You are on page 1of 97

Medicographia

Vol 38, No. 3, 2016 ISSN 0243-3397

Metabolic agents in the contemporary treatment of angina


C hance
Medicographia
favors only
the prepared mind Metabolic agents
Pa s t e u r in the contemporary
treatment of angina

Vol 38, No. 3



Pages 243-336
16 MP 0128 X Printed in France

Servier - 50, rue Carnot - 92284 SureSneS Cedex (FranCe) Quarterly Thematic Journal

of Clinical and Therapeutic Research


www.medicographia.com
2016

www.servier.com
A Servier Publication
Medicographia
a Servier publication

Editor in Chief: Frdric Sesini, PharmD


Medicographia
A Servier publication
Editorial Board: Antoine Beillevaire, MD;
Christophe Charpentier, MD; Philippe
Gonnard, MD; Jean-Christophe Guillerm,
MD; Philippe Malet, DVM; Bernard
Parrin, PharmD; Pascal Poullali, MA;
Hugues Renaut, PharmD; Marion
Schrenk, MD; Franois Vilette, PharmD

Medical Publications Editor:


Catriona Donagh, BAppSc
Philippines Slovenia United Kingdom
Production Editors: Mr P. tte Mr a. MaLLet dr a. LaLLouette
Emmanuelle Pinjon-Casu, PhD Servier Philippines, inc. Servier Pharma d.o.o. Servier Laboratories Ltd
(emmanuelle.pinjon-casu@servier.com) 2 orion Cor. Mercedes St. Pot K Sejmiscu 33 rowley Building, Wexham Springs
Bel air village - Makati 1231 Ljubljana Framewood road
John Plant, BPharm tel: 63-2 897 89 90 to 97 Crnuce Wexham Slough
(john.plant@servier.com) tel: 386 1 56 34 811 Berkshire SL3 6rJ
Poland tel: (44-1753) 662744
dr J. dreWLa South Africa
Editorial Assistant: Sophie Gehrisch Servier Polska Sp. z.o.o. Mr S. SPeLLer Uzbekistan
ui. Jana Kazimierza 10 Servier Laboratories Pty Ltd Mrs S. aSKarova
01-248 Warsaw n4 Country Club estate representative office of
Design: Studio DTC tel: (48-22) 594 90 00 Woodlands drive Les Laboratoires Servier in
Layout: Myriam Bucquoit Woodmead 2191 the republic of uzbekistan
Portugal Gauteng Block C, 6th Floor
and Bernard Crespin dr a. MendeS tel: 27 - 081 700 900 international Business Center
Servier Portugal 107B, amir temur avenue
Medicographia is published avenida antonio augusto Spain 100084 tashkent
de aguiar, 128 Mr S. MaSCarau tel: 99 871 140 7997
by Les Laboratoires Servier 1069-133 Lisboa Laboratorios Servier Sa
50, rue Carnot 92284 Suresnes tel: (351-21) 3122000 avenida de los Madroos 33 Venezuela
Cedex France, and printed 28043 Madrid dr e. LanGHaMer
Republic of Panama tel: (34-91) 7489630 Laboratorios Servier, S.a.
by Imprimerie Drid Mr a. PriGent Calle Sanatorio del vila
Zone Industrielle des Chanoux Servier Centro amrica y Sweden Centro empresarial Ciudad Cen-
Caribe, S.a. Mr F. texier ter, torre F, piso 3
49, rue des Frres-Lumire Costa del este Servier Sverige aB Boleta norte - Caracas 1071 a
93334 Neuilly-sur-Marne Cedex Boulevard Costa del este Frsundaviks all 1 tel: 58212718 71 00
edif. PH torre Panam 16970 Solna
Piso 25, Ciudad de Panam tel: +46 854 25 0800 Vietnam
repblica de Panam. Mr M. FitouSSi
tel.: 507.301.02.27 Switzerland Servier vietnam Company Limited
Printed in France Mr F. BenGoLd Level 11, Continental tower,
Directeur de la Publication: Romania Servier Suisse Sa 85 Ham nghi Street,
Mr S. niCoLLerat 10 rue de la Bergre nguyen thai Binh Ward,
Frdric Sesini, PharmD Servier Pharma, S-Park Case Postale 380 district 1, Ho Chi Minh City
11-15 tipografilor str. CH-1217 Meyrin 1 tel: 84 8 3823 8932
Building a1, 3rd floor, sector 1 tel: (41-22) 7378437
Bucarest code 013714 West Indies
tel: (40) 21 528 52 80 Taiwan (ROC) dr t. Monod
2016 by Les Laboratoires Servier Mr r. Lo Servier West indies Ltd
Russian Federation taiwan Servier (S) Pte Ltd Stafford House
All rights reserved throughout the world Mr J. Gavet 3rd Floor, no. 168 the Garrison, St Michael Barba-
Zao Servier Sung Chiang road dos
and in all languages. No part of this pub- 2 Paveletskaya Pl., Bt. 3 taipei 104 tel: (1-246) 4277667
lication may be reproduced, transmitted, 115054 Moscow - russia tel: 886 2 2568 1380
or stored in any form or by any means tel: (7-495) 937 07 00
Thailand
either mechanical or electronic, including Saudi Arabia Mr G. drianno
photocopying, recording, or through an dr B. renaud Servier thailand Ltd
Les Laboratoires Servier Ploenchit Center Building
information storage and Scientific office, 15th Floor, 2 Sukhumvit road
retrieval system, without the written per- 401 abdullatif Plaza, Klongtoey - Bangkok 10110
mission of the copyright holder. al Faisaliyah dist., tel: 66 2 656 8388
King Fahd St.,
Opinions expressed do not necessarily P.o. Box 112744 Tunisia
reflect the views of the publishers, Jeddah 21371 Mr F. BarBu
tel: 966 2 697 69 97 Servier tunisie
editors, or editorial board. The authors, rue du Lac de Cme
editors, and publishers cannot be held Serbia immeuble driss 4
responsible for errors or for any conse- Mr C. BroudiC Bloc a, 1er tage
Les Laboratoires Servier Les Berges du Lac
quences arising from the use of the Bulevar Mihajla Pupina 10L 1053 tunis
information contained in this journal. 11070 Belgrade tel: +216 71655 857
tel: 381 11 424 00 50
Turkey
Address for correspondence: Singapore Mr P. Mea
Servier International / Mr B. aBdiLi Servier ila ve arastirma aS
Servier (S) Pte Ltd Maslak Giz Kule
Medical Publishing Division 67 ubi avenue 1 Meydan Sokak
50, rue Carnot, #06-08 Starhub Green no 28, Kat: 22-23
92284 Suresnes Cedex FRANCE Singapore 408942 34398 Maslak, istanbul
tel: 65 6509 9811 tel: (90-212) 3291400
Tel. + 33 1 55 72 60 00 / Fax 58 84
Slovak Republic Ukraine
Mr r. aZenCotH Mr M. eSCHenBrenner
Servier Slovensko ro Les Laboratoires Servier
Spol S.r.o., Pribinova 10 24 vorovskogo Street
81109 Bratislava 4th floor - Kiev 01054
tel: 421 2 59204111 tel: 380 44 490 34 41
Vol 38, No. 3, 2016
Medicographia
128
A Ser vier publication

Metabolic agents
in the contemporary
treatment of angina
E DITORIAL
245 Metabolic agents and angina treatment
F. J. Pinto, Portugal

T HEMED ARTICLES
251 Chronic ischemic heart disease: an energy imbalance
G. Guarini, M. Marzilli, Italy

257 Mitochondria as a therapeutic target in ischemia


D. J. Hausenloy, United Kingdom

264 The place of metabolic agents in contemporary coronary


artery disease guidelines
L. H. W. Gowdak, Brazil

271 Revascularization in angina patients: addressing


cardiac metabolism challenges
G. D. Lopaschuk, Canada

277 The endothelium: a therapeutic target in post-PCI patients and the role
of trimetazidine in endothelial function
S. Lim, South Korea

282 Metabolic agents in the protection of patients who undergo


revascularization
Y. Chen, China

288 How effective can trimetazidine be in preventing myocardial and


renal revascularization injury?
Y. Lopatin, Russia

Contents continued on next page


Vol 38, No. 3, 2016
Medicographia
128
A Ser vier publication

C ONTROVERSIAL QUESTION
297 Is targeting only stenosis sufficient to optimally improve angina?
P. S. Farsky, Brazil - L. O. Go, Philippines - H. Hasan-Ali, Egypt -
H. Q. T. Ho, Vietnam - D. Isaza-Restrepo, Colombia - T. Kovarnik,
Czech Republic - O. H. Masoli, Argentina - A. N. Parkhomenko and
O. S. Gurjeva, Ukraine - C. K. Ponde, India - V. Sansoy, Turkey -
D. Vassilev, Bulgaria

VASTAREL MR
309 Trimetazidine: targeting the cardiac cell in stable angina
M. Gliebova, France

I NTERVIEW
317 The ATPCI trial: a new international phase 3 study assessing the clinical
impact of adding a metabolic agent to post-PCI angina treatment
R. Ferrari, Italy

320 F OCUS
Combining hemodynamic and metabolic agents in ischemic
heart disease
G. Fragasso, Italy

U PDATE
328 History and evolution of coronary stenting
M. E. Bertrand, France

EDITORIAL
drugs that act directly by
increasing the energy supply in
cardiac cells are of use, whatever
the causal mechanism involved,
and as such are essential for the
optimal treatment of ischemia. To
Metabolic agents
protect myocardial cells from is-
chemia, energy supply needs to
match energy demand. b-Blockers
and angina treatment
have a positive impact, reducing
energy demand, while metabolic
agents, such as trimetazidine, in-
crease energy supply. That is why
the use of an agent like trimetazi-
dine fully complements b-blocker
therapy.

b y F. J . P i n t o , Po r t u g a l

hronic ischemic heart disease (IHD) and stable angina are a major clinical

C problem worldwide. the prevalence of stable angina is estimated to be


in the region of 20 000 to 50 000 per million in the general population.1,2
According to the rEACH (rEduction of Atherothrombosis for Continued
Health) registry of more than 38 000 patients, 3 in every 20 patients with established
coronary artery disease (CAD) has had a major event or been hospitalized within the
previous year.3

Despite great advances in the management of CAD patients in recent times, symp-
toms are still common in many patients, sometimes even after revascularization.
Fausto J. PINTO, MD, PhD, In the Heart and Soul Study, over a third (38%) of outpatients with stable CAD had
FESC, FACC angina, ischemia, or both.1 A substantial number of patients with typical angina do
Department of Cardiology
University Hospital Santa not have significant coronary atherosclerotic obstructions.4 Furthermore, the preva-
Maria/CHLN, CCUL lence of coronary atherosclerotic obstruction in patients with or without typical angi-
University of Lisbon na is similar and is age-related in both sexes.
Lisbon, PortUgAL

the treatment of stable CAD includes several potential strategies, including revas-
cularization procedures (coronary artery bypass graft [CABg] or percutaneous coro-
nary intervention [PCI]) and pharmacotherapy.5 one of the potential strategies for
the treatment of CAD consists in targeting cardiac cells directly and in particular the
energetic origin of ischemia with the use of a metabolic agent, such as trimetazidine,
on top of b-blockers and other vasoactive agents.5

Myocardial ischemia is multifactorial and, above all, a deficiency


in energy
recent data clearly show that above and beyond coronary atherosclerotic obstruc-
tion, IHD is due to a large number of mechanisms, including coronary vasomotor,
microcirculatory, endothelial, and platelet dysfunction and inflammation, among oth-
ers.6 Because myocardial ischemia is multifactorial in nature, it should, above all,
Address for correspondence:
Professor Fausto J. Pinto, Servio de
be defined as a deficiency in energy (in the form of AtP, adenosine triphosphate)
Cardiologia, Hospital de Santa Maria - at the cardiac cell level.7 Under conditions of energy deficit, drugs that act directly
Centro Hospitalar de Lisboa Norte, by increasing the energy supply in cardiac cells are of use, whatever the causal mech-
E.P.E., Av. Professor Egas Moniz,
Piso 08, 1649-035 Lisboa, Portugal anism involved, and as such are essential for the optimal treatment of ischemia.
(email:
faustopinto@medicina.ulisboa.pt/
fpinto@icvl.pt)
to protect myocardial cells from ischemia, energy supply needs to match energy
demand. b-Blockers have a positive impact, reducing energy demand, while
Medicographia. 2016;38:245-250
metabolic agents, such as trimetazidine, increase energy supply. that is why the
www.medicographia.com use of an agent like trimetazidine fully complements b-blocker therapy.

Metabolic agents and angina treatment Pinto MEDICOGRAPHIA, Vol 38, No. 3, 2016 245
EDITORIAL

Trimetazidine + -blocker: an optimal combination 1000 patients with heart failure, of mainly ischemic origin (93%),
for reducing angina gao et al showed that adding trimetazidine significantly re-
the trIMPoL (trIMetazidine in PoLand) II study was one of duced all-cause mortality as well as cardiovascular events and
the first studies to test the use of a metabolic agent on top hospitalization for heart failure (P<0.01 versus placebo).13 In
of b-blockers in 426 patients with stable CAD.8 It was a ran- postmyocardial infarction patients with stable angina and
domized, multicenter, double-blind, placebo-controlled par- heart failure, the use of modified-release trimetazidine was re-
allel group study. Patients with documented CAD and stable, lated to a significant reduction in major adverse cardiac events
effort-induced angina uncontrolled by metoprolol received (cardiac death, nonfatal myocardial infarction, acute stroke,
either placebo or trimetazidine 20 mg three times daily in ad- need for coronary revascularization, and hospitalization for
dition to metoprolol 50 mg twice daily. In this study, 12 weeks unstable angina or heart failure) after 6 years of follow-up.14
treatment with trimetazidine plus metoprolol significantly im-
proved treadmill exercise test parameters and significantly re- Adding trimetazidine to decrease ischemic
duced clinical symptoms compared with placebo plus meto- reperfusion injury during revascularization and
prolol. this was achieved without any further hemodynamic angina recurrence afterwards
changes in these patients. In addition to its antianginal effica- Labrou et al have investigated whether the administration of
cy, trimetazidine was well tolerated. trimetazidine before and after PCI minimizes procedure-in-
duced myocardial damage and improves left ventricular func-
Michaelides et al performed a randomized, double-blind, con- tion 1 and 3 months after PCI.15 twenty-four hours after PCI,
trolled trial in angina patients who were symptomatic despite troponin I levels were >1 ng/mL in 26% of patients treated
treatment with propranolol.9 the trial demonstrated that adding with trimetazidine versus 44% of patients in the control group.
trimetazidine to treatment significantly decreased the mean Forty-eight hours after revascularization, troponin levels re-
number of angina attacks (63%) twice as much as adding mained elevated in 15% vs 32% of patients. About a fifth
isosorbide dinitrate (31%). this finding might be explained by (22%) of patients in the trimetazidine group had creatine ki-
the mode of action of trimetazidine, which provides a syner- nase MB (CK-MB) levels >5 ng/mL, 24 hours after PCI, com-
gistic and complementary approach to hemodynamic agents, pared with 40% in the control group.
such as b-blockers. In a recent meta-analysis of almost 20 000
angina patients, trimetazidine was shown to be as effective the number of patients with an ejection fraction <50% was
as calcium channel blockers or nitrates at reducing ischemia significantly reduced in the trimetazidine-treated group com-
and angina symptoms.10 pared with the control group at 1 and 3 months after PCI:
11% versus 16% (P=0.046) after 1 month and 4% versus
Nesukay demonstrated that directly adding trimetazidine to 16% (P=0.017) after 3 months. A significant improvement in
b-blockers in over 1400 patients with stable angina allowed for regional wall motion versus placebo was noted after treat-
a quick reduction in angina symptoms, regardless of whether ment with trimetazidine. the use of trimetazidine appeared to
or not these patients who were on b-blockers were also on minimize myocardial reperfusion injury during PCI and im-
nitrates or calcium channel blockers.11 proved global and regional wall motion 1 and 3 months after
PCI, according to the authors.
Adding trimetazidine to -blockers: evidence of
improved prognosis in ischemic patients the incidence of stent restenosis has risen, as increasing
In a recent heart failure registry, approximately 40% of chron- numbers of patients are treated with drug-eluting stents (DES).
ic heart failure patients were found to have heart failure of Chen et al16 evaluated whether long-term treatment with tri-
ischemic origin.12 In a contemporary meta-analysis in nearly metazidine reduced the incidence of stent restenosis in 768
patients who underwent PCI with DES. the group on long-
term trimetazidine treatment had a lower incidence of stent
SELECTED ABBREVIATIONS AND ACRONYMS restenosis compared with the control group (4.2% vs 11.1%;
P=0.001). At the 30-day follow-up, the trimetazidine patients
CABG coronary artery bypass graft
exhibited a higher left ventricular ejection fraction than con-
CAD coronary artery disease trol patients (65.410.7% vs 63.110.4%; P=0.006). the
DES drug-eluting stent incidence of major adverse cerebrovascular or cardiovascular
IHD ischemic heart disease events (MACCEs) was also lower in the trimetazidine group
MACCE major adverse cerebrovascular or cardiovascular event at 1-year follow-up (6.1% vs 10.8%; P=0.032). treatment
with trimetazidine was found to predict a reduction in stent
PCI percutaneous coronary intervention
restenosis (odds ratio [or], 0.376; 95% CI, 0.196 to 0.721;
REACH rEduction of Atherothrombosis for Continued Health
P=0.003). the authors concluded that trimetazidine treat-
TRIMPOL trIMetazidine in PoLand ment effectively reduced the incidence of stent restenosis and
MACCEs 1 year after DES implantation.

246 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Metabolic agents and angina treatment Pinto
EDITORIAL

Xu et al17 also appraised the effects of trimetazidine after DES Conclusion


implantation on recurrent angina pectoris and left ventricular Chronic IHD is still a significant clinical burden in daily practice,
structure in elderly patients with multivessel CAD and with di- and in stable angina b-blockers and revascularization proce-
abetes mellitus and a left ventricular ejection fraction >50%. dures are extensively used treatments. Although the current
After 2 years, patients in the trimetazidine group were shown pharmacological treatment of angina with vasoactive agents
to have significant reductions in the incidence and severity is effective, the addition to therapy of metabolic antianginal
of angina pectoris, compared with the control group, as well agents that act directly at the cardiac cell level could provide
as a reduction in silent myocardial ischemia and increase in further treatment benefits, by targeting alternative mechanisms
angina pectorisfree survival. Left ventricular function and of ischemia. Moreover, use of these metabolic agents during
structure in trimetazidine-treated patients were relatively sta- and after revascularization procedures may also offer addition-
ble after 2 years, but in control patients these parameters de- al benefits, by decreasing ischemic reperfusion injury and de-
teriorated. Adjunctive therapy with trimetazidine after DES creasing angina recurrence. targeting the cardiac cell directly
implantation appears to have a beneficial effect in preventing and addressing the energetic origin of ischemia with metabol-
recurrent angina pectoris as well as in improving left ventric- ic agentssuch as trimetazidineon top of treatment with va-
ular function and structure in elderly multivessel CAD patients soactive agentssuch as b-blockersseems a clinically per-
with diabetes. tinent strategy for managing IHD as effectively as possible.

References
1. gehi AK, Ali S, Na B, Schiller NB, Whooley MA. Inducible ischemia and the with isosorbide dinitrate-propranolol in patients with stable angina. Clin Drug
risk of recurrent cardiovascular events in outpatients with stable coronary heart Invest. 1997;13:8-14.
disease: the heart and soul study: the Heart and Soul Study. Arch Intern Med. 10. Danchin N, Marzilli M, Parkhomenko A, ribeiro JP. Efficacy comparison of tri-
2008;168:1423-1428. metazidine with therapeutic alternatives in stable angina pectoris: a network
2. Leal J, Luengo-Fernndez r, gray A, Petersen S, rayner M Economic burden meta-analysis. Cardiology. 2011;120:59-72.
of cardiovascular diseases in the enlarged European Union. Eur Heart J. 2006; 11. Nesukay E. Assessment of the most effective combination of antianginal med-
27:1610-1619. ications in the treatment of patients with stable angina pectoris. Circulation.
3. Steg Pg, Bhatt DL, Wilson PW, et al; rEACH registry Investigators. one-year 2012;125:e773. doi:10.1161/CIr.0b013e31824fcdb3. Poster P144.
cardiovascular event rates in outpatients with atherothrombosis. JAMA. 2007; 12. Maggioni AP, Dahlstrm U, Filippatos g, et al; Heart Failure Association (HFA) of
297:1197-1206. the European Society of Cardiology. EUrobservational research Programme:
4. Cheng VY, Berman DS, rozanski A, et al. Performance of the traditional age, regional differences and 1-year follow-up results of the Heart Failure Pilot Survey
sex, and angina typicality-based approach for estimating pretest probability of (ESC-HF Pilot). Eur J Heart Fail. 2013;15:808-817.
angiographically significant coronary artery disease in patients undergoing coro- 13. gao D, Ning N, Niu X, Hao g, Meng Z. trimetazidine: a meta-analysis of ran-
nary computed tomographic angiography: results from the multinational coro- domised controlled trials in heart failure. Heart. 2011;97:278-286.
nary Ct angiography evaluation for clinical outcomes: an international multi- 14. Lopatin YM, Ilyukhin oV, Ilyukhina MV, Kalganova EL, tarasov DL, Ivanenko VV.
center registry (CoNFIrM). Circulation. 2011;124:2423-2432. Long-term trimetazidine modified release therapy improves prognosis in post-
5. Montalescot g, Sechtem U, Achenbach S, et al; task Force Members. 2013 myocardial infarction patients with angina pectoris and heart failure. Eur Heart
ESC guidelines on the management of stable coronary artery disease: the task J. 2012;33(suppl abstract):346-347. Abstract 2052.
Force on the management of stable coronary artery disease of the European 15. Labrou A, giannoglou g, Zioutas D, Fragakis N, Katsaris g, Louridas g. trimeta-
Society of Cardiology. Eur Heart J. 2013;34:2949-3003. zidine administration minimizes myocardial damage and improves left ventric-
6. Crea F, Camici Pg, De Caterina r, Lanza gA. Chronic ischaemic heart disease. ular function after percutaneous coronary intervention. Am J Cardiovasc Drugs.
In: Camm AJ, Lscher tF, Serruys PW, eds. The ESC Textbook of Cardiovas- 2007;7:143-150.
cular Medicine. 2nd ed. New York, NY: oxford University Press Inc.; 2009. 16. Chen J, Zhou S, Jin J, et al. Chronic treatment with trimetazidine after dis-
7. Marzilli M, Merz CN, Boden WE, et al. obstructive coronary atherosclerosis and charge reduces the incidence of restenosis in patients who received coronary
ischemic heart disease: an elusive link! J Am Coll Cardiol. 2012;60:951-956. stent implantation: a 1-year prospective follow-up study. Int J Cardiol. 2014;
8. Szwed H, Sadowski Z, Elikowski W, et al. Combination treatment in stable ef- 174:634-639.
fort angina using trimetazidine and metoprolol: results of a randomized, double- 17. Xu X, Zhang W, Zhou Y, et al. Effect of trimetazidine on recurrent angina pec-
blind, multicentre study (trIMPoL II). trIMetazidine in PoLand. Eur Heart J. toris and left ventricular structure in elderly multivessel coronary heart disease
2001;22:2267-2274. patients with diabetes mellitus after drug-eluting stent implantation: a single-
9. Michaelides AP, Spiropoulos K, Dimopoulos K, Athanasiades D, toutouzas P. centre, prospective, randomized, double-blind study at 2-year follow-up. Clin
Antianginal efficacy of the combination of trimetazidine-propranolol compared Drug Investig. 2014;34:251-258.

Keywords: b-blocker; cardiac metabolism; myocardial ischemia; revascularization; stable angina; trimetazidine

Metabolic agents and angina treatment Pinto MEDICOGRAPHIA, Vol 38, No. 3, 2016 247
DITORIAL

les mdicaments qui agis-


sent directement en augmentant
lnergie fournie aux cellules car-
diaques sont essentiels pour trai-
ter lischmie de manire optimale.
Afin de protger les cellules myo-
cardiques de lischmie, lapport
Agents mtaboliques
et traitement de langor
en nergie doit rpondre la de-
mande. Les b-bloquants ont un
impact positif, en rduisant la de-
mande nergtique, tandis que les
agents mtaboliques augmentent
lapport en nergie. Un mdica-
ment comme la trimtazidine com-
plte donc parfaitement laction
des b-bloquants.

p a r F. J . P i n t o , Po r t u g a l

L
a cardiopathie ischmique (CI) chronique et langor stable reprsentent un
problme clinique majeur au niveau mondial. La prvalence de langor sta-
ble est estime environ 20 000 50 000 par million dans la population g-
nrale1,2. Selon le registre REACH (rEduction of Atherothrombosis for Conti-
nued Health), qui comprend plus de 38 000 patients, 3 patients sur 20 ayant une
maladie coronaire tablie ont prsent un vnement majeur ou ont t hospitali-
ss au cours de lanne prcdente3.

Malgr dimportantes avances dans la prise en charge des patients atteints de


MC au cours des dernires annes, de nombreux patients sont encore frquem-
ment symptomatiques, parfois mme aprs une revascularisation. Dans ltude
Heart and Soul, plus dun tiers des patients ambulatoires (38 %) prsentant une MC
stable avaient de langor, une ischmie ou les deux1. Un nombre important de pa-
tients prsentant un angor typique nont pas dobstruction athrosclreuse coronaire
significative4. En outre, la prvalence de lobstruction athrosclreuse coronaire chez
les patients prsentant ou non un angor typique est lie lge et similaire et dans
les deux sexes.

Le traitement de la MC stable repose sur plusieurs stratgies, notamment les pro-


cdures de revascularisation (pontage aorto-coronaire [PAC] ou intervention coro-
naire percutane [ICP]) et la pharmacothrapie5. Lune des stratgies pouvant tre
utilise dans le traitement de la MC consiste cibler les cellules cardiaques direc-
tement, et en particulier lorigine nergtique de lischmie, en utilisant un agent
mtabolique, comme la trimtazidine, en complment des b-bloquants et dautres
agents vasoactifs5.

Lischmie myocardique est multifactorielle, mais cest avant tout


un dficit en nergie
Des donnes rcentes montrent clairement quau-del de lobstruction coronaire
par lathrosclrose, la CI est provoque par un grand nombre de mcanismes, no-
tamment un dysfonctionnement vasomoteur coronaire, microcirculatoire, endoth-
lial et plaquettaire et une inflammation6. Lischmie myocardique tant de nature
multifactorielle, elle devrait avant tout tre considre comme un dficit en nergie
(sous forme dATP [adnosine triphosphate]) au niveau des cellules cardiaques7. En
conditions de dficit nergtique, les mdicaments qui agissent directement en aug-
mentant lnergie fournie aux cellules cardiaques sont utiles, quel que soit le m-
canisme causal impliqu, et sont donc essentiels pour traiter lischmie de manire
optimale.

248 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Agents mtaboliques et traitement de langor Pinto
DITORIAL

Afin de protger les cellules myocardiques de lischmie, lap- Par ailleurs, Nesukay a dmontr que lajout direct de trimta-
port en nergie doit rpondre la demande. Les b-bloquants zidine des b-bloquants chez plus de 1 400 patients atteints
ont un impact positif, en rduisant la demande nergtique, dangor stable permet une rduction rapide des symptmes
tandis que les agents mtaboliques, comme la trimtazidine, angoreux, que les patients traits par les b-bloquants aient
augmentent lapport en nergie. Un mdicament comme la reu simultanment ou non des drivs nitrs ou des inhi-
trimtazidine complte donc parfaitement laction des b-blo- biteurs calciques11.
quants.
Ajout de la trimtazidine aux b -bloquants :
Trimtazidine + b -bloquant : une association amlioration du pronostic chez les patients
optimale pour reduire langor ischmiques
Ltude TRIMPOL II (trIMetazidine in PoLand) a t lune des Lanalyse dun registre rcent de patients souffrant dinsuf-
premires tester lutilisation dun agent mtabolique en com- fisance cardiaque a montr quenviron 40 % des patients in-
plment des b-bloquants chez 426 patients atteints de MC suffisants cardiaques chroniques prsentaient une pathologie
stable8. Dans cette tude randomise, multicentrique, en dou- dorigine ischmique12. Dans une mta-analyse contempo-
ble aveugle, contrle par placebo et en groupes parallles, raine mene chez prs de 1 000 patients atteints dinsuffi-
les patients atteints de MC documente et dangor stable in- sance cardiaque, principalement dorigine ischmique (93 %),
duit par leffort, non contrls par le mtoprolol, ont reu soit Gao et coll. ont montr que laddition de trimtazidine rdui-
un placebo, soit 20 mg de trimtazidine trois fois par jour, en sait significativement la mortalit de toute cause, ainsi que
complment de deux doses quotidiennes de 50 mg de m- les vnements cardio-vasculaires et lhospitalisation pour in-
toprolol. Un traitement de 12 semaines avec lassociation tri- suffisance cardiaque (p < 0,01 vs placebo)13. Chez les patients
mtazidine plus mtoprolol a permis damliorer de manire en post-infarctus du myocarde prsentant un angor stable et
significative les paramtres des preuves deffort sur tapis une insuffisance cardiaque, un suivi de six ans a montr que
roulant et a rduit significativement les symptmes cliniques lutilisation de trimtazidine libration modifie tait lie
par rapport lassociation placebo plus mtoprolol. Ces r- une rduction significative des vnements cardiaques ma-
sultats ont t obtenus sans changements hmodynamiques jeurs (mortalit cardiaque, infarctus du myocarde non fatal,
supplmentaires chez ces patients. Outre son efficacit an- accident vasculaire crbral aigu, ncessit dune revascu-
tiangoreuse, la trimtazidine a t bien tolre. larisation coronaire et hospitalisation pour angor instable ou
insuffisance cardiaque)14.
Michaelides et coll. ont ralis une tude contrle, randomi-
se et en double aveugle chez des patients atteints dangor, Ajout de la trimtazidine pour diminuer
encore symptomatiques malgr un traitement par le propra- les lsions de reperfusion ischmique au cours
nolol9. Cette tude a dmontr que lajout de trimtazidine de la revascularisation et les rcidives dangor
permet de diminuer significativement le nombre moyen de par la suite
crises dangor de manire deux fois plus efficace que lajout du Labrou et coll. ont cherch dterminer si ladministration de
dinitrate disosorbide (63 % vs 31 %). Ce rsultat pourrait trimtazidine avant et aprs une ICP diminue les lsions myo-
sexpliquer par le mode daction de la trimtazidine, qui est cardiques induites par la procdure et amliore la fonction ven-
synergique et complmentaire celui des agents hmodyna- triculaire gauche 1 et 3 mois aprs une ICP15. Vingt-quatre
miques comme les b-bloquants. Dans une rcente mta-ana- heures aprs lICP, 26 % des patients traits par trimtazidine
lyse portant sur prs de 20 000 patients angoreux, la trimta- avaient un taux de troponine I > 1 ng/ml, contre 44 % dans le
zidine sest avre aussi efficace que les inhibiteurs calciques groupe tmoin. Quarante-huit heures aprs la revascularisation,
ou les drivs nitrs pour rduire les symptmes dischmie les taux de troponine restaient levs chez 15 % du groupe
et dangor10. trimetazidine, contre 32 % dans le groupe tmoin. Environ un
cinquime des patients (22 %) du groupe trimtazidine prsen-
taient des taux de cratine kinase MB (CK-MB) > 5 ng/ml,
SELECTED ABBREVIATIONS AND ACRONYMS
24 heures aprs lICP, contre 40 % dans le groupe tmoin. Le
CI cardiopathie ischmique nombre de patients montrant une fraction djection < 50 %
ICP intervention coronaire percutane a diminu de manire significative dans le groupe trait par
MACCE major adverse cerebrovascular or cardiovascular la trimtazidine, par rapport au groupe tmoin : 11 % vs 16 %
event (vnement indsirable vasculaire crbral (p = 0,046) 1 mois aprs lICP, et 4 % vs 16 % (p = 0,017) 3
ou cardio-vasculaire majeur)
mois aprs lICP. Une amlioration significative de la motilit
MC maladie coronaire
paritale locale a t observe aprs le traitement par la tri-
REACH REduction of Atherothrombosis for Continued
Health (rduction de lathrothrombose pour
mtazidine par rapport au placebo. Daprs les auteurs, luti-
le maintien de la sant) lisation de la trimtazidine semble minimiser les lsions myo-
TRIMPOL TRIMetazidine in POLand (trimtazidine en Pologne) cardiques de reperfusion au cours de lICP et amliorer la
motilit paritale globale et rgionale 1 et 3 mois aprs lICP.

Agents mtaboliques et traitement de langor Pinto MEDICOGRAPHIA, Vol 38, No. 3, 2016 249
DITORIAL

Le nombre de patients traits par stents actifs (endoprothses cardique silencieuse et une augmentation de la survie sans
libration de principe actif) ayant augment, lincidence des angor ont t observes chez les patients du groupe trim-
restnoses intra-stent est en progression. Chen et coll.16 ont tazidine par rapport au groupe tmoin. La fonction et la struc-
valu dans quelle mesure un traitement long terme par la ture du ventricule gauche chez les patients traits par la tri-
trimtazidine pouvait rduire lincidence des restnoses in- mtazidine sont restes relativement stables aprs deux ans,
tra-stent chez 768 patients ayant bnfici dune ICP avec un alors que chez les patients tmoins ces paramtres se sont
stent actif. Lincidence des restnoses intra-stent tait inf- dtriors. Un traitement complmentaire par la trimtazidine
rieure dans le groupe ayant reu un traitement long terme aprs la mise en place dun stent actif semble exercer un ef-
par la trimtazidine par rapport au groupe tmoin (4,2 % vs fet bnfique sur la prvention des rcidives dangor, et sur
11,1 % ; p = 0,001). Aprs 30 jours de suivi, la fraction djec- lamlioration de la fonction et de la structure du ventricule
tion ventriculaire gauche des patients sous trimtazidine tait gauche chez des patients gs diabtiques prsentant une
suprieure celle des patients du groupe tmoin (65,4 MC pluritronculaire.
10,7 % vs 63,1 10,4 % ; p = 0,006). Lincidence des v-
nements vasculaires crbraux ou cardio-vasculaires majeurs Conclusion
(major adverse cerebrovascular or cardiovascular events, La CI chronique reste une charge clinique significative en
MACCE) tait galement infrieure dans le groupe recevant pratique courante et, dans langor stable, les b-bloquants et
la trimtazidine lors du suivi un an (6,1 % vs 10,8 % ; p = la revascularisation sont des traitements largement utiliss.
0,032). Le traitement par la trimtazidine sest avr tre un Bien que le traitement pharmacologique actuel de langor par
facteur prdictif de la rduction des restnoses intra-stent des agents vasoactifs savre efficace, laddition dagents an-
(odds ratio, [OR] : 0,376 ; intervalle de confiance [IC] 95 % : tiangoreux mtaboliques agissant directement au niveau des
0,196 0,721 ; p = 0,003). Les auteurs ont conclu quun trai- cellules cardiaques pourrait apporter des bnfices thrapeu-
tement par la trimtazidine rduisait de manire efficace lin- tiques supplmentaires, en ciblant dautres mcanismes de
cidence des restnoses intra-stent et des MACCE un an aprs lischmie. En outre, lutilisation de ces agents mtaboliques
la mise en place dun stent actif. pendant et aprs les procdures de revascularisation aurait
galement lavantage de diminuer les lsions de reperfusion
Xu et coll. ont galement valu les effets de la trimtazidine post-ischmiques et de rduire les rcidives dangor. La stra-
aprs la mise en place dun stent actif, sur la rcidive de lan- tgie consistant cibler directement les cellules cardiaques
gor et sur la structure du ventricule gauche chez des patients et traiter lorigine nergtique de lischmie par des agents
gs atteints de MC pluritronculaire et dun diabte, et pr- mtaboliques comme la trimtazidine en complment des
sentant une fraction djection ventriculaire gauche > 50 %17. agents vasoactifs comme les b-bloquants semble clinique-
Aprs deux ans, des rductions significatives de lincidence ment pertinente pour prendre en charge la CI de la manire
et de la svrit de langor, une diminution de lischmie myo- la plus efficace possible.

250 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Agents mtaboliques et traitement de langor Pinto

M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

The unexpected prevalence


of angina despite optimal medical
therapy plus successful revascu-
larization strongly challenges the
current approach to treating is-
chemic heart disease. Up to this
Chronic ischemic heart
point, cardiologists have focused
on the vascular inability to supply
myocytes with sufficient oxygen
disease: an energy imbalance
and nutrients. Only recently has the
scientific community considered
additional mechanisms that may
contribute to myocardial ischemia.

by G. Guarini and M. Marzilli, Italy

A
lterations in cardiac metabolism have recently been implicated in the
pathophysiology of ischemic heart disease. In normal conditions, the
heart derives most of its energy from b -oxidation of free fatty acids.
However, the healthy heart can easily switch from one substrate to another
according to substrate availability, nutritional status, and exercise level. Par-
adoxically, during prolonged and severe ischemia, the myocardium continues
to derive most of its energy (up to 90%) from b -oxidation. A greater amount
of oxygen is required to completely oxidize a fatty acid with a carbon-chain
length equivalent to that of glucose. Fatty acid oxidation is thought to be detri-
mental in that, while requiring more oxygen, it produces less adenosine tri-

Mario MARZILLI, MD phosphate (ATP) and more reactive oxygen species, thus further reducing mi-
Giacinta GUARINI, MD, PhD tochondrial respiratory efficiency. Aside from metabolic alterations, the process
Cardiovascular Medicine Division of producing and utilizing energy is very complex and includes multiple steps
Cardio Thoracic and Vascular from uptake of metabolites by cardiac myocytes, oxidative phosphorylation in
Department, University of Pisa the mitochondria, and transport of ATP to intracellular components. There-
ITALY
fore, impairment in any one of these steps can have a tremendous impact on
cell homeostasis. In addition to acute and chronic changes in cardiac metab-
olism, mitochondrial dysfunction has been implicated in promoting myocar-
dial ischemia and myocardial damage during the reperfusion phase of an is-
chemic event, thereby further reducing the hearts ability to synthesize and
utilize ATP.
Medicographia. 2016;38:251-256 (see French abstract on page 256)

espite a global reduction in cardiovascular mortality owing to improved

D therapy and well-designed educational programs, ischemic heart disease


(IHD) remains the most important cause of death in Western countries. In
the United States, in the 10-year period from 2003 to 2013, death rates attribut-
able to cardiovascular disease (CVD) declined by 28.8%, while the actual number
of CVD deaths per year declined by 11.7%. Even so, CVD still accounted for 30.8%
of all deaths (ie, 1 out of every 3 deaths) in the United States in 2013. That same
time period witnessed a decline in the prevalence of angina pectoris: between 2009
Address for correspondence:
Professor Mario Marzilli, and 2012, there was an average of 3.4 million people aged 40 years or over in the
Cardiovascular Medicine Division, United States with angina each year compared with 4 million between 1988 and
Cardio Thoracic and Vascular
Department, University of Pisa, Via
1994.1 In Europe, as in the United States, the prevalence of angina increases with
Paradisa n 2, 56100 Pisa, Italy. age, with prevalence ranging from 2%-5% in men aged 45-54 years to 11%-20%
(email: mario.marzilli@med.unipi.it) in men aged over 60; in women, from 0.5%-1% to 10%-14%, respectively.2
www.medicographia.com

Chronic ischemic heart disease: an energy imbalance Guarini and Marzilli MEDICOGRAPHIA, Vol 38, No. 3, 2016 251
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

Myocardial ischemia is often due to coronary atherosclerotic gines receiving system to generate external work. Indeed,
disease, which limits coronary blood flow, causing an imbal- free fatty acids (FFAs) and glucose that are taken up by the
ance between available blood supply and the hearts meta- cells need to be transformed into intermediary components
bolic demands. Anti-ischemic therapy is based on this con- (acetyl coenzyme A [CoA]) by b-oxidation and glycolysis so
cept and focuses on alleviating the problem by removal of the that they can enter the Krebs cycle and produce carbon diox-
coronary obstructions by mechanical means, and/or modu- ide (CO2) and nicotinamide adenine dinucleotide (NADH), a
lating cardiac work and coronary blood flow through pharma- key substrate of oxidative phosphorylation. Respiratory-chain
cological agents. Although such therapeutic strategies aim complexes I through IV transfer electrons from NADH to oxy-
to restore an adequate supply/demand balance, to improve gen, creating a proton electrochemical gradient (Dm H+) across
symptoms, and to prolong survival, available evidence indi- the inner mitochondrial membrane. This gradient is used by
cates that this goal is not always reached. Indeed, a number ATP synthase to phosphorylate adenosine diphosphate (ADP),
of trials report persistent angina in over 30% of patients de- thereby producing the high-energy phosphate compound ATP,
spite optimal medical therapy and despite successful coro- the direct source of energy for all energy-consuming reac-
nary revascularization, both in patients treated by percuta- tions in the heart. Once generated in the mitochondria, ATP is
neous coronary intervention and those treated by coronary transferred by the creatine kinase energy shuttle to myofibrils
artery bypass graft (CABG) surgery.3-5 and to sarcolemmal and sarcoplasmic reticulum ion pumps.
On the basis of these considerations and an overwhelming
The unexpected prevalence of angina despite optimal med- body of evidence, factors other than epicardial stenosis, such
ical therapy plus successful revascularization strongly chal- as mitochondrial dysfunction and metabolic derangement,
lenges the current approach to treating IHD. Up to this point, have been recognized as pathological mechanisms for per-
cardiologists have focused on the vascular inability to supply sistent ischemia.6,7
myocytes with sufficient oxygen and nutrients. Only recently
has the scientific community considered additional mecha- Due to the complex pathophysiology of IHD, some challeng-
nisms that may contribute to myocardial ischemia. Just as ing questions about treatment arise. How should we deal with
there may be multiple paths to a dysfunctional car engine, persistent angina in patients that have already been revas-
there may be multiple paths to myocardial ischemia. In this cularized? Which drugs can be used to treat IHD in patients
analogy, an inadequate blood supply for the heart would be free of coronary stenosis? Indeed, most available antianginal
like a shortage of gasoline for a car; an impaired cellular up- drugs were developed to counteract the effects of a flow-lim-
take of nutrients would be like an altered transmission of gaso- iting stenosis, and their efficacy has been attributed to their
line to the car engine; mitochondrial dysfunction, like an en- ability to either increase coronary blood flow or to decrease
gines inability to transform chemical energy into mechanical myocardial oxygen demand. None of these agents were test-
energy; and an inability to transfer adenosine triphosphate (ATP) ed after the removal of the flow-limiting stenosis. The incom-
to the cellular contractile machinery, like an inability of the en- plete success with current treatment has fostered a large
interest in therapeutic strategies that target the alternative
pathological mechanisms, ie, metabolic modulation. Indeed,
SELECTED ABBREVIATIONS AND ACRONYMS
there is evidence that metabolic modulation therapy may play
Ca2+ calcium a key role in the acute phase of ischemic events, where it
CHF congestive heart failure would affect results of acute interventions on the subsequent
CoA coenzyme A development of heart failure (HF)stunned and hibernated
CVD cardiovascular disease myocardiumas well as for those who experience chronic
DNA deoxyribonucleic acid stable angina.8 Our improved understanding of metabolic
FA fatty acid changes that occur during ischemic events and after reper-
FFA free fatty acid fusion is now being translated into new therapeutic oppor-
HF heart failure tunities.
IHD ischemic heart disease
MPTP mitochondrial permeability transition pore Ischemic heart disease: an energy crisis
NADH nicotinamide adenine dinucleotide Significant progress has been made in recent years in under-
RAAS renin-angiotensin-aldosterone system standing the role of cardiac energy metabolism in the patho-
RISK reperfusion injury salvage kinase [pathway] genesis of myocardial ischemia. As a natural consequence, a
ROS reactive oxygen species better understanding of the metabolic derangements asso-
SAFE survival activating factor enhancement [pathway] ciated with IHD is translating into new therapeutic strategies.
SNS sympathetic nervous system
TACT Trimetazidine in Angina Combination Therapy Under normal conditions, the healthy heart derives approx-
TNF tumor necrosis factor imately two-thirds of its energy (in the form of ATP) from the
FFA pathway; glucose oxidation and pyruvate are the other

252 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Chronic ischemic heart disease: an energy imbalance Guarini and Marzilli
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

source for the remainder of the energy produced. The healthy logical exacerbation of insulin resistance (eg, by diuretics). Of
heart switches easily from one substrate to another as need- these, neurohumoral activation has been the most studied
ed, according to substrate availability, nutritional status, and and is probably the strongest contributor to altered metabo-
exercise level. The myocardium responds to mild-to-mod- lism in HF. Neurohumoral homeostasis is activated in response
erate cardiac ischemia by increasing uptake of glucose so to a long-term depression in cardiac outputcharacterized by
that it can produce the ATP necessary to maintain ionic gra- persistent activation of the SNS and the interlinked RAAS
dients and calcium (Ca2+) homeostasis. Paradoxically and to resulting in increased catecholamine secretion. At the same
detrimental effect, the myocardium continues to rely on b-ox- time, catecholamine reuptake in the heart is decreased. In-
idation for production of most of its energy (90%) during pro- creased levels of catecholamines are directly detrimental to
longed and severe ischemia, despite the elevated lactate pro- the heart, causing substantial enzyme loss as an index of dif-
duction that occurs under these conditions. Furthermore, due fuse myocardial damage, and much oxygen wastage even
to the Randle phenomenona competitive interaction be- in the absence of FFAs in the perfusate. Furthermore, the cat-
tween fatty acid (FA) oxidation and glucose oxidationhigh echolamine norepinephrine promotes both coronary vaso-
rates of FA oxidation inhibit glucose oxidation, already low, constriction and increased plasma FFA levels, further exacer-
even further. Although the complete oxidation of FAs produces bating oxygen wastage. Therefore, addressing the abnormal
more ATP per molecule of CO2 than that produced from the cardiac metabolism in IHD patients may also improve patient
complete oxidation of glucose, more oxygen is used to com- prognosis by halting the progression to HF.
pletely oxidize a FA of equivalent carbon-chain length. There-
fore, glucose oxidation, which produces roughly 15% more Ischemic heart disease: a mitochondrial issue
ATP for a given amount of oxygen used, is considered more Aside from their key role in energy production and metabolic
oxygen sparing than FA oxidation. During ischemia, FA oxi- modulation, mitochondria are essential to cardiomyocyte sur-
dation can become detrimental, because it uses more oxygen vival during ischemia and reperfusion. They are implicated
and produces less ATP and more reactive oxygen species in ATP synthesis, maintenance of Ca2+ homeostasis, cell sur-
(ROS), and so further depresses mitochondrial respiratory vival, and cardioprotection, all of which are regulated by the
efficiency. FFAs promote their own uptake and oxidation and proton gradient across the mitochondrial membrane. Under
they antagonize the uptake of glucose, lactate, and pyruvate, aerobic physiologic conditions, mitochondria are not involved
in part through direct inhibition of pyruvate dehydrogenase. in the beat-to-beat regulation of cytosolic Ca2+ levels, though
The effects of FFAs on the mitochondria include uncoupling a small flux of Ca2+ into the mitochondrial matrix has been ob-
of cellular respiration, resulting in decreased ATP production served. Small increases in mitochondrial Ca2+ concentration
and oxygen wasting. Thus, excessive levels of FFAs in the stimulate the Krebs cycle and the NADH redox potential.
blood lead to lactate and proton accumulation, lowered cel- This fine regulation of mitochondrial Ca2+ is important to en-
lular pH, and disrupted cellular function, as well as impaired hance oxidative phosphorylation and ATP synthesis. Howev-
Ca2+ handling, oxidative stress, reduced activity of the glucose er, under pathological conditions, the mitochondria can take
transporter GLUT-4, and apoptosis of myocytes.9 Such meta- up too much Ca2+, activating a series of steps that trigger a
bolic changes disrupt cell homeostasis and alter membrane vicious cycle that ultimately leads to irreversible cell damage.
structure, and they ultimately lead to cell death. During ischemia, intracellular Ca2+ homeostasis is deranged;
however, mitochondria can still buffer cytosolic Ca2+, suggest-
Interestingly, derangements in myocardial energy metabolism ing that they do not lose their ability to pump Ca2+. Mitochon-
are associated with heart failure (HF) as well10; such altered dria isolated after prolonged periods of ischemia are still able
metabolism is the final common pathway of several cardiac to use oxygen for ATP phosphorylation. Conversely, mitochon-
disorders, such as IHD, cardiomyopathies, hypertension, and dria isolated after reperfusion are structurally altered; their mem-
diabetes-induced HF. Recent data suggest that HF may it- brane pores are open; they contain large amounts of Ca2+;
self promote metabolic changes, such as insulin resistance, they produce large amounts of oxygen free radicals, and the
in part through neurohumoral activation, generating a vicious oxidative phosphorylation system is irreversibly damaged. In
cycle in which metabolic abnormalities further aggravate and addition, ischemia followed by reperfusion induces irreversible
precipitate HF. The associations between altered energy me- deletions in several parts of the mitochondrial genome, im-
tabolism, insulin resistance, and HF may be explained by the pairing ATP production, which is ultimately responsible for car-
following compatible processes: (i) activation of the neuro- diomyocyte death (ischemia-reperfusion injury).
humoral system, including the sympathetic nervous system
(SNS) and the renin-angiotensin-aldosterone system (RAAS); Notably, strategies that confer cardioprotection from myocar-
(ii) inflammation, indicated by increased levels of tumor necro- dial ischemia-reperfusion injury involve the activation of the
sis factor a (TNF-a) and its soluble receptors; (iii) alterations in reperfusion injury salvage kinase (RISK) and survival activat-
skeletal muscle function and mass as a result of reduced phys- ing factor enhancement (SAFE) pathways and the inhibition
ical activity; (iv) endothelial dysfunction; (v) increased adipocy- of mitochondrial permeability transition pore (MPTP) opening.
tokines, such as adiponectin and leptin; and (vi) pharmaco- The MPTP is a nonselective channel located on the inner mi-

Chronic ischemic heart disease: an energy imbalance Guarini and Marzilli MEDICOGRAPHIA, Vol 38, No. 3, 2016 253
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

tochondrial membrane. When this channel is open, the mi- enzymes and the activity of two membrane-bound pumps
tochondrial membrane potential collapses, uncoupling ox- considered to be survival promotingthe sodium-potassi-
idative phosphorylation; this results in the depletion of ATP um ATPase and the Ca2+-uptake pump of the sarcoplasmic
and cell death.11 The MPTP remain closed during myocar- reticulum. Indeed, ischemia-induced derangement of cardiac
dial ischemia and they open only during the first few min- metabolism can be minimized through treatment with meta-
utes of myocardial reperfusion in response to mitochondrial bolic modulators that decrease FA oxidation and increase uti-
Ca2+ overload, oxidative stress, and restoration of physiologic lization of glucose and lactate as energy substrates. The great-
pH.12 The RISK pathway13 involves the protein kinases Akt and est progress in the use of metabolic therapy occurred with
Erk 1 and 2; the SAFE pathway involves activation of TNF-a the advent of the direct inhibitors of myocardial FA oxidation,
and the signal transducer and activator of transcription 3 specifically trimetazidine, discussed next in further detail.19
(STAT3). When specifically activated, these pathways confer
powerful cardioprotection against lethal reperfusion injury.14 u Trimetazidine
Thus, from one perspective, the RISK and SAFE pathways Trimetazidine was the first and, for many years, the only reg-
could be considered to mediate a form of programmed cell istered drug in its class. It is available in over 80 countries
survival. There is extensive evidence that pharmacological or worldwide. It has an established antianginal efficacy, known
mechanical activation of these two pathways, via ischemic even before the discovery of how the drug acts, which is via
preconditioning or postconditioning for example, may reduce partial inhibition of myocardial FA oxidation.19,20 Initial preclinical
myocardial infarct size by up to 50%.15 The cardioprotective studies in animal models of myocardial ischemia and reper-
role of these pathways is believed to be due to inhibition of fusion demonstrated a cytoprotective effect for this drug.21
MPTP opening,16 improved mitochondrial Ca2+ handling,17 and It has been shown by Kantor et al to specifically inhibit the
recruitment of antiapoptotic pathways. Mitochondria offer sev- long-chain activity of the enzyme 3-ketoacyl CoA thiolase
eral potential targets for cardioprotective therapies. These in- (EC 2.3.1.16) (3-KAT),22 the enzyme that catalyzes the last
clude the following: (i) prevention of Ca2+ overload; (ii) pre- step in FA b-oxidation, using long-chain 3-ketoacyl-CoA as a
vention of ROS generation; and (iii) activation of the ATP- substrate to generate acetyl-CoA. Trimetazidines inhibition of
dependent potassium channels (KATP channels) that maintain 3-KAT reduces the NADH/NAD+ and acetyl-CoA/free CoA ra-
the inner mitochondrial membrane integrity, leading to (iv) pre- tios in the mitochondrial matrix, in effect removing the inhibi-
vention of the opening of the nonspecific MPTP complex.18 tion on pyruvate dehydrogenase and thus increasing the rate
of glucose oxidation. Indeed, in the working rat heart, although
Innovative approaches to manage myocardial only modestly reducing the rate of FA oxidation, trimetazidine
ischemia: mitochondria and cardiac energy significantly increases the rate of glucose oxidation.22,23 Tri-
metabolism modulators metazidines efficacy in refractory angina has been demon-
On the basis of this biochemical background, the pharma- strated in clinical trials, which also support the superior ben-
cological manipulation of mitochondria to optimize cardiac efit associated with the addition of this metabolic agent to
energy metabolism makes for an attractive therapeutic option. classic hemodynamic drug therapy, such as b-blockers or
Such an approach is largely based on the promotion of car- nitrates. The efficacy and acceptability of trimetazidine in com-
diac glucose oxidation along with the suppression of b-ox- bination with hemodynamic agents was tested in the TACT
idation, leading to an improvement in cardiac function and study (Trimetazidine in Angina Combination Therapy).24 In that
protection against ischemia-reperfusion injury, as well as at- study, exercise stress test parameters and angina symptoms
tenuated progression to congestive HF (CHF). Owing to the were significantly improved with the addition of trimetazidine
Randle phenomenon, carbohydrate metabolism may be in- to therapy including b-blockers or long-acting nitrates, com-
directly increased by a decreasing rate of FA oxidation. Such pared with addition of placebo. Similar results were observed
a decrease in FA oxidation may be achieved in different ways. in the VASCO-Angina study. This randomized, double-blind,
One of these involves decreasing the availability of FAs as placebo-controlled trial, assessed antianginal efficacy on ex-
an energy substrate; this can be achieved through treatment ercise test parameters and safety of both a standard dosage
with glucose, insulin, and potassium (GIK therapy), which de- (70 mg/day) and a high dosage (140 mg/day) of modified-re-
creases the circulating levels of FFAs and/or their uptake by lease trimetazidine in symptomatic and asymptomatic patients
cardiac myocytes, or through suppression of carnitine palmi- with chronic stable angina who were receiving background
toyl transferase (CPT) I or II to inhibit FA uptake by the mito- b-blocker therapy with atenolol (50 mg/day).25 That study con-
chondria. Another way to decrease FA oxidation is by direct firmed the efficacy and tolerability of both trimetazidine dos-
inhibition of the enzymes involved. Of note, drugs that can ages in improving effort-induced myocardial ischemia and
manipulate FFA oxidation (eg, trimetazidine) have been shown functional capacity in such patients.25 Furthermore, evidence
not only to provide cardioprotection in the acute phase of an from other studies suggest trimetazidine may improve clinical
ischemic event, but also to ameliorate cardiac metabolism manifestation in patients with stable IHD. Indeed, with long-
and angina symptoms in patients with IHD with long-term use. term administration of trimetazidine, the following have been
These findings are supported by a link between key glycolytic observed: a lower average number of weekly attacks, a low-

254 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Chronic ischemic heart disease: an energy imbalance Guarini and Marzilli
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

er mean weekly consumption of short-acting nitrates, im- Conclusions: ischemic heart disease,
provement in quality of life, lessened severity of main clinical an energetic disorder
manifestations of chronic HF, and improved (lowered) func- Historically, IHD has been considered a vascular disease, where
tional class.26-29 Moreover, as trimetazidine has been demon- coronary atherosclerosis causes an imbalance between blood
strated to have similar efficacy in men and women, this met- supply and demand. However, recent evidence suggests that
abolic myocardial cytoprotector can be recommended for cardiac metabolic derangement and the inability of mitochon-
patients with IHD irrespective of sex.30,31 Trimetazidine has also dria to efficiently produce energy are able to induce energy
been used for cardioprotection in patients undergoing coro- starvation similar to that produced by coronary blood flow
nary bypass surgery and percutaneous coronary interven- blockage. In other words, it is conceivable that cardiac meta-
tion.28,32 Consistent with IHD and HF being considered ener- bolic derangements and/or mitochondrial dysfunction can di-
getic disorders, trimetazidine was effective in reducing mortality rectly put the myocardium under ischemic conditions, inde-
and event-free survival in patients with chronic HF in an in- pendently of oxygen and nutrient availability. Accordingly, in an
ternational, multicenter, retrospective cohort study. The ad- experimental model (Zucker obese fatty rat), repairing mito-
dition of trimetazidine to optimal medical therapy improved chondrial DNA damage improved mitochondrial function, re-
long-term survival in these patients.33 That retrospective analy- stored vascular and myocyte properties, and reduced the
sis further confirmed the results of previous small studies in consequences of oxidative stress.35 Similarly, in Zucker lean
patients with chronic HF that had shown that trimetazidine rats in which mitochondrial dysfunction was selectively in-
improves left ventricular function, exercise capacity, and New duced through mitochondrial DNA damage, areas of myocar-
York Heart Association functional class compared with place- dial ischemia, endothelial dysfunction, and depressed con-
bo. Furthermore, the addition of trimetazidine to exercise train- tractile function under cardiac stress were observed in the
ing resulted in greater improvements in functional capacity, absence of coronary atherosclerosis.35 These observations
left ventricular ejection fraction, and endothelium-dependent support the hypothesis that myocardial ischemia should be
dilation in patients with chronic HF.34 considered an energetic disorder. n

References
1. Mozaffarian D, Benjamin EJ, Go AS, et al; American Heart Association Statis- tor signaling in limiting lethal reperfusion injury? Trends Cardiovasc Med. 1999;
tics Committee and Stroke Statistics Subcommittee. Heart disease and stroke 9(8):245-249.
statistic2015 update: a report from the American Heart Association. Circu- 15. Guarini G, Huqi A, Capozza P, Morrone D, Donati F, Marzilli M. Therapy against
lation. 2014;131(4):e29-e322. ischemic injury. Curr Pharm Des. 2013;19(25):4597-4621.
2. Fox K, Garcia MA, Ardissino D, et al; Task Force on the Management of Stable 16. Davidson SM, Hausenloy D, Duchen MR, Yellon DM. Signalling via the reper-
Angina Pectoris of the European Society of Cardiology; ESC Committee for fusion injury signalling kinase (RISK) pathway links closure of the mitochon-
Practice Guidelines (CPG). Guidelines on the management of stable angina pec- drial permeability transition pore to cardioprotection. Int J Biochem Cell Biol.
toris: executive summary. Eur Heart J. 2006;27(11):1341-1381. 2006;38(3):414-419.
3. Boden WE, O'Rourke RA, Teo KK, et al. Optimal medical therapy with or with- 17. Abdallah Y, Gkatzoflia A, Gligorievski D, et al. Insulin protects cardiomyocytes
out PCI for stable coronary disease. N Engl J Med. 2007;356(15):1503-1516. against reoxygenation-induced hypercontracture by a survival pathway tar-
4. Writing Group for the Bypass Angioplasty Revascularization Investigation (BARI) geting SR Ca2+ storage. Cardiovasc Res. 2006;70(2):346-353.
Investigators. Five-year clinical and functional outcome comparing bypass sur- 18. Di Lisa F, Canton M, Carpi A, et al. Mitochondrial injury and protection in ischemic
gery and angioplasty in patients with multivessel coronary disease. A multi- pre- and postconditioning. Antioxid Redox Signal. 2011;14(5):881-891.
center randomized trial. JAMA. 1997;277(9):715-721. 19. Stanley WC. Partial fatty acid oxidation inhibitors for stable angina. Expert Opin
5. Henderson RA, Pocock SJ, Clayton TC, et al. Seven-year outcome in the RITA-2 Investig Drugs. 2002;11(5):615-629.
trial: coronary angioplasty versus medical therapy. J Am Coll Cardiol. 2003;42 20. McClellan KJ, Plosker GL. Trimetazidine. A review of its use in stable angina
(7):1161-1170. pectoris and other coronary conditions. Drugs. 1999;58(1):143-157.
6. Marzilli M, Merz CN, Boden WE, et al. Obstructive coronary atherosclerosis and 21. Vaillant F, Tsibiribi P, Bricca G, et al. Trimetazidine protective effect against is-
ischemic heart disease: an elusive link! J Am Coll Cardiol. 2012;60(11):951- chemia-induced susceptibility to ventricular fibrillation in pigs. Cardiovasc Drugs
956. Ther. 2008;22(1):29-36.
7. Pepine CJ, Douglas PS. Rethinking stable ischemic heart disease: is this the 22. Kantor PF, Lucien A, Kozak R, Lopaschuk GD. The antianginal drug trimetazi-
beginning of a new era? J Am Coll Cardiol. 2012;60(11):957-959. dine shifts cardiac energy metabolism from fatty acid oxidation to glucose ox-
8. Wolff AA, Rotmensch HH, Stanley WC, Ferrari R. Metabolic approaches to the idation by inhibiting mitochondrial long-chain 3-ketoacyl coenzyme A thiolase.
treatment of ischemic heart disease: the clinicians' perspective. Heart Fail Rev. Circ Res. 2000;86(5):580-588.
2002;7(2):187-203. 23. Lopaschuk GD. Optimizing cardiac energy metabolism: how can fatty acid and
9. Ashrafian H, Frenneaux MP, Opie LH. Metabolic mechanisms in heart failure. carbohydrate metabolism be manipulated? Coron Artery Dis. 2001;12(suppl
Circulation. 2007;116(4):434-448. 1):S8-S11.
10. Neubauer S. The failing heartan engine out of fuel. N Engl J Med. 2007;356 24. Chazov EI, Lepakchin VK, Zharova EA, et al. Trimetazidine in Angina Combina-
(11):1140-1151. tion Therapythe TACT study: trimetazidine versus conventional treatment
11. Hausenloy DJ, Yellon DM. New directions for protecting the heart against is- in patients with stable angina pectoris in a randomized, placebo-controlled,
chaemia-reperfusion injury: targeting the reperfusion injury salvage kinase (RISK)- multicenter study. Am J Ther. 2005;12(1):35-42.
pathway. Cardiovasc Res. 2004;61(3):448-460. 25. Vitale C, Spoletini I, Malorni W, Perrone-Filardi P, Volterrani M, Rosano GM.
12. Griffiths EJ, Halestrap AP. Mitochondrial non-specific pores remain closed dur- Efficacy of trimetazidine on functional capacity in symptomatic patients with sta-
ing cardiac ischaemia, but open upon reperfusion. Biochem J.1995;307 (pt 1): ble exertional angina The VASCO-angina study. Int J Cardiol. 2013;168(2):
93-98. 1078-1081.
13. Zhao ZQ, Morris CD, Budde JM, et al. Inhibition of myocardial apoptosis re- 26. Grabczewska Z, Bialoszynski T, Szymanski P, et al. The effect of trimetazidine
duces infarct size and improves regional contractile dysfunction during reper- added to maximal anti-ischemic therapy in patients with advanced coronary
fusion. Cardiovasc Res. 2003;59(1):132-142. artery disease. Cardiol J. 2008;15(4):344-350.
14. Yellon DM, Baxter GF. Reperfusion injury revisited: is there a role for growth fac- 27. Marzilli M. Cardioprotective effects of trimetazidine: a review. Curr Med Res

Chronic ischemic heart disease: an energy imbalance Guarini and Marzilli MEDICOGRAPHIA, Vol 38, No. 3, 2016 255
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

Opin. 2003;19(7):661-672. jury and reperfusion in coronary artery bypass grafting. Arq Bras Cardiol. 2011;
28. Danchin N, Marzilli M, Parkhomenko A, Ribeiro JP. Efficacy comparison of 97(3):209-216.
trimetazidine with therapeutic alternatives in stable angina pectoris: a network 33. Fragasso G, Rosano G, Baek SH, et al. Effect of partial fatty acid oxidation
meta-analysis. Cardiology. 2011;120(2):59-72. inhibition with trimetazidine on mortality and morbidity in heart failure: results
29. Marzilli M. Does trimetazidine prevent myocardial injury after percutaneous coro- from an international multicentre retrospective cohort study. Int J Cardiol. 2013;
nary intervention? Nat Clin Pract Cardiovasc Med. 2008;5(1):16-17. 163(3):320-325.
30. Danchin N. Clinical benefits of a metabolic approach with trimetazidine in revas- 34. Belardinelli R, Lacalaprice F, Faccenda E, Volpe L. Trimetazidine potentiates the
cularized patients with angina. Am J Cardiol. 2006;98(5A):8J-13J. effects of exercise training in patients with ischemic cardiomyopathy referred
31. Vasiuk IuA, Shal'nova SA, Shkol'nik EL, Kulikov KG. The (PRIMA) Study. Com- for cardiac rehabilitation. Eur J Cardiovasc Prev Rehabil. 2008;15(5):533-540.
parison of clinical effect of trimetazidine MR in men and women [in Russian]. 35. Guarini G, Kiyooka T, Ohanyan V, et al. Impaired coronary metabolic dilatation
Kardiologiia. 2011;51(6):11-15. in the metabolic syndrome is linked to mitochondrial dysfunction and mito-
32. Martins GF, Siqueira Filho AG, Santos JB, et al. Trimetazidine on ischemic in- chondrial DNA damage. Basic Res Cardiol. 2016;111(3):29.

Keywords: cardiac metabolism; ischemic heart disease; metabolic agent; mitochondrial dysfunction

LA MALADIE CORONAIRE CHRONIQUE : UN DSQUILIBRE NERGTIQUE


Rcemment, des altrations du mtabolisme cardiaque ont t impliques dans la physiopathologie de la maladie
coronaire. En conditions normales, la plus grande partie de lnergie cardiaque provient de la b -oxydation des acides
gras libres. Cependant, le cur sain peut facilement passer dun substrat un autre selon la disponibilit du subs-
trat, ltat nutritionnel et le niveau deffort. Paradoxalement, au cours dune ischmie prolonge et svre, le myocarde
continue de tirer la plupart de son nergie (jusqu 90 %) de la b -oxydation. Il faut une plus grande quantit doxy-
gne pour oxyder compltement un acide gras avec une chane de carbone dont la longueur est quivalente celle
du glucose. Loxydation des acides gras est considre comme prjudiciable car elle produit moins dATP et plus de
drivs ractifs de loxygne, tout en ncessitant plus doxygne, ce qui rduit donc encore lefficacit de la chaine
respiratoire mitochondriale. Hormis les changements mtaboliques, le processus de production et dutilisation de
lnergie est trs complexe et comprend de nombreuses tapes : labsorption des mtabolites par les myocytes car-
diaques, la phosphorylation oxydative dans les mitochondries et le transport de lATP vers les diffrents comparti-
ments intracellulaires. Cest ainsi quune dtrioration de lune de ces tapes peut avoir un impact norme sur lho-
mostasie cellulaire. En plus des changements aigus et chroniques du mtabolisme cardiaque, il a t montr que
la dysfonction mitochondriale favorise lischmie myocardique et les troubles myocardiques pendant la phase de
reperfusion dun vnement ischmique, ce qui rduit alors encore un peu plus laptitude du cur synthtiser et
utiliser lATP.

256 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Chronic ischemic heart disease: an energy imbalance Guarini and Marzilli

M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

Mitochondria lie at the heart


of a number of cardioprotective
signaling pathways underlying is-
chemic conditioning. As such, a
variety of pharmacological treat-
ment strategies aimed at protect-
Mitochondria as a
ing mitochondria against the detri-
mental effects of acute myocardial
ischemia/reperfusion injury have
therapeutic target in ischemia
been investigated in both experi-
mental and clinical studies, with
mixed results. Of these strategies,
the current treatments that are al-
ready in clinical practice include
trimetazidine and ivabradine.

b y D . J . H a u s e n l oy, U n i t e d K i n g d o m

I
schemic heart disease is the leading cause of death and disability world-
wide. As such, novel therapeutic targets are urgently required to protect the
heart against the detrimental effects of acute ischemia/reperfusion injury
in order to preserve cardiac function and improve clinical outcomes in patients
with ischemic heart disease. In this regard, mitochondria, which are the pow-
erhouses of the cell and which make up one-third of the volume of a cardiomy-
ocyte, are an important target for cardioprotection. Elucidation of the signal-
ing pathways underlying the endogenous cardioprotective phenomenon of
ischemic conditioning, in which the heart can be protected by brief nonlethal
episodes of ischemia and reperfusion, has identified mitochondria to be the
Derek J. HAUSENLOY, MD, PhD end-effector in many of the signal transduction pathways. In this article, we re-
Cardiovascular and Metabolic
view the role of mitochondria as targets for protecting the heart against acute
Disorders Program, Duke-NUS
Graduate Medical School ischemia/reperfusion injury, the therapeutic application of which should help
SINGAPORE improve clinical outcomes in patients with ischemic heart disease.
National Heart Research Institute Medicographia. 2016;38:257-263 (see French abstract on page 263)
Singapore, National Heart Centre
Singapore, SINGAPORE
The Hatter Cardiovascular Institute
schemic heart disease remains the leading cause of death and disability both in

I
Institute of Cardiovascular Science
University College London Europe specifically and worldwide. As such, there remains an urgent need to dis-
London, UNITED KINGDOM
cover novel therapies that can protect the heart against the detrimental effects
National Institute of Health Research
of acute ischemia/reperfusion injury (IRI) in order to preserve left ventricular (LV) sys-
University College London Hospitals
Biomedical Research Centre tolic function and prevent the onset of heart failure. In this article, we review the role
London, UNITED KINGDOM of mitochondria as a therapeutic target for protecting the heart against acute IRI.
In the heart, mitochondria occupy nearly one-third the volume of a cardiomyocyte,
highlighting their importance as the powerhouses of the cell, providing the energy
required for normal cardiac contractile function.

Metabolic and biochemical consequences of acute


ischemia/reperfusion
An acute coronary artery occlusion results in a critical reduction in coronary blood
flow and deprivation of oxygen and nutrients to the affected area of myocardium,
Address for correspondence: and it impairs the clearance of waste metabolites, subjecting cardiomyocytes to the
Professor Derek Hausenloy,
Cardiovascular and Metabolic abrupt metabolic and biochemical changes associated with acute myocardial is-
Disorders Program, Duke-National chemia. If blood flow is restored to the affected area by the removal of the coronary
University of Singapore,
8 College Road, Singapore 169857
artery occlusion, the ischemic myocytes are then exposed to the further metabolic
(email: and biochemical changes associated with the reperfusion process. The combined
derek.hausenloy@duke-nus.edu.sg) injury sustained by the myocardium during these processes is termed acute my-
www.medicographia.com ocardial IRI, and the sequential metabolic and biochemical perturbations that oc-

Mitochondria as a therapeutic target in ischemia Hausenloy MEDICOGRAPHIA, Vol 38, No. 3, 2016 257
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

cur during this process are reviewed below and presented in


Figure 1. Deprivation of oxygen during acute myocardial is-
chemia impairs oxidative phosphorylation by reducing elec-
tron flow through the mitochondrial electron transport chain,
leading to an accumulation of NADH and FADH and cessa-
tion of adenosine triphosphate (ATP) production.1 The mito-
chondrial membrane potential collapses as it is no longer
maintained by the electrochemical gradient across the inner
mitochondrial membrane.2 Intracellular creatine phosphate
is depleted with a concomitant rise in intracellular inorganic
phosphate (Pi), resulting in mitochondria accumulating Pi.3
Residual reserves of ATP are hydrolyzed by F0F1-ATPase in
an attempt to restore the mitochondrial membrane potential,4
resulting in catalytic metabolites such as hypoxanthine, which
are oxidized to release free radicals. The activity of the adenine
nucleotide translocase is reduced, impairing oxidative phos-
Figure 1. The metabolic and biochemical changes that occur
phorylation further still.5
during acute myocardial ischemia and reperfusion.
During acute myocardial ischemia, there is an increase in intracellular calcium
The reduced availability of ATP and oxygen drives anaerobic (Ca2+), inorganic phosphate (Pi), sodium (Na+), reactive oxygen species (ROS),
glycolysis, which results in lactic acid accumulation, leading to NADH, and hydrogen (H+), a fall in adenosine triphosphate (ATP) levels, and
collapse of the mitochondrial membrane potential (Dm). At reperfusion, there
intracellular acidification. The fall in pH activates the sodium is repolarization of the Dm and restoration of mitochondrial ATP production; a
(Na+)/hydrogen (H+) exchanger in an effort to remove cytoso- further increase in Ca2+, Pi, and ROS; oxidation of NADH; and restoration of
neutral pHfactors that mediate cell death by inducing mitochondrial perme-
lic protons, which causes the entry of Na+. The Na+/potassium ability transition pore (MPTP) opening and cardiomyocyte hypercontracture.

(K+)-ATPase, which normally removes excess Na+, is inhibited


SELECTED ABBREVIATIONS AND ACRONYMS
because of the reduced availability of ATP and the increase in
AMI acute myocardial infarction intracellular Pi. This results in a rise in intracellular Na+, which
CABG coronary artery bypass graft triggers the Na+/calcium (Ca2+) exchanger to function in re-
CAD coronary artery disease verse in order to remove cytosolic Na+.6 However, this occurs
CIRCUS does Cyclosporine ImpRove Clinical oUtcome in at the expense of an increase in intracellular Ca2+. The rise in
ST-elevation myocardial infarction patients cytosolic Ca2+ results in the mitochondrial accumulation of
CsA cyclosporin A Ca2+ via the mitochondrial Na+/Ca2+ exchanger.7 The onset of
CYCLE CYCLosporinE A in reperfused acute myocardial rigor contracture coincides with the depletion of ATP,8 and is
infarction [trial] followed by cellular Ca2+ overload.9
EMBRACE Evaluation of Myocardial effects of Bendavia for
STEMI reducing Reperfusion injury in patients with Acute Therefore, after an episode of acute myocardial ischemia, the
Coronary Events [trial] metabolic and biochemical derangements include a low in-
EMIP-FR European Myocardial Infarction Project tracellular pH (<7.0), high intracellular [Ca2+] and [Pi ], and ATP
Free Radicals
depletion (Figure 1), conditions which are exacerbated once
ERK1/2 extracellular signalregulated kinase the acutely ischemic myocardium is reperfused.
IRI ischemia/reperfusion injury
JAK Janus kinase Reperfusion of acutely ischemic myocardium has several im-
LV left ventricular portant consequences: (i) re-energization of the cardiomyo-
MEK1/2 mitogen-activated protein kinase (MAP)/extracellular cyte, causing repolarization of the mitochondrial membrane
signalregulated kinase (ERK) 1/2 potential; (ii) reoxygenation of a reduced mitochondrial res-
MI myocardial infarction piratory chain, resulting in the production of reactive oxygen
MPTP mitochondrial permeability transition pore species (ROS) and oxidation of NADH/FADH; (iii) a drop in in-
PCI percutaneous coronary intervention tracellular Ca2+, but a further influx of Ca2+ into mitochondria
PI3K phosphatidylinositol 3-kinase via the Ca2+-uniporter driven by the restored mitochondrial
PPCI primary percutaneous coronary intervention membrane potential9; and (iv) the wash-out of lactic acid,
RISK reperfusion injury salvage kinase [pathway] which in combination with the reactivation of the Na+/H+ ex-
SAFE survival activating factor enhancement [pathway] change acts to restore a neutral pH. Many of the biochemical
STAT3 signal transducer and activator of transcription 3 and metabolic changes that take place during the first few
STEMI ST-segment elevation myocardial infarction minutes of reperfusion can mediate cardiomyocyte death by
inducing the opening of the mitochondrial permeability tran-

258 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Mitochondria as a therapeutic target in ischemia Hausenloy
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

sition pore (MPTP), a nonselective channel of the inner mi- Inhibiting MPTP opening to protect the heart
tochondrial membrane whose opening uncouples oxidative against acute IRI
phosphorylation, resulting in ATP depletion and cell death by The MPTP is a nonselective channel of the inner mitochon-
necrosis.10 drial membrane, the opening of which mediates cell death
by uncoupling oxidative phosphorylation and inducing mito-
Therefore, the ultimate aim of any cardioprotective strategy chondrial swelling, resulting in ATP depletion and necrotic cell
designed to protect the cardiomyocyte against acute IRI is death.10 The molecular composition of the MPTP is not clear,
to preserve cellular energetic and ionic homeostasis during although it has been suggested that ATP synthase11-13 and
this insult. In this regard, protecting mitochondrial function mitochondrial cyclophilin D14,15 are important components. In
to preserve energy production in response to acute myocar- the setting of an acute myocardial infarction (AMI), it has been
dial IRI is an important strategy of cardioprotection, which has shown to remain closed during acute myocardial ischemia
been investigated in both experimental and clinical studies and to be open in only the first few minutes of reperfusion.
(see Figure 2). Therefore, preventing its opening at the onset of reperfusion is
an important therapeutic strategy
for reducing myocardial infarct
(MI) size after an AMI.16,17 Prevent-
ing MPTP opening at the onset of
reperfusion can be achieved in
various ways as follows10,18,19: (i)
directly by pharmacological MPTP
inhibition; (ii) indirectly through
the activation of signaling path-
ways that converge on the MPTP;
or (iii) indirectly by modifying fac-
tors such as mitochondrial Ca2+
overload and ROS production,
which are known to induce MPTP
opening (see Figure 2).

u Signaling pathways under-


lying ischemic conditioning
that target the MPTP
The heart can be protected from
the detrimental effects of acute
IRI by subjecting it to brief non-
lethal episodes of ischemia and
reperfusion, a phenomenon that
Figure 2. Mitochondrial targets for cardioprotection. has been termed ischemic con-
The diagram provides a simplified scheme of some of the potential mitochondrial targets for cardioprotection, many of ditioning.20,21 Importantly, the pro-
which have been elucidated from studies investigating the signaling pathways underlying ischemic conditioning. These
signal transduction pathways include the RISK (involving PI3K-Akt and MEK1/2-Erk1/2), SAFE (involving JAK-STAT), tective stimulus can be applied
and NO-cGMP pathways, all of which terminate at the mitochondria and, particularly, the mitochondrial permeability directly to the heart before the in-
transition pore (MPTP). These reperfusion salvage pathways have been shown to activate downstream mediators, such
as eNOS, GSK3b, HKII, PKCe, and KATP , which then mediate the inhibitory effect on MPTP opening. The modulation dex ischemic event (ischemic pre-
of mitochondrial energetics by actions on the electron transport chain and adenosine triphosphate production can conditioning)22 or at the onset of
also indirectly prevent MPTP opening in response to acute ischemia/reperfusion injury (IRI).
Cyclosporin A protects against acute IRI by inhibiting MPTP opening via inhibition of CypD. TRO40303 is believed to
reperfusion (ischemic postcon-
protect the heart by inhibiting MPTP opening via attenuation of reactive oxygen species (ROS) production in response ditioning)23 or it can be applied to
to acute IRI. MTP-131 protects the heart against acute IRI by improving mitochondrial energetics via targeting of car- an organ or tissue remote from
diolipin in the inner mitochondrial membrane. Trimetazidine protects the heart against acute IRI by inhibiting fatty acid
oxidation, thereby promoting glucose oxidation, the result of which is improved mitochondrial energetics. Ivabradine the heart (remote ischemic con-
protects the heart against acute IRI by lowering the heart rate and through pleiotropic effects, which include attenua- ditioning).24 A number of different
tion of ROS production and inhibition of MPTP opening.
Abbreviations: cGMP, cyclic guanosine monophosphate; Cardio, cardiolipin; CypD, cyclophilin D; eNOS, endothelial
signal transduction pathways
nitric oxide synthase; Erk1/2, extracellular signalregulated kinase 1/2; ETC, electron transport chain; GSK3, glyco- have been shown to mediate is-
gen synthase kinase 3b; HKII, Hexokinase II; IRI, ischemia/reperfusion injury; JAK, Janus kinase; KATP, mitochondrial chemic conditioning, including
adenosine-triphosphatedependent potassium channel; MEK1/2, mitogen-activated protein kinase (MAP)/extracellular
signalregulated kinase (ERK)1/2; MPTP, mitochon-drial permeability transition pore; NO, nitric oxide; PI3K, phosphatidyli- the reperfusion injury salvage ki-
nositol 3-kinase; PKC, protein kinase C epsilon; PKG, protein kinase G; ROS, reactive oxygen species; SAFE, sur- nase (RISK) pathway (compris-
vivor activating factor enhancement; STAT, signal transducer and activator of transcription; RISK, reperfusion injury
salvage kinase.
ing the PI3K- Akt and MEK1/2-
Modified from reference 64: Hausenloy et al. Basic Res Cardiol. 2009;104(2):189-202. 2009, Springer-Verlag. Erk1/2),25,26 the survival activat-

Mitochondria as a therapeutic target in ischemia Hausenloy MEDICOGRAPHIA, Vol 38, No. 3, 2016 259
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

ing factor enhancement (SAFE) pathway (comprising tumor MITOCARE study (which investigated the efficacy and safety
necrosis factor a, JAK-STAT3),27,28 and the nitric oxide [NO]- of TRO40303 for reduction in reperfusion injury in patients un-
cyclic guanosine monophosphate [cGMP] pathway.29 These dergoing revascularization for STEMI),45 this agent failed to
signaling cascades mediate the ischemic conditioning stim- reduce MI size when administered at the time of PPCI, de-
ulus from the cell surface receptor to the mitochondria where spite careful patient selection (completely occluded infarct-re-
they mediate cardioprotection by inhibiting MPTP opening (see lated artery, large area at risk [AAR]). The neutral findings of
Figure 2). The elucidation of these signaling pathways under- the MITOCARE study may be due in part to ambiguous car-
lying ischemic conditioning has made it possible to use phar- dioprotective effects previously revealed in experimental stud-
macological agents to activate these signal mediators and ies and the fact that the formulation and dosage of TRO40303
recapitulate cardioprotection (Figure 2). Examples of RISK, used in the clinical study differed from that in experimental
SAFE, and NO-cGMP pathway activators that have been studies. Finally, more adverse events were reported in patients
shown to protect the heart against acute IRI include growth administered TRO40303 than in the placebo arm,45 thereby
factors and cytokines, such as atrial natriuretic peptide, in- limiting the clinical application of this therapeutic approach.
sulin, erythropoietin, and glucagon-like peptide-1.25-29
u MTP-131 and myocardial energetics
u Cyclosporin A: a direct inhibitor of the MPTP MTP-131, a mitochondria-targeting peptide, has been shown
The immunosuppressant drug cyclosporin A (CsA) is a po- to optimize mitochondrial energetics and attenuate the pro-
tent inhibitor of MPTP opening that has been demonstrated duction of ROS by selectively targeting cardiolipin in the in-
to reduce MI size in a number of experimental animal stud- ner mitochondrial membrane. It has been reported in both
ies,16,30,31 but not all.32 This therapeutic strategy has been trans- small and large animal experimental studies to reduce MI
lated into the clinical setting in several phase 2 clinical trials in size when administered at the onset of reperfusion and to
AMI, coronary artery bypass graft (CABG) surgery, and stroke, prevent adverse LV remodeling after MI.46,47 However, in the
but the results have been mixed.33-37 The recently completed EMBRACE STEMI clinical trial (Evaluation of Myocardial ef-
CYCLE trial (CYCLosporinE A in reperfused acute myocardial fects of Bendavia for reducing Reperfusion injury in patients
infarction [NCT01650662]), which included 410 ST-segment with Acute Coronary Events),48 intravenous MTP-131 admin-
elevation MI (STEMI) patients, also failed to demonstrate any istered before PPCI failed to reduce enzymatically estimated
benefits with CsA administered before primary percutaneous MI size in a carefully selected population of anterior STEMI
coronary intervention (PPCI), in terms of ST-segment reso- patients (ischemic time <4 hours, no collateral vessels, and
lution and enzymatically estimated MI size.38 Whether MPTP fully occluded coronary artery). The reasons for the neutral re-
inhibition can improve clinical outcomes has been recently sults of this study are not known, but potential reasons may
tested in the CIRCUS trial (does Cyclosporine ImpRove Clin- include a single-targeted approach to cardioprotection, or
ical oUtcome in ST-elevation myocardial infarction patients), pharmacokinetic or pharmacodynamic difficulties in targeting
which involved 970 patients. In that trial, it was shown that mitochondria in STEMI patients.
the administration of CsA immediately before PPCI failed to
improve clinical outcomes at one year (all-cause death, heart Metabolic modulation to protect the heart
failure hospitalization, and adverse LV remodeling) in anterior against acute IRI
STEMI patients.39 Why this large phase 3 trial did not confirm Improving myocardial energetics by modulating mitochondrial
the positive results reported in previous phase 2 studies re- metabolism is an important strategy for cardioprotection that
mains unclear, but potential reasons include the following40: has been extensively investigated over the last 30 to 40 years.
(i) a possible type I error observed in small-size clinical stud- In 1970, Lionel Opie first used insulin to promote glucose ox-
ies; (ii) off-target effects of CsA, as CsA is known to inhibit idation to protect the heart against acute myocardial ische-
cyclophilin A and calcineurin, the results of which may have mia.49 This metabolic approach to protecting the ischemic
counteracted the benefit of inhibiting MPTP opening41; and, heart underlies the cardioprotective effects of trimetazidine.
perhaps, (iii) changes in STEMI patients since the initial phase 2
trial, including a greater use of the new P2Y12 platelet inhib- u Trimetazidine and metabolic modulation
itors (prasugrel, ticagrelor), which are known to reduce MI Trimetazidine is known to improve myocardial glucose uti-
size per se.42 lization by inhibiting fatty acid metabolism. It does this by in-
hibiting long-chain 3-ketoacyl-coenzyme A thiolase, there-
u TRO40303: an indirect inhibitor of the MPTP by blocking b-oxidation of fatty acids and promoting glucose
TRO40303 binds to the translocator protein TSPO in the out- oxidation.50 In the ischemic heart, where oxygen is scarce,
er mitochondrial membrane and is believed to inhibit MPTP glucose oxidation is more beneficial than fatty acid oxidation
opening by attenuating ROS production. It has been report- as the former requires less oxygen consumption than the lat-
ed in small animal experimental studies to reduce MI size,43 ter. This metabolic effect of trimetazidine is central to its anti-
but the cardioprotective effect was not replicated in a clini- anginal effects in patients with stable coronary artery disease
cally relevant porcine MI model.44 In the 163STEMI-patient (CAD).51

260 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Mitochondria as a therapeutic target in ischemia Hausenloy
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

A number of experimental studies have shown that trimetazi- experimental studies have found that this drug can protect
dine can protect the heart against acute IRI, as evidenced by the heart against acute IRI in paced animal hearts,58,60 sug-
reductions in MI size when administered as a pretreatment gesting beneficial pleiotropic effects of this drug, which may
and when administered at the onset of reperfusion.52 This ther- include attenuation of ROS production and inhibition of MPTP
apeutic approach has been investigated in patients present- opening in response to acute IRI.60 Further experimental stud-
ing with an AMI in the large EMIP-FR clinical trial (European ies are required to elucidate the mechanisms underlying the
Myocardial Infarction Project Free Radicals), and although it cardioprotective effect of ivabradine.
did not improve clinical outcomes in AMI patients reperfused
by thrombolysis, it appeared to have a beneficial effect in non- In the clinical setting, by lowering heart rate to reduce my-
reperfused patients, underscoring its anti-ischemic effect.53 ocardial oxygen consumption and increase coronary blood
flow, ivabradine has been shown to be an effective antiang-
Recent meta-analyses have shown that it can decrease pe- inal agent in patients with stable CAD.61 However, this thera-
rioperative and periprocedural myocardial injury in patients peutic approach did not improve clinical outcomes in a group
undergoing coronary revascularization by CABG surgery54 of such patients without heart failure.62 In contrast, it has been
and PCI,55 respectively, suggesting it has a cardioprotective shown to improve clinical outcomes (less cardiovascular mor-
effect in these clinical settings. The beneficial effects from tality or heart failure hospitalization) in stable CAD patients with
modulating mitochondrial metabolism may also be helpful in heart failure and heart rates above 70 beats per minute.63
heart failure, another condition in which disturbances in mi-
tochondrial metabolism play an important role.56 Summary and conclusions
Mitochondria lie at the heart of a number of cardioprotective
u Ivabradine and myocardial energetics signaling pathways underlying ischemic conditioning. As such,
Another effective approach to cardioprotection is to reduce a variety of pharmacological treatment strategies aimed at
the myocardial energy requirements of the heart during acute protecting mitochondria against the detrimental effects of
IRI. This can be achieved with little hemodynamic conse- acute myocardial IRI have been investigated in both experi-
quence by the drug ivabradine, which by inhibiting the If cur- mental and clinical studies, with mixed results. Of these strate-
rent in the sinus node can induce a selective lowering of heart gies, the current treatments that are already in clinical prac-
rate.57 This drug has been demonstrated to lower heart rate tice include trimetazidine and ivabradinefurther studies are
and reduce myocardial ischemia in several experimental stud- required to elucidate the benefit of these agents in acute my-
ies.58-60 However, whether the anti-ischemic effect of ivabra- ocardial IRI and in improvement of clinical outcomes in pa-
dine is secondary to heart rate lowering is not clear, and recent tients with ischemic heart disease. n

References
1. Jennings RB, Reimer KA. The cell biology of acute myocardial ischemia. Annu 12. Carraro M, Giorgio V, Sileikyte J, et al. Channel formation by yeast F-ATP syn-
Rev Med. 1991;42:225-246. thase and the role of dimerization in the mitochondrial permeability transition.
2. Di Lisa F, Blank PS, Colonna R, et al. Mitochondrial membrane potential in J Biol Chem. 2014;289(23):15980-15985.
single living adult rat cardiac myocytes exposed to anoxia or metabolic inhi- 13. Bernardi P, Di LF. The mitochondrial permeability transition pore: molecular na-
bition. J Physiol. 1995;486(pt 1):1-13. ture and role as a target in cardioprotection. J Mol Cell Cardiol. 2015;78:100-106.
3. Allen DG, Orchard CH. Myocardial contractile function during ischemia and 14. Baines CP, Kaiser RA, Purcell NH, et al. Loss of cyclophilin D reveals a critical
hypoxia. Circ Res. 1987;60(2):153-168. role for mitochondrial permeability transition in cell death. Nature. 2005;434
4. Jennings RB, Reimer KA, Steenbergen C. Effect of inhibition of the mitochon- (7033):658-662.
drial ATPase on net myocardial ATP in total ischemia. J Mol Cell Cardiol.1991; 15. Nakagawa T, Shimizu S, Watanabe T et al. Cyclophilin D-dependent mitochon-
23(12):1383-1395. drial permeability transition regulates some necrotic but not apoptotic cell death.
5. Regitz V, Paulson DJ, Hodach RJ, Little SE, Schaper W, Shug AL. Mitochondri- Nature. 2005;434(7033):652-658.
al damage during myocardial ischemia. Basic Res Cardiol.1984;79(2):207-217. 16. Hausenloy DJ, Maddock HL, Baxter GF, Yellon DM. Inhibiting mitochondrial
6. Haigney MC, Miyata H, Lakatta EG, Stern MD, Silverman HS. Dependence of permeability transition pore opening: a new paradigm for myocardial precon-
hypoxic cellular calcium loading on Na+-Ca2+ exchange. Circ Res.1992;71(3): ditioning? Cardiovasc Res. 2002;55(3):534-543.
547-557. 17. Hausenloy DJ, Duchen MR, Yellon DM. Inhibiting mitochondrial permeability
7. Griffiths EJ, Ocampo CJ, Savage JS et al. Mitochondrial calcium transport- transition pore opening at reperfusion protects against ischemia-reperfusion
ing pathways during hypoxia and reoxygenation in single rat cardiomyocytes. injury. Cardiovasc Res. 2003;60(3):617-625.
Cardiovasc Res. 1998;39(2):423-433. 18. Hausenloy DJ, Ong SB, Yellon DM. The mitochondrial permeability transition
8. Bowers KC, Allshire AP, Cobbold PH. Bioluminescent measurement in single pore as a target for preconditioning and postconditioning. Basic Res Cardiol.
cardiomyocytes of sudden cytosolic ATP depletion coincident with rigor. J Mol 2009;104(2):189-202.
Cell Cardiol. 1992;24(3):213-218. 19. Ong SB, Dongworth RK, Cabrera-Fuentes HA, Hausenloy DJ. Role of the
9. Miyata H, Lakatta EG, Stern MD, Silverman HS. Relation of mitochondrial and MPTP in conditioning the heart translatability and mechanism. Br J Pharma-
cytosolic free calcium to cardiac myocyte recovery after exposure to anoxia. col. 2015;172(8):2074-2084.
Circ Res. 1992;71(3):605-613. 20. Hausenloy DJ. Cardioprotection techniques: preconditioning, postcondition-
10. Ong SB, Samangouei P, Kalkhoran SB, Hausenloy DJ. The mitochondrial per- ing and remote conditioning (basic science). Curr Pharm Des. 2013;19(25):
meability transition pore and its role in myocardial ischemia reperfusion injury. 4544-4563.
J Mol Cell Cardiol. 2015;78C:23-34. 21. Bulluck H, Hausenloy DJ. Ischaemic conditioning: are we there yet? Heart. 2015;
11. Giorgio V, von SS, Antoniel M, et al. Dimers of mitochondrial ATP synthase 101(13):1067-1077.
form the permeability transition pore. Proc Natl Acad Sci U S A. 2013;110(15): 22. Murry CE, Jennings RB, Reimer KA. Preconditioning with ischemia: a delay of
5887-5892. lethal cell injury in ischemic myocardium. Circulation. 1986;74(5):1124-1136.

Mitochondria as a therapeutic target in ischemia Hausenloy MEDICOGRAPHIA, Vol 38, No. 3, 2016 261
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

23. Zhao ZQ, Corvera JS, Halkos ME, et al. Inhibition of myocardial injury by is- 46. Dai W, Shi J, Gupta RC, Sabbah HN, Hale SL, Kloner RA. Bendavia, a mito-
chemic postconditioning during reperfusion: comparison with ischemic pre- chondria-targeting peptide, improves postinfarction cardiac function, prevents
conditioning. Am J Physiol Heart Circ Physiol. 2003;285(2):H579-H588. adverse left ventricular remodeling, and restores mitochondria-related gene ex-
24. Przyklenk K, Bauer B, Ovize M, Kloner RA, Whittaker P. Regional ischemic pression in rats. J Cardiovasc Pharmacol. 2014;64(6):543-553.
preconditioning protects remote virgin myocardium from subsequent sus- 47. Shi J, Dai W, Hale SL, et al. Bendavia restores mitochondrial energy metab-
tained coronary occlusion. Circulation. 1993;87(3):893-899. olism gene expression and suppresses cardiac fibrosis in the border zone of
25. Hausenloy DJ, Yellon DM. New directions for protecting the heart against is- the infarcted heart. Life Sci. 2015;141:170-178.
chemia-reperfusion injury: targeting the reperfusion injury salvage kinase (RISK)- 48. Gibson CM, Giugliano RP, Kloner RA, et al. EMBRACE STEMI study: a phase 2a
pathway. Cardiovasc Res. 2004;61(3):448-460. trial to evaluate the safety, tolerability, and efficacy of intravenous MTP-131 on
26. Hausenloy DJ, Yellon DM. Reperfusion injury salvage kinase signalling: taking reperfusion injury in patients undergoing primary percutaneous coronary in-
a RISK for cardioprotection. Heart Fail Rev. 2007;12(3-4):217-234. tervention. Eur Heart J. 2016;37(16):1296-1303.
27. Lecour S. Multiple protective pathways against reperfusion injury: a SAFE path 49. Opie LH. The glucose hypothesis: relation to acute myocardial ischaemia. J Mol
without Aktion? J Mol Cell Cardiol. 2009;46(5):607-609. Cell Cardiol. 1970;1:107-114.
28. Hausenloy DJ, Lecour S, Yellon DM. RISK and SAFE prosurvival signaling path- 50. Kantor PF, Lucien A, Kozak R, Lopaschuk GD. The antianginal drug trimetazi-
ways in ischemic postconditioning: two sides of the same coin. Antioxid Re- dine shifts cardiac energy metabolism from fatty acid oxidation to glucose oxi-
dox Signal. 2010;14(5):893-907. dation by inhibiting mitochondrial long-chain 3-ketoacyl coenzyme A thiolase.
29. Heusch G. Molecular basis of cardioprotection: signal transduction in ischemic Circ Res. 2000;86(5):580-588.
pre-, post-, and remote conditioning. Circ Res. 2015;116(4):674-699. 51. Peng S, Zhao M, Wan J, Fang Q, Fang D, Li K. The efficacy of trimetazidine
30. Argaud L, Gateau-Roesch O, Muntean D, et al. Specific inhibition of the mi- on stable angina pectoris: a meta-analysis of randomized clinical trials. Int J
tochondrial permeability transition prevents lethal reperfusion injury. J Mol Cell Cardiol. 2014;177(3):780-785.
Cardiol. 2005;38(2):367-374. 52. Khan M, Meduru S, Mostafa M, Khan S, Hideg K, Kuppusamy P. Trimetazi-
31. Skyschally A, Schulz R, Heusch G. Cyclosporine A at reperfusion reduces in- dine, administered at the onset of reperfusion, ameliorates myocardial dys-
farct size in pigs. Cardiovasc Drugs Ther. 2010;24(1):85-87. function and injury by activation of p38 mitogen-activated protein kinase and
32. Karlsson LO, Zhou AX, Larsson E, et al. Cyclosporine does not reduce myocar- Akt signaling. J Pharmacol Exp Ther. 2010;333(2):421-429.
dial infarct size in a porcine ischemia-reperfusion model. J Cardiovasc Phar- 53. The EMIP-FR Group. Effect of 48-h intravenous trimetazidine on short- and
macol Ther. 2010;15(2):182-189. long-term outcomes of patients with acute myocardial infarction, with and with-
33. Piot C, Croisille P, Staat P, et al. Effect of cyclosporine on reperfusion injury in out thrombolytic therapy. A double-blind, placebo-controlled, randomized trial.
acute myocardial infarction. N Engl J Med. 2008;359(5):473-481. Eur Heart J. 2000;21(18):1537-1546.
34. Ghaffari S, Kazemi B, Toluey M, Sepehrvand N. The effect of prethrombolytic 54. Zhang N, Lei J, Liu Q, Huang W, Xiao H, Lei H. The effectiveness of preoper-
cyclosporine-A injection on clinical outcome of acute anterior ST-elevation my- ative trimetazidine on myocardial preservation in coronary artery bypass graft
ocardial infarction. Cardiovasc Ther. 2013;31(4):e34-e39. patients: a systematic review and meta-analysis. Cardiology. 2015;131(2):86-96.
35. Chiari P, Angoulvant D, Mewton N, et al. Cyclosporine protects the heart during 55. Zhang Y, Ma XJ, Shi DZ. Effect of trimetazidine in patients undergoing percuta-
aortic valve surgery. Anesthesiology. 2014;121(2):232-238. neous coronary intervention: a meta-analysis. PLoS One. 2015;10(9):e0137775.
36. Hausenloy D, Kunst G, Boston-Griffiths E, et al. The effect of cyclosporin-A on 56. Lopatin YM, Rosano GM, Fragasso G, et al. Rationale and benefits of trimetazi-
peri-operative myocardial injury in adult patients undergoing coronary artery by- dine by acting on cardiac metabolism in heart failure. Int J Cardiol. 2016;203:
pass graft surgery: a randomised controlled clinical trial. Heart. 2014;100(7):544- 909-915.
549. 57. DiFrancesco D, Camm JA. Heart rate lowering by specific and selective I(f) cur-
37. Nighoghossian N, Berthezene Y, Mechtouff L, et al. Cyclosporine in acute is- rent inhibition with ivabradine: a new therapeutic perspective in cardiovascu-
chemic stroke. Neurology. 2015;84(22):2216-2223. lar disease. Drugs. 2004;64(16):1757-1765.
38. Latini R, Limbruno U, La Vecchia L, et al. Effect of cyclosporine a on infarct 58. Heusch G, Skyschally A, Gres P, van CP, Schilawa D, Schulz R. Improvement of
size reduction in reperfused acute myocardial infarction treated with primary regional myocardial blood flow and function and reduction of infarct size with
angioplasty. Circulation. 2014;130:A15211. ivabradine: protection beyond heart rate reduction. Eur Heart J. 2008;29(18):
39. Cung TT, Morel O, Cayla G, et al. Cyclosporine before PCI in patients with 2265-2275.
acute myocardial infarction. N Engl J Med. 2015;373(11):1021-1031. 59. Ceconi C, Cargnoni A, Francolini G, Parinello G, Ferrari R. Heart rate reduc-
40. Hausenloy DJ, Yellon DM. Targeting myocardial reperfusion injurythe search tion with ivabradine improves energy metabolism and mechanical function of
continues. N Engl J Med. 2015;373(11):1073-1075. isolated ischemic rabbit heart. Cardiovasc Res. 2009;84(1):72-82.
41. Hausenloy DJ, Boston-Griffiths EA, Yellon DM. Cyclosporin A and cardioprotec- 60. Kleinbongard P, Gedik N, Witting P, Freedman B, Klocker N, Heusch G. Pleio-
tion: from investigative tool to therapeutic agent. Br J Pharmacol. 2012;165(5): tropic, heart rate-independent cardioprotection by ivabradine. Br J Pharmacol.
1235-1245. 2015;172(17):4380-4390.
42. Yang XM, Liu Y, Cui L, et al. Platelet P2Y12 blockers confer direct postcondi- 61. Borer JS, Fox K, Jaillon P, Lerebours G. Antianginal and antiischemic effects
tioning-like protection in reperfused rabbit hearts. J Cardiovasc Pharmacol Ther. of ivabradine, an If inhibitor, in stable angina: a randomized, double-blind, mul-
2013;18(3):251-262. ticentered, placebo-controlled trial. Circulation. 2003;107(6):817-823.
43. Schaller S, Paradis S, Ngoh GA, et al. TRO40303, a new cardioprotective com- 62. Fox K, Ford I, Steg PG, Tardif JC, Tendera M, Ferrari R. Ivabradine in stable coro-
pound, inhibits mitochondrial permeability transition. J Pharmacol Exp Ther. nary artery disease without clinical heart failure. N Engl J Med. 2014 8;371(12):
2010;333(3):696-706. 1091-1099.
44. Hansson MJ, Llwyd O, Morin D et al. Differences in the profile of protection af- 63. Fox K, Komajda M, Ford I, et al. Effect of ivabradine in patients with left-ven-
forded by TRO40303 and mild hypothermia in models of cardiac ischemia/ tricular systolic dysfunction: a pooled analysis of individual patient data from
reperfusion injury. Eur J Pharmacol. 2015;760:7-19. the BEAUTIFUL and SHIFT trials. Eur Heart J. 2013;34(29):2263-2270.
45. Atar D, Arheden H, Berdeaux A, et al. Effect of intravenous TRO40303 as an 64. Hausenloy DJ, Ong SB, Yellon DM. The mitochondrial permeability transition
adjunct to primary percutaneous coronary intervention for acute ST-elevation my- pore as a target for preconditioning and postconditioning. Basic Res Cardiol.
ocardial infarction: MITOCARE study results. Eur Heart J. 2015;36(2):112-119. 2009;104(2):189-202.

Keywords: cardioprotection; ischemia; ischemic conditioning; mitochondria; therapeutic target; trimetazidine

262 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Mitochondria as a therapeutic target in ischemia Hausenloy
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

LA MITOCHONDRIE , CIBLE THRAPEUTIQUE DANS L ISCHMIE


La maladie coronaire est la cause principale de dcs et dinvalidit dans le monde. Il est donc urgent de trouver de
nouvelles cibles thrapeutiques pour protger le cur des effets dltres des lsions aigus dischmie/reperfu-
sion afin de prserver la fonction cardiaque et damliorer les rsultats cliniques chez les patients coronariens. cet
gard, les mitochondries, qui sont les centrales nergtiques de la cellule et reprsentent le tiers du volume dun
cardiomyocyte, sont une cible importante pour la cardioprotection. La comprhension des voies de signalisation sous-
tendant le phnomne endogne de cardioprotection du conditionnement ischmique, par lequel le cur peut tre
protg par des pisodes brefs non ltaux dischmie et de reperfusion, a permis dtablir que la mitochondrie est
leffecteur terminal dans beaucoup de voies de transduction des signaux. Dans cet article, nous examinons la mito-
chondrie en tant que cible pour la protection du cur contre les lsions dischmie/reperfusion, une approche dont
lapplication thrapeutique pourrait aider amliorer les rsultats cliniques des patients coronariens.

Mitochondria as a therapeutic target in ischemia Hausenloy MEDICOGRAPHIA, Vol 38, No. 3, 2016 263

M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

the less traditional view held


by the Brazilian Society of Cardiol-
ogy recognizes that trimetazidine
may be offered early onbefore
long-acting nitratesin combina-
tion with any hemodynamic agent,
The place of metabolic agents
as long as blood pressure and
heart rate are properly controlled.
With this latter position, a different
in contemporary coronary
view emerges of the treatment of
patients with stable IHD, one that artery disease guidelines
widens the concept of optimal
medical therapy and allows for
the inclusion of trimetazidine be-
fore the commonplace after every-
thing else has failed stance.

b y L . H . W. G o w d a k , B ra z i l

I
n the healthy human heart, free fatty acids (FFAs) supply approximately 60%
to 90% of the energy used to synthesize adenosine triphosphate (ATP); 10%
to 40% comes from glucose and lactate. During ischemia, reduced deliv-
ery of oxygen to cardiomyocytes leads to a decrease in ATP formation by ox-
idative phosphorylation, an increase in the rate of glycolysis, and a high rate of
conversion of pyruvate to lactate. This metabolic disturbance yields a disrup-
tion in cell homeostasis (with accumulation of lactate and H + ions), a fall in
intracellular pH, and a reduction in contractile work. A metabolic shift by direct
inhibition of FFA oxidation in the mitochondria with trimetazidine results in
a decrease in the frequency of angina attacks, increased exercise tolerance,
Luis Henrique Wolff GOWDAK, improvement in quality of life, enhanced myocardial contractility in patients
MD, PhD, FESC
with left ventricular dysfunction, and reduced myocardial damage during my-
Laboratory of Genetics & Molecular
Cardiology and Chronic Coronary ocardial revascularization procedures. The European Society of Cardiology
Artery Unit, Heart Institute (InCor) guidelines on stable angina indicate that trimetazidine may be considered sec-
University of So Paulo Medical
ond-line for the treatment of angina/relief of ischemia in patients already re-
School, So Paulo, BrAzIL
ceiving a b -blocker and/or calcium channel antagonist to control symptoms.
With a different view, the Brazilian guidelines on stable angina recommend
trimetazidine for symptom relief as an add-on therapy right after b -blockers,
but before long-acting nitrates (unless there is a need for better blood pres-
sure control, in which case calcium channel antagonists are preferable).
Medicographia. 2016;38:264-270 (see French abstract on page 270)

hen William Heberden first named and described angina pectoris in

W 1772, its treatment appeared the stuff of far-off dreams: () With re-
spect to the treatment of this complaint, I have little or nothing to ad-
vance: nor indeed is it to be expected we should have made much progress in
the cure of a disease, which has hitherto hardly had a place or a name in medical
books.1 How times have changed. Since the description more than 250 years ago
of the clinical presentation of an entity of which William Heberden could not recol-
lect any mention among medical authors,1 and for which he had little or nothing
to advance regarding its treatment, the modern cardiologist now faces a complete-
Address for correspondence:
Dr Luis Henrique W. Gowdak, ly different challenge: that of devising an adequate therapeutic strategy for patients
Laboratory of Genetics & Molecular with stable angina using the many options available, which include antianginal drugs
Cardiology, Heart Institute (InCor),
Av. Dr. Eneas de Carvalho Aguiar, 44
and myocardial revascularization procedures (percutaneous or surgical). But for
05403-000 So Paulo, Brazil physicians of the past the treatment scenario was very different: it took almost a
(email: luis.gowdak@incor.usp.br) century after Heberdens description before the Scottish physician Sir Thomas Brun-
www.medicographia.com ton2 first used amyl nitrite in the treatment of angina pectoris in 1867. In angina

264 MEDICOGRAPHIA, Vol 38, No. 3, 2016 The place of metabolic agents in contemporary coronary artery disease guidelines Gowdak
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

pectoris we wish a drug which will relax spasm


of the vessels very quickly, but as a rule we do
not require the relaxation to be prolonged, said
Brunton of the type of pharmacological effect
he thought would be useful.3 While Brunton
was using amyl nitrite, another British physician,
William Murrell, began using nitroglycerine in
the treatment of angina pectoris.4 After almost
a century during which the use of nitrates had
gained wide popularity as the only medical treat-
ment for angina, there were two important new
arrivals in the twentieth century: b-blockers5
(early-1960s) and calcium channel antagonists6
(late-1960s to mid-1970s). These drugs formed
the new medical management basis for symp-
tom relief in patients with stable angina; they
could either be used alone or in combination,
Figure 1. Temporal sequence of ischemic events during stress-induced myocar-
as tolerated, in the absence of contraindications dial ischemia. Note that after perfusion heterogeneity sets in, metabolic alteration
to their use. appears before any detectable functional abnormality. Chest pain is usually the
latest manifestation of ongoing myocardial ischemia.
Although many patients certainly benefited from Abbreviation: ECG, electrocardiography.
a combination of these so-called hemodynam- Korean Copied from reference 10: Kim and Youn. J Cardiovasc Ultrasound. 2009;17(2):40-53. 2009,
Society of Echocardiography.
ic agents, it later became apparent that others
remained symptomatic despite the use of maximally tolerat- optimal medical therapy with or without PCI was offered to
ed doses of these drugs. Moreover, even after an increase patients with stable angina, about 40% of patients were still
in the use of myocardial revascularization procedures, includ- complaining of angina after only 3 years of follow-up, regard-
ing a boom in the number of percutaneous coronary inter- less of the therapeutic strategy assigned. These observations,
ventions (PCIs), patients would remain free of angina for dif- which are not unique in the literature, raised the question,
ferent lengths of time, only to return a couple of years later When it comes to offering optimal symptom control, what is
complaining of angina again. In the randomized trial MASS true optimal medical therapy in patients with stable angina?
(Medicine, Angioplasty, or Surgery Study) II,7 for example, in This led to an intensive search for drugs with alternative mech-
patients with nonlimiting angina, multivessel disease, and anisms of action to hemodynamic modulation to be added to
preserved left ventricular function at baseline, 45% of those the therapeutic armamentarium for the management of pa-
initially assigned to optimal medical therapy (a combination of tients with ischemic heart disease (IHD).
b-blockers and/or long-acting nitrates and/or calcium chan-
nel antagonists) were still symptomatic after 5 years of follow- Optimizing energy metabolism as a therapeutic
up; however, it should be noted that even for those who were target in patients with angina
assigned to a myocardial revascularization procedure, the Even though angina is regarded as the clinical hallmark of
prevalence of angina was 22% (PCI group) and 25% (surgical coronary artery disease (CAD), as so eloquently described by
group). In the COUrAGE (Clinical Outcomes Utilizing revas- Heberden, myocardial ischemia cannot be considered an
cularization and AGgressive drug Evaluation) trial,8 in which event precisely circumscribed by the onset and resolution of
angina. In fact, the identification of an ischemic cascade of
pathophysiologic events9 broadened not only our understand-
SELECTED ABBREVIATIONS AND ACRONYMS
ing of different clinical presentations of CAD, but also opened
3-KAT 3-ketoacyl coenzyme A thiolase up opportunities for new therapeutic strategies. Figure 1 shows
ATP adenosine triphosphate the temporal sequence of ischemic events during stress-in-
CAD coronary artery disease duced myocardial ischemia.10
COURAGE Clinical Outcomes Utilizing revascularization and
AGgressive drug Evaluation u Basic concepts in heart metabolism
EMA European Medicines Agency Because metabolic disturbances occur early on during my-
FFA free fatty acid ocardial ischemia, the clinician should bear in mind basic
IHD ischemic heart disease concepts related to heart metabolism. In the healthy human
MASS Medicine, Angioplasty, or Surgery Study heart, free fatty acids (FFAs) supply approximately 60% to
PCI percutaneous coronary intervention 90% of the energy used to synthesize adenosine triphos-
phate (ATP), whereas the remaining 10% to 40% of the ener-

The place of metabolic agents in contemporary coronary artery disease guidelines Gowdak MEDICOGRAPHIA, Vol 38, No. 3, 2016 265
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

gy comes from different energetic substrates, ie, glucose and


lactate.11 Once in the mitochondrion, FFAs undergo b-oxida-
tion, a complex multistep enzymatic process, to produce ATP
for contractile work, calcium uptake by the sarcoplasmic retic-
ulum, and ion homeostasis.12 Glucose is taken up by the my-
ocardium and is either stored as glycogen or broken down
by glycolysis to pyruvate in the cytosol of the cell. Pyruvate
is oxidized to acetyl-CoA in the mitochondria by the enzyme
pyruvate dehydrogenase. Oxidation of glucose and lactate is
strongly inhibited by high rates of FFA oxidation in the heart
(Figure 2).11

u Heart metabolism during ischemia


reduced oxygen delivery to cardiomyocytes during ischemia
leads to mitochondrial metabolic dysfunction, resulting in a
decrease in ATP formation by oxidative phosphorylation and Figure 2. Cardiac energy metabolism under normal aerobic con-
ditions.
an increase in the rate of glycolysis. Pyruvate produced by gly-
Abbreviations: ADP, adenosine diphosphate; ATP, adenosine triphosphate; SR,
colysis is not so readily oxidized in the mitochondria, which sarcoplasmic reticulum.
allows a high rate of conversion of pyruvate to lactate in the Modified from reference 11: Stanley. Eur Heart J Suppl. 2001;3(suppl O):O2-O7.
cytosol and a consequent rise in tissue lactate levels.13 This 2001, The European Society of Cardiology.

metabolic disturbance seen during myocardial ischemia yields


a dramatic disruption in cell homeostasis, with accumulation
of lactate and H+ ions, a fall in intracellular pH, and a reduc-
tion in contractile work (Figure 3).11

u Targeting FFA and carbohydrate oxidation in IHD


Direct inhibition of FFA oxidation in the mitochondria with the
3-ketoacyl coenzyme A thiolase (3-KAT) inhibitor trimetazidine
results in an increase in the rates of glucose and/or lactate
uptake and oxidation.14 The suppression of FFA oxidation and
increased oxidation of pyruvate by pyruvate dehydrogenase
in the mitochondria reduces ischemia-induced disruption of
cardiac metabolism. In other words, inhibiting cardiac FFA
oxidation and increasing the oxidation of pyruvate results in
less lactate production and less of a fall in cell pH, resulting
in clinical benefit for the ischemic patient. This direct meta-
bolic approach is optimally suited to conditions in which there
is sufficient residual oxygen delivery to the myocardium to sup-
port pyruvate oxidation in the mitochondria.15 In other words,
Figure 3. Cardiac energy metabolism during ischemia of moderate
it is important that there be a sufficient rate of acetyl-CoA ox- severity.
idation and oxygen consumption so that increasing the rate Abbreviations: ADP, adenosine diphosphate; ATP, adenosine triphosphate; Pi,
of pyruvate oxidation has a meaningful effect on the rate of inorganic phosphate.
lactate production. This metabolic shift from FFA oxidation to Modified from reference 11: Stanley. Eur Heart J Suppl. 2001;3(suppl O):O2-O7.
2001, The European Society of Cardiology.
glucose oxidation has proven effective in different scenarios
in patients with IHD and/or heart failure. lar antianginal efficacy was observed between the trimetazi-
dine and propranolol groups. No significant differences were
Clinical benefits of trimetazidine in patients with observed between trimetazidine and propranolol as regards
stable angina weekly number of angina attacks, exercise duration, or time
Back in the late 1960s, the use of trimetazidine in patients with to 1-mm ST-segment depression. Similar results were found
angina pectoris had been proposed and successfully used when trimetazidine was compared to nifedipine.18 Moreover,
in small series of patients, although the exact mechanism of trimetazidine can be effectively and safely combined with dif-
action implicated in pain relief was not clear at that time.15,16 ferent antianginal drugs including calcium channel antago-
Later on, the effects of trimetazidine were compared with those nists,19,20 b-blockers,21,22 and long-acting nitrates.23 The com-
of propranolol in a double-blind parallel group multicenter bination of trimetazidine with a hemodynamic agent yielded
study in 149 men with stable angina.17 After 3 months, simi- subjective (a decrease in the frequency of angina attacks) and

266 MEDICOGRAPHIA, Vol 38, No. 3, 2016 The place of metabolic agents in contemporary coronary artery disease guidelines Gowdak
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

objective (increase in exercise tolerance assessed during a u The North American perspective
treadmill test) evidence for its role in the management of pa- Although trimetazidine is not marketed in the United States,
tients with stable angina not sufficiently controlled with a sin- its ability to improve cellular tolerance to ischemia, delay the
gle hemodynamic agent. onset of exercise-induced ischemia, and reduce angina epi-
sodes and nitroglycerin use has been recognized by US ex-
Since most clinical trials of trimetazidine involved a limited perts in the American College of Cardiology/American Heart
number of patients, a recently published study looked at 13 Association guidelines on the management of patients with
randomized controlled trials comprising 1628 patients to de- stable angina.35
termine the efficacy of trimetazidine combined with other anti-
anginal drugs versus other antianginal drugs in the treatment u The Brazilian perspective
of stable angina pectoris.24 Figure 4 (page 268) shows that the Trimetazidine is marketed worldwide and, therefore, included
weekly mean number of angina attacks decreased (panel A) in different national guidelines for the management of patients
and exercise duration improved (panel B) in patients receiving with stable CAD or stable angina. Because a detailed North
trimetazidine on top of conventional antianginal therapy.24 American reference to its use is lacking, many countries
around the world where trimetazidine is available follow the
Trimetazidine in patients with stable angina: recommendations proposed by the European Society of Car-
what do the guidelines say? diology. The Brazilian Society of Cardiology, however, has tak-
The unequivocal benefits of trimetazidine in patients with sta- en a slightly different approach in its placement of trimetazi-
ble IHD include: (i) a decrease in the frequency of angina at- dine in the treatment of patients with stable angina.
tacks and in the need for short-acting nitrates for pain relief 24;
(ii) increased exercise tolerance25; (iii) improvement in quality In the most recent version of the Brazilian guidelines on stable
of life26; (iv) enhanced myocardial contractility in patients with angina,36 b-blockers have kept their position as the first-line
left ventricular dysfunction27; and (v) a reduction in myocardial treatment for the prophylaxis of angina attacks and short-
damage during myocardial revascularization procedures, such acting nitrates remain the cornerstone treatment for the im-
as angioplasty28 or bypass surgery.29 In terms of cardiovascu- mediate relief of chest pain due to CAD. But for patients whose
lar events, trimetazidine may decrease the risk of hospital- symptoms are poorly controlled with b-blockers, the Brazilian
izations in patients with heart failure30; the use of trimetazi- guidelines now recommend that physicians consider adding
dine has been linked to a lower risk of death in patients after trimetazidine early on, provided blood pressure and heart rate
an acute myocardial infarction31 and in patients with heart fail- have been controlled. However, if one needs better control
ure.30,32 So, how do different medical societies value these ben- of blood pressure calcium channel antagonists may be pre-
efits and incorporate trimetazidine in their guidelines on sta- ferred.
ble angina?
There are a couple of studies that may support this approach.
u The European perspective In these studies, the effects of early administration of trimeta-
The most recent guidelines on stable angina issued by the zidine on top of different background antianginal therapies
European Society of Cardiology33 acknowledge trimetazidine were assessed. In one study, 53 patients with symptomatic
as an anti-ischemic metabolic modulator with similar antiang- stable angina receiving propranolol 40 mg tid were random-
inal efficacy to propranolol and devoid of any discernible he- ized to long-acting nitrates or trimetazidine as an add-on ther-
modynamic action. In June 2012, the European Medicines apy for 6 weeks.37 Patients on the combination of a b-block-
Agency (EMA) reviewed available data regarding its efficacy er + long-acting nitrates had a 30% reduction in the number
in effort-induced myocardial ischemia.34 A thorough analysis of angina episodes per week compared to a 62% reduction
of the safety and effectiveness of trimetazidine carried out by seen in patients on the b-blocker + trimetazidine combina-
the EMA concluded that the drug was safe, although move- tion (P=0.001). A treadmill test revealed that the latter com-
ment disorders (including parkinsonism), which were uncom- bination yielded a 6-fold increase in total exercise duration
mon and reversible after drug discontinuation, could not be (95 s versus 16 s).
excluded with the use of trimetazidine. Thus, in patients with
angina pectoris, treatment with trimetazidine should be con- In another study, investigators looked at the benefit of adding
sidered as an add-on to existing treatments in those who are trimetazidine in 1213 highly symptomatic patients with sta-
not adequately controlled by, or who are intolerant to, oth- ble angina being treated with different antianginal strate-
er medicines for angina pectoris. Accordingly, the European gies, comprising b-blocker alone, b-blocker + long-acting
guidelines stated that trimetazidine may be considered for nitrates, or b-blocker + calcium channel antagonist.38 The ad-
the second-line treatment of angina/relief of ischemia in pa- dition of trimetazidine significantly reduced the weekly num-
tients already receiving a b-blocker and/or calcium channel ber of angina attacks and consumption of short-acting ni-
antagonist to control symptoms (class of recommendation: trates in all patients, regardless of the treatment strategy used.
IIb; level of evidence: B).33 But, maybe more importantly, the combination of long-acting

The place of metabolic agents in contemporary coronary artery disease guidelines Gowdak MEDICOGRAPHIA, Vol 38, No. 3, 2016 267
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

A. Weekly number of angina attacks

B. Exercise duration

Figure 4. Forest plot for the aggregate weekly mean number of angina attacks (panel A) and exercise duration at peak exercise (panel B)
in patients received trimetazidine combined with conventional antianginal agents in the treatment of stable angina pectoris, in comparison
with conventional antianginal agents.
Modified from reference 24: Peng et al. Int J Cardiol. 2014;177(3):780-785. 2014, Elsevier Ireland Ltd.

268 MEDICOGRAPHIA, Vol 38, No. 3, 2016 The place of metabolic agents in contemporary coronary artery disease guidelines Gowdak
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

nitrates or calcium channel antagonists with b-blockers did lines not only for stable angina, but also for heart failure. Al-
not provide any additional reduction in the number of angina though there is some consistency in the view of trimetazi-
attacks compared with patients receiving b-blocker + trimeta- dines use as an add-on agent, there is a slight difference in
zidine. perception about how soon trimetazidine should be added
as a second-line treatment. The more traditional viewex-
So, when the time came to review the Brazilian guidelines on pressed by the European Society of Cardiology, for instance
stable angina, the committee decided to give trimetazidine recommends trimetazidine as a second-line agent for use only
a class IIa recommendation (level of evidence: B) for symp- when a combination of conventional hemodynamic agents,
tom relief as a second-line treatment, as an add-on therapy such as b-blockers and calcium channel antagonists, has
right after b-blockers (unless, as previously stated, there is a been unable to control symptoms satisfactorily. On the other
need for better blood pressure control, in which case calci- hand, the less traditional view held by the Brazilian Society
um channel antagonists are preferable). of Cardiology recognizes that trimetazidine, which possess-
es no major safety concerns, may be offered early onbefore
Another particular aspect of this document is that long-acting long-acting nitratesin combination with any hemodynamic
nitrates have been downgraded to a third-line treatment for agent (b-blocker and/or calcium channel antagonist), as long
angina control. The reason being that at least two studies as blood pressure and heart rate are properly controlled.
have demonstrated that the long-term use of long-acting ni-
trates leads to an increase in the risk of cardiovascular events, With this latter position, a different view emerges of the treat-
including death, in healed myocardial infarction patients39 and ment of patients with stable IHD, one that widens the con-
in diabetes patients who underwent elective PCI.40 Worsening cept of optimal medical therapy and allows for the inclusion
of endothelial dysfunction is a potential complication of long- of trimetazidine before the commonplace after everything
acting nitrates that may be linked to adverse outcomes.41 else has failed stance. This proposal may seem original and
new, but it was, in fact, prophetically suggested almost 50
Conclusion years ago by two British investigators, who said that trimeta-
The benefits of trimetazidine in patients with cardiovascular zidine appears to have a place in the long term treatment of
disease have been demonstrated in several studies and meta- angina pectoris (), and may be given to all cases of angi-
analyses, allowing for its incorporation into practice guide- na.16 It is never too late to do what we ought to have done. n

References
1. Heberden, W. Some account of a disorder of the breast. Medical Transactions. 16. Brodbin P, OConnor CA. Trimetazidine in the treatment of angina pectoris. Br J
The Royal College of Physicians of London. 1772;2:59-67. Clin Pract. 1968;22(9):395-396.
2. Brunton TL. On the use of nitrite of amyl in angina pectoris. Lancet. 1867;2: 17. Detry JM, Sellier P, Pennaforte S, Cokkinos D, Dargie H, Mathes P. Trimetazi-
97-98. dine: a new concept in the treatment of angina. Comparison with propranolol in
3. Brunton TL, Bokenham TJ. Note on the effect of amyl nitrite. Pharm J.1888;19: patients with stable angina. Trimetazidine European Multicenter Study Group.
491. Br J Clin Pharmacol. 1994;37(3):279-88.
4. Murrell W. Nitro-glycerine in angina pectoris. Lancet. 1879;1:80-81. 18. Dalla-Volta S, Maraglino G, Della-Valentina P, Viena P, Desideri A. Comparison
5. Black JW, Duncan WA, Shanks rG. Comparison of some properties of pron- of trimetazidine with nifedipine in effort angina: a double-blind, crossover study.
ethalol and propranolol. Br J Pharmacol Chemother. 1965;25(3):577-591. Cardiovasc Drugs Ther. 1990;4(suppl 4):853-839.
6. Fleckenstein A. History of calcium antagonists. Circ Res.1983;52(2 pt 2):I3-I16. 19. Manchanda SC, Krishnaswami S. Combination treatment with trimetazidine and
7. Hueb W, Lopes NH, Gersh BJ, et al. Five-year follow-up of the Medicine, An- diltiazem in stable angina pectoris. Heart. 1997;78(4):353-357.
gioplasty, or Surgery Study (MASS II): a randomized controlled clinical trial of 20. Manchanda SC. Treatment of stable angina with low dose diltiazem in combi-
3 therapeutic strategies for multivessel coronary artery disease. Circulation. nation with the metabolic agent trimetazidine. Int J Cardiol. 2003;88(1):83-89.
2007;115(9):1082-1089. 21. Szwed H, Sadowski z, Elikowski W, et al. Combination treatment in stable ef-
8. Weintraub WS, Spertus JA, Kolm P, et al. Effect of PCI on quality of life in pa- fort angina using trimetazidine and metoprolol: results of a randomized, double-
tients with stable coronary disease. N Engl J Med. 2008;359(7):677-687. blind, multicentre study (TrIMPOL II). TrIMetazidine in POLand. Eur Heart J.
9. Nesto rW, Kowalchuk GJ. The ischemic cascade: temporal sequence of he- 2001;22(24):2267-2274.
modynamic, electrocardiographic and symptomatic expressions of ischemia. 22. Vitale C, Spoletini I, Malorni W, Perrone-Filardi P, Volterrani M, rosano GM. Ef-
Am J Cardiol. 1987;59(7):23C-30C. ficacy of trimetazidine on functional capacity in symptomatic patients with sta-
10. Kim TS, Youn HJ. role of echocardiography in the emergency department. ble exertional anginathe VASCO-angina study. Int J Cardiol. 2013;168(2):
J Cardiovasc Ultrasound. 2009;17(2):40-53. 1078-1081.
11. Stanley WC. Changes in cardiac metabolism: a critical step from stable angi- 23. Chazov EI, Lepakchin VK, zharova EA, et al. Trimetazidine in Angina Combi-
na to ischaemic cardiomyopathy. Eur Heart J Suppl. 2001;3(suppl O):O2-O7. nation Therapythe TACT study: trimetazidine versus conventional treatment
12. Lopaschuk GD, Ussher Jr, Folmes CDL, Jaswal JS, Stanley WC. Myocardial in patients with stable angina pectoris in a randomized, placebo-controlled,
fatty acid metabolism in health and disease. Physiol Rev. 2010;90(1):207-258. multicenter study. Am J Ther. 2005;12(1):35-42.
13. Abozguia K, Shivu GN, Ahmed I, Phan TT, Frenneaux MP. The heart metab- 24. Peng S, zhao M, Wan J, Fang Q, Fang D, Li K. The efficacy of trimetazidine on
olism: pathophysiological aspects in ischaemia and heart failure. Curr Pharm stable angina pectoris: a meta-analysis of randomized clinical trials. Int J Car-
Des. 2009;15(8):827-835. diol. 2014;177(3):780-785.
14. Kantor PF, Lucien A, Kozak r, Lopaschuk GD. The antianginal drug trimetazi- 25. zhao Y, Peng L, Luo Y, et al. Trimetazidine improves exercise tolerance in pa-
dine shifts cardiac energy metabolism from fatty acid oxidation to glucose ox- tients with ischemic heart disease: a meta-analysis. Herz. 2015 Dec 14. Epub
idation by inhibiting mitochondrial long-chain 3-ketoacyl coenzyme A thiolase. ahead of print. doi: 10.1007/s00059-015-4392-2.
Circ Res. 2000;86(5):580-588. 26. Csar LA, Gowdak LH, Mansur AP. The metabolic treatment of patients with
15. Mehrotra TN, Bassadone ET. Trimetazidine in the treatment of angina pectoris. coronary artery disease: effects on quality of life and effort angina. Curr Pharm
Br J Clin Pract. 1967;21(11):553-554. Des. 2009;15(8):841-849.

The place of metabolic agents in contemporary coronary artery disease guidelines Gowdak MEDICOGRAPHIA, Vol 38, No. 3, 2016 269
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

27. Lopatin YM, rosano GM, Fragasso G, et al. rationale and benefits of trimetazi- able at: http://www.ema.europa.eu/docs/en_GB/document_library/referrals_
dine by acting on cardiac metabolism in heart failure. Int J Cardiol. 2016;203: document/Trimetazidine_31/WC500129195.pdf.
909-915. 35. Fihn SD, Gardin JM, Abrams J, et al. 2012 ACCF/AHA/ACP/AATS/PCNA/SCAI/
28. Bonello L, Sbragia P, Amabile N, et al. Protective effect of an acute oral loading STS Guideline for the diagnosis and management of patients with stable is-
dose of trimetazidine on myocardial injury following percutaneous coronary in- chemic heart disease: a report of the American College of Cardiology Founda-
tervention. Heart. 2007;93(6):703-707. tion/American Heart Association Task Force on Practice Guidelines, and the
29. zhang N, Lei J, Liu Q, Huang W, Xiao H, Lei H. The effectiveness of preoper- American College of Physicians, American Association for Thoracic Surgery,
ative trimetazidine on myocardial preservation in coronary artery bypass graft pa- Preventive Cardiovascular Nurses Association, Society for Cardiovascular
tients: a systematic review and meta-analysis. Cardiology. 2015;131(2):86-96. Angiography and Interventions, and Society of Thoracic Surgeons. J Am Coll
30. Fragasso G, rosano G, Baek SH, et al. Effect of partial fatty acid oxidation in- Cardiol. 2012;60(24):e44-e164.
hibition with trimetazidine on mortality and morbidity in heart failure: results from 36. Cesar LA, Ferreira JF, Armaganijan D, et al. Guideline for stable coronary artery
an international multicentre retrospective cohort study. Int J Cardiol. 2013; disease. Arq Bras Cardiol. 2014;103(2 suppl 2):1-56.
163(3):320-325. 37. Michaelides AP, Spiropoulos K, Dimopoulos K, Athanasiades D, Toutouzas P.
31. Kim JS, Kim CH, Chun KJ, et al. Effects of trimetazidine in patients with acute Antianginal efficacy of the combination of trimetazidine-propranolol compared
myocardial infarction: data from the Korean Acute Myocardial Infarction reg- with isosorbide dinitrate-propranolol in patients with stable angina. Clin Drug
istry. Clin Res Cardiol. 2013;102(12):915-922. Invest. 1997;13(1):8-14.
32. Grajek S, Michalak M. The effect of trimetazidine added to pharmacological 38. Nesukay E. Treatment of stable angina in Ukraine: CLASSICA study. Ukr J Car-
treatment on all-cause mortality in patients with systolic heart failure. Cardio- diol. 2014;2:43-47.
logy. 2015;131(1):22-29. 39. Kanamasa K, Hayashi T, Kimura A, Ikeda A, Ishikawa K. Long-term, continu-
33. Montalescot G, Sechtem U, Achenbach S, et al. 2013 ESC guidelines on the ous treatment with both oral and transdermal nitrates increases cardiac events
management of stable coronary artery disease: the Task Force on the manage- in healed myocardial infarction patients. Angiology. 2002;53(4):399-408.
ment of stable coronary artery disease of the European Society of Cardiology. 40. Yiu KH, Pong V, Siu CW, Lau CP, Tse HF. Long-term oral nitrate therapy is as-
Eur Heart J. 2013;34(38):2949-3003. sociated with adverse outcome in diabetic patients following elective percu-
34. European Medicines Agency. Questions and answers on the review of medi- taneous coronary intervention. Cardiovasc Diabetol. 2011;10:52.
cines containing trimetazidine (20 mg tablets, 35 mg modified release tablet and 41. Daiber A, Mnzel T. Organic nitrate therapy, nitrate tolerance, and nitrate-induced
20 mg/ml oral solution): outcome of a procedure under Article 31 of Directive endothelial dysfunction: Emphasis on redox biology and oxidative stress. An-
2001/83/EC as amended. September 3, 2012. Accessed: April 13, 2016. Avail- tioxid Redox Signal. 2015;23(11):899-942.

Keywords: angina; coronary artery disease; guidelines; treatment; trimetazidine

LA PLACE DES AGENTS MTABOLIQUES


DANS LES RECOMMANDATIONS ACTUELLES SUR LA MALADIE CORONAIRE
Dans le cur humain sain, les acides gras libres (AGL) fournissent environ 60 % 90 % de lnergie utilise pour
synthtiser lATP (adnosine triphosphate) ; 10 % 40 % viennent du glucose et du lactate. Pendant lischmie, la
rduction de lapport en oxygne aux cardiomyocytes conduit une diminution de la formation dATP par phospho-
rylation oxydative, une augmentation du taux de la glycolyse et un taux lev de conversion du pyruvate en lac-
tate. Ce trouble mtabolique provoque une perturbation de lhomostasie cellulaire (avec laccumulation de lactate
et dions H+), une chute du pH intracellulaire et une rduction du travail contractile. Avec la trimtazidine, un trans-
fert mtabolique par inhibition directe de loxydation des AGL dans la mitochondrie diminue la frquence des crises
dangor, majore la tolrance leffort, amliore la qualit de vie, augmente la contractilit myocardique des patients
ayant une dysfonction ventriculaire gauche et diminue les lsions myocardiques pendant les procdures de revas-
cularisation myocardique. Daprs les recommandations de la Socit Europenne de Cardiologie sur langor stable,
la trimtazidine peut tre utilise en seconde ligne pour le traitement de langor ou le soulagement de lischmie chez
les patients recevant dj un b -bloquant et/ou un antagoniste calcique pour contrler les symptmes. Voyant les
choses diffremment, les directives brsiliennes sur langor stable recommandent la trimtazidine pour le soulage-
ment des symptmes comme traitement dappoint juste aprs les b -bloquants (sauf sil faut un meilleur contrle de
la pression artrielle, auquel cas les antagonistes calciques sont prfrables), mais avant les drivs nitrs longue
dure daction.

270 MEDICOGRAPHIA, Vol 38, No. 3, 2016 The place of metabolic agents in contemporary coronary artery disease guidelines Gowdak

M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

While revascularization can


improve cardiac function and de-
crease mortality risk, alterations in
cardiac energetics can persist in
the heart after revascularization.
This includes persistent abnormal-
Revascularization in angina
ities in mitochondrial oxidative me-
tabolism, as well as alterations in
energy substrate preference by the
patients: addressing cardiac
heart. In particular, a continued in-
creased reliance on fatty acid ox- metabolism challenges
idation at the expense of glucose
oxidation can occur after reper-
fusion.

b y G . D . Lo p a s c h u k , C a n a d a

T
he onset of an angina pectoris attack is associated with dramatic alter-
ations in cardiac energy metabolism. A mismatch between oxygen (O2 )
demand and O2 supply to the heart muscle results in a decrease in mi-
tochondrial oxidative metabolism, leading to an energy-deficient state in the
heart. In addition, changes in the source of substrates for cardiac mitochon-
drial energy metabolism contribute to contractile dysfunction and to a de-
crease in cardiac efficiency. These changes include an increase in the contri-
bution of cardiac fatty acid (FA) oxidation to residual mitochondrial oxidative
metabolism and an uncoupling of glycolysis from glucose oxidation. Revas-
cularization can lessen angina symptoms predominantly by lessening the mis-
Gary D. LOPASCHUK, PhD, match between O2 demand and O2 supply to the heart muscle. However, while
MSc, BSc
revascularization can improve O2 supply, some of the switches in cardiac me-
Cardiovascular Research Centre
Mazankowski Alberta Heart Institute tabolism persist after revascularization. In particular, the muscle becomes over-
University of Alberta, Edmonton ly reliant on FA oxidation as a source of energy, primarily at the expense of
CANADA
glucose oxidation. This can continue to uncouple glycolysis from glucose ox-
idation, resulting in a continued decrease in cardiac efficiency. As a result, a
challenge in revascularized angina patients is to normalize cardiac energy
metabolism and improve cardiac efficiency. Recent evidence suggests that
therapeutically regulating cardiac energy metabolism by reducing FA oxida-
tionwhich increases glucose oxidationcan improve cardiac efficiency and
cardiac function and lessen the symptoms of angina, even in revascularized
patients. In this article, we review the cardiac mitochondrial energy meta-
bolic changes that occur in the heart in angina patients, and the changes that
occur during revascularization, as well as the potential for targeting FA oxi-
dation to treat angina, both in the presence or absence of revascularization.
Medicographia. 2016;38:271-276 (see French abstract on page 276)

ngina pectoris due to coronary artery disease (CAD) is a major health prob-

A lem in the world. Revascularization of angina patients, either by percutaneous


coronary interventions (PCI) or by coronary artery bypass graft (CABG) sur-
gery, can substantially improve angina symptoms for most patients. However, symp-
Address for correspondence: toms of angina can persist and may return over time, even when taking optimal
Dr Gary Lopaschuk, 423 Heritage antianginal medications.1 As a result, it is imperative to identify new approaches to
Medical Research Centre, University
of Alberta, Edmonton, Canada,
treat angina, even in the revascularized patient. One potentially promising approach
T6G 2S2 to treating angina is to optimize cardiac energy metabolism.2 This would appear to
(email: gary.lopaschuk@ualberta.ca) be a logical approach, as a decrease in oxygen availability and alterations in car-
www.medicographia.com diac energy metabolism are prominent changes in the angina patient. However, in

Revascularization in angina patients: addressing cardiac metabolism challenges Lopaschuk MEDICOGRAPHIA, Vol 38, No. 3, 2016 271
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

order to fully exploit the potential of optimizing cardiac ener-


gy metabolism in angina patients, it is first important to un-
derstand how cardiac energy metabolism is regulated in the
normal patient, in the angina patient, and after revascular-
ization of the angina patient. This paper reviews the cardiac
metabolic changes that occur in the angina patient, and how
optimizing cardiac energy metabolism can be used as an ap-
proach to treat such patients, both those with or without revas-
cularization.

Cardiac energy metabolism in the normal heart


The heart has a very high energy demand, due to the need
to produce large amounts of adenosine triphosphate (ATP)
to support the continuous contractile function of the heart.
There are no significant energy reserves in the heart, and there
is a near complete turnover of the myocardial ATP pool every
5 to 10 seconds.2,3 To meet these high energy demands, the
heart generates ATP by metabolizing a variety of energy sub-
strates, including fatty acids, glucose, lactate, ketone bod-
ies, and amino acids (Figure 1). The contributions of each of
these energy substrates to ATP generation are tightly regu-
lated, and there is a significant degree of plasticity and inter-
dependence in the use of these energy substrates. Under
normal, physiological conditions, fatty acids and carbohy-
Figure 1. Overview of energy metabolism in the heart.
drates (ie, glucose and lactate) represent the primary meta-
Abbreviations: I-IV, complexes I to IV in the electron transport chain; ADP,
bolic fuels that sustain cardiac function, and upwards of 95% adenosine diphosphate; ATP, adenosine triphosphate; CoA, coenzyme A; CPT,
of ATP production is attributable to mitochondrial oxidative carnitine palmitoyl transferase; H +, proton; H2O, water; LDH, lactate dehydro-
genase; MPC, mitochondrial pyruvate carrier; O2 , oxygen; PDH, pyruvate dehy-
phosphorylation.2,3 The remainder of this ATP production pri- drogenase; PDHK, pyruvate dehydrogenase kinase; TCA, tricarboxylic acid.
marily originates from glycolysis, which has the benefit of pro-
ducing ATP without the need for oxygen (Figure 1). The trade Cardiac energy metabolism in angina
off of this anaerobic ATP production is that lactate and pro- Various cardiac pathophysiological states cause perturba-
tons (H+s) are two of the metabolic byproducts of glycolysis tions of the tightly regulated pathways of myocardial energy
if the pyruvate produced from glycolysis is not subsequently substrate metabolism, and these perturbations contribute to
subjected to mitochondrial oxidative metabolism.2 the progression of myocardial injury. In CAD, repeated oxy-
gen (O2) supply and demand mismatches results in down-
The heart normally displays substantial metabolic flexibility, regulation of mitochondrial oxidative metabolism, and an in-
with fatty acid and glucose metabolism having a considerable crease in glucose uptake and glycolysis.5 During an angina
degree of interdependence, a process referred to as the Ran- attack, where O2 availability is not sufficient to meet the O2
dle Cycle or the glucose/fatty acid cycle.4 Increasing fatty acid requirements of the heart, impaired mitochondrial oxidative
oxidation in the heart decreases glucose oxidation, where- metabolism results in a rapid decline in ATP production from
as increasing glucose oxidation inhibits fatty acid oxidation. fatty acid oxidation and glucose oxidation that is proportional
A key site at which fatty acid oxidation decreases glucose to the degree of ischemia (Figure 2A). Glycolysis becomes a
metabolism is at the level of pyruvate dehydrogenase (PDH), very important source of energy during ischemia due to its
the rate-limiting enzyme for glucose oxidation. High rates of ability to generate ATP in the absence of O2. During periods of
fatty acid oxidation inhibit PDH, secondary to activating a PDH mild-to-moderate ischemia, glucose uptake and the mobi-
kinase that phosphorylates and inhibits PDH. lization of endogenous glucose from stored glycogen con-
tribute to increased flux through the glycolytic pathway.2
SELECTED ABBREVIATIONS AND ACRONYMS
Despite the presence of ischemia, mitochondrial fatty acid ox-
CABG coronary artery bypass graft idation remains the predominant source of residual oxidative
CAD coronary artery disease metabolism.6 This is due to a number of ischemia-induced
DCA dichloroacetate changes, which include the following: (i) an increase in cir-
PCI percutaneous coronary intervention culating fatty acids to which the heart is exposed; (ii) an in-
PDH pyruvate dehydrogenase crease in myocardial fatty acid uptake; and (iii) an increase in
mitochondrial fatty acid uptake (see Lopaschuk et al2 for re-

272 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Revascularization in angina patients: addressing cardiac metabolism challenges Lopaschuk
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

view). The increase in the relative contribution of fatty acid tion becomes deregulated. Elevated levels of circulating fatty
oxidation to mitochondrial oxidative metabolism results in a acids combined with an increase in mitochondria fatty acid
parallel decrease in glucose oxidation during ischemia. Since uptake results in an increase in fatty acid oxidation rates dur-
glycolysis is accelerated during ischemia, the hydrolysis of gly- ing reperfusion, with a concomitant marked decrease in glu-
colytically derived ATP uncoupled from subsequent pyruvate cose oxidation rates (Figure 2B).2,6-8 This elevation in circulating
oxidation leads to an increased generation of lactate and H+ fatty acids and cardiac fatty acid oxidation after restoration
(Figure 2A). In severely ischemic hearts, this can result in a de- of blood flow can impair cardiac function and cardiac efficien-
crease in pH in the myocardium, which can lead to cell death. cy (Figure 2B).6,7 The decrease in glucose oxidation after revas-
In milder ischemia (such as seen during angina), the produc- cularization can result in increased uncoupling of glycolysis
tion of H+ from glycolysis leads to disturbances in ionic home- from glucose oxidation and a subsequent increase in produc-
ostasis, which leads to a decrease in cardiac efficiency, as tion of lactate and H+, which can decrease cardiac efficiency
ATP is required to restore these ionic imbalances. and impair heart function.2,6,7 As a result, a challenge in the re-
vascularized angina patient is to normalize cardiac energy me-
Cardiac energy metabolism after revascularization tabolism, particularly by increasing glucose oxidation rates.
Revascularization is an important approach to reducing the
myocardial O2 supply and demand mismatch that can occur While revascularization can improve cardiac function and de-
in the CAD patient. However, during revascularization, the crease mortality risk, alterations in cardiac energetics can per-
short complete interruptions of blood flow produce a pro- sist in the heart after revascularization. This includes persist-
found ischemia, which are accompanied by dramatic alter- ent abnormalities in mitochondrial oxidative metabolism,5 as
ations in cardiac energy metabolism. During reperfusion of well as alterations in energy substrate preference by the heart.
the ischemic heart, overall cardiac fatty acid oxidation rates In particular, a continued increased reliance on fatty acid ox-
are elevated, due, at least partially, to elevated levels of circu- idation at the expense of glucose oxidation can occur after
lating fatty acids (Figure 2B).5,7 In addition, the subcellular con- reperfusion,6-9 providing a challenge in the revascularized pa-
trol of fatty acid oxidation is altered, such that fatty acid oxida- tient to restore normal cardiac energy metabolism.

A B

Figure 2. Alterations in myocardial energy metabolism during ischemia and reperfusion.


During ischemia (A), glycolysis becomes the main source of energy production in the absence of or a decreased supply of oxygen. Fatty acids dominate as the
substrate for residual oxidative metabolism due to increased plasma levels of fatty acids, as well as alterations in the subcellular control of fatty acid oxidation. During
reperfusion (B), glycolytic rates remain high, while fatty acid oxidation dominates over glucose oxidation as the main source of oxidative metabolism. The dominant
fatty acid oxidation rates during reperfusion inhibit glucose oxidation. The uncoupling of glycolysis from glucose oxidation leads to an increase in proton production,
which ultimately leads to myocardial acidosis and calcium overload.
Abbreviations: I-IV, complexes I to IV in the electron transport chain; ADP, adenosine diphosphate; ATP, adenosine triphosphate; CoA, coenzyme A; CPT, carnitine
palmitoyl transferase; H +, proton; H2O, water; LDH, lactate dehydrogenase; MPC, mitochondrial pyruvate carrier; O2 , oxygen; PDH, pyruvate dehydrogenase; PDHK,
pyruvate dehydrogenase kinase; TCA, tricarboxylic acid.

Revascularization in angina patients: addressing cardiac metabolism challenges Lopaschuk MEDICOGRAPHIA, Vol 38, No. 3, 2016 273
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

Targeting fatty acid oxidation to treat angina


Classically, the pharmacological treatment of angina patients
has focused on the use of agents that alter systemic and/or
cardiac hemodynamics. With increasing knowledge of the
mechanisms regulating cardiac energy substrate metabolism,
and with the understanding that alterations in such metab-
olism contribute to the severity of angina symptoms, the
modulation and optimization of energy substrate metabolism
represents a novel and promising area for therapeutic inter-
vention in angina patients. In this regard, pharmacological
agents that shift the balance between the oxidative utilization
of fatty acid and glucose toward glucose oxidation have been
an area of intense research activity.2,3 In particular, pharma-
cological agents that either inhibit fatty acid oxidation and/or
stimulate glucose oxidation are promising anti-ischemic in-
terventions (Figure 3).2,3,8-18

Fatty acid oxidation can be inhibited directly by decreasing


fatty acid uptake into the mitochondria or by inhibiting mito-
chondrial fatty acid oxidation. Fatty acid oxidation can also be
inhibited indirectly by increasing glucose oxidation. Pharma-
cological inhibition of fatty acid oxidation with drugs that low-
er fatty acid levels, block mitochondrial fatty acid uptake, or
directly inhibit fatty acid oxidation all have potential benefit
in the setting of angina. Figure 3. Diagrams for pharmacological targets that decrease
fatty acid oxidation and/or increase glucose oxidation in the heart.
One drug that directly targets mitochondrial fatty acid oxi- Abbreviations: I-IV, complexes I to IV in the electron transport chain; ADP,
adenosine diphosphate; ATP, adenosine triphosphate; CoA, coenzyme A; CPT,
dation is trimetazidine.8-15 Trimetazidine is a partial fatty acid carnitine palmitoyl transferase; DCA, dichloroacetate; H+, proton; H2 O, water;
oxidation inhibitor that competitively inhibits the fatty acid LDH, lactate dehydrogenase; MPC, mitochondrial pyruvate carrier; O2 , oxygen;
PDH, pyruvate dehydrogenase; PDHK, pyruvate dehydrogenase kinase; TCA,
oxidation enzyme, long chain 3-ketoacyl coenzyme A thio- tricarboxylic acid.
lase.8-10 The inhibition of fatty acid oxidation is accompanied
by an increase in glucose oxidation. This is beneficial in both its ability to decrease the release of troponin T and decrease
ischemic myocardium during restoration of blood flow or in ST-segment elevation, respectively, during balloon inflation.17,18
already revascularized myocardium, as the increased glucose A recent meta-analysis of patients undergoing PCI also showed
oxidation decreases the production of H+s arising from the that the use of trimetazidine during the PCI procedure re-
uncoupling of glycolysis from glucose oxidation. sulted in a significant reduction in myocardial injury and in
improved cardiac function,18 effects that would be expected
Results from clinical studies have confirmed the effectiveness by inhibiting fatty acid oxidation in the myocardium.
of trimetazidine as an anti-ischemic agent. Treatment of angi-
na with trimetazidine increases time to 1-mm ST segment de- Although classified as a late sodium current inhibitor, rano-
pression as well as weekly nitrate consumption.11,12 Trimetazi- lazine is also a partial fatty acid oxidation inhibitor, which has
dine has been shown to have comparable effectiveness to been shown to inhibit fatty acid oxidation and reciprocally
propranolol in treating stable angina.11-14 Combination thera- increase glucose oxidation and PDH activity.19 In experi-
py with standard antianginal therapy reduces the number of mental studies, ranolazine attenuates myocardial stunning,
symptomatic episodes of angina and improves time to is- reduces infarct size, increases cardiac ejection fraction, in-
chemia-related electrocardiogram changes on exercise test- creases stroke volume, and increases mechanical efficiency
ing.13 As discussed, many revascularized angina patients still without increasing oxygen consumption.19,20 Clinically, rano-
require antianginal therapy.14,15 The use of metabolic agents lazine, as an anti-ischemic agent, has been shown to increase
during and after revascularization has been shown to provide exercise capacity and time to 1-mm ST-segment depression,
myocardial protection.15,16 Angina patients with previous PCI and reduce the number of weekly angina attacks and nitro-
or CABG show significant improvement in time to 1-mm ST- glycerin consumption as a monotherapy or combined ther-
segment depression, exercise test duration, total workload, apy.21 However, a recent study showed that ranolazine had
time to onset of angina, and weekly angina attacks and ni- no incremental benefit in angina or quality of life in patients
trate consumption.15 Trimetazidine is cardioprotective in pa- with incomplete revascularization after PCI.22 b-Adrenoceptor
tients undergoing CABG and PCI procedures, evidenced by antagonists (b-blockers) are classic drugs used in the setting

274 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Revascularization in angina patients: addressing cardiac metabolism challenges Lopaschuk
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

of angina. This class of drug is believed to exert an oxygen- improvements in left ventricular stroke volume were observed
sparing effect by a reduction in inotropic and chronotropic in the absence of changes in heart rate, left ventricular end
effects, thus reducing cardiac workload. However, blockade diastolic pressure, or myocardial oxygen consumption.25 The
of b-adrenoceptors decreases catecholamine-induced lipol- poor potency and pharmacokinetic profile of DCA make it
ysis and therefore decreases plasma fatty acid availability unlikely that this drug will ever be used clinically.
and extraction. As a result, part of the benefit of b-blockade
in the setting of angina may occur secondary to decreased Conclusion
myocardial fatty acid oxidation. Indeed, carvedilol has been Alterations in cardiac energy metabolism are an important
shown to reduce myocardial free fatty acid uptake by 57% factor in the severity of angina in both the revascularized and
in patients with heart failure.23 nonrevascularized patient. Significant metabolic changes re-
sult in an increase in the contribution of fatty acid oxidation
In addition to inhibiting fatty acid oxidation, directly increas- compared with glucose oxidation to cardiac energy produc-
ing myocardial glucose oxidation may be another approach tion, that leads to a decrease in cardiac efficiency. A challenge
to optimizing energy metabolism in angina. Dichloroacetate in the revascularized angina patient is to normalize cardiac
(DCA) acts via direct stimulation of the mitochondrial PDH energy metabolism and to improve cardiac efficiency. Thera-
complex via the inhibition of the activity of PDH kinase. The peutic strategies aimed at inhibiting fatty acid oxidation are
improved coupling between glycolysis and glucose oxida- one potential approach to optimizing energy metabolism in
tion is believed to be the mechanism by which DCA exerts the angina patient. One such approach is to directly inhibit
its cardioprotective effects.7,24 Experimental studies show that fatty acid oxidation with trimetazidine, which leads to an in-
DCA is cardioprotective in the setting of ischemia and reper- direct increase in glucose oxidation in the heart. The subse-
fusion (see Lopaschuk et al2 for review). However, clinical data quent improvement in cardiac efficiency is associated with
on the use of DCA is scarce. In a small clinical study, where beneficial effects of trimetazidine in decreasing angina symp-
DCA was given to patients with CAD via intravenous infusion, toms, even in the revascularized patient. n

References
1. Weintraub WS, Spertus JA, Kolm P, et al; COURAGE Trial Research Group. 14. Szwed H, Sadowski Z, Pachocki R, et al. The antiischemic effects and toler-
Effect of PCI on quality of life in patients with stable coronary disease. N Engl ability of trimetazidine in coronary diabetic patients. A substudy from TRIM-
J Med. 2008;359:677-687. POL-1. Cardiovasc Drugs Ther. 1999;13(3):217-222.
2. Lopaschuk GD, Ussher JR, Folmes CD, Jaswal JS, Stanley WC. Myocardial 15. Danchin N. Clinical benefits of a metabolic approach with trimetazidine in revas-
fatty acid metabolism in health and disease. Physiol Rev. 2010;90:207-258. cularized patients with angina. Am J Cardiol. 2006;98(5A):8J-13J.
3. Neubauer S. The failing heartan engine out of fuel. N Engl J Med. 2007;356: 16. Zhang Y, Ma XJ, Shi D-Z. Effect of trimetazidine in patients undergoing per-
1140-1151. cutaneous coronary intervention: a meta-analysis. PLoS ONE. 2015;10(9):
4. Randle PJ, Garland PB, Hales CN, Newsholme EA. The glucose fatty-acid cy- e0137775.
cle. Its role in insulin sensitivity and the metabolic disturbances of diabetes mel- 17. Tunerir B, Colak O, Alatas O, Besogul Y, Kural T, Aslan R. Measurement of tro-
litus. Lancet. 1963;1:785-789. ponin T to detect cardioprotective effect of trimetazidine during coronary artery
5. Holley CT, Long EK, Lindsey ME, McFall EO, Kelly RF. Recovery of hibernat- bypass grafting. Ann Thorac Surg. 1999;68(6):2173-2176.
ing myocardium: what is the role of surgical revascularization? J Card Surg. 18. Polonski L, Dec I, Wojnar R, Wilczek K. Trimetazidine limits the effects of my-
2015;30:224-231. ocardial ischaemia during percutaneous coronary angioplasty. Curr Med Res
6. Folmes CD, Sowah D, Clanachan AS, Lopaschuk GD. High rates of residual Opin. 2002;18(7):389-396.
fatty acid oxidation during mild ischemia decrease cardiac work and efficiency. 19. McCormack JG, Barr RL, Wolff AA, Lopaschuk GD. Ranolazine stimulates
J Mol Cell Cardiol. 2009;47:142-148. glucose oxidation in normoxic, ischemic, and reperfused ischemic rat hearts.
7. Liu Q, Docherty JC, Rendell JC, Clanachan AS, Lopaschuk GD. High levels Circulation. 1996;93:135-142.
of fatty acids delay the recovery of intracellular pH and cardiac efficiency in 20. Clarke B, Spedding M, Patmore L, McCormack JG. Protective effects of ra-
post-ischemic hearts by inhibiting glucose oxidation. J Am Coll Cardiol. 2002; nolazine in guinea-pig hearts during low-flow ischaemia and their association
39:718-725. with increases in active pyruvate dehydrogenase. Br J Pharmacol. 1993;109:
8. Kantor PF, Lucien A, Kozak R, Lopaschuk GD. The antianginal drug trimetazi- 748-750.
dine shifts cardiac energy metabolism from fatty acid oxidation to glucose ox- 21. Chaitman BR, Pepine CJ, Parker JO, et al. Effects of ranolazine with atenolol,
idation by inhibiting mitochondrial long-chain 3-ketoacyl coenzyme A thiolase. amlodipine, or diltiazem on exercise tolerance and angina frequency in patients
Circ Res. 2000;86:580-588. with severe chronic angina: a randomized controlled trial. JAMA. 2004;291:
9. Lopaschuk GD, Barr R, Thomas PD, Dyck JR. Beneficial effects of trimetazi- 309-316.
dine in ex vivo working ischemic hearts are due to a stimulation of glucose ox- 22. Alexander KP, Weisz G, Prather K, et al. Effects of ranolazine on angina and
idation secondary to inhibition of long-chain 3-ketoacyl coenzyme A thiolase. quality of life after percutaneous coronary intervention with incomplete revas-
Circ Res. 2003;93:e33-37. cularization: results from the Ranolazine for Incomplete VEssel Revascular-
10. Saeedi R, Grist M, Wambolt RB, Bescond-Jacquet A, Lucien A, Allard MF. ization (RIVER-PCI) Trial. Circulation. 2016;133(1):39-47.
Trimetazidine normalizes postischemic function of hypertrophied rat hearts. 23. Al-Hesayen A, Azevedo ER, Floras JS, Hollingshead S, Lopaschuk GD, Parker
J Pharmacol. Exp Ther. 2005;314:446-454. JD. Selective versus nonselective b-adrenergic receptor blockade in chronic
11. Ciapponi A, Pizarro R, Harrison J. Trimetazidine for stable angina. Cochrane heart failure: differential effects on myocardial energy substrate utilization. Eur
Database Syst Rev. 2005;(4):CD003614. J Heart Fail. 2005;7:618-623.
12. Marzilli M, Klein WW. Efficacy and tolerability of trimetazidine in stable angina: 24. Liu B, Clanachan AS, Schulz R, Lopaschuk GD. Cardiac efficiency is improved
a meta-analysis of randomized, double-blind, controlled trials. Coron Artery after ischemia by altering both the source and fate of protons. Circ Res. 1996;
Dis. 2003;14(2):171-179. 79:940-948.
13. Ruzyllo W, Szwed H, Sadowski Z, et al. Efficacy of trimetazidine in patients 25. Wargovich TJ, MacDonald RG, Hill JA, Feldman RL, Stacpoole PW, Pepine CJ.
with recurrent angina: a subgroup analysis of the TRIMPOL II study. Curr Med Myocardial metabolic and hemodynamic effects of dichloroacetate in coronary
Res Opin. 2004;20(9):1447-1454. artery disease. Am J Cardiol. 1988;6:65-70.

Revascularization in angina patients: addressing cardiac metabolism challenges Lopaschuk MEDICOGRAPHIA, Vol 38, No. 3, 2016 275
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

Keywords: fatty acid oxidation; glucose oxidation; pyruvate dehydrogenase

LA REVASCULARISATION CHEZ LES PATIENTS ANGOREUX :


TRAITER LES PERTURBATIONS DU MTABOLISME CARDIAQUE
La survenue dune crise dangor sassocie des perturbations considrables du mtabolisme nergtique car-
diaque. Un dsquilibre entre besoins et apports en O2 au niveau du muscle cardiaque entrane une diminution du
mtabolisme oxydatif mitochondrial, aboutissant un tat de dficit nergtique dans le cur. De plus, des modi-
fications de la source des substrats du mtabolisme nergtique mitochondrial cardiaque contribuent une dys-
fonction contractile et une diminution de lefficacit cardiaque. Parmi ces modifications on compte une augmen-
tation de la contribution de loxydation des acides gras au mtabolisme oxydatif mitochondrial rsiduel et un d-
couplage entre la glycolyse et loxydation du glucose. La revascularisation peut rduire les symptmes angoreux,
principalement en diminuant le dsquilibre entre besoins et apports en O2 au niveau du muscle cardiaque. Ce-
pendant, mme si la revascularisation permet daccroitre lapport en O2 , certaines altrations du mtabolisme car-
diaque persistent aprs la revascularisation. Notamment, le muscle devient trop dpendant de loxydation des acides
gras comme source dnergie, principalement aux dpens de loxydation du glucose. Ce mcanisme accroitre le
dcouplage entre la glycolyse et loxydation du glucose, aboutissant une diminution prolonge de lefficacit car-
diaque. Cest ainsi que la difficult chez les patients angoreux vasculariss est de normaliser le mcanisme nerg-
tique cardiaque et damliorer lefficacit cardiaque. Daprs des donnes rcentes, un traitement visant rguler
le mtabolisme nergtique cardiaque en diminuant loxydation des acides gras, ce qui augmente loxydation du glu-
cose, peut permettre damliorer lefficacit et la fonction cardiaques et diminuer les symptmes dangor, mme chez
les patients revasculariss. Dans cet article, nous analysons les modifications mtaboliques nergtiques mitochon-
driales cardiaques qui interviennent au niveau du cur chez les patients angoreux et les changements apparais-
sant au cours de la revascularisation, ainsi que la possibilit de cibler loxydation des acides gras pour traiter langor
chez les patients revasculariss ou non revasculariss.

276 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Revascularization in angina patients: addressing cardiac metabolism challenges Lopaschuk

M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

Although the development of


drug-eluting stents to solve in-stent
restenosis has markedly reduced
the extent of restenosis after an-
gioplasty, there is concern about
delayed reendothelialization and
T he endothelium: a therapeutic
late in-stent thrombosis. Various
strategies to prevent restenosis by
stimulating endothelialization or by
target in post-PCI patients
inhibiting vascular smooth muscle
cell (VSMC) proliferation have thus and the role of trimetazidine
been tried. However, few reports
have identified optimal agents with
cell-specific effects on VSMCs and in endothelial function
endothelial cells.

b y S . L i m , S o u t h Ko re a

T
he vascular endothelium, the surface monolayer of the vascular wall,
plays an important role in the maintenance of vascular health. It releas-
es various mediatorssuch as angiotensin II, endothelin-1, endothe-
lium-derived hyperpolarizing factor, nitric oxide, prostacyclin, prostaglandin
H2 , and thromboxane A2 that are involved in vasodilation or vasoconstric-
tion under specific conditions. Dysfunction of the endothelium has been im-
plicated in various vascular pathophysiological processes, including abnor-
mal vascular proliferation, excessive thrombus formation, vasoconstriction, and
vasospasm. It is also associated with restenosis after percutaneous coronary
intervention (PCI). However, there are few clinical studies on the use of phar-
Soo LIM, MD, PHD macological agents to improve endothelial function and decrease the rate
Department of Internal Medicine
of restenosis after PCI. Trimetazidinean agent possessing a broad spectrum
Seoul National University College
of Medicine, Seoul National of pharmacological activities, including protection against damage to the car-
University Bundang Hospital diovascular systemhas recently been shown to improve vascular endothe-
Seongnam, SOUTH KOREA
lial cell function and may reduce the risk of restenosis after PCI. These actions
occur through antioxidative and anti-inflammatory activities, increased adipo-
nectin levels, and decreased insulin resistance. Although there is no direct ev-
idence that trimetazidine can reduce cardiovascular morbidity and mortality,
reductions in these may result indirectly from its effects on endothelial func-
tion. Prospective studies with a cardiovascular end point as a primary objec-
tive are warranted to confirm the cardioprotective effects of trimetazidine.
Medicographia. 2016;38:277-281 (see French abstract on page 281)

orldwide, cardiovascular disease (CVD) and stroke impose huge health

W and economic burdens, with CVD being the number one cause of death
globally.1 Roughly 17.5 million people died from CVD in 2012, repre-
senting 31% of all global deaths. An estimated 7.4 million of these CVD deaths
were due to coronary heart disease and 6.7 million, stroke. In the United States, in
Address for correspondence:
2011, CVD accounted for 31.3% (786 641) of the 2 515 458 deaths overall, with a
Soo Lim, Associate Professor of CVD death rate of 229.6 per 100 000 Americans2 (275.7 in men, 192.3 in women),
Medicine, Department of Internal this after a 30.8% decline in CVD death rate and 15.5% decline in actual number
Medicine, Seoul National University
College of Medicine, Seoul National of CVD deaths from 2001 to 2011.2
University Bundang Hospital,
300, Gumi-dong, Bundang-gu,
Seongnam-city, South Korea The prevalence of diabetes mellitus is increasing rapidly worldwide, and this increase
(Postal code: 463-707) implies an increase in vascular complications. Vascular complications in the heart,
(email: limsoo@snu.ac.kr) brain, and peripheral arteries are more than twice as prevalent in people with dia-
www.medicographia.com betes compared with those without diabetes. Nearly 80% of people with diabetes

Trimetazidine and endothelial function Lim MEDICOGRAPHIA, Vol 38, No. 3, 2016 277
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

Figure 1. Role of endothelial dysfunction


in the development of restenosis after
percutaneous coronary intervention.
Abbreviations: eNOS, endothelial nitric oxide synthase;
PCI, percutaneous coronary intervention;
ROS, reactive oxygen species.

die from CVDs, such as coronary heart dis-


ease, cerebrovascular disease, and periph-
eral artery occlusive disease. Therefore, the
vascular complications of diabetes are now
an important public health priority world-
wide.

A 2015 statistical report on heart disease


and stroke in the United States demonstrat-
ed that the number of percutaneous coro-
nary intervention (PCI) procedures is increas-
ing.2 In patients undergoing PCI, the use
of drug-eluting stents (DESs) has greatly re-
duced the need for reintervention compared
with the use of bare metal stents (BMSs).3,4
However, the incidence of in-stent restenosis in patients af- cated, contribute to restenosis.7 Many people with diabetes
ter PCI, using either DESs or BMSs, remains high.5 Thus, in- have increasingly complex lesion characteristics and disease
stent restenosis continues to be a major problem after coro- comorbidities,6 and the risk for restenosis is high among di-
nary stenting.6 abetics.8

Endothelial dysfunction, a key step in the The vascular endotheliumthe surface monolayer of the vas-
development of restenosis after PCI cular wallplays a critical role in the maintenance of vascu-
Restenosis is the recurrence of narrowing of the coronary ar- lar health. In response to physical and chemical stimuli, it re-
tery in a maladaptive response to damage caused by angio- leases various vasoactive mediators, such as angiotensin II,
plasty. Chronic exposure to cardiovascular risk factors, such endothelin-1, endothelial cell growth factors, endothelium-
as high blood pressure, high blood glucose concentration, dependent hyperpolarizing factor, interleukins, plasminogen
dyslipidemia, smoking, and low physical activity level impair inhibitors, prostacyclin, prostaglandin H2, nitric oxide, and
the defense mechanisms in the vascular endothelium (Fi- thromboxane A2.
gure 1). Chronic inflammation and oxidative stress are well-
known causes of endothelial dysfunction, which along with Although the development of DESs to solve in-stent resteno-
proliferation of vascular cells, production of extracellular ma- sis has markedly reduced the extent of restenosis after angio-
trix, platelet activation, and increased thrombotic activity, plasty,9 there is concern about delayed reendothelialization
processes in which endothelial dysfunction has been impli- and late in-stent thrombosis.10 Various strategies to prevent
restenosis by stimulating endothelialization or by inhibiting vas-
cular smooth muscle cell (VSMC) proliferation have thus been
SELECTED ABBREVIATIONS AND ACRONYMS
tried. However, few reports have identified optimal agents with
CVD cardiovascular disease cell-specific effects on VSMCs and endothelial cells.
DES drug-eluting stent
HUVEC human umbilical vein endothelial cell Role of trimetazidine in preventing the
LysoPC lysophosphatidylcholine
development of endothelial dysfunction and in
reducing the incidence of restenosis after PCI
MCP-1 monocyte chemoattractant protein-1
The piperazine derivative trimetazidine (1-[2,3,4-trimethoxy-
OLETF Otsuka Long-Evans Tokushima Fatty [rat]
benzyl] piperazine dihydrochloride) is an anti-ischemic drug
PCI percutaneous coronary intervention effective in treating patients with angina pectoris (Figure 2).11
ROS reactive oxygen species Protective effects on cardiomyocytes have been shown in pa-
SD Sprague Dawley [rat] tients treated in primary intervention via coronary artery graft
TNF-a tumor necrosis factor a surgery.11 Trimetazidines beneficial effects on heart failure and
VSMC vascular smooth muscle cell ischemic heart disease are related to cardiac energy metab-
olism, which shifts from fatty acid oxidation to glucose oxida-

278 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Trimetazidine and endothelial function Lim
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

In addition, we found that trimetazidine suppresses caspase-3


activity.14 The effect of trimetazidine on apoptosis in HUVECs
was also assessed by measuring mitochondrial membrane
Figure 2. function.14 Compared with LysoPC-treated HUVECs, the num-
Chemical struc- ber of apoptotic cells was markedly lower in the trimetazidine-
ture of trimetazi- treated groups. After LysoPC treatment, the ratio of active
dine (1-[2,3,4- versus inactive caspase-3 was significantly higher than in
trimethoxybenzyl] controls,14 and trimetazidine lowered the active caspase-3
piperazine dihy-
drochloride). ratio in a dose-dependent manner.

tion through trimetazidines inhibition of mitochondrial long- These findings from in vitro cell studies and in vivo animal
chain 3-ketoacyl coenzyme A thiolase; this may contribute studies indicate that trimetazidine helps to prevent damage
to its antianginal effect (Figure 3).12 In addition to trimetaz- to the coronary vasculature and to reduce neointimal prolif-
dines myocardial anti-ischemic effect, it has a vasodilatory eration after vascular injury by way of targeting both vascular
effect on coronary vessels.13 endothelial cells and VSMCs. Recent studies have shown that
trimetazidine has a protective effect against restenosis after
We have recently shown that trimetazidine has beneficial ef- PCI in humans.17-19 In one study, trimetazidine treatment for
fects on the occurrence of restenosis after vascular balloon 10 weeks lessened endothelial damage in the radial artery af-
injury in diabetes.14 We investigated whether trimetazidine ter transradial coronary artery angiography or transradial PCI.19
treatment can lower the extent of restenosis occurring in the Another study of longer duration showed that trimetazidine
carotid artery after balloon injury in animal models of dia- treatment reduced the incidence of in-stent restenosis after
betes, both type 1 (streptozotocin-injected Sprague Dawley PCI with DES implantation measured at the 1-year follow-up.20
[SD] rats) and type 2 (Otsuka Long-Evans Tokushima Fatty There were also fewer major adverse cardiac events in the
[OLETF] rats). OLETF rats represent an obese model of type 2 trimetazidine-treated group than in the control group.
diabetes, in which 5-week-old rats are al-
lowed to grow to 24 weeks of age, at which
time obesity and insulin resistance devel-
op.15 Rats from both models were treated
with trimetazidine at different concentrations
or were sham treated with normal saline,
and a well-established balloon injury pro-
cedure16 was carried out in the carotid artery.
Two weeks after the procedure, the trimeta-
zidine-treated rats in both models showed
significantly less neointimal formation than
controls; in the type 1 diabetes model, trime-
tazidine-treated rats also had lower mean
intima-media ratios (this was dose depend-
ent) and markedly lower in vivo cell prolifer-
ation (as measured by immunostaining for
proliferating cell nuclear antigen) than con-
trols.14

A series of in vitro experimental studies have


shown that reendothelialization after bal-
loon injury is accelerated by trimetazidine Figure 3. Mechanism of action of trimetazidine in energy metabolism in cardiomyocytes.
treatment.17,18 In one study, trimetazidine had Abbreviations: ATP, adenosine triphosphate; CoA, coenzyme A.
a direct effect on endothelial proliferation in
human umbilical vein endothelial cells (HUVECs).14 Prolifer- Antioxidant effects of trimetazidine
ation of HUVECs incubated in medium containing 20 mM There is a large amount of evidence that oxidative stress
lysophosphatidylcholine (LysoPC) was about 20% lower than plays a role in the pathogenesis of diabetes, as well as in di-
in controls. Trimetazidine treatment restored cell proliferation abetic complications such as atherosclerosis and resteno-
in a concentration-dependent manner. Similarly, HUVEC pro- sis.21 Indeed, in the vascular wall, oxidative stress may be a
liferation was decreased by tumor necrosis factor a (TNF-a) key mechanism in processes leading to endothelial dysfunc-
treatment and recovered with trimetazidine treatment.14 tion22; it promotes VSMC migration and proliferation and is

Trimetazidine and endothelial function Lim MEDICOGRAPHIA, Vol 38, No. 3, 2016 279
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

cyte recruitment, and TNF-a and hsCRP


have important roles in the development of
atherosclerosis.29,30 Trimetazidine treatment
has been shown to increase adiponectin
levels and decrease TNF-a and MCP-1
levels.14 These results indicate that the pro-
tective effects of trimetazidine against res-
tenosis occur indirectly in part through the
increase in adiponectin level and decrease
in proinflammatory processes and oxidative
stress.

Here, I have described substantial evidence


to support the concept that trimetazidine
Figure 4. Cardioprotective effects of trimetazidine. plays a positive role in reducing the extent
of restenosis after PCI and have suggested
involved in abnormal VSMC cell growth after balloon injury.7,23 some possible underlying mechanisms. However, there are
We have shown that trimetazidine treatment lowers, in a dose- several issues to note. There are differences between clinical
dependent manner, the cellular production of reactive oxy- balloon angioplasty procedures carried out on diseased vas-
gen species (ROS) in HUVECs treated with LysoPC,14 which culature in human patients and animal balloon injury models,
is known to generate ROS.8 Repeated administration of tri- and the carotid arteries in the balloon injury model cannot
metazidine reduces production of mitochondrial ROS in rats, fully represent the overt atherosclerotic changes found in hu-
and pretreatment with trimetazidineadded to cardioplegic mans.31 In addition, in the rat model, it is VSMCs alone that
solutionreduces oxidative damage in human patients.9,10 contribute to the response observed after balloon injury; in hu-
Also, the ischemia-induced increase in free radical produc- mans, the injurious response in diseased vasculature arises
tion is attenuated by trimetazidine treatment before the onset from the interactions among several cell types, including
of ischemia in rat hearts.24 The antioxidant effects of trimeta- VSMCs, endothelial cells, macrophages, and T cells.32 Further-
zidine occur in various cells of the cardiovascular system.11 more, although the in vitro studies with HUVECs are performed
under high-glucose conditions in an effort to mimic the hy-
Effect of trimetazidine on the inflammatory perglycemic state in diabetics, these conditions may not be
process and on adipocytokines an accurate reflection of the actual state in such patients.
Early after endothelial denudation, an infiltration of inflamma-
tory cells occurs in the vascular wall,25 with the presence of Conclusion
monocytes associated with the neointimal area.26 Neointimal Trimetazidine has a broad spectrum of pharmacological ac-
thickening has been shown to be lessened with the use of tivities that provide cardiovascular protection through its an-
anti-inflammatory agents that block the early recruitment of tioxidative and anti-inflammatory properties, enhancement
monocytes.27 Collectively, these findings suggest a causative of mitochondrial function, reduced fatty acid oxidation, and
role for inflammation and its associated monocyte infiltration improved endothelial dysfunction (Figure 4). Therefore, using
in the development of restenosis. trimetazidine alone or in combination with other antiplatelet
agents or statins may ameliorate endothelial dysfunction and
Previous studies have reported that specific markersinclud- improve antiatherosclerotic and antithrombotic efficacy. Though
ing the adipocytokines adiponectin, monocyte chemoattrac- there is no direct evidence of a trimetazidine benefit for cardio-
tant protein-1 (MCP-1), and TNF-a, and also the inflamma- vascular morbidity and mortality, we speculate that trimetazi-
tory marker high-sensitivity C-reactive protein (hsCRP)can dines effect on endothelial function could affect this indirectly.
affect the development of restenosis and atherosclerosis.28-30 Prospective studies with CVD events as a primary objective
A low adiponectin concentration is a risk factor for the sub- are needed to confirm these potentially beneficial effects of
sequent development of CVDs.28 MCP-1 is involved in mono- trimetazidine. n

References
1. World Health Organization. Cardiovascular diseases (CVDs). Fact sheet No. 317. 2007;370(9591):937-948.
http://www.who.int/mediacentre/factsheets/fs317/en/. Updated January 2015. 4. Kastrati A, Mehilli J, Pache J, et al. Analysis of 14 trials comparing sirolimus-
2. Mozaffarian D, Benjamin EJ, Go AS, et al. Heart disease and stroke statistics eluting stents with bare-metal stents. N Engl J Med. 2007;356(10):1030-1039.
2015 update: a report from the American Heart Association. Circulation. 2015; 5. Lee CH, Tan HC, Lim YT. Update on drug-eluting stents for prevention of re-
131(4):e29-322. stenosis. Asian Cardiovasc Thorac Ann. 2006;14(1):75-82.
3. Stettler C, Wandel S, Allemann S, et al. Outcomes associated with drug-elut- 6. Dangas GD, Claessen BE, Caixeta A, Sanidas EA, Mintz GS, Mehran R. In-stent
ing and bare-metal stents: a collaborative network meta-analysis. Lancet. restenosis in the drug-eluting stent era. J Am Coll Cardiol. 2010;56(23):1897-1907.

280 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Trimetazidine and endothelial function Lim
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

7. Griendling KK, Ushio-Fukai M. Redox control of vascular smooth muscle terv Cardiol. 2012;25(4):411-417.
proliferation. J Lab Clin Med. 1998;132(1):9-15. 20. Chen J, Zhou S, Jin J, et al. Chronic treatment with trimetazidine after dis-
8. Matsubara M, Hasegawa K. Benidipine, a dihydropyridine-calcium channel charge reduces the incidence of restenosis in patients who received coronary
blocker, prevents lysophosphatidylcholine-induced injury and reactive oxygen stent implantation: a 1-year prospective follow-up study. Int J Cardiol. 2014;
species production in human aortic endothelial cells. Atherosclerosis. 2005; 174(3):634-639.
178(1):57-66. 21. Kanellakis P, Nestel P, Bobik A. Angioplasty-induced superoxide anions and
9. Guarnieri C, Muscari C. Beneficial effects of trimetazidine on mitochondrial func- neointimal hyperplasia in the rabbit carotid artery: suppression by the isoflavone
tion and superoxide production in the cardiac muscle of monocrotaline-treat- trans-tetrahydrodaidzein. Atherosclerosis. 2004;176(1):63-72.
ed rats. Biochem Pharmacol. 1988;37(24):4685-4688. 22. Cai H, Harrison DG. Endothelial dysfunction in cardiovascular diseases: the role
10. Fabiani JN, Farah B, Vuilleminot A, et al. Chromosomal aberrations and neu- of oxidant stress. Circ Res. 2000;87(10):840-844.
trophil activation induced by reperfusion in the ischaemic human heart. Eur 23. Satoh K, Nigro P, Berk BC. Oxidative stress and vascular smooth muscle cell
Heart J. 1993;14(suppl G):12-17. growth: a mechanistic linkage by cyclophilin A. Antioxid Redox Signal. 2010;
11. Onay-Besikci A, Ozkan SA. Trimetazidine revisited: a comprehensive review 12(5):675-682.
of the pharmacological effects and analytical techniques for the determina- 24. Maupoil V, Rochette L, Tabard A, Clauser P, Harpey C. Evolution of free radi-
tion of trimetazidine. Cardiovasc Ther. 2008;26(2):147-165. cal formation during low-flow ischemia and reperfusion in isolated rat heart.
12. Kantor PF, Lucien A, Kozak R, Lopaschuk GD. The antianginal drug trimetazi- Cardiovasc Drugs Ther. 1990;4(suppl 4):791-795.
dine shifts cardiac energy metabolism from fatty acid oxidation to glucose ox- 25. Moreno PR, Fallon JT, Murcia AM, et al. Tissue characteristics of restenosis af-
idation by inhibiting mitochondrial long-chain 3-ketoacyl coenzyme A thiolase. ter percutaneous transluminal coronary angioplasty in diabetic patients. J Am
Circ Res. 2000;86(5):580-588. Coll Cardiol. 1999;34(4):1045-1049.
13. Onay-Besikci A, Guner S, Arioglu E, Ozakca I, Ozcelikay AT, Altan VM. The ef- 26. Rogers C, Welt FG, Karnovsky MJ, Edelman ER. Monocyte recruitment and
fects of chronic trimetazidine treatment on mechanical function and fatty acid neointimal hyperplasia in rabbits. Coupled inhibitory effects of heparin. Arte-
oxidation in diabetic rat hearts. Can J Physiol Pharmacol. 2007;85(5):527-535. rioscler Thromb Vasc Biol. 1996;16(10):1312-1318.
14. Yoon JW, Cho BJ, Park HS, et al. Differential effects of trimetazidine on vascu- 27. Mori E, Komori K, Yamaoka T, et al. Essential role of monocyte chemoattrac-
lar smooth muscle cell and endothelial cell in response to carotid artery bal- tant protein-1 in development of restenotic changes (neointimal hyperplasia and
loon injury in diabetic rats. Int J Cardiol. 2013;167(1):126-133. constrictive remodeling) after balloon angioplasty in hypercholesterolemic rab-
15. Mizushige K, Yao L, Noma T, et al. Alteration in left ventricular diastolic filling bits. Circulation. 2002;105(24):2905-2910.
and accumulation of myocardial collagen at insulin-resistant prediabetic stage 28. Pischon T, Girman CJ, Hotamisligil GS, Rifai N, Hu FB, Rimm EB. Plasma adipo-
of a type II diabetic rat model. Circulation. 2000;101(8):899-907. nectin levels and risk of myocardial infarction in men. JAMA. 2004;291(14):
16. Clowes AW, Reidy MA, Clowes MM. Kinetics of cellular proliferation after ar- 1730-1737.
terial injury. I. Smooth muscle growth in the absence of endothelium. Lab In- 29. Pasceri V, Willerson JT, Yeh ET. Direct proinflammatory effect of C-reactive pro-
vest. 1983;49(3):327-333. tein on human endothelial cells. Circulation. 2000;102(18):2165-2168.
17. Belardinelli R, Solenghi M, Volpe L, Purcaro A. Trimetazidine improves en- 30. Tousoulis D, Antoniades C, Stefanadis C. Assessing inflammatory status in car-
dothelial dysfunction in chronic heart failure: an antioxidant effect. Eur Heart J. diovascular disease. Heart. 2007;93(8):1001-1007.
2007;28(9):1102-1108. 31. Nishida M, Miyagawa JI, Tokunaga K, et al. Early morphologic changes of ath-
18. Park KH, Park WJ, Kim MK, et al. Effects of trimetazidine on endothelial dys- erosclerosis induced by ventromedial hypothalamic lesion in the spontaneous-
function after sheath injury of radial artery. Am J Cardiol. 2010;105(12):1723-1727. ly diabetic Goto-Kakizaki rat. J Lab Clin Med. 1997;129(2):200-207.
19. Park KH, Park DW, Kim MK, et al. Effects of sheath injury and trimetazidine on 32. Tulis DA. Rat carotid artery balloon injury model. Methods Mol Med. 2007;139:
endothelial dysfunction of radial artery after transradial catheterization. J In- 1-30.

Keywords: endothelium; inflammation; oxidative stress; percutaneous coronary intervention; restenosis; trimetazidine

LENDOTHLIUM COMME CIBLE THRAPEUTIQUE CHEZ LES PATIENTS


EN POST-ICP ET RLE DE LA TRIMTAZIDINE DANS LA FONCTION ENDOTHLIALE
Lendothlium vasculaire, la monocouche de surface de la paroi vasculaire, joue un rle important dans le maintien
de la sant vasculaire. Il libre diffrents mdiateurs impliqus dans la vasodilatation ou la vasoconstriction en condi-
tions particulires, comme langiotensine 2, lendothline-1, le facteur hyperpolarisant driv de lendothlium, le
dioxyde dazote, la prostaglandine H2 et le thromboxane A2 . Un dysfonctionnement de lendothlium a t impliqu
dans diffrents processus physiopathologiques vasculaires, y compris une prolifration vasculaire anormale, une for-
mation excessive de thrombus, une vasoconstriction et un vasospasme. Ce dysfonctionnement sassocie aussi une
restnose aprs intervention coronaire percutane (ICP). Pourtant, les tudes cliniques sur lutilisation des agents
pharmacologiques pour amliorer la fonction endothliale et diminuer le taux de restnose aprs ICP sont peu nom-
breuses. La trimtazidine, un agent au spectre dactivit pharmacologique large protgeant contre les atteintes du
systme cardiovasculaire, amliore la fonction cellulaire endothliale vasculaire et rduit le risque de restnose aprs
ICP grce ses proprits antioxydantes et anti-inflammatoires, qui augmentent les taux dadiponectine et dimi-
nuent la rsistance linsuline. Aucune donne directe nindique que la trimtazidine rduit la morbimortalit car-
diovasculaire, mais une diminution de la morbidit et de la mortalit pourrait rsulter indirectement de son effet sur
la fonction endothliale. Des tudes prospectives ayant pour objectif principal dvaluer la morbimortalit cardio-
vasculaire sont ncessaires pour confirmer les effets cardioprotecteurs de la trimtazidine.

Trimetazidine and endothelial function Lim MEDICOGRAPHIA, Vol 38, No. 3, 2016 281

M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

[We have] sufficient reason to


believe that for elective percuta-
neous coronary intervention (PCI)
patients, pre- or perioperative treat-
ment with trimetazidine reduces
PCI-related myocardial and vascu-
Metabolic agents in the
lar injury and improves heart func-
tion; and that for acute coronary
syndrome patients, an acute oral
protection of patients who
loading dose of trimetazidine or
long-term treatment with trimetazi- undergo revascularization
dine after stent implantation would
also benefit these patients.

b y Y. C h e n , C h i n a

I
n clinical practice, metabolic agents, including trimetazidine, ranolazine,
and glucose-insulin-potassium, often play an adjunctive role in the treat-
ment of angina pectoris. With the worldwide use of percutaneous coronary
intervention, especially stent implantation, doctors have come to realize that
some patients still suffer from angina, percutaneous coronary intervention
related complications, or poor quality of life. For such reasons, researchers
have used metabolic agents to treat patients who have undergone percuta-
neous coronary interventions, and some progress has been observed in this
area. In this review article, we mainly discuss the protective role of three wide-
ly used agents: trimetazidine, ranolazine, and glucose-insulin-potassium in pa-
Yundai CHEN, MD, PhD tients with coronary artery disease, with a particular focus on patients who
Department of Cardiology
have undergone percutaneous coronary intervention. Aside from these three,
Chinese PLA General Hospital
Beijing, CHINA there are other metabolic agents requiring further investigation to confirm their
benefits for patients undergoing revascularization.
Medicographia. 2016;38:282-287 (see French abstract on page 287)

n recent years, evidence-based medicine has demonstrated that revasculariza-

I tion can save lives in patients with acute coronary syndrome (ACS) and improve
their quality of life. Research has shown that some metabolic agents could further
benefit those patients and reduce ischemia-reperfusion injury or percutaneous coro-
nary intervention (PCI)-related injury. Metabolic agents proven to be useful in pa-
tients who undergo revascularization include trimetazidine, ranolazine, L-carnitine,
glucose-insulin-potassium (GIK), ribose, dichloroacetate, and perhexiline, of which
trimetazidine, ranolazine, and GIK were the most evaluated in recent years.

Metabolic agents as adjunctive therapy in patients undergoing PCI


u Trimetazidine
Trimetazidine, a piperazine derivative, has a long history of research, especially with
regard to cardiac protection. Used worldwide as an adjunctive therapy for stable
angina, trimetazidines protective effect in patients that undergo PCI has become
a research focus.

Address for correspondence: In a small (n=20), randomized, double-blind, placebo-controlled trial, Kober et al found
Professor Yundai Chen, Department
of Cardiology, Chinese PLA General that trimetazidine treatment during percutaneous transluminal coronary angioplas-
Hospital, Beijing, China, 100853. ty (PTCA) decreases intervention-related myocardial ischemia.1 Polonski et al re-
(email: cyundai@vip.163.com) ported that pretreatment with trimetazidine appears to be cardioprotective, helping
www.medicographia.com to prevent myocardial ischemia during PTCA.2 In a study by Steg and his colleagues,

282 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Metabolic agents in the protection of patients who undergo revascularization Chen
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

patients with acute myocardial infarction (AMI) were pre- rat model, Khan et al found that trimetazidine administered
scribed intravenous trimetazidine as an adjunctive therapy to at the onset of reperfusion ameliorates myocardial dysfunc-
primary angioplasty. The authors demonstrated that trimetazi- tion and injury by activation of p38 mitogen-activated pro-
dine was safe and led to earlier resolution of ST-segment ele- tein kinase and Akt signaling.11 In a recent study, Yang et al
vation.3 Later, in a prospective study, Labrou et al demonstrat- revealed that by upregulating microRNA-21 (miR-21) expres-
ed that pretreatment with trimetazidine minimizes myocardial sion, trimetazidine counteracts the apoptotic effect of hypox-
reperfusion injury during PCI and improves global and region- ia/reperfusion.12 Consistent with previous studies, Senturk et
al wall motion at 1 and 3 months after PCI.4 This result was fur- al found that combination of N-acetylcysteine and trimetazi-
ther supported by a multicenter, randomized, and controlled dine effectively decreases oxidative stress, infarct area, and
study aiming to evaluate the myocardial protection of trimetazi- apoptotic activity in a rat model of ischemic reperfusion.13 Tri-
dine during PCI; the results suggested that perioperative tri- metazidine also improves endothelium-dependent relaxation
metazidine therapy can reduce the frequency of angina at- in patients with ischemic cardiomyopathy, owing to its anti-
tacks and myocardial damage during PCI and improve left oxidant properties.14 A recent experimental study suggested
ventricular function during follow-up after PCI.5 A study specif- that trimetazidine ameliorates intracellular calcium (Ca2+) home-
ically aimed at the effect of trimetazidine on recurrent angina ostasis via a switch from lipid metabolism to glucose metab-
and left ventricular structure in elderly multivessel coronary olism, thereby producing its cardioprotective effect and reduc-
heart disease patients with diabetes mellitus after drug-elut- ing damage to hypoxic cardiomyocytes.15
ing stent implantation also found that adjunctive therapy
with trimetazidine had a beneficial effect in these patients.6 On the basis of these findings, Kim et al analyzed data from
the Korean Acute Myocardial Infarction Registry and found
For elective PCI patients, pretreatment with trimetazidine is a that trimetazidine improves clinical outcomes in AMI patients
reasonable option; but what about acute PCI patients? To an- by significantly reducing all-cause mortality and major ad-
swer this question, Bonello et al carried out a study focus- verse cardiac events (MACEs) over 12 months.16 For specific
ing on the protective effect of an acute oral loading dose of patients, such as diabetic patients with renal dysfunction un-
trimetazidine on myocardial injury induced by PCI. The results dergoing elective PCI, a study demonstrated that trimetazi-
suggested that preprocedural acute oral trimetazidine admin- dine administered before elective PCI decreases the incidence
istration significantly reduces PCI-induced myocardial injury.7 of contrast-induced nephropathy (CIN).17
Aside from patients undergoing PCI, researchers also stud-
ied trimetazidines effect on ischemic injury and reperfusion in Our team also demonstrated that long-term treatment with
patients undergoing coronary artery bypass graft (CABG) sur- trimetazidine after stent implantation reduced in-stent resteno-
gery, with the same result.8,9 sis and MACE in a 1-year follow-up study.18 In this study, 768
patients were enrolled and randomized into a trimetazidine
u Cardioprotective mechanism group (n=384) or a control group. After drug-eluting stent im-
A number of investigations have looked into the mechanism in- plantation, all patients were treated with regular medication.
volved in the cardioprotection afforded by trimetazidine. Tritto
et al showed us that trimetazidine protects the post-ischemic In the trimetazidine group, 20 mg trimetazidine was admin-
heart from neutrophil-mediated injury.10 In an ischemia-injury istered three times a day for at least 30 days. All patients re-
ceived follow-up angiography 9-13 months after discharge.
The final analysis included 635 patients (trimetazidine group,
SELECTED ABBREVIATIONS AND ACRONYMS
n=312; control group, n=323). Stent restenosis occurred in
ACS acute coronary syndrome 49 (7.7%) patients. The trimetazidine group had a lower in-
AMI acute myocardial infarction cidence of stent restenosis than the control group (4.2% vs
CABG coronary artery bypass graft 11.1%; P=0.001). At the 30-day follow-up, the trimetazidine
CIN contrast-induced nephropathy group exhibited a higher left ventricular ejection fraction than
GIK glucose-insulin-potassium the control group (65.410.7 vs 63.110.4; P=0.006). The in-
IMMEDIATE Immediate Myocardial Metabolic Enhancement cidence of major adverse cardiac and cerebrovascular events
During Initial Assessment and Treatment in (MACCEs) was also lower in the trimetazidine group at the
Emergency care [trial] 1-year follow-up (6.1% vs 10.8%; P=0.032). Further multivari-
MACE major adverse cardiac event ate analysis revealed that trimetazidine treatment was a pre-
PCI percutaneous coronary intervention dictor for stent restenosis (odds ratio, 0.376; 95% confidence
POAF post-revascularization atrial fibrillation interval, 0.196-0.721; P=0.003). This result was also support-
PTCA percutaneous transluminal coronary angioplasty ed by a Sprague-Dawley rat model experiment that demon-
RIVER-PCI Ranolazine for Incomplete Vessel Revascular- strated that trimetazidine inhibits the proliferation and mi-
ization [trial] gration of vascular smooth muscle cells and promotes the
proliferation of human umbilical vein endothelial cells.19

Metabolic agents in the protection of patients who undergo revascularization Chen MEDICOGRAPHIA, Vol 38, No. 3, 2016 283
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

Aside from these benefits for the heart, trimetazidine also pro- of ranolazine in post-revascularization atrial fibrillation (POAF).
tects the artery from PCI-related injury. Yoon et al reported Tagarakis et al found a protective role for oral ranolazine when
that trimetazidine effectively accelerates re-endothelialization administered preoperatively at a moderate dose in patients
after carotid balloon injury.20 Recent studies have also demon- undergoing on-pump CABG surgery. Their findings suggest
strated that trimetazidine significantly lessens endothelial dys- that perioperative treatment with ranolazine effectively re-
function in the radial artery after catheterization.21,22 In a re- duces the incidence of POAF,36 a result that has been sup-
cent meta-analysis, the authors concluded from nine studies ported by further studies.37-39 In patients that underwent PCI,
involving a total of 778 patients that adjunctive therapy with ranolazine has been found to reduce recurrent ischemic events
trimetazidine in patients undergoing PCI may reduce myocar- and improve quality of life.40,41
dial injury during the procedure and improve cardiac func-
tion.23 However, as presented at the 2015 American Heart Asso-
ciation (AHA) Scientific Sessions, the RIVER-PCI study (Ra-
u Other protective effects nolazine for Incomplete Vessel Revascularization) showed
Besides the protective effect on the cardiovascular system, no incremental benefit in angina or quality of life measures from
recent studies suggest that trimetazidine may have poten- adding ranolazine treatment in an angiographically-identified
tial for use in prevention of CIN. In one meta-analysis includ- population.42 Furthermore, an overall analysis of this study re-
ing three randomized controlled trials in the final analysis, the vealed that ranolazine did not reduce the composite rate of
addition of trimetazidine treatment significantly decreased ischemia-driven revascularization or hospitalization without
the incidence of CIN in patients that underwent coronary an- revascularization in patients with a history of chronic angina
giography.24 The authors pointed out that care should be tak- who had incomplete revascularization after PCI.43
en in the interpretation of this result, taking into account the
small sample size. u Final comments about ranolazine
In our opinion, despite a confirmed role for ranolazine in angi-
u Final comments about trimetazidine na frequency and in quality of life, further investigationwell-
Collectively, these studies provide sufficient reason to believe designed clinical trials, especiallyare warranted to evaluate
that for elective PCI patients, pre- or perioperative treatment its effect in CAD patients undergoing PCI. The use of ranola-
with trimetazidine reduces PCI-related myocardial and vas- zine in patients that are to undergo PCI should not be en-
cular injury and improves heart function; and that for ACS couraged for now.
patients, an acute oral loading dose of trimetazidine or long-
term treatment with trimetazidine after stent implantation would u Glucose-insulin-potassium
also benefit these patients; however, the use of trimetazidine GIK has been used as metabolic therapy in practice for many
for preventing CIN is not recommended as first-line therapy years. Earlier studies found that GIK improves hemodynam-
and still needs to be assessed in more clinical trials. ic performance and is associated with reduced troponin I
release after on-pump CABG surgery.44,45 It also improves
u Ranolazine myocardial perfusion after revascularization and lessens the
Ranolazine is another drug used as an adjunctive therapy for LV remodeling observed at follow-up.46,47 These results are
angina in symptomatic patients who are inadequately con- supported by other experimental research.48,49 However, a
trolled with first-line antianginal therapies.25,26 Among diabet- 1-year follow-up study found that GIK therapy offers no clin-
ic patients that have chronic angina despite treatment with up ical benefit in patients with ST-elevated myocardial infarction
to two agents, ranolazine was found to reduce angina and (STEMI) without signs of heart failure.50 Further meta-analysis
sublingual nitroglycerin use and to be well tolerated.27 A sys- also suggested that GIK does not reduce mortality in patients
tematic review of randomized controlled trials included sev- with AMI.51 Despite these negative results, some important
en studies and concluded that ranolazine reduces anginal studies were carried out to determine the effect of GIK on pa-
symptoms among patients with symptomatic chronic stable tients with CAD. In the IMMEDIATE randomized controlled
angina pectoris28 and is probably cost effective.29 Recent stud- trial (Immediate Myocardial Metabolic Enhancement During
ies have shown additional benefits of ranolazine in patients Initial Assessment and Treatment in Emergency care), Selker
with coronary heart disease. Some experimental studies have et al found that in patients with suspected ACS, out-of-hos-
demonstrated that ranolazine reduces myocardial infarct size pital treatment with GIK did not reduce progression to MI and
and improves left ventricular function.30,31 It also markedly although it did not improve 30-day survival, it was associat-
reduces ventricular arrhythmias induced by ischemia and ed with lower rates of the composite outcome comprising
ischemia-reperfusion, indicating a protective role in PCI in pa- cardiac arrest and in-hospital mortality.52 Similar results were
tients with ACS.32 Possible mechanisms involved in these phe- found in a 1-year follow-up of this study, whereas in those
nomena include reduction in Ca2+ overload and oxidative stress with STEMI, the composite of cardiac arrest or 1-year mortal-
and improvement in mitochondrial integrity.33-35 On the basis ity, and of cardiac arrest, mortality, or hospitalization for heart
of these findings, some clinical research focused on the role failure within 1 year, were significantly reduced. This benefit

284 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Metabolic agents in the protection of patients who undergo revascularization Chen
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

might be limited to AMI patients.53 A further meta-analysis also may involve the different number of trials included in the meta-
revealed that administration of GIK in ACS patients did not analyses (13 vs 5 respectively). On the basis of these incon-
significantly reduce mortality after the onset of symptoms.54 sistent findings, we suggest that clinical trialswell-designed
randomized controlled trials, especiallyare needed to fur-
u Final comments about GIK ther determine the effect of L-carnitine treatment in patients
On the basis of the above, we believe that presently we do with CAD, especially those planned to undergo PCI.
not have enough evidence to support the first-line use of GIK
in patients undergoing PCI. Studies to further investigate the Recent studies have demonstrated that the metabolic agent
role of GIK in these patients are needed. dichloroacetate improves cardiac contractile dysfunction af-
ter ventricular fibrillation59 and also prevents restenosis in pre-
u Other metabolic agents clinical animal models of vessel injury.60 However, until more
L-Carnitine is another adjunctive therapy for angina pectoris evidence is available, care should be exercised when con-
and has been shown to attenuate left ventricular dilation dur- sidering its clinical use in patients undergoing PCI.
ing the first year after an AMI, resulting in smaller left ventric-
ular volumes at follow-up.55 In the stent era, L-carnitine has Additional metabolic agents, such as perhexiline, ribose, and
also been used in patients undergoing PCI, with inconsis- others, still need further investigation to confirm their roles in
tent results. Xue et al found that L-carnitine as an adjunctive patients with CAD, especially for those undergoing PCI.
therapy to PCI was associated with a reduced level of cardiac
markers in patients with non-STEMI.56 A later systematic re- Conclusions
view and meta-analysis found that, compared with placebo There are a number of metabolic agents widely used in ad-
or control, L-carnitine was associated with a 27% reduction junctive therapy for anginal pectoris. Their use in patients that
in all-cause mortality, a 65% reduction in ventricular arrhyth- have undergone PCI has been under investigation for some
mias, and a 40% reduction in anginal symptoms in patients time, with differing levels of support available for the various
experiencing an AMI.57 However, these findings were not con- agents. With regard to the agents discussed in this article,
sistent with results from another meta-analysis in which the we believe a sufficient amount of evidence has accumulated
authors concluded that there was no significant marginal ben- in support of a protective role for trimetazidine, though its use
efit in terms of all-cause mortality, heart failure, unstable angina, in preventing CIN should be further investigated. In our opin-
or myocardial reinfarction in the setting of AMI for oral L-car- ion, ranolazine, GIK, and L-carnitine, among other metabolic
nitine maintenance doses of 2 g or greater per day.58 A pos- agents briefly touched on here, require further investigation re-
sible reason behind the differing results of these two studies garding their use in patients undergoing revascularization. n

References
1. Kober G, Pennaforte S, Buck T, Sievert H, Vallbracht C. Myocardial cytopro- 10. Tritto I, Wang P, Kuppusamy P, Giraldez R, Zweier JL, Ambrosio G. The anti-
tection during percutaneous transluminal coronary angioplasty. Eur Heart J. anginal drug trimetazidine reduces neutrophil-mediated cardiac reperfusion
1993;14(Suppl G):6-11. injury. J Cardiovasc Pharmacol. 2005;46(1):89-98.
2. Poloski L, Dec I, Wojnar R, Wilczek K. Trimetazidine limits the effects of my- 11. Khan M, Meduru S, Mostafa M, Khan S, Hideg K, Kuppusamy PJ. Trimetazi-
ocardial ischaemia during percutaneous coronary angioplasty. Curr Med Res dine, administered at the onset of reperfusion, ameliorates myocardial dysfunc-
Opin. 2002;18(7):389-396. tion and injury by activation of p38 mitogen-activated protein kinase and Akt
3. Steg PG, Grollier G, Gallay P, et al; LIST Study Group. A randomized double- signaling. Pharmacol Exp Ther. 2010;333(2):421-429.
blind trial of intravenous trimetazidine as adjunctive therapy to primary angio- 12. Yang Q, Yang K, Li AY. Trimetazidine protects against hypoxia-reperfusion-in-
plasty for acute myocardial infarction. Int J Cardiol. 2001;77(2-3):263-273. duced cardiomyocyte apoptosis by increasing microRNA-21 expression. Int
4. Labrou A, Giannoglou G, Zioutas D, Fragakis N, Katsaris G, Louridas G. Trimetazi- J Clin Exp Pathol. 2015;8(4):3735-3741.
dine administration minimizes myocardial damage and improves left ventric- 13. entrk T, avun S, Avc B, Yermezler A, Serdar Z, Savc V. Effective inhibition
ular function after percutaneous coronary intervention. Am J Cardiovasc Drugs. of cardiomyocyte apoptosis through the combination of trimetazidine and N-
2007;7(2):143-150. acetylcysteine in a rat model of myocardial ischemia and reperfusion injury.
5. Chen YD, Zhao LK, Tian F, Du ZM, Jiang H, Wei M. Evaluation of the myocar- Atherosclerosis. 2014;237(2):760-766.
dial protection of trimetazidine during percutaneous coronary intervention: a 14. Belardinelli R, Solenghi M, Volpe L, Purcaro A. Trimetazidine improved endo-
multi-center randomized and controlled clinical study. Chin J Itern Med. 2010; thelial dysfunction in chronic heart failure: an antioxidant effect. Eur Heart J.
49(6):473-476. 2007;28(9):1102-1108.
6. Xu X, Zhang W, Zhou Y, et al. Effect of trimetazidine on recurrent angina pec- 15. Wei J, Xu H, Shi L, Tong J, Zhang J. Trimetazidine protects cardiomyocytes
toris and left ventricular structure in elderly multivessel coronary heart disease against hypoxia-induced injury through ameliorates calcium homeostasis. Chem
patients with diabetes mellitus after drug-eluting stent implantation: a single- Biol Interact. 2015;236:47-56.
center, randomized, double-blind study at 2-year follow-up. Clin Drug Investig. 16. Kim JS, Kim CH, Chun KJ, et al. Effects of trimetazidine in patients with acute
2014,34:251-258. myocardial infarction: data from the Korean Acute Myocardial Infarction Reg-
7. Bonello L, Sbragia P, Amabile N, et al. Protective effect of an acute oral load- istry. Clin Res Cardiol. 2013;102(12):915-922.
ing dose of trimetazidine on myocardial injury following percutaneous coronary 17. Shehata M. Impact of trimetazidine on incidence of myocardial injury and con-
intervention. Heart. 2007;93(6):703-707. trast-induced nephropathy in diabetic patients with renal dysfunction undergoing
8. Martins GF, Siqueira Filho AG, Santos JB, et al. Trimetazidine on ischemic in- elective percutaneous coronary intervention. Am J Cardiol. 2014;114(3):389-394.
jury and reperfusion in coronary artery bypass grafting. [Article in English and 18. Chen J, Zhou S, Jin J, et al. Chronic treatment with trimetazidine after discharge
Portuguese]. Arq Bras Cardiol. 2011;97(3):209-216. reduces the incidence of restenosis in patients who received coronary stent
9. Zhang N, Lei J, Liu Q, Huang W, Xiao H, Lei H. The effectiveness of preoper- implantation: a 1-year prospective follow-up study. Int J Cardiol. 2014;174(3):
ative trimetazidine on myocardial preservation in coronary artery bypass graft 634-639.
patients: a systematic review and meta-analysis. Cardiology. 2015;131(2):86-96. 19. Yoon JW, Cho BJ, Park HS, et al. Differential effects of trimetazidine on vascu-

Metabolic agents in the protection of patients who undergo revascularization Chen MEDICOGRAPHIA, Vol 38, No. 3, 2016 285
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

lar smooth muscle cell and endothelial cell in response to carotid artery bal- 2015;11: 469-474.
loon injury in diabetic rats. Int J Cardiol. 2013;167(1):126-133. 41. Gutierrez JA, Karwatowska-Prokopczuk E, Murphy SA, et al. Effects of ra-
20. Yoon JW, Cho BJ, Park HS, et al. Differential effects of trimetazidine on vas- nolazine in patients with chronic angina in patients with and without percuta-
cular smooth muscle cell and endothelial cell in response to carotid artery neous coronary intervention for acute coronary syndrome: observations from
balloon injury in diabetic rats. Int J Cardiol. 2013;167(1):126-133. the MERLIN-TIMI 36 Trial. Clin Cardiol. 2015;38(8):469-475.
21. Park KH, Park WJ, Kim MK, et al. Effects of trimetazidine on endothelial dys- 42. Alexander KP, Weisz G, Prather K, et al. Effects of ranolazine on angina and
function after sheath injury of radial artery. Am J Cardiol. 2010;105:1723-1727. quality of life after percutaneous coronary intervention with incomplete revas-
22. Park KH, Park DW, Kim MK, et al. Effects of sheath injury and trimetazidine on cularization: results from the Ranolazine for Incomplete Vessel Revasculariza-
endothelial dysfunction of radial artery after transradial catheterization. J Interv tion (RIVER-PCI) trial. Circulation. 2016;133(1):39-47.
Cardiol. 2012;25:411-417. 43. Weisz G, Genereux P, Iiguez A, et al. Ranolazine in patients with incomplete
23. Zhang Y, Ma XJ, Shi DZ. Effect of trimetazidine in patients undergoing percuta- revascularization after percutaneous coronary intervention (RIVER-PCI): a multi-
neous coronary intervention: a meta-analysis. PLoS One. 2015;10(9):e0137775. center, randomized, double-blind, placebo-controlled trial. Lancet. 2016;387
24. Nadkarni GN, Konstantinidis I, Patel A, et al. Trimetazidine decreases risk of con- (10014):136-145.
trast-induced nephropathy in patients with chronic kidney disease: a meta- 44. Ranasinghe AM, Quinn DW, Pagano D, et al. Glucose-insulin-potassium and
analysis of randomized controlled trials. J Cardio Pharmacol Ther. 2015;20(6): tri-iodothyronine individually improve hemodynamic performance and are as-
539-546. sociated with reduced troponin I release after on-pump coronary artery by-
25. Chaitman BR, Pepine CJ, Parker JO, et al; Combination Assessment of Rano- pass grafting. Circulation. 2006;114(1 suppl):I245-I250.
lazine In Stable Angina (CARISA) Investigators. Effects of ranolazine with atenolol, 45. Ranasinghe AM, McCabe CJ, Quinn DW, et al. How does glucose insulin potas-
amlodipine, or diltiazem on exercise tolerance and angina frequency in patients sium improve hemodynamic performance? Evidence for altered expression
with severe chronic angina: a randomized controlled trial. JAMA. 2004;291(3): of beta-adrenoreceptor and calcium handling genes. Circulation. 2006;114(1
309-316. suppl):I239-I244.
26. Stone PH, Gratsiansky NA, Blokhin A, Huang IZ, Meng L; ERICA Investigators. 46. Bucciarelli-Ducci C, Bianchi M, De Luca L, et al. Effects of glucose-insulin-
Antianginal efficacy of ranolazine when added to treatment with amlodipine: potassium infusion on myocardial perfusion and left ventricular remodeling in
the ERICA (Efficacy of Ranolazine in Chronic Angina) trial. J Am Coll Cardiol. patients treated with primary angioplasty for ST-elevation acute myocardial
2006;48(3):566-575. infarction. Am J Cardiol. 2006;98(10):1349-1353.
27. Kosiborod M, Arnold SV, Spertus JA, et al. Evaluation of ranolazine in patients 47. Li Y, Zhang L, Zhang L, et al. High-dose glucose-insulin-potassium has hemo-
with type 2 diabetes mellitus and chronic stable angina: results from the TERISA dynamic benefits and can improve cardiac remodeling in acute myocardial in-
randomized clinical trial (Type 2 Diabetes Evaluation of Ranolazine in Subjects farction treated with primary percutaneous coronary intervention: from a ran-
With Chronic Stable Angina). J Am Coll Cardiol. 2013;61(20):2038-2045. domized controlled study. J Cardiovasc Dis Res. 2010;1(3):104-109.
28. Banon D, Filion KB, Budlovsky T, Franck C, Eisenberg MJ. The usefulness of 48. Eiferman D, Perez-Tamayo RA, Abe K, Okum E, Higgins R. Real-time moni-
ranolazine for the treatment of refractory chronic stable angina pectoris as toring of cardiac metabolism using biosensors shows myocardial protection
determined from a systematic review of randomized controlled trials. Am J Car- during ischemia-reperfusion injury with glucose-insulin-potassium administra-
diol. 2014;113(6):1075-1082. tion. Surgery. 2007;142(2):150-155.
29. Coleman CI, Freemantle N, Kohn CG. Ranolazine for the treatment of chron- 49. Suranadi IW, Demaison L, Chat V, Peltier S, Richardson M, Leverve X. An in-
ic stable angina: a cost-effectiveness analysis from the UK perspective. BMJ crease in the redox state during reperfusion contributes to the cardioprotec-
Open. 2015;5(11):e008861. tive effect of GIK solution. J Appl Physiol (1985). 2012;113(5):775-784.
30. Hale SL, Leeka JA, Kloner RA. Improved left ventricular function and reduced ne- 50. Rasoul S, Ottervanger JP, Timmer JR, et al. One year outcomes after glucose-
crosis after myocardial ischemia/reperfusion in rabbits treated with ranolazine, an insulin-potassium in ST elevation myocardial infarction. The Glucose-insulin-
inhibitor of the late sodium channel. J Pharmacol Exp Ther. 2006;318(1):418-423. potassium study II. Int J Cardiol. 2007;122(1):52-55.
31. Hale SL, Kloner RA. The antianginal agent, ranolazine, reduces myocardial in- 51. Zhao YT, Weng CL, Chen ML, et al. Comparison of glucose-insulin-potassium
farct size but does not alter anatomic no-reflow or regional myocardial blood and insulin-glucose as adjunctive therapy in acute myocardial infarction: a con-
flow in ischemia/reperfusion in the rabbit. J Cardiovasc Pharmacol Ther. 2008; temporary meta-analysis of randomised controlled trials. Heart. 2010;96(20):
13(3):226-232. 1622-1626.
32. Dhalla AK, Wang WQ, Dow J, et al. Ranolazine, an antianginal agent, marked- 52. Selker HP, Beshansky JR, Sheehan PR, et al. Out-of-hospital administration of
ly reduces ventricular arrhythmias induced by ischemia and ischemia-reperfu- intravenous glucose-insulin-potassium in patients with suspected acute coro-
sion. Am J Physiol Heart Circ Physiol. 2009;297(5):H1923-H1929. nary syndromes: the IMMEDIATE randomized controlled trial. JAMA. 2012;307
33. Aldakkak M, Camara AK, Heisner JS, Yang M, Stowe DF. Ranolazine reduces (18):1925-1933.
Ca2+ overload and oxidative stress and improves mitochondrial integrity to pro- 53. Selker HP, Udelson JE, Massaro JM, et al. One-year outcomes of out-of-hos-
tect against ischemia reperfusion injury in isolated hearts. Pharmacol Res. 2011; pital administration of intravenous glucose, insulin, and potassium (GIK) in pa-
64(4):381-392. tients with suspected acute coronary syndromes (from the IMMEDIATE [Im-
34. Gadicherla AK, Stowe DF, Antholine WE, Yang M, Camara AK. Damage to mi- mediate Myocardial Metabolic Enhancement During Initial Assessment and
tochondrial complex I during cardiac ischemia reperfusion injury is reduced in- Treatment in Emergency Care] Trial). Am J Cardiol. 2014;113(10):1599-1605.
directly by anti-anginal drug ranolazine. Biochim Biophys Acta. 2012;1817(3): 54. Jin PY, Zhang HS, Guo XY, Liang WF, Han QF. Glucose-insulin-potassium
419-429. therapy in patients with acute coronary syndrome: a meta-analysis of random-
35. Caldern-Snchez EM, Domnguez-Rodrguez A, Lpez-Haldn J, et al. Car- ized controlled trials. BMC Cardiovasc Disord. 2014;14:169.
dioprotective Effect of Ranolazine in the Process of Ischemia-reperfusion in 55. Iliceto S, Scrutinio D, Bruzzi P, et al. Effects of L-carnitine administration on left
Adult Rat Cardiomyocytes. Rev Esp Cardiol (Engl Ed). 2016;69(1):45-53. ventricular remodeling after acute anterior myocardial infarction: the L-Carni-
36. Tagarakis GI, Aidonidis I, Daskalopoulou SS, et al. Effect of ranolazine in pre- tine Ecocardiografia Digitalizzata Infarto Miocardico (CEDIM) Trial. J Am Coll
venting postoperative atrial fibrillation in patients undergoing coronary revas- Cardiol. 1995;26(2):380-387.
cularization surgery. Curr Vasc Pharmacol. 2013;11(6):988-991. 56. Xue YZ, Wang LX, Liu HZ, Qi XW, Wang XH, Ren HZ. L-Carnitine as an ad-
37. Tsu LV, Lee S. Use of ranolazine in the prevention and treatment of postop- junct therapy to percutaneous coronary intervention for non-ST elevation my-
erative atrial fibrillation in patients undergoing cardiac surgery. Ann Pharma- ocardial infarction. Cardiovasc Drugs Ther. 2007;21(6):445-448.
cother. 2014;48(5):633-637. 57. DiNicolantonio JJ, Lavie CJ, Fares H, Menezes AR, OKeefe JH. L-Carnitine
38. Simopoulos V, Tagarakis GI, Daskalopoulou SS, et al. Ranolazine enhances the in the secondary prevention of cardiovascular disease: systematic review and
antiarrhythmic activity of amiodarone by accelerating conversion of new-on- meta-analysis. Mayo Clin Proc. 2013;88(6):544-551.
set atrial fibrillation after cardiac surgery. Angiology. 2014;65(4):294-297. 58. Shang R, Sun Z, Li H. Effective dosing of L-carnitine in the secondary preven-
39. Koskinas KC, Fragakis N, Katritsis D, Skeberis V, Vassilikos V. Ranolazine en- tion of cardiovascular disease: a systematic review and meta-analysis. BMC
hances the efficacy of amiodarone for conversion of recent-onset atrial fibril- Cardiovasc Disord. 2014;14:88.
lation. Europace. 2014;16(7):973-979. 59. Azam MA, Wagg CS, Mass S, et al. Feeding the fibrillating heart: Dichloroac-
40. Shammas NW, Shammas GA, Keyes K, Duske S, Kelly R, Jerin M. Ranola- etate improves cardiac contractile dysfunction following VF. Am J Physiol
zine versus placebo in patients with ischemic cardiomyopathy and persistent Heart Circ Physiol. 2015;309(9):H1543-H1553.
chest pain or dyspnea despite optimal medical and revascularization thera- 60. Deuse T, Hua X, Wang D, et al. Dichloroacetate prevents restenosis in pre-
py: randomized, double-blind crossover pilot study. Ther Clin Risk Manag. clinical animal models of vessel injury. Nature. 2014;509(7502):641-614.

286 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Metabolic agents in the protection of patients who undergo revascularization Chen
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

Keywords: adjunctive therapy; coronary artery disease; glucose-insulin-potassium; metabolic agent; percutaneous coro-
nary intervention; ranolazine; trimetazidine

UTILISATION DES AGENTS MTABOLIQUES POUR


LA CARDIOPROTECTION DES PATIENTS APRS UNE REVASCULARIZATION
En pratique clinique, les agents mtaboliques, dont la trimtazidine, la ranolazine et lassociation glucose-insuline-
potassium (GIK), jouent souvent un rle dappoint dans le traitement de langor. Lintervention coronaire percutane
tant trs utilise au niveau mondial, notamment limplantation de stents, les mdecins se sont rendu compte que
certains patients continuaient souffrir dangor et de complications lies lintervention coronaire percutane ou
quils avaient une mauvaise qualit de vie. Cest pourquoi des agents mtaboliques ont t tests pour traiter les
patients ayant subi ce type dinterventions et des progrs ont t observs. Dans cet article, nous analysons prin-
cipalement le rle protecteur de trois agents largement utiliss dans la maladie coronaire : la trimtazidine, la rano-
lazine et le GIK, avec un accent particulier sur les patients ayant subi une intervention coronaire percutane. Outre
ces trois traitements dappoint, il existe dautres agents mtaboliques dont les effets bnfiques sur les patients ayant
subi une revascularisation restent confirmer par des examens plus approfondis.

Metabolic agents in the protection of patients who undergo revascularization Chen MEDICOGRAPHIA, Vol 38, No. 3, 2016 287

M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

Contrast-induced nephropa-
thy (CIN) or contrast-induced acute
kidney injury is a common but un-
derdiagnosed complication of
coronary diagnostic and interven-
tional procedures that is associat-
How effective
ed with increased in-hospital mor-
bidity and mortality, prolonged
hospital stay, and raised health care
can trimetazidine be
costs.The search continues for
new pharmacologic and nonphar- in preventing myocardial and
macologic interventions for the
prevention of CIN in patients un-
dergoing [such procedures]. renal revascularization injury?

b y Y. Lo p a t i n , R u s s i a

C
urrently, percutaneous coronary intervention is considered one of the
key treatment strategies for the management of occlusive coronary ar-
tery disease. Even with the technical advances in percutaneous coro-
nary intervention that have made the procedure safe, with a minimal rate of
complications, myocardial revascularization procedures per se still cause my-
ocardial or renal injuries. Lately, particular attention has been paid to such
complications, including periprocedural myocardial injury and contrast-in-
duced nephropathy. These complications can occur frequently and are as-
sociated with a worse prognosis. In this regard, the search for strategies that
prevent the development of these periprocedural injuries seems very impor-
Yury LOPATIN, MD, PhD, FHFA tant. Results of experimental and clinical studies suggest promising poten-
Volgograd Medical State University
tial for trimetazidine in the prevention of periprocedural myocardial and renal
Volgograd Regional Cardiology
Centre, Volgograd injuries.
RUSSIA Medicographia. 2016;38:288-295 (see French abstract on page 295)

rimetazidine is an anti-ischemic metabolic modulator that has been approved

T worldwide for symptomatic treatment of chronic stable angina. Moreover, there


is sound evidence to consider appropriate the use of this agent in patients
with heart failure of ischemic etiology.1 In the past few years, several randomized
clinical trials (RCTs) have demonstrated that therapy with trimetazidine after per-
cutaneous coronary intervention (PCI) reduces the incidence of major adverse car-
diac events, recurrent angina pectoris, and stent restenosis, and that it improves
cardiac function.2-4

At the same time, evidence continues to accumulate that myocardial revascular-


ization procedures per se cause myocardial or renal injuries, which can be associ-
ated with worse clinical outcomes.

How effective can the use of trimetazidine be in prevention of myocardial and renal
revascularization injury?
Address for correspondence:
Yury Lopatin, Professor and Head Periprocedural myocardial injury during percutaneous coronary
of the Cardiology Department, intervention
Volgograd Regional Cardiology
Centre, 106, Universitetsky prospekt,
In about one-third of patients undergoing coronary revascularization by PCI, the pro-
Volgograd, Russia cedure itself causes myocardial injury (termed periprocedural myocardial injury,
(email: yu.lopatin@gmail.com) PMI),5,6 which has been associated with an increased rate of major adverse cardiac
www.medicographia.com events, including death.7,8

288 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Trimetazidine and prevention of myocardial and renal revascularization injury Lopatin
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

PMI during PCI is often clinically silent; however, it can be de- strategies to prevent distal embolization and microvascular
tected if the level of serum cardiac enzymes increases above coagulation, and strategies of protecting the myocardium it-
the 99th percentile upper reference limit (ULR). The Joint Eu- self against PMI (cardioprotection).
ropean Society of Cardiology (ESC)/American College of Car-
diology Foundation (ACCF)/American Heart Association (AHA)/ Regarding prevention of side branch occlusion, the current
World Heart Federation (WHF) Task Force Universal Defini- ESC/European Association for Cardiothoracic Surgery (EACTS)
tion of Myocardial Infarction 20129 defined PMI during PCI Guidelines on myocardial revascularization10 favor stent im-
as an elevation of cardiac troponin (cTn) level above the 99th plantation in the main vessel only, followed by provisional bal-
percentile URL after PCI, assuming a normal baseline troponin loon angioplasty with or without stenting of the side branch
value. This document also noted that in patients undergo- rather than routine stenting of both vessels. Several stents,
ing PCI with normal (99th percentile URL) baseline cTn con- designed specifically for treatment of bifurcation lesions, have
centrations, elevations of cTn greater than 5 x 99th percentile undergone extensive evaluation.
URL occurring within 48 hours of the procedure plus ischemic,
angiographic, or imaging findings are already defined as PCI- Strategies to prevent distal embolization and microvascular
related myocardial infarction (MI) (type 4a). The document coagulation include administration of antiplatelet and anti-
also clarifies that when a cTn value is less than or equal to thrombotic agents, use of distal protection devices, or direct
5 x 99th percentile URL after PCI and if the cTn value was stenting of the coronary lesion without predilatation.6
normal before PCIor when the cTn value is greater than 5 x
99th percentile URL in the absence of ischemic, angiograph- Strategies for protecting the myocardium against PMI are
ic, or imaging findingsthe term myocardial injury should based on pharmacological and nonpharmacological interven-
be used. tions. Among these interventions, the most discussed ones
are a high dose of statins,11,12 intracoronary b-blocker or ade-
Herrmann5 in his review classified the key factors that might nosine administration,13-17 trimetazidine,18 cyclosporine A,19,20
determine the incidence and magnitude of PMI into three and remote ischemic preconditioning.21,22 However, the abil-
groups: patient-related, lesion-related, and procedure-relat- ity of some of these interventions to provide effective car-
ed. The most frequently reported among these are older age; dioprotection has not been confirmed in all RCTs.
multivessel diffuse coronary artery disease (CAD); pre-exist-
ing renal impairment; presence of anemia; plaque burden; Trimetazidine for the prevention of periprocedural
number of lesions; presence of bifurcation lesions; tortuosity myocardial injury in patients undergoing coronary
of coronary arteries; suboptimal stenting; and multiple stents. intervention
Of course, the assessment of these factors before the inter- The first clinical trial on this issue was the open-label, random-
vention allows risk stratification for PMI. The most common ized, controlled, two parallel groups study trial performed by
mechanisms of myocardial injury during PCI are distal em- Polonski et al,23 in which 22 patients with one-vessel CAD re-
bolization and side branch occlusion; others are dissection, ceived oral trimetazidine, 60 mg daily, at least 4 days before
thrombus, no reflow/slow flow, or coronary perforation.6 percutaneous transluminal coronary angioplasty. It was found
that, compared with the control group (22 patients) pretreat-
Several strategies to protect from PMI during PCI have been ment with trimetazidine reduced not only angina, rhythm distur-
applied in clinical practice. Babu et al6 have divided them into bances, and ischemic ST-T changes on the electrocardiogram
three subgroups: strategies to prevent side branch occlusion, during the procedure, but also demonstrated a nonsignificant
trend to lower levels of cardiac troponin I (cTnI) 6 and 12 hours
after the procedure.
SELECTED ABBREVIATIONS AND ACRONYMS

CAD coronary artery disease Later, two independent groups from France and Greece re-
CIN contrast-induced nephropathy ceived more clear evidence of the ability of trimetazidine to
CKD chronic kidney disease prevent periprocedural myocardial injury in patients undergo-
CK-MB creatine kinase-MB ing coronary interventions. The single-center, prospective,
CM contrast media randomized evaluation study of Bonello et al24 included 206
cTnI cardiac troponin I stable angina patients with one-vessel CAD. Patients who un-
MI myocardial infarction derwent more than one inflation procedure during PCI were
PCI percutaneous coronary intervention excluded from the study. Half of the patients received an acute
PMI periprocedural myocardial injury loading dose of trimetazidine (60 mg) starting 30 min before
RCT randomized clinical trial recanalization, after which the operator was allowed to pro-
SCr serum creatinine ceed with angioplasty. The main outcome of this study was
ULR upper reference limit the frequency and the increase in the level of cTnI after a suc-
cessful PCI. cTnI levels were measured before and 6, 12, 18,

Trimetazidine and prevention of myocardial and renal revascularization injury Lopatin MEDICOGRAPHIA, Vol 38, No. 3, 2016 289
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

and 24 hours after PCI. It was found that there were no sta- tients (44%) in the placebo group. Forty-eight hours after the
tistically significant differences in the frequency of cTnI levels procedure, cTnI levels remained elevated in 15% of patients
between the trimetazidine group and the control group. How- receiving trimetazidine and in 32% of patients in the place-
ever, postprocedural cTnI levels were significantly reduced bo group. Twenty-four hours after PCI, CK-MB levels were
in the trimetazidine group at all time points (mean [standard above 5 ng/mL in 22% of patients in the trimetazidine group
deviation] for control vs trimetazidine group, respectively: at and 40% of patients in the placebo group. The authors con-
6 hours, 4.2 ng/mL [0.8] vs 1.7 ng/mL [0.2], P<0.001; at 12 cluded that trimetazidine can reduce myocardial reperfusion
hours, 5.5 ng/mL [1.5] vs 2.3 ng/mL [0.4], P<0.001; at 18 injury during PCI. They also stressed the need for further stud-
hours, 9 ng/mL [2.3] vs 3 ng/mL [0.5], P<0.001; and at 24 ies with inclusion of more patients.
hours, 3.2 ng/mL [1.2] vs 1 ng/mL [0.5], P<0.001). Moreover,
the total amount of cTnI released after PCI, as assessed by Xu et al26 in a single-center, prospective, randomized, con-
the area under the curve of serial measurements, was signifi- trolled study again demonstrated that trimetazidine reduced
cantly reduced in the trimetazidine group (P<0.05) (Figure 1). post-PCI cTnI release in patients with unstable angina pec-
toris. A total of 106 patients who underwent successful elec-
tive PCI and drug-eluting stent implantation were random-
ized to a trimetazidine group (n=51, 60 mg trimetazidine oral
loading dose 0.5-1.0 hour before PCI followed by 20 mg three
times daily after PCI on top of standard therapy) or a control
group (standard therapy without trimetazidine, n=55). cTnI
level was measured before and 16-18 hours after PCI. It was
found that cTnI levels after PCI were higher than before the
procedure in both groups of patients (P<0.01). However, post-
procedural cTnI levels increased from 0.02 mg/L (95% CI,
0.01-0.03)] at baseline to 0.11 mg/L (95% CI, 0.07-0.13)]
(P<0.05) at 16-18 hours in the trimetazidine group, whereas
in the control group, it increased from 0.02 mg/L (95% CI,
0.01-0.03)] to 1.31 mg/L (95% CI, 0.44-2.31)] (P<0.05). The
proportion of patients in the trimetazidine group who showed
a postprocedural cTnI level elevation of greater than 0.10 mg/L
was lower than that in the control group (P<0.01).

Figure 1. Time course of cardiac troponin I release. Recently Zhang et al18 have published a meta-analysis that
Mean circulating cardiac troponin I concentrations (error bars showing standard covered data from 9 RCTs with a total of 778 patients hav-
deviation) are indicated for control (open symbols; n=130) and trimetazidine
(closed symbols; n=136) groups. The arrow indicates the time of PCI. ***P<0.001. ing undergone PCI. It was shown that additional use of trimeta-
Abbreviations: cTnI, cardicac troponin I; PCI, percutaneous coronary intervention. zidine in the periprocedural period of PCI significantly im-
From reference 24: Bonello et al. Heart. 2007;93:703-707. 2007, BMJ Pub-
lishing Group Ltd.
proved left ventricular ejection fraction, reduced elevated cTnI
level (relative risk [RR], 0.69; 95% confidence interval [CI], 0.48-
In the other trial, Labrou et al25 included 52 patients hospital- 0.99), angina attacks during PCI (odds ratio [OR], 0.16; 95%
ized for acute coronary syndromes. Coronary angiography CI, 0.07-0.38), and ischemic ST-T changes on the electro-
was performed in all patients, and more specifically, after 6 cardiogram during PCI (RR, 0.76; 95% CI, 0.59-0.98).
days of hospitalization in patients with MI. Patients who had
undergone primary PCI were excluded from this study. All pa- It should be noted that this meta-analysis is in line with anoth-
tients received bare metal stents; drug-eluting stents were er meta-analysis27 that also showed a cardioprotective effect
not used in the study. In addition to conventional antianginal of trimetazidine in patients that underwent coronary artery by-
therapy, 27 patients received 20 mg oral trimetazidine every 8 pass graft surgery. The authors of both meta-analyses have
hours, starting 15 days before PCI and continuing for 3 months noted the superiority of trimetazidine over conventional ther-
after the procedure. The other 25 patients were included in apy during revascularization procedures. However, the au-
the placebo group. For each patient, serum cTnI and crea- thors also emphasize that new clinical trials with large sam-
tine kinase-MB (CK-MB) levels were measured before PCI, ples and rigorous designs are needed.
then at 6, 24, and 48 hours after the procedure. Serum cTnI
and CK-MB measurements were considered negative for my- Several mechanisms are responsible for the prevention of is-
ocardial damage when levels were lower than 0.2 ng/mL and chemic reperfusion injury; one of these is the ability of trimeta-
lower than 5 ng/mL, respectively. It was observed that 24 hours zidine to inhibit the opening of mitochondrial permeability
after PCI, cTnI levels were higher than 1 ng/mL in 7 of 27 pa- transition pores, a crucial event in cardiomyocyte death after
tients (26%) in the trimetazidine group and in 11 of 25 pa- myocardial ischemia-reperfusion.28

290 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Trimetazidine and prevention of myocardial and renal revascularization injury Lopatin
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

Contrast-induced nephropathy following ple CM exposures within the past 72 hours.38 There is no
percutaneous coronary intervention specific treatment for CIN after PCIprevention remains the
Contrast-induced nephropathy (CIN) or contrast-induced acute most effective strategy. The first step in the prevention of CIN
kidney injury is a common but underdiagnosed complication is the identification of patients at high risk. The most com-
of coronary diagnostic and interventional procedures that is monly used scoring system is the Mehran score.39 Preven-
associated with increased in-hospital morbidity and mortal- tive strategies for CIN include the limitation of CM volume;
ity, prolonged hospital stay, and raised health care costs.29-33 use of preheated (37C) iso-osmolar CM; pre-PCI hydration
CIN is the third most common cause of hospital-acquired with normal saline; use of N-acetylcysteine, sodium bicarbon-
acute renal failure.34 ate, and statins; and stopping nephrotoxic drugs 48 hours
before and after CM exposure.40-42 The search continues for
It is known that the administration of contrast media (CM) rap- new pharmacologic and nonpharmacologic interventions for
idly induces intense renal vasoconstriction and subsequent the prevention of CIN in patients undergoing coronary diag-
reduced blood perfusion. This can lead to ischemic and hy- nostic and interventional procedures.
poxic damage of the renal medulla and the production of oxy-
gen free radicals, inducing tubular epithelial damage.35 Ad- Trimetazidine for the prevention of
ditional factors such as hypotension, microembolization of contrast-induced nephropathy in patients
atheromatous debris, or bleeding complications can also be undergoing coronary interventions
responsible for the development of CIN.36 Onbasili et al43 were the first to clinically evaluate the efficacy
of trimetazidine in the prevention of CIN in patients with high
In spite of the growing importance of this complication, there SCr levels undergoing coronary angiography or PCI. A total of
is a lack of consensus on how to define CIN. According to the 82 patients with basal SCr levels between 1.2 and 2.5 mg/dL
most recognized definition, CIN is an absolute (0.5 mg/dL; were enrolled in a prospective double-blind, randomized, con-
44 mmol/L) or relative (25%) increase in baseline serum trolled trial. Indications of the coronary interventions were acute
creatinine (SCr) levels 48-72 hours after an exposure to io- coronary syndrome, stable angina, dilated cardiomyopathy,
dinated CM. and preoperative assessment. Of all patients, 19 had diabetes
mellitus (all of them type 2). In this study, patients were ran-
Generally, the incidence of CIN in individuals with normal re- domized into a trimetazidine group (20 mg three times daily,
nal function who undergo PCI is low (<3%).37 However, it rises orally, for 72 hours starting 48 hours before the procedure)
remarkably in patients with chronic kidney disease (CKD) (up or a control group. The standard parenteral hydration pro-
to 40% and even more).29,37 Besides pre-existing CKD, other tocol was applied to patients in both groups. SCr levels were
predisposing factors for CIN are diabetes, congestive heart measured before the procedure, 48 hours, and 7 days after
failure, hypotension, hypertension, preprocedure shock, re- the procedure. An increase in SCr level exceeding 0.5 mg/day
cent MI, anemia, female sex, advanced age, and concomitant or one-quarter of the baseline value was considered as CIN.
use of nephrotoxic agents.29,38 Procedure-related risk factors It was found that SCr levels in the control group increased sig-
for the development of CIN include high volume of CM, as nificantly 2 days after the procedure (P<0.05) and returned to
well as its high osmolarity, intra-arterial injection, and multi- the baseline values on the seventh day (Figure 2). On the oth-
er hand, they did not change significantly on the second day,
and they even significantly decreased on the seventh day in
the trimetazidine group (P<0.05). CIN developed in 2.5% (1/
40) of patients in the trimetazidine group and in 16.6% (7/42)
of patients in the control group (P<0.05).

Later, Rahman et al44 in a large prospective randomized,


controlled trial again confirmed the ability of trimetazidine to
prevent CIN after coronary angiogram or PCI. A total of 400
patients were enrolled in this study; in contrast to the study
performed by Onbasili et al,43 patients with diabetes were ex-
cluded. Of the 400 enrolled, 200 patients were treated with
trimetazidine plus hydration with normal saline and 200 pa-
tients (control group) were given hydration with normal saline
only. The dose of trimetazidine was 35 mg twice daily for 96
Figure 2. Serum creatinine levels in control group and trimetazidine hours starting 48 hours before the procedure. It was found
group.
that the incidence of CIN was significantly reduced by admin-
Abbreviation: TMZ, trimetazidine.
Adapted from reference 43: Onbasili et al. Heart. 2007;93:698-702. 2007, istration of trimetazidine with saline in comparison with saline
BMJ Publishing Group Ltd. alone (4% vs 14%; P<0.05).

Trimetazidine and prevention of myocardial and renal revascularization injury Lopatin MEDICOGRAPHIA, Vol 38, No. 3, 2016 291
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

While the two above-mentioned trials43,44 enrolled nondiabetic whereas it occurred in 14 patients (28%) of the control group
or a mixture of diabetic and nondiabetic patients with CKD, (P<0.05). Thus, the study performed by Shehata45 was the
the study performed by Shehata45 evaluated the effect of peri- first one to evaluate both the anti-CIN and the anti-PMI ef-
procedural administration of trimetazidine on the incidence fects of trimetazidine in diabetic patients with CKD undergo-
of PCI-induced myocardial injury and CIN in high-risk pa- ing elective PCI.
tients with diabetes and mild-to-moderate renal dysfunction.
The primary end point of the study was the development of Last year Nadkarni et al46 published a meta-analysis that pooled
CIN 72 hours after PCI. A total of 100 consecutive diabetic data from the three above-mentioned RCTs, which altogeth-
patients with chronic stable angina and mild-to-moderate CKD er included 582 patients with CKD and SCr levels ranging
were randomized into a trimetazidine group (35 mg of agent from 1.26 to 2 mg/dL. It was shown that in patients under-
twice daily for 72 hours starting 48 hours before the proce- going coronary angiography, administration of trimetazidine in
dure) and a control group (without trimetazidine). The stan- conjunction with normal saline and/or oral N-acetylcysteine
dard parenteral hydration protocol was applied to all included was associated with a significant reduction in the incidence of
patients. Additionally, N-acetylcysteine (1200 mg) was given CIN by 11% (risk difference 0.11; 95% CI, 0.16-0.06; P<0.01)
to patients in both groups 24 hours before and after the pro- when compared with the control group. The number need-
cedure. There was no statistically significant difference be- ed to treat to prevent 1 episode of CIN was 9. The authors
tween the two groups in terms of the preliminary angiographic concluded that trimetazidine could be considered a poten-
findings and procedural characteristics. However, postpro- tial tool for prevention of CIN in patients with renal dysfunc-
cedural mean cTnI level was significantly higher in the con- tion. On the other hand, Nadkarni et al46 emphasized that
trol group than in the trimetazidine group (6 hours: 80.3 vs considering the small sample size of these studies and the
160.2 pg/mL, P<0.001; 12 hours: 130.9 vs 240.8 pg/mL, level of evidence being 1C, decision making about the use of
P<0.001; and 24 hours: 70.7 vs 140.3 pg/mL, P<0.001). trimetazidine should be individualized to each patient and
The SCr level in the control group significantly increased 3 each clinical context.
days after PCI and decreased on the tenth day. On the other
hand, no significant change was observed in the trimetazidine Recently Liu et al47 have confirmed once again the renopro-
group. Mean cTnI levels as well as mean SCr levels in both tective effect of trimetazidine on CIN in patients with mild-to-
study groups are graphically presented in Figures 3 and 4. moderate renal dysfunction who undergo coronary angiog-
CIN was noted in 6 patients (12%) of the trimetazidine group, raphy or PCI. In this single-center prospective, randomized

Figure 3. Graphic presentation showing changes in mean cardiac Figure 4. Graphic presentation showing changes in mean serum
troponin I levels in control group and trimetazidine group. creatinine levels in control group and trimetazidine group.
Abbreviation: PCI, percutaneous coronary intervention. Abbreviation: PCI, percutaneous coronary intervention.
Adapted from reference 45: Shehata. Am J Cardiol. 2014;114:389-394. 2014, Adapted from reference 45: Shehata. Am J Cardiol. 2014;114:389-394. 2014,
Elsevier Inc. All rights reserved. Elsevier Inc. All rights reserved.

292 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Trimetazidine and prevention of myocardial and renal revascularization injury Lopatin
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

controlled clinical trial, 132 patients with re-


nal dysfunction undergoing coronary an-
giography or PCI (more than half of all pa-
tients) were divided into a control group
(n=70) and a trimetazidine group (n=62). Tri-
metazidine was administered orally 48 hours
before and 24 hours after the procedure.
Standard hydration was used in all included
patients.

Postoperative SCr concentration was signif-


icantly lower in the trimetazidine group than
in the control group at 48 hours but not at
24 hours (P=0.026 and P=0.056, respec-
tively), whereas postoperative cystatin C lev-
el was significantly lower in the trimetazidine
group both at 24 and 48 hours than in the
control group (P=0.000 and P=0.025, re-
spectively). Trimetazidine significantly re-
duced the incidence of CIN (8% vs 20% in Figure 5. Kaplan-Meier survival curve of the timing of adverse events during the follow-
control; P=0.034). Moreover, the incidence up period.
of adverse events within 12 months of fol- Abbreviation: TMZ, trimetazidine.
low-up was significantly lower in the trimeta- Adapted from reference 47: Liu et al. Am J Med Sci. 2015;350:398-402. Copyright 2015 Southern
Society for Clinical Investigation. Published by Elsevier Inc. All rights reserved.
zidine group than in the control group (9.6%
vs 22.8%; P=0.043). The Kaplan-Meier survival curves of ad- It is important to mention that the potent antioxidant effect of
verse events showed that the incidence of adverse events trimetazidine has been demonstrated not only in myocardial
significantly decreased in the trimetazidine group compared and renal, but also hepatic, pulmonary, intestinal, and testic-
with the control group (log rank P=0.035) (Figure 5). A sig- ular ischemia-reperfusion injuries.49-53 This allows us to con-
nificant correlation between the adverse events and the in- sider trimetazidine a promising agent that can prevent develop-
cidence of CIN was also noted. ment of ischemia-reperfusion injuries during revascularization
procedures. However, several issues are still to be clarified.
Akgll et al48 explain the renoprotective properties of trimeta- First of all, it is necessary to identify the optimal duration of the
zidine against CIN by its antioxidant properties. In an exper- pre- and post-procedural administration of trimetazidine. The
imental study, the authors showed significantly higher levels question regarding the optimal dose of trimetazidine also re-
of malondialdehyde and lower levels of superoxide dismu- mains open. Moreover, new studies are needed to evaluate not
tase activity in a CM alone group than in a CM plus trimetazi- only the efficacy but also the safety of trimetazidine in differ-
dine group. Moreover, histopathological analysis demonstrated ent patient populations having undergone revascularization
a significant expansion of Bowmans capsule, tubule epithe- procedures. For example, this agent is contraindicated when
lium degeneration, tubule epithelium necrosis, and intersti- creatinine clearance is below 30 mL/min. The dose of trimeta-
tial infiltration in the CM group compared with the CM plus zidine that will be most effective in the context of prevention of
trimetazidine group. Interestingly, none of the histopatholog- ischemia-reperfusion injury in patients with creatinine clear-
ical scores differed significantly between the CM plus trimetazi- ance of 30-60 mL/min is still to be defined as well. Large-
dine group and the control group (without CM). scale studies are required to answer these questions. n

References
1. Lopatin YM, Rosano GM, Fragasso G, et al. Rationale and benefits of trimetazi- 4. Chen J, Zhou S, Jin J, et al. Chronic treatment with trimetazidine after dis-
dine by acting on cardiac metabolism in heart failure. Int J Cardiol. 2016;203: charge reduces the incidence of restenosis in patients who received coronary
909-915. stent implantation: a 1-year prospective follow-up study. Int J Cardiol. 2014;174:
2. Lopatin YM, Dronova EP. Clinical and pharmacoeconomic evaluation of long- 634-639.
term use of trimetazidine modified release in patients with coronary artery dis- 5. Herrmann J. Peri-procedural myocardial injury: 2005 update. Eur Heart J. 2005;
ease undergoing percutaneous coronary intervention. Heart (Rus). 2011;2: 26:2493-2519.
67-72. 6. Babu GG, Walker JM, Yellon DM, et al. Peri-procedural myocardial injury dur-
3. Xu X, Zhang W, Zhou Y, et al. Effect of trimetazidine on recurrent angina pec- ing percutaneous coronary intervention: an important target for cardioprotec-
toris and left ventricular structure in elderly multivessel coronary heart disease tion. Eur Heart J. 2011;32:23-32.
patients with diabetes mellitus after drug-eluting stent implantation: a single- 7. Ioannidis JP, Karvouni E, Katritsis DG. Mortality risk conferred by small eleva-
centre, prospective, randomized double-blind study at 2-year follow-up. Clin tions of creatine kinase-MB isoenzyme after percutaneous coronary interven-
Drug Investig. 2014;34:251-258. tion. J Am Coll Cardiol. 2003;42:1406-1411.

Trimetazidine and prevention of myocardial and renal revascularization injury Lopatin MEDICOGRAPHIA, Vol 38, No. 3, 2016 293
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

8. Testa L, Van Gaal WJ, Biondi Zoccai GG, et al. Myocardial infarction after per- come after contemporary percutaneous coronary intervention. Am Heart J.
cutaneous coronary intervention: a meta-analysis of troponin elevation apply- 2006;151:146-152.
ing the new universal definition. QJM. 2009;102:369-378. 31. Solomon RJ, Mehran R, Natarajan MK, et al. Contrast-induced nephropathy
9. Thygesen K, Alpert JS, Jaffe AS, et al. Third universal definition of myocardial and long-term adverse events: cause and effect? Clin J Am Soc Nephrol. 2009;
infarction. Eur Heart J. 2012;33:2551-2567. 4:1162-1169.
10. Windecker S, Kolh P, Alfonso F, et al. 2014 ESC/EACTS guidelines on myo- 32. Weisbord SD, Palevsky PM. Contrast-induced acute kidney injury: short and
cardial revascularization. Eur Heart J. 2014;35:2541-2619. long-term implications. Semin Nephrol. 2011;31:300-309
11. Merla R, Reddy NK, Wang FW, et al. Meta-analysis of published reports on the 33. Subramanian S, Tumlin J, Bapat B. Economic burden of contrast-induced
effect of statin treatment before percutaneous coronary intervention on peri- nephropathy: implications for prevention strategies. J Med Econ. 2007;10:119-
procedural myonecrosis. Am J Cardiol. 2007;100:770-776. 134.
12. Pan Y, Tan Y, Li B, Li X. Efficacy of high-dose rosuvastatin preloading in pa- 34. Nash K, Hafeez A, Hou S. Hospital-acquired renal insufficiency. Am J Kidney
tients undergoing percutaneous coronary intervention: a meta-analysis of four- Dis. 2002;39:930-936.
teen randomized controlled trials. Lipids Health Dis. 2015;14:97. 35. Russo D, Minutolo R, Cianciaruso B, et al. Early effects of contrast media on
13. Uretsky BF, Birnbaum Y, Osman A, et al. Distal myocardial protection with in- renal hemodynamics and tubular function in chronic renal failure. J Am Soc
tracoronary beta blocker when added to a GpIIb/IIIa platelet receptor block- Nephrol. 1995;6:1451-1458.
er during percutaneous coronary intervention improves clinical outcome. Ca- 36. Gupta R, Gurm HS, Bhatt DL, et al. Renal failure after percutaneous coronary
theter Cardiovasc Interv. 2008;72:488-497. intervention is associated with high mortality. Catheter Cardiovasc Interv. 2005;
14. Park H, Otani H, Noda T, et al. Intracoronary followed by intravenous admin- 64:442-448
istration of the short-acting b-blocker landiolol prevents myocardial injury in 37. Rihal CS, Textor SC, Grill DE, et al. Incidence and prognostic importance of
the face of elective percutaneous coronary intervention. Int J Cardiol. 2013; acute renal failure after percutaneous coronary intervention. Circulation. 2002;
167:1547-1551. 105:2259-2264.
15. Er F, Dahlem KM, Nia AM, et al. Randomized control of sympathetic drive with 38. Nicola R, Shaqdan KW, Aran K, et al. Contrast-induced nephropathy: identi-
continuous intravenous esmolol in patients with acute ST-Segment elevation fying the risks, choosing the right agent, and reviewing effective prevention
myocardial infarction: the BEtA-blocker Therapy in Acute Myocardial Infarc- and management methods. Curr Probl Diagn Radiol. 2015;44:501-504.
tion (BEAT-AMI) trial. JACC Cardiovasc Interv. 2016;9:231-240. 39. Mehran R, Aymong ED, Nikolsky E, et al. A simple risk score for prediction of
16. Lee CH, Low A, Tai BC, et al. Pretreatment with intracoronary adenosine re- contrast-induced nephropathy after percutaneous coronary intervention: devel-
duces the incidence of myonecrosis after non urgent percutaneous coronary opment and initial validation. J Am Coll Cardiol. 2004;44:1393-1399.
intervention: a prospective randomized study. Eur Heart J. 2007;28:19-25. 40. Owen RJ, Hiremath S, Myers A, et al. Canadian Association of Radiologists
17. De Luca G, Iorio S, Venegoni L, et al. Evaluation of intracoronary adenosine to consensus guidelines for the prevention of contrast-induced nephropathy: up-
prevent periprocedural myonecrosis in elective percutaneous coronary inter- date 2012. Can Assoc Radiol J. 2014;65:96-105.
vention (from the PREVENT-ICARUS Trial). Am J Cardiol. 2012;109:202-207. 41. Wichmann JL, Katzberg RW, Litwin ShE, et al. Contrast-induced nephropathy.
18. Zhang Y, Ma XJ, Shi DZ. Effect of trimetazidine in patients undergoing percuta- Circulation. 2015;132:1931-1936.
neous coronary intervention: a meta-analysis. PLoS One. 2015;10(9):e0137775. 42. Pavlidis AN, Jones DA, Sirker A, et al. Prevention of contrast-induced acute kid-
19. Piot C, Croisille P, Staat P, et al. Effect of cyclosporine on reperfusion injury in ney injury after percutaneous coronary intervention for chronic total coronary
acute myocardial infarction. N Engl J Med. 2008;359:473-481. occlusions. Am J Cardiol. 2015;115:844-851.
20. Ottani F, Latini R, Staszewsky L, et al. for the CYCLE Investigators. Cyclosporine 43. Onbasili AO, Yeniceriglu Y, Agaoglu P, et al. Trimetazidine in the prevention of
A in Reperfused Myocardial Infarction: the multicenter, controlled, open-label contrast-induced nephropathy after coronary procedures. Heart. 2007;93:698-
CYCLE Trial. J Am Coll Cardiol. 2016;67:365-374. 702.
21. Hoole SP, Heck PM, Sharples L, et al. Cardiac Remote Ischemic Precondition- 44. Rahman MM, Haque SS, Rokeya B et al. Trimetazidine in the prevention of con-
ing in Coronary Stenting (CRISP Stent) Study: a prospective, randomized con- trast induced nephropathy after coronary angiogram. Mymensingh Med J. 2012;
trol trial. Circulation. 2009;119:820-827. 21:292-299.
22. Zografos TA, Katritsis GD, Katritsis DG. Remote ischemic preconditioning re- 45. Shehata M. Impact of trimetazidine on incidence of myocardial injury and con-
duces peri-procedural myocardial injury in elective percutaneous coronary in- trast-induced nephropathy in diabetic patients with renal dysfunction under-
tervention: a meta-analysis. Int J Cardiol. 2014;173:530-532. going elective percutaneous coronary intervention. Am J Cardiol. 2014;114:
23. Polonski L, Dec I, Wojnar R, et al. Trimetazidine limits the effects of myocar- 389-394.
dial ischaemia during percutaneous coronary angioplasty. Curr Med Res Opin. 46. Nadkarni GN, Konstantinidis I, Patel A, et al. Trimetazidine decreases risk of con-
2002;18:389-396. trast-induced nephropathy in patients with chronic kidney disease: a meta-analy-
24. Bonello L, Sbragia P, Amabile N, et al. Protective effect of an acute oral load- sis of randomized controlled trials. J Cardiovasc Pharmacol Ther. 2015;20:539-
ing dose of trimetazidine on myocardial injury following percutaneous coro- 546.
nary intervention. Heart. 2007;93:703-707. 47. Liu W, Ming Q, Shen J, et al. Trimetazidine prevention of contrast-induced
25. Labrou A, Giannoglou G, Zioutas D, et al. Trimetazidine administration mini- nephropathy in coronary angiography. Am J Med Sci. 2015;350:398-402.
mizes myocardial damage and improves left ventricular function after percu- 48. Akgll C, Saruhan T, Eryilmaz U, et al. The first histopathological evidence
taneous coronary intervention. Am J Cardiovasc Drug. 2007;7:143-150. of trimetazidine for the prevention of contrast-induced nephropathy. Ren Fail.
26. Xu XH, Zhang WJ, Zhou YJ, et al. Effects of trimetazidine therapy on left ven- 2014;36:575-580.
tricular function after percutaneous coronary intervention. Zhonghua Xin Xue 49. Iskesen I, Saribulbul O, Cerrahoglu M, et al. Trimetazidine reduces oxidative
Guan Bing Za Zhi. 2013;41:205-209. stress in cardiac surgery. Circ J. 2006;70:1169-1173.
27. Zhang N, Lei JY, Liu Q, et al. The effectiveness of preoperative trimetazidine on 50. Singh D, Chopra K. Effect of trimetazidine on renal ischemia/reperfusion injury
myocardial preservation in coronary artery bypass graft patients: a systematic in rats. Pharmacol Res. 2004;50:623-629.
review and meta-analysis. Cardiology. 2015;131:86-96. 51. Inci I, Dutly A, Rousson V, et al. Trimetazidine protects the energy status after
28. Argaud L, Gomez L, Gateau-Roesch O, et al. Trimetazidine inhibits mitochon- ischemia and reduces reperfusion injury in a rat single lung transplant model.
drial permeability transition pore opening and prevents lethal ischemia-reper- J Thorac Cardiovasc Surg. 2001;122:1155-1161.
fusion injury. J Mol Cell Cardiol. 2005;39:893-899. 52. Tetik C, Ozden A, Calli N, et al. Cytoprotective effect of trimetazidine on 60
29. Marenzi G, Lauri G, Assanelli E, et al. Contrast-induced nephropathy in patients minutes of intestinal ischemiareperfusion injury in rats. Transpl Int. 1999;12:
undergoing primary angioplasty for acute myocardial infarction. J Am Coll 108-112.
Cardiol. 2004;44:1780-1785. 53. Unal D, Karatas OF, Savas M, et al. Protective effects of trimetazidine on testic-
30. Blackman DJ, Pinto R, Ross JR, et al. Impact of renal insufficiency on out- ular ischemia-reperfusion injury in rats. Urol Int. 2007;78:356-362.

Keywords: contrast-induced nephropathy; percutaneous coronary intervention; periprocedural myocardial injury;


trimetazidine

294 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Trimetazidine and prevention of myocardial and renal revascularization injury Lopatin
M E TA B O L I C AGENTS IN THE
C O N T E M P O R A R Y T R E AT M E N T O F A N G I N A

COMMENT LA TRIMTAZIDINE PEUT-ELLE TRE EFFICACE


DANS LA PRVENTION DES LSIONS DE REVASCULARISATION RNALE ET MYOCARDIQUE ?
Lintervention coronaire percutane est actuellement considre comme lun des traitements cls dans la prise en
charge de la maladie coronaire occlusive. Les nouvelles techniques dintervention coronaire percutane lont rendue
sre, avec un taux minimal de complications, mais la revascularisation myocardique per se provoque encore des l-
sions rnales ou myocardiques. Rcemment, ces complications ont fait lobjet dune attention particulire, notam-
ment les lsions myocardiques priprocdurales et la nphropathie aux produits de contraste. Ces complications
peuvent arriver frquemment et sont associes un pronostic dfavorable. Cest pourquoi la recherche de strat-
gies visant prvenir le dveloppement de ces lsions priprocdurales est trs importante. Les rsultats des tudes
cliniques et exprimentales laissent entrevoir des rsultats prometteurs pour la trimtazidine dans la prvention des
lsions rnales et myocardiques priprocdurales.

Trimetazidine and prevention of myocardial and renal revascularization injury Lopatin MEDICOGRAPHIA, Vol 38, No. 3, 2016 295
THE QUESTION CONTROVERSIAL QUESTION

oronary revascularization

C procedures are commonly


performed for the sympto-
matic treatment of patients with
myocardial ischemia. Neverthe-
less, current data show that even
Is targeting only stenosis
after successful revascularization
a third of angina patients still suf-
fer from pain. Angina that recurs or
sufficient to optimally
persists after coronary revascular-
ization represents a significant clin-
ical problem and its management
improve angina?
is particularly challenging. There-
fore, this begs the question: are we
doing enough by targeting steno-
sis only?

1. P. S. Farsky, Brazil

2. L. O. Go, Philippines

3. H. Hasan-Ali, Egypt

4. H. Q. T. Ho, Vietnam

5. D. Isaza-Restrepo, Colombia

6. T. Kovarnik, Czech Republic

7. O. H. Masoli, Argentina

8. A. N. Parkhomenko and
O. S. Gurjeva, Ukraine

9. C. K. Ponde, India

10 . V. Sansoy, Turkey

11. D. Vassilev, Bulgaria

Is targeting only stenosis sufficient to optimally improve angina? MEDICOGRAPHIA, Vol 38, No. 3, 2016 297
CONTROVERSIAL QUESTION

1. P. S. Farsky, Brazil
PCI in patients with stable angina, showed that native coro-
nary disease progression exceeded failed revascularization
as the cause of angina after five years.3 Disease progression
Pedro Silvio FARSKY, MD
Doutor em Cincias pela Fac Medicina USP occurred in native untreated arteries in two-thirds of cases.
Fellow da European Society of Cardiology In this study, the myocardial jeopardy score fell following ini-
Instituto Dante Pazzanese de Cardiologia tial revascularization, from 60% to 17% for PCI-treated pa-
Hospital Israelita Albert Einstein
So Paulo, BRAZIL tients compared with a reduction from 60% to 7% for CABG
(email: farskyp@uol.com.br) surgery patients ( P<0.001), but rebounded after five years
to 25% for PCI and 20% for surgery patients ( P=0.01). Myo-

M
yocardial revascularization with percutaneous coro- cardial jeopardy increased between study entry and the five-
nary intervention (PCI) or coronary artery bypass year follow-up in 42% of PCI-treated patients and 51% of
graft surgery (CABG) is indicated when there is sig- CABG-treated patients ( P=0.06).
nificant obstruction of coronary blood flow associated with
myocardial ischemia, in order to relieve symptoms or prolong The MASS II trial (Medicine, Angioplasty, or Surgery Study)
survival. randomly assigned 611 patients with multivessel disease, pre-
served left ventricular systolic function, and stable angina to
Several mechanisms may explain the persistence of angina/ CABG, PCI, or optimal medical therapy.4 After only one year,
ischemia after a revascularization procedure, including graft or 12% of the patients in the CABG group, 21% in the PCI group,
PCI failure, incomplete revascularization, and disease pro- and 54% in the medical therapy group had angina (P<0.0001).
gression in native coronary arteries. Microvascular dysfunc- Furthermore, after 10 years, 64% of patients in the CABG
tion may play a prominent role in the unexpected prevalence group, 59% of patients in the PCI group, and 43% in the op-
of angina after the removal of obstructions in the major coro- timal medical therapy group ( P<0.001) were angina free.
nary branches.
Incomplete coronary revascularization
Graft failure and new atherosclerotic lesions In many patients with chronic stable angina, complete revas-
Angina may recur at any time in the first few months follow- cularization is not achieved at the time of PCI or CABG. Com-
ing apparently successful CABG surgery, and may present plete revascularization of all significantly obstructed coronary
as stable or unstable angina. In the early postoperative peri- segments is the goal of CABG, and recent data has shown
od, angina is usually caused by graft closure due to a tech- that complete revascularization following PCI has a positive
nical problem. One year after CABG, angina may occur as effect on long-term clinical outcomes. However, incomplete
a result of the gradual development of graft stenosis or of the coronary revascularization following CABG or PCI is associat-
progression of new atherosclerotic lesions, either in nonby- ed with increased mortality as well as with an increased inci-
passed vessels or distal to graft anastomosis. dence of myocardial infarction, repeat revascularization, and
major adverse cardiovascular or cerebrovascular events.5
After 10 years, the rate of saphenous vein graft closure is about
50%, and is associated with anatomical factors (eg, artery References
diameter), clinical factors (eg, male sex and aging), and risk 1. Goldman S, Copeland J, Moritz T, et al. Improvement in early saphenous vein
factors for atherosclerotic cardiovascular disease.1 Using the graft patency after coronary artery bypass surgery with antiplatelet therapy: results
of a Veterans Administration Cooperative Study. Circulation. 1988;77:1324-1332.
internal thoracic artery reduces angina recurrence and pro- 2. Fitzgibbon GM, Kafka HP, Leach AJ, Keon WJ, Hooper GD, Burton JR. Coronary
longs survival. bypass graft fate and patient outcome: angiographic follow-up of 5,065 grafts
related to survival and reoperation in 1,388 patients during 25 years. J Am Coll
Cardiol. 1996;28:616-626.
Late recurrent angina after CABG can also result from pro- 3. Alderman EL, Kip KE, Whitlow PL, et al. Native coronary disease progression
gressive atherosclerosis in a native vessel. Studies performed exceeds failed revascularization as cause of angina after five years in the Bypass
before the widespread use of arterial grafting found that saphe- Angioplasty Revascularization Investigation (BARI). J Am Coll Cardiol. 2004;44:
766-774.
nous vein graft (SVG) disease was responsible for 80% of new 4. Hueb W, Soares PR, Gersh BJ, et al. The medicine, angioplasty, or surgery study
angina symptoms, as opposed to new native artery disease, (MASS-II): a randomized, controlled clinical trial of three therapeutic strategies
which was responsible for 54% of the cases.2 Later, an analy- for multivessel coronary artery disease: one-year results. J Am Coll Cardiol. 2004;
43:1743-1751.
sis from the BARI trial (Bypass Angioplasty Revascularization 5. Kereiakes DJ. Reassessing the importance of complete versus incomplete coro-
Investigation), which investigated the use of CABG versus nary revascularization. Rev Cardiovasc Med. 2014;15:24-30.

298 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Is targeting only stenosis sufficient to optimally improve angina?
CONTROVERSIAL QUESTION

2. L. O. Go, Philippines
coronary intervention patients and 23.7% of coronary artery
bypass graft surgery patients.5 And in the most aggressively
managed IHD patient group, those with acute coronary syn-
Loewe O. GO, MD, FACC drome, residual angina at one year of follow-up is found in
H. B. Calleja Heart and Vascular Institute 37% of those patients who received early invasive therapy.6
ST. LUKES MEDICAL CENTER
Quezon City
PHILIPPINES Taken together, these observations suggest that coronary
(email: goloewe@yahoo.com.ph) stenosis or obstruction is not the only causative factor of
symptomatic IHD, and hence revascularization alone cannot

B
ack in the early 1990s, when I was a cardiology fellow relieve angina in all patients. Other factors such as vasospasm,
in Mount Sinai Hospital in New York, coronary angio- microvascular disease, endothelial dysfunction, thrombosis,
plasty was a very popular treatment for treating angina inflammation and even an excessive heart rate can also
in patients with coronary artery disease. However, the direc- provoke angina. And keep in mind that these factors are not
tor of the cardiac catheterization laboratory, Dr John Ambrose, mutually exclusive (ie, several of them can be operating si-
taught us something radical at the time: patients presenting multaneously in the same patient). Thus, guidelines for IHD
with stable and even unstable angina often have nonsignif- management emphasize global risk assessment and recom-
icant or no coronary stenosis during angiography. It turns out mend optimizing treatment by using renin-angiotensin sys-
that this was a very prescient observation. teminhibitors, statins, antithrombotic agents, b-blockers,
and/or other heart ratelowering agents. But whatever the
Two lines of evidence strongly suggest that targeting only etiology of angina, the end result is a metabolic derangement
coronary stenosis is NOT sufficient to optimally improve angi- in the cardiomyocytes leading to an imbalance between en-
na in patients with ischemic heart disease (IHD). First, angina ergy production and consumption. This imbalance produces
actually occurs in patients in the absence of coronary steno- increased lactic acid levels, which then stimulate sensory
sis. In one study of 1630 patients with typical angina, the ob- nerve fibers in the myocardium and manifest as angina. Cor-
served prevalence of angiographically confirmed >50% stenot- recting these derangements requires the use of unique agents,
ic coronary artery disease ranged from 38%-53% in men and such as trimetazidine, which shifts mitochondrial energy pro-
15%-29% in women over 50 years old1which implies that duction from fatty acid oxidation (more oxygen consuming) to
the majority of these patients do not have significant coronary glucose oxidation (more oxygen sparing) and thus can help
obstruction. And in patients with the most dramatic mani- to restore the balance between energy supply and demand,
festations of IHD, namely those with ST-elevation myocar- decrease lactic acidosis, and ultimately reduce angina.
dial infarction (2251 patients) or unstable angina (2406 pa-
References
tients), up to 7%-14% of men and 10%-30% of women have 1. Cheng VY, Berman DS, Rozanski A, et al. Performance of the Traditional Age,
normal coronary artery angiograms.2 Second, multiple stud- Sex, and Angina TypicalityBased Approach for Estimating Pretest Probability
ies show that even after successful revascularization a signif- of Angiographically Significant Coronary Artery Disease in Patients Undergo-
ing Coronary Computed Tomographic Angiography. Results From the Multina-
icant number of patients still suffer from angina. The landmark tional Coronary CT Angiography Evaluation for Clinical Outcomes: An Interna-
COURAGE trial (Clinical Outcomes Utilizing Revasculariza- tional Multicenter Registry (CONFIRM). Circulation. 2011;124:2423-2432.
tion and Aggressive druG Evaluation) showed that after per- 2. Hochman JS, Tamis, JE, Thompson TD, et al. Sex, clinical presentation, and out-
come in patients with acute coronary syndromes. N Engl J Med. 1999;341:226-
cutaneous coronary intervention on top of optimal medical 232.
therapy, many patients remain symptomatic41% at three 3. Weintraub WS, Spertus JA, Kolm P, et al; COURAGE Trial Research Group. Ef-
years and 26% at five years.3 A meta-analysis which includ- fect of PCI on Quality of Life in Patients with Stable Coronary Disease. N Engl
J Med. 2008;359:677-687.
ed studies published from 1992-2007 (RITA-2 [Randomized 4. Wijeysundera HC, Nallamothu BK, Krumholz HM, Tu JV, Ko DT. Meta-analy-
Intervention Treatment of Angina 2], SWISSI II [SWiss Inter- sis: Effects of Percutaneous Coronary Intervention Versus Medical Therapy on
ventional Study on Silent Ischemia 2], MASS II [Medicine, An- Angina Relief. Ann Intern Med. 2010;152:370-379.
5. Cohen DJ, Van Hout B, Serruys PW, et al; SYNTAX Investigators. Quality of Life
gioplasty, or Surgery Study 2], and COURAGE 2007) showed after PCI with Drug-Eluting Stents or Coronary-Artery Bypass Surgery. N Engl
that angina persists in 29%-31% of revascularized patients J Med. 2011;364:1016-1026.
from one to five years out.4 Despite the current technical ad- 6. Fox KA, Poole-Wilson PA, Henderson RA, et al. Interventional versus conserva-
tive treatment for patients with unstable angina or non-ST-elevation myocardial
vancements in revascularization procedures, angina is still infarction: the British Heart Foundation RITA 3 randomised trial. Lancet. 2002;
present at 12 months of follow-up in 29.4% of percutaneous 360:743-751.

Is targeting only stenosis sufficient to optimally improve angina? MEDICOGRAPHIA, Vol 38, No. 3, 2016 299
CONTROVERSIAL QUESTION

3. H. Hasan-Ali, Egypt
According to the so-called plaque-centric hypothesis, it was
thought that removing epicardial coronary stenosis by per-
Hosam HASAN-ALI, MD, PhD cutaneous coronary intervention could cure ischemic heart
A Board Member of the Egyptian Society
of Cardiology disease, and therefore, angina. However, in the COURAGE tri-
Head of Cardiovascular Medicine Department al (Clinical Outcomes Utilizing Revascularization and Aggres-
Assiut University Cardiac Hospital sive druG Evaluation), only 66% of stable angina patients
Assiut University Hospitals
Assiut Governorate, EGYPT treated by percutaneous coronary intervention were free from
(e-mail: hosam_hasan@hotmail.com) angina at one year, and 74% at 5 years.6 The recent reports
of persistent angina occurring after percutaneous coronary

T
raditionally, ischemic heart disease has been linked to intervention with evidence of ischemia in the absence of resid-
the presence of obstructive epicardial coronary artery ual stenosis or restenosis have showed that microvascular
disease.1 However, extensive data have failed to show ischemia may coexist in patients with epicardial obstructive
that all patients who have atherosclerotic obstructions have coronary artery disease.2,5 These findings shifted the para-
ischemic heart disease or, conversely, that all patients who digm of ischemic heart disease pathophysiology from the
have ischemic heart disease present with obstructive coro- traditional plaque-centric hypothesis to a multifactorial hy-
nary atherosclerosis.2 Obstructive coronary artery disease pothesis. This new modelwhich Marzilli et al 2 call the solar
has been reported in asymptomatic individuals and this is system of ischemic heart diseaseis centered around my-
referred to as silent ischemia.1,3 In contrast, obstructive coro- ocardial ischemia, and the orbiting planets are the six fac-
nary artery disease is absent in patients with typical angina1 tors that contribute to ischemia: epicardial coronary artery
and in patients with positive non-invasive testing.3 This is more obstruction, endothelial dysfunction, microvascular dysfunc-
obvious in women than in men.3 More than half the women tion, coronary spasm (microvascular and epicardial), spon-
with stable chest pain undergoing coronary angiography do taneous thrombosis and platelet aggregation, and inflamma-
not have obstructive coronary artery disease, while this is true tion. Since epicardial coronary artery obstruction is only one
for only one-third of men.4 of these factors, targeting epicardial coronary stenosiswhen
it is presentis actually only one step in the management of
Traditionally, this was considered to be due to false positive ischemic heart disease. A more comprehensive approach that
non-invasive tests.3 Patients with stable angina and normal also includes the myocardial cell and microvascular ischemia
coronary arteries or diffuse nonobstructive coronary artery is essential to improve patient morbidity and mortality.
disease were thought to experience little more than a re-
duction in their quality of life and have a benign prognosis; References
1. Cheng VY, Berman DS, Rozanski A, et al. Performance of the traditional age,
however, they actually have elevated risks of major adverse sex, and angina typicality-based approach for estimating pretest probability of
cardiovascular events and all-cause mortality compared with angiographically significant coronary artery disease in patients undergoing coro-
a reference population without ischemic heart disease.4 nary computed tomographic angiography: Results from the multinational coro-
nary CT angiography evaluation for clinical outcomes: An international multicen-
ter registry (confirm). Circulation. 2011;124:2423-2432.
In addition, large myocardial infarction registries have showed 2. Marzilli M, Merz CN, Boden WE, et al. Obstructive coronary atherosclerosis and
an absence of flow-limiting coronary pathology in 5%-25% ischemic heart disease: An elusive link! J Am Coll Cardiol. 2012;60:951-956.
3. Patel MR, Peterson ED, Dai D, et al. Low diagnostic yield of elective coronary
of cases.5 This changed our view of this type of patients and angiography. New Engl J Med. 2010;362:886-895.
the term cardiac syndrome X emerged to describe patients 4. Jespersen L, Hvelplund A, Abildstrom SZ, et al. Stable angina pectoris with no
who show signs of ischemic heart disease in the absence of obstructive coronary artery disease is associated with increased risks of major
adverse cardiovascular events. Eur Heart J. 2012;33:734-744.
obstructive epicardial coronary artery disease.5 In these pa- 5. Cocco G, Jerie P. Angina pectoris in patients without flow-limiting coronary ar-
tients the pathophysiological mechanisms underlying ischemic tery disease (cardiac syndrome x). A forest of a variety of trees. Cardiol J. 2015;
heart disease are endothelial dysfunction and microvascular 22(6):605-612.
6. Boden WE, O'Rourke RA, Teo KK, et al; Group CTR. Optimal medical therapy
dysfunction, sometimes associated with coronary microvas- with or without PCI for stable coronary disease. New Engl J Med. 2007;356:
cular spam and epicardial coronary artery spasm.2,5 1503-1516.

300 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Is targeting only stenosis sufficient to optimally improve angina?
CONTROVERSIAL QUESTION

4 . H. Q. T. Ho, Vietnam
nists, and nitrates) alone or in combination to control their
symptoms. Despite this extensive use of antianginal drugs,
only 66% of patients were free from angina after 3 years of
follow-up. The results of the COURAGE and BARI 2D trials
Huynh Quang Tri HO, MD, PhD indicate that targeting coronary artery stenosis only is far from
Head of Intensive Care Unit, sufficient to optimally improve angina.
Heart Institute of Hochiminh City
VIETNAM
(email: hohuynhquangtri@yahoo.com) On the other hand, myocardial ischemia in the absence of ob-
structive coronary disease is a marker of poor prognosis. In

W
hen treating patients with chronic stable angina, the WISE study (Womens Ischemia Syndrome Evaluation),
clinicians should aim at reducing anginal symp- women with myocardial ischemia (seen on magnetic reso-
toms, which will improve their patients exercise nance spectroscopy) who did not have obstructive coronary
capacity and quality of life. The traditional approach to symp- disease had a similarly high rate of hospitalization for unsta-
tom reduction is the prescription of hemodynamic drugs such ble angina than women with obstructive coronary disease.3
as b-blockers, calcium antagonists, and nitrates. In case of
poor response to medical therapy, coronary revascularization Trimetazidine, an antianginal drug that controls myocardial is-
with percutaneous coronary intervention (PCI) or coronary ar- chemia through intracellular metabolic changes, represents a
tery bypass graft surgery (CABG) is often performed. The ra- useful alternative and can be used as add-on therapy to he-
tionale for prescribing hemodynamic drugs and performing modynamic antianginal drugs. A growing body of evidence
coronary revascularization as the next step in the treatment of supports the antianginal efficacy of trimetazidine, alone or in
angina is the assumption that the unique cause of angina is combination.
coronary artery stenosis.
The data described here point to the conclusion that the treat-
However, numerous clinical studies have proven the short- ment of angina should focus on myocardial ischemia rather
comings of this traditional approach. In the COURAGE trial than solely on coronary artery stenosis. Accordingly, the lat-
(Clinical Outcomes Utilizing Revascularization and Aggressive est ESC guidelines on the management of stable coronary
druG Evaluation), 2287 patients with stable coronary artery artery disease have recognized the important role of meta-
disease were randomized to undergo PCI with optimal med- bolic agents like trimetazidine.4
ical therapy (PCI group) or optimal medical therapy alone.1
Optimal medical therapy included antiplatelet agents, a statin,
References
and hemodynamic antianginal drugs alone or in combination. 1. Boden WE, ORourke RA, Teo KK, et al; COURAGE Trial Research Group. Op-
Although antianginal drugs were widely prescribed, 21.1% timal medical therapy with or without PCI for stable coronary disease. N Engl
of patients in the PCI group needed to undergo additional J Med. 2007;356:1503-1516.
2. Dagenais GR, Lu J, Faxon DP, et al; BARI 2D Study Group. Effects of optimal
revascularization, and at the end of the study, only 74% of medical treatment with or without coronary revascularization on angina and sub-
patients were free of angina. The design of the BARI 2D trial sequent revascularizations in patients with type 2 diabetes mellitus and stable
(Bypass Angioplasty Revascularization Investigation 2 dia- ischemic heart disease. Circulation. 2011;123:1492-1500.
3. Johson BD, Shaw LJ, Buchthal SD, et al. Prognosis in women with myocardial
betes), which was carried out in 2364 patients with coronary ischemia in the absence of obstructive coronary disease. Results from the Na-
artery disease and type 2 diabetes, was similar to that of the tional Institutes of Health-National Heart, Lung, and Blood Institute-sponsored
COURAGE trial.2 In the group of patients who underwent Womens Ischemia Syndrome Evaluation (WISE). Circulation. 2004;109:2993-
2999.
prompt coronary revascularization (PCI or CABG), more than 4. Montalescot G, Sechtem U, Achenbach S, et al. 2013 ESC guidelines on the man-
90% needed antianginal drugs (b-blockers, calcium antago- agement of stable coronary artery disease. Eur Heart J. 2013;34:2949-3003.

Is targeting only stenosis sufficient to optimally improve angina? MEDICOGRAPHIA, Vol 38, No. 3, 2016 301
CONTROVERSIAL QUESTION

5 . D. Isaza-Restrepo, Colombia
effects of dichloroacetate, a drug that increases glucose ox-
idation by stimulating pyruvate dehydrogenase, have been
Daniel ISAZA-RESTREPO, MD, FACC studied in patients with ischemic heart disease.5 However,
Director Coronary Care Unit
Fundacin Cardioinfantil some pharmacokinetic and pharmacodynamic issues need to
Director of Cardiology Fellowship program be clarified before its real clinical benefit can be determined.5
Universidad del Rosario, and Universidad As mentioned above, these methods either require further ev-
del Bosque at Fundacin Cardioinfantil
Bogot, COLOMBIA idence or are not used because of concerns regarding their
(email: disaza@cardioinfantil.org) safety.

C
ardiac energy metabolism alterations are the main Taking into account these issues, trimetazidine appears to
pathophysiological factor involved in many heart con- be the best evidence-based option to optimize metabolism in
ditions, such as ischemic heart disease, where oxy- ischemic heart disease. It competitively inhibits long-chain
gen delivery is impaired by the presence of stenosis.1 Oxygen 3-ketoacyl CoA thiolase, and therefore inhibits b-oxidation and
is required to produce enough energy to maintain cardiac stimulates glucose oxidation. Meta-analyses have proven its
function using substrates such as glucose, free fatty acids clinical benefit in patients suffering from angina pectoris sec-
(FFAs), and proteins. The lack of oxygen caused by ischemic ondary to ischemic heart disease, and have suggested that
heart disease alters the metabolic pathways of individual cells. it may be beneficial in patients with heart failure.6,7
Current data show that even after successful revasculariza-
tion a third of angina patients still suffer from pain. Reasons As exposed previously, we can be sure that modifying the
for this include: mechanical aspects such as neointimal hy- cardiomyocytes metabolic machinery in patients with coro-
perplasia/restenosis, incomplete revascularization, athero- nary artery disease improves ischemic symptoms further than
sclerotic plaque progression, microvascular dysfunction, or mechanical coronary interventions alone. This knowledge
coronary vasospasm. However, the diffuse nature of angina opens up a broad range of possibilities for improving the qual-
and the fact that ischemia affects the metabolism of every car- ity of life of our patients. The results of the ongoing ATPCI clin-
diomyocyte may perpetuate this condition. ical trial (efficAcy and safety of Trimetazidine in Patients with
angina pectoris having been treated by percutaneous Coro-
Modifying the energy substrate supply has been proposed as nary Intervention), which is currently enrolling patients with
a way to improve the metabolic performance of cardiomyo- angina pectoris undergoing PCI who are then randomized to
cytes. Initially, studies that used an infusion of glucose-insulin- trimetazidine or placebo and treated for 2 to 4 years, are ex-
potassium to increase the rate of glycolysis and decrease the pected to further our knowledge in this area.
bioavailability of FFAs to potentiate cardiac metabolism were
not conclusive in showing clinical benefit.2 Other drugs, such References
as fibrates, niacin, and nicotinic acidwhich act as PPAR lig- 1. Taegtmeyer H, Young ME, Lopaschuk GD, et al. Assessing Cardiac Metabolism:
A Scientific Statement From the American Heart Association. Circ Res. 2016;
andactivated nuclear hormone receptor agonists and de- 118(10):1659-1701.
crease triglyceride levels, and therefore FFA levels, thereby 2. Selker HP, Beshansky JR, Sheehan PR, et al. Out-of-hospital administration of
reducing the rate of b-oxidationshowed benefits but their intravenous glucose-insulin-potassium in patients with suspected acute coro-
nary syndromes: the IMMEDIATE randomized controlled trial. JAMA. 2012;307:
side effects casted doubt about their clinical usefulness.3 1925-1933.
3. Canner PL, Berge KG, Wenger NK, et al. Fifteen year mortality in Coronary Drug
Modifying enzyme expression should theoretically be bene- Project patients: long-term benefit with niacin. J Am Coll Cardiol. 1986;8:1245-
1255.
ficial, but more clinical evidence is needed to support the 4. Cole PL, Beamer AD, McGowan N, et al. Efficacy and safety of perhexiline maleate
use of etomoxir, oxfenicine, and perhexiline. These drugs in- in refractory angina. A double-blind placebo-controlled clinical trial of a novel
hibit the action of malonyl-CoA decarboxylase, an enzyme in antianginal agent. Circulation. 1990;81:1260-1270.
5. Wargovich TJ, MacDonald RG, Hill JA, Feldman RL, Stacpoole PW, Pepine CJ.
the fatty acid synthesis pathway, which degrades malonyl- Myocardial metabolic and hemodynamic effects of dichloroacetate in coronary
CoA. In turn, malonyl-CoA acts as an inhibitor of carnitine artery disease. Am J Cardiol. 1988;61:65-70.
palmitoyltransferase-1 (CPT-1), a key enzyme involved in b-ox- 6. Ciapponi A, Pizarro R, Harrison J. Trimetazidine for stable angina. Cochrane Da-
tabase Syst Rev. 2005;CD003614.
idation. This prevents the use of FFAs as an energy substrate, 7. Gao D, Ning N, Niu X, Hao G, Meng Z. Trimetazidine: a meta-analysis of ran-
thereby optimizing energy production in cardiomyocytes.4 The domised controlled trials in heart failure. Heart. 2011;97:278-286.

302 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Is targeting only stenosis sufficient to optimally improve angina?
CONTROVERSIAL QUESTION

6 . T. Kovarnik, Czech Republic


Post-revascularization AP occurs when:
(i) Revascularization is inappropriately indicated. The only un-
questionable indication for coronary revascularization in sta-
ble patients is the presence of significant ischemia with (SAP)
Tomas KOVARNIK, MD, PhD or without (silent ischemia) angina pectoris. Revascularization
Associate Professor of Internal Medicine simply cannot overcome the inherent risk of complications
Charles University Hospital
Prague, CZECH REPUBLIC (in-stent restenosis, periprocedural myocardial necrosis, in-
(email: Tomas.Kovarnik@vfn.cz) stent thrombosis, bleeding during antithrombotic treatment,
potential wound infection, and many others) in patients with-

A
ngina pectoris (AP) refers to chest pain caused by out significant ischemia. Moreover, in patients without evi-
myocardial ischemia and is actually only a symptom. dence of ischemia, chest pain may have other causes, and its
Myocardial ischemia can develop either gradually recurrence after revascularization cannot be considered as
and cause clinical manifestations known as stable angina treatment failure.
pectoris (SAP)or suddenlywith the development of acute (ii) There is a coronary etiology. In this case, AP recurs after
coronary syndrome (ACS). appropriately indicated and well-performed revascularization.
This relapse can be caused by target lesion failure (in-stent
In both clinical scenarios the extent of myocardial ischemia restenosis, in-stent thrombosis) or the progression of a lesion
is the strongest predictor of prognosis. Myocardial ischemia that was not significant at the time of revascularization. Revas-
can be treated by (i) improving blood flow to the myocardi- cularization itself does not affect the process of atherosclero-
um (revascularization), (ii) decreasing myocardial demand on sis, which can continue unless appropriate medication is pre-
blood flow (eg, with b-blockers, calcium channel blockers, scribed. These conditions must be treated according to the
and ivabradinea drug that lowers oxygen consumption and underlying causes.
increases blood flow to the myocardium by decreasing the (iii) AP persists despite appropriately indicated revasculariza-
heart rate), and (iii) increasing myocardial metabolic efficiency tion, which may be due to incomplete revascularization (an-
when blood supply is limited (eg, with trimetazidine, a drug other significant stenosis that has been left untreated), the
that improves the efficiency of energy production in myocar- presence of microvessel disease, a technical complication
dial cells suffering from ischemia by shifting their metabolism during PCI (peripheral embolization, untreated dissection), or
from free-fatty-acid b-oxidation back to glycolysis). Nitrates stretch pain (probably due to extension of vessel adventitia).
fall somewhere between revascularization and the latter two
conservative strategies, because they can enlarge coronary Several smaller studies have suggested that the incidence of
arteries and increase blood flow, while at the same time de- chest pain is lower after implantation of fully biodegradable
creasing preload (by venous dilatation). This action, in turn, stents rather than metallic stents. However, the large ran-
decreases oxygen consumption. domized controlled trial ABSORB 3 did not confirm this find-
ing.3 In this trial, AP occurred in 18% of patients, both in the
The greater the extent of myocardial ischemia, the greater biodegradable and drug-eluting stent groups.
the benefit of revascularization will be. Revascularization not
only limits AP better than conservative therapy, but it also Conclusion
improves the prognosis in patients with significant stenosis For every patient it is necessary to determine whether chest
located in the left main coronary artery; significant stenosis pain is caused by ischemia, and if it is, to what extent. Based on
located in a proximal part of the left anterior descending ar- this information, the treatment can be properly tailored to each
tery; significant stenosis located in two or three major vessels patient to avoid unnecessary revascularization, which would
together with systolic dysfunction of the left ventricle (ejec- not only without any benefit, but could actually be harmful.
tion fraction <45%); myocardial ischemia affecting a large area
(>10%); and significant stenosis located in the last patent coro- References
1. Windecker S, Kolh P, Alfonso F. 2014 ESC/EACTS Guidelines on myocardial
nary artery.1 Revascularization. Eur Heart J. 2014;35:2541-2619.
2. Abbate A, Biondi-Zoccai G, Agostoni P, Lipinski M, Vetrovec G. Recurrent angi-
However, about 30% patients suffer from AP after successful na after coronary revascularization: a clinical challenge. Eur Heart J. 2007;28:
1057-1065.
revascularization, which significantly impairs their quality of 3. Kereiakes D. ABSORB 3 trial. Presented at: TCT 2015; October 2015; San Fran-
life.2 cisco, CA.

Is targeting only stenosis sufficient to optimally improve angina? MEDICOGRAPHIA, Vol 38, No. 3, 2016 303
CONTROVERSIAL QUESTION

7. O. H. Masoli, Argentina
Coronary reserve
A good understanding of the physiology of the coronary cir-
culation is necessary to understand the concept of coronary
reserve. In a nutshell, coronary reserve refers to the ability of
Osvaldo H. MASOLI, MD, FACC, FESC the vascular circuits to adapt to myocardial oxygen consump-
Chief of Cardiac Imaging tion by vasodilation. Depending on the level of endothelial health
Image Department of TCba Salguero
Buenos Aires, ARGENTINA and on the extent of the mechanical obstruction present in
(email: ohmasoli@fibertel.com.ar) the epicardial arteries as a result of the atherosclerotic process,
an adaptive process called vascular remodeling may take place.
Determinants of myocardial flow This process initially leads to an increase in vessel diameter.

T
he coronary circulation is composed of the epicardial When atherosclerosis involves the lumen, the percentage of
coronary arteries, which are of large caliber, and the obstruction will affect the coronary reserve capacity, ie, the
resistance vessels, which are less than 300 m in di- ability to lower vascular resistance, and thus increase flow, so
ameter. Whereas the epicardial vessels exert little or no flow that the arterial blood can reach peripheral tissues without
resistance, flow resistance in the resistance vessels increas- altering cellular metabolism. But this compensation mecha-
es gradually as the vessel diameter decreases to less than nism is limited. Unless we try to stop the progress of ather-
100 m (eg, in arterioles). Exchange of substances between osclerosis or remove the obstruction, with medication and/or
blood and tissue occurs at the capillary level. invasive treatments such as myocardial revascularization pro-
cedures, the damage may be irreversible and permanently
In the myocardium, blood flow largely depends on the pres- alter the affected vessel, and thus tissue function.
sure gradient between the aortic root and the left atrium (coro-
nary pressure). Under normal conditions, coronary pressure Conclusion
is fully maintained in the epicardial vessels, with minimal or no The mechanisms involved in the genesis of myocardial ische-
loss of pressure in the distal epicardial arteries. In contrast, mia and its manifestation as angina are very complex and
intracoronary pressure decreases along the microvascula- go beyond the epicardial arteries. Therefore, efforts aiming to
ture to a pressure of 20-30 mm Hg (with most of the pres- normalize the lumen of the arteries or bypass the obstruction
sure dissipating in vessels with a diameter of 100-300 m). only act on part of the problem. Fortunately, we now have
drugs that can improve cell metabolism, such as trimetazidine,
As a result of a decrease in microvascular resistance caused or new drugs that can reduce the heart rate, such as ivabra-
by metabolic changespossibly involving adenosine, a met- dine, a drug whose pleiotropic effects lead to vasodilatation
abolite of adenosine monophosphate which induces vascu- and the release of nitric oxide, and thus improve endothelium
lar muscle relaxationwork-related myocardial flow increases. function. But we should not forget all the other drugs that
have been shown to have a beneficial effect on the outcome
The resulting flow increase is augmented by endothelium-de- of patients with coronary heart disease, nor the positive ef-
pendent factors, and this faster flow exerts more shear stress fects of healthy eating and physical activity.
on the endothelium, stimulates the enzyme nitric oxide syn-
References
thase (eNOS), triggering the release of nitric oxide, which 1. Masoli O, Balio NP, Sabat D, et al. Effect of endothelial dysfunction on region-
relaxes smooth muscles. In this scenario, endothelial cells and al dysfunction on regional perfusion in myocardial territories supplied by nor-
smooth muscle cells interact closely, which helps the ves- mal and diseased vessels in patients with coronary artery disease. J Nucl Cardiol.
2000;7:199-204.
sels adjust their diameter according to changes in flow rate in 2. Schelbert HR. Anatomy and physiology of coronary blood flow. J Nucl Cardiol.
the microvascular and epicardial vessels. 2010;4:545-554.

304 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Is targeting only stenosis sufficient to optimally improve angina?
CONTROVERSIAL QUESTION

8 . A. N. Parkhomenko and O. S. Gurjeva, Ukraine


creases the number of dysfunctional mitochondria, leads to
collagen deposition, macrophage infiltration, fibrosis, deple-
tion of sarcomeres, and activates apoptosis. More profound
changes occur in stunned myocardium and result from in-
hibition of Na-K-ATPase, increased intracellular Na+ level, Ca2+
overload, stimulation of reactive oxygen species (ROS) pro-
duction, and depression of contractile function in near nor-
mal coronary flow. Revascularization partially attenuates these
Alexander N. PARKHOMENKO, MD, PhD changes, but patients may still experience coronary micro-
Olga S. GURJEVA, MD, PhD, MH embolization and microvascular obstruction, which may lead
Ukrainian Institute of Cardiology - Kiev, UKRAINE
(e-mail: aparkhomenko@yahoo.com) to abnormal gene expression and apoptosis.5 Apoptosis oc-
curs in low-flow states and may be triggered by reperfusion

M
icrovascular angina (MVA) is found in about one- injury and severe energy depletion and follows either intrin-
third of patients undergoing coronary angiography sic mitochondria-mediated or extrinsic membrane-mediated
for angina who have no significant obstructive coro- pathways. The intrinsic pathway involves mechanisms that
nary lesions, and is strongly associated with adverse long- impact the functioning of mitochondrial permeability transi-
term prognosis.1 In patients with normal, near-normal, or tion pores (mPTPs). The extrinsic pathway is receptor-mediat-
restored epicardial flow, several factors contributing to an ed and may be activated by oxidative stress late after reper-
imbalance between oxygen demand and supply may coexist. fusion. Reperfusion injury and slow-reflow states contribute
About half the patients with coronary artery disease (CAD) to necrosis, increased mitochondrial membrane permeability,
have concomitant hypertension, and a growing proportion of cell swelling, lysis, fragmentation of cellular structures, activa-
elderly patients may present with aortic stenosis (AS). In this tion of inflammatory pathways, and leukocyte infiltration.
group of patients, increased oxygen demand is the result of
increased left ventricular end-systolic pressure, left ventricular These maladaptive cascades often partially persist after epi-
hypertrophy, impaired diastolic function, increased heart rate, cardial flow is restored and should be addressed medically.
and increased wall stress. As left ventricular hypertrophy pro- Metabolic agents such as trimetazidine counteract the effect
gresses, coronary flow reserve may be affected by increased of myocardial ischemia on mitochondrial membrane perme-
diastolic filling pressure, which compresses the endocardi- ability by diminishing oxidative stress and inhibiting mPTP
um, impairs perfusion, and reduces capillary distribution.2 In opening, and also reduce caspase 3 activity and apoptosis.6
patients with AS, MVA has been attributed to preexisting ab- The mechanisms that underlie angina are so complex that it
normal resting arteriolar vasodilation in patchily distributed cannot be resolved by relying on revascularization only; other
microvasculature preserving myocardial perfusion and ab- nonmechanistic approaches are therefore needed to man-
normally constricted prearteriolar vessels preventing distal age patients after revascularization.
pressure overload.3
References
MVA may follow an endothelium-dependent or an endothe- 1. Jespersen L, Hvelplund A, Abildstrm S, et al. Stable angina pectoris with no
lium-independent pattern. According to the current in-depth obstructive coronary artery disease is associated with increased risks of major
understanding of the pathways underlying angina symptoms, adverse cardiovascular events. Eur Heart J. 2012;33(6):734-744.
2. Rajappan K, Rimoldi OE, Camici PG, et al. Functional changes in coronary mi-
there is a close correlation between the ratio of oxygen de- crocirculation after valve replacement in patients with aortic stenosis. Circulation.
mand to oxygen supply and cardiac energy metabolism (sub- 2003;107:3170-3175.
strate utilization, production of ATP by oxidative phosphory- 3. Ong P, Athanasiadis A, Borgulya G, Mahrholdt H, Kaski JC, Sechtem U. High
prevalence of a pathological response to acetylcholine testing in patients with sta-
lation, and ATP transfer and utilization).4 The cardiac metabolic ble angina pectoris and unobstructed coronary arteries. The ACOVA Study (Ab-
system is very flexible, and can switch from one energy source normal COronary VAsomotion in patients with stable angina and unobstructed
to another. However, its adaptive capacity decreases in states coronary arteries). J Am Coll Cardiol. 2012;59:655-662.
4. Neubauer S. The failing heartan engine out of fuel. N Engl J Med. 2007;356
associated with increased oxygen demand or steadily de- (11):1140-1151.
creased coronary blood flow, triggering metabolic remodel- 5. Reffelmann T, Kloner RA. The "no-reflow" phenomenon: basic science and clin-
ing. Prolonged energy deficit triggers the expression of fetal ical correlates. Heart. 2002;87(2):162-168.
6. Hu B, Li W, Xu T, Chen T, Guo J. Evaluation of trimetazidine in angina pectoris
genes and a switch from fat to glucose metabolism, stimu- by echocardiography and radionuclide angiography: a meta-analysis of random-
lates glycogen accumulation and changes in cell signaling, in- ized, controlled trials. Clin Cardiol. 2011;34(6):395-400.

Is targeting only stenosis sufficient to optimally improve angina? MEDICOGRAPHIA, Vol 38, No. 3, 2016 305
CONTROVERSIAL QUESTION

9 . C. K. Ponde, India
propriate in those whose probability of having angina is inter-
mediate. Estimation of coronary flow reserve is a boon for those
in whom angiography shows borderline stenosis (70%). A
C. K. PONDE, MD, DM(Card), DNB (Card),
FACC, FSCAI, FCSI, FIAE, FISE, FICC fractional flow reserve (FFR) of 0.75 or less is usually con-
Consultant cardiologist sidered to be a good indication for PCI. The DEFER trial has
Head, Department of Cardiology shown that performing PCI for lesions with a FFR of 0.75 or
P.D. Hinduja National Hospital
Mumbai, INDIA more does not improve symptoms nor prognosis.6
(email: ckpshekhar@yahoo.co.in)
Achieving and maintaining an optimal body weight and fol-

T
he number of percutaneous coronary angioplasty pro- lowing a graded exercise training program are known to im-
cedures (PCI) performed in patients with chronic stable prove exercise capacity in patients with CSA. Achieving an
angina (CSA) has increased tremendously in the last optimal hematocrit and excellent blood pressure control, and
two decades.1 In a series of 2000 patients with CSA (of whom lowering LDL-C levels to below 70 mg/dL are all extremely
39% underwent PCI and 28% CABG) almost a third had mul- important ways to improve the prognosis of these patients.
tiple episodes of angina per week after 6 months of follow-up.2 Post-PCI stretch pain is usually treated with analgesics and is,
Moreover, the Euro Heart Survey reports that 60% of patients thankfully, self-limiting. The functional causes of recurrent angi-
with persistent angina post-PCI are moderately/severely dis- na (microvascular dysfunction/epicardial coronary spasm) re-
abled. spond best to diltiazem and long-acting nitrates. Trimetazidine
(an inhibitor of fatty acid oxidation), which improves angina
The most common causes of persistent/recurrent angina post- by shifting myocardial metabolism toward glucose oxidation
PCI are either structural (stretch pain, in-stent restenosis, (TRIMPOL II study)7 has also been found to be useful in such
in-stent thrombosis, incomplete revascularization, or progres- patients. b-Blockers should be prescribed to all post-PCI pa-
sion of coronary atherosclerosis) or functional (microvascu- tients unless contraindicated. Statins and angiotensin-con-
lar dysfunction or epicardial coronary spasm). A recent meta- verting enzyme (ACE) inhibitors should also be part of OMT in
analysis of randomized clinical trials comparing PCI versus most patients. High compliance rates with OMT, such as those
optimal medical therapy (OMT) in patients with CSA has shown obtained in the COURAGE trial, (90% at 5 years) are not easy
that PCI does not reduce the risk of mortality, cardiovascu- to obtain in clinical practice unless combinations are used to
lar death, nonfatal myocardial infarction, or revascularization reduce the number of pills patients are prescribed. Thankful-
procedures; however, it provides greater relief from angina ly in India various combinations are freely available. Recent-
compared with OMT, at least in the first year.3 Most interna- ly, the BEAUTIFUL and ASSOCIATE trials have shown some
tional guidelines recommend revascularization procedures promising results for the use of ivabradine as an add-on ther-
in CSA only when symptoms are not controlled by OMT. apy or in those in whom b-blockers are contraindicated to
reduce the frequency of angina attacks and the number of
In-stent restenosis usually manifests between 4 and 8 months cardiac events.
after PCI and is associated with angina and objective evi-
References
dence of myocardial ischemia on provocative testing.4 If OMT 1. Ko DT, Tu JV, Samadashvili Z, et al. Temporal trends in the use of percutaneous
fails to control it, repeat revascularization is usually required. coronary intervention and coronary artery bypass surgery in New York State and
However, the use of drug-eluting stents (DESs) has substan- Ontario. Circulation. 2010;121:2635-2644.
2. Vetrovec GW, Watson J, Chaitman B, Cody R, Wenger N. Symptoms persist in
tially reduced its occurrence. patients with chronic angina despite frequent antianginal use and prior revas-
cularization. J Am Coll Cardiol. 2004;43(Suppl. II):A281.
Inappropriate vasoconstriction of small vessels in the distal coro- 3. Pursnani S, Korley F, Gopaul R, et al. Percutaneous coronary intervention versus
optimal medical therapy in stable coronary artery disease, a systematic review
nary bed of the target vessel is a frequent cause of positive and meta-analysis of randomized clinical trials. Circ Cardiovasc Interv. 2012;
stress tests after successful angioplasty.5 Stent implantation 5:476-490.
also induces distal coronary endothelial dysfunction. Exercise- 4. Holmes DR Jr. In-stent restenosis. Rev Cardiovasc Med. 2001;2:115-119.
5. Ito S, Nakasuka K, Morimoto K, et al. Angiographic and clinical characteristics of
induced spasm of a large epicardial coronary artery in the dis- patients with acetylcholine-induced coronary vasospasm on follow-up coronary
tal post-stent segment has been documented and reported. angiography following drug-eluting stent implantation. J Invasive Cardiol. 2011;
The two most frequent causes of early post-CABG angina are 23:57-64.
6. Pijls NH, Van Schaardenburgh P, Manoharan G, et al. Percutaneous coronary
anastomotic site lesions and rapid venous graft degeneration/ intervention of functionally nonsignificant stenosis: 5-year follow-up of the DEFER
thrombosis. A thorough clinical evaluation is crucial in such Study. J Am Coll Cardiol. 2007;49:2105-2111.
patients. In those with established angina, direct coronary an- 7. Szwed H, Sadowski Z, Elikowski W. Combination treatment in stable effort angi-
na using trimetazidine and metoprolol: results of a randomized, double-blind, mul-
giography should be performed, while non-invasive stress tests ticentre study (TRIMPOL II). TRIMetazidine in POLand. Eur Heart J. 2001;22:
(vs myocardial scintigraphy/stress echocardiography) are ap- 2267-2274.

306 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Is targeting only stenosis sufficient to optimally improve angina?
CONTROVERSIAL QUESTION

10. V. Sansoy, Turkey


chemia in elderly patients with multivessel coronary artery dis-
ease and diabetes mellitus. In addition, pretreatment or con-
comitant treatment with trimetazidine seems to have a cardio-
Vedat SANSOY, MD, FESC protective effect in patients undergoing percutaneous
Professor of Cardiology coronary intervention. These studies have shown that
Institute of Cardiology trimetazidine treatment results in an improvement in several
University of Istanbul
Istanbul, TURKEY ischemic parameters, such as a reduction in the frequency of
(email: vedat.sansoy@gmail.com) angina pectoris attacks and of myocardial damage during
percutaneous coronary intervention and coronary artery by-

D
espite the progress made by the various therapeutic pass graft surgery. These benefits are achieved because
methods used in cardiology in the last decades, coro- trimetazidine protects the heart from ischemic damage and
nary artery disease remains the leading cause of mor- oxidative stress. In addition, trimetazidine has also been
tality and morbidity worldwide. Percutaneous coronary inter- shown to improve left ventricular function in the follow-up pe-
vention is an effective and safe treatment to relieve severe riod after percutaneous angioplasty.6
stenosis in patients with coronary artery disease, but studies
have shown that many patients still suffer from recurrent angi- References
1. Hueb W, Soares PR, Gersh BJ, et al. The Medicine, Angioplasty, or Surgery trial
na or silent myocardial ischemia after revascularization.1 (MASS-II): a randomized, controlled, clinical trial of three therapeutic strategies
for multivessel coronary artery disease. One-year results. J Am Coll Cardiol.
Traditional antianginal agents for the treatment of stable angi- 2004;43:1743-1751.
2. Kolh P, Windecker S, Alfonso F, et al; Task Force on Myocardial Revasculariza-
na pectoris include nitrates, calcium antagonists, and b-block- tion of the European Society of Cardiology and the European Association for
ers, which reduce angina attacks either by reducing ATP con- Cardio-Thoracic Surgery; European Association of Percutaneous Cardiovascu-
sumption via a reduction in the heart rate and blood pressure lar Interventions. 2014 ESC/EACTS Guidelines on myocardial revascularization:
the Task Force on Myocardial Revascularization of the European Society of
or by increasing ATP production through an increase in coro- Cardiology (ESC) and the European Association for Cardio-Thoracic Surgery
nary blood flow.2 Optimizing myocardial metabolism with meta- (EACTS). Developed with the special contribution of the European Association
bolic agents is a new strategy that can be used in patients of Percutaneous Cardiovascular Interventions (EAPCI). Eur J Cardiothorac Surg.
2014;46:517-592.
with stenotic coronary artery disease. These drugs represent 3. Kantor PF, Lucien A, Kozak R, Lopaschuk GD. The antianginal drug trimetazidine
a new class in the treatment of ischemic heart disease. Tri- shifts cardiac energy metabolism from fatty acid oxidation to glucose oxidation
metazidine is a metabolic agent, and unlike conventional an- by inhibiting mitochondrial long-chain 3-ketoacyl coenzyme A thiolase. Circ Res.
2000;86:580-588.
tianginal drugs, it restores the balance between myocardial 4. Desideri A, Celegon L. Metabolic management of ischemic heart disease: clin-
oxygen supply and demand by selectively inhibiting the long- ical data with trimetazidine. Am J Cardiol. 1998;82:50K-53K.
chain 3-ketoacyl coenzyme A thiolase, thus partially suppress- 5. Xu X, Zhang W, Zhou Y, et al. Effect of trimetazidine on recurrent angina pec-
toris and left ventricular structure in elderly multivessel coronary heart disease pa-
ing the b-oxidation of fatty acids, stimulating glucose metab- tients with diabetes mellitus after drug-eluting stent implantation: a single-centre,
olism, and increasing myocardial ischemic tolerance.3,4 Xu prospective, randomized, double-blind study at 2-year follow-up. Clin Drug In-
et al 5 have showed that adjunctive therapy with trimetazidine vestig. 2014;34:251-258.
6. Bonello L, Sbragia P, Amabile N, et al. Protective effect of an acute oral loading
after drug-eluting stent implantation reduces the incidence dose of trimetazidine on myocardial injury following percutaneous coronary in-
and the severity of angina pectoris as well as that of silent is- tervention. Heart. 2007;93:703-707.

Is targeting only stenosis sufficient to optimally improve angina? MEDICOGRAPHIA, Vol 38, No. 3, 2016 307
CONTROVERSIAL QUESTION

11. D. Vassilev, Bulgaria


formed through radial access, and a JL 4, 6F guiding catheter
was used to canulate the LM. Two wires were placed in the
LAD and LCX. First, we predilated the LM-LAD with a balloon,
after which two overlapping stents (Biofreedom 3.0x28 mm
Dobrin VASSILEV, PhD and 3.5x24 mm) were implanted into the LAD. There was no
Head of cardiology department significant stenosis in the diagonal ostia (grade III TIMI flow).
Medical University of Sofia
Sofia, BULGARIA Another overlapping stent was placed in the LM and ostio-
(email: dobrinv@gmail.com) proximal part of the LAD (Biofreedom 3.5x24 mm). We then
dilated the stented region (LM and LAD) with noncompliant

I
schemic heart disease is a diffuse disease encompass- (NC) balloons. Because there was 70% stenosis in the LCX
ing not only the epicardial compartment, but also the mi- ostium we performed a balloon dilation of the LAD and LCX
crovascular compartment. This is one of the reasons for ostia. The final angiographic result showed no dissection and
continuing chest pain even after complete mechanical epi- no residual stenosis, and normal flow was restored in all treat-
cardial revascularization: about 40% of all bypassed or stent- ed vessels. A final intravascular ultrasound (IVUS) was per-
ed patients still have angina one year after the procedure.1 We formed in the LAD, LCX and left main artery, and showed no
present a case in which the patient had residual ischemia de- signs of dissection and stent strut malposition. The circum-
spite complete revascularization with the use of last-gener- flex artery ostium was not compromised, so there was no need
ation drug-eluting stents. In cases such as this onein which for additional stenting.
further revascularization is impossiblea metabolic drug that
can reduce ischemia may improve symptoms. The patient was discharged 2 days after the procedure; he
had no angina symptoms, nor any elevation of the markers
A fifty-two year-old male smoker, with a history of hyperten- of myocardial necrosis, and there were no adverse events. He
sion and dyslipidemia, was admitted to our clinic with stable was prescribed rosuvastatin, ramipril, bisoprolol, clopidogrel,
angina triggered by low-level physical activity, which he had and acetylsalicylic acid.
started to experience approximately one year earlier (class
III CCS). His ECG at rest was normal, and did not show any At 1 month of follow-up, there were no adverse events, but
signs of ischemia. Echocardiography did not show any kinet- the patient still had intermittent episodes of chest pain. The
ic dysfunction at rest; the ejection fraction was 53%, the left stress test at 11 METs (metabolic equivalents) was ECG neg-
ventricle was not dilated, and there were no significant valvular ative, with slight chest discomfort. For this reason we decid-
lesions. In addition, the cardiac markers of myocardial necro- ed to add trimetazidine to his treatment. At the next visitthe
sis were normal. following monththe patient was completely asymptomatic.
This case clearly illustrates the fact that even after complete
Coronary angiography, which was performed through right mechanical revascularization microvascular dysfunction can
radial access, showed 80% stenosis in the distal left main (LM) still cause discomfort. Increasing hemodynamic therapy pro-
artery. There was 70% stenosis in the proximal and middle vides no additional benefit in this kind of situation and the only
segments of the left anterior descending (LAD) artery, where option is to directly alter the ischemic threshold by adding a
there were two Medina 110 bifurcation stenotic lesions in the metabolic drug (eg, trimetazidine) according to the currently
first and second diagonal branches, respectively. The SYN- proposed algorithm for the treatment of angina.3
TAX score was 28. According to the ESC Guidelines for My-
ocardial Revascularization, the patient had left main artery dis- References
ease with a SYNTAX score of 23-32, which corresponds to 1. Cohen DJ, Van Hout B, Serruys PW, et al. Quality of life after PCI with drug-elut-
class I and level of evidence B for coronary artery bypass ing stents or coronary-artery bypass surgery. N Engl J Med. 2011;17;364(11):
1016-1026.
graft surgery (CABG).2 Treatment with percutaneous coronary 2. Windecker S, Kolh P, Alfonso F, et al. 2014 ESC/EACTS Guidelines on myocar-
intervention is class IIa with a level B of evidence. However, dial revascularization: The Task Force on Myocardial Revascularization of the
the Heart Team decided that percutaneous coronary interven- European Society of Cardiology (ESC) and the European Association for Cardio-
Thoracic Surgery (EACTS)Developed with the special contribution of the Euro-
tion was an option as the patient was reluctant to go through pean Association of Percutaneous Cardiovascular Interventions (EAPCI). Eur
CABG surgery. Heart J. 2014;35(37):2541-2619.
3. Task Force Members; Montalescot G, Sechtem U, Achenbach S, et al. 2013 ESC
guidelines on the management of stable coronary artery disease: the Task Force
Before the procedure, the patient was preloaded with clopi- on the management of stable coronary artery disease of the European Society
dogrel 600 mg and 500 mg aspirin. The procedure was per- of Cardiology. Eur Heart J. 2013;34(38):2949-3003.

308 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Is targeting only stenosis sufficient to optimally improve angina?

VA STA R E L M R

This overall improvement in


adenosine triphosphate produc-
tion with trimetazidine protects the
cell against acidosis and ionic im-
balance during ischemia. Notably,
trimetazidine has been shown to
Trimetazidine:
prevent calcium overload, the pro-
duction of free radicals, and apop-
tosis in the cardiac cell during re-
targeting the cardiac cell
perfusion. By preventing all these
deleterious effects, trimetazidine
maintains the contractile function
in stable angina
of the cardiac cell and reduces
anginal symptoms.

b y M . G l i e b ova , Fra n c e

A
ngina is now recognized as a multifaceted disease where coronary ar-
tery obstructions are only one among many contributing factors. Be-
cause of its central involvement in a number of ischemic processes,
the cardiac cell is recognized as a major player in cardiac ischemia. At the cell
level, myocardial ischemia is characterized by altered myocardial energy me-
tabolism, which results in an ionic imbalance, misuse of and deficit in energy,
and ultimately functional defects. During ischemia, trimetazidine acts direct-
ly at the cell level by decreasing fatty acid oxidation and increasing glucose
oxidation, which helps restore normal energy metabolism. This leads to an
overall improvement in the general function of the cell and, at the organ level,
Mariia GLIEBOVA, MD provides anti-ischemic activity. By directly targeting the cardiac cell, trimetazi-
Servier International dines mechanism of action complements those of hemodynamically active
Global Medical Strategy
and Information Department
antianginal therapies, thus maximizing clinical efficacy when used in a com-
Cardiovascular bination strategy. Trimetazidine has been shown to provide additional reduc-
Suresnes, FranCe tion in symptoms and increase exercise capacity in angina patients whose
symptoms are inadequately controlled by b-blockers and/or calcium chan-
nel blockers. The beneficial effect of trimetazidine has been demonstrated
in various angina patients, including those with a history of myocardial infarc-
tion, previous percutaneous coronary intervention, diabetes, or left ventricu-
lar dysfunction. In addition to its proven antianginal effect, trimetazidine may
also provide extra benefits in terms of cardioprotection and prognosis in many
cardiovascular patients.
Medicographia. 2015;37:309-316 (see French abstract on page 316)

table angina pectoris, a clinical manifestation of myocardial ischemia, is a chron-

S ic disease associated with poor prognosis and impaired quality of life. Patients
with severe angina are at greater risk for cardiovascular events, including my-
ocardial infarction or death, and the rate of hospitalizations is particularly high in this
population.

One therapeutic option for relieving angina symptoms is the use of invasive pro-
cedures, such as myocardial revascularization. However, this type of approach has
Address for correspondence: unclear effects on survival,1 and its use is restricted to angina patients with obstruc-
Dr Mariia Gliebova,
Servier International, 50 rue Carnot,
tive coronary artery stenosis. Thus, medical therapyby reducing the symptoms
92284 Suresnes Cedex, France of angina, increasing exercise tolerance, and improving quality of liferemains a
(email: mariia.gliebova@servier.com) cornerstone in the management of angina patients.2 antianginal drugs exert their
www.medicographia.com effects by reducing cardiac workload, ie, reducing myocardial oxygen consumption

Trimetazidine: targeting the cardiac cell in stable angina Gliebova MEDICOGRAPHIA, Vol 38, No. 3, 2016 309
VA STA R E L M R

and/or increasing oxygen supply to the heart. This is the mode Targeting the cardiac cell for increased
of action of standard antianginal treatments, such as b-block- clinical efficacy
ers or calcium channel blockers. another approach consists The recognition of myocardial ischemia as a multifactorial
in optimizing oxygen use by directly increasing energy produc- process implies that antianginal management should not sole-
tion at the cardiac level, such as with the anti-anginal agent ly focus on large coronary vessels, but also on the microves-
trimetazidine. In view of their complementary mechanisms sels and cardiac cell. One option would be to adjust treat-
of action, antianginal drugs are often used in combination for ment for each patient according to the underlying causes of
additive or synergic effects. Here we review the pharmaco- angina. However, this would prove difficult to diagnose in prac-
logical rationale and clinical efficacy of trimetazidine in patients
with stable angina pectoris, in light of recent experimental and
clinical findings. Origin Possible mechanisms of ischemia

Vascular
Angina: a single clinical entity with Macrovessels Flow-limiting stenosis (atherosclerosis)
multiple etiologies endothelial dysfunction
angina pectoris is generally recognized as the clinical expres- Vasospasm
sion of underlying coronary artery disease (CaD). In patients Muscle bridge
with CaD, myocardial ischemia results from flow-limiting ob- Inflammation
structions in the epicardial coronary arteries, as documented Microvessels Microvascular dysfunction
by coronary angiography. However, a significant proportion of endothelial dysfunction
patients with typical angina symptoms do not present obstruc- Spasm
tive lesions in their angiograms. In a recent report, 83.5% of Inflammation
Microemboli
angina patients with no previously documented CaD had no
evidence of obstructive CaD.3 These patients often present Nonvascular
with occult coronary abnormalities, which explain their angina Cardiomyocytes Impaired energy metabolism
Defective cellular oxygen transport
symptoms. These vascular defects often occur concomitantly,
Mitochondrial dysfunction
supporting the notion that multiple causative mechanisms in
Inflammation/fibrosis
anginaincluding endothelial dysfunction, microvascular im-
pairment, and myocardial bridgingare common.4 Beyond Table I. Multiple causes of stable ischemic heart disease.
coronary mechanisms, additional nonvascular mechanisms These different mechanisms of ischemia are not mutually exclusive and often
can contribute to myocardial ischemia (Table I). These are occur concomitantly during angina.
particularly relevant in a significant minority of angina patients Modified from reference 6: Pepine and Douglas. J Am Coll Cardiol. 2012;60(11):
957-959.
for whom there is no coronary explanation for their symptoms.4
Because of its central involvement in a number of ischemic tice. a more convenient approach would consist in a glob-
processes, including inflammation and impaired myocardial al therapeutic strategy that encompasses all causes of is-
energy metabolism, the cardiac cell is recognized as a major chemia. In the euro Heart Survey, over half of stable angina
nonvascular player in ischemic heart disease (Table I).5,6 Pro- patients (59%) were prescribed two or more antianginal drugs.7
gression in angina is a complex and multifactorial process, with Combining different anti-ischemic drugs, each with its own
both vascular and nonvascular contributing mechanisms. distinct mechanism of action, may improve clinical efficacy by
Coronary obstruction represents only one piece of the puzzle. additive or even synergistic effects. Moreover, a combination
strategy allows more treatment flexibility, by permitting drug
selection according to a patients comorbidities and/or car-
SELECTED ABBREVIATIONS AND ACRONYM S
diac function.
ATP adenosine triphosphate
ATPCI efficacy and safety of Trimetazidine in Patients with The heart requires a large amount of energy to support its
angina pectoris having been treated by percuta- continuous contractile activity, making energy metabolism a
neous Coronary Intervention
fundamental process in the cardiac cell. Under normal con-
CAD coronary artery disease
ditions, the main source of energy in the cardiac cell comes
DIETRIC estudio prospectivo en pacientes DIabticos de la
from the oxidative metabolism of fatty acids in the mitochon-
efectividad y tolerabilidad de la TrImetazidina en
asociacin al tratamiento previo de su enfermedad dria, which provides 70% of total adenosine triphosphate
Coronaria (aTP). However, the mitochondrial oxidation of glucose re-
MACCE major adverse cardiac and cerebrovascular event mains a more efficient metabolic process, as it requires 12%
METRO Management of angina: a reTrospective cOhort less oxygen than fatty acids for an equivalent aTP produc-
PCI percutaneous coronary intervention tion. During ischemia, where oxygen supply is limited, oxida-
TRIMPOL TrIMetazidine in POLand tive metabolism is dramatically reduced, and glycolysis (ie,
conversion of glucose to pyruvate) becomes a preponderant

310 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Trimetazidine: targeting the cardiac cell in stable angina Gliebova
VA STA R E L M R

source of energy (Figure 1).8 This alteration in myocardial me- imal studies11-13 and confirmed in a clinical setting,14 where
tabolism is accompanied by an uncoupling of glycolysis and treatment with trimetazidine for 3 months was shown to in-
glucose oxidation, an increase in intracellular lactate and pro- crease myocardial levels of high energy phosphates in heart
tons, and an ionic imbalance, with serious consequences at failure patients by 33%. This overall improvement in aTP pro-
the cellular level. These maladaptive metabolic changes cause duction with trimetazidine protects the cell against acidosis
a depletion in the cellular energy store, as evidenced by a de- and ionic imbalance during ischemia.12,15,16 notably, trimeta-
crease in aTP production and other high-energy phosphate zidine has been shown to prevent calcium overload,17 the pro-
levels. In order to maintain ionic homeostasis, aTP-depend- duction of free radicals,18 and apoptosis19 in the cardiac cell
ent ion transporters are activated to eject protons and sodium during reperfusion. By preventing all these deleterious effects,
out of the cardiac cell. This consumption of aTP for noncon- trimetazidine maintains the contractile function of the cardiac
tractile purposes further impairs cardiac function, in a con- cell and reduces anginal symptoms.12,20
text where energy levels are already dramatically reduced.
This vicious cycle has been summarized by Pepine et al in To summarize, trimetazidine exerts its anti-ischemic effects
the following terms: ischemia begets more ischemia.9 directly at the cardiac cell level, by optimizing aTP use and op-
posing deleterious changes that occur during ischemia.
Trimetazidine acts directly at the cell level by inhibiting long-
chain 3-ketoacyl coenzyme a thiolase, a mitochondrial en- Clinical efficacy of trimetazidine in stable angina
zyme involved in fatty acid oxidation. Inhibition of this enzyme The anti-ischemic and antianginal efficacy of trimetazidine has
leads to a decrease in fatty acid oxidation and, as a conse- been demonstrated in a range of randomized clinical trials in-
quence, stimulates glucose oxidation and inhibits glycolysis volving nearly 4000 patients with chronic stable angina world-
(Figure 1).10 This beneficial effect of trimetazidine on energy wide.20 This clinical efficacy is associated with a good safety
metabolism pathways is accompanied by a restoration of in- profile. The positive benefit-risk balance of trimetazidine in sta-
tracellular energy levels. This has been demonstrated in an- ble angina was recently reaffirmed in an assessment of the
european Medicines agency in 2012 and recog-
nized in the latest european Society of Cardiology
guidelines for the treatment of stable CaD in 2013,22
and for the treatment of heart failure in 2016.23

Efficacy of trimetazidine in comparison


with b-blockers
The antianginal efficacy of trimetazidine (60 mg dai-
ly) was compared with that of propranolol (120 mg
daily) in a randomized, double-blind, multicenter
study in 149 patients with stable angina and doc-
umented CaD.24 results indicated that the antiang-
inal efficacy of trimetazidine was similar to that of
the b-blocker.

The improvement in clinical parameters was com-


parable in both groups of patients after 3 months
of treatment (mean propranolol-trimetazidine dif-
ference in number of weekly angina attacks, 2.0
[P=0.117]; and in weekly use of short-acting ni-
trates, 1.1 [P=0.426]). a similar anti-ischemic ef-
fect for trimetazidine and propranolol was observed
during exercise testing.
Figure 1. The alteration in fatty acid oxidation during
cardiac ischemia is accompanied by an increase in
glycolysis and a decrease in glucose oxidation, with
deleterious consequences for cardiac homeostasis
and efficiency (black arrows). The inhibition of fatty acid
oxidation with trimetazidine restores the coupling of
glycolysis to glucose oxidation, which in turn increases
production of ATP and restores cell function (red arrows).
Modified after reference 8: Fillmore et al. Br J Pharmacol. 2014;
171:2080-2090. 2014, The British Pharmacological Society.

Trimetazidine: targeting the cardiac cell in stable angina Gliebova MEDICOGRAPHIA, Vol 38, No. 3, 2016 311
VA STA R E L M R

Efficacy of trimetazidine as a combination therapy


with b-blockers
The antianginal efficacy of trimetazidine as an add-on ther-
apy to b-blockers was evaluated in a series of randomized
clinical studies.27-29 In the TrIMPOL-II (TrIMetazidine in PO-
Land-II) study, a total of 426 stable angina patients with doc-
umented CaD were randomized to trimetazidine (60 mg daily)
or placebo, in addition to receiving a background treatment
of metoprolol (100 mg daily).30 exercise testing, evaluated at
baseline and after 3 months of treatment, revealed a significant
improvement in the trimetazidine-treated group compared
with the placebo group (increase in total exercise duration
of 16% and 10% for trimetazidine and placebo, respectively;
in total workload of 14% and 7%; in time to onset of angina
of 24% and 13%; and in time to 1-mm ST-segment depres-
sion of 29% and 22% [P<0.01 for all]). Clinical symptoms
(number of angina attacks, use of nitrate, and intensity of angi-
nal pain) were also significantly reduced with trimetazidine
plus metoprolol compared with placebo plus metoprolol.

In a separate randomized, double-blind study, the antiangi-


Figure 2. Mean number of weekly angina attacks in patients nal efficacy of trimetazidine in combination with propranolol
whose symptoms were inadequately controlled with propranolol
was evaluated versus isosorbide dinitrate plus propranolol.31
before (D0) and after (D60) add-on treatment with isosorbide
dinitrate (beige bar) or trimetazidine (red bar). Stable angina patients (n=53) for whom symptoms were not
adequately controlled by propranolol (120 mg daily) received
This study shows that trimetazidine possesses the clinical ef- add-on therapy with trimetazidine (60 mg daily) or isosorbide
ficacy of a first-line antianginal agent. The comparable effica- dinitrate (30 mg daily) for 2 months. Patients who received
cy of trimetazidine and b-blocker was acknowledged in the trimetazidine had a greater reduction in the weekly number of
2013 european Society of Cardiology guidelines on the man- angina attacks (Figure 2) and in consumption of short-act-
agement of stable CaD.22 ing nitrates compared with the reference group. Moreover,
exercise test results only improved in the group where trimeta-
Efficacy of trimetazidine in comparison with calcium zidine was used (increase in exercise duration of 14% ver-
channel blockers sus 2% for trimetazidine versus isosorbide dinitrate, respec-
The antianginal efficacy of trimetazidine (60 mg daily) was tively; and in time to 1-mm ST-segment depression of 15%
compared with that of nifedipine (40 mg daily) in a random- versus 3%). Overall, trimetazidine appeared to have better an-
ized, double-blind study with a cross-over design in 39 pa- tianginal efficacy compared with the long-acting nitrate isosor-
tients with stable angina and documented CaD.25 each treat- bide dinitrate.
ment lasted for 6 weeks, separated by a 1-week washout
period. Both treatments reduced the frequency of angina at- The use of trimetazidine in combination with a b-blocker pro-
tacks to a similar degree. The comparable anti-ischemic activ- vides better clinical efficacy than b-blocker alone or another
ity of the two drugs was also shown by a similar improvement combination strategy. This may be related to an additive, or
in exercise test parameters (ie, increase in total workload of possibly synergetic, effect of the two drugs, each of which
24% and 30% for trimetazidine and nifedipine, respectively; works via a distinct mechanism of action. The use of trimetazi-
in duration of exercise of 13% and 17%; and in time to 1-mm dine, which acts directly at the cardiac level, appears a ra-
ST-segment depression of 17% and 19% [with no significant tional choice for optimizing the treatment of patients inade-
difference between treatments]). quately controlled by b-blockers.

In a separate study using a parallel-arm design, trimetazidine Antianginal effectiveness of trimetazidine in


and the nondihydropyridine calcium channel blocker diltiazem different clinical situations
decreased the number of weekly angina attacks and nitrate In everyday practice
consumption to the same extent. Moreover, both drugs had The antianginal effectiveness of trimetazidine in clinical prac-
a similar effect profile with regards to exercise test parame- tice was evaluated in a large prospective cohort of 1213 out-
ters.26 These clinical data suggest that trimetazidine could be patients with stable angina inadequately controlled by stan-
an effective alternative to calcium channel blockers in the treat- dard therapy consisting of b-blocker alone or a combination of
ment of stable angina patients. b-blocker plus long-acting nitrate or calcium channel block-

312 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Trimetazidine: targeting the cardiac cell in stable angina Gliebova
VA STA R E L M R

er.32 all patients received trimetazidine (35 mg modified-re- decreased the number of angina episodes in clinical prac-
lease formulation, twice daily) on top of their standard treat- tice (from 2.8/week at baseline to 0.9/week after treatment
ment for 2 months. In all patients, trimetazidine significant- [P<0.001]). Short-acting nitrate consumption was also re-
ly reduced the number of weekly angina attacks, regardless duced (from 2.5/week at baseline to 0.7/week after treatment
of the background therapy (Figure 3). Moreover, in this [P<0.001]). Interestingly, in a separate study in the same pop-
study maximal antianginal efficacy was achieved with a dual ulation (diabetic patients with CaD), the addition of trimetazi-
combination of trimetazidine and b-blocker only. These data dine on top of standard treatment was able to reduce not only
suggest that combination therapy with b-blocker and symptomatic episodes of myocardial ischemia, but also silent
trimetazidine should be considered for the optimal manage- ischemia, a feature commonly observed in diabetic patients
ment of stable angina patients in real-life situations. and associated with a worse prognosis.35

Trimetazidine has also been demonstrated to have


benefits in challenging clinical situations, such as
in elderly diabetic patients with multivessel coro-
nary heart disease undergoing percutaneous coro-
nary intervention (PCI).36 In this population (n=700),
treatment with trimetazidine (60 mg daily) for two
years on top of conventional treatment significant-
ly reduced the incidence of angina, recurrent angi-
na, and silent ischemia compared with placebo.
Moreover, these beneficial effects of trimetazidine
were associated with a stabilization of cardiac func-
tion and structure.

Overall, these data suggest that the efficacy of tri-


metazidine in angina patients with diabetes goes
beyond the relief of angina symptoms by providing
additional benefits in terms of cardioprotection.
Figure 3. Mean number of weekly angina attacks in outpatients whose symp-
toms were inadequately controlled by standard therapy (b-blocker alone, In angina patients with left ventricular
b-blocker + long-acting nitrate, or b-blocker + calcium channel blocker) before dysfunction
(gray bars) and after (red bars) add-on treatment with trimetazidine for 2 a number of reports tend to indicate that trimetazi-
months. Data are presented in patients who had 7 angina attacks per week
at inclusion. dine could improve the cardiac function of patients
Modified after reference 32: Nesukay. Ukr J Cardiol. 2014;2:43-47. 2014, Ukrcardio.
with ischemic heart disease. In CaD patients with
left ventricular dysfunction, 2 years treatment with
In angina patients with diabetes trimetazidine (60 mg daily) on top of background therapy im-
angina patients often present with associated comorbidities proved both myocardial perfusion and contractile function
that make their therapeutic management particularly challeng- versus placebo, with single photon emission computerized to-
ing. Patients with concomitant diabetes mellitus and CaD mography imaging.37 Trimetazidine was shown to increase left
are considered to be at especially high cardiovascular risk.22 ventricular ejection fraction, reduce left ventricular volumes,
In fact, type 2 diabetes patients without previous myocardial and improve the quality of life of CaD patients with left ven-
infarction have a similar risk of myocardial infarction to that tricular dysfunction in various clinical settings, including in the
of nondiabetic patients with previous myocardial infarction.33 elderly38 and diabetic39 patients.
In diabetic patients, cardiac energy metabolism is significant-
ly altered, and these alterations are further amplified in the Interestingly, the beneficial effects of trimetazidine on cardiac
presence of underlying ischemic heart disease. Trimetazidine function observed in CaD may extend to heart failure, whether
appears an attractive tool for the management of angina pa- of ischemic origin or not. a recent meta-analysis of 17 ran-
tients with diabetes. This hypothesis is supported by findings domized clinical trials in heart failure that included 955 patients
from the DIeTrIC trial (estudio prospectivo en pacientes DIa- treated with trimetazidine or placebo suggests so.40 Treatment
bticos de la efectividad y tolerabilidad de la TrImetazidina with trimetazidine was associated with a significant improve-
en asociacin al tratamiento previo de su enfermedad Coro- ment in left ventricular ejection fraction in patients with is-
naria), a prospective, observational study that investigated chemic heart failure (weighted mean difference with placebo,
the antianginal effect of trimetazidine in 580 outpatients with +7.37%; 95% CI, 6.05 to 8.70; P<0.01) or nonischemic heart
type 2 diabetes and CaD.34 The addition of trimetazidine (60 failure (weighted mean difference, +8.72%; 95% CI, 5.51 to
mg/day) to conventional background therapy for 6 months 11.92; P<0.01). This was accompanied by a reduction in the

Trimetazidine: targeting the cardiac cell in stable angina Gliebova MEDICOGRAPHIA, Vol 38, No. 3, 2016 313
VA STA R E L M R

risks of all-cause mortality (relative risk [rr], 0.29; 95% CI, trimetazidine (60 mg administered as a single dose 30 minutes
0.17 to 0.49; P<0.00001) and of cardiovascular events and before intervention) or no treatment. Treatment with trimetazi-
hospitalization (rr, 0.42; 95% CI, 0.30 to 0.58; P<0.00001). dine prior to PCI limited periprocedural myocardial injury, as
These findings are further supported by a retrospective analy- shown by the significant reduction in cardiac troponin Ic lev-
sis of a cohort of 669 patients with heart failure, in which the el up to 24 hours post-PCI in these patients compared with
use of trimetazidine was associated with improved survival patients who received no treatment. Thus, the direct effect of
from all-cause death and cardiovascular death.41 trimetazidine on the cardiac cell may provide cardioprotection
immediately after PCI.
The addition of trimetazidine to conventional medical therapy
is likely to improve cardiac function and prognosis in patients
with left ventricular dysfunction and heart failure. Importantly,
trimetazidine has a neutral effect at the hemodynamic level
and can thus be safely used in these types of patient. additon-
ally, trimetazidine has been recognized as an effective anti-angi-
nal treatment that is safe to use in heart failure and has been
granted with class IIb of recommendation and level a of evi-
dence in the latest eSC guidelines for heart failure treatment.23

In angina patients with a history of


myocardial infarction
Trimetazidine may also provide benefits in terms of outcomes
in patients after myocardial infarction. The MeTrO study (Man-
agement of angina: a reTrospective cOhort) included 353
patients with stable angina treated with at least one antiang-
inal drug (nitrates, b-blockers, calcium channels blockers,
trimetazidine, or nicorandil) who had previously been hospi-
talized for myocardial infarction.42 Of all the antianginal strate-
gies, only the one that included trimetazidine was associat-
ed with a significant reduction in all-cause mortality over 6 Figure 4. Incidences of stent restenosis and major adverse car-
months following hospital discharge for myocardial infarc- diac and cerebrovascular events (MACCEs)including all-cause
tion (odds ratio [Or], 0.36; 95% CI, 0.15 to 0.86; P=0.022). mortality, nonfatal myocardial infarction, revascularization, stroke,
and cerebral bleedingin the control (gray bars; n=323) and
trimetazidine (red bars; n=312) groups 12 months after percuta-
Similarly, a retrospective analysis of a Korean registry that neous coronary intervention.
included 13 733 patients hospitalized for myocardial infarc-
tion showed that treatment with trimetazidine during the in- a recent report investigated the effect of trimetazidine on the
hospital period was independently associated with a signif- incidence of stent restenosis and major adverse cardiac and
icant reduction in all-cause mortality (hazard ratio [Hr], 0.41; cerebrovascular events (MaCCes) in patients undergoing PCI.46
95% CI, 0.18 to 0. 97; P=0.042) and major adverse cardiac a total of 786 patients were randomized after implantation
events (Hr, 0.24; 95% CI, 0.10 to 0.56; P=0.001) over 12 of a drug-eluting stent to trimetazidine (60 mg daily) on top
months.43 of standard treatment for at least 30 days or standard treat-
ment only, ie, no additional therapy. at 1-year follow-up, pa-
These data suggest that trimetazidine, in addition to its clin- tients in the trimetazidine group had a significantly lower in-
ical antianginal efficacy, may offer long-term cardioprotection cidence of stent restenosis and MaCCes compared with
by improving survival in patients after acute myocardial in- those in the control group who received standard treatment
farction. only (Figure 4). Interestingly, in a rat model of diabetes, trimeta-
zidine decreased the proliferation of vascular smooth muscle
In angina patients with percutaneous coronary cells and promoted re-endothelialization after injury of the
intervention carotid artery.47 Both mechanisms could have contributed
although it is a minimally invasive procedure, PCI leads to my- to the lower rate of stent restenosis observed with trimetazi-
ocardial injury in about one third of patients. a rise in troponin dine after PCI.
level immediately following PCI, a marker for myocardial in-
jury, is correlated with a worse prognosis in these patients.44 Overall, these findings suggest that beyond its antianginal
The effect of trimetazidine has been investigated in this spe- effect, trimetazidine may improve prognosis in patients under-
cific setting in a randomized study involving 266 angina pa- going revascularization procedures. This hypothesis is cur-
tients undergoing elective PCI.45 Patients were randomized to rently being tested in the aTPCI (The efficacy and safety of

314 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Trimetazidine: targeting the cardiac cell in stable angina Gliebova
VA STA R E L M R

Trimetazidine in Patients with angina pectoris having been that directly targets the cardiac cell, trimetazidine provides ro-
treated by percutaneous Coronary Intervention) study, a large bust antianginal efficacy, reduces the burden of myocardial is-
morbidity-mortality trial in angina patients with a post-PCI fol- chemia, and improves exercise capacity in a variety of angina
low-up of 2 to 4 years. patients, including those with a history of myocardial infarc-
tion, previous PCI, diabetes, or left ventricular dysfunction.
Conclusion Moreover, data from clinical trials suggest that trimetazidine
Trimetazidine is an antianginal treatment with well-established may also offer long-term cardioprotection in a broad range
efficacy, whether as monotherapy or part of combination ther- of cardiovascular patients.
apy (for patients inadequately controlled by hemodynami- Acknowledgments: The author would like to thank Julie Salzmann, PhD,
cally active therapy alone). Based on its mechanism of action for providing medical writing assistance.

References
1. Pursnani S, Korley F, Gopaul r, et al. Percutaneous coronary intervention ver- 20. Saeedi r, Grist M, Wambolt rB, Bescond-Jacquet a, Lucien a, allard MF.
sus optimal medical therapy in stable coronary artery disease: a systematic re- Trimetazidine normalizes postischemic function of hypertrophied rat hearts.
view and meta-analysis of randomized clinical trials. Circ Cardiovasc Interv. J Pharmacol Exp Ther. 2005;314:446-454.
2012; 5:476-490. 21. Detry JM. Clinical features of an anti-anginal drug in angina pectoris. Eur Heart J.
2. Husted Se, Ohman eM. Pharmacological and emerging therapies in the treat- 1993;14(suppl G):18-24.
ment of chronic angina. Lancet. 2015;386:691-701. 22. Montalescot G, Sechtem U, achenbach S, et al. 2013 eSC guidelines on the
3. Westermann D, Savvatis K, Wollenberg U, Limberg r, Maier LS, Bauersachs management of stable coronary artery disease: The Task Force on the man-
J. Prevalence of obstructive coronary artery disease in ambulatory patients agement of stable coronary artery disease of the european Society of Cardiol-
with stable angina pectoris. J Clin Exp Cardiolog. 2015;6:387.doi:10.4172/ ogy. Eur Heart J. 2013;34:2949-3003.
2155-9880.1000387- 23. Ponikowski P, Voors aa, anker SD, et al; authors/Task Force Members. 2016
4. Lee BK, Lim HS, Fearon WF, et al. Invasive evaluation of patients with angina eSC Guidelines for the diagnosis and treatment of acute and chronic heart fail-
in the absence of obstructive coronary artery disease. Circulation. 2015;131: ure: The Task Force for the diagnosis and treatment of acute and chronic heart
1054-1060. failure of the european Society of Cardiology. Eur Heart J. 2016;37(27):2129-
5. Marzilli M, Merz Cn, Boden We, et al. Obstructive coronary atherosclerosis and 2200.
ischemic heart disease: an elusive link! J Am Coll Cardiol. 2012;60:951-956. 24. Detry JM, Sellier P, Pennaforte S, Cokkinos D, Dargie H, Mathes P. Trimetazi-
6. Pepine CJ, Douglas PS. rethinking stable ischemic heart disease is this the dine: a new concept in the treatment of angina. Comparison with propranolol
beginning of a new era? J Am Coll Cardiol. 2012;60(11):957-959. in patients with stable angina. Br J Clin Pharmacol. 1994;37:279-288.
7. Daly Ca, Clemens F, Sendon JL, et al. The initial management of stable angina 25. Dalla-Volta S, Maraglino G, Della-Valentina P, Viena P, Desideri a. Comparison
in europe, from the euro Heart Survey: a description of pharmacological man- of trimetazidine with nifedipine in effort angina: a double-blind, crossover study.
agement and revascularization strategies initiated within the first month of pres- Cardiovasc Drugs Ther. 1990;4(suppl 4):853-859.
entation to a cardiologist in the euro Heart Survey of Stable angina. Eur Heart J. 26. Koylan n, Bilge aK, adalet K, Mercanoglu F, Buyukozturk K. Comparison of the
2005;26:1011-1022. effects of trimetazidine and diltiazem on exercise performance in patients with
8. Fillmore n, Mori J, Lopaschuk GD. Mitochondrial fatty acid oxidation alterations coronary heart disease. The Turkish trimetazidine study (TTS). Acta Cardiol.
in heart failure, ischaemic heart disease and diabetic cardiomyopathy. Br J Phar- 2004; 59:644-650.
macol. 2014;171:2080-2090. 27. Sellier P, Broustet JP. assessment of anti-ischemic and antianginal effect at
9. Pepine CJ, nichols WW. The pathophysiology of chronic ischemic heart dis- trough plasma concentration and safety of trimetazidine Mr 35 mg in patients
ease. Clin Cardiol. 2007;30:I4-I9. with stable angina pectoris. a multicenter, double-blind, placebo-controlled
10. Kantor PF, Lucien a, Kozak r, Lopaschuk GD. The antianginal drug trimetazi- study. Am J Cardiovasc Drugs. 2003;3:361-369.
dine shifts cardiac energy metabolism from fatty acid oxidation to glucose ox- 28. Vitale C, Spoletini I, Malorni W, Perrone-Filardi P, Volterrani M, rosano GM. effi-
idation by inhibiting mitochondrial long-chain 3-ketoacyl coenzyme a thiolase. cacy of trimetazidine on functional capacity in symptomatic patients with stable
Circ Res. 2000;86:580-588. exertional anginathe VaSCO-angina study. Int J Cardiol. 2013;168:1078-1081.
11. rossi a, Lavanchy n, Martin J. antiischemic effects of trimetazidine: 31P-nMr 29. Deng B, Huang SZ, Liu M, Li T, Lu Y. Combination therapy of trimetazidine
spectroscopy study in the isolated rat heart. Cardiovasc Drugs Ther. 1990;4 with metoprolol in patients with stable angina pectoris. Chin J New Drugs Clin
(suppl 4):812-813. Rem. 2002;21:197-199.
12. el Banani H, Bernard M, Baetz D, et al. Changes in intracellular sodium and pH 30. Szwed H, Sadowski Z, elikowski W, et al. Combination treatment in stable ef-
during ischaemia-reperfusion are attenuated by trimetazidine. Comparison be- fort angina using trimetazidine and metoprolol. results of a randomized, dou-
tween low- and zero-flow ischaemia. Cardiovasc Res. 2000;47:688-696. ble-blind, multicentre study (TrIMPOL II). Eur Heart J. 2001;22:2267-2274.
13. Inci I, Dutly a, rousson V, Boehler a, Weder W. Trimetazidine protects the ener- 31. Michaelides aP, Spiropoulos K, Dimopouloss K, athanasiades D, Toutouzas P.
gy status after ischemia and reduces reperfusion injury in a rat single lung trans- antianginal efficacy of the combination of trimetazidine-propranolol compared
plant model. J Thorac Cardiovasc Surg. 2001;122:1155-1161. with isosorbide dinitrate-propranolol in patients with stable angina. Clin Drug
14. Fragasso G, Perseghin G, De CF, et al. effects of metabolic modulation by Invest. 1997;13:8-14.
trimetazidine on left ventricular function and phosphocreatine/adenosine tri- 32. nesukay eG. Treatment of stable angina in Ukraine: CLaSSICa study. Ukr J
phosphate ratio in patients with heart failure. Eur Heart J. 2006;27:942-948. Cardiol. 2014;2:43-47.
15. Lagadic-Gossmann D, Le Prigent K, Feuvray D. effects of trimetazidine on pHi 33. Haffner SM, Lehto S, ronnemaa T, Pyorala K, Laakso M. Mortality from coro-
regulation in the rat isolated ventricular myocyte. Br J Pharmacol. 1996;117: nary heart disease in subjects with type 2 diabetes and in nondiabetic subjects
831-838. with and without prior myocardial infarction. N Engl J Med.1998;339:229-234.
16. renaud JF. Internal pH, na+, and Ca2+ regulation by trimetazidine during car- 34. rodriguez PL, Maicas BC, Velazquez MM, Gil PB. [a prospective study on tri-
diac cell acidosis. Cardiovasc Drugs Ther. 1988;1:677-686. metazidine effectiveness and tolerability in diabetic patients in association to
17. Wei J, Xu H, Shi L, Tong J, Zhang J. Trimetazidine protects cardiomyocytes the previous treatment of their coronary disease. DIeTrIC study]. Rev Clin Esp.
against hypoxia-induced injury through ameliorates calcium homeostasis. Chem 2005;205:57-62.
Biol Interact. 2015;236:47-56. 35. Marazzi G, Wajngarten M, Vitale C, et al. effect of free fatty acid inhibition on
18. Guarnieri C, Muscari C. effect of trimetazidine on mitochondrial function and silent and symptomatic myocardial ischemia in diabetic patients with coronary
oxidative damage during reperfusion of ischemic hypertrophied rat myocardi- artery disease. Int J Cardiol. 2007;120:79-84.
um. Pharmacology. 1993;46:324-331. 36. Xu X, Zhang W, Zhou Y, et al. effect of trimetazidine on recurrent angina pectoris
19. ruixing Y, Wenwu L, al-Ghazali r. Trimetazidine inhibits cardiomyocyte apop- and left ventricular structure in elderly multivessel coronary heart disease pa-
tosis in a rabbit model of ischemia-reperfusion. Transl Res. 2007;149:152-160. tients with diabetes mellitus after drug-eluting stent implantation: a single-cen-

Trimetazidine: targeting the cardiac cell in stable angina Gliebova MEDICOGRAPHIA, Vol 38, No. 3, 2016 315
VA STA R E L M R

tre, prospective, randomized, double-blind study at 2-year follow-up. Clin Drug 42. Iyengar SS, rosano GM. effect of antianginal drugs in stable angina on pre-
Investig. 2014;34:251-258. dicted mortality risk after surviving a myocardial infarction: a preliminary study
37. el-Kady T, el-Sabban K, Gabaly M, Sabry a, abdel-Hady S. effects of trimetazi- (MeTrO). Am J Cardiovasc Drugs. 2009;9:293-297.
dine on myocardial perfusion and the contractile response of chronically dys- 43. Kim JS, Kim CH, Chun KJ, et al. effects of trimetazidine in patients with acute
functional myocardium in ischemic cardiomyopathy: a 24-month study. Am J myocardial infarction: data from the Korean acute Myocardial Infarction registry.
Cardiovasc Drugs. 2005;5:271-278. Clin Res Cardiol. 2013;102:915-922.
38. Vitale C, Wajngaten M, Sposato B, et al. Trimetazidine improves left ventricu- 44. Babu GG, Walker JM, Yellon DM, Hausenloy DJ. Peri-procedural myocardial
lar function and quality of life in elderly patients with coronary artery disease. Eur injury during percutaneous coronary intervention: an important target for car-
Heart J. 2004;25:1814-1821. dioprotection. Eur Heart J. 2011;32:23-31.
39. Belardinelli r, Cianci G, Gigli M, Mazzanti M, Lacalaprice F. effects of trimetazi- 45. Bonello L, Sbragia P, amabile n, et al. Protective effect of an acute oral load-
dine on myocardial perfusion and left ventricular systolic function in type 2 di- ing dose of trimetazidine on myocardial injury following percutaneous coro-
abetic patients with ischemic cardiomyopathy. J Cardiovasc Pharmacol. 2008; nary intervention. Heart. 2007;93:703-707.
51:611-615. 46. Chen J, Zhou S, Jin J, et al. Chronic treatment with trimetazidine after discharge
40. Gao D, ning n, niu X, Hao G, Meng Z. Trimetazidine: a meta-analysis of ran- reduces the incidence of restenosis in patients who received coronary stent
domised controlled trials in heart failure. Heart. 2011;97:278-286. implantation: a 1-year prospective follow-up study. Int J Cardiol. 2014;174:
41. Fragasso G, rosano G, Baek SH, et al. effect of partial fatty acid oxidation in- 634-639.
hibition with trimetazidine on mortality and morbidity in heart failure: results from 47. Yoon JW, Cho BJ, Park HS, et al. Differential effects of trimetazidine on vascular
an international multicentre retrospective cohort study. Int J Cardiol. 2013;163: smooth muscle cell and endothelial cell in response to carotid artery balloon
320-325. injury in diabetic rats. Int J Cardiol. 2013;167:126-133.

Keywords: antianginal efficacy; cardiac cell; cardiac energy metabolism; cardioprotection; myocardial ischemia; stable angina
pectoris; trimetazidine

CIBLER LA CELLULE CARDIAQUE DANS LANGOR STABLE GRCE LA TRIMTAZIDINE


Langor est maintenant reconnu comme une maladie multiforme o lobstruction coronaire ne constitue quun fac-
teur de risque parmi dautres. La cellule cardiaque, dont le rle est central dans un grand nombre de processus is-
chmiques, est reconnue comme lun des acteurs principaux de lischmie cardiaque. Au niveau cellulaire, lisch-
mie myocardique se caractrise par un mtabolisme nergtique myocardique modifi, rsultant en un dsquilibre
ionique, un msusage et un dficit en nergie et aboutit finalement un dysfonctionnement. Pendant lischmie,
la trimtazidine agit directement au niveau cellulaire en diminuant loxydation des acides gras et en augmentant loxy-
dation du glucose, ce qui contribue rtablir un mtabolisme nergtique normal. Ce mcanisme conduit une am-
lioration globale du fonctionnement gnral de la cellule et exerce une action anti-ischmique au niveau des organes.
En ciblant directement la cellule cardiaque, le mcanisme daction de la trimtazidine complte celui des traitements
antiangoreux hmodynamiques, optimisant ainsi lefficacit clinique lorsquelle est associe dautres traitements.
La trimtazidine diminue les symptmes et augmente laptitude leffort des patients angoreux dont les symptmes
sont mal contrls par les b-bloquants et/ou les antagonistes calciques. Leffet bnfique de la trimtazidine a t d-
montr chez de nombreux patients angoreux, dont ceux ayant des antcdents dinfarctus du myocarde et ceux
qui ont eu une intervention coronaire percutane, un diabte ou une dysfonction ventriculaire gauche. En plus de
ses effets antiangoreux prouvs, la trimtazidine peut aussi apporter dautres effets bnfiques en termes de car-
dioprotection et de pronostic chez de nombreux patients risque cardiovasculaire.

316 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Trimetazidine: targeting the cardiac cell in stable angina Gliebova

INTERVIEW

ATPCI (efficAcy and safety of


Trimetazidine in Patients with angi-
na pectoris having been treated
by percutaneous Coronary Inter-
ventions [PCI]) will improve our
The ATPCI trial: a new
knowledge about coronary artery
disease and its treatment by PCI. international phase 3 study
It will also identify the role of meta-
bolic therapy in this patient set-
ting. It is the first study to test the assessing the clinical impact of
value of increasing the energy sta-
tus of the ischemic myocyte with
trimetazidine in terms of hard end
adding a metabolic agent to
points such as cardiac death and
hospitalization. post-PCI angina treatment
I n t e r v i e w w i t h R . Fe r ra r i , I t a l y

Can you tell us what ATPCI stands for?

TPCI is the short name of an international, multicenter, randomized, double-

A blind, placebo-controlled study in patients treated with trimetazidine for 2


to 4 years. The full title of the trial is efficAcy and safety of Trimetazidine in
Patients with angina pectoris having been treated by percutaneous Coronary Inter-
ventions. The sponsor of the study is the Institut de Recherches Internationales
Servier (I.R.I.S.) and it is registered with the World Health Organization (WHO) under
the universal trial number (UTN) UIIII-1145-1743.

Roberto FERRARI, MD The acronym ATPCI is particularly appropriate for the study as it targets patients
Department of Cardiology with ischemic heart disease who have received a percutaneous coronary interven-
and LTTA Centre
University Hospital of Ferrara tion (PCI) and pharmacological treatment by trimetazidinethe active drug under
ITALY testing, a piperazine derivative with anti-ischemic properties that are different from
any other anti-ischemic drugsa drug that increases the adenosine triphosphate
(ATP) levels. The standard approach to treat coronary artery disease (CAD) is based
on increasing oxygen supply to the myocardium and/or reducing oxygen consump-
tion by means of hemodynamic effects, acting via the heart rate in the case of iva-
bradine, or via heart rate and myocardial contractility in the case of b-blockers and
verapamil or diltiazem, and via the peripheral arterial coronary and/or venous re-
sistance in the case of dihydropyridine calcium-channel blockers and nitrates. An
alternative approach is to improve the efficiency and the energy of the heart for a
given supply of oxygen, which is reduced during ischemia. In other words, such an
approach aims to increase the level of ATP. This metabolic approach is interesting
considering that trimetazidine does not have hemodynamic effects, but acts as a
modulator of cardiac metabolism.

Under physiological conditions, the energy demand of the heart is met by the me-
tabolism of two main substratesglucose and free fatty acids (FFAs). Glucose
metabolism occurs by means of glycolysis and oxidation. Whereas the oxidative
Address for correspondence: pathway converts pyruvate into acetyl-coenzyme A (CoA), which is incorporated
Roberto Ferrari, MD, Chair of in the Krebs cycle and produces 36 molecules of ATP, the conversion of glucose to
Cardiology, Azienda Ospedaliero-
Universitaria di Ferrara, Ospedale di
pyruvate during glycolysis produces only two molecules of ATP, but this ATP is im-
Cona, Via Aldo Moro 8, 44124 portant as oxygen is not required in the process.1 The FFAs are the main sources
(Cona) Ferrara, Italy of ATP, corresponding to about 70% of the myocardial production, but the oxida-
(email: fri@unife.it)
tion of FFAs requires about 10% more oxygen than that of glucose to produce an
Medicographia. 2016;38:317-319 equivalent amount of energy.1 During ischemia and/or angina, all oxidative process-
www.medicographia.com es are depressed, leading to acetyl-coA accumulation in the mitochondria with a

Adding a metabolic agent to post-PCI angina treatment: the ATPCI trial Ferrari MEDICOGRAPHIA, Vol 38, No. 3, 2016 317
INTERVIEW

block of FFA b-oxidation and of the activity of pyruvate de- The second consideration relies on the unique anti-ischemic
hydrogenase, the enzyme that allows the entry of pyruvate and antianginal effects of trimetazidine, which could be ide-
into the mitochondria. Pyruvate is then converted into lactic al after PCI. The antianginal effect of trimetazidine in stable
acid, which is released from the myocytes into the extracel- angina pectoris is well-known and documented by 25 stud-
lular space, thus reducing the pH and causing pain by irrita- ies, including more than 4000 patients, in which trimetazi-
tion of the myocardial nerve fibers. Thus, in ischemia, glycol- dine was compared with placebo or active comparator. Ef-
ysis, contrary to oxidation, continues in an anaerobic fashion, ficacy has always been assessed on ergometric criteria and
having as final product lactate instead of pyruvate, and be- symptoms.
comes the only and most important source of anaerobic ATP.1
Trimetazidine further inhibits FFA b-oxidation and, indirectly, In addition, a first Cochrane meta-analysis conducted in 2005,
stimulates the activity of pyruvate dehydrogenase, thus di- involving a total of 1378 patients, reported a statistically sig-
recting pyruvate into the mitochondria, avoiding lactic acid nificant and clinically relevant efficacy of trimetazidine in the
formation, and allowing anaerobic glycolysis to continue. This treatment of angina, either alone or in combination with con-
has two important consequencesreduction in intra- and ventional antianginal treatments.4 In 2012, a second Cochrane
extracellular acidosis, thus preventing pain, and improvement meta-analysis, including 2283 patients and two additional
in anaerobic ATP production through glycolysis, ensuring ergometric criteriatotal exercise duration and time to onset
maintenance of cellular homeostasis and viability.2 As ancil- of anginaconfirmed the conclusion of the first meta-analy-
lary properties resulting from the maintenance of ATP, trimetazi- sis (unpublished data). A third network meta-analysis was
dine has also been shown to reduce oxygen free radical pro- then conducted in 2011, including studies with positive re-
duction and oxidative stress and to inhibit the opening of sults as well as studies with inconclusive results, as required
mitochondrial calcium pores with the consequent release of by European guidelines. The conclusion was that trimetazi-
cytochrome C, thus reducing apoptosis and attenuating in- dine is as efficacious as other antianginal agents despite the
flammation.2 different mechanism of action.5

The acronym ATPCI is well-suited to this study, which aims The third consideration is related to the increasing evidence
to test whether treatment with trimetazidine to maintain ATP of microvascular dysfunction as a cause of angina. Trimetazi-
levels after PCI is useful to reduce symptoms and improve dine, by improving energy metabolism, could be particularly
outcomes in patients with ischemic heart diseases. useful in this setting, where classic agents, which mainly act
on the epicardial coronary artery, are less likely to be active.
What is the rationale of the study and what is the
main objective of ATPCI? A previous proof-of-concept study showed a benefit of tri-
metazidine over placebo in reducing the incidence of recur-
TPCI aims to assess the long-term efficacy and safe- rent angina in CAD patients after drug-eluting stent implan-

A ty of trimetazidine versus placebo, when given in ad-


dition to other evidence-based cardiovascular ther-
apies in patients having had a recent PCI. The rationale for
tation.6 Another study showed a reduction in major adverse
cardiovascular events after 1-year follow-up in patients who
received trimetazidine for at least 1 month after PCI.7 A recent
ATPCI is based on three considerations. meta-analysis evaluates the effects of trimetazidine on pa-
tients undergoing PCI in a total of 778 patients.8 The addition-
The first is that, despite the wide use of PCI for patients with al use of trimetazidine, before intervention, reduced angina
chronic or acute angina, it is not clear whether this interven- attacks and electrocardiographic changes during PCI and
tion results in an improvement in outcomes when compared cardiac troponin level. It also improved the left ventricular ejec-
with medical therapy. In addition, several studies have shown tion fraction.8
that, after PCI, not all patients are angina free or stop the an-
tianginal treatment.3 What is the study design and who are the target
patients?
SELECTED ABBREVIATIONS AND ACRONYMS
TPCI is a phase 3, international, multicenter, double-
ATP
ATPCI
adenosine triphosphate
efficAcy and safety of trimetazidine in Patients with
angina pectoris having been treated by percuta-
A blind, placebo-controlled study randomized in two
parallel and balanced armson top of post-PCI rec-
ommended treatment for CAD, patients receive either trimeta-
neous Coronary Interventions zidine 35 mg twice daily or placebofor both secondary
CAD coronary artery disease prevention and regular antianginal therapies, as per current
FFA free fatty acid guidelines. Randomization at inclusion is stratified according
PCI percutaneous coronary intervention to country and nature of PCI procedure, whether elective or
urgent after an acute presentation but without STEMI.

318 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Adding a metabolic agent to post-PCI angina treatment: the ATPCI trial Ferrari
INTERVIEW

A total of 5800 patients from 27 countries will be included or increasing the dose of one of the evidence-based antiang-
within 30 days of PCI and followed-up during a 2- to 4-year inal therapies; and (iv) recurrent or persistent angina, lead-
treatment period with a maximum of 10 visits. ATPCI includes ing to performance of a coronary angiography, all of which will
patients (women or men 21 years old and <85 years old be reviewed by an independent adjudication committee. The
of any ethnic origin) presenting with a single or multivessel primary safety end point is the incidence of serious emer-
CAD and having undergone PCI for at least one stenosis to gency adverse events (in all visits) with trimetazidine as com-
either a native coronary artery or a coronary graft where the pared with placebo.
PCI fell into any of the following categories: (i) indicated be-
cause of angina pectoris occurring either in the context of The secondary end points are the time to first occurrence of
stable angina (elective PCI) or in the context of an acute pres- each of the separate four components of the primary end
entation, such as unstable angina/nonST-segment elevation point, with the addition of evidence of ischemia (documented
myocardial infarction (NSTEMI), but excluding ST-segment by stress imaging and leading to adding, switching, or in-
elevation myocardial infarction (STEMI); (ii) achieved by stent creasing the dose of one of the evidence-based antianginal
implantation or by other acceptable interventional (nonsur- therapies). Other efficacy end points include the following:
gical) means; (iii) successful as planned by the operator and CCS class of angina symptoms, number of angina episodes
with no further revascularization (either percutaneous or sur- per week, number of doses of short-acting nitrates taken per
gical) planned; or (iv) uncomplicated, such that the patients week, number of antianginal drugs taken by the patient, Seat-
discharge was not delayed because of a cardiac or cerebro- tle Angina Questionnaire scores (in countries where a validat-
vascular problem. ed translation is available), EQ-5D-3L Questionnaire scores,
and level of cardiac troponin (before each repeat PCI and be-
A measurement of left ventricular ejection fraction (LVEF) needs tween 6 and 24 hours after).
to be performed within 3 months before inclusion for patients
having undergone elective PCI and between PCI and inclu- What might we expect from the study?
sion for patients having undergone index PCI performed in
the context of an acute syndrome. Patients can be select- TPCI will improve our knowledge of CAD and its treat-
ed after PCI regardless of whether they are asymptomatic
or symptomatic with regard to angina, and regardless of their
Canadian Cardiovascular Society (CCS) class.
A ment by PCI. It will also identify the role of metabolic
therapy in this setting of patients. It is the first study to
test the value of increasing the energy status of the ischemic
myocyte with trimetazidine in terms of hard end points such
What is the primary end point of the study? as cardiac death and hospitalization. This is particularly rele-
vant considering that a recent study with ranolazine, another
he primary efficacy end point of ATPCI is the time to piperazine derivative, in a similar patient setting failed to show

T first occurrence of an event in the composite of: (i) car-


diac death; (ii) hospitalization for a cardiac event; (iii)
recurrent or persistent angina leading to adding, switching,
a benefit.9
Acknowledgments. This work was supported by a grant from Fon-
dazione Anna Maria Sechi per il Cuore (FASC), Italy.

Keywords: angina; cardiac metabolism; percutaneous coronary intervention; trimetazidine

References
1. Ferrari R, Williams AJ. The role of mitochondria in myocardial damage occur- patients with diabetes mellitus after drug-eluting stent implantation: a single-
ring on post-ischaemic reperfusion. J Appl Cardiol. 1986;1:501-519. centre, prospective, randomized, double-blind study at 2-year follow-up. Clin
2. Cargnoni A, Pasini E, Ceconi C, Curello S, Ferrari R. Insight into cytoprotection Drug Investig. 2014;34:251-258.
with metabolic agents. Eur Heart J. 1999;1(suppl O):O40-O48. 7. Chen J, Zhou S, Jin J, et al. Chronic treatment with trimetazidine after discharge
3. Babu GG, Walker J M, Yellon DM, Hausenloy DJ. Peri-procedural myocardial reduces the incidence of restenosis in patients who received coronary stent
injury during percutaneous coronary intervention: an important target for car- implantation: a 1-year prospective follow-up study. Int J Cardiol. 2014;174:634-
dioprotection. Eur Heart J. 2011;32:23-32. 639.
4. Ciapponi A, Pizzarro R, Harrison J. Trimetazidine for stable angina. Cochrane 8. Zhang Y, Xiao-juan M, Da-zhuo S. Effect of trimetazidine in patients undergoing
Database Syst Rev. 2005;(4):CD003614. percutaneous coronary intervention: a meta-analysis. PLoS One. 2015;10(9):
5. Danchin N, Marzilli M, Parkhomenko A, Ribeiro JP. Efficacy comparison of tri- e0137775.
metazidine with therapeutic alternatives in stable angina pectoris: a network 9. Weisz G, Gnreux P, Iiguez A; RIVER-PCI investigators. Ranolazine in patients
meta-analysis. Cardiology. 2011;120:59-72. with incomplete revascularization after percutaneous coronary intervention
6. Xiaohan X, Zhang W, Zhou Y, et al. Effect of trimetazidine on recurrent angina (RIVER-PCI): a multicentre, randomised, double-blind, placebo-controlled trial.
pectoris and left ventricular structure in elderly multivessel coronary heart disease Lancet. 2016;387(10014):136-145.

Adding a metabolic agent to post-PCI angina treatment: the ATPCI trial Ferrari MEDICOGRAPHIA, Vol 38, No. 3, 2016 319

FOCUS

Given thepathophysiology
of ischemic heart disease and the
difficulties encountered when
trying to control the total ischemic
burden with classic hemodynam-
ically active drugs, an adjunctive
Combining hemodynamic
therapeutic option that pharmaco-
logically manipulates cardiac en-
ergy metabolism seems reason-
and metabolic agents
able. This approach is based on
stimulating myocardial glucose ox-
idation to optimize cardiac energy
in ischemic heart disease
metabolism, and is proven to im-
prove cardiac function and protect
myocardial tissue against ische-
mia-reperfusion injury.

by G. Fragasso, Italy

M
yocardial ischemia can be looked at as a metabolic problem, as it
leads to an imbalance in the pathways the normal heart relies on
for energy production. Use of pharmacological agents to optimize
cardiac energy metabolism by stimulating myocardial glucose oxidation can
be an effective therapeutic option. The metabolic agent trimetazidine does
this indirectly by inhibiting fatty acid b-oxidation, in effect changing the en-
ergy substrate preference, promoting a shift from fatty acid metabolism to-
ward glucose metabolism, which is more efficient for ATP production. The ef-
ficacy of trimetazidine in the treatment of angina pectoris has been evaluated
under various conditions: trimetazidine administered as a monotherapy or in
Gabriele FRAGASSO, MD combination, acutely or over a longer-term period, as initial treatment, and in
Heart Failure Unit patients resistant to b-blockers or calcium-channel antagonists. All published
Istituto Scientifico San Raffaele
Milano, ITALY studies employing trimetazidine in patients with chronic ischemic heart dis-
ease have invariably reported beneficial clinical effects without adverse he-
modynamic events. In fact, in chronic ischemic heart disease patients with left
ventricular dysfunction, trimetazidine has been shown to be a particularly ef-
fective adjunctive treatment in terms of improvement in left ventricular me-
tabolism and function. An ongoing randomized clinical study in patients with
revascularized coronary artery disease should clarify whether the reported
experimental and clinical benefits of trimetazidine also translate into improved
prognosis.
Medicographia. 2016;38:320-327 (see French abstract on page 327)

schemic heart disease is a major cause of morbidity and mortality worldwide. In

I patients with acute coronary syndromes, revascularization interventions have been


shown to reduce myocardial infarction and death1,2; however, this is not the case
in patients with chronic stable angina. In fact, according to the European Society
of Cardiology (ESC) and the European Association for Cardio Thoracic Surgery
(EACTS) 2014 guidelines,3 percutaneous coronary intervention (PCI) or coronary ar-
tery bypass grafting (CABG) should be reserved for patients with refractory angina
and for patients in whom such procedures would be expected to provide a survival
benefit; this would be influenced by a number of factors, such as the number of
Address for correspondence: diseased vessels, lesion location and severity, and the presence of left ventricular
Gabriele Fragasso, Heart Failure Unit,
Istituto Scientifico San Raffaele,
dysfunction. Thus, all patients with stable ischemic heart disease, whether they are
Via Olgettina 60, 20132 Milano, Italy asymptomatic or experiencing severe symptoms, should first receive optimal med-
(email: gabriele.fragasso@hsr.it) ical therapy, which would usually be maintained even after revascularization.
www.medicographia.com

320 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Combining hemodynamic and metabolic agents in ischemic heart disease Fragasso
FOCUS

Current anti-ischemic/antianginal therapy focuses on two ma- infarction or heart failure, in which they consistently decreased
jor actions. The first pertains to vascular protection, in this case morbidity and mortality. However, several reports have dis-
aiming to delay progression of atherosclerosis (by use of sta- cussed potential pitfalls in their use in patients with stable coro-
tins, antithrombotic drugs), which would reduce future car- nary artery disease (CAD). In the REACH registry (REduction
diovascular events and death and improve quantity of life. The of Atherothrombosis for Continued Health), b-blockers were
second pertains to improvement in the imbalance between not associated with a lower risk of composite cardiovascular
myocardial oxygen supply and demand (ischemic imbalance), events, and they were associated with higher rates for the sec-
which would reduce the severity and frequency of angina ondary outcome (comprising primary outcome and hospital-
symptoms and also contribute to improvement in quality of ization for atherothrombotic events or a revascularization pro-
life. However, this therapeutic view does not consider the car- cedure) in chronic CAD patients.8 A recent post-hoc analysis
diac metabolic consequences of myocardial ischemia. In fact, of the CHARISMA study (Clopidogrel for High Atherothrom-
ischemia can be thought of as a metabolic problem (previ- botic Risk and Ischemic Stabilization Management and Avoid-
ously discussed in Salerno et al4), because it leads to an im- ance) indicated that use of b-blockers in patients that had a
balance in the pathways the normal heart relies on for energy previous myocardial infarction but no heart failure was asso-
production. Under normoxic conditions, the healthy heart gen- ciated with a better cardiovascular prognosisdetermined by
erates approximately two-thirds of its energy (in the form of reduced reinfarction ratebut did not reduce overall mortal-
adenosine triphosphate, ATP) from the free fatty acid (FFA) ity. Additionally, in patients without previous myocardial infarc-
pathway; the remaining energy production is derived from tion, b-blockers did not reduce cardiovascular events but were
glucose oxidation and lactate.5-7 Under hypoxic conditions, rather associated with a higher incidence of stroke, confirm-
such as mild-to-moderate ischemia, myocardial cells turn to ing previous meta-analyses of studies performed in hyperten-
another more oxygen-efficient pathway to generate sufficient sive patients.9 The hypothesized mechanism to explain these
ATP to support calcium homeostasis and maintenance of ion- potential deleterious effects of b-blockers is related to an in-
ic gradients: their response is to increase glucose uptake, as sufficient reduction in central aortic pressure, potentially re-
glycolysis requires less oxygen per mole of ATP generated lated to heart rate reduction, which in certain contexts would
than FFA oxidation. Severe ischemia, however, rapidly induces not play a positive role. Therefore, there is still no clear evidence
an imbalance between cardiac tissue oxygen demand and from randomized clinical trials for the efficacy of b-blockers
the available coronary blood supply. Changes in myocardial used in first-line treatment in patients with chronic stable angi-
function, metabolism, and morphology ensue, leading to ar- na. Yet, we enthusiastically continue to use them.
rhythmias, contractile failure, and electrophysiological abnor-
malities. Myocardial cell uptake of glucose decreases and If a patient continues to complain of symptoms after the first-
conversion to lactate increases; there is a switch from lactate line treatment scheme has been implemented, other drugs,
uptake to lactate production, and most pyruvate is trans- such as calcium-channel blockers or long-acting nitrates,
formed into lactate, increasing cell acidosis. Concurrently, use could be prescribed. Calcium-channel blockers (mostly dihy-
of the FFA pathway slows, and overall ATP production de- dropyridine derivatives) cause coronary and peripheral vasodi-
creases. The results of such metabolic changes include the lation (but increase heart rate and partially reduce the beneficial
disruption of cell homeostasis, alterations in membrane heart ratelowering effect of b-blockers); the phenylalkylamine
structure, and ultimately cell death. derivative verapamil and, to a lesser extent, diltiazem (benzo-
thiazepine calcium-channel blocker) reduce heart rate and
This review discusses the rationale behind a pharmacological
approach to stop this vicious circle in patients with chronic is-
chemic heart disease. SELECTED ABBREVIATIONS AND ACRONYMS

ATP adenosine triphosphate


Medical treatment of chronic ischemic heart disease ATPCI efficAcy and safety of trimetazidine in Patients
In addition to lifestyle and hygienic dietary measures, guide- with angina pectoris having been treated by
line-recommended first-line treatment for patients with stable percutaneous Coronary Interventions
angina includes aspirin, statins, and b-blockers. This recom- CAD coronary artery disease
mendation is consistent across all guidelines for the diagno- CHARISMA Clopidogrel for High Atherothrombotic Risk
sis and management of this condition. The main probable and Ischemic Stabilization Management and
mechanism by which b-blockers relieve anginal symptoms is Avoidance
a reduction in both heart rate and contractility. However, their FFA free fatty acid
indication as a first-line treatment is still under debate. They PCI percutaneous coronary intervention
are given a class I indication for the treatment of chronic angi- REACH REduction of Atherothrombosis for Continued
na, mainly on the basis of a general agreement on the issue, Health [registry]
whereas they are attributed a level of evidence A on the basis TRIMPOL-I TRIMetazidine in POLand [study]
of studies that were carried out in patients after a myocardial

Combining hemodynamic and metabolic agents in ischemic heart disease Fragasso MEDICOGRAPHIA, Vol 38, No. 3, 2016 321
FOCUS

contractility and are used when b-blockers are contraindicat- to nonadipose tissue exacerbates metabolic abnormalities
ed. Combining verapamil or diltiazem with b-blockers yields characteristic of the insulin resistance syndrome,12 a common
additive effects in terms of bradycardia, heart block, and neg- pattern in patients with ischemic heart disease. Furthermore,
ative inotropic effects. When added to b-blockers or calcium- there is new evidence that elevated levels of FFA may not only
channel blockers, long-acting nitrates improve exercise toler- impair glucose uptake in heart and skeletal muscle but also
ance, increase time to onset of angina, and reduce ST-segment alter metabolism in the vascular endothelium, which leads to
depression during exercise testing; however, their use is limited premature cardiovascular disease.13 These findings suggest
by the development of tolerance on long-term administration. that metabolic therapy could have a beneficial role in glucose
In summary and similarly to b-blockers, there is no clear-cut metabolism homeostasis.
evidence of the prognostic utility of these additional antiang-
inal drugs in chronic CAD. Furthermore, conflicting evidence Manipulation of cardiac energy metabolism through a num-
exists about combining antianginal hemodynamic drugs. ber of approaches has been investigated. Trimetazidine is the
most extensively studied cardiac metabolic drug and has been
Having said this, pharmacological treatment of chronic is- shown to increase glucose oxidation and reduce FFA utiliza-
chemic heart disease continues to be based mostly on b- tion, restoring cardiac coupling between glycolysis and glu-
blockers, calcium-channel blockers, and nitrates. In most cas- cose oxidation. The next section will look more closely at the
es, if symptoms are not brought under control by treatment beneficial effects of cardiac metabolic manipulation by trimeta-
with a single traditional antianginal drug, a drug combination zidine in ischemic heart disease.
would be used, with the addition of a second or third agent.
However, there are no clinical studies demonstrating a real Complementary role of trimetazidine in ischemic
additive efficacy of a combination of classic hemodynamical- heart disease
ly active drugs as compared with monotherapy. Furthermore, Trimetazidines use in patients with ischemic heart disease has
significant side effects may limit the maximal doses that can consistently provided clinical benefits (previously discussed
be used for such drugs, especially in an aged population. In in Salerno et al4). Although its mechanism of action is still un-
such a context, the use of alternative therapeutic approaches der debate,14,15 experimental evidence indicates that trimetazi-
would be warmly welcomed and, with this in mind, pharma- dine exerts its effects predominantly through partial inhibition
cologically addressing the underlying derangements in car- of mitochondrial long-chain 3-ketoacyl coenzyme A thio-
diac metabolism, discussed next in further detail, could be a lase, the last enzyme involved in b-oxidation,16 in effect caus-
rational solution to this problem. ing a switch in energy substrate use away from FFA to glucose
and lactate. As mentioned briefly in the previous section, the
Pharmacological manipulation of cardiac energy resulting reduction in FFA oxidation and increase in glucose
metabolism oxidation restores the myocardial coupling between glycoly-
Given the above-described pathophysiology of ischemic heart sis and carbohydrate oxidation, allowing ATP production with
disease and the difficulties encountered with many patients less consumption of oxygen.17 Trimetazidine also promotes
when trying to control the total ischemic burden with classic membrane phospholipid turnover during ischemia and reper-
hemodynamically active drugs, an adjunctive therapeutic op- fusion, redirecting FFA toward phospholipids and thus increas-
tion that pharmacologically manipulates cardiac energy me- es the cells tolerance to ischemia-reperfusion damage.17-19
tabolism seems reasonable (previously discussed in Salerno Trimetazidines anti-ischemic actions are independent of he-
et al4). This approach is based on stimulating myocardial glu- modynamic changes and are associated with a greater re-
cose oxidation to optimize cardiac energy metabolism, and covery of mechanical function after ischemia.17
is proven to improve cardiac function and protect myocar-
dial tissue against ischemia-reperfusion injury.10 Myocardial Trimetazidines efficacy in the treatment of angina pectoris has
glucose oxidation can be promoted either directly by stimu- been investigated under various conditions: trimetazidine ad-
lating glucose metabolism or indirectly by inhibiting fatty acid ministered as a monotherapy or in combination, acutely or
b-oxidation, producing a shift of energy substrate utilization over a longer-term period, as initial treatment, and in patients
away from fatty acid metabolism and toward glucose metab- resistant to b-blockers or calcium-channel antagonists.20-30
olism, a more oxygen-efficient path to ATP production (more
ATP produced per mole of oxygen used). Indeed, oxygen con- Initially studied in patients with chronic stable effort angina
sumption efficiency in the heart can be improved within the during exercise testing, acute administration of trimetazidine
range of 16% to 26% by the increased use of glucose and increased effort tolerance and delayed the appearance of is-
lactatemore efficient fuels for aerobic respiration.10 chemic symptoms and electrocardiogram changes.20

Additionally, the uptake of glucose in the heart and arm skele- With long-term treatment, the benefits seen with acute admin-
tal muscle has been shown to be inversely related to serum istration were confirmed. Such treatment was well-tolerat-
FFA levels,11 and an increased flux of FFA from adipose tissue ed, with no appreciable side effects, including no significant

322 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Combining hemodynamic and metabolic agents in ischemic heart disease Fragasso
FOCUS

A B

Figure 1. Bar charts showing mean values (+ 1 standard deviation) from dobutamine stress echocardiography in chronic ischemic
heart disease patients taking placebo (grey bars) or trimetazidine (red bars) on top of optimal hemodynamic therapy.
Trimetazidine significantly induced an increase in the administered dobutamine dose (panel A) versus placebo. Despite a higher administered stress, left ventricular
function (assessed by wall motion score index) was significantly less impaired when patients were on trimetazidine (panel B). These findings indicate that metabolic
therapy added to treatment schemes yields a better response to stress.
Based on data from reference 32: Lu et al. Am J Cardiol. 1998;82:898-901.

changes in heart rate and/or aortic pressure.21 In comparison In diabetic patients with chronic stable angina, Marazzi et al
studies, improvement in ischemic threshold and exercise tol- have shown that trimetazidine added to standard medical ther-
erance on trimetazidine treatment is similar to that reported apy reduces the number of episodes of ST-segment depres-
for propranolol and nifedipine, and there was even a lower sion and silent ischemia and reduces total ischemic burden.28
incidence of side effects.22,23 In patients with stable angina pectoris, the efficacy of trime-

Trimetazidines effects were shown to be additive to those of


hemodynamically active drugs, with direct evidence provided
by a randomized, double-blind study in patients with chronic
effort angina and documented CAD. This study compared the
combination of trimetazidine plus the b-blocker propranolol
with nitrates plus propranolol, and found that the combina-
tion including trimetazidine was not only more effective and
better tolerated than the other combination, but also the only
one that showed improvement. The other combination had no
effect on symptoms and exercise capacity.24 In another ran-
domized, double-blind study in patients with angina uncon-
trolled by diltiazem, the addition of trimetazidine to a full-dose
diltiazem treatment scheme yielded beneficial effects.25 It sig-
nificantly reduced the number of ischemic attacks, prolonged
the time to 1-mm ST-segment depression, increased the time
to onset of exercise-induced angina, and increased the max-
imum work at peak exercise, without adverse hemodynamic
Figure 2. Insulin sensitivity. Bar chart showing mean (+1 standard
events or increased side effects. In yet another study, this time deviation) glucose infusion rate at the hyperinsulinemic/euglycemic
in patients with stable effort angina uncontrolled by metoprolol clamp during short-term (15 days) and long-term (6 months)
alone, 12-week treatment with trimetazidine and metoprolol placebo and trimetazidine administration in patients with chronic
significantly reduced clinical symptoms compared with place- ischemic heart disease, left ventricular dysfunction, and diabetes.
bo and metoprolol.26 A previous study, TRIMPOL-I (TRIMetazi- The infusion rate of glucose necessary to maintain euglycemia after an insulin
bolus was greater when patients were on trimetazidine, both in the short and
dine in POLand), had already shown that in diabetic patients, long term. This indicates that in these high-risk patients, trimetazidine, compared
4 weeks of treatment with trimetazidine was associated with with placebo, was also able to significantly improve insulin sensitivity.
Abbreviations: LT, long term; ST, short term.
a significantly lower number of anginal episodes and an im- Based on data from reference 33: Fragasso G et al. Am Heart J. 2003;146: E1-E8
provement in myocardial ischemia and exercise capacity.27 and reference 36: Monti et al. Am J Physiol Endocrinol Metab. 2006; 290:E54-E59.

Combining hemodynamic and metabolic agents in ischemic heart disease Fragasso MEDICOGRAPHIA, Vol 38, No. 3, 2016 323
FOCUS

Figure 3. Baseline and follow-up


Tpeak-Tend dispersion (mean + 1
standard deviation) in patients with
left ischemic and nonischemic left
ventricular dysfunction and treated
with trimetazidine (red columns) or
conventional therapy alone (controls;
greycolumns).
Tpeak-Tend dispersion index is a noninvasive
marker of dispersion of ventricular repolariza-
tion and is positively related to the risk of
arrhythmias. The evidence of a significant
TMZ-induced Tpeak-Tend dispersion reduc-
tion only in patients with post-ischemic left
ventricular dysfunction supports the hypothe-
sis that the potential antiarrhythmic properties
of TMZ could be directly mediated by the
anti-ischemic action of the drug.
Abbreviations: TMZ, trimetazidine.
Based on data from reference 37: Cera et
al. J Cardiovasc Pharmacol Ther. 2010;15:
24-30.

tazidine was found to be comparable to that of other drugs iological effects37 (Figure 3) may also be involved. Long-term
that have no influence on heart rate (ie, other nonheart-rate- administration of trimetazidine has also been shown to im-
lowering antianginal drugs).29 prove survival and event-free survival in patients with ischemic
and nonischemic left ventricular dysfunction (Figure 4).38
Trimetazidine has also been confirmed to be effective in dif-
ferent settings of stable coronary disease,30-32 expanding its Similarly to other established antianginal drugs, the main lim-
use to include patients with heart failure both of ischemic and itations on the wide use of trimetazidine in chronic ischemic
nonischemic origin.33,34 In such contexts, trimetazidine im- heart disease include the paucity of data on mortality and ma-
proves symptoms, cardiac response to ischemia, left ventric- jor cardiovascular events and on direct comparisons between
ular function (Figure 1, page 323),32 and thus quality of life trimetazidine and established antianginal therapies. Neverthe-
as well. The main mechanism of action is probably through less, in 2005, a Cochrane review including 23 studies (1378
a trimetazidine-induced increase in myocardial cellular ener- patients) concluded that trimetazidine is a well-tolerated drug
gy reserve.35 However, improved endothelial function36 and that provides benefit in patients with stable angina, in terms
increased insulin sensitivity33,36 (Figure 2, page 323) may also of patient-reported intake of glyceryl trinitrate tablets and num-
play a role; it is possible that indirect beneficial electrophys- ber of weekly angina episodes when used as monotherapy
and in combination with conventional antianginal agents.39
A more recent meta-analysis (13 studies, 1628 patients) that
compared trimetazidine with conventional antianginal drugs
confirmed the efficacy of trimetazidine treatment for stable
angina pectoris, regardless of treatment duration.40

At present, the European Society of Cardiology indicates tri-


metazidine as an effective adjunctive treatment in patients with
angina not completely controlled by standard hemodynamic
therapy.41

Combined metabolic action of b-blockers and


trimetazidine
Despite their above-described pitfalls, b-blockers continue to
Figure 4. Effects of trimetazidine versus conventional therapy alone be the clinical mainstay of ischemic heart disease treatment.
(control) on 5-year global and cardiovascular mortality in patients They act principally through reduction in oxygen consumption
with ischemic and nonischemic left ventricular dysfunction. by reducing heart rate and inotropism. However, they could
The histogram on the left shows an 11.3% reduction in 5-year global mortality
when trimetazidine (TMZ) is administered in addition to standard medical therapy
have a direct complementary metabolic effect themselves, by
(P=0.015). The histogram on the right shows an 8.7% reduction in 5-year cardio- reducing peripheral lipolysis and reducing FFA availability. In-
vascular mortality in the same patients (P=0.05). deed, there is evidence that b-blockade reduces FFA use in fa-
Abbreviations: CV, cardiovascular; TMZ, trimetazidine.
Based on data from reference 38: : Fragasso G et al. Int J Cardiol. 2013;163: vor of greater glucose use in cardiac patients.42 Such a change
320-325. in cardiac energy metabolism could be a potential mechanism

324 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Combining hemodynamic and metabolic agents in ischemic heart disease Fragasso
FOCUS

for the decreased cardiac oxygen consumption and improve- quent and is frustrating for patients and doctors. In fact, sub-
ment in energy efficiency observed with b-blocker treatment sequent repeat coronary angiography and revascularization
of ischemic heart disease and heart failure.43 Whether nonse- procedures introduce both additional risk for the patient and
lective b-blockers are more efficient than selective ones in shift- cost to the health care system. A more effective medical strat-
ing whole-body substrate utilization from FFA to glucose oxi- egy could certainly improve the management of these pa-
dation44 is still under debate.45 Nonetheless, the better survival tients. In this context, an ongoing international, multicenter,
rates observed with nonselective b-blockers could be explained randomized clinical study would provide the cardiological com-
by their effect on the metabolism.46 Additionally, inhibition of munity with new solid data in a few years time. The purpose
activity in the sympathetic nervous system with the central- of the ongoing ATPCI study (efficAcy and safety of Trimetazi-
acting antihypertensive drug moxonidine has been associ- dine in Patients with angina pectoris having been treated by
ated with increased mortality in patients with chronic heart fail- percutaneous Coronary Intervention; EudraCT Number: 2010-
ure.47 In fact, despite a significant reduction in catecholamine 022134-89) is to evaluate the long-term efficacy and safety of
spillover from the synapses in the sympathethic nervous sys- trimetazidine, in addition to evidence-based cardiovascular
temthus reducing catecholamine levels in the bloodand, therapy, in patients having had a recent percutaneous coro-
consequently, heart rate, moxonidine increases both FFA use nary intervention. The primary objectives are to demonstrate
and myocardial oxygen consumption.48 This could be the rea- the superiority of trimetazidine over placebo in preventing re-
son why central sympathetic inhibition fails to prevent deaths currence or exacerbation of angina pectoris and in reducing
in long-term studies in patients with chronic heart failure; it also cardiac events, and also to document its safety by analyzing
indicates that the main mechanism of action of b-blockers in the occurrence of serious adverse events. Apart from the eval-
cardiac syndromes probably involves something other than uation of the effects of trimetazidine in this widely encoun-
a simple reduction in heart rate. Thus, it is possible that the de-
gree of heart rate reduction is just a marker of the functional
response after the administration of b-blockers, ie, a conse-
quent effect rather than a mechanism. On this basis, we can
hypothesize that b-blockers and trimetazidine have a comple-
mentary, synergistic metabolic action: whereas the former re-
duces FFA availability, the latter decreases their cardiac utiliza-
tion. Overall, this drug-induced metabolic shift could reduce
FFA oxidation and increase flux through pyruvate dehydro-
genase with a consequent energy-sparing effect.35,49

There is evidence to suggest that trimetazidines metabolic


effect may occur in other organs and tissues as well.50 In fact,
apart from a reduction in whole-body energy demand, a trend
for a reduction in whole-body lipid oxidation and in fasting plas-
ma FFA concentration has also been observed (Figure 5).50 Figure 5. Bar chart showing mean (+ 1 SD) resting energy expen-
diture (kcal/day) at baseline and after 3 months of follow-up in
This general metabolic shift could in the end reduce the over- patients with ischemic and nonischemic left ventricular dysfunction
all metabolic requirements of the body, resulting in a very at- treated with conventional therapy plus trimetazidine (red columns)
tractive adaptation strategy in the context of coronary and my- or conventional therapy alone (grey columns).
ocardial insufficiencies. Interestingly, b-blockers have also been Trimetazidine significantly reduces the whole-body energy expenditure, indicating
shown to directly affect whole-body metabolism. In trained a potential role of this metabolic drug on overall metabolic requirements of the body.
Based on data from reference 50: Fragasso et al. Heart. 2011;97:1495-1500.
athletes, b-blockade abolishes the increase in plasma glu-
cose levels that occur during intense exercise, owing to an in- tered patient population, this new study will also provide us
creased peripheral glucose uptake and no significant change with much useful information about the current epidemiolo-
in glucose production.51 Such effects from b-blockade on glu- gy, treatment, and outcome in patients with chronic ischemic
cose kinetics could be mediated directly; they could also be heart disease. It is expected to provide some of the most sol-
indirectly mediated through changes in lipid substrates and/or id data about the treatment of chronic ischemic heart disease
counter-regulatory hormones. ever produced.

Potential role of metabolic therapy in revascularized Conclusions


chronic ischemic heart disease Optimization of cardiac energy metabolism is attractive as an
Despite no clearly demonstrated prognostic gain, nowadays, approach to protect myocardial cells from ischemia and to
revascularization procedures are often considered first for the improve performance of dysfunctioning myocardium. To that
control of angina pectoris. However, recurrent or persistent effect, trimetazadine, which shifts the energy substrate pref-
angina after initially successful revascularization is not infre- erence away from FFA metabolism toward increased glu-

Combining hemodynamic and metabolic agents in ischemic heart disease Fragasso MEDICOGRAPHIA, Vol 38, No. 3, 2016 325
FOCUS

cose oxidation, has been shown by a number of studies to be ence very high morbidity and mortality rates in spite of treat-
an effective adjunctive treatment in patients with chronic is- ment efforts. Furthermore, most cardiac diseases are asso-
chemic heart disease and heart failure, reducing ischemic ciated with derangements in glucose homeostasis, which cer-
burden and improving left ventricular metabolism and func- tainly contribute to primary disease progression. An advantage
tion. Whether the reported experimental and clinical benefits of trimetazidine treatment is the combined beneficial effects
translate into improved prognosis is currently being ascer- that FFA inhibitors have on left ventricular function and glu-
tained by an ongoing international randomized clinical trial. cose metabolism, which would be especially advantageous in
This has potential to be a major therapeutic advance in chron- cardiac patients with coexisting myocardial dysfunction and
ic ischemic heart disease patients, who continue to experi- glucose metabolism abnormalities.

References
1. Anderson JL, Adams CD, Antman EM, et al; American College of Cardiology; 17. Tabbi-Anneni I, Helies-Toussaint C, Morin D, Bescond-Jacquet A, Lucien A,
American Heart Association Task Force on Practice Guidelines (Writing Com- Grynberg A. Prevention of heart failure in rats by trimetazidine treatment: a con-
mittee to Revise the 2002 Guidelines for the Management of Patients With Un- sequence of accelerated phospholipid turnover? J Pharmacol Exp Ther. 2003;
stable Angina/Non-ST-Elevation Myocardial Infarction); American College of 304:1003-1009.
Emergency Physicians; Society for Cardiovascular Angiography and Interven- 18. Sentex E, Sergiel JP, Lucien A, Grynberg A. Is the cytoprotective effect of
tions; Society of Thoracic Surgeons; American Association of Cardiovascular trimetazidine associated with lipid metabolism? Am J Cardiol.1998;82:18K-24K.
and Pulmonary Rehabilitation; Society for Academic Emergency Medicine. ACC/ 19. Sentex E, Sergiel JP, Lucien A, Grynberg A. Trimetazidine increases phos-
AHA 2007 guidelines for management of patients with unstable angina/non- pholipid turnover in ventricular myocyte. Mol Cell Biochem.1997;175:153-162.
ST-elevation myocardial infarction. J Am Coll Cardiol. 2007;50:e1-e57. 20. Sellier P, Audouin P, Payen B, Corona P, Duong TC, Ourbak P. Acute effects of
2. Antman EM, Anbe DT, Armstrong PW, et al; American College of Cardiology/ trimetazidine evaluated by exercise testing. Eur J Clin Pharmacol. 1987;33:205-
American Heart Association Task Force on Practice Guidelines (Writing Com- 207.
mittee to Revise the 1999 Guidelines for the Management of Patients With 21. Szwed H, Hradec J, Preda I. Anti-ischaemic efficacy and tolerability of trimetazi-
Acute Myocardial Infarction). ACC/AHA guidelines for the management of pa- dine administered to patients with angina pectoris: results of three studies. Coron
tients with ST-elevation myocardial infarctionexecutive summary. Circulation. Artery Dis. 2001;12(suppl 1):S25-S28.
2004;110:588-636. 22. Dtry JM, Sellier P, Pennaforte S, Cokkinos D, Dargie H, Mathes P. Trimetazi-
3. Windecker S, Kolh P, Alfonso F, et al. 2014 ESC/EACTS guidelines on myocar- dine: a new concept in the treatment of angina. Comparison with propranolol in
dial revascularization: the Task Force on Myocardial Revascularization of the patients with stable angina. Br J Clin Pharmacol. 1994;37:279-288.
European Society of Cardiology (ESC) and the European Association for Cardio- 23. Dalla-Volta S, Maraglino G, Della-Valentina P, Viena P, Desideri A. Comparison
Thoracic Surgery (EACTS) Developed with the special contribution of the Eu- of trimetazidine with nifedipine in effort angina: a doubleblind, crossover study.
ropean Association of Percutaneous Cardiovascular Interventions (EAPCI). Eur Cardiovasc Drugs Ther. 1990;4(suppl 4):853-859.
Heart J. 2014;35:2541-2619. 24. Michaelides AP, Spiropoulos K, Dimopoulos K, Athanasiades D, Toutouzas P.
4. Salerno A, Fragasso G, Montanaro C, et al. Role of metabolic modulation in the Antianginal efficacy of the combination of trimetazidine-propranolol compared
management of chronic ischemic heart disease. Clin Med Insights Ther. 2010; with isosorbide dinitrate-propranolol in patients with stable angina. Clin Drug
2:577-587. Invest. 1997;13:8-14.
5. Bing RJ, Siegel A, Ungar I, Gilbert M. Metabolism of the human heart. II. Stud- 25. Manchanda SC, Krishnaswami S. Combination treatment with trimetazidine and
ies on fat, ketone and amino acid metabolism. Am J Med. 1954;16:504-515. diltiazem in stable angina pectoris. Heart. 1997;78:353-357.
6. Shipp JC, Opie LH, Challoner D. Fatty acid and glucose metabolism in the per- 26. Szwed H, Sadowski Z, Elikowski W, et al. Combination treatment in stable ef-
fused heart. Nature. 1961;189:1018-1019. fort angina using trimetazidine and metoprolol: results of a randomized, double-
7. Wisnecki JA, Gertz EQ, Neese RA, Mayr M. Myocardial metabolism of free fatty blind, multicentre study (TRIMPOL II). TRIMetazidine in POLand. Eur Heart J.
acids: studies with 14C-labeled substrates in humans. J Clin Invest. 1987;79: 2001;22:2267-74.
359-366. 27. Szwed H, Pachocki R. Domzal-Bochenska M, et al. Efficacy and tolerance of
8. Bangalore S, Steg G, Deedwania P, et al; REACH Registry Investigators. b-Block- trimetazidine, a metabolic antianginal, in combination with a hemodynamic an-
er use and clinical outcomes in stable outpatients with and without coronary tianginal in stable exertion angina. TRIMPOL I, a multicenter study. Presse Med-
artery disease. JAMA. 2012;308:1340-1349. icale. 2000;29:533-538.
9. Bangalore S, Bhatt DL, Steg PG. b-Blockers and cardiovascular events in pa- 28. Marazzi G, Wajngarten M, Vitale C, et al. Effect of free fatty acid inhibition on
tients with and without myocardial infarction: post hoc analysis from the CHA- silent and symptomatic myocardial ischemia in diabetic patients with coronary
RISMA Trial. Circ Cardiovasc Qual Outcomes. 2014;7:872-881. artery disease. Int J Cardiol. 2007;120:79-84.
10. Lopaschuk GD, Stanley WC. Glucose metabolism in the ischemic heart. Cir- 29. Danchin N, Marzilli M, Parkhomenko A, Ribeiro JP. Efficacy comparison of tri-
culation. 1997;95:313-315. metazidine with therapeutic alternatives in stable angina pectoris: a network
11. Nuutila P, Knuuti MJ, Raitakari M, et al. Effect of antilipolysis on heart and skele- meta-analysis. Cardiology. 2011;120:59-72.
tal muscle glucose uptake in overnight fasted humans. Am J Physiol. 1994; 30. Fragasso G, Piatti PM, Monti L, et al. Acute effects of heparin administration
267:E941-E946. on the ischemic threshold of patients with coronary artery disease: evaluation
12. Lewis GF, Carpentier A, Adeli K, Giacca A. Disordered fat storage and mobiliza- of the protective role of the metabolic modulator trimetazidine. J Am Coll Car-
tion in the pathogenesis of insulin resistance and type 2 diabetes. Endocr Rev. diol. 2002;39:413-419.
2002;23:201-229. 31. Fragasso G, Montano C, Perseghin G, et al. The anti-ischemic effect of tri-
13. Steinberg HO, Baron AD. Vascular function, insulin resistance and fatty acids. metazidine in patients with postprandial myocardial ischemia is unrelated to meal
Diabetologia. 2002;45:623-634. composition. Am Heart J. 2006;151:1238.e1-e8.
14. MacInnes A, Fairman DA, Binding P, et al. The antianginal agent trimetazidine 32. Lu C, Dabrowski P, Fragasso G, Chierchia SL. Effects of trimetazidine on is-
does not exert its functional benefit via inhibition of mitochondrial long-chain chemic left ventricular dysfunction in patients with coronary artery disease: a dou-
3-ketoacyl coenzyme A thiolase. Circ Res. 2003;93:e26-e32. ble-blind, placebo-controlled, crossover study. Am J Cardiol.1998;82:898-901.
15. Lopaschuk GD, Barr R, Thomas PD, Dyck JR. Beneficial effects of trimetazi- 33. Fragasso G, Piatti PM, Monti L, et al. Short- and long-term beneficial effects of
dine in ex vivo working ischemic hearts are due to a stimulation of glucose ox- partial free fatty acid inhibition in diabetic patients with ischemic dilated car-
idation secondary to inhibition of long-chain 3-ketoacyl coenzyme A thiolase. diomyopathy. Am Heart J. 2003;146:E18.
Circ Res. 2003;93:e33-e37. 34. Fragasso G, Palloshi A, Puccetti P, et al. A randomized clinical trial of trimetazi-
16. Kantor PF, Lucien A, Kozak R, Lopaschuk GD. The antianginal drug trimetazi- dine, a partial free fatty acid oxidation inhibitor, in patients with systolic dys-
dine shifts cardiac energy metabolism from fatty acid oxidation to glucose oxi- function heart failure. J Am Coll Cardiol. 2006;48:992-998.
dation by inhibiting mitochondrial long-chain 3-ketoacyl coenzyme A thiolase. 35. Fragasso G, Perseghin G, De Cobelli F, et al. Effects of metabolic modulation
Circ Res. 2000;86:580-588. by trimetazidine on left ventricular function and phosphocreatine/adenosine

326 MEDICOGRAPHIA, Vol 38, No. 3, 2016 Combining hemodynamic and metabolic agents in ischemic heart disease Fragasso
FOCUS

triphosphate ratio in patients with heart failure. Eur Heart J. 2006;27:942-948. 44. Podbregar M, Voga G. Effect of selective and nonselective b-blockers on rest-
36. Monti LD, Setola E, Fragasso G, et al. Metabolic and endothelial effects of tri- ing energy production rate and total body substrate utilization in chronic heart
metazidine on forearm skeletal muscle in patients with type 2 diabetes and is- failure. J Cardiac Fail. 2002:8:369-378.
chemic cardiomyopathy. Am J Physiol Endocrinol Metab. 2006;290:E54-E59. 45. Sharma V, Dhillon P, Wambolt R, et al. Metoprolol improves cardiac function
37. Cera M, Salerno A, Fragasso G, et al. Beneficial electrophysiological effects and modulates cardiac metabolism in the streptozotocin (STZ) diabetic rat. Am
of trimetazidine in patients with post-ischemic chronic heart failure. J Cardio- J Physiol Heart Circ Physiol. 2008;294:H1609-H1620.
vasc Pharmacol Ther. 2010;15:24-30. 46. Poole-Wilson P, Swedberg K, Cleland J, et al; Carvedilol Or Metoprolol Euro-
38. Fragasso G, Rosano G, Baek Hong S, et al. Effect of partial acid oxidation pean Trial Investigators. Comparison of carvedilol and metoprolol on clinical out-
inhibition with trimetazidine on mortality and morbidity in heart failure: results comes in patients with chronic heart failure in the Carvedilol Or Metoprolol Eu-
from an international multicentre retrospective cohort study. Int J Cardiol. 2013; ropean Trial (COMET): randomised controlled trial. Lancet. 2003;362:7-13.
163:320-325. 47. Wiltse C, Wright T; MOXCON Investigators. Adverse mortality effect of central
39. Ciapponi A, Pizarro R, Harrison J. Trimetazidine for stable angina. Cochrane sympathetic inhibition with sustained-release moxonidine in patients with heart
Database Syst Rev. 2005;(4):CD003614. failure (MOXCON). Eur J Heart Fail. 2003;5:659-666.
40. Peng S, Zhao M, Wan J, Fang Q, Fang D, Li K. The efficacy of trimetazidine on 48. Mobini R, Jansson PA, Bergh CH, et al. Influence of central inhibition of sym-
stable angina pectoris: a meta-analysis of randomized clinical trials. Int J Car- pathetic nervous activity on myocardial metabolism in chronic heart failure: acute
diol. 2014;177:780-785. effects of the imidazoline I1-receptor agonist moxonidine. Clin Sci. 2006;110:
41. Task Force Members; Montalescot G, Sechtem U, Achenbach S, et al. 2013 329-336.
ESC guidelines on the management of stable coronary artery disease: The Task 49. Lopaschuk GD, Belke DD, Gamble J, Itoi T, Schonekess BO. Regulation of fat-
Force on the management of stable coronary artery disease of the European ty acid oxidation in the mammalian heart in health and disease. Biochim Bio-
Society of Cardiology. Eur Heart J. 2013;34:2949-3003. phys Acta. 1994;1213:263-276.
42. Wallhaus TR, Taylor M, DeGrado TR, Russell DC. Myocardial free fatty acid and 50. Fragasso G, Salerno A, Lattuada G, et al. Effect of partial inhibition of fatty acid
glucose use after carvedilol treatment in patients with congestive heart failure. oxidation by trimetazidine on whole body energy metabolism in patients with
Circulation. 2001;103:2441-2446. chronic heart failure. Heart. 2011;97:1495-1500.
43. Spoladore R, Fragasso G, Perseghin G, et al. Beneficial effects of beta-block- 51. Howlett KF, Watt MJ, Hargreaves M, Febbraio MA. Regulation of glucose ki-
ers on left ventricular function and cellular energy reserve in patients with heart netics during intense exercise in humans: effects of a- and b-adrenergic block-
failure. Fundam Clin Pharmacol. 2013;27:455-64. ade. Metabolism. 2003;52:1615-1620.

Keywords: angina; metabolism; myocardial ischemia; trimetazidine

LASSOCIATION D AGENTS HMODYNAMIQUES ET MTABOLIQUES


DANS LA CARDIOPATHIE ISCHMIQUE
Lischmie myocardique peut tre considre comme un problme mtabolique conduisant un dsquilibre des
voies utilises normalement par le cur pour produire de lnergie. Lutilisation dagents pharmacologiques pour
optimiser le mtabolisme nergtique cardiaque en stimulant loxydation du glucose au niveau du myocarde peut tre
une option thrapeutique efficace. Ainsi, la trimetazidine est un agent mtabolique qui agit indirectement en inhibant
loxydation b des acides gras, ce qui privilgie le mtabolisme du glucose, un substrat nergtique plus efficace pour
la production dATP que les acides gras. Lefficacit de la trimtazidine dans le traitement de langor a t value
dans diffrentes conditions : administration de la trimtazidine en monothrapie ou en association, en priode aigu
ou plus long terme, en traitement initial, et chez les patients rsistants aux b-bloquants ou aux inhibiteurs calciques.
Toutes les tudes publies employant la trimtazidine chez des patients ayant une cardiopathie ischmique chro-
nique ont invariablement rapport des effets cliniques bnfiques sans vnements indsirables hmodynamiques.
En fait, chez les patients ayant une cardiopathie ischmique chronique avec dysfonction ventriculaire gauche, la tri-
mtazidine est un complment particulirement efficace en termes damlioration de la fonction et du mtabolisme
ventriculaires gauches. Une tude clinique randomise en cours chez des patients ayant une maladie coronaire re-
vascularise devrait permettre de dterminer si les bnfices cliniques et exprimentaux de la trimtazidine se tra-
duisent aussi par une amlioration du pronostic.

Combining hemodynamic and metabolic agents in ischemic heart disease Fragasso MEDICOGRAPHIA, Vol 38, No. 3, 2016 327

U P DAT E

Bioresorbable stents are very


promising: they offer the vascular
scaffold for a certain amount of
time and then the implanted ma-
terials are progressively resorbed.
This offers a number of advantages,
History and evolution
including the elimination of foreign
bodies inside the wall, restoration
of endothelial coverage, and pos-
of coronary stenting
sibly restoration of vasomotion.
These biodegradable stents can
be divided into two categories:
metallic stents that are magne-
sium based and those that are
polymeric resorbable.

by M. E. Bertrand, France

C
oronary stenting represents a major step in the history of percutaneous
coronary angioplasty. Jacques Puel performed the first stent implanta-
tion in man in 1986, and research in the area took off immediately. The
primary concern about balloon angioplasty procedures was safety, as there is
a risk of abrupt occlusion during the procedure. This risk and the subsequent
need for emergency bypass surgery were dramatically reduced with stent im-
plantation. Nevertheless, investigators then faced another problem: the risk
of stent thrombosis. However, such risk is suppressed by the use of a dual an-
tiplatelet treatment. Coronary stenting has now played a major role in the fight
against restenosis, with drug-eluting stents considerably reducing this risk,
Michel E. BERTRAND, even in high-risk patients (diabetics). With coronary stenting, coronary inter-
MD, FESC, FRCP, FACC ventional procedures have become the primary approach to myocardial revas-
Hpital Universitaire
Cardiologique, Lille cularization.
FRANCE Medicographia. 2016;38:328-334 (see French abstract on page 334)

he first coronary angioplasty in man was performed by Andreas Gruentzig

T from Zurich (Switzerland) in September 1977.1 This new, noninvasive approach


for myocardial revascularization would have an immediate and great success.
In the years that followed, significant technical improvements were proposed. The
over-the-wire technique, designed by John Simpson,2 and the monorail system3 were
rapidly adopted by the interventional cardiology community. However, new problems
were observed: the most important pertained to the safety of the procedure. During
or immediately after angioplasty, an abrupt occlusion of the vessel was observed in
3% to 5% of cases. This accident was related to an occlusive dissection or an occlu-
sive thrombosis of the vessel. Thus, when use of coronary angioplasty was beginning,
it was recommended to perform the procedure with a surgical backup, enabling im-
mediate emergency bypass surgery if needed. In most cases, this emergency opera-
tion was followed by limited myocardial necrosis. Aside from this immediate problem,
30% to 45% of cases experienced later restenosis in a process such as the follow-
ing: the mechanism of initial enlargement of the lumen was through dissection of the
vessel; and this dissection was followed by a process of healing, which involved mi-
gration and proliferation of smooth muscle cells into a loose extracellular matrix; thus,
Address for correspondence: intimal hyperplasia occurred, contributing to the renarrowing of the dilated vessel.
Michel E. Bertrand,
Hpital Universitaire Cardiologique, Later, it was discovered that another important phenomenon was occurring nega-
Lille, FRANCE tive remodeling of the vessel, leading to shrinkage of the dilated segment. It is with-
(e-mail: mbertrand2007@gmail.com)
in this context that coronary stenting arose and solved, at least partially, the two ma-
www.medicographia.com jor problems of coronary angioplasty, namely abrupt occlusion and restenosis.

328 MEDICOGRAPHIA, Vol 38, No. 3, 2016 History and evolution of coronary stenting Bertrand
U P DAT E

History of stenting percutaneous approach; the solution was found by the engi-
It has been said that the Egyptians tried to cure the narrow- neer Christian Imbert. The self-expandable stent, which could
ing of urethra by introducing a small reed in the urinary canal be implanted via a percutaneous femoral approach, was thus
in order to reestablish a more fluid relationship between the created. Looking for centers to experiment with his device,
internal and external milieu. Christian Imbert met Ulrich Sigwart in Lausanne and the group
of Jacques Puel in Toulouse.
In September 1912, the French surgeon, Alexis Carrel made
a prophetic statement following the publication of his work In cooperation with radiologists from Toulouse, Jacques Puel
on permanent intubation of the thoracic aorta in dogs4: conducted experiments in sheep and dogs. These experi-
The permanent intubation of a large vessel is a simple opera- ments quickly (probably too quickly) met with success and
tion. It may become practical, if the shape and the nature of showed, at least at first glance, that it was easy (possibly too
the tube be modified as to avoid lacerations (). The question easy) to implant the endocoronary prosthesis percutaneous-
of the application of this method to human surgery may then, ly and that rapid endothelialization of the struts occurred. How-
possibly, be considered. ever, these animal experiments did not reveal the high risk of
It took more than 70 years to verify this assumption. subsequent thrombosis. Later, Puel confessed that he had
probably underestimated this risk. Simultaneously in Lau-
The first stent was conceived from the progress in therapeu- sanne, Ulrich Sigwart was conducting experimental implan-
tic intervention initiated by Charles Dotter5; he was a true in- tation in dogs.
ventor and a pioneer in the interventional cardiovascular world.
He opened the way for interventional cardiology, and he start- The first stent implantation in man was performed on March
ed to design the first vascular stent at the end of the 1980s 28, 1986 by Puel using the Wallstent.7 The medical history of
with insertion of plastic tubes and collapsible stainless-steel this first patient is quite simple: the patient was a 63-year-old
prostheses into the femoral or popliteal arteries of dogs. male with arterial hypertension and symptomatic restenosis
6 months after treatment of a midleft anterior descending ar-
In 1978, a young fellow who had recently arrived from Argenti- tery lesion (Figure 1). In 1986, with evidence-based medicine
na attended a lecture by Andreas Gruentzig at a meeting of
the Society of Cardiovascular and Interventional Radiology.
His name was Julio Palmaz, and he thought that the prob-
lems that doctor Gruentzig had with his balloon could be avoid-
ed by inserting some sort of a scaffold at the time of dilata-
tion.6 The chairman of the department said that it could be
a nice research project. After writing a report and making draw-
ings, he started to build a prototype in his garage with cop-
per wire and solder materials. A

The wire was woven in a crisscross mesh around a pencil with


two rows of pins. Solder was used to fix the cross points to
allow the mesh to retain its shape. Once built, the mesh diam-
eter was decreased by compressing it on progressively small-
er wooden dowels and then was crimped by hand on a fold-
ed balloon. However, the material was excessively rigid, and
the slots and the spaces between were inadequate. Julio Pal-
B
maz searched for a manufacturer, but several companies re-
fused to consider this device.

In Switzerland, the story of stenting started during a party: two


Swedish persons metone, Hans Wallstn, was the design-
er of a revolutionary machine intended to manufacture paper; C
the other, Ake Senning, was a well-known cardiovascular sur-
geon. During the meeting, Senning explained that aortic dis- Figure 1. First stent implantation in man.
section was a very serious acute disease and went on to de- Stent implantation in a 63-year-old male with arterial hypertension and sympto-
matic hypertension 6 months after treatment of a midleft anterior descending
tail his concept of a mechanical scaffolding of the arterial wall artery lesion. The procedure was carried out by Jacques Puel in 1986.
by a latticed metallic tube. Wallstn, very excited, decided to A. Angiogram showing high-grade stenosis of the proximal left anterior descend-
ing artery (yellow arrow) before the procedure. B. The bare metal stent used in
take up the problem of metallic endoprostheses. He created the procedure. C. Angiogram showing the stent in place (yellow arrow).
the Wallstent but had some difficulties finding a solution for a All rights reserved.

History and evolution of coronary stenting Bertrand MEDICOGRAPHIA, Vol 38, No. 3, 2016 329
U P DAT E

Figure 2.
Impact of
stenting on
acute occlu-
sion and
emergency
surgical bailout
procedures.
Abbreviation:
Emerg CABG,
emergency
coronary artery
bypass graft
surgery.
All rights
reserved.

still in its infancy, the patient received no antiplatelet drugs These two clinical trials, conducted initially in Toulouse and lat-
or statins in preparation for stent implantation; rather, he re- er in Lausanne, proved the feasibility of the stent implanta-
ceived only subcutaneous heparin during the procedure and tion method. However, they also demonstrated the high po-
within the next 6 weeks. By chance, he had no stent throm- tential thrombogenic risk posed by introduction of this foreign
bosis or in-stent restenosis within that time span; however, body; nevertheless, stents markedly reduced the risk of acute
he did not escape progression of the atherosclerotic process or subacute occlusion. As a result, the need for emergency
and has had recurrent episodes of angina pectoris related to surgical bailout procedures was drastically reduced (Figure 2).
a new lesion on the ostium of the left anterior descending ar-
tery and another one on the circumflex artery, which was treat- The evolution of coronary stenting over the years that followed
ed in 2004 by a new stent implantation. In the weeks after can be divided into three different partstechnical improve-
that first implant, seven other patients received a self-expand- ment, safety improvement, and restenosis prevention and treat-
able Wallstent without any complications. ment. These will be discussed in turn.

In Lausanne, the results obtained from 9 months of animal ex- u Technical improvements in stenting
periments were convincing enough to persuade the Institu- A number of variations of the stent have been proposed over
tional Review Board to give approval in April 1986 for the use the years. The first stent implanted in the coronary arteries in
of stent implants for three indications: abrupt vessel closure man was the Wallstent (Medinvent) (Figure 3A). It was a self-
after balloon angioplasty, restenosis after balloon angioplas- expanding stent composed of 20 strands of 0.06 to 0.09 mm
ty, and stenosis of saphenous vein bypass grafts.8 After a diameter arranged into a self-expanding mesh design. It was
number of deployments in human femoral and iliac arteries, flexible; its length ranging from 15 to 30 mm; and its diam-
the self-expanding mesh stent was first deployed after balloon eter between 3.0 to 6.0 mm. The mesh was compressed and
dilatation of a tight stenosis in a vein bypass procedure. elongated on the delivery catheter owing to a double wall sleeve
membrane. Retraction of this membrane allowed the progres-
Although initial results were promising, they were misleading sive release into the vessel. The aforementioned work of Julio
the next four patients to undergo the stent procedure expe- Palmaz led to creation of a tubular slotted stent; together
rienced a subacute stent thrombosis. With a single antithrom- with Richard Schatz, it was implanted in coronary arteries for
botic treatment using full-dose heparin, the risk of thrombosis the first time in December 1987 (Figure 3B).9 This was a bal-
was very high. loon-expandable stent crimped on the delivery catheter, and
it became very popular. The Gianturco Roubin stent10 (ap-
Later, Ulrich Sigwart would perform the stent implantation pro- proved in the United States in 1993) (Figure 3C) had a poor
cedure under full anticoagulation treatment with heparin fol- radial strength, which was responsible for an increased rate of
lowed by oral anticoagulation with warfarin.8 This medical treat- restenosis and stent thrombosis. Later, Medtronic proposed
ment slightly decreased the risk of stent thrombosis, though a coil stent or Wiktor stent (Figure 3D). Finally, stents covered
it remained very high (occurring in 5% to 10% of cases). by a membrane of polytetrafluoroethylene were proposed

330 MEDICOGRAPHIA, Vol 38, No. 3, 2016 History and evolution of coronary stenting Bertrand
U P DAT E

Figure 3. Four types of stents: the precursors.


Coronary stents: the precursors A number of variations of the stent have been proposed. Shown
here are the (A) Wallstent, (B) Schatz-Palmaz, (C) Gianturco-
A Wallstent: self-expanding C Gianturco-Roubin Roubin, and (D) Wiktor stents.
All rights reserved.

The first drug-eluting stent of this group was the


Cypher (Cordis Corporation). Almost simultane-
ously proposed was paclitaxel, which inhibits cell
replication and is the drug delivered by the Taxus
B Schatz-Palmaz: tubular stents (Boston Scientific). Cypher and Taxus stents
D Wiktor stent: coil were the first-generation drug-eluting stents. These
were followed by second-generation (followed by
a second generation (Xience, Endeavor stents) and
third-generation drug-eluting stents, the latter be-
ing fully bioresorbable vascular scaffolds (BVS), in-
cluding: (Igaki-Tamai (Igaki Medical Planning), BVS
1.0 (Abbott Vascular), DESolve (Elxir Medical Cor-
poration), REVA (Reva Medical), ART 18AZ (Arterial
Remodeling Technologies), and Amaranth stents.

and used in saphenous vein graft stenoses to avoid the em- u Safety improvements in stenting: the fight against
bolization of the friable materials characteristic of these le- acute or subacute stent thrombosis (first revolution in
sions. It was also used for the emergency treatment of coro- coronary stenting)
nary perforations. It would take nearly 10 years to eliminate the frightening risk
of acute or subacute stent thrombosis. A number of strate-
Drug-eluting stents were introduced in 2000. They are com- gies were proposed; these included use of full-dose unfrac-
posed of two parts: the polymer coating the strut (one or sev- tionated heparin, low-molecular-weight heparin, dextran, sulfin-
eral layers) and the drug delivered into the vessel wall. The pyrazone, aspirin, and antivitamin K. The combination of these
drugs act on the cell cycle (Figure 4) and are able to suppress drugs was ineffective and led to significant bleeding at the
smooth muscle cell proliferation without toxicity and with a puncture site resulting in big hematomas that required vas-
low inflammatory risk. Most drug-eluting stents use an analog cular repair. In this context, although stenting was recognized
of sirolimus (drugs used from the limus group include sirolimus, to be effective for the treatment of abrupt periprocedural oc-
everolimus, zotarolimus, biolimus, the sirolimus metabolite no- clusion and to help avoid emergency bypass operations,
volimus, and myolimus, a macrocyclic lactone close to the many investigators were ready to abandon this technique
rapamycin family). for the treatment of restenosis. However, coronary stenting
would be resuscitated by two new findings by An-
tonio Colombo from Italy and Paul Barragan from
Marseille.
Limus family S phase
DNA
replcation Antonio Colombo, through extensive use of intra-
FKBP120
vascular ultrasound, demonstrated that in many
mTOR
cases, stent implantation was far from perfect, with
Paclitaxel
Low dose malapposition and insufficient deployment. From
these observations, he recommended inflation of
G0 phase G1 phase G2 phase the balloon at a higher level of pressure to improve
Cell works but Cell enlarges Preparation
is not actively and makes for division embedding of the stent inside the wall. With a larg-
replicating new proteins
er lumen and a better flow, stent thrombosis might
be avoided.
Paclitaxel
M phase High dose
Figure 4. Impact of the drugs delivered by
Cell division
drug-eluting stents on the cell cycle.
Abbreviations: G0 phase, resting phase of the cell cycle;
CELL DIVISION G1, G2 phases, interphases of the cell cycle; M phase, mitotic
phase of the cell cycle; mTOR, mammalian target of rapamycin.
All rights reserved.

History and evolution of coronary stenting Bertrand MEDICOGRAPHIA, Vol 38, No. 3, 2016 331
U P DAT E

Rutsch) launched a randomized study assessing the role of


coronary stenting in the treatment of restenosis. Although ini-
tially impossible to find support from industry for such a trial,
the tenacity of Serruys, de Jaegere, Kiemeneij and colleagues
led to the launch of the BENESTENT I trial (BElgian-NEther-
lands STENT) in Belgium and the Netherlands. This pilot study
included only 60 patients and took almost 1 year to complete
in four centers with a total number of almost 4000 interven-
tions, demonstrating the skepticism of the interventional car-
diology community for stenting.18 This landmark study demon-
strated the benefit of coronary stenting for the treatment of
restenosis. The results were confirmed by an American trial
(STRESS [STent REStenosis Study]).19 Later, the results were
complemented by the BENESTENT II study, which was per-
Figure 5. Rate of stent thrombosis in patients taking a dual an-
formed with a heparin-coated stent and showed that bleed-
tiplatelet treatment (aspirin + ticlopidine) compared with patients
taking aspirin + a vitamin K antagonist. ing complications almost completely disappeared.20
Abbreviations: ASA, acetylsalicylic acid; FANTASTIC, Full ANTicoagulation versus
ASpirin and TIClopidine (study); ISAR, Intracoronary Stenting and Antithrombotic However, over time, it appeared that in-stent restenosis was
Regimen; STARS, STent Anticoagulation Restenosis Study; Vit, vitamin.
Based on data from references 13-15. All rights reserved.
a true matter of concern. It was discovered that though the
scaffolding role of stenting prevented negative remodeling
Almost simultaneously, Paul Barragan reported that a dual an- the shrinkage of the arteryas a reaction to the metallic for-
tiplatelet treatment with aspirin and ticlopidine was success- eign body, the vascular wall generated a new, intense hyper-
ful.11 Barragan participated in a trial launched by Bertrand et plasia. Solinas et al21 showed the different aspects of in-stent
al (The TACT trial [Ticlopidine Angioplasty Coronary Trial]). restenosis, as follows: in-stent restenosis was focal (margin
The goal of this trial was to verify if an antiplatelet drug (ticlo- or mid-stent) in 42%, diffuse in 22%, proliferative (ie, with ex-
pidine) could prevent restenosis. This study presented at the tension beyond the stent) in 30%, and even occlusive in 6%.
American Heart Association meeting in 1992 had negative re- Several techniques were used to address this new problem;
sults but was able to demonstrate the benefits of ticlopidine these included redilatation by balloon (mainly for focal in-stent
+ aspirin in preventing acute periprocedural complications of restenosis), by the stent-in-stent technique, by Rotablator ther-
balloon angioplasty. Thus, Barragan continued to use this dual apy, and even by brachytherapy. At first glance, brachythera-
anti-platelet treatment after completion of the study. In perform- py seemed promising but was subject to numerous regula-
ing stent implantations, he was the only investigator to have tions due to the rules of radioprotection. This method was
no (or rare) cases of stent thrombosis. Finally, on the basis of never well accepted, was limited to a small number of centers,
a French registry collecting data on the usual strategies, it was and was finally abandoned with the emergence of drug-elut-
observed that the combination of aspirin and ticlopidine was ing stents.
markedly effective for reductions in stent thrombosis and vas-
cular complications.12 Three randomized studies performed u Drug-eluting stents (the second revolution in coronary
initially in Europe13,14 and later in the United States15 demon- stenting)
strated the benefit of this dual antiplatelet treatment (Figure 5). Over the years, several investigatorsparticularly Wim van der
Subsequently, Bertrand et al showed that another thienopy- Giessenattempted to coat the struts of the metallic stents
ridine (clopidogrel) was superior to ticlopidine (the CLASSIC tri- with different substances, the goal being to coat the struts
al [CLopidogrel Aspirin Stent International Cooperative study]).16 with a polymer as a drug-carrier vehicle; these drugs sought
The danger of acute/subacute thrombosis having been over- to inhibit the cell cycle. In July 1999, Cordis Corporation stud-
come, the so-called stentomania began, with an increasing ied a stent coated with a polymer releasing the drug rapa-
number of stent implantation procedures taking place. mycin. The company conducted a pilot study in Rotterdam,
the Netherlands with Patrick Serruys and in Sao Paulo, Brazil
u The fight against restenosis with Eduardo Souza, in which 15 patients were studied with
u The role of stenting angiographic and intravascular ultrasound follow-up. The two
In 1991, Serruys et al published in the New England Journal principal investigators were surprised to discover that at 4
of Medicine the results from the first 105 patients in the world and 12 months follow-up there was no evidence of neointimal
treated with the Wallstent.17 This report essentially addressed hyperplasia. These results led to a randomized clinical trial
the problem of subacute thrombosis, but few readers over- conducted in 237 patients treated by bare-metal stent or the
looked the fact that the rate of restenosis after stenting was Cypher, the first drug-eluting stentthe RAVEL study (RAn-
only 14%. In this context, Dutch investigators began a multi- domized study with the sirolimus-eluting VElocity balloon-ex-
center registry, and several investigators (Serruys, Bertrand, pandable stent in the treatment of patients with de novo na-

332 MEDICOGRAPHIA, Vol 38, No. 3, 2016 History and evolution of coronary stenting Bertrand
U P DAT E

tive coronary artery Lesions).22 This study, presented at the be divided into two categories: metallic stents that are mag-
European Society of Cardiology congress in Vienna (Septem- nesium based and those that are polymeric resorbable
ber 2001) by Marie-Claude Morice, was a new turning point more than 10 stents of this type have been studied, made of
in coronary interventional cardiology, as there was zero re- poly-L-lactic acid (PLLA) and poly-D,L-lactic acid. Absorb
stenosis. BVS (Abbott Vascular) is a fully resorbable stent which has
obtained the CE mark (Conformit Europenne [European
Subsequently, two major trials confirmed these excellent re- conformity]); the ABSORB II trial published in the Lancet in
sults: the SIRIUS trial (SIRolImus-elUting Stent in coronary 2014 compared Absorb BVS vs the Xience metallic drug-
lesions),23 as well as the TAXUS trial (Treatment of de novo eluting stent in a cohort of 501 patients.27-29 At follow-up, there
coronAry disease using a single paclitaXel-elUting Stent),24 was no significant difference in terms of safety and efficacy
which used another eluting drug, paclitaxel. Later, the course between the two devices. Currently, there is great interest for
of drug-eluting stents was disturbed by a frighten-
ing issueat the 2006 European Society of Car-
Before balloon angioplasty
diology congress in Vienna, results suggesting late
stent thrombosis were presented. Fortunately, these Prevention of
Lumen restenosis
scary results were not confirmed,25 but they led to
with stents
a still ongoing debate about the duration of dual
antiplatelet treatment: 6 months, 12 months, or

BMS Stent Drug-eluting stent
more? Nevertheless, the development of drug-elut- After balloon angioplasty
Prevention of shrinkage Prevention of shrinkage
ing stents continued and with the new drug-elut- and hyperplasia
ing stents using a different drug-carrier vehicle, all of
them have been shown to offer efficacy and safety.

Thus, the challenges were overcome: the cage pre- Restenosis


Hyperplasia and
vents negative remodeling and the eluted drug pre- negative remodeling
vents cell proliferation and migration (Figure 6). A fi-
nal step was the development of stents that used
biodegradable polymers and bioresorbable scaf-
folds: drug-eluting stents with biodegradable poly-
mers provide the benefit of drug-eluting stents in Figure 6. Summary of the relationship between restenosis and stenting.
the early days/months and of the bare-metal stents Whereas balloon angioplasty often results in negative remodeling and sometimes in restenosis,
later. A number of prostheses were studied: AX- the use of stents prevents negative remodeling. Drug-eluting stents can provide this benefit
and also prevent cell proliferation and migration.
XESS (Biosensors), Orsiro (Biotronik), DESyne (Elixir), Abbreviation: BMS, bare-metal stent.
Infinium (Sahajanand), Biomine (Meril Life). All rights reserved.

As the polymer may induce side effects, polymer-free drug- these new bioresorbable stents, but it is obvious that a longer
eluting stents have been proposed. The eluting drug may be follow-up is needed in order to reach final conclusions. There
introduced into a microporous surface on metallic stents. Ex- are currently a number of studies underway evaluating these
amples include the Yukon stent (Translumina), BioFreedom new devices.
(Biosensors), VESTAsync (MIV Therapeutics), Nano (Xience),
and Bicare (Lepu Medical). In other examples, the Optima Conclusion
stent (CID Vascular) proposes small reservoirs of tacrolimus Coronary stenting represents one the most important ad-
covered with carbofilm, and the Amazonia PAX stent (MIN- vances in the field of coronary angioplasty. With this technique,
VASYS) is a cobalt-chromium stent coated with paclitaxel. percutaneous coronary interventions are safe in most cases
and the risk for patients to be sent for surgery for emergency
Bioresorbable stents are very promising: they offer the vascu- bailout procedures has become minimal. Additionally, the risk
lar scaffold for a certain amount of time and then the implant- of reintervention after angioplasty is markedly reduced after
ed materials are progressively resorbed. This offers a number drug-eluting stent implantation. With coronary stenting, coro-
of advantages, including the elimination of foreign bodies in- nary interventional procedures, as minimally invasive tech-
side the wall, restoration of endothelial coverage, and possi- niques, have become the primary method of myocardial revas-
bly restoration of vasomotion. These biodegradable stents can cularization in man.

Keywords: coronary stenting; interventional cardiology; myocardial revascularization; percutaneous coronary angioplasty

History and evolution of coronary stenting Bertrand MEDICOGRAPHIA, Vol 38, No. 3, 2016 333
U P DAT E

References
1. Gruntzig A. Transluminal dilatation of coronary-artery stenosis. Lancet. 1978;1 pirin after coronary stenting: the clopidogrel aspirin stent international coop-
(8058):263. erative study (CLASSICS). Circulation. 2000;102(6):624-629.
2. Simpson JB, Baim DS, Robert EW, Harrison DC. A new catheter system for 17. Serruys, PW, Strauss BH, Beatt KJ, et al. Angiographic follow-up after place-
coronary angioplasty. Am J Cardiol. 1982;49(5):1216-1222. ment of a self-expanding coronary-artery stent. N Engl J Med. 1991;324(1):
3. Bonzel T, Wollschlager H, Just H. A new catheter system for the mechanical 13-17.
dilatation of coronary stenoses with exchangeable intracoronary catheters, fast 18. Serruys PW, de Jaegere P, Kiemeneij F, et al. A comparison of balloon-expand-
flow of the contrast agent and improved control [in German]. Biomed Tech (Berl). able-stent implantation with balloon angioplasty in patients with coronary ar-
1986;31(9):195-200. tery disease. Benestent Study Group. N Engl J Med. 1994;331(8):489-495.
4. Carrel A. Permanent intubation of the thoracic aorta. J Exp Med. 1912;16(1): 19. Fischman DL, Leon MB, Baim DS, et al. A randomized comparison of coronary-
17-24. stent placement and balloon angioplasty in the treatment of coronary artery
5. Dotter CT, Judkins MP. Transluminal treatment of arteriosclerotic obstruction. disease. Stent Restenosis Study Investigators. N Engl J Med. 1994;331(8):
Description of a new technic and a preliminary report of its application. Circu- 496-501.
lation. 1964;30:654-670. 20. Serruys PW, van Hout B, Bonnier H, et al. Randomised comparison of implan-
6. Palmaz JC, Sibbitt RR, Reuter SR, Tio FO, Rice WJ. Expandable intraluminal tation of heparin-coated stents with balloon angioplasty in selected patients
graft: a preliminary study. Work in progress. Radiology. 1985;156(1):73-77. with coronary artery disease (BENESTENT II). Lancet.1998;352(9129):673-681.
7. Puel J, Joffre F, Rousseau H, et al. Self-expanding coronary endoprosthesis in 21. Solinas E, Dangas G, Kirtane AJ, et al. Angiographic patterns of drug-eluting
the prevention of restenosis following transluminal angioplasty. Preliminary clin- stent restenosis and one-year outcomes after treatment with repeated percu-
ical study [in French]. Arch Mal Coeur Vaiss. 1987;80(8):1311-1312. taneous coronary intervention. Am J Cardiol. 2008;102(3):311-315.
8. Sigwart U, Puel J, Mirkovitch V, et al. Intravascular stents to prevent occlusion 22. Morice MC, Serruys PW, Sousa JE, et al; RAVEL study group. A randomized
and restenosis after transluminal angioplasty. N Engl J Med. 1987;316(12):701- comparison of a sirolimus-eluting stent with a standard stent for coronary revas-
706. cularization. N Engl J Med. 2002;346(23):1773-1780.
9. Palmaz JC, Richter GM, Nldge G, et al. Intraluminal Palmaz stent implantation. 23. Stone GW, Moses JW, Ellis SG, et al. Safety and efficacy of sirolimus- and
The first clinical case report on a balloon-expanded vascular prosthesis [in Ger- paclitaxel-eluting coronary stents. N Engl J Med. 2007;356(10):998-1008.
man]. Radiologe. 1987;27(12):560-563. 24. Stone GW, Ellis SG, Cox DA, et al; TAXUS-IV Investigators. A polymer-based,
10. Macander PJ, Agrawal SK, Roubin GS. The Gianturco-Roubin balloon-expand- paclitaxel-eluting stent in patients with coronary artery disease. N Engl J Med.
able intracoronary flexible coil stent. J Invasive Cardiol. 1991;3(2):85-94. 2004;350(3):221-231.
11. Barragan P, Pietri P, Villain P, Silvestri M, Roquebert PO. Antiplatelet therapy 25. Kastrati A, Mehilli J, Pache J, et al. Analysis of 14 trials comparing sirolimus-
during coronary endoprosthesis placement [in French]. Arch Mal Coeur Vaiss. eluting stents with bare-metal stents. N Engl J Med. 2007;356(10):1030-1039.
1996;89(11 suppl):1515-1520. 26. Windecker S, Remondino A, Eberli FR, et al. Sirolimus-eluting and paclitaxel-
12. Karrillon GJ, Morice MC, Benveniste E, et al. Intracoronary stent implantation eluting stents for coronary revascularization. N Engl J Med. 2005;353(7):653-
without ultrasound guidance and with replacement of conventional anticoag- 662.
ulation by antiplatelet therapy. 30-Day clinical outcome of the French multi- 27. Diletti R, Farooq V, Girasis C, et al. Clinical and intravascular imaging out-
center registry. Circulation. 1996;94(7):1519-1527. comes at 1 and 2 years after implantation of ABSORB everolimus eluting biore-
13. Schmig A, Neumann FJ, Kastrati A, et al. A randomized comparison of anti- sorbable vascular scaffolds in small vessels. Late lumen enlargement: does
platelet and anticoagulant therapy after the placement of coronary-artery stents. bioresorption matter with small vessel size? Insight from the ABSORB cohort
N Engl J Med. 1996;25;334(17):1084-1089. B trial. Heart. 2013;99(2):98-105.
14. Bertrand ME, Legrand V, Boland J, et al. Randomized multicenter comparison 28. Diletti R, Serruys PW, Farooq V, et al. ABSORB II randomized controlled trial:
of conventional anticoagulation versus antiplatelet therapy in unplanned and a clinical evaluation to compare the safety, efficacy, and performance of the Ab-
elective coronary stenting. The Full Anticoagulation Versus Aspirin and Ticlo- sorb everolimus-eluting bioresorbable vascular scaffold system against the
pidine (FANTASTIC) study. Circulation. 1998;98(16):1597-1603. XIENCE everolimus-eluting coronary stent system in the treatment of subjects
15. Leon MB, Baim DS, Popma JJ, et al. A clinical trial comparing three anti- with ischemic heart disease caused by de novo native coronary artery lesions:
thrombotic-drug regimens after coronary-artery stenting. Stent Anticoagulation rationale and study design. Am Heart J. 2012;164(5):654-663.
Restenosis Study Investigators. N Engl J Med. 1998;339(23):1665-1671. 29. Dudek D, Onuma Y, Ormiston JA, Thuesen L, Miquel-Hebert K, Serruys PW.
16. Bertrand ME, Rupprecht HJ, Urban P, Gershlick AH; CLASSICS Investigators. Four-year clinical follow-up of the ABSORB everolimus-eluting bioresorbable
Double-blind study of the safety of clopidogrel with and without a loading dose vascular scaffold in patients with de novo coronary artery disease: the ABSORB
in combination with aspirin compared with ticlopidine in combination with as- trial. EuroIntervention. 2012;7(9):1060-1061.

HISTOIRE ET VOLUTION DE L IMPLANTATION DES ENDOPROTHSES CORONAIRES


Limplantation des endoprothses coronaires est une tape majeure dans lhistoire de langioplastie coronaire per-
cutane. Jacques Puel a ralis la premire implantation dendoprothse chez lhomme en 1986 et la recherche dans
ce domaine sest dveloppe immdiatement. La premire crainte concernant langioplastie par ballonnet tait la s-
curit, cause du risque docclusion brutale pendant lintervention. Le risque, et donc le besoin dun pontage en ur-
gence, sont considrablement rduits par limplantation dendoprothses. Nanmoins, les mdecins ont t confron-
ts un autre problme : le risque de thrombose de lendoprothse. Ce risque peut, cependant, tre limin grce
un double traitement antiplaquettaire. Limplantation dendoprothses coronaires joue maintenant un rle majeur
dans le combat contre la restnose, les endoprothses libration de principes actifs rduisant considrablement
ce risque, mme chez les patients haut risque (les diabtiques). Avec limplantation dendoprothses, langioplas-
tie coronaire est dsormais la principale mthode de revascularisation myocardique.

334 MEDICOGRAPHIA, Vol 38, No. 3, 2016 History and evolution of coronary stenting Bertrand
Medicographia
A Ser vier publication

I nternational Advisory Committee


Argentina China (PRC) Germany Kazakhstan
Mr G. RENACCO Mr D. DARGENT Mr O. KIRST Mr O. NEGULYAEV
Servier Argentina S.A. Servier (Tianjin) Pharmaceu- Servier Deutschland GmbH Les Laboratoires Servier
Av. del Libertador 5926, Piso 8 tical Co., Beijing Office Elsenheimerstr. 53 Representative Office
C1428ARP - Buenos-Aires 6th Floor, West Building 80687 Mnchen 310 G DOSTYK av, 3rd Floor
Tel: (54-11) 4706 5800/5801 World Financial Centre Tel: (49-89) 5709501 050 020 Almaty
No. 1 East 3rd Ring, Tel: 00 7 727 386 76 62
Armenia Middle Road Greece
Mr G. VELIJANYAN Chaoyang District Mr F. FOUILLOUX Korea
Les Laboratoires Servier Beijing 100020 P.R.C. Servier Hellas Ms C. ROUCHS
Representative Office Tel: 86 10 65610341 72, Ethnikis Antistaseos & Servier Korea Ltd, 5th Floor,
Hyusisain Ave. Agamemnonos str. 215, Seochojungang-ro
1 Building, 5th floor, Colombia 152 31 Chalandri Seocho-gu
office N13 Mr R. FEDRIGO Tel: (302-10) 9391000 Seoul 137-802
NORD Business Centre Laboratorios Servier Tel: 0077 27386763
0001 - Yerevan de Colombia SA Gulf Countries
Tel: +374 10 505074 Edificio Centro Empresarial Mr P. PEREZ Latvia
98 x 28, Piso 4 Les Laboratoires Servier Dr J. LEJA
Australia Transversal 19A - N 98-28 Representative Office SIA Servier Latvia
Mr F. PARISOT Bogot D.C. API World Tower 8th, Flat 801 Duntes Nami, Duntes Street 11
Servier Laboratories Pty Ltd Tel. : (57.1) 742.9369. Sheikh Zayed Road - Dubai LV1013 Riga
Servier House, Tel: 971 4 332 99 03 Tel: 371 6750 20 39
8 Cato Street Croatia
Hawthorn Mr A. MALLET Hong Kong Lithuania
Victoria 3122 Servier Pharma d.o.o. Mr L. GARNIER Dr M. PAPIEVIS
Tel: (61-3) 88237333 Tuskanova 37 Servier Hong Kong Ltd UAB Servier Pharma
10000 Zagreb Room 4201-03, 42/F Europa Business Center
Austria Tel: 385 1 3016 222 248 Queens Road East Konstitucijos av 7
Mrs B. FRITSCHE-SURCHAT Wanchai, Hong Kong LT09308 Vilnius
Servier Austria GmbH Czech Republic Tel: 852 2577 1922 Tel: 370 52 638 615
Mariahilferstrasse 20/5 Mr P. BOYER
1070 Wien Servier SRO Hungary Luxembourg
Tel: (43-1) 524399900 Florentinum Mr J. F. KESSELHUT Mr A. BRETHOUS
Na Florenci 2116/15 Servier Hungaria KFT Servier Luxembourg SA
Azerbaijan 110 00 Praha 1 Westend Office 3a, rue Guillaume Kroll
Dr R. TAHMAZOV Tel: (420-2) 22118602 B tower, 3rd floor, Vci t 1-3 1882 Luxembourg Gasperich
Servier Azerbaijan LLC Budapest 1062 Tel: (352) 49 35 35
3th Floor, Hyatt Tower II Denmark Tel: 36 1 238 7799
1033, Izmir Street Mr F. TEXIER Malaysia
Baku AZE 1 065 Servier Danmark A/S India Dr K. PANSE
Tel: 00 994 12 490 79 00 Roskildevej 39 A Mr Y. GIRARD Servier Malaysia SDN BHD
2000 Frederiksberg Serdia Pharmaceuticals Lot 1301, Level 13,
Belarus Tel: (45-36) 442260 (India Pvt) Ltd, Serdia House Uptown 2
Dr O. MULYARCHIK Off Dr S.S. Rao Road, Parel 2 Jalan SS21/37
Les Laboratoires Servier Egypt Mumbai 400 012 Damansara Uptown
70, Myasnikova Street Mr G. CHARLES Tel: 91 22 24198000 47400 Petaling Jaya
220 030 Minsk Servier Egypt, Selangor Darul Ehsan
Tel: 00 375 17 306 54 51 Scientific Office Indonesia Tel: 603 772 638 66
67, El Horreya St. Mr A. NEROT
Belgium Heliopolis PT Servier Indonesia Mexico
Mr A. BRETHOUS P.O. Box 123 Heliopolis Menara Kadin Indonesia Mr F. DARCHEZ
Servier Benelux SA Tel: (20) 222 90 60 77 18th Floor Laboratorios Servier de
Riverside Business Park JL. H.R. Rasuna Said Blok Mxico S.A. de C.V.,
Boulevard International No. 57 Estonia X-5 Kav. 2-3 Palmas 830
1070 Bruxelles Mr F. de KERTANGUY Jakarta 12950 Lomas de Chapultepec
Tel: (32-2) 5294311 Servier Laboratories O Tel: (62-21) 57903940 C.P. 11 000 - Mxico D.F.
Rotermanni 8 Tel: (52-55) 52 02 33 59
Brazil Tallinn 10111 Ireland
Mr C. SABATHIER Tel: 372 640 00 07 Mr F. DRUGUET Morocco
Laboratorios Servier do Brasil Servier Laboratories Ltd Mr J. Y. GAL
Ltda, Estrada dos Finland Block 2 Servier Maroc
Bandeirantes Mr F. TEXIER West Pier Business Campus Immeuble ZEVACO
4211 - Jacarepagua Servier Finland OY Old Dunleary Road Lotissement Fath 4
Rio de Janeiro, RJ Ayritie 12A Dun Laoghaire, Co. Dublin Route dAzemmour
CEP: 22-775-113 01510 Vantaa Tel: (353-1) 6638110 Casablanca 20180
Tel: (55-21) 2142-1414 Tel: 358 9 279 8080 Tel: +212(0) 522 79 52 52
Italy
Bulgaria France Mr F. DEBAILLON-VESQUE Netherlands
Mr A. BRETON Mr P. ETORRE Servier Italia SpA Mr D. MURPHY
Servier Medical EOOD Les Laboratoires Servier Via Luca Passi, 85 Servier Nederland Farma BV
14 blvd Tzar Osvoboditel 50, rue Carnot 00166 Roma Kanaalpark 140 - 2321 JV Leiden
1000 Sofia 92284 Suresnes Cedex Tel: (39-06) 669 081 Tel: (31-71) 5246700
Tel: 359 87 71 57 00 Tel: 33 1 55 72 60 00
Japan Pakistan
Canada Georgia Mr E. DELARGE Dr R. MAJEED
Mr F. FASANO Dr M. KHETSURIANI Nihon Servier Company Ltd Servier Research &
Servier Canada Inc. LLS Representative Office Hongo MK Building 5F Pharmaceuticals, (Pakistan)
235, boulevard Armand Frappier 44 Kote Apkhazi Street 1-28-34, Hongo, Bunkyo-Ku PVT Ltd, 65 Main Boulevard
Laval, Qubec H7V 4A7 0105 Tbilisi 113-0033 Tokyo Gulberg - Lahore
Tel: (1-450) 9789700 Tel: +995 32 243 93 90 Tel: (81-3) 5842 7111 Tel: (92-42) 5879500
Medicographia
a Servier publication

Editor in Chief: Frdric Sesini, PharmD


Medicographia
A Servier publication
Editorial Board: Antoine Beillevaire, MD;
Christophe Charpentier, MD; Philippe
Gonnard, MD; Jean-Christophe Guillerm,
MD; Philippe Malet, DVM; Bernard
Parrin, PharmD; Pascal Poullali, MA;
Hugues Renaut, PharmD; Marion
Schrenk, MD; Franois Vilette, PharmD

Medical Publications Editor:


Catriona Donagh, BAppSc
Philippines Slovenia United Kingdom
Production Editors: Mr P. tte Mr a. MaLLet dr a. LaLLouette
Emmanuelle Pinjon-Casu, PhD Servier Philippines, inc. Servier Pharma d.o.o. Servier Laboratories Ltd
(emmanuelle.pinjon-casu@servier.com) 2 orion Cor. Mercedes St. Pot K Sejmiscu 33 rowley Building, Wexham Springs
Bel air village - Makati 1231 Ljubljana Framewood road
John Plant, BPharm tel: 63-2 897 89 90 to 97 Crnuce Wexham Slough
(john.plant@servier.com) tel: 386 1 56 34 811 Berkshire SL3 6rJ
Poland tel: (44-1753) 662744
dr J. dreWLa South Africa
Editorial Assistant: Sophie Gehrisch Servier Polska Sp. z.o.o. Mr S. SPeLLer Uzbekistan
ui. Jana Kazimierza 10 Servier Laboratories Pty Ltd Mrs S. aSKarova
01-248 Warsaw n4 Country Club estate representative office of
Design: Studio DTC tel: (48-22) 594 90 00 Woodlands drive Les Laboratoires Servier in
Layout: Myriam Bucquoit Woodmead 2191 the republic of uzbekistan
Portugal Gauteng Block C, 6th Floor
and Bernard Crespin dr a. MendeS tel: 27 - 081 700 900 international Business Center
Servier Portugal 107B, amir temur avenue
Medicographia is published avenida antonio augusto Spain 100084 tashkent
de aguiar, 128 Mr S. MaSCarau tel: 99 871 140 7997
by Les Laboratoires Servier 1069-133 Lisboa Laboratorios Servier Sa
50, rue Carnot 92284 Suresnes tel: (351-21) 3122000 avenida de los Madroos 33 Venezuela
Cedex France, and printed 28043 Madrid dr e. LanGHaMer
Republic of Panama tel: (34-91) 7489630 Laboratorios Servier, S.a.
by Imprimerie Drid Mr a. PriGent Calle Sanatorio del vila
Zone Industrielle des Chanoux Servier Centro amrica y Sweden Centro empresarial Ciudad Cen-
Caribe, S.a. Mr F. texier ter, torre F, piso 3
49, rue des Frres-Lumire Costa del este Servier Sverige aB Boleta norte - Caracas 1071 a
93334 Neuilly-sur-Marne Cedex Boulevard Costa del este Frsundaviks all 1 tel: 58212718 71 00
edif. PH torre Panam 16970 Solna
Piso 25, Ciudad de Panam tel: +46 854 25 0800 Vietnam
repblica de Panam. Mr M. FitouSSi
tel.: 507.301.02.27 Switzerland Servier vietnam Company Limited
Printed in France Mr F. BenGoLd Level 11, Continental tower,
Directeur de la Publication: Romania Servier Suisse Sa 85 Ham nghi Street,
Mr S. niCoLLerat 10 rue de la Bergre nguyen thai Binh Ward,
Frdric Sesini, PharmD Servier Pharma, S-Park Case Postale 380 district 1, Ho Chi Minh City
11-15 tipografilor str. CH-1217 Meyrin 1 tel: 84 8 3823 8932
Building a1, 3rd floor, sector 1 tel: (41-22) 7378437
Bucarest code 013714 West Indies
tel: (40) 21 528 52 80 Taiwan (ROC) dr t. Monod
2016 by Les Laboratoires Servier Mr r. Lo Servier West indies Ltd
Russian Federation taiwan Servier (S) Pte Ltd Stafford House
All rights reserved throughout the world Mr J. Gavet 3rd Floor, no. 168 the Garrison, St Michael Barba-
Zao Servier Sung Chiang road dos
and in all languages. No part of this pub- 2 Paveletskaya Pl., Bt. 3 taipei 104 tel: (1-246) 4277667
lication may be reproduced, transmitted, 115054 Moscow - russia tel: 886 2 2568 1380
or stored in any form or by any means tel: (7-495) 937 07 00
Thailand
either mechanical or electronic, including Saudi Arabia Mr G. drianno
photocopying, recording, or through an dr B. renaud Servier thailand Ltd
Les Laboratoires Servier Ploenchit Center Building
information storage and Scientific office, 15th Floor, 2 Sukhumvit road
retrieval system, without the written per- 401 abdullatif Plaza, Klongtoey - Bangkok 10110
mission of the copyright holder. al Faisaliyah dist., tel: 66 2 656 8388
King Fahd St.,
Opinions expressed do not necessarily P.o. Box 112744 Tunisia
reflect the views of the publishers, Jeddah 21371 Mr F. BarBu
tel: 966 2 697 69 97 Servier tunisie
editors, or editorial board. The authors, rue du Lac de Cme
editors, and publishers cannot be held Serbia immeuble driss 4
responsible for errors or for any conse- Mr C. BroudiC Bloc a, 1er tage
Les Laboratoires Servier Les Berges du Lac
quences arising from the use of the Bulevar Mihajla Pupina 10L 1053 tunis
information contained in this journal. 11070 Belgrade tel: +216 71655 857
tel: 381 11 424 00 50
Turkey
Address for correspondence: Singapore Mr P. Mea
Servier International / Mr B. aBdiLi Servier ila ve arastirma aS
Servier (S) Pte Ltd Maslak Giz Kule
Medical Publishing Division 67 ubi avenue 1 Meydan Sokak
50, rue Carnot, #06-08 Starhub Green no 28, Kat: 22-23
92284 Suresnes Cedex FRANCE Singapore 408942 34398 Maslak, istanbul
tel: 65 6509 9811 tel: (90-212) 3291400
Tel. + 33 1 55 72 60 00 / Fax 58 84
Slovak Republic Ukraine
Mr r. aZenCotH Mr M. eSCHenBrenner
Servier Slovensko ro Les Laboratoires Servier
Spol S.r.o., Pribinova 10 24 vorovskogo Street
81109 Bratislava 4th floor - Kiev 01054
tel: 421 2 59204111 tel: 380 44 490 34 41
Medicographia
Vol 38, No. 3, 2016 ISSN 0243-3397

Metabolic agents in the contemporary treatment of angina


C hance
Medicographia
favors only
the prepared mind Metabolic agents
Pa s t e u r in the contemporary
treatment of angina

Vol 38, No. 3



Pages 243-336
16 MP 0128 X Printed in France

Servier - 50, rue Carnot - 92284 SureSneS Cedex (FranCe) Quarterly Thematic Journal

of Clinical and Therapeutic Research


www.medicographia.com
2016

www.servier.com
A Servier Publication

You might also like