1
|
Russu E, Arbanasi EM, Chirila TV, Muresan AV. Therapeutic strategies based on non-ionizing radiation to prevent venous neointimal hyperplasia: the relevance for stenosed arteriovenous fistula, and the role of vascular compliance. Front Cardiovasc Med 2024; 11:1356671. [PMID: 38374996 PMCID: PMC10875031 DOI: 10.3389/fcvm.2024.1356671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 01/23/2024] [Indexed: 02/21/2024] Open
Abstract
We have reviewed the development and current status of therapies based on exposure to non-ionizing radiation (with a photon energy less than 10 eV) aimed at suppressing the venous neointimal hyperplasia, and consequentially at avoiding stenosis in arteriovenous grafts. Due to the drawbacks associated with the medical use of ionizing radiation, prominently the radiation-induced cardiovascular disease, the availability of procedures using non-ionizing radiation is becoming a noteworthy objective for the current research. Further, the focus of the review was the use of such procedures for improving the vascular access function and assuring the clinical success of arteriovenous fistulae in hemodialysis patients. Following a brief discussion of the physical principles underlying radiotherapy, the current methods based on non-ionizing radiation, either in use or under development, were described in detail. There are currently five such techniques, including photodynamic therapy (PDT), far-infrared therapy, photochemical tissue passivation (PTP), Alucent vascular scaffolding, and adventitial photocrosslinking. The last three are contingent on the mechanical stiffening achievable by the exogenous photochemical crosslinking of tissular collagen, a process that leads to the decrease of venous compliance. As there are conflicting opinions on the role of compliance mismatch between arterial and venous conduits in a graft, this aspect was also considered in our review.
Collapse
Affiliation(s)
- Eliza Russu
- Clinic of Vascular Surgery, Mures County Emergency Hospital, Targu Mures, Romania
- Department of Vascular Surgery, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Targu Mures, Romania
| | - Emil-Marian Arbanasi
- Clinic of Vascular Surgery, Mures County Emergency Hospital, Targu Mures, Romania
- Department of Vascular Surgery, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Targu Mures, Romania
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Targu Mures, Romania
- Centre for Advanced Medical and Pharmaceutical Research (CCAMF), George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Targu Mures, Romania
| | - Traian V. Chirila
- Centre for Advanced Medical and Pharmaceutical Research (CCAMF), George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Targu Mures, Romania
- Queensland Eye Institute, Woolloongabba, QLD, Australia
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Targu Mures, Romania
- School of Chemistry and Physics, Queensland University of Technology, Brisbane, QLD, Australia
- Australian Institute of Bioengineering and Nanotechnology (AIBN), University of Queensland, St Lucia, QLD, Australia
| | - Adrian V. Muresan
- Clinic of Vascular Surgery, Mures County Emergency Hospital, Targu Mures, Romania
- Department of Vascular Surgery, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Targu Mures, Romania
| |
Collapse
|
2
|
Lauder L, da Costa BR, Ewen S, Scholz SS, Wijns W, Lüscher TF, Serruys PW, Edelman ER, Capodanno D, Böhm M, Jüni P, Mahfoud F. Randomized trials of invasive cardiovascular interventions that include a placebo control: a systematic review and meta-analysis. Eur Heart J 2021; 41:2556-2569. [PMID: 32666097 DOI: 10.1093/eurheartj/ehaa495] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 04/01/2020] [Accepted: 05/27/2020] [Indexed: 01/09/2023] Open
Abstract
AIMS The difference in the benefit of invasive cardiovascular interventions compared with placebo controls has not been analysed systematically. METHODS AND RESULTS MEDLINE and Web of Science were searched through 29 March 2020. Randomized, placebo-controlled trials of invasive cardiovascular interventions (including catheter-based interventions and pacemaker-like devices) investigating predefined primary outcomes were included. Standardized mean differences (SMD) and odds ratios were calculated for continuous and dichotomous outcomes, respectively. Meta-regression analyses were performed to assess whether estimates of treatment effects were associated with methodological characteristics of trials. Thirty trials, including 4102 patients, were analysed. The overall risk of bias was judged to be low in only 43% of the trials. Ten trials (33%) demonstrated statistically significant superiority of invasive interventions over placebo controls for the respective predefined primary outcomes. In almost half of the 16 trials investigating continuous predefined primary outcomes, the SMD between the active and placebo procedure indicated a small (n = 4) to moderate (n = 3) treatment effect of active treatment over placebo. In contrast, one trial indicated a small treatment effect in favour of the placebo procedure. In the remaining trials, there was no relevant treatment effect of active treatment over placebo. In trials with a protocol-mandated stable and symmetrical use of co-interventions, the superiority of active procedures vs. invasive placebo procedures was significantly larger as compared with trials with frequent or unbalanced changes in co-interventions (P for interaction 0.027). CONCLUSIONS The additional treatment effect of invasive cardiovascular interventions compared with placebo controls was small in most trials.
Collapse
Affiliation(s)
- Lucas Lauder
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes, Saarland University, Kirrberger Str., Gebäude 41.1, 66421 Homburg/Saar, Germany
| | - Bruno R da Costa
- Applied Health Research Centre, Li Ka Shing Knowledge Institute of St Michael's Hospital, University of Toronto, 155 College Street, Toronto, ON M5T 3M6, Canada.,Institute of Primary Health Care (BIHAM), University of Bern, Mittelstraße 43, 3012 Bern, Switzerland
| | - Sebastian Ewen
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes, Saarland University, Kirrberger Str., Gebäude 41.1, 66421 Homburg/Saar, Germany
| | - Sean S Scholz
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes, Saarland University, Kirrberger Str., Gebäude 41.1, 66421 Homburg/Saar, Germany
| | - William Wijns
- The Lambe Institute for Translational Medicine and CURAM, National University of Ireland, University Road, Galway H91 TK33, Ireland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, Schlieren Campus, University of Zürich, Wagistrasse 12, 8952 Schlieren, Switzerland.,Royal Brompton and Harefield Hospital Trust, Imperial College London, Sydney Street, London SW3 6NP, UK
| | - Patrick W Serruys
- The National Lung and Heart Institute, Imperial College London, Dovehouse St, Chelsea, London SW3 6LY, UK
| | - Elazer R Edelman
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.,Institute for Medical Engineering and Science, MIT, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Davide Capodanno
- Division of Cardiology, C.A.S.T., P.O. "G. Rodolico", Azienda Ospedaliero-Universitaria "Policlinico-Vittorio Emanuele", University of Catania, Via S. Citelli, 31 Catania, Italy
| | - Michael Böhm
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes, Saarland University, Kirrberger Str., Gebäude 41.1, 66421 Homburg/Saar, Germany
| | - Peter Jüni
- Applied Health Research Centre, Li Ka Shing Knowledge Institute of St Michael's Hospital, University of Toronto, 155 College Street, Toronto, ON M5T 3M6, Canada.,Department of Medicine, Institute of Health Policy, Management and Evaluation, University of Toronto, 155 College Street, Suite 425, Toronto, ON M5T 3M6, Canada
| | - Felix Mahfoud
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes, Saarland University, Kirrberger Str., Gebäude 41.1, 66421 Homburg/Saar, Germany.,Institute for Medical Engineering and Science, MIT, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| |
Collapse
|
3
|
Brahmbhatt AN, Misra S. Stem Cell Delivery for the Treatment of Arteriovenous Fistula Failure. STEM CELL THERAPY FOR VASCULAR DISEASES 2021:281-297. [DOI: 10.1007/978-3-030-56954-9_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|
4
|
A randomized feasibility study of the effect of ascorbic acid on post-angioplasty restenosis of hemodialysis vascular access (NCT03524846). Sci Rep 2019; 9:11095. [PMID: 31366980 PMCID: PMC6668477 DOI: 10.1038/s41598-019-47583-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 07/16/2019] [Indexed: 12/16/2022] Open
Abstract
Restenosis remains a significant problem after angioplasty of hemodialysis vascular access. Both experimental and clinical studies have shown a protective effect of antioxidants against post-angioplasty restenosis. A prospective, randomized, feasibility study was conducted to investigate the effect of ascorbic acid to prevent restenosis. Ninety-three hemodialysis patients were randomized into three groups after angioplasty: placebo (n = 31), 300 mg ascorbic acid (n = 31), and 600 mg ascorbic acid (n = 31), treated intravenously 3 times per week for 3 months. Eighty-nine completed the clinical follow-up, and 81 had angiographic follow-up. In the angiographic follow-up, the mean (stand deviation) late loss of luminal diameter for the placebo, 300 mg, and 600 mg groups were 3.15 (1.68) mm, 2.52 (1.70) mm (P = 0.39 vs. placebo group), and 1.59 (1.67) mm (P = 0.006, vs. placebo group), with corresponding angiographic binary restenosis of 79%, 67% (P = 0.38 vs. placebo group), and 54% (P = 0.08 vs. placebo group). The post-interventional primary patency rates at 3 months were 47%, 55% (P = 0.59 vs. placebo group), and 70% (P = 0.18 vs. placebo group) for placebo, 300 mg, and 600 mg groups. Our results demonstrated that intravenous 600 mg ascorbic acid was a feasible therapy and might attenuate restenosis after angioplasty; however, its effect on post-interventional primary patency was modest.
Collapse
|
5
|
Abstract
BACKGROUND To report our experience and evaluate the results of a hybrid procedure comprising of surgical thrombectomy and adjuvant high pressure balloon angioplasty for management of thrombosed arteriovenous grafts (AVGs) with underlying venous anastomotic stenosis. METHODS Between January 2015 and June 2017, 148 patients with first-time thrombosis of AVGs were considered for treatment with surgical thrombectomy followed by high pressure balloon angioplasty of the underlying venous anastomotic lesions. Monitoring and surveillance was subsequently performed using clinical and hemodialysis criteria to detect a failing/failed access. Primary, assisted primary, and secondary patency rates were calculated using Kaplan-Meier analysis. RESULTS This hybrid procedure was technically successful in 135 cases (91.2%) and clinically successful in 131 cases (88.5%). The postintervention primary patency rate of the target lesions was 68.2%, 52.4%, and 44.2% at 3, 6, and 12 months, respectively. Endovascular re-interventions increased significantly the postintervention assisted primary patency to 79.1%, 71.9%, and 66.8% (P=0.0004), and the postintervention secondary patency to 86.5%, 82.2%, and 78.6% (P<0.0001) at the same time points, respectively. CONCLUSIONS Hybrid salvage of thrombosed AVGs using high pressure balloon angioplasty of the venous anastomotic stenosis following surgical thrombectomy by Fogarty catheter is a highly successful and safe procedure, with acceptable short-term primary patency. The need for repeated interventions demonstrates the necessity of continuous graft monitoring and surveillance to improve both assisted primary, and secondary patency rates.
Collapse
Affiliation(s)
- Haitham Ali
- Department of Vascular and Endovascular Surgery, Assiut University Hospitals, Assiut, Egypt -
| | - Khaled Attallah
- Department of Vascular and Endovascular Surgery, Assiut University Hospitals, Assiut, Egypt
| | - Khaled Awad
- Department of Vascular and Endovascular Surgery, Assiut University Hospitals, Assiut, Egypt
| | - Bahgat Thabet
- Department of Vascular and Endovascular Surgery, Assiut University Hospitals, Assiut, Egypt
| |
Collapse
|
6
|
Radiation suppresses neointimal hyperplasia through affecting proliferation and apoptosis of vascular smooth muscle cells. J Vasc Access 2017; 19:153-161. [PMID: 29192723 DOI: 10.5301/jva.5000804] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To study the effect of x-ray radiotherapy on vascular smooth muscle cells (VSMCs) and elucidate the mechanisms in preventing neointimal hyperplasia of prosthetic vascular grafts. MATERIALS AND METHODS In model I, twelve mongrel dogs underwent revascularization with prosthetic grafts and half the dogs underwent irradiation of the grafts at 28 Gy. In model II, human VSMCs (hVSMCs) were maintained and divided into six groups to which external radiation was applied at six different doses: 0 Gy, 2 Gy, 8 Gy, 16 Gy, 24 Gy and 30 Gy. In both models, specimens were harvested and examined by using morphological, immunological, cellular and molecular methods. RESULTS After irradiation, the neointima thickness was significantly lower in irradiated groups (p≤0.01). The radiotherapy could up-regulate p27kip1, and down-regulate proliferating cell nuclear antigen (PCNA) and S phase kinase associated protein 2 (Skp2). X-ray irradiation inhibits the proliferation of hVSMCs via acting on G1/S phase of cell cycle. The apoptosis of hVSMCs increased significantly with dose and time. The expression of PCNA and Skp2 were decreased after a first increasing trend with dose, but had a significant negative correlation with time. The expression of p27kip1 had a significant positive correlation with dose and time. CONCLUSIONS Postoperative external fractionated irradiation after prosthetic vessel replacement of the abdominal aorta suppressed the development of hyperplasia in the graft neointima in the short term. There was a prominent time- and dose-dependent inhibition of VSMC proliferation by radiation when it was administered.
Collapse
|
7
|
Hu H, Patel S, Hanisch JJ, Santana JM, Hashimoto T, Bai H, Kudze T, Foster TR, Guo J, Yatsula B, Tsui J, Dardik A. Future research directions to improve fistula maturation and reduce access failure. Semin Vasc Surg 2016; 29:153-171. [PMID: 28779782 DOI: 10.1053/j.semvascsurg.2016.08.005] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
With the increasing prevalence of end-stage renal disease, there is a growing need for hemodialysis. Arteriovenous fistulae (AVF) are the preferred type of vascular access for hemodialysis, but maturation and failure continue to present significant barriers to successful fistula use. AVF maturation integrates outward remodeling with vessel wall thickening in response to drastic hemodynamic changes in the setting of uremia, systemic inflammation, oxidative stress, and pre-existent vascular pathology. AVF can fail due to both failure to mature adequately to support hemodialysis and development of neointimal hyperplasia that narrows the AVF lumen, typically near the fistula anastomosis. Failure due to neointimal hyperplasia involves vascular cell activation and migration and extracellular matrix remodeling with complex interactions of growth factors, adhesion molecules, inflammatory mediators, and chemokines, all of which result in maladaptive remodeling. Different strategies have been proposed to prevent and treat AVF failure based on current understanding of the modes and pathology of access failure; these approaches range from appropriate patient selection and use of alternative surgical strategies for fistula creation, to the use of novel interventional techniques or drugs to treat failing fistulae. Effective treatments to prevent or treat AVF failure require a multidisciplinary approach involving nephrologists, vascular surgeons, and interventional radiologists, careful patient selection, and the use of tailored systemic or localized interventions to improve patient-specific outcomes. This review provides contemporary information on the underlying mechanisms of AVF maturation and failure and discusses the broad spectrum of options that can be tailored for specific therapy.
Collapse
Affiliation(s)
- Haidi Hu
- Department of Surgery, Yale University School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Department of Vascular and Thyroid Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT
| | - Sandeep Patel
- Department of Surgery, Yale University School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT; Royal Free Hospital, University College London, London, UK
| | - Jesse J Hanisch
- Department of Surgery, Yale University School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT
| | - Jeans M Santana
- Department of Surgery, Yale University School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT
| | - Takuya Hashimoto
- Department of Surgery, Yale University School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT
| | - Hualong Bai
- Department of Surgery, Yale University School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT
| | - Tambudzai Kudze
- Department of Surgery, Yale University School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT
| | - Trenton R Foster
- Department of Surgery, Yale University School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT
| | - Jianming Guo
- Department of Surgery, Yale University School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT
| | - Bogdan Yatsula
- Department of Surgery, Yale University School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT
| | - Janice Tsui
- Royal Free Hospital, University College London, London, UK
| | - Alan Dardik
- Department of Surgery, Yale University School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT; VA Connecticut Healthcare System, West Haven, CT.
| |
Collapse
|
8
|
Ravani P, Quinn RR, Oliver MJ, Karsanji DJ, James MT, MacRae JM, Palmer SC, Strippoli GFM. Pre-emptive correction for haemodialysis arteriovenous access stenosis. Cochrane Database Syst Rev 2016; 2016:CD010709. [PMID: 26741512 PMCID: PMC6486172 DOI: 10.1002/14651858.cd010709.pub2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Guidelines recommend routine arteriovenous (AV) graft and fistula surveillance (technology-based screening) in addition to clinical monitoring (physical examination) for early identification and pre-emptive correction of a stenosis before the access becomes dysfunctional. However, consequences on patient-relevant outcomes of pre-emptive correction of a stenosis in a functioning access as opposed to deferred correction, i.e. correction postponed to when the access becomes dysfunctional, are uncertain. OBJECTIVES We aimed to evaluate 1) whether pre-emptive correction of an AV access stenosis improves clinically relevant outcomes; 2) whether the effects of pre-emptive correction of an AV access stenosis differ by access type (fistula versus graft), aim (primary and secondary prophylaxis), and surveillance method for primary prophylaxis (Doppler ultrasound for the screening of functional and anatomical changes versus measurement of the flow in the access); and 3) whether other factors (dialysis duration, access location, configuration or materials, algorithm for referral for intervention, intervention strategies (surgical versus radiological or other), or study design) explain the heterogeneity that might exist in the effect estimates. SEARCH METHODS We searched the Cochrane Kidney and Transplant Specialised Register to 30 November 2015 using search terms relevant to this review. SELECTION CRITERIA We included all studies of any access surveillance method for early identification and pre-emptive treatment of an AV access stenosis. DATA COLLECTION AND ANALYSIS We extracted data on potentially remediable and irremediable failure of the access (i.e. thrombosis and access loss respectively); infection and mortality; and resource use (hospitalisation, diagnostic and intervention procedures). Analysis was by a random effects model and results expressed as risk ratio (RR), hazard ratio (HR) or incidence rate ratio (IRR) with 95% confidence intervals (CI). MAIN RESULTS We identified 14 studies (1390 participants), nine enrolled adults without a known access stenosis (primary prophylaxis; three studies including people using fistulas) and five enrolled adults with a documented stenosis in a non-dysfunctional access (secondary prophylaxis; three studies in people using fistulas). Study follow-up ranged from 6 to 38 months, and study size ranged from 58 to 189 participants. In low- to moderate-quality evidence (based on GRADE criteria) in adults treated with haemodialysis, relative to no surveillance and deferred correction, surveillance with pre-emptive correction of an AV stenosis reduced the risk of thrombosis (RR 0.79, 95% CI 0.65 to 0.97; I² = 30%; 18 study comparisons, 1212 participants), but had imprecise effect on the risk of access loss (RR 0.81, 95% CI 0.65 to 1.02; I² = 0%; 11 study comparisons, 972 participants). In analyses subgrouped by access type, pre-emptive stenosis correction did not reduce the risk of thrombosis (RR 0.95, 95% CI 0.8 to 1.12; I² = 0%; 11 study comparisons, 697 participants) or access loss in grafts (RR 0.9, 95% CI 0.71 to 1.15; I² = 0%; 7 study comparisons; 662 participants), but did reduce the risk of thrombosis (RR 0.5, 95% CI 0.35 to 0.71; I² = 0%; 7 study comparisons, 515 participants) and the risk of access loss in fistulas (RR 0.5, 95% CI 0.29 to 0.86; I² = 0%; 4 studies; 310 participants). Three of the four studies reporting access loss data in fistulas (199 participants) were conducted in the same centre. Insufficient data were available to assess whether benefits vary by prophylaxis aim in fistulas (i.e. primary and secondary prophylaxis). Although the magnitude of the effects of pre-emptive stenosis correction was considerable for patient-centred outcomes, results were either heterogeneous or imprecise. While pre-emptive stenosis correction may reduce the rates of hospitalisation (IRR 0.54, 95% CI 0.31 to 0.93; I² = 67%; 4 study comparisons, 219 participants) and use of catheters (IRR 0.58, 95% CI 0.35 to 0.98; I² = 53%; 6 study comparisons, 394 participants), it may also increase the rates of diagnostic procedures (IRR 1.78, 95% CI 1.18 to 2.67; I² = 62%; 7 study comparisons, 539 participants), infection (IRR 1.74, 95% CI 0.78 to 3.91; I² = 0%; 3 studies, 248 participants) and mortality (RR 1.38, 95% CI 0.91 to 2.11; I² = 0%; 5 studies, 386 participants).In general, risk of bias was high or unclear in most studies for many domains we assessed. Four studies were published after 2005 and only one had evidence of registration within a trial registry. No study reported information on authorship and/or involvement of the study sponsor in data collection, analysis, and interpretation. AUTHORS' CONCLUSIONS Pre-emptive correction of a newly identified or known stenosis in a functional AV access does not improve access longevity. Although pre-emptive stenosis correction may be promising in fistulas existing evidence is insufficient to guide clinical practice and health policy. While pre-emptive stenosis correction may reduce the risk of hospitalisation, this benefit is uncertain whereas there may be a substantial increase (i.e. 80%) in the use of access-related procedures and procedure-related adverse events (e.g. infection, mortality). The net effects of pre-emptive correction on harms and resource use are thus unclear.
Collapse
Affiliation(s)
- Pietro Ravani
- Cumming School of Medicine, University of CalgaryDepartments of Medicine and Community Health SciencesFoothills Medical Centre1403 29th St NWCalgaryABCanadaT2N 2T9
| | - Robert R Quinn
- Cumming School of Medicine, University of CalgaryDepartments of Medicine and Community Health SciencesFoothills Medical Centre1403 29th St NWCalgaryABCanadaT2N 2T9
| | - Matthew J Oliver
- University of TorontoDepartment of MedicineSunnybrook Health Sciences Centre2075 Bayview Avenue ‐ Room A239TorontoONCanadaM4N 3M5
| | - Divya J Karsanji
- Cumming School of Medicine, University of CalgaryCommunity Health SciencesCalgaryABCanada
| | - Matthew T James
- Cumming School of Medicine, University of CalgaryDepartment of Medicine and Community Health SciencesFoothills Medical Centre1403 29th StCalgaryABCanadaT2N 2T9
| | - Jennifer M MacRae
- Cumming School of Medicine, University of CalgaryDepartment of MedicineFoothills Medical Centre1403 29th St NWCalgaryABCanadaT2N 2T9
| | - Suetonia C Palmer
- University of Otago ChristchurchDepartment of Medicine2 Riccarton AvePO Box 4345ChristchurchNew Zealand8140
| | - Giovanni FM Strippoli
- The Children's Hospital at WestmeadCochrane Kidney and Transplant, Centre for Kidney ResearchWestmeadNSWAustralia2145
- University of BariDepartment of Emergency and Organ TransplantationBariItaly
- DiaverumMedical Scientific OfficeLundSweden
- Diaverum AcademyBariItaly
- The University of SydneySydney School of Public HealthSydneyAustralia
| | | |
Collapse
|
9
|
Terry CM, Dember LM. Novel therapies for hemodialysis vascular access dysfunction: myth or reality? Clin J Am Soc Nephrol 2013; 8:2202-12. [PMID: 24235283 DOI: 10.2215/cjn.07360713] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hemodialysis vascular access dysfunction is a major source of morbidity for patients with ESRD. Development of effective approaches to prevent and treat vascular access failure requires an understanding of the underlying mechanisms, suitable models for preclinical testing, systems for targeted delivery of interventions to maximize efficacy and minimize toxicity, and rigorous clinical trials that use appropriate outcome measures. This article reviews the substantial progress and ongoing challenges in developing novel treatments for arteriovenous vascular access failure and focuses on localized rather than systemic interventions.
Collapse
Affiliation(s)
- Christi M Terry
- Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah, †Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | |
Collapse
|
10
|
Yang B, Janardhanan R, Vohra P, Greene EL, Bhattacharya S, Withers S, Roy B, Nieves Torres EC, Mandrekar J, Leof EB, Mukhopadhyay D, Misra S. Adventitial transduction of lentivirus-shRNA-VEGF-A in arteriovenous fistula reduces venous stenosis formation. Kidney Int 2013; 85:289-306. [PMID: 23924957 PMCID: PMC3844094 DOI: 10.1038/ki.2013.290] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 06/02/2013] [Accepted: 06/20/2013] [Indexed: 12/30/2022]
Abstract
Venous neointimal hyperplasia (VNH) causes hemodialysis vascular access failure. Here we tested whether VNH formation occurs in part due to local vessel hypoxia caused by surgical trauma to the vasa vasorum of the outflow vein at the time of arteriovenous fistula placement. Selective targeting of the adventitia of the outflow vein at the time of fistula creation was performed using a lentivirus-delivered small-hairpin RNA that inhibits VEGF-A expression. This resulted in significant increase in mean lumen vessel area, decreased media/adventitia area, and decreased constrictive remodeling with a significant increase in apoptosis (increase in caspase 3 activity and TUNEL staining) accompanied with decreased cellular proliferation and hypoxia-inducible factor-1α at the outflow vein. There was significant decrease in cells staining positive for α-smooth muscle actin (a myofibroblast marker) and VEGFR-1 expression with a decrease in MMP-2 and MMP-9. These results were confirmed in animals that were treated with humanized monoclonal antibody to VEGF-A with similar results. Since hypoxia can cause fibroblast to differentiate into myofibroblasts, we silenced VEGF-A gene expression in fibroblasts and subjected them to hypoxia. This decreased myofibroblast production, cellular proliferation, cell invasion, MMP-2 activity, and increased caspase 3. Thus, VEGF-A reduction at the time of arteriovenous fistula placement results in increased positive vascular remodeling.
Collapse
Affiliation(s)
- Binxia Yang
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Rajiv Janardhanan
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Pawan Vohra
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Eddie L Greene
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Santanu Bhattacharya
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sarah Withers
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Bhaskar Roy
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Evelyn C Nieves Torres
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Edward B Leof
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sanjay Misra
- 1] Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA [2] Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
11
|
Roy-Chaudhury P, Arnold P, Seigel J, Misra S. From basic biology to randomized clinical trial: the Beta Radiation for Arteriovenous Graft Outflow Stenosis (BRAVO II). Semin Dial 2012; 26:227-32. [PMID: 23067015 DOI: 10.1111/sdi.12000] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The BRAVO-II study was a randomized controlled study of endovascular radiation therapy as compared to sham radiation therapy, following angioplasty of a thrombosed PRFE graft. The results did not show a benefit of endovascular radiation therapy, albeit in the context of an early termination of the study at less than 50% enrollment due to business reasons. Emphasis is laid on the fact that there may still be a role for radiation therapy in specific clinical settings associated with dialysis vascular access dysfunction.
Collapse
Affiliation(s)
- Prabir Roy-Chaudhury
- Dialysis Vascular Research Group, Division of Nephrology, University of Cincinnati and VA Medical Center, Cincinnati, Ohio, USA.
| | | | | | | |
Collapse
|
12
|
Roy-Chaudhury P, El-Khatib M, Campos-Naciff B, Wadehra D, Ramani K, Leesar M, Mistry M, Wang Y, Chan JS, Lee T, Munda R. Back to the Future: How Biology and Technology Could Change the Role of PTFE Grafts in Vascular Access Management. Semin Dial 2012; 25:495-504. [DOI: 10.1111/j.1525-139x.2012.01091.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
13
|
Collins MJ, Li X, Lv W, Yang C, Protack CD, Muto A, Jadlowiec CC, Shu C, Dardik A. Therapeutic strategies to combat neointimal hyperplasia in vascular grafts. Expert Rev Cardiovasc Ther 2012; 10:635-47. [PMID: 22651839 PMCID: PMC3401520 DOI: 10.1586/erc.12.33] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Neointimal hyperplasia (NIH) in bypass conduits such as veins and prosthetic grafts is an important clinical entity that limits the long-term success of vascular interventions. Although the development of NIH in the conduits shares many of the same features of NIH that develops in native arteries after injury, vascular grafts are exposed to unique circumstances that predispose them to NIH, including surgical trauma related to vein handling, hemodynamic changes creating areas of low flow, and differences in biocompatibility between the conduit and the host environment. Multiple different approaches, including novel surgical techniques and targeted gene therapies, have been developed to target and prevent the causes of NIH. Recently, the PREVENT trials, the first molecular biology trials in vascular surgery aimed at preventing NIH, have failed to produce improved clinical outcomes, highlighting the incomplete knowledge of the pathways leading to NIH in vascular grafts. In this review, we aim to summarize the pathophysiologic pathways that underlie the formation of NIH in both vein and synthetic grafts and discuss current and potential mechanical and molecular approaches under investigation that may limit NIH in vascular grafts.
Collapse
Affiliation(s)
- Michael J Collins
- Department of Surgery and the Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, CT, USA
| | - Xin Li
- Department of Surgery and the Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, CT, USA
- Department of Vascular Surgery, Xiangya Second Hospital of Central South University, Changsha, Hunan, China
| | - Wei Lv
- Department of Surgery and the Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, CT, USA
- Department of Vascular Surgery, Shandong Provincial Hospital, Shandong University School of Medicine, Jinan, Shandong, China
| | - Chenzi Yang
- Department of Surgery and the Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, CT, USA
- Department of Vascular Surgery, Xiangya Second Hospital of Central South University, Changsha, Hunan, China
| | - Clinton D Protack
- Department of Surgery and the Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, CT, USA
| | - Akihito Muto
- Department of Thoracic and Cardiovascular Surgery, Mie University Graduate School of Medicine, Mie, Japan
| | - Caroline C Jadlowiec
- Department of Surgery and the Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, CT, USA
| | - Chang Shu
- Department of Vascular Surgery, Xiangya Second Hospital of Central South University, Changsha, Hunan, China
| | - Alan Dardik
- Department of Surgery and the Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT, USA
| |
Collapse
|
14
|
Basile C, Konner K, Lomonte C. The haemodialysis arteriovenous graft: is a new era coming? Nephrol Dial Transplant 2012; 27:876-8. [PMID: 22328734 DOI: 10.1093/ndt/gfs019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
15
|
Ahmed S, Roy-Chaudhury P. Radiation therapy for dialysis access stenosis: unfulfilled promise or false expectations. Semin Dial 2012; 25:464-9. [PMID: 22276964 DOI: 10.1111/j.1525-139x.2011.01006.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Hemodialysis vascular access dysfunction is a major cause of morbidity and hospitalization in the hemodialysis population at a cost of well over $1 billion per annum. Venous stenosis (due to venous neointimal hyperplasia [VNH]) is the most common cause of polytetrafluroethylene PTFE) dialysis access graft and arteriovenous fistula (AVF) failure. Despite the magnitude of the clinical problem, however, there are currently no effective therapies for this condition. We and others have previously demonstrated that VNH in PTFE dialysis grafts and AVF is composed of smooth muscle cells/myofibroblasts, endothelial cells within neointimal microvessels, and peri-graft macrophages. Radiation therapy blocks the proliferation and activation of all these cell types. The current review will dissect out the available in vitro, experimental, and clinical data on the use of radiation therapy for vascular stenosis in general, and for dialysis access dysfunction in particular. It is important to try and identify whether there is still a role for radiation therapy in this specific clinical setting. We believe that this is a critically important question to answer in view of the huge unmet clinical need that is currently associated with hemodialysis vascular access dysfunction.
Collapse
Affiliation(s)
- Syed Ahmed
- Dialysis Vascular Access Research Group, Division of Nephrology, University of Cincinnati, Cincinnati, Ohio 45267-0585, USA
| | | |
Collapse
|
16
|
Stent-Grafts Improve Secondary Patency of Failing Hemodialysis Grafts. J Vasc Access 2011; 13:65-70. [DOI: 10.5301/jva.2011.8485] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2011] [Indexed: 11/20/2022] Open
Abstract
Purpose Failing hemodialysis grafts continue to pose a challenge in the care of patients with end-stage renal disease (ESRD). We review our experience using percutaneous stent-grafts for the treatment of venous outflow stenosis and occlusion in order to assess their efficacy in nonautologous graft salvage. Methods This is a retrospective review of patients treated with percutaneous Viabahn® stent-grafts for failing hemodialysis arteriovenous grafts (AVGs), between 6/2006 and 12/2009. All stent-grafts were deployed across the venous anastomosis to address the outflow obstruction. Patency and re-intervention rates were estimated using Kaplan-Meier analysis. Results Twenty patients had stent-grafts placed. Successful stent-grafting was defined as <30% residual stenosis and the ability to dialyze through the graft post-treatment. Technical success for stent-grafting was 100%. Median follow-up was 23 months (range 3 to 37 months). Stent-graft patency at 6 months: 94.7%, 12 months: 94.7%, 18 months: 82.1%, 24 months: 82.1%, 36 months: 82.1%. Freedom from re-intervention was 69% at 24 months and 50% at 36 months. Two AVGs failed and had to be removed without further interventions, because of complete occlusion and infection. Two patients (10%) required re-intervention for arterial inflow stenosis. One patient (5%) required balloon angioplasty and stenting of stenosis distal to the stent-graft. One patient (5%) developed in-stent stenosis and was treated with placement of a new stent. Graft salvage rate was 80% at 36 months. Conclusions Stent-grafts can be successfully used to improve freedom from re-intervention rates and overall patency rates of failing AVGs.
Collapse
|
17
|
Yevzlin AS, Chan MR, Becker YT, Roy-Chaudhury P, Lee T, Becker BN. "Venopathy" at work: recasting neointimal hyperplasia in a new light. Transl Res 2010; 156:216-25. [PMID: 20875897 PMCID: PMC4310704 DOI: 10.1016/j.trsl.2010.07.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 07/05/2010] [Accepted: 07/13/2010] [Indexed: 12/26/2022]
Abstract
Hemodialysis vascular access is a unique form of vascular anastomosis. Although it is created in a unique disease state, it has much to offer in terms of insights into venous endothelial and anastomotic biology. The development of neointimal hyperplasia (NH) has been identified as a pathologic entity, decreasing the lifespan and effectiveness of hemodialysis vascular access. Subtle hints and new data suggest a contrary idea-that NH, to some extent an expected response, if controlled properly, may play a beneficial role in the promotion of maturation to a functional access. This review attempts to recast our understanding of NH and redefine research goals for an evolving discipline that focuses on a life-sustaining connection between an artery and vein.
Collapse
Affiliation(s)
- Alexander S Yevzlin
- Departments of Medicine and Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wis, USA
| | | | | | | | | | | |
Collapse
|
18
|
Misra S, Fu AA, Misra KD, Shergill UM, Leof EB, Mukhopadhyay D. Hypoxia-induced phenotypic switch of fibroblasts to myofibroblasts through a matrix metalloproteinase 2/tissue inhibitor of metalloproteinase-mediated pathway: implications for venous neointimal hyperplasia in hemodialysis access. J Vasc Interv Radiol 2010; 21:896-902. [PMID: 20434368 DOI: 10.1016/j.jvir.2010.02.030] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Revised: 02/08/2010] [Accepted: 02/18/2010] [Indexed: 01/16/2023] Open
Abstract
PURPOSE Hemodialysis grafts fail because of venous neointimal hyperplasia formation caused by adventitial fibroblasts that have become myofibroblasts (ie, alpha-smooth muscle actin [SMA]-positive cells) and migrate to the neointima. There is increased expression of hypoxia-inducible factor (HIF)-1alpha in venous neointimal hyperplasia formation in experimental animal models and clinical samples. It was hypothesized that, under hypoxic stimulus (ie, HIF-1alpha), fibroblasts will convert to myofibroblasts through a matrix metalloproteinase (MMP)-2-mediated pathway. MATERIALS AND METHODS Murine AKR-2B fibroblasts were made hypoxic or normoxic for 24, 48, and 72 hours. Protein expression for HIF-1alpha, alpha-SMA, MMP-2, MMP-9, tissue inhibitor of metalloproteinase (TIMP)-1, and TIMP-2 was performed to determine the kinetic changes of these proteins. Immunostaining for alpha-SMA, collagen, and fibronectin was performed. RESULTS At all time points, there was significantly increased expression of HIF-1alpha in the hypoxic fibroblasts compared with normoxic fibroblasts (P < .05). There was significantly increased expression of alpha-SMA at all time points, which peaked by 48 hours in hypoxic fibroblasts compared with normoxic fibroblasts (P < .05). There was a significant increase in the expression of active MMP-2 by 48-72 hours and a significant increase in TIMP-1 by 48-72 hours by hypoxic fibroblasts (P < .05). By 72 hours, there was significant increase in TIMP-2 expression (P < .05). Immunohistochemical analysis demonstrated increased expression of alpha-SMA, collagen, and fibronectin as the duration of hypoxia increased. CONCLUSIONS Under hypoxic conditions, fibroblasts will convert to myofibroblasts through an MMP-2-mediated pathway, which may provide insight into the mechanism of venous neointimal hyperplasia.
Collapse
Affiliation(s)
- Sanjay Misra
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
| | | | | | | | | | | |
Collapse
|
19
|
Pathology of explanted polytetrafluoroethylene vascular grafts. Cardiovasc Pathol 2010; 20:213-21. [PMID: 20619685 DOI: 10.1016/j.carpath.2010.06.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Revised: 05/26/2010] [Accepted: 06/03/2010] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Graft occlusion is a well-documented etiology for arteriovenous fistulae failure. However, there is little morphologic information elucidating why synthetic vascular grafts fail. The purpose of this study was to examine the tissue responses occurring within and adjacent to explanted polytetrafluoroethylene grafts that were utilized during cardiovascular procedures and subsequently removed. METHODS Forty explanted polytetrafluoroethylene grafts (including 32 failed vascular grafts) originating from 18 females and 22 males who ranged in age from 6 to 82 years (mean age, 36 years) were evaluated. Duration of engraftment varied from 1 to 255 months (mean engraftment period, 64 months). RESULTS In addition to neointimal hyperplasia, foreign body reaction, and thrombosis, an unexpected finding was calcification involving the graft material, as well as luminal thrombus and adjacent soft tissues. Twenty-seven of forty cases (68%) showed evidence of calcification, either within or adjacent to polytetrafluoroethylene grafts. Histologic examination revealed variable degrees and patterns of calcification within and adjacent to explanted polytetrafluoroethylene membranes and conduits (arterial, arteriovenous, or cardiac grafts). A significantly longer duration of engraftment (P=.015) was identified in calcified versus noncalcified polytetrafluoroethylene materials. Patient age, serum calcium, creatinine level, and blood urea nitrogen level showed no statistically significant differences between patients with calcified grafts and patients without calcified grafts. CONCLUSIONS Interstitial calcification is frequently found within explanted polytetrafluoroethylene grafts and is associated with graft disruption. These findings suggest that calcification of polytetrafluoroethylene biomaterials may play a role in eventual graft failure. A better understanding of the process of polytetrafluoroethylene graft calcification may lead to novel therapies that aid in the prevention of polytetrafluoroethylene vascular graft failure.
Collapse
|
20
|
Huijbregts H, de Borst G, Veldhuis W, Verhagen H, Velema E, Pasterkamp G, Moll F, Blankestijn P, Hoefer I. Cryoplasty of the Venous Anastomosis for Prevention of Intimal Hyperplasia in a Validated Porcine Arteriovenous Graft Model. Eur J Vasc Endovasc Surg 2010; 39:620-6. [DOI: 10.1016/j.ejvs.2009.12.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Accepted: 12/25/2009] [Indexed: 11/26/2022]
|
21
|
Haskal ZJ, Trerotola S, Dolmatch B, Schuman E, Altman S, Mietling S, Berman S, McLennan G, Trimmer C, Ross J, Vesely T. Stent graft versus balloon angioplasty for failing dialysis-access grafts. N Engl J Med 2010; 362:494-503. [PMID: 20147715 DOI: 10.1056/nejmoa0902045] [Citation(s) in RCA: 254] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND The leading cause of failure of a prosthetic arteriovenous hemodialysis-access graft is venous anastomotic stenosis. Balloon angioplasty, the first-line therapy, has a tendency to lead to subsequent recoil and restenosis; however, no other therapies have yet proved to be more effective. This study was designed to compare conventional balloon angioplasty with an expanded polytetrafluoroethylene endovascular stent graft for revision of venous anastomotic stenosis in failing hemodialysis grafts. METHODS We conducted a prospective, multicenter trial, randomly assigning 190 patients who were undergoing hemodialysis and who had a venous anastomotic stenosis to undergo either balloon angioplasty alone or balloon angioplasty plus placement of the stent graft. Primary end points included patency of the treatment area and patency of the entire vascular access circuit. RESULTS At 6 months, the incidence of patency of the treatment area was significantly greater in the stent-graft group than in the balloon-angioplasty group (51% vs. 23%, P<0.001), as was the incidence of patency of the access circuit (38% vs. 20%, P=0.008). In addition, the incidence of freedom from subsequent interventions at 6 months was significantly greater in the stent-graft group than in the balloon-angioplasty group (32% vs. 16%, P=0.03 by the log-rank test and P=0.04 by the Wilcoxon rank-sum test). The incidence of binary restenosis at 6 months was greater in the balloon-angioplasty group than in the stent-graft group (78% vs. 28%, P<0.001). The incidences of adverse events at 6 months were equivalent in the two treatment groups, with the exception of restenosis, which occurred more frequently in the balloon-angioplasty group (P<0.001). CONCLUSIONS In this study, percutaneous revision of venous anastomotic stenosis in patients with a prosthetic hemodialysis graft was improved with the use of a stent graft, which appears to provide longer-term and superior patency and freedom from repeat interventions than standard balloon angioplasty. (ClinicalTrials.gov number, NCT00678249.)
Collapse
Affiliation(s)
- Ziv J Haskal
- Division of Vascular and Interventional Radiology, University of Maryland Medical Center, 22 S. Greene St., GK214, Baltimore, MD 21201, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Roy-Chaudhury P, Lee T, Duncan H, El-Khatib M. Combining novel technologies with improved logistics to reduce hemodialysis vascular access dysfunction. J Vasc Access 2009; 10:1-4. [PMID: 19340792 DOI: 10.1177/112972980901000101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Hemodialysis (HD) vascular access dysfunction is currently a huge clinical problem for which there are no effective therapies. There are, however, a number of promising technologies that are currently at the experimental or clinical trial stage. We believe that the application of these novel technologies in combination with better clinical protocols for vascular access care could significantly reduce the current problems associated with HD vascular access.
Collapse
Affiliation(s)
- P Roy-Chaudhury
- Cincinnati Dialysis Access Program, Cincinnati, OH - USA and University of Cincinnati and the VA Medical Center, Cincinnati, OH 45267-0585 USA.
| | | | | | | |
Collapse
|
23
|
Diskin CJ, Stokes TJ, Dansby LM, Radcliff L, Carter TB. Understanding the pathophysiology of hemodialysis access problems as a prelude to developing innovative therapies. ACTA ACUST UNITED AC 2008; 4:628-38. [DOI: 10.1038/ncpneph0947] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2008] [Accepted: 08/06/2008] [Indexed: 11/09/2022]
|
24
|
Fetuin-A expression in early venous stenosis formation in a porcine model of hemodialysis graft failure. J Vasc Interv Radiol 2008; 19:1477-82. [PMID: 18693047 DOI: 10.1016/j.jvir.2008.06.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2008] [Revised: 06/01/2008] [Accepted: 06/03/2008] [Indexed: 01/16/2023] Open
Abstract
PURPOSE Because fetuin-A is a cytokine with multifunctional effects on vascular smooth muscle cells and fibroblasts, the authors examined the course of its expression in early venous stenosis formation in a porcine model of chronic renal insufficiency with polytetrafluoroethylene (PTFE) arteriovenous (AV) hemodialysis grafts. MATERIALS AND METHODS Pigs had chronic renal insufficiency created by complete embolization of the left kidney and partial embolization of the right kidney. Twenty-eight days later, PTFE AV grafts were placed from the carotid artery to the ipsilateral jugular vein, and the animals were euthanized 3 days (n = 4), 7 days (n = 4), or 14 days (n = 4) later. Expression of fetuin-A was determined by Western blot analysis of the venous stenosis, control veins, and plasma. Immunohistochemical analysis of the venous stenosis and control vein was performed. Blood urea nitrogen (BUN) and creatinine were measured before embolization and at the time of graft placement. RESULTS The mean BUN and creatinine levels at graft placement were significantly higher than before embolization (P < .05). Severe venous neointimal hyperplasia occurred by day 14 and was characterized by primarily alpha-smooth muscle actin-positive cells. By day 14, fetuin-A levels had increased significantly (P < .05) at the venous stenosis compared with control veins and in the serum compared with measurements before embolization. CONCLUSIONS Significantly increased expression of fetuin-A was observed in early venous stenosis by day 14 and in serum compared with baseline measurements. Understanding the role of fetuin-A in venous neointimal hyperplasia could help in improving outcomes in patients undergoing hemodialysis.
Collapse
|
25
|
Expression of hypoxia inducible factor-1 alpha, macrophage migration inhibition factor, matrix metalloproteinase-2 and -9, and their inhibitors in hemodialysis grafts and arteriovenous fistulas. J Vasc Interv Radiol 2008; 19:252-9. [PMID: 18341958 DOI: 10.1016/j.jvir.2007.10.031] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2007] [Revised: 10/14/2007] [Accepted: 10/15/2007] [Indexed: 12/27/2022] Open
Abstract
PURPOSE It is well recognized that arteriovenous fistulas (AVFs) used for hemodialysis access have better primary patency rates with less restenosis than polytetrafluoroethylene (PTFE) grafts; however, the mechanism responsible for this is not known. Recent data suggest that hypoxia inducible factor-1 alpha (HIF-1 alpha) is associated with vascular restenosis, possibly through mechanisms that increase the production of macrophage migration inhibition factor (MIF), matrix metalloproteinase-2 (MMP-2) and MMP-9, and their inhibitors (tissue inhibitor of MMPs; TIMP). The present study tested the hypothesis that there are differences in the expression patterns of HIF-1 alpha, MIF, MMP-2, MMP-9, and TIMPs in specimens removed from patients with AVFs and PTFE grafts. MATERIALS AND METHODS Whole-vessel tissue samples were obtained from the vein distal to the vein-to-PTFE graft anastomosis and the proximal outflow vein (within 6 cm of the arteriovenous anastomosis) of AVFs from 17 patients who required a surgical revision for thrombosis and stenosis. Nonstenotic veins of four patients undergoing hemodialysis vascular access placement were used as controls. PTFE grafts (n = 6), AVFs (n = 6), and control samples (n = 3) underwent Western blot analysis and zymography. A separate group of five patients with PTFE hemodialysis grafts and one control subject were used for immunohistochemical analysis. RESULTS Specimens from patients with PTFE grafts had significantly higher expression of HIF-1 alpha (P = .03), MIF (P = .02), TIMP-1 (P = .0006), pro-MMP-2 (P = .02), and pro-MMP-9 (P = .046) compared with control veins. The expression of only pro-MMP-9 was significantly higher in AVFs compared with control samples (P = .004). There was a significant increase in the expression of MIF (P = .007) and TIMP-1 (P < .0001) in PTFE graft specimens compared with AVFs. MIF and TIMP-1 were localized to the adventitia of the vein distal to the vein-to-PTFE graft anastomosis. CONCLUSIONS There were major differences in the expression patterns of hypoxia (ie, HIF-1 alpha) and proteins regulated by HIF-1?, including MIF, pro-MMP-2, pro-MMP-9, and TIMP-1, in specimens removed from patients with PTFE grafts and AVFs. Understanding the role of HIF-1 alpha and these proteins in hemodialysis access failure can help improve outcomes.
Collapse
|
26
|
|
27
|
Abstract
Optimizing vascular access outcomes remains an ongoing challenge for clinical nephrologists. All other things being equal, fistulas are preferred over grafts, and grafts are preferred over catheters. Mature fistulas have better longevity and require fewer interventions, as compared with mature grafts. The major hurdle to increasing fistula use is the high rate of failure to mature of newly created fistulas. There is a desperate need for enhanced understanding of the mechanisms of failure to mature and the optimal type and timing of interventions to promote maturity. Grafts are prone to frequent stenosis and thrombosis. Surveillance for graft stenosis with preemptive angioplasty may reduce graft thrombosis, but recent randomized clinical trials have questioned the efficacy of this approach. Graft stenosis results from aggressive neointimal hyperplasia, and pharmacologic approaches to slowing this process are being investigated in clinical trials. Catheters are prone to frequent thrombosis and infection. The optimal management of catheter-related bacteremia is a subject of ongoing debate. Prophylaxis of catheter-related bacteremia continues to generate important clinical research. Close collaboration among nephrologists, surgeons, radiologists, and the dialysis staff is required to optimize vascular access outcomes and can be expedited by having a dedicated access coordinator to streamline the process. The goal of this review is to provide an update on the current status of vascular access management.
Collapse
|
28
|
Zhang J, Melhem M, Kassing W, Kelly B, Wang Y, Krishnamoorthy M, Heffelfinger S, Desai P, Roy-Chaudhury P. In vitro Paclitaxel and Radiation Effects on the Cell Types Responsible for Vascular Stenosis: A Preliminary Analysis. Blood Purif 2006; 25:155-60. [PMID: 17179736 DOI: 10.1159/000098018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2006] [Accepted: 08/02/2006] [Indexed: 11/19/2022]
Abstract
Hemodialysis vascular access dysfunction as a result of venous neointimal hyperplasia in dialysis access grafts and fistulae is currently a huge clinical problem. The aim of this study was to assess the effects of paclitaxel and radiation, both singly and in combination on the proliferation of cell types present within the lesion of venous neointimal hyperplasia (vascular smooth muscle cells, fibroblasts and endothelial cells within the neointimal microvessels). Vascular smooth muscle cells, fibroblasts and endothelial cells were plated onto 96-well plates and exposed to different concentrations and doses of paclitaxel and radiation, respectively (both individually and in combination). Growth inhibition was assessed with an MTT assay. Both paclitaxel and radiation resulted in significant growth inhibition of all three cell types. However, even small doses of paclitaxel appeared to attenuate the antiproliferative effect of radiation on these cell types. Further experiments to elucidate the mechanism behind these findings could result in a better understanding of combination antiproliferative therapies.
Collapse
Affiliation(s)
- J Zhang
- Department of Medicine, University of Cincinnati, Cincinnati, Ohio 45267-0585, USA
| | | | | | | | | | | | | | | | | |
Collapse
|