|
Indian Pediatr 2020;57:
165-171 |
|
Antisense Oligonucleotides: A Unique Treatment Approach
|
Abhinaya V Krishnan and Devendra Mishra
From Department of Pediatrics, Maulana Azad Medical College
(University of Delhi), Delhi-110002
Correspondence to: Dr Devendra Mishra, Professor, Department of
Pediatrics, Maulana Azad Medical College (University of Delhi), Delhi
110 002, India.
Email: [email protected]
|
Synthetic Antisense oligonucleotides
(ASOs) are novel and efficient laboratory tools to regulate the
expression of specific genes, and have only recently come into clinical
use. These are synthetic single-stranded DNA analogs, whose sequence is
complementary to a target nucleotide and alter protein synthesis by
several mechanisms. We herein provide a primer on the topic for
pediatricians, as this group of drugs is likely to see many more drugs
for previously incurable diseases.
Keywords: Duchenne muscular dystrophy,
Eteplirsen, Nusinersen, Spinal muscular atrophy, Treatment.
|
N ucleic acids come in two forms: deoxyribonucleic
acids (DNA) and ribonucleic acids (RNA). RNA has much structural variety
with subtypes like messenger RNA (mRNA, that codes for protein)
non-coding RNAs, transfer RNA (tRNA), ribosomal RNA (rRNA), and long-noncoding
RNAs (lncRNAs) – DNA is a much more stable molecule [1]. Genetic
information from DNA encodes to RNA, ie, transcription, which is
then translated into proteins. Most of the available drugs, such as
small molecules and antibodies target mainly proteins due to their
mechanisms of action and chemical properties. In recent years, the use
of compounds that can bind messenger RNAs (mRNAs) has gained increasing
interest, as inhibition of protein expression can be helpful for
controlling the course of inflammatory and neoplastic diseases. The two
major therapeutic approaches in this field are the antisense oligo-nucleotides
(ASOs) that inhibit mRNA translation, and the oligonucleotides, which
function via RNA inter-ference (RNAi) pathway [2].
Synthetic antisense oligonucleotides are a novel and
efficient laboratory tool to regulate the expression of specific genes.
These are synthetic single-stranded DNA analogs; usually 15-30 base
pairs in length, whose sequence (3' to 5') is antisense and
complementary to the sense sequence of the target nucleotide (mRNA)
hence called antisense oligonucleotides [2]. They selectively bind to
specific pre-messenger ribonucleic acid (pre-mRNA)/mRNA sequences and
alter protein synthesis by several mechanisms. First studied in the late
1970s to inhibit oncogenic viral production [3], further research led to
designing of highly modified ASOs with more targeted delivery,
tolerability, safety, with a prominent role in treating life-threatening
diseases that were previously incurable [4].
Mechanism of Action
The mechanism of action of ASOs may briefly be
summarized in three sequential steps, as follows [5]:
Pre-hybridization phase is the phase in which the
ASO enters the cell, distributes within the cell, to achieve sufficient
concentrations at the target RNA site. The internalization of the ASO
within the cell by carrier protein-mediated endocytosis is a complex
process – further, the ASO needs to escape the cellular endosomal
pathway to reach the target site, which is a rate-limiting process [6].
Hybridization phase is the phase in which ASO
sorts through the cellular nucleic acid sequence space to hybridize to
its target RNA site. This is a complex process that involves
interactions with proteins, such as Ago2, or other cellular components
[5].
Post hybridization phase: After binding to the
mRNA site depending on the chemical design of the ASO, a variety of
events may be induced that alter the target RNA to achieve the desired
pharmacological outcome. There are two main mechanisms: the common
mechanism is by induction of endogenous RNAse H activity (ASO-RNase H)
that cleaves the mRNA-ASO hetero-duplex which leads to degradation of
the target toxic mRNA and leaves the ASO intact. The second mechanism
includes binding to the RNA and causing translational inhibition by
steric hindrance, exon skipping, exon inclusion, destabilization of
pre-mRNA in the nucleus, or targeting the destruction of microsomal RNAs
that control the expression of other genes [2,7]. For example, ASOs can
bind to mRNA structures and prevent the 5’-mRNA cap formation or,
alternatively, they modify the polyadenylation site to prevent mRNA
translation or alter RNA stability. Moreover, ASOs can directly stick to
the mRNA and sterically block the 40S and 60S ribosomal subunits from
attaching or running along the mRNA transcript during translation. Other
ASOs bind on pre-mRNA intron/exon junctions and directly modulate
splicing by masking splicing enhancers and repressor sequences, skipping
exons, or forcing the inclusion of otherwise alternatively spliced exons.
These actions are independent of RNase activity, as with Eteplirsen.
|
Fig. 1 RNAse H independent mechanisms
to prevent mRNA translation by ASO. Step 1. Binds to 5’ cap;
Step 2. Binds to poly A tail; Step 3. Stearic hindrance; and
Step 4. Modifies exon splicing.
|
The mechanism of ASO action is shown in Fig.
1. There are many hurdles to incorporating ASOs in therapeutic use,
because of their mechanism of cellular uptake and action (Box
1). Thus, modifications are needed on the native ASOs to overcome
these disadvantages, and this has led to various versions for clinical
use.
Box I Hurdles to Using Anti-sense
Oligonucleo-tides for Therapeutic Purposes
• Nucleic acids are inherently susceptible to
degradation by endogenous nucleases: ASOs in their native forms
have a very short half-life, even before they are filtered out
through the kidney.
• Unfavorable bio-distribution and
pharma-cokinetic properties: Synthetic ASOs are large
(approximately 30 kD) and highly negatively charged molecules
and thus do not cross vascular endothelium, dense extracellular
matrix and cell and nuclear membranes in order to reach their
intracellular DNA or mRNA targets.
• Off-target effects of ASO may lead to a
devastating adverse reaction.
• Synthetic ASOs can be immunogenic.
• Sub-optimal binding affinity for complementary sequences.
|
First Generation ASOs
These are obtained by replacing one of the
non-bridging oxygen atoms in the phosphate group of nucleotide with
either sulfur groups (phosphorothioates), methyl groups (methylphosphonates)
or amines (posphoroamidates). Phosphorothioate substitution was the
earliest and the most commonly used modification that renders the
inter-nucleotide linkage resistant to nuclease degradation, supports
endogenous RNAse H activity to degrade the target mRNA, improves the
pharmacokinetic characteristics by their binding with plasma proteins
which alter the half-life and increases the availability of ASO to the
target site. The other type is thiophosphoramidate substitution, though
it had many side-effects in animal models and in vitro experiments
[2,8].
Second Generation ASOs
Second generation ASOs were developed to overcome the
shortcomings of first-generation ASOs. These second-generation antisense
agents, contain a Phosphorothioate backbone and replacement of the
2-hydroxyl by many different groups but most commonly by 2’-O-methoxy (OMe),
2’-O-methoxyethyl (MOE), and locked nucleic acid (LNA). 2’-OMe
modifications are commonly used in a ‘gapmer’ design, which is a
chimeric oligo nucleotide comprising a DNA sequence core with flanking
2’-MOEnucleotides.2-Methoxyethyl is probably the most common one used in
trials. Their mechanism of action is RNAse independent. The advantages
of these ASOs are improved nuclease resistance, target-binding affinity,
increased thermal stability of complementary hybridization, encourages
tighter binding and allowing use of shorter oligonucleotides [2,4,7].
Third Generation ASOs
These are modified ASOs which help in their
intracellular uptake and effective delivery to the target. These ASOs
are covalently bound to a carrier or ligand, such as lipid particles,
liposomes, nanoparticles, and, more recently, the sugar N-acetyl
galactosamine to enhance safer delivery to the target site [2,9,10].
Pharmacokinetics
The rationale for understanding the pharmacokinetics
of ASOs is to correlate effective dose with clearance rates to optimize
effectiveness while reducing potentially harmful side-effects [6]. The
route of administration of these drugs is parenteral – intravenous,
subcutaneous and intrathecal because oral bioavailability is less than
1% [11].
Absorption: The pharmacokinetic properties of
oligonucleotides following parenteral administration are predominantly
studied in phosphorothioate oligo-nucleotides, which after parenteral
administration, bind to plasma proteins at >90% and transfer rapidly
from blood to tissues with a distribution half-life of 1-2 hour. By 12
hour after dosing, <1% of the administered dose remains in circulation.
At the same time, <5% of the administered dose is recovered in urine and
feces over the first day, and this broad distribution to tissues causes
the rapid disappearance of compound from blood. The highest tissue
accumulation has been observed in kidney, liver, spleen, lymph nodes,
adipocytes and bone marrow. In marked contrast, those oligonucleotides
lacking charge and/or binding to plasma proteins (peptide nucleic acids,
morpholinos) are rapidly cleared from plasma, with substantially higher
excretion in urine in the first day resulting in lower overall tissue
accumulation and ultimate target bioavailability [12].These drugs do not
cross the blood-brain barrier and poorly distribute in skeletal muscle,
heart, and lung.
Distribution: Although distribution out of plasma
to tissue is rapid, the ultimate distribution to the active site within
cells following a single dose is maximally realized at 24-48 h. Thus,
the onset of action for antisense oligonucleotides is slower than the
distribution out of plasma, which is explained due to the intracellular
uptake and the kinetics for transport from the cell surface to the
nucleus. Once distributed to cells, they are slowly cleared with tissue
half-life ranging from 2-4 weeks [11,13].
Metabolism: These drugs are metabolized by
endonucleases, which are expressed in most tissues hence liver
dysfunction does not appear to affect their action. These drugs are not
substrates for cytochrome P450 enzymes, and hence a very low drug-drug
interaction is known [11].
Excretion: Excretion is predominantly renal and
fecal; biliary uptake is minimal.
Clinical Use
Chemically modified 1 st
and 2nd generation ASOs have
generated a new hope in the management of devastating neuromuscular and
few other diseases such as Duchenne Muscular Dystrophy (DMD), Spinal
Muscular atrophy (SMA), Myotonic dystrophy, familial
hypercholester-olemia, Amyotrophic lateral sclerosis, factor IX
thrombosis, Huntington chorea and peripheral neuropathies [6,14,15].
Nearly two decades after their advent, the US FDA approved the first ASO
for therapeutic use in 1998 (Fomiversan). We have come a long way since
then (Box II), with approvals coming for two oligonucleotides in
2016 for use in DMD and SMA [16]. These modify the disease make-up and
progression by an effect at the gene (mRNA) level, hence provide immense
scope for complete cure or at least a better quality of life. A
multitude of other drugs is under development or in trials for the
treatment for various other diseases, including cancers.
Box II Available Anti-sense
Oligonucleotides for Clinical Application
Nusinersen: Spinal muscular atrophy
Eteplirsen:
Duchenne Muscular Dystrophy
Fomiversan: Cytomegalovirus retinitis
secondary to AIDS
Pegabtinib: Age-related macular
degeneration of the retina
Mipomersen: Familial hypercholesterolemia
Defibrotide: Severe HVOD following
high-dose chemotherapy and autologous BMT
Patisiran: Transthyretin amyloidosis
HVOD: hepatic veno-occlusive disease; BMT: bone marrow
transplantation
|
Individual Drugs
Fomiversan: This was the first ASO to be
approved by the FDA in 1998, used clinically in CMV retinitis secondary
to AIDS [17]. At that time, there was a high unmet need for
anti-cytomegalovirus retinitis drugs; however, subsequently, due to the
development of high-activity antiretroviral therapy (HAART), the number
of CMV cases dramatically decreased [16], and its use has declined.
Pegabtinib: It was approved by the FDA in
2004 to treat Age-related macular degeneration (AMD) of the retina
[16,18]. This is caused by the VEGF-stimulated growth of blood vessels (neovascularization)of
the choroid of the eye leading to macular blindness. This molecule
prevents the binding of VEGF to VEGFR receptors. However, with the
advent of cheaper and better alternatives like bevazimumab, its use is
on the downswing [16].
Eteplirsen: It was approved by the FDA for DMD in
2016, which was a remarkable step in the future of treatment for this
disease. DMD is caused by mutations within the dystrophin gene that
disrupt the reading frame or cause premature termination of protein
synthesis [19].This was also the first approved exon skipping ASO to be
used in humans [14]. This molecule is a 30-nucleotide
phosphoro-diamidatemorpholino oligomer type third-generation ASO that
hybridizes to exon 51 of DMD Pre-mRNA and causes it to be skipped during
splicing; this corrects the translational reading frame in certain DMD
gene deletions, resulting in the production of shortened but functional
dystrophin protein similar to what is found in Becker’s muscular
dystrophy. It is effective only in DMD caused by exon 51 deletion (13%),
which, however, is supported by a limited trial including 12 patients.
High costs limit its widespread use [16].
The FDA approval of eteplirsen has been controversial
due to the poor generalizability of the trial. Mendell, et al.
[20] compared the three-year progression of the disease and its effect
on ambulation in those receiving Eteplirsen and compared them to
historic controls (n=13). Ambulatory DMD patients aged between
7-13 years, amenable to exon 51 skipping who were able to walk between
180-440 m on 6-Minute Walk Test and on stable corticosteroids for 24
weeks were randomized into three cohorts (each n=4) viz,
placebo, Eteplirsen 30 mg/kg/week and 50 mg/kg/week. Later all received
the drug in an open labeled trial. Six minute walk test and pulmonary
function tests were done at baseline, 6, 1 and 24 months. A significant
advantage on the walk test and a lower incidence of loss of ambulation
(16%) were seen in the eteliprisen group in comparison to matched
historic controls (46.2%).
Nusinersen: This is the other ASO approved by FDA in
2016 for spinal muscular atrophy (SMA). SMA is most frequently caused by
a homozygous deletion or mutation within the Survival motor neuron1
(SMN1) gene located on chromosome 5. Homozygous deletion of SMN1
exon 7 is confirmatory for the diagnosis of SMA. SMN1 gene
codes for the ubiquitously expressed ‘survival motor neuron’ (SMN)
protein, which is essential for the maintenance of motor neurons. Humans
have one more paralogous SMN1 gene copy, referred to as SMN2,
which differs from SMN1 only by a cytosine-to-thymine mutation in
exon 7 of the SMN2 gene,which leads to alternative splicing
processes with the consequence that exon 7 is omitted from the majority
of SMN2 transcripts [6,14,21]. Yet a small amount (approximately
10%) of functional SMN protein is expressed via the SMN2
gene. This allows for partial compensation of the lost SMN1 exon 7 by
SMN2 synthesis. Clinical phenotype is hence related to the number of
SMN2 copies [21]. Nusinersen is a 2'-OMe phosphorothioate ASO that
induces the inclusion of exon 7 in the SMN1 and SMN2 mRNA
by targeting and blocking an intron 7 internal splice site and producing
functional SMN protein [16]. Nusinersen is now indicated in infants with
types 1, 2, and 3 SMA. Nusinersen has to be given intrathecally as it
does not cross the blood-brain barrier. The mean plasma terminal
elimination half-life is 63-87 days, and the mean CNS terminal
elimination half-life is 135-177 days. A fixed-dose is recommended
because dose-related toxicity has not been demonstrated. The renal route
of elimination is applicable for nusinersen and its inactive metabolites
[22]. It is supplied as 12 mg/5 mL preservative-free solution and given
in a standard dose of 12 mg on days 0, 14, 28, and 63 in two-weekly
intervals followed by repeated applications in 4-month intervals
[22,23].
Nusinersen is probably the most promising ASO
manufactured which could modify the outcome and mortality in infants
with SMA. A randomized, double-blind, sham-controlled trial by Finkel,
et al. [23] in 2017 proved the same. In this trial 122 infants
who were less than 7 months of age at the time of screening, having
onset of symptoms from less than 6months of age with a confirmed
mutation in the SMN1 gene with two copies of SMN2 gene
were randomized in 2:1 ratio. 81 were to receive the drug and 41 the
sham injections. Nusirensen was injected intrathecally 12 mg/ adjusted
according to CSF volume on days on days 1, 15, 29, and 64 and
maintenance doses on days 183 and 302. The primary endpoints were the
motor-milestone response defined according to results on the Hammersmith
infant neurological examination and event-free survival which was the
time to death or the use of permanent assisted ventilation. Secondary
end-points were overall survival and subgroup analyses of event-free
survival according to disease duration at screening. Due to a very
significant result during the interim analysis which showed a motor
milestone response of 41% in test and 0 in control, the trial was
prematurely terminated and everyone received the drug. In the final
analysis, 51% in the test group showed a motor response. Risk of death
or use of permanent assisted ventilation was lower in nusinersen group
47% [hazard ratio (95% CI) 0.53 (0.32-0.89), P = 0.005]. Also the
likelihood of event-free survival and overall survival were
significantly more in the test group – infants with the shortest disease
duration prior to drug administered had the highest likelihood of
event-free survival [23].
In another multi-center, double-blind,
sham-controlled phase 3 trial by Mercuri, et al. [24] in 2018,
126 children with SMA who had symptom onset after 6 months of age were
randomly assigned in a 2:1 ratio, to undergo intrathecal administration
of nusinersen at a dose of 12 mg (nusinersen group) or a sham procedure
(control group) on days 1, 29, 85, and 274. The primary endpoint was the
least squares mean change from baseline in the Hammersmith functional
motor scale-expanded (HFMSE) score at 15 months of treatment; Secondary
endpoints included the percentage of children with a clinically
meaningful increase from baseline in the HFMSE score (³3
points), an outcome that indicates improvement in at least two motor
skills. This trial was also prematurely terminated as the pre-specified
interim analysis showed a least-squares mean increase (by 4.0 points)
from baseline to month 15 in the HFMSE score in the nusinersen group and
a least-squares mean decrease in the control group (by-1.9 points) with
a significant between-group difference favoring nusinersen
(least-squares mean difference in change, 5.9 points; 95% confidence
interval, 3.7 to 8.1; P<0.001). Results of the final analysis
were consistent with results of the interim analysis in which 57% of the
children in the nusinersen group as compared with 26% in the control
group had a significant increase from baseline to month 15 in the HFMSE
score of at least 3 points, and the overall incidence of adverse events
was similar in the nusinersen group and the control group (93% and 100%,
respectively) [24]. Patients from both the above trials were later
enrolled in SHINE, an open-label extension trial. SHINE is structured to
evaluate the effects of longer treatment with nusinersen with respect to
motor function and quality of life which is still ongoing [25]. The
results from the above studies infer that nusinersen could produce
positive changes in the SMA patient’s clinical course however the
results may not be generalizable to all patients with SMA because only
patients with types 1, 2, and 3 were included in the trials.
Additionally, the long-term benefit and safety is not known at this
time. It appears that nusinersen may have more benefit in patients who
are younger with less severe disease or less comorbidity.
The most common adverse reactions of nusinersen are
(>10%) upper and lower-respiratory tract infections (39%-43%),
atelectasis (14%), constipation (30%), headache (50%), back pain (41%),
and post-lumbar puncture syndrome (41%). Its high cost and route of
administration are its major limitations [22,26].
Mipomersen: Mipomersen is the first FDA-approved
systemically-delivered ASO in 2013 for familial hypercholesterolemia
[16]. This is a disease caused loss of function mutations in both
LDL-receptor genes which results in the reduced liver uptake of plasma
LDL cholesterol leading to a very high plasma concentration of
low-density lipoprotein. The core protein of the LDL particle is
apolipoprotein B [27]. Mipomersen is targeted to the coding region of
the apoB mRNA which effectively reduces plasma LDL and cholesterol
levels with less deleterious effects on HDL. Side effects are injection
site reactions and liver toxicity [28].
Other drugs include Defibrotide for severe hepatic
veno-occlusive disease occurring after high dose chemotherapy and
autologous bone marrow transplantation [29], and Revusiran, Patisiran
and Inotersen for Transthyretin amyloidosis [30-32].
Adverse Drug Reactions
Oligonucleotides are prone to a diverse array of
off-target interactions because of their size, negative charge, and
potential to be synthetic [4]. Thus, despite a good overall safety
profile, a few adverse reactions are encountered due to their off-target
effects [33]. However, the number of studies and the sample size
included are too small to determine the general side effect profile, the
dose relationship and class effect for these drugs.
Binding of nucleic acid to cell surface proteins or
to proteins inside cells -oligonucleotides can bind serine/threonine
protein kinase PKR or Toll-like receptors (TLRs) and activate the innate
immune response/alternate complement pathway. These can also bind to
dRNA and DNA by complementary base-pairing. Vasculitis or
glomerulonephritis are rare manifestations of immune activation [9]. All
ASOs and dsRNAs will be at least partially complementary to DNA or RNA
sequences inside cells that are not their intended targets and thus can
modify the actions of genes on these mRNA/DNA, which could be harmful
[4].
Thrombocytopenia is one of the most common
side-effects seen [34]. Two forms of thrombocytopenia are studied. The
more common form is milder, transient and dose-dependent wherein
bleeding episodes are very rare. In humans, thrombocytopenia has been
reported in cancer studies with a number of first-generation ASOs and
occasionally with second-generation ASOs such as Mipomersen [35]. Other
is rarer and severe form with bleeding episodes [36].
Cost
Despite remarkable progress in the development of
ASOs for clinical use, the cost remains a major limitation for
widespread use. For Eteplirsen, the costs were estimated at US$ 57,600
(INR 38,59,000) per month [37]. For Nusinersen, the cost of treatment of
a patient with SMA amounts to US$ 750,000 (INR 5,02,50,000) for the
first year, and half of that every year afterward [38,39].Thus the high
costs are a major setback to any healthcare system plus the poor
validity of the clinical trials in showing efficacy and adverse effects
do not give a definitive risk-benefit advantage.
Conclusions
The invention of ASOs represents a therapeutic
milestone in those diseases for which we do not have a definitive cure
by modifying the disease pathways. ASOs are under development or have
already been tested in clinical trials for the treatment of many other
diseases like myotonic dystrophy, Huntington chorea, Amyotrophic lateral
sclerosis, Hemophilia A, Hereditary neuropathies. There have been some
demands from individual patients and patient-support groups in LMICs
(including India) to permit use of these drugs through publicly-funded
programs. Although these drugs have good safety and tolerability, their
high cost, route of administration, localized target (not applicable to
all variants of disease), and lack of significant clinical trials
describing mechanism of action, target sites, efficacy, and side effects
are the major limitations. Hence, further research is required to better
elucidate these important aspects, before wide-spread use would be a
possibility.
Contributors: Both authors were involved in
planning the manuscript and review of literature. Initial draft: AVK;
Final manuscript: AVK, DM. Both authors approved the final manuscript.
Funding: None; Competing interest: None
stated.
References
1. Chery J. RNA therapeutics: RNAi and antisense
mechanisms and clinical applications. Postdoctoral Journal.
2016;4:35-50.
2. Verma A. Recent advances in antisense
oligonucleotide therapy in genetic neuromuscular diseases. Ann Indian
Acad Neurol. 2018;21:3-7.
3. Stephenson ML, Zamecnik PC. Inhibition of Rous
sarcoma viral RNA translation by a specific oligo-deoxyribo-nucleotide.
Proc Natl Acad Sci USA.1978; 75:285-8.
4. Shen X, Corey DR. Chemistry, mechanism and
clinical status of antisense oligonucleotides and duplex RNAs. Nucleic
Acids Res. 2018;46:1584-600.
5. Crooke ST, Wang S, Vickers TA, Shen W, Liang X.
Cellular uptake and trafficking of antisense oligonucleotides. Nat
Biotechnol. 2017;35:230-7.
6. Miller CM, Harris EN. Antisense oligonucleotides:
Treat-ment strategies and cellular internalization. RNA Dis. 2016;3:4.
7. Dias N, Stein CA. Antisense oligonucleotides:
Basic concepts and mechanisms. Mol Cancer Ther. 2002;1:347-55.
8. Gustincich S, Zucchelli S, Mallamaci A. The Yin
and Yang of nucleic acid-based therapy in the brain. Prog Neurobio.
2017;155:194-211.
9. Chi X, Gatti P, Papoian T. Safety of antisense
oligonucleotide and siRNA-based therapeutics. Drug Discov Today.
2017;22:823-33.
10. Prakash TP, Graham MJ, Yu J, Carty R, Low A,
Chappell A, et al. Targeted delivery of antisense
oligonucleotides to hepatocytes using triantennary N-acetyl
galactosamine improves potency 10-fold in mice. Nucleic Acids Res.
2014;42:8796-807.
11. Geary RS. Antisense oligonucleotide
pharmacokinetics and metabolism. Expert Opin Drug Metab Toxicol. 2009;5:
381-91.
12. Braasch DA, Paroo Z, Constantinescu A, Ren G, Oz
OK, Mason RP, et al. Biodistribution of phosphodiester and
phosphorothioate siRNA. Bioorg Med Chem Lett. 2004;14:1139-43.
13. Bennett CF, Baker BF, Pham N, Swayze E, Geary RS.
Pharmacology of antisense drugs. Annu Rev Pharmacol Toxicol.
2017;57:81-105.
14. Evers MM, Toonen LJA, van Roon-Mom WMC.
Anti-sense oligonucleotides in therapy for neuro-degenerative disorders.
Adv Drug Deliv Rev. 2015;87:90-103.
15. Wurster CD, Ludolph AC. Antisense
oligonucleotides in neurological disorders. TherAdvNeurolDisord.
2018;11: 1-19.
16. Stein CA, Castanotto D. FDA-approved
oligonucleotide therapies in 2017. Mol Ther. 2017;25:1069-75.
17. Vitravene Study Group. A randomized controlled
clinical trial of intra-vitreous fomivirsen for treatment of newly
diagnosed peripheral cytomegalovirus retinitis in patients with AIDS. Am
J Ophthalmol. 2002;133:467-74.
18. Gragoudas ES, Adamis AP, Cunningham ET Jr,
Feinsod M, Guyer DR;VEGF Inhibition Study in Ocular Neovascularization
Clinical Trial Group. Pegaptanib for neovascular age-related macular
degeneration. N Engl J Med. 2004;351:2805-16.
19. Koo T, Wood MJ. Clinical trials using antisense
oligonucleotides in Duchenne muscular dystrophy. Hum Gene Ther.
2013;24:479-88.
20. Mendell JR, Goemans N, Lowes LP, Alfano LN, Berry
K, Shao J, et al. Longitudinal effect of eteplirsen versus
historical control on ambulation in Duchenne muscular dystrophy:
Eteplirsen in DMD. Ann Neurol. 2016;79: 257-71.
21. Castro D, Iannaccone ST. Spinal muscular atrophy:
therapeutic strategies. Curr Treat Options Neurol. 2014; 16:316.
22. Spinraza [package insert]. Cambridge, MA: Biogen
Inc;2017.
23. Finkel RS, Mercuri E, Darras BT, Connolly AM,
Kuntz NL, Kirschner J, et al. Nusinersen versus sham control in
infantile-onset spinal muscular atrophy. N Engl J Med. 2017;377:1723-32.
24. Mercuri E, Darras BT, Chiriboga CA, Day JW,
Campbell C, Connolly AM, Lannaccone ST, et al; CHERISH Study
Group. Nusinersen versus sham control in later-onset spinal muscular
atrophy. N Engl J Med. 2018;378:625-35.
25. National Library of Medicine. 12 Studies found
for: nusinersen. Available from: https://clinicaltrials.gov/ct2/results?cond=&term=nusinersen&cntry=&state=&city=&dist.
Accessed March 17, 2018.
26. Hoy SM. Nusinersen: first global approval. Drugs.
2017;77:473-79.
27. McGowan MP, Tardif J-C, Ceska R, Burgess LJ,
Soran H, Gouni-Berthold I, et al. Randomized, placebo-controlled
trial of mipomersen in patients with severe hypercholesterolemia
receiving maximally tolerated lipid-lowering therapy. PLoS One.
2012;7:e49006.
28. Thomas GS, Cromwell WC, Ali S, Chin W, Flaim JD,
Davidson M. Mipomersen, an apolipoprotein B synthesis inhibitor, reduces
atherogenic lipoproteins in patients with severe hypercholesterolemia at
high cardiovascular risk. J Am Coll Cardiol. 2013;62:2178-84.
29. Richardson PG, Riches ML, Kernan NA, Brochstein
JA, Mineishi S, Termuhlen AM, et al. Phase 3 trial of defibrotide
for the treatment of severe veno-occlusive disease and multi-organ
failure. Blood. 2016;127:1656-65.
30. Ackermann EJ, Guo S, Benson MD, Booten S, Freier
S, Hughes SG, et al. Suppressing transthyretin production in
mice, monkeys and humans using 2nd-generation antisense oligonucleotides.
Amyloid. 2016;23:148-57.
31. Zimmerman TS, Karsten V, Chan A, Chiesa J, Boyce
M, Bettencourt BR et al. Clinical proof of concept for a novel
hepatocyte-targeting GalNAc-siRNA conjugate. Mol Ther. 2017;25:71-8.
32. Suhr OB, Coelho T, BuadesJ, Pouget J, Conceicao
I, Berk J, et al. Efficacy and safety of Patisiran for familial
amyloidotic polyneuropathy: A phase II multi-dose study. Orphanet J Rare
Dis. 2015;10:109.
33. Winkler J, Stessl M, Amartey J, Noe CR.
Off-target effects related to the phosphorothioate modification of
nucleic acids. Chem Med Chem. 2010;5:1344-52.
34. Frazier KS. Antisense oligonucleotide therapies:
the promise and the challenges from a toxicologic pathologist’s
perspective. Toxicol Pathol. 2015;43:78-89.
35. KYNAMRO – Mipomersen sodium injection, solution.
Official Label: http://
www.accessdata.fda.gov/drugsatfda_docs/label/2016/203568s008lbl.pdf.
Accessed 10 May, 2019.
36. Crooke ST, Baker BF, Kwoh TJ, Cheng W, Schulz DJ,
Xia S, et al. Integrated safety assessment of 22 -o-methoxyethyl
chimeric antisense oligonucleotides in nonhuman primates and healthy
human volunteers. Mol Ther. 2016;24:1771-82.
37. Dalakas MC. Gene therapy for Duchenne muscular
dystrophy: balancing good science, marginal efficacy, high emotions and
excessive cost. Ther Adv Neurol Disord. 2017;10:293-6.
38. Gellad WF, Kesselheim AS. Accelerated approval
and expensive drugs - A challenging combination. N Engl J
Med.2017;376:2001-04.
39. Editorial. Treating rare disorders: Time to act
on unfair prices. Lancet Neurol. 2017;16:761.
|
|
|
|