1
Nov
2020

Rebuffed as Overlords, AI Experts Return in Peace, Seeking Partnership with Clinicians

David Shaywitz

Why not healthcare?

That’s the core question at the heart of efforts to apply emerging digital and data technologies to healthcare and life science. 

As Suchi Saria, an entrepreneur and a computer scientist at John Hopkins, where she directs the Machine Learning and Healthcare Lab, puts it, in the 2000s, these technologies transformed sectors, such as banking, in a fashion that was “kind of amazing.” 

Artificial intelligence (AI), operating on these rich data, profoundly changed and improved the way business is done. 

Take, for example, banking fraud detection. The industry “can’t imagine doing it without AI, and with AI they’ve increased sensitivity dramatically, timeliness dramatically” and have far improved specificity, Saria notes.

Contrast this with the AI experience in healthcare. In the last decade, healthcare has “basically spent a ton of our investments” to go “from no data to data, going from no digital infrastructure to additional infrastructure,” says Saria, but yet, “when we think of AI, we think of it as a thing that could be transformative, that has the potential, that is in the future.”

Suchi Saria, Founder, Bayesian Health; John C. Malone Associate Professor and Director of AI & Healthcare, Johns Hopkins University

What we’re missing, insists Saria, is that the future is now – “in reality, today. Now that the data exist, the use of AI in deriving value from the data that’s being collected is the single biggest opportunity in healthcare.”

It’s a hopeful perspective, though of course not universally shared.  But it is hotly debated, perhaps nowhere more intelligently than at the recent, inaugural SAIL pre-symposium (virtual, of course) focused on AI and health, and featuring many of the field’s most thoughtful voices, including data scientists, clinicians, administrators, and even the Editor-in-Chief of the august New England Journal of Medicine (NEJM). All offered comments that were almost invariably germane, focused, and informative, representing the best of what conferences can be. You can watch the whole thing yourself here

Six major themes emerged in my notes:

  1. The challenge of outcomes – what should we, and can we, seek to optimize?
  2. The centrality of bias – the need to ensure AI isn’t perpetuating and exacerbating inequities.
  3. The consensus for the “doctor and AI” mindset – rather than “doctor or
  4. Promising use cases – the “green shoots.”
  5. Opportunities in evidence generation – and why leveraging electronic medical record data remains so hard.
  6. Stubborn hurdles and implementation challenges – including interoperability, data access, and conflating “interesting” and “important.”
The Challenge of Outcomes

Almost by definition, the goal of medicine is to improve outcomes. As NEJM editor Eric Rubin puts it, “we are interested in the impact on the patient,” adding “the closer we can get to something that we care about, the better off we are.”  

Similarly, the lens through which UnitedHealth Group’s Chief Scientific Officer, Ken Ehlert, views potential AI solutions is “are we getting a better outcome?” Entrepreneur and academic ophthalmologist Michael Abramoff also stresses the importance of focusing on outcomes.

But such focus turns out to be easy to say but far more difficult to operationalize, as Harvard’s Zak Kohane points out.

“We’re not very good at looking at outcomes because the systematized capture [of outcomes], whether in trials or EHRs, is noisy, confounded.” He predicts that “many, if not all the AI programs that are going to be deployed in the next 10 years will be poor with respect to outcomes and rich with respect to either human labels or intermediate process measures.”

Zak Kohane, Chair of the Department of Biomedical Informatics, Harvard Medical School

These endpoints can seduce and mislead us, he suggests, leading us to optimize for something we regard as a proxy for meaningful outcomes, yet which may ultimately not be linked to the outcomes as closely as we’d like to imagine.

Kohane (a pediatric endocrinologist) cites the example of diabetologists seeking to improve microvascular disease by focusing on driving down the levels of glycosylated hemoglobin (HbA1c); this turns out to work, to a point, in terms of reducing kidney damage, but pushing “too” intensively for very low HbA1c levels was ultimately found to increase the risk of death. We were “misled by the process outcome in this case,” Kohane says. “For adults, minimizing glycohemoglobin was actually the wrong thing.”

“Medicine,” reflects Kohane, “is a beautiful art but it’s barely a science.  As a result, many of our intuitions of what constitutes a solid correlate to outcomes, again and again, gets proven to us to be wrong.”

It’s also important to recognize, as University of Utah Health’s Chief Medical Information Officer, Maia Hightower, points out, that “the outcomes that we as clinicians may see as important may be different than what our communities see as important.”

The Ubiquity of Bias

Despite, or perhaps because, of a series of high-profile failures (like an AI-powered image classifying program that was able to recognize categories as fine as “Graduation,” yet mislabeled people of color as “Gorillas”), the AI community has tackled this challenge. The AI community has transformed itself from laggards to leaders, as Brian Christian captures in a captivating new book, The Alignment Problem, that I recently reviewed for the Wall Street Journal.

Duke University computer scientist Cynthia Rudin highlights a prismatic example of bias – an insurance company algorithm that aimed to predict which patients might need more care in the future, and thus might benefit from extra (anticipatory) services today. 

The company used “cost as a proxy for care,” in their modeling, Rudin says. “The only problem is that black patients were receiving lower cost health care. They weren’t less ill. They were just receiving lower cost health care.” But these patients would have been systematically underserved by the algorithm’s recommendations. 

Anant Madabhushi, who directs the Center for Computational Imaging and Personalized Diagnostics at Case Western University, offers another example from his own research on prostate cancer.  Black men “tend to have more severe disease,” Madabhushi says, and “potentially higher incidence of prostate cancer,” yet “a lot of the existing risk models that we currently have for prostate cancer have been built largely with a plurality of non-black men represented in those datasets.” 

Attuned to the possibility of racial differences in the disease – as IBM Watson’s Tiffani Bright points out, “you can’t measure what you don’t know about” – Madabhushi uncovered “actual differences in the area around the tumor” in pathology specimens taken from black men and white men.  From this, they “created a dedicated model” that “resulted in a much higher accuracy in predicting risk of recurrence,” compared to a “population-agnostic model.”

At one point, there might have been a collective sense that the best way to avoid bias is to avoid collecting data that might predispose to bias, like race. But a key theme emerging from both this discussion and Christian’s book is that appropriately collecting and thoughtfully considering these data can be essential and invaluable.

“From an operations perspective,” explains Hightower, there’s now the “expectation within the healthcare system [that we’re] capturing all other types of data to tell the complete story of our patients.” She says they “do gender pretty well,” but are “not as good with race. And definitely when we talk about preferred language and LGBTQ+ status, it starts to deteriorate even more.”

As Kohane points out, such information can be critical. Consider hereditary breast and ovarian cancer (HBOC), associated with mutations in the BRCA1 and BRCA2 genes. Ashkenazi Jews are at significantly greater risk of carrying one of these mutations; according to a genetic counselor at Jackson Laboratory, “one in 40 Ashkenazi Jewish individuals versus one in 400 people in the general population carry a mutation in BRCA1 or BRCA2.” If a provider does not customize their calculation, “if they do not discriminate based on the ethnicity of the patient being a Jewish woman,” Kohane notes, “they are actually underserving that patient in a very unfortunate way. And I think that [customizing treatment based on factors that, where appropriate, include ethnicity, for example] is going to be increasingly true as we get more precise about our medicine.”

Marzyeh Ghassemi, Assistant Professor, University of Toronto, Computer Science and Medicine

Marzeyeh Ghassemi, a Canadian computer scientist focused on the application of machine learning to healthcare, and whose lab will be moving to MIT next summer, has thought deeply about fairness and bias. She is especially concerned about “unjust bias” – bias that “perpetuates systemic structural injustice that’s been visited upon a certain group for many reasons.” 

One example she cites: the historical tendency for women’s pain to be “ignored when they go to the doctor.” Consequently, she says, if we feed that data into a model, “we make an algorithm that perpetuates that structure, systemic injustice. That’s a bias that’s bad, and we don’t want to do that.”

One specific suggestion Ghassemi offers: regulators requiring “performance guarantees across different subgroups.” She emphasizes considering such performance proactively, and demonstrating it, represents a better solution than perhaps the more convenient alternative of “narrowing the scope of your claims” – i.e. seeking approval only for a single group. Otherwise, she says, “we going to end up with a lot of devices and algorithms and bells and whistles and treatments that only work on wealthy white people.”

Kohane points out that the computer science community was two decades ahead of medicine in embracing open access publishing. “There is an interesting set of precedents where good societal behavior has actually been pioneered by the computational community,” he says, suggesting that perhaps computer scientists, as they seek to bring AI to medicine, could set another good example here as well.

Doctor and AI

While many journalists seem permanently stuck on the “will AI replace doctors?” storyline, the field itself moved on a long time ago, driven, it seems, less by political expediency – the idea that AI will be an easier sell if doctors are less threatened by it – than by authentic scientific humility.

Many leading AI practitioners have recognized the limitations as well as the power of their computational tools, and see in partnerships with people an opportunity to at once bring out the best from both computer and human while also guarding against some very real concerns.

Among the top worries: the unexpected fragility of AI algorithms. Approaches that seem to work brilliantly in a defined set of circumstances may fail catastrophically when the situation is changed – even imperceptibly. 

For example, fascinating studies involving “adversarial attacks” have revealed that a seemingly sophisticated image recognition algorithm can be tricked by simply altering, in some cases, a single (well-chosen) pixel. Similarly, AI researchers working in healthcare have become increasingly worried about black box algorithms delivering misguided recommendations based on subtle flaws that may lurk undetected – as I discussed at the start of my recent WSJ review of Brian Christian’s The Alignment Problem.

As Duke’s Rudin (also one of the stars of Christian’s book) explains, right now, when thinking about the application of AI to many aspects of healthcare, “we don’t trust our models.” They might be “reasoning about things the wrong way.” 

To leverage the power of AI while mitigating the risks, Rudin’s group is focused on using AI to develop clinical decision aids that are based on simple point schemes, to derive the sorts of scores that physicians are already accustomed to calculating – to ballpark a patient’s cardiovascular risk, for example.  

The trick is using the sophisticated AI to figure out the most relevant variables to measure (a computationally difficult problem) and distill these parameters into simple integers, which a busy physician can still add up and evaluate in the context of the patient.

Cynthia Rudin, professor of computer science, Duke University

The scores produced by this approach, Rudin says, “are just as accurate as any model you can construct.” Plus, they offer the conspicuous advantage of being interpretable – the physician can “really understand how the variables work together jointly to form a final prediction,” she adds. Furthermore, “being able to have the human in the loop actually helps you with the uncertainty that you can’t quantify – the ‘unknown unknowns.’”

There’s also a hope that AI can help aggregate, organize, and prioritize the huge amount of information physicians and other health providers need to contend with, helping to distill for them the information they need – when they need it.

Both Microsoft’s head of research Peter Lee, and UnitedHealth’s Ehlert, for example, envision AI “augmenting what humans can do, absorbing and integrating knowledge for better decisions” as Lee puts it. The ability to process health information in real time, Saria believes, will enable medicine to (finally…) transition from a “reactionary paradigm to an anticipatory paradigm,” anticipating disease in time to prevent it or at least head it off at an early stage.  For example, argues Saria, “AI is pretty much the only way to identify conditions like sepsis, and patients at risk of sepsis, early and precisely.” 

Saria cites the management of stroke as another example, where AI can rapidly identify likely occlusive events, which a radiologist can immediately review and potentially validate, facilitating the timely triage of patients to a comprehensive stroke center for appropriate treatment.

Both Saria and Ehlert also flag the opportunity for AI to offer providers reference values for measurement that are personalized and contextualized for each patient, rather than based on average values for the population as a whole.

Columbia University biomedical informaticist Nick Tatonetti may have captured the shared sentiment the best, observing:

“Medicine is really about human interactions. Caring for somebody and curing someone of a disease is an extremely human activity. And humans should be centered in that process. A lot of technology that’s been introduced in health care has been rightly criticized for getting in the way of the patient doctor relationship, that human connection. There really is an opportunity for technology not to get in the way any longer, but start to disappear into the background and really put that interaction in the center.”

Uses Cases

Several speakers offered concrete examples of the application of AI in medicine. A particularly intriguing example, presented by Greg Hager, a computer scientist and director of the Malone Center for Engineering in Healthcare at Johns Hopkins, focused on surgical training. 

Hager explains that the widespread adoption of the da Vinci surgical system, which enables surgeons to operate with robotic assistance, almost as if they’re playing a video game, offered a remarkable opportunity. The da Vinci’s recording of all aspects of a surgical procedure, from stereo videos to the force applied to the instruments, Hager realized, generates a fantastically useful dataset.  By thoughtfully bringing AI to bear on these data, Hager and his colleagues break procedures down into steps, and “evaluate the quality performance of those steps.”

This analysis can determine “whether it’s an attending [senior physician] or a trainee who’s operating, just by the quality of the performance in that data.” Plus they can feed the data back into training.  “Once we know where you lie in the skill scale,” Hager says, “we can understand where your potential deficits are, and we can turn that into a training regime so we can now say, look, here are the things that would be most useful for you to work on to improve your surgical technique.”

Perhaps not surprisingly, other use cases involved imaging. 

Hager, for example, described the development of an algorithm intended not to replace radiologists, but rather, to enable them to use their skills most effectively. The approach he described would analyze mammograms, and sort them “extremely reliably” into two categories: clearly normal and everything else. This would “use the machine to replace the drudge work, the over and over again work,” and instead “allow radiologists to focus more at the tip of the pyramid, the place where there’s really high value and [the critical need for] human input.”  As he summarizes, “We should be thinking about augmenting people,” and says the way to do this is “to allow them to focus on the place where people have the most value.”

Pathology offers another promising opportunity for the application of AI, Case Western’s Madabhushi points out. In an approach similar to Hager’s, pathology slides might be pre-filtered by a measure of complexity, with the most difficult cases presented to the pathologist when she is the most alert.

He also cited an Israeli company whose software provided “second opinion” reads, reviewing slides that pathologists had already identified as benign. This (theoretically) minimizes the downside risk, while enabling the identification of lesions that initially escaped human detection. (Of course, the concern would be the Peltzman Effect – the worry that pathologists might become less diligent in their initial reads if they thought a computer was likely to double-check their work.)

Ken Ehlert, chief scientific officer, UnitedHealth Group

More generally, both UnitedHealth’s Ehlert and Microsoft’s Lee express hope that AI could also improve our understanding of biology, and complex biological networks. Ehlert also suggests a useful function for AI could be producing for each patient a “patients like mine” function. The idea is that it would be enormously empowering for physicians if an algorithm could review data from millions of patients, presenting the doctor, in real time, with information about how similar patients fared, and what treatment approaches worked best. (This capability, as I’ve described, is painfully absent today.)  

Lee, meanwhile, points to the opportunity for AI to provide doctors with “an intelligent assistant” function, “ambient clinical intelligence” that could listen to a physician interact with a patient and set up a clinical note for her accordingly. 

From EHR to Evidence (?)

The digitization of the electronic health record would seem to present an enormous opportunity for learning and care improvement. Yet, as I’ve repeatedly and perhaps obsessively discussed (see here, here,  and references therein), delivering on this promise has proved exceptionally difficult.

One issue seems to be a misunderstanding of what an EHR is, and isn’t. Essentially, we tend to think we are directly learning about patients, yet what we’re really learning, Kohane reminds us, “is the behavior of doctors.” He adds, “most events, most of the data items, are created by the doctor. So you’re actually learning from the doctor. You’re not learning from the biology.” We need to recognize the difference between the two, he cautions. 

Kohane draws a contrast with the relative feasibility of using images as a substrate for AI.  While acknowledging potential sources of variability in images (a biopsy of a heterogeneous tumor, for example, may happen to catch an unrepresentative sample), Kohane explains,

“I’m going to be much more confident about image-based metrics than I am about  time series, EHR-based metrics, because I just know how much more variation there is [compared to] the slab of tissue that’s obtained in the OR or the retinal image. I assure you, it’s less than the practice of medicine in different cities, and how current, aggressive or venal different doctors are in different systems. That’s going to make our evaluation process for those algorithms that are very doctor-in-the-loop dependent, quite tricky to evaluate.”

Rubin of the NEJM also points to challenges of relying on EHR data. “As someone who contributes to the charts all the time, I’d say that a lot of them are driven by insurance claims rather than caring for the patient or getting the most complete collection of information on that patient.” 

Adds Rubin, we need to think about “the purpose of the data that we’re going to collect, because if we can anticipate that purpose, we can do a better job of collecting data that fulfills that purpose.”

Yet even with their limitations, EHRs still capture data that would seem to be valuable, and provide the opportunity for insight, albeit not through the traditional, gold-standard mechanism of a typical randomized control trial (RCT), with its distinct, highly specified methods of data collection and analysis. 

This presents a dilemma. On the one hand, as Saria asserts, “our reliance on RCTs alone for evidence generation is dramatically slowing down the rate at which we can learn from our data.” 

Rubin’s response (effectively representing the broader medical establishment) is measured. He agrees both that RCTs are “the gold standard right now,” and that they “have tremendous limitations…right now, you can really only ask one question, and it can take 10 years and $100 million to get the answer to that question.” He notes that data from EHRs — “real world data” — is “fundamentally different, and it’s a work in progress to figure out how to make that rigorous,” adding “we have to figure out how to bring rigor, and how to understand the rigor within trials that are not traditionally designed.” Emerging disciplines, Rubin said, need to think about “what do they consider as rigorous,” and to develop “reasonable standards” that can be used for evaluation.

The challenge of leveraging EHR data for evidence generation was experienced directly by Microsoft’s Lee, in the context of his work with the Mayo Clinic on their effort to evaluate convalescent plasma for COVID-19 therapy. This effort enrolled over 100,000 patients, treated over 71,000, and resulted (famously or infamously, depending on your point of view) in the FDA granting Emergency Use Authorization for this treatment.

Peter Lee, Corporate Vice President,
Microsoft Research & Incubations

As Lee candidly describes it;

“For the vast majority of those 71,000 patients, there was tremendous access to clinical histories in electronic form. And so it was almost a perfect situation in modern era where we ought to be able to take all of that clinical experience in a compressed time frame and extract information about safety and efficacy of that experimental therapy…but the process was extremely difficult. And in fact, ultimately, the data was pretty impoverished. And so the amount of instrumentation that we need, the amount of forethought in clinical practice so that the digital exhaust of what we can learn from that clinical practice really feeds into advancing science and ultimately regulatory approvals. Altogether, this is still, in my mind, absolutely the future, but is just much more difficult and subtle than at least I had realized, even as short as one year ago.”

While the spirit to learn from EHR is willing, the flesh (or at least the requite infrastructure), it seems, remains weak.

Hurdles and Barriers

Perhaps predictably, two impediments to the productive application of AI to medicine emerged from the discussion: data sharing and implementation.

The challenge of data access, long lamented, remains a serious problem – perhaps the most significant problem, according to a clearly exasperated Rudin – in bringing AI to medicine. 

Rudin notes:

“The thing that’s stopping a huge amount of scientific research in health care and AI is lack of data. It’s not an AI question, but if we could solve it, it would give a lot of AI answers…you can’t even reproduce a lot of the scientific papers from a few years ago. Sometimes you e-mail the authors, like the lead author of the paper, and they say, well, I never actually had access to that data in the first place. That was done somewhere else by somebody else. Then you email that person and they don’t have the data. It’s impossible to get it. So how are you going to estimate the effect of drugs? How are you going to, you know, reproduce any scientific study and do a better job of it using machine learning if you don’t have access to the data?”

While acknowledging the “tradeoff with privacy,” Rudin says that “if we cannot figure out ways to make data available for scientists to use, then AI is just going to continue to not be used in hospitals, that’s all I can say.”

Asked if the culture, perhaps, is starting to shift, Rudin is blunt: “No, it’s not.” 

Even COVID-19, it seems, couldn’t motivate the necessary change.

Rudin expected that when the virus hit:

“We would be getting e-mails from everywhere saying, hey, we’ve got a bunch of data on COVID patients, here you go — [a dataset that has] a whole medical record for everybody with the COVID information and their survival and all this stuff. No. There’s a few databases that supposedly are available. But the truth is, they’re not. There’s a lot of barriers to even to get into those databases.”

The gap between the many high-profile, data sharing consortia that have sprung up with great fanfare in response to the pandemic, and the apparent difficultly experienced by a top academic computer scientist trying to use these data, seems disappointing (though I might add: hardly surprising).

Microsoft’s Lee also lamented the challenge of accessing the data needed during the COVID-19 crisis.  Working with hospitals in Seattle and elsewhere, Lee says, “we saw this really completely, vividly.”

As Lee tells it, in the early days of COVID-19, it was “critically important” for hospitals and hospital systems “to understand what patients are being seen, what COVID-19 encounters were taking place, what capacity do we have to treat those patients properly? And that capacity is hospital beds, ICUs [intensive care units], PPE [personal protective equipment], testing and so on. And then how is that capacity being utilized?”

Yet, “despite the incredible digitization over the past decade and fifteen years in all manner of health care operations, what we found was we still had frustrating inability to sort of connect the digital dots here,” Lee says. “We had things like PPE tracked in digital ERP [enterprise resource planning] systems, we had encounters uncoded, but in free form text in EHR systems. And we had very little understanding of utilization.”

Even worse, explains Lee, “in terms of fundamental data interoperability standards, we couldn’t quite connect rapidly the identities of people across these various digital silos.”

While Lee insists he’s “optimistic about the future, because the world, and particularly the US, is moving rapidly to address these interoperability issues,” he emphasizes that the crisis forced many to “confront firsthand…some of the work that we still have to do.”

Beyond the (palpably traumatic) interoperability challenges several speakers highlighted, an additional difficulty apparent in the translation of AI into practice concerns. 

In particular, both Saria and Ehlert emphasize that just because a problem is either “interesting” or “solvable,” and might represent an attractive academic research project, that “doesn’t mean it’s actually interesting to be solved in regular practice and regular life,” as Ehlert put it.  He cited the example of a startup that “had put an enormous amount of energy” into developing a device that would use an automated system to instantly measure your height as you walked through the door. “I didn’t realize we had a height problem in health care,” he quips.

Observes Saria “in academia, we think a lot about publishing papers that show new models and evaluating performance of models. But when you start turning into practice,” you need to start “really thinking through all of the uses cases and thinking about harms versus benefit analysis.”

Related more broadly to implementation, Ehlert also highlighted what may be the most significant challenge of all: alignment.  The issue, he points out, isn’t for-profit vs not-for-profit institutions.  “The reality,” he astutely observes, “is everybody has a stakeholder.” He continues, “a hospital does well when there’s more admissions. A physician does well when they treat more patients. A pharmaceutical does well when they sell more pills. An insurance company does well when they manage the risk better.”

Noting the many different business models these different organizations have, it’s perhaps not surprising that, as Ehlert suggests, “One of the biggest issues I think that we all struggle with is how do we get alignment across those things.”

Perhaps, Ehlert says, we need to “look at our fellow humans” and agree “that our real goal” is for “people to have a better health outcome, and have the highest quality of life for the maximum number of years possible.”  

If we’re all aiming for that shared goal, adds Ehlert hopefully, we should be able to “actually align people” to ensure that the “data is collected the way that it needs to be collected to actually make that happen.”

He’s right, of course. But it’s a big “if” – and a big “should.” 

Note: some quotes have been very lightly edited for clarity.

29
Oct
2020

J&J, AZ Back in the Saddle, Regeneron Moves the Ball Downfield, and a Flurry of Deals

Luke Timmerman, founder & editor, Timmerman Report

America, my old conservative friend from a Mountain West state told me on the phone in April, can’t handle the pandemic.

We’re soft, he said.

Like the characters in the 2008 Pixar film “Wall-E,” he said, we’re sucking on our Slurpees and so drunk on cheap 24/7 entertainment that we can’t even stand on our own two feet anymore.

 

Forget about lockdowns, social distancing, or masks to flatten the curve. Won’t happen, he said. We ought to let individuals do what they want, using their best judgment, like in Sweden, as Rand Paul was arguing at the time.

Wrong, I replied. People in pandemics of the past found a way to curb the spread with disciplined social distancing, hand-washing, and masks. Look at the gutsy healthcare workers and tireless essential workers getting us through these hard times. They were sacrificing so much. We only had to sacrifice a little to wrestle the virus to the ground, breaking the chain of community transmission.

While all of us are stuck at home doing our part, I told my friend, look at the biomedical enterprise running on its toes at Olympic pace. We’ve seen nothing comparable since World War II. Government, academia and industry were setting aside their usual differences, and coming together in common cause in an inspiring way. The cavalry was here, I said, to develop mRNA vaccines, therapeutic neutralizing antibodies, more sensitive and rapid tests for SARS-CoV-2.

To go from a raw DNA sequence of a brand new virus to a vaccine candidate injected in a human arm in two months? My head still spins just thinking about it. Within 12 months of that most basic scientific step, we are likely to have at least one vaccine that can be rolled out at scale. Writing that sentence would have been unthinkable one year ago.

Thinking back on this phone conversation with my old friend in April 2020 – between a couple guys who grew up in the same small town in southwestern Wisconsin and grew far apart ideologically as adults – both of us were about half-right.

We have made some terrible choices as a country. We are living with the consequences. The most salient facts are ones that shouldn’t have to be written this far into the pandemic – 80,000 new cases a day, 1,000 new deaths a day, 40+ states with uncontained community spread, no national plan.

The beauty of our democratic system is that we get a chance every so often to correct our mistakes. We do that with our votes. It won’t bring an immediate end to this nightmare. It will take time to halt the spread, and to mend our tattered social fabric.

This has been the hardest year many of us have ever lived through. But people have encountered more difficult hardships before, on vast scales. We will get back on own two feet. We will come out of this with more resilience, more toughness, and more empathy for our fellow human beings.

Plenty of you in biotech are already thriving in this new environment. I look forward to seeing the biotech industry lead the way, showing the rest of the country what a brighter future can look like.

Vaccines

We started the week with good news on the vaccine front. Both AstraZeneca and Johnson & Johnson, developers of viral vector-based vaccines for COVID-19, were able to resume enrollment of their pivotal trials. AZ’s candidate had been on hold in the US since September, while J&J did a voluntary pause to suss out an adverse event in a trial participant. In both cases, the events were not deemed to be caused by the experimental vaccine – so we can all exhale that these serious big-company contenders are still in the mix.

Treatments

Eli Lilly’s neutralizing antibody for COVID-19 didn’t help hospitalized patients, according to the NIH-sponsored ACTIV-3 trial. As Lilly pointed out in a statement, there are other trials running that look at mild to moderate COVID-19 patients. Many experts expect the drug to be more likely to work in this population.

Regeneron’s antibody cocktail produced another slice of encouraging data. Scientifically, the neutralizing antibodies are hammering down the viral loads within the first 7 days, as you’d want to see. That biological phenomenon matched up nicely with the key clinical observation – a 57 percent reduction in hospital visits through Day 29. The difference between drug and placebo was even more meaningful for patient groups with underlying chronic conditions — cardiovascular, metabolic, lung, liver or kidney disease; or immunocompromised. This is good news, especially because efficacy was seen at the low dose – 2.4 grams – meaning that more patients can be served with existing manufacturing capacity. But Regeneron is still saying it has capacity for about 50,000 doses – obviously not enough to fulfill the fanciful promises of antibodies for everyone that are being issued on the campaign trail.

Financings

Boston-based Scorpion Therapeutics raised a $108 million Series A financing to develop precision cancer drugs. Atlas Venture, Omega Funds and Vida Ventures participated, and serial entrepreneur Gary Glick is the founding CEO. (See TR coverage of Glick’s past ventures at IFM Therapeutics, in December 2019, and April 2019).

Novato, Calif.-based Ultragenyx Pharmaceutical, the developer of drugs for rare diseases, raised $400 million in a stock offering at $90 a share.

Shanghai-based LianBio raised $310 million in a crossover financing to develop new medicines for China and other Asian markets. RA Capital, Venrock Healthcare Capital Partners and Chinese investor CMG-SDIC Capital co-led.

Irving, Tex.-based Caris Life Sciences raised $310 million in a financing composed of $235 million in equity and $75 million in debt. Caris said it plans to apply AI to its database of 215,000 patients with molecular and clinical outcome data to advance its precision medicine programs.

Boston-based Puretech Health filed a 20-F listing which would allow it to be publicly traded both in London, and on the NASDAQ.

Madison, Wis.-based Exact Sciences, the maker of molecular diagnostics for cancer, raised $869 million in a stock offering at $101 a share.

Cambridge, Mass.-based Foghorn Therapeutics, the developer of genetic medicines based on emerging science around the chromatin regulatory system, raised $120 million in an IPO priced at $16 a share.

Boston and Copenhagen-based Galecto, a fibrosis drug developer, raised $85 million in an IPO at $15 a share.

Watertown, Mass.-based SQZ Biotech raised $70.6 million in an IPO priced at $16 a share. The company is developing cell therapies.

Boulder, Colo.-based Biodesix raised $72 million in an IPO priced at $18 a share. The company is working on diagnostics for lung diseases.

San Diego-based Arena Pharmaceuticals spun out a startup, Longboard Pharmaceuticals, with a $56 million Series A financing led by Farallon Capital Management. The new company will focus on developing drugs for CNS disorders that can be targeted via G-protein coupled receptors.

Boston-based Cerevel Therapeutics became a public company through a merger with a Special Purpose Acquisition Company (SPAC) sponsored by Perceptive Advisors. The deal is putting $440 million to work at Cerevel. The neuroscience drug developer is led by Tony Coles.

Deals

Exact Sciences agreed to acquire Cambridge, Mass.-based Thrive Earlier Detection for as much as $2.1 billion in cash and stock. About $1.7 billion is upfront, with the rest in milestones. Thrive Earlier Detection is attempting to develop blood-based tests that can catch multiple types of cancer at an earlier stage, when it’s most treatable with standard oncology treatments. (Read in-depth TR coverage of Thrive Earlier Detection’s $110 million Series A round, led by Third Rock Ventures, from May 2019.)

Bayer agreed to acquire AskBio, a leading AAV-vector based gene therapy company, for $2 billion upfront and another $2 billion in milestones.

South San Francisco-based insitro, the developer of a machine learning platform for drug discovery, secured a five-year collaboration with Bristol-Myers Squibb to work on neurological diseases like amyotrophic lateral sclerosis and frontotemporal dementia. Insitro is getting $50 million upfront. (Listen to CEO Daphne Koller on The Long Run podcast, recorded before this deal was announced.)

Novato, Calif.-based Ultragenyx Pharmaceutical struck a deal with Cambridge, Mass.-based Solid Biosciences to co-develop gene therapies for Duchenne Muscular Dystrophy. Solid is pocketing $40 million cash upfront.

Hayward, Calif.-based Arcus Biosciences formed a partnership with AstraZeneca. The smaller company is bringing its anti-TIGIT antibody to the table, and will pair it with the anti-PD-L1 antibody durvalumab (Imfinzi), for a Phase III trial for non-small cell lung cancer.

Novartis agreed to acquire Cambridge, Mass.-based Vedere Bio for $150 million upfront and $130 million in milestones. Vedere is developing optogenetic therapies that are delivered to the retina intravitreally to restore functional vision

San Diego-based Retrophin agreed to pay $90 million upfront to acquire Orphan Technologies, the developer of a treatment for classical homocystinuria.

Manufacturing and Access

Cambridge, Mass.-based Moderna formed an agreement with Takeda Pharmaceutical and the government of Japan to supply 50 million doses of its mRNA COVID-19 vaccine that’s currently in Phase III development. The mRNA vaccine and therapy company reported it had about $4 billion of cash in the bank on Sept. 30. That’s up from about $1.26 billion on Dec. 31, 2019.

Personnel File

Veteran biotech leaders George Golumbeski and Faheem Hasnain joined Vertex Ventures HC as executive advisors.

Boulder, Colo.-based Inscripta, the genome engineering company, said it hired Sri Kosaraju as its new CEO. He replaces Kevin Ness.

Boston-based Monte Rosa Therapeutics, the developer of targeted protein degrading drugs, hired Min Wang as chief operating officer, Ajim Tamboli as chief financial officer and Jullian Jones as senior vice president, head of business development.

Watertown, Mass.-based SQZ Biotech, a cell therapy company, hired Micah Zajic as chief business officer. Marc Schegerin and Sapna Srivastava also were named to the board of directors.

Houston-based Bellicum Pharmaceuticals cut its staff from 68 to 14 – a 79 percent reduction – after a clinical trial of a CAR-T therapy failed in the first four patients.

Public Health

  • Winter is Coming: Time for a Mask Mandate. WSJ Editorial. Oct. 25. (Scott Gottlieb)
  • Preventing the Spread of SARS-CoV-2 With Masks and Other ‘Low Tech’ Interventions. JAMA. Oct. 26. (Andrea Lerner et al)
  • Effectiveness of Face Masks in Preventing Airborne Transmission of SARS-CoV-2. Msphere. Oct. 29. (Hiroshi Ueki et al)
  • COVID-19 Mitigation Behaviors by Age Group – US April-June 2020. CDC Morbidity and Mortality Weekly Report. Oct. 27. (Helena Hutchins et al)
  • Hospitals Are Reeling Under a 46 Percent Spike in COVID-19 Patients. NYT. Oct. 27. (Giulia McDonnell Nieto del Rio, Simon Romero and Mike Baker)
  • President Trump Issues Executive Order, Setting the Stage for Loyalty Tests for Thousands of Federal Civil Servants. Government Executive. Oct. 22. (Donald Kettl)

Science

  • Identification of Required Host Factors for SARS-CoV-2 Infection in Human Cells. Cell. Oct. 24. (Zharko Daniloski et al)
  • SARS-CoV-2 Neutralizing Antibody LY-CoV555 in Outpatients with COVID-19. NEJM. Oct. 28. (Peter Chen et al)
  • SARS-CoV-2 Orf6 Hijacks Nup98 to Block STAT Nuclear Import and Antagonize Interferon Signaling. PNAS. Oct. 23. (Lisa Miorin et al)

Science Features

  • America Is About to Choose How Bad the Pandemic Will Get. The Atlantic. Oct. 28. (Ed Yong)
  • There’s Only One Chance to Do This Right. FDA Panel Wrestles With COVID-19 Vaccine Issues. Science. Oct. 23. (Jon Cohen)
  • The Very, Very Bad Look of Remdesivir, the First FDA Approved COVID-19 Drug. Science. Oct. 28. (Jon Cohen and Kai Kupferschmidt)
  • CRISPR Gene Editing Can Lead to Big Mistakes in Human Embryos. WSJ. Oct. 29. (Amy Dockser Marcus)

Our Shared Humanity

  • I’m a Black Psychiatrist. I’m Never Prepared for the Emptiness and Grief I Feel When Police Shoot a Black Person. STAT. Oct. 23. (Chase Anderson)

Regulatory Action

Cambridge, Mass.-based Foundation Medicine received FDA approval for a companion diagnostic for larotrectinib (Vitrakvi), the cancer treatment designed for solid tumors with NTRK fusions.

29
Oct
2020

What Happened in Switzerland?

Alex Mayweg, managing director, Versant Ventures

Back in March — during the first wave — I reflected on the COVID-19 situation in Switzerland. This small country, at that time, was managing its outbreak and quickly getting it under control.

This was just as the federal government had begun coordinating a response, which had previously been left to local authorities. As I mentioned back then, while Switzerland is rarely the first to take measures or adopt something new, when the country does, it tends to take action extremely well.

Switzerland never formally and fully locked down, but the population adhered to the social distancing rules. And the results spoke for themselves — the numbers during May and the early summer faded to nearly insignificant digits. Switzerland’s flattened curve became the envy of many European countries.

While many Swiss citizens had cancelled the summer holidays they had planned to beautiful locations all around Europe, most (including me!) regretted doing so when the summer arrived and almost seemed — well — sort of normal. Perhaps a bit too normal.

The youth took the opportunity and enjoyed the warm sunny weather in Switzerland, hanging out with friends on the beautiful rivers, lake shores, and mountain resorts. And who would blame them after such a spring?

The mountain resorts recorded strong occupancy in the summer months. It almost seemed like most of Europe decided that hiking in the Swiss mountains might be a reasonable vacation destination, where social distancing — if needed — might be easier. Outdoor and camping equipment became extremely popular purchases.

And while some social distancing and hygiene measures certainly persisted, very few wore masks (except on public transport), as the federal government also returned the power back to the 26 local authorities – known as cantons. Each canton implemented its own measures — or lack of — which could be well justified as the case numbers remained low.

In August and September the numbers started creeping up. Cases rose particularly among the 20-29 year old age group. The famous R-value crept up and hovered just above 1, but after some weeks of increase, there were also brief periods of decreasing case numbers. Were we controlling the situation due to our heightened sensitivity, education and Swiss-efficiency hand washing?

Schools re-opened and mask wearing was still minimal. That changed when some cantons, such as Basel Stadt and a few others, reintroduced masks in supermarkets and shops but not in all public areas. Surprisingly, some cantons didn’t require masks in most public areas. The number of new cases was steadily increasing, but the curve was still pretty flat.

Since most of the cases were in young people, there was very little added pressure being put on hospitals. But even if corrected by age-group, the hospitalization and mortality rate looked much better now than it did during the initial first wave. Doctors were learning how to better treat patients in the hospital, and a couple of new medicines – dexamethasone and remdesivir – were available for those in need. Things seemed to be relatively under control.

Then the autumn school holiday break arrived. Again, the mountain resorts were packed. In many areas mask wearing was minimal. With cooling temperatures, guests were enjoying their meals or afternoon refreshments indoors or in mountain huts. Northern Italy was also a popular destination as the numbers there were even lower than in some parts of Switzerland — so why not go? You’ll never see Venice with this few tourists!

About two weeks later,  cases shot up sharply. Suddenly, we saw a switch from local clusters of COVID-19 to a nationwide surge of cases. Hospitalizations spiked.

What were the factors driving this new nationwide spread?

  1. Mixing of people to new locations during holiday time, which also allows further mixing between age groups
  2. Colder weather with more people spending time indoors together and beginning of the heating season providing drying air and less circulation indoors.
  3. Patchy COVID-19 measures across different cantons, generally insufficient social distancing and limited use of masks.

Within two weeks, Switzerland went from one of the best in Europe to one of the worst, eclipsing the UK, Spain, the Netherlands and also France in new cases per 100K inhabitants.

About 10 days ago, when the sharp incline of the case curve surpassed many European countries, the federal government once again took back control from the inconsistent local authorities and introduced mask law in most public indoor spaces. Once again, the federal government issued a strong recommendation to work from home.

While this was important, I could not help notice that many other European countries had already taken these measures, or even tougher ones, a while ago.

On Wednesday, Oct. 28, the federal government announced further measures, such as requirement for mask wearing in certain public open-air areas, early closing times for bars and restaurants and further limitations for gatherings of certain group sizes. Rapid Covid-19 tests will also be rolled out.

The federal government stopped short of the anticipated “circuit breaker lock downs” that may still yet be implemented next to help flatten the curve as cases are spreading into higher age groups and hospital occupancy is going up.

The number of deaths, which usually increases a few weeks after a surge in new cases, is unfortunately once again following this sad, predictable pattern.  

It is hard to know if too little is being done too late. The lack of measures and discipline in the summer are certainly an issue, but also understandable given the very low numbers of infections at the time. It’s difficult for people to remain vigilant when the threat isn’t visible.

The next weeks and months look to be a bumpy ride. We must do everything now to get this back to some reasonable control. But once again I have faith in my fellow citizens that with good discipline and by working together (albeit socially distanced!) we can hopefully manage this next wave as well or even better than the last.

My hope is that with our united best efforts over the next months and, at some point next year with the aid of emerging vaccines, new drugs, further refinement of treatment protocols, and perhaps a bit of “earlier than expected herd immunity” we can get on top of this virus.

If we do, we can eventually start the Roaring Twenties of this millennium.

28
Oct
2020

The Battle for the Soul of Biopharma: Peter Kolchinsky on The Long Run

Today’s guest on The Long Run is Peter Kolchinsky.

Peter is the managing partner at RA Capital Management. The Boston-based firm invests in public and private life sciences companies with a total of $6.8 billion under management.

Peter Kolchinsky, managing partner, RA Capital

Peter is a virologist by training at Harvard University. It’s obviously a valuable set of skills to have in a year like this. But he’s been spending more time lately on the future viability of the biopharmaceutical ecosystem itself.

The industry, as everyone ought to know, is in deep trouble. Public anger over high drug prices has been festering for years, with no real solution being offered by either party in Washington DC. Peter has seen the good that industry can do with creating new medicines, as well as the misdeeds that have been committed by those in the insurance industry and in pharmaceuticals.

In January, he wrote about some proposed solutions in his book, “The Great American Drug Deal.” It’s brilliant, unorthodox in its solutions, and easy for a layperson to read – unlike a lot of health policy books.

Peter hasn’t been content to just say his piece in a book – he’s now spearheading a nonprofit called No Patient Left Behind. It seeks to advocate for some of the ideas described in the book, urging biotech executives, leaders of industry trade groups, and members of Congress to get on board. He sometimes seeks to advance his thoughts as an occasional contributing writer for Timmerman Report.

In this conversation, I skipped Peter’s backstory. We talked about the crisis in biopharma and the broken profit incentives that have created this mess. He sees an internal battle within the industry between builders who develop innovative new drugs and other products, versus the landlords who are more interested in rent-seeking behavior to maximize profits of old blockbusters. He calls this a “battle for the soul of the industry” – perhaps adapting a phrase from a certain candidate’s rhetorical framework this election year.

One small note before we start: Peter was joined on this call by Chris Morrison, a veteran biotech journalist now working as an editor at RA Capital. You’ll hear Chris chime in at a couple of points, once to clarify a patent expiration date, and another time to remind Peter of the name of an author he was trying to cite.

Peter is brilliant and fiery in this conversation. Anyone who cares about the future of biotech ought to give this a listen.

Now, please join me and Peter Kolchinsky on The Long Run.

27
Oct
2020

How Many COVID-19 Deaths Could Have Been Avoided? More Than 150,000

Ruth Etzioni, PhD, Full Member, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center

For a numbers person like myself, the SARS-CoV-2 pandemic is supremely frustrating. Even after 8 months, there are so many numbers that we still do not know.  Even though there are thousands of researchers trying to fill in the gaps in our knowledge, the gaps persist.

As the officially recorded US death toll from SARS-CoV2 approached 200,000 last month (it’s now over 224,000), one missing number in particular began to disturb me. While I agreed with the many voices blaming the abysmal national response for the grim death count, I knew that even if we had responded optimally, the number of COVID-related deaths would not have been zero.

But how many deaths might we have expected had we responded quickly with sufficient testing, effective contact tracing and compliance with masking and social distancing?

I couldn’t find any published guesses. So I did it myself (with some help, acknowledged below).

I took as my model the country of Germany. It is frequently cited as having “crushed the curve” in March and April, because of a combination of rigorous contact tracing, quarantining, and near universal compliance with local and national lockdowns. In fact, Germany managed to keep cases and deaths ultralow through the summer.

But COVID-19 is a virus that tends to resist tidy narratives and easy explanations. Like much of Europe, despite its early accomplishments, Germany, too, is coping with a new spike in COVID-19 cases. On Sept. 22, when the official US death toll surpassed 200,000, Germany was still in good shape, recording new cases at levels only slightly higher than its summer nadir.

I asked the following question: If the US had been as successful as Germany in managing the virus, how many deaths would we have expected by September 22? By “as successful as Germany.” I meant, “if the likelihood of COVID-19 death in the US were the same as in Germany.”

It is not enough to simply translate the COVID-19 death rate per capita in Germany into a number of US deaths, because the age and demographic structures of the two countries are quite different. The vast majority of Germans are of European ethnicity. In the US, 18 percent of the population is Hispanic or Latino and 13 percent are African American. And the German population skews slightly older.

To accommodate the demographic differences between the two countries I used age-specific cumulative death rates in Germany from the Robert Koch Institute as my baseline for non-Hispanic whites. But African American and Hispanic/Latino populations in the US have had markedly higher COVID-19 mortality rates than non-Hispanic whites. Even if the US had responded properly to the pandemic, I do not expect that it would also have solved its persistent problem of racial disparities in underlying health, which makes minority populations especially vulnerable to a pandemic.

So, for these minority populations I inflated the Germany-based death rate using race-specific death rate ratios (relative to non-Hispanic whites) from the CDC. Putting this all together, I (and my co-authors) concluded that, had we been as successful as Germany at managing the virus, the US would have had 43,187 deaths rather than the recorded cumulative total of 200,000 on Sept. 22. You can read the abstract and full manuscript here at MedRxiv.

I stared for a while at this shocking result. It was much worse than anticipated. These findings suggested that almost 80% of the 200,000 lives lost – 160,000 people! – could perhaps have been saved by a response that met the gravity of the threat, a response that went all out to break the chain of community spread in order to protect American lives. Even though my result was just an approximation, I felt like it was an important data point. I wanted to publish it right away. But I knew better.

I have seen over and over again how science has been politicized during the pandemic. I wrote in these pages about inappropriate, politically motivated interpretations of the changing IHME COVID-19 model projections, and the appropriation of the Mayo Clinic convalescent plasma study for political expediency. I knew that my result could be used to support a variety of political stories.  Mostly I was concerned that it would be dismissed by Administration supporters because of its limitations, chief among them being that the US is not Germany.

So I tried to make it politicization proof. I went through all of the relevant differences that I could think of between the two countries and explained why my result was still defensible. Yes, there are cultural differences, and greater trust in the government in Germany. Yes, Americans are more likely to be obese. And yes, it is possible and even likely that what counts as a COVID death in Germany is not the same as in the US. I re-classified my work as a “thought experiment designed to provide a first quantification of a best-case scenario in this country.” I still thought it deserved to be published.

Unfortunately, the journals that I submitted it to did not agree.  I quickly received two flat-out rejections from journals that are known for publishing commentaries on the state of public health in this country.

Was the work simply too hot to handle? Were my methods at fault? Were the limitations deal breakers?  Did the editors disagree with my message?

I’ll never know. But at least my science has been validated. This weekend CNN published an article titled: “Faulty US COVID-19 response meant 130,000 to 210,000 avoidable deaths, report finds.”

Finally! I breathlessly followed the link and arrived at a report from a very reputable group of researchers at Columbia University. They had exactly the same idea as I had, but they looked not only at Germany but also at South Korea, Japan, Australia, Canada, and France.

By the time they posted their report, the US death toll stood at 217,000. The report concluded that “had the U.S. government implemented an “averaged” approach that mirrored these countries, the U.S. might have limited fatalities to between 38,000 to 85,000 lives – suggesting that a minimum of 130,000 COVID-19 deaths might have been avoidable given alternate policies, implementation, and leadership.”

In academia, there is a term for when someone publishes your idea before you have a chance to do so. It’s called getting “scooped.” Normally, it’s painful. But this time I am not at all perturbed. I’m delighted that others agree that this information is urgently needed and that they are equally determined to unearth and disseminate it.

We have a number for the human cost of our disastrous national response to COVID-19.

Now we just need to make sure everyone knows it.

Thanks to my co-authors on the article, Ivor S Douglas MD and Elan Markowiz, future PhD.

26
Oct
2020

Machine Learning for Drug Discovery: Daphne Koller on The Long Run

Today’s guest on The Long Run is Daphne Koller.

Daphne is the CEO of South San Francisco-based insitro. The company is seeking to develop a new platform for drug discovery that leans on a combination of wet labs and machine learning algorithms to spot new biological targets for drug discovery.

Daphne Koller, founder and CEO, insitro

Artificial intelligence and machine learning have been stirring imaginations in biopharma, and generating more than a little hype, for a few years now. Smart people who know how complex drug discovery is, are pining for something to make drug discovery less risky, less time-consuming, and less expensive. Insitro represents one of the prominent wagers in the AI for drug discovery category, as it raised a $143 million Series B financing in May led by Andreessen Horowitz, and which included earlier investors like ARCH Venture Partners, Foresite Capital, GV, and Third Rock Ventures.

Daphne comes to this challenge with a fresh set of eyes. She was a math prodigy as a kid. She became a tenured professor of computer science at Stanford University. She became a successful entrepreneur, co-founding Coursera, the online learning platform. But when it came time to find challenging new problems to solve intellectually, and important problems to solve for humanity, she decided that tackling disease was the place to be for her next act.

We talked about her journey, and some of the key aspects of her strategy to make insitro succeed in this new frontier.

Now, please join me and Daphne Koller on The Long Run.

22
Oct
2020

Remdesivir’s FDA Approval, Moderna Fully Enrolls & FDA Wrestles With Trust

Catch up on the main events of the week in biotech with Frontpoints.

Luke Timmerman, founder & editor, Timmerman Report

The FDA issued a surprising approval – not another watered-down Emergency Use Authorization – to Gilead Sciences for remdesivir (Veklury) its antiviral against COVID-19. The antiviral, designed to stop the SARS-CoV-2 virus from copying itself, is now the first treatment fully approved by the FDA against this particularly nefarious virus. Gilead’s approval is for hospitalized patients. The company added that it can manufacture enough of the drug to make it widely available at US hospitals.

The main piece of evidence in favor of approval is the ACTT-1 trial, conducted with the National Institute of Allergy and Infectious Disease, which showed that remdesivir could reduce hospitalization time by a median of five days in the overall patient population, and by seven days in patients who were given supplemental oxygen at baseline.

No rigorous randomized study has yet shown that remdesivir can increase the chances that patients with survive.

With that rather large caveat, I’ll call this a piece of good news.

Not everyone agrees.

Immediately, skeptics pointed to the Solidarity study results from the WHO, which came out last week. That trial didn’t show a survival benefit for remdesivir. But for those who look closely at the protocol and conduct of the study, it looks like a hot mess. Patient subgroups weren’t adequately characterized. WHO researchers weren’t able to answer basic questions, like whether remdesivir worked better for patients with mild disease on low-flow oxygen than for more severely ill patients with overactive immune reactions. Local protocols were all over the place, creating inconsistent data collection across 30 countries. There’s also a curious decision to use 99 percent Confidence Intervals in the meta-analysis (Fig. 4), when it would be standard practice to use 95 percent Confidence Intervals.

Why move to that high of a bar? It’s odd – and to me – a largely inconclusive study that represents frantic, panicked efforts by doctors to do their best in an emergency, but ultimately arriving at a sloppy result at best.

You can see some of the Twitter chatter below, from Eric Topol and Peter Bach, who are expressing skepticism, but really come down in the “inconclusive” camp on remdesivir. It is curious to see the FDA give the green light without more evidence. But under the circumstances, I think there’s good reason to prescribe remdesivir. I know I’d demand it right now if I had a family member hospitalized (while also fighting to get into a clinical trial of a neutralizing antibody from Regeneron, Lilly or VIR).

A Word on Trust

Yesterday, I listened to most of the FDA advisory committee evaluating COVID19 vaccines. The most striking moment of the day came during a presentation by representatives from the Reagan-Udall Foundation for the FDA. They surveyed public opinion on the COVID19 vaccine race. Their findings were sad. So sad, for all of us who care about science as a method for seeking the truth.  

Take a look at their key slides below.

 

Now let that sink in for a second. And as a card-carrying member of the scientific enterprise, ask yourself:

What am I doing today, this month, and this year, to build strong relationships outside my immediate professional circles? What am I doing to foster more confidence in science in my community? What am I doing to push back against the tidal wave of cynicism and apathy in our country?

This isn’t someone else’s problem. This is something we all have to work on.

Vaccines

Moderna said it completed enrollment in the Phase III trial of its COVID19 mRNA vaccine candidate. The company pointed out that minority populations represent 37 percent of those enrolled. CEO Stephane Bancel said the company could receive Emergency Use Authorization as soon as December. The minority enrollment, and demographic breakout on safety and efficacy, will surely factor in the public mind as people think about whether to take any COVID19 vaccine. Especially at a time of rising skepticism, and hesitancy to take vaccines.

Pfizer CEO Albert Bourla, who previously has said it could possibly have a positive readout on its COVID19 vaccine candidate in late October, backed away from that aggressive position this week, posting a letter on the company website saying that the company could apply for an Emergency Use Authorization after a key safety milestone is achieved in the third week of November.

Brief Note on the Pandemic

We are heading now to a horrible third peak. More than 70,000 new cases a day are piling up in the US. We are back to seeing 1,000 deaths a day. More than 223,000 are officially dead from COVID19, and the excess death toll now exceeds 300,000. The trendlines look ominous this fall and winter, even though we have seen some tremendous progress has been made with therapeutics and vaccines.

Our president chooses to blame Tony Fauci “and these idiots” who people are tired of listening to.

If we as a country were willing to follow the science, and course-correct when presented with new data on what works and what doesn’t — like Tony Fauci and “those idiots” — we’d be in a far better position than we are today. (See Timmerman Report July 9 editorial on Standing With Fauci)

Science
  • COVID-19: All the Wrong Moves, in All the Wrong Places. Science. Sept. 15. (Christopher Barrett and Michael Yaffe)
  • COVID-19 Can Affect the Heart. Science. Oct. 23. (Eric Topol)
  • Understanding COVID-19 Vaccine Efficacy. Science. Oct. 21. (Marc Lipsitch and Natalie Dean)
  • The Engines of SARS-CoV-2 Spread. Science. Oct. 23. (Elizabeth Lee et al)
  • Will COVID19 Vaccines Save Lives? Current Trials Aren’t Designed to Tell Us. BMJ. Oct. 21. (Peter Doshi)
  • Therapeutic Activity of an Inhaled Potent SARS-CoV-2 Neutralizing Human Monoclonal Antibody in Hamsters. BioRxiv. Oct. 14. (Michael Piepenbrink et al)
  • A Structural Model of the Endogenous Human BAF Complex Informs Disease Mechanisms. Cell. Oct. 13. (Nazar Mashtalir et al)
  • Structural Analysis of Full-Length SARS-CoV-2 Spike Protein from an Advanced Vaccine Candidate. Science. Oct. 20. (Sandhya Bangaru et al)
  • Unbiased Screens Show CD8+ T cells of COVID19 Patients Recognize Shared Epitopes in SARS-CoV-2, Most of Which Are Not Located in the Spike Protein. Immunity. Oct. 20. (Andrew Ferretti et al)
  • Outcomes of Neonates Born to Mothers With SARS-CoV-2 Infections at a Large Medical Center in New York City. JAMA Pediatrics. Oct. 12. (Dani Dumitriu et al)
Science Features
  • Should COVID Be Left to Spread Among the Young and Healthy? Two Clashing Petitions. The Economist. Oct. 21.
  • Studies Point to a Big Drop in COVID19 Death Rates. NPR Shots. Oct. 20. (Geoff Brumfiel)
  • Cryo-Electron Microscopy Breaks the Atomic Resolution Barrier at Last. Science. Oct. 21. (Robert Service)
Public Health
  • The Pandemic Has Caused Nearly 300,000 More Deaths Than Expected in a Typical Year. Washington Post. Oct. 20. (Lenny Bernstein)
  • Excess Deaths Associated With COVID19 by Age, Race and Ethnicity, United States, Jan. 26-Oct. 3, 2020. CDC Morbidity and Mortality Report. Oct. 20. (Lauren Rossen et al)
Manufacturing and Patient Access
  • Where Do I Go to Get My COVID Antibody Cocktail? WSJ. Oct. 18. (Scott Gottlieb and Mark McClellan)
Trust, or Lack Thereof
  • Supporting US Public Health Experts. Nature Bioengineering. Oct. 22. (Jeremy Levin et al)
  • Eight Persistent COVID19 Myths and Why People Believe Them. Scientific American. Oct. 12. (Tanya Lewis)
RIP

Michael Lane, who the New York Times called “a general in the rout of smallpox,” died at the age of 84. His name isn’t one that kids learn in school, but he was one of the American heroes, along with Bill Foege, who oversaw the Herculean task of wiping that scourge from the face of the Earth. Maybe when we emerge from this terrible pandemic, we’ll get better at recognizing some of these unsung heroes of the scientific enterprise.

Financings

RA Capital Management raised $461 million for a second venture fund to invest in biotech startups.

Menlo Ventures raised its 15th fund. This one has $500 million. Part of the focus will be on “therapeutics, particularly companies built around platform technologies and massive datasets that can produce multiple new treatments, and transformative health technologies, including applications of genomics and synthetic biology,” the firm said.

Cambridge, Mass.-based Be Biopharma raised a $52 million Series A financing led by Atlas Venture and RA Capital Management. It’s developing B-cell medicines, based on research from David Rawlings and Richard James at Seattle Children’s Research Institute. Aleks Radovic-Moreno is the founding president.

UK-based Abcam, a biological reagents and tool marketer, raised $157 million in an IPO at $17.50 a share.

Australia-based Opthea raised $128 million in an IPO at $13.50 a share. The company is developing a VEGF inhibitor for the wet form of age-related macular degeneration.

South San Francisco-based Aligos Therapeutics raised $150 million in an IPO at $15 a share. The company is working on antiviral therapies for chronic hepatitis B and coronaviruses.

Boston-based Replimune, the developer of oncolytic virus treatments, raised $250 million in a stock offering at $40 a share.

Evanston, Ill.-based Aptinyx raised $42 million in a stock offering at $3 a share after releasing some encouraging data for

Houston-based Kiromic Biopharma raised $15 million in an IPO to advance an AI platform for immuno-oncology therapy development.

Cambridge, Mass.-based Praxis Precision Medicines raised $219 million in an IPO at $19 a share. The company is working on CNS drugs.

Irvine, Calif.-based Tarsus Pharmaceuticals raised $88 million in an IPO at $16 a share. It’s working on ophthalmic drugs.

San Francisco-based InCarda Therapeutics raised $30 million in the first close of a Series C financing to develop inhalable treatments for cardiovascular disease. Innoviva led the deal, and existing investors Deerfield Management, HealthCap and Morningside Venture also joined the financing.

Boston-based Allonia, a spinout from Ginkgo Bioworks, raised $40 million in a Series A financing to make enzymes to clean up environmental contaminants.

Sarissa Capital Management raised $175 million in an IPO at $10 per unit. It’s a blank check company.

Israel-based Azura Ophthalmics raised $20 million to develop a treatment for dry eye.

Paris-based SparingVision raised 44 million EUR to develop genomic medicines for ocular diseases.

Cambridge, Mass.-based Compass Therapeutics, a cancer immunotherapy company, filed a prospectus with the SEC to raise up to $50 million in an IPO.

This Week in the Opioid Crisis

Purdue Pharma agreed to plead guilty to three criminal charges for its role in fostering the opioid painkiller addiction crisis. Purdue will reportedly make a $225 million direct payment to the federal government, but it will likely not have to cough up the full $8 billion extent of the settlement, because it is now in bankruptcy. While a guilty plea on three federal counts is pretty straightforward, David Sackler told Vanity Fair “we didn’t cause the crisis.”

Personnel File

Cambridge, Mass.-based Seres Therapeutics, the microbiome-based therapy developer, hired David Ege as chief technology officer and Jayne Gansler as chief people officer.

BD hired Patrick Kaltenbach as chief technology officer. Dave Hickey was promoted to president of the life sciences segment of the company.

Cambridge, Mass-based Nimbus Therapeutics hired Erin Cowhig as chief people officer. She was previously VP of HR at Vertex Pharmaceuticals.

Gaithersburg, MD-based Sensei Biotherapeutics, a developer of off-the-shelf cell therapies for cancer, hired Anu Hoey as chief business officer.

Cambridge, Mass.-based Sage Therapeutics said CEO Jeff Jonas was scheduled to undergo surgery yesterday (Oct. 22), and will essentially be on a medical leave until December. “While Dr. Jonas is recuperating, he will be available as needed, and during this time, the Sage leadership team will direct business operations,” the company said in a statement.

Democrats in the nation’s capital, according to STAT, apparently want to fire Moncef Slaoui from Operation Warp Speed because of his pharmaceutical industry track record and network. Bad idea.

Cambridge, Mass.-based AVROBio added Gail Farfel to its board.

UK-based Vaccitech hired Margaret (Meg) Marshall as chief medical officer.

Flagship Pioneering said Tuyen Ong has joined the venture firm as a CEO-partner. He’ll lead Ring Therapeutics, a gene therapy company.

Kudos

The National Academy of Medicine named 100 new members. Wendy Chung, a geneticist at Columbia University, Levi Garraway, now at Genentech, and David Liu of Harvard University, Marc Lipsitch of Harvard University, Aviv Regev of Genentech, Antoni Ribas of UCLA, Pardis Sabeti of the Broad Institute, and Bob Wachter of UCSF are members of this year’s stellar cast that are well-known in the biopharma community. (Full list).

Peter Byers of the University of Washington won the 2020 Victor McKusick Leadership award from the American Society of Human Genetics.

Data That Mattered

Switzerland and Cambridge, Mass.-based CRISPR Therapeutics provided an update on a Phase I trial of CTX110, as a treatment for CD19-positive B-cell malignancies. The company reported that 2 out of 4 patients on Dose Level 3 achieved Complete Responses at 3 months. A patient on Dose Level 4 developed cytokine release syndrome, appeared to have a Complete Response at Day 25, but then took turn a for the worse and died at day 52.

Cambridge, Mass.-based Alnylam Pharmaceuticals reported on Phase III results from the Illuminate-B study of pediatric patients who got lumasiran for Primary Hyperoxaluria Type 1. The drug showed a 72 percent mean reduction in urinary oxalate. The product is a subcutaneously administered RNAi drug.

Cambridge, Mass.-based Biogen said it has discontinued development of opicinumab, the anti-LINGO antibody for multiple sclerosis.

San Francisco-based VIR Biotechnology presented data on a broadly neutralizing antibody against influenza A which showed it could bind with and potentially neutralize all major strains of the virus from the past 100 years. That was a preclinical finding. A separate Phase I study found the antibody, VIR-2482, could last long enough to be given once per flu season. Findings were presented at IDWeek.

Evanston, Ill.-based Aptinyx reported top-line data from a Phase II study in post-traumatic stress disorder (PTSD). More detail will be presented at a scientific meeting.

Merck reported positive Phase III results from two more studies of its 15-valent pneumococcal vaccine candidate. Merck also reported 3-year follow-up survival data for pembrolizumab (Keytruda) in first-line treatment of non-small cell lung cancer.

Regulatory Action

Roche won full FDA approval for venetoclax (Venclexta) for newly diagnosed acute myeloid leukemia in adults 75 years or older, or who have comorbidities that preclude use of intensive induction chemotherapy. The drug had gotten an accelerated approval in November 2018.

Deals

New York-based Nuvation Bio, a cancer drug developer, and Panacea Acqusition (a SPAC backed by EcoR1 Capital) merged in a deal that will bring $500 million in concurrent equity financings.

San Francisco-based Invitae and Menlo Park, Calif.-based PacBio agreed to a research collaboration to use whole genome sequencing to find clinically relevant molecular targets to develop diagnostic tests for kids with epilepsy.

South San Francisco-based insitro, a company using machine learning to advance drug discovery, agreed to acquire Haystack Sciences. Haystack is working on “large, diverse combinatorial chemical libraries encoded by unique DNA sequences called DNA-encoded libraries, or DELs.” Terms weren’t disclosed.

Genentech agreed to work on an AI for drug discovery partnership with Genesis Therapeutics, a spinout from the Pande Lab at Stanford University.

Roche agreed to work with Boston-based Atea Pharmaceuticals on a direct-acting antiviral treatment against COVID-19. Atea’s AT-527 is being developed as an oral nucleotide prodrug, with potential for hospitalized and non-hospitalized patients. It has shown broad potential against human flaviviruses and coronaviruses. Roche agreed to pay $350 million.

Vifor Pharma secured US commercial rights from Cara Therapeutics to IV Korsuva as a treatment for kidney dialysis patients with pruritis (itching). Cara is getting $100 million upfront.

 

19
Oct
2020

Getting Ready for the Next Pandemic With a Comprehensive Response

Ankit Mahadevia, CEO, Spero Therapeutics

The progress in developing therapies and vaccines for COVID-19 has been extraordinary, and has received extraordinary attention.

But there’s another good news story beginning to emerge, about our widening array of defenses against bacterial infectious disease.

On the clinical front in the past month, developers have announced positive Phase III trials in difficult diseases that have been marked with prior failure, such as C. Difficile gut infections and oral treatment of resistant, complex urinary tract infections (cUTI). As we have noted before in this column, the continued growth and sustainability of the antibacterial field depends on choosing medicines that have a high prevalance of unmet need and sustainable reimbursement in the current system (typically outside of the hospital DRG based payment system).

Those things are necessary to build a base upon which to develop new treatments for emerging diseases.    

For infections (primarily hospital-based) that require reimbursement reform before we can develop economically sustainable treatments, the reintroduction of the PASTEUR Act last month in Congress is a welcome step in planning for our preparedness against future pandemics and infectious threats.

The Act would confer substantial upfront payments and payments over time (totaling hundreds of millions of dollars) to sponsors developing agents to treat current and future high-priority bacterial threats. This could provide the kind of steady, predictable revenue streams for future infectious threats that don’t exist in the market today.

Where do we go from here? The impact of infection, even at the highest levels of government, makes the wait for vaccines and therapeutics excruciating. 

Certainly, a comprehensive, long-term approach to developing medicines for current and future threats before they’re in dire need is critical.  Further, as an important part of the economy and the social infrastructure that supports it, the drug development community must be a driver in this solution as it has been with COVID-19, rather than waiting for someone else or another pandemic to propel us into action.

Each of us in the ecosystem has a part to play to keep driving ahead towards a solution in advance, so we won’t be stuck playing catch-up.

Drug Developers: Continue the focus on right drugs for right patients

Those of us that select and prioritize investment in infectious disease portfolios have the highest responsibility to choose therapeutic needs that meet two related priorities: address a need currently not met by existing medications and have the economic potential to sustain a franchise.

Medicines reimbursed outside of the hospital tend to meet these criteria (see here for much more data behind a premise for a sustainable franchise). 

When a company can generate enough product revenue to achieve scale, it then has the means to plow profits back into additional promising therapies. Cubist Pharmaceuticals (built based on the growth of daptomycin in the outpatient setting) and Pfizer (built in part on the growth of linezolid in the outpatient setting) are models for success.

When building off a stable foundation as in these cases, antibacterial developers can have enough wherewithal to acquire additional revenue-generating products, take advantage of some of the substantial regulatory and market exclusivity incentives that exist in this category, and fulfill our industry’s mission to be a source of cures for the most pressing needs of the moment and in the future. 

Large Pharma: Build on a great start 

The commitment of over $1 billion to the infectious disease sector by a consortium of larger companies, through the Antimicrobial Resistance (AMR) Action Fund, is a great start. 

However, money by itself isn’t enough. Given that drug development is the answer to the challenge nature has posed to us with infectious threats, larger drug developers can continue to be central by directly advancing medicines.

The approach of being reactive to threats as they emerge is suboptimal, as evidenced by today’s pandemic. As with biotech, the first step is building these efforts around applications that have economic potential in the current health care environment and can form the basis for efforts at greater scale.   

Recent investments by Roche and Pfizer, continuing investment in the field by Merck, as well as interest from new parties are positive developments. There’s also an increasing recognition of the opportunity to do well for shareholders and patients by using sustainable products to build a base for a pipeline.

Policy makers: Keep up the good work via BARDA

It’s heartening to see the multiple, well thought-out proposals in legislatures around the world for investing in new anti-infectives.  What’s required now is for all stakeholders to coalesce around the right solutions. No plan will be 100% perfect for all stakeholders, but doing nothing will certainly be imperfect for all of us. 

In the meantime, the Biomedical Advanced Research and Development Authority (BARDA) has risen to the challenge of the COVID-19 response. It has also remained focused on infectious threats beyond COVID-19, marshaling its expertise, mandate, and national coordinating platform.    

All of us

We can choose what we value in the health care system. Ultimately, all actors in our health care system follow the incentives created by reimbursement. Our industry has a vast number of pipeline medicines in play (many around the same targets) in oncology, rare diseases, and inflammation.

That is because our system — either intentionally or unintentionally — puts more financial support behind these endeavors. We have fewer medicines in play for psychiatry, and for infectious threats, for the same reason. 

We have been spoiled by a very, very effective infrastructure against infections over the last 50 years. COVID-19, and the relentless, worrisome march of antimicrobial resistance suggests large holes in that armor.

All of the breathtaking innovation in our sector, including the cancer treatments, procedures, and approaches leveraging our immune system are built on an assumption that we already have a strong armamentarium for infectious disease prevention and treatment. The pandemic has exposed a weakness in our defenses, and opened up a new set of great opportunities for biotech to show what it can do. We’ve already made great progress in this most difficult year.

We encourage all of us in the industry to keep doing our part to prevent infectious threats of the future.

 

Special thanks to Tim Hunt, Aleks Enge of the Novo REPAIR fund, and the Spero Team for their contributions.

15
Oct
2020

J&J Vaccine Study Paused, Lilly Antibody Paused, and Regeneron’s Ebola FDA Approval

Luke Timmerman, founder & editor, Timmerman Report

It’s hard to tell yet whether the J&J vaccine and Eli Lilly antibody trials are suffering from momentary blips, or something more serious on the safety front.

The AstraZeneca vaccine trial has been stuck on pause in the US since September, which seems like a rather lengthy delay in pandemic terms. Let’s hope they all get back in the saddle shortly.

Catch up on the main happenings of the week in biotech, and try not to get blinded by the distortions of election season.

Save yourself some time and keep things in perspective with Frontpoints.

Vaccines

Johnson & Johnson said it put a temporary pause on all dosing with its adenoviral vector-based, single-shot vaccine candidate for COVID-19 after observing an “unexplained illness in a study participant.” The halt of all clinical trials with this vaccine candidate includes the ongoing Phase III Ensemble trial, was done in accordance with company policies, and isn’t a clinical hold imposed by the FDA, the company said. I’ll be interviewing J&J chief scientific officer Paul Stoffels on a panel at BIO-Europe Oct. 26, and will be sure to ask him about the latest on this program (which hopefully will be back enrolling participants by then).

Sanofi and Lexington, Mass.-based Translate Bio said their mRNA vaccine candidate passed preclinical studies in mice and monkeys, and is being readied to enter clinical trials in the fourth quarter of 2020. The mRNA vaccine candidate generated dose-dependent levels of binding antibodies and neutralizing antibodies to the SARS-CoV-2 spike protein. Scientists at the companies also saw a Th1-biased T cell responses against virus in mice and monkeys. The vaccine is being developed in a two-shot regimen, given three weeks apart. (Preprint article here.)

Treatments

Remdesivir, the antiviral developed by Gilead Sciences, didn’t offer a survival benefit to hospitalized patients in the long-awaited Solidarity trial of 11,000 patients, according to a copy of the World Health Organization study first reported by the Financial Times. The drug also had little effect on hospitalization time. The preprint summary came out later in the day on MedRxiv.

AstraZeneca said it has advanced its long-acting antibody therapy combination, AZD7442, for COVID-19 into a pair of Phase III trials that will enroll 6,000 patients. The LAABs were optimised with half-life extension and reduced Fc receptor binding, and they should provide six to 12 months of protection from COVID-19, the company said. The US government is set to spend $486 million to get access to 100,000 doses, and it can acquire another 1 million doses, the company said.

Eli Lilly and Incyte said baricitinib (Olumiant), a JAK1/JAK2 inhibitor for rheumatoid arthritis, was found to shorten the time to recovery from COVID-19 when given in tandem with remdesivir, compared with remdesivir alone. Patients on the combo got out of the hospital in a median time of 7 days, compared with 8 days for those in the control group. The combo appeared marginally better on survival rates than remdesivir alone, but the difference wasn’t statistically significant, Lilly said. The data came from the Adaptive COVID-19 Treatment Trial, sponsored by the National Institute of Allergy and Infectious Disease.

The NIH said it paused a clinical trial of Eli Lilly’s monoclonal antibody for COVID-19, LY-CoV555, after a Data Safety Monitoring Board reported that study had reached a “predefined boundary for safety” after five days of treatment.

Regulatory Action

The FDA cleared Regeneron Pharmaceuticals to market atoltivimab, maftivimab, and odesivimab-ebgn (Inmazeb), a three-antibody mixture, as the first treatment for Ebola virus in adults and children. This landmark therapeutic approval comes 10 months after the agency approved the first Ebola vaccine, designed to prevent infections. The Ebola work at Regeneron in recent years set the stage for it to move with clarity and speed on its double-antibody cocktail for SARS-CoV-2.

Waltham, Mass. and Dublin-based Alkermes won a 16-1 positive recommendation from an FDA advisory committee that evaluated ALKS3831 – its combo of olanzapine/samidorphan – as a once-daily, oral atypical antipsychotic drug candidate for adults with schizophrenia and for bipolar I disorder. The advisory committee also voted 13-3 that the company had adequately characterized the safety profile.

Beckman Coulter received an Emergency Use Authorization from the FDA for an IgM antibody test that that the company said demonstrates 99.9% specificity and 98.3% sensitivity for the spike protein of the SARS-CoV-2 virus. It’s part of a suite of serology tests, including an IgG assay cleared in June.

Dallas-based Livmor won FDA 510k clearance for a wearable device that provides continuous monitoring of pulse rhythms to detect atrial fibrillation.

Cambridge, Mass.-based Voyager Therapeutics said its IND to begin clinical trials of a gene therapy for Huntington’s disease was placed on hold by the FDA because of questions about chemistry, manufacturing and controls (CMC).

Deals

Eli Lilly agreed to acquire Cambridge, Mass.-based Disarm Therapeutics for $135 million upfront, plus $1.2 billion in potential milestone payments. Disarm is in preclinical development with SARM1 inhibitors  for axonal degeneration diseases such as peripheral neuropathy, amyotrophic lateral sclerosis, and multiple sclerosis.

Japan-based Astellas Pharma agreed to acquire Berkeley, Calif.-based iota Biosciences for $127 million to get the rest of the company that it doesn’t already own, plus another $176 million to iota shareholders if certain milestones are met. Iota is developing ultra-small implantable medical devices to advance the field of bioelectronics.

Cambridge, Mass.-based Dyno Therapeutics, the developer of AAV-based vectors for gene therapies, struck a partnership with Roche. Dyno is getting an undisclosed upfront payment, and total deal value with milestones worth $1.8 billion.

Data That Mattered

Pfizer’s CDK4/6 inhibitor for HER2-negative breast cancer, palbociclib (Ibrance) failed in a Phase III clinical trial of patients with early breast cancer who have residual invasive disease after completing neoadjuvant chemotherapy.

Boston-based Vertex Pharmaceuticals shut down a Phase II clinical trial of VX-814 for alpha-1 antitrypsin deficiency after seeing elevated liver enzymes in several patients. Vertex stock plunged 20 percent on the news of the new safety signal. The company has another compound, VX-864, that’s structurally distinct and also in Phase II development for AATD.

Cambridge, Mass.-based Sage Therapeutics reported positive interim results from the Phase III Shoreline study that looked at zuranalone, an oral GABAA receptor-positive allosteric modulator for major depressive disorder. The drug is designed to be given in a two-week course of treatment, and therapy can be resumed on an as-needed basis. About 70 percent of patients in the study only needed one or two treatment courses, the company said. Sage said it expects more data in the first half of 2021.

Tarrytown, NY-based Regeneron Pharmaceuticals and Sanofi said dupilumab (Dupixent) passed a Phase III clinical trial for children ages 6-11 with uncontrolled moderate-to-severe asthma. The antibody, which inhibits the inflammatory cytokines IL-4 and IL-13, reduced severe asthma attacks by 65 percent compared with placebo after one year. Researchers also saw improved lung function within two weeks, which continued through the 52-week study.

Belgium-based Galapagos NV and its partner, Servier, reported that an experimental drug for knee osteoarthritis failed in a 52-week Phase II study of 932 patients.

San Diego-based Gossamer Bio said it missed the primary endpoint in a pair of Phase II studies, with its drug candidate for moderate-to-severe eosinophilic asthma, and for chronic rhinosinusitis.

Science
  • Immunogenicity of Novel mRNA COVID19 Vaccine MRT5500 in Mice and Non-Human Primates. BioRxiv. Oct. 14. (Kirill Kalnan et al)
  • Comparative host-coronavirus protein interaction networks reveal pan-viral disease mechanisms. Science. Oct. 15. (QBI COVID-19 Research Group (QCRG), San Francisco)
  • Genomic Evidence for Reinfection With SARS-CoV-2: A Case Study. The Lancet Infectious Diseases. Oct. 12. (Richard Tillett et al)
  • Heterogeneity in Transmissability and Shedding of SARS-CoV-2 Via Droplets and Aerosols. MedRxiv. Oct. 15. (Paul Chen et al)
  • REGN-COV2 Antibodies Prevent and Treat SARS-CoV-2 Infection in Rhesus Macaques and Hamsters. Science. Oct. 9. (Regeneron team)
  • Repurposed Antiviral Drugs for COVID-19, interim WHO Solidarity Trial Results. MedRxiv. Oct. 15. (WHO Solidarity Trial Consortium)
Science Features
  • Immunity and Re-Infection. In the Pipeline. Oct. 14. (Derek Lowe)
  • First, a Vaccine Approval. Then Chaos and Confusion. NYT. Oct. 12. (Carl Zimmer)
  • The Coronavirus Unveiled. NYT. Oct. 9. (Carl Zimmer)
The Infodemic
  • She Hunts Viral Rumors About Real Viruses. Profile of Heidi Larson, director of the Vaccine Confidence Project. NYT. Oct. 15. (Jenny Anderson)
  • Another Unfounded Study on Origins of Virus Spread Online. NYT. Oct. 13. (Katherine Wu)
Strategy
  • It’s Raining Biotech SPACs. LifeSciVC. Oct. 15. (Bruce Booth)
Building Trust, or Eroding Trust?
Manufacturing
  • Merck, after facing years of pressure from urologists to address the shortage of its live BCG therapy for bladder cancer, said it will build a new facility in North Carolina to increase capacity to meet the increased demand. The company said it will 5-6 years to get the new facility up and running, in part because “each batch takes more than 3 months to make, 30 days of which is waiting for the growth of bacteria used to make the medicine.”
Our Shared Humanity
  • Kids Have Suffered Enough. Let Them Have Halloween. NYT. Oct. 15. (Aaron Carroll)
  • What Strength Really Means When You’re Sick. The Atlantic. Oct. 9. (Ed Yong)
  • An Especially Inspiring Nobel Prize. Timmerman Report. Oct. 13. (Jennifer E. Adair)
Investigative Reporting
  • Inside the Fall of the CDC. How the world’s greatest public health organization was brought to its knees by a virus, the president and the capitulation of its own leaders, causing damage that could last much longer than the coronavirus ProPublica. Oct. 15. (James Bandler, Patricia Callahan, Sebastian Rotella and Kirsten Berg)
  • The Inside Story of How the Coronavirus Task Force Coordinator Undermined the World’s Top Health Agency. Science. Oct. 14. (Charles Piller)
Public Health
  • Scientific Consensus on the COVID-19 Pandemic: We Need to Act Now. The Lancet. Oct. 15. (Nireen Alwan and 30 leading epidemiologist co-authors.)
  • Herd Immunity Strategy Endorsed by White House a ‘Ridiculous’ Way to Stop COVID. It Will Just Kill People, Scientists Say. USA Today. Oct. 14. (Elizabeth Weise)
  • White House Blocked CDC From Requiring Masks on Public Transportation. NYT. Oct. 9. (Sheila Kaplan)
  • Federal Official Threatens Nevada for Halting Rapid Testing in Nursing Homes. Nevada Said Tests Had High False Positive Rate. NYT. Oct. 9. (Katherine Wu)
  • FDA Chief Defends Vaccine Trial Halts as Vital to Public Safety. Bloomberg News. Oct. 14. (Drew Armstrong)
  • An Outbreak of COVID-19 Associated With a Recreational Hockey Game in Florida. CDC Weekly Morbidity and Mortality Report. Oct. 16. (David Atrubin, Michael Wiese, Becky Bohinc)
Politics
  • Why Nature Supports Joe Biden for President. Nature. Oct. 14. (The Editors)
  • On Nov. 3, Vote to End Attacks on Science. Scientific American. Oct. 9. (The Editors)
  • Fauci: I’m Not Going Anywhere. Says He’ll Serve Next 4-Year Term, No Matter Who Wins. NBC News. Oct. 12. (Dareh Gregorian)
  • The White House Embraces Herd Immunity Strategy From Great Barrington Declaration. NYT. Oct. 15. (Coronavirus coverage) (Great Barrington Declaration)
Perspective
This Week in Drug Pricing
  • Biden Hints At What Could Be a Model for Drug Pricing Compromise: The Germany Model. Inside Health Policy. Oct. 15. (John Wilkerson)
This Week in the Opioid Crisis

Johnson & Johnson reported in its third quarter earnings statement that it will pony up another $1 billion as part of the settlement with state Attorneys General for its role in the opioid epidemic. The company’s total liability now stands at $5 billion.

Financings

Canaan Partners raised its 12th fund. This one is worth $800 million, and will be put to work in healthcare and technology companies.

Cambridge, Mass.-based Codiak Biosciences, the exosome therapeutics developer, raised $82.5 million in an IPO priced at $15 a share. (See TR coverage of Codiak’s partnership with Sarepta, June 2020).

Cambridge, Mass.-based Nimbus Therapeutics, the small molecule drug discovery operation that leans heavily on structural biology and computational chemistry, secured a $60 million private financing from RA Capital Management and BVF Partners to continue advancing its Tyk2 inhibitor into Phase II, an HPK1 inhibitor into the clinic, and more preclinical candidates. (See TR coverage of Nimbus / Celgene partnership, Oct. 2017).

San Francisco-based Vineti, the developer of a digital platform to manage distribution of personalized gene and cell therapies, expanded its Series C financing by an additional $33 million, bringing the total to $68 million. Cardinal Health led the deal, and Marc Benioff joined the syndicate. (See TR coverage of Vineti’s initial Series C close, Feb. 2020).

San Diego-based RayzeBio raised $45 million in a Series A financing to develop a new platform for radiopharmaceuticals for cancer. Versant Ventures and venBio co-led, and were joined by Samsara BioCapital. (See TR coverage).

Seattle-based NanoString Technologies, the maker of life sciences tools, raised $230 million in a public offering at $40 a share. (See recent TR article by Nanostring’s Sarah Warren and Jason Reeves about spatial biology research into COVID-19).

San Francisco-based Spruce Biosciences raised $103 million in an IPO priced at $15 a share. The company is working on rare endocrine disorders.

Switzerland-based VectivBio raised $110 million to advance Phase III development and commercialization of apraglutide, a GLP-2 analogue to treat short bowel syndrome.

Dublin-based Priothera raised 30 million EUR to develop oral sphingosine 1 phosphate (S1P) receptor modulators for hematological malignancies. Fountain Healthcare Partners and HealthCap co-led.

UK-based Oxford Nanopore raised $100 million to advance its commercial work, and further R&D, of its nanopore-based DNA/RNA sequencing technology.

Beijing and Cambridge, Mass.-based EdiGene raised about $67 million in a Series B financing to advance its gene editing programs into clinical trials.

Watertown, Mass.-based SQZ Biotechnologies, the developer of cell therapies for cancer and infectious disease, filed an S-1 prospectus with the SEC to raise up to $75 million in an IPO.

San Diego-based Evofem Biosciences, a women’s health company, raised $25 million in convertible debt from Adjuvant Capital.

Branford, CT-based Azitra raised $17 million in a Series B financing to advance a microbiome-based therapy for dermatology indications. Leaps by Bayer led.

Personnel File

Cambridge, Mass.-based Dewpoint Therapeutics hired Ameet Nathwani as CEO. (See TR coverage of Dewpoint and its membraneless organelles for drug discovery, Apr. 2019).

Joe Anderson joined France-based Sofinnova Partners a partner in its crossover fund. He comes from Abingworth.

Tom Graney, formerly of Vertex, joined Belgium and Boston-based Oxurion as CFO. The company is working on treating diabetic macular edema.

Germany-based Merck KGaA said Luciano Rossetti is retiring as global head of R&D for healthcare. He’ll be replaced by Danny Bar-Zohar, who joins the company Nov. 1, and Joern-Peter Halle. Bar-Zohar takes on the development work, while Halle will oversee research.

San Carlos, Calif.-based Nautilus Biotechnology, a proteomics company, hired Nick Nelson as chief business officer.

San Diego-based Effector Therapeutics, the developer of selective translation regulator inhibitors (STRIs) for the treatment of cancer, hired Premal Patel as chief medical officer.

Menlo Park, Calif. and Dallas-based ReCode Therapeutics, working on respiratory diseases, hired Julie Eastland as chief operating and chief financial officer.

15
Oct
2020

Corporate Leaders Face Pressure from Investors, Regulators to Act on Diversity

Karl Simpson, CEO, Liftstream

Over the summer, when George Floyd’s horrific death and the disparities of the pandemic combined to spur a new national reckoning with systemic racial injustice, many CEOs issued statements.

Many insisted they would do more than talk – they’d take action in day-to-day business against structural inequalities.

Those loud pronouncements of action are fading fast. The reverberations of their disquietude will soon give way to challenging questions from the intended recipients of those well-crafted statements opposing racism and promising equality.

While the large multinational companies led the way, many smaller companies joined the momentous wave of formal protest that crashed at the feet of those marching the streets in support of BLM, sweeping them along with hope and motivation because “this time was different.”

As the heat of summer subsides, and the mercury falls, the racial justice issue is still burning red-hot. Activists have lost none of the zeal for change. But what action have we seen?

It is too optimistic to hope for immediate change, especially given the historical intractability of the racial issue and the many other difficulties companies currently face.

I do not wish to serve up pre-packed excuses to CEOs. They need to be pressured to follow through with actions. As each day goes by without change, the impatience has undoubtedly intensified. Many ideas for how to bring greater equality aren’t new, so pulling together an action plan should not take too long, but we must allow time for thoughtful and considered action to be implemented.

Very few people would contest that dialogue is crucial to resolving this social tumult. At some point though, words must give way to deeds, and that time has arrived. In an earlier article I wrote for the Timmerman Report, I described the skepticism that follows corporate pledges on diversity. The words, or sentiment, often heartfelt and well-meaning, count for little unless they are linked to direct action.

The NYC Comptroller, Scott Stringer, set out this challenge to CEOs. At the beginning of July, he sent letters to 67 S&P 100 companies, asking the companies to provide evidence of their respective commitments to racial diversity, consistent with the public statements they had made. Among the 67 companies that received the letters were 10 pharmaceutical and biotechnology companies.

The NYC Comptroller, on behalf of the New York City Employees’ Retirement System, Teachers Retirement System of the City of New York and New York City Board of Education Retirement System, requested that the companies publicly disclose their consolidated EEO-1 data. They argue that without this disclosed data, stakeholders are unable to monitor, assess and benchmark a company’s progress and successful practices to hire, retain and promote black employees, other employees of color, and women. After all, it is data they are already obliged to collect under law.

The letters included the request that the companies disclose the consolidated EEO-1 report with the raw data, not percentages, such that it would provide a clear and trackable data set across 10 categories of employment, including senior management.

Investors who evaluate companies on financial metrics wish for this consistent formatting and full disclosure as to enable useful comparisons between periods, and among peer group companies. In response to this letter, about half of the companies — 34, to be precise — have committed to disclose their consolidated EEO-1 report. Abbvie, Amgen, Biogen, Bristol-Myers Squibb, Gilead Sciences, Medtronic and Pfizer are among those following through with disclosure.

This is an encouraging sign of leadership, which others can follow.

This action is in line with a broader effort by institutional investors to get companies to report on a range of human capital metrics and indicators, including diversity and inclusion. One such example is the Human Capital Management Coalition (http://www.uawtrust.org/hcmc) which filed a rulemaking petition with the Securities and Exchange Commission.

This coalition of institutional investors, with $5.9 trillion under management and co-chaired by the UAW Retiree Medical Trust and CalSTRS, are seeking improved reporting and disclosure by companies on human capital metrics, including diversity and inclusion.

Cambria Allen-Retzlaff, co-chair of the Human Capital Management Coalition and Corporate Governance Director for the UAW Retiree Medical Benefits Trust

“The Coalition has long viewed diversity and inclusion data from portfolio companies as critical to investors’ overall understanding about how well a company is managing its workforce. This is why workforce data – focusing on diversity among senior management – is among the four fundamental metrics we believe every company should report to shareholders. These metrics alongside the number of full-time, part-time, contingent employees, turnover in the workforce and total workforce costs, provide a more accurate view of a company’s human capital management and investment,” said Cambria Allen-Ratzlaff, co-chair of the Human Capital Management Coalition and Corporate Governance Director for the UAW Retiree Medical Benefits Trust. “Decisive data showing financial and performance benefits of a diverse workforce makes sense: Why wouldn’t a company seek talent from the largest pool possible?”  

The regulatory and policy sands are clearly shifting, toward more disclosure on D&I, whether at the board, senior management, or organizational level. Proxy firm ISS (Institutional Shareholder Services) recently reported its 2020 policy survey results, in which they asked if corporate boards should disclose the demographics of the board members including directors’ self-identified race and/or ethnicity.

Nearly three quarters (73%) of investors agreed that companies should do this to the full extent possible.

Despite the winds of change blowing forcefully through the business landscape, and a powerful coalition of backers, this proposal for more transparency has stalled. On Aug. 26, the SEC rejected the opportunity to require companies, under Regulation S-K, to report racial and gender workforce data. The amendment to Regulation S-K does introduce principles-based human capital reporting on issues material to the company, which will not result in widespread and uniform reporting by issuers on D&I.

The Human Capital Management Coalition, along with many other institutional investors, will no doubt continue to push the SEC. More specifically, we can expect investors to press their portfolio companies for improved disclosure, and vote accordingly when they aren’t happy with the results.

Running concurrent with the SECs vote on Regulation S-K amendments, was a legislative process in the State of California where Assembly Bill 979 has been signed into law by Gov. Gavin Newsom. This law amends the Corporations Code and requires corporations to appoint directors from underrepresented groups based on the board size. The statute defines a director from an under-represented community as:

“an individual who self-identifies as black, African American, Hispanic, Latino, Asian, Pacific Islander, Native American, Native Hawaiian or Alaska Native, or who self-identifies as gay, lesbian, bisexual or transgender.”

The new law follows similar legislation on gender which has seen many women corporate directors appointed to the boards of life sciences companies throughout California. Companies will have until between 2021 and 2022 to comply, or face fines ranging from $100,000 to $300,000.

Board composition gets a lot of attention when discussing D&I because there is publicly available information, particularly on gender. This information offers an opportunity for analysis and gives advocates the data on which to base their case. The lack of data on diversity at all corporate levels means the board of directors will continue to be an indicator of an organization’s cultural tone.

While board diversity can only signal a correlation with financial performance, we know that it has specific business implications that show up in a company’s financial results. For example, close to half of women (45%) in biotech will reject a job if there are no women involved in their interview and the employer’s board and management are all males. It is not a massive leap to suggest that the same will be true of prospective candidates from racial minorities when people like them are missing from a company’s hierarchy. These data show a direct effect on recruitment, a material risk for many biotech companies, and value.

Any board of directors should act under the societal context in which the company is active. Diversity and equality are very much part of this current and future context, and the judgments that boards and CEOs take at this time will have long-term implications, possibly long after the directors have all moved on. The time horizon to which a board of directors is looking to formulate strategy has a bearing on material issues, and how highly they are prioritizing diversity. If the board has a long-term view, then it should be conscious of how its own diversity is seeding the firm’s cultural identity, and either enabling or limiting management’s ability to promulgate inclusivity throughout the company. 

But board composition is not only limited to the skills around the table. Investors and proxy firms continue to pay attention to the issue of directors serving on too many boards. The specificity of biotech often demands directors with particular expertise and skills. It is, therefore, not uncommon to see directors serving on multiple company boards, both private and public, calling into question their ability to be effective in the role. Diversifying the candidate pool is seen as an obvious remedy to this over-boarding problem.

The trend of “over-boarding” also concerns institutional investors and proxy firms. Among public issuers, they see evidence of greater over-boarding in biotech, pharma and healthcare than in any other industry.

For example, Glass Lewis recommended against U.S. directors 235 times in 2019 for over-boarding, with over a quarter of these occurring at companies in the pharma, biotech and healthcare sector, double any other sector. While proxy firms are only concerned with directorships for listed companies, there remain significant opportunities in biotech to serve on private company boards. While the demands of board service for a company with a venture investor ownership structure may not prove as onerous as a public company, there are limits to what even the most talented person can manage.

Different conclusions can be drawn from this evidence of over-boarding. One is that biotech and pharma companies still favor appointing highly recognized and well networked directors who they believe to have incomparable experience. Another is that companies are not cognizant of appointing directors who are burdened by competing commitments, or care less about their level of engagement.

Replacing such directors by appointing diverse candidates is part of the solution. While they wait for attrition, companies can also expand their boards and add diversity this way too. But unless companies are adapting their approach for appointing board members, looking further afield and in new places, many diverse candidates will soon become over-boarded themselves.  

As for private companies with venture ownership, they require incentives to do more on ED&I. Perhaps the action by Goldman Sachs to refuse to support any company’s IPO without board diversity, thereby limiting access to capital, will be one such incentive for biotechs.

The most significant current incentive though, is that clear evidence of a company’s diversity and inclusive culture will drive decision making among future employees, and influence employee retention. Companies that fail to hire and retain diverse employees will lose out on key talent, and will be taking on material operating risk.

The board and CEO can provide the first spark needed to light the fire. Their response to ED&I must be multifaceted but should include a series of well-described key measures, such as the adoption of a publicly stated diversity policy, containing a clear recruitment search policy for all board and management appointments.

Companies should expand the number of directors on the board to add diversity; even though the board may reduce in size again as terms expire. Companies should consider their governance structure, including the independence of the Chair of the board.

Research that Liftstream published in 2017 showed a correlation between greater board diversity and a separate CEO/Chair structure. Nomination and Governance committees need to have diverse candidates chairing this committee, or at least as members. Companies nearing IPO, or recently listed, should take advantage of any opportunity to add new independent, diverse directors to the board as investor directors rotate off. And boards and CEOs must look internally at how succession plans and internal promotion processes can be made to equally award black and minority employees. 

There are many ways to act in the short term, and the long term. Let us hope those words begin to transfer to deeds, and that those deeds add up over time to the kind of change our society needs.

13
Oct
2020

An Especially Inspiring Nobel Prize, and a Sign of More Work to Do

Dr. Jennifer E. Adair, PhD, associate professor, Clinical Research Division and Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, and research associate professor, University of Washington.

A historic moment for women in science

Early in the morning of Wednesday, Oct. 7, we heard news that buffered the impact of the chaos and tragedy of this year.

For the first time in history, women almost matched men for Nobel Prize awards across all categories.

One in particular stood out.

Two women shared the award in Chemistry: Dr. Jennifer Doudna of the University of California at Berkeley and Dr. Emmanuelle Charpentier of the Max Planck Institute for the Science of Pathogens in Berlin, Germany for their co-development of a method of genome editing.

Just writing that sentence brings tears to my eyes.

There are a couple reasons why. The 2012 discovery by Doudna and Charpentier, that CRISPR systems derived from bacteria and phages can be repurposed to re-write the DNA of nearly any organism, opened the door for a new era in biology. That discovery has helped fuel my own research in human gene therapy as an early-career faculty member at the Fred Hutchinson Cancer Research Center.

However, the reason that gets me running for the tissue box is that they didn’t have to share this award with a man.

Barbara McClintock in the lab, 1947.

To be clear, this hasn’t happened for a Nobel Prize in Chemistry since 1964. That year, Dr. Dorothy Crowfoot Hodgkin became only the second solo female laureate in history for her application of X-ray crystallography to solve protein structures. She was preceded by the only other solo female laureate, Dr. Marie Skłodowska Curie in 1911 for her discovery of the elements radium and polonium.

The Nobel Prize in Medicine has once gone unshared to a woman, Dr. Barbara McClintock in 1983 for her discovery of mobile genetic elements. No female laureate of the Nobel Prize in Physics has ever been honored alone, in fact all four female laureates in Physics have shared their prizes with two male laureates.

One very, very small step towards equity

Between 1901 and 2020, a total of 931 Nobel laureates have been named. Only 58 (6.2%) are women. Women comprise only 15.9% of Literature laureates, 13.7% of Peace laureates, 5.4% of Medicine laureates, 2.4% of the Memorial Prize in Economic Sciences laureates and 1.9% of Physics laureates.

With this year’s announcement, the prize in Chemistry now boasts an impressive 1 percentage point increase in female laureates from 2.7% in 2019, to 3.8% in 2020 (seven women out of a total 186 Chemistry laureates).

That may not sound like much progress, but there is a long-term trend at work. About half of all the female Nobel laureates — 28 out of the 58 in history — were honored between 2001 and 2020. That sounds like significant progress, but it should be considered against the total number of laureates in the same 20-year time period. There were 232 total laureates counting men, women, and organizations, which means that women have only won 12.1 percent of the prizes in the 21st century. Only 12 of these laureates (5.2%) were awarded for Chemistry, Physics or Medicine.

To put this in perspective, women make up about 42 percent of PhD degree recipients in Science, Technology, Engineering or Mathematics (STEM) fields.

The root of the problem

When Presidential candidate Joe Biden announced he would select a woman as his Vice Presidential running mate in 2020, responses were mixed, even among women. I heard echoes of the age-old argument, “Shouldn’t the best candidate win?”

The problem with this argument is that the meritocracy isn’t based on a level playing field – it is biased towards men.

Of all the evidence I could cite here, consider just a few recent findings. A 2019 study in JAMA showed that women in science receive less research funding than their male colleagues, even when grant scores are similar. Another 2020 study by researchers at Northeastern University found that women are biased against in peer-reviewed journals.

Two other studies objectively illustrate just how deep gender bias in science goes. The first was a randomized, double-blind study published in PNAS in 2012.

In this study, science faculty in chemistry, biology and physics were asked to review resumes for a lab manager position. Resumes were identical and were randomly assigned male- or female-associated names. Male-named applicants were consistently ranked higher than female-named applicants, despite identical merit on paper.

This bias was explored further in a more recent study published in Sex Roles last year. In this study, resumes were again randomly assigned names, but this time both gender and race were examined. The position for consideration was postdoctoral fellow. This study supported the known gender bias especially evident in Physics, and further underscored the intersectionality of race and gender as the most biased against across all sciences – again, in the context of equal merit on paper.

It is sad and unsurprising then that of the 22 Hispanic Nobel Prize recipients, only two are women and neither of whom are included in the six Hispanic laureates honored in Chemistry or Medicine. Only six Nobel laureates are known to have identified as LGBTQ. Exactly zero of the Nobel laureates in Chemistry, Physics or Medicine are Black.

How (white) men can and need to help

The Swedish Academy of Sciences committee intended to grant the 1911 Nobel Prize in Chemistry to both Marie and her husband Dr. Pierre Curie, but Pierre died in an accident in 1906 and the award stipulations prevent posthumous award recipients, granting the historic award to Marie alone. With her default-solo Nobel in Chemistry in 1911 following her husband’s death, Dr. Curie also became the only woman in history to win a Nobel Prize twice. She shared her first Nobel Prize in Physics in 1903 with her husband and her doctoral advisor, Dr. Henri Becquerel. Initially only Pierre and Henri were nominated, but after learning of their nomination they decided to lobby the Nobel Prize committee for Marie to be included.

Dr. Hodgkin’s Nobel Prize nomination was solo from the outset, as she had solved both the crystal structures of penicillin in 1945 and Vitamin B12 in 1955. But her nomination didn’t come until two years after colleague Dr. Max Perutz shared the Nobel Prize in Chemistry with Dr. John Kendrew for solving the protein structure of hemoglobin, in 1959.

Indeed, Perutz has been quoted as saying, “I felt embarrassed when I was awarded the Nobel Prize before Dorothy, whose great discoveries had been made with such fantastic skill and chemical insight and had preceded my own.”

Dr. McClintock received her solo 1983 Noble Prize in Medicine more than 33 years after her discovery of mobile genetic elements. In her autobiography, she gives credit to Dr. C.B. Hutchison for inviting her to take his graduate level genetics course at Cornell University, Dr. Lester W. Sharp for his cytology course, and Dr. Rollins A. Emerson, who paired McClintock with fellow graduate students Marcus M. Rhoades and George W. Beadle, who became a Nobel laureate in 1958.

In these cases, and in all of the shared Nobel Prizes awarded to women, men had to open the door. Not only the men named above, but also the nominators, who must meet a set of stringent criteria which greatly favors men, and the prize committees, which have been made up entirely or predominantly of men throughout history. Today, less than 25% of Nobel Prize committee members are female.

When only one group has a majority seat at the table and owns the building the table sits in, they have the ultimate power to invite anyone else in to sit down. They have to actively push for others who do not look like or relate to them to join, and they need to do this by deconstructing their own perceptions of meritocracy.

Drs. Doudna and Charpentier didn’t just have to exceed peer-reviewed publishing standards and funding norms, they had to commercially advance their methodology and simultaneously become global advocates for ethical application of the technology. The result of this momentous effort is a tool that continues to rewrite not just DNA, but nearly all of the life sciences.

Since nomination and committee determinations for Nobel Prizes are made over the course of a year and sealed for a period of 50 years after the award is made, it will be a very long time before we have insight into the decisions behind this year’s awards. Regardless, it is a long overdue positive message to young female scientists like myself, and scientist-hopefuls everywhere.

There are many, many more exceptional candidates working incredibly hard, making groundbreaking discoveries, and waiting and hoping for their chance to someday get the call from Stockholm. Everyone must realize that to the newcomers, the rooms and tables and decorum are unfamiliar and will likely need to change.

True equity and inclusion requires many, many new steps and challenges beyond opening the door, pulling out the chair and sitting down. In short, everyone needs to get uncomfortable for at least some time. The sooner we start seeing and listening to one another at the same tables, the shorter the discomfort will be for everyone.

Even more important, beyond that discomfort lies an infinite number of new ideas whose benefits to the planet and humanity are exponentially diverse.

 

Dr. Jennifer E. Adair, PhD is an Associate Professor in the Clinical Research Division and Cell and Gene Therapy Program at the Fred Hutchinson Cancer Research Center, and a Research Associate Professor at the University of Washington, both in Seattle. Despite an unexpected pregnancy at 19, and another at 20, Jen completed her Bachelor of Science degree in Chemistry at Youngstown State University while on welfare in 2000.

She then went on to complete her doctoral degree in Genetics and Cell Biology at Washington State University in the laboratory of Dr. Raymond Reeves in 2005, during which she gave birth to her third child. After 2 years as a post-doctoral fellow in the laboratory of Dr. Kenneth Olden, the first African American Director of a National Institute of Health, Jen returned to the Pacific Northwest to start her career in gene therapy at Fred Hutch under Dr. Hans-Peter Kiem.

She started her independent laboratory in 2014 to develop tools and methods for delivering gene therapy to patients who need it, anywhere in the world. That same year she adopted her fourth child. In addition to science and science writing, Jen is an inventor, girls soccer coach and volunteer with Boys & Girls Clubs of America, including an interactive program to introduce children to gene therapy science. She is an advocate for the ethical application of gene therapy and diversity in science. Her research has been honored by TEDx, Geekwire and AAAS to name a few. In 2020, Jen was named the Fleischauer Family Endowed Chair in Gene Therapy Translation. You can learn more about Jen here.

12
Oct
2020

Looking at Cancer From a Different Angle: Pearl Huang on The Long Run

Today’s guest on The Long Run is Pearl Huang.

Pearl is the CEO of Cambridge, Mass.-based Cygnal Therapeutics.

Pearl Huang, CEO, Cygnal Therapeutics

Cygnal is a startup dedicated to developing cancer drugs based on some fairly new understanding of the Peripheral Nervous System.

For years, scientists assumed that the PNS was merely a conduit of the central nervous system. But what if the PNS is not a passive actor in disseminating messages from the brain, but an independent force capable of propagating its own neural signals that help facilitate the growth and spread of cancer? Cygnal represents a wager that exoneural biology may be one of the keys to understanding oncogenesis, and one that’s been long overlooked.

Pearl comes to this work with a wealth of experience. She’s a scientist by training, and has worked her way up through the ranks at the Big Pharma companies Merck, GSK and Roche. In between there, she got her first taste for entrepreneurship as the scientific founder of BeiGene.

Pearl grew up in a small college town of the Upper Midwest, a place much like where I grew up. She has some interesting observations about the remote Upper Peninsula of Michigan, moving on to MIT when there weren’t many women around, and then later in life finding herself comfortable with taking on senior leadership roles. It all sets the scene for what drew her to the unorthodox approach to cancer being explored at Cygnal.

Please join me and Pearl Huang on The Long Run.

8
Oct
2020

A Historic Nobel for CRISPR, Pfizer Sticks Up for FDA, & a CDC Legend Speaks

Luke Timmerman, founder & editor, Timmerman Report

What a time to be alive in biomedicine. Catch up on the main events in Frontpoints.

The Babe Ruth of Public Health Speaks Out

William Foege is in his mid-80s. There’s a genome sciences building at the University of Washington named after him because Bill Gates gave money to build it, and he wanted it named after a scientific hero. Foege has nothing to prove to anyone, and doesn’t seek attention. He’s belongs on the Mount Rushmore of global health for his strategically brilliant and relentless campaign to eradicate smallpox from Earth.

So I snapped straight up and read every word Foege wrote when USA Today discovered and published a private letter Foege wrote to current CDC director Robert Redfield, dated Sept. 23.

Foege empathizes with the embattled CDC director in the opening, but like an angry and baffled father, he chastised Redfield for this monumental failing of American leadership in public health. The only real alternative at this point, Foege wrote, is for Redfield to document the horrifying cascade of White House incompetence and mendacity, and resign to save the CDC, a once-proud and now humiliated agency.

Read the full, scathing letter here.

CDC, Finally, Updates Airborne Guidance

We’ve known since January that SARS-CoV-2 is transmitted through the air. There have been debates over the relative extent of droplets versus aerosols, and how far is far enough for social distancing, but still, but the science on tiny aerosol transmission has been clear for some time. And yet, the CDC, has been unable to effectively communicate this to the public. Remember when Dr. Redfield told a Congressional committee that a mask was an even more effective defense tool than a vaccine, and when the President slapped him down for that act of fact-based insubordination? The latest shameful episode in CDC history came when the agency briefly took down guidance from its website in September that emphasized the risk from airborne transmission of the virus. It took until Oct. 5 – yet another day with more than 40,000 new cases – before the CDC was able to update its guidance on its website about airborne transmission. Look at the terrible case of confusing whiplash this gives the public, neatly captured in WSJ editor Jonathan Rockoff’s tweets.

If citizens had been given the straight scoop, straight from the podium of the CDC starting in January and continuously updated from that point forward, I firmly believe this country would have looked more like Germany today. We would have been able to save tens of thousands of lives. But now, as things are, many thousands more are going to die – still 1,000 a day! This horse should never have gotten so far out of the barn. The body politic should never have been so thoroughly poisoned that we’d up in a cynical abyss where large numbers of citizens refuse to wear masks and engage in common sense social distancing.

All of the elected officials who are responsible, and those who continue to enable this daily rolling national train wreck, must be voted out of office.

Kudos

The Nobel Prizes came out, and were quite illustrative of the golden age of biomedicine. On Monday, the Nobel Prize in Physiology or Medicine went to Harvey Alter at the National Institutes of Health; Michael Houghton, now at the University of Alberta; and Charles Rice, now at Rockefeller University. Their work on the hepatitis C virus laid the groundwork for one of the biggest success stories in biotech history – cures for this chronic, liver-damaging infection. Houghton, by the way, did his pioneering work on HCV while at Chiron, one of the early biotech companies (now part of Novartis).  

Then, on Wednesday, came an even bigger award – the Nobel Prize for Chemistry went to Jennifer Doudna and Emmanuelle Charpentier for their discovery of CRISPR-Cas9 gene editing. This discovery of precise editing tools was revolutionary, and is now setting the stage for a new crop of gene-edited medicines with potential to cure diseases like sickle-cell disease, a new generation of drug discovery platforms to speed up precision medicine development, clever agricultural applications, cheap and fast diagnostics and so much more. The cheap, fast, easy nature of CRISPR editing makes it an accessible lab tool widely distributed in labs around the world. Doudna and Charpentier’s contribution not only changed biomedicine, but this dynamic duo is especially inspiring to women scientists and young girls. That was the bigger story than the long-running patent dispute between the University of California-Berkeley and the Broad Institute. George Church and Eric Lander, a couple of leading lions of science with competing claims to novelty in the early work on CRISPR, both made gracious public comments, rather than resort to the kind of occasionally ugly professional jealousy that often swirls around the Nobels. Church, when asked about Doudna and Charpentier’s prize, told STAT “they made the key discovery.”

Isaac Kohane, a professor of biomedical informatics at Harvard Medical School, was awarded the 2020 Morris F. Collen Award of Excellence from the American College of Medical Informatics.

Regulatory Action

The FDA released guidance for COVID-19 vaccine development, and what the standards are for an Emergency Use Authorization, in advance of the upcoming Oct. 22 advisory committee to review the latest safety and efficacy data on this historic effort. The big news in the document (full version here) was that the FDA is calling for at least two months of safety data (see Appendix II) from participants in trials after getting their second and final dose. That means there’s no way an EUA can be issued before the general election on Nov. 3. The White House had reportedly balked at the guidance document, and held it up with questions for FDA. The White House has done considerable damage to the FDA’s reputation for scientific independence this year, but as the President was ill with COVID-19, the White House ultimately backed off, dropping its objections to the balance the agency has been trying to strike between the need for speed, as well as the need for adequate safety data. Given the state of vaccine hesitancy in the public, which perceives a corner-cutting job in the works for electoral purposes, we can be thankful that cooler heads have prevailed.

Albert Bourla, the CEO of Pfizer, may have had a hand in the White House backing off from its aggressive stance, as he said unequivocally that his company wouldn’t try to undercut the independence of the FDA (even if it’s presumably financially advantageous for the company to get a jump on rivals and start selling its COVID vaccine as fast as possible). See his Tweet below.

One final point about COVID-19 vaccine timelines that has gotten less attention: waiting just a few weeks longer for the trials to play out could be the difference between making a momentous decision on a rather skimpy data set, or making it based on a fully mature dataset rich with information on key subgroups like the elderly, and racial and ethnic groups that have been reluctant to enroll in trials (and who especially need vaccines because of the higher-than-average risk minority groups face.) See this comment I tweeted in the moment from Larry Corey of Fred Hutch, one of the leaders of the Operation Warp Speed effort, who spoke at a Johns Hopkins University and University of Washington symposium on Oct. 6.

Tarrytown, NY-based Regeneron Pharmaceuticals submitted a request Oct. 7 to the FDA for Emergency Use Authorization for its REGN-COV2 two-antibody cocktail that’s designed to neutralize the SARS-CoV-2 virus. This comes after the company issued a press release Sept. 29 with the first meaningful slice of clinical data, showing the antibody could significantly reduce viral loads, especially among patients with high amounts of circulating virus. Data from that rolling clinical program haven’t yet been published in a peer-reviewed journal. The move to seek an EUA for wide distribution to patients came the same day the President extolled the virtues of the therapeutic neutralizing antibody from Regeneron, which he received on a Compassionate Use basis, and which he publicly credited with helping him recover from COVID-19. Whether that’s true, of course, we don’t know.

New York-based Mesoblast received a dreaded Complete Response Letter from the FDA, explaining why it wasn’t willing to approve the company’s application to market a new treatment for pediatric steroid-refractory graft-versus-host-disease in children. This was a surprise, given that the FDA’s Oncologic Drugs Advisory Committee voted 9-1 in favor, saying the available data support the drug’s efficacy. The FDA disagreed, saying the company should run at least one additional randomized study in adults or children.

New York-based Y-mAbs Therapeutics received a Refusal to File letter – meaning the FDA declined to review its application to start marketing omburtamab as a treatment of pediatric patients with CNS/leptomeningeal metastasis from neuroblastoma. The agency said the company needs to provide more detail in its Chemistry Manufacturing and Control (CMC) section, and the Clinical section of the Biologics License Application.

San Carlos, Calif.-based Iovance Biotherapeutics reported that its plans to file a Biologics License Application to the FDA have been delayed, based on a disagreement with the agency over “the required potency assays to fully define its TIL therapy.” The company is developing lifileucel as a Tumor-Infiltrating Lymphocyte therapy for metastatic melanoma.

Deals

Bristol-Myers Squibb agreed to pay $13.1 billion in cash, or $225 a share, to acquire Brisbane, Calif.-based Myokardia, the developer of targeted therapies for cardiovascular diseases. Through the deal, BMS obtains mavacamten, a first-in-class treatment for obstructive hypertrophic cardiomyopathy which has gone through Phase III development, and which is expected to go before the FDA with a New Drug Application in the first quarter of 2021. Third Rock Ventures bet $38 million on this company in a Series A financing in September 2012 (see my coverage at the time on Xconomy.)

Pleasanton, Calif.-based 10X Genomics acquired Boston-based ReadCoor for $350 million, a few weeks after its acquisition of Stockholm-based CartaNA. The two acquisitions represent a strategic move into In Situ analysis. (See TR coverage of ReadCoor in this June 2020 feature on “Complexity in Motion.”)

Vancouver, BC-and Seattle-based Chinook Therapeutics closed its merger with Aduro Biotech and completed a $115 million private placement that leaves it with $275 million in cash to develop precision medicine therapies for kidney diseases.

Palo Alto, Calif.-based BridgeBio Pharma agreed to pay $175 million to acquire the remainder of Eidos Therapeutics which it didn’t already own. Eidos is developing acoramidis, a TTR stabilizer for patients with ATTR cardiomyopathy and polyneuropathy.

Cambridge, Mass.-based AVROBio in-licensed a lentiviral gene therapy program from the University of Manchester, to treat patients with Hunter Syndrome. (See TR coverage of AVROBio, September 2020)

The Netherlands and Belgium-based argenx announced a series of partnerships with Chugai Pharmaceuticals, The Clayton Foundation, and Halozyme Therapeutics to expand its antibody engineering capabilities.

Takeda Pharmaceuticals and Arrowhead Pharmaceuticals agreed to work together on further developing ARO-AAT for Alpha-1 Antitrypsin-Associated Liver Disease. Arrowhead is getting $300 million upfront.

Data That Mattered

Eli Lilly had the best clinical data of the week, hands down. The Indianapolis-based pharma company reported that its neutralizing antibody program for COVID-19, developed in collaboration with AbCellera against the spike protein of SARS-CoV-2, nailed an interim analysis of the BLAZE-1 clinical trial that looked at mild-to-moderate COVID-19 patients. The combo therapy of two antibodies did especially well at taking down viral loads at Day 3, Day 7 and Day 11, and reduced COVID-related hospitalization and ER visits. The company said it will seek Emergency Use Authorization from the FDA based on the result. This is the kind of biological and clinical data alignment that we all want to see. Key slide below.

Amgen had a mixed bag of recent results. The company reported positive top-line results for its KRAS G12C directed small molecule drug candidate, sotorasib, in a Phase II study of 126 patients with KRAS G12C mutated forms of advanced non-small cell lung cancer. A few days later, Amgen reported on a failed Phase III result for omecamtiv mecarbil in patients with heart failure with reduced ejection fraction (HFrEF). Technically, the trial hit the primary composite endpoint, but the benefit on that score was miniscule, and the drug failed to demonstrate an improvement in cardiovascular death, a key secondary endpoint. Results from the GALACTIC-HF study, which enrolled, 8,256 patients in 35 countries, will be presented at the American Heart Association’s Scientific Sessions on Nov. 13.

Gilead Sciences and collaborators from the National Institute of Allergy and Infectious Disease published final results for remdesivir in the New England Journal of Medicine. The full analysis confirms the earlier reports that the antiviral can help patients recover more quickly, and get out of the hospital faster. The drug didn’t show a statistically significant benefit on mortality at Day 29, but a post-hoc analysis showed there was a 72 percent mortality benefit in a subgroup of patients who got low-flow oxygen upon hospital admission.

Science
  • Airborne Transmission of SARS-CoV-2. Science. Oct. 5. (Kimberly Prather et al)
  • Cross Reactive Memory T-cells and Herd Immunity to SARS-CoV-2. Nature Reviews Immunology. Oct. 6. (Marc Lipsitch et al)
  • Antigen-based Testing, But Not Real-Time PCR, Correlates With SARS-CoV-2 Virus Culture. MedRxiv. Oct. 5. (Andrew Pekosz et al)
  • Remdesivir for the Treatment of COVID-19. Final Report. New England Journal of Medicine. Oct. 8. (John Biegel et al)
  • Effect of Hydroxychloroquine in Hospitalized Patients With COVID-19. New England Journal of Medicine. Oct. 8. (The RECOVERY Collaborative Group)
  • Remdesivir Targets a Structurally Analogous Region of the Ebola and SARS-CoV-2 Polymerases. Proceedings of the National Academy of Sciences. Oct. 7. (Michael Lo et al)
Science Features
Public Health
  • Get Serious About the People Putting All of Us at Risk. Let’s Enforce Mask Wearing. NYT. Sept. 29. (Elisabeth Rosenthal)
  • Against COVID-19, Imperfect Measures Do the Most Good. NYT. Oct. 4. (Joshua Schiffer)
Strategy
  • Playing to Win: Scenario Planning for a Binary Readout in Biotech. LifeSciVC. Oct. 8. (Ankit Mahadevia)
Politics
  • Why Nature Needs to Cover Politics More Than Ever Now. Nature Editorial. Oct. 6. (The Editors)
  • Dying in a Leadership Vacuum. New England Journal of Medicine Editorial. Oct. 8. (The Editors)
  • Coronavirus Advisor Scott Atlas Hits Back at Fauci and Others Who Doubt His Advice. “I’m Here Because the Country’s Off the Rails.” Business Insider. Oct. 6. (Ashley Collman)
Worth a Listen
  • Moncef Slaoui, The Man Behind America’s Race for a Coronavirus Vaccine. Sway podcast. Oct. 5. (Kara Swisher)
Personnel File

Merck said that Roger Perlmutter, president of Merck Research Laboratories since 2013, is retiring on Jan. 1, 2021. He will be replaced by Dean Li, the company’s senior vice president of discovery sciences and translational medicine. Li is a former professor of medicine and cardiology at the University of Utah. Perlmutter, an immunologist by training, presided over the breakout success of the cancer immunotherapy pembrolizumab (Keytruda).

Brett Zbar joined General Atlantic as global head of life sciences, as the growth equity firm formally established life sciences as its fifth core investment sector. Zbar was previously a managing director with Foresite Capital. (I wrote about Zbar in 2018 as one of “Nine VCs Who Matter, But You Never Read About.”)

Paul Medeiros joined Cambridge, Mass.-based Q-State Biosciences as president and CEO. The company is developing drugs for diseases associated with electrically excitable cells.

San Rafael, Calif.-based BioMarin Pharmaceutical hired Kevin Eggan as Group Vice President, Head of Research and Early Development. He was formerly a tenured professor in the Department of Stem Cell and Regenerative Biology at Harvard University, the Director of Stem Cell Biology for the Stanley Center for Psychiatric Research at the Broad Institute, and an Institute Member of the Broad Institute of MIT and Harvard.

San Diego-based Expansion Therapeutics named Renato Skerlj as its new CEO. The company is developing small molecule drugs against RNA targets.

Sarah Noonberg was hired as chief medical officer of San Francisco-based Maze Therapeutics. (See TR coverage of the $191 million Series A in this company in February 2019, led by Third Rock Ventures and Arch Venture Partners.)

Rick Bright, the former head of the Biomedical Advanced Research and Development Authority who became a whistleblower on the political interference in the COVID-19 response effort, including the unwarranted push for hydroxychloroquine, resigned. In his amended whistleblower complaint, he said he had only been given one assignment since being transferred to the NIH.

Norwood, Mass.-based Corbus Pharmaceuticals cut its workforce by 54 percent.

Name Change

Seattle Genetics officially changed its name to SeaGen Inc. – which has been its informal name for years. The company is actually based in suburban Bothell, Wash. and isn’t exactly a genetics company, it’s a cancer drug developer. It’s never made a lot of buzz for itself, but it has long been the unquestioned bellwether of the Seattle biotech hub with a $35 billion market valuation.

Financings

San Mateo, Calif. and Cambridge, Mass.-based Kronos Bio, a cancer drug developer led by former Gilead CSO Norbert Bischofberger, raised $250 million in an IPO at $19 a share.

Watertown, Mass.-based C4 Therapeutics, the developer of targeted protein degrading drugs, raised $210 million in an IPO at $19 a share.

Silver Spring, Maryland-based Aziyo Biologics raised $50 million in an IPO at $17 a share.

Durham, NC and Austin, Tex.-based Shattuck Labs, a cancer drug developer, raised $202 million in an IPO at $17 a share.

Exton, Penn.-based Immunome, a company working on memory B-cells to develop antibody therapies, raised $39 million in an IPO at $12 a share.

Cambridge, Mass.-based Oncorus, a viral immunotherapy developer for cancer, raised $87 million in an IPO at $15 a share.

Alameda, Calif.-based Scribe Therapeutics raised $20 million in a Series A financing to develop in vivo CRISPR gene editing medicines. Andreesen Horowitz led. It spun out of the Doudna Lab, Oakes Lab, and the Innovative Genomics Institute. Scribe also announced a Biogen partnership to work on neurodegeneration.

South San Francisco-based Federation Bio got started with a $50 million Series A to develop microbiome therapies. Horizons Ventures led, and was joined by existing investors Venrock and Altitude. Emily Drabant Conley, formerly of 23andMe, was named CEO.

Stamford, Conn.-based Springworks Therapeutics, a cancer drug developer, raised $250 million in a stock offering at $51 a share.

7
Oct
2020

Learning From COVID-19: The Lessons For Real World Data

David Shaywitz

The COVID-19 crisis created an urgent need for healthcare data.

For starters, it was necessary to characterize the spread of the pandemic. Quickly, reports were needed on the capacity of healthcare facilities responsible for care of the severely afflicted. Then there was the urgent need to assess the trajectory, and outcomes, of patients admitted to hospitals. 

The profound difficulty our healthcare system has had responding to each of these needs – despite the often heroic efforts of so many dedicated individuals – has revealed critical gaps in the way healthcare data are gathered, shared, and analyzed.

The challenges of defining the spread of COVID-19 relates in part to existing deficiencies – “We don’t really have a public health infrastructure,” Walmart Health’s Senior Vice President Marcus Osborne explains

The Centers for Disease Control and Prevention also made some high-profile blunders early on – perhaps most prominently, the distribution of a flawed initial test for the virus, which forced the country into catch-up mode from the start, as the New York Times and others have discussed. Also a key factor: the Trump’s Administration’s apparent distrust of, and disdain for, the established experts in the public health community; a representative headline, from Axios: “Trump’s war on public health experts.”

The failure experienced by hospitals in assessing capacity has been persuasively documented by Wall Street Journal reporters Melanie Evans and Alexandra Berzon.

The challenge of understanding collectively what happens to patients once they’ve been admitted to a hospital may be less visible, but remains equally problematic.  We are far better at, or at least more diligent at, determining what a patient should be billed for than determining at the most basic level how they actually fared, both individually and for most categories of patients.

This represents the “feedback gap” I recently described, in the context of a July conference on “Establishing a High-Quality Real-World Data Ecosystem” organized by the Duke-Margolis Center.

Recently, the Margolis Center convened another meeting, focused specifically on “Applying Lessons Learned from RWE [real world evidence] in the Time of COVID-19 to the Future.”  While the individual presenters were uniformly hopeful and optimistic, I emerged from the proceedings with the strong sense that the pandemic has thrown into sharp relief a number of persistent and long-standing challenges.

Those who are interested can watch the conference video on YouTube. 

Several topics caught my attention.

First, related directly to the origin of the feedback gap I previously described, is the idea emphasized by UCSF’s Dr. Laura Esserman. In typical care, “we don’t get outcomes on everybody – that’s a problem with medicine.”  She added, “We should track outcomes on everyone, and isn’t that just real-world data?” 

She continued,

“Our current electronic health records are not organized for quality improvement and you shouldn’t have to go to the IRB to get permission to collect the data that allows you to do your job.“

In other words: how can we improve the care we routinely provide if we’re not routinely, and systematically, determining and analyzing how the patients we’re currently taking care of are doing?

Dr. Robert Califf – legendary cardiology clinical trialist, former Commissioner of the FDA, and now Head of Clinical Policy and Strategy at Verily – highlighted a consequence of this failure: the vast amount of clinical practice is not informed by high-quality evidence. He cited a recent study that reported how few of the cardiology guidelines (just 8.5% of the recommendations in the American College of Cardiology/American Heart Association guidelines) are based on the highest level of evidence (supported by multiple randomized controlled trials – RCTs). Unfortunately, this pattern that doesn’t seem to have changed in the last decade.  

While we have a robust clinical trial enterprise, Califf explained, it isn’t meeting a number of critical needs. In particular, he says, “We are not generating the evidence we need to support the healthcare decisions that patients and their doctors have to make every day.”

Fixing this, he says, will require us to “deal with the fragmentation and misaligned incentives in our system.”  

As I’ve argued, a key “reason the information isn’t tracked is, essentially, no one (besides the patient!) really cares, in the sense of being personally invested in (and accountable for) the outcome.”

The consequence of our failure to collect – and the lack of adequate motivation to routinely collect – the information we need to improve care, even at the level of most individual hospitals, much less the regional and national level, has been felt especially acutely by FDA Deputy Commissioner Dr. Amy Abernathy.  An expert in real-world evidence from her Duke and Flatiron Health days, Abernathy has been seeking to organize the incoming COVID-19 data and analyze it through collaborative efforts such as the COVID-19 Evidence Accelerator (in which I’m a participant).

Reflecting on what she’s learned, Abernethy highlighted what struck me as the observational research version of Mike Tyson’s memorable epigram, “Everyone has a plan until they get punched in the mouth.”

In the case of learning from COVID-19 RWE, there was important methodological lesson to be learned from the challenges of even the seemingly most basic elements, such as defining “time zero,” determining what constitutes a hospital admission, and discerning whether a patient was receiving intensive or critical care. 

Many of these issues were surfaced, Abernethy noted, at the Evidence Accelerator, when participants were encouraged to show their work, and get into the critical “nitty-gritty.”  Some of these challenges were also highlighted by conference participants, including Harvard’s Griffin Weber.

Abernethy also pointed out that we’ve become relatively proficient at understanding at a glance what to look for in a high quality RCT, assessing attributes like adequate statistical power and how the blinding was managed.  Now, she said, we need to develop this intuitive understanding for observational studies as well. 

Abernethy also emphasized the need to refine our conception of RWE.  We often tend to view RWE-driven studies as a “replacement product” for RCTs – but this framing may be misleading and distracting.  Everyone would like to have robust RCTs to answer every question, she said, but that’s not possible, and we need RWE “to fill in the gaps.”  

RWE, she emphasized, can be used for a range of purposes, such as understanding patterns of care, or deciding which RCTs should be conducted.

This is a critically important idea: the value of RWE is not as a substitute for RCTs, but rather to more effectively capture the totality of data in the healthcare system, and to provide information about healthcare as it’s actually practiced, within the acknowledged messiness of routine care, as I’ve discussed.

I was also struck by Abernethy’s focus on the importance of high-quality datasets, which would seem to be the cornerstone of meaningful analytics. Abernethy highlighted the problem of data gaps, and the need to link data sets and fill in missing data using different data sources, in effort to approach a level of “completeness” that would enable meaningful study. She noted that technology might be helpful here, in the form of “synthetic controls” (statistically generated comparators based existing data; a nice explainer from Jen Goldsack here) and the use of tokenization (an approach to de-identification of data permitting it to be shared; a useful white paper from Datavant, a leading startup in this space, here).

Abernethy also offered what I thought was spot-on advice regarding the development and application of technology, and some important advice about how healthcare could more effectively engage with technologists and tech companies.

There’s a pervasive problem, she said, with “vendor-think” – the idea that the healthcare stakeholder (hospital, payor, biopharmaceutical company, health services researcher) specifies what a vendor needs to provide, and then the vendor “builds against that list.” 

She described with perfect clarity not just how many large healthcare organizations typically approach large projects, but also the mindset within healthcare organizations that I’ve witnessed and described, where data experts and statisticians are often treated as second-class citizens.

What’s needed, she persuasively argued, is for authentic collaboration, where you have at the same table not just the manager or executive, say, who’s sponsoring the project, but also healthcare domain experts, who understand the subtleties and context of how the data were generated, as well as the technologists – the data scientists and engineers who can build and refine the solution. 

Such ongoing collaboration not only ensures a better mutual understanding of needs, but also enables the work to proceed iteratively, and to evolve as the participants refine their understanding of both the problem to be solved and the solutions that can be envisioned.

Achieving this balance is notoriously difficult, and vanishingly rare to see in practice.  This barrier – a hurdle in organizational dynamics as much as technological expertise – also represents an exceptional opportunity for an integrative and empathetic leader who can not only bring the right people to the table, but (and this is the hard part) ensure their talents are fully elicited and authentically embraced.

7
Oct
2020

Seeing COVID-19 in Context: Applying Spatial Biology to the Lungs

Sarah Warren, senior director, translational science, NanoString Technologies

It’s worth looking back on what is known about the SARS-CoV-2 virus, which was first detected in the US in mid-January, when a man traveled from Wuhan, China to Seattle.

Nearly 10 months later, a tremendous amount of knowledge has been gathered about the virus, how it is transmitted, and the disease symptoms it causes.

We know, for instance, the risk factors that are associated with severity of infection, such as being elderly, or having certain pre-existing conditions like Type 2 diabetes. We are beginning to understand all the ways the virus can be transmitted, particularly through the air. And yet, much remains unknown about the virus, particularly its impact on the body at the molecular level.

Jason Reeves, senior scientist, NanoString Technologies

In our quest to gain a deeper understanding of the molecular and cellular pathology of the disease, investigators have been leveraging every tool and platform available. Applying spatial biology – a strategy for profiling the distribution of cells and molecular pathways within tissues – to samples collected from patients who died from the disease, represents a powerful way to understand at a local level how the virus is reshaping infected tissue and the subsequent immune response.

Our company, Seattle-based NanoString Technologies, recently hosted a virtual Advancing Spatial Biology Conference to feature the work of investigators who are using the company’s GeoMx Digital Spatial Profiler to address a variety of research questions. One of the tracks was dedicated to studies that addressed the impacts of COVID-19 in the lungs. Although the data are still early, the collective results presented by these investigators reveal insights into the underlying biology of the virus and will guide development of therapeutics, supportive care, and vaccines.

Some of the major findings from these studies are summarized below.     

Infected Tissues Are Heterogeneous

The GeoMx technology enables simultaneous profiling of 60-90 proteins, and up to 1800 RNA transcripts (today), from multiple regions of up to half a millimeter in diameter within a tissue section with a profiling area of 14×36 mm.

Capturing this much data, in spatial context from a tissue sample, enables the use of data analysis methodologies suitable for high-plex data to create a deep understanding of how the infection manifests in different patients.

What we see again and again is that the disease varies greatly between individuals. Several investigators during the track pointed out that lung tissue sections from COVID-19 patients were more variable than lung tissue sections from patients that died from other causes. Chris Mason and colleagues at Weill Cornell Medicine in New York were able to compare COVID-19 lung tissues to flu-infected lung tissues, to underscore the wider range of possibilities.

What might be driving this? Partly, the diversity is driven by different courses of the disease that are observed for each patient. Some people succumb rapidly to infection and pass away shortly after diagnosis and presentation in a hospital. These people tend to have evidence of active viral replication and strong interferon response.

Other people have a longer course of disease. They may survive the initial diagnosis but then die from secondary effects of the infection such as inflammatory cytokine storms or other dysregulated immune responses. The lung tissue from these patients tends to be characterized by more tissue repair signaling pathways, perhaps as the lungs attempt to heal themselves.

However, this dichotomization is not universal, so there must be other as yet unidentified causes of the diversity. Viral load may be one contributing factor. Intriguing data presented by Gordon Jiang from Beth Israel Deaconess presented showed that the levels of 5-lipoxygenase, an enzyme involved in the production of inflammatory leukotrienes, is expressed in proportion to viral load.

Despite this inter-patient variability, there were some common patterns of expression, especially in the immune response. Certain innate immune cells, such as macrophages and neutrophils, are activated by the virus and robustly recruited to the sites of viral infection early in the course of disease. The type I interferon response, part of the body’s first line of defense against viral infection, is consistently one of the strongest upregulated pathways.

This suggests that therapeutics targeting these pathways and currently in clinical trials, such as baricitinib, the JAK1/2 inhibitor developed by Lilly that blocks production of interferons, may be effective at tamping down runaway inflammation early in infection, if given in combination with antivirals to control viral load, such as remdesivir, or therapeutic neutralizing antibodies in development. Intriguingly, data presented by the keynote speaker, Dr. Peter Sorger from Harvard Medical School, suggested that similar pathways may also be upregulated in non-human primates infected with COVID-19.

The Virus Affects Tissue Architecture and Immune Response

It is also abundantly evident that the virus is impacting both the immune response and altering the underlying tissue architecture. Regions of the lung with high viral load are characterized by hyaline membranes, sheets of dead cells, surfactant, and proteins that are associated with acute respiratory distress.

These hyaline membranes can be visualized on tissue sections and are accompanied by hyperplasia (excessive growth) of type II alveolar cells. This is a key cell population in the lung that is a progenitor for the gas-exchanging type I alveolar cells. These cells maintain the lubricating surfactants, and recruit immune cells following injury, which are necessary to maintaining normal breathing ability.

Type II alveolar cells are recognized as a reservoir of COVID-19, where the virus hides out, allowing the infection to progress into the lower lungs. These type II alveolar cells provide a fertile breeding ground for the virus, as they express the ACE2 protein that acts as a viral receptor. Spatial profiling of the lungs revealed enrichment of collagen synthesis pathways and extracellular remodeling pathways in the Cornell cohort. Building on that finding, Asa Segerstolpe of the Broad Institute presented evidence of increased keratin expression in infected regions of the lung.  

As previously mentioned, the immune response is dominated by interferon signaling and associated chemokine expression. This was accompanied by robust recruitment of myeloid cells, including monocytes, macrophages, and dendritic cells, to infected regions of the lung, whereas T cells and NK cells were more abundant in patients with high viral load but displayed less specificity for highly infected regions of the lung. In these studies, cell population abundance was inferred through gene expression deconvolution strategies to enable profiling of 14 different immune cell populations. Patients with high viral load also had expression of some immune checkpoints, including PD-L1 and IDO1, but mixed expression of other checkpoints including CTLA4, LAG3, and VISTA.

Impact of Spatial Profiling

One of the most interesting observations from the COVID-19 studies has been the diversity of the localization of immune responses to infection. David Ting, from Massachusetts General Hospital, demonstrated that in addition to variation between patients who have high and low viral titers, there is also variation between regions of interest with high viral load and those without, but only for some signaling pathways.

For example, chemokines CXCL9, CXCL10, and IDO1, which recruit and regulate immune cells, are expressed at higher levels only in areas of the lung with detectable viral transcripts from patients with high viral load. In contrast, antiviral proteins IFITM1, IFIT3, and IFI6, are expressed at approximately equal levels in virus-high vs virus-low areas of the lung. Antigen presenting genes are displayed uniformly throughout the lung regardless of viral load. These observations are only possible with spatial biology tools that enable simultaneous profiling of a large number of targets in parallel.

As these studies and other mature, we are starting to better understand some of the mechanisms at play in COVID-19 infection. However, given the rarity of these samples and the not insignificant risk physicians must undertake to collect them, we must try to learn as much as possible from each sample we have.

Spatial biology represents a powerful tool to directly examine the infected tissue of COVID-19 patients and characterize its inherent complexity, rather than risk getting overly focused on a single variable that’s just one of many factors at work. By applying tools such as GeoMx and methods such as spatial profiling, we can deepen our understanding of the disease and accelerate the development of treatments and vaccines to mitigate the growing burden of this pandemic.

1
Oct
2020

FDA Commissioners Speak Out, An Antibiotic Incentive Proposal and a Drug Price Grilling

Luke Timmerman, founder & editor, Timmerman Report

Rummaging around in the garage can be about more than just tossing out junk.

Going through old files lately, I found a printout from the Carnegie-Knight Task Force in 1997. That was the year I graduated from college and went to work as a local newspaper reporter.

The task force, convened by the Project for Excellence in Journalism, was wrestling with the advent of 24-hour cable news. Internet news was just getting started. Many journalism old-timers worried about an erosion of traditional standards across platforms, and unholy merger of news and entertainment. The O.J. Simpson trial was fresh in people’s minds.

A group of prominent journalists attempted to hash out what they called a Statement of Shared Purpose.

The first bullet point in a 9-point theory of journalism said:

Journalism’s first obligation is to the truth

Democracy depends on citizens having reliable, accurate facts put in a meaningful context. Journalism does not pursue truth in an absolute or philosophical sense, but it can — and must — pursue it in a practical sense. This “journalistic truth” is a process that begins with the professional discipline of assembling and verifying facts. Then journalists try to convey a fair and reliable account of their meaning, valid for now, subject to further investigation. Journalists should be as transparent as possible about sources and methods so audiences can make their own assessment of the information. Even in a world of expanding voices, accuracy is the foundation upon which everything else is built — context, interpretation, comment, criticism, analysis and debate. The truth, over time, emerges from this forum. As citizens encounter an ever greater flow of data, they have more need — not less — for identifiable sources dedicated to verifying that information and putting it in context.

Re-reading this statement now, I think it’s possible to swap in the word “science” where you see “journalism.” These are very different professions, but both are important means people have used to navigate the world since the Enlightenment.  

Science has held up much better than journalism in the past 20 years. In many ways, we are living in a biology Renaissance brimming with new possibility for the treatment and prevention of disease.

Science, of course, can’t give us all the answers with crystal clarity. It isn’t perfect, and neither are the people who work in the scientific enterprise. But science is the best available means we have for asking and answering some of the most vital questions of the day.

There’s a whole lot more heat than light being emitted into our information commons at the moment, but science is the way back onto a clear path. All of us should defend it.

Now, make sure you didn’t miss anything big in biotech this week. Read Frontpoints.

Trust

When seven former FDA commissioners get together to write an op-ed in the Washington Post under the headline “The Trump Administration is Undermining the Credibility of the FDA” — you know you are in uncharted territory. The authors of this piece span 30 years of leadership at the FDA, and they served in Democratic and Republican administrations. They have battle scars from political interests who tried to tip scientific decision-making one way or another. If you read one thing this week, I’d suggest it be this piece by Robert Califf, Scott Gottlieb, Margaret Hamburg, Jane Henney, David Kessler, Mark McClellan and Andy von Eschenbach.

Problem-Solving

Antibiotic development has languished for way too long. How can we get ahead of the curve, and incentivize more antibiotic development now so that we’re better prepared for the next infectious disease calamity? This is not a new issue. It’s a bipartisan issue. It is appropriately being treated as such by US Sens. Michael Bennet of Colorado and Todd Young of Indiana. Bennet is a Democrat and Young is a Republican. These guys will not get cable TV news air time for confronting this serious issue. But they introduced a creative bill this week to set up a subscription model to provide predictable revenue streams for antibiotic developers, along with some provisions for appropriate use of the drugs they develop. It’s just a bill at this point and needs debate. But this is the kind of thing an intelligent, forward-thinking country populated by active citizens ought to debate. See the full bill language here, and a one-page summary here.

Data That Mattered

Tarrytown, NY-based Regeneron Pharmaceuticals released an interim cut of data from 275 patients who enrolled in a rolling clinical trial its therapeutic neutralizing double-antibody combo treatment for COVID-19. REGN-COV2 reduced viral loads and cut down on the time it took to recover from symptoms in non-hospitalized patients. Cutting down viral loads is a good early sign, especially with a clean safety profile. (For background, see Q&A With Regeneron SVP of global clinical development, David Weinreich, TR, June 29, 2020).

Cambridge, Mass.-based Ironwood Pharmaceuticals said it failed in a Phase III clinical trial with IW-3718, an experimental drug for gastroesophageal reflux disease (GERD). The company shut down further clinical development of the drug, and said it plans to cut 100 jobs, or about 35 percent of its workforce.

Cambridge, Mass.-based Alnylam Pharmaceuticals, the RNA interference drug developer, said its experimental treatment lumasiran passed the 18-patient, Phase III Illuminate-B study for primary hyperoxaluria Type 1 in children under the age of six. The results were consistent with the Illuminate-A study that looked at patients ages 6 and older. Alnylam said full results will be presented at a virtual meeting Oct. 22.

Vaccines

Moderna CEO Stephane Bancel said at a Financial Times conference that the company’s mRNA vaccine candidate for COVID-19 won’t be ready for widespread distribution until spring 2021. That should be no surprise to anyone paying close attention to the biotech sector, but it does contradict recent statements by the President.

AstraZeneca’s COVID-19 vaccine candidate has been on clinical hold in the US since Sept. 6, and the FDA has now widened its inquiry into the safety data to date, according to Reuters. The trial was paused after a report of a vaccine subject coming down with transverse myelitis, an immune disorder.

Pfizer CEO Albert Bourla has attracted a ton of scrutiny, rightfully, since he has made repeated claims that the company — in a best-case scenario — might have data as soon as October that says its Phase III COVID-19 vaccine study is a success, and worthy of an Emergency Use Authorization application to the FDA. The caveats and qualifiers have largely been stripped away in our red-hot election season. When the President said at this week’s debate that we are only “weeks away from a vaccine,” Bourla felt compelled to write to Pfizer employees that he was “disappointed” in how vaccines were discussed in political terms. In the memo, posted on LinkedIn Thursday afternoon after reporting by Politico and others, Bourla wrote “we are approaching our goal and despite not having any political considerations with our pre-announced date, we find ourselves in the crucible of the U.S. Presidential election.” He added: “We would never succumb to political pressure.”

Testing
  • Trump Announces Plan To Ship 150 Million Rapid Antigen Tests for COVID-19 for States, Tribes and Territories. NYT. Sept. 28. (Katherine Wu)
  • Rethinking COVID-19 Test Sensitivity. A Strategy for Containment. NEJM. Sept. 30. (Michael Mina et al)
Science Features
  • Cycle Threshold Could Help Reveal How Infectious a COVID-19 Patient Is. Should Test Results Include It? Science. Sept. 29. (Robert Service)
  • Alexa, Do I Have COVID-19? Nature. Sept. 30. (Emily Anthes)
  • Why We Need to Keep Using the Term Long COVID. Oct. 1. (The BMJ Opinion)
  • Science and Scientists Highly Regarded Across the Globe, Pew Survey Says. Oct. 1. (Agence France Presse)
  • HHMI Is the Second Major Funder to Mandate The Research It Pays For Be Published Openly. Nature. Oct. 1. (Holly Else)
Public Health
  • Public Health, Pandemic Response and the 2020 Election. The Lancet Public Health. Oct. 1. (Esther Choo and Aaron Carroll)
  • This Overlooked Variable is Key to the Pandemic. It’s Not R. The Atlantic. Sept. 30. (Zeynep Tufekci)
  • Trump Allies Say the Virus Has Nearly Run Its Course. ‘Nonsense,’ Experts Say. NYT. Sept. 29. (Donald G. McNeil Jr.)
Our Shared Humanity
  • Timothy Ray Brown, First Person Cured of HIV, Dies of Cancer. Associated Press. Sept. 30. (Marilynn Marchione)
Financings

Pfizer agreed to make a $200 million equity investment in Suzhou, China-based CStone Pharmaceuticals. The companies are working together on a PD-L1 antibody for use in mainland China.

China-based InventisBio, a cancer drug developer, raised $147 million in a Series B financing led by Hillhouse affiliate GL Ventures.

Berkeley, Calif.-based Carmot Therapeutics raised $47 million in a Series C financing to advance its incretin receptor modulators through Phase II studies. Amgen joined The Column Group and other existing investors.

Switzerland-based Sophia Genetics raised $110 million in a Series F financing to scale up to meet global demand from clinical and biopharma customers for its “data-driven medicine” offerings. aMoon and Hitachi Ventures led.

New York-based Koneksa raised $16 million in a Series B financing to scale up its work on digital biomarkers for drug development. Spring Mountain Capital led, and was joined by new investors McKesson Ventures, Novartis Pharma and MBX Capital.

Miami Beach-based Vesper Healthcare Acquisition raised $400 million in an IPO. It’s a blank check company intended for M&A. Brent Saunders, the former CEO of Allergan, is the CEO.

Sarissa Capital Management is forming a Specialty Acquisition Company (SPAC) or “blank check” company, seeking to raise $200 million. Sarissa CEO Alex Denner and others at the firm will “ target businesses with valuations of $500 million to $1 billion that have the potential to be substantially greater over time due to their underlying business characteristics and growth opportunities as a public company,” according to the S-1.

New Haven, Conn.-based Biohaven Pharmaceuticals raised $60 million to set up an Asia-Pacific subsidiary.

XtalPi, an AI for drug discovery company, raised $319 million in a Series C deal led by Softbank Vision Fund.

South San Francisco and Seattle-based Sonoma Biotherapeutics expanded its Series A financing to $70 million. The company is working on T-reg cell therapies for autoimmune diseases. Investors included Lyell Immunopharma, Arch Venture Partners, 8VC, LifeForce Capital, and Lilly Asia Ventures Biosciences. (See TR coverage, including interview with CEO Jeff Bluestone, Feb. 6. 2020)

Cambridge, Mass.-based Vedanta Biosciences secured $7.4 million upfront, and potentially another $69.5 million, from BARDA to further develop its experimental defined bacterial consortium therapy for C. difficile infections.

Kudos

Frank Bennett, the longtime chief scientific officer of Carlsbad, Calif.-based Ionis Pharmaceuticals, the antisense drug developer, won the Lifetime Achievement Award from the Oligonucleotide Therapeutics Society.

Personnel File
  • Germany-based Merck KGaA said Belen Garijo will be its new CEO, taking over from Stefan Oschmann. She will start in the top job in May 2021.
  • South San Francisco-based insitro, a company using machine learning to facilitate drug discovery, added Roger Perlmutter, president of Merck Research Laboratories, to its board of directors.
  • Lexington, Mass.-based Kaleido Biosciences named Daniel Manichella its new CEO, replacing Alison Lawton.
  • Boston-based Akouos, a gene therapy for ear diseases company, hired Sachiyo Minegishi as chief financial officer, and promoted Jennifer Wellman from SVP of regulatory and quality to COO.
  • Research Triangle Park, NC-based G1 Therapeutics, a cancer drug developer, named Jack Bailey as its new CEO. He will replace Mark Velleca.
  • Cambridge, Mass.-based Rubius Therapeutics hired Jose “Pepe” Carmona as chief financial officer.
  • Cambridge, Mass.-based Nimbus Therapeutics promoted Abbas Kazimi to chief business officer. He previously had the title of VP of business development.
  • Cambridge, Mass. and New York-based Black Diamond Therapeutics named Robert Ingram its new chairman of the board. Brad Bolzon of Versant Ventures will give up that seat, but remain on the board.
  • Toronto-based Deep Genomics hired Ferdinand Massari as chief medical officer.
  • Cambridge, Mass.-based Thrive Earlier Detection added three new executives. Sam Asgarian was hired as chief medical officer, Frank Diehl is the new executive vice president of product solutions, and Dina Ciarimboli is the full-time chief legal officer.
  • Watertown, Mass.-based Tarveda Therapeutics, a cancer drug developer, promoted Brian Roberts to CEO from CFO.
  • Waltham, Mass.-based Xilio Therapeutics, the developer of tumor-selective immunotherapies for cancer, named Rachel Humphrey to its board of directors.
  • France-based Genfit, the NASH drug developer, cut 40 percent of its workforce.
Regulatory Action

Plymouth Meeting, Penn.-based Inovio said its pivotal clinical trial of a COVID-19 vaccine has been placed on partial clinical hold by the FDA. The agency put the hold in place because of questions about the company’s planned delivery device for the trial. The Cellectra2000 device, the company says, “provides a brief electrical pulse to reversibly open small pores in the local skin area cells resulting in more than a hundred-fold increase in product delivery providing dose sparing and consistency.” Inovio shares fell 28 percent on the setback.

GSK won FDA approval to market mepolizumab (Nucala) as a treatment for Hypereosinophilic Syndrome (HES), a disease in which patients produce too many eosinophils, a type of white blood cell. The drug is an antibody directed against IL-5.

Japan-based Shionogi won FDA clearance to market cefiderocol (Fetroja) for hospital-acquired bacterial pneumonia and ventilator-associated bacterial pneumonia caused by gram-negative bugs like acinetobacter.

CSL Behring won FDA clearance to market Haegarda, (C1 esterase inhibitor Subcutaneous [Human]) to prevent attacks of hereditary angioedema in patients age 6 and older.

Cambridge, Mass.-based Solid Biosciences said the FDA lifted the clinical hold on its gene therapy for Duchenne Muscular Dystrophy. The company said it answered questions that the agency submitted in July, about manufacturing, updated safety and efficacy data, and “direction on total viral load to be administered per patient.”

Brea, Calif.-based Beckman Coulter secured Emergency Use Authorization from the FDA for a fully automated test to detect IL-6 in blood or plasma of COVID-19 patients – a sign of a potentially dangerous cytokine storm in severely ill patients.

Franklin Lakes, NJ-based BD said it received a CE Mark in Europe to market a rapid point-of-care antigen test for COVID-19 that can deliver a result in 15 minutes. The test has been available in the US since July.

Sunnyvale, Calif.-based Cepheid won Emergency Use Authorization from the FDA for its Xpert Xpress that can test simultaneously for SARS-CoV-2, Flu A, Flu B, and RSV infections – which all can present with some similar symptoms. The 4-in-1 test is designed to run on Cepheid’s installed base of 26,000 machines worldwide, and can deliver a result in 36 minutes.

Deals

Norway-based Vaccibody, a neoantigen cancer vaccine developer, formed a partnership with Genentech. The little company stands to collect $200 million in upfront and near-term milestones.

Belgium-based Celyad Oncology said it formed a clinical collaboration with Merck to test Celyad’s non gene-edited allogeneic CAR-T cell therapy candidate in tandem with the PD-1 inhibitor pembrolizumab (Keytruda).

Stamford, Conn.-based Sema4 formed a collaboration with Janssen Pharmaceuticals to use genomic data and data analysis to stratify patients most likely to benefit in its cancer clinical trials.

Switzerland-based Covis Group agreed to acquire Waltham, Mass.-based AMAG Pharmaceuticals for $13.75 a share, or about $647 million.

Tweetworthy

At a U.S. House of Representatives committee hearing on Wednesday, Rep. Katie Porter grilled former Celgene CEO Mark Alles for the years of unjustified price increases for the cancer drug lenalidomide (Revlimid). This public flogging was sharp and succinct. She hit on the classic, and unresolved, core issue in biopharma – how are companies supposed to fairly balance the responsibility to shareholders with the responsibility to patients? (Porter didn’t even get to the part where Alles did a lousy job for shareholders, as he ran Celgene into the ground, sold it to Bristol-Myers Squibb, and then personally walked away with even more money in his pocket). (Video clip, 5:04)

Rep. Porter had more to say about greed on the second day of hearings. This whiteboard drawing from Rep. Porter is worth keeping in mind the next time you hear a pharma CEO pat himself on the back for his company’s heroics.

30
Sep
2020

COVID-19 Diagnostic Testing Needs To Be Open, Not Closed

Alex Dickinson, co-founder and executive chairman, ChromaCode

The U.S. already has the real-time PCR infrastructure to run tens of millions of tests per day. Why don’t we use that capacity?

Our consumer electronics companies are constantly marketing the newest one-touch, automatic machine. They make it sound so simple, so alluring, so irresistible. Think of our smartphones and watches.

That might partly explain the administration’s instant embrace of the Abbott ID NOW point-of-care machine for COVID-19 testing. FDA commissioner Dr. Stephen Hahn touted the box’s features and benefits – a 6.6-pound box, the size of a toaster, that could deliver accurate yes-no test results in 15 minutes — at a Rose Garden news conference back in March. President Trump even seemed to cradle it briefly.

In late March, when horrific images from overwhelmed New York hospitals were dominating television, this seemed like good news that people wanted to hear. The call for the fast and simple answer was on display again this week, as the Trump Administration announced plans to distribute 150 million rapid antigen tests.

If there is a consistent pattern to this Administration’s response, it has been to put faith in quick fixes instead of strategic long-range plans that play out over a period of months. We’ve seen the unfortunate results of this excessive faith in the latest shiny technology object.

Like many things about the COVID-19 pandemic, the truth about point-of-care testing machines is more complicated than their coolly modern façades indicate. Whether these rapid testing instruments are molecular tests that look for the SARS-CoV-2 virus’ signature genetic material, or antigen tests that detect specific proteins on the virus’ surface, there are accuracy trade-offs with the rapid point-of-care tests, when compared with traditional real-time PCR platforms. Multiple academic studies have shown higher false-negative rates than what Abbott has reported on its ID NOW platform.

The issue of test inaccuracy hasn’t received much attention, but it has gotten some notice. Recently, the Governor of Ohio, Republican Mike DeWine, received a positive result from a point-of-care machine that detects antigens. Subsequent RT-PCR testing thankfully showed that the previous point-of-care test was a false positive.

The reliability of these sleek point-of-care machines is not the only thing that should concern us. These are highly automated, closed systems. That sounds good at first glance to most people, because it implies simplicity, speed and consistency. The term “sample to answer” is the diagnostic industry equivalent of “plug and play.” What’s not to like?

Besides the problems with accuracy, these closed systems create capacity constraints.

Our increasing reliance on these closed systems is hampering our ability to ramp up national testing effort to the scale it needs to be. Recent reporting from Reuters assigns blame for the U.S. testing shortfall on these closed testing systems.

These companies operate on the razor and razor blade model. They sell the machine first, and then supply the proprietary consumable supplies such as chemical kits and plastic sample plates and pipettes needed to make it run. It’s similar in concept to the branded printer cartridges in use on your HP or Epson home printer.

The rapid point-of-care COVID-19 diagnostic testing companies – four major manufacturers – simply can’t manufacture enough consumable chemicals, sample plates and pipettes in their special kits to keep their instruments running at full capacity nationwide. This is the primary cause of testing backlogs. And it’s not just the “printer cartridges” that are in shortage – many of these companies can’t make enough of the “printers” themselves to meet demand.

These closed systems can be useful and convenient under normal circumstances. During a typical flu season, hospitals can quickly determine whether a few patients are suffering from the flu or a different respiratory illness and treat accordingly.

But a pandemic is not a normal circumstance. And while some experts have recently called for an increase of less accurate antigen tests to more quickly track and contain the virus — essentially throwing “everything but the kitchen sink” at the problem because the U.S. has struggled to generate a comprehensive national testing plan — there’s no evidence that these manufacturers can produce the instruments and consumables at the necessary scale.

For our country to reasonably manage this pandemic, we need much more than just the test results for a few dozen patients at a time delivered by a quick, easy-to-use black box.

These point-of-care machines do have a role to play, however. They can be invaluable tools for COVID-19 testing in specific environments, such as daily testing of caregivers at assisted living facilities. But as a nation, experts say we need to be doing millions of tests per day — the latest estimate by Harvard’s Global Health Institute is 4 million per day. The network of discrete closed systems cannot handle that kind of volume, nor is it flexible enough to scale up and down as testing needs change.

Fortunately, we already have an open-system network that we can harness to produce the amount of testing needed to safely reopen our economy: an installed base of “old-fashioned” real-time PCR machines.

Every big central lab, government lab, hospital lab and research lab in the country has at least one open-platform real-time PCR instrument. Different manufacturers make these instruments, but they all run in basically the same way, using the same chemical supplies.

These systems aren’t as quick to deliver results and require trained technicians to operate. But they can handle tremendous volumes of tests. These traditional RT-PCR machines can handle 10-100 times higher throughput than the point-of-care tests, and are able to generate >1,000 results per day per machine. In contrast to ID NOW, they are the gold standard of accuracy.

Certainly, open-platform PCR instruments are currently handling a significant portion of the testing. But right now, only labs that are CLIA certified by the federal government are allowed to handle diagnostic testing. That leaves out most academic labs and many, many commercial research labs that are run to exacting specifications, but are not technically CLIA certified. Add to that a large number of PCR instruments that run diagnostics for our pets. We love our pets, but I think most people would agree to delay those diagnoses to free up capacity for the urgent COVID-19 demand.

The federal government could, and should, use its influence to requisition these instruments for use in COVID-19 testing. The capacity exists – it just needs to be properly directed and mobilized. It’s not too late to swing into action.

At a low estimate, the U.S. has 30,000 open-platform PCR instruments. If each instrument is leveraged to run the 1,000 tests per day it can run, that would give us 30 million tests per day. Capacity could be stretched further through the use of robots, pooled testing or the application of data science.

This is a robust infrastructure that will better meet the testing demands this pandemic calls for. Testing at high volume will help us find our way back to something closer to normal until we have a widely distributed vaccine. 

The role for open systems is clear.

As Gary Kobinger, a Canadian researcher best known for his work on Ebola, argues in that same Reuters report that all diagnostics should be done on open platforms. “At one point there will be a new pathogen, and the company that makes the cassette that is controlling everything is not going to be able to supply you,” Kobinger said. “And this is where we are now, right?”

Right.

 

Alex Dickinson, PhD is the co-founder and executive chairman of ChromaCode. He is a life sciences executive with over 25 years of experience leading strategic initiatives that have transformed small companies and enabled large companies to penetrate new markets. Most recently, he was the Senior Vice President of Strategic Initiatives at Illumina, where his responsibilities included working with national governments and large institutions to develop precision medicine programs for healthcare systems.

28
Sep
2020

Small Molecules Against RNA Targets: Jennifer Petter on The Long Run

Today’s guest on The Long Run is Jennifer Petter.

She is the founder and chief scientific officer of Waltham, Massachusetts-based Arrakis Therapeutics.

Jennifer is a medicinal chemist who has spent her career thinking about how to make small molecules with all the classic Lipinski “Rule of 5” characteristics against protein targets.

Jennifer Petter, founder and chief scientific officer, Arrakis Therapeutics

Five years ago, when she was looking for a new entrepreneurial challenge, she attended a Gordon conference. She saw a couple scientific presentations from Matt Disney at Scripps and Kevin Weeks at UNC Chapel Hill that gave her an idea. Might it now be possible to make small molecules against RNA targets?

She was inspired to get going on building a new drug discovery platform at what we now call Arrakis Therapeutics.

Arrakis took this work up several notches this spring, through a new partnership with Roche. The big drugmaker, seeing the possibility for creating multiple small molecules against RNA targets, agreed to pay $190 million upfront to Arrakis to work together on making it happen. Arrakis also recently described its work in detail in its first peer-reviewed publication in ACS Chemical Biology.

Jennifer also used to identify as a man, and was known as Russ Petter. She came out publicly as transgendered in June 2018. CEO Mike Gilman wrote about it on the company blog.

In this conversation, we spent the first part talking about Jennifer’s early life and key steps in her career leading up to her current work at Arrakis. At the end, we talked about her gender transition and how she handled that in the workplace. I think my questions are a little awkward, but it’s OK because the situation was awkward for a lot of people. It’s old news, but I think Jennifer has some timeless thoughts on handling the situation with grace.

Now, please join me and Jennifer Petter on The Long Run.