95 results on '"Tsimikas, Sotirios"'
Search Results
2. Contributors
- Author
-
Balasubramanyam, Ashok, primary, Ballantyne, Christie M., additional, Barter, Philip J., additional, Blumenthal, Roger S., additional, Boffa, Michael B., additional, Brewer, Bryan, additional, Brinton, Eliot A., additional, Brothers, Julie A., additional, Carlson, Lars A., additional, Catapano, Alberico L., additional, Chiavaroli, Laura, additional, Church, Timothy S., additional, Cohen, David E., additional, Creider, Julia C., additional, Daniels, Stephen R., additional, Davidson, David J., additional, Davidson, Michael H., additional, Deedwania, Prakash, additional, deGoma, Emil M., additional, Do, Rose Q., additional, Fayad, Zahi A., additional, Fazio, Sergio, additional, Fellstrøm, Bengt, additional, Fleming, Jennifer, additional, Gillard, Baiba K., additional, Ginsberg, Henry N., additional, Goldberg, Anne Carol, additional, Gotto, Antonio M., additional, Guyton, John R., additional, Harris, William S., additional, Hegele, Robert A., additional, Holdaas, Hallvard, additional, Hoogeveen, Ron C., additional, Jacobson, Terry A., additional, Jardine, Alan G., additional, Jenkins, David J.A., additional, Johnson, Amber E., additional, Jones, Peter, additional, Jones, Peter H., additional, Kastelein, John J.P., additional, Kendall, Cyril W.C., additional, Kobashigawa, Jon, additional, Koschinsky, Marlys L., additional, Kris-Etherton, Penny M., additional, Kuivenhoven, Jan Albert, additional, Lairez, Olivier, additional, Lavie, Carl J., additional, Libby, Peter, additional, Linton, MacRae F., additional, Marcovina, Santica M., additional, Mark, Patrick B., additional, Martin, Seth S., additional, McGovern, Mark E., additional, McKenney, James M., additional, Miller, Michael, additional, Miller, Yury I., additional, Mirrahimi, Arash, additional, Moon, Jennifer E., additional, Moriarty, Patrick M., additional, Motazacker, Mohammad Mahdi, additional, Nambi, Vijay, additional, Negi, Smita I., additional, Nicholls, Stephen J., additional, Nissen, Steven E., additional, Norata, Giuseppe D., additional, Patel, Payal S., additional, Patel, Rajan K., additional, Xavier Pi-Sunyer, F., additional, Pokharel, Yashashwi, additional, Pownall, Henry J., additional, Raal, Frederick, additional, Rader, Daniel J., additional, Reyes-Soffer, Gissette, additional, Ridker, Paul M., additional, Rosales, Corina, additional, Rosenson, Robert S., additional, Roth, Eli M., additional, Rye, Kerry-Anne, additional, Santos, Raul D., additional, Sarzynski, Mark A., additional, Schwartz, Gregory G., additional, Sekhar, Rajagopal V., additional, Singh, Parmanand, additional, Skulas-Ray, Ann C., additional, Srichaikul, Kristie, additional, Stafforini, Diana M., additional, Stein, Evan A., additional, Stone, Neil J., additional, Swift, Damon L., additional, Tawakol, Ahmed, additional, Taylor, Allen J., additional, Tsimikas, Sotirios, additional, Turner, Traci, additional, Vijayaraghavan, Krishnaswami, additional, Virani, Salim S., additional, Wang, David Q.-H., additional, Wenger, Nanette K., additional, Wilkinson, Michael J., additional, and Wong, Julia M.W., additional
- Published
- 2015
- Full Text
- View/download PDF
3. Lipoprotein Oxidation and Modification
- Author
-
Miller, Yury I., primary and Tsimikas, Sotirios, additional
- Published
- 2009
- Full Text
- View/download PDF
4. Contributing Authors
- Author
-
Assmann, Gerd, primary, Bagshaw, Deborah, additional, Balasubramanyam, Ashok, additional, Ballantyne, Christie M., additional, Barter, Philip, additional, Bays, Harold, additional, Blumenthal, Roger S., additional, Brewer, H. Bryan, additional, Brown, B. Greg, additional, Brunzell, John D., additional, Campbell, Catherine Y., additional, Canner, Paul L., additional, Carlson, Lars A., additional, Catapano, A.L., additional, Chahil, Tina J., additional, Church, Timothy S., additional, Cohen, David E., additional, Davidson, Michael H., additional, Deedwania, Prakash C., additional, Després, Jean-Pierre, additional, Devaraj, Sridevi, additional, Devine, Patrick J., additional, Fayad, Zahi A., additional, Fazio, Sergio, additional, Fellstrøm, Bengt, additional, Ganz, Peter, additional, Ginsberg, Henry N., additional, Goldberg, Anne Carol, additional, Gotto, Antonio M., additional, Guyton, John R., additional, Harris, William S., additional, Holdaas, Hallvard, additional, Hoogeveen, Ron C., additional, Jacobson, Terry A., additional, Jardine, Alan G., additional, Jenkins, David J.A., additional, Jialal, Ishwarlal, additional, Jones, Peter H., additional, Josse, Andrea R., additional, Kendall, Cyril W.C., additional, Kobashigawa, Jon A., additional, Koschinsky, Marlys L., additional, Kris-Etherton, Penny M., additional, Kurra, Salila, additional, Lavie, Carl J., additional, Le, Ngoc-Anh, additional, Libby, Peter, additional, Linton, MacRae F., additional, Marcovina, Santica M., additional, Mark, Patrick B., additional, McGovern, Mark E., additional, McKenney, James M., additional, Merz, C. Noel Bairey, additional, Miller, Michael, additional, Miller, Yury I., additional, Mora, Samia, additional, Moriarty, Patrick M., additional, Musunuru, Kiran, additional, Myers, Kelly S., additional, Nambi, Vijay, additional, Nguyen, Tri H., additional, Nicholls, Stephen J., additional, Nissen, Steven E., additional, Norata, G.D., additional, Ohlson, Melissa, additional, Packard, Chris J., additional, Pi-Sunyer, F. Xavier, additional, Polk, Donna, additional, Pownall, Henry J., additional, Rader, Daniel J., additional, Rosenson, Robert S., additional, Rudd, James H.F., additional, Saseen, Joseph S., additional, Schwartz, Gregory G., additional, Seedorf, Udo, additional, Sekhar, Rajagopal V., additional, Stone., Neil J., additional, Taylor, Allen J., additional, Tsimikas, Sotirios, additional, Vijayaraghavan, Krishnaswami, additional, Wang, David Q.-H., additional, Wenger, Nanette K., additional, Wiggins, Barbara S., additional, Wilson, Peter W.F., additional, and Wong, Julia M.W., additional
- Published
- 2009
- Full Text
- View/download PDF
5. Lipoprotein Oxidation, Macrophages, Immunity, and Atherogenesis
- Author
-
Tsimikas, Sotirios, primary, Glass, Christopher K., additional, Steinberg, Daniel, additional, and Witztum, Joseph L., additional
- Published
- 2001
- Full Text
- View/download PDF
6. Molecular Biology of Lipoproteins and Dyslipidemias
- Author
-
Tsimikas, Sotirios, primary and Mooser, Vincent, additional
- Published
- 1999
- Full Text
- View/download PDF
7. Determinants of binding of oxidized phospholipids on apolipoprotein (a) and lipoprotein (a)1
- Author
-
Leibundgut, Gregor, Scipione, Corey, Yin, Huiyong, Schneider, Matthias, Boffa, Michael B., Green, Simone, Yang, Xiaohong, Dennis, Edward, Witztum, Joseph L., Koschinsky, Marlys L., and Tsimikas, Sotirios
- Subjects
Pan troglodytes ,Enzyme-Linked Immunosorbent Assay ,Mice, Transgenic ,QD415-436 ,Biochemistry ,Mice ,Species Specificity ,Tandem Mass Spectrometry ,Animals ,Humans ,Research Articles ,Apolipoproteins A ,Phospholipids ,Binding Sites ,Gorilla gorilla ,kringles ,Pan paniscus ,Mice, Inbred C57BL ,lipoproteins ,Macaca fascicularis ,plasminogen ,lipids (amino acids, peptides, and proteins) ,Oxidation-Reduction ,Lipoprotein(a) ,Papio ,Protein Binding - Abstract
Oxidized phospholipids (OxPLs) are present on apolipoprotein (a) [apo(a)] and lipoprotein (a) [Lp(a)] but the determinants influencing their binding are not known. The presence of OxPLs on apo(a)/Lp(a) was evaluated in plasma from healthy humans, apes, monkeys, apo(a)/Lp(a) transgenic mice, lysine binding site (LBS) mutant apo(a)/Lp(a) mice with Asp(55/57)→Ala(55/57) substitution of kringle (K)IV10)], and a variety of recombinant apo(a) [r-apo(a)] constructs. Using antibody E06, which binds the phosphocholine (PC) headgroup of OxPLs, Western and ELISA formats revealed that OxPLs were only present in apo(a) with an intact KIV10 LBS. Lipid extracts of purified human Lp(a) contained both E06- and nonE06-detectable OxPLs by tandem liquid chromatography-mass spectrometry (LC-MS/MS). Trypsin digestion of 17K r-apo(a) showed PC-containing OxPLs covalently bound to apo(a) fragments by LC-MS/MS that could be saponified by ammonium hydroxide. Interestingly, PC-containing OxPLs were also present in 17K r-apo(a) with Asp(57)→Ala(57) substitution in KIV10 that lacked E06 immunoreactivity. In conclusion, E06- and nonE06-detectable OxPLs are present in the lipid phase of Lp(a) and covalently bound to apo(a). E06 immunoreactivity, reflecting pro-inflammatory OxPLs accessible to the immune system, is strongly influenced by KIV10 LBS and is unique to human apo(a), which may explain Lp(a)'s pro-atherogenic potential.
- Published
- 2013
8. Secretory Phospholipase A(2)-IIA and Cardiovascular Disease
- Author
-
Holmes, Michael V., Simon, Tabassome, J Exeter, Holly, Folkersen, Lasse, Asselbergs, Folkert W., Guardiola, Montse, Cooper, Jackie A., Palmen, Jutta, Hubacek, Jaroslav A., Carruthers, Kathryn F., Horne, Benjamin D., Brunisholz, Kimberly D., Mega, Jessica L., Van Iperen, Erik P A, Li, Mingyao, Leusink, Maarten, Trompet, Stella, Verschuren, Jeffrey J W., Kees Hovingh, G, Dehghan, Abbas, Nelson, Christopher P., Kotti, Salma, Danchin, Nicolas, Scholz, Markus, Haase L., Christiane, Rothenbacher, Dietrich, Swerdlow, Daniel I., Kuchenbaecker, Karoline B., Staines-Urias, Eleonora, Goel, Anuj, van t Hooft, Ferdinand, Gertow, Karl, de Faire, Ulf, Panayiotou, Andrie G., Tremoli, Elena, Baldassarre, Damiano, Veglia, Fabrizio, Holdt, Lesca M., Beutner, Frank, Gansevoort, Ron T., Navis, Gerjan J., Mateo Leach, Irene, Breitling, Lutz P., Brenner, Hermann, Thiery, Joachim, Dallmeier, Dhayana, Franco-Cereceda, Anders, Boer, Jolanda M A., Stephens, Jeffrey W., Hofker, Marten H., Tedgui, Alain, Hofman, Albert, Uitterlinden, Andre G., Adamkova, Vera, Pitha, Jan, Onland-Moret, Charlotte N., Cramer, Maarten J., Nathoe, Hendrik M., Spiering, Wilko, Klungel, Olaf H., Kumari, Meena, Whincup, Peter H., Morrow, David A., Braund, Peter S., Hall, Alistair S., Olsson, Anders G., Doevendans, Pieter A., Trip, Mieke D., Tobin, Martin D., Hamsten, Anders, Watkins, Hugh, Koenig, Wolfgang, Nicolaides, Andrew N., Teupser, Daniel, Day, Ian N M, F Carlquist, John, Gaunt, Tom R., Ford, Ian, Sattar, Naveed, Tsimikas, Sotirios, Schwartz, Gregory G., Lawlor, Debbie A., Morris, Richard W., Sandhu, Manjinder S., Poledne, Rudolf, Maitland-van der Zee, Anke H., Khaw, Kay-Tee, Keating, Brendan J., van der Harst, Pim, Price, Jackie F., Mehta, Shamir R., Yusuf, Salim, Witteman, Jaqueline C M, Franco, Oscar H., Jukema, Wouter J., de Knijff, Peter, Tybjaerg-Hansen, Anne, Rader, Daniel J., Farrall, Martin, Samani, Nilesh J., Kivimaki, Mika, Fox, Keith A A., Humphries, Steve E., Anderson, Jeffrey L., Boekholdt, Matthijs S., Palmer, Tom M., Eriksson, Per, Pare, Guillaume, Hingorani, Aroon D., Sabatine, Marc S., Mallat, Ziad, Casas, Juan P., Talmud, Philippa J., Holmes, Michael V., Simon, Tabassome, J Exeter, Holly, Folkersen, Lasse, Asselbergs, Folkert W., Guardiola, Montse, Cooper, Jackie A., Palmen, Jutta, Hubacek, Jaroslav A., Carruthers, Kathryn F., Horne, Benjamin D., Brunisholz, Kimberly D., Mega, Jessica L., Van Iperen, Erik P A, Li, Mingyao, Leusink, Maarten, Trompet, Stella, Verschuren, Jeffrey J W., Kees Hovingh, G, Dehghan, Abbas, Nelson, Christopher P., Kotti, Salma, Danchin, Nicolas, Scholz, Markus, Haase L., Christiane, Rothenbacher, Dietrich, Swerdlow, Daniel I., Kuchenbaecker, Karoline B., Staines-Urias, Eleonora, Goel, Anuj, van t Hooft, Ferdinand, Gertow, Karl, de Faire, Ulf, Panayiotou, Andrie G., Tremoli, Elena, Baldassarre, Damiano, Veglia, Fabrizio, Holdt, Lesca M., Beutner, Frank, Gansevoort, Ron T., Navis, Gerjan J., Mateo Leach, Irene, Breitling, Lutz P., Brenner, Hermann, Thiery, Joachim, Dallmeier, Dhayana, Franco-Cereceda, Anders, Boer, Jolanda M A., Stephens, Jeffrey W., Hofker, Marten H., Tedgui, Alain, Hofman, Albert, Uitterlinden, Andre G., Adamkova, Vera, Pitha, Jan, Onland-Moret, Charlotte N., Cramer, Maarten J., Nathoe, Hendrik M., Spiering, Wilko, Klungel, Olaf H., Kumari, Meena, Whincup, Peter H., Morrow, David A., Braund, Peter S., Hall, Alistair S., Olsson, Anders G., Doevendans, Pieter A., Trip, Mieke D., Tobin, Martin D., Hamsten, Anders, Watkins, Hugh, Koenig, Wolfgang, Nicolaides, Andrew N., Teupser, Daniel, Day, Ian N M, F Carlquist, John, Gaunt, Tom R., Ford, Ian, Sattar, Naveed, Tsimikas, Sotirios, Schwartz, Gregory G., Lawlor, Debbie A., Morris, Richard W., Sandhu, Manjinder S., Poledne, Rudolf, Maitland-van der Zee, Anke H., Khaw, Kay-Tee, Keating, Brendan J., van der Harst, Pim, Price, Jackie F., Mehta, Shamir R., Yusuf, Salim, Witteman, Jaqueline C M, Franco, Oscar H., Jukema, Wouter J., de Knijff, Peter, Tybjaerg-Hansen, Anne, Rader, Daniel J., Farrall, Martin, Samani, Nilesh J., Kivimaki, Mika, Fox, Keith A A., Humphries, Steve E., Anderson, Jeffrey L., Boekholdt, Matthijs S., Palmer, Tom M., Eriksson, Per, Pare, Guillaume, Hingorani, Aroon D., Sabatine, Marc S., Mallat, Ziad, Casas, Juan P., and Talmud, Philippa J.
- Abstract
Objectives This study sought to investigate the role of secretory phospholipase A(2) (sPLA(2))-IIA in cardiovascular disease. less thanbrgreater than less thanbrgreater thanBackground Higher circulating levels of sPLA(2)-IIA mass or sPLA(2) enzyme activity have been associated with increased risk of cardiovascular events. However, it is not clear if this association is causal. A recent phase III clinical trial of an sPLA(2) inhibitor (varespladib) was stopped prematurely for lack of efficacy. less thanbrgreater than less thanbrgreater thanMethods We conducted a Mendelian randomization meta-analysis of 19 general population studies (8,021 incident, 7,513 prevalent major vascular events [MVE] in 74,683 individuals) and 10 acute coronary syndrome (ACS) cohorts (2,520 recurrent MVE in 18,355 individuals) using rs11573156, a variant in PLA2G2A encoding the sPLA(2)-IIA isoenzyme, as an instrumental variable. less thanbrgreater than less thanbrgreater thanResults PLA2G2A rs11573156 C allele associated with lower circulating sPLA(2)-IIA mass (38% to 44%) and sPLA(2) enzyme activity (3% to 23%) per C allele. The odds ratio (OR) for MVE per rs11573156 C allele was 1.02 (95% confidence interval [CI]: 0.98 to 1.06) in general populations and 0.96 (95% CI: 0.90 to 1.03) in ACS cohorts. In the general population studies, the OR derived from the genetic instrumental variable analysis for MVE for a 1-log unit lower sPLA(2)-IIA mass was 1.04 (95% CI: 0.96 to 1.13), and differed from the non-genetic observational estimate (OR: 0.69; 95% CI: 0.61 to 0.79). In the ACS cohorts, both the genetic instrumental variable and observational ORs showed a null association with MVE. Instrumental variable analysis failed to show associations between sPLA2 enzyme activity and MVE. less thanbrgreater than less thanbrgreater thanConclusions Reducing sPLA(2)-IIA mass is unlikely to be a useful therapeutic goal for preventing cardiovascular events., Funding Agencies|BHG programme grant|RG/10/12/28456|UK Medical Research Council (Population Health Scientist Fellowship)|G0802432|British Heart Foundation|RG 10/12/28456RG008/014RG/10/001/27643PG07/133/24260FS 08/ 048/25628|National Institute of Health Research University College London Hospitals Biomedical Research Centre||Dutch Kidney Foundation|E033|Netherlands Organisation for Health Research and Development (NWO VENI)|916.761.70|Dutch Inter University Cardiology Institute Netherlands (ICIN)||Canada Research Chair in Genetic and Molecular Epidemiology||Europe Against Cancer Programme of the European Commission (SANCO), Dutch Ministry of Public Health, Welfare and Sports (VWS), Netherlands Cancer Registry (NKR), LK Research Funds, Dutch Prevention Funds, Dutch Cancer Society|||EU-LSHM-CT-2006037697
- Published
- 2013
- Full Text
- View/download PDF
9. Associations between lipoprotein(a), oxidized phospholipids, and extracoronary vascular disease.
- Author
-
Bellomo TR, Liu Y, Gilliland TC, Miksenas H, Haidermota S, Wong M, Hu X, Cristino JR, Browne A, Plutzky J, Tsimikas S, Januzzi JL Jr, and Natarajan P
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Vascular Diseases blood, Vascular Diseases metabolism, Lipoprotein(a) blood, Phospholipids blood, Phospholipids metabolism, Oxidation-Reduction
- Abstract
The roles of lipoprotein(a) [Lp(a)] and related oxidized phospholipids (OxPLs) in the development and progression of coronary disease is known, but their influence on extracoronary vascular disease is not well-established. We sought to evaluate associations between Lp(a), OxPL apolipoprotein B (OxPL-apoB), and apolipoprotein(a) (OxPL-apo(a)) with angiographic extracoronary vascular disease and incident major adverse limb events (MALEs). Four hundred forty-six participants who underwent coronary and/or peripheral angiography were followed up for a median of 3.7 years. Lp(a) and OxPLs were measured before angiography. Elevated Lp(a) was defined as ≥150 nmol/L. Elevated OxPL-apoB and OxPL-apo(a) were defined as greater than or equal to the 75th percentile (OxPL-apoB ≥8.2 nmol/L and OxPL-apo(a) ≥35.8 nmol/L, respectively). Elevated Lp(a) had a stronger association with the presence of extracoronary vascular disease compared to OxPLs and was minimally improved with the addition of OxPLs in multivariable models. Compared to participants with normal Lp(a) and OxPL concentrations, participants with elevated Lp(a) levels were twice as likely to experience a MALE (odds ratio: 2.14, 95% confidence interval: 1.03, 4.44), and the strength of the association as well as the C statistic of 0.82 was largely unchanged with the addition of OxPL-apoB and OxPL-apo(a). Elevated Lp(a) and OxPLs are risk factors for progression and complications of extracoronary vascular disease. However, the addition of OxPLs to Lp(a) does not provide additional information about risk of extracoronary vascular disease. Therefore, Lp(a) alone captures the risk profile of Lp(a), OxPL-apoB, and OxPL-apo(a) in the development and progression of atherosclerotic plaque in peripheral arteries., Competing Interests: Conflict of interest This study was sponsored by Novartis as an investigator-initiated study. The sponsor provided feedback on the study design but not data interpretation. X. H., J. R. C., and A. B. are employees of Novartis. S. T. is a coinventor and receives royalties from patents owned by University of California San Diego (UCSD) and is a cofounder and has an equity interest in Oxitope, LLC and its affiliates, Kleanthi Diagnostics, LLC and Covicept Therapeutics, Inc and has a dual appointment at UCSD and Ionis Pharmaceuticals. The terms of this arrangement have been reviewed and approved by the University of California, San Diego, in accordance with its conflict-of-interest policies. J. L. J. is a Trustee of the American College of Cardiology, a Board member of Imbria Pharmaceuticals, a Director at Jana Care, has received grant support from Applied Therapeutics, Innolife, Novartis Pharmaceuticals, and Abbott Diagnostics, consulting income from Abbott, Janssen, Novartis, Prevencio, and Roche Diagnostics and participates in clinical endpoint committees/data safety monitoring boards for Abbott, AbbVie, Bayer, CVRx, Siemens, and Takeda. P.N. reports research grants from Allelica, Amgen, Apple, Boston Scientific, Genentech, Rochee, and Novartis, personal fees from Allelica, Apple, AstraZeneca, Blackstone Life Sciences, Creative Education Concepts, CRISPR Therapeutics, Eli Lilly & Co, Foresite Labs, Genentech, Rochee, GV, HeartFlow, Magnet Biomedicine, Merck, and Novartis, scientific advisory board membership of Esperion Therapeutics, Preciseli, and TenSixteen Bio, scientific co-founder of TenSixteen Bio, equity in MyOme, Preciseli, and TenSixteen Bio, and spousal employment at Vertex Pharmaceuticals, all unrelated to the present work. The remaining authors have nothing to disclose. Although these relationships have been identified for conflict-of-interest management based on the project's overall scope, the research findings included in this particular publication may not necessarily relate to the interests of the above companies., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
10. Corrigendum to "Volanesorsen and triglyceride levels in familial chylomicronemia syndrome: Long-term efficacy and safety data from patients in an open-label extension trial" [Journal of Clinical Lipidology, Volume 17, Issue 3, May-June 2023, Pages 342-355].
- Author
-
Witztum JL, Gaudet D, Arca M, Jones A, Soran H, Gouni-Berthold I, Stroes ESG, Alexander VJ, Jones R, Watts L, Xia S, and Tsimikas S
- Published
- 2024
- Full Text
- View/download PDF
11. Atherothrombotic and thrombolytic biomarkers in incident stroke and atrial fibrillation-related stroke: The Multi-Ethnic Study of Atherosclerosis (MESA).
- Author
-
Lidani KCF, Trainor PJ, Bhatia HS, Nasir K, Blaha MJ, Tsai MY, Gottesman RF, Post WS, Thanassoulis G, Tsimikas S, Heckbert SR, and DeFilippis AP
- Subjects
- Humans, Risk Assessment methods, Biomarkers, Plasminogen, Risk Factors, Atrial Fibrillation complications, Atrial Fibrillation diagnosis, Atrial Fibrillation drug therapy, Ischemic Attack, Transient complications, Stroke diagnosis, Stroke epidemiology, Stroke complications, Atherosclerosis complications
- Abstract
Background and Aims: Although several biomarkers have been studied in thromboembolic stroke, measuring the balance between thrombus formation and thrombolysis and data on its role in predicting stroke and atrial fibrillation (AF)-related stroke is limited. We sought to assess atherothrombotic biomarkers grouped into composite factors that reflect thrombotic and thrombolytic potential, and the balance between these factors as it relates to incident stroke or transient ischemic attack (TIA) and stroke/TIA in AF., Methods: A Thrombotic Factor, derived from fibrinogen, plasmin-antiplasmin complex, factor VIII, D-dimer, and lipoprotein(a); and a Thrombolytic Factor, derived from plasminogen and oxidized phospholipids on plasminogen, were evaluated at baseline in 5,764 Multi-Ethnic Study of Atherosclerosis (MESA) participants. We evaluated the association between these two factors representative of thrombotic and thrombolytic potential and incident stroke/TIA (n = 402), and AF-related stroke/TIA (n = 82) over a median of 13.9 and 3.7 years, respectively. Cox proportional hazard models adjusted for medication use, cardiovascular risk factors and CHA
2 DS2 -VASc score were utilized. Harrell's C-index was estimated to evaluate model performance., Results: In models including both factors, Thrombotic Factor was positively while Thrombolytic Factor was inversely associated with incident stroke/TIA and AF-related stroke/TIA. Incorporating these factors along with the CHA2 DS2 -VASc in adjusted models resulted in a small improvement in risk prediction of incident stroke/TIA and AF-related stroke/TIA compared to models without the factors (C-index from 0.697 to 0.704, and from 0.657 to 0.675, respectively)., Conclusions: Composite biomarker factors, representative of the balance between thrombotic and thrombolytic propensity, provided an improvement in predicting stroke/TIA beyond CHA2 DS2 -VASc score., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
12. Lipoprotein(a) levels and carotid intima-media thickness in children: A 20-year follow-up study.
- Author
-
de Boer LM, Hutten BA, Tsimikas S, Yeang C, Zwinderman AH, Kroon J, Revers A, Kastelein JJP, and Wiegman A
- Subjects
- Adolescent, Child, Female, Humans, Male, Follow-Up Studies, Risk Factors, Carotid Intima-Media Thickness, Hyperlipoproteinemia Type II blood, Lipoprotein(a) blood
- Abstract
Elevated lipoprotein(a) [Lp(a)] is independently associated with cardiovascular disease (CVD). In a recent long-term follow-up study involving children with familial hypercholesterolemia (FH), Lp(a) levels contributed significantly to early atherosclerosis, as measured by carotid intima-media thickness (cIMT). To determine if this holds true for children without FH, we conducted a 20-year follow-up study, examining 88 unaffected siblings (mean age: 12.9 years) of children with FH. No significant association was found between Lp(a) and cIMT during follow-up (ß-adjusted [95% confidence interval] = 0.0001 [-0.008 to 0.008] mm per 50 nmol/L increase Lp(a), p = 0.97). In conclusion, our findings suggest that elevated levels of Lp(a) do not play a significant role in arterial wall thickening among children without FH during the 20-year follow-up period. This leads us to consider the possibility that cIMT may not be a suitable marker for detecting potential subtle changes in the arterial wall mediated by Lp(a) in the young, general population. However, it could also be that elevated Lp(a) is only a significant risk factor for atherosclerosis in the presence of other risk factors such as FH., (Copyright © 2023. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
13. Clinical trials to improve outcomes in patients with elevated Lp(a) undergoing PCI: The time has arrived.
- Author
-
Tsimikas S, Bhatia HS, and Erlinge D
- Subjects
- Humans, Angina Pectoris, Treatment Outcome, Risk Factors, Percutaneous Coronary Intervention, Coronary Artery Disease surgery, Coronary Artery Disease drug therapy
- Published
- 2023
- Full Text
- View/download PDF
14. Lipoprotein(a) and the pooled cohort equations for ASCVD risk prediction: The Multi-Ethnic Study of Atherosclerosis.
- Author
-
Bhatia HS, Rikhi R, Allen TS, Yeang C, Guan W, Garg PK, Tsai MY, Criqui MH, Shapiro MD, and Tsimikas S
- Subjects
- Humans, Risk Assessment, Risk Factors, Lipoprotein(a), Atherosclerosis diagnosis
- Abstract
Background and Aims: Lipoprotein(a) [Lp(a)] is an independent risk factor for atherosclerotic cardiovascular disease (ASCVD) but is not included in the Pooled Cohort Equations (PCE). We aimed to assess how well the PCE predict 10-year event rates in individuals with elevated Lp(a), and whether the addition of Lp(a) improves risk prediction., Methods: We compared observed versus PCE-predicted 10-year ASCVD event rates, stratified by Lp(a) level and ASCVD risk category using Poisson regression, and evaluated the association between Lp(a) > 50 mg/dL and ASCVD risk using Cox proportional hazards models in the Multi-Ethnic Study of Atherosclerosis (MESA). We evaluated the C-index and net reclassification improvement (NRI) with addition of Lp(a) to the PCE., Results: The study population included 6639 individuals (20%, n = 1325 with elevated Lp(a)). The PCE accurately predicted 10-year event rates for individuals with elevated Lp(a) with observed event rates falling within predicted limits. Elevated Lp(a) was associated with increased risk of CVD events overall (HR 1.27, 95% CI 1.00-1.60), particularly in low (HR 2.45, 95% CI 1.40-4.31), and high-risk (HR 1.41, 95% CI 1.02-1.96) individuals. Continuous NRI (95% CI) with the addition of Lp(a) to the PCE for CVD was 0.0963 (0.0158-0.1953) overall, and 0.2999 (0.0876, 0.5525) among low-risk individuals., Conclusions: The PCE performs well for event rate prediction in individuals with elevated Lp(a). However, Lp(a) is associated with increased CVD risk, and the addition of Lp(a) to the PCE improves risk prediction, particularly among low-risk individuals. These results lend support for increasing use of Lp(a) testing for risk assessment., (Copyright © 2023 Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
15. Effects of mixed nut consumption on LDL cholesterol, lipoprotein(a), and other cardiometabolic risk factors in overweight and obese adults.
- Author
-
Nora CL, Zhang L, Castro RJ, Marx A, Carman HB, Lum T, Tsimikas S, and Hong MY
- Subjects
- Humans, Adult, Cholesterol, LDL, Cardiometabolic Risk Factors, Lipoprotein(a), Adiponectin, 8-Hydroxy-2'-Deoxyguanosine metabolism, Obesity diagnosis, Obesity metabolism, Risk Factors, Inflammation diagnosis, Inflammation prevention & control, Inflammation metabolism, Nuts, Overweight diagnosis
- Abstract
Background and Aims: Elevated LDL-C, lipoprotein(a) [Lp(a)], and inflammation are associated with greater risk for atherosclerotic cardiovascular events. Consumption of individual nut types decreases these risk factors but knowledge about the effect of mixed nuts on Lp(a) is limited. The objective of this study was to determine the effects of consuming 42.5 g/day of mixed nuts on LDL-C, Lp(a), and inflammatory markers in individuals with overweight or obesity., Methods and Results: In a 16-week randomized control trial, 29 participants with overweight or obesity (BMI 25-40 kg/m
2 ) consumed either 42.5 g/day of mixed nuts (cashews, almonds, macadamia nuts, Brazil nuts, pecans, pistachios, walnuts, and peanuts) or 69 g/day isocaloric pretzels. Blood samples were collected at baseline, week 8, and week 16 for analysis on total cholesterol (TC), LDL-C, Lp(a), inflammation markers, glucose, insulin, adiponectin and liver function enzymes. No significant differences were seen in TC, LDL-C, HDL-C, Lp(a), or liver function enzymes between the two groups. Participants consuming mixed nuts had significantly lower body fat percentage and diastolic blood pressure, and higher adiponectin (all P ≤ 0.05). C-reactive protein (CRP) and 8-oxo-deoxyguanosis (8-oxodG) showed non-significant decreasing trends and total antioxidant capacity (TAC) had a non-significant increasing trend in the mixed nut group., Conclusion: Consumption of mixed nuts had no evidence of an effect on LDL-C or Lp(a) throughout the intervention. Notably, mixed nut consumption lowered body fat percentage without significant changes in body weight or BMI. Future studies with larger sample sizes investigating the changing trends of CRP, 8-oxodG, and TAC are warranted., Clinical Trial Register: NCT03375866., Competing Interests: Declaration of Competing Interest The authors disclose no conflict of interest., (Copyright © 2023 The Author(s). Published by Elsevier B.V. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
16. Lipoprotein(a) and coronary artery calcium in comparison with other lipid biomarkers: The multi-ethnic study of atherosclerosis.
- Author
-
Jackson CL, Garg PK, Guan W, Tsai MY, Criqui MH, Tsimikas S, and Bhatia HS
- Subjects
- Humans, Biomarkers, Calcium, Cholesterol, Cholesterol, HDL, Cholesterol, LDL, Cohort Studies, Lipoprotein(a), Risk Factors, Atherosclerosis etiology, Coronary Artery Disease complications, Vascular Calcification
- Abstract
Background: Coronary artery calcium (CAC) scoring is often used for atherosclerotic cardiovascular disease (ASCVD) risk stratification in individuals with elevated lipoprotein(a) [Lp(a)]., Objective: To evaluate associations between Lp(a) and baseline CAC (volume/density) and CAC progression compared to other lipid biomarkers., Methods: We utilized data from the Multi-Ethnic Study of Atherosclerosis (MESA), a cohort study of individuals without clinical ASCVD, excluding statin users. We evaluated the associations between Lp(a), low-density lipoprotein-cholesterol (LDL-C), high-density lipoprotein-cholesterol (HDL-C), triglycerides, total cholesterol, apolipoprotein B, and non-HDL-C with baseline CAC and annual CAC progression using multivariable ordinal regression with adjustment for ASCVD risk factors. Analyses were also stratified by median age., Results: In 5,597 participants (2,726 at median 9.5-year follow-up), Lp(a) was not associated with baseline CAC volume or density and was modestly associated with volume progression (OR 1.11, 95% CI 1.03-1.21). However, other biomarkers were positively associated with baseline volume and volume progression (LDL-C: OR 1.26, 95% CI: 1.19-1.33 and OR 1.22, 95% CI: 1.15-1.30, respectively), except HDL-C which was inversely associated. LDL-C, total cholesterol and non-HDL-C were inversely associated with baseline density. In participants <62 years of age, Lp(a) was modestly associated with baseline CAC volume (OR 1.10, 95% CI: 1.00-1.20) and volume progression (OR 1.16 95% CI: 1.04-1.30)., Conclusions: In contrast to other lipid biomarkers, Lp(a) was not associated with baseline CAC volume or density and was only modestly associated with volume progression. Our findings suggest that Lp(a) is not as robustly associated with CAC as other lipid biomarkers., (Copyright © 2023. Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
17. High immunoglobulin-M levels to oxidation-specific epitopes are associated with lower risk of acute myocardial infarction.
- Author
-
Taleb A, Willeit P, Amir S, Perkmann T, Kozma MO, Watzenböck ML, Binder CJ, Witztum JL, and Tsimikas S
- Subjects
- Male, Infant, Newborn, Humans, Epitopes, Phosphorylcholine, Autoantibodies, Immunoglobulin M, Apolipoproteins, Lipoproteins, LDL, Antigen-Antibody Complex, Myocardial Infarction
- Abstract
Immunoglobulin M (IgM) autoantibodies to oxidation-specific epitopes (OSEs) can be present at birth and protect against atherosclerosis in experimental models. This study sought to determine whether high titers of IgM titers to OSE (IgM OSE) are associated with a lower risk of acute myocardial infarction (AMI) in humans. IgM to malondialdehyde (MDA)-LDL, phosphocholine-modified BSA, IgM apolipoprotein B100-immune complexes, and a peptide mimotope of MDA were measured within 24 h of first AMI in 4,559 patients and 4,617 age- and sex-matched controls in the Pakistan Risk of Myocardial Infarction Study. Multivariate-adjusted logistic regression was used to estimate odds ratio (OR) and 95% confidence interval for AMI. All four IgM OSEs were lower in AMI versus controls (P < 0.001 for all). Males, smokers and individuals with hypertension and diabetes had lower levels of all four IgM OSE than unaffected individuals (P < 0.001 for all). Compared to the lowest quintile, the highest quintiles of IgM MDA-LDL, phosphocholine-modified BSA, IgM apolipoprotein B100-immune complexes, and MDA mimotope P1 had a lower OR of AMI: OR (95% confidence interval) of 0.67 (0.58-0.77), 0.64 (0.56-0.73), 0.70 (0.61-0.80) and 0.72 (0.62-0.82) (P < 0.001 for all), respectively. Upon the addition of IgM OSE to conventional risk factors, the C-statistic improved by 0.0062 (0.0028-0.0095) and net reclassification by 15.5% (11.4-19.6). These findings demonstrate that IgM OSE provides clinically meaningful information and supports the hypothesis that higher levels of IgM OSE may be protective against AMI., Competing Interests: Conflict of interest S. T and J. L. W. are co-inventors and receive royalties from patents owned by University of California San Diego (UCSD) and are co-founders and have an equity interest in Oxitope, Pharmaceuticals and Kleanthi Diagnostics. Although these relationships have been identified for conflict-of-interest management based on the overall scope of the project, the research findings included in this publication may not necessarily relate to the interests of the above companies. The terms of this arrangement have been reviewed and approved by the University of California, San Diego in accordance with its conflict-of-interest policies. S. T. has a dual appointment at UCSD and Ionis Pharmaceuticals. J. L. W. is a consultant to Ionis Pharmaceuticals. The other co-authors have nothing to disclose., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
18. APOC3 inhibition with volanesorsen reduces hepatic steatosis in patients with severe hypertriglyceridemia.
- Author
-
Prohaska TA, Alexander VJ, Karwatowska-Prokopczuk E, Tami J, Xia S, Witztum JL, and Tsimikas S
- Subjects
- Humans, Apolipoprotein C-III, Triglycerides, Oligonucleotides pharmacology, Oligonucleotides therapeutic use, Hypertriglyceridemia complications, Hypertriglyceridemia drug therapy, Hypertriglyceridemia chemically induced
- Abstract
ApoC-III inhibits lipoprotein lipase and hepatic uptake of triglyceride-rich lipoproteins. It is unknown whether targeting apoC-III affects hepatic steatosis in patients with hypertriglyceridemia. We studied the effect of volanesorsen, a potent antisense oligonucleotide targeting APOC3 mRNA, on hepatic fat fraction (HFF) assessed by MRI in patients with severe hypertriglyceridemia (SHTG, triglycerides ≥500 mg/dL), familial partial lipodystrophy (FPL, triglycerides ≥200 mg/dL) and familial chylomicronemia syndrome (FCS, triglycerides ≥750 mg/dL). The data were evaluated individually in COMPASS (SHTG), APPROACH (FCS), and BROADEN (FPL) trials. The baseline absolute HFF were elevated in all three trials and ranged from 6.3-18.1%. In COMPASS, compared to placebo, volanesorsen significantly reduced the absolute HFF by -3.02% (95% CI, (-5.60, -0.60), p = 0.009) (placebo-adjusted % change from baseline -24.2%, p = 0.034) from baseline to 6 months. In APPROACH a non-significant absolute -1.0% (95% CI, -2.9, 0.0, p = 0.13) reduction in HFF was noted from baseline to 12 months (placebo-adjusted % change from baseline -37.1%, p = 0.20). In BROADEN volanesorsen significantly reduced the absolute HFF by -8.34% (95% CI, -13.01, -3.67, p = 0.001) from baseline to 12 months (placebo-adjusted % change from baseline -52.7%, p = 0.004). In all 3 trials individually, a strong inverse correlation was present between the baseline HFF and the change in HFF in the volanesorsen groups, but not in the placebo groups. In conclusion, apoC-III inhibition with volanesorsen has favorable effects in HFF in patients with different etiologies of hypertriglyceridemia., (Copyright © 2023. Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
19. Volanesorsen and triglyceride levels in familial chylomicronemia syndrome: Long-term efficacy and safety data from patients in an open-label extension trial.
- Author
-
Witztum JL, Gaudet D, Arca M, Jones A, Soran H, Gouni-Berthold I, Stroes ESG, Alexander VJ, Jones R, Watts L, Xia S, and Tsimikas S
- Subjects
- Humans, Oligonucleotides adverse effects, Apolipoprotein C-III, Triglycerides, Hyperlipoproteinemia Type I drug therapy, Hyperlipoproteinemia Type I genetics
- Abstract
Background: Familial chylomicronemia syndrome (FCS) is a rare, autosomal recessive genetic disorder characterized by a marked increase in plasma triglyceride (TG) levels and recurrent episodes of pancreatitis. The response to conventional TG-lowering therapies is suboptimal. Volanesorsen, an antisense oligonucleotide that targets hepatic apoC-III mRNA, has been shown to significantly reduce TGs in patients with FCS., Objective: To further evaluate the safety and efficacy of extended treatment with volanesorsen in patients with FCS., Methods: This phase 3 open-label extension study evaluated the efficacy and safety of extended treatment with volanesorsen in three groups of patients with FCS: Those who had previously received volanesorsen or placebo in the APPROACH and COMPASS studies, and treatment-naive patients not participating in either study. Key endpoints included change in fasting TG and other lipid measurements, and safety over 52 weeks., Results: Volanesorsen treatment resulted in sustained reductions in plasma TG levels in previously treated patients from the APPROACH and COMPASS studies. Volanesorsen-treated patients from the three populations studied had mean decreases in fasting plasma TGs from index study baseline to months 3, 6, 12 and 24 as follows: decreases of 48%, 55%, 50%, and 50%, respectively (APPROACH); decreases of 65%, 43%, 42%, and 66%, respectively (COMPASS); and decreases of 60%, 51%, 47%, and 46%, respectively (treatment-naive). Common adverse events were injection site reactions and platelet count decrease, consistent with previous studies., Conclusion: Extended open-label treatment with volanesorsen in patients with FCS resulted in sustained reductions of plasma TG levels and safety consistent with the index studies., Competing Interests: Declaration of Competing Interest JLW is a consultant to Ionis Pharmaceuticals and ST is an employee of Ionis Pharmaceuticals. JLW and ST are co-inventors and receive royalties from patents owned by the University of California, San Diego on oxidation-specific antibodies and of biomarkers related to oxidized lipoproteins. They are co-founders and have an equity interest in Oxitope, Inc and its affiliates (“Oxitope”) as well as in Kleanthi Diagnostics, LLC (“Kleanthi”). Although these relationships have been identified for conflict of interest management based on the overall scope of the project and its potential benefit to Oxitope and Kleanthi, the research findings included in this particular publication may not necessarily relate to the interests of Oxitope and Kleanthi. The terms of this arrangement have been reviewed and approved by the University of California, San Diego in accordance with its conflict of interest policies. DG reports grants and personal fees from Ionis Pharmaceuticals during the conduct of the study; grants and personal fees from Allergan, Amgen, Amryt, Arrowhead, Eli Lilly, Novartis, NovoNordisk, Regeneron, and Sanofi outside the submitted work; grants from Acasti, Aegerion, Applied Therapeutics, AstraZeneca, Boehringer-Ingelheim, Ceapro, Dalcor, Esperion, Kowa, The Medicine Company, and Uniqure outside the submitted work; and personal fees from CRISPR Therapeutics, Saliogen, and Verve Therapeutics outside the submitted work. MA has received research grant support and lecturing fees from Alfasigma, Amgen, Amryt, Daiichi Sankyo, Ionis/Akcea, Novartis, Pfizer, Regeneron, and Sanofi. AJ has provided services to the following companies: Akcea, Amgen, Sanofi, and SOBI. HS received research and education grants and honoraria from Akcea Therapeutics, Alexion, Amryt, Synageva, Kowa, MSD, NAPP, Novartis, Pfizer, Sanofi, Synageva, and Takeda. IG-B has received personal honoraria from Aegerion, Akcea, Amarin, Amgen, Daiichi Sankyo, Novartis, Regeneron, and Sanofi; and nonfinancial support from Akcea, Amgen, and Sanofi. ESGS has received advisory board/lecturing fees, paid to institution, from: Amgen, Sanofi, NovoNordisk, AstraZeneca, Esperion, Daiichi-Sankyo, and Ionis/Akcea. No patents/stocks. VJA, LW, and SX are employees of Ionis Pharmaceuticals. RJ was an employee of Akcea Therapeutics at the time of the study and manuscript development and may own stock in Ionis Pharmaceuticals, parent company of Akcea Therapeutics., (Copyright © 2023 National Lipid Association. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
20. Efficacy and safety of pelacarsen in lowering Lp(a) in healthy Japanese subjects.
- Author
-
Karwatowska-Prokopczuk E, Lesogor A, Yan JH, Hurh E, Hoenlinger A, Margolskee A, Xia S, and Tsimikas S
- Subjects
- Humans, Oligonucleotides, Antisense, Oligonucleotides, Healthy Volunteers, Double-Blind Method, Dose-Response Relationship, Drug, East Asian People, Lipoprotein(a)
- Abstract
Background: Pelacarsen is a liver-targeted antisense oligonucleotide that potently lowers lipoprotein(a) [Lp(a)] levels. Its safety and efficacy in diverse populations has not been extensively studied., Objective: To assess the effect of pelacarsen, including monthly dosing of 80 mg, in subjects of Japanese ancestry., Methods: A randomized double-blind, placebo-controlled, study was performed in 29 healthy Japanese subjects treated with single ascending doses (SAD) of pelacarsen 20, 40 and 80 mg subcutaneously or multiple doses (MD) of pelacarsen 80 mg monthly for 4 doses. The primary objective was to assess the safety and tolerability in healthy Japanese subjects; secondary objectives to assess the pharmacokinetics of pelacarsen; and exploratory objective to determine the effect of pelacarsen on plasma Lp(a) levels., Results: No serious adverse events or clinically relevant abnormalities in any laboratory parameters were noted. In the MD cohort, mean plasma concentrations of pelacarsen peaked at ∼4 hours and declined in a bi-exponential manner thereafter. In the SAD cohorts, the placebo-corrected least-square mean (PCLSM) percent changes in Lp(a) at Day 30 were: -55.4% (p=0.0008), -58.9% (p=0.0003) and -73.7% (p<0.0001) for the 20 mg, 40 mg, and 80 mg pelacarsen-treated groups, respectively. In the MD cohort, the PCLSM at Days 29, 85, 113, 176 and 204 were -84.0% (p=0.0003), -106.2% (p<0.0001), -70.0 (p<0.0001), -80.0% (p=0.0104) and -55.8% (p=0.0707), respectively., Conclusions: Pelacarsen demonstrates an acceptable safety and tolerability profile and potently lowers plasma levels of Lp(a) in healthy Japanese subjects, including with the 80 mg monthly dose being evaluated in the Lp(a) HORIZON trial., (Copyright © 2022. Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
21. Assessment of efficacy and safety of volanesorsen for treatment of metabolic complications in patients with familial partial lipodystrophy: Results of the BROADEN study: Volanesorsen in FPLD; The BROADEN Study.
- Author
-
Oral EA, Garg A, Tami J, Huang EA, O'Dea LSL, Schmidt H, Tiulpakov A, Mertens A, Alexander VJ, Watts L, Hurh E, Witztum JL, Geary RS, and Tsimikas S
- Subjects
- Female, Humans, Male, Apolipoprotein C-III, Glycated Hemoglobin, Triglycerides, Lipodystrophy, Familial Partial
- Abstract
Background: Volanesorsen, an antisense oligonucleotide, is designed to inhibit hepatic apolipoprotein C-III synthesis and reduce plasma apolipoprotein C-III and triglyceride concentrations., Objective: The present study assessed efficacy and safety of volanesorsen in patients with familial partial lipodystrophy (FPLD) and concomitant hypertriglyceridemia and diabetes., Methods: BROADEN was a randomized, placebo-controlled, phase 2/3, 52-week study with open-label extension and post-treatment follow-up periods. Patients received weekly subcutaneous volanesorsen 300 mg or placebo. The primary endpoint was percent change from baseline in fasting triglycerides at 3 months. Secondary endpoints included relative percent change in hepatic fat fraction (HFF), visceral adiposity, and glycated hemoglobin levels., Results: Forty patients (11 men, 29 women) were enrolled, majority of whom were aged <65 years (mean, 47 years) and White. Least squares mean (LSM) percent change in triglycerides from baseline to 3 months was -88% (95% CI, -134 to -43) in the volanesorsen group versus -22% (95% CI, -61 to 18) in the placebo group, with a difference in LSM of -67% (95% CI, -104 to -30; P=0.0009). Volanesorsen induced a significant LSM relative reduction in HFF of 53% at month 12 versus placebo (observed mean [SD]: 9.7 [7.65] vs. 18.0 [8.89]; P=0.0039). No statistically significant changes were noted in body volume measurements (fat, liver, spleen, visceral/subcutaneous adipose tissue) or glycated hemoglobin. Serious adverse events in patients assigned to volanesorsen included 1 case each of sarcoidosis, anaphylactic reaction, and systemic inflammatory response syndrome., Conclusion: In BROADEN, volanesorsen significantly reduced serum triglyceride levels and hepatic steatosis in patients with FPLD., Competing Interests: Declarations of Interest E.A.O. has received grant support and worked as a scientific advisor to Ionis Pharmaceuticals and Akcea Therapeutics, has received grant support and provided scientific advising for Regeneron Pharmaceuticals and Aegerion Pharmaceuticals (now owned by Amryt), has received grant support from Gemphire Therapeutics, is actively receiving grant support from GI Dynamics, Rhythm Pharmaceuticals, Fractyl, Regeneron Pharmaceuticals, and Amryt Pharmaceuticals, has provided consulting for Third Rock Ventures and Rejuvenate, and holds a joint patent with Amgen entitled “use of leptin for treating human lipoatrophy and a method of determining predisposition to said treatment” (now owned by Amryt) and is entitled to royalty fees. A.G. has been contracted by Aegerion Pharmaceuticals, Amryt Pharmaceuticals, Ionis Pharmaceuticals, Pfizer, Quintiles, and Regeneron Pharmaceuticals, has received consulting fees from Cello Health, Health Advances, Intarcia Therapeutics, Kyttaro, and Third Rock Ventures, has received honoraria from Aegerion (metreleptin) for a roundtable meeting and from Regeneron for a steering committee and an advisory board, has served as investigator for Intercept Pharmaceuticals, and holds a joint patent with Amgen entitled “use of leptin for treating human lipoatrophy and a method of determining predisposition to said treatment” but receives no financial compensation. J.T., V.J.A., L.W., R.S.G., and S.T. are employees of Ionis Pharmaceuticals Inc. E.A.H. and L.S.L.O. were employees of Akcea Therapeutics Inc. H.S. was a study investigator in the study. A.T. was a principal investigator in the study. A.M. declaration of interest: none. E.H. is an employee of Akcea Therapeutics Inc. J.L.W. and S.T. are coinventors and receive royalties from patents owned by University of California San Diego (UCSD) on oxidation-specific antibodies and of biomarkers related to oxidized lipoproteins, are cofounders and have equity interest in Oxitope and its affiliates (“Oxitope”) and Kleanthi Diagnostics. J.L.W. has received consulting fees from Ionis Pharmaceuticals. S.T. has a dual appointment at Ionis Pharmaceuticals and UCSD, is cofounder of Covicept Therapeutics, consultant to imaware, and has received research funding to UCSD from Novartis. All authors have approved the final article., (Copyright © 2022. Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
22. Effect of olezarsen targeting APOC-III on lipoprotein size and particle number measured by NMR in patients with hypertriglyceridemia.
- Author
-
Karwatowska-Prokopczuk E, Tardif JC, Gaudet D, Ballantyne CM, Shapiro MD, Moriarty PM, Baum SJ, Amour ES, Alexander VJ, Xia S, Otvos JD, Witztum JL, and Tsimikas S
- Subjects
- Humans, Apolipoprotein C-III, Triglycerides, Lipoproteins, Particle Size, Hypertriglyceridemia drug therapy, Hyperlipidemias
- Abstract
Background: Olezarsen is a hepatocyte-targeted, GalNAc-modified antisense oligonucleotide that decreases plasma levels of apolipoprotein C-III (apoC-III) and triglyceride-rich lipoproteins (TRLs)., Objective: To define the effect of olezarsen on NMR-derived lipoprotein particle size and concentration., Methods: Patients (n=114) with or at risk for atherosclerotic cardiovascular disease and fasting triglycerides ≥200 and <500 mg/dL received olezarsen (10 or 50 mg every 4 weeks, 15 mg every 2 weeks, or 10 mg every week) or saline placebo subcutaneously for 6-12 months. NMR LipoProfile® analysis was performed in frozen EDTA plasma samples collected at baseline and at the primary analysis timepoint (PAT) at 6 months., Results: A dose-dependent relationship was generally noted with increasing cumulative doses of olezarsen in TRL particle (TRLP), LDL particle (LDL-P) and HDL (HDL-P) particle concentrations. In the 50 mg every 4 weeks dose, compared to placebo, olezarsen resulted in a significant reduction in total TRL-P (51%, P<0.0001) with largest reductions in large-size (68%, P<0.0001) and medium-size (63%, P<0.0001) TRL-P. Total LDL-P concentration was not changed, but large LDL-P increased by 186% (p=0.0034), and small LDL-P decreased by 39% (p=0.0713). Total HDL-P concentration increased by 15% (P=0.0006), driven primarily by a 32% increase in small HDL subspecies (diameters <8.3 nm) (P=0.0008)., Conclusion: Olezarsen results in favorable changes in lipoprotein concentration and particle size, primarily manifested by reduction in TRLs, remodeling to larger LDL particles, and increase in small HDL-P. These findings suggest that apoC-III inhibition improves the overall atherogenic risk profile., Competing Interests: Disclosures EKP, VJB, SX, and ST are employees of Ionis Pharmaceuticals. J-C T received research grants from Amarin, AstraZeneca, DalCor, Esperion, Ionis, Pfizer and Servier; honoraria from Amarin, DalCor, Pfizer, Sanofi and Servier; minor equity interest in DalCor. DG reports grants and personal fees from Akcea Therapeutics, Ionis Pharmaceuticals during the conduct of the study, Arrowhead and Regeneron, and grants from Kowa and Acasti and grants from Uniqure outside the submitted work. CMB has received grant/research support (to his institution) from Abbott Diagnostic, Akcea, Amgen, Esperion, Ionis, Novartis, Regeneron, and Roche Diagnostic, and has been a consultant for Abbott Diagnostics, Althera, Amarin, Amgen, Arrowhead, AstraZeneca, Corvidia, Denka Seiken, Esperion, Genentech, Gilead, Matinas BioPharma Inc, New Amsterdam, Novartis, Novo Nordisk, Pfizer, Regeneron, Roche Diagnostic, and Sanofi-Synthelabo. MDS serves on scientific advisory boards: Amgen, Esperion, Novartis. PMM is a consultant, speaker, or received Research grants from Amgen, Esperion, Kaneka, Amarin, Stage II Innovations/Renew, Novartis, Ionis, FH Foundation, GB Life Sciences, Aegerion. SJB is consultant on scientific advisory board or speaker for Altimmune, Akcea, Amgen, AstraZeneca, Boehringer Ingelheim, Axcella, Eli Lilly, Esperion, Madrigal, Novartis, Regeneron, Sanofi. JDO is an employee of LabCorp. JLW is a consultant to Ionis. JLW and ST are co-inventors and receive royalties from patents owned by UCSD on oxidation-specific antibodies and of biomarkers related to oxidized lipoproteins and are co-founders and have an equity interest in Oxitope, Inc and its affiliates (“Oxitope”) as well as in Kleanthi Diagnostics, LLC (“Kleanthi”). ST is a co-founder of Covicept Therapeutics. Although these relationships have been identified for conflict of interest management based on the overall scope of the project and its potential benefit to Oxitope and Kleanthi, the research findings included in this particular publication may not necessarily relate to the interests of Oxitope and Kleanthi. The terms of this arrangement have been reviewed and approved by the University of California, San Diego in accordance with its conflict of interest policies. The other authors have no disclosures. All authors have approved the final article should be true and included in the disclosure., (Copyright © 2022. Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
23. Oxidized phospholipids on apolipoprotein B-100 versus plasminogen and risk of coronary heart disease in the PROCARDIS study.
- Author
-
Clarke R, Hammami I, Sherliker P, Valdes-Marquez E, Watkins H, Hill M, Yang X, Tsimikas S, and Hopewell JC
- Subjects
- Apolipoprotein B-100, Apolipoproteins B, Humans, Lipoprotein(a), Oxidation-Reduction, Plasminogen, Coronary Disease diagnosis, Coronary Disease epidemiology, Phospholipids
- Abstract
Background and Aims: Oxidized phospholipids carried on the apolipoprotein B-100 (OxPL-apoB) component of Lp(a) are predictive of coronary heart disease (CHD), but the role of oxidized phospholipids carried on plasminogen (OxPL-PLG) is unknown. We examined the independent effects of OxPL-apoB and OxPL-PLG for risk of CHD before and after adjustment for Lp(a)., Methods: Plasma levels of OxPL-apoB, OxPL-PLG, plasminogen and Lp(a) were measured in the PROCARDIS study of early-onset CHD (906 cases/858 controls). Multivariable logistic regression was used to estimate the odds ratios (OR) for each biomarker with CHD after adjustment for established risk factors., Results: Mean levels of OxPL-apoB were higher in cases than controls, but levels of OxPL-PLG and plasminogen were similar. For OxPL-apoB, individuals in the top vs bottom fifth had 2-fold higher age and sex-adjusted OR of CHD (OR = 2.61 [95%CI: 1.91, 3.55]), which were partially attenuated after adjustment for established risk factors. The findings for OxPL-apoB and CHD in PROCARDIS were comparable with those of a meta-analysis of all such studies. However, the associations of OxPL-apoB with CHD were fully attenuated by additional adjustment for Lp(a) (OR = 0.93 [0.54,1.60]). Neither OxPL-PLG nor plasminogen were associated with CHD. Overall, there were no differences in the predictive value for CHD of high vs normal levels (<20th or >80th percentile) of OxPL-apoB, OxPL-PLG, plasminogen or Lp(a) after stratifying for each other., Conclusions: These results highlight the context-dependency of OxPL in plasma and suggest that their associated risk of CHD is chiefly mediated by their carriage on Lp(a)., (Crown Copyright © 2022. Published by Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
24. Development and validation of an isoform-independent monoclonal antibody-based ELISA for measurement of lipoprotein(a).
- Author
-
Marcovina SM, Navabi N, Allen S, Gonen A, Witztum JL, and Tsimikas S
- Subjects
- Apolipoproteins A, Apoprotein(a), Chromatography, Liquid, Enzyme-Linked Immunosorbent Assay, Epitopes, Humans, Protein Isoforms, Tandem Mass Spectrometry, Antibodies, Monoclonal, Lipoprotein(a)
- Abstract
The study aims were to develop a new isoform-independent enzyme-linked immunoassay (ELISA) for the measurement of lipoprotein(a) [Lp(a)], validate its performance characteristics, and demonstrate its accuracy by comparison with the gold-standard ELISA method and an LC-MS/MS candidate reference method, both developed at the University of Washington. The principle of the new assay is the capture of Lp(a) with monoclonal antibody LPA4 primarily directed to an epitope in apolipoprotein(a) KIV
2 and its detection with monoclonal antibody LPA-KIV9 directed to a single antigenic site present on KIV9 . Validation studies were performed following the guidelines of the Clinical Laboratory Improvement Amendments and the College of American Pathologists. The analytical measuring range of the LPA4/LPA-KIV9 ELISA is 0.27-1,402 nmol/L, and the method meets stringent criteria for precision, linearity, spike and recovery, dilutability, comparison of plasma versus serum, and accuracy. Method comparison with both the gold-standard ELISA and the LC-MS/MS method performed in 64 samples with known apolipoprotein(a) isoforms resulted in excellent correlation with both methods (r=0.987 and r=0.976, respectively). Additionally, the variation in apolipoprotein(a) size accounted for only 0.2% and 2.2% of the bias variation, respectively, indicating that the LPA4/LPA-KIV9 ELISA is not affected by apolipoprotein(a) size polymorphism. Peptide mapping and competition experiments demonstrated that the measuring monoclonal antibodies used in the gold-standard ELISA (a-40) and in the newly developed ELISA (LPA-KIV9) are directed to the same epitope,4076 LETPTVV4082 , on KIV9 . In conclusion, no statistically or clinically significant bias was observed between Lp(a) measurements obtained by the LPA4/LPA-KIV9 ELISA and those obtained by the gold-standard ELISA or LC-MS/MS, and therefore, the methods are considered equivalent., Competing Interests: Conflict of interest S. M. M., N. N., and S. A. are employees of Medpace Reference Laboratories. S. M. reports consulting roles for Roche, Denka, Novartis, and research. A. G., J. L. W., and S. T. are coinventors of monoclonal antibodies directed to Lp(a) owned by UCSD directed to Lp(a). J. L. W. and S. T. receive royalties from patents on oxidation-specific antibodies and of biomarkers related to oxidized lipoproteins and Lp(a) held by UCSD. S. T. and J. L. W. are cofounders and have an equity interest in Oxitope, Inc and its affiliates (“Oxitope”) as well as in Kleanthi Diagnostics, LLC (“Kleanthi”). The terms of this arrangement have been reviewed and approved by the University of California, San Diego, in accordance with its conflict-of-interest policies. S. T. has a dual appointment at UCSD and Ionis Pharmaceuticals. J. L. W. is a consultant to Ionis Pharmaceuticals and A. G. to Kleanthi Diagnostics. All other authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
25. Trends in testing and prevalence of elevated Lp(a) among patients with aortic valve stenosis.
- Author
-
Bhatia HS, Ma GS, Taleb A, Wilkinson M, Kahn AM, Cotter B, Yeang C, DeMaria AN, Patel MP, Mahmud E, Reeves RR, and Tsimikas S
- Subjects
- Aortic Valve surgery, Humans, Lipoprotein(a), Prevalence, Risk Factors, Treatment Outcome, Aortic Valve Stenosis diagnosis, Aortic Valve Stenosis epidemiology, Aortic Valve Stenosis surgery, Heart Valve Prosthesis Implantation methods
- Abstract
Background and Aims: Lipoprotein(a) [Lp(a)] is causally associated with aortic valve stenosis (AS) but Lp(a) testing among AS patients is not broadly incorporated into clinical practice. We evaluated trends in Lp(a) testing in an academic medical center., Methods: Educational efforts and adding Lp(a) to the lipid panel on the electronic medical record (EMR) and pre-procedure order sets were used to increase awareness of Lp(a) as a risk factor in AS. Medical records at University of California San Diego Health (UCSDH) were analyzed from 2010 to 2020 to define the yearly frequency of first time Lp(a) testing in patients with diagnosis codes for AS or undergoing transcatheter aortic valve replacement (TAVR)., Results: Lp(a) testing for any indication increased over 5-fold from 2010 to 2020. A total of 3808 patients had a diagnosis of AS and 417 patients had TAVR. Lp(a) levels >30 mg/dL were present in 37% of AS and 35% of TAVR patients. The rates of Lp(a) testing in AS and TAVR were 14.0% and 65.7%, respectively. In AS, Lp(a) testing increased over time from 8.5% in 2010, peaking at 24.2% in 2017, and declining to 13.9% in 2020 (p < 0.001 for trend). Following implementation of EMR order-sets in 2016, Lp(a) testing in TAVR cases increased to a peak of 88.5% in 2018., Conclusions: Elevated Lp(a) is prevalent in AS and TAVR patients. Implementation of educational efforts and practice pathways resulted in increased Lp(a) testing in patients with AS. This study represents a paradigm that may allow increased global awareness of Lp(a) as a risk factor for AS., (Copyright © 2022 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
26. Generation of cardio-protective antibodies after pneumococcal polysaccharide vaccine: Early results from a randomised controlled trial.
- Author
-
Ren S, Hansbro PM, Srikusalanukul W, Horvat JC, Hunter T, Brown AC, Peel R, Faulkner J, Evans TJ, Li SC, Newby D, Hure A, Abhayaratna WP, Tsimikas S, Gonen A, Witztum JL, and Attia J
- Subjects
- Australia, Biomarkers, Humans, Immunoglobulin G, Immunoglobulin M, Streptococcus pneumoniae, Atherosclerosis prevention & control, Pneumococcal Vaccines
- Abstract
Background and Aims: Observational studies have demonstrated that the pneumococcal polysaccharide vaccine (PPV) is associated with reduced risk of cardiovascular events. This may be mediated through IgM antibodies to OxLDL, which have previously been associated with cardioprotective effects. The Australian Study for the Prevention through Immunisation of Cardiovascular Events (AUSPICE) is a double-blind, randomised controlled trial (RCT) of PPV in preventing ischaemic events. Participants received PPV or placebo once at baseline and are being followed-up for incident fatal and non-fatal myocardial infarction or stroke over 6 years., Methods: A subgroup of participants at one centre (Canberra; n = 1,001) were evaluated at 1 month and 2 years post immunisation for changes in surrogate markers of atherosclerosis, as pre-specified secondary outcomes: high-sensitive C-reactive protein (CRP), pulse wave velocity (PWV), and carotid intima-media thickness (CIMT). In addition, 100 participants were randomly selected in each of the intervention and control groups for measurement of anti-pneumococcal antibodies (IgG, IgG2, IgM) as well as anti-OxLDL antibodies (IgG and IgM to CuOxLDL, MDA-LDL, and PC-KLH)., Results: Concentrations of anti-pneumococcal IgG and IgG2 increased and remained high at 2 years in the PPV group compared to the placebo group, while IgM increased and then declined, but remained detectable, at 2 years. There were statistically significant increases in all anti-OxLDL IgM antibodies at 1 month, which were no longer detectable at 2 years; there was no increase in anti-OxLDL IgG antibodies. There were no significant changes in CRP, PWV or CIMT between the treatment groups at the 2-year follow-up., Conclusions: PPV engenders a long-lasting increase in anti-pneumococcal IgG, and to a lesser extent, IgM titres, as well as a transient increase in anti-OxLDL IgM antibodies. However, there were no detectable changes in surrogate markers of atherosclerosis at the 2-year follow-up. Long-term, prospective follow-up of clinical outcomes is continuing to assess if PPV reduces CVD events., (Copyright © 2022 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
27. Lipoprotein(a), venous thromboembolism and COVID-19: A pilot study.
- Author
-
Nurmohamed NS, Collard D, Reeskamp LF, Kaiser Y, Kroon J, Tromp TR, van den Born BH, Coppens M, Vlaar APJ, Beudel M, van de Beek D, van Es N, Moriarty PM, Tsimikas S, and Stroes ESG
- Subjects
- Humans, Lipoprotein(a), Pilot Projects, Risk Factors, SARS-CoV-2, COVID-19, Venous Thromboembolism diagnosis, Venous Thromboembolism epidemiology
- Abstract
Background and Aims: Thrombosis is a major driver of adverse outcome and mortality in patients with Coronavirus disease 2019 (COVID-19). Hypercoagulability may be related to the cytokine storm associated with COVID-19, which is mainly driven by interleukin (IL)-6. Plasma lipoprotein(a) [Lp(a)] levels increase following IL-6 upregulation and Lp(a) has anti-fibrinolytic properties. This study investigated whether Lp(a) elevation may contribute to the pro-thrombotic state hallmarking COVID-19 patients., Methods: Lp(a), IL-6 and C-reactive protein (CRP) levels were measured in 219 hospitalized patients with COVID-19 and analyzed with linear mixed effects model. The baseline biomarkers and increases during admission were related to venous thromboembolism (VTE) incidence and clinical outcomes in a Kaplan-Meier and logistic regression analysis., Results: Lp(a) levels increased significantly by a mean of 16.9 mg/dl in patients with COVID-19 during the first 21 days after admission. Serial Lp(a) measurements were available in 146 patients. In the top tertile of Lp(a) increase, 56.2% of COVID-19 patients experienced a VTE event compared to 18.4% in the lowest tertile (RR 3.06, 95% CI 1.61-5.81; p < 0.001). This association remained significant after adjusting for age, sex, IL-6 and CRP increase and number of measurements. Increases in IL-6 and CRP were not associated with VTE. Increase in Lp(a) was strongly correlated with increase in IL-6 (r = 0.44, 95% CI 0.30-0.56, p < 0.001)., Conclusions: Increases in Lp(a) levels during the acute phase of COVID-19 were strongly associated with VTE incidence. The acute increase in anti-fibrinolytic Lp(a) may tilt the balance to VTE in patients hospitalized for COVID-19., (Copyright © 2021 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
28. Always Present, But Now Rediscovered: Lp(a) as a Predictor of Long-Term Outcomes in PCI.
- Author
-
Tsimikas S, Reeves RR, and Patel MP
- Subjects
- Humans, Risk Factors, Treatment Outcome, Coronary Artery Disease diagnostic imaging, Coronary Artery Disease therapy, Percutaneous Coronary Intervention adverse effects
- Abstract
Competing Interests: Funding Support and Author Disclosures Dr Tsimikas is a coinventor and receives royalties from patents owned by UCSD; is a cofounder and has an equity interest in Oxitope and its affiliates Kleanthi Diagnostics and Covicept Therapeutics; and has a dual appointment at UCSD, and Ionis Pharmaceuticals. Although these relationships have been identified for conflict-of-interest management on the basis of the overall scope of the project, the research findings included in this particular publication may not necessarily relate to the interests of the aforementioned companies. The terms of this arrangement have been reviewed and approved by UCSD, in accordance with its conflict-of-interest policies. All other authors have reported that they have no relationships relevant to the contents of this paper to disclose.
- Published
- 2021
- Full Text
- View/download PDF
29. Prevalence and influence of LPA gene variants and isoform size on the Lp(a)-lowering effect of pelacarsen.
- Author
-
Karwatowska-Prokopczuk E, Clouet-Foraison N, Xia S, Viney NJ, Witztum JL, Marcovina SM, and Tsimikas S
- Subjects
- Apoprotein(a) genetics, Humans, Prevalence, Protein Isoforms genetics, Apolipoproteins A, Lipoprotein(a) genetics
- Abstract
Background and Aims: Antisense oligonucleotides (ASOs) targeting LPA to lower lipoprotein(a) [Lp(a]] are in clinical trials. Patients have been recruited according to various Lp(a) thresholds, but the prevalence of LPA genetic variants and their effect on efficacy of these ASOs are not well described., Methods: We analyzed data from 4 clinical trials of the ASO pelacarsen targeting apolipoprotein(a) that included 455 patients. Common LPA genetic variants rs10455872 and rs3798220, major and minor isoform size, and changes in Lp(a), LDL-C, apoB, OxPL-apoB and OxPL-apo(a) were analyzed according to categories of baseline Lp(a)., Results: The prevalence of carrier status for rs10455872 and rs3798220 combined ranged from 25.9% in patients with Lp(a) in the 75 - <125 nmol/L range to 77.1% at Lp(a) ≥375 nmol/L. The prevalence of homozygosity for rs3798220, rs10455872 and for double heterozygosity in category of Lp(a) ≥375 nmol/L was 6.3%, 14.6% and 12.5%, respectively. Isoform size decreased with increasing Lp(a) plasma levels, with 99.3% of patients with Lp(a) ≥175 nmol/L having ≤20 KIV repeats in the major isoform. The mean percent reduction from baseline in Lp(a), OxPL-apoB and OxPL-apo(a) in response to pelacarsen was not affected by the presence of rs10455872 and rs3798220, isoform size or baseline Lp(a) at all doses studied., Conclusions: In patients randomized to Lp(a) lowering trials, LPA genetic variants are common, but a sizable proportion do not carry common variants associated with elevated Lp(a). In contrast, the major isoform size was almost uniformly ≤20 KIV repeats in patients with Lp(a) ≥175 nmol/L. The Lp(a) and OxPL lowering effects of pelacarsen were independent of both LPA genetic variants and isoform size., (Copyright © 2021 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
30. Effect of bariatric surgery on plasma levels of oxidised phospholipids, biomarkers of oxidised LDL and lipoprotein(a).
- Author
-
Ho JH, Adam S, Liu Y, Azmi S, Dhage S, Syed AA, Ammori BJ, Donn R, Heald A, Gibson MJ, Malik RA, Yang X, Durrington PN, Tsimikas S, and Soran H
- Subjects
- Humans, Male, Female, Adult, Middle Aged, Prospective Studies, Oxidation-Reduction, Obesity, Morbid surgery, Obesity, Morbid blood, Lipoproteins, LDL blood, Bariatric Surgery, Lipoprotein(a) blood, Biomarkers blood, Phospholipids blood
- Abstract
Background: Obesity is associated with adverse cardiovascular outcomes and this is improved following bariatric surgery. Oxidised phospholipids (OxPL) are thought to reflect the pro-inflammatory effects of lipoprotein(a) [Lp(a)], and both are independent predictors of cardiovascular disease., Objective: Our study sought to determine the impact of bariatric surgery on OxPL, biomarkers of oxidised LDL (OxLDL) and Lp(a)., Methods: This is a prospective, observational study of 59 patients with severe obesity undergoing bariatric surgery. Blood samples were obtained prior to surgery and at 6 and 12 months after. Sixteen patients attending the tertiary medical weight management clinic at the same centre were also recruited for comparison. Lipid and metabolic blood parameters, OxLDL, OxPL on apolipoprotein B-100 (OxPL-apoB), IgG and IgM autoantibodies to MDA-LDL, IgG and IgM apoB-immune complexes and Lp(a) were measured., Results: Reduction in body mass index (BMI) was significant following bariatric surgery, from median 48 kg/m
2 at baseline to 37 kg/m2 at 6 months and 33 kg/m2 at 12 months. OxPL-apoB levels decreased significantly at 12 months following surgery [5.0 (3.2-7.4) to 3.8 (3.0-5.5) nM, p = 0.001], while contrastingly, Lp(a) increased significantly [10.2 (3.8-31.9) to 16.9 (4.9-38.6) mg/dl, p = 0.002]. There were significant post-surgical decreases in IgG and IgM biomarkers, particularly at 12 months, while OxLDL remained unchanged., Conclusions: Bariatric surgery results in a significant increase in Lp(a) but reductions in OxPL-apoB and other biomarkers of oxidised lipoproteins, suggesting increased synthetic capacity and reduced oxidative stress. These biomarkers might be clinically useful to monitor physiological effects of weight loss interventions., (Copyright © 2020 National Lipid Association. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
31. Glucose Control in Patients Undergoing PCI: What Is the Optimal HbA 1c for Long-Term Outcomes?
- Author
-
Tsimikas S and Bhatia HS
- Subjects
- Blood Glucose, Glycated Hemoglobin, Humans, Treatment Outcome, Myocardial Infarction, Percutaneous Coronary Intervention adverse effects
- Abstract
Competing Interests: Funding Support and Author Disclosures This work was partially supported by the National Institutes of Health (grant 5T32HL079891), as part of the University of California-San Diego Integrated Cardiovascular Epidemiology Fellowship. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. Dr. Tsimikas has received research support from the Fondation Leducq (grant 16CVD01) and partial support from National Institutes of Health grants HL136275, HL106579, HL108735, HL136098, HL128550, and HL135737; is co-inventor of and has received royalties from patents owned by the University of California-San Diego on oxidation-specific antibodies and of biomarkers related to oxidized lipoproteins; is co-founder of and has an equity interest in Oxitope (and its affiliates [“Oxitope”]) and Kleanthi Diagnostics (“Kleanthi”); and has a dual appointment at the University of California-San Diego and Ionis Pharmaceuticals; the findings included in this particular publication may not necessarily relate to the interests of Oxitope and Kleanthi; the terms of this arrangement have been reviewed and approved by the University of California-San Diego in accordance with its conflict of interest policies. Dr. Bhatia has reported that he has no relationships relevant to the contents of this paper to disclose.
- Published
- 2021
- Full Text
- View/download PDF
32. Novel method for quantification of lipoprotein(a)-cholesterol: implications for improving accuracy of LDL-C measurements.
- Author
-
Yeang C, Witztum JL, and Tsimikas S
- Subjects
- Humans, Male, Female, Oligonucleotides, Antisense, Middle Aged, Adult, Cholesterol, LDL blood, Lipoprotein(a) blood
- Abstract
Current methods for determining "LDL-C" in clinical practice measure the cholesterol content of both LDL and lipoprotein(a) [Lp(a)-C]. We developed a high-throughput, sensitive, and rapid method to quantitate Lp(a)-C and improve the accuracy of LDL-C by subtracting for Lp(a)-C (LDL-C
corr ). Lp(a)-C is determined following isolation of the Lp(a) on magnetic beads linked to monoclonal antibody LPA4 recognizing apolipoprotein(a). This Lp(a)-C assay does not detect cholesterol in plasma samples lacking Lp(a) and is linear up to 747 nM Lp(a). To validate this method clinically over a wide range of Lp(a) (9.0-822.8 nM), Lp(a)-C and LDL-Ccorr were determined in 21 participants receiving an Lp(a)-specific lowering antisense oligonucleotide and in eight participants receiving placebo at baseline, at 13 weeks during peak drug effect, and off drug. In the groups combined, Lp(a)-C ranged from 0.6 to 35.0 mg/dl and correlated with Lp(a) molar concentration (r = 0.76; P < 0.001). However, the percent Lp(a)-C relative to Lp(a) mass varied from 5.8% to 57.3%. Baseline LDL-Ccorr was lower than LDL-C [mean (SD), 102.2 (31.8) vs. 119.2 (32.4) mg/dl; P < 0.001] and did not correlate with Lp(a)-C. It was demonstrated that three commercially available "direct LDL-C" assays also include measures of Lp(a)-C. In conclusion, we have developed a novel and sensitive method to quantitate Lp(a)-C that provides insights into the Lp(a) mass/cholesterol relationship and may be used to more accurately report LDL-C and reassess its role in clinical medicine., Competing Interests: Conflict of interest Drs Tsimikas and Witztum are coinventors and receive royalties from patents owned by UCSD on oxidation-specific antibodies and of biomarkers related to oxidized lipoproteins and are cofounders and have equity interests in Oxitope, Inc. and Kleanthi Diagnostics, LLC. Although these relationships have been identified for conflict of interest management based on the overall scope of the project and its potential benefit to Oxitope and Kleanthi, the research findings included in this particular publication may not necessarily relate to the interests of Oxitope and Kleanthi. The terms of this arrangement have been reviewed and approved by the UCSD in accordance with its conflict of interest policies. Dr Tsimikas has a dual appointment at UCSD and Ionis Pharmaceuticals, and Dr Witztum is a consultant to Ionis Pharmaceuticals. The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
33. Short-term regulation of hematopoiesis by lipoprotein(a) results in the production of pro-inflammatory monocytes.
- Author
-
Schnitzler JG, Poels K, Stiekema LCA, Yeang C, Tsimikas S, Kroon J, Stroes ESG, Lutgens E, and Seijkens TTP
- Subjects
- Animals, Hematopoiesis, Mice, Mice, Inbred C57BL, Myelopoiesis, Lipoprotein(a), Monocytes
- Abstract
Background: Lipoproteins are important regulators of hematopoietic stem and progenitor cell (HSPC) biology, predominantly affecting myelopoiesis. Since myeloid cells, including monocytes and macrophages, promote the inflammatory response that propagates atherosclerosis, it is of interest whether the atherogenic low-density lipoprotein (LDL)-like particle lipoprotein(a) [Lp(a)] contributes to atherogenesis via stimulating myelopoiesis., Methods & Results: To assess the effects of Lp(a)-priming on long-term HSPC behavior we transplanted BM of Lp(a) transgenic mice, that had been exposed to elevated levels of Lp(a), into lethally-irradiated C57Bl6 mice and hematopoietic reconstitution was analyzed. No differences in HSPC populations or circulating myeloid cells were detected ten weeks after transplantation. Likewise, in vitro stimulation of C57Bl6 BM cells for 24 h with Lp(a) did not affect colony formation, total cell numbers or myeloid populations 7 days later. To assess the effects of elevated levels of Lp(a) on myelopoiesis, C57Bl6 bone marrow (BM) cells were stimulated with lp(a) for 24 h, and a marked increase in granulocyte-monocyte progenitors, pro-inflammatory Ly6
high monocytes and macrophages was observed. Seven days of continuous exposure to Lp(a) increased colony formation and enhanced the formation of pro-inflammatory monocytes and macrophages. Antibody-mediated neutralization of oxidized phospholipids abolished the Lp(a)-induced effects on myelopoiesis., Conclusion: Lp(a) enhances the production of inflammatory monocytes at the bone marrow level but does not induce cell-intrinsic long-term priming of HSPCs. Given the short-term and direct nature of this effect, we postulate that Lp(a)-lowering treatment has the capacity to rapidly revert this multi-level inflammatory response., (Copyright © 2020 The Authors. Published by Elsevier B.V. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
34. Generation and characterization of LPA-KIV9, a murine monoclonal antibody binding a single site on apolipoprotein (a).
- Author
-
Gonen A, Yang X, Yeang C, Alekseeva E, Koschinsky M, Witztum JL, Boffa M, and Tsimikas S
- Subjects
- Animals, Mice, Humans, Apolipoproteins A metabolism, Binding Sites, Antibodies, Monoclonal immunology, Mice, Inbred BALB C
- Abstract
Lipoprotein (a) [Lp(a)] is a risk factor for CVD and a target of therapy, but Lp(a) measurements are not globally standardized. Commercially available assays generally use polyclonal antibodies that detect multiple sites within the kringle (K)IV
2 repeat region of Lp(a) and may lead to inaccurate assessments of plasma levels. With increasing awareness of Lp(a) as a cardiovascular risk factor and the active clinical development of new potential therapeutic approaches, the broad availability of reagents capable of providing isoform independence of Lp(a) measurements is paramount. To address this issue, we generated a murine monoclonal antibody that binds to only one site on apo(a). A BALB/C mouse was immunized with a truncated version of apo(a) that contained eight total KIV repeats, including only one copy of KIV2 We generated hybridomas, screened them, and successfully produced a KIV2 -independent monoclonal antibody, named LPA-KIV9. Using a variety of truncated apo(a) constructs to map its binding site, we found that LPA-KIV9 binds to KIV9 without binding to plasminogen. Fine peptide mapping revealed that LPA-KIV9 bound to the sequence4076 LETPTVV4082 on KIV9 In conclusion, the generation of monoclonal antibody LPA-KIV9 may be a useful reagent in basic research studies and in the clinical application of Lp(a) measurements., Competing Interests: Conflict of interest—A.G., S.T., and J.L.W. are co-inventors and receive royalties from patents owned by the University of California San Diego on oxidation-specific antibodies and of biomarkers related to oxidized lipoproteins. S.T. and J.L.W. are co-founders and have an equity interest in Oxitope, Inc. and its affiliates (Oxitope) as well as in Kleanthi Diagnostics, LLC (Kleanthi). Although these relationships have been identified for conflict of interest management based on the overall scope of the project and its potential benefit to Oxitope and Kleanthi, the research findings included in this particular publication may not necessarily relate to the interests of Oxitope and Kleanthi. The terms of this arrangement have been reviewed and approved by the University of California San Diego in accordance with its conflict of interest policies. S.T. has a dual appointment at University of California San Diego and Ionis Pharmaceuticals. J.L.W. is a consultant to Ionis Pharmaceuticals. M.L.K. is a paid consultant for Ayma Therapeutics and holds a research contract from Abcentra. M.B.B. is a paid consultant for Ayma Therapeutics. The other authors declare that they have no conflicts with the contents of this article., (Copyright © 2020 Gonen et al.)- Published
- 2020
- Full Text
- View/download PDF
35. The dedicated "Lp(a) clinic": A concept whose time has arrived?
- Author
-
Tsimikas S and Stroes ESG
- Subjects
- Biomarkers blood, Cardiovascular Diseases diagnosis, Cardiovascular Diseases epidemiology, Cardiovascular Diseases therapy, Delivery of Health Care, Integrated, Humans, Hyperlipoproteinemias diagnosis, Hyperlipoproteinemias epidemiology, Hyperlipoproteinemias therapy, Patient Care Team, Prevalence, Prognosis, Up-Regulation, Ambulatory Care, Ambulatory Care Facilities, Cardiovascular Diseases blood, Hyperlipoproteinemias blood, Lipoprotein(a) blood
- Abstract
The emergence of pathophysiological, epidemiologic, and genetic data strongly supports the causality for lipoprotein(a) [Lp(a)] in cardiovascular disease (CVD) and calcific aortic valve disease (CAVD). In parallel, novel Lp(a) lowering approaches have been developed that have re-invigorated clinical interest in Lp(a). Because Lp(a) is the most prevalent monogenetic lipid disorder globally, with prevalence of Lp(a) > 50 mg/dL estimated at >1.4 billion people, the rationale for diagnosing and managing Lp(a)-mediated risk is now stronger than ever. Patients with elevated Lp(a) are significantly under-diagnosed and the diagnosis is frequently made ad hoc rather than systematically. Elevated Lp(a) levels are associated with atherothrombotic risk and patients present with varied clinical phenotypes, ranging from stroke in pediatric age groups, to ST-segment elevation myocardial infarction in young males, to CAVD in elderly individuals. A new clinical care paradigm of a dedicated "Lp(a) Clinic" would serve to evaluate and manage such patients who have elevated Lp(a) as the pathophysiological etiology. Such a clinic would include multidisciplinary expertise in lipid metabolism, clinical cardiology, vascular medicine, valvular disease, thrombosis, and pediatric aspects of clinical care. This viewpoint argues for the rationale of an Lp(a) outpatient clinic where patients with elevated Lp(a) and their affected relatives can be referred, evaluated, managed and followed, to ultimately reduce Lp(a)-mediated CVD and CAVD risk., Competing Interests: Declaration of competing interest ST is a co-inventor, receives royalties from patents owned by the University of California San Diego on oxidation-specific antibodies, is a co-founder of Oxitope, Inc and Kleanthi Diagnostics and currently has a dual appointment at UCSD and as an employee of Ionis Pharmaceuticals. ESGS is a consultant for Akcea Therapeutics, Amgen, Sanofi, Esperion, and has received research grants from Sanofi., (Copyright © 2020. Published by Elsevier B.V.)
- Published
- 2020
- Full Text
- View/download PDF
36. Potent reduction of plasma lipoprotein (a) with an antisense oligonucleotide in human subjects does not affect ex vivo fibrinolysis.
- Author
-
Boffa MB, Marar TT, Yeang C, Viney NJ, Xia S, Witztum JL, Koschinsky ML, and Tsimikas S
- Subjects
- Adolescent, Adult, Aged, Female, Fibrinolysis genetics, Humans, Lipoprotein(a) genetics, Male, Middle Aged, Young Adult, Lipoprotein(a) blood, Oligonucleotides, Antisense genetics
- Abstract
It is postulated that lipoprotein (a) [Lp(a)] inhibits fibrinolysis, but this hypothesis has not been tested in humans due to the lack of specific Lp(a) lowering agents. Patients with elevated Lp(a) were randomized to antisense oligonucleotide [IONIS-APO(a)
Rx ] directed to apo(a) ( n = 7) or placebo ( n = 10). Ex vivo plasma lysis times and antigen concentrations of plasminogen, factor XI, plasminogen activator inhibitor 1, thrombin activatable fibrinolysis inhibitor, and fibrinogen at baseline, day 85/92/99 (peak drug effect), and day 190 (3 months off drug) were measured. The mean ± SD baseline Lp(a) levels were 477.3 ± 55.9 nmol/l in IONIS-APO(a)Rx and 362.1 ± 89.9 nmol/l in placebo. The mean± SD percentage change in Lp(a) for IONIS-APO(a)Rx was -69.3 ± 12.2% versus -5.4 ± 6.9% placebo ( P < 0.0010) at day 85/92/99 and -15.6 ± 8.9% versus 3.2 ± 12.2% ( P = 0.003) at day 190. Clot lysis times and coagulation/fibrinolysis-related biomarkers showed no significant differences between IONIS-APO(a)Rx and placebo at all time points. Clot lysis times were not affected by exogenously added Lp(a) at concentrations up to 200 nmol/l to plasma with very low (12.5 nmol/l) Lp(a) levels, whereas recombinant apo(a) had a potent antifibrinolytic effect. In conclusion, potent reductions of Lp(a) in patients with highly elevated Lp(a) levels do not affect ex vivo measures of fibrinolysis; the relevance of any putative antifibrinolytic effects of Lp(a) in vivo needs further study., (Copyright © 2019 Boffa et al.)- Published
- 2019
- Full Text
- View/download PDF
37. Erratum: A monoclonal antibody to assess oxidized cholesteryl esters associated with apoAI and apoB-100 lipoproteins in human plasma.
- Author
-
Gonen A, Choi SH, Miu P, Agatisa-Boyle C, Acks D, Taylor AM, McNamara CA, Tsimikas S, Witztum JL, and Miller YI
- Published
- 2019
- Full Text
- View/download PDF
38. PCSK9 loss-of-function variants and Lp(a) phenotypes among black US adults.
- Author
-
Mefford MT, Marcovina SM, Bittner V, Cushman M, Brown TM, Farkouh ME, Tsimikas S, Monda KL, López JAG, Muntner P, and Rosenson RS
- Subjects
- Female, Humans, Male, Middle Aged, Phenotype, Proprotein Convertase 9 deficiency, Proprotein Convertase 9 metabolism, United States, Black or African American genetics, Lipoprotein(a) genetics, Loss of Function Mutation genetics, Proprotein Convertase 9 genetics
- Abstract
The pharmacologic inhibition of proprotein convertase subtilisin-kexin type 9 (PCSK9) lowers lipoprotein (a) [Lp(a)] concentrations. However, the impact of genetic PCSK9 loss-of-function variants (LOFVs) on Lp(a) is uncertain. We determined the association of PCSK9 LOFVs with Lp(a) measures among black adults. Genotyping for PCSK9 LOFVs was conducted in 10,196 black Reasons for Geographic and Racial Differences in Stroke study participants. Among 241 participants with and 723 randomly selected participants without PCSK9 LOFVs, Lp(a) concentations, apo(a) kringle IV (KIV) repeats (a proxy for isoform size), and oxidized phospholipid (OxPL) apoB levels were measured using validated methods. Median Lp(a) concentrations among participants with and without PCSK9 LOFVs were 63.2 and 80.4 nmol/l, respectively ( P = 0.016). After adjusting for age, sex, estimated glomerular filtration rate, LDL cholesterol, and statin use, participants with versus without a PCSK9 LOFV had a lower median Lp(a) concentration [Δ = -18.8 nmol/l (95% CI: -34.2, -3.3)]. Median apo(a) isoform sizes were 24 and 23 KIV repeats ( P = 0.12) among participants with and without PCSK9 LOFVs, respectively [Δ = 1.1 (95% CI: 0.2, 2.0) after adjustment]. Median OxPL-apoB levels among participants with and without PCSK9 LOFVs were 3.4 and 4.1 nM ( P = 0.20), respectively [Δ = -1.2 nM (95% CI -2.4, -0.04) after adjustment]. Among black adults, PCSK9 LOFVs were associated with lower Lp(a) concentration and OxPL-apoB levels., (Copyright © 2019 Mefford et al.)
- Published
- 2019
- Full Text
- View/download PDF
39. The challenges of measuring Lp(a): A fight against Hydra?
- Author
-
Kronenberg F and Tsimikas S
- Subjects
- Animals, Immunoassay, Lipoprotein(a), Hydra
- Published
- 2019
- Full Text
- View/download PDF
40. Nanobody-Facilitated Multiparametric PET/MRI Phenotyping of Atherosclerosis.
- Author
-
Senders ML, Hernot S, Carlucci G, van de Voort JC, Fay F, Calcagno C, Tang J, Alaarg A, Zhao Y, Ishino S, Palmisano A, Boeykens G, Meerwaldt AE, Sanchez-Gaytan BL, Baxter S, Zendman L, Lobatto ME, Karakatsanis NA, Robson PM, Broisat A, Raes G, Lewis JS, Tsimikas S, Reiner T, Fayad ZA, Devoogdt N, Mulder WJM, and Pérez-Medina C
- Subjects
- Animals, Atherosclerosis genetics, Atherosclerosis immunology, Atherosclerosis pathology, Disease Models, Animal, Disease Progression, Early Diagnosis, Genetic Predisposition to Disease, Lectins, C-Type immunology, Mannose Receptor, Mannose-Binding Lectins immunology, Mice, Knockout, ApoE, Multimodal Imaging, Phenotype, Rabbits, Radiopharmaceuticals pharmacokinetics, Receptors, Cell Surface immunology, Scavenger Receptors, Class E immunology, Single-Domain Antibodies metabolism, Vascular Cell Adhesion Molecule-1 immunology, Atherosclerosis diagnostic imaging, Multiparametric Magnetic Resonance Imaging, Plaque, Atherosclerotic, Positron-Emission Tomography, Radiopharmaceuticals administration & dosage, Single-Domain Antibodies administration & dosage
- Abstract
Objectives: This study sought to develop an integrative positron emission tomography (PET) with magnetic resonance imaging (MRI) procedure for accurate atherosclerotic plaque phenotyping, facilitated by clinically approved and nanobody radiotracers., Background: Noninvasive characterization of atherosclerosis remains a challenge in clinical practice. The limitations of current diagnostic methods demonstrate that, in addition to atherosclerotic plaque morphology and composition, disease activity needs to be evaluated., Methods: We screened 3 nanobody radiotracers targeted to different biomarkers of atherosclerosis progression, namely vascular cell adhesion molecule (VCAM)-1, lectin-like oxidized low-density lipoprotein receptor (LOX)-1, and macrophage mannose receptor (MMR). The nanobodies, initially radiolabeled with copper-64 (
64 Cu), were extensively evaluated in Apoe-/- mice and atherosclerotic rabbits using a combination of in vivo PET/MRI readouts and ex vivo radioactivity counting, autoradiography, and histological analyses., Results: The 3 nanobody radiotracers accumulated in atherosclerotic plaques and displayed short circulation times due to fast renal clearance. The MMR nanobody was selected for labeling with gallium-68 (68 Ga), a short-lived radioisotope with high clinical relevance, and used in an ensuing atherosclerosis progression PET/MRI study. Macrophage burden was longitudinally studied by68 Ga-MMR-PET, plaque burden by T2-weighted MRI, and neovascularization by dynamic contrast-enhanced (DCE) MRI. Additionally, inflammation and microcalcifications were evaluated by fluorine-18 (18 F)-labeled fluorodeoxyglucose (18 F-FDG) and18 F-sodium fluoride (18 F-NaF) PET, respectively. We observed an increase in all the aforementioned measures as disease progressed, and the imaging signatures correlated with histopathological features., Conclusions: We have evaluated nanobody-based radiotracers in rabbits and developed an integrative PET/MRI protocol that allows noninvasive assessment of different processes relevant to atherosclerosis progression. This approach allows the multiparametric study of atherosclerosis and can aid in early stage anti-atherosclerosis drug trials., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
41. ApoC-III ASO promotes tissue LPL activity in the absence of apoE-mediated TRL clearance.
- Author
-
Ramms B, Patel S, Nora C, Pessentheiner AR, Chang MW, Green CR, Golden GJ, Secrest P, Krauss RM, Metallo CM, Benner C, Alexander VJ, Witztum JL, Tsimikas S, Esko JD, and Gordts PLSM
- Subjects
- Animals, Apolipoprotein C-III genetics, Lipoprotein Lipase genetics, Mice, Mice, Knockout, ApoE, Receptors, LDL genetics, Receptors, LDL metabolism, Apolipoprotein C-III metabolism, Apolipoproteins E deficiency, Lipoprotein Lipase metabolism, Triglycerides blood
- Abstract
Hypertriglyceridemia results from accumulation of triglyceride (TG)-rich lipoproteins (TRLs) in the circulation and is associated with increased CVD risk. ApoC-III is an apolipoprotein on TRLs and a prominent negative regulator of TG catabolism. We recently established that in vivo apoC-III predominantly inhibits LDL receptor-mediated and LDL receptor-related protein 1-mediated hepatic TRL clearance and that apoC-III-enriched TRLs are preferentially cleared by syndecan-1 (SDC1). In this study, we determined the impact of apoE, a common ligand for all three receptors, on apoC-III metabolism using apoC-III antisense oligonucleotide (ASO) treatment in mice lacking apoE and functional SDC1 ( Apoe
-/- Ndst1f/f Alb-Cre+ ). ApoC-III ASO treatment significantly reduced plasma TG levels in Apoe-/- Ndst1f/f Alb-Cre+ mice without reducing hepatic VLDL production or improving hepatic TRL clearance. Further analysis revealed that apoC-III ASO treatment lowered plasma TGs in Apoe-/- Ndst1f/f Alb-Cre+ mice, which was associated with increased LPL activity in white adipose tissue in the fed state. Finally, clinical data confirmed that ASO-mediated lowering of APOC-III via volanesorsen can reduce plasma TG levels independent of the APOE isoform genotype. Our data indicate that apoE determines the metabolic impact of apoC-III as we establish that apoE is essential to mediate inhibition of TRL clearance by apoC-III and that, in the absence of functional apoE, apoC-III inhibits tissue LPL activity., (Copyright © 2019 Ramms et al. Published by The American Society for Biochemistry and Molecular Biology, Inc.)- Published
- 2019
- Full Text
- View/download PDF
42. Lipoprotein Apheresis Acutely Reverses Coronary Microvascular Dysfunction in Patients With Severe Hypercholesterolemia.
- Author
-
Wu MD, Moccetti F, Brown E, Davidson BP, Atkinson T, Belcik JT, Giraud G, Duell PB, Fazio S, Tavori H, Tsimikas S, and Lindner JR
- Subjects
- Aged, Animals, Biomarkers blood, Case-Control Studies, Coronary Disease blood, Coronary Disease diagnostic imaging, Coronary Disease etiology, Down-Regulation, Female, Humans, Hypercholesterolemia blood, Hypercholesterolemia complications, Hypercholesterolemia diagnosis, Male, Middle Aged, Peripheral Arterial Disease blood, Peripheral Arterial Disease diagnostic imaging, Peripheral Arterial Disease etiology, Recovery of Function, Severity of Illness Index, Sheep, Domestic, Time Factors, Treatment Outcome, Blood Component Removal adverse effects, Cholesterol, LDL blood, Coronary Circulation, Coronary Disease physiopathology, Hypercholesterolemia therapy, Microcirculation, Muscle, Skeletal blood supply, Peripheral Arterial Disease physiopathology
- Abstract
Objectives: This study evaluated whether lipoprotein apheresis produces immediate changes in resting perfusion in subjects with severe hypercholesterolemia, and whether there is a difference in the response between peripheral and coronary microcirculations., Background: Lipoprotein apheresis is used in patients with severe hypercholesterolemia to reduce plasma levels of low-density lipoprotein cholesterol., Methods: Quantitative contrast-enhanced ultrasound perfusion imaging of the myocardium at rest and skeletal muscle at rest and during calibrated contractile exercise was performed before and immediately after lipoprotein apheresis in 8 subjects with severe hypercholesterolemia, 7 of whom had a diagnosis of familial hypercholesterolemia. Myocardial perfusion imaging was also performed in 14 normal control subjects. Changes in myocardial work and left ventricular function were assessed by echocardiography. Ex vivo ovine coronary and femoral artery ring tension assays were assessed in the presence of pre- and post-apheresis plasma., Results: Apheresis acutely decreased low-density lipoprotein cholesterol (234.9 ± 103.2 mg/dl vs. 67.1 ± 49.5 mg/dl; p < 0.01) and oxidized phospholipid on apolipoprotein B-100 (60.2 ± 55.2 nmol/l vs. 47.0 ± 24.5 nmol/l; p = 0.01), and acutely increased resting myocardial perfusion (55.1 [95% confidence interval: 77.2 to 73.1] IU/s vs. 135 [95% confidence interval: 81.2 to 189.6] IU/s; p = 0.01), without changes in myocardial work. Myocardial longitudinal strain improved in those subjects with reduced pre-apheresis function. Skeletal muscle perfusion at rest and during contractile exercise was unchanged by apheresis. Acetylcholine-mediated dilation of ex vivo ovine coronary but not femoral arteries was impaired in pre-apheresis plasma and was completely reversed in post-apheresis plasma., Conclusions: Lipoprotein apheresis produces an immediate improvement in coronary microvascular function, which increases myocardial perfusion and normalizes endothelial-dependent vasodilation. These changes are not observed in the periphery. (Acute Microvascular Changes With LDL Apheresis; NCT02388633)., (Copyright © 2019 American College of Cardiology Foundation. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
43. A monoclonal antibody to assess oxidized cholesteryl esters associated with apoAI and apoB-100 lipoproteins in human plasma.
- Author
-
Gonen A, Choi SH, Miu P, Agatisa-Boyle C, Acks D, Taylor AM, McNamara CA, Tsimikas S, Witztum JL, and Miller YI
- Subjects
- Animals, Cholesterol Esters immunology, Humans, Mice, Oxidation-Reduction, Antibodies, Monoclonal immunology, Apolipoprotein A-I metabolism, Apolipoprotein B-100 metabolism, Cholesterol Esters blood, Cholesterol Esters metabolism, Enzyme-Linked Immunosorbent Assay methods
- Abstract
Atherosclerosis is associated with increased lipid peroxidation, leading to generation of multiple oxidation-specific epitopes (OSEs), contributing to the pathogenesis of atherosclerosis and its clinical manifestation. Oxidized cholesteryl esters (OxCEs) are a major class of OSEs found in human plasma and atherosclerotic tissue. To evaluate OxCEs as a candidate biomarker, we generated a novel mouse monoclonal Ab (mAb) specific to an OxCE modification of proteins. The mAb AG23 (IgG1) was raised in C57BL6 mice immunized with OxCE-modified keyhole limpet hemocyanin, and hybridomas were screened against OxCE-modified BSA. This method ensures mAb specificity to the OxCE modification, independent of a carrier protein. AG23 specifically stained human carotid artery atherosclerotic lesions. An ELISA method, with AG23 as a capture and either anti-apoAI or anti-apoB-100 as the detection Abs, was developed to assay apoAI and apoB-100 lipoproteins that have one or more OxCE epitopes. OxCE-apoA or OxCE-apoB did not correlate with the well-established oxidized phospholipid-apoB biomarker. In a cohort of subjects treated with atorvastatin, OxCE-apoA was significantly lower than in the placebo group, independent of the apoAI levels. These results suggest the potential diagnostic utility of a new biomarker assay to measure OxCE-modified lipoproteins in patients with CVD., (Copyright © 2019 Gonen et al.)
- Published
- 2019
- Full Text
- View/download PDF
44. Relationship of lipoprotein-associated apolipoprotein C-III with lipid variables and coronary artery disease risk: The EPIC-Norfolk prospective population study.
- Author
-
van Capelleveen JC, Lee SR, Verbeek R, Kastelein JJP, Wareham NJ, Stroes ESG, Hovingh GK, Khaw KT, Boekholdt SM, Witztum JL, and Tsimikas S
- Subjects
- Aged, Apolipoprotein A-I blood, Apolipoprotein B-100 blood, Female, Follow-Up Studies, Humans, Male, Middle Aged, Prospective Studies, Risk Factors, Apolipoprotein C-III blood, Coronary Artery Disease blood
- Abstract
Background: Plasma apolipoprotein C-III (apoC-III) levels are associated with coronary artery disease (CAD) risk., Objective: To assess whether lipoprotein-associated apoC-III levels predict risk of CAD events., Methods: apoC-III associated with apoB, apoAI, and Lp(a) (apoCIII-apoB, apoCIII-apoAI, and apoCIII-Lp(a), respectively) were measured using high-throughput chemiluminescent enzyme-linked immunoassays in 2711 subjects (1879 controls and 832 cases with CAD) in the European Prospective Investigation into Cancer and Nutrition-Norfolk prospective population study with 7.4 years of follow-up. These measures were correlated with a variety of lipid measurements and the presence of CAD. The indices of "total apoCIII-apoB" and "total apoCIII-apoAI" were derived by multiplying plasma apoB and apoAI, respectively., Results: apoCIII-apoB (P = .001), apoCIII-Lp(a) (P < .001), apoCIII-apoAI (P = .005) were higher in cases vs controls; tended to correlate positively with body mass index, hsCRP, apoC-III, low-density lipoprotein (LDL) cholesterol, triglycerides, remnant cholesterol, very low density lipoprotein, LDL and high-density lipoprotein particle number and very low density lipoprotein size; but negatively with LDL and high-density lipoprotein particle size (P < .001 for all). apoCIII-apoB, apoCIII-apoAI, apoCIII-Lp(a), total apoCIII-Lp(a), and total apoCIII-apoB were predictors of CAD after adjustment of age, sex, body mass index, smoking, diabetes, hypertensive and lipid-lowering drug use, but they lost their significance after further adjustment of lipid and lipoprotein variables., Conclusions: This study suggests that enzyme-linked immunoassay-measured lipoprotein-associated apoC-III markers reflect atherogenic lipid particles but do not independently predict risk of CAD events., (Copyright © 2018 National Lipid Association. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
45. Baseline and on-statin treatment lipoprotein(a) levels for prediction of cardiovascular events: individual patient-data meta-analysis of statin outcome trials.
- Author
-
Willeit P, Ridker PM, Nestel PJ, Simes J, Tonkin AM, Pedersen TR, Schwartz GG, Olsson AG, Colhoun HM, Kronenberg F, Drechsler C, Wanner C, Mora S, Lesogor A, and Tsimikas S
- Subjects
- Aged, Biomarkers blood, Cardiovascular Diseases epidemiology, Cholesterol, HDL blood, Cholesterol, LDL blood, Female, Humans, Lipoprotein(a) drug effects, Male, Middle Aged, Randomized Controlled Trials as Topic, Risk Factors, Cardiovascular Diseases blood, Cardiovascular Diseases prevention & control, Hydroxymethylglutaryl-CoA Reductase Inhibitors therapeutic use, Hypercholesterolemia drug therapy, Lipoprotein(a) blood
- Abstract
Background: Elevated lipoprotein(a) is a genetic risk factor for cardiovascular disease in general population studies. However, its contribution to risk for cardiovascular events in patients with established cardiovascular disease or on statin therapy is uncertain., Methods: Patient-level data from seven randomised, placebo-controlled, statin outcomes trials were collated and harmonised to calculate hazard ratios (HRs) for cardiovascular events, defined as fatal or non-fatal coronary heart disease, stroke, or revascularisation procedures. HRs for cardiovascular events were estimated within each trial across predefined lipoprotein(a) groups (15 to <30 mg/dL, 30 to <50 mg/dL, and ≥50 mg/dL, vs <15 mg/dL), before pooling estimates using multivariate random-effects meta-analysis., Findings: Analyses included data for 29 069 patients with repeat lipoprotein(a) measurements (mean age 62 years [SD 8]; 8064 [28%] women; 5751 events during 95 576 person-years at risk). Initiation of statin therapy reduced LDL cholesterol (mean change -39% [95% CI -43 to -35]) without a significant change in lipoprotein(a). Associations of baseline and on-statin treatment lipoprotein(a) with cardiovascular disease risk were approximately linear, with increased risk at lipoprotein(a) values of 30 mg/dL or greater for baseline lipoprotein(a) and 50 mg/dL or greater for on-statin lipoprotein(a). For baseline lipoprotein(a), HRs adjusted for age and sex (vs <15 mg/dL) were 1·04 (95% CI 0·91-1·18) for 15 mg/dL to less than 30 mg/dL, 1·11 (1·00-1·22) for 30 mg/dL to less than 50 mg/dL, and 1·31 (1·08-1·58) for 50 mg/dL or higher; respective HRs for on-statin lipoprotein(a) were 0·94 (0·81-1·10), 1·06 (0·94-1·21), and 1·43 (1·15-1·76). HRs were almost identical after further adjustment for previous cardiovascular disease, diabetes, smoking, systolic blood pressure, LDL cholesterol, and HDL cholesterol. The association of on-statin lipoprotein(a) with cardiovascular disease risk was stronger than for on-placebo lipoprotein(a) (interaction p=0·010) and was more pronounced at younger ages (interaction p=0·008) without effect-modification by any other patient-level or study-level characteristics., Interpretation: In this individual-patient data meta-analysis of statin-treated patients, elevated baseline and on-statin lipoprotein(a) showed an independent approximately linear relation with cardiovascular disease risk. This study provides a rationale for testing the lipoprotein(a) lowering hypothesis in cardiovascular disease outcomes trials., Funding: Novartis Pharma AG., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
46. Relationship of lipoprotein(a) molar concentrations and mass according to lipoprotein(a) thresholds and apolipoprotein(a) isoform size.
- Author
-
Tsimikas S, Fazio S, Viney NJ, Xia S, Witztum JL, and Marcovina SM
- Subjects
- Apoprotein(a) metabolism, Humans, Lipoprotein(a) genetics, Molecular Weight, Polymorphism, Single Nucleotide, Protein Isoforms chemistry, Protein Isoforms metabolism, Apoprotein(a) chemistry, Lipoprotein(a) chemistry, Lipoprotein(a) metabolism
- Abstract
Background: Lipoprotein(a) [Lp(a)] is reported as Lp(a) particle mass (mg/dL) or molar concentration of apolipoprotein(a) [apo(a)] (nmol/L), which is considered the gold standard. Values are often converted from one measurement to the other but the validity of this is unknown., Objectives: To quantify the relationship between Lp(a) molar concentration and Lp(a) mass in the context of various Lp(a) level thresholds and apo(a) isoform size., Methods: In all samples, Lp(a) levels in molar concentration and apo(a) isoform size were determined at the Northwest Lipid Metabolism and Diabetes Research Laboratories (NLMDRL). Lp(a) mass levels were determined at the University of California, San Diego (UCSD) (1635 samples), by 5 commercially available assays: Denka 1 and Denka 2 (each 80 samples), 2 turbidimetric assays (2545 and 2673 samples, respectively), and an enzyme-linked immunosorbent assay (2605 samples). The ratios between Lp(a) molar concentration and mass (eg, nmol/L/mg/dL) were calculated and related to apo(a) isoform size., Results: The mean (SD) ratios for NLMDRL/UCSD, NLMDRL/Denka1, and NLMDRL/Denka2 were 2.42 (1.25), 1.64 (0.18), and 2.02 (0.22), respectively. The ratios for NLMDRL/UCSD, NLMDRL/Denka1, and NLMDRL/Denka2 increased by Lp(a) cutoffs, with ratios of 1.82, 1.52, and 1.87, respectively, for Lp(a) < 75 nmol/L and 2.80, 1.89, and 2.24, respectively, for Lp(a) > 125 nmol/L. For the commercial turbidimetric assays and enzyme-linked immunosorbent assay, the ratios ranged from <1 to >5., Conclusions: Lp(a) molar/mass ratios are threshold, method, and isoform dependent. A single conversion factor between assays is not appropriate. These data support the transition of Lp(a) mass assays to molar concentration to improve diagnostic and clinical interpretation of Lp(a)-mediated risk., (Copyright © 2018 National Lipid Association. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
47. Relationship between "LDL-C", estimated true LDL-C, apolipoprotein B-100, and PCSK9 levels following lipoprotein(a) lowering with an antisense oligonucleotide.
- Author
-
Viney NJ, Yeang C, Yang X, Xia S, Witztum JL, and Tsimikas S
- Subjects
- Female, Humans, Male, Apolipoprotein B-100 blood, Cholesterol, LDL blood, Lipoprotein(a) blood, Lipoprotein(a) genetics, Oligonucleotides, Antisense genetics, Proprotein Convertase 9 blood
- Abstract
Background: The laboratory measurement of "low-density lipoprotein cholesterol (LDL-C)" includes the cholesterol content of lipoprotein(a) (Lp(a)-C)., Objective: To estimate the "true" LDL-C in relation to changes in apolipoprotein B-100 (apoB-100) and assess changes in proprotein convertase subtilisin/kexin 9 (PCSK9) levels in patients with elevated Lp(a) treated with IONIS-APO(a)
Rx. METHODS: A pooled placebo group (n = 29), and cohort A (n = 24, baseline Lp(a) 50-175 mg/dL) and cohort B (n = 8, baseline Lp(a) > 175 mg/dL) treated with IONIS-APO(a)Rx were studied. Lp(a) particle number, ultracentrifugation-measured "LDL-C", apoB-100, total PCSK9, and lipoprotein-associated PCSK9 (PCSK9-Lp(a), PCSK9-apoB, PCSK9-apoAI) were measured. Lp(a)-cholesterol (Lp(a)-C) and LDL-C corrected for Lp(a)-C (LDL-Ccorr ) were calculated., Results: Baseline mean (standard deviation) "LDL-C" was 120 (42), 128 (45), and 112 (39) mg/dL in placebo, cohorts A and B, respectively, whereas LDL-Ccorr was 86 (48), 96 (43), and 57 (37) mg/dL (P < .001 compared with placebo), representing 28%, 25%, and 50% lower levels than "LDL-C". Following IONIS-APO(a)Rx treatment at day 85/99, Lp(a) particle number and Lp(a)-C decreased -66.8% and -71.6%, apoB-100 -10.3% and -17.5%, "LDL-C" -11.8% and -22.7%, (P < .001 for all vs placebo), whereas LDL-Ccorr increased +10.4% (P = .66) and +49.9% (P < .001) in cohorts A and B, respectively. Total PCSK9 did not change but PCSK9-Lp(a) decreased with IONIS-APO(a)Rx vs placebo (-39.0% vs +8.4%, P < .001)., Conclusion: LDL-Ccorr is lower than laboratory "LDL-C" in patients with elevated Lp(a). Following apolipoprotein(a) inhibition and decline in Lp(a) and Lp(a)-C, the decline in apoB-100 is consistent with the notion that LDL devoid of apo(a) is cleared faster than Lp(a). These types of analyses may provide insights into the mechanisms of drugs affecting Lp(a) levels in clinical trials., (Copyright © 2018 National Lipid Association. Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
48. In search of a physiological function of lipoprotein(a): causality of elevated Lp(a) levels and reduced incidence of type 2 diabetes.
- Author
-
Tsimikas S
- Subjects
- Humans, Incidence, Kringles, Lipoprotein(a), Risk Factors, Cardiovascular Diseases, Diabetes Mellitus, Type 2
- Published
- 2018
- Full Text
- View/download PDF
49. Interleukin-1 genotypes modulate the long-term effect of lipoprotein(a) on cardiovascular events: The Ioannina Study.
- Author
-
Naka KK, Bechlioullis A, Marini A, Sionis D, Vakalis K, Triantis G, Wilkins L, Rogus J, Kornman KS, Witztum JL, Doucette-Stamm L, Michalis LK, and Tsimikas S
- Subjects
- Aged, Cardiovascular Diseases diagnosis, Coronary Artery Disease diagnosis, Female, Genotype, Humans, Logistic Models, Male, Middle Aged, Polymorphism, Single Nucleotide, Prospective Studies, Risk Factors, Time Factors, Biomarkers metabolism, Cardiovascular Diseases genetics, Coronary Artery Disease genetics, Interleukin-1 genetics, Lipoprotein(a) genetics
- Abstract
Background: Lipoprotein(a) [Lp(a)] is a genetic risk factor for cardiovascular disease (CVD), and proinflammatory interleukin-1 (IL-1) genotypes may influence Lp(a)-mediated CVD events. The genotype IL-1(+) is associated with higher rates of inflammation than IL-1(-) genotype. Targeting IL-1β was recently shown to decrease CVD events independent of low-density lipoprotein-cholesterol levels., Objective: The objective of the study is to assess the modulatory effect of IL-1 genotypes on risk mediated by Lp(a) METHODS: We assessed whether IL-1 genotypes modulate the effect of Lp(a) on major adverse cardiovascular events (cardiovascular death, myocardial infarction, and stroke/transient ischemic attack) and angiographically determined coronary artery disease (CAD). IL-1 genotypes and Lp(a) were measured in 603 patients without diabetes mellitus undergoing angiography. Major adverse cardiovascular events and CAD were assessed over a median of 45 months., Results: In multivariable-adjusted analysis, Lp(a) was associated with major adverse cardiovascular events (hazard ratio [HR] [95% confidence interval {CI}]: 2.95 [1.16-7.54], P = .023) and CAD (odds ratio [OR] [95% CI]: 1.84 [1.12-3.03], P = .016) comparing quartile 4 vs quartile 1. In Cox regression analysis, IL-1(+) patients with Lp(a) above the median (>9.2 mg/dL) had a worse event-free cumulative survival (HR [95% CI]: 3.59 [1.07-12.03], P = .039) compared to IL-1(-) patients with Lp(a) below the median. In IL-1(+) patients aged ≤60 years, Lp(a) was also associated with angiographically determined CAD (OR [95% CI]: 2.90 [1.07-7.86], P = .036) comparing quartile 4 vs quartile 1 but not IL-1(-) patients., Conclusion: Proinflammatory IL-1(+) genotypes modulate the risk of Lp(a) long-term CVD events and CAD. These data suggest that the dual genetic contributions of elevated Lp(a) levels and IL-1(+) genotypes may identify younger subjects at particularly high risk for CVD events., (Copyright © 2017 National Lipid Association. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
50. Temporal variability in lipoprotein(a) levels in patients enrolled in the placebo arms of IONIS-APO(a) Rx and IONIS-APO(a)-L Rx antisense oligonucleotide clinical trials.
- Author
-
Marcovina SM, Viney NJ, Hughes SG, Xia S, Witztum JL, and Tsimikas S
- Subjects
- Apoprotein(a) antagonists & inhibitors, Apoprotein(a) genetics, Apoprotein(a) metabolism, Cholesterol, LDL blood, Cohort Studies, Double-Blind Method, Humans, Oligonucleotides, Antisense chemistry, Placebo Effect, Treatment Outcome, Acetylgalactosamine chemistry, Cardiovascular Diseases drug therapy, Lipoprotein(a) blood, Oligonucleotides, Antisense therapeutic use
- Abstract
Background: Lipoprotein(a) [Lp(a)] levels are primarily genetically determined, but their natural variability is not well known., Objective: The aim of the study was to evaluate the short-term temporal variability in Lp(a) in 3 placebo groups from the IONIS-APO(a)
Rx and IONIS-APO(a)-LRx trials., Methods: The placebo groups comprised 3 studies: Study 1 with 10 subjects with any Lp(a) concentration; Study 2 with 13 subjects with Lp(a) ≥75 nmol/L (∼30 mg/dL); and Study 3 with 29 patients with Lp(a) ≥125 nmol/L (≥∼50 mg/dL). Lp(a) was measured in serial blood samples (range 7-12 samples up to 190 days of follow-up) and analyzed as absolute change and mean percent change from baseline. Outliers were defined as having a > ±25% difference in Lp(a) from baseline at any future time point., Results: No significant temporal differences in mean absolute Lp(a) levels were present in any group. However, among individuals, the mean change in absolute Lp(a) levels at any time point ranged from -16.2 to +7.0 nmol/L in Study 1, -15.8 to +9.8 nmol/L in Study 2, and -60.2 to +16.6 nmol/L in Study 3. The mean percent change from baseline ranged from -9.4% to +21.6% for Study 1, -13.1% to 2.8% for Study 2, and -12.1% to +4.9% in Study 3. A total of 21 of 52 subjects (40.4%) were outliers, with 13 (62%) >25% up and 8 (38%) >25% down. Significant variability was also noted in other lipid parameters, but no outliers were noted with serum albumin., Conclusion: In subjects randomized to placebo in Lp(a) lowering trials, modest intra-individual temporal variability of mean Lp(a) levels was present. Significant number of subjects had > ±25% variation in Lp(a) in at least 1 time point. Although Lp(a) levels are primarily genetically determined, further study is required to define additional factors mediating short-term variability., (Copyright © 2017 National Lipid Association. Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.