Revolutionizing the Textile Industry from the Factory Floor – A Conversation with Bloom Labs

We talked with two founders of Bloom Labs — Sim Gulati, CEO and Richard Freundlich, CTO — in the basement of IndieBio last week after their company had just been voted “Killer of the Week” by their peers in Batch 14. Each week at IndieBio, the companies in our batch compete based on their top accomplishments just in the previous week as they learn to move at venture speed. Top companies are nominated by the IndieBio SF team, but the winner is ultimately chosen by a voting of their peers within the batch.

Sim’s been a part of the textile industry since before he could walk

Sim started in the textile industry as a kid, helping his family run their clothing factory in India to create export-quality cotton and polyester for big name retail stores like J.C. Penney and LuluLemon.

[Sim]: “After learning the fundamentals as well as the harmful impacts of the textile industry, I became a bit obsessed with how to circularize it. Due to my young age, I wasn’t able to realize this vision within the family business so I knew it was time to set out on my own.”

[Sierra]: Wow so you’ve been an entrepreneur for pretty much your entire life. And this moment gave you the push to start Bloom?

[Sim]: “Not quite – first I formed a company called Dropel where we were innovating fabric to make it more sustainable. I quickly realized that the innovation had to be more upstream to enable a true paradigm shift in the industry – that’s where Bloom came in. At Bloom, we are innovating at the level of the raw material for the fabric. This way we can introduce massive innovation into the textile industry without requiring it to accept major infrastructure changes.”

[Mohan]: How did you settle on protein waste as your feedstock of choice?

[Sim]: “Feedstock for textile fibers has to be abundant and scalable. There is actually over 15 billion pounds of protein waste produced annually by poultry farmers around the globe. Yeah, we humans eat a lotta chicken! We came up with the concept of how to use this feedstock for melt spinning and melt extrusion processes without having to build new capex. In our proprietary process we convert this waste into keratin pellets which can drop into existing textile manufacturing infrastructure today. While we are a startup, we can scale almost immediately with existing equipment.”

After flying planes for 30 years, Richard is ready to repent for his impacts on the ozone layer

After embarking on his mission to circularize the textile industry with waste protein feedstocks, Sim met Richard, CTO, as well as Mike Jaffe, now Bloom’s CSO, who had both previously worked together at Celanese in fiber manufacturing. Richard had actually worked on protein waste conversion technology previously in the context of sustainable food packaging.

[Richard]: “I knew the core technology behind Bloom would work as I had previously done it. Once Sim introduced me to the concept of using the process for fiber creation, I immediately saw immense potential to transform the textile industry and was on board to join full time.”

[Sierra]: “It must be so satisfying to see your technology come full circle like this”

[Richard]: “Bloom is truly providing the right solution at the right time. 30 years ago no one cared about keratin as oil was so cheap. Now that circularity is crucial to enable a sustainable future, our scalable, cost-effective solution is poised to truly disrupt the textile industry…30 years ago is also when I first began my passion of flying planes. I’m thrilled to now be giving back to our climate to more than compensate for the negative impacts of that!”

Progress over perfection: how to manage conflict among an all-star team of engineers and scientists

Each member of the team at Bloom possesses a unique skillset to propel the company forward. From experts in melt spinning to materials science to chemists – Sim has assembled a great team with expertise over every aspect of protein processing and fiber generation. Of course, managing such a team of brilliant scientists and engineers can be challenging.

[Richard]: “Different experts on our team can clash from time to time. But we have constant communication and meetings to make sure everyone is aligned on key objectives and how to meet them.”

[Mohan]: Is managing the priorities and expectations of scientists different compared to doing the same with engineers?

[Sim]: “With tight deadlines it can be difficult for scientists to deliver. We’ve recently accomplished 6 months of work in just 3 weeks to prepare for the IndieBio SF Demo Day. To get there, we’ve had to juggle plenty of unknowns and focus mainly on getting to our demo day goals rather than fully understanding the intricacies of each result. This can drive the scientists insane, but at the end of the day our focus as a disruptive startup is really on progress over perfection.”

Why Bloom won Killer this week

[Sierra]: So what earned you guys the Killer last week?

[Sim]: “Last week, we successfully processed waste protein into uniform pellets for the first time. Next, we’re going to conduct a full scale trial. This is crucial to our development as a company as we are using scalable machinery and have taken the time to fully understand all of our process variables. So we will easily be able to move to the next stage of development using existing commercial scale equipment.”

Through our short conversation we could really sense the commitment of the Bloom team to access an abundant feedstock supply, build a truly unique team, and ultimately become the low cost leader in renewable (and perhaps even nonrenewable) textile manufacturing.  We can’t wait to see how this team has progressed the next time we talk.

Stay tuned for a deep dive into our next Killer!

The FDA and USDA approved Upside’s cultivated meat. Will it ever reach your table? Find out at the SOSV Climate Tech Summit.

UPSIDE Foods has made history yet again. The first company ever to create cultivated meatballs, chicken, and duck now has the first-ever approvals from the U.S. Food and Drug Administration (FDA) and Department of Agriculture (USDA) to sell its chicken commercially. UPSIDE chicken debuted August 4, 2023 at San Francisco’s Bar Crenn in partnership with three-Michelin-star chef Dominique Crenn. 

So, when will UPSIDE chicken be available at grocery stores and restaurants, not just at a $150 six-course meal? 

Cultivated meat still has some hurdles to clear. At the SOSV Climate Tech Summit (Sept 26-27 / free & virtual / register now), UPSIDE Foods founder and CEO Dr. Uma Valeti, MD will discuss the state of cultivated meat with Po Bronson, Managing Director of IndieBio and General Partner at SOSV. 

Cultivated meat is an important climate technology because livestock account for 10 to 20% of greenhouse gas emissions worldwide. Livestock also use almost 80% of the world’s agricultural land (lots and lots of feed…), consume 10% of annually available fresh water, and incentivize deforestation in places like the Amazon. Cultivated meat, on the other hand, is grown in bioreactors and could eliminate that collateral climate damage. The cells are nourished with a culture medium rich in oxygen, amino acids, glucose, vitamins, salts, and proteins, all of which can be sourced from plants and other clean sources.  

Uma’s journey to cultivated meat started during his upbringing in India, where he witnessed the mass slaughter of animals on several occasions and decided to give up meat. Later, as a cardiologist at the Mayo Clinic experimenting with cardiac stem cells, Uma realized he could stem the slaughter by growing animal tissues in bioreactors. He left medicine to prove that tasty, cultivated meat, indistinguishable from the animal-born variety, was possible. 

With SOSV’s IndieBio as his first backer, Uma produced the world’s first cultivated meat at IndieBio’s office in San Francisco.  UPSIDE has gone on to raise over $600M in funding from a mix of institutional, individual, and strategic investors that include TemasekAbu Dhabi Growth Fund (ADG), John Doerr, Bill Gates, Cargill, and Tyson Foods. In 2022, UPSIDE Foods was the subject of Meat the Future, an award-winning documentary about the quest for cultured meat, narrated by Jane Goodall.  

The Good Food Institute, a nonprofit think tank, tracks approximately 100 cultivated meat companies worldwide. The biggest hurdle for the category is cost—cultivated meats remain significantly more expensive than the government-subsidized variety enjoyed in the U.S. and elsewhere. Other challenges include texture and taste (particularly for premium cuts of meat) and public perception.    

What will it take for cultured meat to win in the $1.4 trillion global meat market?

Register today for the SOSV Climate Tech Summit to find out.

The Speakers


Dr. Uma Valeti photo

Dr. Uma Valeti

Dr. Uma Valeti is the CEO and Founder of UPSIDE Foods, the world’s leading cultivated meat company. UPSIDE Foods was the first company focused on growing meat directly from animal cells and has led the industry ever since. Since 2015, UPSIDE Foods has achieved numerous industry-defining milestones, including being the first company to produce multiple species of cultivated meat (beef, chicken and duck), to partner with the existing meat industry, and to sell a cultivated meat product in the United States. UPSIDE Foods has raised over $600 million from a diverse coalition of investors including Richard Branson, John Mackey, Bill Gates, Temasek, ADG, Softbank, Threshold, Baillie Gifford, Norwest, Cargill, Tyson Foods and Whole Foods. Uma’s vision is to enable people around the world to eat the meat they love, while transforming the food system to become more sustainable and humane. Uma has been featured in the Wall Street Journal, the Aspen Ideas Festival and SXSW. In 2019 he was named a “Global Thinker of the Decade” by Foreign Policy magazine. Uma is a Mayo Clinic-trained cardiologist and an adjunct professor in Cardiovascular Medicine at Stanford University.


Po Bronson photo

Po Bronson

Po Bronson is passionate about reconceptualizing complex challenges into more elegant forms, to broaden understanding and highlight priorities. He’s been at IndieBio SF since 2018. Po is a longtime science journalist honored with nine national awards, and author of seven bestselling books that are available in 28 languages worldwide. His work has been cited in 185 academic journals and 503 books. His background is in Economics. He learned finance at Credit Suisse and consulting at a division of Arthur Andersen. Prior to IndieBio he spent four years as a Futurist with Attention Span Media, consulting corporate innovation efforts for globally-recognized brands. Most recently, Po is the author of Decoding the World: A Roadmap for the Questioner, published by Twelve Books, a division of the Hachette Publishing Group.


SOSV Climate Tech Summit: A Conversation with Vinod Khosla and Sean O’Sullivan

Back in 2012 if you had predicted that the climate tech startup category would be worth $2.6 trillion today, you would have been laughed out of the room. That year was the nadir of first “clean tech” investing wave, which poured some $25 billion into earth-saving startups. Less than half of that capital was returned according to an infamous MIT postmortem (which conveniently excluded Tesla). Deep technology in particular “…consumed the most capital and yielded the lowest returns,” said the report. 

But two investors would have believed you. Their venture firms, Khosla Ventures and SOSV, continued to back deep tech, climate-focused companies despite the troubled track record. Today, the firms have two of the best climate portfolios in the world. Vinod Khosla and Sean O’Sullivan joined us at the SOSV Climate Tech Summit (Sept 26-27 / free & virtual) for a one-on-one conversation about the past, present and future of climate tech investing. 

Vinod, born in an Indian army household, migrated to the United States in 1976 and co-founded two landmark companies: Daisy Systems, the first major design software for electrical engineers, and Sun Microsystems, a pioneer in computing workstations and servers that invented the Java programming language. Khosla then joined Kleiner Perkins Caufield and Byers (KPCB) as a General Partner, leading investments in semiconductor company NexGen and internet routing pioneer Juniper Networks (earning a 2,500x return). He founded Khosla Ventures in 2004 partly to fund “science experiments,” as he puts it, and became an OG of the clean tech 1.0 investment wave. 

For Sean, born in New York City, the path to climate tech venture was also marked by early success. As an undergraduate engineer at Rensselaer Polytechnic Institute in 1986, he co-founded his first company, a desktop PC mapping app called MapInfo. Sean took the company public in 1994 and founded SOSV in 1995 to invest as a super angel. His early climate tech investments included Carma (2007), GetAround (2011) and Jump Bikes (2012), and today SOSV’s Climate Tech 100 is a powerful statement of Sean’s commitment to the category.

When most VCs gave up on cleantech around 2012, Vinod and Sean kept exploring and investing. Vinod called for “Reinventing Societal Infrastructure with Technology” in a 2018 blog post. Innovation in food, health, housing, transportation, industry—“The big needs in society”—would inherently produce cleaner, climate-friendly technologies, he argued. Sean, meanwhile, scaled SOSV from a personal investing vehicle into a global firm focused on “deep tech for human and planetary health.” Across industries, there was an opportunity for the “reinvention of all of these different means of production,” as Sean told Bloomberg earlier this year.

Today, Khosla Ventures has $15B under management and has invested in over 30 climate tech companies. Four have gone public—Amyris (bio-based materials), QuantumScape (solid state batteries), View (smart buildings), and LanzaTech (climate-friendly materials and fuels). Khosla also backs several climate unicorns including Commonwealth Fusion Systems (nuclear fusion) and Impossible Foods (plant-based meats).

SOSV has $1.5 billion under management and operates the IndieBio and HAX startup development programs, which launch 100+ pre-seed companies a year in bio-tech and hard tech. Many of those are climate tech companies, and Pitchbook has recognized SOSV as the most active investor in climate globally. The top 100 companies in the SOSV climate portfolio represent $11.69B in value and have raised $3.77B. They include unicorns like Perfect Day (fermentation-based proteins), NotCo (plant-based proteins using AI), UPSIDE Foods (cultured meat), and Formlabs (3D printing). Khosla Ventures and SOSV have co-invested in two climate startups: Oobli (sweet proteins) and Vertical Oceans (shrimp farming).

What can we learn from these titans of climate tech about backing breakthrough “science experiments”?

Catch the full session—and all others from this year’s Summit—here.

Learn more about the Summit.

The Speakers


Vinod Khosla photo

Vinod Khosla

Vinod Khosla is an entrepreneur, investor, and technology fan. He is the founder of Khosla Ventures, focused on impactful technology investments in software, AI, robotics, 3D printing, healthcare and more. Mr. Khosla was a co-founder of Daisy systems and founding CEO of Sun Microsystems where he pioneered open systems and commercial RISC processors. One of Mr. Khosla’s greatest passions is being a mentor to entrepreneurs, assisting entrepreneurs and helping them build technology-based businesses. Mr. Khosla is driven by the desire to make a positive impact through using technology to reinvent societal infrastructure and multiply resources. He is also passionate about Social Entrepreneurship. Vinod holds a Bachelor of Technology in Electrical Engineering from IIT, New Delhi, a Master’s in Biomedical Engineering from Carnegie Mellon University and an MBA from the Stanford Graduate School of Business.


Sean O’Sullivan photo

Sean O’Sullivan

Sean O’Sullivan is an entrepreneur, venture capitalist and philanthropist. He started SOSV in 1995, and today the firm operates two pre-seed startup development programs, HAX and IndieBio, which graduate more than 100 companies each year in bio tech and hard tech addressing the broad mission of “human and planetary health.” The states of New York and New Jersey committed $25 million each to support IndieBio in New York City and HAX in Newark. Sean’s family foundation wrote the first major check for Salman Khan’s Khan Academy and also co-founded CoderDojo and the Autism Impact Fund. Sean holds a Bachelors of Science in Electrical Engineering from Rensselaer Polytechnic Institute and a Masters of Fine Arts in Film Production from the University of Southern California.


Can bio-economy unicorn Solugen decarbonize the chemical industry? Find out at the SOSV Climate Summit

The $4.7 trillion chemical industry accounts for about 4% of global GDP and 5% of carbon emissions. The industry is proving hard to decarbonize due to its use of carbon-intensive feedstock (e.g., petroleum) and high heat in production. Solugen, valued at over $2B, aims to deliver the green  solution. At the SOSV Climate Tech Summit (Sept 26-27 / free & virtual / register now), Solugen co-founder and CEO Dr. Gaurab Chakrabarti, MD, PhD sat down with Dr. John Cumbers of SynBioBeta, a synthetic biology innovation network, to discuss how bio startups can eliminate emissions.   

Solugen is part of the multitrillion-dollar bioeconomy, which gained momentum in September 2022 after President Joe Biden issued an executive order “…to advance biotechnology and biomanufacturing towards innovative solutions in health, climate change, energy, food security, agriculture, supply chain resilience, and national and economic security.” 

It’s a tall order from the White House but tailor-made  for companies like Solugen. The team has developed a carbon-negative molecule factory called a “Bioforge” that uses proprietary engineered enzymes to produce chemicals (it’s “…not your grandpa’s chemical plant,” as Solugen puts it). Solugen claims that it can synthesize 90% of the world’s chemicals using nothing but sugar, water, catalysts, and air (and renewable electricity)—with zero emissions and no wastewater. It also claims that over 90% of these feedstocks are converted to product, meaning Solugen minimizes yield losses that make traditional thermochemical and fermentation processes so expensive.

Currently, Solugen’s product line includes chemicals for plant nutrition, water treatment, concrete production, cleaning agents, skincare products, and, somewhat unexpectedly, oil and gas extraction. To date, Solugen has raised over $640M in funding. Its backers include Kennivik, Lowercarbon Capital, Refactor Capital, BlackRock, GIC, Temasek, Baillie Gifford, Founders Fund, and Fifty Years

How much scale will Solugen need to make a serious dent in emissions? Can they outcompete chemical manufacturers with bio-based substitutes? And how else might bioeconomy startups like Solugen help with decarbonization? 

Catch the full session—and all others from this year’s Summit—here.

Learn more about the Summit.

The Speakers


Dr. Gaurab Chakrabarti, MD, PhD photo

Dr. Gaurab Chakrabarti, MD, PhD

Dr. Gaurab Chakrabarti, MD, PhD is the co-founder and CEO of Solugen. As a physician-scientist, Gaurab took an oath to first do no harm—and for him, that goes beyond patients or medicine. So in 2016, he started Solugen to decarbonize the chemicals industry, one of the most damaging for people and the Earth. Gaurab believes in using biology in unconventional ways to solve incredibly complex problems and is building a world-class team to join him on the journey. Gaurab studied computational neuroscience as an undergraduate at Brown University and received his MD & PhD in cancer biology and enzymology at the University of Texas. He is an author or co-author on more than 20 peer-reviewed publications and patents, and he is a Forbes 30 Under 30 in Industry and Manufacturing.


Dr. John Cumbers photo

Dr. John Cumbers

Dr. John Cumbers is the founder and CEO of SynBioBeta, the leading community of innovators, investors, engineers, and thinkers who share a passion for using synthetic biology to build a better, more sustainable universe. He publishes the weekly SynBioBeta Digest, hosts the SynBioBeta Podcast, and wrote “What’s Your Biostrategy?”, the first book to anticipate how synthetic biology is going to disrupt virtually every industry in the world. John also founded BetaSpace, a space settlement innovation network and community of visionaries, technologists, and investors accelerating the industries needed to sustain human life here and off-planet. John has been involved with multiple startups, he’s an operating partner and investor at the hard tech investment fund Data Collective, and he’s a former bioengineer at NASA. John earned his PhD in Molecular Biology, Cell Biology, and Biochemistry from Brown University and is originally from the UK.


SOSV startup competition focused on CO2 capture and removal. Early stage founders welcome. Application deadline July 28.

As part of the upcoming SOSV Climate Tech Summit—taking place virtually September 26-27th this year— we’re thrilled to announce our partnership with Extreme Tech Challenge (XTC) to host a Startup Innovation Challenge (aka a startup competition!) focused on Carbon Removal and Carbon Capture, Utilization & Storage (CCUS). Apply here—the deadline is July 28.

The competition is open to all early-stage startups (Series A or earlier) that are focusing on Carbon Removal and Carbon Capture, Utilization & Storage anywhere in the world. 

An expert team will review the applications and select five finalists, who will pitch before a live, online audience and judges at the SOSV Climate Tech Summit on September 26. The judges will announce the winner the next day during the summit. 

Why participate? The finalists will pitch live to an online audience of VCs, and video archives from the pitch will be published on SOSV’s site after the event. Overall, we expect 5,000 attendees at the summit. There’s no better place for climate founders to find investors. Check out this year’s speakers.

The third annual SOSV Climate Tech Summit convenes the entire climate tech ecosystem, including technologists, founders, investors, and researchers across the climate tech startup ecosystem. The summit will include two days of main-stage sessions featuring noted climate founders and investors, as well as breakout sessions with more than 20 major climate investors. Don’t forget to register! (It’s free!).

Extreme Tech Challenge (XTC) is the world’s largest startup competition that aims to help startups change the world for the better. Founded by Silicon Valley venture capitalists, Young Sohn and Bill Tai, their mission is to empower entrepreneurs building innovative technologies that improve the world.


One more thing: Separately from the Startup Innovation Challenge, SOSV is also hosting the invitation-only Climate Tech Matchup for founders and VCs that was so successful last year, attracting 500 investors and 1500 founder-investor meetings in 2022.  Applications for the matchup will open in August. 

IndieBio NY Demo Day: Watch 10 pitches from Batch 5

Demo Day 2023

Yesterday, IndieBio presented New York Batch 5 for an online Demo Day, an event that highlights new companies in the IndieBio fold. Unique to this year’s Demo Day was the location of the event: IndieBio’s new 25,000 sq. ft. office, lab, and event space at 7 Penn Plaza in New York City. 

IndieBio’s managing director Stephen Chambers led Demo Day, introducing the 10 companies in New York Batch 5, and SOSV managing general partner Sean O’Sullivan offered closing remarks on the companies’ potential impact on climate change, human health, and beyond. The founders of each company presented on stage about their plans to address existing challenges within the food industry, biomaterials, femtech, industrial biotech, therapeutics, and drug discovery. Learn more about each of the New York Batch 5 companies below—and be sure to check out their Demo Day presentations, too!

BioFluff

BioFluff is developing a biodegradable, cost-competitive, and high-quality alternative to animal and synthetic furs for the luxury clothing market. As the world’s first completely plant-based fur, BioFluff’s product is free from plastic and GMOs and is sourced from organic renewable fiber plants—meaning no pesticides, herbicides, insecticides, petrochemicals or unsustainable raw-oil derivatives in the manufacturing process.  

FluoSphera

FluoSphera is devising the first-ever liquid microphysiological systems (MPS) to revolutionize drug discovery, increase the success rate of clinical trials, and propose superior alternatives to animal experimentation. FluoSphera’s MPS mimics the communication between multiple human organs in vitro to more accurately predict the effects of candidate drugs—even before they reach the first patient—and pinpoint the most promising drugs.

Edge

Edge is solving one of the main roadblocks to the cultivated meat industry: finding a growth factor without extensive downstream processing. To do this, the company has created a novel bioprocess using animal cell factories that self-supply authentic growth factors. Edge’s method doesn’t require the isolation and purification steps of recombinant protein production (common in fermentation and molecular farming), significantly cutting costs while providing a constant supply of growth factors and reducing contamination risk. 

Forte Protein

Forte Protein is building a technology platform to sustainably produce bio-identical animal proteins such as ovalbumin and lactoferrin within plants like lettuce or kale. These proteins can be ingredients for non-GMO, vegan, kosher, and gluten-free supplements, gels, dairy and meat alternatives, and energy drinks. The company can also repurpose their plant waste as feedstock, fertilizer, biofuel, extracts, and other plant compounds to bring in an additional revenue stream. 

Atlantic Fish Co

Atlantic Fish Co is developing cultivated seafood that is both delicious and sustainable. The company harvests cells from fish, feeds nutrients in a bioreactor, and uses scaffolding to yield the texture of a whole fish filet. The resulting seafood is real fish-cell-based cuisine without the environmental and health consequences of conventional fish farming.  

Vader Nanotechnologies

Vader Nanotechnology designs new organisms and enzymes that break down plastic and chemical pollutants. Using automation, image processing and computer vision, and analytical chemistry, the company chooses organisms that can grow on plastics and chemicals that resist natural degradation and optimizes these organisms’ performance. 

Vitarka Therapeutics

Vitarka Therapeutics has developed EndoPore, a drug-delivery solution that uses synthetic biology to create pore forming proteins (PFPs) for targeted, cytosolic delivery of RNA therapeutics. Vitarka Therapeutics is exploiting PFP’s naturally-evolved mechanism of endosomal escape and its mechanism of stabilizing RNA.

Pneuma

Pneuma is creating a living, breathing textile, OXYA, seeded with microalgae that consumes carbon dioxide and produces oxygen. By engineering living materials that can photosynthesize and make oxygen, Pneuma hopes to promote healthier, more sustainable apparel and home-ware industries. 

Bioeutectics

Bioeutectics is making green solvents mainstream with its non-toxic, biodegradable, and sustainable solvents. Using a combination of eutectic technology and green chemistry, Bioeutectics is customizing products for industries spanning food, pharma, and personal care. 

AIMA

AIMA is innovating period pain management with CBD-infused products—the first to be tested being OVY. This vaginal suppository acts as a safe and effective pain management solution for menstruators with dosages based on personalized pain response.

Here are videos for all the sessions at the 2022 SOSV Climate Tech Summit

Title graphic for the SOSV Climate Tech Summit

We just completed our two-day summit (Oct. 25-26), which included 22 panels and fireside chats with leading founders, VCs, researchers and policy experts.

We had a great turnout, with more 6700 people registered this year, from 88 countries. We recognize that the event was not available during waking hours in much of the world so we wanted to make the sessions available on-demand right at the show’s conclusion.

The full recordings for each day are here for Day 1 and Day 2. You can find each session listed with its corresponding YouTube link below.

Welcome to SOSV Climate Tech Summit 

Watch Session on YouTube

SPEAKERS


How to invent climate unicorns

Watch Session on YouTube

MIT’s Dr. Yet-Ming Chiang has an astonishing knack for converting science into indispensable climate tech companies, including unicorns Form Energy and Desktop metal and rising star Sublime Systems. How does he do it? And what spaces does he see in the vast materials space?

SPEAKERS


Who writes those early checks, and why?

Watch Session on YouTube

In venture investing, early checks are usually the riskiest, and when it comes to deep tech climate startups, the risk is especially high. How do VCs who put early capital to work decide which founders to back, and in what climate sectors? How do they coach their founders and what do they expect in the early years?

SPEAKERS


Nuclear fusion, the forever promise

Watch session on YouTube

More than $5 billion in venture capital has heated up the race to produce commercially viable, nuclear fusion power – as soon as the 2030s. Helion Founder David Kirtley is one of the frontrunning founders in the race to make this near perfect energy technology a reality.

SPEAKERS


Is this geothermal’s moment?

Watch Session on YouTube

One cure for the limitations of solar and wind is the geothermal potential right under our feet, but the US produces less than 1% of its energy from geo. Dandelion and Quaise are working on solutions that will transform access to geothermal’s untapped possibilities.

SPEAKERS

  • Carlos Araque, Quaise Energy, Co-Founder and CEO
  • Kathy Hannun, Dandelion Energy, Co-Founder and President
  • Moderator: Candice Ammori, Founder, The Climate Vine

Who writes series A & B checks and why?

Watch Session on YouTube

The value of Series A investments in climate tech grew 76% to $5.3 billion between 2020 and 2021, while Series B investments doubled to $8 billion. Khosla, USV and EIP are in the thick of the competition to write those checks. Hear how these investors pick their bets. 

SPEAKERS


The Swedish Way: How Vargas Built NorthVolt, Polarium and H2 Green Steel

Far from Silicon Valley, Carl-Erik Lagercrantz co-founded Vargas Holding and launched three notable companies to re-shore critical clean industries. Can their novel playbook apply to other industries and geographies?

Watch Session on YouTube

SPEAKERS


When the sun and wind aren’t around, what then?

Watch Session on YouTube

Globally, carbon offsets are traded on ETS generally well below $100/tCO₂. The trouble is that most solutions for carbon capture and storage still cost well above $100 per ton. Who will close the gap? We’ll hear from two founders who believe they have the technology and business model to get there. And to keep the conversation grounded, we’ll run it by a scientist whose life’s work is to figure out carbon capture.

SPEAKERS


What’s the right role for Uncle Sam?

Watch Session on YouTube

Whether it’s consumer incentives for electric cars or FDA approvals for alternative proteins, startup founders often need help from legislators and regulators to succeed. What is the best way for the government to help innovators succeed without playing favorites or making the wrong bets? 

SPEAKERS

  • Sarah Hunter, X (Formerly Google X), Director of Global Public Policy
  • Dr. Jesse Jenkins, Princeton University, Assistant Professor and Macro-scale Energy Systems Engineer
  • Robin Millican, Breakthrough Energy Ventures (BEV), Director, U.S. Policy and Advocacy
  • Moderator: Danny Crichton, Lux Capital, Head of Editorial

The DoE’s $40 billion “bridge to bankability”

Watch Session on YouTube

The US Department of Energy has a $40 billion loan facility to assist critical, sustainable energy projects. It’s Jigar Shah’s job to make the call on those loans, and he’s studying 77 applications. So how does that work, exactly?

SPEAKERS


A rockstar VC returns to “fix” climate and secure humanity’s future

Watch Session on YouTube

Legendary software investor Chris Sacca came out of retirement to raise more than $1 billion for his new climate fund, Lowercarbon, and build a team focused on climate science and investing. In no time, they’ve invested in more than 60 companies. What has Sacca learned so far?

SPEAKERS


Termination Shock, the near now

Watch Session on YouTube

Neal Stephenson’s latest, “Termination Shock,” is a riveting take on what the near-future life might look like when climate goes badly sideways. What does the sci-fi legend really think about humanity’s ability to address the challenge?

SPEAKERS


Can direct air capture save the day?

Watch Session on YouTube

The Biden climate package set aside billions to support carbon dioxide removal technologies, and Climeworks is spear-heading the direct air capture (DAC) globally, with the world’s largest DAC facility and storage installation in operation. Co-founded by Dr. Christoph Gebald, the scale-up is on a journey to climate impact at scale and strives to inspire 1 billion people to act and remove CO2 from the air. How far has the technology developed, and what does it take to scale it up as fast as required? How does direct air capture become a real business? 

SPEAKER


Carbon reduction three ways – forests, algae and making things

Watch Session on YouTube

Our planetary ecosystem scrubs C02. Can we help forests and ocean life do better? Or borrow nature’s photosynthesis to make atmospheric CO2 into valuable materials and fuels? These three founders are putting nature to work to reduce C02. 

SPEAKERS


Singapore is way ahead. They have to be.

Watch Session on YouTube

Climate change is an existential threat to Singapore, but unlike most small island states Singapore has the financial resources, technological capability, and governmental focus to address climate change on fronts. At the center of that effort, is Minister Grace Fu.

SPEAKERS

  • Grace Fu, Singapore, Minister for Sustainability and the Environment.
  • Moderator: Ben Joffe, SOSV, Partner

Can plants and AI curb the livestock GHG problem?

Watch Session on YouTube

Can plants replace the animals in our diet? Livestock accounts for 14.5% of global greenhouse gas emissions, and unicorn Notco has taken on dairy with a plant-based milk and is using an AI-driven platform to design an arrray of plant-based protein replacements. How far can Notco go, and how fast?

SPEAKERS

  • Matias Muchnick, NotCo, Founder and CEO
  • Moderator: Amanda Little, Columnist for Bloomberg and Professor of journalism and science writing at Vanderbilt University

When farming goes indoors

Watch Session on YouTube

Food and climate have a tricky relationship. Unsustainable agriculture is a big GHG source; worsening climate risks food production and famine. Food production in sustainable facilities, If they can scale economically, may be an answer, or so these founders hope.

SPEAKERS


Alternative protein scale-up? There’s a corporate for that.

Watch Session on YouTube

Startups in food tech face daunting capital and know-how challenges to scale production. That’s why brewer giant AB InBev created BioBrew, a new division to build precision fermentation operations (beer, get it?) for the likes of partner The Every Company. How did that collaboration come to pass and how is it working?

SPEAKERS


How can ag tech feed and save the planet at the same time?

Watch Session on YouTube

Industrial agriculture is a major contributor to the greenhouse gasses that drive climate change, and at the same time our ever hotter planet is putting food production everywhere under an unprecedented strain. The challenge for ag tech pioneers is to address both challenges at once.

SPEAKERS

  • Josh Silverman, Aromyx Corporation, CEO
  • Matias Viel, Beeflow, Founder and CEO
  • Toni Wendt, Traitomic – Carlsberg Group, Head of Technology Development and Operations
  • Po Bronson, SOSV General Partner and IndieBio SF Managing Director

A $3.6 billion investment company for future generations towards a net zero world. Singapore, of course.

Watch Session on YouTube

Temasek, a global investment company, was already a global leader in climate tech investing when in June it announced the launch of GenZero, a wholly-owned $3.6 billion investment platform company dedicated to accelerating decarbonisation globally. GenZero CEO Frederick Teo will address how GenZero plans to put that capital to work. 

SPEAKERS


New ways to make materials, minus the carbon.

Watch Session on YouTube

So much of what we take for granted, from plastic, to wood to concrete and steel involve processes that contribute mightily to GHG emissions or other unsustainable processes. These three founders are walking the fine line between new green technologies and promising commercial replacements for legacy approaches. 

SPEAKERS


Welcome to climate tech investing

Watch Session on YouTube

In little more than a year, 72 new climate venture funds and $13 billion in fresh capital pulled up to the climate startup ecosystem. More capital is great for the sector, but how do these new general partners see the opportunity? Which stages and categories do they like? 

SPEAKERS


SOSV Climate Tech Summit Closing Remarks

Watch Session on YouTube

Where do we go from here?

SPEAKERS

Thank you for joining the SOSV Climate Tech Summit!

Kutanios: A Novel Skin-protective Ingredient for All Types of Skin Care

Kutanios produces a peptide-based ingredient that prevents sun damage and aging via topical application. The novel mechanism stops skin cells from degrading the surrounding collagen and promoting inflammation, common causes of sun-damage related aging. Not only does this peptide protect against sun and skin damage, but their ingredient is biodegradable and safe for the environment.

Watch Kutanios present at IndieBio NY Class 3 Demo Day

We spoke to CEO Irina Miller, Ph.D., and CSO Norman Miller, Ph.D., about the discoveries that underlie Kutanios and their vision for skincare’s future.

How did your passion for science evolve into a biotech startup?

Irina: My passion is for science. My passion is for science for medical science, for biophysics, and biochemistry, for science. I studied medicine first, and became a physician. Then my PhD courses were in the field of cardiovascular biochemistry, studying the effect of damage to vascular walls and protection from this damage. 

Since then, I realized that the mechanism underlying their vascular or damaged was collagen exposure, and it’s very similar with what’s happening on the skin surface. Skin is the largest organ in the body, but the capacity of protecting itself is very limited.

We are developing science-based innovative ingredients to detoxify the skin from the harmful effects of solar radiation, tobacco smoke, pollution and other harmful chemicals.

What is the scientific basis of Kutanios’ technology?

Irina: We decided to start Kutanios, which has a totally new approach to skincare, to detoxify the skin of harmful chemicals, all causes of skin aging, dermatological conditions, including acne, and even more severe damaged skin cancers. 

The idea is science based, and it’s based on 3 big discoveries of Professor Norman Miller, who discovered the role of HDL (good cholesterol) in prevention of cardiovascular disease. 

His second big discovery is that a small part of HDL proteins can bind and detoxify toxic lipid peroxides which are damaging to the cells and its surroundings. 

His third discovery was that the concentration of these toxic lipid peroxides in skin is much greater, more than 10 times greater compared with concentration in blood.

How do lipid peroxides affect skin health?

Norman: So the lipid peroxides that are formed in skin, by sunlight and atmospheric pollutants and tobacco smoke, have several effects on skin cells and on the proteins between the skin cells that are important in our appearance. 

For example, there’s a protein in skin called collagen that is important for giving the skin strength. There’s another protein called elastin, which gives elasticity. The cells have many different functions in skin. And also the DNA is important. All of these things that I’ve mentioned can be damaged by lipid peroxides. And there’s good evidence that they all contribute to the aging process.

How do the Kutanios peptides stop lipid peroxides from damaging skin?

Norman: Our peptides enter skin, they bind these lipid peroxides, and act like magnets or sponges to sort of mop up the lipid peroxides. That prevents them from having the harmful effects on the proteins and on the cells and on the DNA. 

At the same time, the the peptides stick to the surfaces of the cells. And so when the cells are lost from the skin by the normal processes, they take the peptide and the lipid peroxides with them, so very little lipid peroxide will enter the blood, most of it will be lost. 

It’s a kind of a cleansing process, a detoxification process, that will protect the skin from the harmful effects of these lipid peroxides.

Why is it important to create novel skin health solutions?

Norman: The role of lipid peroxides in skin conditions has been completely neglected. It’s been known from scientific research for many years that lipid peroxides develop in the skin; it’s been known also that lipid peroxides have bad effects on cells. But nobody has brought these two together. 

At the moment, in the skincare industry, there is no ingredient that specifically targets these chemicals, even though we know they’re being produced all the time, that they’re increased by sunlight–and by all sunlight, not just by ultraviolet light by all sunlight–they’re increased by tobacco smoke, and they’re increased by all common atmospheric pollutants. 

We know that they’re being produced, but there is no ingredient, surprisingly, in skincare in the skincare industry at this point, which targets them. And our product will be the first one.

How do you imagine skincare and skin health products will evolve?

Irina: My envisage of the future is that application of creams will be topical and very targeted to the particular needs of every individual. And that’s what we’re aiming for. 

At the moment, our ingredients, our new component, is also very targeted, and it’s targeted to specific harmful particles, which are produced in more than polluted environments. In the future we will be targeting particles and particular problems within the body, within cells.

AlgiKnit moves its eco-conscious materials production to NC for next stage of growth

Vegconomist reports “Kelp Yarn to Be Produced at Scale as AlgiKnit Opens North Carolina Hub.” The article says AlgiKnit (SOSV RebelBio 05) “aims to cut the fashion industry’s carbon emissions in half” with its eco-conscious biomaterials created from kelp. The company’s new facility in Triangle Park, NC, will house R&D, business, and manufacturing as it prepares to commercialize, scale production, and accelerate growth.

Regarding AlgiKnit’s choice for their new location, co-founder and COO Aleksandra Gosiewski says, “North Carolina’s Research Triangle area is becoming a hub for big tech and life sciences, and we look forward to collaborating with other companies focused on scientific innovation and disruption. We’re also excited for the opportunity to tap into the deep and diverse pool of talent and knowledge in the area to strengthen our scientific and engineering divisions.”

Bio-textile firm Bucha Bio lands new funding

Image courtesy of Bucha Bio.

In its story “Bio-textile firm Bucha Bio lands new funding,” Bio Market Insights reports that the New York-based SOSV IndieBio startup Bucha Bio (IBNY02 2021) has raised $550,000 to accelerate production of its planet- and animal-friendly material Mirai. The creation of Mirai involves fermenting nanocellulose and blending the resulting leather-like material with natural fibers, resulting in what the group describes as “advanced performance and luxuriously soft handfeel.”

Bucha Bio’s mission takes aim at the high-polluting leather industry which produces over 4 million tons of solid waste per year. With this funding from New Climate Ventures, Beni Venture Capital, Lifely VC, QKZ Design, and MicroVentures, Bucha Bio plans to introduce Mirai to companies across the fashion industry as “an ethical alternative to animal leather products including epoxy, vinyl, and latex.”

SOSV HAX alum Opentrons raises $200 million, reaching $1.8 billion valuation

IndieBio’s fellow startup development program, HAX, just recorded a big win, as reported by Bloomberg in “SoftBank Invests in Robotic Company Behind NYC Covid Testing. HAX alum Opentrons Labworks Inc. raised $200 million in round led by Softbank to reach a valuation of $1.8 billion. The Brooklyn-based, laboratory robotics company used its automated systems to decrease Covid-19 testing result times from 14 days to 24 hours and reduced test costs from $2,000 to $28. Opentrons “has grown to support a community of more than 1,000 scientists and 46 countries.” Other participating investors in the funding include Khosla Ventures and ex-Pfizer Inc. CEO Jeff Kindler.

“Biology opens the door to solve many of humanity’s grand challenges. For far too long, scientists and clinicians have been locked-in by slow, expensive, and overly complex lab solutions that underpin their work.” —Brennan-Badal, Chief Executive Officer, Opentrons

Nyoka Design Labs helps engineer the world’s first bioluminescent gaming PC

In this video from from PCGamesN, “World’s first bioluminescent gaming PC is impractically beautiful,” Linus Tech Tips and SOSV IndieBio startup Nyoka Design Labs collaborated on a project to make a gaming PC glow while it cools. Nyoka is a startup that aims to use bioluminescence as a cleantech alternative to the air-polluting fluid contained in glow sticks. The PC cooling project was a fun side challenge. But don’t try this complicated build at home, kids. As explained in the video, “the fluid’s micro-particles could also damage your CPU block. So, while bioluminescent PC lighting is mesmerising, it’s unlikely to replace your usual coolant.”

SOSV HAX’s Opentrons is center stage in New York City’s rapidly growing life sciences sector

NY1 published a segment “How New York City is expanding its life sciences industry” featuring Opentrons, an SOSV HAX startup. If you’ve ever taken a COVID-19 test in New York City, it’s highly likely your sample was processed by Opentrons, a Brooklyn-based company that builds lab robots for biologists. And this is by design—pegged as a key to New York’s economic recovery, the life sciences industry has received $1 billion investment and support from the NYC Economic Development Corporation (EDC) and Mayor Bill de Blasio to attract new businesses, capital, and jobs. This week Opentrons received a $200M investment led by Softbank, boosting the company’s value to $1.8B.

“Beyond all our traditional industries, beyond the tech community that has grown so successfully in New York City, we need to build up life sciences. This is the future.” —New York City Mayor Bill de Blasio

Inside Hermès, where MycoWorks mycelium leather will soon join the product line

Vanity Fair recently published a photo essay, “Inside the Hermès Workshop That Makes Its Iconic Bags,” that discusses the iconic designer’s plans to use IndieBio alum’s MycoWorks‘ “Fine Mycelium,” in an upcoming line of products. From Alexis Cheung’s piece:

“Despite this staunch adherence to tradition, Hermès will introduce a decidedly modern material this fall: mycelium leather. Developed in collaboration with the San Francisco-based biotech company MycoWorks, this “Fine Mycelium,” coined Sylvania by its creators, derives not from cattle but from mushrooms. Fournier insists that its quality and durability meet the same high standards of traditional leathers and that the material continues Hermès’s long legacy of innovation—it was, after all, Thierry’s grandson Émile-Maurice Hermès who introduced the zipper to handbags in 1922.

“We strongly believe that we should not oppose new technology with what we do with the hands and tradition,” says Fournier. “Both are compatible.” Plus, he adds, “It’s a fantastic opportunity for creation, to play with new materials.” (For now, this particular play is reserved for the Victoria handbag from the autumn/winter 2021 collection, constructed at a workshop of its own.)”

SOSV Climate Tech 100 Startups value grows by 44% in 5 months

On Earth Day, April 22, 2021, SOSV published its SOSV Climate Tech 100 list, a collection of the top climate tech companies in our portfolio. The list was notable because the companies up to that point had raised $1.85 billion from investors (including $89 million from SOSV), and had a market cap in aggregate of $5.65 billion.

Techcrunch reported on those numbers and commented that SOSV’s “planetary health’ mission was “paying off.” We subsequently published detailed analyses of the list’s founders and investors, and we announced an Oct. 20–21 event called the SOSV Climate Tech Summit, aimed at helping the climate ecosystem move faster.

Now it’s nearly five months later, and the Climate Tech 100 list financials are due for an update. Thanks to the strength of the companies on the list as well as a powerful surge in climate tech venture investing, the financials for the 100 have taken a big step forward.

  • The SOSV Climate Tech 100 aggregate value has jumped from $5.7 billion to over $8.1 billion, an increase of 44%.
  • Total investment in the 100 increased $508 million to reach $2.36 billion, an increase of nearly 28%.
  • SOSV topped off its investments with $13.8M to reach $103 million, an increase of 15.5%.

Join IndieBio at the SOSV Climate Tech Summit

IndieBio will be a big part of the SOSV Climate Tech Summit on Oct. 20-21. The event is virtual and free. The summit’s purpose is to convene the founders, investors, technologists, corporates, media and anyone else keen to understand and accelerate the climate tech startup ecosystem. Read more about the event here. Register here.

Several IndieBio folks are a part in the programming, including Po Bronson, Arvind Gupta, Pae Wu and Gwen Cheni, and so are IndieBio alums like Dr. Uma Valeti, CEO and founder of UPSIDEFoods. There are also many speakers on the main stage, the majority in fact, who are not from SOSV and represent some the best minds at work across startup climate tech. You can see all the speakers announced so far here.

In order to help both founders and investors, one very special feature of the summit is a series of 18 breakout sessions dedicated to early stage investors, incubators and government agencies that have a strong track record working with pre-seed and seed climate tech startups. They range from SOSV’s IndieBio and HAX, to TechStars, The Engine, DVCV, MassChallenge, the NSF and ARPA-e, Greentown Labs, Energy Impact Partners, and more. Look for the full list to be published soon. The sessions will be led by senior partners at those outfits and focus on what they have to offer climate tech founders. The breakouts will be staged one after another so that founders can easily catch them all. The sessions will also be live with plenty of time for audience questions.

Finally, the summit will offer an Expo that features some of the top climate tech companies from all the programs that are offering breakout sessions. SOSV is offering Expo spots to all the companies that are part of the SOSV Climate Tech 100.

The SOSV team is working hard to produce a great event that really benefits everyone in the climate ecosystem who is working hard on breakthroughs that will help address climate change. Please join us at the event. It’s free and virtual. Register here.

Allozymes looks to upend chemical manufacturing with rapid enzyme engineering and $5M seed

Part of the complex process that turns raw materials into finished products like detergents, cosmetics and flavors relies on enzymes, which facilitate chemical transformations. But finding the right enzyme for a new or proposed drug or additive is a drawn out and almost random process — which Allozymes aims to change with a remarkable new system that could set a new standard in the industry, and has raised a $5 million seed round to commercialize.

Enzymes are chains of amino acids, the “building blocks of life” among the many things encoded in DNA. These large, complex molecules bind to other substances in a way that facilitates a chemical reaction, say turning sugars in a cell into a more usable form of energy.

Carbix: Turning CO2 into Stone

To meet the challenge of our climate crisis requires humanity to reinvent industries on a global scale. Eliminating emissions as fast as possible is critical, and it has become clear we also need to pull CO2 out of the atmosphere, as well as prevent more from collecting. As we do this a new question emerges, where do we put it? Carbix is working on a solution for long-term carbon storage while simultaneously decarbonizing cement, a notoriously hard to clean up industry. I sat down with Quincy, their CEO, to find out how.

 Enhanced Weathering is a term that more and more people are learning about as climate science goes mainstream, yet its geological nature feels so distant to human timescales. How are you learning from nature to take on the challenge of producing carbon negative cement?

 Nature has perfected the capture of CO2, but over timescales that are far beyond the urgency of humanity’s climate crisis. We know from extensively studying the natural carbonation cycle of minerals like calcium silicates and olivine that certain conditions must co-exist in order for CO2 to turn to stone. At Carbix, we have distilled this biological process to engineering parameters optimizing heat, pressure, mixing rate, UV-C and CO2 injection.

We don’t have to pipe CO2 underground near hydrothermal veins to achieve optimal heat and pressure for carbonation. To further speed up the process we learned from the chemistry of the upper atmosphere that UV-C light energy can enhance carbonation reactions at lower pressures than what may be possible on Earth. The Big Reactor in the Sky (Sun + upper atmosphere) has taught us the role that UV-C light plays in generating hydroxyl radicals (OH) to accelerate the carbonation process.

Our X1 reactor has been designed from day one to give us control of these key variables.

 Can you tell us about the X1? What does it look like to potentially deploy in the world to meet the incredible scale of construction?

The X1 reactor scales up to about 150m3 (cubic meters) to meet the demand of the cement and concrete industry. In operation we need a few inputs. Certain minerals, like olivine, have the greatest carbon sequestration potential so we’ll be finding sources with the smallest carbon footprint possible to ship in. The other critical input is CO2, which we can get by partnering with direct air capture (DAC) technologies or at lower concentrations through direct smokestack effluent. While effluent has lower CO2 concentrations, it actually is still a good feedstock since we then forgo DAC costs and capture other pollutants which otherwise end up in our air and lungs. We also will need energy and water, which we hope to get from renewable sources and lower costs with water recirculation and energy recovery devices.

 Ooh sounds sci-fi!

The X1 reactor looks like something out of an “Alien” movie series so I guess that makes it Alien tech hahah.

The design keeps in mind that size and speed matters when it comes to tackling the emissions from cement making, which accounts for up to 8–10% of GHG global emissions! It’s a huge climate issue, and a huge market as cement and concrete products are a nearly $330B annual global market. The use of concrete, as well as heavy CO2 emitters, are distributed across the globe so the X1 can be deployed as a single unit or scaled to multiple to match the rate of emissions or concrete production needed.

 You’re a young company, yet already in deep conversations with large companies about working together. Can you share more about the appetite in industry for climate solutions?

 The interest is strong from the industry to reduce their GHG footprints. The public and private incentives are expanding but already moving the industry in our direction. The US and other major governments have incentives through the 45-Q and LCFS standard (California) to help pull the industry in. The call to action — in a country like Japan for example, are mandates that require cement companies to reduce their GHG footprint by 30% by 2030, no exceptions.

These incentives are important because the scale we’re talking about is so big. One potential customer, Dangote-West Africa as the example, processes nearly 6000 metric tons per day of clinker at one plant!. That’s an annual rate of nearly 2.0Mt (megatons). Other cement plants have production rates of about half that at 1MM(megaton per year), like Mitsubishi.

We’re in talks and even sharing ideas on which product makes sense to pilot. Dangote proposed the idea of creating tiles with carbon negative carbonates and oxides so that every one of their customers can buy carbon negative products. Imagine that — individuals and businesses can beautify their homes and offices and become climate champions while effortlessly installing tiles. We think it’s an amazing idea as it lets consumers vote with their dollars to act on climate change.

 Finally, tell us about yourself and your team. What inspires you all to work on this problem?

 We’re all passionate about protecting our natural environment (and the humans in it) and have been so for most of our lives. It’s an internal drive. We’re also technologists. So for us the pathway to healing the plant comes through technology, like the X1 reactor. Inspiration to take this direct path to removing CO2 from the atmosphere and creating products like cement and concrete is driven in part by the scale of impact we can make. What we’re doing works in parallel with the transition to a clean energy infrastructure by giving the planet some “breathing room“ until the transition is complete.

Myself, I have over 10 years in clean energy design engineering, with a previous finance background. Dr. Vintit Dighe has also been in the cleantech space. He is an expert in fluid dynamics, wind energy, and machine learning. He‘s developing fluid and chemistry solvers to guide enhancements to the X1 reactor kinetics. Samip Desai has a clean energy and finance background in cleantech and is actively working in business development to bring in multinational cement and concrete ready mix producers.

 

AsimicA: Raising the Bar for All Biofermentation

In May 2020, a McKinsey report found that the global bioeconomy is slated to become a $4 trillion gold rush as synthetic biology’s promise to make high-quality, sustainable products gains traction. These products range from food to textiles to medicines, and biofermentation is the manufacturing process that makes all of it instead of having to extract them as natural resources. Today, we sit down with Nik Mushnikov, CEO and co-founder of AsimicA, whose technology promises to solve the bane of biomanufacturing: low yields that formerly could not compete with traditional manufacturing methods. Using his invention, dubbed “microbial stem cells,” Nik thinks he can achieve multi-fold yield increases in product, and keep the bioreactors running longer.

Ok first off, what is a biofactory, and why is everybody talking about them?

Microbial biofactories are basically reactions in which we use microbes (like a bacteria or yeasts) to make the product we want. This was a solution when the product itself was biological, like insulin, and we didn’t know how to engineer a synthetic process that is smarter than a living cell.

Now with genetic engineering, biofermentation is becoming more popular as we are learning how to make other products that aren’t even biological in nature. For example, we used to make plastics with petrochemicals because it was cheaper and more efficient, but the synthetic biology field is learning they can engineer microbes to make specialized plastics.

Wow, so do you think that biofermentation will be used to make everything?

I think petrochemical or chemical synthesis is still more efficient, but it comes with problems like relying on oil as a feedstock and toxic pollutants as a byproduct.

Microbes, on the other hand, have the potential to produce any chemical compound in a sustainable manner, using renewable resources, if you can engineer it correctly. If you factor in the externalities, I think that traditional chemical synthesis is starting to lose its edge, especially when it comes to specialized products.

Why isn’t everyone using biofermentation right now?

Microbial biofactories can’t operate non-stop. As they divide, every generation becomes less productive and you have to restart the entire batch. Every restart is expensive in terms of expensive, specialized labor and downtime. We’re talking several days. Economically this doesn’t fare well, and we need to find ways of making them more efficient and more productive.

People have tried ways to make the microbes live longer or more resistant, but evolutionary genetics eventually catches up and drives the population to become sickly and unproductive. The solutions in the past have been… lackluster.

So I guess that’s where you come in! How are you solving this problem of low yields?

Instead of trying to make the microbes live longer, we found a way to repopulate and replenish biofactories with a fresh generation of microbes during the batch. We’re doing it using our innovation of “Microbial Stem Cells.”

In our bodies, our stem cells essentially replenish the cells in our tissues so that they stay functional for decades, much longer than individual functional cells can live.

Our idea is similar to that — microbial stem cells are constantly replenishing the fraction of productive microbes in the bioreactor. It is a way to bring up new young and strong “workers” to the factory, so to speak. We’ve published mathematical models that show that this replenishment strategy would result in a 2–4 fold higher number of productive microbes in the bioreactor, which translates to higher productivity per reactor, and longer batch runs.

The effect that microbial stem cells can provide on bioreactor productivity can significantly increase the profitability of bio-manufacturing.

How did you come upon this insight? Was this always something you engineered with biofermentation in mind?

I was always very intrigued by the potential of biofactories for manufacturing all sorts of chemicals: pharmaceuticals, fuels, advanced materials, and so on. And I wanted to be involved in designing new strategies for bio-manufacturing using microbes.

When I started my PhD, I had a couple of projects, focused on increasing yields of microbial fermentation. The idea of realizing stem-cell-like behavior in industrial strains of microbes came out from previous fundamental research insights made by my advisor, Dr. Grant Bowman. He was studying a phenomenon of asymmetric cell division in some unique bacterial species. Sometimes their cell division diagram resembles the division of stem cells. Certainly, these species are not applicable to the industry. And molecular mechanisms underlying their asymmetry are way too complex to just copy them.

What we’ve done is that we identified a minimal set of key components that can induce asymmetric division in other species. We borrowed them from several different bacteria and transferred them into E. coli, and our Nature Chemical Biology paper demonstrated that we can indeed induce asymmetric cell division and program differentiation in different cell types.

What kind of products are you able to make and is there any limit to what yields you can increase?

We just got results last week that we are able to make several products from pharma, food, and the cosmetics industries. These are just proof of principle experiments to demonstrate how versatile our platform is. We’re thinking of experimenting with fuels next which are highly toxic products for microbes to make — again, just to flex how broadly applicable our “microbial stem cell” technology is.

Everyone who does biofermentation is dealing with the exhaustion of microbes, and I don’t think there is a better solution than ours to improve yields and lengthen batch reactor runs.

So the sky is the limit in terms of products, but what about the cells? Are you limited to E. Coli?

That is a very good point. Of course, not all biofactories are using E. coli cells as their workhorse. We see a fairly straightforward way to transfer out technology to other species of bacteria. We’re already working with Bacillus subtilis, which is what most of our industrial partners are using. Transferring our ideas to eukaryotic microbes, yeast, and fungi, would be a more R&D extensive project but we’re confident we can get there.

How does a potential partner incorporate their technology into their existing manufacturing process?

Biotech companies won’t need to change much in their manufacturing processes. What we do is a strain engineering service, which changes how the culture of cells in the bioreactor behaves, enabling microbial stem cell properties. But it doesn’t change the production process itself. Simply speaking, our partners would do the same things they were used to, but their strains after AsimicA modified them would have higher productivity.

Why did you study microbiology?

I started to learn about the broad potential of microbes back in high school and my interest kept growing throughout college. Each microbiology course taught me that the potential of microorganisms is unlimited. My first research project was in yeast genetics. I’ve learned some methods and practices of working with microbes, but the research scope was rather fundamental and driven by the needs of clinical biology, whereas I was more interested in applied microbiology.

I finally found The Bowman Lab in Wyoming to do my PhD, where my interests were more aligned with applied bio. There, I could perform my research studies, keeping in mind that we’re creating new tools, new technologies that can be directly applied to the industry. That research dynamic is closer to my heart, and launching AsimicA where I can take the application to industry has been one of the most rewarding experiences I’ve ever had.

What good would this do for the world?

The world needs cleaner economics. Humanity is facing existential challenges, and although we are getting closer to solving those challenges we still have a long way to go and perhaps what will move the needle the most is to change the way we make things — using renewable resources, reducing pollutants, and dropping down our emissions. Our technology can facilitate biofermentation in becoming the primary method for the production of the vast majority of chemicals. Just imagine how much dirty production we can push out if we can increase yields by 2–4 fold across the entire industry. That’s what I think AsimicA could do for the world and that’s the future that I want to help build for us.

Kraken Sense: Pathogens Have Nowhere Else to Hide

A considerable amount of effort is taken to make sure that the water that is used to process and rinse your produce is clean and clear of pathogens like salmonella and legionella. Even with all the regulations that are imposed on our food supply chain to prevent such outbreaks, we are still not impervious to these bacterial threats. The affects not only public health, but also environmental, as millions of pounds of food are thrown away because of the scare. The primary reason is that the current methods for testing are too slow and too cumbersome to alert us fast enough. We sit down with Nisha Sarveswaran to talk about her innovative platform to disrupt the water testing market.

How did you first become interested in water safety? What life experiences led you to this?

Water quality has always been a passion of mine. I was born in Sri Lanka and I knew many people who didn’t have access to clean water, so I have always been conscious of the importance of safe water access. Our continuous progress is only ensured if we can properly manage our basic resources, and having safe water is critical to everyone.

I learned about the importance of water testing while doing research on pathogen-related illnesses and food recalls. It became clear to me that the present testing methods, developed more than fifty years ago, cannot meet the water and food supply challenges of 21st century.

Why not?

The current best practice is to take a sample of water, culture it in the lab for three days and have a trained technician examine the results to determine the presence and the extend of the contamination in the original sample. With our just-in-time logistics network, the produce collected and tested today may already be in the grocery store three days from now, so the current testing methodology takes too long and too limiting in scope.

With our real-time detection methodology, we can identify the contamination issue at the source and prevent the costly recalls that we are always hearing about on the news.

What’s special about your technology?

Ours uses a system based on antibodies on a carbon nanotube, which are tiny materials with very interesting properties. With our specific treatment and manufacturing methods we are able to create thin, narrow, electrically conductive strips with embedded antibodies specific to certain bacteria and even strains. When exposed to water samples that contain the target bacteria, the electrical signal changes in a very unique way, and that allows us to detect the presence and concentration of the bacteria that we want to detect.

Because we are measuring the signals immediately as water is passed through, we can essentially detect pathogens in real time. The only limit is how fast we can concentrate the water, and how fast the antibodies bind to the pathogens to get a noticeable change in our signal. This real time signal means that you can catch pathogens before the food even leaves the door of your facility. Compare that to having to get a water sample, and literally shipping it to a testing facility.

Incredible, that must really save a lot of time and money!

Yes! People need to understand that it’s not just about how many people get food poisoning. Just think about how much perfectly good produce out there gets thrown out because of a bad apple in the market (pun intended).

How do you mean?

For example, once a bad batch of romaine lettuce leaves a facility, it’s hard to track where all of it goes after, and once a few people get sick from it, the entire industry panics and avoids romaine lettuce, which kills the prices and puts the entire romaine market in shock (for good reason). This scare translates to hundreds of millions of dollars of food wasted, which is not only an environmental waste, but also a waste considering how many people are currently food insecure.

Interesting! So this isn’t just about people getting food poisoning, you’re saying this is a much bigger supply chain efficiency problem?

Food consumption is growing rapidly with our rising population and increasing prosperity. Our resources and supply chain will become more strain and will require modern solutions to identify the potential contaminations in real-time. The sensors that are able to detect harmful bacteria, in as little time as possible, are becoming more and more important to ensure food safety.

Moreover, by detecting contamination early, we are not only able to prevent costly recalls and associated health implications, but can also significantly reduce the food waste by providing alternative utilization for food that is no longer fit for human consumption. Currently the food waste from the supply chain accounts for 6% of the total greenhouse gas emissions. Our solution will ensure that the food that is distributed is safe and thus will also reduce the food waste that happens in the supply chain due to recalls.

You’re not a one-trick pony are you? I assume you can test for multiple pathogens?

We are building a multi-pathogen lab-on-a-chip system that can detect multiple pathogens simultaneously in real time. The remarkable advantage of our approach, other than the real-time capability, is that if there are antibodies available for a certain pathogen, we can build a sensor that can detect it and add it to our list of capabilities.

This work however goes beyond simply creating new sensors. In order to ensure that the results can get to the right hands in as little time as possible we have also developed automated water sampling systems and AI based machine learning algorithms running on our cloud platform that can interpret the sensor data and send the results in seconds.

Looks like you can cover a really broad spectrum of pathogens, but how fast can you make a test for other pathogens?

We can develop a new sensor in under 2 months, for example having developed the E. coli sensor we have spent some of the time at IndieBio developing Legionella sensor. This process will only accelerate as our first sensors enter the market and the process of creating new sensors becomes more established.

So is the speed at which KrakenSense is testing going to be the new standard for water testing? Are we going to see you guys across the entire supply chain?

We are working on developing protocols to help increase food safety testing and establish our methodology as the new standard for water testing. We really think that the water testing market won’t be the same after a few facilities can test in real time.

It’s like Amazon’s 2-day shipping: Once people start to get used to the speed, they just can’t imagine going to a much slower system… likewise, we think once we have a few pilots and customers, the rest of the market will start to find their conventional way of testing really outdated, and will want to come to us. In the near future, we see our solutions being present across the supply chain from early detection on the farms, to critical supply chain points that are highly susceptible to contamination.

So what’s on your roadmap now?

We are raising the seed round to further develop the lab on the chip system, expand our detectable bacteria capabilities, and pilot our solution with several key customers that will demonstrate the concept to the industry in general. At the same time, we are developing a suite of tools that will be used in tying it all together with blockchain technology so that every supplier has constant traceability in their food supply chain in real time.

CASPR Biotech: Revolutionizing Molecular Diagnostics

The CRISPR Cas complex has been a game-changing technology for gene editing. CASPR Biotech is using the incredible accuracy of CRISPR for something different — to revolutionize medical diagnostics.

We spoke with company founders Franco Goytia and Carla Giménez.

There are a couple other prominent companies also using CRISPR-Cas systems for diagnostics. What’s different about your focus?

We are rapidly developing a device for hospitals to quickly detect if a patient has an antimicrobial resistant infection. This is a huge problem for hospitals. It’s not just that 2 million people in the US every year get infected with superbugs, or that the infection rates are even higher internationally. When a patient walks into the hospital with a fever of unknown origin, it takes between 24 hours and three days to amplify the DNA of the bacteria to determine if the patient has AMR. During that time, the patient has to be isolated, making it very stressful and expensive. For every one patient found to have AMR, nine more have to go through this isolation. And during that time, they’re started on wide-spectrum antibiotics, which makes the global problem worse.

How does your solution change that?

We can diagnose AMR infections in under an hour. Most superbugs have one of three sequences of DNA that make them resistant. We code our Guide RNA to detect those, and if they do, our Cas enzymes trigger a signal. Not only does it do so in under an hour, but does so more accurately, and cheaper.

You already have this working?

Yes, in the lab. As well, at a hospital in Argentina we have tested our system to detect infectious diseases like Dengue virus. These aren’t yet automated into a device, but our device development is going incredibly well and we are on track to begin a 510K study with the FDA in one year.

You discovered two novel Cas enzymes, one in the Cas 9 family, one in the Cas 12 family. How did you do that, and how they are different?

Existing IP portfolios held by the primary CRISPR institutions were discovered by searching through the public databases of sequenced bacteria. We went through unpublished data, collected by our partners in the extreme environments of Argentina — regions as diverse as volcanos, high deserts, hot springs, and Antarctica. In these environments, bacteria evolved unique ways of defending themselves against viruses.

We are looking at ways our Cas 12 gives us a competitive advantage. It appears to be more stable at higher temperatures, likely due to the environment where it evolved. Using it at higher temperatures may facilitate other reactions in our system, turning what’s a two-step process into a one-step process.

You’ve been able to make incredibly fast progress during your time at IndieBio. What’s been your secret?

Before IndieBio, we were running on a very thin budget. To order reagents, we might be waiting a few months until we could get the money together. At IndieBio, we got them in a day — and shouted in celebration when they arrived. We showed up at IndieBio really hungry to do science at a much faster pace, to make quick decisions. It’s been an opportunity that we’ve been thankful for, and haven’t taken a single moment for granted.

We’ve also been very inspired by the scientists who created the foundational technologies of CRISPR. The way bacteria has defended itself against viruses has existed for hundreds of millions of years. But it took great imagination to reconfigure it for gene editing. This precedent reminds us, daily, that if we work hard, at any moment we too could make more discoveries.

How big could CRISPR-based diagnostics get?

Certainly, from AMR and infectious diseases we will move into flu detection, respiratory infections, sepsis, and urinary tract infections. But the application go beyond healthcare and the hospital. Just like Google allows us to search the entire internet, CRISPR allows us to search the entire genome for any genetic code. Rapid DNA detection has applications beyond humans into pets, livestock, and plant life. Farming, biosecurity, and new domains of agriculture are all possible.

Watch CASPR Biotech pitch on IndieBio Demo Day, Tuesday June 25th in San Francisco or via LiveStream. Register here!

Clinicai: Detecting Colorectal Cancer in Your Toilet

The successful treatment of cancer lies in early detection. If we can detect it as early as Phase 1 and 2, the outcomes for therapy are significantly better. Unfortunately, symptoms oftentimes will not present until Phase 3 and 4 which limits effectivity of therapeutic interventions. Clinicai is making a noninvasive monitor for colorectal cancer with a device that hooks on the side of your toilet. We asked Chun-Hao Huang, co-founder and CEO of Clinicai, about his journey as an entrepreneur.

How did you become interested in the early detection of colorectal cancer?

Chun-Hao: During my Ph.D. I built animal models mimicking human colon disease to find biomarkers and therapeutics. I was passionate about gastrointestinal biology and learned how to monitor mice’s poop every day, dissect the intestine and colon for pathology examination, and realized that, wow, the stool is actually very closely related to our gut system. I also realized that for colorectal cancer, if we can detect early, we can even use current treatments to cure this disease and every single patient would have the chance to survive.

I have now been working in cancer treatment for more than 10 years and have seen that the field puts a lot of effort into trying to develop new therapeutics. However, a lot of diseases, especially colorectal cancer, can be solved even right now if you detect it in time. But the early detection or preventative angles are not so appreciated by the field. So that’s why I started to focus on early detection because I believe the way to actually make cancer manageable today is by monitoring your body to capture the signals at the very outset so we can apply current therapies when they would be most effective.

How did you decide to start the company and how did your team come together?

Chun-Hao: I almost was a scientist for life, and I really wanted to make a big science discovery creating a way to solve cancers. When I was at the Lindau Nobel Laureate Meeting, I heard a lot of top scientists and Nobel laureates talking about the future of medicine and how to manage diseases like cancer. The meeting inspired me but I realized it is actually a very long process from research to benefiting humanity, and I started to wonder if there is a way we can speed up this process and bring biotechnology into our daily lives.

So I asked what is the best way we can accelerate scientific discovery and also bring biotech to our daily life? It’s very interesting to me that the device we use daily is a cell phone when biotech should be the technology we are using every day. I found the answer is to start a company because you can put a lot of resources into building a product actually related to daily life. I met my co-founders Medina Baitemirova, Juan Carlos Guáqueta, and Mr. Toilet Jack Sim at Singularity in NASA Ames, and later on our CTO Dr. Ya-Ju Lin. We all have different expertise, but we share the same vision that we want to detect disease like cancer early. We wanted to make the most comfortable diagnostic platform, which in the end ended up being a toilet. We want to make people actually enjoy health monitoring.

How does your technology work? What is that key insight?

Chun-Hao: It’s pretty interesting, when we started developing this technology we hoped to achieve three things. First thing, if we want to do the most comfortable monitoring and diagnostic at home, then the technology needs to be non-invasive where the user puts in the minimum effort to maximize regular monitoring. Second is digital information; we need to have a technology which can digitalize the substance we detected. Third, it needs to continuously gather the data. The most current methodology, like biomarkers or chemical methods, are not able to do that, so we started testing a bunch of different sensors. We started with a narrow range optical sensor and in the end, we find out that there is a large wavelength hyperspectral imaging originated from space technology that can help us to gather all the information from the stool.

That’s how we started using that technology to check stool samples and that’s how our technology derived. This is not just technology we can use in the lab, or in the hospital, we actually integrated that into a prototype device that will snap on a toilet. Everyone can attach that device at home, I even tried it myself. Then it can turn on when you sit on the toilet and then gather the data. I think these will be pretty powerful in the future, not only for cancer detection but also can help detect other preventable diseases.

What lessons did you learn transitioning from scientist to entrepreneur at IndieBio?

Chun-Hao: As a scientist, every day you go to a lab, you have the experiment you want to do, but some experiments could take a couple weeks or even longer. But when you are designing your experimental plans in a startup environment, like IndieBio, you need to think about how we can leverage different resources to make this process faster. That’s actually what I really like about this environment because you’re still doing the very cool science, but you have a way to speed it up. The other thing which is quite important is that here we’re thinking of how we can actually turn our technology into a real product and then benefit people’s lives. As a scientist in the lab, you try to solve problems, but you don’t usually think that this can actually work in our daily life.Since we joined IndieBio, we have started to talk to the FDA, we talked to insurance companies, we talked to doctors, and especially to the users. We are building things they really want and also doing cool science. This is what makes being a scientist-entrepreneur great.

How do you think your success as a company will change the diagnostics industry?

Chun-Hao: I used a proteomics approach when I was in college to identify biomarkers and in the past 10 years I didn’t see that many biomarkers come into the diagnostic field. I think it’s time that we need something new to disrupt this field. Before we always relied on one or two biomarkers, but at Clinicai we believe the key is to detect signatures or patterns of the disease.

That’s why at Clinicai we are trying to get signatures and patterns from your stool or urine samples and then we use the machine learning to understand those features. I think that that will change the way we think about diagnosing. The other strong advantage we have is to actually bring the diagnostic into our home. Before it’s been pretty difficult and we find our best niche at home where our bathroom and toilet have a lot of information every day. We hope in the future everyone can really enjoy health monitoring and can live longer and healthier.

What milestones are you aiming to hit in the near future?

Chun-Hao: I want to thank IndieBio because we actually met our milestones here. We’ve built our first prototype, which we never thought about that before. The milestone for next year is to improve our prototype and then work with hospitals to install the device in hospital toilets or in patients’ toilets at home to monitor their stool signals for colorectal cancer detection. After that, we plan to go through the FDA path and we are pretty positive about that. After that, we hope our device will be in everyone’s home for future health monitoring and diagnosis.

Watch Clinicai pitch on IndieBio Demo Day, Tuesday Nov. 6th in San Francisco or via LiveStream. Register here!

Stämm: Reinventing the Bioreactor

Biology is the best platform for manufacturing cells. It can grow and produce all kinds of biomaterials very effectively. But for humans to be able to tap into this potential, we have to build bioreactors and figure out how to scale them in order to produce the massive amounts of biomaterials we need. Bioreactors technology is very different when you’re optimizing for one reaction at one liter versus 100,000 liters, but this is where Stämm comes in. They are reinventing the way bioreactors work by developing a modular microfluidic platform that controls all the physical parameters, such as nutrients and dissolved oxygen, in a controlled environment. Unlike traditional bioreactor systems, their microfluidic bioreactor scales by adding more modules to increase the size of the reactor.

We sat down with CEO Juan “Yuyo” Llamazares to learn more. (Fun fact: his name is pronounced like “shoe-show.”)

How did you become interested in building reactors?

Yuyo: My first interaction with microorganisms was to study the interaction between plants and soil bacteria called growth-promoting bacteria. I was amazed by the communication between these microorganisms, this ability to sense the environment and that the bacteria will provide to the plants. I wanted to make use of this knowledge to create a product. I quickly realized that I would have to use bioreactors, and it became clear that the whole industry is based on bioreactors. That’s when I decided that I wanted to learn more about how we create these environments to make the cells divide, or to induce synthesis of protein. That was the first time that I got to see a bioreactor. I was used to working with plants, so working with a bioreactor felt really unintuitive, given what I knew of how cells proliferate in nature. I saw a really big opportunity there to develop new approaches.

How did you come to start this company and how did your co founders come together?

Yuyo: Well, my cofounder is my cousin, so we grew up together. But also, my grandfather taught me how to brew beer when I was 12 years old, and Federico and I saw an opportunity to manufacture yeast for brewers. That was our first time making a product, and it showed us how difficult it is to build biotech facilities. Even people that have a lot of biotech experience have trouble translating their knowledge to the market because it’s hard to develop these facilities. We saw the need for a reinvention of bioreactors so they can be reproduced around the world. We actively started scouting researchers knowledgeable in microfluidics and robotics, and started seeing that we could make this a reality. Bringing the team together, for us, was key. When we communicated our vision for the company, we found that researchers were excited to join us.

It can be difficult to take the leap to make that transition and to advocate for impact and transformation in the real world, coming from an environment that gives you all the tools you need to be comfortable. In Argentina, where I am from, there’s a reason researchers choose to stay in the lab. You have a predictable career and life, and that’s reassuring. But I made a trip to Chile and I learned about spin-off companies. That was not a concept I had been exposed to in Argentina, and I decided I wanted to bring the knowledge at my university to the world through my own company.

How does your technology work and what is the key insight that you had?

Yuyo: The key insight was to try to think about how cells experience the process of biomanufacturing and identifying room for improvement in that process. We found that today, we look at biomanufacturing as a population of cells, so we try to control the cells inside our vessel as a whole. The problem is that each cell at some point is going to be going through different metabolic stages and we take the average of that complexity. With existing bioreactors, we don’t have the tools to control that complexity, so we try to create a different kind of bioreactor: if we can account for each cell’s microenvironment then we open up a way to address the cell complexity. From that insight, we turned to microfluidics because it gave us the ability to look at the individual cell, however, the problem is that it’s hard to scale microchannels. That’s what we’re addressing with a lattice that cells move through in a predictable manner, which provides consistent nutrients and oxygen.

So how do you think your success as a company it will change the bio manufacturing industry?

Yuyo: I think that more people will know what biomanufacturing is. People will recognize it as a tool to solve a problem. Right now, biomanufacturing is “hidden” in big facilities, but with our technology, it can be used by more people at a smaller scale and more people will see the power of biology to manufacture or to solve problems.

What milestones are you aiming to hit in the near future?

Yuyo: We want to manufacture the bioprocessor and sell it to companies so they can optimize their processes as they create their product. We want to prove that this novel approach is robust and that there’s a future there. So far, we have worked with CHO and HEK cells for the manufacture of monoclonal antibodies, and we have seen that in our system, individual cells perform better. This means our process is more efficient and has higher productivity per cell. One of the other companies currently at IndieBio used our system to significantly increase the productivity of their process over the course of 20 days.

Watch STAMM pitch on IndieBio Demo Day, Tuesday Nov. 6th in San Francisco or via LiveStream. Register here!

EMBER: Distributed Emergency Response for Cardiac Arrest

Cardiac arrest is one of the most time critical conditions that can strike anywhere. For the patient, each minute until the ambulance arrives has a huge impact on survival and future medical complications. Ember Medical is solving this by connecting the millions of high-risk patients with CPR-trained medical professionals in the community. Their app alerts nearby first-responders and 911, allowing anyone with cardiac arrest to get stabilized while the ambulance is still on the way.

We chatted with Mohamed (Shadi) Wahba, Co-Founder and CEO of EMBER about the company’s origins and progress so far.

How did you become interested in digital health and emergency medicine?

Shadi: I think it all started because I lost a lot of people in my life. I lost my friend when I was 17 years old to cardiac arrest, and then I lost my mother when I was about 21. At that time I was very interested in technology and finding a way to use it to help people in times of greatest need. This was something I sat with for a long time, and I tried a lot of different projects until I found the right idea and team. That’s how EMBER came to be.

How did the company start and how did you get your team together?

Shadi: It all began on a phone call between myself and my cousin, Mo, who is a cardiologist. He wanted to improve defibrillators, and I had the engineering background to help. We came up with a plan to build a, cheaper, faster, smartphone-enabled defibrillator, but soon realized that a new and improved AED didn’t begin to solve the real problem we were trying to address. The main issue causing survival rates to be very, very low in cardiac arrest, or similar time-sensitive emergencies, is that it takes too long for an ambulance to arrive on the scene.

There are nearly 60 million Americans who are CPR-trained or medical professionals. With those numbers the odds are high one will be near a victim and able to provide life-saving support before an ambulance arrives. We realized the solution to improving survival rates was simply to notify these individuals that their help is needed nearby. After further research, we found that several countries were in the process of adopting a similar idea at a governmental level, and studies have shown that this has increased the survival rate significantly. We had the research and the technology, and we knew that this was a platform that not only had real-world potential but real-world successes.

How does EMBER work if I’m just any person off the street who wants to use your product?

Shadi: Our app is available across all app stores to anyone with a smartphone. If you are walking on the street and you or a loved one has a sudden medical emergency, you can tap the in-app alert button to notify 911 and the nearest medics around you that you need help. Of course, there may be emergencies in which your movement may be limited. Our app allows your loved ones to download the app on their own smartphones—we’ve even enabled voice activation for easier access.

What happens when your app gets notified?

Shadi: Once the alert button is pressed, we simultaneously send your location to 911 and to nearby medics. When the medic is notified, they can choose to respond with a one-tap confirmation. They are then guided through in-app navigation to your location and will provide on-site support in the form of CPR and/or defibrillation until the ambulance arrives.

Our medic network will be responding to cardiac arrest cases in our pilot phase, and will then expand to respond to other time-sensitive emergencies such as opioid overdose, hypoglycemia, and anaphylactic shock. When each minute matters, EMBER ensures that a professional from our network is on their way to help as fast as possible.

What lessons did you learn transitioning from this idea into building a company while at IndieBio?

Shadi: It has absolutely been a transformative experience for me. This all started as an off-hand idea and as what felt like a project with my cousin. It’s totally different from building a company where we now have to think about all the different dynamics involved: who we’re going to hire, how to prioritize, what needs to be built first, where to find funding. It’s a much more complex animal. But the thing I loved about IndieBio is that we always had resources to learn from—whether that was the IndieBio team themselves or our fellow startups with vastly different life experiences. The amount of knowledge that I gained about the startup world and how to manage a company through these last four months is unparalleled. I wouldn’t have accomplished nearly as much without this network. It wasn’t an easy transition, but it was amazingly valuable. I can’t say enough about how much I learned and grew, as both a person and a CEO.

What were some of your greatest successes in the first months of building Ember?

Shadi: In just a few months we’ve formed critical partnerships  to build our network of medics, increase awareness, and expand to potential userbases. What started as an idea is now a full-fledged platform backed by partnerships with the American Nursing Association, Sutter Health, and other international organizations. It is very exciting for us to have partnered with the Qatar Foundation where we will be helping to provide emergency support for the World Cup in 2022.

How do you think EMBER’s success can change the emergency medicine landscape?

Shadi: A large part of what we are doing involves integrating with 911. This is a crucial part of improving the Emergency Medical Services (EMS) system as a whole. 911 technology is very much stuck in the 1970’s and 1980’s, which doesn’t make sense when existing technology is capable of so much more. Lives are at stake. Why not use the technology we have to help the most crucial system in our lives? EMBER eliminates the technology gap and brings EMS out of the dark ages. It’s a comprehensive solution, and I do believe it will be the go-to app for medical emergencies.

We can’t afford to waste time in the multistep process of calling 911 and waiting 15 minutes for an ambulance to arrive. EMBER makes it possible to use existing technology to notify and receive help within 5 minutes. Life is our most valuable commodity, we need to make the most out of the technology we have to preserve it.

What milestones is EMBER aiming for in the near future?

Shadi: We have three big milestones that we want to achieve in the near future. The first is to grow our medic network. The more medics we have, the faster response will be and the more lives we can save.

The next will be full integration with 911 dispatch centers. Our goal is to establish a two-way communication to improve their systems for all callers. For our third milestone, we’ll focus on expanding our emergency response efforts to address all time-sensitive medical emergencies, including opioid overdose, hypoglycemia, and anaphylactic shock.

We’re excited to hit these milestones, and many more, to make EMBER the go-to resource for all medical emergency needs in the near future.

Watch Ember pitch on IndieBio Demo Day, Tuesday Nov. 6th in San Francisco or via LiveStream. Register here!

Call for Applications

Biology is the next big technology and we are looking for scientists that will usher the new wave of iconic life science companies.

“Nothing is normal in the new biotech; it’s inherently cross-disciplinary and purposefully attacks preconceptions of what can’t be done.”- Po Bronson

Why Scientists?

Scientists make amazing entrepreneurs due to their technical expertise, problem solving skills, resourcefulness and persistence. Over the years, the number of life science PhDs has been rising exponentially and at the same time running biological experiments is becoming faster and cheaper. Our ability to read, write, cut, copy and paste DNA more efficiently is significantly decreasing costs and increasing speed and accessibility of experiments. Tying these trends together, IndieBio enables scientists to build radically transformative companies through our unique program.

Our Program

We take a design-driven approach to integrate product and business development in continuous feedback loops. Startups can rapidly prototype and get early customer traction at a pace that is closer to an IT startup rather than traditional biotech.

During our 4-month program in downtown San Francisco, scientists leverage $250,000 in funding with our fully-equipped labs, 300+ mentors and a galvanized ecosystem of industry, academia and investors that enables life science businesses to thrive. Our interdisciplinary team works with the companies every day to enable scientists to de-risk their science and business. Together we build the foundations of a viable and scalable business that can impact billions of people.

 

What types of companies do we look for?

We are here to fund scientists that can translate scientific insights into commercializable products that solve large scale human and planetary problems.

The biggest advantage a startup has is the precise focus on solving a problem from first principles. Our most successful founders build on deep technical knowledge with market insights that come from approaching the problem from all angles. As a result, startups create new business models, reimagine antiquated systems, or build industries from the ground up.

We welcome applications of biology for any industry. Thus far, our companies have represented the eight categories below, but we are most excited about companies that bridge multiple categories or invent new categories.

 

Therapeutics

Despite the billions spent in R&D, we continue to treat symptoms and not the causes of disease. New modalities such as immunomodulation and functional metabolomics are setting a new paradigm in drug discovery and delivery. With gene therapy we will have the ability to directly edit our own genomes to fix inherited diseases and transcend our parents’ genetic material. Across all these modalities there has been a rise in platform technologies enabling repeated target and therapeutic discovery.

Regenerative Medicine

Regenerative medicine and tissue engineering will give us control of how we treat damage to our bodies, from losing limbs to restoring loss of function from paralysis to growing whole replacement organs. Furthermore, we can intervene and reverse the processes of aging.

Neurotechnology

We are only beginning to understand the brain. New biological and digital tools are needed to understand and treat neurodegeneration and mental health. Brain computer interfaces open up new possibilities for human consciousness.

Medical Devices, Tools & Diagnostics

Early detection of diseases that can seamlessly integrate in to healthcare workflows not only enhances decision making through precise real-time biomarkers, but also, eliminates centralized labs and administrative bottlenecks that are burdening the healthcare system. The advancement of research tools is critical for unlocking new knowledge that can lead to life-saving solutions.

Future of Food & Agriculture

Food supply cannot catch up with food demand at the same time supply remains inefficient and unethical. New biotechnologies are changing the unit economics of how we produce protein. Vertical integration of food and agtech can enable us to unbundle the food supply system and increase efficiencies of production.

Consumer Biology

Driven by faster and cheaper science, companies will bring biology direct to consumers in an increasingly personalized manner. The first human genome cost $2.7B. Today people can order an at-home sequencing kit for $100. Products are increasingly putting the power in the hands of the consumer to manage their own health.

Computational Biology, BioData, & AI

Biology is rich with data and complexity and companies are increasingly leveraging bio-processes with machine learning and automation, creating bio-feedback-loops to optimize each stage of a life science company: from discovery to manufacturing.

Industrial Biology, Biomaterials, & Clean Biotech

Not only is our demand for commodities unsustainable, the industrial processes for converting commodities into everyday products remains inefficient. Biology is inherently versatile and scalable. Cells, the building blocks of life, live to divide, and under the right conditions they can be engineered to create bio-materials and novel commodities that can then be scaled exponentially using fermentation without harming the planet.

 

Our Application Process

Online Application. Our application process begins with an online submission at http://indiebio.co/apply.

Technical due diligence. Selected companies are invited to a 30-minute video interview that focuses on technical due diligence. We encourage applicants to ask questions about the IndieBio program throughout the process.

De-risking milestones. If the first call is favorable, our team will set up additional calls to discuss the product, the business plan, and discuss the derisking milestones that the company aims to achieve by the end of the 4-month program. Often, homework is assigned to address certain questions. Once discussions are mutually favorable, an offer is made.

Deeper look into how do we evaluate companies. We evaluate companies based on five key questions.

  1. What is the technical insight that gives you an unfair advantage? This is often the core technology that can be patent-protected, whether it is licensed from an academic institution or developed in-house. What advantages does your technology have over competing technologies? How does your technology address the core problems you are trying to solve?
  2. How is the insight made into a product? Science itself is not a product. Product development starts with understanding the end user. What problem are you solving for the end user? What is the form factor? What is the workflow? What are the parameters for a successful drug? What product do you focus on first when you have a platform technology?
  3. How does the product form a sustainable business? What is the go-to-market strategy for roll-out when the startup is cash limited? How to gain adoption? How to navigate regulatory pathways?
  4. Can this business make $1 billion or touch the lives of 1 billion people? Venture capital investment seeks the potential for big returns and big impact.
  5. Is this the team to make it all happen? Arguably, the most important aspect of selecting teams at an early stage is the founders themselves. Do they have the experience and expertise to turn their technical insight into a viable business and propel the company into a flourishing venture? We look for founders who are coachable, able to make decisions rapidly, take responsibility, are resilient, and are passionate about their work. We look for founders who are self-aware and possess a growth mindset.

The interview style is informal conversations and we often instill a mini preview of the IndieBio program during the interviews. Our application timeline is rolling, with set deadlines that batch the interviews. We encourage applying early and sending periodic updates of progress even if you don’t hear back immediately. Updates are also encourage between interviews as it could take time for both sides to come up with good strategies. We also encourage re-applications if you were not selected for one class. Some of our most successful companies reapplied 6 months later with significant momentum. Most ideas and teams will take a while to mature. (Read “I have an idea. What’s next?” for the starter checklist.)

Lastly, we encourage you to due diligence on us. Learn more about our story and our program featured in Neo.Life. Attend or livestream our next Demo Day on Nov. 6th at the Herbst Theatre or watch the previous ones on Youtube. Talk to founders of any of our alumni companies or attend an event at our space.

 

Apply Now!

We look forward to hearing your world-changing idea! Apply now at http://indiebio.co/apply!

JointechLabs: Point-of-Care Cell Therapy

JointechLabs

Stem cells have been a key source of innovation in regenerative medicine, because they possess the ability to self-renew. Adult stem cells are found within the human body, in bone marrow, tooth enamel, and other locations. When someone is injured, the inflammation from the injury acts as a signal for the stem cells to become activated. The stem cells heal the body by traveling to the affected location and changing into the cells that have been damaged.

This process doesn’t always work fast enough to heal all injuries, so stem cells can be manually harvested from areas where they are abundant, and transferred to areas in the body where they are needed. JointechLabs have created a device which makes this process more accessible and affordable for doctors and patients everywhere. We asked Nishit Pancholi, the President and Chief Medical Officer at JointechLabs, more about the company:

How did you become interested in biotech?

NP: We were always interested in the potential of harnessing the natural healing powers of the human body, and use that to enhance healing, and regeneration. It is painful to see near and dear ones continue to suffer from orthopedic problems or non-healing wounds after receiving conventional treatments. Improving science so that one’s own stem cells can be more accessible to help these patients heal became the founding mission of JointechLabs.

When did you decide to start a company, and where did your team get together?

NP: In 2010, Dr. Nathan Katz, who has extensive experience in stem cell biology, started JointechLabs with Felix Pustilnik, who has experience in business and sales. I have a background in medicine with a focus on clinical applications of stem cells, and I joined the executive team as a co-founder later.

How does your technology work?

NP: JointechLabs has created a device to provide stem cell therapies on demand, in a doctor’s office, for 1/10 of the price of conventional alternatives. Although stem cell therapies have been shown to have tremendous potential and efficacy in treating a variety of illnesses, they are currently underused, as the facilities and doctors that are capable of delivering them are few and far between. Stem cells are not easily transported, and so patients often must travel great distances in order to be treated; so inevitably the treatment is also incredibly expensive.

Our technology enables doctors to practice stem cell therapies and allows for development of stem cell therapies for medical indications with unmet medical need. Our device provides access to a high quality cell fraction at the point-of-care, therefore, a doctor can offer cell treatments in the regenerative medicine segment. Going further, our cell therapies will provide treatments for medical indications with unmet medical needs: orthopedic indications, wound healing, vascular indications.

What lessons did you learn transitioning from science to entrepreneurship at IndieBio?

NP: Stay focused on what’s important for the company development and progress. Identify and clarify your product, development roadmap, regulatory aspects, market strategy and risks mitigation of all listed. Develop an understanding of the costs associated with each step. Prepare for prospective company growth: HR, legal, etc.

How do you think your success as a company would change the healthcare industry?

NP: Patients will receive access to effective treatments for indications with unmet medical needs. Since the condition will be treated in the early stage of development, the cost of indication life-cycle will be reduced dramatically, providing direct benefits to the insurance companies and, therefore, to the patients.

What milestones are you aiming to hit in the near future?

NP: FDA 510K approval for our medical device and brining the device to doctor’s offices. RMAT (Regenerative Medicine Advanced Therapies) FDA designation for our stem cell therapies.

Watch JointechLabs pitch on IndieBio Demo Day, Tuesday April 17th in San Francisco or via LiveStream. Register here!

An Interview With Brendan Griffen of Scaled Biolabs

Scaled Biolabs

A Biomedical Lab the Size of Your Phone.

Scaled Biolabs

Photo: The Scaled Biolabs team – Brendan Griffen (left), Justin Cooper-White, and Drew Titmarsh.

Biological experiments are time-consuming, and space-consuming, while requiring repeated manual motions from lab technicians. But technology has allowed Scaled Biolabs to shrink down the entire system of experimentation down to the size of your phone. Using microfluidics, scientists can conduct thousands of individual tests in one fell swoop and accelerate the rate of discoveries. We talked to the company’s CTO, Brendan Griffen, a few questions about how this all came to be:

Tell me about your background, how did you become interested in biotech?

My academic background is in computational physics with an specialization in astrophysics, theoretical physics, and cosmology. I’ve spent the last ten years working on large projects trying to understand the origin of our universe, the evolution of stars, and galaxies.  From these experiences, I’ve seen first hand the power of a supercomputer in solving some of the most complex problems in the world. This is fundamentally because we’ve been able to control the flow of electrons through circuits at increasingly smaller scales. Biology is similar in many ways but instead of mixing and moving electrons from point A to point B, we’re moving fluids. This is the fundamental way biomedical research is currently done so it always perplexed me as to why we continue to use clunky equipment to interface the human scale with what we’re actually interested in (e.g. cells interacting). This is what excited me about the future of biotechnology, our ability to do biology at nature’s scale. The potential to miniaturize most of biological research means that in the next 50 years, we will likely transform our lives even more dramatically than what computers have done in the past 50 years.

I’ve always been interested in biotechnology but never had the opportunity to apply myself to developing these ideas. When I saw the technology my co-founders Drew Titmarsh and Justin Cooper-White had developed, I immediately understood that a microfluidic approach to experimental biology is exactly what’s needed to make the aforementioned future a reality. In order to be part of this exciting future, we formed our company Scaled Biolabs.

What problem are you working to solve with Scaled Biolabs?

We are accelerating discoveries in biology. We’ve taken the functionality of a modern biomedical R&D lab and shrunk it all down on a system the size of your phone. By shrinking things down we rely on less expensive materials, less highly trained manual labor, and most importantly we can execute numbers of experiments on an unprecedented scale. We can run nearly 10,000 experiments on a single system and additionally resolve every single cell in every one of those experiments. Why is this important? If a biologist can run down every single possible path in a maze of possibilities faster, then they can find the optimal solution which gets them to their desired outcome sooner rather than later.

Stem cells are one such maze and a very active area of research right now — how do we turn stem cells, the ‘blueprint cell’, into different parts of the human body? Our collaborators have already grown human kidney and beating heart tissue in our system because they found the optimal method for getting to those outcomes faster than traditional methods. At Scaled Biolabs, we enable these kinds of breakthroughs by allowing scientists to get more done, cheaper and sooner.

If you could only pick one thing to validate your reason for forming a startup, what would it be? In other words, what would be the single biggest indicator to you that you are doing the right thing?

All discussions of “what has value?” tend to link back to a basic notion of health and wellbeing. I personally believe that our team really does have the potential to deliver fundamental improvements to human health. Take just one example — in 2005, 65 million people died from kidney disease. Being able to regenerate a patient’s kidney from their own stem cells, which won’t be rejected by their immune system, will literally save lives. If we had to boil it down to a single reason for doing all of this then it’s that — accelerating the advent of regenerative medicine to meet the needs of millions of patients who can’t be helped by a pharmaceutical drug.

How do you think success can change your industry?

Apart from meeting patient’s needs, success to us is creating a new status quo in our industry. Biological research is in desperate need of an upgrade, and should our approach be successful, we would be enamored to see it more widely used. We don’t want to stop just there though. Unknown problems on the horizon will require solutions not yet invented, and so being successful will mean not just creating a new status quo, but also continually creating novel technological solutions.

How is your team uniquely able to tackle this? What’s the expertise?

We’ve got complementary talent trained around the world.

Our CEO, Drew Titmarsh, is a trained chemical and biological engineer and co-inventor of the microbioreactor technology which is the workhorse platform of Scaled Biolabs. He has coordinated multidisciplinary projects at the Australian Institute for Bioengineering and Nanotechnogy (Brisbane, Australia), and the Institute for Medical Biology, A*STAR (Singapore) in the areas of tissue engineering and regenerative medicine.

Our CSO Professor Justin Cooper-White is a global leader in tissue engineering and microfluidics, and co-inventor of our microbioreactor technology. With 20 years of expertise in running and funding large research programs, he currently holds the positions of Professor of Bioengineering in the Australian Institute of Bioengineering & Nanotechnology and the School of Chemical Engineering at the University of Queensland, Director of the Australian National Fabrication Facility-Queensland Node, and Office of the Chief Executive Science Leader within CSIRO, Australia’s federal research institution.

I’m the CTO and have ten years in computational physics employing a wide variety of hardware and software tools to create solutions to big data problems. My previous four years research as a postdoctoral fellow at MIT has provided me with a wide range of interdisciplinary skills which are well suited to our challenges ahead.

Any big lessons learned transitioning to startup entrepreneurship?

It’s been quite a dramatic transition. The following three areas are where we’ve found the biggest lessons:

  1. Being OK with moving more quickly than you’re comfortable with. Academia tends to have a perfection focused mindset because the operational timescales are much longer. In industry, things are measured in days. The closest analogy I’ve found is it’s like morphing from a mammal into an insect — your priorities certainly change!
  2. As the phrase goes, “it’s not what you know, it’s who who you know”. Academia tends to (in the ideal case) be more of a meritocracy where what you know really does give you the greatest return. In industry, having strong relationships with people you can lean on for help or to stage a warm introduction often converts into something of great value.
  3. Follow up. No one cares about your business more than you, so you really have to make that extra effort to follow up with people if an email or call thread goes cold. Even if there is not a quid pro quo to be had on the business side, it is always very useful to keep all learning opportunities available. There are large number of tools online now which allow you to maintain several hundred conversations at once and ensure that you don’t let potentially important relationships go cold.

What’s the biggest challenge you’ve encountered so far?

Our biggest challenge has been our messaging and identity (i.e. “what are we?”). This is often the case for platform technologies — you can address multiple problems but the key is to find the underlying compelling narrative which brings it all together. Thinking more fundamentally about our technology and which direction we are heading has really helped us solve this problem. Traction also does wonders to identity woes because you get validation that what you’re building is something people want, so it’s much easier to speak to that than just a lofty garage band idea.

What are the big goals and milestones you’re looking to hit in the short term? Long term?

At the moment we are focused on providing value to customers who seek the advantages of our technology. We are in the early stages of our long term goal of placing our system in every research lab around the world. These are primarily companies either creating high quality stem cells or turning stem cells into different tissues of the human body. In the very long term we want to place our instrument in all doctor’s offices so that unfortunate folks who are diagnosed with cancer can get personalized treatment plans tailored to their own immune system and cancer type. With this two pronged approach we aim to become industry leaders in both regenerative medicine and personalized medicine. We’ve got a long journey but we’re making good strides.

Learn more about Scaled Biolabs by watching them pitch on IndieBio Demo Day Feb. 9th! Register for the event or LiveStream here!

An Interview With Arshia Firouzi of Ravata Solutions

Ravata Solutions

Electronic Embryo Alteration.

Ravata Solutions

Photo: Arshia Firouzi (left) and Gurkern Sufi.

There are numerous reasons why new therapeutics take so long to become accessible to people suffering from life-threatening diseases. Many lab animals which are used in the testing of new treatments, most notably mice, often need to be genetically modified in order for them to best represent the condition that needs to be treated by new substances. The process of altering mouse embryos is extremely manual and time consuming, which is why Ravata Solutions has developed a new device to cut the process down to a fraction of its original time. We asked the company’s’ CEO and founder, Arshia Firouzi, a few questions:

Tell me about your background, how did you become interested in biotech?

I grew up in Southern Illinois and moved to Sacramento, California in late 2001. In 2011 I began my education at UC Davis where I studied Physics and Electrical Engineering. Following my graduation in 2016 I teamed up with my long-time friend and housemate Gurkern Sufi to start Ravata. We were and continue to be very excited about the intersection of electronics and biology. It is our view that the union of the two can accelerate achievement and advances in both fields.

My personal interest in public health stems from my experiences with my epilepsy. I am fortunate to have my seizures controlled but I continue to visit a neurologist and interact with many people having various neurological diseases. One of my favorite moments at Ravata was seeing the neuroscience research being done in mouse models. Knowing that our success means the success of neuroscience has fueled my passion for our work.

To an extent I almost feel like biotech became interested in me. I ended up with friends doing research in bio, professors working with biotech entrepreneurs, and then eventually a research project in biotech that led me to Ravata.

What problem are you working to solve with Ravata Solutions?

We are working to solve the limitations surrounding embryo engineering. Our technology is opening a bottleneck in the way genetically modified animals are created. Today, the process of transforming animal embryos is a manual one. As a result, animals used in medical research and preclinical trials can take over a year to produce. Furthermore, many times these animals are not good models of the disease they are meant to represent. What we are doing at Ravata is providing a cost effective and time efficient method to create quality animal models.  

If you could only pick one thing to validate your reason for forming a startup, what would it be? In other words, what would be the single biggest indicator to you that you are doing the right thing?

The fact that we have a technology to make a difference and a focused team to bring it into reality is how I validated forming a startup.

How do you think success can change your industry?

Our success means that the rodent model industry can produce better animal models for medical research and preclinical trials faster (up to 100X) and more efficiently than ever before. This will significantly shorten the research and drug development timespan.

How is your team uniquely able to tackle this? What’s the expertise?

Our technology involves the intersection of electronics and biology. My team has the necessary experience with electrical engineering, biology, and material science to tackle the challenges associated with the science.

Any big lessons learned transitioning to startup entrepreneurship?

The biggest lesson I have learned transitioning to startup entrepreneurship is that having a great idea is only 1% of having a successful business. There are many great people with many brilliant ideas. It takes a combination of a large network, hard work, and strong mentors to be successful.

What’s the biggest challenge you’ve encountered so far?

The biggest challenge I have encountered so far is learning how to manage my bandwidth. There are always urgent tasks needing to be handled, many of which I have no experience with. In order to get through everything requires a new understanding of what is necessary, how to delegate tasks, and time management.

What are the big goals and milestones you’re looking to hit in the short term? Long term?

In the short term, we are looking to finalize the designs for our current system and enter the rodent model market. In the long term, we are aiming to adapt our device to work with other animal models and eventually other cell types such as plants, fungi, and even human cells.

Learn more about Ravata Solutions by watching Arshia pitch on IndieBio Demo Day Feb. 9th! Register for the event or LiveStream here!

Magnetic Pulses to Combat Depression: An Interview With Mehran Talebinejad of NeuroQore

NeuroQore
NeuroQore

Photo: The NeuroQore team (Mehran on far left). 

Depression is a major burden in many people’s lives who we know. Some of the treatments that are prescribed, like medication, are not always effective or without major side effects. Drug-resistant depression is sometimes treated with electroshock therapy, which is risky, despite being the gold standard. In comes NeuroQore, a new device that aims to treat depression by sending magnetic pulses to a small region of the brain, which is safer than electroshock therapy. We asked NeuroQore’s CEO, Mehran Talebinejad, a few questions:

Tell me about your background, how did you get interested in the biotech space?

As a teenager I was fascinated with brain machine interfaces (BMI) and mind uploading. This drove me to study Biomedical Engineering and go towards neural prosthetics and brain surgery. Fast forward 10 years after getting into the university, and I did my first brain surgery. During this surgery I realized the brain is so extremely complex, and machine BMIs have a long long way to go before being publicly available. I also realized non-invasive brain tools and neuromodulation is super important since we don’t have easy access to the brain while the skull is blocking us!

So how did you try to turn that complexity into something practical you could work on?

Among non-invasive approaches to brain stimulation or neuromodulation there are only two very promising approaches: the first is electroconvulsive therapy (ECT) or electroshock therapy. This is the gold standard for treatment of drug-resistant depression. The second is the magnetic brain stimulation or repetitive transcranial magnetic stimulation (rTMS), which emulates ECT in smaller brain regions without convulsion. ECT requires hospitalization, anesthesia, and has severe cognitive side effects (memory loss) and a risk of death. Less than 1% of patients are willing to endure ECT! On the other hand, rTMS is outpatient, has no systemic side effects and is widely accepted by patients. I saw the potential of rTMS and I had a vision to make it more accessible and more effective for treatment of drug-resistant depression and a range of other brain disorders (psychiatric and neurological).

What problem are you working to solve with your company, NeuroQore?

TMS is a platform tech with a range of applications, but we are focused on drug-resistant depression at this time. Over 16M patients are diagnosed with major depressive disorder (MDD) every year in the US, and more than 4M remain drug-resistant. Which means they do not get satisfactory results from drugs in the first line of therapy. There is an option for them to do ECT, but as I mentioned less than 1% willing to endure ECT (still over 100,000 patients/year). So there is a large unmet need, or I would say crisis, for drug-resistant depression. Depression is among few disorders in medicine where a patient says “I rather die than have this”! NeuroQore is set to make TMS accessible and more effective as an alternative option to ECT.

If you could only pick one thing to validate your reason for forming a startup, what would it be? In other words, what would be the single biggest indicator to you that you are doing the right thing?

The indicator is being able to execute on my own vision and feeling satisfied after seeing patients getting into remission. Patients bring us all types gifts and flowers in the last sessions of their treatment, and almost all of them have been super satisfied, which is very fulfilling for me.

How do you think success can change your industry?

NeuroQore is set to change mental health care and psychiatry as we know it, a condition that has been relatively unchanged since the 1960s. Patients with depression will be able to go to our centers (i.e. the “Apple Store for Depression”), monitor their depression with physical evidence (biomarkers not just anecdotal questioners), and finally get effective outpatient treatment! Today mental health care and psychiatry both suck, it is literally depressing to get depression treatment.

How is your team uniquely able to tackle this? What’s the expertise?

We have a great multi-disciplinary team and have been working together for over a decade. My academic background is in biomedical engineering, neuroscience, and technology management. I was selected as a rising star CEO by Invest Ottawa and I have been recognized and awarded many times for my work at NeuroQore as a co-founder and CEO.

Adrian is my co-inventor and co-founder, and has been working with me for over 14 years. He is an award winning expert in scientific research and development, with academic background in electrical and biomedical engineering specialized in non-invasive approaches.

Jonathan is a pioneering rTMS clinician/psychiatrist. He is very well known and well respected in the psychiatry society, and has had amazing contributions to improve clinical rTMS for depression treatment. He has experienced over 2,000 patients in his practice to date.

Brittany is an angel, she is our anticipatory patient service expert, with an academic background in psychology and mental health neuroscience. Her role is crucial in patient experience, which is very important for mental health. She is helping us change mental health care as we know it with her innovative service approach.

Any big lessons learned transitioning to startup entrepreneurship?

Life can be very exciting and fulfilling, but as an entrepreneur I must be ready for anything above and beyond what I know and have learned. I must be ready to learn on the fly and adapt to new situations.

What’s the biggest challenge you’ve encountered so far?

Educating the government, the public, and clinicians about rTMS and non-drug depression treatment… and removing the stigma of depression.

What are the big goals and milestones you’re looking to hit in the short term? Long term?

In 2017 we will have four centers operational/active in California (SF, LA, Oakland, and Long Beach), and in the long term we are planning to repeat this model across the US with over 100 centers in the next three years.

Learn more about NeuroQore by watching Mehran pitch on IndieBio Demo Day Feb. 9th! Register for the event or LiveStream here!

The Bad Boy of Biosensors: An Interview With Ray Chiu of BioInspira

Bioinspira
Bioinspira

Photo: Ray Chiu (far right) and the BioInspira team. 

Every year, more than 200 natural gas pipeline-related incidents happen on average in the United States, and $5B in economic opportunities are lost as a result of gas leaks. BioInspira is aiming to solve this problem by bringing air chemical detection to the next level.

At IndieBio, we call the founder and CEO of BioInspira, Ray Chiu, the “Bad Boy of Biosensors”. Not only has he helped the company raise over $1.3M, he’s also closed partnerships with a consortium of the largest northern and southern California gas and electricity companies. BioInspira uses biology to change the economics of how we monitor our infrastructures. We asked Ray a few questions:

Tell me about your background, how did you get interested in the biotech space?

My background is in chemical engineering. I’ve had a huge passion for science since I was little, and I was always very keen on learning about breakthroughs in new frontiers of scientific research. Because of this, I made the decision to participate in this task of expanding scientific knowledge. Biotechnology is a relatively unexplored area filled with unknown potentials. Whereas the microelectronic revolution has come and gone with the projected bottleneck from Moore’s Law, there are still many secrets we can still learn about biology, biochemistry, and how they can change our way of life. This wealth of potential for discovery and impact on our life is what propelled me into the biotech space.

What problem are you working to solve with your company, Bioinspira?

Billions of dollars worth of economic opportunities are lost because there are currently very ineffective ways of tracking our infrastructures. This is a huge problem. We envision that, by combining network connectivity with powerful sensors, we can eliminate waste that is generated by the use of Earth’s natural resources. Take natural gas leaks, for example. BioInspira can save as much as 40% of the gas that is leaked from our gas infrastructure while cutting the leak inspection time in half.

Such engineering marvel, if successful, will also provide unprecedented data and insight and lead to a safer smarter world. However, current sensing technologies can’t achieve this goal due to their power consumption, size, cost, and accuracy. BioInspira believes a new revolutionary sensing mechanism is required. And we aim to solve this problem with our technology.

If you could only pick one thing to validate your reason for forming a startup, what would it be? In other words, what would be the single biggest indicator to you that you are doing the right thing?

Passion. I firmly believe that the technology our team is working on will revolutionize people’s way of life and lead to a safer and smarter world. Seeing my technology become successful and actually influence society would be the single biggest indicator that I am doing the right thing.

“I firmly believe that the technology our team is working on will revolutionize people’s way of life and lead to a safer and smarter world.”

How do you think success can change your industry?

If we are successful, we will revolutionize the way in which the industry performs inspections for safety and emission control. Our customers will have constant and real time information on leaks in their systems, leading to efficient leak repair processes. This would help reduce waste and save lives.

How is your team uniquely able to tackle this? What’s the expertise?

We have more than 20 years of professional sensor research experience. We are experts in phage sensor engineering. Our sensor research goes back more than 10 years with more than $2M research funding invested to date. We also have industry experts, thought leaders, and the chief inventor of our technology forming a powerful advisory board. In addition, we have partners with OEM partners on device manufacturing that will ensure the quality of our solution.

Any big lessons learned transitioning to startup entrepreneurship?

Do not take anything for granted. Besides changing our R&D plan to meet the customer’s schedule and needs, we invested most of our time to grow relationships with potential customers and end users. If we do not turn these into a potential sales channel or investment opportunity, or we ruin our relationships, all our time will be wasted.

What’s the biggest challenge you’ve encountered so far?

As this is a completely new sensing mechanism, one of our biggest challenges was to explain the technology to potential end users as well as investors. Most of the end users are experts in sensors, but do not know much about biotechnology. On the other hand, most of the interested investors are very familiar in biotechnology, but do not understand much about the sensor industry.

What are the big goals and milestones you’re looking to hit in the short term? Long term?

Short term: successfully deliver our sensor development kit and complete field tests with customers. Long term: overhaul the sensor industry by providing a revolutionizing sensor platform with improved combined capabilities.

Learn more about BioInspira by watching Ray pitch on IndieBio Demo Day Feb. 9th! Register for the event or LiveStream here!

Creating a Functional Window to the Brain: an Interview with Henrik D. Kjeldsen of Truust Neuroimaging

Creating a Functional Window to the Brain: an Interview with Henrik D. Kjeldsen of Truust Neuroimaging
Creating a Functional Window to the Brain: an Interview with Henrik D. Kjeldsen of Truust Neuroimaging

Modern neuroscience is still relying on old methods that don’t allow us to truly understand what’s happening in the brain in real-time. As a result, we have a limited understanding of brain-related disease and ability to treat conditions early.

Truust’s neuroimaging technology is providing real-time data in order to visualize energy flow in the brain, and, as a result, be able to predict and treat brain-related diseases before they start. I spoke with the Henrik, the CEO, to learn how he discovered this problem, limitations of today’s technology, and how Truust can change the field. Check out his pitch from IndieBio’s Demo Day Livestream!

A: Tell me about your background, how did you get interested in the biotech space?

H: I met my co-founder, Lars, about 15 years ago, while working as an electrical engineer. As a result of the computational problems presented while working on the Semantic Web I got interested in artificial intelligence and did a Master’s of AI in the Netherlands. I realized current AI approaches are not able to handle such complex problems, and we need to study real intelligence to figure it out. So I went to the UK to do a PhD in Experimental Neuroscience, where I faced a new problem; current tools are only able to see very little of what is going on in the brain. This not only makes it very difficult to understand the brain, but also makes it impossible to diagnose brain-related problems early enough to catch disease before they present with behavioral symptoms or become actual structural changes in the brain. This made us realize that we need to improve our neuroimaging tools, and based on the idea of super-resolution we initially created a solution for specialized micro-electrode arrays that worked surprisingly well. Since then I spent some time at CERN, the global leader in experimental particle physics, where I really came to understand the potential of super-resolution signal processing techniques.

A: What problem are you working to solve with your company, Truust?

H: A major problem in neuroscience is that we are relying on statistical analysis to understand connections in the brain since we can’t actually see real-time energy flow. With our super-resolution method, we can see the actual flow of electromagnetic energy from point A to B which means we now have a window into the living dynamic brain for the first time. So using existing EEG hardware we will collect huge amounts of data that machine learning can be applied to and understand EEG biomarkers of a wide range of brain-related problems. We envision a future with an EEG terminal in every doctor’s office as part of every physical; and the doctor does not even have to be an expert, our biomarker system is all the assistance they need.

A: If you could only pick one thing to validate your reason for forming a startup, what would it be? In other words, what would be the single biggest indicator to you that you are doing the right thing?

H: Seeing all different kinds of people, from random kids to experts in the field, get really excited about our prototypes visualizing energy flow in the brain, tells us that we are really doing something new at the cutting edge of understanding the brain

A: How do you think success can change your industry?

We are going to revolutionize neuro-imaging to make it useful and relevant to the general population and their healthcare. It will become much lower cost than current MRI and PET techniques

A: How is your team uniquely able to tackle this? What’s the expertise?

H: We have a unique perspective on the brain being cross-disciplinary between electrical engineering, AI, physics, and neuroscience. This allows us to take a physical and computational approach to what has traditionally been a biological problem. We have worked on these problems in one form or another for a long time, and what we are doing now is a natural culmination of that path.

A: Any big lessons learned transitioning  to startup entrepreneurship?

H: We’ve had the expectation fulfilled that things move very fast. We can do things much quicker than in academia or in large companies. That’s actually one of the reasons we left those to form a  startup. Things were moving very slowly. In a startup we are learning much faster since we’re able to experiment and iterate so much faster.

A: What’s the biggest challenge you’ve encountered so far?

H: Talking about what we do in a way that isn’t too academic. There’s a balance between being technically correct and not being overly technical. These are new ideas that people aren’t generally familiar with, so communicating the nuances while still letting them see the big picture can be tough.

A: What are the big goals and milestones you’re looking to hit in the short term? Long term?

H: In the short term we want to validate our technique with a number of different labs on hard, real world problems, like better seizure localization for epilepsy. Our long term goal is to truly understand the brain, which would open up many more general applications all across medicine and research.

Get in touch with Henrik at henrik.kjeldsen@truustneuroimaging.com

Reprogramming Cancer and the Future of Medicine: an Interview with Andrew Gray of Vali Nanomedical

Reprogramming Cancer and the Future of Medicine: an Interview with Andrew Gray of Vali Nanomedical
Reprogramming Cancer and the Future of Medicine: an Interview with Andrew Gray of Vali Nanomedical

Drug delivery is one of the greatest challenges in treating cancer today. There are a multitude of effective drugs that aren’t able to be delivered to tumor sites, or can not be delivered in combination.

Vali Nanomedical is now solving these drug delivery problems with a revolutionary programmable drug delivery system, and working towards a future where in vivo cellular reprogramming can cure disease without even using drugs. I talked with Andrew, Vali’s CEO, about his path to entrepreneurship, its challenges, and the future of Vali. Check out his pitch from IndieBio’s Demo Day Livestream!

AK: Tell me about your background, how did you get interested in the biotech space?

AG: I have PhD in molecular biology, focusing on cancer and developing cancer vaccines as a grad student. I got involved in nanotech knowing that starting a company was the goal. Early on I took a class on entrepreneurship and knew right away that was the path for me, not traditional academia. Going into startups has been really exciting since it’s been a goal for some time.

AK: What problem are you working to solve with your company, Vali?

AG: The biggest issue in cancer is delivering drugs to where they need to be. There are a lot of great drugs out there that can’t reach the right place or are toxic. At Vali Nanomedical, we created a programmable nanoparticle that homes onto cancer cells and releases drugs there but doesn’t harm healthy cells. It’s like making a smart missile that knows when it has missed and refuses to detonate when it’s in the wrong place.

AK: If you could only pick one thing to validate your reason for forming a startup, what would it be? In other words, what would be the single biggest indicator to you that you are doing the right thing?

AG: This is the first time in my entire career that I’ve felt like I’m at home. Every day flies by and is exciting.

AK: How do you think success can change your industry?

AG: We envision a world where drugs aren’t used at all to treat disease. Instead, we’d use our technology to reprogram cells to either return to a healthy state or, if they’re too far gone like cancer cells, reprogrammed to kill themselves. Ultimately we want to make cancer a disease of the past.

AK: How is your team uniquely able to tackle this? What’s the expertise?

AG: We have a fantastic team with very complementary skills. Prof. Mike Wong is a physician-scientist who has dedicated his career to finding better ways to treat cancer. As I mentioned, I’m an expert in the molecular biology of cancer. Prof. Pin Wang is an absurdly accomplished and productive biomedical engineer at USC. The founding team is backed up by industry veterans, including former director of BD at Amgen Holly Hartman and Prof. John Daniels, who literally launched an industry by inventing and commercialized collagen for injection. Between us, we have all the skills necessary to make our grand dream a reality.

AK: Any big lessons learned transitioning from academia to startup entrepreneurship?

AG: You have to reframe the way you talk about your work. Academics are trained to talk about the things they’re 99% sure they’ve figured out. As an entrepreneur, you talk about problems that you haven’t solved yet and demonstrate you have a plan to get there.

AK: What’s the biggest challenge you’ve encountered so far?

AG: Finding the right first addition to the team. It was only by good luck and timing that I found the perfect person to add to the team. He was an amazing materials scientists named Don Johnson, who’s also a PhD.

AK: What are the big goals and milestones you’re looking to hit in the short term? Long term?

AG: Short term are building multiple partnerships with pharma to deliver drugs they can’t deliver now, either alone or in combination to treat cancer. We’re uniquely capable of doing combination therapies. Like I said, in the long term we want to make cancer a disease of the past.
Get in touch with Andrew at andrew.gray@valinano.com

Bringing Space Technology to Healthcare: an Interview with Jeff Nosanov of V-Sense Medical

Bringing Space Technology to Healthcare: an Interview with Jeff Nosanov of V-Sense Medical
Bringing Space Technology to Healthcare: an Interview with Jeff Nosanov of V-Sense Medical

Tracking vital signs is a crucial means for preventing disease. However, it’s a big time demand on caretakers of the most vulnerable patients and rarely done for much of the general population.

V-Sense is developing monitors using NASA Jet Propulsion Lab radar technology to remotely and continuously monitor key vital signs. I talked to the company’s CEO, Jeff Nosanov, about learning how to apply this new technology, lessons moving from research to startups, and goals for V-Sense. Check out his pitch live on February 4th on IndieBio’s Demo Day Livestream!

A: Tell me about your background, how did you get interested in the biotech space?

J: I always wanted to be an astronaut and started college in engineering because of this. I later switched out to approach the space world from a different angle and got the first ever Space and Telecommunication Law degree. That got me my job at NASA Jet Propulsion Lab (JPL) testing clinical application for a new radar technology. My biotech interest started in 2012 after my newborn was in the NICU for a week and we kept having problems with his vital sign monitors. I found out this is a very common issue in medicine and almost considered commonplace, which I thought was really strange and unfortunate. I knew the technology I was involved with could remotely measure vitals and realized it would be very helpful for this application. Then when my second child was born and had the exact same problem in the NICU, I got a really passionate about applying this technology to medicine. When we moved to Bethesda for my wife’s residency I left NASA and had the freedom to pursue this startup and technology.

A: What problem are you working to solve with your company, V-Sense?

J: Understaffing is a huge problem in nursing homes across the US. Due to this nurses and staff can’t measure vital signs on schedule and as often as needed. This results in missing a lot of important information on patients that could improve quality of care and save money. We’re automating this time-consuming task which is a win for everyone. As we expand into consumer homes we can provide the same service to millions more people.

A: If you could only pick one thing to validate your reason for forming a startup, what would it be? In other words, what would be the single biggest indicator to you that you are doing the right thing?

J: I’m really excited about the day I get a call from our first customer that says we saved someone’s life because of our device.

A: How do you think success can change your industry?

J: For five years at the JPL I would hear from my wife, who was in medical school,  how some techniques doctors use are brand new and others are two thousand years old. We often have a big gap between techniques doctors are using and what modern technology can do. I’m interested in bringing more advanced technology to medicine via the rapid innovation possible in a startup. There’s a lot of great technology sitting in research labs throughout the country but no one is having the light bulb moment to apply it. I want to see more innovative medical technology coming out of space technology research.

A: How is your team uniquely able to tackle this? What’s the expertise?

J: I spent two years working with the team that invented this technology, mostly looking at how to apply it clinically. So I’ve been working with this technology and its users for years. My CTO, Hector, spent over a decade at JPL working on tons of radar technologies and applications. After that, he did a lot of product consulting and is perfectly positioned to bring this technology out to the world. Plus, we still have access to JPL and the tremendous minds working there.

A: Any big lessons learned transitioning from research to startup entrepreneurship?

J: The power of going out and directly talking to your customers. It’s not really clear who our customer is with research since we don’t know if our technology will reach the outside world. In business we can go out and ask the user to design it with us and how to make it as useful as possible for them.

A: What’s the biggest challenge you’ve encountered so far?

J: The medical community is fairly conservative, for understandable reasons. It can be a challenge to communicate possibilities of radar technology since they aren’t trained to know its nuances and applications. We have to communicate in a collaborative manner to explain the potential and value of this new way of doing what they already do. Having a wife who’s a physician has been really helpful since I can interact with and learn from so many different physicians.

A: What are the big goals and milestones you’re looking to hit in the short term? Long term?

J: Raising another round of funding that allows us to get to market in the nursing home space. Once we do that we’ll be reaching a sufficient install base so that the data gathered can be used to predict medical events for nursing home patients. From there we’re releasing a consumer product for the home that does the same thing.

Get in touch with Jeff at jeff@vsensemedical.com

Taking Control of Cellular Protein Output: an Interview with Paul Feldstein of Circularis

Taking Control of Cellular Protein Output: an Interview with Paul Feldstein of Circularis

Cellular protein production is used across industries to create products for medicine, consumers, research, and more. However, the technology to do so in the most efficient and effective ways has lagged behind production.

Circularis is using their expertise in discovering, analyzing, and evolving cellular promoters in order to regulate protein production with revolutionary precision. I talked to the company’s CEO, Paul Feldstein, about his team’s expertise, taking this advanced technology from academia to startups, and how Circularis will push biotech forward. Check out his pitch live on February 4th on IndieBio’s Demo Day Livestream!

A: Tell me about your background, how did you get involved in the biotech space?

P: I received my PhD at UC Davis in biochemistry and worked in one of the pioneering ribozyme (catalytic RNA) labs. So my background is in RNA biochemistry, and I have been working on research with the most primitive molecular parasites. These are made of RNA, are even simpler than viruses, and have led to the development of useful molecular tools.

A: What problem are you working to solve with your company, Circularis?

P: For years we’ve been thinking about better ways to find promoters since the current tools are cumbersome and hard to work with. Our technology can discover, analyze, and evolve promoters. There’s a lot we can do with that. We can regulate protein production to increase or decrease cellular output. We can do diagnostics to see how cells change in disease states. We can do therapeutics. Ultimately, a cell has to turn on a genetic program to respond to signals and that program is driven by promoters.

A: If you could only pick one thing to validate your reason for forming a startup, what would it be? In other words, what would be the single biggest indicator to you that you are doing the right thing?

P: We want to help great companies become even better. There are a lot of companies that are making proteins and facing challenges. We think we can make them better and drive the field of biotech forward.

A: How do you think success can change your industry?

P: Microbes, plant, and animal cells are precision instruments that everyone is trying to manipulate. Right now the whole field of biotech is trying to use sledgehammers to control these small precise instruments. We’re making precision tools to actually control cells effectively.

A: How is your team uniquely able to tackle this? What’s the expertise?

P: My experience in biochemistry is allowing us to develop the fundamental technology. Jim and LeAnn know how to apply it to plant and animal cells. We have decades of experience developing these tools from scratch and working together.

A: Any big lessons learned transitioning from academia to startup entrepreneurship?

P: It’s an entirely different world. I’m used to being judged only on the basis of the science in academia. In startups people think the science is interesting, but they really want to know how we’re going to make money and be a viable company. This a big learning curve and means we’re learning a totally different language. Academics talk for a long time about details, and here we’re giving five-minute pitches to explain everything.

A: What are the big goals and milestones you’re looking to hit in the short term? Long term?

P: In the short term we aim to successfully complete our pilot projects and develop longer term ongoing business relationships with our customers. In the long term, we want to expand our repertoire of organisms that we work on from only microbes to include mammalian and plant cells.

Get in touch with Paul at pafeldstein@circularisbiotech.com

Putting Biological Supercomputers in the Palm of your Hand: an Interview with Oshiorenoya Agabi of Koniku

Putting Biological Supercomputers in the Palm of your Hand: an Interview with Oshiorenoya Agabi of Koniku
Putting Biological Supercomputers in the Palm of your Hand: an Interview with Oshiorenoya Agabi of Koniku

As ambitions to create newer and faster supercomputers grow, so do the challenges. Increasing computational power comes with demands of scale, stability, and accessibility.

Koniku is a startup working to solve this by harnessing the power of biological neurons to create the next generation of supercomputers. I talked with Koniku’s CEO, Oshiorenoya E. Agabi, about his story, goals for Koniku, and changing the science of computation. Check out his pitch live on February 4th on IndieBio’s Demo Day Livestream!

A: Tell me about your background, how did you get interested in the biotech space?

O: I was born and raised in Lagos, Nigeria where I also did my bachelors. While getting my master’s in physics I remember a Russian professor telling me that if I wanted to make an impact I needed to go to a field where not a lot has been done and there’s opportunity to quantify everything. Following his advice, I took a course in neuroscience and knew right away this was it. Since then I’ve been working in computational neuroscience – quantifying how neurons function, engineering how to talk to them, and building tech out of them. After writing my master’s thesis on using biological neurons to do computation, I went to Imperial College in London to do my PhD in Computational Neuroscience and Bioengineering. That’s where I founded Koniku.

A: What problem are you working to solve with your company, Koniku?

O: The question is how to build a truly cognitive system. To do larger applications, we currently have to build massive and unscalable server farms. If we ever want to get to the point of doing significant computations we have to move away from the silicon paradigm. Koniku eventually aims to build a device that is capable of thinking in the biological sense, like a human being. We think we can do this in the next two to five years. We’re currently engineering neurons such that they are sensitive to particles in parts per trillion. With our computational backend, we’ll have a device that can be used for particle detection and be used for industrial, military, and agricultural application.

A: If you could only pick one thing to validate your reason for forming a startup, what would it be? In other words, what would be the single biggest indicator to you that you are doing the right thing?

O: We want to prove  we can connect to neurons and structure biological systems, and have the ability to package that.

A: How do you think success can change your field?

O: If we’re just 60% successful as a company, we’ll change computing as we know it. We’ll see a massive shift away from the silicon computing industry. Similar to what silicon did for the information age, we’ll do this for biotech.

A: How is your team uniquely able to tackle this? What’s the expertise?

O: We have a wealth of business and technical experience and expertise. This is a massive undertaking so we are still looking for more people with experience in computational neuroscience.

A: What’s the biggest challenge you’ve encountered so far?

O: In the near term it has been delivering game-changing devices and generating early revenue. We don’t want to just move fast and break things, we want to deliver an amazing product at the same time. Since we’re building a device we can’t just update it as needed. It has to perform exceptionally well every time a customer or user buys a product. We want to deliver an exponential advantage.

A: What are the big goals and milestones you’re looking to hit in the short term? Long term?

O: In the short term we would like to start delivering our sensing controls and computing devices to customers in the aviation and pharma sectors that have signed with us. We want to deliver an amazing product.

Our long-term goal is building a strong user base that lets other people make money off our devices. In the next two to five years we want this base of people programming and building separate applications for our device. It will be a true platform technology that people can build secondary products out of.

Get in touch with Osh at agabi@koniku.uk

Creating a pick and place machine for DNA synthesis: an interview with Jeff Clayton of Genesis DNA

Creating a pick and place machine for DNA synthesis: an interview with Jeff Clayton of Genesis DNA
Creating a pick and place machine for DNA synthesis: an interview with Jeff Clayton of Genesis DNA

In recent years we’ve seen an exponential ability to sequence DNA. What in the recent past it cost billions of dollars is now available for under one thousand dollars. However, the cost of synthesizing new DNA is still prohibitive and causing bottlenecks in research and industry.

Genesis DNA is working to create new methods to standardize DNA synthesis and make it cheaper, faster, and more reliable. I talked to the company’s CEO, Jeff Clayton, about his background, lessons from creating a start up, and Genesis DNA’s goals. Check out his pitch live on February 4th on IndieBio’s Demo Day Livestream!

Tell me about your background—how did you get interested in the biotech space?

I started off in chemistry as an undergrad since I always had an interest in biochem and biology, but ultimately went in the engineering direction for grad school since I thought there were a lot of opportunity for devices. I saw merging silicon and biological systems as a way to get the best of both worlds. My co-founder, David, also has a chemistry background but went more towards bio-engineering. The idea of Genesis DNA was born from a combination of his expertise in biology and mine in engineering.

What problem are you working to solve with your company, Genesis DNA?

We are developing a next-gen platform for DNA synthesis to standardize the process of assembling genes. The current assembly process is unpredictable. Every time you want to build a gene you have to design and build all the building blocks. This makes it difficult to know how long it will take and how successful a particular build will be. By standardizing the process it becomes easier and more cost effective to build custom DNA sequences.

If you could only pick one thing to validate your reason for forming a startup, what would it be? In other words, what would be the single biggest indicator to you that you are doing the right thing?

Our customer testimonials. It’d be a lot more difficult to work on this problem if we didn’t have people telling us all the time that today’s tools are still inadequate. We’ve heard from almost every biotech company that we’ve talked to that the ability to write DNA has become a huge bottleneck for research.

How do you think success can change your industry?

A great tool for DNA synthesis can do for biology what great microfabrication tools did for electronics. They increase freedom of design and make ideas more accessible and affordable to explore.

How is your team uniquely able to tackle this? What’s the expertise?

Ultimately we’re building a tool that builds biology so the expertise needed is heavy on both the biology and engineering sides. Which our team has.

Any big lessons learned transitioning from academia to startup entrepreneurship?

At IndieBio we’ve parallelized our research much more than we ever did in grad school. In a startup you don’t have the luxury of trying one thing at a time and waiting like in academia. This has been a really gratifying experience because you can achieve a lot of key metrics and get key results much faster.

What’s the biggest challenge you’ve encountered so far?

Besides the tech, learning how to effectively communicate science and its significance to the outside world. It’s not something you get a lot of practice at doing in academia.

What are the big goals and milestones you’re looking to hit in the short term? Long term?

In the short term we’re aiming to validate our chemistry for the synthesis of a short DNA sequence. Our ultimate goal is to greatly simplify the way researchers get DNA. We want it to be delivered in less than a week for simple or complex sequences and an order of magnitude cheaper than what is currently available.

Get in touch with Jeff at jeff@genesisdna.com

Unlocking Gene Therapy for the Masses: an interview with Ryan Pawell of Indee

Unlocking Gene Therapy for the Masses: an interview with Ryan Pawell of Indee

The science of gene therapy poses one of the greatest technical challenges in modern medicine. Researchers and industry face the significant challenge of introducing new functioning genes into cells, and doing so in a scalable and affordable manner.

Indee is a biotech startup developing new tools for gene therapy to bring it to the masses. They’re  currently part of IndieBio’s second class of startups. I talked to the company’s CEO, Ryan Pawell, about his story, transitioning to entrepreneurship, and the future of Indee. Check out his pitch live on February 4th on IndieBio’s Demo Day Livestream!

A: Tell me about your background, how did you get interested in the biotech space?

R: I have a B.S. in Mechanical Engineering. My senior project was developing tools for cervical spinal surgeries, and I also worked for a couple years on portable oxygen concentrators to treat breathing disorders like COPD. I realized I wanted to get into stem cells, and thought microfluidics was a good route since it was a high value and low-cost tech for stem cell manufacturing with good potential business models. All that led me to do a Microfluidics PhD in Australia.

A: What problem are you working to solve with your company, Indee?

R: Indee is solving scalable gene delivery. This is really important for gene therapy since right now scales are limited to just a few thousand wealthy patients per year. Creating a scalable process can treat tens of millions of people that die from multiple diseases every year.

A: If you could only pick one thing to validate your reason for forming a startup, what would it be? In other words, what would be the biggest indicator that you are doing the right thing?

R: I just don’t like having a boss. My first job was working for myself, and it was more rewarding and flexible. The whole point of this startup is to work on something that helps other people while enjoying the benefits of self-employment. Right now at an early stage startup there aren’t really any benefits of self-employment [laughs].

A: How do you think success can change your industry?

R: We’re looking to ultimately manufacture these life-changing therapeutics for tens of millions of people every year.

A: How is your team uniquely able to tackle this? What’s the expertise?

R: Our team consists of two full-time employees (myself included), eight advisors, some high-value contractors, and IndieBio/SOSV. Our expertise is in both microfluidics, fluid dynamics, and cell biology. My co-founder, Matt, handles the biology and business development. I do the engineering and operations. Having a co-founder with complementary and effective skillsets has made a huge difference.

A: Any big lessons learned transitioning from academia to startup entrepreneurship?

R: Startups are exciting, challenging, and fast-paced. Academia… not so much.

A: What’s the biggest challenge you’ve encountered so far?

R: It can be tough to stay on top of everything. We have a great two person team but when the work piles up three would be nice [laughs].

A: What are the big goals and milestones you’re looking to hit in the short term? Long term?

R: Our big goals are to verify our technology and sign our next investment. In the short term we want to show gene editing and be able to process 10,000 samples a week. This is proof of concept for a 50-fold improvement over current manufacturing scales without spending 43 million dollars for a manufacturing plant, because we don’t have that. In the long term we’d like to use that system to manufacture 10,000 therapeutics per week so we could treat half a million patients per year.

Get in touch with Ryan at rpawell@indeetx.com

IndieBio and SpaceGAMBIT – Announce Partnership to Accelerate Space Biology

IndieBio and SpaceGAMBIT - Announce Partnership to Accelerate Space Biology
IndieBio and SpaceGAMBIT - Announce Partnership to Accelerate Space Biology

Today we’re delighted to announce our partnership withSpaceGAMBIT to accelerate Space Biology, we believe one of the weakest links in making humanity a multi-planetary species is the understanding and development of biotechnologies which could be deployed in every aspect of life in space.

In this partnership, IndieBio will be supportingSpaceGAMBIT with open source capacity building biotechnologies that have potential applications in space and SpaceGambit will be supporting IndieBio in developing novel biotechnologies that could have novel applications in space but would require experienced space industry partners.

SpaceGAMBIT democratizes space technology by funding open-source projects that tackle everything holding us back from being a spacefaring species. They work with partners and the maker movement to find out what’s keeping us stuck on Earth, engage the crowd to find solutions, and share the results with the world. They’ve worked with NASA and the White House, and are now adding IndieBio to their list of partners to help hack humanity to the stars.

Onwards and Upwards from your friends at IndieBio and SpaceGAMBIT!

http://www.spacegambit.org/

http://sf.indie.bio/

Reimagining the Future with Biology

Reimagining the Future with Biology
Reimagining the Future with Biology

Today, Bolt Threads, previously Refactored Materials announced that they’d raised $40M in their latest round of financing and expect to have their yeast derived spiders silk (10x stronger than steel per weight) available for sale by 2016.

The new Bolt Threads biomaterial will have applications we can only now dream of and likely many, we have yet to dream and this is only the beginning of the new bioeconomy that’s being built around us, in university, commercial and biohacker labs around the world.

Over the last four months, our first IndieBio class in San Francisco (IB1), we’ve seen what’s possible when scientists, innovators and pioneers join together to accelerate how we build our world with biology.Amazing companies, products and services have been built in a spirit of camaraderie and collaboration (both within the first cohort and with the broader community) which is stark contrast to many of the innovation silos we’ve all experienced in academia and industry.

Our first time founders have worked side by side with each other and veteran entrepreneurs and scientists (across all industries) to reimagine a world in which previously intractable problems might now be solved with applied biology.

  • Pembient has shaken up the world of Rhino conservation by challenging the status quo, sometimes education isn’t enough, if you have an approach which is failing, change it.
  • Clara Foods is helping us to reimagine food with beautiful and delicious Meringues, to start with, which have excited Chef’s globally as a new way to innovating in the kitchen humanely!
  • Extem are powering regenerative medicine with the first and most extensive global stem cell bank, supplying researchers globally with the cells they need to deliver on the promise of regenerative medicine, helping patients in dire need.
  • Arcturus Biocloud have launched the first consumer biotech cloud service, enabling applied biology and science from anywhere with a simple user interface with users signed up on the platform from over 100+ cities globally (and growing).

These are only a few of the companies who you’ll see presenting on our first Demo day, June 11th, in San Francisco, in which we invite you to attend, our general admissions are now sold out due to massive interest but this event is also for the broader global community and will be live streamed, so whether you’re in SF, Mumbai, London or further afield we invite you to join us! We’ll be sharing our livestream calendar invite on @indbio soon!

If you’re a scientist, entrepreneur or biohacker and have a re-imagined vision of the future, built with applied biology that you’re currently working on or would like to build, we’d also love to invite you to apply for our $250k funding package, we’ll be funding our next class of 15 new companies in SF in September and our first early applications deadline is June 30th 2015, get your application in ASAP, don’t wait for the deadline, as we’re currently interviewing teams for consideration into the next class!

The Story of Fire, Stone, and Biology

The Story of Fire, Stone and Biology
The Story of Fire, Stone and Biology

It was the Titan, Prometheus, who created man.

Prometheus glanced down on the Earth and noticed that rainwater was making nature live, without it trees and bushes died, giving way to desert.

Prometheus discovered the power of earth and water, so he mixed clay with water, moulding the shape of the first man.

It was in the shape of the gods that he created mankind.

For some, the well trodden mythology of the theft of fire from the gods by Prometheus, to share with humanity is seen as both a great sacrifice and a parable, that not even a titan is allowed to share the tools of the gods.

But fire and stone isn’t the only lesson which the Titan Prometheus can teach us, even in his punishment, chained to a rock, suffering in agony as his liver is eaten by an eagle, eternally, he gave us one more gift.

Regeneration.

It appears that even the ancient Greek’s knew of the liver’s ability to regenerate after injury, it’s an ancient learning hinted through mythology and yet, only recently have we really started to unravel the mechanisms of regeneration, not just in the liver but across the bodies of humans and other multicellular organisms.

“In Biology, Regeneration is the process of renewal, restoration, and growth that makes genomes, cells, organisms, and ecosystems resilient to natural fluctuations or events that cause disturbance or damage”

So what do we, as humanity now know about regeneration?

  • We know that our DNA has built in repair mechanisms
  • We know that our cells have “shredders” which are constantly on the search for damaged proteins, destroying the old to ensure our cells continue to function.
  • We know that our tissues are constantly replenished by adult stem cells, as we get broken down by wear and tear, our bodies do an incredible job of repairing us, until they can’t.
  • We know that some organisms still retain their regenerative capabilities, long past the embryonic stage.
  • Flatworms can be cut in half and regenerate entirely, salamanders retain the ability to regenerate organs, eyes, tails and many more of their body parts and humans retain a limited amount of regeneration (although not the ability to regenerate damaged organs or limbs… yet).

So can we 3D print human organs yet?

No, not yet but we’re starting to understand some of the most complex technologies on this planet, DNA, proteins, cells and our living tissues, which turn chaos into order.

We have been able to strip hearts of cardiac cells, repopulate their scaffolding and restart them, they beat.

We’ve been able to keep lungs alive outside of the human body for a few hours (previously they’d foul within minutes), match and transplant hearts, lungs, kidneys and more successfully into living donors and this is just the beginning.

The technology of life

We’re beginning to understand the language of the gods, DNA is the code in which life is written and as we remember, in memorial, those who have lost their lives or have been severely wounded in battle for our freedoms, we can now offer them a glimmer of hope.

This year, the Department of Defense launched a new Organ preservation initiative supported by the Organ Preservation Alliance and New Organ. Companies like Organovo are now printing more and more complex tissues, we can, as of today, grow organoids (small organ like tissues) of most major human organs and researchers at universities across the globe are diving deeply into regenerative medicine and the development of a new area of science, organogenesis.

Prometheus, on that rock of eternal damnation, gave us one last gift, regeneration.

IndieBio secures partnership with Transcriptic to Accelerate Biology

IndieBio secures partnership with Transcriptic to Accelerate Biology
IndieBio secures partnership with Transcriptic to Accelerate Biology

Media Contact: Dorothy Lou Bailey, dorothylou@transcriptic.com

San Francisco, CA – IndieBio, the leading global biotech accelerator, today announced an exclusive partnership with the robotic cloud laboratory platform Transcriptic for up to $60,000 per year in free services for the companies they fund and accelerate.

The Transcriptic credits will be divided evenly among the companies in each new class at IndieBio. IndieBio currently has 12 companies in their inaugural San Francisco based class and plans on admitting approximately 30 per year (15 per cycle). This partnership is the first in a series of announcements by IndieBio as they prepare to open applications for their next application cycle.

“We are incredibly excited to be announcing this partnership with Transcriptic and working to accelerate biology together. From the beginning, our aim in founding IndieBio has been to give scientists and entrepreneurs a new, lower cost path to innovation in Independent Biology. We evolve, like the companies we fund, so expect to hear some very big, additional news from us this week on this continued acceleration.”

Ryan Bethencourt, Program Director, IndieBio

“We believe cloud science will enable a better future and we’re excited to partner Transcriptic to help enable that future.”

Arvind Gupta, Partner SOSventures and Founder IndieBio

Transcriptic, a venture-backed on-demand science company that enables researchers to run their experiments remotely through a web based interface, has democratized access to biotech automation. Transcriptic can execute a wide range of common molecular biology workflows with high repeatability and low cost. From their 10,000 sq. ft. Menlo Park facility, Transcriptic services clients spanning the globe with always-on infrastructure. “The platform enables scientists to remove the intense manual labor that encompasses 70% of research and development time. While useful for all, this is especially valuable for small companies who need reliable data and longer capital runways.”

IndieBio views biology as technology and invites applicants from across the life sciences from food, biomaterials to new therapeutics, melding biology, software and robotics to apply for the spring and winter batches. IndieBio funds and accelerates company’s developing solutions with biology to some of the world’s most intractable problems with the broad vision of moving our world from one of scarcity to one of abundance.

“Transcriptic is pivotal to accelerating our R&D program. They allow us to conduct meaningful and reproducible experiments without having to incur the expensive capital cost of buying additional scientific equipment or hiring additional staff to conduct these crucial experiments.”

Leo Wan, CSO & Co-Founder, Ranomics (IB1)

Extem – The future of stem cell based therapy.

Extem - The future of stem cell based therapy.
Extem - The future of stem cell based therapy.

We are a group of scientists and engineers designing a new system to mass-produce adult stem cells. Our pilot project is the rapid large-scale production of mesenchymal stem cells, for use in basic research and bioprinting.

With federal science budget cuts of almost $600M in Canada alone between 2008 and 2013, it is becoming increasingly difficult for researchers to fund their experiments. We are removing the difficulty and expense of isolating primary cells by delivering ready-to-use stem cells straight to your lab.

Our ultimate goal is to empower researchers and other enabling technologies to revolutionize bio-engineering through the integration of stem cell tools. In pioneering a global source of stem cells, we aim to exponentially accelerate stem cell research and open the gate to new stem cell-based therapies in regenerative medicine and tissue engineering.

-the extem team

Stem cells, research, and bio-engineering – the future of stem cell based therapy.

#Sciencehack : Transforming Science through Collaboration

Transforming Science through Collaboration
Thanks to Connor Dickie of Synbiota for this guest post.  I can’t wait to get my hands on a kit!
hands
It’s often said that collaboration is key. Why then, has collaboration in the life-sciences been so limited, when the products (medicine, materials, food, & fuel) are so important? The problem can’t be technical, It’s almost 2015! We’ve got more free and excellent collaboration tools at our fingertips than ever. Why then, is collaboration in life-science not the standard?
A lot of it has to do with an old mentality, one that was born when bringing a biotech product to market would cost hundreds of millions of dollars and take a decade to complete. Just think, if you had a company that spent what amounts to a mountain of cash, and a significant part of a team of highly-paid researchers lives on a single product, you too would want keep your trade secrets pretty safe.
What happens then when bringing a biotech product to market costs less than $50k and takes only a year or two of development, or even less? This dream has not exactly happened yet in biotech, but it regularly happens in the IT sector, particularly with software, so let’s look there for insight as to how things might play out for biotech in the near future if collaboration and the Open Science model becomes as widely adopted as Open Source.
Remember (if you can) what computing was like in the 60’s and 70’s. Back then, a computer took up and entire room, and cost millions of dollars. Chances were that if you had access to a computer you had a PhD in computer science or math, and you were employed by the military, a research institute, or a large bank. Software was expensive, scarce, and cryptic.
Open Source Movement
By the mid 90’s all this had changed. Computers were ubiquitous, and the internet was connecting PCs and people from around the globe. Armed with the right tools and the idea of free and open software, cadres of computer scientists and developers were joined by students, hackers, designers and artists. This ragtag group started to coalesce into a community known as the Open Source Movement, which was built on the idea that by working together, a network of independent people could challenge the status quo. And boy did they ever!
Linux is often referenced as the poster-child of the Open Source movement, and for good reason. Since it’s early days, the dream of Linux was to replace Unix (the standard computer operating system used in mission-critical environments, easily costing over $10,000 per license) with a free and open alternative. Naysayers felt that this dream seemed insurmountable at best, and likely just a crazy pipe dream. But the power of a dedicated community, linked by the internet, wielding accessible tools, and each contributing a small part of the solution, changed the computing world forever.
What would it look like if a similar story played out in biotech? What would the benefits be to society be? Could we realize the dream of free, open, and trustworthy medicines? What if we could do the same for sustainable materials, food, and fuels? What would the world be like then?
While exciting to think about, there is a lot of work that needs to be done before these questions can be fully answered. There are some fundamental issues in biotech that make distributed collaboration difficult, and this is primarily the problem that we’ve addressed at Synbiota. We’ve taken lessons from the Open Source software movement and have applied them to biotech, and already we’re seeing exciting productivity gains for our users.
Open Science Movement
One of the biggest innovations we’ve brought to the Synbiota community is vastly increased project reproducibility when both our tools and integrated wetware are used together. By combining a common suite of free web-based, tools and an inexpensive, easy to use biological wetware standard, researchers and developers are able to hit the ground running, often producing results in weeks for just a few hundred dollars.
With this reduced cost and timeline to create working Synthetic Biology projects, we’re seeing an increasing amount of projects on Synbiota that are released to the public under a creative commons license – effectively Open Sourcing the science, which feeds back into the system making it easier to reproduce and extend existing projects, or even develop new projects based on previous work.
An example of Open Science in motion is the #ScienceHack initiative that we launched at the 2014 SXSW Interactive Festival. #ScienceHack demonstrates that using a common Synthetic Biology wetware kit, and Synbiota in a collaborative manner, it’s now possible to create real medicine at a cost a few orders of magnitude cheaper than Big Pharma.
Violacein
Each #ScienceHack event uses the “Violacein Factory” wetware kit and a shared set of protocols that are freely available under a Creative Commons license. While the first #ScienceHack participants had only their hypothesis to inform their work, each subsequent #ScienceHack was able to leverage the Open Science results of previous #ScienceHacks – resulting in the first ever Synthetic Biology medicine created by a loosely affiliated group of scientists, artists, hackers, and students.
While the efforts of the Synbiota community have yet to change the status quo in biotech, #ScienceHack has successfully demonstrated that it is possible for motivated independent researchers to collaborate, and have a positive effect on the world using Synthetic Biology technology.
We hope this is just the start of something big. We continue to provide the tools, but it’s up to the community to turn this into a global movement that can offer an effective and open alternative to the status quo.