Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Review Article
  • Published: 07 January 2022

Update on the pathophysiology and medical treatment of peripheral artery disease

  • Jonathan Golledge   ORCID: orcid.org/0000-0002-5779-8848 1 , 2 , 3  

Nature Reviews Cardiology volume  19 ,  pages 456–474 ( 2022 ) Cite this article

6091 Accesses

68 Citations

32 Altmetric

Metrics details

  • Pathogenesis
  • Peripheral vascular disease
  • Therapeutics

Approximately 6% of adults worldwide have atherosclerosis and thrombosis of the lower limb arteries (peripheral artery disease (PAD)) and the prevalence is rising. PAD causes leg pain, impaired health-related quality of life, immobility, tissue loss and a high risk of major adverse events, including myocardial infarction, stroke, revascularization, amputation and death. In this Review, I describe the pathophysiology, presentation, outcome, preclinical research and medical management of PAD. Established treatments for PAD include antithrombotic drugs, such as aspirin and clopidogrel, and medications to treat dyslipidaemia, hypertension and diabetes mellitus. Randomized controlled trials have demonstrated that these treatments reduce the risk of major adverse events. The drug cilostazol, exercise therapy and revascularization are the current treatment options for the limb symptoms of PAD, but each has limitations. Novel therapies to promote collateral and new capillary growth and treat PAD-related myopathy are under investigation. Methods to improve the implementation of evidence-based medical management, novel drug therapies and rehabilitation programmes for PAD-related pain, functional impairment and ischaemic foot disease are important areas for future research.

Peripheral artery disease (PAD) is present in 6% of adults and is associated with leg pain, walking impairment and high risk of major adverse cardiovascular events, including amputation and death.

PAD usually presents with leg pain, ischaemic ulceration or gangrene and is usually caused by atherosclerosis and thrombosis, although all current animal models use surgically created hindlimb ischaemia.

Treatment options for the leg symptoms of PAD include cilostazol, exercise therapy and revascularization, with novel therapies under investigation including autologous cell therapy, organic nitrates and antioxidants.

Antithrombotic and LDL-cholesterol-lowering medications, smoking cessation and treatment of hypertension and diabetes mellitus reduce the risk of major adverse events, but programmes to improve uptake of these measures are needed.

This is a preview of subscription content, access via your institution

Access options

Access Nature and 54 other Nature Portfolio journals

Get Nature+, our best-value online-access subscription

24,99 € / 30 days

cancel any time

Subscribe to this journal

Receive 12 print issues and online access

195,33 € per year

only 16,28 € per issue

Buy this article

  • Purchase on Springer Link
  • Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

peripheral vascular disease research paper

Similar content being viewed by others

peripheral vascular disease research paper

The role of Klotho and FGF23 in cardiovascular outcomes of diabetic patients with chronic limb threatening ischemia: a prospective study

peripheral vascular disease research paper

NT-proBNP as a surrogate for unknown heart failure and its predictive power for peripheral artery disease outcome and phenotype

peripheral vascular disease research paper

Development of a two-stage limb ischemia model to better simulate human peripheral artery disease

Nastasi, D. R. et al. The cost-effectiveness of intensive low-density lipoprotein cholesterol lowering in people with peripheral artery disease. J. Vasc. Surg. 73 , 1396–1403 (2021).

Article   PubMed   Google Scholar  

Anand, S. S. et al. Major adverse limb events and mortality in patients with peripheral artery disease: the COMPASS trial. J. Am. Coll. Cardiol. 71 , 2306–2315 (2018).

McDermott, M. M. et al. Leg symptom categories and rates of mobility decline in peripheral arterial disease. J. Am. Geriatr. Soc. 58 , 1256–1262 (2010).

Article   PubMed   PubMed Central   Google Scholar  

Sampson, U. K. et al. Global and regional burden of death and disability from peripheral artery disease: 21 world regions, 1990 to 2010. Glob. Heart 9 , 145–158 (2014).

Song, P. et al. Global, regional, and national prevalence and risk factors for peripheral artery disease in 2015: an updated systematic review and analysis. Lancet Glob. Health 7 , e1020–e1030 (2019).

Criqui, M. H. et al. Lower extremity peripheral artery disease: contemporary epidemiology, management gaps, and future directions: a scientific statement from the American Heart Association. Circulation 144 , e171–e191 (2021).

Golledge, J. & Drovandi, A. Evidence-based recommendations for medical management of peripheral artery disease. J. Atheroscler. Thromb. 28 , 573–583 (2021).

Article   CAS   PubMed   PubMed Central   Google Scholar  

Bonaca, M. P. et al. Low-density lipoprotein cholesterol lowering with evolocumab and outcomes in patients with peripheral artery disease: insights from the FOURIER trial (Further Cardiovascular Outcomes Research with PCSK9 Inhibition in Subjects with Elevated Risk). Circulation 137 , 338–350 (2018).

Article   CAS   PubMed   Google Scholar  

Anand, S. S. et al. Rivaroxaban with or without aspirin in patients with stable peripheral or carotid artery disease: an international, randomised, double-blind, placebo-controlled trial. Lancet 391 , 219–229 (2018).

Cooke, J. P. & Meng, S. Vascular regeneration in peripheral artery disease. Arterioscler. Thromb. Vasc. Biol. 40 , 1627–1634 (2020).

Gerhard-Herman, M. D. et al. 2016 AHA/ACC guideline on the management of patients with lower extremity peripheral artery disease: a report of the American College of Cardiology/American Heart Association task force on clinical practice guidelines. Circulation 135 , e726–e779 (2017).

PubMed   Google Scholar  

Aboyans, V. et al. Editor’s Choice - 2017 ESC Guidelines on the Diagnosis and Treatment of Peripheral Arterial Diseases, in collaboration with the European Society for Vascular Surgery (ESVS). Eur. J. Vasc. Endovasc. Surg. 55 , 305–368 (2018).

Abola, M. T. B. et al. Asia-Pacific consensus statement on the management of peripheral artery disease: a report from the Asian Pacific Society of Atherosclerosis and Vascular Disease Asia-Pacific Peripheral Artery Disease Consensus Statement Project Committee. J. Atheroscler. Thromb. 27 , 809–907 (2020).

Schorr, E. N. & Treat-Jacobson, D. Methods of symptom evaluation and their impact on peripheral artery disease (PAD) symptom prevalence: a review. Vasc. Med. 18 , 95–111 (2013).

Conte, M. S. et al. Global vascular guidelines on the management of chronic limb-threatening ischemia. Eur. J. Vasc. Endovasc. Surg. 58 , S1–S109 (2019).

Fowkes, F. G. et al. Comparison of global estimates of prevalence and risk factors for peripheral artery disease in 2000 and 2010: a systematic review and analysis. Lancet 382 , 1329–1340 (2013).

Hirsch, A. T. et al. A call to action: women and peripheral artery disease: a scientific statement from the American Heart Association. Circulation 125 , 1449–1472 (2012).

Srivaratharajah, K. & Abramson, B. L. Women and peripheral arterial disease: a review of sex differences in epidemiology, clinical manifestations, and outcomes. Can. J. Cardiol. 34 , 356–361 (2018).

Heart Protection Study Collaborative Group. Randomized trial of the effects of cholesterol-lowering with simvastatin on peripheral vascular and other major vascular outcomes in 20,536 people with peripheral arterial disease and other high-risk conditions. J. Vasc. Surg. 45 , 645–654 (2007).

Article   Google Scholar  

Marjoribanks, J., Farquhar, C., Roberts, H., Lethaby, A. & Lee, J. Long-term hormone therapy for perimenopausal and postmenopausal women. Cochrane Database Syst. Rev. 1 , CD004143 (2017).

Choi, J., Joseph, L. & Pilote, L. Obesity and C-reactive protein in various populations: a systematic review and meta-analysis. Obes. Rev. 14 , 232–244 (2013).

Aboyans, V. et al. Intrinsic contribution of gender and ethnicity to normal ankle-brachial index values: the Multi-Ethnic Study of Atherosclerosis (MESA). J. Vasc. Surg. 45 , 319–327 (2007).

Fowkes, F. G. et al. Peripheral artery disease: epidemiology and global perspectives. Nat. Rev. Cardiol. 14 , 156–170 (2017).

Allison, M. A. et al. Ethnic-specific prevalence of peripheral arterial disease in the United States. Am. J. Prev. Med. 32 , 328–333 (2007).

Klarin, D. et al. Genome-wide association study of peripheral artery disease in the Million Veteran Program. Nat. Med. 25 , 1274–1279 (2019).

Harwood, A. E. et al. Exercise training for intermittent claudication: a narrative review and summary of guidelines for practitioners. BMJ Open Sport. Exerc. Med. 6 , e000897 (2020).

Hirsch, A. T. et al. Peripheral arterial disease detection, awareness, and treatment in primary care. JAMA 286 , 1317–1324 (2001).

Zhang, Y. et al. Global disability burdens of diabetes-related lower-extremity complications in 1990 and 2016. Diabetes Care 43 , 964–974 (2020).

Prompers, L. et al. High prevalence of ischaemia, infection and serious comorbidity in patients with diabetic foot disease in Europe. Baseline results from the Eurodiale study. Diabetologia 50 , 18–25 (2007).

Morbach, S. et al. Long-term prognosis of diabetic foot patients and their limbs: amputation and death over the course of a decade. Diabetes Care 35 , 2021–2027 (2012).

Rosi, L. M., Jones, A. S., Topliss, D. J. & Bach, L. A. Demographics and outcomes of inpatients with diabetic foot ulcers treated conservatively and surgically in a metropolitan hospital network. Diabetes Res. Clin. Pract. 175 , 108821 (2021).

Misra, S. et al. Perfusion assessment in critical limb ischemia: principles for understanding and the development of evidence and evaluation of devices: a scientific statement from the American Heart Association. Circulation 140 , e657–e672 (2019).

Criqui, M. H. & Aboyans, V. Epidemiology of peripheral artery disease. Circ. Res. 116 , 1509–1526 (2015).

Golledge, J. et al. High ankle brachial index predicts high risk of cardiovascular events amongst people with peripheral artery disease. PLoS ONE 15 , e0242228 (2020).

McDermott, M. M. et al. The ankle brachial index is associated with leg function and physical activity: the Walking and Leg Circulation Study. Ann. Intern. Med. 136 , 873–883 (2002).

Golledge, J. et al. Relationship between disease specific quality of life measures, physical performance, and activity in people with intermittent claudication caused by peripheral artery disease. Eur. J. Vasc. Endovasc. Surg. 59 , 957–964 (2020).

Nayak, P. et al. Association of six-minute walk distance with subsequent lower extremity events in peripheral artery disease. Vasc. Med. 25 , 319–327 (2020).

McDermott, M. M. et al. Femoral artery plaque characteristics, lower extremity collaterals, and mobility loss in peripheral artery disease. Vasc. Med. 22 , 473–481 (2017).

McDermott, M. M. et al. Unsupervised exercise and mobility loss in peripheral artery disease: a randomized controlled trial. J. Am. Heart Assoc. 4 , e001659 (2015).

McDermott, M. M. et al. Pathophysiological changes in calf muscle predict mobility loss at 2-year follow-up in men and women with peripheral arterial disease. Circulation 120 , 1048–1055 (2009).

McDermott, M. M. et al. Community walking speed, sedentary or lying down time, and mortality in peripheral artery disease. Vasc. Med. 21 , 120–129 (2016).

Morris, D. R. et al. Association of lower extremity performance with cardiovascular and all-cause mortality in patients with peripheral artery disease: a systematic review and meta-analysis. J. Am. Heart Assoc. 3 , e001105 (2014).

McDermott, M. M. et al. Association of 6-minute walk performance and physical activity with incident ischemic heart disease events and stroke in peripheral artery disease. J. Am. Heart Assoc. 4 , e001846 (2015).

McDermott, M. M. et al. Decline in functional performance predicts later increased mobility loss and mortality in peripheral arterial disease. J. Am. Coll. Cardiol. 57 , 962–970 (2011).

Sigvant, B., Lundin, F. & Wahlberg, E. The risk of disease progression in peripheral arterial disease is higher than expected: a meta-analysis of mortality and disease progression in peripheral arterial disease. Eur. J. Vasc. Endovasc. Surg. 51 , 395–403 (2016).

Faglia, E. et al. Early and five-year amputation and survival rate of diabetic patients with critical limb ischemia: data of a cohort study of 564 patients. Eur. J. Vasc. Endovasc. Surg. 32 , 484–490 (2006).

Surinach, J. M. et al. Differences in cardiovascular mortality in smokers, past-smokers and non-smokers: findings from the FRENA registry. Eur. J. Intern. Med. 20 , 522–526 (2009).

Rymer, J. A. et al. Association of disease progression with cardiovascular and limb outcomes in patients with peripheral artery disease: insights from the EUCLID trial. Circ. Cardiovasc. Interv. 13 , e009326 (2020).

Hiatt, W. R. et al. Ticagrelor versus clopidogrel in symptomatic peripheral artery disease. N. Engl. J. Med. 376 , 32–40 (2017).

McDermott, M. M. et al. Associations of borderline and low normal ankle-brachial index values with functional decline at 5-year follow-up: the WALCS (Walking and Leg Circulation Study). J. Am. Coll. Cardiol. 53 , 1056–1062 (2009).

Narcisse, D. I. et al. The association of healthcare disparities and patient-specific factors on clinical outcomes in peripheral artery disease. Am. Heart J. 239 , 135–146 (2021).

Makowski, L. et al. Sex related differences in therapy and outcome of patients with intermittent claudication in a real-world cohort. Atherosclerosis 325 , 75–82 (2021).

Djerf, H. et al. Low risk of procedure related major amputation following revascularisation for intermittent claudication: a population based study. Eur. J. Vasc. Endovasc. Surg. 59 , 817–822 (2020).

Madabhushi, V. et al. Revascularization of intermittent claudicants leads to more chronic limb-threatening ischemia and higher amputation rates. J. Vasc. Surg. 74 , 771–779 (2021).

Kim, T. I. et al. Multiple reinterventions for claudication are associated with progression to chronic limb-threatening ischemia. Ann. Vasc. Surg. 72 , 166–174 (2021).

Verwer, M. C., Wijnand, J. G. J., Teraa, M., Verhaar, M. C. & de Borst, G. J. Long term survival and limb salvage in patients with non-revascularisable chronic limb threatening ischaemia. Eur. J. Vasc. Endovasc. Surg. 62 , 225–232 (2021).

de Donato, G. et al. Evaluation of clinical outcomes after revascularization in patients with chronic limb-threatening ischemia: results from a prospective national cohort study (RIVALUTANDO). Angiology 72 , 480–489 (2021).

Article   PubMed   CAS   Google Scholar  

Wijeyaratne, M. et al. Clinical outcomes following lower extremity vein bypass for chronic limb threatening ischaemia (CLTI) at the University of Colombo, Sri Lanka. Eur. J. Vasc. Endovasc. Surg. 60 , 560–566 (2020).

Kodama, A. et al. Editor’s Choice - relationship between Global Limb Anatomic Staging System (GLASS) and clinical outcomes following revascularisation for chronic limb threatening ischaemia in the bypass versus angioplasty in severe ischaemia of the leg (BASIL)-1 Trial. Eur. J. Vasc. Endovasc. Surg. 60 , 687–695 (2020).

Tay, W. L. et al. Two-year clinical outcomes following lower limb endovascular revascularisation for chronic limb threatening ischaemia at a tertiary Asian vascular centre in Singapore. Singap. Med. J. https://doi.org/10.11622/smedj.2020104 (2020).

Long, C. A. et al. Incidence and factors associated with major amputation in patients with peripheral artery disease: insights from the EUCLID trial. Circ. Cardiovasc. Qual. Outcomes 13 , e006399 (2020).

Gutierrez, J. A. et al. Polyvascular disease and risk of major adverse cardiovascular events in peripheral artery disease: a secondary analysis of the EUCLID trial. JAMA Netw. Open. 1 , e185239 (2018).

Smith, S. L., Matthews, E. O., Moxon, J. V. & Golledge, J. A systematic review and meta-analysis of risk factors for and incidence of 30-day readmission after revascularization for peripheral artery disease. J. Vasc. Surg. 70 , 996–1006 (2019).

Smith, S. L. et al. Outcomes and costs of open and endovascular revascularisation for chronic limb ischaemia in an australian cohort. Heart Lung Circ. 30 , 1552–1561 (2021).

Kaplovitch, E. et al. Rivaroxaban and aspirin in patients with symptomatic lower extremity peripheral artery disease: a subanalysis of the COMPASS randomized clinical trial. JAMA Cardiol. 6 , 21–29 (2021).

Hopley, C. W. et al. Chronic kidney disease and risk for cardiovascular and limb outcomes in patients with symptomatic peripheral artery disease: the EUCLID trial. Vasc. Med. 24 , 422–430 (2019).

Samsky, M. D. et al. Association of heart failure with outcomes among patients with peripheral artery disease: insights from EUCLID. J. Am. Heart Assoc. 10 , e018684 (2021).

Galani, J. et al. Association of chronic obstructive pulmonary disease with morbidity and mortality in patients with peripheral artery disease: insights from the EUCLID trial. Int. J. Chron. Obstruct Pulmon Dis. 16 , 841–851 (2021).

Cronin, O., Morris, D. R., Walker, P. J. & Golledge, J. The association of obesity with cardiovascular events in patients with peripheral artery disease. Atherosclerosis 228 , 316–323 (2013).

Fudim, M. et al. Association of hypertension and arterial blood pressure on limb and cardiovascular outcomes in symptomatic peripheral artery disease: the EUCLID trial. Circ. Cardiovasc. Qual. Outcomes 13 , e006512 (2020).

Boc, V., Bozic Mijovski, M., Pohar Perme, M. & Blinc, A. Diabetes and smoking are more important for prognosis of patients with peripheral arterial disease than some genetic polymorphisms. Vasa 48 , 229–235 (2019).

Rymer, J. A. et al. Association of health status scores with cardiovascular and limb outcomes in patients with symptomatic peripheral artery disease: insights from the EUCLID (Examining Use of Ticagrelor in Symptomatic Peripheral Artery Disease) trial. J. Am. Heart Assoc. 9 , e016573 (2020).

Singh, T. P., Morris, D. R., Smith, S., Moxon, J. V. & Golledge, J. Systematic review and meta-analysis of the association between C-reactive protein and major cardiovascular events in patients with peripheral artery disease. Eur. J. Vasc. Endovasc. Surg. 54 , 220–233 (2017).

Singh, N. et al. Preoperative hemoglobin A1c levels and increased risk of adverse limb events in diabetic patients undergoing infrainguinal lower extremity bypass surgery in the Vascular Quality Initiative. J. Vasc. Surg. 70 , 1225–1234 e1221 (2019).

Kremers, B. et al. Plasma biomarkers to predict cardiovascular outcome in patients with peripheral artery disease: a systematic review and meta-analysis. Arterioscler. Thromb. Vasc. Biol. 40 , 2018–2032 (2020).

Pastori, D. et al. Statins and major adverse limb events in patients with peripheral artery disease: a systematic review and meta-analysis. Thromb. Haemost. 120 , 866–875 (2020).

Kim, C., Yang, Y. S., Ryu, G. W. & Choi, M. Risk factors associated with amputation-free survival for patients with peripheral arterial disease: a systematic review. Eur. J. Cardiovasc. Nurs. 20 , 295–304 (2021).

Kreutzburg, T. et al. Editor’s Choice-the GermanVasc score: a pragmatic risk score predicts five year amputation free survival in patients with peripheral arterial occlusive disease. Eur. J. Vasc. Endovasc. Surg. 61 , 248–256 (2021).

Narula, N., Olin, J. W. & Narula, N. Pathologic disparities between peripheral artery disease and coronary artery disease. Arterioscler. Thromb. Vasc. Biol. 40 , 1982–1989 (2020).

Pandya, Y. K., Lowenkamp, M. N. & Chapman, S. C. Functional popliteal artery entrapment syndrome: a review of diagnostic and management approaches. Vasc. Med. 24 , 455–460 (2019).

Francois, C. J. Peripheral vascular imaging focusing on nonatherosclerotic disease. Radiol. Clin. North Am. 58 , 831–839 (2020).

Yahagi, K. et al. Pathophysiology of native coronary, vein graft, and in-stent atherosclerosis. Nat. Rev. Cardiol. 13 , 79–98 (2016).

Virmani, R., Kolodgie, F. D., Burke, A. P., Farb, A. & Schwartz, S. M. Lessons from sudden coronary death: a comprehensive morphological classification scheme for atherosclerotic lesions. Arterioscler. Thromb. Vasc. Biol. 20 , 1262–1275 (2000).

Fahed, A. C. & Jang, I. K. Plaque erosion and acute coronary syndromes: phenotype, molecular characteristics and future directions. Nat. Rev. Cardiol. 18 , 724–734 (2021).

Golledge, J., Greenhalgh, R. M. & Davies, A. H. The symptomatic carotid plaque. Stroke 31 , 774–781 (2000).

Narula, N. et al. Pathology of peripheral artery disease in patients with critical limb ischemia. J. Am. Coll. Cardiol. 72 , 2152–2163 (2018).

Krishna, S. M., Omer, S. M. & Golledge, J. Evaluation of the clinical relevance and limitations of current pre-clinical models of peripheral artery disease. Clin. Sci. 130 , 127–150 (2016).

Article   CAS   Google Scholar  

Krishna, S. M., Moxon, J. V. & Golledge, J. A review of the pathophysiology and potential biomarkers for peripheral artery disease. Int. J. Mol. Sci. 16 , 11294–11322 (2015).

McDermott, M. M. et al. Skeletal muscle pathology in peripheral artery disease: a brief review. Arterioscler. Thromb. Vasc. Biol. 40 , 2577–2585 (2020).

Annex, B. H. & Cooke, J. P. New directions in therapeutic angiogenesis and arteriogenesis in peripheral arterial disease. Circ. Res. 128 , 1944–1957 (2021).

Farber, A. & Eberhardt, R. T. The current state of critical limb ischemia: a systematic review. JAMA Surg. 151 , 1070–1077 (2016).

Venkatesh, B. A. et al. Baseline assessment and comparison of arterial anatomy, hyperemic flow, and skeletal muscle perfusion in peripheral artery disease: the Cardiovascular Cell Therapy Research Network “Patients with Intermittent Claudication Injected with ALDH Bright Cells” (CCTRN PACE) study. Am. Heart J. 183 , 24–34 (2017).

Tronc, F. et al. Role of matrix metalloproteinases in blood flow-induced arterial enlargement: interaction with NO. Arterioscler. Thromb. Vasc. Biol. 20 , E120–E126 (2000).

Hutchings, G. et al. Molecular mechanisms associated with ROS-dependent angiogenesis in lower extremity artery disease. Antioxidants 10 , 735 (2021).

Mercier, C., Rousseau, M. & Geraldes, P. Growth factor deregulation and emerging role of phosphatases in diabetic peripheral artery disease. Front. Cardiovasc. Med. 7 , 619612 (2020).

Beltran-Camacho, L., Rojas-Torres, M. & Duran-Ruiz, M. C. Current status of angiogenic cell therapy and related strategies applied in critical limb ischemia. Int. J. Mol. Sci. 22 , 2335 (2021).

Morris, D. R., Skalina, T. A., Singh, T. P., Moxon, J. V. & Golledge, J. Association of computed tomographic leg muscle characteristics with lower limb and cardiovascular events in patients with peripheral artery disease. J. Am. Heart Assoc. 7 , e009943 (2018).

McDermott, M. M. et al. Lower extremity ischemia, calf skeletal muscle characteristics, and functional impairment in peripheral arterial disease. J. Am. Geriatr. Soc. 55 , 400–406 (2007).

Baum, O. et al. Capillary ultrastructure and mitochondrial volume density in skeletal muscle in relation to reduced exercise capacity of patients with intermittent claudication. Am. J. Physiol. Regul. Integr. Comp. Physiol. 310 , R943–R951 (2016).

Duscha, B. D. et al. Skeletal muscle capillary density is related to anaerobic threshold and claudication in peripheral artery disease. Vasc. Med. 25 , 411–418 (2020).

Ho, T. K., Rajkumar, V., Black, C. M., Abraham, D. J. & Baker, D. M. Increased angiogenic response but deficient arteriolization and abnormal microvessel ultrastructure in critical leg ischaemia. Br. J. Surg. 93 , 1368–1376 (2006).

White, S. H. et al. Walking performance is positively correlated to calf muscle fiber size in peripheral artery disease subjects, but fibers show aberrant mitophagy: an observational study. J. Transl. Med. 14 , 284 (2016).

Article   PubMed   PubMed Central   CAS   Google Scholar  

Anderson, C. P., Pekas, E. J. & Park, S. Y. Microvascular dysfunction in peripheral artery disease: is heat therapy a viable treatment? Int. J. Env. Res. Public Health 18 , 2384 (2021).

Behroozian, A. & Beckman, J. A. Microvascular disease increases amputation in patients with peripheral artery disease. Arterioscler. Thromb. Vasc. Biol. 40 , 534–540 (2020).

Grenon, S. M. et al. Walking disability in patients with peripheral artery disease is associated with arterial endothelial function. J. Vasc. Surg. 59 , 1025–1034 (2014).

Treat-Jacobson, D. et al. Implementation of supervised exercise therapy for patients with symptomatic peripheral artery disease: a science advisory from the American Heart Association. Circulation 140 , e700–e710 (2019).

Yang, C. et al. Retinal microvascular findings and risk of incident peripheral artery disease: an analysis from the Atherosclerosis Risk in Communities (ARIC) study. Atherosclerosis 294 , 62–71 (2020).

Krishna, S. M. et al. Development of a two-stage limb ischemia model to better simulate human peripheral artery disease. Sci. Rep. 10 , 3449 (2020).

Lane, R., Harwood, A., Watson, L. & Leng, G. C. Exercise for intermittent claudication. Cochrane Database Syst. Rev. 12 , CD000990 (2017).

Parikh, P. P. et al. A reliable mouse model of hind limb gangrene. Ann. Vasc. Surg. 48 , 222–232 (2018).

Gomes de Almeida Schirmer, B. et al. The NO-donor MPC-1011 stimulates angiogenesis and arteriogenesis and improves hindlimb ischemia via a cGMP-dependent pathway involving VEGF and SDF-1alpha. Atherosclerosis 304 , 30–38 (2020).

Yao, Z. et al. Bone marrow mesenchymal stem cell-derived endothelial cells increase capillary density and accelerate angiogenesis in mouse hindlimb ischemia model. Stem Cell Res. Ther. 11 , 221 (2020).

Cui, Y. et al. N-acetylcysteine differentially regulates the populations of bone marrow and circulating endothelial progenitor cells in mice with limb ischemia. Eur. J. Pharmacol. 881 , 173233 (2020).

Gotze, A. M. et al. IL10 alters peri-collateral macrophage polarization and hind-limb reperfusion in mice after femoral artery ligation. Int. J. Mol. Sci. 21 , 2821 (2020).

Article   PubMed Central   CAS   Google Scholar  

Hollander, M. R. et al. Stimulation of collateral vessel growth by inhibition of galectin 2 in mice using a single-domain llama-derived antibody. J. Am. Heart Assoc. 8 , e012806 (2019).

O’Neill, K. M. et al. NOX4 is a major regulator of cord blood-derived endothelial colony-forming cells which promotes post-ischaemic revascularization. Cardiovasc. Res. 116 , 393–405 (2020).

Ryan, T. E. et al. PFKFB3-mediated glycolysis rescues myopathic outcomes in the ischemic limb. JCI Insight 5 , e139628 (2020).

Article   PubMed Central   Google Scholar  

Orfany, A. et al. Mitochondrial transplantation ameliorates acute limb ischemia. J. Vasc. Surg. 71 , 1014–1026 (2020).

Troidl, K. et al. The lipopeptide MALP-2 promotes collateral growth. Cells 9 , 997 (2020).

Article   CAS   PubMed Central   Google Scholar  

Golledge, J., Ward, N. C. & Watts, G. F. Lipid management in people with peripheral artery disease. Curr. Opin. Lipidol. 30 , 470–476 (2019).

MacKeigan, D. T. et al. Updated understanding of platelets in thrombosis and hemostasis: the roles of integrin psi domains and their potential as therapeutic targets. Cardiovasc. Hematol. Disord. Drug Targets 20 , 260–273 (2020).

Montarello, N. J., Nguyen, M. T., Wong, D. T. L., Nicholls, S. J. & Psaltis, P. J. Inflammation in coronary atherosclerosis and its therapeutic implications. Cardiovasc. Drugs Ther. https://doi.org/10.1007/s10557-020-07106-6 (2020).

Singh, A., Tandon, S. & Tandon, C. An update on vascular calcification and potential therapeutics. Mol. Biol. Rep. 48 , 887–896 (2021).

Deppen, J. N. et al. A Swine hind limb ischemia model useful for testing peripheral artery disease therapeutics. J. Cardiovasc Transl. Res https://doi.org/10.1007/s12265-021-10134-8 (2021).

Hashimoto, A., Miyakoda, G., Hirose, Y. & Mori, T. Activation of endothelial nitric oxide synthase by cilostazol via a cAMP/protein kinase A- and phosphatidylinositol 3-kinase/Akt-dependent mechanism. Atherosclerosis 189 , 350–357 (2006).

Kherallah, R. Y., Khawaja, M., Olson, M., Angiolillo, D. & Birnbaum, Y. Cilostazol: a review of basic mechanisms and clinical uses. Cardiovasc. Drugs Ther. https://doi.org/10.1007/s10557-021-07187-x (2021).

Brown, T. et al. Cilostazol for intermittent claudication. Cochrane Database Syst. Rev. 6 , CD003748 (2021).

Desai, K., Han, B., Kuziez, L., Yan, Y. & Zayed, M. A. Literature review and meta-analysis of the efficacy of cilostazol on limb salvage rates after infrainguinal endovascular and open revascularization. J. Vasc. Surg. 73 , 711–721 e713 (2021).

Thanigaimani, S. et al. Network meta-analysis comparing the outcomes of treatments for intermittent claudication tested in randomized controlled trials. J. Am. Heart Assoc. 10 , e019672 (2021).

Castellsague, J. et al. Characterization of new users of cilostazol in the UK, Spain, Sweden, and Germany. Pharmacoepidemiol. Drug. Saf. 26 , 615–624 (2017).

Castaneda, P. R. et al. Outcomes and safety of electronic consult use in vascular surgery. J. Vasc. Surg. 71 , 1726–1732 (2020).

Golledge, J. et al. Risk of major amputation in patients with intermittent claudication undergoing early revascularization. Br. J. Surg. 105 , 699–708 (2018).

Polonsky, T. S. & McDermott, M. M. Lower extremity peripheral artery disease without chronic limb-threatening ischemia: a review. JAMA 325 , 2188–2198 (2021).

McDermott, M. M. et al. Walking exercise therapy effects on lower extremity skeletal muscle in peripheral artery disease. Circ. Res. 128 , 1851–1867 (2021).

Biswas, M. P. et al. Exercise training and revascularization in the management of symptomatic peripheral artery disease. JACC Basic Transl. Sci. 6 , 174–188 (2021).

Treat-Jacobson, D. et al. Optimal exercise programs for patients with peripheral artery disease: a scientific statement from the American Heart Association. Circulation 139 , e10–e33 (2019).

Parmenter, B. J., Mavros, Y., Ritti Dias, R., King, S. & Fiatarone Singh, M. Resistance training as a treatment for older persons with peripheral artery disease: a systematic review and meta-analysis. Br. J. Sports Med. 54 , 452–461 (2020).

Duscha, B. D. et al. Angiogenesis in skeletal muscle precede improvements in peak oxygen uptake in peripheral artery disease patients. Arterioscler. Thromb. Vasc. Biol. 31 , 2742–2748 (2011).

Allen, J. D. et al. Plasma nitrite flux predicts exercise performance in peripheral arterial disease after 3 months of exercise training. Free Radic. Biol. Med. 49 , 1138–1144 (2010).

Woessner, M. et al. Beet the best? Circ. Res. 123 , 654–659 (2018).

Kenjale, A. A. et al. Dietary nitrate supplementation enhances exercise performance in peripheral arterial disease. J. Appl. Physiol. 110 , 1582–1591 (2011).

Harwood, A. E., Smith, G. E., Cayton, T., Broadbent, E. & Chetter, I. C. A systematic review of the uptake and adherence rates to supervised exercise programs in patients with intermittent claudication. Ann. Vasc. Surg. 34 , 280–289 (2016).

Golledge, J. et al. Meta-analysis of clinical trials examining the benefit of structured home exercise in patients with peripheral artery disease. Br. J. Surg. 106 , 319–331 (2019).

Pymer, S. A. et al. An updated systematic review and meta-analysis of home-based exercise programmes for individuals with intermittent claudication. J. Vasc. Surg. 74 , 2076–2085.e20 (2021).

McDermott, M. M. et al. Home-based walking exercise intervention in peripheral artery disease: a randomized clinical trial. JAMA 310 , 57–65 (2013).

McDermott, M. M. et al. Effect of a home-based exercise intervention of wearable technology and telephone coaching on walking performance in peripheral artery disease: the HONOR randomized clinical trial. JAMA 319 , 1665–1676 (2018).

McDermott, M. M. et al. Effect of low-intensity vs high-intensity home-based walking exercise on walk distance in patients with peripheral artery disease: the LITE randomized clinical trial. JAMA 325 , 1266–1276 (2021).

Saratzis, A. et al. Supervised exercise therapy and revascularization for intermittent claudication: network meta-analysis of randomized controlled trials. JACC Cardiovasc. Interv. 12 , 1125–1136 (2019).

Pandey, A. et al. Comparative efficacy of endovascular revascularization versus supervised exercise training in patients with intermittent claudication: meta-analysis of randomized controlled trials. JACC Cardiovasc. Interv. 10 , 712–724 (2017).

Jansen, S. C. P. et al. Successful implementation of the exercise first approach for intermittent claudication in the Netherlands is associated with few lower limb revascularisations. Eur. J. Vasc. Endovasc. Surg. 60 , 881–887 (2020).

Katsanos, K., Spiliopoulos, S., Kitrou, P., Krokidis, M. & Karnabatidis, D. Risk of death following application of paclitaxel-coated balloons and stents in the femoropopliteal artery of the leg: a systematic review and meta-analysis of randomized controlled trials. J. Am. Heart Assoc. 7 , e011245 (2018).

Dan, K. et al. Paclitaxel-related balloons and stents for the treatment of peripheral artery disease: insights from the Food and Drug Administration 2019 Circulatory System Devices Panel Meeting on late mortality. Am. Heart J. 222 , 112–120 (2020).

Dinh, K. et al. Mortality rates after paclitaxel-coated device use in patients with occlusive femoropopliteal disease: an updated systematic review and meta-analysis of randomized controlled trials. J. Endovasc. Ther. 28 , 755–777 (2021).

Hiramoto, J. S., Teraa, M., de Borst, G. J. & Conte, M. S. Interventions for lower extremity peripheral artery disease. Nat. Rev. Cardiol. 15 , 332–350 (2018).

Adam, D. J. et al. Bypass versus angioplasty in severe ischaemia of the leg (BASIL): multicentre, randomised controlled trial. Lancet 366 , 1925–1934 (2005).

International Standard Randomised Controlled Trial Number Registry. ISRCTN https://www.isrctn.com/ISRCTN27728689 (2020).

International Standard Randomised Controlled Trial Number Registry. ISRCTN https://www.isrctn.com/ISRCTN14469736 (2019).

US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02060630 (2021).

Schwartz, G. G. et al. Alirocumab and cardiovascular outcomes after acute coronary syndrome. N. Engl. J. Med. 379 , 2097–2107 (2018).

Jukema, J. W. et al. Alirocumab in patients with polyvascular disease and recent acute coronary syndrome: ODYSSEY OUTCOMES trial. J. Am. Coll. Cardiol. 74 , 1167–1176 (2019).

Ishikawa, Y. et al. Preventive effects of eicosapentaenoic acid on coronary artery disease in patients with peripheral artery disease. Circ. J. 74 , 1451–1457 (2010).

Bhatt, D. L. et al. Cardiovascular risk reduction with icosapent ethyl for hypertriglyceridemia. N. Engl. J. Med. 380 , 11–22 (2019).

Ostergren, J. et al. Impact of ramipril in patients with evidence of clinical or subclinical peripheral arterial disease. Eur. Heart J. 25 , 17–24 (2004).

SPRINT Research Group. A randomized trial of intensive versus standard blood-pressure control. N. Engl. J. Med. 373 , 2103–2116 (2015).

ACCORD Study Group. Effects of intensive blood-pressure control in type 2 diabetes mellitus. N. Engl. J. Med. 362 , 1575–1585 (2010).

Goldman, M. P. et al. Effect of intensive glycemic control on risk of lower extremity amputation. J. Am. Coll. Surg. 227 , 596–604 (2018).

Verma, S. et al. Cardiovascular outcomes and safety of empagliflozin in patients with type 2 diabetes mellitus and peripheral artery disease: a subanalysis of EMPA-REG OUTCOME. Circulation 137 , 405–407 (2018).

Neal, B., Perkovic, V. & Matthews, D. R. Canagliflozin and cardiovascular and renal events in type 2 diabetes. N. Engl. J. Med. 377 , 644–657 (2017).

Cannon, C. P. et al. Cardiovascular outcomes with ertugliflozin in type 2 diabetes. N. Engl. J. Med. 383 , 1425–1435 (2020).

Bonaca, M. P. et al. Dapagliflozin and cardiac, kidney, and limb outcomes in patients with and without peripheral artery disease in DECLARE-TIMI 58. Circulation 142 , 734–747 (2020).

Wiviott, S. D. et al. Dapagliflozin and cardiovascular outcomes in type 2 diabetes. N. Engl. J. Med. 380 , 347–357 (2019).

CAPRIE Steering Committee. A randomised, blinded, trial of clopidogrel versus aspirin in patients at risk of ischaemic events (CAPRIE). Lancet 348 , 1329–1339 (1996).

Bonaca, M. P. et al. Rivaroxaban in peripheral artery disease after revascularization. N. Engl. J. Med. 382 , 1994–2004 (2020).

Hennrikus, D. et al. Effectiveness of a smoking cessation program for peripheral artery disease patients: a randomized controlled trial. J. Am. Coll. Cardiol. 56 , 2105–2112 (2010).

Thomas Manapurathe, D. et al. Cohort study examining the association between blood pressure and cardiovascular events in patients with peripheral artery disease. J. Am. Heart Assoc. 8 , e010748 (2019).

Golledge, J., Quigley, F., Velu, R., Walker, P. J. & Moxon, J. V. Association of impaired fasting glucose, diabetes and their management with the presentation and outcome of peripheral artery disease: a cohort study. Cardiovasc. Diabetol. 13 , 147 (2014).

Machline-Carrion, M. J. et al. Effect of a multifaceted quality improvement intervention on the prescription of evidence-based treatment in patients at high cardiovascular risk in Brazil: the BRIDGE cardiovascular prevention cluster randomized clinical trial. JAMA Cardiol. 4 , 408–417 (2019).

Vernooij, J. W. et al. Internet based vascular risk factor management for patients with clinically manifest vascular disease: randomised controlled trial. BMJ 344 , e3750 (2012).

Peterson, G. G. et al. Effect of the million hearts cardiovascular disease risk reduction model on initiating and intensifying medications: a prespecified secondary analysis of a randomized clinical trial. JAMA Cardiol. 6 , 1050–1059 (2021).

Hong, F. F. et al. Roles of eNOS in atherosclerosis treatment. Inflamm. Res. 68 , 429–441 (2019).

Ismaeel, A. et al. The nitric oxide system in peripheral artery disease: connection with oxidative stress and biopterins. Antioxidants 9 , 590 (2020).

Allen, J. D., Giordano, T. & Kevil, C. G. Nitrite and nitric oxide metabolism in peripheral artery disease. Nitric Oxide 26 , 217–222 (2012).

Loffredo, L. et al. Imbalance between nitric oxide generation and oxidative stress in patients with peripheral arterial disease: effect of an antioxidant treatment. J. Vasc. Surg. 44 , 525–530 (2006).

Loffredo, L. et al. Dark chocolate acutely improves walking autonomy in patients with peripheral artery disease. J. Am. Heart Assoc. 3 , e001072 (2014).

McDermott, M. M. et al. Cocoa to improve walking performance in older people with peripheral artery disease: the COCOA-PAD pilot randomized clinical trial. Circ. Res. 126 , 589–599 (2020).

Park, S. Y. et al. Acute mitochondrial antioxidant intake improves endothelial function, antioxidant enzyme activity, and exercise tolerance in patients with peripheral artery disease. Am. J. Physiol. Heart Circ. Physiol. 319 , H456–H467 (2020).

Zankl, A. R. et al. Telmisartan improves absolute walking distance and endothelial function in patients with peripheral artery disease. Clin. Res. Cardiol. 99 , 787–794 (2010).

Montanari, G. et al. Treatment with low dose metformin in patients with peripheral vascular disease. Pharmacol. Res. 25 , 63–73 (1992).

Sirtori, C. R. et al. Metformin improves peripheral vascular flow in nonhyperlipidemic patients with arterial disease. J. Cardiovasc. Pharmacol. 6 , 914–923 (1984).

McDermott, M. M. et al. Effect of resveratrol on walking performance in older people with peripheral artery disease: the RESTORE randomized clinical trial. JAMA Cardiol. 2 , 902–907 (2017).

Gao, W., Chen, D., Liu, G. & Ran, X. Autologous stem cell therapy for peripheral arterial disease: a systematic review and meta-analysis of randomized controlled trials. Stem Cell Res. Ther. 10 , 140 (2019).

McDermott, M. M. et al. Effect of granulocyte-macrophage colony-stimulating factor with or without supervised exercise on walking performance in patients with peripheral artery disease: the PROPEL randomized clinical trial. JAMA 318 , 2089–2098 (2017).

McDermott, M. M. et al. Racial differences in the effect of granulocyte macrophage colony-stimulating factor on improved walking distance in peripheral artery disease: the PROPEL randomized clinical trial. J. Am. Heart Assoc. 8 , e011001 (2019).

Gu, Y. et al. A randomized, double-blind, placebo-controlled phase II study of hepatocyte growth factor in the treatment of critical limb ischemia. Mol. Ther. 27 , 2158–2165 (2019).

Hammad, T. A. et al. Stromal cell-derived factor-1 plasmid treatment for patients with peripheral artery disease (STOP-PAD) trial: six-month results. J. Endovasc. Ther. 27 , 669–675 (2020).

Gorenoi, V., Brehm, M. U., Koch, A. & Hagen, A. Growth factors for angiogenesis in peripheral arterial disease. Cochrane Database Syst. Rev. 6 , CD011741 (2017).

Forster, R., Liew, A., Bhattacharya, V., Shaw, J. & Stansby, G. Gene therapy for peripheral arterial disease. Cochrane Database Syst. Rev. 10 , CD012058 (2018).

Viney, N. J. et al. Antisense oligonucleotides targeting apolipoprotein(a) in people with raised lipoprotein(a): two randomised, double-blind, placebo-controlled, dose-ranging trials. Lancet 388 , 2239–2253 (2016).

Everett, B. M. et al. Inhibition of interleukin-1beta and reduction in atherothrombotic cardiovascular events in the CANTOS trial. J. Am. Coll. Cardiol. 76 , 1660–1670 (2020).

Ridker, P. M. & Rane, M. Interleukin-6 signaling and anti-interleukin-6 therapeutics in cardiovascular disease. Circ. Res. 128 , 1728–1746 (2021).

Antonopoulos, A. S., Papanikolaou, E., Vogiatzi, G., Oikonomou, E. & Tousoulis, D. Anti-inflammatory agents in peripheral arterial disease. Curr. Opin. Pharmacol. 39 , 1–8 (2018).

Parvar, S. L., Thiyagarajah, A., Nerlekar, N., King, P. & Nicholls, S. J. A systematic review and meta-analysis of gender differences in long-term mortality and cardiovascular events in peripheral artery disease. J. Vasc. Surg. 73 , 1456–1465 (2021).

Download references

Acknowledgements

J.G. received grants from the National Health and Medical Research Council (1180736), Heart Foundation, Tropical Australian Academic Health Centre, Townsville Hospital and Health Service Study, Education and Research Trust Fund and Queensland Government. J.G. holds a Practitioner Fellowship from the National Health and Medical Research Council (1117601) and a Senior Clinical Research Fellowship from the Queensland Government. The author thanks S. Thangaimani, J. Phie and C. Burrows (James Cook University, Australia) for help with production of the figures for initial submission and A. Golledge for help with proof reading. He thanks all the past and current researchers from the Queensland Research Centre for Peripheral Vascular Disease and collaborators for their ongoing research on peripheral artery disease, which has contributed towards the insights included in this Review. Finally, the author apologizes to scientists whose research could not be included in this Review owing to space limitations.

Author information

Authors and affiliations.

Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Queensland, Australia

Jonathan Golledge

The Department of Vascular and Endovascular Surgery, Townsville University Hospital, Townsville, Queensland, Australia

The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland, Australia

You can also search for this author in PubMed   Google Scholar

Corresponding author

Correspondence to Jonathan Golledge .

Ethics declarations

Competing interests.

The author has received honoraria from Amgen and Bayer for giving lectures on PAD.

Additional information

Peer review information.

Nature Reviews Cardiology thanks Brian H. Annex; Iris Baumgartner, who co-reviewed with Dario Häberli; Mary Kavurma; and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Minimally invasive interventional (such as stenting) or open surgical (such as endarterectomy) procedures to improve distal blood supply.

Muscle damage with various causes, including impaired muscle blood supply owing to peripheral artery disease.

The expansion of existing collateral arteries to improve distal blood supply.

The sprouting of new capillaries and formation of new networks of small vessels to improve distal blood supply.

Administering stem or progenitor cells to encourage angiogenesis.

Administering harmless viruses that have been modified to carry a gene of interest into the local tissue.

Rights and permissions

Reprints and permissions

About this article

Cite this article.

Golledge, J. Update on the pathophysiology and medical treatment of peripheral artery disease. Nat Rev Cardiol 19 , 456–474 (2022). https://doi.org/10.1038/s41569-021-00663-9

Download citation

Accepted : 07 December 2021

Published : 07 January 2022

Issue Date : July 2022

DOI : https://doi.org/10.1038/s41569-021-00663-9

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

This article is cited by

Association of lower extremity peripheral arterial disease with quantitative muscle features from computed tomography angiography.

  • Yuexin Chen
  • Zhengyu Jin

Insights into Imaging (2024)

Promoting collateral formation in type 2 diabetes mellitus using ultra-small nanodots with autophagy activation and ROS scavenging

  • Yixuan Wang

Journal of Nanobiotechnology (2024)

Revaskularisation beim diabetischen Fußsyndrom? Wann und wie?

  • Ulrich Rother
  • Hartmut Görtz
  • Christian-Alexander Behrendt

Gefässchirurgie (2024)

Primary results of the brazilian registry of atherothrombotic disease (NEAT)

  • Pedro G. M. de Barros e Silva
  • Charlene Troiani do Nascimento
  • Mauricio Antonio Pompilio

Scientific Reports (2024)

ETV2/ER71, the key factor leading the paths to vascular regeneration and angiogenic reprogramming

  • Tae Min Kim
  • Changwon Park

Stem Cell Research & Therapy (2023)

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

peripheral vascular disease research paper

Information

  • Author Services

Initiatives

You are accessing a machine-readable page. In order to be human-readable, please install an RSS reader.

All articles published by MDPI are made immediately available worldwide under an open access license. No special permission is required to reuse all or part of the article published by MDPI, including figures and tables. For articles published under an open access Creative Common CC BY license, any part of the article may be reused without permission provided that the original article is clearly cited. For more information, please refer to https://www.mdpi.com/openaccess .

Feature papers represent the most advanced research with significant potential for high impact in the field. A Feature Paper should be a substantial original Article that involves several techniques or approaches, provides an outlook for future research directions and describes possible research applications.

Feature papers are submitted upon individual invitation or recommendation by the scientific editors and must receive positive feedback from the reviewers.

Editor’s Choice articles are based on recommendations by the scientific editors of MDPI journals from around the world. Editors select a small number of articles recently published in the journal that they believe will be particularly interesting to readers, or important in the respective research area. The aim is to provide a snapshot of some of the most exciting work published in the various research areas of the journal.

Original Submission Date Received: .

  • Active Journals
  • Find a Journal
  • Proceedings Series
  • For Authors
  • For Reviewers
  • For Editors
  • For Librarians
  • For Publishers
  • For Societies
  • For Conference Organizers
  • Open Access Policy
  • Institutional Open Access Program
  • Special Issues Guidelines
  • Editorial Process
  • Research and Publication Ethics
  • Article Processing Charges
  • Testimonials
  • Preprints.org
  • SciProfiles
  • Encyclopedia

ijms-logo

Article Menu

  • Subscribe SciFeed
  • Recommended Articles
  • PubMed/Medline
  • Google Scholar
  • on Google Scholar
  • Table of Contents

Find support for a specific problem in the support section of our website.

Please let us know what you think of our products and services.

Visit our dedicated information section to learn more about MDPI.

JSmol Viewer

Pathophysiology of peripheral arterial disease (pad): a review on oxidative disorders.

peripheral vascular disease research paper

1. Methodology of Literature Search for Review

1.1. data sources and search, 1.2. data extraction, 2. introduction on topic, 3. biochemistry of the physical exercise: pro and anti oxidative effects, 4. oxidative stress and physical exercise in patients with peripheral arterial disease, 5. antioxidants, and heme oxygenase 1 in peripheral artery disease, 6. discussion, 7. conclusions, author contributions, conflicts of interest.

  • Fowkes, F.G.; Rudan, D.; Rudan, I.; Aboyans, V.; Denenberg, J.O.; McDermott, M.M.; Norman, P.E.; Sampson, U.K.; Williams, L.J.; Mensah, G.A.; et al. Comparison of global estimates of prevalence and risk factors for peripheral artery disease in 2000 and 2010: A systematic review and analysis. Lancet 2013 , 382 , 1329–1340. [ Google Scholar ] [ CrossRef ]
  • Aronow, H.; Hiatt, W.R. The burden of peripheral artery disease and the role of antiplatelet therapy. Postgrad. Med. 2009 , 121 , 123–135. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Shu, J.; Santulli, G. Update on peripheral artery disease: Epidemiology and evidence-based facts. Atherosclerosis 2018 , 275 , 379–381. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Signorelli, S.; Anzaldi, M.; Fiore, V.; Catanzaro, S.; Simili, M.; Torrisi, B.; Neri, S. Study on unrecognized peripheral arterial disease (PAD) by ankle/brachial index and arterial co-morbidity in Catania (Sicily, Italy). Angiology 2010 , 61 , 524–529. [ Google Scholar ] [ CrossRef ]
  • Herrington, W.; Lacey, B.; Sherliker, P.; Armitage, J.; Lewington, S. Epidemiology of Atherosclerosis and the Potential to Reduce the Global Burden of Atherothrombotic Disease. Circ. Res. 2016 , 118 , 535–546. [ Google Scholar ] [ CrossRef ]
  • Signorelli, S.S.; Katsiki, N. Oxidative Stress and Inflammation: Their Role in the Pathogenesis of Peripheral Artery Disease with or Without Type 2 Diabetes Mellitus. Curr. Vasc. Pharmacol. 2018 , 16 , 547–554. [ Google Scholar ] [ CrossRef ]
  • Leung, F.P.; Yung, L.M.; Laher, I.; Yao, X.; Chen, Z.Y.; Huang, Y. Exercise, vascular wall and cardiovascular diseases: An update. Sports Med. 2008 , 38 , 1009–1024. [ Google Scholar ] [ CrossRef ]
  • López-Cruz, R.I.; Zenteno-Savín, T.; Galván-Magaña, F. Superoxide production, oxidative damage and enzymatic antioxidant defenses in shark skeletal muscle. Comp. Biochem. Physiol. A Mol. Integr. Physiol. 2010 , 156 , 50–56. [ Google Scholar ] [ CrossRef ]
  • Lambertucci, R.H.; Levada-Pires, A.C.; Rossoni, L.V.; Curi, R.; Pithon-Curi, T.C. Effects of aerobic exercise training on antioxidant enzyme activities and mRNA levels in soleus muscle from young and aged rats. Mech. Ageing Dev. 2007 , 128 , 267–275. [ Google Scholar ] [ CrossRef ]
  • Di Raimondo, D.; Musiari, G.; Miceli, G.; Arnao, V.; Pinto, A. Preventive and Therapeutic Role of Muscle Contraction against Chronic Diseases. Curr. Pharm. Des. 2016 , 22 , 4686–4699. [ Google Scholar ] [ CrossRef ]
  • Steven, S.; Daiber, A.; Dopheide, J.F.; Münzel, T.; Espinola-Klein, C. Peripheral artery disease, redox signaling, oxidative stress—Basic and clinical aspects. Redox Biol. 2017 , 12 , 787–797. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Walker, M.A.; Hoier, B.; Walker, P.J.; Schulze, K.; Bangsbo, J.; Hellsten, Y.; Askew, C.D. Vasoactive enzymes and blood flow responses to passive and active exercise in peripheral arterial disease. Atherosclerosis 2016 , 246 , 98–105. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Gliemann, L.; Nyberg, M.; Hellsten, Y. Nitric oxide and reactive oxygen species in limb vascular function: What is the effect of physical activity? Free Radic. Res. 2014 , 48 , 71–83. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Barker, G.A.; Green, S.; Green, A.A.; Walker, P.J. Walking performance, oxygen uptake kinetics and resting muscle pyruvate dehydrogenase complex activity in peripheral arterial disease. Clin. Sci. 2004 , 106 , 241–249. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Wang, H.; Hiatt, W.R.; Barstow, T.J.; Brass, E.P. Relationships between muscle mitochondrial DNA content, mitochondrial enzyme activity and oxidative capacity in man: Alterations with disease. Eur. J. Appl. Physiol. Occup. Physiol. 1999 , 80 , 22–27. [ Google Scholar ] [ CrossRef ]
  • Brass, E.P. Skeletal muscle metabolism as a target for drug therapy in peripheral arterial disease. Vasc. Med. 1996 , 1 , 55–59. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Drummond, G.R.; Selemidis, S.; Griendling, K.K.; Sobey, C.G. Combating oxidative stress in vascular disease: NADPH oxidases as therapeutic targets. Nat. Rev. Drug Discov. 2011 , 10 , 453–471. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Semenza, G.L. Vasculogenesis, angiogenesis, and arteriogenesis: Mechanisms of blood vessel formation and remodeling. J. Cell Biochem. 2007 , 102 , 840–847. [ Google Scholar ] [ CrossRef ]
  • Pedersen, B.K. Anti-inflammatory effects of exercise: Role in diabetes and cardiovascular disease. Eur. J. Clin. Investig. 2017 , 47 , 600–611. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Di Raimondo, D.; Tuttolomondo, A.; Musiari, G.; Schimmenti, C.; D’Angelo, A.; Pinto, A. Are the Myokines the Mediators of Physical Activity-Induced Health Benefits? Curr. Pharm. Des. 2016 , 22 , 3622–3647. [ Google Scholar ] [ CrossRef ]
  • Di Raimondo, D.; Miceli, G.; Musiari, G.; Tuttolomondo, A.; Pinto, A. New insights about the putative role of myokines in the context of cardiac rehabilitation and secondary cardiovascular prevention. Ann. Transl. Med. 2017 , 5 , 300. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Nielsen, S.; Pedersen, B.K. Skeletal muscle as an immunogenic organ. Curr. Opin. Pharmacol. 2008 , 8 , 346–351. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Di Raimondo, D. Editorial (Thematic Issue: Myokines and Exercise Training: More Shadows than Lights). Curr. Pharm. Des. 2016 , 22 , 3619–3621. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Finkler, M.; Lichtenberg, D.; Pinchuk, I. The relationship between oxidative stress and exercise. J. Basic Clin. Physiol. Pharmacol. 2014 , 25 , 1–11. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Bloomer, R.J.; Goldfarb, A.H.; Wideman, L.; McKenzie, M.J.; Consitt, L.A. Effects of acute aerobic and anaerobic exercise on blood markers of oxidative stress. J. Strength Cond. Res. 2005 , 19 , 276–285. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Gomez-Cabrera, M.C.; Domenech, E.; Viña, J. Moderate exercise is an antioxidant: Upregulation of antioxidant genes by training. Free Radic. Biol. Med. 2008 , 15 , 126–131. [ Google Scholar ] [ CrossRef ]
  • Radak, Z.; Chung, H.Y.; Goto, S. Systemic adaptation to oxidative challenge induced by regular exercise. Free Radic. Biol. Med. 2008 , 44 , 153–159. [ Google Scholar ] [ CrossRef ]
  • McDermott, M.M. Lower extremity manifestations of peripheral artery disease: The pathophysiologic and functional implications of leg ischemia. Circ. Res. 2015 , 116 , 1540–1550. [ Google Scholar ] [ CrossRef ]
  • McDermott, M.M.; Liu, K.; Greenland, P.; Guralnik, J.M.; Criqui, M.H.; Chan, C.; Chan, C.; Pearce, W.H.; Schneider, J.R.; Ferrucci, L.; et al. Functional decline in peripheral arterial disease: Associations with the ankle brachial index and legsymptoms. JAMA 2004 , 292 , 453–461. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Kiani, S.; Aasen, J.G.; Holbrook, M.; Khemka, A.; Sharmeen, F.; LeLeiko, R.M.; Tabit, C.E.; Farber, A.; Eberhardt, R.T.; Gokce, N.; et al. Peripheral artery disease is associated with severe impairment of vascular function. Vasc. Med. 2013 , 18 , 72–78. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • de Silva, R.C.; Wolosker, N.; Yugar-Toledo, J.C.; Consolim-Colombo, F.M. Vascular reactivity is impaired and associated with walking ability in patients with intermittent claudication. Angiology 2015 , 66 , 680–686. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • McDermott, M.M.; Greenland, P.; Liu, K.; Guralnik, J.M.; Criqui, M.H.; Dolan, N.C.; Chan, C.; Celic, L.; Pearce, W.H.; Schneider, J.R.; et al. Leg symptoms in peripheral arterial disease: Associated clinical characteristics and functional impairment. JAMA 2001 , 286 , 1599–1606. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Ross, R. Atherosclerosis-an inflammatory disease. N. Engl. J. Med. 1999 , 340 , 115–126. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Brevetti, G.; Schiano, V.; Chiariello, M. Endothelial dysfunction: A key to the pathophysiology and natural history of peripheral arterial disease? Atherosclerosis 2008 , 197 , 1–11. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Ridker, P.M.; Cushman, M.; Stampfer, M.J.; Tracy, R.P.; Hennekens, C.H. Plasma concentration of C-reactive protein and risk of developing peripheral vascular disease. Circulation 1998 , 97 , 425–428. [ Google Scholar ] [ CrossRef ]
  • Tzoulaki, I.; Murray, G.D.; Lee, A.J.; Rumley, A.; Lowe, G.D.; Fowkes, F.G. Inflammatory, haemostatic, and rheological markers for incident peripheral arterial disease: Edinburgh Artery Study. Eur. Heart J. 2007 , 28 , 354–362. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Lin, C.W.; Hsu, L.A.; Chen, C.C.; Yeh, J.T.; Sun, J.H.; Lin, C.H.; Chen, S.T.; Hsu, B.R.; Huang, Y.Y. C-reactive protein as an outcome predictor for percutaneous transluminal angioplasty in diabetic patients with peripheral arterial disease and infected foot ulcers. Diabetes Res. Clin. Pract. 2010 , 90 , 167–172. [ Google Scholar ] [ CrossRef ]
  • Jialal, I.; Verma, S.; Devaraj, S. Inhibition of endothelial nitric oxide synthase by C-reactive protein: Clinical relevance. Clin. Chem. 2009 , 55 , 206–208. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Pipinos, I.I.; Judge, A.R.; Selsby, J.T.; Zhu, Z.; Swanson, S.A.; Nella, A.A.; Dodd, S.L. The myopathy of peripheral arterial occlusive disease: Part 1. Functional and histomorphological changes and evidence for mitochondrial dysfunction. Vasc. Endovasc. Surg. 2007 , 41 , 481–489. [ Google Scholar ] [ CrossRef ]
  • Koutakis, P.; Weiss, D.J.; Miserlis, D.; Shostrom, V.K.; Papoutsi, E.; Ha, D.M.; Ha, D.M.; Carpenter, L.A.; McComb, R.D.; Casale, G.P.; et al. Oxidative damage in the gastrocnemius of patients with peripheral artery disease is myofiber type selective. Redox Biol. 2014 , 2 , 921–928. [ Google Scholar ]
  • Thompson, J.R.; Swanson, S.A.; Haynatzki, G.; Koutakis, P.; Johanning, J.M.; Reppert, P.R.; Papoutsi, E.; Miserlis, D.; Zhu, Z.; Casale, G.P.; et al. Protein Concentration and Mitochondrial Content in the Gastrocnemius Predicts Mortality Rates in Patients With Peripheral Arterial Disease. Ann. Surg. 2015 , 261 , 605–610. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Pipinos, I.I.; Judge, A.R.; Zhu, Z.; Selsby, J.T.; Swanson, S.A.; Johanning, J.M.; Baxter, B.T.; Lynch, G.T.; Dodd, S.L. Mitochondrial defects and oxidative damage in patients with peripheral arterial disease. Free Radic. Biol. Med. 2006 , 41 , 262–269. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Hiatt, W.R.; Wolfel, E.E.; Regensteiner, J.G.; Brass, E.P. Skeletal muscle carnitine metabolism in patients with unilateral peripheral arterial disease. J. Appl. Physiol. 1992 , 73 , 346–353. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Pipinos, I.I.; Sharov, V.G.; Shepard, A.D.; Anagnostopoulos, P.V.; Katsamouris, A.; Todor, A.; Filis, K.A.; Sabbah, H.N. Abnormal mitochondrial respiration in skeletal muscle in patients with peripheral arterial disease. J. Vasc. Surg. 2003 , 38 , 827–832. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Paradis, S.; Charles, A.L.; Meyer, A.; Lejay, A.; Scholey, J.W.; Chakfe, N.; Zoll, J.; Geny, B. Chronology of mitochondrial and cellular events during skeletal muscle ischemia-reperfusion. Am. J. Physiol. Cell Physiol. 2016 , 310 , C968–C982. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Arany, Z.; Foo, S.Y.; Ma, Y.; Ruas, J.L.; Bommi-Reddy, A.; Girnun, G.; Marcus Cooper, M.; Dina Laznik, D.; Jessica Chinsomboon, J.; Shamina, M.; et al. HIF independent regulation of VEGF and angiogenesis by the transcriptional coactivator PGC-1alpha. Nature 2008 , 451 , 1008–1012. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Koutakis, P.; Miserlis, D.; Myers, S.A.; Kim, J.K.; Zhu, Z.; Papoutsi, E.; Papoutsi, E.; Swanson, S.A.; Haynatzki, G.; Ha, D.M.; et al. Abnormal accumulation of desmin in gastrocnemiusmyofibers of patients with peripheral artery disease: Associations with altered myofiber morphology and density, mitochondrialdysfunction and impaired limb function. J. Histochem. Cytochem. 2015 , 63 , 256–269. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Ceci, R.; Beltran Valls, M.R.; Duranti, G.; Dimauro, I.; Quaranta, F.; Pittaluga, M.; Sabatini, S.; Caserotti, P.; Parisi, P.; Parisi, A.; et al. Oxidative stress responses to a graded maximal exercise test in older adults following explosive-type resistance training. Redox Biol. 2014 , 2 , 65–72. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Di Meo, S.; Napolitano, G.; Venditti, P. Mediators of Physical Activity Protection against ROS-Linked Skeletal Muscle Damage. Int. J. Mol. Sci. 2019 , 20 , 3024. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Knuuti, J.; Wijns, W.; Saraste, A.; Capodanno, D.; Barbato, E.; Funck-Brentano, C.; Prescott, E.; Storey, R.F.; Deaton, C.; Cuisset, T.; et al. 2019 ESC Guidelines for the diagnosis and management of chronic coronary syndromes. Eur. Heart J. 2019 , 41 . [ Google Scholar ] [ CrossRef ]
  • Melikoglu, M.A.; Kaldirimci, M.; Katkat, D.; Sen, I.; Kaplan, I.; Senel, K. The effect of regular long term training on antioxidant enzymatic activities. J. Sports Med. Phys. Fitness 2008 , 48 , 388–390. [ Google Scholar ]
  • Mrakic-Sposta, S.; Gussoni, M.; Moretti, S.; Pratali, L.; Giardini, G.; Tacchini, P.; Dellanoce, C.; Tonacci, A.; Mastorci, F.; Borghini, A.; et al. Effects of Mountain Ultra-Marathon Running on ROS Production and Oxidative Damage by Micro-Invasive Analytic Techniques. PLoS ONE 2015 , 10 , e0141780. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Vezzoli, A.; Pugliese, L.; Marzorati, M.; Serpiello, F.R.; La Torre, A.; Porcelli, S. Time-course changes of oxidative stress response to high-intensity discontinuous training versus moderate-intensity continuous training in masters runners. PLoS ONE 2014 , 9 , e87506. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Veal, E.; Jackson, T.; Latimer, H. Role/s of ‘Antioxidant’ Enzymes in Ageing. Subcell. Biochem. 2018 , 90 , 425–450. [ Google Scholar ] [ PubMed ]
  • Peake, J.M.; Markworth, J.F.; Nosaka, K.; Raastad, T.; Wadley, G.D.; Coffey, V.G. Modulating exercise-induced hormesis: Does less equal more? J. Appl. Physiol. 2015 , 119 , 172–189. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Nemes, R.; Koltai, E.; Taylor, A.W.; Suzuki, K.; Gyori, F.; Radak, Z. Reactive Oxygen and Nitrogen Species Regulate Key Metabolic, Anabolic, and Catabolic Pathways in Skeletal Muscle. Antioxidants 2018 , 7 , 85. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Nakajima, T.; Kurano, M.; Hasegawa, T.; Takano, H.; Iida, H.; Yasuda, T.; Nakajima, T.; Kurano, M.; Hasegawa, T.; Takano, H.; et al. Pentraxin3 and high-sensitive C-reactive protein are independent inflammatory markers released during high-intensity exercise. Eur. J. Appl. Physiol. 2010 , 110 , 905–913. [ Google Scholar ] [ CrossRef ]
  • Signorelli, S.S.; Mazzarino, M.C.; Di Pino, L.; Malaponte, G.; Porto, C.; Pennisi, G.; Marchese, G.; Costa, M.P.; Digrandi, D.; Celotta, G.; et al. High Circulating Levels of Cytokines (IL-6 and TNFalpha), Adhesion Molecules (VCAM-1 and ICAM-1) and Selectins in Patients With Peripheral Arterial Disease at Rest and After a Treadmill Test. Vasc. Med. 2003 , 8 , 15–19. [ Google Scholar ] [ CrossRef ]
  • Andreozzi, G.M.; Martini, R.; Cordova, R.; D’Eri, A.; Salmistraro, G.; Mussap, M. Plebani M Circulating levels of cytokines (IL-6 and IL-1beta) in patients with intermittent claudication, at rest, after maximal exercise treadmill test and during restore phase. Could they be progression markers of the disease? Int. Angiol. 2007 , 26 , 245–252. [ Google Scholar ]
  • Norgren, L.; Hiatt, W.R.; Dormandy, J.A.; Nehler, M.R.; Harris, K.A.; Fowkes, F.G.R. Inter-society consensus for the management of peripheral arterial disease (TASC II). J. Vasc. Surg. 2007 , 45 , S65–S67. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • McDermott, M.M.; Polonsky, T.S. Home-based exercise. A therapeutic option for peripheral arterial disease. Circulation 2016 , 134 , 1127–1129. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Brevetti, G.; Angelini, C.; Rosa, M.; Carrozzo, R.; Perna, S.; Corsi, M.; Matarazzo, A.; Marcialis, A. Muscle Carnitine Deficiency in Patients With Severe Peripheral Vascular Disease. Circulation 1991 , 84 , 1490–1495. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Hiatt, W.R.; Regensteiner, J.G.; Wolfel, E.E.; Ruff, L.; Brass, E.P. Carnitine and acyl carnitine metabolism during exercise in humans: Dependence on skeletal muscle metabolic state. J. Clin. Investig. 1989 , 84 , 1167–1173. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Brevetti, G.; Chiariello, M.; Ferulano, G.; Policicchio, A.; Nevola, E.; Rossini, A.; Attisano, T.; Ambrosio, G.; Siliprandi, N.; Angelini, C. Increases in walking distance in patients with peripheral vascular disease treated with L-carnitine: A double-blind, cross-over study. Circulation 1988 , 77 , 767–773. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Pignatelli, P.; Lenti, L.; Sanguigni, V.; Frati, G.; Simeoni, I.; Gazzaniga, P.P.; Pulcinelli, F.M.; Violi, F. Carnitine inhibits arachidonic acid turnover, platelet function, and oxidative stress. Am. J. Physiol. Heart Circ. Physiol. 2003 , 284 , H41–H48. [ Google Scholar ] [ CrossRef ]
  • Signorelli, S.S.; Malaponte, G.; Di Pino, L.; Digrandi, D.; Pennisi, G.; Mazzarino, M.C. Effects of ischaemic stress on leukocyte activation processes in patients with chronic peripheral occlusive arterial disease: Role of L-propionyl carnitine administration. Pharmacol. Res. 2001 , 44 , 305–309. [ Google Scholar ] [ CrossRef ]
  • Stasi, M.A.; Scioli, M.G.; Arcuri, G.; Mattera, G.G.; Lombardo, K.; Marcellini, M.; Riccioni, T.; De Falco, S.; Pisano, C.; Luigi Spagnoli, L.G.; et al. Propionyl-L-carnitine improves postischemic blood flow recovery and arteriogenetic revascularization and reduces endothelial NADPH-oxidase 4-mediated superoxide production. Arterioscler. Thromb. Vasc. Biol. 2010 , 30 , 426–435. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Silvestro, A.; Scopacasa, F.; Oliva, G.; de Cristofaro, T.; Iuliano, L.; Brevetti, G. Vitamin C prevents endothelial dysfunction induced by acute exercise in patients with intermittent claudication. Atherosclerosis 2002 , 165 , 277–283. [ Google Scholar ] [ CrossRef ]
  • Carr, A.C.; Zhu, B.Z.; Frei, B. Potential anti-atherogenic mechanisms of ascorbate (vitamin C) and alpha-tocopherol (vitamin E). Circ. Res. 2000 , 87 , 349–354. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Esterbauer, H.; Dieber-Rotheneder, M.; Striegl, G.; Waeg, G. Role of vitamin E in preventing the oxidation of low-density lipoprotein. Am. J. Clin. Nutr. 1991 , 53 (Suppl. 1), 314S–321S. [ Google Scholar ] [ CrossRef ]
  • Stephens, N.G.; Parsons, A.; Schofield, P.M.; Kelly, F.; Cheeseman, K.; Mitchinson, M.J. Randomised controlled trial of vitamin E in patients with coronary disease: Cambridge Heart Antioxidant Study (CHAOS). Lancet 1996 , 347 , 781–786. [ Google Scholar ] [ CrossRef ]
  • Arosio, E.; De Marchi, S.; Zannoni, M.; Prior, M.; Lechi, A. Effect of glutathione infusion on leg arterial circulation, cutaneous microcirculation, and pain-free walking distance in patients with peripheral obstructive arterial disease: A randomized, double-blind, placebo-controlled trial. Mayo Clin. Proc. 2002 , 77 , 754–759. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Loffredo, L.; Marcoccia, A.; Pignatelli, P.; Andreozzi, P.; Borgia, M.C.; Cangemi, R.; Chiarotti, F.; Violi, F. Oxidative-stress-mediated arterial dysfunction in patients with peripheral arterial disease. Eur. Heart J. 2007 , 28 , 608–612. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Carnevale, R.; Loffredo, L.; Nocella, C.; Bartimoccia, S.; Bucci, T.; De Falco, E.; Peruzzi, M.; Chimenti, I.; Biondi-Zoccai, G.; Pignatelli, P.; et al. Epicatechin and catechin modulate endothelial activation induced by platelets of patients with peripheral artery disease. Oxid. Med. Cell Longev. 2014 , 2014 , 691015. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Heumüller, S.; Wind, S.; Barbosa-Sicard, E.; Schmidt, H.H.H.W.; Busse, R.; Schröder, K.; Brandes, R.P. Apocynin is not an inhibitor of vascular NADPH oxidases but an antioxidant. Hypertension 2008 , 51 , 211–217. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Pignatelli, P.; Lenti, L.; Sanguigni, V.; Frati, G.; Simeoni, I.; Gazzaniga, P.P.; Pulcinelli, F.M.; Violi, F. Plasma heme oxygenase 1 is decreased in peripheral artery disease patients. Mol. Med. Rep. 2016 , 14 , 3459–3463. [ Google Scholar ]
  • Gardner, A.W.; Parker, D.E.; Webb, N.; Montgomery, P.S.; Scott, K.J.; Blevins, S.M. Calf Muscle Hemoglobin Oxygen Saturation Characteristics and Exercise Performance in Patients With Intermittent Claudication. J. Vasc. Surg. 2008 , 48 , 644–649. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Hickman, P.; Harrison, D.K.; Hill, A.; McLaren, M.; Tamei, H.; McCollum, P.T.; Belch, J.J. Exercise in patients with intermittent claudication results in the generation of oxygen derived free radicals and endothelial damage. Adv. Exp. Med. Biol. 1994 , 361 , 565–570. [ Google Scholar ]
  • Hamburg, N.M.; Balady, G.J. Exercise rehabilitation in peripheral artery disease: Functional impact and mechanisms of benefits. Circulation 2011 , 123 , 87–97. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Regensteiner, J.G.; Hiatt, W.R.; Coll, J.R.; Criqui, M.H.; Treat-Jacobson, D.; McDermott, M.M.; Hirsch, A.T.; Treat-Jacobson, D.; McDermott, M.M. The impact of peripheral arterial disease on health-related quality of life in the Peripheral Arterial Disease Awareness, Risk, and Treatment: New Resources for Survival (PARTNERS) Program. Vasc. Med. 2008 , 13 , 15–22. [ Google Scholar ] [ CrossRef ]
  • Garg, P.K.; Tian, L.; Criqui, M.H.; Liu, K.; Ferrucci, L.; Guralnik, J.M.; Tan, J.; NcDermott, M.M. Physical activity during daily life and mortality in patients with peripheral arterial disease. Circulation 2006 , 114 , 242–248. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Koutakis, P.; Ismaeel, A.; Farmer, P.; Purcell, S.; SSmith, R.S.; Eidson, J.L.; Bohannon, W.T. Oxidative stress and antioxidant treatment in patients with peripheral arterial disease. Physiol. Rep. 2018 , 6 , e13650. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Dopheide, J.F.; Scheer, M.; Doppler, C.; Obst, V.; Stein, P.; Vosseler, M.; Abegunewardene, N.; Gori, T.; Münzel, T.; Daiber, A.; et al. Change of walking distance in intermittent claudication: Impact on inflammation, oxidative stress and mononuclear cells: A pilot study. Clin. Res. Cardiol. 2015 , 104 , 751–763. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Loffredo, L.; Carnevale, R.; Cangemi, R.; Angelico, F.; Augelletti, T.; Di Santo, S.; Calabrese, C.M.; DellaVolpe, L.; Pignatelli, P.; Perri, L.; et al. NOX2 up-regulation is associated with artery dysfunction in patients with peripheral artery disease. Int. J. Cardiol. 2013 , 165 , 184–192. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Muller, M.D.; Drew, R.C.; Blaha, C.A.; Mast, J.L.; Cui, J.; Reed, A.; Sinoway, L.I. Oxidative stress contributes to the augmented exercise pressor reflex in peripheral arterial disease patients. J. Physiol. 2012 , 590 , 6237–6246. [ Google Scholar ] [ CrossRef ]
  • Lan, Y.; Liu, H.; Liu, J.; Zhao, H.; Wang, H. Is serum total bilirubin a predictor of prognosis in arteriosclerotic cardiovascular disease? A meta-analysis. Medicine 2019 , 98 , e17544. [ Google Scholar ] [ CrossRef ]
  • Maines, M.D. The heme oxygenase system: A regulator of second messenger gases. Annu. Rev. Pharmacol. Toxicol. 1997 , 37 , 517–554. [ Google Scholar ] [ CrossRef ]
  • Itoh, K.; Chiba, T.; Takahashi, S.; Ishii, T.; Igarashi, K.; Katoh, Y.; Oyake, T.; Hayashi, N.; Satoh, K.; Hatayama, I.; et al. An nrf2/small maf heterodimer mediates the induction of phase ii detoxifying enzyme genes through antioxidant response elements. Biochem. Biophys. Res. Commun. 1997 , 236 , 313–322. [ Google Scholar ] [ CrossRef ]
  • Ishikawa, K.; Maruyama, Y. Heme oxygenase as an intrinsic defense system in vascular wall: Implication against atherogenesis. J. Atheroscler. Thromb. 2001 , 8 , 63–70. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Stocker, R.; Yamamoto, Y.; McDonagh, A.F.; Glazer, A.N.; Ames, B.N. Bilirubin is an antioxidant of possible physiological importance. Science 1987 , 235 , 1043–1046. [ Google Scholar ] [ CrossRef ]
  • Kim, J.A.; Territo, M.C.; Wayner, E.; Carlos, T.M.; Parhami, F.; Smith, C.W.; Haberland, M.E.; Fogelman, A.M.; Berliner, J.A. Partial characterization of leukocyte binding molecules on endothelial cells induced byminimally oxidized ldl. Arterioscler. Thromb. 1994 , 14 , 427–433. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Duckers, H.J.; Boehm, M.; True, A.L.; Yet, S.F.; San, H.; Park, J.L.; Clinton Webb, R.; Lee, M.E.; Nabel, G.J.; Nabel, E.G. Heme oxygenase-1 protects against vascular constriction and proliferation. Nat. Med. 2001 , 7 , 693–698. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Mayer, M. Association of serum bilirubin concentration with risk of coronary artery disease. Clin. Chem. 2000 , 46 , 1723–1727. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Ishikawa, K.; Sugawara, D.; Wang, X.; Suzuki, K.; Itabe, H.; Maruyama, Y.; Lusis, A.J. Heme oxygenase-1 inhibits atherosclerotic lesion formation in LDL-receptor knockout mice. Circ. Res. 2001 , 88 , 506–512. [ Google Scholar ] [ CrossRef ]
  • Ishikawa, K.; Sugawara, D.; Goto, J.; Watanabe, Y.; Kawamura, K.; Shiomi, M.; Itabe, H.; Maruyama, Y. Heme oxygenase-1 inhibits atherogenesis in Watanabe heritable hyperlipidemic rabbits. Circulation 2001 , 104 , 1831–1836. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Hayashi, S.; Takamiya, R.; Yamaguchi, T.; Matsumoto, K.; Tojo, S.J.; Tamatani, T.; Kitajima, M.; Makino, N.; Ishimura, Y.; Suematsu, M. Induction of heme oxygenase-1 suppresses venular leukocyte adhesion elicited by oxidative stress: Role of bilirubin generated by the enzyme. Circ. Res. 1999 , 85 , 663–671. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Kishimoto, Y.; Ibe, S.; Saita, E.; Sasaki, K.; Hanako, N.; Miura, K.; Ikegami, Y.; Ohmori, R.; Kondo, K.; Momiyama, Y. Plasma Heme Oxygenase-1 Levels in Patients with Coronary and Peripheral Artery Diseases. Dis. Markers 2018 , 2018 , 6138124. [ Google Scholar ] [ CrossRef ]
  • Li Volti, G.; Seta, F.; Schwartzman, M.L.; Nasjletti, A.; Abraham, N.G. Heme oxygenase attenuates angiotensin II mediated increase in cyclooxygenase-2 activity in human femoral endothelial cells. Hypertension 2003 , 41 , 715–719. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Cao, J.; Peterson, S.J.; Sodhi, K.; Vanella, L.; Barbagallo, I.; Rodella, L.F.; Schwartzman, M.L.; Abraham, N.G.; Kappas, A. Heme Oxygenase Gene Targeting to Adipocytes Attenuates Adiposity and Vascular Dysfunction in Mice Fed a High-Fat Diet. Hypertension 2012 , 60 , 467–475. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Abraham, N.G.; Kappa, A. Pharmacological and clinical aspects of heme oxygenase. Pharmacol. Rev. 2008 , 60 , 79–127. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Suzuki, M.; Iso-o, N.; Takeshita, S.; Tsukamoto, K.; Mori, I.; Sato, T.; Ohno, M.; Nagai, R.; Ishizaka, N. Facilitated angiogenesis induced by heme oxygenase-1 gene transfer in a rat model of hind limb ischemia. Biochem. Biophys. Res. Commun. 2003 , 302 , 138–143. [ Google Scholar ] [ CrossRef ]
  • Lee, T.S.; Chang, C.C.; Zhu, Y.; Shyy, J.Y. Simvastatin induces heme oxygenase-1: A novel mechanism of vessel protection. Circulation 2004 , 110 , 1296–1302. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Vijayan, V.; Wagener, F.; Immenschuh, S. The macrophage heme-heme oxygenase-1 system and its role in inflammation. Biochem. Pharmacol. 2018 , 153 , 159–167. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Kishimoto, Y.; Kondo, K.; Momiyama, Y. The Protective Role of Heme Oxygenase-1 in Atherosclerotic Diseases. Int. J. Mol. Sci. 2019 , 20 , 3628. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Pasini, A.M.; Stranieri, C.; Rigoni, A.M.; De Marchi, S.; Peserico, D.; Mozzini, C.; Cominacini, L.; Garbin, U. Physical Exercise reduces cytotoxicity and up-regulates Nrf2 and UPR expression in circulating cells of peripheral artery disease patients: An hypoxic Adaptation? J. Atheroscler. Thromb. 2018 , 25 , 808–820. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
AntioxidantsEffects and MarkersReferences
-CarnitineFMD and brachial basal diameter significantly increased
Increase in NOx bioavailability
Decrease in 8-OHdG
[ , , , , ]
Reduces OxS walking induced
Reduces arterial pressure response to physical exercise
No reduction of flow mediated dilatation (FMD) by maximal physical exercise
No elevation of TABRS OxS marker
No elevation of soluble CMA-1
[ , , ]
Reduces OxS walking induced [ , ]
Reduces pain free walking distance
Improves macrocirculatory flow after physical exercise
[ , ]


Enhances platelet activation
Increases the release of soluble cell adhesion molecules (sCAMs)
Decreases eNOS activation
Effects on NO bioavailability
[ , ]

Share and Cite

Signorelli, S.S.; Marino, E.; Scuto, S.; Di Raimondo, D. Pathophysiology of Peripheral Arterial Disease (PAD): A Review on Oxidative Disorders. Int. J. Mol. Sci. 2020 , 21 , 4393. https://doi.org/10.3390/ijms21124393

Signorelli SS, Marino E, Scuto S, Di Raimondo D. Pathophysiology of Peripheral Arterial Disease (PAD): A Review on Oxidative Disorders. International Journal of Molecular Sciences . 2020; 21(12):4393. https://doi.org/10.3390/ijms21124393

Signorelli, Salvatore Santo, Elisa Marino, Salvatore Scuto, and Domenico Di Raimondo. 2020. "Pathophysiology of Peripheral Arterial Disease (PAD): A Review on Oxidative Disorders" International Journal of Molecular Sciences 21, no. 12: 4393. https://doi.org/10.3390/ijms21124393

Article Metrics

Article access statistics, further information, mdpi initiatives, follow mdpi.

MDPI

Subscribe to receive issue release notifications and newsletters from MDPI journals

U.S. flag

An official website of the United States government

Here’s how you know

Official websites use .gov A .gov website belongs to an official government organization in the United States.

Secure .gov websites use HTTPS A lock ( A locked padlock ) or https:// means you’ve safely connected to the .gov website. Share sensitive information only on official, secure websites.

  • Heart-Healthy Living
  • High Blood Pressure
  • Sickle Cell Disease
  • Sleep Apnea
  • Information & Resources on COVID-19
  • The Heart Truth®
  • Learn More Breathe Better®
  • Blood Diseases and Disorders Education Program
  • Publications and Resources
  • Blood Disorders and Blood Safety
  • Sleep Science and Sleep Disorders
  • Lung Diseases
  • Health Disparities and Inequities
  • Heart and Vascular Diseases
  • Precision Medicine Activities
  • Obesity, Nutrition, and Physical Activity
  • Population and Epidemiology Studies
  • Women’s Health
  • Research Topics
  • Clinical Trials
  • All Science A-Z
  • Grants and Training Home
  • Policies and Guidelines
  • Funding Opportunities and Contacts
  • Training and Career Development
  • Email Alerts
  • NHLBI in the Press
  • Research Features
  • Past Events
  • Upcoming Events
  • Mission and Strategic Vision
  • Divisions, Offices and Centers
  • Advisory Committees
  • Budget and Legislative Information
  • Jobs and Working at the NHLBI
  • Contact and FAQs
  • NIH Sleep Research Plan
  • < Back To Research Topics

Peripheral Artery Disease (PAD) Research

Language switcher.

Research that improves the health of people with peripheral artery disease (PAD) is part of NHLBI’s larger commitment to understanding and treating cardiovascular disease. PAD is caused by atherosclerosis that restricts blood flow through the arteries supplying the legs. PAD may be silent (without symptoms) or cause muscle pain with exertion such as walking or climbing stairs. Severe disease, termed critical limb ischemia (CLI) and present in only a small percentage of those with PAD, is associated with risk of amputation. Awareness that people with PAD often also have atherosclerosis of arteries supplying other organs such as the heart, and brain, is important because the risk of heart attack and stroke is elevated. Appropriate medical therapy to reduce risk should be an integral part of addressing PAD.

Studies funded and led by the NHLBI focus on ways to treat and prevent both atherosclerosis in general and PAD in particular. NHLBI’s research has revealed specific risk factors for developing PAD, including smoking and having diabetes, which can be treated with lifestyle interventions and medications. Researchers have also been working to improve existing treatments and develop new ones, some of which are described below.

Icon illustration of medical cross symbol

NHLBI research that really made a difference

  • NHLBI’s Claudication: Exercise Versus Endoluminal Revascularization ( CLEVER ) study showed that a supervised exercise program was more effective at improving treadmill walking capacity than angioplasty with or without stenting of an occluded blood vessel, or using or medicines alone. The results of CLEVER and other research studies including the Improving Functioning in Peripheral Arterial Disease trial, led the Centers for Medicare & Medicaid Services to reimburse supervised exercise training (SET) programs for symptomatic PAD.
  • The Low InTensity Exercise Intervention in PAD ( LITE ) trial demonstrated that a home-based coaching program incorporating high-intensity but not low-intensity walking exercise improved walking performance in people with symptomatic PAD. This type of intervention does not require travel to a training center and is likely to be more accessible than traditional in-center SET programs.
  • Researchers found that African Americans taking part in the Jackson Heart Study and the Atherosclerosis Risk in Communities Study had an increased risk of PAD if they smoked. Smoking was associated with PAD in a dose-dependent manner: People who smoked twice as much or for twice as long as others were twice as likely to develop PAD.

Current research funded by the NHLBI

Our Division of Cardiovascular Sciences , which includes the Vascular Biology and Hypertension Branch, oversees much of the research on PAD we fund.

Find related funding opportunities and program contacts in Atherothrombosis and Coronary Artery Disease and Vascular Biology and Hypertension research.

Current research on PAD treatments

PAD is  a challenging condition to treat because the recommended treatment — regular walking — can be painful, difficult to undertake due to other co-existing health conditions such as arthritis, and/or not practicable in the home or community environment. NIH-funded research is working on ways to increase walking endurance and circulation in the legs by using mechanical devices.

  • Conventional ultrasound therapy for PAD involves using sound waves to increase blood flow in the legs. Until now, this therapy has been available only in clinical settings. NHLBI-funded researchers have developed an ultrasound device for people with PAD to wear when resting or sleeping. The device is like a sleeve worn over the legs and has its own portable power generator. Researchers hope to find out if this therapy improves blood flow in the legs.
  • Participants in the INTERCEDE (INTERmittent pneumatic ComprEssion for Disability rEversal in PAD) trial wear inflatable cuffs wrapped around their feet, ankles, and calves for 2 hours every day. Every 20 seconds, the cuffs rapidly inflate and then rapidly deflate. This increases blood flow through the legs. Researchers will look at whether the device improves walking performance, how it changes leg muscles and blood flow, and whether those changes last.
  • Small implantable sensors are being developed to monitor tissue oxygen levels in limbs of PAD patients during and after procedures done to restore blood flow.

Find more NHLBI-funded studies on PAD treatments  at the NIH RePORTER.

Eliminating health disparities in PAD

Health disparities exist whenever a person’s membership in a racial, ethnic, age, or other type of group makes them more likely to get or develop a disease or less likely to be diagnosed or treated for a disease. Researchers have found, for example, that African American smokers are three times more likely to develop PAD than white smokers. NHLBI-funded research is working to determine what health disparities exist, what causes them, and how to address them. One fundamental way NHLBI will achieve these goals is to ensure diversity in enrollment in all PAD research studies that involve people.

  • The NHLBI is funding PASOS (Peripheral Artery Disease Study of SOL, an ancillary study of the Hispanic Community Health Study/Study of Latinos ). As part of the study, researchers screen more than 6,000 participants age 45 and older for early signs of PAD, including reduced blood flow in the legs. They also use fitness trackers to measure activity levels. Participants with borderline poor circulation will be followed more closely to see what factors predict who will develop PAD.
  • The Variant Determinants of African American Limb Pathology in Peripheral Arterial Disease study hopes to learn more about how variants of a gene, BAG3, known to exist in both African Americans with critical limb ischemia (CLI) and with cardiomyopathy, may disrupt normal reparative links between muscle damaged by lack of oxygen and new blood vessel growth. If the hypothesis is correct, this gene may become a therapeutic target. Findings may help explain why African Americans with PAD disproportionately suffer from CLI.

Find more NHLBI-funded studies on health disparities and PAD at the NIH RePORTER.

Current research to better understand PAD

NHLBI-funded research has contributed to the understanding of the basic biology behind PAD. But there is still more to learn. This basic knowledge is what often help scientists develop future treatments.

  • A large NHLBI-funded multisite clinical trial, BEST-CLI , will compare effectiveness of two standard treatment strategies for CLI treatment: Endovascular and open surgical bypass revascularization. Extensive data on clinical course, adverse events, and clinical outcomes including amputation, major reintervention, and death will be collected over a period of 2 years.
  • Hematopoietic stem cells and progenitor cells are found in bone marrow and circulating blood and develop into other cells including blood cells. Researchers want to know whether giving people with PAD a protein that stimulates the release of stem cells from the bone marrow will promote the growth of blood vessels in the legs, improve circulation, and increase walking endurance.
  • Sometimes, to better understand a disease, researchers build computer models based on real-life experiments they have run. These models allow them to change variables and see what the outcomes of those changes might be. NHLBI-funded researchers are doing just that to study PAD. Their state-of-the-art computer model will represent the way new blood vessels form, grow, and provide oxygen-carrying blood to the legs and feet. Researchers hope that this work will provide a better understanding of PAD and help suggest new treatments.
  • Researchers believe they have worked out the details of a cellular pathway that explain the lack of circulation and oxygen in the lower extremities of patients with PAD. They now want to test their hypothesis by giving people with PAD a protein that promotes the growth of blood vessels by blocking another protein that inhibits vessel growth. The researchers hope this will improve circulation in people with PAD.

Find more NHLBI-funded studies on understanding PAD at the NIH RePORTER. 

Illustration of PAD showing a blocked blood vessel

Learn about research that confirms the safety of drug-coated stents and other devices used to open up blocked arteries in the legs of patients with PAD.

PAD research labs at the NHLBI

Our Division of Intramural Research , which includes investigators from the Translational Vascular Medicine Branch and its Experimental Atherosclerosis Laboratory , performs research on PAD.

Related PAD programs

  • Researchers in the NHLBI-funded  Multi-Ethnic Study of Atherosclerosis (MESA) followed a group of study participants for 10 years and found that nearly 5% of them developed PAD. However, participants who scored higher on the American Heart Association’s Life’s Simple 7 checklist for maximizing heart health were less likely to have PAD; their risk decreased about 17% for each of the Life’s Simple 7 criteria they met. Learn more about this study:  Life’s Simple 7 and Peripheral Artery Disease: The Multi-Ethnic Study of Atherosclerosis .
  • NHLBI’s Vascular Interventions/Innovations and Therapeutic Advances (VITA) Program studies and develops promising tests and treatments for blood vessel diseases, including PAD, and vascular access for hemodialysis. VITA has developed a new approach for treating people who have serious complications from PAD. One problem with angioplasty (a common treatment for PAD to open narrowed or blocked arteries) is the risk that the blood vessels will scar and re-narrow. A VITA project developed bioengineered tubes that one day could be used instead of a person’s own blood vessels or donor vessels for bypass revascularization . They are unique because the body’s immune system will not reject them as foreign. Learn more about the VITA program:  Next-generation replacement blood vessels target kidney and heart disease .

Explore more NHLBI research on PAD

The sections above provide you with the highlights of NHLBI-supported research on PAD. You can explore the full list of NHLBI-funded studies on the NIH RePORTER .

To find more studies:

  • Type your search words into the  Quick Search  box and press enter. 
  • Check  Active Projects  if you want current research.
  • Select the  Agencies  arrow, then the  NIH  arrow, then check  NHLBI .

If you want to sort the projects by budget size — from the biggest to the smallest — click on the  FY Total Cost by IC  column heading.

  • Search Menu
  • Sign in through your institution
  • Volume 2024, Issue 6, June 2024 (In Progress)
  • Volume 2024, Issue 5, May 2024
  • Bariatric Surgery
  • Breast Surgery
  • Cardiothoracic Surgery
  • Colorectal Surgery
  • Colorectal Surgery, Upper GI Surgery
  • Gynaecology
  • Hepatobiliary Surgery
  • Interventional Radiology
  • Neurosurgery
  • Ophthalmology
  • Oral and Maxillofacial Surgery
  • Otorhinolaryngology - Head & Neck Surgery
  • Paediatric Surgery
  • Plastic Surgery
  • Transplant Surgery
  • Trauma & Orthopaedic Surgery
  • Upper GI Surgery
  • Vascular Surgery
  • Author Guidelines
  • Submission Site
  • Open Access
  • Reasons to Submit
  • About Journal of Surgical Case Reports
  • Editorial Board
  • Advertising and Corporate Services
  • Journals Career Network
  • Self-Archiving Policy
  • Journals on Oxford Academic
  • Books on Oxford Academic

Issue Cover

Article Contents

Introduction, case presentation.

  • < Previous

An usually early presentation of peripheral arterial disease in a 30-year-old woman

  • Article contents
  • Figures & tables
  • Supplementary Data

Herbert Oye, Arham Aslam, An usually early presentation of peripheral arterial disease in a 30-year-old woman, Journal of Surgical Case Reports , Volume 2021, Issue 10, October 2021, rjab457, https://doi.org/10.1093/jscr/rjab457

  • Permissions Icon Permissions

Peripheral vascular disease is commonly thought to be a disease of elderly patients. However, with certain environmental factors and predetermined genetics, it can present much earlier in life. This case report demonstrates a case of severe peripheral vascular disease in a 30-year-old woman with an 11-year history of smoking. We report the case of a 30-year-old woman who presented to our clinic with a 9-year history of peripheral vascular disease. She complained of worsening claudication in both lower extremities and intermittent pain at rest. A CT arteriogram was conducted and showed significant stenosis of both left and right iliac arteries, as well as stenosis of the distal aorta. In the setting of certain genetic and environmental factors, the presentation of peripheral vascular disease can occur significantly earlier in life.

With roughly 6.5 million Americans over the age of 40 affected by peripheral vascular disease [ 1 ], it is commonly thought to be a disease of the aged. In the setting of certain risk factors, such as diabetes, hypercholesterolemia, hypertension and smoking, atherosclerotic plaques take years to build-up enough to cause significant stenosis. Although all vessels may be affected, the lower extremities are more commonly involved in the symptoms of peripheral vascular disease. We report the case of a 30-year-old woman who presented to our clinic with worsening symptoms of peripheral vascular disease.

The patient was a 30-year-old woman who was diagnosed with peripheral vascular disease 9 years prior to this encounter. She presented to our clinic with complaints of worsening pain in the lower extremities with activity and sometimes pain at rest. The patient had a medical history of hypertension and admitted to non-compliance with medication. She has been smoking cigarettes for 11 years and usually consumes 1 pack per day. In addition, she reported a strong family history of coronary artery disease. The patient was taken to the office-based lab located within the clinic and was prepared sterilely for a catheter arteriogram.

Catheter arteriogram results showed significant occlusion of the left iliac artery ( Figs 3 and 4 ), the right iliac artery ( Fig. 1 ), as well as the distal abdominal aorta ( Fig. 2 ). There were numerous collateral vessels noted, indicating the presence of long-standing proximal stenosis ( Fig. 1 ). The decision was made to use angioplasty to help widen the areas of stenosis. An 8 mm × 40 mm Passeo balloon was advanced from the right femoral artery into the left iliac artery ( Fig. 7 ) and insufflated to 6 mmHg ( Fig. 6 ). The balloon was allowed to remain expanded for 1 min and then was deflated. Next, the distal aorta was repaired in a similar fashion. The balloon was advanced into the distal aorta from the right femoral access and insufflated to 12 mmHg ( Fig. 8 ). The balloon was allowed to remain expanded for 90 s.

Fluoroscopy of right iliac artery showing vast collateral vascular formation, indicating long-standing proximal obstruction.

Fluoroscopy of right iliac artery showing vast collateral vascular formation, indicating long-standing proximal obstruction.

Fluoroscopy of distal aorta showing narrowing.

Fluoroscopy of distal aorta showing narrowing.

Fluoroscopy of bilateral iliac arteries showing significant arterial disease.

Fluoroscopy of bilateral iliac arteries showing significant arterial disease.

Fluoroscopy of bilateral iliac arteries showing significant arterial disease.

Angioplasty of right iliac artery being performed.

Angioplasty of left iliac artery being performed.

Angioplasty of left iliac artery being performed.

Fluoroscopy of guidewire being passed into left iliac artery.

Fluoroscopy of guidewire being passed into left iliac artery.

Angioplasty of distal aorta being performed.

Angioplasty of distal aorta being performed.

Fluoroscopy showing dilation of the distal aorta post-angioplasty.

Fluoroscopy showing dilation of the distal aorta post-angioplasty.

Angioplasty of right iliac artery being performed.

Fluoroscopy showing dilation of the right iliac vessel post-angioplasty.

Fluoroscopy of right iliac artery showing improved perfusion.

Fluoroscopy of right iliac artery showing improved perfusion.

Fluoroscopy of left iliac artery showing improved perfusion.

Fluoroscopy of left iliac artery showing improved perfusion.

Fluoroscopy of left popliteal artery showing good perfusion.

Fluoroscopy of left popliteal artery showing good perfusion.

Fluoroscopy of right popliteal artery showing good perfusion.

Fluoroscopy of right popliteal artery showing good perfusion.

Lastly, the right iliac artery ( Fig. 5 ) was treated using the same method ( Fig. 10 ). After completion of the angioplasty procedure, catheter arteriogram was repeated to visualize the effectiveness of the procedure in relieving the extensive stenosis. The imaging revealed almost complete restoration of the diameter of both left and right iliac arteries ( Figs 11 and 12 ). The distal aorta also showed improved diameter after the procedure ( Fig. 9 ). Angiogram of both lower extremities was also performed. The catheter was passed distal to the stenosis that was previously present in the left iliac artery, to help visualize perfusion of its distal branches ( Figs 13 and 14 ). The catheter was removed from the right femoral artery and reinserted in a distal fashion, to help visualize downstream perfusion. The right popliteal artery was well visualized on angiogram ( Fig. 15 ), signifying strong perfusion. The patient was strongly advised to discontinue smoking cigarettes. In addition, the patient was advised to maintain regular physical exercise as tolerated and take baby aspirin daily to prevent thrombotic events.

The patient in this case developed peripheral vascular disease at an unusually early age than is expected. This may be a result of modifiable risk factors, such as hypertension, obesity and smoking. While pain in the lower extremities in young adults is commonly caused by trauma or muscular pathologies, the differential of peripheral arterial disease should also be considered. This is especially true in the setting of comorbid conditions and risk factors. The treatment of peripheral arterial disease focuses on reducing exposure to the risk factors, such as quitting smoking and regular exercise. Pharmacological options include antiplatelet agents, such as aspirin, to prevent thrombotic events. Cilostazol, a phosphodiesterase-3 inhibitor, has been shown to reduce the symptoms of claudication in patients with peripheral arterial disease.

The unique nature of significant peripheral arterial disease in a relatively young patient with a strong family history of coronary artery disease and smoking, carries significant comorbidity concerns for the future. We elected not to stent the vessels at this time, due to the small caliber of the vessels. With continued smoking, the placement of stents in this patient’s vessels may induce thrombosis.

Endovascular intervention in this young patient in the form of angioplasty alone, showed good promise and avoided the need for stenting. Endovascular intervention can delay the need for a major aortobifemoral bypass in this young patient, as well as thrombotic complications associated with stenting.

Virani   SS , Alonso   A , Benjamin   EJ , Bittencourt   MS , Callaway   CW , Carson   AP , et al.    Heart disease and stroke statistics—2020 update: a report from the American Heart Associationexternal icon . Circulation.   2020 ; 141 : e139 – 596 .

Google Scholar

  • arteriography
  • peripheral vascular diseases
  • environmental factors
  • constriction, pathologic
  • iliac artery
  • older adult
  • distal zone of aorta
Month: Total Views:
October 2021 55
November 2021 70
December 2021 38
January 2022 13
February 2022 27
March 2022 29
April 2022 52
May 2022 37
June 2022 43
July 2022 49
August 2022 31
September 2022 52
October 2022 73
November 2022 62
December 2022 34
January 2023 32
February 2023 50
March 2023 74
April 2023 74
May 2023 74
June 2023 81
July 2023 88
August 2023 91
September 2023 59
October 2023 58
November 2023 70
December 2023 77
January 2024 67
February 2024 78
March 2024 97
April 2024 76
May 2024 70
June 2024 26

Email alerts

Citing articles via, affiliations.

  • Online ISSN 2042-8812
  • Copyright © 2024 Oxford University Press and JSCR Publishing Ltd
  • About Oxford Academic
  • Publish journals with us
  • University press partners
  • What we publish
  • New features  
  • Open access
  • Institutional account management
  • Rights and permissions
  • Get help with access
  • Accessibility
  • Advertising
  • Media enquiries
  • Oxford University Press
  • Oxford Languages
  • University of Oxford

Oxford University Press is a department of the University of Oxford. It furthers the University's objective of excellence in research, scholarship, and education by publishing worldwide

  • Copyright © 2024 Oxford University Press
  • Cookie settings
  • Cookie policy
  • Privacy policy
  • Legal notice

This Feature Is Available To Subscribers Only

Sign In or Create an Account

This PDF is available to Subscribers Only

For full access to this pdf, sign in to an existing account, or purchase an annual subscription.

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings
  • My Bibliography
  • Collections
  • Citation manager

Save citation to file

Email citation, add to collections.

  • Create a new collection
  • Add to an existing collection

Add to My Bibliography

Your saved search, create a file for external citation management software, your rss feed.

  • Search in PubMed
  • Search in NLM Catalog
  • Add to Search

Endothelial function in cardiovascular medicine: a consensus paper of the European Society of Cardiology Working Groups on Atherosclerosis and Vascular Biology, Aorta and Peripheral Vascular Diseases, Coronary Pathophysiology and Microcirculation, and Thrombosis

Affiliations.

  • 1 Centre for Bioscience, Faculty of Science & Engineering, Manchester Metropolitan University, Manchester, UK.
  • 2 Institute of Clinical Chemistry, University and University Hospital Zurich, University Heart Center, Zurich, Switzerland.
  • 3 Laboratory of Translational Nutrition Biology, Swiss Federal Institute of Technology, Zurich, Switzerland.
  • 4 Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, Medical Faculty, University of Basel, Basel, Switzerland.
  • 5 Leviev Heart Center, Chaim Sheba Medical Center, Tel Hashomer, Israel.
  • 6 Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
  • 7 Cardiology Department, Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia.
  • 8 Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
  • 9 Department of Cardiology, Dupuytren University Hospital, Inserm U-1094, Limoges University, Limoges, France.
  • 10 Department of Cardiology, Center for Molecular Medicine, Karolinska University Hospital, Solna, Stockholm, Sweden.
  • 11 INSERM U1116, Université de Lorraine, Centre Hospitalier Régional Universitaire de Nancy, Vandoeuvre les Nancy, France.
  • 12 Cardiovascular Program-ICCC, IR-Hospital de la Santa Creu i Sant Pau, CiberCV, Autonomous University of Barcelona, Barcelona, Spain.
  • 13 Unit of Cardiology, Karolinska Institute and Karolinska University Hospital, Solna, Stockholm, Sweden.
  • 14 Catheterization Laboratory, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy.
  • 15 'CarolDavila' University of Medicine and Pharmacy, Bucharest, Romania.
  • 16 Vanderbilt University School of Medicine, Nashville, TN, USA.
  • 17 Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK.
  • 18 Department of Medicine, Jagiellonian University Collegium Medicum, Cracow, Poland.
  • 19 Laboratory of Clinical Chemistry and Hematology, University Medical Centre Utrecht, The Netherlands.
  • 20 Department of Infection, Immunity and Cardiovascular Disease, Bateson Centre & INSIGNEO Institute, University of Sheffield, Sheffield S10 2RX, UK.
  • 21 Insigneo Institute for In Silico Medicine, Sheffield, UK.
  • 22 Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
  • 23 Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
  • 24 Department of Cardiovascular Medicine, Medical Faculty, University of Münster, Münster, Germany.
  • 25 SRH Central Hospital Suhl, Suhl, Germany.
  • 26 Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillian-Universität (LMU) München, Munich, Germany.
  • 27 German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany.
  • 28 Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
  • 29 Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands.
  • 30 Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
  • PMID: 32282914
  • PMCID: PMC7797212
  • DOI: 10.1093/cvr/cvaa085

Endothelial cells (ECs) are sentinels of cardiovascular health. Their function is reduced by the presence of cardiovascular risk factors, and is regained once pathological stimuli are removed. In this European Society for Cardiology Position Paper, we describe endothelial dysfunction as a spectrum of phenotypic states and advocate further studies to determine the role of EC subtypes in cardiovascular disease. We conclude that there is no single ideal method for measurement of endothelial function. Techniques to measure coronary epicardial and micro-vascular function are well established but they are invasive, time-consuming, and expensive. Flow-mediated dilatation (FMD) of the brachial arteries provides a non-invasive alternative but is technically challenging and requires extensive training and standardization. We, therefore, propose that a consensus methodology for FMD is universally adopted to minimize technical variation between studies, and that reference FMD values are established for different populations of healthy individuals and patient groups. Newer techniques to measure endothelial function that are relatively easy to perform, such as finger plethysmography and the retinal flicker test, have the potential for increased clinical use provided a consensus is achieved on the measurement protocol used. We recommend further clinical studies to establish reference values for these techniques and to assess their ability to improve cardiovascular risk stratification. We advocate future studies to determine whether integration of endothelial function measurements with patient-specific epigenetic data and other biomarkers can enhance the stratification of patients for differential diagnosis, disease progression, and responses to therapy.

Keywords: Cardiovascular; Endothelial function.

Published on behalf of the European Society of Cardiology. All rights reserved. © The Author(s) 2020. For permissions, please email: [email protected].

PubMed Disclaimer

Endothelial dysfunction describes multiple phenotypic…

Endothelial dysfunction describes multiple phenotypic states. Left panel: In homeostatic conditions, the healthy…

Schematic representation of the endothelial…

Schematic representation of the endothelial factors underlying cardiovascular risk. ECFCs, endothelial colony-forming cells;…

Similar articles

  • Methods for evaluating endothelial function: a position statement from the European Society of Cardiology Working Group on Peripheral Circulation. Lekakis J, Abraham P, Balbarini A, Blann A, Boulanger CM, Cockcroft J, Cosentino F, Deanfield J, Gallino A, Ikonomidis I, Kremastinos D, Landmesser U, Protogerou A, Stefanadis C, Tousoulis D, Vassalli G, Vink H, Werner N, Wilkinson I, Vlachopoulos C. Lekakis J, et al. Eur J Cardiovasc Prev Rehabil. 2011 Dec;18(6):775-89. doi: 10.1177/1741826711398179. Eur J Cardiovasc Prev Rehabil. 2011. PMID: 21450600 Review.
  • Prognostic Value of Flow-Mediated Vasodilation in Brachial Artery and Fingertip Artery for Cardiovascular Events: A Systematic Review and Meta-Analysis. Matsuzawa Y, Kwon TG, Lennon RJ, Lerman LO, Lerman A. Matsuzawa Y, et al. J Am Heart Assoc. 2015 Nov 13;4(11):e002270. doi: 10.1161/JAHA.115.002270. J Am Heart Assoc. 2015. PMID: 26567372 Free PMC article. Review.
  • Expert consensus and evidence-based recommendations for the assessment of flow-mediated dilation in humans. Thijssen DHJ, Bruno RM, van Mil ACCM, Holder SM, Faita F, Greyling A, Zock PL, Taddei S, Deanfield JE, Luscher T, Green DJ, Ghiadoni L. Thijssen DHJ, et al. Eur Heart J. 2019 Aug 7;40(30):2534-2547. doi: 10.1093/eurheartj/ehz350. Eur Heart J. 2019. PMID: 31211361
  • Endothelial dysfunction and cardiovascular risk prediction in peripheral arterial disease: additive value of flow-mediated dilation to ankle-brachial pressure index. Brevetti G, Silvestro A, Schiano V, Chiariello M. Brevetti G, et al. Circulation. 2003 Oct 28;108(17):2093-8. doi: 10.1161/01.CIR.0000095273.92468.D9. Epub 2003 Oct 6. Circulation. 2003. PMID: 14530195 Clinical Trial.
  • Non-invasive endothelial function testing and the risk of adverse outcomes: a systematic review and meta-analysis. Xu Y, Arora RC, Hiebert BM, Lerner B, Szwajcer A, McDonald K, Rigatto C, Komenda P, Sood MM, Tangri N. Xu Y, et al. Eur Heart J Cardiovasc Imaging. 2014 Jul;15(7):736-46. doi: 10.1093/ehjci/jet256. Epub 2014 Jan 7. Eur Heart J Cardiovasc Imaging. 2014. PMID: 24399339 Review.
  • Investigations of Antioxidant and Anti-Cancer Activities of 5-Aminopyrazole Derivatives. Rapetti F, Spallarossa A, Russo E, Caviglia D, Villa C, Tasso B, Signorello MG, Rosano C, Iervasi E, Ponassi M, Brullo C. Rapetti F, et al. Molecules. 2024 May 14;29(10):2298. doi: 10.3390/molecules29102298. Molecules. 2024. PMID: 38792163 Free PMC article.
  • Role and molecular mechanism of NOD2 in chronic non-communicable diseases. Kong L, Cao Y, He Y, Zhang Y. Kong L, et al. J Mol Med (Berl). 2024 Jun;102(6):787-799. doi: 10.1007/s00109-024-02451-7. Epub 2024 May 14. J Mol Med (Berl). 2024. PMID: 38740600 Review.
  • Cytosolic DNA initiates a vicious circle of aging-related endothelial inflammation and mitochondrial dysfunction via STING: the inhibitory effect of Cilostazol. Zheng ZH, Wang JJ, Lin JG, Ye WL, Zou JM, Liang LY, Yang PL, Qiu WL, Li YY, Yang SJ, Zhao M, Zhou Q, Li CZ, Li M, Li ZM, Zhang DM, Liu PQ, Liu ZP. Zheng ZH, et al. Acta Pharmacol Sin. 2024 Apr 30. doi: 10.1038/s41401-024-01281-0. Online ahead of print. Acta Pharmacol Sin. 2024. PMID: 38689095
  • Remnant cholesterol trajectory and subclinical arteriosclerosis: a 10-year longitudinal study of Chinese adults. Yang PT, Tang L, Yang SQ, Shi QL, Wang YQ, Qin YX, Wang JG, Li Y. Yang PT, et al. Sci Rep. 2024 Apr 19;14(1):9037. doi: 10.1038/s41598-024-59173-6. Sci Rep. 2024. PMID: 38641617 Free PMC article.
  • Repressive H3K27me3 drives hyperglycemia-induced oxidative and inflammatory transcriptional programs in human endothelium. Sánchez-Ceinos J, Hussain S, Khan AW, Zhang L, Almahmeed W, Pernow J, Cosentino F. Sánchez-Ceinos J, et al. Cardiovasc Diabetol. 2024 Apr 5;23(1):122. doi: 10.1186/s12933-024-02196-0. Cardiovasc Diabetol. 2024. PMID: 38580969 Free PMC article.
  • Kalucka J, Bierhansl L, Conchinha NV, Missiaen R, Elia I, Bruning U, Scheinok S, Treps L, Cantelmo AR, Dubois C, de Zeeuw P, Goveia J, Zecchin A, Taverna F, Morales-Rodriguez F, Brajic A, Conradi LC, Schoors S, Harjes U, Vriens K, Pilz GA, Chen R, Cubbon R, Thienpont B, Cruys B, Wong BW, Ghesquiere B, Dewerchin M, De Bock K, Sagaert X, Jessberger S, Jones EAV, Gallez B, Lambrechts D, Mazzone M, Eelen G, Li X, Fendt SM, Carmeliet P.. Quiescent endothelial cells upregulate fatty acid beta-oxidation for vasculoprotection via redox homeostasis. Cell Metab 2018;28:881–894.e13. - PubMed
  • Shah AV, Birdsey GM, Peghaire C, Pitulescu ME, Dufton NP, Yang Y, Weinberg I, Osuna Almagro L, Payne L, Mason JC, Gerhardt H, Adams RH, Randi AM.. The endothelial transcription factor ERG mediates angiopoietin-1-dependent control of Notch signalling and vascular stability. Nat Commun 2017;8:16002. - PMC - PubMed
  • Cardner M, Yalcinkaya M, Goetze S, Luca E, Balaz M, Hunjadi M, Hartung J, Shemet A, Kränkel N, Radosavljevic S, Keel M, Othman A, Karsai G, Hornemann T, Claassen M, Liebisch G, Carreira E, Ritsch A, Landmesser U, Krützfeldt J, Wolfrum C, Wollscheid B, Beerenwinkel N, Rohrer L, von Eckardstein A.. Structure-function relationships of HDL in diabetes and coronary heart disease. JCI Insight 2020;5:e131491. - PMC - PubMed
  • Li X, Sun X, Carmeliet P.. Hallmarks of endothelial cell metabolism in health and disease. Cell Metab 2019;30:414–433. - PubMed
  • Dikalov S, Itani H, Richmond B, Arslanbaeva L, Vergeade A, Rahman SMJ, Boutaud O, Blackwell T, Massion PP, Harrison DG, Dikalova A.. Tobacco smoking induces cardiovascular mitochondrial oxidative stress, promotes endothelial dysfunction, and enhances hypertension. Am J Physiol Heart Circ Physiol 2019;316:H639–H646. - PMC - PubMed

Publication types

  • Search in MeSH

Related information

Grants and funding.

  • R35 HL140016/HL/NHLBI NIH HHS/United States
  • 692511/ERC_/European Research Council/International
  • WT_/Wellcome Trust/United Kingdom
  • 214567/Z/18/Z/WT_/Wellcome Trust/United Kingdom
  • RG/19/10/34506/BHF_/British Heart Foundation/United Kingdom
  • P01 HL129941/HL/NHLBI NIH HHS/United States

LinkOut - more resources

Full text sources.

  • Archivio Istituzionale della Ricerca Unimi
  • Europe PubMed Central
  • Ovid Technologies, Inc.
  • PubMed Central
  • Silverchair Information Systems
  • White Rose Research Online
  • Genetic Alliance
  • MedlinePlus Health Information

full text provider logo

  • Citation Manager

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Characteristics of motor evoked potentials in patients with peripheral vascular disease

  • Sarai, Pawandeep
  • Luff, Charlotte
  • Rohani-Shukla, Cyrus
  • Strutton, Paul H.

With an aging population, it is common to encounter people diagnosed with peripheral vascular disease (PVD). Some will undergo surgeries during which the spinal cord may be compromised and intraoperative neuromonitoring with motor evoked potentials (MEPs) is employed to help mitigate paralysis. No data exist on characteristics of MEPs in older, PVD patients, which would be valuable for patients undergoing spinal cord at-risk surgery or participating in neurophysiological research. Transcranial magnetic stimulation, which can be delivered to the awake patient, was used to stimulate the motor cortex of 20 patients (mean (±SD)) age 63.2yrs (±11.5) with confirmed PVD, every 10 minutes for one hour with MEPs recorded from selected upper and lower limb muscles. Data were compared to that from 20 healthy volunteers recruited for a protocol development study (28yrs (±7.6)). MEPs did not differ between patient's symptomatic and asymptomatic legs. MEP amplitudes were not different for a given muscle between patients and healthy participants. Except for vastus lateralis, disease severity did not correlate with MEP amplitude. There were no differences over time in the coefficient of variation of MEP amplitude at each time point for any muscle in patients or in healthy participants. Although latencies of MEPs were not different between patients and healthy participants for a given muscle, they were longer in older participants. The results obtained suggest PVD alone does not impact MEPs; there were no differences between more symptomatic and less symptomatic legs. Further, in general, disease severity did not corelate with MEP characteristics. With an aging population, more patients with PVD and cardiovascular risk factors will be participating in neurophysiological studies or undergoing surgery where spinal cord integrity is monitored. Our data show that MEPs from these patients can be easily evoked and interpreted.

heart health center / heart a-z list / peripheral vascular disease article

Peripheral Vascular Disease (PVD)

  • Medical Author: Melissa Conrad Stöppler, MD
  • Medical Editor: Daniel Lee Kulick, MD, FACC, FSCAI

What is peripheral vascular disease (PVD)?

Are atherosclerosis and peripheral vascular disease related, what are the symptoms of peripheral vascular disease, who is at risk for peripheral vascular disease, how does atherosclerosis cause disease, is there a test to diagnose peripheral vascular disease, what is the treatment for peripheral vascular disease, medications for peripheral vascular disease, angioplasty to treat peripheral vascular disease, surgery to treat peripheral vascular disease, what are the complications of peripheral vascular disease, preventing peripheral vascular disease.

Peripheral vascular disease (PVD) refers to diseases of the blood vessels (arteries and veins) located outside the heart and brain. While there are many causes of peripheral vascular disease, doctors commonly use the term peripheral vascular disease to refer to peripheral artery disease (peripheral arterial disease, PAD), a condition that develops when the arteries that supply oxygen-rich blood to the internal organs, arms, and legs become completely or partially blocked as a result of atherosclerosis .

Atherosclerosis is a gradual process whereby hard cholesterol substances (plaques) are deposited in the walls of the arteries. This buildup of cholesterol plaques causes hardening of the artery walls and narrowing of the inner channel (lumen) of the artery. When this happens in the peripheral circulation, peripheral vascular disease is the result. The atherosclerosis process begins early in life (as early as teens in some people). When atherosclerosis is mild and the arteries are not substantially narrowed, atherosclerosis causes no symptoms. Therefore, many adults typically are unaware that their arteries are gradually accumulating cholesterol plaques. However, when atherosclerosis becomes advanced with aging , it can cause critical occlusive disease of the arteries resulting in tissue ischemia (lack of blood and oxygen).

Arteries that are narrowed by advanced atherosclerosis can cause diseases in different organs. For example, advanced atherosclerosis of the coronary arteries (arteries that supply heart muscles) can lead to angina , coronary heart disease , and heart attacks . Advanced atherosclerosis of the carotid and cerebral arteries (arteries that supply blood to the brain) can lead to strokes and transient ischemic attacks ( TIA ). Advanced atherosclerosis in the lower extremities can lead to pain while walking or exercising ( claudication ), deficient wound healing, and/or leg ulcers.

Picture of carotid artery disease and plaque buildup.

Atherosclerosis is often generalized, meaning it affects arteries throughout the body. Therefore, patients with heart attacks are also more likely to develop strokes and peripheral vascular disease, and vice versa.

Approximately half of the people with peripheral artery disease do not experience any symptoms. For patients with symptoms, the most common symptoms are intermittent claudication and rest pain .

  • Intermittent claudication refers to arm or leg pain or cramping in the arms or legs that occurs with exercise and goes away with rest. The severity and location of the pain of intermittent claudication vary depending upon the location and extent of blockage of the involved artery. The most common location of intermittent claudication is the calf muscle of the leg, leading to calf or leg pain while walking. The pain in the calf muscle occurs only during exercise such as walking, and the pain steadily increases with continued walking until the patient has to stop due to intolerable pain. Then the pain quickly subsides during rest. Intermittent claudication can affect one or both legs.
  • Rest pain in the legs occurs when the artery occlusion is so critical that there is not enough blood and oxygen supply to the legs even at rest which represents a more serious form of the condition. The pain typically affects the feet , is usually severe, and occurs at night when the patient is lying down, face up.

Other symptoms and signs of peripheral artery disease include:

  • Numbness of the legs or feet
  • Weakness and atrophy (diminished size and strength) of the calf muscle
  • A feeling of coldness in the legs or feet
  • Changes in the color of the feet; feet turn pale when they are elevated, and turn dusky red during independent position
  • Hair loss over the top of the feet and thickening of the toenails
  • Poor wound healing in the legs or feet
  • Painful ulcers and/or gangrene in areas of the feet where blood supply is lost; typically in the toes

peripheral vascular disease research paper

Peripheral artery disease (or peripheral arterial disease) affects approximately 10 million adults in the U.S. About 5% of people over the age of 50 are believed to suffer from peripheral artery disease. Peripheral artery disease is slightly more common in men than in women and most often occurs in older persons (over the age of 50). The known risk factors for peripheral artery disease are those that predispose to the development of atherosclerosis.

Risk factors for peripheral artery disease include:

  • High blood cholesterol (elevated levels of the "bad" LDL cholesterol and triglycerides
  • Low blood levels of the "good" HDL cholesterol
  • Cigarette smoking
  • Diabetes mellitus (both type 1 and type 2 diabetes )
  • High blood pressure ( hypertension ) or a family history of hypertension
  • A family history of atherosclerotic disease
  • Chronic renal failure
  • Overweight or obesity
  • Physical inactivity

In peripheral artery disease, the risk factors are additive, so a person with a combination of two risk factors -- diabetes and smoking , for example -- has an increased likelihood of developing more severe peripheral artery disease than a person with only one risk factor.

Health News

  • Taking a GLP-1 Medicine? Here's What Experts Say You Should Eat
  • FDA Gives Nod to RSV Vaccine for People in Their 50s
  • Study Supports Safety of High-Dose General Anesthesia
  • Glowing Dye Helps Surgeons Track & Destroy Prostate Cancer
  • Many Louisiana Residents May Be Exposed to Sky-High Levels of Toxic Gas

Atherosclerosis causes disease in two ways. 1) Atherosclerosis can limit the ability of the narrowed arteries to increase delivery of blood and oxygen to the tissues of the body during times when oxygen demand needs to be increased, for example, during exertion; or 2) complete obstruction of an artery by a thrombus or embolus (thrombus and embolus are forms of blood clots ), which results in tissue death (necrosis). Exertional angina and intermittent claudication are two examples of insufficient delivery of blood and oxygen to meet tissue demand; whereas strokes and heart attacks are examples of death of tissue caused by complete artery obstruction by blood clots.

There are many similarities between coronary heart disease (or coronary artery disease , which is atherosclerosis involving the arteries of the heart) and peripheral artery disease, and the two conditions may coexist in the same individual. For example, patients with exertional angina typically have no symptoms at rest. However, during exertion, the critically narrowed coronary arteries are incapable of increasing blood and oxygen delivery to meet the increased oxygen needs of the heart muscles. Lack of blood and oxygen causes chest pain (exertional angina). Exertional angina typically subsides when the patient rests. In patients with intermittent claudication, the narrowed arteries in the lower extremities (for example, a narrowed artery at the groin) cannot increase blood and oxygen delivery to the calf muscles during walking. These patients experience pain in the calf muscles that will only subside after resting.

Patients with unstable angina have critically narrowed coronary arteries that cannot deliver enough blood and oxygen to the heart muscle even at rest. These patients have chest pain at rest and are at imminent risk of developing heart attacks. Patients with severe artery occlusion in the legs can develop rest pain (usually in the feet). Rest pain represents such severe occlusion that there is the insufficient blood supply to the feet even at rest. They are at risk of developing foot ulcers and gangrene.

When atherosclerosis narrows the arteries, blood tends to clot in the narrowed areas, forming a thrombus, a type of blood clot (plural thrombi). Sometimes pieces of the blood clot break off and travel in the bloodstream until they are trapped in a narrower point in the artery beyond which they cannot pass. A thrombus or piece of thrombus that travels to another point is called an embolus. Thrombi and emboli can cause sudden and complete artery blockage, leading to tissue necrosis (death of tissue due to lack of oxygen).

For example, complete blockage of a coronary artery by a thrombus causes heart attack , while complete blockage of the carotid or cerebral artery causes ischemic stroke . Emboli originating from atherosclerosis in the aorta (the main artery delivering blood to the body) can obstruct small arteries in the feet, resulting in painful and blue (cyanotic) toes, foot ulcers, and even gangrene.

What are collateral vessels or collateral circulation?

Sometimes, despite the presence of a severe blockage in an artery, the involved area does not become painful or lose its blood supply due to the presence of collateral vessels. Collateral circulation means that the particular area is supplied by more than one artery so that the blockage of a single vessel does not result in a severe degree of blood loss. Collateral circulation can develop over time to help provide oxygenated blood to an area where an artery is narrowed. Doctors believe that regular supervised exercise can stimulate the growth and development of collateral circulation and relieve symptoms of intermittent claudication.

What are the other causes of peripheral vascular diseases?

Several conditions such as vasculitis may cause damage to blood vessels throughout the body. Injuries to blood vessels (from accidents such as auto accidents or sports injuries), blood-clotting disorders, and damage to blood vessels during surgery can also lead to the inadequate blood supply to body tissues (ischemia).

Tissue ischemia can also occur in the absence of atherosclerosis or other abnormalities of arteries. One example of a condition in which the blood vessels themselves are not damaged is Raynaud's disease , which is believed to occur due to spasms in blood vessels brought on by stress or a cold environment.

Since atherosclerosis of the peripheral arteries (PAD) is by far the most common cause of peripheral vascular disease, the rest of this article focuses on peripheral artery disease.

The doctor will perform a physical exam and look for signs and symptoms of peripheral artery disease, for example, weak or absent artery pulses in the extremities, bruits (sounds that can be heard through a stethoscope), blood pressure changes, and skin color and nail changes

In addition to the history of symptoms and the physical signs of peripheral artery disease imaging tests can be used in the diagnosis of the condition. Imaging tests include:

  • Doppler ultrasound - This form of ultrasound (measurement of high-frequency sound waves that are reflected off of tissues) can detect and measure blood flow. Doppler ultrasound is used to measure blood pressures behind the knees and at the ankles. In patients with significant peripheral artery disease in the legs, the blood pressure in the ankles will be lower than the blood pressure in the arms (brachial blood pressure). The ankle-brachial index (ABI) is a number derived from dividing the ankle blood pressure by the brachial blood pressure. An ankle-brachial index of 0.9 to 1.3 is normal, an ABI less than 0.9 indicate the presence of peripheral artery disease in the arteries in the legs, and an ABI below 0.5 usually indicates severe arterial occlusion in the legs.
  • Duplex ultrasound - This is a color-assisted non-invasive technique to study the arteries. Ultrasound probes can be placed on the skin overlying the arteries and can accurately detect the site of artery stenosis as well as to measure the degree of obstruction.
  • Angiography - An angiography is an imaging procedure to study the blood vessels of the extremities, similar to the way a coronary angiogram provides an image of the blood vessels supplying the heart. It is the most accurate test to detect the location(s) and severity of artery occlusion, as well as collateral circulations. Small hollow plastic tubes (catheters) are advanced from a small skin puncture at the groin (or the arm), under X-ray guidance, to the aorta and the arteries. Iodine contrast "dye," is then injected into the arteries while an X-ray video is recorded. Angiogram gives the doctor a picture of the location and severity of narrowed artery segments. This information is important in helping the doctor select patients for angioplasty or surgical bypass (see below).
  • Because X-ray angiography is invasive with potential side effects (such as injury to blood vessels and contrast dye reactions), it is not used for the initial diagnosis of peripheral artery disease. It is only used when a patient with severe peripheral artery disease symptoms is considered for angioplasty or surgery. Several different imaging methods have been used in angiography examinations, including X-rays , magnetic resonance imaging ( MRI ), and computed tomography (CT) scans.
  • Magnetic resonance imaging (MRI) angiography uses magnetism, radio waves, and a computer to produce images of body structures and has the advantage of avoiding X-ray radiation exposure.

Subscribe to MedicineNet's General Health Newsletter

By clicking Submit, I agree to the MedicineNet's Terms & Conditions & Privacy Policy and understand that I may opt out of MedicineNet's subscriptions at any time.

Treatment goals for peripheral artery disease include:

  • Relieve the pain of intermittent claudication.
  • Improve exercise tolerance by increasing the walking distance before the onset of claudication.
  • Prevent critical artery occlusion that can lead to foot ulcers, gangrene, and amputation.
  • Prevent heart attacks and strokes.

Treatment of peripheral artery disease includes lifestyle measures, supervised exercises, medications, angioplasty, and surgery.

Lifestyle changes

  • Smoking cessation eliminates a major risk factor for disease progression, and it lowers the incidences of pain at rest and amputations. Smoking cessation also is important to prevent heart attacks and strokes.
  • A healthy diet can help lower blood cholesterol and other lipid levels and may help control blood pressure.
  • Keep other risk fa ctors, such as diabetes, lipid levels, and blood pressure under control by following medical advice regarding medications and lifestyle changes.

Supervised exercise

Proper exercise can condition the muscles to use oxygen effectively and can speed the development of collateral circulation. Clinical trials have shown that regular supervised exercise can reduce symptoms of intermittent claudication and allow individuals to walk longer before the onset of claudication. Ideally, your health care provider should prescribe an exercise program tailored to your specific needs.

Rehabilitation programs supervised by healthcare professionals such as nurses or physical therapists may help. Exercise at least three times a week, each session lasting longer than 30 to 45 minutes for the best results. Exercise usually involves walking on a monitored treadmill until claudication develops; walking time is then gradually increased with each session. Patients are also monitored for the development of chest pain or heart rhythm irregularities during exercise.

While lifestyle changes may be enough treatment for some people with peripheral artery disease, others may require medication. Examples of medications used to treat peripheral artery disease include antiplatelet or anti-clotting agents, cholesterol-lowering drugs such as statins , and medications that increase blood supply to the extremities such as cilostazol ( Pletal ) and pentoxifylline ( Trental ), and medications that control high blood pressure .

  • Antiplatelet medications (such as aspirin and clopidogrel [ Plavix ]) make the blood platelets less likely to stick to one another to form blood clots. Low-dose aspirin (81 to 325 mg/day) is usually prescribed indefinitely because it is also helpful in preventing strokes and heart attacks in patients with peripheral artery disease. Clopidogrel (Plavix) is an alternative to aspirin for those who are allergic or cannot tolerate aspirin. Antiplatelet medications also help prevent the occlusion of blood vessels after angioplasty or bypass surgery.
  • Anticoagulant medications act to prevent blood clotting. Both heparin and warfarin ( Coumadin , Jantoven ) are anticoagulant medications. Anticoagulants are sometimes prescribed for people with peripheral artery disease if they are at increased risk for the formation of blood clots; these agents are used much less frequently than antiplatelet agents in patients with peripheral artery disease.
  • Cholesterol-lowering drugs of the statin class have been shown in numerous large clinical trials to help prevent heart attacks and strokes and prolong survival among patients with atherosclerosis. Statins have also been shown to slow the progression of peripheral artery disease, decrease atherosclerosis in the arteries, and improve claudication symptoms.
  • Cilostazol (Pletal) is a medication that can help increase physical activity (enabling one to walk a greater distance without the pain of claudication). Cilostazol works by causing dilation of the arteries and an increased supply of oxygenated blood to be delivered to the arms and legs. Cilostazol is recommended for some patients with claudication when lifestyle modifications and exercise are ineffective. Side effects are generally mild and include headache , diarrhea , and dizziness . Cilostazol should not be used in patients with heart failure because of concern over increased mortality in heart failure patients using medications similar to cilostazol.
  • Pentoxifylline (Pentoxyl, Trental) improves blood flow to the extremities by decreasing the viscosity ("stickiness") of blood, enabling more efficient blood flow. Side effects are fewer than with cilostazol, but its benefits are weaker and have not been conclusively proven by all studies.
  • Dru gs to control hypertension may also be prescribed. Current recommendations are to treat hypertension in patients with peripheral artery disease to prevent strokes and heart attacks.

Angioplasty, also known as percutaneous transluminal angioplasty, or PTA, is a non-surgical procedure that can widen a narrowed or blocked artery. A thin tube (catheter) is inserted into an artery in the groin or arm and advanced to the area of narrowing. A tiny balloon on the tip of the catheter is then inflated to enlarge the narrowing in the artery. This procedure is also commonly performed to dilate narrowed areas in the coronary arteries that supply blood to the heart muscle.

Sometimes the catheter technique is used to insert a stent (a cylindrical wire mesh tube) into the affected area of the artery to keep the artery open. In other cases, thrombolytic medications (medications that dissolve blood clots) may be delivered to the blocked area via a catheter.

Angioplasty does not require general anesthesia. Usually, a local anesthetic at the area of catheter insertion and a mild sedative is given. Major complications of angioplasty are rare but can occur. These include damage to the artery or blood clot formation, excessive bleeding from the catheter insertion site, and abrupt vessel closure (blockage of the treated area occurring within 24 hours of the procedure).

Despite these risks, the overall incidence of complications is low and the benefits of angioplasty (no general anesthesia, no surgical incision, and the ability to return to normal activities within a couple of days) outweigh its risks. Usually, a one-night hospital stay is required when angioplasty is performed.

Angioplasty is used when a patient has claudication that limits his or her activities and does not respond to exercise medications, and lifestyle measures. Most doctors also recommend angioplasty when the disease is very severe, and there is a focal, localized narrowing that is accessible via a catheter. If a patient is too ill to have surgery and has severe ischemia (decreased oxygen in the tissues) that threatens the loss of a limb, angioplasty may also be attempted.

Some cases of peripheral artery disease may be more difficult to treat by angioplasty. For example, blockages in multiple small arteries of the legs or blockages in extremely small vessels may not be treatable by this method.

Surgical treatment for peripheral artery disease involves either bypass vascular surgery performed by a vascular surgeon or an endarterectomy. Indications for surgical treatment of peripheral artery disease include lesions that, for anatomical reasons, may be difficult to treat by angioplasty. Examples include lesions covering long segments of a vessel, vessels with multiple narrowed areas, or long areas of narrowing. Bypass surgery involves using a vein from your body or a portion of a synthetic vessel (known as grafts) to create a detour around the blockage. One end of the graft is sewn to the damaged artery above the blockage and the other end is sewn below the blocked area. Blood flow is then able to bypass the area of narrowing or blockage Bypass surgery is a major surgical procedure requiring general anesthesia and a hospital stay.

Endarterectomy is a procedure in which the surgeon cleans out plaque buildup inside the artery of the affected leg or arm.

In rare cases, the decreased circulation to the extremities characteristic of peripheral artery disease can lead to open, non-healing sores, ulcers, gangrene, or other limb-threatening injuries to the extremities. The areas that do not receive adequate blood flow are also more prone to develop infections and, in extreme cases, amputation may be necessary.

Peripheral vascular disease related to atherosclerosis can be prevented by minimizing the risk factors that are controllable, such as eating a heart-healthy diet , maintaining a h ealthy weight, not smoking, getting regular exercise, and maintaining good control of blood sugar levels if you have diabetes.

Top Peripheral Vascular Disease Related Articles

peripheral vascular disease research paper

Heart Healthy Diet: 25 Foods You Should Eat

peripheral vascular disease research paper

Blood Clots (in the Leg)

peripheral vascular disease research paper

C-Reactive Protein (CRP) Test

peripheral vascular disease research paper

CT Scan (Computerized Tomography)

peripheral vascular disease research paper

Chest pain is a common complaint by a patient in the ER. Causes of chest pain include broken or bruised ribs, pleurisy, pneumothorax, shingles, pneumonia, pulmonary embolism, angina, heart attack, costochondritis, pericarditis, aorta or aortic dissection, and reflux esophagitis.

Diagnosis and treatment of chest pain depends upon the cause and clinical presentation of the patient's chest pain.

peripheral vascular disease research paper

Cholesterol Levels: What's Normal and How to Lower High Cholesterol

peripheral vascular disease research paper

Cholesterol Test

peripheral vascular disease research paper

CT Scan vs. MRI

peripheral vascular disease research paper

CAD Slideshow

peripheral vascular disease research paper

Heart Disease: Causes of a Heart Attack

peripheral vascular disease research paper

Heart Disease Quiz

peripheral vascular disease research paper

Illustrations of the Heart

peripheral vascular disease research paper

Cholesterol Quiz

peripheral vascular disease research paper

Kidney Failure

peripheral vascular disease research paper

Muscle Cramps

peripheral vascular disease research paper

Salt & Low Sodium Diet Quiz

peripheral vascular disease research paper

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

Preview improvements coming to the PMC website in October 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • Oxford University Press - PMC COVID-19 Collection

Logo of pheoup

Progress in aorta and peripheral cardiovascular disease research

Lucia mazzolai.

1 Division of Angiology, Heart and Vessel Department, Lausanne University Hospital (CHUV), Lausanne, Switzerland

Adriano Alatri

Alessandra bura rivière.

2 Division of Vascular Medicine, Heart, Vessels and Metabolisms Department, Toulouse University Hospital, Toulouse, France

Marco De Carlo

3 Cardiothoracic and Vascular Department, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy

Christian Heiss

4 Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK

Christine Espinola-Klein

5 Section Angiology, Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany

Oliver Schlager

6 Division of Angiology, Department of Medicine II, Medical University of Vienna, Waehringer Gürtel 18-20, 1090 Vienna, Austria

Henrik Sillesen

7 Department of Vascular Surgery, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark

Daniel Staub

8 Division of Angiology, University Hospital Basel, University of Basel, Basel, Switzerland

José F Rodriguez-Palomares

9 Cardiology Department, Hospital General Universitari Vall d’Hebron, Vall d’Hebron Research Institute (VHIR), CIBER-CV, Barcelona, Spain

Aline Verstraeten

10 Centre of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium

Victor Aboyans

11 Department of Cardiology, Dupuytren-2 University Hospital, and Inserm 1094 & IRD, Limoges University, Limoges, France

Although coronavirus disease 2019 seems to be the leading topic in research number of outstanding studies have been published in the field of aorta and peripheral vascular diseases likely affecting our clinical practice in the near future. This review article highlights key research on vascular diseases published in 2020. Some studies have shed light in the pathophysiology of aortic aneurysm and dissection suggesting a potential role for kinase inhibitors as new therapeutic options. A first proteogenomic study on fibromuscular dysplasia (FMD) revealed a promising novel disease gene and provided proof-of-concept for a protein/lipid-based FMD blood test. The role of NADPH oxidases in vascular physiology, and particularly endothelial cell differentiation, is highlighted with potential for cell therapy development. Imaging of vulnerable plaque has been an intense field of research. Features of plaque vulnerability on magnetic resonance imaging as an under-recognized cause of stroke are discussed. Major clinical trials on lower extremity peripheral artery disease have shown added benefit of dual antithrombotic (aspirin plus rivaroxaban) treatment.

1. Introduction

The year 2020 was overshadowed by the spread of the novel coronavirus disease 2019 (COVID-19). Four months after the first cases in China, Varga et al. 1 described the involvement of the vascular endothelium in patients with COVID-19.

Nevertheless, the year 2020 was also characterized by outstanding publications in the field of aortic and peripheral vascular diseases, potentially improving the clinical management in the future.

This article summarizes the main findings from key studies published in 2020 on the basic science, epidemiology, imaging, and clinical trials addressing aortic and peripheral arterial and venous diseases. A specific chapter on vascular complications secondary to COVID-19 infection is also addressed. For this review, the authors performed a systematic review of published research papers in major vascular areas (arterial, venous, aorta, basic science, clinical trials, and COVID). The final selection was discussed within the group through a consensus.

2. Basic science

2.1 aortic aneurysm and dissection.

In 2020, multiple putative aortic aneurysm and dissection (AoAD) therapeutical targets were identified, two of which are detailed here.

In two small-scale case–control cohorts, a common missense variant in the alcohol-metabolizing aldehyde dehydrogenase 2 protein (ALDH2 p.Glu504Lys; gnomAD v2.1.1 MAF total : 1.8% vs. MAF Asians : 10.1%) previously linked to increased coronary artery disease risk, was suggested to be associated with protection from AoAD. 2 Further proof of a direct relationship between ALDH2 activity and AoAD risk was next pursued in two different AoAD mouse models. In angiotensin II- or 3-aminopropionitrile fumarate-infused mice, ALDH2 inhibition by the isoflavone daidzin was shown to partially rescue the aortopathy phenotype as demonstrated by mitigation of elastic fibre fragmentation, near-normalization of aortic wall thickness, and a concomitant reduction in the incidence of AoAD. 2 Further disentanglement of molecular mechanisms underlying this protective effect exposed a critical role for miR-31-5p-dependent inhibition of the pathological contractile-to-synthetic vascular smooth muscle cell (VSMC) phenotype switch. Specifically, ALDH2 loss-of-function represses the expression of miR-31-5p, which results in elevated myocardin levels and, consequently, increased expression of the VSMCs’ contractile apparatus genes (i.e. α-SMA, SM22-α, and calponin). 2

AoAD is also an important complication of the vascular Ehlers–Danlos syndrome (vEDS), caused by heterozygous mutations in the collagen Type III alpha 1 chain ( COL3A1 ) gene. Contrary to some other heritable aortopathies (e.g. Marfan syndrome), vEDS-related dissections or ruptures can occur without prior aneurysm formation, and involve also muscular arteries. Bowen et al. 3 created two vEDS mouse models ( Col3a1 G209S/+ and Col3a1 G938D/+ ) corresponding to known human vEDS-causing mutations and presenting with sudden death due to aortic dissection or rupture in the absence of prior dilatation and major architectural wall deterioration. Comparative aortic transcriptome analysis of mutant mice and their wild-type (WT) littermates suggested excessive signalling through the PLC/IP3/PKC/ERK axis as a key disease culprit. Whereas administration of blood pressure-lowering agents such as beta-blockers did not improve survival rates, pharmacological treatment with ERK1/2 or PKCβ inhibitors (i.e. riboxistaurin, cobimetinib, and hydralazine) improved survival from 52% to 90–97%, conferring considerable translational weight to the transcriptome findings. Similar to humans, pregnancy/lactation- and male puberty-associated exacerbation of aortic dissection/rupture risk were observed in the vEDS mice, all of which could be rescued by oxytocin and androgen signalling attenuation, respectively. Altogether, these studies put forward ALDH2, miR-31-5p, and the PLC/IP3/PKC/ERK axis as novel targets for AoAD prevention and, in the latter case, emphasized the role of androgen signalling and breastfeeding (oxytocin) in vEDS-related aortic dissection/rupture risk.

Niacin is known to prevent atherosclerosis via anti-inflammatory effects by activating the G protein-coupled receptor GPR109A on immune cells. An experimental study in mice indicates now that niacin could prevent abdominal aortic aneurysm (AAA) development independent of GPR109A. 4 The authors show that niacin markedly blunted AAA formation in two mouse models (angiotensin II and CaCl 2 ) with lowered inflammatory responses and matrix degradation. Importantly, deletion of GPR109A gene did not prevent the protective effects. Nicotinamide led to very similar results with increases in NAD+ concentrations and Sirt1 activity suggesting that both niacin and nicotinamide could become novel therapeutic agents to prevent AAA. Nicotinamide or related molecules may have the advantage of not causing flushing, a side effect of niacin linked to GPR109A.

2.2 Fibromuscular dysplasia

Fibromuscular dysplasia (FMD) affects predominantly middle-aged women and causes stenosis, tortuosity, aneurysm, and/or dissection of medium-sized arteries. It is often asymptomatic, and its aetiology poorly understood. A first FMD plasma proteomics and lipidomics study in 90 multifocal FMD patients and 100 age/sex-matched control individuals revealed differential abundance of 105 proteins and 16 lipid sub-classes (particularly triglycerides and fatty acids). 5 Of these, 37 proteins and 10 lipid sub-classes were confirmed as being up- or downregulated in an independent validation cohort of 23 FMD patients and 28 controls. Using machine learning approaches, a combined protein and lipid signature reaching, respectively, a sensitivity and specificity of 78.3% and 64.3% was developed. Additionally, protein quantitative trait locus mapping and subsequent Bayesian network classification suggested CD2AP, PODXL2, and TACC3 to be upstream of FMD development and, therefore, to be candidate disease drivers. Moreover, an independent genetic association study in 506 FMD patients and 876 healthy individuals revealed a significant relationship between single-nucleotide polymorphisms in the upstream region of CD2AP expressed by endothelial cells of medium-sized arteries. 5 Like PODXL2, CD2AP has been suggested to play a role in vascular leucocyte adhesion and rolling, and increased FMD risk [top-hit: rs9296551; ∼odds ratio (OR) 1.36]. This first proteogenomic FMD study revealed a promising novel disease gene and provided proof-of-concept for a protein/lipid-based FMD blood test.

2.3 Atherosclerosis

Chronic inflammation and autoimmunity play important roles in the atherosclerosis development and stability of atherosclerotic plaques. Recently, relevance of co-stimulatory immune checkpoint protein glucocorticoid-induced tumour necrosis factor receptor family-related protein (GITR) in atherogenesis has been shown. 6 GITR is known for activating and regulating effects on T cells. A novel role of GITR is proposed, in driving myeloid cell recruitment and activation in atherosclerosis, thereby inducing plaque growth and vulnerability. GITR expression was elevated in carotid endarterectomy specimens from 100 patients with symptomatic carotid disease vs. those extracted from asymptomatic patients ( n  = 93). GITR was essentially found in macrophages, endothelial cells, and T cells, and similar patterns were also observed in femoral endarterectomy samples. 6 GITR expression correlated with signs of plaque vulnerability. Furthermore, patients with cardiovascular (CV) disease showed elevated soluble plasma GITR levels compared to healthy controls. In 28-week-old Gitr −/− Apoe −/− mice extension of aortic atherosclerosis was reduced, and plaques showed a more stable phenotype with fewer macrophages, smaller necrotic core, and a thicker fibrous cap. Lymphocytes were not affected by GITR deficiency. Monocyte and macrophage cell migration, activation, and mitochondrial function were differently modulated in Apoe −/− and Gitr −/− Apoe −/− mice. In Gitr −/− Apoe −/− mice, monocytes showed decreased integrins and reactive oxygen species (ROS) levels as well as reduced endothelium recruitment. Along the same line, macrophages showed reduced migratory capacity and lower cytokines production. Altogether ( Figure  1 ), these data indicate that GITR plays a pivotal role and is a potential therapeutic target in atherosclerosis.

An external file that holds a picture, illustration, etc.
Object name is cvab144f1.jpg

GITR-mediated atherogenesis (reproduced with permission from Shami et al. 6 ). GITR, glucocorticoid-induced tumour necrosis factor receptor family-related protein.

Cell membrane exteriorization of phosphatidylserine and phosphatidylethanolamine (PE) occurring during apoptosis within plaques contributes to several high-risk features of vulnerable plaque. Recently, molecular imaging with PE-avid radiolabeled oxytetracycline in experimental atherosclerotic lesions was investigated in rabbits. 7 The 99m Tc-oxytetracycline uptake was >two-fold higher in the test group of 21 rabbits on high-fat diet for 16 weeks vs. 6 rabbits on normal chow (controls) and 6 negative radiotracer control animals ( 99m Tc-linear oxytetracycline without PE-binding capability). On histology, oxytetracycline uptake correlated to lesion severity and macrophage burden. This study represents a notable step towards atherosclerosis molecular imaging using radiolabeled oxytetracycline to localize lipid-rich areas with high levels of apoptotic macrophages in an experimental model. Further clinical studies in humans are awaited to detect vulnerable high-risk plaques.

A few interesting papers highlighted the important and complex role of NADPH oxidases in vascular physiology beyond their role as ROS producers and atherosclerosis mediators. A review of a large body of literature shows that endothelial cell differentiation requires ROS and NADPH oxidases (Nox1, 2, 4, and 5) and these are important local modulators of the signalling networks regulating differentiation of stem cells to endothelial cells. 8

Understanding the specific roles of NADPH oxidases may also help to further develop cell-based therapies. For instance, in cord blood-derived endothelial colony-forming cells, Nox4 could be a future therapeutic target as it enhances the reparative functions of these cells supporting the creation of a pro-reparative microenvironment and effective post-ischaemic revascularization. 9 The requirement for Nox4-derived ROS in vivo was highlighted by a mouse study demonstrating that Nox4-derived H 2 O 2 plays a key role in exercise-induced vascular adaptations. 10 Exercise led to an increased H 2 O 2 release in the aorta of WT mice with adaptations of the eNOS and Ppargc1a pathway, and intracellular calcium release. In Nox4 −/− mice, the physical activity performance and vascular protective effects of exercise were inhibited.

3. Epidemiology and prevention

A recent analysis from the Framingham Heart Study and the Danish nationwide administrative registries showed strong familial association of aortic aneurysm. 11 Children of parents with aorta sized in the upper quartile (adjusted for age, sex, and body surface area) had a three-fold increased risk of being themselves in that aortic diameter upper quartile. Additionally, first-degree relatives of patients with ascending aortic aneurysm had a 6.7-fold increased risk in developing an ascending aortic aneurysm, and a 9.2-fold risk for aortic dissection. These observations support the use of systematic screening for aortic diseases in affected families. 12

Patients with lower extremity artery disease (LEAD) have a high risk for major adverse cardiovascular events (MACE) and major adverse limb events (MALE). Recent analysis of UK electronic health reports showed a 15% decrease in LEAD incidence between 2006 and 2015 [from 236 to 202 per 100 000 person-years; adjusted incidence rate ratio (IRR) 0.85, 95% confidence interval (CI) 0.82–0.88]. However, CV mortality for incident LEAD did not decline significantly (adjusted IRR 0.84, 95% CI 0.70–1.00), at variance with the significant 43% fall in mortality for incident coronary artery disease. 13

New insights on the pathophysiological mechanisms underlying the dismal prognosis of patients with chronic limb-threatening ischaemia come from a study demonstrating the association between CD34+ cell migration and long-term CV mortality. In coculture, CD34+ cells imprinted naive endothelial cells, increasing apoptosis and reducing network formation. 14 An altered paracrine signalling from CD34+ cells to the endothelium may contribute to the increased CV risk in these patients.

The lack of improvement in CV prognosis of LEAD patients extends also to limb prognosis, as demonstrated by a new analysis from the 2017 Global Burden of Disease study. 15 Data from 16 European countries, Canada, Australia, and the USA between 1990 and 2017 showed wide time trend variability among countries and between sexes regarding amputation proximal to toes, in the absence of uniform improvements. New data on the impact of revascularization on limb prognosis in claudicants were reported in a retrospective analysis of 11 887 elective endovascular procedures. 16 One-year amputation rate was 1.1%; independent predictors of major amputation were congestive heart failure (OR 6.5, 95% CI 2.4–17.2), American Society of Anesthesiologists Class IV (OR 9.3, 95% CI 1.9–44.9), non-white race (OR 3.3, 95% CI 1.5–7.4), and tibial-level intervention (OR 6.3, 95% CI 1.5–26.1).

Women were previously considered to have poorer prognosis when affected by LEAD, but this has been contradicted by two recent post hoc analyses of large randomized-controlled studies (RCTs). In the EUCLID (Examining Use of Ticagrelor in PAD) trial, women with LEAD were at lower risk for MACE compared with men [9.5% vs. 11.2%; adjusted hazard ratio (HR) 0.77; P  < 0.001], but had similar rates of MALE (2.6% vs. 3.0%; adjusted HR 0.90; P  = 0.37). 17 Similarly, in the COMPASS (Cardiovascular Outcomes for People Using Anticoagulant Strategies) trial, women had similar rates for MACE and major bleeding. There were no sex-related interaction regarding the benefits in reducing MACE under low-dose rivaroxaban plus aspirin vs. aspirin alone (women: HR 0.72; men: HR 0.76; P interaction = 0.75) or major bleeding (women: HR 2.22; men: HR 1.60; P interaction = 0.19). 18

Benefits of a more potent antithrombotic therapy in LEAD patients were recently confirmed in a meta-analysis encompassing seven RCTs comparing various antithrombotic regimens. 19 More vs. less intense antithrombotic therapy reduced significantly the risk of limb revascularization, limb amputation, and stroke ( Figure  2 ), without significant effects on myocardial infarction and CV death, but at cost of increased risk of (major) bleeding, highlighting the importance of individualized risk/benefit assessment.

An external file that holds a picture, illustration, etc.
Object name is cvab144f2.jpg

Forest plot of risk of adverse events with more intense vs. less intense antithrombotic therapy in patients with lower extremity artery disease. The 95% CIs are denoted by lines (modified from Savarese et al. 19 ). CI, confidence interval; CV, cardiovascular.

The largest diameter of the aorta is considered as the main risk factor for aortic rupture and dissection and, therefore, this criterion is recommended to guide interventions. However, the measurement of aortic diameter (especially at the aortic root) has multiple limitations. Several imaging techniques and measurement protocols exist suggesting that established thresholds of aortic diameters might not be fully adequate. Therefore, it is highly recommended to be very exhaustive when performing aortic measurements, indicating in detail the methodology used. 20

To overcome these limitations, new parameters have been recently introduced. Ascending aortic length (length > 13 cm) has been associated with a five-fold increase in aortic events (rupture, dissection, or death). 21 Furthermore, increased aortic stiffness (aortic strain reduction) correlated with higher incidence of dissection or surgery in Marfan patients. 22 A recent study showed that both ascending aortic length and volume improve prediction of aortic dissection in case of ascending aortic aneurysms. 23 Patients ( n  = 25) with type A acute aortic dissection who had a computed tomography (CT) within the prior 2 years were compared to those with thoracic aortic aneurysm and no-acute aortic dissection ( n  = 75), and healthy controls ( n  = 258). Aortic diameter was similar between patients with and without acute aortic dissection (45 vs. 46 mm, respectively, P  = 0.075), as well as aortic volume (126 vs. 124 cm 3 , respectively, P  = 0.909) with differences in aortic length (90 vs. 84 mm, respectively, P  = 0.031). Aortic volume and length showed, respectively, a five-fold and a seven-fold sensitivity in predicting acute aortic dissection while the aortic length presented 70% positive predictive value.

Not only the carotid artery stenosis severity, but also the atherosclerotic plaque composition and morphology are increasingly recognized as important features determining stroke risk. High-resolution, contrast-enhanced carotid magnetic resonance imaging (MRI) enables non-invasive characterization of carotid artery plaques (CAP), including features of plaque vulnerability as intraplaque haemorrhage, thin and/or ruptured fibrous cap, large lipid-rich and/or necrotic core, and mural thrombus. The CAPIS study investigated a causal role between complicated CAP and specific MRI plaque features with cryptogenic stroke. 24 Using a subtle prospective study, 234 patients with acute ischaemic stroke restricted to single carotid artery territory on brain MRI, and unilateral or bilateral CAP were recruited. Among patients with cryptogenic stroke ( n  = 104), prevalence of ipsilateral complicated CAP on MRI was significantly higher ipsilateral (31%) vs. contralateral to the infarct (12%; P  = 0.0005). The prevalence of ipsilateral complicated CAP in cryptogenic stroke was also significantly higher than in patients with cardioembolic or small vessel stroke (15%; P  = 0.02), but significantly lower than in those with large artery stroke and ipsilateral 50–69% carotid stenosis (68%; P  = 0.003). These finding substantiate the role of complicated non-stenotic CAP with imaging features of plaque vulnerability on MRI as an under-recognized cause of stroke. Further studies are needed to determine a possible interventional approach in patients with such lesions.

Non-invasive assessment of limb perfusion and its impact in predicting wound healing and amputation outcome could be of particular importance in patients with critical limb ischaemia (CLI). Different methods for skin or muscle perfusion imaging have been developed, using hyperspectral, laser Doppler, MR-based methods, and contrast-enhanced ultrasound techniques, with poor clinical application. In a prospective study of 25 patients with diabetes and CLI, pedal perfusion was assessed before and after revascularization, using 99m Tc-tetrofosimin single-photon-emission computed tomography (SPECT)/CT perfusion imaging of segmented angiosomes of the foot. 25 SPECT/CT detected a significantly lower regional microvascular perfusion response in patients undergoing amputation compared to those with saved limbs at 3 and 12 months after revascularization. The amputation-free survival rate was significantly higher at 3 and 12 months for high-perfusion than low-perfusion responders to revascularization. Therefore, SPECT/CT imaging could provide new prognostic information on regional perfusion response to lower extremity revascularization.

5. Clinical trials

Several clinical trials have been published in 2020 ( Table  1 ). The TEDY study investigated whether telmisartan 40 mg daily could slow small AAAs growth, as measured by ultrasound and CT scanning. 26 Among the 207 patients included in the intention-to-treat analysis, no significant difference in ultrasound-assessed AAA growth rates was found among those under telmisartan (1.68 mm/year) vs. placebo (1.78 mm/year, P  = 0.66). Similarly, telmisartan did not significantly affect AAA growth assessed by CT-measured AAA diameter or volume.

Summary of major clinical trials in the field of aorta and peripheral vascular diseases in 2020

TrialType and aimComparison Setting (indication)Primary endpointMain hypothesis validated?
Aorta
 TEDY Multicentre RCT to assess the effect of telmisartan on aneurysm growth in patients with AAATelmisartan vs. placebo210Patients with AAA (diameter 35–49 mm)Between treatment group difference in AAA growth over 2 yearsNo: [−0.11 mm (−0.60 to 0.38),  = 0.66]
 TicAAA Multicentre RCT to assess the effect of ticagrelor on aneurysm growth in patients with AAATicagrelor vs. placebo144Patients with AAA (diameter 35–49 mm)Between treatment group difference in change of AAA volume (MRI measurement) after 1 yearNo: [baseline-adjusted mean ratio (ticagrelor/placebo) 1.013 (0.993–1.034),  = 0.205]
Peripheral vascular diseases
 VOYAGER PAD Multicentre RCT to assess dual pathway inhibition after peripheral revascularization in LEADAspirin + rivaroxaban vs. aspirin + placebo6564Symptomatic LEAD with peripheral revascularizationALI, vascular-related major amputation, MI, ischaemic stroke, or cardiovascular deathYes: HR 0.85 (0.76–0.96),  = 0.0085
SWEDEPAD Multicentre open-label, registry-based RCT to assess all-cause mortality after the use of paclitaxel-coated devices after peripheral revascularization in LEADPaclitaxel-coated devices vs. uncoated devices2289Symptomatic LEAD with peripheral revascularizationAll-cause mortalityNo: HR 1.06 (0.92–1.22)
 ODYSSEY OUTCOMES (prespecified analysis)Prespecifed analysis of multicentre RCT to assess the effect of alirocumab on the occurrence of LEAD events and VTEAlirocumab vs. placebo18 924Patients with recent ACS and elevated levels of lipoproteins despite intensive or maximum-tolerated statin treatmentOccurrence of LEAD events (critical limb ischaemia, limb revascularization, amputation for ischaemia) and VTE (DVT or PE) within the trial period (median duration of follow-up: 2.8 years)Yes: combined endpoint LEAD + VTE HR 0.69 (0.55–0.86),  < 0.001 (LEAD only HR 0.69 (0.54–0.89),  = 0.004; VTE only HR 0.67 (0.44–1.01),  = 0.06)
 FOURIER ( analysis) analysis of multicentre RCT to assess the effect of evolocumab on the occurrence of VTEEvolocumab vs. placebo27 564Patients (≥40 to ≤85 years of age) with cardiovascular disease and additional predefined cardiovascular risk factorsOccurrence of DVT or PE within the trial period (median duration of follow-up: 2.2 years)Yes: HR 0.71 (0.50–1.00),  = 0.05
 CARAVAGGIO Multicentre RCT to compare the efficacy of apixaban with dalteparin in the risk reduction of recurrent VTE in cancerApixaban vs. dalteparin (non-inferiority of apixaban)1170Cancer patients with symptomatic or incidental DVT or PERecurrent VTE within 6 monthsYes: HR 0.63 (0.37–1.07),  < 0.001

AAA, abdominal aortic aneurysm; ACS, acute coronary syndrome; ALI, acute limb ischaemia; DVT, deep vein thrombosis; HR, hazard ratio; LEAD, lower extremity artery disease; MI, myocardial infarction; MRI, magnetic resonance imaging; PAD, peripheral arterial disease; PE, pulmonary embolism; RCT, randomized-controlled study; VTE, venous thromboembolism.

The presence of mural thrombosis is considered as contributive to the AAA growth. The TicAAA study, randomized 144 patients with AAA to receive either ticagrelor 90 mg twice daily or placebo. 27 After 12 months, the AAA volume growth rate assessed by MRI did not differ between the ticagrelor and the placebo groups (9.1% vs. 7.5% , P  =   0.205). Neither the AAA diameter nor the intraluminal thrombus volume change differed between treatment groups.

Both TEDY and TicAAA trials were of short duration (1 year) and potential benefits with longer treatment durations need further investigations. In addition, the percentage proportion of female patients was low (TEDY 12%, TicAAA 4.2%), which does not allow any conclusion with respect to sex differences in treatment effects.

The VOYAGER trial tested whether rivaroxaban 2.5 mg b.i.d. + aspirin 100 mg (dual pathway inhibition, DPI) was superior to aspirin 100 mg alone (control) in preventing thrombotic events after lower limbs revascularization. This multicentre, prospective RCT included 6564 patients (26% females) undergoing infra-iliac arterial revascularization. 28 Primary efficacy outcome was a composite of acute limb ischaemia, major amputation for vascular causes, myocardial infarction, ischaemic stroke, or death from CV causes. It occurred in 17.3% patients in the DPI vs. 19.9% in the control group (HR 0.85, 95% CI 0.76–0.96; P  = 0.009). The most frequent component of the primary endpoint, acute limb ischaemia, occurred in 5.2% in the DPI and 7.8% in the control group (HR 0.67, 95% CI 0.55–0.82). No heterogeneity with respect to patients' sex was reported. TIMI major bleeding occurred in 2.65% patients in the DPI and in 1.87% in the control group (HR 1.43, 95% CI 0.97–2.10; P  = 0.07). Overall, for every 10 000 patients treated for 1 year DPI would prevent 181 primary efficacy outcome events at the cost of 29 principal safety outcome events. The number needed to treat at 3 years was 39.

These results challenge the concept of single or dual antiplatelet therapy (DAPT) after peripheral revascularization. DPI was superior to aspirin monotherapy but was not tested against DAPT, although clopidogrel was allowed in the trial for up to 6 months in both arms. However, the use of DAPT after peripheral endovascular therapy is empirical and has never been validated in a specific RCT. In addition, an economic evaluation showed that combination of rivaroxaban 2.5 mg b.i.d. with aspirin 100 mg is a cost-effective treatment option for patients with chronic CAD or LEAD compared to aspirin alone. 33

Regarding endovascular treatment of LEAD, the use of paclitaxel-coated balloons and stents has been challenged following a systematic review and meta-analysis in 2018 suggesting mortality excess. 34 These safety concerns initiated an unplanned interim analysis of the randomized, open-label, registry-based SWEDEPAD trial, aiming to assess potential effects of drug-eluting technology on the incidence of amputation among patients with chronic limb-threatening ischaemia and health-related quality of life among 2289 patients (45% females) with intermittent claudication. 29 Over a mean follow-up of 2.5 years, the mortality did not differ between the drug-coated device group and the uncoated device group (HR 1.06, 95% CI 0.92–1.22). 29 Furthermore, the mortality did not differ between patients with chronic limb-threatening ischaemia (HR 1.04, 95% CI 0.90–1.21) and those with intermittent claudication (HR 1.18, 95% CI 0.72–1.93). While these findings relieve concerns on a suggested mortality risk after paclitaxel use in LEAD interventions, SWEDEPAD still has to be interpreted with caution as an unintended interim analysis.

5.3 Venous thromboembolism

The Caravaggio study was a prospective open-label non-inferiority RCT comparing apixaban to dalteparin in 1155 cancer patients (51% females) with a venous thromboembolism (VTE) episode. 32 Recurrent VTE occurred in 5.6% in the apixaban group and 7.9% in the dalteparin group (HR 0.63, P  < 0.001 for non-inferiority, P  = 0.09 for superiority). In patients <65 years apixaban was more effective than dalteparin in the prevention of recurrent VTE. No difference in major bleeding was observed between both arms. The rate of the combined cumulative incidence of recurrent VTE or major bleeding did not differ between groups.

Together with previous trials, 35–37 the Caravaggio trial further promotes the use of direct oral anticoagulants in patients with cancer. Clinicians should take into consideration individual bleeding risks, concomitant medication, and cancer types (e.g. patients with brain tumours, known intracerebral metastases, or acute leukaemia were excluded from Caravaggio) in cancer patients with VTE.

5.4 Proprotein convertase subtilisin/kexin type 9 inhibition in VTE and LEAD

Proprotein convertase subtilisin/kexin type 9 (PCSK9) degrades low-density lipoprotein cholesterol (LDL-C) receptors and subsequently raises LDL-C. 38 Modulation of VTE (deep vein thrombosis or pulmonary embolism) and LEAD (CLI, limb revascularization, or amputation for ischaemia) by PCSK9 inhibitors was assessed through prespecified analysis of two large clinical trials. The ODYSSEY-OUTCOMES trial compared alirocumab to placebo in 18 924 patients with recent acute coronary syndrome and uncontrolled dyslipidaemia with maximum-tolerated statin treatment. 30 After a median follow-up of 2.8 years, LEAD-related events occurred in 246 patients and VTE events in 92 patients. Alirocumab significantly reduced the risk of LEAD events (HR 0.69; P  = 0.004). The reduction was proportional with baseline lipoprotein(a) ( P trend  = 0.03), but not with LDL-C levels ( P trend  = 0.50). Fewer, although non-significant, VTE events were recorded in the alirocumab group (HR 0.67; P  = 0.06). VTE risk was numerically higher in the highest baseline quartile of lipoprotein(a) without significant trend across quartiles ( P trend  = 0.22) and without association with baseline quartile of LDL-C corrected ( P trend  = 0.85).

A post hoc analysis of the FOURIER trial (Further Cardiovascular Outcomes Research with PCSK9 Inhibition in Subjects with Elevated Risk) also assessed the VTE risk in 27 564 patients with stable atherosclerosis and hyperlipidaemia on statin therapy, randomized to evolocumab or placebo. 31 After a median follow-up of 2.2 years, the risk of VTE was reduced under evolocumab (HR 0.71; P  = 0.05). There was no relation between baseline LDL-C levels and magnitude of VTE risk reduction. In patients with higher baseline lipoprotein(a) levels, evolocumab reduced VTE risk by 48% (HR 0.52; P  = 0.017), whereas, in those with lower baseline lipoprotein(a) levels, evolocumab had no effect on VTE risk ( P interaction  = 0.087 for HR; P heterogeneity  = 0.037 for absolute risk reduction). There was a significant interaction between baseline lipoprotein(a) concentrations and magnitude of VTE risk reduction ( P interaction  = 0.04).

In both trials, female patients as well as non-white patients were underrepresented. Nevertheless, when combining data from both trials in a meta-analysis, a 31% relative risk reduction in VTE was observed with PCSK9 inhibition vs. placebo (HR 0.69; P  = 0.007). 31

In conclusion, PCSK9 inhibition reduce the risk of LEAD events and could reduce VTE risk. However, further specific studies are required to confirm this class effect.

Although COVID-19 is primary known as a respiratory disease caused by the SARS-CoV-2 virus, vascular complications including coagulopathy, arterial ischaemic events, and VTE are also common. 39 Indeed, hyperinflammatory and prothrombotic states characterize COVID-19 disease. Vascular endothelial cells play a pivotal role in the COVID-19 disease pathophysiology, both as target organ and as contributing contributor to inflammation and thrombosis. 39 SARS-CoV-2 infects directly the endothelium via the angiotensin-converting enzyme 2 receptor inducing endotheliitis. The systemic cytokine storm induced by SARS-CoV-2 virus affects also the endothelium. These changes result in endothelium dysfunction, likely contributing to poor patient outcome ( Figure  3 ). 39

An external file that holds a picture, illustration, etc.
Object name is cvab144f3.jpg

Potential endothelial dysregulation by SARS-CoV-2 (reproduced with permission from Evans et al. 39 ). ACE2, angiotensin-converting enzyme 2 receptor; ROS, reactive oxygen species.

COVID-19 is associated with high VTE prevalence. In a recent meta-analysis, including 48 observational studies (18 093 patients) reporting VTE incidence in hospitalized patients with COVID-19, 40 overall VTE incidence was 17.0%, with 7.1% in patients admitted to the ward and 27.9% in those admitted to the intensive care unit. Most of these patients were receiving pharmacological prophylaxis, suggesting the need for intensive anticoagulation. A number of studies, evaluating optimal dose and course of thromboprophylaxis in hospitalized COVID-19 patients, are ongoing.

Several autopsy studies contributed to clarify the pathophysiology of COVID-19 disease. Ackermann et al . 41 evaluated features of seven lungs from COVID-19 infected patients, to those of patients died from acute respiratory distress syndrome secondary to influenza A (H1N1), and control lungs of age-matched uninfected patients. Both COVID-19 and H1N1 influenza patients shared some histologic pattern such as diffuse alveolar damage with perivascular T-cell infiltration. However, COVID-19 patients presented a typical pattern including severe endothelial injury associated with intracellular SARS-CoV-2 virus, and disrupted endothelial cell membranes. In addition, COVID-19 lungs showed widespread vascular thrombosis with microangiopathy and occlusion of alveolar capillaries. Finally, significant angiogenesis was shown in the lungs of affected patients. 41 In light of these findings, many of the pulmonary embolisms described in the literature in COVID-19 patients may actually be episodes of in situ thrombosis.

Finally, concomitant CV disease or risk factors may aggravate the clinical course of COVID-19 disease as shown in a number of meta-analyses. Evaluating 25 studies and 65 484 patients, Ssentongo et al. 42 showed the association of CV disease and 10 pre-existing comorbidities with COVID-19 mortality. Compared to those without comorbidities, risk of death was significantly higher in patients with CV disease (RR 2.25), hypertension (RR 1.82), diabetes (RR 1.48), congestive heart failure (RR 2.03), chronic kidney disease (RR 3.25), and cancer (RR 1.47). 42

7. Conclusions

During this very special past year, a number of studies paved the way towards a better understanding of several vascular diseases, including atherosclerosis, FMD, aneurysms, and elastopathies, identifying new targets for diagnosis or therapy. In the clinical field, several seminal trials filled the big gap of knowledge on the use of antithrombotic therapies in vascular diseases. COVID-19, the unpredicted ‘guest-star’ of research in 2020 showed major repercussions in the CV system, and here again, the better understanding of its pathophysiology and extensive use of antithrombotic therapies improved the outcome of millions of patients within <10 months although optimal antithrombotic dosage in these patients remains to be determined.

Authors’ contributions

L.M. and V.A. conceived and designed the manuscript, drafted the manuscript, and revised it for important intellectual content. A.A., A.B.R., M.D.C., C.H., C.E.-K., O.S., H.S., D.S., J.R.P., and A.V. made substantial contributions to the conception and design of the manuscript, and drafted the manuscript or revised it for important intellectual content. All authors approved the final version of the manuscript and agreed to be accountable for the work.

Conflict of interest: none declared.

IMAGES

  1. Peripheral vascular disease

    peripheral vascular disease research paper

  2. A Review of Peripheral Vascular Disease

    peripheral vascular disease research paper

  3. Peripheral Artery Disease Concept Map

    peripheral vascular disease research paper

  4. (PDF) Management of Peripheral Vascular Disease

    peripheral vascular disease research paper

  5. (PDF) Management of chronic peripheral artery disease patients with

    peripheral vascular disease research paper

  6. A Compendium on Peripheral Arterial Disease

    peripheral vascular disease research paper

VIDEO

  1. Seminar Peripheral Vascular Disease VID

  2. Peripheral Vascular Disease

  3. peripheral vascular disease

  4. Peripheral Artery Disease: What you Need to Know

  5. Peripheral vascular disease || easy explanation in hindi || Medical surgical nursing ||

  6. What measures help with peripheral vascular disease? #vasculardisease #peripheralarterydisease

COMMENTS

  1. Evidence-Based Medical Management of Peripheral Artery Disease

    Highlights. Peripheral artery disease is a disease of the lower extremity arteries associated with high cardiovascular mortality. Medical management of peripheral artery disease includes cholesterol reduction, antiplatelet therapy, anticoagulation, peripheral vasodilators, blood pressure control, exercise therapy, and smoking cessation, all of which have the capacity to reduce mortality ...

  2. Update on peripheral artery disease: Epidemiology and evidence-based

    1. Definition. Peripheral artery disease (PAD) is a progressive disorder characterized by stenosis and/or occlusion of large and medium-sized arteries, other than those that supply the heart (coronary artery disease, CAD) or the brain (cerebrovascular disease). PAD affects the lower extremities more commonly than the upper extremity vessels ...

  3. Peripheral Arterial Disease: A Narrative Review

    Abstract. Peripheral arterial disease (PAD) describes the partial or complete occlusion of blood flow in the distal arteries of the body. A decreased arterial patency may occur due to a reduction in the elasticity or diameter of the vessel. The goal of interventions is to decrease incidence and reduce complications by identifying and minimizing ...

  4. Peripheral Artery Disease

    Key Clinical PointsPeripheral Artery Disease. •. Atherosclerotic peripheral artery disease affects more than 200 million persons worldwide, including at least 8.5 million persons in the United ...

  5. Peripheral vascular disease

    Peripheral vascular disease is a pathological condition in which one or more blood vessels outside of the heart (for example, in the legs or arms) is narrowed or blocked, usually, but not always ...

  6. Peripheral Artery Disease: A Comprehensive Updated Review

    Abstract. Peripheral arterial disease is estimated to affect more than 200 million people worldwide. Although more than 50% of those affected are asymptomatic, it accounts for 3%-4% of amputations and a crude 5-year death rate of 82.4 deaths per 1000 patient-years when adjusted for duration of follow-up. Additionally, peripheral artery disease ...

  7. Update on the pathophysiology and medical treatment of peripheral

    Peripheral artery disease (PAD) is a growing global public health problem owing to its high and rising prevalence and association with adverse outcomes. In this Review, Golledge summarizes the ...

  8. Free Full-Text

    Despite its significant impact on patients' lives and the healthcare system, peripheral arterial disease (PAD) has long been overshadowed by other cardiovascular diseases [ 1 ]. Originally viewed as a mere complication of conditions such as diabetes and hypertension, PAD is now recognized as an independent and associated risk factor [ 2 ].

  9. Pathophysiology of Peripheral Arterial Disease (PAD): A Review on

    Peripheral arterial disease (PAD) is an atherosclerotic disease that affects a wide range of the world's population, reaching up to 200 million individuals worldwide. PAD particularly affects elderly individuals (>65 years old). ... Feature papers represent the most advanced research with significant potential for high impact in the field. A ...

  10. Peripheral Vascular Disease

    Peripheral vascular disease (PVD) includes peripheral arterial disease (PAD) and venous disease. PAD is a chronic progressive atherosclerotic disease leading to partial or total peripheral vascular occlusion. PAD typically affects the abdominal aorta, iliac arteries, lower limbs, and occasionally the upper extremities.[1] PAD affects nearly 200 million people worldwide with increasing global ...

  11. Health Literacy and Disease Knowledge of Patients With Peripheral

    This scoping review summarises health literacy and disease knowledge in patients with abdominal aortic aneurysm (AAA) or peripheral arterial disease (PAD) and the influencing factors. Data Sources A systematic search was conducted in PubMed, Embase, PsychINFO, and CINAHL covering the period January 2012 to October 2022.

  12. Rivaroxaban in Peripheral Artery Disease after Revascularization

    METHODS. In a double-blind trial, patients with peripheral artery disease who had undergone revascularization were randomly assigned to receive rivaroxaban (2.5 mg twice daily) plus aspirin or ...

  13. Peripheral Artery Disease (PAD) Research

    Research that improves the health of people with peripheral artery disease (PAD) is part of NHLBI's larger commitment to understanding and treating cardiovascular disease. PAD is caused by atherosclerosis that restricts blood flow through the arteries supplying the legs. PAD may be silent (without symptoms) or cause muscle pain with exertion ...

  14. Peripheral Artery Disease: Current Insight Into the Disease and Its

    Abstract. Peripheral artery disease (PAD), which comprises atherosclerosis of the abdominal aorta, iliac, and lower-extremity arteries, is underdiagnosed, undertreated, and poorly understood by the medical community. Patients with PAD may experience a multitude of problems, such as claudication, ischemic rest pain, ischemic ulcerations ...

  15. usually early presentation of peripheral arterial disease in a 30-year

    Peripheral vascular disease is commonly thought to be a disease of elderly patients. However, with certain environmental factors and predetermined genetics, it can present much earlier in life. This case report demonstrates a case of severe peripheral vascular disease in a 30-year-old woman with an 11-year history of smoking.

  16. Endothelial function in cardiovascular medicine: a consensus paper of

    Endothelial cells (ECs) are sentinels of cardiovascular health. Their function is reduced by the presence of cardiovascular risk factors, and is regained once pathological stimuli are removed. In this European Society for Cardiology Position Paper, we describe endothelial dysfunction as a spectrum o …

  17. Epidemiology of Peripheral Artery Disease and Polyvascular Disease

    PERIPHERAL VASCULAR DISEASE COMPENDIUM Circulation Research. 2021;128:1818-1832. DOI: 10.1161/CIRCRESAHA.121.318535 June 11, 2021 1819 Aday and Matsushita Epidemiology of PAD as patients with PAD, regardless of leg symptoms, have poorer prognosis and reduced limb function compared with those without.

  18. Characteristics of motor evoked potentials in patients with peripheral

    With an aging population, it is common to encounter people diagnosed with peripheral vascular disease (PVD). Some will undergo surgeries during which the spinal cord may be compromised and intraoperative neuromonitoring with motor evoked potentials (MEPs) is employed to help mitigate paralysis. No data exist on characteristics of MEPs in older, PVD patients, which would be valuable for ...

  19. Peripheral Arterial Disease

    Patients with peripheral arterial disease (PAD) have decreased lower extremity arterial perfusion which is commonly referred to as "poor circulation." In most cases of PAD, atherosclerotic plaques narrow the arterial flow lumen which restricts blood flow to the distal extremity. Reduced blood flow can cause thigh or calf pain with walking due to temporary ischemia of the leg muscles during ...

  20. What is Peripheral Artery Disease?

    Peripheral artery disease, or PAD, is a narrowing of the peripheral arteries that carry blood away from the heart to other parts of the body. The most common type is lower-extremity PAD, in which blood flow is reduced to the legs and feet. Upper-extremity PAD (arms, hands and fingers) is less common but affects about 10% of the population.

  21. Diabetes and peripheral artery disease: A review

    Peripheral arterial disease (PAD) refers to partial or complete occlusion of the peripheral vessels of the upper and lower limbs. It usually occurs as part of systemic atherosclerosis in the coronary and cerebral arteries. The prevalence of PAD is expected to continue to increase in the foreseeable future owing to the rise in the occurrence of ...

  22. Peripheral Vascular Disease (PVD)

    Atherosclerosis is a gradual process whereby hard cholesterol substances (plaques) are deposited in the walls of the arteries. This buildup of cholesterol plaques causes hardening of the artery walls and narrowing of the inner channel (lumen) of the artery. When this happens in the peripheral circulation, peripheral vascular disease is the result.

  23. Progress in aorta and peripheral cardiovascular disease research

    Abstract. Although coronavirus disease 2019 seems to be the leading topic in research number of outstanding studies have been published in the field of aorta and peripheral vascular diseases likely affecting our clinical practice in the near future. This review article highlights key research on vascular diseases published in 2020.