Updates from PTC Therapeutics and uniQure on their huntingtin-lowering trials

On 21st June, both PTC Therapeutics and uniQure shared data from their respective clinical trials, both testing huntingtin-lowering as an approach to treat HD, but with different types of therapies. In this article we go through the data they each presented, what it all means and the next steps the companies will be taking.

Treating HD with huntingtin-lowering

Both PTC and uniQure’s approaches to treating Huntington’s disease draw on the basics of HD genetics. A gene called huntingtin becomes expanded, leading to an extra-long protein that is thought to damage brain cells. Dozens of pharmaceutical and biotech companies are working on therapies that try to decrease the amount of that long, faulty huntingtin protein, an approach known as huntingtin lowering. PTC and uniQure are two such companies which have ongoing clinical trials in this area although their approaches are quite different.

PTC: getting at genes by popping a pill?

Most huntingtin lowering drugs in development are aimed at the middle-man between the gene and the protein, a genetic messenger known as RNA. PTC’s drug, PTC-518, performs an intricate cut-and-paste step, so that now the middle-man is holding a stop sign. The cell’s machinery sees the stop sign and decides not to move forward with making the protein.

This type of drug is known as a splice modulator, and one major benefit of this approach is that it can be given by mouth. Based on data from animals we know that PTC-518 taken orally can reach many parts of brain and body without invasive procedures like a spinal injection or a brain surgery. PTC-518 targets both the expanded and regular forms of huntingtin so both versions of the protein are lowered following treatment with this drug.

uniQure: one shot therapy to lower huntingtin, forever?

We’ve written several times before about uniQure’s unique approach to treating HD – the first of its kind. Gene therapies create a fundamental change to a person’s genetics to try and treat or cure a disease. Although still targeting the genetic message molecule, uniQure’s approach with their drug, AMT-130, is quite different to PTC’s.

AMT-130 is a piece of man-made genetic material, packaged inside an empty, harmless virus, delivered to the deep parts of the brain via a surgery. The idea is that this one-time procedure will allow the therapy to spread into many brain cells, setting up little factories that continue to produce a genetic “antidote” for many years to come. This should prevent the huntingtin RNA message from producing so much huntingtin protein, in each brain cell that AMT-130 enters.

Updates from PTC

A pivot for PIVOT-HD

HDBuzz reported on the start as well as updates to the ongoing trial of PTC-518, so let’s recap. This roughly 3-month trial was planned to involve around 160 participants at sites throughout the US, Canada, Europe, and Australia. Participants would receive placebo or PTC-518 by mouth at a few different doses (5 mg or 10 mg), and visit study sites for evaluations related to safety, side effects, huntingtin levels in blood, and tests related to their movement, mood, and thinking abilities. Those who completed the study would have the option to enroll in an “open-label extension,” in which everyone receives PTC-518 and continues to have study visits periodically.

One unique aspect of this trial is that it was designed for people with very early signs of HD, potentially even before they are experiencing movement symptoms or major changes in their day-to-day abilities. But partway through, PTC announced a few changes. They decided to expand the study to include people with measurable movement symptoms and early difficulties with daily tasks, sometimes known as “manifest HD”. Additionally, they extended the drug trial period from 3 months to 12 months. Because of this lengthening, there were delays in getting approval to move forward from the USA’s Federal Drug Administration, so recruitment for the trial was paused in the United States but continued as planned elsewhere. We wrote more about these announcements in November of 2022.

Data shared from PIVOT-HD shows PTC-518 lowers huntingtin levels

At the same time that PTC shared these changes to the trial, they announced that they’d share data from the first, 3-month portion of the study in the first half of 2023. A helpful tidbit: when a company announces their intention to deliver news during a particular time window, it’s usually the case that they share in the latest part of that time window – in this case, late June 2023.

PTC held a meeting with investors and issued a statement sharing their findings from the PIVOT-HD trial to date. One of the key findings at this stage was huntingtin levels were reduced in people who received PTC-518, and that the group who received a higher dose of the drug, had a greater reduction in their huntingtin levels. This is positive news as it suggests that the effect on huntingtin levels is dose-dependent, i.e. the more drug you give, the greater the effect, so this will help guide future dosing strategies if this might need tweaking in subsequent phases of clinical testing.

The trial measured levels both of the genetic message and of the huntingtin protein molecule itself in blood samples from the participants. There was good agreement between these two measures which is what we would expect based on how the drug works by targeting the genetic message so this was an encouraging finding.

PTC-518 can move from the blood into the central nervous system

One common worry with drugs which are taken by mouth that are designed to treat brain disorders, is that it can be very difficult for these molecules to pass from the bloodstream to the central nervous system.
In the study data presented, PTC measured levels of the drug in the bloodstream and spinal fluid and showed that PTC-518 does indeed make its way into the spinal fluid that surrounds the brain. The balance of drug levels in the blood and spinal fluid was fairly equal which is good news although it doesn’t give us information about whether the drug is able to get to the regions of the brain important in HD, such as the striatum, that PTC hope to target.

Treatment with PTC-518 appears to be well tolerated

Following the disappointing news from the Novartis VIBRANT-HD clinical trial which tested branaplam, a drug similar to PTC-518, which was halted due to bad side effects, everyone was very keen to learn how the PIVOT-HD trial might fare in terms of safety.

The data PTC presented from this small study was encouraging, showing that no treatment-related serious adverse events occurred and that any minor side effects experienced by participants in the trial (e.g. headache) were found at equal levels in both treatment groups as well as the placebo group, suggesting they’re not related to the drug itself.

Another readout of brain health are the levels of a protein called NfL. Levels of NfL increase when the brain is sick and it is well documented that NfL levels increase in people with HD over time as their disease progresses. PTC measured the levels of NfL in the spinal fluid of trial participants and saw small decreases for people who were treated with each dose of the drug compared to placebo. This is good news as other huntingtin-lowering trials have actually seen spikes or increased levels of NfL. However, the data are fairly variable, and only come from a short duration of treatment, so it’s not yet clear how significant this decrease is until more people are treated for longer.

Updates from uniQure

The HD-Gene-TRX trials so far

Given the novelty of HD gene therapy, current clinical trials of AMT-130 are in the early stages and are focused on ensuring safety. Across multiple small trials in the USA and Europe, only around 40 people with early HD symptoms have undergone the surgery, up to 26 in the USA and 15 in Europe.

A low and a high dose of AMT-130 are being tested in this trial. A few of those were “sham” surgeries, with no drug delivered, as a comparison group. Participants are very carefully monitored for a couple of weeks after the surgery, and then followed closely for a year with less frequent visits up to 5 years. They go to study visits and complete blood work, neurological exams, and assessments of their HD, like thinking and movement tests.

Last June 2022, uniQure shared some early data from the first cohort of ten people in the low-dose group – they didn’t observe any major safety issues, and huntingtin levels, though only measurable in a very small group, were trending down.

Then, in August, some dangerous neurological side effects were reported following 3 of the high dose surgeries, leading to a short pause. These were resolved for all the participants, such that in November, new surgeries could again move forward with some extra monitoring in place.

What we learned today about AMT-130

Since last November, we’ve been waiting for more data from uniQure about the ongoing trial of AMT-130, expected in June. This release involves two years of data from the first, low-dose cohort of ten, and one year of data from the second, high dose cohort of sixteen US participants.

The good news from uniQure is that the NfL spike which occurs after the surgery to deliver the drug does seem to return close to baseline by about 18 months and no further increases are observed. The change in NfL levels compared to controls in the long run are not really clear just yet though. No significant changes were observed in total brain volume either which is positive.

In terms of symptoms, uniQure reported several measurements made in treated patients. This includes a set of assessments that takes into account many aspects of a persons movement, called the total motor score. Compared to the expected trajectory of these changes in movements, patients treated with AMT-130 seemed to be doing a little bit better over 18 months.

A measure called total functional capacity encompasses how people are doing in their tasks of daily life. Patients treated with AMT-130 appeared to show a stabilization of this measurement, which includes milestones such as continued work, ability to do household finances, etc. Consistent with that, formal tests of people’s ability to think flexibly also seemed to stabilize, compared to the expected trajectory for HD patients.

However, some of the data shared by uniQure are a bit confusing to make sense of. When they looked at levels of huntingtin in the spinal fluid, this was decreased in the low dose cohort but increased in the high dose cohort when looking at the cohort averages. This could be due to a highly noisy dataset across just a small number of participants, or perhaps some technical issue with the huntingtin level measurements, but that isn’t known just yet.

It’s worth remembering that with AMT-130, in particular, that extreme caution is required, and we in the field are straddling a tricky divide. On the one hand, we want to have larger numbers of people treated with AMT-130 so that we can see robust changes in the measurements being done on the subjects. But remember, this is a very novel gene therapy with a virus that cannot be shut off! And so uniQure and regulators have to walk a tightrope between enrolling enough people to generate robust data and patient safety.

The bottom line and what’s next

Following many disappointing trial outcomes from other companies testing huntingtin-lowering therapies in the clinic, it’s encouraging to see progress from two different approaches.

The data presented by PTC are broadly encouraging and show that the drug appears to be working to lower huntingtin levels, as intended, with minimal side effects. It’s important to note that this trial is currently very small – data from a total of just 22 people were reported in this particular update. How these findings might change in a study with a larger cohort remains to be seen. We also don’t yet know if the observed huntingtin-lowering will lead to a halting or slowing of symptoms in people with HD. PTC stated in this update that they will use the data presented in this update to argue to the FDA for the trial enrollment to be recommenced at the US sites where it was previously paused. They will also now continue enrollment in their European sites.

Whilst the uniQure data are not necessarily discouraging, they are frankly not clear cut either. This is often the case with small trials where variability is high between participants so trying to work out if a drug is having a desired effect can be challenging. uniQure plan to continue recruitment of their trial in both the US and Europe, the former of which will also investigate treatment of AMT-130 and steroids at the same time, to hopefully reduce some of the side effects they have seen with this drug.

We will keep you posted on all fronts as things develop.

Huntington’s disease therapeutics conference 2023 – Day 3

Welcome to the third and final day of HD science, live from Dubrovnik, Croatia!

Our Twitter updates are compiled below. Continue to follow live updates for the final day of the conference with the hashtag #HDTC2023.
Check out our coverage of Day 1 here: https://en.hdbuzz.net/343 and day 2 here: https://en.hdbuzz.net/344.

Biomarkers

This morning’s session will focus on biomarkers, things we can measure to get a picture of a person’s health or their response to a drug. Different types of biomarker measurements might focus on predicting onset, monitoring a person’s HD, or checking drug safety. As we heard last night, NfL levels can help us get a better picture of brain health, but there are other proteins also being studied for this purpose.

HD Clarity

First up is Dr. Niels Henning Skotte from the University of Copenhagen who will be telling us about his work studying biomarkers from HD patient samples. He uses samples from a large spinal fluid collection study called HDClarity https://hdclarity.net/. Niels is first talking about the importance of “quality control” in the spinal fluid samples – tests they do to ensure that they are uncontaminated and properly stored. He also presented some statistics to show how many are needed to answer different types of questions about HD. Many of the proteins which are potential biomarkers are only present in tiny amounts in patient samples.

There are special graphs called “volcano plots” that allow researchers to see which proteins found in spinal fluid differ the most between people with and without the HD gene. Some of the potential biomarkers even show differences between gene-negative and pre-symptomatic gene carriers, which could be helpful in the hunt for treatments that could be administered before symptom onset. When certain protein levels differ between people with and without HD in both blood and spinal fluid, researchers look more closely at them to understand how, and possibly why, their levels shift at different stages of HD. When a protein change is consistent across many people, then it may be considered as a useful biomarker of HD. The role of different proteins in the body is considered, and how they interact with one another, which can give us clues about biological processes affected in HD.

In this age of artificial intelligence or AI, scientists can feed large datasets to computer systems and ask them to consider complex sets of factors to determine which proteins would make the best biomarkers. Niels is using machine learning approaches to do just this. In the future, measuring changes across groups of many proteins prior to the development of symptoms might be used to better track exactly where a person is in disease progression or to decide when they should begin an HD treatment.

Fat molecules as a biomarker for HD

The next speaker is Dr. William Griffiths from Swansea University who will be telling us about how cholesterol and other fat molecules could be used as possible biomarkers for HD. William reminds us that about 25% of the body’s cholesterol is in the brain, and a lot of this is made on site. Some types are able to exit the brain, so we might be able to measure their levels to gain an understanding of brain health. Disruption of the cholesterol-making process and changes in levels of cholesterol have been observed in HD, and in fact there are drug development efforts focused on correcting these changes.

William’s work focuses on measuring the differences between cholesterol levels in people with HD and without HD to see if these molecules could be used as a biomarker. Measuring and analyzing cholesterols requires fancy biochemistry techniques. The specific molecules they are looking for are hard to detect even with top end equipment available, so they had to tweak the system using a cool technique called “click chemistry”. This boosts the signal of the cholesterol from grass-size to tree-size, as William explains. William’s group has found that one form of cholesterol, which is only generated in neurons, is diminished in blood samples from HD patients, making it a potential biomarker.

Somatic instability as a biomarker of HD

Up now is Dr. Darren Monckton from the University of Glasgow who will be telling us about his group’s research on whether some aspects of somatic instability could be a biomarker of HD.

Scientists can measure the levels of somatic instability of the CAG repeat part of the HD gene in all types of different patient samples. Darren uses fancy sequencing techniques to do this as accurately as possible in the DNA from blood donated by people with HD. The Monckton group has mapped out how the expansion of CAG repeats over time (somatic instability) changes at different rates in blood samples depending on the age of the person and their original CAG number.

They have also looked at blood samples from the same individual collected 7 years apart. This gives clues about how somatic instability increases in each person over time. Even over this huge timeframe, the changes are generally very subtle and happen slowly. Being able to measure these small changes is very important, because potential drugs which will alter the rate of somatic instability will also likely have very subtle effects.

These techniques will probably prove to be very useful in some of the clinical trials in the pipeline. HD is not the only disease that has somatic instability, and the techniques in development by the Monckton lab for measuring subtle DNA changes over time will be useful to apply to the study of other genetic diseases and corresponding treatments outside the field of HD.

Tracking Huntington with PET tracing

We are back from our coffee break and now we will be hearing from Dr. Mette Skinbjerg about a huntingtin PET tracer which allows tracking of the toxic clumps of protein in the brain which accumulate over time. We wrote about this previously here. Without a tracer, the only way we can see how huntingtin protein accumulates in the human brain is to look at samples after someone has died. Tracers are a safe way to look in living people and could be a great way to see how drugs might be working.

CHDI has been working with academic partners to make a tracer for HD which they have extensively characterised in many different HD animal models including mice and monkeys which allows tracking of the build up of protein clumps over time. Now they are moving beyond the animal models to test their tracer in people. Tracers are labelled with radioactivity so scientists can measure where they stick to the target – in this case the clumps. It’s important tracers leave the body after dosing so exposure to radioactivity is in the safe range.

Although the tracer seems to be safe to use in people, unfortunately the signal in the brain didn’t track with what the scientists expected. This is disappointing but this program taught us a lot about making a tracer for HD which can be used for making better ones in the future. Now the team is working on a new generation of tracers which they are hoping will perform much better. Things are progressing forward in the lab with lots of testing in HD animals so hopefully this next round will work better.

Biomarkers and machine learning

The next speaker is Dr. Peter Wijeratne from the University of Sussex. Peter’s group aims to use biomarkers and machine learning to characterise and predict HD progression in individuals. Very cool!

As researchers continue to identify all sorts of different biomarkers from biofluids, imaging, etc, many biomarkers for one person could be combined for better predictions. But combining and understanding all of this data together is hard for people to do, this is where AI can help us out!

Peter used a fun ChatGPT example to explain machine learning – an algorithm that can adapt and make inferences from patterns in data. He showed how quickly these systems can learn new information and make informed decisions – very cool! To “train” the AI system, you need lots and lots of high quality training data where scientists already know the answers. The AI system can then learn to spot patterns in this data, enabling it to spot similar and related patterns in test data where the answers aren’t known yet.

Peter’s group are looking at brain imaging data from three different studies which looked at how different brain structures change in HD over time. Training AI on these complex and rich datasets, they hope that they will be able to make robust predictions of disease. Turns out, good predictions for disease onset could be made, and the results agreed well with the HD-ISS staging system. They hope this will prove useful for making predictions at the individual level in the future.

HTT levels and tominersen

In the next talk, Dr. Blair Leavitt, a clinician/researcher from the University of British Columbia, will speak about his study of samples from the GENERATION HD1 trial of tominersen. He is diving deeper into how huntingtin levels change with tominersen treatment.

Blair starts by thanking HD family members who so generously and selflessly share biological samples with scientists to create a biobank resource. This is invaluable to scientists to understand HD and how drugs may change the path of this disease. Blair is focusing on one individual in particular, who felt very strongly about donating his brain when he passed. A rich data set and many samples are available from the tominersen trials he participated in, along with his brain tissue, which offers a rare window into drug effects.

Looking at the brain, scientists on Blair’s team were able to measure levels of the drug in different regions and compare this to the exposure levels predicted by earlier monkey experiments. Generally these showed the predictions were pretty good. Next they looked at huntingtin levels in different regions of the brain and how these compare with control brains. As expected for a huntingtin-lowering treatment, the levels in this individual were much lower than in controls. Disappointingly, the levels of huntingtin in the spinal fluid were too low to be quantified for this individual. This means we don’t know how well the brain and spinal fluid levels of huntingtin correlate, for this trial participant at least.

It’s hard to overstate how precious this tissue is for scientists to be able to truly examine in depth how a treatment has affected the brain. Collaborators in the audience will ask additional questions using this tissue so generously donated by this one trial participant.

Clinical trials

After a lunch break, we are back for the last scientific session of the #HDTC2023 conference. This afternoon’s session will be focused on clinical trials, and we’ll be hearing about study design and progress in human research.

Classifying HD stages

Dr. Jeff Long of the University of Iowa is speaking about the HD-ISS, a staging system for HD. It’s a clinical research tool that allows researchers to better classify people in the early stages of HD for more efficient trial design and recruitment.

Now that this new tool is in wider use, Jeff’s team is developing a database of information from large observational trials, like IMAGE-HD, PREDICT-HD, TRACK-HD, and ENROLL-HD, to better understand the timecourse of progression through HD-ISS stages. Having built these tools, Jeff’s team is working to understand how they’d be useful in a clinical trial – how many HD patients at a particular disease stage would be needed to generate robust findings to convince us whether or not a drug worked.

Since the HD-ISS incorporates data from brain images, biomarkers, and genetics, many variables can be considered to define which clinical measurements are the best ones to use to show whether a new drug might be working. Using observational trial databases, researchers like Jeff can apply statistical techniques to better predict how many participants and what types of assessments are likely to be needed to show the benefit of a drug. This is complex and important math that illustrates the importance of participation in observational research.

The proof is in the pudding (or the data)

The next talk is a highly anticipated one sharing the very early results of the PROOF-HD Trial of Pridopidine. We learned from press on Tuesday that the trial did not meet its primary endpoints, but now we are seeing the data.

Dr. Michael Hayden, the CEO of Prilenia, is giving this presentation. He is first explaining the mechanism of how the drug is believed to affect nerve cells, in particular its action on a type of receptor that facilitates communication between neurons, known as sigma 1.

The PROOF-HD trial was designed to use certain clinical assessments, a combination of movement, behavior, and thinking tests, to see if pridopidine helped slow the worsening of HD symptoms over the course of about a year and a half. Trials are designed with “primary,” “secondary,” and “exploratory” endpoints. Showing that a drug affects primary endpoints is usually how decisions are made to continue developing the drug and eventually to get it approved. This trial recruited fast, and most participants stayed in it the entire time, a testament to the mobilization and commitment of HD patients. When the vast majority of people continue in a long trial, it can also speak to the safety and tolerability of the drug.

The main overall finding is that the trial’s primary endpoints were not met. In this case it was a measurement of people’s ability to function day-to-day. On average, people taking pridopidine and people taking placebo functioned similarly for the duration of the trial. Another important endpoint was a combination of different movement, behavioral, and thinking measures, and these also did not improve for people taking pridopidine. Pridopidine might have shown some benefits in one movement measurement, called the Q-motor, but this was not statistically significant.

When a trial is designed, but before it begins, the sponsor (in this case Prilenia) has to make decisions about what types of statistics and analyses it will perform once the results are in. In PROOF-HD, Prilenia decided they would separate groups of participants into those taking drugs called neuroleptics (also known as neuropsychotics) and those not taking neuroleptics. This is because pridopidine affects some of the same biological pathways as neuroleptics. When Prilenia looked at data just from people who weren’t taking neuroleptic drugs, the potential benefit was more obvious using some clinical measurements, especially in the first year. Ultimately this also was not statistically significant.

Michael is now showing data from a trial of pridopidine in people with ALS, a disease which has some shared biology with HD. This trial also showed some potential benefits on secondary outcomes.

Prilenia believes that there remains some promise for pridopidine for treating HD, and the company will now focus on delving deeper into the data. They especially need to understand how different neuroleptic treatments affect response to the drug.

Red light for branaplam

Up next, Dr. Beth Borowsky, of Novartis, describes the results of their trial with a drug called branaplam in HD patients. We wrote about this drug and its surprising mechanism of action here.

While branaplam was safe in children with another disease (spinal muscular atrophy), some animal studies had indicated there was a possibility of damage in the nerves that project from the brain to the skin and muscles of the body. Based on that concern, Novartis included specialized experts in that kind of nerve damage amongst treated HD patients, just in case such a symptom emerged during the trial.

Unfortunately, over a few weeks, some subtle movement and lab measurements started to suggest that the feared risks had actually emerged. In consultation with their independent expert safety monitors, Novartis decided to initially pause dosing. At the time of pause the patients had gotten treatment from between 5 and 22 weeks. After careful review, they found that 78% of the treated patients showed one or more signs that could indicate nerve damage, and also some changes in brain structures called ventricles.

Based on a very careful analysis of the benefit and the risk for patients, Novartis made the decision to halt the trial in December 2022. We covered this here. Currently, all the participants in the trial are continuing to be monitored for symptoms of nerve damage and to track how that may change over time after they stopped taking the drug.

Beth has brought a snapshot of data that Novartis collected to bring the HD community up to speed on what they found. First – as hoped, branaplam lowered Huntingtin levels by around 25% in the spinal fluid, suggesting that branaplam was able to lower levels of Huntingtin in the brain.

Unfortunately, Novartis also found higher levels of a protein called neurofilament light, or NfL, a marker of unhappy brain cells. We’ve talked about NfL before, since it increases in the normal course of HD, and it was a big focus of the biomarkers session at this meeting. We’d hope that if an HD drug works, levels of NfL will go down over time. But in the branaplam study, Novartis discovered that NfL levels in the blood and spinal fluid increased with treatment. This is one of the findings that helped encourage Novartis to pause the study.

In parallel to these lab tests, physicians were carrying out careful nerve function studies in each participant. About 86% of participants had some kind of neurological symptom, and brain imaging showed larger fluid-filled cavities known as lateral ventricles.

All together, it seems like Novartis’s drug did what they thought it would do – reduce Huntingtin levels in the brain. Unfortunately, this was accompanied by serious side effects, so there is not a safe path forward for this drug. Importantly, Novartis is continuing to monitor the trial participants and to analyze data to inform drug development moving forward.

AMT-130 marches forward

Up next, Dr. Talaha Ali from uniQure is giving an update on their study of an HD gene therapy for HD called AMT-130. This relies on the injection of harmless viruses that carry instructions to teach brain cells how to reduce HTT levels.

The amazing thing about these viral gene therapies is that they theoretically require only a single injection, as the viruses persist in the brain for many years – perhaps forever. The downside of this is that it requires surgery to deliver the viruses into the brain. This surgical approach is being tested in two separate trials – one in the US and one in Europe. Because this is such cutting edge stuff, only small numbers of people are being included – around 40 patients in total.

Patients in the trial receive very careful, very slow, injection of the drug into different parts of the deep brain structures that are most impacted in HD. The trial is testing a low dose and a high dose of AMT-130, and closely monitoring participants for the first year and then more frequently for up to 5 years. uniQure will be sharing new data and updates soon, likely by the end of June 2023.

As previously covered, along the way uniQure had some concerning reactions in three patients. After careful examination, the independent doctors monitoring these symptoms decided that the risk seemed acceptable, and the trial was continued.

Excitingly, uniQure has some evidence that AMT-130 reduces brain levels of Huntingtin in the CSF – but to date, the number of treated patients is much too small to make accurate estimates. Hopefully more exciting data to come next time we hear from uniQure!

Narrowing the targets for tominersen

The next talk is by Dr. Peter McColgan from Roche, which is developing a drug called tominersen for HD. He’ll talk about the history of the program, what they’re learning from tominersen trials, and what’s happening with the ongoing GENERATION HD2 trial.

Ionis originally developed tominersen, a spinally delivered genetic drug called an ASO. In early, short safety trials, it was the first drug able to lower huntingtin levels in humans. This was followed by a very large trial to test effects on HD symptoms, known as GENERATION HD1. We learned in March of 2021 that GENERATION HD1 had been halted because of safety concerns – tominersen wasn’t helping HD patients, and at the highest dose it may even have been hurting.

Later, Roche dived deeper into the data, and found that some participants in GENERATION HD1 may have benefitted from tominersen, specifically those who started the trial at a younger age and with less severe symptoms. For this reason, Roche designed and launched the GENERATION HD2 trial, which is a smaller study testing tominersen in a younger population of people in the earliest stages of HD. This study is recruiting now and eventually there will be up to 75 sites in 15 countries.

Peter is now showing data on NfL, a protein that can serve as a marker of damage to nerve cells. New analyses of data from GENERATION HD1 show that giving tominersen at lower doses is likely safer based on lower levels of NfL. The GENERATION HD2 trial is testing two different, lower levels of tominersen, and mathematical modeling predicts that these lower doses will be safer because they will not lead to such large increases in NfL.

Now Peter is sharing new NfL data from the GENERATION HD1 trial. Towards the end of the trial, it actually looks as though NfL levels are going down with tominersen, which is further evidence that the lower doses being tested in GENERATION HD2 could have some promise.

Here’s Peter’s whole presentation on tominersen.

That concludes the research talks at the 2023 HD Therapeutics Conference. Thanks for following along, and visit http://hdbuzz.net to read summaries of Day 1, Day 2, and Day 3!

Huntington’s disease therapeutics conference 2023 – Day 2

Welcome to the second full day of HD science, live from Dubrovnik! After yesterday’s amazing basic science talks, today begins with a session focused on companies developing new experimental treatments for HD.

Our Twitter updates are compiled below. Continue to follow live updates for the rest of the conference with the hashtag #HDTC2023.

Check out our coverage of Day 1 here: https://en.hdbuzz.net/343 . We’ll post summaries in article format for each day of the conference.

HD therapeutic candidates

A unique approach to ASOs

The first speaker of the morning is Dr. Nicole Datson, from a company called VICO – they work with a type of drug familiar to many HD families, Antisense Oligonucleotides, or ASOs. Several companies, including Wave Life Sciences and Roche, are already testing ASO drugs in HD, but Vico’s approach is unique. VICO’s ASO directly targets the genetic mutation that causes HD – an expanded “CAG” sequence near the beginning of the huntingtin gene. This has very interesting implications, because HD is not the only “CAG repeat expansion” disease – at least 8 other human diseases are caused by the same exact genetic change, but in different genes across the genome. So, if the drug works, it could potentially be applied to any one of this family of diseases that are associated with the same genetic change.

Dr. Nicole Datson, Vico’s Chief Scientific Officer, gives an overview of Vico’s approach. Even the regular huntingtin gene has a long CAG sequence (~17-20 in most people without HD), so it’s difficult for these drugs to specifically bind only the disease form of huntingtin. But Vico suggests that while their ASOs targeting CAG don’t only recognize the longer form that causes the disease, it seems to prefer these longer CAGs, so there’s more impact of the drug on the expanded CAG repeat. Nicole is showing their data from HD patient cells after treatment with their ASO drug, snappily called VO659. Treatment with higher doses of the ASO leads to greater reduction of the expanded huntingtin protein, the bad guy of HD.

Excitingly, the same ASO also has effects in cells from patients with two other brain diseases involving CAG expansion, forms of ataxia called SCA1 and SCA3. In both cases, the ASO also prefers the mutant form of the gene with the longer CAG repeats. If it works, the cool implication of this is that a single drug could potentially work for a whole alphabet soup of diseases caused by CAG repeat expansions including HD, SCA1, SCA3, DRPLA, and SBMA.

Moving away from cells, Nicole shows that treating HD mice with ASO also leads to reductions in the huntingtin protein. The behavior and brain anatomy of the HD mice was also improved by the ASO treatment – very cool. Vico’s data shows that the ASO stays in the brains of animals for a long time, suggesting that they might be able to have fairly large intervals between treatments. This would be a big benefit for ASOs that require injections into the spine to reach the brain.

In mouse models of two other CAG-expansion diseases, SCA1 and SCA3, they saw similar benefits: the disease versions of the gene were switched off to a greater degree, leading to improvements in disease-like symptoms. VICO have also tested their drug in monkeys and saw that their drug spreads fairly well throughout different regions of the brain and stays in the brain for a long time, same as they saw in the mice. Based on their animal data, Vico thinks that for both HD and the other diseases they could inject their drug as infrequently as a couple times per year. This would make treatment much easier on families, making it easier to participate in the trial. Given these promising animal and cell studies, Vico launched an early stage human study in patients with HD, SCA1, and SCA3 – very cool they’re targeting these conditions all in one “basket” trial.

RNA interference

Up next is Dr. William Cantley, a researcher at Alnylam Pharmaceuticals, a leading company in the area of “RNA interference”, also called RNAi. This is a totally distinct chemistry from ASOs, but has a very similar goal – to reduce levels of a target protein. In theory RNAi is more potent than ASOs, meaning you need less drug for the same effect, but to date ASOs are being widely used in brain diseases. Part of the problem is delivery – how to get RNAi drugs to the 84 billion brain cells that might need to get treated in HD. Getting RNAi drugs into other cell types, such as the liver, has been improved by sticking little molecular keys onto RNAi drugs. When cells in the body have a matching lock on their cell surface, they can take up RNAi drugs much more easily. Alnylam has a lot of experience with these lock-and-key tricks to get RNAi drugs into liver cells. But treating the liver is less important in HD. So Alnylam has developed a new key, “C16,” that unlocks important brain cell types that we care about in HD.

William shows very cool data in another brain disease, Alzheimer’s, in which tagging an RNAi drug with C16 seems to work very well to get into the brains of monkeys. William is announcing – for the first time publicly – that Alnylam is working on an RNAi drug for HD using this cool new C16 key approach.

After walking the crowd through Alnylam’s strategic decision process, William gives an overview of some early work in HD mice with a new C16-RNAi drug. Using two different drugs, targeting different parts of the HD gene, they see reduction of huntingtin protein. In addition to these HD mouse studies, Alnylam also conducted studies in monkey brains which show very big reductions of HTT protein levels in the cortex, the wrinkly outside part of the brain, that’s affected HD. Exciting to have another arrow in the quiver of HTT lowering drugs, as RNAi drugs and ASO drugs have different risks and benefits. We won’t know until we’re done which of these approaches provides the most benefits, so great to have another runner in the race.

Making a huntingtin protein degrader

The final talk before the first coffee break of the day is from Adam Hendricson, who works at Arvinas Operations. Adam will be telling us about their work to make a huntingtin protein degrader – a small molecule drug which can reduce levels of huntingtin by sending it to the cell’s rubbish bin.

In recent years, there has been a scientific explosion in a new kind of technology focused on breaking down disease proteins with small molecules called PROTACs. This is exciting as these drugs COULD be taken by mouth if they work out. PROTACs work by bringing a rubbish-labeling protein in our cells into close contact with a target protein, in our case the huntingtin protein. This would lead to huntingtin being tagged in a special way that tells the cell to treat it like trash. Arvinas is one of the first companies to bring this technology into a clinical trial. They are currently testing one of their drugs to treat cancer, showing it is possible for this new approach to be more than just a cool tool for lab scientists. They are working on a number of different brain diseases including Alzheimer’s, Parkinson’s, and HD.

Arvinas are hoping to target the “soluble” huntingtin protein, the form that precedes the formation of the more solidly structured protein clumps. They are also looking for drug molecules which have preference for mutant over normal huntingtin protein. In cells in a dish, they show that the PROTAC they have identified binds both the mutant huntingtin protein, and the trash-labelling protein. These molecules bind very tightly which is what drug-hunters like Adam are looking for!

The PROTAC molecule from Arvinas can in fact lower the levels of both the soluble and clumped form of the expanded huntingtin which is good news, as scientists think reducing levels of both of these proteins would be beneficial in patients. Most of this work so far has been experiments in cells in a dish but Arvinas are now starting to test their molecule in mice. The good news is that it looks like the PROTAC is able to get into the brain, which is often very challenging for drugs to do.

Tracking the superhumans

Next up we’re hearing from Dr. Donna Finch of Alchemab Therapeutics. Their focus is targeting the immune system, using antibodies that could potentially be protective to brain cells in HD. Their approach is to find naturally occurring protective antibodies in patients who are resilient i.e. very long-lived or less affected by disease genes. Then they do all sorts of screens to figure out why their antibodies are protective, and go on to test the best ones in cells and in animals.

This is like tracking down the natural “super humans” among us and working out why disease seems to affect them less than most people, and using that information to help treat others. Alchemab has come to this conference to find out how they might use their approach for HD – very exciting to have another company come to work on a new avenue of therapeutics! They collaborate with organizations across the world that have helped them to gather samples from people with different diseases and vulnerabilities. They have compiled immune system info into a “data cube” that allows them to do large scale analysis across many individuals.

We are living in the age of BIG data! Companies like Alchemab use absolutely huge data sets and lots of clever computer tools to find small signals in the data which could uncover new biology. They’ve found that antibodies against the huntingtin protein are generated in resilient individuals, even in other diseases like Alzheimer’s. An antibody has been identified by Alchemab, called ATLX-1095, which binds to a fragment of the huntingtin protein. The idea is that this antibody could tell the immune system to focus on clearing away the harmful protein.

In early assays this antibody prevents pieces of harmful huntingtin from sticking together and starting to form larger clumps which are thought to be toxic. Alchemab work with a number of academic collaborators who are now repeating these experiments – it’s a cool partnership! Alchemab has also shown that ATLX-1095 increases the amount of huntingtin cleanup being performed by a type of support cell called microglia. So the drug is helping to “gobble up” trash in cells, as Donna puts it. The drug is also able to penetrate into the brain. While these antibodies originally come directly from human cells, they need to be manufactured on a larger scale in order to do more experiments and eventually, human trials. Alchemab has had early success in being able to produce the antibody in a laboratory. Now they are testing this potential drug in mice, so we will watch this space as they progress through these next stages of preclinical work!

Targeting exon 1

Next up is Dr. Pavlina Konstantinova who is from VectorY. They work on a number of different brain diseases, including ALS, Parkinson’s, and HD. VectorY, like many other companies we’ve heard from, are trying to specifically target the mutant copy of the huntingtin protein while sparing the normal copy. They are specifically focused on targeting the part of the protein that arises from the CAG repeat expansion, which occurs right at the beginning of the gene, known as exon 1. This encodes a long string of glutamines which is what makes this protein sticky and clumpy.

VectorY is also developing a new technique known as VecTab, similar to PROTAC that we heard about earlier. It’s another way of putting a “trash tag” on the harmful huntingtin protein that tells the cell to break it down and throw it away. Pavlina is sharing data showing that these “VecTabs” reduce the amount of huntingtin clumps found inside cells in a dish. They have also treated HD mice, and so far they are seeing reduced huntingtin clumps as well as some improvement in their movement symptoms.

They are also working on a treatment for ALS using delivery with viruses, and have tested its ability to spread through the brain in mice and larger animals like pigs and primates. So far they are seeing good spread, meaning that the treatment reaches many areas of the brain. VectorY is also working on improving the technologies needed to direct their targets for clearance by the cell, to deliver the drugs to the right parts of the brain, and to manufacture the drugs on the scale needed to bring it to patients.

Targeting MSH3

The final talk before a break for lunch is from Dr. Todd Carter at Voyager Therapeutics – that’s right, yet another company working on HD, very exciting! Voyager is working on targeting MSH3, the genetic modifier we heard a lot about yesterday. Voyager is a “gene therapy” company – meaning they focus on delivering therapies to the body’s cells using harmless virus particles. Voyager’s particular focus is a type of virus called AAV that’s naturally really good at breaking into brain cells, the most important target for HD.

Todd describes that Voyager’s new viral particles, which they call TRACER AAV, have a very unique capability. Unlike natural AAV’s, this engineered virus is able to jump into the brain after being injected into the bloodstream.
Most AAV’s are totally stopped by the blood brain barrier (BBB), a tight seal that protects the brain from harmful things like viruses and other toxic molecules found occasionally in the blood. Most AAV’s, if injected in the blood, are totally excluded from the brain.

Using clever technology, Voyager has constructed AAVs that can make their way through the BBB into the brain after a simple injection in the blood. If this works, it could simplify gene therapy for HD compared to previous approaches which relied on direct brain injections. Compared to natural viruses, Voyager’s new AAVs get to a larger portion of the brains of monkeys when they tested them out. This suggests that there’s a good chance that this might provide a way to deliver useful payloads to the brains of people with HD. One cool feature of this technology is it could be used to carry multiple therapeutics for delivery into the brain at the same time. For example, huntingtin lowering and MSH3 targeting drugs which could be used in a combination treatment for people with HD.

Biomarkers and HD: the keynote speech

After a short break we are back for the keynote speaker this afternoon. The talk will be from Dr. Henrik Zetterberg, who is affiliated with University of Gothenburg and University College London. This talk will be focussed on biomarkers for HD, including neurofilament light, also called NfL.

Henrik is starting with a history of the people, the technology, and the discoveries that led us to the use of NfL as a biomarker – something we can reliably measure to track the progression of HD as well as other brain diseases. Work from teams at University College London, including HDBuzz’s own Prof. Ed Wild, has shown that there is a relationship between levels of NfL and aspects of HD, such as CAG repeat length and timing of symptom onset. NfL can be used as a marker of brain damage in injury and disease; for example Henrik has shown increased levels of NfL in boxers right after they spar, especially when they take many hits. NfL levels also drop after successful treatment of nervous system diseases like multiple sclerosis, spinal muscular atrophy, HIV, and more recently forms of Batten disease and Alzheimer’s. Researchers can measure NfL in biofluids like blood and CSF (the fluid that bathes the brain and spinal cord). In some diseases, like HD and Alzheimer’s, NfL levels go up just as symptoms begin to appear.

Henrik’s lab is also using NfL to try and tease out whether a person experiencing new symptoms might have a psychiatric disease, like major depression or schizophrenia (low NfL), versus a neurodegenerative disease, like HD or Alzheimer’s (high NfL). Another area in which NfL measurement could be useful is to help doctors understand whether someone is experiencing neurological injury or unexpected side effects following general anesthesia, cancer treatment, or surgery. New methods are making it easier and easier to measure NfL – rather than having to take a sample of spinal fluid and preserve it carefully for analysis, sample collection could be done at home with a finger prick! He’s also looking at NfL from some unexpected and rather wacky angles – like showing that the brain’s support cells “eat” NfL, showing it goes up in bears when they hibernate, or that it increases in astronauts who go to space! Overall, Henrik emphasizes that NfL can be useful as a measurement of disease onset, as a way to understand the safety of a drug or a procedure, and as another angle to observe other factors that influence HD symptoms, like age and genetic variations.

That’s all for day 2! We will be back tomorrow morning to report on the third and final day of HDTC2023! Remember to continue to follow live updates for the rest of the conference on Twitter, with the hashtag #HDTC2023.

Huntington’s disease therapeutics conference 2023 – Day 1

Hello from Dubrovnik, Croatia, where the 2023 CHDI Therapeutics Conference will be taking place from Monday, April 24th, through Thursday, April 27th!

This conference is a big one for HD researchers worldwide, from industry, academia, and nonprofit. Dozens of scientists will give talks on all things HD, from genetics, to therapeutics, to clinical trial news.

The HDBuzz Editorial team will be on the scene starting on the morning of Tuesday, April 24th, live-tweeting scientific talks and updates on the progress of clinical trials. Our Twitter updates are compiled below. Continue to follow live updates for the rest of the conference with the hashtag #HDTC2023.

For a summary of last year’s conference, start here: https://en.hdbuzz.net/320 We’ll post summaries in article format for each day of the conference.

Knowing what we need to know

The first talk of the morning comes from Dr. Vahri Beaumont from CHDI, who will give an overview of what we still don’t understand about HD and what we need to know to better develop therapeutics. She is first discussing the history of our understanding of HD genetics and brain changes, from CAG repeats, to loss of brain cells and circuits, which scientists have come to understand through the study of human brain tissue donations and brain imaging.

We have known for a while now that people with HD who have the same CAG number can start to get symptoms at different ages. One reason for this is other genetic differences in a person’s DNA code. Scientists are studying these DNA letter changes to better understand how they might alter HD onset and might also be exploited for making new medicines for HD. Many of these other genetic differences affect “somatic instability,” in which the CAG mutation that causes HD mutates even further in some brain cells, getting even longer. Long CAG repeats in the huntingtin gene leads to an expanded huntingtin protein, which over time can be toxic to different parts of brain cells. Vahri reminds us that there is still a lot we don’t understand about the precise sequence of events which link the HTT gene expansion to the symptoms people with HD experience.

For example, we are still unclear on exactly which HTT gene product is the key player in disease – is it the message molecule? The protein? Protein clumps? Perhaps they all play a role in HD. Another question that remains unanswered is whether the “bad” copy of huntingtin is messing things up, or whether the loss of one “good” copy leaves brain cells without some function. Regardless of these questions, several therapeutic approaches that target HD genetics are already being tested in the clinic. Some focus on total huntingtin, both normal and expanded; others target only the expanded form.

The good news is that there are many different companies testing out all kinds of approaches in the clinic, testing many different hypotheses. Perhaps a combination of these therapies may be the best way to treat HD. Thanks to the generosity of brain donations from people with HD after they pass, scientists are continuing to make breakthroughs to understand the disease in people using these very precious tissue samples.

Scientists are using the most cutting-edge technology to understand what is happening with huntingtin messages and protein in different types of cells and why certain types might be more vulnerable. Using many different kinds of animal models, researchers are building a better picture of what happens inside the brain during HD and how we might intervene.
Animal models also allow us to test interventions like drugs at very early stages of HD.

Vahri points out that there are some limitations to mouse models which don’t show all the symptoms of HD in people. Scientists are continuing to develop and use multiple models so they might best test drugs before they are used in people in the clinic.

One of the biggest goals in HD research is to be able to start treatment before symptom onset. This is not easy, but a very strategic staging system, the HD-ISS https://en.hdbuzz.net/325, will help scientists achieve this.

Data Sharing

Dr. David Howland from CHDI is introducing the first official data sharing session of the conference. It will focus on huntingtin DNA and how our understanding of its structure can inform the development of therapies.

DNA interruptions

First up is Dr. Galen Wright from the University of Manitoba, who will be discussing how small variations in the huntingtin gene affect the course of HD. 2023 marks the 30th anniversary since the mapping of the HD gene, huntingtin. This gene is VERY big. Much bigger than most other genes in our bodies, which can make it challenging for scientists to study.

Galen is recapping what we’ve learned about the HD gene, the tendency of CAG repeats to expand in some brain cells over time (somatic instability), and the other genes that influence this expansion.

Three DNA letters code for a single amino acid, the building blocks of proteins. CAG codes for glutamine. Interestingly, CAA also codes for glutamine and it turns out that most folks with HD have a CAA “interruption” in their CAGs. People who don’t have this CAA interruption in their huntingtin gene get disease a lot earlier on in life, even though the protein coded for by the gene is exactly the same. This happens very rarely, but it suggests there is something about the DNA code which is important in HD. Scientists thought that these CAA interruptions would alter how the huntingtin gene might change through somatic instability, but it turns out that isn’t the case. This means there is still more work to do to understand what is going on.

When people undergo predictive testing, the overall length of the CAG repeats is measured. Although small DNA letter changes can make a big difference in HD symptoms, we are not at a point of measuring this in individuals to understand their likelihood of early or late onset.
Interestingly, there are other diseases which are caused by DNA letter changes in the huntingtin gene, including Rett syndrome and another disease called LOMARS. These diseases also affect the central nervous system like HD.

Galen’s team mined large open datasets which bring together gene association data from many different studies, not necessarily focussed on HD. They found that the huntingtin gene is linked to traits like aging and psychological symptoms. Together, this means that the huntingtin gene is probably important in lots of different roles in our nerve cells and that the biology of huntingtin is complex. Galen rightly points out that the more we learn, the more questions we have about huntingtin.

Dissecting DNA repair

Next up is Dr. Anna Pluciennik from Thomas Jefferson University. Anna’s lab studies how mutations in our DNA letter code occur and how these might lead to disease. The mutations are caused by damage to DNA, which happens an estimated 50,000 times daily! We have evolved many ways to repair DNA to avoid a build up of mutations.

Anna’s team studies a specific type of DNA repair called mis-match repair, which corrects a situation where the 2 strands of the DNA helix aren’t properly matched so the helix structure is a bit wonky. These wonky structures are recognised by special machinery which can then try to fix these problems to correct the DNA letter code. Ironically, in some cases (like with CAG repeats) this machinery actually makes things worse.

Anna’s lab studies biochemistry and she likened it to disassembling a car into its thousands of parts to understand how they all work together. This allows her team to work out details that can’t necessarily be observed in complex cell cultures or in animal models. In her lab, Anna’s team makes a proxy for the HD mutation to understand how repair machinery might recognise and try to fix it. She studies the expansion of CAG repeats, which can cause them to stick out of the DNA helix, a structure known as an “extrusion.”

Using this proxy, Anna’s lab is dissecting which DNA repair proteins do what. This type of dissection is important for future studies that might target such proteins with drugs which could help to treat people with HD. Anna’s work is helping to understand how different amounts of each protein might tip the balance to decide whether the machinery corrects DNA damage as it should, or inadvertently makes things worse.

DNA structure influences function

The final talk of this first morning session is from Dr. Natalia Gromak, from the University of Oxford. Natalia’s team studies special structures called R-loops which may be important in HD. R-loops are formed when the messenger copy of the DNA code, called RNA, is being made. If the message RNA copy interacts with the DNA like a zipper, it forms a sort of bubble in the DNA.

These structures have important roles in certain functions in cells, but can also interfere with things causing disease, so they must be balanced carefully. Very early on, a link was made between R-loops and neurodegenerative disease including ALS. Natalia’s group has generated a list of proteins that interact with R-loop structures in hopes of understanding their roles in biology and how these might go wrong, causing disease. 50+ diseases have repetitive DNA sequences which are expanded – just like in HD.

The Gromak lab found that R-loops are formed in regions with repetitive DNA, and have studied R-loops in Friedreich’s ataxia. The question for this conference of course is whether R-loops play a role in HD. Natalia’s group found that there are more R-loops in blood cells derived from HD patients, and found the same result in HD mutation-bearing neurons grown in a dish. There is also more DNA damage in both of these cell types. Next questions for the team are whether R-loops form on the repetitive sequence in the HD gene, whether they can affect the further expansion of this region (somatic instability), and whether huntingtin lowering has any effect on the R-loops seen in HD cells.

CRISPR and HD

Up next following a caffeine break is Dr. Michael Brodsky, from UMass Chan Medical School. Michael’s lab uses CRISPR technologies which can be deployed in the lab to make very precise edits of genomic DNA sequences. Targeting the root cause of HD, the CAG expansion in the huntingtin gene, is the most sensical way to treat the disease, but this is easier said than done. Gene editing would be one way to do this, but we’ve had to wait for the technology to catch up.

10 years ago, this was all a pipedream but technologies have improved so rapidly that we are now very seriously studying gene editing as a possible therapy for HD which is very exciting! Michael points out that gene editing is permanent, so much care must go into making sure there are no unintended changes. Another challenge for using gene editing for HD is that the drug must be delivered into neurons, which is no mean feat. The gene editing must also be very precise. This means that ideally only the expanded huntingtin gene is targeted, so there are limited or no changes to the normal huntingtin gene – also a tall order.

Michael’s group is taking two approaches to specifically gene edit the expanded huntingtin gene. The first is to target small letter changes (called SNPs) in the rest of the huntingtin gene DNA which tend to be associated with the expanded version. The Brodsky lab is first trying out these experiments in all sorts of different HD mouse models, results of which suggest that they are able to specifically edit just the expanded huntingtin gene – great news!

An alternative approach to specifically gene edit the expanded huntingtin gene is to actually reduce the size of the CAG expansion back to the normal range. Michael’s group has been successful doing this in HD mice and cells in a dish. There are still some kinks to work out before this can be developed as a potential treatment for HD but they are cautiously optimistic that further research will help to define a path forward.

More CRISPR and HD!

The next talk, from Dr. Ben Kleinstiver of Harvard/MGH, will also focus on DNA editing. He runs a genomic technology development group that is working on how to alter the expansion of CAG repeats and eventually create therapeutics. Ben’s lab focuses on the many ways that CRISPR can be used to make many different types of changes to DNA. They are engineering the CRISPR machinery to tailor these changes even further.

His main research question is, “what genome editing tools can be used to alter or shorten CAG repeats?” The lab takes different approaches to cutting repeats, interrupting them, or replacing single DNA letters or sequences. Because CRISPR evolved as a way for bacteria to combat virus attacks, there are still some limitations to using the CRISPR machinery to treat diseases. Ben’s group is working on overcoming these limitations to allow better access to different parts of the HD gene. Techniques include using different types of DNA-cutting or letter-replacing enzymes, and applying different methods to direct them at DNA sequences. Then they measure whether CAG repeats get shorter. The goal is to fine-tune the editing and customize it for the huntingtin gene.

This is Ben’s first HD conference! It’s exciting to see how CRISPR experts are directing their efforts towards HD. As technologies continue to advance we hope they can be applied to future human therapeutics.

Even more CRISPR!

Next up is Kathryn Woodburn from Life Edit Therapeutics who will give the last talk before we break for lunch. Kathryn works on ways to target the expanded copy of the huntingtin gene with editing technologies. Life Edit Therapeutics is looking at how different versions of the CRISPR machinery, especially those found in plants, can be used to customize editing of the expanded huntingtin gene.

Their approach to treating HD will be to use viruses to deliver their editing machinery to the brain. So far they have tried this in different kinds of HD mice with different versions and different doses of their gene editing drugs. They are able to decrease levels of harmful huntingtin protein by 40% while leaving the healthy protein intact! To get expanded huntingtin gene specificity, their approach is to target specific DNA signatures which are only found in the expanded version of the gene. Life Edit Therapeutics are looking at a few different signatures to do this and so far the data looks promising.

Making sure that there are no unwanted off-target effects is a challenging task and the scientists at Life Edit are working to get this figured out as quickly as possible. That’s all for the morning’s session!

HD Genetic Modifiers

Day One’s afternoon’s session will focus on progress being made in the study of HD genetic modifiers.

Understanding MSH3 in HD

Large scale human genetic studies, known as GWAS, have allowed researchers to identify these genetic modifiers, other genes that influence when HD symptoms begin. The first talk is from CHDI scientists, Dan Felsenfeld and Tasir Haque, who will be telling us all about their big team effort studying a gene identified in the GWAS called MSH3, and how they might be able to make drugs targeting this protein.

MSH3, as you may remember from the earlier talks, recognises wonky bits of mismatched DNA which need to be fixed. The CAG expansion in the huntingtin gene is prone to create these wonky bits, and it’s thought that MSH3 activity at the huntingtin gene may inadvertently increase the number of CAG repeats in brain cells (somatic instability). Scientists think that MSH3 could be a good target for medicines as switching off this gene seems to be beneficial in animal models of HD, as it reduces somatic instability – the expansion of the CAG number – in the huntingtin gene.

Completely switching off a gene is quite challenging to do in people, so instead scientists are making so-called “small molecule” drugs, which could potentially be taken by mouth, that aim to stop MSH3 working so well in cells. Dan’s team has considered different ways to inhibit MSH3 and created a toolbox of materials and protocols to study their small molecules. This will help other researchers hoping to make drugs targeting MSH3. To make better drugs, it helps to be able to “see” the MSH3 protein. Using clever techniques, it is possible to create 3D models of the protein, and then the scientists can see where and how their molecule binds.

Tasir Haque is now showing some animations that zoom in on different parts of the MSH3 protein and where the drugs fit in. Lots of squiggles that have great meaning for structural biologists! Using these models, they can work out how to better improve these early stage drug molecules to better fit into all the nooks and crannies of the MSH3 protein surface, which should improve their properties.

Drugs targeting MSH3

Next up is Caroline Benn from LoQus23 Therapeutics, a company that is also working to develop drugs targeting MSH3 – it’s a hot area! LoQus23 is taking a slightly different approach to the CHDI program on MSH3 – they are making molecules which target a different region of the protein. This is good news for the HD field as it’s great to be able to test out multiple approaches! Although their approach of targeting different regions of MSH3 is more difficult, they have pulled it off and found two such series of molecules which are very potent and selective, meaning they bind the MSH3 protein very tightly without affecting other proteins.

LoQus23 has also established a way to measure somatic instability in cells in a dish to test how well their molecules work. These are complex experiments which take weeks from start to finish.They will also be able to use this platform to find new targets, besides MSH3, which play a similar role in this part of DNA damage repair which is so important in HD.

Next up is James Fleming from Pfizer. This company is also developing drugs to target the pathway involving MSH3. Pfizer are taking a similar approach to the CHDI team, and like the other folks, have developed a suite of tools and methods to test their molecules for the ability to stop expansion of CAG repeats.

Like other pharmaceutical companies, Pfizer takes a series of steps to screen potential drug compounds, understand how they interact with the proteins they target, and then test them in cells and in animals. They too are using 3D models and chemical tests to show that their drugs can stick to the protein complex that MSH3 is a part of, which has helped them make these molecules better and better over time. The next step is to test these drugs in cells grown in a dish. A lot of this work is focused on the minute details of the protein chemistry, structure, and energetics. Suffice it to say that mathematics figures into the drug development process!

To bring these studies into animals and then later into humans will require a drug with the right properties: the ability to target MSH3, the ability for the body to break it down, and the ability for the drug to get into the brain. Not a small task! After better understanding the properties of a new drug in cells and animals, it may then be tested for safety in people. Right now this is a bit far in the future for all of the compounds presented today, but it’s exciting to see that companies are moving the work forward.

Massive datasets to identify genetic modifiers

Kicking off the last session of the day is Jim Gusella from Harvard, who will be telling us about genetic modifiers of HD on behalf of a large consortium of scientists who study HD genetics.

Jim begins his talk by acknowledging all of the HD families who have so generously shared their data and samples with HD researchers over the years, without whom, these large-scale analyses would not be possible. An interesting finding we have known about for a while now, is that folks with the same CAG number may start to have symptoms at very different ages. Genetic modifiers are markers in the DNA which can explain this early or late onset of symptoms.

More and more evidence is pointing to a particular driver of the onset and speed at which HD gets worse over time: the expansion of CAG repeats in some cells. This process, known as somatic instability, seems to be linked directly to genetic modifiers. The power of these modifier studies comes from the number of patient samples which are analysed – more data means higher confidence conclusions. In the most recent study, 11698 participants’ data were analysed which is amazing!

A problem with HD research, and science in general, is that many of the samples analysed are from Europeans or folks of European heritage. In this later dataset, the team is working to include a more diverse group of patients in the data. With such a wealth and diversity of data, it is possible to zoom out on a large scale and make general predictions of how genetic modifiers – tiny changes in other genes – affect when people with HD might reach certain stages of HD.

It is important to emphasize that this is a way to have more confidence about what other genes most affect HD in people. This is different from being able to predict onset or disease course in an individual person with HD. Jim’s team and the genetic consortium are also looking at how subtle differences and “interruptions” in the sequence of CAGs in the huntingtin gene affect the DNA structure and the tendency of the repeats to become unstable and grow longer. The good news from this most recent dataset is that MSH3, the topic of the previous session, is still a very significant modifier by all of the analyses Jim and colleagues used. That lends a lot of credibility to all of these approaches targeting somatic instability and trying to stop the expansion of the CAG repeat or shrink it.

CAG expansions in specific brain cells

Next up is Nathaniel Heintz, based at Rockefeller University, who will be talking to us about his work on understanding which genes are switched on or off in HD. The Heintz lab developed a series of techniques that allow scientists to “sort” the nuclei of many different cells and look at genetic messages in many cell types. This has become an important way to study why certain cells are most vulnerable in HD and other diseases. These analyses use post-mortem brain tissue samples, enabled by the amazing generosity of the HD community.

The striatum, an area in the center of the brain, is most deeply affected by HD. Heintz’s team is able to sort through different kinds of cells in the striatum and have discovered that CAG repeat expansion occurs most often in one type of cell, medium spiny neurons.

We have known for a long time that medium spiny neurons (MSNs) are lost in great numbers in HD. There are different types of MSNs and oddly it turns out that both those which are vulnerable in HD and those which survive are subject to CAG repeat expansion. The reason these medium spiny neurons seem to have higher levels of expansion could be due to the higher levels of MSH3 which are found in these cells but that link is not yet proved. Medium spiny neurons also have huge numbers of genes that are switched on or off in HD – 1000 turned on and 500 turned off! Many of the genes affected are involved in DNA damage repair – again reinforcing the important role this likely plays in HD.

Ongoing work is addressing the questions of when medium spiny neurons are affected in HD and how best to intervene. They are also looking at other brain areas and going layer by layer to understand exactly which types of cells are becoming damaged or lost.

Speed of CAG expansions

The last speaker for the day is Steve McCarroll, from Harvard. Steve’s lab looks at which genes are switched on or off at the levels of single cells, instead of a big mixture of loads of different types of cells – an incredibly detailed approach. He uses a fruit analogy to talk about the power of this technique – you can compare cell types like different kinds of berries, the same cell type in different people like apples to apples, the differences between different cells of the same type, like looking at two blueberries.

From these single cell analyses, they can work out which cells disappear over the course of HD, confirming previous findings that medium spiny neurons and spiny projection neurons are the most vulnerable kinds of nerve cells. They can also work out exactly which cells have CAG expansions – this seems to suggest that the vulnerable medium spiny neurons are the most likely to have the most expansion. The CAG expansion during a person’s lifetime in these cells seems to be very specific JUST to the huntingtin gene, and only the HD gene, not other genes which have similar kinds of DNA code. The majority of these vulnerable brain cells have moderate expansion in the CAG repeat, but a small subset have huge expansions which scientists haven’t quite figured out the reason for just yet.

According to the McCarroll lab’s data, the moderate expansions seem to happen very slowly over time, but the more exaggerated expansions happen much more rapidly. The key question is, at what threshold of CAG repeat number does the expansion speed up, causing damage and death of these vulnerable nerve cells? To try and figure this out, the McCarroll lab can compare individual neurons with different CAG numbers, and split them into groups to better understand what lengths are most problematic. They grouped cells by CAG number and strangely didn’t see too many differences in genes turning on and off at lower repeat lengths. The most profound changes occur in cells with very, very long CAG repeats, more than 180.

McCarroll is proposing a very different way of thinking about HD pathology and how the disease works over time. There is some interesting chatter in the audience! But this is why it’s so good that all these scientists can get together at this meeting to debate all these ideas.

Tune in tomorrow!

That’s all for today, folks. We’re breaking for the night, but will be back tomorrow morning! Remember to continue to follow live updates for the rest of the conference with the hashtag #HDTC2023.

PROOF-HD study of pridopidine ends with negative result

Top line results of the PROOF-HD study, run by Prilenia Therapeutics and testing pridopidine, have been announced at the American Academy of Neurology convention. Sadly the trial outcome was negative. We recap the history of pridopidine in Huntington’s disease, review the trial results, and figure out where this disappointing result leaves us.

The drug: pridopidine

Pridopidine has been under investigation as a possible treatment for Huntington’s disease since the early 2000s and has a long and colourful history. It was initially developed by the Swedish company Neurosearch who called it Huntexil.

Neurosearch thought pridopidine was able to stabilise levels of dopamine, which is important for movement control. They hoped it might therefore suppress involuntary movements and improve voluntary movements. They ran two trials called MermaiHD and HART, but the drug did not show conclusive benefits for movement control.

In 2012, Teva Pharmaceuticals purchased the right to develop pridopidine, and ran a third study called PRIDE-HD which tested several different doses of pridopidine, again with the aim of improving movement function.

The PRIDE-HD study ended in 2016 with a negative result for movement improvement, but a curious finding when the data were scrutinised after the event. For one of the dose levels tested, there was an apparent stabilisation in a clinical score called total functional capacity or TFC.

TFC is a score out of 13 that estimates someone’s ability to work, undertake household tasks, care for themselves and so on. TFC tends to decline steadily as Huntington’s disease progresses, and a drug to slow or halt the decline in TFC would be very appealing.

One mystery at the time was how pridopidine might have a beneficial effect on function without actually impacting movement control, which is what it was supposed to do.

The twist: a change of mechanism

While Teva was studying pridopidine in the PRIDE-HD study, its scientists were making new discoveries about how the drug actually worked.

Unexpectedly they found that its main action was nothing to do with dopamine, but was instead targeting a protein called sigma-1 receptor or S1R, which is involved in helping neurons survive under conditions of stress. You can read about this in detail in this HDBuzz article.

These findings about pridopidine caused a rethink about what the drug might be able to do in the brain. Improving movement control would be a symptomatic benefit, while extending the survival of neurons would be a disease-modifying result that could actually slow progression of HD.

Prilenia and PROOF-HD

The rights to pridopidine were then moved to a new company called Prilenia Therapeutics. Buoyed by the new findings about S1R, Prilenia launched the PROOF-HD trial in 2020.

PROOF-HD would be pridopidine’s fourth attempt to meaningfully impact Huntington’s disease. The trial enrolled 499 participants with HD and tested one dose of pridopidine (45mg per day) against placebo.

The primary outcome measure was TFC, so the trial was asking whether pridopidine could slow the progression of HD over 15 months, by comparing TFC changes for participants receiving the drug or placebo.

PROOF-HD was classified as a phase 3 trial, meaning a positive result would let Prilenia get approval for pridopidine to be prescribed to HD patients.

A negative result

PROOF-HD finished in March of this year, and the top-line results were announced today at the American Academy of Neurology meeting in Boston, USA, by the trial’s Principal Investigator, Dr Andy Feigin.

We won’t sugar-coat this: the trial results were unfortunately negative. The drug did not slow progression of HD as measured by the TFC.

Failing to meet its primary endpoint means that pridopidine will not get licensed by the FDA and other regulatory agencies.

All trials have secondary endpoints, which are measurements of special interest that might suggest the drug is doing something useful even if it doesn’t meet its primary endpoint. Unfortunately Feigin reported that PROOF-HD failed to meet its secondary endpoints, too.

Where now?

The news of a negative result for PROOF-HD will of course be a big disappointment for everyone who took part in the trial and the whole HD community.

The HD community is becoming all too used to hearing news from clinical trials which do not pan out as we would have hoped. But it’s worth reminding ourselves of a few fundamental truths.

First: a negative trial is not a failed trial, as long as we learn everything we can from it. The data from PROOF-HD will help us learn more about the effect of pridopidine and how to design better drugs and trials. Our daily updates from the currently-running HD Therapeutics Conference will give you a very thorough rundown of what’s being developed and what trials are underway.

Second: Every trial will be negative until one is positive. Many other trials are running or coming soon that test drugs targeting solid, known features of HD.

And finally: this community is robust, smart, and determined. We know how to turn sadness and disappointment into a positive, unstoppable drive to succeed. We know how to get right back on the horse and test the next promising drug, to ensure that not a single day is lost in bringing about the future we are all working for: a future we create together where we have safe, effective treatments to slow or prevent HD.