Hostname: page-component-7bb8b95d7b-nptnm Total loading time: 0 Render date: 2024-09-29T18:49:03.801Z Has data issue: false hasContentIssue false

Pulmonary reperfusion injury in post-palliative intervention of oligaemic cyanotic CHD: a new catastrophic consequence or just revisiting the same old story?

Published online by Cambridge University Press:  18 October 2023

Sisca Natalia Siagian
Affiliation:
Division of Pediatric Cardiology and Congenital Heart Disease, Department of Cardiology and Vascular Medicine, National Cardiovascular Centre Harapan Kita, Universitas Indonesia, Jakarta, Indonesia
Made Satria Yudha Dewangga
Affiliation:
Department of Cardiology and Vascular Medicine, Sanglah General Hospital, Denpasar, Indonesia
Bayushi Eka Putra
Affiliation:
Division of Pediatric Cardiology and Congenital Heart Disease, Department of Cardiology and Vascular Medicine, National Cardiovascular Centre Harapan Kita, Universitas Indonesia, Jakarta, Indonesia
Christianto Christianto*
Affiliation:
Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
*
Corresponding author: C. Christianto; Email: [email protected]
Rights & Permissions [Opens in a new window]

Abstract

Pulmonary reperfusion injury is a well-recognised clinical entity in the setting pulmonary artery angioplasty for pulmonary artery stenosis or chronic thromboembolic disease, but not much is known about this complication in post-palliative intervention of oligaemic cyanotic CHD. The pathophysiology of pulmonary reperfusion injury in this population consists of both ischaemic and reperfusion injury, mainly resulting in oxidative stress from reactive oxygen species generation, followed by endothelial dysfunction, and cytokine storm that may induce multiple organ dysfunction. Other mechanisms of pulmonary reperfusion injury are “no-reflow” phenomenon, overcirculation from high pressure in pulmonary artery, and increased left ventricular end-diastolic pressure. Chronic hypoxia in cyanotic CHD eventually depletes endogenous antioxidant and increased the risk of pulmonary reperfusion injury, thus becoming a concern for palliative interventions in the oligaemic subgroup. The incidence of pulmonary reperfusion injury varies depending on multifactors. Despite its inconsistence occurrence, pulmonary reperfusion injury does occur and may lead to morbidity and mortality in this population. The current management of pulmonary reperfusion injury is supportive therapy to prevent deterioration of lung injury. Therefore, a general consensus on pulmonary reperfusion injury is necessary for the diagnosis and management of this complication as well as further studies to establish the use of novel and potential therapies for pulmonary reperfusion injury.

Type
Review
Copyright
© The Author(s), 2023. Published by Cambridge University Press

Background

Patients with cyanotic CHD and decreased pulmonary blood flow may require palliative interventions before the corrective surgeries, especially those with complex lesions or severe desaturation. Blalock–Taussig–Thomas shunt is one of the surgical approach of these palliative interventions. Reference Quandt, Ramchandani and Stickley1,Reference Sandoval, Chaturvedi and Benson2 However, transcatheter approaches such as patent ductus arteriosus stenting in duct-dependent CHD or the recently developed right ventricular outflow tract stenting in tetralogy of Fallot are preferable for selected cases in some centres compared to surgical approach which is associated with significant mortality and morbidity. Reference Li, Zhou and Li3 Although these palliative procedures aim to increase pulmonary blood flow, the sudden increase of blood flow in the previously underperfused pulmonary vasculature may cause acute pulmonary reperfusion injury. Reference Asija, Roth and Hanley4,Reference Yacouby, Meador and Mossad5

Pulmonary reperfusion injury is a well-recognised clinical entity in the setting of pulmonary artery angioplasty for pulmonary artery stenosis or chronic thromboembolic disease. Pulmonary reperfusion injury manifests as temporary and localised pulmonary oedema in an area of increased lung tissue perfusion which impairs gas exchange. Reference Yacouby, Meador and Mossad5,Reference Ejiri, Ogawa, Fujii, Ito and Matsubara6 One study reported that 10 out of 46 patients (22%) developed pulmonary reperfusion injury after pulmonary artery angioplasty. Reference Yacouby, Meador and Mossad5 Another study reported three out of four patients with Williams syndrome developed clinical pulmonary reperfusion injury after pulmonary artery balloon angioplasty, thus indicating higher risk of pulmonary reperfusion injury in these patients. Reference Maskatia, Feinstein, Newman, Hanley and Roth7 Nevertheless, the incidence of pulmonary reperfusion injury in post-palliative intervention in cyanotic CHD is unknown. The exact mechanism of pulmonary reperfusion injury in these specific population is still poorly understood. Therefore, this review aims to evaluate and collect the evidence as well as to review the mechanism of pulmonary reperfusion injury in post-palliative intervention in cyanotic CHD.

Pulmonary reperfusion injury and its clinical manifestations

Ischemia-reperfusion injury is a complex pathological process as a result of paradoxical exacerbation of cellular dysfunction and death, following restoration of blood flow to previously ischaemic tissues. Reference Cowled, Fitridge, Fitridge and Thompson8 All tissues and organs are indeed susceptible to ischaemia, but there is difference between the susceptibility to an ischaemic insult. Reference Zarbock, Eroglu, Erturk, Ince and Westphal9 Despite being resistant to ischaemia due to the availability of oxygen from both alveolar gas exchange and the blood supply via a dual circulatory system (pulmonary and bronchial arteries), lungs can still suffer from ischaemia in situations that impair alveolar oxygenation or pulmonary blood flow. Therefore, ischaemia-reperfusion injury may affect lungs, otherwise known as pulmonary reperfusion injury. Reference den Hengst, Gielis, Lin, Van Schil, De Windt and Moens10

Pulmonary reperfusion injury or lung ischaemia-reperfusion injury is a complex pathological process triggered by lack of oxygen supply, followed by a period of reperfusion in the lung. Reference Weyker, Webb, Kiamanesh and Flynn11 Pulmonary reperfusion injury causes increased pulmonary vascular resistance and increased vascular permeability manifesting as varying levels of non-cardiogenic pulmonary oedema with pulmonary wedge pressures typically less than 18 mmHg. Pulmonary oedema will cause ventilation/perfusion (V/Q) mismatch which affects the oxygenation. Chest X-ray of pulmonary reperfusion injury usually shows ranging severity from mild infiltrates to diffuse opacifications, similar to an acute respiratory distress syndrome. Reference Weyker, Webb, Kiamanesh and Flynn11

There is currently no specific diagnosis criteria for pulmonary reperfusion injury; thus, it is primarily a diagnosis of exclusion. There were various criteria used by a few reports, but they were not yet established for the diagnosis of pulmonary reperfusion injury. Reference Khemani, Patel, Bart and Newth12Reference Christie, Carby, Bag, Corris, Hertz and Weill14 The International Society of Heart and Lung Transplant (ISHLT) criteria was the best available criteria to identify and grade segmental pulmonary reperfusion injury, though they are designed for whole-lung graft reperfusion. The criteria stated that PaO2/FiO2 ratio of 200–300 indicates pulmonary reperfusion injury and a ratio of less than 200 indicates severe pulmonary reperfusion injury. Reference Christie, Carby, Bag, Corris, Hertz and Weill14 Post-intervention pulmonary oedema or haemorrhage, unilateral or bilateral chest radiograph infiltrates, ICU admission, as well as unplanned intubation and post-operative mechanical ventilation may serve as a predictive clinical features of pulmonary reperfusion injury. Reference Zarbock, Eroglu, Erturk, Ince and Westphal9

Aetiology of pulmonary reperfusion injury

Lung ischaemia occurs when oxygen supply fails to meet the metabolic demands of the pulmonary parenchyma either due to decreased ventilation or limited blood flow. Therefore, we can divide the causes of pulmonary reperfusion injury into two groups: ventilated ischaemia and anoxic ischaemia. In ventilated ischaemia, the ventilation is normal while the blood supply to the lung is interrupted, which can be seen in thrombotic situations (acute chest syndrome in sickle cell patients and pulmonary artery embolic phenomena) and stenotic situations (pulmonary artery stenosis). In this group, pulmonary reperfusion injury follows the correction of the interrupted blood supply by thromboendarterectomy or catheter-directed thrombolysis and balloon angioplasty. On the contrary, there is complete cessation of blood flow and ventilation in anoxic ischaemia. Pulmonary reperfusion injury occurs less frequently in this group which includes the cold ischaemia time occuring in cardiopulmonary bypass and lung transplantation. Factors possibly associated with pulmonary reperfusion injury are length of ischaemic time, hypotension, overzealous fluid administration, mechanical ventilation, infection, cause of death of lung transplant donor, and extent of immune response. Reference Weyker, Webb, Kiamanesh and Flynn11

Pathogenesis and pathophysiology of pulmonary reperfusion injury

The five main cellular mechanisms in pulmonary reperfusion injury include sterile immunity, complement activation, activation of coagulation, activation of cell death pathways, and finally, endothelial dysfunction. Sterile immunity includes the activation of adaptive and innate immunity, while complement activation amplifies the immune response. Activation of coagulation includes platelet activation and aggregation, microvascular vasoconstriction, and thrombus formation. Reference Weyker, Webb, Kiamanesh and Flynn11 Endothelial dysfunction causes increased vascular permeability (pulmonary oedema), activation of complement and coagulation systems, imbalance of vasoconstricting and vasodilating factors, as well as “no re-flow phenomenon.” Reference Eltzschig and Carmeliet15Reference Yemisci, Gursoy-Ozdemir, Vural, Can, Topalkara and Dalkara17 The end result of these pathologic processes may be seen microscopically as alveoli with marked perivascular oedema, focal interstitial and intraalveolar leucocyte infiltration, and proteinous exudate. Reference Weyker, Webb, Kiamanesh and Flynn11

The pathological processes of pulmonary reperfusion injury can be divided into two parts: ischaemia injury and reperfusion injury. Reference Zarbock, Eroglu, Erturk, Ince and Westphal9,Reference Wu, Yiang and Liao18 Imbalance of oxygen supply and demand in ischaemia will cause hypoxia, leading to dysfunction in the electron transport chain in mitochondria. Decreasing adenosine triphosphate production will induce anaerobic metabolism, dysfunction of sodium–potassium pumps (Na+/K+-adenosine triphosphate synthase), and detachment of ribosomes. Reference Yellon and Hausenloy16,Reference Mura, Andrade and Han19 Anaerobic metabolism produces a lower level of adenosine triphosphate and antioxidative agents in the cells as well as lactic acidosis which may lead to metabolic acidosis. Failure of sodium–potassium pumps and calcium pumps (Ca2+-adenosine triphosphate synthase) on the cell surface will cause retention of sodium and calcium in cells and potassium out of cells. Activity of sodium–hydrogen exchanger pumps (Na+/H+) is decreased as a result of a higher level of sodium in cells, thus causing accumulation of hydrogen in cells. Dysfunctional calcium pumps on endoplasmic reticulum will also limit calcium reuptake. The cells will accumulate sodium, calcium, and hydrogen ions resulting in hyperosmoloarity and cell swelling. Decreased cellular pH also leads to impaired enzyme activity and clumping of nuclear chromatin. Detachment of ribosomes decreases synthesis of protein. Reference Wu, Yiang and Liao18

Despite the believe that the lung is resistant to ischaemic injury because of its independent source of oxygen in the alveolar space, multiple studies have described that the lung becomes vulnerable to ischaemia-reperfusion injury because of this feature. Reference Deffebach, Charan, Lakshminarayan and Butler20Reference Adkins and Taylor22 Alveolar oxygen does help maintain aerobic metabolism and prevent hypoxic reactive oxygen species formation. Reference Eckenhoff, Dodia, Tan and Fisher23 However, a high concentration of oxygen in the gas mixture in addition to the absence of blood flow with a low oxygen gradient in the lung tissue can cause reactive oxygen species formation, even more in the case of ventilation. Reference Fisher, Dodia, Tan, Ayene and Eckenhoff24

In the reperfusion phase, restoring the blood flow to the ischaemic lung tissue will not only restore the oxygen supply but also increase the generation of reactive oxygen species, such as superoxide anions, hydrogen peroxide, and the most unstable, hydroxyl radicals leading to secondary injury. Reperfusion in condition of mitochondrial damage and electrolyte imbalance promotes oxidative stress from three major systems: xanthine oxidase system, nitric oxide synthase system, and reduced nicotinamide adenine dinucleotide phosphate oxidase system. A compensatory mechanism is activated to nullify the effect of the reactive oxygen species in normal circumstances. However, the large amount of reactive oxygen species generated from both ischaemic phase and reperfusion phase as well as the lower concentration of antioxidative agents in the ischaemic cells can lead to oxidative stress that promotes endothelial dysfunction, deoxyribonucleic acid damage, and local inflammatory responses. Reference Weyker, Webb, Kiamanesh and Flynn11,Reference Wu, Yiang and Liao18

These inflammatory cascades and oxidative stress may lead to a cytokine storm, resulting in damage to cellular structures. The cell response is dependent on the severity of tissue injury and can be correlated with the duration of ischaemia-reperfusion injury. A shorter duration or mild ischaemia-reperfusion injury may activate cell survival programmes to control reactive oxygen species generation and cell damage, while moderate ischaemia-reperfusion injury cause cell dysfunction by autophagy but may activate recovery systems for survival. However, prolonged ischaemia-reperfusion injury may lead to cell death via four pathways: autophagy, mitoptosis, necrosis and necroptosis, and apoptosis. Reference Wu, Yiang and Liao18,Reference Verma, Fedak and Weisel25 Ischaemia-reperfusion injury of a single organ, the lung in this case, causes the release of various pro-inflammatory mediators which may induce infllammation in other organ, thus potentially contributing to multiple organ dysfunction or failure. Reference Zarbock, Eroglu, Erturk, Ince and Westphal9,Reference Mura, Andrade and Han19

As mentioned before, “no-reflow phenomenon” may occur as a consequence of endothelial dysfunction during the reperfusion phase. Activation of leucocyte, platelet, and complement may lead to endothelial dysfunction and subsequently, formation of thrombi and impaired vascular relaxation, thereby decreased microvascular flow despite reperfusion. This may further delay the recovery of the ischaemic cells in ischaemia-reperfusion injury as shown in some cases of delayed graft function or persistent organ dysfunction despite adequate reperfusion. Reference Weyker, Webb, Kiamanesh and Flynn11

Another hypothesis on the pathophysiology of pulmonary reperfusion injury is the exposure of the pulmonary circulation to higher pressure after angioplasty as shown by the sudden increase of pulmonary artery mean distal pressure exceeding 20 mmHg or a sudden increase of the pressure more than 150 per cent from the baseline, or possibly the increase in the end-diastolic volume of a non-compliant left ventricle. Reference Zarbock, Eroglu, Erturk, Ince and Westphal9,Reference Bergersen and Lock26 This pathophysiology concept was in line with a case series by Ho et al which reported pulmonary reperfusion injury following stent implantation in patients with ductal-dependent unilateral disconnected pulmonary artery. Ho et al proposed that in the early phase, unilateral pulmonary oedema results primarily from acute pulmonary reperfusion injury to the stented lung, exacerbated by increase in left ventricular end-diastolic pressure due to the acute increase in preload. Following recovery from the early phase, there was evidence of a high pulmonary blood flow state to both lungs leading to pre- and post-capillary pulmonary hypertension as shown by the evidence of high right ventricular pressures and marked right ventricular dilatation. Reference Ho, Salmon, Hribernik, Hayes, Thomson and Bentham27

Pulmonary reperfusion injury in cyanotic CHD

Cyanotic CHD can be broadly classified into two subgroups: decreased pulmonary blood flow and increased pulmonary blood flow (Fig. 1). The main cause of pulmonary injury in cyanotic CHD is hypoxia which may be acute or chronic. Acute hypoxia leads to acidosis and depletion of glycogen and adenosine triphosphate causing the heart to rely on anaerobic metabolism. Similarly, chronic hypoxia leads to cyanosis as well as glycogen and adenosine triphosphate depletion though in a much longer period of time. Reference Silverman, Kohler, Levitsky, Pavel, Fang and Feinberg28

Figure 1. Classification of cyanotic CHD. *ASD = atrial septal defect; AVSD = atrioventricular septal defect; BTTS = Blalock–Taussig–Thomas shunt; ccTGA = congenitally corrected transposition of the great arteries; DORV = double-outlet right ventricle; HLHS = hypoplastic left heart syndrome; PAH = pulmonary artery hypertension; PBF = pulmonary blood flow; PDA = patent ductus arteriosus; PS = pulmonary stenosis; PV = pulmonary vein; RVOT = right ventricular outflow tract; TAPVD = total anomalous of pulmonary vein drainage; TGA = transposition of the great arteries; VSD = ventricular septal defect. Reference Laubach and Sharma29,Reference Ossa Galvis, Bhakta, Tarmahomed and Mendez30

It is known that infants with normoxic myocardium have increased tolerant to ischaemia because of adaptive mechanism. Reference Teoh, Mickle and Weisel31,Reference Hammon32 An animal model study also showed that CHD rats under hypoxia conditions and volume-overloaded hearts had higher n-3/n-6 polyunsaturated fatty acid ratios which may upregulate antioxidant signalling pathway, thus providing cardioprotection against ischaemia-reperfusion injury. Reference Asada, Itoi, Nakamura and Hamaoka33 However, chronic hypoxia in cyanotic CHD eventually depletes endogenous antioxidant in myocardium and other organs making them more susceptible to oxygen-mediated injury, thereby increasing the risk of pulmonary reperfusion injury. Reference Asada, Itoi, Nakamura and Hamaoka33,Reference Allen and Ilbawi34

The risk of pulmonary reperfusion injury in cyanotic CHD raises a concern for patients in the decreased pulmonary blood flow subgroup. Reference Quandt, Ramchandani and Stickley1,Reference Sandoval, Chaturvedi and Benson2 Despite the temporary relieve of cyanosis by reperfusion of the pulmonary vasculature, palliative interventions may paradoxically cause acute pulmonary reperfusion injury. Reference Asija, Roth and Hanley4,Reference Yacouby, Meador and Mossad5 Yacouby et al reported incidence of pulmonary reperfusion injury in 22% of patients (10 out of 46 subjects) who underwent pulmonary artery balloon angioplasty. Reference Yacouby, Meador and Mossad5 Several case reports also showed that patients who underwent pulmonary artery balloon angioplasty were more vulnerable to acute pulmonary reperfusion injury when they had pre-existing long-standing pulmonary stenosis or persistently high pulmonary artery pressure after dilation. Therefore, cyanotic CHD with decreased pulmonary blood flow may have similar risk of pulmonary reperfusion injury. Reference Walker, Bateman, Rigby and Brookes35Reference Ostovan, Kamali and Zolghadrasli38 The traditional palliative intervention for cyanotic CHD is Blalock–Taussig–Thomas shunt surgery. Aside from the surgical approach, there is room for transcatheter approach which is minimally invasive. Patent ductus arteriosus stenting or right ventricular outflow tract stenting/balloon are feasible to increase pulmonary blood flow in severe right ventricular outflow tract obstruction and duct-dependent cyanotic CHD. Reference Laubach and Sharma29

Palliative intervention in oligaemic cyanotic CHD

Surgical aortopulmonary shunt (Blalock–Taussig–Thomas shunt)

Blalock–Taussig–Thomas shunt is a surgical procedure of bridging the subclavian or innominate artery to pulmonary artery, first reported in 1945. Reference Blake and Yancy39 Despite being considered as the sole treatment for patients with tetralogy of Fallot in the past, Blalock-Taussig-Thomas shunt is currently indicated for infants with complex single-ventricle physiology or patients with late presentation. Primary tetralogy of Fallot repair is the definitive management of tetralogy of Fallot, though many centres still palliate infants before the definitive procedures. Reference Kaskinen, Keski-Nisula and Martelius40 Modified Blalock–Taussig–Thomas shunt, originated from the Great Ormond Street Group, is developed from Blalock–Taussig–Thomas shunt by interposing a prosthetic tube between the subclavian artery and pulmonary artery. This technique is more advanced with less dissection compared to the previous technique while guaranteeing a proximal and distal anastomosis of large diameter. The maximal flow is regulated by the patient’s uninterrupted subclavian artery diameter, thus allowing for a possible increase in shunt flow with growth. However, Blalock–Taussig–Thomas shunt has been associated with high morbidity and mortality (10–20%) even with the advancement in technology and technique, thus making transcatheter approach more of an attractive alternative. Reference Kaskinen, Keski-Nisula and Martelius40,Reference Caputo, Mokhtari and Rogers41

The incidence of pulmonary reperfusion injury in Blalock–Taussig–Thomas shunt varied between studies. Two patients with univentricular heart complex and pulmonary stenosis had pulmonary reperfusion injury after undergoing modified Blalock–Taussig–Thomas shunt using polytetrafluoroethylene tube and classsic Blalock–Taussig–Thomas shunt. Reference Okita, Miki and Kusuhara42 Tassig et al observed that from the 601 patients who survived Blalock–Taussig–Thomas shunt, 20 patients (3.3%) developed pulmonary hypertension. Reference Tassig, Crocetti and Eshaghpour43 Hofschire et al added that incidence of pulmonary vascular disease increased with time following the Blalock–Taussig–Thomas shunt. There was no patient, from total of 36 patients, developed severe pulmonary vascular changes in less than 8 years following the Blalock–Taussig–Thomas shunt, while 10 of 20 patients (50%) developed some degree of pulmonary vascular disease after having Blalock–Taussig–Thomas shunt 8 years or longer. Reference Hofschire, Rosenquist, Ruckerman, Moller and Edwards44

The mechanism for pulmonary reperfusion injury in Blalock–Taussig–Thomas shunt was thought simply due to overshunting which is depicted by signs of shock and congested lungs with wide pulse pressure from six readings within 6 hours and saturation above 85 on room air. Reference Ismail, Almazmi and Khokhar45 Overshunting might be caused by a bigger shunt size leading to a diastolic run-off which ends up with decreased coronary and systemic perfusion. However, this notion was denied in the study by Ismail et al, which indicated that a larger shunt size did not correlate with overshunting. Reference Ismail, Almazmi and Khokhar45,Reference Dirks, Prêtre and Knirsch46 Degree of shunting or pulmonary flow to systemic flow ratio (Qp:Qs) in Blalock–Taussig–Thomas shunt is also dependent on the ratio of pulmonary to systemic vascular resistance. In case of overshunting, we have to optimise the balance of Qp:Qs by increasing the pulmonary vascular resistance and reducing the systemic vascular resistance. Pulmonary vascular resistance may be increased by reducing fraction of inspired oxygen to 0.21 despite suctioning or nebulising, avoiding hyperventilation with targeted permissive hypercapnea, administering high positive end-expiratory pressure of 6–9, and maintaining blood pH of 7.35–7.40. Systemic vascular resistance may be reduced by administering inodilators. Reference Ismail, Almazmi and Khokhar45,Reference Wardoyo, Makdinata and Wijayanto47

The long duration of Blalock–Taussig–Thomas shunt procedure is one of the risks of pulmonary reperfusion injury because ischaemia-reperfusion injury of the lungs occurs not only after the procedure but also during the surgical procedure by utilisation of cardiopulmonary bypass. Previous animal studies performed in dogs have shown that cardiopulmonary bypass procedures increase malondialdehyde content in the lung interstitium with simultaneous downregulation of superoxide dismutase activity. Malondialdehyde is the end product of lipid peroxidation which is an indicator of the extent of lipid peroxidation, while superoxide dismutase is an oxygen free radical scavenger which reflects the antioxidant activity. Furthermore, the expression of transforming growth factor-beta 1 also increases during cardiopulmonary bypass. Transforming growth factor-beta 1 is usually related with the increase of pulmonary endothelial and alveolar epithelial permeability. Additionally, transforming growth factor-beta 1 also leads to decreased sodium channel on the apical surface of alveolar epithelial cells, thus impairing the removal of water and salt for alveolar lumen. Increased oxygen free radicals could activate pro-inflammatory nuclear factors, such as Nuclear factor kappa B and activator protein 1, which lead to upregulation of transforming growth factor-beta 1. Transforming growth factor-beta 1 can reversely promote the expression of oxygen free radicals, thus forming the endless circle of oxygen free radical-transforming growth factor-beta 1-lung injury. All these evidences show that cardiopulmonary bypass use tilts the balance in the lung to pro-inflammatory state which may increase the risk of pulmonary reperfusion injury. Reference Qu, Li, Wang, Yang and Wang48

The simplest solution to prevent cardiopulmonary bypass-induced pulmonary injury is not using cardiopulmonary bypass in the cardiac surgery (inversion to off-pump operation) as shown in off-pump coronary artery bypass graft surgery. However, it is still impossible to perform most cardiac surgeries without cardiopulmonary bypass. Reference Apostolakis, Koletsis, Baikoussis, Siminelakis and Papadopoulos49 There were some strategies explored to minimise these deleterious effects of cardiopulmonary bypass from improved cardiopulmonary bypass devices and methods to pharmacological pre-conditioning and post-conditioning. However, the effectiveness of these strategies are still unclear. Reference Luc, Aboelnazar and Himmat50Reference Slottosch, Liakopoulos and Kuhn52

Cardiopulmonary bypass machines impose patients with very high oxygen tension of 400–500 mmHg. Reference Gauduel, Menasche and Duvelleroy53,Reference Joachimsson, Sjöberg, Forsman, Johansson, Ahn and Rutberg54 Administration of 100% oxygen may cause oxygen toxicity or absorptive atelectasis as shown in a previous study which reported exacerbation of post-operative lung injury with the administration of 100% oxygen during cardiopulmonary bypass. Reference Sasson, Sherman and Ezri55 Therefore, keeping normoxia condition (PO2 of 80–100 mmHg) of the cardiopulmonary bypass is required to reduce the extent of myocardial and pulmonary dysfunction in regard to reoxygenation injury. Reference Joachimsson, Sjöberg, Forsman, Johansson, Ahn and Rutberg54Reference Arthur E.Baue, Graeme, Hillel and Keith56

Aside from hyperoxia, leucocytes might cause reoxygenation injury by generating oxygen-derived radicals. The method of leucocyte depletion strategy by using leucocyte depletion filters may reduce the effect of reoxygenation injury along with keeping the oxygen tension at a normal level. Reference Allen and Ilbawi34 Additionally, in vivo study by Qiu et al reported that reperfused lung tissues increased the expression of enzymatic product inosine monophosphate deaminase which regulates neutrophil trafficking in the microvessel. This finding may provide an alternative prospect for minimising ischaemic-reperfusion-associated lung injury besides the depletion strategy. On the contrary, an ex vivo study by Luc et al showed that there was increased pro-inflammatory cytokines and leucocytes in the perfusate despite the use of leucocyte filter as it became saturated over 12 hours of ex vivo lung perfusion. Reference Luc, Aboelnazar and Himmat50 Therefore, there is still no objective evidence for the routine use of leucocyte filter in cardiopulmonary bypass.

Circuits coating has been used for the prevention of inflammatory reaction in cardiopulmonary bypass use, especially heparin-coated circuit which is the first and most extensively studied. The concept behind heparin coating is mimicking the endothelial surface that contains heparin sulphate, thus reducing complement activation, reducing inflammatory response, and improving clinical outcomes. Most studies showed improved pulmonary function with heparin-coated circuit, but there was no influence on the ICU stay and intubation time of the patients. Reference Apostolakis, Koletsis, Baikoussis, Siminelakis and Papadopoulos49

A small observational study conducted by Su et al suggested that deep hypothermic circulatory arrest increased the risk of post-operative pulmonary dysfunction compared to that of the deep hypothermic low flow during cardiopulmonary bypass. Although deep hypothermic circulatory arrest confers predisposition to worse hypoxic conditions, longer exposure of blood in the cardiopulmonary bypass circuit during deep hypothermic low flow may lead to more complement-related injury in the pulmonary endothelium, thus exacerbating the risk of lung injury. Reference Qiu, Wada, Stahl and Serhan57

The use of prime solution in cardiopulmonary bypass circuits may cause increased body fluid volume (hemodilution), thus posing a problem for children who have poor ability to regulate and excrete body fluids content. The excessive increase of body fluids volume may result in haemostatic impairment, coagulation disorders, inflammatory response, and myocardial and pulmonary oedema leading to increased post-operative mortality and morbidity. Reference Hu, Li, Chen, Yan, Zhang and Zhang58 Ultrafiltration is used to minimise hemodilution by removing volume of priming and reducing overall post-operative oedema, specifically that of lungs for better post-operative oxygenation in cardiac surgery. Additionaly, it has been shown that ultrafiltration also removes destructive and inflammatory substances, inflammatory cyokines, and toxin from the circulation as well as increases the colloid oncotic pressure which subsequently prevents the development of pulmonary interstitial oedema. Reference Apostolakis, Koletsis, Baikoussis, Siminelakis and Papadopoulos49

Both conventional ultrafiltration with or without modified ultrafiltration are used routinely in cardiopulmonary bypass. Conventional ultrafiltration is carried during the running of cardiopulmonary bypass to maintain moderate hemodilution and minimal venous reservoir blood. Modified ultrafiltration, which has greater efficiency in removing excess fluid compared to conventional ultrafiltration, is performed immediately after the termination of cardiopulmonary bypass. Despite its advantage, modified ultrafiltration has some potential disadvantageous such as technical complications, surface-induced inflammatory responses, as well as longer duration and additional cost. Reference Hu, Li, Chen, Yan, Zhang and Zhang58,Reference Kuratani, Bunsangjaroen, Srimueang, Masaki, Suzuki and Katogi59 A meta-analysis of paediatric cardiac surgery showed that conventional ultrafiltration with increased ultrafiltration volume and post-cardiopulmonary bypass haematocrit is compared with conventional ultrafiltration alone. However, modified ultrafiltration did not significantly influence the post-operative aortic occlusion, cardiopulmonary bypass, and hospital stay duration. Reference Hu, Li, Chen, Yan, Zhang and Zhang58 Another meta-analysis of randomised controlled trial of paediatric cardiac surgery also showed that there was significant improvement of clinical conditions in the immediate postbypass period with modified ultrafiltration over conventional ultrafiltration, but the post-operative outcome parameters were not significantly influenced. Reference Kuratani, Bunsangjaroen, Srimueang, Masaki, Suzuki and Katogi59 Therefore, the advantage of conventional ultrafiltration with modified ultrafiltration over conventional ultrafiltration only in children is still unclear.

Steroids have been used as post-operative lung protection after cardiopulmonary bypass for nearly 30 years, but there is still conflicting evidence regarding its effectiveness to reduce post-operative mortality and to improve post-operative complications after cardiopulmonary bypass. Reference Whitlock, Devereaux and Teoh60Reference Glumac, Kardum and Sodic63 An experimental study showed improvement of post-operative lung function (alveolar-arterial oxygen gradient, pulmonary vascular resistance, and extracellular lung water) by using methylprednisolone as pre-treatment. Reference Lodge, Chai, Daggett, Ungerleider and Jaggers64 However, this was denied by clinical studies by Chaney et al with contradicting results. Reference Chaney, Durazo-Arvizu, Nikolov, Blakeman and Bakhos65,Reference Chaney, Nikolov, Blakeman, Bakhos and Slogoff66 A recent meta-analysis showed that low-dose corticosteroid prophylaxis significantly decreased inflammatory factor concentrations and improved the overall outcome of cardiac surgery with cardiopulmonary bypass in adult patients. However, there was increased risk of myocardial infarction and hyperglycaemia requiring insulin infusion. In children, corticosteroid was associated with shortened cardiopulmonary bypass time, increased risk of insulin infusion, and no substantial changes in mortality and other outcomes. Reference Chai, Zhuang and Tian67 Another review concluded that perioperative corticosteroid did not improve mortality rate or other secondary outcomes such as stroke, renal failure, myocardial infarction, and infection, but there were some benefits: reduced risk of pneumonia and respiratory failure, and shortened length of ICU and hospital stay. Reference Perchermeier and Tassani-Prell68 Despite this contradicting evidence, many centres still use this fast-track recovery protocol as a fundamental strategy. Reference Hall, Smith and Rocker69Reference Tassani, Richter and Barankay71

Patent ductus arteriosus stenting

Patent ductus arteriosus stent implantation is a procedure to maintain pulmonary blood flow through the ductus arteriosus, especially in ductal-dependent cyanotic CHD. Reference Fitridge72 This procedure has become more popular as it is minimally invasive, avoiding a median sternotomy or lateral thoracostomy, as well as exposure to cardiopulmonary bypass. Reference Fitridge72,Reference Glatz, Petit and Goldstein73 Glatz et al reported no difference in mortality or unplanned intervention between patent ductus arteriosus stenting and Blalock–Taussig–Thomas shunt. Nevertheless, patent stenting had fewer procedural complications, shorter length of stay and ICU stay, lower diuretic use, and more symmetrical with larger size of pulmonary arteries which are beneficial for primary repair. Reference Glatz, Petit and Goldstein73 Bentham and colleagues also found that patent ductus arteriosus stenting had a significant mortality benefit, less extracorporeal support, fewer ventilation days, shoreter ICU stay, and overall length of stay. Reference Bentham, Zava and Harrison74 These results were generally similar to a study by Nasser et al, though there is no difference in growth and symmetry of pulmonary artery between both groups. Reference Nasser, Abdulrahman, Qwaee, Alakfash, Mohamad and Kabbani75 Despite the aforementioned superiority, patent ductus arteriosus stenting usually required more reintervention with higher rate of failure (17%) which is followed by switching to surgical Blalock–Taussig–Thomas shunt. Reference Bentham, Zava and Harrison74,Reference Nasser, Abdulrahman, Qwaee, Alakfash, Mohamad and Kabbani75 The incidence of pulmonary reperfusion injury in patent ductus arteriosus stenting is rare, but it still may occur as a complication compromising haemodynamic. This is evidenced by a study by Bahaidarah et al founding one case of pulmonary reperfusion injury from 43 patent ductus arteriosus stenting procedures in patients with cyanotic CHD. Reference Bahaidarah, Al-Ata and Alkhushi76,Reference Alwi77

Right ventricular outflow tract stenting or balloon procedure

Severely desaturated infants with acidosis metabolic who are prostaglandin-dependent in cases of stenotic antegrade pulmonary blood flow as a consequence of the narrowing of the right ventricular outflow tract (infundibular or valvar stenosis) or disturbances in the pulmonary arterial tree such as hypoplastic or the presence of multiple aortopulmonary collateral arteries may require early intervention. Comorbidities such as prematurity, low weight, infection, neurological injury, and other conditions requiring non-cardiac surgery may increase the risk or even delay the primary cardiac repair in neonates. Similarly, Blalock–Taussig–Thomas shunt procedures performed in infants with prematurity, low weight, and hypoplastic pulmonary arteries have increased risk of complications such as pulmonary artery stenosis and pulmonary overcirculation. Hence, this becomes a dillema as these patients are at increased risk of morbidity and mortality for either primary repair bridging treatment of Blalock–Taussig–Thomas shunt. Reference Sandoval, Chaturvedi and Benson2,Reference Castleberry, Gudausky, Berger, Tweddell and Pelech78 Right ventricular outflow tract stenting even becomes the primary palliation procedure for tetralogy of Fallot in some centres. Reference Quandt, Ramchandani and Stickley1

A few studies reported that right ventricular outflow tract stenting may alleviate both infundibular and pulmonary valve obstruction, increasing oxygen saturation and delaying the requirement for early surgery. This procedure also allowed pulmonary arterial and somatic growth with clinical results comparable to early surgical repair in more favourable patients. Reference Sandoval, Chaturvedi and Benson2,Reference Castleberry, Gudausky, Berger, Tweddell and Pelech78 A systematic review and meta-analysis reported that patients with marked obstruction of pulmonary blood flow, low birth weight, or small pulmonary artery size who underwent right ventricular outflow tract stenting had improved conditions as well as pulmonary blood flow. Reference Laurentius, Wiyono, Dominique Subali and Natalia Siagian79 Similar to other palliative interventions, right ventricular outflow tract has risk of developing pulmonary reperfusion injury after reperfusion of the previously underperfused pulmonary circulation. One study of tetralogy of Fallot patients undergoing right ventricular outflow tract stenting reported 9 out of 12 children (75%) develops post-procedural pulmonary reperfusion injury which mostly resolved in 72 hours. Reference Garcia, Sandoval Jones and Aristizabal Villa80 A review hypothesised that the immediate death observed during early post-operative monitoring of right ventricular outflow tract stenting may be caused by acute respiratory distress syndrome induced by pulmonary reperfusion injury or sepsis. Reference Laurentius, Wiyono, Dominique Subali and Natalia Siagian79 Gradual increment of pulmonary blood flow by redilatation of stent may be required in cases of severe form of tetralogy of Fallot physiology with late presentation to reduce the risk of perioperative death from pulmonary reperfusion injury. Reference Begum, Bhuiyan and Khan81

Management of acute pulmonary reperfusion injury in post-palliative procedure

Management of pulmonary reperfusion injury post-palliative procedure should focus on diuretics, oxygen, and mechanical ventilatory support until 72 hours. Monitoring should be done in the ICU as titration of diuretics should be done carefully. Reference Bergersen and Lock26 Although conservative fluid management using diuretics in the children has shown to lower mortality rate as well as shorten ICU stay and mechanical ventilation days, the optimal guideline for fluid management in pulmonary reperfusion injury remains controversial. It is due to the heterogeneous nature of population phenotypes with the disease of interest in real-life settings. Reference Lee, Corl and Levy82

In any chosen mode for ventilatory support, parameter controls must maintain the concept of open-lung ventilation or permissive hypercapnia strategy. This setting would provide a protective effect on the lung, instead of aggravating the lung damage. The recommended adjustment of alveolar protective tidal volume ranges between 4 and 8 mL/kg of ideal weight to prevent unwanted volutrauma. Supplemental oxygen must be adjusted to maintain saturation between 92 and 97%, while ensuring the positive end-expiratory pressure stays below 10 mmHg. Reference Coronado-Muñoz and Escalante-Kanashiro83

Severe heart failure due to pulmonary overflow is uncommon but may occur if the flow rate is too high thus requiring reduction. This condition may require the use of systemic vasodilator if the systolic blood pressure is above 75 mmHg and diastolic pressure above 30 mmHg. Milrinone may be started from 0.3 to 1 mcg/kg/min in neonates. After milrinone is titrated up to 1 mcg/kg/min, sodium nitroprusside may be started from 0.5 mcg/kg/min and titrated up to 5 mcg/kg/min. Reference Ismail, Almazmi and Khokhar45 Although the concept of bigger shunt size correlating with overshunting is still debatable, minimising overshunting by reducing the shunt size is an option. This may be achieved by replacing the shunt, decreasing the diameter of ductal stent by adding several stents (Russian Doll technique), or increasing the length of tube. Multiple coronary stents would be required to decrease the stent diameter, while increasing the length of the tube can be achieved by using a covered stent. By reducing the diameter and lengthening the duct, this will increase the resistance and decrease the flow rate. Reference Yamashita, Akamine and Sumida84Reference Dreyer, Mühlfeld and Fehrenbach88

There are many potential treatments for thetherapy and prevention of pulmonary reperfusion injury based on its complex pathophysiology. Various methods of preconditioning such as ischaemic, hyperthermic, and chemical preconditioning may stimulate the production of heat-shock proteins in the cells which give protective benefits prior to ischaemic reperfusion insult. Therapeutic gases such as nitric oxide, hydrogen, hydrogen sulfide, and carbon monoxide, administration of prostaglandin E1, intravenous injection of soluble complement receptor type 1, administration of platelet-activating factor antagonists, and exogenous surfactant therapy also show potential in prophylaxis and therapy of pulmonary reperfusion injury. All these treatments are still in research, mostly only until in vivo experiments; hence, their clinical evidence remains unelucidated. Reference Yamashita, Akamine and Sumida84Reference Dreyer, Mühlfeld and Fehrenbach88

Conclusion

Pulmonary reperfusion injury is a complex disease trigerred by ischaemia and reperfusion involving multiple molecular and cellular mechanism. The risk of pulmonary reperfusion injury has been recognised a long time ago and is still an important issue today. The incidence of pulmonary reperfusion injury varies depending on multifactors such as the type of cyanotic CHD, baseline condition of the patients including the cardiac and pulmonary function, type of palliative intervention, and other risk factors. The lack of specific diagnosis criteria for pulmonary reperfusion injury also leads to the unawareness of this disease entity. However, pulmonary reperfusion injury does occur and may lead to morbidity and mortality in post-palliative procedures in oligaemic cyanotic CHD. Understanding the risk and mechanisms of this complication will prepare the heart team for this complication, improve periprocedural management and post-operative care, and eventually improve the outcome for the patients. It is necessary to construct a general consensus for pulmonary reperfusion injury as currently the cornerstone of management for pulmonary reperfusion injury is supportive therapy to prevent further deterioration of lung injury. Further studies are also required to establish the benefit of novel and potential treatment for prophylaxis and therapy of pulmonary reperfusion injury.

Acknowledgements

We would like to thank those who have supported us in the making of this review. We are especially grateful to the Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Indonesia, for their guidance and assistance in teaching the authors about research methodology as well as literature search and data analysis for a review.

Financial support

This research received no specific grant from any funding agency, commercial, or not-for-profit sectors.

Competing interests

None.

Author contribution

Conceptualisation: S.N.S; literature search and data analysis: S.N.S, M.S.Y.D., B.E.P., and C.C.; supervising: S.N.S.; drafting and revising: S.N.S. and C.C.; All authors have read and agreed to the published version of the manuscript.

References

Quandt, D, Ramchandani, B, Stickley, J, et al. Stenting of the right ventricular outflow tract promotes better pulmonary arterial growth compared with modified blalock-taussig shunt palliation in tetralogy of Fallot-type lesions. JACC Cardiovasc Interv 2017; 10: 17741784.10.1016/j.jcin.2017.06.023CrossRefGoogle ScholarPubMed
Sandoval, JP, Chaturvedi, RR, Benson, L, et al. Right ventricular outflow tract stenting in tetralogy of Fallot infants with risk factors for early primary repair. Circ Cardiovasc Interv 2016; 9: e003979.10.1161/CIRCINTERVENTIONS.116.003979CrossRefGoogle ScholarPubMed
Li, D, Zhou, X, Li, M. Arterial duct stent versus surgical shunt for patients with duct-dependent pulmonary circulation: a meta-analysis. BMC Cardiovasc Disord 2021; 21: 9.10.1186/s12872-020-01817-2CrossRefGoogle ScholarPubMed
Asija, R, Roth, SJ, Hanley, FL, et al. Reperfusion pulmonary edema in children with tetralogy of Fallot, pulmonary atresia, and major aortopulmonary collateral arteries undergoing unifocalization procedures: a pilot study examining potential pathophysiologic mechanisms and clinical significance. J Thorac Cardiovasc Surg 2014; 148: 15601565.10.1016/j.jtcvs.2014.01.017CrossRefGoogle ScholarPubMed
Yacouby, S, Meador, M, Mossad, E. Lung reperfusion injury in patients after balloon angioplasty for pulmonary artery stenosis. J Cardiothorac Vasc Anesth 2014; 28: 502505.10.1053/j.jvca.2013.10.024CrossRefGoogle ScholarPubMed
Ejiri, K, Ogawa, A, Fujii, S, Ito, H, Matsubara, H. Vascular injury is a major cause of lung injury after balloon pulmonary angioplasty in patients with chronic thromboembolic pulmonary hypertension. Circ Cardiovasc Interv 2018; 11: e005884.10.1161/CIRCINTERVENTIONS.117.005884CrossRefGoogle Scholar
Maskatia, SA, Feinstein, JA, Newman, B, Hanley, FL, Roth, SJ. Pulmonary reperfusion injury after the unifocalization procedure for tetralogy of Fallot, pulmonary atresia, and major aortopulmonary collateral arteries. J Thorac Cardiovasc Surg 2012; 144: 184189.10.1016/j.jtcvs.2011.12.030CrossRefGoogle ScholarPubMed
Cowled, P, Fitridge, R. Pathophysiology of Reperfusion Injury. In: Fitridge, R, Thompson, M (eds). Mechanisms of Vascular Disease: A Reference Book for Vascular Specialists, University of Adelaide Press, Adelaide (AU), 2011.Google Scholar
Zarbock, A, Eroglu, A, Erturk, E, Ince, C, Westphal, M. Ischemia-reperfusion injury and anesthesia. Biomed Res Int 2014; 2014: 980318.10.1155/2014/980318CrossRefGoogle ScholarPubMed
den Hengst, WA, Gielis, JF, Lin, JY, Van Schil, PE, De Windt, LJ, Moens, AL. Lung ischemia-reperfusion injury: a molecular and clinical view on a complex pathophysiological process. Am J Physiol Heart Circ Physiol 2010; 299: H128399.10.1152/ajpheart.00251.2010CrossRefGoogle ScholarPubMed
Weyker, PD, Webb, CA, Kiamanesh, D, Flynn, BC. Lung ischemia reperfusion injury: a bench-to-bedside review. Semin Cardiothorac Vasc Anesth 2013; 17: 2843.10.1177/1089253212458329CrossRefGoogle ScholarPubMed
Khemani, RG, Patel, NR, Bart, RD 3rd, Newth, CJL. Comparison of the pulse oximetric saturation/fraction of inspired oxygen ratio and the PaO2/fraction of inspired oxygen ratio in children. Chest 2009; 135: 662668.10.1378/chest.08-2239CrossRefGoogle ScholarPubMed
El-Khatib, MF, Jamaleddine, GW. A new oxygenation index for reflecting intrapulmonary shunting in patients undergoing open-heart surgery. Chest 2004; 125: 592596.10.1378/chest.125.2.592CrossRefGoogle ScholarPubMed
Christie, JD, Carby, M, Bag, R, Corris, P, Hertz, M, Weill, D. Report of the ISHLT working group on primary lung graft dysfunction part II: definition. A consensus statement of the international society for heart and lung transplantation. J Heart Lung Transplant 2005; 24: 14541459.10.1016/j.healun.2004.11.049CrossRefGoogle Scholar
Eltzschig, HK, Carmeliet, P. Hypoxia and inflammation. N Engl J Med 2011; 364: 656665.10.1056/NEJMra0910283CrossRefGoogle ScholarPubMed
Yellon, DM, Hausenloy, DJ. Myocardial reperfusion injury. N Engl J Med 2007; 357: 11211135.10.1056/NEJMra071667CrossRefGoogle ScholarPubMed
Yemisci, M, Gursoy-Ozdemir, Y, Vural, A, Can, A, Topalkara, K, Dalkara, T. Pericyte contraction induced by oxidative-nitrative stress impairs capillary reflow despite successful opening of an occluded cerebral artery. Nat Med 2009; 15: 10311037.10.1038/nm.2022CrossRefGoogle ScholarPubMed
Wu, MY, Yiang, GT, Liao, WT, et al. Current mechanistic concepts in ischemia and reperfusion injury. Cell Physiol Biochem 2018; 46: 16501667.10.1159/000489241CrossRefGoogle ScholarPubMed
Mura, M, Andrade, CF, Han, B, et al. Intestinal ischemia-reperfusion-induced acute lung injury and oncotic cell death in multiple organs. Shock 2007; 28: 227238.10.1097/01.shk.0000278497.47041.e3CrossRefGoogle ScholarPubMed
Deffebach, ME, Charan, NB, Lakshminarayan, S, Butler, J. The bronchial circulation. Small, but a vital attribute of the lung. Am Rev Respir Dis 1987; 135: 463481.Google ScholarPubMed
Esme, H, Fidan, H, Koken, T, Solak, O. Effect of lung ischemia--reperfusion on oxidative stress parameters of remote tissues. Eur J Cardiothorac Surg 2006; 29: 294298.10.1016/j.ejcts.2005.12.008CrossRefGoogle ScholarPubMed
Adkins, WK, Taylor, AE. Role of xanthine oxidase and neutrophils in ischemia-reperfusion injury in rabbit lung. J Appl Physiol 1990; 69: 20122018.10.1152/jappl.1990.69.6.2012CrossRefGoogle ScholarPubMed
Eckenhoff, RG, Dodia, C, Tan, Z, Fisher, AB. Oxygen-dependent reperfusion injury in the isolated rat lung. J Appl Physiol 1992; 72: 14541460.10.1152/jappl.1992.72.4.1454CrossRefGoogle ScholarPubMed
Fisher, AB, Dodia, C, Tan, ZT, Ayene, I, Eckenhoff, RG. Oxygen-dependent lipid peroxidation during lung ischemia. J Clin Invest 1991; 88: 674679.10.1172/JCI115352CrossRefGoogle ScholarPubMed
Verma, S, Fedak, PW, Weisel, RD, et al. Fundamentals of reperfusion injury for the clinical cardiologist. Circulation 2002; 105: 23322336.10.1161/01.CIR.0000016602.96363.36CrossRefGoogle ScholarPubMed
Bergersen, L, Lock, JE. What is the current option of first choice for treatment of pulmonary arterial stenosis? Cardiol Young 2006; 16: 329338.10.1017/S1047951106000679CrossRefGoogle ScholarPubMed
Ho, AB, Salmon, TP, Hribernik, I, Hayes, N, Thomson, JD, Bentham, JR. A case series of three patients with unilateral disconnected pulmonary artery supplied by an ipsilateral patent ductus arteriosus: neonatal ductal stenting as palliation to preserve pulmonary arterial patency. Eur Heart J Case Rep 2020; 4: 17.10.1093/ehjcr/ytaa422CrossRefGoogle ScholarPubMed
Silverman, NA, Kohler, J, Levitsky, S, Pavel, DG, Fang, RB, Feinberg, H. Chronic hypoxemia depresses global ventricular function and predisposes to the depletion of high-energy phosphates during cardioplegic arrest: implications for surgical repair of cyanotic congenital heart defects. Ann Thorac Surg 1984; 37: 304308.10.1016/S0003-4975(10)60735-7CrossRefGoogle Scholar
Laubach, VE, Sharma, AK. Mechanisms of lung ischemia-reperfusion injury. Curr Opin Organ Transplant 2016; 21: 246252.10.1097/MOT.0000000000000304CrossRefGoogle ScholarPubMed
Ossa Galvis, MM, Bhakta, RT, Tarmahomed, A, Mendez, MD. Cyanotic Heart Disease. StatPearls. StatPearls Publishing, Treasure Island (FL), 2022.Google Scholar
Teoh, KH, Mickle, DA, Weisel, RD, et al. Effect of oxygen tension and cardiovascular operations on the myocardial antioxidant enzyme activities in patients with tetralogy of Fallot and aorta-coronary bypass. J Thorac Cardiovasc Surg 1992; 104: 159164.10.1016/S0022-5223(19)34848-2CrossRefGoogle ScholarPubMed
Hammon, JW Jr. Myocardial protection in the immature heart. Ann Thorac Surg 1995; 60: 839842.10.1016/0003-4975(95)00573-4CrossRefGoogle ScholarPubMed
Asada, D, Itoi, T, Nakamura, A, Hamaoka, K. Tolerance to ischemia reperfusion injury in a congenital heart disease model. Pediatr Int 2016; 58: 12661273.10.1111/ped.13022CrossRefGoogle Scholar
Allen, BS, Ilbawi, MN. Hypoxia, reoxygenation and the role of systemic leukodepletion in pediatric heart surgery. Perfusion 2001; 16: 1929.10.1177/026765910101600i104CrossRefGoogle ScholarPubMed
Walker, CP, Bateman, CJ, Rigby, ML, Brookes, CI. Acute pulmonary edema after percutaneous balloon valvuloplasty for pulmonary valve stenosis. J Cardiothorac Vasc Anesth 2001; 15: 480482.10.1053/jcan.2001.24998CrossRefGoogle ScholarPubMed
Mohanty, S, Pandit, BN, Tyagi, S. Balloon pulmonary valvotomy--not just a simple balloon dilatation. Indian Heart J 2014; 66: 462465.10.1016/j.ihj.2014.05.007CrossRefGoogle ScholarPubMed
Tefera, E, Qureshi, SA, Bermudez-Cañete, R, Rubio, L. Percutaneous balloon dilation of severe pulmonary valve stenosis in patients with cyanosis and congestive heart failure. Catheter Cardiovasc Interv 2014; 84: E715.10.1002/ccd.25324CrossRefGoogle ScholarPubMed
Ostovan, MA, Kamali, M, Zolghadrasli, A. A case of fatal acute lung injury after balloon valvuloplasty of pulmonary stenosis: case report and review of literature. J Cardiovasc Thorac Res 2015; 7: 7880.10.15171/jcvtr.2015.18CrossRefGoogle ScholarPubMed
Blake, K, Yancy, CW. Change the name of the blalock-taussig shunt to blalock-thomas-taussig shunt. JAMA Surg 2022; 157: 287288.10.1001/jamasurg.2021.5611CrossRefGoogle ScholarPubMed
Kaskinen, AK, Keski-Nisula, J, Martelius, L, et al. Lung injury after neonatal congenital cardiac surgery is mild and modifiable by corticosteroids. J Cardiothorac Vasc Anesth 2021; 35: 21002107.10.1053/j.jvca.2021.01.017CrossRefGoogle ScholarPubMed
Caputo, M, Mokhtari, A, Rogers, CA, et al. The effects of normoxic versus hyperoxic cardiopulmonary bypass on oxidative stress and inflammatory response in cyanotic pediatric patients undergoing open cardiac surgery: a randomized controlled trial. J Thorac Cardiovasc Surg 2009; 138: 206214.10.1016/j.jtcvs.2008.12.028CrossRefGoogle ScholarPubMed
Okita, Y, Miki, S, Kusuhara, K, et al. Acute pulmonary edema after blalock-taussig anastomosis. Ann Thorac Surg 1992; 53: 684685.10.1016/0003-4975(92)90334-ZCrossRefGoogle ScholarPubMed
Tassig, HB, Crocetti, A, Eshaghpour, E, et al. Long-time observations on the blalock-taussig operation. 3. Common complications. Johns Hopkins Med J 1971; 129: 274289.Google ScholarPubMed
Hofschire, PJ, Rosenquist, GC, Ruckerman, RN, Moller, JH, Edwards, JE. Pulmonary vascular disease complicating the blalock-taussig anastomosis. Circulation 1977; 56: 124126.10.1161/01.CIR.56.1.124CrossRefGoogle ScholarPubMed
Ismail, SR, Almazmi, MM, Khokhar, R, et al. Effects of protocol-based management on the post-operative outcome after systemic to pulmonary shunt. Egypt Heart J 2018; 70: 271278.10.1016/j.ehj.2018.09.007CrossRefGoogle ScholarPubMed
Dirks, V, Prêtre, R, Knirsch, W, et al. Modified blalock taussig shunt: a not-so-simple palliative procedure. Eur J Cardiothorac Surg 2013; 44: 10961102.10.1093/ejcts/ezt172CrossRefGoogle ScholarPubMed
Wardoyo, S, Makdinata, W, Wijayanto, MA. Perioperative strategy to minimize mortality in neonatal modified blalock-taussig–Thomas shunt: a literature review. Cirugía Cardiovascular 2022; 29: 3135.10.1016/j.circv.2021.04.002CrossRefGoogle Scholar
Qu, X, Li, Q, Wang, X, Yang, X, Wang, D. N-acetylcysteine attenuates cardiopulmonary bypass-induced lung injury in dogs. J Cardiothorac Surg 2013; 8: 107.10.1186/1749-8090-8-107CrossRefGoogle ScholarPubMed
Apostolakis, EE, Koletsis, EN, Baikoussis, NG, Siminelakis, SN, Papadopoulos, GS. Strategies to prevent intraoperative lung injury during cardiopulmonary bypass. J Cardiothorac Surg 2010; 5: 1.10.1186/1749-8090-5-1CrossRefGoogle ScholarPubMed
Luc, JGY, Aboelnazar, NS, Himmat, S, et al. A Leukocyte filter does not provide further benefit during ex vivo lung perfusion. Asaio J 2017; 63: 672678.10.1097/MAT.0000000000000550CrossRefGoogle Scholar
Ziyaeifard, M, Alizadehasl, A, Massoumi, G. Modified ultrafiltration during cardiopulmonary bypass and postoperative course of pediatric cardiac surgery. Res Cardiovasc Med 2014; 3: e17830.Google ScholarPubMed
Slottosch, I, Liakopoulos, O, Kuhn, E, et al. Controlled lung reperfusion to reduce pulmonary ischaemia/reperfusion injury after cardiopulmonary bypass in a porcine model. Interact Cardiovasc Thorac Surg 2014; 19: 962970.10.1093/icvts/ivu270CrossRefGoogle ScholarPubMed
Gauduel, Y, Menasche, P, Duvelleroy, M. Enzyme release and mitochondrial activity in reoxygenated cardiac muscle: relationship with oxygen-induced lipid peroxidation. Gen Physiol Biophys 1989; 8: 327340.Google ScholarPubMed
Joachimsson, PO, Sjöberg, F, Forsman, M, Johansson, M, Ahn, HC, Rutberg, H. Adverse effects of hyperoxemia during cardiopulmonary bypass. J Thorac Cardiovasc Surg 1996; 112: 812819.10.1016/S0022-5223(96)70069-7CrossRefGoogle ScholarPubMed
Sasson, L, Sherman, A, Ezri, T, et al. Mode of ventilation during cardiopulmonary bypass does not affect immediate postbypass oxygenation in pediatric cardiac patients. J Clin Anesth 2007; 19: 429433.10.1016/j.jclinane.2007.03.007CrossRefGoogle Scholar
Arthur E.Baue, ASG, Graeme, LH, Hillel, L, Keith, SN. Glenn’s Thoracic and Cardiovascular Surgery. Appleton & Lange, Minneapolis, 1991.Google Scholar
Qiu, FH, Wada, K, Stahl, GL, Serhan, CN. IMP and AMP deaminase in reperfusion injury down-regulates neutrophil recruitment. Proc Natl Acad Sci U S A 2000; 97: 42674272.10.1073/pnas.97.8.4267CrossRefGoogle ScholarPubMed
Hu, J, Li, P, Chen, X, Yan, J, Zhang, J, Zhang, C. Effects of modified ultrafiltration and conventional ultrafiltration combination on perioperative clinical outcomes in pediatric cardiac surgery: a meta-analysis. Medicine (Baltimore) 2021; 100: e24221.10.1097/MD.0000000000024221CrossRefGoogle ScholarPubMed
Kuratani, N, Bunsangjaroen, P, Srimueang, T, Masaki, E, Suzuki, T, Katogi, T. Modified versus conventional ultrafiltration in pediatric cardiac surgery: a meta-analysis of randomized controlled trials comparing clinical outcome parameters. J Thorac Cardiovasc Surg 2011; 142: 861867.10.1016/j.jtcvs.2011.04.001CrossRefGoogle ScholarPubMed
Whitlock, RP, Devereaux, PJ, Teoh, KH, et al. Methylprednisolone in patients undergoing cardiopulmonary bypass (SIRS): a randomised, double-blind, placebo-controlled trial. Lancet 2015; 386: 12431253.10.1016/S0140-6736(15)00273-1CrossRefGoogle ScholarPubMed
Whitlock, R, Teoh, K, Vincent, J, et al. Rationale and design of the steroids in cardiac surgery trial. Am Heart J 2014; 167: 660665.10.1016/j.ahj.2014.01.018CrossRefGoogle ScholarPubMed
Glumac, S, Kardum, G, Sodic, L, Supe-Domic, D, Karanovic, N. Effects of dexamethasone on early cognitive decline after cardiac surgery: a randomised controlled trial. Eur J Anaesthesiol 2017; 34: 776784.10.1097/EJA.0000000000000647CrossRefGoogle ScholarPubMed
Glumac, S, Kardum, G, Sodic, L, et al. Longitudinal assessment of preoperative dexamethasone administration on cognitive function after cardiac surgery: a 4-year follow-up of a randomized controlled trial. BMC Anesthesiol 2021; 21: 129.10.1186/s12871-021-01348-zCrossRefGoogle ScholarPubMed
Lodge, AJ, Chai, PJ, Daggett, CW, Ungerleider, RM, Jaggers, J. Methylprednisolone reduces the inflammatory response to cardiopulmonary bypass in neonatal piglets: timing of dose is important. J Thorac Cardiovasc Surg 1999; 117: 515522.10.1016/S0022-5223(99)70331-4CrossRefGoogle ScholarPubMed
Chaney, MA, Durazo-Arvizu, RA, Nikolov, MP, Blakeman, BP, Bakhos, M. Methylprednisolone does not benefit patients undergoing coronary artery bypass grafting and early tracheal extubation. J Thorac Cardiovasc Surg 2001; 121: 561569.10.1067/mtc.2001.112343CrossRefGoogle Scholar
Chaney, MA, Nikolov, MP, Blakeman, B, Bakhos, M, Slogoff, S. Pulmonary effects of methylprednisolone in patients undergoing coronary artery bypass grafting and early tracheal extubation. Anesth Analg 1998; 87: 2733.10.1213/00000539-199807000-00007CrossRefGoogle ScholarPubMed
Chai, T, Zhuang, X, Tian, M, et al. Meta-analysis: shouldn’t prophylactic corticosteroids be administered during cardiac surgery with cardiopulmonary bypass? Front Surg 2022; 9: 832205.10.3389/fsurg.2022.832205CrossRefGoogle ScholarPubMed
Perchermeier, S, Tassani-Prell, P. The use of corticosteroids for cardiopulmonary bypass in adults. Curr Anesthesiol Rep 2021; 11: 292297.10.1007/s40140-021-00468-zCrossRefGoogle Scholar
Hall, RI, Smith, MS, Rocker, G. The systemic inflammatory response to cardiopulmonary bypass: pathophysiological, therapeutic, and pharmacological considerations. Anesth Analg 1997; 85: 766782.10.1213/00000539-199710000-00011CrossRefGoogle ScholarPubMed
Richter, JA, Meisner, H, Tassani, P, Barankay, A, Dietrich, W, Braun, SL. Drew-anderson technique attenuates systemic inflammatory response syndrome and improves respiratory function after coronary artery bypass grafting. Ann Thorac Surg 2000; 69: 7783.10.1016/S0003-4975(99)01131-5CrossRefGoogle ScholarPubMed
Tassani, P, Richter, JA, Barankay, A, et al. Does high-dose methylprednisolone in aprotinin-treated patients attenuate the systemic inflammatory response during coronary artery bypass grafting procedures? J Cardiothorac Vasc Anesth 1999; 13: 165172.10.1016/S1053-0770(99)90081-2CrossRefGoogle ScholarPubMed
Fitridge, R. Mechanisms of Vascular Disease: A Textbook for Vascular Specialists. Springer, Adelaide, 2020, 755.10.1007/978-3-030-43683-4CrossRefGoogle Scholar
Glatz, AC, Petit, CJ, Goldstein, BH, et al. Comparison between patent ductus arteriosus stent and modified blalock-taussig shunt as palliation for infants with ductal-dependent pulmonary blood flow: insights from the congenital catheterization research collaborative. Circulation 2018; 137: 589601.10.1161/CIRCULATIONAHA.117.029987CrossRefGoogle ScholarPubMed
Bentham, JR, Zava, NK, Harrison, WJ, et al. Duct stenting versus modified blalock-taussig shunt in neonates with duct-dependent pulmonary blood flow: associations with clinical outcomes in a multicenter national study. Circulation 2018; 137: 581588.10.1161/CIRCULATIONAHA.117.028972CrossRefGoogle Scholar
Nasser, BA, Abdulrahman, M, Qwaee, AAL, Alakfash, A, Mohamad, T, Kabbani, MS. Impact of stent of ductus arteriosus and modified blalock-taussig shunt on pulmonary arteries growth and second-stage surgery in infants with ductus-dependent pulmonary circulation. J Saudi Heart Assoc 2020; 32: 8692.Google ScholarPubMed
Bahaidarah, S, Al-Ata, J, Alkhushi, N, et al. Outcome of ductus arteriosus stenting including vertical tubular and convoluted tortuous ducts with emphasis on technical considerations. Egypt Heart J 2021; 73: 83.10.1186/s43044-021-00210-4CrossRefGoogle ScholarPubMed
Alwi, M. Stenting the ductus arteriosus: case selection, technique and possible complications. Ann Pediatr Cardiol 2008; 1: 3845.10.4103/0974-2069.41054CrossRefGoogle ScholarPubMed
Castleberry, CD, Gudausky, TM, Berger, S, Tweddell, JS, Pelech, AN. Stenting of the right ventricular outflow tract in the high-risk infant with cyanotic teratology of Fallot. Pediatr Cardiol 2014; 35: 423430.10.1007/s00246-013-0796-zCrossRefGoogle ScholarPubMed
Laurentius, A, Wiyono, L, Dominique Subali, A, Natalia Siagian, S. Evaluation of right ventricular outflow tract stenting as palliative treatment in severely cyanotic tetralogy of fallot: a systematic review and meta-analysis of observational studies. J Tehran Heart Cent 2021; 16: 135146.Google ScholarPubMed
Garcia, O, Sandoval Jones, J, Aristizabal Villa, G. Pulmonary reperfusion injury following right ventricular outflow tract stenting. Eur Soc Rad 2017; 2017: B-1379.Google Scholar
Begum, NNF, Bhuiyan, NI, Khan, AA. Stenting of right ventricular out flow tract: analysis of 32 Cases from catheterization laboratory of a paediatric cardiac centre. Bangladesh Heart J 2020; 35: 15.10.3329/bhj.v35i1.49136CrossRefGoogle Scholar
Lee, J, Corl, K, Levy, MM. Fluid therapy and acute respiratory distress syndrome. Crit Care Clin 2021; 37: 867875.10.1016/j.ccc.2021.05.012CrossRefGoogle ScholarPubMed
Coronado-Muñoz, Á., Escalante-Kanashiro, R. Pediatric acute respiratory distress syndrome: how to protect the lungs during mechanical ventilation? Bol Med Hosp Infant Mex 2021; 78: 181190.Google ScholarPubMed
Yamashita, H, Akamine, S, Sumida, Y, et al. Inhaled nitric oxide attenuates apoptosis in ischemia-reperfusion injury of the rabbit lung. Ann Thorac Surg 2004; 78: 292297.10.1016/j.athoracsur.2003.12.025CrossRefGoogle ScholarPubMed
de Perrot, M, Fischer, S, Liu, M, et al. Prostaglandin E1 protects lung transplants from ischemia-reperfusion injury: a shift from pro- to anti-inflammatory cytokines. Transplantation 2001; 72: 15051512.10.1097/00007890-200111150-00006CrossRefGoogle ScholarPubMed
Gillinov, AM, DeValeria, PA, Winkelstein, JA, et al. Complement inhibition with soluble complement receptor type 1 in cardiopulmonary bypass. Ann Thorac Surg 1993; 55: 619624.10.1016/0003-4975(93)90264-ICrossRefGoogle ScholarPubMed
Curtin, ML. Current status of platelet-activating factor antagonists. Expert Opin Ther Pat 1998; 8: 703711.10.1517/13543776.8.6.703CrossRefGoogle Scholar
Dreyer, N, Mühlfeld, C, Fehrenbach, A, et al. Exogenous surfactant application in a rat lung ischemia reperfusion injury model: effects on edema formation and alveolar type II cells. Respir Res 2008; 9: 5.10.1186/1465-9921-9-5CrossRefGoogle Scholar
Figure 0

Figure 1. Classification of cyanotic CHD. *ASD = atrial septal defect; AVSD = atrioventricular septal defect; BTTS = Blalock–Taussig–Thomas shunt; ccTGA = congenitally corrected transposition of the great arteries; DORV = double-outlet right ventricle; HLHS = hypoplastic left heart syndrome; PAH = pulmonary artery hypertension; PBF = pulmonary blood flow; PDA = patent ductus arteriosus; PS = pulmonary stenosis; PV = pulmonary vein; RVOT = right ventricular outflow tract; TAPVD = total anomalous of pulmonary vein drainage; TGA = transposition of the great arteries; VSD = ventricular septal defect.29,30