Functional Analysis of LDLR (Low-Density Lipoprotein Receptor) Variants in Patient Lymphocytes to Assess the Effect of Evinacumab in Homozygous Familial Hypercholesterolemia Patients With a Spectrum of LDLR Activity

Poulabi Banerjee, Kuo-Chen Chan, Michel Tarabocchia, Asier Benito-Vicente, Ana C Alves, Kepa B Uribe, Mafalda Bourbon, Paul J Skiba, Robert Pordy, Daniel A Gipe, Daniel Gaudet, Cesar Martin, Poulabi Banerjee, Kuo-Chen Chan, Michel Tarabocchia, Asier Benito-Vicente, Ana C Alves, Kepa B Uribe, Mafalda Bourbon, Paul J Skiba, Robert Pordy, Daniel A Gipe, Daniel Gaudet, Cesar Martin

Abstract

Objective: Homozygous familial hypercholesterolemia is a rare disease usually caused by LDLR (low-density lipoprotein receptor) mutations. Homozygous familial hypercholesterolemia is characterized by markedly elevated LDL-C (low-density lipoprotein cholesterol) levels and an extremely high risk of premature atherosclerotic cardiovascular disease. A phase 2, proof-of-concept study (NCT02265952) demonstrated that evinacumab, a fully human monoclonal antibody to ANGPTL3 (angiopoietin-like 3 protein), reduced LDL-C levels in 9 patients with genotypically confirmed homozygous familial hypercholesterolemia and was well tolerated. The aim of this study was to analyze the effects of evinacumab on LDLR activity in lymphocytes purified from patients in the proof-of-concept study. Approach and Results: LDLR activity was assessed in patient lymphocytes before and after treatment with evinacumab and versus lymphocytes carrying wild-type LDLR, and also in an LDLR-defective Chinese hamster ovary cell line (CHO-ldlA7) transfected with plasmids encoding the LDLR variants. Overall mean peak reduction in LDL-C with evinacumab was -58±18%, occurring between Week 4 and Week 12. Mutations identified in the 9 patients were shown to be pathogenic, with loss of LDLR activity versus wild type. Two of the LDLR variants, p.(Cys681*) and p.(Ala627Profs*38), were class 2 type mutations that are retained in the endoplasmic reticulum. Six variants were class 3 type mutations with impaired LDL-C binding activity: p.(Trp87Gly), occurring in 2 patients, p.(Gln254Pro), p.(Ser177Leu), p.(Gly335Val), and p.(Ser306Leu). Evinacumab had no effect on LDLR activity.

Conclusions: These results suggest that evinacumab is effective for lowering LDL-C in patients with homozygous familial hypercholesterolemia, and the inhibition of ANGPTL3 in humans lowers LDL-C in a mechanism independent of the LDLR.

Keywords: hypercholesterolemia; lipoproteins; mutations; proof of concept study; rare disease.

Figures

Figure 1.
Figure 1.
LDLR (low-density lipoprotein receptor) activity in lymphocytes from 9 homozygous familial hypercholesterolemia (HoFH) patients before and after evinacumab treatment. LDLR expression (A); LDL binding (B); and LDL uptake (C). LDLR activity and expression was quantified by flow cytometry as described in the Materials and Methods section. The values represent the mean of 3 independent experiments; error bars represent ±SD. wt=wild-type (non-HoFH) control; c.[191-?_694+?del] and p.(Trp87*)=LDLR defective heterozygous patient controls (see Methods). (1;2) represents the mean±SD of patients 1 and 2 which carry the same LDLR variants; the lymphocytes from these 2 patients were analyzed independently. The sex of the patients 1 to 9 from which the lymphocytes were sourced is indicated in the Table. HoFH indicates homozygous familial hypercholesterolemia; LDL, low-density lipoprotein; and LDLR, low-density lipoprotein receptor. *P<0.001 comparing wt with each variant before and after treatment. No statistical significance was found when after treatment vs before treatment LDLR activity data (expression, binding, and uptake) of each variant was compared.
Figure 2.
Figure 2.
Expression of wild-type LDLR and LDLR variants in Chinese hamster ovary-ldlA7 transfected cells. LDLR (low-density lipoprotein receptor) expression (upper) and GAPDH expression (A and C; lower); LDLR to GAPDH densitometric analysis (B and D). Cells were transfected with the corresponding plasmids carrying the LDLR mutations of interest and immunoblot analysis was performed, as described in Materials and Methods. The relative band intensity of mature LDLR protein expression was calculated as the ratio of 160 kDa LDLR band intensity to that of GAPDH. A representative experiment from 3 independently performed assays is shown in A. Levels of significance were determined by a 2-tailed Student t test, and a CI of >95% (P<0.05) was used to establish statistical significance. No statistically significant differences were found among LDLR expression for variants analyzed in A and B, but differences were significant for variants analyzed in C and D. wt indicates wild-type.
Figure 3.
Figure 3.
LDLR (low-density lipoprotein receptor) activity in Chinese hamster ovary-ldlA7-transfected cells. LDLR expression (A); LDL (low-density lipoprotein) binding (B); and LDL uptake (C). LDLR activity and expression was quantified by flow cytometry, as described in the Materials and Methods section. The values represent the mean of 3 independent experiments; error bars represent ±SD. wt indicates wild-type. *P<0.001 comparing wt with each variant.
Figure 4.
Figure 4.
Class 2 type mutation assignment of variants found in homozygous familial hypercholesterolemia patients. LDLR (low-density lipoprotein receptor) expression and colocalization with calregulin in the endoplasmic reticulum (A); LDL (low-density lipoprotein)-LDLR binding activity (B); and LDL internalization activity (C). Confocal laser scanning microscopy was used to analyze LDLR activity, as described in the Materials and Methods section. wt indicates wild-type.
Figure 5.
Figure 5.
Class 3 type mutation assignment of variants found in homozygous familial hypercholesterolemia patients. LDLR (low-density lipoprotein receptor) expression at cellular membrane (A); LDL (low-density lipoprotein)-LDLR binding activity (B); and LDL internalization activity (C). Confocal laser scanning microscopy was used to analyze LDLR activity, as described in the Materials and Methods section. wt indicates wild-type.

References

    1. Cuchel M, Bruckert E, Ginsberg HN, Raal FJ, Santos RD, Hegele RA, Kuivenhoven JA, Nordestgaard BG, Descamps OS, Steinhagen-Thiessen E, et al. European Atherosclerosis Society Consensus Panel on Familial Hypercholesterolaemia. Homozygous familial hypercholesterolaemia: new insights and guidance for clinicians to improve detection and clinical management. A position paper from the Consensus Panel on Familial Hypercholesterolaemia of the European Atherosclerosis Society. Eur Heart J. 2014;35:2146–2157. doi: 10.1093/eurheartj/ehu274.
    1. Marais AD. Familial hypercholesterolaemia. Clin Biochem Rev. 2004;25:49–68.
    1. Goldstein JL, Hobbs HH, Brown MS. Familial hypercholesterolemia. In: Scriver CR, Beaudet A, Sly WS, Valle D, editors. In: The Metabolic and Molecular Bases of Inherited Disease. New York: McGraw-Hill; 2001. pp. 2863–2913.
    1. Hobbs HH, Brown MS, Goldstein JL. Molecular genetics of the LDL receptor gene in familial hypercholesterolemia. Hum Mutat. 1992;1:445–466. doi: 10.1002/humu.1380010602.
    1. Nordestgaard BG, Chapman MJ, Humphries SE, Ginsberg HN, Masana L, Descamps OS, Wiklund O, Hegele RA, Raal FJ, Defesche JC, et al. European Atherosclerosis Society Consensus Panel. Familial hypercholesterolaemia is underdiagnosed and undertreated in the general population: guidance for clinicians to prevent coronary heart disease: consensus statement of the European Atherosclerosis Society. Eur Heart J. 2013;34:3478–390a. doi: 10.1093/eurheartj/eht273.
    1. Gaudet D, Tremblay G, Perron P, Gagné C, Ouadahi Y, Moorjani S. [Familial hypercholesterolemia in eastern Quebec: a public health problem? The experience of the hyperlipidemia clinic of Chicoutimi]. Union Med Can. 1995;124:54–60.
    1. Raal FJ, Sjouke B, Hovingh GK, Isaac BF. Phenotype diversity among patients with homozygous familial hypercholesterolemia: a cohort study. Atherosclerosis. 2016;248:238–244. doi: 10.1016/j.atherosclerosis.2016.03.009.
    1. Raal FJ, Pilcher GJ, Panz VR, van Deventer HE, Brice BC, Blom DJ, Marais AD. Reduction in mortality in subjects with homozygous familial hypercholesterolemia associated with advances in lipid-lowering therapy. Circulation. 2011;124:2202–2207. doi: 10.1161/CIRCULATIONAHA.111.042523.
    1. Chora JR, Medeiros AM, Alves AC, Bourbon M. Analysis of publicly available LDLR, APOB, and PCSK9 variants associated with familial hypercholesterolemia: application of ACMG guidelines and implications for familial hypercholesterolemia diagnosis. Genet Med. 2018;20:591–598. doi: 10.1038/gim.2017.151.
    1. Etxebarria A, Benito-Vicente A, Alves AC, Ostolaza H, Bourbon M, Martin C. Advantages and versatility of fluorescence-based methodology to characterize the functionality of LDLR and class mutation assignment. PLoS One. 2014;9:e112677. doi: 10.1371/journal.pone.0112677.
    1. Strøm TB, Laerdahl JK, Leren TP. Mutation p.L799R in the LDLR, which affects the transmembrane domain of the LDLR, prevents membrane insertion and causes secretion of the mutant LDLR. Hum Mol Genet. 2015;24:5836–5844. doi: 10.1093/hmg/ddv304.
    1. Strøm TB, Tveten K, Laerdahl JK, Leren TP. Mutation G805R in the transmembrane domain of the LDL receptor gene causes familial hypercholesterolemia by inducing ectodomain cleavage of the LDL receptor in the endoplasmic reticulum. FEBS Open Bio. 2014;4:321–327. doi: 10.1016/j.fob.2014.03.007.
    1. Zodda D, Giammona R, Schifilliti S.Treatment strategy for dyslipidemia in cardiovascular disease prevention: focus on old and new drugs. Pharmacy (Basel) 20186pii: E10.
    1. France M. Homozygous familial hypercholesterolaemia: update on management. Paediatr Int Child Health. 2016;36:243–247. doi: 10.1080/20469047.2016.1246640.
    1. Raal FJ, Honarpour N, Blom DJ, Hovingh GK, Xu F, Scott R, Wasserman SM, Stein EA TESLA Investigators. Inhibition of PCSK9 with evolocumab in homozygous familial hypercholesterolaemia (TESLA Part B): a randomised, double-blind, placebo-controlled trial. Lancet. 2015;385:341–350. doi: 10.1016/S0140-6736(14)61374-X.
    1. Raal FJ, Hovingh GK, Blom D, Santos RD, Harada-Shiba M, Bruckert E, Couture P, Soran H, Watts GF, Kurtz C, et al. Long-term treatment with evolocumab added to conventional drug therapy, with or without apheresis, in patients with homozygous familial hypercholesterolaemia: an interim subset analysis of the open-label TAUSSIG study. Lancet Diabetes Endocrinol. 2017;5:280–290. doi: 10.1016/S2213-8587(17)30044-X.
    1. Stein EA, Honarpour N, Wasserman SM, Xu F, Scott R, Raal FJ. Effect of the proprotein convertase subtilisin/kexin 9 monoclonal antibody, AMG 145, in homozygous familial hypercholesterolemia. Circulation. 2013;128:2113–2120. doi: 10.1161/CIRCULATIONAHA.113.004678.
    1. Thedrez A, Blom DJ, Ramin-Mangata S, Blanchard V, Croyal M, Chemello K, Nativel B, Pichelin M, Cariou B, Bourane S, et al. Homozygous Familial Hypercholesterolemia Patients With Identical Mutations Variably Express the LDLR (Low-Density Lipoprotein Receptor): Implications for the Efficacy of Evolocumab. Arterioscler Thromb Vasc Biol. 2018;38:592–598. doi: 10.1161/ATVBAHA.117.310217.
    1. Gaudet D, Gipe DA, Pordy R, Ahmad Z, Cuchel M, Shah PK, Chyu KY, Sasiela WJ, Chan KC, Brisson D, et al. ANGPTL3 Inhibition in Homozygous Familial Hypercholesterolemia. N Engl J Med. 2017;377:296–297. doi: 10.1056/NEJMc1705994.
    1. Hassan M. ANGPLT3: a novel modulator of lipid metabolism. Glob Cardiol Sci Pract. 2017;2017:e201706. doi: 10.21542/gcsp.2017.6.
    1. Minicocci I, Santini S, Cantisani V, Stitziel N, Kathiresan S, Arroyo JA, Martí G, Pisciotta L, Noto D, Cefalù AB, et al. Clinical characteristics and plasma lipids in subjects with familial combined hypolipidemia: a pooled analysis. J Lipid Res. 2013;54:3481–3490. doi: 10.1194/jlr.P039875.
    1. Romeo S, Yin W, Kozlitina J, Pennacchio LA, Boerwinkle E, Hobbs HH, Cohen JC. Rare loss-of-function mutations in ANGPTL family members contribute to plasma triglyceride levels in humans. J Clin Invest. 2009;119:70–79. doi: 10.1172/JCI37118.
    1. Dewey FE, Gusarova V, Dunbar RL, O’Dushlaine C, Schurmann C, Gottesman O, McCarthy S, Van Hout CV, Bruse S, Dansky HM, et al. Genetic and pharmacologic inactivation of ANGPTL3 and cardiovascular disease. N Engl J Med. 2017;377:211–221. doi: 10.1056/NEJMoa1612790.
    1. Etxebarria A, Palacios L, Stef M, Tejedor D, Uribe KB, Oleaga A, Irigoyen L, Torres B, Ostolaza H, Martin C. Functional characterization of splicing and ligand-binding domain variants in the LDL receptor. Hum Mutat. 2012;33:232–243. doi: 10.1002/humu.21630.
    1. Benito-Vicente A, Alves AC, Etxebarria A, Medeiros AM, Martin C, Bourbon M. The importance of an integrated analysis of clinical, molecular, and functional data for the genetic diagnosis of familial hypercholesterolemia. Genet Med. 2015;17:980–988. doi: 10.1038/gim.2015.14.
    1. Melzer S, Nunes CS, Endringer DC, de Andrade TU, Tarnok A, Lenz D. Trypan blue as an affordable marker for automated live-dead cell analysis in image cytometry. Scanning. 2016;38:857–863. doi: 10.1002/sca.21335.
    1. Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, Grody WW, Hegde M, Lyon E, Spector E, et al. ACMG Laboratory Quality Assurance Committee. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17:405–424. doi: 10.1038/gim.2015.30.
    1. Lang W, Frishman WH. Angiopoietin-Like 3 protein inhibition: a new frontier in lipid-lowering treatment. Cardiol Rev. 2019;27:211–217. doi: 10.1097/CRD.0000000000000258.
    1. Tikka A, Jauhiainen M. The role of ANGPTL3 in controlling lipoprotein metabolism. Endocrine. 2016;52:187–193. doi: 10.1007/s12020-015-0838-9.
    1. Etxebarria A, Benito-Vicente A, Palacios L, Stef M, Cenarro A, Civeira F, Ostolaza H, Martin C. Functional characterization and classification of frequent low-density lipoprotein receptor variants. Hum Mutat. 2015;36:129–141. doi: 10.1002/humu.22721.
    1. Rodríguez-Jiménez C, Pernía O, Mostaza J, Rodríguez-Antolín C, de Dios García-Díaz J, Alonso-Cerezo C, García-Polo I, Blanco A, Lahoz C, Arrieta F, et al. Functional analysis of new variants at the low-density lipoprotein receptor associated with familial hypercholesterolemia. Hum Mutat. 2019;40:1181–1190. doi: 10.1002/humu.23801.
    1. Silva S, Alves AC, Patel D, Malhó R, Soutar AK, Bourbon M. In vitro functional characterization of missense mutations in the LDLR gene. Atherosclerosis. 2012;225:128–134. doi: 10.1016/j.atherosclerosis.2012.08.017.
    1. Di Taranto MD, Benito-Vicente A, Giacobbe C, Uribe KB, Rubba P, Etxebarria A, Guardamagna O, Gentile M, Martín C, Fortunato G. Identification and in vitro characterization of two new PCSK9 Gain of Function variants found in patients with Familial Hypercholesterolemia. Sci Rep. 2017;7:15282. doi: 10.1038/s41598-017-15543-x.
    1. Moorjani S, Roy M, Torres A, Bétard C, Gagné C, Lambert M, Brun D, Davignon J, Lupien P. Mutations of low-density-lipoprotein-receptor gene, variation in plasma cholesterol, and expression of coronary heart disease in homozygous familial hypercholesterolaemia. Lancet. 1993;341:1303–1306. doi: 10.1016/0140-6736(93)90815-x.
    1. Thompson GR, Blom DJ, Marais AD, Seed M, Pilcher GJ, Raal FJ. Survival in homozygous familial hypercholesterolaemia is determined by the on-treatment level of serum cholesterol. Eur Heart J. 2018;39:1162–1168. doi: 10.1093/eurheartj/ehx317.
    1. Aegerion Pharmaceuticals Inc. JUXTAPID (lomitapide) capsules, for oral use. Highlights of prescribing information. 2017. Available at: . Accessed September 4, 2018.
    1. Kastle Therapeutics. KYNAMRO (mipomersen sodium) Injection. Solution for Subcutaneous Injection. Highlights of Prescribing Information. 2016. Available at: . Accessed September 4, 2018.
    1. Amgen Inc. Repatha prescribing information. 2015. Available at: . Accessed July 2, 2018.
    1. Sanofi-aventis U.S. LLC. Praluent prescribing information. 2015. Available at: . Accessed July 2, 2018.
    1. Catapano AL, Graham I, De Backer G, Wiklund O, Chapman MJ, Drexel H, Hoes AW, Jennings CS, Landmesser U, Pedersen TR, et al. ESC Scientific Document Group. 2016 ESC/EAS guidelines for the management of dyslipidaemias. Eur Heart J. 2016;37:2999–3058. doi: 10.1093/eurheartj/ehw272.
    1. Landmesser U, Chapman MJ, Stock JK, Amarenco P, Belch JJF, Borén J, Farnier M, Ference BA, Gielen S, Graham I, et al. 2017 Update of ESC/EAS Task Force on practical clinical guidance for proprotein convertase subtilisin/kexin type 9 inhibition in patients with atherosclerotic cardiovascular disease or in familial hypercholesterolaemia. Eur Heart J. 2018;39:1131–1143. doi: 10.1093/eurheartj/ehx549.
    1. Lloyd-Jones DM, Morris PB, Ballantyne CM, Birtcher KK, Daly DD, Jr, DePalma SM, Minissian MB, Orringer CE, Smith SC., Jr. 2016 ACC expert consensus decision pathway on the role of non-statin therapies for LDL-cholesterol lowering in the management of atherosclerotic cardiovascular disease risk: a report of the American College of Cardiology Task Force on Clinical Expert Consensus Documents. J Am Coll Cardiol. 2016;68:92–125.
    1. Farnier M, Gaudet D, Valcheva V, Minini P, Miller K, Cariou B. Efficacy of alirocumab in high cardiovascular risk populations with or without heterozygous familial hypercholesterolemia: Pooled analysis of eight ODYSSEY Phase 3 clinical program trials. Int J Cardiol. 2016;223:750–757. doi: 10.1016/j.ijcard.2016.08.273.
    1. Raal FJ, Stein EA, Dufour R, Turner T, Civeira F, Burgess L, Langslet G, Scott R, Olsson AG, Sullivan D, et al. RUTHERFORD-2 Investigators. PCSK9 inhibition with evolocumab (AMG 145) in heterozygous familial hypercholesterolaemia (RUTHERFORD-2): a randomised, double-blind, placebo-controlled trial. Lancet. 2015;385:331–340. doi: 10.1016/S0140-6736(14)61399-4.
    1. Stein EA, Gipe D, Bergeron J, Gaudet D, Weiss R, Dufour R, Wu R, Pordy R. Effect of a monoclonal antibody to PCSK9, REGN727/SAR236553, to reduce low-density lipoprotein cholesterol in patients with heterozygous familial hypercholesterolaemia on stable statin dose with or without ezetimibe therapy: a phase 2 randomised controlled trial. Lancet. 2012;380:29–36. doi: 10.1016/S0140-6736(12)60771-5.

Source: PubMed

3
订阅