular disease.

This collaboration highlights the broad applicability of our product engine to discover and develop new treatments in genetically defined rare diseases with high unmet need, said Robert J. Gould, Ph.D., Fulcrums President and Chief Executive Officer. We are pleased to partner with MyoKardia, a leader in the field of precision cardiovascular medicine and look forward to leveraging their unique capabilities to rapidly advance potential treatments to serve patients in urgent need.

About MyoKardia MyoKardia is a clinical-stage biopharmaceutical company discovering and developing targeted therapies for the treatment of serious cardiovascular diseases. The company is pioneering a precision medicine approach to its discovery and development efforts by (1) understanding the biomechanical underpinnings of disease; (2) targeting the proteins that modulate a given condition; (3) identifying patient populations with shared disease characteristics; and (4) applying learnings from research and clinical studies to inform and guide pipeline growth and product advancement. MyoKardias initial focus is on small molecule therapeutics aimed at the proteins of the heart that modulate cardiac muscle contraction to address diseases driven by excessive contraction, impaired relaxation, or insufficient contraction. Among its discoveries are three clinical-stage therapeutics: mavacamten (formerly MYK-461); danicamtiv (formerly MYK-491) and MYK-224.

MyoKardias mission is to change the world for people with serious cardiovascular disease through bold and innovative science.

About Fulcrum TherapeuticsFulcrum Therapeutics is a clinical-stage biopharmaceutical company focused on improving the lives of patients with genetically defined rare diseases in areas of high unmet medical need. Fulcrums proprietary product engine identifies drug targets which can modulate gene expression to treat the known root cause of gene mis-expression. The company has advanced losmapimod to Phase 2 clinical development for the treatment of facioscapulohumeral muscular dystrophy (FSHD) and is advancing losmapimod to Phase 3 for the treatment of COVID-19. Fulcrum also anticipates a regulatory filing in the second half of 2020 with FTX-6058 for the treatment of sickle cell disease.

Please visit http://www.fulcrumtx.com.

Continued here:

MyoKardia and Fulcrum Therapeutics Announce Multi-Target Collaboration to Discover Novel Targeted Therapies for Genetic Cardiomyopathies -...

Treating Colorectal Cancer in the COVID-19 Era – Medscape

This interview was originally published as part of MDedge's Blood & Cancer Podcast series. In this episode, podcast host David H. Henry, MD, of Pennsylvania Hospital, Philadelphia, spoke with David J. Kerr, CBE, MD, DSc, professor of cancer medicine at the University of Oxford in England and past president of the European Society for Medical Oncology. Kerr recently cowrote an article outlining recommendations for treating colorectal cancer during the COVID-19 pandemic. In their discussion, Henry and Kerr review those recommendations and compare notes on the practice of cancer medicine in the United States and the United Kingdom. This transcript has been edited for length and clarity.

David H. Henry, MD: As a general hematologist/oncologist, I usually think of stage II colorectal cancer (CRC) as something not to treat unless it meets several conditions. What typically pushes you to treat a patient with stage II CRC with adjuvant therapy?

David J. Kerr, CBE, MD, DSc: I was a part of the QUASAR (Quick And Simple And Reliable) trial group that published a study in 2007 showing that there was a survival benefit for offering adjuvant chemotherapy to stage II patients. The benefit was modest, with an approximately 4% survival improvement, but I feel that's enough to at least discuss adjuvant chemotherapy with patients who are under 70 and fit. We find that maybe 60% of the younger patients are keen to go ahead with adjuvant treatment, even with that very modest survival improvement. Older patients tend to look at the pros and cons and more often than not say no. But I think it's legitimate and intellectually reasonable to have the discussion with the patient.

Henry: That's something that also occurs in adjuvant chemotherapy in breast cancer, where the benefit is around 2%.

Kerr: Exactly. The QUASAR trial had about 3000 patients in it, so its reliability comes from large numbers and tight confidence intervals. Around 75% of these patients were not considered high risk.

I've got a wee bit of an ax to grind about using pathologic markers of high risk in stage II. When its capacity to identify patients with, for example, vascular and lymphatic invasion has been studied, the degree of correlation between different pathology centers can be as low as 50%. We think that these risk factors are carved in stone, yet they're often more subjective than objective. Therefore, I tend not to be too swayed by them. That's why we've got a big research group looking at more objective molecular and other markers to define those truly at high risk.

Henry: In breast cancer, we do the Oncotype DX testing all the time. Do you think that's ready for us to use yet in CRC?

Kerr: Our QUASAR group validated the Oncotype DX for colon cancer. The problem is that the risk factors associated with this assay for colon cancer are not as strong as for breast cancer. Although it is used and is a validated discriminant, the hazard ratios are relatively small, around 1.5. Whereas some of the more recent work, particularly looking at digital pathology and artificial intelligence, appears a bit more powerful, with hazard ratios up to 3 and 4. There are much stronger discriminants out there.

Henry: Discussing another patient scenario: If you saw less than 12 lymph nodes or perforation, would that push you to offer this patient adjuvant therapy?

Kerr: That would be true for the latter, but these days, less so for the first one. In the past decade and possibly longer, I've seen a remarkable global improvement in the quality of surgery and the uniformity of pathology. It's unusual now for anybody in a significant cancer hospital to see a pathology report that's got less than 12 nodes. And although I agree that it's a recognized prognostic factor, it just doesn't count as much now.

Henry: Shifting to the COVID-19 era, what are you giving and for how long?

Kerr: We use a lot of oral capecitabine. In the original placebo-controlled QUASAR studies, we were using bolus 5-fluorouracil (5-FU)/leucovorin, the so-called Mayo Clinic regimen. When the X-ACT trial came out comparing capecitabine with that regimen, the capecitabine came out very nicely. It's a much more cost-effective way for us to deliver the drug. It keeps patients out of the hospital facility. It's not a trivial drug; we still need to watch over patients. But we wanted to try to reduce the comorbidity of chemotherapy in the time of COVID, keep patients out of hospital, and reduce the burden on our chemotherapy front-line staff that is, fiddling around with infusional pumps and so on. We wanted to take the hassle factor down as low as we could. We use 6 months of capecitabine in a conventional dose (2 weeks on, 1 week off), as was reported in the X-ACT study. We were pretty happy with that.

Henry: You've also recommended measuring for enzymes to see if there's fluoropyrimidine toxicity. Do you check that on your capecitabine patients?

Kerr: Increasingly in the United Kingdom, we do that in all of our patients receiving fluoropyrimidine treatment. 5-FU, which is one of the most widely used anticancer drugs in the world, is broken down and degraded by an enzyme called dihydropyrimidine dehydrogenase. If there are genetic variants that reduce the activity of the enzyme, then consequently you get higher-than-average levels of 5-FU in the bloodstream. With some of these genetic variants, it's associated with an almost 100% risk for death.

We run genetic screens in all of our patients. That allows us to identify rarely but importantly patients who would die if we gave them 5-FU, patients who are at a high risk for grade 4 toxicity, particularly neutropenia, septic neutropenic fever, grade 4 hematology drops, and so on. In the 6 months since we instituted this, we found a significant reduction in the number of patients being admitted to our wards because of fluoropyrimidine toxicity, so that's a nice thing. It's a test that we helped to invent. We've got a fantastic genetics outfit in Oxford, and we've done quite a bit of work in improving the sensitivity and specificity of the test.

Henry: Patients with Gilbert syndrome don't metabolize irinotecan very well. Is this is a different enzyme system than with Gilbert syndrome?

Kerr: It is. But you're exactly right to mention it. It's encompassed within a whole field of pharmacogenetics, in that if we can understand what the genetic variants are that may cause somebody to either overmetabolize or undermetabolize a drug, it would allow us to individualize those.

Precision medicine has been taken over by the molecular biologists, which we like. But you and I both know, being sort of graybeards, that the thing that we control most about the delivery of any drug to any patient is its dose and schedule. And that will be determined by a host of other factors. We overdose patients, but we also underdose them. If we could individualize that better, that's got to be an important component of precision medicine.

Henry: It's certainly a step in the right direction. Sadly, I've given capecitabine to patients and one or two times had someone admitted for weeks with diarrhea that wouldn't stop tremendous toxicity by proper meter-squared dosing.

Here where I practice, there's been a bid to keep people out of the hospital and clinic by using alternate-week capecitabine. Would you be a fan of that?

Kerr: It's a clever idea for reducing patterns of toxicity. We've gone with the enzyme because we're pretty comfortable with it. But we're also prepared to dose-reduce in those patients over the age of 70. In terms of clinical pharmacology, we old folk tend to be a wee bit gentler with the chemotherapy anyway, maybe offering 80% of the conventional dose up front, for sensible reasons.

Henry: In the COVID-19 era, what are you giving your patients with stage III T3N1 disease?

Kerr: I think the T3N1 patients do as well as the stage II patients. We wouldn't be inclined to offer them anything different. I don't think there's any role for the use of oxaliplatin in the treatment of stage II disease. If we think that the clinical outcomes, natural history, and survival patterns are similar for T3N1, as has been shown repeatedly in large community studies, we treat them in the same way.

Now, wouldn't it be lovely just to give 3 months of capecitabine? But that feels a step too far now. We try to base everything on published evidence, and 6 months of capecitabine feels fine to me. My wife Rachel [Kerr, an oncologist specializing in gastrointestinal cancers at the University of Oxford] was part of the SCOT trial, which contributed to that lovely New England Journal of Medicine publication in 2018, in which there was good evidence that 3 months was as effective as 6 months. If we were seeing patients with T4N2 disease in normal circumstances, we would often offer 6 months of treatment rather than 3, splitting hairs a little. But in the time of COVID, we just wouldn't do it. We'd do 3 months.

Henry: Are you persuaded by the International Duration Evaluation of Adjuvant Therapy (IDEA) trial showing that FOLFOX (fluorouracil, leucovorin, and oxaliplatin) was beat out by CAPOX (capecitabine/oxaliplatin)?

Kerr: We cannot work in Oxford and not be terrified of Sir Richard Peto, who is the enemy of all subgroup analyses. Richard has beat us over the head with that. So I think there's a huge statistical instability in subgroup analysis.

It would be surprising if CAPOX is better than FOLFOX, because if you look at the data overall, they're indistinguishable. If anything, the needle slightly favors FOLFOX. In this particular case, the needle went the other way. I didn't pay too much attention to it. But during COVID-19, we recommend 3 months of CAPOX for reasons of reducing hassle of infusion and making life that bit safer and easier, hopefully for our patients as well as our frontline chemotherapy staff.

Henry: You offered recommendations for advanced cancer as well. I just had a patient with sigmoid colon cancer with a couple of very large metastases in the liver. We began her on CAPOX. Is that the regimen you're suggesting?

Kerr: It is. That's sort of our default go-to chemotherapy at the moment.

Henry: What are your thoughts about using bevacizumab? Not just during the COVID-19 era, but before it as well.

Kerr: We don't use bevacizumab. This is a stark difference between our two healthcare systems. I used to be quite involved in governmental health policy around cancer planning. Our National Institute for Health and Care Excellence (NICE) decided early on that the clinical benefits accrued by bevacizumab were too small to warrant its at that stage enormous cost. So we don't use bevacizumab at all.

Henry: That's very interesting, because we're looking at the same data across the Atlantic and practicing differently. I can't disagree with you. I've been underwhelmed with the difference, yet our patients sometimes say, "What's the most you could possibly give to me?" Of course, then we'd start clicking off the bevacizumab and sink the national healthcare budget.

Kerr: The study that provided initial data in metastatic CRC randomized patients to receive bevacizumab in addition to bolus 5-FU, irinotecan, and leucovorin, which is a regimen that's not widely used at all. It showed a very significant benefit for bevacizumab. I suspect that the bevacizumab was making up the gap for a somewhat inferior chemotherapy regimen.

In the big, 2000-patient studies looking at CAPOX or FOLFOX plus or minus bevacizumab, the benefits almost disappeared. There were some improvements in progression-free survival but nothing seen in overall survival at all. Therefore, in our taxation-based healthcare system, it's actually an easy decision to say, sorry, we need to pass on this one.

Henry: If you've gotten 3 months in and a patient's carcinoembryonic antigen is falling and their lesions are smaller, do you press on or give a break?

Kerr: We've done two large trials in the United Kingdom, both of which were published in The Lancet, in which we took patients who were responding to chemotherapy after 3 months and randomized them into stop-and-start or continuous chemotherapy. There was no difference in overall survival in either of those groups. These are old trials with somewhat inferior chemotherapy, but nevertheless it gave us an intellectual basis for offering chemotherapy holidays. It's a nice way to present it to patients.

If patients have got small-volume disease and are relatively well, indolent, and asymptomatic, we would give them a couple of months' holiday break and then scan again. If there's any evidence of progression, we'd restart the chemotherapy.

If patients present with large-volume disease that we know we'll lose control of as soon as we stop the chemotherapy, we'll probably move from CAPOX to single-agent capecitabine, with the same regimen, doing the scan again in a couple of months. If patients were on epidermal growth factor receptor inhibitors, then we would keep that biologic going, probably with single-agent capecitabine, dose reduced.

Henry: My patient a couple of weeks ago had a sigmoid primary, was not obstructed, in pain, or bleeding, and had a couple of liver lesions that I could feel and were a bit tender. What's your threshold for deferring start of chemotherapy in metastatic disease like that?

Kerr: You present interesting cases. With that patient, I think I'd want to do something. That feels intuitively right.

But say we have somebody who has had their primary out, so we're not worried about any obstructive problems, and they come with a couple of small liver or pulmonary metastases. If they're elderly, a bit frail, or comorbid, then I'd consider watching them for a couple of months to see what happened.

There's very little trial evidence to suggest when you and I should start chemotherapy. When I was younger, there were two trials that I knew of, one Italian and one northern European, both of which were very small. Each recruited only about 120 patients. The idea was you observe and give chemotherapy when your patients become symptomatic, or give chemotherapy straightaway and randomize to one or the other management plan. One trial was positive for early intervention and the other was negative. These are very small, difficult trials to run.

Also, I think you and I suffer from two things. First, there's a lack of clear trial information. Second, there's a lack of biological markers that allow us to tell the patient sitting in our consulting room that we think they're going to get indolent disease and we can wait, or we've seen small volume in the scans but it's nasty, so we need to get on with treatment. These are things that we and many other labs around the world are looking at. Can we get a marker for aggressiveness? Wouldn't that be a handy thing to have?

Henry: The last of your recommendations for advanced CRC dealt with chemotherapy after resection and metastases. I believe there are some data pointing to the fact that postoperative chemotherapy may make for better outcomes. But would you hold that or give it in the COVID-19 era?

Kerr: We wouldn't recommend it. I think the whole literature around both neoadjuvant and classically post-resection adjuvant chemotherapy is difficult. It's an area that we've contributed to in the past and which is littered with small trials, some almost single-center. There have been great results with intrahepatic arterial infusion, but those are difficult to apply at other centers, even Penn and Oxford. So it's a rather confused area. And if I have a degree of confusion about benefits in the time of COVID-19, we don't feel it's worthwhile considering.

Henry: I also wanted to ask about rectal cancer. We've been persuaded a lot by data on this side of the world about total neoadjuvant therapy, which I've always been a fan of in my oncology career. In breast cancer, you and the patient can see if it's working or not. And here, you can do the same thing and probably get more drug in, as opposed to waiting. Are you a fan of the total neoadjuvant therapy? And are you somewhat curtailing the radiation piece in the COVID-19 era?

Kerr: I think we are becoming fans of the total neoadjuvant therapy. It's taking a bit longer to be taken up across the United Kingdom. But the data look good; we agree about that.

As things stand now, we are trying to curtail everything. We're going from complex radio-chemotherapeutic regimens, from long-course to short-course treatment. We're looking to omit some of the drugs for example, taking oxaliplatin out rather than giving combination chemo-radiotherapy. Our colleagues in the radiation field again are looking to compress and simplify as best they can and as logically as they can.

Read more here:

Treating Colorectal Cancer in the COVID-19 Era - Medscape