Acessibilidade / Reportar erro

Gene Silencing Therapeutics in Cardiology: A Review Article

Abstract

Therapeutics that inhibit enzymes, receptors, ion channels, and cotransporters have long been the mainstay of cardiovascular medicine. Now, oligonucleotide therapeutics offer a modern variation on this paradigm of protein inhibition. Rather than target a protein, however, small interfering ribonucleic acids and antisense oligonucleotides target the messenger RNA (mRNA) from which a protein is translated. Endogenous, cellular mechanisms enable the oligonucleotides to bind a selected sequence on a target mRNA, leading to its degradation. The catalytic nature of the process confers an advantage over the stoichiometric binding of traditional small molecule therapeutics to their respective protein targets. Advances in nucleic acid chemistry and delivery have enabled development of oligonucleotide therapeutics against a wide range of diseases, including hyperlipidemias and hereditary transthyretin-mediated amyloidosis with polyneuropathy. While most of these therapeutics were initially designed for rare diseases, recent clinical trials highlight the potential impact of oligonucleotides on more common forms of cardiovascular disease.

Keywords:
Cardiovascular Disease; Gene Silencing; RNA Interference, Oligonucleotides; Antisense; Amyloidosis

Introduction

Almost every drug targets a protein, the last step in the flow of genetic information from DNA through transcription and translation. However, as early as 1978, Zamecnik envisioned targeting the prior step and demonstrated that a 13-base oligonucleotide complementary to a sequence in the Rous sarcoma virus inhibited translation of viral messenger RNA (mRNA) and the consequent oncogenic transformation of cells in culture. Furthermore, a chemical modification of the oligonucleotide prolonged its efficacy, presumably by slowing its degradation.11. Stephenson ML, Zamecnik PC. Inhibition of Rous sarcoma viral RNA translation by a specific oligodeoxyribonucleotide. Proc Natl Acad Sci U S A. 1978;75(1):285-8.,22. Zamecnik PC, Stephenson ML. Inhibition of Rous sarcoma virus replication and cell transformation by a specific oligodeoxynucleotide. Proc Natl Acad Sci U S A. 1978;75(1):280-4. This latter observation foreshadowed the chemistry that enables the remarkable duration of action of oligonucleotide therapeutics today.

Silencing the expression of a gene by targeting its mRNA via an oligonucleotide expands the therapeutic armamentarium, overcoming major hurdles such as the identification of “druggable” proteins and therapeutics that bind them. Furthermore, target proteins may not have a suitable drug binding site, or binding to off-target proteins may cause undesirable side effects. Oligonucleotides can circumvent these limitations and expedite the drug discovery process. With a target protein and the sequence of its coding gene, an antisense oligonucleotide (ASO) or a small interfering ribonucleic acid (siRNA) can be designed to bind to a complementary sequence on an mRNA with exquisite specificity. Different cellular mechanisms are co-opted to degrade the mRNA: RNase H1 (ASO) or RNA inhibition (RNA interference [RNAi]). The physiologic functions of RNase H1 and RNAi include antiviral defense,33. Broecker F, Moelling K. Evolution of immune systems from viruses and transposable elements. Front Microbiol. 2019;10:51. and the RNAi pathway is fundamental to controlling gene expression in diverse eukaryotic species.44. Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K, Tuschl T. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature. 2001;411(6836):494-8.,55. Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature. 1998;391(6669):806-11.

ASOs are single-stranded oligonucleotides of 15-20 bases, designed to be complementary (antisense) to a target site on the mRNA transcript,66. Cook PD. Antisense medicinal chemistry. In: Crooke ST, ed. Antisense research and application. Vol 131. Switzerland: Springer Nature; 1998:51-101.,77. Chi X, Gatti P, Papoian T. Safety of antisense oligonucleotide and siRNA-based therapeutics. Drug Discov Today. 2017;22(5):823-33. to which they bind via Watson–Crick base pairing. The ASO chemical modification pattern determines whether the target mRNA is degraded or processed. The most widely used mechanism involves an endogenous nuclease, RNase H1, which recognizes chimeric DNA/RNA duplexes formed by the ASO and target mRNA (Figure 1A). Hence the ASO needs to contain at least 8-10 deoxyribonucleotide residues in the center, which can be flanked by other chemically modified residues. After recognizing? the ASO/mRNA duplex, RNase H1 cleaves the mRNA, thereby reducing production of the target protein.

Figure 1
ASO and RNAi mechanisms for cleavage of target mRNA.

siRNAs are double-stranded RNAs, 19-25 base pairs in length, which utilize the highly conserved natural RNAi pathway. The siRNAs are designed to recruit and bind to an enzyme complex called RNA-induced silencing complex (RISC) to mediate degradation of their respective mRNA targets.44. Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K, Tuschl T. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature. 2001;411(6836):494-8.,55. Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature. 1998;391(6669):806-11. During the loading process into RISC, the 2 strands of the siRNAs are separated (Figure 1B). The sense “passenger” strand is ejected, and the antisense “guide” strand remains loaded and “guides” the RISC to a complementary site on the target mRNA, where it binds through Watson–Crick base pairing.88. Nykänen A, Haley B, Zamore PD. ATP requirements and small interfering RNA structure in the RNA interference pathway. Cell. 2001;107(3):309-21. After cleaving the mRNA, the antisense strand remains bound to the enzyme; thus, in the catalytic action of siRNAs, a single molecule serves to cleave a large number of target mRNAs.

In this review, key technical advances in the evolution of oligonucleotide therapeutics, including ASOs and siRNAs approved or in late-stage clinical development for cardiovascular diseases, are discussed.

Methods

Based on standard review methods, a comprehensive literature search of the PubMed database was conducted to find relevant published articles on gene silencing in cardiology. Search terms were gene+silencing+cardiac (2157 results), RNAi+cardiac (491), and antisense+oligonucleotide+cardiac (1131). Adding “clinical” and/or “phase” refined searches to include only therapeutics in, or previously in, clinical trials, resulting in 78 publications under consideration. Only English language articles were reviewed, no date limits were specified, and searches were undertaken up to July 20, 2020. Relevant publications identified from references of the retrieved articles were also included. Up-to-date information on clinical trials of siRNA and ASO therapeutics in cardiology was sought on the website https://clinicaltrials.gov.

Oligonucleotide therapeutics: evolution of a novel class of medicines

Oligonucleotide therapeutics, with ASOs and siRNAs representing the most prominent gene silencing technologies, have emerged as a new generation of precision medicines for an increasing number of diseases with high unmet medical needs, including cardiovascular diseases. However, unmodified oligonucleotides lack drug-like properties, and decades were spent optimizing their chemistry and developing advanced drug delivery systems that were required for these molecules to become drugs.

Oligonucleotide therapeutics are rapidly degraded and eliminated without the chemical modifications that assure stability and mediate delivery to the intended tissue. Ubiquitous nucleases hydrolyze the bond between phosphodiester groups in DNA/RNA and the sugar moiety of each base, thus degrading nucleic acids. A phosphorothioate modification (Figure 2A) protects the phosphodiester bond in ASOs and siRNAs against enzymatic hydrolysis99. Dowdy SF. Overcoming cellular barriers for RNA therapeutics. Nat Biotechnol. 2017;35(3):222-9. and improves delivery of ASOs by increasing hydrophobicity and improving protein binding. Formation of ASO-protein complexes increases the ASO’s half-life in circulation and reduces urinary excretion to facilitate distribution (predominantly) to liver, spleen, and kidney. While a phosphorothioate backbone confers advantages, it also elicits unwanted interactions with proteins, e.g., the platelet-specific glycoprotein receptor VI or complement, which may mediate thrombocytopenia or glomerulonephritis associated with ASOs.77. Chi X, Gatti P, Papoian T. Safety of antisense oligonucleotide and siRNA-based therapeutics. Drug Discov Today. 2017;22(5):823-33.

Figure 2
(A) Commonly used chemical modifications and (B) clinically validated delivery systems for oligonucleotide therapeutics.

Chemical modifications of sugar moieties in an ASO or siRNA, such as 2’-O-methoxyethyl (2’-MOE), 2’-O-methyl (2’-O-Me), and 2’-fluoro (2’-F) (Figure 2A), are commonly incorporated to increase stability against nucleases, enhance avidity for the target mRNA, and mitigate any innate immune response.99. Dowdy SF. Overcoming cellular barriers for RNA therapeutics. Nat Biotechnol. 2017;35(3):222-9. Of note, 2’-F and 2’-O-Me substitutions along the entire length of an siRNA (Figure 2A) can dramatically increase potency and duration of action.1010. Foster DJ, Brown CR, Shaikh S, Trapp C, Schlegel MK, Qian K, et al. Advanced siRNA designs further improve in vivo performance of GalNAc-siRNA conjugates. Mol Ther. 2018;26(3):708-17.,1111. Nair JK, Willoughby JL, Chan A, Charisse K, Alam MR, Wang Q, et al. Multivalent N-acetylgalactosamine-conjugated siRNA localizes in hepatocytes and elicits robust RNAi-mediated gene silencing. J Am Chem Soc. 2014;136(49):16958-61.

Alongside advances in nucleic acid chemistry, highly effective drug delivery platforms were developed. Encapsulation of an siRNA in a lipid nanoparticle (LNP) represented a significant early advance in systemic delivery (Figure 2B). Demonstrating proof-of-concept, an LNP-siRNA formulation reduced hepatic apolipoprotein B (ApoB) mRNA and levels of ApoB and cholesterol in nonhuman primates.1212. Zimmermann TS, Lee AC, Akinc A, Bramlage B, Bumcrot D, Fedoruk MN, et al. RNAi-mediated gene silencing in non-human primates. Nature. 2006;441(7089):111-4. Second-generation LNPs showed approximately 100-fold improved potency in mouse models.1313. Jayaraman M, Ansell SM, Mui BL, Tam YK, Chen J, Du X, et al. Maximizing the potency of siRNA lipid nanoparticles for hepatic gene silencing in vivo. Angew Chem Int Ed Engl. 2012;51(34):8529-33. The improvement was translated to humans, leading to the first siRNA therapeutic, patisiran, for patients with hereditary transthyretin-mediated (hATTR) amyloidosis with polyneuropathy.1414. Akinc A, Maier MA, Manoharan M, Fitzgerald K, Jayaraman M, Barros S, et al. The Onpattro story and the clinical translation of nanomedicines containing nucleic acid-based drugs. Nat Nanotechnol. 2019;14(12):1084-7.,1515. Coelho T, Adams D, Silva A, Lozeron P, Hawkins PN, Mant T, et al. Safety and efficacy of RNAi therapy for transthyretin amyloidosis. N Engl J Med. 2013;369(9):819-29.

LNPs effectively deliver siRNAs to the liver, but manufacturing is complicated, and administration is intravenous. A simpler solution targets ASOs and siRNAs to cell surface receptors via a conjugated ligand that triggers endocytosis. A current strategy involves conjugation to an N-acetylgalactosamine (GalNAc) ligand (Figure 2B), which binds the asialoglycoprotein receptor (ASGPR). The ASGPR recognizes N-acetylgalactosamine residues on glycoproteins and is highly expressed on hepatocytes.1616. Ashwell G, Morell AG. The role of surface carbohydrates in the hepatic recognition and transport of circulating glycoproteins. Adv Enzymol Relat Areas Mol Biol. 1974;41(0):99-128.,1717. Biessen EA, Beuting DM, Roelen HC, van de Marel GA, van Boom JH, van Berkel TJ. Synthesis of cluster galactosides with high affinity for the hepatic asialoglycoprotein receptor. J Med Chem. 1995;38(9):1538-46. High-affinity, multivalent GalNAc ligands suitable for subcutaneous administration have been developed for ASOs and siRNAs.1111. Nair JK, Willoughby JL, Chan A, Charisse K, Alam MR, Wang Q, et al. Multivalent N-acetylgalactosamine-conjugated siRNA localizes in hepatocytes and elicits robust RNAi-mediated gene silencing. J Am Chem Soc. 2014;136(49):16958-61.,1818. 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(13):8796-807.

Chemically modified nucleic acids and GalNAc ligands have greatly expanded the potential of oligonucleotide therapeutics for diseases that can be treated by suppressing hepatic synthesis of a protein. The combination of stabilized siRNAs with a high-affinity GalNAc ligand, termed enhanced stabilization chemistry (ESC) conjugates, has enabled potent and durable gene silencing in the liver with infrequent administration at low doses.1111. Nair JK, Willoughby JL, Chan A, Charisse K, Alam MR, Wang Q, et al. Multivalent N-acetylgalactosamine-conjugated siRNA localizes in hepatocytes and elicits robust RNAi-mediated gene silencing. J Am Chem Soc. 2014;136(49):16958-61. The first GalNAc-siRNA conjugate approved was givosiran for the treatment of acute intermittent porphyria.1919. Balwani M, Sardh E, Ventura P, Peiró PA, Rees DC, Stölzel U, et al. Phase 3 trial of RNAi therapeutic givosiran for acute intermittent porphyria. N Engl J Med. 2020;382(24):2289-301. The safety of the GalNAc-siRNA platform has been established in large clinical trials of inclisiran, discussed below.2020. Raal FJ, Kallend D, Ray KK, Turner T, Koenig W, Wright RS, et al. Inclisiran for the treatment of heterozygous familial hypercholesterolemia. N Engl J Med. 2020;382(16):1520-30.,2121. Ray KK, Wright RS, Kallend D, Koenig W, Leiter LA, Raal FJ, et al. Two phase 3 trials of inclisiran in patients with elevated LDL cholesterol. N Engl J Med. 2020;382(16):1507-19. Several other GalNAc-ASO and -siRNA conjugates for cardiovascular indications are currently in clinical trials (Table 1).

Table 1
Oligonucleotide therapies in mid- to late-stage development or approved for cardiovascular indications

Gene silencing in hyperlipidemias

Loss-of-function variants of proprotein convertase subtilisin/kexin type 9 (PCSK9), angiopoietin-like protein-3 (ANGPTL3), APOC3 (ApoC-III), and LPA (ApoA) are associated with reduced low-density lipoprotein (LDL), cholesterol (LDL-C), or triglyceride levels and protection from coronary heart disease.2222. Cohen JC, Boerwinkle E, Mosley TH, Jr., Hobbs HH. Sequence variations in PCSK9, low LDL, and protection against coronary heart disease. N Engl J Med. 2006;354(12):1264-72.,2323. Laina A, Gatsiou A, Georgiopoulos G, Stamatelopoulos K, Stellos K. RNA therapeutics in cardiovascular precision medicine. Front Physiol. 2018;9:953. Human genetics suggested that lowering circulating levels of these proteins could be used to treat hyperlipidemias and reduce cardiovascular risk. Large clinical trials of monoclonal antibodies against PCSK9 have since validated this therapeutic hypothesis.2424. Sabatine MS, Giugliano RP, Keech AC, Honarpour N, Wiviott SD, Murphy SA, et al. Evolocumab and clinical outcomes in patients with cardiovascular disease. N Engl J Med. 2017;376(18):1713-22.2626. Szarek M, White HD, Schwartz GG, Alings M, Bhatt DL, Bittner VA, et al. Alirocumab reduces total nonfatal cardiovascular and fatal events: the ODYSSEY OUTCOMES trial. J Am Coll Cardiol. 2019;73(4):387-96. Oligonucleotide therapeutics targeting each of the genes above have been developed (Table 1). We discuss below those approved or having phase 3 clinical trial results.

Mipomersen, an ASO that suppresses ApoB synthesis, was the first oligonucleotide therapeutic approved for a cardiovascular indication. The rationale for its development in familial hypercholesterolemia was based in part on the phenotype of individuals with familial hypobetalipoproteinemia. Protein-truncating variants of APOB are associated with very low levels of LDL-C and ApoB,2727. Young SG, Bertics SJ, Curtiss LK, Dubois BW, Witztum JL. Genetic analysis of a kindred with familial hypobetalipoproteinemia. Evidence for two separate gene defects: one associated with an abnormal apolipoprotein B species, apolipoprotein B-37; and a second associated with low plasma concentrations of apolipoprotein B-100. J Clin Invest. 1987;79(6):1842-51. and early clinical observations of humans who carried such APOB mutations suggested a reduced risk of atherosclerotic disease.2828. Glueck CJ, Gartside PS, Mellies MJ, Steiner PM. Familial hypobeta-lipoproteinemia: studies in 13 kindreds. Trans Assoc Am Physicians. 1977;90:184-203. In initial clinical studies, mipomersen given weekly to patients with homozygous familial hypercholesterolemia reduced ApoB by 26.8% and LDL-C by 24.7% (from baseline 441 ± 139 mg/dL).2929. Raal FJ, Santos RD, Blom DJ, Marais AD, Charng MJ, Cromwell WC, et al. Mipomersen, an apolipoprotein B synthesis inhibitor, for lowering of LDL cholesterol concentrations in patients with homozygous familial hypercholesterolaemia: a randomised, double-blind, placebo-controlled trial. Lancet. 2010;375(9719):998-1006. In subsequent phase 3 trials, mipomersen reduced LDL-C by 28-37% in patients with heterozygous familial hypercholesterolemia or severe hypercholesterolemia, on maximally tolerated lipid-lowering therapeutics.3030. McGowan MP, Tardif JC, 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(11):e49006.3232. 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: a randomized, double-blind, placebo-controlled trial. J Am Coll Cardiol. 2013;62(23):2178-84. In all these trials, some patients developed elevations of hepatic transaminases and hepatic steatosis that resolved following discontinuation of mipomersen.3030. McGowan MP, Tardif JC, 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(11):e49006.3232. 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: a randomized, double-blind, placebo-controlled trial. J Am Coll Cardiol. 2013;62(23):2178-84. Familial hypobetalipoproteinemia is likewise associated with hepatic steatosis because ApoB is necessary for export of triglycerides from the liver.3333. Schonfeld G, Patterson BW, Yablonskiy DA, Tanoli TS, Averna M, Elias N, et al. Fatty liver in familial hypobetalipoproteinemia: triglyceride assembly into VLDL particles is affected by the extent of hepatic steatosis. J Lipid Res. 2003;44(3):470-8. Mipomersen was withdrawn from the market.

Volanesorsen, an ASO that suppresses ApoC3 synthesis, is approved in the European Union (EU) for the treatment of familial chylomicronemia syndrome (FCS).3434. European Medicines Agency. Summary of product characteristics: Waylivra 285 mg solution for injection in pre-filled syringe. 2019. [Cited in 2020 September 28]. Available from: https://www.ema.europa.eu/en/medicines/human/EPAR/waylivra#product-information-section.
https://www.ema.europa.eu/en/medicines/h...
Characterized by hypertriglyceridemia and pancreatitis, FCS is caused by a deficiency in the lipoprotein lipase (LPL) enzyme that hydrolyzes triglycerides in chylomicrons and very low-density LDL particles. In contrast to the action of LPL, ApoC3 inhibits the clearance of triglyceride-rich lipoproteins via inhibition of LPL and LPL-independent mechanisms.3535. Gaudet D, Brisson D, Tremblay K, Alexander VJ, Singleton W, Hughes SG, et al. Targeting APOC3 in the familial chylomicronemia syndrome. N Engl J Med. 2014;371(23):2200-6. In a phase 3 study of patients with FCS, volanesorsen decreased ApoC3 levels by 84% from baseline. Notably, total triglyceride fell 77% from mean 2267 ± 1259 mg/dL at baseline to 590 ± 497 mg/dL at 3 months with once-weekly subcutaneous injections.3636. Witztum JL, Gaudet D, Freedman SD, Alexander VJ, Digenio A, Williams KR, et al. Volanesorsen and triglyceride levels in familial chylomicronemia syndrome. N Engl J Med. 2019;381(6):531-42. While the decrease in triglycerides was significant, almost 50% of patients developed a fall in platelet counts below 100 000 per microliter, and 2 patients’ counts fell below 25 000, leading to treatment discontinuation.3636. Witztum JL, Gaudet D, Freedman SD, Alexander VJ, Digenio A, Williams KR, et al. Volanesorsen and triglyceride levels in familial chylomicronemia syndrome. N Engl J Med. 2019;381(6):531-42. In the United States (US), volanesorsen has not been granted Food and Drug Administration approval. A phase 2/3 study of volanesorsen for the treatment of familial partial lipodystrophy and a phase 2 study of a second-generation ApoC3 GalNAc-ASO conjugate for the treatment of hypertriglyceridemia in patients with cardiovascular disease are ongoing (Table 1).

Inclisiran, a GalNAc-siRNA conjugate that suppresses PCSK9 synthesis, significantly reduced LDL-C levels in large, phase 3 trials of patients with familial hypercholesterolemia, atherosclerotic cardiovascular disease (ASCVD), or high risk of ASCVD.2020. Raal FJ, Kallend D, Ray KK, Turner T, Koenig W, Wright RS, et al. Inclisiran for the treatment of heterozygous familial hypercholesterolemia. N Engl J Med. 2020;382(16):1520-30.,2121. Ray KK, Wright RS, Kallend D, Koenig W, Leiter LA, Raal FJ, et al. Two phase 3 trials of inclisiran in patients with elevated LDL cholesterol. N Engl J Med. 2020;382(16):1507-19. LDL receptors on the hepatocyte cell surface remove LDL-C from circulation, following which the receptor is recycled back to the cell surface. Secreted by the liver, PCSK9 regulates the number of LDL receptors by binding them and thus diverting them from recycling to degradation. Gain-of-function variants of PCSK9, hence, lower LDL receptor numbers resulting in higher LDL-C and cardiovascular risk.3737. Abifadel M, Varret M, Rabès JP, Allard D, Ouguerram K, Devillers M, et al. Mutations in PCSK9 cause autosomal dominant hypercholesterolemia. Nat Genet. 2003;34(2):154-6. Loss-of-function variants have the opposite effect.2222. Cohen JC, Boerwinkle E, Mosley TH, Jr., Hobbs HH. Sequence variations in PCSK9, low LDL, and protection against coronary heart disease. N Engl J Med. 2006;354(12):1264-72.

Inclisiran has a sustained duration of action and potency. In ORION-1, a phase 2 study of patients with ASCVD and elevated LDL-C (on maximally tolerated doses of statins), a single 300-mg subcutaneous dose lowered LDL-C by 38.4% from baseline on day 180. A second dose (day 90) resulted in a 52.6% decrease from baseline on day 180.3838. Ray KK, Landmesser U, Leiter LA, Kallend D, Dufour R, Karakas M, et al. Inclisiran in patients at high cardiovascular risk with elevated LDL cholesterol. N Engl J Med. 2017;376(15):1430-40. Data from phase 3 ORION-10 and ORION-11 trials has validated these results.2121. Ray KK, Wright RS, Kallend D, Koenig W, Leiter LA, Raal FJ, et al. Two phase 3 trials of inclisiran in patients with elevated LDL cholesterol. N Engl J Med. 2020;382(16):1507-19. These 2 studies enrolled patients with ASCVD or an ASCVD equivalent (type 2 diabetes, familial hypercholesterolemia, or a 10-year risk of a cardiovascular event of ≥ 20% as assessed by Framingham Risk Score for Cardiovascular Disease or equivalent), who had a baseline LDL-C of ≥ 70 mg/dL (100 mg/dL for ASCVD equivalent) on a maximally tolerated dose of statins.2121. Ray KK, Wright RS, Kallend D, Koenig W, Leiter LA, Raal FJ, et al. Two phase 3 trials of inclisiran in patients with elevated LDL cholesterol. N Engl J Med. 2020;382(16):1507-19. Administered every 6 months, inclisiran reduced LDL-C by 52.3% and 49.9% on day 510 in ORION-10 and ORION-11, respectively.2121. Ray KK, Wright RS, Kallend D, Koenig W, Leiter LA, Raal FJ, et al. Two phase 3 trials of inclisiran in patients with elevated LDL cholesterol. N Engl J Med. 2020;382(16):1507-19. ORION-9 tested the same regimen in patients with heterozygous familial hypercholesterolemia. On day 510, inclisiran reduced LDL-C by 39.7% from a baseline level of 151.4 ± 50.4 mg/dL.2020. Raal FJ, Kallend D, Ray KK, Turner T, Koenig W, Wright RS, et al. Inclisiran for the treatment of heterozygous familial hypercholesterolemia. N Engl J Med. 2020;382(16):1520-30. Adverse events were similar between the inclisiran and placebo groups in ORION-9, -10, and -11. Injection-site reactions were more common in the inclisiran group and were generally mild. ORION-4, an ongoing trial of 15 000 patients, will assess the impact of inclisiran on cardiovascular outcomes.3939. Stoekenbroek RM, Kallend D, Wijngaard PL, Kastelein JJ. Inclisiran for the treatment of cardiovascular disease: the ORION clinical development program. Future Cardiol. 2018;14(6):433-42. Together, the results indicate that, if approved, inclisiran has the potential to be a novel, safe, effective approach to treating hypercholesterolemia in high-risk patients. The pharmacodynamic profile supporting infrequent (biannual) administration additionally suggests this approach could minimize patient noncompliance with treatment and allow hypercholesterolemia to be managed with a dosing strategy more akin to routine vaccinations for diseases such as influenza. Approval of inclisiran is under review in the US and Europe.

Gene silencing in transthyretin-mediated amyloidosis

Transthyretin-mediated (ATTR) amyloidosis is a rare, underdiagnosed, rapidly progressive, debilitating, and fatal disease caused by misfolded transthyretin (TTR) (prealbumin) that accumulates as amyloid deposits in multiple tissues.4040. Hanna M. Novel drugs targeting transthyretin amyloidosis. Curr Heart Fail Rep. 2014;11(1):50-7.,4141. Hawkins PN, Ando Y, Dispenzeri A, Gonzalez-Duarte A, Adams D, Suhr OB. Evolving landscape in the management of transthyretin amyloidosis. Ann Med. 2015;47(8):625-38. Cardiomyopathy and polyneuropathy are the most prominent features, but systemic manifestations arise from amyloid infiltration of other tissues, including the gastrointestinal (GI) tract.4141. Hawkins PN, Ando Y, Dispenzeri A, Gonzalez-Duarte A, Adams D, Suhr OB. Evolving landscape in the management of transthyretin amyloidosis. Ann Med. 2015;47(8):625-38.,4242. Conceição I, Gonzalez-Duarte A, Obici L, Schmidt HH, Simoneau D, Ong ML, et al. "Red-flag" symptom clusters in transthyretin familial amyloid polyneuropathy. J Peripher Nerv Syst. 2016;21(1):5-9. Polyneuropathy includes sensory, motor, and autonomic impairment and has an aggressive course leading to deteriorating quality of life (QOL) and loss of function.4141. Hawkins PN, Ando Y, Dispenzeri A, Gonzalez-Duarte A, Adams D, Suhr OB. Evolving landscape in the management of transthyretin amyloidosis. Ann Med. 2015;47(8):625-38. Cardiac involvement typically presents as heart failure with preserved ejection fraction (HFpEF) or unexplained left ventricular (LV) wall thickening;4343. Maurer MS, Bokhari S, Damy T, Dorbala S, Drachman BM, Fontana M, et al. Expert consensus recommendations for the suspicion and diagnosis of transthyretin cardiac amyloidosis. Circ Heart Fail. 2019;12(9):e006075. progressive symptoms of heart failure (HF) and cardiac arrhythmias are experienced, with death typically occurring 2.5-5.5 years after cardiomyopathy diagnosis.4444. Lane T, Fontana M, Martinez-Naharro A, Quarta CC, Whelan CJ, Petrie A, et al. Natural history, quality of life, and outcome in cardiac transthyretin amyloidosis. Circulation. 2019;140(1):16-26.,4545. Ruberg FL, Maurer MS, Judge DP, Zeldenrust S, Skinner M, Kim AY, et al. Prospective evaluation of the morbidity and mortality of wild-type and V122I mutant transthyretin amyloid cardiomyopathy: the Transthyretin Amyloidosis Cardiac Study (TRACS). Am Heart J. 2012;164(2):222-8 e1. In 2 studies, 13% of patients with HFpEF and 16% of older patients who underwent transcatheter aortic valve replacement for severe aortic stenosis had evidence of cardiac amyloid involvement.4646. Castano A, Narotsky DL, Hamid N, Khalique OK, Morgenstern R, DeLuca A, et al. Unveiling transthyretin cardiac amyloidosis and its predictors among elderly patients with severe aortic stenosis undergoing transcatheter aortic valve replacement. Eur Heart J. 2017;38(38):2879-87.,4747. Gonzalez-Lopez E, Gallego-Delgado M, Guzzo-Merello G, de Haro-Del Moral FJ, Cobo-Marcos M, Robles C, et al. Wild-type transthyretin amyloidosis as a cause of heart failure with preserved ejection fraction. Eur Heart J. 2015;36(38):2585-94. While delays in diagnosis are common,4848. Adams D, Suhr OB, Hund E, Obici L, Tournev I, Campistol JM, et al. First European consensus for diagnosis, management, and treatment of transthyretin familial amyloid polyneuropathy. Curr Opin Neurol. 2016;29(Suppl. 1):S14-26. recognition of the early signs of ATTR amyloidosis (e.g., connective tissue manifestations such as carpal tunnel syndrome)4343. Maurer MS, Bokhari S, Damy T, Dorbala S, Drachman BM, Fontana M, et al. Expert consensus recommendations for the suspicion and diagnosis of transthyretin cardiac amyloidosis. Circ Heart Fail. 2019;12(9):e006075. could be an important opportunity for therapeutic intervention before cardiac (and/or neurologic) symptoms develop.4949. Sperry BW, Reyes BA, Ikram A, Donnelly JP, Phelan D, Jaber WA, et al. Tenosynovial and cardiac amyloidosis in patients undergoing carpal tunnel release. J Am Coll Cardiol. 2018;72(17):2040-50.

Pharmacotherapies that target steps in the amyloidogenic pathway are available. TTR is predominantly produced in the liver and normally circulates as a stable tetramer. However, abnormal destabilization of the protein causes dissociation into monomers that misfold, deposit, and accumulate as amyloid in various tissues, including heart (Figure 3), nerves, and GI tract.5050. Kelly JW. Amyloid fibril formation and protein misassembly: a structural quest for insights into amyloid and prion diseases. Structure. 1997;5(5):595-600. The disease has 2 types, hereditary and wild-type (hATTR and wtATTR amyloidosis, respectively).4040. Hanna M. Novel drugs targeting transthyretin amyloidosis. Curr Heart Fail Rep. 2014;11(1):50-7.,4141. Hawkins PN, Ando Y, Dispenzeri A, Gonzalez-Duarte A, Adams D, Suhr OB. Evolving landscape in the management of transthyretin amyloidosis. Ann Med. 2015;47(8):625-38. In hATTR amyloidosis, also known as ATTRv amyloidosis, TTR gene variants destabilize the protein.5050. Kelly JW. Amyloid fibril formation and protein misassembly: a structural quest for insights into amyloid and prion diseases. Structure. 1997;5(5):595-600. hATTR amyloidosis can have a heterogeneous presentation, differing according to TTR variant, age of onset, and geography.5151. Planté-Bordeneuve V, Kerschen P. Transthyretin familial amyloid polyneuropathy. In: Said G, Krarup C, eds. Peripheral nerve disorders. Vol 115. Amsterdam, The Netherlands: Elsevier B.V.; 2013:643-58. Historically, certain variants have been associated with predominant cardiomyopathy or neuropathy; however, a majority of patients develop a mixed phenotype.5252. Rapezzi C, Quarta CC, Obici L, Perfetto F, Longhi S, Salvi F, et al. Disease profile and differential diagnosis of hereditary transthyretin-related amyloidosis with exclusively cardiac phenotype: an Italian perspective. Eur Heart J. 2013;34(7):520-8.5454. Benson MD, Waddington-Cruz M, Berk JL, Polydefkis M, Dyck PJ, Wang AK, et al. Inotersen treatment for patients with hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):22-31. Several geographies are endemic for hATTR amyloidosis, including Brazil, where the most common variant, V30M, was found in 92% of patients.5555. Cruz MW, Foguel D, Berensztejn AC, Pedrosa RC, Mundayat R, Ong ML, et al. The demographic, genetic, and clinical characteristics of Brazilian subjects enrolled in the Transthyretin Amyloidosis Outcomes Survey. Amyloid. 2017;24(sup1):103-4. This variant has previously been associated with predominant polyneuropathy, although a Brazilian study reported cardiac symptoms in at least 40% of patients with ATTRV30M amyloidosis.5656. Pinto MV, Pinto LF, Dias M, Rosa RS, Mundayat R, Pedrosa RC, et al. Late-onset hereditary ATTR V30M amyloidosis with polyneuropathy: characterization of Brazilian subjects from the THAOS registry. J Neurol Sci. 2019;403:1-6. In contrast, patients with wtATTR amyloidosis do not carry a TTR variant and disease is associated with aging;5757. Rubin J, Maurer MS. Cardiac amyloidosis: overlooked, underappreciated, and treatable. Annu Rev Med. 2020;71:203-19. patients typically present with predominant cardiomyopathy.5858. Maurer MS, Hanna M, Grogan M, Dispenzieri A, Witteles R, Drachman B, et al. Genotype and phenotype of transthyretin cardiac amyloidosis: THAOS (transthyretin amyloid outcome survey). J Am Coll Cardiol. 2016;68(2):161-72.

Figure 3
Cardiac amyloidosis. The amyloidogenic cascade leads to the production of toxic TTR oligomers and amyloid deposits, which impact cardiac structure and function.

Orthotopic liver transplantation (OLT) was the first available treatment for patients with hATTR amyloidosis with polyneuropathy, and it acts by eliminating the production of variant TTR.5959. Ando Y, Coelho T, Berk JL, Cruz MW, Ericzon BG, Ikeda S, et al. Guideline of transthyretin-related hereditary amyloidosis for clinicians. Orphanet J Rare Dis. 2013;8:31. In patients with early-stage polyneuropathy and absence of cardiac involvement, certain symptoms can improve or stabilize in the short term following OLT,5959. Ando Y, Coelho T, Berk JL, Cruz MW, Ericzon BG, Ikeda S, et al. Guideline of transthyretin-related hereditary amyloidosis for clinicians. Orphanet J Rare Dis. 2013;8:31. supporting the therapeutic rationale for silencing the TTR gene in this disease. However, disease progression can occur after OLT; death of 38% of patients due to cardiac events post OLT was reported in a retrospective study.6060. Algalarrondo V, Antonini T, Théaudin M, Chemla D, Benmalek A, Castaing D, et al. Cause of death analysis and temporal trends in survival after liver transplantation for transthyretin familial amyloid polyneuropathy. Amyloid. 2018;25(4):253-60. End-stage HF and sudden death have been observed, due to continued deposition of wild-type (wt) TTR at the site of pre-existing cardiac deposits (e.g., in the myocardium).5959. Ando Y, Coelho T, Berk JL, Cruz MW, Ericzon BG, Ikeda S, et al. Guideline of transthyretin-related hereditary amyloidosis for clinicians. Orphanet J Rare Dis. 2013;8:31.,6161. Yazaki M, Mitsuhashi S, Tokuda T, Kametani F, Takei YI, Koyama J, et al. Progressive wild-type transthyretin deposition after liver transplantation preferentially occurs onto myocardium in FAP patients. Am J Transplant. 2007;7(1):235-42.

Thyroid hormone binding to TTR stabilizes the tetramer,6262. Miroy GJ, Lai Z, Lashuel HA, Peterson SA, Strang C, Kelly JW. Inhibiting transthyretin amyloid fibril formation via protein stabilization. Proc Natl Acad Sci U S A. 1996;93(26):15051-6. motivating the development of small-molecule stabilizers (e.g., diflunisal and tafamidis). Diflunisal, a nonsteroidal anti-inflammatory therapeutic, slows the worsening of neuropathy in patients with hATTR amyloidosis.6363. Berk JL, Suhr OB, Obici L, Sekijima Y, Zeldenrust SR, Yamashita T, et al. Repurposing diflunisal for familial amyloid polyneuropathy: a randomized clinical trial. JAMA. 2013;310(24):2658-67. However, adverse effects on renal and platelet function have limited its use in patients with cardiomyopathy,6464. Sekijima Y, Tojo K, Morita H, Koyama J, Ikeda S. Safety and efficacy of long-term diflunisal administration in hereditary transthyretin (ATTR) amyloidosis. Amyloid. 2015;22(2):79-83. and diflunisal is not approved in any region for the treatment of ATTR amyloidosis. Tafamidis can delay peripheral neurologic impairment6565. Coelho T, Maia LF, Martins da Silva A, Waddington Cruz M, Plante-Bordeneuve V, Lozeron P, et al. Tafamidis for transthyretin familial amyloid polyneuropathy: a randomized, controlled trial. Neurology. 2012;79(8):785-92. and is available for the treatment of early-stage neuropathy in regions including Latin America (e.g., Brazil, Mexico), EU, and Japan.6666. Waddington Cruz M, Benson MD. A review of tafamidis for the treatment of transthyretin-related amyloidosis. Neurol Ther. 2015;4(2):61-79.,6767. Pinto MV, Barreira AA, Bulle AS, Freitas MRG, França MC, Jr., Gondim FAA, et al. Brazilian consensus for diagnosis, management and treatment of transthyretin familial amyloid polyneuropathy. Arq Neuropsiquiatr. 2018;76(9):609-21. Tafamidis has not been approved in the US as a treatment for hATTR amyloidosis with polyneuropathy. Tafamidis has, however, been recently approved for treatment of ATTR amyloidosis with cardiomyopathy following a study of patients with wtATTR and hATTR amyloidosis with New York Heart Association class 1, 2, or 3 HF.6868. Maurer MS, Schwartz JH, Gundapaneni B, Elliott PM, Merlini G, Waddington-Cruz M, et al. Tafamidis treatment for patients with transthyretin amyloid cardiomyopathy. N Engl J Med. 2018;379(11):1007-16. At 30 months, tafamidis was associated with lower all-cause mortality than placebo (hazard ratio [HR], 0.70; 95% confidence interval [CI], 0.51-0.96) and a lower rate of cardiovascular-related hospitalizations (relative risk ratio 0.68 per year; 95% CI, 0.56-0.81). Secondary analyses demonstrated lower rates of decline in 6-minute walk test (6-MWT) distance and in Kansas City Cardiomyopathy Questionnaire overall score compared with placebo.6868. Maurer MS, Schwartz JH, Gundapaneni B, Elliott PM, Merlini G, Waddington-Cruz M, et al. Tafamidis treatment for patients with transthyretin amyloid cardiomyopathy. N Engl J Med. 2018;379(11):1007-16.

Phase 3 studies of 2 oligonucleotide therapeutics, inotersen and patisiran, validated TTR gene silencing in the liver as an effective therapeutic strategy in hATTR amyloidosis with polyneuropathy5353. Adams D, Gonzalez-Duarte A, O'Riordan WD, Yang CC, Ueda M, Kristen AV, et al. Patisiran, an RNAi therapeutic, for hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):11-21.,5454. Benson MD, Waddington-Cruz M, Berk JL, Polydefkis M, Dyck PJ, Wang AK, et al. Inotersen treatment for patients with hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):22-31. and offered insights into the potential of this approach in patients with cardiomyopathy. Indeed, patisiran and 2 other oligonucleotide therapeutics are being evaluated in ongoing trials for ATTR amyloidosis with cardiomyopathy (Table 1).

The efficacy of inotersen and patisiran was evaluated via 2 measures in the pivotal studies of patients with hATTR amyloidosis with polyneuropathy: the modified Neuropathy Impairment Score+7 tests (mNIS+7), a composite measure of neuropathy (higher scores indicate more impairment6969. Suanprasert N, Berk JL, Benson MD, Dyck PJ, Klein CJ, Gollob JA, et al. Retrospective study of a TTR FAP cohort to modify NIS+7 for therapeutic trials. J Neurol Sci. 2014;344(1-2):121-8.); and the Norfolk Quality of Life-Diabetic Neuropathy (Norfolk QOL-DN) questionnaire, which assesses patients’ perception of their QOL (higher scores indicate poorer QOL7070. Vinik EJ, Vinik AI, Paulson JF, Merkies IS, Packman J, Grogan DR, et al. Norfolk QOL-DN: validation of a patient reported outcome measure in transthyretin familial amyloid polyneuropathy. J Peripher Nerv Syst. 2014;19(2):104-14.). Of note, the scoring range of mNIS+7 differed between the trials of inotersen (-22.4-346.3) and patisiran (0-304), reflecting differences in the measurement of sensation, nerve conduction, and autonomic function.5353. Adams D, Gonzalez-Duarte A, O'Riordan WD, Yang CC, Ueda M, Kristen AV, et al. Patisiran, an RNAi therapeutic, for hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):11-21.,5454. Benson MD, Waddington-Cruz M, Berk JL, Polydefkis M, Dyck PJ, Wang AK, et al. Inotersen treatment for patients with hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):22-31.

Inotersen is an ASO administered weekly via subcutaneous injection. In the phase 3 NEURO-TTR trial, inotersen reduced serum TTR protein levels to a median nadir of 79% from baseline.5454. Benson MD, Waddington-Cruz M, Berk JL, Polydefkis M, Dyck PJ, Wang AK, et al. Inotersen treatment for patients with hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):22-31. This resulted in significant improvement compared with placebo in mNIS+7 scores (least-squares mean change from baseline on week 66 of +5.8 points in the inotersen group compared with +25.5 points in the placebo group) and in Norfolk QOL-DN scores (mean change from baseline was +1.0 and +12.7 points for inotersen and placebo groups, respectively).5454. Benson MD, Waddington-Cruz M, Berk JL, Polydefkis M, Dyck PJ, Wang AK, et al. Inotersen treatment for patients with hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):22-31.

Among patients in the NEURO-TTR trial, 63% had echocardiographic signs of cardiac amyloid involvement, but no significant changes were observed in echocardiographic variables at the end of the study.5454. Benson MD, Waddington-Cruz M, Berk JL, Polydefkis M, Dyck PJ, Wang AK, et al. Inotersen treatment for patients with hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):22-31. A small, uncontrolled study of inotersen for patients with hATTR or wtATTR amyloidosis with cardiomyopathy suggested disease improvement relative to baseline after 1-3 years of treatment, with declines in LV mass, LV wall thickness, and N-terminal prohormone of brain-type natriuretic peptide (NT-proBNP), as well as an improvement in global longitudinal strain (GLS).7171. Dasgupta NR, Rissing SM, Smith J, Jung J, Benson MD. Inotersen therapy of transthyretin amyloid cardiomyopathy. Amyloid. 2020;27(1):52-8. Functional capacity appeared to stabilize or improve relative to baseline over the first 2 years of treatment in patients with hATTR amyloidosis, as quantified by the 6-MWT.7171. Dasgupta NR, Rissing SM, Smith J, Jung J, Benson MD. Inotersen therapy of transthyretin amyloid cardiomyopathy. Amyloid. 2020;27(1):52-8. In contrast, data from natural history studies show a progressive decline in the 6-MWT over a similar period.4444. Lane T, Fontana M, Martinez-Naharro A, Quarta CC, Whelan CJ, Petrie A, et al. Natural history, quality of life, and outcome in cardiac transthyretin amyloidosis. Circulation. 2019;140(1):16-26.,4545. Ruberg FL, Maurer MS, Judge DP, Zeldenrust S, Skinner M, Kim AY, et al. Prospective evaluation of the morbidity and mortality of wild-type and V122I mutant transthyretin amyloid cardiomyopathy: the Transthyretin Amyloidosis Cardiac Study (TRACS). Am Heart J. 2012;164(2):222-8 e1.

Inotersen is approved in several countries, including Brazil, for hATTR amyloidosis with polyneuropathy, but treatment requires regular monitoring for thrombocytopenia and acute glomerulonephritis. Each complication occurred in 3% of patients in NEURO-TTR; 1 death was associated with a case of grade 4 thrombocytopenia.5454. Benson MD, Waddington-Cruz M, Berk JL, Polydefkis M, Dyck PJ, Wang AK, et al. Inotersen treatment for patients with hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):22-31. Inotersen is not being developed for cardiomyopathy; however, a GalNAc-ASO conjugate with a similar sequence and design to inotersen is being evaluated in a phase 3 trial of patients with hATTR or wtATTR amyloidosis with cardiomyopathy (Table 1).

Patisiran, an siRNA formulated in an LNP, is administered intravenously every 3 weeks. In the phase 3 APOLLO trial, patisiran reduced TTR protein levels by median 81% from baseline.5353. Adams D, Gonzalez-Duarte A, O'Riordan WD, Yang CC, Ueda M, Kristen AV, et al. Patisiran, an RNAi therapeutic, for hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):11-21. Patisiran resulted in improvements in mNIS+7 and Norfolk QOL-DN scores relative to both placebo and to the patients’ own baseline.5353. Adams D, Gonzalez-Duarte A, O'Riordan WD, Yang CC, Ueda M, Kristen AV, et al. Patisiran, an RNAi therapeutic, for hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):11-21. In the patisiran and placebo arms, the least-squares mean changes from baseline were -6.0 and +28.0 in mNIS+7 and -6.7 and +14.4 in Norfolk QOL-DN, respectively. Patisiran also improved gait speed in a 10-meter walk test (change relative to baseline was +0.08 and -0.24 meters per second in the patisiran and placebo arms, respectively).5353. Adams D, Gonzalez-Duarte A, O'Riordan WD, Yang CC, Ueda M, Kristen AV, et al. Patisiran, an RNAi therapeutic, for hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):11-21.

Data from APOLLO suggest that patisiran may also improve measures of cardiac structure and function in patients with cardiomyopathy. Prespecified exploratory analyses assessed patisiran and placebo in a subpopulation with evidence of cardiac amyloid involvement, defined by a baseline LV wall thickness ≥ 13 mm and no history of aortic valve disease or hypertension.7272. Solomon SD, Adams D, Kristen A, Grogan M, Gonzalez-Duarte A, Maurer MS, et al. Effects of patisiran, an RNA interference therapeutic, on cardiac parameters in patients with hereditary transthyretin-mediated amyloidosis. Circulation. 2019;139(4):431-43. Reflecting the multisystem nature of hATTR amyloidosis, the cardiac subpopulation comprised 56% of APOLLO patients who were enrolled on the basis of polyneuropathy. Compared with placebo, patisiran reduced mean LV wall thickness, increased end-diastolic volume, decreased GLS, increased cardiac output, and lowered NT-proBNP. A decrease in GLS in the patisiran arm is notable since an absolute 1% increase has previously been associated with an increased risk of death (HR, 1.1, 95% CI, 1.01-1.19).7373. Quarta CC, Solomon SD, Uraizee I, Kruger J, Longhi S, Ferlito M, et al. Left ventricular structure and function in transthyretin-related versus light-chain cardiac amyloidosis. Circulation. 2014;129(18):1840-9. Patisiran demonstrated a positive benefit:risk profile in both the overall and cardiac subpopulations. In post hoc analyses of safety data, among the entire APOLLO population, the exposure-adjusted rates of cardiac hospitalizations and all-cause death were 18.7 and 10.1 per 100 patient-years in the placebo and patisiran groups, respectively (Andersen–Gill HR, 0.54; 95% CI, 0.28-1.01).7272. Solomon SD, Adams D, Kristen A, Grogan M, Gonzalez-Duarte A, Maurer MS, et al. Effects of patisiran, an RNA interference therapeutic, on cardiac parameters in patients with hereditary transthyretin-mediated amyloidosis. Circulation. 2019;139(4):431-43. While these data suggest that improvement in cardiac parameters following patisiran treatment may lead to improved outcomes, the APOLLO study was not designed to investigate clinical outcomes such as death or cardiovascular hospitalization, and further studies are needed.

Patisiran is approved in > 30 countries globally, including Brazil, for the treatment of hATTR amyloidosis with polyneuropathy (specific indications vary by country/region).7474. Kristen AV, Ajroud-Driss S, Conceicao I, Gorevic P, Kyriakides T, Obici L. Patisiran, an RNAi therapeutic for the treatment of hereditary transthyretin-mediated amyloidosis. Neurodegener Dis Manag. 2019;9(1):5-23.7878. Alnylam Pharmaceuticals Inc. Alnylam announces approval in Japan of ONPATTRO® for the treatment of hereditary ATTR amyloidosis with polyneuropathy [press release]. 2019. [Cited in 2020 September 28]. Available from: https://investors.alnylam.com/press-release?id=23886.
https://investors.alnylam.com/press-rele...
Except for infusion-related reactions, the overall incidence and types of adverse effects were similar between patisiran and placebo arms of the APOLLO trial. Infusion-related reactions occurred in 19% and 9% of patisiran- and placebo-treated patients, respectively.5353. Adams D, Gonzalez-Duarte A, O'Riordan WD, Yang CC, Ueda M, Kristen AV, et al. Patisiran, an RNAi therapeutic, for hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):11-21. They were generally mild or moderate, spontaneously resolved, and decreased in frequency with subsequent infusions; risk was reduced by premedicating patisiran-treated patients. An ongoing trial, APOLLO-B, is evaluating the safety and efficacy of patisiran in ATTR amyloidosis with cardiomyopathy (Table 1).

Vutrisiran, a second-generation investigational RNAi therapeutic targeting TTR, uses a GalNAc-siRNA ESC conjugate, similar to that used for inclisiran, with chemical modifications that confer increased potency and high metabolic stability. Administered subcutaneously once every 3 months in a phase 1 study of healthy volunteers, a single 25-mg dose resulted in a maximum 80% TTR reduction sustained for approximately 90 days.7979. Habtemariam BA, Karsten V, Attarwala H, Goel V, Melch M, Clausen VA, et al. Single dose pharmacokinetics and pharmacodynamics of transthyretin targeting GalNAc-siRNA conjugate, vutrisiran, in healthy subjects. Clin Pharmacol Ther. 2020;Jun 29. doi: 10.1002/cpt.1974. Online ahead of print.
https://doi.org/10.1002/cpt.1974...
This vutrisiran regimen is predicted to achieve 88-90% TTR reduction, similar to patisiran in the APOLLO trial. Two phase 3 trials are currently ongoing to evaluate vutrisiran for the treatment of hATTR amyloidosis with polyneuropathy (HELIOS-A) and ATTR amyloidosis with cardiomyopathy (HELIOS-B; Table 1).

Summary

More than 40 years after Zamecnik envisioned targeting RNA as a therapeutic modality, oligonucleotides have emerged as cutting-edge medicines that exploit endogenous mRNA degradation mechanisms. By silencing the expression of disease-causing proteins with genetic precision, siRNAs and ASOs expand the range of biologic targets beyond those that require direct drug-protein binding. Advances in nucleic acid chemistry and oligonucleotide delivery to the liver have yielded novel therapeutic approaches to cardiovascular diseases. They include common conditions like hypercholesterolemia and rarer ones like ATTR amyloidosis, which may be more prevalent than previously thought. GalNAc-siRNA conjugates have demonstrated positive benefit:risk profiles in clinical studies and have potential to be convenient treatments for conditions that are difficult to manage. Their characteristics also raise the possibility that these therapeutics may be useful for both symptomatic and presymptomatic individuals. For example, early diagnosis and quarterly treatment with an siRNA in patients with noncardiac signs of ATTR amyloidosis may prevent the development of congestive HF. Also, semiannual injections of inclisiran for high-risk individuals with hypercholesterolemia may achieve better outcomes than current medications whose compliance is inconsistent. While oligonucleotide-based therapeutics are relatively new, their ability to target the fundamental bases of diseases is already clear, and their impact on medicine is certain to grow.

  • Sources of Funding
    This study was funded by Alnylam Pharmaceuticals.
  • Study Association
    This study is not associated with any thesis or dissertation work.
  • Ethics approval and consent to participate
    This article does not contain any studies with human participants or animals performed by any of the authors.

References

  • 1
    Stephenson ML, Zamecnik PC. Inhibition of Rous sarcoma viral RNA translation by a specific oligodeoxyribonucleotide. Proc Natl Acad Sci U S A. 1978;75(1):285-8.
  • 2
    Zamecnik PC, Stephenson ML. Inhibition of Rous sarcoma virus replication and cell transformation by a specific oligodeoxynucleotide. Proc Natl Acad Sci U S A. 1978;75(1):280-4.
  • 3
    Broecker F, Moelling K. Evolution of immune systems from viruses and transposable elements. Front Microbiol. 2019;10:51.
  • 4
    Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K, Tuschl T. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature. 2001;411(6836):494-8.
  • 5
    Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature. 1998;391(6669):806-11.
  • 6
    Cook PD. Antisense medicinal chemistry. In: Crooke ST, ed. Antisense research and application. Vol 131. Switzerland: Springer Nature; 1998:51-101.
  • 7
    Chi X, Gatti P, Papoian T. Safety of antisense oligonucleotide and siRNA-based therapeutics. Drug Discov Today. 2017;22(5):823-33.
  • 8
    Nykänen A, Haley B, Zamore PD. ATP requirements and small interfering RNA structure in the RNA interference pathway. Cell. 2001;107(3):309-21.
  • 9
    Dowdy SF. Overcoming cellular barriers for RNA therapeutics. Nat Biotechnol. 2017;35(3):222-9.
  • 10
    Foster DJ, Brown CR, Shaikh S, Trapp C, Schlegel MK, Qian K, et al. Advanced siRNA designs further improve in vivo performance of GalNAc-siRNA conjugates. Mol Ther. 2018;26(3):708-17.
  • 11
    Nair JK, Willoughby JL, Chan A, Charisse K, Alam MR, Wang Q, et al. Multivalent N-acetylgalactosamine-conjugated siRNA localizes in hepatocytes and elicits robust RNAi-mediated gene silencing. J Am Chem Soc. 2014;136(49):16958-61.
  • 12
    Zimmermann TS, Lee AC, Akinc A, Bramlage B, Bumcrot D, Fedoruk MN, et al. RNAi-mediated gene silencing in non-human primates. Nature. 2006;441(7089):111-4.
  • 13
    Jayaraman M, Ansell SM, Mui BL, Tam YK, Chen J, Du X, et al. Maximizing the potency of siRNA lipid nanoparticles for hepatic gene silencing in vivo. Angew Chem Int Ed Engl. 2012;51(34):8529-33.
  • 14
    Akinc A, Maier MA, Manoharan M, Fitzgerald K, Jayaraman M, Barros S, et al. The Onpattro story and the clinical translation of nanomedicines containing nucleic acid-based drugs. Nat Nanotechnol. 2019;14(12):1084-7.
  • 15
    Coelho T, Adams D, Silva A, Lozeron P, Hawkins PN, Mant T, et al. Safety and efficacy of RNAi therapy for transthyretin amyloidosis. N Engl J Med. 2013;369(9):819-29.
  • 16
    Ashwell G, Morell AG. The role of surface carbohydrates in the hepatic recognition and transport of circulating glycoproteins. Adv Enzymol Relat Areas Mol Biol. 1974;41(0):99-128.
  • 17
    Biessen EA, Beuting DM, Roelen HC, van de Marel GA, van Boom JH, van Berkel TJ. Synthesis of cluster galactosides with high affinity for the hepatic asialoglycoprotein receptor. J Med Chem. 1995;38(9):1538-46.
  • 18
    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(13):8796-807.
  • 19
    Balwani M, Sardh E, Ventura P, Peiró PA, Rees DC, Stölzel U, et al. Phase 3 trial of RNAi therapeutic givosiran for acute intermittent porphyria. N Engl J Med. 2020;382(24):2289-301.
  • 20
    Raal FJ, Kallend D, Ray KK, Turner T, Koenig W, Wright RS, et al. Inclisiran for the treatment of heterozygous familial hypercholesterolemia. N Engl J Med. 2020;382(16):1520-30.
  • 21
    Ray KK, Wright RS, Kallend D, Koenig W, Leiter LA, Raal FJ, et al. Two phase 3 trials of inclisiran in patients with elevated LDL cholesterol. N Engl J Med. 2020;382(16):1507-19.
  • 22
    Cohen JC, Boerwinkle E, Mosley TH, Jr., Hobbs HH. Sequence variations in PCSK9, low LDL, and protection against coronary heart disease. N Engl J Med. 2006;354(12):1264-72.
  • 23
    Laina A, Gatsiou A, Georgiopoulos G, Stamatelopoulos K, Stellos K. RNA therapeutics in cardiovascular precision medicine. Front Physiol. 2018;9:953.
  • 24
    Sabatine MS, Giugliano RP, Keech AC, Honarpour N, Wiviott SD, Murphy SA, et al. Evolocumab and clinical outcomes in patients with cardiovascular disease. N Engl J Med. 2017;376(18):1713-22.
  • 25
    Schwartz GG, Steg PG, Szarek M, Bhatt DL, Bittner VA, Diaz R, et al. Alirocumab and cardiovascular outcomes after acute coronary syndrome. N Engl J Med. 2018;379(22):2097-107.
  • 26
    Szarek M, White HD, Schwartz GG, Alings M, Bhatt DL, Bittner VA, et al. Alirocumab reduces total nonfatal cardiovascular and fatal events: the ODYSSEY OUTCOMES trial. J Am Coll Cardiol. 2019;73(4):387-96.
  • 27
    Young SG, Bertics SJ, Curtiss LK, Dubois BW, Witztum JL. Genetic analysis of a kindred with familial hypobetalipoproteinemia. Evidence for two separate gene defects: one associated with an abnormal apolipoprotein B species, apolipoprotein B-37; and a second associated with low plasma concentrations of apolipoprotein B-100. J Clin Invest. 1987;79(6):1842-51.
  • 28
    Glueck CJ, Gartside PS, Mellies MJ, Steiner PM. Familial hypobeta-lipoproteinemia: studies in 13 kindreds. Trans Assoc Am Physicians. 1977;90:184-203.
  • 29
    Raal FJ, Santos RD, Blom DJ, Marais AD, Charng MJ, Cromwell WC, et al. Mipomersen, an apolipoprotein B synthesis inhibitor, for lowering of LDL cholesterol concentrations in patients with homozygous familial hypercholesterolaemia: a randomised, double-blind, placebo-controlled trial. Lancet. 2010;375(9719):998-1006.
  • 30
    McGowan MP, Tardif JC, 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(11):e49006.
  • 31
    Stein EA, Dufour R, Gagne C, Gaudet D, East C, Donovan JM, et al. Apolipoprotein B synthesis inhibition with mipomersen in heterozygous familial hypercholesterolemia: results of a randomized, double-blind, placebo-controlled trial to assess efficacy and safety as add-on therapy in patients with coronary artery disease. Circulation. 2012;126(19):2283-92.
  • 32
    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: a randomized, double-blind, placebo-controlled trial. J Am Coll Cardiol. 2013;62(23):2178-84.
  • 33
    Schonfeld G, Patterson BW, Yablonskiy DA, Tanoli TS, Averna M, Elias N, et al. Fatty liver in familial hypobetalipoproteinemia: triglyceride assembly into VLDL particles is affected by the extent of hepatic steatosis. J Lipid Res. 2003;44(3):470-8.
  • 34
    European Medicines Agency. Summary of product characteristics: Waylivra 285 mg solution for injection in pre-filled syringe. 2019. [Cited in 2020 September 28]. Available from: https://www.ema.europa.eu/en/medicines/human/EPAR/waylivra#product-information-section
    » https://www.ema.europa.eu/en/medicines/human/EPAR/waylivra#product-information-section
  • 35
    Gaudet D, Brisson D, Tremblay K, Alexander VJ, Singleton W, Hughes SG, et al. Targeting APOC3 in the familial chylomicronemia syndrome. N Engl J Med. 2014;371(23):2200-6.
  • 36
    Witztum JL, Gaudet D, Freedman SD, Alexander VJ, Digenio A, Williams KR, et al. Volanesorsen and triglyceride levels in familial chylomicronemia syndrome. N Engl J Med. 2019;381(6):531-42.
  • 37
    Abifadel M, Varret M, Rabès JP, Allard D, Ouguerram K, Devillers M, et al. Mutations in PCSK9 cause autosomal dominant hypercholesterolemia. Nat Genet. 2003;34(2):154-6.
  • 38
    Ray KK, Landmesser U, Leiter LA, Kallend D, Dufour R, Karakas M, et al. Inclisiran in patients at high cardiovascular risk with elevated LDL cholesterol. N Engl J Med. 2017;376(15):1430-40.
  • 39
    Stoekenbroek RM, Kallend D, Wijngaard PL, Kastelein JJ. Inclisiran for the treatment of cardiovascular disease: the ORION clinical development program. Future Cardiol. 2018;14(6):433-42.
  • 40
    Hanna M. Novel drugs targeting transthyretin amyloidosis. Curr Heart Fail Rep. 2014;11(1):50-7.
  • 41
    Hawkins PN, Ando Y, Dispenzeri A, Gonzalez-Duarte A, Adams D, Suhr OB. Evolving landscape in the management of transthyretin amyloidosis. Ann Med. 2015;47(8):625-38.
  • 42
    Conceição I, Gonzalez-Duarte A, Obici L, Schmidt HH, Simoneau D, Ong ML, et al. "Red-flag" symptom clusters in transthyretin familial amyloid polyneuropathy. J Peripher Nerv Syst. 2016;21(1):5-9.
  • 43
    Maurer MS, Bokhari S, Damy T, Dorbala S, Drachman BM, Fontana M, et al. Expert consensus recommendations for the suspicion and diagnosis of transthyretin cardiac amyloidosis. Circ Heart Fail. 2019;12(9):e006075.
  • 44
    Lane T, Fontana M, Martinez-Naharro A, Quarta CC, Whelan CJ, Petrie A, et al. Natural history, quality of life, and outcome in cardiac transthyretin amyloidosis. Circulation. 2019;140(1):16-26.
  • 45
    Ruberg FL, Maurer MS, Judge DP, Zeldenrust S, Skinner M, Kim AY, et al. Prospective evaluation of the morbidity and mortality of wild-type and V122I mutant transthyretin amyloid cardiomyopathy: the Transthyretin Amyloidosis Cardiac Study (TRACS). Am Heart J. 2012;164(2):222-8 e1.
  • 46
    Castano A, Narotsky DL, Hamid N, Khalique OK, Morgenstern R, DeLuca A, et al. Unveiling transthyretin cardiac amyloidosis and its predictors among elderly patients with severe aortic stenosis undergoing transcatheter aortic valve replacement. Eur Heart J. 2017;38(38):2879-87.
  • 47
    Gonzalez-Lopez E, Gallego-Delgado M, Guzzo-Merello G, de Haro-Del Moral FJ, Cobo-Marcos M, Robles C, et al. Wild-type transthyretin amyloidosis as a cause of heart failure with preserved ejection fraction. Eur Heart J. 2015;36(38):2585-94.
  • 48
    Adams D, Suhr OB, Hund E, Obici L, Tournev I, Campistol JM, et al. First European consensus for diagnosis, management, and treatment of transthyretin familial amyloid polyneuropathy. Curr Opin Neurol. 2016;29(Suppl. 1):S14-26.
  • 49
    Sperry BW, Reyes BA, Ikram A, Donnelly JP, Phelan D, Jaber WA, et al. Tenosynovial and cardiac amyloidosis in patients undergoing carpal tunnel release. J Am Coll Cardiol. 2018;72(17):2040-50.
  • 50
    Kelly JW. Amyloid fibril formation and protein misassembly: a structural quest for insights into amyloid and prion diseases. Structure. 1997;5(5):595-600.
  • 51
    Planté-Bordeneuve V, Kerschen P. Transthyretin familial amyloid polyneuropathy. In: Said G, Krarup C, eds. Peripheral nerve disorders. Vol 115. Amsterdam, The Netherlands: Elsevier B.V.; 2013:643-58.
  • 52
    Rapezzi C, Quarta CC, Obici L, Perfetto F, Longhi S, Salvi F, et al. Disease profile and differential diagnosis of hereditary transthyretin-related amyloidosis with exclusively cardiac phenotype: an Italian perspective. Eur Heart J. 2013;34(7):520-8.
  • 53
    Adams D, Gonzalez-Duarte A, O'Riordan WD, Yang CC, Ueda M, Kristen AV, et al. Patisiran, an RNAi therapeutic, for hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):11-21.
  • 54
    Benson MD, Waddington-Cruz M, Berk JL, Polydefkis M, Dyck PJ, Wang AK, et al. Inotersen treatment for patients with hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):22-31.
  • 55
    Cruz MW, Foguel D, Berensztejn AC, Pedrosa RC, Mundayat R, Ong ML, et al. The demographic, genetic, and clinical characteristics of Brazilian subjects enrolled in the Transthyretin Amyloidosis Outcomes Survey. Amyloid. 2017;24(sup1):103-4.
  • 56
    Pinto MV, Pinto LF, Dias M, Rosa RS, Mundayat R, Pedrosa RC, et al. Late-onset hereditary ATTR V30M amyloidosis with polyneuropathy: characterization of Brazilian subjects from the THAOS registry. J Neurol Sci. 2019;403:1-6.
  • 57
    Rubin J, Maurer MS. Cardiac amyloidosis: overlooked, underappreciated, and treatable. Annu Rev Med. 2020;71:203-19.
  • 58
    Maurer MS, Hanna M, Grogan M, Dispenzieri A, Witteles R, Drachman B, et al. Genotype and phenotype of transthyretin cardiac amyloidosis: THAOS (transthyretin amyloid outcome survey). J Am Coll Cardiol. 2016;68(2):161-72.
  • 59
    Ando Y, Coelho T, Berk JL, Cruz MW, Ericzon BG, Ikeda S, et al. Guideline of transthyretin-related hereditary amyloidosis for clinicians. Orphanet J Rare Dis. 2013;8:31.
  • 60
    Algalarrondo V, Antonini T, Théaudin M, Chemla D, Benmalek A, Castaing D, et al. Cause of death analysis and temporal trends in survival after liver transplantation for transthyretin familial amyloid polyneuropathy. Amyloid. 2018;25(4):253-60.
  • 61
    Yazaki M, Mitsuhashi S, Tokuda T, Kametani F, Takei YI, Koyama J, et al. Progressive wild-type transthyretin deposition after liver transplantation preferentially occurs onto myocardium in FAP patients. Am J Transplant. 2007;7(1):235-42.
  • 62
    Miroy GJ, Lai Z, Lashuel HA, Peterson SA, Strang C, Kelly JW. Inhibiting transthyretin amyloid fibril formation via protein stabilization. Proc Natl Acad Sci U S A. 1996;93(26):15051-6.
  • 63
    Berk JL, Suhr OB, Obici L, Sekijima Y, Zeldenrust SR, Yamashita T, et al. Repurposing diflunisal for familial amyloid polyneuropathy: a randomized clinical trial. JAMA. 2013;310(24):2658-67.
  • 64
    Sekijima Y, Tojo K, Morita H, Koyama J, Ikeda S. Safety and efficacy of long-term diflunisal administration in hereditary transthyretin (ATTR) amyloidosis. Amyloid. 2015;22(2):79-83.
  • 65
    Coelho T, Maia LF, Martins da Silva A, Waddington Cruz M, Plante-Bordeneuve V, Lozeron P, et al. Tafamidis for transthyretin familial amyloid polyneuropathy: a randomized, controlled trial. Neurology. 2012;79(8):785-92.
  • 66
    Waddington Cruz M, Benson MD. A review of tafamidis for the treatment of transthyretin-related amyloidosis. Neurol Ther. 2015;4(2):61-79.
  • 67
    Pinto MV, Barreira AA, Bulle AS, Freitas MRG, França MC, Jr., Gondim FAA, et al. Brazilian consensus for diagnosis, management and treatment of transthyretin familial amyloid polyneuropathy. Arq Neuropsiquiatr. 2018;76(9):609-21.
  • 68
    Maurer MS, Schwartz JH, Gundapaneni B, Elliott PM, Merlini G, Waddington-Cruz M, et al. Tafamidis treatment for patients with transthyretin amyloid cardiomyopathy. N Engl J Med. 2018;379(11):1007-16.
  • 69
    Suanprasert N, Berk JL, Benson MD, Dyck PJ, Klein CJ, Gollob JA, et al. Retrospective study of a TTR FAP cohort to modify NIS+7 for therapeutic trials. J Neurol Sci. 2014;344(1-2):121-8.
  • 70
    Vinik EJ, Vinik AI, Paulson JF, Merkies IS, Packman J, Grogan DR, et al. Norfolk QOL-DN: validation of a patient reported outcome measure in transthyretin familial amyloid polyneuropathy. J Peripher Nerv Syst. 2014;19(2):104-14.
  • 71
    Dasgupta NR, Rissing SM, Smith J, Jung J, Benson MD. Inotersen therapy of transthyretin amyloid cardiomyopathy. Amyloid. 2020;27(1):52-8.
  • 72
    Solomon SD, Adams D, Kristen A, Grogan M, Gonzalez-Duarte A, Maurer MS, et al. Effects of patisiran, an RNA interference therapeutic, on cardiac parameters in patients with hereditary transthyretin-mediated amyloidosis. Circulation. 2019;139(4):431-43.
  • 73
    Quarta CC, Solomon SD, Uraizee I, Kruger J, Longhi S, Ferlito M, et al. Left ventricular structure and function in transthyretin-related versus light-chain cardiac amyloidosis. Circulation. 2014;129(18):1840-9.
  • 74
    Kristen AV, Ajroud-Driss S, Conceicao I, Gorevic P, Kyriakides T, Obici L. Patisiran, an RNAi therapeutic for the treatment of hereditary transthyretin-mediated amyloidosis. Neurodegener Dis Manag. 2019;9(1):5-23.
  • 75
    Alnylam Pharmaceuticals Inc. Alnylam announces approval in Brazil of ONPATTRO® for the treatment of hereditary ATTR amyloidosis with polyneuropathy [press release]. 2020. [Cited in 2020 September 28]. Available from: https://investors.alnylam.com/press-release?id=24606
    » https://investors.alnylam.com/press-release?id=24606
  • 76
    CADTH. Patisiran. 2019. [Cited in 2020 September 28]. Available from: https://www.cadth.ca/patisiran
    » https://www.cadth.ca/patisiran
  • 77
    swissmedic. Abbreviated information for health care professionals for ONPATTRO 10 mg/5 ml, concentrate for solution for infusion (version September 2019). 2019. [Cited in 2020 September 28]. Available from: www.swissmedicinfo.ch
    » www.swissmedicinfo.ch
  • 78
    Alnylam Pharmaceuticals Inc. Alnylam announces approval in Japan of ONPATTRO® for the treatment of hereditary ATTR amyloidosis with polyneuropathy [press release]. 2019. [Cited in 2020 September 28]. Available from: https://investors.alnylam.com/press-release?id=23886
    » https://investors.alnylam.com/press-release?id=23886
  • 79
    Habtemariam BA, Karsten V, Attarwala H, Goel V, Melch M, Clausen VA, et al. Single dose pharmacokinetics and pharmacodynamics of transthyretin targeting GalNAc-siRNA conjugate, vutrisiran, in healthy subjects. Clin Pharmacol Ther. 2020;Jun 29. doi: 10.1002/cpt.1974. Online ahead of print.
    » https://doi.org/10.1002/cpt.1974

Publication Dates

  • Publication in this collection
    26 July 2021
  • Date of issue
    Sep-Oct 2022

History

  • Received
    29 Sept 2020
  • Reviewed
    17 Feb 2021
  • Accepted
    17 Feb 2021
Sociedade Brasileira de Cardiologia Avenida Marechal Câmara, 160, sala: 330, Centro, CEP: 20020-907, (21) 3478-2700 - Rio de Janeiro - RJ - Brazil
E-mail: revistaijcs@cardiol.br