Specialist
Former executive at Precision NanoSystems Inc
Agenda
- LNPs (lipid nanoparticles) as a drug delivery vehicle and demand outlook across RNA therapeutics, mRNA vaccines, in-vivo gene therapies and more
- LNP supplier landscape analysis – key players' competitive positioning, sources of differentiation, plus potential patent considerations as the industry scales
- Outlook for LNPs vs competing transfection and transduction technologies including electroporation and viral vectors respectively across in vivo and ex vivo settings
- Next-generation LNP technologies review and complex API (active pharmaceutical ingredient) delivery vehicle constructs
Questions
1.
We are all somewhat familiar with LNPs given they are used as part of the mRNA vaccines today, but by way of introduction, what an LNP is and why they so useful for delivering complex APIs [active pharmaceutical ingredients] delivery?
2.
You mentioned high production capabilities and scalability as major advantage to LNP as a delivery vehicle. My understanding is that LNPs have further advantages in terms of their packaging capacity, lack of immunogenicity, biodegradability and so on. How significant are these?
3.
What are the purposes of the various LNP classes? What are their respective applications? The MC3 class seems fairly prevalent.
4.
We’ll circle back to the tissue targeting point later in the call. To what extent do LNPs have to be tailored to the specific application or indication in question? For example, the MC3 class was specifically designed over multiple iterations for therapeutics such as Onpattro [patisiran]. Is there such thing as a plug-and-play LNP? How should we be thinking about the scalability and potential inflection in demand for LNPs going forward?
5.
MC3 is an ionisable lipid. At pH 4 it combines with mRNA and this neutralises the charge so in physiological conditions there is less toxicity. That being said, toxicity and tolerability issues are still there even with these ionisable lipid nanoparticles. In your opinion, what will the next-generation lipids comprise of and how will the new constructs address the tolerability issue?
6.
Putting cargo design aside, what modifications might happen in next-generation LNPs change to widen the therapeutic window? I’ve read, for example, that removing PEG could be part of the solution. Even it’s not proven unequivocally, there is increasing evidence that PEG is playing part of the early allergic reaction and adverse reactions we’ve seen with the COVID-19 vaccine.
7.
Do you expect LNPs or variations of what we have today to remain the delivery vehicle of choice for complex APIs going forward? Alternatively, might we notice a shift towards lipoplexes, so cationic liposomes, polyplexes, exosomes, etc?
8.
How long do you think it would take for LNPs being made redundant by exosomes, polyplexes and so on? What’s the useful life?
9.
As discussed, new LNP classes are being developed and modifications made to widen the therapeutic window, and there may be a shift towards the lipo and polyplexes, how conserved are the underlying equipment technology or input materials required across those different delivery constructs, given you said exosomes is a separate beast? I’m curious to understand whether existing LNP manufacturers can easily pivot into these new delivery constructs and whether the existing equipment would have use here.
10.
Do you expect steady demand for the sterile filtration mix equipment and other enabling technologies here?
11.
Going one level deeper than the LNP constructs we’ve been discussing? As an example, I understand Biotage sells its Flash 400 chromatography columns to Croda for the purification of LNPs used for Pfizer and BioNtech’s mRNA vaccine. As demand for LNPs increases, for what instruments or technologies will demand equally increase?
12.
Could you comment on the patent landscape? What issues may rise from the IP estates around all of this as the industry scales out?
13.
What do you think will be the core demand driver for nanoparticles going forward? Would it be RNA-based therapeutics, vaccines, gene therapy or anything else?
14.
You noted that LNP-mediated transfection could be a major competitive threat to electroporation in ex vivo gene therapy applications. We have indeed heard that electroporation impacts cell viability and function, but efficiency is very high and there are scale and time-to-market advantages for this approach. What are the salient advantages beyond viability for LNP-mediated transfection? How significant a threat this represent to combined microfluidics and electroporation technologies which hope to have a far smaller impact on viability, such as that from a player such as Kytopen?
15.
On what timeline could a commercial grade LNP transfection platform compete with electroporation platforms?
16.
Perhaps a gross oversimplification, but electroporation involves an electric pulse opens up the cell membrane and genetic material, or other cargo, can diffuse across the membrane. By what mechanism would LNP transfection work? Would you need additional stimulus to mediate transfection, for example?
17.
I understand electroporation is very application specific, in that one needs to optimise the protocols to ensure optimal efficiency and cell viability. In essence, this is a highly time consuming process and fairly difficult to do and doesn’t really represent a plug and play solution. Would LNPs equally application specific given specific tropisms for certain cells or one needs to add certain proteins such as ApoE [apolipoprotein E]? Or, could we have a plug-and-play LNP-based transfection platform irrespective of cell type?
18.
My understanding is that electroporation cannot be used for knock-in edit –if you want to introduce a CAR construct into a T cell you would still have to use a viral vector, so electroporation is predominantly used for knock-out edits such as TCR knock-down. Would an LNP mediated platform be able to perform both knock-in and knock-out edits?
19.
Who are the leading players for the ex vivo nanoparticle-mediated transfection?
20.
Moving to the in-vivo side of things, is it reasonable to assume that LNPs will be the enabling technology for CRISPR-Cas gene editing in vivo as opposed to adeno or lentiviral vectors, with reference to the data that coming out of Intellia?
21.
How sanguine are you about nanoparticles being able to tissue-target beyond the liver, eye and lung? To your point, liver diseases – as targeted by Intellia’s NTLA-2001 – are the low-hanging fruit, given LNPs preferentially accumulate in the literal sense of the physiological closed compartment, and that the lungs are an option because LNPs can be nebulised.
22.
Forgive me if I’m being slow here, but I understand that even with a tropism for the heart, we would still have non-functional LNP delivery in the liver, given LNPs preferentially accumulate there. To that extent, would any tissue targeting require higher and potentially toxic doses? We know LNPs are associated with dose-dependent toxicities, so would an extra-hepatic dose require a quantity that would be toxic for the liver? Could it be a major challenge to find a therapeutic window for targeted LNP delivery?
23.
What would be the use case for viral vectors in in-vivo gene therapy if LNPs can one day molecularly target tissues? LNP coupled with mRNA have certain advantages including the transient protein expression of the CRISPR-Cas9 complex, which limits potential off-target effects – we don’t want gene editing machinery sticking around in the cells for too long. In essence, I’m curious to understand whether LNP and mRNA may make viral vector mediated in-vivo transduction obsolete?
24.
In summary, it sounds as if we could move away from viral vectors across cell and gene applications, whether ex vivo or in vivo. For ex vivo applications viral vectors are used for knock-in edits, but if we move towards LNP-mediated transfection technology, which can do both, we don’t need them anymore. In ex vivo, as we move towards CRISPR-Cas9, should they be able to demonstrate specific tropism, given the immunogenicity and perhaps even the integrating concerns of the latter. This seems to be bad news for the viral vector players. Am I missing anything?
25.
What’s not to like about the use of LNPs in cell or gene therapies? We discussed the barriers to molecular tissue targeting but those seem surmountable.
26.
Let’s revisit Intellia NTLA-2001 as a potential case study if you will. The data we have is for 0.3mg per kg dose that led to an 87% mean reduction in serum TTR with a maximum of 96%. There was a strong dose response relationship. Intellia now going to move the trial up to a 1mg per kg and then 2mg per kilogram. Can we assume an equal efficacy for serum TTR or could safety become an issue?
27.
Do you expect low safety risks for in vivo gene therapies using LNP delivery vehicles? Are these de-risked given the mRNA COVID-19 vaccines?
28.
What are your closing thoughts on the demand outlook for LNP or nanoparticle delivery systems? Who is well placed to win here?