Open access peer-reviewed chapter - ONLINE FIRST

VA ECMO and Drug Intoxication

Written By

Mohamed Nassef and Nashat Abdulhalim

Submitted: 29 December 2023 Reviewed: 04 June 2024 Published: 27 June 2024

DOI: 10.5772/intechopen.1005793

Evolving Therapies and Technologies in Extracorporeal Membrane Oxygenation IntechOpen
Evolving Therapies and Technologies in Extracorporeal Membrane Ox... Edited by Michael S. Firstenberg

From the Edited Volume

Evolving Therapies and Technologies in Extracorporeal Membrane Oxygenation [Working Title]

Dr. Michael S. Firstenberg

Chapter metrics overview

2 Chapter Downloads

View Full Metrics

Abstract

ECMO support can help in maintaining tissue perfusion and oxygenation in acutely intoxicated patients until the drug or toxin is either eliminated by the body with natural metabolism and excretory processes or possibly renal replacement therapy may be instituted to enhance the elimination. Once cardiotoxic substances reach systemic circulation and are distributed in the various tissues, the patient is present with cardiovascular dysfunction, arrhythmia, or cardiovascular collapse. Cardiovascular collapse may require temporary support of the circulatory function. This helps in hepatic detoxification over time while providing reliable tissue perfusion and allowing sufficient antidote circulation. Extracorporeal membrane oxygenation (ECMO) use in cardiotoxic drug overdose is increasing. Calcium channel blockers (CCB) along with β-blockers constitute more than 65% of deaths from cardiovascular medications whereas in the 2021, American Association of Poison Control Centers’ National Poison Data System Annual report, Tricyclic Antidepressant (TCA) accounted for 4705 single exposures and 15 deaths. VA ECMO is effective in critically ill poisoned patients who do not respond to conventional therapies and did not show any improvement along with refractory shock and acute renal failure like in CCB drug overdose and TCA-induced cardiac toxicity and cardiogenic shock, where the recovery depends mainly on maintaining perfusion. The procedure is considered as a lifesaving bridge either to recovery, to antidote, to toxin elimination with renal replacement therapy, or to transplant.

Keywords

  • intoxication
  • veno-arterial
  • ECMO
  • indication
  • shock
  • lifesaving
  • poisoning
  • cardiotoxic
  • calcium channel blockers
  • β-Blockers

1. Introduction

Although there has been a significant advancement in management and improved outcomes of drug overdose-induced cardiovascular shock in the last 3 decades, this is contributed by improvement in bedside hemodynamic monitoring, a better understanding of shock, and aggressive supportive therapy for hemodynamic optimization [1, 2]. Still, the incidence of cardiovascular collapse in patients with acute intoxication is as high as 17% [3]. Sudden cardiac death in a younger and otherwise healthy population is most likely due to intoxication [4]. The onset of cardiovascular effects after drug ingestion is short and depends not only on the ingested quantity but on the severity of the toxic profile, and type of drug as well. One of the most aggressive supportive modalities available is extracorporeal membrane oxygenation (ECMO). ECMO is an external device that supports the cardiopulmonary system by providing oxygenation and cardiac function for a patient in cardiac and respiratory failure. ECMO has been successfully used in all ages for various medical and surgical conditions leading to cardiovascular collapse, respiratory failure, cardiogenic shock, or refractory hypotension [5, 6]. ECMO has also been used in toxicology cases when cardiac arrest or refractory cardiogenic shock develops. This has been studied in both animal models and human cases [7, 8]. In this chapter, we explore the use of VA ECMO for drug intoxication, especially those associated with cardiotoxic effects. Most of the available literature is from the Western countries, which describes the use of ECMO in acute drug intoxication. The experimental evidence with three different trials using lidocaine [9], desipramine [10], and amitriptyline [11] in dogs and swine, respectively, has shown better outcomes with extracorporeal life support (ECLS) in comparison to the conventional treatment using fluid, vasopressor, inotropes, antiarrhythmic agents, etc. The major drawback about all three studies was that the animals were put on ECLS immediately after collapse and the duration of ECLS was short. This may not hold true in actual human intoxication because they are not supported with ECLS immediately and duration of ECLS is longer. However, these results are promising and encourage the use of ECMO in refractory shock due to poisoning.

Advertisement

2. Clinical epidemiology and mortality in drug intoxication

Drug intoxication ranges from mild symptoms to severe and life-threatening cardiovascular dysfunction. Intoxication may be intentional (suicidal, homicidal, misuse, or abuse) or unintentional (accidental) [12]. Usually, 85% of patients presenting with drug intoxication have minor toxic effects and usually do not require aggressive treatment. Intentional exposures have more severe effects and poor outcomes for obvious reasons [13]. An analysis between the years 2000 and 2014 of cases reported to United States poison control centers revealed that although there are numerous different substance exposures, the ones associated with the highest morbidity and mortality have consistently been related to TCA overdose [14]. In 1992, TCA overdose accounted for 1.12 exposures per 10,000 population of the USA [15]. Acute intoxication with cardiotoxic drugs is associated with high mortality, which is around 12% worldwide, however, mortality associated with psychotropic drugs and non-cardiotoxic analgesics is around 4%. CAs were the number one cause of fatality from drug ingestion until the last decade, when they were surpassed by analgesics. Only 2–3% of (CAs) overdose cases that reach a healthcare facility result in death [16].

Advertisement

3. Drugs with cardiotoxic potential

The primary reasons for drug-induced cardiotoxicity and hypotension include systemic vasodilation leading to circulatory failure, myocardial depression & dysfunction, life-threatening arrhythmias. The acute toxic cardiac dysfunction is primarily associated with systolic dysfunction secondary to reduced myocardial contractility [17]. Drug-induced cardiotoxicity, commonly in the form of cardiac muscle dysfunction that may progress to cardiogenic shock, represents a major adverse effect of some common traditional anti-neoplastic agents, e.g., anthracyclines, cyclophosphamide, 5 fluorouracil, as well as newer agents such as biological monoclonal antibodies, e.g., trastuzumab, bevacizumab, and nivolumab; tyrosine kinase inhibitors, e.g., sunitinib and nilotinib; antiretroviral drugs, e.g., zidovudine; antidiabetics, e.g., rosiglitazone; as well as some illicit drugs such as alcohol, cocaine, methamphetamine, ecstasy, and synthetic cannabinoids. Most of the affected patients had no prior manifestation of the disease [18].

The cardiotoxic potential is not restricted to cardiovascular drugs only, the mortality remains higher in other drugs having membrane stabilizing activity (Tables 1 and 2) [19].

  1. Anti-arrhythmic agents (Vaughan Williams class I)

  2. Beta Blockers

  3. Dopamine and norepinephrine uptake inhibitors (bupropion)

  4. Anti-epileptics (Phenytoin/Carbamazepine)

  5. Antimalarial agents (Quinine/Chloroquine)

  6. Polycyclic antidepressants (Imipramine, Desipramine, etc.)

  7. Opioids (dextropropoxyphene)

  8. Recreational Agents such as Cocaine

Table 1.

Drugs with Membrane Stabilizing Activity (MSA).

  1. Calcium channel blockers

  2. Meprobamate

  3. Colchicine

  4. Cardiac glycosides (digoxin)

  5. H1 Antihistaminic

  6. Beta Blockers (not associated with membrane stabilizing activity)

Table 2.

Other drugs with cardiotoxic potential [18, 19].

Advertisement

4. Mechanism of toxicity and clinical manifestations

The most common cardiotoxicity clinical Case Series were reported with Tricyclic Antidepressant (TCA) toxicity and other Cardiovascular medication overdoses like Calcium channel blockers (CCBs) and beta blockers. The exact mechanism of drug-induced cardiotoxicity remains unclear, though it is likely to be multifactorial.

4.1 Tricyclic antidepressant (TCA) toxicity

Tricyclic antidepressants (TCAs) were one of the first classes of commonly prescribed antidepressants to be approved by the Food and Drug Administration (FDA). However, due to severe and potentially fatal side effects, their use in the U.S. has now been replaced by newer and safer alternatives.

Tricyclic antidepressants (TCAs) are still being used for the treatment of various conditions, including depression, migraine, headache prophylaxis, neuropathic pain, obsessive-compulsive disorder, and nocturnal enuresis. Because of their neuropsychiatric indications, they are accessible for suicidal attempts and overdose in patients with major depression [20].

Unfurtunatlly TCAs medication pharmacokinetic properities represent an ideal properities enhancing overdose toxicity; Long elimination half-life (25 to 81 hours) [21]. Large volume of distribution (10–20 L/kg) [22]. Highly bound to plasma protein (up to 95%), high lipid solubility, narrow therapeutic index (200–300 ng/mL), non-dialyzable and hemodialysis is ineffective. Major toxicity and deaths are associated with concentrations above 1000 ng/ml (Table 3) [18, 24].

Possible mechanismIts effect
Blockade of fast sodium channels in myocardial cells.
  1. Prolongation of phase “0” of the myocardial action potential.

  2. QRS prolongation.

  3. Bradycardia.

Blockade of potassium channel.
  1. QT prolongation.

  2. Torsade’s de pointes.

A quinidine-like toxic effect.
  1. Myocardial depression.

  2. Hypotension.

Blockade of peripheral alpha-adrenergic receptors.
  • Hypotension.

Table 3.

Mechanisms of TCA-induced cardiac toxicity [23].

TCA poisoning-related cardiovascular toxicity is evident in the presence of arrhythmia and hypotension in patients. Commonly, it causes both tachyarrhythmias and bradyarrhythmia, along with a prolongation of the QT interval seen on ECGs thus, possibly leading to Torsade de pointes [23]. TCA-associated wide complex tachyarrhythmias that originate from a ventricular origin are commonly ventricular tachycardia and ventricular fibrillation [25]. The QRS prolongation has been a vital role in determining and predicting the severity of TCA poisoning. This is why ECGs are pivotal during acute TCA poisoning and have become preferable over routine laboratory testing [26]. Bradyarrhythmia also typically occurs due to an atrioventricular block. However, the most common arrhythmia to occur due to TCA toxicity is sinus tachycardia and this is due to the anticholinergic properties of TCAs and the inhibition of NE [27]. Hypotension occurs due to a reduction in myocardial contraction and reduced systemic vascular resistance due to the alpha-adrenergic blockade [28]. This is why patients with TCA poisoning may develop acidemia due to a metabolic component from under perfused end organs and eventually respiratory acidosis due to respiratory depression, thus causing a mixed acidotic picture [29, 30]. Overall decrease in myocardial function will inevitably lead to worsening tissue hypoperfusion and hyperlacticaemia (Figures 14).

Figure 1.

ECG with Wide complex tachycardia, concerning for polymorphic ventricular tachycardia.

Figure 2.

12-lead electrocardiogram in the emergency department showing widened QRS (duration ˜170 ms), prominent R waves in aVR, and frequent ventricular premature complexes.

Figure 3.

CA toxicity, characteristics, and monitoring (created by Mohamed Nassef 09dec 2023).

Figure 4.

Approach for selecting severely poisoned patient for VA ECMO [31].

4.2 Cardiovascular medication overdose

Cardiovascular medication overdose constitutes about 3.5% of all drug overdoses, and it carries a mortality of about 16% [32]. Calcium channel blockers (CCBs) are among the most used cardiovascular drugs in the adult population. Like most other medications, when calcium channel blockers are taken beyond the appropriate recommended dosage, they can have untoward toxicities with a wide range of complications that can even be fatal. Calcium channel blockers (CCB) along with β-blockers and angiotensin receptor blockers constitute more than 65% of deaths from cardiovascular medications. Overdose can cause serious mortality and morbidity. Half-life of amlodipine and telmisartan are 34 to 50 hours and 24 hours, respectively [33]. The overdose of these drugs can cause profound refractory hypotension, bradyarrhythmia, and shock. The combination of these drugs may blunt the sympathetic and vasoconstrictive responses and worsen CCB toxicity [34]. Toxicity can be delayed up to 16 hours after the ingestion of sustained-release formulations. Initial symptoms may be as nonspecific as dizziness, fatigue, and lightheadedness, and in severe toxicities, it may rapidly decline to alter mental status, coma, hypotension, and bradycardia.

Hypotension and bradycardia, when progressive, can eventually lead to cardiogenic shock. Also, hyperglycemia is common with all subclasses of CCBs and can be a useful clinical marker for poisoning severity. Both effects lead directly to metabolic acidosis [35, 36, 37]. It is also common to develop mild hypokalemia and mild to severe hypocalcemia [38]. All CCBs are very well absorbed orally across the subtypes, undergo extensive hepatic first-pass metabolism, are lipophilic, bind readily to plasma proteins, and have a large volume of distribution (> 2 liters/kg). Elimination by hemodialysis or hemofiltration is ineffective because they are highly protein-bound with a large volume of distribution [39, 40]. The most common ECG abnormalities involving calcium channel blockers other than dihydropyridines are sinus bradycardia, variable degrees of atrioventricular blocks, bundle branch block, QT prolongation, and junctional rhythms. Dihydropyridines maintain normal sinus rhythm and can cause reflex sinus tachycardia. Echocardiography may show good left ventricular function, normal functioning valves, and an ejection fraction of 40–60%.

Because of their similarity of end-organ affinity, pharmacological action, and target patients, beta blockers are the most common confounders. Compared to CCBs, beta blockers are less likely to cause hyperglycemia in adult patients [41]. However, an initial presentation with bradycardia and hypotension should prompt consideration of a wide array of pharmacological agents, including:

  • Beta blocker toxicity

  • Tricyclic antidepressant toxicity

  • Digoxin toxicity

  • Clonidine overdose

  • Sedative-hypnotic toxicity

  • Opiate overdose

  • Organophosphate poisoning

Advertisement

5. Evidence behind the role of VA ECMO in drug intoxication

The available literature is restricted to case reports or case series with few patients only. However, these results are promising and encouraging, the use of VA ECMO in refractory shock due to drug intoxication. The experiences in human subject include an observational cohort which shows better outcome in six patients with severe intoxication with profound shock who received VA ECMO as compared to use of ECMO for other indications [42]. VA ECMO is a lifesaving therapeutic option, considered a bridge for recovery in TCA and Venlafaxine-induced cardiac toxicity. Since 1993, Goodwin DA and colleagues reported a case of near-fatal TCA overdose that failed to respond to standard therapy but was resuscitated using extracorporeal circulation [43]. Weinberg et al. described the successful use of VA ECMO in two patients with refractory vasodilatory shock associated with amlodipine poisoning [44]. C. Vignesh and colleagues reported a clinical Case Series using veno-arterial extracorporeal membrane oxygenation (VA ECMO) circulatory support as a salvage therapy in patients resistant to maximal medical management for drug overdose [45]. The cornerstone of medical management for drug overdoses consists of limiting their absorption and hemodynamic support until they are metabolized and eliminated from the body. Gastric decontamination with activated charcoal to limit the absorption can be attempted when done early [46]. Volume resuscitation with fluids to overcome the relative hypovolemia associated with vasodilation and inotropic/vasopressor use are the mainstay. Treatment with sodium bicarbonate remains the standard of care for TCA poisoning [47]. In CCB overdose, calcium infusion can be initiated to overcome the calcium channel antagonism. The increased transcellular calcium gradient drives calcium intracellularly and antagonizes the effect of CCBs [48].

5.1 Role of VA ECMO in drug intoxication

The reversibility in cardiotoxicity, especially in the case of refractory shock not responding to conventional measures, may raise questions regarding the role of VA ECMO as a lifesaving procedure. VA ECMO may be useful in providing adequate cardiac output and maintaining tissue perfusion, which helps in redistribution of the toxic substances and their metabolites from central circulation and facilitates the metabolism and excretion of drugs by improving hepatic and renal blood flow [49].

5.1.1 Bridge to recovery

Toxic substances having potential for cardiotoxicity, where antidote is not available, may be fatal even with conventional support. VA ECMO can support the cardiac function in poisoned patients with severe cardiotoxicity, who are having severe left or right ventricular dysfunction, persistent life-threatening arrhythmias, or even cardiac arrest unresponsive to conventional management. Cardiovascular function starts recovering once the toxic substance is either metabolized or excreted from the body. The duration of VA ECMO support depends on several factors such as severity of toxicity and recovery cardiac dysfunction, half-life of toxin, and organ dysfunction at the time of initiation of VA ECMO, etc. [50, 51] Veno-arterial (VA) ECMO reduced cardiac oxygen consumption and provided both hemodynamic and respiratory support as a bridge to recovery [52]. Even in cases of multiple drug intoxication or unknown poisoning with cardiogenic shock, ECMO support can be beneficial (Table 4) [55, 56].

Published case reportsIngested agentThe duration of ECMO (hours)
Goodwin et al. 1993Desipramine60
Williams et al. 1994Imipramine7
Kobayashi et al. 2011Nortriptyline14
Kejiri et al. 2021Amitriptyline27

Table 4.

Duration of mechanical support as a bridge to recovery [43, 44, 45, 53, 54].

5.1.2 Bridge to antidote

VA ECMO can be helpful in life-threatening arrhythmia or cardiovascular collapse with those toxins that can be managed successfully with antidote, but which is not available readily due to short shelf life and cost. Digoxin-specific antibodies fragments (Fab) rapidly improve the digitalis-induced arrhythmias and cardiac toxicity. However, Digoxin poisoning is uncommon and Fab fragments are expensive with limited shelf life. Patients can be supported with VA ECMO until Fab is administered [57, 58].

5.1.3 Bridge to toxin elimination with renal replacement therapy

VA ECMO may be considered to maintain hemodynamics. The RRT may be added to ECMO circuit or may be started separately to eliminate the toxin [59]. The various techniques used for toxin removal include dialysis, hemoperfusion, and hemofiltration. The basic principle of dialysis involves diffusion through a semipermeable membrane, whereas in hemoperfusion, the toxin is adsorbed on the adsorbent surface of the dialyzer. The mechanism of hemofiltration is convection across the membrane. These therapies may be used as a single or in combination. The decision about dialyzing the toxic substance depends on the molecular weight, protein binding, and volume of distribution of the toxin. Large molecular weight medications are poorly dialyzed. Toxic substances that are highly protein-bound are less available for removal through renal replacement therapy (RRT). If the toxic substance has a large volume of distribution, elimination of toxic substance will be prolonged because RRT will remove toxic substance from plasma space only [60].

5.1.4 Bridge to transplant

VA ECMO can be useful as a bridge to a permanent assist device and heart transplant in patients with persistent irreversible cardiac failure.

Advertisement

6. Approach for selecting severely poisoned patients for VA ECMO

Advertisement

7. Hemodynamic aspects of VA ECMO support

VA ECMO has the highest capability to reduce myocardial pressure-volume area (sum of myocardial potential energy and myocardial stroke work) in patients with cardiogenic shock CS reducing left ventricular end-diastolic volume (LVEDV) and left ventricular end-diastolic pressure (LVEDP) while providing complete hemodynamic and respiratory support. The myocardial pressure-volume area is further reduced by the weaning of inotropes and vasopressors. All these prevent the vicious cycle of maladaptive neurohormonal and vascular mechanisms. Also, native RV function is not critical to provide systemic perfusion due to its reduced reliance on transpulmonary flow. VA ECMO improves systemic perfusion by increasing the main arterial blood pressure MAP, reducing central venous pressure CVP, and increasing the systemic arteriovenous pressure gradient. This maintains the organ function and reduces the generation and accumulation of toxic metabolites. This may be particularly relevant to improving blood flow in organs with portal circulation, such as the liver and kidney. Fluid removal and reducing venous congestion can be further enhanced by splicing a continuous renal replacement therapy using veno-venous hemodialysis mode (CVVHD) into the VA ECMO circuit [61, 62, 63, 64, 65].

Advertisement

8. Conclusion

VA ECMO has been recognized as a pivotal advancement in the treatment of severe drug overdoses, particularly those leading to cardiovascular shock and complications. It transcends the capabilities of traditional therapies by offering critical, lifesaving support in instances of severe cardiotoxicity and refractory shock. Its primary functions include the stabilization of cardiovascular dysfunction, management of shock, and support during acute renal failure, often seen in overdoses involving cardiotoxic drugs like TCAs and CCBs. The importance of VA ECMO lies in its ability to ensure continuous tissue perfusion and oxygenation, thereby facilitating the detoxification process and aiding in the elimination of toxic substances from the body. This makes it an essential bridge to recovery or transplant, especially in cases where the patient’s condition does not improve with conventional treatment methods. Furthermore, VA ECMO plays a vital role in reducing cardiac stress and maintaining systemic circulation, which is crucial in managing the high mortality and morbidity associated with drug overdoses.

Overall, the increasing reliance on VA ECMO in the realm of critical care highlights its effectiveness in handling complex, life-threatening scenarios caused by drug intoxication. This underscores its indispensable role in modern medical practice, marking it as a cornerstone in the management of drug overdose-induced complications.

Advertisement

Acknowledgments

We extend our deep thanks to the Emirates Health Services (EHS) and its leadership for the unlimited support for the continuation of the ECMO program.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

Notes/thanks/other declarations

We would like to thank Dr. Javed khan, head of the ECMO program at Al Qassimi Hospital, and all the program members.

Thanks to Dr. Sumaya Abdulatif CMO of Al Qassimi Hospital.

References

  1. 1. Riou B, Barriot P, Rimailho A, Baud FJ. Treatment of severe chloroquine poisoning. The New England Journal of Medicine. 1988;318(1-6):17
  2. 2. Clemessy JL, Taboulet P, Hoffman JR, Hantson P, Barriot P, Bismuth C, et al. Treatment of acute chloroquine poisoning: A 5-year experience. Critical Care Medicine. 1996;24:1189-1195
  3. 3. Manini AF, Nelson LS, Stimmel B, et al. Incidence of adverse cardiovascular events in adults following drug overdose. Academic Emergency Medicine. 2012;19(7):843-849
  4. 4. Huikuri HV, Castellanos A, Myerburg RJ. Sudden death due to cardiac arrhythmias. The New England Journal of Medicine. 2001;345:1473-1482
  5. 5. Chapter 7 Drug Induced Cardiotoxicity: Mechanism, Prevention and Management. Available from: https://www.intechopen.com/chapters/62447 [Accessed: December 4, 2023]
  6. 6. Simone KE. Thirty US poison centers report later: Greater demand, more difficult problems. Clinical Toxicology (Philadelphia, Pa.). 2014;52(2):91-92. DOI: 10.3109/15563650.2013.878949
  7. 7. Rehder KJ, Turner DA, Cheifetz IM. Extracorporeal membrane oxygenation for neonatal and pediatric respiratory failure: An evidence-based review of the past decade (2002-2012). Pediatric Critical Care Medicine. 2013;14(9):851-861. DOI: 10.1097/PCC.0b013e3182a5540d
  8. 8. Zampieri FB, Mendes PV, Ranzani OT, et al. Extracorporeal membrane oxygenation for severe respiratory failure in adult patients: A systematic review and meta-analysis of current evidence. Journal of Critical Care. 2013;28(6):998-1005. DOI: 10.1016/j.jcrc.2013.07.047
  9. 9. Panzeri C, Bacis G, Ferri F, et al. Extracorporeal life support in severe Taxus baccata poisoning. Clinical Toxicology (Philadelphia, Pa.). 2010;48(5):463-465. DOI: 10.3109/15563650.2010.487487
  10. 10. Freedman MD, Gal J, Freed CR. Extracorporeal pump assistance – Novel treatment for acute lidocaine poisoning. European Journal of Clinical Pharmacology. 1982;22:129-135
  11. 11. Martin TG, Klain MM, Molner RL, et al. Extracorporeal life support vs thumper after lethal desipramine OD (abstract). Veterinary and Human Toxicology. 1990;32:349
  12. 12. Larkin GL, Graeber GM, Hollingsed MJ. Experimental amitriptyline poisoning: Treatment of severe cardiovascular toxicity with cardiopulmonary bypass. Annals of Emergency Medicine. 1994;23:480-486
  13. 13. Gummin DD, Mowry JB, Spyker DA, Brooks DE, Fraser MO, Banner W. 2016 annual report of the American Association of Poison Control Centers' National Poison Data System (NPDS): 34th annual report. Clinical Toxicology (Philadelphia, Pa.). 2017;55(10):1072-1252. DOI: 10.1080/15563650.2017.1388087. Epub 2017 Nov 29. Erratum in: Clin Toxicol (Phila). 2017;55(10):1256
  14. 14. Nelson JC, Spyker DA. Morbidity and mortality associated with medications used in the treatment of depression: An analysis of cases reported to U.S. poison control centers, 2000-2014. The American Journal of Psychiatry. 2017;174(5):438-450
  15. 15. Khalid MM, Waseem M. Toxicity, Tricyclic Antidepressant. Treasure Island (FL): StatPearls Publishing; 2017, [Updated: March 16, 2017]. Available from: https://www.ncbi.nlm.nih.gov/books/NBK430931 [Accessed: December 9, 2023]
  16. 16. Beneke LLS, Vearrier D. Tricyclic Antidepressant Toxicity. Medscape. Updated: June 04, 2024. Available from: https://emedicine.medscape.com/article/819204-overview#a6 [Accessed: December 3, 2023]
  17. 17. Drug Induced Cardiotoxicity: Mechanism, Prevention and Management. Available from: https://www.researchgate.net/publication/328948622_Drug_Induced_Cardiotoxicity_Mechanism_Prevention_and_Management#fullTextFileContent [Accessed: December 4, 2023]
  18. 18. Henry JA, Cassidy SL. Membrane stabilizing activity: A major cause of fatal poisoning. Lancet. 1986;1:1414-1417
  19. 19. Nadeem UK, Ricardo PN, Nudrat S, et al. Intentional and unintentional poisoning in Pakistan: A pilot study using the emergency departments surveillance project. BMC Emergency Medicine. 2015;15(Suppl. 2):S2
  20. 20. Jones AL, Dargan PI. Churchill’s Pocketbook of Toxicology. London: Churchill Livingstone; 2001
  21. 21. Kolecki PF, Curry SC. Poisoning by sodium channel blocking agents. Critical Care Clinics. 1997;13:829-848
  22. 22. Dawson AH. The tricyclic antidepressants. In: Dart RC, editor. Medical Toxicology. 3rd ed. Vol. 1. Baltimore: Lippincott Williams & Wilkins; 2004. pp. 851-861
  23. 23. Giwa A, Oey E. The return of an old nemesis: Survival after severe tricyclic antidepressant toxicity, a case report. Toxicology Reports. 10 Mar 2018;5:357-362. DOI: 10.1016/j.toxrep.2018.03.009. [Accessed: December 4, 2023]
  24. 24. Tricyclic Antidepressants (TCAs), Mayo Clinic. 2016. Available from: https://www.mayoclinic.org/diseases-conditions/depression/in-depth/antidepressants/art-20046983 [Accessed: December 29, 2017]
  25. 25. Spiker D, Biggs J, Tricyclic antidepressants. Prolonged plasma levels after overdose. JAMA. 1976;236:1711-1712
  26. 26. James LP, Kearns GL. Cyclic antidepressant toxicity in children and adolescents. Journal of Clinical Pharmacology. 1995;35:343-350
  27. 27. Zapalska-Pozarowska K, Szponar J, Tchorz M, Kostek H, Pozarowski W. Severe tricyclic antidepressants poisoning? A case study. Przeglaṃd Lekarski. 2012;69(8):621-623
  28. 28. Goldberg RJ, Capone RJ, Hunt JD. Cardiac complications following tricyclic antidepressant overdose. Issues for monitoring policy. JAMA. 1985;254(13):1772-1775
  29. 29. Boehnert MT, Lovejoy FH Jr. Value of the QRS duration versus the serum drug level in predicting seizures and ventricular arrhythmias after an acute overdose of tricyclic antidepressants. The New England Journal of Medicine. 1985;313(8):474-479
  30. 30. Kerr GW, McGuffie AC, Wilkie S. Tricyclic antidepressant overdose: A review. Emergency Medicine Journal. 2001;18(4):236-241
  31. 31. Gupta V, Gupta R, Wander GS. Role of ECMO in life threatening intoxication/the Egyptian. Journal of Critical Care Medicine. 2018;6:103-109
  32. 32. Lai MW, Klein-Schwartz W, Rodgers GC, Abrams JY, Haber DA, Bronstein AC, et al. 2005 annual report of the American Association of Poison Control Centers’ national poisoning and exposure database. Clinical Toxicology (Philadelphia, Pa.). 2006;44:803-932
  33. 33. Bronstein AC, Spyker DA, Cantilena LR Jr, Rumack BH, Dart RC. 2011 annual report of the American Association of Poison Control Centers’ National Poison Data System (NPDS): 29th annual report. Clinical Toxicology (Philadelphia, Pa.). 2012;50(10):911-1164
  34. 34. Abernethy DR. The pharmacokinetic profile of amlodipine. American Heart Journal. 1989;118:1100-1103
  35. 35. Hofer CA, Smith JK, Tenholder MF. Verapamil intoxication: A literature review of overdoses and discussion of therapeutic options. The American Journal of Medicine. 1993;95(4):431-438
  36. 36. Kuo MJ, Tseng YZ, Chen TF, Fong DE. Verapamil overdose and severe hypocalcemia. Journal of Toxicology. Clinical Toxicology. 1992;30(2):309-311
  37. 37. Reith DM, Dawson AH, Epid D, Whyte IM, Buckley NA, Sayer GP. Relative toxicity of beta blockers in overdose. Journal of Toxicology. Clinical Toxicology. 1996;34(3):273-278
  38. 38. Howarth DM, Dawson AH, Smith AJ, Buckley N, Whyte IM. Calcium channel blocking drug overdose: An Australian series. Human & Experimental Toxicology. 1994;13(3):161-166
  39. 39. Smith SW, Ferguson KL, Hoffman RS, Nelson LS, Greller HA. Prolonged severe hypotension following combined amlodipine and valsartan ingestion. Clinical Toxicology (Philadelphia, Pa.). 2008;46(5):470-474
  40. 40. Dewitt CR, Waksman JC. Pharmacology, pathophysiology and management of calcium channel blocker and beta-blocker toxicity. Toxicological Reviews. 2004;23:223-238
  41. 41. Levine M, Brooks DE, Franken A, Graham R. Delayed-onset seizure and cardiac arrest after amitriptyline overdose, treated with intravenous lipid emulsion therapy. Pediatrics. 2012;130(2):e432-e438
  42. 42. Vanzetto G, Akret C, Bach V, et al. Percutaneous extracorporeal life support in acute severe hemodynamic collapses: Single centre experience in 100 consecutive patients. The Canadian Journal of Cardiology. 2009;25:e179-e186
  43. 43. Goodwin DA, Lally KP, Null DM Jr. Extracorporeal membrane oxygenation support for cardiac dysfunction from tricyclic antidepressant overdose. Critical Care Medicine. 1993;21:625-627
  44. 44. Weinberg RL, Bouchard NC, Abrams DC, Bacchetta M, Dzierba AL, Burkart KM, et al. Venoarterial extracorporeal membrane oxygenation for the management of massive amlodipine overdose. Perfusion. 2014;29:53-56
  45. 45. Vignesh C, Kumar M, Venkataraman R, Rajagopal S, Ramakrishnan N, Abraham BK, et al. Extracorporeal membrane oxygenation in drug overdose. Indian Journal of Critical Care Medicine. 2018;22(2):111-115. DOI: 10.4103/ijccm.IJCCM_417_17A Clinical Case Series
  46. 46. Kenny J. Treating overdose with calcium channel blockers. BMJ. 16 Apr 1994;308:992-993
  47. 47. Salhanick SD. Tricyclic antidepressant poisoning. In: Post TW, editor. UpToDate. Waltham, MA: UpToDate. [Accessed: November 25, 2013]
  48. 48. Rizvi I, Ahmad A, Gupta A, Zaman S. Life-threatening calcium channel blocker overdose and its management. BML Case Reports. 2012;2012:bcr0120125643
  49. 49. Albertson TE, Dawson A, De Latorre F, Hoffman RS, Hollander JE, Jaeger A, et al. TOX-ACLS: Toxicologic-oriented advanced cardiac life support. Annals of Emergency Medicine. 2001;37:S78-S90
  50. 50. Jae-Hwan L, Hyun-Sook K, Jae-Hyeong P, et al. Incidence and clinical course of left ventricular systolic dysfunction in patients with carbon monoxide poisoning. Korean Circulation Journal. 2016;46(5):665-671
  51. 51. Hossein HM, Nasim Z, Mitra R, et al. Successful treatment of aluminium phosphide poisoning by extracorporeal membrane oxygenation. Basic & Clinical Pharmacology & Toxicology. 2016;118:243-246
  52. 52. Chung M, Shiloh AL, Carlese A. Monitoring of the adult patient on venoarterialextracorporeal membrane oxygenation. Scientific World Journal. 2014;39:325-328
  53. 53. Kobayashi K, Miyajima M, Tanaka T, et al. Tricyclic antidepressant overdose rescued by percutanenous cardiopulmonary support: Report of two cases [Japanese]. Chūdoku Kenkyū. 2011;24:46-50
  54. 54. Ikejiri K, Akama Y, Ieki Y, Kawamoto E, Suzuki K, Yokoyama K, et al. Veno-arterial extracorporeal membrane oxygenation and targeted temperature management in tricyclic antidepressant-induced cardiac arrest. A case report and literature review. Medicine (Baltimore). 2021;100(9):e24980
  55. 55. Mohamed YA, Akram AA, Mohamed MK. Veno-venous extracorporeal membrane oxygenation in a case of organophosphorus poisoning. Egyptian Journal of Critical Care Medicine. 2016;4:43-46
  56. 56. Roberto R, Barbara C, Roberto M, et al. Extra-corporeal life support for near fatal multi-drug intoxication: A case report. Journal of Medical Case Reports. 2011;5:231
  57. 57. Idialisoa R, Jouffroy R, Lamhaut L, et al. Extra corporeal life support in life threatening digoxin overdose: A bridge to antidote Austin. Emergency Medicine. 2016;2(5):1029
  58. 58. Chan BS, Buckley NA. Digoxin-specific antibody fragments in the treatment of digoxin toxicity. Clinical Toxicology (Philadelphia, Pa.). 2014;52:824-836
  59. 59. Daubin C, Lehoux P, Ivascau C, Tasle M, Bousta M, Lepage O, et al. Extracorporeal life support in severe drug intoxication: A retrospective cohort study of seventeen cases. Critical Care. 2009;13:187-193
  60. 60. Holubek WJ, Hoffman RS, Goldfarb DS, Nelson LS. Use of hemodialysis and hemoperfusion in poisoned patients. Kidney International. 2008;74:1327-1334
  61. 61. Eddleston M. Pattern and problems of deliberate self poisoning in the developing world. The Quarterly Journal of Medicine. 2000;93:715-731
  62. 62. Zihni S, Edvin P, Aurel D, et al. Early clinical outcome of acute poisoning cases treated in intensive care unit. Medicinski Arhiv. 2015;69(6):400-404
  63. 63. Müller D, Herbert D. Common causes of poisoning: Etiology, diagnosis and treatment. Deutsches Ärzteblatt International. 2013;110(41):690-700
  64. 64. Vilma SS, Maria CPM, Flavia FN. Occupational pesticide poisoning, 2000-2009, Brazil. Revista de Saúde Pública. 2013;47(3):1-8
  65. 65. Mokhlesi B, Leikin JB, Murray P, et al. Adult toxicology in critical care: Part II: Specific poisonings. Chest. 2003;123:897-922

Written By

Mohamed Nassef and Nashat Abdulhalim

Submitted: 29 December 2023 Reviewed: 04 June 2024 Published: 27 June 2024