Submit manuscript...
Journal of
eISSN: 2373-6437

Anesthesia & Critical Care: Open Access

Correspondence:

Received: January 01, 1970 | Published: ,

Citation: DOI:

Download PDF

Abstract

CSA-AKI is common after cardiac surgery, and even small increases in serum creatinine are associated with significant increases morbidity and mortality. A variety of risk factors have been identified for the development of CSA-AKI, but a precise precipitating event or events which lead to this complication have yet to be identified. Moreover, current diagnostic methods for detecting AKI lag behind the point of injury by 24-48 hours. Current technologies to close this gap include biomarkers and cerebral autoregulation monitoring data, but clear methodologies for their use have yet to be perfected. There have similarly been many failed interventions aimed at ameliorating AKI, though goal-directed perfusion strategies on CPB may hold promise.

Keywords: kidney, renal function failure, acute kidney injury, cardiac surgery

Abbreviations

CSA-AKI, cardiac surgery associated acute kidney injury; ADQI, acute dialysis quality initiative; RIFLE, risk-injury-failure-loss-end stage renal disease; AKIN, acute kidney injury network; CPB, cardiopulmonary bypass; CHF, congestive heart failure; IGFBP7, insulin-like growth-factor binding protein 7; TIMP-2, tissue inhibitor metallo proteinases; RRT, renal replacement therapy

Scope of the problem

Cardiac surgery associated acute kidney injury (CSA-AKI) develops in 5% to 40% of patients.1-5 The precise etiology of CSA-AKI is unclear, but can result from a variety of factors including ischemia resulting from hypotension, hemorrhage, or circulatory arrest, nephrotoxic agents, and the development of postoperative complications including sepsis. Unfortunately, mortality rates for new patients requiring dialysisremain staggeringly high, at around 1/3 in mixed patient populations.6,7 Patients who double their serum creatinine or need acute dialysis have a 2 to 5-fold higher risk of death.8 Even mild or subclinical deteriorations in renal function profoundly increase the risk for major adverse outcomes after cardiac surgery.3,4,9,10

Definition of renal failure in cardiac surgery

In 2004 the American Society of Nephrology Renal Research Group introduced the term “acute kidney injury” (AKI) in an effort to systematize terminology and risk stratification for patients. This terminology was adopted to reflect the entire spectrum of the disease: from minimal elevations in serum creatinine to anuric renal failure; from functional deviations to structural changes; and from pre-renal azotemia to acute tubular necrosis. A consensus definition of AKI was proposed by the Acute Dialysis Quality Initiative (ADQI), which introduced the RIFLE (Risk-Injury-Failure-Loss-End Stage Renal Disease) criteria.11 These criteria have since been modified by the Acute Kidney Injury Network (AKIN), which specifies a timeframe of 48 hours within which AKI occurs,along withthree classifications describing increases in serum creatinine relative to baseline(Table 1).12 In most patients after cardiac surgery, serum creatinine will increase only by 0.1-0.2mg/dL. If creatinine increases more than 0.3mg/dL, AKI is diagnosed.

RIFLE

AKIN

Stage

Cr/GFR criteria

Urine output criteria

Stage

Cr criteria

Urine output criteria

Risk

Increased Cr x 1.5-2.0 or GFR decreased > 25%

UO < 0.5mL/kg/hr x 6hr

1

Increased Cr x 1.5-2.0 or ≥ 0.3mg/dL

UO<0.5mL/kg/hr x 6hr

Injury

Increased Cr x 2.0-3.0 or GFR decreased > 50%

UO < 0.5mL/kg/hr x 12hr

2

Increased Cr x 2.0-3.0

UO < 0.5mL/kg/hr x 12hr

Failure

Increased Cr x 3.0 or GFR decreased > 75% or Cr ≥4 mg/dL (with acute rise of ≥ 0.5 mg/dL

UO < 0.3mL/kg/hrx 24 hr or anuria x 12hr

3

Increased Cr x 3.0 or Cr ≥4 mg/dL (with acute rise of ≥ 0.5mg/dL)

UO<0.3mL/kg/hr x24hr or anuria x 12hr

Loss

Persistent ARF =complete loss of renal function for >4weeks

Patients who receive renal replacement therapy (RRT) are considered to have met AKINcritera for stage 3 regardless of the stage they are in prior to RRT initiation.

ESRD

End Stage Renal Disease

Table 1 RIFLE and AKIN classifications for AKI12,40
Increases specified are defined as increases from baseline serum creatinine (i.e.: a patient with AKIN stage 1 AKI will have a serum creatinine 1.5-2.0 times baseline: or 50-100% higher than baseline).
AKI, acute kidney injury; RIFLE, risk-injury-failure-loss guidlines; AKIN, acute kidney injury network guidelines; CR, serum creatinine; UO, urine output; GFR, glomerular filtration rate; ARF, acute renal failure; ESRD, end-stage renal disease; RRT, renal replacement therapy

Risk factors

In general, clinical AKI is thought to be multifactorial, and seemingly minor insults may result in renal failure in high-risk patients. In humans,the exact cause of CSA-AKI is unknown but thought to be precipitated by an ischemic event. Many authors have attempted to identify risk factors for AKI after cardiac surgery. These have been reported to include age,13,14 preoperative renal impairment,14,15 diabetes,16 cerebrovascular disease,16 prolonged cardiopulmonary bypass (CPB) time,13-15,17 valve procedures,13-15,17,18 postoperative hypotension,14 or hemolysis and ensuing pigment nephropathy.19

The kidneys are similar to the brain in that they have highly efficient intrinsic auto regulation mechanisms that maintain an RBF over a wide range (MAP 60 to 120mm Hg) of renal arterial perfusion pressures.20 Disruption of this mechanism causes pre-renal azotemia, a term which has been replaced with concept of volume-responsive AKI by the ADQI and AKIN groups, since pre-renal azotemia and acute tubular necrosis develop in parallel in AKI. The main mechanism is a reduction in renal perfusion with a modest reduction of GFR, an increase in creatinine and BUN, which are often accompanied by oliguria. Non-volume-responsive AKI is thought to be caused by renal ischemia with subsequent rapidly progressive and profound reduction in GFR.21

The issue: when exactly CSA-AKI happens and why?

Currently, there are no evidence-based guidelines to support any specific fluid use or vasoactive medication to improve renal function. Similarly, there are no devices or biomarkers which can identify either optimal renal perfusion or ischemia in real time to prevent CSA-AKI. What we do have is a general consensus that volume depletion and congestive heart failure (CHF) symptoms should be addressed and corrected as part of the management of AKI.22

The major issue with CSA-AKI management, however, is the delay indiagnosis caused by using standard clinical diagnostic criteria (serum creatinine and calculated GFR). These detect AKI when it is already well established, 24-48 hours after the injury. Moreover, serum creatinine has limited sensitivity and specificity because its serum levels depend significantly on non-renal factors such as age, sex and muscle mass. To prevent CSA-AKI, detection of AKI must happen within a “window of opportunity” for interventions, just as now exists for stroke and MI treatment. As such, there is a need to develop a clinically viable, real-time, continuous, and non-invasive monitor to assess renal function or at least renal perfusion.

Novel strategies at detection

The AKI research network evaluated several novel biomarkers in clinical trials with the goal of diagnosing CSA-AKI within the first 24hours after the injury. These biomarkers are neutrophil gelatinase-associated lipocalin (NGAL), cystatin C, kidney injury molecule 1 and interleukin-18.23,24 Their levels increase within 2 hours of injury and denote a 5-6-fold increased risk of AKI.

As of September 5, 2014, Nephro Check, a test by Astute Medical, was approved by the FDA to detect the presence of an insulin-like growth-factor binding protein 7 (IGFBP7) and the tissue inhibitor metallo proteinases (TIMP-2) in the urine, which are associated with acute kidney injury.25 Within 20minutes, the test provides a score based on the amount of the proteins present which correlates to the patient’s risk of developing AKI within 12hours of the test being performed. However, a recent paper called into question whether these markers can adequately detect patients who will develop AKI on the day of surgery, as opposed to the first postoperative day.26

During hypoperfusion, renovascular autoregulation is compromised earlier and more severely than cerebrovascular autoregulation.27 Innovative technology to determine the lower limit of the cerebral autoregulation threshold has been developed and validated at Hopkins. Excursions of MAP below the lower limit of cerebral autoregulation were independently associated with AKI.28 Because of this, observations of cerebral perfusion may be used as a surrogate for renal perfusion.

Novel strategies on prevention and treatment

A list of attempted and failed strategies to prevent CSA-AKI includes:diuretics (furosemide, mannitol);29 renal vasodilators (dopamine, fenoldopan, Ca channel blockers); atrial natriuretic peptides (nesiritide); anti-oxidants (N-acetylcysteine); ACE-inhibitors;30 anti-inflammatory drugs (steroids); ultrafiltration; anti-apoptotic agents; urinary alkalinization by sodium bicarbonate;31 statins;32 human recombinant erythropoietin;33 and remote ischemic pre conditioning.34 Trials conducted on renotropic progenitor cells administration were prematurely terminated by the company (AlloCure).

Currently, the most promising strategy to reduce CSA-AKI is goal-directed perfusion management using oxygen delivery (DO2 level>270ml/min/m2) and its ratio to CO2 production (DO2/VCO2>5.3).35 Maintaining cardiopulmonary bypass (CPB) flows above 54mL/kg/min, mean arterial pressureless than 26mmHg from baseline and avoiding hyperthermia (core temperature >37 degrees Celsius) have been proposed as protective strategies during bypass.19,36,37

Finally, prolonged loss of kidney function requires renal replacement therapy (RRT). Accepted indications for RRT are presented in Table 2. Whether initiation of earlier or prophylactic dialysis prior to the development of symptoms and signs of renal failure offers any clinical or survival benefit is unproven. Several trials have shown that the modality of RRT-either intermittent or continuous-hasno impact on outcomes in patients with AKI.38 Treatment with higher intensity continuous RRT or with early RRT also did not provide any additional benefit.39

1

Refractory fluid overload

2

Hyperkalemia (plasma potassium concentration >6.5mEg/L) or rapidly rising potassium levels

3

Signs of uremia: such as pericarditis: neuropathy: or an otherwise unexplained decline in mental status

4

Metabolic acidosis (pH<7.1)

5

Certain alcohol and drug intoxications

Table 2 Accepted Indications for Renal Replacement Therapy in Patients with AKI

Conclusion

In summary, knowledge of CSA-AKI is critical to the provider caring for cardiac surgery patients. To the extent that etiologies such as hypotension and low oxygen delivery have been identified as risk factors for CSA-AKI, clinicians should apply this knowledge to their routine clinical practice.

Acknowledgments

None.

Conflicts of interest

The authors declare that there are no conflicts of interest.

Funding

None.

References

  1. Brown JR, Cochran RP, MacKenzie TA, et al. Long–term survival after cardiac surgery is predicted by estimated glomerular filtration rate. Ann Thorac Surg. 2008;86(1):4–11.
  2. Chang TI, Leong TK, Boothroyd DB, et al. Acute kidney injury after CABG versus PCI: an observational study using 2 cohorts. J Am Coll Cardiol. 2014;64(10):985–994.
  3. Hobson CE, Yavas S, Segal MS, et al. Acute kidney injury is associated with increased long–term mortality after cardiothoracic surgery. Circulation. 2009;19(18):2444–2453.
  4. Lassnigg A, Schmidlin D, Mouhieddine M, Bachmann LM, Druml W, et al. (2004) Minimal changes of serum creatinine predict prognosis in patients after cardiothoracic surgery: a prospective cohort study. J Am Soc Nephrol 15(6):1597–1605.
  5. Provenchère S, Plantefève G, Hufnagel G, et al. Renal dysfunction after cardiac surgery with normothermic cardiopulmonary bypass:incidence, risk factors, and effect on clinical outcome. Anesth Analg. 2003;96(5):1258–1264.
  6. Xue JL, Daniels F, Star RA, et al. Incidence and mortality of acute renal failure in Medicare beneficiaries, 1992 to 2001. J Am Soc Nephrol. 2006;17(4):1135–1142.
  7. Lameire N, Van Biesen W, Vanholder R. The rise of prevalence and the fall of mortality of patients with acute renal failure: what the analysis of two databases does and does not tell us. J Am Soc Nephrol. 2006;17(4):923–925.
  8. Chertow GM, Levy EM, Hammermeister KE, et al. Independent association between acute renal failure and mortality following cardiac surgery. Am J Med. 1998;104(4):343–348.
  9. Lopez–Delgado JC, Esteve F, Torrado H, et al. Influence of acute kidney injury on short– and long–term outcomes in patients undergoing cardiac surgery:risk factors and prognostic value of a modified RIFLE classification. Crit Care. 2013;17(6):R293.
  10. Elmistekawy E, McDonald B, Hudson C, et al. Clinical impact of mild acute kidney injury after cardiac surgery. Ann Thorac Surg. 2014;98(3):815–822.
  11. Bellomo R, Ronco C, Kellum JA, et al. Acute renal failure – definition, outcome measures, animal models, fluid therapy and information technology needs: the Second International Consensus Conference of the Acute Dialysis Quality Initiative (ADQI) Group. Crit Care. 2004;8(4):R204–212.
  12. Okusa MD, Davenport A. Reading between the (guide) lines––the KDIGO practice guideline on acute kidney injury in the individual patient. Kidney Int. 2014;85(1):39–48.
  13. Sirvinskas E, Andrejaitiene J, Raliene L, et al. Cardiopulmonary bypass management and acute renal failure: risk factors and prognosis. Perfusion. 2008;23(6):323–327.
  14. Suen WS, Mok CK, Chiu SW, et al. Risk factors for development of acute renal failure (ARF) requiring dialysis in patients undergoing cardiac surgery. Angiology. 1998;49(10):789–800.
  15. Mangos GJ, Brown MA, Chan WY, et al. Acute renal failure following cardiac surgery: incidence, outcomes and risk factors. Aust N Z J Med. 1995;25(4):284–289.
  16. Hu Y, Li Z, Chen J, et al. Risk factors for acute kidney injury in patients undergoing same admission coronary angiography and valve replacement. J Card Surg. 2013;28(6):627–631.
  17. Tuttle KR, Worrall NK, Dahlstrom LR, et al. Predictors of ARF after cardiac surgical procedures. Am J Kidney Dis. 2003;41(1):76–83.
  18. Grayson AD, Khater M, Jackson M,  et al.  Valvular heart operation is an independent risk factor for acute renal failure. Ann Thorac Surg. 2003;75(6):1829–1835.              
  19. Haase M, Bellomo R, Story D, et al. Effect of mean arterial pressure, haemoglobin and blood transfusion during cardiopulmonary bypass on post–operative acute kidney injury. Nephrol Dial Transplant. 2012;27(1):153–160.
  20. Walker M, Harrison–Bernard LM, et al. Dynamic interaction between myogenic and TGF mechanisms in afferent arteriolar blood flow autoregulation. Am J Physiol Renal Physiol. 2000;279(5):F858–865.
  21. Garwood S. Cardiac surgery–associated acute renal injury:new paradigms and innovative therapies. J Cardiothorac Vasc Anesth. 2010;24(6):990–1001.
  22. Tolwani A, Paganini E, Joannidis M, et al. Treatment of patients with cardiac surgery associated–acute kidney injury. Int J Artif Organs. 2008;31(2):190–196.
  23. Parikh CR, Coca SG, Thiessen–Philbrook H, et al. Postoperative biomarkers predict acute kidney injury and poor outcomes after adult cardiac surgery. J Am Soc Nephrol. 2011;22(9):1748–1757.
  24. Shaw A. Update on acute kidney injury after cardiac surgery. J Thorac Cardiovasc Surg. 2012;143(3):676–681.
  25. Kashani K, Al–Khafaji A, Ardiles T, et al. Discovery and validation of cell cycle arrest biomarkers in human acute kidney injury. Crit Care. 2013;17(1):R25.
  26. Wetz AJ, Richardt EM, Wand S, et al. Quantification of urinary TIMP–2 and IGFBP–7–an adequate diagnostic test to predict acute kidney injury after cardiac surgery? Crit Care. 2013;19(1):3.
  27. Rhee CJ, Kibler KK, Easley RB, et al. Renovascular reactivity measured by near–infrared spectroscopy. J Appl Physiol. 1985;113(2):307–314.
  28. Ono M, Arnaoutakis GJ, Fine DM, et al. Blood pressure excursions below the cerebral autoregulation threshold during cardiac surgery are associated with acute kidney injury. Crit Care Med. 2013;41(2):464–471.
  29. van der Voort PH, Boerma EC, Koopmans M, et al. Furosemide does not improve renal recovery after hemofiltration for acute renal failure in critically ill patients: a double blind randomized controlled trial. Crit Care Med. 2009;37(2):533–538.
  30. Zacharias M, Gilmore IC, Herbison GP, et al. Interventions for protecting renal function in the perioperative period. Cochrane Database Syst Rev. 2013;3:CD003590.
  31. Tie HT, Luo MZ, Luo MJ, et al. Sodium bicarbonate in the prevention of cardiac surgery–associated acute kidney injury: a systematic review and meta–analysis. Crit Care. 2014;18(5):517.
  32. Prowle JR, Calzavacca P, Licari E, et al. Pilot double–blind, randomized controlled trial of short–term atorvastatin for prevention of acute kidney injury after cardiac surgery. Nephrology (Carlton). 2012;17(3):215–224.
  33. Kim JH, Shim JK, Song JW, et al. Effect of erythropoietin on the incidence of acute kidney injury following complex valvular heart surgery:a double blind, randomized clinical trial of efficacy and safety. Crit Care. 2013;17(5):R254.
  34. Healy DA, Khan WA, Wong CS, et al. Remote preconditioning and major clinical complications following adult cardiovascular surgery: systematic review and meta–analysis. Int J Cardiol. 2014;176(1):20–31.
  35. de Somer F, Mulholland JW, Bryan MR, et al. O2 delivery and CO2 production during cardiopulmonary bypass as determinants of acute kidney injury: time for a goal–directed perfusion management? Crit Care. 2011;15(4):R192.
  36. Kanji HD, Schulze CJ, Hervas–Malo M, et al. Difference between pre–operative and cardiopulmonary bypass mean arterial pressure is independently associated with early cardiac surgery–associated acute kidney injury. J Cardiothorac Surg. 2010;5:71.
  37. Newland RF, Tully PJ, Baker RA.  Hyperthermic perfusion during cardiopulmonary bypass and postoperative temperature are independent predictors of acute kidney injury following cardiac surgery. Perfusion. 2013;28(3):223–231.
  38. Schefold JC, Haehling S, Pschowski R, et al. The effect of continuous versus intermittent renal replacement therapy on the outcome of critically ill patients with acute renal failure (CONVINT):a prospective randomized controlled trial. Crit Care. 2014;18(1):R11.
  39. Bellomo R, Cass A, Cole L, et al. Intensity of continuous renal–replacement therapy in critically ill patients. N Engl J Med. 2009;361(17):1627–1638.
  40. Valette X, du Cheyron DA. critical appraisal of the accuracy of the RIFLE and AKIN classifications in defining "acute kidney insufficiency" in critically ill patients. J Crit Care. 2013;28(2):116–125.
Creative Commons Attribution License

© . This is an open access article distributed under the terms of the, which permits unrestricted use, distribution, and build upon your work non-commercially.