Podcast appearances and mentions of George Church

  • 212PODCASTS
  • 317EPISODES
  • 45mAVG DURATION
  • 1MONTHLY NEW EPISODE
  • Mar 28, 2025LATEST

POPULARITY

20172018201920202021202220232024


Best podcasts about George Church

Latest podcast episodes about George Church

Grow Everything Biotech Podcast
122. Ecohacking the Planet: Daniel Goodwin of Homeworld Collective Brews Up Planetary-Scale Solutions

Grow Everything Biotech Podcast

Play Episode Listen Later Mar 28, 2025 60:27


Karl and Erum sit down with Daniel Goodwin, co-founding Director of Homeworld Collective alongside his friend and collaborator Paul Reginato. Together, they explore how to build resilient, values-aligned infrastructure for a flourishing bioeconomy—beyond the venture model.From climate biotech to systems thinking, Daniel shares how Homeworld Collective is more than just a network—it's an emergent, mycelial community focused on care, collaboration, and collective intelligence. The conversation weaves through everything from the limits of "techno-solutionism" to the power of story in science, and the need to design not just tools, but the conditions for innovation to thrive.If you're thinking about the future of biotech, and how culture, community, and infrastructure intersect to make it real—this episode is for you.Grow Everything brings the bioeconomy to life. Hosts Karl Schmieder and Erum Azeez Khan share stories and interview the leaders and influencers changing the world by growing everything. Biology is the oldest technology. And it can be engineered. What are we growing?Learn more at ⁠⁠⁠⁠⁠⁠⁠www.messaginglab.com/groweverything⁠⁠⁠⁠⁠⁠⁠ Chapters:00:00:00 – Welcome to the Biofuture: Erum and Karl kick things off in full grind mode.00:00:16 – Biotech Boom Times: Why the field never sleeps (and neither do the hosts).00:01:01 – Inside the AI Emergence Summit: Smell farms, AGI hype, and real talk on moats.00:03:21 – DIY Healthcare with AI: What happens when you feed your genome to a chatbot?00:05:35 – Can AI Decode Biology? Drug discovery and the limitations of human cognition.00:07:49 – Mammoths, Models & Media: Colossal Biosciences and the narrative power of science.00:11:32 – Meet Daniel Goodwin: Systems thinker, community builder, biotech whisperer.00:12:03 – On George Church: The unintentional influence that shaped Homeworld's DNA.00:18:07 – What Is Homeworld Collective? Building more than a Slack—building a system.00:20:47 – Imagining the Next Bio Era: Biotech's renaissance as culture, not just tech.00:26:52 – Idea Porn & Biotech Fiction: When storytelling shapes science (and vice versa).00:29:54 – Collaborating with Biology: Designing with, not against, nature's blueprint.00:30:07 – Rivers as Systems: What infrastructure can learn from ecosystems.00:30:54 – Biomanufacturing Is Hard: Real talk on what it takes to scale with biology.00:32:08 – Carbon Capture, Naturally: Biodesign, fermentation, and the future of CO₂.00:33:45 – Biosecurity + Ethics: Guardrails, governance, and growing with intention.00:41:18 – Prototype Like a Scientist: What biotech can borrow from product design.00:49:07 – Cleaning Up Chemistry: Synbio's role in reshaping industrial pollution.00:52:46 – Looking Ahead: Final reflections, future infrastructure, and quiet revolutions.Links and Resources:Homeworld CollectiveThe Climate Biotech PodcastKarl and Erum on the Climate Biotech PodcastAI Circle Osmo.AIDoctors Told Him He Was Going to Die. Then A.I. Saved His Life.Colossal BiosciencesHoping to revive mammoths, scientists create 'woolly mice'George Church episode, interviewed by Homeworld CollectiveNeuroscience Needs Hackers, Scientific American Liquid Trees A Natural History of the Future: What the Laws of Biology Tell Us about the Destiny of the Human Species by Robb DunnTopics Covered: climate biotech, George Church, Churchian Juxtaposition, idea porn, genetic engineering, geo biotech, environmental engineering, Have a question or comment? Message us here:Text or Call (804) 505-5553 ⁠⁠⁠⁠⁠⁠⁠Instagram⁠⁠⁠⁠⁠⁠⁠  / ⁠⁠⁠⁠⁠⁠⁠Twitter⁠⁠⁠⁠⁠⁠⁠ / ⁠⁠⁠⁠⁠⁠⁠LinkedIn⁠⁠⁠⁠⁠⁠⁠ / ⁠⁠⁠⁠⁠⁠⁠Youtube⁠⁠⁠⁠⁠⁠⁠ / ⁠⁠⁠⁠⁠⁠⁠Grow Everything⁠⁠⁠⁠⁠Email: groweverything@messaginglab.comMusic by: Nihilore

Shortcut – Schneller mehr verstehen
Mammut-Comeback für den Klimaschutz?

Shortcut – Schneller mehr verstehen

Play Episode Listen Later Mar 18, 2025 20:46


Ein Start-up plant für 2028 die Geburt eines Mammuts. Zuletzt hat die Firma immerhin flauschige Mäuse erschaffen. Wer steckt hinter der Idee und was kann daraus werden? SPIEGEL-Redakteur Johann Grolle ordnet den Plan ein. Sagt uns, wie euch Shortcut gefällt. Hier geht's zur Umfrage. »SPIEGEL Shortcut« – Schneller mehr verstehen. Wir erklären euch jeden Tag ein wichtiges Thema – kurz und verständlich. Für alle, die informiert mitreden wollen. Neue Folgen von Shortcut gibt es von Montag bis Freitag auf Spiegel.de, YouTube und überall, wo es Podcasts gibt. Links zur Folge: Sind diese Mäuse ein erster Schritt zum Comeback der Mammuts? Forscher finden fast vollständig erhaltenes Mammutbaby Flauschig, scheu – und vom Aussterben bedroht ► Host: Maximilian Sepp ► Redaktion: Ilyass Alaoui, Florian Hofmann ► Redaktionelle Leitung: Jannis Schakarian, Marius Mestermann ► Produktion: Christian Weber ► Postproduktion: Florian Hofmann, Christian Weber ► Musik: Above Zero ►►► Lob, Kritik, Themenvorschläge? Schreibt uns: hallo.shortcut@spiegel.de +++ Alle Infos zu unseren Werbepartnern finden Sie hier. Die SPIEGEL-Gruppe ist nicht für den Inhalt dieser Seite verantwortlich. +++ Den SPIEGEL-WhatsApp-Kanal finden Sie hier. Alle SPIEGEL Podcasts finden Sie hier. Mehr Hintergründe zum Thema erhalten Sie mit SPIEGEL+. Entdecken Sie die digitale Welt des SPIEGEL, unter spiegel.de/abonnieren finden Sie das passende Angebot. Informationen zu unserer Datenschutzerklärung.

The Naked Scientists Podcast
What is mirror life?

The Naked Scientists Podcast

Play Episode Listen Later Feb 25, 2025 32:44


We're taking you through the looking glass to explore 'mirror life': could we be about to flip biology on its head? Like this podcast? Please help us by supporting the Naked Scientists

Grow Everything Biotech Podcast
115. Gene-ius at Work: George Church on Climate Hacks & CRISPR Stacks – A Homeworld Collective Interview

Grow Everything Biotech Podcast

Play Episode Listen Later Feb 7, 2025 50:32


In this special Grow Everything episode swap with The Climate Biotech Podcast, Erum and Karl introduce a mind-blowing conversation with the one and only George Church. If you're into synthetic biology, you already know he's a legend—co-founding 50+ biotech companies and pushing the boundaries of what's possible with DNA. In this episode, George talks about scaling microscopic innovations to solve massive global challenges—think rewilding ecosystems, bio-mining for new molecular tools, and engineering biology to fight climate change. He even throws out wild ideas like testing space colonies on Earth before heading to Mars. Stick around after the episode for Erum and Karl's takeaways and a heads-up on a funding opportunity from Homeworld Collective! Grow Everything brings the bioeconomy to life. Hosts Karl Schmieder and Erum Azeez Khan share stories and interview the leaders and influencers changing the world by growing everything. Biology is the oldest technology. And it can be engineered. What are we growing? Learn more at ⁠www.messaginglab.com/groweverything⁠  Chapters: 00:00:00 – Microscopic, but make it massive 00:00:17 – Erum and Karl are back—let's grow everything 00:00:36 – The Homeworld Collective: where biotech meets climate 00:01:44 – Enter George Church (yes, that George Church) 00:03:34 – From river dolphins to rewriting life—George's early days 00:05:25 – Synthetic biology's glow-up (and growing pains) 00:07:18 – Climate biotech: hurdles, hype, and huge potential 00:11:32 – Can biotech be infinitely scalable and atomically precise? 00:21:36 – Inorganic synthetic biology: when life meets metal 00:24:50 – Biotech's biggest climate bets—what's next? 00:27:02 – Genetic engineering, but make it ecosystem-sized 00:29:04 – Space colonies on Earth? Let's test before Mars 00:37:26 – Rapid-fire Q&A—George Church vs. the audience 00:39:49 – Curiosity is the best lab equipment—George's advice 00:42:26 – Final thoughts from Karl and Erum, next steps, and a funding opportunity Links and Resources: The Climate Biotech Podcast Homeworld Collective Garden Grants: Biotech in Greenhouse Gas Removal Colossal Biosciences Topics Covered:  Genetic engineering, biotech, bioeconomy, research and development, climate biotechnology Have a question or comment? Message us here: Text or Call (804) 505-5553  ⁠Instagram⁠  / ⁠Twitter⁠ / ⁠LinkedIn⁠ / ⁠Youtube⁠ / ⁠Grow Everything⁠ Email: groweverything@messaginglab.com Music by: Nihilore Production by: Amplafy Media

Escuta Essa
Extinção

Escuta Essa

Play Episode Listen Later Dec 18, 2024 38:16


Deveríamos tentar fazer com que grandes pedaços de terra voltem a ser como eram há 10 mil anos? O que é deixar a natureza ser selvagem e o que é brincar de deus? Na busca pelas respostas, o auroque, o dodô e o mamute, três das mais famosas espécies já extintas, podem voltar à vida. Este é mais um episódio do Escuta Essa, podcast semanal em que Denis e Danilo trocam histórias de cair o queixo e de explodir os miolos. Todas as quartas-feiras, no seu agregador de podcasts favorito, é a vez de um contar um causo para o outro. Não deixe de enviar os episódios do Escuta Essa para aquela pessoa com quem você também gosta de compartilhar histórias e aproveite para mandar seus comentários e perguntas no Spotify, nas redes sociais , ou no e-mail escutaessa@aded.studio. A gente sempre lê mensagens no final de cada episódio! ... NESTE EPISÓDIO •⁠ ⁠O parque Oostvaardersplassen, nos Países Baixos, teve sua história contada pela jornalista Elizabeth Kolbert ⁠na revista New Yorker em 2012⁠. A matéria foi traduzida ⁠para a revista piauí em 2013⁠. •⁠ ⁠Em 2022, ⁠⁠o jornal Guardian falou sobre como cresceu o intervencionismo⁠⁠ humano na área selvagem. •⁠ ⁠O livro que detalha a história dos auroques e sua importância para as culturas asiática e europeia é o ⁠"Retracing The Aurochs: History, Morphology & Ecology of an Extinct Wild Ox"⁠, de Cis Van Vuure. •⁠ ⁠A startup que pretende recriar mamutes e dodôs ⁠é a Colossal Biosciences⁠, fundada por George Church. •⁠ ⁠Para conhecer mais sobre o projeto de fazer a Europa selvagem de novo, você pode conferir ⁠o site do grupo Rewilding Europe⁠. ... AD&D STUDIO A AD&D produz podcasts e vídeos que divertem e respeitam sua inteligência! Acompanhe todos os episódios em ⁠aded.studio ⁠para não perder nenhuma novidade.

The Unadulterated Intellect
#82 – William Leonard Pickard, George Church, Glenn Cohen, Ruth L. Okedij, Tina Liu, and Alex Zhavoronkov: Petrie-Flom Center Open House – Health Law, Biotechnology, and the Future (09/19/2024)

The Unadulterated Intellect

Play Episode Listen Later Dec 12, 2024 66:13


(00:00) Introduction (02:27) Petrie-Flom Center Open House – Health Law, Biotechnology, and the Future (51:55) Q&A --- Support this podcast: https://podcasters.spotify.com/pod/show/theunadulteratedintellect/support

The Foresight Institute Podcast
Existential Hope Podcast: Adam Marblestone | Solving Science's Biggest Gaps

The Foresight Institute Podcast

Play Episode Listen Later Nov 29, 2024 51:24


Adam Marblestone is the CEO of Convergent Research. He is working with a large and growing network of collaborators and advisors to develop a strategic roadmap for future FROs. Outside of CR, he serves on the boards of several non-profits pursuing new methods of funding and organizing scientific research including Norn Group and New Science, and as an interviewer for the Hertz Foundation. Previously, he was a Schmidt Futures Innovation Fellow, a Fellow with the Federation of American Scientists (FAS), a research scientist at Google DeepMind, Chief Strategy Officer of the brain-computer interface company Kernel, a research scientist at MIT, a PhD student in biophysics with George Church and colleagues at Harvard, and a theoretical physics student at Yale. He has also previously helped to start companies like BioBright, and advised foundations such as Open Philanthropy.Session SummaryIn this episode of the Existential Hope Podcast, our guest is Adam Marblestone, CEO of Convergent Research. Adam shares his journey from working on nanotechnology and neuroscience to pioneering a bold new model for scientific work and funding: Focused Research Organizations (FROs). These nonprofit, deep-tech startups are designed to fill critical gaps in science by building the infrastructure needed to accelerate discovery. Tune in to hear how FROs are unlocking innovation, tackling bottlenecks across fields, and inspiring a new approach to advancing humanity's understanding of the world.Full transcript, list of resources, and art piece: https://www.existentialhope.com/podcastsExistential Hope was created to collect positive and possible scenarios for the future so that we can have more people commit to creating a brighter future, and to begin mapping out the main developments and challenges that need to be navigated to reach it. Existential Hope is a Foresight Institute project.Hosted by Allison Duettmann and Beatrice ErkersFollow Us: Twitter | Facebook | LinkedIn | Existential Hope InstagramExplore every word spoken on this podcast through Fathom.fm. Hosted on Acast. See acast.com/privacy for more information.

Aging-US
Aging's Commitment to Advancing Research: Sponsoring the “Future of Aging Research Mixer”

Aging-US

Play Episode Listen Later Nov 20, 2024 4:02


Aging (Aging-US) was a proud sponsor of the “Future of Aging Research Mixer 2024” hosted by the Aging Initiative at Harvard University on November 15 in Boston. This event united a vibrant community of students, researchers and technologists, all driven by a shared mission: advancing innovations in aging research and longevity science. Key Highlights from the Future of Aging Research Mixer 2024 The event kicked off with inspiring opening remarks and a keynote by George Church, professor at Harvard Medical School, founding member of the Wyss Institute, and co-founder of over 50 biotech companies. He was joined by Kat Kajderowicz, an MIT PhD student and Principal at age1. Together, they highlighted the interdisciplinary nature of aging research and its immense potential to drive transformative advancements. Jesse Poganik, HMS Instructor in Medicine and Executive Co-Director of the Biomarkers of Aging Consortium, discussed the evolution of aging science and the critical role biomarkers play in understanding aging processes and assessing the effectiveness of interventions aimed at slowing or reversing age-related changes. Alex Colville, co-founder and general partner at age1, explained how venture capital can accelerate innovation in longevity biotechnology. He shared career advice for aspiring researchers and paid tribute to his mentor, Dr. David Sinclair, a pioneer in aging research. These talks highlighted the importance of mentorship, interdisciplinary collaboration, and investment in driving progress in the aging research field. Read the full summary - https://aging-us.org/2024/11/agings-commitment-to-advancing-research-sponsoring-the-future-of-aging-research-mixer/ About Aging-US The mission of the journal is to understand the mechanisms surrounding aging and age-related diseases, including cancer as the main cause of death in the modern aged population. The journal aims to promote 1) treatment of age-related diseases by slowing down aging, 2) validation of anti-aging drugs by treating age-related diseases, and 3) prevention of cancer by inhibiting aging. (Cancer and COVID-19 are age-related diseases.) Please visit our website at https://www.Aging-US.com​​ and connect with us: Facebook - https://www.facebook.com/AgingUS/ X - https://twitter.com/AgingJrnl Instagram - https://www.instagram.com/agingjrnl/ YouTube - https://www.youtube.com/@AgingJournal LinkedIn - https://www.linkedin.com/company/aging/ Pinterest - https://www.pinterest.com/AgingUS/ Spotify - https://open.spotify.com/show/1X4HQQgegjReaf6Mozn6Mc MEDIA@IMPACTJOURNALS.COM

Do you really know?
What is de-extinction?

Do you really know?

Play Episode Listen Later Sep 29, 2024 4:25


What is de-extinction? The process of de-extinction is all about bringing extinct species back to life. It became known in the 2010s, thanks to an ambitious plan from American geneticist George Church, who dreamed of resurrecting the woolly mammoth, an animal which has been extinct for 3700 years. Fast forward to the 2020s and Church's bioscience startup Colossal is now eding closer towards making that a reality. As of March 2022, Colossal announced that it had raised a total of $75M for the project. Is it really possible to revive extinct animal species? Why would we want to bring extinct species back to life? How is the idea of de-extinction being received by the scientific community? In under 3 minutes, we answer your questions! To listen to the last episodes, you can click here: What are happy hormones? What is erectile dysfunction? Who are Arcade Fire? A podcast written and realised by Joseph Chance. First broadcast: 10/05/2022 Learn more about your ad choices. Visit megaphone.fm/adchoices

Pharma and BioTech Daily
Pharma and Biotech Daily: Your Daily Dose of What Matters in the Industry

Pharma and BioTech Daily

Play Episode Listen Later Sep 23, 2024 3:54


Good morning from Pharma and Biotech daily: the podcast that gives you only what's important to hear in Pharma and Biotech world.Senseonics has received FDA clearance for a one-year continuous glucose monitoring system. The company's commercial partner, Ascensia, is in talks with insulin pump manufacturers to create an automated insulin delivery system. Zimmer is pulling its hip implant off the market due to fracture risk, Bausch + Lomb is considering a sale, and Masimo investor claims a proxy war win as CEO Joe Kiani fails to retain a board seat. The wearables market is expected to see significant growth, with adhesives playing a vital role in development. Other noteworthy stories include DermaSensor's skin cancer detection device, a Senate committee holding Steward CEO in contempt, and Medtronic detailing a study of adaptive neurostimulation in Parkinson's disease.Brisk Iced Tea brought back its classic claymation ad campaign featuring Doja Cat to target Gen Z. E.l.f. launched a skincare campaign with "Sinfluencer" Megan Stalter. Pizza Hut put resumes on boxes to help job seekers stand out. Frito-Lay used burner phones in a campaign targeting Gen Z for Flamin' Hot Cheetos. Chipotle planned a digital scavenger hunt to give out $1 million in free quesadillas. Customers' trust in AI is low, impacting purchasing decisions.Cigna is scaling back its Medicare Advantage footprint in 8 states next year, affecting roughly 5,400 members, mostly in Florida. Johnson & Johnson is facing sanctions over a plan to rebate 340B drugs, while Express Scripts is suing the FTC over a report criticizing pharmacy benefit managers. The US is behind other wealthy nations in healthcare despite spending the most, with a Commonwealth Fund analysis ranking the US last in access to care and health outcomes.Cigna is reducing its Medicare Advantage footprint in 8 states, affecting around 5,400 members, mainly in Florida. CarePoint Health may lay off 2,600 employees due to financial struggles. The House committee passed a bill to extend telehealth flexibilities in Medicare for another two years. Steward Health Care auctioned assets, with Christus Health set to purchase a Texas-based medical center for $4.5 million.MilliporeSigma is partnering with an experienced viral vector CDMO to offer solutions for the production of cell and gene therapies. They invite attendees of the Mesa conference in Phoenix to visit their booth or schedule a meeting to learn about their viral vector development and manufacturing capabilities.The Senate has voted to hold Steward CEO in contempt for defying a congressional subpoena. The US healthcare system lags behind other wealthy nations in performance despite spending the most. Oak Street Health pays $60 million to settle allegations of a kickback scheme, and J&J faces sanctions over a plan to rebate 340B drugs.Medtech news from September 19, 2024, includes updates on various companies and developments in the industry. Masimo's proxy fight is coming to an end, with a shareholder vote looming. Axonics wins a patent dispute against Medtronic, while Boston Scientific's proposal to buy Axonics is still under review.A startup from George Church's lab has raised $75 million to develop 'supercell' medicines, claiming it can create off-the-shelf therapies from induced pluripotent stem cells faster and simpler than current methods. Roche's Xofluza has shown promising results in reducing flu transmission in a phase 3 study, marking the first time an antiviral drug has achieved this in a global trial.The text discusses the latest advances in oncology research presented at the ESMO conference, focusing on the challenges faced by the pharmaceutical industry in expanding patient access and improving cancer research outcomes.The text promotes a webinar by QT Group on September 26th focusing on continuous compliance for medical software, specifically addressing how to maintain software quality and compliance with medical standa

Learning With Lowell
Dr. Peter Lidsky: Cracking the Aging Code: Investigating Programmed Adaptation – 249

Learning With Lowell

Play Episode Listen Later Aug 13, 2024 61:48


Dr. Peter Lidsky posits that aging is not merely a process of accumulating damages, as commonly believed, but rather a programmed adaptation akin to a part of the immune system. His hypothesis suggests that aging evolved to eliminate older individuals who might be susceptible to severe chronic diseases. Check out these other great views as well:Interview with Aubrey De Grey: https://youtu.be/DHvoPlcWwg4Interview with George Church: https://youtu.be/yNplpykf0B4Interview with Lisa Fabiny-Kiser: https://youtu.be/bE5jEGE5-OMInterview with Matt Kaeberlein: https://youtu.be/9QJ_ak3a05QInterview with Nir Barzilai : https://youtu.be/LuOAShcjOt0Interview with Michael Levin : https://youtu.be/5XvDdbYzwf8 About the showThe Learning With Lowell show is a series for the everyday mammal. In this show we'll learn about leadership, science, and people building their change into the world. The goal is to dig deeply into people who most of us wouldn't normally ever get to hear. The Host of the show – Lowell Thompson- is a lifelong autodidact, serial problem solver, and founder of startups. Learning with lowell linksYoutube: https://www.youtube.com/channel/UCzri06unR-lMXbl6sqWP_-QYoutube clips:https://www.youtube.com/channel/UC-B5x371AzTGgK-_q3U_KfAWebsite: https://www.learningwithlowell.com/ Peter Lidsky linkshttps://twitter.com/LidskyPeterhttps://www.linkedin.com/in/peter-lidsky-71997540 Timestamps00:00 Start00:10 who is Peter Lidsky / research / longevity01:30 Academia differences04:20 Aging explained and the Squirrel test07:20 Pathogen control hypothesis15:50 Lifespans different organisms (fanq)17:55 Natural experiments to prove/disprove (fanq)24:30 Implications of Theory of aging25:00 Senescent cells31:32 Therapeutics / interventions from his research33:00 Implications of his theory37:50 Limits of aging40:10 What do people need to see to believe his theory44:30 Show him the drug that is better than rapamycin46:40 Data sharing / organization to push forward48:50 What is slowing aging research49:50 Pleiotropic genes / aging52:00 Research at Hong Kong52:40 Focus of future research53:55 Devices for research56:00 Bryan Johnson Blueprint59:20 People to follow01:00:00 place to stay up to date01:01:30 Thanks for all the fish longevity #aging #agingresearch

Learning With Lowell
Dr. David Katz: Lifespan Clock, Bodies Destroying Themselves, & Evolution Selecting for Aging – 250

Learning With Lowell

Play Episode Listen Later Aug 13, 2024 94:23


“My long-term goal is to model the pathways involved in human aging in order to develop therapeutic interventions. I organize an annual aging conference in the Midwestern United States with the dual goal of promoting the aging as an adaptation theory and promoting the aging as a disease mindset. The conference website is curing-aging.com.” Check out these other great views as well:Interview with Aubrey De Grey: https://youtu.be/DHvoPlcWwg4Interview with George Church: https://youtu.be/yNplpykf0B4Interview with Lisa Fabiny-Kiser: https://youtu.be/bE5jEGE5-OMInterview with Matt Kaeberlein: https://youtu.be/9QJ_ak3a05QInterview with Nir Barzilai : https://youtu.be/LuOAShcjOt0Interview with Michael Levin : https://youtu.be/5XvDdbYzwf8. PODCAST INFO:The Learning With Lowell show is a series for the everyday mammal. In this show we'll learn about leadership, science, and people building their change into the world. The goal is to dig deeply into people who most of us wouldn't normally ever get to hear. The Host of the show – Lowell Thompson- is a lifelong autodidact, serial problem solver, and founder of startups. LINKSSpotify: https://open.spotify.com/show/66eFLHQclKe5p3bMXsCTRHRSS: https://www.learningwithlowell.com/feed/podcast/Youtube: https://www.youtube.com/channel/UCzri06unR-lMXbl6sqWP_-QYoutube clips: https://www.youtube.com/channel/UC-B5x371AzTGgK-_q3U_KfAWebsite: https://www.learningwithlowell.com David Katz linkshttps://www.linkedin.com/in/davidwarrenkatz/https://www.youtube.com/watch?v=zhbOo_INMwg&t=65s&ab_channel=CuringAginghttps://www.curing-aging.com/ Timestamps00:00 start00:20 Programed aging hypothesis explained01:20 Michael Levin / bioelectricity02:15 Telomerase / concerns around copies of copies04:15 Ability for immortality in animals04:44 Immortal jellyfish06:02 Butterfly brains06:40 Programmed aging in practice for therapies07:16 FDA aging as a disease08:33 Curing aging objectionable09:15 We need to work against our bodies10:18 Fasting / Michael Rae / Caloric restriction11:55 Highly against Richard Dawkins selfish gene and ideas around evolution13:20 Start of slides13:50 Why do we age / David's argument18:20 Different types of aging23:20 Induce damage to test hypothesis25:10 David St Clair work26:20 Evolution has consistently selected for aging29:20 Aging and cancer30:00 exponential population growth31:00 Evolution as an ecological thing32:00 Animals getting bigger and smaller on small island / evolution as multivariable equation35:11 Mole Rats and rats36:25 people who get castrated live longer37:22 Bryan Johnson / interfering with body38:00 Testosterone / women live longer39:22 Mechanism of the body working against itself40:15 Easy interventions / keeping track of time for puberty, death, etc44:30 Squirrels having a year long internal clock46:00 Uniform life events / aging clock / evidence that convinces people50:50 ATF4 Pathway / muscle mass52:00 Not even trying to make therapies ATF454:40 Developing therapies against the body55:30 VCs at conference56:10 Exploring this hypothesis58:40 Tricking body to “think” it's younger59:10 Aubrey De Grey like hallmarks of aging clock1:00:00 Epigenetic clocks / biologic clocks01:01:30 Disrupting dreams / sleep01:02:50 Sleep affecting aging01:04:08 People who stop sleeping die within a number of weeks01:04:40 Does the biological clock have state and turning it back01:10:20 Steel man argument against the Programed aging hypothesis01:10:55 A good case for the clock / time of day, year, and stage of year01:13:00 Dolphins and sleep01:13:30 Target therapy with this theory01:15:40 Children's hospital01:16:05 Kids better able to fight off cancer / medicine divided01:17:11 PhD Tensor decomposition presentation / fundamental relationship of matrices01:18:40 implications and pushback for the tensor decomposition01:19:30 What are the trade offs / stop measuring algorithms based off complexity01:21:18 Real life example / chromosomes01:24:30 SENS and the human genome li...

Learning With Lowell
Kim Rahir: From Blind & Wheelchair Bound to Successful Olympic Weight Lifting & Powerlifting – 251

Learning With Lowell

Play Episode Listen Later Aug 13, 2024 85:49


Kim takes you to the next level of health and performance without the standard prescription of hours of cardio or chicken and broccoli.With laser focus on what has been proven to work for improved performance, sleep, cognitive function, metabolic health and stress levels, you will get maximum results with minimum effort. Work smarter, not harder to make your body look and perform the way you intend it to.The Fabulous in 15 approach is a minimalist protocol that takes you from stressed and tired to vibrant and energetic in the shortest possible amount of time. Check out these other great views as well:Interview with Aubrey De Grey: https://youtu.be/DHvoPlcWwg4Interview with George Church: https://youtu.be/yNplpykf0B4Interview with Lisa Fabiny-Kiser: https://youtu.be/bE5jEGE5-OMInterview with Matt Kaeberlein: https://youtu.be/9QJ_ak3a05QInterview with Nir Barzilai : https://youtu.be/LuOAShcjOt0Interview with Michael Levin : https://youtu.be/5XvDdbYzwf8. PODCAST INFO:The Learning With Lowell show is a series for the everyday mammal. In this show we'll learn about leadership, science, and people building their change into the world. The goal is to dig deeply into people who most of us wouldn't normally ever get to hear. The Host of the show – Lowell Thompson- is a lifelong autodidact, serial problem solver, and founder of startups. LINKSSpotify: https://open.spotify.com/show/66eFLHQclKe5p3bMXsCTRHRSS: https://www.learningwithlowell.com/feed/podcast/Youtube: https://www.youtube.com/channel/UCzri06unR-lMXbl6sqWP_-QYoutube clips: https://www.youtube.com/channel/UC-B5x371AzTGgK-_q3U_KfAWebsite: https://www.learningwithlowell.com Kim rahir linkshttps://kimrahir.com/https://www.linkedin.com/in/kim-rahir/?originalSubdomain=eshttps://www.instagram.com/kim.rahir/?hl=en Timestamps / chapters00:00 Start00:05 Kim Rahir00:50 Accomplishments after MS diagnosis / Olympic weight lifting02:04 Olympic weight lifting03:00 Deadlift04:30 Powerlifting07:55 Mike Tyson, Adversity and training10:10 Rejuvenating after training13:00 Getting through lack of motivations / 5 minute rule16:55 Only 5 minutes19:15 Lifelong learning24:20 Wheelchair to power lifting29:40 MS32:55 202434:55 Dream number36:55 Meditation / mindfulness39:00 Learn mediation / headspace41:45 Strength training43:40 Weeks until getting better45:55 Train to failure48:00 Health and energy focused49:30 Time intensity52:40 Diet / food!54:55 Family dinners and not over eating57:55 Celebrities / interesting meals01:01:45 Community01:05:10 Saying what you want01:08:55 how to get family on board01:11:30 Women vs men advantages and disadvantages01:13:10 Adversity01:14:30 Self Advocacy / MS diagnosis01:22:00 Post having kids01:23:55 One thing you can do right now01:24:55 Her Clients01:25:40 Thank you for all the fish weightlifting #multiplesclerosis #olympicweightlifting

Pharma and BioTech Daily
Pharma and Biotech Daily: Keeping You Informed on Industry Trends and Developments

Pharma and BioTech Daily

Play Episode Listen Later Jul 22, 2024 3:32


Good morning from Pharma and Biotech daily: the podcast that gives you only what's important to hear in Pharma and Biotech world.Mountain Dew introduced the Mountain Dude character in a new campaign called "Do the Dew" created by Goodby Silverstein & Partners. The campaign features a stylish brand character and encourages consumers to get active. Amazon released a back-to-school ad promoting savings, Heinz launched a campaign inspired by superheroes for their condiments, and Kraft Heinz named former Pepsi marketer Todd Kaplan as their North America CMO. Hershey also handed US media duties to Publicis. Industries are adopting the media network model to replicate the success of companies like Amazon and Walmart. Additionally, America's most trusted brands like Nvidia, Sony, and Adidas are discussed in a sponsored content piece. Other trending topics include Havas agencies losing B Corp status, Google accused of misleading consumers, the WNBA securing lucrative media rights deals, and Meta in talks to buy a stake in eyewear giant EssilorLuxottica.Transitioning to the world of biotech, Revolution Medicines is advancing its cancer drug, a ras inhibitor, to phase 3 trials after demonstrating tumor reduction in pancreatic cancer patients. The drug showed promising results but also had high rates of side effects like rash and nausea. Roche's obesity pill showed significant weight loss in a small study, while Gilead's Chief Medical Officer is set to depart next year. The biotech industry in 2024 is seeing progress in gene editing, mRNA, and cell therapies, offering optimism for the future of medicine. Caribou is cutting its workforce, Sionna is exploring abandoned cystic fibrosis drugs from AbbVie, and the industry is focusing on successful commercialization strategies. Overall, the industry is evolving with new treatments and developments shaping the landscape of biotech and pharma.Shifting gears to healthcare news, the House Committee has urged the FDA to suspend the lab-developed test rule, citing concerns about potential alterations to the United States' laboratory testing infrastructure. Steward executives received significant compensation before the company declared bankruptcy, and UnitedHealth's cyberattack response costs are expected to exceed $2.3 billion this year. Senators have introduced bipartisan healthcare cybersecurity legislation, while a Chicago children's hospital faces class action lawsuits after a cyberattack. Branded calling is highlighted as a way to increase patient answer rates, and AI is showcased as a tool to address healthcare challenges. Overall, the healthcare industry is facing various challenges related to cybersecurity, financial pressures, and patient care.In the realm of pharmaceuticals, Boehringer Ingelheim has partnered with GoodRx to offer its Humira biosimilar at a 92% discount. This move is aimed at capitalizing on Humira's decreasing market share. Artiva Biotherapeutics has announced an upsized IPO of $167 million to support the development of its therapy for systemic lupus erythematosus. George Church's startup has raised $60 million for its investigational therapy for gout, while Aveo's Fotivda combination therapy did not meet its primary efficacy endpoint in a phase III study. Invitro Cell Research is also working on preventive and regenerative medicine to help people live healthier lives. Other news includes the FDA rejecting Orexo's opioid overdose drug and granting a third indication for Phathom's Voquezna. Pfizer's once-daily weight loss pill and ongoing disputes over CRISPR patents are also highlighted.That's all for today's episode of Pharma and Biotech daily. Stay informed and have a great day!

Nina’s Notes Podcast

Hi Friends,   It's Nina's Note #80!In honor of this milestone, I have something new for you today, a debut interview for the Nina's Notes Podcast. On this inaugural episode, I am thrilled to welcome Michael Geer, a visionary determined to redefine our approach to aging.

Night Science
George Church and shooting for the stars

Night Science

Play Episode Listen Later Apr 29, 2024 36:18


George Church, professor of genetics at Harvard Medical School, leads a large research group at the Wyss Institute for Biologically Inspired Engineering. A pioneer in the fields of personalized genomics and synthetic biology, he has co-founded over 50 biotech companies. In 2017, Time magazine named him one of the 100 most influential people in the world. In this conversation, we discuss the importance of embracing outliers and taking calculated risks – it's not about never failing, it's about failing a million times a day. As Yogi Berra said, "When you come to a fork in the road, take it!”  George argues that you can change the world as long as you don't care who gets the credit. He recommends shooting for the stars – maybe you'll hit the moon. This episode was supported by Research Theory (researchtheory.org). For more information on Night Science, visit https://www.biomedcentral.com/collections/night-science .

The Genetics Podcast
EP 132: From mammoths to genome editing, trailblazing genetics with Dr. George Church

The Genetics Podcast

Play Episode Listen Later Apr 25, 2024 47:41


In this episode, we are excited to welcome George Church, Professor at Harvard Medical School and MIT, and Co-founder of Eugit Therapeutics, Colossal Biosciences, Arrived AI, and eGenesis. He is also the Director of PersonalGenomes.org, the world's only open-access information hub for human genomic, environmental, and trait data, as well as an IARPA BRAIN Project and the NIH Center for Excellence in Genomic Science. If that weren't enough, George was named one of the 100 most influential people in the world in Time Magazine's 2017 Time 100 list. In 2022, he was featured among the most influential people in biopharma by FiercePharma, where he was listed among the top 8 most famous geneticists in human history. All to say, you won't want to miss this episode with a genetics superstar!

FYI - For Your Innovation
George Church: Decoding the Future of Genomics

FYI - For Your Innovation

Play Episode Listen Later Apr 11, 2024 30:06


On this episode of FYI, ARK Multiomics analyst Nemo Marjanovic sits down with Professor George Church, a renowned geneticist and entrepreneur from Harvard. They discuss the evolution of genomic sequencing technologies, the intersection of genomics and blockchain for privacy, and the fascinating potential of DNA for data storage. They delve into multiomic approaches for enhanced biological understanding, the transformative impact of artificial intelligence (AI) in medical advancements, and George's groundbreaking research on aging. Key Points From This Episode:Evolution of genomic sequencing: from short to long readsBalancing privacy with blockchain in genomicsThe potential of DNA as a universal data storage mediumAdvancements and challenges in writing DNA for data encodingThe growing importance and integration of multiomic approachesHow AI and high-throughput screening revolutionize drug discoveryInsights into startups from George's lab leveraging machine learning and multiomicsRevolutionary research on aging: targeting multiple pathways simultaneouslyThe concept of protective gene therapies against all virusesEmerging technologies in genome synthesis and developmental biology

FYI - For Your Innovation
Resurrecting the Woolly Mammoth: CEO Ben Lamm's Colossal Quest

FYI - For Your Innovation

Play Episode Listen Later Feb 29, 2024 47:36


In this episode of For Your Innovation, Ali Urman chats with Ben Lamm, CEO of Colossal, about the groundbreaking venture aimed at resurrecting extinct species, starting with the woolly mammoth. Lamm shares insights into his journey from tech to biotech, highlighting the fusion of artificial intelligence (AI), automation, and genetic editing to challenge the boundaries of science and conservation. He delves into the ecological and scientific ramifications of bringing back extinct species and addresses the technological, regulatory, and ethical challenges faced along the way. Join us as we explore how Colossal is not just redefining de-extinction but also contributing significantly to conservation efforts and genetic research."I don't have a background in biology. My background is in finance and accounting. And then I've always been passionate about starting businesses. And I've always just been massively curious. And it's funny, my massive curiosity kind of led me to the mammoth" -@federallammKey Points From This Episode:Ben Lamm discusses transitioning from tech entrepreneurship to biotech with ColossalColossal aims to bring the woolly mammoth back from extinction, inspired by George ChurchConvergence of AI, automation, and biotech in de-extinction and conservation effortsFormBio: A spin-off providing AI tools for cell and gene therapy, aiding in biotech researchThe role of computational biology and gene editing in reconstructing extinct speciesThe potential ecological benefits of reintroducing extinct species like the woolly mammothColossal's involvement in conservation efforts and genetic rescue, beyond de-extinctionChallenges and advancements in xenotransplantation and disease resistance in wildlifeRegulatory considerations for biotech innovations in de-extinction and gene editingLamm's admiration for innovative minds like George Church, breaking barriers in science

Papa Phd Podcast
La bourse Pre-Amp avec Virginie Petel-Légaré et Michael Mee d’Amplitude Ventures

Papa Phd Podcast

Play Episode Listen Later Feb 15, 2024 48:04


Bienvenue à ce nouvel épisode d'Au-delà de la thèse avec Papa PhD en collaboration avec la bourse Pre-Amp d'Amplitude Ventures !Cette semaine, je parle avec Michael Mee et Virginie Petel-Légaré de la bourse Pre-Amp, un programme d'été qui transporte les jeunes scientifiques ua doctorat dans l'univers de la création d'entreprises et du capital de risque. Michael Mee est directeur chez Amplitude Ventures, le principal fonds de capital-risque du Canada dans le domaine de la biotechnologie et de la santé. Après avoir obtenu son doctorat en ingénierie biomédicale dans le laboratoire de George Church à HMS, Michael a travaillé chez Flagship Pioneering dans le domaine de la création d'entreprises. Chez Flagship, il s'est concentré sur le développement de deux entreprises de microbiome dans les domaines agricole (Indigo) et thérapeutique (Kaleido), sur la cofondation et le lancement d'une nouvelle plateforme thérapeutique et d'une entreprise de thérapie génique (Cobalt Biomedicine/Sana Therapeutics) et, plus récemment, sur le développement de nouvelles entreprises dans les domaines de la mise au point de médicaments basés sur l'IA et de l'édition de gènes. L'un de ses objectifs à long terme a toujours été de ramener chez lui les enseignements tirés de Boston afin de contribuer à l'écosystème biotechnologique canadien. À cette fin, il a récemment déménagé à Montréal et se réjouit de pouvoir atteindre cet objectif avec l'équipe grandissante d'Amplitude et de Pre-Amp.Virginie Petel-Légaré est actuellement Venture Associate chez Pre-Amp. Elle a fait partie de la première cohorte de la bourse Pre-Amp en 2021 et travaille avec Amplitude depuis, d'abord en tant que consultante, puis en tant qu'employée à temps plein. Virginie a terminé son baccalauréat en physiologie à l'Université McGill et a décidé de se concentrer sur les neurosciences. Elle a obtenu une maîtrise en neurosciences de l'Université de Montréal, où elle a étudié les déficits de la mémoire de travail en relation avec les modifications anatomiques de la connectivité pariéto-frontale. Virginie a terminé son doctorat à l'Institut neurologique de Montréal (Université McGill) en se concentrant sur le rôle du dysfonctionnement mitochondrial dans la SLA et la maladie de Parkinson. Ce que tu apprendras dans cette entrevue : Comment la bourse Pre-Amp se distingue par son focus sur l'apprentissage par l'expérience tout et comment elle offre un tremplin vers des carrières diversifiées hors de l'universitéComment le fait de travailler sur des projets hors de ton champ de compétence habituel peut débloquer ta créativité et contribuer à l'innovation.Comment tes compétences académiques peuvent être transférables et précieuses dans le secteur entrepreneurial.Et bien plus.Bonne écoute !La bourse Pre-Amp t'intéresse ? Envoie ton CV et ta lettre de motivation à > cette adresse courriel < - tu as jusqu'au 31 mars 2024 pour postuler ! Les ressources de cet épisode : Bourse Pre-Amp | Page WebAmplitude Ventures | Site Web Tu aimeras aussi ces épisodes : Marie Itoiz – Se préparer à l'après-doctorat : PapaPhD.com/marie-itoiz-adtAlexandre Bran - Le doctorat, une expérience transformatrice : PapaPhD.com/260Rémi Quirion – Projeter les jeunes chercheur.e.s dans des carrières d'avenir : PapaPhD.com/203Nathalie Ouellette – L'astrophysique à la portée de tous : PapaPhD.com/nathalie-ouellette/

Wake Up!
Wake Up! Monday, January 22, 2024

Wake Up!

Play Episode Listen Later Jan 22, 2024 45:40


We're live with Deacon Terry Bellon, Deacon at St. George Church in Baton Rouge talks about why men should attend a Catholic retreat. Alan Migliorato, co-author of Failing Forward: Leadership Lessons for Catholic Teens Today joins us and Jordan and Lisa Tabor, Catholic business people and founders of Rain Will Bring Flowers Foundation, talk about the event Planting Seeds of Hope focusing on suicide prevention.

The Colin McEnroe Show
Back from the dead: Exploring the cutting edge of de-extinction

The Colin McEnroe Show

Play Episode Listen Later Dec 21, 2023 49:00


At the beginning of the year, the company behind the public effort to de-extinct the wooly mammoth announced it will also be de-extincting the dodo. The announcement stirred up a lot of excitement and questions about whether we can – or should – bring back species once they're gone. So this hour we're talking about de-extinction! We'll hear about what it takes to bring back extinct animals, efforts to build a safety net for plants that might go extinct in the future, and walk through some fun de-extinction thought experiments. GUESTS: Helen Pilcher: Science and comedy writer with a PhD in cell biology who wrote Bring Back the King: The New Science of De-extinction Ben Lamm: CEO of the de-extinction company Colossal, which he co-founded with George Church. Carlos de la Rosa: President and CEO of the Center for Plant Conservation Join the conversation on Facebook and Twitter. The Colin McEnroe Show is available as a podcast on Apple Podcasts, Spotify, Google Podcasts, Stitcher, or wherever you get your podcasts. Subscribe and never miss an episode! Subscribe to The Noseletter, an email compendium of merriment, secrets, and ancient wisdom brought to you by The Colin McEnroe Show. Colin McEnroe and Cat Pastor contributed to this show, which originally aired on May 18, 2023.Support the show: http://www.wnpr.org/donateSee omnystudio.com/listener for privacy information.

Ground Truths
David Liu: A Master Class on the Future of Genome Editing

Ground Truths

Play Episode Listen Later Dec 10, 2023 47:52


David Liu is an gifted molecular biologist and chemist who has pioneered major refinements in how we are and will be doing genome editing in the future, validating the methods in multiple experimental models, and establishing multiple companies to accelerate their progress.The interview that follows here highlights why those refinements beyond the CRISPR Cas9 nuclease (used for sickle cell disease) are vital, how we can achieve better delivery of editing packages into cells, ethical dilemmas, and a future of somatic (body) cell genome editing that is in some ways is up to our imagination, because of its breadth, over the many years ahead. Recorded 29 November 2023 (knowing the FDA approval for sickle cell disease was imminent)Annotated with figures, external links to promote understanding, highlights in bold or italics, along with audio links (underlined)Eric Topol (00:11):Hello, this is Eric Topol with Ground Truths and I'm so thrilled to have David Liu with me today from the Broad Institute, Harvard, and an HHMI Investigator. David was here visiting at Scripps Research in the spring, gave an incredible talk which I'll put a link to. We're not going to try to go over all that stuff today, but what a time to be able to get to talk with you about what's happening, David. So welcome.David Liu (00:36):Thank you, and I'm honored to be here.Eric Topol (00:39):Well, the recent UK approval (November 16, 2023) of the first genome editing after all the years that you put into this, along with many other colleagues around the world, is pretty extraordinary. Maybe you can just give us a sense of that threshold that's crossed with the sickle cell and beta thalassemia also imminently [FDA approval granted for sickle-cell on 8 December 2023] likely to be getting that same approval here in the U.S.David Liu (01:05):Right? I mean, it is a huge moment for the field, for science, for medicine. And just to be clear and to give credit where credit is due, I had nothing to do with the discovery or development of CRISPR Cas9 as a therapeutic, which is what this initial gene editing CRISPR drug is. But of course, the field has built on the work of many scientists with respect to CRISPR Cas9, including Emmanuel Charpentier and Jennifer Doudna and George Church and Feng Zhang and many, many others. But it is, I think surprisingly rapid milestone in a long decade's old effort to begin to take some control over our genetic features by changing DNA sequences of our choosing into sequences that we believe will offer some therapeutic benefit. So this initial drug is the CRISPR Therapeutics /Vertex drug. Now we can say it's actually a drug approved drug, which is a Crispr Cas9 nuclease programmed to cut a DNA sequence that is involved in silencing fetal hemoglobin genes. And as you know, when you cut DNA, you primarily disrupt the sequence that you cut. And so if you disrupt the DNA sequence that is required for silencing your backup fetal hemoglobin genes, then they can reawaken and serve as a way to compensate for adult hemoglobin genes like the defective sickle cell alleles that sickle cell anemia patients have. And so that's the scientific basis of this initial drug.Eric Topol (03:12):So as you aptly put— frame this—this is an outgrowth of about a decade's work and it was using a somewhat constrained, rudimentary form of editing. And your work has taken this field considerably further with base and prime editing whereby you're not just making a double strand cut, you're doing nicks, and maybe you can help us understand this next phase where you have more ways you can intervene in the genome than was possible through the original Cas9 nucleases.David Liu (03:53):Right? So gene editing is actually a several decades old field. It just didn't quite become as popular as it is now until the discovery of CRISPR nucleases, which are just much easier to reprogram than the previous programmable zinc finger or tail nucleases, for example. So the first class of gene editing agents are all nuclease enzymes, meaning enzymes that take a piece of DNA chromosome and literally cut it breaking the DNA double helix and cutting the chromosome into two pieces. So when the cell sees that double strand DNA break, it responds by trying to get the broken ends of the chromosome back together. And we think that most of the time, maybe 90% of the time that end joining is perfect, it just regenerates the starting sequence. But if it regenerates the starting sequence perfectly and the nuclease is still around, then it can just cut the rejoin sequence again.(04:56):So this cycle of cutting and rejoining and cutting and rejoining continues over and over until the rejoining makes the mistake that changes the DNA sequence at the cut site because when those mistakes accumulate to a point that the nuclease no longer recognizes the altered sequence, then it's a dead end product. That's how you end up with these disrupted genes that result from cutting a target DNA sequence with a nuclease like Crispr Cas9. So Crispr Cas9 and other nucleases are very useful for disrupting genes, but one of their biggest downsides is in the cells that are most relevant to medicine, to human therapy like the cells that are in your body right now, you can't really control the sequence of DNA that comes out of this process when you cut a DNA double helix inside of a human cell and allow this cutting and rejoining process to take place over and over again until you get these mistakes.(06:03):Those mistakes are generally mixtures of insertions and deletions that we can't control. They are usually disruptive to a gene. So that can be very useful when you're trying to disrupt the function of a gene like the genes that are involved in silencing fetal hemoglobin. But if you want to precisely fix a mutation that causes a genetic disease and convert it, for example, back into a healthy DNA sequence, that's very hard to do in a patient using DNA cutting scissors because the scissors themselves of course don't include any information that allows you to control what sequence comes out of that repair process. You can add a DNA template to this cutting process in a process called HDR or Homology Directed Repair (figure below from the Wang and Doudna 10-year Science review), and sometimes that template will end up replacing the DNA sequence around the cut site. But unfortunately, we now know that that HDR process is very inefficient in most of the types of cells that are relevant for human therapy.(07:12):And that explains why if you look at the 50 plus nuclease gene editing clinical trials that are underway or have taken place, all but one use nucleases for gene disruption rather than for gene correction. And so that's really what inspired us to develop base editing in 2016 and then prime editing in 2019. These are methods that allow you to change a DNA sequence of your choosing into a different sequence of your choosing, where you get to specify the sequence that comes out of the editing process. And that means you can, for the first time in a general way, programmable change a DNA sequence, a mutation that causes a genetic disease, for example, into a healthy sequence back into the normal, the so-called wild type sequence, for example. So base editors work by actually performing chemistry on an individual DNA base, rearranging the atoms of that base to become a different base.(08:22):So base editors can efficiently and robustly change A's into G's G's, into A's T's into C's or C's into T's. Those four changes. And those four changes for interesting biochemical reasons turn out to be four of the most common ways that our DNA mutates to cause disease. So base editors can be used and have been used in animals and now in six clinical trials to treat a wide variety of diseases, high cholesterol and sickle cell disease, and T-cell leukemia for example. And then in prime editors we developed a few years later to try to address the types of changes in our genomes that caused genetic disease that can't be fixed with a base editor, for example. You can't use a base editor to efficiently and selectively change an A into a T. You can't use a base editor to perform an insertion of missing DNA letters like the three missing letters, CTT, that's the most common cause of cystic fibrosis accounting for maybe 70% of cystic fibrosis patients.(09:42):You can't use a base editor to insert missing DNA letters like the missing TATC. That is the most common cause of Tay-Sachs disease. So we develop prime editors as a third gene editing technology to complement nucleases and base editors. And prime editors work by yet another mechanism. They don't, again, they don't cut the DNA double helix, at least they don't cause that as the required mechanism of editing. They don't perform chemistry on an individual base. Instead, prime editors take a target DNA sequence and then write a new DNA sequence onto the end of one of the DNA strands and then sort of help the cell navigate the DNA repair processes to have that newly written DNA sequence replace the original DNA sequence. And in the process it's sort of true search and replace gene editing. So you can basically take any DNA sequence of up to now hundreds of base pairs and replace it with any other sequence of your choosing of up to hundreds of base pairs. And if you integrate prime editing with other enzymes like recombinase, you can actually perform whole gene integration of five or 10,000 base pairs, for example, this way. So prime editing's hallmark is really its versatility. And even though it's the newest of the three ways that have been robustly used to edit mammalian cells and rescue animal models of genetic disease, it is arguably the most versatile by far,Eric Topol (11:24):Right? Well, in fact, if you just go back to the sickle cell story as you laid out the Cas9 nuclease, that's now going into commercial approval in the UK and the US, it's more of a blunt instrument of disruption. It's indirect. It's not getting to the actual genomic defect, whereas you can do that now with these more refined tools, these new, and I think that's a very important step forward. And that is one part of some major contributions you've made. Of course, there are many. One of the things, of course, that's been a challenge in the field is delivery whereby we'd like to get this editing done in many parts of the body. And of course it's easy, perhaps I put that in quotes, easy when you're taking blood out and you're going to edit those cells and them put it back in. But when you want to edit the liver or the heart or the brain, it gets more challenging. Now, you did touch on one recent report, and this is of course the people with severe familial hypercholesterolemia. The carriers that have LDL cholesterol several hundred and often don't respond to even everything we have on the shelf today. And there were 10 people with this condition that was reported just a few weeks ago. So that's a big step forward.David Liu (13:09):That was also a very exciting milestone. So that clinical trial was led by scientists at Verve Therapeutics and Beam Therapeutics, and it was the first clinical readout of an in vivo base editing clinical trial. There was previously at the end of 2022, the first clinical readout of an ex vivo base editing clinical trial using CAR T cells, ex vivo  base edited to treat T-cell leukemia in pediatric patients in the UK. Ffigure from that NEJM paper below). But as you point out, there are only a small fraction of the full range of diseases that we'd like to treat with gene editing and the types of cells we'd like to edit that can be edited outside of the body and then transplanted back into the body. So-called ex vivo editing. Basically, you can do this with cells of some kind of blood lineage, hematopoietic stem cells, T-cells, and really not much else in terms of editing outside the body and then putting back into the body as you point out.(14:17):No one's going to do that with the brain or the heart anytime soon. So what was very exciting about the Verve Beam clinical trial is that Verve sought to disrupt the function of PCSK9 storied, gene validated by human genetics, because there are humans that naturally have mutations in PCSK9, and they tend to have much lower incidences of heart disease because their LDL, so-called bad cholesterol, is much lower than it would otherwise be without those mutations. So Verve set out to simply disrupt PCSK9 through gene editing. They didn't care whether they used a nuclease or a base editor. So they compared side-by-side the results of disrupting PCSK9 with Cas9 nuclease versus disrupting it by installing a precise single letter base edit using an adenine base editor. And they actually concluded that the base editor gave them higher efficacy and fewer unwanted consequences.(15:28):And so they went with the base editor. So the clinical trial that just read out were patients treated in New Zealand, in which they were given a lipid nanoparticle mRNA complex of an adenine base editor programmed with a guide RNA to install a specific A to G mutation in a splice site in PCSK9 that inactivates the gene so that it can no longer make functional PCSK9 protein. And the exciting result that read out was that in patients that receive this base editor, a single intravenous injection of the base editor lipid nanoparticle complex, as you know, lipid nanoparticles very efficiently go to the liver. In most cases, PCSK9 was edited in the liver and the result was substantial reduction in LDL cholesterol levels in these patients. And the hope and the anticipation is that that one-time treatment should be durable, should be more or less permanent in these patients. And I think while the patients who are at highest risk of coronary artery disease because of their genetics that give them absurdly high LDL cholesterol levels, that makes the most sense to go after those patients first because they are at extremely high risk of heart attacks and strokes. If the treatment proves to be efficacious and safe, then I think it's tempting to speculate that a larger and larger population of people who would benefit from having lower LDL cholesterol levels, which is probably most people, that they would also be candidates for this kind of therapy.Eric Topol (17:22):Yeah, no, it's actually pretty striking how that could be achieved. And I know in the primates that were done prior to the people in New Zealand, there was a very durable effect that went on well over I think a year or even two years. So yeah, that's right. Really promising. So now that gets us to a couple of things. One of them is the potential for off-target effects. As you've gotten more and more with these tools to be so precise, is the concern that you could have off-target effects just completely, of course inadvertent, but potential for other downstream in time known unknowns, if you will. What are your thoughts about that?David Liu (18:15):Yeah, I have many thoughts on this issue. It's very important the FDA and regulatory bodies are right to be very conservative about off-target editing because we anticipate those off targets will be permanent, those off-target edits will be permanent. And so we definitely have a responsibility to minimize adding to the mutational burden that all humans have as a function of existing on this planet, eating what we eat, being bombarded by cosmic rays and sunlight and everything else. But I think it's also important to put off-target editing into some context. One context is I think virtually every substance we've ever put into a person, including just about every medicine we've ever put into a person, has off-target effects, meaning modulates the function of biological molecules other than the intended target. Of course, the stakes are higher when those are gene editing agents because those modifications can be permanent.(19:18):I think most off-target edits are very likely to have no consequence because most of our genome, if you mutate in the kinds of small ways like making an individual base pair change for a base editor are likely to have no consequence. We sort of already know this because we can measure the mutational burden that we all face as a function of living and it's measurable, it's low, but measurable. I've read some papers that estimate that of the roughly 27 trillion [should be ~37] cells in an adult person, that there are billions and possibly hundreds of billions of mutations that accumulate every day in those 27 [37] trillion cells. So our genomes are not quite the static vaults that we'd like to think that they are. And of course, we have already purposefully given life extending medicines to patients that work primarily by randomly mutating their genomes. These are chemotherapeutic agents that we give to cancer patients.(20:24):So I think that history of giving chemotherapeutic agents, even though we know those agents will mess up the genomes of these patients and potentially cause cancer far later down the road, demonstrates that there are risk benefit situations where the calculus favors treatment, even if you know you are causing mutations in the genome, if the condition that the patient faces and their prognosis is sufficiently grave. All that said, as I mentioned, we don't want to add to the mutational burden of these patients in any clinically relevant way. So I think it is appropriate that the early gene editing clinical candidates that are in trials or approved now are undergoing lots and lots of scrutiny. Of course, doing an off-target analysis in an animal is of limited value because the animal's genome is quite different than the human genome. So the off targets won't align, but doing off-target analysis in human cells and then following up these patients for a long time to confirm hopefully that there isn't clinical evidence of quality of life or lifespan deterioration caused by off-target editing, that's all very, very important.(21:55):I also think that people may not fully appreciate that on target editing consequences also need to be examined and arguably examined with even more urgency than off-target edits. Because when you are cutting a chromosome at a target site with a nucleus, for example, you generate a complex mixture of different products of different DNA sequences that come out, and the more sequences you sequence, the more different products you realize are generated. And I don't think it's become routine to try to force the companies, the clinical groups that are running these trials to characterize the top 1000 on target products for their biological consequence. That would be sort of impractical to do and would probably slow down greatly the benefit of these early nuclease clinical trials for patients. But those are actually the products that are generated with much higher frequency typically than the off-target edits. And that's part of why I think it makes more sense from a clinical safety perspective to use more precise gene editing methods like base editing and prime editing where we know the products that are generated are mostly the products that we want are not uncontrolled mixtures of different deletion and insertion products.(23:27):So I think paying special attention to the on-target products, which are generated typically 70 to 100% of the time as opposed to the off targets which may be generated at a 0.1 to 1% level and usually not that many at that level once it reaches a clinical candidate. I think that's all important to do.Eric Topol (23:51):You've made a lot of great points there and thanks for putting that in perspective. Well, let's go on to the delivery issue. You mentioned nanoparticles, viral vectors, and then you've come up with small virus-like neutered viruses if you will. I think a company Nvelop that you've created to push on that potential. What are your thoughts about where we stand since you've become a force for coming up with much better editing, how about much better and more diverse delivery throughout the body? What are your thoughts about that?David Liu (24:37):Yeah, great. Great question. I think one of the legacies of gene editing is and will be that it inspired many more scientists to work hard on macromolecular delivery technologies. All of these gene editing agents are macromolecules, meaning they're proteins and or nucleic acids. None of them are small molecules that you can just pop a pill and swallow. So they all require special technologies to transfer the gene editing agent from outside of the cell into the cell. And the fact that taking control of our genetic features has become such a popular aspiration of medicine means that there's a lot of scientists as measured, most importantly by the young scientists, by the graduate students and the postdocs and the young professors of which I'm no longer one sadly, who have decided that they're going to devote a big part of their program to delivery. So you summarized many of the clinically relevant, clinically validated delivery technologies already, somewhat sadly, because if there were a hundred of these technologies, you probably wouldn't need to ask this question. But we have lipid nanoparticles that are particularly good at delivering messenger RNA, that was used to deliver the covid vaccine into billions of people. Now also used to deliver, for example, the adenine base editor mRNA into the livers of those hypercholesterolemia patients in the Verve/Beam clinical trial.(26:20):So those lipid nanoparticles are very well matched for gene editing delivery as long as it's liver. And they also are particularly well matched because their effect is transient. They cause a burst of gene editing agents to be produced in the liver and then they go away. The gene editing agents can't persist, they can't integrate into the genome despite what some conspiracy theorists might worry about. Not that you've had any encounter with any of those people. I'm sure that's actually what you want for a gene editing agent. You ideally want a delivery method that exposes the cell only for the shortest amount of time needed to make the on-target edit at the desired level. And then you want the gene editing agent to disappear and never come back because it shouldn't need to. DNA edits to our genome for durable cells should be permanent. So that's one method.(27:25):And then there are a variety of other methods that researchers have used to deliver to other cells, but they each carry some trade-offs. So if you're trying to edit hematopoietic stem cells, you can take them out of the body. Once they're out of the body, you have many more methods you can use to deliver efficiently into them. You can electroporated messenger, RNA or even ribonuclear proteins. You can treat with lipids or viruses, you can edit and then put them back into the body. But as you already mentioned, that's sort of a unique feature of blood cells that isn't applicable to the heart or the brain, for example, or the eyes. So then that brings us to viral vectors. There are a variety of clinically validated viral methods for delivery. AAV— adeno associated virus— is probably the most diverse, most relevant, and one of the best tolerated viral delivery methods. The beauty of AAV is that it can deliver to a variety of tissues. AAV can deliver into spinal cord neurons, for example, into retinal cells, into the heart, into the liver, into a few other tissues as well.(28:48):And that diversity of being able to choose AAV capsids that are known to get into the types of tissues that you're trying to target is a great strength of that approach. One of the downsides of AAV for gene editing agents is that their delivery tends to be fairly durable. You can engineer AAVs into next generation capsids that sort of get rid of themselves or the gene editing agents get rid of themselves. But classic AAV tends to stay around in patients for a long time, at least months, for example, and possibly years. And we also don't yet have a good way, clinically validated way of re-dosing AAV. And once you administer high doses of AAV in a patient that tends to provoke high-titer, neutralizing antibodies against those AAVs making it difficult to then come back six months or a year later and dose again with an AAV.(29:57):So researchers are on the bright side, have become very good at engineering and evolving in the laboratory next generation AAVs that can go to greater diversity issues that can be more potent. Potency is important because if you can back off the dose, maybe you can get around some of these immunogenicity issues. And I think we will see a renaissance with AAV that will further broaden its clinical scope. Even though I appreciate that the decisions by a couple large pharma companies to sort of pull out of using AAV for gene therapy seemed to cause people to, I think prematurely conclude that AAV has fallen out of favor. I think for gene therapy, it's quite different than gene editing. Gene therapy, meaning you are delivering a healthy copy of the gene, and you need to keep that healthy copy of the gene in the patient for the rest of the patient's life.(30:59):That's quite different than gene editing where you just need the edit to take place over days to weeks, and then you want the editing agent to actually go away and you never want to come back. I think AAV will used to deliver gene editing agents will avoid some of the clinical challenges like how do we redose? Because you shouldn't need to redose if the gene editing clinical trial proceeds as you hope. And then you mentioned these virus-like particles. So we became interested in virus-like particles as other labs have because they offer some of the best strengths of non-viral and viral approaches like non-viral approaches such as LMPs. They deliver the transient form of a gene editing agent. In fact, they can deliver the fully assembled protein RNA complex of a base editor or a prime editor or a CRISPR nuclease. So in its final form, and that means the exposure of the cell to the editing agent is minimized.(32:15):You can treat with these virus-like particles, deliver the protein form of these gene editing agents, allow the on-target site to get edited. And then since the half-life of these proteins tends to be very small, roughly 24 hours for example, by a week later, there should be very little of the material left in the animal or prospectively in the patient virus-like particles, as you call them, neutered viruses, they lack viral DNA or RNA. They don't have the ability to integrate a virus's genome into the human genome, which can cause some undesired consequences. They don't randomly introduce DNA into our genomes, therefore, and they disappear more transiently than viruses like AAV or adenoviruses or other kinds of lentiviruses that have been used in the clinic. So these virus-like particles or VLP offer really some of the best strengths on paper at least of both viral and non-viral delivery.(33:30):Their limitation thus far has been that there really haven't been examples of potent in vivo delivery of cargoes like gene editing agents using virus-like particles. And so we recently set out to figure out why, and we identified several bottlenecks, molecular bottlenecks that seemed to be standing in the way of virus-like particles, doing a much more efficient job at delivering inside of an animal. (Figure from that paper below.) And we engineered solutions to each of these first three molecular bottlenecks, and we've identified a couple more since. And that resulted in what we call VLPs engineered virus-like particles. And as you pointed out, Keith Joung and myself, co-founded a company called Nvelop to try to bring these technologies and other kinds of molecular delivery technologies, next generation delivery technologies to patients.Eric Topol (34:28):Well, that gets me to the near wrapping up, and that is the almost imagination you could use about where all this can go in the future. Recently, I spoke to a mutual friend Fyodor Urnov, who talked about wouldn't it be amazing if for people with chronic pain you could just genome edit neurons their spinal cord? As you already touched on recently, Jennifer Doudna, who we both know talked about editing to prevent Alzheimer's disease. Well, that may be a little far off in time, but at least people are talking about these things that is not, we're not talking about germline editing, we're just talking about somatic cell and being able to approach conditions that have previously been either unapproachable or of limited success and potential of curing. So this field continues to evolve and you and all your colleagues are a big part of how this has evolved as quickly as it has. What are your thoughts about, are there any bounds to the potential in the longer term for genome editing? Right.David Liu (35:42):It's a great question because all of the early uses of gene editing in people are appropriately focused on people who are at dire risk of having shorter lives or very poor quality of life as it should be for a new kind of therapeutic because the risks are high until we continue to validate the clinical benefit of these gene editing treatments. And therefore we want to choose patients the highest that face the poorest prognosis where the risk benefit ratio favors treatment as strongly as possible. But your question, I think very accurately highlights that our genome and changes to it determine far more than whether you have a serious genetic disorder like Sickle Cell Disease or Progeria or Cystic Fibrosis or Familial Hypercholesterolemia or Tay-Sachs disease. And being able to not just correct mutations that are associated with devastating genetic disorders, but perhaps take control of our genomes in more sophisticated way that you pointed out two examples that I think are very thought provoking to treat chronic pain permanently to lower the risk of horrible diseases that affect so many families devastating to economies worldwide as well, like Alzheimer's disease, Parkinson's disease, the genetic risk factors that are the strongest genetic determinants of diseases like Alzheimer's disease are actually, there are several that are known already.(37:36):And an interesting possibility for the future, it isn't going to happen in the next few years, but it might happen within the next 10 or 20 years, might be to use gene editing to precisely change some of those most grievous alleles that are risk factors for Alzheimer's disease like a apoE4, to change them to the genetic forms that have normal or even reduced risk for Alzheimer's disease. That's a very tough clinical trial to run, but I'd say not any tougher than the dozens of most predominantly failed Alzheimer's clinical trials that have probably collectively accounted for hundreds of billions of dollars of investmentEric Topol (38:28):Easily.David Liu (38:31):And all of that speaks to the fact that Alzheimer's disease, for example, is enormous burden on society by every measure. So it's worth investing and major resources and taking major risks to try to create perhaps preventative treatments that just lower our risk globally. Getting there will require that these pioneering early clinical trials for gene editing are smashing successes. I'm optimistic that they will be, there will be bumps in the road because there always are bumps in the road. There will be patients who have downturns in their health and everyone will wonder whether those patients had a downturn because of a gene editing treatment they received. And ascertaining whether that's the case will be very important. But as these trials continue to progress, and as they continue hopefully on this quite positive trajectory to date, it's tempting to imagine a future where we can use precise gene editing methods. For example, you can install a variety using prime editing, a variety of alleles that naturally occur in people that reduce the risk of Alzheimer's disease or Parkinson's disease like the mutation that 0.1% of Icelandic people and almost nobody else has in amyloid precursor protein changing alanine 673 to threonine (A673T).(40:09):It is very thought provoking, and I don't think society is ready now to take that step, but I think if things continue to proceed on this promising trajectory, it's inevitable because arguably, the defining trait of our species is that we use every ounce of our talents and our gifts and our resources and our creativity to try to improve our lives and those of our children. And I don't think if we have ways of treating genetic diseases or even of reducing grievous genetic disease risk, that we will be able to sit on our hands and not take steps towards that kind of future solon as those technologies continue to be validated in the clinic as being safe and efficacious. It's, I teach a gene editing class and I walk them through a slippery slope at the end of five ethics cases, starting with progeria, where most people would say having a single C of T mutation in one gene that you, by definition didn't inherit from mom or dad.(41:17):It just happened spontaneously. That gives you an average lifespan of 14 and a half years and strongly affects other aspects of the quality of your life and your family's life that if you can change as we did in animals that T back into a C and correct the disease and rescue many of the phenotypes and extend lifespan, that that's an ethical use of gene editing. Treating genetic deafness is the second case. It's a little bit more complicated because many people in the deaf community don't view deafness as a disability. It's at least a more subjective situation than progeria. But then there are other cases like changing apoE4 to apoE3 or even apoE2 with the lower than normal risk of Alzheimer's disease, or installing that Icelandic mutation and amyloid precursor protein that substantially lowers risk of Alzheimer's disease. And then finally, you can, I always provoke a healthy debate in the class at the end by pointing out that in the 1960s, one of the long distance cross country alpine skiing records was set by a man who had a naturally occurring mutation in his EPO receptor, his erythropoietin receptor, so that his body always thought he was on EPO as if he were dosing on EPO, although that was of course before the era of EPO dosing was really possible, but it was just a naturally occurring mutation in this case, in his family.(42:48):And when I first started teaching this class, most students could accept using gene editing to treat progeria, but very few were willing to go even past that, even to genetic deafness, certainly not to changing a ApoE risk factors for Alzheimer's. Nowadays, I'd say the 50% vote point is somewhere between case three and case four, most people are actually say, yeah, especially since they have family members who've been through Alzheimer's disease. If they are a apoE4, some of them are a apoE4/apoE4 [homozygotes], why not change that to a apoE3 or even an ApoE2 or as one student challenged the class this year, if you were born with a apoE2, would you want to change it to a ApoE3 so you could be more normal? Most people would say, no, there's no way I would do that.(43:49):And for the first time this year, there were one or two students who actually even defended the idea of putting in a mutation in erythropoietin receptor to increased increase their endurance under low oxygen conditions. Of course, it's also presumably useful if you ever, God forbid, are treated with a cancer chemotherapeutic. Normally you get erythropoietin to try to restore some, treat some of the anemia that can result, and this student was making a case, well, why wouldn't we? If this is a naturally occurring mutation that's been shown to benefit certain people doing certain things. I don't think that's a general societal view. And I am a little bit skeptical we'll ever get widespread acceptance of case number five. But I think all of it is healthy stimulates a healthy discussion around the surprisingly gentle continuum between disease treatment, disease prevention, and what some would call human improvement.And it used to be that even the word human improvement was sort of an anathema. I think now at least the students in my class are starting to rethink what does that really mean? We improving ourselves a number of ways genetically and otherwise by virtue of our lifestyles, by virtue of who we choose to procreate with. So it's a really interesting debate, and I think the rapid development and now clinical progression and now approval, regulatory approval of gene editing drugs will play a central role in this discussion.Eric Topol (45:38):No question. I mean, also just to touch on the switch from a apoE4 to apoE2, you would get a potential 2-fer of lesser risk for Alzheimer's and a longer lifespan. So I mean, there's a lot of things here. The thing that got me years ago, I mean, this is many years ago at a meeting with George Church and he says, we're going to just edit 60 genes and then we can do all sorts of xeno-pig transplants and forget the problem of donors. And it's happening now.David Liu (46:11):Yeah, I mean, he used a base editor to edit hundreds of genes at once, if not thousands ofEric Topol (46:16):That's why it's just, yeah, no, it's just extraordinary. And I think people need to be aware that opportunities here, as you say, with potential bumps along the way, unquestionably, is almost limitless. So this has been a masterclass thanks to you, David, in where we are, where we're headed in genome editing at a very extraordinary time where we've really seeing things click. And I just want to also add that you're going to be here with a conference in La Jolla in January, I think, on base and prime editing. Is that right? So for those who are listeners who are into this topic, maybe they can also hear the latest, I'm sure there'll be more between now and next. Well, several weeks from now at your, it's aDavid Liu (47:12):Conference on, it's the fifth international conference on base and prime editing and associated enzymes, the somewhat baroque name. And I will at least be giving a virtual talk there. It actually overlaps with the talk I'm giving at Rockefeller that time. Ah, okay, cool. But I'm speaking at the conference either in person or virtually.Eric Topol (47:34):Yeah. Well, anytime we get to hear from you and the field, of course it's enlightening. So thanks so much for joining. Thank youDavid Liu (47:42):For having me. And thank you also for all of your, I think, really important public service in connecting appropriately the ground truths about science and vaccines and other things to people. I think that's very much appreciated by scientists like myself.Eric Topol (48:00):Oh, thanks David.Thanks for listening, reading, and subscribing to Ground Truths. To be clear, this is a hybrid format, roughly alternating between analytical newsletters/essays and podcasts with exceptional people, attempting to achieve about 2 posts per week. It's all related to cutting-edge advances in life science, medicine, and information tech (A.I.)All content is free. If you wish to become a paid subscriber know that all proceeds go to Scripps Research. Get full access to Ground Truths at erictopol.substack.com/subscribe

Biotalk
Biotalk Episode 12: A Conversation with Soufiane Aboulhouda of Nucleate

Biotalk

Play Episode Listen Later Dec 4, 2023 32:27


On this episode of Biotalk, Locust Walk‘s Geoff Meyerson welcomes Soufiane Aboulhouda, Co-Founder and Chairman of the Board of Nucleate, a global non-profit dedicated to empowering future biotech leaders through education and fostering innovation in life sciences companies. The discussion explores Soufiane's journey into early-stage biotech and delves into the founding story of Nucleate, highlighting the organization's mission and its pivotal role in shaping the biotech landscape. Soufiane, who served as President of Nucleate for over 5 years, shares valuable lessons learned during his tenure. Listeners also gain insights into Soufiane's current work at the Wyss Institute under George Church, exploring the intricacies of his research in the lab. The episode concludes with Soufiane addressing the biggest challenges facing the biotech industry and Nucleate, expressing optimism for the organization's future. The positive note of the conversation emphasizes the reasons behind Soufiane ‘s confidence in the promising future of Nucleate. We invite you to tune in to this compelling discussion on biotech, education, and innovation. Subscribe or follow Biotalk on Apple Podcasts | Spotify.    Timestamps:   1:21: To kick things off, I'd love for you to provide our audience with some background as to what led you to your initial exploration of early-stage biotech?  5:15: What is the founding story of behind Nucleate? What you're trying to solve and how did it come about   8:56: You also served as President of Nucleate for over 5 years. What lessons have you learned throughout your time as president?  16:04: You currently work at the Wyss Institute under George Church. Tell us more about your work in that lab.   19:37: In terms of Nucleate as an organizations, where do you operate, how many members do you have?  22:58: How has the biotech market downturn impacted Nucleate? Your lab? Your research?  26:48: At Locust Walk, we believe that dislocations are a great way to spot opportunity.  What would you say are the kinds of opportunities that are revealing themselves to you as the industry goes through a little bit of a struggle?    28:54: To wrap up a great conversation, do you believe there is a biggest issue currently facing the industry, or potentially impacting Nucleate?  30:44: Why are you optimistic about the future of Nucleate? 

NEJM AI Grand Rounds
Machines & Genes: The Future of AI in Biology with Dr. George Church

NEJM AI Grand Rounds

Play Episode Listen Later Oct 25, 2023 49:48 Transcription Available


In this episode, vanguard geneticist Dr. George Church recounts his storied career from his early fascination with computers and science to his pioneering work in genomics and synthetic biology. Dr. Church developed innovative DNA sequencing methods that enabled the first sequencing of the human genome. He was also instrumental in developing CRISPR gene editing technology. Dr. Church discusses his controversial ideas around resurrecting wooly mammoths and using genome sequencing in dating apps to prevent genetic diseases. He also provides insights into founding genomics companies and the role of AI in advancing biotechnology. As a professor of genetics at Harvard Medical School and the founder of multiple genomics companies, Dr. Church emphasizes the potential for synthetic biology and genetics to transform medicine and society. Transcript.

Modern Healthspan
Immune System Rejuvenation : Path To The Clinic | Dr Stan Wang Ep2

Modern Healthspan

Play Episode Listen Later Oct 22, 2023 13:10


In this video Dr Wang talks about the current state of trials and how the therapy might eventually look. Stan Wang is Founder and CEO of Thymmune Therapeutics, a cell therapy company working to address high unmet needs across immunology. Previously, Stan was Founding Chief Scientific Officer at Cellino Biotech, where he led R&D and application of its technology to rapidly engineer cells. Stan received his Ph.D. from the University of Cambridge and his M.D. from Columbia University. He was a postdoctoral fellow in cell and gene therapy with George Church at Harvard Medical School.

Modern Healthspan
Promising Therapy To Restoring Our Immune System & Combat Aging | Dr Stan Wang Ep1

Modern Healthspan

Play Episode Listen Later Oct 19, 2023 22:40


In this episode Dr Wang explains about the thymus gland and his technology for not only restoring the thymus but building new thymuses from iPCSs at scale. Stan Wang is Founder and CEO of Thymmune Therapeutics, a cell therapy company working to address high unmet needs across immunology. Previously, Stan was Founding Chief Scientific Officer at Cellino Biotech, where he led R&D and application of its technology to rapidly engineer cells. Stan received his Ph.D. from the University of Cambridge and his M.D. from Columbia University. He was a postdoctoral fellow in cell and gene therapy with George Church at Harvard Medical School.

Really? no, Really?
Cloning Mammoths, Cultivating Organs + Giant Beavers

Really? no, Really?

Play Episode Listen Later Sep 19, 2023 39:04 Transcription Available


Jason and Peter heard that a biotech company claimed that the long-deceased woolly mammoth will be brought back from extinction by 2027…Really, no really! Are we on the cusp of a real-life Jurassic Park? With new technology available to de-extinct long dead animals, potentially change the genetic code of human babies and the possibility of creating synthetic life, we needed to get some clarity. We had to find and talk to the man leading the science…and that man is Dr. George Church, known as the "father of synthetic biology. He is the Professor of Genetics at Harvard Medical School and Professor of Health Sciences & Technology at Harvard and MIT. In 2021 Dr. Church and entrepreneur Ben Lamm founded Colossal Biosciences, a synthetic biology company tasked in part with sequencing the remains of woolly mammoths which would allow them, in theory to “de-extinct” the ancient creatures.   In this episode: How do you de-extinct a woolly mammoth? The dangers lurking beneath melting permafrost. Redefining death. Creating human organs inside pigs. Bringing back those cryogenically frozen. Cold resistant Asian Elephants. Cultured meat and mammoth meatballs The dodo & Tasmanian tiger are coming back too?     Follow Dr. Church: X (Twitter): @GeoChurch   Follow us: www.reallynoreally.com Instagram YouTube TikTok Facebook Threads X (Twitter)See omnystudio.com/listener for privacy information.

Little Curiosities With Kendall Long
Colossal Interview: Resurrecting the Mammoth

Little Curiosities With Kendall Long

Play Episode Listen Later Aug 22, 2023 29:03


What if you could bring back an animal that's gone extinct? Well, that's exactly what scientists are doing at Colossal Biosciences. On this episode of Little Curiosities, I sit down with Colossal CEO and Founder Ben Lamm and Head of Biological Sciences Eriona Hysolli, Ph.D. and hear about animals they are bringing back, where they plan to release them, and so much more. Let's talk about it shall we? ~~ Follow Kendall Long on Instagram: @itskendalllong Tik Tok: @itskendalllong ~~ ABOUT COLOSSAL BIOSCIENCES Colossal Biosciences was founded by emerging technology and software entrepreneur Ben Lamm and world-renowned geneticist and serial biotech entrepreneur George Church, Ph.D., and is the first to apply CRISPR technology for the purposes of species de-extinction. Colossal creates innovative technologies for species restoration, critically endangered species protection and the repopulation of critical ecosystems that support the continuation of life on Earth. Colossal is accepting humanity's duty to restore Earth to a healthier state, while also solving for the future economies and biological necessities of the human condition through cutting-edge science and technologies. To follow along, please visit: https://colossal.com/. ~~ COLOSSAL WEBSITE & SOCIALS Website: https://colossal.com Twitter/X: https://twitter.com/itiscolossal Instagram: https://www.instagram.com/itiscolossal/ LinkedIn: https://www.linkedin.com/company/itiscolossal/ Facebook: https://www.facebook.com/itiscolossal YouTube: https://www.youtube.com/@itiscolossal ~~ Little Curiosities With Kendall Long is brought to you by QCODE. To advertise on the show, contact us! Learn more about your ad choices. Visit megaphone.fm/adchoices

The Munk Debates Podcast
Be it Resolved, let's engineer a better human being

The Munk Debates Podcast

Play Episode Listen Later Aug 8, 2023 43:43


We've come a long way since DNA was first discovered in the mid 19th century. Today's scientists are using powerful engineering techniques to edit genes in human eggs and sperm, curing diseases and repairing defective genes before a child is even born. Some scientists are excited about these therapies, championing them as an exciting opportunity to create immunity to viruses, eliminate serious illnesses like AIDS, Alzheimer's, and cancer, and possibly reverse aging. Like prior innovations in medicine and technology, why wouldn't we embrace a science that allows people to live longer, healthier, and happier lives? Others are alarmed. They are worried that these new techniques raise a host of profound ethical issues. While eliminating genetic diseases is a worthwhile endeavor, many parents might be inclined to use this science to create designer babies: children who are smarter, taller, or have other supposedly desirable traits. And these tools aren't cheap. They will surely be available to the rich first, creating a terrifying new dimension to the growing economic inequality crisis. Scientists also point out that ‘playing god' and editing genes will alter our DNA code forever, and one mistake could inadvertently introduce new diseases into the human gene pool. While the desire to cure genetic diseases is a noble one, the manipulation of our DNA is more likely than not to push humanity towards a dangerous and dystopian future no one wants.  Arguing for the motion is George Church, Professor of Genetics at Harvard Medical School, Professor of Health Sciences and Technology at Harvard and MIT Arguing against the motion is Joyce Harper, Professor of Reproductive Science at the Institute for Women's Health, University College London.   Sources:  ABC News, France24, Today Show, NBC News, VICE, PBS, Gattaca, Critical Past The host of the Munk Debates is Rudyard Griffiths - @rudyardg.   Tweet your comments about this episode to @munkdebate or comment on our Facebook page https://www.facebook.com/munkdebates/ To sign up for a weekly email reminder for this podcast, send an email to podcast@munkdebates.com.   To support civil and substantive debate on the big questions of the day, consider becoming a Munk Member at https://munkdebates.com/membership Members receive access to our 10+ year library of great debates in HD video, a free Munk Debates book, newsletter and ticketing privileges at our live events. This podcast is a project of the Munk Debates, a Canadian charitable organization dedicated to fostering civil and substantive public dialogue - https://munkdebates.com/ The Munk Debates podcast is produced by Antica, Canada's largest private audio production company - https://www.anticaproductions.com/ Senior Producer: Ricki Gurwitz Editor: Reza Dahya  

What I Know
Flashback: Ben Lamm of Colossal: Value Your Critics

What I Know

Play Episode Listen Later Jul 31, 2023 48:44


By the time he teamed up with Harvard geneticist George Church to found Colossal Biosciences, Ben Lamm had founded, built, and sold five companies. This one would be the most audacious yet: Its goal is to create disruptive conservation technologies, including, to de-extinct the woolly mammoth. Yes, it is actively working to edit elephant genes to create a cold-hearty herbivore to help decelerate melting of the arctic permafrost, and, thus, prevent release of 600 tons of carbon a year. It's also working with existing species-conservation efforts globally–and hopes to apply its technology to save animal populations from going extinct. But with the audacious mission comes a lot of questions–and many critics. Lamm told host Christine Lagorio-Chafkin that he learns more from his detractors than from his supporters–and he welcomes both hearing from them, and, in a couple cases, he's actually hired them to work with him.

Faster, Please! — The Podcast

On Faster, Please! — The Podcast, I've interviewed guests on exciting new technologies like artificial intelligence, fusion energy, and reusable rockets. But today's episode explores another Next Big Thing: biotechnology. To discuss recent advances in CRISPR gene editing and their applications for medicine, I'm sitting down with Kevin Davies.Kevin is executive editor of The CRISPR Journal and author of the excellent 2020 book, Editing Humanity: The CRISPR Revolution and the New Era of Genome Editing.In This Episode* CRISPR advances over the past decade (1:13)* What CRISPR therapies will come next? (8:46)* Non-medical applications of gene editing (13:11)* Bioweapons and the ethics of CRISPR (18:43)* Longevity and genetic enhancements (25:48)Faster, Please! is a reader-supported publication. To receive new posts and support my work, consider becoming a free or paid subscriber.Below is an edited transcript of our conversationCRISPR advances over the past decadeWhen people talk about AI, for instance, they might be talking about different versions or applications of AI—machine learning being one. So when we talk about CRISPR, are we just talking about one technique, the one they figured out back in 2012? Are there different ones? Are there improvements? So it's really a different technique. So how has that progressed?You're right. CRISPR has become shorthand for genome editing. But the version of CRISPR that was recognized with the Nobel Prize three years ago in 2020 to Jennifer Doudna and Emmanuelle Charpentier was for one, we can call it the traditional form of CRISPR. And if I refer to it again, I'll call it CRISPR-Cas9. Cas9 is the shorthand name for the enzyme that actually does the cutting of the DNA. But we are seeing extraordinary progress in developing new and even more precise and more nuanced forms of genome editing. They still kind of have a CRISPR backbone. They still utilize some of the same molecular components as the Nobel Prize–winning form of CRISPR. But in particular, I'm thinking of techniques called base editing and prime editing, both of which have commercial, publicly funded biotech companies pushing these technologies into the clinic. And I think over the next five to 10 years, increasingly what we refer to as “CRISPR genome editing” will be in the form of these sort of CRISPR 2.0 technologies, because they give us a much broader portfolio of DNA substitutions and changes and edits, and give the investigators and the clinicians much more precision and much more subtlety and hopefully even more safety and more guarantees of clinical efficiency.Right. That's what I was going to ask. One advantage is the precision, because you don't want to do it wrong. You don't want mutations. Do no harm first. A big advantage is maybe limiting some of the potential downsides.In the ideal gene-editing scenario, you would have a patient with, say, a genetic disease that you can pinpoint to a single letter of the genetic code. And we want to fix that. We want to zero in on that one letter—A, C, T, or G is the four-letter alphabet of DNA, as I hope most of your listeners know—and we want to revert that back to whatever most normal, healthy people have in their genetic code at that specific position. CRISPR-Cas9, which won the Nobel Prize, is not the technology to do that sort of single base edit. It can do many other things, and the success in the clinic is unquestionable already in just a few years. But base editing and, in particular, prime editing are the two furthest developed technologies that allow investigators to pinpoint exactly where in the genome we want to make the edit. And then without completely cutting or slicing the double helix of DNA, we can lay up the section of DNA that we want to replace and go in and just perform chemistry on that one specific letter of DNA. Now, this hasn't been proven in the clinic just yet. But the early signs are very, very promising that this is going to be the breakthrough genome-editing technology over the next 10 to 20 years.Is CRISPR in the wild yet, or are we still in the lab?No, we're in the clinic. We are in human patients. There are at least 200 patients who have already been in or are currently enrolled in clinical trials. And so far, the early results—there are a few caveats and exceptions—but so far the overwhelming mood of the field is one of bullish enthusiasm. I don't want to complete this interview without singling out this one particular story, which is the clinical trial that has been sponsored by CRISPR Therapeutics and Vertex Pharmaceuticals for sickle cell disease. These are primarily African-American patients in this country because the sickle cell mutation arose in Africa some 7,000 years ago.We're talking about a pretty big share of the African-American population.This is about 100,000 patients just in America, in the US alone. And it's been a neglected disease for all kinds of reasons, probably beyond the scope of our discussion. But the early results in the first few dozen patients who have been enrolled in this clinical trial called the exa-cel clinical trial, they've all been cured. Pretty much all cured, meaning no more blood transfusions, no more pain crises, no more emergency hospitalizations. It is a pretty miraculous story. This therapy is now in the hands of the FDA and is speeding towards—barring some unforeseen complication or the FDA setting the bar so high that they need the investigators to go back and do some further checks—this should be approved before the end of this year.There's a catch, though. This will be a therapy that, in principle, will become—once approved by the FDA and the EMA in Europe, of course—will become available to any sickle cell patient. The catch will, of course, be the cost or the price that the companies set, because they're going to look for a return on their investment. It's a fascinating discussion and there's no easy answer. The companies need to reward their shareholders, their investors, their employees, their staff, and of course build a war chest to invest in the next wave, the next generation of CRISPR therapies. But the result of that means that probably we're going to be looking at a price tag of, I mean, I'm seeing figures like $1.9 million per patient. So how do you balance that? Is a lifetime cure for sickle cell disease worth $2, maybe $3 million? Will this patient population be able to afford that? In many cases, the answer to that will be simply, no. Do you have to remortgage your house and go bankrupt because you had a genetic quirk at birth? I don't know quite how we get around this.Different countries will have different answers with different health systems. Do you have a sense of what that debate is going to be like in Washington, DC?It's already happening in other contexts. Other gene therapies have been approved over the last few years, and they come with eye-watering price tags. The highest therapy price that I've seen now is $3.5 million. Yes, there are discounts and waiver programs and all this sort of stuff. But it's still a little obscene. Now, when those companies come to negotiate, say, with the UK National Health Service, they'll probably come to an agreement that is much lower, because the Brits are not going to say that they're going to be able to afford that for their significant sickle cell population.Is it your best guess that this will be a treatment the government pays for?What's interesting and what may potentially shift the calculus here is that this particular therapy is the disease affects primarily African-Americans in the United States. That may change the political calculus, and it may indeed change the corporate calculus in the boardrooms of Vertex and CRISPR Therapeutics, who may not want the backlash that they're going to get when they say, “Oh, by the way, guys, it's $2 million or you're out of luck.”There are companies that are studying using CRISPR to potentially correct the mutations that cause genetic forms of blindness, genetic forms of liver disease.What CRISPR therapies will come next?And after this CRISPR treatment for sickle cell disease is available, what therapies will come next?Probably a bunch of diseases that most people, unless they are unfortunate enough to have it in their family, won't have heard of. There are companies that are studying using CRISPR to potentially correct the mutations that cause genetic forms of blindness, genetic forms of liver disease. It turns out the liver is an organ that is very amenable to taking up medicines that we can inject in the blood. The other big clinical success story has come from another company in the Boston area called Intellia Therapeutics. Also publicly traded. They've developed CRISPR therapies that you can inject literally into the body, rather than taking cells out and doing it in the lab and then putting those cells back in, as in the case of sickle cell.I'm not sure that was actually even clear: that you can do it more than one way.Yes.And obviously it sounds like it would be better if they could just inject you.Exactly. That's why people are really excited about this, because this now opens up the doors for treating a host of diseases. And I think over the next few years we will see a growing number of diseases, and it won't just be these rare sort of genetic diseases with often unpronounceable names. It may be things like heart disease. There's another company—they're all in Boston, it seems—Verve Therapeutics, which is taking one of these more recent gene-editing technologies that we talked about a minute ago, base editing, and saying that there's a gene that they're going to target that has been clearly linked with cholesterol levels. And if we can squash production of this gene, we can tap down cholesterol levels. That will be useful, in the first instance, for patients with genetic forms of high cholesterol. Fair enough. But if it works in them, then the plan is to roll this out for potentially thousands if not millions of adults in this country who maybe don't feel that they have a clearly defined genetic form of high cholesterol, but this method may still be an alternative that they will consider versus taking Atorvastatin for the rest of your life, for example.Where are the CRISPR cancer treatments?They're also making progress, too. Those are in clinical trials. A little more complicated. Of course, cancer is a whole slew of different diseases, so it's a little hard to say, “Yeah, we're making progress here, less so there.” But I think one of the most heartwarming stories—this is an n of one, so it's an anecdotal story—but there was a teenager in the UK treated at one of the premier London medical schools who had a base editing form of CAR T therapy. A lot of people have heard of CAR T therapy for various cancers. And she is now in remission. So again, early days, but we're seeing very positive signs in these early clinical tests.It sounds like we went from a period where it was all in the lab and that we might be in a period over the next five years where it sounds like a wave of potential treatments.I think so, yeah.And for as much as we've seen articles about “The Age of AI,” it really sounds like this could be the age of biotechnology and the age of CRISPR…I think CRISPR, as with most new technologies, you get these sort of hype cycles, right? Two and a half years ago, CRISPR, all the stocks were at peak valuations. And I went on a podcast to say, why are the CRISPR stocks so high? I wasn't really sure, but I was enjoying it at the time. And then, of course, we entered the pandemic. And the biotech sector, perversely, ironically, has really been hit hard by the economy and certainly by the market valuations. So all of the CRISPR gene-editing companies—and there are probably at least eight or 10 now that are publicly traded and many more poised to join them—their valuations are a fraction of what they were a couple of years ago. But I suspect as these first FDA approvals and more scientific peer review papers, of course, but more news of the clinical success to back up and extend what has already been clearly proven as a breakthrough technology in the lab with the Nobel Prize—doesn't get much better than that, does it?—then I think we're going to start to see that biotech sector soar once again.Certainly, there are a lot of computational aspects to CRISPR in terms of designing the particular stretches of nucleic acid that you're going to use to target a specific gene. And AI can help you in that quest to make those ever more precise.Non-medical applications of gene editingThere are also non-medical applications. Can you just give me a little state of play on how that's looking?I think one of the—when CRISPR…And agriculture.Feeding the planet, you could say.That's certainly a big application.It's a human health application—arguably the biggest application.I think one of the fun ones is the work of George Church at Harvard Medical School, who's been on 60 Minutes and Stephen Colbert and many other primetime shows, talking about his work using CRISPR to potentially resurrect the woolly mammoth, which sort of sounds like, “That's Jurassic Park on steroids. That's crazy.” But his view is that, no, if we had herds—if that's the technical term—of woolly mammoths—roaming Siberia and the frozen tundra, they'll keep the ground, the surface packed down and stop the gigatons of methane from leaching out into the atmosphere. We have just seen a week, I've been reading on social media, of the hottest temperatures in the world since records began. And that's nothing compared to what we're potentially going to see if all these greenhouse gases that are just under the surface in places like Siberia further leach into the atmosphere. So that's the sort of environmental cause that Church is on. I think many people think this is a rather foolish notion, but he's launched a company to get this off the ground called Colossal Biosciences, and they're raising a lot of money, it appears. I'm curious to see how it goes. I wish him well.Also, speaking of climate change, making crops more resilient to the heat. That's another I've heard…One of the journals I'm involved in, called GEN Biotechnology, just published a paper in which investigators in Korea have used CRISPR to modify a particular gene in the tomato genome to make it a higher source of vitamin D. And that may not seem to be the most urgent need, but the point is, we can now engineer the DNA of all kinds of plants and crops, many of which are under threat, whether it's from drought or other types of climate change or pests, bacteria, parasites, viruses, fungi, you name it. And in my book Editing Humanity, which came out a couple of years ago, there was a whole chapter listing a whole variety of threats to our favorite glass of orange juice in the morning. That's not going to exist. If we want that all-natural Florida orange juice, we're not going to have that option. We've either got to embrace what technology will allow us to do to make these orange crops more resistant to the existential threat that they're facing, or we're going to have to go drink something else.I started out talking about AI and machine learning. Does that play a role in CRISPR, either helping the precision of the technology or in some way refining the technology?Yeah, hopefully you'll invite me back in a year and I'll be able to give you a more concrete answer. I think the short answer is, yes. Certainly, there are a lot of computational aspects to CRISPR in terms of designing the particular stretches of nucleic acid that you're going to use to target a specific gene. And AI can help you in that quest to make those ever more precise. When you do the targeting in a CRISPR experiment, the one thing you don't want to have happen is for the little stretch of DNA that you've synthesized to go after the gene in question, you don't want that to accidentally latch onto or identify another stretch of DNA that just by statistical chance has the same stretch of 20 As, Cs, Ts, and Gs. AI can help give us more confidence that we're only honing in on the specific gene that we want to edit, and we're not potentially going to see some unforeseen, off-target editing event.Do you think when we look back at this technology in 10 years, not only will we see a wider portfolio of potential treatments, but we'll look at the actual technique and think, “Boy, back in 2012, it was a butchery compared to what we're doing; we were using meat cleavers, and now we're using lasers”?I think, yeah. That's a slightly harsh analogy. With this original form of CRISPR, published in 2012, Nobel Prize in 2020, one of the potential caveats or downsides of the technology is that it involves a complete snip of the double helix, the two strands of DNA, in order to make the edit. Base editing and prime editing don't involve that double-stranded severance. It's just a nick of one strand or the other. So it's a much more genetically friendly form of gene editing, as well as other aspects of the chemistry. We look forward to seeing how base and prime editing perform in the clinic. Maybe they'll run into some unforeseen hurdles and people will say, “You know what? There was nothing wrong with CRISPR. Let's keep using the originally developed system.” But I'm pretty bullish on what base and prime editing can do based on all of the early results have been published in the last few years on mice and monkeys. And now we're on the brink of going into the clinic.One medical scenario that they laid out would be, what if two people with a deadly recessive disease like sickle cell disease, or perhaps a form of cystic fibrosis, wanted to have a healthy biological child?Bioweapons and the ethics of CRISPRThis podcast is usually very optimistic. So we're going to leave all the negative stuff for this part of the podcast. We're going to rush through all the downsides very quickly.First question: Especially after the pandemic, a lot more conversation about bioweapons. Is this an issue that's discussed in this community, about using this technology to create a particularly lethal or virulent or targeted biological weapon?Not much. If a rogue actor or nation wanted to develop some sort of incredibly virulent bioweapon, there's a whole wealth of genetic techniques, and they could probably do it without involving CRISPR. CRISPR is, in a way, sort of the corollary of another field called synthetic biology or synthetic genomics that you may have talked about on your show. We've got now the facility, not just to edit DNA, but to synthesize custom bits of DNA with so much ease and affordability compared to five or 10 years ago. And we've just seen a global pandemic. When I get that question, I've had it before, I say, “Yeah, did we just not live through a global pandemic? Do we really need to be engineering organisms?” Whether you buy the lab leak hypothesis or the bioengineering hypothesis, or it was just a natural transfer from some other organism, nature can do a pretty good job of hurting human beings. I don't know that we need to really worry too much about bioweapons at this point.In 2018, there was a big controversy over a Chinese researcher who created some genome-edited babies. Yeah. Is there more to know about that story? Has that become a hotter topic of discussion as CRISPR has advanced?The Chinese scientist, He Jiankui, who performed those pretty abominable experiments was jailed for the better part of three years. He got early release in China and slowly but surely he's being rehabilitated. He's literally now moved his operation from Shenzhen to Beijing. He's got his own lab again, and he's doing genome editing experiments again. I saw again on social media recently, he's got a petition of muscular dystrophy families petitioning Jack Ma, the well-known Chinese billionaire, to fund his operation to devise a new gene editing therapy for patients with Duchenne muscular dystrophy and other forms of muscular dystrophy. I wouldn't want He Jiankui let within a thousand miles of my kids, because I just wouldn't trust him. And he's now more recently put out a manifesto stating he thinks we should start editing embryos again. So I don't know quite what is going on.It seems the Chinese threw the book at him. Three years is not a trivial prison sentence. He was fined about half a million dollars. But somebody in the government there seems to be okay with him back at the bench, back in the lab, and dabbling in CRISPR. And I don't know that he's been asked, does he have any regrets over the editing of Lulu and Nana. There was a third child born a few months later as well. All he will say is, “We moved too fast.” That is the only caveat that he has allowed himself to express publicly.We know nothing more about the children. They're close to five years old now. There's one particular gene that was being edited was pretty messed up. But we know it's not an essential gene in our bodies, because there are many people walking around who don't have a functional copy of this CCR5 receptor gene, and they're HIV resistant. That was the premise for He Jiankui's experiment. But he has said, “No, they are off limits. The authorities are not going to reveal their identities. We are monitoring them, and we will take care of them if anything goes wrong.” But I think a lot of people in the West would really like to help, to study them, to offer any medical assistance. Obviously, we have to respect their privacy. The twin girls and the third child who was born a bit later, maybe they're being protected for their own good. How would you like it if you grew up through childhood and into your teenage years, to walk around knowing that you were this human experiment? That may be a very difficult thing to live with. So more to come on that.There's no legitimate discussion about changing that in the West or anywhere else?Obviously, in the wake of what He Jiankui did, there were numerous blue ribbon panels, including one just organized by the National Academy of Sciences, just a stone's throw from where we're talking today. And I thought that report was very good. It did two things. This was published a couple of years ago. Two important things came out of it. One is this all-star group of geneticists and other scientists said, “We don't think that human embryo editing should be banned completely. There may be scenarios down the road where we actually would want to reserve this technology because nothing else would help bring about a particular medical outcome that we would like.” And the one medical scenario that they laid out would be, what if two people with a deadly recessive disease like sickle cell disease, or perhaps a form of cystic fibrosis, wanted to have a healthy biological child?There are clinics around the country and around the world now doing something called pre-implantation genetic diagnosis. If you have a family history of a genetic disease, you can encourage the couple to do IVF. We form an embryo or bunch of embryos in the test tube or on the Petri dish. And then we can do a little biopsy of each embryo, take a quick sneak peek at the DNA, look to see if it's got the bad gene or perhaps the healthy gene, and then sort of tag the embryos and only implant the embryos that we think are healthy. This is happening around the country as we speak for hundreds, if not thousands, of different genetic diseases. But it won't work if mom and dad have a recessive, meaning two copies of a bad gene, because there's no healthy gene that you can select in any of those embryos. It would be very rare, but in those scenarios, maybe embryo editing is a way we would want to go. But I don't see a big clamor for this right now. And the early results have been published using CRISPR on embryos in the wake of He Jiankui did have said, “It's a messy technique. It is not safe to use. We don't fully understand how DNA editing and DNA repair works in the human embryo, so we really need to do a whole lot more basic science, as we did in the original incarnation of CRISPR, before we even dare to revisit editing human embryos.” Longevity is interesting because, of course, in the last 18 months there's a company in Silicon Valley called Altos, funded by Yuri Milner, employing now two dozen of the top aging researchers who've been lured away from academia into this transnational company to find hopefully cures or insights into how to postpone aging. Longevity and genetic enhancementsAnother area is using these treatments not to fix things, but to enhance people, whether it's for intelligence or some other trait. A lot of money pouring into longevity treatments from Silicon Valley. Do we know more about the potential of CRISPR for either extending lifespans or selecting for certain desirable traits in people?This sort of scenario is never going to go away. When it comes up, if I hear someone say, “Could we use CRISPR or any gene editing technology to boost intelligence or mathematical ability or music musical ability, or anything that we might want…”Or speed in the hundred meters.“…or speed in the hundred meters, to enhance our perfect newborn?” I would say, what gene are you going to enhance? Intelligence—are you kidding me? Half of the 10,000 genes are expressed in the human brain. You want to start meddling with those? You wouldn't have a prayer of having a positive outcome. I think we can pretty much rule that out. Longevity is interesting because, of course, in the last 18 months there's a company in Silicon Valley called Altos, funded by Yuri Milner, employing now two dozen of the top aging researchers who've been lured away from academia into this transnational company to find hopefully cures or insights into how to postpone aging. That's going to be a long, multi-decade quest to go from that to potentially, “Oh, let's edit a little embryo, our newborn son or daughter so they have the gift of 120 years on this decaying, overheating planet…” Yes, there's a lot to wade through on that.And you have another book coming out. Can you give us a preview of that?I'm writing a book called Curved Air, which is about the story of sickle cell disease. It was first described in a paper from physicians in Chicago in 1910 who were studying the curious anemia of a dental student who walked into their hospital one day. That gentleman, Walter Noel, is now buried back in his homeland, the island of Grenada. But in the 1940s, it was described and characterized as the first molecular disease. We know more about sickle cell disease than almost any other genetic disease. And yet, as we touched on earlier, patients with this who have not had the wealth, the money, the influence, they've been discriminated against in many walks of life, including the medical arena.We're still seeing terribly, tragically, videos and stories and reports of sickle cell patients who are being turned away from hospital rooms, emergency rooms, because the medical establishment just looks at a person of color in absolute agony with one of these pain crises and just assumed, “Oh, they want another opioid hit. Sickle cell? What is that?” There's a lot of fascinating science. There's all this hope in the gene editing and now in the clinic. And there's all this socioeconomic and other history. So I'm going to try to weave all this together in a format that hopefully everyone will enjoy reading.Hopefully a book with a happy ending. Not every book about a disease has a wonderful…I think a positive note to end on is the first American patient treated in this CRISPR clinical trial for sickle cell disease four years ago,Victoria Gray, has become something of a poster child now. She's been featured on National Public Radio on awhole series of interviews and just took her first overseas flight earlier this year to London to speak at a CRISPR gene editing conference. She gave a lovely 15-minute personal talk, shaking with nerves, about her personal voyage, her faith in God, and what's brought her here now, pain-free, traveling the world, and got a standing ovation. You don't see many standing ovations at medical conferences or genetics conferences. And if ever anybody deserved it, somebody like Victoria Gray did. Early days, but a very positive journey that we're on. This is a public episode. If you'd like to discuss this with other subscribers or get access to bonus episodes, visit fasterplease.substack.com/subscribe

Moonshots with Peter Diamandis
How AI Is Helping Solve Extinction w/ Ben Lamm | EP #53

Moonshots with Peter Diamandis

Play Episode Listen Later Jul 6, 2023 90:23


In this episode, Peter and Ben discuss the possibility of bringing extinct species back to life through Ben's startup, Colossal.  02:05 | De Extinction: Unveiling Our Future 21:10 | Gene-Swapping for Species Survival 59:32 | The Return of Extinct Species Ben Lamm is a renowned entrepreneur and visionary in technology and AI. With successful startups like Chaotic Moon Studios and Conversable, he has made significant contributions to the industry. As the founder of Hypergiant Industries and Colossal, a venture capital-backed startup focused on genetic engineering and reproductive technology in support of George Church's de-extinction efforts, Lamm continues to push the boundaries of innovation, focusing on AI solutions and genetic engineering. Learn about Colossal. _____________ I only endorse products and services I personally use. To see what they are,  please support this podcast by checking out our sponsors:  Experience the future of sleep with Eight Sleep. Visit https://www.eightsleep.com/moonshots/ to save $150 on the Pod Cover.  _____________ I send weekly emails with the latest insights and trends on today's and tomorrow's exponential technologies. Stay ahead of the curve, and sign up now:  Tech Blog _____________ Connect With Peter: Twitter Instagram Youtube Moonshots and Mindsets Learn more about your ad choices. Visit megaphone.fm/adchoices

Best New Ideas in Money
A mammoth mission

Best New Ideas in Money

Play Episode Listen Later Jul 6, 2023 21:13


A buzzy de-extinction startup is using genetic engineering and restorative biology for big projects — like bringing back the wooly mammoth. Gene-editing pioneer George Church joins us to discuss the venture. Learn more about your ad choices. Visit megaphone.fm/adchoices

MoneyBall Medicine
Non-standard Amino Acids in the Development of New Medical Therapies

MoneyBall Medicine

Play Episode Listen Later Jul 5, 2023 60:16


In the same way that written English is built around an alphabet of just 26 letters, all life on Earth is built around a standard set of just 20 amino acids, which are the building blocks of all proteins. And just as we've invented special characters like emoji to go beyond our standard letters, it turns out that biologists can expand their repertoire of powers using non-standard amino acids—those that either occur rarely in nature, or that can only be made in the lab. GRO Biosciences, a spinout from the laboratory of the renowned synthetic biology pioneer George Church at Harvard Medical School, is one of the companies working to explore the exciting applications of non-standard amino acids (NSAAs), and Harry's guest this weeks is GRO's co-founder and CEO, Dan Mandell. He says NSAAs could help overcome some of the limitations that keep today's gene and protein therapies from being used more widely, while also expanding the kinds of jobs that protein-based therapies can do.For a full transcript of this episode, please visit our episode page at http://www.glorikian.com/podcast Please rate and review The Harry Glorikian Show on Apple Podcasts! Here's how to do that from an iPhone, iPad, or iPod touch:1. Open the Podcasts app on your iPhone, iPad, or Mac. 2. Navigate to The Harry Glorikian Show podcast. You can find it by searching for it or selecting it from your library. Just note that you'll have to go to the series page which shows all the episodes, not just the page for a single episode.3. Scroll down to find the subhead titled "Ratings & Reviews."4. Under one of the highlighted reviews, select "Write a Review."5. Next, select a star rating at the top — you have the option of choosing between one and five stars. 6. Using the text box at the top, write a title for your review. Then, in the lower text box, write your review. Your review can be up to 300 words long.7. Once you've finished, select "Send" or "Save" in the top-right corner. 8. If you've never left a podcast review before, enter a nickname. Your nickname will be displayed next to any reviews you leave from here on out. 9. After selecting a nickname, tap OK. Your review may not be immediately visible.That's it! Thanks so much.

The Colin McEnroe Show
Back from the dead: Exploring the cutting edge of de-extinction

The Colin McEnroe Show

Play Episode Listen Later May 18, 2023 49:00


At the beginning of the year, the company behind the public effort to de-extinct the wooly mammoth announced it will also be de-extincting the dodo. The announcement stirred up a lot of excitement and questions about whether we can – or should – bring back species once they're gone. So this hour we're talking about de-extinction! We'll hear about what it takes to bring back extinct animals, efforts to build a safety net for plants that might go extinct in the future, and walk through some fun de-extinction thought experiments. GUESTS: Helen Pilcher: a science and comedy writer with a PhD in cell biology who wrote Bring Back the King: The New Science of De-extinction Ben Lamm: CEO of the de-extinction company Colossal, which he co-founded with George Church. Carlos de la Rosa: President and CEO of the Center for Plant Conservation Join the conversation on Facebook and Twitter.Support the show: http://www.wnpr.org/donateSee omnystudio.com/listener for privacy information.

Clearer Thinking with Spencer Greenberg
Science is learning from start-ups (with Adam Marblestone)

Clearer Thinking with Spencer Greenberg

Play Episode Listen Later May 15, 2023 73:39


Read the full transcript here. What are focused research organizations? Which kinds of research projects lend themselves to the FRO model? Researchers in academia frequently complain about the incentive structures around funding and publishing; so how do FROs change those dynamics? Why must FROs be time-limited, especially if they're successful? Who's in charge in an FRO? How does "field-building" help to improve science? What effects might large language models have on science?Adam Marblestone is the CEO of Convergent Research. He's been launching Focused Research Organizations (FROs) such as E11 bio and Cultivarium. He also serves on the boards of several non-profits pursuing new methods of funding and organizing scientific research including Norn Group and New Science. Previously, he was a Schmidt Futures Innovation Fellow, a consultant for the Astera Institute, a Fellow with the Federation of American Scientists (FAS), a research scientist at Google DeepMind, Chief Strategy Officer of the brain-computer interface company Kernel, a research scientist at MIT, a PhD student in biophysics with George Church and colleagues at Harvard, and a theoretical physics student at Yale. He also previously helped to start companies like BioBright and advised foundations such as the Open Philanthropy Project. His work has been recognized with a Technology Review 35 Innovators Under 35 Award (2018), a Fannie and John Hertz Foundation Fellowship (2010), and a Goldwater Scholarship (2008). Learn more about him at adammarblestone.org. [Read more]

Ask The Tech Guys (Audio)
ATTG 1964: Podcast Dreamin' - Google Takeout, Headphones for Sleep, Windows to Mac

Ask The Tech Guys (Audio)

Play Episode Listen Later Mar 6, 2023 144:12 Very Popular


Twitter cut off its API access, and developers of apps like Tweetbot and Twitterrific are offering specific options to help them out. A new iMac is coming out soon? Why can't I access my files from the Google Takeout service? Is there another service I can use to access my Google data? Is there an iOS shortcut for this? The Father of Modern Genomics, Dr. George Church. Lucid dreaming & headphones you can wear to sleep. Is there an easy way to move contacts over to Fastmail? Ant Pruitt & moving from Windows to Mac. Is it worth setting up IoT devices on its own network? Does Apple send out a notification to your old iPhone when your device reaches its end of life? Hosts: Leo Laporte and Mikah Sargent Guest: Ant Pruitt Get episodes ad-free with Club TWiT at https://twit.tv/clubtwit Show notes and links for this episode are available at: https://twit.tv/shows/ask-the-tech-guys/episodes/1964 Download or subscribe to this show at: https://twit.tv/shows/ask-the-tech-guys Sponsors: lectricebikes.com fortra.com

All TWiT.tv Shows (MP3)
Ask The Tech Guys 1964: Podcast Dreamin'

All TWiT.tv Shows (MP3)

Play Episode Listen Later Mar 6, 2023 144:12


Twitter cut off its API access, and developers of apps like Tweetbot and Twitterrific are offering specific options to help them out. A new iMac is coming out soon? Why can't I access my files from the Google Takeout service? Is there another service I can use to access my Google data? Is there an iOS shortcut for this? The Father of Modern Genomics, Dr. George Church. Lucid dreaming & headphones you can wear to sleep. Is there an easy way to move contacts over to Fastmail? Ant Pruitt & moving from Windows to Mac. Is it worth setting up IoT devices on its own network? Does Apple send out a notification to your old iPhone when your device reaches its end of life? Hosts: Leo Laporte and Mikah Sargent Guest: Ant Pruitt Get episodes ad-free with Club TWiT at https://twit.tv/clubtwit Show notes and links for this episode are available at: https://twit.tv/shows/ask-the-tech-guys/episodes/1964 Download or subscribe to this show at: https://twit.tv/shows/all-twittv-shows Sponsors: lectricebikes.com fortra.com

Radio Leo (Audio)
Ask The Tech Guys 1964: Podcast Dreamin'

Radio Leo (Audio)

Play Episode Listen Later Mar 6, 2023 144:12


Twitter cut off its API access, and developers of apps like Tweetbot and Twitterrific are offering specific options to help them out. A new iMac is coming out soon? Why can't I access my files from the Google Takeout service? Is there another service I can use to access my Google data? Is there an iOS shortcut for this? The Father of Modern Genomics, Dr. George Church. Lucid dreaming & headphones you can wear to sleep. Is there an easy way to move contacts over to Fastmail? Ant Pruitt & moving from Windows to Mac. Is it worth setting up IoT devices on its own network? Does Apple send out a notification to your old iPhone when your device reaches its end of life? Hosts: Leo Laporte and Mikah Sargent Guest: Ant Pruitt Get episodes ad-free with Club TWiT at https://twit.tv/clubtwit Show notes and links for this episode are available at: https://twit.tv/shows/ask-the-tech-guys/episodes/1964 Download or subscribe to this show at: https://twit.tv/shows/total-leo Sponsors: lectricebikes.com fortra.com

The Tech Guy (Video HI)
ATTG 1964: Podcast Dreamin' - Google Takeout, Headphones for Sleep, Windows to Mac

The Tech Guy (Video HI)

Play Episode Listen Later Mar 6, 2023 144:52


Twitter cut off its API access, and developers of apps like Tweetbot and Twitterrific are offering specific options to help them out. A new iMac is coming out soon? Why can't I access my files from the Google Takeout service? Is there another service I can use to access my Google data? Is there an iOS shortcut for this? The Father of Modern Genomics, Dr. George Church. Lucid dreaming & headphones you can wear to sleep. Is there an easy way to move contacts over to Fastmail? Ant Pruitt & moving from Windows to Mac. Is it worth setting up IoT devices on its own network? Does Apple send out a notification to your old iPhone when your device reaches its end of life? Hosts: Leo Laporte and Mikah Sargent Guest: Ant Pruitt Get episodes ad-free with Club TWiT at https://twit.tv/clubtwit Show notes and links for this episode are available at: https://twit.tv/shows/ask-the-tech-guys/episodes/1964 Download or subscribe to this show at: https://twit.tv/shows/ask-the-tech-guys Sponsors: lectricebikes.com fortra.com

All TWiT.tv Shows (MP3)
Triangulation 433: The Father of Modern Genomics

All TWiT.tv Shows (MP3)

Play Episode Listen Later Mar 3, 2023 60:24


In a wide-ranging conversation, Leo Laporte talks with Dr. George Church about the history and future of genome sequencing. Dr. Church explains why he is very excited about how they made an organism resistant to all viruses- both known and unknown.   They also talk about:  How Dr. Church made sequencing affordable (from $3BN to $600) Where are we on sequencing the entire human genome, and why would we even want to? How close are we to reversing aging? Should we use gene therapy to double the human lifespan? Why genetically engineering cold-resistant elephants could be important for climate change, and what about ultimately bringing back the Mammoth? He explains the difference between the hype and reality of CRISPR sequencing, synthesis, and delivery What are the privacy concerns about companies sharing your genome? They discuss the controversy and benefit of reconstructing old viruses like the 1918 flu and even inventing new viruses. Dr. Church is a professor of genetics at Harvard Medical School and a pioneer in personal genetics. He is credited with developing the first methods for sequencing the first genome and also the CRISPR technology. He created several companies, including Nebula Genomics and Veritas Genetics. Host: Leo Laporte Guest: Dr. George Church Download or subscribe to this show at https://twit.tv/shows/triangulation.

All TWiT.tv Shows (MP3)
TWiT Events 15: The Father of Modern Genomics

All TWiT.tv Shows (MP3)

Play Episode Listen Later Mar 3, 2023 60:24


In a wide-ranging conversation, Leo Laporte talks with Dr. George Church about the history and future of genome sequencing. Dr. Church explains why he is very excited about how they made an organism resistant to all viruses- both known and unknown.   They also talk about:  How Dr. Church made sequencing affordable (from $3BN to $600) Where are we on sequencing the entire human genome, and why would we even want to? How close are we to reversing aging? Should we use gene therapy to double the human lifespan? Why genetically engineering cold-resistant elephants could be important for climate change, and what about ultimately bringing back the Mammoth? He explains the difference between the hype and reality of CRISPR sequencing, synthesis, and delivery What are the privacy concerns about companies sharing your genome? They discuss the controversy and benefit of reconstructing old viruses like the 1918 flu and even inventing new viruses. Dr. Church is a professor of genetics at Harvard Medical School and a pioneer in personal genetics. He is credited with developing the first methods for sequencing the first genome and also the CRISPR technology. He created several companies, including Nebula Genomics and Veritas Genetics. Host: Leo Laporte Guest: Dr. George Church Download or subscribe to this show at https://twit.tv/shows/twit-events. Get episodes ad-free with Club TWiT at https://twit.tv/clubtwit

Radio Leo (Audio)
Triangulation 433: The Father of Modern Genomics

Radio Leo (Audio)

Play Episode Listen Later Mar 3, 2023 60:24


In a wide-ranging conversation, Leo Laporte talks with Dr. George Church about the history and future of genome sequencing. Dr. Church explains why he is very excited about how they made an organism resistant to all viruses- both known and unknown.   They also talk about:  How Dr. Church made sequencing affordable (from $3BN to $600) Where are we on sequencing the entire human genome, and why would we even want to? How close are we to reversing aging? Should we use gene therapy to double the human lifespan? Why genetically engineering cold-resistant elephants could be important for climate change, and what about ultimately bringing back the Mammoth? He explains the difference between the hype and reality of CRISPR sequencing, synthesis, and delivery What are the privacy concerns about companies sharing your genome? They discuss the controversy and benefit of reconstructing old viruses like the 1918 flu and even inventing new viruses. Dr. Church is a professor of genetics at Harvard Medical School and a pioneer in personal genetics. He is credited with developing the first methods for sequencing the first genome and also the CRISPR technology. He created several companies, including Nebula Genomics and Veritas Genetics. Host: Leo Laporte Guest: Dr. George Church Download or subscribe to this show at https://twit.tv/shows/triangulation.

Radio Leo (Audio)
TWiT Events 15: The Father of Modern Genomics

Radio Leo (Audio)

Play Episode Listen Later Mar 3, 2023 60:24


In a wide-ranging conversation, Leo Laporte talks with Dr. George Church about the history and future of genome sequencing. Dr. Church explains why he is very excited about how they made an organism resistant to all viruses- both known and unknown.   They also talk about:  How Dr. Church made sequencing affordable (from $3BN to $600) Where are we on sequencing the entire human genome, and why would we even want to? How close are we to reversing aging? Should we use gene therapy to double the human lifespan? Why genetically engineering cold-resistant elephants could be important for climate change, and what about ultimately bringing back the Mammoth? He explains the difference between the hype and reality of CRISPR sequencing, synthesis, and delivery What are the privacy concerns about companies sharing your genome? They discuss the controversy and benefit of reconstructing old viruses like the 1918 flu and even inventing new viruses. Dr. Church is a professor of genetics at Harvard Medical School and a pioneer in personal genetics. He is credited with developing the first methods for sequencing the first genome and also the CRISPR technology. He created several companies, including Nebula Genomics and Veritas Genetics. Host: Leo Laporte Guest: Dr. George Church Download or subscribe to this show at https://twit.tv/shows/twit-events. Get episodes ad-free with Club TWiT at https://twit.tv/clubtwit

Blazing Trails
Can Bringing Back the Woolly Mammoth Help Save the Planet?

Blazing Trails

Play Episode Listen Later Jan 25, 2023 20:32


Could bringing back the woolly mammoth be a key to saving the planet? Tech entrepreneur Ben Lamm and Harvard geneticist George Church are betting on it. Together they launched Colossal Bioscience, a genetic engineering company focused on using gene editing technology to advance de-extinction. But the company has a much larger goal, re-introducing native populations to help restore natural ecosystems. Join us to hear Ben and George talk about their pioneering work. For more information on Colossal go to www.colossal.com.

Real Coffee with Scott Adams
Episode 1975 Scott Adams: 2023 Starts With Some Excellent Fake News, Conspiracy Theories And More

Real Coffee with Scott Adams

Play Episode Listen Later Jan 1, 2023 51:25 Very Popular


My new book LOSERTHINK, available now on Amazon https://tinyurl.com/rqmjc2a Find my "extra" content on Locals: https://ScottAdams.Locals.com Content: Charlottesville and Richard Spencer Could I hypnotize AI? Andrew Tate Ben Garrison, my new nemesis George Church aging reversal progress Alzheimer treatment breakthrough ~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~ If you would like to enjoy this same content plus bonus content from Scott Adams, including micro-lessons on lots of useful topics to build your talent stack, please see scottadams.locals.com for full access to that secret treasure. --- Support this podcast: https://anchor.fm/scott-adams00/support

Open to Debate
Should We Use Gene Editing to Make Better Babies?

Open to Debate

Play Episode Listen Later Oct 7, 2022 53:19 Very Popular


A genetic disease runs in your family. Your doctor tells you that, should you wish to have a child, that child is likely to also carry the disease. But a new gene-editing technology could change your fate. It could ensure that your baby is -- and remains -- healthy. What do you do? It's is not without its perils. Critics say the technology will exacerbate inequality, pressure all parents (and nations) into editing their children to stay competitive, and meddle with the most basic aspect of our humanity. So, should we use gene editing to make better babies?   Arguing in favor of the motion is geneticist George Church and futurist Amy Webb. Arguing against the motion is policy advocate Marcy Darnovsky and philosopher Françoise Baylis. Emmy award-winning journalist John Donvan moderates.  Learn more about your ad choices. Visit megaphone.fm/adchoices

The Doctor's Farmacy with Mark Hyman, M.D.
How Reprogramming Our Genes Can Extend Our Lives with George Church

The Doctor's Farmacy with Mark Hyman, M.D.

Play Episode Listen Later Aug 31, 2022 73:03 Very Popular


This episode is brought to you by Rupa Health, Cozy Earth, and InsideTracker.All living things are programmed with a certain lifespan, which can be dramatically different from species to species. Humans now, in general, live twice as long as our ancestors, thanks to environmental changes and advances in medicine. But most of us spend the last years of our lives without the quality of life we really want. We've come to identify a slew of symptoms and chronic diseases as a natural part of aging, but do they really have to be?Today I'm excited to talk to Dr. George Church all about the latest science on reprogramming our genes to extend our healthspan and lifespan. Dr. George Church is a professor of genetics at Harvard Medical School, a founding member of the Wyss Institute, and director of PersonalGenomes.org, the world's only open-access information on human genomic, environmental, and trait data. Dr. Church is known for pioneering the fields of personal genomics and synthetic biology. He developed the first methods for the first genome sequence and his team invented CRISPR for human stem cell genome editing and other synthetic biology technologies and applications—including new ways to create organs for transplantation, gene therapies for aging reversal, and gene drives to eliminate Lyme disease and malaria. Dr. Church is the director of IARPA & NIH BRAIN Projects and the National Institutes of Health Center for Excellence in Genomic Science. He is the author of Regenesis.This episode is brought to you by Rupa Health, Cozy Earth, and InsideTracker.Rupa Health is a place where Functional Medicine practitioners can access more than 2,000 specialty lab tests. You can check out a free, live demo with a Q&A or create an account at RupaHealth.com.Right now, get 40% off your Cozy Earth sheets. Just head over to cozyearth.com and use code MARK40.InsideTracker is a personalized health and wellness platform like no other. Right now they're offering my community 20% off at insidetracker.com/drhyman.Here are more details from our interview (audio version / Apple Subscriber version):The top things that prevent disease and enhance longevity (7:06 / 4:13) Defining aging as a disease and treating it as such (8:24 / 5:40) Reprogramming and repairing cells to a younger state (12:24 / 9:35) The future, and challenges, of delivering Yamanaka factors for cell reprogramming (15:05 / 11:50) Gene editing vs gene therapy (28:02 / 22:36) Using gene therapy to reverse disease and lengthen life span (34:44 / 30:55) Animal-to-human organ transplants (37:34 / 33:04) Can we live youthfully to 100 years old and beyond? (44:31 / 40:34) How much and what types of protein do we need for healthy aging? (50:30 / 45:50) Using gene editing to bring back mammoths to restore damaged ecosystems (1:00:53 / 56:50) Get a copy of George Church and Ed Regis' book, Regenesis: How Synthetic Biology Will Reinvent Nature and Ourselves, here. Our GDPR privacy policy was updated on August 8, 2022. Visit acast.com/privacy for more information.

FoundMyFitness
#077 Rewriting genomes to eradicate disease and aging | Dr. George Church

FoundMyFitness

Play Episode Listen Later Aug 24, 2022 128:20 Very Popular


George Church, Ph.D. is a professor of genetics at Harvard Medical School and of health sciences and technology at both Harvard and the Massachusetts Institute of Technology. Dr. Church played an instrumental role in the Human Genome Project and is widely recognized as one of the premier scientists in the fields of gene editing technology and synthetic biology.  In this episode, we discuss: 07:13 - History of the Human Genome Project 15:20 - Manufacturing cell phones (with biology) 17:34 - Genome Project-Write 20:03 - Writing a human Y chromosome (from scratch) 20:48 - What if you could eliminate viral disease? 22:51 - De-extinction and reinstating lost traits and genes 27:06 - The Vertebrate Genomes Project 29:47 - AlphaFold and other AI tools 41:27 - CRISPR vs. Base Editing (emerging tools of genetic engineering) 49:40 - Why multiplex editing will change the world 52:18 - Molecular flight recorder 53:31 - Preventing viral spillover and enhancing livestock 57:40 - PCSK9 gene therapy for cholesterol 01:00:30 - Is aging an evolved program? 01:05:21 - Treating aging with a combination gene treatment 01:09:04 - Does animal research help us understand human aging? 01:11:40 - Human organoids as a model and therapeutic 01:13:34 - Could engineered transplant organs become better than the originals? 01:16:17 - Embryo editing controversy 01:28:41 - Gene editing for space travel 01:30:40 - Can synthetic biology alleviate poverty? 01:34:07 - Is in vitro fertilization and embryo selection practically similar to editing? 01:39:12 - The occasional cost of brilliance 01:45:45 - Eradicating disease with Gene Drive 01:48:55 - Technologies to solve Lyme disease 01:51:57 - Dr. Church's experience with narcolepsy as a bridge to creative insights 02:00:42 - Why George encoded his book in DNA Watch this episode on YouTube Show notes are available by clicking here Join over 300,000 people and get the latest distilled information straight to your inbox weekly: https://www.foundmyfitness.com/newsletter Become a FoundMyFitness premium member to get access to exclusive episodes, emails, live Q+A's with Rhonda and more: https://www.foundmyfitness.com/premium Learn more about the premium podcast The Aliquot: https://www.foundmyfitness.com/aliquot