Cardiol Res
Cardiology Research, ISSN 1923-2829 print, 1923-2837 online, Open Access
Article copyright, the authors; Journal compilation copyright, Cardiol Res and Elmer Press Inc
Journal website http://www.cardiologyres.org

Case Report

Volume 9, Number 1, February 2018, pages 46-49


Critical Management of Severe Hypotension Caused by Amlodipine Toxicity Managed With Hyperinsulinemia/Euglycemia Therapy Supplemented With Calcium Gluconate, Intravenous Glucagon and Other Vasopressor Support: Review of Literature

Kishore Kumara, c, Madhavi Biyyama, Bharat Bajantrib, Sureshkumar Nayudua

aDivision of Gastroenterology, Department of Medicine, Bronx Lebanon Hospital Center, Bronx, NY, USA
bDivision of Pulmonary and Critical Care, Department of Medicine, Bronx Lebanon Hospital Center, Bronx, NY, USA
cCorresponding Author: Kishore Kumar, Division of Gastroenterology, Department of Medicine, Bronx Lebanon Hospital Center, Bronx, NY, USA

Manuscript submitted December 3, 2017, accepted December 12, 2017
Short title: Hypotension Caused by Amlodipine Toxicity
doi: https://doi.org/10.14740/cr646w

Abstract▴Top 

Calcium channel blocker (CCB ) overdose, whether intentional or accidental, is a common clinical scenario and can be very lethal. Conventional treatments for CCB overdose include intravenous (IV) fluids, calcium salts, dopamine, dobutamine, norepinephrine, phosphodiesterase inhibitors, and glucagon. However, the conventional therapies are unsuccessful in reversing the cardiovascular toxicity of CCB, so they commonly fail to improve the hemodynamic condition of the patient. Blockade of the L-type calcium channels that mediate the antihypertensive effect of CCBs also decreases the release of insulin from pancreatic β-islet cells and reduces glucose uptake by tissues (insulin resistance). By targeting this insulin-mediated pathway, hyperinsulinemia/euglycemia therapy (HIET) appears to have a distinct role, and its clinical potential is underrecognized in the management of severe CCB toxicity. We present a case of young man with amlodipine toxicity successfully managed with high dose of IV insulin therapy.

Keywords: Calcium channel blocker toxicity; Hyperinsulinemia/euglycemia therapy; Shock

Introduction▴Top 

Calcium channel blocker (CCB) overdose, whether intentional or accidental, is a common clinical scenario and can be very lethal. Conventional treatments for CCB overdose include intravenous (IV) fluids, calcium salts, dopamine, dobutamine, norepinephrine, phosphodiesterase inhibitors, and glucagon. Most of these therapies are intended to increase transmembrane calcium flow (calcium salts) or increase cyclic adenosine monophosphate (cAMP) concentration by stimulating production of adenylate cyclase (with norepinephrine and glucagon) or by inhibiting production of phosphodiesterase (with amrinone and milrinone). However, the conventional therapies are unsuccessful in reversing the cardiovascular toxicity of CCB, so they commonly fail to improve the hemodynamic condition of the patient. Blockade of the L-type calcium channels that mediate the antihypertensive effect of CCBs also decreases the release of insulin from pancreatic β-islet cells and reduces glucose uptake by tissues (insulin resistance). By targeting this insulin-mediated pathway, hyperinsulinemia/euglycemia therapy (HIET) appears to have a distinct role, and its clinical potential is underrecognized in the management of severe CCB toxicity. There is growing experimental and clinical evidence of the value and the safety of HIET in the management of CCB poisoning. Although the mechanism of this beneficial action is not fully explained, HIET should be considered in patients with CBB-induced cardiovascular compromise. Additional clinical research and prospective clinical studies are needed to confirm the safety and efficacy of HIET and to support more formal guidelines and therapeutic regimens, but some rational recommendations can be made based on the available data. Author suggested careful monitoring of blood glucose, serum potassium concentrations and electrocardiogram is required.

Case Report▴Top 

An 18-year-old obese man was brought to our emergency department after inadvertent intake of anti-hypertensive medications under the influence of alcohol which were originally prescribed to his mother. According to patient, he took pills around 12:30 am after returning home and remembered waking up at 4:00 am with episodes of non-bilious, non-bloody vomiting. Patient reports being drunk when he ingested multiple pills of amlodipine 5 mg, metformin 500 mg and combination pill of lisinopril 20 mg/hydrochlorothiazide 25 mg which were half bottle full as per family. However, he continues to deny suicidal attempt or intentional intake of prescribed medication. He has no history of suicidal attempt, mood disorder or any other past psychiatric illness. He has no medical problem and does not take any medication at home. He admits smoking marijuana and tobacco and drinks alcohol regularly. On presentation, his temperature was 98.7 °F, blood pressure was 68/50 mm Hg, pulse was 82 beats per minute, respiratory rate was 14 cycles per minute and BMI was 41.1 kg/m2. Chest exam showed bilateral air entry without any adventitious sounds. The cardiovascular exam showed normal heart sounds without murmurs, gallops, or rubs. The abdomen was soft, with no visceromegaly and with normal bowel sounds. Extremities were without edema, cyanosis, or clubbing. Electrocardiogram showed sinus tachycardia with no evidence of any conduction delays. In the emergency room, he received intravenous bolus of normal saline with transient improvement in his blood pressure; however, his blood pressure remained low requiring vasopressor support. Initially he was treated with activated charcoal, aggressive intravenous hydration and calcium infusion. Poison control was consulted and suggested HIET. He was started on high dose insulin of 250 units/h that was increased to 450 units/h, dose was up titrated based on blood pressure response along with up titrating doses of intravenous dextrose to avoid hypoglycemia. Blood sugar levels were checked every 30 min during the time of insulin/dextrose administration. Although most of the time while receiving intravenous insulin/dextrose therapy his blood sugar levels were at higher sides, he developed total of three episodes of asymptomatic hypoglycemia range from 47 to 61 mg/dL well responded to increasing dose of dextrose. Beside serum glucose, serum lactic acid, serum creatinine, serum potassium, serum calcium, PH and intake/output were closely monitored. Echocardiogram showed normal wall motion, contractility and valvular functions. He received insulin therapy for about 42 h and intravenous glucagon therapy at rate of 10 mg/h for about 30 h. During the period of refractory hypotension, he received vasopressor support with phenylephrine and norepinephrine. He required total of 3 days monitoring in critical care unit without airway compromise or need of positive pressure ventilation. Subsequently patient was transferred to inpatient psychiatry unit to evaluate for undiagnosed underlying psychiatric; however, he remained asymptomatic and subsequently discharged home with outpatient follow-up for the management of substance dependency.

Discussion▴Top 

CCBs are a heterogeneous group of chemicals that inhibit L-type calcium channels. These channels control myocardial and vascular smooth muscle contractility as well as the cardiac conduction system and pacemaker cells. CCBs are used extensively to treat angina pectoris, hypertension, Raynaud’s syndrome, supraventricular tachycardia and migraine headaches. CCBs are divided based on their primary physiologic effects. Dihydropyridine CCBs (DHPs) such as amlodipine and nifedipine, preferentially block calcium channels in the vasculature, acting as vasodilators and in toxic doses leading to myocardial depression, impairing cardiac conduction and QRS prolongation [1-3]. All other CCBs are grouped together as non-dihydropyridine (non-DHP) CCB. The most common are verapamil and diltiazem. These act on cardiomyocytes to reduce vascular permeability and affect cardiac contractility and conduction, though they also have some mild vasodilatory action [4].

Conventional therapy for CCB poisoning included the administration of fluids, calcium salts, glucagon, and vasopressor [5, 6]. Intravenous calcium has been a frequently used agent for calcium channel overdose. The goal is to competitively overcome the antagonism of the CCB. Calcium may be given as either calcium gluconate or calcium chloride. While calcium chloride contains three times more calcium for the same volume, calcium gluconate is less irritating to the veins and is preferred in most instances. Ionized calcium levels should be monitored, with the goal being two times the normal [1]. Glucagon is another frequently recommended antidote for CCB overdose. Glucagon stimulates adenyl cyclase via G proteins, resulting in increased intracellular cyclic AMP, which in turn leads to stimulation of muscle contraction. The clinical effect of glucagon resides in its positive inotropic and chronotropic effects as confirmed in multiple animal studies, but not in human clinical trials [2]. The main side effects of this therapy are nausea, vomiting, hyperglycemia, and ileus. The significant incidence of vomiting mandates ensuring a protected airway prior to glucagon administration [1]. In severe symptomatic bradycardia not responding to atropine or isoprenaline infusion other modalities of treatment described in literature are transvenous pacing [7], plasma exchange [8] and extracorporeal membrane oxygenation [9]. Extracorporeal elimination by conventional hemofiltration and dialysis is not recommended because these agents bind to plasma proteins and have a large volume of distribution. In our case, the patient presented with severely decreased blood pressure, which was reversed after administration of the HIET along with conventional therapy and tapering dose of vasopressors (norepinephrine and phenylephrine). HIET is considered to be the most advanced therapy first reported in humans in 1999, that showed improvement in circulatory shock [10, 11] interrupting the vicious cycle that is responsible for progressive hemodynamic deterioration and, ultimately, patient death. The efficacy and safety of this treatment have been demonstrated in several cases of CCB poisoning [6, 12, 13].

CCB overdose is frequently complicated by metabolic acidosis secondary to renal failure from prolonged hypoperfusion and end-organ ischemia [4], though in our patient renal failure and lactic acidosis were partly contributed by overdose of metformin and lisinopril. Another pathophysiologic mechanism for acidosis in patients with calcium channel toxicity decreases insulin production due to the blockage of the L-type calcium channels in the pancreas leading to impairment of the myocardial energy supply. In normal circumstances, myocardial cells use the oxidation of free fatty acids as an energy substrate for aerobic metabolism. However, in a shock state, the myocardium switches to glucose use which requires the presence of insulin in order to enter in the cell. Therefore, CCB toxicity causes decrease insulin release from pancreas hence during a state of hypoinsulinemia and acquired insulin resistance, myocardial cells are unable to use glucose as an energy source, leading to decreased myocardial contractility and hypotension. The lack of energy substrate exacerbates cardiovascular depression, which is already compromised by the blocking of the calcium channels. Non-cardiogenic pulmonary edema is also one of the complications secondary to CCB overdose [14-16]. HIET may lead to reversal of cardiovascular collapse in CCB toxicity by improving myocardial utilization of carbohydrates, increasing plasma levels of ionized calcium, improving hyperglycemic acidotic state, as well as by clearing the cytosol of lactic acid and other glycolytic byproducts [2, 17, 18]. In addition, insulin has a direct positive inotropic activity that may contribute to its clinical effects [2, 17-19]. Boyer and colleagues proposed a protocol for the initiation of HIET wherein an initial insulin bolus of as much as 1 IU/kg is given, followed by insulin infusions of 0.5 IU/kg/h and 10% dextrose in half-normal saline. Blood glucose must be checked at least once every 30 min and hypertonic glucose must be infused to maintain blood glucose in the upper normal range. Up to 20 - 30 g/h may be needed in adults. Supplemental potassium is required only to prevent severe hypokalemia [20]. The duration of HIET should be guided by the clinical response, especially hemodynamic parameters: the goal should be hemodynamic stability after the withdrawal of vasoactive agents [20, 21]. Methylene blue has been used as a novel approach to treat refractory CCB overdose based on reports of its use to counteract post-coronary artery bypass vasoplegia when added to vasopressors and HIET. Methylene blue resolves vasoplegia by decreasing intracellular cyclic guanosine monophosphate (cGMP), scavenging nitric oxide, and inhibiting nitric oxide synthesis, all in direct opposition to the action of CCBs [22, 23]. There is no definitive evidence that either gastrointestinal decontamination in the form of activated charcoal or whole bowel irrigation alters the clinical outcome in the CCB overdose. However, gastrointestinal (GI) decontamination is still advocated because of the potential lethal nature of this overdose and lack of a specific efficacious antidote. But potential risks of GI decontamination should be kept in mind, e.g. gastric lavage should probably be withheld in patients who are already bradycardic or have conduction disturbances [15]. There are case reports that have suggested the importance of whole bowel irrigation in patients who ingested sustained release preparations of calcium channel blockers [3, 24] (Table 1).

Table 1.
Click to view
Table 1. Various Treatment Methods Used in CCB Overdose
 

Conclusion

CCB overdose, whether intentional or accidental, can be lethal. There is growing experimental and clinical evidence of the value and the safety of HIET in the management of CCB poisoning. Although the mechanism of this beneficial action is not fully explained, HIET should be considered in patients with CBB-induced cardiovascular compromise. Although not effective in all cases, HIET often improves arterial blood pressure, myocardial contractility and metabolic acidosis. Whatever the main hemodynamic effect involved, observations support the value of HIET in patients with CCB intoxication and circulatory compromise and suggest that this therapy should probably be considered earlier in the management of the condition rather than being used as a rescue option. Indeed, some cases in which HIET was introduced late in the treatment and failed to improve the patient’s condition have also been reported. The effectiveness of HIET is often limited to an improvement of hypotension and acidosis that is observed within 30 - 45 min of starting insulin administration. Direct actions on bradycardia and cardiac conduction are variable and are hardly differentiated from effects due to the improvement of hemodynamic status. Additional clinical research and prospective clinical studies are needed to confirm the safety and efficacy of HIET and to support more formal guidelines and therapeutic regimens, but some rational recommendations can be made on the basis of the available data. Author suggested careful monitoring of blood glucose, serum potassium concentrations and the electrocardiogram is required.

Conflict of Interest

The authors declare that there is no conflict of interest regarding the publication of this paper.


References▴Top 
  1. Shah SK, Goswami SK, Babu RV, Sharma G, Duarte AG. Management of calcium channel antagonist overdose with hyperinsulinemia-euglycemia therapy: case series and review of the literature. Case Rep Crit Care. 2012;2012:927040.
    doi
  2. Kerns W, 2nd. Management of beta-adrenergic blocker and calcium channel antagonist toxicity. Emerg Med Clin North Am. 2007;25(2):309-331; abstract viii.
    doi pubmed
  3. Salhanick SD, Shannon MW. Management of calcium channel antagonist overdose. Drug Saf. 2003;26(2):65-79.
    doi
  4. DeWitt CR, Waksman JC. Pharmacology, pathophysiology and management of calcium channel blocker and beta-blocker toxicity. Toxicol Rev. 2004;23(4):223-238.
    doi
  5. Abeysinghe N, Aston J, Polouse S. Diltiazem overdose: a role for high-dose insulin. Emerg Med J. 2010;27(10):802-803.
    doi pubmed
  6. Megarbane B, Karyo S, Baud FJ. The role of insulin and glucose (hyperinsulinaemia/euglycaemia) therapy in acute calcium channel antagonist and beta-blocker poisoning. Toxicol Rev. 2004;23(4):215-222.
    doi pubmed
  7. Kenny J. Treating overdose with calcium channel blockers. BMJ. 1994;308(6935):992-993.
    doi pubmed
  8. Ezidiegwu C, Spektor Z, Nasr MR, Kelly KC, Rosales LG. A case report on the role of plasma exchange in the management of a massive amlodipine besylate intoxication. Ther Apher Dial. 2008;12(2):180-184.
    doi pubmed
  9. Durward A, Guerguerian AM, Lefebvre M, Shemie SD. Massive diltiazem overdose treated with extracorporeal membrane oxygenation. Pediatr Crit Care Med. 2003;4(3):372-376.
    doi pubmed
  10. Burkes R, Wendorf G. A multifaceted approach to calcium channel blocker overdose: a case report and literature review. Clin Case Rep. 2015;3(7):566-569.
    doi pubmed
  11. Yuan TH, Kerns WP, 2nd, Tomaszewski CA, Ford MD, Kline JA. Insulin-glucose as adjunctive therapy for severe calcium channel antagonist poisoning. J Toxicol Clin Toxicol. 1999;37(4):463-474.
    doi pubmed
  12. Greene SL, Gawarammana I, Wood DM, Jones AL, Dargan PI. Relative safety of hyperinsulinaemia/euglycaemia therapy in the management of calcium channel blocker overdose: a prospective observational study. Intensive Care Med. 2007;33(11):2019-2024.
    doi pubmed
  13. Min L, Deshpande K. Diltiazem overdose haemodynamic response to hyperinsulinaemia-euglycaemia therapy: a case report. Crit Care Resusc. 2004;6(1):28-30.
    pubmed
  14. Humbert VH, Jr., Munn NJ, Hawkins RF. Noncardiogenic pulmonary edema complicating massive diltiazem overdose. Chest. 1991;99(1):258-259.
    doi pubmed
  15. Lam YM, Tse HF, Lau CP. Continuous calcium chloride infusion for massive nifedipine overdose. Chest. 2001;119(4):1280-1282.
    doi pubmed
  16. Saravu K, Balasubramanian R. Near-fatal amlodipine poisoning. J Assoc Physicians India. 2004;52:156-157.
    pubmed
  17. Das UN. Insulin: an endogenous cardioprotector. Curr Opin Crit Care. 2003;9(5):375-383.
    doi pubmed
  18. Proano L, Chiang WK, Wang RY. Calcium channel blocker overdose. Am J Emerg Med. 1995;13(4):444-450.
    doi
  19. Patel NP, Pugh ME, Goldberg S, Eiger G. Hyperinsulinemic euglycemia therapy for verapamil poisoning: a review. Am J Crit Care. 2007;16(5):498-503.
    pubmed
  20. Boyer EW, Duic PA, Evans A. Hyperinsulinemia/euglycemia therapy for calcium channel blocker poisoning. Pediatr Emerg Care. 2002;18(1):36-37.
    doi pubmed
  21. Lheureux PE, Zahir S, Gris M, Derrey AS, Penaloza A. Bench-to-bedside review: hyperinsulinaemia/euglycaemia therapy in the management of overdose of calcium-channel blockers. Crit Care. 2006;10(3):212.
    doi pubmed
  22. Aggarwal N, Kupfer Y, Seneviratne C, Tessler S. Methylene blue reverses recalcitrant shock in beta-blocker and calcium channel blocker overdose. BMJ Case Rep. 2013;2013.
  23. Pagni S, Austin EH. Use of intravenous methylene blue for the treatment of refractory hypotension after cardiopulmonary bypass. J Thorac Cardiovasc Surg. 2000;119(6):1297-1298.
    doi pubmed
  24. Isbister GK. Delayed asystolic cardiac arrest after diltiazem overdose; resuscitation with high dose intravenous calcium. Emerg Med J. 2002;19(4):355-357.
    doi pubmed


This article is distributed under the terms of the Creative Commons Attribution Non-Commercial 4.0 International License, which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.


Cardiology Research is published by Elmer Press Inc.

 

Browse  Journals  

 

Journal of Clinical Medicine Research

Journal of Endocrinology and Metabolism

Journal of Clinical Gynecology and Obstetrics

 

World Journal of Oncology

Gastroenterology Research

Journal of Hematology

 

Journal of Medical Cases

Journal of Current Surgery

Clinical Infection and Immunity

 

Cardiology Research

World Journal of Nephrology and Urology

Cellular and Molecular Medicine Research

 

Journal of Neurology Research

International Journal of Clinical Pediatrics

 

 
       
 

Cardiology Research, bimonthly, ISSN 1923-2829 (print), 1923-2837 (online), published by Elmer Press Inc.                     
The content of this site is intended for health care professionals.

This is an open-access journal distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 International License, which permits unrestricted
non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
Creative Commons Attribution license (Attribution-NonCommercial 4.0 International CC-BY-NC 4.0)


This journal follows the International Committee of Medical Journal Editors (ICMJE) recommendations for manuscripts submitted to biomedical journals,
the Committee on Publication Ethics (COPE) guidelines, and the Principles of Transparency and Best Practice in Scholarly Publishing.

website: www.cardiologyres.org   editorial contact: editor@cardiologyres.org
Address: 9225 Leslie Street, Suite 201, Richmond Hill, Ontario, L4B 3H6, Canada

© Elmer Press Inc. All Rights Reserved.


Disclaimer: The views and opinions expressed in the published articles are those of the authors and do not necessarily reflect the views or opinions of the editors and Elmer Press Inc. This website is provided for medical research and informational purposes only and does not constitute any medical advice or professional services. The information provided in this journal should not be used for diagnosis and treatment, those seeking medical advice should always consult with a licensed physician.