Eli Lilly and Company (NYSE:LLY) Lilly Oncology ASCO Investor Event June 2, 2024 8:30 PM ET
Company Participants
Jake Van Naarden – Executive VP & President of Loxo
Lillian Smyth – Senior Vice President and Global Development Head for Breast Cancer at Loxo
John Pagel – Vice President and Head of Global Hematology at Loxo
Geoff Oxnard – VP of Clinical Development, Global Head Thoracic Cancer at Loxo
Arjun Balar – MD, Vice President, Global Clinical Development at Loxo
Barry Taylor – Chief Scientific Officer of Loxo
Kara Clinton – Senior Vice President Oncology Medical Affairs
Winselow Tucker – Senior Vice President & Chief Commercial Officer for Loxo
Conference Call Participants
Evan Seigerman – BMO Capital Markets
Seamus Fernandez – Guggenheim Securities
Geoff Meacham – Bank of America
Steve Scala – TD Cowen
Chris Schott – JPMorgan
Akash Tewari – Jefferies
Dave Risinger – Leerink Partners
Jake Van Naarden
All right. Can everyone hear me, yes. Okay. I think we’re going to get started. All right. Thanks, everyone, for coming tonight. And for those online, thanks for being patient as we let people get in and take their seats. I know we’re starting a little bit late. My name is Jake Van Naarden. I lead the oncology unit here at Lilly, which, in the case of oncology, actually spans our discovery efforts all the way through our commercial medicines, and you’ll see that reflected actually in what we walked through here tonight.
Over the past couple of years, we’ve been working to modernize the portfolio and strategy in oncology here at Lilly. And we’re doing this event tonight because I think we’re really entering a new phase. And so tonight, we’re going to tell you a little bit about how we think our philosophy about (indiscernible) creation, what we’ve been up to over the past couple of years in these efforts, walk through the portfolio itself and importantly, actually introduce you to a bunch of members of the team who don’t do as much IR and don’t do so much media but are actually the ones doing all the hard work, getting our medicines built and bringing them to patients. So I’m excited for you to meet them here tonight.
This is the safe harbor provision. So in terms of agenda, I’m going to start and kick things off, talk a little bit about our strategy, our vision, the kinds of medicines we make, tell you a little bit about (indiscernible) the changes in both the research and development portfolio and philosophy over the past couple of years, give an update on the commercial performance of our approved medicines. And then hand it over to my teammates, my colleagues who are going to walk through a bunch of the actual medicines in the portfolio that we’re excited about. And then we’ll take some Q&A at the end.
Speaking of the team, really excited about the team we’ve built here in Lilly Oncology. This is a group of people who the vast majority of whom are new to the company over the past couple of years. Many of them are new to industry. I just couldn’t be more thrilled with the kind of talent that we’ve been able to attract, which we think is a big reflection actually of the medicines we’re working on. Great people want to work on great medicines. We’ve had the bring a really great team together that I think rivals the best, frankly, among our peer set. And so you’ll hear from them tonight as we talk through the portfolio.
So a couple of years ago, in 2019, Lilly bought Loxo and I came to the company from that acquisition. And in the ensuing year, A bunch of us had the opportunity to take on increasing roles in leadership we were reporting an opportunity to reshape what oncology look like here at the company.
But Lilly’s have a long history in oncology. This is our first gambit. Some of the first chemotherapies ever invented or Lilly products, there, many of them are in use today are still standard of care in tens of thousands, hundreds of thousands of patients globally as generic medicines. Some of the first model antibodies in oncology or Lilly products still in today. And of course, Verzenio medicine has made a huge impact for people with breast cancer has, of course, been an important business contributor as well.
Verzenio aside, the past called decade of oncology research at Lilly has not been as successful. And that was the opportunity we had a few years ago to change what that looked like.
So starting with strategy. How do we think about oncology medicine creation and delivery? Really, 3 things. And it starts with a conviction in biology. The entire effort is biology oriented. It’s not around where the tumor rose inside of the body. It’s not around some categories of biology. It’s about — it starts with identifying targets, proteins or other parts of the cell in, on around a cancer cell that we think that if you build the right medicine, step 2, that you can actually get the tumors out of die and you observe that both preclinically and eventually, you can observe that clinically on conventional imaging.
Monotherapy activity clinically is what we’re looking for in the right patients. We beat these medicines up a ton preclinically, and we build them to pretty exacting standards. Of course, they’re not always going to be perfectly replicable when you put it in the clinic, but we think that through this approach, we can build a portfolio with a reasonably high probability of delivering medicines come the end of clinical trials.
And so when we enter the clinic, yes, we’re looking for monotherapy activity. Sometimes it’s going to be robust enough to allow for an accelerated approval in a late-line population where there’s a significant unmet need. We’ve done those. Sometimes it’s just enough to tell you have something real that you can invest more capital into in randomized trials and prove out through other endpoints, and we’ve done that as well. And so this is the approach we take. And it really centers on biologic conviction, target selection and building the right medicines.
How do we do it? We do it through a combination of our internal discovery efforts and business development. A couple of years ago, we recognize that we really needed business development to help accelerate this modernization effort. And so we’ve done a lot of deal making since the Loxo acquisition. We’ve purchased companies both public and private. We’ve done discovery partnerships. We’ve done licensing transactions. We’ve gravitated towards earlier-stage deals. That’s where we think the biggest opportunity is to actually create value. And I think we will continue to be active in business development.
It’s really a philosophy of agnosticism as to where good medicines come from. Our discovery teams are actually at the table with our business development professionals and identifying these opportunities. We don’t sort of have a first principle about where a good medicine comes from. We have a lot of smarter people here. There’s a lot of smart people out there in the external environment. And if we can find programs that we otherwise wouldn’t have the ability to make or can de-risk things we’re already working on forward, integrate things we’re already working on, we love to be able to bring those things in from the outside world.
So let’s talk about the portfolio. So in late 2019, early 2020, this was the portfolio of approved and investigational medicines that we inherited and had to decide to do it.
Unfortunately, most of the medicines, certainly in the first 2 columns on this slide we’re either sort of already failing or we thought were destined to do so. And so we took the bold initiative in the first couple of months of assuming these roles to terminate nearly the entire pipeline. And we made a bet. And we made a bet that in the ensuing years, through a combination of internal discovery efforts, leveraging the philosophy I just talked about, through business development and through extreme focused, we could build back to a pipeline of size that would rival what we inherited, if not larger, with a portfolio probability that had a much higher chance of delivering real medicines that made a big impact to patients come the end of their journey while at the same time taking the handful of medicines that did look real and actually progress them through the development portfolio and get them approved launch, et cetera.
And I’m excited tonight to report in this way that I think we’re well on our way to doing that. Now do we yet have the portfolio of clearly working medicines in the clinic that resulted from these efforts? No, we’re not there yet. But I think we’re entering a new phase of what that could look like. Importantly, as you can see on this slide, that dotted box on the upper left represents entirely new projects since that 2019, 2020 time frame. These were literally like ideas on a sheet of paper that we either brought in from the outside world and perfected or we built from scratch. And of course, we’ve moved important medicines from the left to the right. You’ll hear about a lot of those later this evening.
We’ve also diversified the modalities. A couple of years ago, if you wanted to make a cancer medicine at Lilly, you could make a small molecule or you can make a vacant antibody, and that was basically it. And our observation and looking at the increasingly complex landscape, patient need and biologic opportunities was that we needed the ability to diversify the modalities for which we could make cancer medicines, which meant diversifying the technology toolkit that we actually had. And I’m proud to say we’ve been doing this. Of course, we’re continuing to make selective potent oral small molecules. You’ll hear about a bunch of them tonight. We’re putting our first antibody drug conjugates into the clinic, where we’ll have our first T-cell redirector into the clinic next year. And by way, at least initially of the POINT Biopharma acquisition, we’re now making radioligand therapies with our first one of those in a wholly owned way in the clinic now as well. So I expect over the ensuing years, the color diversity will continue, in fact, perhaps even get more balanced over time. But I’m excited that we — when we identify a target now, we identify a product profile that we think can work in a patient and be differentiated, we now have a technologic toolkit that we can apply in a diverse way of how to make that medicine.
While we were retooling the discovery portfolio, we, of course, were trying to jump-start pivotal development for the medicines that we thought warranted late-stage development kickstarted a suite of late-stage development for a handful of different programs a couple of years ago when this all happened, and interestingly, this resulted in a pretty concentrated clustering of readouts, 8 randomized trial readouts in the span of 12 months. It’s probably the most concentrated number of late-phase readouts in oncology that Lilly have ever had. And most of them have been successful. Of course, they’re never all going to be successful. That’s just not the way this business works. I wish it were, but it usually isn’t. But most of them have been, and that’s really great for patients. It’s really great for advancing our medicines towards approvals and new indications. There’s still one more coming soon. This is the imlunestrant EMBER-3 Phase III study, the first Phase III study for the oral SERD in second-line ER-positive metastatic breast cancer. We’ll hear a little bit more about that later tonight from Lillian. Important Phase III study coming later this year for that medicine.
And of course, all of the discovery retooling has led to an unusually active agenda for new clinical trial starts in calendar 2024, 8 new clinical trial starts for new molecular entities this year, 3 of whom have already dosed their first patients. The rest are still on track to happen throughout the remainder of the year. Perhaps not coincidental that this is such a robust year for new clinical starts. We started this process, as I mentioned about 4 years ago. That’s roughly how long it takes in sort of a best-case scenario to make a new medicine from scratch against the standards that we hold ourselves to. And so it’s really this bolus of early phase momentum that I think allows us to feel like we’re starting a new chapter for oncology R&D here at Lilly.
While we’ve been doing this on the R&D side, of course, we’ve been busy (indiscernible) launching new indications for other medicines. And that’s been going pretty well, especially as of late. The growth that we’ve seen commercial in the portfolio has been pretty robust, especially across our 3 key growth brands, Verzenio, Retevmo and Jaypirca. Verzenio bemaciclib, a medicine that we all know well, has really had a tremendous couple of years for patients, particularly those with high-risk early breast cancer. We’re about 2 years into the launch of that indication for this medicine, and it is very clearly now the standard of care for these patients. It’s a 2-year regimen and then patients are finished that I think has really bent the curve for patients with high-risk EBC, especially with the increased follow-up that we’ve had on the monarchE study and the duration and frankly, expansion of benefit even when patients have finished the regimen.
Uptake in this indication has been fairly brisk. We estimate that about 60% of the eligible population now receives this regimen, which is sort of an interesting work shack test of how you think that is. If there were any other therapeutic area, 60% 2 years into a launch, we feel pretty good. Unfortunately, in oncology, especially in a curative setting, we’re not satisfied with that. So we have work to do to make sure that every eligible patient in the curative setting of high-risk EBC has the potential to benefit from this regimen. And we’re going to continue working until we get as close to 100% as we can.
Jaypirca, pirtobrutinib is our newest approved medicine in oncology. This is the non-covalent BTK inhibitor. We have 2 indications under accelerated approval that were both approved last year, first in relapsed mantle cell lymphoma; second, in relapsed CLL both admittedly small indications to start. Although interestingly, we found that the CLL indication, which is for patients who’ve already received at least a covalent BTK inhibitor and venetoclax or BCL-2 inhibitor, it’s probably a larger indication than we initially thought. The growth in those medicines over the past couple of years have led to an increasing population of patients who’ve now relapsed those medicines and have extremely poor outcomes that Jaypirca can really offer a nice durable disease control for them. So we’re continuing to execute commercially to bring this medicine to patients.
It’s been interesting. We’ve gotten a lot of anecdotal feedback from patients and from physicians about their initial experience with this medicine, particularly those who weren’t involved in the clinical trial program. So this is really their first experience and just great things to say about what the medicine has been able to do for patients in terms of both efficacy and tolerability. I say that because we’re still really in the earliest innings of the overall life cycle of this brand. You’ll hear from John a little bit later tonight about the large development portfolio here. Most of which hasn’t actually read out yet. So there’s a lot more indications to come for this drug. And the early signs of its adoption as well as just the experience that patients and physicians have had, I think, bode really nicely for the medicine’s potential future.
So you’re going to hear now from the team about our investigational portfolio for the most part. And as we’ve been working over the past couple of years to speed our medicine delivery to patients. We have worked tirelessly to squeeze out every ounce of efficiency from every part of the drug development cycle and yet we still face a pretty significant bottleneck, which is actually clinical trial enrollment. It remains by far biggest bottleneck to getting our medicines to patients. Our trials just do not enroll as quickly as we want them to. And yet ironically, particularly in the United States, clinical trial enrollment is not a priority for the health care system. 7% of U.S. cancer patients ever enroll in a clinical trial despite it being a great way to access great care, to access standard of care oftentimes to access new investigational medicine. So I mentioned it here tonight because you’re going to be hearing more from us in the future about this.
We are — we’ve initiated a new effort this year to try and change perceptions about clinical trials in the United States, in particular, and really do more to broaden clinical trial access for patients in this country. You’ll hear a little bit later tonight about the first Phase III trial for olomorasib, or KRAS G12C inhibitor in lung cancer, which is really the first study where we’re piloting actually a lot of new ideas for trial implementation to try and broaden access. So I’ll just give you a flavor for what that’s like and you hear about the study from Jeff in a bit.
We’re allowing entirely local biomarker testing. So whatever patients and providers are using to identify let’s say, PD-L1 levels or KRAS mutations, that liquid biopsy or solid tumor biopsy makes them eligible for the study.
We’re allowing a cycle of standard of care medicines prior to even coming on to the study because we recognize that for patients who are newly diagnosed, they may be waiting for those biomarker results. They may be nervous about a clinical trial and not know how to think about it. They may be going through the study screening procedures. We’re allowing a cycle of standard of care before coming on to our program.
And interestingly, we’re doing something that I think is a first, actually, certainly a first at Lilly, I think, a first for industry, which is that we’re actually paying for all standard of care procedures on the study in addition to the medicines themselves. So CAT scans, labs, all of the things that would normally just be built to insurance, we’re actually paying for. This is actually the normal course for every other disease that we work on at Lilly. We probably should have implemented this in oncology years ago, but we’re doing it now because it’s the right thing to do to. and if we’re going to change the way that patients and providers think about clinical trials, particularly in the community, we really need to eliminate every barrier we possibly can. So you’ll be hearing more about this from us over the coming months and years. I hope we’re able to make it dent here.
And in the meantime, I’m going to hand the mic over to my colleague, Dr. Lillian Smyth, who’s going to walk us through our breast cancer portfolio.
Lillian Smyth
Thanks, Jake. Hi, everyone. Good to see you. I’m Lillian Smyth. I’m a medical oncologist. I was previously on faculty with the breast medicine and early drug development service of Memorial Sloan Kettering. So I used to see breast cancer patients in my (indiscernible). I was also a clinical trial investigator enrolling to many studies. In the last few years, I joined Lilly, and now I lead development of our breast cancer medicines.
So let’s jump right into Verzenio. So as you know, abemaciclib is our oral potent CDK4/6 inhibitor with greater selectivity for CDK4 than 6, which allows us to continually dose this medicine due to less myelosuppression. Shown here are the pivotal studies that we’ve conducted across the entire hormone-positive HER2-negative breast cancer continuum that have led to key indications for abemaciclib. We know that abemaciclib is approved in advanced breast cancer is monotheraphy with a combination with (indiscernible) therapy, and abemaciclib is the first and only CDK4/6 inhibitor approved for the treatment of patients with high-risk early breast cancer.
I’m also going to talk to you a little bit about the post-MONARCH study, which you may have seen was presented at ASCO yesterday, addressing a very important question for the community about CDK4/6 inhibitor sequencing with fulvestrant and abemaciclib after progression on a CDK4/6 inhibitor.
So obviously, there’s been a lot of noise about the evolving adjuvant CDK4/6 inhibitor landscape. So I just wanted to remind you about the data that we have with monarchE. As you know, monarchE was the only adjuvant CDK4/6 inhibitor study designed actually for patients with high-risk disease. And we now have a very mature data set with all patients off therapy and in long-term follow-up.
Shown here is the 5-year outcome data, which is a really important landmark for this disease. And you can see clear separation of the curves well beyond the treatment period. This is what we call carryover effect that we’re seeing from abemaciclib Ultimately, of 5 years, we have seen a 32% reduction in the risk of developing an IDFS event, which translates to almost an 8% absolute difference in the risk of IDFS at 5 years.
Abemaciclib and endocrine therapy is the globally approved standard adjuvant therapy for high-risk early breast cancer, with the category 1 NCCN designation and a strong ASCO guideline recommendation.
Now let’s touch on the post-MONARCH study. So we saw these data presented by Kevin Klinsky yesterday. And to remind you, the post-MONARCH study addressed a question about continuing CDK4/6 inhibition beyond progression on a CDK4/6 inhibitor first line. Patients received abemaciclib plus fulvestrant or fulvestrant alone. And you can see here at the primary analysis, abemaciclib added to fulvestrant, improved investigator-assessed progression-free survival with a hazard ratio of 0.73. And just to put these data in context, we know that there is a really high unmet need to optimize therapy in that post-CDK4/6 inhibitor setting. And when you look at these data and when you consider that the standard care is quite fractured actually in the second line with CDK setting. Targeted therapy options are limited and are associated with varying toxicity.
Fulvestrant and abemaciclib offers a simple treatment solution with an observed treatment effect that is at least directionally similar to what we’ve seen with approved targeted therapies. The conclusions from this study is that post-MONARCH is the first Phase III randomized placebo-controlled study to demonstrate the benefit of continued CDK4/6 inhibition after progression on a CDK4/6 inhibitor. In this trial, abemaciclib improved progression-free survival despite the control arm performing better than expected with a 27% risk reduction for developing a PFS event, consistent benefit across multiple prespecified and clinically relevant subgroups, including key biomarker subgroups and a consistent improvement across key efficacy endpoints. Safety in this study was consistent with what we know about abemaciclib and discontinuation rate importantly was low at 6%.
Abemaciclib and fulvestrant offers a targeted therapy option after disease progression on a CDK4/6 inhibitor for patients with hormone positive HER2-negative advanced breast cancer not selected for biomarker status. And we think that these results really have the opportunity to complement EMBER-3 as what Jake mentioned, which is our Phase III study, which will read out later next year or later this year with our oral third imlunestrant.
Now talking about imlunesterant. This is our brain-penetrant oral search designed for continuous ER target inhibition, including an ESR1 mutant breast cancer. As I mentioned, there is no clear standard of care after receipt of a CDK4/6 inhibitor in the first-line setting, although I’ve shown you just some evidence for abemaciclib from post-MONARCH-in this setting.
There is also no SERD approved in combination with the CDK4/6 inhibitor or indeed in the adjuvant setting. So the thesis of our program was twofold: to develop an oral SERD that could differentiate in the advanced breast cancer setting by combining with abemaciclib, including in that pretreated CDK4/6 population. And then secondly, to displace standard of care adjuvant endocrine therapy in the adjuvant setting.
In terms of the Phase I data that we’ve generated from our EMBER Phase I study with imlunestrant, we’ve seen encouraging clinical efficacy, particularly in that second line post-CDK4/6 inhibitor setting, which is the subject of our ongoing Phase III EMBER study and we’ve seen efficacy of imlunestrant alone but also in combination with standard of care targeted therapies shown here on the far right panel, we’ve evaluated imlunestrant with abemaciclib with everolimus and alpelisib, all therapies that we use in the clinic.
From a safety perspective, imlunestrant’s monotherapy is well tolerated, and importantly, it safely combines with standard of care targeted therapies.
As I mentioned, our pivotal registration studies are ongoing, and include EMBER-3, which is our first Phase III study in the advanced breast cancer setting. This study enrolls patients who have received an imlunestrant inhibitor with or without a CDK4/6 inhibitor across 3 study arms: imlunestrant alone, investigator’s choice endocrine therapy or imlunestrant in combination with abemaciclib. We have dual end point evaluation in both the ITT and ESR1 mutant population. And this study represents the first evaluation of (indiscernible), in combination with the CDK4/6 inhibitor in a Phase III study in the second-line setting. This should position us for dual approval, both as monotherapy and in combination with Verzenio. And although we are aware of the risks of studies like this, we are looking forward to seeing the data later this year. And if positive, this study positions imlunestrant to be the second oral third to market and the first in combination with the CDK4/6 inhibitor.
EMBER-4 is our adjuvant study, enrolling early breast cancer patients who have already received 2 to 5 years of adjuvant endocrine therapy, which may have included a CDK4/6 inhibitor. We are randomizing 6,000 patients across 2 study arms, 5 years of imlunestrant or 5 years of continued standard adjuvant endocrine therapy. This trial evaluates a sequencing strategy for patients who write increased risk of recurrence and really positions us to address the full continuum of the adjuvant treatment journey. And you can envisage a future state if EMBER-4 is positive, where patients are sequencing from up-front abemaciclib and endocrine therapy then to imlunesterant to complete their adjuvant therapy journey, which for many patients is 7 to 10 years, particularly for those that increased risk. This is the largest oncology trial we have ever conducted it clearly, and we’re looking forward to see this study complete in 2027.
So finally, ending on LOXO-783. This is our highly mutant selective H1047R allosteric inhibitor. And just to grant you in the landscape, we know that these H1047R mutation, they’re actually the most common (indiscernible) mutations that we see in our clinic. They occur in 15% of breast cancer. And the limitation of current therapies, current AKT and PI3-kinase inhibitors are not wild-type sparing, are not mutant selective same thing, meaning that they lead to wild-type mediated toxicities, particularly hybrid lycemia, skin toxicity and GI toxicity. And so the thesis of our program was to develop a mutant selective inhibitor that spares wild type entirely and would lead to superior tolerability and efficacy in the clinic.
783 preclinically shown on the far right panel here achieves significant tumor regression as monotherapy when compared to (indiscernible) but importantly, does not lead to increases in insulin or C peptide.
We’ve also generated other preclinical data of 783 in combination with endocrine therapy and CDK4/6 inhibitors, and here, we see additive efficacy with those combinations.
Our Phase I study the (indiscernible) trial is ongoing, and we’re looking forward to sharing data later this year and (indiscernible) to say that LOXO-783 is part of a broad discovery campaign that we have against this target. And in fact, we’re continuing to advance next-generation molecule to be clinic ready in 2025 as we continue to find the clinical profile of 783. As you know, we’ve been planning for a little while now, evaluating 783 as monotherapy and in combination and continuing to refine our understanding of the efficacy profile.
Really, the first important goal of this program was to establish have we developed a truly mutant selective inhibitor that spares wild type and avoids hyperglycemia? We have. LOXO-783 does not cause hyperglycemia in the clinic.
From an efficacy perspective, this medicine has efficacy, and we’re continuing to refine whether that efficacy is good enough in the context of the evolving therapeutic landscape and also the advances we’ve made in our own discovery campaign with molecules that are near clinic-ready for 2025. So with that, I’ll hand you over to Dr. John Pagel to continue the portfolio review.
John Pagel
Good evening, everyone. I’m the guy that brought the water up, struggling a bit with my voice. So bear with me if you could. Thanks for joining us. I am the Global Head of Development for Hematology here at Lilly. I’m going to give you a little bit more about the development of Jaypirca, although, of course, Jake’s introduced that a little bit for us tonight. And I’m going to tell you about a new drug that’s just entered the clinic targeting the full receptor using a novel antibody drug conjugate.
About 2.5 years ago, I joined Lilly, but before that time, I spent more than 20 years in academia doing translational research. I had a research lab, and I saw patients with blood cancers. And it was during that time where I was an investigator on the Phase I/II BRUIN study exploring the use of Jaypirca in patients with relapsed peel malignancies. And I saw firsthand on what this drug did for patients. And because of that, I wanted to be closer to its development.
Of course, you know a lot about Jaypirca. I’ll remind you that it is a Bruton’s tyrosine kinase inhibitor, but it’s a different type of BTK inhibitor. It is very highly selective, but importantly, it is a noncovalent BTK inhibitor, and it’s really been designed to overcome the limitations of the covalent BTK inhibitors. And as you have heard and based on our strong efficacy and well-tolerated safety results from the Phase I/II BRUIN study, the FDA-approved Jaypirca for relapsed mantle cell lymphoma patients after 2 prior lines of therapy, including a BTK inhibitor, and in CLL after 2 prior lines of therapy after both a BTK and BCL-2 inhibitor.
And you can see on this slide, we have a comprehensive suite of Phase III studies, and I’m going to mention a little bit more in detail in just a subsequent slide. But first, I want to tell you a little more again about the efficacy of Jaypirca. These are the waterfall plots showing tumor size reduction in patients on the left with CLL. All of these patients had received prior covalent BTK inhibitor and in mantle cell lymphoma, which is on the right. And again, all of these patients had received prior BTK inhibitor.
These patients are very difficult patients. Not only had they received prior BTK inhibitor therapy, but many other classes of therapy. And in particular, if you see on the left, those CLL patients, all again, who had received a covalent BTK inhibitor, many of them had received as well a BCL-2 inhibitor, venetoclax. And that’s represented by all the aspects that are at the bottom of those vertical bars representing an individual patient tumor size reduction. That’s important because that is a patient population that historically has been of high unmet need due to actually very poor survival.
Similarly, on the right, we see a pattern of patients treated again with a covalent BTK inhibitor, but also many other lines of therapy, including lines of chemo immunotherapy as well. And this is also a patient of high unmet need. This patient population historically has had survival of 6 months or less.
We have built on these robust responses from this study to develop a comprehensive advanced series of Phase III studies, and those are shown here. We have tried to explore the use of Jaypirca through multiple different treatment settings. This includes first-line second line using continuous therapy as well as fixed duration therapy.
We recognize that there’s a need for improving on the therapies and as well as improving on the tolerability profile of the medicine. And therefore, we are comparing Jaypirca to globally established combination therapies, including covalent BTK inhibitors or chemoimmunotherapy.
Also, we recognize that there are some patients who, where a fixed duration approach might be important for them. So we are enrolling patients in a relapsed CLL trial using Jaypirca in combination with venetoclax. That study is now continuing to enroll.
The other 4 company-sponsored studies shown here have all completed enrollment. We expect to enroll about 2,200 patients over these company-sponsored studies and look forward to them reading out over the next couple of years.
I will note the BRUIN 321 study, that will be our first readout of a randomized Phase III study that’s our study comparing Jaypirca to idelisiband rituximab or bendamustine and rituximab and we anticipate those results being reported again later this year.
Lastly, I want to point out on the far right, the BRUIN CLL-18 trial. We’re excited about this trial. This is a trial that will be run through the German CLL study group. It’s anticipated to begin later this year. This is a trial using Jaypirca in combination with venetoclax and a fixed duration approach in the first line.
Overall, I think you can see that these studies show the confidence not only in relapsed/refractory CLL of Jaypirca but also in the first line and with fixed duration therapy.
Okay. I told you that I’ve lead development in hematology. However, we have lots of medicines that are entering the clinic as you can see and many of them simultaneously. So I do lead other programs, and this is one of them.
Of course, we also are very aware that recently, the FDA-approved an antibody drug conjugate targeting the full receptor for patients with relapsed ovarian carcinoma and that’s Elahere. Elahere was shown to be superior to investigator’s choice of chemotherapy in third-line ovarian cancer patients, but only those who have high folate receptor expression. We also know that Elahere has some significant toxicities, including ocular toxicity. So we recognize that there’s an opportunity to establish a new standard of care, not only targeting those patients who might have a high folate expression, but low and moderate expression levels as well. And hopefully, that would be relatively well tolerated.
This is an antibody drug conjugate that uses a topoisomerase payload and has a drug antibody ratio that’s high, and we have significant excitement around this product, and it’s actually now just entered the clinic.
And I just want to show you a little bit of preclinical data to support our conviction that this drug will be active in those patients with perhaps low and moderate folate receptor expression levels. On the left, you can see a model of high folate expression, but in the middle 2, our moderate and low folate expression tumor models where Elahere is much less sensitive. And even in a model on the right of a non-ovarian cancer or colorectal where we see significant activity with our fully directed antibody drug conjugate.
As I said, this program has now entered the clinic, and we’re excited to see where this goes over the next many months. And I think with that, I’ll turn it over to my colleague, Dr. Jeff Oxnard. Thank you.
Geoff Oxnard
Good evening. I’m Geoff Oxnard. I lead lung cancer clinical development at Lilly. I’m going to tell you about a couple of molecules focused in GI and thoracic cancers. I’m a lung cancer doc, myself. I still see patients part time at Boston Medical Center, a safety net hospital in Boston. I joined Lilly last year, previously lone faculty at Dana-Farber and Harvard Medical School for about 10 years, where I was deeply involved in targeted therapy development and biomarker development. And I was one of the lead investigators of the first in human in the positive patients over the years and to see its impact. That’s one of the joint Lilly last year.
Retevmo, I think, nicely demonstrates the 3 principles that Jake told you about earlier, find a target that is important in cancer, RET develop a drug that differentiates compared to the available therapies. We know multi-kinase inhibitors had their limitations in terms of tolerability and potency limitations that Retevmo overcomes. And three, develop the drug into the need. Retevmo has been exhaustively developed. This drug has tumor-agnostic approval in RET-positive cancers in the U.S. and in the EU. It recently received pediatric approval in the U.S. It’s the only RET inhibitor indicated in patients aged 2 and above with RET-positive cancers. It’s underdeveloped in adjuvant lung cancer, a timely space for targeted therapy development. And finally, we’ve just described 2 positive first-line trials in RET-positive non-small cell lung cancer and RET-positive thyroid cancer. These are those 2 trials, both were reported last year at ESMO, both were co-published in the England Journal of Medicine and both demonstrate the impact of this drug when given first line compared to standard of care, PFS hazard ratio of 0.47 on the left in lung cancer of 0.28 on the right in RET-positive thyroid cancer.
This is the activity one would expect from an effective RET-targeted therapy, but these were difficult trials. People said these trials couldn’t be done. We are proud to have completed these first-line trials in rare biomarker positive populations to demonstrate the impact of the drug and to enable access to this drug globally where randomized trial data is really important.
Of course, a RET inhibitor only works if you test for RET. And so enabling access and adoption of molecular testing is critical for this molecule and other molecules in our programs, and we continue to advocate for this.
If you do cell molecular testing, the most common result you get back is KRAS. I’m going to tell you about our KRAS portfolio. These are 3 drugs on the left, olomorasib, a second-generation potent KRAS G12C selective targeted therapy in the middle, KRAS G12D targeted therapy, and only bioavailable drug. This drug is more focused in GI cancers, G12D is the most common KRAS variant. And finally, a pan-KRAS inhibitor, an ambitious drug also orally bioavailable. Olomorasib is entering late phase clinical development. The other 2 drugs are entering the clinic later on this year. I’ll tell you more in a moment.
Let’s start with G12C. G12C is 15% of non-small cell lung cancer discovery of G12C inhibitors create enormous excitement about the ability to target KRAS. The FDA approval of these has moved the bar. And yet some of that optimism has not come to fruition. These drugs are mostly making an impact in previously treated non-small cell lung cancer. First-line lung cancer remains a meaningful unmet need where we think we can make a difference.
Our thesis was that by developing a selective potent, tolerable drug with a wide therapeutic index would allow us to do the difficult thing that has not yet been done in lung cancer, combined a targeted therapy up-front with immunotherapy. Something we think we are well positioned to do.
On the upper right, I’m showing you the target inhibition or targeted occupancy of this second-generation G12C inhibitor compared to first-line — first generation inhibitors, where we see more consistent and more effective target inhibition.
And on the bottom right, I show you pan-tumor activity. We recently showed at ASCO, demonstrating the effect of this drug. But the real reason to believe is data like this, this is new data of olomorasib after treatment with a prior G12C inhibitor.
What you see on the left is patients who discontinued due to toxicity or on the right patients who discontinued progression. Most of these patients, they’re immediate prior therapy, was a G12C inhibitor, and we see a response rate above 40%. We see in PFS of 8 months in this heavily pretreated population. We don’t think this drug is overcoming resistance. We think this drug is overcoming the target inhibition and PK limitations of first-generation drugs.
Importantly, it has differentiated tolerability. The predominant AE range is grade 1. 1 patient discontinued due to treatment-related adverse events even in patients who have intolerance to prior drugs, we see tolerance of this drug. (indiscernible)investigators in the study tell me that this is a drug people can be on for durable periods. We think this is a differentiated tolerability that will enable first-line combination with (indiscernible) therapy.
This is CNS activity we just reported. As we know with Retevmo, CNS activity is important to enable durable response and durable impact in advanced non-small cell lung cancer, developing drugs with CNS activity, you can imagine is a principle across all of our early phase programs.
And so this is the data then combining olomorasib with pembrolizumab. We reported at ASCO 64 patients combined pembrolizumab plus olomorasib but 2 drugs undergoing ongoing dose optimization development with pembro, 50 milligrams and 100 milligrams. This is efficacy data in the first line. You see a 77% response rate across a range of PD-L1 levels. You see a favorable median PFS. We acknowledge this is an early cohort, but nonetheless gives us enthusiasm that this is a regimen that can make a difference in first-line lung cancer.
And here is the tolerability profile. We are pleased to see that discontinuation due to treatment-related adverse events are infrequent. Fundamentally, this is a tox profile we think is consistent with first-line development motivating the SUNRAY-01 trial that I’ll tell you about now.
Jake alluded to this study earlier. This is a study that’s been designed for fundamentally Phase III (indiscernible) across a wide range of Phase III sites. It is a master vertical. Sites love this, right? You start with a dose optimization experience. When that is completed, the trial seamlessly transitions then to 2 parallel Phase III trials. We expect that transition to Phase III later on this year. Those 2 trials are meant to represent real-world practice in KRAS-positive lung cancers. You get a local biomarker result that comes back. You could either put that patient on to an immunotherapy-based regimen or a chemoimmunotherapy-based regimen based on neuro clinical judgment and their biomarker results. As Jake mentioned, this allows enrollment after one cycle of standard of care therapy. To give time for that patient to get started urgently on treatment, get biomarker results. We’ve got enormous positive feedback from sites of how that enables them to enroll effectively. We really think this is designed for rapid enrollment. It’s already active and across a wide range of global sites.
I’m going to move on to the 2 other drugs in the KRAS portfolio. First, KRAS G12D. KRAS G12D is the most common KRAS variant. It is more prevalent in GI cancers, especially colon cancer and pancreatic cancer. This is potential for a big impact. But the development of an orally bioavailable KRAS G12D inhibitor has been challenging. In our R&D unit, we have been able to learn from the development of olomorasib. And so through persistent iterative R&D, we have identified an orally bioavailable G12D inhibitor, a drug that has excellent PK properties so far as we can tell in our preclinical models and motivates us to get this quickly into patients. We think this can make a real impact, especially in GI cancers.
Here, you see the selectivity for G12D compared to other KRAS targets. On the right, you see activity across a range of cell line models. This drug is potent. This drug, we expect to be highly selective and our hope getting into the clinic later this year, we will quickly get to effective and tolerable doses, thus allowing the kind of creative combination regimens that we’ve seen, and we’ve been trying with olomorasib. So again, we can make an impact quickly here into the clinic later this year.
Finally, I’ll end with the pan-KRAS inhibitor. We realized that this is more ambitious. At the same time, this is 1 in 7 cancers. So if we can develop a drug across all of KRAS, there’s a potential for broad impact. There are a lot of ways to target KRAS. There is a unique thesis to this molecule, which is that we can target KRAS activating alterations, wild-type KRAS dependency, but spare a HRAS and NRAS. By sparing HRAS and NRAS, we believe we can get around the rash and other intolerance that other multi-KRAS inhibitors have exhibited. And we see this already in preclinical molecules, making us hopeful that this drug will be able to get to potent effective doses without the toxicity challenges.
Again, on the left, you see selectivity for a range of G12 and G13 mutations, activity in wild-type KRAS dependency, but sparing HRAS and NRAS motivating our thesis and on the right activity across a range of cell line models. Again, this is also going into the clinic later this year. Our expectation is this drug will have a slightly more measured dose escalation in a slightly more narrow therapeutic index as compared to the selective G12D inhibitor. But nonetheless, with this KRAS portfolio, we think we’ll be positioned to make an impact across a wide range of cancers.
And with that, I’m going to hand off to under to Dr. Arjun Balar.
Arjun Balar
Thanks so much, Geoff. So my name is Arjun Balar. I’m a medical oncologist by training, much like my colleagues. And after 10 years in the clinic at NY Langone Health leading the GU Cancers program there and also the clinical trials office. I was really thrilled to join Lilly about 2 years ago to lead development programs across a variety of different modalities.
I want to focus on 2 programs, LOXO-435 and also our Nectin-4 targeted ADCs which are really poised to change treatment options for patients with metastatic urothelial cancer and which is a personal area of research interest for me.
Let’s jump right in. So LOXO-435, this is our isoform-selective FGFR3 inhibitor, A little bit of background. FGFR alterations are activating are present in about 15% to 20% of patients with advanced urothelial cancer. There is an approved available agent, sertofitinib, responses and also a survival benefit against single-agent chemotherapy. However, it comes with a little bit of baggage, a lot of toxicities. These include things like alopecia, fatigue, hand-foot syndrome, diarrhea, ocular toxicity. And these are due to the fact that it’s essentially a pan inhibitor. There are other isoforms of FGFR that we need to know about, such as FGFR4 but in particular, FGFR2 and FGFR1. Targeting these off-targets is what drives these toxicities.
So our core hypothesis here was that let’s develop a truly isoform selective molecule, focuses on the target of interest, and avoid those other targets that we really don’t like. That enables us to have a better safety profile but also captures the full benefit of patients receiving therapy, staying on therapy long enough and not needing dose reductions. These are some of the challenges that patients on erdafitinib have so that we can actually maximize the benefit.
We’ve been in the clinic since ’23, early 2023. But I want to share with you a little bit about the preclinical profile that gives us a sense of how this drug will perform in the clinic.
On the left is an enzyme innovation assay that measures IC50 and looking at nanomolar concentrations. The first 4 rows there, just to make things simple, are representative pan-inhibitors, erdafitinib, obviously at the top. The nanomolar concentration, they’re essentially about the same, and that really defines what a pan-inhibitor is.
At the far bottom, you’ll see LOXO-435. At 0.3 nanomolar, you have exquisite potency against FGFR3, but then if you look at the other concentrations, we’re obviously very much sparing those other targets that we don’t like.
On the far right, you’ll essentially see how we look at selectivity, which is basic arithmetic. Inhibitors are doing what they should be doing, which is really nonpreferentially targeting FGFR3 and the other isoforms, but we’ve shown exquisite sensitivity in these in vitro models — I’m sorry, selectivity in these in vitro models for FGFR3, over FGFR1 and also FGFR3 and FGFR2.
The last thing I’ll mention is that middle column, FGFR3 V55M. What is that? So this is an acquired gatekeeper resistance mutation. It develops in the context of progression on pan-inhibitors such as erdafitinib, and these drugs lose their effectiveness and best represented in the IC50s there. We maintain potency even against the gatekeeper resistance mutation.
Pharmacodynamically, one of the things that we measure are things like hyperphosphatemia. That’s actually specifically due to FGFR1 engagement, erdafitinib comparator causes hyperphosphatemia in preclinical models and all the dose ranges that we tested showed no hyperphosphatemia for LOXO-435. In the middle you’ve got representative examples of tumor growth inhibition, what we expect to see preclinically.
I’m excited to report that after a little over a year in the clinic, we’ve hit therapeutic exposures, showing really nice responses and also a safety profile that’s consistent with our thesis.
I’ll continue on with our Nectin-4 program. Nectin-4 is a well-validated target in metastatic urothelial cancer, but also some other solid tumor types that we’re learning about.
(indiscernible) demonstrated meaningful transformation in the metastatic urothelial cancer in particular, when combined with KEYTRUDA has displaced platinum-based chemotherapy in the first-line setting. But what we know is that treatment resistance will develop, so patients will need options in the post Padcev space. We’ve shown preclinically, and I’ll talk about this in our 2 programs that the primary mechanism of resistance is actually the payload, primarily through drug efflux pumps. And Nectin-4 remains the most important target in urothelial cancer even after progression on Padcev.
We have 2 programs that are entering the clinic, and you’ll see them on your bottom right, LY4101174 which I’ll call 1174 for simplicity and then LY4052031. I’ll call that 2031.
1174, we acquired last year as part of our acquisition of a Biotech in Western Europe called Emergence. They were developing antibody drug conjugates with technologies that we really liked. And internally, by coincidence, we happen to have an internal program that looked very similar, but add some unique differences that I’m going to walk you through.
Let’s zoom in on these 2 programs. On the left, you have 1174. And on the right, you have 203. What’s common? IgG1 antibody with an Fc gamma receptor silent structure here primarily to avoid things like drug antibodies. And let’s talk about where things are different. And also actually, both are DAR8, highly homogeneous and in vitro studies, we’ve shown excellent linker palo stability.
But what’s different here? On the left, it’s a novel (indiscernible) linker, cleaved by beta-glucuronidase. This helps us concentrate this drug more in tumors. The payload, however, is tried and true. It’s exatecan well-known TOPO 1 class inhibitor.
And on the right, you have 2031, a conventional GGFG-linker, cathepsin mediated cleavage but a novel payload (CAMP-98). It’s not been tested in the clinic, but preclinically has shown higher CD and perhaps more potency. You can see, we like both assets. And this reflects our conviction in this target and putting this in the clinic.
I’m excited to report that 1174, we treated our first set of patients, and we’re proceeding through escalation. And with 2031, we just received our safety proceed letter from the FDA, and we’ll soon treat our very first patients later this month.
So I’m going to hand it off to Barry, who’s going to do his best Mariano Rivera and close this out.
Barry Taylor
So all downhill from here with that introduction. Good evening, everyone. My name is Barry Taylor. I am the Scientific Officer of Oncology at Lilly. I joined Lilly in the summer of 2020, having served as an academic faculty member, running a translational cancer research laboratory first at UCSF and then for many years at Memorial Sloan Kettering Cancer Center in New York City. It’s a pleasure to be here. I guess this is technically the anchor leg of our teams relay tonight. And I’m looking forward to walking you through a few additional clinical stage projects that are emerging from our portfolio this year.
The first of these is our selective SMARCA2 selective inhibitor program. This is actually a collaboration with a partner, Foghorn Therapeutics. The thesis of this program is to exploit the so-called synthetic dependency in cancer cells. So what do I mean when I say that, SMARCA2 and it’s closely related partner SMARCA4 together comprise the catalytic engine that drives a large multiprotein complex called the BAF complex. And the BAF complex is what rearranges and remodels chromatin to facilitate gene expression in every cell of our body, but tumor-specific loss of that partner SMARCA4, which arises in 10% of lung cancer through loss of function and truncating mutations forces a lung cancer cell to then rely on SMARCA2 to meet the BAF complex function. And that creates a therapeutic opportunity for us, if we were to be able to engineer potent and selective SMARCA2 inhibitor, we could exploit a synthetic lethality created by SMARCA4 loss. And I say selective because the inhibition of both SMARCA2 and SMARCA4 is not particularly well tolerated normal cells, and we’ll get back to the selectivity piece in the second, but this synthetic lethality is a debt to the pioneering work of PARP inhibitor therapy in patients with BRCA 1 and 2 mutations, a very similar concept. In fact, over the last, say, 10 years of academic research, cancer genomic profiling as well as functional genetics SMARCA2, SMARCA4 synthetic lethal relationship is among the strongest associations in all of (indiscernible). So we’re came to test this mechanism in patients.
As such, we developed a molecule called LY4050784, again with our colleagues at Foghorn Therapeutics. This is an oral SMARCA22 selective inhibitor. It has about a 30-fold margin over SMARCA4 in vitro. And so it demonstrates potent antiproliferative activity in SMARCA4 mutant, but not SMARCA4 wild-type lung cancer. Cell lines on the right, you’re seeing just an example of that, we’ve now profiled this in vitro over a much larger panel of SMARCA4 mutant lines that really represent the mutational diversity of human lung cancers.
And most exciting of all is that this result has translated in vivo. And I want to spend a moment on this slide to give you a historical perspective. These are 3 xenograft models of lung cancer, (indiscernible) models with lung cancer. And on the far right, we see something we actually saw a lot of is we were canvassing the academic literature and the work of our peer companies when we embarked on this project, which was tumor growth inhibition. But we really embarked on this journey not knowing if we could deliver on the kinds of (indiscernible) tumor regressions but suspected that if we were able to engineer the right molecule at the right properties, that would be on the table, which you see in the 2 in vivo models on the far left and in the middle, these are 2 SMARCA4 mutant lung cancer models where we can observe single-agent reverses at multiple dose levels. And actually, these are quite aggressive models of the human disease. Multiple of these models have co-mutations like p53, KRAS and actually maybe most notably, STK11 and KEAP1 mutations, which actually define a particularly poor prognosis set of lung cancers and one with particularly poor outcomes to immunotherapy containing regimens. So with these attributes and molecules excellent oral properties, we’re excited to test this clinically starting later in the year.
And this was prior to place in our larger portfolio of mechanism-based therapies in lung cancer is typified by the mutant selective and isoform selective KRAS inhibitors that Geoff was a little bit earlier.
So switching gears slightly and entirely consistent with something Jake mentioned at the front, which was a very deliberate path toward therapeutic modality diversification in the oncology pipeline. We’re incredibly excited to bring radiopharmaceuticals into the portfolio in oncology at Lilly.
This began with in of POINT Biopharma, a little less than a year ago. This is a deeply experienced team with a current clinical presence of next-generation molecules, which I’ll talk about in a second but also deep radiochemistry experience, a robust isotope supply chain as well as internal commercial scale manufacturing capabilities, and we supplemented that subsequently with our recently announced partnership with Aktis Oncology, which is to bring a novel targeting moiety to the development of RLTs, and we’re excited about that as well.
But we’ve also devoted a substantial effort internally in our own discovery labs for the development of RLTs. And this helps let us take advantage of the deep experience we have in structure-guided small molecule design as well as to tap into the broader Lilly capabilities, be therapeutic peptides or otherwise.
Of course, the thesis with radioligand therapy is highly tumor-specific targeting of radionuclides. And the beauty of the thisteronostic approach, which payers an imaging agent with a therapeutic agent, is if you can see it, and you can deliver high-dose financing radiation locally, those cancer cells will die. And that’s because this is fundamentally not indexed on the often intractable nuanced and heterogeneous biology of individual cancer cells.
And so this process is schematized here on the slide. On the far left is a patient with advanced metastatic prostate cancer whose tumor expresses PSMA. If you take a PSMA ligand and through a linker and a chelator deliver an imaging agent, be it gallium or otherwise, you can do PET imaging baseline and identify, in this case, widespread metastatic disease.
But now if you replace that imaging agent with a therapeutic isotope, leveraging the exact same PSMA ligand as you’re targeting agent, you can deliver potentially transformative antitumor activity in the patient as indicated on the far right, while really sparing normal healthy organs. It’s this kind of result that got us excited and really built our conviction in the therapeutic modality, and we’re keen to work as part of a community to really develop radiopharmaceuticals as a pillar of cancer care across therapeutic indications in the future.
And so I was mentioning about a current clinical presence. This is our active Phase I program called Excel. This is currently enrolling in Canada, though we hope to bring it to the U.S. quite soon. This is testing a novel PSMA ligand to deliver an alpha-emitting actinium isotope. So this is a shorter range but higher energy isotope.
The fundamental thesis with this program is really one of biodistribution because in prostate cancer, unfortunately, these aren’t the only cells in the body that express PSMA. In fact, PSMA as expressed in the salivary gland. And what we and others in the community have seen is significant off-target uptake in the salivary gland of first-generation PSMA targeting radioligands. And so the question here was, can we develop a PSMA ligand that not just improves its affinity and leads to increased internalization, but fundamentally increase the tumor uptake of the radioisotope, thereby sparing salivary and renal toxicities.
And so here is just a summary of those key characteristics. PSMA2 is at novel ligand juxtaposed here to 2 additional first-generation PSMA ligands, both I&T, which is part of the lutetium-based .2002 product as well as PSMA-617, which is the ligand in (indiscernible). And here, this just encapsulates what I was mentioning before of improved affinity, greater degree of internalization leading to a significant increase in tumor uptake in what we hope will be reduced normal organ uptake. And so we’re incredibly excited that this study is enrolling metastatic castrate-resistant prostate cancer is now though after initial safety data, we hope to expand enrollment to even earlier stage hormone-sensitive prostate cancers in the not-too-distant future.
But hopefully, this reflects an effort on our part to really build out a rich portfolio of RLTs at Lilly and really established this as a mainstay of oncology care for patients in need of transformative therapies.
So I just wanted to end and this settles back to something that Jake was describing at the beginning. We are enormously excited by the large number of clinical starts that we’re embarking on this year. But I think when you combine that with our advanced assets, we hope this reflects, and we believe it does a significant expansion in our R&D capabilities. Again, through the deliberate diversification of our therapeutic modalities and new technologies, it allows us to remain focused on the highest conviction areas of cancer biology, in disease indications in need of real fundamental transformative medicines.
And so to bring us back to something that Jake said, this big bet we took 4 years ago to refactor the oncology pipeline at Lilly. We hope this is at least an initial down payment on the output of that process, though we’re keen to continue that work with a rich discovery engine for years to come. So with that, I’ll give it back to Jake.
Jake Van Naarden
Thank you, Barry and team. Before we go into Q&A, I think when you look at this slide, you look at particularly the new clinical starts. In addition to, hopefully, we’ve convinced you about our conviction in the biology ideas here, but really, the theme that connects a lot of these that I think actually connects back to some of our initial successes with — that are now approved medicines is the idea of bringing what I think we were used to calling precision medicine to larger populations.
I think when we think about these great precision medicines in oncology, drugs like larotrectinib Vitrakvi from the original Loxo experiment, drugs like selpercatinib Retevmo, drugs like alectinib and at this meeting, even lorlatinib, these are medicines that have huge effect sizes for the patient populations that they deliver for. But unfortunately, for like the universe those patient populations are not large relative to the global impact that cancer diagnoses have.
And so we tried to learn from those efforts and say, how can we choose targets, how can we make medicines that can deliver that kind of benefit risk, but for larger populations and make a much bigger impact on cancer care.
So thanks, everyone, for joining tonight. I think we’ll now take some questions, and I’ll ask my colleagues to come back on stage. I think the Lilly IR team is sitting around with some microphones. And we’ll answer your questions. Thank you.
Question-and-Answer Session
Q – Evan Seigerman
Evan Seigerman from BMO Capital Markets. Thank you for the fantastic presentation today. I wanted to touch on KRAS G12C, really kind of diving into a little bit on not the commercial challenges of sotorasib and adagrasib, but mainly, what really gives you excitement about the profile of olomorasib, especially given the challenges we’ve seen in developing these 2 assets and really getting them to the front line, what’s differentiated about the profile? And how can you accelerate that development to get this to patients more quickly.
Jake Van Naarden
Geoff, do you want to take that?
Geoff Oxnard
Sure. I mean, we’re aware that there are a lot of these KRAS inhibitors, right? And so there are needs that are adequately addressed perhaps by some of the drugs. But there was a need every lung cancer don’t acknowledge is that first line, we give these treatments and they’re just unreliable, like sometimes the IO works and sometimes it doesn’t work. And sometimes chemo works 2 cycles and the patient progresses. And so I think in breast cancer, we’ve learned to like build and do more. And in lung cancer, that we’re starting to do it. So I think we, as lung cancer docs are ready to build and do more. And so these combinations are built to make the impact upfront, make that impact durable and fundamental bent the curves and get more patients on to the flat part of the curve where the durable from these drugs.
I think the trial is designed to enroll. I won’t deny that tolerability is the key barrier. And we’ve heard very clearly, this drug has overcome that barrier. We’ve identified efficacy, CNS activity, activity in previously treated patients and now tolerability that together allow us to combine with immunotherapy. We see them in our day, and that’s what motivates the first-line trial. I think it’s going to be — I mean, it’s a worthy experiment. We can enroll this trial, and I’m hopeful it will play out.
Seamus Fernandez
Seamus Fernandez from Guggenheim Securities. So just a couple of questions. Jake, when you think about partibrutinib and the sort of magnitude of the opportunity for that product, can you just give us a sense of kind of your vision and Lilly’s vision for how far that product can go and relative to some of the sales that we’ve seen in ibrutinib, Calquence, et cetera, where do you really see that product positioned?
And then just the second question is on the folate targeted ADC. I think we had the (indiscernible) fully targeted ADC, and it saw some fairly meaningful toxicity following a partnership with Bristol. Is there toxicity risk associated with full ADCs in general that you guys are trying to avoid? Or have you studied some of those molecules to really see what those issues potentially were? And so they really view that as a well-targeted therapies.
Jake Van Naarden
So yes, I can start with the Jaypirca commercial question. It’s interesting. We folks who follow that program have probably seen. We’ve been on a journey with this medicine that when we started thinking it was sort of a niche product in previously treated patients, maybe those only with C481S resistance mutations. And over time, just as we’ve grown — I would say, as we’ve gathered more clinical data, we’ve seen the medicine can be a lot bigger than that.
And so I think in our space, it’s probably true for like all drug development, but it’s definitely true in oncology. It’s pretty hard to project out to I think what will be the ultimate peak opportunity for any medicine in its early innings, and that’s really what this one is.
In this class, in particular, we’ve seen sort of early winners and later winners, and I think it’s still an evolving class actually with some of the recent data that’s emerged. So where does that put us? Well, as John talked about, we’re just trying to develop the medicine in all the right places with all the right combination partners to result in positive data readouts that are label-enabling so that physicians can use the medicine however they want. And that actually is increasingly a theme we hear from physicians when we talk to them about the medicine, especially now that they’ve had the chance to use it commercially, they just say, “Oh, I want to use it continuous dosing in the first line. I want to use it post BTK in the second line. I want to use it after BTK and BCL2.”
I think there are a lot of different potential ways this medicine can benefit patients. And there’s a lot of variety, frankly, in how physicians both in the United States and elsewhere may want to use it.
Our job is just to enable that use. And I think if we can do that, this medicine to make a big impact for patients and probably be a meaningfully large product for the business.
On the full it ADC question, I may actually have Barry answer that because it sort of has a lot to do with linker payload design as much as anything else.
Barry Taylor
Yes, thank you for the question. I think what we’re learning and even as evident from today in the last several days at ASCO is that the devils in the details with ADCs. It’s really a 3-component system. And so is your question, is this really a fundamental attribute of the target? Is this toxicities associated with the release of the payload in circulation, not necessarily in an antigen-mediated way? Is this pharmacology related to the linker? I think all of those things for different products have sort of reared their heads, I think in many ways. And we’re integrating all of those lessons, not just studying molecules preclinically but even sort of the first principles by which we design our own medicines, really have to solve the ternary problem of those 3 pieces simultaneously. And so I think these different medicines are reading out the answer to your question in different ways, actually.
John Pagel
If you don’t mind, Jake, let me jump at too.
Jake Van Naarden
Yes, please.
John Pagel
We actually do think based on our preclinical data, of course, as well that this drug we have in development will differentiate itself from, I only hear from an ocular toxicity standpoint. It’s probably not because of folate receptor involving the eye, it’s really the payload. And I say that here, we’re differentiated with the tube summaries payload. But if you look at the MMA, payloads that are typically associated with ocular toxicity. And I think that will be probably the biggest differentiating factor. And as I said, our nonhuman primates, and we don’t see that type of toxicity. Of course, it’s not the same experiment as we see in humans. We’ll see, but we’re pretty confident that we’ll have a differentiated product there.
Geoff Meacham
Geoff Meacham of Bank of America. I had a couple for you, Jake. In the spirit of tonight, I wanted to ask you maybe a higher level, you have a lot of newer technologies, newer assets. You’re developing clinical trials in a novel way, right, in oncology. I guess the question is, what is the appetite for taking target risk to do through basic discovery and to go after novel targets at Lilly? Is that something that you would say as a priority or not?
And the second question is when you look at life cycle management for Verzenio down the road, I know you’re studying the SERD in combination, but what’s the appetite for looking at other CDKs like some of your other competitors are?
Jake Van Naarden
Yes. Sure. I’m going to start on that on the first one and then have Barry chime in, too. When we started a couple of years ago revitalizing the pipeline, I think one of the things we wanted to make sure is that the early wave of medicines had a higher probability than that which we inherited. And so part of the way to do that is to work a little bit more geared towards validated targets with what we think are better medicines. Coming from behind the things we didn’t talk about tonight are way less validated ideas.
The other interesting thing that we didn’t touch on tonight in the prepared section is the opportunity for novel targets, particularly in the context of radioligand therapies because those are not targets that you need to have any biologic role in the cancer cell. And that’s sort of a truism for ADC targets, too except for RLT, you can actually target interest out of their proteins. So there’s a whole suite of interesting ideas for targets that are overexpressed in cancer but actually have nothing to do with the cancer itself that if you can bring high-energy radiation locally to, you can actually kill the cancer cells. So the RLT portfolio actually is going to probably over time end up being very geared towards targets and ideas that like you don’t see walking around the halls of ASCO or AACR at all. But Barry, why don’t you should chime in further on the topic?
Barry Taylor
Yes. No. I mean, exceptionally well said. I think outside of the radio pharmaceutical target base, the pillars of cancer biology are unchanged. They’ve lasted for 25, 30, 35 years of cancer research experience, and that’s like partly why we see a lot of clustering around core target ideas. We are in our earliest phased projects taking on a little bit more target risk, but staying central to the sort of fundamental tenants of cancer biology for the non-radiopharmaceutical products in our portfolio.
Cancer is a little bit different, I think, than many other indications, even both at Lilly and elsewhere in this regard and there’s quite a bit of clustering around core tenants that we’re really planning the vast majority of our target work around.
John Pagel
And then on the CDK work, Lillian, you should probably chime in here because we’ve thought about this idea and looked at a lot of these programs.
Lillian Smyth
Yes. I mean I think that the post-MONARCH data, at least reinforce to us, right, the importance of CYCLONEs in HER2-negative breast cancer. I mean, I think, obviously, there’s a couple of others looking at CDK2, and CDK4 inhibitors. I think as I think about the development of those kind of molecules, there probably is potential to build upon what we’re seeing in post-MONARCH with CDK4/6 inhibition, there’s, by adding on some synergistic mechanism of action agents. However, the CDK4 piece is a little trickier in terms of development, right? Because you’re looking at potentially head-to-head studies against very potent CDK4/6 inhibitors like abemaciclib, which we already know is very CDK4 selective. So I think that really, it’s about integrating what the development path looks like for those molecules into decisions there.
Steve Scala
Steve Scala from TD Cowen. Jake, I believe consensus suggests Lilly Oncology will contribute a high teens percent of Lilly 2024 revenue, and that likely will be a single-digit percentage in about a decade. So a relatively small contributor to the overall company. You sit on the Lilly Executive Committee, so you’re privy to the 10-year plan. Is that likely how it plays out? Or are we underestimating Lilly Oncology or overestimating something else?
And secondly, Dr. Smyth, you might not have intended it this way, but you basically referred to the Novartis Kisqali NATALEE data as noise. Novartis has a pretty formidable data package. So what’s the weaknesses in it?
Jake Van Naarden
Why don’t you take NATALEE first?
Lillian Smyth
Yes. Okay. So NATALEE, obviously, it’s great for patients to generate more data with more molecules, even in broader population. So it’s a positive study and it’s great to see that data. When you sort of talk about, okay, what are the weakness as well, I mean, first of all, follow-up and maturity of data sets is crucially important in natural breast cancer. You’re talking about cure, which is clearly paramount, but you’re also talking about toxicities, of these therapies which are not insignificant. And so that benefit risk assessment is crucially important in the adjuvant setting. So maturity of data set, degree of follow-up crucially important. Clearly, there are differences between the monarchE study and naturally relative to that point of maturity of data sets. So that’s number one.
Number two, we need to evaluate the duration of the therapy, right? We know that 3 years of therapy is what was evaluated in NATALEE. The question is, is that needed is the additional year be, particularly for that high-risk patient population for which you already have abemaciclib available to you, is that additional year of therapy justified in that context. We need to see with more maturity of data, those curves are looking in that off-treatment period. We know that there are patients still on that therapy still on the study. So that’s important to see patients off-therapy and see how those curves are behaving.
The no negative group that was presented at ASCO represents a relatively small group that was enrolled to the NATALEE study. Clearly, they had high-risk features, but I think, particularly for that group, when you’re considering 3 years of deal adjuvant therapy, it will be very important to see a mature data set and how those curves look because we know there’s node negative, although they’re high-risk node negative patients, they do need longer follow-up to see how those events come in.
Kara Clinton
I would just add one other comment, which is I think that there is still a number of patients with high-risk breast cancer who are not receiving treatment today, and so in some ways, we welcome having another organization, we’re talking about those patients in the need therapy.
I think if you want to also look at the ASCO guidelines that were recently released, I think they were pretty clear about how they view all of the issues that Lillian just talked about but also the importance of the separation and deepening of the benefit over the course of time.
Jake Van Naarden
Your first question, Steve. Look, Lilly is at an incredibly interesting moment in time right now. I think we all appreciate that, and the health care benefit in portfolio are delivering for patients is extremely significant and position the broader company, I hope, for a lot of success over the next decade.
One of the knock-on effects of that is the financial strength of the company allows us to invest significant capital in the other disease areas that we focus in, including in oncology. And so when I sit around that table that you referenced,, the success that we’re having with the incretin portfolio makes me not just, of course, excited for what it can do for society and for patients, but it really affords us, I think, a unique opportunity sort of among our peer set to invest in oncology, in addition to neuroscience, in addition to chronic immunologic diseases. So I think as we think about accelerating clinical trials, doing things differently, taking more risk, putting lots more medicines into the patient into clinic all at once, a lot of the reason we’re able to do that actually is because of the success that the rest of the business is having. So I don’t know if consensus 10 years from now is right or wrong, but I think the success the company is having right now really is a sort of rising tide that list the boat of all the other therapeutic areas we invested in, including oncology.
Chris Schott
Chris Schott at JPMorgan. Just two questions for me. Maybe coming back to the POINT acquisition. How quickly can you move those additional targets forward as you think about kind of building out a suite of these opportunities? And at this point, do you have all those capabilities in-house? Or do we have to think about additional acquisitions to really get to the POINT you want it to be there?
The second one is just a bigger picture question, more commercial. There’s been a number of questions around the Part D redesign and some of the changing payer landscape. When I think about a drug like Verzenio or any oral kind of Part D potential medication, do you anticipate there’ll be greater efforts from the payer universe to manage formularies in oncology differently than the past? Or do you think this will still be a category where that’s less frequently occurring.
Jake Van Naarden
Barry, do you want to take the RLT question and then I’m going to have Winselow Tucker, who leads commercial for us in oncology, take your second question.
Barry Taylor
I think with regard to how quickly we can continue to move forward, RLT programs that are in the discovery phase of our pipeline, the intense focus we bring to all of the other therapeutic modalities to get them to patients in a competitive time frame, we apply to the RLT and several of those programs, depending on the specific question and feature of the molecule we’re optimizing for can be moved along quite quickly, actually. But I think it also gives us the opportunity to do something that perhaps radioligand therapy hasn’t enjoyed per se to date and historically, which is to bring real professional molecule creation to radiopharmaceuticals. We obviously have cut our teeth for many years, engineering medicines against really tough complex and nuanced TPPs to bind and inhibit targets to avoid off targets to solve challenging ADME and oral pharmacology, and we are really looking forward to bringing the kind of die in the wall professional drug development to RLTs but do it at a cadence that’s competitive. And to move these programs along, we I think, have every expectation we can deliver on exactly that.
And we do have the vast majority of the capabilities necessary in-house. I don’t think that necessarily precludes additional external innovation, which, as Jake put it at the very beginning, we’re constantly looking for typified by the recent Aktis Oncology partnership. Thank you for your question.
Winselow Tucker
Thank you for your question. I think when it comes to the Part D benefit redesign, there may be a desire to pay to more and do more managing. But oncology has been had a bit of a protected class in some ways. And so we see that continuing over some time, like there may be some effort, but I do think it’s still going to be one that’s going to be hard fought for a major change in that space.
Akash Tewari
Akash for Jefferies. All right. So I have a couple. In your earlier studies for your SERD, you had a 6-month ET cutoff you remove that in your Phase III study. I think if you look at some of the emerging SERD data sets, it’s clear that having patients on a CDK4/6 enriches for ESR expression. And then number two, you’re kind of kicking out some of these ETFs progressors. How much of that is a risk with your upcoming EMERALD study? And what’s your confidence that you’re going to be able to get both a wild type and ESR1 label?
Number two, when we look at the DESTINY-06 data, there’s this kind of question, well, what’s going to happen with HR positive going forward? I mean you’re showing a 13.2 benefit in HER2 low. There’s a 75% overlap between HER2 low and HR positive, do you feel like that’s going to be kind of a changing for your franchise?
Jake Van Naarden
Lillian, do you want to take a stab at both of those?
Lillian Smyth
Yes. I want to make sure I remember all of them. Okay. So first of all, with respect to minimum duration of prior therapy, yes, you’re right, we have a requirement in there in terms of number of months on prior endocrine or CDK4/6 inhibitor nor did the EMERALD study. So I think that we are in a position still, particularly given the sample size that we’ve designed here, 860 patients enrolled, I think we will be in a position to be able to appreciate that benefit robustly.
And to your point about ESR1 mutation acquisition, I think we expect that it should be in that range of around 30%. And as I mentioned, that end point is one of our primary end points. So I think that positions us well in terms of our statistical design to be able to see a positive study. The next question versus about…
Jake Van Naarden
(indiscernible) versus ITT.
Lillian Smyth
Yes. Okay. So I mean, we — I think it’s, obviously, we have to look at the data sets that have been generated thus far and across other programs, including the approved agent. There does appear to be an enrichment to benefit in the ESR1 mutants. Having said that, we would also hope that we can demonstrate at least that this agent is also equally efficacious without added tolerability issues in the wild-type patient population. We appreciate that, that was not the case in EMERALD and that restricted label occurred for the ESR1 mutants. However, we feel and having spoken to a lot of physicians and patients about this, that, that alternative route of administration is crucial for patients. We know that they want to be able to take medicines at home. So I think, obviously, we need to see the data to enable that risk-benefit argument with the regulators, but certainly, that is, I think, an important attribute.
With respect to then, I think you had a third, DESTINY.
Jake Van Naarden
Let me I just want to chime in on EMBER for a second. Look, I think there’s no question that seeing benefit over fulvestrant in the ESR1 wild types is a riskier proposition. I think we know that from all the other studies that we’ve seen. But over the past couple of years, in particular, we’ve had a lot of conversations with society about like access to medicine, access to care equity in the context of that. And yet we’re demanding superior efficacy for an oral agent that patients can take at home versus a painful intramuscular injection that requires extra visits to the health care office, hours of lost time, lost wages, et cetera. I don’t think that we, frankly, as a society should be looking at this through conventional lenses of superiority on PFS end points for methods of administration that are so radically different. So we’ll see what the data show, but we hear consistently from physicians that the oral route of administration is valuable even in the event of the same PFS, and I don’t think that’s what we’re going to show either, but it just gives you a sense for sort of how the treating world is thinking about this.
And Lillian, you want to take the D-B06 Yes. I mean it was great to see the D-B06 data. Obviously, collectively, for T-DXd, the D-B06 and D-B04 data tell you that you can offer this therapy for patients in that first- or second-line chemo appropriate. And I think that’s the point. I don’t think that the community will swing from maximizing and optimizing endocrine and oral therapy options for patients. We know it’s important that they don’t want to jump immediately to infusional therapy nor in fact, do the data tells that they need to do that. I think that we’re seeing in that first-line chemo appropriate setting, obviously, the patient population that was enrolled to D-B06 had a median number of 2 prior therapies. So most of these patients had obviously received, all of them have received a CDK4/6 inhibitor and in fact, more had received a second line of endocrine therapy.
So I do think there’s opportunity for us to build in that second-line post CDK setting, post-MONARCH is the start. Hopefully, EMBER-3 builds upon that. We see nice data with imlunestrant in combination with abemaciclib. And I think that, that second-line setting is really where we need to intensify for patients to delay the need for infusional therapy.
Unidentified Analyst
Maybe just a follow-up on EMBER-3, I guess, at ESMO last year, we heard about the diarrhea profile overlapping with Verzenio. So maybe just confidence in the tolerability profile as you think about EMBER-3 and then the adjuvant setting.
And then the second question is just if EMBER-3 only shows a benefit in the ESR1 population, what does that imply for EMBER-4 in terms of your confidence level?
Lillian Smyth
Thanks for the question. So with respect to the tolerability profile of imlunestrant in combination with abemaciclib, we’re pretty confident. We’ve generated a relatively robust Phase I data set with at least 80 patients that have received the combination. And when we looked at the comparative adverse events in those patients compared to the MONARCH 2 population, it certainly appeared no worse. So our expectation is that imlunestrant will not have additional significant toxicity over what we see with fulvestrant abemaciclib. So from a tolerability perspective, we’re confident that, that will play out well in the Phase III setting.
With respect to your question about ESR1-mutant restriction. So I mean, I think that it’s important to recognize that early breast cancer and advanced breast cancer do have meaningful biological differences. Obviously, ESR1 mutations are not seen in the early breast cancer setting, but ESR1 mutations are surrogates for EOR dependency. And we know that most year positive breast cancer is EOR dependent. So I think that the question still remains relevant regardless if the label or study is confined to an ESR1-mutant benefit. I still think that, that has important and relevant read-through to the adjuvant EMBER-4 setting.
And remember, in December 4 study, we’re evaluating patients that have already demonstrated a degree of endocrine sensitivity by definition, they’ve gotten past that 2 years of therapy initial adjuvant therapy. So I think the question still remains relevant and important. And also recognize that the EMBER-4 study is a different comparator arm to EMBER-3. The hurdle is to be improving upon standard of care adjuvant endocrine therapy, not fulvestrant, which is obviously at the comparator EMBER-3.
Dave Risinger
Dave Risinger from Leerink Partners. So Jake, I was hoping that you could talk a little bit about post-MONARCH in terms of the commercial opportunity, when you think guidelines might change and when we might see that added to the labels globally?
Jake Van Naarden
Yes. So I could start and then Kara and Winselow should really chime in. The idea behind the study was not really motivated by commercial forces, so to speak. The medicine is already labeled in this population. And a lot of physicians have actually already been using the medicine for years now sort of in this way, and they can because it is labeled here.
This was really an evidence generation question that we just had been hearing for years that physicians wanted an actual robust Phase III trial. And of course, now within imlunestrant, it complements that program as well. So it’s just — it’s not, it’s not been a main motivation. I think the feedback we hear at this meeting is that the results in many ways are what docs expected to see from this regimen. And so for those who are already doing it, they sort of feel validated in that approach. And for those who maybe weren’t doing it, maybe they feel like they can have more comfort doing it now that there’s Phase III randomized data. But I mean, Kara, you should talk about guidelines.
Kara Clinton
Yes, I think a couple of things. One is, I think that people still are largely unsatisfied for patients that don’t have a biology-driven cancer and second-line metastatic breast cancer. So I think the enthusiasm has been relatively high, and people were really interested in answering this question in a randomized Phase III setting.
I think overall, we haven’t — overall, we would anticipate that this information will be submitted to appropriate guidelines, and I would expect because it’s a positive study with a PFS benefit of 27%. So that will be included in the guidelines and then we’ll go through the categorization process for NCCN and others, right? What Jacob said, I think currently today in most markets, not only in the U.S. but in the world, if people are choosing to do this, it’s within a labeled indication. So we — I wouldn’t say there’s a significant amount of commercial upside.
Jake Van Naarden
We’re at time. All right. All right. Thanks, everyone, for coming tonight. Hopefully, we gave you a flavor of what we’ve been up to over the past couple of years. And as you can tell, like we still have a lot more work to do. We want to see these medicines start working against the profiles that we expect for these specific patient populations. And hopefully, we’ll be doing one of these events in the near future. We can show you some of those emerging clinical data. So thanks, everyone, for coming.