25
Mar
2021

A Glimpse Into a Price-Controlled Future

Luke Timmerman, founder & editor, Timmerman Report

Imagine if H.R. 3 — the drug price-control bill that has significant support in the House of Representatives — were to become law.

What would it look like if some of the bill’s provisions, like indexing US prices to 120 percent of prices in Europe, were enacted?

How would entrepreneurs adapt? What types of drug discovery programs might be prioritized, and which ones cast aside as impractical? How would management teams have to adjust their strategy, and pitches, to successfully attract investors? How would certain patient groups be affected? What would it mean for the sector?

There are many questions. It’s fascinating, when you dig in, to think through the implications of this thought experiment.

Students at the University of Texas Southwestern Medical Center were challenged to think along these lines through a business plan competition on Mar. 20. Small teams of postdocs, grad students and MBAs were asked to think of a product profile, a relevant disease indication, and — assuming success in clinical development and regulatory approval — a price that could withstand the hard calculations of Incremental Cost-Effectiveness Ratios (ICER) and Quality-Adjusted Life Years (QALY).

The goal was to come out with an investment pitch that investors might buy into.

RA Capital Management, the Office for Technology Development at UTSW, Biotech+ Hub at Pegasus Park and McKinsey supported the competition. RA Capital managing partner Peter Kolchinsky served as a judge with Jeb Keiper, CEO of Nimbus Therapeutics, and Sara Nayeem, partner at Avoro Ventures. I observed.

The students clearly did their homework.

The results were eye-opening.

One group imagined it had an antibody-drug conjugate for lung cancer that delivered an 85 percent Complete Response rate, few side effects, and an average improvement in life expectancy of 10-11 years. Such a drug could be administered for about two years on average, and priced at $210,000 a year in Europe and $250,000 a year in the US, they figured. At that price, the fictional company would beat existing PD-1 inhibitors from Merck and Bristol Myers Squibb on a QALY-based comparison.

Kolchinsky shook his head. Sure, payers would pay for that. But there is no such drug on the market, or in R&D.

Peter Kolchinsky, managing partner, RA Capital

“We never see this kind of efficacy,” Kolchinsky told the students. Turning to his fellow judges, he said: “What they are asking us to do is stop investing in plausible, realistic science and start investing in fantasy.”

Another group sized up a new treatment for irritable bowel syndrome with constipation or diarrhea, and imagined generating three times as much sales volume in Europe to make up for an anticipated shortfall in the US.

Next came a team with an mRNA vaccine for HIV that elicits broadly neutralizing antibodies. The students thought the vaccine could be given to everyone in the US. They considered a low price for a national population and a higher price for a subpopulation deemed to be “high risk.”

The team concluded that it didn’t make sense to develop such a product as a low-volume/high-priced ($9,000) product for the high risk population. Forget about it.

A few days later, I asked the judges to reflect. How were students forced to adapt to the new environment? What did it tell us?

Kolchinsky wrote:

“The participants made an admirable effort, but every single team was forced to make unrealistic assumptions to try to make their business pitches seem fundable despite the limitations of price controls.

“A lot of the submissions were based on actual drugs in development by real companies. But the actual programs are funded because those companies and investors have an expectation that, if successful, those drugs will get reimbursed at prices close to those drugs we have on the market now, which are all higher than prices that HR-3 would allow. And when these teams tried to win funding for these programs but asked investors to accept a much lower price, they couldn’t. Some teams resorted to asking the judges to imagine that these drugs were better than they actually were, had a higher chance of succeeding that they actually have, or would treat more patients than could possibly be true. Bottom line, price controls killed investment in the R&D that is actually possible and forced teams to resort to making promises they couldn’t keep…”

If health economists who run conventional cost-effectiveness are put in charge of setting price, innovators probably won’t be able to win funding from any knowledgeable investors. There may some investors who fall for a pitch claiming a high probability of success and larger market than is real, but in time they will lose enough money to realize the price controls set rewards too low for the risks involved in making new medicines.”

Jeb Keiper, CEO, Nimbus Therapeutics

Jeb Keiper of Nimbus said the pressures would cause entrepreneurs to overpromise. “Like snake oil salesmen, [they] promise a cure in every bottle,” he said. “The constraints forced business plan competitors’ strategies to subvert price-control legislation by promising miracles.” 

That was one survival strategy. The winning team had a different plan. It pitched a reformulation of a proven drug. But in a hostile pricing environment, the students said they would need to accept a lower valuation in the next funding round — crushing the ownership stakes of earlier investors.

Keiper ruefully asked the team who was the CEO. That person would likely be fired.

“The implications are chilling,” Keiper wrote. “Existing biotech investors (including large scale mutual funds and pensions which many Americans have their savings in) get gutted, new medicines stop being developed for patients that need them, and the public only gets incremental improvements that industry watchdogs rightly hound against.”

Pain would be spread among entrepreneurs, investors, and patients. The patients most likely to bear the brunt, though, would be ones with the greatest needs.

Sara Nayeem, partner, Avoro Ventures

Sara Nayeem of Avoro Ventures wrote:

“Drug development for diseases where there have been many historic failures would be especially hard to fund.  For instance, pancreatic cancer is a devastating diagnosis for which we need breakthrough drugs; but because the historical probability of success is much lower than in other areas of oncology (per BIO, a drug for pancreatic cancer entering Phase 1 trials has only a 1.1% chance of approval), investors would need to know that a drug that shows needle-moving efficacy would be reimbursed at a higher price than the average cancer drug…groups like ICER don’t take into account the low success rates in certain diseases; nor do they quantify the value to society into perpetuity of branded drugs going generic.”

Some readers might think this boils down to the same old industry talking points about price controls causing investment to dry up.

But the students weren’t trying to score political points. They were imagining a future drug pricing environment, and trying to prepare.

It was a useful and humbling exercise. Lawmakers would be wise to think a few steps ahead, like this, when considering big changes to a complex market like this with so much at stake.

That’s not to say we should slavishly stick with the status quo. Biotech as an industry is creative and resilient under pressure. Management teams were forced to cut expenses and operate in more lean ways in the Great Recession years of 2008-2012. People in Congress warned the Affordable Care Act would crush incentives for innovation. That didn’t happen.

Instead, a crop of battle-tested, productive companies emerged. We have reaped the harvest of those innovations from the early 2010s. Year by year, the FDA approvals tell the story.

Markets have their vicissitudes. But the secular trend in biomedicine is upward. There’s a lot here in academia, government and industry that’s worth preserving and building upon.

Our elected leaders ought to guide regulations and set guardrails that keep companies honest, with an eye toward preserving a dynamic environment where startups can come along and knock off incumbents.

Kolchinsky, in his book “The Great American Drug Deal,” advocates for contractual genericization as one way to provide balance to pharmaceutical markets that are too often dominated by companies with incentives to primarily protect aging franchises. Many of these aging franchises should have gone generic years ago. Too many drugs, because of patenting games or other perverse incentives that involve middlemen, command sky-high prices that can’t be justified. All of these shenanigans end up wasting money, harming patients and discouraging entrepreneurs.

This publication believes in standing up for the proverbial little guy. Policymakers, health economists, and industry leaders should take action to rein in the excesses of the big guys, without forgetting that little guy (or gal) with a startup dream. That person should have an incentive, and a chance. That person shouldn’t become collateral damage.

If we want to create an environment where scientific entrepreneurs can flourish, we can do it. With scalpels, not sledgehammers.

 

Damaged Public Trust

If you missed Tuesday’s Frontpoints, it predicted this week’s AstraZeneca vaccine debacle. The NIH statement of Mar. 23 pointed out the company’s press release claiming 79 percent efficacy was based on outdated data. The NYT and Washington Post got ahold of a seething letter from the Data Safety Monitoring Board. AZ tried to do some damage control in a revised press release on Mar. 25, which dragged efficacy down to 76 percent. Given the company’s recent pattern of bad behavior and incompetence, I plan to read every line of the FDA briefing documents, and listen to every minute of the FDA advisory committee hearing for this vaccine candidate.

This is a test. Regulators, doing their jobs, will go over everything with a fine-toothed comb and review the application with full transparency. Then we can make informed decisions.

A lot of leeway has been granted to companies in this emergency, born from necessity and a generous spirit of “we’re all in this together.” Some standards about not doing science by press release, and not accepting company statements at face value, have gone out the window in the name of expediency. We put a lot of trust in companies and they have mostly delivered the past year.

But when a company abuses trust repeatedly, the credibility of science and industry itself gets called into question. AstraZeneca needs to make things right. This isn’t one of those cases where PR window dressing or a carpet-bombing of advertisements will make controversy go away. They need to fix things.

This Week in Drug Pricing

Are you a biopharma executive who breathed a sigh of relief about how the drug pricing pit bulls snarling at the industry have been neutered during the glory days of vaccine development?

Not so fast. Public opinion can be fickle.

The AZ imbroglio (see above) feeds into the confirmation bias of the industry’s harshest critics. While a majority are grateful for the vaccines, we still have perverse incentives in our health insurance system, healthcare profiteering, millions of our most vulnerable are being left in the cold, and we still have an information commons that downgrades fact-based discourse and amplifies conspiracy mongering.

Without systemic fixes to how we pay for healthcare — and how we provide fair access to healthcare for everyone — we could easily fall back into the brain-dead rhetorical rut about sticking it to those evil price-gouging drugmakers.

See this NYT op-ed, headlined “Taxpayers Fund Research and Drug Companies Make a Fortune” in which a patient advocate imagines a day when COVID-19 vaccines might cost money (the horror!).

I agree individuals shouldn’t be paying out of pocket for this most valuable preventive medicine. We should be paying for it with our tax dollars. We should be grateful about it because we’re getting an amazing bargain from vaccines that will get the global economy moving again and bring tremendous peace of mind.

Science Policy

President Biden is showing vision and guts. Who says we can’t solve problems, so why bother trying? Biden isn’t buying that nihilistic garbage. He’s acting like a young man in a hurry. Let’s ask serious questions that citizens in a serious participatory democracy ought to ask. Like whether we invest enough in science as a country. What more could we be doing to advance cures, fight climate change, and create meaningful career opportunities for younger generations?

A big investment like the one suggested below inspire a new generation to be drawn to science, and to value science.

Science Features
  • Pandemic whistle-blower: we need a non-political way to track viruses. Interview with Rick Bright. Nature. Mar. 19. (Amy Maxmen)
  • What scientists do and don’t know about the Oxford–AstraZeneca COVID vaccine. Nature. Mar. 24. (Smriti Mallapaty and Ewen Callaway)
  • Unlocking the COVID Code. NYT Magazine. Mar. 25. (Jon Gertner)
Science of SARS-CoV-2
  • Dynamics of SARS-CoV-2 neutralising antibody responses and duration of immunity: a longitudinal study. The Lancet Microbe. Mar. 23. (Wan Chia et al Duke-NUS Medical School, Singapore)
  • Early Evidence of the Effect of SARS-CoV-2 Vaccine at One Medical Center. NEJM. Mar. 23. (William Daniel et al UT Southwestern)
Data That Mattered

Bristol Myers Squibb said it met the primary endpoint – progression free survival — in a Phase III clinical trial with its LAG-3 targeting antibody, relatlimab, in combination with nivolumab (Opdivo), the PD-1 directed antibody. The study looked at patients newly diagnosed with metastatic or unresectable melanoma. Overall survival data aren’t yet available. It’s the first time a checkpoint inhibitor for LAG-3 has passed a pivotal study.

Financings

Eric Schmidt, the former Google CEO, donated $150 million to establish a new center at the Broad Institute to support machine learning analysis of emerging biological datasets, including next-generation DNA sequencing, single-cell genomics, and advanced medical imaging. The Schmidt Center will be co-directed by Caroline Uhler, Associate Professor of Electrical Engineering and Computer Science and the Institute for Data, Systems, and Society at MIT and an associate member of the Broad Institute; and Anthony Philippakis, Broad’s chief data officer. 

Rivervest Venture Partners, with offices in St. Louis and San Diego, said it raised $275 million for Fund V to invest in early stage biopharma and medical device companies.

Seattle-based Eliem Therapeutics raised $80 million in a Series A financing led by RA Capital Management to develop treatments for chronic pain and depression. (TR coverage).

Emeryville, Calif.-based Zymergen, an industrial biotech company, filed an IPO prospectus to raise up to $100 million.

Cambridge, Mass.-based Apnimed raised $25 million in a Series B financing to develop a once-daily oral pill for obstructive sleep apnea. Morningside Ventures led.

San Francisco-based Ginger said it raised $100 million in a Series E financing led by Blackstone to improve mental healthcare offerings for employers, health plans, and strategic partners. (TR Frontpoints column on mental health, Mar. 4, 2021)

Cambridge, Mass.-based 1910 Genetics said it raised $26 million to advance its technology to design small molecules and protein drugs with AI and automated tools. M12 – Microsoft’s Venture Fund and Playground Global co-led.

Personnel File

Moncef Slaoui, the former head of R&D at GSK and leader of the Operation Warp Speed program for COVID-19 vaccines, was fired from the board of a GSK / Verily entity (Galvani Bioelectronics), and stepped down immediately from a new job as chief scientific officer of Centessa Pharmaceuticals, after GSK said it had learned of sexual harassment allegations and an outside law firm investigated the claim.

Amy Abernethy, the principal deputy commissioner of the FDA, will step down from her position next month. She has overseen a push to modernize the agency’s data practices.

Novartis said it’s closing down a factory in Colorado where it was planning to manufacture Zolgensma, the gene therapy for spinal muscular atrophy type 1. The company plans to shut down the plant in July, and lay off 400 workers. It apparently overestimated demand for the therapy, which it obtained through the acquisition of AveXis for $8.7 billion. (FiercePharma coverage).

Atul Dandekar joined South San Francisco-based Maze Therapeutics, a precision medicine company, as chief strategy officer. (TR coverage of Maze, Feb. 2019)

Josh Makower was named the new director of the Stanford Byers Center for Biodesign. He replaces the legendary founder of the center, Paul Yock, a medical device innovator. Makower will retain a relationship with NEA, in keeping with the center’s longstanding focus on academic-industry partnerships.

Merck promoted Caroline Litchfield from treasurer to CFO. She replaces Robert Davis, who is being promoted to president on Apr. 1 as part of a succession plan for him to take over from Ken Frazier as CEO on July 1.

Regulatory Action

Takeda Pharmaceuticals said its submitted an application for its dengue vaccine to regulators in Europe, and in dengue-endemic countries.

Our Shared Humanity

Bruce Booth of Atlas Venture wrote an unusually personal piece on his LifeSciVC blog Mar. 16. I let some time pass before asking him about the feedback from this article about divorce.

It was a rare example of a powerful business leader showing vulnerability.

If you missed it, this is a message that bears repeating. We should all try to be a little more empathetic to friends and colleagues at work who may be stressed by personal issues just beneath the surface. By listening a little more, and extending some understanding and grace, we do ourselves a favor and everyone around us.

I was really happy to hear Booth tell me yesterday that the feedback has been “uniformly positive.”

Hundreds of emails poured in. These weren’t the usual “Hey, nice article” notes that writers sometimes get.

Many of these correspondents got specific and personal, Booth said.

“They went on for paragraphs about a sick kid, a sick spouse, feeling unable to talk about it at work. There were other people who went through divorces, and talked about making it through. They were just sharing their stories. I got very emotional reading all the responses. It was really powerful.”

It took courage to write the article.

We men are taught to be brave from an early age, in a gladiatorial, competitive sense. Never let ‘em see you sweat. Bullies on the playground will bury you at the slightest hint of “weakness.” But at some point in life, we realize that “putting on your game face” at work, as Booth put it, isn’t always a show of strength.

Showing some vulnerability, and allowing people to be human beings with each other in your midst, can be a source of leadership strength.

The positive reaction to Booth’s article lifts my spirits. It reminds me that people in this industry are capable of many good things, in science and beyond.

22
Mar
2021

AZ Tells Us Its Vaccine is OK. Rebuilding Trust Will Take a While

Luke Timmerman, founder & editor, Timmerman Report

Credibility can be lost in a heartbeat.

It can take years to rebuild.

That maxim kept running through the back of my mind when reading the press release on the AstraZeneca Phase III clinical trial conducted with 32,449 participants in the US, Peru and Chile, in partnership with the NIH’s COVID-19 Prevention Network.

The top line was encouraging — 79 percent efficacy at preventing COVID-19 and 100 percent efficacy at preventing severe illness and hospitalization.

The company, following a rough couple weeks of rumors and innuendo, said independent data monitors found no increased risk of blood clots in people who got the vaccine.

“I’m thrilled,” said Ashish Jha, dean of Brown University’s School of Public Health, in reacting to the breaking news in Science. “This is the vaccine that I had always assumed would vaccinate a large chunk of the world.”

It’s a view rooted in practical reality. AZ’s is an adenoviral vector vaccine, given in two doses four weeks apart (or potentially further apart, according to UK officials). It’s manufacturable at global scale, easy to ship in refrigerators, and cheap — $2 to $6 a dose.

So far, so good.

I hope that everything in the company statement is right, and that there are no serious flies in the ointment. But despite our yearning for positive vaccine news, we have reason to withhold judgment until we get a good, hard look at the data.

This company has shot itself in the foot multiple times, damaging its credibility in the past year.

Consider the sequence of vaccine events:

May 21—company says it’s getting $1 billion from the Biomedical Advanced Research and Development Authority (BARDA) to develop, produce and deliver its COVID-19 vaccine in the US. Plan is to run a 30,000-participant Phase III trial, and deliver at least 400 million doses in 2020 and 2021. This was an assuring sign of a major league pharma company with a major league academic partner (Oxford), getting a major league vote of confidence from the richest and most scientifically powerful country in the world. At a time when mRNA was still seen as fairly speculative, this looked like a safe bet.

Sept. 6—company receives a report of an adverse event — transverse myelitis, an inflammation of the spine — in a vaccine participant in Britain. The company halts enrollment while investigating the adverse event, looking to see if its vaccine might have played a causative role.

Sept. 8—company has conference call with FDA officials, seeking clarity on what’s necessary for US regulatory approval. Company neglects to mention the transverse myelitis adverse event in the UK, and the decision to halt trial enrollment. The FDA was blindsided a few hours later when it heard about the revelations on the news. (The only reason we know this is because of a New York Times report from four months later). This confirms the absolute worst impressions critics have of the pharmaceutical industry. Not only that, but if you were working for the FDA and in the room that day, wouldn’t you be fuming that the company didn’t disclose the event, and didn’t discuss how to get to the bottom of the issue in partnership?

Sept. 9—As reports of the adverse event swirl, AstraZeneca CEO Pascal Soriot goes into a private conference call organized by J.P. Morgan for clients of the investment bank. From this well-controlled safe space, he assures everyone that the woman who suffered the severe spinal inflammation was improving and likely to be discharged from the hospital soon. We only learned this because STAT broke the story that day. Again, this was news of international biomedical significance, given in private to wealthy clients of JP Morgan — before the public.

Sept. 9—All AstraZeneca COVID-19 vaccine trials go on clinical hold as researchers seek to suss out what happened with the woman who got transverse myelitis.

Sept. 12—AstraZeneca vaccine trial resumes enrollment in the UK. Importantly, the big 30,000-participant study in the US, the one designed to yield the most rigorous evidence, remains on hold.

Oct. 23—the US FDA lifts the clinical hold, allowing AZ to resume enrollment of the 30,000-participant vaccine study. The clinical hold lasted more than six weeks – an eternity in a fast-moving pandemic. Pfizer/BioNTech, Moderna and J&J leaped ahead.

Nov. 23—AZ reports on a pooled analysis of 23,000 participants who got the vaccine in the UK and Brazil. Because of a dosing screw-up, a half-dose appeared to be 90 percent effective in a subpopulation of 2,100 subjects, while the full dose appeared to be 62 percent effective. The dataset, on its own, was a mess. Regulators here, in the best-resourced drug regulatory agency, already had good reason to be suspicious at the company for hiding a crucial piece of information. Now AZ is telling the world that its trial was bungled? There was no realistic path forward for the company to seek FDA Emergency Use Authorization on the basis of this unconvincing dataset.

Dec. 8—The NYT publishes its expose on AstraZeneca’s failure to disclose the adverse event to the FDA in September. Days later, the FDA approved the first two COVID-19 vaccines – one from Pfizer/BioNTech, the other from Moderna.  

Feb. 8.—South Africa health officials halt administration of the AZ vaccine, saying it was offering “minimal protection” against the B.1.351 variant of SARS-CoV-2 that was in wide circulation by then.

Feb.15—the World Health Organization authorizes the AZ vaccine for use in low and middle-income countries.

Mar. 15—European countries temporarily halt administration of the AZ vaccine amid anecdotal scary reports of blood clots in people who received the vaccine. Despite no compelling evidence to point to the vaccine causing those adverse events, health authorities in multiple countries, fearing the worst, shut down mass vaccinations with the AZ product, partly to allay public fears.

Mar. 22—AZ reports the long-awaited results from the US, Peru and Chile, in 32,000 participants. The top-line efficacy of 79 percent looks solid, if not spectacular. Some public health experts, like Ashish Jha at Brown University, cheer this development as a step toward vaccinating the world.  

For sure, 79 percent is solid efficacy. The more vaccines we have, the better. But before getting too excited, I want to see some more details on how the vaccine is performing against the variants and in subpopulations and what the immunogenicity data look like.

Part of me really prefers to withhold judgment for now, wondering if there’s another shoe to drop. Given the track record, it makes sense to wait and see for what the FDA staff come up with when they comb through the dataset with a kind of rigor that makes peer-review look like a stroll in the park.

Not only is the FDA rigorous, it has a long institutional memory. It can really put the screws to companies behind the scenes in multiple ways.

That’s one of the important lessons I learned many years ago in an FDA law class at Harvard Law School taught by Peter Barton Hutt, the attorney at Covington and a legendary former FDA counsel. (I’m not a Harvard Law graduate, but was able to audit classes at MIT and Harvard during the 2005-2006 academic year via the Knight Science Journalism Fellowship at MIT).

I practically heard shades of Peter Barton Hutt and FDA staff sharpening their pencils this morning when I watched a TV appearance by Anthony Fauci.

The FDA is going to very, very carefully go over all of these data,” Fauci said, “You can rest assured, that the FDA will apply a great deal of scrutiny in every aspect of these data.”

Fauci is surely aware that while the US public may have forgotten some of AZ’s missteps, the FDA has not.

Even if the company turns in a thorough and squeaky-clean application and regulators agree that it deserves an Emergency Use Authorization (the most likely outcome), that will be just one step in building back public trust.

Yesterday morning, on cue with the company press release, the NYT published an op-ed from Heidi Larson, the preeminent voice on vaccine hesitancy.  

There’s a lot of work to do in building vaccine trust, she wrote.

Indeed.

[Update 6:45 am PT, Mar. 23: After this column was published, the National Institute of Allergy and Infectious Disease issued the following statement: “AstraZeneca may have included outdated information from that trial, which may have provided an incomplete view of the efficacy data. We urge the company to work with the DSMB to review the efficacy data and ensure the most accurate, up-to-date efficacy data be made public as quickly as possible.” AstraZeneca responded with its own statement, saying it would provide more updated data within 48 hours.

Financings

Taicang, China and San Diego-based Connect Biopharma raised $191 million in an IPO at $17 a share. The company is working on T-cell driven inflammatory diseases. Qiming Venture Partners, RA Capital Management, and Advantech Capital were among the principal shareholders heading into the liquidity event. Shares inched up to $18.61 at yesterday’s close.

Dallas, Texas-based Instil Bio, the developer of T-infiltrating lymphocyte therapies for cancer, raised $320 million in an IPO at $20 a share. It climbed to $26.80 at yesterday’s close, with a market valuation of $3.3 billion. CEO Bronson Crouch controls the Curative Ventures entity that holds 29.7 percent ownership in the company after the IPO. The other big stakeholders in the company include Venrock, CPMG and Vivo Capital.

Somerville, Mass.-based Finch Therapeutics, a microbiome therapeutics developer, raised $128 million in an IPO at $17 a share. A member of the Walton family, heirs to the Walmart fortune, is among the big winners in this IPO. Shares traded up to $19.15 at yesterday’s close.

Boston Immune Technologies and Therapeutics (BITT) said it completed a $10 million Series A/A1 financing to develop novel antibodies against members of the TNF superfamily. BeiGene participated, along with Hatteras Venture Partners and EGP Investments.

Malvern, Penn.-based Xylocor Therapeutics said it completed a $41.9 million Series A round to advance its gene therapy for coronary artery disease. Fountain Healthcare Partners led and was joined by new investors Longwood Fund and Lumira Ventures.

Seattle-based Dexcare, a new digital health startup, said it raised $20 million in investment led by Define Ventures. The round included Frist Cressey Ventures, Kaiser Permanente Ventures, SpringRock Ventures and Providence Ventures. DexCare describes itself in a statement as “an intelligent digital care operating system that manages health system capacity and demand across all lines of care.” It was developed internally at Providence, a large hospital chain on the West Coast, in 2016 and is now being spun out aas a business with a half-dozen customers.

Cambridge, Mass.-based Aura Biosciences said it raised $80 million in financing to advance virus-like drug conjugate therapies for cancer. Matrix Capital Management and Surveyor Capital led.

Legal Corner

The SEC brought charges against uBiome for allegedly defrauding investors out of $60 million. The complaint was brought in the US District Court in Northern California.

Science Policy
  • Put this one in the ‘Never Forget’ Folder: Tom Frieden on the ‘Mind-Boggling’ Interference of the Trump Administration in COVID-19. The BMJ. Mar. 17. (Joanne Silberner)
  • Where’s the Science Behind CDC’s 6-Foot Decree and Transmissability? WSJ. Mar. 21. (Scott Gottlieb)
  • The Pandemic Year: The Hyper-Acceleration of the Life Sciences. WSJ. Mar. 19. (Eric Topol)
  • Five Myths About Coronavirus Vaccines. Washington Post. Mar. 19. (Peter Hotez and Maria Elena Bottazzi)
Science of SARS-CoV-2
  • Post-acute COVID-19 syndrome. Nature Medicine. Mar. 22. (Ani Nalbandian et al New York-Presbyterian/Columbia University Irving Medical Center)
  • Neutralizing Antibodies Against SARS-CoV-2 Variants After Infection and Vaccination. JAMA. Mar. 19 (Venkata Viswanadh Edara et al Emory University).
  • Infection and vaccine-induced antibody binding and neutralization of the B.1.351 SARS-CoV-2 variant. Cell. Mar. 20. (Venkata Viswanadh Edara et al Emory University).
  • SARS-CoV-2 variants B.1.351 and P.1 escape from neutralizing antibodies. Cell. Mar. 20. (Markus Hoffmann et al University Gottingen)
  • Difference in SARS-CoV-2 attack rate between children and adults may reflect bias. Oxford Clinical Infectious Diseases preprint. Feb. 26. (Zoe Hyde et al University of Western Australia)
  • Transmissibility and transmission of respiratory viruses. Nature Reviews Microbiology. Mar. 22. (Nancy Leung)
  • Single-component, self-assembling, protein nanoparticles presenting the receptor binding domain and stabilized spike as SARS-CoV-2 vaccine candidates. Science Advances. Mar. 19. (Linling He et al The Scripps Research Institute)
Data That Mattered

Roche/Genentech said its PD-L1 directed antibody atezolizumab (Tecentriq), hit the primary endpoint of extending disease-free survival in the Phase III Impower010 study. The trial looked at patients getting adjuvant therapy after surgery and chemotherapy for Stage II-IIIA populations with non-small cell lung cancer. Participants were randomized to the drug group, or best supportive care. The company said the magnitude of disease-free survival benefit “was particularly pronounced in the PD-L1-positive population.” The company said it doesn’t yet have mature data on Overall Survival.

Roche/Genentech also said it shut down a Phase III clinical trial for tominersen, an antisense oligonucleotide for Huntington’s disease in-licensed from Ionis Pharmaceuticals. The drug was designed to reduce production of huntingtin protein (HTT), including its mutated variant, mHTT. An independent data monitoring committee made the recommendation. See Sek Kathiresan’s succinct summary of this head-scratcher below. (Phase I results in NEJM, 2019).

Exton, Penn.-based Idera Pharmaceuticals failed in a pivotal trial of tilsotolimod in combination with ipilimumab versus ipilimumab alone in patients with anti-PD-1 refractory advanced melanoma. The drug is a Toll-like receptor 9 agonist, being combined with the CTLA-4 inhibitor in this case. Shares lost two-thirds of their value.

Our Broken US Healthcare System

Major donors to South Florida hospital foundation got early vaccine access. Politico. Mar. 19. (Arek Sarkissian and Matt Dixon)

RIP

Jose Baselga, the prominent cancer researcher and leader of oncology R&D at AstraZeneca, died at age 61. (STAT obituary).

Personnel File

Diana Brainard was hired as CEO at Cambridge, Mass.-based AlloVir, a cell therapy company focused on viral diseases patients with weakened immune systems. She starts May 17. Brainard is currently senior vice president of virology therapeutics at Gilead Sciences. She oversaw teams that developed curative therapies for hepatitis C and remdesivir for COVID-19, among other areas. She replaces David Hallal, who is moving upstairs to be executive chairman. Hallal is also CEO of ElevateBio, the largest shareholder in AlloVir. (Disclosure: Brainard is married to TR healthtech columnist David Shaywitz.)

Jean-Frédéric Viret was hired as chief financial officer at South San Francisco-based Blade Therapeutics, a developer of treatments for fibrotic diseases. He was previously CFO at Coherus Biosciences.

South San Francisco-based Sutro Biopharma, the developer of treatments for cancer and autoimmunity, promoted David Pauling to General Counsel and Robert Kiss was promoted to senior vice president of process and analytical development.

South San Francisco-based Veracyte, a molecular diagnostics company, added Muna Bhanji to its board of directors.

Chart of the Day

The data out of Brazil are a concern.

Source: Outbreak.info, based on data from Johns Hopkins University Center for Systems Science and Engineering, New York Times, COVID Tracking Project, GISAID Initiative

20
Mar
2021

Enticing Some With Social Cues, Others With Health, Exercise Rewards Body And Mind

David Shaywitz

I recently discussed the rise of digital fitness, and specifically how companies like Peloton are succeeding by delivering an engaging experience.

The new crop of digital fitness companies have figured out how to make health-promoting activities that are intrinsically tedious – like riding a stationary bike – into something compelling and sustaining. A New York Times writer, Amanda Hess, captures the essence of this magnificently in her piece, “Your Brain on Peloton.”

I was reminded of another important component of healthy activities this week when I was chatting with my barber (as one does), and asked about his fitness routine. He told me he goes to his gym 3-4 times a week, and has for years. 

I wondered what gets him there each day. Simple, he said. He has a group of buddies there.  Sometimes, he says, they’ll go for drinks afterwards, or their families will go over to one another’s house afterward for dinner. 

In short: he’s motivated by a sense of community.

An anecdote is hardly data, but the idea that healthy behaviors may be linked to interpersonal influences is well established. 

An influential paper published by Nicholas Christakis and James Fowler in the New England Journal of Medicine in 2007, for example, examined decades of Framingham Heart Study data, and concluded that “obesity may spread in social networks.” They note “people are embedded in social networks,” which “suggests that both bad and good behaviors might spread over a range of social ties.”

Another Christakis paper, from 2016, looked at two years of Gallup surveys for clues about the influence of social networks. Analyzing the data, Christakis and colleagues concluded that individuals who tend to associate with a lot of heavy people are more likely to want to lose weight, but are less likely to be successful. Conversely, associating with thinner people is linked to more successful weight loss.   

“Gains and losses of even a single social tie with a thinner or heavier individual show important links to the probability of obesity,” the authors of the 2016 paper wrote. In other words: the probability of obesity increases with additional ties to heavier people and decreases with additional ties to thinner people, and vice-versa.

A very readable overview of the “power of community” in cultivating consistent healthy fitness behaviors, by industry analyst Anthony Vennare, can be found here.

My barber’s story also reminds us of Clay Christensen’s advice: make sure you understand the problem to be solved. In Christensen’s classic (and perhaps not so healthy) milkshake example, many customers purchasing a shake from a fast-food drive-through turned out not to be looking for a tasty beverage so much as for something to occupy them in the morning, while they were driving to work. 

By recognizing this initially obscure need, the vendor was able to increase sales by making the shakes thicker (to last longer) and adding bits of fruit (to make it more interesting).

Which brings me back to what the real issue is for the barber at the gym. He’s apparently not looking for the most efficient or effective workout. Instead, he’s looking for camaraderie, while engaged in a healthy activity. These social factors are often what draw customers to the gym, and keeps them coming back.  While these individuals may achieve a healthy outcome, their pursuit of physical health is only one motivator, and often not the primary one.

Of course, not everyone is motivated in the same way. Many people are drawn to exercise explicitly for the physical benefits (such as better endurance, an improved cardiovascular risk profile, a beach body, or wanting to keep up with the grandkids). Some then discover significant mental health dividends along the way.

As modern life becomes increasingly busy, with ever more demands on our attention, “exercise time” may represent our last protected space, a cocoon of time we give to ourselves.

Over the last several years (as I’ve discussed here and here), and continuing through the pandemic, I have savored my morning exercise routine. I’ve used that time — whether sweating inside on the treadmill, elliptical, or weight machine, or outside on my bike — to lose myself in audiobooks and podcasts. I avoid work during this early hour, never checking email or text messages, and avoid all but the most essential calls. I love this daily routine. I find it grounds me, and puts me in a relaxed and positive mindset – an ideal headspace – to start the day. 

I’m certainly not alone.

I also recognize that many people — including me — would have a really tough time taking an hour out of each day for reflection, meditation, or other self-soothing activities. For many similar Type A’s, it would feel difficult to justify, and easy to encroach upon. 

Yet because the time is allocated for an activity that’s both nominally health promoting and physically unpleasant (at least compared to sitting on the couch), it somehow seems easier to rationalize – a modestly uncomfortable sacrifice made in the name of disease prevention. 

In short, exercise creates the permission structure to give ourselves the headspace we so desperately need.

In short, exercise creates the permission structure to give ourselves the headspace we so desperately need.

This also makes me wonder how much of the benefits attributed to the physical aspects of exercise may actually come from the state of mind that exercise enables us to inhabit. Parsing the relative contribution of each seems difficult. 

The bottom line is less ambiguous. Whether you go to the gym to socialize and wind up exercising (delighting your cardiologist), or jump on the bike to stay fit and wind up rejuvenated because of your protected “me-time,” (pleasing your psychiatrist), the results are joyously similar: a healthier body, a happier mind, and a better you.

16
Mar
2021

Why Digital Fitness Companies Like Peloton and Tonal Are Exciting For Healthcare

David Shaywitz

A truism in healthcare is that a medicine only works if it’s taken. Unfortunately, many people don’t take the medicines they are prescribed.

Adherence rates for many drugs – especially for preventive medicines like statins – tends to be remarkably low, as I’ve discussed in the New York Times

About half of patients who start taking statins to reduce their risk of cardiovascular disease quit taking them in the first year, according to a 2019 paper. The authors note that adherence goes downhill from there.  

People find it even harder to stick with lasting healthy behaviors. Adherence to diet and exercise regimens is notoriously poor – just losing weight, for example, is hard enough, and keeping it off is profoundly more difficult; 95% of people who lose a significant amount of weight regain it relatively quickly.  (This is a fate I’ve thus far avoided on the Virta low-carb regimen, which I started in 2018 and lost 80 pounds, as I discussed here; I’ve kept the weight  off, as I wrote in TR, here, and I remain a self-pay, non-diabetic subscriber). 

It’s common for those who make New Year’s fitness resolutions to lose remaining resolve around now; after all – food is delicious and exercise is hard.

And yet, there’s hope: more than 90% of Peloton users are apparently still using the bikes a year later. This was true before the pandemic, and it’s still true. 

Moreover, as CNBC reported last month:

“the cycle maker continues to look for ways to entice its customers to exercise more…It recently launched a feature where users can ‘stack’ classes back to back, and have them play automatically one after another. It also recently added Pilates classes to its catalog.”

Physicians may be tempted to contrast this success rate with their own experience advising patients around diet and exercise. The comparison, of course, isn’t quite fair, given the self-selection of Peloton purchasers, whose level of motivation — and disposable income — are likely not representative of the population as a whole.

Yet doctors might see a pattern here. Recall the remarkable success of the lifestyle modification arm of the legendary Diabetes Prevention Program (DPP) study. It demonstrated that in vulnerable patients, impending diabetes could be significantly forestalled. The approach used in the trial – documented here – included individualized programs for each participant, tailored and delivered by “life-style coaches” who maintained intensive contact with participants. This program, as I’ve discussed, is also the basis of digital health companies like Omada.

A similar degree of engagement may be required to realize the benefit of exercise in depression, as highlighted on a 2019 episode of Wendy Zukerman’s wonderful “Science Vs” podcast.  Zukerman points to several relatively recent clinical trials that seem to show a benefit of exercise in patients with depression, in contrast to previous studies. 

Zukerman perceptively observes:

“One reason that these newer studies might be finding a benefit is that these scientists were really tenacious about getting people to do the exercise. Like in one study researchers gave participants free gym memberships and personal trainers – who hounded them if they don’t show up.” 

The genius of Peloton, it would seem, is in motivating participants to remain committed to the program, which they assiduously cultivate through a range of efforts intended to make the cycling both engaging and social.

Other digital fitness companies such as Tonal (strength training) and Hydrow (rowing) are trying to follow suit, as are traditional exercise hardware manufacturers like NordicTrack, now a brand of ICON fitness, who have launched iFit to participate in the connected trend (though based on some reviews – here, also here — it seems like the software has a way to go).

Apple’s Fitness+ app has also arrived on the scene; it’s “Time To Walk” feature – featuring celebrities like Dolly Parton and Draymond Green telling stories while they walk – seems both well thought-out and expandable to other formats.   

As Zukerman reminds us, while you may not be able to exercise your way to weight loss (which is driven largely by what you eat), exercise contributes to your health in many other ways.

Accessing this benefit will require companies to master a range of competencies, including hardware, software, and media, given the importance of an engaging and immersive experience in motivating and sustaining participation. 

Successful connected fitness platforms will improve the health and well-being of their members — including, by the way, a number of academic physicians and scientists, many biopharma researchers and executives, and even some regulators. 

In learning how to sustain engagement with exercise, these digital solutions may point the way to helping us pursue other difficult but healthy activities, like diet modification or rehabilitation after injury. 

Finally, the volume and specificity of data around exercise (which leaps out from this podcast with Tonal founder Aly Orady) would seem to provide powerful collaboration opportunities for a wide range of motivated physicians and medical researchers – a real chance to explore and advance the state of the science. 

In short, the new generation of connected fitness companies offer not only the immediate promise of improved health, but also the intriguing possibility of future insights into both the physiology of health and the psychology of adherence. 

Ride on!   

15
Mar
2021

Software for the Cell & Gene Therapy Wave: Vineti’s Amy DuRoss on The Long Run

Today’s guest on The Long Run is Amy DuRoss.

Amy is the co-founder and CEO of San Francisco-based Vineti. The company provides software to manage the delicate logistical dance for cell and gene therapies.

Amy DuRoss, co-founder and CEO, Vineti

Vineti has raised about $115 million in three venture rounds of financing. Its backers include Cardinal Health, the big medical distributor, as well as traditional venture firms like Canaan Partners and Section32, plus the big cell therapy players Novartis and Gilead Sciences.

All those people are apparently converging on a challenge of the fast-growing world of cell and gene therapy. With more than 1000 Investigational New Drug Applications on file at the FDA, does it make sense to have 1000 different bespoke software and logistical solutions for cell therapies, or does it make sense to have a standard operating system of sorts to make life easier for investigators, for companies, for regulators, and for payers?

Vineti represents a wager on this field moving to standardized software, and a desire to be the company that sets and delivers on that standard.

This has taken a while to sink in with the biotech industry. Amy described the challenge to me a year ago in this Timmerman Report article, and provided an update in this recent conversation.

Before we dive in to the interview, a word from the sponsor of The Long Run.

Synthego is a genome engineering company that enables the acceleration of life science research and development in the pursuit of improved human health.

The company leverages gene editing, machine learning, and automation to build platforms for science at scale. By enabling unprecedented access to cutting-edge genome engineering, Synthego is at the forefront of innovation, accelerating the development of truly engineered biology.

Synthego’s expertise in CRISPR, combined with their proprietary software and technology, means you have a trusted partner whether you are at early stages of basic research or ready to take your therapy to the clinic.

Visit Synthego.com/timmerman to learn more.

Now, please join me and Amy DuRoss on The Long Run.

14
Mar
2021

One Vaccine Dose or Two? We Need Our Best Defenses Against the Variants

Larry Corey, MD

When there aren’t enough doses of vaccine to go around against the SARS-CoV-2 virus, and you’re trying to protect as many people as possible as fast as possible, what’s the right thing to do?

Does it make sense to take the Pfizer and Moderna vaccines and give them as single shots, in order to stretch out our existing supplies and vaccinate twice as many people right away? Or would we be better off sticking with the two-dose regimen that’s been shown to stimulate the immunity needed to keep people out of the hospital and alive?

The “one-shot or two-shot” question is a legitimate subject of debate among scientists, including leading epidemiologists. The media are giving it prominent air time.

My view, based on the underlying immunology and the clinical trial results, is that it’s better to stick with the proven two-dose regimen. Allow me to elaborate.

Proponents of the one-shot strategy acknowledge that a second dose should be given, but they don’t think that the timing of that second dose is as essential as one-dose partial protection in an urgent moment. Some say it’s OK to have a lag time between the first dose and the second, and that the second dose could be given two or three months later. The vaccines were tested in two-dose regimens given three or four weeks apart. By stretching out the time between doses, they contend, we’ll get more of the population partially protected, while buying a little time for the manufacturers to ramp up all the supplies needed for the second round of dosing.

When numerous esteemed people are saying the same thing, a reasonable observer should listen and consider the argument. These people have good faith reasons for taking that position.

But let’s pause and unpack these ideas a bit.

There are substantial data demonstrating partial efficacy after one dose of the Pfizer and Moderna vaccines. Data from the initial Pfizer trial showed that between day 12 and day 21—between the first and second dose—people in the vaccine group were less likely to get COVID-19 infection than their counterparts in the placebo group. The Pfizer vaccine’s efficacy as a single shot was 52% (39 participants in the vaccine group acquired disease compared with 82 infections in the placebo group).

Data from Israel, which had one of the most efficient vaccine rollouts of any country, confirms that a single shot of the Pfizer vaccine delivers about 50 percent effectiveness in the real world with all its messiness. The data in the Moderna trial were essentially the same. From randomization to dose two, there were 7 cases of infection in the vaccine group and 46 COVID-19 cases in the placebo group.

So, on purely clinical grounds, an epidemiologist can argue that more good could be derived for the whole population by vaccinating two persons with a 50% effective vaccine compared with one person taking a 95% effective vaccine.

This poses an interesting decision to make on a personal level with respect to informed consent. What would you rather have as an individual – 95 percent efficacy in two doses three or four weeks apart, or 50 percent efficacy while waiting another 2-3 months for the next dose?

I don’t think the real answer lies in a debate about the ethics of reduced efficacy from single-dose immunization or the efficiency of public health versus individual rights. The important question is: Would we actually be doing more harm than good by administering one dose of the Pfizer or Moderna vaccine to more people?

Why do I raise this question?

I say this as a virologist, knowing that the viruses we’re encountering today and the ones we will encounter in the next several months are not the same viruses that we tested the vaccines against and upon which the single-dose data are based on.

The SARS-CoV-2 virus in wide circulation today, with its myriad variants, is a more formidable adversary. The variants that are emerging and taking over have higher infectivity and have evolved new capabilities to escape from neutralizing antibodies. We have learned that the seeds of that escape are found among people with low/partial immune responses from natural infection. That means the virus, when it gets inside a host with only partial immunity, has a greater opportunity linger inside the body and keep replicating until it develops certain evolutionary advantages.

In this context, while the body is mounting a weak immune response and the virus is continuing to replicate, we’re likely to see a prolonged period of viral shedding from the nose and mouth. That raises the likelihood of sustained community transmission. Essentially, the viruses are stressed but not eliminated by partial immunity.

In such a situation, it makes sense to make the virus face off with our most formidable immune defenses. We force it to go up against people who have been fully immunized with the two-dose vaccine regimen.

There are data to support this view. When we look at the protective antibody levels at day 21 after the first dose of the Pfizer vaccine, we see that the 50% serum neutralization titer is 29. When people get the second dose, and their blood is evaluated for antibodies just 7 days later, neutralizing antibody titers shoot up to 270. After another 7 days – a full two weeks after the second vaccine dose – neutralizing antibody titers are all the way up to 437!

The data with Moderna are similar: after one dose, the mean titer of neutralization is only 18; 7 days after dose two, it’s 256; and 7 days after that, it’s 344.

For people unfamiliar with neutralizing antibody titers, a level of 29 is equal to a low-dose convalescent plasma treatment. We know at this point that low-dose convalescent plasma is associated with more harm than good — it is associated with being a precursor for escape variants to emerge among immunosuppressed persons.

It’s also important to recall that the data we have on efficacy are derived from the clinical trials that were designed to match the vaccine strain with the original—or what some call “wild type” (Washington strain)—virus. The new variants change the game somewhat, and require us to take another look at their impact on vaccine efficacy.

Data from the lab, looking at live virus versus neutralizing antibodies in the petri dish, shows two-or three-fold more antibodies are required to neutralize the B.117 variant now spreading throughout the UK and Europe. Lab results further show 8 to 10-fold more neutralizing antibodies are required for the B.1.351 variant emerging from South Africa.

This resistance to neutralization is associated with the reduction in efficacy observed in the latest clinical trials in geographies where those variants circulating. The reduced efficacy was seen in both the Novavax vaccine (93% efficacy in the UK and 43% in South Africa) and Johnson and Johnson vaccine (73% efficacy in the US and 58% in South Africa).  

This necessity for higher amounts of antibodies against the new variants is why I am concerned about having large numbers of people walking around for an extended time period with less-than-optimal levels of immunity. A 2-3 fold reduction in a neutralizing titer of 20 after one dose of mRNA equates to a titer of 5. That’s essentially no detectable neutralizing activity. An 8-fold drop results in neutralizing titers that “fall off the grid”. This is not the case with the second dose when a two-fold reduction from 450 is still a neutralizing titer of 225 and an 8-fold reduction is 60 — still three times the neutralizing levels above that seen with one dose of mRNA vaccine. 

The argument for the two-dose regimen is even stronger when you look at vulnerable subpopulations. Many elderly people have weak immune systems. One dose of the mRNA vaccine often elicits antibody titers of less than 20. The elderly invariably require two doses to get a detectable neutralizing response.

A similar situation is also seen among persons with a wide variety of chronic diseases; their less-than-robust immune response will have an even greater impact on levels of protection after one dose. Data on the origin of escape variants points to immune-suppressed patients and persons with untreated HIV as reservoirs for the selection of escape mutations, especially with the E484K mutation harbored within the B.1.351 variant.

My compatriots say if one dose works for the Johnson & Johnson vaccine, why not do the same with Pfizer and Moderna? There is a difference: there is a much greater CD8+ killer T cell immune response in the J&J vaccine at one dose than with the mRNA vaccines after one dose. It’s really a different mechanism of protection with the adenoviral vector-based J&J vaccine.

While my musings are inferential, so are the views espoused by those advocating for the one-shot strategy. This is the nature of pandemic response. We are all looking at the same data, asking pertinent questions, and doing our best to navigate the uncertainty in the moment.

It behooves us to solve the challenge of vaccinating our population by first immunizing with the regimens that we know have demonstrated extremely high levels efficacy. Even with our absolute best immune defense, the variants will pose a challenge. I believe we will do more harm than good by markedly increasing the population of people with partial protective immunity.

Weakening our best defense may be well-intended, but it could come back to haunt us. Depending on the past is not how an RNA virus behaves. It uses the past to change its future in order to survive. Good epidemiology does not necessarily mean good virology.

We need all the strength we can muster in the vaccine regimens. For the two mRNA vaccines, I want my friends and family to be immunized with the full two-dose schedules authorized by regulators at the FDA and the European Medicines Agency.

Dr. Larry Corey is the leader of the COVID-19 Prevention Network (CoVPN ) Operations Center, which was formed by the National Institute of Allergy and Infectious Diseases at the U.S. National Institutes of Health to respond to the global pandemic, and the Chair of the ACTIV COVID-19 Vaccine Clinical Trials Working Group. He was intimately involved in the planning of the phase 3 vaccine studies conducted under the funding auspices of Operation Warp Speed. He is past President and Director and Professor in the Vaccine and Infectious Disease Division of Fred Hutchinson Cancer Research Center; and Professor of Medicine and Virology at University of Washington.

10
Mar
2021

Once Vaccinated, What To Do With Masks?

Larry Corey, MD

Contributing editor: Chris Beyrer, MD

Once I’m fully vaccinated, should I still wear a mask?

This is probably the biggest public health policy question facing us today. It’s an issue each one of us will have to ask ourselves as the U.S. mass vaccination campaign continues to roll out, especially when many people around us aren’t yet fully vaccinated.  

No one likes masks, including me. But I understand how many gazillion viral particles can be put on the head of a pin and how many gazillion viral particles can be emitted through a sneeze, or while talking, or eating. I know how easy it is for other people around us to inhale those viral particles, and then come down with a serious case of COVID-19. And I understand how evolution has made this the mode of transmission for hundreds and hundreds of viruses—far too numerous to list here—little critters that were here on the planet well before us.

The good news in the fight against COVID-19 is that we now have three very effective vaccines—two mRNA (Pfizer and Moderna), and one Ad26 (Johnson & Johnson)—approved in the U.S. under Emergency Use Authorization. These vaccines provide personal protection against symptomatic COVID-19. That’s great news, now that more than 30 million Americans have been fully immunized, and perhaps 100 million Americans have some measure of natural immunity from prior bouts with SARS-CoV-2 viral infection.

But what about those unvaccinated persons around you? Are they protected? Can you still acquire the virus unknowingly and transmit it? The CDC put out new guidance on social distancing for fully immunized people on Mar. 8. The headlines focused on fully vaccinated people being able to gather indoors with small groups of unvaccinated friends and family, unmasked.

Essentially, it was a green light to go visit the grandkids and hug them.

We need to think about this new guidance and what we know and don’t know about COVID-19 transmission.

First, let’s look at what happens when you come into contact with the virus and how protection works in your body.

When the virus lands on your hands and you touch your eyes or your nose or your mouth, it really becomes a race between your immune response and the virus’s ability to invade cells and start replicating.

The winner of that race is determined by a number of factors. There’s the infectivity of the virus itself, the inoculation load, the strain of the virus, and your own innate and adaptive immune response.

How good are the first “soldiers,” the innate immune system cells often found in the mucosal lining of the nose, and just under the skin? These cells are often not primed by vaccination. Then there’s the question of how quick are the second soldiers, the B cells that produce specific antibodies, and the killer T cells that have memory for certain pathogens. Do these second-line defenders get into the nose? If so, how many get into the nose?

There’s a fair amount of work that’s been done on what we call the battlefield between the host and the virus. And for many fast-replicating viruses, the half-life of a viral-infected cell is 20 to 40 minutes. So, every 20-minute delay in an immune response can lead to a doubling, sometimes a tripling of the virus. This interplay tends to happen in what I’ll call staccato time; it certainly is not adagio (slow tempo).

Once you’re vaccinated, we know from well-controlled clinical trials that you are protected from getting severely ill or dying. What we don’t yet know is: can the virus still infect you and replicate at a high enough titer that you could unwittingly transmit it to someone else?

That’s the question we’re asking today. If the person is vaccinated, this may not be a big deal. But if they’re not vaccinated and they have an underlying medical condition, and they breathe in SARS-CoV-2 viral particles, then we could have a problem. A problem that leads to hospitalization or worse, death.

The potential severity of COVID-19 continues to stagger the imagination. It’s not like the flu, despite some public assertions from a year ago.

This graph illustrates the point:

It’s this knowledge that I carry with me from day to day.  

I try to remind myself, “Larry, you’re vaccinated, but a lot of people aren’t. In fact, 90% of the people walking around are not vaccinated and you do not have the luxury to give up your responsibility to protect them.”

This brings me back to the issue of wearing masks. Once you’re vaccinated, it’s really not about you at all. It’s about the other person. Maybe a friend or family member or neighbor or colleague that attends the same event or gathering but has not had the opportunity to be vaccinated. Or even someone who has chosen not to be vaccinated: do they deserve to get ill?

One might ask, a year into this pandemic, why don’t we know whether being vaccinated will prevent transmission? We don’t know this because the kinds of studies required to look at transmission are different from the ones designed to test vaccine efficacy. The clinical trials testing vaccine efficacy showed personal benefit—in other words, if you get vaccinated, will you be protected? And the answer to that as we’ve seen is Yes.

But looking at transmission is a different matter altogether, and requires an entirely different study design, a different set of volunteers, and all the time and money required to get a rigorous answer we can count on.

In the phase 3 trials we conducted through Operation Warp Speed, there were thousands and thousands (approximately 30,000 participants) who were tested regularly (approximately once a week) to see if they got symptomatic COVID-19. But to test whether the virus was colonizing in the nose — without visibly detectable symptoms — would have required near-daily testing. And if you pause to consider that—30,000 persons over five months—with each person coming in daily to get their noses swabbed, it would have become impossible to test all the cultures in a timely way. We would have been diverted from defining vaccine efficacy.

It is possible to take fewer people and do what we call a transmission study, which is currently being conducted with the mRNA vaccines. We will look at vaccinated versus unvaccinated persons and define if vaccination prevents you from transmitting the virus.

This will require intensive contact tracing—looking at the contacts, who got infected, who didn’t? Who came first; who came second? Can we define exactly whether this person got it from that person? Sometimes the genetics of the virus allows one to do that.

The emerging variants also complicate things because they have been shown to be more infectious. For example, the B.1.1.7 strain first seen in the UK has been shown to cause 30% more infections. It appears to be shed for a substantially longer period of time than the G614D strain that has been circulating in the US for the last 10 months. That means people have more opportunities to spread it to others when they are doing ordinary things out and about in the community.

As the virus adapts, these new strains will cause a stress on vaccinations. But I think we can almost be happy that we’re doing the transmission study during the period of time when the virus is changing because we really are getting to the relevant issue of how well these vaccines perform when the virus is starting to mutate.

We are getting to answers, but it will take some time. With a little luck, the transmission study will show the mRNA vaccines are spectacular at preventing people from getting sick from COVID-19, AND preventing people from transmitting the SARS-CoV-2 to others. Or if you do acquire the virus from a vaccinated person, perhaps we’ll see the viral loads you take in are trivial, easier for your immune system to handle, making it far less likely that you would transmit it to anyone else.

I’m hoping that’s the result because I want to take off my mask. But until we know this with reasonable certainty, or at least until more of our population is vaccinated, then public policy—and individual conscience—should mandate mask use in public spaces and no more than small gatherings in our homes. The CDC guidance of Mar. 8 is clear that fully immunized people can meet each other at home, in small groups, without mask wearing and social distancing, but we’re going to have to maintain vigilance in public spaces, at work, on public transport and at schools as they reopen. Because mathematical modeling shows that without adhering to these measures, we could double the deaths.

How each one of us behaves makes a difference. Together, we can markedly influence the surges associated with this virus and potentially save lives.

 

Dr. Larry Corey is the leader of the COVID-19 Prevention Network (CoVPN ) Operations Center, which was formed by the National Institute of Allergy and Infectious Diseases at the U.S. National Institutes of Health to respond to the global pandemic, and the Chair of the ACTIV COVID-19 Vaccine Clinical Trials Working Group. He was intimately involved in the planning of the phase 3 vaccine studies conducted under the funding auspices of Operation Warp Speed. He is past President and Director and Professor in the Vaccine and Infectious Disease Division of Fred Hutchinson Cancer Research Center; and Professor of Medicine and Virology at University of Washington.

Chris Beyrer, MD, MPH is the Desmond Tutu Professor in Public Health and Human Rights at the Johns Hopkins Bloomberg School of Public Health. A professor of Epidemiology, Nursing, and Medicine, he now serves as Senior Scientific Liaison to the COVID-19 Vaccine Prevention Network, and as co-editor of this blog series.

9
Mar
2021

The Rise in Asian American Hate Crimes: A Biopharma Perspective and Call For Action

Kevin Kwok, Pharm.D, Parkinson’s Patient Activist & Biopharma Veteran

I have been a biopharma executive for 30 years. For the last 12 years, I’ve been living with Young-Onset Parkinson’s Disease.

I have been an Asian American even longer…all my life.

The biopharma industry has been very good to me, offering constant growth and opportunity for advancement and reinvention.

My most recent job was in patient engagement. It suited me well, given my journey with a neurodegenerative disease. This job required reaching out and actively listening to patients.

Too often, their stories were untold or fell on insensitive ears.

Our industry is moving toward being more patient-centric in both words and deeds. It’s an encouraging trend, as we learn a lot by actually talking with, and listening to, patients. Patients have long had to silently soldier on with their ailments, as the medical community often doesn’t listen well, barreling ahead with treatment plans based on preconceived biases of thinking it knows what patients need.

Listening to people, and giving voice to the voiceless, has been one of the most rewarding things I’ve done in biopharma.

I have learned from actively listening to patients. Silence doesn’t mean everything is OK.

This brings me back to the beginning. Growing up Asian American, I can relate on a certain level. Silence doesn’t mean everything is OK here either. We were raised by the generation before us to work hard, put your nose to the grindstone, and to not rock the boat. We are given lessons from childhood to “Get a good education, get a technical job; work hard… this is the American Way to success.”

Many Asian Americans have followed these lessons and found success in this country. But that doesn’t mean everything is OK. It creates the Asian American model minority myth. These same behaviors keep too many people silent, and blind too many others to the suffering in our midst.

You may have heard recent news stories of a surge in hateful actions, and sometimes violent attacks, on Asian Americans. Reports of hate crimes directed at us have reached an all-time high. It’s the direct result of political leaders who have recklessly politicized COVID-19 as “the Chinese virus” or “Kung Flu,” with no apparent concern for how followers might act on those hostile words.

These are not one-off incidences. The video of a frail, elderly Asian American man, Vichar Ratanapakdee in San Francisco being violently and fatally pushed to the ground was horrifying.

In New York, a middle-aged man riding the subway had “smile” viciously carved from ear to ear by a deranged slasher.

In Maryland, a young Chinese American man riding his bike on a neighborhood bike path, while on a date, was chased and pelted with rocks by kids screaming racial epithets.

There have been countless reports of health care providers being verbally assaulted and spit on, told to “go back to your country and bring your Chinese virus with you.”

Ironically, many of these same Asian American frontline nurses and physicians were on their way to the ICU to battle for the lives of COVID-19 patients.

Many of these victims of racial abuse in the healthcare system are second and third generation Asian Americans. Over the past four years, I have personally experienced racial intolerance, which I haven’t experienced since early childhood.

I had naively thought that overt racism was from a bygone era, and that we now lived in a racially tolerant world. I worked and raised my bi-racial children in San Francisco, the melting pot of all melting pots. My former SF home was only blocks away from Mr. Ratanapakdee’s home in the Richmond District.

The rise in Asian hate crimes, especially over the past year, has changed my perception of our country as a place making steady progress as a multi-ethnic, multi-racial melting pot. So did the horrible murder of George Floyd.

These are scars on American society. What can biopharma do to help the country heal?

There are a few things within our control. Biopharma has plenty of room to improve diversity hiring. It’s true that biopharma already does hire and employ a high percentage of Asian Americans and Asian Nationals. Many continue their daily work without complaint, and without rocking the boat. But silence shouldn’t be interpreted as thinking everything is OK.

Last week, I was invited to provide patient input to an Asian based startup. This company is eager to incorporate patient perspectives in their drug development and commercial strategy for its Parkinson’s drug candidate.

In our Zoom call, the killing of Vichar Ratanapakdee, the 84-year-old man in San Francisco, came up. The pain from employees was evident, coming right through my screen.

Afterwards, the co-founder of this company, based in Asia, told me that Mr. Ratanapakdee’s death was on the front page of every Thai newspaper, and media channels were stating that the US was not a safe place for Asians to work.

The Asian American community has been alarmed and awakened by this escalation in hate crimes. Every night in the past weeks there have been Clubhouse chatroom sessions hosted by Andrew Yang, Lucy Ling, Daniel Dae Kim and other outraged Asian American leaders.

Like disease advocacy, ethnic advocacy gets a boost from celebrity headliners, but the real execution comes from grassroots involvement.

I have been reflecting of late on our own industry and what we can do to foster more of the grassroots engagement that leads to lasting change.

For any company on a quest for talent, every good hire is gold dust. We need to hire, retain, promote and develop all our employees. One key departure of any single pivotal employee can set us back on our project timelines. Racial intolerance threatens our companies’ often razor thin chance for success. Overt and institutional racism is clearly not within the remit of any one group to remedy. What can our industry do to convert this into an opportunity for the greater good?

There are things we can do to influence our biopharma ecosystem. BIO and other biopharmaceutical trade associations can play an influential role.

Here are a few thoughts:

  • Encourage top leadership of member companies to roll up their sleeves and directly engage in open dialogue with all employees part of historically underrepresented groups, including Asian Americans. For Asian staff, bring up your awareness of the “model minority” myth. Diversity and inclusion in the workforce has been a token buzzword for decades. That’s not good enough. Leadership and change needs to be demonstrated at the C-Suite and not delegated to task force assignment. Diversity and inclusion is everyone’s issue.
  • Create patient trust at the point of care. Elderly Asian Americans, fearful of racial violence, might not venture out alone to seek vaccinations at healthcare institutions. We can support patient-centric vaccination programs where vaccines can be brought to where the people are, and where they are comfortable – in this case, Asian American community centers and churches. This patient-centric approach has worked well in other underserved communities.
  • Build patient trust beyond the Asian American community to all the patients we serve. Industry can have more impact in providing support at the local level with community leaders and local chapters of patient advocacy groups that directly work with patients. Local patient groups have evolved greatly during the pandemic, driven by patient need.
  • Expand national and local public relations campaigns that educate the public about the value that diversity in biopharma brings to all Americans, as we regain the upper hand in this pandemic.
  • Above all, treat this issue with urgency and action. Racial intolerance in our industry cannot be acceptable. I am proud of our biopharma industry. At our industry’s core, it discovers, develops and commercializes medicines to meet the needs of patients…ALL patients. The industry has developed multiple COVID-19 vaccines in record time. It now has another watershed opportunity to show it is responsive to racial intolerance directed at our Asian American employees and their families, and dedicated to patients of all races.

This is much bigger than Asian American intolerance. It is the essence and mission of our industry.

[Editor’s Note: Paul Hastings, CEO of Nkarta Therapeutics and vice chair of BIO, provided encouragement and feedback on this piece.]

8
Mar
2021

Science Policy Chat: Join Rep. Auchincloss and Me on Clubhouse Mar. 10

Science is poised for a comeback in the public mind.

Most can agree that science, after years of getting beaten up, is leading us out of the crisis. It’s a source of national competitive advantage. This creates an opportunity to double down on investments, to test new ways of working, to shake some things up that need shaking.

Mark your calendars for 7:30 pm ET / 4:30 pm PT Mar. 15 on Clubhouse. [NOTE: The date has been rescheduled from Mar. 10.]

I’m happy to discuss ideas for US science policy with freshman Congressman Jake Auchincloss. I’ll interview him for the first half-hour, followed by another half-hour of audience Q&A.

Rep. Jake Auchincloss, (Democrat, Massachusetts District 4)

Rep. Auchincloss is a Democrat from the Fourth District of Massachusetts. The district extends from the Boston suburbs of Brookline and Newton to the blue-collar industrial towns that stretch to Rhode Island.

He’s an ex-Marine who commanded infantry in Afghanistan and special operations in Panama. Before being elected to Congress in November, he served on the Newton City Council.

Science runs in the family – his father, Hugh Auchincloss, is the principal deputy director to Tony Fauci at the National Institute of Allergy and Infectious Diseases. His mother is Laurie Glimcher, CEO of the Dana-Farber Cancer Institute.

Rep. Auchincloss strikes me as well-schooled on the issues and eager to learn more about biotech. He’s potentially well-positioned as a young and energetic new member of Congress who doesn’t carry some of the baggage of past partisan fights. He identifies as an “Obama-Baker” voter. He’s thinking about bipartisan issues like NIH funding, and ways to invest in young people for bio-manufacturing jobs.

If you are new to Clubhouse, it’s audio-only, invitation-only, and iPhone-only at this point. It appears to be engineered in a way that elevates substantive conversations. It’s sometimes fun. It can be spontaneous, like old-school call-in radio. I’m looking forward to trying out a forum here with Congressman Auchincloss.

For those on Clubhouse already, follow Jake Auchincloss to save this session on your calendar. If you are an active TR subscriber and would like an invitation to join, ask me luke@timmermanreport.com

See you at 7:30 pm ET / 4:30 pm PT Mar. 10 on Clubhouse.

4
Mar
2021

Mental Health: A New Frontier for Biotech

Luke Timmerman, founder & editor, Timmerman Report

Mental health problems were mounting heading into this pandemic.

Now, the challenges are bigger and coming in waves.

There’s the grief. Think about all the family and friends of the more than 520,000 people who have died.

There’s anxiety about getting infected. There’s depression, and loneliness, that stems from social distancing. Addictions to alcohol and drugs are on the rise as people seek ways to cope. Post-traumatic stress disorder is a concern among our healthcare workers. The social media platforms are designed to maximize engagement — they keep us angry and outraged and ultimately exhausted by efforts to sell us stuff.

Data are starting to tell a story.

Depression rates have tripled during the pandemic, according to one study published in JAMA in September. Suicidal thinking among young people was rising before 2020, and surged last year to the point where one in four young adults in the US contemplated suicide, according to the CDC. Heavy drinking episodes shot up 41 percent among women, according to a RAND Corp. study.

Biotech has sprung into action like never before against the virus. Incredible progress has been made there.

What can industry do about mental health?

This week, some of you may have heard my interview with Bob Nelsen of ARCH Venture Partners on The Long Run podcast. The longtime VC, known for being ahead of the curve, spoke of increasing investment in mental health. This isn’t exactly new for him. Nelsen previously invested in schizophrenia drug developer Karuna Therapeutics and depression drug developer Sage Therapeutics.

What struck me most was that Nelsen wasn’t speaking exclusively about the next Karuna and other “hard science” approaches. He also spoke of the “soft” kind of interventions that incorporate some combination of other elements like, presumably, talk therapy, cognitive behavioral therapy, more convenient access, maybe some passive data collection from smartphones equipped with useful analytical capabilities, and maybe some pharmacologic therapy. He didn’t specifically mention the term “digital therapeutics” but there’s a lot of room to re-think mental health treatment in a broader way.

Thinking about those comments, I called a friend.

D.A. Wallach is a general partner at Time BioVentures, a biotech investment firm in the Los Angeles area. He’s curious about science and startups, and a longtime TR subscriber. He’s a successful recording artist. He kindly gave me one of his unpublished songs to start The Long Run podcast in 2017.

That music still fills me with joy each episode.

D.A. Wallach, general partner, Time Bioventures

I wanted to check in with D.A. partly for his thoughts on mental health investing, but moreso for his personal experience with the system.

He suffered a death in the family last year and has sought grief counseling.

Grief, like death, is something our society doesn’t like to talk about. “Nobody knows what to say to you,” D.A. told me.

“Grief isn’t a mental illness, but it’s sort of the equivalent of a mental injury,” he added. “I’d think of it almost like rehab. If you have an injury to your leg, you go to rehab. If you have an injury to your brain, you should go to rehab.”

Like so many people in America, he discovered that it’s a lot easier to get rehab for a leg injury.

First off, it was hard to find someone qualified to talk to. It didn’t take long to figure out why. A market failure was staring him in the face.

Healthcare insurance reimbursement rates are so low in greater Los Angeles that all the best providers refuse to take insurance, D.A. said. They take cash only. Some of the best charge up to $400 an hour.

That means they end up serving primarily wealthy clientele, leaving the average and the below-average practitioners for everyone else with insurance. “You’d have to be a saint to be an incredibly talented therapist and take insurance. You’re choosing to make less money,” D.A. said.

D.A. ended up finding a counselor. He said he’s satisfied with the quality of care he’s been getting via telemedicine.

What’s gnawing him now is the broken system. Los Angeles has developed what amounts to a two-tiered system – high-quality cash-only mental healthcare for the wealthy and lower-quality, insurance-based mental healthcare for everyone else.

That might be too generous of a description. It assumes people who aren’t wealthy can even get access to mental health services at all.

Limited access is the downstream manifestation of deeper ills. Our social-cultural stigma around mental health issues feeds directly into a political system that, as a result, provides only the most parsimonious support for mental healthcare. Naturally, we are left with this narrow funnel of qualified professionals attempting to grapple with millions of people in need.

The gap is enormous. One pre-pandemic study, conducted by the Cohen Veterans Network, a national nonprofit, and National Council for Behavioral Health, attempted to get its arms around this access problem with its inaugural “America’s Mental Health” report in 2018. (Full report).

According to the report:

  • 42 percent of the population considers cost and inadequate insurance coverage as a barrier to accessing mental healthcare.
  • Nearly one in five Americans, 17%, said they have had to choose between getting treatment for a physical health condition and a mental health condition because of their insurance.
  • Almost two-thirds of Americans, 64%, who have sought mental health treatment believe the U.S. government needs to do more to improve mental health services.
  • More than one-third of Americans, 38%, have had to wait longer than one week for mental health treatments.
  • Almost half of Americans, 46%, have had to or know someone who has had to drive more than an hour roundtrip to seek treatment.

These numbers were disturbing in 2018. One year into the COVID-19 pandemic, we need an update.

Jonathan Shaywitz, psychiatrist

Jonathan Shaywitz, a board-certified psychiatrist in Los Angeles with a medical degree from Harvard Medical School, said the demand for mental health services is “the highest it has been for some time.” (If the name’s familiar, he’s the younger brother of TR healthtech columnist David Shaywitz.)

Jon Shaywitz currently handles outpatient services, as well as inpatient services at five facilities. He’s busy. Despite efficiency gains that make it easier to accommodate new patients through telemedicine, Shaywitz said the waiting list for new patients extends out for three months.

Even with increasing awareness of the need, mental healthcare still doesn’t get reimbursed at the same level as physical healthcare. As Jon Shaywitz said:

“This is an area that still needs work — while the law has attempted to create parity between physical and mental health — we are still lacking regarding parity of reimbursement for mental health. Due to the lack of procedures in mental health, our currency is time and talking and unfortunately insurance companies don’t reimburse for time/talking as they do for procedures or imaging. While some of my colleagues still do private practice, the majority are leaving practice and joining large organizations/companies (community psychiatry/Kaiser) where they are salaried and who take insurance.”

With 520,000 dead from COVID-19, how many family members and friends of the deceased out there could use some help coping with their grief? How many people could use help for depression and anxiety?

What can biotech do that might make a difference?

D.A. hasn’t yet invested in mental health, but he’s been studying the opportunities. It’s an area that can’t simply be addressed with pharmacologic solutionism, where the pill is everything, he says.

A more integrated approach that might involve some combination of tech-enabled diagnostic data, behavioral coaching, new treatments, and the empathetic touch from skilled professionals is more likely to succeed. Smoking cessation is one potential model, where the drug is just one tool. Psychedelic therapies for mood disorders are another potential arena for integrated investigation (see in-depth TR coverage of psychedelic drug R&D, September 2020).

Those of us who aren’t neuroscientists or psychiatrists or venture capitalists can do a few things, too.

We can create situations in our companies when it’s OK for people to take a break when they need it. We can lean on our health insurance providers to improve access to mental health services. We can pressure elected officials to raise awareness and provide more funding for reimbursement.

We can support more basic research, from people like Harmit Malik, a member of the National Academy of Sciences. He bravely admitted in a public forum — see below — that many scientists are struggling.  

We can make ourselves aware of the suffering of our friends and colleagues that’s often buried beneath the surface. We can choose to uplift. We can be kind.

We can do more for mental health.

TR readers can continue on for a roundup of the deals and financings and other major developments of the week in biotech.

Vaccines

Delayed Large Local Reactions to mRNA-1273 Vaccine against SARS-CoV-2. NEJM. Mar. 3. (Kimberly G. Blumenthal et al Mass General Brigham)

Fauci: U.S. must stick with two-shot strategy for Pfizer-BioNTech, Moderna vaccines. Washington Post. Mar. 1. (Dan Diamond)

India-based Bharat Biotech reports vaccine trial results from 25,800-volunteer study with 81 percent efficacy against COVID-19. (Bharat Biotech statement)

Science of SARS-CoV-2

High resolution profiling of MHC-II peptide presentation capacity, by Mammalian Epitope Display, reveals SARS-CoV-2 targets for CD4 T cells and mechanisms of immune-escape. BioRxiv. Mar. 2. (Jan Kisielow et al Repertoire Immune Medicines in Cambridge, Mass.)

Estimated transmissibility and impact of SARS-CoV-2 lineage B.1.1.7 in England. Science. Mar. 3. (Nicholas Davies et al London School of Hygiene and Tropical Medicine)

Science Features

Data That Mattered

San Francisco-based VIR Biotechnology and GSK had some bad news from the NIH-sponsored ACTIV-3 clinical trial that evaluated its therapeutic neutralizing antibody against COVID-19 in hospitalized patients. The Data Safety Monitoring Board recommended enrollment in the drug arm be closed after seeing data “that raised concerns about the magnitude of the benefit.” There were no reported safety signals, the companies said. VIR shares fell 28 percent on the news. (NIH Statement on ACTIV-3, which includes antibodies from VIR and Durham, NC and Beijing-based Brii Biosciences.)

Financings

Cambridge, Mass.-based eGenesis said it raised $125 million in a Series C financing. The company uses CRISPR to engineer animal tissues for transplant into humans – starting with kidneys for kidney failure and islet cells for type 1 diabetes.

Foresite Capital raised $969 million in its Fund V and Opportunity Fund.

Paris-based Sofinnova Partners, a European-focused life science fund, raised a new $540 million late-stage crossover fund.

Berkeley, Calif.-based Caribou Biosciences raised $115 million in a Series C financing to use its CRISPR technology to further advance work on allogeneic, off-the-shelf engineered immune cell therapies for cancer. Farallon Capital Management, PFM Health Sciences, and Ridgeback Capital Investments co-led.

Philadelphia-based Century Therapeutics raised $160 million in a Series C financing. The company is developing induced pluripotent stem cell-derived therapies for cancer. Casdin Capital led. (Listen to Century chief strategy officer Janelle Anderson on The Long Run podcast, Aug. 2019).

Waltham, Mass.-based Morphic Therapeutic seized upon its surging stock price to put together a new offering of shares at $70 apiece. The company, the developer of oral small molecule integrin inhibitors, took home a fresh $245 million from the offering. (See Tuesday’s TR coverage of Morphic’s proof-of-concept clinical data).

San Diego-based Janux Therapeutics raised $56 million in a Series A financing led by Avalon Ventures. The company is developing “Tumor Activated T Cell Engager” technology.

South San Francisco-based Amunix Pharmaceuticals said it raised $117 million in a Series B financing led by Viking Global Investors. The company is developing protease-activated T cell engagers and cytokines for the treatment of solid tumors.

UK-based Exscientia, an AI drug discovery company, said it raised $100 million in a Series C financing that included Blackrock funds, Novo, Bristol Myers Squibb and others.

MassBio released a report this week finding that Massachusetts biotech companies raised a record-breaking $5.8 billion combined in venture capital in 2020. That shattered the previous record of $4.8 billion from 2018. A remarkable 21 biotech companies from Massachusetts alone went public last year, raising another $3.9 billion, the trade group said. (Summary and link to full report).

Access and Manufacturing

Merck, likely suffering from a bruised corporate ego after its COVID-19 vaccine candidates failed, agreed to help Johnson & Johnson manufacture its adenoviral vaccine that was just authorized for distribution by the FDA. The Biomedical Advanced Research and Development Authority (BARDA) agreed to provide $269 million to Merck to set up the necessary processes to make the J&J vaccine. President Biden later said the plan is to make enough vaccine to vaccinate all American adults by May 31. Previously, Novartis and Sanofi said they would help produce the Pfizer / BioNTech vaccine.

WuXi Apptec completed its acquisition of Oxgene, a UK-based contract manufacturer of cell and gene therapies.

San Diego and Boston-based Resilience acquired two new biotech drug manufacturing facilities. One is the 310,000-square foot landmark along the Charles River in Boston, originally developed by Genzyme and now part of Sanofi. The other is a 136,000-square foot facility in Mississauga, Ontario. Resilience said it has now assembled a network of factories with 750,000 square feet in North America. It will continue to invest in upgrading and adapting the facilities for production. (Bob Nelsen of ARCH Venture Partners, co-founder of Resilience, discussed the company in my latest Long Run podcast).

Cambridge, Mass.-based Biogen said it plans to build a new gene therapy manufacturing plant in Research Triangle Park, North Carolina. It’s planning to scale up future gene therapies for neurological disorders. Biogen plans to hire 90 new workers there, and invest $200 million.

Germany-based CureVac, a less-known mRNA vaccine and therapeutics developer, agreed to work with Novartis to help scale up production of its mRNA vaccine candidates in the second quarter of 2021. The companies said they expect to be able to make 50 million doses at a Novartis facility in Austria in 2021, and another 200 million in 2022.

Deals

Amgen agreed to pay $1.9 billion to acquire South San Francisco-based Five Prime Therapeutics, a company that’s been on a 20-year odyssey in biologic drug discovery and development. Amgen pointed to its interest in bemarituzumab, an anti-FGFR2b antibody being prepped for Phase III study in gastric cancer. (Five Prime founder Lewis T. “Rusty” Williams is a member of my Everest Base Camp trekking expedition to benefit Fred Hutch. It’s been on hold because of the pandemic, and now looks like a Spring 2022 event. Can’t wait to ask him about Five Prime on the trails of Nepal).

Takeda Pharmaceuticals took back full rights to soticlestat (TAK-935/OV935) for developmental and epileptic encephalopathies, including Dravet syndrome and Lennox-Gastaut syndrome. New York-based Ovid Therapeutics had been its development partner since 2017, and helped drive the asset through Phase II testing. To get back full global rights, Takeda agreed to pay Ovid $196 million at closing and another $660 million in milestones, plus royalties. Ovid stock, beaten down after a clinical failure with its candidate for Angelman syndrome, jumped back up 37 percent on the news that it was getting the cash infusion.

AbbVie obtained an exclusive option to acquire San Francisco-based Mitokinin once it completes IND-enabling studies. Mitokinin is developing drugs to increase the activity of PINK1 a master regulator of mitochondria. The company, spun out of work at UCSF by Nicholas Hertz and Kevan Shokat, is betting that by increasing the activity of PINK1, it can address one of the underlying problems that contributes to Parkinson’s disease. Terms weren’t disclosed. The startup was backed by Mission Bay Capital.

Santa Clara, Calif.-based Agilent Technologies agreed to pay $550 million in cash at closing, plus $145 million in milestones, to acquire Kirkland, Wash.-based Resolution Bioscience, which uses liquid biopsies and next-gen sequencing technology for the diagnosis of cancer.

The Defense Advanced Research Projects Agency (DARPA), the government agency that made a visionary investment in mRNA vaccines for pandemic preparedness in 2013, awarded a grant to Georgia Tech Research Institute to develop technology to detect SARS-CoV-2 in the air in real-time. San Diego-based Cardea Bio is a subcontractor on the deal. If successful, the detectors could conceivably be used in schools, offices, restaurants and other indoor environments where people might wonder about whether the virus is circulating.

Seattle-based Presage Biosciences said it raised $13 million in a round that included $7 million from new investors LabCorp Venture Fund, Bristol Myers Squibb, and InHarv Partners Ltd. Presage also said it established new research agreements with Merck and Maverick Therapeutics. The company uses intratumoral microdosing to evaluate several cancer drugs simultaneously in Phase O trials.

Grail, the Menlo Park, Calif.-based developer of next-gen sequencing enabled tests for early detection of cancer, said it struck an agreement with Providence, a major West Coast hospital network, to offer Grail’s Galleri test that’s scheduled to become available in the second quarter of 2021. It’s not a surprise that Providence would be an early adopter of this next-gen sequencing enabled healthcare application – it acquired the Institute for Systems Biology, led by DNA sequencing pioneer Leroy Hood, in Mar. 2016. (Read “Hood: Trailblazer of the Genomics Age”).

Our Shared Humanity

The California Life Sciences Association (CLSA) published a Racial & Social Equity Plan, which includes specific actions and a $1 million commitment over three years to get the ball rolling.

Regulatory Action

The FDA released its Data Modernization Action Plan, about 18 months in the making. There are three main elements – identifying valuable demonstration projects, developing consistent and repeatable data practices across the agency, and build up and sustain a stronger talent network (internally and with external partners). (Executive Summary)

Merck said it’s voluntarily withdrawing one of its approved indications for the PD-1 inhibitor pembrolizumab (Keytruda). It’s for metastatic small cell lung cancer. The drug was granted accelerated approval by the FDA in 2019 on a surrogate endpoint – tumor response – but full approval was supposed to be contingent on demonstrating an Overall Survival benefit. The drug failed to clear that higher bar of evidence in January 2020, and now the indication is being taken out of the prescribing label.

The FDA, in an unusual public statement, waded into a mess around Brainstorm Cell Therapeutics and its experimental NurOwn treatment for amyotrophic lateral sclerosis. The company announced Feb. 22 what looked like unequivocal bad news – that the FDA said it doesn’t have enough evidence of efficacy to submit a Biologics License Application. The company, however, appeared to muddy the waters and perhaps stir some false hope by adding that “FDA advised that this recommendation does not preclude Brainstorm from proceeding with a BLA submission.”

The FDA — under pressure for years to release full Complete Response Letters to investors, doctors and patients so that companies can’t engage in one-sided and misleading communications about new drug applications – decided to clear things up. The agency said:

Although FDA generally cannot provide confidential information about unapproved products, given the tremendous public interest in this product, we have concluded that it is important to provide high-level information about the status of the NurOwn development program.

With the recent completion of a randomized phase 3 controlled clinical trial comparing NurOwn to placebo, it has become clear that data do not support the proposed clinical benefit of this therapy. Data indicated that none of the primary or secondary endpoints were met in the group of patients who were randomized.

The agency got into some more specifics in case anyone is wondering if this is a close call, or the agency is being wishy-washy. The take-home lesson: the FDA has to step up its communications game in the age of transparency, especially with companies who are leaving people with misleading impressions.

The FDA granted a label expansion to Pfizer to start marketing lorlatinib (Lorbrena) as a treatment for patients with newly diagnosed ALK-positive non-small cell lung cancer. The FDA also converted an accelerated approval from 2018 into a full approval, based on results from the CROWN study which showed a 72 percent reduced risk of disease worsening or death for ALK-positive patients on the medicine.

Personnel File

Cambridge, Mass.-based Fulcrum Therapeutics said CEO Robert Gould is retiring at the end of March, and will remain on the board of directors and function as an advisor. The rare disease drug developer is promoting chief operating officer Bryan Stuart to president and CEO. (TR coverage of Fulcrum, Sept. 2018).

Cambridge, Mass.-based Solid Biosciences, the developer of gene therapy for Duchenne Muscular Dystrophy, said Erin Powers Brennan was hired as chief legal officer and Joel Schneider was promoted to chief operating officer.

Dilawar Syed was nominated by President Biden to be the deputy administrator of the Small Business Administration. He’s currently CEO at Lumiata, an AI-for-healthcare company.

Waltham, Mass.-based Dyne Therapeutics, the developer of treatments for muscle diseases, said it hired Wildon Farwell as chief medical officer. He was VP of neuromuscular at Biogen.

Cambridge, Mass.-based eGenesis, the CRISPR for xenotransplantation company that recently raised $125 million in a Series C financing and is almost surely getting dressed up for an IPO, hired Sapna Srivastava as chief financial officer. She previously worked as chief financial and strategy officer at Abide Therapeutics, and before that had a similar job at Intellia Therapeutics.

Chicago-based OMX Ventures, a new fund that says it’s investing in companies “at the intersection of biology, big data and engineering,” hired Jamie Kasuboski as a vice president on its team of investing professionals. Jamie previously worked at RA Capital, and Boehringer Ingelheim Venture Fund. Kevin Ness, former CEO of Inscripta, also agreed to become an advisor to OMX.

San Diego-based Viracta Therapeutics, a company working on treatment for viral-associated cancer, added Stephen Rubino and Barry Simon to its board of directors. The company also announced a series of promotions in operating roles.

4
Mar
2021

What Pharma Data Scientists Can Learn from SpaceX, Health System Barriers, & the FDA

David Shaywitz

Three quick data science items:

1) How pharma companies could engage more constructively with data scientists.

2) How health system barriers to data sharing inhibit robust evaluation of the underlying science.

3) The savvy way the FDA is thinking about data science.

  1. Learning From SpaceX

On Wednesday, Elon Musk’s SpaceX landed a prototype spacecraft vertically on the ground — a remarkable engineering accomplishment, and one with important lessons for pharma companies harboring digital aspirations.

SpaceX has been working on this vertical landing for a while. Two previous prototypes crashed to earth with spectacular explosions; even Wednesday’s successful landing was followed in minutes by another large explosion, perhaps because of a “leak in a propellant tank,” the New York Times suggested.

While presumably not pleased by these failures, Musk appeared to see them for what they were, part of the inevitable iterative learning experience that’s required for success with any new technology.

In contrast, many of the data scientists I know in pharma companies feel they have far less room for error. The institutional powers that originally hadn’t welcomed such data scientists at all now have let them in, but often under the equivalent of Dean Wormer’s “Double Secret Probation” (cue up the scene from “Animal House”). 

Many data science teams in pharma feel they have a single chance to prove themselves, and if whatever they are trying to do doesn’t work brilliantly, the data scientists worry they’ll be booted, the technology dismissed as not ready for prime time. 

This seems precisely the wrong mindset for effective technology implementation. The truth is that it takes time and engagement to figure out any new technology – to learn how to use it effectively in a particular context. The way you do this is by trying something provisionally, seeing what works and what doesn’t, making adjustments, and quickly trying again.

It’s an iterative process that allows for rapid adaptation. This allows far more agility than more classic organizational approaches that insist on the pre-specification of almost everything.

Giving new technology a single shot, and requiring perfection straight out of the gate, sets it up for failure. This sort of rigid thinking ultimately doesn’t allow pharma companies to access the power and benefits that data science and emerging technologies have to offer.

  1. Irreproducible

The core premise of science lies in its reproducibility; my description of an experiment should afford you the opportunity to evaluate my math and methods, and to obtain the same results in your own hands.

But this can be a real challenge when the underlying data aren’t shareable – as is often the case with health data studies. 

This came up most recently in a departmental journal club I attended, where a guest professor led a captivating discussion of a just-published paper (not hers) that described a particular application of EHR data. 

Towards the end of the hour, the focus turned to replication – were the raw data underlying the conclusions in the paper available for review?

Here’s what the text actually stated:

“DATA AVAILABILITY
The data used for this study are available from the [redacted] health system,
but restrictions apply to the availability of these data, which were used under license for the current study, and so are not publicly available. Data are, however, available from the authors upon reasonable request and with the permission of [redacted] Health System.“ (redactions mine).

The way this statement was universally interpreted by 30 or so experienced attendees of this journal club was, as several people actually said, “good luck.” 

It was universally understood that no one was ever going to actually access these data. 

A couple of points: the first is that health systems typically see no upside in sharing their data. They generally only share the minimum amount possible and only after the maximum amount of duress.  Many reasons are often invoked, but a key driver is that there’s no economic incentive for health systems to share, and plenty of reasons not to (including fear of divulging information to competitors). Hence, we are stuck in something of a rut with minimal data sharing.

It’s not just health systems, however. Individual researchers tend not to be especially eager to share their data either.

This isn’t universally true, of course, and important exceptions exist. Some scientists are quite open with their data, and have laudably managed to simultaneously advance both science and their careers – the pioneering work of Daniel MacArthur on the Exome Aggregation Consortium (ExAc) comes to mind.  

Nevertheless, many investigators resist sharing “their” data – for reasons that are understandable, if perhaps not always justified. After going through the often arduous process of gathering clinical data, researchers are inclined to guard it so their own team can benefit from this intense upfront effort – a key component of the recent “data parasite” debate. Other times, researchers worry the data will be coarsely reanalyzed, perhaps without adequate understanding of relevant nuance, and legitimate conclusions called into questions by naïve, crusading, attention-seeking critics — a plausible concern.

From the perspective of many researchers, what’s the upside?  Many would say: just about none.

This situation evokes the classic academic joke in which a young faculty member is advised to study seven-year locusts – by the time the work can be called into question, the professor will have already achieved tenure!

But to the extent that conclusions drawn from health data can’t be re-evaluated by others because of issues with data access, the science may not be adequately pressure-tested, and flawed methods may remain unchallenged. 

This is bad for the discipline of health data science, and ultimately bad for patients.

  1. Good News From FDA

Finally, some welcome good news from the FDA, which just released its Data Modernization Action Plan (DMAP) – a companion to the previously released Technology Modernization Action Plan.

I was especially struck (though not surprised, given what DMAP co-author and deputy FDA commissioner Dr. Amy Abernethy has consistently preached) by the emphasis on “high value driver projects.” Dr. Janet Woodcock, acting FDA commissioner, is the other co-author.

As the document states:

“The DMAP is anchored on driver projects that help generate value while building critical capabilities. Driver projects for DMAP are defined as initiatives with measurable value that help multiple stakeholders envision what is possible, allow technical and data experts to identify needed solutions, and develop foundational capabilities. This strategy is distinctly different from focusing on data collection and then looking for questions the data can answer.” (emphasis in original).

In short, this approach gets right what so many get wrong – the importance of collecting and evaluating data with a specific purpose in mind. The intended purpose strongly influences what data are needed and the degree of subsequent validation and refinement required. 

This mindset is likely to be far more productive than approaches that robotically dump all data into some kind of lake and then proudly reports how big the lake is. 

The DMAP approach is so much savvier, and far more likely to yield meaningful results. This methodology also inherently refines the mechanism of fit-for-purpose data collection; as Dr. Abernethy has stated on a number of occasions, you don’t truly understand a dataset, including its limitations, until you really start to use it.

If FDA starts with these well-defined driver projects, and has success, then there will be more natural momentum to spread the successful practices across the agency.

The pragmatic driver project approach (versus creating a data lake upfront and hoping to figure out how to capture value later) is one that many pharma companies and healthcare organizations would do well to emulate.

3
Mar
2021

J&J Vaccine: One Dose Delivers Strong Immune Response and Protection

Larry Corey, MD

The mRNA vaccines from Pfizer / BioNTech and Moderna have understandably dominated the news, but this past week was Johnson & Johnson’s turn in the spotlight with another important contribution to the COVID-19 vaccination effort.

The FDA’s vaccine advisory committee recommended, and the FDA authorized the vaccine, based on results from the 44,000-person Operation Warp Speed–sponsored ENSEMBLE clinical trial evaluating the Johnson & Johnson (J&J) Ad26 vaccine.

This vaccine uses a different way for the spike protein transcript to be introduced into the cells. This transcript, essentially the same as with the mRNA vaccines, is carried in—not by a lipid particle encasing the RNA of the SARS-CoV-2 spike protein—but by a genetically engineered type 26 adenovirus, which has the SARS-CoV-2 spike gene encoded into it.

What happens next is similar to RNA vaccines—the spike protein is made, and the immune cells begin to recognize the new structure and initiate the process of defining anti-SARS-CoV-2 antibodies and T cells to defend against it.

The J&J vaccine has several different immunological characteristics than the RNA vaccines. The neutralizing antibody levels generated by a single dose are not nearly as high as those seen in either the Moderna or Pfizer vaccines. But the T-cell responses in the J&J vaccine, what we call cell-mediated immune responses, appear higher, especially in those we call killer T cells, or the CD8+ cytotoxic T cells, which seek out and destroy cells that are infected with the virus.

These killer T cells are an important part of the story, especially because they tend to both arise earlier and can persist at effective levels with less variation in titer than neutralizing antibodies.

Another important difference in the J&J vaccine trials is that they were designed through an international lens: J&J is a worldwide company with high production capabilities, which eases the burden of distribution. The Ad26 vaccine doesn’t have the cold-chain requirements of the mRNA vaccines and is quite stable at 4 degrees Celsius for extended periods of time.

The Ad26 vaccine platform, importantly, has a long track record and is well-known around the world. It has been used in Africa for preventing Ebola infections, in which the Ebola surface protein is placed into the Ad26 shuttle vector. The Ebola vaccine has been WHO (World Health Organization) authorized for preventing Ebola infections and has been widely administered to women of child-bearing age, pregnant women, and children. The Ad26 platform is also under investigation in elderly adults for RSV (Respiratory Syncytial Virus) infections and in Africa and South America for a preventive HIV vaccine.

The ENSEMBLE Trial was conducted in the United States, Mexico, South Africa and in South America—Argentina, Brazil, Chile, Colombia, Peru. The trial enrolled approximately 44,000 persons, of which 19,302 were in the US; 479 in Mexico; 17,426 in South America; and 6,576 in South Africa. The geographic diversity turned out to be an important feature of the data set, in the context of a pandemic virus with evolving variants becoming prevalent in different parts of the world.

The initial analysis from the J&J trial data provided an extensive look at the vaccine’s effects. More than 400 cases of COVID-19 were reported in the data set: this large data set allowed for a reliable look at the efficacy by country and region without analytical problems posed by too-small sample sizes.

Trial planning was done before the new variants were identified. Hence, it was both fortuitous and extremely lucky that within two months of identifying variants of concern, we “caught” the wave of the introduction of these variants in both Brazil and South Africa. Thus, we now have real-world data on how the B.1.351 variant that has multiple resistant mutations does against a COVID-19 vaccine with the Wuhan-like virus as the immunizing strain.

The point estimates for mild-to-moderate disease showed some difference by region—after one dose, the point estimate was 72% in the US; in Brazil, 71.5%; and in South Africa, 57.3%. This was far better than the point estimate of the AstraZeneca vaccine to the B.1.351 variant (10.1%).

Fortunately, and importantly, the efficacy against severe COVID-19 disease was high in all countries. In the United States, there was only 1 case of severe disease in the vaccine group, 7 in placebo. For South Africa, it was 4 in the vaccine group, 22 in placebo; for Colombia, 1 in the vaccine group, 11 in placebo; and in Brazil, 1 in vaccine, 8 for placebo. So, in this one trial, the J&J one-dose vaccine offered anywhere from 82% to 90% protection against severe disease. The 6 deaths from COVID-19 in South Africa were all in the placebo group.

There is a question about whether the J&J vaccine will be more effective in a two-dose formulation. We don’t know that answer yet. But for a one-dose vaccine, these data are better than I initially expected. To illustrate this, I’ll include the two Kaplan-Meier curves below, which nicely demonstrate the J&J vaccine efficacy on moderate-to-severe disease in South Africa and the disease in severe COVID-19 for the entire study.

The ENSEMBLE study outlines the complex nature of the human immune response to vaccination and the subsequent outcome.

Let’s come back to the question about what the vaccine’s effect on the immune response, and how that immune response translates into protection against severe disease.

The antibody levels with the one-dose J&J vaccine are much lower than what we see with the mRNA vaccines and yet its effectiveness against preventing severe disease—particularly against the B.1.351 South African variant—is quite similar.

Why?

What components of the immune response are central to this protection for severe disease? Antibodies? T cells? Both? These are issues we must decipher, and many of us expect that having high levels of CD8+ T cells activated to recognize the virus is an important part of the reason this adenoviral vaccine provides such excellent prevention against severe disease. It is a nice scientific problem to have, how to go from 85% to 100% efficacy.

The question in front of us today is how do we use this vaccine in our country and globally? To date, it is the only vaccine in which we have good clinical data about both its effects in mild disease as well as severe disease with the South African variant — the cause for recent concern worldwide.

The data from the two mRNA vaccines suggest that they are able to effectively handle the other emerging B.1.1.7 and the California 20.C variants in the US. It is, however, the South African variant that we are concerned about with respect to all the platforms in our current portfolio. As I mentioned earlier, the South African variant is one in which the two-dose AstraZeneca vaccine (chimp adenovirus-based vector) showed no effectiveness in reducing even mild-to-moderate disease in young, healthy South Africans. The point prevalence was 10% with the two-dose AstraZeneca vaccine versus 59% in the one-dose J&J vaccine.

Seeing the disappointing efficacy against the B.1.351 variant that was quickly evolving into the dominant local strain, the South African government—after purchasing and receiving more than a million doses of the AstraZeneca vaccine—made the decision to halt administration of the AstraZeneca vaccine to healthcare workers. Instead, South African authorities opted to work with J&J to obtain its vaccine in an expanded access program. This dynamic interaction between clinical trial data and health care policy is what all these trials have been about. It’s about following the data and adjusting accordingly.

The J&J vaccine is an extremely well-tolerated vaccine—less than 0.5% of enrollees had any grade 3 side effects. It is easy to transport and distribute without the strict cold-chain requirements. There is extensive clinical experience with pregnant women and children. Even though there is somewhat less efficacy with the J&J vaccine in preventing illness—such as a cough, fever, headache, or sore throat—than with the mRNA vaccines, the data from the J&J trial show that the severity of illness is significantly less after vaccination.

This vaccine will keep people out of the hospital, and prevent people from dying. I am gratified to see that both the FDA and CDC ACIP (Advisory Committee on Immunization Practices) advisors endorsed use of this vaccine for those 18 years of age and older. We are pushing J&J to conduct its pediatric trials expeditiously so the advantages of a one-dose vaccine can be used to help get children immunized and back to school by the fall term.

Combating this outbreak requires achieving high vaccine coverage – the percentage of the overall population that’s received a vaccine. The higher that number goes, the greater the chances are that we can stop the spread of new variants.

For all these reasons, the data support the widespread use of this vaccine in our country and around the world.

Dr. Larry Corey is the leader of the COVID-19 Prevention Network (CoVPN ) Operations Center, which was formed by the National Institute of Allergy and Infectious Diseases at the U.S. National Institutes of Health to respond to the global pandemic, and the Chair of the ACTIV COVID-19 Vaccine Clinical Trials Working Group. He is a Professor of Medicine and Virology at University of Washington and a Professor in the Vaccine and Infectious Disease Division and past President and Director of Fred Hutchinson Cancer Research Center.