Restenosis Following Coronary Angioplasty: Current Status

Article information

Korean J Intern Med. 2001;16(2):51-55
From the Cardiac Catheterization Laboratory, New York Presbyterian Hospital - Cornell University, New York, NY
Address reprint requests to : Mun K. Hong, M.D., Director, Cardiovascular Intervention and Research, Cornell University - New York Presbyterian Hospital 520 E. 70th St. Starr Pavilion 4, New York, NY 10021 mkh2003@med.cornell.edu

INTRODUCTION

Since introduction almost 25 years ago by Dr. Andreas Gruentzig, percutaneous transluminal coronary angioplasty (PTCA) has established itself as a viable non-surgical treatment option for patients with symptomatic coronary artery disease. With improvement in operation techniques and angioplasty equipments, especially with the availability of stent, the PTCA procedural success rate approaches over 95% in most lesions and complication rates around 1%. This remarkable procedural success, however, has not translated into sustained long-term outcomes. Restenosis or renarrowing at the site of successful PTCA has been the major limitation of this therapy since its inception. However, we now understand the mechanisms responsible for the restenosis process and have made tremendous progress in reducing the restenosis rates (Table 1). This review paper will highlight these strategies to combat restenosis.

Mechanisms of restenosis and potential treatments

RESTENOSIS: PAST

In the era of balloon angioplasty, restenosis was a frequent event, occurring in approximately 50% of patients by follow-up angiography and in about 30% of patients clinically within six months following a successful procedure1,2). The mechanism of restenosis, based on limited animal studies in normal vessels and human autopsy findings, was felt to be initial thrombus formation and subsequent neointimal hyperplasia36). This partially correct assumption led to numerous pharmacological studies targeting reduction in thrombus deposition or neointimal hyperplasia. These investigations were initially conducted in animal models of overstretch balloon injury and when the results were positive, subsequently in patients. Although the animal studies were in general encouraging, the results of randomized clinical studies were uniformly disappointing7). The negative results were then felt to be a dosing issue, as the weight-adjusted drug dosage was at least a log order higher in animals. Similar amounts in patients would have resulted in severe systemic side effects. Thus, there was an intense interest and research into local drug delivery. The hypothesis was that the amount of drug necessary for preventing restenosis if given systemically would be toxic to the patients, but a smaller amount of the drug delivered at the site of angioplasty using these local drug delivery catheters would provide greater efficacy8). This approach also failed, because the drug delivery was inefficient (less than one percent of the drug was successfully delivered to the vessel wall) and the deposited drug did not stay around for a sufficient period of time for efficacy. More importantly, these therapies were based on incorrect assumption regarding the predominant mechanism of restenosis. Finally, the initial suboptimal results, not too infrequent after balloon angioplasty, also contributed to the high restenosis rates. Thus, new angioplasty devices, such as directional coronary or rotational atherectomy catheters, were developed with the hope of modifying the plaque and reducing restenosis with a mechanical approach911). The results of randomized trials comparing the new angioplasty devices versus balloon angioplasty were also negative12,13). Not until the seminal intravascular ultrasound (IVUS) observations of chronic geometric remodeling (vessel shrinkage at the site of angioplasty) being the predominant mechanism of non-stent angioplasty restenosis14) did we begin to understand how to address the prevention of restenosis in a logical manner (Figure 1). It became clear that a mechanical approach with vessel scaffolding, such as the metallic stents, was the only way to prevent chronic vessel shrinkage. Indeed, the randomized trials comparing the Palmaz-Schatz stents versus balloon angioplasty were the first clinical restenosis studies to provide positive results15,16). However, even these encouraging results showed that stents reduce but do not eliminate restenosis. Subsequent studies in unfavorable lesions, such as small vessels (<3.0 mm in diameter) or diffuse lesions (>10 mm in length), showed that stents are still associated with high restenosis rates, not too dissimilar from those after balloon angioplasty17,18). Furthermore, IVUS observations in patients documented that in-stent restenosis is purely neointimal hyperplasia19), as stents do not recoil over time (Figure 2). Thus, investigators have attempted to define the variables associated with in-stent restenosis. These studies, especially with the aid of IVUS, showed that ostial location, pre-intervention plaque burden and post-intervention lumen dimensions assessed by IVUS20), and diabetes mellitus21) were predictors of eventual in-stent restenosis. Thus, the stent implantation techniques evolved to high-pressure adjunct balloon angioplasty to optimize the initial stent implantation22). However, refined techniques still cannot modify the restenosis rates at the ostial location or in diabetic patients. Furthermore, for those lesions not amenable to stent implantation, the restenosis rate is still unacceptably high.

Figure 1.

Angiographic (top panels) and intravascular ultrasound (bottom panels) findings in a patient with ostial left anterior descending coronary artery lesion (Left panels). The lesion was successfully treated with directional coronary atherectomy (DCA; Middle panels). However, seven months later, the patient returned with recurrent symptoms and restenosis (Right panels). Intravascular ultrasound demonstrated chronic geometric remodeling as the predominant mechanism of restenosis rather than neointimal hyperplasia. (Reproduced by permission from reference 7).

Figure 2.

The ostial right coronary artery lesion was successfully treated with a Palmaz-Schatz stent (JJIS; Left panels). When the patient returned with restenosis (Right panels), intravascular ultrasound showed no change in stent dimension but severe neointimal hyperplasia as the cause of the in-stent restenosis. (Reproduced by permission from reference 7).

RESTENOSIS: PRESENT

It is clear that stents are the only angioplasty device to have an impact on restenosis. However, there are still unfavorable lesion subsets, such as small vessels, diffuse lesions, ostial lesions, saphenous vein graft lesions and diabetic population whose restenosis rates have not been altered by stents. Especially for these lesions, new approaches, such as drug-coated stents and intravascular brachytherapy, are actively investigated to reduce restenosis.

The most exciting development has been the drug-coated stents. The rationale for this approach is that the metallic stent would provide the scaffolding to prevent geometric remodeling and that the antiproliferative agent impregnated on the stent platform would prevent neointimal hyperplasia, thus addressing both mechanisms of restenosis with the same device. Despite the theoretical difficulties of finding biocompatible polymers for drug impregnation, drugs effective at low dose, and unknown pharmacokinetics, there have been encouraging animal studies23,24). Based on these results, the rapamycin-coated stents have been studied in 30 patients with de novo single vessel disease25). The results of this preliminary registry study have been astounding, with no angiographic or clinical restenosis at one-year follow-up. This stent as well as other drug-eluting stents, such as paclitaxel-coated stents and actinomycin-eluting stents, are undergoing randomized trials against bare metal stents to determine the efficacy of this approach for the prevention of restenosis. Whether this therapy would be effective in unfavorable lesions is not known.

Another novel approach, which has been more successful in treating the in-stent restenosis than the prevention of restenosis for de novo lesions, is the intravascular brachytherapy26). The mechanism of brachytherapy is that the ionizing radiation would prevent the dividing cells, such as the smooth muscle cells causing the neointimal hyperplasia, from proliferation by damaging the nucleus of the cells. Several randomized trials have documented the beneficial effect of this approach with both gamma2628) and beta radiation in reducing the recurrent in-stent restenosis29). The preliminary result of the beta radiation for prevention of restenosis following angioplasty of de novo lesions has been discouraging30). However, whether gamma radiation or different beta system would prevent the initial restenosis, especially in combination with stents, needs further evaluation.

RESTENOSIS: FUTURE

Future efforts should try to prevent restenosis completely rather than merely reduce the rates. This may involve a combination of mechanical devices and adjunct pharmacology. The restenosis process is a culmination of multiple factors, including the initial thrombus formation, inflammation, geometric remodeling and neointimal hyperplasia. Potent pharmacological agents could prevent the first two processes, stents would prevent the third event and effective antiproliferative agents, ideally from the stent platform, could address the last process. Potentially more effective drug-eluting stent platform may be drug impregnation of stent grafts, as the graft material would allow impregnation of the drug in a much larger area (close to 90% of the lesion area versus 15–20% for bare metal stents). This device would have limited use in native coronary arteries, however, due to the presence of many side branches and the occlusion of these branches by the graft material.

Alternatively, the angioplasty procedure may have to prevent only the recurrence of ischemia in the target vessel, even if the angioplasty site should become renarrowed. This approach may involve the drug-eluting stent and administration of antigenic growth factors or gene therapy to myocardium supplied by the treated vessel31). Thus, even if the stent should become restenosed, the angiogenesis in the target area may prevent the occurrence of ischemia and the need for repeat intervention.

CONCLUSIONS

The restenosis process is an uncontrolled progression of the natural healing process. Thus, therapies aimed at allowing the necessary healing to occur (i.e., re-endothelialization) and yet preventing the excess scarring (both adventitial and intimal) could prevent restenosis. With rapidly evolving technologies and continuing research, restenosis will be prevented in the majority of patients in the near future and the minority with restenosis still may benefit from a combination therapy aimed at preventing ischemia, not necessarily restenosis.

References

1. Serruys PW, Luijten HE, Beatt KJ, Geuskens R, Feyter PJ, van den Brand M, Reiber JH, ten Kanten UJ, Van Es GA, Hugenholtz PG. Incidence of restenosis after successful coronary angioplasty: a time-related phenomenon. Circulation 77:361–371. 1988;
2. Nobuyoshi M, Kimura T, Nosaka H, Mioka S, Heno K, Yokoi H, Hamasaki N, Horiuchi H, Ohishi H. Restenosis after successful percutaneous transluminal coronary angioplasty: serial angiographic follow-up of 229 patients. J Am Coll Cardiol 12:616–622. 1988;
3. Steele PM, Chesbro JH, Stanson AW, Holmes DR Jr, Dewaujee MK, Badimon L, Fuster V. Balloon angioplasty: natural history of the pathophysiological response to injury in a pig model. Circ Res 57:105–112. 1985;
4. Schwartz RS, Murphy JG, Edwards WD, Camrud AR, Vliestra RE, Holmes DR. Restenosis after balloon angioplasty: a practical proliferative model in porcine coronary arteries. Circulation 82:2190–2200. 1990;
5. Austin GE, Ratliff NB, Hollman J, Tabei S, Phillips DF. Intimal proliferation of smooth muscle cells as an explanation for recurrent coronary artery stenosis after percutaneous transluminal coronary angioplasty. J Am Coll Cardiol 6:369–375. 1985;
6. Farb A, Virmani R, Atkinson JB, Klodgie FD. Plaque morphology and pathologic changes in arteries from patients dying after coronary balloon angioplasty. J Am Coll Cardiol 16:1421–1429. 1990;
7. Hong MK, Mehran R, Mintz GS, Leon MB. Restenosis after coronary angioplasty. Current Problems in Cardiology 22:1–36. 1997;
8. Lincoff AM, Topol EJ, Ellis SG. Local drug delivery for the prevention of restenosis: fact, fancy, and future. Circulation 90:2070–2084. 1994;
9. Hinohara T, Rowe MH, Robertson GC, Selmon MR, Brader L, Leggett JH, Vetter JW, Simpson JB. Effect of lesion characteristics on outcome of directional coronary atherectomy. J Am Coll Cardiol 17:1112–1120. 1991;
10. Ellis SG, Popma JJ, Buchbinder M, Franco I, Leon MB, Kent KM, Richard AD, Satler LF, Topol EJ, Whitlow PL. Relation of clinical presentation, restenosis morphology, and operator technique to the procedural results of rotational atherectomy and rotational atherectomy-facilitated angioplasty. Circulation 89:882–892. 1994;
11. Cook SL, Eigler NL, Shefer A, Golenberg T, Forrester JS, Litvack F. Percutaneous excimer laser coronary angioplasty of lesions not ideal for balloon angioplasty. Circulation 84:632–643. 1991;
12. Reifart N, Vandormael M, Krajcar M, et al. Randomized comparison of angioplasty of complex coronary lesions at a single center: excimer laser, rotational atherectomy, and balloon comparison (ERBAC) study. Circulation 84:632–643. 1991;
13. Topol E, Leya F, Pinkerton C, Pinkerton CA, Whitlow PL, Hofling B, Simonton CA, Masden RR, Serruys PW, Leon MB. A comparison of directional atherectomy with coronary angioplasty in patients with coronary artery disease: The CAVEAT study group. N Engl J Med 329:221–227. 1993;
14. Mintz GS, Popma JJ, Pichard AD, Kent KM, Satler LF, Wong C, Hong MK, Korach JA, Leon MB. Arterial remodeling after coronary angioplasty: a serial intravascular ultrasound study. Circulation 94:35–43. 1996;
15. Fischman DL, Leon MB, Baim DS, Schatz RA, Savage MP, Penn I, Detre K, Veltri L, Ricci D, Nobuyoshi M, et al. A randomized comparison of coronary stent placement and balloon angioplasty in the treatment of coronary artery disease. N Engl J Med 331:496–501. 1994;
16. Serruys PW, de Jaegere P, Kiemeneij F, Macaya C, Rutsch W, Hendrickx G, Emanuelsson H, Marco J, Legrand V, Materne P, et al. A comparison of balloon-expandable-stent implantation with balloon angioplasty in patients with coronary artery disease. N Engl J Med 331:489–495. 1994;
17. Akiyama T, Moussa I, Reimers B, Ferraro M, Kobayashi Y, Blengino S, Di Francesco L, Colombo A. Angiographic and clinical outcome following coronary stenting of small vessels. A comparison with coronary stenting of large vessels. J Am Coll Cardiol 32:1610–1618. 1998;
18. Kobayashi Y, De Gregorio J, Kobayashi N, Akiyama T, Reimers B, Finci L, Di Mario C, Colombo A. Stented segment length as an independent predictor of restenosis. J Am Coll Cardiol 34:651–659. 1999;
19. Hoffmann R, Mintz GS, Dussaillant GR, Popma JJ, Pichard AD, Salter LF, Kent KM, Griffin J, Leon MB. Patterns and mechanisms of instent restenosis: a serial intravascular ultrasound study. Circulation 94:1247–1254. 1996;
20. Hoffmann R, Mintz GS, Mehran R, Pichard AD, Kent KM, Salter LF, Popma JJ, Wu H, Leon MB. Intravascular ultrasound predictors of angiographic restenosis in lesions treated with Palmaz-Schatz stents. J Am Coll Cardiol 31:43–49. 1998;
21. Kornowski R, Mintz GS, Kent KM, Pichard AP, Satler LF, Bueher TA, Hong MK, Popma JJ, Leon MB. Increased restenosis in diabetes mellitus after coronary interventions is due to exaggerated intimal hyperplasia. A serial intravascular ultrasound study. Circulation 95:1366–1369. 1997;
22. Colombo A, Hall P, Nakamura S, Almagor Y, Maiello LA, Sousa AG, Falotico , Jaeger J, Popma JJ, Serruys PW. Intracoronary stenting without anticoagulation accomplished with intravascular ultrasound guidance. Circulation 91:1676–1688. 1995;
23. Carter AJ, Bailey LR, Llanos G, et al. Stent based sirolimus delivery reduces neointimal proliferation in a porcine coronary model of restenosis. J Am Coll Cardiol 35(Suppl A)13A(Abstr). 2000;
24. Klugherz BD, Llanos G, Lieuallen W, et al. Stent-based delivery of sirolimus for the prevention of restenosis. J Am Coll Cardiol 35(Suppl A)58A(Abstr). 2000;
25. Sousa JE, Costa MA, Abizaid AS, Feres F, Pino IM, Seixas AS, Staico R, Matto LA, Sousa AG, Falotico , Jaeger J, Popma JJ, Serruys PW. Lack of neointimal proliferation after implantation of sirolimus-coated stents in human coronary arteries. A quantitative coronary angiography and three-dimensional intravascular ultrasound study. Circulation 103:192–195. 2001;
26. Teirstein PS, Massullo V, Jani S, Popma JJ, Mintz GS, Russo RJ, Schatz RA, Guarneri EN, Steuterman S. Catheter-based radiotherapy to inhibit restenosis after coronary stenting. N Engl J Med 336:1697–1703. 1997;
27. Waksman R, White RL, Chan RC, Bass BG, Geirlach L, Mintz GS, Satler LF, Mehran R, Serruys PW, Lansky AJ, Fitzgerald P, Bhargava B, Kent KM, Pichard AD, Leon MB. Intracoronary gamma-radiation therapy after angioplasty inhibits recurrence in patients with in-stent restenosis. Circulation 101:2165–2171. 2000;
28. Leon MB, Teirstein PS, Moses JW, Tripuraneni P, Lansky AJ, Jani S, Wong SC, Fish D, Kumtz RE. Localized intracoronary gamma-radiation therapy to inhibit the recurrence of restenosis after stenting. N Engl J Med 344:250–256. 2001;
29. Raizner AE, Oesterle SN, Waksman R, Serruys PW, Colombo A, Lim YL, Yeung AC, van der Giessen Wj, Vandertie L, Chiu JK, White LR, Fitzgerald PJ, Kaluza GL, Ali NM. Inhibition of restenosis with beta-emitting radiotherapy: Report of the proliferation reduction with vascular energy trial (PREVENT). Circulation 102:951–958. 2000;
30. Kuntz R. Late breaking trials. American College of Cardiology 2001.
31. Rosengart TK, Lee LY, Patel SR, Sanborn SR, Sanborn TA, Parkh M, Bergman GW, Hachamovitch R, Szuic M, Kligfield Pd, Okin PM, Hahn RT, Devereux RB, Post MR, Hackett NR, Foster T, Grasso TM, Lesser ML, Isom OW, Crystal RG. Angiogenesis gene therapy. Phase I assessment of direct intramyocardial administration of an adenovirus vector expressing VEGF121 cDNA to individuals with clinically significant severe coronary artery disease. Circulation 100:468–474. 1999;

Article information Continued

Figure 1.

Angiographic (top panels) and intravascular ultrasound (bottom panels) findings in a patient with ostial left anterior descending coronary artery lesion (Left panels). The lesion was successfully treated with directional coronary atherectomy (DCA; Middle panels). However, seven months later, the patient returned with recurrent symptoms and restenosis (Right panels). Intravascular ultrasound demonstrated chronic geometric remodeling as the predominant mechanism of restenosis rather than neointimal hyperplasia. (Reproduced by permission from reference 7).

Figure 2.

The ostial right coronary artery lesion was successfully treated with a Palmaz-Schatz stent (JJIS; Left panels). When the patient returned with restenosis (Right panels), intravascular ultrasound showed no change in stent dimension but severe neointimal hyperplasia as the cause of the in-stent restenosis. (Reproduced by permission from reference 7).

Table 1.

Mechanisms of restenosis and potential treatments

Mechanism Treatment Availability
Acute Recoil Stent Yes
Thrombus Deposition Antiplatelet therapy including glycoprotein IIb/IIIa inhibitors Yes
Inflammation Anti-inflammatory agent Yes
Chronic Geometric Remodeling Stent Yes
Neointimal Hyperplasia Antiproliferative agent/drug-eluting stent Undergoing investigation
In-stent neointimal hyperplasia Brachytherapy Yes