Article
Ischemic heart disease
REC Interv Cardiol. 2019;1:21-25
Access to side branches with a sharply angulated origin: usefulness of a specific wire for chronic occlusions
Acceso a ramas laterales con origen muy angulado: utilidad de una guía específica de oclusión crónica
Servicio de Cardiología, Hospital de Cabueñes, Gijón, Asturias, España
ABSTRACT
Introduction and objectives: The optimal treatment of nonculprit angiographic intermediate lesions (diameter stenosis 40%-69%) in patients with ST-segment elevation myocardial infarction (STEMI) is still unknown. Lesions with fractional flow reserve (FFR) ≤ 0.80 are indicative of ischemia and benefit from revascularization. However, lesions with FFR > 0.80 and optical coherence tomography (OCT) findings of vulnerability have been hypothesized to cause adverse events during follow-up. The study aims to compare the efficacy of a preventive treatment with stent implantation plus optimal medical therapy vs optimal medical therapy alone for nonculprit intermediate lesions with FFR > 0.80 and OCT findings of plaque vulnerability in STEMI patients at 4 years of follow-up.
Methods: This parallel-group, multicenter, controlled, single-blind, and 1:1 randomized trial will enroll a total of 600 STEMI patients with ≥ 1 intermediate nonculprit lesions with FFR > 0.80 and OCT findings of plaque vulnerability. The primary endpoint is target vessel failure, defined as the composite of cardiac death, target vessel myocardial infarction, or target vessel revascularization. The study will include a parallel registry of patients with FFR > 0.80 but without OCT findings of vulnerability. Vulnerable plaques are defined as lipid-rich fibroathermas with plaque burden ≥ 70% and a thin fibrous cap (≤ 80 μm).
Results: The VULNERABLE trial will reveal the role of preventive treatment with stent implantation for nonculprit and functionally nonsignificant vulnerable plaques in STEMI patients.
Conclusions: This is the first randomized trial of OCT-guided treatment of vulnerables plaques. Registered at ClinicalTrials.gov (NCT05599061).
Keywords: Fractional flow reserve. Optical coherence tomography. ST-segment elevation myocardial infarction. Vulnerable plaque.
RESUMEN
Introducción y objetivos: El tratamiento óptimo de las lesiones angiográficas intermedias (diámetro de estenosis 40-69%) no culpables en pacientes con infarto agudo de miocardio con elevación del segmento ST (IAMCEST) está por determinar. La reserva fraccional de flujo (RFF) permite diagnosticar lesiones causantes de isquemia (RFF ≤ 0,80) que se benefician de una revascularización. No obstante, las lesiones con RFF > 0,80 y criterios de vulnerabilidad por tomografía de coherencia óptica (OCT) también se ha hipotetizado que pueden causar eventos adversos en el seguimiento. El objetivo es comparar la eficacia del tratamiento preventivo con implantación de stent más tratamiento médico óptimo de lesiones intermedias no culpables con RFF > 0,80 y características de placa vulnerable frente a solo tratamiento médico óptimo en pacientes con IAMCEST a 4 años de seguimiento.
Métodos: Estudio de grupos paralelos, multicéntrico, controlado, aleatorizado 1:1 y simple ciego. Se incluirán 600 pacientes con IAMCEST y al menos una lesión intermedia no culpable que presenten RFF > 0,80 y características de placa vulnerable por OCT. El objetivo primario se define como fallo del vaso diana, compuesto de muerte cardiaca, infarto del vaso diana y necesidad de revascularización del vaso diana. El estudio incluye un registro paralelo para pacientes con RFF > 0,80 sin características de placa vulnerable. Se define placa vulnerable como fibroateromas lipídicos con carga de placa ≥ 70% y capa fibrosa fina (≤ 80 µm).
Resultados: El estudio VULNERABLE permitirá conocer el papel del tratamiento preventivo con stent de placas vulnerables no culpables funcionalmente no significativas en pacientes con IAMCEST.
Conclusiones: Se trata del primer estudio aleatorizado para el tratamiento de placas vulnerables guiado por OCT. Registrado en ClinicalTrials.gov (NCT05599061).
Palabras clave: Reserva fraccional de flujo. Tomografía de coherencia óptica. Infarto agudo de miocardio con elevación del segmento ST. Placa vulnerable.
Abbreviations FFR: fractional flow reserve. MLA: minimum lumen area. OCT: optical coherence tomography. OMT: optimal medical therapy. PDE: percent diameter stenosis. STEMI: ST-segment elevation myocardial infarction.
INTRODUCTION
The presence of multivessel disease, defined as angiographic lesions with a percent diameter stenosis (PDS) ≥ 50% by visual estimation in patients with ST-segment elevation myocardial infarction (STEMI), is estimated to be approximately 50%.1 The COMPLETE trial compared angiography-guided preventive revascularization with stent implantation added to optimal medical therapy (OMT) for nonculprit lesions with a PDS ≥ 70% vs OMT alone.2 The trial found that angiography-guided preventive revascularization significantly reduced adverse cardiovascular events at 3 years of follow-up.2 Although the COMPLETE trial required physiological assessment using fractional flow reserve (FFR) for lesions with a PDS between 50% and 69% to guide the decision on revascularization, in practice, it was performed in only a very small percentage of patients.
The FLOWER-MI and FRAME-AMI trials3,4 investigated preventive stenting of FFR-guided nonculprit lesions—obtained through intracoronary pressure wire—compared with angiography-guided complete revascularization (visual estimation). Both trials mainly included intermediate lesions and demonstrated that pressure wire-guided preventive revascularization significantly reduces the need for revascularization, with similar or superior efficacy to angiography-guided complete revascularization.3,4 Despite these findings, clinical practice guidelines based on the COMPLETE trial recommend preventive stenting of nonculprit lesions guided by angiography alone.5,6
It is important to note that FFR is considered the gold standard for detecting myocardial ischemia (FFR ≤ 0.80). However, deferring treatment of nonculprit lesions that do not cause ischemia (FFR > 0.80) through OMT raises concerns in selected cases in which the anatomical features of the lesion suggest signs of vulnerability. In the FLOWER-MI trial, the group of patients randomized to undergo pressure-wire-guided revascularization with an FFR > 0.80 (referred for OMT) had more adverse events than those in the same group with FFR values ≤ 0.80 (referred for percutaneous revascularization).7 Several studies using intravascular imaging modalities have also demonstrated an association between the presence of fibro-lipid plaques with high lipid content and thin fibrous caps—known as vulnerable plaques—and the development of future adverse events due to plaque rupture.8,11
The VULNERABLE trial aims to evaluate the efficacy of a combined strategy using intracoronary physiological techniques and intravascular imaging to guide the treatment of intermediate nonculprit lesions in STEMI patients. The study hypothesis is that preventive stenting—in addition to OMT—in intermediate nonculprit lesions with FFR values > 0.80 and characteristics of vulnerable plaque will be superior to OMT alone. The present article includes the rationale and design of the study.
METHODS
Design
The VULNERABLE trial (NCT05599061) includes 3 groups based on the results obtained during the combined functional and anatomical assessment using pressure wires and optical coherence tomography (OCT). Figure 1 shows the study flowchart, which illustrates the 3 groups: patients with FFR ≤ 0.80 treated with stent (search failures), patients with FFR > 0.80 without vulnerable plaque characteristics (included in the registry group), and patients with FFR > 0.80 and vulnerable plaque characteristics (included in the randomized clinical trial).
This is a multicenter, controlled, prospective, randomized, parallel-group, single-blind study with patients included in the clinical trial group. The study will be conducted in accordance with the recommendations outlined in the Declaration of Helsinki on clinical research and has been approved by the lead ethics committee (Hospital Universitari de Bellvitge) and endorsed by the remaining ethics committees of participating centers. The participating centers and principal investigators are shown in table 1 of the supplementary data.
Primary endpoint |
---|
Compare the percentage of TVF between the 2 groups of patients assigned to the randomized clinical trial (FFR > 0.80 with characteristics of vulnerable plaque by OCT): preventive revascularization with stent + OMT vs OMT alone |
Key secondary endpoints |
Compare the percentage of TVF between patients allocated to the registry group (FFR > 0.80 without characteristics of vulnerable plaque by OCT and treated with the OMT) and patients allocated to the randomized OMT group (FFR > 0.80 with characteristics of vulnerable plaque) |
Other secondary endpoints |
Compare the rate of all-cause mortality reported between the 2 subgroups of randomized patients |
Compare the percentage of cardiac deaths reported between the 2 subgroups of randomized patients |
Compare the percentage of all myocardial infarctions reported between the 2 subgroups of randomized patients |
Compare the percentage of target vessel myocardial infarctions reported between the 2 subgroups of randomized patients |
Compare the percentage of target vessel revascularization needs between the 2 subgroups of randomized patients |
Evaluate the percentage of restenosis and stent thrombosis in the preventive revascularization group with stent + OMT of the randomized clinical trial |
* Although all objectives are marked with a complete 4-year follow-up, an interim study will be conducted at 2 years. ** All objectives will be calculated on an intention-to-treat basis according to the statistical plan. An exploratory per-protocol analysis will also be conducted based on the assessment by the study’s core imaging laboratory. |
FFR: fractional flow reserve; OCT: optical coherence tomograph; OMT: optimal medical treatment; TVF: target vessel failure. |
The study has been entirely designed and initiated by researchers and is sponsored by the Spanish Society of Cardiology Working Group on Intracoronary Diagnostic Techniques, which includes a steering committee, a data and safety monitoring board, and an independent event adjudication committee. The members of these committees are listed in table 2 of the supplementary data. The steering committee and all study investigators are committed to accurate data collection and adherence to the study protocol. The funding entity (Abbott Vascular, United States) plays no role in the study design, data collection, analysis, or the writing of the study results. The study sponsor (Foundation for Education in Interventional Cardiology Procedures [EPIC]), along with the principal investigators, is responsible for data management and confidentiality.
Inclusion criteria |
---|
Patients older than 18 years |
With STEMI (ST-segment elevation > 1 mm in, at least, 2 contiguous leads or true posterior ST-segment elevation with > 2 mm depression in anterior leads or new onset left bundle branch block) treated with successful revascularization of the culprit lesion within 72 hours from symptom onset |
Presenting with multivessel disease with, at least, 1 angiographically intermediate lesion (PDS of 40% up to 69% by visual estimation) in a native vessel different from the culprit vessel |
Planned FFR-guided percutaneous revascularization with a single 2.0 mm-to- 4.5 mm stent |
Between 1 and 60 days after the index procedure (revascularization of the STEMI culprit vessel) |
Exclusion criteria |
Life expectancy < 4 years |
Women of childbearing age who wish to become pregnant |
Known intolerance to acetylsalicylic acid, heparin, everolimus, or iodinated contrast |
Unresolved mechanical complications or infarct-related cardiogenic shock |
Lesions suitable for the study located in the left main coronary artery, vessels with previous revascularization, in coronary bifurcations with > 2.5 mm side branches, severe angulations, or segments with severe calcification |
History of severe asthma |
Chronic kidney disease with glomerular filtration rate < 45 mL/min |
FFR: fractional flow reserve; PDS: percent diameter stenosis; STEMI: ST-segment elevation myocardial infarction. |
Endpoints
The primary objective of the VULNERABLE study (NCT05599061) is to compare the efficacy of preventive stenting combined with OMT vs OMT alone for intermediate lesions in noninfarct-related arteries with an FFR > 0.80 and vulnerable plaque characteristics as identified by OCT over a 4-year follow-up period. The primary endpoint of the study is the rate of target vessel failure (TVF), which is defined as a composite of cardiac death, target vessel myocardial infarction, or the need for target vessel revascularization.
The study also aims to evaluate several secondary endpoints, which are summarized in table 1. Among these secondary objectives, a key focus is the comparison of the TVF rate (the primary endpoint) between the registry group (patients with FFR > 0.80 without vulnerable plaque characteristics treated with OMT) and the randomized OMT arm of the clinical trial (patients with FFR > 0.80 and vulnerable plaque characteristics). The study endpoints are defined in table 3 of the supplementary data.12,13
Patient inclusion and exclusion criteria
The inclusion and exclusion criteria for the study are detailed in table 2. In brief, all patients with STEMI who have undergone successful revascularization of the culprit lesion and have at least 1 intermediate lesion (visually defined as having a DS of 40%-69%) in a noninfarct-related artery will be eligible for the study if percutaneous revascularization with a single stent guided by FFR is being considered. The study procedure must be conducted between 1 and 60 days after the revascularization of the culprit lesion. Patients must provide informed consent prior to the elective procedure for evaluating the nonculprit lesion.
Study protocol for nonculprit lesions and randomization
Eligible lesions will first be assessed with a pressure wire following the standard procedures in each center. Lesions with an FFR ≤ 0.80 will be considered search failures, and revascularization will be recommended based on clinical indications.5,6
Lesions with an FFR > 0.80 will be further evaluated with OCT according to the standard acquisition methods to detect vulnerable plaques in each center. The decision on whether a lesion meets the criteria for vulnerable plaque will be made by an accredited local investigator during the study procedure.
Patients with at least 1 lesion with an FFR > 0.80 without vulnerable plaque characteristics on OCT will be included in the registry group of the study. The protocol recommends OMT for all lesions with an FFR > 0.80 without vulnerable plaque characteristics. These patients will receive the same clinical follow-up as those in the randomized clinical trial group.
Patients with at least 1 lesion with an FFR > 0.80 that meets the criteria for a vulnerable plaque on OCT will be included in the clinical trial group. These patients will be randomized 1:1 to either preventive stenting combined with OMT or OMT alone (figure 1). Randomization will be conducted without stratification by center or clinical condition, using telematic algorithms. This process will be carried out online via the data collection platform provided by pInvestiga (Pontevedra, Spain).
The supplementary data provide additional details on the FFR assessment method, including special situations where the lesion under study could not be fully evaluated, instances of unstable nonculprit plaques, complications related to diagnostic techniques, or patients with more than 1 nonculprit lesion.
Study device and implantation procedure
Patients with an FFR > 0.80 and vulnerable plaque characteristics identified by OCT assigned to the percutaneous coronary intervention group will be treated with an everolimus-eluting stent (Xience, Abbott, United States). According to the protocol, stent implantation must be guided by OCT. The criteria for OCT-guided stent implantation are detailed in table 4 of the supplementary data.
Optimal medical therapy
All patients included in both the randomized clinical trial and the registry must receive treatment in accordance with the European Society of Cardiology guidelines for managing acute coronary syndromes.5 The study protocol emphasizes managing modifiable risk factors—such as diet, smoking, obesity, exercise, and psychological status—as well as nonmodifiable risk factors, with set targets for blood pressure (systolic < 130 mmHg and diastolic < 80 mmHg), low-density lipoprotein cholesterol (< 55 mg/dL), and glycated hemoglobin A1c (< 7%). Pharmacological therapy should include beta-blockers and renin-angiotensin system inhibitors. Dual antiplatelet therapy is also recommended, but only during the first year after the index procedure, at the discretion of each center. As per the protocol, patient treatment details will be reported annually, and 2 lipid profile tests will be conducted throughout the study.
Vulnerable plaque criteria on optical coherence tomography and investigator training
Based on histopathological data, a plaque is defined as vulnerable when it is caused by a fibroatheroma with a large necrotic core composed of cellular debris and a high number of inflammatory cells, covered by a thin fibrous cap (≤ 65 µm).14 The criteria for identifying a vulnerable plaque in the study are adapted from the classic histopathological definition but modified for OCT assessment. These criteria are shown in figure 2.
According to the protocol, 3 simultaneous criteria are required to define a vulnerable plaque by OCT:
The presence of a fibro-lipid plaque with a necrotic core covering more than 90º of the perimeter of the vessel over a length of more than 5 mm. A necrotic core is defined as a hypointense image with poorly defined borders that attenuates the OCT light beam, preventing visualization of the artery behind the core.
The presence of a thin fibrous cap, defined as ≤ 80 µm (65 + 15 µm axial resolution) in ≥ 3 consecutive images. The fibrous cap is defined as the tissue separating the necrotic core from the vessel lumen. Investigators will be trained to differentiate other findings that could be mistaken for a thin cap on OCT. Figure 3 shows examples of analogous OCT images that may mimic a thin fibrous cap but do not correspond to vulnerable plaques.
Investigators will be required to measure a plaque burden of ≥ 70% in the cross-sectional area corresponding to the minimal luminal area (MLA) within the lesion. To perform this assessment, it is necessary to measure the vessel perimeter by delineating the external elastic membrane (EEM). Due to the difficulty of assessing the vessel perimeter in fibro-lipid plaques, especially at the MLA site, investigators will be trained to choose a section as close as possible to the MLA, where at least 60% of the vessel perimeter can be visualized if it is not possible at the same point. This allows for calculation using the following formula (figure 4):
As per protocol, at least 1 local investigator from each participating center must have completed an online training course for the detection and assessment of vulnerable plaques using OCT, following the study criteria. Upon completing this course and passing a specific questionnaire, the investigator will be certified and approved to participate in the study.
Angiographic and optimal coherence tomography quantification analyses
The study includes an independent imaging laboratory for angiographic quantification and OCT analysis (Barcelona Cardiac Imaging Core Laboratory [BARCICORE-Lab]) to monitor adherence to the study criteria for diagnosing vulnerable plaques. A blinded analysis of the study results will be conducted, and patients will be assigned according to the protocol for exploratory analysis. A detailed explanation of the angiographic and OCT analysis conducted by the study laboratory is shown in the supplementary data.
Clinical follow-up and blinding
Patients in both the registry group and the randomized clinical trial group will undergo clinical follow-up for 4 years. Follow-up will include telephone consultations at 1 and 3 years, and in-person visits at 2 and 4 years. Each follow-up will involve an electrocardiogram and blood tests with cholesterol determination.
Patients in the randomized clinical trial group will be blinded to their assigned treatment group (single-blind). The details of blinding and monitoring are specified in the supplementary data.
Sample size calculation
The sample size has been calculated for the randomized clinical trial group. The number of patients included in the registry and search failures will depend on the total number needed to achieve the estimated sample size for the randomized trial.
According to previous studies on patients with acute coronary syndrome, theTVF rate for nonculprit lesions meeting vulnerable plaque criteria treated with OMT is estimated to be around 8% to 10% at 4 years. In similar lesions treated with stenting, the rate is approximately 4%.2,7,9 The studies used for the sample size calculation are summarized in table 5 of the supplementary data. Based on the study hypothesis, preventive stenting in nonculprit lesions with an FFR > 0.80 and vulnerable plaque characteristics is expected to reduce the primary endpoint by 60%. The estimated rate of TVF in the OMT group at 4 years is 10%. Assuming an annual loss to follow-up rate of 1.5% (total 6%), randomizing 600 participants 1:1 to preventive stenting plus OMT vs OMT alone will provide 80% power to demonstrate the superiority of preventive stenting with a 2-sided alpha error of .05.
Statistical analysis plan
The primary and secondary endpoints will be analyzed using the intention-to-treat principle at the 4-year follow-up. Comparisons will estimate event proportions between groups using logistic regression and will be reported as odds ratios with 95% confidence intervals. Only 1 event per patient will be counted for the primary endpoint. P values < .05 will be considered statistically significant for the primary endpoint. Kaplan-Meier curves will be used to visualize the time to the first event between groups.
For primary endpoint composites with missing data, a specific monitoring plan will determine if the missing data are random. In cases where data are adjudicated as missing at random, imputation methods will be used. For nonrandom missing data, sensitivity analyses using worst-case and last observation carried forward methods will be conducted.
Subgroup analyses will be performed for the primary and secondary endpoints, which involves comparing TVF rates between registry patients and those randomized to OMT in the clinical trial. Prespecified subgroups include: age > 75 years, sex, diabetes mellitus, left ventricular ejection fraction ≤ 35% at the time of the procedure, lesions in the proximal or mid-left anterior descending artery, and lesions in vessels with a reference diameter ≤ 2.75 mm.
Additionally, a hypothesis-generating parallel analysis will be conducted according to the study protocol. Patients will be included in the analysis only if the imaging laboratory confirms that their assigned treatment group, as determined by the local investigator, is consistent with the presence of vulnerable plaque identified by OCT. Patients will be excluded if there is a discrepancy between the investigator’s assignment and the imaging laboratory’s findings.
Interim analysis
After 2 years of follow-up, an interim analysis of the data is planned to monitor the primary endpoint in the randomized clinical trial group. Clinical follow-up will be extended if the events observed in the OMT arm of the randomized clinical trial are less than 4%.
DISCUSSION
The VULNERABLE trial aims to investigate the combined use of intracoronary physiology and images to guide the treatment of intermediate nonculprit lesions in STEMI patients.
Several lipid-lowering and anti-inflammatory drugs have been shown to reduce thrombotic events in patients with STEMI, likely by stabilizing functionally nonsignificant vulnerable plaques.15,17 In the PACMAN-AMI trial, treatment with alirocumab in addition to statins significantly reduced atheroma, decreased lipid content, and led to thickening of the fibrous cap compared with placebo in coronary regions with angiographically nonobstructive atherosclerosis (DS, 20%-50%).18 However, it is noteworthy that only 31% of patients in that study exhibited all 3 markers of reduced atherosclerosis, and data on more significant plaques (eg, 40%-69% stenosis with vulnerability criteria) were not specified.19
The use of stents in patients with vulnerable plaques is intended to enhance neointimal healing of the struts, which thickens the fibrous cap and stabilizes the plaque. The randomized PREVENT trial assessed the effectiveness of preventive stenting for functionally nonsignificant vulnerable lesions in patients with chronic coronary syndrome compared with OMT. Vulnerable plaques were identified using various intravascular imaging techniques, with most being guided solely by intravascular ultrasound. The study found that preventive stenting resulted in a statistically significant reduction in the rate of TVF at 2 years of follow-up (0.4% vs 3.4%; P = .0003).11
Finally, several observational trials have demonstrated that OCT is an effective method for detecting vulnerable plaques and monitoring the response to intensive treatments aimed at stabilizing these plaques through fibrous cap thickening.18,20 The PECTUS-obs trial included 438 acute coronary syndrome patients with nonculprit lesions with FFR > 0.80 treated with the OMT alone.10 All lesions were examined using OCT, with criteria similar to those used in the VULNERABLE trial to define vulnerable plaques. In that study, 34% of patients had at least 1 vulnerable lesion, which was associated with a higher risk of adverse events (15.4% vs 8.2% for the composite endpoint of death, myocardial infarction, or revascularization in the groups with and without vulnerable plaques, respectively). The VULNERABLE trial is the first to use OCT to guide the treatment of vulnerable plaques in functionally nonsignificant lesions.
CONCLUSIONS
The VULNERABLE trial aims to evaluate the effectiveness of preventive stenting plus OMT vs OMT alone for vulnerable plaques, as defined by OCT, in functionally nonsignificant intermediate lesions in nonculprit vessels of patients with STEMI. In addition, the study will provide information on the clinical relevance of the presence of vulnerable plaques in nonculprit lesions.
FUNDING
This study has been funded by Abbott Vascular.
ETHICAL CONSIDERATIONS
The study is being conducted following the recommendations outlined in the Declaration of Helsinki on clinical research, has been approved by Hospital Universitari de Bellvitge research ethics committee, and endorsed by the remaining ethics committees of participating centers. Informed consent acceptance and signature are required prior to performing any elective procedures to study the nonculprit lesion. Potential sex and gender biases are considered.
STATEMENT ON THE USE OF ARTIFICIAL INTELLIGENCE
No artificial intelligence was used in the drafting of this manuscript.
AUTHORS’ CONTRIBUTIONS
J. Gómez-Lara and E. Gutiérrez-Ibañes drafted this document. The remaining signatories reviewed the document, made changes at their discretion, and approved the final text.
CONFLICTS OF INTEREST
J. Gómez-Lara and E. Gutiérrez-Ibañes received a grant from Abbott Vascular for this study. A. Jurado-Román has received fees from Abbott, Boston, and Shockwave. E. Fernández received fees from Abbott and Hexacath. C. Cortés received a Río Hortega Contract from Instituto de Salud Carlos III. S. Brugaletta received fees from Abbott, Microport, and General Electric. T. García-Camarero received fees from Medtronic and Boston. J.A. Linares Vicente received fees from Abbott Vascular, Braun, AstraZeneca, Bayer, and IZASA. O. Rodríguez-Leor received fees from Shockwave, WorlsMedica, and Medtronic. S. Ojeda received fees from Abbott, Boston, WorldMedica, and Biosensors. A. Pérez de Prado received grants and fees from Abbot, Boston, iVascular, and Terumo. H.M. García-García received fees from ACIST, Boston Scientific, Medis, Biotronik, InfraRedx/Nipro, Chiesi, and Cordis. S. Ojeda and A. Pérez de Prado are associate editors of REC: Interventional Cardiology; the journal’s editorial procedure to ensure impartial processing of the manuscript has been followed. The remaining authors declared no conflicts of interest whatsoever.
WHAT IS KNOWN ABOUT THE TOPIC?
- Thin-cap fibroatheromas, also known as vulnerable plaques, are responsible for most acute coronary syndromes. Approximately 50% of patients with STEMI have additional angiographic lesions beyond the culprit lesion, which are associated with a significant number of adverse ischemic events. Preventive stenting for severe nonculprit lesions (DS ≥ 70%) has been shown to reduce the number of adverse events. However, the effectiveness of preventive stenting for angiographically intermediate nonculprit lesions (SD, 40%-69%) that have characteristics of vulnerable plaques remains to be determined.
WHAT DOES THIS STUDY ADD?
- VULNERABLE is the first randomized trial to evaluate the preventive treatment of angiographically intermediate, nonculprit lesions that exhibit features of vulnerability identified by OCT in patients with STEMI.
REFERENCES
1. Park DW, Clare RM, Schulte PJ, et al. Extent, location, and clinical significance of non-infarct-related coronary artery disease among patients with ST-elevation myocardial infarction. JAMA 2014;312:2019-2027.
2. Mehta SR, Wood DA, Storey RF, et al. Complete Revascularization with Multivessel PCI for Myocardial Infarction. N Engl J Med. 2019;381:1411-1421.
3. Lee JM, Kim HK, Park KH, et al. Fractional flow reserve versus angiography-guided strategy in acute myocardial infarction with multivessel disease:a randomized trial. Eur Heart J. 2023;44:473-484.
4. Puymirat E, Cayla G, Simon T, et al. Multivessel PCI Guided by FFR or Angiography for Myocardial Infarction. N Engl J Med. 2021;385:297-308.
5. Byrne RA, Rossello X, Coughlan JJ, et al. 2023 ESC Guidelines for the management of acute coronary syndromes. Eur Heart J. 2023;44:3720-3826.
6. Lawton JS, Tamis-Holland JE, Bangalore S, et al. 2021 ACC/AHA/SCAI Guideline for Coronary Artery Revascularization:Executive Summary:A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. Circulation. 2022;145:4-17.
7. Denormandie P, Simon T, Cayla G, et al. Compared Outcomes of ST-Elevation Myocardial Infarction Patients with Multivessel Disease Treated with Primary Percutaneous Coronary Intervention and Preserved Fractional Flow Reserve of Non-Culprit Lesions Treated Conservatively and of Those with Low Fractional Flow Reserve Managed Invasively:Insights from the FLOWER MI trial. Circ Cardiovasc Interv. 2021;14:011314.
8. Stone GW, Maehara A, Lansky AJ, et al. A prospective natural-history study of coronary atherosclerosis. N Engl J Med. 2011;364:226-235.
9. Erlinge D, Maehara A, Ben-Yehuda O, et al. Identification of vulnerable plaques and patients by intracoronary near-infrared spectroscopy and ultrasound (PROSPECT II):a prospective natural history study. Lancet. 2021;397:985-995.
10. Mol JQ, Volleberg R, Belkacemi A, et al. Fractional Flow Reserve-Negative High-Risk Plaques and Clinical Outcomes After Myocardial Infarction. JAMA Cardiol. 2023;8:1013-1021.
11. Park SJ, Ahn JM, Kang DY, et al. Preventive percutaneous coronary intervention versus optimal medical therapy alone for the treatment of vulnerable atherosclerotic coronary plaques (PREVENT):a multicentre, open-label, randomised controlled trial. Lancet. 2024;403:1753-1765.
12. Cutlip DE, Windecker S, Mehran R, et al. Clinical end points in coronary stent trials:a case for standardized definitions. Circulation. 2007;115:2344-2351.
13. Thygesen K, Alpert JS, Jaffe AS, et al. Fourth universal definition of myocardial infarction (2018). Eur Heart J. 2019;40:237-269.
14. Virmani R. Pathology of the vulnerable plaque. J Am Coll Cardiol. 2006;47:C13-8.
15. Cannon CP, Braunwald E, McCabe CH, et al. Intensive versus moderate lipid lowering with statins after acute coronary syndromes. N Engl J Med. 2004;350:1495-1504.
16. Schwartz GG, Steg PG, Szarek M, et al. Alirocumab and Cardiovascular Outcomes after Acute Coronary Syndrome. N Engl J Med. 2018;379:2097-2107.
17. Tardif JC, Kouz S, Waters DD, et al. Efficacy and Safety of Low-Dose Colchicine after Myocardial Infarction. N Engl J Med. 2019;381:2497-2505.
18. Raber L, Ueki Y, Otsuka T, et al. Effect of Alirocumab Added to High-Intensity Statin Therapy on Coronary Atherosclerosis in Patients With Acute Myocardial Infarction:The PACMAN-AMI Randomized Clinical Trial. JAMA. 2022;327:1771-1781.
19. Biccire FG, Haner J, Losdat S, et al. Concomitant Coronary Atheroma Regression and Stabilization in Response to Lipid-Lowering Therapy. J Am Coll Cardiol. 2023;82:1737-1747.
20. Kedhi E, Berta B, Roleder T, et al. Thin-cap fibroatheroma predicts clinical events in diabetic patients with normal fractional flow reserve:the COMBINE OCT-FFR trial. Eur Heart J. 2021;42:4671-4679.
* Corresponding author.
E-mail address: gomezjosep@hotmail.com (J. Gómez-Lara).
ABSTRACT
Introduction and objectives: Distal radial access (DRA) for coronary procedures is currently recognized as an alternative to conventional transradial access, with documented advantages primarily related to access-related complications. However, widespread adoption of DRA as the default approach remains limited. Therefore, this prospective cohort study aimed to present our initial experience with DRA for coronary procedures in any clinical settings.
Methods: From August 2020 to November 2023, we included 1000 DRA procedures (943 patients) conducted at a single center. The study enrolled a diverse patient population. We recommended pre- and postprocedural ultrasound evaluations of the radial artery course, with ultrasound-guided DRA puncture. The primary endpoint was DRA success, while secondary endpoints included coronary procedure success, DRA performance metrics, and the incidence of access-related complications.
Results: The DRA success rate was 97.4% (n = 974), with coronary procedure success at 96.9% (n = 969). The median DRA time was 40 [interquartile range, 30-60] seconds. Diagnostic procedures accounted for 64% (n = 644) of cases, while 36% (n = 356) involved percutaneous coronary intervention (PCI), including primary PCI in 13% (n = 128). Pre-procedure ultrasound evaluation and ultrasound-guided DRA were performed in 83% (n = 830) and 85% (n = 848) of cases, respectively. Access-related complications occurred in 2.9% (n = 29).
Conclusions: This study shows the safety and feasibility of DRA for coronary procedures, particularly when performed under ultrasound guidance in a diverse patient population. High rates of successful access and coronary procedure outcomes were observed, together with a low incidence of access-related complications. The study was registered on ClinicalTrials.gov (NTC06165406).
Keywords: Vascular access. Distal radial artery. Coronary angiography. Percutaneous transluminal coronary angioplasty. Doppler ultrasound. Access-related complications.
RESUMEN
Introducción y objetivos: Actualmente, el acceso radial distal (ARD) para procedimientos coronarios es una alternativa al acceso radial convencional, con algunas ventajas descritas principalmente en términos de complicaciones relacionadas con el acceso. A pesar de la evidencia, pocos centros han establecido el ARD como acceso sistemático para procedimientos coronarios. El objetivo de esta cohorte prospectiva es presentar la experiencia inicial en nuestro centro con el ARD en pacientes con indicación de procedimientos coronarios en cualquier escenario clínico.
Métodos: Se incluyeron 1.000 procedimientos de ARD (943 pacientes) realizados en un único centro de agosto de 2020 a noviembre de 2023. El estudio fue realizado con pacientes en cualquier escenario clínico. Se recomendó la valoración por ultrasonido del trayecto de la arteria radial antes y después del procedimiento, así como la punción ecoguiada. El objetivo principal fue el éxito del ARD. Como objetivos secundarios se consideraron el éxito del procedimiento coronario, el desempeño del ARD y las complicaciones relacionadas con el acceso.
Resultados: El éxito del ARD fue del 97,4% (n = 974) y el éxito del procedimiento coronario fue del 96,9% (n = 969). El tiempo de acceso del ARD fue de 40 segundos [rango intercuartílico, 30-60]. Se realizaron procedimientos diagnósticos en el 64% (n = 644) e intervencionismo coronario percutáneo (ICP) en el 36% (n = 356), incluyendo ICP primario en el 13% (n = 128) de los pacientes. La valoración por ultrasonido antes del procedimiento se llevó a cabo en el 83% (n = 830) y la punción ecoguiada en el 85% (n = 848). La incidencia de complicaciones relacionadas con el acceso fue del 2,9% (n = 29).
Conclusiones: Este estudio muestra la viabilidad y la seguridad del ARD principalmente guiado por ultrasonido para los procedimientos coronarios en cualquier escenario clínico, con un alto porcentaje de éxito del acceso y de éxito del procedimiento, además de una baja incidencia de complicaciones relacionadas con el acceso. El estudio fue registrado en ClinicalTrials.gov (NTC06165406).
Palabras clave: Acceso vascular. Arteria radial distal. Coronariografía. Angioplastia coronaria transluminal percutánea. Ultrasonido Doppler. Complicaciones relacionadas con el acceso.
Abbreviations CAG: coronary angiography. DRA: distal radial access. DRart: distal radial artery. PRart: proximal radial artery. TRA: transradial access.
INTRODUCTION
Currently, distal radial access (DRA) in the anatomical snuffbox for both noncoronary and coronary procedures is gaining popularity. Since its introduction by Babunashvili et al.,1 in 2011, several observational studies have validated the feasibility and safety of DRA,2-4 comparing it with conventional transradial access (TRA). DRA has shown advantages such as a lower incidence of radial artery occlusion (RAO) and shorter hemostasis time, with minimal access-related complications.5,6 The usefulness of ultrasound to guide DRA and evaluate access-related complications has also been described.7,8 Recent randomized trials comparing DRA with TRA have reported conflicting results regarding RAO incidence, crossover rates, and access times.9-11 Nevertheless, meta-analyses consistently support the benefits of DRA, albeit with a higher crossover rate.12-13 One of the limitations of most studies on DRA is the restricted inclusion of patients in emergent situations or complex percutaneous coronary interventions (PCI), such as ST-segment elevation myocardial infarction (STEMI); therefore, the feasibility of the approach in this context is somewhat scarce.2,9-11,14 Despite current evidence, the use of DRA as the default access for coronary procedures is still not widely implemented in most centers. Hence, this prospective single-center cohort aimed to present the experience of our first 1000 DRA in patients undergoing coronary procedures in any clinical settings.
METHODS
Population and study design
The Distal Radial Access for Diagnostic and Interventional Coronary Procedures in an all-comer population (DISTAL) registry is a prospective observational investigation aiming to assess the performance of DRA and compare clinical and procedural characteristics in a diverse population undergoing coronary procedures. This interim analysis presents our initial experience with DRA conducted at a single center. All DRA procedures performed by 4 experienced operators, previously proficient in TRA, were included in the study from August 2020 to November 2023.
This study was approved by the Ethics Committee of our institution (CEIC-2804) and was conducted following the principles of the Declaration of Helsinki. All patients gave their informed written consent before the procedure.
Inclusion and exclusion criteria
The study included patients aged 18 years and older undergoing diagnostic or therapeutic coronary procedures using DRA in any clinical setting. Patients with an unsuitable distal radial artery (DRart) assessed by ultrasound (non-permeable or diameter <1 .8 mm) were excluded, as were patients with no palpable pulse of DRart with such unsuitability characteristics. Additional exclusion criteria encompassed participation in other clinical trials, known allergy to iodinated contrast, inability to provide informed consent, and women of childbearing age without a negative pregnancy test. While the Barbeau test was recommended, it was not mandatory for inclusion.15
Endpoints
The primary endpoint was the success of DRA and the main secondary endpoint was the success of the coronary procedure. Other secondary endpoints included DRA procedure time, total procedure duration, the incidence of radial artery spasm, exposure to ionizing radiation, patient comfort levels, hemostasis time, access-related complications, and the impact of ultrasound guidance on DRA performance. Detailed definitions of these endpoints are provided in the supplementary data.
Distal radial access technique
The DRA technique has been previously described,2,4,16-18 and is explained in detail in the supplementary data. Key aspects of interest included patient selection, the decision to use ultrasound-guided puncture19 (figure 1) vs blind with palpation puncture at the discretion of the operator, patient positioning for right (r) or left (l) DRA, the puncture technique itself, and the hemostasis procedure (figure 2).
Statistical analysis
Sample size and statistical power calculations were performed using the GRANMO calculator.20 A sample size of 1000 procedures was determined to provide a statistical power greater than 99% to detect a difference of 3% or more in the proportion of DRA success (primary endpoint) at our center, assuming an alpha risk of 1%. This calculation was based on a reference proportion from previous medical literature estimated around 95%.11,18,21
Categorical variables are presented as counts (percentages), while continuous variables were assessed for normal distribution using the Kolmogorov-Smirnov test. Normally distributed variables are expressed as mean (standard deviation), and nonnormally distributed variables as median [interquartile range].
To evaluate the impact of the learning curve, comparisons were made among quartiles of the study period for variables including access failure, DRA time, total procedure time, and access-related complications. Analysis of variance or the Kruskal-Wallis test was used depending on the normality of the variable. Logistic regression analysis (logit command) was used with the first quartile as the reference to compare percentages among quartiles.
Statistical analyses were conducted using SPSS Statistics 20.0 software (IBM, United States) and STATA 12 (StataCorp, College Station, United States). A p-value < 0.05 was considered statistically significant for all tests.
RESULTS
From August 2020 to November 2023, a total of 1000 DRA procedures (943 patients) were performed. Table 1 shows the patients’ baseline clinical characteristics. The mean age was 68 years, and 29% of the patients were women. A total of 47% of the procedures were performed on an outpatient basis. In 35% of cases, the indication was acute coronary syndrome (13% STEMI).
Clinical characteristics | n = 1000 |
---|---|
Age, (years), mean (SD) | 68.1 (11.7) |
Female, n (%) | 289 (28.9) |
Weight, (kg), mean (SD) | 78.0 (14.8) |
Height, (cm), mean (SD) | 167.9 (8.1) |
Body mass index, (kg/m2), mean (SD) | 28.0 (4.5) |
Hypertension, n (%) | 735 (73.5) |
Dyslipidemia, n (%) | 578 (57.8) |
Diabetes mellitus, n (%) | 353 (35.3) |
Current smoker, n (%) | 246 (24.6) |
Family history of premature coronary heart disease, n (%) | 54 (5.4) |
Previous peripheral artery disease, n (%) | 50 (0.5) |
Previous stroke, n (%) | 41 (4.1) |
Previous heart failure, n (%) | 252 (25.2) |
GFR (mL/minute/1.73m2), mean (SD) | 72.4 (20.0) |
Dialysis, n (%) | 27 (2.7) |
Left ventricular ejection fraction, mean (SD) | 52.6 (16.2) |
Atrial fibrillation, n (%) | 170 (17.0) |
OAC | |
Acenocoumarin, n (%) | 170 (17.0) |
Direct OAC, n (%) | 81 (8.1) |
Previous CAG, n (%) | 251 (25.1) |
Previous CABG, n (%) | 43 (4.3) |
Previous PCI, n (%) | 218 (21.8) |
Previous ischemic heart disease | |
Previous STEMI, n (%) | 133 (13.3) |
Previous NSTEMI, n (%) | 69 (6.9) |
Previous CCS, n (%) | 53 (5.3) |
CAG indication | |
Chronic coronary syndrome, n (%) | 207 (20.7) |
STEMI, n (%) | 128 (12.8) |
NSTEMI, n (%) | 224 (22.4) |
Staged PCI, n (%) | 60 (6.0) |
Diagnostic, n (%) | 381 (38.1) |
Preoperative CAG in patients with VHD, n (%) | 183 (18.3) |
Dilated cardiomyopathy, n (%) | 158 (15.8) |
Ventricular tachycardia, n (%) | 24 (2.4) |
Others, n (%) | 16 (1.6) |
Outpatient coronary arteriography, n (%) | 470 (47) |
CABG, coronary artery bypass grafting; CAG, coronary angiography; CCS, chronic coronary syndrome; GFR, glomerular filtration rate; NSTEMI, non−ST-segment elevation myocardial infarction; OAC, oral anticoagulation; PCI, percutaneous coronary intervention; STEMI, ST-segment elevation myocardial infarction; VHD, valvular heart disease. Data are expressed as No. (%) or mean ± standard deviation. |
Table 2 presents the characteristics of the radial artery and the DRA procedure. High rates of preprocedure ultrasound evaluation and ultrasound-guided technique for DRA were noted (83% and 85%, respectively). Notably, the percentage of coronary procedures showing insufficient catheter length due to DRA was low (3.7%).
Procedure characteristics | n = 1000 |
---|---|
Preprocedure characteristics | |
Arterial pulse strength scale | |
Absent, n (%) | 12 (1.2) |
Weak, n (%) | 167 (16.7) |
Normal, n (%) | 652 (65.2) |
Strong, n (%) | 169 (16.9) |
Radial artery preprocedure ultrasound evaluation, n (%) | 830 (83.0) |
Arterial tortuosity | |
Radial, n (%) | 23 (2.3) |
Subclavian, n (%) | 62 (6.2) |
Calcified radial artery, n (%) | 26 (2.6) |
Distal radial artery size, mm (SD) | 2.3 (0.3) |
Proximal radial artery size, mm (SD) | 2.5 (0.4) |
Depth of the distal radial artery, mm (SD) | 3.8 (1.0) |
DRA technique | |
CAG by the same DRA, n (%) | 57 (5.7) |
Ultrasound-guided access, n (%) | 848 (84.8) |
DRA side | |
Right DRA, n (%) | 627 (62.7) |
Left DRA, n (%) | 373 (37.3) |
Introducer size | |
5 French, n (%) | 256 (25.6) |
6 French, n (%) | 744 (74.4) |
Introducer sheath type | |
Prelude Ideal (Merit Medical) Introducer Kit, n (%) | 950 (95.0) |
Radifocus Introducer II Kit A (Terumo Corporation), n (%) | 50 (5.0) |
Short length of the radial catheter | 37 (3.7) |
Postprocedure arterial patency evaluation, n (%) | 907 (90.7) |
Postprocedure puncture site bleeding, n (%) | 55 (5.5) |
CAG, coronary angiography; DRA, distal radial access. Data are expressed as No. (%) or mean ± standard deviation. |
Table 3 summarizes the characteristics of coronary procedures, including the extent of coronary artery disease, types of procedures, and features of patients who underwent PCI. In general, 64% of the procedures were only diagnostic, while 36% included PCI.
Procedure characteristics | n = 1000 |
---|---|
Coronary disease extent | |
One vessel, n (%) | 285 (28.5) |
Two vessels, n (%) | 174 (17.4) |
Three vessels, n (%) | 176 (17.6) |
LMCAD, n (%) | 55 (5.5) |
Coronary bypass graft, n (%) | 27 (2.7) |
Characteristics of the coronary procedure | |
Type of coronary procedures | |
Diagnostic, n (%) | 644 (64.4) |
PCI, n (%) | 356 (35.6) |
Ambulatory PCI, n (%) | 90 (9.0) |
PCI culprit lesion | |
LMCAD, n (%) | 9 (0.9) |
Left anterior descending artery, n (%) | 164 (16.4) |
Circumflex coronary artery, n (%) | 95 (9.5) |
Right coronary artery, n (%) | 100 (10.0) |
Coronary bypass graft | 2 (0.2) |
Specific techniques | |
Wire-based intracoronary physiological assessment, n (%) | 57 (5.7) |
Optical coherence tomography, n (%) | 21 (2.1) |
Intravascular ultrasound, n (%) | 30 (3.0) |
Guide catheter extension system, n (%) | 15 (1.5) |
Rotational atherectomy, n (%) | 16 (1.6) |
Cutting balloon, n (%) | 34 (3.4) |
Intracoronary lithotripsy, n (%) | 8 (8.0) |
Thrombus aspiration, n (%) | 81 (8.1) |
Intracoronary perfusion catheter, n (%) | 7 (0.7) |
Special PCI procedures | |
Complex bifurcation, n (%) | 60 (6.0) |
Chronic total occlusion, n (%) | 16 (1.6) |
Volume of contrast, (mL), mean (SD) | 85.0 (53.1) |
Heparin dose, (IU), median [IQR] | 5000 (3000-8500) |
LMCAD, left main coronary artery disease; PCI, percutaneous coronary intervention. |
Table 4 depicts the clinical endpoints. The DRA success rate was 97.4% and the coronary procedure success rate was 96.9%. The median access time was 40 (interquartile range [IQR], 30-60) seconds, and 4% of patients experienced radial artery spasm. The overall rate of access-related complications was low (2.9%).
Clinical endpoints | n = 1000 |
---|---|
Primary endpoint | |
DRA success, n (%) | 974 (97.4) |
Coronary procedure success by DRA, n (%) | 969 (96.9) |
Secondary endpoints | |
Access time, (sec), median [IQR] | 40 (30-60) |
Procedure time, (min), median [IQR] | 29.0 [17.3-45.0] |
Radial artery spasm, n (%) | 44 (4.4) |
DAP, (Gy.m2), median [IQR] | 32.7 [19.2-63.0] |
Fluoroscopy time (min), median [IQR] | 4.6 [2.5-10.0] |
VAS patient comfort for access, mean (SD) | 2.2 (0.6) |
VAS patient comfort for hemostasis, mean (SD) | 2.1 (0.4) |
Hemostasis time, (hour), mean, (SD) | 2.9 (1.1) |
Access-related complications (all), n (%) | 29 (2.9) |
Radial artery occlusion, n (%) | 10 (1.0) |
Hematoma, n (%) | |
Type I-a, n (%) | 11 (1.1) |
Type I-b, n (%) | 1 (0.1) |
Type II, n (%) | 1 (0.1) |
Type III, n (%) | 1 (0.1) |
Type IV, n (%) | 0 (0) |
Radial pseudoaneurysm, n (%) | 0 (0) |
Radial dissection, n (%) | 5 (0.5) |
Arteriovenous fistula, n (%) | 0 (0) |
DAP, dose-area product; DRA, distal radial access; VAS, visual analog scale. Data are expressed as No. (%), mean ± standard deviation, or median [interquartile range]. |
Combined preprocedure ultrasound evaluation and ultrasound-guided puncture were performed in 82.8% of cases, with successful DRA achieved in 97.7% compared with 95.9% in those who did not undergo ultrasound guidance (P = .183). Based on the strength of the arterial pulse—absent, weak, normal, and strong—ultrasound-guided puncture was performed in 100%, 91%, 89.7%, and 45.5% of cases, respectively. Access time was longer with ultrasound-guided puncture than with nonultrasound-guided puncture (40 s [30-70] vs 35 s [30-45]; P < .001). The success of DRA in relation to the use of ultrasound-guided technique among all strengths of arterial pulse is detailed in table 1 of the supplementary data.
Arterial patency after removal of the hemostatic device was assessed in 907 patients (90.7%), revealing RAO in only 1% (n = 10).
In the quartile analysis, a shift in the selection of DRA side was observed, with lDRA initially more commonly used, shifting to rDRA as the preferred access in later quartiles (figure 3A). DRA failure rates were low in all quartiles but decreased significantly from the third quartile onwards (figure 3B). Access time decreased significantly from the second quartile onwards and remained stable thereafter (figure 3C). However, no significant differences were found in total procedure duration between quartiles (figure 3D).
DISCUSSION
Using data from a large prospective registry of patients who underwent DRA for coronary procedures, with high use of ultrasound-guided techniques, our study showed that DRA achieves high rates of access and procedural success, coupled with a low incidence of access-related complications in an all-comer population.
The usefulness of ultrasound in the distal radial access technique
Understanding the anatomy of the anatomical snuffbox is crucial for successful DRA, and ultrasound serves as a valuable tool in achieving this, offering demonstrated advantages.5,16,17,22 In our study, preprocedure ultrasound evaluation and ultrasound-guided DRA techniques were used in most patients. In addition to assessing arterial diameters and evaluating calcification and tortuosity, ultrasound enabled us to exclude patients with unsuitable distal radial arteries. Overall, we found no significant differences between ultrasound-guided and nonultrasound-guided DRA, although the former was associated with longer access times. However, the role of ultrasound is particularly noteworthy in cases of weak or absent arterial pulses, which are often underrepresented in prior studies. The presence of a suboptimal arterial pulse can stem from various factors, including small DRart, hypotension, collateral blood supply, or depth of DRart.11 In our study, most patients with weak pulses underwent ultrasound-guided puncture, with a favorable trend toward successful access in those who did. However, in patients with normal to strong pulses, no differences in DRA success were found, and even prolongation of access time was observed with its use. Therefore, in this type of pulse, an ultrasound-guided puncture is probably not necessary.
Feasibility, safety, and technical issues in distal radial access
This study corroborates the previously reported advantages of DRA,3,9,10,12,13,18 such as a low rate of RAO, acceptable access time, short hemostasis time, and adequate patient comfort.
Furthermore, the absence of an increased risk of hand dysfunction after DRA has been demonstrated,23 even compared with TRA at 12 months of follow-up, documented by Al-Azizi et al.24 Here, we focus on controversial issues that may have hampered wider adoption of this technique, and our results may provide additional support for DRA.
High success rates of DRA in coronary procedures have been reported in numerous studies.2-4,17,18,25 In addition, recent clinical trials and meta-analyzes describe a higher crossover rate compared with TRA.9-13
In contrast to our results, trials comparing DRA with TRA have reported lower access success and longer puncture times.9-11 Conversely, our study demonstrates remarkably high success rates for DRA and coronary procedures, as well as shorter access time, consistent with registries in which DRA is the default approach among experienced operators, as shown by the largest registries published to date, the DISTRACTION and KODRA studies.2-4,18,21
The KODRA trial included 4977 DRA procedures from a Korean registry.21 The authors reported a DRA success rate of 94.4%, with a crossover rate of 6.7%. In contrast to our work, the use of ultrasound-guided puncture in KODRA was low (6.4%). Additionally, the authors found predictors of DRA failure, such as the presence of a weak pulse and limited operator experience (less than 100 cases).
The equivalence of rDRA and lDRA has previously been demonstrated, and contemporary studies use mainly rDRA.9-11,17 As in the first registries, which suggested a potential advantage of lDRA, we started our experience with lDRA but, based on operator comfort and preference, the use of the rDRA increased over time.
Although the feasibility and benefits of DRA over TRA in STEMI have been observed, the literature on the topic remains scarce.2,9-11 In our registry, all attempted DRA procedures in patients with STEMI were successful. However, the first DRA in STEMI was performed after the operators had surpassed the learning curve for the technique (up to case 320). Similarly, the use of DRA for complex PCI has been previously described.22,26,27 In our cohort, all complex PCI procedures were performed without crossover.
The puncture site in DRA, situated 5 cm distal to TRA, may lead to an inadequate catheter length in specific contexts (such as tall patients, dilated aorta, subclavian artery tortuosity, and the need for retrograde access to PCI for chronic total occlusions).28 We found a low incidence of short catheter length during DRA procedures, with only 1 crossover due to severe tortuosity of the subclavian artery.
DRA-related complications have been consistently reported to be low.2,9-11,18 Similarly, we found a very low rate of complications, the most common being type I-a hematoma. In our study, the incidence of in-hospital RAO was 1%.
The number of DRA procedures to overcome the learning curve and maintain a success rate above 94% is around 150 to 200.2,8 However, in our early experience, we achieved this percentage after the first 20 cases per operator.17 In this study, operators navigated the learning curve in the first quartile; however, success significantly improved to more than 99% in the last 2 quartiles, probably because DRA became the default access for coronary procedures among operators.
Limitations
First, this study was an interim analysis of the leading participating site and coordinator of the DISTAL registry (NTC06165406), conducted because substantial enrollment from other sites was lacking. Although the data cannot be fully extrapolated to other centers, recalculation of the sample size was considered sufficient to evaluate the results.
Second, patient enrollment was not consecutive because the decision to use DRA was at the operators’ discretion. Only one-third of coronary procedures during the study period used this approach. However, we included all patients in whom operators intended to use DRA in any clinical setting were included, with only 21 patients excluded due to DRart ≤1.8mm. Third, this was a descriptive cohort of DRA, without a comparison control group. Fourth, the scale used to assess the arterial pulse is subjective. However, this scale is widely used in routine clinical practice and has been used in multiple DRA studies. Finally, radial artery patency was not evaluated in 9.7% of the patients before discharge, and no evaluation was conducted at 1 month; therefore, the in-hospital rate of radial artery occlusion may be underestimated and no mid-term data are available on the patency of the DRart.
CONCLUSIONS
This study shows the safety and feasibility of DRA primarily guided by ultrasound for coronary procedures in an all-comer population, with high rates of both access and procedural success, in addition to a very low rate of access-related complications.
FUNDING
None declared.
ETHICAL CONSIDERATIONS
This study was approved by the Ethics Committee of our institution (CEIC-2804) and was conducted following the principles of the Declaration of Helsinki. All patients gave their informed written consent before the procedure.
STATEMENT ON THE USE OF ARTIFICIAL INTELLIGENCE
Not used.
AUTHORS’ CONTRIBUTIONS
K. Rivera and D. Fernández-Rodríguez conceived and designed the study. K. Rivera, D. Fernández-Rodríguez, M. García-Guimarães, J. Casanova-Sandoval, and J. L. Ferreiro analyzed data, and drafted the manuscript. All authors contributed to the treatment of patients, data acquisition and mining, and review and approval of the final version of the manuscript.
CONFLICTS OF INTEREST
J. L. Ferreiro reports a) honoraria for lectures from Eli Lilly Co, Daiichi Sankyio, Inc, AstraZeneca, Pfizer, Abbott, Boehringer Ingelheim, Bistol-Myers Squibb, Rovi, Terumo and Ferrer; b) consulting fees from AstraZeneca, Eli Lilly Co, Ferrer, Boston Scientific, Pfizer, Boehringer Ingelheim, Daiichi Sankyo, Inc, Bristol-Myers Squibb and Biotronik; c) research grants from AstraZeneca. The remaining authors have no conflicts of interest to declare.
WHAT IS KNOWN ABOUT THE TOPIC?
- Previous studies have demonstrated the safety and feasibility and safety DRA. Compared with TRA, DRA has several advantages, despite the high prevalence of crossover and controversial incidence of radial artery occlusion.
WHAT DOES THIS STUDY ADD?
- The results of this cohort show the safety and feasibility of DRA in an all-comer population throughout the spectrum of DRart pulses. Our study demonstrates that preprocedure ultrasound evaluation and the ultrasound-guided DRA technique help to achieve a low crossover rate, which is especially useful in patients with an unfavorable arterial pulse. According to our observations, DRA in urgent/emergent procedures and complex PCI is feasible and safe once the learning curve has been overcome and the operator is familiar with the technique.
REFERENCES
1. Babunashvili A, Dundua D. Recanalization and reuse of early occluded radial artery within 6 days after previous transradial diagnostic procedure. Catheter Cardiovasc Interv. 2011;77:530-536.
2. Lee JW, Park SW, Son JW, Ahn SG, Lee SH. Real-world experience of the left distal transradial approach for coronary angiography and percutaneous coronary intervention:A prospective observational study (LeDRA). EuroIntervention. 2018;14:e995-e1003.
3. Oliveira MDP, Navarro EC, Kiemeneij F. Distal transradial access as default approach for coronary angiography and interventions. Cardiovasc Diagn Ther. 2019;9:513-519.
4. Kiemeneij F. Left distal transradial access in the anatomical snuffbox for coronary angiography (ldTRA) and interventions (ldTRI). EuroIntervention. 2017;13:851-857.
5. Sgueglia GA, Di Giorgio A, Gaspardone A, Babunashvili A. Anatomic Basis and Physiological Rationale of Distal Radial Artery Access for Percutaneous Coronary and Endovascular Procedures. JACC Cardiovasc Interv. 2018;11:2113-2119.
6. Lu H, Wu D, Chen X. Comparison of Distal Transradial Access in Anatomic Snuffbox Versus Transradial Access for Coronary Angiography. Heart Surg Forum. 2020;23:E407-E410.
7. Ghose T, Kachru R, Dey J, Khan WU, et al. Safety and Feasibility of Ultrasound-Guided Access for Coronary Interventions through Distal Left Radial Route. J Interv Cardiol. 2022;2022:2141524.
8. Roh JW, Kim Y, Lee OH, et al. The learning curve of the distal radial access for coronary intervention. Sci Rep. 2021;11:13217.
9. Tsigkas G, Papageorgiou A, Moulias A, et al. Distal or Traditional Transradial Access Site for Coronary Procedures:A Single-Center, Randomized Study. JACC Cardiovasc Interv. 2022;15:22-32.
10. Aminian A, Sgueglia GA, Wiemer M, et al. Distal Versus Conventional Radial Access for Coronary Angiography and Intervention:The DISCO RADIAL Trial. JACC Cardiovasc Interv. 2022;15:1191-1201.
11. Kozin´ski Ł, Orzałkiewicz Z, Da˛browska-Kugacka A. Feasibility and Safety of the Routine Distal Transradial Approach in the Anatomical Snuffbox for Coronary Procedures:The ANTARES Randomized Trial. J Clin Med. 2023;12:7608.
12. Ferrante G, Condello F, Rao SV, et al. Distal vs Conventional Radial Access for Coronary Angiography and/or Intervention:A Meta-Analysis of Randomized Trials. JACC Cardiovasc Interv. 2022;15:2297-2311.
13. Barbarawi M, Barbarawi O, Jailani M, Al-Abdouh A, Mhanna M, Robinson P. Traditional versus distal radial access for coronary angiography:A meta-Analysis of randomized controlled trials. Coron Artery Dis. 2023;34:274-280.
14. Erdem K, Kurtogˇlu E, Küçük MA, Ilgenli TF, Kizmaz M. Distal transradial versus conventional transradial access in acute coronary syndrome. Turk Kardiyoloji Dernegi Arsivi. 2021;49:257-265.
15. Valgimigli M, Campo G, Penzo C, Tebaldi M, Biscaglia S, Ferrari R. Transradial coronary catheterization and intervention across the whole spectrum of allen test results. J Am Coll Cardiol. 2014;63:1833-1841.
16. Sgueglia GA, Lee BK, Cho BR, et al. Distal Radial Access:Consensus Report of the First Korea-Europe Transradial Intervention Meeting. JACC Cardiovasc Interv. 2021;14:892-906.
17. Rivera K, Fernández-Rodríguez D, Casanova-Sandoval J, et al. Comparison between the Right and Left Distal Radial Access for Patients Undergoing Coronary Procedures:A Propensity Score Matching Analysis. J Interv Cardiol. 2022;2022:7932114.
18. Oliveira MD, Navarro EC, Caixeta A. Distal transradial access for coronary procedures:A prospective cohort of 3,683 all-comers patients from the DISTRACTION registry. Cardiovasc Diagn Ther. 2022;12:208-219.
19. Hadjivassiliou A, Kiemeneij F, Nathan S, Klass D. Ultrasound-guided access to the distal radial artery at the anatomical snuffbox for catheter-based vascular interventions:A technical guide. EuroIntervention. 2021;16:1342-1348.
20. Calculadora de tamaño muestral GRANMO. Available at:https://www.imim.cat/media/upload/arxius/granmo/granmo_v704.html. Accessed 25 Mar 2024.
21. Lee JW, Kim Y, Lee BK, et al. Distal Radial Access for Coronary Procedures in a Large Prospective Multicenter Registry:The KODRA Trial. JACC Cardiovasc Interv. 2024;17:329-340.
22. Zong B, Liu Y, Han B, Feng CG. Safety and feasibility of a 7F thin-walled sheath via distal transradial artery access for complex coronary intervention. Front Cardiovasc Med. 2022;9:959197.
23. Sgueglia GA, Hassan A, Harb S, et al. International Hand Function Study Following Distal Radial Access:The RATATOUILLE Study. JACC Cardiovasc Interv. 2022;15:1205-1215.
24. Al-Azizi K, Moubarak G, Dib C, et al. Distal Versus Proximal Radial Artery Access for Cardiac Catheterization:1-Year Outcomes. Am J Cardiol. 2024;220:102-110.
25. Rivera K, Fernández-Rodríguez D, Bullones J, et al. Impact of sex differences on the feasibility and safety of distal radial access for coronary procedures:a multicenter prospective observational study. Coron Artery Dis. 2024;35(5):360-367.
26. Rivera K, Fernández-Rodríguez D, García-Guimarães M, Ramírez Martínez T, Casanova-Sandoval J. Intravascular ultrasound-guided percutaneous exclusion of a complicated coronary artery aneurysm presenting as ST-segment elevation myocardial infarction. Coron Artery Dis. 2023;34:527-528.
27. Nikolakopoulos I, Patel T, Jefferson BK, et al. Distal Radial Access in Chronic Total Occlusion Percutaneous Coronary Intervention:Insights From the PROGRESS-CTO Registry. J Invasive Cardiol. 2021;33:E717-E722.
28. Davies RE, Gilchrist IC. Back hand approach to radial access:The snuff box approach. Cardiovasc Revasc Med. 2018;19:324-326.
* Corresponding authors.
E-mail addresses: psrivera.lleida.ics@gencat.cat (K. Rivera); dfernandez.lleida.ics@gencat.cat (D. Fernández-Rodríguez).
ABSTRACT
Introduction and objectives: Drug-eluting balloons (DEB) are an established treatment option for in-stent restenosis (ISR). This study aimed to assess the safety and efficacy of a novel DEB in patients with ISR.
Methods: This prospective, single-center study enrolled a consecutive cohort of patients diagnosed with ISR who underwent coronary angioplasty with a new second-generation paclitaxel-eluting balloon. The 3 main endpoints were myocardial infarction, target lesion revascularization, and target vessel revascularization. Baseline variables were collected, including patient and procedure characteristics. Follow-up data were collected through medical records or telephone contact.
Results: The study included 160 consecutive patients with 206 treated lesions (mean age, 71.4 ± 14.9 years, 15.5% women) undergoing percutaneous coronary intervention with DEB for ISR. A total of 53.3% of patients had acute coronary syndrome. The average diameter of the treated vessel was 3.10 ± 0.7 mm. The DEB used had a mean diameter of 3.1 ± 0.6 mm and a mean length of 23.1 ± 6.8 mm. Predilatation was performed in 98% of the lesions, and a noncompliant balloon was used in 80%. Intracoronary imaging was used in 24% of cases. At the end of the procedure, 98.5% of patients had Thrombolysis in Myocardial Infarction flow grade 3, residual stenosis was > 30% in 3.4%, and dissection occurred in 1.4%. Bail-out stenting was required in 4.8% of patients. Mortality was nil during follow-up (maximum 768 days). The incidence of myocardial infarction, target lesion revascularization, and target vessel revascularization were 5.4% (95%CI, 0.69-10.1), 8.4% (95%CI, 0-17.8), and 14.2% (95%CI, 3.61-24.78), respectively.
Conclusions: In this cohort of patients with ISR treated with DEB, we observed a low rate of adverse events in both the short- and mid-term. These results support the safety and efficacy of this new generation of DEB for treating ISR.
Keywords: In-stent restenosis. Drug-eluting balloon. Paclitaxel.
RESUMEN
Introducción y objetivos: El balón farmacoactivo (BFA) es un tratamiento establecido para tratar la reestenosis intrastent (RIS). El objetivo de este estudio fue valorar la eficacia y la seguridad de un nuevo BFA en pacientes con RIS.
Métodos: Cohorte prospectiva, unicéntrica y consecutiva de pacientes con RIS tratados con angioplastia coronaria con un nuevo balón liberador de paclitaxel de segunda generación. Los 3 eventos principales del estudio fueron infarto de miocardio, revascularización de la lesión diana y revascularización del vaso diana. Se recogieron variables basales, incluidas las características del paciente y del procedimiento. Los datos referentes al seguimiento se obtuvieron de registros médicos o por contacto telefónico.
Resultados: Se incluyeron 160 pacientes consecutivos con 206 lesiones tratadas (71,4 ± 14,9 años, el 15,5% mujeres) que fueron tratados con una intervención coronaria percutánea con BFA debido a RIS. El 53,3% de los pacientes presentaban síndrome coronario agudo. El diámetro medio del vaso tratado fue de 3,1 ± 0,7 mm. El diámetro y la longitud del BFA empleado fueron de 3,1 ± 0,6 mm y 23,1 ± 6,8, respectivamente. El 98% de las lesiones se predilataron y en el 80% se empleó un balón no distensible. El 24% de las angioplastias fueron guiadas por imagen intracoronaria. El 98,5% de los pacientes presentaban un flujo Thrombolysis in Myocardial Infarction de grado 3 al final de la angioplastia. Hubo estenosis residual > 30% en el 3,4%, y el 1,4% presentaron disección. El 4,8% de los pacientes requirieron stent de rescate. Al finalizar el seguimiento (máximo 768 días), ningún paciente había fallecido. Las incidencias de infarto de miocardio, de revascularización de la lesión diana y de revascularización del vaso diana fueron del 5,4% (IC95%, 0,69-10,1), el 8,4% (IC95%, 0-17,8) y el 14,2% (IC95%, 3,61-24,78), respectivamente.
Conclusiones: En esta cohorte de pacientes con RIS tratados con BFA se observa una baja tasa de eventos clínicos adversos, tanto a corto como a mediano plazo. Estos resultados respaldan la eficacia y la seguridad de esta nueva generación de BFA para pacientes con RIS.
Palabras clave: Reestenosis intrastent. Balón farmacoactivo. Paclitaxel.
Abbreviations DEB: drug-eluting balloon. ISR: in-stent restenosis. TLR: target lesion revascularization. TVR: target vessel revascularization.
INTRODUCTION
Patients with coronary in-stent restenosis (ISR) represent a clinical challenge.1 Evidence indicates that these patients are at increased risk of recurrent symptoms, myocardial infarction, and repeated coronary revascularizations.2 The use of drug-eluting balloons (DEB) is a novel alternative therapeutic strategy in patients with ISR.1,3,4 The effect of DEBs in coronary angioplasty is based on the rapid and uniform transfer of antiproliferative drugs into the vessel wall using a single balloon through a lipophilic matrix without the need for permanent implants.5
Over time, new DEB technologies are developed and launched onto the market. The Essential Pro (iVascular, Spain) is a paclitaxel-eluting balloon catheter with advancements to enhance catheter pushability and drug delivery. We believe it is essential to report outcomes from real-world settings. In this study, we report our findings on the safety and efficacy of this new DEB in patients with ISR.
METHODS
Design and population
This prospective, single-center study included a cohort of consecutive patients undergoing DEB angioplasty with the Essential Pro. The center treating these patients performs more than 1500 percutaneous coronary interventions per year. The 2 inclusion criteria for this analysis were: a) use of an Essential Pro DEB and b) its application for ISR treatment. ISR was defined as stenosis more than 50% within the stented segment, and treatment was indicated according to the treating physician’s judgment.6 The use of the Essential Pro DEB was prioritized during the study period to treat all eligible patients for DEB angioplasty, while other DEB devices were rarely used due to inventory constraints. There were no exclusion criteria. Patients may have undergone stent coronary angioplasty of other lesions in the same or a different setting.
Drug-eluting balloon characteristics
The Essential Pro is a paclitaxel-eluting balloon with a uniform 3 μg/mm2 eluting formulation, consisting of paclitaxel (80%) and a biocompatible amphiphilic excipient (20%).7 The balloon incorporates the proprietary TransferTech technology (iVascular, Spain), which is based on the ultrasonic deposition of nanodrops, followed by a dry-off process, resulting in a homogeneous microcrystalline drug coating. This allows more uniform and complete treatment of the vessel with the antiproliferative drug. The microcrystalline structure, coupled with the lipophilic nature of both paclitaxel and the excipient, facilitates drug transfer within 45 to 60 seconds. The Essential Pro balloon has been designed with a smooth transition and a very low tip profile of 0.016 inches, enhancing flexibility, trackability, and device crossability. The balloon is compatible with 5-Fr sheaths in all available diameters.
Procedures
All procedures and decisions in this study reflect real-world clinical practice. Therefore, clinical indications, the use and selection of DEBs, procedural steps, and medical treatments were decided by treating physicians without following any specific guidelines. All coronary angiograms performed during follow-up were part of routine clinical practice and were assessed by our research team when available. Baseline and follow-up data were collected in a single anonymized dedicated database. Procedural aspects, as well as both baseline and follow-up angiograms, were independently evaluated by 3 different interventional cardiologists. Physicians were trained to consult senior staff if they had doubts when assessing angiograms or clinical records. Follow-up was conducted using clinical records, and patients with no on-site clinical visits during follow-up were contacted by telephone following standard clinical practice in our institution. This study was approved by our local institutional review board and patients provided consent for the use of their anonymized information for research purposes before inclusion. This was an investigator-initiated study with no sponsoring or funding.
Outcome definitions
Device delivery was defined as successful DEB insufflation in the affected coronary segment. Procedural, angiographic, and other standard outcomes were defined according to the Second Academic Research Consortium criteria.8 Cardiovascular mortality was defined as any death without a clear noncardiovascular cause. Acute myocardial infarction was defined as any myocardial infarction meeting the fourth version of the Universal Myocardial Infarction Criteria.9 Target lesion revascularization (TLR) was defined as any revascularization within or 5 mm beyond the treated segment.8 Target vessel revascularization (TVR) was defined as revascularization of the index treated vessel.8 Coronary-related hospitalization was defined as a new hospitalization in which a coronary origin was suspected as the main reason for admission. The 3 main efficacy outcomes were myocardial infarction, TLR, and TVR.
Statistical analysis
Categorical variables are presented as percentages, and continuous variables as mean standard deviation (SD) when appropriate. Since the same patient may receive more than 1 DEB (for the same or different territory), the denominator for balloon-specific variables was based on the total DEBs used (such as treated vessel, vessel diameter, DEB diameter, and length), while the denominator of patient-level variables (such as age, sex, or clinical outcomes) was each single individual. Clinical outcomes during follow-up are presented at 30 days, 1 year, and total follow-up. The Kaplan-Meier method was used for estimating both the total follow-up risk and generating survival curves. Data were analyzed using IBM SPSS Statistics 25.
RESULTS
From December 2020 to June 2023, 290 patients with 352 coronary lesions were treated with DEB. Among them, 160 patients (206 lesions) underwent DEB angioplasty due to ISR. Out of the 160 patients receiving DEB for ISR, 46 patients (29%) received more than 1 DEB angioplasty for ISR, either during the same procedure or staged to a different lesion.
The patients’ baseline characteristics are summarized in table 1. The mean age was 71.4 ± 14.9 years, 15.5% were women, and 35.5% had diabetes. Clinical presentation was stable angina in 29.7%, unstable angina in 30.5%, non–ST-segment elevation myocardial infarction in 9.9%, ST-segment elevation myocardial infarction in 12.9%, and 16.7% were asymptomatic.
Patient characteristics | |
Age, y | 71.4 (14.9) |
Sex women | 20 (15.5) |
BMI, kg/m2 | 29.2 (10.5) |
Hypertension | 115 (87.7) |
Active smoking | 8 (6.1) |
Diabetes mellitus | 46 (35.3) |
Previous MI | 67 (51.5) |
Previous CABG | 26 (20) |
Reduced LVEF (< 30%) | 10 (7.6) |
Laboratory parameters | |
Hemoglobin, g/dL | 13.9 (1.5) |
GFR, mL/min/1.73 m2 | 82.9 (25.4) |
Active medication | |
Aspirin | 110 (84.6) |
Clopidogrel | 75 (57.6) |
Ticagrelor | 3 (2.3) |
Prasugrel | 2 (1.5) |
Anticoagulation | 20 (15.2) |
Clinical presentation | |
Silent ischemia | 22 (16.7) |
Stable angina | 39 (29.7) |
Unstable angina | 40 (30.5) |
NSTEMI | 13 (9.9) |
STEMI | 17 (12.9) |
BMI, body mass index; CABG, coronary artery bypass grafting; GFR, glomerular filtration rate; LVEF, left ventricular ejection fraction; MI, myocardial infarction; NSTEMI, non–ST-segment elevation myocardial infarction; STEMI, ST-segment elevation myocardial infarction. Data are expressed as No. (%). |
Procedural characteristics are detailed in table 2. The most commonly treated vessel was the left anterior descending artery (48.7%), followed by the left circumflex (30.7%), and the right coronary artery (17%). Bifurcation was present in 10.7%. Lesion preparation was performed in 98.2% of cases (80% with a noncompliant balloon). Intracoronary imaging was used in 24% of patients. None of the patients underwent rotational atherectomy, and 2.4% underwent balloon lithotripsy before DEB delivery. The mean vessel diameter was 3.1 ± 0.65 mm. The mean DEB diameter was 3.1 ± 0.6 mm, and the mean length was 23.1 ± 6.8 mm. Device delivery was successful in 100% of cases (figure 1). The final angiographic assessment revealed a final dissection in 1.4%, Thrombolysis in Myocardial Infarction flow less than 3 in 1.5%, and residual stenosis more than 30% in 3.4%. Bail-out stenting was needed in 4.8%.
Treated vessel | |
LAD | 100 (48.7) |
LCx | 63 (30.7) |
Right coronary artery | 35 (17) |
Left main coronary artery | 5 (2.4) |
Graft | 2 (0.9) |
Anatomical characteristics | |
Bifurcation lesion | 22 (10.7) |
Vessel diameter, mm | 3.1 (0.65) |
Procedural characteristics | |
IVUS-guided PCI | 51 (24) |
Lesion predilatation | 202 (98) |
Predilatation with NC balloon | 165 (80) |
Intravascular lithotripsy | 5 (2.4) |
DEB diameter, mm | 3.1 (0.6) |
DEB length, mm | 23.1 (6.8) |
Result after DEB PCI | |
Vessel dissection | 3 (1.4) |
TIMI flow 3 | 203 (98.5) |
Residual stenosis > 30% | 194 (3.4) |
Bail-out stenting | 10 (4.8) |
DEB, drug-eluting balloon; IVUS, intravascular ultrasound; LAD, left anterior descending artery; LCx, left circumflex artery; NC, noncompliant; PCI, percutaneous coronary intervention; RCA, right coronary artery; TIMI, Thrombolysis in Myocardial Infarction. Data are expressed as No. (%). |
After discharge, 93.3% of the patients were successfully contacted. The median follow-up was 361 days, including censored patients, with a maximum of 768 days. At 30 days of follow-up, there were no deaths or TLR, there was 1 myocardial infarction (0.6%), TVR occurred in 0.6%, and 6 patients were readmitted to hospital due to a coronary syndrome (4.1%). At the 1-year follow-up, mortality was 0%, myocardial infarction occurred in 3.4%, TLR in 2.5%, TVR in 6.3%, and coronary-related rehospitalizations in 11.8%. At 18 months, the TLR rate was 4.3%. When all available follow-up was included (figure 2), mortality was 0%, myocardial infarction occurred in 5.4% (95% confidence interval [95%CI], 0.69-10.1), TLR in 8.4% (95%CI, 0-17.8), and TVR in 14.2% (95%CI, 3.61-24.78). During follow-up, none of the patients underwent surgical revascularization.
DISCUSSION
This is the first study to describe a real-world experience with the Essential Pro DEB for the treatment of ISR. In this cohort, all attempts at DEB delivery were successful, and less than 1 in 20 patients required bail-out stenting. The use of this new-generation DEB catheter was associated with high efficacy and a low incidence of adverse clinical outcomes during follow-up.
Patients with ISR are at higher risk of recurrent events than those undergoing non-ISR angioplasty.10 The annual rate of ISR requiring TLR is around 2%,3 representing up to 11% of all percutaneous coronary interventions performed in the United States.11,12 Notably, 52% of patients presenting with symptomatic ISR have unstable angina, and up to 27% have an acute myocardial infarction.12 Therefore, ISR poses a significant clinical challenge due to both its frequency and severity. The use of DEB in the ISR scenario avoids the addition of extra stent layers, which may have detrimental effects in the long term.
The use of DEB in ISR poses certain challenges. DEB platforms commonly have lower lesion crossability than regular coronary balloon catheters. DEBs also have larger profiles than conventional balloons making it difficult to cross the lesion and requiring aggressive maneuvers that could lead to a loss of coating drug during delivery.13 However, in our study, all attempted DEB deployments were successful. This high success rate may be due to improvements in second-generation DEBs, as well as better lesion evaluation and lesion preparation.
In the present study, TLR occurred in 2.5% of the patients and TVR in 6.3% at 1 year, while TLR occurred in 4.3% at 18 months. This event rate may seem low when compared with a prior systematic review of randomized and observational studies, which reported a TVR rate after DEB treatment of 11.3% with a calculated weighted mean follow-up of 18 months.14 In a recent investigational device exemption randomized trial for a paclitaxel-coated balloon in ISR, the rate of TLR at 1 year was 13%.15 However, prior evidence stems from diverse settings, designs, and populations, making it difficult to draw strong conclusions.
The rate of TLR with the previous generation of the Essential Pro DEB in a smaller cohort (n = 31) was 10% at 6 months.16 While this rate may seem higher than that reported in our study, the small number of events (n = 3) makes comparisons challenging.
Limitations
This study has some limitations. First, it was based on a real-world cohort involving different operators from the same center, which does not follow specific protocols. Only a quarter of the patients underwent angioplasty assessment guided by intracoronary imaging. The lack of sponsorship to cover intracoronary imaging costs and its limited use reflects the usual clinical practice of this center. During the performance of this study, few patients with ISR were treated with other DEB catheters due to the lack of specific DEB sizes in stock. Since this situation was rare and was unrelated to clinical or medical coverage characteristics, it is unlikely to introduce significant bias. Since this was a substudy of a larger DEB cohort, some variables specific to ISR, such as the time from prior stent implantation or the type of stent used, were not available.
Second, there were no dedicated follow-up visits for this study. Although most of these patients were followed up by local cardiologists who maintained regular medical records, some required telephone contact for follow-up. Third, angiographic assessment was not duplicated, and no core lab was available. Finally, the number of events was low despite consecutive enrollment from late 2020, impacting the precision of Kaplan-Meier estimates for key clinical outcomes. Some limitations are related to real-world practice settings, which, on the other hand, enhance external validity with less selection bias compared with other more controlled designs.
CONCLUSIONS
Among patients with ISR, the Essential Pro DEB catheter had a high delivery rate and a low incidence of adverse clinical outcomes during follow-up. These results further underscore the safety and efficacy of this new-generation DEB for patients with ISR.
FUNDING
This work received no industry sponsoring or funding.
ETHICAL CONSIDERATIONS
This study was approved by our local institutional review board at the Instituto Cardiovascular de Buenos Aires, and patients provided written informed consent to use their anonymized information for research purposes before their inclusion. Possible sex/gender biases have been considered in the preparation of this paper.
STATEMENT ON THE USE OF ARTIFICIAL INTELLIGENCE
No artificial intelligence tool was used in the preparation of this study.
AUTHORS’ CONTRIBUTIONS
L. Padilla conceived and oversaw all the process. F. Liberman, J. Tello, P. Rosas, P. Spaletra, G. Pedernera, P. Mascolo, S. Ordoñez, P. Santilli, and A. Candiello collected data and analyzed coronary angiograms. F. Cura and J. Belardi provided senior advice. P. Lamelas performed the statistical analysis and generated the first draft of the manuscript.
CONFLICTS OF INTEREST
L. Padilla has received proctoring and consulting honoraria from Terumo and Boston Scientific. P. Spaletra has received honoraria from Boston Scientific. F. Cura has received honoraria from Medtronic, Boston Scientific, Terumo, and Meril. P. Lamelas has received proctoring and consulting honoraria from Medtronic, Boston Scientific, Meril, Microport. The remaining authors have no conflicts of interest to declare.
WHAT IS KNOWN ABOUT THE TOPIC?
- Patients with ISR are at high risk of recurrent events and are commonly treated with DEB. New or newer generation DEBs are frequently launched onto the market. It is important to report the real-world safety and efficacy of interventional devices. The Essential Pro is a secondgeneration paclitaxel-eluting balloon. Enhancements of this DEB include improvements in forward pushability, crossover capacity, and drug delivery capabilities.
WHAT DOES THIS STUDY ADD?
- Using this new-generation DEB, all attempts at treating ISR (n = 206) were successful. Intravascular ultrasound was used in 24%. The incidence of adverse events, from the procedure to mid-term follow-up, was infrequent and probably lower than that previously reported. These realworld results emphasize the safety and efficacy of this novel generation DEB for patients with ISR.
REFERENCES
1. Giacoppo D, Saucedo J, Scheller B. Coronary Drug-Coated Balloons for De Novo and In-Stent Restenosis Indications. J Soc Cardiovasc Angiogr Interv. 2023. https://doi.org/10.1016/j.jscai.2023.100625.
2. Pleva L, Kukla P, Hlinomaz O. Treatment of coronary in-stent restenosis:A systematic review. J Geriatr Cardiol. 2018;15:173-184.
3. Giustino G, Colombo A, Camaj A, et al. Coronary In-Stent Restenosis:JACC State-of-the-Art Review. J Am Coll Cardiol. 2022;80:348-372.
4. Indermuehle A, Bahl R, Lansky AJ, et al. Drug-eluting balloon angioplasty for in-stent restenosis:A systematic review and meta-analysis of randomised controlled trials. Heart. 2012;99:327-333.
5. Jeger R V., Eccleshall S, Wan Ahmad WA, et al. Drug-Coated Balloons for Coronary Artery Disease:Third Report of the International DCB Consensus Group. JACC Cardiovasc Interv. 2020;13:1391-1402.
6. Klein LW, Nathan S, Maehara A, et al. SCAI Expert Consensus Statement on Management of In-Stent Restenosis and Stent Thrombosis. J Soc Cardiovasc Angiogr Interv. 2023;2:100971.
7. Pérez de Prado A, Pérez-Martínez C, Cuellas Ramón C, et al. Safety and Efficacy of Different Paclitaxel-eluting Balloons in a Porcine Model. Rev Esp Cardiol. 2014;67:456-462.
8. Garcia-Garcia HM, McFadden EP, Farb A, et al. Standardized end point definitions for coronary intervention trials:The academic research consortium 2 consensus document. Circulation. 2018;137:2635-2650.
9. Domienik-Karlowicz J, Kupczyn´ska K, Michalski B, et al. Fourth universal definition of myocardial infarction. Selected messages from the european society of cardiology document and lessons learned from the new guidelines on st-segment elevation myocardial infarction and non-st-segment elevation-acute coronary syndrome. Cardiol J. 2021;28:195-201.
10. Steinberg DH, Pinto Slottow TL, Buch AN, et al. Impact of In-Stent Restenosis on Death and Myocardial Infarction. Am J Cardiol. 2007;100:1109-13.
11. Madhavan M V, Kirtane AJ, Redfors B, et al. Stent-Related Adverse Events >1 Year After Percutaneous Coronary Intervention. J Am Coll Cardiol. 2020;75:590-604.
12. Moussa ID, Mohananey D, Saucedo J, et al. Trends and Outcomes of Restenosis After Coronary Stent Implantation in the United States. J Am Coll Cardiol. 2020;76:1521-1531.
13. Yoshida R, Ishii H, Morishima I, et al. Impact of adjunctive use of guide extension catheter on midterm outcome of drug-coated balloon angioplasty. EuroIntervention. 2019;15:688-691.
14. Cui KY, Lyu SZ, Zhang M, Song XT, Yuan F, Xu F. Drug-Eluting Balloon versus New-Generation Drug-Eluting Stent for the Treatment of In-Stent Restenosis:An Updated Systematic Review and Meta-Analysis. Chin Med J (Engl). 2018;131:600-607.
15. Yeh RW, Shlofmitz R, Moses J, et al. Paclitaxel-Coated Balloon vs Uncoated Balloon for Coronary In-Stent Restenosis:The AGENT IDE Randomized Clinical Trial. JAMA. 2024;331:1015-1024.
16. de la Torre Hernández JM, Garcia Camarero T, Lozano Ruiz-Poveda F, et al. Angiography and Optical Coherence Tomography Assessment of the Drug-Coated Balloon ESSENTIAL for the Treatment of In-Stent Restenosis. Cardiovasc Revasc Med. 2020;21:508-513.
* Corresponding author.
E-mail address: Ltpadilla@icba.com.ar (L. Padilla).
ABSTRACT
Introduction and objectives: Functional assessment of coronary stenosis severity with the piezo-electric sensor pressure wire has shown a discrepancy of up to 20% between hyperemic and nonhyperemic indexes. No data are available with fiber-optic pressure wires. The aim of this study was to evaluate the incidence and factors related to the diagnostic discordance between these indexes with a fiber-optic pressure wire. Secondary aims were to assess diagnostic reproducibility in 2 consecutive measurements of fractional flow reserve (FFR) and diastolic pressure ratio (dPR) and evaluate the drift rate.
Methods: We conducted a prospective, observational multicenter study in patients undergoing functional assessment with a fiber-optic pressure wire. We took 2 consecutive measurements of the dPR (cutoff point 0.89) and FFR (cut-off point 0.80) in each lesion analyzed. The diagnostic correlation between 2 measurements with the same technique and between the 2 techniques (dPR and FFR) was assessed. Clinical and angiographic factors associated with discordance (FFR−/dPR+ and FFR+/dPR−) between the 2 techniques were analyzed.
Results: We included 428 cases of stenosis (361 patients). Diagnostic reproducibility was 95.8% for the dPR, with a correlation coefficient between the 2 measurements (dPR1 and dPR2) of 0.974 (P < .0001). For FFR, the diagnostic reproducibility was 94.9% with a correlation coefficient (FFR1 and FFR2) of 0.942 (P < .0001). The diagnostic discordance was 18.2% (FFR+/dPR− 8.2% and FFR−/dPR+ 10%). Among the variables analyzed, the factors significantly associated with FFR−/dPR+ discordance in the multivariate analysis were hypertension and intracoronary adenosine. The only factors significantly associated with FFR+/dPR− discordance were age < 75 years and stenosis > 60%. The drift rate was 5.7%.
Conclusions: Although FFR and dPR measurements with a fiber-optic pressure wire have excellent reproducibility and a low drift rate, the discordance rate remains similar to those in previous studies with a piezo-electric pressure wire. FFR−/dPR+ discordance is associated with intracoronary adenosine and hypertension. FFR+/dPR− discordance is related to age < 75 years old and stenosis > 60%.
Keywords: Coronary physiology. Fractional flow reserve. Nonhyperemic index. Discordance. Drift.
RESUMEN
Introducción y objetivos: La valoración funcional de las estenosis coronarias con guías de presión de sensor piezoeléctrico ha mostrado hasta un 20% de discordancia entre los índices hiperémico y no hiperémico. No hay datos disponibles con guía de presión de sensor óptico. El objetivo del estudio es evaluar la incidencia y los factores relacionados con la discordancia diagnóstica entre estos índices con guía de presión de sensor óptico. Como objetivos secundarios se evaluó la reproducibilidad diagnóstica en dos determinaciones consecutivas de la reserva fraccional de flujo (RFF) y la diastolic pressure ratio (dPR). También se evaluó la tasa de drift.
Métodos: Estudio observacional, prospectivo, multicéntrico, en pacientes a quienes se realiza una valoración funcional con guía de presión de sensor óptico. Se hicieron dos mediciones consecutivas de dPR (umbral 0,89) y RFF (umbral 0,80) en cada lesión analizada. Se valoró la correlación diagnóstica entre dos mediciones con la misma técnica y entre ambas técnicas (dPR y RFF). Se analizaron factores clínicos y angiográficos asociados a la discordancia (RFF−/dPR+ y RFF+/dPR−) entre ambas técnicas.
Resultados: Se incluyeron 428 estenosis (361 pacientes). La reproducibilidad diagnóstica fue del 95,8% para dPR, con un coeficiente de correlación entre ambas mediciones (dPR1 y dPR2) de 0,974 (p < 0,0001). Para RFF la reproducibilidad diagnóstica fue del 94,9%, con un coeficiente de correlación (RFF1 y RFF2) de 0,942 (p < 0,0001). La discordancia diagnóstica fue del 18,2% (RFF+/dPR− 8,2% y RFF−/dPR+ 10%). Entre las variables analizadas, en el análisis multivariado, la hipertensión arterial y la administración intracoronaria de adenosina se asociaron de manera significativa con la discordancia RFF−/dPR+. Solo la edad < 75 años y la estenosis > 60% se asociaron de manera significativa con la discordancia RFF+/dPR−. La tasa de drift fue del 5,7%.
Conclusiones: Aunque las mediciones de RFF y dPR con guía de presión de sensor óptico tienen una excelente reproducibilidad y una baja incidencia de drift, la tasa de discordancia permanece similar a la de estudios previos con guía de presión de sensor piezoeléctrico. La adenosina intracoronaria y la hipertensión arterial se asocian con la discordancia RFF−/dPR+. La edad < 75 años y la estenosis > 60% se asocian a discordancia RFF+/dPR−.
Palabras clave: Fisiología coronaria. Reserva fraccional de flujo. Índice no hiperémico. Discordancia. Drift.
Abbreviations dPR: diastolic pressure ratio. FFR: fractional flow reserve. FOSW: fiber-optic sensor wire. iFR: instantaneous wave-free ratio: PPSW: piezoelectric pressure sensor wire.
INTRODUCTION
Fractional flow reserve (FFR) measurement is an invasive procedure performed during coronary angiography to determine the functional significance of coronary stenoses.
In recent years, the instantaneous wave-free ratio (iFR) resting index has been developed to assess the functional significance of coronary stenoses without the need for adenosine administration. The optimal iFR cutoff value—equivalent to 0.80 in FFR—was initially established at 0.89.1 In 2017, 2 clinical studies comparing FFR with iFR found no significant differences in clinical outcomes at follow-up.2-3 After the publication of these 2 studies, the European Society of Cardiology guidelines on myocardial revascularization4 assigned resting indices the same grade of recommendation as FFR for the functional assessment of coronary lesions.
Despite the validation of these 2 techniques in clinical trials and their inclusion in clinical practice guidelines, up to 20% discordance has been reported between iFR+/FFR− or iFR−/FFR+5 Several clinical factors, such as diabetes,6 and anatomical factors, such as lesion location in the left main or proximal left anterior descending coronary arteries, have been identified in association with this discordance.7
Previous studies comparing FFR with iFR using a piezoelectric pressure sensor wire (PPSW) calculated the mean distal-to-aortic pressure ratio beginning 25% into diastole and ending 5 ms before end diastole.1
Recently, a new resting index—the diastolic pressure ratio (dPR)—has been developed to calculate the mean distal-to-aortic pressure ratio over the entire diastolic phase (from the lowest point of the dicrotic notch up to 50 ms before the onset of the upstroke of the next beat)8 using a fiber-optic sensor wire (FOSW).
A study that compared the values of different resting indices (iFR, dPR, dPR25-75, dPRmid, iFRmatlab, iFR50ms, and iFR100ms) revealed that all were numerically identical,8 meaning that the results obtained with the iFR can be extrapolated to other resting indices.
To date, no study has compared the agreement between dPR and FFR measured using a FOSW. One advantage of the FOSW over the PPSW is the lower loss of mean pressure matching in the wire compared with the measurement obtained in the guide catheter (drift).9 Although various iFR studies state that drifts < ± 0.02 are considered acceptable, the drifts reported with the FOSW were even lower at < ± 0.01.10
The diagnostic reproducibility of PPSW decreases significantly when close to the threshold value of 0.80 and is approximately 80% when measurements are < 0.77 or > 0.83, and around 90% with values < 0.76 or > 0.84.11 Since the FOSW is less sensitive to changes in humidity and temperature, greater reproducibility of results can be expected when the measurement is repeated.
Considering that most discordant measurements have been associated with cutoff values, the better reproducibility of measurements and practically nonexistent drift of the FOSW can more accurately determine FFR and dPR measurements and reduce discrepancies.
METHODS
Study design
In this prospective, observational, and multicenter registry of consecutive coronary stenoses, we conducted a study with FOSW based on our routine clinical practice.
We included consecutive patients with clinical signs and coronary angiography findings suggesting the need for a functional study with a pressure wire. We excluded patients with cardiogenic shock, heart failure, severe anemia (hemoglobin < 10 mg/dL), heart rate < 50 or > 100 bpm, baseline systolic blood pressure < 90 mmHg or > 160 mmHg, severe coronary artery lesions in distal segments, and contraindications for the administration of adenosine.
Objective
The aim of this study was to evaluate the incidence and factors related to diagnostic discrepancies between these indices using the FOSW. Secondary aims consisted of assessing the diagnostic reproducibility of FOSW in 2 consecutive measurements of FFR and dPR and evaluating the drift rate.
Procedure
The study was approved by the Drugs Research Ethics Committee of the Basque Country (internal code PS 2019039). All patients received information on the study and were asked to sign a written informed consent form prior to their participation in the study.
We performed coronary angiography using standard methods, with visual estimation of severity after intracoronary nitroglycerin administration. We included lesions with up to 50% to 75% percent diameter stenosis and collected data on the reference luminal diameter, minimum luminal diameter, lesion length, calcification, and vessel tortuosity for each studied lesion.
We performed 2 consecutive measurements of dPR (threshold, 0.89) and FFR (threshold, 0.80) for each studied lesion and analyzed the clinical and angiographic factors to determine their correlation with discordance (FFR−/dPR+ and FFR+/dPR−). We took dPR1 and FFR1 as reference values for discrepancy analysis.
We conducted the FOSW functional study with 5-, 6-, or 7-Fr guide catheters without side holes, using an OptoWire (Opsens Medical, Canada). After advancing the wire toward the tip of the guide catheter, we removed the introducer sheath and flushed the system with saline solution to prevent damping of the pressure wire resulting in equal pressure of the wire and the guide catheter at the tip of the catheter. After advancing the pressure wire distally, we administered 200 μg of intracoronary nitroglycerin before taking any measurements. We took the 2 dPR measurements after waiting the necessary time to obtain confirmation of a stable baseline distal-to-aortic coronary pressure ratio (Pd/Pa).
Subsequently, we took 2 different FFR measurements. Hyperemia was induced according to standard practice in each center (through intracoronary or IV adenosine infusion). If intracoronary adenosine was infused, for the second measurement, we waited until the baseline heart rate, blood pressure, and Pd/Pa were regained and then infused the same dose of adenosine. If IV adenosine was infused, the infusion was stopped until baseline heart rate, blood pressure, and Pd/Pa were regained, and then we infused adenosine at the same rate.
We evaluated the presence of drift upon removal of the pressure wire from the guide catheter. Drift was defined as a difference in Pd/Pa of at least ± 0.02 upon removal of the pressure wire from the guide catheter. In the presence of significant drift, measurements were repeated.
Cutoff values
The cutoff value was ≤ 0.80 for FFR and ≤ 0.89 for dPR.10 We categorized all studied vessels based on dPR and FFR values into 4 groups: concordant positive group (FFR ≤ 0.80 and dPR ≤ 0.89), concordant negative group (FFR > 0.80 and dPR > 0.89), discordant FFR+/dPR− group (FFR ≤ 0.80 and dPR > 0.89), and discordant FFR−/dPR+ group (FFR > 0.80 and dPR ≤ 0.89).
Statistical analysis
Continuous variables are expressed as mean and standard deviation, while categorical variables are expressed as percentages. We measured the association between continuous variables using Pearson’s correlation coefficient. To determine differences in variables in the FFR/dPR concordance groups we used ANOVA (for continuous variables) and the chi-square test (for categorical variables). We used the chi-square test to assess how each variable impacted FFR−/dPR+ and FFR+/dPR− discrepancies, and a multiple logistic regression model with backward elimination to determine the factors impacting FFR−/dPR+ and FFR+/dPR− discrepancies. On univariate analysis, we included variables with P < .1 in the logistic regression analysis and excluded those with a total n < 10. The analysis was conducted using SPSS software (version 20.1) and R (version 4.0.4).
RESULTS
We included a total of 428 stenoses in 361 patients. Table 1 and table 2 show the patients’ baseline characteristics, clinical presentation, and procedural characteristics.
N = 361 | |
---|---|
Age (years) | 65.80 ± 10.5 |
Male sex | 76.9 |
Hypertension | 63.3 |
Diabetes mellitus | 31 |
Hypercholesterolemia | 60.4 |
Active/former smoker | 19.7/40.5 |
Previous acute coronary syndrome | 30.5 |
Atrial fibrillation | 14.7 |
Heart failure/dysfunction | 15.4 |
Peripheral artery disease | 10 |
Valvular heart disease, previous bypass, stroke | < 6 |
Data are expressed as No. (%) mean ± standard deviation. |
N = 361 | |
---|---|
Clinical presentation | N = 361 |
Chest pain | 45.8 |
Acute coronary syndrome | 23.1 |
Unstable angina | 7.1 |
Left ventricular dysfunction | 9.9 |
Others | 14.2 |
Procedural characteristics | |
Baseline systolic blood pressure (mmHg) | 132 ± 24 |
Systolic blood pressure during hyperemia (mmHg) | 125 ± 25 |
Baseline heart rate (bpm) | 70 ± 12 |
Heart rate during hyperemia (bpm) | 69 ± 15 |
Reference luminal diameter (mm) | 3.09 ± 0.53 |
Stenosis (%) | 54 ± 8 |
Lesion length (mm) | 17.9 ± 12.2 |
IV/intracoronary adenosine | 33/67 |
Catheter size (5-Fr/6-Fr) | 17.5/81 |
Drift ≥ ± 0.02 | 5.7 |
dPR | 0.90 ± 0.08 |
FFR | 0.83 ± 0.08 |
dPR, diastolic pressure ratio; FFR, fractional flow reserve. Data are expressed as No. (%) mean ± standard deviation. |
Sixty-seven percent of the patients received intracoronary adenosine; the mean doses of intracoronary adenosine administered were 324 μg (standard deviation [SD] ± 152) via the right coronary artery and 442 μg (SD ± 234) via the left coronary artery.
The medians of dPR measurements were 0.90 and 0.90 (SD ± 0.08) for the first and second measurements, with positivity rates of 27.4% and 27.9%, respectively. For FFR, the medians were 0.83 and 0.83 (SD ± 0.08) for the first and second measurements, with positivity rates of 28.1% and 30%, respectively.
The most widely studied vessel was the left anterior descending coronary artery (63%), followed by the left circumflex (20%) and right coronary arteries (16%).
The left anterior descending coronary artery showed a higher positivity rate (dPR+, 35.3%; FFR, 34%) than the left circumflex (dPR, 11.9%; FFR, 20.5%) and right coronary arteries (dPR, 15.9%; FFR, 17.4%).
Diagnostic reproducibility was 95.8% for dPR, with a correlation coefficient between the 2 measurements (dPR1 and dPR2) of 0.974 (P < .0001) and a mean difference of 0.019 (max, 0.12; min, −0.17). For dPR values < 0.86 or > 0.92, diagnostic reproducibility was 99.6%, decreasing to 90.7% when values were ≥ 0.86 or ≤ 0.92. For FFR, diagnostic reproducibility was 94.9%, with a correlation coefficient (FFR1 and FFR2) of 0.942 (P < .0001) and a mean difference of 0.029 (max, 0.14; min, −0.18) (figure 1). Values < 0.77 or > 0.83 showed a diagnostic reproducibility of 98.6%, decreasing to 86.4% when these values were ≥ 0.77 or ≤ 0.83.
The diagnostic concordance (figure 2) between FFR and dPR was 82%, with a correlation coefficient of 0.721 (P < .0001), while diagnostic discordance was 18.2% (FFR+/dPR–, 8.2% and FFR–/dPR+, 10.0%). In the FFR+/dPR– discordant group, FFR was 0.76 ± 0.04 and dPR, 0.93 ± 0.03. In the FFR–/dPR+ discordant group, FFR was 0.84 ± 0.03 and dPR, 0.86 ± 0.03.
Out of the 75 discordant results reported, the measurements at the cutoff value (7 stenoses with FFR 0.80 and 18 stenoses with dPR 0.89) showed a discordance rate of 72%, which decreased as it moved away from the cutoff value (figure 3).
Table 1 of the supplementary data illustrates the association between clinical and anatomical characteristics and the extent of agreement between FFR and dPR.
Out of all the variables analyzed in the multivariate analysis, hypertension (odds ratio [OR], 3.48, 95% confidence interval [95%CI], 1.01-11.98; P = .043) and intracoronary adenosine (OR, 7.04; 95%CI, 1.63-30.3; P = .001) were significantly associated with FFR–/dPR+ discordance. Age younger than 75 years (OR, 4.52; 95%CI, 1.03-20; P = .016) and percent diameter stenosis > 60% (OR, 6.69; 95%CI, 2.79-16; P < .001) were significantly associated with FFR+/dPR– discordance (table 3).
Variables | FFR+/dPR− | FFR−/dPR+ | ||||||
---|---|---|---|---|---|---|---|---|
Univariate analysis | Multivariate logistic regression | Univariate analysis | Multivariate logistic regression | |||||
OR (95%CI) | P | OR (95%CI) | P | OR (95%CI) | P | OR (95%CI) | P | |
Age < 75 years | 9.5 vs 3.3 | .039 | 4.52 (1.03-20) | .016 | 7.1 vs 8.9 | .347 | ||
Female sex | 5.7 vs 8.9 | .231 | 11.4 vs 6.2 | .079 | ||||
Hypertension | 7.2 vs 10.4 | .178 | 10 vs 2.2 | .002 | 3.48 (1.01-11.98) | .043 | ||
Diabetes mellitus | 7.3 vs 8.6 | .406 | 11.7 vs 5.3 | .018 | 2.11 (0.95-4.69) | .064 | ||
Dyslipidemia | 7.5 vs 9.5 | .304 | 7.1 vs 6.7 | .525 | ||||
HF/LV dysfunction | 4.4 vs 9.0 | .154 | 11.8 vs 6.7 | .118 | ||||
Valvular heart disease | 7.7 vs 8.3 | .635 | 19.2 vs 6.8 | .037 | ||||
Coronary calcification | 6.7 vs 8.6 | .387 | 5.3 vs 8 | .302 | ||||
Moderate/severe tortuosity | 7.8 vs 8.3 | .516 | 10.6 vs 5.7 | .054 | ||||
Left main coronary artery | 0 vs 7.9 | .612 | 50 vs 6.9 | .007 | ||||
Left anterior descending coronary artery | 7.9 vs 7.6 | .531 | 9.4 vs 4.4 | .041 | ||||
Right coronary artery | 10.8 vs 7.1 | .176 | 2.4 vs 8.8 | .029 | ||||
Left circumflex artery | 4.3 vs 8.5 | .179 | 2.9 vs 8.4 | .081 | ||||
RLD > 3 mm | 6.6 vs 13.3 | .049 | 7.5 vs 8 | .518 | ||||
Length > 20 mm | 12.5 vs 5.4 | .010 | 7.8 vs 7.3 | .492 | ||||
Stenosis > 60% | 16 vs 3 | < .001 | 6.69 (2.79-16) | < .001 | 5.8 vs 8.3 | .227 | ||
Heart rate > 80 bpm | 8.4 vs 8 | .527 | 10.8 vs 6.8 | .155 | ||||
Intracoronary adenosine | 7.4 vs 8.5 | .713 | 13 vs 3.8 | .004 | 7.04 (1.63-30.3) | .001 | ||
95%CI, 95% confidence interval; dPR, diastolic pressure ratio; FFR, fractional flow reserve; HF, heart failure; LV, left ventricle; OR, odds ratio; RLD, reference luminal diameter. Data are expresed in %. |
The drift rate was 5.7%.
DISCUSSION
We present the results of the first study conducted with a FOSW capable of measuring the diagnostic variability of 2 consecutive determinations of nonhyperemic and hyperemic indices, as well as the diagnostic discordance between the 2 techniques.
Previous discordance studies between the 2 indices with PPSW revealed discordance rates ranging from 12% to 22%,12,13 largely depending on the proximity of the values to the cutoff point. In a study by Lee et al.,12 the mean iFR and FFR values were 0.95 ± 0.10 and 0.87 ± 0.11, respectively, with a discordance rate of 12%, while in a study by Warisawa et al.,13 the mean iFR and FFR values were 0.89 ± 0.05 and 0.80 ± 0.03, respectively, with a discordance rate of 22%. In our study, the discordance rate was 18.2%, with a mean dPR of 0.90 (SD ± 0.08) and a mean FFR of 0.83 (SD ± 0.08), which is a slightly lower discordance rate than that reported by previous studies on PPSW and mean iFR and FFR values close to the cutoff point, which may be indicative of the accuracy of measurements obtained with FOSW.
The main findings of this study were the excellent diagnostic reproducibility of the FOSW, the clinical and anatomical variables related to FFR/dPR discordance, and the low drift rate reported in the measurements.
Diagnostic reproducibility with the fiber-optic sensor wire
Diagnostic reproducibility with the FOSW was excellent, with a variation between 2 consecutive measurements < 0.02 for dPR and < 0.03 for FFR. This accuracy in measurement confers excellent diagnostic reproducibility. These data are better than those previously reported with PPSW.11
Clinical and anatomical variables associated with FFR/dPR discordance
For FFR+/dPR− discordance, in the multivariate analysis, only age younger than 75 years and percent diameter stenosis > 60% were significantly associated with FFR+/dPR− discordance. This discordance in participants younger than 75 years could be explained by a slower baseline flow and a greater coronary flow reserve in younger patients with preserved microvascular function.14,15 Although discordance due to a higher percent diameter stenosis has already been described in previous studies,15,16 such discordance requires a preserved coronary flow reserve.6 When arterial flow velocity significantly increases during hyperemia, the pressure gradient does so too, decreasing distal coronary pressure during hyperemia substantially compared with baseline values, resulting in a low FFR value.
For FFR−/dPR+ discordance, in the multivariate analysis, the associated variables were hypertension and the administration of intracoronary adenosine. Although hypertension has not been associated with FFR−/dPR+ discordance in previous studies, it is known that patients with hypertension and left ventricular hypertrophy have a reduced coronary flow reserve17 and a possible lack of vasodilatory response to adenosine due to an increased left ventricular end-diastolic pressure. These 2 factors could play a key role in the association between hypertension and FFR−/dPR+ discordance.
Although IV adenosine is the most widely studied route of administration to achieve maximum hyperemia, intracoronary adenosine at doses > 300 μg may be equally or more effective in achieving maximum hyperemia18 and with fewer adverse events.19 In our study, the FFR−/dPR+ discordance reported when intracoronary adenosine was used could be a result of a failure to achieve adequate hyperemia.
These variables related to discordance demonstrate that dPR and FFR measure different aspects of coronary circulation, which may be affected differently in distinct patients or myocardial territories, leading to discordant FFR values and nonhyperemic indices.20
Drift in the fiber-optic pressure wire
The incidence of drift in clinical studies of pressure wires is not well known, and the drift considered acceptable has varied over the years. Previously, FFR measurement was repeated when drift was > 5 mmHg,21 while in more recent studies, drift > 3 mmHg has been considered significant. When FFR is between 0.77 and 0.82, drift ≤ 3 mmHg can reclassify 18.7% of stenoses,22 and this reclassification may be higher when a nonhyperemic diastolic or whole-cycle index is used.23 In the CONTRAST trial analysis of the PPSW, the drift rate (Pd/Pa ± 0.03) was 17.5%,24 while a more recent study comparing drift between FOSW and PPSW revealed a significantly lower rate with the FOSW (4.8% vs 26.7%; P = .02).9 In our study, the drift rate was 5.7%, which is consistent with other studies on FOSW, and much lower than that reported with PPSW, facilitating the use of pressure wire in routine clinical practice.
Limitations
Our study has several limitations. Both the severity and length of coronary lesions were quantified by the operator’s visual estimation at the time of the procedure, and since this was a study without a core laboratory, we cannot rule out the possibility that some of the discrepancies found were due to technical problems in determining the indices. Since the study was based on our routine clinical practice, most patients received intracoronary adenosine, and the protocol did not specify the intracoronary infusion comprehensively, which may have resulted in the lower hyperemia reported in some patients.
Target lesion revascularization was based on dPR or FFR values according to the operators’ decision. Patient selection for pressure guidance evaluation was also left to the treating physician’s discretion, which may have resulted in biases. However, our intention was to study dPR and FFR indices under real-world conditions.
CONCLUSIONS
Although FFR and dPR measurements with FOSW have excellent reproducibility and a low incidence of drift, the discordance rate remains similar to that reported by previous studies with PPSW, and largely depends on the proximity of values to the cutoff point. Intracoronary adenosine and hypertension, which imply a lack of hyperemia or increased microvascular resistance, are associated with FFR−/dPR+ discordance. Age younger than 75 years and the severity of stenosis, which may be associated with a preserved coronary flow reserve, are related to FFR+/dPR− discordance.
FUNDING
This study received no funding.
ETHICAL CONSIDERATIONS
This study was approved by the Drugs Research Ethics Committee of the Basque Country (internal code PS 2019039) for its implementation. All patients received a patient information sheet about the study and signed an informed consent form before enrollment. The study took into consideration sex and gender variables before drafting this article.
STATEMENT ON THE USE OF ARTIFICIAL INTELLIGENCE
No artificial intelligence has been used.
AUTHORS’ CONTRIBUTIONS
M. Sádaba Sagredo drafted the protocol, included patients as the lead investigator of his center, and drafted the manuscript. A. Subinas Elorriaga and A. Quirós contributed to the statistical analysis and drafting of the manuscript. The remaining authors are lead investigators of the READI EPIC-14 trial in their respective centers and contributed to patient inclusion and article review.
CONFLICTS OF INTEREST
None declared.
WHAT IS KNOWN ABOUT THE TOPIC?
- Determination of fractional flow reserve (FFR) is a widely used technique to establish the functional significance of coronary stenoses. In recent years, resting indices have been developed to assess the functional significance of coronary stenoses without the need for adenosine administration. The optimal cutoff value—equivalent to 0.80 in FFR—has been established at 0.89. Despite its validation in clinical trials and endorsement in clinical practice guidelines, discordant results are obtained in up to 20% of the cases between the 2 techniques.
WHAT DOES THIS STUDY ADD?
- Studies on discordance between hyperemic and nonhyperemic indices are conducted with piezoelectric pressure sensor wires. Fiber-optic sensor wires are not sensitive to temperature or humidity changes, making measurements more reproducible and drift rates very low.
- No previous studies have compared the concordance between hyperemic and nonhyperemic indices with the use of a fiber-optic sensor wire. –Despite the low diagnostic variability of diastolic pressure ratio (dPR) and FFR (4.2% for dPR and 5.1% for FFR) in 2 consecutive measurements, and a similarly low drift rate (5.7%), the discrepancy between the 2 indices remains similar to that reported by previous studies (18.2%), indicating that discrepancies are more related to clinical and anatomical variables and proximity to the cutoff value than to the pressure wire used.
SUPPLEMENTARY DATA
Supplementary data associated with this article can be found in the online version available at https://doi.org/10.24875/ RECIC.M24000448.
ACKNOWLEDGEMENTS
We wish to thank M.ª Ángeles Carmona for her support in data collection and patient inclusion.
REFERENCES
1. Escaned J, Echavarría-Pinto M, Garcia-Garcia HM, et al. Prospective Assessment of the Diagnostic Accuracy of Instantaneous Wave-Free Ratio to Assess Coronary Stenosis Relevance:Results of ADVISE II International, Multicenter Study (ADenosine Vasodilator Independent Stenosis Evaluation II). JACC Cardiovasc Interv. 2015;8:824-833.
2. Davies JE, Sen S, Dehbi HM, et al. Use of the Instantaneous Wave-free Ratio or Fractional Flow Reserve in PCI. N Engl J Med. 2017;376:1824-1834.
3. Götberg M, Christiansen EH, Gudmundsdottir IJ, et al. Instantaneous Wave-free Ratio versus Fractional Flow Reserve to Guide PCI. N Engl J Med. 2017;376:1813-1823.
4. Neumann FJ, Sousa-Uva M, Ahlsson A, et al. 2018 ESC/EACTS Guidelines on myocardial revascularization. Eur Heart J. 2019;40:87-165.
5. Jeremias A, Maehara A, Généreux P, et al. Multicenter Core Laboratory Comparison of the Instantaneous Wave-Free Ratio and Resting Pd/Pa With Fractional Flow Reserve. The RESOLVE Study. J Am Coll Cardiol. 2014;63:1253-1261.
6. Cook CM, Jeremias A, Petraco R, et al. Fractional Flow Reserve/Instantaneous Wave-Free Ratio Discordance in Angiographically Intermediate Coronary Stenoses:An Analysis Using Doppler-Derived Coronary Flow Measurements. JACC Cardiovasc Interv. 2017;10:2514-2524.
7. Kobayashi Y, Johnson NP, Berry C, et al. The Influence of Lesion Location on the Diagnostic Accuracy of Adenosine-Free Coronary Pressure Wire Measurements. JACC Cardiovasc Interv. 2016;9:2390-2399.
8. Van't Veer M, Pijls N, Hennigan B, et al. Comparison of Different Diastolic Resting Indexes to iFR. Are They All Equal?J Am Coll Cardiol. 2017;70:3088-3096.
9. Haddad K, Potter B, Matteau A, et al. Assessing the Accuracy of a Second-Generation Optical Sensor Pressure Wire in a Wire-to-Wire Comparison (The ACCURACY Study). Cardiovasc Revasc Med. 2022;35:51-56.
10. Kern M. Comparing FFR Tools:New Wires and a Pressure Microcatheter. Cath Lab Digest. 2016;24(5).
11. Petraco R, Sen S, Echavarria-Pinto M, et al. Fractional Flow Reserve-Guided Revascularization. Practical Implications of a Diagnostic Gray Zone and Measurements Variability on Clinical Decisions. JACC Cardiovasc Interv. 2013;6:222-225.
12. Lee JM, Shin E-S, Nam C-W, et al. Discrepancy Between Fractional Flow Reserve and Instantaneous Wave-free Ratio:Clinical and Angiographic Characteristics. Int J Cardiol. 2017;245:63-68.
13. Warisawa T, Cook C, Howard JP, et al. Physiological Pattern of Disease Assessed by Pressure-Wire Pullback Has an Influence on Fractional Flow Reserve/Instantaneous Wave-Free Ratio Discordance. Insights From the Multicenter AJIP Registry. Circ Cardiovasc Interv. 2019;12:e007494.
14. Lim HS, Pim AL, De Bruyne B, et al. The impact of age on fractional flow reserve-guided percutaneous coronary intervention:A FAME (Fractional Flow Reserve versus Angiography for Multivessel Evaluation) trial substudy. Int J Cardiol. 2014;177:66-70.
15. Dérimay F, Johnson NP, Zimmermann FM, et al. Predictive factors of discordance between the instantaneous wave-free ratio and fractional flow reserve. Catheter Cardiovasc Interv. 2019;94:356-363.
16. Wienemann H, Meyer A, Mauri V, et al. Comparison of Resting Full-Cycle Ratio and Fractional Flow Reserve in a German Real-World Cohort. Front Cardiovasc Med. 2021;8:744181.
17. Schäfer S, Kelm, Mingers S, Strauer BE. Left ventricular remodeling impairs coronary flow reserve in hypertensive patients. J Hypertens. 2002;20:1431-1437.
18. Lopez-Palop R, Carrillo P, Frutos A, et al. Comparison of effectiveness of high-dose intracoronary adenosine versus intravenous administration on the assessment of fractional flow reserve in patients with coronary heart disease. Am J Cardiol. 2013;111:1277-1283.
19. Abo-Aly M, Lolay G, Adams C, et al. Comparison of Intracoronary versus Intravenous Adenosine-induced Maximal Hyperemia for Fractional Flow Reserve Measurement:A Systematic Review and Meta-analysis. Catheter Cardiovasc Interv. 2019;94:714-721.
20. Nijjer SS, De Waard GA, Sen S, et al. Coronary pressure and flow relationships in humans:phasic analysis of normal and pathological vessels and the implications for stenosis assessment:a report from the Iberian–Dutch–English (IDEAL) collaborators. Eur Heart J. 2016;37:2069-2080.
21. Vranckx P, Cutlip DE, McFadden EP, Kern MJ, Mehran R, Muller O. Coronary Pressure–Derived Fractional Flow Reserve Measurements. Recommendations for Standardization, Recording, and Reporting as a Core Laboratory Technique. Proposals for Integration in Clinical Trials. Circ Cardiovasc Interv. 2012;5:312-317.
22. Wakasa N, Kuromochi T, Mihashi N, et al. Impact of Pressure Signal Drift on Fractional Flow Reserve-Based Decision-Making for Patients With Intermediate Coronary Artery Stenosis. Circ J. 2016;80:1812-1819.
23. Cook C, Ahmad Y, Shun-Shin MJ, et al. Quantification of the Effect of Pressure Wire Drift on the Diagnostic Performance of Fractional Flow Reserve, Instantaneous Wave-Free Ratio, and Whole-Cycle Pd/Pa. Circ Cardiovasc Interv. 2016;9:e002988.
24. Matsumura M, Johnson N, Fearon WF, et al. Accuracy of Fractional Flow Reserve Measurements in Clinical Practice:Observations From a Core Laboratory Analysis. JACC Cardiovasc Interv. 2017;10:1392-1401.
ABSTRACT
Introduction and objectives: Although drug-eluting stents are the main treatment in percutaneous coronary interventions (PCI), drug-coated balloons (DCB) represent an appealing alternative as they eliminate the risk of stent thrombosis and avoid leaving any metal structure in the vessel wall. However, limited evidence has been published to date on the vessel wall healing processes, plaque remodeling, plaque composition, and the impact on the coronary microcirculation after percutaneous coronary intervention with DCB (DCB-PCI).
Methods: This is investigator-initiated, single-center, single-arm, open-label, pilot study of 30 patients with native vessel disease undergoing DCB-PCI. Intravascular ultrasound and angiography-derived index of microvascular resistance (IMRangio) will be performed before and immediately after PCI, and at 3 months of follow-up.
Conclusions: The study aims to provide new evidence on the modification of atherosclerotic plaque in patients with de novo lesions undergoing PCI with DCB. This will be assessed by examining the change in the percentage of atheroma volume and late lumen enlargement using intravascular ultrasound and by evaluating changes in the microcirculation using IMRangio.
Registered at Clinicaltrials.gov (NCT06080919).
Keywords: Drug-coated balloon. Intravascular ultrasound. Angiography-derived index of microvascular resistance.
RESUMEN
Introducción y objetivos: Pese a que los stents farmacoactivos son el tratamiento principal en las angioplastias coronarias, los balones farmacoactivos representan una alternativa interesante dado que eliminan el riesgo de trombosis del stent sin dejar ningún tipo de estructura metálica en la pared del vaso. No obstante, la evidencia en cuanto a los procesos de cicatrización de la pared del vaso, el remodelado, los cambios en la composición de la placa ateroesclerótica y el impacto en la microcirculación coronaria tras el intervencionismo coronario percutáneo (ICP) con balón farmacoactivo aún no se ha esclarecido.
Métodos: Estudio piloto abierto, de un solo grupo, iniciado por el investigador, de 30 pacientes con enfermedad de vaso nativo sometidos a ICP con balón farmacoactivo. Se realizará ecografía intravascular y se determinará el índice de resistencia microvascular derivado de la angiografía (angio-IRM) antes, inmediatamente después y a los 3 meses de seguimiento de la angioplastia.
Conclusiones: Se aportará nueva evidencia sobre la modificación de la placa en pacientes con enfermedad de vaso nativo tratados con balón farmacoactivo, evaluando el cambio en el porcentaje del volumen de ateroma y el aumento luminal tardío, así como los cambios en la microcirculación mediante angio-IRM.
Registrado en Clinicaltrials.gov (NCT06080919).
Palabras clave: Balón farmacoactivo. Ecografía intravascular. Índice de resistencia microvascular derivado de la angiografía.
Abbreviations DCB: drug-coated balloon. EEM: external elastic membrane. IMRangio: angiography-derived index of microcirculatory resistance. IVUS: intravascular ultrasound. PCI: Percutaneous coronary intervention.
INTRODUCTION
Coronary artery disease is the leading single cause of mortality worldwide, accounting for more than 7 million deaths annually1 and its prevalence has been increasing in the last 20 years.2 Percutaneous coronary intervention (PCI) has been crucial in the treatment of coronary artery disease.3,4 The advent of drug-eluting stents (DES) has substantially reduced restenosis rates through the deposition of antiproliferative drugs in the vessel wall. DES have evolved over the years and have become the gold standard in PCI.5 Drug-coated balloons (DCB) represent an alternative in the setting of PCI. DCB consist of a balloon coated with antiproliferative agents encapsulated in a polymer matrix.6 Upon inflation, the balloon brings the antiproliferative drug into contact with the vessel wall. The main goal of DCB is to eliminate the risk of stent thrombosis and achieve lower restenosis rates by not leaving any type of metal structure in the treated segment.6
The safety and efficacy of DCB have been extensively studied in de novo coronary artery disease.6 In small vessel disease, DCB have demonstrated noninferiority to DES in several randomized clinical trials.7 A recent meta-analysis has shown that the use of DCB, compared with that of DES, is associated with a lower risk of vessel thrombosis and a trend toward a lower risk of acute myocardial infarction.8 In large vessel de novo lesions, current data do not support the widespread use of DCB over DES, although DCB appear to be safe and effective.9,10 Nevertheless, there is a need to elucidate the elution on the vessel wall, healing processes, plaque remodeling, plaque composition and the impact on the coronary microcirculation following PCI with DCB.
The present report describes the design and rationale for a study of plaque modification and impact on the microcirculation after PCI with DCB (the PLAMI study).
METHODS
The study will be an investigator-initiated, single-center, single-arm, open-label, pilot study in patients undergoing PCI with DCB for de novo lesions. The study has been approved by the hospital ethics committee on research involving medical products. The study has been registered in ClinicalTrials.gov (NCT06080919).
Procedure
Eligible patients will be informed about the study and will be required to provide signed informed consent prior to inclusion. Patients will undergo DCB-PCI under intravascular ultrasound (IVUS) guidance. Angiography-derived coronary physiology will be assessed after the procedure using Angio Plus software (Pulse Medical Imaging Technology, China). The angiography images will be used to obtain the angiography-derived index of microcirculatory resistance (IMRangio) values, before and after DCB-PCI. All procedures will be performed according to current European guidelines5: the target lesion will be predilated with semicompliant balloons or noncompliant balloons, with a diameter equal to the reference vessel diameter and with an appropriate length. Multiple predilations will be accepted. The DCB will be the paclitaxel-coated balloon Pantera Lux (BIOTRONIK AG, Switzerland).
The lesion will then be treated with a DCB with a reference vessel diameter/balloon diameter ratio of 1:1. DCB length will be equal to lesion length + 5 mm. DCB inflation time will be set at 45 to 60 seconds to guarantee correct and complete drug elution. The prespecified reasons for DES implantation after DCB-PCI will be residual stenosis > 30%, dissections > type B and TIMI flow < 3.6 Angiographic follow-up with IVUS and IMRangio evaluation will be performed 3 months after the index procedure. The study timeline is summarized in figure 1.
IVUS images will be taken before the DCB-PCI, immediately after, and at 3 months of follow-up using the Opticross HD 60 MHz (Boston Scientific Corp, United States) system. All IVUS studies will be performed after intracoronary administration of 200 μg of nitroglycerin. The IVUS images will be acquired at 30 frames per second with an automatic transducer pull back (at 0.5 mm/second) to the proximal reference vessel lesion. As there will be no stents to take as a reference, the proximal and distal side branches adjacent to the treated lesion will serve as references, matching the coronary angiographic images (figure 2). All IVUS images will be analyzed by an independent core lab.
Angiography-derived assessment of coronary physiology will be performed with Angio Plus software (Pulse Medical Imaging Technology, China). For the evaluation of each lesion, at least 2 projections with a difference of > 25° will be selected. The operator will manually mark the points proximal and distal to the lesion, and the system automatically outlines the contours of the detected vessel. If the traced vessel trajectory deviates from the normal lumen, the necessary manual modifications will be performed. The artificial intelligence-assisted software combines the intravascular imaging information with the estimated vessel flow to obtain the IMRangio. All the angiography images will be analyzed by an independent core lab to obtain the IMRangio.
Study population and enrolment criteria
Patients will be screened to ensure they meet the inclusion criteria and none of the exclusion criteria prior to study enrolment. Inclusion criteria consist of an indication to undergo PCI for a de novo lesion according to current guidelines (with no restrictions regarding vessel size).5 Inclusion and exclusion criteria are summarized in table 1.
Inclusion criteria | Exclusion criteria |
---|---|
Patient with CAD undergoing PCI with DCB with no limitation to vessel size | Age < 18 years |
Cardiogenic shock | |
ST-segment elevation myocardial infarction | |
Use of mechanical circulatory support | |
Complex coronary lesions* including chronic total occlusions, bifurcation lesions, left main coronary artery disease, severe calcified lesions, graft interventions and in-stent restenosis | |
Inability to provide informed consent | |
Unable to understand and follow study-related instructions or unable to comply with study protocol | |
Currently participating in another trial | |
Pregnant women | |
CAD, coronary artery disease; DCB, drug-coated balloon; PCI, percutaneous coronary intervention. |
Sample size
Because of the exploratory nature of this study, no formal sample size calculation is required. Based on previous pilot studies with similar designs,12 a sample of 30 lesions is planned to evaluate the impact of DCB on coronary healing and the microcirculatory territory.
Study endpoints
The primary endpoint is the change in percentage atheroma volume evaluated by IVUS from baseline to 3 months of follow-up. Secondary endpoints will include a) lumen change from baseline to 3 months of follow-up (minimum, maximum, average areas), b) the percentage of progressors and percentage of regressors, c) external elastic membrane (EEM) change from baseline to post DCB-PCI (average), d) EEM change post-DCB-PCI to the 3-month follow-up (average), e) the percentage of remodeling types (neutral, negative, and positive), f) IMRangio change from baseline to post- DCB-PCI, g) IMRangio change from post-DCB-PCI to the 3-month follow-up.
An independent clinical event committee, consisting of cardiologists not participating in the trial, will review and adjudicate all major adverse cardiac events according to the study protocol.
Considering the luminal area as the area delimited by the luminal border, the minimal luminal area is defined as the smallest lumen area within the length of the treated lesion.13,14 The atheroma or plaque burden is defined as the ratio of atheroma area to the vessel EEM and is calculated by dividing the sum of plaque and media cross-sectional area (CSA) by the EEM CSA.13,14 As the atheroma area can be calculated in each frame, the total atheroma volume is obtained by taking the sum of the differences between the EEM CSA area and the luminal CSA for all available images.15 The percent of the volume of the EEM occupied by atheroma is called the percentage atheroma volume.15,16
Serial arterial remodeling types will be classified as usual: neutral if there is no change in EEM, negative if there is a decrease in the EEM and positive if vice versa.
Statistical considerations
Continuous variables will be described as mean ± standard deviation or median [interquartile range]. Categorical variables will be described as percentages. The paired t-test will be used to compare continuous variables measured before and after treatment in the same patient, and differences in proportions will be tested with the chi-square or Fisher exact test. A P value less than .05 (typically ≤ .05) will be considered statistically significant. Statistical analyses will be performed using Stata software version 13.1 (StataCorp LP, United States).
DISCUSSION
Although the use of DES remains predominant in the performance of PCI, complications such as stent thrombosis and in-stent restenosis led to the development of DCB. DCB have the theoretical benefit of not leaving metallic material in the vascular lumen, thereby reducing the possibility of mechanical complications such as malapposition, stent fracture, and stent thrombosis. This could potentially reduce neointimal proliferation and shorten the duration of dual antiplatelet therapy.6 Current guidelines assign a level IA recommendation to the treatment of in-stent restenosis.5 While the use of DCB in de novo lesions seems promising, it is not yet widespread. In addition, PCI is not without risks, as it involves a certain degree of injury to the artery wall from balloon inflations and stent struts.17,19 The vascular response to endothelial cell and smooth muscle cell injury represents a complex network of biochemical responses that involve the immune system. All these factors regulate the processes of neointimal hyperplasia, vascular remodeling, and normal reendothelialization of the arterial wall.17
The pathophysiology of restenosis and lumen loss after angioplasty is a complex process involving various factors and is not limited to neointimal hyperplasia.19 Acutely, plain old balloon angioplasty (POBA) generates an increase in luminal area that is mainly due to an expansion of the EEM, mainly attributed to the elastic properties of the vessel rather than to plaque compression or removal.20 Subsequently, within the first few minutes after PCI, there is an “acute recoil” due to the elastic properties of the arterial wall. In the chronic phase, IVUS data indicate that luminal loss is mainly due to a progressive reduction in EEM rather than an increase in atherosclerotic plaque volume. Unlike the acute phase where loss of area is solely due to elastic properties, “chronic recoil” leading to the loss of area also involves a combination factors such as fibrosis, apoptosis, and changes in the extracellular matrix.19,21 Interestingly, not all patients show negative remodeling with a decrease in EEM; around 25% show a persistent increase in EEM, which is correlated with a reduced restenosis rate. Consequently, restenosis appears to be primarily due to the direction and magnitude of changes in arterial remodeling,19 although neointimal hyperplasia also plays a role .
Nevertheless, the existing evidence is based on analysis after the use of traditional balloons. With DCB-PCI, late lumen enlargement has been observed compared with POBA.20 Although this finding has been partly attributed to the inhibition of neointimal proliferation by antiproliferative drugs,22 the role of plaque modification or vessel healing phenomena in influencing this process cannot be excluded. A previous study showed that late lumen enlargement was higher in areas with the highest plaque burden; however, that study was a retrospective assessment and used a quantitative coronary angiography protocol.23 It could be hypothesized that, by inducing controlled damage to the artery wall, together with the antiproliferative effect of DCB, positive vessel remodeling might be achieved, reducing restenosis rates without the need for DES. Therefore, with DCB-PCI, we are able to treat coronary stenosis not only from a mechanical point of view, but can also change the natural history of the disease and restenosis. In this regard, IVUS analysis will be essential to evaluate the reasons behind the gain or loss of luminal area. The dynamic changes produced after PCI are depicted in figure 3.
The DCB that will be used in our study, paclitaxel, has been extensively analyzed as a balloon-coating drug due to its lipophilic properties and its ability to elute into the vessel wall.24 Moreover, the available paclitaxel-DCB have shown good results in patients undergoing PCI for native vessel disease.25 In contrast, because of the hydrophobic characteristics of sirolimus, maintaining an adequate percentage in the wall over the mid-term poses technical challenges. However, advances in the formulation of the new generation of sirolimus DCB are anticipated to address this issue by facilitating adequate drug release into the vessel wall.24
As previously mentioned, the coronary microcirculation is closely related to proper coronary functioning and the pathophysiology of coronary artery disease. While it is believed that the performance of PCI, as well as the injury and healing of the coronary artery, may affect the coronary microcirculation, the evidence regarding DCB-PCI is scarce. Moreover, the plaque rupture, intimal dissections and thrombus formation that occur during balloon angioplasty are a potential source of embolism to the microvascular bed.
Since direct visualization of the microcirculation is not feasible in clinical practice,26 its assessment relies on parameters reflecting its functional status, usually coronary flow reserve and the IMR. Coronary flow reserve is defined as the ratio between hyperemic flow in response to nonendothelial vasodilation and resting blood flow. It is crucial to exclude epicardial stenosis before using coronary flow reserve, as it provides an integrated measurement of both epicardial and coronary microcirculation.26 IMR is calculated as the product of distal coronary pressure at maximal hyperemia multiplied by the hyperemic mean transit time.
In our study, we will perform a noninvasive, nonhyperemic assessment of the coronary microcirculation using IMRangio. This approach aims to characterize the baseline status of the microcirculation and assess the microvasculature changes induced by PCI and their variation over a 3-month period.
By monitoring IMRangio before and after treating the stenotic epicardial lesion, we will be able to assess the effects of acute fracture of the atherosclerotic plaque and injury to the arterial wall in the microvascular bed. We also aim to investigate whether these collateral harmful changes provoked during angioplasty remain consistent or vary significantly at 3 months of follow-up. In this same context, the analysis of IVUS during follow-up will allow us to correlate the changes in the arterial wall and atherosclerotic plaque after DCB-PCI with microcirculation physiology. To date, no insights into the anatomical and physiological process of healing of the injured arterial wall after DCB-PCI have been available in the published literature.
CONCLUSIONS
The PLAMI study is a first-in-man pilot study that aims to provide new information on the modification of atherosclerotic plaque assessed by intracoronary imaging in patients with de novo lesions undergoing PCI with DCB.
FUNDING
None reported.
ETHICAL CONSIDERATIONS
The study has been approved by the hospital ethics committee on research involving medical products. Eligible patients will be informed about the study and must provide written informed consent prior to inclusion in the study. Possible gender/sex biases have been considered.
STATEMENT ON THE USE OF ARTIFICIAL INTELLIGENCE
No artificial intelligence has been used in the preparation of this article.
AUTHORS’ CONTRIBUTIONS
J.A Sorolla Romero, A.Teira Calderón, J. Sanz Sánchez and H.M. Garcia-Garcia contributed to the conception, design, drafting and revision of the article. J.P. Vílchez Tschischke, P. Aguar Carrascosa, F.Ten Morro, L. Andrés Lalaguna, L. Martínez Dolz and J.L. Díez Gil contributed to the critical revision of the intellectual content.
CONFLICTS OF INTEREST
None declared.
WHAT IS KNOWN ABOUT THE TOPIC?
- DCB have proven clinical effectiveness in cases of in-stent restenosis and de novo lesions involving small vessel coronary artery disease.
- Several studies in small vessel coronary artery disease have shown a benefit of DCB in the vessel wall, with late lumen enlargement during follow-up.
- However, there is little evidence of their use in larger vessels.
- In addition, the impact of DCB on the coronary microcirculation has not been evaluated to date.
WHAT DOES THIS STUDY ADD?
- The PLAMI study aims to characterize vessel healing using IVUS after DCB-PCI in patients with native vessel disease and to correlate these findings with the impact on microcirculation.
REFERENCES
1. Ralapanawa U, Sivakanesan R. Epidemiology and the Magnitude of Coronary Artery Disease and Acute Coronary Syndrome:A Narrative Review. J Epidemiol Glob Health. 2021;11:169-177.
2. Arnett DK, Blumenthal RS, Albert MA, et al. 2019 ACC/AHA Guideline on the Primary Prevention of Cardiovascular Disease:A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation. 2019;140:e596-e646.
3. Canfield J, Totary-Jain H. 40 Years of Percutaneous Coronary Intervention:History and Future Directions. J Pers Med. 2018;8:33.
4. Stefanini GG, Alfonso F, Barbato E, et al. Management of myocardial revascularisation failure:an expert consensus document of the EAPCI. EuroIntervention. 2020;16:e875-e890.
5. Neumann F, Sousa-Uva M, Ahlsson A, et al. 2018 ESC/EACTS Guidelines on myocardial revascularization. Eur Heart J. 2019;40:87-165.
6. Jeger RV, Eccleshall S, Wan Ahmad WA, et al. Drug-Coated Balloons for Coronary Artery Disease:Third Report of the International DCB Consensus Group. JACC Cardiovasc Interv. 2020;13:1391-1402.
7. Tang Y, Qiao S, Su X, et al. Drug-Coated Balloon Versus Drug-Eluting Stent for Small-Vessel Disease:The RESTORE SVD China Randomized Trial. JACC Cardiovasc Interv. 2018;11:2381-2392.
8. Sanz Sánchez J, Chiarito M, Cortese B, et al. Drug-Coated balloons vs drug-eluting stents for the treatment of small coronary artery disease:A meta-analysis of randomized trials. Catheter Cardiovasc Interv. 2021;98:66-75.
9. Yerasi C, Case BC, Forrestal BJ, et al. Drug-Coated Balloon for de Novo Coronary Artery Disease:JACC State-of-the-Art Review. J Am Coll Cardiol. 2020;75:1061-1073.
10. Nishiyama N, Komatsu T, Kuroyanagi T, et al. Clinical value of drug-coated balloon angioplasty for de novo lesions in patients with coronary artery disease. Int J Cardiol. 2016;222:113-118.
11. Lawton JS, Tamis-Holland JE, Bangalore S, et al. 2021 ACC/AHA/SCAI Guideline for Coronary Artery Revascularization. J Am Coll Cardiol. 2022;79:e21-e129.
12. Joner M, Finn AV, Farb A, et al. Pathology of drug-eluting stents in humans:delayed healing and late thrombotic risk. J Am Coll Cardiol. 2006;48:193-202.
13. Xu J, Lo S. Fundamentals and role of intravascular ultrasound in percutaneous coronary intervention. Cardiovasc Diagn Ther. 2020;10:1358-1370.
14. Mintz GS, Nissen SE, Anderson WD, et al. American College of Cardiology Clinical Expert Consensus Document on Standards for Acquisition, Measurement and Reporting of Intravascular Ultrasound Studies (IVUS). A report of the American College of Cardiology Task Force on Clinical Expert Consensus Documents. J Am Coll Cardiol. 2001;37:1478-1492.
15. Gogas BD, Farooq V, Serruys PW, Garcia-Garcia HM. Assessment of coronary atherosclerosis by IVUS and IVUS-based imaging modalities:progression and regression studies, tissue composition and beyond. Int J Cardiovasc Imaging. 2011;27:225-237.
16. Tobis JM, Perlowski A. Atheroma Volume by Intravascular Ultrasound as a Surrogate for Clinical End Points. J Am Coll Cardiol. 2009;53:1116-1118.
17. Feinberg MW. Healing the injured vessel wall using microRNA-facilitated gene delivery. J Clin Invest. 2014;124:3694-3697.
18. Inoue T, Croce K, Morooka T, Sakuma M, Node K, Simon DI. Vascular Inflammation and Repair:Implications for Reendothelialization, Restenosis, and Stent Thrombosis. JACC Cardiovasc Interv. 2011;4:1057-1066.
19. Mintz GS, Popma JJ, Pichard AD, et al. Arterial Remodeling After Coronary Angioplasty. Circulation. 1996;94:35-43.
20. Her AY, Ann SH, Singh GB, et al. Comparison of Paclitaxel-Coated Balloon Treatment and Plain Old Balloon Angioplasty for De Novo Coronary Lesions. Yonsei Med J. 2016;57:337-341.
21. Geary RL, Nikkari ST, Wagner WD, Williams JK, Adams MR, Dean RH. Wound healing:A paradigm for lumen narrowing after arterial reconstruction. J Vasc Surg. 1998;27:96-108.
22. Sogabe K, Koide M, Fukui K, et al. Optical coherence tomography analysis of late lumen enlargement after paclitaxel-coated balloon angioplasty for de-novo coronary artery disease. Catheter Cardiovasc Interv. 2021;98:E35-E42.
23. Kleber FX, Schulz A, Waliszewski M, et al. Local paclitaxel induces late lumen enlargement in coronary arteries after balloon angioplasty. Clin Res Cardiol. 2015;104:217-225.
24. Yerasi C, Case BC, Forrestal BJ, et al. Drug-Coated Balloon for De Novo Coronary Artery Disease:JACC State-of-the-Art Review. J Am Coll Cardiol. 2020;75:1061-1073.
25. Venetsanos D, Omerovic E, Sarno G, et al. Long term outcome after treatment of de novo coronary artery lesions using three different drug coated balloons. Int J Cardiol. 2021;325:30-36.
26. Kunadian V, Chieffo A, Camici PG, et al. An EAPCI Expert Consensus Document on Ischaemia with Non-Obstructive Coronary Arteries in Collaboration with European Society of Cardiology Working Group on Coronary Pathophysiology &Microcirculation Endorsed by Coronary Vasomotor Disorders International Study Group. Eur Heart J. 2020;41:3504-3520.
* Corresponding authors.
E-mail addresses: sjorge4@gmx.com (J. Sanz Sánchez); hector.m.garciagarcia@medstar.net; hect2701@gmail.com (H.M. Garcia-Garcia).
- On- vs off-hours primary percutaneous coronary intervention: a single-center 5-year experience
- Angina or ischemia with no obstructed coronary arteries: a specific diagnostic and therapeutic protocol
- Excimer laser coronary atherectomy in severely calcified lesions: time to bust the myth
- Effectiveness of the DyeVert Power XT system during percutaneous coronary interventions
Debate
Debate: ECMO in patients with cardiogenic shock due to myocardial infarction
A researcher’s perspective
aHeart Center Leipzig at Leipzig University, Department of Internal Medicine/Cardiology, Leipzig, Germany
bLeipzig Heart Science, Leipzig, Germany
A clinician’s perspective
Unidad de Cuidados Agudos Cardiológicos, Servicio de Cardiología, Hospital Universitario la Paz, IdiPAZ, Madrid, Spain
Editorials
Clinical evaluation requirements under the new European Union medical device regulation
Centro Nacional de Certificación de Productos Sanitarios, Agencia Española de Medicamentos y Productos Sanitarios (AEMPS), Madrid, Spain
Review Articles
Scientific letters
Practical concepts of catheter-directed aspiration thrombectomy in ECMO-supported patients
aServicio de Cardiología, Hospital Universitario La Paz, Madrid, Spain
bServicio de Cardiología, Hospital Universitario 12 de Octubre, Madrid, Spain