Monday, February 3, 2020

Treating achondroplasia: statins' role for the treatment of achondroplasia

Statins restore bone growth in an animal model of achondroplasia

As many of you may know, a few years ago a Japanese group published an elegant work showing that the statins, a family of drugs widely used to reduce cholesterol levels, were able to restore bone growth in an animal model of achondroplasia (1). However, the researchers were not able to find out how these medicines were working in their model.  Later, the group leaded by Pavel Krejci published a study in which they ruled out any direct action of statins on the fibroblast growth factor receptor 3 (FGFR3) pathways (2).

Now, it seems that the mechanism by which statins could restore or promote bone growth might have been unveiled, with the publication of a new study where researchers observed that fluvastatin, one of the statins, was able to increase the activity of a key enzyme axis that positively regulates bone growth, the IHH-PTHrP pathway (IHH:
Indian Hedgehog; PTHrP: peptide (or protein) related to Parathyroid Hormone) (3).I have already reviewed it in an article from October last year.

This sounds complicated but in summary, bone growth is a consequence of a cell process within structures called growth plates that are located in the extremities of the long bones (Figure 1). There, the chondrocytes (the master cells of bone growth), reacting to many agents, go from a resting state to a highly proliferation frenzy and finally enlarge several times from their baseline size (Figure 1), then giving space to new bone tissue. It is the continuous chondrocyte's "awake, proliferate, enlarge" cell cycle that elongate bones. This cell cycle is tightly regulated and both
IHH and FGFR3 are fundamental to modulate how chondrocytes multiply and enlarge. The IHH-PTHrP axis is specially important in the proliferation stage (4) and mutations in the PTHrP receptor that impair its normal activity cause a rare skeletal dysplasia called Jansen metaphyseal dysplasia (5) in which bones are severely shortened. Furthermore, previous studies showed that FGFR3 reduces the activity of the IHH-PTHrP pathway (6), and that PTH was able to restore growth in mouse models of achondroplasia (7,8). 

Figure 1. Cartilage growth plate.


  
In normal state, FGFR3 inhibits both chondrocytes' proliferation and hypertrophy to an extent that allows balanced bone growth through a couple of enzymatic cascades inside the chondrocyte, the STAT1 and the RAS-RAF-MEK-ERK (also called MAPK) pathways (Figure 2). In achondroplasia, due to the mutation in its structure, FGFR3 is working excessively, thus quite blocking the entire growth process. Under this model, upon the overactivity of FGFR3, fewer chondrocytes awake from the resting state and fewer will proliferate and enlarge to enable new bone to be built.

Figure 2. FGFR3 pathways.


Signaling pathways activated by FGF/FGFR. FGFs induce dimerization, kinase activation and transphosphorylation of tyrosine residues of FGFRs, leading to activation of downstream signaling pathways. Multiple pathways are stimulated by FGF/FGFR signaling such as Ras-MAP kinase, PI-3 kinase/AKT and PLC-γ pathways. Furthermore, FGF signaling can also stimulate STAT1/p21 pathway. FGF/FGFR signaling also phosphorylates the Shc and Src protein. FGF/FGFR play crucial roles in the regulation of proliferation, differentiation and apoptosis of chondrocytes via downstream signaling pathways. From Su N et al. 2014. Reproduced here for educational purposes only.


Current therapeutic strategies

Currently, there are four therapies for achondroplasia in clinical development exploring three different strategies. The most advanced one is an analogue of C-type natriuretic peptide (CNP) called vosoritide (9). Another CNP analogue is also being tested (10). CNP works naturally counteracting the effects of one of the FGFR3 pathways, called MAPK, which controls the pace of chondrocytes' hypertrophy, but it has minimum or no effect in the FGFR3 pathway responsible for reducing chondrocytes' proliferation, according to the evidence so far (11) (Figure 3). There are several articles in this blog where we review CNP, you just have to browse the index page to learn more about it.


Figure 3. Therapeutic strategies for achondroplasia.

The figure above shows the site of action of CNP, meclizine and also of the tyrosine kinase inhibitors (TKI) NF449 and A31, which work like infigratinib. From Matsushita M et al. (2013). Reproduced here for educational purposes only.

The other two strategies target FGFR3 directly, but using distinct approaches. One of the drugs, called recifercept, is in fact a modified form of FGFR3 lacking the hook which normally anchors this enzyme to the chondrocyte cell membrane, allowing this molecule to circulate freely when administered (12). That's why it is called a "soluble receptor" (remember that FGFR3 is a receptor enzyme). So, how does it work? As you may know, FGFR3 is a kind of power switch on the wall of the chondrocytes: it needs a finger (the FGFs, the ligands) to turn on (activate) and exerts its functions (Figure 4). By circulating freely in the body, recifercept can reach the growth plates and capture those FGFs before they engage FGFR3 (it works outside the cell), explaining why it is also called a ligand trap. The consequence is that if FGFR3 is not activated, then it cannot block the chondrocyte's cell cycle, and growth can be restored (12). Here, you see that recifercept might be able to inhibit all FGFR3 pathways, so would have effects both in the chondrocyte's proliferation and hipertrophy phases. In theory it would be more potent than CNP analogues.

Figure 4. Ligand trap.


An illustration of the ligand trap strategy. The switch on the wall represents FGFR3 and the finger a FGFR ligand (a FGF). FGFR3 is activated when a FGF binds to it. The trap is made of a "free" form of FGFR3 which competes with the cell wall switch, preventing its activation.


The third strategy is being explored with infigratinib (13). This molecule is called a tyrosine kinase inhibitor (TKI) and it is able to bind the part of FGFR3 which is responsible for activating its pathways inside the chondrocyte (Figure 3). In this case, FGFR3 keeps being activated outside the cell by the FGFs, but it is unable to turn on its pathways inside the cell. This means that infigratinib could be more potent than CNP analogues, also having effects on both key chondrocyte's phases of the cell cycle.


How do those findings about the statins fit in the achondroplasia's therapeutic landscape ? 

As our understanding of how statins work in the growth plate is advancing we could think on a strategy where combining them with CNP analogues may result in improved effect on bone growth. This concept is also applicable to meclizine, which also works inhibiting the same MAPK pathway CNP does (14) (Figure 3). By one side CNP (or meclizine) would work on restoring the ability of chondrocytes to enlarge and mature (hypertrophy) while statins would work restoring the activity of the IHH-PTHrP axis, which in turn would help chondrocytes to regain their proliferation capacity and leading to a better overall response on bone growth.

Nevertheless, it is clear that this hypothesis needs to be tested in an appropriate pre-clinical model as a proof of concept before any further step. For instance, one could think in a study with four arms: control (sham), CNP-only, statin-only and CNP-statin combo, which would allow researchers to determine if there were any sinergistic effects with that combination.

Combination therapies might also allow a reduction of doses needed to reach the desired effects on bone growth by each of the agents being tested, therefore reducing risks of undesired effects. For example, one known risk associated with TKIs against FGFRs used in cancer is hyperphosphatemia (15). We know that the dose of infigratinib tested for achondroplasia is far lower than those used for cancer (13) so the risk of this kind of side effect would be also lower, but what if the combination with a statin could reduce that risk even more, by allowing an even lower dose of the TKI?

As I said above, appropriate models must be tested for safety and efficacy before any of these ideas could be put in march, but the goal here is to share them and inspire researchers interested in finding solutions for achondroplasia and many other skeletal dysplasias. 

If you follow this blog it is possible that you already know that therapies for FGFR3-related dysplasias might not only be applicable for several other types of bone dysplasias where FGFR3 plays a role in the mechanism of disease, but also for other non FGFR3-related dysplasias as well. One example comes from the developer of vosoritide, which has started a program for some forms of idiopathic short stature (see more info here and here; these links will take you to two pdf presentations. You just have to browse them for achondroplasia. The first one has more details). Another example comes from a study with infigratinib in which the researchers found out that the inhibition of FGFR3 had positive effects in two animal models of severe dysplasias associated with mutations in the sulphate transporter gene, which is also the cause of diastrophic dysplasia (16).

Things are getting better, and although definitive results from all the ongoing and upcoming initiatives will still take a few years to become available, it is reassuring that  not in a distant future many children will be spared of enduring the many medical complications that often occur in bone dysplasias and will enjoy better quality-of-life.

ps. You can find much more information about all strategies briefly reviewed here in other articles of the blog. Try the index page. 



References

1. Yamashita A et al. Statin treatment rescues FGFR3 skeletal dysplasia phenotypes. Nature 2014; 513 (7519):507-11. 

2. Fafilek B et al. Statins do not inhibit the FGFR signaling in chondrocytes. Osteoarthritis Cartilage 2017; (9):1522-30.

3. Ishikawa M et al. The effects of fluvastatin on Indian Hedgehog pathway in endochondral ossification. Cartilage 2019; 22:1947603519862318. doi: 10.1177/1947603519862318. [Epub ahead of print] 

4. Kronenberg HM. Developmental regulation of the growth plate. Nature 2003; 423(6937):332-6.

5. Calvi LM, Schipani E. The PTH/PTHrP receptor in Jansen's metaphyseal chondrodysplasia. J Endocrinol Invest 2000;23(8):545-54.

6. Chen L et al. A Ser(365)-->Cys mutation of fibroblast growth factor receptor 3 in mouse downregulates Ihh/PTHrP signals and causes severe achondroplasia. Hum Mol Genet 2001; 10(5):457-65.

7. Ueda K et al. PTH has the potential to rescue disturbed bone growth in achondroplasia. Bone 2007;41(1):13-8. 

8. Xie Y et al. Intermittent PTH (1-34) injection rescues the retarded skeletal development and postnatal lethality of mice mimicking human achondroplasia and thanatophoric dysplasia. Hum Mol Genet 2012; 21(18):3941-55.

9.
Savarirayan R et al. C-Type Natriuretic Peptide Analogue Therapy in Children with Achondroplasia. N Engl J Med. 2019;381(1):25-35.

10.
Breinholt VM et al. TransCon CNP, a Sustained-Release C-Type Natriuretic Peptide Prodrug, a Potentially Safe and Efficacious New Therapeutic Modality for the Treatment of Comorbidities Associated with Fibroblast Growth Factor Receptor 3-Related Skeletal Dysplasias. J Pharmacol Exp Ther 2019 Sep;370(3):459-71.

11. Lorget F et al. Evaluation of the therapeutic potential of a CNP analog in a Fgfr3 mouse model recapitulating achondroplasia. Am J Hum Genet. 2012;91(6):1108-14.
Free access. 

12. Garcia S et al. Postnatal soluble FGFR3 therapy rescues achondroplasia symptoms and restores bone growth in mice. Sci Transl Med. 2013 Sep 18;5(203):203ra124. Free access.

13. Komla-Ebri D et al. Tyrosine kinase inhibitor NVP-BGJ398 functionally improves FGFR3-related dwarfism in mouse model. J Clin Invest 2016;126(5):1871-84. Free access. 

14. Matsushita M et al. Meclozine facilitates proliferation and differentiation of chondrocytes by attenuating abnormally activated FGFR3 signaling in achondroplasia. PLoS One. 2013 Dec 4;8(12):e81569. Free access.15. Kelly CM et al. A phase Ib study of BGJ398, a pan-FGFR kinase inhibitor in combination with imatinib in patients with advanced gastrointestinal stromal tumor. Invest New Drugs 2019;37(2):282-90. Free access.
 
15.
Nogova L et al. Evaluation of BGJ398, a Fibroblast Growth Factor Receptor 1-3 Kinase Inhibitor, in Patients With Advanced Solid Tumors Harboring Genetic Alterations in Fibroblast Growth Factor Receptors: Results of a Global Phase I, Dose-Escalation and Dose-Expansion Study. J Clin Oncol 2017;35(2):157-65. Free access.

16. Zheng C et al. Suppressing UPR-dependent overactivation of FGFR3 signaling ameliorates SLC26A2-deficient chondrodysplasias. EBioMedicine 2019;40:695-709. Free access.








No comments:

Post a Comment